Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 40(9): 939-956.e16, 2022 09 12.
Article in English | MEDLINE | ID: mdl-35985343

ABSTRACT

Mutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they seem to be less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1-mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH-mutant gliomas. Mechanistically, this reflects an obligate dependence of glioma cells on the de novo pyrimidine synthesis pathway and mutant IDH's ability to sensitize to DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.


Subject(s)
Brain Neoplasms , Glioma , Leukemia , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Enzyme Inhibitors/therapeutic use , Glioma/drug therapy , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Salicylanilides , Triazoles
2.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34784297

ABSTRACT

The genetic bases for the congenital disorders of glycosylation (CDG) continue to expand, but how glycosylation defects cause patient phenotypes remains largely unknown. Here, we combined developmental phenotyping and biochemical studies in a potentially new zebrafish model (pmm2sa10150) of PMM2-CDG to uncover a protease-mediated pathogenic mechanism relevant to craniofacial and motility phenotypes in mutant embryos. Mutant embryos had reduced phosphomannomutase activity and modest decreases in N-glycan occupancy as detected by matrix-assisted laser desorption ionization mass spectrometry imaging. Cellular analyses of cartilage defects in pmm2sa10150 embryos revealed a block in chondrogenesis that was associated with defective proteolytic processing, but seemingly normal N-glycosylation, of the cell adhesion molecule N-cadherin. The activities of the proconvertases and matrix metalloproteinases responsible for N-cadherin maturation were significantly altered in pmm2sa10150 mutant embryos. Importantly, pharmacologic and genetic manipulation of proconvertase activity restored matrix metalloproteinase activity, N-cadherin processing, and cartilage pathology in pmm2sa10150 embryos. Collectively, these studies demonstrate in CDG that targeted alterations in protease activity create a pathogenic cascade that affects the maturation of cell adhesion proteins critical for tissue development.


Subject(s)
Cadherins/metabolism , Peptide Hydrolases/metabolism , Phosphotransferases (Phosphomutases)/metabolism , Animals , Disease Models, Animal , Humans , Mice
3.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31852778

ABSTRACT

Enteric viruses infect the gastrointestinal tract, and bacteria can promote replication and transmission of several enteric viruses. Viruses can be inactivated by exposure to heat or bleach, but poliovirus, coxsackievirus B3, and reovirus can be stabilized by bacteria or bacterial polysaccharides, limiting inactivation and aiding transmission. We previously demonstrated that certain N-acetylglucosamine (GlcNAc)-containing polysaccharides can stabilize poliovirus. However, the detailed virus-glycan binding specificity and glycan chain length requirements, and thus the mechanism of virion stabilization, have been unclear. A previous limitation was our lack of defined-length glycans to probe mechanisms and consequences of virus-glycan interactions. Here, we generated a panel of polysaccharides and oligosaccharides to determine the properties required for binding and stabilization of poliovirus. Poliovirus virions are nonenveloped icosahedral 30-nm particles with 60 copies of each of four capsid proteins, VP1 to VP4. VP1 surrounds the 5-fold axis, and our past work indicates that this region likely contains the glycan binding site. We found that relatively short GlcNAc oligosaccharides, such as a six-unit GlcNAc oligomer, can bind poliovirus but fail to enhance virion stability. Virion stabilization required binding of long GlcNAc polymers of greater than 20 units. Our data suggest a model where GlcNAc polymers of greater than 20 units bind and bridge adjacent 5-fold axes, thus aiding capsid rigidity and stability. This study provides a deeper understanding of enteric virus-bacterial glycan interactions, which are important for virion environmental stability and transmission.IMPORTANCE Enteric viruses are transmitted through the fecal-oral route, but how enteric viruses survive in the environment is unclear. Previously, we found that bacterial polysaccharides enhance poliovirus stability against heat or bleach inactivation, but the specific molecular requirements have been unknown. Here, we showed that certain short-chain oligosaccharides can bind to poliovirus but do not increase virion stability. Long-chain polysaccharides bind and may bridge adjacent sites on the viral surface, thus increasing capsid rigidity and stability. This work defines the unique interactions of poliovirus and glycans, which provides insight into virion environmental stability and transmission.


Subject(s)
Enterovirus/metabolism , Oligosaccharides/metabolism , Poliovirus/physiology , Polysaccharides , Virion/physiology , Animals , Bacteria/metabolism , Capsid Proteins/metabolism , Chlorocebus aethiops , Enterovirus Infections/virology , HeLa Cells , Humans , Lipopolysaccharides/metabolism , Vero Cells
4.
Nat Commun ; 10(1): 2377, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31147550

ABSTRACT

Glycans from microbial pathogens are well known pathogen-associated molecular patterns that are recognized by the host immunity; however, little is known about whether and how mammalian self-glycans activate the host immune response, especially in the context of autoimmune disease. Using biochemical fractionation and two-dimensional HPLC, we identify an abundant and bioactive free glycan, the Manß1-4GlcNAc disaccharide in TREX1-associated autoimmune diseases. We report that both monosaccharide residues and the ß1-4 linkage are critical for bioactivity of this disaccharide. We also show that Manß1-4GlcNAc is produced by oligosaccharyltransferase hydrolysis of lipid-linked oligosaccharides in the ER lumen, followed by ENGase and mannosidase processing in the cytosol and lysosomes. Furthermore, synthetic Manß1-4GlcNAc disaccharide stimulates a broad immune response in vitro, which is in part dependent on the STING-TBK1 pathway, and enhances antibody response in vivo. Together, our data identify Manß1-4GlcNAc as a novel innate immune modulator associated with chronic autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , Disaccharides/immunology , Immunity, Innate/immunology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Autoimmune Diseases/genetics , Disease Models, Animal , Endoplasmic Reticulum , Exodeoxyribonucleases/genetics , Fibroblasts , Mice , Phosphoproteins/genetics , RAW 264.7 Cells
5.
FASEB J ; 33(6): 6801-6812, 2019 06.
Article in English | MEDLINE | ID: mdl-30811219

ABSTRACT

Herpes simplex virus 1 (HSV-1) is a contagious neurotropic herpesvirus responsible for oral lesions and herpesviral encephalitis. The HSV-1 envelope contains N-glycosylated proteins involved in infection and that are candidate drug targets. NGI-1 is a small-molecule inhibitor of oligosaccharyltransferase (OST) complexes STT3A-OST and STT3B-OST, which catalyze cotranslational and post-translational N-glycosylation, respectively. Because host OSTs attach HSV-1 glycans, NGI-1 might have anti-HSV-1 activity. We evaluated HSV-1 function using NGI-1 and human embryonic kidney 293 knockout lines for OST isoform-specific catalytic and accessory subunits. N-glycosylation of 2 representative envelope proteins (gC and gD) was primarily dependent upon STT3A-OST, but to a large extent replaceable by STT3B-OST. Knockouts impairing STT3A- or STT3B-OST activity, by themselves, did not appreciably affect HSV-1 function (plaque-forming units, normalized to viral particles measured by unglycosylated capsid protein VP5 content). However, with cells lacking STT3B-OST activity (missing the catalytic subunit STT3B or the oxidoreductase subunits magnesium transporter 1/tumor suppressor candidate 3) and thus solely dependent upon STT3A-OST for N-glycosylation, NGI-1 treatment resulted in HSV-1 having cell type-dependent dysfunction (affecting infectivity with Vero cells much more than with the 293 lines). Ablation of post-translational N-glycosylation can therefore make HSV-1 infectivity, and possibly masking of immunogenic peptide epitopes by glycans, highly sensitive to pharmacological inhibition of cotranslational N-glycosylation.-Lu, H., Cherepanova, N. A., Gilmore, R., Contessa, J. N., Lehrman, M. A. Targeting STT3A-oligosaccharyltransferase with NGI-1 causes herpes simplex virus 1 dysfunction.


Subject(s)
Benzamides/pharmacology , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Hexosyltransferases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Sulfonamides/pharmacology , Animals , Chlorocebus aethiops , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Glycosylation , HEK293 Cells , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Vero Cells
6.
Glycobiology ; 29(2): 106-109, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30388226

ABSTRACT

Discovered 40 years ago, the Lec5 glycosylation mutant cell line has a complex recessive genotype and is characterized by accumulation of lipid-linked oligosaccharide assembly intermediates, reduced conversion of polyprenols to dolichols, and an unusual phenotypic dependence upon cell culture conditions such as temperature, plating density and medium quality. The heritable defect in Lec5 is unknown. Here we demonstrate an unexpected epigenetic basis for Lec5, with a surprising linkage to increased expression of homeobox genes, which in turn is associated with increased transcription of cholesterol biosynthesis genes. These results suggest testable hypotheses for the biochemical abnormalities of the Lec5 mutant.


Subject(s)
Cholesterol/genetics , Genes, Homeobox/genetics , Lectins/genetics , Lipopolysaccharides/genetics , Cholesterol/metabolism , Glycosylation , Humans , Lectins/metabolism , Lipopolysaccharides/metabolism , Mutation
7.
Proc Natl Acad Sci U S A ; 115(38): 9557-9562, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30181269

ABSTRACT

Oligosaccharyltransferases (OSTs) N-glycosylate proteins by transferring oligosaccharides from lipid-linked oligosaccharides (LLOs) to asparaginyl residues of Asn-Xaa-Ser/Thr acceptor sequons. Mammals have OST isoforms with STT3A or STT3B catalytic subunits for cotranslational or posttranslational N-glycosylation, respectively. OSTs also hydrolyze LLOs, forming free oligosaccharides (fOSs). It has been unclear whether hydrolysis is due to one or both OSTs, segregated from N-glycosylation, and/or regulated. Transfer and hydrolysis were assayed in permeabilized HEK293 kidney and Huh7.5.1 liver cells lacking STT3A or STT3B. Transfer by both STT3A-OST and STT3B-OST with synthetic acceptors was robust. LLO hydrolysis by STT3B-OST was readily detected and surprisingly modulated: Without acceptors, STT3B-OST hydrolyzed Glc3Man9GlcNAc2-LLO but not Man9GlcNAc2-LLO, yet it hydrolyzed both LLOs with acceptors present. In contrast, LLO hydrolysis by STT3A-OST was negligible. STT3A-OST however may be regulatory, because it suppressed STT3B-OST-dependent fOSs. TREX1, a negative innate immunity factor that diminishes immunogenic fOSs derived from LLOs, acted through STT3B-OST as well. In summary, only STT3B-OST hydrolyzes LLOs, depending upon LLO quality and acceptor site occupancy. TREX1 and STT3A suppress STT3B-OST-dependent fOSs. Without strict kinetic limitations during posttranslational N-glycosylation, STT3B-OST can thus moonlight for LLO hydrolysis. In contrast, the STT3A-OST/translocon complex preserves LLOs for temporally fastidious cotranslational N-glycosylation.


Subject(s)
Hexosyltransferases/metabolism , Lipopolysaccharides/metabolism , Membrane Proteins/metabolism , Oligosaccharides/metabolism , Protein Processing, Post-Translational/physiology , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Gene Knockout Techniques , Glycosylation , Hexosyltransferases/genetics , Humans , Hydrolysis , Isoenzymes , Membrane Proteins/genetics , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport/physiology
8.
Nat Chem Biol ; 12(12): 1023-1030, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27694802

ABSTRACT

Asparagine (N)-linked glycosylation is a protein modification critical for glycoprotein folding, stability, and cellular localization. To identify small molecules that inhibit new targets in this biosynthetic pathway, we initiated a cell-based high-throughput screen and lead-compound-optimization campaign that delivered a cell-permeable inhibitor, NGI-1. NGI-1 targets oligosaccharyltransferase (OST), a hetero-oligomeric enzyme that exists in multiple isoforms and transfers oligosaccharides to recipient proteins. In non-small-cell lung cancer cells, NGI-1 blocks cell-surface localization and signaling of the epidermal growth factor receptor (EGFR) glycoprotein, but selectively arrests proliferation in only those cell lines that are dependent on EGFR (or fibroblast growth factor, FGFR) for survival. In these cell lines, OST inhibition causes cell-cycle arrest accompanied by induction of p21, autofluorescence, and cell morphology changes, all hallmarks of senescence. These results identify OST inhibition as a potential therapeutic approach for treating receptor-tyrosine-kinase-dependent tumors and provides a chemical probe for reversibly regulating N-linked glycosylation in mammalian cells.


Subject(s)
Benzamides/pharmacology , Cellular Senescence/drug effects , Enzyme Inhibitors/pharmacology , Hexosyltransferases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfonamides/pharmacology , Benzamides/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Hexosyltransferases/metabolism , High-Throughput Screening Assays , Humans , Membrane Proteins/metabolism , Molecular Structure , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry
9.
Mol Biol Cell ; 27(8): 1220-34, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26912795

ABSTRACT

Activation of the unfolded protein response (UPR) can be either adaptive or pathological. We term the pathological UPR that causes fatty liver disease a "stressed UPR." Here we investigate the mechanism of stressed UPR activation in zebrafish bearing a mutation in thetrappc11gene, which encodes a component of the transport protein particle (TRAPP) complex.trappc11mutants are characterized by secretory pathway defects, reflecting disruption of the TRAPP complex. In addition, we uncover a defect in protein glycosylation intrappc11mutants that is associated with reduced levels of lipid-linked oligosaccharides (LLOs) and compensatory up-regulation of genes in the terpenoid biosynthetic pathway that produces the LLO anchor dolichol. Treating wild-type larvae with terpenoid or LLO synthesis inhibitors phenocopies the stressed UPR seen intrappc11mutants and is synthetically lethal withtrappc11mutation. We propose that reduced LLO level causing hypoglycosylation is a mechanism of stressed UPR induction intrappc11mutants. Of importance, in human cells, depletion of TRAPPC11, but not other TRAPP components, causes protein hypoglycosylation, and lipid droplets accumulate in fibroblasts from patients with theTRAPPC11mutation. These data point to a previously unanticipated and conserved role for TRAPPC11 in LLO biosynthesis and protein glycosylation in addition to its established function in vesicle trafficking.


Subject(s)
Oligosaccharides/metabolism , Unfolded Protein Response , Vesicular Transport Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Atorvastatin/pharmacology , Dolichols/biosynthesis , Dolichols/genetics , Glycosylation , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Larva/drug effects , Larva/metabolism , Lipids/chemistry , Liver/metabolism , Liver/pathology , Mutation , Oligosaccharides/chemistry , Terpenes/metabolism , Terpenes/pharmacology , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , Vesicular Transport Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
10.
Trends Biochem Sci ; 40(12): 715-717, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26476576

ABSTRACT

The mechanism for flipping large lipid-linked oligosaccharides across membranes has remained a paradox. Perez et al. now report the structure of the PglK protein of C. jejuni, a flippase for a bacterial lipid-linked oligosaccharide, and reveal an unexpected whip-like mechanism.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Biocatalysis , Campylobacter jejuni/enzymology , Lipopolysaccharides/metabolism
11.
Leuk Res ; 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26346348

ABSTRACT

BCR-ABL positive (+) acute lymphoblastic leukemia (ALL) accounts for ∼30% of cases of ALL. We recently demonstrated that 2-deoxy-d-glucose (2-DG), a dual energy (glycolysis inhibition) and ER-stress (N-linked-glycosylation inhibition) inducer, leads to cell death in ALL via ER-stress/UPR-mediated apoptosis. Among ALL subtypes, BCR-ABL+ ALL cells exhibited the highest sensitivity to 2-DG suggesting BCR-ABL expression may be linked to this increased vulnerability. To confirm the role of BCR-ABL, we constructed a NALM6/BCR-ABL stable cell line and found significant increase in 2-DG-induced apoptosis compared to control. We found that Mcl-1 was downregulated by agents inducing ER-stress and Mcl-1 levels correlated with ALL sensitivity. In addition, we showed that Mcl-1 expression is positively regulated by the MEK/ERK pathway, dependent on BCR-ABL, and further downregulated by combining ER-stressors with TKIs. We determined that energy/ER stressors led to translational repression of Mcl-1 via the AMPK/mTOR and UPR/PERK/eIF2α pathways. Taken together, our data indicate that BCR-ABL+ ALL exhibits heightened sensitivity to induction of energy and ER-stress through inhibition of the MEK/ERK pathway, and translational repression of Mcl-1 expression via AMPK/mTOR and UPR/PERK/eIF2α pathways. This study supports further consideration of strategies combining energy/ER-stress inducers with BCR-ABL TKIs for future clinical translation in BCR-ABL+ ALL patients.

12.
Immunity ; 43(3): 463-74, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26320659

ABSTRACT

TREX1 is an endoplasmic reticulum (ER)-associated negative regulator of innate immunity. TREX1 mutations are associated with autoimmune and autoinflammatory diseases. Biallelic mutations abrogating DNase activity cause autoimmunity by allowing immunogenic self-DNA to accumulate, but it is unknown how dominant frameshift (fs) mutations that encode DNase-active but mislocalized proteins cause disease. We found that the TREX1 C terminus suppressed immune activation by interacting with the ER oligosaccharyltransferase (OST) complex and stabilizing its catalytic integrity. C-terminal truncation of TREX1 by fs mutations dysregulated the OST complex, leading to free glycan release from dolichol carriers, as well as immune activation and autoantibody production. A connection between OST dysregulation and immune disorders was demonstrated in Trex1(-/-) mice, TREX1-V235fs patient lymphoblasts, and TREX1-V235fs knock-in mice. Inhibiting OST with aclacinomycin corrects the glycan and immune defects associated with Trex1 deficiency or fs mutation. This function of the TREX1 C terminus suggests a potential therapeutic option for TREX1-fs mutant-associated diseases.


Subject(s)
Cytosol/enzymology , Exodeoxyribonucleases/metabolism , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Aclarubicin/analogs & derivatives , Aclarubicin/pharmacology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Exodeoxyribonucleases/antagonists & inhibitors , Exodeoxyribonucleases/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Frameshift Mutation , HEK293 Cells , HeLa Cells , Hexosyltransferases/genetics , Humans , Immunity, Innate/genetics , Immunoblotting , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Polysaccharides/metabolism , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
13.
Cell ; 156(6): 1179-1192, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24630721

ABSTRACT

The hexosamine biosynthetic pathway (HBP) generates uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) for glycan synthesis and O-linked GlcNAc (O-GlcNAc) protein modifications. Despite the established role of the HBP in metabolism and multiple diseases, regulation of the HBP remains largely undefined. Here, we show that spliced X-box binding protein 1 (Xbp1s), the most conserved signal transducer of the unfolded protein response (UPR), is a direct transcriptional activator of the HBP. We demonstrate that the UPR triggers HBP activation via Xbp1s-dependent transcription of genes coding for key, rate-limiting enzymes. We further establish that this previously unrecognized UPR-HBP axis is triggered in a variety of stress conditions. Finally, we demonstrate a physiologic role for the UPR-HBP axis by showing that acute stimulation of Xbp1s in heart by ischemia/reperfusion confers robust cardioprotection in part through induction of the HBP. Collectively, these studies reveal that Xbp1s couples the UPR to the HBP to protect cells under stress.


Subject(s)
Biosynthetic Pathways , DNA-Binding Proteins/metabolism , Hexosamines/metabolism , Transcription Factors/metabolism , Unfolded Protein Response , Animals , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Humans , Male , Mice , Mice, Transgenic , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Nitrogenous Group Transferases/genetics , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
14.
Cancer Chemother Pharmacol ; 73(2): 349-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24292700

ABSTRACT

BACKGROUND: As tumors evolve, they upregulate glucose metabolism while also encountering intermittent periods of glucose deprivation. Here, we investigate mechanisms by which pancreatic cancer cells respond to therapeutic (2-deoxy-D-glucose, 2-DG) and physiologic (glucose starvation, GS) forms of glucose restriction. METHODS: From a tumor cell line (1420) that is unusually sensitive to 2-DG under normoxia, low (14DG2)- and high (14DG5)-dose resistant cell lines were selected and used to probe the metabolic pathways involved with their response to different forms of glucose deprivation. RESULTS: Muted induction of the unfolded protein response was found to correlate with resistance to 2-DG. Additionally, 14DG2 displayed reduced 2-DG uptake, while 14DG5 was cross-resistant to tunicamycin, suggesting it has enhanced ability to manage glycosylation defects. Conversely, 2-DG-resistant cell lines were more sensitive than their parental cell line to GS, which coincided with lowered levels of glycogen phosphorylase (PYGB) and reduced breakdown of glycogen to glucose in the 2-DG-resistant cell lines. Moreover, by inhibiting PYGB in the parental cell line, sensitivity to GS was increased. CONCLUSIONS: Overall, the data demonstrate that the manner in which glucose is restricted in tumor cells, i.e., therapeutic or physiologic, leads to differential biological responses involving distinct glucose metabolic pathways. Moreover, in evolving tumors where glucose restriction occurs, the identification of PYGB as a metabolic target may have clinical application.


Subject(s)
Deoxyglucose/pharmacology , Glucose/deficiency , Glucose/metabolism , Glycogen Phosphorylase/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Down-Regulation , Glycolysis , Humans , Isoenzymes , Pancreatic Neoplasms/enzymology , Unfolded Protein Response
16.
Methods Mol Biol ; 1022: 277-82, 2013.
Article in English | MEDLINE | ID: mdl-23765668

ABSTRACT

Oligosaccharyltransferase (OT) catalyzes the signature reaction of the asparagine-linked glycosylation pathway, namely, the transfer of preformed glycans from the lipid-linked oligosaccharide Glc3Man9GlcNAc2-P-P-Dolichol (G3M9Gn2-LLO) to appropriate asparaginyl residues on acceptor polypeptides. We have identified a reaction, possibly catalyzed by OT, that results in the hydrolysis or "transfer to water" of host LLOs in response to viral infection with release of a free G3M9Gn2 glycan. The loss of LLO ostensibly hinders N-glycosylation of viral polypeptides. This response is achieved by a novel stress-activated signaling pathway in which free mannose-6-phosphate (M6P) acts as a second-messenger. Here, we describe methods with permeabilized mammalian cells for activation of the M6P-regulated LLO hydrolysis, or transfer of glycan to water, in vitro.


Subject(s)
Lipopolysaccharides/metabolism , Mannosephosphates/metabolism , Animals , Bacterial Proteins/metabolism , Cell Culture Techniques/methods , Cell Membrane Permeability , Glycosylation , Hydrolysis , Polyisoprenyl Phosphate Sugars/metabolism , Polysaccharides/metabolism , Streptolysins/metabolism , Water/metabolism
17.
J Biol Chem ; 288(28): 20616-23, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720757

ABSTRACT

Mature dolichol-linked oligosaccharides (mDLOs) needed for eukaryotic protein N-glycosylation are synthesized by a multistep pathway in which the biosynthetic lipid intermediate Man5GlcNAc2-PP-dolichol (M5-DLO) flips from the cytoplasmic to the luminal face of the endoplasmic reticulum. The endoplasmic reticulum membrane protein Rft1 is intimately involved in mDLO biosynthesis. Yeast genetic analyses implicated Rft1 as the M5-DLO flippase, but because biochemical tests challenged this assignment, the function of Rft1 remains obscure. To understand the role of Rft1, we sought to analyze mDLO biosynthesis in vivo in the complete absence of the protein. Rft1 is essential for yeast viability, and no Rft1-null organisms are currently available. Here, we exploited Trypanosoma brucei (Tb), an early diverging eukaryote whose Rft1 homologue functions in yeast. We report that TbRft1-null procyclic trypanosomes grow nearly normally. They have normal steady-state levels of mDLO and significant N-glycosylation, indicating robust M5-DLO flippase activity. Remarkably, the mutant cells have 30-100-fold greater steady-state levels of M5-DLO than wild-type cells. All N-glycans in the TbRft1-null cells originate from mDLO indicating that the M5-DLO excess is not available for glycosylation. These results suggest that rather than facilitating M5-DLO flipping, Rft1 facilitates conversion of M5-DLO to mDLO by another mechanism, possibly by acting as an M5-DLO chaperone.


Subject(s)
Eukaryotic Cells/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Flow Cytometry , Glucose/pharmacology , Glycoproteins/genetics , Glycosylation , Lysosomal Membrane Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Mutation , Polyisoprenyl Phosphate Oligosaccharides/metabolism , Polysaccharides/metabolism , Protein Biosynthesis , Protozoan Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transformation, Genetic , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/growth & development
18.
Dis Model Mech ; 6(1): 95-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22899857

ABSTRACT

Individuals with congenital disorders of glycosylation (CDG) have recessive mutations in genes required for protein N-glycosylation, resulting in multi-systemic disease. Despite the well-characterized biochemical consequences in these individuals, the underlying cellular defects that contribute to CDG are not well understood. Synthesis of the lipid-linked oligosaccharide (LLO), which serves as the sugar donor for the N-glycosylation of secretory proteins, requires conversion of fructose-6-phosphate to mannose-6-phosphate via the phosphomannose isomerase (MPI) enzyme. Individuals who are deficient in MPI present with bleeding, diarrhea, edema, gastrointestinal bleeding and liver fibrosis. MPI-CDG patients can be treated with oral mannose supplements, which is converted to mannose-6-phosphate through a minor complementary metabolic pathway, restoring protein glycosylation and ameliorating most symptoms, although liver disease continues to progress. Because Mpi deletion in mice causes early embryonic lethality and thus is difficult to study, we used zebrafish to establish a model of MPI-CDG. We used a morpholino to block mpi mRNA translation and established a concentration that consistently yielded 13% residual Mpi enzyme activity at 4 days post-fertilization (dpf), which is within the range of MPI activity detected in fibroblasts from MPI-CDG patients. Fluorophore-assisted carbohydrate electrophoresis detected decreased LLO and N-glycans in mpi morphants. These deficiencies resulted in 50% embryonic lethality by 4 dpf. Multi-systemic abnormalities, including small eyes, dysmorphic jaws, pericardial edema, a small liver and curled tails, occurred in 82% of the surviving larvae. Importantly, these phenotypes could be rescued with mannose supplementation. Thus, parallel processes in fish and humans contribute to the phenotypes caused by Mpi depletion. Interestingly, mannose was only effective if provided prior to 24 hpf. These data provide insight into treatment efficacy and the broader molecular and developmental abnormalities that contribute to disorders associated with defective protein glycosylation.


Subject(s)
Congenital Disorders of Glycosylation/diet therapy , Congenital Disorders of Glycosylation/enzymology , Mannose-6-Phosphate Isomerase/deficiency , Mannose-6-Phosphate Isomerase/genetics , Mannose/administration & dosage , Animals , Base Sequence , Congenital Disorders of Glycosylation/genetics , Dietary Supplements , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Mice , Morpholinos/administration & dosage , Morpholinos/genetics , Mutation , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
19.
J Clin Invest ; 123(1): 455-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257357

ABSTRACT

Postprandially, the liver experiences an extensive metabolic reprogramming that is required for the switch from glucose production to glucose assimilation. Upon refeeding, the unfolded protein response (UPR) is rapidly, though only transiently, activated. Activation of the UPR results in a cessation of protein translation, increased chaperone expression, and increased ER-mediated protein degradation, but it is not clear how the UPR is involved in the postprandial switch to alternate fuel sources. Activation of the inositol-requiring enzyme 1 (IRE1) branch of the UPR signaling pathway triggers expression of the transcription factor Xbp1s. Using a mouse model with liver-specific inducible Xbp1s expression, we demonstrate that Xbp1s is sufficient to provoke a metabolic switch characteristic of the postprandial state, even in the absence of caloric influx. Mechanistically, we identified UDP-galactose-4-epimerase (GalE) as a direct transcriptional target of Xbp1s and as the key mediator of this effect. Our results provide evidence that the Xbp1s/GalE pathway functions as a novel regulatory nexus connecting the UPR to the characteristic postprandial metabolic changes in hepatocytes.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Hepatocytes/metabolism , Liver/metabolism , Postprandial Period/physiology , Transcription Factors/metabolism , UDPglucose 4-Epimerase/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Hepatocytes/cytology , Liver/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rabbits , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , UDPglucose 4-Epimerase/genetics , X-Box Binding Protein 1
20.
Mol Biol Cell ; 23(21): 4175-87, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956764

ABSTRACT

Congenital disorder of glycosylation (PMM2-CDG) results from mutations in pmm2, which encodes the phosphomannomutase (Pmm) that converts mannose-6-phosphate (M6P) to mannose-1-phosphate (M1P). Patients have wide-spectrum clinical abnormalities associated with impaired protein N-glycosylation. Although it has been widely proposed that Pmm2 deficiency depletes M1P, a precursor of GDP-mannose, and consequently suppresses lipid-linked oligosaccharide (LLO) levels needed for N-glycosylation, these deficiencies have not been demonstrated in patients or any animal model. Here we report a morpholino-based PMM2-CDG model in zebrafish. Morphant embryos had developmental abnormalities consistent with PMM2-CDG patients, including craniofacial defects and impaired motility associated with altered motor neurogenesis within the spinal cord. Significantly, global N-linked glycosylation and LLO levels were reduced in pmm2 morphants. Although M1P and GDP-mannose were below reliable detection/quantification limits, Pmm2 depletion unexpectedly caused accumulation of M6P, shown earlier to promote LLO cleavage in vitro. In pmm2 morphants, the free glycan by-products of LLO cleavage increased nearly twofold. Suppression of the M6P-synthesizing enzyme mannose phosphate isomerase within the pmm2 background normalized M6P levels and certain aspects of the craniofacial phenotype and abrogated pmm2-dependent LLO cleavage. In summary, we report the first zebrafish model of PMM2-CDG and uncover novel cellular insights not possible with other systems, including an M6P accumulation mechanism for underglycosylation.


Subject(s)
Congenital Disorders of Glycosylation/enzymology , Congenital Disorders of Glycosylation/pathology , Neurogenesis , Phosphotransferases (Phosphomutases)/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cartilage/drug effects , Cartilage/embryology , Cartilage/pathology , Cell Shape/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Congenital Disorders of Glycosylation/genetics , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/pathology , Disease Models, Animal , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Gene Expression Regulation, Developmental/drug effects , Glycosylation/drug effects , Lipopolysaccharides/metabolism , Mannose-6-Phosphate Isomerase/metabolism , Mannosephosphates/metabolism , Morpholinos/pharmacology , Motor Neurons/drug effects , Motor Neurons/pathology , Movement/drug effects , Neurogenesis/drug effects , Phosphotransferases (Phosphomutases)/deficiency , Phosphotransferases (Phosphomutases)/genetics , Skull/abnormalities , Skull/drug effects , Skull/embryology , Spinal Cord/drug effects , Spinal Cord/embryology , Spinal Cord/pathology , Substrate Specificity/drug effects , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...