• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 23
  • 11
  • 5
  • 4
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 57
  • 16
  • 16
  • 15
  • 14
  • 14
  • 12
  • 12
  • 11
  • 9
  • 8
  • 8
  • 8
  • 8
  • 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.
1

Evading the anti-tumour immune response : a novel role for Focal Adhesion Kinase

Lund, Thomas Anthony January 2016 (has links)
Here I describe a new function of Focal Adhesion Kinase (FAK) in driving anti-tumour immune evasion. The kinase activity of FAK in squamous cancer cells drives the recruitment of regulatory T-cells (Tregs) by transcriptionally regulating chemokine/cytokine and ligand-receptor networks, including the transcription of CCL5 and TGFβ, which are required for enhanced Treg recruitment. In turn, these changes inhibit antigen-primed cytotoxic CD8+ T-cell activity in the tumour microenvironment, permitting survival and growth of FAK-expressing tumours. I show that immune evasion requires FAK’s catalytic activity, and a small molecule FAK kinase inhibitor, VS-4718, which is currently in clinical development, drives depletion of Tregs and permits CD8+ T-cell-mediated tumour clearance. It is therefore likely that FAK inhibitors may trigger immune-mediated tumour regression, providing previously unrecognized therapeutic benefit.
2

Integrin alphα5/fibronectin1 and focal adhesion kinase are required for lens fiber morphogenesis in zebrafish

Hayes, Julie Marie 17 December 2010 (has links)
Fibronectin (fn) and integrin α5 (itgα5) are both key players in cell adhesion and intracellar signaling, however the specific in vivo role of these proteins has never been analyzed in the vertebrate lens. The results presented here indicate that Fn1 and Itgα5 proteins are essential for the proper development of the lens. The loss of Fn1 protein in the zebrafish embryo results in distinct adhesion defects, defects in lens fiber morphogenesis, and cataracts. These results were phenocopied in zebrafish itga5 mutants, thereby indicating an essential role for Fn1 and Itgα5 during lens development. Furthermore, embryos with reduced levels of ptk2.1 (focal adhesion kinase – FAK) also phenocopied the defective fn1 and itgα5 lens, suggesting that FAK is a major player in the intracellular signaling mediated by Fn1/Itgα5 interactions in the lens. / text
3

Characterizing ErbB2-induced mammary tumourigenesis

Oliver, Joseph James 18 September 2007 (has links)
Approximately 30% of human breast cancers demonstrate overexpression of the receptor tyrosine kinase ErbB2/HER2/Neu, with these cases correlating with recurrence and poor prognosis. While therapy targeting ErbB2 has met with some success, particularly in early-stage breast cancers, transformation and progression towards a later-stage metastatic phenotype is likely sustained by aberrant signaling from additional players, for instance that downstream of integrins. In fact, treatment with integrin-blocking antibodies and ß1-integrin ablation leads to reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo. Moreover, ErbB2 has recently been found to interact with the ß4-integrin subunit to promote tumour formation and progression, as deletion of the ß4-integrin signaling domain led to suppression of mammary tumour onset and invasive growth, coupled with decreases in ErbB2-dependent signaling. ErbB2 may interact with integrin subunits by direct binding or via the intracellular kinases Src and focal adhesion kinase (FAK), both known to be activated downstream of ErbB2 and integrins and having well-established roles in cell adhesion, migration, and invasion. Using an inducible model of ErbB2 activation, we have demonstrated that controlled ErbB2 activation in human mammary epithelial cells leads to phosphorylation of Src Tyr215 and FAK Tyr861, consistent with a recently published clinical study examining phosphorylated forms of Src and FAK in ErbB2-positive human breast tumour samples. We have also confirmed that ErbB2 activation increases the capacity of cells for survival: Normally, MCF10A human mammary epithelial cells cultured in three-dimensional, laminin-rich extracellular matrix gel form mammary acini-like spheroids with hollow lumen surrounded by a single layer of polarized epithelial cells. However, ErbB2 activation prevents luminal clearance and induces luminal filling in acini formed in three-dimensional culture, and leads to activation of Akt, a known survival signal. Taken together these data indicate a potential role for ErbB2 at the apex of cell survival signaling via Src, FAK, and Akt, contributing to luminal cell survival in three-dimensional culture. We have thus confirmed Src, FAK, and Akt as potential players in early onset of breast cancer, and targeting these signaling players concurrently with ErbB2 may prove effective, especially in early stage breast cancers. / Thesis (Master, Pathology & Molecular Medicine) -- Queen's University, 2007-08-31 16:25:59.917
4

Mechanismen der sensiblen Mechanotransduktion in Kardiomyozyten

Schreiber, Anna 14 November 2016 (has links) (PDF)
Die vorgelegte Dissertationsschrift dient der Identifizierung des Mechanismus, über welchen neonatale Kardiomyozyten der Ratte biaxialen zyklischen Stretch wahrnehmen. Angriffspunkt dafür bildeten sowohl Stretch Activated Ion Channels (SAIC) als auch die mit Integrinen assoziierte Focal Adhesion Kinase (FAK). Die Inhibition der SAIC erfolgte mit Gadolinium und eine Blockade der FAK konnte durch den FAK-Inhibitor II erreicht werden. Eigens angelegte Zellkulturen wurden unter definierten Parametern für 24 Stunden unter Einwirkung genannter Stoffe gestretcht und anschließend einer Analyse mittels Immunfluoreszenz und Western Blot unterzogen. Gestretchte Kardiomyozyten richteten sich schräg zur Achse der einwirkenden Kraft aus und wiesen eine Polarisierung von Connexin 43 auf, außerdem zeigte sich dessen gesteigerte Expression. Durch die Blockade der FAK konnte lediglich eine aufgehobene Polarisierung von Connexin 43 bei unveränderter Expression und Ausrichtung der Kardiomyozyten festgestellt werden. Auch die Mikrotubuli veränderten nach Stretch ihre Orientierung bezogen auf die Zellachse. Die zunächst annähernde Parallelität der Fasern zeigte sich nach Inhibition der FAK deutlich aufgelockert. Für die Aktinfilamente konnte dies nicht nachgewiesen werden. Die Integrine dienen demnach der Wahrnehmung von Stretch und vermitteln die Polarisierung von Connexin 43 sowie die Orientierung der Mikrotubuli über die FAK. Für die anderen genannten Prozesse ist die Aktivierung eines FAK-unabhängigen Integrin-Signalweges denkbar. Gadolinium hatte insgesamt keinen Effekt auf beschriebene Veränderungen, sodass ein Einfluss der SAIC auf Stretch-induzierte Veränderungen in Kardiomyozyten ausgeschlossen werden konnte. Gleichzeitig konnte durch die Beobachtung der Polarisierung von Microtubule Organizing Center, Kinesin und Golgi-Apparat die Hypothese der sich durch Stretch orientierenden neonatalen Kardiomyozyte unterstützt werden, welche die Voraussetzung für die Anordnung von Connexin 43 an den Zellpolen darstellen könnte. Sowohl die Selbstorganisation des Myokards im Rahmen der Embryogenese als auch die Pathophysiologie verschiedener kardialer Erkrankungen könnte dadurch womöglich besser verstanden werden.
5

Estímulo por soro em fibroblastos quiescentes induz a fosforilação da miosina-Va e sua localização em adesões focais / Serum by stimulation in quiescent fibroblasts induces phosphorylation of myosin - Va and its location in focal adhenosis

