Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Fertil Steril ; 107(3): 699-706.e6, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28259259

ABSTRACT

OBJECTIVE: To identify metabolites that are associated with and predict the presence of endometriosis. DESIGN: Metabolomics study using state-of-the-art mass spectrometry approaches. SETTING: University hospital and universities. PATIENT(S): Twenty-five women with laparoscopically confirmed endometriosis (cases) and 19 women with laparoscopically documented absence of endometriosis (controls). None of the women included in this study had received oral contraception or GnRH agonists for a minimum of 1 month before blood collection. INTERVENTION(S): Plasma collection. MAIN OUTCOME MEASURE(S): Metabolite profiles were generated and interrogated using multiple mass spectrometry methods, that is, high performance liquid chromatography coupled with negative mode electrospray ionization tandem mass spectrometry, UPLC-MS/MS, and ultra performance liquid chromatography-electroSpray ionization-quadrupole time-of-flight (UPLC-ESI-Q-TOF). Metabolite groups investigated included phospholipids, glycerophospholipids, ether-phospholipids, cholesterol-esters, triacylglycerol, sphingolipids, free fatty acids, steroids, eicosanoids, and acylcarnitines. RESULT(S): A panel of acylcarnitines predicted the presence of endometriosis with 88.9% specificity and 81.5% sensitivity in human plasma, with a positive predictive value of 75%. However, due to data limitations the outcome of the receiver operating characteristic curve analysis was not significant. CONCLUSION(S): A diagnostic model based on acylcarnitines has the potential to predict the presence and stage of endometriosis.


Subject(s)
Carnitine/analogs & derivatives , Endometriosis/blood , Lipids/blood , Metabolomics , Adult , Area Under Curve , Belgium , Biomarkers/blood , Carnitine/blood , Case-Control Studies , Chromatography, High Pressure Liquid , Endometriosis/diagnosis , Female , Hospitals, University , Humans , Laparoscopy , Metabolomics/methods , Pilot Projects , Predictive Value of Tests , ROC Curve , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
3.
Sci Rep ; 5: 10944, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26091097

ABSTRACT

Regular exercise has emerged as one of the best therapeutic strategies to prevent and treat type-2-diabetes. Exercise-induced changes in the muscle secretome, consisting of myokines and metabolites, may underlie the inter-organ communication between muscle and other organs. To investigate this crosstalk, we developed an in vitro system in which mouse C2C12 myotubes underwent electric pulse stimulation (EPS) to induce contraction. Subsequently the effects of EPS-conditioned media (EPS-CM) on hepatocytes were investigated. Here, we demonstrate that EPS-CM induces Metallothionein 1/2 and Slc30a2 gene expression and reduces Cyp2a3 gene expression in rat hepatocytes. When testing EPS-CM that was generated in the absence of C2C12 myotubes (non-cell EPS-CM) no decrease in Cyp2a3 expression was detected. However, similar inductions in hepatic Mt1/2 and Slc30a2 expression were observed. Non-cell EPS-CM were also applied to C2C12 myotubes and compared to C2C12 myotubes that underwent EPS: here changes in AMPK phosphorylation and myokine secretion largely depended on EPS-induced contraction. Taken together, these findings indicate that EPS can alter C2C12 myotube function and thereby affect gene expression in cells subjected to EPS-CM (Cyp2a3). However, EPS can also generate non-cell-mediated changes in cell culture media, which can affect gene expression in cells subjected to EPS-CM too. While EPS clearly represents a valuable tool in exercise research, care should be taken in experimental design to control for non-cell-mediated effects.


Subject(s)
Gene Expression Regulation , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Animals , Cell Line , Electric Stimulation , Mice , Muscle Fibers, Skeletal/cytology , Rats
4.
Blood ; 125(11): 1782-92, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25568349

ABSTRACT

C/EBPε, a member of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors, is exclusively expressed in myeloid cells and regulates transition from the promyelocytic stage to the myelocytic stage of neutrophil development, being indispensable for secondary and tertiary granule formation. Knowledge concerning the functional role of C/EBPε posttranslational modifications is limited to studies concerning phosphorylation and sumoylation. In the current study, using ectopic expression and ex vivo differentiation of CD34(+) hematopoietic progenitor cells, we demonstrate that C/EBPε is acetylated, which was confirmed by mass spectrometry analysis, identifying 4 acetylated lysines in 3 distinct functional domains. Regulation of C/EBPε acetylation levels by the p300 acetyltransferase and the sirtuin 1 deacetylase controls transcriptional activity, which can at least in part be explained by modulation of DNA binding. During neutrophil development, acetylation of lysines 121 and 198 were found to be crucial for terminal neutrophil differentiation and the expression of neutrophil-specific granule proteins, including lactoferrin and collagenase. Taken together, our data illustrate a critical role for acetylation in the functional regulation of C/EBPε activity during terminal neutrophil development.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Acetylation , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , COS Cells , Cell Differentiation , Cell Line, Tumor , Chlorocebus aethiops , Collagenases/metabolism , HL-60 Cells , Humans , Lactoferrin/metabolism , Lysine/chemistry , Myelopoiesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sirtuin 1/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
5.
J Immunol ; 194(4): 1856-66, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25582855

ABSTRACT

During secondary immune responses, Ab-opsonized bacteria are efficiently taken up via FcRs by dendritic cells. We now demonstrate that this process induces cross-talk between FcRs and TLRs, which results in synergistic release of several inflammatory cytokines, as well as altered lipid metabolite profiles. This altered inflammatory profile redirects Th1 polarization toward Th17 cell responses. Interestingly, GM-CSF-producing Th cells were synergistically evoked as well, which suggests the onset of polyfunctional Th17 cells. Synergistic cytokine release was dependent on activation via MyD88 and ITAM signaling pathways through TLRs and FcRs, respectively. Cytokine regulation occurred via transcription-dependent mechanisms for TNF-α and IL-23 and posttranscriptional mechanisms for caspase-1-dependent release of IL-1ß. Furthermore, cross-talk between TLRs and FcRs was not restricted to dendritic cells. In conclusion, our results support that bacteria alone initiate fundamentally different immune responses compared with Ab-opsonized bacteria through the combined action of two classes of receptors and, ultimately, may refine new therapies for inflammatory diseases.


Subject(s)
Dendritic Cells/immunology , Receptor Cross-Talk/immunology , Receptors, Fc/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptors/immunology , Antibodies, Bacterial/immunology , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Enterobacteriaceae Infections/immunology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/immunology , Flow Cytometry , Humans , Immunologic Memory/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Phenotype , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology
6.
Cell Rep ; 10(3): 339-345, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25600869

ABSTRACT

The biogenesis of ribosomes and their coordination of protein translation consume an enormous amount of cellular energy. As such, it has been established that the inhibition of either process can extend eukaryotic lifespan. Here, we used next-generation sequencing to compare ribosome-associated RNAs from normal strains of Caenorhabditis elegans to those carrying the life-extending daf-2 mutation. We found a long noncoding RNA (lncRNA), transcribed telomeric sequence 1 (tts-1), on ribosomes of the daf-2 mutant. Depleting tts-1 in daf-2 mutants increases ribosome levels and significantly shortens their extended lifespan. We find tts-1 is also required for the longer lifespan of the mitochondrial clk-1 mutants but not the feeding-defective eat-2 mutants. In line with this, the clk-1 mutants express more tts-1 and fewer ribosomes than the eat-2 mutants. Our results suggest that the expression of tts-1 functions in different longevity pathways to reduce ribosome levels in a way that promotes life extension.

7.
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
8.
Obesity (Silver Spring) ; 22(5): 1296-308, 2014 May.
Article in English | MEDLINE | ID: mdl-24339422

ABSTRACT

OBJECTIVE: Extracellular vesicles (EVs) released by human adipocytes or adipose tissue (AT)-explants play a role in the paracrine interaction between adipocytes and macrophages, a key mechanism in AT inflammation, leading to metabolic complications like insulin resistance (IR) were determined. METHODS: EVs released from in vitro differentiated adipocytes and AT-explants ex vivo were characterized by electron microscopy, Western blot, multiplex adipokine-profiling, and quantified by flow cytometry. Primary monocytes were stimulated with EVs from adipocytes, subcutaneous (SCAT) or omental-derived AT (OAT), and phenotyped. Macrophage supernatant was subsequently used to assess the effect on insulin signaling in adipocytes. RESULTS: Adipocyte and AT-derived EVs differentiated monocytes into macrophages characteristic of human adipose tissue macrophages (ATM), defined by release of both pro- and anti-inflammatory cytokines. The adiponectin-positive subset of AT-derived EVs, presumably representing adipocyte-derived EVs, induced a more pronounced ATM-phenotype than the adiponectin-negative AT-EVs. This effect was more evident for OAT-EVs versus SCAT-EVs. Furthermore, supernatant of macrophages pre-stimulated with AT-EVs interfered with insulin signaling in human adipocytes. Finally, the number of OAT-derived EVs correlated positively with patients HOMA-IR. CONCLUSIONS: A possible role for human AT-EVs in a reciprocal pro-inflammatory loop between adipocytes and macrophages, with the potential to aggravate local and systemic IR was demonstrated.


Subject(s)
Adipocytes/ultrastructure , Cell Communication , Macrophages/metabolism , Adipokines/metabolism , Adiponectin/metabolism , Adipose Tissue/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Humans , Immunologic Factors/pharmacology , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance , Monocytes/cytology , Monocytes/metabolism , Obesity/metabolism , Signal Transduction
9.
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
10.
Ageing Res Rev ; 12(4): 918-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24080227

ABSTRACT

Accumulation of oxidative damage has been proposed to be causal to aging as defined by the Free radical Theory of Aging, which has been subject to recent debate. However, a major hurdle in understanding the biological roles of reactive oxygen species (ROS) signaling and their oxidative damage has been the widely recognized methodological difficulties to measure oxidative damage and ROS in vivo. In this review we describe the various novel approaches that have recently been developed to overcome this challenge in the nematode Caenorhabditis elegans, which is a paradigm invertebrate model organism for studying aging and age-related disease given its short lifespan, easy genetics and transparency. In addition, we also discuss these methods in other important model organisms of aging, including the budding yeast Saccharomyces cerevisiae, the fruitfly Drosophila melanogaster and the mouse Mus musculus. After an introduction on the various ROS that can be encountered, we discuss approaches for the detection and quantification of ROS and ROS damage of DNA, lipids and proteins, highlighting examples from literature to demonstrate the applicability and caveats of each method. As will become clear, combinations of approaches have now become possible and will prove essential for thoroughly understanding the involvement of ROS and ROS damage in the biology of aging and disease.


Subject(s)
Aging/metabolism , Caenorhabditis elegans/metabolism , Models, Animal , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Aging/genetics , Animals , Caenorhabditis elegans/genetics , Free Radicals/analysis , Free Radicals/metabolism , Humans , Reactive Oxygen Species/analysis
11.
Nat Commun ; 4: 2656, 2013.
Article in English | MEDLINE | ID: mdl-24141283

ABSTRACT

Transcriptional coregulators, including the acetyltransferase Tip60, have a key role in complex cellular processes such as differentiation. Whereas post-translational modifications have emerged as an important mechanism to regulate transcriptional coregulator activity, the identification of the corresponding demodifying enzymes has remained elusive. Here we show that the expression of the Tip60 protein, which is essential for adipocyte differentiation, is regulated through polyubiquitination on multiple residues. USP7, a dominant deubiquitinating enzyme in 3T3-L1 adipocytes and mouse adipose tissue, deubiquitinates Tip60 both in intact cells and in vitro and increases Tip60 protein levels. Furthermore, inhibition of USP7 expression and activity decreases adipogenesis. Transcriptome analysis reveals several cell cycle genes to be co-regulated by both Tip60 and USP7. Knockdown of either factor results in impaired mitotic clonal expansion, an early step in adipogenesis. These results reveal deubiquitination of a transcriptional coregulator to be a key mechanism in the regulation of early adipogenesis.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue/metabolism , Histone Acetyltransferases/genetics , Protein Processing, Post-Translational , Trans-Activators/genetics , Ubiquitin-Specific Proteases/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Lysine Acetyltransferase 5 , Male , Mice , Mice, Inbred C57BL , Mitosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription, Genetic , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
12.
Immunity ; 39(2): 259-71, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973222

ABSTRACT

Stable Foxp3 expression is required for the development of functional regulatory T (Treg) cells. Here, we demonstrate that the expression of the transcription factor Foxp3 can be regulated through the polyubiquitination of multiple lysine residues, resulting in proteasome-mediated degradation. Expression of the deubiquitinase (DUB) USP7 was found to be upregulated and active in Treg cells, being associated with Foxp3 in the nucleus. Ectopic expression of USP7 decreased Foxp3 polyubiquitination and increased Foxp3 expression. Conversely, either treatment with DUB inhibitor or USP7 knockdown decreased endogenous Foxp3 protein expression and decreased Treg-cell-mediated suppression in vitro. Furthermore, in a murine adoptive-transfer-induced colitis model, either inhibition of DUB activity or USP7 knockdown in Treg cells abrogated their ability to resolve inflammation in vivo. Our data reveal a molecular mechanism in which rapid temporal control of Foxp3 expression in Treg cells can be regulated by USP7, thereby modulating Treg cell numbers and function.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin Thiolesterase/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/genetics , Disease Models, Animal , Endopeptidases/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA Interference , RNA, Small Interfering , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
13.
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
14.
Article in English | MEDLINE | ID: mdl-23502461

ABSTRACT

Upon exposure to platinum analogs, mesenchymal stem cells were recently found to excrete minute amounts of specific lipid mediators that induce chemotherapy resistance. One of these lipids is hexadeca-4,7,10,13-tetraenoic acid (FA(16:4)n-3). Importantly, FA(16:4)n-3 is present in high concentrations in certain fish oils and algae and oral intake of these products also potently induced chemotherapy resistance. These findings suggested that certain foods could negatively affect clinical chemotherapy treatment. In order to allow further study of the relation between FA(16:4)n-3 and clinical chemotherapy resistance, a method for the detection and quantification of FA(16:4)n-3 in plasma is required. Therefore, a quantification method for FA(16:4)n-3 in human and mouse plasma was developed consisting of a liquid-liquid extraction, solid phase clean-up and LC-MS/MS (MRM) analysis. The method was fully validated over a period of three weeks according to the standard protocols and requirements. The linearity range of the method is 1-100 nmol/L (r(2)>0.99) using deuterated FA(16:3)n-3 as an internal standard. The limits of quantification and detection are 1.0 nmol/L and 0.8 nmol/L, respectively. Recoveries for spiked concentrations range from 103 to 108%. The intra-day and inter-day mean precision amounts to 98-106% and 100-108%, respectively. No significant loss of FA(16:4)n-3 is observed upon storage at -80 °C. The developed assay for the detection and quantification of FA(16:4)n-3 in human plasma is robust and reproducible. The validation parameters are within limits of acceptance.


Subject(s)
Chromatography, Liquid/methods , Fatty Acids, Unsaturated/blood , Tandem Mass Spectrometry/methods , Animals , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/isolation & purification , Humans , Limit of Detection , Linear Models , Liquid-Liquid Extraction , Mice , Reproducibility of Results , Solid Phase Extraction
15.
Aging Cell ; 12(2): 214-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23279719

ABSTRACT

Oxidative damage is thought to be a major cause in development of pathologies and aging. However, quantification of oxidative damage is methodologically difficult. Here, we present a robust liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach for accurate, sensitive, and linear in vivo quantification of endogenous oxidative damage in the nematode Caenorhabditis elegans, based on F3-isoprostanes. F3-isoprostanes are prostaglandin-like markers of oxidative damage derived from lipid peroxidation by Reactive Oxygen Species (ROS). Oxidative damage was quantified in whole animals and in multiple cellular compartments, including mitochondria and peroxisomes. Mutants of the mitochondrial electron transport proteins mev-1 and clk-1 showed increased oxidative damage levels. Furthermore, analysis of Superoxide Dismutase (sod) and Catalase (ctl) mutants uncovered that oxidative damage levels cannot be inferred from the phenotype of resistance to pro-oxidants alone and revealed high oxidative damage in a small group of chemosensory neurons. Longitudinal analysis of aging nematodes revealed that oxidative damage increased specifically with postreproductive age. Remarkably, aging of the stress-resistant and long-lived daf-2 insulin/IGF-1 receptor mutant involved distinct daf-16-dependent phases of oxidative damage including a temporal increase at young adulthood. These observations are consistent with a hormetic response to ROS.


Subject(s)
Aging/metabolism , Caenorhabditis elegans/metabolism , Isoprostanes/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , Aging/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Catalase/genetics , Catalase/metabolism , Cytochromes b , Forkhead Transcription Factors , Gene Expression , Insulin/genetics , Insulin/metabolism , Isoprostanes/analysis , Mutation , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sensory Receptor Cells , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
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
17.
Endocrinology ; 153(12): 6104-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23041671

ABSTRACT

Progesterone regulates multiple behavioral, physiological, and pathological aspects of female reproductive biology through its two progesterone receptors (PRs), PR-B and the truncated PR-A. PR-B is necessary for mammary gland development in mice and, compared with PR-A, is overall a stronger transactivator of target genes due to an additional activation function 3 (AF3) domain. In dogs, known for their high sensitivity to progesterone-induced mammary cancer, the PR-B function was studied. Canine PR (cPR)-B appeared to contain multiple mutations within AF3 core sequence motifs and lacks N-terminal ligand-independent posttranslational modifications. Consequently, cPR-B has a weak transactivation potential on progesterone-responsive mouse mammary tumor virus-luc and progesterone response element 2-luc reporters transiently transfected in hamster, human, or canine cells and also on known target genes FKBP5 and SGK in doxycycline-inducible, stable transfected cPR-B in canine mammary cells. The cPR-B function was restored to the level of human PR-B by the replacement of canine AF3 domain with the human one. The lack of AF3 domain-dependent transcriptional activity was unique for canids (gray wolf, red fox, and raccoon dog) and not present in closely related caniform species (brown bear, gray seal, and domestic ferret). Despite the limited transactivation potential, canids develop normal mammary glands and frequently mammary tumors. Therefore, these results question the role of PR-B in breast cancer development and may explain unique features of canid reproduction.


Subject(s)
Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Dogs , Female , Foxes , Humans , Ligands , Mammary Glands, Animal , Mass Spectrometry/methods , Mice , Molecular Sequence Data , Mutation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Raccoon Dogs , Sequence Homology, Amino Acid , Species Specificity , Wolves
18.
Anal Bioanal Chem ; 404(5): 1413-26, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22814969

ABSTRACT

Oxylipins, including eicosanoids, affect a broad range of biological processes, such as the initiation and resolution of inflammation. These compounds, also referred to as lipid mediators, are (non-) enzymatically generated by oxidation of polyunsaturated fatty acids such as arachidonic acid (AA). A plethora of lipid mediators exist which makes the development of generic analytical methods challenging. Here we developed a robust and sensitive targeted analysis platform for oxylipins and applied it in a biological setting, using high performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) operated in dynamic multiple reaction monitoring (dMRM). Besides the well-described AA metabolites, oxylipins derived from linoleic acid, dihomo-γ-linolenic acid, α-linolenic acid, eicosapentaenoic acid and docosahexaenoic acid were included. Our comprehensive platform allows the quantitative evaluation of approximately 100 oxylipins down to low nanomolar levels. Applicability of the analytical platform was demonstrated by analyzing plasma samples of patients undergoing cardiac surgery. Altered levels of some of the oxylipins, especially in certain monohydroxy fatty acids such as 12-HETE and 12-HEPE, were observed in samples collected before and 24 h after cardiac surgery. These findings indicate that this generic oxylipin profiling platform can be applied broadly to study these highly bioactive compounds in relation to human disease.


Subject(s)
Oxylipins/blood , Tandem Mass Spectrometry/methods , Chromatography, High Pressure Liquid/methods , Humans , Male , Middle Aged , Oxylipins/analysis , Sensitivity and Specificity , Thoracic Surgery
19.
Cancer Cell ; 20(3): 370-83, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21907927

ABSTRACT

The development of resistance to chemotherapy is a major obstacle for lasting effective treatment of cancer. Here, we demonstrate that endogenous mesenchymal stem cells (MSCs) become activated during treatment with platinum analogs and secrete factors that protect tumor cells against a range of chemotherapeutics. Through a metabolomics approach, we identified two distinct platinum-induced polyunsaturated fatty acids (PIFAs), 12-oxo-5,8,10-heptadecatrienoic acid (KHT) and hexadeca-4,7,10,13-tetraenoic acid (16:4(n-3)), that in minute quantities induce resistance to a broad spectrum of chemotherapeutic agents. Interestingly, blocking central enzymes involved in the production of these PIFAs (cyclooxygenase-1 and thromboxane synthase) prevents MSC-induced resistance. Our findings show that MSCs are potent mediators of resistance to chemotherapy and reveal targets to enhance chemotherapy efficacy in patients.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclooxygenase 1/metabolism , Drug Resistance, Neoplasm , Fatty Acids, Unsaturated/metabolism , Fatty Acids/metabolism , Mesenchymal Stem Cells/metabolism , Platinum Compounds/pharmacology , Thromboxane-A Synthase/metabolism , Animals , Apoptosis/drug effects , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cyclooxygenase Inhibitors , Humans , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Oxaliplatin , Thromboxane-A Synthase/antagonists & inhibitors , Tumor Cells, Cultured
20.
Mol Cell ; 41(4): 445-57, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21329882

ABSTRACT

Expression of the Myc oncoprotein is downregulated in response to stress signals to allow cells to cease proliferation and escape apoptosis, but the mechanisms involved in this process are poorly understood. Cell cycle arrest in response to DNA damage requires downregulation of Myc via a p53-independent signaling pathway. Here we have used siRNA screening of the human kinome to identify MAPKAPK5 (MK5, PRAK) as a negative regulator of Myc expression. MK5 regulates translation of Myc, since it is required for expression of miR-34b and miR-34c that bind to the 3'UTR of MYC. MK5 activates miR-34b/c expression via phosphorylation of FoxO3a, thereby promoting nuclear localization of FoxO3a and enabling it to induce miR-34b/c expression and arrest proliferation. Expression of MK5 in turn is directly activated by Myc, forming a negative feedback loop. MK5 is downregulated in colon carcinomas, arguing that this feedback loop is disrupted during colorectal tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Down-Regulation , Feedback, Physiological , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HCT116 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...