• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 20
  • 3
  • 1
  • 1
  • Tagged with
  • 30
  • 19
  • 19
  • 10
  • 9
  • 8
  • 8
  • 8
  • 7
  • 6
  • 6
  • 5
  • 5
  • 5
  • 5
  • 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.
11

Analysis of Mph1 kinase and its substrates in spindle checkpoint signalling

Zich, Judith January 2010 (has links)
Accurate chromosome segregation is crucial as mis-segregation results in aneuploidy, which can lead to severe diseases such as cancer. The spindle checkpoint monitors sister-chromatid attachment and inhibits the onset of anaphase until all chromosomes are correctly bi-oriented on the mitotic spindle. The spindle checkpoint machinery of S.pombe is composed of many proteins, one of which is the kinase Mph1 (Mps1p-like pombe homolog). It previously has been shown that Mph1 is essential for the spindle checkpoint but not whether this is due to its kinase activity. In this study we determined the role of Mph1 kinase activity in the spindle checkpoint. To do so a kinase-dead version of Mph1, which had no detectable kinase activity, was analysed. Using this kinase-dead allele we showed that lack of Mph1 kinase activity abolished the spindle checkpoint and led to chromosome missegregation. As a result of these two defects cell viability of cells lacking Mph1 kinase activity was severely impaired. These results led to the question of how Mph1 kinase activity regulates the spindle checkpoint. Spindle checkpoint signalling is thought to mainly take place at two sites, at the kinetochore and at the anaphase promoting complex (APC). The APC is an E3 ubiquitin ligase that drives cells into anaphase by targeting the separase inhibitor securin and cyclin B for degradation by the 26 S proteasome. Upon activation of the spindle checkpoint the APC is inhibited by the mitotic checkpoint complex (MCC) composed of Slp1, Mad2 and Mad3. In this study we wanted to test whether the regulatory role of Mph1 kinase in the spindle checkpoint is via MCC binding to the APC. Using the kinase-dead version of Mph1 we showed that Mad2 and Mad3 binding to the APC is severely impaired in the absence of Mph1 kinase activity. This result led to the hypothesis that Mph1 might regulate Mad2 and Mad3 binding Using kinase assays Mad2 and Mad3 were identified as in vitro substrates of Mph1 and phosphorylation sites in Mad2 and Mad3 were determined by mass spectrometry. Phosphorylation mutants of Mad2 and Mad3 showed spindle checkpoint defects, indicating that they are important Mph1 substrates.
12

Initiating the Spindle Assembly Checkpoint Signal: Checkpoint Protein Mad1 Associates with Outer Kinetochore Protein Ndc80 in Budding Yeast

Weirich, Alexandra 14 June 2013 (has links)
The spindle assembly checkpoint (SAC) is an evolutionarily conserved mechanism that delays the initiation of anaphase by inhibiting the Anaphase Promoting Complex (APC) until all kinetochores have achieved bipolar attachment on the mitotic spindle. Mad1-3, Bub1, and Bub3, components of the SAC, are conserved from yeast to humans. These proteins localize to unattached kinetochores, though it is unknown with which kinetochore proteins they interact and how these interactions transduce information about microtubule attachement. Here, purification of the checkpoint proteins from Saccharomyces cerevisiae suggests that Mad1 interacts with the outer kinetochore protein Ndc80 in a SAC, cell cycle, and DNA dependent manner. Ndc80 is thought to mediate attachment of kinetochores to microtubules so the interaction between Mad1 and Ndc80 suggests a mechanism by which cells sense kinetochore-microtubule attachment. The SAC is of special importance in some types of cancer where genetic damage and aneuploidy is correlated with mutated SAC genes. A better understanding of the SAC mechanism will aid in the development of targetted cancer therpeutics.
13

Light Intermediate Chain 1: a Multifunctional Cargo Binder for Cytoplasmic Dynein 1: a Dissertation

