1
|
Bai H, Liu T, Wang H, Li Y, Wang Z. Regulating Effects of Three Polysaccharides on Gut Microbiota in Felines and Canines: Insights from In Vitro Digestion, Fecal Fermentation, and Lactic Acid Bacterial Fermentation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9618-9636. [PMID: 40211571 DOI: 10.1021/acs.jafc.4c12847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
This study evaluated the effects of laminaria polysaccharide (FU), aloe polysaccharide (AL), and yeast β-glucan (YG) on in vitro digestion, fecal fermentation, and lactic acid bacterial fermentation in felines and canines. FU (Mw 27 713 g/mol), AL (44 425 g/mol), and YG (29 296 g/mol) were primarily composed of glucose, galacturonic acid, and glucuronic acid. The FU strongly promoted the fermentation of Bifidobacterium, Lactobacillus, and Pediococcus. The AL slightly supported Lactobacillus. The YG showed minimal activity. Molecular weights decreased after digestion with negligible monosaccharide release. The FU and AL were almost completely metabolized during fecal fermentation, while YG had ∼50% utilization. The FU produced the highest lactate concentration. The YG significantly increased SCFA levels, especially in canines, surpassing fructooligosaccharides. The FU:AL:YG mixture (0.8:0.2:1, FAY) achieved the best overall effects. 16S rRNA analysis revealed the enrichment of SCFA-producing genera (e.g., Megamonas, Parabacteroides) and suppression of odor-related bacteria (e.g., Fusobacterium, Desulfovibrio). Metabolomics showed reduced levels of odor compounds (e.g., 2-oxindole, 5-hydroxyindole) and increased secondary bile acids (e.g., deoxycholic acid, glycocholic acid). The FU, AL, and FAY upregulated bile secretion, neuroactive pathways, and arachidonic acid metabolism, while YG activated the ABC transporter pathway.
Collapse
Affiliation(s)
- Huasong Bai
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Tong Liu
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Hengyan Wang
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Yunliang Li
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, PR China
| | - Zhanzhong Wang
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| |
Collapse
|
2
|
Jiang S, Chen H, Chen S, Chen N, Yang H, Duan Y, Ao S, Wang R, Wang X, Zhang Y, Yuan J. Genetically Encoded Biosensors for Constrained Biological Functions in Probiotic Escherichia coli Nissle. ACS Synth Biol 2025; 14:296-303. [PMID: 39772427 DOI: 10.1021/acssynbio.4c00747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The probiotic Escherichia coli Nissle (EcN) is an exceptional strain that has attracted significant attention not only for its clinical efficacy in the treatment and prevention of gastrointestinal disorders but also as a burgeoning microbial chassis for living therapeutic applications. However, there is an immediate necessity to develop conditional expression systems that confine the activity of EcN specifically in the gastrointestinal tract, to avoid influencing the environment. Here, we constructed two genetically encoded interchangeable sensors responsive to body temperature at 37 °C, and small molecules such as protocatechuic acid (PCA), a metabolite found in green tea. By employing dCpf1 targeted deactivation of the LacI gene, we thereby coupled the above sensing modules with the Ptrc-lacO system and achieved improved signal outputs and relatively high ON/OFF ratios. Subsequently, we validated the biological function of engineering EcN using the enhanced green fluorescent protein (eGFP) in an animal model of mice. Taken together, the construction of genetically encoded sensors to restrict the biological functions of EcN would be applicable for the real-world implementation of living therapeutics or drug delivery.
Collapse
Affiliation(s)
- Shan Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Haofeng Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Shiyao Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Na Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Haofeng Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Yiyang Duan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Shiqi Ao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Ruoxi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Xin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Yalin Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
| | - Jifeng Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| |
Collapse
|
3
|
Doranga S, Krogfelt KA, Cohen PS, Conway T. Nutrition of Escherichia coli within the intestinal microbiome. EcoSal Plus 2024; 12:eesp00062023. [PMID: 38417452 PMCID: PMC11636361 DOI: 10.1128/ecosalplus.esp-0006-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/03/2023] [Indexed: 03/01/2024]
Abstract
In this chapter, we update our 2004 review of "The Life of Commensal Escherichia coli in the Mammalian Intestine" (https://doi.org/10.1128/ecosalplus.8.3.1.2), with a change of title that reflects the current focus on "Nutrition of E. coli within the Intestinal Microbiome." The earlier part of the previous two decades saw incremental improvements in understanding the carbon and energy sources that E. coli and Salmonella use to support intestinal colonization. Along with these investigations of electron donors came a better understanding of the electron acceptors that support the respiration of these facultative anaerobes in the gastrointestinal tract. Hundreds of recent papers add to what was known about the nutrition of commensal and pathogenic enteric bacteria. The fact that each biotype or pathotype grows on a different subset of the available nutrients suggested a mechanism for succession of commensal colonizers and invasion by enteric pathogens. Competition for nutrients in the intestine has also come to be recognized as one basis for colonization resistance, in which colonized strain(s) prevent colonization by a challenger. In the past decade, detailed investigations of fiber- and mucin-degrading anaerobes added greatly to our understanding of how complex polysaccharides support the hundreds of intestinal microbiome species. It is now clear that facultative anaerobes, which usually cannot degrade complex polysaccharides, live in symbiosis with the anaerobic degraders. This concept led to the "restaurant hypothesis," which emphasizes that facultative bacteria, such as E. coli, colonize the intestine as members of mixed biofilms and obtain the sugars they need for growth locally through cross-feeding from polysaccharide-degrading anaerobes. Each restaurant represents an intestinal niche. Competition for those niches determines whether or not invaders are able to overcome colonization resistance and become established. Topics centered on the nutritional basis of intestinal colonization and gastrointestinal health are explored here in detail.
Collapse
Affiliation(s)
- Sudhir Doranga
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Karen A. Krogfelt
- Department of Science and Environment, Pandemix Center Roskilde University, Roskilde, Denmark
| | - Paul S. Cohen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, Rhode Island, USA
| | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
4
|
Jandl B, Dighe S, Gasche C, Makristathis A, Muttenthaler M. Intestinal biofilms: pathophysiological relevance, host defense, and therapeutic opportunities. Clin Microbiol Rev 2024; 37:e0013323. [PMID: 38995034 PMCID: PMC11391705 DOI: 10.1128/cmr.00133-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.
Collapse
Affiliation(s)
- Bernhard Jandl
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Satish Dighe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christoph Gasche
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- Loha for Life, Center for Gastroenterology and Iron Deficiency, Vienna, Austria
| | - Athanasios Makristathis
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Alles M, Gunasena M, Zia T, D'Mello A, Bhattarai S, Mulhern W, Terry L, Scherger T, Wijeratne S, Singh S, Wijeratne AJ, Kasturiratna D, Tettelin H, Weyand NJ, Liyanage NPM. Unveiling the immune dynamics of Neisseria persistent oral colonization. Infect Immun 2024; 92:e0004824. [PMID: 38814083 PMCID: PMC11238562 DOI: 10.1128/iai.00048-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/27/2024] [Indexed: 05/31/2024] Open
Abstract
Commensal bacteria are crucial in maintaining host physiological homeostasis, immune system development, and protection against pathogens. Despite their significance, the factors influencing persistent bacterial colonization and their impact on the host still need to be fully understood. Animal models have served as valuable tools to investigate these interactions, but most have limitations. The bacterial genus Neisseria, which includes both commensal and pathogenic species, has been studied from a pathogenicity to humans perspective but lacks models that study immune responses in the context of long-term persistence. Neisseria musculi, a recently described natural commensal of mice, offers a unique opportunity to study long-term host-commensal interactions. In this study, for the first time, we have used this model to study the transcriptional, phenotypic, and functional dynamics of immune cell signatures in the mucosal and systemic tissue of mice in response to N. musculi colonization. We found key genes and pathways vital for immune homeostasis in palate tissue, validated by flow cytometry of immune cells from the lung, blood, and spleen. This study offers a novel avenue for advancing our understanding of host-bacteria dynamics and may provide a platform for developing efficacious interventions against mucosal persistence by pathogenic Neisseria.
Collapse
Affiliation(s)
- Mario Alles
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Manuja Gunasena
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Tauqir Zia
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Adonis D'Mello
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Saroj Bhattarai
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
| | - Will Mulhern
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Luke Terry
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Trenton Scherger
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Saranga Wijeratne
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Sachleen Singh
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, USA
| | - Asela J. Wijeratne
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, USA
| | - Dhanuja Kasturiratna
- Department of Mathematics and Statistics, Northern Kentucky University, Highland Heights, Kentucky, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nathan J. Weyand
- Department of Biological Sciences, Ohio University, Athens, Ohio, USA
- The Infectious and Tropical Disease Institute, Ohio University, Athens, Ohio, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio, USA
| | - Namal P. M. Liyanage
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Zhang Y, Huang H, Luo C, Zhang X, Chen Y, Yue F, Xie B, Chen T, Zou C. The Next-Generation Probiotic E. coli 1917-pSK18a-MT Ameliorates Cadmium-Induced Liver Injury by Surface Display of Metallothionein and Modulation of Gut Microbiota. Nutrients 2024; 16:1468. [PMID: 38794706 PMCID: PMC11124084 DOI: 10.3390/nu16101468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Cadmium (Cd) is recognized as being linked to several liver diseases. Currently, due to the limited spectrum of drugs available for the treatment of Cd intoxication, developing and designing antidotes with superior detoxification capacity and revealing their underlying mechanisms remains a major challenge. Therefore, we developed the first next-generation probiotic E. coli 1917-pSK18a-MT that delivers metallothionein (MT) to overcome Cd-induced liver injury in C57BL/6 mice by utilizing bacterial surface display technology. The results demonstrate that E. coli 1917-pSK18a-MT could efficiently express MT without altering the growth and probiotic properties of the strain. Moreover, we found that E. coli 1917-pSK18a-MT ameliorated Cd contamination-induced hepatic steatosis, inflammatory cell infiltration, and liver fibrosis by decreasing the expression of aminotransferases along with inflammatory factors. Activation of the Nrf2-Keap1 signaling pathway also further illustrated the hepatoprotective effects of the engineered bacteria. Finally, we showed that E. coli 1917-pSK18a-MT improved the colonic barrier function impaired by Cd induction and ameliorated intestinal flora dysbiosis in Cd-poisoned mice by increasing the relative abundance of the Verrucomicrobiota. These data revealed that the combination of E. coli 1917 and MT both alleviated Cd-induced liver injury to a greater extent and restored the integrity of colonic epithelial tissues and bacterial dysbiosis.
Collapse
Affiliation(s)
- Yan Zhang
- School of Resources and Environment, Nanchang University, Nanchang 330031, China; (Y.Z.); (H.H.)
| | - Hong Huang
- School of Resources and Environment, Nanchang University, Nanchang 330031, China; (Y.Z.); (H.H.)
| | - Chuanlin Luo
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China;
| | - Xinfeng Zhang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China;
| | - Yanjing Chen
- Department of Obstetrics & Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Fenfang Yue
- School of Life Sciences, Nanchang University, Nanchang 330031, China;
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Bingqing Xie
- Department of Obstetrics & Gynecology, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Tingtao Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China;
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Changwei Zou
- School of Resources and Environment, Nanchang University, Nanchang 330031, China; (Y.Z.); (H.H.)
| |
Collapse
|
7
|
Giliazeva A, Akosah Y, Noack J, Mardanova A. Adhesion of Klebsiella oxytoca to bladder or lung epithelial cells is promoted by the presence of other opportunistic pathogens. Microb Pathog 2024; 190:106642. [PMID: 38599551 DOI: 10.1016/j.micpath.2024.106642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
The intestinal and respiratory tracts of healthy individuals serve as habitats for a diverse array of microorganisms, among which Klebsiella oxytoca holds significance as a causative agent in numerous community- and hospital-acquired infections, often manifesting in polymicrobial contexts. In specific circumstances, K. oxytoca, alongside other constituents of the gut microbiota, undergoes translocation to distinct physiological niches. In these new environments, it engages in close interactions with other microbial community members. As this interaction may progress to co-infection where the virulence of involved pathogens may be promoted and enhance disease severity, we investigated how K. oxytoca affects the adhesion of commonly co-isolated bacteria and vice versa during co-incubation of different biotic and abiotic surfaces. Co-incubation was beneficial for the adhesion of at least one of the two co-cultured strains. K. oxytoca enhanced the adhesion of other enterobacteria strains to polystyrene and adhered more efficiently to bladder or lung epithelial cell lines in the presence of most enterobacteria strains and S. aureus. This effect was accompanied by bacterial coaggregation mediated by carbohydrate-protein interactions occurring between bacteria. These interactions occur only in sessile, but not planktonic populations, and depend on the features of the surface. The data are of particular importance for the risk assessment of the urinary and respiratory tract infections caused by K. oxytoca, including those device-associated. In this paper, we present the first report on K. oxytoca ability to acquire increased adhesive capacities on epithelial cells through interactions with common causal agents of urinary and respiratory tract infections.
Collapse
Affiliation(s)
- Adeliia Giliazeva
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, Building 15, 01968, Senftenberg, Germany.
| | - Yaw Akosah
- Department of Molecular Pathobiology, College of Dentistry, New York University, 345 E. 24th St., 10010, New York, USA
| | - Jonas Noack
- Medipan GmbH, Computer Science, Ludwig-Erhard-Ring 3, 15827, Dahlewitz, Germany
| | - Ayslu Mardanova
- Department of Microbiology, Institute of Fundamental Medicine and Biology, Kazan (Volga region) Federal University, Kremlyovskaya 18, 420008, Kazan, Russia
| |
Collapse
|
8
|
Bedoya-Correa CM, Betancur-Giraldo S, Franco J, Arango-Santander S. Probiotic Effect of Streptococcus dentisani on Oral Pathogens: An In Vitro Study. Pathogens 2024; 13:351. [PMID: 38787203 PMCID: PMC11123734 DOI: 10.3390/pathogens13050351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Probiotics, including Streptococcus dentisani, have been proposed as an alternative to re-establish the ecology of the oral cavity and inhibit the formation of pathogenic biofilms. The main objective of this work was to assess the probiotic ability of S. dentisani against Streptococcus mutans, Streptococcus mitis, and Candida albicans biofilms. The ability of the strains to form a monospecies biofilm and the probiotic potential of S. dentisani using the competition, exclusion, and displacement strategies were determined. All strains were moderate biofilm producers. The ability of S. dentisani to compete with and exclude S. mutans and S. mitis during biofilm formation was not significant. However, S. dentisani significantly reduced pathologic streptococcal biofilms using the displacement strategy. Also S. dentisani reduced the formation of the C. albicans biofilm mainly through competition and displacement. In vitro, S. dentisani exhibited probiotic potential to reduce the formation of potentially pathogenic biofilms. Further investigation is required to understand the biofilm-inhibiting mechanisms exhibited by this probiotic strain.
Collapse
Affiliation(s)
- Claudia María Bedoya-Correa
- GIOM Group, Faculty of Dentistry, Universidad Cooperativa de Colombia, Medellin 055421, Colombia; (J.F.); (S.A.-S.)
| | | | - John Franco
- GIOM Group, Faculty of Dentistry, Universidad Cooperativa de Colombia, Medellin 055421, Colombia; (J.F.); (S.A.-S.)
- Salud y Sostenibilidad Group, School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia
| | - Santiago Arango-Santander
- GIOM Group, Faculty of Dentistry, Universidad Cooperativa de Colombia, Medellin 055421, Colombia; (J.F.); (S.A.-S.)
| |
Collapse
|
9
|
Liu H, Ma J, Yang P, Geng F, Li X, Lü J, Wang Y. Comparative analysis of biofilm characterization of probiotic Escherichia coli. Front Microbiol 2024; 15:1365562. [PMID: 38559351 PMCID: PMC10978722 DOI: 10.3389/fmicb.2024.1365562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Biofilms are thought to play a vital role in the beneficial effects of probiotic bacteria. However, the structure and function of probiotic biofilms are poorly understood. In this work, biofilms of Escherichia coli (E. coli) Nissle 1917 were investigated and compared with those of pathogenic and opportunistic strains (E. coli MG1655, O157:H7) using crystal violet assay, confocal laser scanning microscopy, scanning electron microscopy and FTIR microspectroscopy. The study revealed significant differences in the morphological structure, chemical composition, and spatial heterogeneity of the biofilm formed by the probiotic E. coli strain. In particular, the probiotic biofilm can secrete unique phospholipid components into the extracellular matrix. These findings provide new information on the morphology, architecture and chemical heterogeneity of probiotic biofilms. This information may help us to understand the beneficial effects of probiotics for various applications.
Collapse
Affiliation(s)
- Huiping Liu
- College of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jingfang Ma
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Pan Yang
- College of Pharmacy, Binzhou Medical University, Yantai, China
| | - Feng Geng
- College of Pharmacy, Binzhou Medical University, Yantai, China
| | - Xueling Li
- Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Junhong Lü
- College of Pharmacy, Binzhou Medical University, Yantai, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Yadi Wang
- College of Pharmacy, Binzhou Medical University, Yantai, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
10
|
Wang S, Mu L, Yu C, He Y, Hu X, Jiao Y, Xu Z, You S, Liu SL, Bao H. Microbial collaborations and conflicts: unraveling interactions in the gut ecosystem. Gut Microbes 2024; 16:2296603. [PMID: 38149632 PMCID: PMC10761165 DOI: 10.1080/19490976.2023.2296603] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/14/2023] [Indexed: 12/28/2023] Open
Abstract
The human gut microbiota constitutes a vast and complex community of microorganisms. The myriad of microorganisms present in the intestinal tract exhibits highly intricate interactions, which play a crucial role in maintaining the stability and balance of the gut microbial ecosystem. These interactions, in turn, influence the overall health of the host. The mammalian gut microbes have evolved a wide range of mechanisms to suppress or even eliminate their competitors for nutrients and space. Simultaneously, extensive cooperative interactions exist among different microbes to optimize resource utilization and enhance their own fitness. This review will focus on the competitive mechanisms among members of the gut microorganisms and discuss key modes of actions, including bacterial secretion systems, bacteriocins, membrane vesicles (MVs) etc. Additionally, we will summarize the current knowledge of the often-overlooked positive interactions within the gut microbiota, and showcase representative machineries. This information will serve as a reference for better understanding the complex interactions occurring within the mammalian gut environment. Understanding the interaction dynamics of competition and cooperation within the gut microbiota is crucial to unraveling the ecology of the mammalian gut microbial communities. Targeted interventions aimed at modulating these interactions may offer potential therapeutic strategies for disease conditions.
Collapse
Affiliation(s)
- Shuang Wang
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- Department of Biopharmaceutical Sciences (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lingyi Mu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chong Yu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Yuting He
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Xinliang Hu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Yanlei Jiao
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Ziqiong Xu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Shaohui You
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Shu-Lin Liu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| | - Hongxia Bao
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, State-Province Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD) College of Pharmacy, Harbin Medical University, Harbin, China
- Harbin Medical University-University of Calgary Cumming School of Medicine Centre for Infection and Genomics, Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Mukherjee S, Bhattacharjee S, Paul S, Nath S, Paul S. Biofilm-a Syntrophic Consortia of Microbial Cells: Boon or Bane? Appl Biochem Biotechnol 2023; 195:5583-5604. [PMID: 35829902 DOI: 10.1007/s12010-022-04075-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
Biofilm is the conglomeration of microbial cells which is associated with a surface. In the recent times, the study of biofilm has gained popularity and vivid research is being done to know about the effects of biofilm and that it consists of many organisms which are symbiotic in nature, some of which are human pathogens. Here, in this study, we have discussed about biofilms, its formation, relevance of its presence in the biosphere, and the possible remediations to cope up with its negative effects. Since removal of biofilm is difficult, emphasis has been made to suggest ways to prevent biofilm formation and also to devise ways to utilize biofilm in an economically and environment-friendly method.
Collapse
Affiliation(s)
- Susmita Mukherjee
- Department of Biotechnology, University of Engineering and Management, University Area, Plot No. III - B/5, New Town, Action Area - III, Kolkata, West Bengal, 700160, India
| | - Shreya Bhattacharjee
- Department of Biotechnology, University of Engineering and Management, University Area, Plot No. III - B/5, New Town, Action Area - III, Kolkata, West Bengal, 700160, India
| | - Sharanya Paul
- Department of Biotechnology, University of Engineering and Management, University Area, Plot No. III - B/5, New Town, Action Area - III, Kolkata, West Bengal, 700160, India
| | - Somava Nath
- Department of Biotechnology, University of Engineering and Management, University Area, Plot No. III - B/5, New Town, Action Area - III, Kolkata, West Bengal, 700160, India
| | - Sonali Paul
- Department of Biotechnology, University of Engineering and Management, University Area, Plot No. III - B/5, New Town, Action Area - III, Kolkata, West Bengal, 700160, India.
| |
Collapse
|
12
|
Emani SS, Kan A, Storms T, Bonanno S, Law J, Ray S, Joshi NS. Periplasmic stress contributes to a trade-off between protein secretion and cell growth in Escherichia coli Nissle 1917. Synth Biol (Oxf) 2023; 8:ysad013. [PMID: 37601821 PMCID: PMC10439730 DOI: 10.1093/synbio/ysad013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/29/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Maximizing protein secretion is an important target in the design of engineered living systems. In this paper, we characterize a trade-off between cell growth and per-cell protein secretion in the curli biofilm secretion system of Escherichia coli Nissle 1917. Initial characterization using 24-h continuous growth and protein production monitoring confirms decreased growth rates at high induction, leading to a local maximum in total protein production at intermediate induction. Propidium iodide (PI) staining at the endpoint indicates that cellular death is a dominant cause of growth reduction. Assaying variants with combinatorial constructs of inner and outer membrane secretion tags, we find that diminished growth at high production is specific to secretory variants associated with periplasmic stress mediated by outer membrane secretion and periplasmic accumulation of protein containing the outer membrane transport tag. RNA sequencing experiments indicate upregulation of known periplasmic stress response genes in the highly secreting variant, further implicating periplasmic stress in the growth-secretion trade-off. Overall, these results motivate additional strategies for optimizing total protein production and longevity of secretory engineered living systems Graphical Abstract.
Collapse
Affiliation(s)
| | - Anton Kan
- Department of Materials, ETH Zürich, Zürich, Switzerland
| | - Timothy Storms
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Shanna Bonanno
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Jade Law
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Sanhita Ray
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
| | - Neel S Joshi
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Yang G, Yang L, Zhou X. Inhibition of bacterial swimming by heparin binding of flagellin FliC from Escherichia coli strain Nissle 1917. Arch Microbiol 2023; 205:286. [PMID: 37452842 DOI: 10.1007/s00203-023-03622-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
Escherichia coli Nissle1917 (EcN) is a non-pathogenic probiotic strain widely used to maintain gut health, treat gastrointestinal disorders, and modulate the gut microbiome due to its anti-inflammatory and competitive exclusion effects against pathogenic bacteria. Heparin, abundant on intestinal mucosal surfaces, is a highly sulfated glycosaminoglycan primarily produced by mast cells. Currently, the interaction between EcN surface protein and heparin has remained elusive. In this study, the flagellin FliC responsible for EcN's movement was separated and characterized as a heparin binding protein by mass spectrometry (MS) analysis. The recombinant FliC protein, expressed by plasmid pET28a( +)-fliC, was further prepared to confirm the interaction between FliC and heparin. The results showed that heparin-Sepharose's ability to bind FliC was 48-fold higher than its ability to bind the negative control, bovine serum albumin (BSA). Neither the knockout of gene fliC nor the addition of heparin affects the growth of EcN, but both significantly inhibit the swimming of EcN. Adding 10 mg/ml heparin reduced the swimming diameter of the wild type and the complemented strain to 29-41% of the original, but that did not affect the swimming ability of the knockout strains. These results demonstrate that heparin interacts with EcN flagellin FliC and inhibits bacteria swimming. Exploring this interaction could improve our understanding of the relationship between hosts and microorganisms and provide a potential basis for disease treatment.
Collapse
Affiliation(s)
- Guixia Yang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Lingkang Yang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Xianxuan Zhou
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China.
| |
Collapse
|
14
|
Forsyth JH, Barron NL, Scott L, Watson BNJ, Chisnall MAW, Meaden S, van Houte S, Raymond B. Decolonizing drug-resistant E. coli with phage and probiotics: breaking the frequency-dependent dominance of residents. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001352. [PMID: 37418300 PMCID: PMC10433417 DOI: 10.1099/mic.0.001352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
Widespread antibiotic resistance in commensal bacteria creates a persistent challenge for human health. Resident drug-resistant microbes can prevent clinical interventions, colonize wounds post-surgery, pass resistance traits to pathogens or move to more harmful niches following routine interventions such as catheterization. Accelerating the removal of resistant bacteria or actively decolonizing particular lineages from hosts could therefore have a number of long-term benefits. However, removing resident bacteria via competition with probiotics, for example, poses a number of ecological challenges. Resident microbes are likely to have physiological and numerical advantages and competition based on bacteriocins or other secreted antagonists is expected to give advantages to the dominant partner, via positive frequency dependence. Since a narrow range of Escherichia coli genotypes (primarily those belonging to the clonal group ST131) cause a significant proportion of multidrug-resistant infections, this group presents a promising target for decolonization with bacteriophage, as narrow-host-range viral predation could lead to selective removal of particular genotypes. In this study we tested how a combination of an ST131-specific phage and competition from the well-known probiotic E. coli Nissle strain could displace E. coli ST131 under aerobic and anaerobic growth conditions in vitro. We showed that the addition of phage was able to break the frequency-dependent advantage of a numerically dominant ST131 isolate. Moreover, the addition of competing E. coli Nissle could improve the ability of phage to suppress ST131 by two orders of magnitude. Low-cost phage resistance evolved readily in these experiments and was not inhibited by the presence of a probiotic competitor. Nevertheless, combinations of phage and probiotic produced stable long-term suppression of ST131 over multiple transfers and under both aerobic and anaerobic growth conditions. Combinations of phage and probiotic therefore have real potential for accelerating the removal of drug-resistant commensal targets.
Collapse
Affiliation(s)
- Jessica H. Forsyth
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
- Present address: Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Natalie L. Barron
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | - Lucy Scott
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | | | | | - Sean Meaden
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
- Present address: Department of Biology, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Stineke van Houte
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | - Ben Raymond
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| |
Collapse
|
15
|
Zhu YX, You Y, Chen Z, Xu D, Yue W, Ma X, Jiang J, Wu W, Lin H, Shi J. Inorganic Nanosheet-Shielded Probiotics: A Self-Adaptable Oral Delivery System for Intestinal Disease Treatment. NANO LETTERS 2023; 23:4683-4692. [PMID: 36912868 DOI: 10.1021/acs.nanolett.3c00118] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The oral delivery of probiotics is commonly adopted for intestinal disease treatments in clinical settings; however, the probiotics suffer from a strong acidic attack in the gastric area and the low-efficiency intestinal colonization of naked probiotics. Coating living probiotics with synthetic materials has proven effective in enabling the adaption of bacteria to gastrointestinal environments, which, unfortunately, may shield the probiotics from initiating therapeutic responses. In this study, we report a copolymer-modified two-dimensional H-silicene nanomaterial (termed SiH@TPGS-PEI) that can facilitate probiotics to adapt to diverse gastrointestinal microenvironments on-demand. Briefly, SiH@TPGS-PEI electrostatically coated on the surface of probiotic bacteria helps to resist erosive destruction in the acidic stomach and spontaneously degrades by reacting with water to generate hydrogen, an anti-inflammatory gas in response to the neutral/weakly alkaline intestinal environment, thus exposing the probiotic bacteria for colitis amelioration. This strategy may shed new light on the development of intelligent self-adaptive materials.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| | - Yanling You
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zhixin Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Deliang Xu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wenwen Yue
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| | - Xinxin Ma
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P. R. China
| | - Junjie Jiang
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Wencheng Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
| | - Han Lin
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
| | - Jianlin Shi
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
16
|
Aljohani AM, El-Chami C, Alhubail M, Ledder RG, O’Neill CA, McBain AJ. Escherichia coli Nissle 1917 inhibits biofilm formation and mitigates virulence in Pseudomonas aeruginosa. Front Microbiol 2023; 14:1108273. [PMID: 36970701 PMCID: PMC10031955 DOI: 10.3389/fmicb.2023.1108273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/31/2023] [Indexed: 03/10/2023] Open
Abstract
In the quest for mitigators of bacterial virulence, cell-free supernatants (CFS) from 25 human commensal and associated bacteria were tested for activity against Pseudomonas aeruginosa. Among these, Escherichia coli Nissle 1917 CFS significantly inhibited biofilm formation and dispersed extant pseudomonas biofilms without inhibiting planktonic bacterial growth. eDNA was reduced in biofilms following exposure to E. coli Nissle CFS, as visualized by confocal microscopy. E. coli Nissle CFS also showed a significant protective effect in a Galleria mellonella-based larval virulence assay when administrated 24 h before challenge with the P. aeruginosa. No inhibitory effects against P. aeruginosa were observed for other tested E. coli strains. According to proteomic analysis, E. coli Nissle CFS downregulated the expression of several P. aeruginosa proteins involved in motility (Flagellar secretion chaperone FliSB, B-type flagellin fliC, Type IV pilus assembly ATPase PilB), and quorum sensing (acyl-homoserine lactone synthase lasI and HTH-type quorum-sensing regulator rhlR), which are associated with biofilm formation. Physicochemical characterization of the putative antibiofilm compound(s) indicates the involvement of heat-labile proteinaceous factors of greater than 30 kDa molecular size.
Collapse
Affiliation(s)
- Ahmad M. Aljohani
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Ministry of Education, Riyadh, Saudi Arabia
| | - Cecile El-Chami
- Division of Musculoskeletal and Dermatological Science, Faculty of Biology, Medicine and Health, School of Biological Science, The University of Manchester, Manchester, United Kingdom
| | - Muna Alhubail
- Division of Musculoskeletal and Dermatological Science, Faculty of Biology, Medicine and Health, School of Biological Science, The University of Manchester, Manchester, United Kingdom
| | - Ruth G. Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Catherine A. O’Neill
- Division of Musculoskeletal and Dermatological Science, Faculty of Biology, Medicine and Health, School of Biological Science, The University of Manchester, Manchester, United Kingdom
| | - Andrew J. McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- *Correspondence: Andrew J. McBain,
| |
Collapse
|
17
|
Marković KG, Grujović MŽ, Koraćević MG, Nikodijević DD, Milutinović MG, Semedo-Lemsaddek T, Djilas MD. Colicins and Microcins Produced by Enterobacteriaceae: Characterization, Mode of Action, and Putative Applications. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11825. [PMID: 36142096 PMCID: PMC9517006 DOI: 10.3390/ijerph191811825] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 06/15/2023]
Abstract
Enterobacteriaceae are widely present in many environments related to humans, including the human body and the food that they consume, from both plant or animal origin. Hence, they are considered relevant members of the gastrointestinal tract microbiota. On the other hand, these bacteria are also recognized as putative pathogens, able to impair human health and, in food, they are considered indicators for the microbiological quality and hygiene status of a production process. Nevertheless, beneficial properties have also been associated with Enterobacteriaceae, such as the ability to synthesize peptides and proteins, which can have a role in the structure of microbial communities. Among these antimicrobial molecules, those with higher molecular mass are called colicins, while those with lower molecular mass are named microcins. In recent years, some studies show an emphasis on molecules that can help control the development of pathogens. However, not enough data are available on this subject, especially related to microcins. Hence, this review gathers and summarizes current knowledge on colicins and microcins, potential usage in the treatment of pathogen-associated diseases and cancer, as well as putative applications in food biotechnology.
Collapse
Affiliation(s)
- Katarina G. Marković
- Institute for Information Technologies, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Mirjana Ž. Grujović
- Institute for Information Technologies, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Maja G. Koraćević
- Innovation Center, University of Niš, 18000 Niš, Serbia
- Faculty of Medicine, Department of Pharmacy, University of Niš, 18000 Niš, Serbia
| | - Danijela D. Nikodijević
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Milena G. Milutinović
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Teresa Semedo-Lemsaddek
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Milan D. Djilas
- Institute for Public Health of Vojvodina, Futoška 121, 21000 Novi Sad, Serbia
| |
Collapse
|
18
|
Gelfat I, Aqeel Y, Tremblay JM, Jaskiewicz JJ, Shrestha A, Lee JN, Hu S, Qian X, Magoun L, Sheoran A, Bedenice D, Giem C, Manjula-Basavanna A, Pulsifer AR, Tu HX, Li X, Minus ML, Osburne MS, Tzipori S, Shoemaker CB, Leong JM, Joshi NS. Single domain antibodies against enteric pathogen virulence factors are active as curli fiber fusions on probiotic E. coli Nissle 1917. PLoS Pathog 2022; 18:e1010713. [PMID: 36107831 PMCID: PMC9477280 DOI: 10.1371/journal.ppat.1010713] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/29/2022] [Indexed: 11/18/2022] Open
Abstract
Enteric microbial pathogens, including Escherichia coli, Shigella and Cryptosporidium species, take a particularly heavy toll in low-income countries and are highly associated with infant mortality. We describe here a means to display anti-infective agents on the surface of a probiotic bacterium. Because of their stability and versatility, VHHs, the variable domains of camelid heavy-chain-only antibodies, have potential as components of novel agents to treat or prevent enteric infectious disease. We isolated and characterized VHHs targeting several enteropathogenic E. coli (EPEC) virulence factors: flagellin (Fla), which is required for bacterial motility and promotes colonization; both intimin and the translocated intimin receptor (Tir), which together play key roles in attachment to enterocytes; and E. coli secreted protein A (EspA), an essential component of the type III secretion system (T3SS) that is required for virulence. Several VHHs that recognize Fla, intimin, or Tir blocked function in vitro. The probiotic strain E. coli Nissle 1917 (EcN) produces on the bacterial surface curli fibers, which are the major proteinaceous component of E. coli biofilms. A subset of Fla-, intimin-, or Tir-binding VHHs, as well as VHHs that recognize either a T3SS of another important bacterial pathogen (Shigella flexneri), a soluble bacterial toxin (Shiga toxin or Clostridioides difficile toxin TcdA), or a major surface antigen of an important eukaryotic pathogen (Cryptosporidium parvum) were fused to CsgA, the major curli fiber subunit. Scanning electron micrographs indicated CsgA-VHH fusions were assembled into curli fibers on the EcN surface, and Congo Red binding indicated that these recombinant curli fibers were produced at high levels. Ectopic production of these VHHs conferred on EcN the cognate binding activity and, in the case of anti-Shiga toxin, was neutralizing. Taken together, these results demonstrate the potential of the curli-based pathogen sequestration strategy described herein and contribute to the development of novel VHH-based gut therapeutics.
Collapse
Affiliation(s)
- Ilia Gelfat
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts, United States of America
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Yousuf Aqeel
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Jacqueline M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Justyna J. Jaskiewicz
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Anishma Shrestha
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - James N. Lee
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Shenglan Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Xi Qian
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Loranne Magoun
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Abhineet Sheoran
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Daniela Bedenice
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Colter Giem
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Avinash Manjula-Basavanna
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Amanda R. Pulsifer
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Hann X. Tu
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Xiaoli Li
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, United States of America
| | - Marilyn L. Minus
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, United States of America
| | - Marcia S. Osburne
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Tufts University, Medford, Massachusetts, United States of America
| | - Neel S. Joshi
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| |
Collapse
|
19
|
Azam MW, Khan AU. CRISPRi -mediated suppression of E. coli Nissle 1917 virulence factors: A strategy for creating an engineered probiotic using csgD gene suppression. Front Nutr 2022; 9:938989. [PMID: 35978963 PMCID: PMC9376613 DOI: 10.3389/fnut.2022.938989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background Biofilm formation is a complex phenomenon, and it is the causative agent of several human infections. Bacterial amyloids are involved in biofilm formation leading to infection persistence. Due to antibiotic resistance, their treatment is a great challenge for physicians. Probiotics, especially E. coli Nissle 1917 (EcN), are used to treat human intestinal disorders and ulcerative colitis. It also expresses virulence factors associated with biofilm and amyloid formation. EcN produces biofilm equivalent to the pathogenic UPEC strains. Methods CRISPRi was used to create the knockdown mutants of the csgD gene (csgD-KD). The qRT-PCR was performed to assess the expression of the csgD gene in csgD-KD cells. The csgD-KD cells were also evaluated for the expression of csgA, csgB, fimA, fimH, ompR, luxS, and bolA genes. The gene expression data obtained was further confirmed by spectroscopic, microscopic, and other assays to validate our study. Results CRISPRi-mediated knockdown of csgD gene shows reduction in curli amyloid formation, biofilm formation, and suppression of genes (csgA, csgB, fimA, fimH, ompR, bolA, and luxS) involved in virulence factors production. Conclusion Curli amyloid fibers and fimbriae fibers play a critical role in biofilm formation leading to pathogenicity. CsgD protein is the master regulator of curli synthesis in E. coli. Hence, curli amyloid inhibition through the csgD gene may be used to improve the EcN and different probiotic strains by suppressing virulence factors.
Collapse
Affiliation(s)
- Mohd W Azam
- Medical Microbiology and Molecular Biology Lab, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Asad U Khan
- Medical Microbiology and Molecular Biology Lab, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
20
|
Gaio D, DeMaere MZ, Anantanawat K, Eamens GJ, Falconer L, Chapman TA, Djordjevic S, Darling AE. Phylogenetic diversity analysis of shotgun metagenomic reads describes gut microbiome development and treatment effects in the post-weaned pig. PLoS One 2022; 17:e0270372. [PMID: 35749534 PMCID: PMC9232140 DOI: 10.1371/journal.pone.0270372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Intensive farming practices can increase exposure of animals to infectious agents against which antibiotics are used. Orally administered antibiotics are well known to cause dysbiosis. To counteract dysbiotic effects, numerous studies in the past two decades sought to understand whether probiotics are a valid tool to help re-establish a healthy gut microbial community after antibiotic treatment. Although dysbiotic effects of antibiotics are well investigated, little is known about the effects of intramuscular antibiotic treatment on the gut microbiome and a few studies attempted to study treatment effects using phylogenetic diversity analysis techniques. In this study we sought to determine the effects of two probiotic- and one intramuscularly administered antibiotic treatment on the developing gut microbiome of post-weaning piglets between their 3rd and 9th week of life. Shotgun metagenomic sequences from over 800 faecal time-series samples derived from 126 post-weaning piglets and 42 sows were analysed in a phylogenetic framework. Differences between individual hosts such as breed, litter, and age, were found to be important contributors to variation in the community composition. Host age was the dominant factor in shaping the gut microbiota of piglets after weaning. The post-weaning pig gut microbiome appeared to follow a highly structured developmental program with characteristic post-weaning changes that can distinguish hosts that were born as little as two days apart in the second month of life. Treatment effects of the antibiotic and probiotic treatments were found but were subtle and included a higher representation of Mollicutes associated with intramuscular antibiotic treatment, and an increase of Lactobacillus associated with probiotic treatment. The discovery of correlations between experimental factors and microbial community composition is more commonly addressed with OTU-based methods and rarely analysed via phylogenetic diversity measures. The latter method, although less intuitive than the former, suffers less from library size normalization biases, and it proved to be instrumental in this study for the discovery of correlations between microbiome composition and host-, and treatment factors.
Collapse
Affiliation(s)
- Daniela Gaio
- iThree Institute, University of Technology Sydney, Ultimo, Australia
- * E-mail:
| | | | - Kay Anantanawat
- iThree Institute, University of Technology Sydney, Ultimo, Australia
| | - Graeme J. Eamens
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, Australia
| | - Linda Falconer
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, Australia
| | - Toni A. Chapman
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, Australia
| | - Steven Djordjevic
- iThree Institute, University of Technology Sydney, Ultimo, Australia
| | - Aaron E. Darling
- iThree Institute, University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
21
|
Porter SB, Johnston BD, Kisiela D, Clabots C, Sokurenko EV, Johnson JR. Bacteriophage Cocktail and Microcin-Producing Probiotic Escherichia coli Protect Mice Against Gut Colonization With Multidrug-Resistant Escherichia coli Sequence Type 131. Front Microbiol 2022; 13:887799. [PMID: 35547133 PMCID: PMC9082999 DOI: 10.3389/fmicb.2022.887799] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Non-antibiotic measures are needed to reduce the rate of infections due to multidrug-resistant organisms (MDROs), including by eliminating the commensal reservoir that underlies such strains’ dissemination and leads to recurrent infections. Here, we tested a cocktail of pre-selected bacteriophages and an engineered microcin C7-producing probiotic Escherichia coli Nissle-1917 strain for their ability to reduce gut colonization by an E. coli strain from sequence type 131 (ST131)-H30R, which is the major clonal group of MDROs among extraintestinal clinical E. coli isolates. Although the bacteriophage cocktail was highly effective against ST131-H30R strains both in vitro and in a murine model of subcutaneous sepsis, it was only weakly and transiently effective against gut colonization by the target ST131-H30R strain (0.5 log10 decrease on d + 1: p < 0.001; no significant effect on d + 4 and beyond). The probiotic strain, while also highly active against ST131-H30R in vitro, was ineffective against ST131-H30R gut colonization despite its abundant presence in feces. Nonetheless, despite failing as decolonizing agents when administered separately, when co-administered the bacteriophage cocktail and probiotic strain exhibited striking synergy against ST131-H30R gut colonization. This combinatory effect was most pronounced on d + 1 (3.3 log10 target strain decrease: p < 0.001), and persisted until d + 7 (0.5 log10 decrease; p < 0.02.). Although by d + 10 the ST131-H30R load was fully restored, these findings provide proof of concept for combined bacteriophage-plus-probiotic administration to reduce or, possibly, to prevent gut colonization with MDROs in high-risk individuals.
Collapse
Affiliation(s)
- Stephen B Porter
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States
| | - Brian D Johnston
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Dagmara Kisiela
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Connie Clabots
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States
| | - Evgeni V Sokurenko
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - James R Johnson
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States.,Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
22
|
Foster-Nyarko E, Pallen MJ. The microbial ecology of Escherichia coli in the vertebrate gut. FEMS Microbiol Rev 2022; 46:fuac008. [PMID: 35134909 PMCID: PMC9075585 DOI: 10.1093/femsre/fuac008] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli has a rich history as biology's 'rock star', driving advances across many fields. In the wild, E. coli resides innocuously in the gut of humans and animals but is also a versatile pathogen commonly associated with intestinal and extraintestinal infections and antimicrobial resistance-including large foodborne outbreaks such as the one that swept across Europe in 2011, killing 54 individuals and causing approximately 4000 infections and 900 cases of haemolytic uraemic syndrome. Given that most E. coli are harmless gut colonizers, an important ecological question plaguing microbiologists is what makes E. coli an occasionally devastating pathogen? To address this question requires an enhanced understanding of the ecology of the organism as a commensal. Here, we review how our knowledge of the ecology and within-host diversity of this organism in the vertebrate gut has progressed in the 137 years since E. coli was first described. We also review current approaches to the study of within-host bacterial diversity. In closing, we discuss some of the outstanding questions yet to be addressed and prospects for future research.
Collapse
Affiliation(s)
- Ebenezer Foster-Nyarko
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Mark J Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TU, United Kingdom
| |
Collapse
|
23
|
Werneburg GT. Catheter-Associated Urinary Tract Infections: Current Challenges and Future Prospects. Res Rep Urol 2022; 14:109-133. [PMID: 35402319 PMCID: PMC8992741 DOI: 10.2147/rru.s273663] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/27/2022] [Indexed: 12/15/2022] Open
Abstract
Catheter-associated urinary tract infection (CAUTI) is the most common healthcare-associated infection and cause of secondary bloodstream infections. Despite many advances in diagnosis, prevention and treatment, CAUTI remains a severe healthcare burden, and antibiotic resistance rates are alarmingly high. In this review, current CAUTI management paradigms and challenges are discussed, followed by future prospects as they relate to the diagnosis, prevention, and treatment. Clinical and translational evidence will be evaluated, as will key basic science studies that underlie preventive and therapeutic approaches. Novel diagnostic strategies and treatment decision aids under development will decrease the time to diagnosis and improve antibiotic accuracy and stewardship. These include several classes of biomarkers often coupled with artificial intelligence algorithms, cell-free DNA, and others. New preventive strategies including catheter coatings and materials, vaccination, and bacterial interference are being developed and investigated. The antibiotic pipeline remains insufficient, and new strategies for the identification of new classes of antibiotics, and rational design of small molecule inhibitor alternatives, are under development for CAUTI treatment.
Collapse
Affiliation(s)
- Glenn T Werneburg
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
24
|
Manzhalii E, Moyseyenko V, Kondratiuk V, Molochek N, Falalyeyeva T, Kobyliak N. Effect of a specific Escherichia coli Nissle 1917 strain on minimal/mild hepatic encephalopathy treatment. World J Hepatol 2022; 14:634-646. [PMID: 35582294 PMCID: PMC9055191 DOI: 10.4254/wjh.v14.i3.634] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/01/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatic encephalopathy (HE) can be considered a result of dysregulated gut-liver-brain axis function, where cognitive impairment can be reversed or prevented by the beneficial effects induced by "gut-centric" therapies, such as the administration of nonabsorbable disaccharides, nonabsorbable antibiotics, probiotics and prebiotics. AIM To assess the short-term efficacy and safety of the probiotic Escherichia coli Nissle (EcN) 1917 strain compared to lactulose and rifaximin in patients with minimal/mild HE. METHODS From January 2017 to March 2020, a total of 45 patients with HE were enrolled in this prospective, single-centre, open-label, randomized study. Participants were randomly assigned at a ratio of 1:1:1 to one of the treatment groups: The EcN group (n = 15), lactulose group (n = 15) or rifaximin group (n = 15) for a 1 mo intervention period. The main primary outcomes of the study were changes in serum ammonia and Stroop test score. The secondary outcomes were markers of a chronic systemic inflammatory response (ІL-6, ІL-8, and IFN-γ) and bacteriology of the stool flora evaluated by specialized nonculture techniques after a 1 mo intervention period. RESULTS Patients who were given rifaximin or EcN showed a more significant reduction in serum ammonia and normalization of Bifidobacteria and Lactobacilli abundance compared to the lactulose group. However, the most pronounced restoration of the symbiotic microflora was associated with EcN administration and characterized by the absence of E. coli with altered properties and pathogenic enterobacteria in patient faeces. In the primary outcome analysis, improvements in the Stroop test parameters in all intervention groups were observed. Moreover, EcN-treated patients performed 15% faster on the Stroop test than the lactulose group patients (P = 0.017). Both EcN and rifaximin produced similar significant reductions in the proinflammatory cytokines INF-γ, IL-6 and IL-8. EcN was more efficient than lactulose in reducing proinflammatory cytokine levels. CONCLUSION The use of the probiotic EcN strain was safe and quite efficient for HE treatment. The probiotic reduced the ammonia content and the level of serum proinflammatory cytokines, normalized the gut microbiota composition and improved the cognitive function of patients with HE. The application of the EcN strain was more effective than lactulose treatment.
Collapse
Affiliation(s)
- Elina Manzhalii
- Department of Propedeutics of Internal Medicine, Bogomolets National Medical University, Kyiv 01601, Ukraine.
| | - Valentyna Moyseyenko
- Department of Propedeutics of Internal Medicine, Bogomolets National Medical University, Kyiv 01601, Ukraine
| | - Vitalii Kondratiuk
- Department of Propedeutics of Internal Medicine, Bogomolets National Medical University, Kyiv 01601, Ukraine
| | - Nataliia Molochek
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
- Department of Pediatrics, Bogomolets National Medical University, Kyiv 01601, Ukraine
| | - Tetyana Falalyeyeva
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
- Department of Scientific, Medical Laboratory CSD, Kyiv 01004, Ukraine
| | - Nazarii Kobyliak
- Department of Endocrinology, Bogomolets National Medical University, Kyiv 01601, Ukraine
- Department of Scientific, Medical Laboratory CSD, Kyiv 01004, Ukraine.
| |
Collapse
|
25
|
The Effect of Tannin-Rich Witch Hazel on Growth of Probiotic Lactobacillus plantarum. Antibiotics (Basel) 2022; 11:antibiotics11030395. [PMID: 35326857 PMCID: PMC8944479 DOI: 10.3390/antibiotics11030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/09/2022] [Accepted: 03/13/2022] [Indexed: 11/23/2022] Open
Abstract
Probiotic bacteria help maintain microbiome homeostasis and promote gut health. Maintaining the competitive advantage of the probiotics over pathogenic bacteria is a challenge, as they are part of the gut microbiome that is continuously exposed to digestive and nutritional changes and various stressors. Witch hazel that is rich in hamamelitannin (WH, whISOBAXTM) is an inhibitor of growth and virulence of pathogenic bacteria. To test for its effect on probiotic bacteria, WH was tested on the growth and biofilm formation of a commercially available probiotic Lactobacillus plantarum PS128. As these bacteria are aerotolerant, the experiments were carried out aerobically and in nutritionally inadequate/poor (nutrient broth) or adequate/rich (MRS broth) conditions. Interestingly, despite its negative effect on the growth and biofilm formation of pathogenic bacteria such as Staphylococcus epidermidis, WH promotes the growth of the probiotic bacteria in a nutritionally inadequate environment while maintaining their growth under a nutritionally rich environment. In the absence of WH, no significant biofilm is formed on the surfaces tested (polystyrene and alginate), but in the presence of WH, biofilm formation was significantly enhanced. These results indicate that WH may thus be used to enhance the growth and survival of probiotics.
Collapse
|
26
|
Kenneally C, Murphy CP, Sleator RD, Culligan EP. The Urinary Microbiome and Biological Therapeutics: Novel Therapies For Urinary Tract Infections. Microbiol Res 2022; 259:127010. [DOI: 10.1016/j.micres.2022.127010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
|
27
|
Yadav MK, Yadav P, Dhiman M, Tewari S, Tiwari SK. Plantaricin LD1 purified from Lactobacillus plantarum LD1 inhibits biofilm formation of Enterococcus faecalis ATCC 29212 in tooth model. Lett Appl Microbiol 2022; 75:623-631. [PMID: 35146783 DOI: 10.1111/lam.13668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/14/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022]
Abstract
Plantaricin LD1 was purified to homogeneity using activity-guided chromatography. Enterococcus faecalis ATCC 29212 was found to be sensitive to plantaricin LD1 showing 13 ± 0.21 mm zone of growth inhibition. The minimum inhibitory concentration (MIC) was found to be 50 µg ml-1 against Ent. faecalis ATCC 29212. The in vitro biofilm formation by Ent. faecalis ATCC 29212 was observed which was completely inhibited in the presence of bacteriocin. Similarly, biofilm formation was also observed on the teeth surface showing purple colour whereas, treated-teeth were clean indicated no biofilm formation. Further, untreated cells of Ent. faecalis ATCC 29212 were found normal and plantaricin LD1-treated cells were ruptured seen under light microscope suggesting killing of target cells. These findings have proven the initial leads for antimicrobial and anti-biofilm activity of plantaricin LD1 against Ent. faecalis and its possible application for the treatment of endodontic diseases.
Collapse
Affiliation(s)
- Manoj Kumar Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Priyanka Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Meenu Dhiman
- Department of Conservative Dentistry and Endodontics, Post Graduate Institute of Dental Sciences, Rohtak-124001, Haryana, India
| | - Sanjay Tewari
- Department of Conservative Dentistry and Endodontics, Post Graduate Institute of Dental Sciences, Rohtak-124001, Haryana, India
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| |
Collapse
|
28
|
Mourand G, Paboeuf F, Grippon P, Lucas P, Bougeard S, Denamur E, Kempf I. Impact of Escherichia coli probiotic strains ED1a and Nissle 1917 on the excretion and gut carriage of extended-spectrum beta-lactamase-producing E. coli in pigs. Vet Anim Sci 2021; 14:100217. [PMID: 34825108 PMCID: PMC8604716 DOI: 10.1016/j.vas.2021.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 11/05/2022] Open
Abstract
The inoculated cefotaxime-resistant E. coli was a good pig gut colonizer. Probiotics could not reduce faecal excretion of resistant E. coli in inoculated pigs. Resistant E. coli titers were lower in digestive tracts of the probiotic-treated pigs. No transfer of the blaCTX−M-1 gene was detected.
We evaluated the impact of the administration of two Escherichia coli probiotic strains (ED1a and Nissle 1917) to pigs on the gut carriage or shedding of extended-spectrum beta-lactamase-producing E. coli. The probiotics were given to four sows from 12 days before farrowing to the weaning day, and to the 23 piglets (infected treated group (IPro)) from birth to the age of 49 days. Four other sows and their 24 piglets (infected non-treated group (INT)) did not receive the probiotics. IPro and INT piglets (n = 47) were orally inoculated with the strain E. coli 17–348F-RifR carrying the blaCTX−M-1 gene and resistant to rifampicin. Cefotaxime-resistant (CTXR) E. coli and rifampicin-resistant (RifR) E. coli were cultured and excretion of probiotics was studied using PCR on individual faecal and post-mortem samples, and from manure collected after the challenge with resistant E. coli. CTXR and RifRE.coli isolates were characterized to detect transfer of the blaCTX−M-1 to other strains.. Overall, there was no significant reduction in faecal excretion of CTXR and RifRE. coli in IPro pigs compared with INT pigs, although the CTXR and RifRE. coli titres were slightly, but significantly lower in the colon, caecum and rectum at post mortem. Excretion of the probiotics decreased with age, but Nissle 1917 was detected in most pigs at post-mortem. No transfer of the blaCTX−M-1 gene to probiotic and other E. coli strains was detected. In conclusion, in our experimental conditions, the used probiotics did not reduce shedding of the challenge strain.
Collapse
Affiliation(s)
| | - Frédéric Paboeuf
- ANSES, Laboratoire de Ploufragan-Plouzané-Niort, Ploufragan, France
| | - Pauline Grippon
- ANSES, Laboratoire de Ploufragan-Plouzané-Niort, Ploufragan, France
| | - Pierrick Lucas
- ANSES, Laboratoire de Ploufragan-Plouzané-Niort, Ploufragan, France
| | | | - Erick Denamur
- Université de Paris, IAME, INSERM UMR 1137, Paris, France.,APHP, Hôpital Bichat Claude-Bernard, Laboratoire de Génétique Moléculaire, Paris, France
| | - Isabelle Kempf
- ANSES, Laboratoire de Ploufragan-Plouzané-Niort, Ploufragan, France
| |
Collapse
|
29
|
Guli M, Winarsih S, Barlianto W, Illiandri O, Sumarno SP. Mechanism of Lactobacillus reuteri Probiotic in Increasing Intestinal Mucosal Immune System. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Probiotics are defined as live microorganisms which, when consumed in adequate quantities as food ingredients, provide health benefits to the host. Lactobacillus, Bifidobacterium, and Saccharomyces, are three probiotics that are intensively used as probiotics in humans and animals. Probiotics have beneficial effects on health when given adequate amounts. The concept of probiotics on human health, namely modulating the gut microbiota and its effect on the host. Probiotics play an important role in maintaining intestinal integrity through a number of different interactions, including changes in cytokine expression in the mucosa. Probiotics compete with intestinal pathogens for mucosal receptors, thereby increasing interepithelial resistance. Probiotics such as Lactobacillus casei sp GG strain was used as a prophylaxis that could increase the expression of epithelial mucin, thereby reducing the translocation of pathogenic bacteria. Abnormal local immune response is characterized by decreased secretion of IgA, thus allowing enterocyte attachment and local translocation of bacterial antigens, which are the main stimulation of pathological events. Colonic stasis can promote the growth of pathogenic bacteria which allows malignant porin bacterial strains to thrive. The gut microbiota has a major influence on human health. The microbial population has an important role in the host, such as the metabolic activity of probiotics producing energy and nutrient absorption, developing the host immune system, and preventing colonization and infection of pathogens. Lactobacillus reuteri is a hetero-fermentative bacterium that lives in the digestive tract of humans. L. reuteri has been used to treat infant necrotizing pseudomembrane. In this paper, the mechanism of L reuteri to increase host immunological response will be reviewed.
Collapse
|
30
|
Seco EM, Fernández LÁ. Efficient markerless integration of genes in the chromosome of probiotic E. coli Nissle 1917 by bacterial conjugation. Microb Biotechnol 2021; 15:1374-1391. [PMID: 34755474 PMCID: PMC9049610 DOI: 10.1111/1751-7915.13967] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 01/30/2023] Open
Abstract
The probiotic strain Escherichia coli Nissle 1917 (EcN) is a common bacterial chassis in synthetic biology developments for therapeutic applications given its long track record of safe administration in humans. Chromosomal integration of the genes of interest (GOIs) in the engineered bacterium offers significant advantages in genetic stability and to control gene dose, but common methodologies relying on the transformation of EcN are inefficient. In this work, we implement in EcN the use of bacterial conjugation in combination with markerless genome engineering to efficiently insert multiple GOIs at different loci of EcN chromosome, leaving no antibiotic resistance genes, vector sequences or scars in the modified bacterium. The resolution of cointegrants that leads to markerless insertion of the GOIs requires expression of I-SceI endonuclease and its efficiency is enhanced by λ Red proteins. We show the potential of this strategy by integrating different genes encoding fluorescent and bioluminescent reporters (i.e. GFP, mKate2, luxCDABE) both individually and sequentially. We also demonstrate its application for gene deletions in EcN (ΔflhDC) and to replace the endogenous regulation of chromosomal locus (i.e. flhDC) by heterologous regulatory elements (e.g. tetR-Ptet) in order to have an ectopic control of gene expression in EcN with an external inducer to alter bacterial behaviour (e.g. flagellar motility). Whole-genome sequencing confirmed the introduction of the designed modifications without off-target alterations in the genome. This straightforward approach accelerates the generation of multiple modifications in EcN chromosome for the generation of living bacterial therapeutics.
Collapse
Affiliation(s)
- Elena M Seco
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus UAM Cantoblanco, Madrid, 28049, Spain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus UAM Cantoblanco, Madrid, 28049, Spain
| |
Collapse
|
31
|
Al-Sarraj F. The effect of antibiotics and photodynamic therapy on extended-spectrum beta-lactamase (ESBL) positive of Escherichia coli and Klebsiella pneumoniae in urothelial cells. Saudi J Biol Sci 2021; 28:5561-5567. [PMID: 34588866 PMCID: PMC8459124 DOI: 10.1016/j.sjbs.2021.05.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 01/10/2023] Open
Abstract
Background/aim Urinary tract infections are commonly caused by the bacteria Escherichia coli and Klebsiella pneumoniae (UTI). The emergence of extended-spectrum -lactamase (ESBL)-producing bacteria strains has made UTI treatment more difficult. Materials and methods The aim of this study was to characterize E. coli and K. pneumoniae strains' cytotoxic effects, antibiotic sensitivity, interaction with urothelial cells, and reaction to photodynamic therapy. Results As demonstrated by the higher number of colonies formed, the ESBL + E. coli and K. Pneumonia showed a higher degree of binding with human urothelial cells. With the urothelial cells, K. Pneumonia had the highest binding ability. The cytotoxicity of non-ESBL generating E. coli and K. Pneumonia, on the other hand, was higher. With longer incubation, the discrepancy between the cytotoxic effects of non-ESBL producer and ESBL + E. coli decreased. K. Pneumonia was the opposite. The concentration of ESBL-negative E. coli was easily decreased by photodynamic therapy; however, after a two-hour incubation period, the number of E. coli ESBL + colonies increased from 124 percent to 294 percent. Conclusion With the duration of the incubation period, the number of non-ESBL-producing K. Pneumonia increased. Even with longer incubation times, the number of K. Pneumonia ESBL + colonies decreased, contrary to expectations. The findings show that the two bacterial species differed in terms of cytotoxicity, interaction with urothelial cells, and photodynamic therapy response.
Collapse
Affiliation(s)
- Faisal Al-Sarraj
- Department of Biological Sciences, Faculty of Science, P.O. Box 80203, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
32
|
Serra DO, Hengge R. Bacterial Multicellularity: The Biology of Escherichia coli Building Large-Scale Biofilm Communities. Annu Rev Microbiol 2021; 75:269-290. [PMID: 34343018 DOI: 10.1146/annurev-micro-031921-055801] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biofilms are a widespread multicellular form of bacterial life. The spatial structure and emergent properties of these communities depend on a polymeric extracellular matrix architecture that is orders of magnitude larger than the cells that build it. Using as a model the wrinkly macrocolony biofilms of Escherichia coli, which contain amyloid curli fibers and phosphoethanolamine (pEtN)-modified cellulose as matrix components, we summarize here the structure, building, and function of this large-scale matrix architecture. Based on different sigma and other transcription factors as well as second messengers, the underlying regulatory network reflects the fundamental trade-off between growth and survival. It controls matrix production spatially in response to long-range chemical gradients, but it also generates distinct patterns of short-range matrix heterogeneity that are crucial for tissue-like elasticity and macroscopic morphogenesis. Overall, these biofilms confer protection and a potential for homeostasis, thereby reducing maintenance energy, which makes multicellularity an emergent property of life itself. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Diego O Serra
- Instituto de Biología Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario (UNR), 2000 Rosario, Argentina
| | - Regine Hengge
- Institut für Biologie/Mikrobiologie, Humboldt-Universität zu Berlin, 10115 Berlin, Germany;
| |
Collapse
|
33
|
Zeng XY, Li M. Looking into key bacterial proteins involved in gut dysbiosis. World J Methodol 2021; 11:130-143. [PMID: 34322365 PMCID: PMC8299906 DOI: 10.5662/wjm.v11.i4.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/11/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal microbiota plays a pivotal role in health and has been linked to many diseases. With the rapid accumulation of pyrosequencing data of the bacterial composition, the causal-effect relationship between specific dysbiosis features and diseases is now being explored. The aim of this review is to describe the key functional bacterial proteins and antigens in the context of dysbiosis related-diseases. We subjectively classify the key functional proteins into two categories: Primary key proteins and secondary key proteins. The primary key proteins mainly act by themselves and include biofilm inhibitors, toxin degraders, oncogene degraders, adipose metabolism modulators, anti-inflammatory peptides, bacteriocins, host cell regulators, adhesion and invasion molecules, and intestinal barrier regulators. The secondary key proteins mainly act by eliciting host immune responses and include flagellin, outer membrane proteins, and other autoantibody-related antigens. Knowledge of key bacterial proteins is limited compared to the rich microbiome data. Understanding and focusing on these key proteins will pave the way for future mechanistic level cause-effect studies of gut dysbiosis and diseases.
Collapse
Affiliation(s)
- Xin-Yu Zeng
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ming Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumors, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
34
|
Parker MT, Kunjapur AM. Deployment of Engineered Microbes: Contributions to the Bioeconomy and Considerations for Biosecurity. Health Secur 2021; 18:278-296. [PMID: 32816583 DOI: 10.1089/hs.2020.0010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Engineering at microscopic scales has an immense effect on the modern bioeconomy. Microbes contribute to such disparate markets as chemical manufacturing, fuel production, crop optimization, and pharmaceutical synthesis, to name a few. Due to new and emerging synthetic biology technologies, and the sophistication and control afforded by them, we are on the brink of deploying engineered microbes to not only enhance traditional applications but also to introduce these microbes to sectors, contexts, and formats not previously attempted. In microbially managed medicine, microbial engineering holds promise for increasing efficacy, improving tissue penetration, and sustaining treatment. In the environment, the most effective areas for deployment are in the management of crops and protection of ecosystems. However, caution is warranted before introducing engineered organisms to new environments where they may proliferate without control and could cause unforeseen effects. We summarize ideas and data that can inform identification and assessment of the risks that these tools present to ensure that realistic hazards are described and unrealistic ones do not hinder advancement. Further, because modes of containment are crucial complements to deployment, we describe the state of the art in microbial biocontainment strategies, current gaps, and how these gaps might be addressed through technological advances in synthetic engineering. Collectively, this work highlights engineered microbes as a foundational and expanding facet of the bioeconomy, projects their utility in upcoming deployments outside the laboratory, and identifies knowns and unknowns that will be necessary considerations and points of focus in this endeavor.
Collapse
Affiliation(s)
- Michael T Parker
- Michael T. Parker, PhD, is an Assistant Dean, Office of the Dean, Georgetown University, Washington, DC. Aditya M. Kunjapur, PhD, is an Assistant Professor, Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Aditya M Kunjapur
- Michael T. Parker, PhD, is an Assistant Dean, Office of the Dean, Georgetown University, Washington, DC. Aditya M. Kunjapur, PhD, is an Assistant Professor, Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| |
Collapse
|
35
|
Park K, Park S, Nagappan A, Ray N, Kim J, Yoon S, Moon Y. Probiotic Escherichia coli Ameliorates Antibiotic-Associated Anxiety Responses in Mice. Nutrients 2021; 13:nu13030811. [PMID: 33804493 PMCID: PMC8000835 DOI: 10.3390/nu13030811] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 01/14/2023] Open
Abstract
Despite the beneficial actions of antibiotics against bacterial infections, the use of antibiotics is a crucial etiological factor influencing microbial dysbiosis-associated adverse outcomes in human health. Based on the assumption that gut microbial dysbiosis can provoke behavioral or psychological disorders, the present study evaluated anxiety-linked behavioral changes in a mouse model of streptomycin-induced dysbiosis. Measuring anxiety-like behavior using the light-dark box and elevated plus maze tests indicated that streptomycin treatment caused acute anxiety in mice. As an intervention for dysbiosis-associated distress, the probiotic strain Escherichia coli Nissle 1917 (EcN) was evaluated for its effects on streptomycin-induced behavioral changes in mice. EcN supplementation persistently ameliorated anxiety responses in mice with streptomycin-induced dysbiosis. As an outcome of anxiety, body weight changes were marginally affected by antibiotic treatment. However, mice supplemented with EcN displayed acute retardation of body weight gain, since EcN is known to reduce food intake and increase energy expenditure. Taken together, EcN treatment prominently counteracted streptomycin-induced anxiety in mice, with the metabolically beneficial retardation of body weight gain. The present model simulates psychological disorders in antibiotic users. As a promising intervention, EcN treatment can facilitate psychological relief under conditions of dysbiotic stress by blocking the pathologic gut-brain circuit.
Collapse
Affiliation(s)
- Kiwoong Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
- Department of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Suhyeon Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
- Department of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Arulkumar Nagappan
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
| | - Navin Ray
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
| | - Sik Yoon
- Department of Anatomy, College of Medicine, Pusan National University, Yangsan 50612, Korea;
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Biomedical Research Institute, Pusan National University, Yangsan 50612, Korea; (K.P.); (S.P.); (A.N.); (N.R.); (J.K.)
- Department of Medicine, Pusan National University, Yangsan 50612, Korea
- Correspondence: ; Tel.: +82-51-510-8094
| |
Collapse
|
36
|
Baishya J, Bisht K, Rimbey JN, Yihunie KD, Islam S, Al Mahmud H, Waller JE, Wakeman CA. The Impact of Intraspecies and Interspecies Bacterial Interactions on Disease Outcome. Pathogens 2021; 10:96. [PMID: 33494265 PMCID: PMC7909810 DOI: 10.3390/pathogens10020096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 11/16/2022] Open
Abstract
The human microbiota is an array of microorganisms known to interact with the host and other microbes. These interactions can be competitive, as microbes must adapt to host- and microorganism-related stressors, thus producing toxic molecules, or cooperative, whereby microbes survive by maintaining homeostasis with the host and host-associated microbial communities. As a result, these microbial interactions shape host health and can potentially result in disease. In this review, we discuss these varying interactions across microbial species, their positive and negative effects, the therapeutic potential of these interactions, and their implications on our knowledge of human well-being.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine A. Wakeman
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (J.B.); (K.B.); (J.N.R.); (K.D.Y.); (S.I.); (H.A.M.); (J.E.W.)
| |
Collapse
|
37
|
Carvalho FM, Teixeira-Santos R, Mergulhão FJM, Gomes LC. The Use of Probiotics to Fight Biofilms in Medical Devices: A Systematic Review and Meta-Analysis. Microorganisms 2020; 9:microorganisms9010027. [PMID: 33374844 PMCID: PMC7824608 DOI: 10.3390/microorganisms9010027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 12/25/2022] Open
Abstract
Medical device-associated infections (MDAI) are a critical problem due to the increasing usage of medical devices in the aging population. The inhibition of biofilm formation through the use of probiotics has received attention from the medical field in the last years. However, this sparse knowledge has not been properly reviewed, so that successful strategies for biofilm management can be developed. This study aims to summarize the relevant literature about the effect of probiotics and their metabolites on biofilm formation in medical devices using a PRISMA-oriented (Preferred Reporting Items for Systematic reviews and Meta-Analyses) systematic search and meta-analysis. This approach revealed that the use of probiotics and their products is a promising strategy to hinder biofilm growth by a broad spectrum of pathogenic microorganisms. The meta-analysis showed a pooled effect estimate for the proportion of biofilm reduction of 70% for biosurfactants, 76% for cell-free supernatants (CFS), 77% for probiotic cells and 88% for exopolysaccharides (EPS). This review also highlights the need to properly analyze and report data, as well as the importance of standardizing the in vitro culture conditions to facilitate the comparison between studies. This is essential to increase the predictive value of the studies and translate their findings into clinical applications.
Collapse
|
38
|
Wu D, Li X, Yu Y, Gong B, Zhou X. Heparin stimulates biofilm formation of Escherichia coli strain Nissle 1917. Biotechnol Lett 2020; 43:235-246. [PMID: 33011901 DOI: 10.1007/s10529-020-03019-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Escherichia coli strain Nissle 1917 (EcN), a gut probiotic competing with pathogenic bacteria, has been used to attenuate various intestinal dysfunctions. Heparin is a sulfated glycosaminoglycan enriched in the human and animal intestinal mucosa, which has a close connection with bacterial biofilm formation. However, the characteristics of heparin affecting bacterial biofilm formation remain obscure. In this study, we investigated the influence of heparin and its derivatives on EcN biofilm formation. RESULTS Here, we found that heparin stimulated EcN biofilm formation in a dose-dependent manner. With the addition of native heparin, the EcN biofilm formation increased 6.9- to 10.8-fold than that without heparin, and was 1.4-, 3.1-, 3.0-, and 3.8-fold higher than that of N-desulfated heparin (N-DS), 2-O-desulfated heparin (2-O-DS), 6-O-desulfated heparin (6-O-DS), and N-/2-O-/6-O-desulfated heparin (N-/2-O-/6-O-DS), respectively. Depolymerization of heparin produced chain-shortened heparin fragments with decreased molecular weight. The depolymerized heparins did not stimulate EcN biofilm formation. The OD570 value of EcN biofilm with the addition of chain-shortened heparin fragments was 8.7-fold lower than that of the native heparin. Furthermore, the biofilm formation of Salmonella enterica serovar Typhimurium was also investigated with the addition of heparin derivatives, and the results were consistent with that of EcN biofilm formation. CONCLUSIONS We conclude that heparin stimulated EcN biofilm formation. Both the sulfation and chain-length of heparin contributed to the enhancement of EcN biofilm formation. This study increases the understanding of how heparin affects biofilm formation, indicating the potential role of heparin in promoting intestinal colonization of probiotics that antagonize pathogen infections.
Collapse
Affiliation(s)
- Dandan Wu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiaomei Li
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yanying Yu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Bingxue Gong
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xianxuan Zhou
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
39
|
Ohno M, Hasegawa M, Hayashi A, Caballero-Flores G, Alteri CJ, Lawley TD, Kamada N, Núñez G, Inohara N. Lipopolysaccharide O structure of adherent and invasive Escherichia coli regulates intestinal inflammation via complement C3. PLoS Pathog 2020; 16:e1008928. [PMID: 33027280 PMCID: PMC7571687 DOI: 10.1371/journal.ppat.1008928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/19/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
Gut dysbiosis associated with intestinal inflammation is characterized by the blooming of particular bacteria such as adherent-invasive E. coli (AIEC). However, the precise mechanisms by which AIEC impact on colitis remain largely unknown. Here we show that antibiotic-induced dysbiosis worsened chemically-induced colitis in IL-22-deficient mice, but not in wild-type mice. The increase in intestinal inflammation was associated with the expansion of E. coli strains with genetic and functional features of AIEC. These E. coli isolates exhibited high ability to out compete related bacteria via colicins and resistance to the host complement system in vitro. Mutation of wzy, the lipopolysaccharide O polymerase gene, rendered AIEC more sensitive to the complement system and more susceptible to engulfment and killing by phagocytes while retaining its ability to outcompete related bacteria in vitro. The wzy AIEC mutant showed impaired fitness to colonize the intestine under colitic conditions, but protected mice from chemically-induced colitis. Importantly, the ability of the wzy mutant to protect from colitis was blocked by depletion of complement C3 which was associated with impaired intestinal eradication of AIEC in colitic mice. These studies link surface lipopolysaccharide O-antigen structure to the regulation of colitic activity in commensal AIEC via interactions with the complement system.
Collapse
Affiliation(s)
- Masashi Ohno
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Mizuho Hasegawa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Atsushi Hayashi
- Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Miyarisan Pharmaceutical Co., Ltd., Central Research Institute, Saitama, Japan
| | - Gustavo Caballero-Flores
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Christopher J. Alteri
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, United States of America
| | - Trevor D. Lawley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Nobuhiko Kamada
- Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Gabriel Núñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Naohiro Inohara
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
40
|
The Cu(II) Reductase RclA Protects Escherichia coli against the Combination of Hypochlorous Acid and Intracellular Copper. mBio 2020; 11:mBio.01905-20. [PMID: 32994322 PMCID: PMC7527725 DOI: 10.1128/mbio.01905-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Enterobacteria, including Escherichia coli, bloom to high levels in the gut during inflammation and strongly contribute to the pathology of inflammatory bowel diseases. To survive in the inflamed gut, E. coli must tolerate high levels of antimicrobial compounds produced by the immune system, including toxic metals like copper and reactive chlorine oxidants such as hypochlorous acid (HOCl). Here, we show that extracellular copper is a potent detoxifier of HOCl and that the widely conserved bacterial HOCl resistance enzyme RclA, which catalyzes the reduction of copper(II) to copper(I), specifically protects E. coli against damage caused by the combination of HOCl and intracellular copper. E. coli lacking RclA was highly sensitive to HOCl when grown in the presence of copper and was defective in colonizing an animal host. Our results indicate that there is unexpected complexity in the interactions between antimicrobial toxins produced by innate immune cells and that bacterial copper status is a key determinant of HOCl resistance and suggest an important and previously unsuspected role for copper redox reactions during inflammation.IMPORTANCE During infection and inflammation, the innate immune system uses antimicrobial compounds to control bacterial populations. These include toxic metals, like copper, and reactive oxidants, including hypochlorous acid (HOCl). We have now found that RclA, a copper(II) reductase strongly induced by HOCl in proinflammatory Escherichia coli and found in many bacteria inhabiting epithelial surfaces, is required for bacteria to resist killing by the combination of intracellular copper and HOCl and plays an important role in colonization of an animal host. This finding indicates that copper redox chemistry plays a critical and previously underappreciated role in bacterial interactions with the innate immune system.
Collapse
|
41
|
Deng Z, Luo XM, Liu J, Wang H. Quorum Sensing, Biofilm, and Intestinal Mucosal Barrier: Involvement the Role of Probiotic. Front Cell Infect Microbiol 2020; 10:538077. [PMID: 33102249 PMCID: PMC7546212 DOI: 10.3389/fcimb.2020.538077] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022] Open
Abstract
The intestine is a particularly dynamic environment in which the host constantly interacts with trillions of symbiotic bacteria called the microbiota. Using quorum sensing (QS) communication, bacteria can coordinate their social behavior and influence host cell activities in a non-invasive manner. Nowadays, a large amount of research has greatly spurred the understanding of how bacterial QS communication regulates bacterial cooperative behaviors due to coexistence and host-microbe interactions. In this review, we discuss bacterial QS in the gut and its role in biofilm formation. As a biological barrier, the mucosal immune system can effectively prevent pathogenic microorganisms and other immunogenic components from entering the internal environment of the host. We focus on the relationship between biofilm and intestinal mucosal immunity, and how probiotic bacteria may regulate them. This review is to provide a theoretical basis for the development of new techniques including probiotics targeting the intestinal barrier function, thereby improving gut health.
Collapse
Affiliation(s)
- Zhaoxi Deng
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jianxin Liu
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Haifeng Wang
- Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Science, Zhejiang University, Hangzhou, China
- *Correspondence: Haifeng Wang
| |
Collapse
|
42
|
Role of Lactobacillus biofilms in Listeria monocytogenes adhesion to glass surfaces. Int J Food Microbiol 2020; 334:108804. [PMID: 32818764 DOI: 10.1016/j.ijfoodmicro.2020.108804] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Listeria monocytogenes can form long-lasting biofilms on food-contact surfaces. Lactic acid bacteria (LAB) have shown promise in antagonizing this microorganism in liquid media. However, the ecological relationships differ when cells are forming biofilms. In this work, we propose the use of Lactobacillus biofilms as surface "conditioners" to modulate the adhesion of L. monocytogenes. For this, the biofilm formation ability of Lactobacillus fermentum MP26 and Lactobacillus salivarius MP14 (human milk origin), fluorescently labeled by transfer of the mCherry-encoding pRCR12 plasmid, was first evaluated. Then, mature biofilms of these strains transformed with pRCR12 for expressing the fluorescent protein mCherry were used as adhesion substrate for GFP-tagged L. monocytogenes Scott A. The resulting biofilms were studied in terms of cellular population and attached biomass (cells plus matrix). Species distribution inside the biofilm structure was revealed by confocal laser scanning microscopy (CLSM). Although none of the Lactobacillus spp. strains reduced the adhesion of L. monocytogenes Scott A, species interactions seem to interfere with the synthesis of extracellular polymeric substances and species distribution inside the biofilms. In dual-species biofilms, CLSM images revealed that Lactobacillus cells were trapping those of L. monocytogenes Scott A. When surfaces were conditioned with Lactobacillus biofilms, the spatial distribution of L. monocytogenes Scott A cells was species-specific, suggesting these interactions are governing the ultimate biofilm structure. The results here obtained open new possibilities for controlling L. monocytogenes dispersal using these Lactobacillus spp. biofilms as a "natural" immobilization way. Whether species interactions could modify the virulence of L. monocytogenes still remains unclear.
Collapse
|
43
|
Xu J, Xia K, Li P, Qian C, Li Y, Liang X. Functional investigation of the chromosomal ccdAB and hipAB operon in Escherichia coli Nissle 1917. Appl Microbiol Biotechnol 2020; 104:6731-6747. [PMID: 32535695 PMCID: PMC7293176 DOI: 10.1007/s00253-020-10733-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/23/2020] [Accepted: 06/07/2020] [Indexed: 12/31/2022]
Abstract
Toxin-antitoxin systems (TASs) have attracted much attention due to their important physiological functions. These small genetic factors have been widely studied mostly in commensal Escherichia coli strains, whereas the role of TASs in the probiotic E. coli Nissle 1917 (EcN) is still elusive. Here, the physiological role of chromosomally encoded type II TASs in EcN was examined. We showed that gene pair ECOLIN_00240-ECOLIN_00245 and ECOLIN_08365-ECOLIN_08370 were two functional TASs encoding CcdAB and HipAB, respectively. The homologs of CcdAB and HipAB were more conserved in E. coli species belonging to pathogenic groups, suggesting their important roles in EcN. CRISPRi-mediated repression of ccdAB and hipAB significantly reduced the biofilm formation of EcN in the stationary phase. Moreover, ccdAB and hipAB were shown to be responsible for the persister formation in EcN. Biofilm and persister formation of EcN controlled by the ccdAB and hipAB were associated with the expression of genes involved in DNA synthesis, SOS response, and stringent response. Besides, CRISPRi was proposed to be an efficient tool in annotating multiple TASs simultaneously. Collectively, our results advance knowledge and understanding of the role of TASs in EcN, which will enhance the utility of EcN in probiotic therapy. Key points • Two TASs in EcN were identified as hipAB and ccdAB. • Knockdown of HipAB and CcdAB resulted in decreased biofilm formation of EcN. • Transcriptional silencing of hipAB and ccdAB affected the persister formation of EcN. • An attractive link between TASs and stress response was unraveled in EcN. • CRISPRi afforded a fast and in situ annotation of multiple TASs simultaneously.
Collapse
Affiliation(s)
- Jun Xu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Kai Xia
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Pinyi Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Chenggong Qian
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yudong Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Xinle Liang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| |
Collapse
|
44
|
Samuels AN, Roggiani M, Smith KA, Zhu J, Goulian M, Kohli RM. Deciphering the Role of Colicins during Colonization of the Mammalian Gut by Commensal E. coli. Microorganisms 2020; 8:microorganisms8050664. [PMID: 32370119 PMCID: PMC7284606 DOI: 10.3390/microorganisms8050664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Colicins are specific and potent toxins produced by Enterobacteriaceae that result in the rapid elimination of sensitive cells. Colicin production is commonly found throughout microbial populations, suggesting its potential importance for bacterial survival in complex microbial environments. Nonetheless, as colicin biology has been predominately studied using synthetic models, it remains unclear how colicin production contributes to survival and fitness of a colicin-producing commensal strain in a natural environment. To address this gap, we took advantage of MP1, an E. coli strain that harbors a colicinogenic plasmid and is a natural colonizer of the murine gut. Using this model, we validated that MP1 is competent for colicin production and then directly interrogated the importance of colicin production and immunity for MP1 survival in the murine gut. We showed that colicin production is dispensable for sustained colonization in the unperturbed gut. A strain lacking colicin production or immunity shows minimal fitness defects and can resist displacement by colicin producers. This report extends our understanding of the role that colicin production may play for E. coli during gut colonization and suggests that colicin production is not essential for a commensal to persist in its physiologic niche in the absence of exogenous challenges.
Collapse
Affiliation(s)
- Amanda N. Samuels
- Department of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Graduate Group on Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Manuela Roggiani
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.R.); (K.A.S.); (M.G.)
| | - Kathryn A. Smith
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.R.); (K.A.S.); (M.G.)
- Department of Biology, Solenis LLC., Wilmington, DE 19803, USA
| | - Jun Zhu
- Graduate Group on Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Mark Goulian
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.R.); (K.A.S.); (M.G.)
| | - Rahul M. Kohli
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: ; Tel.: +1-(215)-573-7523
| |
Collapse
|
45
|
Fang K, Park OJ, Hong SH. Controlling biofilms using synthetic biology approaches. Biotechnol Adv 2020; 40:107518. [PMID: 31953206 PMCID: PMC7125041 DOI: 10.1016/j.biotechadv.2020.107518] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 12/22/2022]
Abstract
Bacterial biofilms are formed by the complex but ordered regulation of intra- or inter-cellular communication, environmentally responsive gene expression, and secretion of extracellular polymeric substances. Given the robust nature of biofilms due to the non-growing nature of biofilm bacteria and the physical barrier provided by the extracellular matrix, eradicating biofilms is a very difficult task to accomplish with conventional antibiotic or disinfectant treatments. Synthetic biology holds substantial promise for controlling biofilms by improving and expanding existing biological tools, introducing novel functions to the system, and re-conceptualizing gene regulation. This review summarizes synthetic biology approaches used to eradicate biofilms via protein engineering of biofilm-related enzymes, utilization of synthetic genetic circuits, and the development of functional living agents. Synthetic biology also enables beneficial applications of biofilms through the production of biomaterials and patterning biofilms with specific temporal and spatial structures. Advances in synthetic biology will add novel biofilm functionalities for future therapeutic, biomanufacturing, and environmental applications.
Collapse
Affiliation(s)
- Kuili Fang
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Oh-Jin Park
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA; Department of Biological and Chemical Engineering, Yanbian University of Science and Technology, Yanji, Jilin, People's Republic of China
| | - Seok Hoon Hong
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA.
| |
Collapse
|
46
|
Leech J, Golub S, Allan W, Simmons MJH, Overton TW. Non-pathogenic Escherichia coli biofilms: effects of growth conditions and surface properties on structure and curli gene expression. Arch Microbiol 2020; 202:1517-1527. [PMID: 32222779 PMCID: PMC7355273 DOI: 10.1007/s00203-020-01864-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/21/2020] [Accepted: 03/12/2020] [Indexed: 11/28/2022]
Abstract
Biofilm formation is a harmful phenomenon in many areas, such as in industry and clinically, but offers advantages in the field of biocatalysis for the generation of robust biocatalytic platforms. In this work, we optimised growth conditions for the production of Escherichia coli biofilms by three strains (PHL644, a K-12 derivative with enhanced expression of the adhesin curli; the commercially-used strain BL21; and the probiotic Nissle 1917) on a variety of surfaces (plastics, stainless steel and PTFE). E. coli PHL644 and PTFE were chosen as optimal strain and substratum, respectively, and conditions (including medium, temperature, and glucose concentration) for biofilm growth were determined. Finally, the impact of these growth conditions on expression of the curli genes was determined using flow cytometry for planktonic and sedimented cells. We reveal new insights into the formation of biofilms and expression of curli in E. coli K-12 in response to environmental conditions.
Collapse
Affiliation(s)
- James Leech
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NH, UK
| | - Stacey Golub
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Wendy Allan
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Mark J H Simmons
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Tim W Overton
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK. .,Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
47
|
Abstract
Mucus in the gastrointestinal (GI) tract is the primary point-of-interaction between humans and their gut microbiota. This intimates that mucus not only ensures protection against endogenous and exogenous opportunists but also provisions for the human microbiota to reside and flourish. With the emergence of living therapeutics, engineered microbes can deliver and produce increasingly complex medicine, and controlling the mucoadhesive properties of different microbial chassis can dictate dose-response in a patient. Here we present a redesigned, in vitro, plate-based assay to measure the mucus adhesion of various probiotics. Cell-mucus interactions were isolated by immobilizing mucus to the plate surface. Binding parameters were derived for each probiotic strain by measuring cell adhesion over a wide range of cell concentrations, providing dose-dependent adhesion metrics. Surface proteins and cell components known to influence mucoadhesion were then heterologously expressed or altered in Lactococcus lactis MG1363 and Escherichia coli Nissle 1917 to control mucus-binding capacity, avidity, and cooperativity.
Collapse
Affiliation(s)
- Zachary J. S. Mays
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Todd C. Chappell
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Nikhil U. Nair
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
48
|
Ma J, Li C, Wang J, Gu J. Genetically Engineered Escherichia coli Nissle 1917 Secreting GLP-1 Analog Exhibits Potential Antiobesity Effect in High-Fat Diet-Induced Obesity Mice. Obesity (Silver Spring) 2020; 28:315-322. [PMID: 31970910 DOI: 10.1002/oby.22700] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study aimed to investigate the potential antiobesity effect of genetically modified Escherichia coli Nissle 1917 (EcN-GM) in mice fed a high-fat diet (HFD). METHODS The mice were randomly divided into six groups: a normal diet group (ND), a HFD group, a HFD + EcN group, and three HFD + EcN-GM groups. The effects of EcN-GM on body weight, food intake, fat pad and organ weight, and an oral glucose tolerance test were measured, in addition to hepatic biochemistry and histological analysis. The mRNA expression of neuropeptides related to food intake regulation in the hypothalamus was also detected. RESULTS The results showed that EcN-GM decreased body weight, body weight gain, food intake, fat pad weight, and hepatic weight of HFD mice. There were beneficial effects of EcN-GM on blood glucose, hepatic biochemistry, and hepatic histological alterations. A dramatic switch of food intake-regulating gene expression in the hypothalamus was also observed in mice. CONCLUSIONS This work has revealed that a modified live bacterial therapeutic, EcN-GM, has potential beneficial effects on obesity. This effect may be related to the regulating of the neuropeptide expression of energy intake and expenditure in the hypothalamus. This study demonstrates a successful example of engineered EcN-GM as a novel approach for weight management.
Collapse
Affiliation(s)
- Jie Ma
- Department of Research and Development, LiTong Bio-Medical Science, Chengdu, Sichuan, P.R. China
| | - Cuiying Li
- Department of Blood Transfusion, Air Force Characteristic Medical Center, Beijing, P.R. China
| | - Junrui Wang
- Department of Orthopaedics, Chengdu Second People's Hospital, Chengdu, Sichuan, P.R. China
| | - Jianwen Gu
- Department of Neurosurgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, P.R. China
| |
Collapse
|
49
|
Soundararajan M, von Bünau R, Oelschlaeger TA. K5 Capsule and Lipopolysaccharide Are Important in Resistance to T4 Phage Attack in Probiotic E. coli Strain Nissle 1917. Front Microbiol 2019; 10:2783. [PMID: 31849915 PMCID: PMC6895014 DOI: 10.3389/fmicb.2019.02783] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
Rapidly growing antibiotic resistance among gastrointestinal pathogens, and the ability of antibiotics to induce the virulence of these pathogens makes it increasingly difficult to rely on antibiotics to treat gastrointestinal infections. The probiotic Escherichia coli strain Nissle 1917 (EcN) is the active component of the pharmaceutical preparation Mutaflor® and has been successfully used in the treatment of gastrointestinal disorders. Gut bacteriophages are dominant players in maintaining the microbial homeostasis in the gut, however, their interaction with incoming probiotic bacteria remains to be at conception. The presence of bacteriophages in the gut makes it inevitable for any probiotic bacteria to be phage resistant, in order to survive and successfully colonize the gut. This study addresses the phage resistance of EcN, specifically against lytic T4 phage infection. From various experiments we could show that (i) EcN is resistant toward T4 phage infection, (ii) EcN's K5 polysaccharide capsule plays a crucial role in T4 phage resistance and (iii) EcN's lipopolysaccharide (LPS) inactivates T4 phages and notably, treatment with the antibiotic polymyxin B which neutralizes the LPS destroyed the phage inactivation ability of isolated LPS from EcN. Combination of these identified properties in EcN was not found in other tested commensal E. coli strains. Our results further indicated that N-acetylglucosamine at the distal end of O6 antigen in EcN's LPS could be the interacting partner with T4 phages. From our findings, we have reported for the first time, the role of EcN's K5 capsule and LPS in its defense against T4 phages. In addition, by inactivating the T4 phages, EcN also protects E. coli K-12 strains from phage infection in tri-culture experiments. Our research highlights phage resistance as an additional safety feature of EcN, a clinically successful probiotic E. coli strain.
Collapse
Affiliation(s)
- Manonmani Soundararajan
- Institute for Molecular Infection Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | | | - Tobias A Oelschlaeger
- Institute for Molecular Infection Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| |
Collapse
|
50
|
Invited review: Tannins as a potential alternative to antibiotics to prevent coliform diarrhea in weaned pigs. Animal 2019; 14:95-107. [PMID: 31571564 DOI: 10.1017/s1751731119002143] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In addition to a multifactorial etiology of nutritional, social and environmental stressors, post-weaning diarrhea (PWD) in pigs is often related to infection with specific pathogens such as enterotoxigenic Escherichia coli (ETEC). In swine farming operations, the incidence of PWD is a global concern and is associated with an unbalanced gut status, resulting in poor performance and high antimicrobial consumption via prophylaxis and metaphylaxis. Increases in antimicrobial resistance are reinforcing an already-urgent need for sustainable, alternative solutions for maintaining optimal gut health in livestock. Tannin-rich plants and extracts contain bioactive compounds that could be of great interest in this respect. This review describes how the use of tannins around weaning could be beneficial for pigs, with special emphasis on the reduction of ETEC-related PWD. An overview of the broad chemical diversity of tannins is presented together with their physicochemical and biological properties, as well as how they may be metabolized in the digestive tract. The pharmacological effects exerted by tannins are summarized; more precisely, the possible mechanisms by which tannins can disrupt the different steps of the pathogenesis of ETEC-related PWD are highlighted. The factors affecting the bioactivity of tannins are also discussed, shedding light on the importance of chemical structure among different tannins.
Collapse
|