Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 13(5): e1006829, 2017 May.
Article in English | MEDLINE | ID: mdl-28558063

ABSTRACT

All cells respond to osmotic stress by implementing molecular signaling events to protect the organism. Failure to properly adapt can lead to pathologies such as hypertension and ischemia-reperfusion injury. Mitogen-activated protein kinases (MAPKs) are activated in response to osmotic stress, as well as by signals acting through G protein-coupled receptors (GPCRs). For proper adaptation, the action of these kinases must be coordinated. To identify second messengers of stress adaptation, we conducted a mass spectrometry-based global metabolomics profiling analysis, quantifying nearly 300 metabolites in the yeast S. cerevisiae. We show that three branched-chain amino acid (BCAA) metabolites increase in response to osmotic stress and require the MAPK Hog1. Ectopic addition of these BCAA derivatives promotes phosphorylation of the G protein α subunit and dampens G protein-dependent transcription, similar to that seen in response to osmotic stress. Conversely, genetic ablation of Hog1 activity or the BCAA-regulatory enzymes leads to diminished phosphorylation of Gα and increased transcription. Taken together, our results define a new class of candidate second messengers that mediate cross talk between osmotic stress and GPCR signaling pathways.


Subject(s)
Amino Acids/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Osmotic Pressure , Saccharomyces cerevisiae/metabolism , Signal Transduction , GTP-Binding Protein alpha Subunits/genetics , Gene Expression Regulation, Fungal , Metabolome , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
2.
Am J Respir Crit Care Med ; 190(4): 445-55, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25054455

ABSTRACT

RATIONALE: Sepsis is a leading cause of morbidity and mortality. Currently, early diagnosis and the progression of the disease are difficult to make. The integration of metabolomic and transcriptomic data in a primate model of sepsis may provide a novel molecular signature of clinical sepsis. OBJECTIVES: To develop a biomarker panel to characterize sepsis in primates and ascertain its relevance to early diagnosis and progression of human sepsis. METHODS: Intravenous inoculation of Macaca fascicularis with Escherichia coli produced mild to severe sepsis, lung injury, and death. Plasma samples were obtained before and after 1, 3, and 5 days of E. coli challenge and at the time of killing. At necropsy, blood, lung, kidney, and spleen samples were collected. An integrative analysis of the metabolomic and transcriptomic datasets was performed to identify a panel of sepsis biomarkers. MEASUREMENTS AND MAIN RESULTS: The extent of E. coli invasion, respiratory distress, lethargy, and mortality was dependent on the bacterial dose. Metabolomic and transcriptomic changes characterized severe infections and death, and indicated impaired mitochondrial, peroxisomal, and liver functions. Analysis of the pulmonary transcriptome and plasma metabolome suggested impaired fatty acid catabolism regulated by peroxisome-proliferator activated receptor signaling. A representative four-metabolite model effectively diagnosed sepsis in primates (area under the curve, 0.966) and in two human sepsis cohorts (area under the curve, 0.78 and 0.82). CONCLUSIONS: A model of sepsis based on reciprocal metabolomic and transcriptomic data was developed in primates and validated in two human patient cohorts. It is anticipated that the identified parameters will facilitate early diagnosis and management of sepsis.


Subject(s)
Bacteremia/blood , Bacteremia/diagnosis , Metabolomics/methods , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/diagnosis , Transcriptome/physiology , Animals , Biomarkers/blood , Cohort Studies , Disease Models, Animal , Early Diagnosis , Female , Humans , Macaca , Male
3.
Kidney Int ; 85(5): 1214-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24429397

ABSTRACT

Here we studied plasma metabolomic profiles as determinants of progression to end-stage renal disease (ESRD) in patients with type 2 diabetes (T2D). This nested case-control study evaluated 40 cases who progressed to ESRD during 8-12 years of follow-up and 40 controls who remained alive without ESRD from the Joslin Kidney Study cohort. Controls were matched with cases for baseline clinical characteristics, although controls had slightly higher eGFR and lower levels of urinary albumin excretion than cases. Plasma metabolites at baseline were measured by mass spectrometry-based global metabolomic profiling. Of the named metabolites in the library, 262 were detected in at least 80% of the study patients. The metabolomic platform recognized 78 metabolites previously reported to be elevated in ESRD (uremic solutes). Sixteen were already elevated in the baseline plasma of our cases years before ESRD developed. Other uremic solutes were either not different or not commonly detectable. Essential amino acids and their derivatives were significantly depleted in the cases, whereas certain amino acid-derived acylcarnitines were increased. All findings remained statistically significant after adjustment for differences between study groups in albumin excretion rate, eGFR, or HbA1c. Uremic solute differences were confirmed by quantitative measurements. Thus, abnormal plasma concentrations of putative uremic solutes and essential amino acids either contribute to progression to ESRD or are a manifestation of an early stage(s) of the disease process that leads to ESRD in T2D.


Subject(s)
Amino Acids, Essential/blood , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Kidney Failure, Chronic/etiology , Metabolomics , Uremia/etiology , Aged , Biomarkers/blood , Boston , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetic Nephropathies/blood , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/physiopathology , Disease Progression , Female , Glomerular Filtration Rate , Glycated Hemoglobin/analysis , Humans , Kidney/physiopathology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/physiopathology , Male , Mass Spectrometry , Metabolomics/methods , Middle Aged , Time Factors , Uremia/blood , Uremia/diagnosis , Uremia/physiopathology
4.
J Proteome Res ; 12(4): 1946-55, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23448543

ABSTRACT

Oxidative stress is a determinant of liver steatosis and the progression to more severe forms of disease. The present study investigated the effect of paraoxonase-1 (PON1) deficiency on histological alterations and hepatic metabolism in mice fed a high-fat high-cholesterol diet. We performed nontargeted metabolomics on liver tissues from 8 male PON1-deficient mice and 8 wild-type animals fed a high-fat, high-cholesterol diet for 22 weeks. We also measured 8-oxo-20-deoxyguanosine, reduced and oxidized glutathione, malondialdehyde, 8-isoprostanes and protein carbonyl concentrations. Results indicated lipid droplets in 14.5% of the hepatocytes of wild-type mice and in 83.3% of the PON1-deficient animals (P < 0.001). The metabolomic assay included 322 biochemical compounds, 169 of which were significantly decreased and 16 increased in PON1-deficient mice. There were significant increases in lipid peroxide concentrations and oxidative stress markers. We also found decreased glycolysis and the Krebs cycle. The urea cycle was decreased, and the pyrimidine cycle had a significant increase in orotate. The pathways of triglyceride and phospholipid synthesis were significantly increased. We conclude that PON1 deficiency is associated with oxidative stress and metabolic alterations leading to steatosis in the livers of mice receiving a high-fat high-cholesterol diet.


Subject(s)
Aryldialkylphosphatase/deficiency , Cholesterol/adverse effects , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Lipid Metabolism/drug effects , Amino Acids/metabolism , Animals , Aryldialkylphosphatase/genetics , Biomarkers/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose/metabolism , Glutathione/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Metabolomics/methods , Mice , Mice, Inbred C57BL , Orotic Acid/metabolism , Oxidative Stress
5.
Mol Cell Biol ; 26(1): 77-87, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16354681

ABSTRACT

YAP is a multifunctional adapter protein and transcriptional coactivator with several binding partners well described in vitro and in cell culture. To explore in vivo requirements for YAP, we generated mice carrying a targeted disruption of the Yap gene. Homozygosity for the Yap(tm1Smil) allele (Yap-/-) caused developmental arrest around E8.5. Phenotypic characterization revealed a requirement for YAP in yolk sac vasculogenesis. Yolk sac endothelial and erythrocyte precursors were specified as shown by histology, PECAM1 immunostaining, and alpha globin expression. Nonetheless, development of an organized yolk sac vascular plexus failed in Yap-/- embryos. In striking contrast, vasculogenesis proceeded in both the allantois and the embryo proper. Mutant embryos showed patterned gene expression domains along the anteroposterior neuraxis, midline, and streak/tailbud. Despite this evidence of proper patterning and tissue specification, Yap-/- embryos showed developmental perturbations that included a notably shortened body axis, convoluted anterior neuroepithelium, caudal dysgenesis, and failure of chorioallantoic fusion. These results reveal a vital requirement for YAP in the developmental processes of yolk sac vasculogenesis, chorioallantoic attachment, and embryonic axis elongation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chorioallantoic Membrane/abnormalities , Chorioallantoic Membrane/blood supply , Neovascularization, Physiologic/genetics , Phosphoproteins/genetics , Yolk Sac/abnormalities , Yolk Sac/blood supply , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Embryo, Mammalian/abnormalities , Embryo, Mammalian/blood supply , Embryo, Mammalian/cytology , Embryonic Development/genetics , Gene Expression , Gene Targeting , Genes, Lethal , Homozygote , Mice , Mice, Mutant Strains , Mutation , Phosphoproteins/metabolism , Proteins/genetics , Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins , Yolk Sac/cytology
6.
Mol Cell Biol ; 23(13): 4559-72, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808097

ABSTRACT

Most eukaryotic cells contain nearly equimolar amounts of nucleosomes and H1 linker histones. Despite their abundance and the potential functional specialization of H1 subtypes in multicellular organisms, gene inactivation studies have failed to reveal essential functions for linker histones in vivo. Moreover, in vitro studies suggest that H1 subtypes may not be absolutely required for assembly of chromosomes or nuclei. By sequentially inactivating the genes for three mouse H1 subtypes (H1c, H1d, and H1e), we showed that linker histones are essential for mammalian development. Embryos lacking the three H1 subtypes die by mid-gestation with a broad range of defects. Triple-H1-null embryos have about 50% of the normal ratio of H1 to nucleosomes. Mice null for five of these six H1 alleles are viable but are underrepresented in litters and are much smaller than their littermates. Marked reductions in H1 content were found in certain tissues of these mice and in another compound H1 mutant. These results demonstrate that the total amount of H1 is crucial for proper embryonic development. Extensive reduction of H1 in certain tissues did not lead to changes in nuclear size, but it did result in global shortening of the spacing between nucleosomes.


Subject(s)
Histones/physiology , Nucleosomes/physiology , Alleles , Animals , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatography, High Pressure Liquid , Chromosomes/metabolism , DNA/metabolism , Gene Expression Regulation, Developmental , Genotype , Heterozygote , Histones/chemistry , Histones/metabolism , Homozygote , Mice , Mice, Knockout , Micrococcal Nuclease/metabolism , Models, Biological , Mutation , Nucleosomes/metabolism , Phenotype , Polymerase Chain Reaction , Thymus Gland/metabolism , Time Factors
7.
PLoS One ; 10(5): e0128231, 2015.
Article in English | MEDLINE | ID: mdl-26024523

ABSTRACT

Hematopoietic regeneration after high dose chemotherapy necessitates activation of the stem cell pool. There is evidence that serum taken after chemotherapy comprises factors stimulating proliferation and self-renewal of CD34(+) hematopoietic stem and progenitor cells (HSPCs)--however, the nature of these feedback signals is yet unclear. Here, we addressed the question if specific microRNAs (miRNAs) or metabolites are affected after high dose chemotherapy. Serum taken from the same patients before and after chemotherapy was supplemented for in vitro cultivation of HSPCs. Serum taken after chemotherapy significantly enhanced HSPC proliferation, better maintained a CD34(+) immunophenotype, and stimulated colony forming units. Microarray analysis revealed that 23 miRNAs changed in serum after chemotherapy--particularly, miRNA-320c and miRNA-1275 were down-regulated whereas miRNA-3663-3p was up-regulated. miRNA-320c was exemplarily inhibited by an antagomiR, which seemed to increase proliferation. Metabolomic profiling demonstrated that 44 metabolites were less abundant, whereas three (including 2-hydroxybutyrate and taurocholenate sulphate) increased in serum upon chemotherapy. Nine of these metabolites were subsequently tested for effects on HSPCs in vitro, but none of them exerted a clear concentration dependent effect on proliferation, immunophenotype and colony forming unit formation. Taken together, serum profiles of miRNAs and metabolites changed after chemotherapy. Rather than individually, these factors may act in concert to recruit HSPCs into action for hematopoietic regeneration.


Subject(s)
Antineoplastic Agents/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/drug effects , MicroRNAs/blood , Antigens, CD34/metabolism , Case-Control Studies , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Lymphoma/blood , Lymphoma/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Serum
8.
Cancer Res ; 74(3): 787-96, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24351290

ABSTRACT

The relevance of cysteine metabolism in cancer has gained considerable interest in recent years, largely focusing on its role in generating the antioxidant glutathione. Through metabolomic profiling using a combination of high-throughput liquid and gas chromatography-based mass spectrometry on a total of 69 patient-derived glioma specimens, this report documents the discovery of a parallel pathway involving cysteine catabolism that results in the accumulation of cysteine sulfinic acid (CSA) in glioblastoma. These studies identified CSA to rank as one of the top metabolites differentiating glioblastoma from low-grade glioma. There was strong intratumoral concordance of CSA levels with expression of its biosynthetic enzyme cysteine dioxygenase 1 (CDO1). Studies designed to determine the biologic consequence of this metabolic pathway identified its capacity to inhibit oxidative phosphorylation in glioblastoma cells, which was determined by decreased cellular respiration, decreased ATP production, and increased mitochondrial membrane potential following pathway activation. CSA-induced attenuation of oxidative phosphorylation was attributed to inhibition of the regulatory enzyme pyruvate dehydrogenase. Studies performed in vivo abrogating the CDO1/CSA axis using a lentiviral-mediated short hairpin RNA approach resulted in significant tumor growth inhibition in a glioblastoma mouse model, supporting the potential for this metabolic pathway to serve as a therapeutic target. Collectively, we identified a novel, targetable metabolic pathway involving cysteine catabolism contributing to the growth of aggressive high-grade gliomas. These findings serve as a framework for future investigations designed to more comprehensively determine the clinical application of this metabolic pathway and its contributory role in tumorigenesis.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cysteine/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Metabolic Networks and Pathways , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cysteine/analogs & derivatives , Cysteine/pharmacology , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/genetics , Cysteine Dioxygenase/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Gene Expression , Glioblastoma/genetics , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Grading , Pyruvate Dehydrogenase Complex/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
9.
Clin J Am Soc Nephrol ; 8(3): 363-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23220422

ABSTRACT

BACKGROUND AND OBJECTIVES: CKD is a common public health problem. Identifying biomarkers adds prognostic/diagnostic value by contributing to an understanding of CKD at the molecular level and possibly defining new drug targets. Metabolomics provides a snapshot of biochemical events at a particular time in the progression of CKD. This cross-sectional metabolomics study ascertained whether plasma metabolite profiles are significantly different in CKD stages 2, 3, and 4. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: An analysis of plasma metabolites, using gas and liquid chromatography coupled to mass spectrometry, was conducted on 30 nondiabetic men ages 40-52 years, with 10 participants each in CKD stages 2, 3, and 4 based on their estimated GFR (calculated by the Modified Diet in Renal Disease formula). Participants were recruited in late 2008, and plasma samples were tested at Metabolon Inc and analyzed in 2012. RESULTS: Comparison of stage 3/stage 2 identified 62 metabolites that differed (P ≤ 0.05), with 39 higher and 23 lower in stage 3 compared with stage 2; comparisons of stage 4/stage 2 identified 111 metabolites, with 66 higher and 45 lower; and comparisons of stage 4/stage 3 identified 11 metabolites, with 7 higher and 4 lower. Major differences in metabolite profiles with increasing stage of CKD were observed, including altered arginine metabolism, elevated coagulation/inflammation, impaired carboxylate anion transport, and decreased adrenal steroid hormone production. CONCLUSIONS: Global metabolite profiling of plasma uncovered potential biomarkers of stages of CKD. Moreover, these biomarkers provide insight into possible pathophysiologic processes that may contribute to progression of CKD.


Subject(s)
Metabolomics , Renal Insufficiency, Chronic/blood , Adult , Biomarkers/blood , Chromatography, Liquid , Cross-Sectional Studies , Disease Progression , Gas Chromatography-Mass Spectrometry , Glomerular Filtration Rate , Humans , Male , Metabolomics/methods , Middle Aged , Predictive Value of Tests , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/physiopathology , Severity of Illness Index , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
10.
Toxicol Sci ; 132(1): 235-49, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23152189

ABSTRACT

Chloracne is commonly observed in humans exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD); yet, the mechanism of toxicity is not well understood. Using normal human epidermal keratinocytes, we investigated the mechanism of TCDD-mediated enhancement of epidermal differentiation by integrating functional genomic, metabolomic, and biochemical analyses. TCDD increased the expression of 40% of the genes of the epidermal differentiation complex found on chromosome 1q21 and 75% of the genes required for de novo ceramide biosynthesis. Lipid analysis demonstrated that eight of the nine classes of ceramides were increased by TCDD, altering the ratio of ceramides to free fatty acids. TCDD decreased the expression of the glucose transporter, SLC2A1, and most of the glycolytic transcripts, followed by decreases in glycolytic intermediates, including pyruvate. NADH and Krebs cycle intermediates were decreased, whereas NAD(+) was increased. Mitochondrial glutathione (GSH) reductase activity and the GSH/glutathione disulfide ratio were decreased by TCDD, ultimately leading to mitochondrial dysfunction, characterized by decreased inner mitochondrial membrane potential and ATP production, and increased production of the reactive oxygen species (ROS), hydrogen peroxide. Aryl hydrocarbon receptor (AHR) antagonists blocked the response of many transcripts to TCDD, and the endpoints of decreased ATP production and differentiation, suggesting regulation by the AHR. Cotreatment of cells with chemical antioxidants or the enzyme catalase blocked the TCDD-mediated acceleration of keratinocyte cornified envelope formation, an endpoint of terminal differentiation. Thus, TCDD-mediated ROS production is a critical step in the mechanism of this chemical to accelerate keratinocyte differentiation.


Subject(s)
Cell Differentiation/drug effects , Keratinocytes/drug effects , Polychlorinated Dibenzodioxins/toxicity , Reactive Oxygen Species/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Gene Expression/drug effects , Glycolysis/drug effects , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mitochondria/drug effects , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
11.
PLoS One ; 8(12): e83756, 2013.
Article in English | MEDLINE | ID: mdl-24358308

ABSTRACT

BACKGROUND: Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease and a risk factor for cirrhosis, hepatocellular carcinoma and liver failure. Previously, we reported that dietary docosahexaenoic acid (DHA, 22:6,n-3) was more effective than eicosapentaenoic acid (EPA, 20:5,n-3) at reversing western diet (WD) induced NASH in LDLR(-/-) mice. METHODS: Using livers from our previous study, we carried out a global non-targeted metabolomic approach to quantify diet-induced changes in hepatic metabolism. RESULTS: Livers from WD + olive oil (WD + O)-fed mice displayed histological and gene expression features consistent with NASH. The metabolomic analysis of 320 metabolites established that the WD and n-3 polyunsaturated fatty acid (PUFA) supplementation had broad effects on all major metabolic pathways. Livers from WD + O-fed mice were enriched in saturated (SFA) and monounsaturated fatty acids (MUFA), palmitoyl-sphingomyelin, cholesterol, n-6 PUFA, n-6 PUFA-containing phosphoglycerolipids, n-6 PUFA-derived oxidized lipids (12-HETE) and depleted of C20-22 n-3 PUFA-containing phosphoglycerolipids, C20-22 n-3 PUFA-derived oxidized lipids (18-HEPE, 17,18-DiHETE) and S-lactoylglutathione, a methylglyoxal detoxification product. WD + DHA was more effective than WD + EPA at attenuating WD + O-induced changes in NASH gene expression markers, n-6 PUFA and oxidized lipids, citrate and S-lactosyl glutathione. Diet-induced changes in hepatic MUFA and sphingolipid content were associated with changes in expression of enzymes involved in MUFA and sphingolipid synthesis. Changes in hepatic oxidized fatty acids and S-lactoylglutathione, however, correlated with hepatic n-3 and n-6 C20-22 PUFA content. Hepatic C20-22 n-3 PUFA content was inversely associated with hepatic α-tocopherol and ascorbate content and positively associated with urinary F2- and F3-isoprostanes, revealing diet effects on whole body oxidative stress. CONCLUSION: DHA regulation of hepatic SFA, MUFA, PUFA, sphingomyelin, PUFA-derived oxidized lipids and S-lactoylglutathione may explain the protective effects of DHA against WD-induced NASH in LDLR(-/-) mice.


Subject(s)
Diet , Fatty Acids, Omega-3/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Receptors, LDL/genetics , Animals , Carbon/metabolism , Disease Models, Animal , Endotoxins/blood , Energy Metabolism , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/metabolism , Lipid Peroxidation , Liver/metabolism , Liver/pathology , Male , Metabolome , Metabolomics , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Oxidative Stress , Phospholipids/metabolism , Sphingomyelins/metabolism
12.
Cancer Res ; 72(22): 5878-88, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23026133

ABSTRACT

Although considerable progress has been made toward understanding glioblastoma biology through large-scale genetic and protein expression analyses, little is known about the underlying metabolic alterations promoting their aggressive phenotype. We conducted global metabolomic profiling on patient-derived glioma specimens and identified specific metabolic programs differentiating low- and high-grade tumors, with the metabolic signature of glioblastoma reflecting accelerated anabolic metabolism. When coupled with transcriptional profiles, we identified the metabolic phenotype of the mesenchymal subtype to consist of accumulation of the glycolytic intermediate phosphoenolpyruvate and decreased pyruvate kinase activity. Unbiased hierarchical clustering of metabolomic profiles identified three subclasses, which we term energetic, anabolic, and phospholipid catabolism with prognostic relevance. These studies represent the first global metabolomic profiling of glioma, offering a previously undescribed window into their metabolic heterogeneity, and provide the requisite framework for strategies designed to target metabolism in this rapidly fatal malignancy.


Subject(s)
Glioblastoma/metabolism , Glioma/metabolism , Gas Chromatography-Mass Spectrometry , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Glioma/pathology , Humans , Mesoderm/metabolism , Mesoderm/pathology , Metabolomics , Neoplasm Grading , Phenotype , Phosphoenolpyruvate/metabolism , Pyruvate Kinase/metabolism , Signal Transduction
13.
Autophagy ; 8(10): 1477-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22906967

ABSTRACT

Autophagy is a catabolic process that functions in recycling and degrading cellular proteins, and is also induced as an adaptive response to the increased metabolic demand upon nutrient starvation. However, the prosurvival role of autophagy in response to metabolic stress due to deprivation of glutamine, the most abundant nutrient for mammalian cells, is not well understood. Here, we demonstrated that when extracellular glutamine was withdrawn, autophagy provided cells with sub-mM concentrations of glutamine, which played a critical role in fostering cell metabolism. Moreover, we uncovered a previously unknown connection between metabolic responses to ATG5 deficiency and glutamine deprivation, and revealed that WT and atg5 (-/-) MEFs utilized both common and distinct metabolic pathways over time during glutamine deprivation. Although the early response of WT MEFs to glutamine deficiency was similar in many respects to the baseline metabolism of atg5 (-/-) MEFs, there was a concomitant decrease in the levels of essential amino acids and branched chain amino acid catabolites in WT MEFs after 6 h of glutamine withdrawal that distinguished them from the atg5 (-/-) MEFs. Metabolomic profiling, oxygen consumption and pathway focused quantitative RT-PCR analyses revealed that autophagy and glutamine utilization were reciprocally regulated to couple metabolic and transcriptional reprogramming. These findings provide key insights into the critical prosurvival role of autophagy in maintaining mitochondrial oxidative phosphorylation and cell growth during metabolic stress caused by glutamine deprivation.


Subject(s)
Autophagy , Glutamine/metabolism , Oxygen Consumption , Adenosine Triphosphate/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5 , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Glutamine/deficiency , Glutamine/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Metabolome/drug effects , Metabolome/genetics , Metabolomics , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
PLoS One ; 6(8): e23641, 2011.
Article in English | MEDLINE | ID: mdl-21853158

ABSTRACT

Hepatitis C virus (HCV) is capable of disrupting different facets of lipid metabolism and lipids have been shown to play a crucial role in the viral life cycle. The aim of this study was to examine the effect HCV infection has on the hepatocyte metabolome. Huh-7.5 cells were infected using virus produced by the HCV J6/JFH1 cell culture system and cells were harvested 24, 48, and 72-hours following infection. Metabolic profiling was performed using a non-targeted multiple platform methodology combining ultrahigh performance liquid chromatography/tandem mass spectrometry (UHPLC/MS/MS(2)) and gas chromatography/mass spectrometry (GC/MS). There was a significant increase in a number of metabolites involved in nucleotide synthesis and RNA replication during early HCV infection. NAD levels were also significantly increased along with several amino acids. A number of lipid metabolic pathways were disrupted by HCV infection, resulting in an increase in cholesterol and sphingolipid levels, altered phospholipid metabolism and a possible disruption in mitochondrial fatty acid transport. Fluctuations in 5'-methylthioadenosine levels were also noted, along with alterations in the glutathione synthesis pathway. These results highlight a number of previously unreported metabolic interactions and give a more in depth insight into the effect HCV has on host cell biochemical processes.


Subject(s)
Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatocytes/metabolism , Hepatocytes/virology , Metabolomics , Cell Line, Tumor , Cholesterol/metabolism , Deoxyadenosines/metabolism , Fatty Acids/metabolism , Homeostasis , Humans , Oxidation-Reduction , Phospholipids/metabolism , Sphingolipids/metabolism , Thionucleosides/metabolism , Time Factors
15.
Dev Cell ; 16(3): 398-410, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19289085

ABSTRACT

Outside cells of the preimplantation mouse embryo form the trophectoderm (TE), a process requiring the transcription factor Tead4. Here, we show that transcriptionally active Tead4 can induce Cdx2 and other trophoblast genes in parallel in embryonic stem cells. In embryos, the Tead4 coactivator protein Yap localizes to nuclei of outside cells, and modulation of Tead4 or Yap activity leads to changes in Cdx2 expression. In inside cells, Yap is phosphorylated and cytoplasmic, and this involves the Hippo signaling pathway component Lats. We propose that active Tead4 promotes TE development in outside cells, whereas Tead4 activity is suppressed in inside cells by cell contact- and Lats-mediated inhibition of nuclear Yap localization. Thus, differential signaling between inside and outside cell populations leads to changes in cell fate specification during TE formation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blastocyst Inner Cell Mass/metabolism , DNA-Binding Proteins/metabolism , Muscle Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Trophoblasts/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CDX2 Transcription Factor , Cell Cycle Proteins , Cells, Cultured , DNA-Binding Proteins/genetics , Ectoderm/metabolism , Embryo Culture Techniques , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Muscle Proteins/genetics , Phosphoproteins/genetics , Pregnancy , Protein Serine-Threonine Kinases/genetics , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , YAP-Signaling Proteins
16.
Mol Biol Cell ; 19(7): 3080-96, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18480408

ABSTRACT

Phosphoinositides (PIPs) are ubiquitous regulators of signal transduction events in eukaryotic cells. PIPs are degraded by various enzymes, including PIP phosphatases. The integral membrane Sac1 phosphatases represent a major class of such enzymes. The central role of lipid phosphatases in regulating PIP homeostasis notwithstanding, the biological functions of Sac1-phosphatases remain poorly characterized. Herein, we demonstrate that functional ablation of the single murine Sac1 results in preimplantation lethality in the mouse and that Sac1 insufficiencies result in disorganization of mammalian Golgi membranes and mitotic defects characterized by multiple mechanically active spindles. Complementation experiments demonstrate mutant mammalian Sac1 proteins individually defective in either phosphoinositide phosphatase activity, or in recycling of the enzyme from the Golgi system back to the endoplasmic reticulum, are nonfunctional proteins in vivo. The data indicate Sac1 executes an essential household function in mammals that involves organization of both Golgi membranes and mitotic spindles and that both enzymatic activity and endoplasmic reticulum localization are important Sac1 functional properties.


Subject(s)
Golgi Apparatus/metabolism , Membrane Proteins/physiology , Phosphoric Monoester Hydrolases/chemistry , Spindle Apparatus , Alleles , Animals , Cell Nucleus/metabolism , Embryonic Stem Cells/cytology , Endoplasmic Reticulum/metabolism , Genetic Complementation Test , HeLa Cells , Humans , Membrane Proteins/metabolism , Mice , Mitosis , Mutagenesis, Site-Directed , Phosphoric Monoester Hydrolases/metabolism
17.
Genes Dev ; 20(16): 2202-7, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16882971

ABSTRACT

MicroRNAs (miRNAs) are short, noncoding RNAs that post-transcriptionally regulate gene expression. While hundreds of mammalian miRNA genes have been identified, little is known about the pathways that regulate the production of active miRNA species. Here we show that a large fraction of miRNA genes are regulated post-transcriptionally. During early mouse development, many miRNA primary transcripts, including the Let-7 family, are present at high levels but are not processed by the enzyme Drosha. An analysis of gene expression in primary tumors indicates that the widespread down-regulation of miRNAs observed in cancer is due to a failure at the Drosha processing step. These data uncover a novel regulatory step in miRNA function and provide a mechanism for miRNA down-regulation in cancer.


Subject(s)
Embryo, Mammalian/metabolism , MicroRNAs/metabolism , Neoplasms/genetics , RNA Processing, Post-Transcriptional , Animals , Blotting, Northern , Cell Line , Cell Line, Tumor , Down-Regulation , Embryonic Development , Female , Mice , MicroRNAs/genetics , Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/metabolism , Stem Cells/metabolism , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Up-Regulation
18.
Development ; 129(16): 3851-60, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12135923

ABSTRACT

The segmental heritage of all vertebrates is evident in the character of the vertebral column. And yet, the extent to which direct translation of pattern from the somitic mesoderm and de novo cell and tissue interactions pattern the vertebral column remains a fundamental, unresolved issue. The elements of vertebral column pattern under debate include both segmental pattern and anteroposterior regional specificity. Understanding how vertebral segmentation and anteroposterior positional identity are patterned requires understanding vertebral column cellular and developmental biology. In this study, we characterized alignment of somites and vertebrae, distribution of individual sclerotome progeny along the anteroposterior axis and development of the axial skeleton in zebrafish. Our clonal analysis of zebrafish sclerotome shows that anterior and posterior somite domains are not lineage-restricted compartments with respect to distribution along the anteroposterior axis but support a 'leaky' resegmentation in development from somite to vertebral column. Alignment of somites with vertebrae suggests that the first two somites do not contribute to the vertebral column. Characterization of vertebral column development allowed examination of the relationship between vertebral formula and expression patterns of zebrafish Hox genes. Our results support co-localization of the anterior expression boundaries of zebrafish hoxc6 homologs with a cervical/thoracic transition and also suggest Hox-independent patterning of regionally specific posterior vertebrae.


Subject(s)
Somites/cytology , Spine/embryology , Zebrafish Proteins , Zebrafish/embryology , Animals , Body Patterning , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/genetics , Models, Biological , Spine/cytology , Tail/embryology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL