• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 81
  • 37
  • 14
  • 11
  • 5
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 175
  • 61
  • 49
  • 34
  • 24
  • 21
  • 20
  • 16
  • 16
  • 13
  • 13
  • 13
  • 12
  • 12
  • 12
  • 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.
31

CDK8 : une cible de la voie KRAS/MAP Kinase dans la carcinogénèse colorectale

Placet, Morgane January 2014 (has links)
La voie KRAS/BRAF/MEK/ERK MAP Kinase joue un rôle clé dans le contrôle de la prolifération des cellules épithéliales intestinales normales et cancéreuses. En effet, on retrouve des mutations du gène KRAS dans près de 35 à 40% des cancers colorectaux et une mutation du gène BRAF dans 10 à 15% des cas. Ces mutations de type gain-de-fonction sont mutuellement exclusives, ce qui suggère que la signalisation MEK/ERK qui est en aval de BRAF joue possiblement un rôle crucial dans le développement de plus de 60% des cancers colorectaux. Notre laboratoire a d’ailleurs rapporté que l’expression d’une forme mutante hyperactive de MEK1 est suffisante pour induire la transformation des cellules épithéliales intestinales normales en culture. Cette transformation est caractérisée par une transition épithélium-mésenchyme (EMT) conférant aux cellules des capacités tumorales, invasives et métastatiques. Afin de mieux comprendre les mécanismes moléculaires impliqués dans les effets transformant de MEK1, une analyse comparative par micropuces d’ADN (Affymetrix) a été effectuée et celle-ci a montré que le gène codant pour la protéine CDK8, une kinase dépendante des cyclines, est un des gènes les plus induits (12 fois) par l’hyperactivation de MEK1. Ce résultat suggèrerait l’implication de CDK8 dans l’oncogenèse colorectale induite par l’hyperactivation de la voie KRAS/MAP Kinase. De manière intéressante, nous avons d’abord mis en évidence que CDK8 était surexprimée dans des tumeurs de patients atteints de cancer colorectal de différents stades ainsi que dans des lignées cancéreuses colorectales humaines. Parmi ces lignées cellulaires analysées, nous avons mis en évidence que cette surexpression était en partie dépendante de l’activité MEK. Nous avons aussi confirmé la surexpression de CDK8 dans des lignées de cellules épithéliales intestinales de rat exprimant les oncogènes KRAS ou BRAF ou le mutant de MEK1 constitutivement actif. La baisse d’expression de CDK8 par l’utilisation d’un shARN a révélé que CDK8 contribue à l’hyperprolifération cellulaire ainsi qu’à la croissance en indépendance d’ancrage induite par l’expression du mutant hyperactif de MEK1. De plus, la baisse d’expression de CDK8 atténue le phénotype fibroblastique des cellules transformées par l’oncogène BRAF ou le mutant de MEK1 constitutivement actif, qui exhibent un phénotype plus épithélial. Nous avons pu mettre en évidence que CDK8 serait impliqué dans l’expression de gènes liés à la morphologie cellulaire tel que Snail1, Snail2 et Gem. Nos résultats montrent donc que CDK8 contribue au potentiel oncogénique de la voie MAP Kinase dans les cellules épithéliales intestinales en modulant leurs capacités prolifératives et leur transformation morphologique.
32

In Vitro Modeling of Pancreatic Duct Cell Carcinogenesis

Leung, Lisa 20 June 2014 (has links)
Pancreatic adenocarcinoma (PDAC) putatively arises from the pancreatic duct, thus usage of the normal human pancreatic duct epithelial (HPDE) cell line is an ideal model to examine the successive accumulation of genetic alterations involved in carcinogenesis. KRAS mutations have been reported in 90% of PDACs. Oncogenic KRAS elicits activation of downstream pathways involved in survival, motility, and cell cycle progression. KRASG12V introduction in the HPDE cell line upregulates Lipocalin-2 (LCN2) expression. LCN2 has been identified in numerous carcinomas and is associated with survival, tumorigenicity, and invasion. In this work, LCN2 was found to be commonly expressed in high grade pancreatic duct neoplastic precursor lesions and PDAC illustrating its potential as a biomarker. Moreover, in vitro and in vivo studies demonstrate that high LCN2 expression promotes gemcitabine resistance, MMP-9 activity, angiogenesis, and tumorigenicity. Loss of Smad4 function is found in 55% of PDAC cases. Smad4 is a critical component in the TGF-β signaling which mediates the transcription of genes involved in processes such as cell cycle arrest, apoptosis, and invasion. This work examined the consequences of KRASG12V expression and Smad4 loss in the HPDE model. Cellular invasion was promoted by KRASG12V expression or knocking down Smad4 by 80% in the HPDE model. A TGF-β resistant HPDE cell line, TβR, was shown to lack Smad4 expression due to deletion, promoter methylation, and nonsense mutation. KRASG12V expression in the TβR model (TβR KRAS) promoted neoplastic transformation and tumour formation in immunodeficient mice with complete penetrance. Smad4 expression in the TβR KRAS cell line reinstated TGF-β signaling, delayed tumour formation, and decreased metastatic spread. This study provides evidence that Smad4 acts as a restriction point in the transformation of HPDE cells. Overall, this work examines the contribution of genes involved in transformation, and identifies a potential therapeutic and diagnostic biomarker in PDAC.
33

Evaluation of Altered Kras Codon Bias and NOS Inhibition During Lung Tumorigenesis

Pershing, Nicole L. January 2014 (has links)
<p>The small GTPases <italic>HRAS, <italic>NRAS and <italic>KRAS are mutated in approximately one-third of all human cancers, rendering the proteins constitutively active and oncogenic. Lung cancer is the leading cause of cancer deaths worldwide, and more than 20% of human lung cancers harbor mutations in <italic>RAS, with 98% of those occurring in the <italic>KRAS isoform. While there have been many advances in the understanding of <italic>KRAS&ndash;driven lung tumorigenesis, it remains a therapeutic challenge. To further this understanding and assess novel approaches for treatment, I have investigated two aspects of <italic>Kras&ndash;driven tumorigenesis in the lung:</p><p>(<italic>I) Despite nearly identical protein sequences, the three <italic>RAS proto-oncogenes exhibit divergent codon usage. Of the three isoforms, <italic>KRAS contains the most rare codons resulting in lower levels of KRAS protein expression relative to <italic>HRAS and <italic>NRAS. To determine the consequences of rare codon bias during <italic>de <italic>novo tumorigenesis, we created a knock-in <italic>Kras<super>ex3op mouse in which synonymous mutations in exon 3 converted codons from rare to common. These mice had reduced tumor burden and fewer oncogenic mutations in the <italic>Kras<super>ex3op allele following carcinogen exposure. The reduction in tumorigenesis appeared to be a product of rare codons affecting both the oncogenic and non&ndash;oncogenic alleles. Converting rare codons to common codons yielded a more potent oncogenic allele that promoted growth arrest and enhanced tumor suppression by the non-oncogenic allele. Thus, rare codons play an integral role in <italic>Kras tumorigenesis.</p><p>(<italic>II) Lung cancer patients exhale higher levels of NO and <italic>iNOS<super>-/- mice are resistant to chemically induced lung tumorigenesis. I hypothesize that NO promotes <italic>Kras&ndash;driven lung adenocarcinoma, and NOS inhibition may decrease <italic>Kras&ndash;driven lung tumorigenesis. To test this hypothesis, I assessed efficacy of the NOS inhibitor L&ndash;NAME in a genetically engineered mouse model of <italic>Kras-driven lung adenocarcinoma. Adenoviral Cre recombinase was delivered into the lungs intranasally, resulting in expression of oncogenic <italic>Kras<super>G12D and dominant-negative <italic>Trp53<super>R172H in lung epithelial cells. L&ndash;NAME treatment was provided in the water and continued until survival endpoints. In this model, L&ndash;NAME treatment decreased tumor growth and prolonged survival. These data establish a potential clinical role for NOS inhibition in lung cancer treatment.</p> / Dissertation
34

The role of epithelial cell-derived tumour necrosis Factor Alpha in pancreatic carcinogenesis

Bossard, Maud January 2012 (has links)
Activating mutations of the kras proto-oncogene are found in more than 90% of human pancreatic ductal adenocarcinoma (PDAC) and can result in increased activity of the NF-κB pathway, leading to constitutive production of proinflammatory cytokines such as TNF-α. Pancreatic cancer progression occurs through a series of pre-invasive lesions, pancreatic intraepithelial neoplasias (PanIN lesions), which progress into invasive carcinoma. The aim of this thesis is to understand the autocrine role of TNF-α produced by premalignant epithelial cells in pancreatic tumour progression. This cytokine has already been shown to be involved in the progression of cancer. The major hypothesis therefore tested was that TNF-α secreted by pre-malignant epithelial cells promotes the early stages of pancreatic carcinogenesis by sustaining an inflamed microenvironment. In the spontaneous kras+/LSL-G12D; pdx1-cre mouse model of pancreatic cancer, concomitant genetic deletion of the TNF-α/IKK2 pathway substantially delayed pancreatic cancer progression and resulted in downregulation of the classical Notch target genes hes1 and hey1. Cell lines from the different PanIN bearing mice were established and used to dissect the cooperation between TNF-α/IKK2 and Notch signalling during PanIN progression. Optimal expression of Notch target genes was induced upon TNF-α stimulation of the canonical NF-κB signalling pathway, in cooperation with basal Notch signals. Mechanistically, TNF-α stimulation resulted in phosphorylation of histone H3 at the hes1 promoter and this signal was lost upon ikk2 genetic deletion. HES1 suppressed the expression of pparg, which encodes for the anti-inflammatory nuclear receptor PPAR-γ. Thus, crosstalk between TNF-α/IKK2 and Notch sustained an intrinsic inflammatory profile of the transformed cells. The treatment of PanIN bearing mice with rosiglitazone, a PPAR-γ agonist, also delayed PanIN progression. A malignant cell-autonomous, low-grade inflammatory process was shown to operate from the very early stages of kras-driven pancreatic carcinogenesis, which may cooperate with the Notch signalling pathway to promote pancreatic cancer progression.
35

STK38L kinase ablation promotes loss of cell viability in a subset of KRAS-dependent pancreatic cancer Cell lines

Grant, Trevor James 01 November 2017 (has links)
Pancreatic ductal adenocarcinomas (PDACs) are highly aggressive malignancies, associated with poor clinical prognosis and limited therapeutic options. The KRAS oncogene is mutated in over 90% of PDACs and plays a pivotal role in tumor progression. Global gene expression profiling of PDAC reveals 3-4 major molecular subtypes with distinct phenotypic traits and pharmacological vulnerabilities, including variations in oncogenic KRAS pathway dependencies. PDAC cell lines of the aberrantly differentiated endocrine exocrine (ADEX) subtype are robustly KRAS-dependent for survival. The KRAS gene is located on chromosome 12p11-12p12, a region amplified in 5-10% of primary PDACs. Within this amplicon, we identified co-amplification of KRAS with the STK38L gene in a subset of primary human PDACs and PDAC cell lines. This provided rationale to determine whether PDAC cell lines are dependent on STK38L expression for proliferation and viability. STK38L (also known as NDR2) encodes a nuclear Dbf2-related (NDR) serine/threonine kinase, which shares homology with Hippo pathway LATS1/2 kinases. We show that STK38L expression levels are elevated in a subset of primary PDACs and PDAC cell lines that display ADEX subtype characteristics, including overexpression of mutant KRAS. RNAi-mediated depletion of STK38L in a subset of ADEX subtype cell lines results in decreased cellular proliferation and increased apoptotic cell death. Concomitant with cytostatic and cytotoxic effects, STK38L depletion causes increased expression of the LATS2 kinase and the cell cycle regulator p21. LATS2 depletion partially rescues the cell proliferation and viability effects of STK38L depletion. Lastly, high STK38L mRNA expression is associated with worse patient prognosis compared to low STK38L expression in PDACs. Taken together, our study uncovers STK38L as a candidate, targetable vulnerability in a subset of molecularly defined PDACs. / 2019-11-01T00:00:00Z
36

Rôles des isoformes de PI3K p110α et p110ß dans l'initiation de la carcinogenèse pancréatique induite par Kras oncogénique / Role of Pl3K isoforms p110alpha and p110beta in Kras-induced pancreatic cancerogenesis

Baer, Romain 14 September 2015 (has links)
L'adénocarcinome pancréatique (PDAC) est un des cancers les plus létaux, tuant environ 95% des patients diagnostiqués à 5 ans. A l'heure actuelle, aucune chimiothérapie curative n'est disponible. La voie PI3K est une des voies les plus altérées dans les cancers. En particulier, dans la moitié des cancers pancréatiques, une augmentation d'activité de la voie PI3K/Akt/mTOR a été décrite et corrélée à un mauvais pronostic. Les enzymes de signalisation phosphoinositide-3-kinase (PI3K) de classe I sont présentes chez les mammifères sous plusieurs isoformes (p110a, p110ß p110d et p110y). Les isoformes p110a et p110ß sont ubiquitaires alors que p110d et p110y sont majoritairement exprimées dans les cellules immunitaires. Toutes les isoformes de classe I présentent un domaine d'activation par Ras, dont la fonctionnalité reste à être approfondie. Les PI3Ks de classe I phosphorylent le PIP2 en PIP3, un second messager lipidique capable de réguler des fonctions biologiques très variées telles que la prolifération ou la différenciation cellulaire. Bien que les rôles physiologiques des différentes isoformes de PI3K commencent à être connus, le débat actuel dans le domaine des PI3Ks est de savoir quelle isoforme cibler dans le traitement des cancers. Il a récemment été publié que la dépendance à la voie PI3K/Akt d'une tumeur est liée, selon l'organe et les altérations génétiques associées, à l'activité d'une isoforme. La mutation activatrice de l'oncogène KrasG12D est retrouvée dans plus de 90% des cas d'adénocarcinomes pancréatiques et est reconnue comme la mutation " initiatrice " du PDAC. Les souris qui présentent une activation constitutive de l'oncogène KrasG12D reproduisent toutes les étapes de la carcinogenèse pancréatique humaine, des lésions prénéoplasiques jusqu'à l'adénocarcinome (5% des souris à 12 mois). Partant des constats que les PI3K et Kras interagissent physiquement et que seules p110a et p110ß sont exprimées dans le pancréas sain, l'objectif de mon projet de thèse était de déterminer si les deux isoformes ubiquitaires de PI3K pouvaient jouer des rôles différents dans l'initiation de la carcinogenèse pancréatique induite par Kras muté. Pour cela, nous avons généré un nouveau modèle murin qui associe la mutation de l'oncogène KrasG12D avec une inactivation conditionnelle de l'activité kinase de p110a ou de p110ß, mimant ainsi l'utilisation d'un inhibiteur pharmacologique spécifique. Durant ma thèse, j'ai démontré que seule l'isoforme p110a est nécessaire à l'initiation de la carcinogenèse pancréatique induite par Kras muté. De manière dose-dépendante, l'inactivation d'une seule copie de p110a prolonge significativement la survie des animaux, tandis que les souris présentant une inactivation complète de l'activité kinase de p110a ne développent pas de lésion prénéoplasique. L'inactivation de cette seule isoforme bloque totalement la transdifférenciation des acini, normalement induite par Kras muté ou lors d'une pancréatite aigüe. p110a contrôle cette reprogrammation cellulaire, ainsi que les modifications morphologiques associées, en régulant les petites GTPases Rho, intermédiaires de signalisation essentielles au remodelage du cytosquelette d'actine. De plus, l'activité kinase de p110a est essentielle au maintien d'autres voies de signalisation oncogéniques comme les voies EGFR/MAPK, NF-kB ou encore IL6/STAT3. Dans un modèle de carcinogenèse pancréatique agressif où le suppresseur de tumeur p53 est partiellement inactivé, p110a exprimée dans l'épithélium est impliquée dans la progression tumorale en régulant la prolifération des cellules canalaires mais également, celle des cellules du microenvironnement par une action possiblement paracrine. Cette étude est la première démonstration génétique in vivo de l'implication d'une isoforme de PI3K dans le cancer du pancréas, et apporte un rationnel en faveur de l'utilisation des inhibiteurs spécifiques de p110a dans le traitement du cancer du pancréas. / Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers, with a 5-yr survival rate <5% of diagnosed patients. No efficient chemotherapy is currently available. PI3K signaling is one of the most altered signaling pathways in cancer. In half of PDAC, PI3K/Akt/mTOR activation is increased and correlated with poor prognosis. Mammals have four isoforms of Class I PI3K (p110a, p110ß p110d and p110y)which activate the Akt/mTOR signaling pathway. p110a and p110ß are ubiquitously expressed whereas p110d and p110y are mainly expressed in immune cells. All PI3K isoforms possess a Ras-binding domain whether it is functional for all isoforms remains to be demonstrated. Upon cell stimulation by cell surface receptors, PI3K generate the phosphatidylinositol tri-phosphate (PIP3), a lipid messenger involved in many cellular processes such as cell growth, proliferation, migration or cell differentiation. PI3K isoforms have nonredundant roles under physiological, nondisease conditions. In cancer, this redundancy of PI3K isoforms appears more complex and intensely debated. It is thus crucial to delineate and understand which PI3K isoform we have to target for each cancer type. Recently, it was shown that PI3K/Akt tumour dependency is tissue-specific and depends on genetic alterations. KrasG12D -activating mutation is found in more than 90% of PDAC and known as the PDAC-initiative mutation. Pancreatic specific expression of a KrasG12D allele in mice faithfully reproduces human pancreatic cancerogenesis, from preneoplastic lesions to adenocarcinoma. Based on the fact that PI3K isoforms and Kras interacts physically and that Kras is the major mutation in PDAC, the aim of my PhD project aims to determine if the two ubiquitous PI3K isoforms, p110a and p110ß, could have non redundant roles during Kras-driven pancreatic cancer initiation. In order to dissect the putative divergent role of p110a and p110ß in the inititation of Kras-driven pancreatic cancerogenesis, we generated new mice models which combined the expression of mutated-Kras and an inactivation of the p110a or p110ß-kinase domains in the pancreas using a conditional targeting strategy, mimicking pharmacological blockade of p110a or p110ß activity. During my PhD, I demonstrated that PI3K p110a isoform is required for Kras-driven pancreatic cancerogenesis. Inactivation of one copy of the pik3ca gene is sufficient to prevent mouse lethality and complete inactivation of p110a catalytic activity completely blocked the occurrence of all types of preneoplastic lesions induced by mutated Kras. Inactivation of this single isoform abrogates the transition of exocrine acinar cells into pancreatic preneoplastic ductal lesions induced by oncogenic Kras and/or pancreatic injury. p110a signaling through small GTPase Rho and actin cytoskeleton controls the reprogramming of acinar cells and regulates cell morphology in vivo and in vitro. Moreover, p110a kinase activity is required for the maintenance of other oncogenic signaling pathways as EGFR/MAPK, NF-kB or IL6/STAT3 axis. Finally, epithelial p110a was necessary for pancreatic ductal cancers to arise from Kras-induced pancreatic preneoplastic lesions by increasing epithelial and stromal cells proliferation in the context of mutated p53. This is the first genetic demonstration of the physiopathological implication of one PI3K isoform in PDAC in vivo and my data provide a strong rational for the use of selective p110a inhibitors in pancreatic cancer therapeutic strategy.
37

Genetic heterogeneity of EGFR and KRAS mutations in primary tumor tissue from non-small cell lung cancer patients

Mattsson, Johanna January 2011 (has links)
Activated epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations characterize molecular subgroups of non-small cell lung cancer (NSCLC) and have a strong predictive value for response to EGFR inhibitor therapy. Recently, EGFR mutation testing was included in the diagnostic algorithm of NSCLC. However, there is a controversy about the clonal stability of the mutation during the progression of the disease. The aim of this study was to analyze NSCLC tumor tissue for the presence of both EGFR and KRAS mutations in morphologically different parts of the primary tumor. Formaldehyd fixed and paraffin embedded lung cancer specimens from primary resected NSCLC patients were selected; five cases harboring EGFR and five with KRAS mutations. From each tumor, three morphologically different tumor sites were manually micro-dissected and analyzed for the presence of EGFR and KRAS mutations. Additionally, normal lung tissue at a distance from the primary tumor as well as in close vicinity was tested.The EGFR and KRAS status were consistent in the three different areas of the primary tumors of all ten cases. EGFR as well as KRAS mutations were as well detectable in close and in some distant normal lung parenchyma in 7 of 10 analyzed patient samples. In conclusion, we found consistent KRAS and EGFR mutation status in primary NSCLC tumors. This finding is of importance for clinical practice, because it indicates that any part of the tumor, independent of intratumoral histological pattern, is representative for EGFR and KRAS mutation testing.
38

Molekylär klassificering av tjocktarmscancer : PAM-klusteranalys för identifiering av undergrupper

Arvidsson, Per, Snickars, Samuel January 2012 (has links)
The main objective of this study is to divide a number of colorectal cancer cases into subgroups based on their molecular features using cluster analysis. The data used is supplied by a research group at Pathology, the Department of Medical Biosciences, Umeå University, and consists, after some preparation, of 455 observations which is a larger data set than many similar studies. The molecular variables that the clustering is based on are CIMP (CpG Island Methylator Phenotype), MSI (Micro Satellite Instability), BRAF- and KRAS-mutations. These are categorical variables and consequently the clustering method used is PAM (Partitioning Around Medoids) which is particularly useful with data on diverse variable level. The final analysis results in four subgroups that are represented by different combinations of attributes on the aforementioned variables. The disparity between the clusters are then evaluated by, for instance, comparing the survival time for their pertaining patients and it appears that two of the clusters are significantly different in this aspect. Other patient related and tumor specific characteristics are also linked with the separate cancer types and tested if they occur in varying extent. The locations of the tumors in the colon are for instance significantly different between the groups. Cluster analyses are exploratory tools so the choice of useful variables and subsequent interpretation of the results can be complicated and require relevant subject knowledge. / Huvudsyftet med denna studie är att med hjälp av klusteranalys dela in en mängd tjocktarmscancerfall i undergrupper baserat på deras molekylära egenskaper. Materialet som används tillhandahålls av en forskningsgrupp vid Patologi, Institutionen för medicinsk biovetenskap, Umeå universitet, och består efter viss bearbetning av 455 observationer vilket är en större datamängd än flera liknande studier. De molekylära variabler som ligger till grund för klusterindelningen är nivån på CIMP (CpG Island Methylator Phenotype), MSI (Mikrosatellitinstabillitet), BRAF- och KRAS-mutationer. Dessa är kategoriska variabler och därför används PAM (Partitioning Around Medoids) som är en särskild klusterteknik lämpad vid data på varierade variabelnivåer. I det slutliga resultatet fås fyra undergrupper som representeras av olika kombinationer av utfallen på ovannämnda variabler. Klustren utvärderas bland annat genom att jämföra överlevnadstiden för varje kluster, och det visar sig att två av klustren skiljer sig signifikant åt i detta avseende. Även andra patientrelaterade och tumörspecifika egenskaper kopplas samman till de olika cancertyperna och testas om de förekommer i varierande utsträckning. Var någonstans tumören är placerad är till exempel signifikant skilt mellan grupperna. Klusteranalyser är explorativa redskap så valet av variabler och sedermera tolkningar av resultat kan vara komplicerade och kräva stor sakkunskap.
39

The cooperation of the tumor suppressor gene Dlc1 and the oncogene Kras in tumorigenesis

Buse, Cordula 25 October 2012 (has links)
This thesis investigated the cooperation of the Kras2 oncogene with the tumor suppressor gene Dlc1 in lung tumor development. Dlc1 is a negative regulator of RhoGTPase proteins, which are mainly involved in the regulation of the actin cytoskeleton and cell migration. We hypothesized that loss of Dlc1 expression leads to more aggressive tumors, which should also result in increased incidence of metastasis. All experiments were performed in mice containing a heterozygous oncogenic Kras allele and a heterozygous gene trapped Dlc1 allele (KD) and in mice only carrying the oncogenic Kras allele (K+). Throughout all experiments we have consistently found no significant differences between the two groups in terms of tumor burden (tumor numbers, sizes and areas), metastases or methylation patterns. These results suggest that heterozygous downregulation of Dlc1 is not enough to increase tumor formation and metastasis development in the Kras lung tumors.
40

In Vitro Modeling of Pancreatic Duct Cell Carcinogenesis

Leung, Lisa 20 June 2014 (has links)
Pancreatic adenocarcinoma (PDAC) putatively arises from the pancreatic duct, thus usage of the normal human pancreatic duct epithelial (HPDE) cell line is an ideal model to examine the successive accumulation of genetic alterations involved in carcinogenesis. KRAS mutations have been reported in 90% of PDACs. Oncogenic KRAS elicits activation of downstream pathways involved in survival, motility, and cell cycle progression. KRASG12V introduction in the HPDE cell line upregulates Lipocalin-2 (LCN2) expression. LCN2 has been identified in numerous carcinomas and is associated with survival, tumorigenicity, and invasion. In this work, LCN2 was found to be commonly expressed in high grade pancreatic duct neoplastic precursor lesions and PDAC illustrating its potential as a biomarker. Moreover, in vitro and in vivo studies demonstrate that high LCN2 expression promotes gemcitabine resistance, MMP-9 activity, angiogenesis, and tumorigenicity. Loss of Smad4 function is found in 55% of PDAC cases. Smad4 is a critical component in the TGF-β signaling which mediates the transcription of genes involved in processes such as cell cycle arrest, apoptosis, and invasion. This work examined the consequences of KRASG12V expression and Smad4 loss in the HPDE model. Cellular invasion was promoted by KRASG12V expression or knocking down Smad4 by 80% in the HPDE model. A TGF-β resistant HPDE cell line, TβR, was shown to lack Smad4 expression due to deletion, promoter methylation, and nonsense mutation. KRASG12V expression in the TβR model (TβR KRAS) promoted neoplastic transformation and tumour formation in immunodeficient mice with complete penetrance. Smad4 expression in the TβR KRAS cell line reinstated TGF-β signaling, delayed tumour formation, and decreased metastatic spread. This study provides evidence that Smad4 acts as a restriction point in the transformation of HPDE cells. Overall, this work examines the contribution of genes involved in transformation, and identifies a potential therapeutic and diagnostic biomarker in PDAC.

Page generated in 0.1118 seconds