1
|
Oliveira M, Barbosa J, Teixeira P. Listeria monocytogenes gut interactions and listeriosis: Gut modulation and pathogenicity. Microbiol Res 2025; 297:128187. [PMID: 40279724 DOI: 10.1016/j.micres.2025.128187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Following ingestion via contaminated food, Listeria monocytogenes faces multiple hurdles through the human digestive system, thereby influencing its capacity to cause infection. This review provides a comprehensive overview of the multifaceted mechanisms employed by L. monocytogenes to overcome gastrointestinal hurdles and interact with the host's microbiota, facing chemical and physical barriers such as saliva, stomach acidity, bile salts and mechanical clearance. Proposed evasion strategies will be highlighted, exploring the bacteriocins produced by L. monocytogenes, such as the well-described bacteriocin Listeriolysin S (LLS), a bacteriocin that inhibits inflammogenic species - Lmo2776, and a phage tail-like bacteriocin, monocin. The competitive dynamic interactions within the gut microbiota, as well as the modulation of microbiota composition and immune responses, will also be explored. Finally, the adhesion and invasion of the intestinal epithelium by L. monocytogenes is described, exploring the mechanism of pathogenesis, biofilm and aggregation capacities and other virulence factors. Unlike previous reviews that may focus on individual aspects of L. monocytogenes pathogenicity, this review offers a holistic perspective on the bacterium's ability to persist and cause infection, integrating information about survival strategies, including bacteriocin production, immune modulation, and virulence factors. By connecting recent findings on microbial interactions and infection dynamics, this review incorporates recent developments in the field and connects various lines of research that explore both host and microbial factors influencing infection outcomes.
Collapse
Affiliation(s)
- M Oliveira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - J Barbosa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - P Teixeira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal.
| |
Collapse
|
2
|
Nasher F, Wren BW. Unravelling mechanisms of bacterial recognition by Acanthamoeba: insights into microbial ecology and immune responses. Front Microbiol 2024; 15:1405133. [PMID: 39247694 PMCID: PMC11377244 DOI: 10.3389/fmicb.2024.1405133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Acanthamoeba, are ubiquitous eukaryotic microorganisms, that play a pivotal role in recognizing and engulfing various microbes during predation, offering insights into microbial dynamics and immune responses. An intriguing observation lies in the apparent preference of Acanthamoeba for Gram-negative over Gram-positive bacteria, suggesting potential differences in the recognition and response mechanisms to bacterial prey. Here, we comprehensively review pattern recognition receptors (PRRs) and microbe associated molecular patterns (MAMPs) that influence Acanthamoeba interactions with bacteria. We analyze the molecular mechanisms underlying these interactions, and the key finding of this review is that Acanthamoeba exhibits an affinity for bacterial cell surface appendages that are decorated with carbohydrates. Notably, this parallels warm-blooded immune cells, underscoring a conserved evolutionary strategy in microbial recognition. This review aims to serve as a foundation for exploring PRRs and MAMPs. These insights enhance our understanding of ecological and evolutionary dynamics in microbial interactions and shed light on fundamental principles governing immune responses. Leveraging Acanthamoeba as a model organism, provides a bridge between ecological interactions and immunology, offering valuable perspectives for future research.
Collapse
Affiliation(s)
- Fauzy Nasher
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Brendan W Wren
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
3
|
Tang M, Tian S, Chen K, Zhang Q, Lei Y, Tang T, Zeng J, Wang C. Membrane vesicles derived from Listeria monocytogenes might be a potential antigen delivery vector. Int J Pharm 2023; 644:123275. [PMID: 37516216 DOI: 10.1016/j.ijpharm.2023.123275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Membrane vesicles (MVs) derived from Listeria monocytogenes (LM) have a natural nanoscale size and contain a variety of bacterial components. We speculated that LM MVs may be a novel delivery vector, but it is necessary to evaluate the safety and immunogenicity of LM MVs in vivo. Here, we isolated LM MVs and tested their safety and immunogenicity both in vitro and in vivo. The results showed that LM MVs stimulated RAW264.7 cells and DC2.4 cells to secrete the inflammatory cytokines IL-1β, TNF-α, IL-6 and IL-10. Intraperitoneal injection of LM MVs at 80 μg per C57BL/6 mouse did not cause lethal effects or irreversible pathological changes in major organs, indicating that LM MVs were safe. Intraperitoneal immunization of C57BL/6 mice twice with LM MVs mainly induced a high level of LM MV-specific IgG antibodies. In addition, we subcutaneously injected C57BL/6 mice with a mixture of ovalbumin and LM MVs and found that LM MVs exhibited a humoral immune adjuvant effect equal to that of the same amount of alum. The results of this study indicated that LM MVs have good safety and effective immunogenicity and may act as humoral immune adjuvants. Therefore, LM MVs are a potential new choice for antigen and drug delivery vectors.
Collapse
Affiliation(s)
- Mingyuan Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Sicheng Tian
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Kehan Chen
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Qiuyang Zhang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Yao Lei
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Tian Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Jumei Zeng
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China
| | - Chuan Wang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610061, China.
| |
Collapse
|
4
|
Oliveira AH, Tiensuu T, Guerreiro D, Tükenmez H, Dessaux C, García-del Portillo F, O’Byrne C, Johansson J. The Virulence and Infectivity of Listeria monocytogenes Are Not Substantially Altered by Elevated SigB Activity. Infect Immun 2023; 91:e0057122. [PMID: 37125941 PMCID: PMC10269059 DOI: 10.1128/iai.00571-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/26/2023] [Indexed: 05/02/2023] Open
Abstract
Listeria monocytogenes is a bacterial pathogen capable of causing severe infections but also thriving outside the host. To respond to different stress conditions, L. monocytogenes mainly utilizes the general stress response regulon, which largely is controlled by the alternative sigma factor Sigma B (SigB). In addition, SigB is important for virulence gene expression and infectivity. Upon encountering stress, a large multicomponent protein complex known as the stressosome becomes activated, ultimately leading to SigB activation. RsbX is a protein needed to reset a "stressed" stressosome and prevent unnecessary SigB activation in nonstressed conditions. Consequently, absence of RsbX leads to constitutive activation of SigB even without prevailing stress stimulus. To further examine the involvement of SigB in the virulence of this pathogen, we investigated whether a strain with constitutively active SigB would be affected in virulence factor expression and/or infectivity in cultured cells and in a chicken embryo infection model. Our results suggest that increased SigB activity does not substantially alter virulence gene expression compared with the wild-type (WT) strain at transcript and protein levels. Bacteria lacking RsbX were taken up by phagocytic and nonphagocytic cells at a similar frequency to WT bacteria, both in stressed and nonstressed conditions. Finally, the absence of RsbX only marginally affected the ability of bacteria to infect chicken embryos. Our results suggest only a minor role of RsbX in controlling virulence factor expression and infectivity under these conditions.
Collapse
Affiliation(s)
- Ana H. Oliveira
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre of Microbial Research, Umeå University, Umeå, Sweden
| | - Teresa Tiensuu
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre of Microbial Research, Umeå University, Umeå, Sweden
| | - Duarte Guerreiro
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre of Microbial Research, Umeå University, Umeå, Sweden
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hasan Tükenmez
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre of Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Charlotte Dessaux
- Laboratory of Intracellular Bacterial Pathogens, National Center of Biotechnology, (CNB)-CSIC, Madrid, Spain
| | | | - Conor O’Byrne
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Jörgen Johansson
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre of Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
5
|
Tasneem M, Gupta SD, Momin MB, Hossain KM, Osman TB, Rabbi MF. In silico annotation of a hypothetical protein from Listeria monocytogenes EGD-e unfolds a toxin protein of the type II secretion system. Genomics Inform 2023; 21:e7. [PMID: 37037465 PMCID: PMC10085738 DOI: 10.5808/gi.22071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/03/2023] [Indexed: 04/03/2023] Open
Abstract
The gram-positive bacterium Listeria monocytogenes is an important foodborne intracellular pathogen that is widespread in the environment. The functions of hypothetical proteins (HP) from various pathogenic bacteria have been successfully annotated using a variety of bioinformatics strategies. In this study, a HP Imo0888 (NP_464414.1) from the Listeria monocytogenes EGD-e strain was annotated using several bioinformatics tools. Various techniques, including CELLO, PSORTb, and SOSUIGramN, identified the candidate protein as cytoplasmic. Domain and motif analysis revealed that the target protein is a PemK/MazF-like toxin protein of the type II toxin-antitoxin system (TA) which was consistent with BLASTp analysis. Through secondary structure analysis, we found the random coil to be the most frequent. The Alpha Fold 2 Protein Structure Prediction Database was used to determine the three-dimensional (3D) structure of the HP using the template structure of a type II TA PemK/MazF family toxin protein (DB ID_AFDB: A0A4B9HQB9) with 99.1% sequence identity. Various quality evaluation tools, such as PROCHECK, ERRAT, Verify 3D, and QMEAN were used to validate the 3D structure. Following the YASARA energy minimization method, the target protein's 3D structure became more stable. The active site of the developed 3D structure was determined by the CASTp server. Most pathogens that harbor TA systems create a crucial risk to human health. Our aim to annotate the HP Imo088 found in Listeria could offer a chance to understand bacterial pathogenicity and identify a number of potential targets for drug development.
Collapse
Affiliation(s)
- Maisha Tasneem
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Shipan Das Gupta
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Monira Binte Momin
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Kazi Modasser Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Tasnim Binta Osman
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md. Fazley Rabbi
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
- Corresponding author: E-mail:
| |
Collapse
|
6
|
Adhikari P, Florien N, Gupta S, Kaushal A. Recent Advances in the Detection of Listeria monocytogenes. Infect Dis (Lond) 2023. [DOI: 10.5772/intechopen.109948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Listeria monocytogenes is the third-most severe pathogen causing a yearly outbreak of food poisoning in the world that proliferates widely in the environment. Infants, pregnant mothers, and immuno-compromised people are at high risk. Its ability to grow in both biotic and abiotic environments leads to epidemics that infect 5 out of 10 people annually. Because of the epithelial adhesion (by E-cadherin binding), it can suppress immune cells and thrive in the gastrointestinal tract till the brain through blood flow (E-cadherin). Microbial culture is still used as a gold standard, but takes a long time and often yields false positive results due to incompetence and temperature variations. Therefore, in order to treat it rather than using broad spectrum antibiotics, a standardized time-saving and highly specific technology for early detection is very important. It has been observed that the production of a particular antibody is delaying (so does the detection process) as a result of the inadequate understanding of the pathophysiology of the bacteria. This book chapter provides a brief summary of a pathogen as well as the scientific advances that led to its identification more easily.
Collapse
|
7
|
Zakrzewski AJ, Kurpas M, Zadernowska A, Chajęcka-Wierzchowska W, Fraqueza MJ. A Comprehensive Virulence and Resistance Characteristics of Listeria monocytogenes Isolated from Fish and the Fish Industry Environment. Int J Mol Sci 2023; 24:ijms24043581. [PMID: 36834997 PMCID: PMC9967382 DOI: 10.3390/ijms24043581] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Listeria monocytogenes is an important pathogen, often associated with fish, that can adapt and survive in products and food processing plants, where it can persist for many years. It is a species characterized by diverse genotypic and phenotypic characteristics. Therefore, in this study, a total of 17 L. monocytogenes strains from fish and fish-processing environments in Poland were characterized for their relatedness, virulence profiles, and resistance genes. The Core Genome Multilocus Sequence Typing (cgMLST) analysis revealed that the most frequent serogroups were IIa and IIb; sequence types (ST) were ST6 and ST121; and clonal complexes (CC) were CC6 and CC121. Core genome multilocus sequence typing (cgMLST) analysis was applied to compare the present isolates with the publicly available genomes of L. monocytogenes strains recovered in Europe from humans with listeriosis. Despite differential genotypic subtypes, most strains had similar antimicrobial resistance profiles; however, some of genes were located on mobile genetic elements that could be transferred to commensal or pathogenic bacteria. The results of this study showed that molecular clones of tested strains were characteristic for L. monocytogenes isolated from similar sources. Nevertheless, it is worth emphasizing that they could present a major public health risk due to their close relation with strains isolated from human listeriosis.
Collapse
Affiliation(s)
| | - Monika Kurpas
- Department of Immunobiology and Environmental Microbiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Anna Zadernowska
- Department of Industrial and Food Microbiology, University of Warmia and Mazrui, 10-726 Olsztyn, Poland
- Correspondence:
| | | | - Maria João Fraqueza
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
8
|
Tran BM, Linnik DS, Punter CM, Śmigiel WM, Mantovanelli L, Iyer A, O’Byrne C, Abee T, Johansson J, Poolman B. Super-resolving microscopy reveals the localizations and movement dynamics of stressosome proteins in Listeria monocytogenes. Commun Biol 2023; 6:51. [PMID: 36641529 PMCID: PMC9840623 DOI: 10.1038/s42003-023-04423-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
The human pathogen Listeria monocytogenes can cope with severe environmental challenges, for which the high molecular weight stressosome complex acts as the sensing hub in a complicated signal transduction pathway. Here, we show the dynamics and functional roles of the stressosome protein RsbR1 and its paralogue, the blue-light receptor RsbL, using photo-activated localization microscopy combined with single-particle tracking and single-molecule displacement mapping and supported by physiological studies. In live cells, RsbR1 is present in multiple states: in protomers with RsbS, large clusters of stressosome complexes, and in connection with the plasma membrane via Prli42. RsbL diffuses freely in the cytoplasm but forms clusters upon exposure to light. The clustering of RsbL is independent of the presence of Prli42. Our work provides a comprehensive view of the spatial organization and intracellular dynamics of the stressosome proteins in L. monocytogenes, which paves the way towards uncovering the stress-sensing mechanism of this signal transduction pathway.
Collapse
Affiliation(s)
- Buu Minh Tran
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Dmitrii Sergeevich Linnik
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Christiaan Michiel Punter
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Wojciech Mikołaj Śmigiel
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Luca Mantovanelli
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Aditya Iyer
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Conor O’Byrne
- Microbiology, School of Biological & Chemical Sciences, Ryan Institute, University of Galway, Galway, Ireland
| | - Tjakko Abee
- grid.4818.50000 0001 0791 5666Laboratory of Food Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Jörgen Johansson
- grid.12650.300000 0001 1034 3451Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Bert Poolman
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Alegbeleye O, Sant'Ana AS. Impact of temperature, soil type and compost amendment on the survival, growth and persistence of Listeria monocytogenes of non-environmental (food-source associated) origin in soil. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 843:157033. [PMID: 35777564 DOI: 10.1016/j.scitotenv.2022.157033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Listeria monocytogenes of varied sources including food-related sources may reach the soil. Associated food safety and environmental health risks of such contamination depend significantly on the capacity of L. monocytogenes to survive in the soil. This study assessed the survival of 13 L. monocytogenes strains isolated from food and food processing environments and a cocktail of three of the strains in two types of soils (loam and sandy) under controlled temperature conditions: 5, 10, 20, 25, 30℃ and 'uncontrolled' ambient temperature conditions in a tropical region. The impact of compost amendment on the survival of L. monocytogenes in the two different types of soils was also assessed. Soil type, temperature and compost amendment significantly (P <0.001) impacted the survival of L. monocytogenes in soil. Temperature variations affected the survival of L. monocytogenes in soil, where some strains such as strain 732, a L. monocytogenes 1/2a strain survived better at lower temperature (5°C), for which counts of up to 10.47 ± 0.005 log CFU/g were recovered in compost-amended sandy soil, 60 days post-inoculation. Some other strains such as strain 441, a L. monocytogenes 1/2a survived best at intermediate temperature (25 and 30 °C), while others such as 2739 (L. monocytogenes 1/2b) thrived at higher temperature (between 30 °C - 37 °C). There were significant correlations between the influence of temperature and soil type, where lower temperature conditions (5°C - 20°C) were generally more suitable for survival in sandy soil compared to higher temperature conditions. For some of the strains that thrived better in sandy soil at lower temperature, Pearson correlation analysis found significant correlations between temperature and soil type. Steady, controlled temperature generally favored the survival of the strains compared to uncontrolled ambient temperature conditions, except for the cocktail. The cocktail persisted until the last day of post-inoculation storage (60th day) in all test soils and under all incubation temperature conditions. Loam soil was more favorable for the survival of L. monocytogenes and compost amendment improved the survival of the strains, especially in compost-amended sandy soil. Listeria monocytogenes may exhibit variable survival capacity in soil, depending on conditions such as soil type, compost amendment and temperature.
Collapse
Affiliation(s)
- Oluwadara Alegbeleye
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Anderson S Sant'Ana
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
10
|
Di Renzo L, De Angelis ME, Torresi M, Di Lollo V, Di Teodoro G, Averaimo D, Defourny SVP, Di Giacinto F, Profico C, Olivieri V, Pomilio F, Cammà C, Ferri N, Di Francesco G. First Report of Septicaemic Listeriosis in a Loggerhead Sea Turtle (Caretta caretta) Stranded along the Adriatic Coast: Strain Detection and Sequencing. Animals (Basel) 2022; 12:ani12182364. [PMID: 36139224 PMCID: PMC9495059 DOI: 10.3390/ani12182364] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Although there are increasing reports on the prevalence of Listeria monocytogenes in wild species, this is the first case of listeriosis in sea turtle. An adult female Caretta caretta was rescued after being stranded alive along the coast of the Abruzzo region (Italy) in summer 2021. The turtle died in 6 days due to respiratory failure. The necropsy showed widespread organ lesions, such as yellow foci of necrosis in many organs, gastrointestinal erosions, pericarditis, and granulomatous pneumonia. Microbiological and histological analyses were performed on several organs. Listeria monocytogenes was isolated from multiple organs, indicating a case of septicaemic listeriosis, and the genome was sequenced and characterized. All the colonies analysed belonged to the same strain serogroup IVb, ST388, and CC388.
Collapse
Affiliation(s)
- Ludovica Di Renzo
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
- Centro Studi Cetacei Onlus, Centro Recupero e Riabilitazione Tartarughe Marine “L.Cagnolaro”, 65125 Pescara, PE, Italy
- Correspondence: ; Tel.: +39-08613321
| | | | - Marina Torresi
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Valeria Di Lollo
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Giovanni Di Teodoro
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Daniela Averaimo
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | | | - Federica Di Giacinto
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Chiara Profico
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
- Centro Studi Cetacei Onlus, Centro Recupero e Riabilitazione Tartarughe Marine “L.Cagnolaro”, 65125 Pescara, PE, Italy
| | - Vincenzo Olivieri
- Centro Studi Cetacei Onlus, Centro Recupero e Riabilitazione Tartarughe Marine “L.Cagnolaro”, 65125 Pescara, PE, Italy
| | - Francesco Pomilio
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Cesare Cammà
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Nicola Ferri
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| | - Gabriella Di Francesco
- Istituto Zooprofilattico Sperimentale (IZS) dell’Abruzzo e Molise “G. Caporale”, 64100 Teramo, TE, Italy
| |
Collapse
|
11
|
Septicaemic Listeriosis in a White-Faced Saki (Pithecia pithecia). J Comp Pathol 2022; 194:7-13. [DOI: 10.1016/j.jcpa.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/16/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022]
|
12
|
Kim J, Park J, Choi Z, Hong M. Structure-based molecular characterization of the LltR transcription factor from Listeria monocytogenes. Biochem Biophys Res Commun 2022; 600:142-149. [PMID: 35219103 DOI: 10.1016/j.bbrc.2022.02.067] [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] [Received: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/18/2022]
Abstract
Listeria monocytogenes is a psychrotrophic food-borne pathogenic bacterium that causes listeriosis. Due to its unusual adaptation, an ability to grow at extended temperatures ranging from 4 to 45 °C, L. monocytogenes is notoriously hard to control in food-manufacturing processes. In addition, the growing number of antibiotic-resistant L. monocytogenes strains have made listeriosis steadily refractory to clinical treatments and can lead to serious life-threatening diseases, such as sepsis and meningitis, in immunocompromised persons and neonates. Transcription factors that belong to the PadR family play a key role in bacterial survival against unfavorable environmental stresses. The LltR protein from L. monocytogenes was identified as a PadR-type transcription factor and was shown to be required for bacterial growth adaptation at low temperatures. Despite the functional significance of LltR in listeria survival and pathogenesis, our molecular understanding of the LltR-mediated transcriptional regulation is highly limited. Here, we report the crystal structure of LltR and reveal the operator DNA recognition mechanism used by LltR. LltR dimerizes into an isosceles triangle-like shape and requires a winged helix-turn-helix motif for dsDNA recognition. Indeed, LltR and putative operator dsDNA binding was observed and suggests a transcriptional repression of the llfR-lmo0600-lmo0601 operon by direct interaction between the LltR transcription factor and its promoter region. Structure-based comparative and mutational analyses showed that LltR interacts with dsDNA via a unique strategy that combines both LltR-specific and PadR family-common mechanisms.
Collapse
Affiliation(s)
- Junghun Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Jaewan Park
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Zion Choi
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Minsun Hong
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| |
Collapse
|
13
|
The Bactericidal Efficacy and the Mechanism of Action of Slightly Acidic Electrolyzed Water on Listeria monocytogenes' Survival. Foods 2021; 10:foods10112671. [PMID: 34828952 PMCID: PMC8621911 DOI: 10.3390/foods10112671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
In the present work, the bactericidal efficacy and mechanism of slightly acidic electrolyzed water (SAEW) on L. monocytogenes were evaluated. The results showed that the strains of L. monocytogenes were killed completely within 30 s by SAEW whose available chlorine concentration (ACC) was higher than 12 mg/L, and it was confirmed that ACC is the main factor affecting the disinfection efficacy of SAEW. Moreover, our results demonstrated that SAEW could destroy the cell membrane of L. monocytogenes, which was observed by SEM and FT-IR, thus resulting in the leakage of intracellular substances including electrolyte, protein and nucleic acid, and DNA damage. On the other hand, the results found that SAEW could disrupt the intracellular ROS balance of L. monocytogenes by inhibiting the antioxidant enzyme activity, thus promoting the death of L. monocytogenes. In conclusion, the bactericidal mechanism of SAEW on L. monocytogenes was explained from two aspects including the damage of the cell membrane and the breaking of ROS balance.
Collapse
|
14
|
Shi D, Anwar TM, Pan H, Chai W, Xu S, Yue M. Genomic Determinants of Pathogenicity and Antimicrobial Resistance for 60 Global Listeria monocytogenes Isolates Responsible for Invasive Infections. Front Cell Infect Microbiol 2021; 11:718840. [PMID: 34778102 PMCID: PMC8579135 DOI: 10.3389/fcimb.2021.718840] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Listeria monocytogenes remains a significant public health threat, causing invasive listeriosis manifested as septicemia, meningitis, and abortion, with up to 30% of cases having a fatal outcome. Tracking the spread of invasive listeriosis requires an updated knowledge for virulence factors (VFs) and antimicrobial resistance features, which is an essential step toward its clinical diagnosis and treatment. Taking advantage of high-throughput genomic sequencing, we proposed that the differential genes based on the pathogenomic composition could be used to evaluate clinical observations and therapeutic options for listeriosis. Here, we performed the comparative genomic analysis of 60 strains from five continents with a diverse range of sources, representing serotypes 1/2a, 1/2b, 1/2c, and 4b, comprising lineage I and lineage II and including 13 newly contributed Chinese isolates from clinical cases. These strains were associated with globally distributed clonal groups linked with confirmed foodborne listeriosis outbreak and sporadic cases. We found that L. monocytogenes strains from clonal complex (CC) CC8, CC7, CC9, and CC415 carried most of the adherence and invasive genes. Conversely, CC1, CC2, CC4, and CC6 have the least number of adherence and invasive genes. Additionally, Listeria pathogenicity island-1 (LIPI-1), LIPI-2, intracellular survival, surface anchoring, and bile salt resistance genes were detected in all isolates. Importantly, LIPI-3 genes were harbored in CC3, CC224, and ST619 of the Chinese isolates and in CC1, CC4, and CC6 of other worldwide isolates. Notably, Chinese isolates belonging to CC14 carried antibiotic resistance genes (ARGs) against β-lactams (blaTEM-101, blaTEM-105) and macrolide (ermC-15), whereas CC7 and CC8 isolates harbored ARGs against aminoglycoside (aadA10_2, aadA6_1), which may pose a threat to therapeutic efficacy. Phylogenomic analysis showed that CC8, CC7, and CC5 of Chinese isolates, CC8 (Swiss and Italian isolates), and CC5 and CC7 (Canadian isolates) are closely clustered together and belonged to the same CC. Additionally, CC381 and CC29 of Chinese isolates shared the same genomic pattern as CC26 of Swiss isolate and CC37 of Canadian isolate, respectively, indicating strong phylogenomic relation between these isolates. Collectively, this study highlights considerable clonal diversity with well-recognized virulence and antimicrobial-resistant determinants among Chinese and worldwide isolates that stress to design improved strategies for clinical therapies.
Collapse
Affiliation(s)
- Dawei Shi
- Division II of In Vitro Diagnostics for Infectious Diseases, Institute for In Vitro Diagnostics Control, National Institutes for Food and Drug Control, Beijing, China
| | - Tanveer Muhammad Anwar
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Hang Pan
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Wenqin Chai
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Sihong Xu
- Division II of In Vitro Diagnostics for Infectious Diseases, Institute for In Vitro Diagnostics Control, National Institutes for Food and Drug Control, Beijing, China
| | - Min Yue
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
| |
Collapse
|
15
|
Shahid AD, Lu Y, Iqbal MA, Lin L, Huang S, Jiang X, Chen S. Listeria monocytogenes crosses blood brain barrier through Rho GTPases induced migration of macrophages and inflammatory interleukin expression. Microb Pathog 2021; 159:105143. [PMID: 34400281 DOI: 10.1016/j.micpath.2021.105143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Listeria monocytogenes crossing the blood-brain barrier in the form of "Trojan Horse" is of great significance for the establishment of bacterial encephalitis and meningitis. Induction of cell migration and crossing the blood-brain barrier is very important to understand the Listeria pathogenesis. The Rho GTPases family is considered a key factor in regulating cell migration. This study was designed to investigate the expression of Rho GTPases and their effect on the behavior of cell migration and the stimulation of immune factors. Selective Rho GTPases were investigated by real-time PCR and Western blot. Among these, the expression of RhoA was significantly increased following the infection of Listeria monocytogenes in macrophages. Further, we found that RhoA improves the migration of macrophages and expression of IL-1β, IL-6, and TNF-α. The expression of IL-1β, IL-6 and TNF-α possibly facilitates the migration and adhesion of macrophages to cross the blood-brain barrier. This study provides preliminary ground to investigate the detailed mechanism of Listeria monocytogenes crossing the blood-brain barrier.
Collapse
Affiliation(s)
| | - Ye Lu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China; Department of Clinical Laboratory, Yixing People's Hospital, Affiliated Jiangsu University, Wuxi, 214200, China
| | | | - Lin Lin
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Shuang Huang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xugan Jiang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Shengxia Chen
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
16
|
Li M, Carpenter CE, Broadbent JR. Organic Acid Exposure Enhances Virulence in Some Listeria monocytogenes Strains Using the Galleria mellonella Infection Model. Front Microbiol 2021; 12:675241. [PMID: 34295317 PMCID: PMC8290484 DOI: 10.3389/fmicb.2021.675241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023] Open
Abstract
Prior research has suggested that the use of organic acids in the food industry may unintentionally enhance pathogenicity of Listeria monocytogenes strain N1-227 and R2-499. This study explored the connection between habituation to L-lactic acid or acetic acid and virulence in L. monocytogenes strains N1-227 and R2-499 using selected gene expression analysis and the in vivo Galleria mellonella wax worm model for infection. Expression of transcription factors (sigB and prfA) and genes related to acid resistance (gadD2, gadD3, and arcA) and bile resistance (bsh and bilE) or to virulence (inlA, inlB, hly, plcA, plcB, uhpT, and actA) was investigated by quantitative real-time PCR (qRT-PCR), while in vivo virulence was assessed by following the lethal time to 50% population mortality (LT50) of G. mellonella larvae after injection of untreated and habituated L. monocytogenes. Twenty minutes of habituation to the organic acids at pH 6.0 significantly increased expression of key acid and bile stress response genes in both strains, while expression of virulence genes was strain-dependent. The expression of transcription factor sigB was strain-dependent and there was no significant change in the expression of transcription factor prfA in both strains. Habituation to acid increased virulence of both strains as evidenced by decreased LT50 of G. mellonella larvae injected with Listeria habituated to either acid. In summary, habituation of both L. monocytogenes strains to organic acids up-regulated expression of several stress and virulence genes and concurrently increased virulence as measured using the G. mellonella model.
Collapse
Affiliation(s)
- Minghao Li
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Charles E Carpenter
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Jeff R Broadbent
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
17
|
Zhou A, Wang L, Zhang J, Yang X, Ou Z, Zhao L. Survival of viable but nonculturable Cronobacter sakazakii in macrophages contributes to infections. Microb Pathog 2021; 158:105064. [PMID: 34171399 DOI: 10.1016/j.micpath.2021.105064] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/09/2021] [Accepted: 06/16/2021] [Indexed: 02/04/2023]
Abstract
Cronobacter sakazakii (C. sakazakii), a pathogen that exists in dry and low-moisture environments, such as powder infant formula (PIF), can enter a viable but nonculturable (VBNC) state under harsh conditions, which enables it to escape traditional detection methods and thus poses a potential public health risk. This study aimed at assessing the virulent nature of VBNC C. sakazakii. Our results showed that VBNC C. sakazakii induced intestinal inflammation in neonatal rats. However, the degree of inflammation was significantly lower than that of culturable bacteria due to decreasing endotoxin production, motility, adhesion, and invasion ability in the VBNC state. From the perspective of bacterial translocation, the numbers of C. sakazakii in the blood, liver, and spleen of rats treated with VBNC cells were in the same order of magnitude as those treated with its culturable counterpart and may lead to the same degree of bacteremia. According to the macrophage survival assays, the survival rate of VBNC C. sakazakii within macrophages was 4.7 times higher than that of culturable cells. Based on these findings, we hypothesize that VBNC C. sakazakii evaded the host immune defense system, penetrated the tissue barrier, and translocated to the bloodstream, liver, and spleen through macrophages. Thus, our study reveals that VBNC C. sakazakii could be a potential risk for infants' health.
Collapse
Affiliation(s)
- Aidi Zhou
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, PR China
| | - Li Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China.
| | - Jingfeng Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, PR China
| | - Xiaoqing Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China
| | - Zhihua Ou
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China
| | - Lichao Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Wu Shan Road 483, Guangzhou 510642, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, PR China.
| |
Collapse
|
18
|
Transcriptome Analysis of Listeria monocytogenes Exposed to Beef Fat Reveals Antimicrobial and Pathogenicity Attenuation Mechanisms. Appl Environ Microbiol 2021; 87:AEM.03027-20. [PMID: 33608290 DOI: 10.1128/aem.03027-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Listeria monocytogenes is a deadly intracellular pathogen mostly associated with consumption of ready-to-eat foods. This study investigated the effectiveness of total beef fat (BF-T) from flaxseed-fed cattle and its fractions enriched with monounsaturated fatty acids (BF-MUFA) and polyunsaturated fatty acids (BF-PUFA), along with commercially available long-chain fatty acids (LC-FA), as natural antimicrobials against L. monocytogenes BF-T was ineffective at concentrations up to 6 mg/ml, while L. monocytogenes was susceptible to BF-MUFA and BF-PUFA, with MICs at pH 7 of 0.33 ± 0.21 mg/ml and 0.06 ± 0.03 mg/ml, respectively. The MIC of C14:0 was significantly lower than those of C16:0 and C18:0 (P < 0.05). Fatty acids c9-C16:1, C18:2n-6, and C18:3n-3 showed stronger inhibitory activity than c9-C18:1 and conjugated C18:2, with MICs of <1 mg/ml. Furthermore, global transcriptional analysis by transcriptome sequencing (RNA-seq) was performed to characterize the response of L. monocytogenes to selected fatty acids. Functional analysis indicated that antimicrobial LC-UFA repressed the expression of genes associated with nutrient transmembrane transport, energy generation, and oxidative stress resistance. On the other hand, upregulation of ribosome assembly and translation process is possibly associated with adaptive and repair mechanisms activated in response to LC-UFA. Virulence genes and genes involved in bile, acid, and osmotic stresses were largely downregulated, and more so for c9-C16:1, C18:2n-6, and C18:3n-3, likely through interaction with the master virulence regulator PrfA and the alternative sigma factor σB IMPORTANCE Listeria monocytogenes is a bacterial pathogen known for its ability to survive and thrive under adverse environments and, as such, its control poses a significant challenge, especially with the trend of minimally processed and ready-to-eat foods. This work investigated the effectiveness of fatty acids from various sources as natural antimicrobials against L. monocytogenes and evaluated their potential role in L. monocytogenes pathogenicity modulation, using the strain ATCC 19111. The findings show that long-chain unsaturated fatty acids (LC-UFA), including unsaturated beef fat fractions from flaxseed-fed cattle, could have the potential to be used as effective antimicrobials for L. monocytogenes through controlling growth as well as virulence attenuation. This not only advances our understanding of the mode of action of LC-UFA against L. monocytogenes but also suggests the potential for use of beef fat or its fractions as natural antimicrobials for controlling foodborne pathogens.
Collapse
|
19
|
Mohan V, Cruz CD, van Vliet AHM, Pitman AR, Visnovsky SB, Rivas L, Gilpin B, Fletcher GC. Genomic diversity of Listeria monocytogenes isolates from seafood, horticulture and factory environments in New Zealand. Int J Food Microbiol 2021; 347:109166. [PMID: 33838478 DOI: 10.1016/j.ijfoodmicro.2021.109166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/28/2021] [Accepted: 03/06/2021] [Indexed: 11/28/2022]
Abstract
Listeria monocytogenes is a foodborne human pathogen that causes systemic infection, fetal-placental infection in pregnant women causing abortion and stillbirth and meningoencephalitis in elderly and immunocompromised individuals. This study aimed to analyse L. monocytogenes from different sources from New Zealand (NZ) and to compare them with international strains. We used pulsed-field gel electrophoresis (PFGE), multilocus sequence typing (MLST) and whole-genome single nucleotide polymorphisms (SNP) to study the population structure of the NZ L. monocytogenes isolates and their relationship with the international strains. The NZ isolates formed unique clusters in PFGE, MLST and whole-genome SNP comparisons compared to the international isolates for which data were available. PFGE identified 31 AscI and 29 ApaI PFGE patterns with indistinguishable pulsotypes being present in seafood, horticultural products and environmental samples. Apart from the Asc0002:Apa0002 pulsotype which was distributed across different sources, other pulsotypes were site or factory associated. Whole-genome analysis of 200 randomly selected L. monocytogenes isolates revealed that lineage II dominated the NZ L. monocytogenes populations. MLST comparison of international and NZ isolates with lineage II accounted for 89% (177 of 200) of the total L. monocytogenes population, while the international representation was 45.3% (1674 of 3473). Rarefaction analysis showed that sequence type richness was greater in NZ isolates compared to international trend, however, it should be noted that NZ isolates predominantly came from seafood, horticulture and their respective processing environments or factories, unlike international isolates where there was a good mixture of clinical, food and environmental isolates.
Collapse
Affiliation(s)
- Vathsala Mohan
- The New Zealand Institute for Plant & Food Research Limited, Auckland, New Zealand.
| | - Cristina D Cruz
- The New Zealand Institute for Plant & Food Research Limited, Auckland, New Zealand
| | - Arnoud H M van Vliet
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Daphne Jackson Road, Guildford GU2 7AL, Surrey, United Kingdom
| | - Andrew R Pitman
- The New Zealand Institute for Plant & Food Research Limited, Lincoln, New Zealand.
| | - Sandra B Visnovsky
- The New Zealand Institute for Plant & Food Research Limited, Lincoln, New Zealand
| | - Lucia Rivas
- Institute of Environmental Science and Research Limited, Christchurch, New Zealand
| | - Brent Gilpin
- Institute of Environmental Science and Research Limited, Christchurch, New Zealand
| | - Graham C Fletcher
- The New Zealand Institute for Plant & Food Research Limited, Auckland, New Zealand
| |
Collapse
|
20
|
Tran BM, Prabha H, Iyer A, O'Byrne C, Abee T, Poolman B. Measurement of Protein Mobility in Listeria monocytogenes Reveals a Unique Tolerance to Osmotic Stress and Temperature Dependence of Diffusion. Front Microbiol 2021; 12:640149. [PMID: 33679676 PMCID: PMC7925416 DOI: 10.3389/fmicb.2021.640149] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/21/2021] [Indexed: 11/18/2022] Open
Abstract
Protein mobility in the cytoplasm is essential for cellular functions, and slow diffusion may limit the rates of biochemical reactions in the living cell. Here, we determined the apparent lateral diffusion coefficient (DL) of GFP in Listeria monocytogenes as a function of osmotic stress, temperature, and media composition. We find that DL is much less affected by hyperosmotic stress in L. monocytogenes than under similar conditions in Lactococcus lactis and Escherichia coli. We find a temperature optimum for protein diffusion in L. monocytogenes at 30°C, which deviates from predicted trends from the generalized Stokes-Einstein equation under dilute conditions and suggests that the structure of the cytoplasm and macromolecular crowding vary as a function of temperature. The turgor pressure of L. monocytogenes is comparable to other Gram-positive bacteria like Bacillus subtilis and L. lactis but higher in a knockout strain lacking the stress-inducible sigma factor SigB. We discuss these findings in the context of how L. monocytogenes survives during environmental transmission and interaction with the human host.
Collapse
Affiliation(s)
- Buu Minh Tran
- Department of Biochemistry, University of Groningen, Groningen, Netherlands
| | - Haritha Prabha
- Department of Biochemistry, University of Groningen, Groningen, Netherlands
| | - Aditya Iyer
- Department of Biochemistry, University of Groningen, Groningen, Netherlands
| | - Conor O'Byrne
- School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Tjakko Abee
- Laboratory of Food Microbiology, Wageningen University Research, Wageningen, Netherlands
| | - Bert Poolman
- Department of Biochemistry, University of Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
Karam J, Méresse S, Kremer L, Daher W. The roles of tetraspanins in bacterial infections. Cell Microbiol 2020; 22:e13260. [PMID: 32902857 DOI: 10.1111/cmi.13260] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Tetraspanins, a wide family composed of 33 transmembrane proteins, are associated with different types of proteins through which they arbitrate important cellular processes such as fusion, adhesion, invasion, tissue differentiation and immunological responses. Tetraspanins share a comparable structural design, which consists of four hydrophobic transmembrane domains with cytoplasmic and extracellular loops. They cooperate with different proteins, including other tetraspanins, receptors or signalling proteins to compose functional complexes at the cell surface, designated tetraspanin-enriched microdomains (TEM). Increasing evidences establish that tetraspanins are exploited by numerous intracellular pathogens as a doorway for entering and replicating within human cells. Although previous surveys focused mainly on viruses and parasites, it is now becoming clear that bacteria interact with tetraspanins, using TEM as a "gateway" to infection. In this review, we examine the biological functions of tetraspanins that are relevant to bacterial infective procedures and consider the available data that reveal how different bacteria benefit from host cell tetraspanins in infection and in the pathogenesis of diseases. We will also emphasise the stimulating potentials of targeting tetraspanins for preventing bacterial infectious diseases, using specific neutralising antibodies or anti-adhesion peptide-based therapies. Such innovative therapeutic opportunities may deliver alternatives for fighting difficult-to-manage and drug-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Jona Karam
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | | | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| | - Wassim Daher
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| |
Collapse
|
22
|
Sharma R, Gal L, Garmyn D, Bisaria VS, Sharma S, Piveteau P. Evidence of Biocontrol Activity of Bioinoculants Against a Human Pathogen, Listeria monocytogenes. Front Microbiol 2020; 11:350. [PMID: 32218775 PMCID: PMC7078112 DOI: 10.3389/fmicb.2020.00350] [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: 12/05/2019] [Accepted: 02/17/2020] [Indexed: 11/13/2022] Open
Abstract
Due to rhizodeposits and various microbial interactions, the rhizosphere is an extremely dynamic system, which provides a conductive niche not only for bacteria beneficial to plants but also for those that might pose a potential threat to humans. The importance of bioinoculants as biocontrol agents to combat phytopathogens has been widely recognized. However, little information exists with respect to their role in inhibiting human pathogens in the rhizosphere. The present study is an attempt to understand the impact of an established bacterial consortium, Azotobacter chroococcum, Bacillus megaterium, and Pseudomonas fluorescens, on the survivability of Listeria monocytogenes in the rhizosphere of Cajanus cajan and Festuca arundinacea. An experiment conducted in Hoagland's medium in the presence of C. cajan demonstrated that the presence of bioinoculants impaired growth of L. monocytogenes compared to that observed in their absence. On the other hand, in the presence of F. arundinacea, no significant differences were observed in the population dynamics of L. monocytogenes in the presence or absence of the bioinoculants. Agar plate assay through cross streak method revealed the inhibition of L. monocytogenes by bioinoculants. Potential bioactive compounds were identified by ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). These results suggest that agricultural amendments can act as protective agents against human pathogens while enforcing plant growth promotion.
Collapse
Affiliation(s)
- Richa Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Laurent Gal
- Agroécologie, AgroSup Dijon, Institut National de la Recherche Agronomique, Université Bourgogne – Franche-Comté, Dijon, France
| | - Dominique Garmyn
- Agroécologie, AgroSup Dijon, Institut National de la Recherche Agronomique, Université Bourgogne – Franche-Comté, Dijon, France
| | - V. S. Bisaria
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Shilpi Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Pascal Piveteau
- Agroécologie, AgroSup Dijon, Institut National de la Recherche Agronomique, Université Bourgogne – Franche-Comté, Dijon, France
| |
Collapse
|
23
|
Wang G, Zhao H, Zheng B, Li D, Yuan Y, Han Q, Tian Z, Zhang J. TLR2 Promotes Monocyte/Macrophage Recruitment Into the Liver and Microabscess Formation to Limit the Spread of Listeria Monocytogenes. Front Immunol 2019; 10:1388. [PMID: 31297109 PMCID: PMC6607897 DOI: 10.3389/fimmu.2019.01388] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 06/03/2019] [Indexed: 12/24/2022] Open
Abstract
TLR2 signaling plays a critical protective role against acute Listeria monocytogenes (Lm) infection by up-regulating inflammatory cytokines and promoting macrophage antimicrobial capabilities. However, the underlying mechanism by which TLR2 regulates hepatic macrophage-mediated anti-Lm immune responses remains poorly understood. In this study, we found that both the absolute number and proportion of monocyte/macrophage (Mo/MΦ) in the liver and spleen of Tlr2 -/- mice were significantly lower compared to wild type mice. Changes in TLR2 signaling in both hepatocytes and Mo/MΦs were associated with the infiltration of Mo/MΦs in response to Lm-infection. Analyses by proteome profiler array and ELISA revealed that hepatocytes recruited Mo/MΦs via TLR2-dependent secretion of CCL2 and CXCL1, which was confirmed by receptor blocking and exogenous chemokine administration. Importantly, we found that TLR2 contributed to macrophage mobility in the liver through a TLR2/NO/F-actin pathway, facilitating the formation of macrophage-associated hepatic microabscesses. Moreover, TLR2 activation induced the expression of several PRRs on hepatic macrophages associated with the recognition of Lm and augmented macrophage bacterial clearance activity. Our findings provide insight into the intrinsic mechanisms of TLR2-induced Mo/MΦ migration and mobility, as well as the interaction between macrophages and hepatocytes in resistance to Lm infection.
Collapse
Affiliation(s)
- Guan Wang
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Huajun Zhao
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Bingqing Zheng
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Dongxuan Li
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Qiuju Han
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhigang Tian
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
24
|
Lee C, Kim MI, Park J, Hong M. Structure-based molecular characterization and regulatory mechanism of the LftR transcription factor from Listeria monocytogenes: Conformational flexibilities and a ligand-induced regulatory mechanism. PLoS One 2019; 14:e0215017. [PMID: 30970033 PMCID: PMC6457526 DOI: 10.1371/journal.pone.0215017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/25/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis and can lead to serious clinical problems, such as sepsis and meningitis, in immunocompromised patients and neonates. Due to a growing number of antibiotic-resistant L. monocytogenes strains, listeriosis can steadily become refractory to antibiotic treatment. To develop novel therapeutics against listeriosis, the drug resistance mechanism of L. monocytogenes needs to be determined. The transcription factor LftR from L. monocytogenes regulates the expression of a putative multidrug resistance transporter, LieAB, and belongs to the PadR-2 subfamily of the PadR family. Despite the functional significance of LftR, our molecular understanding of the transcriptional regulatory mechanism for LftR and even for the PadR-2 subfamily is highly limited. Here, we report the crystal structure of LftR, which forms a dimer and protrudes two winged helix-turn-helix motifs for DNA recognition. Structure-based mutational and comparative analyses showed that LftR interacts with operator DNA through a LftR-specific mode as well as a common mechanism used by the PadR family. Moreover, the LftR dimer harbors one intersubunit cavity in the center of the dimeric structure as a putative ligand-binding site. Finally, conformational flexibilities in the LftR dimer and in the cavity suggest that a ligand-induced regulatory mechanism would be used by the LftR transcription factor.
Collapse
Affiliation(s)
- Choongdeok Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Meong Il Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Jaewan Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Minsun Hong
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
- * E-mail:
| |
Collapse
|
25
|
Rao DM, Phan DT, Choo MJ, Owen AL, Perraud AL, Gally F. Mice Lacking Fatty Acid-Binding Protein 5 Are Resistant to Listeria monocytogenes. J Innate Immun 2019; 11:469-480. [PMID: 30884482 DOI: 10.1159/000496405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/18/2018] [Indexed: 02/01/2023] Open
Abstract
To investigate the role of fatty acid-binding protein 5 (FABP5) in infectious diseases, FABP5-deficient mice were challenged with Listeria monocytogenes, a facultative intracellular bacterial pathogen. Interestingly, FABP5-deficient animals were able to clear the infection within 3 days whereas control wild-type (WT) animals showed comparatively higher bacterial burdens in the liver and spleen. Sections of infected tissues showed an increase in inflammatory foci in WT mice compared to FABP5-deficient mice. FABP5-deficient mice had lower circulating inflammatory cytokines and increased inducible nitric oxide synthase production. FABP5-deficient mouse bone marrow-derived macrophages produced higher levels of nitrite anion than their WT counterparts in response to various stimuli. Additionally, in contrast to FABP5-/- mice, transgenic mice overexpressing FABP5 in myeloid cells (LysM-Cre driven) showed decreased survival rates and increased bacterial burden and inflammatory cytokines. Overall, these findings suggest that increased FABP5 levels correlate with a higher L. monocytogenes bacterial burden and elevated subsequent inflammation.
Collapse
Affiliation(s)
- Deviyani M Rao
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Della T Phan
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Michelle J Choo
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Amie L Owen
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Anne-Laure Perraud
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Fabienne Gally
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA,
| |
Collapse
|
26
|
Pinheiro J, Lisboa J, Pombinho R, Carvalho F, Carreaux A, Brito C, Pöntinen A, Korkeala H, dos Santos NM, Morais-Cabral JH, Sousa S, Cabanes D. MouR controls the expression of the Listeria monocytogenes Agr system and mediates virulence. Nucleic Acids Res 2018; 46:9338-9352. [PMID: 30011022 PMCID: PMC6182135 DOI: 10.1093/nar/gky624] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes (Lm) causes invasive infection in susceptible animals and humans. To survive and proliferate within hosts, this facultative intracellular pathogen tightly coordinates the expression of a complex regulatory network that controls the expression of virulence factors. Here, we identified and characterized MouR, a novel virulence regulator of Lm. Through RNA-seq transcriptomic analysis, we determined the MouR regulon and demonstrated how MouR positively controls the expression of the Agr quorum sensing system (agrBDCA) of Lm. The MouR three-dimensional structure revealed a dimeric DNA-binding transcription factor belonging to the VanR class of the GntR superfamily of regulatory proteins. We also showed that by directly binding to the agr promoter region, MouR ultimately modulates chitinase activity and biofilm formation. Importantly, we demonstrated by in vitro cell invasion assays and in vivo mice infections the role of MouR in Lm virulence.
Collapse
Affiliation(s)
- Jorge Pinheiro
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto 4200-135, Portugal
| | - Johnny Lisboa
- Group of Fish Immunology & Vaccinology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
| | - Rita Pombinho
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto 4200-135, Portugal
| | - Filipe Carvalho
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto 4200-135, Portugal
| | - Alexis Carreaux
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
- SDV - UFR Sciences Du Vivant: Université Paris Diderot-Paris 7, Paris 75013, France
| | - Cláudia Brito
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto 4200-135, Portugal
| | - Anna Pöntinen
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Hannu Korkeala
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Nuno M S dos Santos
- Group of Fish Immunology & Vaccinology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
| | - João H Morais-Cabral
- Group of Structural Biochemistry, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, IBMC – Institute for Molecular and Cell Biology; i3S – Institute for Research and Innovation in Health, Porto 4200-135, Portugal
| |
Collapse
|
27
|
Rahman A, Munther D, Fazil A, Smith B, Wu J. With-in host dynamics of L. monocytogenes and thresholds for distinct infection scenarios. J Theor Biol 2018; 454:80-90. [PMID: 29842866 DOI: 10.1016/j.jtbi.2018.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/21/2018] [Accepted: 05/24/2018] [Indexed: 10/16/2022]
Abstract
The case fatality and illness rates associated with L. monocytogenes continue to pose a serious public health burden despite the significant efforts and control protocol administered by private and public sectors. Due to the advance in surveillance and improvement in detection methodology, the knowledge of sources, transmission routes, growth potential in food process units and storage, effect of pH and temperature are well understood. However, the with-in host growth and transmission mechanisms of L. monocytogenes, particularly within the human host, remain unclear, largely due to the limited access to scientific experimentation on the human population. In order to provide insight towards the human immune response to the infection caused by L. monocytogenes, we develop a with-in host mathematical model. The model explains, in terms of biological parameters, the states of asymptomatic infection, mild infection and systemic infection leading to listeriosis. The activation and proliferation of T-cells are found to be critical for the susceptibility of the infection. Utilizing stability analysis and numerical simulation, the ranges of the critical parameters relative to infection states are established. Bifurcation analysis shows the impact of the differences of these parameters on the dynamics of the model. Finally, we present model applications in regards to predicting the risk potential of listeriosis relative to the susceptible human population.
Collapse
Affiliation(s)
- Ashrafur Rahman
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, Ontario M3J 1P3, Canada.
| | - Daniel Munther
- Department of Mathematics, Cleveland State University, Cleveland, OH 44115, USA
| | - Aamir Fazil
- National Microbiology Laboratory, Public Health Agency of Canada Guelph, Ontario N1G 5B2, Canada
| | - Ben Smith
- National Microbiology Laboratory, Public Health Agency of Canada Guelph, Ontario N1G 5B2, Canada
| | - Jianhong Wu
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
28
|
Rychli K, Stessl B, Szakmary-Brändle K, Strauß A, Wagner M, Schoder D. Listeria monocytogenes Isolated from Illegally Imported Food Products into the European Union Harbor Different Virulence Factor Variants. Genes (Basel) 2018; 9:E428. [PMID: 30142903 PMCID: PMC6162745 DOI: 10.3390/genes9090428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
Abstract
Unregulated international flow of foods poses a danger to human health, as it may be contaminated with pathogens. Recent studies have investigated neglected routes of pathogen transmission and reported the occurrence of Listeria monocytogenes in food illegally imported into the European Union (EU), either confiscated at four international airports or sold illegally on the Romanian black market. In this study we investigated the genotype diversity and the amino acid sequence variability of three main virulence factors of 57 L. monocytogenes isolates. These isolates were derived from 1474 food samples illegally imported into the EU and originated from 17 different countries. Multilocus sequence typing revealed 16 different sequence types (STs) indicating moderate genotype diversity. The most prevalent STs were ST2, ST9, and ST121. The pulsed-field gel electrophoresis (PFGE) analysis resulted in 34 unique pulsotypes. PFGE types assigned to the most prevalent STs (ST2, ST9, and ST121) were highly related in their genetic fingerprint. Internalin A (InlA) was present in 20 variants, including six truncated InlA variants, all harbored by isolates of ST9 and ST121. We detected eight ST-specific listeriolysin O (LLO) variants, and among them, one truncated form. The actin-assembly-inducing protein ActA was present in 15 different ST-specific variants, including four ActA variants with an internal truncation. In conclusion, this study shows that L. monocytogenes, isolated from illegally imported food, have moderate genotype diversity, but diverse virulence factors variants, mainly of InlA.
Collapse
Affiliation(s)
- Kathrin Rychli
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Beatrix Stessl
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Kati Szakmary-Brändle
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Anja Strauß
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Martin Wagner
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Dagmar Schoder
- Institute of Milk Hygiene, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| |
Collapse
|
29
|
Rahman A, Munther D, Fazil A, Smith B, Wu J. Advancing risk assessment: mechanistic dose-response modelling of Listeria monocytogenes infection in human populations. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180343. [PMID: 30225020 PMCID: PMC6124125 DOI: 10.1098/rsos.180343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/25/2018] [Indexed: 05/16/2023]
Abstract
The utility of characterizing the effects of strain variation and individual/subgroup susceptibility on dose-response outcomes has motivated the search for new approaches beyond the popular use of the exponential dose-response model for listeriosis. While descriptive models can account for such variation, they have limited power to extrapolate beyond the details of particular outbreaks. By contrast, this study exhibits dose-response relationships from a mechanistic basis, quantifying key biological factors involved in pathogen-host dynamics. An efficient computational algorithm and geometric interpretation of the infection pathway are developed to connect dose-response relationships with the underlying bistable dynamics of the model. Relying on in vitro experiments as well as outbreak data, we estimate plausible parameters for the human context. Despite the presence of uncertainty in such parameters, sensitivity analysis reveals that the host response is most influenced by the pathogen-immune system interaction. In particular, we show how variation in this interaction across a subgroup of the population dictates the shape of dose-response curves. Finally, in terms of future experimentation, our model results provide guidelines and highlight vital aspects of the interplay between immune cells and particular strains of Listeria monocytogenes that should be examined.
Collapse
Affiliation(s)
- Ashrafur Rahman
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, Ontario, CanadaM3J 1P3
| | - Daniel Munther
- Department of Mathematics, Cleveland State University, Cleveland, OH 44115, USA
| | - Aamir Fazil
- National Microbiology Laboratory, Public Health Agency of Canada, Guelph, Ontario, CanadaN1G 5B2
| | - Ben Smith
- National Microbiology Laboratory, Public Health Agency of Canada, Guelph, Ontario, CanadaN1G 5B2
| | - Jianhong Wu
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, Ontario, CanadaM3J 1P3
| |
Collapse
|
30
|
Carvalho F, Sousa S, Cabanes D. l-Rhamnosylation of wall teichoic acids promotes efficient surface association of Listeria monocytogenes virulence factors InlB and Ami through interaction with GW domains. Environ Microbiol 2018; 20:3941-3951. [PMID: 29984543 DOI: 10.1111/1462-2920.14351] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 07/02/2018] [Indexed: 11/29/2022]
Abstract
Wall teichoic acids (WTAs) are important surface glycopolymers involved in various physiological processes occurring in the Gram-positive cell envelope. We previously showed that the decoration of Listeria monocytogenes (Lm) WTAs with l-rhamnose conferred resistance against antimicrobial peptides. Here, we show that WTA l-rhamnosylation also contributes to physiological levels of autolysis in Lm through a mechanism that requires efficient association of Ami, a virulence-promoting autolysin belonging to the GW protein family, to the bacterial cell surface. Importantly, WTA l-rhamnosylation also controls the surface association of another GW protein, the invasin internalin B (InlB), that promotes Lm invasion of host cells. Whereas WTA N-acetylglucosaminylation is not a prerequisite for GW protein surface association, lipoteichoic acids appear to also play a role in the surface anchoring of InlB. Strikingly, while the GW domains of Ami, InlB and Auto (another autolysin contributing to cell invasion and virulence) are sufficient to mediate surface association, this is not the case for the GW domains of the remaining six uncharacterized Lm GW proteins. Overall, we reveal WTA l-rhamnosylation as a bacterial surface modification mechanism that contributes to Lm physiology and pathogenesis by controlling the surface association of GW proteins involved in autolysis and infection.
Collapse
Affiliation(s)
- Filipe Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sandra Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Didier Cabanes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
Wang G, Lin A, Han Q, Zhao H, Tian Z, Zhang J. IFN-γ protects from apoptotic neutrophil-mediated tissue injury during acute Listeria monocytogenes infection. Eur J Immunol 2018; 48:1470-1480. [PMID: 29935120 DOI: 10.1002/eji.201847491] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/24/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023]
Abstract
Listeria monocytogenes (LM) is a foodborne Gram-positive intracellular pathogen that can cause listeriosis in humans and animals. Although phagocytes are known to be involved in the response to this infection, the role of neutrophils is not entirely clear. Here, we have demonstrated that soon after LM infection, a large number of IFN-γ-producing neutrophils quickly accumulated in the spleen, blood, and peritoneal cavity. Both in vivo and in vitro experiments demonstrated that neutrophils were an important source of IFN-γ. IFN-γ played a critical protective role against acute LM infection, as demonstrated by the poor survival of Ifng-/- mice. Moreover, IFN-γ promoted bacterial clearance by the neutrophils, thereby inhibiting LM-induced neutrophil apoptosis and spleen damage. In addition to this, IFN-γ could effectively drive macrophage-mediated phagocytosis of apoptotic neutrophils, which was accompanied with TGF-β secretion and was involved in protection against tissue injury. Importantly, by phagocytizing apoptotic neutrophils, macrophages obtained myeloperoxidase, an important bactericidal molecule only produced by neutrophils, which further promoted the antibacterial activity of macrophages. These findings demonstrate that neutrophils are an important source of IFN-γ at the early stage of LM infection, which is characterized by both LM elimination and tissue-protective effects.
Collapse
Affiliation(s)
- Guan Wang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Ang Lin
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhigang Tian
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
32
|
Shrinet J, Bhavesh NS, Sunil S. Understanding Oxidative Stress in Aedes during Chikungunya and Dengue Virus Infections Using Integromics Analysis. Viruses 2018; 10:v10060314. [PMID: 29890729 PMCID: PMC6024870 DOI: 10.3390/v10060314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Arboviral infection causes dysregulation of cascade of events involving numerous biomolecules affecting fitness of mosquito to combat virus. In response of the viral infection mosquito’s defense mechanism get initiated. Oxidative stress is among the first host responses triggered by the vector. Significant number of information is available showing changes in the transcripts and/or proteins upon Chikungunya virus and Dengue virus mono-infections and as co-infections. In the present study, we collected different -omics data available in the public database along with the data generated in our laboratory related to mono-infections or co-infections of these viruses. We analyzed the data and classified them into their respective pathways to study the role of oxidative stress in combating arboviral infection in Aedes mosquito. The analysis revealed that the oxidative stress related pathways functions in harmonized manner.
Collapse
Affiliation(s)
- Jatin Shrinet
- Vector Borne Diseases, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Neel Sarovar Bhavesh
- Transcriptional Regulation, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Sujatha Sunil
- Vector Borne Diseases, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
33
|
Zilelidou EA, Skandamis PN. Growth, detection and virulence of Listeria monocytogenes in the presence of other microorganisms: microbial interactions from species to strain level. Int J Food Microbiol 2018; 277:10-25. [PMID: 29677551 DOI: 10.1016/j.ijfoodmicro.2018.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 01/28/2023]
Abstract
Like with all food microorganisms, many basic aspects of L. monocytogenes life are likely to be influenced by its interactions with bacteria living in close proximity. This pathogenic bacterium is a major concern both for the food industry and health organizations since it is ubiquitous and able to withstand harsh environmental conditions. Due to the ubiquity of Listeria monocytogenes, various strains may contaminate foods at different stages of the supply chain. Consequently, simultaneous exposure of consumers to multiple strains is also possible. In this context even strain-to-strain interactions of L. monocytogenes play a significant role in fundamental processes for the life of the pathogen, such as growth or virulence, and subsequently compromise food safety, affect the evolution of a potential infection, or even introduce bias in the detection by classical enrichment techniques. This article summarizes the impact of microbial interactions on the growth and detection of L. monocytogenes primarily in foods and food-associated environments. Furthermore it provides an overview of L. monocytogenes virulence in the presence of other microorganisms.
Collapse
Affiliation(s)
- Evangelia A Zilelidou
- Agricultural University of Athens, Department of Food Science and Human Nutrition, Laboratory of Food Quality Control and Hygiene, Iera odos 75, 11855 Athens, Greece
| | - Panagiotis N Skandamis
- Agricultural University of Athens, Department of Food Science and Human Nutrition, Laboratory of Food Quality Control and Hygiene, Iera odos 75, 11855 Athens, Greece.
| |
Collapse
|
34
|
Rismondo J, Wamp S, Aldridge C, Vollmer W, Halbedel S. Stimulation of PgdA-dependent peptidoglycanN-deacetylation by GpsB-PBP A1 inListeria monocytogenes. Mol Microbiol 2017; 107:472-487. [DOI: 10.1111/mmi.13893] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 11/29/2017] [Accepted: 12/03/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Jeanine Rismondo
- FG11 Division of Enteropathogenic bacteria and Legionella; Robert Koch Institute, Burgstrasse 37; Wernigerode 38855 Germany
| | - Sabrina Wamp
- FG11 Division of Enteropathogenic bacteria and Legionella; Robert Koch Institute, Burgstrasse 37; Wernigerode 38855 Germany
| | - Christine Aldridge
- Institute for Cell and Molecular Biosciences, The Centre for Bacterial Cell Biology; Newcastle University; Newcastle upon Tyne NE2 4AX UK
| | - Waldemar Vollmer
- Institute for Cell and Molecular Biosciences, The Centre for Bacterial Cell Biology; Newcastle University; Newcastle upon Tyne NE2 4AX UK
| | - Sven Halbedel
- FG11 Division of Enteropathogenic bacteria and Legionella; Robert Koch Institute, Burgstrasse 37; Wernigerode 38855 Germany
| |
Collapse
|
35
|
Schardt J, Jones G, Müller-Herbst S, Schauer K, D'Orazio SEF, Fuchs TM. Comparison between Listeria sensu stricto and Listeria sensu lato strains identifies novel determinants involved in infection. Sci Rep 2017; 7:17821. [PMID: 29259308 PMCID: PMC5736727 DOI: 10.1038/s41598-017-17570-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/28/2017] [Indexed: 01/01/2023] Open
Abstract
The human pathogen L. monocytogenes and the animal pathogen L. ivanovii, together with four other species isolated from symptom-free animals, form the “Listeria sensu stricto” clade. The members of the second clade, “Listeria sensu lato”, are believed to be solely environmental bacteria without the ability to colonize mammalian hosts. To identify novel determinants that contribute to infection by L. monocytogenes, the causative agent of the foodborne disease listeriosis, we performed a genome comparison of the two clades and found 151 candidate genes that are conserved in the Listeria sensu stricto species. Two factors were investigated further in vitro and in vivo. A mutant lacking an ATP-binding cassette transporter exhibited defective adhesion and invasion of human Caco-2 cells. Using a mouse model of foodborne L. monocytogenes infection, a reduced number of the mutant strain compared to the parental strain was observed in the small intestine and the liver. Another mutant with a defective 1,2-propanediol degradation pathway showed reduced persistence in the stool of infected mice, suggesting a role of 1,2-propanediol as a carbon and energy source of listeriae during infection. These findings reveal the relevance of novel factors for the colonization process of L. monocytogenes.
Collapse
Affiliation(s)
- Jakob Schardt
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Grant Jones
- Department of Microbiology, Immunology, & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Stefanie Müller-Herbst
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Kristina Schauer
- Lehrstuhl für Hygiene und Technologie der Milch, Tiermedizinische Fakultät, Ludwig-Maximilians-Universität München, Schönleutner Str. 8, 85764, Oberschleißheim, Germany
| | - Sarah E F D'Orazio
- Department of Microbiology, Immunology, & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Thilo M Fuchs
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany. .,Friedrich-Loeffler-Institut, Institut für Molekulare Pathogenese, Naumburger Str. 96a, 07743, Jena, Germany.
| |
Collapse
|
36
|
Chen K, Bao Z, Gong W, Tang P, Yoshimura T, Wang JM. Regulation of inflammation by members of the formyl-peptide receptor family. J Autoimmun 2017; 85:64-77. [PMID: 28689639 PMCID: PMC5705339 DOI: 10.1016/j.jaut.2017.06.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with a variety of diseases. The hallmark of inflammation is leukocyte infiltration at disease sites in response to pathogen- or damage-associated chemotactic molecular patterns (PAMPs and MAMPs), which are recognized by a superfamily of seven transmembrane, Gi-protein-coupled receptors (GPCRs) on cell surface. Chemotactic GPCRs are composed of two major subfamilies: the classical GPCRs and chemokine GPCRs. Formyl-peptide receptors (FPRs) belong to the classical chemotactic GPCR subfamily with unique properties that are increasingly appreciated for their expression on diverse host cell types and the capacity to interact with a plethora of chemotactic PAMPs and MAMPs. Three FPRs have been identified in human: FPR1-FPR3, with putative corresponding mouse counterparts. FPR expression was initially described in myeloid cells but subsequently in many non-hematopoietic cells including cancer cells. Accumulating evidence demonstrates that FPRs possess multiple functions in addition to controlling inflammation, and participate in the processes of many pathophysiologic conditions. They are not only critical mediators of myeloid cell trafficking, but are also implicated in tissue repair, angiogenesis and protection against inflammation-associated tumorigenesis. A series recent discoveries have greatly expanded the scope of FPRs in host defense which uncovered the essential participation of FPRs in step-wise trafficking of myeloid cells including neutrophils and dendritic cells (DCs) in host responses to bacterial infection, tissue injury and wound healing. Also of great interest is the FPRs are exploited by malignant cancer cells for their growth, invasion and metastasis. In this article, we review the current understanding of FPRs concerning their expression in a vast array of cell types, their involvement in guiding leukocyte trafficking in pathophysiological conditions, and their capacity to promote the differentiation of immune cells, their participation in tumor-associated inflammation and cancer progression. The close association of FPRs with human diseases and cancer indicates their potential as targets for the development of therapeutics.
Collapse
Affiliation(s)
- Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Zhiyao Bao
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Department of Pulmonary & Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Peng Tang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| |
Collapse
|
37
|
Ur Rahman S, Stanton M, Casey PG, Spagnuolo A, Bensi G, Hill C, Francis KP, Tangney M, Gahan CGM. Development of a Click Beetle Luciferase Reporter System for Enhanced Bioluminescence Imaging of Listeria monocytogenes: Analysis in Cell Culture and Murine Infection Models. Front Microbiol 2017; 8:1797. [PMID: 29018414 PMCID: PMC5622934 DOI: 10.3389/fmicb.2017.01797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/05/2017] [Indexed: 01/22/2023] Open
Abstract
Listeria monocytogenes is a Gram-positive facultative intracellular pathogen that is widely used as a model organism for the analysis of infection biology. In this context, there is a current need to develop improved reporters for enhanced bioluminescence imaging (BLI) of the pathogen in infection models. We have developed a click beetle red luciferase (CBR-luc) based vector (pPL2CBRopt) expressing codon optimized CBR-luc under the control of a highly expressed Listerial promoter (PHELP) for L. monocytogenes and have compared this to a lux-based system expressing bacterial luciferase for BLI of the pathogen using in vitro growth experiments and in vivo models. The CBR-luc plasmid stably integrates into the L. monocytogenes chromosome and can be used to label field isolates and laboratory strains of the pathogen. Growth experiments revealed that CBR-luc labeled L. monocytogenes emits a bright signal in exponential phase that is maintained during stationary phase. In contrast, lux-labeled bacteria produced a light signal that peaked during exponential phase and was significantly reduced during stationary phase. Light from CBR-luc labeled bacteria was more efficient than the signal from lux-labeled bacteria in penetrating an artificial tissue depth assay system. A cell invasion assay using C2Bbe1 cells and a systemic murine infection model revealed that CBR-luc is suited to BLI approaches and demonstrated enhanced sensitivity relative to lux in the context of Listeria infection models. Overall, we demonstrate that this novel CBR reporter system provides efficient, red-shifted light production relative to lux and may have significant applications in the analysis of L. monocytogenes pathogenesis.
Collapse
Affiliation(s)
- Sadeeq Ur Rahman
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,College of Veterinary Sciences and Animal Husbandry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Michael Stanton
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Pat G Casey
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | | | | | - Colin Hill
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | | | - Mark Tangney
- APC Microbiome Institute, University College Cork, Cork, Ireland.,Cork Cancer Research Centre, University College Cork, Cork, Ireland.,SynBio Centre, University College Cork, Cork, Ireland
| | - Cormac G M Gahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,SynBio Centre, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
38
|
Hülskötter K, Schmidtke D, Dubicanac M, Siesenop U, Zimmermann E, Gerhauser I, Baumgärtner W, Herder V. Spontaneous listeriosis in grey mouse lemurs (Microcebus murinus), but not in Goodman’s mouse lemurs (Microcebus lehilahytsara) of the same colony. Vet Microbiol 2017; 208:94-96. [DOI: 10.1016/j.vetmic.2017.07.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 12/30/2022]
|
39
|
Sinha S, Kuo CY, Ho JK, White PJ, Jazayeri JA, Pouton CW. A suicidal strain of Listeria monocytogenes is effective as a DNA vaccine delivery system for oral administration. Vaccine 2017; 35:5115-5122. [PMID: 28822642 DOI: 10.1016/j.vaccine.2017.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 07/10/2017] [Accepted: 08/07/2017] [Indexed: 01/24/2023]
Abstract
In this study we determined the in vivo activity of model ovalbumin vaccines delivered by direct intramuscular delivery of plasmid DNA or oral delivery using a recombinant suicidal Listeria monocytogenes strain (rsΔ2). In a previous report we described how rsΔ2 is capable of delivering luciferase, as protein or DNA, in vitro, into non-dividing intestinal epithelial cells (Kuo et al., 2009). This is achieved by engineering a dual expression shuttle vector, pDuLX-Luc, that replicates in E. coli and rsΔ2 and drives gene expression from the Listeria promoter (Phly) as well as the eukaryotic cytomegalovirus promoter (CMV), thereby delivering both protein and plasmid DNA to the cell cytoplasm. For the current in vivo study rsΔ2 containing pDuLX-OVA was used to deliver both ovalbumin protein and the mammalian expression plasmid by the oral route. Controls were used to investigate the activity of this system versus positive and negative controls, as well as quantifying activity against direct intramuscular injection of expression plasmids. Oral administration of rsΔ2(pDuLX-OVA) produced significant titres of antibody and was effective at inducing targeted T-cell lysis (approximately 30% lysis relative to an experimental positive control, intravenous OVA-coated splenocytes+lipopolysaccharide). Intramuscular injection of plasmids pDuLX-OVA or p3L-OVA (which lacks the prokaryotic promoter) also produced significant CTL-mediated cell lysis. The delivery of the negative control rsΔ2 (pDuLX-Luc) confirmed that the observed activity was induced specifically by the ovalbumin vaccination. The data suggest that the oral activity of rsΔ2(pDuLX-OVA) is explained by delivery of OVA protein, expressed in rsΔ2 from the prokaryotic promoter present in pDuLX-OVA, but transfection of mammalian cells in vivo may also play a role. Antibody titres were also produced by oral delivery (in rsΔ2) of the p3L-OVA plasmid in which does not include a prokaryotic promoter.
Collapse
Affiliation(s)
- Shubhra Sinha
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Cheng-Yi Kuo
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Joan K Ho
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Paul J White
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Jalal A Jazayeri
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia.
| |
Collapse
|
40
|
Kumar V, Ahmad A. Targeting calpains: A novel immunomodulatory approach for microbial infections. Eur J Pharmacol 2017; 814:28-44. [PMID: 28789934 DOI: 10.1016/j.ejphar.2017.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 02/09/2023]
Abstract
Calpains are a family of Ca2+ dependent cytosolic non-lysosomal proteases with well conserved cysteine-rich domains for enzymatic activity. Due to their functional dependency on Ca2+ concentrations, they are involved in various cellular processes that are regulated by intracellular ca2+ concentration (i.e. embryo development, cell development and migration, maintenance of cellular architecture and structure etc.). Calpains are widely studied proteases in mammalian (i.e. mouse and human) physiology and pathophysiology due to their ubiquitous presence. For example, these proteases have been found to be involved in various inflammatory disorders such as neurodegeneration, cancer, brain and myocardial ischemia and infarction, cataract and muscular dystrophies etc. Besides their role in these sterile inflammatory conditions, calpains have also been shown to regulate a wide range of infectious diseases (i.e. sepsis, tuberculosis, gonorrhoea and bacillary dysentery etc.). One of these regulatory mechanisms mediated by calpains (i.e. calpain 1 and 2) during microbial infections involves the regulation of innate immune response, inflammation and cell death. Thus, the major emphasis of this review is to highlight the importance of calpains in the pathogenesis of various microbial (i.e. bacterial, fungal and viral) diseases and the use of calpain modulators as potential immunomodulators in microbial infections.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Paediatrics and Child Health, Children's Health Queensland Clinical Unit, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| | - Ali Ahmad
- Laboratory of innate immunity, CHU Ste-Justine Research Center/Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, 3175 Cote Ste Catherine, Montreal, Quebec, Canada H3T 1C5.
| |
Collapse
|
41
|
Schmitter S, Fieseler L, Klumpp J, Bertram R, Loessner MJ. TetR-dependent gene regulation in intracellularListeria monocytogenesdemonstrates the spatiotemporal surface distribution of ActA. Mol Microbiol 2017; 105:413-425. [DOI: 10.1111/mmi.13706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Sibylle Schmitter
- Institute of Food, Nutrition and Health; ETH Zurich; Schmelzbergstrasse 7 Zurich CH-8092 Switzerland
| | - Lars Fieseler
- Institute of Food, Nutrition and Health; ETH Zurich; Schmelzbergstrasse 7 Zurich CH-8092 Switzerland
| | - Jochen Klumpp
- Institute of Food, Nutrition and Health; ETH Zurich; Schmelzbergstrasse 7 Zurich CH-8092 Switzerland
| | - Ralph Bertram
- Lehrbereich Mikrobielle Genetik; Eberhard-Karls-Universität Tübingen; Auf der Morgenstelle 28 Tübingen D-72076 Germany
| | - Martin J. Loessner
- Institute of Food, Nutrition and Health; ETH Zurich; Schmelzbergstrasse 7 Zurich CH-8092 Switzerland
| |
Collapse
|
42
|
NicAogáin K, O’Byrne CP. The Role of Stress and Stress Adaptations in Determining the Fate of the Bacterial Pathogen Listeria monocytogenes in the Food Chain. Front Microbiol 2016; 7:1865. [PMID: 27933042 PMCID: PMC5120093 DOI: 10.3389/fmicb.2016.01865] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/04/2016] [Indexed: 12/15/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes is a highly adaptable organism that can persist in a wide range of environmental and food-related niches. The consumption of contaminated ready-to-eat foods can cause infections, termed listeriosis, in vulnerable humans, particularly those with weakened immune systems. Although these infections are comparatively rare they are associated with high mortality rates and therefore this pathogen has a significant impact on food safety. L. monocytogenes can adapt to and survive a wide range of stress conditions including low pH, low water activity, and low temperature, which makes it problematic for food producers who rely on these stresses for preservation. Stress tolerance in L. monocytogenes can be explained partially by the presence of the general stress response (GSR), a transcriptional response under the control of the alternative sigma factor sigma B (σB) that reconfigures gene transcription to provide homeostatic and protective functions to cope with the stress. Within the host σB also plays a key role in surviving the harsh conditions found in the gastrointestinal tract. As the infection progresses beyond the GI tract L. monocytogenes uses an intracellular infectious cycle to propagate, spread and remain protected from the host's humoral immunity. Many of the virulence genes that facilitate this infectious cycle are under the control of a master transcriptional regulator called PrfA. In this review we consider the environmental reservoirs that enable L. monocytogenes to gain access to the food chain and discuss the stresses that the pathogen must overcome to survive and grow in these environments. The overlap that exists between stress tolerance and virulence is described. We review the principal measures that are used to control the pathogen and point to exciting new approaches that might provide improved means of control in the future.
Collapse
Affiliation(s)
| | - Conor P. O’Byrne
- Bacterial Stress Response Group, Microbiology, School of Natural Sciences, College of Science, National University of IrelandGalway, Ireland
| |
Collapse
|
43
|
Pensinger DA, Boldon KM, Chen GY, Vincent WJB, Sherman K, Xiong M, Schaenzer AJ, Forster ER, Coers J, Striker R, Sauer JD. The Listeria monocytogenes PASTA Kinase PrkA and Its Substrate YvcK Are Required for Cell Wall Homeostasis, Metabolism, and Virulence. PLoS Pathog 2016; 12:e1006001. [PMID: 27806131 PMCID: PMC5091766 DOI: 10.1371/journal.ppat.1006001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/14/2016] [Indexed: 12/02/2022] Open
Abstract
Obstacles to bacterial survival and replication in the cytosol of host cells, and the mechanisms used by bacterial pathogens to adapt to this niche are not well understood. Listeria monocytogenes is a well-studied Gram-positive foodborne pathogen that has evolved to invade and replicate within the host cell cytosol; yet the mechanisms by which it senses and responds to stress to survive in the cytosol are largely unknown. To assess the role of the L. monocytogenes penicillin-binding-protein and serine/threonine associated (PASTA) kinase PrkA in stress responses, cytosolic survival and virulence, we constructed a ΔprkA deletion mutant. PrkA was required for resistance to cell wall stress, growth on cytosolic carbon sources, intracellular replication, cytosolic survival, inflammasome avoidance and ultimately virulence in a murine model of Listeriosis. In Bacillus subtilis and Mycobacterium tuberculosis, homologues of PrkA phosphorylate a highly conserved protein of unknown function, YvcK. We found that, similar to PrkA, YvcK is also required for cell wall stress responses, metabolism of glycerol, cytosolic survival, inflammasome avoidance and virulence. We further demonstrate that similar to other organisms, YvcK is directly phosphorylated by PrkA, although the specific site(s) of phosphorylation are not highly conserved. Finally, analysis of phosphoablative and phosphomimetic mutants of YvcK in vitro and in vivo demonstrate that while phosphorylation of YvcK is irrelevant to metabolism and cell wall stress responses, surprisingly, a phosphomimetic, nonreversible negative charge of YvcK is detrimental to cytosolic survival and virulence in vivo. Taken together our data identify two novel virulence factors essential for cytosolic survival and virulence of L. monocytogenes. Furthermore, our data demonstrate that regulation of YvcK phosphorylation is tightly controlled and is critical for virulence. Finally, our data suggest that yet to be identified substrates of PrkA are essential for cytosolic survival and virulence of L. monocytogenes and illustrate the importance of studying protein phosphorylation in the context of infection.
Collapse
Affiliation(s)
- Daniel A. Pensinger
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Kyle M. Boldon
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Grischa Y. Chen
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - William J. B. Vincent
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Kyle Sherman
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Meng Xiong
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Adam J. Schaenzer
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Emily R. Forster
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina
| | - Rob Striker
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
- W. S. Middleton Memorial Veteran’s Hospital, Madison, Wisconsin
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
44
|
Quereda JJ, García-Del Portillo F, Pucciarelli MG. Listeria monocytogenes remodels the cell surface in the blood-stage. ENVIRONMENTAL MICROBIOLOGY REPORTS 2016; 8:641-648. [PMID: 27085096 DOI: 10.1111/1758-2229.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
After crossing the intestinal barrier, the bacterial pathogen Listeria monocytogenes disseminates via the blood to the liver, spleen, brain and placenta. Transcriptomic studies have shown that L. monocytogenes changes expression of many genes during this blood-stage. However, no comparable data at the protein level are known. As main interactors with the environment, we focused in surface proteins produced by L. monocytogenes in an ex vivo bovine blood model. Bacteria exposed to blood alter selectively the amount of several surface proteins compared with bacteria grown in laboratory media. Increased levels were detected for Lmo0514 and Internalin A, two surface proteins covalently bound to peptidoglycan, and the moonlighting protein alcohol-acetaldehyde dehydrogenase, also known as Lap for 'Listeria adhesion protein'. Lmo0514, induced by L. monocytogenes inside epithelial cells, is required for survival in plasma and for virulence in mice at early infection stages. Lmo0514 is also important to cope with low pH stress. By contrast, L. monocytogenes down-regulates other surface proteins following exposure to blood and plasma such as Internalin I. These data provide evidence for remodelling of the L. monocytogenes cell surface during the blood-stage, which it could facilitate pathogen dissemination to deep organs.
Collapse
Affiliation(s)
- Juan J Quereda
- Laboratory of Intracellular Bacterial Pathogens, Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Francisco García-Del Portillo
- Laboratory of Intracellular Bacterial Pathogens, Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - M Graciela Pucciarelli
- Laboratory of Intracellular Bacterial Pathogens, Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO-CSIC), Departamento de Biología Molecular, Universidad Autónoma de Madrid, Spain
| |
Collapse
|
45
|
Rahman SA, Munther D, Fazil A, Smith B, Wu J. Unraveling the dose-response puzzle of L. monocytogenes: A mechanistic approach. Infect Dis Model 2016; 1:101-114. [PMID: 29928724 PMCID: PMC5963320 DOI: 10.1016/j.idm.2016.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/14/2016] [Accepted: 09/21/2016] [Indexed: 12/27/2022] Open
Abstract
Food-borne disease outbreaks caused by Listeria monocytogenes continue to impose heavy burdens on public health in North America and globally. To explore the threat L. monocytogenes presents to the elderly, pregnant woman and immuno-compromised individuals, many studies have focused on in-host infection mechanisms and risk evaluation in terms of dose-response outcomes. However, the connection of these two foci has received little attention, leaving risk prediction with an insufficient mechanistic basis. Consequently, there is a critical need to quantifiably link in-host infection pathways with the dose-response paradigm. To better understand these relationships, we propose a new mathematical model to describe the gastro-intestinal pathway of L. monocytogenes within the host. The model dynamics are shown to be sensitive to inoculation doses and exhibit bi-stability phenomena. Applying the model to guinea pigs, we show how it provides useful tools to identify key parameters and to inform critical values of these parameters that are pivotal in risk evaluation. Our preliminary analysis shows that the effect of gastro-environmental stress, the role of commensal microbiota and immune cells are critical for successful infection of L. monocytogenes and for dictating the shape of the dose-response curves.
Collapse
Affiliation(s)
- S.M. Ashrafur Rahman
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada
| | - Daniel Munther
- Department of Mathematics, Cleveland State University, Cleveland, OH 44115, United States
| | - Aamir Fazil
- National Microbiology Laboratory, Public Health Agency of Canada, Guelph, ON N1G 5B2, Canada
| | - Ben Smith
- National Microbiology Laboratory, Public Health Agency of Canada, Guelph, ON N1G 5B2, Canada
| | - Jianhong Wu
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
46
|
Wang S, Liu P, Wei J, Zhu Z, Shi Z, Shao D, Ma Z. Tumor suppressor p53 protects mice against Listeria monocytogenes infection. Sci Rep 2016; 6:33815. [PMID: 27644341 PMCID: PMC5028743 DOI: 10.1038/srep33815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023] Open
Abstract
Tumor suppressor p53 is involved in regulating immune responses, which contribute to antitumor and antiviral activity. However, whether p53 has anti-bacterial functions remains unclear. Listeria monocytogenes (LM) causes listeriosis in humans and animals, and it is a powerful model for studying innate and adaptive immunity. In the present study, we illustrate an important regulatory role of p53 during LM infection. p53 knockout (p53KO) mice were more susceptible to LM infection, which was manifested by a shorter survival time and lower survival rate. p53KO mice showed significant impairments in LM eradication. Knockdown of p53 in RAW264.7 and HeLa cells resulted in increased invasion and intracellular survival of LM. Furthermore, the invasion and intracellular survival of LM was inhibited in p53-overexpressing RAW264.7 and HeLa cells. LM-infected p53KO mice exhibited severe clinical symptoms and organ injury, presumably because of the abnormal production of the pro-inflammatory cytokines TNF-α, IL-6, IL-12, and IL-18. Decreased IFN-γ and GBP1 productions were observed in LM-infected p53-deficient mice or cells. The combination of these defects likely resulted in the overwhelming LM infection in the p53KO mice. These observations indicate that p53 serves as an important regulator of the host innate immune that protects against LM infection.
Collapse
Affiliation(s)
- Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Pingping Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zixiang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China.,State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Zixue Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| |
Collapse
|
47
|
Gelbíčová T, Pantůček R, Karpíšková R. Virulence factors and resistance to antimicrobials in Listeria monocytogenes
serotype 1/2c isolated from food. J Appl Microbiol 2016; 121:569-76. [DOI: 10.1111/jam.13191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 12/14/2022]
Affiliation(s)
- T. Gelbíčová
- Veterinary Research Institute; Brno Czech Republic
| | - R. Pantůček
- Department of Experimental Biology; Faculty of Science; Masaryk University; Brno Czech Republic
| | | |
Collapse
|
48
|
Rehaiem A, Fhoula I, Slim AF, Ben Boubaker IB, Chihi AB, Ouzari HI. Prevalence, acquired antibiotic resistance and bacteriocin production of Enterococcus spp. isolated from tunisian fermented food products. Food Control 2016. [DOI: 10.1016/j.foodcont.2015.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
49
|
Li L, Chen K, Xiang Y, Yoshimura T, Su S, Zhu J, Bian XW, Wang JM. New development in studies of formyl-peptide receptors: critical roles in host defense. J Leukoc Biol 2016; 99:425-35. [PMID: 26701131 PMCID: PMC4750370 DOI: 10.1189/jlb.2ri0815-354rr] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/29/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
Formyl-peptide receptors are a family of 7 transmembrane domain, Gi-protein-coupled receptors that possess multiple functions in many pathophysiologic processes because of their expression in a variety of cell types and their capacity to interact with a variety of structurally diverse, chemotactic ligands. Accumulating evidence demonstrates that formyl-peptide receptors are critical mediators of myeloid cell trafficking in the sequential chemotaxis signal relays in microbial infection, inflammation, and immune responses. Formyl-peptide receptors are also involved in the development and progression of cancer. In addition, one of the formyl-peptide receptor family members, Fpr2, is expressed by normal mouse-colon epithelial cells, mediates cell responses to microbial chemotactic agonists, participates in mucosal development and repair, and protects against inflammation-associated tumorigenesis. These novel discoveries greatly expanded the current understanding of the role of formyl-peptide receptors in host defense and as potential molecular targets for the development of therapeutics.
Collapse
Affiliation(s)
- Liangzhu Li
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Keqiang Chen
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yi Xiang
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Teizo Yoshimura
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaobo Su
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jianwei Zhu
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiu-wu Bian
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ji Ming Wang
- *Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA; Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China; and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
50
|
Schultze T, Hilker R, Mannala GK, Gentil K, Weigel M, Farmani N, Windhorst AC, Goesmann A, Chakraborty T, Hain T. A detailed view of the intracellular transcriptome of Listeria monocytogenes in murine macrophages using RNA-seq. Front Microbiol 2015; 6:1199. [PMID: 26579105 PMCID: PMC4627465 DOI: 10.3389/fmicb.2015.01199] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/15/2015] [Indexed: 01/21/2023] Open
Abstract
Listeria monocytogenes is a bacterial pathogen and causative agent for the foodborne infection listeriosis, which is mainly a threat for pregnant, elderly, or immunocompromised individuals. Due to its ability to invade and colonize diverse eukaryotic cell types including cells from invertebrates, L. monocytogenes has become a well-established model organism for intracellular growth. Almost 10 years ago, we and others presented the first whole-genome microarray-based intracellular transcriptome of L. monocytogenes. With the advent of newer technologies addressing transcriptomes in greater detail, we revisit this work, and analyze the intracellular transcriptome of L. monocytogenes during growth in murine macrophages using a deep sequencing based approach. We detected 656 differentially expressed genes of which 367 were upregulated during intracellular growth in macrophages compared to extracellular growth in Brain Heart Infusion broth. This study confirmed ∼64% of all regulated genes previously identified by microarray analysis. Many of the regulated genes that were detected in the current study involve transporters for various metals, ions as well as complex sugars such as mannose. We also report changes in antisense transcription, especially upregulations during intracellular bacterial survival. A notable finding was the detection of regulatory changes for a subset of temperate A118-like prophage genes, thereby shedding light on the transcriptional profile of this bacteriophage during intracellular growth. In total, our study provides an updated genome-wide view of the transcriptional landscape of L. monocytogenes during intracellular growth and represents a rich resource for future detailed analysis.
Collapse
Affiliation(s)
- Tilman Schultze
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Rolf Hilker
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany ; Bioinformatics and Systems Biology, Justus Liebig University Giessen, Germany
| | - Gopala K Mannala
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Katrin Gentil
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Markus Weigel
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Neda Farmani
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Anita C Windhorst
- Institute of Medical Informatics, Justus Liebig University Giessen, Germany
| | - Alexander Goesmann
- Bioinformatics and Systems Biology, Justus Liebig University Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - Torsten Hain
- Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| |
Collapse
|