Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 19(6)2018 May 27.
Article in English | MEDLINE | ID: mdl-29861494

ABSTRACT

The Ras oncogene (Rat Sarcoma oncogene, a small GTPase) is a key driver of human cancer, however alone it is insufficient to produce malignancy, due to the induction of cell cycle arrest or senescence. In a Drosophila melanogaster genetic screen for genes that cooperate with oncogenic Ras (bearing the RasV12 mutation, or RasACT), we identified the Drosophila Src (Sarcoma virus oncogene) family non-receptor tyrosine protein kinase genes, Src42A and Src64B, as promoting increased hyperplasia in a whole epithelial tissue context in the Drosophila eye. Moreover, overexpression of Src cooperated with RasACT in epithelial cell clones to drive neoplastic tumourigenesis. We found that Src overexpression alone activated the Jun N-terminal Kinase (JNK) signalling pathway to promote actin cytoskeletal and cell polarity defects and drive apoptosis, whereas, in cooperation with RasACT, JNK led to a loss of differentiation and an invasive phenotype. Src + RasACT cooperative tumourigenesis was dependent on JNK as well as Phosphoinositide 3-Kinase (PI3K) signalling, suggesting that targeting these pathways might provide novel therapeutic opportunities in cancers dependent on Src and Ras signalling.


Subject(s)
Carcinogenesis , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Cell Differentiation , Cell Polarity , Compound Eye, Arthropod/enzymology , Compound Eye, Arthropod/metabolism , Compound Eye, Arthropod/pathology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Epithelium/enzymology , Epithelium/metabolism , Epithelium/physiopathology , Female , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , ras Proteins/physiology
2.
PLoS One ; 10(7): e0132987, 2015.
Article in English | MEDLINE | ID: mdl-26207831

ABSTRACT

During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers we used Drosophila epithelial tumor models, which are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors we used a comparative microarray analysis to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, our work suggests that EMT-promoting signals in human cancers could similarly utilize networks of these proteins to promote cancer stem cell states.


Subject(s)
Carcinogenesis/genetics , Drosophila Proteins/physiology , Genes, ras/physiology , Oncogenes/physiology , Receptors, Notch/physiology , Zinc Fingers/genetics , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Microarray Analysis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Interaction Domains and Motifs/genetics
3.
Biol Open ; 4(8): 1024-39, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26187947

ABSTRACT

The deregulation of cell polarity or cytoskeletal regulators is a common occurrence in human epithelial cancers. Moreover, there is accumulating evidence in human epithelial cancer that BTB-ZF genes, such as Bcl6 and ZBTB7A, are oncogenic. From our previous studies in the vinegar fly, Drosophila melanogaster, we have identified a cooperative interaction between a mutation in the apico-basal cell polarity regulator Scribble (Scrib) and overexpression of the BTB-ZF protein Abrupt (Ab). Herein, we show that co-expression of ab with actin cytoskeletal regulators, RhoGEF2 or Src64B, in the developing eye-antennal epithelial tissue results in the formation of overgrown amorphous tumours, whereas ab and DRac1 co-expression leads to non-cell autonomous overgrowth. Together with ab, these genes affect the expression of differentiation genes, resulting in tumours locked in a progenitor cell fate. Finally, we show that the expression of two mammalian genes related to ab, Bcl6 and ZBTB7A, which are oncogenes in mammalian epithelial cancers, significantly correlate with the upregulation of cytoskeletal genes or downregulation of apico-basal cell polarity neoplastic tumour suppressor genes in colorectal, lung and other human epithelial cancers. Altogether, this analysis has revealed that upregulation of cytoskeletal regulators cooperate with Abrupt in Drosophila epithelial tumorigenesis, and that high expression of human BTB-ZF genes, Bcl6 and ZBTB7A, shows significant correlations with cytoskeletal and cell polarity gene expression in specific epithelial tumour types. This highlights the need for further investigation of the cooperation between these genes in mammalian systems.

4.
PLoS Genet ; 9(7): e1003627, 2013.
Article in English | MEDLINE | ID: mdl-23874226

ABSTRACT

The capacity of tumour cells to maintain continual overgrowth potential has been linked to the commandeering of normal self-renewal pathways. Using an epithelial cancer model in Drosophila melanogaster, we carried out an overexpression screen for oncogenes capable of cooperating with the loss of the epithelial apico-basal cell polarity regulator, scribbled (scrib), and identified the cell fate regulator, Abrupt, a BTB-zinc finger protein. Abrupt overexpression alone is insufficient to transform cells, but in cooperation with scrib loss of function, Abrupt promotes the formation of massive tumours in the eye/antennal disc. The steroid hormone receptor coactivator, Taiman (a homologue of SRC3/AIB1), is known to associate with Abrupt, and Taiman overexpression also drives tumour formation in cooperation with the loss of Scrib. Expression arrays and ChIP-Seq indicates that Abrupt overexpression represses a large number of genes, including steroid hormone-response genes and multiple cell fate regulators, thereby maintaining cells within an epithelial progenitor-like state. The progenitor-like state is characterised by the failure to express the conserved Eyes absent/Dachshund regulatory complex in the eye disc, and in the antennal disc by the failure to express cell fate regulators that define the temporal elaboration of the appendage along the proximo-distal axis downstream of Distalless. Loss of scrib promotes cooperation with Abrupt through impaired Hippo signalling, which is required and sufficient for cooperative overgrowth with Abrupt, and JNK (Jun kinase) signalling, which is required for tumour cell migration/invasion but not overgrowth. These results thus identify a novel cooperating oncogene, identify mammalian family members of which are also known oncogenes, and demonstrate that epithelial tumours in Drosophila can be characterised by the maintenance of a progenitor-like state.


Subject(s)
Carcinogenesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , MAP Kinase Signaling System/genetics , Neoplasms, Glandular and Epithelial/genetics , Nuclear Proteins/genetics , Animals , Cell Proliferation , Disease Models, Animal , Drosophila Proteins/metabolism , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Glandular and Epithelial/pathology , Nuclear Proteins/metabolism , Oncogene Protein p65(gag-jun)/genetics , Oncogene Protein p65(gag-jun)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
Dis Model Mech ; 6(3): 661-78, 2013 May.
Article in English | MEDLINE | ID: mdl-23324326

ABSTRACT

The Ras oncogene contributes to ≈ 30% of human cancers, but alone is not sufficient for tumorigenesis. In a Drosophila screen for oncogenes that cooperate with an activated allele of Ras (Ras(ACT)) to promote tissue overgrowth and invasion, we identified the GTP exchange factor RhoGEF2, an activator of Rho-family signalling. Here, we show that RhoGEF2 also cooperates with an activated allele of a downstream effector of Ras, Raf (Raf(GOF)). We dissect the downstream pathways through which RhoGEF2 cooperates with Ras(ACT) (and Raf(GOF)), and show that RhoGEF2 requires Rho1, but not Rac, for tumorigenesis. Furthermore, of the Rho1 effectors, we show that RhoGEF2 + Ras (Raf)-mediated tumorigenesis requires the Rho kinase (Rok)-Myosin-II pathway, but not Diaphanous, Lim kinase or protein kinase N. The Rho1-Rok-Myosin-II pathway leads to the activation of Jun kinase (JNK), in cooperation with Ras(ACT). Moreover, we show that activation of Rok or Myosin II, using constitutively active transgenes, is sufficient for cooperative tumorigenesis with Ras(ACT), and together with Ras(ACT) leads to strong activation of JNK. Our results show that Rok-Myosin-II activity is necessary and sufficient for Ras-mediated tumorigenesis. Our observation that activation of Myosin II, which regulates Filamentous actin (F-actin) contractility without affecting F-actin levels, cooperates with Ras(ACT) to promote JNK activation and tumorigenesis, suggests that increased cell contractility is a key factor in tumorigenesis. Furthermore, we show that signalling via the Tumour necrosis factor (TNF; also known as Egr)-ligand-JNK pathway is most likely the predominant pathway that activates JNK upon Rok activation. Overall, our analysis highlights the need for further analysis of the Rok-Myosin-II pathway in cooperation with Ras in human cancers.


Subject(s)
Cell Transformation, Neoplastic/pathology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Myosin Type II/metabolism , Signal Transduction , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Apoptosis , Arthropod Antennae/growth & development , Cell Cycle Proteins , Cell Differentiation , Cell Shape , Cell Transformation, Neoplastic/metabolism , Clone Cells , Drosophila melanogaster/enzymology , Enzyme Activation , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Larva/metabolism , MAP Kinase Signaling System , Models, Biological , Proto-Oncogene Proteins c-raf/metabolism , Pupa/metabolism , Up-Regulation , rac GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
6.
Dis Model Mech ; 6(2): 521-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996645

ABSTRACT

Anti-cancer drug development involves enormous expenditure and risk. For rapid and economical identification of novel, bioavailable anti-tumour chemicals, the use of appropriate in vivo tumour models suitable for large-scale screening is key. Using a Drosophila Ras-driven tumour model, we demonstrate that tumour overgrowth can be curtailed by feeding larvae with chemicals that have the in vivo pharmacokinetics essential for drug development and known efficacy against human tumour cells. We then develop an in vivo 96-well plate chemical screening platform to carry out large-scale chemical screening with the tumour model. In a proof-of-principle pilot screen of 2000 compounds, we identify the glutamine analogue, acivicin, a chemical with known activity against human tumour cells, as a potent and specific inhibitor of Drosophila tumour formation. RNAi-mediated knockdown of candidate acivicin target genes implicates an enzyme involved in pyrimidine biosynthesis, CTP synthase, as a possible crucial target of acivicin-mediated inhibition. Thus, the pilot screen has revealed that Drosophila tumours are glutamine-dependent, which is an emerging feature of many human cancers, and has validated the platform as a powerful and economical tool for in vivo chemical screening. The platform can also be adapted for use with other disease models, thus offering widespread applications in drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drosophila melanogaster/drug effects , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Biological Availability , Cell Proliferation/drug effects , Cytidine Triphosphate/biosynthesis , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drosophila melanogaster/cytology , Glutamine/metabolism , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Pharmacogenetics , Pilot Projects
7.
BMC Dev Biol ; 11: 57, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21955824

ABSTRACT

BACKGROUND: Epithelial neoplasias are associated with alterations in cell polarity and excessive cell proliferation, yet how these neoplastic properties are related to one another is still poorly understood. The study of Drosophila genes that function as neoplastic tumor suppressors by regulating both of these properties has significant potential to clarify this relationship. RESULTS: Here we show in Drosophila that loss of Scribbled (Scrib), a cell polarity regulator and neoplastic tumor suppressor, results in impaired Hippo pathway signaling in the epithelial tissues of both the eye and wing imaginal disc. scrib mutant tissue overgrowth, but not the loss of cell polarity, is dependent upon defective Hippo signaling and can be rescued by knockdown of either the TEAD/TEF family transcription factor Scalloped or the transcriptional coactivator Yorkie in the eye disc, or reducing levels of Yorkie in the wing disc. Furthermore, loss of Scrib sensitizes tissue to transformation by oncogenic Ras-Raf signaling, and Yorkie-Scalloped activity is required to promote this cooperative tumor overgrowth. The inhibition of Hippo signaling in scrib mutant eye disc clones is not dependent upon JNK activity, but can be significantly rescued by reducing aPKC kinase activity, and ectopic aPKC activity is sufficient to impair Hippo signaling in the eye disc, even when JNK signaling is blocked. In contrast, warts mutant overgrowth does not require aPKC activity. Moreover, reducing endogenous levels of aPKC or increasing Scrib or Lethal giant larvae levels does not promote increased Hippo signaling, suggesting that aPKC activity is not normally rate limiting for Hippo pathway activity. Epistasis experiments suggest that Hippo pathway inhibition in scrib mutants occurs, at least in part, downstream or in parallel to both the Expanded and Fat arms of Hippo pathway regulation. CONCLUSIONS: Loss of Scrib promotes Yorkie/Scalloped-dependent epithelial tissue overgrowth, and this is also important for driving cooperative tumor overgrowth with oncogenic Ras-Raf signaling. Whether this is also the case in human cancers now warrants investigation since the cell polarity function of Scrib and its capacity to restrain oncogene-mediated transformation, as well as the tissue growth control function of the Hippo pathway, are conserved in mammals.


Subject(s)
Cell Polarity/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Cell Proliferation , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epithelial Cells/metabolism , Eye/embryology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins , Mutation , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oncogene Protein p21(ras)/metabolism , Protein Kinase C/biosynthesis , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Wings, Animal/embryology , YAP-Signaling Proteins , raf Kinases/metabolism
8.
Genetics ; 188(1): 105-25, 2011 May.
Article in English | MEDLINE | ID: mdl-21368274

ABSTRACT

We have shown previously that mutations in the apico-basal cell polarity regulators cooperate with oncogenic Ras (Ras(ACT)) to promote tumorigenesis in Drosophila melanogaster and mammalian cells. To identify novel genes that cooperate with Ras(ACT) in tumorigenesis, we carried out a genome-wide screen for genes that when overexpressed throughout the developing Drosophila eye enhance Ras(ACT)-driven hyperplasia. Ras(ACT)-cooperating genes identified were Rac1 Rho1, RhoGEF2, pbl, rib, and east, which encode cell morphology regulators. In a clonal setting, which reveals genes conferring a competitive advantage over wild-type cells, only Rac1, an activated allele of Rho1 (Rho1(ACT)), RhoGEF2, and pbl cooperated with Ras(ACT), resulting in reduced differentiation and large invasive tumors. Expression of RhoGEF2 or Rac1 with Ras(ACT) upregulated Jun kinase (JNK) activity, and JNK upregulation was essential for cooperation. However, in the whole-tissue system, upregulation of JNK alone was not sufficient for cooperation with Ras(ACT), while in the clonal setting, JNK upregulation was sufficient for Ras(ACT)-mediated tumorigenesis. JNK upregulation was also sufficient to confer invasive growth of Ras(V12)-expressing mammalian MCF10A breast epithelial cells. Consistent with this, HER2(+) human breast cancers (where human epidermal growth factor 2 is overexpressed and Ras signaling upregulated) show a significant correlation with a signature representing JNK pathway activation. Moreover, our genetic analysis in Drosophila revealed that Rho1 and Rac are important for the cooperation of RhoGEF2 or Pbl overexpression and of mutants in polarity regulators, Dlg and aPKC, with Ras(ACT) in the whole-tissue context. Collectively our analysis reveals the importance of the RhoGEF/Rho-family/JNK pathway in cooperative tumorigenesis with Ras(ACT).


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Genes, ras , Guanine Nucleotide Exchange Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Precancerous Conditions/enzymology , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cell Shape , Cell Survival , Clone Cells , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Eye/cytology , Eye/growth & development , Eye/ultrastructure , Genes, Insect/genetics , Humans , MAP Kinase Signaling System , Precancerous Conditions/pathology , Protein Kinase C/metabolism , Reproducibility of Results , Rho Guanine Nucleotide Exchange Factors , Up-Regulation/genetics
9.
BMC Biol ; 7: 62, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19778415

ABSTRACT

BACKGROUND: Metastatic neoplasias are characterized by excessive cell proliferation and disruptions to apico-basal cell polarity and tissue architecture. Understanding how alterations in cell polarity can impact upon tumour development is, therefore, a central issue in cancer biology. The Drosophila gene scribble (scrib) encodes a PDZ-domain scaffolding protein that regulates cell polarity and acts as a tumour suppressor in flies. Increasing evidence also implicates the loss of human Scrib in cancer. In this report, we investigate how loss of Scrib promotes epithelial tumourigenesis in Drosophila, both alone and in cooperation with oncogenic mutations. RESULTS: We find that genetically distinct atypical protein kinase C (aPKC)-dependent and Jun N-terminal kinase (JNK)-dependent alterations in scrib mutants drive epithelial tumourigenesis. First, we show that over-expression of the apical cell polarity determinants Crumbs (Crb) or aPKC induces similar cell morphology defects and over-proliferation phenotypes as scrib loss-of-function. However, the morphological and proliferative defects in scrib mutants are independent of Crb function, and instead can be rescued by a dominant negative (kinase dead) aPKC transgene. Secondly, we demonstrate that loss of Scrib promotes oncogene-mediated transformation through both aPKC and JNK-dependent pathways. JNK normally promotes apoptosis of scrib mutant cells. However, in cooperation with oncogenic activated Ras or Notch signalling, JNK becomes an essential driver of tumour overgrowth and invasion. aPKC-dependent signalling in scrib mutants cooperates with JNK to significantly enhance oncogene-mediated tumour overgrowth. CONCLUSION: These results demonstrate distinct aPKC and JNK-dependent pathways through which loss of Scrib promotes tumourigenesis in Drosophila. This is likely to have a direct relevance to the way in which human Scrib can similarly restrain an oncogene-mediated transformation and, more generally, on how the outcome of oncogenic signalling can be profoundly perturbed by defects in apico-basal epithelial cell polarity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Epithelium/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/genetics , Mutant Proteins/metabolism , Neoplasms/enzymology , Protein Kinase C/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Polarity , Cell Proliferation , Cell Shape , Cell Survival , Cell Transformation, Neoplastic , Clone Cells , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Epithelium/enzymology , MAP Kinase Signaling System , Membrane Proteins/metabolism , Models, Biological , Mutant Proteins/genetics , Mutation/genetics , Neoplasms/pathology , Phenotype
10.
Dev Biol ; 311(1): 106-23, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17870065

ABSTRACT

In homozygous mutants of Drosophila lethal-2-giant larvae (lgl), tissues lose apico-basal cell polarity and exhibit ectopic proliferation. Here, we use clonal analysis in the developing eye to investigate the effect of lgl null mutations in the context of surrounding wild-type tissue. lgl- clones in the larval eye disc exhibit ectopic expression of the G1-S regulator, Cyclin E, and ectopic proliferation, but do not lose apico-basal cell polarity. Decreasing the perdurance of Lgl protein in larval eye disc clones, by forcing extra proliferation of lgl- tissue (using a Minute background), leads to a loss in cell polarity and to more extreme ectopic cell proliferation. Later in development at the pupal stage, lgl mutant photoreceptor cells show aberrant apico-basal cell polarity, but this is not associated with ectopic proliferation, presumably because cells are differentiated. Thus in a clonal context, the ectopic proliferation and cell polarity defects of lgl- mutants are separable. Furthermore, lgl- mosaic eye discs have alterations in the normal patterns of apoptosis: in larval discs some lgl- and wild-type cells at the clonal boundary undergo apoptosis and are excluded from the epithelia, but apoptosis is decreased elsewhere in the disc, and in pupal retinas lgl- tissue shows less apoptosis.


Subject(s)
Cell Polarity , Cell Proliferation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Drosophila/cytology , Eye/cytology , Eye/embryology , Mutation
11.
Nat Rev Cancer ; 5(8): 626-39, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16034367

ABSTRACT

The development of human cancer is a multistep process, involving the cooperation of mutations in signalling, cell-cycle and cell-death pathways, as well as interactions between the tumour and the tumour microenvironment. To dissect the steps of tumorigenesis, simple animal models are needed. This article discusses the use of the genetically amenable, multicellular organism, the vinegar fly Drosophila melanogaster. In particular, recent studies have highlighted the power of D. melanogaster for examining cooperative interactions between tumour suppressors and oncogenes and for generating in vivo models of tumour development and metastasis.


Subject(s)
Drosophila melanogaster/genetics , Neoplasms/genetics , Oncogenes/genetics , Signal Transduction/genetics , Animals , Humans , Models, Animal
12.
Curr Opin Cell Biol ; 16(6): 614-22, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15530771

ABSTRACT

Studies in animal models have revealed many surprises regarding the importance of key cell cycle regulators during animal development and homeostasis, underscoring the plasticity and redundancy of cell cycle circuitry within a whole-animal context. Moreover, checkpoint regulators, which are not essential for viability in yeast and cultured cells, play important roles in cell cycle control during development.


Subject(s)
Cell Cycle , Gene Expression Regulation , Models, Animal , Animals , Cell Survival , Circadian Rhythm , DNA Replication , G1 Phase , Humans , Mice , Mice, Knockout , Mitosis
13.
Oncogene ; 22(58): 9225-30, 2003 Dec 18.
Article in English | MEDLINE | ID: mdl-14681682

ABSTRACT

Scribble (scrib), discs large (dlg) and lethal giant larvae (lgl) encode proteins that regulate cell polarity and have been identified as neoplastic tumour suppressor genes in Drosophila melanogaster. Here, we have used the Drosophila model system to provide the first functional evidence that human Scribble (hScrib) can act as a tumour suppressor. We show that hScrib protein displays highly polarized localization in mammalian epithelial cells and colocalizes with mammalian Dlg, similar to D. melanogaster Scribble (DmScrib) distribution in Drosophila epithelium. Furthermore, hScrib can rescue the polarity and tumorous overgrowth defects of scrib mutant Drosophila. hScrib therefore can act as an effective tumour suppressor in vivo, regulating both apical-basal polarity and cellular proliferation in a manner similar to that of DmScrib in Drosophila. These data demonstrate that hScrib is a functional homologue of DmScrib and therefore predict an important role for hScrib in the suppression of mammalian tumorigenesis.


Subject(s)
Drosophila Proteins/genetics , Genes, Tumor Suppressor , Membrane Proteins/genetics , Mutation , Alleles , Animals , Drosophila melanogaster , Female , Genotype , Green Fluorescent Proteins , Heterozygote , Humans , Luminescent Proteins/metabolism , Male , Microscopy, Fluorescence , Models, Genetic , Phenotype , Tumor Suppressor Proteins
14.
EMBO J ; 22(21): 5769-79, 2003 Nov 03.
Article in English | MEDLINE | ID: mdl-14592975

ABSTRACT

Cancer is a multistep process involving cooperation between oncogenic or tumor suppressor mutations and interactions between the tumor and surrounding normal tissue. Here we present the first description of cooperative tumorigenesis in Drosophila, by using a system that mimics the development of tumors in mammals. We have used the MARCM system to generate mutant clones of the apical-basal cell polarity tumor suppressor gene, scribble, in the context of normal tissue. We show that scribble mutant clones in the eye disc exhibit ectopic expression of cyclin E and ectopic cell cycles, but do not overgrow due to increased cell death mediated by the JNK pathway and the surrounding wild-type tissue. In contrast, when oncogenic Ras or Notch is expressed within the scribble mutant clones, cell death is prevented and neoplastic tumors develop. This demonstrates, for the first time in Drosophila, that activated alleles of Ras and Notch can act as cooperating oncogenes in the development of epithelial tumors, and highlights the importance of epithelial polarity regulators in restraining oncogenes and preventing tumor formation.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Genes, Insect , Membrane Proteins/genetics , Mutation , Animals , Apoptosis , Cyclin E/genetics , Drosophila/growth & development , Drosophila/physiology , Drosophila Proteins/physiology , Eye/growth & development , Gene Expression Regulation, Neoplastic , Genes, ras , JNK Mitogen-Activated Protein Kinases , Membrane Proteins/physiology , Mitogen-Activated Protein Kinases/physiology , Models, Biological , Neoplasms, Experimental/etiology , Neoplasms, Experimental/genetics , Receptors, Notch , Up-Regulation
15.
EMBO J ; 21(13): 3377-89, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12093739

ABSTRACT

Cyclin E-Cdk2 is essential for S phase entry. To identify genes interacting with cyclin E, we carried out a genetic screen using a hypomorphic mutation of Drosophila cyclin E (DmcycE(JP)), which gives rise to adults with a rough eye phenotype. Amongst the dominant suppressors of DmcycE(JP), we identified brahma (brm) and moira (mor), which encode conserved core components of the Drosophila Brm complex that is highly related to the SWI-SNF ATP-dependent chromatin remodeling complex. Mutations in genes encoding other Brm complex components, including snr1 (BAP45), osa and deficiencies that remove BAP60 and BAP111 can also suppress the DmcycE(JP) eye phenotype. We show that Brm complex mutants suppress the DmcycE(JP) phenotype by increasing S phases without affecting DmcycE protein levels and that DmcycE physically interacts with Brm and Snr1 in vivo. These data suggest that the Brm complex inhibits S phase entry by acting downstream of DmcycE protein accumulation. The Brm complex also physically interacts weakly with Drosophila retinoblastoma (Rbf1), but no genetic interactions were detected, suggesting that the Brm complex and Rbf1 act largely independently to mediate G(1) arrest.


Subject(s)
Cell Cycle Proteins/physiology , Cyclin E/physiology , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , G1 Phase/physiology , S Phase/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Alleles , Animals , Animals, Genetically Modified , Cell Cycle Proteins/genetics , Cyclin E/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Eye/ultrastructure , G1 Phase/genetics , Gene Dosage , Gene Expression Regulation, Developmental , Genes, Suppressor , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Larva/metabolism , Larva/ultrastructure , Macromolecular Substances , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phenotype , Protein Interaction Mapping , S Phase/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transgenes , Wings, Animal/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...