51
|
Pitts MG, D'Orazio SEF. Enrichment of Neutrophils and Monocytes From the Liver Following Either Oral or Intravenous Listeria monocytogenes Infection. ACTA ACUST UNITED AC 2021; 130:e102. [PMID: 32710703 DOI: 10.1002/cpim.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Listeria monocytogenes is a foodborne pathogen that causes serious, often deadly, systemic disease in susceptible individuals such as neonates and the elderly. These facultative intracellular bacteria have been an invaluable tool in immunology research for more than three decades. Intravenous (i.v.) injection is the most commonly used transmission route in mice, but oral models of infection have also been developed in recent years, and these may be more appropriate for many studies. This article includes detailed instructions for use of either foodborne or i.v. inoculation of mice and discusses the rationale for choosing either model. Additionally, a protocol is provided for enrichment of neutrophils and monocytes from the infected liver in a manner that allows for determination of bacterial burden while still providing sufficient cells for use in flow cytometric analysis or in vitro assays. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Foodborne L. monocytogenes infection Support Protocol 1: Preparing L. monocytogenes for foodborne infection Basic Protocol 2: Intravenous L. monocytogenes infection Support Protocol 2: Preparing L. monocytogenes for intravenous infection Basic Protocol 3: Enrichment of non-parenchymal cells from the infected liver.
Collapse
Affiliation(s)
- Michelle G Pitts
- University of Kentucky College of Pharmacy, Department of Pharmaceutical Sciences, Lexington, Kentucky
| | - Sarah E F D'Orazio
- University of Kentucky College of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Lexington, Kentucky
| |
Collapse
|
52
|
CBP22, a Novel Bacteriocin Isolated from Clostridium butyricum ZJU-F1, Protects against LPS-Induced Intestinal Injury through Maintaining the Tight Junction Complex. Mediators Inflamm 2021; 2021:8032125. [PMID: 34158805 PMCID: PMC8187061 DOI: 10.1155/2021/8032125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/14/2020] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
A novel bacteriocin secreted by Clostridium butyricum ZJU-F1 was isolated using ammonium sulfate fractionation, cation exchange chromatography, affinity chromatography, and reverse-phase high-performance liquid chromatography (RP-HPLC). The bacteriocin, named CBP22, contained 22 amino acids with the sequence PSAWQITKCAGSIAWALGSGIF. Analysis of its structure and physicochemical properties indicated that CBP22 had a molecular weight of 2264.63 Da and a +1 net charge. CBP22 showed activity against E. col K88, E. coli ATCC25922, and S. aureus ATCC26923. The effects and potential mechanisms of bacteriocin CBP22 on the innate immune response were investigated with a lipopolysaccharide- (LPS-) induced mouse model. The results showed that pretreatment with CBP22 prevented LPS-induced impairment in epithelial tissues and significantly reduced serum levels of IgG, IgA, IgM, TNF-α, and sIgA. Moreover, CBP22 treatment increased the expression of the zonula occludens and reduced permeability as well as apoptosis in the jejunum in LPS-treated mice. In summary, CBP22 inhibits the intestinal injury and prevents the gut barrier dysfunction induced by LPS, suggesting the potential use of CBP22 for treating intestinal damage.
Collapse
|
53
|
He Y, Yang Q, Tian L, Zhang Z, Qiu L, Tao X, Wei H. Protection of surface layer protein from Enterococcus faecium WEFA23 against Listeria monocytogenes CMCC54007 infection by modulating intestinal permeability and immunity. Appl Microbiol Biotechnol 2021; 105:4269-4284. [PMID: 33990856 DOI: 10.1007/s00253-021-11240-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Enterococcus faecium WEFA23 was previously found effectively against adherence and colonization of Listeria monocytogenes CMCC54007, which might be closely related to its surface layer protein (SLP). In this study, the protective of SLP of E. faecium WEFA23 against infection of L. monocytogenes CMCC54007 was systemically investigated. In vitro assay showed that SLP actively inhibited L. monocytogenes internalization into Caco-2 cell line, with decreasing mRNA level of pro-inflammation cytokines and virulence factors and restoring destroyed intestinal barrier. In vivo assay through excluding SLP of E. faecium WEFA23 by 5 M LiCl represented that SLP increased body weight, reduced mortality and cell counts of L. monocytogenes CMCC54007 in tissues of mice. Further researches showed that SLP protected against L. monocytogenes CMCC54007 infection by modulation of intestinal permeability and immunity, namely, it decreased fluorescein isothiocyanate (FITC)-Dextran in serum, ameliorated destroyed colon structure, and increased number of goblet cells and protein level of TJ protein (Claudin-1, Occludin, and ZO-1) in colon. For immunity, SLP decreased number of CD4+ and CD8+ T cells in liver, mRNA level, and content of pro-inflammatory factors IL-6, IL-1β, IFN-γ ,TNF-α, and NO, and restored the structure of liver and spleen. Key Points•SLP of E. faecium inhibited L. monocytogenes internalization and colonization•SLP of E. faecium ameliorated host intestinal barrier dysfunction•SLP of E. faecium decreased pro-inflammatory cytokines and cells.
Collapse
Affiliation(s)
- Yao He
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Qin Yang
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Linlin Tian
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Zhihong Zhang
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Liang Qiu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, People's Republic of China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Hua Wei
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China.
| |
Collapse
|
54
|
Lopes-Luz L, Mendonça M, Bernardes Fogaça M, Kipnis A, Bhunia AK, Bührer-Sékula S. Listeria monocytogenes: review of pathogenesis and virulence determinants-targeted immunological assays. Crit Rev Microbiol 2021; 47:647-666. [PMID: 33896354 DOI: 10.1080/1040841x.2021.1911930] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Listeria monocytogenes is one of the most invasive foodborne pathogens and is responsible for numerous outbreaks worldwide. Most of the methods to detect this bacterium in food require selective enrichment using traditional bacterial culture techniques that can be time-consuming and labour-intensive. Moreover, molecular methods are expensive and need specific technical knowledge. In contrast, immunological approaches are faster, simpler, and user-friendly alternatives and have been developed for the detection of L. monocytogenes in food, environmental, and clinical samples. These techniques are dependent on the constitutive expression of L. monocytogenes antigens and the specificity of the antibodies used. Here, updated knowledge on pathogenesis and the key immunogenic virulence determinants of L. monocytogenes that are used for the generation of monoclonal and polyclonal antibodies for the serological assay development are summarised. In addition, immunological approaches based on enzyme-linked immunosorbent assay, immunofluorescence, lateral flow immunochromatographic assays, and immunosensors with relevant improvements are highlighted. Though the sensitivity and specificity of the assays were improved significantly, methods still face many challenges that require further validation before use.
Collapse
Affiliation(s)
- Leonardo Lopes-Luz
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| | - Marcelo Mendonça
- Curso de Medicina Veterinária, Universidade Federal do Agreste de Pernambuco, Garanhuns, Brasil
| | | | - André Kipnis
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| | - Arun K Bhunia
- Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Samira Bührer-Sékula
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| |
Collapse
|
55
|
Pizarro-Cerdá J. Pascale Cossart: Listeria monocytogenes, host-pathogen interactions & beyond. Cell Microbiol 2021; 22:e13165. [PMID: 32185896 DOI: 10.1111/cmi.13165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Javier Pizarro-Cerdá
- Yersinia Research Unit, Institut Pasteur, Paris, France.,Plague Maintenance, Spread and Evolution Institut Pasteur International Unit, Paris, France.,Plague and Other Yersinioses French National Reference Laboratory, Institut Pasteur, Paris, France.,Yersinia WHO Collaborative Research and Reference Centre, Institut Pasteur, Paris, France
| |
Collapse
|
56
|
Feeding in spatangoids: the case of Abatus Cordatus in the Kerguelen Islands (Southern Ocean). Polar Biol 2021. [DOI: 10.1007/s00300-021-02841-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
57
|
Listeria spp. Isolated from Tonsils of Wild Deer and Boars: Genomic Characterization. Appl Environ Microbiol 2021; 87:AEM.02651-20. [PMID: 33397708 DOI: 10.1128/aem.02651-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 01/11/2023] Open
Abstract
Listeria monocytogenes is a major human and animal foodborne pathogen. However, data from environmental reservoirs remain scarce. Here, we used whole-genome sequencing to characterize Listeria species isolates recovered over 1 year from wild animals in their natural habitats in Spain. Three different Listeria spp. (L. monocytogenes [n = 19], Listeria ivanovii subsp. londoniensis [n = 4], and Listeria innocua [n = 3]) were detected in 23 animal tonsils (9 deer, 14 wild boars) and 2 feeding troughs. No Listeria species was detected in feces. L. monocytogenes was detected in tonsils of 44.4% (8 out of 18) of deer and 40.7% (11 out of 27) of wild boars. L. monocytogenes isolates belonged to 3 different core genome multilocus sequence typing (cgMLST) types (CTs) of 3 distinct sublineages (SL1, SL387, and SL155) from lineages I and II. While cgMLST type L1-SL1-ST1-CT5279 (IVb; clonal complex 1 [CC1]) occurred only in one animal, types L1-SL387-ST388-CT5239 (IVb; CC388) and L2-SL155-ST155-CT1170 (IIa; CC155) were retrieved from multiple animals. In addition, L1-SL387-ST388-CT5239 (IVb; CC388) isolates were collected 1 year apart, revealing their long-term occurrence within the animal population and/or environmental reservoir. The presence of identical L. monocytogenes strains in deer and wild boars suggests contamination from a common food or environmental source, although interhost transmission cannot be excluded. Pathogenicity islands LIPI-1, LIPI-3, and LIPI-4 were present in 100%, 5%, and 79% of the L. monocytogenes isolates, respectively, and all L. monocytogenes lineage II isolates (n = 3) carried SSI-1 stress islands. This study highlights the need for monitoring L. monocytogenes environmental contamination and the importance of tonsils as a possible L. monocytogenes intrahost reservoir.IMPORTANCE Listeria monocytogenes is a foodborne bacterial pathogen responsible for listeriosis. Whole-genome sequencing has been extensively used in public health and food industries to characterize circulating Listeria isolates, but genomic data on isolates occurring in natural environments and wild animals are still scarce. Here, we show that wild animals carry pathogenic Listeria and that the same genotypes can be found at different time points in different host species. This work highlights the need of Listeria species monitoring of environmental contamination and the importance of tonsils as a possible L. monocytogenes intrahost reservoir.
Collapse
|
58
|
Making Sense of the Biodiversity and Virulence of Listeria monocytogenes. Trends Microbiol 2021; 29:811-822. [PMID: 33583696 DOI: 10.1016/j.tim.2021.01.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/21/2023]
Abstract
Listeria monocytogenes is a foodborne pathogen responsible for listeriosis, an infection that can manifest in humans as bacteremia, meningoencephalitis in immunocompromised patients and the elderly, and fetal-placental infection in pregnant women. Reference strains from this facultative intracellular bacterium have been instrumental in the investigation of basic mechanisms in microbiology, immunology, and cell biology. The integration of bacterial population genomics with environmental, epidemiological, and clinical data allowed the uncovering of new factors involved in the virulence of L. monocytogenes and its adaptation to different environments. This review illustrates how these investigations have led to a better understanding of the bacterium's virulence and the driving forces that shaped it.
Collapse
|
59
|
Huang J, Zhou C, Zhou G, Li H, Ye K. Effect of Listeria monocytogenes on intestinal stem cells in the co-culture model of small intestinal organoids. Microb Pathog 2021; 153:104776. [PMID: 33548482 DOI: 10.1016/j.micpath.2021.104776] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 02/08/2023]
Abstract
Listeria monocytogenes is a foodborne pathogen that causes systemic infections by crossing the intestinal barrier. However, in vitro analysis of the interaction of L. monocytogenes and small intestinal epithelium has yet to be fully elucidated. To study host responses from intestinal epithelium during L. monocytogenes infection, we used the co-culture model of small intestinal organoids and L. monocytogenes. Results showed that L. monocytogenes mediated damage to intestinal epithelium, especially intestinal stem cells. L. monocytogenes was found to reduce budding rate and increase mortality of organoids. Moreover, it affected the proliferation of epithelial cells and numbers of secretory cells. In addition, it was demonstrated that L. monocytogenes stimulated a reduction in the number of Lgr5+ stem cells. Furthermore, L. monocytogenes affected the expression of Hes1, Math1 and Sox9 to interfere with the differentiation of intestinal stem cells. Collectively, our findings reveal the effects of L. monocytogenes infection on intestinal stem cells and demonstrate that small intestinal organoid is a suitable experimental model for studying intestinal epithelium-pathogen interactions.
Collapse
Affiliation(s)
- Jie Huang
- Key Laboratory of Meat Processing and Quality Control, MOE, China-US Joint Research Center for Food Safety and Quality, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology; Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Cong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, China-US Joint Research Center for Food Safety and Quality, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology; Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, China-US Joint Research Center for Food Safety and Quality, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology; Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Haokun Li
- Key Laboratory of Meat Processing and Quality Control, MOE, China-US Joint Research Center for Food Safety and Quality, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology; Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Keping Ye
- Key Laboratory of Meat Processing and Quality Control, MOE, China-US Joint Research Center for Food Safety and Quality, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology; Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
60
|
Chow JTH, Gall AR, Johnson AK, Huynh TN. Characterization of Listeria monocytogenes isolates from lactating dairy cows in a Wisconsin farm: Antibiotic resistance, mammalian cell infection, and effects on the fecal microbiota. J Dairy Sci 2021; 104:4561-4574. [PMID: 33516554 DOI: 10.3168/jds.2020-18885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 11/04/2020] [Indexed: 12/26/2022]
Abstract
Listeria monocytogenes is an invasive foodborne pathogen that is ubiquitously present in the dairy farm environment. Although cattle are a reservoir of L. monocytogenes, most adult animals do not exhibit clinical symptoms, suggesting a homeostasis between this pathogen and the bovine gastrointestinal ecosystem. Nevertheless, substantial prevalence of L. monocytogenes fecal shedding by dairy cattle has been reported in many studies, posing threats of transmission within the herd and contamination of the human food supply. Accordingly, understanding the L. monocytogenes ecology within the bovine gastrointestinal tract is important to prevent clinical illness in the animal host, reduce transmission, and guide intervention strategies. In this study, we conducted a longitudinal sampling of fecal samples from 20 lactating dairy cows in one Wisconsin farm over a 29-d period and found a strikingly high incidence of L. monocytogenes shedding, in 90% of sampled animals. The L. monocytogenes isolates were genetically diverse, representing all common serotypes previously identified from cattle. Additionally, most tested isolates were resistant to ampicillin, and a few were also resistant to gentamicin or trimethoprim/sulfamethoxazole. Most isolates effectively infected human epithelial cells (Caco-2) and murine fibroblasts (L2), suggesting that they are all capable of causing systemic infection if the intestinal barrier is breached. Finally, we investigated the effects of L. monocytogenes colonization on the gastrointestinal tract microbiota by analyzing the fecal bacterial communities of some shedding and nonshedding cows. Whereas L. monocytogenes did not affect the α and β diversity of tested animals, a subset of shedding cows exhibited different abundances of certain operational taxonomic units within the Bacteroidetes and Firmicutes phyla compared with nonshedding cows. Overall, our findings highlight the threat of antibiotic resistance among some L. monocytogenes isolates, emphasize the need for a strain-specific approach in listeriosis treatment, and suggest the potential negative influence of subclinical L. monocytogenes carriage on animal gut health.
Collapse
Affiliation(s)
- Justin T H Chow
- Department of Food Science, University of Wisconsin, Madison 53706
| | - Aaron R Gall
- Department of Food Science, University of Wisconsin, Madison 53706
| | | | - TuAnh N Huynh
- Department of Food Science, University of Wisconsin, Madison 53706.
| |
Collapse
|
61
|
Hery L, Guidez A, Durand AA, Delannay C, Normandeau-Guimond J, Reynaud Y, Issaly J, Goindin D, Legrave G, Gustave J, Raffestin S, Breurec S, Constant P, Dusfour I, Guertin C, Vega-Rúa A. Natural Variation in Physicochemical Profiles and Bacterial Communities Associated with Aedes aegypti Breeding Sites and Larvae on Guadeloupe and French Guiana. MICROBIAL ECOLOGY 2021; 81:93-109. [PMID: 32621210 PMCID: PMC7794107 DOI: 10.1007/s00248-020-01544-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/15/2020] [Indexed: 05/10/2023]
Abstract
Aedes aegypti develop in aquatic habitats in which mosquito larvae are exposed to physicochemical elements and microorganisms that may influence their life cycle and their ability to transmit arboviruses. Little is known about the natural bacterial communities associated with A. aegypti or their relation to the biotic and abiotic characteristics of their aquatic habitats. We characterized the physicochemical properties and bacterial microbiota of A. aegypti breeding sites and larvae on Guadeloupe and in French Guiana. In addition, we explored whether geographic location, the type of breeding site and physicochemical parameters influenced the microbiota associated with this mosquito species. We used large-scale 16S rRNA gene sequencing of 160 breeding sites and 147 pools of A. aegypti larvae and recorded 12 physicochemical parameters at the sampled breeding sites. Ordination plots and multiple linear regression were used to assess the influence of environmental factors on the bacterial microbiota of water and larvae. We found territory-specific differences in physicochemical properties (dissolved oxygen, conductivity) and the composition of bacterial communities in A. aegypti breeding sites that influenced the relative abundance of several bacteria genera (e.g., Methylobacterium, Roseoccocus) on the corresponding larvae. A significant fraction of the bacterial communities identified on larvae, dominated by Herbiconiux and Microvirga genera, were consistently enriched in mosquitoes regardless the location. In conclusion, territory-specific differences observed in the biotic and abiotic properties of A. aegypti breeding sites raise concern about the impact of these changes on pathogen transmission by different A. aegypti populations.
Collapse
Affiliation(s)
- Lyza Hery
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Amandine Guidez
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | | | - Christelle Delannay
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | | | - Yann Reynaud
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Jean Issaly
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | - Daniella Goindin
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Grégory Legrave
- Laboratory of Environment and Food Hygiene, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Joel Gustave
- Regional Health Agency of Guadeloupe, Gourbeyre, Guadeloupe France
| | - Stéphanie Raffestin
- Laboratory of Environment and Hygiene, Institut Pasteur of French Guiana, Cayenne, French Guiana France
| | - Sebastien Breurec
- Transmission, Reservoir and Diversity of Pathogens Unit, Institut Pasteur of Guadeloupe, Pointe-à-Pitre, France
- Hyacinthe Bastaraud Faculty of Medicine, University of Antilles, Pointe-à-Pitre, France
- INSERM Centre for Clinical Investigation 1424, Pointe-à-Pitre, Les Abymes France
| | - Philippe Constant
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec Canada
| | - Isabelle Dusfour
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | - Claude Guertin
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec Canada
| | - Anubis Vega-Rúa
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| |
Collapse
|
62
|
Girardot F, Allégra S, Pfendler S, Conord C, Rey C, Gillet B, Hughes S, Bouchardon AE, Hua A, Paran F, Bouchardon JL, Faure O. Bacterial diversity on an abandoned, industrial wasteland contaminated by polychlorinated biphenyls, dioxins, furans and trace metals. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 748:141242. [PMID: 32827890 DOI: 10.1016/j.scitotenv.2020.141242] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
Most former industrial sites are contaminated by mixtures of trace elements and organic pollutants. Levels of pollutants do not provide information regarding their biological impact, bioavailability and possible interactions between substances. There is genuine interest in combining chemical analyses with biological investigations. We studied a brownfield where several industrial activities were carried out starting in the 1970s, (incineration of pyralene transformers, recovery of copper by burning cables in the open air). Four representative plots showing different levels of polychlorobiphenyls were selected. Organic and trace metal levels were measured together with soil pedological characteristics. The bacterial community structure and functional diversity were assessed by 16S metagenomics with deep sequencing and community-level physiological profiling. Additionally, a vegetation survey was performed. Polychlorobiphenyls (8 mg.kg-1 to 1500 mg.kg-1) were from 2.4 × 103-fold to 6 × 105-fold higher than the European background level of 2.5 μg.kg-1. Polychlorinated dibenzo-p-dioxins and dibenzofurans ranged from 0.5 to 8.0 μg.kg-1. The soil was also contaminated with trace metals, i.e., Cu > 187, Zn > 217 and Pb > 372 mg.kg-1. Location within the study area, trace metal content and soil humidity were stronger determinants than organic pollutants of bacterial community structures and activities. Thus, the highest biological activity and the greatest bacteriological richness were observed in the plot that was less contaminated with trace metals, despite the high level of organic pollutants in the plot. Moreover, trace element pollution was associated with a relatively low presence of Actinobacteria and Rhizobia. The plot with the highest metal contamination was rich in metal-resistant bacteria such as Sphingomonadales, Geodermatophilaceae and KD4-96 (Chloroflexi phylum). Acidobacteria and Sphingomonadales, capable of resisting trace metals and degrading persistent organic pollutants, were dominant in the plots that had accumulated metal and organic contamination, but bacterial activity was lower in these plots than in the other plots.
Collapse
Affiliation(s)
- Françoise Girardot
- Université de Lyon, Université Jean Monnet Saint-Etienne, CNRS, EVS-ISTHME UMR 5600, F-42023 Saint-Étienne, France; Université de Lyon, Université Jean Monnet Saint-Etienne, Institut Universitaire de Technologie, F-42023 Saint- Étienne, France.
| | - Séverine Allégra
- Université de Lyon, Université Jean Monnet Saint-Etienne, CNRS, EVS-ISTHME UMR 5600, F-42023 Saint-Étienne, France; Université de Lyon, Université Jean Monnet Saint-Etienne, Institut Universitaire de Technologie, F-42023 Saint- Étienne, France
| | - Stéphane Pfendler
- Université de Lyon, Université Jean Monnet Saint-Etienne, CNRS, EVS-ISTHME UMR 5600, F-42023 Saint-Étienne, France
| | - Cyrille Conord
- Université de Lyon, Université Jean Monnet Saint-Etienne, CNRS, EVS-ISTHME UMR 5600, F-42023 Saint-Étienne, France
| | - Carine Rey
- Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon (ENSL), Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, F-69634 Lyon, France; Master de Biologie, ENSL, Université Claude Bernard Lyon I, Université de Lyon, F-69342 Lyon, France
| | - Benjamin Gillet
- Université de Lyon, Université Claude Bernard Lyon 1, ENSL, CNRS, Institut de Génomique Fonctionnelle de Lyon, UMR 5242, F-69007 Lyon, France
| | - Sandrine Hughes
- Université de Lyon, Université Claude Bernard Lyon 1, ENSL, CNRS, Institut de Génomique Fonctionnelle de Lyon, UMR 5242, F-69007 Lyon, France
| | - Anne Emilie Bouchardon
- Ecole Nationale Supérieure des Mines de Saint-Étienne (ENSM-SE), Centre SPIN-EVS, UMR5600, F-42023 Saint- Étienne tienne, France
| | - Anna Hua
- Université de Lyon, Université Jean Monnet Saint-Etienne, CNRS, EVS-ISTHME UMR 5600, F-42023 Saint-Étienne, France
| | - Frédéric Paran
- Ecole Nationale Supérieure des Mines de Saint-Étienne (ENSM-SE), Centre SPIN-EVS, UMR5600, F-42023 Saint- Étienne tienne, France
| | - Jean Luc Bouchardon
- Ecole Nationale Supérieure des Mines de Saint-Étienne (ENSM-SE), Centre SPIN-EVS, UMR5600, F-42023 Saint- Étienne tienne, France
| | - Olivier Faure
- Ecole Nationale Supérieure des Mines de Saint-Étienne (ENSM-SE), Centre SPIN-EVS, UMR5600, F-42023 Saint- Étienne tienne, France
| |
Collapse
|
63
|
Effect of gut microbiota on depressive-like behaviors in mice is mediated by the endocannabinoid system. Nat Commun 2020; 11:6363. [PMID: 33311466 PMCID: PMC7732982 DOI: 10.1038/s41467-020-19931-2] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
Depression is the leading cause of disability worldwide. Recent observations have revealed an association between mood disorders and alterations of the intestinal microbiota. Here, using unpredictable chronic mild stress (UCMS) as a mouse model of depression, we show that UCMS mice display phenotypic alterations, which could be transferred from UCMS donors to naïve recipient mice by fecal microbiota transplantation. The cellular and behavioral alterations observed in recipient mice were accompanied by a decrease in the endocannabinoid (eCB) signaling due to lower peripheral levels of fatty acid precursors of eCB ligands. The adverse effects of UCMS-transferred microbiota were alleviated by selectively enhancing the central eCB or by complementation with a strain of the Lactobacilli genus. Our findings provide a mechanistic scenario for how chronic stress, diet and gut microbiota generate a pathological feed-forward loop that contributes to despair behavior via the central eCB system.
Collapse
|
64
|
Bergsten E, Mestivier D, Sobhani I. The Limits and Avoidance of Biases in Metagenomic Analyses of Human Fecal Microbiota. Microorganisms 2020; 8:microorganisms8121954. [PMID: 33317070 PMCID: PMC7764459 DOI: 10.3390/microorganisms8121954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 01/02/2023] Open
Abstract
An increasing body of evidence highlights the role of fecal microbiota in various human diseases. However, more than two-thirds of fecal bacteria cannot be cultivated by routine laboratory techniques. Thus, physicians and scientists use DNA sequencing and statistical tools to identify associations between bacterial subgroup abundances and disease. However, discrepancies between studies weaken these results. In the present study, we focus on biases that might account for these discrepancies. First, three different DNA extraction methods (G’NOME, QIAGEN, and PROMEGA) were compared with regard to their efficiency, i.e., the quality and quantity of DNA recovered from feces of 10 healthy volunteers. Then, the impact of the DNA extraction method on the bacteria identification and quantification was evaluated using our published cohort of sample subjected to both 16S rRNA sequencing and whole metagenome sequencing (WMS). WMS taxonomical assignation employed the universal marker genes profiler mOTU-v2, which is considered the gold standard. The three standard pipelines for 16S RNA analysis (MALT and MEGAN6, QIIME1, and DADA2) were applied for comparison. Taken together, our results indicate that the G’NOME-based method was optimal in terms of quantity and quality of DNA extracts. 16S rRNA sequence-based identification of abundant bacteria genera showed acceptable congruence with WMS sequencing, with the DADA2 pipeline yielding the highest congruent levels. However, for low abundance genera (<0.5% of the total abundance) two pipelines and/or validation by quantitative polymerase chain reaction (qPCR) or WMS are required. Hence, 16S rRNA sequencing for bacteria identification and quantification in clinical and translational studies should be limited to diagnostic purposes in well-characterized and abundant genera. Additional techniques are warranted for low abundant genera, such as WMS, qPCR, or the use of two bio-informatics pipelines.
Collapse
Affiliation(s)
- Emma Bergsten
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
| | - Denis Mestivier
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
- Bioinformatics Core Facility, Institut Mondor de Recherche Biomédicale, UMR 955—Institut National de la Santé et de la Recherche Médicale—UPEC, 94010 Créteil, France
| | - Iradj Sobhani
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
- Service de Gastroenterologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, 94010 Créteil, France
- Correspondence: ; Tel.: +33-149814358
| |
Collapse
|
65
|
Zong X, Fu J, Xu B, Wang Y, Jin M. Interplay between gut microbiota and antimicrobial peptides. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:389-396. [PMID: 33364454 PMCID: PMC7750803 DOI: 10.1016/j.aninu.2020.09.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
The gut microbiota is comprised of a diverse array of microorganisms that interact with immune system and exert crucial roles for the health. Changes in the gut microbiota composition and functionality are associated with multiple diseases. As such, mobilizing a rapid and appropriate antimicrobial response depending on the nature of each stimulus is crucial for maintaining the balance between homeostasis and inflammation in the gut. Major players in this scenario are antimicrobial peptides (AMP), which belong to an ancient defense system found in all organisms and participate in a preservative co-evolution with a complex microbiome. Particularly increasing interactions between AMP and microbiota have been found in the gut. Here, we focus on the mechanisms by which AMP help to maintain a balanced microbiota and advancing our understanding of the circumstances of such balanced interactions between gut microbiota and host AMP. This review aims to provide a comprehensive overview on the interplay of diverse antimicrobial responses with enteric pathogens and the gut microbiota, which should have therapeutic implications for different intestinal disorders.
Collapse
Affiliation(s)
- Xin Zong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bocheng Xu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
66
|
Srivastava S, Sharma SK, Srivastava V, Kumar A. Proteomic Exploration of Listeria monocytogenes for the Purpose of Vaccine Designing Using a Reverse Vaccinology Approach. Int J Pept Res Ther 2020; 27:779-799. [PMID: 33144851 PMCID: PMC7595573 DOI: 10.1007/s10989-020-10128-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
Listeriosis is a major foodborne infection provoked by a bacterium known as Listeria monocytogenes. It is one of the predominant causes of death in pregnant women, infants, and immunocompromised persons. Despite such fatal effects, until now there is no proper medication or drug available for such a serious foodborne infection. One of the most promising ways to deal with this challenge is vaccination. This present study aims at the prediction of B cell epitopes for subunit vaccine designing against Listeria monocytogenes using a reverse vaccinology approach. Among screened out 299 epitopes of strain F2365 of Listeria monocytogenes, based on the VaxiJen score, the top 20 epitopes were selected. 3D modeling of epitopes and alleles was generated by PEPstrMOD and Swiss Model respectively. Molecular docking reveals 4 epitopes viz., MKFLFPLKL, CEETFGIRL, FLKIDPPIL, and VRHHGGGHK based on binding energy. All 4 epitopes were investigated for non-toxicity, binding affinity, and population coverage. After vigorous investigation, epitope FLKIDPPIL was anticipated as the best vaccine contender. The stability of the FLKIDPPIL-HLA DRB1 _0101 complex was proved by performing the simulation. Here, predicted peptide through the Insilico approach may become a potential remedy against listeriosis, after the wet-lab approach and clinical trials.
Collapse
Affiliation(s)
- Shivani Srivastava
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University Uttar Pradesh, Kanpur, 209217 India
| | - Suraj Kumar Sharma
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University Uttar Pradesh, Kanpur, 209217 India
| | - Vivek Srivastava
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University Uttar Pradesh, Kanpur, 209217 India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University Uttar Pradesh, Kanpur, 209217 India
| |
Collapse
|
67
|
Gómez-Laguna J, Cardoso-Toset F, Meza-Torres J, Pizarro-Cerdá J, Quereda JJ. Virulence potential of Listeria monocytogenes strains recovered from pigs in Spain. Vet Rec 2020; 187:e101. [PMID: 33024008 DOI: 10.1136/vr.105945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/04/2020] [Accepted: 08/16/2020] [Indexed: 11/03/2022]
Abstract
BACKGROUND Listeria monocytogenes is a foodborne bacterial pathogen that causes listeriosis, an infectious disease in animals and people, with pigs acting as asymptomatic reservoirs. In August 2019 an outbreak associated with the consumption of pork meat caused 222 human cases of listeriosis in Spain. Determining the diversity as well as the virulence potential of strains from pigs is important to public health. METHODS The behaviour of 23 L monocytogenes strains recovered from pig tonsils, meat and skin was compared by studying (1) internalin A, internalin B, listeriolysin O, actin assembly-inducing protein and PrfA expression levels, and (2) their invasion and intracellular growth in eukaryotic cells. RESULTS Marked differences were found in the expression of the selected virulence factors and the invasion and intracellular replication phenotypes of L monocytogenes strains. Strains obtained from meat samples and belonging to serotype 1/2a did not have internalin A anchored to the peptidoglycan. Some strains expressed higher levels of the studied virulence factors and invaded and replicated intracellularly more efficiently than an epidemic L monocytogenes reference strain (F2365). CONCLUSION This study demonstrates the presence of virulent L monocytogenes strains with virulent potential in pigs, with valuable implications in veterinary medicine and food safety.
Collapse
Affiliation(s)
- Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, University of Cordoba, Cordoba, Spain
| | | | - Jazmín Meza-Torres
- Yersinia Research Unit, Microbiology Department, Institut Pasteur, Paris, France
| | - Javier Pizarro-Cerdá
- Yersinia Research Unit, Microbiology Department, Institut Pasteur, Paris, France
- World Health Organization Collaborating Research & Reference Centre for Plague, Microbiology Department, Institut Pasteur, F-75724 Paris, France
| | - Juan J Quereda
- Departamento Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| |
Collapse
|
68
|
Neurotropic Lineage III Strains of Listeria monocytogenes Disseminate to the Brain without Reaching High Titer in the Blood. mSphere 2020; 5:5/5/e00871-20. [PMID: 32938704 PMCID: PMC7494839 DOI: 10.1128/msphere.00871-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Progress in understanding the two naturally occurring central nervous system (CNS) manifestations of listeriosis (meningitis/meningoencephalitis and rhombencephalitis) has been limited by the lack of small animal models that can readily distinguish between these distinct infections. We report here that certain neurotropic strains of Listeria monocytogenes can spread to the brains of young otherwise healthy mice and cause neurological deficits without causing a fatal bacteremia. The novel strains described here fall within phylogenetic lineage III, a small collection of L. monocytogenes isolates that have not been well characterized to date. The animal model reported here mimics many features of human rhombencephalitis and will be useful for studying the mechanisms that allow L. monocytogenes to disseminate to the brain stem following natural foodborne transmission. Listeria monocytogenes is thought to colonize the brain using one of three mechanisms: direct invasion of the blood-brain barrier, transportation across the barrier by infected monocytes, and axonal migration to the brain stem. The first two pathways seem to occur following unrestricted bacterial growth in the blood and thus have been linked to immunocompromise. In contrast, cell-to-cell spread within nerves is thought to be mediated by a particular subset of neurotropic L. monocytogenes strains. In this study, we used a mouse model of foodborne transmission to evaluate the neurotropism of several L. monocytogenes isolates. Two strains preferentially colonized the brain stems of BALB/cByJ mice 5 days postinfection and were not detectable in blood at that time point. In contrast, infection with other strains resulted in robust systemic infection of the viscera but no dissemination to the brain. Both neurotropic strains (L2010-2198, a human rhombencephalitis isolate, and UKVDL9, a sheep brain isolate) typed as phylogenetic lineage III, the least characterized group of L. monocytogenes. Neither of these strains encodes InlF, an internalin-like protein that was recently shown to promote invasion of the blood-brain barrier. Acute neurologic deficits were observed in mice infected with the neurotropic strains, and milder symptoms persisted for up to 16 days in some animals. These results demonstrate that neurotropic L. monocytogenes strains are not restricted to any one particular lineage and suggest that the foodborne mouse model of listeriosis can be used to investigate the pathogenic mechanisms that allow L. monocytogenes to invade the brain stem. IMPORTANCE Progress in understanding the two naturally occurring central nervous system (CNS) manifestations of listeriosis (meningitis/meningoencephalitis and rhombencephalitis) has been limited by the lack of small animal models that can readily distinguish between these distinct infections. We report here that certain neurotropic strains of Listeria monocytogenes can spread to the brains of young otherwise healthy mice and cause neurological deficits without causing a fatal bacteremia. The novel strains described here fall within phylogenetic lineage III, a small collection of L. monocytogenes isolates that have not been well characterized to date. The animal model reported here mimics many features of human rhombencephalitis and will be useful for studying the mechanisms that allow L. monocytogenes to disseminate to the brain stem following natural foodborne transmission.
Collapse
|
69
|
Lourenço M, Chaffringeon L, Lamy-Besnier Q, Pédron T, Campagne P, Eberl C, Bérard M, Stecher B, Debarbieux L, De Sordi L. The Spatial Heterogeneity of the Gut Limits Predation and Fosters Coexistence of Bacteria and Bacteriophages. Cell Host Microbe 2020; 28:390-401.e5. [DOI: 10.1016/j.chom.2020.06.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/30/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023]
|
70
|
Differential Modulation of Listeria monocytogenes Fitness, In Vitro Virulence, and Transcription of Virulence-Associated Genes in Response to the Presence of Different Microorganisms. Appl Environ Microbiol 2020; 86:AEM.01165-20. [PMID: 32591377 DOI: 10.1128/aem.01165-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/20/2020] [Indexed: 02/03/2023] Open
Abstract
Interactions between Listeria monocytogenes and food-associated or environmental bacteria are critical not only for the growth but also for a number of key biological processes of the microorganism. In this regard, limited information exists on the impact of other microorganisms on the virulence of L. monocytogenes In this study, the growth of L. monocytogenes was evaluated in a single culture or in coculture with L. innocua, Bacillus subtilis, Lactobacillus plantarum, or Pseudomonas aeruginosa in tryptic soy broth (10°C/10 days and 37°C/24 h). Transcriptional levels of 9 key virulence genes (inlA, inlB, inlC, inlJ, sigB, prfA, hly, plcA, and plcB) and invasion efficiency and intracellular growth in Caco-2 cells were determined for L. monocytogenes following growth in mono- or coculture for 3 days at 10°C or 9 h at 37°C. The growth of L. monocytogenes was negatively affected by the presence of L. innocua and B. subtilis, while the effect of cell-to-cell contact on L. monocytogenes growth was dependent on the competing microorganism. Cocultivation affected the in vitro virulence properties of L. monocytogenes in a microorganism-specific manner, with L. innocua mainly enhancing and B. subtilis reducing the invasion of the pathogen in Caco-2 cells. Assessment of the mRNA levels of L. monocytogenes virulence genes in the presence of the four tested bacteria revealed a complex pattern in which the observed up- or downregulation was only partially correlated with growth or in vitro virulence and mainly suggested that L. monocytogenes may display a microorganism-specific transcriptional response.IMPORTANCE Listeria monocytogenes is the etiological agent of the severe foodborne disease listeriosis. Important insight regarding the physiology and the infection biology of this microorganism has been acquired in the past 20 years. However, despite the fact that L. monocytogenes coexists with various microorganisms throughout its life cycle and during transmission from the environment to foods and then to the host, there is still limited knowledge related to the impact of surrounding microorganisms on L. monocytogenes' biological functions. In this study, we showed that L. monocytogenes modulates specific biological activities (i.e., growth and virulence potential) as a response to coexisting microorganisms and differentially alters the expression of virulence-associated genes when confronted with different bacterial genera and species. Our work suggests that the interaction with different bacteria plays a key role in the survival strategies of L. monocytogenes and supports the need to incorporate biotic factors into the research conducted to identify mechanisms deployed by this organism for establishment in different environments.
Collapse
|
71
|
Uengwetwanit T, Uawisetwathana U, Arayamethakorn S, Khudet J, Chaiyapechara S, Karoonuthaisiri N, Rungrassamee W. Multi-omics analysis to examine microbiota, host gene expression and metabolites in the intestine of black tiger shrimp ( Penaeus monodon) with different growth performance. PeerJ 2020; 8:e9646. [PMID: 32864208 PMCID: PMC7430268 DOI: 10.7717/peerj.9646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/12/2020] [Indexed: 12/20/2022] Open
Abstract
Understanding the correlation between shrimp growth and their intestinal bacteria would be necessary to optimize animal's growth performance. Here, we compared the bacterial profiles along with the shrimp's gene expression responses and metabolites in the intestines between the Top and the Bottom weight groups. Black tiger shrimp (Penaeus monodon) were collected from the same population and rearing environments. The two weight groups, the Top-weight group with an average weight of 36.82 ± 0.41 g and the Bottom-weight group with an average weight of 17.80 ± 11.81 g, were selected. Intestines were aseptically collected and subjected to microbiota, transcriptomic and metabolomic profile analyses. The weighted-principal coordinates analysis (PCoA) based on UniFrac distances showed similar bacterial profiles between the two groups, suggesting similar relative composition of the overall bacterial community structures. This observed similarity was likely due to the fact that shrimp were from the same genetic background and reared under the same habitat and diets. On the other hand, the unweighted-distance matrix revealed that the bacterial profiles associated in intestines of the Top-weight group were clustered distinctly from those of the Bottom-weight shrimp, suggesting that some unique non-dominant bacterial genera were found associated with either group. The key bacterial members associated to the Top-weight shrimp were mostly from Firmicutes (Brevibacillus and Fusibacter) and Bacteroidetes (Spongiimonas), both of which were found in significantly higher abundance than those of the Bottom-weight shrimp. Transcriptomic profile of shrimp intestines found significant upregulation of genes mostly involved in nutrient metabolisms and energy storage in the Top-weight shrimp. In addition to significantly expressed metabolic-related genes, the Bottom-weight shrimp also showed significant upregulation of stress and immune-related genes, suggesting that these pathways might contribute to different degrees of shrimp growth performance. A non-targeted metabolome analysis from shrimp intestines revealed different metabolic responsive patterns, in which the Top-weight shrimp contained significantly higher levels of short chain fatty acids, lipids and organic compounds than the Bottom-weight shrimp. The identified metabolites included those that were known to be produced by intestinal bacteria such as butyric acid, 4-indolecarbaldehyde and L-3-phenyllactic acid as well as those produced by shrimp such as acyl-carnitines and lysophosphatidylcholine. The functions of these metabolites were related to nutrient absorption and metabolisms. Our findings provide the first report utilizing multi-omics integration approach to investigate microbiota, metabolic and transcriptomics profiles of the host shrimp and their potential roles and relationship to shrimp growth performance.
Collapse
Affiliation(s)
- Tanaporn Uengwetwanit
- Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Umaporn Uawisetwathana
- Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Sopacha Arayamethakorn
- Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Juthatip Khudet
- Shrimp Genetic Improvement Center, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Sage Chaiyapechara
- Aquaculture Service Development Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Nitsara Karoonuthaisiri
- Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Wanilada Rungrassamee
- Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| |
Collapse
|
72
|
Volant S, Lechat P, Woringer P, Motreff L, Campagne P, Malabat C, Kennedy S, Ghozlane A. SHAMAN: a user-friendly website for metataxonomic analysis from raw reads to statistical analysis. BMC Bioinformatics 2020; 21:345. [PMID: 32778056 PMCID: PMC7430814 DOI: 10.1186/s12859-020-03666-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/16/2020] [Indexed: 01/04/2023] Open
Abstract
Background Comparing the composition of microbial communities among groups of interest (e.g., patients vs healthy individuals) is a central aspect in microbiome research. It typically involves sequencing, data processing, statistical analysis and graphical display. Such an analysis is normally obtained by using a set of different applications that require specific expertise for installation, data processing and in some cases, programming skills. Results Here, we present SHAMAN, an interactive web application we developed in order to facilitate the use of (i) a bioinformatic workflow for metataxonomic analysis, (ii) a reliable statistical modelling and (iii) to provide the largest panel of interactive visualizations among the applications that are currently available. SHAMAN is specifically designed for non-expert users. A strong benefit is to use an integrated version of the different analytic steps underlying a proper metagenomic analysis. The application is freely accessible at http://shaman.pasteur.fr/, and may also work as a standalone application with a Docker container (aghozlane/shaman), conda and R. The source code is written in R and is available at https://github.com/aghozlane/shaman. Using two different datasets (a mock community sequencing and a published 16S rRNA metagenomic data), we illustrate the strengths of SHAMAN in quickly performing a complete metataxonomic analysis. Conclusions With SHAMAN, we aim at providing the scientific community with a platform that simplifies reproducible quantitative analysis of metagenomic data.
Collapse
Affiliation(s)
- Stevenn Volant
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France
| | - Pierre Lechat
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France
| | - Perrine Woringer
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France
| | - Laurence Motreff
- Biomics - Département Génomes et Génétique, Institut Pasteur, 28 Rue du Docteur Roux, Paris, 75015, France
| | - Pascal Campagne
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France
| | - Christophe Malabat
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France
| | - Sean Kennedy
- Biomics - Département Génomes et Génétique, Institut Pasteur, 28 Rue du Docteur Roux, Paris, 75015, France
| | - Amine Ghozlane
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 Rue Du Docteur Roux, Paris, 75015, France. .,Biomics - Département Génomes et Génétique, Institut Pasteur, 28 Rue du Docteur Roux, Paris, 75015, France.
| |
Collapse
|
73
|
Joerger RD. Salmonella enterica's "Choice": Itaconic Acid Degradation or Bacteriocin Immunity Genes. Genes (Basel) 2020; 11:genes11070797. [PMID: 32679707 PMCID: PMC7397319 DOI: 10.3390/genes11070797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 11/30/2022] Open
Abstract
Itaconic acid is an immunoregulatory metabolite produced by macrophages in response to pathogen invasion. It also exhibits antibacterial activity because it is an uncompetitive inhibitor of isocitrate lyase, whose activity is required for the glyoxylate shunt to be operational. Some bacteria, such as Yersinia pestis, encode enzymes that can degrade itaconic acid and therefore eliminate this metabolic inhibitor. Studies, primarily with Salmonella enterica subspecies enterica serovar Typhimurium, have demonstrated the presence of similar genes in this pathogen and the importance of these genes for the persistence of the pathogen in murine hosts. This minireview demonstrates that, based on Blast searches of 1063 complete Salmonella genome sequences, not all Salmonella serovars possess these genes. It is also shown that the growth of Salmonella isolates that do not possess these genes is sensitive to the acid under glucose-limiting conditions. Interestingly, most of the serovars without the three genes, including serovar Typhi, harbor DNA at the corresponding genomic location that encodes two open reading frames that are similar to bacteriocin immunity genes. It is hypothesized that these genes could be important for Salmonella that finds itself in strong competition with other Enterobacteriacea in the intestinal tract—for example, during inflammation.
Collapse
Affiliation(s)
- Rolf D Joerger
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
74
|
NASQAR: a web-based platform for high-throughput sequencing data analysis and visualization. BMC Bioinformatics 2020; 21:267. [PMID: 32600310 PMCID: PMC7322916 DOI: 10.1186/s12859-020-03577-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 06/01/2020] [Indexed: 01/23/2023] Open
Abstract
Background As high-throughput sequencing applications continue to evolve, the rapid growth in quantity and variety of sequence-based data calls for the development of new software libraries and tools for data analysis and visualization. Often, effective use of these tools requires computational skills beyond those of many researchers. To ease this computational barrier, we have created a dynamic web-based platform, NASQAR (Nucleic Acid SeQuence Analysis Resource). Results NASQAR offers a collection of custom and publicly available open-source web applications that make extensive use of a variety of R packages to provide interactive data analysis and visualization. The platform is publicly accessible at http://nasqar.abudhabi.nyu.edu/. Open-source code is on GitHub at https://github.com/nasqar/NASQAR, and the system is also available as a Docker image at https://hub.docker.com/r/aymanm/nasqarall. NASQAR is a collaboration between the core bioinformatics teams of the NYU Abu Dhabi and NYU New York Centers for Genomics and Systems Biology. Conclusions NASQAR empowers non-programming experts with a versatile and intuitive toolbox to easily and efficiently explore, analyze, and visualize their Transcriptomics data interactively. Popular tools for a variety of applications are currently available, including Transcriptome Data Preprocessing, RNA-seq Analysis (including Single-cell RNA-seq), Metagenomics, and Gene Enrichment.
Collapse
|
75
|
Gonçalves P, El Daker S, Vasseur F, Serafini N, Lim A, Azogui O, Decaluwe H, Guy-Grand D, Freitas AA, Di Santo JP, Rocha B. Microbiota stimulation generates LCMV-specific memory CD8 + T cells in SPF mice and determines their TCR repertoire during LCMV infection. Mol Immunol 2020; 124:125-141. [PMID: 32563081 DOI: 10.1016/j.molimm.2020.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/16/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
Abstract
Both mouse and human harbour memory phenotype CD8+ T cells specific for antigens in hosts that have not been previously exposed to these antigens. The origin and the nature of the stimuli responsible for generation of CD44hi CD8+ T cells in specific pathogen-free (SPF) mice remain controversial. It is known that microbiota plays a crucial role in the prevention and resolution of systemic infections by influencing myelopoiesis, regulating dendritic cells, inflammasome activation and promoting the production of type I and II interferons. By contrast, here we suggest that microbiota has a direct effect on generation of memory phenotype CD44hiGP33+CD8+ T cells. In SPF mice, it generates a novel GP33+CD44hiCD8+ T cell sub-population associating the properties of innate and genuine memory cells. These cells are highly enriched in the bone marrow, proliferate rapidly and express immediate effector functions. They dominate the response to LCMV and express particular TCRβ chains. The sequence of these selected TCRβ chains overlaps with that of GP33+CD8+ T cells directly selected by microbiota in the gut epithelium of SPF mice, demonstrating a common selection mechanism in gut and peripheral CD8+ T cell pool. Therefore microbiota has a direct role in priming T cell immunity in SPF mice and in the selection of TCRβ repertoires during systemic infection. We identify a mechanism that primes T cell immunity in SPF mice and may have a major role in colonization resistance and protection from infection.
Collapse
Affiliation(s)
- Pedro Gonçalves
- Population Biology Unit, CNRS URA 196, Institut Pasteur, Paris 75015, France; INSERM, U1151, CNRS, UMR8253, Institut Necker Enfants Malades, Université Paris Descartes, Paris 75015, France; Innate Immunity Unit, INSERM, U668, Institut Pasteur, Paris 75015, France.
| | - Sary El Daker
- Population Biology Unit, CNRS URA 196, Institut Pasteur, Paris 75015, France
| | - Florence Vasseur
- INSERM, U1151, CNRS, UMR8253, Institut Necker Enfants Malades, Université Paris Descartes, Paris 75015, France
| | - Nicolas Serafini
- Innate Immunity Unit, INSERM, U668, Institut Pasteur, Paris 75015, France; INSERM U1223, Paris 75015, France
| | | | - Orly Azogui
- INSERM, U1151, CNRS, UMR8253, Institut Necker Enfants Malades, Université Paris Descartes, Paris 75015, France
| | - Helene Decaluwe
- Population Biology Unit, CNRS URA 196, Institut Pasteur, Paris 75015, France
| | - Delphine Guy-Grand
- INSERM U1223, Paris 75015, France; Lymphopoiesis Unit, INSERM U668, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris 75015, France
| | - Antonio A Freitas
- Population Biology Unit, CNRS URA 196, Institut Pasteur, Paris 75015, France
| | - James P Di Santo
- Innate Immunity Unit, INSERM, U668, Institut Pasteur, Paris 75015, France; INSERM U1223, Paris 75015, France
| | - Benedita Rocha
- Population Biology Unit, CNRS URA 196, Institut Pasteur, Paris 75015, France; INSERM, U1151, CNRS, UMR8253, Institut Necker Enfants Malades, Université Paris Descartes, Paris 75015, France.
| |
Collapse
|
76
|
Cartography of Free-Living Amoebae in Soil in Guadeloupe (French West Indies) Using DNA Metabarcoding. Pathogens 2020; 9:pathogens9060440. [PMID: 32512696 PMCID: PMC7350318 DOI: 10.3390/pathogens9060440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022] Open
Abstract
Free-living amoebae (FLA) are ubiquitous protists. Pathogenic FLA such as N. fowleri can be found in hot springs in Guadeloupe, soil being the origin of this contamination. Herein, we analyzed the diversity and distribution of FLA in soil using a targeted metataxonomic analysis. Soil samples (n = 107) were collected from 40 sites. DNA was extracted directly from soil samples or from FLA cultivated at different temperatures (30, 37 and 44 °C). Metabarcoding studies were then conducted through FLA 18SrDNA amplicons sequencing; amplicon sequence variants (ASV) were extracted from each sample and taxonomy assigned against SILVA database using QIIME2 and SHAMAN pipelines. Vermamoeba were detected in DNA extracted directly from the soil, but to detect other FLA an amoebal enrichment step was necessary. V. vermiformis was by far the most represented species of FLA, being detected throughout the islands. Although Naegleria were mainly found in Basse-Terre region, N. fowleri was also detected in Grand Terre and Les Saintes Islands. Acanthamoeba were mainly found in areas where temperature is approx. 30 °C. Vannella and Vahlkampfia were randomly found in Guadeloupe islands. FLA detected in Guadeloupe include both pathogenic genera and genera that can putatively harbor microbial pathogens, therefore posing a potential threat to human health.
Collapse
|
77
|
Pathogenic Biohacking: Induction, Modulation and Subversion of Host Transcriptional Responses by Listeria monocytogenes. Toxins (Basel) 2020; 12:toxins12050294. [PMID: 32380645 PMCID: PMC7290974 DOI: 10.3390/toxins12050294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
During infection, the foodborne bacterial pathogen Listeria monocytogenes dynamically influences the gene expression profile of host cells. Infection-induced transcriptional changes are a typical feature of the host-response to bacteria and contribute to the activation of protective genes such as inflammatory cytokines. However, by using specialized virulence factors, bacterial pathogens can target signaling pathways, transcription factors, and epigenetic mechanisms to alter host gene expression, thereby reprogramming the response to infection. Therefore, the transcriptional profile that is established in the host is delicately balanced between antibacterial responses and pathogenesis, where any change in host gene expression might significantly influence the outcome of infection. In this review, we discuss the known transcriptional and epigenetic processes that are engaged during Listeria monocytogenes infection, the virulence factors that can remodel them, and the impact these processes have on the outcome of infection.
Collapse
|
78
|
Angebault C, Payen M, Woerther PL, Rodriguez C, Botterel F. Combined bacterial and fungal targeted amplicon sequencing of respiratory samples: Does the DNA extraction method matter? PLoS One 2020; 15:e0232215. [PMID: 32343737 PMCID: PMC7188255 DOI: 10.1371/journal.pone.0232215] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
Background High-throughput sequencing techniques are used to analyse the diversity of the respiratory microbiota in health and disease. Although extensive data are available regarding bacterial respiratory microbiota, its fungal component remains poorly studied. This is partly due to the technical issues associated with fungal metagenomics analyses. In this study, we compared two DNA extraction protocols and two fungal amplification targets for combined bacterial and fungal targeted amplicon sequencing analyses of the respiratory microbiota. Methods Six sputa, randomly selected from routine samples in Mondor Hospital (Creteil, France) and treated anonymously, were tested after bacterial and fungal routine culture. Two of which were spiked with Aspergillus Fumigati and Aspergillus Nigri (105 conidia/mL). After mechanical lysis, DNA was extracted using automated QIAsymphony® extraction (AQE) or manual PowerSoil® MoBio extraction (MPE). DNA yield and purity were compared. DNA extracted from spiked sputa was subjected to (i) real-time PCR for Aspergillus DNA detection and (ii) combined metagenomic analyses targeting barcoded primers for fungal ITS1 and ITS2, and bacterial V1-V2 and V3-V4 16S regions. Amplicon libraries were prepared using MiSeq Reagent V3 kit on Illumina platform. Data were analysed using PyroMIC© and SHAMAN software, and compared with culture results. Results AQE extraction provided a higher yield of DNA (AQE/MPE DNA ratio = 4.5 [1.3–11]) in a shorter time. The yield of Aspergillus DNA detected by qPCR was similar for spiked sputa regardless of extraction protocol. The extraction moderately impacted the diversity or relative abundances of bacterial communities using targeted amplicon sequencing (2/43 taxa impacted). For fungi, the relative abundances of 4/11 major taxa were impacted and AQE results were closer to culture results. The V1-V2 or V3-V4 and ITS1 or ITS2 targets assessed similarly the diversity of bacterial and fungal major taxa, but ITS2 and V3-V4 detected more minor taxa. Conclusion Our results showed the importance of DNA extraction for combined bacterial and fungal targeted metagenomics of respiratory samples. The extraction protocol can affect DNA yield and the relative abundances of few bacterial but more fungal taxa. For fungal analysis, ITS2 allowed the detection of a greater number of minor taxa compared with ITS1.
Collapse
Affiliation(s)
- Cécile Angebault
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, Assistance Publique des Hôpitaux de Paris (APHP), Créteil, France
- EA DYNAMiC 7380, Faculté de Santé, Univ Paris-Est Créteil, Créteil, France
- EA DYNAMiC 7380, Ecole nationale vétérinaire d’Alfort, USC Anses, Maison-Alfort, France
- * E-mail:
| | - Mathilde Payen
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, Assistance Publique des Hôpitaux de Paris (APHP), Créteil, France
| | - Paul-Louis Woerther
- EA DYNAMiC 7380, Faculté de Santé, Univ Paris-Est Créteil, Créteil, France
- Unité de Bactériologie-Hygiène, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, APHP, Créteil, France
| | - Christophe Rodriguez
- Next-Generation Sequencing Platform”Génomiques”, INSERM U955, APHP, IMRB Créteil, Créteil, France
| | - Françoise Botterel
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, Assistance Publique des Hôpitaux de Paris (APHP), Créteil, France
- EA DYNAMiC 7380, Faculté de Santé, Univ Paris-Est Créteil, Créteil, France
- EA DYNAMiC 7380, Ecole nationale vétérinaire d’Alfort, USC Anses, Maison-Alfort, France
| |
Collapse
|
79
|
Chen Y, Chen M, Wang J, Wu Q, Cheng J, Zhang J, Sun Q, Xue L, Zeng H, Lei T, Pang R, Ye Q, Wu S, Zhang S, Wu H, Li W, Kou X. Heterogeneity, Characteristics, and Public Health Implications of Listeria monocytogenes in Ready-to-Eat Foods and Pasteurized Milk in China. Front Microbiol 2020; 11:642. [PMID: 32351479 PMCID: PMC7174501 DOI: 10.3389/fmicb.2020.00642] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 03/20/2020] [Indexed: 12/20/2022] Open
Abstract
Listeria monocytogenes is a foodborne pathogen with a high mortality rate in humans. This study aimed to identify the pathogenic potential of L. monocytogenes isolated from ready-to-eat (RTE) foods and pasteurized milk in China on the basis of its phenotypic and genotypic characteristics. Approximately 7.7% (44/570) samples tested positive for L. monocytogenes among 10.8% (39/360) RTE and 2.4% (5/210) pasteurized milk samples, of which 77.3% (34/44) had < 10 MPN/g, 18.2% (8/44) had 10-110 MPN/g, and 4.5% (2/44) had > 110 MPN/g. A total of 48 strains (43 from RTE foods and five from milk samples) of L. monocytogenes were isolated from 44 positive samples. PCR-serogroup analysis revealed that the most prevalent serogroup was II.2 (1/2b-3b-7), accounting for 52.1% (25/48) of the total, followed by serogroup I.1 (1/2a-3a) accounting for 33.3% (16/48), serogroup I.2 (1/2c-3c) accounting for 12.5% (6/48), and serogroup II.1 (4b-4d-4e) accounting for 2.1%. All isolates were grouped into 11 sequence types (STs) belonging to 10 clonal complexes (CCs) and one singleton (ST619) via multi-locus sequence typing. The most prevalent ST was ST87 (29.2%), followed by ST8 (22.9%), and ST9 (12.5%). Virulence genes determination showed that all isolates harbored eight virulence genes belonging to Listeria pathogenicity islands 1 (LIPI-1) (prfA, actA, hly, mpl, plcA, plcB, and iap) and inlB. Approximately 85.4% isolates carried full-length inlA, whereas seven isolates had premature stop codons in inlA, six of which belonged to ST9 and one to ST5. Furthermore, LLS (encoded by llsX gene, representing LIPI-3) displays bactericidal activity and modifies the host microbiota during infection. LIPI-4 enhances neural and placental tropisms of L. monocytogenes. Results showed that six (12.5%) isolates harbored the llsX gene, and they belonged to ST1/CC1, ST3/CC3, and ST619. Approximately 31.3% (15/48) isolates (belonging to ST87/CC87 and ST619) harbored ptsA (representing LIPI-4), indicating the potential risk of this pathogen. Antimicrobial susceptibility tests revealed that > 95% isolates were susceptible to 16 antimicrobials; however, 60.4 and 22.9% isolates were intermediately resistant to streptomycin and ciprofloxacin, respectively. The results show that several isolates harbor LIPI-3 and LIPI-4 genes, which may be a possible transmission route for Listeria infections in consumers.
Collapse
Affiliation(s)
- Yuetao Chen
- College of Food Science, South China Agricultural University, Guangzhou, China
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Moutong Chen
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Qingping Wu
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Jianheng Cheng
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Jumei Zhang
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Qifan Sun
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Liang Xue
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Haiyan Zeng
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Tao Lei
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Rui Pang
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Qinghua Ye
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Shi Wu
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Shuhong Zhang
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Haoming Wu
- Guangdong Institute of Microbiology, Guangdong Academic of Science, State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou, China
| | - Wenzhi Li
- Infinitus (China) Company, Ltd., Guangzhou, China
| | - Xiuying Kou
- Infinitus (China) Company, Ltd., Guangzhou, China
| |
Collapse
|
80
|
Jabs S, Biton A, Bécavin C, Nahori MA, Ghozlane A, Pagliuso A, Spanò G, Guérineau V, Touboul D, Giai Gianetto Q, Chaze T, Matondo M, Dillies MA, Cossart P. Impact of the gut microbiota on the m 6A epitranscriptome of mouse cecum and liver. Nat Commun 2020; 11:1344. [PMID: 32165618 PMCID: PMC7067863 DOI: 10.1038/s41467-020-15126-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/17/2020] [Indexed: 12/28/2022] Open
Abstract
The intestinal microbiota modulates host physiology and gene expression via mechanisms that are not fully understood. Here we examine whether host epitranscriptomic marks are affected by the gut microbiota. We use methylated RNA-immunoprecipitation and sequencing (MeRIP-seq) to identify N6-methyladenosine (m6A) modifications in mRNA of mice carrying conventional, modified, or no microbiota. We find that variations in the gut microbiota correlate with m6A modifications in the cecum, and to a lesser extent in the liver, affecting pathways related to metabolism, inflammation and antimicrobial responses. We analyze expression levels of several known writer and eraser enzymes, and find that the methyltransferase Mettl16 is downregulated in absence of a microbiota, and one of its target mRNAs, encoding S-adenosylmethionine synthase Mat2a, is less methylated. We furthermore show that Akkermansia muciniphila and Lactobacillus plantarum affect specific m6A modifications in mono-associated mice. Our results highlight epitranscriptomic modifications as an additional level of interaction between commensal bacteria and their host.
Collapse
Affiliation(s)
- Sabrina Jabs
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, U604 Institut National de la Santé et de la Recherche Médicale, USC 2020 Institut National de la Recherche Agronomique, 25 rue du Dr Roux, F-75015, Paris, France.
| | - Anne Biton
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 rue du Dr Roux, F-75015, Paris, France
| | - Christophe Bécavin
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 rue du Dr Roux, F-75015, Paris, France
| | - Marie-Anne Nahori
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, U604 Institut National de la Santé et de la Recherche Médicale, USC 2020 Institut National de la Recherche Agronomique, 25 rue du Dr Roux, F-75015, Paris, France
| | - Amine Ghozlane
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 rue du Dr Roux, F-75015, Paris, France
| | - Alessandro Pagliuso
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, U604 Institut National de la Santé et de la Recherche Médicale, USC 2020 Institut National de la Recherche Agronomique, 25 rue du Dr Roux, F-75015, Paris, France
| | - Giulia Spanò
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, U604 Institut National de la Santé et de la Recherche Médicale, USC 2020 Institut National de la Recherche Agronomique, 25 rue du Dr Roux, F-75015, Paris, France
| | - Vincent Guérineau
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
| | - David Touboul
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
| | - Quentin Giai Gianetto
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 rue du Dr Roux, F-75015, Paris, France
- Unité de spectrométrie de masse et Protéomique, CNRS USR 2000, Institut Pasteur, 28 rue du Dr Roux, F-75015, Paris, France
| | - Thibault Chaze
- Unité de spectrométrie de masse et Protéomique, CNRS USR 2000, Institut Pasteur, 28 rue du Dr Roux, F-75015, Paris, France
| | - Mariette Matondo
- Unité de spectrométrie de masse et Protéomique, CNRS USR 2000, Institut Pasteur, 28 rue du Dr Roux, F-75015, Paris, France
| | - Marie-Agnès Dillies
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 28 rue du Dr Roux, F-75015, Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, U604 Institut National de la Santé et de la Recherche Médicale, USC 2020 Institut National de la Recherche Agronomique, 25 rue du Dr Roux, F-75015, Paris, France.
| |
Collapse
|
81
|
Rolhion N, Chassaing B, Nahori MA, de Bodt J, Moura A, Lecuit M, Dussurget O, Bérard M, Marzorati M, Fehlner-Peach H, Littman DR, Gewirtz AT, Van de Wiele T, Cossart P. A Listeria monocytogenes Bacteriocin Can Target the Commensal Prevotella copri and Modulate Intestinal Infection. Cell Host Microbe 2020; 26:691-701.e5. [PMID: 31726031 PMCID: PMC6854461 DOI: 10.1016/j.chom.2019.10.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/05/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Understanding the role of the microbiota components in either preventing or favoring enteric infections is critical. Here, we report the discovery of a Listeria bacteriocin, Lmo2776, which limits Listeria intestinal colonization. Oral infection of conventional mice with a Δlmo2776 mutant leads to a thinner intestinal mucus layer and higher Listeria loads both in the intestinal content and deeper tissues compared to WT Listeria. This latter difference is microbiota dependent, as it is not observed in germ-free mice. Strikingly, it is phenocopied by pre-colonization of germ-free mice before Listeria infection with Prevotella copri, an abundant gut-commensal bacteria, but not with the other commensals tested. We further show that Lmo2776 targets P. copri and reduces its abundance. Together, these data unveil a role for P.copri in exacerbating intestinal infection, highlighting that pathogens such as Listeria may selectively deplete microbiota bacterial species to avoid excessive inflammation. L. monocytogenes secretes a bacteriocin (Lmo2776) homologous to the lactococcin 972 Lmo2776 controls Listeria intestinal colonization in a microbiota-dependent manner Lmo2776 targets the abundant gut commensal Prevotella copri Presence of P. copri exacerbates infection
Collapse
Affiliation(s)
- Nathalie Rolhion
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France
| | - Benoit Chassaing
- Neurosciences Institute, Georgia State University (GSU), Atlanta, GA 30303, USA; Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, GSU, Atlanta, GA 30303, USA
| | - Marie-Anne Nahori
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France
| | - Jana de Bodt
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Alexandra Moura
- Institut Pasteur, Unité Biologie des Infections, 75015 Paris, France; Inserm, U1117, 75015 Paris, France
| | - Marc Lecuit
- Institut Pasteur, Unité Biologie des Infections, 75015 Paris, France; Inserm, U1117, 75015 Paris, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Institut Imagine, 75743 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France; Université de Paris, 75013 Paris, France
| | - Marion Bérard
- Animalerie Centrale, Department of Technology and Scientific Programmes, Institut Pasteur, 75015 Paris, France
| | - Massimo Marzorati
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Hannah Fehlner-Peach
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, GSU, Atlanta, GA 30303, USA
| | - Tom Van de Wiele
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France.
| |
Collapse
|
82
|
Mitri C, Bischoff E, Belda Cuesta E, Volant S, Ghozlane A, Eiglmeier K, Holm I, Dieme C, Brito-Fravallo E, Guelbeogo WM, Sagnon N, Riehle MM, Vernick KD. Leucine-Rich Immune Factor APL1 Is Associated With Specific Modulation of Enteric Microbiome Taxa in the Asian Malaria Mosquito Anopheles stephensi. Front Microbiol 2020; 11:306. [PMID: 32174902 PMCID: PMC7054466 DOI: 10.3389/fmicb.2020.00306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/11/2020] [Indexed: 01/14/2023] Open
Abstract
The commensal gut microbiome is contained by the enteric epithelial barrier, but little is known about the degree of specificity of host immune barrier interactions for particular bacterial taxa. Here, we show that depletion of leucine-rich repeat immune factor APL1 in the Asian malaria mosquito Anopheles stephensi is associated with higher midgut abundance of just the family Enterobacteraceae, and not generalized dysbiosis of the microbiome. The effect is explained by the response of a narrow clade containing two main taxa related to Klebsiella and Cedecea. Analysis of field samples indicate that these two taxa are recurrent members of the wild Anopheles microbiome. Triangulation using sequence and functional data incriminated relatives of C. neteri and Cedecea NFIX57 as candidates for the Cedecea component, and K. michiganensis, K. oxytoca, and K.sp. LTGPAF-6F as candidates for the Klebsiella component. APL1 presence is associated with host ability to specifically constrain the abundance of a narrow microbiome clade of the Enterobacteraceae, and the immune factor may promote homeostasis of this clade in the enteric microbiome for host benefit.
Collapse
Affiliation(s)
- Christian Mitri
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Emmanuel Bischoff
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Eugeni Belda Cuesta
- Integromics Unit, Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Stevenn Volant
- Bioinformatics and Biostatistics Hub, Department of Computational Biology, Institut Pasteur, Paris, France
- CNRS USR 3756, Institut Pasteur, Paris, France
| | - Amine Ghozlane
- Bioinformatics and Biostatistics Hub, Department of Computational Biology, Institut Pasteur, Paris, France
- CNRS USR 3756, Institut Pasteur, Paris, France
| | - Karin Eiglmeier
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Inge Holm
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Constentin Dieme
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Emma Brito-Fravallo
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Wamdaogo M. Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - N’Fale Sagnon
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Michelle M. Riehle
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kenneth D. Vernick
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| |
Collapse
|
83
|
Bacteriocins of Listeria monocytogenes and Their Potential as a Virulence Factor. Toxins (Basel) 2020; 12:toxins12020103. [PMID: 32033406 PMCID: PMC7076858 DOI: 10.3390/toxins12020103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 12/28/2022] Open
Abstract
Intestinal microbiota exerts protective effects against the infection of various bacterial pathogens, including Listeria monocytogenes, a major foodborne pathogen whose infection can lead to a disease (listeriosis) with a high fatality rate. As a strategy to mitigate the action of the intestinal microbiota, pathogens often produce antimicrobial proteinaceous compounds such as bacteriocins. In this review, we summarize the information currently available for the well-characterized L. monocytogenes bacteriocin listeriolysin S, with the emphasis on its intriguing mode of action as a virulence factor, which promotes the infection of L. monocytogenes by changing the composition of the intestinal microbiota. We then discuss another intriguing L. monocytogenes bacteriocin Lmo2776 that specifically inhibits the inflammogenic species, Prevotella copri, in the intestinal microbiota, reducing superfluous inflammation while weakening virulence. In addition, we describe relatively less studied phage tail-like Listeria bacteriocins (monocins) and elaborate on the possibility that these monocins could be involved in enhancing pathogenicity. In spite of the burgeoning interest in the roles played by the intestinal microbiota against the L. monocytogenes infection, our understanding on the virulence factors affecting the intestinal microbiota is still lacking, calling for further studies on bacteriocins that could function as novel virulence factors.
Collapse
|
84
|
Tavares RDM, Silva DALD, Camargo AC, Yamatogi RS, Nero LA. Interference of the acid stress on the expression of llsX by Listeria monocytogenes pathogenic island 3 (LIPI-3) variants. Food Res Int 2020; 132:109063. [PMID: 32331684 DOI: 10.1016/j.foodres.2020.109063] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/13/2022]
Abstract
Listeria monocytogenes harbor different virulence factors, with a highly heterogeneous distribution between distinct lineages and serotypes. The Listeria Pathogenicity Island 3 (LIPI-3), mainly described in lineage I, encodes for Listeriolysin S (LLS), a virulence factor expressed by L. monocytogenes in the gastrointestinal tract during in vivo infections. The aim of this study was to carry out a comparative genotypic analysis of LIPI-3 identified in L. monocytogenes isolates obtained in Brazil and subjected to whole genomic sequencing (WGS). In addition, transcription of llsX expression under different acid stress conditions was evaluated by RT-PCR. Homologues of the eight LIPI-3 genes (llsAGHXBYDP) were identified in 15 isolates (all from lineage I) representative of different sequence types: ST1 (n = 3), ST3 (n = 6), ST218 (n = 5) and ST288 (n = 1). Single nucleotide polymorphism (SNP) analysis revealed that genetic variation resulted in modification of the final peptide LLS for ST218 (serogroup IVb-v1) and ST288 (serogroup IIb). Selected strains from ST3 and ST288 were subjected to acid stress conditions and the expression of llsX, a LIPI-3 gene, was observed: only F2365 (4b/ST1) presented llsX expression after six hours of acid stress, indicating relevant differences when compared to isolates IIb (ST3 and 288). The results highlight the presence of genomic variations on LIPI-3 and llsX expression under acid stress conditions, demanding further studies to evaluate if these mutations have an impact on L. monocytogenes virulence in vivo.
Collapse
Affiliation(s)
- Rafaela de Melo Tavares
- Universidade Federal de Viçosa, Departamento de Veterinária, Laboratório de Inspeção de Produtos de Origem Animal, Campus UFV, Centro, 36570 900, Viçosa, MG, Brazil
| | - Danilo Augusto Lopes da Silva
- Universidade Federal de Viçosa, Departamento de Veterinária, Laboratório de Inspeção de Produtos de Origem Animal, Campus UFV, Centro, 36570 900, Viçosa, MG, Brazil
| | - Anderson Carlos Camargo
- Universidade Federal de Viçosa, Departamento de Veterinária, Laboratório de Inspeção de Produtos de Origem Animal, Campus UFV, Centro, 36570 900, Viçosa, MG, Brazil
| | - Ricardo Seiti Yamatogi
- Universidade Federal de Viçosa, Departamento de Veterinária, Laboratório de Inspeção de Produtos de Origem Animal, Campus UFV, Centro, 36570 900, Viçosa, MG, Brazil
| | - Luís Augusto Nero
- Universidade Federal de Viçosa, Departamento de Veterinária, Laboratório de Inspeção de Produtos de Origem Animal, Campus UFV, Centro, 36570 900, Viçosa, MG, Brazil.
| |
Collapse
|
85
|
Antibody-coated microbiota in nasopharynx of healthy individuals and IVIg-treated patients with hypogammaglobulinemia. J Allergy Clin Immunol 2020; 145:1686-1690.e4. [PMID: 31954779 DOI: 10.1016/j.jaci.2020.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 11/23/2022]
|
86
|
Li Y, Rebuffat S. The manifold roles of microbial ribosomal peptide-based natural products in physiology and ecology. J Biol Chem 2020; 295:34-54. [PMID: 31784450 PMCID: PMC6952617 DOI: 10.1074/jbc.rev119.006545] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ribosomally synthesized and posttranslationally modified peptides (RiPPs), also called ribosomal peptide natural products (RPNPs), form a growing superfamily of natural products that are produced by many different organisms and particularly by bacteria. They are derived from precursor polypeptides whose modification by various dedicated enzymes helps to establish a vast array of chemical motifs. RiPPs have attracted much interest as a source of potential therapeutic agents, and in particular as alternatives to conventional antibiotics to address the bacterial resistance crisis. However, their ecological roles in nature are poorly understood and explored. The present review describes major RiPP actors in competition within microbial communities, the main ecological and physiological functions currently evidenced for RiPPs, and the microbial ecosystems that are the sites for these functions. We envision that the study of RiPPs may lead to discoveries of new biological functions and highlight that a better knowledge of how bacterial RiPPs mediate inter-/intraspecies and interkingdom interactions will hold promise for devising alternative strategies in antibiotic development.
Collapse
Affiliation(s)
- Yanyan Li
- Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM, UMR 7245 CNRS-MNHN), National Museum of Natural History (MNHN), CNRS, CP 54, 57 rue Cuvier 75005, Paris, France.
| | - Sylvie Rebuffat
- Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM, UMR 7245 CNRS-MNHN), National Museum of Natural History (MNHN), CNRS, CP 54, 57 rue Cuvier 75005, Paris, France.
| |
Collapse
|
87
|
Louie A, Zhang T, Becattini S, Waldor MK, Portnoy DA. A Multiorgan Trafficking Circuit Provides Purifying Selection of Listeria monocytogenes Virulence Genes. mBio 2019; 10:e02948-19. [PMID: 31848289 PMCID: PMC6918090 DOI: 10.1128/mbio.02948-19] [Citation(s) in RCA: 27] [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: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Listeria monocytogenes can cause a life-threatening illness when the foodborne pathogen spreads beyond the intestinal tract to distant organs. Many aspects of the intestinal phase of L. monocytogenes pathogenesis remain unknown. Here, we present a foodborne infection model using C57BL/6 mice that have been pretreated with streptomycin. In this model, as few as 100 L. monocytogenes CFU were required to cause self-limiting enterocolitis, and systemic dissemination followed previously reported routes. Using this model, we report that listeriolysin O (LLO) and actin assembly-inducing protein (ActA), two critical virulence determinants, were necessary for intestinal pathology and systemic spread but were dispensable for intestinal growth. Sequence tag-based analysis of microbial populations (STAMP) was used to investigate the within-host population dynamics of wild-type and LLO-deficient strains. The wild-type bacterial population experienced severe bottlenecks over the course of infection, and by 5 days, the intestinal population was highly enriched for bacteria originating from the gallbladder. In contrast, LLO-deficient strains did not efficiently disseminate and gain access to the gallbladder, and the intestinal population remained diverse. These findings suggest that systemic spread and establishment of a bacterial reservoir in the gallbladder imparts an intraspecies advantage in intestinal occupancy. Since intestinal L. monocytogenes is ultimately released into the environment, within-host population bottlenecks may provide purifying selection of virulence genes.IMPORTANCEListeria monocytogenes maintains capabilities for free-living growth in the environment and for intracellular replication in a wide range of hosts, including livestock and humans. Here, we characterized an enterocolitis model of foodborne L. monocytogenes infection. This work highlights a multiorgan trafficking circuit and reveals a fitness advantage for bacteria that successfully complete this cycle. Because virulence factors play critical roles in systemic dissemination and multiple bottlenecks occur as the bacterial population colonizes different tissue sites, this multiorgan trafficking circuit likely provides purifying selection of virulence genes. This study also serves as a foundation for future work using the L. monocytogenes-induced enterocolitis model to investigate the biology of L. monocytogenes in the intestinal environment.
Collapse
Affiliation(s)
- Alexander Louie
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Ting Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Simone Becattini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
88
|
Umu ÖCO, Gueimonde M, Oostindjer M, Ovchinnikov KV, de los Reyes-Gavilán CG, Arbulu S, Hernández PE, Martínez B, Diep DB, Salazar N. Use of Fecal Slurry Cultures to Study In Vitro Effects of Bacteriocins on the Gut Bacterial Populations of Infants. Probiotics Antimicrob Proteins 2019; 12:1218-1225. [DOI: 10.1007/s12602-019-09614-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
89
|
Camargo AC, Moura A, Avillan J, Herman N, McFarland AP, Sreevatsan S, Call DR, Woodward JJ, Lecuit M, Nero LA. Whole-genome sequencing reveals Listeria monocytogenes diversity and allows identification of long-term persistent strains in Brazil. Environ Microbiol 2019; 21:4478-4487. [PMID: 31251828 PMCID: PMC7644123 DOI: 10.1111/1462-2920.14726] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/12/2019] [Accepted: 06/24/2019] [Indexed: 01/06/2023]
Abstract
Advances in whole-genome sequencing (WGS) technologies have documented genetic diversity and epidemiology of the major foodborne pathogen Listeria monocytogenes (Lm) in Europe and North America, but data concerning South America are scarce. Here, we examined the population structure and genetic diversity of this major foodborne pathogen collected in Brazil. Based on core genome multilocus sequence typing (cgMLST), isolates from lineages I (n = 22; 63%) and II (n = 13; 37%) were distributed into 10 different sublineages (SLs) and represented 31 new cgMLST types (CTs). The most prevalent SLs were SL9 (n = 9; 26%), SL3 (n = 6; 17%) and SL2 and SL218 (n = 5; 14%). Isolates belonging to CTs L2-SL9-ST9-CT4420 and L1-SL315-ST520-CT4429 were collected 3 and 9 years apart, respectively, revealing long-term persistence of Lm in Brazil. Genetic elements associated with stress survival were present in 60% of isolates (57% SSI-1 and 3% SSI-2). Pathogenic islands were present in 100% (LIPI-1), 43% (LIPI-3) and 6% (LIPI-4) of the isolates. Mutations leading to premature stop codons were detected in the prfA and inlA virulence genes. This study is an important contribution to understanding the genomic diversity and epidemiology of Lm in South America. In addition, the results highlight the importance of using WGS to reveal Lm long-term persistence.
Collapse
Affiliation(s)
- Anderson C. Camargo
- Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Alexandra Moura
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
| | - Johannetsy Avillan
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, USA
| | - Nicole Herman
- Veterinary Population Medicine Department, University of Minnesota, St. Paul, MN, USA
| | | | - Srinand Sreevatsan
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Douglas R. Call
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, USA
| | | | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
- Université de Paris, Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, APHP, Institut Imagine, Paris, France
| | - Luís A. Nero
- Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| |
Collapse
|
90
|
Mohammadzadeh R, Azadegan A, Kalani BS. Listeriolysin S may inhibit the anti-listerial properties of Lactobacillus plantarum. Microb Pathog 2019; 137:103744. [PMID: 31521800 DOI: 10.1016/j.micpath.2019.103744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 11/16/2022]
Abstract
Listeriosis is a serious infection linked to the consumption of food contaminated with Listeria monocytogenes. Outbreaks and mortality rates associated with this infection make it a significant public health concern. As biocontrol agents, probiotics such as Lactobacillus plantarum had been of interest for the promotion of antilisterial activities. However, a recent bacteriocin from epidemic L. monocytogenes strains called listeriolysin S (LLS) has been identified with the ability to target the prokaryotic cells that may hinder the anti-listerial properties of L. plantarum. The present study was designed to investigate the interplay between serotypes 4b (lineage I, LLS-producing strain) and 1/2a (NCTC7973, lineage II, non LLS-producing strain) L. monocytogenes and L. plantarum ATCC13643. According to the results of the co-culture assay, L. plantarum significantly reduced the growth of LLS- L. monocytogenes. However, there was a significant reduction in the growth of L. plantarum when co-cultured with LLS + L. monocytogenes. Moreover, according to the results of the culture assay using Caco-2 cell line, there was a significant reduced intracellular count of LLS- L. monocytogenes after L. plantarum exposure, whereas, no major differences were observed in the intracellular count of LLS + L. monocytogenes. These results suggest that L. plantarum may be unable to inhibit infections caused by LLS-producing L. monocytogenes. Also, phylogenetic studies showed the presence of LLS-like proteins in several environmental isolates including L. innocua which suggests a role for LLS in survival and bacterial colonization in harsh conditions. In overall, the ability of LLS to target certain bacterial cells should be taken into consideration during the development of anti-listerial probiotics. Future experiments are required to elucidate the exact mechanisms by which LLS achieves bacterial killing.
Collapse
Affiliation(s)
- Rokhsareh Mohammadzadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Azadegan
- Department of Microbiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
91
|
Phylogenetically Defined Isoforms of Listeria monocytogenes Invasion Factor InlB Differently Activate Intracellular Signaling Pathways and Interact with the Receptor gC1q-R. Int J Mol Sci 2019; 20:ijms20174138. [PMID: 31450632 PMCID: PMC6747193 DOI: 10.3390/ijms20174138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/10/2019] [Accepted: 08/15/2019] [Indexed: 01/19/2023] Open
Abstract
The pathogenic Gram-positive bacterium Listeria monocytogenes has been evolving into a few phylogenetic lineages. Phylogenetically defined substitutions were described in the L. monocytogenes virulence factor InlB, which mediates active invasion into mammalian cells via interactions with surface receptors c-Met and gC1q-R. InlB internalin domain (idInlB) is central to interactions with c-Met. Here we compared activity of purified recombinant idInlB isoforms characteristic for L. monocytogenes phylogenetic lineage I and II. Size exclusion chromatography and intrinsic fluorescence were used to characterize idInlBs. Western blotting was used to study activation of c-Met-dependent MAPK- and PI3K/Akt-pathways. Solid-phase microplate binding and competition assay was used to quantify interactions with gCq1-R. Isogenic recombinant L. monocytogenes strains were used to elucidate the input of idInlB isoforms in HEp-2 cell invasion. Physicochemical parameters of idInlB isoforms were similar but not identical. Kinetics of Erk1/2 and Akt phosphorylation in response to purified idInlBs was lineage specific. Lineage I but not lineage II idInlB specifically bound gC1q-R. Antibody against gC1q-R amino acids 221–249 inhibited invasion of L. monocytogenes carrying lineage I but not lineage II idInlB. Taken together, obtained results suggested that phylogenetically defined substitutions in idInlB provide functional distinctions and might be involved in phylogenetically determined differences in virulence potential.
Collapse
|
92
|
Ducarmon QR, Zwittink RD, Hornung BVH, van Schaik W, Young VB, Kuijper EJ. Gut Microbiota and Colonization Resistance against Bacterial Enteric Infection. Microbiol Mol Biol Rev 2019; 83:e00007-19. [PMID: 31167904 PMCID: PMC6710460 DOI: 10.1128/mmbr.00007-19] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome is critical in providing resistance against colonization by exogenous microorganisms. The mechanisms via which the gut microbiota provide colonization resistance (CR) have not been fully elucidated, but they include secretion of antimicrobial products, nutrient competition, support of gut barrier integrity, and bacteriophage deployment. However, bacterial enteric infections are an important cause of disease globally, indicating that microbiota-mediated CR can be disturbed and become ineffective. Changes in microbiota composition, and potential subsequent disruption of CR, can be caused by various drugs, such as antibiotics, proton pump inhibitors, antidiabetics, and antipsychotics, thereby providing opportunities for exogenous pathogens to colonize the gut and ultimately cause infection. In addition, the most prevalent bacterial enteropathogens, including Clostridioides difficile, Salmonella enterica serovar Typhimurium, enterohemorrhagic Escherichia coli, Shigella flexneri, Campylobacter jejuni, Vibrio cholerae, Yersinia enterocolitica, and Listeria monocytogenes, can employ a wide array of mechanisms to overcome colonization resistance. This review aims to summarize current knowledge on how the gut microbiota can mediate colonization resistance against bacterial enteric infection and on how bacterial enteropathogens can overcome this resistance.
Collapse
Affiliation(s)
- Q R Ducarmon
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - R D Zwittink
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - B V H Hornung
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - W van Schaik
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - V B Young
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - E J Kuijper
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Clinical Microbiology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Donor Feces Bank, Leiden, Netherlands
| |
Collapse
|
93
|
Zhang Y, Dong S, Chen H, Chen J, Zhang J, Zhang Z, Yang Y, Xu Z, Zhan L, Mei L. Prevalence, Genotypic Characteristics and Antibiotic Resistance of Listeria monocytogenes From Retail Foods in Bulk in Zhejiang Province, China. Front Microbiol 2019; 10:1710. [PMID: 31402906 PMCID: PMC6672743 DOI: 10.3389/fmicb.2019.01710] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 07/11/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes is an important foodborne pathogen causing public concern. A total of 3354 retail foods in bulk were sampled and screened for L. monocytogenes. Seventy-three (2.2%) samples including 21 ready-to-eat (RTE) foods and 52 raw foods were confirmed positive for L. monocytogenes. Sushi and salmon sashimi occupied the top two slots in RTE foods with relatively high presence rate of 12.9 and 6.9%, respectively. Meanwhile, L. monocytogenes was found to be distributed unequally in raw foods; the presence rates in raw meat (3.5%) and poultry (3.8%) were significantly higher than that in raw seafood (1.3%). Notably, L. monocytogenes was not detected in raw freshwater food. The L. monocytogenes isolates belonged to four serotypes, 1/2a, 1/2b, 1/2c, and 4b, with the most prevalent serotype being 1/2a (47.9%). Eighteen sequence types (STs) and eighteen virulence types (VTs) containing four newly assigned VTs (VT180, VT181, VT182, and VT183) were determined via multilocus sequence typing (MLST) and multi-virulence-locus sequence typing (MVLST). Among the 73 L. monocytogenes isolates, 23 (31.5%) belonged to epidemic clones (ECs) including ECI, ECIV, ECV, ECVI, ECVIII and ECXI among which ECV was predominant. Antibiotic susceptibility tests revealed a high resistance rate (11.0%) to tetracycline. Moreover, we identified the distribution patterns of virulence genes of four Listeria pathogenicity islands (LIPI) in L. monocytogenes isolates. prfA, hly, plcA, plcB, mpl, actA genes in LIPI-1 and inlA, inlB, inlC, inlJ genes in LIPI-2 were detected in approximately all L. monocytogenes isolates. The distribution of both LIPI-3 genes and LIPI-4 genes exhibited association with lineage and ST. LIPI-4 genes were present exclusively in ST87 isolates. Relatedness analysis revealed the absence of distinct association between STs, ECs, LIPI-3 and LIPI-4 distribution and specific food groups. This study provided fundamental data for Chinese food safety authorities to grasp the contamination status of L. monocytogenes in foods, assess the potential risk of this pathogen and further address the safety issue of retail foods in bulk in China.
Collapse
Affiliation(s)
- Yunyi Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Shilei Dong
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, China
| | - Honghu Chen
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jiancai Chen
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Junyan Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhen Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yong Yang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ziyan Xu
- Department of Biotechnology, Wenzhou Medical University, Wenzhou, China
| | - Li Zhan
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lingling Mei
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
94
|
Oviedo JM, Surmann K, Gorgojo JP, Valdez H, Dhople VM, Lamberti Y, Völker U, Rodriguez ME. Shotgun proteomic analysis of Bordetella parapertussis provides insights into the physiological response to iron starvation and potential new virulence determinants absent in Bordetella pertussis. J Proteomics 2019; 206:103448. [PMID: 31325608 DOI: 10.1016/j.jprot.2019.103448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/19/2019] [Accepted: 07/11/2019] [Indexed: 01/05/2023]
Abstract
Bordetella parapertussis is one of the pathogens that cause whooping cough. Even though its incidence has been rising in the last decades, this species remained poorly investigated. This study reports the first extensive proteome analysis of this bacterium. In an attempt to gain some insight into the infective phenotype, we evaluated the response of B. parapertussis to iron starvation, a critical stress the bacteria face during infection. Among other relevant findings, we observed that the adaptation to this condition involves significant changes in the abundance of two important virulence factors of this pathogen, namely, adenylate cyclase and the O-antigen. We further used the proteomic data to search for B. parapertussis proteins that are absent or classified as pseudogenes in the genome of Bordetella pertussis to unravel differences between both whooping cough causative agents. Among them, we identified proteins involved in stress resistance and virulence determinants that might help to explain the differences in the pathogenesis of these species and the lack of cross-protection of current acellular vaccines. Altogether, these results contribute to a better understanding of B. parapertussis biology and pathogenesis. SIGNIFICANCE: Whooping cough is a reemerging disease caused by both Bordetella pertussis and Bordetella parapertussis. Current vaccines fail to induce protection against B parapertussis and the incidence of this species has been rising over the years. The proteomic analysis of this study provided relevant insights into potential virulence determinants of this poorly-studied pathogen. It further identified proteins produced by B. parapertussis not present in B. pertussis, which might help to explain both the differences on their respective infectious process and the current vaccine failure. Altogether, the results of this study contribute to the better understanding of B. parapertussis pathogenesis and the eventual design of improved preventive strategies against whooping cough.
Collapse
Affiliation(s)
- Juan Marcos Oviedo
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Kristin Surmann
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Juan Pablo Gorgojo
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Hugo Valdez
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Vishnu M Dhople
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Yanina Lamberti
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - María Eugenia Rodriguez
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| |
Collapse
|
95
|
Johansson J, Freitag NE. Regulation of Listeria monocytogenes Virulence. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0064-2019. [PMID: 31441398 PMCID: PMC10957223 DOI: 10.1128/microbiolspec.gpp3-0064-2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Indexed: 02/07/2023] Open
Abstract
Whereas obligate human and animal bacterial pathogens may be able to depend upon the warmth and relative stability of their chosen replication niche, environmental bacteria such as Listeria monocytogenes that harbor the ability to replicate both within animal cells and in the outside environment must maintain the capability to manage life under a variety of disparate conditions. Bacterial life in the outside environment requires adaptation to wide ranges of temperature, available nutrients, and physical stresses such as changes in pH and osmolarity as well as desiccation. Following ingestion by a susceptible animal host, the bacterium must adapt to similar changes during transit through the gastrointestinal tract and overcome a variety of barriers associated with host innate immune responses. Rapid alteration of patterns of gene expression and protein synthesis represent one strategy for quickly adapting to a dynamic host landscape. Here, we provide an overview of the impressive variety of strategies employed by the soil-dwelling, foodborne, mammalian pathogen L. monocytogenes to straddle diverse environments and optimize bacterial fitness both inside and outside host cells.
Collapse
Affiliation(s)
- Jörgen Johansson
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden (MIMS) and Umeå Centre for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| | - Nancy E Freitag
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago IL
| |
Collapse
|
96
|
Valente GLC, Acurcio LB, Freitas LPV, Nicoli JR, Silva AM, Souza MR, Penna CFAM. Short communication: In vitro and in vivo probiotic potential of Lactobacillus plantarum B7 and Lactobacillus rhamnosus D1 isolated from Minas artisanal cheese. J Dairy Sci 2019; 102:5957-5961. [PMID: 31128873 DOI: 10.3168/jds.2018-15938] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/23/2019] [Indexed: 12/15/2022]
Abstract
Some Lactobacillus strains may contribute to the health of the host when administered in adequate concentrations, demonstrating their probiotic potential. In contrast, Listeria monocytogenes is a foodborne pathogen that can cause enteropathy, meningoencephalitis, abortion, and septicemia. The aim of this survey was to evaluate the in vitro and in vivo probiotic potential of Lactobacillus plantarum B7 and Lactobacillus rhamnosus D1, isolated from Minas artisanal cheese of the Serra da Canastra (Minas Gerais, Brazil), against Lis. monocytogenes. We submitted B7 and D1 to in vitro testing (antibiogram, tolerance to bile salts and artificial gastric fluid, and spot-on-lawn) and in vivo testing (relative weight gain in mice). Both Lactobacillus strains demonstrated in vitro inhibitory activity against Lis. monocytogenes, as well as sensitivity to antimicrobials and resistance to gastric acids and bile salts. In the in vivo assays, mice treated with D1 gained more weight than mice in the other groups. These results indicate that D1 could have higher probiotic potential than B7 because improvements in feed conversion may help animals fight infection.
Collapse
Affiliation(s)
- G L C Valente
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - L B Acurcio
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - L P V Freitas
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - J R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - A M Silva
- Departamento de Engenharia de Alimentos, Universidade Federal de São João del-Rei, Sete Lagoas, 35701-970, Brazil
| | - M R Souza
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - C F A M Penna
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| |
Collapse
|
97
|
Chen M, Cheng J, Zhang J, Chen Y, Zeng H, Xue L, Lei T, Pang R, Wu S, Wu H, Zhang S, Wei X, Zhang Y, Ding Y, Wu Q. Isolation, Potential Virulence, and Population Diversity of Listeria monocytogenes From Meat and Meat Products in China. Front Microbiol 2019; 10:946. [PMID: 31134008 PMCID: PMC6514097 DOI: 10.3389/fmicb.2019.00946] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/15/2019] [Indexed: 12/23/2022] Open
Abstract
Listeria monocytogenes is a globally notorious foodborne pathogen. This study aimed to qualitatively and quantitatively detect L. monocytogenes from meat and meat products in China and to establish their virulence profiles and population diversity. From 1212 meat and meat product samples, 362 (29.9%) were positive for L. monocytogenes. Of these positive samples, 90.6% (328/362) had less than 10 MPN/g, 5.5% (20/364) samples had 10-110 MPN/g, and 3.9% (14/362) of the positive samples had over 110 MPN/g. Serogroup analysis showed that the most prevalent serogroup of L. monocytogenes was I.1 (1/2a-3a), which accounted for 45.0% (123/458) of the total, followed by serogroup I.2 (1/2c-3c) that comprised 26.9%, serogroup II.1 (4b-4d-4e) that comprised 4.8%, and serogroup II.2 (1/2b-3b-7) that comprised 23.3%. A total of 458 isolates were grouped into 35 sequence types (STs) that belonged to 25 clonal complexes (CCs) and one singleton (ST619) by multi-locus sequence typing. The most prevalent ST was ST9 (26.9%), followed by ST8 (17.9%), ST87 (15.3%), ST155 (9.4%), and ST121 (7.6%). Thirty-seven isolates harbored the llsX gene (representing LIPI-3), and they belonged to ST1/CC1, ST3/CC3, ST288/CC288, ST323/CC288, ST330/CC288, ST515/CC1, and ST619, among which ST323/CC288, ST330/CC288, and ST515/CC1 were newly reported to carry LIPI-3. Seventy-five isolates carried ptsA, and they belonged to ST87/CC87, ST88/CC88, and ST619, indicating that consumers may be exposed to potential hypervirulent L. monocytogenes. Antibiotics susceptibility tests revealed that over 90% of the isolates were susceptible to 11 antibiotics; however, 40.0% of the isolates exhibited resistance against ampicillin and 11.8% against tetracycline; further, 45.0 and 4.6% were intermediate resistant and resistant to ciprofloxacin, respectively. The rise of antibiotic resistance in L. monocytogenes suggests that stricter regulations should be formulated to restrict the use of antibiotic agents in human listeriosis treatment and livestock breeding.
Collapse
Affiliation(s)
- Moutong Chen
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Jianheng Cheng
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Jumei Zhang
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Yuetao Chen
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Haiyan Zeng
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Liang Xue
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Tao Lei
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Rui Pang
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Shi Wu
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Haoming Wu
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Shuhong Zhang
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Xianhu Wei
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Youxiong Zhang
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| | - Yu Ding
- Department of Food Science and Technology, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Institute of Microbiology, State Key Laboratory of Applied Microbiology Southern China, Guangdong Open Laboratory of Applied Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangzhou, China
| |
Collapse
|
98
|
Chung LK, Raffatellu M. G.I. pros: Antimicrobial defense in the gastrointestinal tract. Semin Cell Dev Biol 2019; 88:129-137. [PMID: 29432952 PMCID: PMC6087682 DOI: 10.1016/j.semcdb.2018.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 01/11/2023]
Abstract
The gastrointestinal tract is a complex environment in which the host immune system interacts with a diverse array of microorganisms, both symbiotic and pathogenic. As such, mobilizing a rapid and appropriate antimicrobial response depending on the nature of each stimulus is crucial for maintaining the balance between homeostasis and inflammation in the gut. Here we focus on the mechanisms by which intestinal antimicrobial peptides regulate microbial communities during dysbiosis and infection. We also discuss classes of bacterial peptides that contribute to reducing enteric pathogen outgrowth. This review aims to provide a comprehensive overview on the interplay of diverse antimicrobial responses with enteric pathogens and the gut microbiota.
Collapse
Affiliation(s)
- Lawton K Chung
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La Jolla, CA, 92093-0704, United States
| | - Manuela Raffatellu
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La Jolla, CA, 92093-0704, United States; Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla CA, United States.
| |
Collapse
|
99
|
Vilchis-Rangel RE, Espinoza-Mellado MDR, Salinas-Jaramillo IJ, Martinez-Peña MD, Rodas-Suárez OR. Association of Listeria monocytogenes LIPI-1 and LIPI-3 marker llsX with invasiveness. Curr Microbiol 2019; 76:637-643. [PMID: 30888475 DOI: 10.1007/s00284-019-01671-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 03/13/2019] [Indexed: 12/29/2022]
Abstract
Listeria monocytogenes is an opportunistic pathogen that is widely distributed in the environment. The evolution of its genome has exhibited differences in virulence among strains of the same species. Listeria monocytogenes LIPI-3 (Listeria Pathogenicity Island 3) and LIPI-1 (Listeria Pathogenicity Island 1) are considered responsible for the increased virulence in some strains. The aim of this study was to detect LIPI-1 genes and the llsX gene belonging to LIPI-3 in invasive strains of L. monocytogenes and to establish whether there is a relationship among the invasiveness, presence of the llsX and LIPI-1 genes, and the source of the strains. The results showed that 70% of the strains were invasive, and all these strains except one possessed LIPI-1, which suggests that although there is a correlation between LIPI-1 and invasiveness, the independent mechanisms of LIPI-1 may contribute to invasiveness. In contrast, 35% of the total strains were positive for llsX and were invasive; thus, the results revealed that there is a strong association between llsX and the invasiveness of L. monocytogenes in HEp-2 cells (HeLa contaminant/epithelial in origin). In addition, there is no other association with any other variable in this study. Moreover, the authors found that LIPI-1 and llsX are more frequently found in fresh than in frozen vegetables. Together, the findings provide an approximation for the better understanding of Listeriolysin S (LLS) and its role in the pathogenesis of L. monocytogenes, and a possible relation between virulence factors and food-storage temperature.
Collapse
Affiliation(s)
- Rodolfo Erik Vilchis-Rangel
- Laboratorio de Bacteriología Médica, Depto. Microbiología, Escuela Nacional de Ciencias Biológicas-Instituto Politécnico Nacional (ENCB-IPN), Prolongación de Carpio y Plan de Ayala, 11340, Mexico City, CDMX, Mexico
| | - María Del Rosario Espinoza-Mellado
- Depto. Investigación, Central de Instrumentación de Microscopia, ENCB-IPN, Prolongación de Carpio y Plan de Ayala, 11340, Mexico City, CDMX, Mexico.
| | - Irving Jesús Salinas-Jaramillo
- Laboratorio de Microbiología General, Depto. Microbiología, ENCB-IPN, Prolongación de Carpio y Plan de Ayala, 11340, Mexico City, CDMX, Mexico
| | - Marcos Daniel Martinez-Peña
- Laboratorio de Recursos Genéticos Microbianos, Centro Nacional de Recursos Genéticos (CNRG), Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias Boulevard de la Biodiversidad 400, Rancho las Cruces, C.P. 47600, Tepatitlán de Morelos, Jalisco, Mexico
| | - Oscar Rodolfo Rodas-Suárez
- Laboratorio de Microbiología General, Depto. Microbiología, ENCB-IPN, Prolongación de Carpio y Plan de Ayala, 11340, Mexico City, CDMX, Mexico
| |
Collapse
|
100
|
Riaz A, Noureen S, Liqat I, Arshad M, Arshad N. Antilisterial efficacy of Lactobacillus brevis MF179529 from cow: an in vivo evidence. Altern Ther Health Med 2019; 19:37. [PMID: 30709347 PMCID: PMC6359795 DOI: 10.1186/s12906-019-2444-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 01/18/2019] [Indexed: 01/14/2023]
Abstract
Background Listeria monocytogenes is an opportunistic foodborne pathogen that causes human Listeriosis and high mortality particularly in immunocompromised individuals. Pregnant women are more prone to L. monocytogenes infection resulting in abortions. In the present study, antilisterial activity of Lactobacillus brevis (LB) MF179529, a probiotic bacterial strain, was investigated in a murine model. Methods Initially a pilot study was conducted to determine the dose of L. monocytogenes required to cause symptomatic listeriosis. In the main trial, mice were divided into 4 groups. Group I was kept as negative control, group II was exposed to L. monocytogenes and maintained as positive control. Group III was fed with L. brevis only, while group IV received L. brevis for 3 days prior to L. monocytogenes infection. A volume of 200 μl of L. monocytogenes ATCC 19115 and L. brevis MF179529 bacterial suspension corresponding to cell density of 109CFU/ml were given to respective groups by intragastric route. Progress of infection was monitored for 7 days including general health scoring, listeria dispersion in organs, bacterial load in intestine and blood biochemistry were recorded on 3rd, 5th and 7th days post infection (dpi). Results Clinical listeriosis was induced by 109CFU/ml of L. monocytogenes ATCC 19115 in mice. Animals of group IV displayed minor signs of infection. L. brevis supplementation resulted in significant reduction in dispersion and propagation of L. monocytogenes in liver, spleen and intestine. L. brevis MF179529 consumption led to a significant elevation of number of lactic acid bacteria and reduction of total plate count, anaerobic count and coliform population in intestine. Moreover, total leukocyte and neutrophil counts of treated animals were similar to the negative control while positive control group displayed higher number. Safety evaluation of L. brevis was performed by monitoring general health, hematological and serological parameters of L. brevis fed and negative control group (group III and I). No significant difference in feed intake, body temperature, body weight and blood picture could be detected in L. brevis supplemented and control groups. Conclusion Our results indicate ameliorative role of L. brevis in L. monocytogenes infection and suggest that L. brevis could be used for prophylactic measure. Electronic supplementary material The online version of this article (10.1186/s12906-019-2444-5) contains supplementary material, which is available to authorized users.
Collapse
|