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











Publication year range
1.
Nat Cancer ; 3(4): 471-485, 2022 04.
Article in English | MEDLINE | ID: mdl-35484422

ABSTRACT

Aberrant expression of MYC transcription factor family members predicts poor clinical outcome in many human cancers. Oncogenic MYC profoundly alters metabolism and mediates an antioxidant response to maintain redox balance. Here we show that MYCN induces massive lipid peroxidation on depletion of cysteine, the rate-limiting amino acid for glutathione (GSH) biosynthesis, and sensitizes cells to ferroptosis, an oxidative, non-apoptotic and iron-dependent type of cell death. The high cysteine demand of MYCN-amplified childhood neuroblastoma is met by uptake and transsulfuration. When uptake is limited, cysteine usage for protein synthesis is maintained at the expense of GSH triggering ferroptosis and potentially contributing to spontaneous tumor regression in low-risk neuroblastomas. Pharmacological inhibition of both cystine uptake and transsulfuration combined with GPX4 inactivation resulted in tumor remission in an orthotopic MYCN-amplified neuroblastoma model. These findings provide a proof of concept of combining multiple ferroptosis targets as a promising therapeutic strategy for aggressive MYCN-amplified tumors.


Subject(s)
Ferroptosis , Neuroblastoma , Cell Death , Child , Cysteine/therapeutic use , Ferroptosis/genetics , Glutathione/therapeutic use , Humans , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics
2.
Commun Biol ; 1: 210, 2018.
Article in English | MEDLINE | ID: mdl-30511023

ABSTRACT

Cytotoxic activities of several Golgi-dispersing compounds including AMF-26/M-COPA, brefeldin A and golgicide A have previously been shown to induce autophagy or apoptosis. Here, we demonstrate that these Golgi disruptors also trigger ferroptosis, a non-apoptotic form of cell death characterized by iron-dependent oxidative degradation of lipids. Inhibitors of ferroptosis not only counteract cell death, but they also protect from Golgi dispersal and inhibition of protein secretion in response to several Golgi stress agents. Furthermore, the application of sublethal doses of ferroptosis-inducers such as erastin and sorafenib, low cystine growth conditions, or genetic knockdown of SLC7A11 and GPX4 all similarly protect cells from Golgi stress and lead to modulation of ACSL4, SLC7A5, SLC7A11 or GPX4 levels. Collectively, this study suggests a previously unrecognized function of the Golgi apparatus, which involves cellular redox control and prevents ferroptotic cell death.

3.
Mol Biol Cell ; 29(1): 42-52, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29118074

ABSTRACT

The secretory pathway is a major determinant of cellular homoeostasis. While research into secretory stress signaling has so far mostly focused on the endoplasmic reticulum (ER), emerging data suggest that the Golgi itself serves as an important signaling hub capable of initiating stress responses. To systematically identify novel Golgi stress mediators, we performed a transcriptomic analysis of cells exposed to three different pharmacological compounds known to elicit Golgi fragmentation: brefeldin A, golgicide A, and monensin. Subsequent gene-set enrichment analysis revealed a significant contribution of the ETS family transcription factors ELK1, GABPA/B, and ETS1 to the control of gene expression following compound treatment. Induction of Golgi stress leads to a late activation of the ETS upstream kinases MEK1/2 and ERK1/2, resulting in enhanced ETS factor activity and the transcription of ETS family target genes related to spliceosome function and cell death induction via alternate MCL1 splicing. Further genetic analyses using loss-of-function and gain-of-function experiments suggest that these transcription factors operate in parallel.


Subject(s)
Alternative Splicing/genetics , Golgi Apparatus/metabolism , MAP Kinase Signaling System , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-ets/metabolism , Stress, Physiological , Transcription, Genetic , A549 Cells , Alternative Splicing/drug effects , Apoptosis/drug effects , Brefeldin A/pharmacology , Cytoprotection/drug effects , Gene Expression Profiling , Gene Knockdown Techniques , Golgi Apparatus/drug effects , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Monensin/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Spliceosomes/drug effects , Spliceosomes/metabolism , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects
4.
Cell Death Dis ; 8(10): e3069, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28981087

ABSTRACT

Disruption of the Golgi apparatus can induce a distinct form of programmed cell death that has not been thoroughly characterized. We found that pharmacological application of Golgi stress leads to induction of death receptors (DRs) 4 and 5. DR4 appears to be primarily responsible for the initiation of cell death downstream of Golgi stress, whereas DR5 seems to be more important for cell death triggered by endoplasmic reticulum (ER) stress in specific cancer cell lines. DR induction downstream of either Golgi or ER stress mainly causes intracellular accumulation of DR4 presumably at the Golgi, rather than increased expression on the cell surface. Nevertheless, cells treated with secretory pathway stressors displayed an increased susceptibility to TRAIL (tumor necrosis factor related apoptosis inducing ligand), the endogenous ligand of DR4/5, probably due to intracellular sequestration of the caspase-8 regulator CFLAR (caspase-8 and FADD-like apoptosis regulator). These findings have implications for the treatment of cancer with DR agonists and our general understanding of DR signaling while highlighting the role of the Golgi apparatus as a cell death signaling platform.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology
5.
Mol Biol Cell ; 28(26): 3756-3772, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29074567

ABSTRACT

The Golgi apparatus is increasingly recognized as a major hub for cellular signaling and is involved in numerous pathologies, including neurodegenerative diseases and cancer. The study of Golgi stress-induced signaling pathways relies on the selectivity of the available tool compounds of which currently only a few are known. To discover novel Golgi-fragmenting agents, transcriptomic profiles of cells treated with brefeldin A, golgicide A, or monensin were generated and compared with a database of gene expression profiles from cells treated with other bioactive small molecules. In parallel, a phenotypic screen was performed for compounds that alter normal Golgi structure. Histone deacetylase (HDAC) inhibitors and DNA-damaging agents were identified as novel Golgi disruptors. Further analysis identified HDAC1/HDAC9 as well as BRD8 and DNA-PK as important regulators of Golgi breakdown mediated by HDAC inhibition. We provide evidence that combinatorial HDACi/(+)-JQ1 treatment spurs synergistic Golgi dispersal in several cancer cell lines, pinpointing a possible link between drug-induced toxicity and Golgi morphology alterations.


Subject(s)
Azepines/pharmacology , Drug Evaluation, Preclinical/methods , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Triazoles/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Gene Expression Profiling/methods , Golgi Apparatus/drug effects , Histone Deacetylase 1/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans
6.
J Cell Sci ; 130(14): 2251-2265, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28536105

ABSTRACT

Tether complexes play important roles in endocytic and exocytic trafficking of lipids and proteins. In yeast, the multisubunit transport protein particle (TRAPP) tether regulates endoplasmic reticulum (ER)-to-Golgi and intra-Golgi transport and is also implicated in autophagy. In addition, the TRAPP complex acts as a guanine nucleotide exchange factor (GEF) for Ypt1, which is homologous to human Rab1a and Rab1b. Here, we show that human TRAPPC13 and other TRAPP subunits are critically involved in the survival response to several Golgi-disrupting agents. Loss of TRAPPC13 partially preserves the secretory pathway and viability in response to brefeldin A, in a manner that is dependent on ARF1 and the large GEF GBF1, and concomitant with reduced caspase activation and ER stress marker induction. TRAPPC13 depletion reduces Rab1a and Rab1b activity, impairs autophagy and leads to increased infectivity to the pathogenic bacterium Shigella flexneri in response to brefeldin A. Thus, our results lend support for the existence of a mammalian TRAPPIII complex containing TRAPPC13, which is important for autophagic flux under certain stress conditions.


Subject(s)
Antigens, Neoplasm/metabolism , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , A549 Cells , ADP-Ribosylation Factor 1/metabolism , Anti-Bacterial Agents/pharmacology , Antigens, Neoplasm/drug effects , Autophagy/physiology , Brefeldin A/pharmacology , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/metabolism , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Shigella flexneri/drug effects , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/drug effects
7.
Traffic ; 18(8): 530-544, 2017 08.
Article in English | MEDLINE | ID: mdl-28485883

ABSTRACT

The Golgi apparatus is part of the secretory pathway and of central importance for modification, transport and sorting of proteins and lipids. ADP-ribosylation factors, whose activation can be blocked by brefeldin A (BFA), play a major role in functioning of the Golgi network and regulation of membrane traffic and are also involved in proliferation and migration of cancer cells. Due to high cytotoxicity and poor bioavailability, BFA has not passed the preclinical stage of drug development. Recently, AMF-26 and golgicide A have been described as novel inhibitors of the Golgi system with antitumor or bactericidal properties. We provide here further evidence that AMF-26 closely mirrors the mode of action of BFA but is less potent. Using several human cancer cell lines, we studied the effects of AMF-26, BFA and golgicide A on cell homeostasis including Golgi structure, endoplasmic reticulum (ER) stress markers, secretion and viability, and found overall a significant correlation between these parameters. Furthermore, modulation of ADP-ribosylation factor expression has a profound impact on Golgi organization and survival in response to Golgi stress inducers.


Subject(s)
Cell Survival , Golgi Apparatus/metabolism , Stress, Physiological , ADP-Ribosylation Factors/drug effects , ADP-Ribosylation Factors/metabolism , Brefeldin A/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Naphthols/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology
8.
Nat Cell Biol ; 15(12): 1473-85, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185178

ABSTRACT

Treatment of cells with brefeldin A (BFA) blocks secretory vesicle transport and causes a collapse of the Golgi apparatus. To gain more insight into the cellular mechanisms mediating BFA toxicity, we conducted a genome-wide haploid genetic screen that led to the identification of the small G protein ADP-ribosylation factor 4 (ARF4). ARF4 depletion preserves viability, Golgi integrity and cargo trafficking in the presence of BFA, and these effects depend on the guanine nucleotide exchange factor GBF1 and other ARF isoforms including ARF1 and ARF5. ARF4 knockdown cells show increased resistance to several human pathogens including Chlamydia trachomatis and Shigella flexneri. Furthermore, ARF4 expression is induced when cells are exposed to several Golgi-disturbing agents and requires the CREB3 (also known as Luman or LZIP) transcription factor, whose downregulation mimics ARF4 loss. Thus, we have uncovered a CREB3-ARF4 signalling cascade that may be part of a Golgi stress response set in motion by stimuli compromising Golgi capacity.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Golgi Apparatus/physiology , Stress, Physiological , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factors/genetics , Brefeldin A/pharmacology , Chlamydia trachomatis/physiology , Disease Susceptibility , Gene Knockdown Techniques , Golgi Apparatus/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Protein Transport/drug effects , RNA, Small Interfering/genetics , Shigella flexneri/physiology , Signal Transduction , Transcriptional Activation , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 108(29): 11756-65, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21677192

ABSTRACT

Tunicamycin (TM) inhibits eukaryotic asparagine-linked glycosylation, protein palmitoylation, ganglioside production, proteoglycan synthesis, 3-hydroxy-3-methylglutaryl coenzyme-A reductase activity, and cell wall biosynthesis in bacteria. Treatment of cells with TM elicits endoplasmic reticulum stress and activates the unfolded protein response. Although widely used in laboratory settings for many years, it is unknown how TM enters cells. Here, we identify in an unbiased genetic screen a transporter of the major facilitator superfamily, major facilitator domain containing 2A (MFSD2A), as a critical mediator of TM toxicity. Cells without MFSD2A are TM-resistant, whereas MFSD2A-overexpressing cells are hypersensitive. Hypersensitivity is associated with increased cellular TM uptake concomitant with an enhanced endoplasmic reticulum stress response. Furthermore, MFSD2A mutant analysis reveals an important function of the C terminus for correct intracellular localization and protein stability, and it identifies transmembrane helical amino acid residues essential for mediating TM sensitivity. Overall, our data uncover a critical role for MFSD2A by acting as a putative TM transporter at the plasma membrane.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Transport Proteins/metabolism , Protein Folding/drug effects , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Tunicamycin/pharmacology , Blotting, Western , Cell Line , Cell Survival , Glycosylation/drug effects , Humans , Immunoprecipitation , Microscopy, Confocal , Symporters , Tumor Suppressor Proteins/genetics
10.
Nature ; 462(7269): 108-12, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19847166

ABSTRACT

The proto-oncogene KRAS is mutated in a wide array of human cancers, most of which are aggressive and respond poorly to standard therapies. Although the identification of specific oncogenes has led to the development of clinically effective, molecularly targeted therapies in some cases, KRAS has remained refractory to this approach. A complementary strategy for targeting KRAS is to identify gene products that, when inhibited, result in cell death only in the presence of an oncogenic allele. Here we have used systematic RNA interference to detect synthetic lethal partners of oncogenic KRAS and found that the non-canonical IkappaB kinase TBK1 was selectively essential in cells that contain mutant KRAS. Suppression of TBK1 induced apoptosis specifically in human cancer cell lines that depend on oncogenic KRAS expression. In these cells, TBK1 activated NF-kappaB anti-apoptotic signals involving c-Rel and BCL-XL (also known as BCL2L1) that were essential for survival, providing mechanistic insights into this synthetic lethal interaction. These observations indicate that TBK1 and NF-kappaB signalling are essential in KRAS mutant tumours, and establish a general approach for the rational identification of co-dependent pathways in cancer.


Subject(s)
Genes, ras/genetics , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Alleles , Apoptosis , Cell Line, Tumor , Cell Survival , Gene Expression Profiling , Genes, Lethal , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins c-rel/metabolism , Signal Transduction , bcl-X Protein/metabolism
11.
Cancer Cell ; 16(1): 21-32, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19573809

ABSTRACT

Dysregulation of the phosphatidylinositol 3-kinase (PI3K) signaling pathway occurs frequently in human cancer. PTEN tumor suppressor or PIK3CA oncogene mutations both direct PI3K-dependent tumorigenesis largely through activation of the AKT/PKB kinase. However, here we show through phosphoprotein profiling and functional genomic studies that many PIK3CA mutant cancer cell lines and human breast tumors exhibit only minimal AKT activation and a diminished reliance on AKT for anchorage-independent growth. Instead, these cells retain robust PDK1 activation and membrane localization and exhibit dependency on the PDK1 substrate SGK3. SGK3 undergoes PI3K- and PDK1-dependent activation in PIK3CA mutant cancer cells. Thus, PI3K may promote cancer through both AKT-dependent and AKT-independent mechanisms. Knowledge of differential PI3K/PDK1 signaling could inform rational therapeutics in cancers harboring PIK3CA mutations.


Subject(s)
Breast Neoplasms/genetics , Mutation , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/physiology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Survival/genetics , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation , Female , Gene Expression Profiling , Humans , Neoplasms/metabolism , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction/genetics
12.
Mol Cell ; 29(5): 533-5, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18342598

ABSTRACT

In this issue of Molecular Cell, Ozcan et al. (2008) show that the loss of the tuberous sclerosis tumor suppressor complex induces endoplasmic reticulum stress, leading to attenuation of insulin receptor signaling activity via the unfolded protein response.


Subject(s)
Oxidative Stress , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Endoplasmic Reticulum/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Neoplasms/metabolism , Proteins , Receptor, Insulin/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Trends Pharmacol Sci ; 28(4): 149-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17350110

ABSTRACT

Evidence of off-target effects (OTEs) associated with small interfering (si)RNAs (19-29bp) in mammalian cells has existed for several years. Two recent articles demonstrate that short sequences within long double-stranded (ds)RNAs frequently cause undesirable OTEs in cultured Drosophila cells. These results reveal the potential for high false-positive rates in RNA interference (RNAi) screens using long dsRNAs and highlight the need for screening with multiple, non-overlapping long dsRNAs or siRNAs. Discovering multiple potent siRNAs with minimal off-target profiles for each target transcript will be invaluable for genome-based studies of gene function and for personalized RNAi therapeutics.


Subject(s)
Drosophila/genetics , RNA Interference , RNA, Double-Stranded/genetics , Animals , Gene Library , Genes, Insect/genetics
14.
Curr Biol ; 15(1): 24-30, 2005 Jan 11.
Article in English | MEDLINE | ID: mdl-15649360

ABSTRACT

The control of cellular growth is tightly linked to the regulation of protein synthesis. A key function in translation initiation is fulfilled by the 5' cap binding eukaryotic initiation factor 4E (eIF4E), and dysregulation of eIF4E is associated with malignant transformation and tumorigenesis . In mammals, the activity of eIF4E is modulated by phosphorylation at Ser209 by mitogen-activated protein kinases (MAPK)-interacting kinases 1 and 2 (Mnk1 and Mnk2) , which themselves are activated by ERK and p38 MAPK in response to mitogens, cytokines or cellular stress . Whether phosphorylation of eIF4E at Ser209 exerts a positive or inhibitory effect on translation efficiency has remained controversial. Here we provide a genetic characterization of the Drosophila homolog of Mnk1/2, Lk6. Lk6 function is dispensable under a high protein diet, consistent with the recent finding that mice lacking both Mnk1 and Mnk2 are not growth-impaired . Interestingly, loss of Lk6 function causes a significant growth reduction when the amino acid content in the diet is reduced. Overexpression of Lk6 also results in growth inhibition in an eIF4E-dependent manner. We propose a model of eIF4E regulation that may reconcile the contradictory findings with regard to the role of phosphorylation by Mnk1/2.


Subject(s)
Diet , Drosophila/growth & development , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Biosynthesis/physiology , Amino Acid Sequence , Amino Acids/metabolism , Animals , Body Weights and Measures , Drosophila/metabolism , Drosophila Proteins , Female , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Models, Biological , Molecular Sequence Data , Mutagenesis, Insertional , Oxidative Stress/genetics , Phosphorylation , Sequence Alignment , Wings, Animal/growth & development
15.
Genes Dev ; 18(23): 2893-904, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15545625

ABSTRACT

Mammalian target of rapamycin (mTOR) is a central regulator of protein synthesis whose activity is modulated by a variety of signals. Energy depletion and hypoxia result in mTOR inhibition. While energy depletion inhibits mTOR through a process involving the activation of AMP-activated protein kinase (AMPK) by LKB1 and subsequent phosphorylation of TSC2, the mechanism of mTOR inhibition by hypoxia is not known. Here we show that mTOR inhibition by hypoxia requires the TSC1/TSC2 tumor suppressor complex and the hypoxia-inducible gene REDD1/RTP801. Disruption of the TSC1/TSC2 complex through loss of TSC1 or TSC2 blocks the effects of hypoxia on mTOR, as measured by changes in the mTOR targets S6K and 4E-BP1, and results in abnormal accumulation of Hypoxia-inducible factor (HIF). In contrast to energy depletion, mTOR inhibition by hypoxia does not require AMPK or LKB1. Down-regulation of mTOR activity by hypoxia requires de novo mRNA synthesis and correlates with increased expression of the hypoxia-inducible REDD1 gene. Disruption of REDD1 abrogates the hypoxia-induced inhibition of mTOR, and REDD1 overexpression is sufficient to down-regulate S6K phosphorylation in a TSC1/TSC2-dependent manner. Inhibition of mTOR function by hypoxia is likely to be important for tumor suppression as TSC2-deficient cells maintain abnormally high levels of cell proliferation under hypoxia.


Subject(s)
Hypoxia/physiopathology , Protein Kinases/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , 3T3 Cells , Animals , Cell Division/physiology , Down-Regulation , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
16.
Genes Dev ; 18(23): 2879-92, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15545626

ABSTRACT

Diverse extrinsic and intrinsic cues must be integrated within a developing organism to ensure appropriate growth at the cellular and organismal level. In Drosophila, the insulin receptor/TOR/S6K signaling network plays a fundamental role in the control of metabolism and cell growth. Here we show that scylla and charybdis, two homologous genes identified as growth suppressors in an EP (enhancer/promoter) overexpression screen, act as negative regulators of growth. The simultaneous loss of both genes generates flies that are more susceptible to reduced oxygen concentrations (hypoxia) and that show mild overgrowth phenotypes. Conversely, scylla or charybdis overactivation reduces growth. Growth inhibition is associated with a reduction in S6K but not PKB/Akt activity. Together, genetic and biochemical analysis places Scylla/Charybdis downstream of PKB and upstream of TSC. Furthermore, we show that scylla and charybdis are induced under hypoxic conditions and that scylla is a target of Drosophila HIF-1 (hypoxia-inducible factor-1) like its mammalian counterpart RTP801/REDD1, thus establishing a potential cross-talk between growth and oxygen sensing.


Subject(s)
Drosophila Proteins/physiology , Drosophila/genetics , Hypoxia/metabolism , Ribosomal Protein S6 Kinases/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Drosophila Proteins/biosynthesis , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL