Mass spectrometry based targeted metabolomics precisely characterized new functional metabolites that regulate biofilm formation in Escherichia coli

2021 ◽  
Vol 1145 ◽  
pp. 26-36
Author(s):  
Rui Guo ◽  
Xialin Luo ◽  
Jingjing Liu ◽  
Haitao Lu
2005 ◽  
Vol 187 (1) ◽  
pp. 382-387 ◽  
Author(s):  
Yoshikane Itoh ◽  
Xin Wang ◽  
B. Joseph Hinnebusch ◽  
James F. Preston ◽  
Tony Romeo

ABSTRACT Polymeric β-1,6-N-acetyl-d-glucosamine (poly-β-1,6-GlcNAc) has been implicated as an Escherichia coli and Staphylococcus epidermidis biofilm adhesin, the formation of which requires the pgaABCD and icaABCD loci, respectively. Enzymatic hydrolysis of poly-β-1,6-GlcNAc, demonstrated for the first time by chromatography and mass spectrometry, disrupts biofilm formation by these species and by Yersinia pestis and Pseudomonas fluorescens, which possess pgaABCD homologues.


2019 ◽  
Author(s):  
Haitao Lu ◽  
Yumei Que ◽  
Xia Wu ◽  
Tianbing Guan ◽  
Hao Guo

ABSTRACTBiofilm formation plays a key role in many bacteria causing infections, which mostly accounts for high-frequency infectious recurrence and antibiotics resistance. In this study, we sought to compare modified metabolism of biofilm and planktonic populations with UIT89, a predominant agent of urinary tract infection, by combining mass spectrometry based untargeted and targeted metabolomics methods, as well as cytological visualization, which enable us to identify the driven metabolites and associated metabolic pathways underlying biofilm formation. Surprisingly, our finding revealed distinct differences in both phenotypic morphology and metabolism between two patterns. Furthermore, we identified and characterized 38 differential metabolites and associated three metabolic pathways involving glycerolipid metabolism, amino acid metabolism and carbohydrate metabolism that were altered mostly during biofilm formation. This discovery in metabolic phenotyping permitted biofilm formation shall provide us a novel insight into the desperation of biofilm, which enable to develop novel biofilm based treatments against pathogen causing infections, with lower antibiotic resistance.


2017 ◽  
Vol 17 (1) ◽  
pp. 69-77
Author(s):  
Tu Lijun ◽  
Sun Hanju ◽  
He Shudong ◽  
Zhu Yongsheng ◽  
Yu Ming ◽  
...  

The aim of this study was to investigate epigallocatechin gallate (EGCG) prebiotics activities systematically which was reported as a bioactive substance. Therefore, EGCG was separated by water extraction, resin purification and prep-HPLC. Then the production of EGCG was confirmed by HPLC and mass spectrometry (MS) analysis and its purify was 97.23%. EGCG extractive and green tea extract (GTE) were further incubated with Bifidobacterium infantis, B. adolescentis, B. bifidum and Lactobacillus acidophilus to study its effect on microbial populations and medium pH. Finally, Escherichia coli, Salmonella, Staphylococcus aureus and Candida albicans were employed as pathogenic bacteria to explore the antimicrobial activity of EGCG and GTE. The results demonstrated that EGCG extractive could be beneficial for the proliferation of Bifidobacterium and L. acidophilus and also inhibit some pathogenic bacteria. In conclusion, both EGCG extractive and GTE had prebiotics activities and the effects of EGCG extractive were superior to those of GTE.


Biofouling ◽  
2021 ◽  
pp. 1-11
Author(s):  
Ahmed Mathlouthi ◽  
Nabil Saadaoui ◽  
Eugenia Pennacchietti ◽  
Daniela De Biase ◽  
Mossadok Ben-Attia

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Huiyi Song ◽  
Ni Lou ◽  
Jianjun Liu ◽  
Hong Xiang ◽  
Dong Shang

Abstract Background Escherichia coli (E. coli) is the principal pathogen that causes biofilm formation. Biofilms are associated with infectious diseases and antibiotic resistance. This study employed proteomic analysis to identify differentially expressed proteins after coculture of E. coli with Lactobacillus rhamnosus GG (LGG) microcapsules. Methods To explore the relevant protein abundance changes after E. coli and LGG coculture, label-free quantitative proteomic analysis and qRT-PCR were applied to E. coli and LGG microcapsule groups before and after coculture, respectively. Results The proteomic analysis characterised a total of 1655 proteins in E. coli K12MG1655 and 1431 proteins in the LGG. After coculture treatment, there were 262 differentially expressed proteins in E. coli and 291 in LGG. Gene ontology analysis showed that the differentially expressed proteins were mainly related to cellular metabolism, the stress response, transcription and the cell membrane. A protein interaction network and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis indicated that the differentiated proteins were mainly involved in the protein ubiquitination pathway and mitochondrial dysfunction. Conclusions These findings indicated that LGG microcapsules may inhibit E. coli biofilm formation by disrupting metabolic processes, particularly in relation to energy metabolism and stimulus responses, both of which are critical for the growth of LGG. Together, these findings increase our understanding of the interactions between bacteria under coculture conditions.


Sign in / Sign up

Export Citation Format

Share Document