Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biotechnol Bioeng ; 119(1): 226-235, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34687217

RESUMO

Solventogenesis and sporulation of clostridia are the main responsive adaptations to the acidic environment during acetone-butanol-ethanol (ABE) fermentation. It was hypothesized that five orphan histidine kinases (HKs) including Cac3319, Cac0323, Cac0903, Cac2730, and Cac0437 determined the cell fates between sporulation and solventogenesis. In this study, the comparative genomic analysis revealed that a mutation in cac0437 appeared to contribute to the nonsporulating feature of ATCC 55025. Hence, the individual and interactive roles of five HKs in regulating cell growth, metabolism, and sporulation were investigated. The fermentation results of mutants with different HK expression levels suggested that cac3319 and cac0437 played critical roles in regulating sporulation and acids and butanol biosynthesis. Morphological analysis revealed that cac3319 knockout abolished sporulation (Stage 0) whereas cac3319 overexpression promoted spore development (Stage VII), and cac0437 knockout initiated but blocked sporulation before Stage II, indicating the progression of sporulation was altered through engineering HKs. By combinatorial HKs knockout, the interactive effects between two different HKs were investigated. This study elucidated the regulatory roles of HKs in clostridial differentiation and demonstrated that HK engineering can be effectively used to control sporulation and enhance butanol biosynthesis.


Assuntos
Proteínas de Bactérias , Butanóis/metabolismo , Clostridium acetobutylicum , Histidina Quinase , Esporos Bacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética , Clostridium acetobutylicum/metabolismo , Clostridium acetobutylicum/fisiologia , Fermentação , Histidina Quinase/genética , Histidina Quinase/metabolismo , Engenharia Metabólica
2.
J Microbiol Biotechnol ; 31(10): 1393-1400, 2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34584034

RESUMO

Acetone-butanol-ethanol (ABE) fermentation by the anaerobic bacterium Clostridium acetobutylicum has been considered a promising process of industrial biofuel production. Phosphotransbutyrylase (phosphate butyryltransferase, PTB) plays a crucial role in butyrate metabolism by catalyzing the reversible conversion of butyryl-CoA into butyryl phosphate. Here, we report the crystal structure of PTB from the Clostridial host for ABE fermentation, C. acetobutylicum, (CaPTB) at a 2.9 Å resolution. The overall structure of the CaPTB monomer is quite similar to those of other acyltransferases, with some regional structural differences. The monomeric structure of CaPTB consists of two distinct domains, the N- and C-terminal domains. The active site cleft was formed at the interface between the two domains. Interestingly, the crystal structure of CaPTB contained eight molecules per asymmetric unit, forming an octamer, and the size-exclusion chromatography experiment also suggested that the enzyme exists as an octamer in solution. The structural analysis of CaPTB identifies the substrate binding mode of the enzyme and comparisons with other acyltransferase structures lead us to speculate that the enzyme undergoes a conformational change upon binding of its substrate.


Assuntos
Proteínas de Bactérias/química , Clostridium acetobutylicum/enzimologia , Fosfato Acetiltransferase/química , Acetona/metabolismo , Acil Coenzima A , Sequência de Aminoácidos , Butanóis/metabolismo , Domínio Catalítico , Etanol/metabolismo , Fermentação , Estrutura Quaternária de Proteína
3.
Nucleic Acids Res ; 49(19): e113, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34417598

RESUMO

DNA methylation is widespread amongst eukaryotes and prokaryotes to modulate gene expression and confer viral resistance. 5-Methylcytosine (m5C) methylation has been described in genomes of a large fraction of bacterial species as part of restriction-modification systems, each composed of a methyltransferase and cognate restriction enzyme. Methylases are site-specific and target sequences vary across organisms. High-throughput methods, such as bisulfite-sequencing can identify m5C at base resolution but require specialized library preparations and single molecule, real-time (SMRT) sequencing usually misses m5C. Here, we present a new method called RIMS-seq (rapid identification of methylase specificity) to simultaneously sequence bacterial genomes and determine m5C methylase specificities using a simple experimental protocol that closely resembles the DNA-seq protocol for Illumina. Importantly, the resulting sequencing quality is identical to DNA-seq, enabling RIMS-seq to substitute standard sequencing of bacterial genomes. Applied to bacteria and synthetic mixed communities, RIMS-seq reveals new methylase specificities, supporting routine study of m5C methylation while sequencing new genomes.


