Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.253
Filter
1.
Methods Enzymol ; 703: 263-297, 2024.
Article in English | MEDLINE | ID: mdl-39260999

ABSTRACT

Rieske-type non-heme iron oxygenases (ROs) are an important family of non-heme iron enzymes. They catalyze a diverse range of transformations in secondary metabolite biosynthesis and xenobiotic bioremediation. ROs typically shuttle electrons from NAD(P)H to the oxygenase component via reductase component(s). This chapter describes our recent biochemical characterization of stachydrine demethylase Stc2 from Sinorhizobium meliloti. In this work, the eosin Y/sodium sulfite pair serves as the photoreduction system to replace the NAD(P)H-reductase system. We describe Stc2 protein purification and quality control details as well as a flow-chemistry to separate the photo-reduction half-reaction and the oxidation half-reaction. Our study demonstrates that the eosin Y/sodium sulfite photo-reduction pair is a NAD(P)H-reductase surrogate for Stc2-catalysis in a flow-chemistry setting. Experimental protocols used in this light-driven Stc2 catalysis are likely to be applicable as a photo-reduction system for other redox enzymes.


Subject(s)
Oxidation-Reduction , Sinorhizobium meliloti , Sinorhizobium meliloti/genetics , Sinorhizobium meliloti/enzymology , Sinorhizobium meliloti/metabolism , Oxygenases/metabolism , Oxygenases/genetics , Oxygenases/chemistry , Demethylation , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
2.
Methods Enzymol ; 703: 3-28, 2024.
Article in English | MEDLINE | ID: mdl-39261002

ABSTRACT

Rieske oxygenases are known as catalysts that enable the cleavage of aromatic and aliphatic C-H bonds in structurally diverse biomolecules and recalcitrant organic environmental pollutants through substrate oxygenations and oxidative heteroatom dealkylations. Yet, the unproductive O2 activation, which is concomitant with the release of reactive oxygen species (ROS), is typically not taken into account when characterizing Rieske oxygenase function. Even if considered an undesired side reaction, this O2 uncoupling allows for studying active site perturbations, enzyme mechanisms, and how enzymes evolve as environmental microorganisms adapt their substrates to alternative carbon and energy sources. Here, we report on complementary methods for quantifying O2 uncoupling based on mass balance or kinetic approaches that relate successful oxygenations to total O2 activation and ROS formation. These approaches are exemplified with data for two nitroarene dioxygenases (nitrobenzene and 2-nitrotoluene dioxygenase) which have been shown to mono- and dioxygenate substituted nitroaromatic compounds to substituted nitrobenzylalcohols and catechols, respectively.


Subject(s)
Biodegradation, Environmental , Oxygen , Oxygenases , Oxygen/metabolism , Oxygenases/metabolism , Oxygenases/chemistry , Nitrobenzenes/metabolism , Nitrobenzenes/chemistry , Reactive Oxygen Species/metabolism , Toluene/metabolism , Toluene/analogs & derivatives , Toluene/chemistry , Kinetics , Oxidation-Reduction , Dioxygenases/metabolism , Dioxygenases/chemistry , Environmental Pollutants/metabolism
3.
Methods Enzymol ; 703: 65-85, 2024.
Article in English | MEDLINE | ID: mdl-39261004

ABSTRACT

Oxygenases catalyze crucial reactions throughout all domains of life, cleaving molecular oxygen (O2) and inserting one or two of its atoms into organic substrates. Many oxygenases, including those in the cytochrome P450 (P450) and Rieske oxygenase enzyme families, function as multicomponent systems, which require one or more redox partners to transfer electrons to the catalytic center. As the identity of the reductase can change the reactivity of the oxygenase, characterization of the latter with its cognate redox partners is critical. However, the isolation of the native redox partner or partners is often challenging. Here, we report the preparation and characterization of PbdB, the native reductase partner of PbdA, a bacterial P450 enzyme that catalyzes the O-demethylation of para-methoxylated benzoates. Through production in a rhodoccocal host, codon optimization, and anaerobic purification, this procedure overcomes conventional challenges in redox partner production and allows for robust oxygenase characterization with its native redox partner. Key lessons learned here, including the value of production in a related host and rare codon effects are applicable to a broad range of Fe-dependent oxygenases and their components.


Subject(s)
Oxidation-Reduction , Oxygenases , Oxygenases/metabolism , Oxygenases/chemistry , Oxygenases/genetics , Oxygenases/isolation & purification , Oxidoreductases/metabolism , Oxidoreductases/chemistry , Oxidoreductases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/isolation & purification , Rhodococcus/enzymology , Rhodococcus/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/chemistry
4.
Methods Enzymol ; 703: 299-328, 2024.
Article in English | MEDLINE | ID: mdl-39261001

ABSTRACT

The biotechnological potential of Rieske Oxygenases (ROs) and their cognate reductases remains unmet, in part because these systems can be functionally short-lived. Here, we describe a set of experiments aimed at identifying both the functional and structural stability limitations of ROs, using terephthalate (TPA) dioxygenase (from Comamonas strain E6) as a model system. Successful expression and purification of a cofactor-complete, histidine-tagged TPA dioxygenase and reductase protein system requires induction with the Escherichia coli host at stationary phase as well as a chaperone inducing cold-shock and supplementation with additional iron, sulfur, and flavin. The relative stability of the Rieske cluster and mononuclear iron center can then be assessed using spectroscopic and functional measurements following dialysis in an iron chelating buffer. These experiments involve measurements of the overall lifetime of the system via total turnover number using both UV-Visible absorbance and HPLC analyses, as well specific activity as a function of temperature. Important methods for assessing the stability of these multi-cofactor, multi-protein dependent systems at multiple levels of structure (secondary to quaternary) include differential scanning calorimetry, circular dichroism, and metallospectroscopy. Results can be rationalized in terms of three-dimensional structures and bioinformatics. The experiments described here provide a roadmap to a detailed characterization of the limitations of ROs. With a few notable exceptions, these issues are not widely addressed in current literature.


Subject(s)
Enzyme Stability , Oxygenases/chemistry , Oxygenases/metabolism , Oxygenases/genetics , Circular Dichroism/methods , Temperature , Chromatography, High Pressure Liquid/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Spectrophotometry, Ultraviolet/methods
5.
Methods Enzymol ; 703: 167-192, 2024.
Article in English | MEDLINE | ID: mdl-39260995

ABSTRACT

Rieske non-heme iron-dependent oxygenases (ROs) are a versatile group of enzymes traditionally associated with the degradation of aromatic xenobiotics. In addition, ROs have been found to play key roles in natural product biosynthesis, displaying a wide catalytic diversity with typically high regio- and stereo- selectivity. However, the detailed characterization of ROs presents formidable challenges due to their complex structural and functional properties, including their multi-component composition, cofactor dependence, and susceptibility to reactive oxygen species. In addition, the substrate availability of natural product biosynthetic intermediates, the limited solubility of aromatic hydrocarbons, and the radical-mediated reaction mechanism can further complicate functional assays. Despite these challenges, ROs hold immense potential as biocatalysts for pharmaceutical applications and bioremediation. Using cumene dioxygenase (CDO) from Pseudomonas fluorescens IP01 as a model enzyme, this chapter details techniques for characterizing ROs that oxyfunctionalize aromatic hydrocarbons. Moreover, potential pitfalls, anticipated complications, and proposed solutions for the characterization of novel ROs are described, providing a framework for future RO research and strategies for studying this enzyme class. In particular, we describe the methods used to obtain CDO, from construct design to expression conditions, followed by a purification procedure, and ultimately activity determination through various activity assays.


Subject(s)
Oxygenases , Pseudomonas fluorescens , Pseudomonas fluorescens/enzymology , Oxygenases/metabolism , Oxygenases/chemistry , Dioxygenases/metabolism , Dioxygenases/chemistry , Dioxygenases/genetics , Enzyme Assays/methods , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Electron Transport Complex III
6.
Methods Enzymol ; 703: 215-242, 2024.
Article in English | MEDLINE | ID: mdl-39260997

ABSTRACT

The Rieske non-heme iron oxygenases (Rieske oxygenases) comprise a class of metalloenzymes that are involved in the biosynthesis of complex natural products and the biodegradation of aromatic pollutants. Despite this desirable catalytic repertoire, industrial implementation of Rieske oxygenases has been hindered by the multicomponent nature of these enzymes and their requirement for expensive reducing equivalents in the form of a reduced nicotinamide adenine dinucleotide cosubstrate (NAD(P)H). Fortunately, however, some Rieske oxygenases co-occur with accessory proteins, that through a downstream reaction, recycle the needed NAD(P)H for catalysis. As these pathways and accessory proteins are attractive for bioremediation applications and enzyme engineering campaigns, herein, we describe methods for assembling Rieske oxygenase pathways in vitro. Further, using the TsaMBCD pathway as a model system, in this chapter, we provide enzymatic, spectroscopic, and crystallographic methods that can be adapted to explore both Rieske oxygenases and their co-occurring accessory proteins.


Subject(s)
NAD , NAD/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Oxygenases/metabolism , Oxygenases/chemistry , Oxygenases/isolation & purification , Crystallography, X-Ray/methods , Electron Transport Complex III/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/isolation & purification , NADP/metabolism
7.
Methods Enzymol ; 704: 27-38, 2024.
Article in English | MEDLINE | ID: mdl-39300651

ABSTRACT

Rieske non-heme iron oxygenases are ubiquitously expressed in prokaryotes. These enzymes catalyze a wide variety of reactions, including cis-dihydroxylation, mono-hydroxylation, sulfoxidation, and demethylation. They contain a Rieske-type [2Fe-2S] cluster and an active site with a mono-nuclear iron bound to a 2-His carboxylate triad. Naphthalene 1,2 dioxygenase, a representative of this family, catalyzes the conversion of naphthalene to (+)-cis-(1R,2S)-dihydroxy-1,2-dihydronaphthalene. This transformation requires naphthalene, two electrons, and an oxygen molecule. The first structure of the terminal oxygenase component of a Rieske non-heme iron oxygenase to be determined was naphthalene 1,2 dioxygenase (NDO-O). In this article, we describe in detail the methods used to recombinantly express and purify NDO-O in rich and minimal salts media, the crystallization of NDO-O for structure determination by X-ray crystallography, the challenges faced, and the methods used for the preparation of enzyme ligand complexes. The methods used here resulted in the determination of several NDO-O complexes with aromatic substrates, nitric oxide, oxygen molecule, and products, leading to an initial understanding of the mechanism of enzyme catalysis and the molecular determinants of the regio- and stereo-specificity of this class of enzymes.


Subject(s)
Dioxygenases , Dioxygenases/chemistry , Dioxygenases/metabolism , Dioxygenases/genetics , Crystallography, X-Ray/methods , Naphthalenes/chemistry , Naphthalenes/metabolism , Oxygenases/chemistry , Oxygenases/metabolism , Catalytic Domain , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Crystallization/methods , Models, Molecular , Multienzyme Complexes
8.
BMC Genomics ; 25(1): 872, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294571

ABSTRACT

BACKGROUND: Carotenoid cleavage oxygenases (CCOs) are a group of enzymes that catalyze the oxidative cleavage of carotenoid molecules. These enzymes widely exist in plants, fungi, and certain bacteria, and are involved in various biological processes. It would be of great importance and necessity to identify CCO members in birch and characterize their responses upon abiotic stresses. RESULTS: A total of 16 BpCCOs, including 8 BpCCDs and 8 BpNCEDs were identified in birch, and phylogenetic tree analysis showed that they could be classified into six subgroups. Collinearity analysis revealed that BpCCOs have the largest number of homologous genes in Gossypium hirsutum and also have more homologous genes in other dicotyledons. In addition, promoter analysis revealed that the promoter regions of BpCCOs contained many abiotic stress-related and hormone-responsive elements. The results of qRT-PCR showed that most of the BpCCOs were able to respond significantly to ABA, PEG, salt and cold stresses. Finally, the prediction of the interacting proteins of BpCCOs by STRING revealed several proteins that may interact with BpCCOs and be involved in plant growth and development/abiotic stress processes, such as HEC1 (bHLH), ATABA1, ATVAMP714, etc. CONCLUSION: In this study, the CCO members were identified in birch in a genome-wide scale. These results indicate that BpCCO genes may play important roles in the abiotic stress responses of birch plants.


Subject(s)
Betula , Gene Expression Regulation, Plant , Multigene Family , Oxygenases , Phylogeny , Stress, Physiological , Betula/genetics , Stress, Physiological/genetics , Oxygenases/genetics , Oxygenases/metabolism , Promoter Regions, Genetic , Plant Proteins/genetics , Plant Proteins/metabolism , Genome, Plant , Gene Expression Profiling
9.
Methods Enzymol ; 704: 113-142, 2024.
Article in English | MEDLINE | ID: mdl-39300645

ABSTRACT

Oxazinomycin is a C-nucleoside natural product characterized by a 1,3-oxazine ring linked to ribose via a C-C glycosidic bond. Construction of the 1,3-oxazine ring depends on the activity of OzmD, which is a mononuclear non-heme iron-dependent enzyme from a family of enzymes that contain a domain of unknown function (DUF) 4243. OzmD catalyzes an unusual oxidative ring rearrangement of a pyridine derivative that releases cyanide as a by-product in the final stage of oxazinomycin biosynthesis. The intrinsic sensitivity of the OzmD substrate to oxygen along with the oxygen dependency of catalysis presents significant challenges in conducting in vitro enzymatic assays. This chapter describes the detailed procedures that have been used to characterize OzmD, including protein preparation, activity assays, and reaction by-product identification.


Subject(s)
Bacterial Proteins , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Streptomyces/genetics , Streptomyces/enzymology , Streptomyces/metabolism , Oxygenases/metabolism , Oxygenases/genetics , Oxygenases/chemistry , Oxygenases/isolation & purification , Enzyme Assays/methods , Oxazines/chemistry , Oxazines/metabolism , Iron/metabolism , Iron/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Nonheme Iron Proteins/metabolism , Nonheme Iron Proteins/chemistry , Nonheme Iron Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/chemistry
10.
Methods Enzymol ; 704: 3-25, 2024.
Article in English | MEDLINE | ID: mdl-39300653

ABSTRACT

Extradiol dioxygenases are a class of non-heme iron-dependent enzymes found in eukaryotes and prokaryotes that play a vital role in the aerobic catabolism of aromatic compounds. They are generally divided into three evolutionarily independent superfamilies with different protein folds. Our recent studies have shed light on the catalytic mechanisms and structure-function relationships of two specific extradiol dioxygenases: 3-hydroxyanthranilate-3,4-dioxygenase, a Type III enzyme essential in mammals for producing a precursor for nicotinamide adenine dinucleotide, and L-3,4-dihydroxyphenylalanine dioxygenase, an uncommon form of Type I enzymes involved in natural product biosynthesis. This work details the expression and isolation methods for these extradiol dioxygenases and introduces approaches to achieve homogeneity and high occupancy of the enzyme metal centers. Techniques such as ultraviolet-visible and electron paramagnetic resonance spectroscopies, as well as oxygen electrode measurements, are discussed for probing the interaction of the non-heme iron center with ligands and characterizing enzymatic activities. Moreover, protein crystallization has been demonstrated as a powerful tool to study these enzymes. We highlight in crystallo reactions and single-crystal spectroscopic methods to further elucidate enzymatic functions and protein dynamics.


Subject(s)
Lens, Crystalline , Lens, Crystalline/enzymology , Lens, Crystalline/metabolism , Animals , Oxygenases/metabolism , Oxygenases/chemistry , Oxygenases/genetics , Electron Spin Resonance Spectroscopy/methods , Dioxygenases/metabolism , Dioxygenases/chemistry , Dioxygenases/genetics
11.
Inorg Chem ; 63(37): 17056-17066, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39238331

ABSTRACT

Alkane monooxygenase (AlkB) is a membrane-spanning metalloenzyme that catalyzes the terminal hydroxylation of straight-chain alkanes involved in the microbially mediated degradation of liquid alkanes. According to the cryoEM structures, AlkB features a unique multihistidine ligand coordination environment with a long Fe-Fe distance in its active center. Up to now, how AlkB employs the diiron center to activate dioxygen and which species is responsible for triggering the hydroxylation are still elusive. In this work, we constructed computational models and performed quantum mechanics/molecular mechanics (QM/MM) calculations to illuminate the electronic characteristics of the diiron active center and how AlkB carries out the terminal hydroxylation. Our calculations revealed that the spin-spin interaction between two irons is rather weak. The dioxygen may ligate to either the Fe1 or Fe2 atom and prefers to act as a linker to increase the spin-spin interaction of two irons, facilitating the dioxygen cleavage to generate the highly reactive Fe(IV)═O. Thus, AlkB employs Fe(IV)═O to trigger the hydrogen abstraction. In addition, the previously suggested mechanism that AlkB uses both the dioxygen and Fe-coordinated water to perform hydroxylation was calculated to be unlikely. Besides, our results indicate that AlkB cannot use the Fe-coordinated dioxygen to directly trigger hydrogen abstraction.


Subject(s)
Alkanes , Oxygen , Alkanes/chemistry , Alkanes/metabolism , Hydroxylation , Oxygen/chemistry , Oxygen/metabolism , Oxygenases/chemistry , Oxygenases/metabolism , Iron/chemistry , Iron/metabolism , Molecular Structure , Models, Molecular , Density Functional Theory , Quantum Theory , Electrons
12.
Methods Enzymol ; 704: 173-198, 2024.
Article in English | MEDLINE | ID: mdl-39300647

ABSTRACT

α-Ketoglutarate-dependent non-heme iron (α-KG NHI) oxygenases compose one of the largest superfamilies of tailoring enzymes that play key roles in structural and functional diversifications. During the biosynthesis of meroterpenoids, α-KG NHI oxygenases catalyze diverse types of chemical reactions, including hydroxylation, desaturation, epoxidation, endoperoxidation, ring-cleavage, and skeletal rearrangements. Due to their catalytic versatility, keen attention has been focused on functional analyses of α-KG NHI oxygenases. This chapter provides detailed methodologies for the functional analysis of the fungal α-KG NHI oxygenase SptF, which plays an important role in the structural diversification of andiconin-derived meroterpenoids. The procedures included describe how to prepare the meroterpenoid substrate using a heterologous fungal host, measure the in vitro enzymatic activity of SptF, and how to perform structural and mutagenesis studies on SptF. These protocols are also applicable to functional analyses of other α-KG NHI oxygenases.


Subject(s)
Ketoglutaric Acids , Terpenes , Terpenes/metabolism , Terpenes/chemistry , Ketoglutaric Acids/metabolism , Oxygenases/metabolism , Oxygenases/genetics , Oxygenases/chemistry , Fungal Proteins/metabolism , Fungal Proteins/genetics , Nonheme Iron Proteins/metabolism , Nonheme Iron Proteins/chemistry , Nonheme Iron Proteins/genetics , Fungi/metabolism , Fungi/genetics , Fungi/enzymology , Enzyme Assays/methods , Substrate Specificity
13.
Cardiovasc Diabetol ; 23(1): 299, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143579

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is associated with systemic inflammation, obesity, metabolic syndrome, and gut microbiome changes. Increased trimethylamine-N-oxide (TMAO) levels are predictive for mortality in HFpEF. The TMAO precursor trimethylamine (TMA) is synthesized by the intestinal microbiome, crosses the intestinal barrier and is metabolized to TMAO by hepatic flavin-containing monooxygenases (FMO). The intricate interactions of microbiome alterations and TMAO in relation to HFpEF manifestation and progression are analyzed here. METHODS: Healthy lean (L-ZSF1, n = 12) and obese ZSF1 rats with HFpEF (O-ZSF1, n = 12) were studied. HFpEF was confirmed by transthoracic echocardiography, invasive hemodynamic measurements, and detection of N-terminal pro-brain natriuretic peptide (NT-proBNP). TMAO, carnitine, symmetric dimethylarginine (SDMA), and amino acids were measured using mass-spectrometry. The intestinal epithelial barrier was analyzed by immunohistochemistry, in-vitro impedance measurements and determination of plasma lipopolysaccharide via ELISA. Hepatic FMO3 quantity was determined by Western blot. The fecal microbiome at the age of 8, 13 and 20 weeks was assessed using 16s rRNA amplicon sequencing. RESULTS: Increased levels of TMAO (+ 54%), carnitine (+ 46%) and the cardiac stress marker NT-proBNP (+ 25%) as well as a pronounced amino acid imbalance were observed in obese rats with HFpEF. SDMA levels in O-ZSF1 were comparable to L-ZSF1, indicating stable kidney function. Anatomy and zonula occludens protein density in the intestinal epithelium remained unchanged, but both impedance measurements and increased levels of LPS indicated an impaired epithelial barrier function. FMO3 was decreased (- 20%) in the enlarged, but histologically normal livers of O-ZSF1. Alpha diversity, as indicated by the Shannon diversity index, was comparable at 8 weeks of age, but decreased by 13 weeks of age, when HFpEF manifests in O-ZSF1. Bray-Curtis dissimilarity (Beta-Diversity) was shown to be effective in differentiating L-ZSF1 from O-ZSF1 at 20 weeks of age. Members of the microbial families Lactobacillaceae, Ruminococcaceae, Erysipelotrichaceae and Lachnospiraceae were significantly differentially abundant in O-ZSF1 and L-ZSF1 rats. CONCLUSIONS: In the ZSF1 HFpEF rat model, increased dietary intake is associated with alterations in gut microbiome composition and bacterial metabolites, an impaired intestinal barrier, and changes in pro-inflammatory and health-predictive metabolic profiles. HFpEF as well as its most common comorbidities obesity and metabolic syndrome and the alterations described here evolve in parallel and are likely to be interrelated and mutually reinforcing. Dietary adaption may have a positive impact on all entities.


Subject(s)
Disease Models, Animal , Disease Progression , Gastrointestinal Microbiome , Heart Failure , Methylamines , Stroke Volume , Ventricular Function, Left , Animals , Heart Failure/physiopathology , Heart Failure/microbiology , Heart Failure/metabolism , Methylamines/metabolism , Methylamines/blood , Male , Obesity/microbiology , Obesity/physiopathology , Obesity/metabolism , Oxygenases/metabolism , Oxygenases/genetics , Liver/metabolism , Biomarkers/blood , Feces/microbiology , Rats , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Bacteria/metabolism , Dysbiosis
14.
Microb Biotechnol ; 17(8): e70000, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39160605

ABSTRACT

Methane capture via oxidation is considered one of the 'Holy Grails' of catalysis (Tucci and Rosenzweig, 2024). Methane is also a primary greenhouse gas that has to be reduced by 1.2 billion metric tonnes in 10 years to decrease global warming by only 0.23°C (He and Lidstrom, 2024); hence, new technologies are needed to reduce atmospheric methane levels. In Nature, methane is captured aerobically by methanotrophs and anaerobically by anaerobic methanotrophic archaea; however, the anaerobic process dominates. Here, we describe the history and potential of using the two remarkable enzymes that have been cloned with activity for capturing methane: aerobic capture via soluble methane monooxygenase and anaerobic capture via methyl-coenzyme M reductase. We suggest these two enzymes may play a prominent, sustainable role in addressing our current global warming crisis.


Subject(s)
Methane , Oxidoreductases , Oxygenases , Recombinant Proteins , Methane/metabolism , Oxygenases/genetics , Oxygenases/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Oxidation-Reduction , Anaerobiosis , Aerobiosis , Archaea/enzymology , Archaea/genetics , Archaea/metabolism
15.
FEMS Microbiol Ecol ; 100(9)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39122657

ABSTRACT

Methanotrophs are the sole biological sink of methane. Volatile organic compounds (VOCs) produced by heterotrophic bacteria have been demonstrated to be a potential modulating factor of methane consumption. Here, we identify and disentangle the impact of the volatolome of heterotrophic bacteria on the methanotroph activity and proteome, using Methylomonas as model organism. Our study unambiguously shows how methanotrophy can be influenced by other organisms without direct physical contact. This influence is mediated by VOCs (e.g. dimethyl-polysulphides) or/and CO2 emitted during respiration, which can inhibit growth and methane uptake of the methanotroph, while other VOCs had a stimulating effect on methanotroph activity. Depending on whether the methanotroph was exposed to the volatolome of the heterotroph or to CO2, proteomics revealed differential protein expression patterns with the soluble methane monooxygenase being the most affected enzyme. The interaction between methanotrophs and heterotrophs can have strong positive or negative effects on methane consumption, depending on the species interacting with the methanotroph. We identified potential VOCs involved in the inhibition while positive effects may be triggered by CO2 released by heterotrophic respiration. Our experimental proof of methanotroph-heterotroph interactions clearly calls for detailed research into strategies on how to mitigate methane emissions.


Subject(s)
Carbon Dioxide , Methane , Microbial Interactions , Volatile Organic Compounds , Methane/metabolism , Volatile Organic Compounds/metabolism , Carbon Dioxide/metabolism , Methylomonas/metabolism , Methylomonas/genetics , Proteomics , Proteome , Heterotrophic Processes , Oxygenases/metabolism , Oxygenases/genetics
16.
Commun Biol ; 7(1): 1054, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191965

ABSTRACT

The kidney is vulnerable to ischemia and reperfusion (I/R) injury that can be fatal after major surgery. Currently, there are no effective treatments for I/R-induced kidney injury. Trimethylamine N-oxide (TMAO) is a gut-derived metabolite linked to many diseases, but its role in I/R-induced kidney injury remains unclear. Here, our clinical data reveals an association between preoperative systemic TMAO levels and postoperative kidney injury in patients after post-cardiopulmonary bypass surgery. By genetic deletion of TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) and dietary supplementation of choline to modulate TMAO levels, we found that TMAO aggravated acute kidney injury through the triggering of endoplasmic reticulum (ER) stress and worsened subsequent renal fibrosis through TGFß/Smad signaling activation. Together, our study underscores the negative role of TMAO in I/R-induced kidney injury and highlights the therapeutic potential through the modulation of TMAO levels by targeting FMO3, thereby mitigating acute kidney injury and preventing subsequent renal fibrosis.


Subject(s)
Acute Kidney Injury , Kidney , Methylamines , Oxygenases , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Oxygenases/metabolism , Oxygenases/genetics , Mice , Male , Methylamines/metabolism , Kidney/metabolism , Kidney/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Mice, Knockout , Mice, Inbred C57BL , Humans , Fibrosis , Signal Transduction , Endoplasmic Reticulum Stress , Transforming Growth Factor beta/metabolism
17.
Molecules ; 29(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39124879

ABSTRACT

Flavin-containing monooxygenase from Methylophaga sp. (mFMO) was previously discovered to be a valuable biocatalyst used to convert small amines, such as trimethylamine, and various indoles. As FMOs are also known to act on sulfides, we explored mFMO and some mutants thereof for their ability to convert prochiral aromatic sulfides. We included a newly identified thermostable FMO obtained from the bacterium Nitrincola lacisaponensis (NiFMO). The FMOs were found to be active with most tested sulfides, forming chiral sulfoxides with moderate-to-high enantioselectivity. Each enzyme variant exhibited a different enantioselective behavior. This shows that small changes in the substrate binding pocket of mFMO influence selectivity, representing a tunable biocatalyst for enantioselective sulfoxidations.


Subject(s)
Oxygenases , Oxygenases/metabolism , Oxygenases/chemistry , Substrate Specificity , Biocatalysis , Oxidation-Reduction , Sulfides/metabolism , Sulfides/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Sulfoxides/chemistry , Sulfoxides/metabolism , Catalysis , Flavins/metabolism , Flavins/chemistry , Stereoisomerism , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics
18.
Mol Med ; 30(1): 128, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39180015

ABSTRACT

BACKGROUND: Tissue fibrosis is a common pathway to failure in many organ systems and is the cellular and molecular driver of myriad chronic diseases that are incompletely understood and lack effective treatment. Recent studies suggest that gut microbe-dependent metabolites might be involved in the initiation and progression of fibrosis in multiple organ systems. MAIN BODY OF THE MANUSCRIPT: In a meta-organismal pathway that begins in the gut, gut microbiota convert dietary precursors such as choline, phosphatidylcholine, and L-carnitine into trimethylamine (TMA), which is absorbed and subsequently converted to trimethylamine N-oxide (TMAO) via the host enzyme flavin-containing monooxygenase 3 (FMO3) in the liver. Chronic exposure to elevated TMAO appears to be associated with vascular injury and enhanced fibrosis propensity in diverse conditions, including chronic kidney disease, heart failure, metabolic dysfunction-associated steatotic liver disease, and systemic sclerosis. CONCLUSION: Despite the high prevalence of fibrosis, little is known to date about the role of gut dysbiosis and of microbe-dependent metabolites in its pathogenesis. This review summarizes recent important advances in the understanding of the complex metabolism and functional role of TMAO in pathologic fibrosis and highlights unanswered questions.


Subject(s)
Fibrosis , Gastrointestinal Microbiome , Methylamines , Methylamines/metabolism , Humans , Animals , Dysbiosis/metabolism , Oxygenases/metabolism
19.
Sheng Wu Gong Cheng Xue Bao ; 40(8): 2444-2456, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39174464

ABSTRACT

Indigo, as a water-soluble non-azo colorant, is widely used in textile, food, pharmaceutical and other industrial fields. Currently, indigo is primarily synthesized by chemical methods, which causes environmental pollution, potential safety hazards, and other issues. Therefore, there is an urgent need to find a safer and greener synthetic method. In this study, a dual-enzyme cascade pathway was constructed with the tryptophan synthase (tryptophanase, EcTnaA) from Escherichia coli and flavin-dependent monooxygenase (flavin-dependent monooxygenase, MaFMO) from Methylophaga aminisulfidivorans to synthesize indigo with L-tryptophan as substrate. A recombinant strain EM-IND01 was obtained. The beneficial mutant MaFMOD197E was obtained by protein engineering of the rate-limiting enzyme MaFMO. MaFMOD197E showed the specific activity and kcat/Km value 2.36 times and 1.34 times higher than that of the wild type, respectively. Furthermore, MaFMOD197E was introduced into the strain EM-IND01 to construct the strain EM-IND02. After the fermentation conditions were optimized, the strain achieved the indigo titer of (1 288.59±7.50) mg/L, the yield of 0.86 mg/mg L-tryptophan, and the productivity of 26.85 mg/(L·h) in a 5 L fermenter. Protein engineering was used to obtain mutants with increased MaFMO activity in this study, which laid a foundation for industrial production of indigo.


Subject(s)
Escherichia coli , Indigo Carmine , Tryptophan , Indigo Carmine/metabolism , Tryptophan/metabolism , Tryptophan/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Engineering , Tryptophanase/genetics , Tryptophanase/metabolism , Tryptophan Synthase/metabolism , Tryptophan Synthase/genetics , Fermentation , Oxygenases/genetics , Oxygenases/metabolism
20.
N Biotechnol ; 83: 82-90, 2024 Nov 25.
Article in English | MEDLINE | ID: mdl-39053683

ABSTRACT

Previously, some bacteria were shown to harbour enzymes capable of catalysing the oxidative cleavage of the double bond of t-anethole and related compounds. The cofactor dependence of these enzymes remained enigmatic due to a lack of biochemical information. We report on catalytic and structural details of a representative of this group of oxidative enzymes: t-anethole oxygenase from Stenotrophomonas maltophilia (TAOSm). The bacterial enzyme could be recombinantly expressed and purified, enabling a detailed biochemical study that has settled the dispute on its cofactor dependence. We have established that TAOSm contains a tightly bound b-type heme and merely depends on dioxygen for catalysis. It was found to accept t-anethole, isoeugenol and O-methyl isoeugenol as substrates, all being converted into the corresponding aromatic aldehydes without the need of any cofactor regeneration. The elucidated crystal structure of TAOSm has revealed that it contains a unique active site architecture that is conserved for this distinct class of heme-containing bacterial oxygenases. Similar to other hemoproteins, TAOSm has a histidine (His121) as proximal ligand. Yet, unique for TAOs, an arginine (Arg89) is located at the distal axial position. Site directed mutagenesis confirmed crucial roles for these heme-liganding residues and other residues that form the substrate binding pocket. In conclusion, the results reported here reveal a new class of bacterial heme-containing oxygenases that can be used for the cleavage of alkene double bonds, analogous to ozonolysis in organic chemistry.


Subject(s)
Heme , Stenotrophomonas maltophilia , Heme/metabolism , Heme/chemistry , Stenotrophomonas maltophilia/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Oxygenases/metabolism , Oxygenases/chemistry , Models, Molecular , Substrate Specificity , Catalytic Domain
SELECTION OF CITATIONS
SEARCH DETAIL