Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 295(25): 8460-8469, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32358064

RESUMO

Prions are lipidated proteins that interact with endogenous lipids and metal ions. They also assemble into multimers and propagate into the infectious scrapie form known as PrPSc The high-resolution structure of the infectious PrPSc state remains unknown, and its analysis largely relies on detergent-based preparations devoid of endogenous ligands. Here we designed polymers that allow isolation of endogenous membrane:protein assemblies in native nanodiscs without exposure to conventional detergents that destabilize protein structures and induce fibrillization. A set of styrene-maleic acid (SMA) polymers including a methylamine derivative facilitated gentle release of the infectious complexes for resolution of multimers, and a thiol-containing version promoted crystallization. Polymer extraction from brain homogenates from Syrian hamsters infected with Hyper prions and WT mice infected with Rocky Mountain Laboratories prions yielded infectious prion nanoparticles including oligomers and microfilaments bound to lipid vesicles. Lipid analysis revealed the brain phospholipids that associate with prion protofilaments, as well as those that are specifically enriched in prion assemblies captured by the methylamine-modified copolymer. A comparison of the infectivity of PrPSc attached to SMA lipid particles in mice and hamsters indicated that these amphipathic polymers offer a valuable tool for high-yield production of intact, detergent-free prions that retain in vivo activity. This native prion isolation method provides an avenue for producing relevant prion:lipid targets and potentially other proteins that form multimeric assemblies and fibrils on membranes.


Assuntos
Encéfalo/metabolismo , Lipídeos/química , Maleatos/química , Nanoestruturas/química , Poliestirenos/química , Proteínas Priônicas/metabolismo , Animais , Cricetinae , Maleatos/síntese química , Maleatos/metabolismo , Metilaminas/química , Camundongos , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Poliestirenos/síntese química , Poliestirenos/metabolismo , Proteínas Priônicas/química , Proteínas Priônicas/isolamento & purificação , Compostos de Sulfidrila/química
2.
Biotechnol Bioeng ; 118(5): 1840-1850, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33512000

RESUMO

Maleate is one of the most important unsaturated four-carbon dicarboxylic acids. It serves as an attractive building block in cosmetic, polymer, and pharmaceutical industries. Currently, industrial production of maleate relies mainly on chemical synthesis using benzene or butane as the starting materials under high temperature, which suffers from strict reaction conditions and low product yield. Here, we propose a novel biosynthetic pathway for maleate production in engineered Escherichia coli. We screened a superior salicylate 5-hydroxylase that can catalyze hydroxylation of salicylate into gentisate with high conversion rate. Then, introduction of salicylate biosynthetic pathway and gentisate ring cleavage pathway allowed the synthesis of maleate from glycerol. Further optimizations including enhancement of precursors supply, disruption of competing pathways, and construction of a pyruvate recycling system, boosted maleate titer to 2.4 ± 0.1 g/L in shake flask experiments. Subsequent scale-up biosynthesis of maleate in a 3-L bioreactor under fed-batch culture conditions enabled the production of 14.5 g/L of maleate, indicating a 268-fold improvement compared with the titer generated by the wildtype E. coli strain carrying the entire maleate biosynthetic pathway. This study provided a promising microbial platform for industrial level synthesis of maleate, and demonstrated the highest titer of maleate production in microorganisms so far.


Assuntos
Escherichia coli/genética , Maleatos/metabolismo , Engenharia Metabólica/métodos , Ácido Chiquímico/metabolismo , Técnicas de Cultura Celular por Lotes , Vias Biossintéticas/genética , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Glicerol/metabolismo , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo
3.
Int J Mol Sci ; 21(2)2020 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-31963306

RESUMO

Alcoholic beverages stimulate pancreatic enzyme secretions by inducing cholecystokinin (CCK) release. CCK is the major stimulatory hormone of pancreatic exocrine secretions, secreted from enteroendocrine I-cells of the intestine. Fermentation products of alcoholic beverages, such as maleic and succinic acids, influence gastric acid secretions. We hypothesize that maleic and succinic acids stimulate pancreatic exocrine secretions during beer and wine ingestion by increasing CCK secretions. Therefore, the effects of maleic and succinic acids on CCK release were studied in duodenal mucosal cells and the enteroendocrine cell line STC-1. Mucosal cells were perfused for 30 min with 5 min sampling intervals, STC-1 cells were studied under static incubation for 15 min, and supernatants were collected for CCK measurements. Succinate and maleate-induced CCK release were investigated. Succinate and maleate doses dependently stimulated CCK secretions from mucosal cells and STC-1 cells. Diltiazem, a calcium channel blocker, significantly inhibited succinate and maleate-induced CCK secretions from mucosal cells and STC-1 cells. Maleate and succinate did not show cytotoxicity in STC-1 cells. Our results indicate that succinate and maleate are novel CCK-releasing factors in fermented alcoholic beverages and could contribute to pancreatic exocrine secretions and their pathophysiology.


Assuntos
Colecistocinina/metabolismo , Mucosa Intestinal/citologia , Bebidas Alcoólicas , Animais , Linhagem Celular , Diltiazem/metabolismo , Fermentação/fisiologia , L-Lactato Desidrogenase/metabolismo , Maleatos/metabolismo , Ratos , Ácido Succínico/metabolismo
4.
Appl Environ Microbiol ; 85(19)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31324629

RESUMO

l-Aspartate has been widely used in medicine and the food and chemical industries. In this study, Serratia marcescens maleate cis-trans isomerase (MaiA) and Escherichia coli aspartase (AspA) were coupled and coexpressed in an engineered E. coli strain in which the byproduct metabolic pathway was inactivated. The engineered E. coli strain containing the dual-enzyme system (pMA) was employed to bioproduce l-aspartate from maleate with a conversion of 98%. We optimized the activity ratio of double enzymes through ribosome binding site (RBS) regulation and molecular modification of MaiA, resulting in an engineered strain: pMA-RBS4-G27A/G171A. The conversion of l-aspartate biotransformed from maleate using the pMA-RBS4-G27A/G171A strain was almost 100%. It required 40 min to complete the whole-cell catalysis, without the intermediate product and byproduct, compared to 120 min before optimization. The induction timing and the amount of inducer in a 5-liter fermentor were optimized for scale-up of the production of l-aspartate. The amount of produced l-aspartate using the cells obtained by fermentation reached 419.8 g/liter (3.15 M), and the conversion was 98.4%. Our study demonstrated an environmentally responsible and efficient method to bioproduce l-aspartate from maleate and provided an available pathway for the industrial production of l-aspartate. This work should greatly improve the economic benefits of l-aspartate, which can now be simply produced from maleate by the engineered strain constructed based on dual-enzyme coupling.IMPORTANCE l-Aspartate is currently produced from fumarate by biological methods, and fumarate is synthesized from maleate by chemical methods in industry. We established a biosynthesis method to produce l-aspartate from maleate that is environmentally responsible, convenient, and efficient. Compared to conventional l-aspartate production, no separation and purification of intermediate products is required, which could greatly improve production efficiency and reduce costs. As environmental issues are attracting increasing attention, conventional chemical methods gradually will be replaced by biological methods. Our results lay an important foundation for the industrialization of l-aspartate biosynthesis from maleate.


Assuntos
Ácido Aspártico/biossíntese , Escherichia coli/metabolismo , Maleatos/metabolismo , Serratia marcescens/enzimologia , Proteínas de Bactérias/metabolismo , Catálise , Escherichia coli/genética , Fermentação , Engenharia Metabólica , Serratia marcescens/genética , cis-trans-Isomerases/metabolismo
5.
Mol Cell Biochem ; 458(1-2): 99-112, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31032535

RESUMO

Maleic acid (MA), which has been reported to be highly excreted in propionic acidemia (PAcidemia), was demonstrated to cause nephropathy by bioenergetics impairment and oxidative stress, but the effects on kidney mitochondrial respiration has not yet been properly investigated. Therefore, the present study investigated the effects of MA (0.05-5 mM), as well as of propionic (PA) and 3-hydroxypropionic (3OHPA) acids (5 mM) that accumulate in PAcidemia, on mitochondrial respiration supported by glutamate, glutamate plus malate or succinate in mitochondrial fractions and homogenates from rat kidney, as well as in permeabilized kidney cells. MA markedly decreased oxygen consumption in state 3 (ADP-stimulated) and uncoupled (CCCP-stimulated) respiration in glutamate and glutamate plus malate-respiring mitochondria, with less prominent effects when using succinate. We also found that PA significantly decreased state 3 and uncoupled respiration in glutamate- and glutamate plus malate-supported mitochondria, whereas 3OHPA provoked milder or no changes. Furthermore, glutamate dehydrogenase and α-ketoglutarate dehydrogenase activities necessary for glutamate oxidation were significantly inhibited by MA in a dose-dependent and competitive fashion. The MA-induced decrease of state 3 and uncoupled respiration found in mitochondrial fractions were also observed in homogenates and permeabilized renal cells that better mimic the in vivo cellular milieu. Taken together, our data indicate that MA, and PA to a lesser extent, disturb mitochondrial-oxidative metabolism in the kidney with the involvement of critical enzymes for glutamate oxidation. It is postulated that our present findings may be possibly involved in the chronic renal failure observed in patients with PAcidemia.


Assuntos
Glutamato Desidrogenase/metabolismo , Ácido Glutâmico/metabolismo , Complexo Cetoglutarato Desidrogenase/metabolismo , Rim/metabolismo , Maleatos/metabolismo , Mitocôndrias/metabolismo , Animais , Masculino , Oxirredução , Ratos , Ratos Wistar
6.
J Phys Chem A ; 123(36): 7710-7719, 2019 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-31433182

RESUMO

Maleamate amidohydrolase (NicF) is a key enzyme in vitamin B3 metabolism that catalyzes the hydrolysis of maleamate to produce maleic acid and ammonia. Unlike most members from the amidohydrolase superfamily it does not require a metal ion. Here, we use multiscale computational enzymology to investigate the catalytic mechanism, substrate binding, oxyanion hole, and roles of key active site residues of NicF from Bordetella bronchiseptica. In particular, molecular dynamics (MD) simulations, quantum mechanics/molecular mechanics (QM/MM) and QTAIM methods have been applied. The mechanism of the NicF-catalyzed reaction proceeds by a nucleophilic addition-elimination sequence involving the formation of a thioester enzyme intermediate (IC2 in stage 1) followed by hydrolysis of the thioester bond to form the products (stage 2). Consequently, the formation of IC2 in stage 1 is the rate-limiting step with a barrier of 88.8 kJ·mol-1 relative to the reactant complex, RC. Comparisons with related metal-dependent enzymes, particularly the zinc-dependent nicotinamidase from Streptococcus pneumonia (SpNic), have also been made to further illustrate unique features of the present mechanism. Along with -NH- donor groups of the oxyanion hole (i.e., HN-Thr146, HN-Cys150), the active site ß-hydroxyl of threonine (HO-ßThr146) is concluded to play a role in stabilizing the carbonyl oxygen of maleamate during the mechanism.


Assuntos
Amidoidrolases/química , Amidoidrolases/metabolismo , Biocatálise , Maleatos/metabolismo , Simulação de Dinâmica Molecular , Teoria Quântica , Bordetella bronchiseptica/enzimologia , Hidrólise , Maleatos/química , Estrutura Molecular
7.
Pestic Biochem Physiol ; 158: 47-53, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31378360

RESUMO

Buprofezin is a chitin synthesis inhibitor that is very effective against Homopteran pests, such as the white-backed planthopper (WBPH), S. furcifera (Horvath). In the present study, resistance selection, cross-resistance and mechanisms of buprofezin resistance were investigated in this planthopper species. However, the mechanism associated with resistance to growth regulator insecticides (IGRs) remains largely unknown. A resistant strain (Bup-R) with a resistance level (22-fold) to buprofezin was developed through continuous selection for 47 generations from a laboratory susceptible strain (Bup-S). The results showed that the Bup-R exhibited no cross-resistance to other tested insecticides. Synergism tests showed that piperonyl butoxide (PBO) (SR = 3.9-fold) and diethyl maleate (DEM) (SR = 1.8-fold) had synergistic effects on buprofezin toxicity in the resistant strain (F47). Enzyme activity results revealed an approximate 5.7-fold difference in cytochrome P450 monooxygenase and a 2-fold difference in glutathione S-transferase (GST) between the resistant and susceptible strains, suggesting that the increased activity of these two enzymes is likely the main detoxification mechanism involved in resistance to buprofezin in this species. Furthermore, the mRNA expression levels of cytochrome P450 (CYP) and GST genes by quantitative real-time PCR results indicated that sixteen P450 and one GST gene were significantly overexpressed in the Bup-R strain, among which thirteen P450 genes and one GST gene were >2-fold higher than in the Bup-S strain. The present study increases our knowledge of the buprofezin resistance mechanism in S. furcifera and provides a useful reference for integrated pest management (IPM) strategies.


Assuntos
Hemípteros/efeitos dos fármacos , Inseticidas/farmacologia , Tiadiazinas/farmacologia , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Hemípteros/metabolismo , Proteínas de Insetos/genética , Resistência a Inseticidas/genética , Maleatos/metabolismo , Butóxido de Piperonila/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
Bioprocess Biosyst Eng ; 42(9): 1483-1494, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31076865

RESUMO

Due to environmental concern, the research to date has tended to focus on how textile dye removal can be carried out in a greener manner. Therefore, this study aims to evaluate the decolorization and biotransformation pathway of Mordant Orange-1 (MO-1) by Cylindrocephalum aurelium RY06 (C. aurelium RY06). Decolorization study was conducted in a batch experiment including the investigation of the effects of physio-chemical parameters. Enzymatic activity of C. aurelium RY06 during the decolorization was also investigated. Moreover, transformation and biodegradation of MO-1 by C. aurelium RY06 were observed using the gas chromatography-mass spectrometry. Manganese peroxidase, lignin peroxidase, laccase, 1,2-dioxygenase, and 2,3-dioxygenase enzymes were detected during the decolorization. In general, the present work concluded that the MO-1 was successfully degraded by C. aurelium RY06 and transformed to be maleic acid and to be isophtalic acid.


Assuntos
Compostos Azo/metabolismo , Corantes/metabolismo , Fungos/metabolismo , Têxteis , Biotransformação , Proteínas Fúngicas/metabolismo , Maleatos/metabolismo , Oxirredutases/metabolismo , Ácidos Ftálicos/metabolismo
9.
Biochim Biophys Acta Bioenerg ; 1859(3): 215-225, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29291373

RESUMO

Intracytoplasmic vesicles (chromatophores) in the photosynthetic bacterium Rhodobacter sphaeroides represent a minimal structural and functional unit for absorbing photons and utilising their energy for the generation of ATP. The cytochrome bc1 complex (cytbc1) is one of the four major components of the chromatophore alongside the reaction centre-light harvesting 1-PufX core complex (RC-LH1-PufX), the light-harvesting 2 complex (LH2), and ATP synthase. Although the membrane organisation of these complexes is known, their local lipid environments have not been investigated. Here we utilise poly(styrene-alt-maleic acid) (SMA) co-polymers as a tool to simultaneously determine the local lipid environments of the RC-LH1-PufX, LH2 and cytbc1 complexes. SMA has previously been reported to effectively solubilise complexes in lipid-rich membrane regions whilst leaving lipid-poor ordered protein arrays intact. Here we show that SMA solubilises cytbc1 complexes with an efficiency of nearly 70%, whereas solubilisation of RC-LH1-PufX and LH2 was only 10% and 22% respectively. This high susceptibility of cytbc1 to SMA solubilisation is consistent with this complex residing in a locally lipid-rich region. SMA solubilised cytbc1 complexes retain their native dimeric structure and co-purify with 56±6 phospholipids from the chromatophore membrane. We extended this approach to the model cyanobacterium Synechocystis sp. PCC 6803, and show that the cytochrome b6f complex (cytb6f) and Photosystem II (PSII) complexes are susceptible to SMA solubilisation, suggesting they also reside in lipid-rich environments. Thus, lipid-rich membrane regions could be a general requirement for cytbc1/cytb6f complexes, providing a favourable local solvent to promote rapid quinol/quinone binding and release at the Q0 and Qi sites.


Assuntos
Proteínas de Bactérias/química , Complexo Citocromos b6f/química , Complexo III da Cadeia de Transporte de Elétrons/química , Maleatos/química , Lipídeos de Membrana/química , Poliestirenos/química , Cromatóforos Bacterianos/química , Cromatóforos Bacterianos/metabolismo , Cromatóforos Bacterianos/ultraestrutura , Proteínas de Bactérias/metabolismo , Complexo Citocromos b6f/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Transferência de Energia , Complexos de Proteínas Captadores de Luz/química , Complexos de Proteínas Captadores de Luz/metabolismo , Maleatos/metabolismo , Lipídeos de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Complexo de Proteína do Fotossistema II/química , Complexo de Proteína do Fotossistema II/metabolismo , Poliestirenos/metabolismo , Rhodobacter sphaeroides/metabolismo , Solubilidade , Synechocystis/metabolismo , Tilacoides/química , Tilacoides/metabolismo , Tilacoides/ultraestrutura
10.
J Am Chem Soc ; 140(44): 14557-14561, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30347979

RESUMO

Rhomboid proteases form a paradigm for intramembrane proteolysis and have been implicated in several human diseases. However, their study is hampered by difficulties in solubilization and purification. We here report on the use of polymers composed of maleic acid and either diisobutylene or styrene for solubilization of rhomboid proteases in lipid nanodiscs, which proceeds with up to 48% efficiency. We show that the activity of rhomboids in lipid nanodiscs is closer to that in the native membrane than rhomboids in detergent. Moreover, a rhomboid that was proteolytically unstable in detergent turned out to be stable in lipid nanodiscs, underlining the benefit of using these polymer-stabilized nanodiscs. The systems are also compatible with the use of activity-based probes and can be used for small molecule inhibitor screening, allowing several downstream applications.


Assuntos
Alcenos/metabolismo , Lipídeos/química , Maleatos/metabolismo , Nanopartículas/metabolismo , Polímeros/metabolismo , Serina Proteases/metabolismo , Alcenos/química , Humanos , Maleatos/química , Modelos Moleculares , Estrutura Molecular , Nanopartículas/química , Tamanho da Partícula , Polímeros/química , Serina Proteases/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
11.
Biochim Biophys Acta Biomembr ; 1860(2): 257-263, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29056560

RESUMO

Discovering how membrane proteins recognize signals and passage molecules remains challenging. Life depends on compartmentalizing these processes into dynamic lipid bilayers that are technically difficult to work with. Several polymers have proven adept at separating the responsible machines intact for detailed analysis of their structures and interactions. Styrene maleic acid (SMA) co-polymers efficiently solubilize membranes into native nanodiscs and, unlike amphipols and membrane scaffold proteins, require no potentially destabilizing detergents. Here we review progress with the SMA lipid particle (SMALP) system and its impacts including three dimensional structures and biochemical functions of peripheral and transmembrane proteins. Polymers systems are emerging to tackle the remaining challenges for wider use and future applications including in membrane proteomics, structural biology of transient or unstable states, and discovery of ligand and drug-like molecules specific for native lipid-bound states.


Assuntos
Membrana Celular/química , Bicamadas Lipídicas/química , Maleatos/química , Nanoestruturas/química , Poliestirenos/química , Membrana Celular/metabolismo , Bicamadas Lipídicas/metabolismo , Maleatos/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Moleculares , Estrutura Molecular , Poliestirenos/metabolismo , Conformação Proteica , Solubilidade
12.
Biochim Biophys Acta Biomembr ; 1860(4): 809-817, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28865797

RESUMO

New technologies for the purification of stable membrane proteins have emerged in recent years, in particular methods that allow the preparation of membrane proteins with their native lipid environment. Here, we look at the progress achieved with the use of styrene-maleic acid copolymers (SMA) which are able to insert into biological membranes forming nanoparticles containing membrane proteins and lipids. This technology can be applied to membrane proteins from any host source, and, uniquely, allows purification without the protein ever being removed from a lipid bilayer. Not only do these SMA lipid particles (SMALPs) stabilise membrane proteins, allowing structural and functional studies, but they also offer opportunities to understand the local lipid environment of the host membrane. With any new or different method, questions inevitably arise about the integrity of the protein purified: does it retain its activity; its native structure; and ability to perform its function? How do membrane proteins within SMALPS perform in existing assays and lend themselves to analysis by established methods? We outline here recent work on the structure and function of membrane proteins that have been encapsulated like this in a polymer-bound lipid bilayer, and the potential for the future with this approach. This article is part of a Special Issue entitled: Beyond the Structure-Function Horizon of Membrane Proteins edited by Ute Hellmich, Rupak Doshi and Benjamin McIlwain.


Assuntos
Bicamadas Lipídicas/química , Lipídeos de Membrana/química , Proteínas de Membrana/química , Polímeros/química , Bicamadas Lipídicas/metabolismo , Maleatos/química , Maleatos/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Modelos Moleculares , Polímeros/metabolismo , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Estirenos/química , Estirenos/metabolismo
13.
Appl Microbiol Biotechnol ; 102(17): 7377-7388, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29926142

RESUMO

Mesaconate, a branched unsaturated dicarboxylic acid, has drawn great interest because of its versatile applications. In this work, we optimized the fermentation efficiency of Escherichia coli to produce mesaconate from glucose. We first drove the carbon flux to 2-ketoglutarate by overexpressing genes involved in TCA precursor pathway and anaplerotic pathways. Then, to increase the pool of phosphoenolpyruvate (PEP), an upstream precursor for 2-ketoglutarate, the phosphotransferase system (PTS) of E. coli was inactivated by deleting glucose PTS permease and the import of glucose was altered by overexpressing galactose/H+ symporter GalP. Further, production optimization was achieved by deleting a class I fumarase (FumA) to block the hydration of mesaconate. Finally, we overexpressed PEP synthase (PpsA) to increase the availability of phosphoenolpyruvate and accelerate the production of mesaconate. These genetic modifications led to mesaconate production with a titer of 23.1 g L-1 and a yield of 0.46 g g-1 glucose (64% of the theoretical maximum). This work demonstrates the possibility of engineering a highly efficient bacteria strain that converts glucose into mesaconate with promising titer, rate, and yield.


Assuntos
Ciclo do Carbono/genética , Escherichia coli/metabolismo , Fumaratos/metabolismo , Glucose/metabolismo , Microbiologia Industrial , Maleatos/metabolismo , Transporte Biológico , Proteínas de Ligação ao Cálcio/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fermentação , Deleção de Genes , Expressão Gênica , Proteínas de Transporte de Monossacarídeos/genética , Proteínas Periplásmicas de Ligação/genética , Fosfoenolpiruvato/metabolismo , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Piruvato Sintase/genética
14.
Exp Dermatol ; 26(8): 685-690, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27376248

RESUMO

Fumaric acid esters (FAEs) are used as an oral treatment for psoriasis. Dimethylfumarate (DMF) and its metabolite monomethylfumarate (MMF) are regarded as the pharmacologically active moieties. Indoleamine 2,3-dioxygenase (IDO) is the key enzyme for the metabolism of tryptophan. The kynurenine pathway is established as a major regulator of innate and adaptive immunity. Here, we investigated the effect of DMF and MMF on IDO activity and expression in human peripheral blood mononuclear cells (PBMCs). IDO activity was determined by measuring the concentration of kynurenine in the culture medium using a HPLC technique. IDO and kynureninase protein expressions were analysed by Western blot. Our results demonstrated that DMF and MMF dose-dependently reduced the levels of L-kynurenine in PBMCs activated by interferon-γ (IFN-γ). Furthermore, MMF had an inhibitory effect on IDO activity in vitro with an ED50 of 10 µmol/L, a value within the therapeutic concentration range for this molecule. We also observed that IDO and kynureninase expressions were reduced in PBMCs in a dose-dependent manner by DMF and MMF. The results of our study show that DMF and MMF (in therapeutic concentrations) inhibited IDO and kynureninase activity and expression in a NF-κB-dependent manner in PBMCs while also decreasing the level of L-kynurenine in these cells. As we found that FAEs inhibit both IDO expression and enzymatic activity leading to a modulation of tryptophan degradation, we believe this effect may contribute to the clinical efficacy of this drug in psoriasis by downregulating pro-inflammatory mediators generated by the kynurenine pathway.


Assuntos
Fumarato de Dimetilo/metabolismo , Fumaratos/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cinurenina/metabolismo , Leucócitos Mononucleares/enzimologia , Maleatos/metabolismo , Fumarato de Dimetilo/uso terapêutico , Voluntários Saudáveis , Humanos , Hidrolases/metabolismo , Cinurenina/uso terapêutico , Cultura Primária de Células , Psoríase/tratamento farmacológico
15.
Proteins ; 84(8): 1029-42, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27040018

RESUMO

Maleylacetate reductase plays a crucial role in catabolism of resorcinol by catalyzing the NAD(P)H-dependent reduction of maleylacetate, at a carbon-carbon double bond, to 3-oxoadipate. The crystal structure of maleylacetate reductase from Rhizobium sp. strain MTP-10005, GraC, has been elucidated by the X-ray diffraction method at 1.5 Å resolution. GraC is a homodimer, and each subunit consists of two domains: an N-terminal NADH-binding domain adopting an α/ß structure and a C-terminal functional domain adopting an α-helical structure. Such structural features show similarity to those of the two existing families of enzymes in dehydroquinate synthase-like superfamily. However, GraC is distinct in dimer formation and activity expression mechanism from the families of enzymes. Two subunits in GraC have different structures from each other in the present crystal. One subunit has several ligands mimicking NADH and the substrate in the cleft and adopts a closed domain arrangement. In contrast, the other subunit does not contain any ligand causing structural changes and adopts an open domain arrangement. The structure of GraC reveals those of maleylacetate reductase both in the coenzyme, substrate-binding state and in the ligand-free state. The comparison of both subunit structures reveals a conformational change of the Tyr326 loop for interaction with His243 on ligand binding. Structures of related enzymes suggest that His243 is likely a catalytic residue of GraC. Mutational analyses of His243 and Tyr326 support the catalytic roles proposed from structural information. The crystal structure of GraC characterizes the maleylacetate reductase family as a third family in the dehydroquinate synthase-like superfamily. Proteins 2016; 84:1029-1042. © 2016 Wiley Periodicals, Inc.


Assuntos
Adipatos/química , Proteínas de Bactérias/química , Maleatos/química , NAD/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Rhizobium/química , Adipatos/metabolismo , Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Maleatos/metabolismo , Modelos Moleculares , Mutação , NAD/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Multimerização Proteica , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rhizobium/enzimologia , Homologia Estrutural de Proteína
16.
Environ Microbiol ; 18(12): 4920-4930, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27318186

RESUMO

The sensor kinase DcuS of Escherichia coli co-operates under aerobic conditions with the C4 -dicarboxylate transporter DctA to form the DctA/DcuS sensor complex. Under anaerobic conditions C4 -dicarboxylate transport in fumarate respiration is catalyzed by C4 -dicarboxylate/fumarate antiporter DcuB. (i) DcuB interacted with DcuS as demonstrated by a bacterial two-hybrid system (BACTH) and by co-chromatography of the solubilized membrane-proteins (mHPINE assay). (ii) In the DcuB/DcuS complex only DcuS served as the sensor since mutations in the substrate site of DcuS changed substrate specificity of sensing, and substrates maleate or 3-nitropropionate induced DcuS response without affecting the fumarate site of DcuB. (iii) The half-maximal concentration for induction of DcuS by fumarate (1 to 2 mM) and the corresponding Km for transport (50 µM) differ by a factor of 20 to 40. Therefore, the fumarate sites are different in transport and sensing. (iv) Increasing levels of DcuB converted DcuS from the permanent ON (DcuB deficient) state to the fumarate responsive form. Overall, the data show that DcuS and DcuB form a DcuB/DcuS complex representing the C4 -dicarboxylate responsive form, and that the sensory site of the complex is located in DcuS whereas DcuB is required for converting DcuS to the sensory competent state.


Assuntos
Antiporters/metabolismo , Transportadores de Ácidos Dicarboxílicos/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Quinases/metabolismo , Antiporters/genética , Transporte Biológico/fisiologia , Transportadores de Ácidos Dicarboxílicos/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Fumaratos/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Maleatos/metabolismo , Nitrocompostos/metabolismo , Propionatos/metabolismo , Proteínas Quinases/genética
17.
Metab Eng ; 38: 285-292, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27697562

RESUMO

Dicarboxylic acids are attractive biosynthetic targets due to their broad applications and their challenging manufacturing process from fossil fuel feedstock. Mesaconate is a branched, unsaturated dicarboxylic acid that can be used as a co-monomer to produce hydrogels and fire-retardant materials. In this study, we engineered nonphosphorylative metabolism to produce mesaconate from d-xylose and l-arabinose. This nonphosphorylative metabolism is orthogonal to the intrinsic pentose metabolism in Escherichia coli and has fewer enzymatic steps and a higher theoretical yield to TCA cycle intermediates than the pentose phosphate pathway. Here mesaconate production was enabled from the d-xylose pathway and the l-arabinose pathway. To enhance the transportation of d-xylose and l-arabinose, pentose transporters were examined. We identified the pentose/proton symporter, AraE, as the most effective transporter for both d-xylose and l-arabinose in mesaconate production process. Further production optimization was achieved by operon screening and metabolic engineering. These efforts led to the engineered strains that produced 12.5g/l and 13.2g/l mesaconate after 48h from 20g/l of d-xylose and l-arabinose, respectively. Finally, the engineered strain overexpressing both l-arabinose and d-xylose operons produced 14.7g/l mesaconate from a 1:1 d-xylose and l-arabinose mixture with a yield of 85% of the theoretical maximum. (0.87g/g). This work demonstrates an effective system that converts pentoses into a value-added chemical, mesaconate, with promising titer, rate, and yield.


Assuntos
Proteínas de Escherichia coli/genética , Escherichia coli/fisiologia , Fumaratos/metabolismo , Maleatos/metabolismo , Engenharia Metabólica/métodos , Redes e Vias Metabólicas/genética , Pentoses/metabolismo , Arabinose/metabolismo , Vias Biossintéticas/genética , Proteínas de Escherichia coli/metabolismo , Fumaratos/isolamento & purificação , Melhoramento Genético/métodos , Lignina/metabolismo , Maleatos/isolamento & purificação , Fosforilação/genética , Xilose/metabolismo
18.
Mol Pharm ; 13(10): 3439-3448, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27568865

RESUMO

There is a paucity of data describing the impact of salt counterions on the biological performance of inhaled medicines in vivo. The aim of this study was to determine if the coadministration of salt counterions influenced the tissue permeability and airway smooth muscle relaxation potential of salbutamol, formoterol, and salmeterol. The results demonstrated that only salbutamol, when formulated with an excess of the 1-hydroxy-2-naphthoate (1H2NA) counterion, exhibited a superior bronchodilator effect (p < 0.05) compared to salbutamol base. The counterions aspartate, maleate, fumarate, and 1H2NA had no effect on the ability of formoterol or salmeterol to reduce airway resistance in vivo. Studies using guinea pig tracheal sections showed that the salbutamol:1H2NA combination resulted in a significantly faster (p < 0.05) rate of tissue transport compared to salbutamol base. Furthermore, when the relaxant activity of salbutamol was assessed in vitro using electrically stimulated, superfused preparations of guinea pig trachea, the inhibition of contraction by salbutamol in the presence of 1H2NA was greater than with salbutamol base (a total inhibition of 94.13%, p < 0.05). The reason for the modification of salbutamol's behavior upon administration with 1H2NA was assigned to ion-pair formation, which was identified using infrared spectroscopy. Ion-pair formation is known to modify a drug's physicochemical properties, and the data from this study suggested that the choice of counterion in inhaled pharmaceutical salts should be considered carefully as it has the potential to alter drug action in vivo.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/química , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Albuterol/química , Albuterol/farmacologia , Naftóis/química , Traqueia/efeitos dos fármacos , Animais , Ácido Aspártico , Cromatografia Líquida de Alta Pressão , Composição de Medicamentos , Fumaratos/metabolismo , Cobaias , Técnicas In Vitro , Masculino , Maleatos/metabolismo , Traqueia/metabolismo
19.
Mol Microbiol ; 91(5): 1009-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24397579

RESUMO

N-heterocyclic compounds from industrial wastes, including nicotine, are environmental pollutants or toxicants responsible for a variety of health problems. Microbial biodegradation is an attractive strategy for the removal of N-heterocyclic pollutants, during which carbon-nitrogen bonds in N-heterocycles are converted to amide bonds and subsequently severed by amide hydrolases. Previous studies have failed to clarify the molecular mechanism through which amide hydrolases selectively recognize diverse amide substrates and complete the biodenitrogenation process. In this study, structural, computational and enzymatic analyses showed how the N-formylmaleamate deformylase Nfo and the maleamate amidase Ami, two pivotal amide hydrolases in the nicotine catabolic pathway of Pseudomonas putida S16, specifically recognize their respective substrates. In addition, comparison of the α-ß-α groups of amidases, which include Ami, pinpointed several subgroup-characteristic residues differentiating the two classes of amide substrates as containing either carboxylate groups or aromatic rings. Furthermore, this study reveals the molecular mechanism through which the specially tailored active sites of deformylases and amidases selectively recognize their unique substrates. Our work thus provides a thorough elucidation of the molecular mechanism through which amide hydrolases accomplish substrate-specific recognition in the microbial N-heterocycles biodenitrogenation pathway.


Assuntos
Amidas/metabolismo , Hidrolases/química , Hidrolases/metabolismo , Pseudomonas putida/enzimologia , Sequência de Aminoácidos , Aminoácidos/metabolismo , Biodegradação Ambiental , Domínio Catalítico , Cristalografia por Raios X , Compostos Heterocíclicos , Hidrólise , Cinética , Maleatos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Mutação/genética , Relação Estrutura-Atividade , Especificidade por Substrato
20.
Metab Eng ; 30: 190-196, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26070834

RESUMO

Mesaconate is an intermediate in the glutamate degradation pathway of microorganisms such as Clostridium tetanomorphum. However, metabolic engineering to produce mesaconate has not been reported previously. In this work, two enzymes involved in mesaconate production, glutamate mutase and 3-methylaspartate ammonia lyase from C. tetanomorphum, were recombinantly expressed in Escherichia coli. To improve mesaconate production, reactivatase of glutamate mutase was discovered and adenosylcobalamin availability was increased. In addition, glutamate mutase was engineered to improve the in vivo activity. These efforts led to efficient mesaconate production at a titer of 7.81 g/L in shake flask with glutamate feeding. Then a full biosynthetic pathway was constructed to produce mesaconate at a titer of 6.96 g/L directly from glucose. In summary, we have engineered an efficient system in E. coli for the biosynthesis of mesaconate.


Assuntos
Proteínas de Bactérias/biossíntese , Clostridium tetanomorphum/genética , Escherichia coli , Fumaratos/metabolismo , Transferases Intramoleculares/biossíntese , Maleatos/metabolismo , Proteínas de Bactérias/genética , Clostridium tetanomorphum/enzimologia , Escherichia coli/genética , Escherichia coli/metabolismo , Transferases Intramoleculares/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA