• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 38
  • 6
  • 5
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 64
  • 32
  • 17
  • 16
  • 15
  • 12
  • 12
  • 10
  • 9
  • 8
  • 8
  • 7
  • 7
  • 7
  • 7
  • 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.
21

Targeting Protein Phosphatase 2a as a Therapeutic Strategy for Chronic Lymphocytic Leukemia

Liu, Qing 22 October 2008 (has links)
No description available.
22

Novel Small Molecules Regulating The Histone Marking, AR Signaling, And AKT Inhibition In Prostate Cancer

Huang, Po-Hsien 23 August 2010 (has links)
No description available.
23

B55alpha modulates the phosphorylation status of the pRb-related p107 and p130 proteins

Jayadeva, Girish January 2010 (has links)
The retinoblastoma family of phosphoproteins consisting of the retinoblastoma protein (pRB) and the two structurally related proteins p130 and p107 play an important role in the negative regulation of cell cycle progression. Hypophosphorylated pocket proteins interact with the different members of the E2F family and repress the transcription of E2F-dependent genes and consequently suppress cell cycle progression through the G0/G1 transition and the restriction point in G1. Mitogenic stimulation results in sequential activation of cyclin/CDK complexes in mid to late G1, leading to subsequent hyperphosphorylation at multiple Ser/Thr sites of pocket proteins triggering dissociation of pocket protein/E2F complexes. This disruption leads to de-repression of many E2F dependent genes whose products are essential for cell cycle progression. The traditional view has been that pocket proteins continue to be hyperphosphorylated through the S and G2 phases and following cyclin/CDK inactivation during mitotic exit become dephosphorylated by action of PP1. However, our lab observed that upon treatment of asynchronously growing cells with the CDK inhibitor Flavopiridol or CHX, pocket proteins, are rapidly dephosphorylated correlating with the inactivation of G1/CDKs and down regulation of D-type cyclins, respectively. Pocket protein dephosphorylation was prevented by pre-treating these cells with phosphtase inhibitors at a concentration selective for PP2A, implicating PP2A or PP2A-like serine/threonine phosphatase in this iii process. The involvement of PP2A on pocket protein dephosphorylation was further strengthened by the observation that SV40 small t antigen (ST) delays/prevents p107 dephosphorylation. Moreover, a physical association between PP2A/C and p130/p107 was observed throughout the cell cycle that was not affected by CHX treatment, strongly suggesting that CHX-induced dephosphorylation is not the result of increased pocket protein targeting by PP2A, but rather that a dynamic equilibrium between CDKs and PP2A is shifted to dephosphorylation when CDK activity is compromised. This dynamic equilibrium operates throughout the cell cycle. PP2A is a trimeric enzyme complex consisting of a catalytic C, a structural A and substrate specific B subunit. There are four families of regulatory B subunits designated B, B’, B’’ and B’’’, each with several members encoded by genes with multiple splice variants that mediate substrate specificity and subcellular localization. It has been reported recently that in excess of 200 functional distinct PP2A holoenzymes can assemble with distinct specificities. Therefore, to gain insight into the mechanisms that regulate the steady state phosphorylation of pocket proteins throughout the cell cycle, it was essential to identify the specific holoenzyme complexes involved. To this end, it was identified that a PP2A trimeric holoenzyme containing B55α specifically targets and dephosphorylates p107/p130 both in vitro and in mammalian cells. B55α associates directly with the spacer of p107 and this interaction seems to be indirectly enhanced by the C-terminus of p107. The decreased association of p107 with PP2A/C of the B55α/PP2A holoenzyme complex upon treatment with ST further confirmed the role of B55α in mediating p107-PP2A/C interaction. Our data also revealed an interaction between B55α and p130, but not pRb, which appears to prefer a PR70, suggesting selectivity in the interaction of pocket proteins with distinct PP2A holoenzymes. In accordance with this, recombinant purified B55α dephosphorylates p107 in vitro. Limited ectopic expression of B55α but not other subunits, result in ST sensitive dephosphorylation of p107 and p130 in cells. Further shRNA mediated knockdown of B55α results in hyperphosphorylation of p107 and p130. This suggests that the cellular levels of B55α are critical in modulating the phosphorylation status of p107/p130 rather than just catalyzing the dephosphorylation of these proteins when the activity of CDKs is compromised. Since ST disrupts the B55α/PP2A holoenzyme complex by binding to the PP2A-A-C dimer and leads to hyperphosphorylation of pocket proteins it is conceivable that ST mediates its effects on cell proliferation at least in part, via inactivation of the PP2A holoenzymes that activates pocket proteins. Given the sensitivity of p107 phosphorylation to the cellular levels of B55α, future analyses should ascertain if deregulation of B55α leads to hyperphosphorylation of pocket proteins and abnormal cell cycle progression. / Molecular Biology and Genetics
24

