• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 156
  • 52
  • 31
  • 12
  • 10
  • 8
  • 7
  • 7
  • 5
  • 3
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 335
  • 117
  • 54
  • 51
  • 42
  • 34
  • 32
  • 31
  • 29
  • 28
  • 28
  • 27
  • 26
  • 26
  • 24
  • 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.
61

Differential effects of epidermal growth factor receptor inhibitors on glioblastoma multiforme

Blazar, Ilyse Natasha 08 April 2016 (has links)
OBJECTIVE: Glioblastoma Multiforme (GBM), one of the most malignant forms of primary brain tumors, is characterized by its highly heterogenous genetic composition, aggressive infiltration of surrounding tissue, and resistance to current treatments. Gene expression analysis has characterized GBM into four main types, with a significant portion belonging to the Classical subtype, typified by overexpression and/or mutation of the epidermal growth factor receptor (EGFR). Also common to this subtype of GBM is the loss of crucial tumor suppressor genes Ink4A/ARF and PTEN, which contribute to the invasive nature and unregulated proliferation that underlie the GBM pathology. The high rate of tumor recurrence post treatment with surgical resection, chemotherapy, and radiation has driven the pursuit of more effective molecularly targeted therapies. This study was undertaken to determine the effects of two types of small molecule tyrosine kinase inhibitors on cells overexpressing wild-type EGFR in the context of their respective complements of tumor suppressor genes. METHODS: Several cell lines were established from mouse models of EGFR wild-type (EGFRWT) driven gliomagenesis and treated with 10 μM of type I tyrosine kinase inhibitors Gefitinib (Iressa®, Astra Zeneca), CI-1033 (Canertinib, Pfizer), or Dimethyl Sulfoxide vehicle. Cells were exposed to each drug treatment as part of a time course ranging from 0 to 24 hours and then evaluated by trypan blue exclusion and Western blot analysis for cell viability and molecular and biochemical effects respectively. RESULTS: Evaluation of cell viability indicated that CI-1033 caused a greater increase in cell death than gefitinib when compared to control treated cells regardless of the tumor suppressors lost. Gefitinib was found to cause cell death only in cells expressing the PTEN tumor suppressor whereas CI-1033 showed similar levels of cell death for cells deficient in Ink4A/ARF or both Ink4A/ARF and PTEN tumor suppressors. Western blot analysis revealed that CI-1033 more effectively inhibited EGFR compared to gefitinib. Treatment with both gefitinib and CI-1033 effectively blocked phosphorylation of EGFR, but this effect was less pronounced with gefitinib treatment. Further analysis of downstream signaling molecules showed a greater presence of cleaved caspase 3, a hallmark of apoptosis, in gefitinib treated cells expressing PTEN than in those cells treated with CI-1033. Cells deficient in both Ink4A/ARF and PTEN did not demonstrate any induction of cleaved caspase 3 following either treatment. CONCLUSIONS: Based on the significant differences in cell viability between treatments, CI-1033 is an overall more effective inhibitor of EGFRWT expressing cells lacking PTEN, while gefitinib and CI-1033 were found to be similarly effective in cells expressing PTEN. The results of western blot analysis indicate that total and irreversible EGFR inhibition may be necessary to induce cell death in a manner that effectively terminates downstream cell signaling. It is likely that CI-1033, unlike gefitinib, induces apoptosis in a caspase-independent manner, which may be one of the many differences in downstream effects produced by these two drugs. Further research is necessary to determine the extent to which each inhibitor shuts down proliferative cell signaling pathways such as PI3K-AKT and MEK-ERK signaling pathways downstream of EGFR. Overall, these data indicate that genotype plays an important role in the determination of therapeutic response and may aid in the evaluation of clinical prognoses.
62

Skillnaden i progressionfri överlevnad för patinter som behandlades med reversibla jämfört med ickereversibla EGFR-kinas-inhibitorer under cancerbehandling.

Kathier, Somaya January 2019 (has links)
Bakgrund: Lungcancer är den femte vanligaste cancerformen i Sverige, i vilken cirka 3650 personer dör varje år. Den ökar bland kvinnor och minskar bland män och den uppkommer vanligast i 70-årsåldern. Lungcancer orsaker mer än en miljon dödsfall varje år över hela världen där rökning utgör den största riskfaktorn. Lungcancer delas följaktligen in i två huvudgrupper baserad på cellform: småcellig och icke-småcellig lungcancer. Icke-småcellig lungcancer förkortas till NSCLC (non-small cell lung cancer) och utgör cirka 80% av all lungcancerfall. Syfte: Med detta arbete vill jag undersökaSkillnaden i progressionfri överlevnad för patinter som behandlades med reversibla jämfört med icke-reversibla EGFR-kinasinhibitorer under cancerbehandling. Metod: Arbetet har baserats på fem vetenskapliga artiklar som hämtades från databasen PubMed. I artiklarna undersöktes effektivitet och säkerhet hos reversibla och icke reversibla EGFR-tyrosinkinashämmare som har administrerats patienter drabbade av lungcancer med EGFR-mutationer. Resultat: Följaktligen visade det sig att icke- reversibel EGFR-tyrosinkinashämmaren osimertinib har en signifikant förbättring i progressionsfri överlevnad jämfört med reversibla EGFR-tyrosinkinashämmare gefitinib och erlotinib. Den progressionsfria medianöverlevnaden var längre vid användning av osimertinib än de reversibla hämmareerlotinib och gefitinib. Detta resulterade icirka 18 månader överlevnadvid användning av osimertinib och 10 månader vid användning av erlotinib och gefitinib. Slutsats:Jag visarhär att icke-reversibel EGFR-tyrosinkinashämmare osimertinibkan användas för patienter med icke-småcellig lungcancer NSCLC. Den har visat en signifikant förbättrad effekt som förstahandsbehandling jämfört med reversibla första generationens EGFR-hämmare gefitinib och erlotinib. Osimertinib är aktiv mot vanliga EGFR- mutationer, framför allt del-19 och L858R, och mot resistensmutationen T790M i exon 20 som bildas vid behandling med första generationens reversibla EGFR-kinasinhibitorer. / Background: Lung cancer is the fifth most common cancer type in Sweden, of which about 3650 people die every year. Its incidence increases among women and decreases among men and it is most common at the age of 70. Approximately there are more than one million deaths each year worldwide caused by lung cancer. Smoking is the most common risk factor. Lung cancer is divided into two main groups: small cell and non-small cell lung cancer. Non-small cell lung cancer is shortened to NSCLC accounts for about 80% of all lung cancers.  Purpose: The goal of this paper is to determine the difference in drug resistance for reversible compared to non-reversible EGFR kinase inhibitors during lung cancer treatment. Based on five scientific articles retrieved from the PubMed database, I investigated the efficacy and safety of reversible and non-reversible EGFR tyrosine kinase inhibitors in patients who have lung cancer with EGFR mutations.  Result: The results showed that the non-reversible EGFR tyrosine kinase inhibitor osimertinib doens't give a significant improvement in progression-free survival compared to reversible the EGFR tyrosine kinase inhibitors gefitinib and erlotinib. For osimertinib the median progression-free survival was longer when compared to the reversible inhibitors erlotinib and gefitinib: approximately 18 months in comparison to 10 months, respectively. Conclusion: The non-reversible EGFR tyrosine kinase inhibitors osimertinib may be preferably used in patients with non-small cell lung cancer NSCLC. It has shown an improved significant efficacy in the first-hand compared to reversible first-generation EGFR tyrosine kinase inhibitors (gefitinib, erlotinib). It is active against common EGFR mutations, especially del 19 and L858R, and against resistance mutation T790M in exon 20 generated during first generation treatment reversible EGFR kinase inhibitors erlotinib and gefitinib.
63

Mechanisms of the anti-proliferative actions of the schweinfurthins in cancer cells

Sheehy, Ryan Michael 01 May 2015 (has links)
Schweinfurthins are intriguing natural product chemotherapeutics due to their potent yet selective activity and their unknown mechanism of growth inhibition in cancer. Much progress has been made in characterizing the intracellular effects of the schweinfurthins since they were first isolated from Macaranga schweinfurthii in 1986. Here, the L-type calcium channel and P- glycoprotein (Pgp) inhibitor verapamil has been found to enhance schweinfurthin- induced growth inhibition. Verapamil induces an increase in the intracellular concentration of a fluorescent schweinfurthin. However, the synergistic relationship between the schweinfurthins and verapamil is complex and not obvious in that verapamil fails to increase the intracellular concentration of a schweinfurthin analogue that is a known substrate of Pgp. Schweinfurthins are also found to induce alterations to cholesterol homeostasis by increasing the expression of the cholesterol efflux pump ABCA1 in an apparent liver X receptor- independent fashion. In addition, schweinfurthin treatment blunts epidermal growth factor downstream activation and phosphorylation of Akt. Lastly, a schweinfurthin-resistant cell line has been created and characterized for resistance to schweinfurthin-induced growth inhibition. The variety of intracellular effects characteristic of schweinfurthin treatment described here provide mechanistic framework for identifying the potential target and mechanism of growth inhibition for the schweinfurthins.
64

Modulação do EGFR em complexos cumulus-oócitos bovinos cultivados in vitro e seus efeitos sobre o metabolismo, maturação e aquisição da competência oocitária e a transcrição gênica das células do cumulus e dos embriões /

Dall'Acqua, Priscila Chediek. January 2018 (has links)
Orientador: Gisele Zoccal Mingoti / Resumo: O receptor do fator de crescimento epidermal (EGFR) está relacionado com a retomada da meiose induzida por estímulo gonadotrófico. Nós utilizamos um inibidor do EGFR (AG1478) para inibir a retomada da meiose durante a pré-maturação (PMIV). No experimento I, foi avaliada a maturação oocitária, a expansão das células do cumulus, a funcionalidade das junções gap, a distribuição dos microfilamentos de actina e o conteúdo lipídico em oócitos e, a expressão gênica nas células do cumulus. No experimento II, foi avaliado o desenvolvimento embrionário, a taxa de eclosão, o número total de células, a taxa de apoptose, o conteúdo lipídico e expressão gênica nos embriões. No experimento I, os COC foram PMIV por 8h em meio com 1 μM de AG1478, inibidor do EGFR, (EGFR-_PreIVM), em seguida foram MIV por 22h (EGFR-_IVM); foram feitos 4 grupos controle: oócitos imaturos imediatamente após sua remoção do folículo (Control_Immat), oócitos MIV por 8h (Control_PreIVM), por 22h (Control_IVM_22h) e por 30h (Control_IVM_30h). No experimento II, os COC foram PMIV por 8h em meio com 1 μM do inibidor do EGFR, em seguida foram MIV por 22h (EGFR-); os COC do grupo controle foram MIV por 22h (Control). Em seguida, os oócitos foram FIV e cultivados por até 9 dias. No experimento I, o grupo EGFR-_PreIVM apresentou 53,1% dos oócitos em GV, com a maioria em GV1 (42,9%; P>0,05) ou em GV3 [28,6%, semelhante ao grupo Control_Immat (P>0,05) e menor que o grupo Control_PreIVM (P<0,05)]; após a MIV não foram encontr... (Resumo completo, clicar acesso eletrônico abaixo) / Abstract: Epidermal growth factor receptor (EGFR) is related with meiosis resumption by gonadotropic stimulus. We used an EGFR inhibitor (AG1478) to block meiosis resumption during prematuration (PIVM). In experiment I, was evaluated oocyte maturation, cumulus cells expansion, gap junctions functionality, microfilaments distribution and lipid content in oocytes and, gene expression in cumulus cells. In experiment II, was assessed embryo development, hatching rates, total cell number, apoptosis, lipid content and gene expression in embryos. For experiment I, COC were PIVM for 8h in medium supplemented with 1μM of AG1478, an EGFR inhibitor, (EGFR-_PreIVM), followed by 22h IVM (EGFR-_IVM); four control groups were evaluated: immature oocytes immediately after follicle removal (Control_Immat), oocytes IVM for 8h (Control_PreIVM), oocytes IVM for 22h (Control_IVM_22h) and oocytes IVM for 30h (Control_IVM_30h). For experiment II, COC were PIVM for 8h in medium supplemented with 1μM of EGFR inhibitor, followed by 22h IVM (EGFR-); COC from control group were matured for 22h (Control). After that, oocytes were IVF and zygotes were cultured up to 9 days. In experiment I, EGFR-_PreIVM group had 53.1% of the oocytes blocked in GV, the majority in GV1 (42.9%; P>0.05) or in GV3 [28.6%, similar to Control_Immat (P>0.05) and lower than Control_PreIVM (P<0.05)]. After IVM, no differences (P>0.05) were found between groups in MII rates, but Control_IMV_30h (9.1%) had higher (P<0.05) rates of degenerated... (Complete abstract click electronic access below) / Doutor
65

Caractérisation des cellules souches cancéreuses de la peau humaine : Implication de la voie de signalisation de l'Epidermal Growth Factor Receptor dans le contrôle de la différenciation des cellules souches de l'épiderme

Le Roy, Hélène 22 July 2009 (has links) (PDF)
L'épiderme humain est un tissu en renouvellement constant où l'équilibre entre les cellules nouvellement formées et les cellules perdues par desquamation est maintenu par la division asymétrique des cellules souches produisant deux cellules filles différentes : l'une possédant la capacité de s'auto-renouveler et l'autre engagée dans la différenciation en plusieurs lignées cellulaires reconstituant l'épiderme et ses appendices. De récentes découvertes suggèrent que les cellules souches altérées, appelées cellules initiatrices de tumeur ou cellules souches cancéreuses, seraient à l'origine du développement tumoral. Par conséquent, l'éradication ciblée de ces cellules pourrait traiter les cancers. Nos études avaient pour but d'identifier et de caractériser les cellules souches cancéreuses dans des tumeurs de peau et d'évaluer l'implication de la voie de signalisation de l'Epidermal Growth Factor Receptor (EGFR) dans le contrôle de leur auto-renouvellement et leur potentiel de différenciation. Nous avons trouvé de rare kératinocytes mitotiques avec une distribution asymétrique de l'EGFR et nous avons déterminé que sa présence à la surface était liée au destin normal ou cancéreux des cellules. Bien qu'essentiel pour la prolifération, la différenciation et la survie des cellules épithéliales, EGFR n'était pas présent à la surface des cellules aux propriétés de cellules souches comme la quiescence, leur compétence à produire des cellules filles fonctionnellement différentes, leurs potentiels de prolifération et de clonogénicité élevés, leur capacité à former des sphères et l'expression de marqueurs de cellules souches. Au contraire, les kératinocytes exposant l'EGFR ont acquis un phénotype plus différencié, démontrant que l'EGFR contrôle un basculement du compartiment de cellules souches à celui de cellules d'amplification transitoire. Ce basculement était associé avec des changements dans le profil d'expression de protéines du cycle cellulaire ou contrôlant la survie et la mitochondrie qui variaient entre les cellules normales et cancéreuses. A partir de nos résultats, nous proposons un modèle dans lequel EGFR fonctionne comme un déterminant du destin des kératinocytes qui équilibre entre la quiescence et la prolifération/différenciation des cellules souches épidermiques durant la mitose. Cet équilibre semble clairement mal fonctionner dans le cancer. Nous avons précédemment montré que la différenciation des kératinocytes partage certains aspects de la signalisation apoptotique et est stimulée par les ROS, nous proposons donc que l'acquisition de l'EGFR à la surface cellulaire active la prolifération cellulaire et par conséquent, la production des ROS. De façon importante, notre étude suggère fortement que les cellules EGFR négatives peuvent constituer le compartiment reproductif de la tumeur responsable du maintien et du développement tumoral, donc fournir un aperçu mécanistique de l'inefficacité des thérapies actuelles anti-EGFR.
66

A Short Thesis about Growth Factors in Gliomas

Hesselager, Göran January 2003 (has links)
<p>Glioblastoma multiforme (GBM) is the most common form of primary brain tumor in humans. Its aggressive and infiltrative growth into the brain, and, at best, only partial sensitivity to radiotherapy and chemotherapy, renders it extremely difficult to treat and survival remains dismal. </p><p>Growth factors, such as platelet-derived growth factor (PDGF) and epidermal growth factor (EGF), and their corresponding receptors are seen in glioma tissue, suggesting the presence of autocrine stimulatory loops. <i>PDGFB</i> and a mutated EGF receptor were also identified as cellular homologues of two oncoviruses, thereby indicating a role in tumorigenesis. This thesis presents a brain tumor model in mice, developed using a <i>PDGFB</i> coding retrovirus to induce overexpression of PDGF-B in neonatal mouse brain. Immature tumors, with histological characteristics of primary brain tumors developed at relatively high frequencies. Mice injected with a non-coding retrovirus did not develop tumors, indicating the crucial role of PDGF stimulation in this system. Tumor cells were also shown to continue to depend on PDGF stimulation when cultured <i>in vitro</i>. </p><p>In human glioblastomas, growth factor receptor signaling is present in conjunction with defects in cell cycle arrest pathways. When the <i>PDGFB</i>-virus model was used with <i>p53</i> or <i>Ink4a-Arf</i> deficient mice, tumors arose with shorter latency and higher frequency. Loss of <i>p53</i> or <i>Ink4a-Arf</i> seemed to facilitate signaling through the PI3K/Akt pathway. Thus, a functional role for the co-existence of p53 loss of function and PDGF signaling in a subset of gliomas is presented. </p><p>Human GBM samples were collected and analyzed with respect to expression and activation of the EGFR and PDGFRα. Most tumors expressed the both receptors at moderate to high levels, but high activation of either receptor seemed mutually exclusive.</p>
67

A Short Thesis about Growth Factors in Gliomas

Hesselager, Göran January 2003 (has links)
Glioblastoma multiforme (GBM) is the most common form of primary brain tumor in humans. Its aggressive and infiltrative growth into the brain, and, at best, only partial sensitivity to radiotherapy and chemotherapy, renders it extremely difficult to treat and survival remains dismal. Growth factors, such as platelet-derived growth factor (PDGF) and epidermal growth factor (EGF), and their corresponding receptors are seen in glioma tissue, suggesting the presence of autocrine stimulatory loops. PDGFB and a mutated EGF receptor were also identified as cellular homologues of two oncoviruses, thereby indicating a role in tumorigenesis. This thesis presents a brain tumor model in mice, developed using a PDGFB coding retrovirus to induce overexpression of PDGF-B in neonatal mouse brain. Immature tumors, with histological characteristics of primary brain tumors developed at relatively high frequencies. Mice injected with a non-coding retrovirus did not develop tumors, indicating the crucial role of PDGF stimulation in this system. Tumor cells were also shown to continue to depend on PDGF stimulation when cultured in vitro. In human glioblastomas, growth factor receptor signaling is present in conjunction with defects in cell cycle arrest pathways. When the PDGFB-virus model was used with p53 or Ink4a-Arf deficient mice, tumors arose with shorter latency and higher frequency. Loss of p53 or Ink4a-Arf seemed to facilitate signaling through the PI3K/Akt pathway. Thus, a functional role for the co-existence of p53 loss of function and PDGF signaling in a subset of gliomas is presented. Human GBM samples were collected and analyzed with respect to expression and activation of the EGFR and PDGFRα. Most tumors expressed the both receptors at moderate to high levels, but high activation of either receptor seemed mutually exclusive.
68

Studies of LRIG1 and the ERBB receptor family in breast and colorectal cancer

Ljuslinder, Ingrid January 2009 (has links)
The LRIG1 gene (leucine-rich repeats and immunoglobulin like domains-1) at chromosome 3p14 is a proposed tumour suppressor gene whose gene product negatively regulates various receptor tyrosine kinases. This function has been the basis for classifying LRIG1 as a potential tumour suppressor gene (TSG). The ERBB receptor family is important in malignant cellular functions such as proliferation, survival, adhesion, migration and differentiation. In breast cancer, amplification of the ERBB2 proto-oncogene is an important negative prognostic factor. The epidermal growth factor receptor (EGFR/ERBB1), is expressed in colorectal cancer and has been correlated to a worse prognosis. Until recently, immunohistochemical analysis of EGFR expression was used to select patients suitable for treatment with EGFR targeted antibodies. This thesis characterizes LRIG1 in breast and colorectal cancer to gain further knowledge of the gene and its expression. Also, the EGFR expression in metastases and the invasive margin of colorectal cancers was investigated to correlate changes to clinical factors. Breast cancer samples and matched normal tissues were evaluated for LRIG1 and the ERBB receptors at gene, RNA and protein levels. An increase in copy number of the LRIG1 gene was evident. Also, increased LRIG1 copy number was associated with high levels of ERBB2 mRNA. Another set of breast cancer tumours were analysed for LRIG1 by FISH analysis. The results were coherent with the previous results. To further analyze the correlation to ERBB2, tumours with LRIG1 increased copy number were analysed for ERBB2. The data showed that 89% of tumours with increased LRIG1 copy number were either ERBB2 amplified or had an increased copy number of ERBB2. To investigate LRIG1 and the EGFR in colorectal cancer, the gene and protein expression was analysed by several methods in tumours and corresponding normal tissues. There were no significant changes at gene level found, but at the protein level, both over- and under expression were seen. No evident correlation between LRIG1 and EGFR expression was detected. The ERBB receptor family expression in colorectal cancer tumours and corresponding metastases was investigated to explore if the expression was altered in the metastatic lesion. The results showed that the EGFR expression was lost in the corresponding metastases in 33% of the tumours and that the same percentage of tumours gained expression in the metastases. Co-expression of the ERBB family members was also analysed; there was a significant increase of ERBB3/ERBB4 co-expression in late stage tumours. EGFR expression at the invasive margin of colorectal cancers was analysed to clarify whether expression correlated to the patient’s prognosis. Significant correlation to survival and the presence of budding was seen. In conclusion, 34% of the breast cancer tumours studied had an increased copy number of LRIG1 with a significant co-incidental increase in ERBB2 copy number. This raises the question of a functional correlation between LRIG1 and ERBB2, a finding that might be of clinical importance. The studies of EGFR and the ERBB receptors in colorectal cancer reflect the heterogeneity of EGFR expression in tumours. In addition, these findings suggest that survival of the patients correlates to an increasing EGFR expression at the invasive margin.
69

Novel Methods for Analysis of Heterogeneous Protein-Cell Interactions : Resolving How the Epidermal Growth Factor Binds to Its Receptor

Björkelund, Hanna January 2013 (has links)
Cells are complex biological units with advanced signalling systems, a dynamic capacity to adapt to its environment, and the ability to divide and grow. In fact, they are of such high level of complexity that it has deemed extremely difficult or even impossible to completely understand cells as complete units. The search for comprehending the cell has instead been divided into small, relatively isolated research fields, in which simplified models are used to explain cell biology. The result produced through these reductionistic investigations is integral for our current description of biology. However, there comes a time when it is possible to go beyond such simplifications and investigate cell biology at a higher level of complexity. That time is now. This thesis describes the development of mathematical tools to investigate intricate biological systems, with focus on heterogeneous protein interactions. By the use of simulations, real-time measurements and kinetic fits, standard assays for specificity measurements and receptor quantification were scrutinized in order to find optimal experimental settings and reduce labour time as well as reagent cost. A novel analysis platform, called Interaction Map, was characterized and applied on several types of interactions. Interaction Map decomposes a time-resolved binding curve and presents information on the kinetics and magnitude of each interaction that contributed to the curve. This provides a greater understanding of parallel interactions involved in the same biological system, such as a cell. The heterogeneity of the epidermal growth factor receptor (EGFR) system was investigated with Interaction Map applied on data from the instrument LigandTracer, together with complementing manual assays. By further introducing disturbances to the system, such as tyrosine kinase inhibitors and variation in temperature, information was obtained about dimerization, internalization and degradation rates. In the long term, analysis of binding kinetics and combinations of parallel interactions can improve the understanding of complex biomolecular mechanisms in cells and may explain some of the differences observed between cell lines, medical treatments and groups of patients.
70

Scaffolding functions of MAGI-2 in the PTEN mediated attenuation of the PI3K/Akt signalling pathway

Poland, Sharon Franceska 24 September 2009
Activated receptor tyrosine kinase (RTK), such as the epidermal growth factor (EGF) receptor (EGFR) and the platelet-derived growth factor (PDGF) receptor (PDGFR), recruit downstream signalling proteins, including phosphatidylinositol 3-kinase (PI3K). PI3K, composed of a regulatory p85 subunit and a catalytic p110 subunit, phosphorylates phosphatidylinositol 4,5-bisphosphate at the 3 position to generate phosphatidylinositol 3,4,5-trisphosphate. This lipid second messenger activates Akt, which promotes cell growth, cell cycle entry and progression, as well as cell survival and cellular migration. PTEN, a tumor suppressor protein, dephosphorylates phosphatidylinositol 3,4,5-trisphosphate at the 3 position, turning off Akt signalling. PTEN contains a C-terminal PDZ binding motif that binds to the PDZ2 domain of MAGI-2, a scaffolding protein that localizes signalling molecules to the plasma membrane. MAGI-2 has ten domains that potentially mediate multiple protein-protein interactions simultaneously. A PTEN associated-complex (PAC) has been described and may contain MAGI-2, PTEN and p85. The PAC is hypothesized to form at the plasma membrane at appropriate sites for PTEN to gain access to its lipid substrates, since the binding of PTEN to MAGI-2 has been shown to enhance the suppression of PI3K-mediated Akt signalling. In order to better understand the role of the PAC in attenuation of the Akt signalling pathway, regions of the MAGI-2 scaffolding protein were mapped to identify the interactions taking place in the PAC. MAGI-2, and its individual domains, were expressed as GST fusion proteins. These were immobilized onto beads and allowed to bind to cellular proteins including PTEN, p85, PDGFR and EGFR using a GST pull-down experiment. The proteins bound to GST-MAGI-2 were identified using an immunoblot analysis. In vitro pull-down experiments revealed that MAGI-2 PDZ2 and PDZ5 domains bind to PTEN, and both MAGI-2 WW domains were shown to bind to p85. EGFR and PDGFR did not bind to the PDZ domains of MAGI-2 under the conditions studied. In order to study protein-protein interactions in cells, immunoprecipitation assays were also performed. Full length MAGI-2 was expressed tagged to a Myc epitope. This was used in immunoprecipitation assays to determine if MAGI-2 could co-immunoprecipitate with proteins involved in the Akt signalling pathway, such as PTEN, p85, PDGFR and EGFR. MAGI-2 can co-immunoprecipitate with PTEN upon 5 min EGF stimulation however, this result was inconclusive because replicate experiments did not verify this initial observation. MAGI-2 does not co-immunoprecipitate with the EGFR nor p85, under the conditions tested. We examined for these interactions after 5 min of growth factor stimulation and more experiments that test different time points after growth factor stimulation would reveal if these interactions are present at shorter time points. MAGI-2 has been shown to bind to PTEN and p85 in vitro and therefore has the potential to regulate the attenuation of the PI3K/Akt signalling pathway in response to activated EGFR and/or PDGFR.

Page generated in 0.0339 seconds