Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107.174
Filtrar
1.
Food Chem ; 462: 140991, 2025 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-39208721

RESUMO

Shewanella baltica is a specific spoilage organism of golden pomfret. This study aims to explore the antibacterial mechanism of slightly acidic electrolysed water (SAEW) against S. baltica (strains ABa4, ABe2 and BBe1) in golden pomfret broths by metabolomics, proteomics and bioinformatics analyses. S. baltica was decreased by at least 3.94 log CFU/mL after SAEW treatment, and strain ABa4 had the highest resistance. Under SAEW stress, amino acids and organic acids in S. baltica decreased, and nucleotide related compounds degraded. Furthermore, 100 differentially expressed proteins (DEPs) were identified. Most DEPs of strains ABe2 and BBe1 were down-regulated, while some DEPs of strain ABa4 were up-regulated, especially those oxidative stress related proteins. These results suggest that the modes of SAEW against S. baltica can be traced to the inhibition of amino acid, carbon, nucleotide and sulphur metabolisms, and the loss of functional proteins for temperature regulation, translation, motility and protein folding.


Assuntos
Proteínas de Bactérias , Shewanella , Shewanella/metabolismo , Shewanella/química , Shewanella/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Água/metabolismo , Água/química , Eletrólise , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Antibacterianos/química , Concentração de Íons de Hidrogênio , Vigna/química , Vigna/microbiologia , Vigna/metabolismo
2.
Clin Chim Acta ; 564: 119927, 2025 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-39153656

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) infects over 50% of the global population and is a significant risk factor for gastric cancer. The pathogenicity of H. pylori is primarily attributed to virulence factors such as vacA. Timely and accurate identification, along with genotyping of H. pylori virulence genes, are essential for effective clinical management and controlling its prevalence. METHODS: In this study, we developed a dual-target RAA-LFD assay for the rapid, visual detection of H. pylori genes (16s rRNA, ureA, vacA m1/m2), using recombinase aided amplification (RAA) combined with lateral flow dipstick (LFD) methods. Both 16s rRNA and ureA were selected as identification genes to ensure reliable detection accuracy. RESULTS: A RAA-LFD assay was developed to achieve dual-target amplification at a stable 37 °C within 20 min, followed by visualization using the lateral flow dipstick (LFD). The whole process, from amplification to results, took less than 30 min. The 95 % limit of detection (LOD) for 16 s rRNA and ureA, vacA m1, vacA m2 were determined as 3.8 × 10-2 ng/µL, 5.8 × 10-2 ng/µL and 1.4 × 10-2 ng/µL, respectively. No cross-reaction was observed in the detection of common pathogens including Escherichia coli, Klebsiella pneumoniae, Enterococcus faecalis, Staphylococcus aureus, Pseudomonas aeruginosa, and Bacillus subtilis, showing the assay's high specificity. In the evaluation of the clinical performance of the RAA-LFD assay. A total of 44 gastric juice samples were analyzed, immunofluorescence staining (IFS) and quantitative polymerase chain reaction (qPCR) were used as reference methods. The RAA-LFD results for the 16s rRNA and ureA genes showed complete agreement with qPCR findings, accurately identifying H. pylori infection as confirmed by IFS in 10 out of the 44 patients. Furthermore, the assay successfully genotyped vacA m1/m2 among the positive samples, showing complete agreement with qPCR results and achieving a kappa (κ) value of 1.00. CONCLUSION: The dual-target RAA-LFD assay developed in this study provides a rapid and reliable method for detecting and genotyping H. pylori within 30 min, minimizing dependency on sophisticated laboratory equipment and specialized personnel. Clinical validation confirms its efficacy as a promising tool for effectively control of its prevalence and aiding in the precise treatment of H. pylori-associated diseases.


Assuntos
Proteínas de Bactérias , Helicobacter pylori , Helicobacter pylori/genética , Helicobacter pylori/isolamento & purificação , Proteínas de Bactérias/genética , Humanos , RNA Ribossômico 16S/genética , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/microbiologia , Técnicas de Amplificação de Ácido Nucleico/métodos
3.
Nat Commun ; 15(1): 7839, 2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-39244607

RESUMO

Burkholderia cenocepacia is an opportunistic and infective bacterium containing an orphan DNA methyltransferase called M.BceJIV with roles in regulating gene expression and motility of the bacterium. M.BceJIV recognizes a GTWWAC motif (where W can be an adenine or a thymine) and methylates N6 of the adenine at the fifth base position. Here, we present crystal structures of M.BceJIV/DNA/sinefungin ternary complex and allied biochemical, computational, and thermodynamic analyses. Remarkably, the structures show not one, but two DNA substrates bound to the M.BceJIV dimer, with each monomer contributing to the recognition of two recognition sequences. We also show that methylation at the two recognition sequences occurs independently, and that the GTWWAC motifs are enriched in intergenic regions in the genomes of B. cenocepacia strains. We further computationally assess the interactions underlying the affinities of different ligands (SAM, SAH, and sinefungin) for M.BceJIV, as a step towards developing selective inhibitors for limiting B. cenocepacia infection.


Assuntos
Proteínas de Bactérias , Burkholderia cenocepacia , Metilação de DNA , DNA Bacteriano , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Epigênese Genética , Regulação Bacteriana da Expressão Gênica , Cristalografia por Raios X , Motivos de Nucleotídeos , Ligação Proteica
4.
Nat Commun ; 15(1): 7848, 2024 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-39245746

RESUMO

The accidental human pathogen Legionella pneumophila (Lp) is the etiological agent for a severe atypical pneumonia known as Legionnaires' disease. In human infections and animal models of disease alveolar macrophages are the primary cellular niche that supports bacterial replication within a unique intracellular membrane-bound organelle. The Dot/Icm apparatus-a type IV secretion system that translocates ~300 bacterial proteins within the cytosol of the infected cell-is a central virulence factor required for intracellular growth. Mutant strains lacking functional Dot/Icm apparatus are transported to and degraded within the lysosomes of infected macrophages. The early foundational work from Dr. Horwitz's group unequivocally established that Legionella does not replicate extracellularly during infection-a phenomenon well supported by experimental evidence for four decades. Our data challenges this paradigm by demonstrating that macrophages and monocytes provide the necessary nutrients and support robust Legionella extracellular replication. We show that the previously reported lack of Lp extracellular replication is not a bacteria intrinsic feature but rather a result of robust restriction by serum-derived nutritional immunity factors. Specifically, the host iron-sequestering protein Transferrin is identified here as a critical suppressor of Lp extracellular replication in an iron-dependent manner. In iron-overload conditions or in the absence of Transferrin, Lp bypasses growth restriction by IFNγ-primed macrophages though extracellular replication. It is well established that certain risk factors associated with development of Legionnaires' disease, such as smoking, produce a chronic pulmonary environment of iron-overload. Our work indicates that iron-overload could be an important determinant of severe infection by allowing Lp to overcome nutritional immunity and replicate extracellularly, which in turn would circumvent intracellular cell intrinsic host defenses. Thus, we provide evidence for nutritional immunity as a key underappreciated host defense mechanism in Legionella pathogenesis.


Assuntos
Ferro , Legionella pneumophila , Doença dos Legionários , Legionella pneumophila/imunologia , Doença dos Legionários/microbiologia , Doença dos Legionários/imunologia , Ferro/metabolismo , Animais , Camundongos , Humanos , Macrófagos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Transferrina/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Feminino
5.
Methods Enzymol ; 703: 195-213, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39260996

RESUMO

Nonheme iron enzymes are versatile biocatalysts for a broad range of unique and powerful transformations, such as hydroxylation, chlorination, and epimerization as well as cyclization/ring-opening of organic molecules. Beyond their native biological functions, these enzymes are robust for engineering due to their structural diversity and high evolvability. Based on enzyme promiscuity and directed evolution as well as inspired by synthetic organic chemistry, nonheme iron enzymes can be repurposed to catalyze reactions previously only accessible with synthetic catalysts. To this end, our group has engineered a series of nonheme iron enzymes to employ non-natural radical-relay mechanisms for new-to-nature radical transformations. In particular, we have demonstrated that a nonheme iron enzyme, (4-hydroxyphenyl)pyruvate dioxygenase from streptomyces avermitilis (SavHppD), can be repurposed to enable abiological radical-relay process to access C(sp3)-H azidation products. This represents the first known instance of enzymatic radical relay azidation reactions. In this chapter, we describe the detailed experimental protocol to convert promiscuous nonheme iron enzymes into efficient and selective biocatalyst for radical relay azidation reactions. One round of directed evolution is described in detail, which includes the generation and handling of site-saturation mutagenesis, protein expression and whole-cell reactions screening in a 96-well plate. These protocol details might be useful to engineer various nonheme iron enzymes for other applications.


Assuntos
Biocatálise , Engenharia de Proteínas , Streptomyces , Engenharia de Proteínas/métodos , Streptomyces/enzimologia , Streptomyces/genética , Ferroproteínas não Heme/química , Ferroproteínas não Heme/metabolismo , Ferroproteínas não Heme/genética , 4-Hidroxifenilpiruvato Dioxigenase/genética , 4-Hidroxifenilpiruvato Dioxigenase/metabolismo , 4-Hidroxifenilpiruvato Dioxigenase/química , Azidas/química , Azidas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo
6.
Methods Enzymol ; 703: 263-297, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39260999

RESUMO

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.


Assuntos
Oxirredução , Sinorhizobium meliloti , Sinorhizobium meliloti/genética , Sinorhizobium meliloti/enzimologia , Sinorhizobium meliloti/metabolismo , Oxigenases/metabolismo , Oxigenases/genética , Oxigenases/química , Desmetilação , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química
7.
Methods Enzymol ; 703: 65-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39261004

RESUMO

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.


Assuntos
Oxirredução , Oxigenases , Oxigenases/metabolismo , Oxigenases/química , Oxigenases/genética , Oxigenases/isolamento & purificação , Oxirredutases/metabolismo , Oxirredutases/química , Oxirredutases/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Sistema Enzimático do Citocromo P-450/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/isolamento & purificação , Rhodococcus/enzimologia , Rhodococcus/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/química
8.
Elife ; 132024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39221912

RESUMO

The hox operon in Synechocystis sp. PCC 6803, encoding bidirectional hydrogenase responsible for H2 production, is transcriptionally upregulated under microoxic conditions. Although several regulators for hox transcription have been identified, their dynamics and higher-order DNA structure of hox region in microoxic conditions remain elusive. We focused on key regulators for the hox operon: cyAbrB2, a conserved regulator in cyanobacteria, and SigE, an alternative sigma factor. Chromatin immunoprecipitation sequencing revealed that cyAbrB2 binds to the hox promoter region under aerobic conditions, with its binding being flattened in microoxic conditions. Concurrently, SigE exhibited increased localization to the hox promoter under microoxic conditions. Genome-wide analysis revealed that cyAbrB2 binds broadly to AT-rich genome regions and represses gene expression. Moreover, we demonstrated the physical interactions of the hox promoter region with its distal genomic loci. Both the transition to microoxic conditions and the absence of cyAbrB2 influenced the chromosomal interaction. From these results, we propose that cyAbrB2 is a cyanobacterial nucleoid-associated protein (NAP), modulating chromosomal conformation, which blocks RNA polymerase from the hox promoter in aerobic conditions. We further infer that cyAbrB2, with altered localization pattern upon microoxic conditions, modifies chromosomal conformation in microoxic conditions, which allows SigE-containing RNA polymerase to access the hox promoter. The coordinated actions of this NAP and the alternative sigma factor are crucial for the proper hox expression in microoxic conditions. Our results highlight the impact of cyanobacterial chromosome conformation and NAPs on transcription, which have been insufficiently investigated.


Assuntos
Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Hidrogenase , Regiões Promotoras Genéticas , Synechocystis , Synechocystis/genética , Synechocystis/metabolismo , Synechocystis/enzimologia , Hidrogenase/metabolismo , Hidrogenase/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Fermentação , Óperon
9.
Methods Enzymol ; 703: 215-242, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39260997

RESUMO

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.


Assuntos
NAD , NAD/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Oxigenases/metabolismo , Oxigenases/química , Oxigenases/isolamento & purificação , Cristalografia por Raios X/métodos , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/química , Complexo III da Cadeia de Transporte de Elétrons/isolamento & purificação , NADP/metabolismo
10.
Arch Microbiol ; 206(10): 401, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39261350

RESUMO

Staphylococcus aureus is a notorious pathogen predominantly involved in skin and soft tissue infections, exhibiting a distinct innate sex bias. This study explores the influence of testosterone on the virulence of S. aureus and elucidates its underlying mechanisms. Utilizing a skin abscess model in intact and castrated male mice, we assessed the effects of testosterone on S. aureus pathogenicity. Compared to controls, castrated mice showed significantly reduced abscess sizes and decreased bacterial loads, highlighting the role of testosterone in modulating the severity of S. aureus infections. In vitro experiments revealed that testosterone enhances the hemolytic activity, cytotoxicity, and oxidative stress resistance of S. aureus. Real-time quantitative PCR analysis showed a significant upregulation of the genes encoding α-hemolysin (hla) and phenol-soluble modulin (psmα). Importantly, testosterone treatment significantly enhanced the expression of the accessory gene regulator (Agr) quorum-sensing system components (agrC, agrA, agrB, agrD), while the SaeRS system (saeR, saeS, and sbi) exhibited only slight changes. Gene knockout experiments revealed that deletion of agrC, rather than saeRS and agrBD, abolishes the testosterone-induced enhancement of hemolysis and gene expression, underscoring the key role of AgrC. Molecular docking simulations indicated a direct interaction between testosterone and AgrC protein, with a strong binding affinity at the active site residue SER201. This study provides new insights into the mechanistic basis of how testosterone enhances the pathogenicity of S. aureus, potentially contributing to increased male susceptibility to S. aureus infections and offering a targeted approach for therapeutic interventions.


Assuntos
Proteínas de Bactérias , Infecções Estafilocócicas , Staphylococcus aureus , Testosterona , Masculino , Testosterona/farmacologia , Testosterona/metabolismo , Animais , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Camundongos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Virulência , Infecções Estafilocócicas/microbiologia , Transativadores/genética , Transativadores/metabolismo , Regulação Bacteriana da Expressão Gênica , Percepção de Quorum , Simulação de Acoplamento Molecular , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Abscesso/microbiologia , Hemólise , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/genética
11.
Proc Natl Acad Sci U S A ; 121(40): e2403842121, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39264745

RESUMO

Modern molecular microbiology elucidates the organizational principles of bacterial biofilms via detailed examination of the interplay between signaling and gene regulation. A complementary biophysical approach studies the mesoscopic dependencies at the cellular and multicellular levels with a distinct focus on intercellular forces and mechanical properties of whole biofilms. Here, motivated by recent advances in biofilm research and in other, seemingly unrelated fields of biology and physics, we propose a perspective that links the biofilm, a dynamic multicellular organism, with the physical processes occurring in the extracellular milieu. Using Bacillus subtilis as an illustrative model organism, we specifically demonstrate how such a rationale explains biofilm architecture, differentiation, communication, and stress responses such as desiccation tolerance, metabolism, and physiology across multiple scales-from matrix proteins and polysaccharides to macroscopic wrinkles and water-filled channels.


Assuntos
Bacillus subtilis , Biofilmes , Biofilmes/crescimento & desenvolvimento , Bacillus subtilis/fisiologia , Bacillus subtilis/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética
12.
Methods Enzymol ; 704: 113-142, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39300645

RESUMO

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.


Assuntos
Proteínas de Bactérias , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Streptomyces/genética , Streptomyces/enzimologia , Streptomyces/metabolismo , Oxigenases/metabolismo , Oxigenases/genética , Oxigenases/química , Oxigenases/isolamento & purificação , Ensaios Enzimáticos/métodos , Oxazinas/química , Oxazinas/metabolismo , Ferro/metabolismo , Ferro/química , Escherichia coli/genética , Escherichia coli/metabolismo , Ferroproteínas não Heme/metabolismo , Ferroproteínas não Heme/química , Ferroproteínas não Heme/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/química
13.
Methods Enzymol ; 704: 143-172, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39300646

RESUMO

The isonitrile group is a compact, electron-rich moiety coveted for its commonplace as a building block and bioorthogonal functionality in synthetic chemistry and chemical biology. Hundreds of natural products containing an isonitrile group with intriguing bioactive properties have been isolated from diverse organisms. Our recent discovery of a conserved biosynthetic gene cluster in some Actinobacteria species highlighted a novel enzymatic pathway to isonitrile formation involving a non-heme iron(II) and α-ketoglutarate-dependent dioxygenase. Here, we focus this chapter on recent advances in understanding and probing the biosynthetic machinery for isonitrile synthesis by non-heme iron(II) and α-ketoglutarate-dependent dioxygenases. We will begin by describing how to harness isonitrile enzymatic machinery through heterologous expression, purification, synthetic strategies, and in vitro biochemical/kinetic characterization. We will then describe a generalizable strategy to probe the mechanism for isonitrile formation by combining various spectroscopic methods. The chapter will also cover strategies to study other enzyme homologs by implementing coupled assays using biosynthetic pathway enzymes. We will conclude this chapter by addressing current challenges and future directions in understanding and engineering isonitrile synthesis.


Assuntos
Nitrilas , Nitrilas/metabolismo , Nitrilas/química , Ácidos Cetoglutáricos/metabolismo , Oxirredutases/metabolismo , Oxirredutases/genética , Oxirredutases/química , Família Multigênica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Ferro/metabolismo , Ferro/química , Vias Biossintéticas , Dioxigenases/metabolismo , Dioxigenases/genética , Dioxigenases/química , Cinética , Actinobacteria/enzimologia , Actinobacteria/genética , Actinobacteria/metabolismo
14.
Commun Biol ; 7(1): 1173, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39294212

RESUMO

The essential L,D-transpeptidase of Mycobacterium tuberculosis (LdtMt2) catalyses the formation of 3 → 3 cross-links in cell wall peptidoglycan and is a target for development of antituberculosis therapeutics. Efforts to inhibit LdtMt2 have been hampered by lack of knowledge of how it binds its substrate. To address this gap, we optimised the isolation of natural disaccharide tetrapeptide monomers from the Corynebacterium jeikeium bacterial cell wall through overproduction of the peptidoglycan sacculus. The tetrapeptides were used in binding / turnover assays and biophysical studies on LdtMt2. We determined a crystal structure of wild-type LdtMt2 reacted with its natural substrate, the tetrapeptide monomer of the peptidoglycan layer. This structure shows formation of a thioester linking the catalytic cysteine and the donor substrate, reflecting an intermediate in the transpeptidase reaction; it informs on the mode of entrance of the donor substrate into the LdtMt2 active site. The results will be useful in design of LdtMt2 inhibitors, including those based on substrate binding interactions, a strategy successfully employed for other nucleophilic cysteine enzymes.


Assuntos
Mycobacterium tuberculosis , Peptidil Transferases , Mycobacterium tuberculosis/enzimologia , Cristalografia por Raios X , Peptidil Transferases/metabolismo , Peptidil Transferases/química , Especificidade por Substrato , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Peptidoglicano/metabolismo , Peptidoglicano/química , Domínio Catalítico , Modelos Moleculares , Parede Celular/metabolismo , Corynebacterium/enzimologia
15.
BMC Vet Res ; 20(1): 421, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39304865

RESUMO

BACKGROUND: Mycoplasmal pneumonia of sheep and goats (MPSG) is an important infectious disease that threatens sheep and goat production worldwide, and Mycoplasma ovipneumoniae (Movi) is one of the major aetiological agents causing MPSG. The aim of this study was to investigate the immunological activity of the Hsp70‒P113 fusion protein derived from Movi and to develop a serological assay for the detection of Movi. METHODS: This study involved codon optimization of the dominant antigenic regions of Movi heat shock protein 70 (Hsp70) and adhesin P113. Afterwards, the optimized sequences were inserted into the prokaryotic expression vector pET-30a( +) through tandem linking with the aid of a linker. Once a positive recombinant plasmid (pET-30a-rHsp70-P113) was successfully generated, the expression conditions were further refined. The resulting double gene fusion target protein (rHsp70‒P113) was subsequently purified using ProteinIso® Ni-NTA resin, and the reactivity of the protein was confirmed via SDS‒PAGE and Western blot analysis. An indirect enzyme-linked immunosorbent assay (i-ELISA) technique was developed to detect Movi utilizing the fusion protein as the coating antigen. The specificity, sensitivity, and reproducibility of all methods were assessed after each reaction parameter was optimized. RESULTS: The resulting rHsp70-P113 protein had a molecular weight of approximately 51 kDa and was predominantly expressed in the supernatant. Western blot analysis demonstrated its favourable reactivity. The optimal parameters for the i-ELISA technique were as follows: the rHsp70-P113 protein was encapsulated at a concentration of 5 µg/mL; the serum was diluted at a ratio of 1:50; the HRP-labelled donkey anti-goat IgG was diluted at a ratio of 1:6,000. The results of the cross-reactivity assays revealed that the i-ELISA was not cross-reactive with other goat-positive sera against Mycoplasma mycodies subsp. capri (Mmc), Mycoplasma capricolum subsp. capripneumoniae (Mccp), Mycoplasma arginini (Marg), orf virus (ORFV) or enzootic nasal tumour virus of goats (ENTV-2). The sensitivity of this method is high, with a maximum dilution of up to 1:640. The results of the intra- and inter-batch replication tests revealed that the coefficients of variation were both less than 10%, indicating excellent reproducibility. The analysis of 108 clinical serum samples via i-ELISA and indirect haemagglutination techniques yielded significant findings. Among these samples, 43 displayed positive results, whereas 65 presented negative results, resulting in a positivity rate of 39.8% for the i-ELISA method. In contrast, the indirect haemagglutination technique identified 20 positive samples and 88 negative samples, resulting in a positivity rate of 18.5%. Moreover, a comparison between the two methods revealed a conformity rate of 78.7%. CONCLUSION: The results obtained in this study lay the groundwork for advancements in the use of an Movi antibody detection kit, epidemiological inquiry, and subunit vaccines.


Assuntos
Ensaio de Imunoadsorção Enzimática , Doenças das Cabras , Cabras , Proteínas de Choque Térmico HSP70 , Mycoplasma ovipneumoniae , Pneumonia por Mycoplasma , Proteínas Recombinantes de Fusão , Doenças dos Ovinos , Animais , Mycoplasma ovipneumoniae/imunologia , Mycoplasma ovipneumoniae/genética , Proteínas de Choque Térmico HSP70/imunologia , Proteínas de Choque Térmico HSP70/genética , Doenças das Cabras/diagnóstico , Doenças das Cabras/imunologia , Doenças das Cabras/microbiologia , Ensaio de Imunoadsorção Enzimática/veterinária , Ensaio de Imunoadsorção Enzimática/métodos , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/diagnóstico , Doenças dos Ovinos/microbiologia , Ovinos , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/genética , Pneumonia por Mycoplasma/veterinária , Pneumonia por Mycoplasma/diagnóstico , Pneumonia por Mycoplasma/imunologia , Adesinas Bacterianas/imunologia , Adesinas Bacterianas/genética , Anticorpos Antibacterianos/sangue , Sensibilidade e Especificidade , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/genética
16.
Microbiology (Reading) ; 170(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39222353

RESUMO

Phase variation is defined as the rapid and reversible switching of gene expression, and typically occurs in genes encoding surface features in small genome bacterial pathogens. Phase variation has evolved to provide an extra survival mechanism in bacteria that lack multiple 'sense-and-respond' gene regulation systems. Many bacterial pathogens also encode DNA methyltransferases that are phase-variable, controlling systems called 'phasevarions' (phase-variable regulons). This primer will summarize the current understanding of phase variation, describing the role of major phase-variable factors, and phasevarions, in bacterial pathobiology.


Assuntos
Bactérias , Regulação Bacteriana da Expressão Gênica , Bactérias/genética , Bactérias/metabolismo , Adaptação Fisiológica/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos , Regulon
17.
Enzymes ; 55: 1-29, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39222988

RESUMO

Bacterial carbonic anhydrases (BCAs, EC 4.2.1.1) are indispensable enzymes in microbial physiology because they facilitate the hydration of carbon dioxide (CO2) to bicarbonate ions (HCO3-) and protons (H+), which are crucial for various metabolic processes and cellular homeostasis. Their involvement spans from metabolic pathways, such as photosynthesis, respiration, to organic compounds production, which are pivotal for bacterial growth and survival. This chapter elucidates the diversity of BCA genetic families, categorized into four distinct classes (α, ß, γ, and ι), which may reflect bacterial adaptation to environmental niches and their metabolic demands. The diversity of BCAs is essential not only for understanding their physiological roles but also for exploring their potential in biotechnology. Knowledge of their diversity enables researchers to develop innovative biocatalysts for industrial applications, including carbon capture technologies to convert CO2 emissions into valuable products. Additionally, BCAs are relevant to biomedical research and drug development because of their involvement in bacterial pathogenesis and microbial survival within the host. Understanding the diversity and function of BCAs can aid in designing targeted therapeutics that interfere with bacterial metabolism and potentially reduce the risk of infections.


Assuntos
Bactérias , Anidrases Carbônicas , Anidrases Carbônicas/metabolismo , Anidrases Carbônicas/genética , Bactérias/enzimologia , Bactérias/genética , Dióxido de Carbono/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
18.
Enzymes ; 55: 121-142, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39222989

RESUMO

Recent research has identified a novel class of carbonic anhydrases (CAs), designated ι-CA, predominantly found in marine diatoms, eukaryotic algae, cyanobacteria, bacteria, and archaea genomes. This class has garnered attention owing to its unique biochemical properties and evolutionary significance. Through bioinformatic analyses, LCIP63, a protein initially annotated with an unknown function, was identified as a potential ι-CA in the marine diatom Thalassiosira pseudonana. Subsequent biochemical characterization revealed that LCIP63 has CA activity and its preference for manganese ions over zinc, indicative of evolutionary adaptation to marine environments. Further exploration of bacterial ι-CAs, exemplified by Burkholderia territorii ι-CA (BteCAι), demonstrated catalytic efficiency and sensitivity to sulfonamide and inorganic anion inhibitors, the classical CA inhibitors (CAIs). The classification of ι-CAs into two variant types based on their sequences, distinguished by the COG4875 and COG4337 domains, marks a significant advancement in our understanding of these enzymes. Structural analyses of COG4337 ι-CAs from eukaryotic microalgae and cyanobacteria thereafter revealed a distinctive structural arrangement and a novel catalytic mechanism involving specific residues facilitating CO2 hydration in the absence of metal ion cofactors, deviating from canonical CA behavior. These findings underscore the biochemical diversity within the ι-CA class and highlight its potential as a target for novel antimicrobial agents. Overall, the elucidation of ι-CA properties and mechanisms advances our knowledge of carbon metabolism in diverse organisms and underscores the complexity of CA evolution and function.


Assuntos
Anidrases Carbônicas , Anidrases Carbônicas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bactérias/efeitos dos fármacos , Burkholderia , Diatomáceas , Cianobactérias , Inibidores da Anidrase Carbônica/farmacologia , Inibidores da Anidrase Carbônica/química
19.
Enzymes ; 55: 343-381, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39222997

RESUMO

Mycobacterium tuberculosis (Mtb), which causes tuberculosis (TB), is still a major global health problem. According to the World Health Organization (WHO), TB still causes more deaths worldwide than any other infectious agent. Drug-sensitive TB is treatable using first-line drugs; treatment of multidrug-resistant (MDR) and extensively drug-resistant (XDR) TB requires second- and third-line drugs. However, due to the long duration of treatment, the noncompliance of patients with different levels of resistance of Mtb to these drugs has worsened the situation. Previously developed anti-TB drugs targeted the replication machinery, protein synthesis, and cell wall biosynthesis pathways of Mtb. Therefore, novel drugs targeting alternate pathways crucial for the survival and pathogenesis of Mtb in the human host are needed. The genome of Mtb encodes three ß-carbonic anhydrases (CAs) that are fundamental for pH homeostasis, hypoxia, survival, and pathogenesis. Recently, several studies have shown that the ß-CAs of Mtb could be inhibited both in vitro and in vivo using small chemical molecules, suggesting that these enzymes could be novel targets for developing anti-TB compounds that are devoid of resistance by Mtb. In addition, homologs of ß-CAs are absent in humans; therefore, drugs developed to target these enzymes might have minimal off-target effects. In this work, we describe the roles of ß-CAs in Mtb and discuss bioinformatics and cheminformatics tools used in development and discovery of novel inhibitors of these enzymes. In addition, we summarize the in vitro and in vivo studies demonstrating that the ß-CAs of Mtb are indeed druggable targets.


Assuntos
Antituberculosos , Inibidores da Anidrase Carbônica , Anidrases Carbônicas , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Humanos , Anidrases Carbônicas/metabolismo , Antituberculosos/farmacologia , Inibidores da Anidrase Carbônica/farmacologia , Inibidores da Anidrase Carbônica/uso terapêutico , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética
20.
Microb Genom ; 10(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39222339

RESUMO

While conducting genomic surveillance of carbapenemase-producing Enterobacteriaceae (CPE) from patient colonisation and clinical infections at Birmingham's Queen Elizabeth Hospital (QE), we identified an N-type plasmid lineage, pQEB1, carrying several antibiotic resistance genes, including the carbapenemase gene bla KPC-2. The pQEB1 lineage is concerning due to its conferral of multidrug resistance, its host range and apparent transmissibility, and its potential for acquiring further resistance genes. Representatives of pQEB1 were found in three sequence types (STs) of Citrobacter freundii, two STs of Enterobacter cloacae, and three species of Klebsiella. Hosts of pQEB1 were isolated from 11 different patients who stayed in various wards throughout the hospital complex over a 13 month period from January 2023 to February 2024. At present, the only representatives of the pQEB1 lineage in GenBank were carried by an Enterobacter hormaechei isolated from a blood sample at the QE in 2016 and a Klebsiella pneumoniae isolated from a urine sample at University Hospitals Coventry and Warwickshire (UHCW) in May 2023. The UHCW patient had been treated at the QE. Long-read whole-genome sequencing was performed on Oxford Nanopore R10.4.1 flow cells, facilitating comparison of complete plasmid sequences. We identified structural variants of pQEB1 and defined the molecular events responsible for them. These have included IS26-mediated inversions and acquisitions of multiple insertion sequences and transposons, including carriers of mercury or arsenic resistance genes. We found that a particular inversion variant of pQEB1 was strongly associated with the QE Liver speciality after appearing in November 2023, but was found in different specialities and wards in January/February 2024. That variant has so far been seen in five different bacterial hosts from six patients, consistent with recent and ongoing inter-host and inter-patient transmission of pQEB1 in this hospital setting.


Assuntos
Surtos de Doenças , Plasmídeos , beta-Lactamases , Humanos , Plasmídeos/genética , beta-Lactamases/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/epidemiologia , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/isolamento & purificação , Klebsiella pneumoniae/efeitos dos fármacos , Proteínas de Bactérias/genética , Enterobacter cloacae/genética , Enterobacter cloacae/isolamento & purificação , Enterobacter cloacae/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/genética , Infecção Hospitalar/microbiologia , Antibacterianos/farmacologia , Citrobacter freundii/genética , Citrobacter freundii/isolamento & purificação , Enterobacteriáceas Resistentes a Carbapenêmicos/genética , Enterobacteriáceas Resistentes a Carbapenêmicos/isolamento & purificação , Hospitais , Enterobacter
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA