Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 375
Filter
Add more filters

Publication year range
1.
Microb Cell Fact ; 23(1): 173, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867236

ABSTRACT

BACKGROUND: The microbial chiral product (R)-3-hydroxybutyrate (3-HB) is a gateway to several industrial and medical compounds. Acetyl-CoA is the key precursor for 3-HB, and several native pathways compete with 3-HB production. The principal competing pathway in wild-type Escherichia coli for acetyl-CoA is mediated by citrate synthase (coded by gltA), which directs over 60% of the acetyl-CoA into the tricarboxylic acid cycle. Eliminating citrate synthase activity (deletion of gltA) prevents growth on glucose as the sole carbon source. In this study, an alternative approach is used to generate an increased yield of 3-HB: citrate synthase activity is reduced but not eliminated by targeted substitutions in the chromosomally expressed enzyme. RESULTS: Five E. coli GltA variants were examined for 3-HB production via heterologous overexpression of a thiolase (phaA) and NADPH-dependent acetoacetyl-CoA reductase (phaB) from Cupriavidus necator. In shake flask studies, four variants showed nearly 5-fold greater 3-HB yield compared to the wild-type, although pyruvate accumulated. Overexpression of either native thioesterases TesB or YciA eliminated pyruvate formation, but diverted acetyl-CoA towards acetate formation. Overexpression of pantothenate kinase similarly decreased pyruvate formation but did not improve 3-HB yield. Controlled batch studies at the 1.25 L scale demonstrated that the GltA[A267T] variant produced the greatest 3-HB titer of 4.9 g/L with a yield of 0.17 g/g. In a phosphate-starved repeated batch process, E. coli ldhA poxB pta-ackA gltA::gltA[A267T] generated 15.9 g/L 3-HB (effective concentration of 21.3 g/L with dilution) with yield of 0.16 g/g from glucose as the sole carbon source. CONCLUSIONS: This study demonstrates that GltA variants offer a means to affect the generation of acetyl-CoA derived products. This approach should benefit a wide range of acetyl-CoA derived biochemical products in E. coli and other microbes. Enhancing substrate affinity of the introduced pathway genes like thiolase towards acetyl-CoA will likely further increase the flux towards 3-HB while reducing pyruvate and acetate accumulation.


Subject(s)
3-Hydroxybutyric Acid , Acetyl Coenzyme A , Citrate (si)-Synthase , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Acetyl Coenzyme A/metabolism , Citrate (si)-Synthase/metabolism , Citrate (si)-Synthase/genetics , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/biosynthesis , Metabolic Engineering/methods , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Ketone Oxidoreductases/metabolism , Ketone Oxidoreductases/genetics , Alcohol Oxidoreductases
2.
Hum Mol Genet ; 29(7): 1168-1179, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32160276

ABSTRACT

Glutaric aciduria type 1 (GA1) is an inborn error of lysine degradation characterized by a specific encephalopathy that is caused by toxic accumulation of lysine degradation intermediates. Substrate reduction through inhibition of DHTKD1, an enzyme upstream of the defective glutaryl-CoA dehydrogenase, has been investigated as a potential therapy, but revealed the existence of an alternative enzymatic source of glutaryl-CoA. Here, we show that loss of DHTKD1 in glutaryl-CoA dehydrogenase-deficient HEK-293 cells leads to a 2-fold decrease in the established GA1 clinical biomarker glutarylcarnitine and demonstrate that oxoglutarate dehydrogenase (OGDH) is responsible for this remaining glutarylcarnitine production. We furthermore show that DHTKD1 interacts with OGDH, dihydrolipoyl succinyltransferase and dihydrolipoamide dehydrogenase to form a hybrid 2-oxoglutaric and 2-oxoadipic acid dehydrogenase complex. In summary, 2-oxoadipic acid is a substrate for DHTKD1, but also for OGDH in a cell model system. The classical 2-oxoglutaric dehydrogenase complex can exist as a previously undiscovered hybrid containing DHTKD1 displaying improved kinetics towards 2-oxoadipic acid.


Subject(s)
Acyl Coenzyme A/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Ketoglutarate Dehydrogenase Complex/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Cells, Cultured , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , HEK293 Cells , Humans , Ketone Oxidoreductases/genetics , Substrate Specificity/genetics
3.
Biochem Biophys Res Commun ; 587: 153-159, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34875534

ABSTRACT

Pyruvate dehydrogenase kinase 1 (PDK1) is a Ser/Thr kinase that inactivates mitochondrial pyruvate dehydrogenase (PDH), leading to switch of glucose metabolism from mitochondrial oxidation to aerobic glycolysis. We previously reported that PDK1 inhibition is a potent therapeutic strategy in multiple myeloma (MM). However, availability of PDK1 inhibitors, which are effective at low concentrations, are limited at present, making PDK1 inhibition difficult to apply in the clinic. In the present study, we examined the efficacy and mechanism of action of JX06, a novel PDK1 inhibitor, against MM cells. We confirmed that PDK1 is highly expressed in normal plasma cells and MM cells using publicly available gene expression datasets. JX06 suppressed cell growth and induced apoptosis against MM cells from approximately 0.5 µM JX06 treatment reduced PDH phosphorylation, suggesting that JX06 is indeed inhibiting PDK1. Intracellular metabolite analysis revealed that JX06 treatment reduced metabolites associated with glucose metabolism of MM cells. Additionally, JX06 in combination with a well-known proteasome inhibitor, bortezomib, significantly increased MM cell death, which raises the possibility of combination use of JX06 with proteasome inhibitors in the clinic. These findings demonstrate that PDK1 can be potentially targeted by JX06 in MM through glycolysis inhibition, leading to a novel therapeutic strategy in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Disulfiram/analogs & derivatives , Enzyme Inhibitors/pharmacology , Glycolysis/drug effects , Morpholines/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/genetics , Bortezomib/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Datasets as Topic , Disulfiram/pharmacology , Drug Synergism , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phosphorylation/drug effects , Plasma Cells/drug effects , Plasma Cells/enzymology , Plasma Cells/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
4.
J Biol Chem ; 295(23): 8078-8095, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32303640

ABSTRACT

2-Oxoadipate dehydrogenase (E1a, also known as DHTKD1, dehydrogenase E1, and transketolase domain-containing protein 1) is a thiamin diphosphate-dependent enzyme and part of the 2-oxoadipate dehydrogenase complex (OADHc) in l-lysine catabolism. Genetic findings have linked mutations in the DHTKD1 gene to several metabolic disorders. These include α-aminoadipic and α-ketoadipic aciduria (AMOXAD), a rare disorder of l-lysine, l-hydroxylysine, and l-tryptophan catabolism, associated with clinical presentations such as developmental delay, mild-to-severe intellectual disability, ataxia, epilepsy, and behavioral disorders that cannot currently be managed by available treatments. A heterozygous missense mutation, c.2185G→A (p.G729R), in DHTKD1 has been identified in most AMOXAD cases. Here, we report that the G729R E1a variant when assembled into OADHc in vitro displays a 50-fold decrease in catalytic efficiency for NADH production and a significantly reduced rate of glutaryl-CoA production by dihydrolipoamide succinyl-transferase (E2o). However, the G729R E1a substitution did not affect any of the three side-reactions associated solely with G729R E1a, prompting us to determine the structure-function effects of this mutation. A multipronged systematic analysis of the reaction rates in the OADHc pathway, supplemented with results from chemical cross-linking and hydrogen-deuterium exchange MS, revealed that the c.2185G→A DHTKD1 mutation affects E1a-E2o assembly, leading to impaired channeling of OADHc intermediates. Cross-linking between the C-terminal region of both E1a and G729R E1a with the E2o lipoyl and core domains suggested that correct positioning of the C-terminal E1a region is essential for the intermediate channeling. These findings may inform the development of interventions to counter the effects of pathogenic DHTKD1 mutations.


Subject(s)
Genetic Variation , Ketone Oxidoreductases/chemistry , Ketone Oxidoreductases/metabolism , Lysine/metabolism , Fibroblasts/chemistry , Fibroblasts/metabolism , Genetic Variation/genetics , Humans , Ketoglutarate Dehydrogenase Complex , Ketone Oxidoreductases/genetics , Kinetics , Lysine/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
5.
PLoS Biol ; 16(4): e2002907, 2018 04.
Article in English | MEDLINE | ID: mdl-29659562

ABSTRACT

A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose-fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Longevity/genetics , Obesity/genetics , Octamer Transcription Factor-1/genetics , Thiamine Deficiency/genetics , Thiamine/metabolism , Animals , Blood Glucose/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Acids/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Octamer Transcription Factor-1/deficiency , Signal Transduction , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology , Triglycerides/blood
6.
FASEB J ; 33(7): 8094-8109, 2019 07.
Article in English | MEDLINE | ID: mdl-30939245

ABSTRACT

Skeletal muscle satellite cells (SMSCs), the major stem cells responsible for the regeneration of skeletal muscle, are normally cell cycle arrested but differentiate to generate myocytes upon muscle damage, forming new myofibers along with self-renewing stem cells in preparation for subsequent injury. In this study, we investigated which factors stimulate the proliferation and differentiation of SMSCs and found that pyruvate, the end product of glycolysis, stimulates their differentiation. Pyruvate antagonizes the effects of hypoxia on preferential self-renewal of SMSCs through dephosphorylation or activation of pyruvate dehydrogenase (PDH), which mediates opening of the gateway from glycolysis to the tricarboxylic acid (TCA) cycle by producing acetyl coenzyme A from pyruvate. PDH kinase 1, highly expressed under hypoxia, is down-regulated under normoxic conditions, leading to an increase in dephosphorylated PDH. Conditional deletion of PDH in SMSCs affects cell divisions generating myocytes and subsequent myotube formation, inefficient skeletal muscle regeneration upon injury, and aggravated pathogenesis of a dystrophin-deficient mouse model of Duchenne muscular dystrophy. Thus, the flow from glycolysis to the TCA cycle mediated by PDH plays a pivotal role in the differentiation of SMSCs, which is critical for the progression of skeletal muscle regeneration.-Hori, S., Hiramuki, Y., Nishimura, D., Sato, F., Sehara-Fujisawa, A. PDH-mediated metabolic flow is critical for skeletal muscle stem cell differentiation and myotube formation during regeneration in mice.


Subject(s)
Cell Differentiation , Ketone Oxidoreductases/metabolism , Muscle Fibers, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/enzymology , Animals , Cell Line , Citric Acid Cycle , Gene Deletion , Glycolysis , Ketone Oxidoreductases/genetics , Mice , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology
7.
Molecules ; 25(18)2020 Sep 19.
Article in English | MEDLINE | ID: mdl-32961811

ABSTRACT

Di-n-butyl phthalate (DBP) is an extensively used plasticizer. Most investigations on DBP have been concentrated on its environmental distribution and toxicity to humans. However, information on the effects of plasticizers on algal species is scarce. This study verified the impacts of endocrine disruptor di-n-butyl phthalate ester on microalga Chlorella vulgaris by approaches of proteomics and gene ontology. The algal acute biotoxicity results showed that the 24h-EC50 of DBP for C. vulgaris was 4.95 mg L-1, which caused a decrease in the chlorophyll a content and an increase in the DBP concentration of C. vulgaris. Proteomic analysis led to the identification of 1257 C. vulgaris proteins. Sixty-one more proteins showed increased expression, compared to proteins with decreased expression. This result illustrates that exposure to DBP generally enhances protein expression in C. vulgaris. GO annotation showed that both acetolactate synthase (ALS) and GDP-L-fucose synthase 2 (GER2) decreased more than 1.5-fold after exposure to DBP. These effects could inhibit both the valine biosynthetic process and the nucleotide-sugar metabolic process in C. vulgaris. The results of this study demonstrate that DBP could inhibit growth and cause significant changes to the biosynthesis-relevant proteins in C. vulgaris.


Subject(s)
Chlorella vulgaris/drug effects , Dibutyl Phthalate/toxicity , Endocrine Disruptors/toxicity , Proteome/analysis , Proteomics/methods , Acetolactate Synthase/genetics , Chlorella vulgaris/genetics , Chlorella vulgaris/metabolism , Chlorophyll A/metabolism , Chromatography, High Pressure Liquid , Down-Regulation/drug effects , Gene Ontology , Ketone Oxidoreductases/genetics , Mass Spectrometry , Up-Regulation/drug effects
8.
J Biol Chem ; 293(45): 17402-17417, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30232153

ABSTRACT

In vertebrate cells, mitochondrial Ca2+ uptake by the mitochondrial calcium uniporter (MCU) leads to Ca2+-mediated stimulation of an intramitochondrial pyruvate dehydrogenase phosphatase (PDP). This enzyme dephosphorylates serine residues in the E1α subunit of pyruvate dehydrogenase (PDH), thereby activating PDH and resulting in increased ATP production. Although a phosphorylation/dephosphorylation cycle for the E1α subunit of PDH from nonvertebrate organisms has been described, the Ca2+-mediated PDP activation has not been studied. In this work, we investigated the Ca2+ sensitivity of two recombinant PDPs from the protozoan human parasites Trypanosoma cruzi (TcPDP) and T. brucei (TbPDP) and generated a TcPDP-KO cell line to establish TcPDP's role in cell bioenergetics and survival. Moreover, the mitochondrial localization of the TcPDP was studied by CRISPR/Cas9-mediated endogenous tagging. Our results indicate that TcPDP and TbPDP both are Ca2+-sensitive phosphatases. Of note, TcPDP-KO epimastigotes exhibited increased levels of phosphorylated TcPDH, slower growth and lower oxygen consumption rates than control cells, an increased AMP/ATP ratio and autophagy under starvation conditions, and reduced differentiation into infective metacyclic forms. Furthermore, TcPDP-KO trypomastigotes were impaired in infecting cultured host cells. We conclude that TcPDP is a Ca2+-stimulated mitochondrial phosphatase that dephosphorylates TcPDH and is required for normal growth, differentiation, infectivity, and energy metabolism in T. cruzi Our results support the view that one of the main roles of the MCU is linked to the regulation of intramitochondrial dehydrogenases.


Subject(s)
Chagas Disease/enzymology , Energy Metabolism , Ketone Oxidoreductases/metabolism , Protozoan Proteins/metabolism , Trypanosoma cruzi/enzymology , Cell Line , Chagas Disease/genetics , Chagas Disease/pathology , Gene Knockdown Techniques , Humans , Ketone Oxidoreductases/genetics , Phosphorylation/genetics , Protozoan Proteins/genetics , Trypanosoma cruzi/genetics
9.
Mol Genet Metab ; 126(4): 388-396, 2019 04.
Article in English | MEDLINE | ID: mdl-30709776

ABSTRACT

Inbred mouse strains are a cornerstone of translational research but paradoxically many strains carry mild inborn errors of metabolism. For example, α-aminoadipic acidemia and branched-chain ketoacid dehydrogenase deficiency are known in C57BL/6J mice. Using RNA sequencing, we now reveal the causal variants in Dhtkd1 and Bckdhb, and the molecular mechanism underlying these metabolic defects. C57BL/6J mice have decreased Dhtkd1 mRNA expression due to a solitary long terminal repeat (LTR) in intron 4 of Dhtkd1. This LTR harbors an alternate splice donor site leading to a partial splicing defect and as a consequence decreased total and functional Dhtkd1 mRNA, decreased DHTKD1 protein and α-aminoadipic acidemia. Similarly, C57BL/6J mice have decreased Bckdhb mRNA expression due to an LTR retrotransposon in intron 1 of Bckdhb. This transposable element encodes an alternative exon 1 causing aberrant splicing, decreased total and functional Bckdhb mRNA and decreased BCKDHB protein. Using a targeted metabolomics screen, we also reveal elevated plasma C5-carnitine in 129 substrains. This biochemical phenotype resembles isovaleric acidemia and is caused by an exonic splice mutation in Ivd leading to partial skipping of exon 10 and IVD protein deficiency. In summary, this study identifies three causal variants underlying mild inborn errors of metabolism in commonly used inbred mouse strains.


Subject(s)
Metabolism, Inborn Errors/genetics , Mice, Inbred Strains/genetics , Animals , DNA Transposable Elements/genetics , Ketone Oxidoreductases/genetics , Male , Metabolism, Inborn Errors/diagnosis , Metabolomics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Phenotype , Sequence Analysis, RNA
10.
Biochem Genet ; 57(3): 443-454, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30644007

ABSTRACT

3-Methyl-1-butanol (3MB) is a promising biofuel that can be produced from 2-ketoisocaproate via the common L-leucine biosynthesis pathway. Corynebacterium glutamicum was chosen as a host bacterium because of its strong resistance to isobutanol. In the current study, several strategies were designed to overproduce 3MB in C. glutamicum through a non-fermentation pathway. The engineered C. glutamicum mutant was obtained by silencing the pyruvate dehydrogenase gene complex (aceE) and deleting the lactic dehydrogenase gene (ldh), followed by mutagenesis with diethyl sulfate (DES) and selection with Fmoc-3-4-thiazolyl-L-alanine (FTA). The mutant could produce 659 mg/L of 3MB after 12 h of incubation. To facilitate carbon flux to 3MB biosynthesis, the engineered recombinant was also constructed without branched-chain acid aminotransferase (ilvE) activity by deleting the ilvE gene. This recombinant could produce 697 mg/L of 3MB after 12 h of incubation.


Subject(s)
Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Genetic Engineering , Mutation , Pentanols/metabolism , Chromosomes, Bacterial , Genes, Bacterial , Ketone Oxidoreductases/genetics
11.
Gastroenterology ; 152(1): 193-205.e10, 2017 01.
Article in English | MEDLINE | ID: mdl-27639802

ABSTRACT

BACKGROUND & AIMS: De novo synthesis of guanosine diphosphate (GDP)-fucose, a substrate for fucosylglycans, requires sequential reactions mediated by GDP-mannose 4,6-dehydratase (GMDS) and GDP-4-keto-6-deoxymannose 3,5-epimerase-4-reductase (FX or tissue specific transplantation antigen P35B [TSTA3]). GMDS deletions and mutations are found in 6%-13% of colorectal cancers; these mostly affect the ascending and transverse colon. We investigated whether a lack of fucosylation consequent to loss of GDP-fucose synthesis contributes to colon carcinogenesis. METHODS: FX deficiency and GMDS deletion produce the same biochemical phenotype of GDP-fucose deficiency. We studied a mouse model of fucosylation deficiency (Fx-/- mice) and mice with the full-length Fx gene (controls). Mice were placed on standard chow or fucose-containing diet (equivalent to a control fucosylglycan phenotype). Colon tissues were collected and analyzed histologically or by enzyme-linked immunosorbent assays to measure cytokine levels; T cells also were collected and analyzed. Fecal samples were analyzed by 16s ribosomal RNA sequencing. Mucosal barrier function was measured by uptake of fluorescent dextran. We transplanted bone marrow cells from Fx-/- or control mice (Ly5.2) into irradiated 8-week-old Fx-/- or control mice (Ly5.1). We performed immunohistochemical analyses for expression of Notch and the hes family bHLH transcription factor (HES1) in colon tissues from mice and a panel of 60 human colorectal cancer specimens (27 left-sided, 33 right-sided). RESULTS: Fx-/- mice developed colitis and serrated-like lesions. The intestinal pathology of Fx-/- mice was reversed by addition of fucose to the diet, which restored fucosylation via a salvage pathway. In the absence of fucosylation, dysplasia appeared and progressed to adenocarcinoma in up to 40% of mice, affecting mainly the right colon and cecum. Notch was not activated in Fx-/- mice fed standard chow, leading to decreased expression of its target Hes1. Fucosylation deficiency altered the composition of the fecal microbiota, reduced mucosal barrier function, and altered epithelial proliferation marked by Ki67. Fx-/- mice receiving control bone marrow cells had intestinal inflammation and dysplasia, and reduced expression of cytokines produced by cytotoxic T cells. Human sessile serrated adenomas and right-sided colorectal tumors with epigenetic loss of MutL homolog 1 (MLH1) had lost or had lower levels of HES1 than other colorectal tumor types or nontumor tissues. CONCLUSIONS: In mice, fucosylation deficiency leads to colitis and adenocarcinoma, loss of Notch activation, and down-regulation of Hes1. HES1 loss correlates with the development of human right-sided colorectal tumors with epigenetic loss of MLH1. These findings indicate that carcinogenesis in a subset of colon cancer is consequent to a molecular mechanism driven by fucosylation deficiency and/or HES1-loss.


Subject(s)
Adenocarcinoma/etiology , Carbohydrate Epimerases/deficiency , Colitis/etiology , Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/etiology , Intestinal Mucosa/metabolism , Ketone Oxidoreductases/deficiency , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow Transplantation , Carbohydrate Epimerases/genetics , Carcinogenesis , Cecum/pathology , Cell Proliferation , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Colonic Neoplasms/chemistry , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Feces/microbiology , Female , Fucose/administration & dosage , Gastrointestinal Microbiome , Guanosine Diphosphate Fucose/biosynthesis , Guanosine Diphosphate Fucose/deficiency , Humans , Ketone Oxidoreductases/genetics , Male , Mice , Mice, Knockout , Middle Aged , Permeability , RNA, Messenger/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Transcription Factor HES-1/analysis , Transcription Factor HES-1/metabolism , Young Adult
12.
Biotechnol Bioeng ; 114(3): 632-644, 2017 03.
Article in English | MEDLINE | ID: mdl-27666939

ABSTRACT

During antibody dependent cell cytotoxicity (ADCC) the target cells are killed by monocytes and natural killer cells. ADCC is enhanced when the antibody heavy chain's core N-linked glycan lacks the fucose molecule(s). Several strategies have been utilized to generate fully afucosylated antibodies. A commonly used and efficient approach has been knocking out the FUT8 gene of the Chinese hamster ovary (CHO) host cells, which results in expression of antibody molecules with fully afucosylated glycans. However, a major drawback of the FUT8-KO host is the requirement for undertaking two separate cell line development (CLD) efforts in order to obtain both primarily fucosylated and fully afucosylated antibody species for comparative studies in vitro and in vivo. Even more challenging is obtaining primarily fucosylated and FUT8-KO clones with similar enough product quality attributes to ensure that any observed ADCC advantage(s) can be strictly attributed to afucosylation. Here, we report generation and use of a FX knockout (FXKO) CHO host cell line that is capable of expressing antibody molecules with either primarily fucosylated or fully afucosylated glycan profiles with otherwise similar product quality attributes, depending on addition of fucose to the cell culture media. Hence, the FXKO host not only obviates the requirement for undertaking two separate CLD efforts, but it also averts the need for screening many colonies to identify clones with comparable product qualities. Finally, FXKO clones can express antibodies with the desired ratio of primarily fucosylated to afucosylated glycans when fucose is titrated into the production media, to allow achieving intended levels of FcγRIII-binding and ADCC for an antibody. Biotechnol. Bioeng. 2017;114: 632-644. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antibodies/chemistry , Fucose/metabolism , Ketone Oxidoreductases/genetics , Protein Engineering/methods , Recombinant Proteins/chemistry , Animals , Antibodies/genetics , Antibodies/metabolism , CHO Cells , CRISPR-Cas Systems , Cricetinae , Cricetulus , Fucose/chemistry , Gene Editing , Gene Knockout Techniques , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Biosci Biotechnol Biochem ; 81(7): 1386-1393, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28463550

ABSTRACT

In Euglena gracilis, pyruvate:NADP+ oxidoreductase, in addition to the pyruvate dehydrogenase complex, functions for the oxidative decarboxylation of pyruvate in the mitochondria. Furthermore, the 2-oxoglutarate dehydrogenase complex is absent, and instead 2-oxoglutarate decarboxylase is found in the mitochondria. To elucidate the central carbon and energy metabolisms in Euglena under aerobic and anaerobic conditions, physiological significances of these enzymes involved in 2-oxoacid metabolism were examined by gene silencing experiments. The pyruvate dehydrogenase complex was indispensable for aerobic cell growth in a glucose medium, although its activity was less than 1% of that of pyruvate:NADP+ oxidoreductase. In contrast, pyruvate:NADP+ oxidoreductase was only involved in the anaerobic energy metabolism (wax ester fermentation). Aerobic cell growth was almost completely suppressed when the 2-oxoglutarate decarboxylase gene was silenced, suggesting that the tricarboxylic acid cycle is modified in Euglena and 2-oxoglutarate decarboxylase takes the place of the 2-oxoglutarate dehydrogenase complex in the aerobic respiratory metabolism.


Subject(s)
Carboxy-Lyases/metabolism , Energy Metabolism/genetics , Euglena gracilis/enzymology , Ketone Oxidoreductases/metabolism , Mitochondria/metabolism , Protozoan Proteins/metabolism , Aerobiosis/genetics , Amino Acid Sequence , Anaerobiosis/genetics , Carboxy-Lyases/genetics , Cloning, Molecular , Culture Media/chemistry , Decarboxylation , Escherichia coli/genetics , Escherichia coli/metabolism , Euglena gracilis/genetics , Fermentation , Gene Expression , Gene Expression Regulation , Glucose/metabolism , Ketone Oxidoreductases/genetics , Kinetics , Mitochondria/genetics , Oxidation-Reduction , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
15.
Mol Microbiol ; 96(6): 1136-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25766665

ABSTRACT

The Campylobacter jejuni capsular polysaccharide is important for virulence and often contains a modified heptose. In strain ATCC 700819 (a.k.a. NCTC 11168), the modified heptose branches off from the capsular backbone and is directly exposed to the environment. We reported previously that the enzymes encoded by wcaG, mlghB and mlghC are involved in heptose modification. Here, we show that inactivation of any of these genes leads to production of capsule lacking modified heptose and alters the transcription of other capsule modification genes differentially. Inactivation of mlghB or mlghC, but not of wcaG, decreased susceptibility to bile salts and abrogated invasion of intestinal cells. All mutants showed increased sensitivity to serum killing, especially wcaG::cat, and had defects in colonization and persistence in chicken intestine, but did not show significant differences in adhesion, phagocytosis and intracellular survival in murine macrophages. Together, our findings suggest that the capsular heptose modification pathway contributes to bacterial resistance against gastrointestinal host defenses and supports bacterial persistence via its role in serum resistance and invasion of intestinal cells. Our data further suggest a dynamic regulation of expression of this pathway in the gastrointestinal tract.


Subject(s)
Bacterial Capsules/metabolism , Campylobacter jejuni/pathogenicity , Heptoses/metabolism , Polysaccharides, Bacterial/metabolism , Animals , Bacterial Capsules/genetics , Bile Acids and Salts/metabolism , Caco-2 Cells , Campylobacter Infections/microbiology , Campylobacter jejuni/enzymology , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Carbohydrate Sequence , Chickens , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/microbiology , Gene Knockout Techniques , Heptoses/genetics , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Molecular Sequence Data , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , RAW 264.7 Cells , Virulence
16.
Tumour Biol ; 37(4): 4963-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26531722

ABSTRACT

TSTA3 participates in enzyme metabolism and affects glycosylation processes, and abnormal glycosylation influences the malignant transformation of cells and tumor development. However, studies have not examined the molecular biological function of TSTA3 in breast cancer (BC). The expression of TSTA3 was examined in BC tissues and cell lines. Kaplan-Meier survival tests and Cox regression were used to analyze prognosis. TSTA3 depletion was used to analyze cell function. The upstream miRNAs of TSTA3 were predicted, and the downstream target gene was analyzed using a RT2 Profiler™ PCR array. Our results show that TSTA3 was highly expressed in BC tissues and cells and was correlated with poor survival. The expression of TSTA3 was correlated with the TNM status (P < 0.01) and served as an independent prognostic factor (P = 0.041). TSTA3-siRNA decreased cell invasion and proliferation in vitro. miR-125a-5p and miR-125b are upstream targets of TSTA3, and a PCR array revealed that TSTA3 affects the CXCR4-CXCL12 genes. The findings suggest that miR-125a-5p/miR-125b suppress the expression of TSTA3, which controls cell proliferation and invasion by regulating CXCR4 expression. In conclusion, a high expression of TSTA3 exerts a proto-oncogenic effect during carcinogenesis and serves as an independent molecular marker for BC patients.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Carbohydrate Epimerases/biosynthesis , Carcinogenesis/genetics , Ketone Oxidoreductases/biosynthesis , MicroRNAs/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Carbohydrate Epimerases/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Ketone Oxidoreductases/genetics , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Staging , Prognosis , Proportional Hazards Models , RNA, Small Interfering
17.
J Inherit Metab Dis ; 39(6): 781-793, 2016 11.
Article in English | MEDLINE | ID: mdl-27586888

ABSTRACT

Lipoic acid (LA) is an essential cofactor required for the activity of five multienzymatic complexes that play a central role in the mitochondrial energy metabolism: four 2-oxoacid dehydrogenase complexes [pyruvate dehydrogenase (PDH), branched-chain ketoacid dehydrogenase (BCKDH), 2-ketoglutarate dehydrogenase (2-KGDH), and 2-oxoadipate dehydrogenase (2-OADH)] and the glycine cleavage system (GCS). LA is synthesized in a complex multistep process that requires appropriate function of the mitochondrial fatty acid synthesis (mtFASII) and the biogenesis of iron-sulphur (Fe-S) clusters. Defects in the biosynthesis of LA have been reported to be associated with multiple and severe defects of the mitochondrial energy metabolism. In recent years, disease-causing mutations in genes encoding for proteins involved in LA metabolism have been reported: NFU1, BOLA3, IBA57, LIAS, GLRX5, LIPT1, ISCA2, and LIPT2. These studies represented important progress in understanding the pathophysiology and molecular bases underlying these disorders. Here we review current knowledge regarding involvement of LA synthesis defects in human diseases with special emphasis on the diagnostic strategies for these disorders. The clinical and biochemical characteristics of patients with LA synthesis defects are discussed and a workup for the differential diagnosis proposed.


Subject(s)
Energy Metabolism/genetics , Thioctic Acid/biosynthesis , Thioctic Acid/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Amino Acid Oxidoreductases/genetics , Animals , Carrier Proteins/genetics , Diagnosis, Differential , Humans , Ketone Oxidoreductases/genetics , Mitochondria/genetics , Multienzyme Complexes/genetics , Transferases/genetics
18.
Biotechnol Lett ; 38(10): 1747-52, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27314478

ABSTRACT

OBJECTIVES: To achieve multienzymatic cascade synthesis of fucosyl oligosaccharide from D-mannose by two-step fermentation pathway in Escherichia coli. RESULTS: E. coli BL21(DE3) harboring pET-22b(+) vectors with six genes, i.e., glucokinase (Glk), phosphomannomutase (ManB), mannose-1-phosphate guanylytransferase (ManC), GDP-mannose 4,6-dehydratase (Gmd), GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase/4-reductase (WcaG), and α-1,2-fucosyltransferase (Fuct) were co-inoculated, and the multienzyme synthetic pathway was constructed to produce fucosyloligosaccharide using D-mannose as substrate. The product, analyzed by LC/MS, fucosyloligosaccharide was formed under the catalysis of Fuct using GDP-fucose as donor substrate and lactose as acceptor substrate. Fucosyloligosaccharides reached 22 mM by a two-step fermentation compared to 3.7 mM with a one-pot fermentation. CONCLUSIONS: Fucosyloligosaccharide was produced by a two-step fermentation to avoid the inhibitory effect of GDP-fucose on Gmd. Two-step fermentation is a rational synthetic pathway for accumulating fucosyloligosaccharide.


