1
|
Yuan X, Song X, Zhang X, Hu L, Zhou D, Zhang J, Dai C. Unraveling host-pathogen dynamics in a murine Model of septic peritonitis induced by vancomycin-resistant Enterococcus faecium. Virulence 2024; 15:2367659. [PMID: 38951957 PMCID: PMC11221476 DOI: 10.1080/21505594.2024.2367659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/09/2024] [Indexed: 07/03/2024] Open
Abstract
Vancomycin-resistant Enterococcus faecium (E. faecium) infection is associated with higher mortality rates. Previous studies have emphasized the importance of innate immune cells and signalling pathways in clearing E. faecium, but a comprehensive analysis of host-pathogen interactions is lacking. Here, we investigated the interplay of host and E. faecium in a murine model of septic peritonitis. Following injection with a sublethal dose, we observed significantly increased murine sepsis score and histological score, decreased weight and bacterial burden, neutrophils and macrophages infiltration, and comprehensive activation of cytokine-mediated signalling pathway. In mice receiving a lethal dose, hypothermia significantly improved survival, reduced bacterial burden, cytokines, and CD86 expression of MHC-II+ recruited macrophages compared to the normothermia group. A mathematical model constructed by observational data from 80 animals, recapitulated the host-pathogen interplay, and further verified the benefits of hypothermia. These findings indicate that E. faecium triggers a severe activation of cytokine-mediated signalling pathway, and hypothermia can improve outcomes by reducing bacterial burden and inflammation.
Collapse
Affiliation(s)
- Xin Yuan
- School of Life Sciences, Ludong University, Yantai, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolin Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xi Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China
| | - Chenxi Dai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
2
|
Daca A, Jarzembowski T. From the Friend to the Foe- Enterococcus faecalis Diverse Impact on the Human Immune System. Int J Mol Sci 2024; 25:2422. [PMID: 38397099 PMCID: PMC10888668 DOI: 10.3390/ijms25042422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Enterococcus faecalis is a bacterium which accompanies us from the first days of our life. As a commensal it produces vitamins, metabolizes nutrients, and maintains intestinal pH. All of that happens in exchange for a niche to inhabit. It is not surprising then, that the bacterium was and is used as an element of many probiotics and its positive impact on the human immune system and the body in general is hard to ignore. This bacterium has also a dark side though. The plasticity and relative ease with which one acquires virulence traits, and the ability to hide from or even deceive and use the immune system to spread throughout the body make E. faecalis a more and more dangerous opponent. The statistics clearly show its increasing role, especially in the case of nosocomial infections. Here we present the summarization of current knowledge about E. faecalis, especially in the context of its relations with the human immune system.
Collapse
Affiliation(s)
- Agnieszka Daca
- Department of Physiopathology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Tomasz Jarzembowski
- Department of Microbiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
3
|
Bai Y, Miyanaga K, Yamamoto N. Enhanced tight junction in Caco-2 cells by the pretreatment with Lactobacillus johnsonii strain MG. Biosci Biotechnol Biochem 2023; 87:1532-1536. [PMID: 37704400 DOI: 10.1093/bbb/zbad131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
We investigated roles of Lactobacillus johnsonii MG (MG) isolated from mice with interaction with tight junction on gut barrier function with Caco-2 cell model. Pretreatment with MG enhanced barrier function and showed protective effect against Enterococcus faecium provided damage. MG treatment increased the gene expressions of transcriptional regulator NFKB and major tight junction protein, ZO-1.
Collapse
Affiliation(s)
- Yuying Bai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
- Department of Infection and Immunity, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-Shi, Tochigi, Japan
| | - Naoyuki Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Akkermansia muciniphila Reduces Peritonitis and Improves Intestinal Tissue Wound Healing after a Colonic Transmural Defect by a MyD88-Dependent Mechanism. Cells 2022; 11:cells11172666. [PMID: 36078075 PMCID: PMC9454966 DOI: 10.3390/cells11172666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Anastomotic leakage is a major complication following colorectal surgery leading to peritonitis, complications, and mortality. Akkermansia muciniphila has shown beneficial effects on the gut barrier function. Whether A. muciniphila reduces peritonitis and mortality during colonic leakage is unknown. Whether A. muciniphila can directly modulate the expression of genes in the colonic mucosa in humans has never been studied. We investigated the effects of a pretreatment (14 days) with live A. muciniphila prior to surgical colonic perforation on peritonitis, mortality, and wound healing. We used mice with an inducible intestinal-epithelial-cell-specific deletion of MyD88 (IEC-MyD88 KO) to investigate the role of the innate immune system in this context. In a proof-of-concept pilot study, healthy humans were exposed to A. muciniphila for 2 h and colonic biopsies taken before and after colonic instillation for transcriptomic analysis. Seven days after colonic perforation, A.-muciniphila-treated mice had significantly lower mortality and severity of peritonitis. This effect was associated with significant improvements of wound histological healing scores, higher production of IL22, but no changes in the mucus layer thickness or genes involved in cell renewal, proliferation, or differentiation. All these effects were abolished in IEC-MyD88 KO mice. Finally, human subjects exposed to A. muciniphila exhibited an increased level of the bacterium at the mucus level 2 h after instillation and significant changes in the expression of different genes involved in the regulation of cell cycling, gene transcription, immunity, and inflammation in their colonic mucosa. A. muciniphila improves wound healing during transmural colonic wall defect through mechanisms possibly involving IL22 signaling and requiring MyD88 in the intestinal cells. In healthy humans, colonic administration of A. muciniphila is well tolerated and changes the expression of genes involved in the immune pathways.
Collapse
|
5
|
Modulation of Bovine Endometrial Cell Receptors and Signaling Pathways as a Nanotherapeutic Exploration against Dairy Cow Postpartum Endometritis. Animals (Basel) 2021; 11:ani11061516. [PMID: 34071093 PMCID: PMC8224678 DOI: 10.3390/ani11061516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The provision of updated information on the molecular pathogenesis of bovine endometritis with host-pathogen interactions and the possibility of exploring the cellular sensors mechanism in a nanotechnology-based drug delivery system against persistent endometritis were reported in this review. The mechanism of Gram-negative bacteria and their ligands has been vividly explored, with the paucity of research detail on Gram-positive bacteria in bovine endometritis. The function of cell receptors, biomolecules proteins, and sensors were reportedly essential in transferring signals into cell signaling pathways to induce immuno-inflammatory responses by elevating pro-inflammatory cytokines. Therefore, understanding endometrial cellular components and signaling mechanisms across pathogenesis are essential for nanotherapeutic exploration against bovine endometritis. The nanotherapeutic discovery that could inhibit infectious signals at the various cell receptors and signal transduction levels, interfering with transcription factors activation and pro-inflammatory cytokines and gene expression, significantly halts endometritis. Abstract In order to control and prevent bovine endometritis, there is a need to understand the molecular pathogenesis of the infectious disease. Bovine endometrium is usually invaded by a massive mobilization of microorganisms, especially bacteria, during postpartum dairy cows. Several reports have implicated the Gram-negative bacteria in the pathogenesis of bovine endometritis, with information dearth on the potentials of Gram-positive bacteria and their endotoxins. The invasive bacteria and their ligands pass through cellular receptors such as TLRs, NLRs, and biomolecular proteins of cells activate the specific receptors, which spontaneously stimulates cellular signaling pathways like MAPK, NF-kB and sequentially triggers upregulation of pro-inflammatory cytokines. The cascade of inflammatory induction involves a dual signaling pathway; the transcription factor NF-κB is released from its inhibitory molecule and can bind to various inflammatory genes promoter. The MAPK pathways are concomitantly activated, leading to specific phosphorylation of the NF-κB. The provision of detailed information on the molecular pathomechanism of bovine endometritis with the interaction between host endometrial cells and invasive bacteria in this review would widen the gap of exploring the potential of receptors and signal transduction pathways in nanotechnology-based drug delivery system. The nanotherapeutic discovery of endometrial cell receptors, signal transduction pathway, and cell biomolecules inhibitors could be developed for strategic inhibition of infectious signals at the various cell receptors and signal transduction levels, interfering on transcription factors activation and pro-inflammatory cytokines and genes expression, which may significantly protect endometrium against postpartum microbial invasion.
Collapse
|
6
|
Pundir P, Liu R, Vasavda C, Serhan N, Limjunyawong N, Yee R, Zhan Y, Dong X, Wu X, Zhang Y, Snyder SH, Gaudenzio N, Vidal JE, Dong X. A Connective Tissue Mast-Cell-Specific Receptor Detects Bacterial Quorum-Sensing Molecules and Mediates Antibacterial Immunity. Cell Host Microbe 2019; 26:114-122.e8. [PMID: 31278040 DOI: 10.1016/j.chom.2019.06.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/09/2019] [Accepted: 06/10/2019] [Indexed: 01/20/2023]
Abstract
Quorum-sensing molecules (QSMs) are secreted by bacteria to signal population density. Upon reaching a critical concentration, QSMs induce transcriptional alterations in bacteria, which enable virulence factor expression and biofilm formation. It is unclear whether mammalian hosts can recognize QSMs to trigger responsive antibacterial immunity. We report that mouse mast-cell-specific G-protein-coupled receptor Mrgprb2 and its human homolog MRGPRX2 are receptors for Gram-positive QSMs, including competence-stimulating peptide (CSP)-1. CSP-1 activates Mrgprb2 and MRGPRX2, triggering mast cell degranulation, which inhibits bacterial growth and prevents biofilm formation. Such antibacterial functions are reduced in Mrgprb2-deficient mast cells, while wild-type mast cells fail to inhibit the growth of bacterial strains lacking CSP-1. Mrgprb2-knockout mice exhibit reduced bacterial clearance, while pharmacologically activating Mrgprb2 in vivo eliminates bacteria and improves disease score. These findings identify a host defense mechanism that uses QSMs as an "Achilles heel" and suggest MRGPRX2 as a potential therapeutic target for controlling bacterial infections.
Collapse
Affiliation(s)
- Priyanka Pundir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rui Liu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chirag Vasavda
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nadine Serhan
- Unité de Différenciation Epithéliale et Autoimmunité Rhumatoïde, INSERM, Université de Toulouse, Toulouse 31000, France
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rebecca Yee
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yingzhuan Zhan
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xueqing Wu
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicolas Gaudenzio
- Unité de Différenciation Epithéliale et Autoimmunité Rhumatoïde, INSERM, Université de Toulouse, Toulouse 31000, France
| | - Jorge E Vidal
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Fiore E, Van Tyne D, Gilmore MS. Pathogenicity of Enterococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0053-2018. [PMID: 31298205 PMCID: PMC6629438 DOI: 10.1128/microbiolspec.gpp3-0053-2018] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Indexed: 12/19/2022] Open
Abstract
Enterococci are unusually well adapted for survival and persistence in a variety of adverse environments, including on inanimate surfaces in the hospital environment and at sites of infection. This intrinsic ruggedness undoubtedly played a role in providing opportunities for enterococci to interact with other overtly drug-resistant microbes and acquire additional resistances on mobile elements. The rapid rise of antimicrobial resistance among hospital-adapted enterococci has rendered hospital-acquired infections a leading therapeutic challenge. With about a quarter of a genome of additional DNA conveyed by mobile elements, there are undoubtedly many more properties that have been acquired that help enterococci persist and spread in the hospital setting and cause diseases that have yet to be defined. Much remains to be learned about these ancient and rugged microbes, particularly in the area of pathogenic mechanisms involved with human diseases.
Collapse
Affiliation(s)
- Elizabeth Fiore
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Daria Van Tyne
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Michael S Gilmore
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
8
|
Popović N, Djokić J, Brdarić E, Dinić M, Terzić-Vidojević A, Golić N, Veljović K. The Influence of Heat-Killed Enterococcus faecium BGPAS1-3 on the Tight Junction Protein Expression and Immune Function in Differentiated Caco-2 Cells Infected With Listeria monocytogenes ATCC 19111. Front Microbiol 2019; 10:412. [PMID: 30891021 PMCID: PMC6411766 DOI: 10.3389/fmicb.2019.00412] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Listeria monocytogenes, the common foodborne pathogenic bacteria species, compromises the intestinal epithelial barrier, leading to development of the listeriosis, a severe disease especially among immunocompromised individuals. L. monocytogenes infection usually requires antibiotic treatment, however, excessive use of antibiotics promotes emergence of antibiotic resistance and the destruction of gut microbiota. Probiotics, including lactic acid bacteria (LAB), have been repeatedly proven as an alternative approach for the treatment of various infections. We have analyzed the potential of Enterococcus faecium BGPAS1-3, a dairy isolate exhibiting strong direct antilisterial effect, to modulate the response of differentiated Caco-2 intestinal epithelial cells to L. monocytogenes ATCC 19111 infection. We showed that the molecule with antilisterial effect is a bacterial cell-wall protein that is highly resistant to the high-temperature treatment. When we tested the antilisterial potential of heat-killed BGPAS1-3, we found that it could prevent tight junction disruption in differentiated Caco-2 monolayer infected with L. monocytogenes ATCC 19111, induce antilisterial host response mechanisms, and stimulate the production of protective TGF-β in intestinal epithelial cells. We also showed that the modulation of MyD88 dependent TLR2 and TLR4 pathways by BGPAS1-3 are involved in host response against L. monocytogenes ATCC 19111. Since heat-killed BGPAS1-3 possess strong antilisterial effects, such postbiotic could be used as a controllable and safe therapeutic.
Collapse
Affiliation(s)
| | - Jelena Djokić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | |
Collapse
|
9
|
Enterococcus faecium TIR-Domain Genes Are Part of a Gene Cluster Which Promotes Bacterial Survival in Blood. Int J Microbiol 2019; 2018:1435820. [PMID: 30631364 PMCID: PMC6304867 DOI: 10.1155/2018/1435820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/14/2018] [Indexed: 11/29/2022] Open
Abstract
Enterococcus faecium has undergone a transition to a multidrug-resistant nosocomial pathogen. The population structure of E. faecium is characterized by a sharp distinction of clades, where the hospital-adapted lineage is primarily responsible for bacteremia. So far, factors that were identified in hospital-adapted strains and that promoted pathogenesis of nosocomial E. faecium mainly play a role in adherence and biofilm production, while less is known about factors contributing to survival in blood. This study identified a gene cluster, which includes genes encoding bacterial Toll/interleukin-1 receptor- (TIR-) domain-containing proteins (TirEs). The cluster was found to be unique to nosocomial strains and to be located on a putative mobile genetic element of phage origin. The three genes within the cluster appeared to be expressed as an operon. Expression was detected in bacterial culture media and in the presence of human blood. TirEs are released into the bacterial supernatant, and TirE2 is associated with membrane vesicles. Furthermore, the tirE-gene cluster promotes bacterial proliferation in human blood, indicating that TirE may contribute to the pathogenesis of bacteremia.
Collapse
|
10
|
Group IIA-Secreted Phospholipase A 2 in Human Serum Kills Commensal but Not Clinical Enterococcus faecium Isolates. Infect Immun 2018; 86:IAI.00180-18. [PMID: 29784864 DOI: 10.1128/iai.00180-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/18/2018] [Indexed: 11/20/2022] Open
Abstract
Human innate immunity employs cellular and humoral mechanisms to facilitate rapid killing of invading bacteria. The direct killing of bacteria by human serum is attributed mainly to the activity of the complement system, which forms pores in Gram-negative bacteria. Although Gram-positive bacteria are considered resistant to killing by serum, we uncover here that normal human serum effectively kills Enterococcus faecium Comparison of a well-characterized collection of commensal and clinical E. faecium isolates revealed that human serum specifically kills commensal E. faecium strains isolated from normal gut microbiota but not clinical isolates. Inhibitor studies show that the human group IIA secreted phospholipase A2 (hGIIA), but not complement, is responsible for killing of commensal E. faecium strains in human normal serum. This is remarkable since the hGIIA concentration in "noninflamed" serum was considered too low to be bactericidal against Gram-positive bacteria. Mechanistic studies showed that serum hGIIA specifically causes permeabilization of commensal E. faecium membranes. Altogether, we find that a normal concentration of hGIIA in serum effectively kills commensal E. faecium and that resistance of clinical E. faecium to hGIIA could have contributed to the ability of these strains to become opportunistic pathogens in hospitalized patients.
Collapse
|
11
|
Gram-Positive Uropathogens, Polymicrobial Urinary Tract Infection, and the Emerging Microbiota of the Urinary Tract. Microbiol Spectr 2017; 4. [PMID: 27227294 DOI: 10.1128/microbiolspec.uti-0012-2012] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gram-positive bacteria are a common cause of urinary-tract infection (UTI), particularly among individuals who are elderly, pregnant, or who have other risk factors for UTI. Here we review the epidemiology, virulence mechanisms, and host response to the most frequently isolated Gram-positive uropathogens: Staphylococcus saprophyticus, Enterococcus faecalis, and Streptococcus agalactiae. We also review several emerging, rare, misclassified, and otherwise underreported Gram-positive pathogens of the urinary tract including Aerococcus, Corynebacterium, Actinobaculum, and Gardnerella. The literature strongly suggests that urologic diseases involving Gram-positive bacteria may be easily overlooked due to limited culture-based assays typically utilized for urine in hospital microbiology laboratories. Some UTIs are polymicrobial in nature, often involving one or more Gram-positive bacteria. We herein review the risk factors and recent evidence for mechanisms of bacterial synergy in experimental models of polymicrobial UTI. Recent experimental data has demonstrated that, despite being cleared quickly from the bladder, some Gram-positive bacteria can impact pathogenic outcomes of co-infecting organisms. When taken together, the available evidence argues that Gram-positive bacteria are important uropathogens in their own right, but that some can be easily overlooked because they are missed by routine diagnostic methods. Finally, a growing body of evidence demonstrates that a surprising variety of fastidious Gram-positive bacteria may either reside in or be regularly exposed to the urinary tract and further suggests that their presence is widespread among women, as well as men. Experimental studies in this area are needed; however, there is a growing appreciation that the composition of bacteria found in the bladder could be a potentially important determinant in urologic disease, including susceptibility to UTI.
Collapse
|
12
|
Karwaciak I, Gorzkiewicz M, Bartosz G, Pulaski L. TLR2 activation induces antioxidant defence in human monocyte-macrophage cell line models. Oncotarget 2017; 8:54243-54264. [PMID: 28903338 PMCID: PMC5589577 DOI: 10.18632/oncotarget.17342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 04/10/2017] [Indexed: 01/08/2023] Open
Abstract
When monocytes are recruited to inflammation/infection sites, extravasate and differentiate into macrophages, they encounter increasing levels of oxidative stress, both from exogenous and endogenous sources. In this study, we aimed to determine whether there are specific biochemical mechanisms responsible for an increase in oxidative stress resistance in differentiating macrophages. We performed experiments on in vitro cell line models of the monocyte-macrophage differentiation axis (less differentiated THP-1 cells and more differentiated Mono Mac 6 cells). At the same time, we verified the hypothesis that activating monocyte/macrophage innate immune response by pathogens (exemplified by stimulating the TLR2 pattern recognition receptor) would further strengthen cellular antioxidant defences. We found that resistance to exogenous oxidative stress increased substantially both during differentiation and upon activation of TLR2. This increase in antioxidant resistance was accompanied by decrease in free radical damage to cellular proteins. On the molecular level, this resistance was mediated especially by increased levels and activity of glutathione, glutathione-related antioxidant enzymes and Mn superoxide dismutase, as shown by gene expression assays, Western blotting and enzyme activity assays. Moreover, upon TLR2 activation additional molecular mechanisms came into play, conferring additional resistance levels even upon differentiated macrophage-like cells, mainly related to thioredoxin-linked antioxidant enzymes.
Collapse
Affiliation(s)
- Iwona Karwaciak
- Laboratory of Transcriptional Regulation, Institute of Medical Biology PAS, Lodz, Poland
| | - Michal Gorzkiewicz
- Laboratory of Transcriptional Regulation, Institute of Medical Biology PAS, Lodz, Poland.,Department of General Biophysics, Faculty of Biology and Environmental Sciences, University of Lodz, Lodz, Poland
| | - Grzegorz Bartosz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Sciences, University of Lodz, Lodz, Poland
| | - Lukasz Pulaski
- Laboratory of Transcriptional Regulation, Institute of Medical Biology PAS, Lodz, Poland.,Department of Molecular Biophysics, Faculty of Biology and Environmental Sciences, University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Wan J, Shan Y, Fan Y, Fan C, Chen S, Sun J, Zhu L, Qin L, Yu M, Lin Z. NF-κB inhibition attenuates LPS-induced TLR4 activation in monocyte cells. Mol Med Rep 2016; 14:4505-4510. [PMID: 27748869 PMCID: PMC5101966 DOI: 10.3892/mmr.2016.5825] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptor (TLR) family are receptors for extracellular or intracellular signaling, such as lipopolysaccharide (LPS), or 12-O-tetradecanoylphorbol-13-acetate. TLR induces the differentiation of human myeloid monocytic‑leukemia cells (THP-1) to macrophages. However, the relationship between extracellular or intracellular signaling and the TLR protein level remain to be determined. Using RT-PCR and western blot analysis, the aim of the present study was to determine whether TLR4, a major TLR family member, could be moderately upregulated by high concentration of LPS and whether it promoted the maturation of THP1 cells. The results showed that, upregulated TLR4 at the protein level and mRNA level enriched the TLR4 modulation style. In addition, TLR4 expression was blocked by nuclear factor (NF)-κB inhibitor, and LPS stimulated NF-κB binding in the TLR4 gene promoter. Therefore, the increased expression of TLR4 in the responsiveness of LPS-treated THP1 cells occurred in response to the upregulation of their respective receptors, as well as a tight binding of NF-κB in the TLR4 gene promoter.
Collapse
Affiliation(s)
- Jian Wan
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Yi Shan
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Yibo Fan
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Conghui Fan
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Song Chen
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Jie Sun
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Lili Zhu
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Long Qin
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Mengjin Yu
- Department of Emergency and Critical Care Medicine, The People's Hospital of Pudong New Area, Shanghai University of Health and Science, Shanghai 201200, P.R. China
| | - Zhaofen Lin
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
14
|
Lipoteichoic acids as a major virulence factor causing inflammatory responses via Toll-like receptor 2. Arch Pharm Res 2016; 39:1519-1529. [PMID: 27498542 DOI: 10.1007/s12272-016-0804-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/28/2016] [Indexed: 12/29/2022]
Abstract
Lipoteichoic acid (LTA), a major cell wall component of Gram-positive bacteria, is associated with various inflammatory diseases ranging from minor skin diseases to severe sepsis. It is known that LTA is recognized by Toll-like receptor 2 (TLR2), leading to the initiation of innate immune responses and further development of adaptive immunity. However, excessive immune responses may result in the inflammatory sequelae that are involved in severe diseases such as sepsis. Although numerous studies have tried to identify the molecular basis for the pathophysiology of Gram-positive bacterial infection, the exact role of LTA during the infection has not been clearly elucidated. This review provides an overview of LTA structure and host recognition by TLR2 that leads to the activation of innate immune responses. Emphasis is placed on differential immunostimulating activities of LTAs of various Gram-positive bacteria at the molecular level.
Collapse
|
15
|
Zou J, Shankar N. Roles of TLR/MyD88/MAPK/NF-κB Signaling Pathways in the Regulation of Phagocytosis and Proinflammatory Cytokine Expression in Response to E. faecalis Infection. PLoS One 2015; 10:e0136947. [PMID: 26317438 PMCID: PMC4552673 DOI: 10.1371/journal.pone.0136947] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022] Open
Abstract
Enterococcus faecalis is a commensal bacterium residing in the gastrointestinal tract of mammals, but in certain situations it is also an opportunistic pathogen which can cause serious disease. Macrophages have been shown to play a critical role in controlling infections by commensal enterococci and also have an important role in mediating chromosomal instability and promoting colon cancer during high-level enterococcal colonization in genetically susceptible mice. However, the molecular mechanisms involved in the interaction of macrophages with enterococci during infection are not fully understood. In this study, using BMDM and RAW264.7 macrophages we show that enterococcal infection activates ERK, JNK and p38 MAPK as well as NF-κB, and drives polarization of macrophages towards the M1 phenotype. Inhibition of NF-κB activation significantly reduced the expression of TNF-α and IL-1β, as did the inhibition of ERK, JNK and p38 MAPK, although to differing extent. Enterococci-induced activation of these pathways and subsequent cytokine expression was contact dependent, modest compared to activation by E. coli and, required the adaptor protein MyD88. Phagocytosis of enterococci by macrophages was enhanced by preopsonization with E. faecalis antiserum and involved the ERK and JNK signaling pathways, with the adaptor protein MyD88 as an important mediator. This study of the interaction of macrophages with enterococci could provide a foundation for studying the pathogenesis of infection by this opportunistic pathogen and to developing new therapeutic approaches to combat enterococcal infection.
Collapse
Affiliation(s)
- Jun Zou
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Nathan Shankar
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
16
|
Theilacker C, Diederich AK, Otto A, Sava IG, Wobser D, Bao Y, Hese K, Broszat M, Henneke P, Becher D, Huebner J. Enterococcus faecalis Glycolipids Modulate Lipoprotein-Content of the Bacterial Cell Membrane and Host Immune Response. PLoS One 2015; 10:e0132949. [PMID: 26172831 PMCID: PMC4501811 DOI: 10.1371/journal.pone.0132949] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 06/21/2015] [Indexed: 12/25/2022] Open
Abstract
In this study, we investigated the impact of the cell membrane composition of E. faecalis on its recognition by the host immune system. To this end, we employed an E. faecalis deletion mutant (ΔbgsA) that does not synthesize the major cell membrane glycolipid diglycosyl-diacylglycerol (DGlcDAG). Proteomic analysis revealed that 13 of a total of 21 upregulated surface-associated proteins of E. faecalis ΔbgsA were lipoproteins. This led to a total lipoprotein content in the cell membrane of 35.8% in ΔbgsA compared to only 9.4% in wild-type bacteria. Increased lipoprotein content strongly affected the recognition of ΔbgsA by mouse macrophages in vitro with an increased stimulation of TNF-α production by heat-fixed bacteria and secreted antigens. Inactivation of the prolipoprotein diacylglycerol transferase (lgt) in ΔbgsA abrogated TNF-α induction by a ΔbgsA_lgt double mutant indicating that lipoproteins mediate increased activation of mouse macrophages by ΔbgsA. Heat-fixed ΔbgsA bacteria, culture supernatant, or cell membrane lipid extract activated transfected HEK cells in a TLR2-dependent fashion; the same was not true of wild-type bacteria. In mice infected intraperitoneally with a sublethal dose of E. faecalis we observed a 70% greater mortality in mice infected with ΔbgsA compared with wild-type-infected mice. Increased mortality due to ΔbgsA infection was associated with elevated plasma levels of the inflammatory cytokines TNF-α, IL-6 and MIP-2. In summary, our results provide evidence that an E. faecalis mutant lacking its major bilayer forming glycolipid DGlcDAG upregulates lipoprotein expression leading to increased activation of the host innate immune system and virulence in vivo.
Collapse
Affiliation(s)
- Christian Theilacker
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- * E-mail:
| | - Ann-Kristin Diederich
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
- Department of Microbiology, Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Andreas Otto
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Irina G. Sava
- Research Center for Nutrition and Food Science, Technical University Munich, Freising, Germany
| | - Dominique Wobser
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Yinyin Bao
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Katrin Hese
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
| | - Melanie Broszat
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Paediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Dörte Becher
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Johannes Huebner
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
17
|
Fensterseifer ICM, Silva ON, Malik U, Ravipati AS, Novaes NRF, Miranda PRR, Rodrigues EA, Moreno SE, Craik DJ, Franco OL. Effects of cyclotides against cutaneous infections caused by Staphylococcus aureus. Peptides 2015; 63:38-42. [PMID: 25451333 DOI: 10.1016/j.peptides.2014.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/31/2014] [Accepted: 10/31/2014] [Indexed: 12/31/2022]
Abstract
The main bacterium associated with skin infection is Staphylococcus aureus, occurring especially in infections acquired via surgical wounds, commonly leading to lethal hospital-acquired infections, emphasizing the importance of identifying new antimicrobial compounds. Among them, cyclotides have gained interest due to their high stability and multifunctional properties. Here, cycloviolacin 2 (CyO2) and kalata B2 (KB2) were evaluated to determinate their anti-staphylococcal activities using a subcutaneous infection model. Anti-staphylococcal activities of 50mM for KB2 and 25mM for CyO2 were detected with no cytotoxic activities against RAW 264.7 monocytes. In the in vivo assays, both cyclotides reduced bacterial load and CyO2 demonstrated an increase in the phagocytosis index, suggesting that the CyO2 in vivo anti-staphylococcal activity may be associated with phagocytic activity, additionally to direct anti-pathogenic activity.
Collapse
Affiliation(s)
- Isabel C M Fensterseifer
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil; Molecular Pathology Post-graduate Program, University of Brasilia, Brasília 70910-900, Brazil
| | - Osmar N Silva
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil
| | - Uru Malik
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia
| | - Anjaneya S Ravipati
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia
| | - Natasha R F Novaes
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil
| | - Paulo R R Miranda
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil
| | - Elaine A Rodrigues
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil
| | - Susana E Moreno
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil
| | - David J Craik
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, Queensland, Australia
| | - Octavio L Franco
- Centro de Análises, Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790-160 Brazil; Molecular Pathology Post-graduate Program, University of Brasilia, Brasília 70910-900, Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; S-Inova, Pos-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Caampo Grande, MS, Brazil.
| |
Collapse
|
18
|
Johannessen M, Askarian F, Sangvik M, Sollid JE. Bacterial interference with canonical NFκB signalling. MICROBIOLOGY-SGM 2013; 159:2001-2013. [PMID: 23873783 PMCID: PMC3799228 DOI: 10.1099/mic.0.069369-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The human body is constantly challenged by a variety of commensal and pathogenic micro-organisms that trigger the immune system. Central in the first line of defence is the pattern-recognition receptor (PRR)-induced stimulation of the NFκB pathway, leading to NFκB activation. The subsequent production of pro-inflammatory cytokines and/or antimicrobial peptides results in recruitment of professional phagocytes and bacterial clearance. To overcome this, bacteria have developed mechanisms for targeted interference in every single step in the PRR–NFκB pathway to dampen host inflammatory responses. This review aims to briefly overview the PRR–NFκB pathway in relation to the immune response and give examples of the diverse bacterial evasion mechanisms including changes in the bacterial surface, decoy production and injection of effector molecules. Targeted regulation of inflammatory responses is needed and bacterial molecules developed for immune evasion could provide future anti-inflammatory agents.
Collapse
Affiliation(s)
- Mona Johannessen
- Research Group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Fatemeh Askarian
- Research Group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Maria Sangvik
- Research Group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Johanna E Sollid
- Research Group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
19
|
Teixeira N, Varahan S, Gorman MJ, Palmer KL, Zaidman-Remy A, Yokohata R, Nakayama J, Hancock LE, Jacinto A, Gilmore MS, de Fátima Silva Lopes M. Drosophila host model reveals new enterococcus faecalis quorum-sensing associated virulence factors. PLoS One 2013; 8:e64740. [PMID: 23734216 PMCID: PMC3667150 DOI: 10.1371/journal.pone.0064740] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 04/17/2013] [Indexed: 01/30/2023] Open
Abstract
Enterococcus faecalis V583 is a vancomycin-resistant clinical isolate which belongs to the hospital-adapted clade, CC2. This strain harbours several factors that have been associated with virulence, including the fsr quorum-sensing regulatory system that is known to control the expression of GelE and SprE proteases. To discriminate between genes directly regulated by Fsr, and those indirectly regulated as the result of protease expression or activity, we compared gene expression in isogenic mutants of V583 variously defective in either Fsr quorum sensing or protease expression. Quorum sensing was artificially induced by addition of the quorum signal, GBAP, exogenously in a controlled manner. The Fsr regulon was found to be restricted to five genes, gelE, sprE, ef1097, ef1351 and ef1352. Twelve additional genes were found to be dependent on the presence of GBAP-induced proteases. Induction of GelE and SprE by GBAP via Fsr resulted in accumulation of mRNA encoding lrgAB, and this induction was found to be lytRS dependent. Drosophila infection was used to discern varying levels of toxicity stemming from mutations in the fsr quorum regulatory system and the genes that it regulates, highlighting the contribution of LrgAB and bacteriocin EF1097 to infection toxicity. A contribution of SprE to infection toxicity was also detected. This work brought to light new players in E. faecalis success as a pathogen and paves the way for future studies on host tolerance mechanisms to infections caused by this important nosocomial pathogen.
Collapse
Affiliation(s)
- Neuza Teixeira
- ITQB Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Departments of Ophthalmology, and Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- CEDOC Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Sriram Varahan
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Matthew J. Gorman
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Kelli L. Palmer
- Departments of Ophthalmology, and Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anna Zaidman-Remy
- CEDOC Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ryoji Yokohata
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Jiro Nakayama
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Lynn E. Hancock
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - António Jacinto
- CEDOC Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Michael S. Gilmore
- Departments of Ophthalmology, and Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maria de Fátima Silva Lopes
- ITQB Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- IBET Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
20
|
Enterococcus faecalis overcomes foreign body-mediated inflammation to establish urinary tract infections. Infect Immun 2012; 81:329-39. [PMID: 23132492 DOI: 10.1128/iai.00856-12] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Urinary catheterization elicits major histological and immunological changes that render the bladder susceptible to microbial invasion, colonization, and dissemination. However, it is not understood how catheters induce these changes, how these changes act to promote infection, or whether they may have any protective benefit. In the present study, we examined how catheter-associated inflammation impacts infection by Enterococcus faecalis, a leading cause of catheter-associated urinary tract infection (CAUTI), a source of significant societal and clinical challenges. Using a recently optimized murine model of foreign body-associated UTI, we found that the implanted catheter itself was the primary inducer of inflammation. In the absence of the silicone tubing implant, E. faecalis induced only minimal inflammation and was rapidly cleared from the bladder. The catheter-induced inflammation was only minimally altered by subsequent enterococcal infection and was not suppressed by inhibitors of the neurogenic pathway and only partially by dexamethasone. Despite the robust inflammatory response induced by urinary implantation, E. faecalis produced biofilm and high bladder titers in these animals. Induction of inflammation in the absence of an implanted catheter failed to promote infection, suggesting that the presence of the catheter itself is essential for E. faecalis persistence in the bladder. Immunosuppression prior to urinary catheterization enhanced E. faecalis colonization, suggesting that implant-mediated inflammation contributes to the control of enterococcal infection. Thus, this study underscores the need for novel strategies against CAUTIs that seek to reduce the deleterious effects of implant-mediated inflammation on bladder homeostasis while maintaining an active immune response that effectively limits bacterial invaders.
Collapse
|
21
|
Wang J, Roderiquez G, Norcross MA. Control of adaptive immune responses by Staphylococcus aureus through IL-10, PD-L1, and TLR2. Sci Rep 2012; 2:606. [PMID: 22930672 PMCID: PMC3428601 DOI: 10.1038/srep00606] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/25/2012] [Indexed: 11/09/2022] Open
Abstract
Microbes induce innate immune responses in hosts. It is critical to know how different microbes control adaptive responses through innate pathways. The impact of gram-positive bacteria on the innate and adaptive responses is unclear. Herein we report that Staphylococcus aureus induces IL-10, Th17-inducing cytokines IL-6 and IL-23, chemokines, and regulates dendritic cell markers. S. aureus inhibits T-cell IL-2 responses through modulation of HLA-DR, CD86 and PD-L1. IFN-gamma, Src kinase inhibitors, or TLR2 antibodies prevented the down-modulation of HLA-DR by S. aureus. Our data demonstrate that innate TLR signaling induces multi-dimensional inhibition of adaptive immune responses, which may contribute to the lack of protective immunity to bacteria or microbe tolerance. IL-10 and PD-L1 antagonists may boost immunity to vaccines for S. aureus and other microbes.
Collapse
Affiliation(s)
- Jinhai Wang
- Laboratory of Immunology, Division of Therapeutic Proteins, Office of Biotechnology Products, Center for Drug Evaluation and Research , Food and Drug Administration, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
22
|
Traore K, Zirkin B, Thimmulappa RK, Biswal S, Trush MA. Upregulation of TLR1, TLR2, TLR4, and IRAK-2 Expression During ML-1 Cell Differentiation to Macrophages: Role in the Potentiation of Cellular Responses to LPS and LTA. ISRN ONCOLOGY 2012; 2012:641246. [PMID: 22685674 PMCID: PMC3364600 DOI: 10.5402/2012/641246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 03/06/2012] [Indexed: 12/27/2022]
Abstract
12-O-tetradecanoylphorbol 13-acetate (TPA) induces the differentiation of human myeloid ML-1 cells to macrophages. In the current study, the expression, responsiveness, and regulation of toll-like receptors (TLRs) in TPA-induced ML-1-derived macrophages were investigated. We have found that TPA-induced differentiation of ML-1 cells was accompanied by the upregulation of TLR1, TLR2, TLR4, and CD14 expression at both the mRNA and protein levels. Interestingly, TLR1 and TLR4 protein expression on ML-1 cells could be blocked by pretreatment with U0126, suggesting the role of an Erk1/2-induced differentiation signal in this process. In addition, the expression of IRAK-2, a key member of the TLR/IRAK-2/NF-κB-dependent signaling cascade was also induced in response to TPA. Accordingly, we demonstrated an increased cellular release of inflammatory cytokines (TNF-α and various interleukins) upon stimulation with LPS and LTA ligands for TLR4 and TLR2, respectively. Furthermore, using luminol-dependent chemiluminescence, addition of LPS and LTA induces a sustained DPI-inhibitable generation of reactive oxygen species (ROS) by the differentiated ML-1 cells. Together, these data suggest that the increase in the responsiveness of TPA-treated ML-1 cells to LPS and LTA occurs in response to the upregulation of their respective receptors as well as an induction of the IRAK-2 gene expression.
Collapse
Affiliation(s)
- Kassim Traore
- Department of Chemistry Geology & Physics, Elizabeth City State University, Elizabeth City, NC 27909, USA
| | | | | | | | | |
Collapse
|
23
|
Oliveira-Nascimento L, Massari P, Wetzler LM. The Role of TLR2 in Infection and Immunity. Front Immunol 2012; 3:79. [PMID: 22566960 PMCID: PMC3342043 DOI: 10.3389/fimmu.2012.00079] [Citation(s) in RCA: 493] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 03/28/2012] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptors (TLRs) are recognition molecules for multiple pathogens, including bacteria, viruses, fungi, and parasites. TLR2 forms heterodimers with TLR1 and TLR6, which is the initial step in a cascade of events leading to significant innate immune responses, development of adaptive immunity to pathogens and protection from immune sequelae related to infection with these pathogens. This review will discuss the current status of TLR2 mediated immune responses by recognition of pathogen-associated molecular patterns (PAMPS) on these organisms. We will emphasize both canonical and non-canonical responses to TLR2 ligands with emphasis on whether the inflammation induced by these responses contributes to the disease state or to protection from diseases.
Collapse
Affiliation(s)
- Laura Oliveira-Nascimento
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine Boston, MA, USA
| | | | | |
Collapse
|
24
|
Daw K, Baghdayan AS, Awasthi S, Shankar N. Biofilm and planktonic Enterococcus faecalis elicit different responses from host phagocytes in vitro. ACTA ACUST UNITED AC 2012; 65:270-82. [PMID: 22333034 DOI: 10.1111/j.1574-695x.2012.00944.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 02/08/2012] [Accepted: 02/08/2012] [Indexed: 12/12/2022]
Abstract
Enterococcus faecalis is a commensal organism of the gastrointestinal tract but can also cause serious opportunistic infections. In addition to high levels of antibiotic resistance, the ability to form biofilms on abiotic surfaces and on in-dwelling devices within the host complicates treatment strategies and successful outcomes of antibiotic therapy. Despite rapid advances made in recent years in understanding the genomics and virulence of this organism, much remains to be learned regarding the host response to enterococcal infections. In this study, we investigated the interaction of RAW264.7 macrophages and JAWS II dendritic cells with biofilm and planktonic E. faecalis, in vitro. Specifically, we compared phagocytosis, intracellular survival, secretion of proinflammatory cytokines, and the activation and maturation of phagocytes. Our results revealed that both macrophages and dendritic cells phagocytize biofilm mode cells at levels equal to or better than their planktonic counterparts. Internalized biofilm bacteria showed relatively greater survival at 24 h in macrophages than in dendritic cells and led to slightly higher expression of phagocyte activation markers. Macrophages infected with biofilm cells also secreted lower levels of proinflammatory cytokines studied. Overall, these results suggest that biofilm E. faecalis may be better adapted to overcome host defenses in vivo.
Collapse
Affiliation(s)
- Kasturee Daw
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
| | | | | | | |
Collapse
|
25
|
Lee HK, Park DW, Bae JH, Kim HJ, Shin DG, Park JS, Lee JG, Lee SJ, Bae YS, Baek SH. RGS2 is a negative regulator of STAT3-mediated Nox1 expression. Cell Signal 2012; 24:803-9. [DOI: 10.1016/j.cellsig.2011.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 11/07/2011] [Indexed: 10/15/2022]
|
26
|
Burger-van Paassen N, van der Sluis M, Bouma J, Korteland-van Male AM, Lu P, Van Seuningen I, Boehm G, van Goudoever JB, Renes IB. Colitis development during the suckling-weaning transition in mucin Muc2-deficient mice. Am J Physiol Gastrointest Liver Physiol 2011; 301:G667-78. [PMID: 21700902 DOI: 10.1152/ajpgi.00199.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The mucin Muc2 is the structural component of the colonic mucus layer. Adult Muc2 knockout (Muc2(-/-)) mice suffer from severe colitis. We hypothesized that Muc2 deficiency induces inflammation before weaning of mother's milk [postnatal day (P) 14] with aggravation of colitis after weaning (P28). Muc2(-/-) and wild-type mice were killed at embryonic day 18.5 and P1.5, P7.5, P14, P21, and P28. Colonic morphology, influx of T cells, and goblet cell-specific protein expression was investigated by (immuno)histochemistry. Cytokine and Toll-like receptor (TLR) profiles in the colon were analyzed by quantitative RT-PCR. Muc2(-/-) mice showed an increased and persistent influx of Cd3ε-positive T cells in the colonic mucosa as of P1.5. This was accompanied by mucosal damage at P28 in the distal colon but not in the proximal colon. At P14, the proinflammatory immune response [i.e., increased interleukin (IL)-12 p35, IL-12 p40, and tumor necrosis factor-α, expression] in the distal colon of Muc2(-/-) mice presented with an immune suppressive response [i.e., increased Foxp3, transforming growth factor (TGF)-β1, IL-10, and Ebi3 expression]. In contrast, at P28, a proinflammatory response remained in the distal colon, whereas the immune suppressive response (i.e., Foxp3 and TGF-β1 expression) declined. The proximal colon of Muc2(-/-) mice did not show morphological damage and was dominated by an immune suppressive response at P14 and P28. Interestingly, changes in expression of TLRs and TLR-related molecules were observed in the distal colon at P14 and P28 and in the proximal colon only at P28. Colitis in Muc2(-/-) mice is limited before weaning by immune suppressive responses and exacerbates in the distal colon after weaning because of the decline in the immune suppressive response.
Collapse
|
27
|
Leichtle A, Lai Y, Wollenberg B, Wasserman SI, Ryan AF. Innate signaling in otitis media: pathogenesis and recovery. Curr Allergy Asthma Rep 2011; 11:78-84. [PMID: 21049294 PMCID: PMC3020300 DOI: 10.1007/s11882-010-0158-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Otitis media (OM) is the most prevalent childhood disease in developed countries. Involvement of innate immunity mediated by Toll-like receptors (TLRs) in OM has been implicated primarily in cell lines and by association studies of innate immune gene polymorphisms with OM prevalence. However, the precise role of innate immunity in OM is incompletely understood. We review recent research that has advanced our understanding of how innate immunity in the middle ear is mediated by the interaction of pathogen molecules with receptors such as the TLRs, leading to the activation of adaptor molecules and production of proinflammatory cytokines. TLR genes and signaling molecules are upregulated in OM in a murine model. Deletion of several key innate immune genes results in persistent OM in mice, coupled with an inability to clear bacterial infection from the middle ear. It is concluded that an intact innate immune signaling system is critical to recovery from bacterial OM.
Collapse
Affiliation(s)
- Anke Leichtle
- Department of Otolaryngology, University of Lübeck, Ratzeburger Allee 160, 23564, Lübeck, Germany.
| | | | | | | | | |
Collapse
|
28
|
Hoffmann M, Messlik A, Kim SC, Sartor RB, Haller D. Impact of a probiotic Enterococcus faecalis
in a gnotobiotic mouse model of experimental colitis. Mol Nutr Food Res 2011; 55:703-13. [DOI: 10.1002/mnfr.201000361] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 11/12/2010] [Accepted: 11/15/2010] [Indexed: 12/12/2022]
|
29
|
Abstract
Enterococcus faecalis and Enterococcus faecium have emerged as multi-resistant nosocomial pathogens in immunocompromised and critically ill patients. Multi-resistant strains have acquired virulence genes resulting in hospital-adapted clones. The following review summarizes several proteins and carbohydrate- or glycoconjugates that have been identified as putative virulence factors involved in the pathogenesis of enterococcal infections and may be used as targets for alternative therapies. Several studies describing the host immune response against enterococci are also summarized.
Collapse
Affiliation(s)
- I G Sava
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Germany
| | | | | |
Collapse
|
30
|
Gröbner S, Fritz E, Schoch F, Schaller M, Berger AC, Bitzer M, Autenrieth IB. Lysozyme activates Enterococcus faecium to induce necrotic cell death in macrophages. Cell Mol Life Sci 2010; 67:3331-44. [PMID: 20458518 PMCID: PMC11115887 DOI: 10.1007/s00018-010-0384-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/31/2010] [Accepted: 04/22/2010] [Indexed: 11/29/2022]
Abstract
Enterococci are commensal organisms in the alimentary tract. However, they can cause a variety of life-threatening infections, especially in nosocomial settings. We hypothesized that induction of cell death might enable these facultative pathogenic bacteria to evade the innate immune response and to cause infections of their host. We demonstrate that E. faecium when exposed to lysozyme induces cell death in macrophages in vitro and in vivo. Flow cytometric analyses of J774A.1 macrophages infected with E. faecium revealed loss of cell membrane integrity indicated by uptake of propidium iodide and decrease of the inner mitochondrial transmembrane potential DeltaPsi(m). Inhibition of caspases, treatment of macrophages with cytochalasin D, or rifampicin did not prevent cells from dying, suggesting cell death mechanisms that are independent of caspase activation, bacterial uptake, and intracellular bacterial replication. Characteristics of necrotic cell death were demonstrated by both lack of procaspase 3 activation and cell shrinkage, electron microscopy, and release of lactate dehydrogenase. Pretreatment of E. faecium with lysozyme and subsequently with broad spectrum protease considerably reduced cell death, suggesting that a bacterial surface protein is causative for cell death induction. Moreover, in a mouse peritonitis model we demonstrated that E. faecium induces cell death of peritoneal macrophages in vivo. Altogether, our results show that enterococci, under specific conditions such as exposure to lysozyme, induce necrotic cell death in macrophages, which might contribute to disseminated infections by these facultative pathogenic bacteria.
Collapse
Affiliation(s)
- Sabine Gröbner
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Fabrizio K, Manix C, Tian H, van Rooijen N, Pirofski LA. The efficacy of pneumococcal capsular polysaccharide-specific antibodies to serotype 3 Streptococcus pneumoniae requires macrophages. Vaccine 2010; 28:7542-50. [PMID: 20800700 DOI: 10.1016/j.vaccine.2010.08.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 08/01/2010] [Accepted: 08/12/2010] [Indexed: 10/19/2022]
Abstract
The efficacy of antibody immunity against Streptococcus pneumoniae stems from the ability of opsonic, serotype (ST)-specific antibodies to pneumococcal capsular polysaccharide (PPS) to facilitate killing of the homologous ST by host phagocytes. However, PPS-specific antibodies have been identified that are protective in mice, but do not promote opsonic killing in vitro, raising the question of how they mediate protection in vivo. To probe this question, we investigated the dependence of antibody efficacy against lethal systemic (intraperitoneal, i.p.) infection with Streptococcus pneumoniae serotype 3 (ST3) on macrophages and neutrophils for the following PPS3-specific monoclonal antibodies (MAbs) in survival experiments in mice using a non-opsonic human IgM (A7), a non-opsonic mouse IgG1 (1E2) and an opsonic mouse IgG1 (5F6). The survival of A7- and PPS3-specific and isotype control MAb-treated neutrophil-depleted and neutrophil-sufficient and macrophage-depleted and macrophage-sufficient mice were determined after i.p. challenge with ST3 strains 6303 and WU2. Neutrophils were dispensable for A7 and the mouse MAbs to mediate protection in this model, but macrophages were required for the efficacy of A7 and optimal mouse MAb-mediated protection. For A7-treated mice, macrophage-depleted mice had higher blood CFU, cytokines and peripheral neutrophil levels than macrophage-sufficient mice, and macrophage-sufficient mice had lower tissue bacterial burdens than control MAb-treated mice. These findings demonstrate that macrophages contribute to opsonic and non-opsonic PPS3-specific MAb-mediated protection against ST3 infection by enhancing bacterial clearance and suggest that neutrophils do not compensate for the absence of macrophages in the model used in this study.
Collapse
Affiliation(s)
- Kevin Fabrizio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
32
|
Loof TG, Goldmann O, Gessner A, Herwald H, Medina E. Aberrant inflammatory response to Streptococcus pyogenes in mice lacking myeloid differentiation factor 88. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:754-63. [PMID: 20019195 DOI: 10.2353/ajpath.2010.090422] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several in vitro studies have emphasized the importance of toll-like receptor/myeloid differentiation factor 88 (MyD88) signaling in the inflammatory response to Streptococcus pyogenes. Since the extent of inflammation has been implicated in the severity of streptococcal diseases, we have examined here the role of toll-like receptor/MyD88 signaling in the pathophysiology of experimental S. pyogenes infection. To this end, we compared the response of MyD88-knockout (MyD88(-/-)) after subcutaneous inoculation with S. pyogenes with that of C57BL/6 mice. Our results show that MyD88(-/-) mice harbored significantly more bacteria in the organs and succumbed to infection much earlier than C57BL/6 animals. Absence of MyD88 resulted in diminished production of inflammatory cytokines such as interleukin-12, interferon-gamma, and tumor necrosis factor-alpha as well as chemoattractants such as monocyte chemotactic protein-1 (MCP-1) and Keratinocyte-derived chemokine (KC), and hampered recruitment of effector cells involved in bacterial clearance (macrophages and neutrophils) to the infection site. Furthermore, MyD88(-/-) but not C57BL/6 mice exhibited a massive infiltration of eosinophils in infected organs, which can be explained by an impaired production of the regulatory chemokines, gamma interferon-induced monokine (MIG/CXCL9) and interferon-induced protein 10 (IP-10/CXCL10), which can inhibit transmigration of eosinophils. Our results indicate that MyD88 signaling targets effector cells to the site of streptococcal infection and prevents extravasation of cells that can induce tissue damage. Therefore, MyD88 signaling may be important for shaping the quality of the inflammatory response elicited during infection to ensure optimal effector functions.
Collapse
Affiliation(s)
- Torsten G Loof
- Department of Microbial Pathogenesis, Infection Immunology Research Group, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
33
|
Enterococcus faecalis capsular polysaccharide serotypes C and D and their contributions to host innate immune evasion. Infect Immun 2009; 77:5551-7. [PMID: 19805541 DOI: 10.1128/iai.00576-09] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has become increasingly difficult to treat infections caused by Enterococcus faecalis due to its high levels of intrinsic and acquired antibiotic resistance. However, few studies have explored the mechanisms that E. faecalis employs to circumvent the host innate immune response and establish infection. Capsular polysaccharides are important virulence factors that are associated with innate immune evasion. We demonstrate, using cultured macrophages (RAW 264.7), that capsule-producing E. faecalis strains of either serotype C or D are more resistant to complement-mediated opsonophagocytosis than unencapsulated strains. We show that differences in opsonophagocytosis are not due to variations in C3 deposition but are due to the ability of capsule to mask bound C3 from detection on the surface of E. faecalis. Similarly, E. faecalis capsule masks lipoteichoic acid from detection, which correlates with decreased tumor necrosis factor alpha production by cultured macrophages in the presence of encapsulated strains compared to that in the presence of unencapsulated strains. Our studies confirm the important role of the capsule as a virulence factor of E. faecalis and provide several mechanisms by which the presence of the capsule influences evasion of the innate immune response and suggest that the capsule could be a potential target for developing alternative therapies to treat E. faecalis infections.
Collapse
|
34
|
Capsular polysaccharide production in Enterococcus faecalis and contribution of CpsF to capsule serospecificity. J Bacteriol 2009; 191:6203-10. [PMID: 19684130 DOI: 10.1128/jb.00592-09] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many bacterial species produce capsular polysaccharides that contribute to pathogenesis through evasion of the host innate immune system. The gram-positive pathogen Enterococcus faecalis was previously reported to produce one of four capsule serotypes (A, B, C, or D). Previous studies describing the four capsule serotypes of E. faecalis were based on immunodetection methods; however, the underlying genetics of capsule production did not fully support these findings. Previously, it was shown that capsule production for serotype C (Maekawa type 2) was dependent on the presence of nine open reading frames (cpsC to cpsK). Using a novel genetic system, we demonstrated that seven of the nine genes in the cps operon are essential for capsule production, indicating that serotypes A and B do not make a capsular polysaccharide. In support of this observation, we showed that serotype C and D capsule polysaccharides mask lipoteichoic acid from detection by agglutinating antibodies. Furthermore, we determined that the genetic basis for the difference in antigenicity between serotypes C and D is the presence of cpsF in serotype C strains. High-pH anion-exchange chromatography with pulsed amperometric detection analysis of serotype C and D capsules indicated that cpsF is responsible for glucosylation of serotype C capsular polysaccharide in E. faecalis.
Collapse
|
35
|
Leichtle A, Hernandez M, Pak K, Webster NJ, Wasserman SI, Ryan AF. The toll-Like receptor adaptor TRIF contributes to otitis media pathogenesis and recovery. BMC Immunol 2009; 10:45. [PMID: 19656404 PMCID: PMC2736931 DOI: 10.1186/1471-2172-10-45] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 08/05/2009] [Indexed: 11/17/2022] Open
Abstract
Background Toll-like receptor (TLR) signalling is crucial for innate immune responses to infection. The involvement of TLRs in otitis media (OM), the most prevalent childhood disease in developed countries, has been implicated by studies in middle ear cell lines, by association studies of TLR-related gene polymorphisms, and by altered OM in mice bearing mutations in TLR genes. Activated TLRs signal via two alternative intracellular signaling molecules with differing effects; MyD88 (Myeloid differentiation primary response gene 88) inducing primarily interleukin expression and TRIF (Tir-domain-containing adaptor inducing interferon β) mediating type I interferon (IFN) expression. We tested the hypothesis that TRIF and type I IFN signaling play a role in OM, using a murine model of OM induced by non-typeable Haemophilus influenzae (NTHi). The ME inflammatory response to NTHi was examined in wild-type (WT) and TRIF-/- mice by qPCR, gene microarray, histopathology and bacterial culture. Results Expression of TRIF mRNA was only modesty enhanced during OM, but both type I IFN signalling genes and type I IFN-inducible genes were significantly up-regulated in WT mice. TRIF-deficient mice showed reduced but more persistent mucosal hyperplasia and less leukocyte infiltration into the ME in response to NTHi infection than did WT animals. Viable bacteria could be cultured from MEs of TRIF-/- mice for much longer in the course of disease than was the case for middle ears of WT mice. Conclusion Our results demonstrate that activation of TRIF/type I IFN responses is important in both the pathogenesis and resolution of NTHi-induced OM.
Collapse
Affiliation(s)
- Anke Leichtle
- Department of Surgery/Otolaryngology University of California, San Diego, 9500 Gilman Avenue, La Jolla, California 92093, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Leichtle A, Hernandez M, Pak K, Yamasaki K, Cheng CF, Webster NJ, Ryan AF, Wasserman SI. TLR4-mediated induction of TLR2 signaling is critical in the pathogenesis and resolution of otitis media. Innate Immun 2009; 15:205-15. [PMID: 19586996 DOI: 10.1177/1753425909103170] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Otitis media is the most prevalent childhood disease in developed countries. The involvement of Toll-like receptors (TLRs) in otitis media pathophysiology has been implicated by studies in cell lines and association studies of TLR gene polymorphisms. However, precise functions of TLRs in the etiology of otitis media in vivo have not been examined. We investigated the inflammatory response to nontypeable Haemophilus influenzae using a model of otitis media in wild-type, TLR2(- /-) and TLR4(-/ -) mice by gene microarray, qPCR, immunohistochemistry, Western blot analysis and histopathology. Toll-like receptor-2(- /-) and TLR4(- /-) mice exhibited a more profound, persistent inflammation with impaired bacterial clearance compared to controls. While wild-type mice induced tumor necrosis factor-a (TNF) after non-typeable H. influenzae challenge, TLR2(-/-) and TLR4(-/-) mice lack TNF induction in the early phase of otitis media. Moreover, lack of TLR2 resulted in a late increase in IL-10 expression and prolonged failure to clear bacteria. Toll-like receptor-4(-/- ) mice showed impaired early bacterial clearance and loss of TLR2 induction in early otitis media. Our results demonstrate that both TLR2 and TLR4 signalling are critical to the regulation of infection in non-typeable H. influenzae-induced otitis media. Toll-like receptor-4 signalling appears to induce TLR2 expression, and TLR2 activation is critical for bacterial clearance and timely resolution of otitis media.
Collapse
Affiliation(s)
- Anke Leichtle
- Department of Surgery/Otolaryngology, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Hovius JWR, Bijlsma MF, van der Windt GJW, Wiersinga WJ, Boukens BJD, Coumou J, Oei A, de Beer R, de Vos AF, van 't Veer C, van Dam AP, Wang P, Fikrig E, Levi MM, Roelofs JJTH, van der Poll T. The urokinase receptor (uPAR) facilitates clearance of Borrelia burgdorferi. PLoS Pathog 2009; 5:e1000447. [PMID: 19461880 PMCID: PMC2678258 DOI: 10.1371/journal.ppat.1000447] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 04/25/2009] [Indexed: 12/22/2022] Open
Abstract
The causative agent of Lyme borreliosis, the spirochete Borrelia
burgdorferi, has been shown to induce expression of the urokinase
receptor (uPAR); however, the role of uPAR in the immune response against
Borrelia has never been investigated. uPAR not only acts as
a proteinase receptor, but can also, dependently or independently of ligation to
uPA, directly affect leukocyte function. We here demonstrate that uPAR is
upregulated on murine and human leukocytes upon exposure to B.
burgdorferi both in vitro as well as in vivo. Notably, B.
burgdorferi-inoculated C57BL/6 uPAR knock-out mice harbored
significantly higher Borrelia numbers compared to WT controls.
This was associated with impaired phagocytotic capacity of B.
burgdorferi by uPAR knock-out leukocytes in vitro. B.
burgdorferi numbers in vivo, and phagocytotic capacity in vitro,
were unaltered in uPA, tPA (low fibrinolytic activity) and PAI-1 (high
fibrinolytic activity) knock-out mice compared to WT controls. Strikingly, in
uPAR knock-out mice partially backcrossed to a B. burgdorferi
susceptible C3H/HeN background, higher B. burgdorferi numbers
were associated with more severe carditis and increased local TLR2 and
IL-1β mRNA expression. In conclusion, in B. burgdorferi
infection, uPAR is required for phagocytosis and adequate eradication of the
spirochete from the heart by a mechanism that is independent of binding of uPAR
to uPA or its role in the fibrinolytic system. Lyme borreliosis is caused by the spirochete Borrelia
burgdorferi and is transmitted through ticks. Since its discovery
approximately 30 years ago it has become the most important vector-borne disease
in the Western world. The pathogenesis of this complex zoonosis is still not
entirely understood. We here demonstrate that the urokinase receptor (uPAR) is
upregulated in mice and humans upon exposure to B. burgdorferi
in vitro and in vivo. Importantly, we describe the function of uPAR in the
immune response against the spirochete; using uPAR knock-out mice, we show that
uPAR plays an important role in phagocytosis of B. burgdorferi
by leukocytes both in vitro as well as in vivo. In addition, we show that the
mechanism by which uPAR is involved in the phagocytosis of B.
burgdorferi is independent of ligation to its natural ligand uPA or
uPAR's role in fibrinolysis. Our study contributes to the understanding
of the pathogenesis of Lyme borreliosis and might contribute to the development
of innovative novel treatment strategies for Lyme borreliosis.
Collapse
Affiliation(s)
- Joppe W R Hovius
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, AMC, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Leendertse M, Willems RJL, Giebelen IAJ, Roelofs JJTH, van Rooijen N, Bonten MJM, van der Poll T. Peritoneal macrophages are important for the early containment of Enterococcus faecium peritonitis in mice. Innate Immun 2009; 15:3-12. [PMID: 19201820 DOI: 10.1177/1753425908100238] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The increasing incidence of infections with multi-drug resistant Enterococcus faecium necessitates studies to increase knowledge on the pathogenesis of these infections. In this study, the contribution of peritoneal macrophages during E. faecium peritonitis was investigated. In an ex vivo setting, peritoneal macrophages harvested from C57BL/6 mice were responsive to, and able to phagocytose and kill, E. faecium. In vivo, peritoneal macrophages were depleted by intraperitoneal injection of clodronate-encapsulated liposomes, prior to inducing E. faecium peritonitis. Depletion of resident peritoneal macrophages caused a clear delay in peritoneal clearance of E. faecium with increased systemic dissemination. Mice depleted of peritoneal macrophages were able to recruit macrophages and neutrophils to the peritoneal cavity after infection, comparable to control mice. Furthermore, increased levels of peritoneal cytokines and chemokines were found in mice depleted of peritoneal macrophages. This study indicates that peritoneal macrophages are important in the early containment of E. faecium peritonitis and for the regulation of the inflammatory response.
Collapse
Affiliation(s)
- Masja Leendertse
- Center for Infection and Immunity Amsterdam, Center for Experimental and Molecular Medicine.
| | | | | | | | | | | | | |
Collapse
|
39
|
Baqai FP, Gridley DS, Slater JM, Luo-Owen X, Stodieck LS, Ferguson V, Chapes SK, Pecaut MJ. Effects of spaceflight on innate immune function and antioxidant gene expression. J Appl Physiol (1985) 2009; 106:1935-42. [PMID: 19342437 DOI: 10.1152/japplphysiol.91361.2008] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Spaceflight conditions have a significant impact on a number of physiological functions due to psychological stress, radiation, and reduced gravity. To explore the effect of the flight environment on immunity, C57BL/6NTac mice were flown on a 13-day space shuttle mission (STS-118). In response to flight, animals had a reduction in liver, spleen, and thymus masses compared with ground (GRD) controls (P < 0.005). Splenic lymphocyte, monocyte/macrophage, and granulocyte counts were significantly reduced in the flight (FLT) mice (P < 0.05). Although spontaneous blastogenesis of splenocytes in FLT mice was increased, response to lipopolysaccharide (LPS), a B-cell mitogen derived from Escherichia coli, was decreased compared with GRD mice (P < 0.05). Secretion of IL-6 and IL-10, but not TNF-alpha, by LPS-stimulated splenocytes was increased in FLT mice (P < 0.05). Finally, many of the genes responsible for scavenging reactive oxygen species were upregulated after flight. These data indicate that exposure to the spaceflight environment can increase anti-inflammatory mechanisms and change the ex vivo response to LPS, a bacterial product associated with septic shock and a prominent Th1 response.
Collapse
Affiliation(s)
- Farnaz P Baqai
- Department of Radiation Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Leendertse M, Willems RJ, Giebelen IA, Florquin S, van den Pangaart PS, Bonten MJ, van der Poll T. Cecal ligation and puncture induced sepsis impairs host defense against Enterococcus faecium peritonitis. Intensive Care Med 2009; 35:924-32. [DOI: 10.1007/s00134-009-1440-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Accepted: 01/23/2009] [Indexed: 12/22/2022]
|
41
|
Brzezinska AA, Johnson JL, Munafo DB, Ellis BA, Catz SD. Signalling mechanisms for Toll-like receptor-activated neutrophil exocytosis: key roles for interleukin-1-receptor-associated kinase-4 and phosphatidylinositol 3-kinase but not Toll/IL-1 receptor (TIR) domain-containing adaptor inducing IFN-beta (TRIF). Immunology 2008; 127:386-97. [PMID: 19019092 DOI: 10.1111/j.1365-2567.2008.02980.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Lipopolysaccharide (LPS) stimulates exocytosis in neutrophils. The signalling molecules involved in the regulation of this mechanism are currently unknown. Using neutrophils from interleukin-1-receptor-associated kinase (IRAK)-4- and Toll/IL-1 receptor (TIR) domain-containing adaptor inducing IFN-beta (TRIF)-deficient mice, we dissected the signalling pathways that control exocytosis. We analysed exocytosis of peroxidase-negative and azurophilic granules by following the mobilization of the beta2-integrin subunit CD11b and myeloperoxidase (MPO)-containing granules, respectively. IRAK-4-null neutrophils showed marked defects in both peroxidase-negative and azurophilic granule exocytosis in response to LPS. In contrast, the exocytic response to LPS of TRIF-deficient neutrophils was not different from that of wild-type cells. No differences were observed in the exocytosis of secretory organelles between IRAK-4-null and wild-type neutrophils when they were stimulated with the phorbol ester phorbol 12-myristate 13-acetate (PMA). Electron microscopy analysis showed that no morphological abnormalities were present in the granules of IRAK-4-deficient neutrophils, suggesting that the lack of exocytic response to LPS is not attributable to developmental abnormalities. Using pharmacological inhibitors, we found that p38 mitogen-activated protein kinase (p38MAPK) is essential for the exocytosis of all neutrophil secretory organelles in response to LPS. Interestingly, we found that phosphatidylinositol 3-kinase (PI3K) is essential for azurophilic granule exocytosis but not for the mobilization of other neutrophil granules in response to LPS. Azurophilic granule exocytosis in response to Listeria monocytogenes was dependent on PI3K but not IRAK-4 activity, suggesting that alternative signalling pathways are activated in IRAK-4-deficient neutrophils exposed to whole bacteria. Our results identified IRAK-4, p38MAPK and PI3K as important regulatory components with different roles in the signalling pathways that control Toll-like receptor ligand-triggered neutrophil exocytosis.
Collapse
Affiliation(s)
- Agnieszka A Brzezinska
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
42
|
Differential mRNA expression of the avian-specific toll-like receptor 15 between heterophils from Salmonella-susceptible and -resistant chickens. Immunogenetics 2008; 61:71-7. [DOI: 10.1007/s00251-008-0340-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 10/22/2008] [Indexed: 01/13/2023]
|
43
|
Neutrophils are essential for rapid clearance of Enterococcus faecium in mice. Infect Immun 2008; 77:485-91. [PMID: 19001080 DOI: 10.1128/iai.00863-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A progressive increase in infections with multiresistant Enterococcus faecium has been reported, especially in cancer patients and neutropenic patients. Despite its increasing importance as a nosocomial pathogen, knowledge of the pathogenesis of E. faecium infections is highly limited. In this study, we investigated the role of neutrophils during peritonitis with subsequent bacteremia caused by E. faecium. Therefore, we depleted neutrophils by intraperitoneal injections of monoclonal antibody RB6-8C5. Mice were followed for 5 days, and the enterococcal outgrowth and inflammatory response were compared between neutropenic mice and immunoglobulin G-injected control mice. Neutropenic mice demonstrated a severe delay in enterococcal clearance from all cultured organs (peritoneal fluid, blood, and lung and liver tissue). In particular, neutropenic mice remained bacteremic for up to 3 days, whereas all nonneutropenic mice had cleared the bacteria from circulation by 2 days. Furthermore, neutropenic mice displayed elevated peritoneal cytokine and chemokine levels 1 day after the infection and attracted fewer macrophages into the peritoneal cavity. In the circulation, a prolonged elevation of tumor necrosis factor alpha, interleukin-6, and the acute-phase proteins serum amyloid A and complement 3 were measured in neutropenic mice. In conclusion, attraction of neutrophils to the primary site of E. faecium infection is important for a rapid clearance of this bacterium, thereby attenuating a systemic inflammatory response.
Collapse
|
44
|
Leendertse M, Willems RJL, Giebelen IAJ, van den Pangaart PS, Bonten MJM, van der Poll T. The acute-phase response impairs host defence against Enterococcus faecium peritonitis. Immunology 2008; 128:e335-42. [PMID: 19175794 DOI: 10.1111/j.1365-2567.2008.02967.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Enterococcus faecium is an emerging pathogen that causes infections in hospitalized patients with various co-morbid diseases. These underlying diseases are often associated with an acute-phase response that renders patients vulnerable to nosocomial infections. To study the influence of the acute-phase response induced by sterile tissue injury on host defence against E. faecium, mice were injected subcutaneously with either turpentine or casein 1 day before intraperitoneal infection with E. faecium. Control mice were subcutaneously injected with saline or sodium bicarbonate, respectively. Turpentine and casein induced an acute-phase response as reflected by increases in the plasma concentrations of interleukin-6, serum amyloid P and C3. A pre-existent acute-phase response in mice was associated with a strongly reduced capacity to clear E. faecium, resulting in prolonged bacteraemia for several days. The inflammatory response to E. faecium was impaired in mice with an acute-phase response, as shown by reduced capacity to mount a neutrophilic leucocytosis in peripheral blood and by decreased local cytokine concentrations. These data indicate that the acute-phase response impairs host defence against E. faecium, suggesting that this condition may contribute to the increased vulnerability of critically ill patients to enterococcal infections.
Collapse
Affiliation(s)
- Masja Leendertse
- Centre for Infection and Immunity Amsterdam (CINIMA), Amsterdam, the Netherlands.
| | | | | | | | | | | |
Collapse
|
45
|
MyD88 dependent signaling contributes to protective host defense against Burkholderia pseudomallei. PLoS One 2008; 3:e3494. [PMID: 18946505 PMCID: PMC2566818 DOI: 10.1371/journal.pone.0003494] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 10/01/2008] [Indexed: 01/07/2023] Open
Abstract
Background Toll-like receptors (TLRs) have a central role in the recognition of pathogens and the initiation of the innate immune response. Myeloid differentiation primary-response gene 88 (MyD88) and TIR-domain-containing adaptor protein inducing IFNβ (TRIF) are regarded as the key signaling adaptor proteins for TLRs. Melioidosis, which is endemic in SE-Asia, is a severe infection caused by the gram-negative bacterium Burkholderia pseudomallei. We here aimed to characterize the role of MyD88 and TRIF in host defense against melioidosis. Methodology and Principal Findings First, we found that MyD88, but not TRIF, deficient whole blood leukocytes released less TNFα upon stimulation with B. pseudomallei compared to wild-type (WT) cells. Thereafter we inoculated MyD88 knock-out (KO), TRIF mutant and WT mice intranasally with B. pseudomallei and found that MyD88 KO, but not TRIF mutant mice demonstrated a strongly accelerated lethality, which was accompanied by significantly increased bacterial loads in lungs, liver and blood, and grossly enhanced liver damage compared to WT mice. The decreased bacterial clearance capacity of MyD88 KO mice was accompanied by a markedly reduced early pulmonary neutrophil recruitment and a diminished activation of neutrophils after infection with B. pseudomallei. MyD88 KO leukocytes displayed an unaltered capacity to phagocytose and kill B. pseudomallei in vitro. Conclusions MyD88 dependent signaling, but not TRIF dependent signaling, contributes to a protective host response against B. pseudomallei at least in part by causing early neutrophil recruitment towards the primary site of infection.
Collapse
|