The B55α/PP2A Holoenzyme in Cell Cycle Exit, Maturation/Differentiation, and Cancer

Kurimchak, Alison January 2014 (has links)
The cell cycle is negatively regulated by members of the pocket protein family, which consists of the tumor suppressor pRB and two closely related paralogs, p107 and p130. In their hypophosphorylated state, they are associated with E2F transcription factors which result in the repression of transcription of E2F-dependent genes that are required for cell cycle progression. The phosphorylation state of pocket proteins during the cell cycle is determined at least in part by an equilibrium between inducible CDKs and the serine/threonine protein phosphatase PP2A. Protein Phosphatase 2A (PP2A), is a serine/threonine phosphatase that functions as as a collection of trimeric holoenzymes. The trimeric PP2A holoenzyme is composed of the "A" scaffolding subunit, the "C" catalytic subunit, and a "B" regulatory subunit. The B subunit is the major determinant in substrate specificity and subcellular localization. Two holoenzymes consisting of the core PP2A dimer and either the B55α or PR70 regulatory subunits have been implicated in the activation of p107/p130 and pRB, respectively. While the phosphorylation state of p107 is very sensitive to forced changes of B55α levels in human cell lines, regulation of p107 in response to physiological modulation of PP2A/B55α has not been previously elucidated. In this thesis, I show that FGF1, which induces maturation and cell cycle exit in chondrocytes, triggers rapid accumulation of p107/PP2A/B55α complexes coinciding with p107 dephosphorylation without an increase in B55α protein expression in RCS cells. Reciprocal solution-based mass-spectrometry analysis identified the PP2A/B55α complex as a major component of a subset of p107 complexes, which also contain E2F/DPs, DREAM subunits and cyclin/CDK complexes. p107 is one of the major partners of B55α, which also associates with pRB in RCS cells. FGF1 induces dephosphorylation of p107, its translocation to the nucleus, remodeling of p107 complexes, and enhances its interaction with E2F4 and other p107 partners. Consistent with an essential role of B55α in the rapid activation of p107 in chondrocytes, limited ectopic expression of B55α results in marked dephosphorylation of p107, while B55α knockdown results in hyperphosphorylation. More importantly, limited knockdown of B55α dramatically delays FGF1 induced dephosphorylation of p107. Moreover, dephosphorylation of p107 in response to FGF1 treatment results in selective recruitment of p107 to regulated genes including CMYC. Our results suggest a model where FGF1 mediates rapid dephosphorylation and activation of p107 independently of the CDK activities that maintain p130 and pRB hyperphosphorylated for several hours post p107 dephosphorylation in maturing chondrocytes. Additionally, we provide preliminary evidence that PPP2R2A may act as a haploinsufficient tumor suppressor in prostate cancer cell lines. PPP2R2A is hemizygously deleted in various prostate cancer cell lines and tumor samples. We identified three cell lines that express less B55α the gene product of PPP2R2A, than cell lines that are reported to have both alleles intact. Furthermore, ectopic expression of B55α in PC3 cells results in a phenotype reminiscent of senescence, ultimately leading to cell death. These cells are unable to form colonies in soft agar and have increased DNA content and euploidy. Combined with their larger cell and nuclear size, this suggests that ectopic expression of B55α in PC3 cells results in endoreplication. Altogether these suggest that reduced B55α expression in these cells confers a growth advantage in PCa cell lines, which is extinguished when B55α is reintroduced, supporting the notion that hemizygous deletion of PPP2R2A in prostate tumors may help promote tumorigenesis. / Molecular Biology and Genetics
25

PPP2R2A Prostate Cancer Haploinsufficiency is Associated with Worse Prognosis and a High Vulnerability to B55α/PP2A Reconstitution that Triggers Centrosome Destabilization and Inhibits Cell Invasion

Zhao, Ziran January 2020 (has links)
The PPP2R2A gene encodes the B55α regulatory subunit of PP2A. Here we report that PPP2R2A is hemizygously lost in ~42% of prostate adenocarcinomas, correlating with reduced expression, poorer prognosis, and an increased incidence of hemizygous loss (>75%) in metastatic disease. Of note, PPP2R2A homozygous loss is less common (5%) and not increased at later tumor stages. Reduced expression of B55α is also seen in prostate tumor tissue and cell lines. Consistent with the possibility that complete loss of PPP2R2A is detrimental in prostate tumors, PPP2R2A deletion in cells with reduced but present B55α reduces cell proliferation by slowing progression through multiple phases in the cell cycle. Remarkably, B55α-low cells also appear addicted to lower B55α expression, as even moderate increases in B55α expression are toxic. Reconstitution of B55α expression in prostate cancer (PCa) cell lines with low B55α expression reduces proliferation, inhibits transformation and blocks xenograft tumorigenicity. Mechanistically, we show B55α reconstitution reduces phosphorylation of proteins essential for centrosomal maintenance, and induces centrosome collapse and chromosome segregation failure; a first reported link between B55α/PP2A and the vertebrate centrosome. These effects are dependent on a prolonged metaphase to anaphase checkpoint and are lethal to PCa cells addicted to low levels of B55α. Thus, we propose the reduction in B55α levels associated with hemizygous loss is necessary for centrosomal integrity in PCa cells, leading to selective lethality of B55α reconstitution. Such a vulnerability could be targeted therapeutically in the large pool of patients with hemizygous PPP2R2A deletions, using pharmacologic approaches that enhance PP2A/B55α activity. With that aim and considering the limitations of conventional 2D cell culture in mimicking the tumor environment and predicting drug responses in animal models and humans, we also established 3D organoid cultures of PCa cells and immortalized human prostate epithelial cells (hPrEC) in Matrigel. This allowed us to explore cell to extracellular matrix (ECM) interactions. PC3 cells initially form round organoids in Matrigel, followed by an invasive switch to where cell protrusions invade the surrounding ECM. Strikingly, B55α reconstitution, dramatically suppressed rupture of the basement lamina and ECM invasion, while proliferation appeared not affected. Tracking organoid growth at defined time points or using live imaging, shows that protrusions in PC3 organoids are very dynamic and resemble invadopodia. Interestingly, reconstitution of B55α in PC3 organoids just prior the invasive switch results in reduction of invasive leads and those protrusions that appear to initiate keep forming and collapsing, with most organoids remain round. Our previous phosphoproteomics data in 2D culture suggests that cell-to-ECM signaling is likely altered with B55α reconstitution, identifying potential B55α/PP2A substrates among key mediators of integrin signaling. In sum, reconstitution of B55α suppresses invasion in PC3 organoids, possibly by regulating potential B55α substrates in focal adhesion signaling, such as Paxillin and/or Talin. Alternatively, centrosomal defects due to dephosphorylation of B55α substrates (e.g. HAUS6, NEDD1) may cause microtubule defects that preclude invasion. Further studies are required to identify the mechanism. Moreover, because our studies presented above are based on prostate cancer cell lines with undefined genetic alterations, we have immortalized primary human PrEC by a novel approach to generate a cell model to study cooperation of PPP2R2A loss with step-wise introduction of specific oncogenes and/or tumor suppressor gene alterations in transformation, tumorigenicity and invasion. Our newly develop method combines expression of hTERT, which appears insufficient for immortalization of hPrEC with CDKN2A knockout, which we predicted will prevent stress-induced replicative senescence. We have obtained two independent immortalized clones (hPrEC-T-ΔN2A) using this method and confirmed their identity using PCR and western blot analyses. Although cytogenetic analysis showed these two clones are of mixed population with minor alterations in karyotype, 4 out of 11 cells examined in clone 1 appear completely normal. We also find that the clones exit the cell cycle upon contact inhibition and induce p53 expression when treated with flavopiridol, further supporting that hPrEC-T-ΔN2A clones exhibit the features of normal cells. Characterization in 3D culture reveals that the clones are likely of basal epithelial origin. Finally, soft agar and clonogenic assays show hPrEC-T-ΔN2A clones are highly proliferative but not transformed. We are using these cell models to dissect the role of PPP2R2A depletion in step-wise transformation of immortalized PrEC and hope to develop a defined 3D organoid system to study invasion, which could also be suitable for drug screens. Altogether our work has very significantly advanced our understanding of B55α in suppressing transformation in prostate cancer cells and has developed novel tools for further mechanistic characterization of PPP2R2A haploinsufficiency and the development of potential pharmacologic therapeutic agents. / Biomedical Sciences
26

Spatiotemporal regulation of the Greatwall : PP2A axis is required for mitotic progression

Wang, Peng 09 1900 (has links)
Le cycle cellulaire est hautement régulé par la phosphorylation réversible de plusieurs effecteurs. La kinase dépendante des cyclines Cdk1 déclenche la mitose en induisant le bris de l’enveloppe nucléaire, la condensation des chromosomes et la formation du fuseau mitotique. Chez les animaux métazoaires, ces évènements sont contrés par la protéine phosphatase PP2A-B55, qui déphosphoryle plusieurs substrats de Cdk1. La kinase Greatwall (Gwl) est activée par le complexe cycline B-Cdk1 en début de mitose et induit ensuite l’inhibition de PP2A-B55 via Endos/Arpp19. Toutefois, les mécanismes moléculaires qui régulent Gwl sont encore peu connus. Nous avons montré que Gwl a une activité s’opposant à PP2A-B55, qui collabore avec la kinase Polo pour assurer l’attachement du centrosome au noyau et la progression du cycle cellulaire dans le syncytium de l’embryon de la drosophile. Ensuite, nous avons trouvé dans des cellules de drosophile que Gwl est localisée au noyau pendant l’interphase, mais qu’elle se relocalise au cytoplasme dès la prophase, avant le bris de l’enveloppe nucléaire. Nous avons montré que cette translocation de Gwl est cruciale pour sa fonction et qu’elle dépend de la phosphorylation de plusieurs résidus de la région centrale de Gwl par les kinases Polo et Cdk1. Cette région centrale contient également deux séquences de localisation nucléaire (respectivement NLS1 et NLS2). De plus, nos résultats suggèrent que la phosphorylation de Gwl par la kinase Polo promeut sa liaison avec la protéine 14-3-3ε, ce qui favorise la rétention cytoplasmique de Gwl. Le rôle de Cdk1 dans cette translocation reste quant à lui inconnu. De plus, nous avons montré que le complexe cycline B-Cdk1 entre dans le noyau avant que Gwl ne soit transportée dans le cytoplasme. Cdk1 pourrait donc activer Gwl et phosphoryler ses substrats nucléaires, à l’abri de PP2A-B55 qui est largement cytoplasmique. Gwl est ensuite exclue du noyau et relocalisée dans le cytoplasme afin d’induire l’inhibition de PP2A-B55. Cela permet de synchroniser les événements de phosphorylation se produisant dans le noyau et dans le cytoplasme. Fait intéressant, un mécanisme de régulation de la localisation de Gwl similaire à cela a été découvert chez l’humain et chez la levure, suggérant que ce mécanisme est conservé entre différentes espèces. / Reversible phosphorylation of proteins, triggered by cyclically activated kinases and phosphatases, is a key mechanism to control cell cycle progression. CyclinB-Cdk1 is a crucial kinase phosphorylating a large number of substrates to trigger mitotic entry. However, in metazoans, it is counteracted mainly by a Protein Phosphatase 2A carrying the B55 regulatory subunit (PP2A-B55). On the other hand, the Greatwall (Gwl) kinase is activated by CyclinB-Cdk1 upon mitotic entry and subsequently induces the inhibition of PP2A-B55 by Endos/Arpp19, thus promoting mitotic entry and maintenance. Nonetheless, the regulatory mechanisms of Gwl are less clear. We demonstrated that in Drosophila syncytial embryos, PP2A-B55 is negatively regulated by Gwl, but collaborates with Polo kinase to ensure both nucleus attachment of centrosome and faithful cell cycle progression. Later, we discovered that in Drosophila, the subcellular localization of Gwl changes dramatically throughout the cell cycle. Gwl is nuclear in interphase but suddenly becomes mostly cytoplasmic in prophase before nuclear envelope breakdown. Such translocation is important for Gwl’s function and requires the phosphorylation of Gwl by both Polo kinase and Cdk1 in the region containing two Nuclear Localization Signals (NLSs). Phosphorylation of Gwl by Polo likely promotes its association with14-3-3ε thereby promoting Gwl cytoplasmic retention, whereas Cdk1’s role in this translocation remains elusive. Moreover, I found that most cyclin B is imported into the nucleus before Gwl translocates to the cytoplasm. Therefore, Cdk1 can activate Gwl and phosphorylate its nuclear substrates without the perturbation of PP2A-B55 which is largely cytoplasmic. Subsequently, Gwl translocates into cytoplasm to mediate the inhibition of PP2A-B55 so that the phosphorylation events can be synchronized between the nucleus and the cytoplasm. Interestingly, similar spatial regulation of Gwl was also uncovered in mammal cells and in yeast, implying a conserved regulatory mechanism across species.
27

Charakterisierung der Mikrotubulus-assoziierten PP2A und ihrer Zielproteine

Krauß, Sybille Ellen 23 November 2005 (has links)
In der vorliegenden Arbeit sollten Ziel-Proteine der Mikrotubulus-assoziierten PP2A gefunden werden. Anhand phänotypischer Ähnlichkeiten zwischen OS- und Greig-, Acrocallosal- bzw. Pallister-Hall-Syndrom-Patienten wurde eine mögliche Interaktion zwischen dem MID1-alpha4-PP2A-Komplex und GLI3, einem zentralen Transkriptionsfaktor der SHH-Signaltransduktionskaskade, postuliert. In einer Reihe von zellbiologischen und proteinbiochemischen Experimenten konnte gezeigt werden, dass sowohl die intrazelluläre Lokalisation des GLI3 als auch der Phosphorylierungsstatus von Fu, einem Interaktionspartner von GLI3, über den MID1-alpha4-PP2A-Komplex und Mikrotubulus-assoziierter PP2A-Aktivität reguliert werden. Erhöhte Aktivität der Mikrotubulus-assoziierten PP2A führt hierbei zur Dephosphorylierung von Fu und zu einer Akkumulation des GLI3 im Cytosol, während verringerte PP2A-Aktivität zu einer Anreicherung der hyperphosphorylierten Form des Fu und zur Akkumulation des GLI3 im Nukleus führt. Darüber hinaus konnte GSK3beta als die der Mikrotubulus-assoziierten PP2A entgegenwirkende Kinase identifiziert werden. Eine verringerte Aktivität der GSK3beta führt zur Dephosphorylierung von Fu und zu einer Akkumulation des GLI3 im Cytosol. Außerdem wurde in der vorliegenden Arbeit eine Interaktion zwischen GLI3 und der hyperphosphorylierten Form des Fu beschrieben. Die Hyperphosphorylierung von Fu wird über die gegenläufigen Aktivitäten der Mikrotubulus-assoziierten PP2A und GSK3beta reguliert. Durch die Interaktion des hyperphosphorylierten Fu mit cytosolischem, nicht phosphorylierten GLI3 wird dessen Phosphorylierung gesteuert. Phosphoryliertes GLI3 reichert sich im Zellkern an und die Transkription von SHH-Zielgenen wird induziert. Die in dieser Arbeit identifizierten Mechanismen sind ein möglicher zellbiologischer Hintergrund der Übereinstimmung in den klinischen Erscheinungsbildern von OS und Syndromen, die mit Genen der SHH-Signaltransduktionskaskade assoziiert sind. / Misregulation of microtubule-associated phosphatase 2A (PP2A) activity as a result of mutations in the ubiquitin ligase MID1 plays a central role in the pathogenesis of Opitz BBB/G syndrome (OS). Features typical for OS are shared by patients with mutations in GLI3 and PATCHED1 (PTC1), two members of the Sonic Hedgehog (SHH) pathway. These observations suggest that MID1 / PP2A may also be involved in the transduction of the SHH signal. Here we demonstrate that nuclear translocation of the transcription factor GLI3, a major effector of the SHH pathway, is regulated by the activity of the microtubule-associated pool of PP2A. This effect is reproduced pharmacologically by lithium chloride (LiCl), a potent inhibitor of glycogen synthase kinase 3beta (GSK3beta), and correlates with the phosphorylation status of human Fused (hFu), a GLI3 interaction partner. Our data suggest an antagonistic relationship between PP2A and GSK3beta as regulators of SHH signaling and provide a molecular basis for the phenotypic overlap between patients with OS and SHH pathway mutations.
28

Functional investigation of the class II tumor suppressor gene H-REV107-1

Nazarenko, Irina 16 October 2003 (has links)
Das Klasse II Tumorsuppressor-Gen H-REV107-1, ist in normalen Geweben ubiquitär exprimiert, zeigt jedoch häufig Expressionsverluste, vorzugsweise in Tumoren des Brustgewebes, des Ovars und der Lunge. Das H-REV107-1 Protein wirkt in vitro und in vivo als Wachstumssuppressor. Um den Mechanismus der H-REV107-1 bedingten Wachstumshemmung zu verstehen, haben wir mit Hilfe des LexA-basierten Hefe-2-Hybrid Systems interagierende Proteine identifiziert. Diese Suche wurde mit einem H-REV107-1 Deletionskonstrukt durchgeführt, dem die Membran-bindende Domäne fehlte. Diese Analyse lieferte eine Vielzahl von potentiellen Interaktionspartnern, darunter der Retinsäure Rezeptor RARG, das Calcium-bindende Proteine S100A6, der Translations-Elongationsfaktor ETF und das weitgehend unbekannte Protein p14.5 Die Bindungen des H-REV107-1 Proteins an die beiden potentiellen Kandidaten, den Transkriptionsfactor PC4 und die regulatorische Untereinheit der Protein Phosphatase 2A (PR65), wurden genauer untersucht. Wir haben dabei einen Proteinkomplex aus H-REV107-1, PC4 und STAT1 (Signal Transducer and Activator of Transcription 1) identifiziert, der vermutlich eine Rolle in der IFNgamma - abhängigen Wachstumshemmung in Ovarialkarzinom Zellen spielt. Da sich die Expression des H-REV107-1 Gens durch IFNgamma über den Transkriptionsfaktor IRF-1 stimulieren läßt, und in verschiedenen Zelllinien sowohl zur Hemmung des Wachstums, als auch zur Apoptose führt, vermuteten wir verschiedene Mechanismen der Wachstumshemmung durch den IFNgamma-Signalweg und H-REV107-1. Weitere Analysen der H-REV107-1 - vermittelten Apoptose zeigten, daß die Interaktion zwischen H-REV107-1 und PR65 eine wichtige Rolle in diesem Prozeß spielt. Um die Proteindomäne zu identifizieren, welche für die direkte Wechselwirkung von H-REV107-1 mit PR65 verantwortlich ist, wurden H-REV107-1 Mutanten generiert und mittels Co-Immunpräzipitation getestet. Die Prolin-reiche Sequenz am N-Terminus des H-REV107-1 Proteins konnte als verantwortliche Domäne für die Interaktion bestimmt werden. Die funktionelle Analyse dieser Interaktion zeigte die Hemmung der Protein Phosphatase 2A (PP2A) Aktivität in Ovarialkarzinom Zellen durch H-REV107-1. Der Einsatz der Mutanten im Phosphatase-Aktivitätstest zeigte, daß die selbe Domäne, die die Interaktion vermittelt, auch für die Hemmung der Phosphatase 2A verantwortlich ist. Diese Fakten deuteten auf eine wichtige Rolle der Phosphatase 2A in Ovarialkarzinom Zellen hin, weil sowohl die Verwendung des PP2A Inhibitors (Okadainsäure), als auch die Transfektion der Zellen mit einem H-REV107-1 - Expressionsplasmid zur Apoptose führten. Damit konnten wir zeigen, daß PP2A für das Überleben der Ovarialkarzinomzellen notwendig ist, und die Reaktivierung des H-REV107-1 Proteins durch IFNgamma zur Hemmung der Phosphatase und damit zur Apoptose führt. / The H-REV107-1 class II tumor suppressor gene is ubiquitously expressed in normal tissues and down-regulated in human breast, ovarian and lung tumours. H-REV107-1 has the capacity to suppress growth of tumour cells in vitro and in vivo. To understand the mechanism of H-REV107-1 mediated growth suppression I performed a search for H-REV107-1 interacting proteins using a LexA-based yeast two-hybrid system. I screened a human kidney cDNA library with a truncated form of the H-REV107-1 as a bait. This analysis revealed numerous potential interaction partners. Among them the retinoic acid receptor gamma (RARG), the calcium-binding protein S100A6, the translation termination factor ETF1, and the potential translational inhibitor protein P14.5. The interaction of H-REV107-1 with the transcriptional co-activator PC4 and with the regulatory subunit A of protein phosphatase 2A (PR65) was analysed in detail. H-REV107-1 binds ectopically expressed and endogenous PC4. In addition, a multiprotein complex between H-REV107-1, PC4 and the signal transducer and activator of transcription 1 (STAT1) was demonstrated. This complex is likely to be involved in IFNgamma-mediated growth suppression of ovarian carcinoma cells. Endogenous H-REV107-1 can be induced after application of IFNgamma through the IRF-1 transcription factor. This up-regulation of H-REV107-1 leads either to growth suppression via a G1 arrest or to apoptosis depending on the cell line, suggesting different mechanisms of IFNgamma-, and H-REV107-1- mediated growth suppression. Further investigation of the mechanism of H-REV107-1-dependent apoptosis revealed an important role of the interaction between H-REV107-1 and the PR65 protein. The use of several H-REV107-1 mutant proteins generated after disruption of the highly conserved domains identified the proline-rich N-terminal domain responsible for the interaction with PR65 in Co-immunoprecipitation studies. Functional investigation of the H-REV107-1 - PR65 interaction demonstrated that wild-type H-REV107-1 is able to inhibit PP2A activity, however a mutant protein lacking the N-terminal domain was devoid of this function. We sought to identify the functional relevance of the PP2A activity in ovarian carcinoma cells with normally have suppressed the H-REV107-1 gene. Treatment of OVCAR-3 cells with the PP2A inhibitor Okadaic acid and transient transfection of the cells with wild-type H-REV107-1 resulted in the activation of caspase-9, suggesting a role for PP2A in the survival of ovarian carcinoma cells. We suggest, that the down-regulation of H-REV107-1 in ovarian carcinomas is a prerequisite for the PP2A-dependent activation of yet unknown signalling pathways mediating tumour cell survival. Reactivation of H-REV107-1 gene expression via IFNgamma leads to the inhibition of PP2A activity and tumour cell death.
29

Cdc55 controls the balance of phosphatases to coordinate spindle assembly and chromosome disjunction during budding yeast meiosis

Bizzari, Farid Fouad Mahmoud January 2012 (has links)
Meiosis is the process by which haploid gametes are produced from a diploid cell. It is a specialised form of cell division which involves one round of DNA replication followed by two rounds of chromosome segregation. Errors in the segregation process can give rise to aneuploidy, which can result in miscarriages and birth defects. In the first meiotic division homologous chromosomes are segregated, and sister chromatids are segregated in the second division. This is coordinated with two rounds of spindle microtubule assembly and disassembly. How these two processes are coordinated is unknown. In my PhD, I study the role of the protein phosphatase 2A (PP2A) regulatory subunit, Cdc55, in budding yeast meiosis. PP2A is a conserved heterotrimeric enzyme that has important roles in mitosis and meiosis. These roles are dictated by binding to either of its two regulatory subunits, Rts1 and Cdc55, in budding yeast . I show that Cdc55 is required for the proper assembly of a meiotic spindle in meiosis I, through the maintenance of the Cdc14 phosphatase in the nucleolus early in meiosis. In addition, Cdc55 is also required to limit the formation of PP2A complexes with the Rts1 regulatory subunit, and this is essential for the timely dissolution of sister chromatid cohesion. Thus, Cdc55 couples spindle assembly with chromosome segregation through its interactions with Cdc14 and PP2ARts1. Finally, I show some preliminary studies looking at the possible downstream effectors of Cdc14 that are important in this mechanism.
30

p63 and potential p63 targets in squamous cell carcinoma of the head and neck

Boldrup, Linda January 2008 (has links)
Squamous cell carcinoma of the head and neck (SCCHN), the 6th most common cancer worldwide, has a low 5-year survival. Disease as well as treatment often causes patients severe functional and aesthetic problems. In order to improve treatment and diagnosis at earlier stages of tumour development it is important to learn more about the molecular mechanisms behind the disease. p63, an important regulator of epithelial formation, has been suggested to play a role in the development of SCCHN. Six different isoforms of p63 have been found and shown to have various functions. The aim of the studies in this thesis was to learn more about the role of p63 and proteins connected to p63 in SCCHN. Expression of p63, Cox-2, EGFR, beta-catenin, PP2A and p53 isoforms was mapped in tumours and normal tumour adjacent tissue from patients with SCCHN using western blot or RT-PCR. Results showed no significant difference between tumours and normal tumour adjacent tissue concerning expression of EGFR and beta-catenin. Cox-2 and PP2A showed significantly higher expression in tumours while p63 was more expressed in normal tumour adjacent tissue. However, expression of all these proteins in normal tumour adjacent tissue differed from tissue from disease-free non-smoking individuals. Smoking in itself did not affect expression of these proteins. The p53 isoforms p53, p53beta, p53gamma, ∆133p53, ∆133p53beta and ∆133p53gamma were expressed at RNA level in samples both from tumours and normal tumour adjacent tissue, though most of them at fairly low levels. The functional properties of the different p63 isoforms have not been fully mapped. By establishing stable cell lines over-expressing the different p63 isoforms we investigated their specific effect on tumour cells from SCCHN. Only the ∆Np63 isoforms could be stably over-expressed, whereas no clones over-expressing TAp63 could be established. Using microarray technique, cell lines stably expressing the ∆Np63 isoforms were studied and CD44, Keratins 4, 6, 14, 19 and Cox-2 were found to be regulated by p63. In conclusion, the present project adds new data to the field of p63 and SCCHN. For example, we have shown that clinically normal tumour adjacent tissue is altered compared to normal oral mucosa in non tumour patients, and that smoking does not change expression of p63, Cox-2, EGFR, beta-catenin or PP2A in oral mucosa. Novel p53 isoforms are expressed in SCCHN, and even though levels are very low they should not be overlooked. Furthermore, CD44, keratins 4, 6, 14, 19 and Cox-2 were identified as p63 targets in SCCHN.

Page generated in 0.4105 seconds