Assuntos
5-Metilcitosina/metabolismo , Metilases de Modificação do DNA/metabolismo , Enzimas de Restrição do DNA/metabolismo , Escherichia coli K12/genética , Genoma Bacteriano , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Acinetobacter calcoaceticus/enzimologia , Acinetobacter calcoaceticus/genética , Aeromonas hydrophila/enzimologia , Aeromonas hydrophila/genética , Bacillus amyloliquefaciens/enzimologia , Bacillus amyloliquefaciens/genética , Sequência de Bases , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas de Restrição do DNA/genética , Escherichia coli K12/enzimologia , Regulação Bacteriana da Expressão Gênica , Haemophilus/enzimologia , Haemophilus/genética , Haemophilus influenzae/enzimologia , Haemophilus influenzae/genética , Humanos , Microbiota/genética , Análise de Sequência de DNA , Pele/microbiologia
4.
J Am Chem Soc ; 143(1): 335-348, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33372786

RESUMO

Catalysis by canonical radical S-adenosyl-l-methionine (SAM) enzymes involves electron transfer (ET) from [4Fe-4S]+ to SAM, generating an R3S0 radical that undergoes regioselective homolytic reductive cleavage of the S-C5' bond to generate the 5'-dAdo· radical. However, cryogenic photoinduced S-C bond cleavage has regioselectively yielded either 5'-dAdo· or ·CH3, and indeed, each of the three SAM S-C bonds can be regioselectively cleaved in an RS enzyme. This diversity highlights a longstanding central question: what controls regioselective homolytic S-C bond cleavage upon SAM reduction? We here provide an unexpected answer, founded on our observation that photoinduced S-C bond cleavage in multiple canonical RS enzymes reveals two enzyme classes: in one, photolysis forms 5'-dAdo·, and in another it forms ·CH3. The identity of the cleaved S-C bond correlates with SAM ribose conformation but not with positioning and orientation of the sulfonium center relative to the [4Fe-4S] cluster. We have recognized the reduced-SAM R3S0 radical is a (2E) state with its antibonding unpaired electron in an orbital doublet, which renders R3S0 Jahn-Teller (JT)-active and therefore subject to vibronically induced distortion. Active-site forces induce a JT distortion that localizes the odd electron in a single priority S-C antibond, which undergoes regioselective cleavage. In photolytic cleavage those forces act through control of the ribose conformation and are transmitted to the sulfur via the S-C5' bond, but during catalysis thermally induced conformational changes that enable ET from a cluster iron generate dominant additional forces that specifically select S-C5' for cleavage. This motion also can explain how 5'-dAdo· subsequently forms the organometallic intermediate Ω.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo Enxofre/química , S-Adenosilmetionina/química , Proteínas de Bactérias/química , Proteínas de Bactérias/efeitos da radiação , Biocatálise , Domínio Catalítico , Clostridium acetobutylicum/enzimologia , Teoria da Densidade Funcional , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/efeitos da radiação , Luz , Modelos Químicos , Estrutura Molecular , Oxirredução/efeitos da radiação , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/efeitos da radiação , Fotólise , S-Adenosilmetionina/efeitos da radiação , Thermotoga maritima/enzimologia
5.
J Ind Microbiol Biotechnol ; 47(8): 609-620, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32851482

RESUMO

This study addressed the functionality of genetic circuits carrying natural regulatory elements of Clostridium acetobutylicum ATCC 824 in the presence of the respective inducer molecules. Specifically, promoters and their regulators involved in diverse carbon source utilization were characterized using mCherryOpt or beta-galactosidase as a reporter. Consequently, most of the genetic circuits tested in this study were functional in Clostridium acetobutylicum ATCC 824 in the presence of an inducer, leading to the expression of reporter proteins. These genetic sensor-regulators were found to be transferable to another Clostridium species, such as Clostridium beijerinckii NCIMB 8052. The gradual expression of reporter protein was observed as a function of the carbohydrates of interest. A xylose-inducible promoter allows a titratable and robust expression of a reporter protein with stringency and efficacy. This xylose-inducible circuit was seen to enable induction of the expression of reporter proteins in the presence of actual sugar mixtures incorporated in woody hydrolysate wherein glucose and xylose are present as predominant carbon sources.


Assuntos
Clostridium acetobutylicum/genética , Regiões Promotoras Genéticas , beta-Galactosidase/genética , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/metabolismo , Clostridium beijerinckii/genética , Clostridium beijerinckii/metabolismo , Fermentação , Genes Reguladores , Genes Reporter , Glucose/metabolismo , Plasmídeos , Transformação Bacteriana , Xilose/metabolismo , beta-Galactosidase/metabolismo
6.
BMC Res Notes ; 13(1): 281, 2020 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-32522254

RESUMO

OBJECTIVE: We previously described the structure and activity of a glycoside hydrolase family 30 subfamily 8 (GH30-8) endoxylanase, CaXyn30A, from Clostridium acetobutylicum which exhibited novel glucuronic acid (GA)-independent activity. Immediately downstream from CaXyn30A is encoded another GH30-8 enzyme, CaXyn30B. While CaXyn30A deviated substantially in the highly conserved ß7-α7 and ß8-α8 loop regions of the catalytic cleft which are responsible for GA-dependence, CaXyn30B maintains these conserved subfamily 8 amino acid residues thus predicting canonical GA-dependent activity. In this report, we show that CaXyn30B functions as a canonical GA-dependent GH30-8 endoxylanase in contrast to its GA-independent neighbor, CaXyn30A. RESULTS: A clone expressing the catalytic domain of CaXyn30B (CaXyn30B-CD) exhibited GA-dependent endoxylanase activity. Digestion of glucuronoxylan generated a ladder of aldouronate limit products as anticipated for canonical GA-dependent GH30-8 enzymes. Unlike the previously described CaXyn30A-CD, CaXyn30B-CD showed no activity on arabinoxylan or the generation of appreciable neutral oligosaccharides from glucuronoxylan substrates. These results are consistent with amino acid sequence comparisons of the catalytic cleft and phylogenetic analysis.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridium acetobutylicum/enzimologia , Endo-1,4-beta-Xilanases/metabolismo , Ácido Glucurônico/metabolismo , Proteínas de Bactérias/química , Endo-1,4-beta-Xilanases/química
7.
Appl Environ Microbiol ; 86(13)2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32385078

RESUMO

Although Clostridium acetobutylicum is the model organism for the study of acetone-butanol-ethanol (ABE) fermentation, its characterization has long been impeded by the lack of efficient genome editing tools. In particular, the contribution of alcohol dehydrogenases to solventogenesis in this bacterium has mostly been studied with the generation of single-gene deletion strains. In this study, the three butanol dehydrogenase-encoding genes located on the chromosome of the DSM 792 reference strain were deleted iteratively by using a recently developed CRISPR-Cas9 tool improved by using an anti-CRISPR protein-encoding gene, acrIIA4 Although the literature has previously shown that inactivation of either bdhA, bdhB, or bdhC had only moderate effects on the strain, this study shows that clean deletion of both bdhA and bdhB strongly impaired solvent production and that a triple mutant ΔbdhA ΔbdhB ΔbdhC was even more affected. Complementation experiments confirmed the key role of these enzymes and the capacity of each bdh copy to fully restore efficient ABE fermentation in the triple deletion strain.IMPORTANCE An efficient CRISPR-Cas9 editing tool based on a previous two-plasmid system was developed for Clostridium acetobutylicum and used to investigate the contribution of chromosomal butanol dehydrogenase genes during solventogenesis. Thanks to the control of cas9 expression by inducible promoters and of Cas9-guide RNA (gRNA) complex activity by an anti-CRISPR protein, this genetic tool allows relatively fast, precise, markerless, and iterative modifications in the genome of this bacterium and potentially of other bacterial species. As an example, scarless mutants in which up to three genes coding for alcohol dehydrogenases are inactivated were then constructed and characterized through fermentation assays. The results obtained show that in C. acetobutylicum, other enzymes than the well-known AdhE1 are crucial for the synthesis of alcohol and, more globally, to perform efficient solventogenesis.


Assuntos
Oxirredutases do Álcool/genética , Proteínas de Bactérias/genética , Sistemas CRISPR-Cas/genética , Clostridium acetobutylicum/genética , Oxirredutases do Álcool/metabolismo , Proteínas de Bactérias/metabolismo , Clostridium acetobutylicum/enzimologia , Edição de Genes
8.
ACS Synth Biol ; 9(3): 468-474, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32149502

RESUMO

Climate change necessitates the development of CO2 neutral or negative routes to chemicals currently produced from fossil carbon. In this paper we demonstrate a pathway from the renewable resource glucose to next generation biofuel isopentanol by pairing the isovaleryl-CoA biosynthesis pathway from Myxococcus xanthus and a butyryl-CoA reductase from Clostridium acetobutylicum. The best plasmid and Escherichia coli strain combination makes 80.50 ± 8.08 (SD) mg/L of isopentanol after 36 h under microaerobic conditions with an oleyl alcohol overlay. In addition, the system also shows a strong preference for isopentanol production over prenol in microaerobic conditions. Finally, the pathway requires zero adenosine triphosphate and can be paired theoretically with nonoxidative glycolysis, the combination being redox balanced from glucose thus avoiding unnecessary carbon loss as CO2. These pathway properties make the isovaleryl-CoA pathway an attractive isopentanol production route for further optimization.


Assuntos
Trifosfato de Adenosina/metabolismo , Biocombustíveis , Carbono/metabolismo , Myxococcus xanthus/metabolismo , Pentanóis/metabolismo , Biologia Sintética/métodos , Acil Coenzima A/metabolismo , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética , Escherichia coli/genética , Hidroliases/genética , Hidroliases/metabolismo , Modelos Biológicos , Myxococcus xanthus/genética , Plasmídeos/genética
9.
J Ind Microbiol Biotechnol ; 47(2): 169-181, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31807968

RESUMO

Gene CA_C0816 codes for a serine hydrolase protein from Clostridium acetobutylicum (ATCC 824) a member of hormone-sensitive lipase of lipolytic family IV. This gene was overexpressed in E. coli strain BL21and purified using Ni2+-NTA affinity chromatography. Size exclusion chromatography revealed that the protein is a dimer in solution. Optimum pH and temperature for recombinant Clostridium acetobutylicum esterase (Ca-Est) were found to be 7.0 and 60 °C, respectively. This enzyme exhibited high preference for p-nitrophenyl butyrate. KM and kcat/KM of the enzyme were 24.90 µM and 25.13 s-1 µM-1, respectively. Sequence analysis of Ca-Est predicts the presence of catalytic amino acids Ser 89, His 224, and Glu 196, presence of novel GYSMG conserved sequence (instead of GDSAG and GTSAG motif), and undescribed variation of HGSG motif. Site-directed mutagenesis confirmed that Ser 89 and His 224 play a major role in catalysis. This study reports that Ca-Est is hormone-sensitive lipase with novel GYSMG pentapeptide motif at a catalytic domain.


Assuntos
Domínio Catalítico , Clostridium acetobutylicum/enzimologia , Esterases/metabolismo , Sequência de Aminoácidos , Biocatálise , Clostridium acetobutylicum/genética , Esterases/química , Esterases/genética , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Análise de Sequência de Proteína , Temperatura
10.
Photosynth Res ; 143(2): 155-163, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31673863

RESUMO

Photosystem I complexes from the menB deletion mutant of Synechocystis sp. PCC 6803 were previously wired to a Pt nanoparticle via a molecular wire consisting of 15-(3-methyl-1,4-naphthoquinone-2-yl)]pentadecyl sulfide. In the presence of a sacrificial electron donor and an electron transport mediator, the PS I-NQ(CH2)15S-Pt nanoconstruct generated dihydrogen at a rate of 44.3 µmol of H2 mg Chl-1 h-1 during illumination at pH 8.3. The menB deletion strain contains an interruption in the biosynthetic pathway of phylloquinone, which results in the presence of a displaceable plastoquinone-9 in the A1A/A1B sites. The synthesized quinone contains a headgroup identical to the native phylloquinone along with a 15-carbon long tail that is terminated in a thiol. The thiol on the molecular wire is used to bind the Pt nanoparticle. In this short communication, we replaced the Pt nanoparticle with an [FeFe]H2ase variant from Clostridium acetobutylicum that contains an exposed iron on the distal [4Fe-4S] cluster afforded by mutating the surface exposed Cys97 residue to Gly. The thiol on the molecular wire is then used to coordinate the corner iron atom of the iron-sulfur cluster. When all three components are combined and illuminated in the presence of a sacrificial electron donor and an electron transport mediator, the PS I-NQ(CH2)15S-[FeFe]H2ase nanoconstruct generated dihydrogen at a rate of 50.3 ± 9.96 µmol of H2 mg Chl-1 h-1 during illumination at pH 8.3. This successful in vitro experiment sets the stage for assembling a PS I-NQ(CH2)15S-[FeFe]H2ase nanoconstruct in vivo in the menB mutant of Synechocystis sp. PCC 6803.


Assuntos
Hidrogênio/metabolismo , Hidrogenase/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Complexo de Proteína do Fotossistema I/metabolismo , Clostridium acetobutylicum/enzimologia , Transporte de Elétrons , Concentração de Íons de Hidrogênio , Modelos Biológicos , Modelos Moleculares , Complexo de Proteína do Fotossistema I/química , Quinonas/química , Synechocystis/metabolismo
11.
J Biol Inorg Chem ; 24(6): 783-792, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31493152

RESUMO

[FeFe]-hydrogenase catalyzes the reversible reduction of protons to H2 at a complex metallocofactor site, the H-cluster. Biosynthesis of this active-site H-cluster requires three maturation enzymes: the radical S-adenosylmethionine enzymes HydE and HydG synthesize the nonprotein ligands, while the GTPase HydF provides a scaffold for assembly of the 2Fe subcluster of the H-cluster ([2Fe]H) prior to its transfer to hydrogenase. To delineate the assembly and delivery steps for the 2Fe precursor cluster coordinated to HydF ([2Fe]F), we have heterologously expressed HydF in the presence of HydE alone (HydFE) or HydG alone (HydFG), and characterized the resulting purified HydFE and HydFG using UV-visible, EPR, and FTIR spectroscopies and biochemical assays. The iron-sulfur clusters on HydF are modified by co-expression with HydE or HydG, as evidenced by the changes in the visible, EPR, and FTIR spectral features. Further, biochemical assays show that HydFE is capable of activating HydAΔEFG to a limited extent (~ 1% of WT) even though the normal source of CO and CN- ligands of [2Fe]H (HydG) was absent. Activation assays performed with HydFG, in contrast, exhibit no ability to mature HydAΔEFG. It appears that in the case of HydFE, trace diatomics from the cellular environment are incorporated into a [2Fe]F-like precursor on HydF in the absence of HydG. We conclude that the product of HydE, presumably the dithiomethylamine ligand of [2Fe]H, is absolutely essential to the activation process, while the diatomic products of HydG can be provided from alternate sources.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridium acetobutylicum/metabolismo , Hidrogenase/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Proteínas de Bactérias/química , Clostridium acetobutylicum/enzimologia , Espectroscopia de Ressonância de Spin Eletrônica , Hidrogenase/química , Proteínas Ferro-Enxofre/química , Conformação Proteica , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
12.
J Appl Microbiol ; 126(4): 1128-1139, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30656788

RESUMO

AIMS: To establish the biotechnology platforms for production of bio-based chemicals in various micro-organisms is considered as a promising target to improve renewable production of isoprene. METHODS AND RESULTS: In this study, we heterologously expressed the mevalonate (MVA) isoprene biosynthesis pathway, and explored three strategies of increasing isoprene production in Escherichia coli. We first manipulated the expression levels of the MVA pathway genes through changing the gene cassettes and promoters. To introduce cofactor engineering, we then overexpressed NADP-dependent glyceraldehyde-3-phosphate dehydrogenase gene from Clostridium acetobutylicum to supply available NADPH. To reduce the inhibitory by-product accumulation, we finally knocked out acetate-producing genes, phosphate acetyl transferase and pyruvate oxidase B in E. coliJM109 (DE3), decreasing acetate accumulation 89% and increasing isoprene production 39%. The strategies described here finally increased the isoprene titre to 92 mg l-1 in two-gene deletion strain JMAB-4T7P1Trc, increasing 2·6-fold comparing to strain JM7T7. CONCLUSION: The multimodularly engineering approaches including promoter engineering, cofactor engineering and by-product reducing could be used to improve isoprene production in E. coli. SIGNIFICANCE AND IMPACT OF THE STUDY: The metabolic strategies in this study show us directions for further studies to promote transformation of renewable sources to isoprene.


Assuntos
Vias Biossintéticas/genética , Escherichia coli/metabolismo , Hemiterpenos/biossíntese , Engenharia Metabólica/métodos , Ácido Mevalônico/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Butadienos , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Expressão Gênica , Engenharia Genética , NADP/metabolismo
13.
mBio ; 10(1)2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670620

RESUMO

Butanol production by Clostridium acetobutylicum is accompanied by coproduction of acetone and ethanol, which reduces the yield of butanol and increases the production cost. Here, we report development of several clostridial aldehyde/alcohol dehydrogenase (AAD) variants showing increased butanol selectivity by a series of design and analysis procedures, including random mutagenesis, substrate specificity feature analysis, and structure-based butanol selectivity design. The butanol/ethanol ratios (B/E ratios) were dramatically increased to 17.47 and 15.91 g butanol/g ethanol for AADF716L and AADN655H, respectively, which are 5.8-fold and 5.3-fold higher than the ratios obtained with the wild-type AAD. The much-increased B/E ratio obtained was due to the dramatic reduction in ethanol production (0.59 ± 0.01 g/liter) that resulted from engineering the substrate binding chamber and the active site of AAD. This protein design strategy can be applied generally for engineering enzymes to alter substrate selectivity.IMPORTANCE Renewable biofuel represents one of the answers to solving the energy crisis and climate change problems. Butanol produced naturally by clostridia has superior liquid fuel characteristics and thus has the potential to replace gasoline. Due to the lack of efficient genetic manipulation tools, however, clostridial strain improvement has been slower than improvement of other microorganisms. Furthermore, fermentation coproducing various by-products requires costly downstream processing for butanol purification. Here, we report the results of enzyme engineering of aldehyde/alcohol dehydrogenase (AAD) to increase butanol selectivity. A metabolically engineered Clostridium acetobutylicum strain expressing the engineered aldehyde/alcohol dehydrogenase gene was capable of producing butanol at a high level of selectivity.


Assuntos
Álcool Desidrogenase/metabolismo , Aldeído Desidrogenase/metabolismo , Butanóis/metabolismo , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/metabolismo , Engenharia Metabólica , Acetona/metabolismo , Álcool Desidrogenase/química , Álcool Desidrogenase/genética , Aldeído Desidrogenase/química , Aldeído Desidrogenase/genética , Domínio Catalítico , Etanol/metabolismo , Fermentação , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida
14.
Appl Microbiol Biotechnol ; 103(5): 2441-2447, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30673808

RESUMO

Cyanobacteria are oxygen-evolving photosynthetic bacteria. Established genetic manipulation methods and recently developed gene-regulation tools have enabled the photosynthetic conversion of carbon dioxide to biofuels and valuable chemicals in cyanobacteria, especially in unicellular cyanobacteria. However, the oxygen sensitivity of enzyme(s) introduced into cyanobacteria hampers productivity in some cases. Anabaena sp. PCC 7120 is a filamentous cyanobacterium consisting of a few hundred of vegetative cells, which perform oxygenic photosynthesis. Upon nitrogen deprivation, heterocysts, which are specialized cells for nitrogen fixation, are differentiated from vegetative cells at semiregular intervals. The micro-oxic environment within heterocysts protects oxygen-labile nitrogenase from oxygen. This study aimed to repurpose the heterocyst as a host for the production of chemicals with oxygen-sensitive enzymes under photosynthetic conditions. Herein, Anabaena strains expressing enzymes of 1-butanol synthetic pathway from the anaerobe Clostridium acetobutylicum within heterocysts were created. A strain that expressed a highly oxygen-sensitive Bcd/EtfAB complex produced 1-butanol even under photosynthetic conditions. Furthermore, the 1-butanol production per heterocyst cell of a butanol-producing Anabaena strain was fivefold higher than that per cell of unicellular cyanobacterium with the same set of 1-butanol synthetic pathway genes. Thus, our study showed the usefulness of Anabaena heterocysts as a chassis for anaerobic production driven by oxygen-evolving photosynthesis.


Assuntos
Anabaena/metabolismo , Butanóis/metabolismo , Engenharia Metabólica/métodos , Oxigênio/metabolismo , Fotossíntese/fisiologia , Anabaena/classificação , Anabaena/genética , Anaerobiose , Reatores Biológicos/microbiologia , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética
15.
Carbohydr Res ; 472: 72-75, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30500476

RESUMO

N-acetyltransferases are a family of enzymes that catalyze the transfer of the acetyl moiety (COCH3) from acetyl coenzyme A (Acetyl-CoA) to a primary amine of acceptor substrates from small molecules such as aminoglycoside to macromolecules of various proteins. In this study, the substrate selectivity of three N-acetyltransferases falling into different phylogenetic groups was probed against a series of hexosamines and synthetic peptides. GlmA from Clostridium acetobutylicum and RmNag from Rhizomucor miehei, which have been defined as glucosamine N-acetyltransferases, were herein demonstrated to be also capable of acetylating the free amino group on the very first glycine residue of peptide in spite of varied catalytic efficiency. The human recombinant N-acetyltransferase of Naa10p, however, prefers primary amine groups in the peptides as opposed to glucosamine. The varied preference of GlmA, RmNag and Naa10p probably arose from the divergent evolution of these N-acetyltransferases. The expanded knowledge of acceptor specificity would as well facilitate the application of these N-acetyltransferases in the acetylation of hexosamines or peptides.


Assuntos
Acetiltransferases/metabolismo , Clostridium acetobutylicum/enzimologia , Hexosaminas/química , Peptídeos/química , Rhizomucor/enzimologia , Acetilação , Proteínas de Bactérias/metabolismo , Proteínas Fúngicas/metabolismo , Hexosaminas/metabolismo , Humanos , Acetiltransferase N-Terminal A/metabolismo , Acetiltransferase N-Terminal E/metabolismo , Peptídeos/metabolismo , Filogenia , Especificidade por Substrato
16.
Proc Natl Acad Sci U S A ; 115(51): 12967-12972, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30510005

RESUMO

Ferredoxin-dependent thioredoxin reductase was identified 35 y ago in the fermentative bacterium Clostridium pasteurianum [Hammel KE, Cornwell KL, Buchanan BB (1983) Proc Natl Acad Sci USA 80:3681-3685]. The enzyme, a flavoprotein, was strictly dependent on ferredoxin as reductant and was inactive with either NADPH or NADH. This early work has not been further pursued. We have recently reinvestigated the problem and confirmed that the enzyme, here designated ferredoxin-dependent flavin thioredoxin reductase (FFTR), is a flavoprotein. The enzyme differs from ferredoxin-thioredoxin reductase (FTR), which has a signature [4Fe-4S] cluster, but shows structural similarities to NADP-dependent thioredoxin reductase (NTR). Comparative amino acid sequence analysis showed that FFTR is present in a number of clostridial species, some of which lack both FTR and an archetypal NTR. We have isolated, crystallized, and determined the structural properties of FFTR from a member of this group, Clostridium acetobutylicum, both alone and in complex with Trx. The structures showed an elongated FFTR homodimer, each monomer comprising two Rossmann domains and a noncovalently bound FAD cofactor that exposes the isoalloxazine ring to the solvent. The FFTR structures revealed an alternative domain organization compared with NTR that enables the enzyme to accommodate Fdx rather than NADPH. The results suggest that FFTR exists in a range of conformations with varying degrees of domain separation in solution and that the stacking between the two redox-active groups for the transfer of reducing equivalents results in a profound structural reorganization. A mechanism in accord with the findings is proposed.


Assuntos
Clostridium acetobutylicum/enzimologia , Ferredoxinas/química , Flavoproteínas/química , Cristalografia por Raios X , Flavoproteínas/metabolismo , Flavoproteínas/fisiologia , Modelos Moleculares , NADP/química , Oxirredução , Conformação Proteica , Análise de Sequência de Proteína , Homologia de Sequência
17.
Biotechnol Bioeng ; 115(12): 2951-2961, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30199090

RESUMO

One potential advantage of an extremely thermophilic metabolic engineering host (T opt ≥ 70°C) is facilitated recovery of volatile chemicals from the vapor phase of an active fermenting culture. This process would reduce purification costs and concomitantly alleviate toxicity to the cells by continuously removing solvent fermentation products such as acetone or ethanol, a process we are calling "bio-reactive distillation." Although extremely thermophilic heterologous metabolic pathways can be inspired by existing mesophilic versions, they require thermostable homologs of the constituent enzymes if they are to be utilized in extremely thermophilic bacteria or archaea. Production of acetone from acetyl-CoA and acetate in the mesophilic bacterium Clostridium acetobutylicum utilizes three enzymes: thiolase, acetoacetyl-CoA: acetate CoA transferase (CtfAB), and acetoacetate decarboxylase (Adc). Previously reported biocatalytic pathways for acetone production were demonstrated only as high as 55°C. Here, we demonstrate a synthetic enzymatic pathway for acetone production that functions up to at least 70°C in vitro, made possible by the unusual thermostability of Adc from the mesophile C. acetobutylicum, and heteromultimeric acetoacetyl-CoA:acetate CoA-transferase (CtfAB) complexes from Thermosipho melanesiensis and Caldanaerobacter subterraneus, composed of a highly thermostable α-subunit and a thermally labile ß-subunit. The three enzymes produce acetone in vitro at temperatures of at least 70°C, paving the way for bio-reactive distillation of acetone using a metabolically engineered extreme thermophile as a production host.


Assuntos
Acetona/metabolismo , Proteínas de Bactérias/metabolismo , Carboxiliases/metabolismo , Clostridium acetobutylicum/enzimologia , Biologia Sintética/métodos , Proteínas de Bactérias/genética , Carboxiliases/genética , Clostridium acetobutylicum/genética , Clostridium acetobutylicum/metabolismo , Estabilidade Enzimática , Temperatura Alta , Engenharia Metabólica , Redes e Vias Metabólicas/genética
18.
J Biotechnol ; 280: 49-54, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-29885337

RESUMO

Adipic acid (AA) is an important dicarboxylic acid used for the manufacture of nylon and polyurethane plastics. In this study, a novel adipic acid biosynthetic pathway was designed by extending the cis,cis-muconic acid (MA) biosynthesis through biohydrogenation. Enoate reductase from Clostridium acetobutylicum (CaER), an oxygen-sensitive reductase, was demonstrated to have in vivo enzyme activity of converting cis,cis-muconic acid to adipic acid under microaerobic condition. Engineered Escherichia coli strains were constructed to express the whole pathway and accumulated 5.8 ±â€¯0.9 mg/L adipic acid from simple carbon sources. Considering the different oxygen demands between cis,cis-muconic acid biosynthesis and its degradation, a co-culture system was constructed. To improve production, T7 promoter instead of lac promoter was used for higher level expression of the key enzyme CaER and the titer of adipic acid increased to 18.3 ±â€¯0.6 mg/L. To decrease the oxygen supply to downstream strains expressing CaER, Vitreoscilla hemoglobin (VHb) was introduced to upstream strains for its ability on oxygen obtaining. This attempt further improved the production of this novel pathway and 27.6 ±â€¯1.3 mg/L adipic acid was accumulated under microaerobic condition.


Assuntos
Adipatos/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Oxigênio/farmacologia , Ácido Sórbico/análogos & derivados , Aerobiose , Proteínas de Bactérias/farmacologia , Biocatálise , Vias Biossintéticas , Carbono/farmacologia , Clostridium acetobutylicum/enzimologia , Técnicas de Cocultura , Hidrogenação , Ácido Salicílico/metabolismo , Ácido Sórbico/metabolismo , Hemoglobinas Truncadas/farmacologia
19.
J Am Chem Soc ; 140(24): 7623-7628, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29792026

RESUMO

The [FeFe]-hydrogenases ([FeFe] H2ases) catalyze reversible H2 activation at the H-cluster, which is composed of a [4Fe-4S]H subsite linked by a cysteine thiolate to a bridged, organometallic [2Fe-2S] ([2Fe]H) subsite. Profoundly different geometric models of the H-cluster redox states that orchestrate the electron/proton transfer steps of H2 bond activation have been proposed. We have examined this question in the [FeFe] H2ase I from Clostridium acetobutylicum (CaI) by Fourier-transform infrared (FTIR) spectroscopy with temperature annealing and H/D isotope exchange to identify the relevant redox states and define catalytic transitions. One-electron reduction of Hox led to formation of HredH+ ([4Fe-4S]H2+-FeI-FeI) and Hred' ([4Fe-4S]H1+-FeII-FeI), with both states characterized by low frequency µ-CO IR modes consistent with a fully bridged [2Fe]H. Similar µ-CO IR modes were also identified for HredH+ of the [FeFe] H2ase from Chlamydomonas reinhardtii (CrHydA1). The CaI proton-transfer variant C298S showed enrichment of an H/D isotope-sensitive µ-CO mode, a component of the hydride bound H-cluster IR signal, Hhyd. Equilibrating CaI with increasing amounts of NaDT, and probed at cryogenic temperatures, showed HredH+ was converted to Hhyd. Over an increasing temperature range from 10 to 260 K catalytic turnover led to loss of Hhyd and appearance of Hox, consistent with enzymatic turnover and H2 formation. The results show for CaI that the µ-CO of [2Fe]H remains bridging for all of the "Hred" states and that HredH+ is on pathway to Hhyd and H2 evolution in the catalytic mechanism. These results provide a blueprint for designing small molecule catalytic analogs.


Assuntos
Proteínas de Bactérias/química , Hidrogênio/química , Hidrogenase/química , Proteínas Ferro-Enxofre/química , Catálise , Clostridium acetobutylicum/enzimologia , Elétrons , Cinética , Oxirredução , Prótons , Temperatura
20.
Appl Microbiol Biotechnol ; 102(13): 5775-5783, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29691627

RESUMO

The conversion of solar energy into hydrogen represents a highly attractive strategy for the production of renewable energies. Photosynthetic microorganisms have the ability to produce H2 from sunlight but several obstacles must be overcome before obtaining a sustainable and efficient H2 production system. Cyanobacteria harbor [NiFe] hydrogenases required for the consumption of H2. As a result, their H2 production rates are low, which makes them not suitable for a high yield production. On the other hand, [FeFe] enzymes originating from anaerobic organisms such as Clostridium exhibit much higher H2 production activities, but their sensitivity to O2 inhibition impairs their use in photosynthetic organisms. To reach such a goal, it is therefore important to protect the hydrogenase from O2. The diazotrophic filamentous cyanobacteria protect their nitrogenases from O2 by differentiating micro-oxic cells called heterocysts. Producing [FeFe] hydrogenase in the heterocyst is an attractive strategy to take advantage of their potential in a photosynthetic microorganism. Here, we present a biological engineering approach for producing an active [FeFe] hydrogenase (HydA) from Clostridium acetobutylicum in the heterocysts of the filamentous cyanobacterium Nostoc PCC7120. To further decrease the O2 amount inside the heterocyst, the GlbN cyanoglobin from Nostoc commune was coproduced with HydA in the heterocyst. The engineered strain produced 400 µmol-H2 per mg Chlorophyll a, which represents 20-fold the amount produced by the wild type strain. This result is a clear demonstration that it is possible to associate oxygenic photosynthesis with H2 production by an O2-sensitive hydrogenase.


Assuntos
Clostridium acetobutylicum/enzimologia , Hidrogênio/metabolismo , Hidrogenase/genética , Hidrogenase/metabolismo , Microbiologia Industrial/métodos , Nostoc/genética , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA