Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Bioresour Technol ; 397: 130500, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38423487

RESUMO

This study investigates the behaviors and effects of F-53B, an alternative to perfluorooctane sulfonate on anaerobic ammonium oxidation (anammox) processes. Results showed that the nitrogen removal efficiency (NRE) reached 83.8 % at a F-53B concentration of 0.5 mg·L-1, while NRE decreased to 66.9 % with 5 mg·L-1 of F-53B. The defluorination rates of 17.8 % (0.5 mg·L-1) and 9.3 % (5 mg·L-1) were observed, respectively, suggesting the occurrence of F-53B degradation. The relative abundance of Ca. Kuenenia decreased from 26.1 % to 16.2 % with the F-53B concentration increasing from 0.5 mg·L-1 to 5 mg·L-1. Meanwhile, Denitratisoma was selectively enriched with a relative abundance of 40.7 % at an F-53B concentration of 0.5 mg·L-1. Ca. Kuenenia could reduce reactive oxygen species induced by F-53B to maintain the balance of oxidative stress. This study gains insight into the behaviors and metabolic mechanisms of F-53B in anammox consortia, suggesting the feasibility of anammox processes for industrial wastewater.


Assuntos
Oxidação Anaeróbia da Amônia , Éter , Animais , Éter/metabolismo , Desnitrificação , Peixe-Zebra/metabolismo , Alcanossulfonatos/metabolismo , Nitrogênio/metabolismo , Oxirredução , Reatores Biológicos
2.
Environ Toxicol Chem ; 43(1): 170-181, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37861387

RESUMO

High levels of 6:2 chlorinated polyfluorinated ether sulfonate (F-53B), which is a substitute for perfluorooctane sulfonate (PFOS), are detected in various environmental matrices, wildlife, and humans. Chlorinated polyfluorinated ether sulfonate has received increased attention due to its potential risk to ecosystems. However, its toxicity in the soil organisms remains unclear. In the present study, a comparative investigation was conducted on the toxicities of 6:2 Chlorinated polyfluorinated ether sulfonate (F-53B) and PFOS to the earthworm Eisenia. fetida. F-53B was significantly more acutely toxic to earthworms than PFOS, with median lethal concentrations of 1.43 and 1.83 mmol/kg dry soil (~816 and 984 mg/kg dry soil), respectively. Although both F-53B and PFOS, at 0.4 mmol/kg dry soil (=228 and 215 mg/kg dry soil) caused oxidative stress in earthworms, as evidenced by increased superoxide dismutase, peroxidase, and catalase activities as well as malondialdehyde level, the stress caused by F-53B was higher than that caused by PFOS. In transcriptomic and metabolomic studies, negative effects of PFOS and F-53B were observed on several metabolic processes in earthworms, including protein digestion and amino acid absorption, lipid metabolism, and the immune response. Compared with PFOS, F-53B exhibited a weaker disruption of lipid metabolism, comparable potency for toxicity to the immune response, and a stronger potency in extracellular matrix destruction along with apoptosis and ferroptosis induction. Hence, our data suggest that F-53B is more toxic than PFOS to earthworms. The findings provide some new insights into the potential toxicity of F-53B to soil organisms. Environ Toxicol Chem 2024;43:170-181. © 2023 SETAC.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Oligoquetos , Humanos , Animais , Éter/metabolismo , Ecossistema , Peixe-Zebra/metabolismo , Ácidos Alcanossulfônicos/toxicidade , Ácidos Alcanossulfônicos/metabolismo , Alcanossulfonatos/metabolismo , Alcanossulfonatos/toxicidade , Fluorocarbonos/metabolismo , Solo
3.
Environ Health Perspect ; 131(11): 117011, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37995155

RESUMO

BACKGROUND: Perfluorohexane sulfonate (PFHxS) is a frequently detected per- and polyfluoroalkyl substance in most populations, including in individuals who are pregnant, a period critical for early life development. Despite epidemiological evidence of exposure, developmental toxicity, particularly at realistic human exposures, remains understudied. OBJECTIVES: We evaluated the effect of gestational exposure to human-relevant body burden of PFHxS on fetal and placental development and explored mechanisms of action combining alternative splicing (AS) and gene expression (GE) analyses. METHODS: Pregnant ICR mice were exposed to 0, 0.03, and 0.3µg/kg/day from gestational day 7 to day 17 via oral gavage. Upon euthanasia, PFHxS distribution was measured using liquid chromatography-tandem mass spectrometry. Maternal and fetal phenotypes were recorded, and histopathology was examined for placenta impairment. Multiomics was adopted by combining AS and GE analyses to unveil disruptions in mRNA quality and quantity. The key metabolite transporters were validated by quantitative real-time PCR (qRT-PCR) for quantification and three-dimensional (3D) structural simulation by AlphaFold2. Targeted metabolomics based on liquid chromatography-tandem mass spectrometry was used to detect amino acid and amides levels in the placenta. RESULTS: Pups developmentally exposed to PFHxS exhibited signs of intrauterine growth restriction (IUGR), characterized by smaller fetal weight and body length (p<0.01) compared to control mice. PFHxS concentration in maternal plasma was 5.01±0.54 ng/mL. PFHxS trans-placenta distribution suggested dose-dependent transfer through placental barrier. Histopathology of placenta of exposed dams showed placental dysplasia, manifested with an attenuated labyrinthine layer area and deescalated blood sinus counts and placental vascular development index marker CD34. Combined GE and AS analyses pinpointed differences in genes associated with key biological processes of placental development, proliferation, metabolism, and transport in placenta of exposed dams compared to that of control dams. Further detection of placental key transporter gene expression, protein structure simulation, and amino acid and amide metabolites levels suggested that PFHxS exposure during pregnancy led to impairment of placental amino acid transportation. DISCUSSION: The findings from this study suggest that exposure to human-relevant very-low-dose PFHxS during pregnancy in mice caused IUGR, likely via downregulating of placental amino acid transporters, thereby impairing placental amino acid transportation, resulting in impairment of placental development. Our findings confirm epidemiological findings and call for future attention on the health risk of this persistent yet ubiquitous chemical in the early developmental stage and provide a new approach for understanding gene expression from both quantitative and qualitative omics approaches in toxicological studies. https://doi.org/10.1289/EHP13217.


Assuntos
Fluorocarbonos , Placentação , Humanos , Gravidez , Camundongos , Animais , Feminino , Placenta , Processamento Alternativo , Camundongos Endogâmicos ICR , Fluorocarbonos/toxicidade , Fluorocarbonos/metabolismo , Alcanossulfonatos/metabolismo , Alcanossulfonatos/farmacologia , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Perfilação da Expressão Gênica
4.
J Biol Chem ; 299(8): 105010, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37414148

RESUMO

The obligately anaerobic sulfite-reducing bacterium Bilophila wadsworthia is a common human pathobiont inhabiting the distal intestinal tract. It has a unique ability to utilize a diverse range of food- and host-derived sulfonates to generate sulfite as a terminal electron acceptor (TEA) for anaerobic respiration, converting the sulfonate sulfur to H2S, implicated in inflammatory conditions and colon cancer. The biochemical pathways involved in the metabolism of the C2 sulfonates isethionate and taurine by B. wadsworthia were recently reported. However, its mechanism for metabolizing sulfoacetate, another prevalent C2 sulfonate, remained unknown. Here, we report bioinformatics investigations and in vitro biochemical assays that uncover the molecular basis for the utilization of sulfoacetate as a source of TEA (STEA) for B. wadsworthia, involving conversion to sulfoacetyl-CoA by an ADP-forming sulfoacetate-CoA ligase (SauCD), and stepwise reduction to isethionate by NAD(P)H-dependent enzymes sulfoacetaldehyde dehydrogenase (SauS) and sulfoacetaldehyde reductase (TauF). Isethionate is then cleaved by the O2-sensitive isethionate sulfolyase (IseG), releasing sulfite for dissimilatory reduction to H2S. Sulfoacetate in different environments originates from anthropogenic sources such as detergents, and natural sources such as bacterial metabolism of the highly abundant organosulfonates sulfoquinovose and taurine. Identification of enzymes for anaerobic degradation of this relatively inert and electron-deficient C2 sulfonate provides further insights into sulfur recycling in the anaerobic biosphere, including the human gut microbiome.


Assuntos
Bilophila , Humanos , Alcanossulfonatos/metabolismo , Bilophila/metabolismo , Sulfitos/metabolismo , Enxofre/metabolismo , Taurina/metabolismo , Microbioma Gastrointestinal
5.
Appl Environ Microbiol ; 89(7): e0061723, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37404184

RESUMO

Sulfoquinovose (SQ, 6-deoxy-6-sulfo-glucose) constitutes the polar head group of plant sulfolipids and is one of the most abundantly produced organosulfur compounds in nature. Degradation of SQ by bacterial communities contributes to sulfur recycling in many environments. Bacteria have evolved at least four mechanisms for glycolytic degradation of SQ, termed sulfoglycolysis, producing C3 sulfonate (dihydroxypropanesulfonate and sulfolactate) and C2 sulfonate (isethionate) by-products. These sulfonates are further degraded by other bacteria, leading to the mineralization of the sulfonate sulfur. The C2 sulfonate sulfoacetate is widespread in the environment and is also thought to be a product of sulfoglycolysis, although the mechanistic details are yet unknown. Here, we describe a gene cluster in an Acholeplasma sp., from a metagenome derived from deeply circulating subsurface aquifer fluids (GenBank accession no. QZKD01000037), encoding a variant of the recently discovered sulfoglycolytic transketolase (sulfo-TK) pathway that produces sulfoacetate instead of isethionate as a by-product. We report the biochemical characterization of a coenzyme A (CoA)-acylating sulfoacetaldehyde dehydrogenase (SqwD) and an ADP-forming sulfoacetate-CoA ligase (SqwKL), which collectively catalyze the oxidation of the transketolase product sulfoacetaldehyde into sulfoacetate, coupled with ATP formation. A bioinformatics study revealed the presence of this sulfo-TK variant in phylogenetically diverse bacteria, adding to the variety of mechanisms by which bacteria metabolize this ubiquitous sulfo-sugar. IMPORTANCE Many bacteria utilize environmentally widespread C2 sulfonate sulfoacetate as a sulfur source, and the disease-linked human gut sulfate- and sulfite-reducing bacteria can use it as a terminal electron receptor for anaerobic respiration generating toxic H2S. However, the mechanism of sulfoacetate formation is unknown, although it has been proposed that sulfoacetate originates from bacterial degradation of sulfoquinovose (SQ), the polar head group of sulfolipids present in all green plants. Here, we describe a variant of the recently discovered sulfoglycolytic transketolase (sulfo-TK) pathway. Unlike the regular sulfo-TK pathway that produces isethionate, our biochemical assays with recombinant proteins demonstrated that a CoA-acylating sulfoacetaldehyde dehydrogenase (SqwD) and an ADP-forming sulfoacetate-CoA ligase (SqwKL) in this variant pathway collectively catalyze the oxidation of the transketolase product sulfoacetaldehyde into sulfoacetate, coupled with ATP formation. A bioinformatics study revealed the presence of this sulfo-TK variant in phylogenetically diverse bacteria and interpreted the widespread existence of sulfoacetate.


Assuntos
Bactérias , Transcetolase , Humanos , Bactérias/genética , Bactérias/metabolismo , Alcanossulfonatos/metabolismo , Oxirredutases , Trifosfato de Adenosina , Enxofre/metabolismo , Ligases
6.
Environ Pollut ; 233: 8-15, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29049944

RESUMO

Chlorinated polyfluorinated ether sulfonate (Cl-PFESA) is a novel alternative compound for perfluorooctane sulfonate (PFOS), with its environmental risk not well known. The bioaccumulation and toxic effects of Cl-PFESA in the freshwater alga is crucial for the understanding of its potential hazards to the aquatic environment. Scenedesmus obliquus was exposed to Cl-PFESA at ng L-1 to mg L-1, with the exposure regime beginning at the environmentally relevant level. The total log BAF of Cl-PFESA in S. obliquus was 4.66, higher than the reported log BAF of PFOS in the freshwater plankton (2.2-3.2). Cl-PFESA adsorbed to the cell surface accounted for 33.5-68.3% of the total concentrations. The IC50 of Cl-PFESA to algal growth was estimated to be 40.3 mg L-1. Significant changes in algal growth rate and chlorophyll a/b contents were observed at 11.6 mg L-1 and 13.4 mg L-1 of Cl-PFESA, respectively. The sample cell membrane permeability, measured by the fluorescein diacetate hydrolyzation, was increased by Cl-PFESA at 5.42 mg L-1. The mitochondrial membrane potential, measured by Rh123 staining, was also increased, indicating the hyperpolarization induced by Cl-PFESA. The increasing ROS and MDA contents, along with the enhanced SOD, CAT activity, and GSH contents, suggested that Cl-PFESA caused oxidative damage in the algal cells. It is less possible that current Cl-PFESA pollution in surface water posed obvious toxic effects on the green algae. However, the bioaccumulation of Cl-PFESA in algae would contribute to its biomagnification in the aquatic food chain and its effects on membrane property could potentially increase the accessibility and toxicity of other coexisting pollutants.


Assuntos
Scenedesmus/metabolismo , Poluentes Químicos da Água/metabolismo , Alcanossulfonatos/metabolismo , Ácidos Alcanossulfônicos , Clorofila , Clorofila A , Éter , Éteres/metabolismo , Fluorocarbonos , Cadeia Alimentar , Água Doce , Scenedesmus/efeitos dos fármacos
7.
Salud pública Méx ; 56(4): 333-347, jul.-ago. 2014. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-733312

RESUMO

Objetivo. Corregir la mala clasificación y mejorar la calidad de la información sobre la mortalidad materna en México. Material y métodos. A través de los registros clínicos y autopsias verbales, se estudiaron todas las defunciones certificadas como maternas y una selección de defunciones de mujeres en edad fértil, cuyas causas fueron consideradas como sospechosas de encubrir una muerte materna; todas ocurridas durante 2011 en México. Resultados. La búsqueda intencionada y reclasificación de muertes maternas permitió rescatar más de 100 muertes que no habían sido registradas ni codificadas inicialmente como maternas y se ratificaron o rectificaron las causas anotadas en los certificados de defunción. Este procedimiento también permitió reclasificar como muertes maternas 297 defunciones de la base preliminar del Instituto Nacional de Estadística y Geografía. Conclusiones. La Búsqueda Intencionada y Reclasificación de Muertes Maternas es un procedimiento muy útil para mejorar la calidad de la información sobre la mortalidad materna.


Objective. To correct the misclassification and improve the quality of information on maternal mortality in Mexico. Materials and methods. Using clinical records and verbal autopsies, we studied all deaths certified as maternal deaths as well as a selection of deaths of women of childbearing age whose causes were considered as suspected of hiding a maternal death, all of which occurred during 2011 within Mexico. Results. The deliberate search of maternal deaths and reclassification allowed the rescue of just over 100 deaths that were not originally registered or coded as maternal and confirmed or corrected the causes of death recorded on death certificates as confirmed maternal deaths. This procedure also allowed the reclassification of 297 maternal deaths of women in the groundwork of the National Institute of Statistics and Geography. Conclusions. International Search and Reclassification of Maternal Deaths is a very useful procedure for improving the classification of cases that were not classified as maternal deaths and the effect was greater with the coding of indirect obstetric deaths.


Assuntos
Clorofenóis/metabolismo , Euryarchaeota/metabolismo , Pentaclorofenol/metabolismo , Alcanossulfonatos/metabolismo , Anaerobiose , Bactérias Anaeróbias/metabolismo , Biodegradação Ambiental , Cloretos/metabolismo , Modelos Químicos , Molibdênio
8.
Toxicol Pathol ; 39(2): 325-36, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21270424

RESUMO

The dual peroxisome-proliferator-activated receptor (PPAR) α/γ agonist tesaglitazar has been shown to produce fibrosarcomas in rats. Here, the authors studied morphology, proliferation, differentiation, and inflammation markers in adipose tissue from rats exposed to 1, 3, or 10 µmol/kg tesaglitazar for 2 or 12 weeks, including recovery groups (12 weeks treatment followed by 12 weeks recovery), and 3 or 10 µmol/kg tesaglitazar for 24 weeks. Subcutaneous white and brown fat revealed reversible dose-related histopathological alterations and after 12 and 24 weeks developed areas of thickened skin (fatty lumps). There was a dose-dependent increase in proliferation of interstitial cells in white and brown fat as shown by increased mitotic index in all dose groups after 2 weeks. This was limited to the high dose after 12 and 24 weeks in white fat. Gene expression analyses showed that while tesaglitazar induced differentiation of adipose tissue characterized with a switch in cyclin D1 and D3 mRNA by 12 weeks, longer exposure at high doses reversed this differentiation concurrent with a reappearance of early adipocyte and inflammatory markers. These data suggest that sustained increased turnover of mesenchymal cells in adipose tissues, concomitant with onset of inflammation and fibrosis, drives development of fibrosarcomas in rats treated with tesaglitazar.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Fibrossarcoma/induzido quimicamente , PPAR alfa/agonistas , PPAR gama/agonistas , Adipócitos , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Alcanossulfonatos/sangue , Alcanossulfonatos/metabolismo , Análise de Variância , Animais , Biomarcadores , Proliferação de Células , Fibrossarcoma/patologia , Expressão Gênica , Inflamação/induzido quimicamente , Masculino , Fenilpropionatos/sangue , Fenilpropionatos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
9.
Microbiology (Reading) ; 156(Pt 5): 1556-1564, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20150239

RESUMO

2,3-Dihydroxypropane-1-sulfonate (DHPS) is a widespread intermediate in plant and algal transformations of sulfoquinovose (SQ) from the plant sulfolipid sulfoquinovosyl diacylglycerol. Further, DHPS is recovered quantitatively during bacterial degradation of SQ by Klebsiella sp. strain ABR11. DHPS is also a putative precursor of sulfolactate in e.g. Ruegeria pomeroyi DSS-3. A bioinformatic approach indicated that some 28 organisms with sequenced genomes might degrade DHPS inducibly via sulfolactate, with three different desulfonative enzymes involved in its degradation in different organisms. The hypothesis for Cupriavidus pinatubonensis JMP134 (formerly Ralstonia eutropha) involved a seven-gene cluster (Reut_C6093-C6087) comprising a LacI-type transcriptional regulator, HpsR, a major facilitator superfamily uptake system, HpsU, three NAD(P)(+)-coupled DHPS dehydrogenases, HpsNOP, and (R)-sulfolactate sulfo-lyase (SuyAB) [EC 4.4.1.24]. HpsOP effected a DHPS-racemase activity, and HpsN oxidized (R)-DHPS to (R)-sulfolactate. The hypothesis for Roseovarius nubinhibens ISM was similar, but involved a tripartite ATP-independent transport system for DHPS, HpsKLM, and two different desulfonative enzymes, (S)-cysteate sulfo-lyase [EC 4.4.1.25] and sulfoacetaldehyde acetyltransferase (Xsc) [EC 2.3.3.15]. Representative organisms were found to grow with DHPS and release sulfate. C. pinatubonensis JMP134 was found to express at least one NAD(P)(+)-coupled DHPS dehydrogenase inducibly, and three different peaks of activity were separated by anion-exchange chromatography. Protein bands (SDS-PAGE) were subjected to peptide-mass fingerprinting, which identified the corresponding genes (hpsNOP). Purified HpsN converted DHPS to sulfolactate. Reverse-transcription PCR confirmed that hpsNOUP were transcribed inducibly in strain JMP134, and that hpsKLM and hpsNOP were transcribed in strain ISM. DHPS degradation is widespread and diverse, implying that DHPS is common in marine and terrestrial environments.


Assuntos
Álcool Desidrogenase/isolamento & purificação , Alcanossulfonatos/metabolismo , Proteínas de Bactérias/isolamento & purificação , Cupriavidus necator/metabolismo , Álcool Desidrogenase/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Carbono/metabolismo , Cupriavidus necator/enzimologia , Cupriavidus necator/genética , Eletroforese em Gel de Poliacrilamida , Metabolismo Energético , Genoma Bacteriano , Família Multigênica , Rhodobacteraceae/enzimologia , Rhodobacteraceae/genética , Rhodobacteraceae/metabolismo , Transcrição Gênica
10.
Biochim Biophys Acta ; 1804(1): 97-105, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19770075

RESUMO

Detailed kinetic studies were performed in order to determine the role of the single cysteine residue in the desulfonation reaction catalyzed by SsuD. Mutation of the conserved cysteine at position 54 in SsuD to either serine or alanine had little effect on FMNH(2) binding. The k(cat)/K(m) value for the C54S SsuD variant increased 3-fold, whereas the k(cat)/K(m) value for C54A SsuD decreased 6-fold relative to wild-type SsuD. An initial fast phase was observed in kinetic traces obtained for the oxidation of flavin at 370 nm when FMNH(2) was mixed against C54S SsuD (k(obs), 111 s(-1)) in oxygenated buffer that was 10-fold faster than wild-type SsuD (k(obs), 12.9 s(-1)). However, there was no initial fast phase observed in similar kinetic traces obtained for C54A SsuD. This initial fast phase was previously assigned to the formation of the C4a-(hydro)peroxyflavin in studies with wild-type SsuD. There was no evidence for the formation of the C4a-(hydro)peroxyflavin with either SsuD variant when octanesulfonate was included in rapid reaction kinetic studies, even at low octanesulfonate concentrations. The absence of any C4a-(hydro)peroxyflavin accumulation correlates with the increased catalytic activity of C54S SsuD. These results suggest that the conservative serine substitution is able to effectively take the place of cysteine in catalysis. Conversely, decreased accumulation of the C4a-(hydro)peroxyflavin intermediate with the C54A SsuD variant may be due to decreased activity. The data described suggest that Cys54 in SsuD may be either directly or indirectly involved in stabilizing the C4a-(hydro)peroxyflavin intermediate formed during catalysis through hydrogen bonding interactions.


Assuntos
Cisteína/genética , Proteínas de Escherichia coli/genética , NADH NADPH Oxirredutases/genética , Alcanossulfonatos/metabolismo , Sítios de Ligação/genética , Catálise , Cisteína/metabolismo , Proteínas de Escherichia coli/metabolismo , Flavinas/metabolismo , Cinética , Oxigenases de Função Mista , NADH NADPH Oxirredutases/metabolismo
11.
BMC Genomics ; 9: 483, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18854009

RESUMO

BACKGROUND: Regulation of sulphur metabolism in Corynebacterium glutamicum ATCC 13032 has been studied intensively in the last few years, due to its industrial as well as scientific importance. Previously, the gene cg0156 was shown to belong to the regulon of McbR, a global transcriptional repressor of sulphur metabolism in C. glutamicum. This gene encodes a putative ROK-type regulator, a paralogue of the activator of sulphonate utilisation, SsuR. Therefore, it is an interesting candidate for study to further the understanding of the regulation of sulphur metabolism in C. glutamicum. RESULTS: Deletion of cg0156, now designated cysR, results in the inability of the mutant to utilise sulphate and aliphatic sulphonates. DNA microarray hybridisations revealed 49 genes with significantly increased and 48 with decreased transcript levels in presence of the native CysR compared to a cysR deletion mutant. Among the genes positively controlled by CysR were the gene cluster involved in sulphate reduction, fpr2 cysIXHDNYZ, and ssuR. Gel retardation experiments demonstrated that binding of CysR to DNA depends in vitro on the presence of either O-acetyl-L-serine or O-acetyl-L-homoserine. Mapping of the transcription start points of five transcription units helped to identify a 10 bp inverted repeat as the possible CysR binding site. Subsequent in vivo tests proved this motif to be necessary for CysR-dependent transcriptional regulation. CONCLUSION: CysR acts as the functional analogue of the unrelated LysR-type regulator CysB from Escherichia coli, controlling sulphide production in response to acceptor availability. In both bacteria, gene duplication events seem to have taken place which resulted in the evolution of dedicated regulators for the control of sulphonate utilisation. The striking convergent evolution of network topology indicates the strong selective pressure to control the metabolism of the essential but often toxic sulphur-containing (bio-)molecules.


Assuntos
Proteínas de Bactérias/genética , Corynebacterium glutamicum/genética , Regulon , Proteínas Repressoras/genética , Sulfatos/metabolismo , Alcanossulfonatos/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Corynebacterium glutamicum/metabolismo , Cisteína/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Família Multigênica , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos , Regiões Promotoras Genéticas , RNA Bacteriano/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sítio de Iniciação de Transcrição , Transcrição Gênica
12.
Biophys J ; 95(6): 2924-38, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18567630

RESUMO

We present a quantitative analysis of conformational changes of the nucleotide-binding subunits, MalK(2), of the maltose ATP-binding cassette importer MalFGK(2) during the transport cycle. Distance changes occurring between selected residues were monitored in the full transporter by site-directed spin-labeling electron paramagnetic resonance spectroscopy and site-directed chemical cross-linking. We considered S83C and A85C from the conserved Q-loop and V117C located on the outer surface of MalK. Additionally, two native cysteines (C350, C360) were included in the study. On ATP binding, small rearrangements between the native sites, and no distance changes between positions 117 were detected. In contrast, positions 85 come closer together in the ATP-bound state and in the vanadate-trapped intermediate and move back toward the apo-state after ATP hydrolysis. The distance between positions 83 is shown to slightly decrease on ATP binding, and to further decrease after ATP hydrolysis. Results from cross-linking experiments are in agreement with these findings. The data are compared with in silico spin-labeled x-ray structures from both isolated MalK(2) and the MalFGK(2)-E complex. Our results are consistent with a slightly modified "tweezers-like" model of closure and reopening of MalK(2) during the catalytic cycle, and show an unforeseen potential interaction between MalK and the transmembrane subunit MalG.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/metabolismo , Nucleotídeos/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Alcanossulfonatos/metabolismo , Fenômenos Biomecânicos , Catálise , Biologia Computacional , Reagentes de Ligações Cruzadas/metabolismo , Cristalografia por Raios X , Cisteína , Dimerização , Espectroscopia de Ressonância de Spin Eletrônica , Proteínas de Transporte de Monossacarídeos/genética , Mutação , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Solubilidade , Marcadores de Spin
13.
J Bacteriol ; 189(5): 1675-88, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16997956

RESUMO

Two genes encoding transcriptional regulators involved in sulfur assimilation pathways in Burkholderia cenocepacia strain 715j have been identified and characterized functionally. Knockout mutations in each of the B. cenocepacia genes were constructed and introduced into the genome of 715j by allelic replacement. Studies on the utilization of various sulfur sources by 715j and the obtained mutants demonstrated that one of the B. cenocepacia regulators, designated CysB, is preferentially involved in the control of sulfate transport and reduction, while the other, designated SsuR, is required for aliphatic sulfonate utilization. Using transcriptional promoter-lacZ fusions and DNA-binding experiments, we identified several target promoters for positive control by CysB and/or SsuR--sbpp (preceding the sbp cysT cysW cysA ssuR cluster), cysIp (preceding the cysI cysD1 cysN cysH cysG cluster), cysD2p (preceding a separate cluster, cysD2 cysNC), and ssuDp (located upstream of the ssuDCB operon)--and we demonstrated overlapping functions of CysB and SsuR at particular promoters. We also demonstrated that the cysB gene is negatively controlled by both CysB and SsuR but the ssuR gene itself is not significantly regulated as a separate transcription unit. The function of B. cenocepacia CysB (in vivo and in vitro) appeared to be independent of the presence of acetylserine, the indispensable coinducer of the CysB regulators of Escherichia coli and Salmonella. The phylogenetic relationships among members of the "CysB family" in the gamma and beta subphyla are presented.


Assuntos
Proteínas de Bactérias/fisiologia , Burkholderia/metabolismo , Regulação Bacteriana da Expressão Gênica , Enxofre/metabolismo , Fatores de Transcrição/fisiologia , Alcanossulfonatos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Burkholderia/genética , Clonagem Molecular , DNA/metabolismo , Genoma Bacteriano , Dados de Sequência Molecular , Fenótipo , Filogenia , Regiões Promotoras Genéticas , Serina/análogos & derivados , Serina/farmacologia , Sulfatos/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-16511173

RESUMO

The alkanesulfonate FMN reductase (SsuE) from Escherichia coli catalyzes the reduction of FMN by NADPH to provide reduced flavin for the monooxygenase (SsuD) enzyme. The vapor-diffusion technique yielded single crystals that grow as hexagonal rods and diffract to 2.9 A resolution using synchrotron X-ray radiation. The protein crystallizes in the primitive hexagonal space group P622. The SsuE protein lacks any cysteine or methionine residues owing to the role of the SsuE enzyme in the acquisition of sulfur during sulfate starvation. Therefore, substitution of two leucine residues (Leu114 and Leu165) to methionine was performed to obtain selenomethionine-containing SsuE for MAD phasing. The selenomethionine derivative of SsuE has been expressed and purified and crystals of the protein have been obtained with and without bound FMN. These preliminary studies should lead to the structure solution of SsuE. It is anticipated that this new protein structure will provide detailed structural information on specific active-site regions of the protein and insight into the mechanism of flavin reduction and transfer of reduced flavin.


Assuntos
Alcanossulfonatos/metabolismo , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , FMN Redutase/química , Cristalização , Cristalografia por Raios X
15.
Int J Biol Macromol ; 34(1-2): 49-54, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15178009

RESUMO

Dynamic light scattering (DLS) measurements were performed to study the binding of anionic surfactant alpha olefin sulfonate (AOS) to gelatin chains at various NaCl concentrations at 30 degrees C in aqueous sodium phosphate buffer (pH = 6.8) solutions. The surfactant concentration was varied from 0 to 80 mM and the NaCl concentrations chosen were 0.025, 0.05, and 0.1 M. AOS exhibited electrostatic binding to the positively charged sites of the polypeptide chain resulting in considerable reduction in its hydrodynamic radius up to critical micellar concentration (cmc = 8 mM for no salt, 0.01 and 0.025 M, and 5 mM for 0.05 M and 2 mM for 0.1 M solutions). The correlation function revealed the presence of two types of structures above cmc; namely the micelles of AOS and gelatin-AOS micelle complexes. The micellar radii (Rm), the effective gelatin-surfactant complex radii (Rc), have been determined as a function of salt concentration. No critical aggregation concentration (cac) was observed. The inter-gelatin-surfactant complex (kD1) and inter-micellar interactions (kD2), were determined by fitting the concentration dependence of Rm and Rc to a virial expansion in reduced concentration (c - cmc), which are compared. While kD1 showed strong ionic strength dependence, kD2 remained invariant of the same. The protein to surfactant binding ratio was found to be smaller than normal. Results have been discussed within the framework of the necklace-bead model of polymer-surfactant interactions.


Assuntos
Alcanossulfonatos/química , Gelatina/metabolismo , Tensoativos/química , Alcanossulfonatos/metabolismo , Alcenos/química , Gelatina/química , Interações Hidrofóbicas e Hidrofílicas , Luz , Modelos Químicos , Nefelometria e Turbidimetria/métodos , Concentração Osmolar , Espalhamento de Radiação , Eletricidade Estática , Tensoativos/metabolismo , Termodinâmica
16.
Arch Microbiol ; 176(6): 406-14, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11734883

RESUMO

Novel isolates of Achromobacter xylosoxidans, Tsukamurella wratislaviensis and a Rhodococcus sp. are described. These grew with short-chain alkanesulfonates as their sole source of carbon and energy. T. wratislaviensis strain SB2 grew well with C(3)-C(6) linear alkanesulfonates, isethionate and taurine, Rhodococcus sp. strain CB1 used C(3)-C(10) linear alkanesulfonates, taurine and cysteate, but neither strain grew with ethanesulfonate. In contrast, A. xylosoxidans strain AE4 grew well with ethanesulfonate, making it the first bacterium to be described which can grow with this compound. It also grew with unsubstituted C(3)-C(5) alkanesulfonates and isethionate. Hydrolysis was excluded as a mechanism for alkanesulfonate metabolism in these strains; and evidence is given for a diversity of uptake and desulfonatase systems. We provide evidence for an initial monooxygenase-dependent desulfonation in the metabolism of ethanesulfonate and propanesulfonate by A. xylosoxidans strain AE4.


Assuntos
Actinomycetales/metabolismo , Alcaligenes/enzimologia , Alcanossulfonatos/metabolismo , Oxigenases/metabolismo , Rhodococcus/metabolismo , Actinomycetales/crescimento & desenvolvimento , Alcaligenes/crescimento & desenvolvimento , Ácidos Alcanossulfônicos/metabolismo , Oxirredução , Rhodococcus/crescimento & desenvolvimento , Especificidade da Espécie , Sulfitos/metabolismo
17.
FEMS Microbiol Lett ; 205(2): 271-5, 2001 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11750815

RESUMO

Twenty-three Escherichia coli strains were tested for their ability to use taurine, methanesulfonate, L-cysteate and other alkanesulfonates as sole sulfur sources for growth. One strain was unable to use any of the alkanesulfonates offered as sole sulfur sources for growth but grew with sulfate. Seven strains (class I) used alkanesulfonates for this purpose, but not methanesulfonate or L-cysteate. A further seven strains (class II) grew with all compounds tested, except with L-cysteate, and eight strains (class III) utilized all compounds tested as sulfur sources. Sulfur assimilation from methanesulfonate and L-cysteate was absolutely dependent on the ssuEADCB operon that encodes an alkanesulfonate uptake system (SsuABC) and a two-component monooxygenase (SsuDE) involved in the release of sulfite from alkanesulfonates. Long-term exposure of class I strains to methanesulfonate and of class II strains to L-cysteate selected for derivatives that utilized these two sulfur sources as efficiently as sulfate. The nucleotide sequence of the ssuEADCB operon in the methanesulfonate- and L-cysteate-utilizing derivative EC1250Me+ was identical to that in the class I wild-type EC1250. Gain of the ability to utilize methanesulfonate and L-cysteate as sulfur sources thus appears to result from increased expression of ssu genes rather than from a change in the quality of one or several of the Ssu proteins.


Assuntos
Ácido Cisteico/metabolismo , Escherichia coli/metabolismo , Mesilatos/metabolismo , Alcanossulfonatos/metabolismo , Sequência de Bases , Meios de Cultura , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli , Humanos , Oxigenases de Função Mista , Mutação , NADH NADPH Oxirredutases/genética , Óperon , Análise de Sequência de DNA , Enxofre/metabolismo
18.
Arch Microbiol ; 176(1-2): 1-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11479697

RESUMO

In the absence of sulfate and cysteine, Escherichia coli can use aliphatic sulfonates as a source of sulfur for growth. Starvation for sulfate leads to the expression of the tauABCD and ssuEADCB genes. Each of these gene clusters encodes an ABC-type transport system required for uptake of aliphatic sulfonates and a desulfonation enzyme. The TauD protein is an alpha-ketoglutarate-dependent dioxygenase that preferentially liberates sulfite from taurine (2-aminoethanesulfonic acid). SsuD is a monooxygenase that catalyzes the oxygenolytic desulfonation of a range of aliphatic sulfonates other than taurine. Its cosubstrate is FMNH2, which is provided by SsuE, an NAD(P)H-dependent FMN reductase. In contrast to many other bacteria, E. coli is unable to grow with arylsulfonates or with sulfate esters as sulfur source. The tau and ssu systems thus provide all genes for the utilization of known organosulfur sources by this organism, except the as yet unidentified gene(s) that enable some E. coli strains to grow with methanesulfonate or cysteate as a sulfur source. Expression of the tau and ssu genes requires the LysR-type transcriptional regulatory proteins CysB and Cbl. Synthesis of Cbl itself is under control of the CysB protein, and the CysB protein may therefore be regarded as the master regulator for sulfur assimilation in E. coli, while the Cbl protein functions as an accessory element specific for utilization of sulfur from organosulfur sources.


Assuntos
Alcanossulfonatos/metabolismo , Escherichia coli/metabolismo , Sulfatos/metabolismo , Enxofre/metabolismo , Alcanossulfonatos/química , Transporte Biológico , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Oxigenases/química , Oxigenases/metabolismo , Regulon/genética , Sulfatases/química , Sulfatases/metabolismo , Sulfatos/química , Enxofre/química
19.
Toxicol Sci ; 55(1): 36-43, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10788557

RESUMO

Chronic administration of alachlor has been shown to produce neoplastic responses in the nasal turbinate mucosa, glandular stomach mucosa, and thyroid follicular epithelium of rats. Subsequent studies have shown that specific metabolic activation of alachlor is required for nasal tumor formation, and that non-genotoxic, threshold-sensitive processes produce all three tumors. The herbicide alachlor is degraded in the soil by microbial action to the tertiary ethane sulfonate metabolite (ESA). The acute and subchronic toxicity of ESA is very low, and the metabolite did not produce developmental toxicity or genotoxicity. The studies described here were conducted to determine whether ESA shares a common mechanism of oncogenicity with alachlor in rats. Specifically, we studied ESA's pharmacokinetics and ability to produce changes that are causally associated with the oncogenicity of alachlor. These studies demonstrated that ESA was poorly absorbed and underwent minor metabolism, which contrasted with the significant absorption and substantial metabolism observed with alachlor. ESA was also excreted more quickly than alachlor and showed no evidence of accumulation in the nasal turbinates, a site of oncogenicity for alachlor in the rat. In addition, ESA did not elicit the characteristic preneoplastic changes observed in the development of alachlor-induced nasal, stomach, and thyroid tumors. The results of these studies support the conclusion that ESA does not share a common oncogenic mechanism with alachlor and would not be expected to produce the same oncogenic responses observed following chronic alachlor exposure in rats.


Assuntos
Acetamidas/metabolismo , Acetamidas/toxicidade , Alcanossulfonatos/toxicidade , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Herbicidas/metabolismo , Herbicidas/toxicidade , Acetamidas/farmacocinética , Alcanossulfonatos/metabolismo , Alcanossulfonatos/farmacocinética , Animais , Autorradiografia , Carcinógenos/farmacocinética , Divisão Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Fezes/química , Feminino , Mucosa Gástrica/patologia , Herbicidas/farmacocinética , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Mucosa Nasal/patologia , Tamanho do Órgão/efeitos dos fármacos , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/patologia , Ratos , Ratos Long-Evans , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/patologia , Distribuição Tecidual
20.
J Bacteriol ; 182(10): 2687-95, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10781534

RESUMO

The Escherichia coli tauABCD and ssuEADCB gene clusters are required for the utilization of taurine and alkanesulfonates as sulfur sources and are expressed only under conditions of sulfate or cysteine starvation. tauD and ssuD encode an alpha-ketoglutarate-dependent taurine dioxygenase and a reduced flavin mononucleotide-dependent alkanesulfonate monooxygenase, respectively. These enzymes are responsible for the desulfonation of taurine and alkanesulfonates. The amino acid sequences of SsuABC and TauABC exhibit similarity to those of components of the ATP-binding cassette transporter superfamily, suggesting that two uptake systems for alkanesulfonates are present in E. coli. Chromosomally located in-frame deletions of the tauABC and ssuABC genes were constructed in E. coli strain EC1250, and the growth properties of the mutants were studied to investigate the requirement for the TauABC and SsuABC proteins for growth on alkanesulfonates as sulfur sources. Complementation analysis of in-frame deletion mutants confirmed that the growth phenotypes obtained were the result of the in-frame deletions constructed. The range of substrates transported by these two uptake systems was largely reflected in the substrate specificities of the TauD and SsuD desulfonation systems. However, certain known substrates of TauD were transported exclusively by the SsuABC system. Mutants in which only formation of hybrid transporters was possible were unable to grow with sulfonates, indicating that the individual components of the two transport systems were not functionally exchangeable. The TauABCD and SsuEADCB systems involved in alkanesulfonate uptake and desulfonation thus are complementary to each other at the levels of both transport and desulfonation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Alcanossulfonatos/metabolismo , Escherichia coli/metabolismo , Oxigenases/metabolismo , Taurina/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transporte Biológico , Escherichia coli/genética , Proteínas de Escherichia coli , FMN Redutase , Deleção de Genes , Teste de Complementação Genética , Oxigenases de Função Mista , Mutagênese , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , Óperon , Oxigenases/genética , Enxofre/metabolismo , Taurina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA