Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Sci Rep ; 10(1): 5656, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32221390

RESUMO

Human phosphoglucomutase 1 (PGM1) is an evolutionary conserved enzyme that belongs to the ubiquitous and ancient α-D-phosphohexomutases, a large enzyme superfamily with members in all three domains of life. PGM1 catalyzes the bi-directional interconversion between α-D-glucose 1-phosphate (G1P) and α-D-glucose 6-phosphate (G6P), a reaction that is essential for normal carbohydrate metabolism and also important in the cytoplasmic biosynthesis of nucleotide sugars needed for glycan biosynthesis. Clinical studies have shown that mutations in the PGM1 gene may cause PGM1 deficiency, an inborn error of metabolism previously classified as a glycogen storage disease, and PGM1 deficiency was recently also shown to be a congenital disorder of glycosylation. Here we present three crystal structures of the isoform 2 variant of PGM1, both as a free enzyme and in complex with its substrate and product. The structures show the longer N-terminal of this PGM1 variant, and the ligand complex structures reveal for the first time the detailed structural basis for both G1P substrate and G6P product recognition by human PGM1. We also show that PGM1 and the paralogous gene PGM5 are the results of a gene duplication event in a common ancestor of jawed vertebrates, and, importantly, that both PGM1 isoforms are conserved and of functional significance in all vertebrates. Our finding that PGM1 encodes two equally conserved and functionally important isoforms in the human organism should be taken into account in the evaluation of disease-related missense mutations in patients in the future.


Assuntos
Fosfoglucomutase/genética , Fosfotransferases (Fosfomutases)/genética , Isoformas de Proteínas/genética , Animais , Domínio Catalítico/genética , Citoplasma/genética , Glucose-6-Fosfato/genética , Glucofosfatos/genética , Doença de Depósito de Glicogênio/genética , Glicosilação , Humanos , Ligantes , Mutação de Sentido Incorreto/genética , Vertebrados/genética
2.
Biochim Biophys Acta Gen Subj ; 1864(7): 129601, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32179131

RESUMO

BACKGROUND: Escherichia coli cells contain a homolog of presumed 5-keto-4-deoxyuronate isomerase (KduI) from pectin-degrading soil bacteria, but the catalytic activity of the E. coli protein (o-KduI) was never demonstrated. METHODS: The known three-dimensional structure of E. coli o-KduI was compared with the available structures of sugar-converting enzymes. Based on the results of this analysis, sugar isomerization activity of recombinant o-KduI was tested against a panel of D-sugars and their derivatives. RESULTS: The three-dimensional structure of o-KduI exhibits a close similarity with Pyrococcus furiosus cupin-type phosphoglucose isomerase. In accordance with this similarity, o-KduI was found to catalyze interconversion of glucose-6-phosphate and fructose-6-phosphate and, less efficiently, conversion of glucuronate to fructuronate. o-KduI was hexameric in crystals but represented a mixture of inactive hexamers and active dimers in solution and contained a tightly bound Zn2+ ion. Dilution, substrate binding and Zn2+ removal shifted the hexamer ⇆ dimer equilibrium to the dimers. CONCLUSIONS: Our findings identify o-KduI as a novel phosphosugar isomerase in E. coli, whose activity may be regulated by changes in oligomeric structure. GENERAL SIGNIFICANCE: More than 5700 protein sequences are annotated as KduI, but their enzymatic activity has not been directly demonstrated. E. coli o-KduI is the first characterized member of this group, and its enzymatic activity was found to be different from the predicted activity.


Assuntos
Aldose-Cetose Isomerases/genética , Glucose-6-Fosfato Isomerase/genética , Conformação Proteica , Aldose-Cetose Isomerases/ultraestrutura , Sequência de Aminoácidos/genética , Metabolismo dos Carboidratos/genética , Catálise , Cristalografia por Raios X , Escherichia coli/enzimologia , Frutosefosfatos/genética , Glucose-6-Fosfato/genética , Glucose-6-Fosfato Isomerase/ultraestrutura , Pyrococcus furiosus/enzimologia
3.
Nucleic Acids Res ; 48(2): 996-1009, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31799627

RESUMO

Dynamic regulation is an effective strategy for fine-tuning metabolic pathways in order to maximize target product synthesis. However, achieving dynamic and autonomous up- and down-regulation of the metabolic modules of interest simultaneously, still remains a great challenge. In this work, we created an autonomous dual-control (ADC) system, by combining CRISPRi-based NOT gates with novel biosensors of a key metabolite in the pathway of interest. By sensing the levels of the intermediate glucosamine-6-phosphate (GlcN6P) and self-adjusting the expression levels of the target genes accordingly with the GlcN6P biosensor and ADC system enabled feedback circuits, the metabolic flux towards the production of the high value nutraceutical N-acetylglucosamine (GlcNAc) could be balanced and optimized in Bacillus subtilis. As a result, the GlcNAc titer in a 15-l fed-batch bioreactor increased from 59.9 g/l to 97.1 g/l with acetoin production and 81.7 g/l to 131.6 g/l without acetoin production, indicating the robustness and stability of the synthetic circuits in a large bioreactor system. Remarkably, this self-regulatory methodology does not require any external level of control such as the use of inducer molecules or switching fermentation/environmental conditions. Moreover, the proposed programmable genetic circuits may be expanded to engineer other microbial cells and metabolic pathways.


Assuntos
Bacillus subtilis/isolamento & purificação , Técnicas Biossensoriais , Engenharia Metabólica/métodos , Redes e Vias Metabólicas/genética , Acetoína/metabolismo , Acetilglucosamina/metabolismo , Bacillus subtilis/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Redes Reguladoras de Genes/genética , Glucosamina/análogos & derivados , Glucosamina/genética , Glucosamina/metabolismo , Glucose/química , Glucose/genética , Glucose-6-Fosfato/análogos & derivados , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo
4.
J Mol Biol ; 431(19): 3933-3942, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31306666

RESUMO

The molecular mechanisms of pathological non-synonymous single-nucleotide polymorphisms are still the object of intensive research. To this end, we explore here whether non-synonymous single-nucleotide polymorphisms can work via allosteric mechanisms. Using structure-based statistical mechanical model of allostery and analyzing energetics of the effects of mutations in a set of 27 proteins with at least 50 pathological SNPs in each molecule, we found that, indeed, some SNPs can work allosterically. We illustrate the molecular basis of disease phenotypes caused by allosteric SNPs with the case studies of human galactose 1-phosphate uridyltransferase (GALT) and glucose-6-phosphate dehydrogenase (G6PD). We also found that mutations of a number of other residues in the protein may cause modulation comparable to those observed for known pathological SNPs. In order to explain this, we propose a notion of allosteric polymorphism, which implies the presence of a number of critical positions in the protein sequence, whose mutations can allosterically disrupt the protein function and result in a disease phenotype. We conclude that the emerging importance of allosteric polymorphism calls for the development of computational framework for analyzing the allosteric effects of mutations and their role in the modulation of protein activity.


Assuntos
Polimorfismo de Nucleotídeo Único/genética , Regulação Alostérica/genética , Glucose-6-Fosfato/genética , Humanos , Modelos Moleculares , Mutação/genética , UTP-Hexose-1-Fosfato Uridililtransferase/genética
5.
RNA Biol ; 16(8): 1055-1065, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31116083

RESUMO

Many bacterial small RNAs (sRNAs) are processed resulting in variants with roles potentially distinct from the primary sRNAs. In Enterobacteriaceae sRNA GlmZ activates expression of glmS by base-pairing when the levels of glucosamine-6-phosphate (GlcN6P) are low. GlmS synthesizes GlcN6P, which is required for cell envelope biosynthesis. When dispensable, GlmZ is cleaved by RNase E in the base-pairing sequence. Processing requires protein RapZ, which binds GlmZ and recruits RNase E by interaction. Cleavage is counteracted by the homologous sRNA GlmY, which accumulates upon GlcN6P scarcity and sequesters RapZ. Here, we report a novel role for a processed sRNA. We observed that processing of GlmZ is never complete in vivo. Even upon RapZ overproduction, a fraction of GlmZ remains full-length, while the 5' cleavage product (GlmZ*) accumulates. GlmZ* retains all elements required for RapZ binding. Accordingly, GlmZ* can displace full-length GlmZ from RapZ and counteract processing in vitro. To mimic GlmZ* in vivo, sRNA chimeras were employed consisting of foreign 3' ends including a terminator fused to the 3' end of GlmZ*. In vitro, these chimeras perform indistinguishable from GlmZ*. Expression of the chimeras in vivo inhibited processing of endogenous GlmZ, causing moderate upregulation of GlmS synthesis. Hence, accumulation of GlmZ* prevents complete GlmZ turnover. This mechanism may serve to adjust a robust glmS basal expression level that is buffered against fluctuations in RapZ availability.


Assuntos
Proteínas de Bactérias/genética , Endorribonucleases/genética , Proteínas de Escherichia coli/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Escherichia coli/genética , Retroalimentação Fisiológica , Regulação Bacteriana da Expressão Gênica/genética , Glucosamina/análogos & derivados , Glucosamina/genética , Glucose-6-Fosfato/análogos & derivados , Glucose-6-Fosfato/genética , RNA Bacteriano/genética , RNA Mensageiro/genética , Pequeno RNA não Traduzido/genética
6.
Biotechnol J ; 14(3): e1800264, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30105781

RESUMO

Glucosamine-6-phosphate N-acetyltransferase (GNA1) that catalyzes acetyl transfer from acetyl-coenzyme A to glucosamine-6-phosphate (GlcN-6P), and glutamine-fructose-6-phosphate aminotransferase (GlmS) that catalyzes the formation of GlcN-6P from fructose-6-phosphate (Fru-6P), are two key enzymes in Bacillus subtilis for the bioproduction of N-acetylglucosamine (GlcNAc), a nutraceutical that has various applications in healthcare. In this study, the expression of GNA1 and GlmS is fine-tuned by 5'-terminus fusion engineering to improve GlcNAc production. Specifically, the expression level of GNA1 is enhanced at the translational level via fusion of an epitope tag to the 5'-terminus of GNA1 gene and ribosome binding site (RBS) sequence engineering. Next, enhanced expression of GlmS is achieved at the transcriptional and translational levels by fusing an mRNA stabilizer to the 5'-terminus of GlmS gene. Under the control of GNA1 (fusion with cMyc tag and with the optimum RBS M-Rm) and GlmS (fusion with mRNA stabilizer ΔermC+14/7A), the GlcNAc titer and yield in the shake flask increase to 18.5 g L-1 and 0.37 g GlcNAc/g glucose, which are 2.9-fold and 2.3-fold that of the control, respectively. This synthetic pathway fine-tuning method at the transcriptional and translational levels by combinatorial modulation of regulatory elements, including epitope tag, RBS sequence, and mRNA stabilizer, might represent a general and effective approach for the construction of microbial cell factories.


Assuntos
Acetilglucosamina/genética , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Glucosamina 6-Fosfato N-Acetiltransferase/genética , Sítios de Ligação/genética , Glucosamina/análogos & derivados , Glucosamina/genética , Glucose/genética , Glucose-6-Fosfato/análogos & derivados , Glucose-6-Fosfato/genética , Engenharia Metabólica/métodos , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , Ribossomos/genética , Transcrição Gênica/genética
7.
Mol Ther ; 26(7): 1771-1782, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784585

RESUMO

Glycogen storage diseases (GSDs) of the liver are devastating disorders presenting with fasting hypoglycemia as well as hepatic glycogen and lipid accumulation, which could lead to long-term liver damage. Diet control is frequently utilized to manage the potentially dangerous hypoglycemia, but there is currently no effective pharmacological treatment for preventing hepatomegaly and concurrent liver metabolic abnormalities, which could lead to fibrosis, cirrhosis, and hepatocellular adenoma or carcinoma. In this study, we demonstrate that inhibition of glycogen synthesis using an RNAi approach to silence hepatic Gys2 expression effectively prevents glycogen synthesis, glycogen accumulation, hepatomegaly, fibrosis, and nodule development in a mouse model of GSD III. Mechanistically, reduction of accumulated abnormally structured glycogen prevents proliferation of hepatocytes and activation of myofibroblasts as well as infiltration of mononuclear cells. Additionally, we show that silencing Gys2 expression reduces hepatic steatosis in a mouse model of GSD type Ia, where we hypothesize that the reduction of glycogen also reduces the production of excess glucose-6-phosphate and its subsequent diversion to lipid synthesis. Our results support therapeutic silencing of GYS2 expression to prevent glycogen and lipid accumulation, which mediate initial signals that subsequently trigger cascades of long-term liver injury in GSDs.


Assuntos
Doença de Depósito de Glicogênio Tipo III/genética , Glicogênio Sintase/genética , Glicogênio/genética , Cirrose Hepática/genética , Cirrose Hepática/patologia , Fígado/patologia , Interferência de RNA/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Fibroblastos/patologia , Glucose-6-Fosfato/genética , Doença de Depósito de Glicogênio Tipo III/patologia , Hepatócitos/patologia , Hepatomegalia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Hum Mol Genet ; 26(22): 4395-4405, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28973635

RESUMO

Glycogen storage disease type-Ib (GSD-Ib), deficient in the glucose-6-phosphate transporter (G6PT), is characterized by impaired glucose homeostasis, myeloid dysfunction, and long-term risk of hepatocellular adenoma (HCA). We examined the efficacy of G6PT gene therapy in G6pt-/- mice using recombinant adeno-associated virus (rAAV) vectors, directed by either the G6PC or the G6PT promoter/enhancer. Both vectors corrected hepatic G6PT deficiency in murine GSD-Ib but the G6PC promoter/enhancer was more efficacious. Over a 78-week study, using dose titration of the rAAV vectors, we showed that G6pt-/- mice expressing 3-62% of normal hepatic G6PT activity exhibited a normalized liver phenotype. Two of the 12 mice expressing < 6% of normal hepatic G6PT activity developed HCA. All treated mice were leaner and more sensitive to insulin than wild-type mice. Mice expressing 3-22% of normal hepatic G6PT activity exhibited higher insulin sensitivity than mice expressing 44-62%. The levels of insulin sensitivity correlated with the magnitudes of hepatic carbohydrate response element binding protein signaling activation. In summary, we established the threshold of hepatic G6PT activity required to prevent tumor formation and showed that mice expressing 3-62% of normal hepatic G6PT activity maintained glucose homeostasis and were protected against age-related obesity and insulin resistance.


Assuntos
Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Animais , Antiporters/genética , Antiporters/metabolismo , Modelos Animais de Doenças , Vetores Genéticos , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Homeostase , Humanos , Resistência à Insulina , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Regiões Promotoras Genéticas
9.
Metab Eng ; 41: 212-221, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28465173

RESUMO

Oleaginous microalgae have great prospects in the fields of feed, nutrition, biofuel, etc. However, biomass and lipid productivity in microalgae remain a major economic and technological bottleneck. Here we present a novel regulatory target, glucose-6-phosphate dehydrogenase (G6PD) from the pentose phosphate pathway (PPP), in boosting microalgal lipid accumulation. G6PD, involved in the formation of NADPH demanded in fatty acid biosynthesis as reducing power, was characterized in oleaginous microalga Phaeodactylum tricornutum. In G6PD overexpressing microalgae, transcript abundance of G6PD increased by 4.4-fold, and G6PD enzyme activity increased by more than 3.1-fold with enhanced NADPH production. Consequently, the lipid content increased by 2.7-fold and reached up to 55.7% of dry weight, while cell growth was not apparently affected. The fatty acid composition exhibited significant changes, including a remarkable increase in monounsaturated fatty acids C16:1 and C18:1 concomitant with a decrease in polyunsaturated fatty acids C20:5 and C22:6. G6PD was localized to the chloroplast and its overexpression stimulated an increase in the number and size of oil bodies. Proteomic and metabolomic analyzes revealed that G6PD play a key role in regulating pentose phosphate pathway and subsequently upregulating NADPH consuming pathways such as fatty acid synthesis, thus eventually leading to lipid accumulation. Our findings show the critical role of G6PD in microalgal lipid accumulation by enhancing NADPH supply and demonstrate that G6PD is a promising target for metabolic engineering.


Assuntos
Proteínas de Cloroplastos , Diatomáceas , Ácidos Graxos Insaturados , Glucosefosfato Desidrogenase , Microalgas , NADP , Proteínas de Cloroplastos/genética , Proteínas de Cloroplastos/metabolismo , Diatomáceas/enzimologia , Diatomáceas/metabolismo , Ácidos Graxos Insaturados/biossíntese , Ácidos Graxos Insaturados/genética , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Microalgas/enzimologia , Microalgas/genética , NADP/genética , NADP/metabolismo , Via de Pentose Fosfato/genética
10.
Biomed Res Int ; 2017: 5470241, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197413

RESUMO

In recent years, multidrug resistance of Escherichia coli has become a serious problem. However, resistance to fosfomycin (FOM) has been low. We screened E. coli clinical isolates with reduced susceptibility to FOM and characterized molecular mechanisms of resistance and reduced susceptibility of these strains. Ten strains showing reduced FOM susceptibility (MIC ≥ 8 µg/mL) in 211 clinical isolates were found and examined. Acquisition of genes encoding FOM-modifying enzyme genes (fos genes) and mutations in murA that underlie high resistance to FOM were not observed. We examined ability of FOM incorporation via glucose-6-phosphate (G6P) transporter and sn-glycerol-3-phosphate transporter. In ten strains, nine showed lack of growth on M9 minimum salt agar supplemented with G6P. Eight of the ten strains showed fluctuated induction by G6P of uhpT that encodes G6P transporter expression. Nucleotide sequences of the uhpT, uhpA, glpT, ptsI, and cyaA shared several deletions and amino acid mutations in the nine strains with lack of growth on G6P-supplemented M9 agar. In conclusion, reduction of uhpT function is largely responsible for the reduced sensitivity to FOM in clinical isolates that have not acquired FOM-modifying genes or mutations in murA. However, there are a few strains whose mechanisms of reduced susceptibility to FOM are still unclear.


Assuntos
Infecções por Escherichia coli/tratamento farmacológico , Proteínas de Escherichia coli/biossíntese , Escherichia coli/efeitos dos fármacos , Fosfomicina/administração & dosagem , Proteínas de Transporte de Monossacarídeos/biossíntese , Alquil e Aril Transferases/genética , Sequência de Bases , Resistência a Múltiplos Medicamentos/genética , Escherichia coli/genética , Escherichia coli/patogenicidade , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Humanos , Proteínas de Transporte de Monossacarídeos/genética , Mutação
11.
Methods ; 106: 76-81, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27130889

RESUMO

Uniquely among known natural ribozymes that cleave RNA sequence-specifically, the glmS ribozyme-riboswitch employs a small molecule, glucosamine-6-phosphate (GlcN6P) as a catalytic cofactor. In vitro selection was employed to search for coenzyme-independent variants of this ribozyme. In addition to shedding light on the catalytic mechanism of the ribozyme, such variants could resemble the evolutionary ancestors of the modern, GlcN6P-regulated ribozyme-riboswitch. A mutant pool was constructed such that the secondary structure elements, which define the triply-pseudoknotted global fold of the ribozyme, was preserved. A stringent selection scheme that relies on thiol-mercury affinity chromatography for separating active and inactive sequences ultimately yielded a triple mutant with a cleavage rate exceeding 3min(-1) that only requires divalent cations for activity. Mutational analysis demonstrated that a point reversion of the variant toward the wild-type sequence was sufficient to partially restore GlcN6P-dependence, suggesting that coenzyme dependence can be readily be acquired by RNAs that adopt the glmS ribozyme fold. The methods employed to perform this selection experiment are described in detail in this review.


Assuntos
Glucosamina/análogos & derivados , Glucose-6-Fosfato/análogos & derivados , RNA Catalítico/genética , Riboswitch/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Catálise , Coenzimas/química , Coenzimas/genética , Glucosamina/química , Glucosamina/genética , Glucose-6-Fosfato/química , Glucose-6-Fosfato/genética , Mutação , Conformação de Ácido Nucleico , RNA Catalítico/química
12.
PLoS One ; 10(11): e0142121, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26540271

RESUMO

The strictly anaerobic, Gram-positive bacterium, Thermoanaerobacterium aotearoense SCUT27, is capable of producing ethanol, hydrogen and lactic acid by directly fermenting glucan, xylan and various lignocellulosically derived sugars. By using non-metabolizable and metabolizable sugars as substrates, we found that cellobiose, galactose, arabinose and starch utilization was strongly inhibited by the existence of 2-deoxyglucose (2-DG). However, the xylose and mannose consumptions were not markedly affected by 2-DG at the concentration of one-tenth of the metabolizable sugar. Accordingly, T. aotearoense SCUT27 could consume xylose and mannose in the presence of glucose. The carbon catabolite repression (CCR) related genes, ccpA, ptsH and hprK were confirmed to exist in T. aotearoense SCUT27 through gene cloning and protein characterization. The highly purified Histidine-containing Protein (HPr) could be specifically phosphorylated at Serine 46 by HPr kinase/phosphatase (HPrK/P) with no need to add fructose-1,6-bisphosphate (FBP) or glucose-6-phosphate (Glc-6-P) in the reaction mixture. The specific protein-interaction of catabolite control protein A (CcpA) and phosphorylated HPr was proved via affinity chromatography in the absence of formaldehyde. The equilibrium binding constant (KD) of CcpA and HPrSerP was determined as 2.22 ± 0.36 nM by surface plasmon resonance (SPR) analysis, indicating the high affinity between these two proteins.


Assuntos
Proteínas de Bactérias/genética , Carbono/metabolismo , Repressão Catabólica/genética , Metabolismo/genética , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Proteínas Serina-Treonina Quinases/genética , Thermoanaerobacterium/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica/genética , Glucose-6-Fosfato/genética , Histidina/genética
13.
Metab Eng ; 28: 104-113, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542851

RESUMO

Control of native enzyme levels is important when optimizing strains for overproduction of heterologous compounds. However, for many central metabolic enzymes, static knockdown results in poor growth and protein expression. We have developed a strategy for dynamically modulating the abundance of native enzymes within the host cell and applied this to a model system for myo-inositol production from glucose. This system relies on controlled degradation of a key glycolytic enzyme, phosphofructokinase-I (Pfk-I). Through tuning Pfk-I levels, we have been able to develop an Escherichia coli strain with a growth mode close to wild type and a production mode with an increased glucose-6-phosphate pool available for conversion into myo-inositol. The switch to production mode is trigged by inducer addition, allowing yield, titer, and productivity to be managed through induction time. By varying the time of Pfk-I degradation, we were able to achieve a two-fold improvement in yield and titers of myo-inositol.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Técnicas de Silenciamento de Genes , Glucose-6-Fosfato , Inositol , Fosfofrutoquinase-1 , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Inositol/genética , Inositol/metabolismo , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo
14.
Nat Chem Biol ; 9(12): 805-10, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24096303

RESUMO

Uniquely among known ribozymes, the glmS ribozyme-riboswitch requires a small-molecule coenzyme, glucosamine-6-phosphate (GlcN6P). Although consistent with its gene-regulatory function, the use of GlcN6P is unexpected because all of the other characterized self-cleaving ribozymes use RNA functional groups or divalent cations for catalysis. To determine what active site features make this ribozyme reliant on GlcN6P and to evaluate whether it might have evolved from a coenzyme-independent ancestor, we isolated a GlcN6P-independent variant through in vitro selection. Three active site mutations suffice to generate a highly reactive RNA that adopts the wild-type fold but uses divalent cations for catalysis and is insensitive to GlcN6P. Biochemical and crystallographic comparisons of wild-type and mutant ribozymes show that a handful of functional groups fine-tune the RNA to be either coenzyme or cation dependent. These results indicate that a few mutations can confer new biochemical activities on structured RNAs. Thus, families of structurally related ribozymes with divergent function may exist.


Assuntos
Proteínas de Bactérias/metabolismo , Dobramento de Proteína , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Glucosamina/análogos & derivados , Glucosamina/genética , Glucosamina/metabolismo , Glucose-6-Fosfato/análogos & derivados , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Cinética , Modelos Moleculares , Conformação Proteica , RNA/genética , RNA/metabolismo , RNA Catalítico , Thermoanaerobacter/genética , Thermoanaerobacter/metabolismo
15.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 10): 2008-16, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24100319

RESUMO

The first structure of a bacterial α-phosphoglucomutase with an overall fold similar to eukaryotic phosphomannomutases is reported. Unlike most α-phosphoglucomutases within the α-D-phosphohexomutase superfamily, it belongs to subclass IIb of the haloacid dehalogenase superfamily (HADSF). It catalyzes the reversible conversion of α-glucose 1-phosphate to glucose 6-phosphate. The crystal structure of α-phosphoglucomutase from Lactococcus lactis (APGM) was determined at 1.5 Šresolution and contains a sulfate and a glycerol bound at the enzyme active site that partially mimic the substrate. A dimeric form of APGM is present in the crystal and in solution, an arrangement that may be functionally relevant. The catalytic mechanism of APGM and its strict specificity towards α-glucose 1-phosphate are discussed.


Assuntos
Proteínas de Bactérias/química , Lactococcus lactis/enzimologia , Fosfotransferases (Fosfomutases)/química , Proteínas de Bactérias/genética , Domínio Catalítico/genética , Cristalografia por Raios X , Glucose-6-Fosfato/química , Glucose-6-Fosfato/genética , Glucofosfatos/química , Glucofosfatos/genética , Hidrolases/química , Hidrolases/classificação , Hidrolases/genética , Lactococcus lactis/genética , Mimetismo Molecular/genética , Família Multigênica , Fosfotransferases (Fosfomutases)/classificação , Fosfotransferases (Fosfomutases)/genética , Ligação Proteica/genética , Especificidade por Substrato/genética
16.
J Biol Chem ; 287(43): 36455-64, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22932905

RESUMO

Cellular methylation processes enable expression of gluconeogenic enzymes and metabolism of the nutrient selenium. Selenium status has been proposed to relate to type II diabetes risk, and plasma levels of selenoprotein P (SEPP1) have been positively correlated with insulin resistance. Increased expression of gluconeogenic enzymes glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase 1 (PCK1) has negative consequences for blood glucose management in type II diabetics. Transcriptional regulation of SEPP1 is directed by the same transcription factors that control the expression of G6PC and PCK1, and these factors are activated by methylation of arginine residues. We sought to determine whether expression of SEPP1 and the aforementioned glucoconeogenic enzymes are regulated by protein methylation, the levels of which are reliant upon adequate S-adenosylmethionine (SAM) and inhibited by S-adenosylhomocysteine (SAH). We treated a human hepatocyte cell line, HepG2, with inhibitors of adenosylhomocysteine hydrolase (AHCY) known to increase concentration of SAH before analysis of G6PC, PCK1, and SEPP1 expression. Increasing SAH decreased 1) the SAM/SAH ratio, 2) protein-arginine methylation, and 3) expression of SEPP1, G6PC, and PCK1 transcripts. Furthermore, hormone-dependent induction of gluconeogenic enzymes was reduced by inhibition of protein methylation. When protein-arginine methyltransferase 1 expression was reduced by siRNA treatment, G6PC expression was inhibited. These findings demonstrate that hepatocellular SAM-dependent protein methylation is required for both SEPP1 and gluconeogenic enzyme expression and that inhibition of protein arginine methylation might provide a route to therapeutic interventions in type II diabetes.


Assuntos
Regulação da Expressão Gênica , Gluconeogênese , S-Adenosilmetionina/metabolismo , Selenoproteína P/biossíntese , Adenosil-Homocisteinase/biossíntese , Adenosil-Homocisteinase/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metilação , Fosfoenolpiruvato Carboxiquinase (GTP)/biossíntese , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Proteína-Arginina N-Metiltransferases/biossíntese , Proteína-Arginina N-Metiltransferases/genética , S-Adenosilmetionina/genética , Selenoproteína P/genética
18.
J Cell Biochem ; 112(1): 30-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20506195

RESUMO

Branched-chain amino acids (BCAAs) have a potential to improve glucose metabolism in cirrhotic patients; however, the contribution of liver in this process has not been clarified. To estimate the effect of BCAA on glucose metabolism in liver, we evaluated the mRNA expression levels of glucose-sensing apparatus genes in HepG2 cells and in rat liver after oral administration of BCAA. HepG2 cells were cultured in low glucose (100 mg/dl) or high glucose (400 mg/dl) in the absence or presence of BCAA. The mRNA expression levels and protein levels of GLUT2 and liver-type glucokinase (L-GK) were estimated using RT-PCR and immunoblotting. The expression levels of transcriptional factors, including SREBP-1c, ChREBP, PPAR-γm and LXRα, were estimated. The mRNA expression levels of transcriptional factors, glycogen synthase, and genes involved in gluconeogenesis were evaluated in rat liver at 3 h after the administration of BCAA. BCAA accelerated the expression of GLUT2 and L-GK in HepG2 cells in high glucose. Expression levels of ChREBP, SREBP-1c, and LXRα were also increased in this condition. BCAA administration enhanced the mRNA expression levels of L-GK, SREBP-1c, and LXRα and suppressed the expression levels of G-6-Pase in rat liver, without affecting the expression levels of glycogen synthase or serum glucose concentrations. BCAA administration enhanced the bioactivity of the glucose-sensing apparatus, probably via the activation of a transcriptional mechanism, suggesting that these amino acids may improve glucose metabolism through the accelerated utility of glucose and glucose-6-phosphate in the liver.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Glucose/metabolismo , Fígado/metabolismo , Aminoácidos de Cadeia Ramificada/administração & dosagem , Animais , Glucoquinase/genética , Glucoquinase/metabolismo , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Células Hep G2 , Humanos , Fígado/enzimologia , Masculino , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
19.
FEBS Lett ; 584(14): 3073-9, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20621833

RESUMO

In this study, we observed that low glucose or fructose reduces the increase in hypoxia-inducible factor-1alpha (HIF-1alpha) protein under hypoxic conditions. 6-Aminonicotinamide (6-AN), an inhibitor of the pentose phosphate pathway (PPP), also inhibited the increase of HIF-1alpha protein under hypoxic conditions, while the reduced protein levels of HIF-1alpha by low glucose were apparently recovered by the addition of MG-132 or NADPH. Moreover, siRNA for glucose-6-phosphate dehydrogenase, which produces NADPH, reduced the increase in HIF-1alpha protein. On the other hand, cobalt-induced expression of HIF-1alpha protein was not affected by low glucose or 6-AN under normoxic conditions. In conclusion, glucose metabolism through the PPP, but not in glycolysis, plays an important role in the stabilization of HIF-1alpha protein under hypoxic conditions.


Assuntos
Glucose/metabolismo , Hipóxia/metabolismo , 6-Aminonicotinamida , Glucose-6-Fosfato/genética , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Glicólise/genética , Humanos , Hipóxia/genética , Leupeptinas , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Via de Pentose Fosfato/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA