1
|
Chowdhury S, Sadhukhan P, Mahata N. Immunoinformatics investigation on pathogenic Escherichia coli proteome to develop an epitope-based peptide vaccine candidate. Mol Divers 2024:10.1007/s11030-024-11034-0. [PMID: 39516450 DOI: 10.1007/s11030-024-11034-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Escherichia coli (E. coli), a gram-negative bacterium, quickly colonizes in the human gastrointestinal tract after birth and typically sustains a long-term, symbiotic relationship with the host. However, certain virulent strains of E. coli can cause diseases such as urinary tract infections, meningitis, and enteric disorders. The rising antibiotic resistance among these strains has heightened the urgency for an effective vaccine. This study employs immunoinformatics and a reverse vaccinology technique to identify prospective antigens and create an efficient vaccine construct. In this study, we reported the "Attaching and Effacing Protein" a novel outer-membrane protein conserved in all pathogenic E. coli strains, based on proteome screening. We developed an in silico multi-epitope vaccine that includes helper T lymphocyte (HTL), cytotoxic T lymphocyte (CTL), B cell lymphocyte (BCL), and pan HLA DR-binding reactive epitope (PADRE) sequences, along with appropriate linkers and adjuvants. Machine Learning algorithms were used to evaluate antigenicity, solubility, stability, and non-allergenicity of the vaccine construct. Additionally, molecular docking analysis revealed that vaccine construct has a strong predicted binding affinity for human toll-like receptors on the cell surface. In this context, laboratory validations are necessary to demonstrate the effectiveness of the possible vaccine design that showed encouraging findings through computational validation.
Collapse
Affiliation(s)
- Soham Chowdhury
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Pinkan Sadhukhan
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| |
Collapse
|
2
|
Kalchev Y, Argirova P, Boev I, Yaneva A, Vatev N, Stoycheva M, Murdjeva M. Cytokine profile in patients with acute bacterial meningitis. Cytokine 2023; 170:156315. [PMID: 37544134 DOI: 10.1016/j.cyto.2023.156315] [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: 06/14/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Bacterial meningitis is a life-threatening disease with high mortality and common long-term sequelae. The inflammatory response in the subarachnoid space, modulated by different cytokines, plays a major role in the pathogenesis of acute central nervous system infections. We aimed to examine correlations of interleukin (IL)-6, IL-8, IL-10, IL-12(p40), and tumor necrosis factor (TNF)-α levels with disease severity, complications, and outcome in patients with acute bacterial meningitis. METHODS The study involved 30 patients with bacterial meningitis/meningoencephalitis admitted to the University Hospital St. George, Plovdiv over a period of 4 years. Patients were selected based on clinical presentation and laboratory abnormalities, consistent with a neuroinfection. Enzyme-linked immunosorbent assay was used to measure the studied cytokines in both cerebrospinal fluid (CSF) and serum in parallel. For microbiological diagnosis multiplex, polymerase chain reaction, and CSF culture were used. RESULTS In patients with acute bacterial meningitis CSF levels of IL-6, IL-8, IL-10, and TNF-α are significantly increased than in serum. CSF TNF-α, CSF IL-8, and CSF IL-10 had a moderate negative correlation to CSF glucose. It was found that serum IL-8 is significantly elevated in patients who experienced neurological complications, have severe clinical course, and in deceased patients. CSF IL-10 is increased only in patients with severe acute bacterial meningitis. CONCLUSION Among patients with acute bacterial meningitis serum IL-8 could delineate these with increased risk of neurological complications, severe clinical course, and fatal outcome. Serum IL-8 and CSF IL-10 could be used as indicators of disease severity.
Collapse
Affiliation(s)
- Y Kalchev
- Department of Medical Microbiology and Immunology "Prof. Dr. Elissay Yanev", Faculty of Pharmacy, Medical University - Plovdiv, Bulgaria; Laboratory of Microbiology, University Hospital St. George, Plovdiv, Bulgaria; Research Institute at Medical University - Plovdiv, Bulgaria.
| | - P Argirova
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria
| | - I Boev
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria
| | - A Yaneva
- Department of Medical Informatics, Biostatistics, and eLearning, Faculty of Public Health, Medical University - Plovdiv, Bulgaria
| | - N Vatev
- Department of Epidemiology and Disaster Medicine, Faculty of Public Health, Medical University - Plovdiv, Bulgaria
| | - M Stoycheva
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria; Clinic of Infectious Diseases, University Hospital St. George, Plovdiv, Bulgaria
| | - M Murdjeva
- Department of Medical Microbiology and Immunology "Prof. Dr. Elissay Yanev", Faculty of Pharmacy, Medical University - Plovdiv, Bulgaria; Laboratory of Microbiology, University Hospital St. George, Plovdiv, Bulgaria; Research Institute at Medical University - Plovdiv, Bulgaria
| |
Collapse
|
3
|
Manda-Handzlik A, Cieloch A, Kuźmicka W, Mroczek A, Stelmaszczyk-Emmel A, Demkow U, Wachowska M. Secretomes of M1 and M2 macrophages decrease the release of neutrophil extracellular traps. Sci Rep 2023; 13:15633. [PMID: 37730741 PMCID: PMC10511515 DOI: 10.1038/s41598-023-42167-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
The release of neutrophil extracellular traps (NETs) can be either beneficial or detrimental for the host, thus it is necessary to maintain a balance between formation and clearance of NETs. Multiple physiological factors eliciting NET release have been identified, yet the studies on natural signals limiting NET formation have been scarce. Accordingly, our aim was to analyze whether cytokines or immune cells can inhibit NET formation. To that end, human granulocytes were incubated with interleukin (IL)-4, IL-10, transforming growth factor beta-2 or adenosine and then stimulated to release NETs. Additionally, neutrophils were cultured in the presence of natural killer (NK) cells, regulatory T cells (Tregs), pro-inflammatory or anti-inflammatory macrophages (M1 or M2 macrophages), or in the presence of NK/Tregs/M1 macrophages or M2 macrophages-conditioned medium and subsequently stimulated to release NETs. Our studies showed that secretome of M1 and M2 macrophages, but not of NK cells and Tregs, diminishes NET formation. Co-culture experiments did not reveal any effect of immune cells on NET release. No effect of cytokines or adenosine on NET release was found. This study highlights the importance of paracrine signaling at the site of infection and is the first to show that macrophage secretome can regulate NET formation.
Collapse
Affiliation(s)
- Aneta Manda-Handzlik
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland.
| | - Adrianna Cieloch
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Weronika Kuźmicka
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Agnieszka Mroczek
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Małgorzata Wachowska
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| |
Collapse
|
4
|
Li J, Pu F, Peng C, Wang Y, Zhang Y, Wu S, Wang S, Shen X, Cheng R, He F. Antibiotic cocktail-induced gut microbiota depletion in different stages could cause host cognitive impairment and emotional disorders in adulthood in different manners. Neurobiol Dis 2022; 170:105757. [PMID: 35588989 DOI: 10.1016/j.nbd.2022.105757] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/23/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023] Open
Abstract
Gut microbiota depletion may result in cognitive impairment and emotional disorder. This study aimed to determine the possible association between host gut microbiota, cognitive function, and emotion in various life stages and its related underlying mechanisms. Seventy-five neonatal mice were randomly divided into five groups (n = 15 per group). Mice in the vehicle group were administered distilled water from birth to death, and those in the last four groups were administered antibiotic cocktail from birth to death, from birth to postnatal day (PND) 21 (infancy), from PND 21 to 56 (adolescence), and from PND 57 to 84 (adulthood), respectively. Antibiotic exposure consistently altered the gut microbiota composition and decreased the diversity of gut microbiota. Proteobacteria were the predominant bacteria instead of Firmicutes and Bacteroidetes after antibiotic exposure in different life stages. Long-term and infant gut microbiota depletion resulted in anxiety- and depression-like behaviors, memory impairments, and increased expression of γ-aminobutyric acid type A receptor α1 of adult mice. Long-term antibiotic exposure also significantly decreased serum interleukin (IL)-1β, IL-10, and corticosterone of adult mice. Gut microbiota depletion in adolescence resulted in anxiety-like behaviors, short-term memory decline, decreased serum interferon-γ (IFN-γ), mRNA expression of 5-hydroxytryptamine receptor 1A, and neuropeptide Y receptor Y2 in the prefrontal cortex of adult mice. Antibiotic exposure in adulthood damaged short-term memory and decreased serum IL-10, IFN-γ, and increased γ-aminobutyric acid type B receptor 1 mRNA expression of adult mice. These results suggest that antibiotic-induced gut microbiota depletion in the long term and infancy resulted in the most severe cognitive and emotional disorders followed by depletion in adolescence and adulthood. These results also suggest that gut microbes could influence host cognitive function and emotion in a life stage-dependent manner by affecting the function of the immune system, hypothalamic-pituitary-adrenal axis, and the expression of neurochemicals in the brain.
Collapse
Affiliation(s)
- Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Fangfang Pu
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Chenrui Peng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Yimei Wang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Yujie Zhang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Silu Wang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China.
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, PR China.
| |
Collapse
|
5
|
The Impact of Cytokines on Neutrophils' Phagocytosis and NET Formation during Sepsis-A Review. Int J Mol Sci 2022; 23:ijms23095076. [PMID: 35563475 PMCID: PMC9101385 DOI: 10.3390/ijms23095076] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is an overwhelming inflammatory response to infection, resulting in multiple-organ injury. Neutrophils are crucial immune cells involved in innate response to pathogens and their migration and effector functions, such as phagocytosis and neutrophil extracellular trap (NET) formation, are dependent on cytokine presence and their concentration. In the course of sepsis, recruitment and migration of neutrophils to infectious foci gradually becomes impaired, thus leading to loss of a crucial arm of the innate immune response to infection. Our review briefly describes the sepsis course, the importance of neutrophils during sepsis, and explains dependence between cytokines and their activation. Moreover, we, for the first time, summarize the impact of cytokines on phagocytosis and NET formation. We highlight and discuss the importance of cytokines in modulation of both processes and emphasize the direction of further investigations.
Collapse
|
6
|
Chambers CA, Dadelahi AS, Moley CR, Olson RM, Logue CM, Skyberg JA. Nucleotide receptors mediate protection against neonatal sepsis and meningitis caused by alpha-hemolysin expressing Escherichia coli K1. FASEB J 2022; 36:e22197. [PMID: 35147989 DOI: 10.1096/fj.202101485r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/30/2021] [Accepted: 01/20/2022] [Indexed: 01/15/2023]
Abstract
Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.
Collapse
Affiliation(s)
- Catherine A Chambers
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Alexis S Dadelahi
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Charles R Moley
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Rachel M Olson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Catherine M Logue
- Department of Population Heath, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jerod A Skyberg
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
7
|
Yanik BM, Dauch JR, Cheng HT. Interleukin-10 Reduces Neurogenic Inflammation and Pain Behavior in a Mouse Model of Type 2 Diabetes. J Pain Res 2020; 13:3499-3512. [PMID: 33402846 PMCID: PMC7778525 DOI: 10.2147/jpr.s264136] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Neurogenic inflammation is a major component of chronic neuropathic pain. Previously, we established the db/db mouse as an animal model of painful diabetic neuropathy (PDN) of type 2 diabetes. In the current study, we investigate the roles of interleukin (IL)-10, an anti-inflammatory cytokine, in the development of neurogenic inflammation and pain behavior in db/db mouse. Materials and Methods We first studied IL-10 expression in lumbar dorsal root ganglion (LDRG) neurons of db/db mice using immunohistochemistry, immunoblots, and reverse transcription polymerase chain reaction during the period of pain behavior (from 8 to 16 wk of age). To determine if the reduced IL-10 expression mediates the mechanical allodynia in db/db mice, we administered recombinant mouse IL-10 or saline (control) intraperitoneally to control db/+ and db/db mice starting at 8 wk of age. IL-10 treatment was repeated every other day for 2 wk until the mice reached 10 wk of age. Results During the period of pain behavior, reduction of IL-10 protein and gene expression was detected in LDRG of db/db mice. Treatment with recombinant IL-10, from 8 to 10 wk of age, alleviates pain behaviors in db/db mice without affecting other diabetic parameters. In parallel, IL-10 treatment reduced the upregulation of nerve growth factor (NGF), inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF)-α, and high-affinity NGF receptor (Trk A) in LDRG, as well as the numbers of iNOS-positive Langerhans cells and CD-68-positive dermal dendritic cells in the hind-foot-pad skin in db/db mice. Conclusion Our findings suggest that the reduction in neuronal IL-10 increases inflammatory phenomena, ultimately contributing to PDN. These results suggest that the dysregulation of cytokine-mediated inflammation contributes to the development of PDN in db/db mice. Targeting this pathophysiologic mechanism could be an effective approach for treating PDN in type 2 diabetes.
Collapse
Affiliation(s)
- Brandon M Yanik
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jacqueline R Dauch
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Hsinlin T Cheng
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, Michigan, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Mukhopadhyay S, Heinz E, Porreca I, Alasoo K, Yeung A, Yang HT, Schwerd T, Forbester JL, Hale C, Agu CA, Choi YH, Rodrigues J, Capitani M, Jostins-Dean L, Thomas DC, Travis S, Gaffney D, Skarnes WC, Thomson N, Uhlig HH, Dougan G, Powrie F. Loss of IL-10 signaling in macrophages limits bacterial killing driven by prostaglandin E2. J Exp Med 2020; 217:132614. [PMID: 31819956 PMCID: PMC7041704 DOI: 10.1084/jem.20180649] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/09/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022] Open
Abstract
Cytokines and lipid mediators are key regulators of inflammation; but how they are mechanistically linked is poorly understood. Here, Mukhopadhyay et al. show a novel regulation between cytokine IL-10 and lipid mediator PGE2 that functionally connects them to intestinal inflammation. Loss of IL-10 signaling in macrophages (Mφs) leads to inflammatory bowel disease (IBD). Induced pluripotent stem cells (iPSCs) were generated from an infantile-onset IBD patient lacking a functional IL10RB gene. Mφs differentiated from IL-10RB−/− iPSCs lacked IL-10RB mRNA expression, were unable to phosphorylate STAT3, and failed to reduce LPS induced inflammatory cytokines in the presence of exogenous IL-10. IL-10RB−/− Mφs exhibited a striking defect in their ability to kill Salmonella enterica serovar Typhimurium, which was rescuable after experimentally introducing functional copies of the IL10RB gene. Genes involved in synthesis and receptor pathways for eicosanoid prostaglandin E2 (PGE2) were more highly induced in IL-10RB−/− Mφs, and these Mφs produced higher amounts of PGE2 after LPS stimulation compared with controls. Furthermore, pharmacological inhibition of PGE2 synthesis and PGE2 receptor blockade enhanced bacterial killing in Mφs. These results identify a regulatory interaction between IL-10 and PGE2, dysregulation of which may drive aberrant Mφ activation and impaired host defense contributing to IBD pathogenesis.
Collapse
Affiliation(s)
- Subhankar Mukhopadhyay
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Medical Research Council Centre for Transplantation, Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Eva Heinz
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | | | - Kaur Alasoo
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Amy Yeung
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Huei-Ting Yang
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Swiss Precision Dignostics Development Company Limited, Bedford, UK
| | - Tobias Schwerd
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jessica L Forbester
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | | | | | - Yoon Ha Choi
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | | | - Melania Capitani
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Luke Jostins-Dean
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - David C Thomas
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Simon Travis
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | - William C Skarnes
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Nicholas Thomson
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Fiona Powrie
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.,The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Kang MJ, Jang AR, Park JY, Ahn JH, Lee TS, Kim DY, Lee MS, Hwang S, Jeong YJ, Park JH. IL-10 Protects Mice From the Lung Infection of Acinetobacter baumannii and Contributes to Bacterial Clearance by Regulating STAT3-Mediated MARCO Expression in Macrophages. Front Immunol 2020; 11:270. [PMID: 32153580 PMCID: PMC7047127 DOI: 10.3389/fimmu.2020.00270] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/03/2020] [Indexed: 11/13/2022] Open
Abstract
Interleukin-10 plays important, yet contrasting, roles in host protection against bacterial infections and in the septic response. To determine the role of IL-10 in the host defense against Acinetobacter baumannii infection, wild-type (WT) and IL-10-deficient mice were infected intranasally with the bacteria. IL-10-deficient mice exhibited increased mortality, severe pathology, and excess production of proinflammatory cytokines and chemokines in the lungs, and increased bacterial burdens in bronchoalveolar lavage (BAL) fluids and lung homogenates after A. baumannii infection, compared to WT mice. Intranasal administration of recombinant IL-10 rescued mice from the lethality of the bacterial infection by promoting bacterial clearance and reducing production of cytokines and chemokines in the lungs. In vitro experiments revealed that IL-10 enhanced phagocytosis and bacterial killing by macrophages by upregulating the macrophage receptor with collagenous structure (MARCO). In addition, A. baumannii-induced activation of STAT3 was impaired in IL-10-deficient macrophages, which was essential for expression of MARCO. Intranasal adoptive transfer of WT macrophages resulted in significant increases in mice survival and bacterial clearance in IL-10-deficient mice infected with A. baumannii. Our results show that IL-10 played an important role in the host defense against pulmonary infection of A. baumannii by promoting the antibacterial function of macrophages by regulating MARCO expression through the STAT3-mediated pathway.
Collapse
Affiliation(s)
- Min-Jung Kang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Ji-Yeon Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Dong-Yeon Kim
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Moo-Seung Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Seungwoo Hwang
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yu-Jin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
10
|
Jafari M, Mohammadzadeh Jahani P, Choopanizadeh M, Jamalidoost M, Pourabbas B, Pouladfar G, Kalani M. Investigating the role of T helper related cytokines in cerebrospinal fluid for the differential diagnosis of bacterial meningitis in pre-treated paediatric patients. Biomarkers 2020; 25:171-178. [DOI: 10.1080/1354750x.2020.1714737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Maedeh Jafari
- Department of Pediatrics, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Maral Choopanizadeh
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Jamalidoost
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Pourabbas
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Pouladfar
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Kim BJ, Shusta EV, Doran KS. Past and Current Perspectives in Modeling Bacteria and Blood-Brain Barrier Interactions. Front Microbiol 2019; 10:1336. [PMID: 31263460 PMCID: PMC6585309 DOI: 10.3389/fmicb.2019.01336] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/29/2019] [Indexed: 01/18/2023] Open
Abstract
The central nervous system (CNS) barriers are highly specialized cellular barriers that promote brain homeostasis while restricting pathogen and toxin entry. The primary cellular constituent regulating pathogen entry in most of these brain barriers is the brain endothelial cell (BEC) that exhibits properties that allow for tight regulation of CNS entry. Bacterial meningoencephalitis is a serious infection of the CNS and occurs when bacteria can cross specialized brain barriers and cause inflammation. Models have been developed to understand the bacterial - BEC interaction that lead to pathogen crossing into the CNS, however, these have been met with challenges due to these highly specialized BEC phenotypes. This perspective provides a brief overview and outlook of the in vivo and in vitro models currently being used to study bacterial brain penetration, and opinion on improved models for the future.
Collapse
Affiliation(s)
- Brandon J Kim
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, United States
| | - Kelly S Doran
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
12
|
E. coli induced larger neutrophils in the peritoneal cavity of mice with severe septic peritonitis. Mol Immunol 2018; 105:86-95. [PMID: 30500625 DOI: 10.1016/j.molimm.2018.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 12/26/2022]
Abstract
Neutrophils, classified as professional phagocytes, are crucial in killing bacteria and preventing inflammation. When studying the roles of neutrophils in the development of the septic peritonitis induced by E. coli, we noticed some of the larger cells existed among peritoneal lavage fluid cells (PLCs). Besides the large size, their nuclei are segmented and flat, and squeezed to the marginal zone of the inner membrane. The cells, therefore, were designated as E. coli induced larger neutrophils (e-Neus). Further studies showed that, the e-Neus were ly6G positive, indicating the e-Neus were a type of neutrophils. The enlarged cell size and marginal nucleus of the e-Neus were caused by engulfing abundant of E. coli, marking the active participation of the e-Neus in clearance of E. coli. Functionally, the e-Neus generated reactive oxygen species (ROS) and IL-10. Furthermore, the occurrence and accumulation of the e-Neus were closely correlated with the severity of septic peritonitis and mortality of the mice. Overall, the e-Neus presented here may enrich the understandings on neutrophil transitions in response to various insults, and could be used to evaluate the severity of septic peritonitis induced by E. coli.
Collapse
|
13
|
Permanent neuroglial remodeling of the retina following infiltration of CSF1R inhibition-resistant peripheral monocytes. Proc Natl Acad Sci U S A 2018; 115:E11359-E11368. [PMID: 30442669 PMCID: PMC6275537 DOI: 10.1073/pnas.1807123115] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This work contributes to the understanding of the enigmatic progressive retinal damage following acute ocular surface injury. Clinical findings in patients suggest that such injuries can adversely affect the retina. This study demonstrates that corneal injury leads to rapid infiltration of blood-derived monocytes into the retina and to subsequent remodeling of the neuroglial system. In contrast to previously held belief, this study shows that the blood-derived monocytes engraft permanently into the retina and differentiate into microglia-like cells. Although these cells are morphologically indistinguishable from native microglia, they retain a distinct signature and insensitivity to CSF1R inhibition and exhibit a reactive phenotype which persists long after the noxious stimuli is removed, ultimately contributing to progressive neuroretinal degeneration. Previous studies have demonstrated that ocular injury can lead to prompt infiltration of bone-marrow–derived peripheral monocytes into the retina. However, the ability of these cells to integrate into the tissue and become microglia has not been investigated. Here we show that such peripheral monocytes that infiltrate into the retina after ocular injury engraft permanently, migrate to the three distinct microglia strata, and adopt a microglia-like morphology. In the absence of ocular injury, peripheral monocytes that repopulate the retina after depletion with colony-stimulating factor 1 receptor (CSF1R) inhibitor remain sensitive to CSF1R inhibition and can be redepleted. Strikingly, consequent to ocular injury, the engrafted peripheral monocytes are resistant to depletion by CSF1R inhibitor and likely express low CSF1R. Moreover, these engrafted monocytes remain proinflammatory, expressing high levels of MHC-II, IL-1β, and TNF-α over the long term. The observed permanent neuroglia remodeling after injury constitutes a major immunological change that may contribute to progressive retinal degeneration. These findings may also be relevant to other degenerative conditions of the retina and the central nervous system.
Collapse
|
14
|
Rogers LM, Anders AP, Doster RS, Gill EA, Gnecco JS, Holley JM, Randis TM, Ratner AJ, Gaddy JA, Osteen K, Aronoff DM. Decidual stromal cell-derived PGE 2 regulates macrophage responses to microbial threat. Am J Reprod Immunol 2018; 80:e13032. [PMID: 30084522 DOI: 10.1111/aji.13032] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022] Open
Abstract
PROBLEM Bacterial chorioamnionitis causes adverse pregnancy outcomes, yet host-microbial interactions are not well characterized within gestational membranes. The decidua, the outermost region of the membranes, is a potential point of entry for bacteria ascending from the vagina to cause chorioamnionitis. We sought to determine whether paracrine communication between decidual stromal cells and macrophages shaped immune responses to microbial sensing. METHOD OF STUDY Decidual cell-macrophage interactions were modeled in vitro utilizing decidualized, telomerase-immortalized human endometrial stromal cells (dTHESCs) and phorbol ester-differentiated THP-1 macrophage-like cells. The production of inflammatory mediators in response to LPS was monitored by ELISA for both cell types, while phagocytosis of bacterial pathogens (Escherichia coli and Group B Streptococcus (GBS)) was measured in THP-1 cells or primary human placental macrophages. Diclofenac, a non-selective cyclooxygenase inhibitor, and prostaglandin E2 (PGE2 ) were utilized to interrogate prostaglandins as decidual cell-derived paracrine immunomodulators. A mouse model of ascending chorioamnionitis caused by GBS was utilized to assess the colocalization of bacteria and macrophages in vivo and assess PGE2 production. RESULTS In response to LPS, dTHESC and THP-1 coculture demonstrated enhancement of most inflammatory mediators, but a potent suppression of macrophage TNF-α generation was observed. This appeared to reflect a paracrine-mediated effect of decidual cell-derived PGE2 . In mice with GBS chorioamnionitis, macrophages accumulated at sites of bacterial invasion with increased PGE2 in amniotic fluid, suggesting such paracrine effects might hold relevance in vivo. CONCLUSION These data suggest key roles for decidual stromal cells in modulating tissue responses to microbial threat through release of PGE2 .
Collapse
Affiliation(s)
- Lisa M Rogers
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anjali P Anders
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Ryan S Doster
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Juan S Gnecco
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jacob M Holley
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tara M Randis
- Department of Pediatrics, New York University School of Medicine, New York, New York.,Department of Microbiology, New York University School of Medicine, New York, New York
| | - Adam J Ratner
- Department of Pediatrics, New York University School of Medicine, New York, New York.,Department of Microbiology, New York University School of Medicine, New York, New York
| | - Jennifer A Gaddy
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Veteran Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - Kevin Osteen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Veteran Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - David M Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
15
|
Zhao WD, Liu DX, Wei JY, Miao ZW, Zhang K, Su ZK, Zhang XW, Li Q, Fang WG, Qin XX, Shang DS, Li B, Li QC, Cao L, Kim KS, Chen YH. Caspr1 is a host receptor for meningitis-causing Escherichia coli. Nat Commun 2018; 9:2296. [PMID: 29895952 PMCID: PMC5997682 DOI: 10.1038/s41467-018-04637-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 05/03/2018] [Indexed: 12/17/2022] Open
Abstract
Escherichia coli is the leading cause of neonatal Gram-negative bacterial meningitis, but the pathogenesis of E. coli meningitis remains elusive. E. coli penetration of the blood–brain barrier (BBB) is the critical step for development of meningitis. Here, we identify Caspr1, a single-pass transmembrane protein, as a host receptor for E. coli virulence factor IbeA to facilitate BBB penetration. Genetic ablation of endothelial Caspr1 and blocking IbeA–Caspr1 interaction effectively prevent E. coli penetration into the brain during meningitis in rodents. IbeA interacts with extracellular domain of Caspr1 to activate focal adhesion kinase signaling causing E. coli internalization into the brain endothelial cells of BBB. E. coli can invade hippocampal neurons causing apoptosis dependent on IbeA–Caspr1 interaction. Our results indicate that E. coli exploits Caspr1 as a host receptor for penetration of BBB resulting in meningitis, and that Caspr1 might be a useful target for prevention or therapy of E. coli meningitis. Penetration of the blood–brain barrier (BBB) is crucial for development of E. coli-caused meningitis. Here, the authors show that a host membrane protein, Caspr1, acts as a receptor for a bacterial virulence factor to facilitate BBB penetration and entry of E. coli into brain neurons.
Collapse
Affiliation(s)
- Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China.
| | - Dong-Xin Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Jia-Yi Wei
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Zi-Wei Miao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Zheng-Kang Su
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Qiang Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Wen-Gang Fang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Xiao-Xue Qin
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - De-Shu Shang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Bo Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Qing-Chang Li
- Department of Pathology, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Liu Cao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe St, Room 3157, Baltimore, MD, 21287, USA
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China.
| |
Collapse
|
16
|
Schlieckau F, Schulz D, Fill Malfertheiner S, Entleutner K, Seelbach-Goebel B, Ernst W. A novel model to study neonatal Escherichia coli sepsis and the effect of treatment on the human immune system using humanized mice. Am J Reprod Immunol 2018; 80:e12859. [PMID: 29672989 DOI: 10.1111/aji.12859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/27/2018] [Indexed: 01/03/2023] Open
Abstract
PROBLEM Neonatal sepsis is a serious threat especially for preterm infants. As existing in vitro and in vivo models have limitations, we generated a novel neonatal sepsis model using humanized mice and tested the effect of Betamethasone and Indomethacin which are used in the clinic in case of premature birth. METHOD OF STUDY Humanized mice were infected with Escherichia coli (E. coli). Subsequently, the effect of the infection itself, and treatment with Betamethasone and Indomethacin on survival, recovery, bacterial burden, leukocyte populations, and cytokine production, was analyzed. RESULTS The human immune system in the animals responded with leukocyte trafficking to the site of infection and granulopoiesis in the bone marrow. Treatment with Indomethacin had no pronounced effect on the immune system or bacterial burden. Betamethasone induced a decline of splenocytes. CONCLUSION The human immune system in humanized mice responds to the infection, making them a suitable model to study neonatal E. coli sepsis and the immune response of the neonatal immune system. Treatment with Betamethasone could have potential negative long-term effects for the immune system of the child.
Collapse
Affiliation(s)
- Florian Schlieckau
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Daniela Schulz
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Sara Fill Malfertheiner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Kathrin Entleutner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Birgit Seelbach-Goebel
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Wolfgang Ernst
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Paschalis EI, Lei F, Zhou C, Kapoulea V, Thanos A, Dana R, Vavvas DG, Chodosh J, Dohlman CH. The Role of Microglia and Peripheral Monocytes in Retinal Damage after Corneal Chemical Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1580-1596. [PMID: 29630857 DOI: 10.1016/j.ajpath.2018.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/04/2018] [Accepted: 03/26/2018] [Indexed: 12/17/2022]
Abstract
Eyes that have experienced alkali burn to the surface are excessively susceptible to subsequent severe glaucoma and retinal ganglion cell loss, despite maximal efforts to prevent or slow down the disease. Recently, we have shown, in mice and rabbits, that such retinal damage is neither mediated by the alkali itself reaching the retina nor by intraocular pressure elevation. Rather, it is caused by the up-regulation of tumor necrosis factor-α (TNF-α), which rapidly diffuses posteriorly, causing retinal ganglion cell apoptosis and CD45+ cell activation. Herein, we investigated the involvement of peripheral blood monocytes and microglia in retinal damage. Using CX3CR1+/EGFP::CCR2+/RFP reporter mice and bone marrow chimeras, we show that peripheral CX3CR1+CD45hiCD11b+MHC-II+ monocytes infiltrate into the retina from the optic nerve at 24 hours after the burn and release further TNF-α. A secondary source of peripheral monocyte response originates from a rare population of patrolling myeloid CCR2+ cells of the retina that differentiate into CX3CR1+ macrophages within hours after the injury. As a result, CX3CR1+CD45loCD11b+ microglia become reactive at 7 days, causing further TNF-α release. Prompt TNF-α inhibition after corneal burn suppresses monocyte infiltration and microglia activation, and protects the retina. This study may prove relevant to other injuries of the central nervous system.
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Chengxin Zhou
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Vassiliki Kapoulea
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Aristomenis Thanos
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Demetrios G Vavvas
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Claes H Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
19
|
Breland EJ, Eberly AR, Hadjifrangiskou M. An Overview of Two-Component Signal Transduction Systems Implicated in Extra-Intestinal Pathogenic E. coli Infections. Front Cell Infect Microbiol 2017; 7:162. [PMID: 28536675 PMCID: PMC5422438 DOI: 10.3389/fcimb.2017.00162] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022] Open
Abstract
Extra-intestinal pathogenic E. coli (ExPEC) infections are common in mammals and birds. The predominant ExPEC types are avian pathogenic E. coli (APEC), neonatal meningitis causing E. coli/meningitis associated E. coli (NMEC/MAEC), and uropathogenic E. coli (UPEC). Many reviews have described current knowledge on ExPEC infection strategies and virulence factors, especially for UPEC. However, surprisingly little has been reported on the regulatory modules that have been identified as critical in ExPEC pathogenesis. Two-component systems (TCSs) comprise the predominant method by which bacteria respond to changing environments and play significant roles in modulating bacterial fitness in diverse niches. Recent studies have highlighted the potential of manipulating signal transduction systems as a means to chemically re-wire bacterial pathogens, thereby reducing selective pressure and avoiding the emergence of antibiotic resistance. This review begins by providing a brief introduction to characterized infection strategies and common virulence factors among APEC, NMEC, and UPEC and continues with a comprehensive overview of two-component signal transduction networks that have been shown to influence ExPEC pathogenesis.
Collapse
Affiliation(s)
- Erin J Breland
- Department of Pharmacology, Vanderbilt University Medical CenterNashville, TN, USA
| | - Allison R Eberly
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashville, TN, USA
| | - Maria Hadjifrangiskou
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashville, TN, USA.,Department of Urology, Vanderbilt University Medical CenterNashville, TN, USA
| |
Collapse
|
20
|
Zhao M, Zhang L, Lv S, Zhang C, Wang L, Chen H, Zhou Y, Lou J. IQGAP1 Mediates Hcp1-Promoted Escherichia coli Meningitis by Stimulating the MAPK Pathway. Front Cell Infect Microbiol 2017; 7:132. [PMID: 28469997 PMCID: PMC5395654 DOI: 10.3389/fcimb.2017.00132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/31/2017] [Indexed: 01/14/2023] Open
Abstract
Escherichia coli-induced meningitis remains a life-threatening disease despite recent advances in the field of antibiotics-based therapeutics, necessitating continued research on its pathogenesis. The current study aims to elucidate the mechanism through which hemolysin-coregulated protein 1 (Hcp1) induces the apoptosis of human brain microvascular endothelial cells (HBMEC). Co-immunoprecipitation coupled with mass spectrometric (MS) characterization led to the identification of IQ motif containing GTPase activating protein 1 (IQGAP1) as a downstream target of Hcp1. IQGAP1 was found to be up-regulated by Hcp1 treatment and mediate the stimulation of HBMEC apoptosis. It was shown that Hcp1 could compete against Smurf1 for binding to IQGAP1, thereby rescuing the latter from ubiquitin-dependent degradation. Subsequent study suggested that IQGAP1 could stimulate the MAPK signaling pathway by promoting the phosphorylation of ERK1/2, an effect that was blocked by U0126, an MAPK inhibitor. Furthermore, U0126 also demonstrated therapeutic potential against E. coli meningitis in a mouse model. Taken together, our results suggested the feasibility of targeting the MAPK pathway as a putative therapeutic strategy against bacterial meningitis.
Collapse
Affiliation(s)
- Mingna Zhao
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Lingfei Zhang
- Center for RNA Research, State Key Laboratory of Molecular Biology-University of Chinese Academy of Sciences, Institute of Biochemistry and Cell Biology Shanghai Institutes for Biological Sciences Chinese Academy of SciencesShanghai, China.,Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Shaogang Lv
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Chenzi Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Lin Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Hong Chen
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Yan Zhou
- Department of Microbiology and Immunobiology, Harvard Medical SchoolBoston, MA, USA
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
21
|
Peñaloza HF, Schultz BM, Nieto PA, Salazar GA, Suazo I, Gonzalez PA, Riedel CA, Alvarez-Lobos MM, Kalergis AM, Bueno SM. Opposing roles of IL-10 in acute bacterial infection. Cytokine Growth Factor Rev 2016; 32:17-30. [PMID: 27522641 DOI: 10.1016/j.cytogfr.2016.07.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Interleukin-10 (IL-10) is recognized as an anti-inflammatory cytokine that downmodulates inflammatory immune responses at multiple levels. In innate cells, production of this cytokine is usually triggered after pathogen recognition receptor (PRR) engagement by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patters (DAMPs), as well as by other soluble factors. Importantly, IL-10 is frequently secreted during acute bacterial infections and has been described to play a key role in infection resolution, although its effects can significantly vary depending on the infecting bacterium. While the production of IL-10 might favor host survival in some cases, it may also result harmful for the host in other circumstances, as it can prevent appropriate bacterial clearance. In this review we discuss the role of IL-10 in bacterial clearance and propose that this cytokine is required to recover from infection caused by extracellular or highly pro-inflammatory bacteria. Altogether, we propose that IL-10 drives excessive suppression of the immune response upon infection with intracellular bacteria or in non-inflammatory bacterial infections, which ultimately favors bacterial persistence and dissemination within the host. Thus, the nature of the bacterium causing infection is an important factor that needs to be taken into account when considering new immunotherapies that consist on the modulation of inflammation, such as IL-10. Indeed, induction of this cytokine may significantly improve the host's immune response to certain bacteria when antibiotics are not completely effective.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Barbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Geraldyne A Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Isidora Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pablo A Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Chile
| | - Manuel M Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
22
|
Krishnan S, Chang AC, Stoltz BM, Prasadarao NV. Escherichia coli K1 Modulates Peroxisome Proliferator-Activated Receptor γ and Glucose Transporter 1 at the Blood-Brain Barrier in Neonatal Meningitis. J Infect Dis 2016; 214:1092-104. [PMID: 27456707 DOI: 10.1093/infdis/jiw306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/13/2016] [Indexed: 12/14/2022] Open
Abstract
Escherichia coli K1 meningitis continues to be a major threat to neonatal health. Previous studies demonstrated that outer membrane protein A (OmpA) of E. coli K1 interacts with endothelial cell glycoprotein 96 (Ecgp96) in the blood-brain barrier to enter the central nervous system. Here we show that the interaction between OmpA and Ecgp96 downregulates peroxisome proliferator-activated receptor γ (PPAR-γ) and glucose transporter 1 (GLUT-1) levels in human brain microvascular endothelial cells, causing disruption of barrier integrity and inhibition of glucose uptake. The suppression of PPAR-γ and GLUT-1 by the bacteria in the brain microvessels of newborn mice causes extensive pathophysiology owing to interleukin 6 production. Pretreatment with partial or selective PPAR-γ agonists ameliorate the pathological outcomes of infection by suppressing interleukin 6 production in the brain. Thus, inhibition of PPAR-γ and GLUT-1 by E. coli K1 is a novel pathogenic mechanism in meningitis, and pharmacological upregulation of PPAR-γ and GLUT-1 levels may provide novel therapeutic avenues.
Collapse
Affiliation(s)
- Subramanian Krishnan
- Division of Infectious Diseases, Department of Pediatrics Department of Surgery, Children's Hospital Los Angeles
| | - Alexander C Chang
- Division of Infectious Diseases, Department of Pediatrics Department of Surgery, Children's Hospital Los Angeles
| | - Brian M Stoltz
- Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena
| | - Nemani V Prasadarao
- Division of Infectious Diseases, Department of Pediatrics Department of Surgery, Children's Hospital Los Angeles Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
23
|
Doran KS, Fulde M, Gratz N, Kim BJ, Nau R, Prasadarao N, Schubert-Unkmeir A, Tuomanen EI, Valentin-Weigand P. Host-pathogen interactions in bacterial meningitis. Acta Neuropathol 2016; 131:185-209. [PMID: 26744349 PMCID: PMC4713723 DOI: 10.1007/s00401-015-1531-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/26/2022]
Abstract
Bacterial meningitis is a devastating disease occurring worldwide with up to half of the survivors left with permanent neurological sequelae. Due to intrinsic properties of the meningeal pathogens and the host responses they induce, infection can cause relatively specific lesions and clinical syndromes that result from interference with the function of the affected nervous system tissue. Pathogenesis is based on complex host–pathogen interactions, some of which are specific for certain bacteria, whereas others are shared among different pathogens. In this review, we summarize the recent progress made in understanding the molecular and cellular events involved in these interactions. We focus on selected major pathogens, Streptococcus pneumonia, S. agalactiae (Group B Streptococcus), Neisseria meningitidis, and Escherichia coli K1, and also include a neglected zoonotic pathogen, Streptococcus suis. These neuroinvasive pathogens represent common themes of host–pathogen interactions, such as colonization and invasion of mucosal barriers, survival in the blood stream, entry into the central nervous system by translocation of the blood–brain and blood–cerebrospinal fluid barrier, and induction of meningeal inflammation, affecting pia mater, the arachnoid and subarachnoid spaces.
Collapse
|
24
|
Kimmel DW, Rogers LM, Aronoff DM, Cliffel DE. Prostaglandin E2 Regulation of Macrophage Innate Immunity. Chem Res Toxicol 2015; 29:19-25. [PMID: 26656203 DOI: 10.1021/acs.chemrestox.5b00322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Globally, maternal and fetal health is greatly impacted by extraplacental inflammation. Group B Streptococcus (GBS), a leading cause of chorioamnionitis, is thought to take advantage of the uterine environment during pregnancy in order to cause inflammation and infection. In this study, we demonstrate the metabolic changes of murine macrophages caused by GBS exposure. GBS alone prompted a delayed increase in lactate production, highlighting its ability to redirect macrophage metabolism from aerobic to anaerobic respiration. This production of lactate is thought to aid in the development and propagation of GBS throughout the surrounding tissue. Additionally, this study shows that PGE2 priming was able to exacerbate lactate production, shown by the rapid and substantial lactate increases seen upon GBS exposure. These data provide a novel model to study the role of GBS exposure to macrophages with and without PGE2 priming.
Collapse
Affiliation(s)
| | - Lisa M Rogers
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - David M Aronoff
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University , Nashville, Tennessee 37232, United States
| | | |
Collapse
|
25
|
Abstract
Neonatal sepsis is one of the most fulminating conditions in neonatal intensive care units. Antipathogen and supportive care are administered routinely, but do not deliver satisfactory results. In addition, the efforts to treat neonatal sepsis with anti-inflammatory agents have generally shown to be futile. The accumulating data imply that intracellular redox changes intertwined into neonatal sepsis redox cycle represent the main cause of dysfunction of mitochondria and cells in neonatal sepsis. Our aim here is to support the new philosophy in neonatal sepsis treatment, which involves the integration of mechanisms that are responsible for cellular dysfunction and organ failure, the recognition of the most important targets, and the selection of safe agents that can stop the neonatal sepsis redox cycle by hitting the hot spots. Redox-active agents that could be beneficial for neonatal sepsis treatment according to these criteria include lactoferrin, interleukin 10, zinc and selenium supplements, ibuprofen, edaravone, and pentoxifylline.
Collapse
|
26
|
Krishnan S, Chang AC, Hodges J, Couraud PO, Romero IA, Weksler B, Nicholson BA, Nolan LK, Prasadarao NV. Serotype O18 avian pathogenic and neonatal meningitis Escherichia coli strains employ similar pathogenic strategies for the onset of meningitis. Virulence 2015; 6:777-86. [PMID: 26407066 DOI: 10.1080/21505594.2015.1091914] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Neonatal meningitis Escherichia coli K1 (NMEC) are thought to be transmitted from mothers to newborns during delivery or by nosocomial infections. However, the source of E. coli K1 causing these infections is not clear. Avian pathogenic E. coli (APEC) have the potential to cause infection in humans while human E. coli have potential to cause colibacillosis in poultry, suggesting that these strains may lack host specificity. APEC strains are capable of causing meningitis in newborn rats; however, it is unclear whether these bacteria use similar mechanisms to that of NMEC to establish disease. Using four representative APEC and NMEC strains that belong to serotype O18, we demonstrate that these strains survive in human serum similar to that of the prototypic NMEC strain E44, a derivative of RS218. These bacteria also bind and enter both macrophages and human cerebral microvascular endothelial cells (HCMEC/D3) with similar frequency as that of E44. The amino acid sequences of the outer membrane protein A (OmpA), an important virulence factor in the pathogenesis of meningitis, are identical within these representative APEC and NMEC strains. Further, these strains also require FcγRI-α chain (CD64) and Ecgp96 as receptors for OmpA in macrophages and HCMEC/D3, respectively, to bind and enter these cells. APEC and NMEC strains induce meningitis in newborn mice with varying degree of pathology in the brains as assessed by neutrophil recruitment and neuronal apoptosis. Together, these results suggest that serotype O18 APEC strains utilize similar pathogenic mechanisms as those of NMEC strains in causing meningitis.
Collapse
Affiliation(s)
- Subramanian Krishnan
- a Division of Infectious Diseases and Department of Pediatrics; Children's Hospital Los Angeles , CA USA
| | - Alexander C Chang
- a Division of Infectious Diseases and Department of Pediatrics; Children's Hospital Los Angeles , CA USA
| | - Jacqueline Hodges
- a Division of Infectious Diseases and Department of Pediatrics; Children's Hospital Los Angeles , CA USA
| | - Pierre-Olivier Couraud
- b Inserm; Institut Cochin, Paris, France; Université Paris Descartes; Sorbonne Paris Cité , Paris , France
| | - Ignacio A Romero
- c Department of Life ; Health and Chemical Sciences; Open University ; Milton Keynes , UK
| | - Babette Weksler
- d Division of Hematology and Medical Oncology; Weill Cornell Medical College ; New York , NY USA
| | - Bryon A Nicholson
- e Department of Veterinary Microbiology and Preventive Medicine ; College of Veterinary Medicine; Iowa State University ; Ames , IA USA
| | - Lisa K Nolan
- e Department of Veterinary Microbiology and Preventive Medicine ; College of Veterinary Medicine; Iowa State University ; Ames , IA USA
| | - Nemani V Prasadarao
- a Division of Infectious Diseases and Department of Pediatrics; Children's Hospital Los Angeles , CA USA.,f Department of Surgery ; Children's Hospital Los Angeles; University of Southern California ; Los Angeles , CA USA
| |
Collapse
|
27
|
Jin C, Londono I, Mallard C, Lodygensky GA. New means to assess neonatal inflammatory brain injury. J Neuroinflammation 2015; 12:180. [PMID: 26407958 PMCID: PMC4583178 DOI: 10.1186/s12974-015-0397-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/10/2015] [Indexed: 01/23/2023] Open
Abstract
Preterm infants are especially vulnerable to infection-induced white matter injury, associated with cerebral palsy, cognitive and psychomotor impairment, and other adverse neurological outcomes. The etiology of such lesions is complex and multifactorial. Furthermore, timing and length of exposure to infection also influence neurodevelopmental outcomes. Different mechanisms have been posited to mediate the observed brain injury including microglial activation followed by subsequent release of pro-inflammatory species, glutamate-induced excitotoxicity, and vulnerability of developing oligodendrocytes to cerebral insults. The prevalence of such neurological impairments requires an urgent need for early detection and effective neuroprotective strategies. Accordingly, noninvasive methods of monitoring disease progression and therapy effectiveness are essential. While diagnostic tools using biomarkers from bodily fluids may provide useful information regarding potential risks of developing neurological diseases, the use of magnetic resonance imaging/spectroscopy has emerged as a promising candidate for such purpose. Various pharmacological agents have demonstrated protective effects in the immature brain in animal models; however, few studies have progressed to clinical trials with promising results.
Collapse
Affiliation(s)
- Chen Jin
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Irene Londono
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Carina Mallard
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden.
| | - Gregory A Lodygensky
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada. .,Montreal Heart Institute, 5000 Rue Bélanger, Montréal, Québec, Canada. .,Department of Neuroscience and Pharmacology, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
28
|
Impaired hydrogen sulfide synthesis and IL-10 signaling underlie hyperhomocysteinemia-associated exacerbation of colitis. Proc Natl Acad Sci U S A 2014; 111:13559-64. [PMID: 25187563 DOI: 10.1073/pnas.1413390111] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Vitamin B deficiencies, which can lead to hyperhomocysteinemia (Hhcy), are commonly reported in patients with inflammatory bowel disease (IBD) and may be a causative underlying factor. However, the mechanism for this effect is not known. Hydrogen sulfide (H2S) is a gaseous mediator that promotes tissue repair and resolution of inflammation. In experimental colitis, a marked increase in colonic H2S synthesis drives ulcer healing and resolution of inflammation. Because H2S synthesis is in part dependent upon enzymes that require vitamin B6 as a cofactor, we tested the hypothesis that Hhcy in rodent models would increase the susceptibility to colitis. In all three models tested, diet-induced Hhcy significantly exacerbated colitis. The usual elevation of colonic H2S synthesis after induction of colitis was absent in all three models of colitis. Administration of an H2S donor to Hhcy rats significantly decreased the severity of colitis. Compared with wild-type mice, interleukin (IL) 10-deficient mice on a normal diet had decreased levels of colonic H2S synthesis, a 40% increase in serum homocysteine, and a phenotype similar to wild-type mice with Hhcy. IL-10-deficient mice fed the vitamin B-deficient diet exhibited more severe colonic inflammation, but the normal elevation of colonic H2S synthesis was absent. Administration of IL-10 to the IL-10-deficient mice restored colonic H2S synthesis and significantly decreased serum homocysteine levels. These results suggest that the exacerbation of colitis in Hhcy is due in part to impaired colonic H2S synthesis. Moreover, IL-10 plays a novel role in promoting H2S production and homocysteine metabolism, which may have therapeutic value in conditions characterized by Hhcy.
Collapse
|
29
|
Mediators of monocyte migration in response to recovery modalities following resistance exercise. Mediators Inflamm 2014; 2014:145817. [PMID: 24987193 PMCID: PMC4060064 DOI: 10.1155/2014/145817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/14/2014] [Accepted: 05/05/2014] [Indexed: 11/14/2022] Open
Abstract
Mediators of monocyte migration, complement receptor-3 (CR3), and chemokine ligand-4 (CCL4) were measured in response to recovery modalities following resistance exercise. Thirty resistance-trained men (23.1 ± 2.9 y; 175.2 ± 7.1 cm; 82.1 ± 8.4 kg) were given neuromuscular electric stimulation (NMES), cold water immersion (CWI), or control (CON) treatments immediately following resistance exercise. Blood samples were obtained preexercise (PRE), immediately (IP), 30 minutes (30 P), 24 hours (24 H), and 48 hours (48 H) after exercise for measurement of circulating CCL4 and CR3 expression on CD14+ monocytes, by assay and flow cytometry. Circulating CCL4 showed no consistent changes. Inferential analysis indicated that CR3 expression was likely greater in CON at 30 P than NMES (90.0%) or CWI (86.8%). NMES was likely lower than CON at 24 H (92.9%) and very likely lower at 48 H (98.7%). Expression of CR3 following CWI was very likely greater than CON (96.5%) at 24 H. The proportion of CR3+ monocytes was likely greater following CWI than NMES (85.8%) or CON (85.2%) at 24 H. The change in proportion of CR3+ monocytes was likely (86.4%) greater following NMES than CON from IP to 30 P. The increased expression of CR3 and increased proportion of CR3+ monocytes following CWI at 24 H indicate a potentially improved ability for monocyte adhesion to the endothelium, possibly improving phagocytosis of damaged tissues.
Collapse
|
30
|
Zigmond E, Bernshtein B, Friedlander G, Walker CR, Yona S, Kim KW, Brenner O, Krauthgamer R, Varol C, Müller W, Jung S. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 2014; 40:720-33. [PMID: 24792913 DOI: 10.1016/j.immuni.2014.03.012] [Citation(s) in RCA: 441] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/18/2014] [Indexed: 02/07/2023]
Abstract
Interleukin-10 (IL-10) is a pleiotropic anti-inflammatory cytokine produced and sensed by most hematopoietic cells. Genome-wide association studies and experimental animal models point at a central role of the IL-10 axis in inflammatory bowel diseases. Here we investigated the importance of intestinal macrophage production of IL-10 and their IL-10 exposure, as well as the existence of an IL-10-based autocrine regulatory loop in the gut. Specifically, we generated mice harboring IL-10 or IL-10 receptor (IL-10Rα) mutations in intestinal lamina propria-resident chemokine receptor CX3CR1-expressing macrophages. We found macrophage-derived IL-10 dispensable for gut homeostasis and maintenance of colonic T regulatory cells. In contrast, loss of IL-10 receptor expression impaired the critical conditioning of these monocyte-derived macrophages and resulted in spontaneous development of severe colitis. Collectively, our results highlight IL-10 as a critical homeostatic macrophage-conditioning agent in the colon and define intestinal CX3CR1(hi) macrophages as a decisive factor that determines gut health or inflammation.
Collapse
Affiliation(s)
- Ehud Zigmond
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel; The Research Center for Digestive Tract and Liver Diseases, Tel Aviv-Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Biana Bernshtein
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gilgi Friedlander
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Catherine R Walker
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Simon Yona
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ki-Wook Kim
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ori Brenner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rita Krauthgamer
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv-Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
31
|
Krishnan S, Shanmuganathan MV, Behenna D, Stoltz BM, Prasadarao NV. Angiotensin II receptor type 1--a novel target for preventing neonatal meningitis in mice by Escherichia coli K1. J Infect Dis 2013; 209:409-19. [PMID: 24041786 DOI: 10.1093/infdis/jit499] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The increasing incidence of Escherichia coli K1 meningitis due to escalating antibiotic resistance warrants alternate treatment options to prevent this deadly disease. We screened a library of small molecules from the National Institutes of Health clinical collection and identified telmisartan, an angiotensin II receptor type 1 (AT1R) blocker, as a potent inhibitor of E. coli invasion into human brain microvascular endothelial cells (HBMECs). Immunoprecipitation studies revealed that AT1R associates with endothelial cell gp96, the receptor in HBMECs for E. coli outer membrane protein A. HBMECs pretreated with telmisartan or transfected with AT1R small interfering RNA were resistant to E. coli invasion because of downregulation of protein kinase C-α phosphorylation. Administration of a soluble derivative of telmisartan to newborn mice before infection with E. coli prevented the onset of meningitis and suppressed neutrophil infiltration and glial cell migration in the brain. Therefore, telmisartan has potential as an alternate treatment option for preventing E. coli meningitis.
Collapse
|
32
|
Humanized mice, a new model to study the influence of drug treatment on neonatal sepsis. Infect Immun 2013; 81:1520-31. [PMID: 23439310 DOI: 10.1128/iai.01235-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Bacterial infection with group B Streptococcus (GBS) represents a prominent threat to neonates and fetuses in the Western world, causing severe organ damage and even death. To improve current therapeutic strategies and to investigate new approaches, an appropriate in vivo model to study the immune response of a human immune system is needed. Therefore, we introduced humanized mice as a new model for GBS-induced sepsis. Humanized mice feature deficiencies similar to those found in neonates, such as lower immunoglobulin levels and myeloid cell dysfunction. Due to the husbandry in specific-pathogen-free (SPF) facilities, the human immune cells in these mice also exhibit a naive phenotype which mimics the conditions in fetuses/neonates. Following infection, cytokine release and leukocyte trafficking from the bone marrow to the lymphoid organ (spleen) and into the peritoneum (site of infection) as well as bacterial spreading and clearance were traceable in the humanized mice. Furthermore, we investigated the effects of betamethasone and indomethacin treatment using this novel sepsis model. Although both drugs are commonly used in perinatal care, little is known about their effects on the neonatal immune system. Treatment of infected humanized mice not only induced the reduction of human leukocytes in the spleen but also increased the bacterial load in all analyzed organs, including the brain, which did not show infiltration of live GBS in untreated controls. These studies demonstrate the utility of the humanized mice as a new model to study an immature human immune response during bacterial infection and allow the investigation of side effects induced by various treatments.
Collapse
|
33
|
Prostaglandin E2 and the suppression of phagocyte innate immune responses in different organs. Mediators Inflamm 2012; 2012:327568. [PMID: 23024463 PMCID: PMC3449139 DOI: 10.1155/2012/327568] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/19/2012] [Accepted: 05/03/2012] [Indexed: 12/15/2022] Open
Abstract
The local and systemic production of prostaglandin E2 (PGE2) and its actions in phagocytes lead to immunosuppressive conditions. PGE2 is produced at high levels during inflammation, and its suppressive effects are caused by the ligation of the E prostanoid receptors EP2 and EP4, which results in the production of cyclic AMP. However, PGE2 also exhibits immunostimulatory properties due to binding to EP3, which results in decreased cAMP levels. The various guanine nucleotide-binding proteins (G proteins) that are coupled to the different EP receptors account for the pleiotropic roles of PGE2 in different disease states. Here, we discuss the production of PGE2 and the actions of this prostanoid in phagocytes from different tissues, the relative contribution of PGE2 to the modulation of innate immune responses, and the novel therapeutic opportunities that can be used to control inflammatory responses.
Collapse
|
34
|
|
35
|
Le HT, Tran VG, Kim W, Kim J, Cho HR, Kwon B. IL-33 Priming Regulates Multiple Steps of the Neutrophil-Mediated Anti-Candida albicansResponse by Modulating TLR and Dectin-1 Signals. THE JOURNAL OF IMMUNOLOGY 2012; 189:287-95. [DOI: 10.4049/jimmunol.1103564] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
36
|
Aronoff DM, Bergin IL, Lewis C, Goel D, O'Brien E, Peters-Golden M, Mancuso P. E-prostanoid 2 receptor signaling suppresses lung innate immunity against Streptococcus pneumoniae. Prostaglandins Other Lipid Mediat 2012; 98:23-30. [PMID: 22575745 DOI: 10.1016/j.prostaglandins.2012.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/22/2012] [Accepted: 03/23/2012] [Indexed: 10/28/2022]
Abstract
Pneumonia is a major global health problem. Prostaglandin (PG) E(2) is an immunomodulatory lipid with anti-inflammatory, immunosuppressive, and pro-resolving actions. Data suggest that the E-prostanoid (EP) 2 receptor mediates immunomodulatory effects of PGE(2), but the extent to which this occurs in Streptococcus pneumoniae infection is unknown. Intratracheal lung infection of C57BL/6 mice possessing (EP2(+/+)) or lacking (EP2(-/-)) the EP2 receptor was performed, as were in vitro studies of alveolar macrophage (AM) host defense functions. Bacterial clearance and survival were significantly improved in vivo in EP2(-/-) mice and it correlated with greater neutrophilic inflammation and higher lung IL-12 levels. Upon ex vivo challenge with pneumococcus, EP2(-/-)cells expressed greater amounts of TNF-α and MIP-2 than did EP2(+/+) AMs, and had improved phagocytosis, intracellular killing, and reactive oxygen intermediate generation. These data suggest that PGE(2)-EP2 signaling may provide a novel pharmacological target for treating pneumococcal pneumonia in combination with antimicrobials.
Collapse
Affiliation(s)
- David M Aronoff
- Divisions of Infectious Diseases, The University of Michigan, Ann Arbor, MI 48109-5680, United States.
| | | | | | | | | | | | | |
Collapse
|
37
|
Kalinski P. Regulation of immune responses by prostaglandin E2. THE JOURNAL OF IMMUNOLOGY 2012; 188:21-8. [PMID: 22187483 DOI: 10.4049/jimmunol.1101029] [Citation(s) in RCA: 1321] [Impact Index Per Article: 101.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PGE(2), an essential homeostatic factor, is also a key mediator of immunopathology in chronic infections and cancer. The impact of PGE(2) reflects the balance between its cyclooxygenase 2-regulated synthesis and 15-hydroxyprostaglandin dehydrogenase-driven degradation and the pattern of expression of PGE(2) receptors. PGE(2) enhances its own production but suppresses acute inflammatory mediators, resulting in its predominance at late/chronic stages of immunity. PGE(2) supports activation of dendritic cells but suppresses their ability to attract naive, memory, and effector T cells. PGE(2) selectively suppresses effector functions of macrophages and neutrophils and the Th1-, CTL-, and NK cell-mediated type 1 immunity, but it promotes Th2, Th17, and regulatory T cell responses. PGE(2) modulates chemokine production, inhibiting the attraction of proinflammatory cells while enhancing local accumulation of regulatory T cells cells and myeloid-derived suppressor cells. Targeting the production, degradation, and responsiveness to PGE(2) provides tools to modulate the patterns of immunity in a wide range of diseases, from autoimmunity to cancer.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
38
|
de Breij A, Eveillard M, Dijkshoorn L, van den Broek PJ, Nibbering PH, Joly-Guillou ML. Differences in Acinetobacter baumannii strains and host innate immune response determine morbidity and mortality in experimental pneumonia. PLoS One 2012; 7:e30673. [PMID: 22347396 PMCID: PMC3275605 DOI: 10.1371/journal.pone.0030673] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 12/20/2011] [Indexed: 11/18/2022] Open
Abstract
Despite many reports documenting its epidemicity, little is known on the interaction of Acinetobacter baumannii with its host. To deepen our insight into this relationship, we studied persistence of and host response to different A. baumannii strains including representatives of the European (EU) clones I–III in a mouse pneumonia model. Neutropenic mice were inoculated intratracheally with five A. baumannii strains and an A. junii strain and at several days morbidity, mortality, bacterial counts, airway inflammation, and chemo- and cytokine production in lungs and blood were determined. A. baumannii RUH875 and RUH134 (EU clone I and II, respectively) and sporadic strain LUH8326 resulted in high morbidity/mortality, whereas A. baumannii LUH5875 (EU clone III, which is less widespread than clone I and II) caused less symptoms. A. baumannii type strain RUH3023T and A. junii LUH5851 did not cause disease. All strains, except A. baumannii RUH3023T and A. junii LUH5851, survived and multiplied in the lungs for several days. Morbidity and mortality were associated with the severity of lung pathology and a specific immune response characterized by low levels of anti-inflammatory (IL-10) and specific pro-inflammatory (IL-12p40 and IL-23) cytokines at the first day of infection. Altogether, a striking difference in behaviour among the A. baumannii strains was observed with the clone I and II strains being most virulent, whereas the A. baumannii type strain, which is frequently used in virulence studies appeared harmless.
Collapse
Affiliation(s)
- Anna de Breij
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
39
|
gp96 expression in neutrophils is critical for the onset of Escherichia coli K1 (RS218) meningitis. Nat Commun 2011; 2:552. [PMID: 22109526 PMCID: PMC3537828 DOI: 10.1038/ncomms1554] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 10/19/2011] [Indexed: 01/01/2023] Open
Abstract
Despite the fundamental function of neutrophils (PMNs) in innate immunity, their role in Escherichia coli K1 (EC-K1) induced meningitis is unexplored. Here we show that PMN-depleted mice are resistant to EC-K1 (RS218) meningitis. EC-K1 survives and multiplies in PMNs for which outer membrane protein A (OmpA) expression is essential. EC-K1infection of PMNs increases the cell surface expression of gp96, which acts as a receptor for bacterial entry. Suppression of gp96 expression in newborn mice prevents the onset of EC-K1 meningitis. Infection of PMNs with EC-K1 suppresses oxidative burst by down regulating rac1, rac2 and gp91phox transcription both in vitro and in vivo. The interaction of loop 2 of OmpA with gp96 is essential for EC-K1-mediated inhibition of oxidative burst. These results reveal that EC-K1 exploits surface expressed gp96 in PMNs to prevent oxidative burst for the onset of neonatal meningitis.
Collapse
|
40
|
Mittal R, Krishnan S, Gonzalez-Gomez I, Prasadarao NV. Deciphering the roles of outer membrane protein A extracellular loops in the pathogenesis of Escherichia coli K1 meningitis. J Biol Chem 2011; 286:2183-93. [PMID: 21071448 PMCID: PMC3023514 DOI: 10.1074/jbc.m110.178236] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/05/2010] [Indexed: 11/06/2022] Open
Abstract
Outer membrane protein A (OmpA) has been implicated as an important virulence factor in several gram-negative bacterial infections such as Escherichia coli K1, a leading cause of neonatal meningitis associated with significant mortality and morbidity. In this study, we generated E. coli K1 mutants that express OmpA in which three or four amino acids from various extracellular loops were changed to alanines, and we examined their ability to survive in several immune cells. We observed that loop regions 1 and 2 play an important role in the survival of E. coli K1 inside neutrophils and dendritic cells, and loop regions 1 and 3 are needed for survival in macrophages. Concomitantly, E. coli K1 mutants expressing loop 1 and 2 mutations were unable to cause meningitis in a newborn mouse model. Of note, mutations in loop 4 of OmpA enhance the severity of the pathogenesis by allowing the pathogen to survive better in circulation and to produce high bacteremia levels. These results demonstrate, for the first time, the roles played by different regions of extracellular loops of OmpA of E. coli K1 in the pathogenesis of meningitis and may help in designing effective preventive strategies against this deadly disease.
Collapse
Affiliation(s)
- Rahul Mittal
- From the Division of Infectious Diseases, Department of Pediatrics, and
| | | | | | - Nemani V. Prasadarao
- From the Division of Infectious Diseases, Department of Pediatrics, and
- Surgery
- Saban Research Institute, Childrens Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California 90027
| |
Collapse
|
41
|
Mittal R, Sukumaran SK, Selvaraj SK, Wooster DG, Babu MM, Schreiber AD, Verbeek JS, Prasadarao NV. Fcγ receptor I alpha chain (CD64) expression in macrophages is critical for the onset of meningitis by Escherichia coli K1. PLoS Pathog 2010; 6:e1001203. [PMID: 21124939 PMCID: PMC2987830 DOI: 10.1371/journal.ppat.1001203] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 10/21/2010] [Indexed: 11/18/2022] Open
Abstract
Neonatal meningitis due to Escherichia coli K1 is a serious illness with unchanged morbidity and mortality rates for the last few decades. The lack of a comprehensive understanding of the mechanisms involved in the development of meningitis contributes to this poor outcome. Here, we demonstrate that depletion of macrophages in newborn mice renders the animals resistant to E. coli K1 induced meningitis. The entry of E. coli K1 into macrophages requires the interaction of outer membrane protein A (OmpA) of E. coli K1 with the alpha chain of Fcγ receptor I (FcγRIa, CD64) for which IgG opsonization is not necessary. Overexpression of full-length but not C-terminal truncated FcγRIa in COS-1 cells permits E. coli K1 to enter the cells. Moreover, OmpA binding to FcγRIa prevents the recruitment of the γ-chain and induces a different pattern of tyrosine phosphorylation of macrophage proteins compared to IgG2a induced phosphorylation. Of note, FcγRIa−/− mice are resistant to E. coli infection due to accelerated clearance of bacteria from circulation, which in turn was the result of increased expression of CR3 on macrophages. Reintroduction of human FcγRIa in mouse FcγRIa−/− macrophages in vitro increased bacterial survival by suppressing the expression of CR3. Adoptive transfer of wild type macrophages into FcγRIa−/− mice restored susceptibility to E. coli infection. Together, these results show that the interaction of FcγRI alpha chain with OmpA plays a key role in the development of neonatal meningitis by E. coli K1. Escherichia coli K1 is the most common cause of meningitis in premature infants; the mortality rate of this disease ranges from 5% to 30%. A better understanding of the pathogenesis of E. coli K1 meningitis is needed to develop new preventative strategies. We have shown that outer membrane protein A (OmpA) of E. coli K1, independent of antibody opsonization, is critical for bacterial entrance and survival within macrophages. Using a newborn mouse model, we found that depletion of macrophages renders the animals resistant to E. coli K1 induced meningitis. OmpA binds to α-chain of Fcγ-receptor I (FcγRIa) in macrophages, but does not induce expected gamma chain association and signaling. FcγRIa knockout mice are resistant to E. coli K1 infection because their macrophages express more CR3 and are thus able to kill bacteria with greater efficiency, preventing the development of high-grade bacteremia, a pre-requisite for the onset of meningitis. These novel observations demonstrate that inhibiting OmpA binding to FcγRIa is a promising therapeutic target for treatment or prevention of neonatal meningitis.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Bacterial Outer Membrane Proteins/metabolism
- Binding, Competitive
- Blotting, Western
- Brain/immunology
- Brain/metabolism
- Brain/microbiology
- COS Cells
- Chlorocebus aethiops
- Escherichia coli/growth & development
- Escherichia coli/pathogenicity
- Flow Cytometry
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Immunoprecipitation
- Macrophage-1 Antigen/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/microbiology
- Meningitis, Escherichia coli/etiology
- Meningitis, Escherichia coli/metabolism
- Meningitis, Escherichia coli/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Phagocytosis
- Phosphorylation
- RNA, Messenger/genetics
- Receptors, IgG/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Rahul Mittal
- Division of Infectious Diseases, The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sunil K. Sukumaran
- Division of Infectious Diseases, The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Suresh K. Selvaraj
- Division of Infectious Diseases, The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - David G. Wooster
- Division of Infectious Diseases, The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - M. Madan Babu
- Structural Studies Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Alan D. Schreiber
- Hematology and Oncology Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - J. Sjef Verbeek
- Department of Human Genetics, University Medical Center, Leiden, Netherlands
| | - Nemani V. Prasadarao
- Division of Infectious Diseases, The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Mittal R, Gonzalez-Gomez I, Prasadarao NV. Escherichia coli K1 promotes the ligation of CD47 with thrombospondin-1 to prevent the maturation of dendritic cells in the pathogenesis of neonatal meningitis. THE JOURNAL OF IMMUNOLOGY 2010; 185:2998-3006. [PMID: 20675593 DOI: 10.4049/jimmunol.1001296] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are professional APCs providing a critical link between adaptive and innate immune responses. Our previous studies have shown that Escherichia coli K1 internalization of myeloid DCs suppressed the maturation of the cells for which outer membrane protein A (OmpA) expression is essential. In this study, we demonstrate that infection of DCs with OmpA(+) E. coli significantly upregulates the expression of CD47, an integrin-associated protein, and its natural ligand thrombospondin 1 (TSP-1). Pretreatment of DCs with anti-CD47 blocking Ab or knocking down the expression of CD47 or TSP-1, but not signal regulatory protein alpha by small interfering RNA, abrogated the suppressive effect of E. coli K1. Ligation of CD47 with a mAb prevented the maturation and cytokine production by DCs upon stimulation with LPS similar to the inhibitory effect induced by OmpA(+) E. coli. In agreement with the in vitro studies, suppression of CD47 or TSP-1 expression in newborn mice by a novel in vivo small interfering RNA technique protected the animals against E. coli K1 meningitis. Reconstitution of CD47 knockdown mice with CD47(+) DCs renders the animals susceptible to meningitis by E. coli K1, substantiating the role of CD47 expression in DCs for the occurrence of meningitis. Our results demonstrate a role for CD47 for the first time in bacterial pathogenesis and may be a novel target for designing preventive approaches for E. coli K1 meningitis.
Collapse
Affiliation(s)
- Rahul Mittal
- Division of Infectious Diseases, Saban Research Institute, Children's Hospital, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|