1
|
Campos Muzquiz LG, Martínez Gómez D, Reyes Cruz T, Méndez Olvera ET. Evaluation of intracellular survival of Campylobacter fetus subsp. fetus in bovine endometrial cells by qPCR. IRANIAN JOURNAL OF VETERINARY RESEARCH 2021; 22:94-99. [PMID: 34306105 PMCID: PMC8294819 DOI: 10.22099/ijvr.2021.38693.5632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/02/2021] [Accepted: 02/28/2021] [Indexed: 09/30/2022]
Abstract
BACKGROUND Campylobacter fetus subsp. fetus is the causal agent of sporadic abortion and infertility in bovines that produces economic losses in livestock. AIMS This study evaluates the capability of C. fetus subsp. fetus to invade and survive in bovine endometrial epithelial cells and attempts to describe a pathogenic mechanism of this microorganism. METHODS Primary culture of bovine endometrial epithelial cells was challenged with C. fetus subsp. fetus. Intracellular bacteria, represented by the number of genomic copies (g.c.) were quantified at 0, 2, 4, 10, and 24 hours post-infection (h.p.i.), by quantitative polymerase chain reaction (qPCR). The presence of intracellular bacteria was evaluated by immunofluorescence and immunohistochemistry. RESULTS The results showed that only viable C. fetus subsp. fetus could invade endometrial cells. The g.c. number in assays with viable C. fetus subsp. fetus reached an average value of 656 g.c., remained constant until 4 h.p.i., then decreased to 100 g.c, at 24 h.p.i. In assays with non-viable microorganisms, the average value of g.c. was less than 1 g.c. and never changed. The intracellular presence of this bacteria was confirmed at 2 h.p.i. by immunofluorescence and immunohistochemistry. CONCLUSION The results suggest that only C. fetus subsp. fetus viable can invade bovine endometrial epithelial cells but will not replicate in them, indicating that the endometrial cells do not represent a replication niche for this pathogen. Nonetheless, this invasion capability suggests that this type of cell could be employed by the pathogen to spread to other tissues.
Collapse
Affiliation(s)
- L. G. Campos Muzquiz
- Department of Genetics and Virology, Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Mexico
| | - D. Martínez Gómez
- Laboratory of Agricultural Microbiology, Department of Agricultural and Animal Production, Metropolitan Autonomous University-Xochimilco, Mexico City, Mexico
| | - T. Reyes Cruz
- Biological and Health Sciences Division, Department of Agricultural and Animal Production, Metropolitan Autonomous University-Xochimilco, Mexico City, Mexico
| | - E. T. Méndez Olvera
- Laboratory of Molecular Biology, Department of Agricultural and Animal Production, Metropolitan Autonomous University-Xochimilco, Mexico City, Mexico
| |
Collapse
|
2
|
Sachdeva K, Sundaramurthy V. The Interplay of Host Lysosomes and Intracellular Pathogens. Front Cell Infect Microbiol 2020; 10:595502. [PMID: 33330138 PMCID: PMC7714789 DOI: 10.3389/fcimb.2020.595502] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Lysosomes are an integral part of the intracellular defense system against microbes. Lysosomal homeostasis in the host is adaptable and responds to conditions such as infection or nutritional deprivation. Pathogens such as Mycobacterium tuberculosis (Mtb) and Salmonella avoid lysosomal targeting by actively manipulating the host vesicular trafficking and reside in a vacuole altered from the default lysosomal trafficking. In this review, the mechanisms by which the respective pathogen containing vacuoles (PCVs) intersect with lysosomal trafficking pathways and maintain their distinctness are discussed. Despite such active inhibition of lysosomal targeting, emerging literature shows that different pathogens or pathogen derived products exhibit a global influence on the host lysosomal system. Pathogen mediated lysosomal enrichment promotes the trafficking of a sub-set of pathogens to lysosomes, indicating heterogeneity in the host-pathogen encounter. This review integrates recent advancements on the global lysosomal alterations upon infections and the host protective role of the lysosomes against these pathogens. The review also briefly discusses the heterogeneity in the lysosomal targeting of these pathogens and the possible mechanisms and consequences.
Collapse
|
3
|
Liu T, Song X, Khan S, Li Y, Guo Z, Li C, Wang S, Dong W, Liu W, Wang B, Cao H. The gut microbiota at the intersection of bile acids and intestinal carcinogenesis: An old story, yet mesmerizing. Int J Cancer 2019; 146:1780-1790. [DOI: 10.1002/ijc.32563] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Tianyu Liu
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Samiullah Khan
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Yun Li
- Department of Pharmacy, General HospitalTianjin Medical University Tianjin China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Chuqiao Li
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| |
Collapse
|
4
|
Sarkar A, Tindle C, Pranadinata RF, Reed S, Eckmann L, Stappenbeck TS, Ernst PB, Das S. ELMO1 Regulates Autophagy Induction and Bacterial Clearance During Enteric Infection. J Infect Dis 2019; 216:1655-1666. [PMID: 29029244 DOI: 10.1093/infdis/jix528] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macrophages are specialized phagocytic cells involved in clearing invading pathogens. Previously we reported that engulfment and cell motility protein 1 (ELMO1) in macrophages mediates bacterial internalization and intestinal inflammation. Here we studied the role of ELMO1 in the fate of internalized targets. ELMO1 is present in the intracellular vesicles and enhances accumulation of the protein LC3B following engulfment of Salmonella or treatment with autophagy-inducing rapamycin. The protein ATG5 and the kinase ULK1 are involved in classical autophagy, while LC3-associated phagocytosis is ULK1 independent. ATG5 but not ULK1 cooperated with ELMO1 in LC3 accumulation after infection, suggesting the ELMO1 preferentially regulated LC3-associated phagocytosis. Because LC3-associated phagocytosis delivers cargo for degradation, the contribution of ELMO1 to the lysosome degradation pathways was evaluated by studying pH and cathepsin B activity. ELMO1-depleted macrophages showed a time-dependent increase in pH and a decrease in cathepsin B activity associated with bacterial survival. Together, ELMO1 regulates LC3B accumulation and antimicrobial responses involved in the clearance of enteric pathogens. This paper investigated how innate immune pathways involving ELMO1 work in a coordinated fashion to eliminate bacterial threats. ELMO1 is present in the phagosome and enhances bacterial clearance by differential regulation of lysosomal acidification and enzymatic activity.
Collapse
Affiliation(s)
- Arup Sarkar
- Department of Pathology, University of California-San Diego
| | | | | | - Sharon Reed
- Department of Pathology, University of California-San Diego
| | - Lars Eckmann
- Department of Medicine, University of California-San Diego
| | | | - Peter B Ernst
- Department of Pathology, University of California-San Diego
| | - Soumita Das
- Department of Pathology, University of California-San Diego
| |
Collapse
|
5
|
Radomski N, Rebbig A, Leonhardt RM, Knittler MR. Xenophagic pathways and their bacterial subversion in cellular self-defense - παντα ρει - everything is in flux. Int J Med Microbiol 2017; 308:185-196. [PMID: 29126745 DOI: 10.1016/j.ijmm.2017.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/09/2023] Open
Abstract
Autophagy is an evolutionarily ancient and highly conserved eukaryotic mechanism that targets cytoplasmic material for degradation. Autophagic flux involves the formation of autophagosomes and their degradation by lysosomes. The process plays a crucial role in maintaining cellular homeostasis and responds to various environmental conditions. While autophagy had previously been thought to be a non-selective process, it is now clear that it can also selectively target cellular organelles, such as mitochondria (referred to as mitophagy) and/or invading pathogens (referred to as xenophagy). Selective autophagy is characterized by specific substrate recognition and requires distinct cellular adaptor proteins. Here we review xenophagic mechanisms involved in the recognition and autolysosomal or autophagolysosomal degradation of different intracellular bacteria. In this context, we also discuss a recently discovered cellular self-defense pathway, termed mito-xenophagy, which occurs during bacterial infection of dendritic cells and depends on a TNF-α-mediated metabolic switch from oxidative phosphorylation to glycolysis.
Collapse
Affiliation(s)
- Nadine Radomski
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Annica Rebbig
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Ralf M Leonhardt
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Michael R Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany.
| |
Collapse
|
6
|
Nie YF, Hu J, Yan XH. Cross-talk between bile acids and intestinal microbiota in host metabolism and health. J Zhejiang Univ Sci B 2016; 16:436-46. [PMID: 26055905 DOI: 10.1631/jzus.b1400327] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bile acid (BA) is de novo synthesized exclusively in the liver and has direct or indirect antimicrobial effects. On the other hand, the composition and size of the BA pool can be altered by intestinal microbiota via the biotransformation of primary BAs to secondary BAs, and subsequently regulate the nuclear farnesoid X receptor (FXR; NR1H4). The BA-activated FXR plays important roles in BA synthesis and metabolism, glucose and lipid metabolism, and even hepatic autophagy. BAs can also play a role in the interplays among intestinal microbes. In this review, we mainly discuss the interactions between BAs and intestinal microbiota and their roles in regulating host metabolism, and probably the autophagic signaling pathway.
Collapse
Affiliation(s)
- Yang-fan Nie
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | | | | |
Collapse
|
7
|
Banerjee S, Gummadidala PM, Rima RA, Ahmed RU, Kenne GJ, Mitra C, Gomaa OM, Hill J, McFadden S, Banaszek N, Fayad R, Terejanu G, Chanda A. Quantitative acoustic contrast tomography reveals unique multiscale physical fluctuations during aflatoxin synthesis in Aspergillus parasiticus. Fungal Genet Biol 2014; 73:61-8. [PMID: 25312859 DOI: 10.1016/j.fgb.2014.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/01/2014] [Accepted: 10/06/2014] [Indexed: 01/14/2023]
Abstract
Fungal pathogens need regulated mechanical and morphological fine-tuning for pushing through substrates to meet their metabolic and functional needs. Currently very little is understood on how coordinated colony level morphomechanical modifications regulate their behavior. This is due to an absence of a method that can simultaneously map, quantify, and correlate global fluctuations in physical properties of the expanding fungal colonies. Here, we show that three-dimensional ultrasonic reflections upon decoding can render acoustic contrast tomographs that contain information on material property and morphology in the same time scale of one important phytopathogen, Aspergillus parasiticus, at multiple length scales. By quantitative analysis of the changes in acoustic signatures collected as the A. parasiticus colony expands with time, we further demonstrate that the pathogen displays unique acoustic signatures during synthesis and release of its hepatocarcinogenic secondary metabolite, aflatoxin, suggesting an involvement of a multiscale morphomechanical reorganization of the colony in this process. Our studies illustrate for the first time, the feasibility of generating in any invading cell population, four-dimensional maps of global physical properties, with minimal physical perturbation of the specimens. Our developed method that we term quantitative acoustic contrast tomography (Q-ACT), provides a novel diagnostic framework for the identification of in-cell molecular factors and discovery of small molecules that may modulate pathogen invasion in a host.
Collapse
Affiliation(s)
- Sourav Banerjee
- Integrated Material Assessment and Predictive Simulation Laboratory (i-MAPS), Department of Mechanical Engineering, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States.
| | - Phani M Gummadidala
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States
| | - Rowshan A Rima
- Integrated Material Assessment and Predictive Simulation Laboratory (i-MAPS), Department of Mechanical Engineering, University of South Carolina, Columbia, SC 29208, United States
| | - Riaz U Ahmed
- Integrated Material Assessment and Predictive Simulation Laboratory (i-MAPS), Department of Mechanical Engineering, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States
| | - Gabriel J Kenne
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Chandrani Mitra
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States
| | - Ola M Gomaa
- Microbiology Department, National Center for Radiation Research and Technology, Cairo, Egypt
| | - Jasmine Hill
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Sandra McFadden
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Nora Banaszek
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Raja Fayad
- Department of Exercise Science, Arnold School of Public Health University of South Carolina, Columbia, SC 29208, United States; Center for Colon Cancer Research, University of South Carolina, United States
| | - Gabriel Terejanu
- Department of Computer Science and Engineering, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States
| | - Anindya Chanda
- Laboratory of Fungal Pathogenesis and Secondary Metabolism, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States; Center for Uncertainty Driven Computational Biomechanics, University of South Carolina, Columbia, SC 29208, United States.
| |
Collapse
|
8
|
Bowden SD, Hopper-Chidlaw AC, Rice CJ, Ramachandran VK, Kelly DJ, Thompson A. Nutritional and metabolic requirements for the infection of HeLa cells by Salmonella enterica serovar Typhimurium. PLoS One 2014; 9:e96266. [PMID: 24797930 PMCID: PMC4010460 DOI: 10.1371/journal.pone.0096266] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 04/07/2014] [Indexed: 12/04/2022] Open
Abstract
Salmonella is the causative agent of a spectrum of human and animal diseases ranging from gastroenteritis to typhoid fever. It is a food - and water - borne pathogen and infects via ingestion followed by invasion of intestinal epithelial cells and phagocytic cells. In this study we employed a mutational approach to define the nutrients and metabolic pathways required by Salmonella enterica serovar Typhimurium during infection of a human epithelial cell line (HeLa). We deleted the key glycolytic genes, pfkA and pfkB to show that S. Typhimurium utilizes glycolysis for replication within HeLa cells; however, glycolysis was not absolutely essential for intracellular replication. Using S. Typhimurium strains deleted for genes encoding components of the phosphotransferase system and glucose transport, we show that glucose is a major substrate required for the intracellular replication of S. Typhimurium in HeLa cells. We also deleted genes encoding enzymes involved in the utilization of gluconeogenic substrates and the glyoxylate shunt and show that neither of these pathways were required for intracellular replication of S. Typhimurium within HeLa cells.
Collapse
Affiliation(s)
- Steven D. Bowden
- Institute of Food Research, Norwich Research Park, Colney, Norwich, United Kingdom
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | | | | | - Vinoy K. Ramachandran
- Institute of Food Research, Norwich Research Park, Colney, Norwich, United Kingdom
- Department of Plant Sciences, University of Oxford, Oxford, United Kingdom
| | - David J. Kelly
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Arthur Thompson
- Institute of Food Research, Norwich Research Park, Colney, Norwich, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Abstract
Autophagy is used by the cell to degrade various substrates; this is achieved either through the canonical, non-selective autophagy pathway or through selective autophagy. Both pathways proceed via distinct key steps and use specific molecular mechanisms. The canonical autophagy pathway has been studied in detail in mammalian cells and in model organisms, such as yeast. The molecular mechanisms underlying non-canonical autophagy, in addition to alternative pathways that are independent of some of the key autophagy machinery, are beginning to become clear. Besides degradation of cellular proteins, autophagy proteins are also involved in many other functions, some of which are important during bacterial infections. Autophagy functions as an antibacterial mechanism. The induction and recognition mechanisms for several bacterial species have been elucidated. Bacteria can escape killing by autophagy and some can even use autophagy to promote infection of host cells, through the interaction between bacterial effector proteins and autophagy components. The knowledge about bacteria–autophagy interactions will inform the design of new drugs and treatments against bacterial infections.
Autophagy not only degrades components of host cells but can also target intracellular bacteria and thus contribute to host defences. Here, Huang and Brumell discuss the canonical and selective pathways of antibacterial autophagy, as well as the ways in which bacteria can escape from them and sometimes even use them to promote infection. Autophagy is a cellular process that targets proteins, lipids and organelles to lysosomes for degradation, but it has also been shown to combat infection with various pathogenic bacteria. In turn, bacteria have developed diverse strategies to avoid autophagy by interfering with autophagy signalling or the autophagy machinery and, in some cases, they even exploit autophagy for their growth. In this Review, we discuss canonical and non-canonical autophagy pathways and our current knowledge of antibacterial autophagy, with a focus on the interplay between bacterial factors and autophagy components.
Collapse
|
10
|
Pareja MEM, Colombo MI. Autophagic clearance of bacterial pathogens: molecular recognition of intracellular microorganisms. Front Cell Infect Microbiol 2013; 3:54. [PMID: 24137567 PMCID: PMC3786225 DOI: 10.3389/fcimb.2013.00054] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/02/2013] [Indexed: 01/01/2023] Open
Abstract
Autophagy is involved in several physiological and pathological processes. One of the key roles of the autophagic pathway is to participate in the first line of defense against the invasion of pathogens, as part of the innate immune response. Targeting of intracellular bacteria by the autophagic machinery, either in the cytoplasm or within vacuolar compartments, helps to control bacterial proliferation in the host cell, controlling also the spreading of the infection. In this review we will describe the means used by diverse bacterial pathogens to survive intracellularly and how they are recognized by the autophagic molecular machinery, as well as the mechanisms used to avoid autophagic clearance.
Collapse
Affiliation(s)
- Maria Eugenia Mansilla Pareja
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Médicas, Instituto de Histología y Embriología-CONICET, Universidad Nacional de Cuyo Mendoza, Argentina
| | | |
Collapse
|
11
|
In vitro prevention of salmonella lipopolysaccharide-induced damages in epithelial barrier function by various lactobacillus strains. Gastroenterol Res Pract 2013; 2013:973209. [PMID: 23840201 PMCID: PMC3690232 DOI: 10.1155/2013/973209] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 05/07/2013] [Indexed: 01/01/2023] Open
Abstract
Background. Lactobacillus shows beneficial anti-inflammatory effects on Salmonella infection. The maintenance of the tight junction (TJ) integrity plays an importance role in avoiding bacterial invasion. Whether Lactobacillus could be used to regulate the TJ protein expression and distribution in inflamed intestinal epithelial cells was determined. Methods. Using the transwell coculture model, Salmonella lipopolysaccharide (LPS) was apically added to polarized Caco-2 cells cocultured with peripheral blood mononuclear cells in the basolateral compartment. LPS-stimulated Caco-2 cells were incubated with various Lactobacillus strains. TJ integrity was determined by measuring transepithelial electrical resistance across Caco-2 monolayer. Expression and localization of TJ proteins (zonula-occludens- (ZO-) 1) were determined by Western blot and immunofluorescence microscopy. Results. Various strains of Lactobacillus were responsible for the different modulations of cell layer integrity. LPS was specifically able to disrupt epithelial barrier and change the location of ZO-1. Our data demonstrate that Lactobacillus could attenuate the barrier disruption of intestinal epithelial cells caused by Salmonella LPS administration. We showed that Lactobacillus strains are associated with the maintenance of the tight junction integrity and appearance. Conclusion. In this study we provide insight that live probiotics could improve epithelial barrier properties and this may explain the potential mechanism behind their beneficial effect in vivo.
Collapse
|
12
|
Role for neutrophils in host immune responses and genetic factors that modulate resistance to Salmonella enterica serovar typhimurium in the inbred mouse strain SPRET/Ei. Infect Immun 2010; 78:3848-60. [PMID: 20643856 DOI: 10.1128/iai.00044-10] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Infection with Salmonella enterica serovar Typhimurium is a complex disease in which the host-bacterium interactions are strongly influenced by genetic factors of the host. We demonstrate that SPRET/Ei, an inbred mouse strain derived from Mus spretus, is resistant to S. Typhimurium infections. The kinetics of bacterial proliferation, as well as histological examinations of tissue sections, suggest that SPRET/Ei mice can control bacterial multiplication and spreading despite significant attenuation of the cytokine response. The resistance of SPRET/Ei mice to S. Typhimurium infection is associated with increased leukocyte counts in the circulation and enhanced neutrophil influx into the peritoneum during the course of infection. A critical role of neutrophils was confirmed by neutrophil depletion: neutropenic SPRET/Ei mice were sensitive to infection with S. Typhimurium and showed much higher bacterial loads. To identify genes that modulate the natural resistance of SPRET/Ei mice to S. Typhimurium infection, we performed a genome-wide study using an interspecific backcross between C3H/HeN and SPRET/Ei mice. The results of this analysis demonstrate that at least two loci, located on chromosomes 6 and 11, affect survival following lethal infection with S. Typhimurium. These two loci contain several interesting candidate genes which may have important implications for the search for genetic factors controlling Salmonella infections in humans and for our understanding of complex host-pathogen interactions in general.
Collapse
|
13
|
Abstract
Autophagy is an innate immune defense mechanism against various intracellular bacterial pathogens, such as Salmonella enterica serovar Typhimurium (S. typhimurium), Listeria monocytogenes and Shigella flexneri. S. typhimurium uses type three secretion systems (T3SSs) to invade mammalian cells and replicate in Salmonella-containing vacuoles (SCVs). A small population of intracellular S. typhimurium is targeted by autophagy shortly after infection. Evidence suggests that these bacteria are present within SCVs that have been damaged by high levels of T3SS activity. Autophagy limits the growth of S. typhimurium in host cells. Therefore, autophagy can be considered to protect the cytosol of eukaryotic cells from bacterial colonization. L. monocytogenes secretes the pore-forming cytolysin listeriolysin O (LLO) to disrupt the phagosome and escape into the cytosol, where it acquires actin-based motility. Autophagy can target L. monocytogenes in the cytosol under specific experimental conditions. However, L. monocytogenes utilizes several virulence factors to evade being killed by the autophagy system. A newly appreciated population of L. monocytogenes undergoes slow growth in specialized vacuoles termed spacious Listeria-containing phagosomes (SLAPs), the formation of which requires bacterial LLO and host autophagy. In the cytosol, S. flexneri can also be a target for autophagy in the absence of a T3SS effector, IcsB, that normally impairs the interaction between Atg5 and wild-type bacteria. Therefore, autophagy can recognize intracellular bacteria in a variety of ways, leading to different fates for these bacteria in host cells. The inefficient autophagy of enteric bacteria in genetically compromised individuals may contribute to the pathogenesis of Crohn's disease.
Collapse
Affiliation(s)
- Ju Huang
- Cell Biology Program, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | |
Collapse
|
14
|
Arnold R, Brandmaier S, Kleine F, Tischler P, Heinz E, Behrens S, Niinikoski A, Mewes HW, Horn M, Rattei T. Sequence-based prediction of type III secreted proteins. PLoS Pathog 2009; 5:e1000376. [PMID: 19390696 PMCID: PMC2669295 DOI: 10.1371/journal.ppat.1000376] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 03/11/2009] [Indexed: 12/30/2022] Open
Abstract
The type III secretion system (TTSS) is a key mechanism for host cell interaction used by a variety of bacterial pathogens and symbionts of plants and animals including humans. The TTSS represents a molecular syringe with which the bacteria deliver effector proteins directly into the host cell cytosol. Despite the importance of the TTSS for bacterial pathogenesis, recognition and targeting of type III secreted proteins has up until now been poorly understood. Several hypotheses are discussed, including an mRNA-based signal, a chaperon-mediated process, or an N-terminal signal peptide. In this study, we systematically analyzed the amino acid composition and secondary structure of N-termini of 100 experimentally verified effector proteins. Based on this, we developed a machine-learning approach for the prediction of TTSS effector proteins, taking into account N-terminal sequence features such as frequencies of amino acids, short peptides, or residues with certain physico-chemical properties. The resulting computational model revealed a strong type III secretion signal in the N-terminus that can be used to detect effectors with sensitivity of approximately 71% and selectivity of approximately 85%. This signal seems to be taxonomically universal and conserved among animal pathogens and plant symbionts, since we could successfully detect effector proteins if the respective group was excluded from training. The application of our prediction approach to 739 complete bacterial and archaeal genome sequences resulted in the identification of between 0% and 12% putative TTSS effector proteins. Comparison of effector proteins with orthologs that are not secreted by the TTSS showed no clear pattern of signal acquisition by fusion, suggesting convergent evolutionary processes shaping the type III secretion signal. The newly developed program EffectiveT3 (http://www.chlamydiaedb.org) is the first universal in silico prediction program for the identification of novel TTSS effectors. Our findings will facilitate further studies on and improve our understanding of type III secretion and its role in pathogen-host interactions.
Collapse
Affiliation(s)
- Roland Arnold
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Stefan Brandmaier
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Frederick Kleine
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Patrick Tischler
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Eva Heinz
- Institute for Bioinformatics and Systems Biology (MIPS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Sebastian Behrens
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Antti Niinikoski
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| | - Hans-Werner Mewes
- Institute for Bioinformatics and Systems Biology (MIPS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Matthias Horn
- University of Vienna, Department of Microbial Ecology, Vienna, Austria
| | - Thomas Rattei
- Technische Universität München, Department of Genome Oriented Bioinformatics, Wissenschaftszentrum Weihenstephan, Freising, Germany
| |
Collapse
|
15
|
Salyers AA, Moon K, Schlessinger D. The human intestinal tract – a hotbed of resistance gene transfer? Part II. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.clinmicnews.2007.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Lawley TD, Chan K, Thompson LJ, Kim CC, Govoni GR, Monack DM. Genome-wide screen for Salmonella genes required for long-term systemic infection of the mouse. PLoS Pathog 2006; 2:e11. [PMID: 16518469 PMCID: PMC1383486 DOI: 10.1371/journal.ppat.0020011] [Citation(s) in RCA: 277] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Accepted: 01/11/2006] [Indexed: 11/20/2022] Open
Abstract
A microarray-based negative selection screen was performed to identify Salmonella enterica serovar Typhimurium (serovar Typhimurium) genes that contribute to long-term systemic infection in 129X1/SvJ (Nramp1r) mice. A high-complexity transposon-mutagenized library was used to infect mice intraperitoneally, and the selective disappearance of mutants was monitored after 7, 14, 21, and 28 d postinfection. One hundred and eighteen genes were identified to contribute to serovar Typhimurium infection of the spleens of mice by 28 d postinfection. The negatively selected mutants represent many known aspects of Salmonella physiology and pathogenesis, although the majority of the identified genes are of putative or unknown function. Approximately 30% of the negatively selected genes correspond to horizontally acquired regions such as those within Salmonella pathogenicity islands (SPI 1–5), prophages (Gifsy-1 and −2 and remnant), and the pSLT virulence plasmid. In addition, mutations in genes responsible for outer membrane structure and remodeling, such as LPS- and PhoP-regulated and fimbrial genes, were also selected against. Competitive index experiments demonstrated that the secreted SPI2 effectors SseK2 and SseJ as well as the SPI4 locus are attenuated relative to wild-type bacteria during systemic infection. Interestingly, several SPI1-encoded type III secretion system effectors/translocases are required by serovar Typhimurium to establish and, unexpectedly, to persist systemically, challenging the present description of Salmonella pathogenesis. Moreover, we observed a progressive selection against serovar Typhimurium mutants based upon the duration of the infection, suggesting that different classes of genes may be required at distinct stages of infection. Overall, these data indicate that Salmonella long-term systemic infection in the mouse requires a diverse repertoire of virulence factors. This diversity of genes presumably reflects the fact that bacteria sequentially encounter a variety of host environments and that Salmonella has evolved to respond to these selective forces in a way that permits both the bacteria and the host to survive. Bacteria belonging to the genus Salmonella are capable of establishing a long-term systemic infection in a variety of hosts, including humans, rodents, fowl, and cattle. The ability of Salmonella to subvert the active immune response of the host represents millions of years of co-evolution and is the result of specialized virulence factors that promote long-term infection. This study describes a microarray-based genome-wide screen designed to identify genes required by Salmonella enterica serovar Typhimurium (serovar Typhimurium) to persist and replicate in the spleen and liver of mice for up to 28 days. The results demonstrate that serovar Typhimurium utilizes a diverse repertoire of virulence factors, including both known and novel virulence genes, to establish infection and to persist in the host. The authors' data further established a previously unappreciated role for Salmonella pathogenicity island 1 in maintaining a persistent systemic infection. In addition, a progressive selection against serovar Typhimurium mutants based upon the duration of the infection was observed, suggesting that certain classes of genes are required at specific times during infection and providing a foundation to further dissect Salmonella pathogenesis into distinct temporal phases.
Collapse
Affiliation(s)
- Trevor D Lawley
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Alemán A, Rodríguez-Escudero I, Mallo GV, Cid VJ, Molina M, Rotger R. The amino-terminal non-catalytic region of Salmonella typhimurium SigD affects actin organization in yeast and mammalian cells. Cell Microbiol 2005; 7:1432-46. [PMID: 16153243 DOI: 10.1111/j.1462-5822.2005.00568.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The internalization of Salmonella into epithelial cells relies on the function of bacterial proteins which are injected into the cell by a specialized type III secretion system. Such bacterial effectors interfere with host cell signalling and induce local cytoskeletal rearrangements. One of such effectors is SigD/SopB, which shares homology with mammalian inositol phosphatases. We made use of the Saccharomyces cerevisiae model for elucidating new aspects of SigD function. Endogenous expression of SigD in yeast caused severe growth inhibition. Surprisingly, sigD alleles mutated in the catalytic site or even deleted for the whole C-terminal phosphatase domain still inhibited yeast growth by inducing loss of actin polarization and precluding the budding process. Accordingly, when expressed in HeLa cells, the same sigD alleles lost the ability of depleting phosphatidylinositol 4,5-bisphosphate from the plasma membrane, but still caused disappearance of actin fibres and loss of adherence. We delineate a region of 25 amino acids (residues 118-142) that is necessary for the effect of SigD on actin in HeLa cells. Our data indicate that SigD exerts a toxic effect linked to its N-terminal region and independent of its phosphatase activity.
Collapse
Affiliation(s)
- Ainel Alemán
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| | | | | | | | | | | |
Collapse
|
18
|
Intracellular Voyeurism: Examining the Modulation of Host Cell Activities bySalmonella enterica Serovar Typhimurium. EcoSal Plus 2005; 1. [PMID: 26443522 DOI: 10.1128/ecosalplus.2.2.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Salmonella spp. can infect host cells by gaining entry through phagocytosis or by inducing host cell membrane ruffling that facilitates bacterial uptake. With its wide host range, Salmonella enterica serovar Typhimurium has proven to be an important model organism for studying intracellular bacterial pathogenesis. Upon entry into host cells, serovar Typhimurium typically resides within a membrane-bound compartment termed the Salmonella-containing vacuole (SCV). From the SCV, serovar Typhimurium can inject several effector proteins that subvert many normal host cell systems, including endocytic trafficking, cytoskeletal rearrangements, lipid signaling and distribution, and innate and adaptive host defenses. The study of these intracellular events has been made possible through the use of various imaging techniques, ranging from classic methods of transmission electron microscopy to advanced livecell fluorescence confocal microscopy. In addition, DNA microarrays have now been used to provide a "snapshot" of global gene expression in serovar Typhimurium residing within the infected host cell. This review describes key aspects of Salmonella-induced subversion of host cell activities, providing examples of imaging that have been used to elucidate these events. Serovar Typhimurium engages specific host cell machinery from initial contact with the host cell to replication within the SCV. This continuous interaction with the host cell has likely contributed to the extensive arsenal that serovar Typhimurium now possesses, including two type III secretion systems, a range of ammunition in the form of TTSS effectors, and a complex genetic regulatory network that coordinates the expression of hundreds of virulence factors.
Collapse
|
19
|
Rhee SJ, Walker WA, Cherayil BJ. Developmentally regulated intestinal expression of IFN-gamma and its target genes and the age-specific response to enteric Salmonella infection. THE JOURNAL OF IMMUNOLOGY 2005; 175:1127-36. [PMID: 16002714 DOI: 10.4049/jimmunol.175.2.1127] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Young infants are highly susceptible to systemic dissemination of enteric pathogens such as Salmonella typhimurium when compared with older individuals. The mechanisms underlying this differential susceptibility have not been defined clearly. To better understand this phenomenon, we examined the responses of adult mice and preweaned pups to oral infection by S. typhimurium. We found clear age-specific differences, namely, an attenuated intestinal inflammatory response and a higher systemic bacterial burden in the pups compared with the adults. To elucidate the molecular basis for these differences, we obtained a microarray-based profile of gene expression in the small intestines of uninfected adult and preweaned animals. The results indicated a striking age-dependent increase in the intestinal expression of a number of IFN-gamma-regulated genes involved in antimicrobial defense. This finding was confirmed by real-time quantitative PCR, which also demonstrated an age-dependent increase in intestinal expression of IFN-gamma. The developmental up-regulation of the IFN-gamma-regulated genes was dependent on both IFN-gamma and a normal commensal microflora, as indicated by experiments in IFN-gamma-knockout mice and germfree mice, respectively. However, the increase in expression of IFN-gamma itself was independent of the commensal flora. The functional importance of IFN-gamma in the immunological maturation of the intestine was confirmed by the observation that the response of adult IFN-gamma-knockout animals to S. typhimurium infection resembled that of the wild-type pups. Our findings thus reveal a novel role for IFN-gamma in the developmental regulation of antimicrobial responses in the intestine.
Collapse
MESH Headings
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/deficiency
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/physiology
- Aging/genetics
- Aging/immunology
- Animals
- Animals, Newborn
- Enteritis/genetics
- Enteritis/immunology
- Enteritis/microbiology
- Enteritis/pathology
- Gene Expression Regulation, Developmental/immunology
- Germ-Free Life
- Immunity, Innate/genetics
- Interferon-gamma/biosynthesis
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/pathology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/microbiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Salmonella Infections, Animal/genetics
- Salmonella Infections, Animal/immunology
- Salmonella Infections, Animal/microbiology
- Salmonella Infections, Animal/pathology
- Salmonella typhimurium/growth & development
- Salmonella typhimurium/immunology
Collapse
Affiliation(s)
- Sue J Rhee
- Mucosal Immunology Laboratory, Pediatric Gastroenterology and Nutrition Unit, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
20
|
Smith AC, Cirulis JT, Casanova JE, Scidmore MA, Brumell JH. Interaction of the Salmonella-containing vacuole with the endocytic recycling system. J Biol Chem 2005; 280:24634-41. [PMID: 15886200 DOI: 10.1074/jbc.m500358200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Upon entry of the pathogen Salmonella enterica serovar Typhimurium into host cells, the majority of bacteria reside in a membrane-bound compartment called the Salmonella-containing vacuole (SCV). Previous studies have established that the SCV transiently interacts with early endosomes but only acquires a subset of late endosomal/lysosomal proteins. However, the complete set of interactions between the SCV and the endocytic machinery has yet to be characterized. In this study, we have shown that four characterized regulators of endocytic recycling were present on the SCV after invasion. Interaction kinetics were different for each of the regulators; ARF6 and Rab4 associated immediately, but their presence was diminished 60 min post-infection, whereas syntaxin13 and Rab11 association peaked at 60 min. Using a dominant negative approach, we determined that Rab11 regulates the recycling of CD44 from the vacuole but had no effect on major histocompatibility complex (MHC) class I recycling. In contrast, syntaxin13 regulated the recycling of MHC class I but not of CD44. We also determined that maturation of the SCV, measured by the acquisition of lysosomal associated membrane protein-1, slowed when recycling was impaired. These findings suggest that protein movement through the endocytic recycling system is regulated through at least two concurrent pathways and that efficient interaction with these pathways is necessary for maturation of the Salmonella-containing vacuole. We also demonstrate the utility of using Salmonella invasion as a model of endosomal recycling events.
Collapse
Affiliation(s)
- Adam C Smith
- Infection, Immunity, Injury, and Repair Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | |
Collapse
|
21
|
Reinicke AT, Hutchinson JL, Magee AI, Mastroeni P, Trowsdale J, Kelly AP. A Salmonella typhimurium effector protein SifA is modified by host cell prenylation and S-acylation machinery. J Biol Chem 2005; 280:14620-7. [PMID: 15710609 DOI: 10.1074/jbc.m500076200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
SifA is a Salmonella effector protein that is required for maintenance of the vacuolar membrane that surrounds replicating bacteria. It associates with the Salmonella-containing vacuole but how it interacts with the membrane is unknown. Here we show by immunofluorescence, S100 fractionation and Triton X-114 partitioning that the membrane association and targeting properties of SifA are influenced by a motif encoded within the C-terminal six amino acids. This sequence shares homology with both CAAX and Rab geranylgeranyl transferase prenylation motifs. We characterized the post-translational processing of SifA and showed that the cysteine residue within the CAAX motif is modified by isoprenoid addition through the action of protein geranylgeranyl transferase I. SifA was additionally modified by S-acylation of an adjacent cysteine residue. Similar modifications to host cell proteins regulate numerous functions including protein targeting, membrane association, protein-protein interaction, and signal transduction. This is the only known example of a bacterial effector protein that is modified both by mammalian cell S-acylation and prenylation machinery.
Collapse
Affiliation(s)
- Anna T Reinicke
- Division of Immunology, Department of Pathology, Center for Veterinary Science, University of Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
22
|
Huang FC, Werne A, Li Q, Galyov EE, Walker WA, Cherayil BJ. Cooperative interactions between flagellin and SopE2 in the epithelial interleukin-8 response to Salmonella enterica serovar typhimurium infection. Infect Immun 2004; 72:5052-62. [PMID: 15321998 PMCID: PMC517448 DOI: 10.1128/iai.72.9.5052-5062.2004] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Flagellin is an important stimulus for epithelial interleukin-8 (IL-8) secretion because of its ability to activate Toll-like receptor 5 (TLR5). SopE2, a Salmonella guanine nucleotide exchange factor (GEF), is also involved in intestinal inflammation. To clarify the proinflammatory mechanisms of these proteins, we examined their effects on IL-8 secretion and intracellular signaling in T84 epithelial cells. A Salmonella strain lacking SopE2 (and its homolog SopE) induced lower levels of IL-8 than the wild type and exhibited reduced activation of mitogen-activated protein kinases (MAPKs). Overexpression of wild-type SopE2 in this strain restored MAPK activation and augmented IL-8 production, whereas a mutant lacking GEF activity failed to increase IL-8 expression. Additional effects on signaling were demonstrated in transient transfection experiments, in which SopE2 enhanced the ability of TRAF6, a signal transducer downstream of TLR5, to activate the NF-kappaB transcription factor in 293 cells. Flagellin was also found to be required for IL-8 induction in T84 cells. In its absence, the ability of SopE2 overexpression to increase IL-8 secretion was impaired. Part of this impairment was related to the decreased motility of the flagellin-deficient strain, but lack of flagellin also affected translocation of SopE2 into the infected cells. Our results indicate that flagellin and SopE2 interact functionally at multiple levels to increase IL-8 secretion by epithelial cells-flagellin facilitating the translocation of SopE2, and SopE2 enhancing signaling pathways activated by flagellin. These observations offer a mechanistic explanation for the involvement of these proteins in the pathogenesis of Salmonella-induced gastroenteritis.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Mucosal Immunology Laboratory, Pediatric Gastroenterology and Nutrition Unit, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | | | | | | | | | |
Collapse
|
23
|
Sha J, Fadl AA, Klimpel GR, Niesel DW, Popov VL, Chopra AK. The two murein lipoproteins of Salmonella enterica serovar Typhimurium contribute to the virulence of the organism. Infect Immun 2004; 72:3987-4003. [PMID: 15213144 PMCID: PMC427434 DOI: 10.1128/iai.72.7.3987-4003.2004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Septic shock due to Salmonella and other gram-negative enteric pathogens is a leading cause of death worldwide. The role of lipopolysaccharide in sepsis is well studied; however, the contribution of other bacterial outer membrane components, such as Braun (murein) lipoprotein (Lpp), is not well defined. The genome of Salmonella enterica serovar Typhimurium harbors two copies of the lipoprotein (lpp) gene. We constructed a serovar Typhimurium strain with deletions in both copies of the lpp gene (lpp1 and lpp2) by marker exchange mutagenesis. The integrity of the cell membrane and the secretion of the effector proteins through the type III secretion system were not affected in the lpp double-knockout mutant. Subsequently, the virulence potential of this mutant was examined in a cell culture system using T84 intestinal epithelial and RAW264.7 macrophage cell lines and a mouse model of salmonellosis. The lpp double-knockout mutant was defective in invading and inducing cytotoxic effects in T84 and RAW264.7 cells, although binding of the mutant to the host cell was not affected when compared to the wild-type (WT) serovar Typhimurium. The motility of the mutant was impaired, despite the finding that the number of flagella was similar in the lpp double knockout mutant and the WT serovar Typhimurium. Deletion in the lpp genes did not affect the intracellular survival and replication of Salmonella in macrophages and T84 cells. Induction of the proinflammatory cytokines tumor necrosis factor alpha and interleukin-8 (IL-8) was significantly reduced in macrophages and T84 cells infected with the lpp double-knockout mutant. The levels of IL-8 remained unaffected in T84 cells when infected with either live or heat-killed WT and lpp mutant, indicating that invasion was not required for IL-8 production and that Toll-like receptor 2 signaling might be affected in the Lpp double-knockout mutant. These effects of the Lpp protein could be restored by complementation of the isogenic mutant. The lpp double-knockout mutant was avirulent in mice, and animals infected with this mutant were protected from a lethal challenge dose of WT serovar Typhimurium. The severe combined immunodeficient mice, on the other hand, were susceptible to infection by the lpp double-knockout mutant. The serovar Typhimurium mutants from which only one of the lpp (lpp1 or lpp2) genes was deleted were also avirulent in mice. Taken together, our data indicated that Lpp specifically contributed to the virulence of the organism.
Collapse
Affiliation(s)
- J Sha
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, USA
| | | | | | | | | | | |
Collapse
|
24
|
Takaya A, Suzuki A, Kikuchi Y, Eguchi M, Isogai E, Tomoyasu T, Yamamoto T. Derepression of Salmonella pathogenicity island 1 genes within macrophages leads to rapid apoptosis via caspase-1- and caspase-3-dependent pathways. Cell Microbiol 2004; 7:79-90. [PMID: 15617525 DOI: 10.1111/j.1462-5822.2004.00435.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Salmonella enterica serovar Typhimurium has been reported to induce apoptosis in infected macrophages within 14 h from the time of infection by a caspase-1-dependent mechanism. Here, we demonstrate that depletion of Lon protease in serovar Typhimurium induces rapid and massive apoptosis in macrophages by a mechanism involving both caspases-1 and -3. This excessive induction of apoptosis was abrogated by disruption of invF, which is required for the expression of the Salmonella pathogenicity island 1 (SPI1) genes. Expression of hilA, a central regulator of SPI1 transcription, was repressed in the macrophages after phagocytosis, but this gene was continuously expressed when the DeltaLon mutant grew within the macrophages, so the SPI1 proteins accumulated. Thus, the increase in macrophage apoptosis induced by the DeltaLon mutant could result from continued expression of SPI1 genes under conditions where they are normally repressed. Once Salmonella has established a systemic infection, excess apoptosis of macrophages cells upon which the organism is reliant would be detrimental to the pathogen. Therefore, the Lon protease may be required to suppress apoptosis sufficiently to allow time for the bacterium to replicate, escape and invade new macrophages.
Collapse
Affiliation(s)
- Akiko Takaya
- Department of Microbiology and Molecular Genetics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Norimatsu M, Chance V, Dougan G, Howard CJ, Villarreal-Ramos B. Live Salmonella enterica serovar Typhimurium (S. Typhimurium) elicit dendritic cell responses that differ from those induced by killed S. Typhimurium. Vet Immunol Immunopathol 2004; 98:193-201. [PMID: 15010228 DOI: 10.1016/j.vetimm.2003.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 12/05/2003] [Accepted: 12/05/2003] [Indexed: 11/16/2022]
Abstract
The immune response of bovine monocytes-derived dendritic cells (DC) exposed to either live or killed Salmonella enterica serovar Typhimurium was compared. Both live and killed bacteria induced changes in morphology with distinctive formation of processes and up-regulation of the ability of DC to stimulate allogeneic T-cell proliferation. Also, both live and killed bacteria up-regulated the expression of MHC-I, MHC-II and CD80. However, live bacteria induced greater up-regulation of the expression of CD40 and CD86 than killed bacteria. Live bacteria also induced greater up-regulation of transcription for IL-6, IL-12 and GM-CSF than killed bacteria as measured by quantitative RT-PCR. These data suggest that blood-monocyte-derived DC may follow distinct maturation pathways following exposure to live or killed bacteria. These differences are likely to have consequences for the priming of the adaptive immune responses.
Collapse
Affiliation(s)
- Mari Norimatsu
- Institute for Animal Health, Compton, Newbury, Berkshire RG20 7NN, UK.
| | | | | | | | | |
Collapse
|
26
|
Forsberg M, Blomgran R, Lerm M, Särndahl E, Sebti SM, Hamilton A, Stendahl O, Zheng L. Differential effects of invasion by and phagocytosis of Salmonella typhimurium on apoptosis in human macrophages: potential role of Rho-GTPases and Akt. J Leukoc Biol 2003; 74:620-9. [PMID: 12960245 DOI: 10.1189/jlb.1202586] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In addition to direct activation of caspase-1 and induction of apoptosis by SipB, invasive Salmonella stimulates multiple signaling pathways that are key regulators of host cell survival. Nevertheless, little is known about the relative contributions of these pathways to Salmonella-mediated death of macrophages. We studied human monocytic U937 cells and found that apoptosis was induced by invading wild-type Salmonella typhimurium but not by phagocytosed, serum-opsonized, noninvasive Salmonella mutants. Pretreating U937 cells with inhibitors of tyrosine kinases or phosphatidylinositol-3 kinase (PI-3K) completely blocked phagocytosis of opsonized Salmonella mutants but did not affect invasion by wild-type Salmonella or the apoptosis caused by invasion. However, pretreatment with GGTI-298, a geranylgeranyltransferase-1 inhibitor that prevents prenylation of Cdc42 and Rac1, suppressed Salmonella-induced apoptosis by approximately 70%. Transduction of Tat fusion constructs containing dominant-negative Cdc42 or Rac1 significantly inhibited Salmonella-induced cell death, indicating that the cytotoxicity of Salmonella requires activation of Cdc42 and Rac. In contrast to phagocytosis of opsonized bacteria, invasion by S. typhimurium stimulated Cdc42 and Rac1, regardless of the activities of tyrosine- or PI-3K. Moreover, Salmonella infection activated Akt protein in a tyrosine-kinase or PI-3K-dependent manner, and a reduced expression of Akt by antisense transfection rendered the cells more sensitive to apoptosis induced by opsonized Salmonella. These results indicate that direct activation of Cdc42 and Rac1 by invasive Salmonella is a prerequisite of Salmonella-mediated death of U937 cells, whereas the simultaneous activation of Akt by tyrosine kinase and PI-3K during receptor-mediated phagocytosis protects cells from apoptosis.
Collapse
Affiliation(s)
- Maria Forsberg
- Division of Medical Microbiology, IMK, Linköping University, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The Gram-negative pathogen Salmonella enterica can survive and replicate within a variety of mammalian cells. Regardless of the cell type, internalized bacteria survive and replicate within the Salmonella-containing vacuole, the biogenesis of which is dependent on bacterially encoded virulence factors. In particular, Type III secretion systems translocate bacterial effector proteins into the eukaryotic cell where they can specifically interact with a variety of targets. Salmonella has two distinct Type III secretion systems that are believed to have completely different functions. The SPI2 system is induced intracellularly and is required for intracellular survival in macrophages; it plays no role in invasion but is categorized as being required for Salmonella-containing vacuole biogenesis. In contrast, the SPI1 Type III secretion system is induced extracellularly and is essential for invasion of nonphagocytic cells. Its role in post-invasion processes has not been well studied. Recent studies indicate that Salmonella-containing vacuole biogenesis may be more dependent on SPI1 than previously believed. Other non-SPI2 virulence factors and the host cell itself may play critical roles in determining the intracellular environment of this facultative intracellular pathogen. In this review we discuss the recent advances in determining the mechanisms by which Salmonella regulate Salmonella-containing vacuole biogenesis and the implications of these findings.
Collapse
Affiliation(s)
- Leigh A Knodler
- Host-Parasite Interactions Section, Laboratory of Intracellular Parasites, National Institutes of Allergy and Infectious Diseases/NIH, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | | |
Collapse
|
28
|
Khullar M, Singh RD, Smriti M, Ganguly NK. Anaerobiosis-induced virulence of Salmonella enterica subsp. enterica serovar Typhimurium: role of phospholipase Cgamma signalling cascade. J Med Microbiol 2003; 52:741-745. [PMID: 12909648 DOI: 10.1099/jmm.0.05186-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) can initiate entry into non-phagocytic epithelial cells by triggering certain signal transduction pathways, thereby allowing the pathogen to invade and establish a niche within host cells. Anaerobiosis has been shown to be an important inducer of the invasion process of S. Typhimurium. However, the effect of anaerobiosis on modulation of cell signalling cascades by S. Typhimurium is not known. In the present study, the phospholipase Cgamma signalling cascade was investigated in mice enterocytes, following interaction with S. Typhimurium grown under aerobic and anaerobic growth conditions. Significant increases in enterocyte intracellular calcium and inositol 1,4,5-triphosphate levels were observed on interaction with S. Typhimurium grown anaerobically compared with S. Typhimurium grown aerobically. An increased membrane/cytosolic ratio of protein kinase C was also seen with anaerobic S. Typhimurium in enterocytes compared with aerobic S. Typhimurium. These data suggest that anaerobically grown organisms are more efficient in initiating cell-signalling events than are aerobically grown bacteria. These enhanced cell signals may contribute to the increased virulence of S. Typhimurium grown anaerobically.
Collapse
Affiliation(s)
- Madhu Khullar
- Departments of Experimental Medicine and Biotechnology1 and Medical Microbiology2, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Raman Deep Singh
- Departments of Experimental Medicine and Biotechnology1 and Medical Microbiology2, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Manu Smriti
- Departments of Experimental Medicine and Biotechnology1 and Medical Microbiology2, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Nirmal Kumar Ganguly
- Departments of Experimental Medicine and Biotechnology1 and Medical Microbiology2, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
29
|
Tafazoli F, Magnusson KE, Zheng L. Disruption of epithelial barrier integrity by Salmonella enterica serovar typhimurium requires geranylgeranylated proteins. Infect Immun 2003; 71:872-81. [PMID: 12540569 PMCID: PMC145360 DOI: 10.1128/iai.71.2.872-881.2003] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epithelial cells that line the human intestinal mucosa constitute the initial sites of host invasion by bacterial pathogens. A number of bacteria, such as Salmonella and Yersinia spp., have been shown to disrupt the integrity of the epithelial barrier, although little is known about the mechanisms underlying that effect. We found that polarized MDCK-1 epithelial cells infected with invasive Salmonella enterica serovar Typhimurium SL1344 exhibited marked changes in F-actin organization, an increase in the paracellular flux of dextran, and a rapid decrease in transepithelial electrical resistance (TER). In contrast, infection with an isogenic noninvasive mutant (hilA) increased the TER in these cells. Pretreating MDCK-1 cells with the inhibitors for tyrosine kinase (genistein) or phosphatidylinositol 3-kinase (wortmannin) did not affect invasion and subsequent perturbation of the epithelial barrier by serovar Typhimurium. Instead, the geranylgeranyltransferase 1 inhibitor GGTI-298, but not the farnesyltransferase inhibitor FTI-277, clearly reversed the capacity of serovar Typhimurium to disrupt the epithelial barrier. The substrates for GGTI-298 include Rho family GTPases, as indicated by inhibiting prenylation of Rac1 and Cdc42. Infection with wild-type serovar Typhimurium increased the level of activated Rac1 and Cdc42 and caused these proteins to accumulate apically in MDCK-1 cells. This Salmonella-induced accumulation of Rac1 and Cdc42 and alteration of the junction-associated proteins ZO-1, occludin, and E-cadherin in MDCK-1 cells were markedly inhibited by GGTI-298. These results suggest that activation of geranylgeranylated proteins, including Rac1 and Cdc42, is critical for disruption of barrier integrity by serovar Typhimurium in polarized MDCK-1 cells.
Collapse
Affiliation(s)
- Farideh Tafazoli
- Division of Medical Microbiology, Department of Molecular and Clinical Medicine, Linköping University, Sweden.
| | | | | |
Collapse
|
30
|
Barocchi MA, Ko AI, Reis MG, McDonald KL, Riley LW. Rapid translocation of polarized MDCK cell monolayers by Leptospira interrogans, an invasive but nonintracellular pathogen. Infect Immun 2002; 70:6926-32. [PMID: 12438371 PMCID: PMC132952 DOI: 10.1128/iai.70.12.6926-6932.2002] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic spirochetes of the genus Leptospira are a major cause of human zoonotic infectious disease worldwide. After gaining entry through the skin, the organism causes disease by hematogenously disseminating to multiple organs. The mechanism by which it penetrates the mammalian cell barriers to disseminate is not well understood. In this study, we used a low-passage-number isolate of Leptospira interrogans to elucidate the invasive potential of this spirochete. Quantification of bacteria by dark-field microscopy revealed that pathogenic spirochetes were able to translocate through polarized MDCK cell monolayers at a rate significantly greater than that of nonpathogenic Leptospira or a recognized invasive bacterial pathogen, Salmonella: In contrast to Salmonella, L. interrogans did not alter transepithelial electrical resistance during cell translocation. Both transmission and scanning electron microscopy revealed tight association of the extracellular spirochetes with the host cell plasma membrane, without membrane perturbations suggestive of cytoskeletal rearrangement. Spirochetes were not observed within intercellular junctions or membrane-bound compartments inside cells. They were found within the cytoplasm of only 8% of the counted cells. These results indicate that Leptospira is an invasive but not a facultative intracellular organism. We propose that the rapid translocation of mammalian cells by pathogenic Leptospira is a mechanism designed to evade killing by host cells that permits the organism to quickly reach the bloodstream and disseminate to multiple organs.
Collapse
Affiliation(s)
- Michele A Barocchi
- Division of Infectious Diseases and Immunity, School of Public Health, University of California, Berkeley, 94720, USA
| | | | | | | | | |
Collapse
|
31
|
Clements MO, Eriksson S, Thompson A, Lucchini S, Hinton JCD, Normark S, Rhen M. Polynucleotide phosphorylase is a global regulator of virulence and persistency in Salmonella enterica. Proc Natl Acad Sci U S A 2002; 99:8784-9. [PMID: 12072563 PMCID: PMC124376 DOI: 10.1073/pnas.132047099] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2002] [Indexed: 11/18/2022] Open
Abstract
For many pathogens, the ability to regulate their replication in host cells is a key element in establishing persistency. Here, we identified a single point mutation in the gene for polynucleotide phosphorylase (PNPase) as a factor affecting bacterial invasion and intracellular replication, and which determines the alternation between acute or persistent infection in a mouse model for Salmonella enterica infection. In parallel, with microarray analysis, PNPase was found to affect the mRNA levels of a subset of virulence genes, in particular those contained in Salmonella pathogenicity islands 1 and 2. The results demonstrate a connection between PNPase and Salmonella virulence and show that alterations in PNPase activity could represent a strategy for the establishment of persistency.
Collapse
Affiliation(s)
- Mark O Clements
- Microbiology and Tumor Biology Center, Karolinska Institute, Nobels Väg 16, 171 77 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
32
|
Brumell JH, Tang P, Zaharik ML, Finlay BB. Disruption of the Salmonella-containing vacuole leads to increased replication of Salmonella enterica serovar typhimurium in the cytosol of epithelial cells. Infect Immun 2002; 70:3264-70. [PMID: 12011022 PMCID: PMC127987 DOI: 10.1128/iai.70.6.3264-3270.2002] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2001] [Revised: 01/05/2002] [Accepted: 03/10/2002] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that inhabits a vacuolar compartment, called the Salmonella-containing vacuole (SCV), in infected host cells. Maintenance of the SCV is accomplished by SifA, and mutants of this Salmonella pathogenicity island 2 type III effector replicate more efficiently in epithelial cells. Here we demonstrate that enhanced replication of sifA mutants occurs in the cytosol of these cells. Increased replication of wild-type bacteria was also observed in cells treated with wortmannin or expressing Rab5 Q79L or Rab7 N125I, all of which caused a loss of SCV integrity. Our findings demonstrate the requirement of the host cell endosomal system for maintenance of the SCV and that loss of this compartment allows increased replication of serovar Typhimurium in the cytosol of epithelial cells.
Collapse
Affiliation(s)
- John H Brumell
- Biotechnology Laboratory and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | | | | | | |
Collapse
|
33
|
Abstract
After uptake, Salmonella resides within a unique organelle, the Salmonella-containing vacuole (SCV) in which it eventually replicates. Here we recapitulate the knowledge about how Salmonella controls SCV maturation and the different steps of its intracellular trafficking.
Collapse
Affiliation(s)
- J P Gorvel
- Centre d'Immunologie Inserm-CNRS-Univ. Med. de Marseille-Luminy, Parc Scientifique de Luminy, Case 906, 13288 Marseille cedex 9, France
| | | |
Collapse
|
34
|
Abstract
Environmental pH is one the major factors affecting the composition, biological activities, and pathogenic potential of the biofilms colonizing supragingival surfaces. In periodontal diseases, small changes in pH from the metabolism of amino acids and urea may influence the activity of proteolytic enzymes of host and bacterial origin. Still, there is a significant void in the understanding of pH-dependent gene expression in bacteria, in general, and this is of course a more acute problem when one considers there is virtually no information about gene expression in response to pH in biofilms. The development of new methods and applications of some of the techniques detailed above should help to ameliorate this situation and to generate much-needed data about the role of pH in biofilm composition, stability, and activity.
Collapse
Affiliation(s)
- R A Burne
- Department of Microbiology and Immunology, and Center for Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | |
Collapse
|
35
|
Brumell JH, Tang P, Mills SD, Finlay BB. Characterization of Salmonella-induced filaments (Sifs) reveals a delayed interaction between Salmonella-containing vacuoles and late endocytic compartments. Traffic 2001; 2:643-53. [PMID: 11555418 DOI: 10.1034/j.1600-0854.2001.20907.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Salmonella typhimurium is a facultative intracellular pathogen that colonizes host cells throughout the course of infection. A unique feature of this pathogen is its ability to enter into (invade) epithelial cells and elongate the vacuole within which it resides into tubular structures called Salmonella-induced filaments (Sifs). In this study we sought to characterize the mechanism of Sif formation by immunofluorescence analysis using subcellular markers. The late endosomal lipid lysobisphosphatidic acid associated in a punctate pattern with the Salmonella-containing vacuole, starting 90 min after infection and increasing thereafter. Lysobisphosphatidic acid-rich vesicles were also found to interact with Sifs, at numerous sites along the tubules. Similarly, cholesterol-rich vesicles were also found in association with intracellular bacteria and Sifs. The lysosomal hydrolase cathepsin D was present in Sifs, both in a punctate pattern and, at later times, predominantly in an uninterrupted linear pattern. Rab7 associated with Sifs and expression of the N125I dominant negative mutant of this GTPase inhibited Sif formation. Transfection of HeLa cells with a vector encoding SifA fused to the green fluorescent protein caused swelling and aggregation of lysobisphosphatidic acid-containing compartments, suggesting that this virulence factor directs membrane fusion events involving late endosomes. Our findings demonstrate that Sif formation involves fusion of late endocytic compartments with the Salmonella-containing vacuole, and suggest that SifA modulates this event.
Collapse
Affiliation(s)
- J H Brumell
- Biotechnology Laboratory and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T-1Z3, Canada
| | | | | | | |
Collapse
|
36
|
Henson PM, Bratton DL, Fadok VA. The phosphatidylserine receptor: a crucial molecular switch? Nat Rev Mol Cell Biol 2001; 2:627-33. [PMID: 11483996 DOI: 10.1038/35085094] [Citation(s) in RCA: 236] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The uptake and removal of necrotic or lysed cells involves inflammation and an immune response, due in part to processes that involve members of the collectin family, surface calreticulin and CD91. Clearance of apoptotic cells, by contrast, does not induce either inflammation or immunity. Could the phosphatidylserine receptor be the molecular switch that determines what the outcome will be?
Collapse
Affiliation(s)
- P M Henson
- Program in Cell Biology, Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson Street, Denver, Colorado 80206, USA.
| | | | | |
Collapse
|
37
|
Lu L, Walker WA. Pathologic and physiologic interactions of bacteria with the gastrointestinal epithelium. Am J Clin Nutr 2001; 73:1124S-1130S. [PMID: 11393190 DOI: 10.1093/ajcn/73.6.1124s] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Communication between microorganisms and the gastrointestinal epithelium, ie, bacterial-epithelial "crosstalk," is examined. Because most basic research on the molecular interaction of bacteria with the gut epithelium relates to pathogen-enterocyte interaction, crosstalk with pathologic bacterial is considered in detail. Through their interactions with the intestinal epithelium, pathogens can modify epithelium function to enhance their penetration across the epithelial barrier and to exploit mucosal host defenses for their own benefit. Three representative pathogens are used to illustrate the various adaptive techniques used to colonize and penetrate the mucosal barrier. Salmonella enterica typhimurium interacts with the physiologic receptor for epidermal growth factor to co-opt the receptor's signal transduction mechanisms. Enteropathic Escherichia coli secretes a receptor (type III secretion) into the microvillus surface of enterocytes that disrupts the microvillus and alters its actin structure to form a dome-like anchoring site. Shigella flexneri is used to illustrate how pathogens use the follicular epithelial cell (M cell), the physiologic conduit for antigens to reach gut associated-lymphoid tissues, for penetration of the epithelial barrier. Shigella organisms attached to M cells use their endocytotic properties to enter the cell. Once inside the cell, the organism lyses the endocytic vacuole and co-opts actin and myosin to form a propelling tail for further penetration of the epithelium through the basolateral surface. Probiotics can protect the intestine by competing with pathogens for attachment, strengthening tight junctions between enterocytes, and enhancing the mucosal immune response to pathogens. However, additional molecular studies are needed to define more precisely the mechanism of probiotic-epithelial crosstalk.
Collapse
Affiliation(s)
- L Lu
- Harvard Medical School, Boston, USA
| | | |
Collapse
|
38
|
Hess P, Daryab N, Michaelis K, Reisenauer A, Oelschlaeger TA. Type 1 pili of Citrobacter freundii mediate invasion into host cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2001; 485:225-35. [PMID: 11109110 DOI: 10.1007/0-306-46840-9_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Affiliation(s)
- P Hess
- Institut für Molekulare Infektionsbiologie, Universität Würzburg, Germany
| | | | | | | | | |
Collapse
|
39
|
Beck T, Delley PA, Hall MN. Control of the actin cytoskeleton by extracellular signals. Results Probl Cell Differ 2001; 32:231-62. [PMID: 11131835 DOI: 10.1007/978-3-540-46560-7_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- T Beck
- Department of Biochemistry, Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | | |
Collapse
|
40
|
Coppolino MG, Kong C, Mohtashami M, Schreiber AD, Brumell JH, Finlay BB, Grinstein S, Trimble WS. Requirement for N-ethylmaleimide-sensitive factor activity at different stages of bacterial invasion and phagocytosis. J Biol Chem 2001; 276:4772-80. [PMID: 11092884 DOI: 10.1074/jbc.m007792200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bacterial invasion, like the process of phagocytosis, involves extensive and localized protrusion of the host cell plasma membrane. To examine the molecular mechanisms of the membrane remodeling that accompanies bacterial invasion, soluble NSF attachment protein receptor (SNARE)-mediated membrane traffic was studied in cultured cells during infection by Salmonella typhimurium. A green fluorescent protein-tagged chimera of VAMP3, a SNARE characteristic of recycling endosomes, was found to accumulate at sites of Salmonella invasion. To analyze the possible role of SNARE-mediated membrane traffic in bacterial infection, invasion was measured in cells expressing a dominant-negative form of N-ethylmaleimide-sensitive factor (NSF), an essential regulator of membrane fusion. Inhibition of NSF activity did not affect cellular invasion by S. typhimurium nor the associated membrane remodeling. By contrast, Fcgamma receptor-mediated phagocytosis was greatly reduced in the presence of the mutant NSF. Most important, dominant-negative NSF significantly impaired the fusion of Salmonella-containing vacuoles with endomembranes. These observations indicate that the membrane protrusions elicited by Salmonella invasion, unlike those involved in phagocytosis, occur via an NSF-independent mechanism, whereas maturation of Salmonella-containing vacuoles is NSF-dependent.
Collapse
Affiliation(s)
- M G Coppolino
- Cell Biology Programme, Research Institute, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Jesenberger V, Procyk KJ, Rüth J, Schreiber M, Theussl HC, Wagner EF, Baccarini M. Protective role of Raf-1 in Salmonella-induced macrophage apoptosis. J Exp Med 2001; 193:353-64. [PMID: 11157055 PMCID: PMC2195927 DOI: 10.1084/jem.193.3.353] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Invasive Salmonella induces macrophage apoptosis via the activation of caspase-1 by the bacterial protein SipB. Here we show that infection of macrophages with Salmonella causes the activation and degradation of Raf-1, an important intermediate in macrophage proliferation and activation. Raf-1 degradation is SipB- and caspase-1-dependent, and is prevented by proteasome inhibitors. To study the functional significance of Raf-1 in this process, the c-raf-1 gene was inactivated by Cre-loxP-mediated recombination in vivo. Macrophages lacking c-raf-1 are hypersensitive towards pathogen-induced apoptosis. Surprisingly, activation of the antiapoptotic mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and nuclear factor kappaB pathways is normal in Raf-1-deficient macrophages, and mitochondrial fragility is not increased. Instead, pathogen-mediated activation of caspase-1 is enhanced selectively, implying that Raf-1 antagonizes stimulus-induced caspase-1 activation and apoptosis.
Collapse
Affiliation(s)
| | | | - Jochen Rüth
- Department of Cell and Microbiology, Institute of Microbiology and Genetics
| | - Martin Schreiber
- Department of Cell and Microbiology, Institute of Microbiology and Genetics
| | | | - Erwin F. Wagner
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Manuela Baccarini
- Department of Cell and Microbiology, Institute of Microbiology and Genetics
| |
Collapse
|
42
|
Jepson MA, Schlecht HB, Collares-Buzato CB. Localization of dysfunctional tight junctions in Salmonella enterica serovar typhimurium-infected epithelial layers. Infect Immun 2000; 68:7202-8. [PMID: 11083857 PMCID: PMC97842 DOI: 10.1128/iai.68.12.7202-7208.2000] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Infection of polarized MDCK epithelial layers by Salmonella enterica serovar Typhimurium is accompanied by increased tight junction permeability and by contraction of perijunctional actinomyosin. We localized dysfunctional tight junctions in serovar Typhimurium-infected MDCK layers by imaging apical-basolateral intramembrane diffusion of fluorescent lipid and found that loss of the apical-basolateral diffusion barrier (tight junction fence function) was most marked in areas of prominent perijunctional contraction. The protein kinase inhibitor staurosporine prevented perijunctional contraction but did not reverse the effects of serovar Typhimurium on tight junction barrier function. Hence, perijunctional contraction is not required for Salmonella-induced tight junction dysfunction and this epithelial response to infection may be multifactorial.
Collapse
Affiliation(s)
- M A Jepson
- Cell Imaging Facility and Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | | | | |
Collapse
|
43
|
Abstract
The first sigma(54) promoters in Chlamydia trachomatis L2 were mapped upstream of hypothetical proteins CT652.1 and CT683. Comparative genomics indicated that these sigma(54) promoters and potential upstream activation binding sites are conserved in orthologous C. trachomatis D, C. trachomatis mouse pneumonitis strain, and Chlamydia pneumoniae (CWL029 and AR39) genes.
Collapse
Affiliation(s)
- S A Mathews
- Centre for Molecular Biotechnology, School of Life Sciences, Queensland University of Technology, Brisbane, Queensland, Australia.
| | | |
Collapse
|
44
|
Jesenberger V, Procyk KJ, Yuan J, Reipert S, Baccarini M. Salmonella-induced caspase-2 activation in macrophages: a novel mechanism in pathogen-mediated apoptosis. J Exp Med 2000; 192:1035-46. [PMID: 11015444 PMCID: PMC2193309 DOI: 10.1084/jem.192.7.1035] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The enterobacterial pathogen Salmonella induces phagocyte apoptosis in vitro and in vivo. These bacteria use a specialized type III secretion system to export a virulence factor, SipB, which directly activates the host's apoptotic machinery by targeting caspase-1. Caspase-1 is not involved in most apoptotic processes but plays a major role in cytokine maturation. We show that caspase-1-deficient macrophages undergo apoptosis within 4-6 h of infection with invasive bacteria. This process requires SipB, implying that this protein can initiate the apoptotic machinery by regulating components distinct from caspase-1. Invasive Salmonella typhimurium targets caspase-2 simultaneously with, but independently of, caspase-1. Besides caspase-2, the caspase-1-independent pathway involves the activation of caspase-3, -6, and -8 and the release of cytochrome c from mitochondria, none of which occurs during caspase-1-dependent apoptosis. By using caspase-2 knockout macrophages and chemical inhibition, we establish a role for caspase-2 in both caspase-1-dependent and -independent apoptosis. Particularly, activation of caspase-1 during fast Salmonella-induced apoptosis partially relies on caspase-2. The ability of Salmonella to induce caspase-1-independent macrophage apoptosis may play a role in situations in which activation of this protease is either prevented or uncoupled from the induction of apoptosis.
Collapse
Affiliation(s)
- V Jesenberger
- Department of Cell- and Microbiology, Institute of Microbiology and Genetics, Vienna Biocenter, Vienna, Austria
| | | | | | | | | |
Collapse
|
45
|
Finlay BB, Brumell JH. Salmonella interactions with host cells: in vitro to in vivo. Philos Trans R Soc Lond B Biol Sci 2000; 355:623-31. [PMID: 10874735 PMCID: PMC1692772 DOI: 10.1098/rstb.2000.0603] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Salmonellosis (diseases caused by Salmonella species) have several clinical manifestations, ranging from gastroenteritis (food poisoning) to typhoid (enteric) fever and bacteraemia. Salmonella species (especially Salmonella typhimurium) also represent organisms that can be readily used to investigate the complex interplay that occurs between a pathogen and its host, both in vitro and in vivo. The ease with which S. typhimurium can be cultivated and genetically manipulated, in combination with the availability of tissue culture models and animal models, has made S. typhimurium a desirable organism for such studies. In this review, we focus on Salmonella interactions with its host cells, both in tissue culture (in vitro) and in relevant animal models (in vivo), and compare results obtained using these different models. The recent advent of sophisticated imaging and molecular genetic tools has facilitated studying the events that occur in disease, thereby confirming tissue culture results, yet identifying new questions that need to be addressed in relevant disease settings.
Collapse
Affiliation(s)
- B B Finlay
- Biotechnology Laboratory, University of British Columbia, Vancouver, Canada.
| | | |
Collapse
|
46
|
Sebastiani G, Leveque G, Larivière L, Laroche L, Skamene E, Gros P, Malo D. Cloning and characterization of the murine toll-like receptor 5 (Tlr5) gene: sequence and mRNA expression studies in Salmonella-susceptible MOLF/Ei mice. Genomics 2000; 64:230-40. [PMID: 10756091 DOI: 10.1006/geno.2000.6115] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Toll-like receptors (TLRs) are a group of evolutionarily conserved pattern recognition receptors involved in the activation of the immune system in response to various pathogens. In this paper, we describe the cloning and characterization of the mouse homologue of human TLR5. Mouse Tlr5 encodes a 859-amino-acid protein that contains an N-terminal signal sequence, a leucine-rich repeat extracellular domain, a short transmembrane domain typical of type I transmembrane proteins, and a Toll/interleukin-1R signaling domain characteristic of all TLR proteins. The mouse Tlr5 protein shows 81% homology to human TLR5 and approximately 40% similarity to other TLR family members. Northern blot analysis reveals that Tlr5 is expressed predominantly in liver and lung with low-level expression in most other tissues examined. We have mapped Tlr5 to distal chromosome 1 using the (C57BL/6J x Mus spretus) x C57BL/6J Jackson BSB panel as well as a (C57BL/6J x MOLF/Ei)F(2) panel with the following position: D1Mit112-8.0 cM-Tlr5-9.6 cM-D1Mit17. The presence of a quantitative trait locus for susceptibility to Salmonella typhimurium on distal chromosome 1 prompted the examination of Tlr5 in susceptible MOLF/Ei mice. Polymorphic sequence variants in Tlr5 allowed us to identify a unique 4-allele haplotype in MOLF/Ei. Furthermore, using both Northern blot analysis and reverse transcription-polymerase chain reaction, we have shown a reduced expression of Tlr5 during infection of MOLF/Ei mice with Salmonella. The assignment of Tlr5 to a chromosomal region known to harbor a Salmonella-susceptibility locus together with decreased expression of Tlr5 mRNA in liver of susceptible MOLF/Ei mice suggests the possibility that, as with other members of this family, Tlr5 may play a role in host response to bacterial gram-negative infections.
Collapse
Affiliation(s)
- G Sebastiani
- Department of Biochemistry, Center for the Study of Host Resistance, McGill University, Montreal General Hospital, Quebec, H3G 1A4, Canada
| | | | | | | | | | | | | |
Collapse
|
47
|
Kjemtrup S, Nimchuk Z, Dangl JL. Effector proteins of phytopathogenic bacteria: bifunctional signals in virulence and host recognition. Curr Opin Microbiol 2000; 3:73-8. [PMID: 10679421 DOI: 10.1016/s1369-5274(99)00054-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Phytopathogenic bacteria deliver effectors of disease into plant hosts via a Type III secretion system. These Type III effectors have genetically determined roles in virulence. They also are among the components recognized by the putative receptors of the plant innate immune system. Recent breakthroughs include localization of some of these Type III effectors to specific host cell compartments, and the first dissection of pathogenicity islands that carry them.
Collapse
Affiliation(s)
- S Kjemtrup
- Department of Biology and Curriculum in Genetics and Molecular Biology (JLD), University of North Carolina, Chapel Hill, NC 27599-380, USA
| | | | | |
Collapse
|
48
|
Marcus SL, Brumell JH, Pfeifer CG, Finlay BB. Salmonella pathogenicity islands: big virulence in small packages. Microbes Infect 2000; 2:145-56. [PMID: 10742687 DOI: 10.1016/s1286-4579(00)00273-2] [Citation(s) in RCA: 288] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Reflecting a complex set of interactions with its host, Salmonella spp. require multiple genes for full virulence. Many of these genes are found in 'pathogenicity islands' in the chromosome. Salmonella typhimurium possesses at least five such pathogenicity islands (SPI), which confer specific virulence traits and may have been acquired by horizontal transfer from other organisms. We highlight recent progress in characterizing these SPIs and the function of some of their genes. The role of virulence genes found on a highly conserved plasmid is also discussed. Collectively, these packages of virulence cassettes are essential for Salmonella pathogenesis.
Collapse
Affiliation(s)
- S L Marcus
- Biotechnology Laboratory, and Departments of Biochemistry & Molecular Biology and Microbiology & Immunology, University of British Columbia, Wesbrook Building 237, 6174 University Boulevard, Vancouver, Canada
| | | | | | | |
Collapse
|