Zenzen, Johnny Alex Rockenbach 11 March 2016 (has links)
A montagem e desmontagem das adesões focais (AF) desempenham um papel fundamental em diversos processos celulares, incluindo migração celular e sobrevivência. Resultados prévios do nosso laboratório mostram que fibroblastos nulos ou silenciados para miosina-Va sofrem um atraso na desmontagem das adesões, sugerindo um papel para a miosinaVa neste processo. Neste trabalho, visamos analisar a dinâmica de montagem das AF em fibroblastos murinos imortalizados NIH3T3, utilizando sondas fluorescentes para visualização de componentes de adesão focal. A formação das AF foi analisada após estímulo por soro de células quiescentes, o que leva a intensa polimerização de actina, reorganização do citoesqueleto e montagem das AF. A cinética de montagem das AF foi observada em ensaios ao longo do tempo, de células fixadas em 0, 5, 15, 30, 120 minutos após estímulo, e marcadas para miosina-Va fosforilada (p-miosina-Va, S1650), FAK fosforilada (p-FAK, Y397), vinculina, dinamina-2, integrina-?1, faloidina, Ki67 e DAPI. Os nossos resultados mostraram um aumento de fluorescência de p-miosina-Va por todo o citoplasma após a estímulo com soro, e revelaram que a p-miosina-Va co-localiza com pFAK nas AF logo após o estímulo, essa localização da p-miosina-Va nas AF diminui ao passar do tempo e retorna após 120 minutos. Isto é consistente com os resultados anteriores de um papel da miosina-Va na dinâmica das AF. Também é possível perceber uma maior concentração de p-miosina-Va e dinamina-2 na região perinuclear, 5 minutos após estímulo, e o espalhamento de ambas as proteínas pelo citoplasma com o passar do tempo. Demonstramos, por Western blotting, que o estímulo por soro não causa alteração na quantidade total de miosina-Va em nenhum dos tempos analisados em relação à condição de quiescência, mas induz, após 5 e 15 minutos, um aumento apreciável de p-miosina-Va, que sofre queda e variações nos tempos posteriores. Para nosso conhecimento, esta é a primeira demonstração de que a fosforilação da miosina-Va aumenta em resposta ao soro e estamos investigando se este evento está ligado à dinâmica das adesões focais em fibroblastos / The assembly and disassembly of focal adhesions (FA) play a critical role in several cellular process, including cell migration and survival. Previous work from our laboratory showed that fibroblasts without myosin-Va show a delay in focal adhesion disassembly, suggesting a role for myosin-Va in this process. In this work, we aim at imaging the dynamics of focal adhesion disassembly and reassembly in cells, with fluorescent probes for visualization of focal adhesion components. Here, we used murine NIH3T3 fibroblasts to analyze FA formation after serum stimulation of quiescent cells, which leads to intense polymerization of actin and reorganization of the cytoskeleton and FA assembly. The kinetics of FA assembly was observed in a time-course assay of cells fixed at 0, 5, 15, 30 and 120 min after serum stimulation, and stained for phosphorylated myosin-Va (p-myosin-Va, S1650), phosphorylated FAK (p-FAK, Y397), vinculin, phalloidin and DAPI. Our results showed an increase of pmyosin-Va staining throughout the cytoplasm upon serum stimulation, and revealed that pmyosin-Va does not colocalize with FAK in FA at early time points. However, colocalization is observed after 30 to 120 min. This is consistent with previous results of a role for myosin-Va in FA disassembly. It is also possible to observe a higher concentration of p-myosin-Va and dynamin-2 in the perinuclear region 5 minutes after stimulation, and the spreading of both proteins in the cytoplasm over time. We demonstrate by Western blotting that serum stimulation does not cause change in total amount of myosin-Va, in any of the times analyzed in relation to the quiescent condition, but induces, after 5 and 15 minutes, an appreciable increase of pmyosin-Va suffering drop and variations in the later times. To our knowledge, this is the first demonstration that phosphorylation of myosin-Va increases in response to serum and we are investigating whether this event is connected to the dynamics of focal adhesions in fibroblasts
6

Rôle et régulation biochimique de la Protéine HEF1 et de sa phosphorylation sur sérine dans les tumeurs colorectales / Role and Biochemical Regulation of HEF1 Protein and its Serine Phosphorylation in Colorectal Cancer

Ibrahim, Rama 15 October 2014 (has links)
HEF1 (Human Enhancer of Filamentation 1) est une protéine d’ancrage multi-domaine de la famille CAS. Ces protéines sont impliquées dans des modifications du cytosquelette et sont des médiateurs de l’activation par les intégrines des cascades de signalisation au niveau des adhérences focales. Des données récentes ont mis en évidence une expression élevée de HEF1 associée à une capacité de migration et d’invasion cellulaire accrue dans plusieurs types de cancer. Ainsi, un rôle de la protéine dans la progression tumorale et le développement métastatique a été suggéré. HEF1 est une phosphoprotéine. Outre sa phosphorylation sur résidus tyrosine par les kinases FAK et Src bien décrite, elle est également phosphorylée sur des sérines: la sérine 296 et la sérine 369 identifiée par notre laboratoire comme étant un régulateur de la dégradation protéosomale de la protéine. A ce jour, peu d’informations sont disponibles sur le rôle de HEF1 dans la progression tumorale colorectale, ou sur l’implication de sa phosphorylation sur sérine dans ses fonctions biologiques.Mon travail de thèse a permis de montrer une régulation positive de l’expression de HEF1 dans des échantillons du cancer de côlon, associée à une augmentation de la migration cellulaire dans des lignées tumorale colorectales. L’induction de la migration de la lignée HCT116 lors de la surexpression de HEF1 est corrélée à une augmentation de la phosphorylation sur tyrosine de FAK de manière dépendante de la kinase Src. Dans un deuxième temps, nous nous sommes intéressé à la phosphorylation sur sérine de HEF1, et avons montré que, comparativement à la protéine sauvage, la mutation sur la Sér-369 amplifie son effet pro-migratoire ainsi que la phosphorylation de FAK régulée par HEF1 consécutivement à une stabilisation de la protéine. Nous avons également caractérisé, pour la première fois, une mutation fonctionnelle de HEF1 sur l’Arg-367 qui mime l'effet de la mutation sur la Sér-369. Enfin, nous avons identifié des protéines adaptatrices de la famille BCAR3 comme partenaires de HEF1 et démontré que l’effet pro-migratoire de HEF1 est requiert son interaction avec BCAR3. En conclusion, ce travail nous a permis d’identifier une mutation de HEF1 (R367Q) qui stabilise la protéine, accroissant ainsi son effet pro-migratoire. Nous avons également associé la migration induite par HEF1 à son interaction avec BCAR3 et à une augmentation de la phosphorylation sur tyrosine de FAK. / Human Enhancer of Filamentation 1 (HEF1) is a member of the p130Cas family of docking proteins. HEF1 is present at focal adhesions and is involved in integrin-mediated cytoskeleton reorganization associated with cell migration. Elevated expression of HEF1 promotes invasion and metastasis in multiple cancer cell types. To date, little is known on the role of HEF1 in CRC tumor progression. HEF1 is phosphorylated on several Ser/Thr residues but the effects of these post-translational modifications on the functions of HEF1 are poorly understood. In this manuscript, we investigated the role of HEF1 in colorectal adeno-carcinoma migration capacities. First, we found that HEF1 expression was augmented in high stages CRC samples and then showed that overexpression of HEF1 in colo-carcinoma cell line HCT116 increases cell migration. Moreover, in these cells, HEF1 increases Src-mediated phosphorylation of FAK. We then showed that HEF1 mutation on Ser-369 enhances HEF1-induced migration and FAK phosphorylation as a result of protein stabilization. We also, for the first time characterized a functional mutation of HEF1 on Arg-367 which mimics the effect of Ser-369 to Ala mutation. Finally through mass spectrometry experiments, we identified BCAR3 as an essential interactor and mediator of HEF1-induced migration. We demonstrated that single amino acid mutations that prevent formation of the HEF1-BCAR3 complex impair HEF1-mediated migration. Therefore, amino-acid substitutions that prevent Ser-369 phosphorylation stabilize HEF1 which increases the migration of CRC cells and this requires the interaction of HEF1 with the NSP family adaptor protein BCAR3. Collectively, these data reveal the importance of HEF1 expression level in cancer cell motility and then support the utilization of HEF1 as a biomarker of tumor progression.
7

Contrôle de l'invasion par la protéine kinase C thêta dans les cancers du sein / Control of breast cancer invasion by the protein kinase C theta

Chadelle, Lucie 16 November 2017 (has links)
Entre 15 et 20% des cancers du sein diagnostiqués sont des cancers du sein triple-négatifs (CSTN). Ce sous-type de cancer du sein est caractérisé par l'absence ou le faible niveau d'expression du récepteur au facteur de croissance épidermique de type 2 (HER2) et des récepteurs hormonaux à l'œstrogène et à la progestérone. Par définition, les patientes atteintes de CSTN ne peuvent bénéficier des traitements antihormonaux ou des thérapies ciblées anti-HER2 qui ont nettement amélioré la prise en charge thérapeutique des autres sous-types de cancers du sein. En marge de ces progrès, les CSTN sont ainsi principalement traités par chimiothérapies cytotoxiques, des thérapies ne parvenant pas toujours à empêcher leur dissémination métastatique. Par conséquent, les CSTN sont aujourd'hui associés à des pronostics relativement mauvais et l'identification de nouvelles cibles thérapeutiques constitue un enjeu majeur de la recherche sur le cancer du sein. C'est dans ce contexte qu'une cible thérapeutique potentielle contre les CSTN a récemment été identifiée: la sérine-thréonine kinase PKC thêta. Cette PKC nouvelle est fortement exprimée dans les CSTN alors qu'elle ne l'est pas, ou très faiblement, dans des cancers du sein exprimant le récepteur aux œstrogènes et dans les tissus mammaires non transformés. L'objectif de ma thèse a été d'étudier la fonction de PKC thêta dans le contrôle de l'invasion de cellules tumorales mammaires, une étape clé de la formation de métastases. Nos travaux montrent qu'une inhibition de PKC thêta aboutit à une nette diminution des capacités invasives de lignées de cellules CSTN in vitro. De même, in vivo cette inhibition limite fortement la formation de métastases chez la souris. Nous identifions le mécanisme moléculaire par lequel PKC thêta contrôle l'invasion: PKC thêta est capable d'activer la voie des adhérences focales en phosphorylant directement la kinase des adhérences focales (FAK) sur des sites de phosphorylations encore jamais identifiés, les sérines 892 et 893. Ces phosphorylations sont essentielles aux effets positifs de PKC thêta sur l'invasion et la FAK phosphorylée de la sorte est retrouvée spécifiquement au front de cellules CSTN en migration. De façon intéressante, ces phosphorylations de FAK par PKC thêta permettent une modification de la dynamique de formation des adhérences cellule/matrice ainsi que celle des protrusions. Le contrôle de ces protrusions passe très certainement par une altération de la dynamique d'activité des RhoGTPases induite par PKC thêta De surcroît, l'utilisation d'une PKC thêta activable par la rapamycine nous permet de finement étudier la temporalité des effets de PKC thêta sur la génération des protrusions et des adhérences cellule/matrice. Enfin, concernant le contrôle de l'activité de PKC thêta en amont, nous constatons que son activation de même que ses effets sur la voie FAK et l'invasion dépendent entièrement de CDCP1 (Cub Domain-Containing Protein 1), un récepteur transmembranaire associé à l'agressivité de plusieurs cancers, dont les CSTN. Mes travaux mettent ainsi en évidence un mécanisme inédit de contrôle de la voie FAK permettant l'invasion de cellules tumorales mammaires. De plus, ils valident PKC thêta en tant que cible thérapeutique potentielle dont l'inhibition pourrait permettre de limiter la dissémination métastatique des CSTN et ce sans effets secondaires majeurs, la fonction physiologique de PKCthêta étant non essentielle. / Triple-negative breast cancer is a subtype of breast cancer that primarily affects women under the age of 40. TNBCs account for 15 to 20% of all diagnosed breast cancers and are defined by tumour cells lacking or weakly expressing the oestrogen receptor, the progesterone receptor and the human epidermal growth factor receptor 2 (HER2). By definition, these cancers cannot benefit from hormonal therapies or targeted therapies against the HER2 that have both proved to be very efficient in other breast cancer subtypes. Hence, therapies against TNBC are based essentially on classical cytotoxic chemotherapies that are not always able to block metastatic dissemination of those cancers. As a consequence, TNBCs are associated with poor prognosis and thus, finding new therapeutic targets for TNBC constitutes a major challenge for breast cancer research. In that context, a new potential TNBC therepeutical target has recently been identified: the serine threonine PKC theta. This novel PKC is highly expressed in TNBC whereas it is not expressed in breast cancer expressing the oestrogen receptor or non-transformed mammary tissues. The aim of my PhD was to study the function of PKC theta in the control of breast cancer cells invasion, a key step to metastasis formation that can be defined as the ability of cells to migrate through an extracellular matrix. We have shown that PKC theta inhibition leads to strong diminution of the invasive abilities of TNBC cell lines in vitro. Accordingly, we show in vivo that PKC theta inhibition strongly impairs metastasis formation in mice. We have identified the molecular mechanisms through which PKC theta controls invasion: PKC theta is able to activate focal adhesion signalling by directly phosphorylating FAK (Focal Adhesion Kinase) at newly identified phosphorylation sites, serines 892 and 893. We observe that this phosphorylated FAK localizes to nascent adhesions at the leading edge of migrating breast cancer cells and that those phosphorylations are essential to PKC theta control of invasion. PKC theta phosphorylations of FAK also modify the dynamic of cell/matrix adhesions and protrusion formation. The effects on protrusion formation are most certainly linked to PKC theta alteration of Rho GTPases activity dynamic. Furthermore, the use of an activatable PKC theta enables us to precisely study the temporality of PKC theta effects on both protrusions and adhesions. Protrusions and adhesions being essential to cell migration, those results explain PKC theta positive effects on invasion. Finally, regarding the upstream control of PKC theta we observe that PKC theta activation, together with its effect on FAK and invasion, depend on CDCP1, a transmembrane receptor that has been linked to the aggressiveness of several cancers, including TNBC. Altogether, my work reveals a new mechanism of FAK pathway activation leading to breast cancer cell invasion. Moreover, it further defines PKC theta as an interesting therepeutical target, whose inhibition could limit metastatic dissemination of TNBC without major secondary effects, as PKC theta has a non-essential physiological function.
8

Estímulo por soro em fibroblastos quiescentes induz a fosforilação da miosina-Va e sua localização em adesões focais / Serum by stimulation in quiescent fibroblasts induces phosphorylation of myosin - Va and its location in focal adhenosis

Johnny Alex Rockenbach Zenzen 11 March 2016 (has links)
A montagem e desmontagem das adesões focais (AF) desempenham um papel fundamental em diversos processos celulares, incluindo migração celular e sobrevivência. Resultados prévios do nosso laboratório mostram que fibroblastos nulos ou silenciados para miosina-Va sofrem um atraso na desmontagem das adesões, sugerindo um papel para a miosinaVa neste processo. Neste trabalho, visamos analisar a dinâmica de montagem das AF em fibroblastos murinos imortalizados NIH3T3, utilizando sondas fluorescentes para visualização de componentes de adesão focal. A formação das AF foi analisada após estímulo por soro de células quiescentes, o que leva a intensa polimerização de actina, reorganização do citoesqueleto e montagem das AF. A cinética de montagem das AF foi observada em ensaios ao longo do tempo, de células fixadas em 0, 5, 15, 30, 120 minutos após estímulo, e marcadas para miosina-Va fosforilada (p-miosina-Va, S1650), FAK fosforilada (p-FAK, Y397), vinculina, dinamina-2, integrina-?1, faloidina, Ki67 e DAPI. Os nossos resultados mostraram um aumento de fluorescência de p-miosina-Va por todo o citoplasma após a estímulo com soro, e revelaram que a p-miosina-Va co-localiza com pFAK nas AF logo após o estímulo, essa localização da p-miosina-Va nas AF diminui ao passar do tempo e retorna após 120 minutos. Isto é consistente com os resultados anteriores de um papel da miosina-Va na dinâmica das AF. Também é possível perceber uma maior concentração de p-miosina-Va e dinamina-2 na região perinuclear, 5 minutos após estímulo, e o espalhamento de ambas as proteínas pelo citoplasma com o passar do tempo. Demonstramos, por Western blotting, que o estímulo por soro não causa alteração na quantidade total de miosina-Va em nenhum dos tempos analisados em relação à condição de quiescência, mas induz, após 5 e 15 minutos, um aumento apreciável de p-miosina-Va, que sofre queda e variações nos tempos posteriores. Para nosso conhecimento, esta é a primeira demonstração de que a fosforilação da miosina-Va aumenta em resposta ao soro e estamos investigando se este evento está ligado à dinâmica das adesões focais em fibroblastos / The assembly and disassembly of focal adhesions (FA) play a critical role in several cellular process, including cell migration and survival. Previous work from our laboratory showed that fibroblasts without myosin-Va show a delay in focal adhesion disassembly, suggesting a role for myosin-Va in this process. In this work, we aim at imaging the dynamics of focal adhesion disassembly and reassembly in cells, with fluorescent probes for visualization of focal adhesion components. Here, we used murine NIH3T3 fibroblasts to analyze FA formation after serum stimulation of quiescent cells, which leads to intense polymerization of actin and reorganization of the cytoskeleton and FA assembly. The kinetics of FA assembly was observed in a time-course assay of cells fixed at 0, 5, 15, 30 and 120 min after serum stimulation, and stained for phosphorylated myosin-Va (p-myosin-Va, S1650), phosphorylated FAK (p-FAK, Y397), vinculin, phalloidin and DAPI. Our results showed an increase of pmyosin-Va staining throughout the cytoplasm upon serum stimulation, and revealed that pmyosin-Va does not colocalize with FAK in FA at early time points. However, colocalization is observed after 30 to 120 min. This is consistent with previous results of a role for myosin-Va in FA disassembly. It is also possible to observe a higher concentration of p-myosin-Va and dynamin-2 in the perinuclear region 5 minutes after stimulation, and the spreading of both proteins in the cytoplasm over time. We demonstrate by Western blotting that serum stimulation does not cause change in total amount of myosin-Va, in any of the times analyzed in relation to the quiescent condition, but induces, after 5 and 15 minutes, an appreciable increase of pmyosin-Va suffering drop and variations in the later times. To our knowledge, this is the first demonstration that phosphorylation of myosin-Va increases in response to serum and we are investigating whether this event is connected to the dynamics of focal adhesions in fibroblasts
9

Understanding how focal adhesion proteins sense and respond to mechanical signals

Stutchbury, Benjamin January 2016 (has links)
The mechanical properties of the tissue vary widely around the body, from the soft brain to the rigid bone. Tissue cells are able to sense mechanical signals from their environment, which influence many aspects of cell behaviour such as migration, proliferation and differentiation. Focal adhesions (FAs) are large protein complexes that form the bridge between the extracellular matrix (ECM)-binding integrins and the contractile actin cytoskeleton. Here, they sense the rigidity of the local environment and translate this information into a cellular response, a process known as mechanotransduction. However, the FA proteins required for mechanotransduction, and the molecular mechanisms involved in this fundamental process, remain to be elucidated. Talin, vinculin, FAK and paxillin are four core FA-associated proteins that are thought to be involved in mechanotransduction. These proteins associate and dissociate from the complex in a constant state of flux. Using a live-cell imaging approach, I found that the rate of dynamic exchange of an FA protein correlates to its function. The FA appears to have a modular organisation; the slowest proteins have a structural role, such as talin and vinculin, responsible for directly linking integrin to actin and sensing the ECM stiffness. The signalling proteins are turned over more rapidly, including FAK and paxillin, and are responsible for directing the cellular response to force-generated signals from the ECM.The second results chapter focused on the force-dependent interactions between talin, vinculin and actin. The talin domains R2R3 were identified as the key mechanosensitive vinculin-binding sites, which are exposed upon the application of force across the talin rod. Vinculin binding to R2R3 led to actin associating with the central actin-binding site in the talin rod (ABS2), which is required for the transmission of actomyosin tension onto the underlying substrate as cellular traction force. Finally, the protein turnover data were incorporated into two mathematical models, describing talin and vinculin turnover, which were able to simulate the dynamic exchange of various talin and vinculin mutants in response to changing ECM stiffness. Using these models, the talin ABS2-actin and vinculin tail-actin interactions were found to be extremely important for sensing the stiffness of the ECM. These findings significantly increase our knowledge of the molecular mechanisms underpinning cellular mechanotransduction. Increased understanding of how mechanical signals are sensed and interpreted by the cell could lead to a number of novel therapies for a wide range of associated diseases, such as atherosclerosis, muscular dystrophy and cancer.
10

Σηματοδοτικά πολυπρωτεϊνικά σύμπλοκα ρυθμίζουν την μεταγωγή μηνυμάτων κατά την κυτταροφαγία των αιμοκυττάρων της μύγας της Μεσογείου. Ο ρυθμιστικός ρόλος της FAK και η συμμετοχή των ιντεγκρινών, των MAPCs και άλλων σηματοδοτικών μορίων / Multiprotein signal transduction complexes regulate the phagocytosis of E.coli in Medfly hemocytes suspension. The role of FAK and the participation of integrins, PI3K, ERK, ELK-1 and other cytoskeletal and regulatory proteins

Φερτάκης, Βασίλειος 29 June 2007 (has links)
Η κινάση εστιακής προσκόλλησης (FAK) είναι μια πρωτεϊνική κινάση τυροσίνης (nRPTK) με κυτταροπλασματική κατανομή στην περιφέρεια του κυττάρου. Εντοπίζεται κυρίως σε περιοχές του κυττάρου γνωστές ως εστίες προσκόλλησης, με τις οποίες το κύτταρο προσκολλάται στην εξωκυτταρική ύλη. Συμμετέχει σε σημαντικές κυτταρικές διεργασίες όπως στην κυτταρική κίνηση και μετανάστευση, στον κυτταρικό πολλαπλασιασμό, στην κυτταρική επιβίωση και απόπτωση. Η FAK ενεργοποιείται από πολλά ερεθίσματα που μπορούν να επάγουν φωσφορυλίωση της σε αμινοξέα τυροσίνης (Υ397, Υ576, Υ577, Υ861, Υ925) όπως για παράδειγμα αυξητικοί παράγοντες, νευροπεπτίδια, παράγοντες που ενεργοποιούν υποδοχείς συνδεδεμένους με G-πρωτεΐνες και μηχανικά ερεθίσματα αλλά κυρίως μετά από τη διέγερση των ιντεγκρινικών υποδοχέων των κυττάρων. Ο ρόλος της FAK στην μεταγωγή μηνυμάτων, στο πλαίσιο της κυτταρικής επικοινωνίας είναι διττός: δρα ως κινάση φωσφορυλιώνοντας τα υποστρώματα της αλλά και ως πρωτεΐνη συνδετήρας (adaptor protein) δημιουργώντας πολυπρωτεϊνικά σηματοδοτικά σύμπλοκα. Δεδομένης, λοιπόν, της ιδιαίτερης φύσης του μορίου, η FAK θα μπορούσε να χαρακτηριστεί ως ενεργοποιούμενη πρωτεΐνη ικρίωμα (activable scaffold protein). Στην παρούσα εργασία μελετήσαμε την ενεργοποίηση της FAK κατά την κυτταροφαγία του βακτηρίου E. coli. Έτσι, χρησιμοποιήθηκαν φαρμακολογικοί αναστολείς για να ανιχνευθεί η συμμετοχή τελεστών της μεταγωγής σημάτων στην ενεργοποίηση της FAK. Επιπλέον, διερευνήθηκε με ανοσοκατακρημνίσεις η συμπλοκοποίηση της FAK με πρωτεΐνες-κλειδιά σημαντικών κυτταρικών μονοπατιών σε κύτταρα που βρίσκονταν σε εναιώρημα. / Cells respond to extracellular stimuli with the recruitment of multiple cytoskeletal and regulatory proteins, which form specialized complexes. These complexes are usually involved in the promotion of integrin-mediated signals to the nucleus. In this study, we investigated whether any signal transduction complexes are constitutively present, in Medfly hemocyte suspension, in the absence of external stimuli. Hemocytes from the 3rd instar larvae were isolated and protein crude extracts were prepared. In hemocyte lysates, the presence of FAK (focal adhesion kinase), Integrin β1 and the adaptor protein Paxillin was identified with immunoprecipitation and immunoblot analysis. The presence of these proteins was also confirmed with immunofluorescence microscopy, in attached hemocytes on glass slides. Co-immunoprecipitation with FAK and immunoblot analysis with anti-Paxillin, anti-tubulin, anti-actin and anti-ERK revealed the complex formation of FAK with Paxillin, Tubulin, Actin and ERK in hemocyte suspensions. The profiles of this complex and of each one of these proteins, separately, varies, during development and phagocytosis of E.coli. Consequently, it was demonstrated that FAK forms complex with cytoskeletal proteins like paxillin, tubulin, actin and signalling proteins such as ERK, in Medfly hemocytes, without the influence of any extracellular stimuli.

Page generated in 0.4103 seconds