Subject(s)
Escherichia coli/growth & development , Fucose/chemistry , Mannose/metabolism , Multienzyme Complexes/genetics , Oligosaccharides/biosynthesis , Biosynthetic Pathways , Carbohydrate Dehydrogenases/genetics , Carbohydrate Epimerases/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Fermentation , Fucosyltransferases/genetics , Genetic Vectors/genetics , Glucokinase/genetics , Guanosine Diphosphate Fucose/chemistry , Ketone Oxidoreductases/genetics , Lactose/chemistry , Multienzyme Complexes/metabolism , Nucleotidyltransferases/genetics , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Phosphotransferases (Phosphomutases)/genetics , Transformation, Bacterial
19.
J Biol Chem ; 289(36): 25382-92, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25053415

ABSTRACT

myc(-/-) rat fibroblasts (KO cells) differ from myc(+/+) (WT) cells and KO cells with enforced Myc re-expression (KO-Myc cells) with respect to mitochondrial structure and function, utilization of glucose and glutamine as energy-generating substrates, and ATP levels. Specifically, KO cells demonstrate low levels of glycolysis and oxidative phosphorylation, dysfunctional mitochondria and electron transport chain complexes, and depleted ATP stores. We examined here how these cells adapt to their energy-deficient state and how they differ in their uptake and utilization of long- and medium-chain fatty acids such as palmitate and octanoate, respectively. Metabolic tracing of these molecules showed that KO cells preferentially utilize them as ß-oxidation substrates and that, rather than directing them into phospholipids, preferentially store them as neutral lipids. KO cell transcriptional profiling and functional assays revealed a generalized up-regulation of pathways involved in fatty acid transport and catabolism as well as evidence that these cells attempt to direct acetyl-CoA into the tricarboxylic acid (TCA) cycle for ATP production rather than utilizing it for anabolic purposes. Additional evidence to support this idea included the finding that AMP-dependent protein kinase was constitutively activated in KO cells. The complex control of pyruvate dehydrogenase, which links glycolysis to the TCA cycle, was also maximized to ensure the conversion of pyruvate to acetyl-CoA. Despite these efforts to maximize acetyl-CoA for energy-generating purposes, its levels remained chronically low in KO cells. This suggests that tumor cells with Myc deregulation might be susceptible to novel therapies that limit acetyl-CoA availability.


Subject(s)
Acetyl Coenzyme A/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Proto-Oncogene Proteins c-myc/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Citric Acid Cycle , Fibroblasts/cytology , Gene Expression Profiling , Gene Knockout Techniques , Glycolysis , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lipid Metabolism , Metabolic Networks and Pathways/genetics , Oxidation-Reduction , Oxidative Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Pyruvic Acid/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction
20.
Biochim Biophys Acta ; 1844(4): 736-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24491525

ABSTRACT

Heterodimeric 2-oxoacid:ferredoxin oxidoreductase (OFOR) from Sulfolobus tokodaii (StOFOR) has only one [4Fe-4S]²âº cluster, ligated by 4 Cys residues, C12, C15, C46, and C197. The enzyme has no other Cys. To elucidate the role of these Cys residues in holding of the iron-sulfur cluster in the course of oxidative decarboxylation of a 2-oxoacid, one or two of these Cys residues was/were substituted with Ala to yield C12A, C15A, C46A, C197A and C12/15A mutants. All the mutants showed the loss of iron-sulfur cluster, except the C197A one which retained some unidentified type of iron-sulfur cluster. On addition of pyruvate to OFOR, the wild type enzyme exhibited a chromophore at 320nm and a stable large EPR signal corresponding to a hydroxyethyl-ThDP radical, while the mutant enzymes did not show formation of any radical intermediate or production of acetyl-CoA, suggesting that the intact [4Fe-4S] cluster is necessary for these processes. The stable radical intermediate in wild type OFOR was rapidly decomposed upon addition of CoA in the absence of an electron acceptor. Non-oxidative decarboxylation of pyruvate, yielding acetaldehyde, has been reported to require CoA for other OFORs, but StOFOR catalyzed acetaldehyde production from pyruvate independent of CoA, regardless of whether the iron-sulfur cluster is intact [4Fe-4S] type or not. A comprehensive reaction scheme for StOFOR with a single cluster was proposed.


Subject(s)
Acetyl Coenzyme A/metabolism , Archaeal Proteins/metabolism , Cysteine/metabolism , Iron-Sulfur Proteins/metabolism , Ketone Oxidoreductases/metabolism , Sulfolobus/enzymology , Acetyl Coenzyme A/chemistry , Alanine/chemistry , Alanine/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Cysteine/chemistry , Decarboxylation , Escherichia coli/genetics , Escherichia coli/metabolism , Free Radicals , Iron/chemistry , Iron/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Ketone Oxidoreductases/chemistry , Ketone Oxidoreductases/genetics , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfolobus/chemistry , Sulfolobus/genetics , Sulfur/chemistry , Sulfur/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL