451
|
Pizarro-Cerdá J, Cossart P. Microbe Profile: Listeria monocytogenes: a paradigm among intracellular bacterial pathogens. MICROBIOLOGY-SGM 2019; 165:719-721. [PMID: 31124782 DOI: 10.1099/mic.0.000800] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Listeria monocytogenes is a food-borne bacterial pathogen that is responsible for listeriosis, a disease characterized by occasional febrile gastroenteritis in immunocompetent individuals, abortions in pregnant women, meningitis in the newborn and fatal bacteraemia in immunocompromised individuals or the elderly. The ability of L. monocytogenes to produce disease is intimately associated with its potential to traverse several human barriers (including the intestinal, placental and blood/brain barriers), to promote its internalization within diverse populations of epithelial cells and to proliferate in the intra-ic environment while escaping host immune responses. L. monocytogenes is often regarded as a paradigm for intracellular parasitism.
Collapse
Affiliation(s)
- Javier Pizarro-Cerdá
- Yersinia Research Unit, Department of Microbiology, Institut Pasteur - 75015 Paris, France
| | - Pascale Cossart
- Bacteria-Cell Interactions Unit, Department of Cell Biology and Infection, Institut Pasteur - 75015 Paris, France
| |
Collapse
|
452
|
Muchaamba F, Eshwar AK, Stevens MJA, von Ah U, Tasara T. Variable Carbon Source Utilization, Stress Resistance, and Virulence Profiles Among Listeria monocytogenes Strains Responsible for Listeriosis Outbreaks in Switzerland. Front Microbiol 2019; 10:957. [PMID: 31130938 PMCID: PMC6510287 DOI: 10.3389/fmicb.2019.00957] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022] Open
Abstract
A combination of phenotype microarrays, targeted stress resistance and virulence assays and comparative genome analysis was used to compare a set of Listeria monocytogenes strains including those involved in previous Swiss foodborne listeriosis outbreaks. Despite being highly syntenic in gene content these strains showed significant phenotypic variation in utilization of different carbon (C)-sources as well as in resistance of osmotic and pH stress conditions that are relevant to host and food associated environments. An outbreak strain from the 2005 Swiss Tomme cheese listeriosis outbreak (Lm3163) showed the highest versatility in C-sources utilized whereas the strain responsible for the 1983 to 1987 Vacherin Montd'or cheese listeriosis outbreak (LL195) showed the highest tolerance to both osmotic and pH stress conditions among the examined strains. Inclusion of L-norvaline led to enhanced resistance of acidic stress in all the examined strains and there were strain-strain-specific differences observed in the ability of other amino acids and urea to enhance acid stress resistance in L. monocytogenes. A strain dependent inhibition pattern was also observed upon inclusion of β-phenylethylamine under alkaline stress conditions. In targeted phenotypic analysis the strain-specific differences in salt stress tolerance uncovered in phenotypic microarrays were corroborated and variations in host cell invasion and virulence among the examined strains were also revealed. Outbreak associated strains representing lineage I serotype 4b showed superior pathogenicity in a zebrafish infection model whilst Lm3163 a lineage II serotype 1/2a outbreak strain demonstrated the highest cellular invasion capacity amongst the tested strains. A genome wide sequence comparison of the strains only revealed few genetic differences between the strains suggesting that variations in gene regulation and expression are largely responsible for the phenotypic differences revealed among the examined strains. Our results have generated data that provides a potential basis for the future design of improved Listeria specific media to enhance routine detection and isolation of this pathogen as well as provide knowledge for developing novel methods for its control in food.
Collapse
Affiliation(s)
- Francis Muchaamba
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| | - Athmanya K. Eshwar
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| | - Marc J. A. Stevens
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| | | | - Taurai Tasara
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| |
Collapse
|
453
|
Zhu S, Zeng M, Guo W, Feng G, Wu H. Catalase-mimetic gold nanoparticles inhibit the antagonistic action of Lactobacillus gasseri toward foodborne enteric pathogens in associative cultures. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:55-66. [PMID: 31017521 DOI: 10.1080/10590501.2019.1591698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gold nanoparticles (AuNPs) have been previously shown to induce gut dysbiosis during colitis in mice, but the underlying mechanism is not clear yet. Here, we evaluated the effects of AuNPs (5 nm diameter, coated with tannic acid, polyvinylpyrrolidone or citrate) on H2O2 accumulation and pathogen antagonization by an intestinal strain of Lactobacillus gasseri under aerobic cultural conditions. AuNPs (0.65 μg/mL) reduced over 50% of H2O2 accumulation by L. gasseri, and significantly inhibited the antagonistic action of L. gasseri on growth of four foodborne enteric pathogens, i.e. Salmonella enterica serovar Typhimurium, Escherichia coli, Listeria monocytogenes, and Staphylococcus aureus in associative cultures.
Collapse
Affiliation(s)
- Suqin Zhu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Mingyong Zeng
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Wei Guo
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Guangxin Feng
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Haohao Wu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| |
Collapse
|
454
|
Rahmeh R, Akbar A, Kishk M, Al-Onaizi T, Al-Azmi A, Al-Shatti A, Shajan A, Al-Mutairi S, Akbar B. Distribution and antimicrobial activity of lactic acid bacteria from raw camel milk. New Microbes New Infect 2019; 30:100560. [PMID: 31193267 PMCID: PMC6522851 DOI: 10.1016/j.nmni.2019.100560] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
Consumer demand for natural pathogen-control agents for substitution of synthetic food preservatives and traditional antibiotics is increasing. This study aimed to reveal the distribution of lactic acid bacteria (LAB) in raw camel milk and to characterize their antimicrobial traits. The genetic identification by 16S rRNA sequencing of 58 LAB isolates showed the predominance of Enterococcus (24.2%), Lactococcus (22.4%) and Pediococcus (20.7%) genera in raw camel milk. These genera exhibited inhibitory activity against a broad spectrum of Gram-positive and Gram-negative bacteria including multidrug-resistant Salmonella. Among these LAB, two isolates-identified as Pediococcus pentosaceus CM16 and Lactobacillus brevis CM22-were selected for their strong bacteriocinogenic anti-listerial activity estimated at 1600 and 800 AU/mL, respectively. The bacteriocins produced were partially purified by ammonium sulphate precipitation and gel filtration and then biochemically characterized. The proteinaceous nature of bacteriocins was confirmed by the susceptibility to enzymes. These bacteriocins showed significant technological characteristics such as heat-resistance, and stability over a wide range of pH (2.0-10.0). In conclusion, these results indicated that Pediococcus pentosaceus CM16 and Lactobacillus brevis CM22 could be useful as potential probiotics. Moreover, their partially purified bacteriocins may play an important role as food preservatives and feed additives. To our knowledge, this is the first report describing the distribution of LAB population in raw camel milk and the characterization of their bacteriocins from the Arabian Peninsula of western Asia.
Collapse
Affiliation(s)
- R Rahmeh
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - A Akbar
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - M Kishk
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - T Al-Onaizi
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - A Al-Azmi
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - A Al-Shatti
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - A Shajan
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - S Al-Mutairi
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - B Akbar
- Biotechnology Programme, Environment & Life Sciences Research Centre, Kuwait Institute for Scientific Research, Safat, Kuwait
| |
Collapse
|
455
|
Mathipa MG, Thantsha MS, Bhunia AK. Lactobacillus casei expressing Internalins A and B reduces Listeria monocytogenes interaction with Caco-2 cells in vitro. Microb Biotechnol 2019; 12:715-729. [PMID: 30989823 PMCID: PMC6559204 DOI: 10.1111/1751-7915.13407] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes has been implicated in a number of outbreaks including the recent largest outbreak in South Africa. Current methods for prevention of foodborne L. monocytogenes infection are inadequate, thus raising a need for an alternative strategy. Probiotic bioengineering is considered a prevailing approach to enhance the efficacy of probiotics for targeted control of pathogens. Here, the ability of Lactobacillus casei expressing the L. monocytogenes invasion proteins Internalins A and B (inlAB) to prevent infection was investigated. The inlAB operon was cloned and surface‐expressed on L. casei resulting in a recombinant strain, LbcInlAB, and subsequently, its ability to inhibit adhesion, invasion and translocation of L. monocytogenes through enterocyte‐like Caco‐2 cells was examined. Cell surface expression of InlAB on the LbcInlAB was confirmed by Western blotting and immunofluorescence staining. The LbcInlAB strain showed significantly higher (P < 0.0001) adherence, invasion and translocation of Caco‐2 cells than the wild‐type L. casei strain (LbcWT), as well as reduced L. monocytogenes adhesion, invasion and transcellular passage through the cell monolayer than LbcWT. Furthermore, pre‐exposure of Caco‐2 cells to LbcInlAB significantly reduced L. monocytogenes‐induced cell cytotoxicity and epithelial barrier dysfunction. These results suggest that InlAB‐expressing L. casei could be a potential practical approach for prevention of listeriosis.
Collapse
Affiliation(s)
- Moloko G Mathipa
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa.,Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Mapitsi S Thantsha
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
456
|
Hofer U. Listeria pioneers. Nat Rev Microbiol 2019; 17:196-197. [DOI: 10.1038/s41579-019-0161-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
457
|
Tan L, Yuan X, Liu Y, Cai X, Guo S, Wang A. Non-muscle Myosin II: Role in Microbial Infection and Its Potential as a Therapeutic Target. Front Microbiol 2019; 10:401. [PMID: 30886609 PMCID: PMC6409350 DOI: 10.3389/fmicb.2019.00401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/15/2019] [Indexed: 01/11/2023] Open
Abstract
Currently, the major measures of preventing and controlling microbial infection are vaccinations and drugs. However, the appearance of drug resistance microbial mounts is main obstacle in current anti-microbial therapy. One of the most ubiquitous actin-binding proteins, non-muscle myosin II (NM II) plays a crucial role in a wide range of cellular physiological activities in mammals, including cell adhesion, migration, and division. Nowadays, growing evidence indicates that aberrant expression or activity of NM II can be detected in many diseases caused by microbes, including viruses and bacteria. Furthermore, an important role for NM II in the infection of some microbes is verified. Importantly, modulating the expression of NM II with small hairpin RNA (shRNA) or the activity of it by inhibitors can affect microbial-triggered phenotypes. Therefore, NM II holds the promise to be a potential target for inhibiting the infection of microbes and even treating microbial-triggered discords. In spite of these, a comprehensive view on the functions of NM II in microbial infection and the regulators which have an impact on the roles of NM II in this context, is still lacking. In this review, we summarize our current knowledge on the roles of NM II in microbial-triggered discords and provide broad insights into its regulators. In addition, the existing challenge of investigating the multiple roles of NM II in microbial infection and developing NM II inhibitors for treating these microbial-triggered discords, are also discussed.
Collapse
Affiliation(s)
- Lei Tan
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research and Development Center for Animal Reverse Vaccinology of Hunan Province, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xiaomin Yuan
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research and Development Center for Animal Reverse Vaccinology of Hunan Province, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Yisong Liu
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research and Development Center for Animal Reverse Vaccinology of Hunan Province, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shiyin Guo
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Aibing Wang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Research and Development Center for Animal Reverse Vaccinology of Hunan Province, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| |
Collapse
|
458
|
Abstract
The Gram-positive pathogen Listeria monocytogenes is able to promote its entry into a diverse range of mammalian host cells by triggering plasma membrane remodeling, leading to bacterial engulfment. Upon cell invasion, L. monocytogenes disrupts its internalization vacuole and translocates to the cytoplasm, where bacterial replication takes place. Subsequently, L. monocytogenes uses an actin-based motility system that allows bacterial cytoplasmic movement and cell-to-cell spread. L. monocytogenes therefore subverts host cell receptors, organelles and the cytoskeleton at different infection steps, manipulating diverse cellular functions that include ion transport, membrane trafficking, post-translational modifications, phosphoinositide production, innate immune responses as well as gene expression and DNA stability.
Collapse
|
459
|
Costa AC, Carvalho F, Cabanes D, Sousa S. Stathmin recruits tubulin to Listeria monocytogenes-induced actin comets and promotes bacterial dissemination. Cell Mol Life Sci 2019; 76:961-975. [PMID: 30506415 PMCID: PMC11105747 DOI: 10.1007/s00018-018-2977-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/24/2018] [Accepted: 11/22/2018] [Indexed: 01/18/2023]
Abstract
The tubulin cytoskeleton is one of the main components of the cytoarchitecture and is involved in several cellular functions. Here, we examine the interplay between Listeria monocytogenes (Lm) and the tubulin cytoskeleton upon cellular infection. We show that non-polymeric tubulin is present throughout Lm actin comet tails and, to a less extent, in actin clouds. Moreover, we demonstrate that stathmin, a regulator of microtubule dynamics, is also found in these Lm-associated actin structures and is required for tubulin recruitment. Depletion of host stathmin results in longer comets containing less F-actin, which may be correlated with higher levels of inactive cofilin in the comet, thus suggesting a defect on local F-actin dynamics. In addition, intracellular bacterial speed is significantly reduced in stathmin-depleted cells, revealing the importance of stathmin/tubulin in intracellular Lm motility. In agreement, the area of infection foci and the total bacterial loads are also significantly reduced in stathmin-depleted cells. Collectively, our results demonstrate that stathmin promotes efficient cellular infection, possibly through tubulin recruitment and control of actin dynamics at Lm-polymerized actin structures.
Collapse
Affiliation(s)
- Ana Catarina Costa
- Group of Molecular Microbiology, i3S-Instituto de Investigação e Inovação em Saúde, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Filipe Carvalho
- Group of Molecular Microbiology, i3S-Instituto de Investigação e Inovação em Saúde, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, 25 Rue du Dr Roux, 75015, Paris, France
| | - Didier Cabanes
- Group of Molecular Microbiology, i3S-Instituto de Investigação e Inovação em Saúde, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, i3S-Instituto de Investigação e Inovação em Saúde, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
| |
Collapse
|
460
|
De Abrew Abeysundara P, Dhowlaghar N, Nannapaneni R. Influence of cold stress on the survival of Listeria monocytogenes Bug600 and ScottA in lethal alkali, acid and oxidative stress. Lebensm Wiss Technol 2019. [DOI: 10.1016/j.lwt.2018.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
461
|
Las Heras V, Clooney AG, Ryan FJ, Cabrera-Rubio R, Casey PG, Hueston CM, Pinheiro J, Rudkin JK, Melgar S, Cotter PD, Hill C, Gahan CGM. Short-term consumption of a high-fat diet increases host susceptibility to Listeria monocytogenes infection. MICROBIOME 2019; 7:7. [PMID: 30658700 PMCID: PMC6339339 DOI: 10.1186/s40168-019-0621-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/04/2019] [Indexed: 05/02/2023]
Abstract
BACKGROUND A westernized diet comprising a high caloric intake from animal fats is known to influence the development of pathological inflammatory conditions. However, there has been relatively little focus upon the implications of such diets for the progression of infectious disease. Here, we investigated the influence of a high-fat (HF) diet upon parameters that influence Listeria monocytogenes infection in mice. RESULTS We determined that short-term administration of a HF diet increases the number of goblet cells, a known binding site for the pathogen, in the gut and also induces profound changes to the microbiota and promotes a pro-inflammatory gene expression profile in the host. Host physiological changes were concordant with significantly increased susceptibility to oral L. monocytogenes infection in mice fed a HF diet relative to low fat (LF)- or chow-fed animals. Prior to Listeria infection, short-term consumption of HF diet elevated levels of Firmicutes including Coprococcus, Butyricicoccus, Turicibacter and Clostridium XIVa species. During active infection with L. monocytogenes, microbiota changes were further exaggerated but host inflammatory responses were significantly downregulated relative to Listeria-infected LF- or chow-fed groups, suggestive of a profound tempering of the host response influenced by infection in the context of a HF diet. The effects of diet were seen beyond the gut, as a HF diet also increased the sensitivity of mice to systemic infection and altered gene expression profiles in the liver. CONCLUSIONS We adopted a systems approach to identify the effects of HF diet upon L. monocytogenes infection through analysis of host responses and microbiota changes (both pre- and post-infection). Overall, the results indicate that short-term consumption of a westernized diet has the capacity to significantly alter host susceptibility to L. monocytogenes infection concomitant with changes to the host physiological landscape. The findings suggest that diet should be a consideration when developing models that reflect human infectious disease.
Collapse
Affiliation(s)
- Vanessa Las Heras
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Adam G Clooney
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Feargal J Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Pat G Casey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Cara M Hueston
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jorge Pinheiro
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Justine K Rudkin
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Cormac G M Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
462
|
Drolia R, Bhunia AK. Crossing the Intestinal Barrier via Listeria Adhesion Protein and Internalin A. Trends Microbiol 2019; 27:408-425. [PMID: 30661918 DOI: 10.1016/j.tim.2018.12.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/11/2018] [Accepted: 12/14/2018] [Indexed: 12/24/2022]
Abstract
The intestinal epithelial cell lining provides the first line of defense, yet foodborne pathogens such as Listeria monocytogenes can overcome this barrier; however, the underlying mechanism is not well understood. Though the host M cells in Peyer's patch and the bacterial invasion protein internalin A (InlA) are involved, L. monocytogenes can cross the gut barrier in their absence. The interaction of Listeria adhesion protein (LAP) with the host cell receptor (heat shock protein 60) disrupts the epithelial barrier, promoting bacterial translocation. InlA aids L. monocytogenes transcytosis via interaction with the E-cadherin receptor, which is facilitated by epithelial cell extrusion and goblet cell exocytosis; however, LAP-induced cell junction opening may be an alternative bacterial strategy for InlA access to E-cadherin and its translocation. Here, we summarize the strategies that L. monocytogenes employs to circumvent the intestinal epithelial barrier and compare and contrast these strategies with other enteric bacterial pathogens. Additionally, we provide implications of recent findings for food safety regulations.
Collapse
Affiliation(s)
- Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
463
|
Fan Z, Xie J, Li Y, Wang H. Listeriosis in mainland China: A systematic review. Int J Infect Dis 2019; 81:17-24. [PMID: 30641204 DOI: 10.1016/j.ijid.2019.01.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/26/2018] [Accepted: 01/05/2019] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE The aim of this study was to conduct a systematic review to better understand the epidemiological and clinical characteristics of listeriosis patients in mainland China. METHODS The six most widely used Chinese and English language databases were searched. The records of patients with listeriosis in mainland China reported during the years 2011-2017 were extracted. The clinical data of patients and information on clinical isolates of Listeria were collected and analyzed. RESULTS In total, 136 records were identified, reporting 562 patients with listeriosis. The number of patients was much higher than that reported in the previous decade. The 227 non-perinatal listeriosis patients included had a mortality rate of 23.78%. Of the 231 perinatal listeriosis patients, 32.68% resulted in abortion and/or newborn death. All listeriosis cases were reported as being sporadic. The listeriosis was traced to infection via a meat product in only three patients, while 33.12% were healthcare-associated infections. CONCLUSIONS The number of patients with listeriosis in mainland China may have been underestimated previously. Perinatal cases in mainland China account for a much higher proportion than is usually described. Considering the high number of listeriosis patients in China, a comprehensive monitoring system for Listeria is urgently needed.
Collapse
Affiliation(s)
- Zhangling Fan
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xie
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Li
- The Institute of Medical Information (IMI) and Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanling Wang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
464
|
Systemic Listeria monocytogenes Infection as a Model to Study T Helper Cell Immune Responses. Methods Mol Biol 2019; 1960:149-160. [PMID: 30798529 DOI: 10.1007/978-1-4939-9167-9_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Listeria monocytogenes, a Gram-positive facultative intracellular pathogen, has been widely used as a model for studying the immune response. Here, we describe a protocol for the systemic infection of mice with L. monocytogenes, followed by isolation of lymphocytes from spleens and lymph nodes. We also include details on how to culture and store L. monocytogenes, as well as the specifics for fluorescence-activated cell sorting (FACS) for CD4+ cells in response to the systemic infection. This protocol can be adapted by changing the dosage of L. monocytogenes for a more or less aggressive infection and/or sorting for other immune cell subtypes of interest.
Collapse
|
465
|
Role and regulation of the stress activated sigma factor sigma B (σ B) in the saprophytic and host-associated life stages of Listeria monocytogenes. ADVANCES IN APPLIED MICROBIOLOGY 2019; 106:1-48. [PMID: 30798801 DOI: 10.1016/bs.aambs.2018.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The stress activated sigma factor sigma B (σB) plays a pivotal role in allowing the food-borne bacterial pathogen Listeria monocytogenes to modulate its transcriptional landscape in order to survive in a variety of harsh environments both outside and within the host. While we have a comparatively good understanding of the systems under the control of this sigma factor much less is known about how the activity of σB is controlled. In this review, we present a current model describing how this sigma factor is thought to be controlled including an overview of what is known about stress sensing and the early signal transduction events that trigger its activation. We discuss the known regulatory overlaps between σB and other protein and RNA regulators in the cell. Finally, we describe the role of σB in surviving both saprophytic and host-associated stresses. The complexity of the regulation of this sigma factor reflects the significant role that it plays in the persistence of this important pathogen in the natural environment, the food chain as well as within the host during the early stages of an infection. Understanding its regulation will be a critical step in helping to develop rational strategies to prevent its growth and survival in the food destined for human consumption and in the prevention of listeriosis.
Collapse
|
466
|
Chua MD, Walker BD, Jin JP, Guttman JA. Calponins Are Recruited to Actin-Rich Structures Generated by Pathogenic Escherichia coli, Listeria, and Salmonella. Anat Rec (Hoboken) 2018; 301:2103-2111. [PMID: 30312538 DOI: 10.1002/ar.23956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/12/2023]
Abstract
The ingestion of enteropathogenic Escherichia coli (EPEC), Listeria monocytogenes, or Salmonella enterica serovar Typhimurium leads to their colonization of the intestinal lumen, which ultimately causes an array of ailments ranging from diarrhea to bacteremia. Once in the intestines, these microbes generate various actin-rich structures to attach, invade, or move within the host intestinal epithelial cells. Although an assortment of actin-associated proteins has been identified to varying degrees at these structures, the localization of many actin stabilizing proteins have yet to be analyzed. Here, we examined the recruitment of the actin-associated proteins, calponin 1 and 2 at EPEC pedestals, L. monocytogenes actin clouds, comet tails and listeriopods, and S. Typhimurium membrane ruffles. In other systems, calponins are known to bind to and stabilize actin filaments. In EPEC pedestals, calponin 1 was recruited uniformly throughout the structures while calponin 2 was enriched at the apical tip. During L. monocytogenes infections, calponin 1 was found through all the actin-rich structures generated by the bacteria, while calponin 2 was only present within actin-rich structures formed by L. monocytogenes near the host cell membrane. Finally, both calponins were found within S. Typhimurium-generated membrane ruffles. Taken together, we have shown that although calponin 1 is recruited to actin-rich structures formed by the three bacteria, calponin 2 is specifically recruited to only membrane-bound actin-rich structures formed by the bacteria. Thus, our findings suggest that calponin 2 is a novel marker for membrane-bound actin structures formed by pathogenic bacteria. Anat Rec, 301:2103-2111, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael Dominic Chua
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Julian A Guttman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
467
|
Zhang M, Gillaspy AF, Gipson JR, Cassidy BR, Nave JL, Brewer MF, Stoner JA, Chen J, Drevets DA. Neuroinvasive Listeria monocytogenes Infection Triggers IFN-Activation of Microglia and Upregulates Microglial miR-155. Front Immunol 2018; 9:2751. [PMID: 30538705 PMCID: PMC6277692 DOI: 10.3389/fimmu.2018.02751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/08/2018] [Indexed: 11/17/2022] Open
Abstract
MicroRNA (miR) miR-155 modulates microglial activation and polarization, but its role in activation of microglia during bacterial brain infection is unclear. We studied miR-155 expression in brains of C57BL/6 (B6.WT) mice infected i.p. with the neuro-invasive bacterial pathogen Listeria monocytogenes (L. monocytogenes). Infected mice were treated with ampicillin starting 2 days (d) post-infection (p.i.) and analyzed 3d, 7d, and 14d p.i. Virulent L. monocytogenes strains EGD and 10403s upregulated miR-155 in whole brain 7 d p.i. whereas infection with avirulent, non-neurotropic Δhly or ΔactA L. monocytogenes mutants did not. Similarly, infection with virulent but not mutated bacteria upregulated IFN-γ mRNA in the brain at 7 d p.i. Upregulation of miR-155 in microglia was confirmed by qPCR of flow cytometry-sorted CD45intCD11bpos brain cells. Subsequently, brain leukocyte influxes and gene expression in sorted microglia were compared in L. monocytogenes-infected B6.WT and B6.Cg-Mir155tm1.1Rsky/J (B6.miR-155−/−) mice. Brain influxes of Ly-6Chigh monocytes and upregulation of IFN-related genes in microglia were similar to B6.WT mice at 3 d p.i. In contrast, by d 7 p.i. expressions of microglial IFN-related genes, including markers of M1 polarization, were significantly lower in B6.miR-155−/− mice and by 14 d p.i., influxes of activated T-lymphocytes were markedly reduced. Notably, CD45highCD11bpos brain cells from B6.miR-155−/− mice isolated at 7 d p.i. expressed 2-fold fewer IFN-γ transcripts than did cells from B6.WT mice suggesting reduced IFN-γ stimulation contributed to dampened gene expression in B6.miR-155−/− microglia. Lastly, in vitro stimulation of 7 d p.i. brain cells with heat-killed L. monocytogenes induced greater production of TNF in B6.miR-155−/− microglia than in B6.WT microglia. Thus, miR-155 affects brain inflammation by multiple mechanisms during neuroinvasive L. monocytogenes infection. Peripheral miR-155 promotes brain inflammation through its required role in optimal development of IFN-γ-secreting lymphocytes that enter the brain and activate microglia. Microglial miR-155 promotes M1 polarization, and also inhibits inflammatory responses to stimulation by heat-killed L. monocytogenes, perhaps by targeting Tab2.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Allison F Gillaspy
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jenny R Gipson
- Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Benjamin R Cassidy
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jessica L Nave
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Misty F Brewer
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Julie A Stoner
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jie Chen
- Histology and Immunohistochemistry Core, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Douglas A Drevets
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
468
|
Fares E, McCloskey CB, Gutierrez A, Princiotta M, Salinas LJ, Drevets DA. Vaccine strain Listeria monocytogenes bacteremia occurring 31 months after immunization. Infection 2018; 47:489-492. [PMID: 30430399 DOI: 10.1007/s15010-018-1249-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Listeria monocytogenes is a food-borne, facultative intracellular bacterium that causes severe diseases such as sepsis and meningoencephalitis in immunocompromised hosts. Because it stimulates robust T-lymphocyte-mediated responses, attenuated L. monocytogenes are candidate vaccine vectors for tumor immunotherapy. CASE We report a case of bacteremia caused by vaccine strain L. monocytogenes (Axalimogene filolisbac) occurring 31 months after immunization against human papilloma virus (HPV) associated cervical cancer. CONCLUSION Receipt of a L. monocytogenes-based vaccine is a novel risk factor for delayed L. monocytogenes bacteremia.
Collapse
Affiliation(s)
- Elias Fares
- Section of Infectious Diseases, Department of Internal Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd, Suite 7300, Oklahoma City, OK, 73104, USA
- , Erie, USA
| | - Cindy B McCloskey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | | | - Linda J Salinas
- Section of Infectious Diseases, Department of Internal Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd, Suite 7300, Oklahoma City, OK, 73104, USA
| | - Douglas A Drevets
- Section of Infectious Diseases, Department of Internal Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd, Suite 7300, Oklahoma City, OK, 73104, USA.
- Medical Services, Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
469
|
BoseDasgupta S, Pieters J. Macrophage-microbe interaction: lessons learned from the pathogen Mycobacterium tuberculosis. Semin Immunopathol 2018; 40:577-591. [PMID: 30306257 DOI: 10.1007/s00281-018-0710-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
Macrophages, being the cornerstone of the immune system, have adapted the ancient nutrient acquisition mechanism of phagocytosis to engulf various infectious organisms thereby helping to orchestrate an appropriate host response. Phagocytosis refers to the process of internalization and degradation of particulate material, damaged and senescent cells and microorganisms by specialized cells, after which the vesicle containing the ingested particle, the phagosome, matures into acidic phagolysosomes upon fusion with hydrolytic enzyme-containing lysosomes. The destructive power of the macrophage is further exacerbated through the induction of macrophage activation upon a variety of inflammatory stimuli. Despite being the end-point for many phagocytosed microbes, the macrophage can also serve as an intracellular survival niche for a number of intracellular microorganisms. One microbe that is particularly successful at surviving within macrophages is the pathogen Mycobacterium tuberculosis, which can efficiently manipulate the macrophage at several levels, including modulation of the phagocytic pathway as well as interfering with a number of immune activation pathways that normally would lead to eradication of the internalized bacilli. M. tuberculosis excels at circumventing destruction within macrophages, thus establishing itself successfully for prolonged times within the macrophage. In this contribution, we describe a number of general features of macrophages in the context of their function to clear an infection, and highlight the strategies employed by M. tuberculosis to counter macrophage attack. Interestingly, research on the evasion tactics employed by M. tuberculosis within macrophages not only helps to design strategies to curb tuberculosis, but also allows a better understanding of host cell biology.
Collapse
Affiliation(s)
- Somdeb BoseDasgupta
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
| | - Jean Pieters
- Department of Biochemistry, Biozentrum, University of Basel, 50-70 Klingelbergstrasse, 4056, Basel, Switzerland.
| |
Collapse
|
470
|
Wałecka-Zacharska E, Gmyrek R, Skowron K, Kosek-Paszkowska K, Bania J. Duration of Heat Stress Effect on Invasiveness of L. monocytogenes Strains. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1457480. [PMID: 30402461 PMCID: PMC6198540 DOI: 10.1155/2018/1457480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/25/2018] [Accepted: 09/13/2018] [Indexed: 01/17/2023]
Abstract
During food production and food conservation, as well as the passage through the human gastrointestinal (GI) tract, L. monocytogenes is exposed to many adverse conditions which may elicit a stress response. As a result the pathogen may become more resistant to other unpropitious factors and may change its virulence. It has been shown that low and high temperature, salt, low pH, and high pressure affect the invasion capacity of L. monocytogenes. However, there is a scarcity of data on the duration of the stress effect on bacterial biology, including invasiveness. The aim of this work was to determine the period during which L. monocytogenes invasiveness remains altered under optimal conditions following exposure of bacteria to mild heat shock stress. Ten L. monocytogenes strains were exposed to heat shock at 54°C for 20 minutes. Then both heat-treated and nontreated control bacteria were incubated under optimal growth conditions, 37°C, for up to 72 hours and the invasion capacity was tested. Additionally, the expression of virulence and stress response genes was investigated in 2 strains. We found that heat stress exposure significantly decreases the invasiveness of all tested strains. However, during incubation at 37°C the invasion capacity of heat-treated strains recovered to the level of nontreated controls. The observed effect was strain-dependent and lasted from less than 24 hours to 72 hours. The invasiveness of 6 out of the 10 nontreated strains decreased during incubation at 37°C. The expression of inlAB correlated with the increase of invasiveness but the decrease of invasiveness did not correlate with changes of the level of these transcripts. Conclusions. The effect of heat stress on L. monocytogenes invasiveness is strain-dependent and was transient, lasting up to 72 hours.
Collapse
Affiliation(s)
- Ewa Wałecka-Zacharska
- Department of Food Hygiene and Consumer Health Protection, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Renata Gmyrek
- Department of Food Hygiene and Consumer Health Protection, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Krzysztof Skowron
- Department of Microbiology, Nicolaus Copernicus University in Toruń, Collegium Medicum of L. Rydygier in Bydgoszcz, Bydgoszcz, Poland
| | - Katarzyna Kosek-Paszkowska
- Department of Food Hygiene and Consumer Health Protection, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Jacek Bania
- Department of Food Hygiene and Consumer Health Protection, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| |
Collapse
|
471
|
Abstract
Listeria monocytogenes is an intracellular bacterial pathogen that is frequently associated with food-borne infection. The ability of L. monocytogenes to cross the blood-brain barrier (BBB) is concerning as it can lead to life-threatening meningitis and encephalitis. The BBB protects the brain microenvironment from various toxic metabolites and microbial pathogens found in the blood following infection, and therefore supports brain homeostasis. The mechanisms by which L. monocytogenes present in the bloodstream cross the BBB to cause brain infections are not fully understood and there is also a lack of a robust model system to study brain infections by L. monocytogenes. Here, we present a simple mouse infection model to determine whether bacteria have crossed the BBB and to quantitate the burden of bacteria that have colonized the brain in vivo. In this method, animals were infected intravenously with L. monocytogenes and were humanely euthanized by exposure to CO2 followed by cervical dislocation. Cardiac perfusion of the animals was performed prior to harvesting infected organs. Blood was collected before perfusion and the number of bacteria per organ or mL of blood was determined by plating dilutions of the blood or organ homogenates on agar plates and counting the number of colonies formed. This method can be used to study novel receptor-ligand interactions that enhance infection of the brain by L. monocytogenes and can be easily adapted for the study of multiple bacterial pathogens.
Collapse
Affiliation(s)
- Pallab Ghosh
- Department of Microbiology and Immunobiology, Harvard Medical School;
| | - Darrren E Higgins
- Department of Microbiology and Immunobiology, Harvard Medical School;
| |
Collapse
|
472
|
Roberts MF, Khan HM, Goldstein R, Reuter N, Gershenson A. Search and Subvert: Minimalist Bacterial Phosphatidylinositol-Specific Phospholipase C Enzymes. Chem Rev 2018; 118:8435-8473. [DOI: 10.1021/acs.chemrev.8b00208] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Mary F. Roberts
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | | | - Rebecca Goldstein
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | | | - Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
473
|
Marini E, Magi G, Ferretti G, Bacchetti T, Giuliani A, Pugnaloni A, Rippo MR, Facinelli B. Attenuation of Listeria monocytogenes Virulence by Cannabis sativa L. Essential Oil. Front Cell Infect Microbiol 2018; 8:293. [PMID: 30186775 PMCID: PMC6113369 DOI: 10.3389/fcimb.2018.00293] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022] Open
Abstract
Anti-virulence strategies are being explored as a novel approach to combat pathogens. Such strategies include inhibition of surface adhesion, tissue invasion, toxin production, and/or interference with the gene regulation of other virulence traits. Listeria monocytogenes, the causative agent of listeriosis, is a facultative intracellular food pathogen characterized by a wide distribution in the environment. Its ability to persist within biofilms and to develop resistance to sanitizers is the cause of significant problems in food processing plants and of steep costs for the food industry. In humans, the treatment of listeriosis is hampered by the intracellular location of listeriae and the poor intracellular penetration of some antibiotics. Eleven L. monocytogenes isolates from patients who were diagnosed with invasive listeriosis in Italy in 2014-2016 were studied. This in vitro and in vivo study explored the antibacterial and anti-virulence properties of a steam-distilled essential oil of Cannabis sativa L., which is being intensively investigated for its high content in powerful bioactive phytochemicals. Susceptibility experiments demonstrated a moderate bactericidal activity of the essential oil (Minimum Bactericidal Concentration > 2048 μg/mL). Assessment of the effects of sublethal concentrations of the essential oil on L. monocytogenes virulence traits demonstrated a significant action on motility. Listeriae were non-motile after exposure to the essential oil. Light and scanning electron microscopy documented aggregates of listeriae with the flagella trapped inside the cluster. Real-time RT-PCR experiments showed downregulation of flagellar motility genes and of the regulatory gene prfA. The ability to form biofilm and to invade Caco-2 cells was also significantly reduced. Galleria mellonella larvae infected with L. monocytogenes grown in presence of sublethal concentrations of the essential oil showed much higher survival rates compared with controls, suggesting that the extract inhibited tissue invasion. Food contamination with L. monocytogenes is a major concern for the food industry, particularly for plants making ready-to-eat and processed food. The present work provides a baseline in the study of the anti-virulence properties of the C. sativa essential oil against L. monocytogenes. Further studies are needed to understand if it could be used as an alternative agent for the control of L. monocytogenes in food processing plants.
Collapse
Affiliation(s)
- Emanuela Marini
- Unit of Microbiology, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Gloria Magi
- Unit of Microbiology, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Gianna Ferretti
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Tiziana Bacchetti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Angelica Giuliani
- Division of Pathology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Armanda Pugnaloni
- Division of Pathology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Maria Rita Rippo
- Division of Pathology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Bruna Facinelli
- Unit of Microbiology, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
474
|
Flickinger JC, Rodeck U, Snook AE. Listeria monocytogenes as a Vector for Cancer Immunotherapy: Current Understanding and Progress. Vaccines (Basel) 2018; 6:E48. [PMID: 30044426 PMCID: PMC6160973 DOI: 10.3390/vaccines6030048] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 01/10/2023] Open
Abstract
Listeria monocytogenes, a Gram-positive facultative anaerobic bacterium, is becoming a popular vector for cancer immunotherapy. Indeed, multiple vaccines have been developed utilizing modified Listeria as a tool for generating immune responses against a variety of cancers. Moreover, over a dozen clinical trials testing Listeria cancer vaccines are currently underway, which will help to understand the utility of Listeria vaccines in cancer immunotherapy. This review aims to summarize current views on how Listeria-based vaccines induce potent antitumor immunity and the current state of Listeria-based cancer vaccines in clinical trials.
Collapse
Affiliation(s)
- John C Flickinger
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Ulrich Rodeck
- Department of Dermatology, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
475
|
King MT, Huh I, Shenai A, Brooks TM, Brooks CL. Structural basis of V HH-mediated neutralization of the food-borne pathogen Listeria monocytogenes. J Biol Chem 2018; 293:13626-13635. [PMID: 29976754 DOI: 10.1074/jbc.ra118.003888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/01/2018] [Indexed: 12/12/2022] Open
Abstract
Listeria monocytogenes causes listeriosis, a potentially fatal food-borne disease. The condition is especially harmful to pregnant women. Listeria outbreaks can originate from diverse foods, highlighting the need for novel strategies to improve food safety. The first step in Listeria invasion is internalization of the bacteria, which is mediated by the interaction of the internalin family of virulence factors with host cell receptors. A crucial interaction for Listeria invasion of the placenta, and thus a target for therapeutic intervention, is between internalin B (InlB) and the receptor c-Met. Single-domain antibodies (VHH, also called nanobodies, or sdAbs) from camel heavy-chain antibodies are a novel solution for preventing Listeria infections. The VHH R303, R330, and R326 all bind InlB with high affinity; however, the molecular mechanism behind their mode of action was unknown. We demonstrate that despite a high degree of sequence and structural diversity, the VHH bind a single epitope on InlB. A combination of gentamicin protection assays and florescent microscopy establish that InlB-specific VHH inhibit Listeria invasion of HeLa cells. A high-resolution X-ray structure of VHH R303 in complex with InlB showed that the VHH binds at the c-Met interaction site on InlB, thereby acting as a competitive inhibitor preventing bacterial invasion. These results point to the potential of VHH as a novel class of therapeutics for the prevention of listeriosis.
Collapse
Affiliation(s)
- Moeko Toride King
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Ian Huh
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Akhilesh Shenai
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Teresa M Brooks
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Cory L Brooks
- From the Department of Chemistry, California State University, Fresno, California 93740
| |
Collapse
|
476
|
Zhu X, Liu D, Singh AK, Drolia R, Bai X, Tenguria S, Bhunia AK. Tunicamycin Mediated Inhibition of Wall Teichoic Acid Affects Staphylococcus aureus and Listeria monocytogenes Cell Morphology, Biofilm Formation and Virulence. Front Microbiol 2018; 9:1352. [PMID: 30034372 PMCID: PMC6043806 DOI: 10.3389/fmicb.2018.01352] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/05/2018] [Indexed: 12/14/2022] Open
Abstract
The emergence of bacterial resistance to therapeutic antibiotics limits options for treatment of common microbial diseases. Subinhibitory antibiotics dosing, often aid in the emergence of resistance, but its impact on pathogen’s physiology and pathogenesis is not well understood. Here we investigated the effect of tunicamycin, a cell wall teichoic acid (WTA) biosynthesis inhibiting antibiotic at the subinhibitory dosage on Staphylococcus aureus and Listeria monocytogenes physiology, antibiotic cross-resistance, biofilm-formation, and virulence. Minimum inhibitory concentration (MIC) of tunicamycin to S. aureus and L. monocytogenes was 20–40 μg/ml and 2.5–5 μg/ml, respectively, and the subinhibitory concentration was 2.5–5 μg/ml and 0.31–0.62 μg/ml, respectively. Tunicamycin pre-exposure reduced cellular WTA levels by 18–20% and affected bacterial cell wall ultrastructure, cell membrane permeability, morphology, laser-induced colony scatter signature, and bacterial ability to form biofilms. It also induced a moderate level of cross-resistance to tetracycline, ampicillin, erythromycin, and meropenem for S. aureus, and ampicillin, erythromycin, vancomycin, and meropenem for L. monocytogenes. Pre-treatment of bacterial cells with subinhibitory concentrations of tunicamycin also significantly reduced bacterial adhesion to and invasion into an enterocyte-like Caco-2 cell line, which is supported by reduced expression of key virulence factors, Internalin B (InlB) and Listeria adhesion protein (LAP) in L. monocytogenes, and a S. aureus surface protein A (SasA) in S. aureus. Tunicamycin-treated bacteria or the bacterial WTA preparation suppressed NF-κB and inflammatory cytokine production (TNFα, and IL-6) from murine macrophage cell line (RAW 264.7) indicating the reduced WTA level possibly attenuates an inflammatory response. These results suggest that at the subinhibitory dosage, tunicamycin-mediated inhibition of WTA biosynthesis interferes with cell wall structure, pathogens infectivity and inflammatory response, and ability to form biofilms but promotes the development of antibiotic cross-resistance.
Collapse
Affiliation(s)
- Xingyue Zhu
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States.,College of Science, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Dongqi Liu
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Atul K Singh
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Xingjian Bai
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Shivendra Tenguria
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, United States.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
477
|
Ireton K, Van Ngo H, Bhalla M. Interaction of microbial pathogens with host exocytic pathways. Cell Microbiol 2018; 20:e12861. [PMID: 29797532 DOI: 10.1111/cmi.12861] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/13/2018] [Indexed: 02/06/2023]
Abstract
Many microbial pathogens co-opt or perturb host membrane trafficking pathways. This review covers recent examples in which microbes interact with host exocytosis, the fusion of intracellular vesicles with the plasma membrane. The bacterial pathogens Listeria monocytogenes and Staphylococcus aureus subvert recycling endosomal pathways of exocytosis in order to induce their entry into human cells. By contrast, entry of the protozoan pathogen Trypanosoma cruzi or the virus adenovirus into host cells involves exploitation of lysosomal exocytosis. Toxins produced by Bacillus anthracis or Vibrio cholerae interfere with exocytosis pathways mediated by the GTPase Rab11 and the exocyst complex. By doing so, anthrax or cholera toxins impair recycling of cadherins to cell-cell junctions and disrupt the barrier properties of endothelial cells or intestinal epithelial cells, respectively. Uropathogenic Escherichia coli (UPEC) is expelled from bladder epithelial cells through two different exocytic routes that involve sensing of bacteria in vacuoles by host Toll-like receptor 4 (TLR4) or monitoring of the pH of lysosomes harbouring UPEC. The TLR4 pathway is mediated by multiple Rab GTPases and the exocyst, whereas the other pathway involves exocytosis of lysosomes. Expulsion of UPEC through these pathways is thought to benefit the host.
Collapse
Affiliation(s)
- Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Hoan Van Ngo
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
478
|
Roy B, Philippe C, Loessner MJ, Goulet J, Moineau S. Production of Bacteriophages by Listeria Cells Entrapped in Organic Polymers. Viruses 2018; 10:E324. [PMID: 29899227 PMCID: PMC6024803 DOI: 10.3390/v10060324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/29/2022] Open
Abstract
Applications for bacteriophages as antimicrobial agents are increasing. The industrial use of these bacterial viruses requires the production of large amounts of suitable strictly lytic phages, particularly for food and agricultural applications. This work describes a new approach for phage production. Phages H387 (Siphoviridae) and A511 (Myoviridae) were propagated separately using Listeria ivanovii host cells immobilised in alginate beads. The same batch of alginate beads could be used for four successive and efficient phage productions. This technique enables the production of large volumes of high-titer phage lysates in continuous or semi-continuous (fed-batch) cultures.
Collapse
Affiliation(s)
- Brigitte Roy
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, QC G1V OA6, Canada.
- Département des Sciences des Aliments, Faculté des Sciences de L'agriculture et de L'alimentation, Université Laval, Québec, QC G1V OA6, Canada.
- Félix d'Hérelle Reference Center for Bacterial Viruses and GREB, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V OA6, Canada.
| | - Cécile Philippe
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, QC G1V OA6, Canada.
- Félix d'Hérelle Reference Center for Bacterial Viruses and GREB, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V OA6, Canada.
| | - Martin J Loessner
- ETH Zurich, Institute of Food, Nutrition and Health, Schmelzbergstrasse, 7CH-8092 Zürich, Switzerland.
| | - Jacques Goulet
- Département des Sciences des Aliments, Faculté des Sciences de L'agriculture et de L'alimentation, Université Laval, Québec, QC G1V OA6, Canada.
| | - Sylvain Moineau
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, QC G1V OA6, Canada.
- Félix d'Hérelle Reference Center for Bacterial Viruses and GREB, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V OA6, Canada.
| |
Collapse
|
479
|
Castellano P, Pérez Ibarreche M, Longo Borges L, Niño Arias FC, Ross GR, De Martinis ECP. Lactobacillus spp. impair the ability of Listeria monocytogenes FBUNT to adhere to and invade Caco-2 cells. Biotechnol Lett 2018; 40:1237-1244. [PMID: 29948513 DOI: 10.1007/s10529-018-2572-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/18/2018] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The objective of this study was to evaluate the ability of Lactobacillus curvatus CRL705, CRL1532, and CRL1533 and Lactobacillus sakei CRL1613 to survive under simulated gastrointestinal conditions. Moreover, a microencapsulation approach was proposed to improve gastrointestinal survival. Finally, experiments were performed to demonstrate that Lactobacillus spp. can modulate the ability of Listeria monocytogenes FBUNT to adhere to and invade Caco-2 cells. RESULTS Lactobacillus strains were encapsulated in alginate beads to enhance the survival of bacteria under in vitro gastrointestinal conditions. All strains hydrolyzed bile salts using chenodeoxycholic acid as a substrate and adhered to Caco-2 cells. Cell-free supernatants (CFSs) showed antimicrobial activity against L. monocytogenes as demonstrated by agar diffusion assays. The average percentages of L. monocytogenes adhesion decreased from 67.74 to 41.75 and 38.7% in the presence of 50 and 90% (v/v), respectively, for all CFSs tested. The highest concentrations of CFSs completely inhibited the L. monocytogenes invasion of Caco-2 cells. CONCLUSIONS The studied Lactobacillus strains have protective effects against the adhesion and invasion of L. monocytogenes FBUNT. Alginate encapsulation of these bacteria improved gastrointestinal tolerance such that they could be further studied as potential probiotics against intestinal pathogenic bacteria.
Collapse
Affiliation(s)
- P Castellano
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, 4000, Tucumán, Argentina.
| | - M Pérez Ibarreche
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, 4000, Tucumán, Argentina
| | - L Longo Borges
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - USP, Av. do Café s/n, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - F C Niño Arias
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - USP, Av. do Café s/n, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - G R Ross
- Instituto de Biotecnología farmacéutica y alimentaria (INBIOFAL-CONICET), Av Kichner, 4000, Tucumán, Argentina
| | - E C Pereira De Martinis
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - USP, Av. do Café s/n, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| |
Collapse
|
480
|
Siqueira MDS, Ribeiro RDM, Travassos LH. Autophagy and Its Interaction With Intracellular Bacterial Pathogens. Front Immunol 2018; 9:935. [PMID: 29875765 PMCID: PMC5974045 DOI: 10.3389/fimmu.2018.00935] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Cellular responses to stress can be defined by the overwhelming number of changes that cells go through upon contact with and stressful conditions such as infection and modifications in nutritional status. One of the main cellular responses to stress is autophagy. Much progress has been made in the understanding of the mechanisms involved in the induction of autophagy during infection by intracellular bacteria. This review aims to discuss recent findings on the role of autophagy as a cellular response to intracellular bacterial pathogens such as, Streptococcus pyogenes, Mycobacterium tuberculosis, Shigella flexneri, Salmonella typhimurium, Listeria monocytogenes, and Legionella pneumophila, how the autophagic machinery senses these bacteria directly or indirectly (through the detection of bacteria-induced nutritional stress), and how some of these bacterial pathogens manage to escape from autophagy.
Collapse
Affiliation(s)
- Mariana da Silva Siqueira
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato de Moraes Ribeiro
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
481
|
Bierne H, Milohanic E, Kortebi M. To Be Cytosolic or Vacuolar: The Double Life of Listeria monocytogenes. Front Cell Infect Microbiol 2018; 8:136. [PMID: 29868493 PMCID: PMC5962784 DOI: 10.3389/fcimb.2018.00136] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/18/2018] [Indexed: 01/06/2023] Open
Abstract
Intracellular bacterial pathogens are generally classified into two types: those that exploit host membrane trafficking to construct specific niches in vacuoles (i.e., "vacuolar pathogens"), and those that escape from vacuoles into the cytosol, where they proliferate and often spread to neighboring cells (i.e., "cytosolic pathogens"). However, the boundary between these distinct intracellular phenotypes is tenuous and may depend on the timing of infection and on the host cell type. Here, we discuss recent progress highlighting this phenotypic duality in Listeria monocytogenes, which has long been a model for cytosolic pathogens, but now emerges as a bacterium also capable of residing in vacuoles, in a slow/non-growing state. The ability of L. monocytogenes to enter a persistence stage in vacuoles might play a role during the asymptomatic incubation period of listeriosis and/or the carriage of this pathogen in asymptomatic hosts. Moreover, persistent vacuolar Listeria could be less susceptible to antibiotics and more difficult to detect by routine techniques of clinical biology. These hypotheses deserve to be explored in order to better manage the risks related to this food-borne pathogen.
Collapse
Affiliation(s)
- Hélène Bierne
- Epigenetics and Cellular Microbiology Team, Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | | |
Collapse
|
482
|
de Grandi AZ, Pinto UM, Destro MT. Dual-species biofilm of Listeria monocytogenes and Escherichia coli on stainless steel surface. World J Microbiol Biotechnol 2018; 34:61. [PMID: 29651554 DOI: 10.1007/s11274-018-2445-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/09/2018] [Indexed: 11/25/2022]
Abstract
Listeria monocytogenes is a Gram-positive bacterium commonly associated with foodborne diseases. Due its ability to survive under adverse environmental conditions and to form biofilm, this bacterium is a major concern for the food industry, since it can compromise sanitation procedures and increase the risk of post-processing contamination. Little is known about the interaction between L. monocytogenes and Gram-negative bacteria on biofilm formation. Thus, in order to evaluate this interaction, Escherichia coli and L. monocytogenes were tested for their ability to form biofilms together or in monoculture. We also aimed to evaluate the ability of L. monocytogenes 1/2a and its isogenic mutant strain (ΔprfA ΔsigB) to form biofilm in the presence of E. coli. We assessed the importance of the virulence regulators, PrfA and σB, in this process since they are involved in many aspects of L. monocytogenes pathogenicity. Biofilm formation was assessed using stainless steel AISI 304 #4 slides immersed into brain heart infusion broth, reconstituted powder milk and E. coli preconditioned medium at 25 °C. Our results indicated that a higher amount of biofilm was formed by the wild type strain of L. monocytogenes than by its isogenic mutant, indicating that prfA and sigB are important for biofilm development, especially maturation under our experimental conditions. The presence of E. coli or its metabolites in preconditioned medium did not influence biofilm formation by L. monocytogenes. Our results confirm the possibility of concomitant biofilm formation by L. monocytogenes and E. coli, two bacteria of major significance in the food industry.
Collapse
Affiliation(s)
- Aline Zago de Grandi
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580 B.14, 05508-900, São Paulo, SP, Brazil
| | - Uelinton Manoel Pinto
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580 B.14, 05508-900, São Paulo, SP, Brazil.
- Food Research Center (FoRC), Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580 B.14, 05508-900, São Paulo, SP, Brazil.
| | - Maria Teresa Destro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580 B.14, 05508-900, São Paulo, SP, Brazil
| |
Collapse
|
483
|
Listeria Adhesion Protein Induces Intestinal Epithelial Barrier Dysfunction for Bacterial Translocation. Cell Host Microbe 2018; 23:470-484.e7. [PMID: 29606495 DOI: 10.1016/j.chom.2018.03.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/30/2017] [Accepted: 03/09/2018] [Indexed: 12/24/2022]
Abstract
Intestinal epithelial cells are the first line of defense against enteric pathogens, yet bacterial pathogens, such as Listeria monocytogenes, can breach this barrier. We show that Listeria adhesion protein (LAP) induces intestinal epithelial barrier dysfunction to promote bacterial translocation. These disruptions are attributed to the production of pro-inflammatory cytokines TNF-α and IL-6, which is observed in mice challenged with WT and isogenic strains lacking the surface invasion protein Internalin A (ΔinlA), but not a lap- mutant. Additionally, upon engagement of its surface receptor Hsp60, LAP activates canonical NF-κB signaling, facilitating myosin light-chain kinase (MLCK)-mediated opening of the epithelial barrier via cellular redistribution of the epithelial junctional proteins claudin-1, occludin, and E-cadherin. Pharmacological inhibition of MLCK or NF-κB in cells or genetic ablation of MLCK in mice prevents mislocalization of junctional proteins and L. monocytogenes translocation. Thus, L. monocytogenes uses LAP to exploit epithelial defenses and cross the intestinal epithelial barrier.
Collapse
|
484
|
|