21 research outputs found

    Opposing effects of TIGAR- and RAC1-derived ROS on Wnt-driven proliferation in the mouse intestine

    Get PDF
    Reactive oxygen species (ROS) participate in numerous cell responses, including proliferation, DNA damage, and cell death. Based on these disparate activities, both promotion and inhibition of ROS have been proposed for cancer therapy. However, how the ROS response is determined is not clear. We examined the activities of ROS in a model of Apc deletion, where loss of the Wnt target gene Myc both rescues APC loss and prevents ROS accumulation. Following APC loss, Myc has been shown to up-regulate RAC1 to promote proliferative ROS through NADPH oxidase (NOX). However, APC loss also increased the expression of TIGAR, which functions to limit ROS. To explore this paradox, we used three-dimensional (3D) cultures and in vivo models to show that deletion of TIGAR increased ROS damage and inhibited proliferation. These responses were suppressed by limiting damaging ROS but enhanced by lowering proproliferative NOX-derived ROS. Despite having opposing effects on ROS levels, loss of TIGAR and RAC1 cooperated to suppress intestinal proliferation following APC loss. Our results indicate that the pro- and anti-proliferative effects of ROS can be independently modulated in the same cell, with two key targets in the Wnt pathway functioning to integrate the different ROS signals for optimal cell proliferation

    MYC is a metastasis gene for non-small-cell lung cancer.

    Get PDF
    Metastasis is a process by which cancer cells learn to form satellite tumors in distant organs and represents the principle cause of death of patients with solid tumors. NSCLC is the most lethal human cancer due to its high rate of metastasis. Lack of a suitable animal model has so far hampered analysis of metastatic progression. We have examined c-MYC for its ability to induce metastasis in a C-RAF-driven mouse model for non-small-cell lung cancer. c-MYC alone induced frank tumor growth only after long latency at which time secondary mutations in K-Ras or LKB1 were detected reminiscent of human NSCLC. Combination with C-RAF led to immediate acceleration of tumor growth, conversion to papillary epithelial cells and angiogenic switch induction. Moreover, addition of c-MYC was sufficient to induce macrometastasis in liver and lymph nodes with short latency associated with lineage switch events. Thus we have generated the first conditional model for metastasis of NSCLC and identified a gene, c-MYC that is able to orchestrate all steps of this process. Potential markers for detection of metastasis were identified and validated for diagnosis of human biopsies. These markers may represent targets for future therapeutic intervention as they include genes such as Gata4 that are exclusively expressed during lung development

    E-cadherin can limit the transforming properties of activating β-catenin mutations

    Get PDF
    Wnt pathway deregulation is a common characteristic of many cancers. But only Colorectal Cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of pancreas) have activating mutations in β-catenin (CTNNB1). We have compared the dynamics and the potency of β-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of β-catenin took much longer to achieve a Wnt deregulation and acquire a crypt-progenitor-cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of β-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of β-catenin mutation to differentially transform the SI versus the colon correlated with significantly higher expression of the β-catenin binding partner E-cadherin. This increased expression is associated with a higher number of E-cadherin:β-catenin complexes at the membrane. Reduction of E-cadherin synergised with an activating mutation of β-catenin so there was now a rapid CPC phenotype within the colon and SI. Thus there is a threshold of β-catenin that is required to drive transformation and E-cadherin can act as a buffer to prevent β-catenin accumulation

    Expression of B-RAF V600E in Type II Pneumocytes Causes Abnormalities in Alveolar Formation, Airspace Enlargement and Tumor Formation in Mice

    Get PDF
    Growth factor induced signaling cascades are key regulatory elements in tissue development, maintenance and regeneration. Perturbations of these cascades have severe consequences, leading to developmental disorders and neoplastic diseases. As a major function in signal transduction, activating mutations in RAF family kinases are the cause of human tumorigenesis, where B-RAF V600E has been identified as the prevalent mutant. In order to address the oncogenic function of B-RAF V600E, we have generated transgenic mice expressing the activated oncogene specifically in lung alveolar epithelial type II cells. Constitutive expression of B-RAF V600E caused abnormalities in alveolar epithelium formation that led to airspace enlargements. These lung lesions showed signs of tissue remodeling and were often associated with chronic inflammation and low incidence of lung tumors. The inflammatory cell infiltration did not precede the formation of the lung lesions but was rather accompanied with late tumor development. These data support a model where the continuous regenerative process initiated by oncogenic B-RAF-driven alveolar disruption provides a tumor-promoting environment associated with chronic inflammation

    Colorectal tumors require NUAK1 for protection from oxidative stress

    Get PDF
    The authors wish to thank the staff of the CRUK Beatson Institute Biological Services Unit for animal husbandry and assistance with in vivo experiments; the staff of the CRUK BI Histology core facility and William Clark of the NGS core facility; David McGarry, Rene Jackstadt, Jiska Van der Reest, Justin Bower and Heather McKinnon for many helpful discussions, and countless colleagues at the CRUK BI and Glasgow Institute of Cancer Sciences for support; Prem Premsrirut & Mirimus Inc. for design and generation of dox-inducible Nuak1 shRNA expressing mice Nathanael Gray for initial provision of NUAK1 inhibitors. Funding was provided by the University of Glasgow and the CRUK Beaton Institute. J.P. was supported by European Commission Marie Curie actions C.I.G. 618448 “SERPLUC” to D.J.M.; N.M. was supported through Worldwide Cancer (formerly AICR) grant 15-0279 to O.J.S. & D.J.M.; B.K. was funded through EC Marie Curie actions mobility award 705190 “NuSiCC”; T.M. was funded through British Lung Foundation grant APHD13-5. The laboratories of S.R.Z. (A12935), O.J.S. (A21139) and M.D. (A17096) are funded by Cancer Research UK. O.J.S. was additionally supported by European Research Council grant 311301 “ColoCan”.Peer reviewedPostprin

    Polycomb Group Protein Bmi1 Is Required for Growth of RAF Driven Non-Small-Cell Lung Cancer

    Get PDF
    Background: We have previously described a RAF oncogene driven transgenic mouse model for non small cell lung cancer (NSCLC). Here we examine whether tumor initiation and growth requires the stem cell self-renewal factor Bmi1. Principal Findings: In order to evaluate Bmi1 function in NSCLC two founder lines that differ in incidence and latency of tumor formation were compared. Ablation of Bmi1 expression in both lines had a dramatically decreased tumor growth. As the line with shorter latency matched the life span of Bmi1 knock out mice, these mice were chosen for further study. The absence of Bmi1 did not decrease the number of tumor initiation in these mice as only the size and not the number of tumors decreased. Reduction in tumor growth resulted from an increase in cell death and decrease in cell cycle progression that corresponded with up-regulation of the p16 INK4a and p19 ARF. Significance: The data identifies Bmi1 as an important factor for expansion but not initiation of RAF driven NSCLC

    Aanlyse der Rolle des E-(Epithelischer) Cadherin in der Lunge Tumorigenesis

    No full text
    Beim humanen nichtkleinzelligen Bronchialkarzinom ist die schrittweise Progression vom gutartigen Tumor zur malignen Metastasierung weitestgehend ungeklärt. In einem transgenen Mausmodell für das humane nichtkleinzellige Bronchialkarzinom, in dem in Lungenepithelzellen eine onkogene Mutante der Proteinkinase C-RAF exprimiert wird, können einzelne Schritte im Prozess der malignen Progression entschlüsselt werden. Die durch C-RAF induzierten Adenome zeichnen sich durch eine hohe genomische Stabilität in den Tumorzellen, durch starke interzelluläre Adhäsionskontakte zwischen den Tumorzellen und das Fehlen einer malignen Progression aus. Hier wurde demzufolge untersucht, ob die Auflösung der E-Cadherin-vermittelten Zellkontakte zwischen den einzelnen Tumorzellen eine Metastasierung auslösen könnte. Es wurden zwei genetische Ansätze verfolgt, um die Rolle der Tumorzelladhäsion im C-RAF Modell zu bewerten, die konditionelle Eliminierung des E-Cadheringens Cdh1 sowie die regulierbare transgene Expression von dominant-negativem E-Cadherin. Die Auflösung der E-Cadherin-vermittelten Zelladhäsion führte zur Neubildung von Tumorgefäßen, welche in der frühen Phase der Gefäßbildung durch Wiederherstellung des Zellkontakts reversibel war. Die vaskularisierten Tumore wuchsen schneller, bildeten invasive Fronten aus und führten zur Ausbildung von Mikrometastasen. Es konnte gezeigt werden, dass Beta-Catenin für die Induktion der Angiogenesefaktoren VEGF-A und VEGF-C in Lungentumorzellinien des Menschen und der Maus essentiell war. Lungentumorzellen aus den in situ Tumoren mit aufgelösten E-Cadherin-vermittelten Zellkontakten exprimierten Gene endodermaler und anderer Zellabstammung, was epigenetische Reprogrammierung in Tumorzellen als den Mechanismus bei der malignen Progression vermuten lässt.Steps involved in the progression of non-small cell lung cancer (NSCLC) to metastasis are poorly understood. Expression of oncogenic C-RAF in lung epithelial cells has yielded a model for non-small cell lung cancer (NSCLC). The induced adenomas are characterised by high genomic stability, a lack of tumor progression and pronounced cell-cell contacts raising the question whether disruption of E-cadherin complexes would promote progression to metastasis. Two genetic approaches were used to evaluate the role of adherens junctions in a C-RAF driven mouse model for NSCLC: conditional ablation of the Cdh1 gene and expression of dominant negative (dn) E-cadherin. Disruption of E-cadherin function caused massive formation of intratumoral vessels that was reversible in the early phase of induction. Vascularized tumors grew more rapidly, developed invasive fronts and gave rise to micrometastasis. ß-catenin was identified as a critical effector of E-cadherin disruption leading to up-regulation of angiogenic inducers (VEGF-A and VEGF-C) in mouse and human lung tumor cell lines. In vivo, lung tumor cells with disrupted E-cadherin expressed ß-catenin target genes of endodermal and other lineages suggesting that reprogramming may be involved in metastatic progression

    Conditional Expression of Oncogenic C-RAF in Mouse Pulmonary Epithelial Cells Reveals Differential Tumorigenesis and Induction of Autophagy Leading to Tumor Regression12

    Get PDF
    Here we describe a novel conditional mouse lung tumor model for investigation of the pathogenesis of human lung cancer. On the basis of the frequent involvement of the Ras-RAF-MEK-ERK signaling pathway in human non-small cell lung carcinoma (NSCLC), we have explored the target cell availability, reversibility, and cell type specificity of transformation by oncogenic C-RAF. Targeting expression to alveolar type II cells or to Clara cells, the two likely precursors of human NSCLC, revealed differential tumorigenicity between these cells. Whereas expression of oncogenic C-RAF in alveolar type II cells readily induced multifocal macroscopic lung tumors independent of the developmental state, few tumors with type II pneumocytes features and incomplete penetrance were found when targeted to Clara cells. Induced tumors did not progress and were strictly dependent on the initiating oncogene. Deinduction of mice resulted in tumor regression due to autophagy rather than apoptosis. Induction of autophagic cell death in regressing lung tumors suggests the use of autophagy enhancers as a treatment choice for patients with NSCLC
    corecore