• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 3
  • 1
  • 1
  • Tagged with
  • 12
  • 12
  • 6
  • 5
  • 5
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 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

In-vitro- und in-vivo-Hemmung des AKT-Signalweges in Melanomzellen durch einen neuen kleinmolekularen Inhibitor / In-vitro- and in-vivo-inhibition of the AKT-pathway in melanoma by a novel small-molecule inhibitor

Schneider, Philine 05 February 2013 (has links)
Konstitutiv aktivierte Signalwege sind verantwortlich für die malignen Veränderungen in Melanozyten, die zur Entstehung des Melanoms beitragen. Im Mittelpunkt dieser Veränderungen stehen der PI3Kinase-AKT-Signalweg und der MAPK-ERK-Signalweg als wichtige Schlüsselwege in der Zellzykluskontrolle. Daher zielen viele neue Therapieversuche im Kampf gegen das Melanom auf die Kontrolle und Regulation dieser Wege. In dieser vorliegenden Arbeit wurden erstmals die Effekte eines neuen PI3K-Inhibitors, BAY-80-6946, und Wortmannin alleine und in Kombinationsbehandlungen mit den MEK1/2-Inhibitoren PD98059 und U0126 in vitro und in vivo untersucht. Zunächst wurden humane Melanomzellen auf konstitutiv aktivierte Signalwege in vitro per Western Blot untersucht und ihre Wachstumsraten im Mausmodell ermittelt. Die humane Melanomzelllinie LOX zeigte eine hohe konstitutive Expression von aktiviertem AKT und ERK, während A375 nur eine geringe Aktivität dieser beiden Signalwege aufwies. Dennoch besaßen diese beiden Zelllinien ein großes Wachstumspotential im Mausmodell im Vergleich zu anderen getesteten Zelllinien. A375 und LOX wurden in Zellkulturexperimenten mit den PI3Kinase-Inhibitoren Bay-80-6946 und Wortmannin sowie den MEK1/2-Inhibitoren PD98059 und U0126 behandelt und Tumor-relevante Zellfunktionen wie Proliferation und Apoptose gemessen. Die Zelllinien zeigten ein unterschiedliches Ansprechen auf die verschiedenen Inhibitoren und keine der Behandlungen wies eine zufriedenstellendes Ergebnis hinsichtlich der Anti-tumoralen Funktion auf. Bei dualer PI3Kinase- und MEK1/2-Hemmung zeigten sich jedoch deutliche synergistische Effekte, so dass diese Behandlungsform einen vielversprechenden Ansatz im Kampf gegen das Melanom darstellt.
2

The scaffold protein KSR1, a novel therapeutic target for the treatment of Merlin-deficient tumors

Zhou, L., Lyons-Rimmer, J., Ammoun, S., Muller, Jurgen, Lasonder, E., Sharma, V., Ercolano, E., Hilton, D., Taiwo, I., Barczyk, M., Hanemann, C.O. 09 November 2015 (has links)
Yes / Merlin has broad tumor-suppressor functions as its mutations have been identified in multiple benign tumors and malignant cancers. In all schwannomas, the majority of meningiomas and 1/3 of ependymomas Merlin loss is causative. In neurofibromatosis type 2, a dominantly inherited tumor disease because of the loss of Merlin, patients suffer from multiple nervous system tumors and die on average around age 40. Chemotherapy is not effective and tumor localization and multiplicity make surgery and radiosurgery challenging and morbidity is often considerable. Thus, a new therapeutic approach is needed for these tumors. Using a primary human in vitro model for Merlin-deficient tumors, we report that the Ras/Raf/mitogen-activated protein, extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) scaffold, kinase suppressor of Ras 1 (KSR1), has a vital role in promoting schwannomas development. We show that KSR1 overexpression is involved in many pathological phenotypes caused by Merlin loss, namely multipolar morphology, enhanced cell-matrix adhesion, focal adhesion and, most importantly, increased proliferation and survival. Our data demonstrate that KSR1 has a wider role than MEK1/2 in the development of schwannomas because adhesion is more dependent on KSR1 than MEK1/2. Immunoprecipitation analysis reveals that KSR1 is a novel binding partner of Merlin, which suppresses KSR1's function by inhibiting the binding between KSR1 and c-Raf. Our proteomic analysis also demonstrates that KSR1 interacts with several Merlin downstream effectors, including E3 ubiquitin ligase CRL4(DCAF1). Further functional studies suggests that KSR1 and DCAF1 may co-operate to regulate schwannomas formation. Taken together, these findings suggest that KSR1 serves as a potential therapeutic target for Merlin-deficient tumors.
3

Rôle de PP2A dans l'activation constitutive de MEK1/2 de cellules MDCK transformées par le virus du sarcome de Moloney

Guérard, Karl-Philippe January 2007 (has links)
Mémoire numérisé par la Division de la gestion de documents et des archives de l'Université de Montréal.
4

Rôle de PP2A dans l'activation constitutive de MEK1/2 de cellules MDCK transformées par le virus du sarcome de Moloney

Guérard, Karl-Philippe January 2007 (has links)
Mémoire numérisé par la Division de la gestion de documents et des archives de l'Université de Montréal
5

Genomweite Suche neuer Modulatoren der Signaltransduktion in kardialer Hypertrophie und Herzinsuffizienz / Genome wide cDNA library screen for new signaling associated modulators of cardiac hypertrophy and heart failure

Kramann, Nadine 18 January 2011 (has links)
No description available.
6

Implication de MEK1 et MEK2 dans l'initiation et la progression du cancer colorectal

Duhamel, Stéphanie 08 1900 (has links)
Une dérégulation de la voie de signalisation Ras/Raf/MEK/ERK1/2 est observée dans plus de 30% des cancers et des mutations activatrices de RAS sont observées dans 30% à 50% des adénomes colorectaux. À la suite d’une analyse extensive de biopsies de tumeurs colorectales humaines par micromatrices tissulaires (TMA), nous avons observé que 44% des tissus cancéreux exprimaient MEK1/2 phosphorylés, contre 10% des tissus normaux. L'analyse des TMA a également révélé que 79% des tumeurs arboraient un marquage nucléaire de MEK1/2 phosphorylés, contre 4 % pour les tissus normaux. Bien que la voie MEK/ERK1/2 soit fréquemment activée dans les cancers, le rôle précis des isoformes de MEK1 et de MEK2 n'a jamais été clairement établie. De même, l'impact de cette localisation nucléaire aberrante de phospho-MEK1/2, dans l'initiation et la progression des cancers colorectaux, est inconnu. Lors d'un premier projet, nous avons démontré, que l’expression de MEK1 ou MEK2 activé est suffisante pour transformer in vitro des cellules intestinales épithéliales de rat (IEC-6). L'expression des mutants actifs de MEK1 ou MEK2 est suffisante pour induire une dérégulation de la prolifération cellulaire et engendrer la formation d'adénocarcinomes invasifs dans un modèle de greffe orthotopique du côlon chez la souris. Nous avons également démontré que l'inhibition de MEK2 par shRNA supprime complètement la prolifération des lignées humaines de cancer du côlon, alors que la suppression de MEK1 a peu d'effet sur la capacité de prolifération. Le deuxième projet, nous a permis d'observer que l'expression d'un mutant nucléaire de MEK1 dans les cellules IEC-6 transforme drastiquement les cellules. Une augmentation de prolifération, une résistance à l'anoikose, un dérèglement du cycle cellulaire, de l'instabilité chromosomique (CIN), de la tétra/aneuploïdie sont observés. La caractérisation des mécanismes responsables de cette localisation aberrante de MEK1/2 phosphorylés, a permis d'identifier la protéine Sef, un régulateur de la localisation cytoplasmique de MEK/ERK1/2. Nous avons démontré que l'expression d'une forme oncogénique de Ras (H-RasV12) inhibe l'expression de Sef, engendrant alors une accumulation nucléaire de MEK1/2 activés. Plus encore, la réexpression de Sef restaure la localisation cytoplasmique de MEK1/2 et renverse les propriétés tumorigéniques ainsi que l'aneuploïdie induite par Ras activé. Un troisième projet, visant la caractérisation des mécanismes associés à la CIN et à l'aneuploïde engendrés par l'activation aberrante de la voie de Ras-ERK1/2, a permis d'observer que l'hyperactivation de ERK1/2 induit des anomalies mitotiques menant à la binucléation. Une localisation erronée et une surexpression de la kinase Aurora A, de même que des protéines de passage du complexe chromosomique (CPC), Aurora B, Survivine et INCENP, sont observées. L'inhibition partielle de l'activation de ERK1/2 par de faible dose de PD184352, un inhibiteur de MEK1/2, est suffisante pour renverser la surexpression de ces régulateurs mitotiques, de même que corriger les anomalies de la mitose et réduire la tétra/aneuploïdie engendrée par Ras oncogénique. Ainsi, nous avons démontré, pour la première fois, que la voie des MAP kinases ERK1/2 est impliquée dans la CIN, la tétraploïdie et l'aneuploïdie. Nos résultats suggèrent que la perte de Sef est un événement oncogénique précoce, qui contribue à la localisation nucléaire aberrante de MEK1/2 qui est observée dans les tumeurs colorectales. Cette localisation anormale de MEK1/2 est associée à l'initiation de la transformation, la progression tumorale et la CIN, via l'activité soutenue de ERK1/2. Ces informations sont capitales et démontrent l’importance de la voie de signalisation Ras/Raf/MEK/ERK1/2 dans le processus de tumorigénèse colorectale. / The Ras-dependent Raf/MEK/ERK1/2 signaling pathway is frequently hyperactivated in human cancer as a result of receptor tyrosine kinase overexpression or gain-of-function mutations in RAS or RAF genes. More specificaly, activating mutation in RAS genes are found in ~ 30-50% of colorectal adenomas and phosphorylation of ERK1/2 is frequently observed in human colorectal cancer cells and tumor specimens. In a large TMA analysis, we found that MEK1/MEK2 are aberrantly activated in 44% of human colorectal cancers. In addition, our analysis revealed that 79% of colorectal cancers exhibit aberrant phospho-MEK1/2 staining in the nucleus, as compared to 4% of normal tissue. How dysregulation and mislocalization of MEK1/2 contribute to tumor initiation and progression is not well understood. In order to determine the exact contribution of MEK1 and MEK2 to the pathogenesis of colorectal cancer, wild type and constitutively active forms of MEK1 and MEK2 were ectopically expressed by retroviral gene transfer in the normal intestinal epithelial cell line IEC-6. We found that the expression of activated MEK1 or MEK2 is sufficient to morphologically transform intestinal epithelial cells, dysregulate cell proliferation and induce the formation of high-grade adenocarcinomas after orthotopic transplantation in mice. A large proportion of these intestinal tumors metastasize to the liver and lung. Importantly, we show that silencing of MEK2 expression completely suppresses the proliferation of human colon carcinoma cell lines, whereas inactivation of MEK1 has a much weaker effect. In a second project, we have investigated the impact of the nuclear mislocalization of phosphorylated MEK1/2 observed in colorectal tumors. We show that oncogenic activation of Ras is sufficient to induce the nuclear accumulation of phosphorylated MEK1/2 and ERK1/2 in intestinal epithelial cells. To evaluate the biological impact of the mislocalization of MEK1/2, we have forced the localization of MEK1 in the nucleus of epithelial cells. We found that sustained nuclear MEK1 signaling leads to hyperactivation of ERK1/2 and to enhanced cell proliferation. Nuclear localization of MEK1 also leads to tetraploidization, chromosomal instability (CIN) and tumorigenesis. Importantly, we show that oncogenic Ras downregulates the spatial regulator Sef, concomitant to nuclear accumulation of activated MEK1/2. Moreover, re-expression of Sef is sufficient to restore the normal localization of MEK1/2 and to revert the cell cycle defects and tumorigenesis induced by oncogenic Ras. Another project was initiated to characterize the tetraploidy and CIN observed upon hyperactivation of the Ras-ERK1/2 pathway. Aneuploidy and CIN are observed in the majority of colorectal cancers and are associated with a poorer prognosis. We show that hyperactivation of ERK1/2 by oncogenic Ras or sustained nuclear MEK-ERK1/2 signaling induces mitotic defects that lead to tetraploidy, aneuploidy and CIN. We also found that dysregulation of Ras-ERK1/2 signaling alters the expression and localization of Aurora A and the Chromosomal passenger complex proteins. In conclusion, we show for the first time that the MEK/ERK1/2 signaling pathway is implicated in aneuploidy and CIN. Our results suggest that sustained nuclear ERK1/2 signaling may contribute to the initiation and progression of colorectal cancer by rapidly inducing aneuploidy and CIN. We suggest that loss of Sef is an early oncogenic event that contributes to genetic instability and tumor progression by sustaining nuclear ERK1/2 signaling. These observations are significant and highlight the importance of the Ras-ERK1/2 signaling pathway in colorectal tumorigenesis.
7

Use of an ex vivo model of human colorectal tumours to study response to the MEK1/2 inhibitor AZD6244

Novo, Sonia Marisa January 2013 (has links)
Colorectal cancer is the second most common cause of cancer death in Western Europe and North America. Current therapies are largely ineffective and are associated with considerable morbidity. Activating mutations in KRAS and BRAF genes are frequent in colorectal cancer, especially at later stages of the disease, and result in constitutive activity of the MAPK pathway, leading to increased proliferation and tumour survival. The MEK1/2 inhibitor AZD6244, that targets the MAPK pathway downstream of these mutations, has been tested as novel therapy for colorectal cancer. However, clinical trials have been disappointing due to an apparent intrinsic and/or acquired resistance to treatment. Mechanisms underlying this resistance have been studied using cell lines and tumour xenografts. However, the relevance of these data to advanced human colorectal cancer is unclear. One of the difficulties in testing and developing novel therapies for colorectal cancer is the lack of representative models of human disease. Thus, the initial aim of my PhD was to develop a method to culture human colorectal cancers ex vivo in order to use this as a platform for investigating response to AZD6244 and other therapies. These studies indicated that regardless of growth conditions, colonic tumour explants suffered extensive apoptosis in the first 24h in culture, which limited their application in drug response assays. Therefore, as an alternative to long term culture of human colorectal explants, I tested the effects of AZD6244 using acute treatments. Twenty three fresh colonic tumours were obtained from patients and treated for 1h with AZD6244 ex vivo in dose response studies. In all samples, MEK1/2 inhibition occurred within 1h of treatment. In one group of particularly sensitive tumours, the drug also had a distinct phenotypic effect. In these tumours, I found that the agent induced a dose-dependent decrease in proliferation and increase in apoptosis within 1h of treatment. Analysis of markers for this sensitivity indicated it was not clearly dependent of the presence of KRAS or BRAF mutations, which have previously been shown to confer sensitivity. Other markers of sensitivity / resistance were also examined. In addition to studies with AZD6244 alone, I examined the combined effects of this agent and aspirin in colon cancer cells lines and in tumour explants, with promising results. Whilst the use of fresh patient tumour tissue has some technical and logistical challenges, these data suggest that such methodologies are worthy of further investigation as a means to examine determinants of sensitivity and resistance to novel therapies, or their likely activity in combination.
8

Differential regulation of the EMT axis by MEK1/2 and MEK5 in triple-negative breast cancer

January 2016 (has links)
acase@tulane.edu / Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) whereby cells adopt a motile and invasive phenotype through loss of epithelial markers, namely Cadherin 1/E-Cadherin (CDH1), and acquisition of mesenchymal markers, such as vimentin (VIM) and Cadherin 2/N-Cadherin (CDH2). While MAPK/ERK1/2 kinase inhibitors (MEKi) have shown promise as antitumor agents in the preclinical setting, application has had limited success clinically. Activation of compensatory signaling, potentially contributing to the emergence of drug resistance, has shifted the therapeutic strategy to combine MEK1/2 inhibitors with agents targeting oncoproteins (RAF) or parallel growth pathways (PI3K). Conventional MAPK family members have been well-characterized in modulation of cellular processes involved in tumor initiation and progression, yet the role of MEK5-ERK5 in cancer biology is not completely understood. Recent studies have highlighted the importance of the MEK5 pathway in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast. Furthermore, elevated levels of ERK5 expression and activity observed in breast carcinomas are linked to worse prognosis in TNBC patients. The purpose of this work is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide rationale for combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance. / 1 / Van Hoang
9

Implication de MEK1 et MEK2 dans l'initiation et la progression du cancer colorectal

Duhamel, Stéphanie 08 1900 (has links)
Une dérégulation de la voie de signalisation Ras/Raf/MEK/ERK1/2 est observée dans plus de 30% des cancers et des mutations activatrices de RAS sont observées dans 30% à 50% des adénomes colorectaux. À la suite d’une analyse extensive de biopsies de tumeurs colorectales humaines par micromatrices tissulaires (TMA), nous avons observé que 44% des tissus cancéreux exprimaient MEK1/2 phosphorylés, contre 10% des tissus normaux. L'analyse des TMA a également révélé que 79% des tumeurs arboraient un marquage nucléaire de MEK1/2 phosphorylés, contre 4 % pour les tissus normaux. Bien que la voie MEK/ERK1/2 soit fréquemment activée dans les cancers, le rôle précis des isoformes de MEK1 et de MEK2 n'a jamais été clairement établie. De même, l'impact de cette localisation nucléaire aberrante de phospho-MEK1/2, dans l'initiation et la progression des cancers colorectaux, est inconnu. Lors d'un premier projet, nous avons démontré, que l’expression de MEK1 ou MEK2 activé est suffisante pour transformer in vitro des cellules intestinales épithéliales de rat (IEC-6). L'expression des mutants actifs de MEK1 ou MEK2 est suffisante pour induire une dérégulation de la prolifération cellulaire et engendrer la formation d'adénocarcinomes invasifs dans un modèle de greffe orthotopique du côlon chez la souris. Nous avons également démontré que l'inhibition de MEK2 par shRNA supprime complètement la prolifération des lignées humaines de cancer du côlon, alors que la suppression de MEK1 a peu d'effet sur la capacité de prolifération. Le deuxième projet, nous a permis d'observer que l'expression d'un mutant nucléaire de MEK1 dans les cellules IEC-6 transforme drastiquement les cellules. Une augmentation de prolifération, une résistance à l'anoikose, un dérèglement du cycle cellulaire, de l'instabilité chromosomique (CIN), de la tétra/aneuploïdie sont observés. La caractérisation des mécanismes responsables de cette localisation aberrante de MEK1/2 phosphorylés, a permis d'identifier la protéine Sef, un régulateur de la localisation cytoplasmique de MEK/ERK1/2. Nous avons démontré que l'expression d'une forme oncogénique de Ras (H-RasV12) inhibe l'expression de Sef, engendrant alors une accumulation nucléaire de MEK1/2 activés. Plus encore, la réexpression de Sef restaure la localisation cytoplasmique de MEK1/2 et renverse les propriétés tumorigéniques ainsi que l'aneuploïdie induite par Ras activé. Un troisième projet, visant la caractérisation des mécanismes associés à la CIN et à l'aneuploïde engendrés par l'activation aberrante de la voie de Ras-ERK1/2, a permis d'observer que l'hyperactivation de ERK1/2 induit des anomalies mitotiques menant à la binucléation. Une localisation erronée et une surexpression de la kinase Aurora A, de même que des protéines de passage du complexe chromosomique (CPC), Aurora B, Survivine et INCENP, sont observées. L'inhibition partielle de l'activation de ERK1/2 par de faible dose de PD184352, un inhibiteur de MEK1/2, est suffisante pour renverser la surexpression de ces régulateurs mitotiques, de même que corriger les anomalies de la mitose et réduire la tétra/aneuploïdie engendrée par Ras oncogénique. Ainsi, nous avons démontré, pour la première fois, que la voie des MAP kinases ERK1/2 est impliquée dans la CIN, la tétraploïdie et l'aneuploïdie. Nos résultats suggèrent que la perte de Sef est un événement oncogénique précoce, qui contribue à la localisation nucléaire aberrante de MEK1/2 qui est observée dans les tumeurs colorectales. Cette localisation anormale de MEK1/2 est associée à l'initiation de la transformation, la progression tumorale et la CIN, via l'activité soutenue de ERK1/2. Ces informations sont capitales et démontrent l’importance de la voie de signalisation Ras/Raf/MEK/ERK1/2 dans le processus de tumorigénèse colorectale. / The Ras-dependent Raf/MEK/ERK1/2 signaling pathway is frequently hyperactivated in human cancer as a result of receptor tyrosine kinase overexpression or gain-of-function mutations in RAS or RAF genes. More specificaly, activating mutation in RAS genes are found in ~ 30-50% of colorectal adenomas and phosphorylation of ERK1/2 is frequently observed in human colorectal cancer cells and tumor specimens. In a large TMA analysis, we found that MEK1/MEK2 are aberrantly activated in 44% of human colorectal cancers. In addition, our analysis revealed that 79% of colorectal cancers exhibit aberrant phospho-MEK1/2 staining in the nucleus, as compared to 4% of normal tissue. How dysregulation and mislocalization of MEK1/2 contribute to tumor initiation and progression is not well understood. In order to determine the exact contribution of MEK1 and MEK2 to the pathogenesis of colorectal cancer, wild type and constitutively active forms of MEK1 and MEK2 were ectopically expressed by retroviral gene transfer in the normal intestinal epithelial cell line IEC-6. We found that the expression of activated MEK1 or MEK2 is sufficient to morphologically transform intestinal epithelial cells, dysregulate cell proliferation and induce the formation of high-grade adenocarcinomas after orthotopic transplantation in mice. A large proportion of these intestinal tumors metastasize to the liver and lung. Importantly, we show that silencing of MEK2 expression completely suppresses the proliferation of human colon carcinoma cell lines, whereas inactivation of MEK1 has a much weaker effect. In a second project, we have investigated the impact of the nuclear mislocalization of phosphorylated MEK1/2 observed in colorectal tumors. We show that oncogenic activation of Ras is sufficient to induce the nuclear accumulation of phosphorylated MEK1/2 and ERK1/2 in intestinal epithelial cells. To evaluate the biological impact of the mislocalization of MEK1/2, we have forced the localization of MEK1 in the nucleus of epithelial cells. We found that sustained nuclear MEK1 signaling leads to hyperactivation of ERK1/2 and to enhanced cell proliferation. Nuclear localization of MEK1 also leads to tetraploidization, chromosomal instability (CIN) and tumorigenesis. Importantly, we show that oncogenic Ras downregulates the spatial regulator Sef, concomitant to nuclear accumulation of activated MEK1/2. Moreover, re-expression of Sef is sufficient to restore the normal localization of MEK1/2 and to revert the cell cycle defects and tumorigenesis induced by oncogenic Ras. Another project was initiated to characterize the tetraploidy and CIN observed upon hyperactivation of the Ras-ERK1/2 pathway. Aneuploidy and CIN are observed in the majority of colorectal cancers and are associated with a poorer prognosis. We show that hyperactivation of ERK1/2 by oncogenic Ras or sustained nuclear MEK-ERK1/2 signaling induces mitotic defects that lead to tetraploidy, aneuploidy and CIN. We also found that dysregulation of Ras-ERK1/2 signaling alters the expression and localization of Aurora A and the Chromosomal passenger complex proteins. In conclusion, we show for the first time that the MEK/ERK1/2 signaling pathway is implicated in aneuploidy and CIN. Our results suggest that sustained nuclear ERK1/2 signaling may contribute to the initiation and progression of colorectal cancer by rapidly inducing aneuploidy and CIN. We suggest that loss of Sef is an early oncogenic event that contributes to genetic instability and tumor progression by sustaining nuclear ERK1/2 signaling. These observations are significant and highlight the importance of the Ras-ERK1/2 signaling pathway in colorectal tumorigenesis.
10

Microglial activation decreases retention of the protease inhibitor saquinavir: implications for HIV treatment

Dallas, Shannon, Block, Michelle, Thompson, Deborah, Bonini, Marcelo, Ronaldson, Patrick, Bendayan, Reina, Miller, David January 2013 (has links)
BACKGROUND:Active HIV infection within the central nervous system (CNS) is confined primarily to microglia. The glial cell compartment acts as a viral reservoir behind the blood-brain barrier. It provides an additional roadblock to effective pharmacological treatment via expression of multiple drug efflux transporters, including P-glycoprotein. HIV/AIDS patients frequently suffer bacterial and viral co-infections, leading to deregulation of glial cell function and release of pro-inflammatory mediators including cytokines, chemokines, and nitric oxide.METHODS:To better define the role of inflammation in decreased HIV drug accumulation into CNS targets, accumulation of the antiretroviral saquinavir was examined in purified cultures of rodent microglia exposed to the prototypical inflammatory mediator lipopolysaccharide (LPS).RESULTS:3H]-Saquinavir accumulation by microglia was rapid, and was increased up to two-fold in the presence of the specific P-glycoprotein inhibitor, PSC833. After six or 24 hours of exposure to 10 ng/ml LPS, saquinavir accumulation was decreased by up to 45%. LPS did not directly inhibit saquinavir transport, and did not affect P-glycoprotein protein expression. LPS exposure did not alter RNA and/or protein expression of other transporters including multidrug resistance-associated protein 1 and several solute carrier uptake transporters.CONCLUSIONS:The decrease in saquinavir accumulation in microglia following treatment with LPS is likely multi-factorial, since drug accumulation was attenuated by inhibitors of NF-kappabeta and the MEK1/2 pathway in the microglia cell line HAPI, and in primary microglia cultures from toll-like receptor 4 deficient mice. These data provide new pharmacological insights into why microglia act as a difficult-to-treat viral sanctuary site.

Page generated in 0.414 seconds