Wadzinski, Thomas 11 September 2006 (has links)
Cells as dynamic, interactive, and self contained units of life have a need for molecular motors that can create physical forces to move cargoes within the cell. Cytoplasmic dynein 1 is one such molecular motor that has many functions in the cell. The number and variety of functions that involve cytoplasmic dynein 1 suggest that there are a number of different binding sites on dynein for different proteins. Cytoplasmic dynein 1 is a multiprotein complex made up of six different subunit families. The many different combinations of subunits that could be used to make up a cytoplasmic dynein 1 holocomplex provides the variety of different binding sites for cargoes that can be individually regulated. The following chapters flush out how light intermediate chain 1 (LIC1), a subunit of cytoplasmic dynein 1, is involved with multiple dynein functions involving the binding of different cargoes to the cytoplasmic dynein 1 holocomplex, and how the binding of these cargoes can be regulated. First, LIC1 is found to be involved in the spindle assembly checkpoint. LIC1 appears to facilitate the removal of Mad1-Mad2, a complex important in producing a wait anaphase signal, from kinetochores. Second, the involvement of LIC1 in the spindle assembly checkpoint requires the phosphorylation of LIC1 at a putative Cdk1 phosphorylation site. This site is located in a domain of LIC1 that binds various proteins suggesting that this phosphorylation could also regulate these interactions. Third, LIC1 is involved in the centrosomal assembly of pericentrin, an important centrosomal protein. From the data presented herein, LIC1 is shaping up as a multifunctional cargo binder for cytoplasmic dynein 1 that requires regulation of its various cargoes.
14

Initiating the Spindle Assembly Checkpoint Signal: Checkpoint Protein Mad1 Associates with Outer Kinetochore Protein Ndc80 in Budding Yeast

Weirich, Alexandra January 2013 (has links)
The spindle assembly checkpoint (SAC) is an evolutionarily conserved mechanism that delays the initiation of anaphase by inhibiting the Anaphase Promoting Complex (APC) until all kinetochores have achieved bipolar attachment on the mitotic spindle. Mad1-3, Bub1, and Bub3, components of the SAC, are conserved from yeast to humans. These proteins localize to unattached kinetochores, though it is unknown with which kinetochore proteins they interact and how these interactions transduce information about microtubule attachement. Here, purification of the checkpoint proteins from Saccharomyces cerevisiae suggests that Mad1 interacts with the outer kinetochore protein Ndc80 in a SAC, cell cycle, and DNA dependent manner. Ndc80 is thought to mediate attachment of kinetochores to microtubules so the interaction between Mad1 and Ndc80 suggests a mechanism by which cells sense kinetochore-microtubule attachment. The SAC is of special importance in some types of cancer where genetic damage and aneuploidy is correlated with mutated SAC genes. A better understanding of the SAC mechanism will aid in the development of targetted cancer therpeutics.
15

How to Assemble a Functional Mitotic Checkpoint Complex

Tipton, Aaron R. 20 September 2012 (has links)
No description available.
16

Regulation of the anaphase promoting complex (APC/C) in the mitotic and meiotic cell cycle of Saccharomyces cerevisiae / Regulation des Anaphase promoting Komplex (APC/C) im mitotischen und meiotischen Zellzyklus von Saccharomyces cerevisiae

Bolte, Melanie 22 January 2004 (has links)
No description available.
17

Mechanism of APC/C activation and substrate specificity in mitosis

Zhang, Suyang January 2018 (has links)
In eukaryotes, cell proliferation and cell cycle transitions are strictly controlled by the anaphase-promoting complex/cyclosome (APC/C). The APC/C is an E3 ubiquitin ligase that regulates chromatid segregation at the metaphase to anaphase transition, exit from mitosis and the establishment and maintenance of G1. The APC/C’s catalytic activity and substrate specificity are controlled by its interactions with two coactivators, Cdc20 and Cdh1. In contrast to Cdh1, APC/C activation by Cdc20 during mitosis requires hyper-phosphorylation of APC/C subunits by cyclin-dependent kinase (Cdk) and polo kinase. The aim of the first part of this thesis was to understand how mitotic phosphorylation regulates APC/C activity. Using cryo-electron microscopy and biochemical analysis, we found that an auto-inhibitory segment of the Apc1 subunit acts as a molecular switch that in apo unphosphorylated APC/C interacts with a coactivator-binding site (C-box binding site), thereby obstructing engagement of Cdc20. Phosphorylation of the auto-inhibitory segment displaces it from the C-box binding site to relieve APC/C auto-inhibition. Efficient phosphorylation of the auto-inhibitory segment requires the recruitment of the kinase Cdk-cyclin-Cks to a hyper-phosphorylated loop of Apc3. In addition to regulation of APC/C activity by phosphorylation, ordered cell progression is ensured by the ability of the APC/C to target substrate degradation in a defined order. At mitosis onset, degradation of securin and cyclin B1 is inhibited by the spindle assembly checkpoint, exerted by the mitotic checkpoint complex (MCC), whereas both cyclin A2 and Nek2A are not subject to MCC inhibition. The aim of the second part of the thesis was to elucidate the mechanism of how the APC/C achieves its substrate specificity. Our biochemical analysis showed that the resistance of cyclin A2 to MCC inhibition is due to its ABBA motif and the Cdk-associated Cks2 subunit. Furthermore, we found that it is the Cdc20 molecule of the MCC that binds to the ABBA motif to allow for cyclin A2 ubiquitination. Strikingly, mutating all three known degrons (KEN box, D box and ABBA motif) of cyclin A did not affect its ubiquitination by APC/CCdc20. Deletion of a potential novel degron found within residues 60-80 of cyclin A2 impaired cyclin A2 ubiquitination.
18

HnRNP E1 Protects Chromosomal Stability Through Post-Transcriptional Regulation of Cdc27 Expression

Schwartz, Laura Link 30 November 2015 (has links)
No description available.
19

Post-replicative resolution of under-replication

Carrington, James T. January 2017 (has links)
The evolutionary pressure to prevent re-replication by inactivating licensing during S phase leaves higher-eukaryotes with large genomes, such as human cells, vulnerable to replication stresses. Origins licensed in G1 must be sufficient to complete replication as new origins cannot be licensed in response to irreversible replication fork stalling. Interdisciplinary approaches between cellular biology and biophysics predict that replication of the genome is routinely incomplete in G2, even in the absence of external stressors. The frequency of converging replication forks that never terminate due to irreversible stalling (double fork stall), which result in a segment of unreplicated DNA, was modelled using high quality origin-mapping data in HeLa and IMR-90 cells. From this, hypotheses were generated that related an increase in unreplicated segments of DNA to reduced functional origin number. Presented in this thesis is the confirmation of this relation by quantifying chromosome mis-segregation and DNA damage responses when origin number was reduced using RNAi against licensing factors. The number of ultrafine anaphase bridges and 53BP1 nuclear bodies are in remarkable concordance with the theoretical predictions for the number of double fork stalls, indicating that cells are able to tolerate under-replication through such post-replicative cellular responses. 53BP1 preferentially binds to chromatin associated with large replicons, and functions synergistically with dormant origins to protect the stability of the genome. Additional candidates, inspired by common fragile site research, have also been characterised as responders to spontaneous under-replication, and include FANCD2 and MiDAS, which function in early mitosis to facilitate completion of replication before cells enter anaphase. In conclusion, a series of mechanisms that sequentially function throughout the cell cycle protects the stability of the human genome against inevitable spontaneous under-replication brought about by its large size.
20

Le rôle du APC (Anaphase-Promoting Complex) <br />au cours de la phase G2/M après dommage de l'ADN

Lee, Jinho 29 October 2007 (has links) (PDF)
Les agents permettant de créer des dommages sur l'ADN sont principalement utilisés dans les traitements contre le cancer. L'activation de points de contrôle du cycle cellulaire après lésion de l'ADN entraîne un arrêt du cycle des cellules. De la connaissance des mécanismes moléculaires de l'arrêt du cycle cellulaire par ces points de contrôle dépend l'efficacité du traitement. Dans les cellules humaines, ces points de contrôle sont primordiaux puisque leur inactivation entraîne la carcinogenèse (génération de cancers). Après traitement par des agents chimiothérapiques et des rayons X, les cellules s'arrêtent en phase G-1 et G-2/Mitose (M) du cycle cellulaire. Si de nombreuses études ont permis de clarifier les mécanismes de l'arrêt en phase G-1 pour des cellules dont l'ADN est endommagé, peu de données sont disponibles concernant l'arrêt en phase G-2/M. Parmi ces points de contrôle, le point de contrôle G-2/M est particulièrement important car il prévient l'entrée en mitose (phase M) des cellules dont l'ADN est endommagé. <br /> Nous avons analysé le rôle du complex appelé APC (Anaphase-Promoting Complex) dans les points de contrôle G-2/M après lésion de l'ADN. Les lésions de l'ADN sont induites dans les cellules synchronisées en phase S. Suite à ces dommages, les cellules montrent un retard et s'arrêtent en phase G-2 avec 4N chromosomes. Afin d'identifier les bases biochimiques de l'arrêt en G-2/M après traitement avec des agents endommageant l'ADN, nous allons concentrer notre recherche sur un complexe composé de multiples protéines possédant une activité de ligation de l'ubiquitine de type E3 (ubiquitin-ligase E3). Ce complexe APC est necessaire pour la dégradation des inhibiteurs d'entrée en anaphase, cyclins mitotiques, et plusieurs kinases mitotiques pour la complétion de la sortie de la mitose. Nous avons analysé et déterminé que l'absence d'activité du complexe APC inhibe l'activation du point de contrôle G-2/M lors de dommages de l'ADN.

Page generated in 0.058 seconds