Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Genes Dev ; 38(9-10): 360-379, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38811170

ABSTRACT

Glioblastoma (GBM) is the most aggressive primary brain cancer. These tumors exhibit high intertumoral and intratumoral heterogeneity in neoplastic and nonneoplastic compartments, low lymphocyte infiltration, and high abundance of myeloid subsets that together create a highly protumorigenic immunosuppressive microenvironment. Moreover, heterogeneous GBM cells infiltrate adjacent brain tissue, remodeling the neural microenvironment to foster tumor electrochemical coupling with neurons and metabolic coupling with nonneoplastic astrocytes, thereby driving growth. Here, we review heterogeneity in the GBM microenvironment and its role in low-to-high-grade glioma transition, concluding with a discussion of the challenges of therapeutically targeting the tumor microenvironment and outlining future research opportunities.


Subject(s)
Brain Neoplasms , Glioblastoma , Tumor Microenvironment , Humans , Glioblastoma/therapy , Glioblastoma/physiopathology , Brain Neoplasms/therapy , Brain Neoplasms/physiopathology , Brain Neoplasms/pathology , Animals
2.
STAR Protoc ; 3(3): 101609, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35990742

ABSTRACT

This protocol describes a genetic model system we developed for glioblastoma (GBM) in Drosophila melanogaster, which can be used to explore the pathogenic phenotypic effects of mutated genetic pathways and to identify potential therapeutic targets for tumors with these mutations. We present genetic schemes and experimental steps needed to create neoplastic glial brain tumors in larval Drosophila. We also provide steps to manipulate genes in this model and to perform brain fixation, immunostaining, and imaging of neoplastic larval brains. For complete details on the use and execution of this protocol, please refer to Read et al., (2009).


Subject(s)
Glioblastoma , Glioma , Animals , Brain/diagnostic imaging , Drosophila melanogaster/genetics , Glioblastoma/genetics , Glioma/pathology , Humans , Larva/genetics
4.
Pharmaceuticals (Basel) ; 14(2)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525329

ABSTRACT

Glioblastoma multiforme (GBM) is a highly malignant primary brain tumor. The current standard of care for GBM is the Stupp protocol which includes surgical resection, followed by radiotherapy concomitant with the DNA alkylator temozolomide; however, survival under this treatment regimen is an abysmal 12-18 months. New and emerging treatments include the application of a physical device, non-invasive 'tumor treating fields' (TTFs), including its concomitant use with standard of care; and varied vaccines and immunotherapeutics being trialed. Some of these approaches have extended life by a few months over standard of care, but in some cases are only available for a minority of GBM patients. Extensive activity is also underway to repurpose and reposition therapeutics for GBM, either alone or in combination with the standard of care. In this review, we present select molecules that target different pathways and are at various stages of clinical translation as case studies to illustrate the rationale for their repurposing-repositioning and potential clinical use.

5.
Clin Cancer Res ; 27(5): 1553-1569, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33172899

ABSTRACT

PURPOSE: Glioblastomas (GBMs), neoplasms derived from glia and neuroglial progenitor cells, are the most common and lethal malignant primary brain tumors diagnosed in adults, with a median survival of 14 months. GBM tumorigenicity is often driven by genetic aberrations in receptor tyrosine kinases, such as amplification and mutation of EGFR. EXPERIMENTAL DESIGN: Using a Drosophila glioma model and human patient-derived GBM stem cells and xenograft models, we genetically and pharmacologically tested whether the YAP and TAZ transcription coactivators, effectors of the Hippo pathway that promote gene expression via TEA domain (TEAD) cofactors, are key drivers of GBM tumorigenicity downstream of oncogenic EGFR signaling. RESULTS: YAP and TAZ are highly expressed in EGFR-amplified/mutant human GBMs, and their knockdown in EGFR-amplified/mutant GBM cells inhibited proliferation and elicited apoptosis. Our results indicate that YAP/TAZ-TEAD directly regulates transcription of SOX2, C-MYC, and EGFR itself to create a feedforward loop to drive survival and proliferation of human GBM cells. Moreover, the benzoporphyrin derivative verteporfin, a disruptor of YAP/TAZ-TEAD-mediated transcription, preferentially induced apoptosis of cultured patient-derived EGFR-amplified/mutant GBM cells, suppressed expression of YAP/TAZ transcriptional targets, including EGFR, and conferred significant survival benefit in an orthotopic xenograft GBM model. Our efforts led us to design and initiate a phase 0 clinical trial of Visudyne, an FDA-approved liposomal formulation of verteporfin, where we used intraoperative fluorescence to observe verteporfin uptake into tumor cells in GBM tumors in human patients. CONCLUSIONS: Together, our data suggest that verteporfin is a promising therapeutic agent for EGFR-amplified and -mutant GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Mutation , Transcription Factors/antagonists & inhibitors , Transcriptional Coactivator with PDZ-Binding Motif Proteins/antagonists & inhibitors , Verteporfin/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Drosophila melanogaster , ErbB Receptors/genetics , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells , Photosensitizing Agents/pharmacology , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Brain ; 144(1): 53-69, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33300045

ABSTRACT

Paediatric high-grade gliomas (HGGs) account for the most brain tumour-related deaths in children and have a median survival of 12-15 months. One promising avenue of research is the development of novel therapies targeting the properties of non-neoplastic cell-types within the tumour such as tumour associated macrophages (TAMs). TAMs are immunosuppressive and promote tumour malignancy in adult HGG; however, in paediatric medulloblastoma, TAMs exhibit anti-tumour properties. Much is known about TAMs in adult HGG, yet little is known about them in the paediatric setting. This raises the question of whether paediatric HGGs possess a distinct constituency of TAMs because of their unique genetic landscapes. Using human paediatric HGG tissue samples and murine models of paediatric HGG, we demonstrate diffuse midline gliomas possess a greater inflammatory gene expression profile compared to hemispheric paediatric HGGs. We also show despite possessing sparse T-cell infiltration, human paediatric HGGs possess high infiltration of IBA1+ TAMs. CD31, PDGFRß, and PDGFB all strongly correlate with IBA1+ TAM infiltration. To investigate the TAM population, we used the RCAS/tv-a system to recapitulate paediatric HGG in newborn immunocompetent mice. Tumours are induced in Nestin-positive brain cells by PDGFA or PDGFB overexpression with Cdkn2a or Tp53 co-mutations. Tumours driven by PDGFB have a significantly lower median survival compared to PDGFA-driven tumours and have increased TAM infiltration. NanoString and quantitative PCR analysis indicates PDGFB-driven tumours have a highly inflammatory microenvironment characterized by high chemokine expression. In vitro bone marrow-derived monocyte and microglial cultures demonstrate bone marrow-derived monocytes are most responsible for the production of inflammatory signals in the tumour microenvironment in response to PDGFB stimulation. Lastly, using knockout mice deficient for individual chemokines, we demonstrate the feasibility of reducing TAM infiltration and prolonging survival in both PDGFA and PDGFB-driven tumours. We identify CCL3 as a potential key chemokine in these processes in both humans and mice. Together, these studies provide evidence for the potent inflammatory effects PDGFB has in paediatric HGGs.


Subject(s)
Brain Neoplasms/immunology , Encephalitis/immunology , Proto-Oncogene Proteins c-sis/immunology , Tumor-Associated Macrophages/immunology , Adolescent , Adult , Animals , Brain Neoplasms/genetics , Cells, Cultured , Chemokines/genetics , Child , Child, Preschool , Encephalitis/genetics , Female , Glioma , Humans , Infant , Infant, Newborn , Male , Mice, Inbred C57BL , Transcriptome , Young Adult
7.
Glia ; 68(10): 2148-2166, 2020 10.
Article in English | MEDLINE | ID: mdl-32639068

ABSTRACT

Glioblastoma (GBM) is the most aggressive primary brain tumor. In addition to being genetically heterogeneous, GBMs are also immunologically heterogeneous. However, whether the differences in immune microenvironment are driven by genetic driver mutation is unexplored. By leveraging the versatile RCAS/tv-a somatic gene transfer system, we establish a mouse model for Classical GBM by introducing EGFRvIII expression in Nestin-positive neural stem/progenitor cells in adult mice. Along with our previously published Nf1-silenced and PDGFB-overexpressing models, we investigate the immune microenvironments of the three models of human GBM subtypes by unbiased multiplex profiling. We demonstrate that both the quantity and composition of the microenvironmental myeloid cells are dictated by the genetic driver mutations, closely mimicking what was observed in human GBM subtypes. These myeloid cells express high levels of the immune checkpoint protein PD-L1; however, PD-L1 targeted therapies alone or in combination with irradiation are unable to increase the survival time of tumor-bearing mice regardless of the driver mutations, reflecting the outcomes of recent human trials. Together, these results highlight the critical utility of immunocompetent mouse models for preclinical studies of GBM, making these models indispensable tools for understanding the resistance mechanisms of immune checkpoint blockade in GBM and immune cell-targeting drug discovery.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Immune Checkpoint Inhibitors/therapeutic use , Mutation/physiology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Cells, Cultured
8.
Adv Exp Med Biol ; 1167: 207-224, 2019.
Article in English | MEDLINE | ID: mdl-31520357

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary malignant adult brain tumor. Genomic amplifications, activating mutations, and overexpression of receptor tyrosine kinases (RTKs) such as EGFR, and genes in core RTK signaling transduction pathways such as PI3K are common in GBM. However, efforts to target these pathways have been largely unsuccessful in the clinic, and the median survival of GBM patients remains poor at 14-15 months. Therefore, to improve patient outcomes, there must be a concerted effort to elucidate the underlying biology involved in GBM tumorigenesis. Drosophila melanogaster has been a highly effective model for furthering our understanding of GBM tumorigenesis due to a number of experimental advantages it has over traditional mouse models. For example, there exists extensive cellular and genetic homology between humans and Drosophila, and 75% of genes associated with human disease have functional fly orthologs. To take advantage of these traits, we developed a Drosophila GBM model with constitutively active variants of EGFR and PI3K that effectively recapitulated key aspects of GBM disease. Researchers have utilized this model in forward genetic screens and have expanded on its functionality to make a number of important discoveries regarding requirements for key components in GBM tumorigenesis, including genes and pathways involved in extracellular matrix signaling, glycolytic metabolism, invasion/migration, stem cell fate and differentiation, and asymmetric cell division. Drosophila will continue to reveal novel biological pathways and mechanisms involved in gliomagenesis, and this knowledge may contribute to the development of effective treatment strategies to improve patient outcomes.


Subject(s)
Brain Neoplasms/pathology , Drosophila melanogaster , Glioblastoma/pathology , Adult , Animals , Cell Transformation, Neoplastic , Disease Models, Animal , Humans , Mice , Signal Transduction
9.
Sci Rep ; 9(1): 13611, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541170

ABSTRACT

Medulloblastoma is a malignant pediatric tumor that arises from neural progenitors in the cerebellum. Despite a five-year survival rate of ~70%, nearly all patients incur adverse side effects from current treatment strategies that drastically impact quality of life. Roughly one-third of medulloblastoma are driven by aberrant activation of the Sonic Hedgehog (SHH) signaling pathway. However, the scarcity of genetic mutations in medulloblastoma has led to investigation of other mechanisms contributing to cancer pathogenicity including epigenetic regulation of gene expression. Here, we show that Helicase, Lymphoid Specific (HELLS), a chromatin remodeler with epigenetic functions including DNA methylation and histone modification, is induced by Sonic Hedgehog (SHH) in SHH-dependent cerebellar progenitor cells and the developing murine cerebella. HELLS is also up-regulated in mouse and human SHH medulloblastoma. Others have shown that HELLS activity generally results in a repressive chromatin state. Our results demonstrate that increased expression of HELLS in our experimental systems is regulated by the oncogenic transcriptional regulator YAP1 downstream of Smoothened, the positive transducer of SHH signaling. Elucidation of HELLS as one of the downstream effectors of the SHH pathway may lead to novel targets for precision therapeutics with the promise of better outcomes for SHH medulloblastoma patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , DNA Helicases/genetics , Medulloblastoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Adult , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Child , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , Epigenesis, Genetic/genetics , Female , Hedgehog Proteins/metabolism , Hedgehog Proteins/physiology , Humans , Male , Medulloblastoma/metabolism , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Quality of Life , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation , YAP-Signaling Proteins
10.
Cancer Res ; 79(6): 1085-1097, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30530503

ABSTRACT

Glioblastoma (GBM) and lower grade gliomas (LGG) are the most common primary malignant brain tumors and are resistant to current therapies. Genomic analyses reveal that signature genetic lesions in GBM and LGG include copy gain and amplification of chromosome 7, amplification, mutation, and overexpression of receptor tyrosine kinases (RTK) such as EGFR, and activating mutations in components of the PI3K pathway. In Drosophila melanogaster, constitutive co-activation of RTK and PI3K signaling in glial progenitor cells recapitulates key features of human gliomas. Here we use this Drosophila glioma model to identify death-associated protein kinase (Drak), a cytoplasmic serine/threonine kinase orthologous to the human kinase STK17A, as a downstream effector of EGFR and PI3K signaling pathways. Drak was necessary for glial neoplasia, but not for normal glial proliferation and development, and Drak cooperated with EGFR to promote glial cell transformation. Drak phosphorylated Sqh, the Drosophila ortholog of nonmuscle myosin regulatory light chain (MRLC), which was necessary for transformation. Moreover, Anillin, which is a binding partner of phosphorylated Sqh, was upregulated in a Drak-dependent manner in mitotic cells and colocalized with phosphorylated Sqh in neoplastic cells undergoing mitosis and cytokinesis, consistent with their known roles in nonmuscle myosin-dependent cytokinesis. These functional relationships were conserved in human GBM. Our results indicate that Drak/STK17A, its substrate Sqh/MRLC, and the effector Anillin/ANLN regulate mitosis and cytokinesis in gliomas. This pathway may provide a new therapeutic target for gliomas.Significance: These findings reveal new insights into differential regulation of cell proliferation in malignant brain tumors, which will have a broader impact on research regarding mechanisms of oncogene cooperation and dependencies in cancer.See related commentary by Lathia, p. 1036.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Myosin Light Chains/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Biomarkers, Tumor/genetics , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mitosis , Myosin Light Chains/genetics , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured
11.
Development ; 145(23)2018 12 04.
Article in English | MEDLINE | ID: mdl-30327326

ABSTRACT

Stem cells reside in specialized microenvironments, called niches, that regulate their development and the development of their progeny. However, the development and maintenance of niches are poorly understood. In the Drosophila brain, cortex glial cells provide a niche that promotes self-renewal and proliferation of neural stem cell-like cells (neuroblasts). In the central brain, neuroblasts and their progeny control post-embryonic morphogenesis of cortex glia through PDGF-like ligands, and this PDGFR receptor tyrosine kinase (RTK) signaling in cortex glia is required for expression of DE-cadherin, which sustains neuroblasts. Thus, through an RTK-dependent feed-forward loop, neuroblasts and their glial niche actively maintain each other. When the EGFR RTK is constitutively activated in cortex glia, they overexpress PDGF orthologs to stimulate autocrine PDGFR signaling, which uncouples their growth and survival from neuroblasts, and drives neoplastic glial transformation and elimination of neuroblasts. These results provide fundamental insights into glial development and niche regulation, and show that niche-neural stem cell feed-forward signaling becomes hijacked to drive neural tumorigenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Morphogenesis , Neural Stem Cells/cytology , Neuroglia/cytology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Autocrine Communication , Brain/growth & development , Cell Proliferation , Cell Survival , Drosophila melanogaster/enzymology , Embryo, Nonmammalian/enzymology , Genetic Testing , Neural Stem Cells/metabolism , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism
12.
PLoS Genet ; 9(2): e1003253, 2013.
Article in English | MEDLINE | ID: mdl-23459592

ABSTRACT

Glioblastoma, the most common primary malignant brain tumor, is incurable with current therapies. Genetic and molecular analyses demonstrate that glioblastomas frequently display mutations that activate receptor tyrosine kinase (RTK) and Pi-3 kinase (PI3K) signaling pathways. In Drosophila melanogaster, activation of RTK and PI3K pathways in glial progenitor cells creates malignant neoplastic glial tumors that display many features of human glioblastoma. In both human and Drosophila, activation of the RTK and PI3K pathways stimulates Akt signaling along with other as-yet-unknown changes that drive oncogenesis. We used this Drosophila glioblastoma model to perform a kinome-wide genetic screen for new genes required for RTK- and PI3K-dependent neoplastic transformation. Human orthologs of novel kinases uncovered by these screens were functionally assessed in mammalian glioblastoma models and human tumors. Our results revealed that the atypical kinases RIOK1 and RIOK2 are overexpressed in glioblastoma cells in an Akt-dependent manner. Moreover, we found that overexpressed RIOK2 formed a complex with RIOK1, mTor, and mTor-complex-2 components, and that overexpressed RIOK2 upregulated Akt signaling and promoted tumorigenesis in murine astrocytes. Conversely, reduced expression of RIOK1 or RIOK2 disrupted Akt signaling and caused cell cycle exit, apoptosis, and chemosensitivity in glioblastoma cells by inducing p53 activity through the RpL11-dependent ribosomal stress checkpoint. These results imply that, in glioblastoma cells, constitutive Akt signaling drives RIO kinase overexpression, which creates a feedforward loop that promotes and maintains oncogenic Akt activity through stimulation of mTor signaling. Further study of the RIO kinases as well as other kinases identified in our Drosophila screen may reveal new insights into defects underlying glioblastoma and related cancers and may reveal new therapeutic opportunities for these cancers.


Subject(s)
Cell Transformation, Neoplastic , Glioblastoma , Multiprotein Complexes , Oncogene Protein v-akt , Phosphatidylinositol 3-Kinases , TOR Serine-Threonine Kinases , Animals , Apoptosis/genetics , Astrocytes/cytology , Astrocytes/metabolism , Cell Proliferation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Neoplastic , Genome, Insect , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neuroglia/metabolism , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
13.
Glia ; 59(9): 1364-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21538561

ABSTRACT

Glioblastomas (GBM), the most common primary brain tumors, infiltrate the brain, grow rapidly, and are refractory to current therapies. Signature genetic lesions in glioblastomas include mutation of the epidermal growth factor receptor tyrosine kinase (EGFR) receptor tyrosine kinase and activating mutations in components of the PI-3 kinase (PI3K) pathway. Despite years of study, how these pathways specifically regulate glial pathogenesis is unclear. To address the genetic and cellular origins of this disease, a novel Drosophila GBM model has been developed in which glial progenitor cells give rise to proliferative and invasive neoplastic cells that create transplantable tumors in response to constitutive co-activation of the EGFR-Ras and PI3K pathways. Standing with a rich literature demonstrating the direct relevance of Drosophila to studies on human cancer, neurological disease, and neurodevelopment, this model represents a robust cell-type specific Drosophila neurological disease model in which malignant cells are created by mutations in genetic pathways thought to be driving forces in a homologous human disease. Using lineage analysis and cell-type specific markers, neoplastic glial cells were found to originate from committed glial progenitor cells, rather than from multipotent neuroblasts. Genetic analyses demonstrated that EGFR-Ras and PI3K induce fly glial neoplasia through activation of a combinatorial genetic network composed, in part, of other genetic pathways also commonly mutated in human glioblastomas. In the future, large-scale forward genetic screens with this model may reveal new insights into the origins and treatments of human glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Drosophila melanogaster/physiology , Animals , Biomarkers, Tumor , Central Nervous System/chemistry , Central Nervous System/physiology , Disease Models, Animal , Glioblastoma/pathology , Glioma/pathology , Humans , Neuroglia/pathology , Neuroglia/physiology
14.
PLoS Genet ; 5(2): e1000374, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19214224

ABSTRACT

Gliomas, the most common malignant tumors of the nervous system, frequently harbor mutations that activate the epidermal growth factor receptor (EGFR) and phosphatidylinositol-3 kinase (PI3K) signaling pathways. To investigate the genetic basis of this disease, we developed a glioma model in Drosophila. We found that constitutive coactivation of EGFR-Ras and PI3K pathways in Drosophila glia and glial precursors gives rise to neoplastic, invasive glial cells that create transplantable tumor-like growths, mimicking human glioma. Our model represents a robust organotypic and cell-type-specific Drosophila cancer model in which malignant cells are created by mutations in signature genes and pathways thought to be driving forces in a homologous human cancer. Genetic analyses demonstrated that EGFR and PI3K initiate malignant neoplastic transformation via a combinatorial genetic network composed primarily of other pathways commonly mutated or activated in human glioma, including the Tor, Myc, G1 Cyclins-Cdks, and Rb-E2F pathways. This network acts synergistically to coordinately stimulate cell cycle entry and progression, protein translation, and inappropriate cellular growth and migration. In particular, we found that the fly orthologs of CyclinE, Cdc25, and Myc are key rate-limiting genes required for glial neoplasia. Moreover, orthologs of Sin1, Rictor, and Cdk4 are genes required only for abnormal neoplastic glial proliferation but not for glial development. These and other genes within this network may represent important therapeutic targets in human glioma.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Glioma/genetics , Phosphatidylinositol 3-Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , ErbB Receptors/genetics , Glioma/metabolism , Glioma/pathology , Humans , Mutation , Neuroglia/cytology , Neuroglia/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , ras Proteins/genetics
15.
Genetics ; 171(3): 1057-81, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15965261

ABSTRACT

Dominant mutations in the Ret receptor tyrosine kinase lead to the familial cancer syndrome multiple endocrine neoplasia type 2 (MEN2). Mammalian tissue culture studies suggest that RetMEN2 mutations significantly alter Ret-signaling properties, but the precise mechanisms by which RetMEN2 promotes tumorigenesis remain poorly understood. To determine the signal transduction pathways required for RetMEN2 activity, we analyzed analogous mutations in the Drosophila Ret ortholog dRet. Overexpressed dRetMEN2 isoforms targeted to the developing retina led to aberrant cell proliferation, inappropriate cell fate specification, and excessive Ras pathway activation. Genetic analysis indicated that dRetMEN2 acts through the Ras-ERK, Src, and Jun kinase pathways. A genetic screen for mutations that dominantly suppress or enhance dRetMEN2 phenotypes identified new genes that are required for the phenotypic outcomes of dRetMEN2 activity. Finally, we identified human orthologs for many of these genes and examined their status in human tumors. Two of these loci showed loss of heterozygosity (LOH) within both sporadic and MEN2-associated pheochromocytomas, suggesting that they may contribute to Ret-dependent oncogenesis.


Subject(s)
Disease Models, Animal , Drosophila/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Amino Acid Sequence , Animals , Drosophila/growth & development , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Retina/growth & development , Retina/metabolism , ras Proteins/genetics , ras Proteins/metabolism
16.
Mol Cell Biol ; 24(15): 6676-89, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254235

ABSTRACT

Src family kinases regulate multiple cellular processes including proliferation and oncogenesis. C-terminal Src kinase (Csk) encodes a critical negative regulator of Src family kinases. We demonstrate that the Drosophila melanogaster Csk ortholog, dCsk, functions as a tumor suppressor: dCsk mutants display organ overgrowth and excess cellular proliferation. Genetic analysis indicates that the dCsk(-/-) overgrowth phenotype results from activation of Src, Jun kinase, and STAT signal transduction pathways. In particular, blockade of STAT function in dCsk mutants severely reduced Src-dependent overgrowth and activated apoptosis of mutant tissue. Our data provide in vivo evidence that Src activity requires JNK and STAT function.


Subject(s)
Mitogen-Activated Protein Kinases/chemistry , Protein-Tyrosine Kinases/chemistry , Signal Transduction , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Cycle , Cell Division , DNA-Binding Proteins/genetics , Drosophila , Drosophila Proteins , Expressed Sequence Tags , Flow Cytometry , Genotype , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Larva/metabolism , Microscopy, Electron, Scanning , Models, Biological , Models, Genetic , Mutation , Phenotype , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Time Factors , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...