Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38721669

ABSTRACT

Accounting for 10-20% of breast cancer cases, triple-negative breast cancer (TNBC) is associated with a disproportionate number of breast cancer deaths. One challenge in studying TNBC is its genomic profile: with the exception of TP53 loss, most breast cancer tumors are characterized by a high number of copy number alterations (CNAs), making modeling the disease in whole animals challenging. We computationally analyzed 186 CNA regions previously identified in breast cancer tumors to rank genes within each region by likelihood of acting as a tumor driver. We then used a Drosophila p53-Myc TNBC model to identify 48 genes as functional drivers. To demonstrate the utility of this functional database, we established six 3-hit models; altering candidate genes led to increased aspects of transformation as well as resistance to the chemotherapeutic drug fluorouracil. Our work provides a functional database of CNA-associated TNBC drivers, and a template for an integrated computational/whole-animal approach to identify functional drivers of transformation and drug resistance within CNAs in other tumor types.


Subject(s)
DNA Copy Number Variations , Disease Models, Animal , Triple Negative Breast Neoplasms , Animals , DNA Copy Number Variations/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Female , Drosophila melanogaster/genetics , Humans , Drug Resistance, Neoplasm/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Gene Expression Regulation, Neoplastic , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Cell Transformation, Neoplastic/genetics
2.
Oncogenesis ; 13(1): 1, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38172609

ABSTRACT

Throughout an individual's life, somatic cells acquire cancer-associated mutations. A fraction of these mutations trigger tumour formation, a phenomenon partly driven by the interplay of mutant and wild-type cell clones competing for dominance; conversely, other mutations function against tumour initiation. This mechanism of 'cell competition', can shift clone dynamics by evaluating the relative status of clonal populations, promoting 'winners' and eliminating 'losers'. This review examines the role of cell competition in the context of tumorigenesis, tumour progression and therapeutic intervention.

3.
J Invest Dermatol ; 143(8): 1378-1387, 2023 08.
Article in English | MEDLINE | ID: mdl-37330719

ABSTRACT

Neurofibromatosis type 1 (NF1) is caused by a nonfunctional copy of the NF1 tumor suppressor gene that predisposes patients to the development of cutaneous neurofibromas (cNFs), the skin tumor that is the hallmark of this condition. Innumerable benign cNFs, each appearing by an independent somatic inactivation of the remaining functional NF1 allele, form in nearly all patients with NF1. One of the limitations in developing a treatment for cNFs is an incomplete understanding of the underlying pathophysiology and limitations in experimental modeling. Recent advances in preclinical in vitro and in vivo modeling have substantially enhanced our understanding of cNF biology and created unprecedented opportunities for therapeutic discovery. We discuss the current state of cNF preclinical in vitro and in vivo model systems, including two- and three-dimensional cell cultures, organoids, genetically engineered mice, patient-derived xenografts, and porcine models. We highlight the models' relationship to human cNFs and how they can be used to gain insight into cNF development and therapeutic discovery.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Skin Neoplasms , Mice , Humans , Animals , Swine , Neurofibromatosis 1/genetics , Neurofibromatosis 1/therapy , Mutation , Neurofibroma/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Alleles
4.
PLoS Genet ; 19(6): e1010792, 2023 06.
Article in English | MEDLINE | ID: mdl-37267433

ABSTRACT

Experimental models that capture the genetic complexity of human disease and allow mechanistic explorations of the underlying cell, tissue, and organ interactions are crucial to furthering our understanding of disease biology. Such models require combinatorial manipulations of multiple genes, often in more than one tissue at once. The ability to perform complex genetic manipulations in vivo is a key strength of Drosophila, where many tools for sophisticated and orthogonal genetic perturbations exist. However, combining the large number of transgenes required to establish more representative disease models and conducting mechanistic studies in these already complex genetic backgrounds is challenging. Here we present a design that pushes the limits of Drosophila genetics by allowing targeted combinatorial ectopic expression and knockdown of multiple genes from a single inducible transgene. The polycistronic transcript encoded by this transgene includes a synthetic short hairpin cluster cloned within an intron placed at the 5' end of the transcript, followed by two protein-coding sequences separated by the T2A sequence that mediates ribosome skipping. This technology is particularly useful for modeling genetically complex diseases like cancer, which typically involve concurrent activation of multiple oncogenes and loss of multiple tumor suppressors. Furthermore, consolidating multiple genetic perturbations into a single transgene further streamlines the ability to perform combinatorial genetic manipulations and makes it readily adaptable to a broad palette of transgenic systems. This flexible design for combinatorial genetic perturbations will also be a valuable tool for functionally exploring multigenic gene signatures identified from omics studies of human disease and creating humanized Drosophila models to characterize disease-associated variants in human genes. It can also be adapted for studying biological processes underlying normal tissue homeostasis and development that require simultaneous manipulation of many genes.


Subject(s)
Drosophila , Genetic Techniques , Animals , Humans , Drosophila/genetics , Transgenes , Animals, Genetically Modified , Introns
5.
J Invest Dermatol ; 143(8): 1358-1368, 2023 08.
Article in English | MEDLINE | ID: mdl-37245145

ABSTRACT

Cutaneous neurofibromas (cNFs) are the most common tumor in people with the rasopathy neurofibromatosis type 1. They number in hundreds or even thousands throughout the body, and currently, there are no effective interventions to prevent or treat these skin tumors. To facilitate the identification of novel and effective therapies, essential studies including a more refined understanding of cNF biology and the role of RAS signaling and downstream effector pathways responsible for cNF initiation, growth, and maintenance are needed. This review highlights the current state of knowledge of RAS signaling in cNF pathogenesis and therapeutic development for cNF treatment.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Skin Neoplasms , Humans , Neurofibroma/metabolism , Neurofibroma/pathology , Neurofibromatosis 1/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Signal Transduction
6.
iScience ; 24(3): 102212, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33733072

ABSTRACT

Adenoid cystic carcinoma (ACC) is a rare cancer type that originates in the salivary glands. Tumors commonly invade along nerve tracks in the head and neck, making surgery challenging. Follow-up treatments for recurrence or metastasis including chemotherapy and targeted therapies have shown limited efficacy, emphasizing the need for new therapies. Here, we report a Drosophila-based therapeutic approach for a patient with advanced ACC disease. A patient-specific Drosophila transgenic line was developed to model the five major variants associated with the patient's disease. Robotics-based screening identified a three-drug cocktail-vorinostat, pindolol, tofacitinib-that rescued transgene-mediated lethality in the Drosophila patient-specific line. Patient treatment led to a sustained stabilization and a partial metabolic response of 12 months. Subsequent resistance was associated with new genomic amplifications and deletions. Given the lack of options for patients with ACC, our data suggest that this approach may prove useful for identifying novel therapeutic candidates.

7.
Sci Rep ; 11(1): 1111, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441820

ABSTRACT

Human papillomavirus (HPV) is the leading cause of cervical cancer and has been implicated in several other cancer types including vaginal, vulvar, penile, and oropharyngeal cancers. Despite the recent availability of a vaccine, there are still over 310,000 deaths each year worldwide. Current treatments for HPV-mediated cancers show limited efficacy, and would benefit from improved understanding of disease mechanisms. Recently, we developed a Drosophila 'HPV 18 E6' model that displayed loss of cellular morphology and polarity, junctional disorganization, and degradation of the major E6 target Magi; we further provided evidence that mechanisms underlying HPV E6-induced cellular abnormalities are conserved between humans and flies. Here, we report a functional genetic screen of the Drosophila kinome that identified IKK[Formula: see text]-a regulator of NF-κB-as an enhancer of E6-induced cellular defects. We demonstrate that inhibition of IKK[Formula: see text] reduces Magi degradation and that this effect correlates with hyperphosphorylation of E6. Further, the reduction in IKK[Formula: see text] suppressed the cellular transformation caused by the cooperative action of HPVE6 and the oncogenic Ras. Finally, we demonstrate that the interaction between IKK[Formula: see text] and E6 is conserved in human cells: inhibition of IKK[Formula: see text] blocked the growth of cervical cancer cells, suggesting that IKK[Formula: see text] may serve as a novel therapeutic target for HPV-mediated cancers.


Subject(s)
Compound Eye, Arthropod/abnormalities , DNA-Binding Proteins/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Oncogene Proteins, Viral/metabolism , Uterine Cervical Neoplasms/pathology , Animals , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Viral , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/metabolism , Drosophila , Female , Humans , Nucleoside-Phosphate Kinase/metabolism , PDZ Domains , Phosphorylation , Proteolysis , Ubiquitin-Protein Ligases/metabolism
8.
Dev Cell ; 49(3): 317-324, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063751

ABSTRACT

Cancer has joined heart disease as the leading source of mortality in the US. In an era of organoids, patient-derived xenografts, and organs on a chip, model organisms continue to thrive with a combination of powerful genetic tools, rapid pace of discovery, and affordability. Model organisms enable the analysis of both the tumor and its associated microenvironment, aspects that are particularly relevant to our understanding of metastasis and drug resistance. In this Perspective, we explore some of the strengths of fruit flies and zebrafish for addressing fundamental cancer questions and how these two organisms can contribute to identifying promising therapeutic candidates.


Subject(s)
Disease Models, Animal , Neoplasms/metabolism , Neoplasms/pathology , Animals , Drosophila melanogaster , Humans , Organoids/metabolism , Organoids/physiology , Tumor Microenvironment/physiology , Zebrafish
9.
Nucleic Acids Res ; 47(W1): W183-W190, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31069376

ABSTRACT

High-throughput experiments produce increasingly large datasets that are difficult to analyze and integrate. While most data integration approaches focus on aligning metadata, data integration can be achieved by abstracting experimental results into gene sets. Such gene sets can be made available for reuse through gene set enrichment analysis tools such as Enrichr. Enrichr currently only supports gene sets compiled from human and mouse, limiting accessibility for investigators that study other model organisms. modEnrichr is an expansion of Enrichr for four model organisms: fish, fly, worm and yeast. The gene set libraries within FishEnrichr, FlyEnrichr, WormEnrichr and YeastEnrichr are created from the Gene Ontology, mRNA expression profiles, GeneRIF, pathway databases, protein domain databases and other organism-specific resources. Additionally, libraries were created by predicting gene function from RNA-seq co-expression data processed uniformly from the gene expression omnibus for each organism. The modEnrichr suite of tools provides the ability to convert gene lists across species using an ortholog conversion tool that automatically detects the species. For complex analyses, modEnrichr provides API access that enables submitting batch queries. In summary, modEnrichr leverages existing model organism databases and other resources to facilitate comprehensive hypothesis generation through data integration.


Subject(s)
Databases, Genetic , Gene Expression/genetics , Gene Library , Genomic Library , Software , Animals , Computational Biology , Gene Ontology , Humans , Metadata
10.
Sci Adv ; 5(5): eaav6528, 2019 05.
Article in English | MEDLINE | ID: mdl-31131321

ABSTRACT

Colorectal cancer remains a leading source of cancer mortality worldwide. Initial response is often followed by emergent resistance that is poorly responsive to targeted therapies, reflecting currently undruggable cancer drivers such as KRAS and overall genomic complexity. Here, we report a novel approach to developing a personalized therapy for a patient with treatment-resistant metastatic KRAS-mutant colorectal cancer. An extensive genomic analysis of the tumor's genomic landscape identified nine key drivers. A transgenic model that altered orthologs of these nine genes in the Drosophila hindgut was developed; a robotics-based screen using this platform identified trametinib plus zoledronate as a candidate treatment combination. Treating the patient led to a significant response: Target and nontarget lesions displayed a strong partial response and remained stable for 11 months. By addressing a disease's genomic complexity, this personalized approach may provide an alternative treatment option for recalcitrant disease such as KRAS-mutant colorectal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, ras , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Zoledronic Acid/administration & dosage , Animals , Colorectal Neoplasms/pathology , Disease Progression , Drosophila/genetics , Drug Administration Schedule , Drug Screening Assays, Antitumor , Female , Genomics , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis , Precision Medicine
11.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Article in English | MEDLINE | ID: mdl-31026276

ABSTRACT

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine , Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinases , Thyroid Neoplasms , Animals , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/metabolism , Computational Biology/methods , Drosophila , Models, Biological , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/metabolism , Protein Kinases/drug effects , Protein Kinases/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/metabolism
12.
Cancer Res ; 78(15): 4344-4359, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29844121

ABSTRACT

A key tool of cancer therapy has been targeted inhibition of oncogene-addicted pathways. However, efficacy has been limited by progressive emergence of resistance as transformed cells adapt. Here, we use Drosophila to dissect response to targeted therapies. Treatment with a range of kinase inhibitors led to hyperactivation of overall cellular networks, resulting in emergent resistance and expression of stem cell markers, including Sox2. Genetic and drug screens revealed that inhibitors of histone deacetylases, proteasome, and Hsp90 family of proteins restrained this network hyperactivation. These "network brake" cocktails, used as adjuncts, prevented emergent resistance and promoted cell death at subtherapeutic doses. Our results highlight a general response of cells, transformed and normal, to targeted therapies that leads to resistance and toxicity. Pairing targeted therapeutics with subtherapeutic doses of broad-acting "network brake" drugs may provide a means of extending therapeutic utility while reducing whole body toxicity.Significance: These findings with a strong therapeutic potential provide an innovative approach of identifying effective combination treatments for cancer. Cancer Res; 78(15); 4344-59. ©2018 AACR.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Drosophila/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasms/metabolism , SOXB1 Transcription Factors/metabolism
13.
Endocr Relat Cancer ; 25(2): T91-T104, 2018 02.
Article in English | MEDLINE | ID: mdl-29348307

ABSTRACT

Twenty-five years ago, RET was identified as the primary driver of multiple endocrine neoplasia type 2 (MEN2) syndrome. MEN2 is characterized by several transformation events including pheochromocytoma, parathyroid adenoma and, especially penetrant, medullary thyroid carcinoma (MTC). Overall, MTC is a rare but aggressive type of thyroid cancer for which no effective treatment currently exists. Surgery, radiation, radioisotope treatment and chemotherapeutics have all shown limited success, and none of these approaches have proven durable in advanced disease. Non-mammalian models that incorporate the oncogenic RET isoforms associated with MEN2 and other RET-associated diseases have been useful in delineating mechanisms underlying disease progression. These models have also identified novel targeted therapies as single agents and as combinations. These studies highlight the importance of modeling disease in the context of the whole animal, accounting for the complex interplay between tumor and normal cells in controlling disease progression as well as response to therapy. With convenient access to whole genome sequencing data from expanded thyroid cancer patient cohorts, non-mammalian models will become more complex, sophisticated and continue to complement future mammalian studies. In this review, we explore the contributions of non-mammalian models to our understanding of thyroid cancer including MTC, with a focus on Danio rerio and Drosophila melanogaster (fish and fly) models.


Subject(s)
Disease Models, Animal , Multiple Endocrine Neoplasia Type 2a , Animals , Humans , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
14.
Nat Chem Biol ; 14(3): 291-298, 2018 03.
Article in English | MEDLINE | ID: mdl-29355849

ABSTRACT

Synthetic tailoring of approved drugs for new indications is often difficult, as the most appropriate targets may not be readily apparent, and therefore few roadmaps exist to guide chemistry. Here, we report a multidisciplinary approach for accessing novel target and chemical space starting from an FDA-approved kinase inhibitor. By combining chemical and genetic modifier screening with computational modeling, we identify distinct kinases that strongly enhance ('pro-targets') or limit ('anti-targets') whole-animal activity of the clinical kinase inhibitor sorafenib in a Drosophila medullary thyroid carcinoma (MTC) model. We demonstrate that RAF-the original intended sorafenib target-and MKNK kinases function as pharmacological liabilities because of inhibitor-induced transactivation and negative feedback, respectively. Through progressive synthetic refinement, we report a new class of 'tumor calibrated inhibitors' with unique polypharmacology and strongly improved therapeutic index in fly and human MTC xenograft models. This platform provides a rational approach to creating new high-efficacy and low-toxicity drugs.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Carcinoma/metabolism , Drosophila/metabolism , Protein Kinase Inhibitors/pharmacology , Thyroid Neoplasms/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Drug Design , Female , HCT116 Cells , Humans , Male , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Neoplasm Transplantation , Protein Isoforms , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , Sorafenib/pharmacology
16.
Nat Commun ; 7: 13615, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27897178

ABSTRACT

The multigenic nature of human tumours presents a fundamental challenge for cancer drug discovery. Here we use Drosophila to generate 32 multigenic models of colon cancer using patient data from The Cancer Genome Atlas. These models recapitulate key features of human cancer, often as emergent properties of multigenic combinations. Multigenic models such as ras p53 pten apc exhibit emergent resistance to a panel of cancer-relevant drugs. Exploring one drug in detail, we identify a mechanism of resistance for the PI3K pathway inhibitor BEZ235. We use this data to identify a combinatorial therapy that circumvents this resistance through a two-step process of emergent pathway dependence and sensitivity we term 'induced dependence'. This approach is effective in cultured human tumour cells, xenografts and mouse models of colorectal cancer. These data demonstrate how multigenic animal models that reference cancer genomes can provide an effective approach for developing novel targeted therapies.


Subject(s)
Colorectal Neoplasms/genetics , Drosophila melanogaster/genetics , Genome , Genomics , Acetates/pharmacology , Acetates/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzopyrans/pharmacology , Benzopyrans/therapeutic use , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epistasis, Genetic/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Signal Transduction/drug effects
17.
Cell Rep ; 16(11): 3052-3061, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27626672

ABSTRACT

We generated and compared Drosophila models of RET fusions CCDC6-RET and NCOA4-RET. Both RET fusions directed cells to migrate, delaminate, and undergo EMT, and both resulted in lethality when broadly expressed. In all phenotypes examined, NCOA4-RET was more severe than CCDC6-RET, mirroring their effects on patients. A functional screen against the Drosophila kinome and a library of cancer drugs found that CCDC6-RET and NCOA4-RET acted through different signaling networks and displayed distinct drug sensitivities. Combining data from the kinome and drug screens identified the WEE1 inhibitor AZD1775 plus the multi-kinase inhibitor sorafenib as a synergistic drug combination that is specific for NCOA4-RET. Our work emphasizes the importance of identifying and tailoring a patient's treatment to their specific RET fusion isoform and identifies a multi-targeted therapy that may prove effective against tumors containing the NCOA4-RET fusion.


Subject(s)
Drosophila melanogaster/metabolism , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Animals , Cell Movement/drug effects , Disease Models, Animal , Drosophila melanogaster/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Up-Regulation/drug effects
18.
Cell Rep ; 14(6): 1477-1487, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26832408

ABSTRACT

We have developed a Drosophila lung cancer model by targeting Ras1(G12V)--alone or in combination with PTEN knockdown--to the Drosophila tracheal system. This led to overproliferation of tracheal tissue, formation of tumor-like growths, and animal lethality. Screening a library of FDA-approved drugs identified several that improved overall animal survival. We explored two hits: the MEK inhibitor trametinib and the HMG-CoA reductase inhibitor fluvastatin. Oral administration of these drugs inhibited Ras and PI3K pathway activity, respectively; in addition, fluvastatin inhibited protein prenylation downstream of HMG-CoA reductase to promote survival. Combining drugs led to synergistic suppression of tumor formation and rescue lethality; similar synergy was observed in human A549 lung adenocarcinoma cells. Notably, fluvastatin acted both within transformed cells and also to reduce whole-body trametinib toxicity in flies. Our work supports and provides further context for exploring the potential of combining statins with MAPK inhibitors such as trametinib to improve overall therapeutic index.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Fatty Acids, Monounsaturated/pharmacology , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Lung Neoplasms/drug therapy , Pyridones/pharmacology , Pyrimidinones/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drug Combinations , Drug Synergism , Fluvastatin , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Survival Rate
19.
Elife ; 4: e08501, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26573956

ABSTRACT

Cancer cells demand excessive nutrients to support their proliferation but how cancer cells sense and promote growth in the nutrient favorable conditions remain incompletely understood. Epidemiological studies have indicated that obesity is a risk factor for various types of cancers. Feeding Drosophila a high dietary sugar was previously demonstrated to not only direct metabolic defects including obesity and organismal insulin resistance, but also transform Ras/Src-activated cells into aggressive tumors. Here we demonstrate that Ras/Src-activated cells are sensitive to perturbations in the Hippo signaling pathway. We provide evidence that nutritional cues activate Salt-inducible kinase, leading to Hippo pathway downregulation in Ras/Src-activated cells. The result is Yorkie-dependent increase in Wingless signaling, a key mediator that promotes diet-enhanced Ras/Src-tumorigenesis in an otherwise insulin-resistant environment. Through this mechanism, Ras/Src-activated cells are positioned to efficiently respond to nutritional signals and ensure tumor growth upon nutrient rich condition including obesity.


Subject(s)
Carcinogenesis , Dietary Sucrose/metabolism , Drosophila/physiology , Phosphotransferases/metabolism , Salts/metabolism , Signal Transduction , Animals , Cell Proliferation , Cells, Cultured
20.
Cell Rep ; 12(4): 636-47, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26190114

ABSTRACT

Diabetic nephropathy is a major cause of end-stage kidney disease. Characterized by progressive microvascular disease, most efforts have focused on injury to the glomerular endothelium. Recent work has suggested a role for the podocyte, a highly specialized component of the glomerular filtration barrier. Here, we demonstrate that the Drosophila nephrocyte, a cell analogous to the mammalian podocyte, displays defects that phenocopy aspects of diabetic nephropathy in animals fed chronic high dietary sucrose. Through functional studies, we identify an OGT-Polycomb-Knot-Sns pathway that links dietary sucrose to loss of the Nephrin ortholog Sns. Reducing OGT through genetic or drug means is sufficient to rescue loss of Sns, leading to overall extension of lifespan. We demonstrate upregulation of the Knot ortholog EBF2 in glomeruli of human diabetic nephropathy patients and a mouse ob/ob diabetes model. Furthermore, we demonstrate rescue of Nephrin expression and cell viability in ebf2(-/-) primary podocytes cultured in high glucose.


Subject(s)
Diabetic Nephropathies/metabolism , Podocytes/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Dietary Carbohydrates/adverse effects , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Podocytes/pathology , Podocytes/physiology , Sucrose/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL