Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(4): 101113, 2021 10.
Article in English | MEDLINE | ID: mdl-34437902

ABSTRACT

There are five known general catalytic mechanisms used by enzymes to catalyze carbohydrate epimerization. The amino sugar epimerase N-acetylmannosamine-6-phosphate 2-epimerase (NanE) has been proposed to use a deprotonation-reprotonation mechanism, with an essential catalytic lysine required for both steps. However, the structural determinants of this mechanism are not clearly established. We characterized NanE from Staphylococcus aureus using a new coupled assay to monitor NanE catalysis in real time and found that it has kinetic constants comparable with other species. The crystal structure of NanE from Staphylococcus aureus, which comprises a triosephosphate isomerase barrel fold with an unusual dimeric architecture, was solved with both natural and modified substrates. Using these substrate-bound structures, we identified the following active-site residues lining the cleft at the C-terminal end of the ß-strands: Gln11, Arg40, Lys63, Asp124, Glu180, and Arg208, which were individually substituted and assessed in relation to the mechanism. From this, we re-evaluated the central role of Glu180 in this mechanism alongside the catalytic lysine. We observed that the substrate is bound in a conformation that ideally positions the C5 hydroxyl group to be activated by Glu180 and donate a proton to the C2 carbon. Taken together, we propose that NanE uses a novel substrate-assisted proton displacement mechanism to invert the C2 stereocenter of N-acetylmannosamine-6-phosphate. Our data and mechanistic interpretation may be useful in the development of inhibitors of this enzyme or in enzyme engineering to produce biocatalysts capable of changing the stereochemistry of molecules that are not amenable to synthetic methods.


Subject(s)
Bacterial Proteins/chemistry , Carbohydrate Epimerases/chemistry , Hexosamines/chemistry , Staphylococcus aureus/enzymology , Sugar Phosphates/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Carbohydrate Epimerases/genetics , Catalysis , Hexosamines/genetics , Hexosamines/metabolism , Mutation, Missense , Protein Conformation, beta-Strand , Protein Domains , Staphylococcus aureus/genetics , Sugar Phosphates/genetics , Sugar Phosphates/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 321(3): L576-L594, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34318710

ABSTRACT

The paramyxoviridae, respiratory syncytial virus (RSV), and murine respirovirus are enveloped, negative-sense RNA viruses that are the etiological agents of vertebrate lower respiratory tract infections (LRTIs). We observed that RSV infection in human small airway epithelial cells induced accumulation of glycosylated proteins within the endoplasmic reticulum (ER), increased glutamine-fructose-6-phosphate transaminases (GFPT1/2) and accumulation of uridine diphosphate (UDP)-N-acetylglucosamine, indicating activation of the hexosamine biosynthetic pathway (HBP). RSV infection induces rapid formation of spliced X-box binding protein 1 (XBP1s) and processing of activating transcription factor 6 (ATF6). Using pathway selective inhibitors and shRNA silencing, we find that the inositol-requiring enzyme (IRE1α)-XBP1 arm of the unfolded protein response (UPR) is required not only for activation of the HBP, but also for expression of mesenchymal transition (EMT) through the Snail family transcriptional repressor 1 (SNAI1), extracellular matrix (ECM)-remodeling proteins fibronectin (FN1), and matrix metalloproteinase 9 (MMP9). Probing RSV-induced open chromatin domains by ChIP, we find XBP1 binds and recruits RNA polymerase II to the IL6, SNAI1, and MMP9 promoters and the intragenic superenhancer of glutamine-fructose-6-phosphate transaminase 2 (GFPT2). The UPR is sustained through RSV by an autoregulatory loop where XBP1 enhances Pol II binding to its own promoter. Similarly, we investigated the effects of murine respirovirus infection on its natural host (mouse). Murine respirovirus induces mucosal growth factor response, EMT, and the indicators of ECM remodeling in an IRE1α-dependent manner, which persists after viral clearance. These data suggest that IRE1α-XBP1s arm of the UPR pathway is responsible for paramyxovirus-induced metabolic adaptation and mucosal remodeling via EMT and ECM secretion.


Subject(s)
Endoribonucleases/metabolism , Epithelial Cells/metabolism , Hexosamines/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Respiratory Mucosa/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/physiology , Unfolded Protein Response , Virus Replication , X-Box Binding Protein 1/metabolism , Animals , Cell Line, Transformed , Endoribonucleases/genetics , Epithelial Cells/pathology , Epithelial Cells/virology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Hexosamines/genetics , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/pathology , X-Box Binding Protein 1/genetics
3.
Curr Genet ; 67(1): 79-83, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33063175

ABSTRACT

Protein phosphorylation is an essential regulatory mechanism that controls most cellular processes, integrating a variety of environmental signals to drive cellular growth. Isr1 is a negative regulator of the hexosamine biosynthesis pathway (HBP), which produces UDP-GlcNAc, an essential carbohydrate that is the building block of N-glycosylation, GPI anchors and chitin. Isr1 was recently shown to be regulated by phosphorylation by the nutrient-responsive CDK kinase Pho85, allowing it to be targeted for degradation by the SCFCDC4. Here, we show that while deletion of PHO85 stabilizes Isr1 in asynchronous cells, Isr1 is still unstable in mitotically arrested cells in a pho85∆ strain. We provide evidence to suggest that this is through phosphorylation by CDK1. Redundant targeting of Isr1 by two distinct kinases may allow for tight regulation of the HBP in response to different cellular signals.


Subject(s)
CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinases/genetics , F-Box Proteins/genetics , Mitosis/genetics , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Biosynthetic Pathways/genetics , Cell Cycle/genetics , Glucosamine/analogs & derivatives , Glucosamine/genetics , Glycosylation , Hexosamines/genetics , Phosphorylation/genetics , Saccharomyces cerevisiae/genetics , Signal Transduction/genetics
4.
Oncogene ; 39(20): 4103-4117, 2020 05.
Article in English | MEDLINE | ID: mdl-32235891

ABSTRACT

Different evidence has indicated metabolic rewiring as a necessity for pancreatic cancer (PC) growth, invasion, and chemotherapy resistance. A relevant role has been assigned to glucose metabolism. In particular, an enhanced flux through the Hexosamine Biosynthetic Pathway (HBP) has been tightly linked to PC development. Here, we show that enhancement of the HBP, through the upregulation of the enzyme Phosphoacetylglucosamine Mutase 3 (PGM3), is associated with the onset of gemcitabine (GEM) resistance in PC. Indeed, mRNA profiles of GEM sensitive and resistant patient-derived tumor xenografts (PDXs) indicate that PGM3 expression is specifically increased in GEM-resistant PDXs. Of note, PGM3 results also overexpressed in human PC tissues as compared to paired adjacent normal tissues and its higher expression in PC patients is associated with worse median overall survival (OS). Strikingly, genetic or pharmacological PGM3 inhibition reduces PC cell growth, migration, invasion, in vivo tumor growth and enhances GEM sensitivity. Thus, combined treatment between a specific inhibitor of PGM3, named FR054, and GEM results in a potent reduction of xenograft tumor growth without any obvious side effects in normal tissues. Mechanistically, PGM3 inhibition, reducing protein glycosylation, causes a sustained Unfolded Protein Response (UPR), a significant attenuation of the pro-tumorigenic Epidermal Growth Factor Receptor (EGFR)-Akt axis, and finally cell death. In conclusion this study identifies the HBP as a metabolic pathway involved in GEM resistance and provides a strong rationale for a PC therapy addressing the combined treatment with the PGM3 inhibitor and GEM.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Pancreatic Neoplasms , Proto-Oncogene Proteins c-akt , Signal Transduction , Unfolded Protein Response/drug effects , Animals , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hexosamines/genetics , Hexosamines/metabolism , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Unfolded Protein Response/genetics , Xenograft Model Antitumor Assays , Gemcitabine
5.
Am J Med Sci ; 359(2): 79-83, 2020 02.
Article in English | MEDLINE | ID: mdl-32039769

ABSTRACT

BACKGROUND: The hexosamine biosynthesis pathway (HBP) is hypothesized to mediate many of the adverse effects of hyperglycemia. We have shown previously that increased flux through this pathway leads to induction of the growth factor transforming growth factor-α (TGF-α) and to insulin resistance in cultured cells and transgenic mice. TGF-ß is regulated by glucose and is involved in the development of diabetic nephropathy. We therefore hypothesized that the HBP was involved in the regulation of TGF-ß by glucose in rat vascular and kidney cells. METHODS: A plasmid containing the promoter region of TGF-ß1 cloned upstream of the firefly luciferase gene was electroporated into rat aortic smooth muscle, mesangial, and proximal tubule cells. Luciferase activity was measured in cellular extracts from cells cultured in varying concentrations of glucose and glucosamine. RESULTS: Glucose treatment of all cultured cells led to a time- and dose-dependent stimulation in TGF-ß1 transcriptional activity, with high (20 mM) glucose causing a 1.4- to 2.0-fold increase. Glucose stimulation did not occur until after 12 hours and disappeared after 72 hours of treatment. Glucosamine was more potent than glucose, with 3 mM stimulating up to a 4-fold increase in TGFß1-transcriptional activity. The stimulatory effect of glucosamine was also dose-dependent but was slower to develop and longer lasting than that of glucose. CONCLUSIONS: The metabolism of glucose through the HBP mediates extracellular matrix production, possibly via the stimulation of TGF-ß in kidney cells. Hexosamine metabolism therefore, may play a role in the development of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Gene Expression Regulation/drug effects , Glucose/pharmacology , Hexosamines/biosynthesis , Kidney Tubules, Proximal/metabolism , Mesangial Cells/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta1/biosynthesis , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glucose/metabolism , Hexosamines/genetics , Humans , Kidney Tubules, Proximal/pathology , Mesangial Cells/pathology , Mice , Mice, Transgenic , Rats , Time Factors , Transforming Growth Factor beta1/genetics
6.
J Biol Chem ; 295(7): 2018-2033, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31915250

ABSTRACT

The hexosamine biosynthesis pathway (HBP) branches from glycolysis and forms UDP-GlcNAc, the moiety for O-linked ß-GlcNAc (O-GlcNAc) post-translational modifications. An inability to directly measure HBP flux has hindered our understanding of the factors regulating protein O-GlcNAcylation. Our goals in this study were to (i) validate a LC-MS method that assesses HBP flux as UDP-GlcNAc (13C)-molar percent enrichment (MPE) and concentration and (ii) determine whether glucose availability or workload regulate cardiac HBP flux. For (i), we perfused isolated murine working hearts with [U-13C6]glucosamine (1, 10, 50, or 100 µm), which bypasses the rate-limiting HBP enzyme. We observed a concentration-dependent increase in UDP-GlcNAc levels and MPE, with the latter reaching a plateau of 56.3 ± 2.9%. For (ii), we perfused isolated working hearts with [U-13C6]glucose (5.5 or 25 mm). Glycolytic efflux doubled with 25 mm [U-13C6]glucose; however, the calculated HBP flux was similar among the glucose concentrations at ∼2.5 nmol/g of heart protein/min, representing ∼0.003-0.006% of glycolysis. Reducing cardiac workload in beating and nonbeating Langendorff perfusions had no effect on the calculated HBP flux at ∼2.3 and 2.5 nmol/g of heart protein/min, respectively. To the best of our knowledge, this is the first direct measurement of glucose flux through the HBP in any organ. We anticipate that these methods will enable foundational analyses of the regulation of HBP flux and protein O-GlcNAcylation. Our results suggest that in the healthy ex vivo perfused heart, HBP flux does not respond to acute changes in glucose availability or cardiac workload.


Subject(s)
Acetylglucosamine/metabolism , Glucose/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational/genetics , Animals , Biosynthetic Pathways/genetics , Glycolysis/genetics , Glycosylation , Heart/drug effects , Heart/physiopathology , Hexosamines/biosynthesis , Hexosamines/genetics , Humans , Mice , Myocardium/pathology
7.
J Proteome Res ; 18(9): 3447-3460, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31424945

ABSTRACT

Type II epithelial-mesenchymal transition (EMT) plays a vital role in airway injury, repair, and remodeling. Triggered by growth factors, such as transforming growth factor beta (TGFß), EMT induced a biological process that converts epithelial cells into secretory mesenchymal cells with a substantially increased production of extracellular matrix (ECM) proteins. Epithelial cells are not professional secretory cells and produce few ECM proteins under normal conditions. The molecular mechanism underlying the transformation of the protein factory and secretory machinery during EMT is significant because ECM secretion is central to the pathogenesis of airway remodeling. Here we report that type II EMT upregulates the protein N-glycosylation of ECMs. The mechanism study reveals that the substantial increase in synthesis of ECM proteins in EMT activates the inositol-requiring protein 1 (IRE1α)-X-box-binding protein 1 (XBP1) axis of the unfolded protein response (UPR) coupled to the hexosamine biosynthesis pathway (HBP). These two pathways coordinately up-regulate the protein N-glycosylation of ECM proteins and increase ER folding capacity and ER-associated degradation (ERAD), which improve ER protein homeostasis and protect transitioned cells from proteotoxicity. Inhibition of the alternative splicing of XBP1 or protein N-glycosylation blocks ECM protein secretion, indicating the XBP1-HBP plays a prominent role in regulating the secretion of ECM proteins in the mesenchymal transition. Our data suggest that the activation of XBP1-HBP pathways and elevation of protein N-glycosylation is an adaptive response to maintain protein quality control and facilitate the secretion of ECM proteins during the mesenchymal transition. The components of the XBP1-HBP pathways may be therapeutic targets to prevent airway remodeling.


Subject(s)
Airway Remodeling/genetics , Endoribonucleases/genetics , Lung Injury/genetics , Protein Serine-Threonine Kinases/genetics , X-Box Binding Protein 1/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Glycosylation , Hexosamines/genetics , Hexosamines/metabolism , Humans , Lung Injury/metabolism , Lung Injury/pathology , Proteostasis/genetics , Signal Transduction/genetics
8.
Mol Cell ; 75(2): 357-371.e7, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31227231

ABSTRACT

Carbohydrate response element binding protein (ChREBP) is a key transcriptional regulator of de novo lipogenesis (DNL) in response to carbohydrates and in hepatic steatosis. Mechanisms underlying nutrient modulation of ChREBP are under active investigation. Here we identify host cell factor 1 (HCF-1) as a previously unknown ChREBP-interacting protein that is enriched in liver biopsies of nonalcoholic steatohepatitis (NASH) patients. Biochemical and genetic studies show that HCF-1 is O-GlcNAcylated in response to glucose as a prerequisite for its binding to ChREBP and subsequent recruitment of OGT, ChREBP O-GlcNAcylation, and activation. The HCF-1:ChREBP complex resides at lipogenic gene promoters, where HCF-1 regulates H3K4 trimethylation to prime recruitment of the Jumonji C domain-containing histone demethylase PHF2 for epigenetic activation of these promoters. Overall, these findings define HCF-1's interaction with ChREBP as a previously unappreciated mechanism whereby glucose signals are both relayed to ChREBP and transmitted for epigenetic regulation of lipogenic genes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Homeodomain Proteins/genetics , Host Cell Factor C1/genetics , Lipogenesis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Animals , Carbohydrates/genetics , Epigenesis, Genetic , Gene Expression Regulation , Glucose/metabolism , Hexosamines/genetics , Hexosamines/metabolism , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology , Promoter Regions, Genetic/genetics , Protein Interaction Maps/genetics
9.
Mol Cancer Res ; 17(6): 1338-1350, 2019 06.
Article in English | MEDLINE | ID: mdl-30885991

ABSTRACT

The metabolic reprogramming associated with characteristic increases in glucose and glutamine metabolism in advanced cancer is often ascribed to answering a higher demand for metabolic intermediates required for rapid tumor cell growth. Instead, recent discoveries have pointed to an alternative role for glucose and glutamine metabolites as cofactors for chromatin modifiers and other protein posttranslational modification enzymes in cancer cells. Beyond epigenetic mechanisms regulating gene expression, many chromatin modifiers also modulate DNA repair, raising the question whether cancer metabolic reprogramming may mediate resistance to genotoxic therapy and genomic instability. Our prior work had implicated N-acetyl-glucosamine (GlcNAc) formation by the hexosamine biosynthetic pathway (HBP) and resulting protein O-GlcNAcylation as a common means by which increased glucose and glutamine metabolism can drive double-strand break (DSB) repair and resistance to therapy-induced senescence in cancer cells. We have examined the effects of modulating O-GlcNAcylation on the DNA damage response (DDR) in MCF7 human mammary carcinoma in vitro and in xenograft tumors. Proteomic profiling revealed deregulated DDR pathways in cells with altered O-GlcNAcylation. Promoting protein O-GlcNAc modification by targeting O-GlcNAcase or simply treating animals with GlcNAc protected tumor xenografts against radiation. In turn, suppressing protein O-GlcNAcylation by blocking O-GlcNAc transferase activity led to delayed DSB repair, reduced cell proliferation, and increased cell senescence in vivo. Taken together, these findings confirm critical connections between cancer metabolic reprogramming, DDR, and senescence and provide a rationale to evaluate agents targeting O-GlcNAcylation in patients as a means to restore tumor sensitivity to radiotherapy. IMPLICATIONS: The finding that the HBP, via its impact on protein O-GlcNAcylation, is a key determinant of the DDR in cancer provides a mechanistic link between metabolic reprogramming, genomic instability, and therapeutic response and suggests novel therapeutic approaches for tumor radiosensitization.


Subject(s)
Acylation/genetics , Cell Proliferation/genetics , Cellular Senescence/genetics , DNA Repair/genetics , Animals , Biosynthetic Pathways/genetics , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded , Epigenesis, Genetic/genetics , Female , Genomic Instability/genetics , Glucose/genetics , Glutamine/genetics , HEK293 Cells , Hexosamines/genetics , Humans , MCF-7 Cells , Mice , Mice, Nude , N-Acetylglucosaminyltransferases/genetics , Protein Processing, Post-Translational/genetics , Proteomics/methods
10.
Biotechnol J ; 14(4): e1800186, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30221828

ABSTRACT

Sodium butyrate (NaBu) is not only well-known for enhancing protein production, but also degrades glycan quality. In this study, butyrate supplied by the precursor molecule 1,3,4-O-Bu3 ManNAc is applied to overcome the negative effects of NaBu on glycan quality while simultaneously increasing the productivity of the model recombinant erythropoietin (EPO). The beneficial impact of 1,3,4-O-Bu3 ManNAc on EPO glycan quality, while evident in wild-type CHO cells, is particularly pronounced in glycoengineered CHO cells with stable overexpression of ß-1,4- and ß-1,6-N-acetylglucosaminyltransferases (GnTIV and GnTV) and α-2,6-sialyltransferase (ST6) enzymes responsible for N-glycan antennarity and sialylation. Supplementation of 1,3,4-O-Bu3 ManNAc achieves approximately 30% sialylation enhancement on EPO protein in wild-type CHO cells. Overexpression of GnTIV/GnTV/ST6 in CHO cells increases EPO sialylation about 40%. Combining 1,3,4-O-Bu3 ManNAc treatment in glyocengineered CHO cells promotes EPO sialylation about 75% relative to EPO from wild-type CHO cells. Moreover, a detailed mass spectrometric ESI-LC-MS/MS characterization of glycans at each of the three N-glycosylation sites of EPO showed that the 1st N-site is highly sialylated and either the negative impact of NaBu or the beneficial effect 1,3,4-O-Bu3 ManNAc treatments mainly affects the 2nd and 3rd N-glycan sites of EPO protein. In summary, these results demonstrate 1,3,4-O-Bu3 ManNAc can compensate for the negative effect of NaBu on EPO glycan quality while simultaneously enhancing recombinant protein yields. In this way, a platform that integrates glycoengineering with metabolic supplementation can result in synergistic improvements in both production and glycosylation in CHO cells.


Subject(s)
Butyric Acid/chemistry , Erythropoietin/chemistry , Hexosamines/chemistry , Polysaccharides/chemistry , Animals , CHO Cells , Chromatography, Liquid , Cricetinae , Cricetulus , Erythropoietin/genetics , Glycosylation/drug effects , Hexosamines/genetics , Humans , Polysaccharides/biosynthesis , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Tandem Mass Spectrometry
11.
J Clin Invest ; 128(11): 4924-4937, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30130254

ABSTRACT

Mutant KRAS drives glycolytic flux in lung cancer, potentially impacting aberrant protein glycosylation. Recent evidence suggests aberrant KRAS drives flux of glucose into the hexosamine biosynthetic pathway (HBP). HBP is required for various glycosylation processes, such as protein N- or O-glycosylation and glycolipid synthesis. However, its function during tumorigenesis is poorly understood. One contributor and proposed target of KRAS-driven cancers is a developmentally conserved epithelial plasticity program called epithelial-mesenchymal transition (EMT). Here we showed in novel autochthonous mouse models that EMT accelerated KrasG12D lung tumorigenesis by upregulating expression of key enzymes of the HBP pathway. We demonstrated that HBP was required for suppressing KrasG12D-induced senescence, and targeting HBP significantly delayed KrasG12D lung tumorigenesis. To explore the mechanism, we investigated protein glycosylation downstream of HBP and found elevated levels of O-linked ß-N-acetylglucosamine (O-GlcNAcylation) posttranslational modification on intracellular proteins. O-GlcNAcylation suppressed KrasG12D oncogene-induced senescence (OIS) and accelerated lung tumorigenesis. Conversely, loss of O-GlcNAcylation delayed lung tumorigenesis. O-GlcNAcylation of proteins SNAI1 and c-MYC correlated with the EMT-HBP axis and accelerated lung tumorigenesis. Our results demonstrated that O-GlcNAcylation was sufficient and required to accelerate KrasG12D lung tumorigenesis in vivo, which was reinforced by epithelial plasticity programs.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/enzymology , Mutation, Missense , Protein Processing, Post-Translational , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , Acylation , Amino Acid Substitution , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Glucose/genetics , Glucose/metabolism , HEK293 Cells , Hexosamines/genetics , Hexosamines/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/genetics
12.
J Biol Chem ; 291(46): 24105-24120, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27758869

ABSTRACT

Cancer stem cells (CSCs) represent a small subpopulation of self-renewing oncogenic cells. As in many other stem cells, metabolic reprogramming has been implicated to be a key characteristic of CSCs. However, little is known about how the metabolic features of cancer cells are controlled to orchestrate their CSC-like properties. We recently demonstrated that hyaluronan (HA) overproduction allowed plastic cancer cells to revert to stem cell states. Here, we adopted stable isotope-assisted tracing and mass spectrometry profiling to elucidate the metabolic features of HA-overproducing breast cancer cells. These integrated approaches disclosed an acceleration of metabolic flux in the hexosamine biosynthetic pathway (HBP). A metabolic shift toward glycolysis was also evident by quantitative targeted metabolomics, which was validated by the expression profiles of key glycolytic enzymes. Forced expression of glutamine:fructose-6-phosphate amidotransferase 1 (GFAT1), an HBP rate-limiting enzyme, resembled the results of HA overproduction with regard to HIF-1α accumulation and glycolytic program, whereas GFAT1 inhibition significantly decreased HIF-1α protein level in HA-overproducing cancer cells. Moreover, inhibition of the HBP-HIF-1 axis abrogated HA-driven glycolytic enhancement and reduced the CSC-like subpopulation. Taken together, our results provide compelling evidence that HA production regulates the metabolic and CSC-like properties of breast cancer cells via HBP-coupled HIF-1 signaling.


Subject(s)
Hexosamines/biosynthesis , Hyaluronic Acid/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Female , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Hexosamines/genetics , Hyaluronic Acid/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Nitrogenous Group Transferases/genetics , Nitrogenous Group Transferases/metabolism
13.
Biochim Biophys Acta ; 1860(8): 1640-54, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26721333

ABSTRACT

BACKGROUND: Congenital disorders of glycosylation are caused by defects in the glycosylation of proteins and lipids. Classically, gene defects with multisystem disease have been identified in the ubiquitously expressed glycosyltransferases required for protein N-glycosylation. An increasing number of defects are being described in sugar supply pathways for protein glycosylation with tissue-restricted clinical symptoms. SCOPE OF REVIEW: In this review, we address the hexosamine and sialic acid biosynthesis pathways in sugar metabolism. GFPT1, PGM3 and GNE are essential for synthesis of nucleotide sugars uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) and cytidine-5'-monophospho-N-acetylneuraminic acid (CMP-sialic acid) as precursors for various glycosylation pathways. Defects in these enzymes result in contrasting clinical phenotypes of congenital myasthenia, immunodeficiency or adult-onset myopathy, respectively. We therefore discuss the biochemical mechanisms of known genetic defects in the hexosamine and CMP-sialic acid synthesis pathway in relation to the clinical phenotypes. MAJOR CONCLUSIONS: Both UDP-GlcNAc and CMP-sialic acid are important precursors for diverse protein glycosylation reactions and for conversion into other nucleotide-sugars. Defects in the synthesis of these nucleotide sugars might affect a wide range of protein glycosylation reactions. Involvement of multiple glycosylation pathways might contribute to disease phenotype, but the currently available biochemical information on sugar metabolism is insufficient to understand why defects in these pathways present with tissue-specific phenotypes. GENERAL SIGNIFICANCE: Future research on the interplay between sugar metabolism and different glycosylation pathways in a tissue- and cell-specific manner will contribute to elucidation of disease mechanisms and will create new opportunities for therapeutic intervention. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Glycoproteins , Hexosamines , N-Acetylneuraminic Acid , Adult , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Hexosamines/genetics , Hexosamines/metabolism , Humans , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , Phosphoglucomutase/genetics , Phosphoglucomutase/metabolism
14.
J Inherit Metab Dis ; 37(2): 297-308, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24136589

ABSTRACT

BACKGROUND: UDP-GlcNAc 2-epimerase/ManNAc 6-kinase (GNE) is a bifunctional enzyme responsible for the first committed steps in the synthesis of sialic acid, a common terminal monosaccharide in both protein and lipid glycosylation. GNE mutations are responsible for a rare autosomal recessive neuromuscular disorder, GNE myopathy (also called hereditary inclusion body myopathy). The connection between the impairment of sialic acid synthesis and muscle pathology in GNE myopathy remains poorly understood. METHODS: Glycosphingolipid (GSL) analysis was performed by HPLC in multiple models of GNE myopathy, including patients' fibroblasts and plasma, control fibroblasts with inhibited GNE epimerase activity through a novel imino sugar, and tissues of Gne(M712T/M712T) knock-in mice. RESULTS: Not only neutral GSLs, but also sialylated GSLs, were significantly increased compared to controls in all tested models of GNE myopathy. Treatment of GNE myopathy fibroblasts with N-acetylmannosamine (ManNAc), a sialic acid precursor downstream of GNE epimerase activity, ameliorated the increased total GSL concentrations. CONCLUSION: GNE myopathy models have increased total GSL concentrations. ManNAc supplementation results in decrease of GSL levels, linking abnormal increase of total GSLs in GNE myopathy to defects in the sialic acid biosynthetic pathway. These data advocate for further exploring GSL concentrations as an informative biomarker, not only for GNE myopathy, but also for other disorders of sialic acid metabolism.


Subject(s)
Glycosphingolipids/metabolism , Multienzyme Complexes/metabolism , Muscular Diseases/metabolism , Animals , Case-Control Studies , Cells, Cultured , Female , Fibroblasts/metabolism , Glycosphingolipids/blood , Glycosphingolipids/genetics , Hexosamines/blood , Hexosamines/genetics , Hexosamines/metabolism , Humans , Mice , Mice, Inbred C57BL , Multienzyme Complexes/blood , Multienzyme Complexes/genetics , Muscles/metabolism , Muscular Diseases/blood , Muscular Diseases/genetics , Mutation , N-Acetylneuraminic Acid/blood , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism
15.
Cancer Res ; 73(16): 5277-87, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23720054

ABSTRACT

Metabolic disruptions that occur widely in cancers offer an attractive focus for generalized treatment strategies. The hexosamine biosynthetic pathway (HBP) senses metabolic status and produces an essential substrate for O-linked ß-N-acetylglucosamine transferase (OGT), which glycosylates and thereby modulates the function of its target proteins. Here, we report that the HBP is activated in prostate cancer cells and that OGT is a central regulator of c-Myc stability in this setting. HBP genes were overexpressed in human prostate cancers and androgen regulated in cultured human cancer cell lines. Immunohistochemical analysis of human specimens (n = 1987) established that OGT is upregulated at the protein level and that its expression correlates with high Gleason score, pT and pN stages, and biochemical recurrence. RNA interference-mediated siliencing or pharmacologic inhibition of OGT was sufficient to decrease prostate cancer cell growth. Microarray profiling showed that the principal effects of OGT inhibition in prostate cancer cells were related to cell-cycle progression and DNA replication. In particular, c-MYC was identified as a candidate upstream regulator of OGT target genes and OGT inhibition elicited a dose-dependent decrease in the levels of c-MYC protein but not c-MYC mRNA in cell lines. Supporting this relationship, expression of c-MYC and OGT was tightly correlated in human prostate cancer samples (n = 1306). Our findings identify HBP as a modulator of prostate cancer growth and c-MYC as a key target of OGT function in prostate cancer cells.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Androgens/genetics , Androgens/metabolism , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA Replication/drug effects , Hexosamines/biosynthesis , Hexosamines/genetics , Hexosamines/metabolism , Humans , Male , Metabolic Networks and Pathways , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , Prostatic Neoplasms/enzymology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Small Molecule Libraries/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
16.
Glycobiology ; 20(1): 107-17, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19797319

ABSTRACT

Sialic acids are widely expressed as terminal carbohydrates on glycoconjugates of eukaryotic cells. They are involved in a variety of cellular functions, such as cell adhesion or signal recognition. The key enzyme of sialic acid biosynthesis is the bifunctional UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE), which catalyzes the first two steps of sialic acid biosynthesis in the cytosol. Inactivation of GNE causes early embryonic lethality. In this study, we analyzed wild-type and GNE-deficient embryonic stem cells from mice. We found for the first time that proliferation is directly correlated with GNE-expression and the cellular sialic acid concentration. Furthermore, we identified growth-related genes that are differentially expressed in GNE-deficient embryonic stem cells compared to wild-type embryonic stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Hexosamines/genetics , Alleles , Animals , Cell Proliferation , Gene Expression , Genotype , Hexosamines/chemistry , Mice , N-Acetylneuraminic Acid/metabolism , Oligonucleotide Array Sequence Analysis , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
17.
J Appl Microbiol ; 108(5): 1780-8, 2010 May.
Article in English | MEDLINE | ID: mdl-19895652

ABSTRACT

AIMS: Paromamine is a vital and common intermediate in the biosynthesis of 4,5 and 4,6-disubstituted 2-deoxystreptamine (DOS)-containing aminoglycosides. Our aim is to develop an engineered Escherichia coli system for heterologous production of paromamine. METHODS AND RESULTS: We have constructed a mutant of E. coli BL21 (DE3) by disrupting glucose-6-phosphate isomerase (pgi) of primary metabolic pathway to increase glucose-6-phosphate pool inside the host. Disruption was carried out by lambda Red/ET recombination following the protocol mentioned in the kit. Recombinants bearing 2-deoxy-scyllo-inosose (DOI), DOS and paromamine producing genes were constructed from butirosin gene cluster and heterologously expressed in engineered host designed as E. coli BL21 (DE3) Delta pgi. Secondary metabolites produced by the recombinants fermentated in 2YTG medium were extracted, and analysis of the extracts showed there is formation of DOI, DOS and paromamine. CONCLUSIONS: Escherichia coli system is engineered for heterologous expression of paromamine derivatives of aminoglycoside biosynthesis. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first report of heterologous expression of paromamine gene set in E. coli. Hence a new platform is established in E. coli system for the production of paromamine which is useful for the exploration of novel aminoglycosides by combinatorial biosynthesis of 4,5- and 4,6-disubtituted route of DOS-containing aminoglycosides.


Subject(s)
Aminoglycosides/biosynthesis , Aminoglycosides/genetics , Escherichia coli/genetics , Bacillus/genetics , Escherichia coli Proteins/genetics , Gene Expression , Glucose-6-Phosphate Isomerase/genetics , Hexosamines/biosynthesis , Hexosamines/genetics , Inositol/analogs & derivatives , Inositol/biosynthesis , Inositol/genetics , Multigene Family , Protein Engineering
18.
Curr Opin Chem Biol ; 13(5-6): 565-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19747874

ABSTRACT

Metabolic glycoengineering, a technique pioneered almost two decades ago wherein monosaccharide analogs are utilized to install non-natural sugars into the glycocalyx of mammalian cells, has undergone a recent flurry of advances spurred by efforts to make the methodology more efficient. This article describes the versatility of metabolic glycoengineering, which is a prime example of 'chemical glycobiology,' and gives an overview of its capability to endow complex carbohydrates in living cells and animals with interesting (and useful!) functionalities. Then an overview is provided describing how acylated monosaccharides, a class of molecules originally intended to be efficiently-used, membrane-permeable metabolic intermediates, have led to the discovery that a subset of these compounds (e.g. tributanoylated hexosamines) display unanticipated 'scaffold-dependent' activities; this finding establishes these molecules as a versatile platform for drug discovery.


Subject(s)
Bioengineering/methods , Drug Discovery/methods , Hexosamines/chemistry , Hexosamines/metabolism , Animals , Biological Products/chemistry , Biological Products/genetics , Biological Products/metabolism , Biopolymers/chemistry , Biopolymers/genetics , Biopolymers/metabolism , Hexosamines/genetics , Humans
19.
J Antibiot (Tokyo) ; 62(9): 471-81, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19644520

ABSTRACT

Biosynthetic studies of aminoglycoside antibiotics have progressed remarkably during the last decade. Many biosynthetic gene clusters for aminoglycoside antibiotics including streptomycin, kanamycin, butirosin, neomycin and gentamicin have been identified to date. In addition, most butirosin and neomycin biosynthetic enzymes have been functionally characterized using recombinant proteins. Herein, we reanalyze biosynthetic genes for structurally related 2-deoxystreptamine (2DOS)-containing aminoglycosides, such as kanamycin, gentamicin and istamycin, based on genetic information including characterized biosynthetic enzymes in neomycin and butirosin biosynthetic pathways. These proposed enzymatic functions for uncharacterized enzymes are expected to support investigation of the complex biosynthetic pathways for this important class of antibiotics.


Subject(s)
Aminoglycosides/genetics , Anti-Bacterial Agents/biosynthesis , Bacteria/enzymology , Genes, Bacterial , Aminoglycosides/biosynthesis , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Bacteria/genetics , Enzymes/genetics , Enzymes/metabolism , Hexosamines/biosynthesis , Hexosamines/genetics , Lyases/genetics , Multigene Family/physiology , Substrate Specificity
20.
Mol Cells ; 27(5): 601-8, 2009 May 31.
Article in English | MEDLINE | ID: mdl-19466609

ABSTRACT

The 2-deoxystreptamine and paromamine are two key intermediates in kanamycin biosynthesis. In the present study, pSK-2 and pSK-7 recombinant plasmids were constructed with two combinations of genes: kanABK and kanABKF and kacA respectively from kanamycin producer Streptomyces kanamyceticus ATCC12853. These plasmids were heterologously expressed into Streptomyces lividans TK24 independently and generated two recombinant strains named S. lividans Sk-2/SL and S. lividans SK-7/SL, respectively. ESI/ MS and ESI-LC/MS analysis of the metabolite from S. lividans SK-2/SL showed that the compound had a molecular mass of 163 [M + H]+, which corresponds to that of 2-deoxystreptamine. ESI/MS and MS/MS analysis of metabolites from S. lividans SK-7/SL demonstrated the production of paromamine with a molecular mass of 324 [M + H]+. In this study, we report the production of paromamine in a heterologous host for the first time. This study will evoke to explore complete biosynthetic pathways of kanamycin and related aminoglycoside antibiotics.


Subject(s)
Aminoglycosides/metabolism , Anti-Bacterial Agents/biosynthesis , Kanamycin/biosynthesis , Streptomyces lividans/genetics , Streptomyces lividans/metabolism , Aminoglycosides/genetics , Anti-Bacterial Agents/chemistry , Biotechnology , Genetic Engineering , Hexosamines/genetics , Hexosamines/metabolism , In Vitro Techniques , Kanamycin/chemistry , Mass Spectrometry , Molecular Weight , Species Specificity , Streptomyces lividans/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...