1
|
Chen N, Xi J, Du N, Shen R, Zhao R, He W, Peng T, Yang Y, Zhang Y, Yu L, Tan W, Yuan Q. Framework nucleic acid strategy enables closer microbial contact for programming short-range interaction. SCIENCE ADVANCES 2024; 10:eadr4399. [PMID: 39661693 PMCID: PMC11633756 DOI: 10.1126/sciadv.adr4399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Programming precise and specific microbial intraspecies or interspecies interaction would be powerful for microbial metabolic regulation, signal pathway mechanism understanding, and therapeutic application. However, it is still of great challenge to develop a simple and universal method to artificially encode the microbial interactions without interfering with the intrinsic cell metabolism. Here, we proposed an extensible and flexible framework nucleic acid strategy for encoding the specific and precise microbial interactions upon self-assembly. With this spatial manipulation tool, we propose the microbial spatial heterogeneity and short-range interaction mechanism that the microbial assembly facilitates the gene expressions of the surface sensors including flagella and pili in Pseudomonas aeruginosa, leading to a more sensitive response to quorum sensing. The microbial interaction programming strategy proposed in this work is expected to provide a powerful and designable nanoplatform for better understanding of distance-dependent bacterial communication networks.
Collapse
Affiliation(s)
- Na Chen
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Jing Xi
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Na Du
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Ruichen Shen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Rui Zhao
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Wei He
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Tianhuang Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Yanbing Yang
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Lilei Yu
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Quan Yuan
- Renmin Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Institute of Molecular Medicine, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
2
|
Zhao Y, Li S, Zhang R, Zhang X, Shen Q, Zhang X, Tian T, Hou X. Characteristics of gut microbiota of premature infants in the early postnatal period and their relationship with intraventricular hemorrhage. BMC Microbiol 2024; 24:513. [PMID: 39623318 PMCID: PMC11610090 DOI: 10.1186/s12866-024-03675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Studies have shown correlations between gut microbiota and neurocognitive function, but little was known about the early postnatal gut microbiota and intraventricular hemorrhage (IVH). We aimed to explore the characteristics of gut microbiota in premature infants and their relationship with IVH, further exploring potential therapeutic targets. METHODS Premature infants delivered at Peking University First Hospital from February 2023 to August 2023 were recruited as a cohort. Feces samples were collected on postnatal days 1, 3, and 5. Premature infants were divided into normal, mild IVH, and severe IVH groups based on cranial ultrasound. 16S rRNA amplicon sequencing technology was used to determine the fecal microbiota, and the results were analyzed. RESULTS Thirty-eight premature infants were enrolled. There was a significant difference in alpha and beta diversity among the three groups. The relative abundance of E. coli and A. muciniphila was different among the three groups. Further random forest analysis indicated that S. lutetiensis, L. mirabilis, and N. macacae can effectively distinguish premature infants with IVH. Finally, the phylogenetic investigation of communities by reconstruction of unobserved states2 (PICRUSt2) functional gene analysis predicted significant differences in energy metabolism, carbohydrate metabolism, metabolism of cofactors and vitamins, and membrane transport between normal and severe IVH groups. CONCLUSIONS The gut microbiota in the early postnatal period of premature infants is closely associated with the IVH status. As age increases, the differences in gut microbiota of premature infants with different degrees of IVH continue to increase, and the trend of changes with severity of IVH becomes more and more obvious. E. coli, A. muciniphila, S. lutetiensis, L. mirabilis, N. macacae, G. haemolysans, and S. oralis can effectively distinguish between IVH infants and normal premature infants. The results indicate that gut microbiota is expected to provide effective therapeutic targets for the diagnosis and treatment of IVH.
Collapse
Affiliation(s)
- Yunlong Zhao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Shan Li
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Rui Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xin Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Qiuyue Shen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xingyun Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Tian Tian
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xinlin Hou
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
| |
Collapse
|
3
|
Coyago-Cruz E, Alarcón A, Guachamin A, Méndez G, Osorio E, Heredia-Moya J, Zuñiga-Miranda J, Beltrán-Sinchiguano E, Vera E. Functional, Antioxidant, Antibacterial, and Antifungal Activity of Edible Flowers. Antioxidants (Basel) 2024; 13:1297. [PMID: 39594439 PMCID: PMC11590945 DOI: 10.3390/antiox13111297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Edible flowers have been used since ancient times, but their potential for improving human health has not been explored. This study aimed to evaluate the profile of bioactive compounds (organic acids, phenolics, and carotenoids) and the antioxidant and antimicrobial activity of nine flower varieties with high concentrations of carotenoids or total phenolic compounds. Ninety-three edible flowers were analysed for physicochemical characteristics, total phenolic and carotenoid concentrations, and antioxidant activity (ABTS). Bioactive profiles were determined by rapid resolution liquid chromatography (RRLC), and antimicrobial activity was determined against Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa and Streptococcus mutans, and Candida albicans and Candida tropicalis. Chrysanthemum x hybrid orange, Helianthus annuus yellow, Tagetes patula orange, Canna indica red, and Hibiscus rosa-sinensis (orange1 and yellow) showed significant concentrations of total carotenoids. In contrast, Pelargonium hortorum orange2, Hibiscus rosa-sinensis red1, and Rosa x hybrid variety medium yellow showed high levels of total phenolics. The predominant compounds in these species were citric acid (991.4 mg/g DW in Hibiscus rosa-sinensis red1), 4-hydroxybenzoic acid (936.2 mg/100 g DW in P. hortorum orange2), kaempferol (971. 9 mg/100 g DW in T. patula orange), quercetin glucoside (958.8 in C. x hybrid), quercetin (919.3 mg/100 g DW in T. patula), α-carotene, and β-carotene in T. patula orange (989.5 and 601.2 mg/100 g DW, respectively). Regarding antimicrobial activity, T. patula orange and P. hortorum orange2 inhibited bacterial growth, while C. x hybrid orange and P. hortorum orange2 inhibited Candida albicans, and the latter inhibited Candida tropicalis. These results indicate the potential of edible flowers as a natural source of bioactive compounds and as a tool in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Elena Coyago-Cruz
- Carrera de Ingeniería en Biotecnología de los Recursos Naturales, Universidad Politécnica Salesiana, Sede Quito, Campus El Girón, Av. 12 de octubre N2422 y Wilson, Quito 170109, Ecuador
| | - Alejandro Alarcón
- Maestría en Productos Farmacéuticos Naturales, Universidad Politécnica Salesiana, Sede Quito, Campus El Girón, Av. 12 de octubre N2422 y Wilson, Quito 170109, Ecuador
| | - Aida Guachamin
- Carrera de Ingeniería en Biotecnología de los Recursos Naturales, Universidad Politécnica Salesiana, Sede Quito, Campus El Girón, Av. 12 de octubre N2422 y Wilson, Quito 170109, Ecuador
| | - Gabriela Méndez
- Carrera de Ingeniería en Biotecnología de los Recursos Naturales, Universidad Politécnica Salesiana, Sede Quito, Campus El Girón, Av. 12 de octubre N2422 y Wilson, Quito 170109, Ecuador
| | - Edison Osorio
- Carrera de Ingeniería en Biotecnología de los Recursos Naturales, Universidad Politécnica Salesiana, Sede Quito, Campus El Girón, Av. 12 de octubre N2422 y Wilson, Quito 170109, Ecuador
| | - Jorge Heredia-Moya
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Johana Zuñiga-Miranda
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | | | - Edwin Vera
- Departamento de Ciencia de los Alimentos y Biotecnología, Facultad de Ingeniería Química, Escuela Politécnica Nacional, Quito 170524, Ecuador
| |
Collapse
|
4
|
Pepoyan A. Gut Akkermansia muciniphila, Prevotellaceae, and Enterobacteriaceae spp. as Possible Markers in Women-Related Nutritional and Clinical Trials: Familial Mediterranean Fever Disease. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2024; 5:785-793. [PMID: 39439767 PMCID: PMC11491588 DOI: 10.1089/whr.2024.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
Background Studies have shown that the gut microbiota of healthy men and men with familial Mediterranean fever (FMF) disease respond differently to placebo. Given the fact that the composition of the gut microbiota is different in men and women, this study aimed to describe in detail the placebo response of the gut microbiota in healthy and FMF women. Materials and Methods The bacterial response to placebo was fully evaluated on a previous PhyloChip™ DNA microarray-based assay (GEO Series; accession number GSE111835). Results The change in the total number of operational taxonomic units in healthy women exposed to placebo is more than that of healthy men, in contrast to FMF people (704 vs. 140 and 409 vs. 7560, respectively [p < 0.05]). Gut Firmicutes diversities are more sensitive to placebo, whereas Akkermansia muciniphila remained unchanged after the placebo administration for both healthy and FMF people. Gut Prevotellaceae and Enterobacteriaceae diversities of healthy subjects and FMF women are also almost unchanged from placebo. Meanwhile, only 56.35% of gut Enterobacteriaceae diversities in FMF men were placebo resistant. Conclusion The response to a placebo varies depending on a person's gender and health status. Healthy and FMF women's placebo study groups could be avoided by excluding placebo-sensitive 704 of 18,725 and 409 of 18,725 bacterial diversities, respectively. Because the placebo causes changes in all gut bacterial phyla in healthy and FMF women, and only the representatives of Enterobacteriaceae and Prevotellaceae families and A. muciniphila spp. are not affected by placebo, these bacteria can be considered as possible markers in women-related nutritional/clinical trials. Data on the response of the gut microbiota in healthy women to placebo might be used in studies of diseases other than FMF. The response of gut bacteria from different taxonomic affiliations to placebo may provide a basis for uncovering the role of these bacteria in the gut-brain axis.
Collapse
Affiliation(s)
- Astghik Pepoyan
- Food Safety and Biotechnology Department, Scientific Research Institute of Food Science and Biotechnology, Armenian National Agrarian University, Yerevan, Armenia
- International Association for Human and Animals Health Improvement, Yerevan, Armenia
| |
Collapse
|
5
|
Nguyen BD, Sintsova A, Schubert C, Sichert A, Scheidegger C, Näf J, Huttman J, Lentsch V, Keys T, Rutschmann C, Christen P, Kiefer P, Keller P, Barthel M, Cuenca M, Christen B, Sauer U, Slack E, Vorholt JA, Sunagawa S, Hardt WD. Salmonella Typhimurium screen identifies shifts in mixed-acid fermentation during gut colonization. Cell Host Microbe 2024; 32:1758-1773.e4. [PMID: 39293436 DOI: 10.1016/j.chom.2024.08.015] [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: 11/23/2023] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
How enteric pathogens adapt their metabolism to a dynamic gut environment is not yet fully understood. To investigate how Salmonella enterica Typhimurium (S.Tm) colonizes the gut, we conducted an in vivo transposon mutagenesis screen in a gnotobiotic mouse model. Our data implicate mixed-acid fermentation in efficient gut-luminal growth and energy conservation throughout infection. During initial growth, the pathogen utilizes acetate fermentation and fumarate respiration. After the onset of gut inflammation, hexoses appear to become limiting, as indicated by carbohydrate analytics and the increased need for gluconeogenesis. In response, S.Tm adapts by ramping up ethanol fermentation for redox balancing and supplying the TCA cycle with α-ketoglutarate for additional energy. Our findings illustrate how S.Tm flexibly adapts mixed fermentation and its use of the TCA cycle to thrive in the changing gut environment. Similar metabolic wiring in other pathogenic Enterobacteriaceae may suggest a broadly conserved mechanism for gut colonization.
Collapse
Affiliation(s)
- Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Clio Scheidegger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julien Huttman
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Tim Keys
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | | | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Philipp Keller
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Miguelangel Cuenca
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Beat Christen
- Institute of Microbiology, University of Stuttgart, Stuttgart, Germany
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
6
|
Hu H, Wang Y, Sun J, Wang Y, Zhou J, Shi Q, Han X, Jiang Y, Wu D, Huang X, Yu Y. Risk factors and molecular epidemiology of intestinal colonization by carbapenem-resistant Gram-negative bacteria in patients with hematological diseases: a multicenter case‒control study. Microbiol Spectr 2024; 12:e0429923. [PMID: 38847538 PMCID: PMC11218473 DOI: 10.1128/spectrum.04299-23] [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] [Received: 01/03/2024] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
Patients with hematological diseases are considered to be at high risk for intestinal colonization by carbapenem-resistant Gram-negative bacteria (CR-GNB). However, the epidemiological data regarding risk factors and molecular characteristics of intestinal colonized CR-GNB isolates in this population are insufficient in China. A multicenter case‒control study involving 4,641 adult patients with hematological diseases from 92 hospitals across China was conducted. Following culture of collected rectal swabs, mass spectrometry and antimicrobial susceptibility tests were performed to identify GNB species and CR phenotype. Risk factors were assessed through retrospective clinical information. Whole-genome sequencing was used to analyze the molecular characteristics of CR-GNB isolates. This trial is registered with ClinicalTrials.gov as NCT05002582. Our results demonstrated that among 4,641 adult patients, 10.8% had intestinal colonization by CR-GNB. Of these, 8.1% were colonized by carbapenem-resistant Enterobacterales (CRE), 2.6% were colonized by carbapenem-resistant Pseudomonas aeruginosa (CRPA), and 0.3% were colonized by carbapenem-resistant Acinetobacter baumannii (CRAB). The risk factors for CR-GNB colonization include male gender, acute leukemia, hematopoietic stem cell transplantation, β-lactam antibiotic usage, and the presence of non-perianal infections within 1 week. Compared with CRPA-colonized patients, patients using carbapenems were more likely to be colonized with CRE. NDM was the predominant carbapenemase in colonized CRE. This study revealed a high CR-GNB intestinal colonization rate among adult patients with hematological diseases in China, with CRE being the predominant one. Notably, a significant proportion of CRE exhibited metallo-β-lactamase production, indicating a concerning trend. These findings emphasize the importance of active screening for CR-GNB colonization in patients with hematological diseases.IMPORTANCECarbapenem-resistant Gram-negative bacteria (CR-GNB) has emerged as a significant threat to public health. Patients with hematological diseases are at high risk of CR-GNB infections due to their immunosuppressed state. CR-GNB colonization is an independent risk factor for subsequent infection. Understanding the risk factors and molecular characteristics of CR-GNB associated with intestinal colonization in patients with hematological diseases is crucial for empirical treatment, particularly in patients with febrile neutropenia. However, the epidemiology data are still insufficient, and our study aims to determine the intestinal colonization rate of CR-GNB, identify colonization risk factors, and analyze the molecular characteristics of colonized CR-GNB isolates.
Collapse
Affiliation(s)
- Huangdu Hu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinping Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Sun
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Critical Care Medicine, Lishui Central Hospital, Lishui, China
| | - Yuting Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junxin Zhou
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiucheng Shi
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhong Han
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yan Jiang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojun Huang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Muñoz-Cazalla A, de Quinto I, Álvaro-Llorente L, Rodríguez-Beltrán J, Herencias C. The role of bacterial metabolism in human gut colonization. Int Microbiol 2024:10.1007/s10123-024-00550-6. [PMID: 38937311 DOI: 10.1007/s10123-024-00550-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
Can we anticipate the emergence of the next pandemic antibiotic-resistant bacterial clone? Addressing such an ambitious question relies on our ability to comprehensively understand the ecological and epidemiological factors fostering the evolution of high-risk clones. Among these factors, the ability to persistently colonize and thrive in the human gut is crucial for most high-risk clones. Nonetheless, the causes and mechanisms facilitating successful gut colonization remain obscure. Here, we review recent evidence that suggests that bacterial metabolism plays a pivotal role in determining the ability of high-risk clones to colonize the human gut. Subsequently, we outline novel approaches that enable the exploration of microbial metabolism at an unprecedented scale and level of detail. A thorough understanding of the constraints and opportunities of bacterial metabolism in gut colonization will foster our ability to predict the emergence of high-risk clones and take appropriate containment strategies.
Collapse
Affiliation(s)
- Ada Muñoz-Cazalla
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Ignacio de Quinto
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Laura Álvaro-Llorente
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Jerónimo Rodríguez-Beltrán
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas-CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Cristina Herencias
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas-CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Ungor I, Apidianakis Y. Bacterial synergies and antagonisms affecting Pseudomonas aeruginosa virulence in the human lung, skin and intestine. Future Microbiol 2024; 19:141-155. [PMID: 37843410 DOI: 10.2217/fmb-2022-0155] [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: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Pseudomonas aeruginosa requires a significant breach in the host defense to cause an infection. While its virulence factors are well studied, its tropism cannot be explained only by studying its interaction with the host. Why are P. aeruginosa infections so rare in the intestine compared with the lung and skin? There is not enough evidence to claim specificity in virulence factors deployed by P. aeruginosa in each anatomical site, and host physiology differences between the lung and the intestine cannot easily explain the observed differences in virulence. This perspective highlights a relatively overlooked parameter in P. aeruginosa virulence, namely, potential synergies with bacteria found in the human skin and lung, as well as antagonisms with bacteria of the human intestine.
Collapse
Affiliation(s)
- Izel Ungor
- Department of Biological Sciences, University of Cyprus, Nicosia, 2109, Cyprus
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia, 2109, Cyprus
| |
Collapse
|
9
|
Fooladi S, Rabiee N, Iravani S. Genetically engineered bacteria: a new frontier in targeted drug delivery. J Mater Chem B 2023; 11:10072-10087. [PMID: 37873584 DOI: 10.1039/d3tb01805a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Genetically engineered bacteria (GEB) have shown significant promise to revolutionize modern medicine. These engineered bacteria with unique properties such as enhanced targeting, versatility, biofilm disruption, reduced drug resistance, self-amplification capabilities, and biodegradability represent a highly promising approach for targeted drug delivery and cancer theranostics. This innovative approach involves modifying bacterial strains to function as drug carriers, capable of delivering therapeutic agents directly to specific cells or tissues. Unlike synthetic drug delivery systems, GEB are inherently biodegradable and can be naturally eliminated from the body, reducing potential long-term side effects or complications associated with residual foreign constituents. However, several pivotal challenges such as safety and controllability need to be addressed. Researchers have explored novel tactics to improve their capabilities and overcome existing challenges, including synthetic biology tools (e.g., clustered regularly interspaced short palindromic repeats (CRISPR) and bioinformatics-driven design), microbiome engineering, combination therapies, immune system interaction, and biocontainment strategies. Because of the remarkable advantages and tangible progress in this field, GEB may emerge as vital tools in personalized medicine, providing precise and controlled drug delivery for various diseases (especially cancer). In this context, future directions include the integration of nanotechnology with GEB, the focus on microbiota-targeted therapies, the incorporation of programmable behaviors, the enhancement in immunotherapy treatments, and the discovery of non-medical applications. In this way, careful ethical considerations and regulatory frameworks are necessary for developing GEB-based systems for targeted drug delivery. By addressing safety concerns, ensuring informed consent, promoting equitable access, understanding long-term effects, mitigating dual-use risks, and fostering public engagement, these engineered bacteria can be employed as promising delivery vehicles in bio- and nanomedicine. In this review, recent advances related to the application of GEB in targeted drug delivery and cancer therapy are discussed, covering crucial challenging issues and future perspectives.
Collapse
Affiliation(s)
- Saba Fooladi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia.
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| |
Collapse
|
10
|
Wang KC, Lerche MH, Ardenkjær-Larsen JH, Jensen PR. Formate Metabolism in Shigella flexneri and Its Effect on HeLa Cells at Different Stages during the Infectious Process. Microbiol Spectr 2023; 11:e0063122. [PMID: 37042762 PMCID: PMC10269805 DOI: 10.1128/spectrum.00631-22] [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: 03/25/2022] [Accepted: 03/16/2023] [Indexed: 04/13/2023] Open
Abstract
Shigellosis caused by Shigella is one of the most important foodborne illnesses in global health, but little is known about the metabolic cross talk between this bacterial pathogen and its host cells during the different stages of the infection process. A detailed understanding of the metabolism can potentially lead to new drug targets remedying the pressing problem of antibiotic resistance. Here, we use stable isotope-resolved metabolomics as an unbiased and fast method to investigate how Shigella metabolizes 13C-glucose in three different environments: inside the host cells, adhering to the host cells, and alone in suspension. We find that especially formate metabolism by bacteria is sensitive to these different environments. The role of formate in pathogen metabolism is sparsely described in the literature compared to the roles of acetate and butyrate. However, its metabolic pathway is regarded as a potential drug target due to its production in microorganisms and its absence in humans. Our study provides new knowledge about the regulatory effect of formate. Bacterial metabolism of formate is pH dependent when studied alone in culture medium, whereas this effect is less pronounced when the bacteria adhere to the host cells. Once the bacteria are inside the host cells, we find that formate accumulation is reduced. Formate also affects the host cells resulting in a reduced infection rate. This was correlated to an increased immune response. Thus, intriguingly formate plays a double role in pathogenesis by increasing the virulence of Shigella and at the same time stimulating the immune response of the host. IMPORTANCE Bacterial infection is a pressing societal concern due to development of resistance toward known antibiotics. Central carbon metabolism has been suggested as a potential new target for drug development, but metabolic changes upon infection remain incompletely understood. Here, we used a cellular infection model to study how the bacterial pathogen Shigella adapts its metabolism depending on the environment starting from the extracellular medium until Shigella successfully invaded and proliferated inside host cells. The mixed-acid fermentation of Shigella was the major metabolic pathway during the infectious process, and the glucose-derived metabolite formate surprisingly played a divergent role in the pathogen and in the host cell. Our data show reduced infection rate when both host cells and bacteria were treated with formate, which correlated with an upregulated immune response in the host cells. The formate metabolism in Shigella thus potentially provides a route toward alternative treatment strategies for Shigella prevention.
Collapse
Affiliation(s)
- Ke-Chuan Wang
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mathilde Hauge Lerche
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jan Henrik Ardenkjær-Larsen
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Pernille Rose Jensen
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
11
|
Pal R, Athamneh AI, Deshpande R, Ramirez JAR, Adu KT, Muthuirulan P, Pawar S, Biazzo M, Apidianakis Y, Sundekilde UK, de la Fuente-Nunez C, Martens MG, Tegos GP, Seleem MN. Probiotics: insights and new opportunities for Clostridioides difficile intervention. Crit Rev Microbiol 2023; 49:414-434. [PMID: 35574602 PMCID: PMC9743071 DOI: 10.1080/1040841x.2022.2072705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/17/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023]
Abstract
Clostridioides difficile infection (CDI) is a life-threatening disease caused by the Gram-positive, opportunistic intestinal pathogen C. difficile. Despite the availability of antimicrobial drugs to treat CDI, such as vancomycin, metronidazole, and fidaxomicin, recurrence of infection remains a significant clinical challenge. The use of live commensal microorganisms, or probiotics, is one of the most investigated non-antibiotic therapeutic options to balance gastrointestinal (GI) microbiota and subsequently tackle dysbiosis. In this review, we will discuss major commensal probiotic strains that have the potential to prevent and/or treat CDI and its recurrence, reassess the efficacy of probiotics supplementation as a CDI intervention, delve into lessons learned from probiotic modulation of the immune system, explore avenues like genome-scale metabolic network reconstructions, genome sequencing, and multi-omics to identify novel strains and understand their functionality, and discuss the current regulatory framework, challenges, and future directions.
Collapse
Affiliation(s)
- Rusha Pal
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Ahmad I.M. Athamneh
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | | | - Jose A. R Ramirez
- ProbioWorld Consulting Group, James Cook University, 4811, Queensland, Australia
| | - Kayode T. Adu
- ProbioWorld Consulting Group, James Cook University, 4811, Queensland, Australia
- Cann Group, Walter and Eliza Hall Institute, La Trobe University, Victoria 3083, Australia
| | | | - Shrikant Pawar
- The Anlyan Center Yale Center for Genomic Analysis, Yale School of Medicine, New Haven CT USA
| | - Manuele Biazzo
- The Bioarte Ltd Laboratories at Life Science Park, San Gwann, Malta
| | | | | | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark G. Martens
- Reading Hospital, Tower Health, West Reading, PA 19611, USA
- Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - George P. Tegos
- Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Mohamed N. Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
- Center for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
12
|
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
|
13
|
Colicins of Escherichia coli Lead to Resistance against the Diarrhea-Causing Pathogen Enterotoxigenic E. coli in Pigs. Microbiol Spectr 2022; 10:e0139622. [PMID: 36190425 PMCID: PMC9603048 DOI: 10.1128/spectrum.01396-22] [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] [Indexed: 12/30/2022] Open
Abstract
Gut microbes can affect host adaptation to various environment conditions. Escherichia coli is a common gut species, including pathogenic strains and nonpathogenic strains. This study was conducted to investigate the effects of different E. coli strains in the gut on the health of pigs. In this study, the complete genomes of two E. coli strains isolated from pigs were sequenced. The whole genomes of Y18J and the enterotoxigenic E. coli strain W25K were compared to determine their roles in pig adaptation to disease. Y18J was isolated from feces of healthy piglets and showed strong antimicrobial activity against W25K in vitro. Gene knockout experiments and complementation analysis followed by modeling the microbe-microbe interactions demonstrated that the antagonistic mechanism of Y18J against W25K relied on the bacteriocins colicin B and colicin M. Compared to W25K, Y18J is devoid of exotoxin-coding genes and has more secondary-metabolite-biosynthetic gene clusters. W25K carries more genes involved in genome replication, in accordance with a shorter cell cycle observed during a growth experiment. The analysis of gut metagenomes in different pig breeds showed that colicins B and M were enriched in Laiwu pigs, a Chinese local breed, but were scarce in boars and Duroc pigs. IMPORTANCE This study revealed the heterogeneity of E. coli strains from pigs, including two strains studied by both in silico and wet experiments in detail and 14 strains studied by bioinformatics analysis. E. coli Y18J may improve the adaptability of pigs toward disease resistance through the production of colicins B and M. Our findings could shed light on the pathogenic and harmless roles of E. coli in modern animal husbandry, leading to a better understanding of intestinal-microbe-pathogen interactions in the course of evolution.
Collapse
|
14
|
Liu Y, Feng J, Pan H, Zhang X, Zhang Y. Genetically engineered bacterium: Principles, practices, and prospects. Front Microbiol 2022; 13:997587. [PMID: 36312915 PMCID: PMC9606703 DOI: 10.3389/fmicb.2022.997587] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022] Open
Abstract
Advances in synthetic biology and the clinical application of bacteriotherapy enable the use of genetically engineered bacteria (GEB) to combat various diseases. GEB act as a small 'machine factory' in the intestine or other tissues to continuously produce heterologous proteins or molecular compounds and, thus, diagnose or cure disease or work as an adjuvant reagent for disease treatment by regulating the immune system. Although the achievements of GEBs in the treatment or adjuvant therapy of diseases are promising, the practical implementation of this new therapeutic modality remains a grand challenge, especially at the initial stage. In this review, we introduce the development of GEBs and their advantages in disease management, summarize the latest research advances in microbial genetic techniques, and discuss their administration routes, performance indicators and the limitations of GEBs used as platforms for disease management. We also present several examples of GEB applications in the treatment of cancers and metabolic diseases and further highlight their great potential for clinical application in the near future.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Hangcheng Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Functional amyloid in a lipid-like environment: a merry dance of many steps. Essays Biochem 2022; 66:1035-1046. [PMID: 36205438 DOI: 10.1042/ebc20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Abstract
Functional amyloid, which unlike its pathological counterpart serves a biological purpose, is produced in a carefully orchestrated sequence of events. In bacteria, the major amyloid component is transported over the periplasm and through the outer membrane to assemble on the bacterial cell surface. During its life time, the amyloid protein may be exposed to both membrane lipids and extracellular surfactant, making it relevant to study its interactions with these components in vitro. Particularly for charged surfactants, the interaction is quite complex and highly dependent on the surfactant:protein molar ratio. Low ratios typically promote aggregation, likely by binding the proteins to micelles and thus increasing the local concentration of proteins, while higher concentrations see an inhibition of the same process as the protein is diluted out and immobilized on individual micelles. This is particularly pronounced for strongly anionic surfactants like SDS; the naturally occurring biosurfactant rhamnolipid interacts more weakly with the protein, which still not only allows aggregation but also leads to less detrimental effects at higher ratios. Similarly, anionic vesicle-forming lipids largely stimulate aggregation likely because of weaker interactions. Anionic lysolipids, thanks to their micelle-forming properties, resemble SDS in their impact on fibrillation. There are also examples of systems where membrane binding sequesters an otherwise amyloidogenic sequence and prevents fibrillation or-quite the opposite- liberates another part of the protein to engage in self-assembly. Thus, membranes and surfactants have very varied roles to play in the biogenesis and function of bacterial amyloid.
Collapse
|
16
|
Xu Y, Kong X, Zhu Y, Xu J, Mao H, Li J, Zhang J, Zhu X. Contribution of gut microbiota toward renal function in sepsis. Front Microbiol 2022; 13:985283. [PMID: 36147845 PMCID: PMC9486003 DOI: 10.3389/fmicb.2022.985283] [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: 07/03/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis most often involves the kidney and is one of the most common causes of acute kidney injury. The prevalence of septic acute kidney injury has increased significantly in recent years. The gut microbiota plays an important role in sepsis. It interacts with the kidney in a complex and multifactorial process, which is not fully understood. Sepsis may lead to gut microbiota alteration, orchestrate gut mucosal injury, and cause gut barrier failure, which further alters the host immunological and metabolic homeostasis. The pattern of gut microbiota alteration also varies with sepsis progression. Changes in intestinal microecology have double-edged effects on renal function, which also affects intestinal homeostasis. This review aimed to clarify the interaction between gut microbiota and renal function during the onset and progression of sepsis. The mechanism of gut–kidney crosstalk may provide potential insights for the development of novel therapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Yaya Xu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Xiangmei Kong
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Yueniu Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Jiayue Xu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Haoyun Mao
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Jiru Li
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Zhang
- Department of Pediatric Respiratory, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jianhua Zhang,
| | - Xiaodong Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
- Xiaodong Zhu,
| |
Collapse
|
17
|
Rubini E, Schenkelaars N, Rousian M, Sinclair KD, Wekema L, Faas MM, Steegers-Theunissen RPM, Schoenmakers S. Maternal obesity during pregnancy leads to derangements in one-carbon metabolism and the gut microbiota: implications for fetal development and offspring wellbeing. Am J Obstet Gynecol 2022; 227:392-400. [PMID: 35452650 DOI: 10.1016/j.ajog.2022.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 11/01/2022]
Abstract
A healthy diet before and during pregnancy is beneficial in acquiring essential B vitamins involved in 1-carbon metabolism, and in maintaining a healthy gut microbiota. Each play important roles in fetal development, immune-system remodeling, and pregnancy-nutrient acquisition. Evidence shows that there is a reciprocal interaction between the one-carbon metabolism and the gut microbiota given that dietary intake of B vitamins has been shown to influence the composition of the gut microbiota, and certain gut bacteria also synthesize B vitamins. This reciprocal interaction contributes to the individual's overall availability of B vitamins and, therefore, should be maintained in a healthy state during pregnancy. There is an emerging consensus that obese pregnant women often have derangements in 1-carbon metabolism and gut dysbiosis owing to high intake of nutritiously poor foods and a chronic systemic inflammatory state. For example, low folate and vitamin B12 in obese women coincide with the decreased presence of B vitamin-producing bacteria and increased presence of inflammatory-associated bacteria from approximately mid-pregnancy. These alterations are risk factors for adverse pregnancy outcomes, impaired fetal development, and disruption of fetal growth and microbiota formation, which may lead to potential long-term offspring metabolic and neurologic disorders. Therefore, preconceptional and pregnant obese women may benefit from dietary and lifestyle counseling to improve their dietary nutrient intake, and from monitoring their B vitamin levels and gut microbiome by blood tests and microbiota stool samples. In addition, there is evidence that some probiotic bacteria have folate biosynthetic capacity and could be used to treat gut dysbiosis. Thus, their use as an intervention strategy for obese women holds potential and should be further investigated. Currently, there are many knowledge gaps concerning the relationship between one-carbon metabolism and the gut microbiota, and future research should focus on intervention strategies to counteract B vitamin deficiencies and gut dysbiosis in obese pregnant women, commencing with the use of probiotic and prebiotic supplements.
Collapse
Affiliation(s)
- Eleonora Rubini
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Melek Rousian
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kevin D Sinclair
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Lieske Wekema
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Characterization of Antimicrobial Composite Edible Film Formulated from Fermented Cheese Whey and Cassava Peel Starch. MEMBRANES 2022; 12:membranes12060636. [PMID: 35736343 PMCID: PMC9230007 DOI: 10.3390/membranes12060636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023]
Abstract
Antimicrobial composite edible film can be a solution for environmentally friendly food packaging, which can be made from fermented cheese whey containing an antimicrobial agent and cassava peel waste that contains starch. The research aims to determine the formulation of fermented cheese whey and cassava peel waste starch, resulting in an antimicrobial composite edible film with the best physical, mechanical, and water vapour permeability (WVP) properties, as well as with high antimicrobial activity. This research was conducted using experimental methods with nine composite edible film formulation treatments with three replications. Three variations in the fermented cheese whey and cassava peel starch ratio (v/v) (1:3, 1:1, 3:1) were combined with variations in the addition of glycerol (20%, 33%, 45%) (w/w) in the production of the composite edible film. Then, the physical characteristics such as elongation at break, tensile strength, WVP, colour, and antimicrobial effect of its film-forming solution were observed. The results showed that 24 h of whey fermentation with Candida tropicalis resulted in an 18.50 mm inhibition zone towards Pseudomonas aeruginosa. The best characteristic of the film was obtained from the formulation of a whey:starch ratio of 1:3 and 33% glycerol, which resulted in a thickness value of 0.21 mm, elongation at break of 19.62%, tensile strength of 0.81 N/mm2, WVP of 3.41 × 10−10·g/m·s·Pa at a relative humidity (RH) of 100%–35%, and WVP of 9.84 × 10−10·g/m·s·Pa at a RH of 75%–35%, with an antimicrobial activity towards P. aeruginosa of 5.11 mm.
Collapse
|
19
|
Charalambous A, Grivogiannis E, Dieronitou I, Michael C, Rahme L, Apidianakis Y. Proteobacteria and Firmicutes Secreted Factors Exert Distinct Effects on Pseudomonas aeruginosa Infection under Normoxia or Mild Hypoxia. Metabolites 2022; 12:449. [PMID: 35629953 PMCID: PMC9146490 DOI: 10.3390/metabo12050449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Microbiota may alter a pathogen's virulence potential at polymicrobial infection sites. Here, we developed a multi-modal Drosophila assay, amenable to the assessment of human bacterial interactions using fly survival or midgut regeneration as a readout, under normoxia or mild hypoxia. Deploying a matrix of 12 by 33 one-to-one Drosophila co-infections via feeding, we classified bacterial interactions as neutral, synergistic, or antagonistic, based on fly survival. Twenty six percent of these interactions were antagonistic, mainly occurring between Proteobacteria. Specifically, Pseudomonas aeruginosa infection was antagonized by various Klebsiella strains, Acinetobacter baumannii, and Escherichia coli. We validated these interactions in a second screen of 7 by 34 one-to-one Drosophila co-infections based on assessments of midgut regeneration, and in bacterial co-culture test tube assays, where antagonistic interactions depended on secreted factors produced upon high sugar availability. Moreover, Enterococci interacted synergistically with P. aeruginosa in flies and in test tubes, enhancing the virulence and pyocyanin production by P. aeruginosa. However, neither lactic acid bacteria nor their severely hypoxic culture supernatants provided a survival benefit upon P. aeruginosa infection of flies or mice, respectively. We propose that at normoxic or mildly hypoxic sites, Firmicutes may exacerbate, whereas Proteobacteria secreted factors may ameliorate, P. aeruginosa infections.
Collapse
Affiliation(s)
- Anna Charalambous
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Evangelos Grivogiannis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Irene Dieronitou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Christina Michael
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Laurence Rahme
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| |
Collapse
|
20
|
Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 2022; 38:110505. [PMID: 35263602 DOI: 10.1016/j.celrep.2022.110505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key regulator of metabolism and interacts with the intestinal microbiome. Here, we study the role of the Drosophila intestinal stem cell (ISC)-specific biotin transporter Smvt in midgut homeostasis, infection-induced regeneration, and tumorigenesis. We show that Smvt-transported biotin in ISCs is necessary for ISC mitosis. Smvt deficiency impairs intestinal maintenance, which can be rescued by the human Smvt, encoded by SLC5A6. ISC-specific, Smvt-silenced flies exhibit microbial dysbiosis, whereby the growth of Providencia sneebia, an opportunistic pathogen, is favored. Dysbiosis correlates with increased Nox expression, reactive oxygen species (ROS), and enterocyte apoptosis. Flies acquire biotin from their diet and microbiota. We show that, when dietary biotin is scarce, biotin-producing commensals, e.g., E. coli, can rescue reduced ISC mitosis. Smvt and commensals also control intestinal tumor growth. Our findings suggest that direct modification of the gut microbiome by biotin can serve as an approach for the treatment of dysbiosis-promoted diseases and tumorigenesis control.
Collapse
Affiliation(s)
- Constantina Neophytou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus.
| |
Collapse
|
21
|
Phillippi DT, Daniel S, Pusadkar V, Youngblood VL, Nguyen KN, Azad RK, McFarlin BK, Lund AK. Inhaled diesel exhaust particles result in microbiome-related systemic inflammation and altered cardiovascular disease biomarkers in C57Bl/6 male mice. Part Fibre Toxicol 2022; 19:10. [PMID: 35135577 PMCID: PMC8827295 DOI: 10.1186/s12989-022-00452-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The gut microbiota plays a vital role in host homeostasis and is associated with inflammation and cardiovascular disease (CVD) risk. Exposure to particulate matter (PM) is a known mediator of inflammation and CVD and is reported to promote dysbiosis and decreased intestinal integrity. However, the role of inhaled traffic-generated PM on the gut microbiome and its corresponding systemic effects are not well-characterized. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) alters the gut microbiome and promotes microbial-related inflammation and CVD biomarkers. 4-6-week-old male C57Bl/6 mice on either a low-fat (LF, 10% fat) or high-fat (HF, 45% fat) diet were exposed via oropharyngeal aspiration to 35 μg DEP suspended in 35 μl saline or saline only (CON) 2x/week for 30 days. To determine whether probiotics could prevent diet or DEP exposure mediated alterations in the gut microbiome or systemic outcomes, a subset of animals on the HF diet were treated orally with 0.3 g/day (~ 7.5 × 108 CFU/day) of Winclove Ecologic® Barrier probiotics throughout the study. RESULTS Our results show that inhaled DEP exposure alters gut microbial profiles, including reducing Actinobacteria and expanding Verrucomicrobia and Proteobacteria. We observed increased circulating LPS, altered circulating cytokines (IL-1α, IL-3, IL-13, IL-15, G-CSF, LIF, MIP-2, and TNF-α), and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in DEP-exposed and/or HF diet mice. Furthermore, probiotics attenuated the observed reduction of Actinobacteria and expansion of Proteobacteria in DEP-exposed and HF-diet mice. Probiotics mitigated circulating cytokines (IL-3, IL-13, G-CSF, RANTES, and TNF- α) and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in respect to DEP-exposure and/or HF diet. CONCLUSION Key findings of this study are that inhaled DEP exposure alters small intestinal microbial profiles that play a role in systemic inflammation and early CVD biomarkers. Probiotic treatment in this study was fundamental in understanding the role of inhaled DEP on the microbiome and related systemic inflammatory and CVD biomarkers.
Collapse
Affiliation(s)
- Danielle T. Phillippi
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Sarah Daniel
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Vaidehi Pusadkar
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
| | - Victoria L. Youngblood
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Kayla N. Nguyen
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Rajeev K. Azad
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- Department of Mathematics, University of North Texas, Denton, TX 76203 USA
| | - Brian K. McFarlin
- Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- UNT Applied Physiology Laboratory, University of North Texas, Denton, TX 76203 USA
| | - Amie K. Lund
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| |
Collapse
|
22
|
Adsorption of extracellular proteases and pyocyanin produced by Pseudomonas aeruginosa using a macroporous magnesium oxide-templated carbon decreases cytotoxicity. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100160. [DOI: 10.1016/j.crmicr.2022.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
|
23
|
Prenatal versus Postnatal Initial Colonization of Healthy Neonates' Colon Ecosystem by the Enterobacterium Escherichia coli. Microbiol Spectr 2021; 9:e0037921. [PMID: 34817225 PMCID: PMC8612161 DOI: 10.1128/spectrum.00379-21] [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] [Indexed: 12/20/2022] Open
Abstract
The human colon is a microbial ecosystem whose initial bacterial colonization in neonates is an important step in establishing a beneficial microbiota for the body's health. This study investigated the occurrence of viable culturable Escherichia coli in first-day meconium versus subsequent days' stool to explore the prenatal versus postnatal initial colonization of the colon by E. coli in healthy neonates. E. coli occurrence was investigated on eosin-methylene blue (EMB) agar, followed by morphological and biochemical characterizations and phylogenetic analysis of 16S rRNA-encoding gene sequences. Viable culturable E. coli was not detected in meconium of healthy male or female neonates delivered either vaginally or by cesarean section. Neonates delivered surgically also showed no E. coli colonization on the second and third days, confirming postnatal colonization of the colon by this enterobacterium. E. coli's initial colonization in the colon of neonates delivered vaginally occurred on the second day, which can be attributed to inoculation from the vaginal canal during delivery and, in comparison to the colonization in neonates delivered surgically, leads to the inference that the bacterium is not originally found in meconium. This study suggests no viability of the meconium microbiome in healthy neonates, possibly due to antimicrobial action in the prenatal colon's meconium protecting babies' gut from infection during delivery. IMPORTANCE The results of this study suggest that the initial postnatal colonization of neonates' colon by beneficial bacteria is a naturally controlled process in which the prenatal colon's meconium might play a role in protecting against infection of the babies' gut during delivery.
Collapse
|
24
|
Fontana F, Mancabelli L, Lugli GA, Taracchini C, Alessandri G, Longhi G, Anzalone R, Viappiani A, Famo R, Brognan M, Micondo KH, Turroni F, Ventura M, D'Alfonso R, Milani C. Investigating the infant gut microbiota in developing countries: worldwide metagenomic meta-analysis involving infants living in sub-urban areas of Côte d'Ivoire. ENVIRONMENTAL MICROBIOLOGY REPORTS 2021; 13:626-636. [PMID: 34152069 PMCID: PMC8518733 DOI: 10.1111/1758-2229.12960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 06/13/2023]
Abstract
In recent decades, infants' gut microbiota has aroused constant scientific interest, primarily due to early- and long-term repercussions on the host health. In this context, nutritional challenges such as those found in less developed countries can influence infants' gut microbiota development, thus generating potentially critical health outcomes. However, comprehensive investigations regarding species-level differences in the infant gut microbiota's composition between urbanized and rural countries are still missing. In this study, 16S rRNA and Shallow Shotgun metagenomics sequencing were exploited to dissect the microbial community's species-level composition of 11 faecal samples collected from infants living in a semi-urban area of Sub-Saharan Africa, i.e. Côte d'Ivoire. Moreover, the generated data were coupled with those retrieved from public available metagenomic repositories, including two rural communities and 13 urban communities of industrialized countries. The meta-analysis led to the identification of Infant Species Community States Type (ISCSTs) and microbial species covariances, which were exploited to reveal key signatures of infants living in rural and semi-urban societies. Remarkably, analysis of rural and semi-urban datasets revealed shifts from ISCSTs prevalent in urbanized populations with putative health implications. Thus, indicating the need for population-wide investigations aimed to define the factors determining such potentially harmful gut microbial communities' signatures.
Collapse
Affiliation(s)
- Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- GenProbio srlParmaItaly
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Chiara Taracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- GenProbio srlParmaItaly
| | | | | | - Roch Famo
- Centre Médical Don Orione AnyamaAnyamaCôte d'Ivoire
| | - Marc Brognan
- Centre Médical Don Orione AnyamaAnyamaCôte d'Ivoire
| | | | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Rossella D'Alfonso
- Centre Médical Don Orione AnyamaAnyamaCôte d'Ivoire
- Pediatric Service of Hospital Military D'AbidjanAbidjanCôte d'Ivoire
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| |
Collapse
|
25
|
Oldenburg M, Rüchel N, Janssen S, Borkhardt A, Gössling KL. The Microbiome in Childhood Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:cancers13194947. [PMID: 34638430 PMCID: PMC8507905 DOI: 10.3390/cancers13194947] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
For almost 30 years, the term "holobiont" has referred to an ecological unit where a host (e.g., human) and all species living in or around it are considered together. The concept highlights the complex interactions between the host and the other species, which, if disturbed may lead to disease and premature aging. Specifically, the impact of microbiome alterations on the etiology of acute lymphoblastic leukemia (ALL) in children is not fully understood, but has been the focus of much research in recent years. In ALL patients, significant reductions in microbiome diversity are already observable at disease onset. It remains unclear whether such alterations at diagnosis are etiologically linked with leukemogenesis or simply due to immunological alteration preceding ALL onset. Regardless, all chemotherapeutic treatment regimens severely affect the microbiome, accompanied by severe side effects, including mucositis, systemic inflammation, and infection. In particular, dominance of Enterococcaceae is predictive of infections during chemotherapy. Long-term dysbiosis, like depletion of Faecalibacterium, has been observed in ALL survivors. Modulation of the microbiome (e.g., by fecal microbiota transplant, probiotics, or prebiotics) is currently being researched for potential protective effects. Herein, we review the latest microbiome studies in pediatric ALL patients.
Collapse
Affiliation(s)
- Marina Oldenburg
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (M.O.); (N.R.); (A.B.)
| | - Nadine Rüchel
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (M.O.); (N.R.); (A.B.)
| | - Stefan Janssen
- Algorithmic Bioinformatics, Department of Biology and Chemistry, Justus Liebig University Gießen, 35390 Gießen, Germany;
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (M.O.); (N.R.); (A.B.)
| | - Katharina L. Gössling
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (M.O.); (N.R.); (A.B.)
- Correspondence:
| |
Collapse
|
26
|
Song CH, Kim N, Nam RH, Choi SI, Yu JE, Nho H, Surh YJ. Changes in Microbial Community Composition Related to Sex and Colon Cancer by Nrf2 Knockout. Front Cell Infect Microbiol 2021; 11:636808. [PMID: 34249773 PMCID: PMC8261249 DOI: 10.3389/fcimb.2021.636808] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
The frequency of azoxymethane/dextran sulfate sodium (AOM/DSS)-induced carcinogenesis in male mice is higher than that in female mice. Previous studies have reported that 17β-estradiol inhibits tumorigenesis in males by modulating nuclear factor-erythroid 2-related factor 2 (Nrf2). This study aimed to investigate the changes in mouse gut microbiome composition based on sex, AOM/DSS-induced colorectal cancer (CRC), and Nrf2 genotype. The gut microbiome composition was determined by 16S rRNA gene sequencing fecal samples obtained at week 16 post-AOM administration. In terms of sex differences, our results showed that the wild-type (WT) male control mice had higher alpha diversity (i.e. Chao1, Shannon, and Simpson) than the WT female control mice. The linear discriminant analysis effect size (LEfSe) results revealed that the abundances of Akkermansia muciniphila and Lactobacillus murinus were higher in WT male control mice than in WT female controls. In terms of colon tumorigenesis, the alpha diversity of the male CRC group was lower than that of the male controls in both WT and Nrf2 KO, but did not show such changes in females. Furthermore, the abundance of A. muciniphila was higher in male CRC groups than in male controls in both WT and Nrf2 KO. The abundance of Bacteroides vulgatus was higher in WT CRC groups than in WT controls in both males and females. However, the abundance of L. murinus was lower in WT female CRC and Nrf2 KO male CRC groups than in its controls. The abundance of A. muciniphila was not altered by Nrf2 KO. In contrast, the abundances of L. murinus and B. vulgatus were changed differently by Nrf2 KO depending on sex and CRC. Interestingly, L. murinus showed negative correlation with tumor numbers in the whole colon. In addition, B. vulgatus showed positive correlation with inflammatory markers (i.e. myeloperoxidase and IL-1β levels), tumor numbers, and high-grade adenoma, especially, developed mucosal and submucosal invasive adenocarcinoma at the distal part of the colon. In conclusion, Nrf2 differentially alters the gut microbiota composition depending on sex and CRC induction.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jeong Eun Yu
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Heewon Nho
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
27
|
Di Menna L, Busceti CL, Ginerete RP, D'Errico G, Orlando R, Alborghetti M, Bruno V, Battaglia G, Fornai F, Leoni L, Rampioni G, Visca P, Monn JA, Nicoletti F. The bacterial quorum sensing molecule, 2-heptyl-3-hydroxy-4-quinolone (PQS), inhibits signal transduction mechanisms in brain tissue and is behaviorally active in mice. Pharmacol Res 2021; 170:105691. [PMID: 34044128 DOI: 10.1016/j.phrs.2021.105691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/25/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022]
Abstract
Interkingdom communication between bacteria and host organisms is one of the most interesting research topics in biology. Quorum sensing molecules produced by Gram-negative bacteria, such as acylated homoserine lactones and quinolones, have been shown to interact with host cell receptors, stimulating innate immunity and bacterial clearance. To our knowledge, there is no evidence that these molecules influence CNS function. Here, we have found that low micromolar concentrations of the Pseudomonas aeruginosa quorum sensing autoinducer, 2-heptyl-3-hydroxy-4-quinolone (PQS), inhibited polyphosphoinositide hydrolysis in mouse brain slices, whereas four selected acylated homoserine lactones were inactive. PQS also inhibited forskolin-stimulated cAMP formation in brain slices. We therefore focused on PQS in our study. Biochemical effects of PQS were not mediated by the bitter taste receptors, T2R4 and T2R16. Interestingly, submicromolar concentrations of PQS could be detected in the serum and brain tissue of adult mice under normal conditions. Levels increased in five selected brain regions after single i.p. injection of PQS (10 mg/kg), peaked after 15 min, and returned back to normal between 1 and 4 h. Systemically administered PQS reduced spontaneous locomotor activity, increased the immobility time in the forced swim test, and largely attenuated motor response to the psychostimulant, methamphetamine. These findings offer the first demonstration that a quorum sensing molecule specifically produced by Pseudomonas aeruginosa is centrally active and influences cell signaling and behavior. Quorum sensing autoinducers might represent new interkingdom signaling molecules between ecological communities of commensal, symbiotic, and pathogenic microorganisms and the host CNS.
Collapse
Affiliation(s)
| | | | | | | | - R Orlando
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Phamacology, Sapienza University, Roma, Italy
| | - M Alborghetti
- Department of Neuroscience, Mental Health, and Sensory Organs, Sapienza University, Italy
| | - V Bruno
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Phamacology, Sapienza University, Roma, Italy
| | - G Battaglia
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Phamacology, Sapienza University, Roma, Italy
| | - F Fornai
- IRCCS Neuromed, Pozzilli, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - L Leoni
- Department of Science, Roma Tre University, Roma, Italy
| | - G Rampioni
- Department of Science, Roma Tre University, Roma, Italy; Fondazione Santa Lucia IRCCS, Roma, Italy
| | - P Visca
- Department of Science, Roma Tre University, Roma, Italy; Fondazione Santa Lucia IRCCS, Roma, Italy
| | | | - F Nicoletti
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Phamacology, Sapienza University, Roma, Italy.
| |
Collapse
|
28
|
Kato S, Sato T, Fujita H, Kawatani M, Yamada Y. Effects of GLP-1 receptor agonist on changes in the gut bacterium and the underlying mechanisms. Sci Rep 2021; 11:9167. [PMID: 33911125 PMCID: PMC8080802 DOI: 10.1038/s41598-021-88612-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/13/2021] [Indexed: 01/07/2023] Open
Abstract
There is a close relationship between the gut microbiota and metabolic disorders. In this study, acute administration of the glucagon-like peptide-1 receptor agonist (GLP-1RA) liraglutide to mice increased the cecal levels of caseinolytic protease B, a component of Escherichia coli, and of norepinephrine. Chemical sympathectomy blocked these events. Norepinephrine was found to pass into the intestinal lumen in vitro. c-Fos staining of the intermediolateral nucleus was identified as indirect evidence of sympathetic nervous system activation of the intestinal tract by GLP-1RA. Under normal conditions, the increase in E. coli did not affect the host. However, in mice with colitis, bacterial translocation was observed with attenuation of tight junction gene expression. This is the first study to investigate the unique underlying mechanisms related the effects of GLP-1RA on changes in the gut bacterium.
Collapse
Affiliation(s)
- Shunsuke Kato
- Departments of Endocrinology, Diabetes, and Geriatric Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Takehiro Sato
- Departments of Endocrinology, Diabetes, and Geriatric Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroki Fujita
- Departments of Endocrinology, Diabetes, and Geriatric Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Masahiro Kawatani
- Departments of Neurophysiology, Akita University Graduate School of Medicine, Akita, Japan
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yuichiro Yamada
- Departments of Endocrinology, Diabetes, and Geriatric Medicine, Akita University Graduate School of Medicine, Akita, Japan.
- Kansai Electric Power Medical Research Institute, 2-1-7 Fukushima, Fukushima-ku, Osaka, Japan.
| |
Collapse
|
29
|
Adsorption of Phenazines Produced by Pseudomonas aeruginosa Using AST-120 Decreases Pyocyanin-Associated Cytotoxicity. Antibiotics (Basel) 2021; 10:antibiotics10040434. [PMID: 33924459 PMCID: PMC8068879 DOI: 10.3390/antibiotics10040434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 02/02/2023] Open
Abstract
AST-120 (Kremezin) is used to treat progressive chronic kidney disease by adsorbing uremic toxin precursors produced by the gut microbiota, such as indole and phenols. Previously, we found that AST-120 decreased drug tolerance and virulence in Escherichia coli by adsorbing indole. Here, we show that AST-120 adsorbs phenazine compounds, such as pyocyanin, produced by Pseudomonas aeruginosa including multidrug-resistant P. aeruginosa strains, and suppresses pyocyanin-associated toxicity in A-549 (alveolar adenocarcinoma) and Caco-2 (colon adenocarcinoma) cells. Addition of fosfomycin, colistin and amikacin, which are often used to treat P. aeruginosa, inhibited the bacterial growth, regardless of the presence or absence of AST-120. These results suggest a further benefit of AST-120 that supports anti-Pseudomonas chemotherapy in addition to that of E. coli and propose a novel method to treat P. aeruginosa infection.
Collapse
|
30
|
Mayorgas A, Dotti I, Martínez-Picola M, Esteller M, Bonet-Rossinyol Q, Ricart E, Salas A, Martínez-Medina M. A Novel Strategy to Study the Invasive Capability of Adherent-Invasive Escherichia coli by Using Human Primary Organoid-Derived Epithelial Monolayers. Front Immunol 2021; 12:646906. [PMID: 33854511 PMCID: PMC8039293 DOI: 10.3389/fimmu.2021.646906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last decades, Adherent-Invasive Escherichia coli (AIEC) has been linked to the pathogenesis of Crohn’s Disease. AIEC’s characteristics, as well as its interaction with the gut immune system and its role in intestinal epithelial barrier dysfunction, have been extensively studied. Nevertheless, the currently available techniques to investigate the cross-talk between this pathogen and intestinal epithelial cells (IECs) are based on the infection of immortalized cell lines. Despite their many advantages, cell lines cannot reproduce the conditions in tissues, nor do they reflect interindividual variability or gut location-specific traits. In that sense, the use of human primary cultures, either healthy or diseased, offers a system that can overcome all of these limitations. Here, we developed a new infection model by using freshly isolated human IECs. For the first time, we generated and infected monolayer cultures derived from human colonic organoids to study the mechanisms and effects of AIEC adherence and invasion on primary human epithelial cells. To establish the optimal conditions for AIEC invasion studies in human primary organoid-derived epithelial monolayers, we designed an infection-kinetics study to assess the infection dynamics at different time points, as well as with two multiplicities of infection (MOI). Overall, this method provides a model for the study of host response to AIEC infections, as well as for the understanding of the molecular mechanisms involved in adhesion, invasion and intracellular replication. Therefore, it represents a promising tool for elucidating the cross-talk between AIEC and the intestinal epithelium in healthy and diseased tissues.
Collapse
Affiliation(s)
- Aida Mayorgas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | - Isabella Dotti
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | - Marta Martínez-Picola
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | - Miriam Esteller
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | - Queralt Bonet-Rossinyol
- Laboratory of Molecular Microbiology, Department of Biology, Universitat de Girona, Girona, Spain
| | - Elena Ricart
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | - Azucena Salas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBER-EHD, Barcelona, Spain
| | | |
Collapse
|
31
|
Shahriar S, Ahsan T, Khan A, Akhteruzzaman S, Shehreen S, Sajib AA. Aspartame, acesulfame K and sucralose- influence on the metabolism of Escherichia coli. Metabol Open 2020; 8:100072. [PMID: 33336183 PMCID: PMC7732866 DOI: 10.1016/j.metop.2020.100072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/18/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Gut microbes play a crucial role in the maintenance of human health. Components in the diet of the host affect their metabolism and diversity. Here, we investigated the influences of three commonly used non-caloric artificial sweeteners-aspartame, acesulfame K and sucralose-on the growth and metabolism of an omnipresent gut microbe Escherichia coli K-12. Methods: Growth of E. coli in the presence of aspartame, acesulfame K and sucralose in media was assessed and the influences of these artificial sweeteners on metabolism were investigated by relative expression analysis of genes encoding the rate limiting steps of important metabolic pathways as well as their global metabolomic profiles. Results: As a whole, E. coli growth was inhibited by aspartame and induced by acesulfame potassium, while the effect of sucralose on growth was less prominent. Although the expressions of multiple key enzymes that regulate important metabolic pathways were significantly altered by all three sweeteners, acesulfame K caused the most notable changes in this regard. In multivariate analysis with the metabolite profiles, the sucralose-treated cells clustered the closest to the untreated cells, while the acesulfame potassium treated cells were the most distant. These sweeteners affect multiple metabolic pathways in E. coli, which include propanoate, phosphonate, phosphinate and fatty acid metabolism, pentose phosphate pathway, and biosynthesis of several amino acids including lysine and the aromatic amino acids. Similar to the gene expression pattern, acesulfame potassium treated E. coli showed the largest deviation in their metabolite profiles compared to the untreated cells.
Collapse
Affiliation(s)
- Shayan Shahriar
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Tamim Ahsan
- Department of Genetic Engineering & Biotechnology, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Abira Khan
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Sharif Akhteruzzaman
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Saadlee Shehreen
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
32
|
Javed I, Cui X, Wang X, Mortimer M, Andrikopoulos N, Li Y, Davis TP, Zhao Y, Ke PC, Chen C. Implications of the Human Gut-Brain and Gut-Cancer Axes for Future Nanomedicine. ACS NANO 2020; 14:14391-14416. [PMID: 33138351 DOI: 10.1021/acsnano.0c07258] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent clinical and pathological evidence have implicated the gut microbiota as a nexus for modulating the homeostasis of the human body, impacting conditions from cancer and dementia to obesity and social behavior. The connections between microbiota and human diseases offer numerous opportunities in medicine, most of which have limited or no therapeutic solutions available. In light of this paradigm-setting trend in science, this review aims to provide a comprehensive and timely summary of the mechanistic pathways governing the gut microbiota and their implications for nanomedicines targeting cancer and neurodegenerative diseases. Specifically, we discuss in parallel the beneficial and pathogenic relationship of the gut microbiota along the gut-brain and gut-cancer axes, elaborate on the impact of dysbiosis and the gastrointestinal corona on the efficacy of nanomedicines, and highlight a molecular mimicry that manipulates the universal cross-β backbone of bacterial amyloid to accelerate neurological disorders. This review further offers a forward-looking section on the rational design of cancer and dementia nanomedicines exploiting the gut-brain and gut-cancer axes.
Collapse
Affiliation(s)
- Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiaoyu Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Nikolaos Andrikopoulos
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuhuan Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
33
|
Agarwal A, Agashe D. The red flour beetle Tribolium castaneum: A model for host-microbiome interactions. PLoS One 2020; 15:e0239051. [PMID: 33006995 PMCID: PMC7531845 DOI: 10.1371/journal.pone.0239051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/30/2020] [Indexed: 11/18/2022] Open
Abstract
A large body of ongoing research focuses on understanding the mechanisms and processes underlying host-microbiome interactions, and predicting their ecological and evolutionary outcomes. To draw general conclusions about such interactions and understand how they are established, we must synthesize information from a diverse set of species. We analysed the microbiome of an important insect model-the red flour beetle Tribolium castaneum-which is a widespread generalist pest of stored cereals. The beetles complete their entire life cycle in flour, which thus serves multiple functions: habitat, food, and a source of microbes. We determined key factors that shape the T. castaneum microbiome, established protocols to manipulate it, and tested its consequences for host fitness. We show that the T. castaneum microbiome is derived from flour-acquired microbes, and varies as a function of (flour) resource and beetle density. Beetles gain multiple fitness benefits from their microbiome, such as higher fecundity, egg survival, and lifespan; and reduced cannibalism. In contrast, the microbiome has a limited effect on development rate, and does not enhance pathogen resistance. Importantly, the benefits are derived only from microbes in the ancestral resource (wheat flour), and not from novel resources such as finger millet, sorghum, and corn. Notably, the microbiome is not essential for beetle survival and development under any of the tested conditions. Thus, the red flour beetle is a tractable model system to understand the ecology, evolution and mechanisms of host-microbiome interactions, while closely mimicking the host species' natural niche.
Collapse
Affiliation(s)
- Aparna Agarwal
- National Centre for Biological Sciences, Bangalore, India
| | - Deepa Agashe
- National Centre for Biological Sciences, Bangalore, India
| |
Collapse
|
34
|
Javed I, Zhang Z, Adamcik J, Andrikopoulos N, Li Y, Otzen DE, Lin S, Mezzenga R, Davis TP, Ding F, Ke PC. Accelerated Amyloid Beta Pathogenesis by Bacterial Amyloid FapC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001299. [PMID: 32999841 PMCID: PMC7509637 DOI: 10.1002/advs.202001299] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/10/2020] [Indexed: 05/03/2023]
Abstract
The gut-brain axis has attracted increasing attention in recent years, fueled by accumulating symptomatic, physiological, and pathological findings. In this study, the aggregation and toxicity of amyloid beta (Aβ), the pathogenic peptide associated with Alzheimer's disease (AD), seeded by FapC amyloid fragments (FapCS) of Pseudomonas aeruginosa that colonizes the gut microbiome through infections are examined. FapCS display favorable binding with Aβ and a catalytic capacity in seeding the peptide amyloidosis. Upon seeding, twisted Aβ fibrils assume a much-shortened periodicity approximating that of FapC fibrils, accompanied by a 37% sharp rise in the fibrillar diameter, compared with the control. The robust seeding capacity for Aβ by FapCS and the biofilm fragments derived from P. aeruginosa entail abnormal behavior pathology and immunohistology, as well as impaired cognitive function of zebrafish. Together, the data offer the first concrete evidence of structural integration and inheritance in peptide cross-seeding, a crucial knowledge gap in understanding the pathological correlations between different amyloid diseases. The catalytic role of infectious bacteria in promoting Aβ amyloidosis may be exploited as a potential therapeutic target, while the altered mesoscopic signatures of Aβ fibrils may serve as a prototype for molecular assembly and a biomarker for screening bacterial infections in AD.
Collapse
Affiliation(s)
- Ibrahim Javed
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Zhenzhen Zhang
- Department of Physics and AstronomyClemson UniversityClemsonSC29634USA
| | - Jozef Adamcik
- Food & Soft MaterialsDepartment of Health Science & TechnologyETH ZurichSchmelzbergstrasse 9, LFO, E23Zurich8092Switzerland
| | - Nicholas Andrikopoulos
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Yuhuan Li
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Center (iNANO)University of AarhusAarhus CAarhus8000Denmark
| | - Sijie Lin
- College of Environmental Science and EngineeringBiomedical Multidisciplinary Innovation Research InstituteShanghai East HospitalShanghai Institute of Pollution Control and Ecological SecurityTongji University1239 Siping RoadShanghai200092China
| | - Raffaele Mezzenga
- Food & Soft MaterialsDepartment of Health Science & TechnologyETH ZurichSchmelzbergstrasse 9, LFO, E23Zurich8092Switzerland
| | - Thomas P. Davis
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Feng Ding
- Department of Physics and AstronomyClemson UniversityClemsonSC29634USA
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Zhongshan HospitalFudan University111 Yixueyuan Rd, Xuhui DistrictShanghai200032China
| |
Collapse
|
35
|
Suzhaeva LV, Makarova MA, Kaftyreva LA. [Phylogenetic groups and virulence genes of Escherichia coli strains isolated from the children gut microbiota.]. Klin Lab Diagn 2020; 65:251-257. [PMID: 32227732 DOI: 10.18821/0869-2084-2020-65-4-251-257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 11/17/2022]
Abstract
Escherichia coli is characterized by a wide intraspecific diversity. The species includes both commensals and pathogens that cause diarrhea and extra-intestinal diseases. Pathogenic strains differ from non-pathogenic ones by the presence of virulence factors and their genes. The phylogenetic structure of the species is represented by four main groups (A, B1, B2, D), which differ in their prevalence among residents of different geographical regions. Pathogenic members of the species have been studied in detail, while non-pathogenic strains have not received such attention. This report presents the results of a study of 511 E. coli strains isolated from the gut microbiota of children without diarrhea and urinary tract infections, aged from 1 month to 17 years, living in St. Petersburg. The main phylogenetic groups were determined by PCR, and E. coli virulence genes associated with diarrhea and extra-intestinal diseases were identified. Results: population structure of E. coli is represented by the following groups: A-33.3%, B1-6.7%, B2-34.0%, D-26%. In the studied population 2.5% of strains belonded to EPEC and 4.5% to EAggEC. EPEC virulence genes were more often detected in strains of phylogroup B1, and EAggEC virulence genes in isolates of phylogroup D. The prevalence of extra - intestinal virulence genes was as follows: pap - 29.5%; sfa - 19.8%; afa - 3.3%; hly - 20.9%; cnf - 17.4%; aer-20.0%. The pap, sfa, hly, and cnf genes were detected mostly in the B2 phylogenetic group. Obtained data shows the similarity of E. coli phylogenetic groups structure in St. Petersburg with E. coli populations isolated from residents of Paris and Sydney. Analysis of the virulence genes prevalence showed the dependence of their presence on the genetic background bacteria.
Collapse
Affiliation(s)
- L V Suzhaeva
- Saint-Petersburg Pasteur Institute, 197101, Saint-Petersburg, Russia
| | - M A Makarova
- Saint-Petersburg Pasteur Institute, 197101, Saint-Petersburg, Russia.,State Educational Institution of the Higher Professional Education «North-Western state medical University n.a. I.I. Mechnikov» of the Ministry of Health of the Russian Federation
| | - L A Kaftyreva
- Saint-Petersburg Pasteur Institute, 197101, Saint-Petersburg, Russia.,State Educational Institution of the Higher Professional Education «North-Western state medical University n.a. I.I. Mechnikov» of the Ministry of Health of the Russian Federation
| |
Collapse
|
36
|
Kundu M, Gucchait A, Misra AK. Convergent synthesis of a pentasaccharide corresponding to the cell wall O-polysaccharide of enteropathogenic Escherichia coli O115. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.130952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|