Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 19(5): e1011308, 2023 05.
Article in English | MEDLINE | ID: mdl-37126534

ABSTRACT

The global spread of the SARS-CoV-2 virus has resulted in emergence of lineages which impact the effectiveness of immunotherapies and vaccines that are based on the early Wuhan isolate. All currently approved vaccines employ the spike protein S, as it is the target for neutralizing antibodies. Here we describe two SARS-CoV-2 isolates with unusually large deletions in the N-terminal domain (NTD) of the spike. Cryo-EM structural analysis shows that the deletions result in complete reshaping of the NTD supersite, an antigenically important region of the NTD. For both spike variants the remodeling of the NTD negatively affects binding of all tested NTD-specific antibodies in and outside of the NTD supersite. For one of the variants, we observed a P9L mediated shift of the signal peptide cleavage site resulting in the loss of a disulfide-bridge; a unique escape mechanism with high antigenic impact. Although the observed deletions and disulfide mutations are rare, similar modifications have become independently established in several other lineages, indicating a possibility to become more dominant in the future. The observed plasticity of the NTD foreshadows its broad potential for immune escape with the continued spread of SARS-CoV-2.


Subject(s)
COVID-19 , Humans , SARS-CoV-2/genetics , Antibodies, Neutralizing , Disulfides , Immunotherapy , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral
2.
iScience ; 25(4): 104056, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35345457

ABSTRACT

Castration-resistant prostate cancer (CRPC) is incurable and remains a significant worldwide challenge (Oakes and Papa, 2015). Matched untargeted multi-level omic datasets may reveal biological changes driving CRPC, identifying novel biomarkers and/or therapeutic targets. Untargeted RNA sequencing, proteomics, and metabolomics were performed on xenografts derived from three independent sets of hormone naive and matched CRPC human cell line models of local, lymph node, and bone metastasis grown as murine orthografts. Collectively, we tested the feasibility of muti-omics analysis on models of CRPC in revealing pathways of interest for future validation investigation. Untargeted metabolomics revealed NAA and NAAG commonly accumulating in CRPC across three independent models and proteomics showed upregulation of related enzymes, namely N-acetylated alpha-linked acidic dipeptidases (FOLH1/NAALADL2). Based on pathway analysis integrating multiple omic levels, we hypothesize that increased NAA in CRPC may be due to upregulation of NAAG hydrolysis via NAALADLases providing a pool of acetyl Co-A for upregulated sphingolipid metabolism and a pool of glutamate and aspartate for nucleotide synthesis during tumor growth.

3.
Front Microbiol ; 11: 574872, 2020.
Article in English | MEDLINE | ID: mdl-33042085

ABSTRACT

The molecule guanosine tetraphophosphate (ppGpp) is most commonly considered an alarmone produced during acute stress. However, ppGpp is also present at low concentrations during steady-state growth. Whether ppGpp controls the same cellular targets at both low and high concentrations remains an open question and is vital for understanding growth rate regulation. It is widely assumed that basal ppGpp concentrations vary inversely with growth rate, and that the main function of basal ppGpp is to regulate transcription of ribosomal RNA in response to environmental conditions. Unfortunately, studies to confirm this relationship and to define regulatory targets of basal ppGpp are limited by difficulties in quantifying basal ppGpp. In this Perspective we compare reported concentrations of basal ppGpp in E. coli and quantify ppGpp within several strains using a recently developed analytical method. We find that although the inverse correlation between ppGpp and growth rate is robust across strains and analytical methods, absolute ppGpp concentrations do not absolutely determine RNA synthesis rates. In addition, we investigated the consequences of two separate RNA polymerase mutations that each individually reduce (but do not abolish) sensitivity to ppGpp and find that the relationship between ppGpp, growth rate, and RNA content of single-site mutants remains unaffected. Both literature and our new data suggest that environmental conditions may be communicated to RNA polymerase via an additional regulator. We conclude that basal ppGpp is one of potentially several agents controlling ribosome abundance and DNA replication initiation, but that evidence for additional roles in controlling macromolecular synthesis requires further study.

4.
mBio ; 11(4)2020 08 18.
Article in English | MEDLINE | ID: mdl-32817111

ABSTRACT

Every cell must produce enough membrane to contain itself. However, the mechanisms by which the rate of membrane synthesis is coupled with the rate of cell growth remain unresolved. By comparing substrate and enzyme concentrations of the fatty acid and phospholipid synthesis pathways of Escherichia coli across a 3-fold range of carbon-limited growth rates, we show that the rate of membrane phospholipid synthesis during steady-state growth is determined principally through allosteric control of a single enzyme, PlsB. Due to feedback regulation of the fatty acid pathway, PlsB activity also indirectly controls synthesis of lipopolysaccharide, a major component of the outer membrane synthesized from a fatty acid synthesis intermediate. Surprisingly, concentrations of the enzyme that catalyzes the committed step of lipopolysaccharide synthesis (LpxC) do not differ across steady-state growth conditions, suggesting that steady-state lipopolysaccharide synthesis is modulated primarily via indirect control by PlsB. In contrast to steady-state regulation, we found that responses to environmental perturbations are triggered directly via changes in acetyl coenzyme A (acetyl-CoA) concentrations, which enable rapid adaptation. Adaptations are further modulated by ppGpp, which regulates PlsB activity during slow growth and growth arrest. The strong reliance of the membrane synthesis pathway upon posttranslational regulation ensures both the reliability and the responsiveness of membrane synthesis.IMPORTANCE How do bacterial cells grow without breaking their membranes? Although the biochemistry of fatty acid and membrane synthesis is well known, how membrane synthesis is balanced with growth and metabolism has remained unclear. This is partly due to the many control points that have been discovered within the membrane synthesis pathways. By precisely establishing the contributions of individual pathway enzymes, our results simplify the model of membrane biogenesis in the model bacterial species Escherichia coli Specifically, we found that allosteric control of a single enzyme, PlsB, is sufficient to balance growth with membrane synthesis and to ensure that growing E. coli cells produce sufficient membrane. Identifying the signals that activate and deactivate PlsB will resolve the issue of how membrane synthesis is synchronized with growth.


Subject(s)
Acetyltransferases/metabolism , Cell Membrane/metabolism , Escherichia coli/growth & development , Escherichia coli/genetics , Phospholipids/biosynthesis , Acetyltransferases/genetics , Biosynthetic Pathways , Lipopolysaccharides/biosynthesis , Mass Spectrometry , Protein Processing, Post-Translational
5.
Nat Chem Biol ; 14(8): 794-800, 2018 08.
Article in English | MEDLINE | ID: mdl-29942079

ABSTRACT

Carbapenems, a family of ß-lactam antibiotics, are among the most powerful bactericidal compounds in clinical use. However, as rational engineering of native carbapenem-producing microbes is not currently possible, the present carbapenem supply relies upon total chemical synthesis of artificial carbapenem derivatives. To enable access to the full diversity of natural carbapenems, we have engineered production of a simple carbapenem antibiotic within Escherichia coli. By increasing concentrations of precursor metabolites and identifying a reducing cofactor of a bottleneck enzyme, we improved productivity by 60-fold over the minimal pathway and surpassed reported titers obtained from carbapenem-producing Streptomyces species. We stabilized E. coli metabolism against antibacterial effects of the carbapenem product by artificially inhibiting membrane synthesis, which further increased antibiotic productivity. As all known naturally occurring carbapenems are derived from a common intermediate, our engineered strain provides a platform for biosynthesis of tailored carbapenem derivatives in a genetically tractable and fast-growing species.


Subject(s)
Carbapenems/biosynthesis , Escherichia coli/metabolism , Metabolic Engineering , Carbapenems/chemistry
7.
Nature ; 544(7650): 372-376, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28425994

ABSTRACT

The non-essential amino acids serine and glycine are used in multiple anabolic processes that support cancer cell growth and proliferation (reviewed in ref. 1). While some cancer cells upregulate de novo serine synthesis, many others rely on exogenous serine for optimal growth. Restriction of dietary serine and glycine can reduce tumour growth in xenograft and allograft models. Here we show that this observation translates into more clinically relevant autochthonous tumours in genetically engineered mouse models of intestinal cancer (driven by Apc inactivation) or lymphoma (driven by Myc activation). The increased survival following dietary restriction of serine and glycine in these models was further improved by antagonizing the anti-oxidant response. Disruption of mitochondrial oxidative phosphorylation (using biguanides) led to a complex response that could improve or impede the anti-tumour effect of serine and glycine starvation. Notably, Kras-driven mouse models of pancreatic and intestinal cancers were less responsive to depletion of serine and glycine, reflecting an ability of activated Kras to increase the expression of enzymes that are part of the serine synthesis pathway and thus promote de novo serine synthesis.


Subject(s)
Glycine/deficiency , Intestinal Neoplasms/diet therapy , Intestinal Neoplasms/metabolism , Lymphoma/diet therapy , Lymphoma/metabolism , Serine/deficiency , Animals , Antioxidants/metabolism , Biguanides/pharmacology , Cell Line, Tumor , Diet , Disease Models, Animal , Female , Food Deprivation , Glycine/metabolism , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Lymphoma/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nutritional Status , Oxidative Phosphorylation/drug effects , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Serine/biosynthesis , Serine/metabolism , Serine/pharmacology , Survival Rate
8.
Methods Enzymol ; 561: 171-96, 2015.
Article in English | MEDLINE | ID: mdl-26358905

ABSTRACT

Here we discuss our methods to analyze small polar compounds involved in central carbon metabolism using LC-MS. Methods described include sample extraction procedures for cells and medium, as well as for plasma/serum, urine, CSF, and tissue samples. Different extraction solvents are assessed. Our methods for using (13)C stable isotope tracers to examine the kinetics and distributions of mass isotopologues of many metabolites are discussed. Quantification methods are described for (13)C stable isotope tracer experiments as well as for unlabeled experiments. These methods were applied in a fumarate hydratase deficient cell model to show how isotope tracing can demonstrate shifts in metabolic pathways and, together with metabolite exchange rates, can be used to gain insights into changes in cell metabolism.


Subject(s)
Carbon/metabolism , Chromatography, Liquid/methods , Isotope Labeling/methods , Mass Spectrometry/methods , Animals , Carbon Isotopes , Humans
9.
Nat Cell Biol ; 17(10): 1317-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26302408

ABSTRACT

Succinate dehydrogenase (SDH) is a heterotetrameric nuclear-encoded complex responsible for the oxidation of succinate to fumarate in the tricarboxylic acid cycle. Loss-of-function mutations in any of the SDH genes are associated with cancer formation. However, the impact of SDH loss on cell metabolism and the mechanisms enabling growth of SDH-defective cells are largely unknown. Here, we generated Sdhb-ablated kidney mouse cells and used comparative metabolomics and stable-isotope-labelling approaches to identify nutritional requirements and metabolic adaptations to SDH loss. We found that lack of SDH activity commits cells to consume extracellular pyruvate, which sustains Warburg-like bioenergetic features. We further demonstrated that pyruvate carboxylation diverts glucose-derived carbons into aspartate biosynthesis, thus sustaining cell growth. By identifying pyruvate carboxylase as essential for the proliferation and tumorigenic capacity of SDH-deficient cells, this study revealed a metabolic vulnerability for potential future treatment of SDH-associated malignancies.


Subject(s)
Aspartic Acid/biosynthesis , Cell Proliferation , Pyruvic Acid/metabolism , Succinate Dehydrogenase/metabolism , Animals , Carboxylic Acids/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Humans , Immunoblotting , Kidney/cytology , Kidney/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Metabolomics/methods , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pyruvate Carboxylase/metabolism , RNA Interference , Succinate Dehydrogenase/genetics
10.
Cancer Cell ; 27(1): 57-71, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25584894

ABSTRACT

A functional genomics study revealed that the activity of acetyl-CoA synthetase 2 (ACSS2) contributes to cancer cell growth under low-oxygen and lipid-depleted conditions. Comparative metabolomics and lipidomics demonstrated that acetate is used as a nutritional source by cancer cells in an ACSS2-dependent manner, and supplied a significant fraction of the carbon within the fatty acid and phospholipid pools. ACSS2 expression is upregulated under metabolically stressed conditions and ACSS2 silencing reduced the growth of tumor xenografts. ACSS2 exhibits copy-number gain in human breast tumors, and ACSS2 expression correlates with disease progression. These results signify a critical role for acetate consumption in the production of lipid biomass within the harsh tumor microenvironment.


Subject(s)
Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Fatty Acids/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/metabolism , Stress, Physiological
11.
PLoS One ; 9(5): e98438, 2014.
Article in English | MEDLINE | ID: mdl-24875183

ABSTRACT

The Forkhead transcription factor FOXA2 plays a fundamental role in controlling metabolic homeostasis in the liver during fasting. The precise molecular regulation of FOXA2 in response to nutrients is not fully understood. Here, we studied whether FOXA2 could be controlled at a post-translational level by acetylation. By means of LC-MS/MS analyses, we identified five acetylated residues in FOXA2. Sirtuin family member SIRT1 was found to interact with and deacetylate FOXA2, the latter process being dependent on the NAD+-binding catalytic site of SIRT1. Deacetylation by SIRT1 reduced protein stability of FOXA2 by targeting it towards proteasomal degradation, and inhibited transcription from the FOXA2-driven G6pase and CPT1a promoters. While mutation of the five identified acetylated residues weakly affected protein acetylation and stability, mutation of at least seven additional lysine residues was required to abolish acetylation and reduce protein levels of FOXA2. The importance of acetylation of FOXA2 became apparent upon changes in nutrient levels. The interaction of FOXA2 and SIRT1 was strongly reduced upon nutrient withdrawal in cell culture, while enhanced Foxa2 acetylation levels were observed in murine liver in vivo after starvation for 36 hours. Collectively, this study demonstrates that SIRT1 controls the acetylation level of FOXA2 in a nutrient-dependent manner and in times of nutrient shortage the interaction between SIRT1 and FOXA2 is reduced. As a result, FOXA2 is protected from degradation by enhanced acetylation, hence enabling the FOXA2 transcriptional program to be executed to maintain metabolic homeostasis.


Subject(s)
Animal Feed , Hepatocyte Nuclear Factor 3-beta/metabolism , Sirtuin 1/metabolism , Acetylation , Active Transport, Cell Nucleus , Animals , Catalysis , Cell Line , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Liver/metabolism , Mice , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Starvation , Transcription, Genetic
12.
Cell Rep ; 7(4): 1248-58, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813884

ABSTRACT

Previous work has shown that some cancer cells are highly dependent on serine/glycine uptake for proliferation. Although serine and glycine can be interconverted and either might be used for nucleotide synthesis and one-carbon metabolism, we show that exogenous glycine cannot replace serine to support cancer cell proliferation. Cancer cells selectively consumed exogenous serine, which was converted to intracellular glycine and one-carbon units for building nucleotides. Restriction of exogenous glycine or depletion of the glycine cleavage system did not impede proliferation. In the absence of serine, uptake of exogenous glycine was unable to support nucleotide synthesis. Indeed, higher concentrations of glycine inhibited proliferation. Under these conditions, glycine was converted to serine, a reaction that would deplete the one-carbon pool. Providing one-carbon units by adding formate rescued nucleotide synthesis and growth of glycine-fed cells. We conclude that nucleotide synthesis and cancer cell proliferation are supported by serine--rather than glycine--consumption.


Subject(s)
Carbon/metabolism , Glycine/metabolism , Neoplasms/metabolism , Serine/metabolism , Cell Growth Processes/physiology , Glycine/administration & dosage , Glycine/pharmacokinetics , HCT116 Cells , Humans , MCF-7 Cells , Metabolic Networks and Pathways , Neoplasms/pathology , Serine/administration & dosage , Serine/pharmacokinetics
13.
PLoS One ; 8(11): e80935, 2013.
Article in English | MEDLINE | ID: mdl-24244726

ABSTRACT

Cellular systems are essential model systems to study reactive oxygen species and oxidative damage but there are widely accepted technical difficulties with available methods for quantifying endogenous oxidative damage in these systems. Here we present a stable isotope dilution UPLC-MS/MS protocol for measuring F2-isoprostanes as accurate markers for endogenous oxidative damage in cellular systems. F2-isoprostanes are chemically stable prostaglandin-like lipid peroxidation products of arachidonic acid, the predominant polyunsaturated fatty acid in mammalian cells. This approach is rapid and highly sensitive, allowing for the absolute quantification of endogenous lipid peroxidation in as little as ten thousand cells as well as damage originating from multiple ROS sources. Furthermore, differences in the endogenous cellular redox state induced by transcriptional regulation of ROS scavenging enzymes were detected by following this protocol. Finally we showed that the F2-isoprostane 5-iPF2α-VI is a metabolically stable end product, which is excreted from cells. Overall, this protocol enables accurate, specific and sensitive quantification of endogenous lipid peroxidation in cellular systems.


Subject(s)
F2-Isoprostanes/analysis , Arachidonic Acid/analysis , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Chromatography, High Pressure Liquid , F2-Isoprostanes/chemistry , F2-Isoprostanes/metabolism , Hep G2 Cells , Humans , Lipid Peroxidation/physiology , Tandem Mass Spectrometry
14.
Mol Syst Biol ; 9: 679, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23820781

ABSTRACT

Mutations in the daf-2 gene of the conserved Insulin/Insulin-like Growth Factor (IGF-1) pathway double the lifespan of the nematode Caenorhabditis elegans. This phenotype is completely suppressed by deletion of Forkhead transcription factor daf-16. To uncover regulatory mechanisms coordinating this extension of life, we employed a quantitative proteomics strategy with daf-2 mutants in comparison with N2 and daf-16; daf-2 double mutants. This revealed a remarkable longevity-specific decrease in proteins involved in mRNA processing and transport, the translational machinery, and protein metabolism. Correspondingly, the daf-2 mutants display lower amounts of mRNA and 20S proteasome activity, despite maintaining total protein levels equal to that observed in wild types. Polyribosome profiling in the daf-2 and daf-16;daf-2 double mutants confirmed a daf-16-dependent reduction in overall translation, a phenotype reminiscent of Dietary Restriction-mediated longevity, which was independent of germline activity. RNA interference (RNAi)-mediated knockdown of proteins identified by our approach resulted in modified C. elegans lifespan confirming the importance of these processes in Insulin/IGF-1-mediated longevity. Together, the results demonstrate a role for the metabolism of proteins in the Insulin/IGF-1-mediated extension of life.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Insulin-Like Growth Factor I/genetics , Insulin/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Transcription Factors/genetics , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Forkhead Transcription Factors , Gene Expression Regulation , Genotype , Insulin-Like Growth Factor I/metabolism , Longevity/genetics , Mutation , Phenotype , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
15.
Biochem J ; 451(1): 45-53, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23320500

ABSTRACT

Reversible phosphorylation is a widespread molecular mechanism to regulate the function of cellular proteins, including transcription factors. Phosphorylation of the nuclear receptor PPARγ (peroxisome-proliferator-activated receptor γ) at two conserved serine residue (Ser(112) and Ser(273)) results in an altered transcriptional activity of this transcription factor. So far, only a very limited number of cellular enzymatic activities has been described which can dephosphorylate nuclear receptors. In the present study we used immunoprecipitation assays coupled to tandem MS analysis to identify novel PPARγ-regulating proteins. We identified the serine/threonine phosphatase PPM1B [PP (protein phosphatase), Mg(2+)/Mn(2+) dependent, 1B; also known as PP2Cß] as a novel PPARγ-interacting protein. Endogenous PPM1B protein is localized in the nucleus of mature 3T3-L1 adipocytes where it can bind to PPARγ. Furthermore we show that PPM1B can directly dephosphorylate PPARγ, both in intact cells and in vitro. In addition PPM1B increases PPARγ-mediated transcription via dephosphorylation of Ser(112). Finally, we show that knockdown of PPM1B in 3T3-L1 adipocytes blunts the expression of some PPARγ target genes while leaving others unaltered. These findings qualify the phosphatase PPM1B as a novel selective modulator of PPARγ activity.


Subject(s)
Adipocytes/metabolism , Cell Nucleus/metabolism , PPAR gamma/metabolism , Phosphoprotein Phosphatases/metabolism , Transcription, Genetic/physiology , 3T3-L1 Cells , Active Transport, Cell Nucleus/physiology , Adipocytes/cytology , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Humans , Magnesium/metabolism , Manganese/metabolism , Mice , PPAR gamma/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation/physiology , Protein Phosphatase 2C
16.
Cancer Res ; 70(21): 8526-36, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959475

ABSTRACT

Oncogene-induced senescence (OIS) is a potent tumor-suppressive mechanism that is thought to come at the cost of aging. The Forkhead box O (FOXO) transcription factors are regulators of life span and tumor suppression. However, whether and how FOXOs function in OIS have been unclear. Here, we show a role for FOXO4 in mediating senescence by the human BRAF(V600E) oncogene, which arises commonly in melanoma. BRAF(V600E) signaling through mitogen-activated protein kinase/extracellular signal-regulated kinase kinase resulted in increased reactive oxygen species levels and c-Jun NH(2) terminal kinase-mediated activation of FOXO4 via its phosphorylation on Thr(223), Ser(226), Thr(447), and Thr(451). BRAF(V600E)-induced FOXO4 phosphorylation resulted in p21(cip1)-mediated cell senescence independent of p16(ink4a) or p27(kip1). Importantly, melanocyte-specific activation of BRAF(V600E) in vivo resulted in the formation of skin nevi expressing Thr(223)/Ser(226)-phosphorylated FOXO4 and elevated p21(cip1). Together, these findings support a model in which FOXOs mediate a trade-off between cancer and aging.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Melanocytes/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism , Animals , Apoptosis , Blotting, Western , Cell Cycle Proteins , Cell Proliferation , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Forkhead Transcription Factors , Humans , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Melanocytes/pathology , Melanoma/genetics , Melanoma/metabolism , Mice , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Xenograft Model Antitumor Assays
17.
FASEB J ; 24(11): 4271-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20570964

ABSTRACT

In this study, we searched for proteins regulating the tumor suppressor and life-span regulator FOXO4. Through an unbiased tandem-affinity purification strategy combined with mass spectrometry, we identified the heterodimer Ku70/Ku80 (Ku), a DNA double-strand break repair component. Using biochemical interaction studies, we found Ku70 to be necessary and sufficient for the interaction. FOXO4 mediates its tumor-suppressive function in part through transcriptional regulation of the cell cycle arrest p27(kip1) gene. Immunoblotting, luciferase reporter assays, and flow cytometry showed that Ku70 inhibited FOXO4-mediated p27(kip1) transcription and cell cycle arrest induction by >40%. In contrast, Ku70 RNAi but not control RNAi significantly increased p27(kip1) transcription. In addition, in contrast to wild-type mouse embryonic stem (ES) cells, Ku70(-/-) ES cells showed significantly increased FOXO activity, which was rescued by Ku70 reexpression. Immunofluorescence studies demonstrated that Ku70 sequestered FOXO4 in the nucleus. Interestingly, the Ku70-FOXO4 interaction stoichiometry followed a nonlinear dose-response curve by hydrogen peroxide-generated oxidative stress. Low levels of oxidative stress increased interaction stoichiometry up to 75%, peaking at 50 µM, after which dissociation occurred. Because the Ku70 ortholog in the roundworm Caenorhabditis elegans was shown to regulate life span involving C. elegans FOXO, our findings suggest a conserved critical Ku70 role for FOXO function toward coordination of a survival program, regulated by the magnitude of oxidative damage.


Subject(s)
Antigens, Nuclear/metabolism , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Multiprotein Complexes , Stress, Physiological , Transcription Factors/metabolism , Animals , Antigens, Nuclear/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Ku Autoantigen , Mice , Multiprotein Complexes/pharmacology , Oxidative Stress/drug effects , Tandem Mass Spectrometry
18.
PLoS One ; 5(4): e9864, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20386698

ABSTRACT

BACKGROUND: The evolutionarily conserved Ccr4-Not and Bur1/2 kinase complexes are functionally related in Saccharomyces cerevisiae. In this study, we further explore the relationship between the subunits Not4p and Bur2p. METHODOLOGY/PRINCIPAL FINDINGS: First, we investigated the presence of post-translational modifications on the Ccr4-Not complex. Using mass spectrometry analyses we identified several SP/TP phosphorylation sites on its Not4p, Not1p and Caf1p subunits. Secondly, the influence of Not4p phosphorylation on global H3K4 tri-methylation status was examined by immunoblotting. This histone mark is severely diminished in the absence of Not4p or of Bur2p, but did not require the five identified Not4p phosphorylation sites. Thirdly, we found that Not4p phosphorylation is not affected by the kinase-defective bur1-23 mutant. Finally, phenotypic analyses of the Not4p phosphomutant (not4S/T5A) and bur2Delta strains showed overlapping sensitivities to drugs that abolish cellular stress responses. The double-mutant not4S/T5A and bur2Delta strain even revealed enhanced phenotypes, indicating that phosphorylation of Not4p and BUR2 are active in parallel pathways for drug tolerance. CONCLUSIONS: Not4p is a phospho-protein with five identified phosphorylation sites that are likely targets of a cyclin-dependent kinase(s) other than the Bur1/2p complex. Not4p phosphorylation on the five Not4 S/T sites is not required for global H3K4 tri-methylation. In contrast, Not4p phosphorylation is involved in tolerance to cellular stresses and acts in pathways parallel to BUR2 to affect stress responses in Saccharomyces cerevisiae.


Subject(s)
Cyclins/physiology , Saccharomyces cerevisiae Proteins/physiology , Stress, Physiological , Ubiquitin-Protein Ligases/physiology , Cyclins/metabolism , Histones/metabolism , Methylation , Multiprotein Complexes , Mutation , Phosphorylation , Repressor Proteins , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
19.
Cancer Res ; 68(18): 7597-605, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794148

ABSTRACT

The Forkhead box O (FOXO) protein family is an evolutionarily conserved subclass of transcription factors recently identified as bona fide tumor suppressors. Preventing the accumulation of cellular damage due to oxidative stress is thought to underlie its tumor-suppressive role. Oxidative stress, in turn, also feedback controls FOXO4 function. Regulation of this process, however, is poorly understood but may be relevant to the ability of FOXO to control tumor suppression. Here, we characterize novel FOXO4 phosphorylation sites after increased cellular oxidative stress and identify the isomerase Pin1, a protein frequently found to be overexpressed in cancer, as a critical regulator of p27(kip1) through FOXO4 inhibition. We show that Pin1 requires these phosphorylation events to act negatively on FOXO4 transcriptional activity. Consistent with this, oxidative stress induces binding of Pin1 to FOXO, thereby attenuating its monoubiquitination, a yet uncharacterized mode of substrate modulation by Pin1. We have previously shown that monoubiquitination is involved in controlling nuclear translocation in response to cellular stress, and indeed, Pin1 prevents nuclear FOXO4 accumulation. Interestingly, Pin1 acts on FOXO through stimulation of the activity of the deubiquitinating enzyme HAUSP/USP7. Ultimately, this results in decreased transcriptional activity towards target genes, including the cell cycle arrest gene p27(kip1). Notably, in a primary human breast cancer panel, low p27(kip1) levels inversely correlated with Pin1 expression. Thus, Pin1 is identified as a novel negative FOXO regulator, interconnecting FOXO phosphorylation and monoubiquitination in response to cellular stress to regulate p27(kip1).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Peptidylprolyl Isomerase/metabolism , Transcription Factors/antagonists & inhibitors , 3T3 Cells , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Cycle Proteins , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Forkhead Transcription Factors , Humans , Hydrogen Peroxide/pharmacology , Mice , Mutagenesis, Site-Directed , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7 , Ubiquitination/drug effects
20.
PLoS One ; 3(7): e2819, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18665269

ABSTRACT

BACKGROUND: The Forkhead box O (FOXO) class of transcription factors are involved in the regulation of several cellular responses including cell cycle progression and apoptosis. Furthermore, in model organisms FOXOs act as tumor suppressors and affect aging. Previously, we noted that FOXOs and p53 are remarkably similar within their spectrum of regulatory proteins. For example, the de-ubiquitinating enzyme USP7 removes ubiquitin from both FOXO and p53. However, Skp2 has been identified as E3 ligase for FOXO1, whereas Mdm2 is the prime E3 ligase for p53. PRINCIPAL FINDINGS/METHODOLOGY: Here we provide evidence that Mdm2 acts as an E3 ligase for FOXO as well. In vitro incubation of Mdm2 and FOXO results in ATP-dependent (multi)mono-ubiquitination of FOXO similar to p53. Furthermore, in vivo co-expression of Mdm2 and FOXO induces FOXO mono-ubiquitination and consistent with this result, siRNA-mediated depletion of Mdm2 inhibits mono-ubiquitination of FOXO induced by hydrogen peroxide. Regulation of FOXO ubiquitination by Mdm2 is likely to be direct since Mdm2 and FOXO co-immunoprecipitate. In addition, Mdm2-mediated ubiquitination regulates FOXO transcriptional activity. CONCLUSIONS/SIGNIFICANCE: These data identify Mdm2 as a novel E3 ligase for FOXOs and extend the analogous mode of regulation between FOXO and p53.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Cycle Proteins , Cell Line , Cell Line, Tumor , Humans , Mice , Models, Biological , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL
...