1
|
Kavanagh D, Barratt J, Schubart A, Webb NJA, Meier M, Fakhouri F. Factor B as a therapeutic target for the treatment of complement-mediated diseases. Front Immunol 2025; 16:1537974. [PMID: 40028332 PMCID: PMC11868072 DOI: 10.3389/fimmu.2025.1537974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
The complement system, consisting of three initiating pathways-classical, lectin and alternative, is an important part of innate immunity. Dysregulation of the complement system is implicated in the pathogenesis of several autoimmune and inflammatory diseases. Therapeutic inhibition of the complement system has been recognized as a viable approach to drug development and has been successful with the approval of a small number of complement inhibitors for diseases such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, neuromyelitis optica, myasthenia gravis and geographic atrophy. More recently, therapies selectively targeting the alternative pathway (AP), which drives the amplification of the complement responses, are being evaluated for these complement-mediated diseases. Complement Factor B, a serine protease, is a unique component of the AP that is essential for the catalytic activity of AP C3 convertase and AP C5 convertase. Inhibition of Factor B blocks the activity of the alternative pathway and the amplification loop, and subsequent generation of the membrane attack complex downstream; however, it has no effect on the initial activation mediated by the classical and lectin complement pathways. Therefore, Factor B is an attractive target for diseases in which the AP is overactivated. In this review, we provide an overview of Factor B and its critical role in the AP, discuss the benefit-risk of Factor B inhibition as a targeted therapeutic strategy, and describe the various Factor B inhibitors that are approved and/or in clinical development.
Collapse
Affiliation(s)
- David Kavanagh
- National Renal Complement Therapeutics Centre, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, The John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Anna Schubart
- Department of Immunology, Novartis BioMedical Research, Basel, Switzerland
| | | | | | - Fadi Fakhouri
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois, UNIL, Lausanne, Switzerland
| |
Collapse
|
2
|
Single-Cell RNA-Seq Analysis Reveals Macrophages Are Involved in the Pathogenesis of Human Sporadic Acute Type A Aortic Dissection. Biomolecules 2023; 13:biom13020399. [PMID: 36830768 PMCID: PMC9952989 DOI: 10.3390/biom13020399] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Macrophages play an important role in the progression of sporadic acute type A aortic dissection (ATAAD). The aim of this study was to characterize the cellular heterogeneity of macrophages in ATAAD tissues by scRNA-seq. Ascending aortic wall tissue from six ATAAD patients and three heart transplant donors was assessed by scRNA-seq and then analyzed and validated by various bioinformatic algorithms and histopathology experiments. The results revealed that the proportion of macrophages in ATAAD tissues (24.51%) was significantly higher than that in normal tissues (13.69%). Among the six macrophage subclusters, pro-inflammatory macrophages accounted for 14.96% of macrophages in the AD group and 0.18% in the normal group. Chemokine- and inflammation-related genes (CCL2, CCL20, S100A8, and S100A9) were expressed more intensively in macrophages in ATAAD tissue than in those in normal tissue. Additionally, intercellular communication analysis and transcription factor analysis indicated the activation of inflammation and degradation of the extracellular matrix in ATAAD tissue. Finally, immunohistochemistry, immunofluorescence, and Western blot experiments confirmed the overexpression of macrophage marker genes (CD68 and CD163) and matrix metalloproteinases (MMP9 and MMP2) in ATAAD tissue. Collectively, our study provides a preliminary evaluation of the role of macrophages in ATAAD, and the results could aid in the development of therapeutic options in the future.
Collapse
|
3
|
Lian M, Wang Q, Chen S, Yang Y, Hong G. The association of serum immunoglobulin and complement levels and liver fibrosis and inflammation stage in patients with chronic hepatitis B. J Viral Hepat 2023; 30:437-447. [PMID: 36695325 DOI: 10.1111/jvh.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
The utility of measurement of serum immunoglobulin and complement in chronic hepatitis B (CHB) patients remains controversial. This study aimed to investigate the association of serum immunoglobulin and complement levels and liver fibrosis and inflammation stage in CHB patients. A total of 687 patients with CHB who underwent liver biopsy were enrolled. Serum immunoglobulin and complement were measured before liver biopsy, and liver pathological results were recorded. Associations of serum immunoglobulin and complement levels and liver fibrosis and inflammation stage were analysed. C3, C4, IgG and IgG1 had statistically significant differences among different fibrosis and different inflammation groups. Both C3 and C4 negatively correlated with fibrosis and inflammation stage, but IgG and IgG1 showed opposite results. C3, C4, IgG and IgG1 had statistical significance to predict ≥S2, ≥S3 and S4, and also had statistical significance to predict ≥G2, ≥G3 and G4. The area under curve (AUC) of the combination of C3, C4 and IgG (C3 + C4 + IgG) for predicting ≥S2, ≥S3 and S4 was 0.640 (95% CI: 0.603, 0.676), 0.674 (95% CI: 0.638, 0.709) and 0.744 (95% CI: 0.710, 0.776), respectively. The AUC of C3 + C4 + IgG for predicting ≥G2, ≥G3 and G4 was 0.723 (95% CI: 0.688, 0.756), 0.674 (95% CI: 0.638, 0.709) and 0.771 (95% CI: 0.738, 0.802), respectively. C3, C4, IgG and IgG1 are correlated with liver fibrosis and inflammation stage in CHB patients. C3, C4, IgG and IgG1 have diagnostic value for liver fibrosis and inflammation. C3 + C4 + IgG may improve diagnostic accuracy.
Collapse
Affiliation(s)
- Mingjian Lian
- Department of clinical laboratory, Xiamen Key Laboratory of Genetic Testing, the first affiliated hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qianming Wang
- Department of clinical laboratory, Xiamen Key Laboratory of Genetic Testing, the first affiliated hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shidong Chen
- Department of clinical laboratory, Xiamen Key Laboratory of Genetic Testing, the first affiliated hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuanyuan Yang
- Department of clinical laboratory, Xiamen Key Laboratory of Genetic Testing, the first affiliated hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guolin Hong
- Department of clinical laboratory, Xiamen Key Laboratory of Genetic Testing, the first affiliated hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
5
|
Schulz K, Trendelenburg M. C1q as a target molecule to treat human disease: What do mouse studies teach us? Front Immunol 2022; 13:958273. [PMID: 35990646 PMCID: PMC9385197 DOI: 10.3389/fimmu.2022.958273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
The complement system is a field of growing interest for pharmacological intervention. Complement protein C1q, the pattern recognition molecule at the start of the classical pathway of the complement cascade, is a versatile molecule with additional non-canonical actions affecting numerous cellular processes. Based on observations made in patients with hereditary C1q deficiency, C1q is protective against systemic autoimmunity and bacterial infections. Accordingly, C1q deficient mice reproduce this phenotype with susceptibility to autoimmunity and infections. At the same time, beneficial effects of C1q deficiency on disease entities such as neurodegenerative diseases have also been described in murine disease models. This systematic review provides an overview of all currently available literature on the C1q knockout mouse in disease models to identify potential target diseases for treatment strategies focusing on C1q, and discusses potential side-effects when depleting and/or inhibiting C1q.
Collapse
Affiliation(s)
- Kristina Schulz
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- *Correspondence: Kristina Schulz,
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
6
|
Kim BG, Choi DY, Kim MG, Jang AS, Suh MW, Lee JH, Oh SH, Park MK. Effect of Angiogenesis and Lymphangiogenesis in Diesel Exhaust Particles Inhalation in Mouse Model of LPS Induced Acute Otitis Media. Front Cell Infect Microbiol 2022; 12:824575. [PMID: 35646744 PMCID: PMC9132252 DOI: 10.3389/fcimb.2022.824575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/11/2022] [Indexed: 11/27/2022] Open
Abstract
Lymphangiogenesis and angiogenesis might have significant involvement in the pathogenesis of otitis media with effusion. This study investigated the effect of diesel exhaust particles (DEP) on inflammation and lymphangiogenesis in a mouse model of acute otitis media (AOM). BALB/c mice were injected with LPS and exposed to 100 µg/m3 DEP. The mice were divided into four groups: control (no stimulation), AOM, AOM + DEP, and DEP + AOM. The effects of DEP inhalation pre- and post-DEP induction were estimated based on measurements of the auditory brainstem response, mRNA levels of lymphangiogenesis-related genes and cytokines, and histology of the middle ear. Cell viability of human middle ear epithelial cells decreased in a dose-response manner at 24 and 48 hours post-DEP exposure. DEP alone did not induce AOM. AOM-induced mice with pre- or post-DEP exposure showed thickened middle ear mucosa and increased expression of TNF-α and IL1-β mRNA levels compared to the control group, but increased serum IL-1β levels were not found in the AOM + Post DEP. The mRNA expression of TLR4, VEGFA, VEGFAC, and VEGFR3 was increased by pre-AOM DEP exposure. The expression of VEFGA protein was stronger in the AOM + Post DEP group than in any other group. The expression of CD31 and CD45 markers in the mouse middle ear tissue was higher in the Pre DEP + AOM group than in the AOM group. This result implies that pre-exposure to DEP more strongly increases inflammation and lymphangiogenesis in a mouse model of acute otitis media.
Collapse
Affiliation(s)
- Byeong-Gon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
| | - Da Yeon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Min-Gyoung Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - An-Soo Jang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
| | - Seung Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
- *Correspondence: Moo Kyun Park, ;
| |
Collapse
|
7
|
Abstract
Objective Otitis media (OM) is a common reason for children to be prescribed antibiotics and undergo surgery but a thorough understanding of disease mechanisms is lacking. We evaluate the evidence of a dysregulated immune response in the pathogenesis of OM. Methods A comprehensive systematic review of the literature using search terms [otitis media OR glue ear OR AOM OR OME] OR [middle ear AND (infection OR inflammation)] which were run through Medline and Embase via Ovid, including both human and animal studies. In total, 82 955 studies underwent automated filtering followed by manual screening. One hundred studies were included in the review. Results Most studies were based on in vitro or animal work. Abnormalities in pathogen detection pathways, such as Toll-like receptors, have confirmed roles in OM. The aetiology of OM, its chronic subgroups (chronic OM, persistent OM with effusion) and recurrent acute OM is complex; however, inflammatory signalling mechanisms are frequently implicated. Host epithelium likely plays a crucial role, but the characterisation of human middle ear tissue lags behind that of other anatomical subsites. Conclusions Translational research for OM presently falls far behind its clinical importance. This has likely hindered the development of new diagnostic and treatment modalities. Further work is urgently required; particularly to disentangle the respective immune pathologies in the clinically observed phenotypes and thereby work towards more personalised treatments.
Collapse
|
8
|
Gauthier A, Wagner E, Thibeault R, Lavoie A. A Novel Case of Complement Factor B Deficiency. J Clin Immunol 2020; 41:277-279. [PMID: 33165708 DOI: 10.1007/s10875-020-00906-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/30/2020] [Indexed: 01/16/2023]
Affiliation(s)
- Amélie Gauthier
- Department of Allergy and Immunology, CHU de Québec-CHUL, Laval University Hospital Center, Laval University, Quebec City, G1V 4G2, Canada.
| | - Eric Wagner
- Immunology and Histocompatibility Laboratory, CHU de Québec-Université Laval and Department of Microbiology-Infectiology and Immunology, Laval University, Quebec City, QC, G1V 4G2, Canada
| | - Roseline Thibeault
- Department of Pediatrics, CHU de Québec-CHUL, Laval University, Quebec City, G1V 4G2, Canada
| | - Aubert Lavoie
- Department of Allergy and Immunology, CHU de Québec-CHUL, Laval University Hospital Center, Laval University, Quebec City, G1V 4G2, Canada
| |
Collapse
|
9
|
Li Q, Lu Q, Zhu MQ, Huang C, Yu KK, Huang YX, Zhao X, Luo XG, Zheng JM. Lower level of complement component C3 and C3a in the plasma means poor outcome in the patients with hepatitis B virus related acute-on-chronic liver failure. BMC Gastroenterol 2020; 20:106. [PMID: 32293297 PMCID: PMC7158068 DOI: 10.1186/s12876-020-01258-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/31/2020] [Indexed: 12/17/2022] Open
Abstract
Background The purpose of this study is to investigate whether or not the complement system is systemically activated and to specify the clinical and prognostic implications of its components during hepatitis B virus related acute-on-chronic liver failure (HBV-ACLF). Methods Blood samples were taken from twenty-seven patients diagnosed with HBV-ACLF, twenty-five patients diagnosed with chronic hepatitis B but without liver failure (CHB), and nine healthy volunteers (the control group). Plasma complement components were measured with Enzyme-linked immunosorbent assay. Correlative analysis were assessed between the levels of complement components and the liver failure related index. Results The concentrations of C3 was 6568 μg/ml in the HBV-ACLF group, 8916 μg/ml in the CHB group and 15,653 μg/ml in the control group, respectively (P < 0.05). The concentrations of C3a was 852 ng/ml in the HBV-ACLF group, 1008 ng/ml in the CHB group and 1755 ng/ml in the control group, respectively (P < 0.05). The concentrations of C1q was 50,509 ng/ml in the HBV-ACLF group, 114,640 ng/ml in the CHB group and 177,001 ng/ml in the control group, respectively (P < 0.05). The concentrations of C1q, C3, C3a, C4, C4a and sC5b-9 were significantly higher in the control group than those in the HBV-ACLF group (3.5, 2.4, 2.1, 1.4, 1.3 and 6.0 fold, respectively). However, there was no statistical significance of the differences in the plasma concentrations of mannose binding lectin and factor B between the HBV-ACLF group and control group. The levels of C3 and C3a were inversely correlated with MELDs or CLIF-C OFs (P < 0.05). Conclusions Our analysis demonstrated that the activation of the classical pathway mediated by C1q may play an important role in the pathogenesis of HBV-ACLF. Furthermore, the plasma levels of C3 and C3a may be potential novel biomarkers in predicting the outcome of HBV-ACLF.
Collapse
Affiliation(s)
- Qian Li
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Qing Lu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Meng-Qi Zhu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Chong Huang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Kang-Kang Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Yu-Xian Huang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China
| | - Xu Zhao
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China.,Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Xing-Guang Luo
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Jian-Ming Zheng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, No.12 Wurumqi Middle Road, Jing'an district, Room 508, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
10
|
Interleukin-17A Aggravates Middle Ear Injury Induced by Streptococcus pneumoniae through the p38 Mitogen-Activated Protein Kinase Signaling Pathway. Infect Immun 2017; 85:IAI.00438-17. [PMID: 28739823 DOI: 10.1128/iai.00438-17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/11/2017] [Indexed: 12/26/2022] Open
Abstract
Acute otitis media (AOM) is one of the most common bacterial infectious diseases in children aged 2 to 7 years worldwide. We previously demonstrated that interleukin-17A (IL-17A) promotes an acute inflammatory response characterized by the influx of neutrophils into the middle ear cavity during Streptococcus pneumoniae-induced AOM. In general, the inflammatory response is viewed as an effector that frequently causes local tissue damage. However, little is known about the pathogenic effects of IL-17A in AOM. Here, we investigated the pathogenic effects of IL-17A by using wild-type (WT) and IL-17A knockout (KO) mouse models. The results showed that the pathogenic effects of AOM, including weight loss, histopathological changes, and proinflammatory cytokine production, were more severe in WT mice than in IL-17A KO mice, suggesting that IL-17A aggravates tissue damage in AOM. Furthermore, these pathogenic effects were found to be dependent on p38 mitogen-activated protein kinase (MAPK) and could be reversed in the presence of a p38 MAPK-specific inhibitor. It was also demonstrated that IL-17A promoted the production of neutrophil myeloperoxidase (MPO) through the p38 MAPK signaling pathway, which was responsible for the middle ear tissue injury. These data support the conclusion that IL-17A contributes to middle ear injury through the p38 MAPK signaling pathway.
Collapse
|
11
|
Habets MN, van Selm S, van Opzeeland FJ, Simonetti E, Hermans PWM, de Jonge MI, Diavatopoulos DA. Role of antibodies and IL17-mediated immunity in protection against pneumococcal otitis media. Vaccine 2016; 34:5968-5974. [PMID: 27771185 DOI: 10.1016/j.vaccine.2016.09.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 09/12/2016] [Accepted: 09/29/2016] [Indexed: 11/19/2022]
Abstract
Widespread vaccination against Streptococcus pneumoniae (the pneumococcus) has significantly reduced pneumococcal disease caused by vaccine serotypes. Despite vaccination, overall pneumococcal colonization rates in children have not reduced and otitis media (OM) by non-vaccine serotypes remains one of the most common childhood infections. Pneumococcal surface protein A (PspA) has been shown to be a promising protein antigen to induce broad protection against pneumococcal colonization. However, its ability to protect against OM remains unclear. Using our previously established mouse model of influenza-virus induced pneumococcal OM, we here show that intranasal vaccination of mice with PspA together with the mucosal adjuvant CTB results in a decrease in pneumococcal load in the middle ears. This decrease correlated with the induction of PspA-specific IgA, a balanced IgG1:IgG2a antibody response and the induction of a mucosal Th17 response. Our data suggests that the IL-17 response to PspA is more important for protection against OM, whilst the presence of antibodies may be less important, as determined in mice deficient in IL-17 signaling or antibody production. Together, these results suggest that mucosal vaccination with PspA may not only protect against colonization, but also against the development of virus-induced pneumococcal OM.
Collapse
Affiliation(s)
- Marrit N Habets
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Saskia van Selm
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Fred J van Opzeeland
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Elles Simonetti
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter W M Hermans
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 10 (Route 412), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Cho CG, Gong SH, Kim HB, Song JJ, Park JH, Lim YS, Park SW. Role of group 3 innate lymphoid cells during experimental otitis media in a rat model. Int J Pediatr Otorhinolaryngol 2016; 88:146-52. [PMID: 27497403 DOI: 10.1016/j.ijporl.2016.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 12/13/2022]
Abstract
The objective of this study was to evaluate the role of group 3 innate lymphoid cells (ILC3) in the middle ear (ME) mucosal response to bacterial infection in a rat model. To confirm the role of ILC3 in bacterially induced otitis media (OM), the serum concentrations of IL-17 and IL-22 were determined by ELISA, and the tissue expression of IL-17 and IL-22 in infected ME mucosa was assessed by immunohistochemical staining. Immunohistochemical staining of specific cell surface markers was also assessed to confirm the origin of the cells expressing IL-17 and IL-22. Twenty Sprague-Dawley rats were used in the surgically-induced animal model of OM. OM was induced by inoculation of non-typeable Haemophilus influenzae into the ME cavity of the rats. The rats were divided into four experimental groups: three infected groups and one control group. Infected groups were subdivided into sets of 5 rats, one for each of the three time points (1, 4 and 7 days post-inoculation). For determination of rat IL-17 and IL-22 levels in infected rats and control rats, infected or control ME mucosa sections were analyzed by immunohistochemistry with specific antibodies directed against IL-17 and IL-22. Immunohistochemical staining for CD3, RORγt, and NKp46 were also conducted on the samples to confirm the origin of cells expressing IL-17 and IL-22. IL-17 and IL-22 serum concentrations were significantly increased in the infected rats compared to control rats. Immunohistochemical staining revealed increased IL-17 and IL-22 expressions in all infected ME mucosae from the first day after inoculation. In addition, the results of tissue staining for the specific surface markers were negative for CD3 and NKp46, but were highly positive for RORγt. IL-17 and IL-22 revealed their association with the bacterially induced proliferative and hyperplastic responses of ME mucosa, which are characteristic features in pathogenesis of OM. Surface marker examination showed that the source cells for IL-17 and IL-22 seemed to be lymphoid tissue inducer (LTi) cells. The results suggest that LTi cells release IL-17 and IL-22, and play a significant role in both the early phase of OM induction and recovery from it.
Collapse
Affiliation(s)
- Chang Gun Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea.
| | - Sung Ho Gong
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea
| | - Hee-Bok Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joo Hyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea
| | - Yun-Sung Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea
| | - Seok-Won Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongguk University Hospital, Goyang, Republic of Korea
| |
Collapse
|
13
|
Schnabolk G, Coughlin B, Joseph K, Kunchithapautham K, Bandyopadhyay M, O'Quinn EC, Nowling T, Rohrer B. Local production of the alternative pathway component factor B is sufficient to promote laser-induced choroidal neovascularization. Invest Ophthalmol Vis Sci 2015; 56:1850-63. [PMID: 25593023 DOI: 10.1167/iovs.14-15910] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE Complement factor B (CFB) is a required component of the alternative pathway (AP) of complement, and CFB polymorphisms are associated with age-related macular degeneration (AMD) risk. Complement factor B is made in the liver, but expression has also been detected in retina and retinal pigment epithelium (RPE)-choroid. We investigated whether production of CFB by the RPE can promote AP activation in mouse choroidal neovascularization (CNV). METHODS Transgenic mice expressing CFB under the RPE65 promoter were generated and crossed onto factor B-deficient (CFB-KO) mice. Biological activity was determined in vitro using RPE monolayers and in vivo using laser-induced CNV. Contribution of systemic CFB was investigated using CFB-KO reconstituted with CFB-sufficient serum. RESULTS Transgenic mice (CFB-tg) expressed CFB in RPE-choroid; no CFB was detected in serum. Cultured CFB-tg RPE monolayers secreted CFB apically and basally upon exposure to oxidative stress that was biologically active. Choroidal neovascularization sizes were comparable between wild-type and CFB-tg mice, but significantly increased when compared to lesions in CFB-KO mice. Injections of CFB-sufficient serum into CFB-KO mice resulted in partial reconstitution of systemic AP activity and significantly increased CNV size. CONCLUSIONS Mouse RPE cells express and secrete CFB sufficient to promote RPE damage and CNV. This further supports that local complement production may regulate disease processes; however, the reconstitution experiments suggest that additional components may be sequestered from the bloodstream. Understanding the process of ocular complement production and regulation will further our understanding of the AMD disease process and the requirements of a complement-based therapeutic.
Collapse
Affiliation(s)
- Gloriane Schnabolk
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States
| | - Beth Coughlin
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Kusumam Joseph
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Kannan Kunchithapautham
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Mausumi Bandyopadhyay
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Elizabeth C O'Quinn
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Tamara Nowling
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bärbel Rohrer
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
14
|
Gisselsson-Solén M, Hermansson A, Melhus A, Brodszki N. Immunologic findings in young children with early onset of acute otitis media. Acta Otolaryngol 2014; 134:1022-1028. [PMID: 25220724 DOI: 10.3109/00016489.2014.902539] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION No significant differences in the number of immune aberrations were seen between children with or without severe recurrent acute otitis media (rAOM); however, subnormal values of immunological markers were found more often than expected, and 4 of the 60 children had treatment-requiring immune deficiencies. OBJECTIVE Minor immunologic aberrations have been reported to be more frequent in children with rAOM. Immune investigation is recommended in children with severe rAOM, defined as six or more AOM episodes per year. The purpose of this study was to describe immunological findings in young children at high risk of developing rAOM, and to relate these to the number of expected aberrations and to the presence of severe rAOM. METHODS A total of 109 children at risk of developing rAOM were offered immune investigation including complement function, immunoglobulins with subclasses and cellular immunity. RESULTS Sixty patients were tested, 31 of whom had severe rAOM and 12 of whom did not develop rAOM. Low levels of IgG2 (27%), C1q (31%) and mannan-binding lectin (21%) were found up to eight times as often as expected. Although subnormal values were more frequent among children with severe rAOM, the study was too small to provide reliable evidence of any difference. Four children were diagnosed with immune deficiencies that required treatment.
Collapse
Affiliation(s)
- Marie Gisselsson-Solén
- Department of Otorhinolaryngology, Head and Neck Surgery, Lund University Hospital , Lund
| | | | | | | |
Collapse
|
15
|
Tong HH, Lambert G, Li YX, Thurman JM, Stahl GL, Douthitt K, Clancy C, He Y, Bowman AS. Deletion of the complement C5a receptor alleviates the severity of acute pneumococcal otitis media following influenza A virus infection in mice. PLoS One 2014; 9:e95160. [PMID: 24740152 PMCID: PMC3989264 DOI: 10.1371/journal.pone.0095160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 03/24/2014] [Indexed: 12/04/2022] Open
Abstract
There is considerable evidence that influenza A virus (IAV) promotes adherence, colonization, and superinfection by S. pneumoniae (Spn) and contributes to the pathogenesis of otitis media (OM). The complement system is a critical innate immune defense against both pathogens. To assess the role of the complement system in the host defense and the pathogenesis of acute pneumococcal OM following IAV infection, we employed a well-established transtympanically-induced mouse model of acute pneumococcal OM. We found that antecedent IAV infection enhanced the severity of acute pneumococcal OM. Mice deficient in complement C1qa (C1qa−/−) or factor B (Bf −/−) exhibited delayed viral and bacterial clearance from the middle ear and developed significant mucosal damage in the eustachian tube and middle ear. This indicates that both the classical and alternative complement pathways are critical for the oto-immune defense against acute pneumococcal OM following influenza infection. We also found that Spn increased complement activation following IAV infection. This was characterized by sustained increased levels of anaphylatoxins C3a and C5a in serum and middle ear lavage samples. In contrast, mice deficient in the complement C5a receptor (C5aR) demonstrated enhanced bacterial clearance and reduced severity of OM. Our data support the concept that C5a-C5aR interactions play a significant role in the pathogenesis of acute pneumococcal OM following IAV infection. It is possible that targeting the C5a-C5aR axis might prove useful in attenuating acute pneumococcal OM in patients with influenza infection.
Collapse
Affiliation(s)
- Hua Hua Tong
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America
| | - Garrett Lambert
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America
| | - Yong Xing Li
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America
| | - Joshua M Thurman
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Gregory L Stahl
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kelsey Douthitt
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America
| | - Caitlin Clancy
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America
| | - Yujuan He
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, United States of America; Department of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, Sichuan, P. R. China
| | - Andrew S Bowman
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
16
|
Interleukin 17A promotes pneumococcal clearance by recruiting neutrophils and inducing apoptosis through a p38 mitogen-activated protein kinase-dependent mechanism in acute otitis media. Infect Immun 2014; 82:2368-77. [PMID: 24664502 DOI: 10.1128/iai.00006-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive and human-restricted pathogen colonizing the nasopharynx with an absence of clinical symptoms as well as a major pathogen causing otitis media (OM), one of the most common childhood infections. Upon bacterial infection, neutrophils are rapidly activated and recruited to the infected site, acting as the frontline defender against emerging microbial pathogens via different ways. Evidence shows that interleukin 17A (IL-17A), a neutrophil-inducing factor, plays important roles in the immune responses in several diseases. However, its function in response to S. pneumoniae OM remains unclear. In this study, the function of IL-17A in response to S. pneumoniae OM was examined using an in vivo model. We developed a model of acute OM (AOM) in C57BL/6 mice and found that neutrophils were the dominant immune cells that infiltrated to the middle ear cavity (MEC) and contributed to bacterial clearance. Using IL-17A knockout (KO) mice, we found that IL-17A boosted neutrophil recruitment to the MEC and afterwards induced apoptosis, which was identified to be conducive to bacterial clearance. In addition, our observation suggested that the p38 mitogen-activated protein kinase (MAPK) signaling pathway was involved in the recruitment and apoptosis of neutrophils mediated by IL-17A. These data support the conclusion that IL-17A contributes to the host immune response against S. pneumoniae by promoting neutrophil recruitment and apoptosis through the p38 MAPK signaling pathway.
Collapse
|
17
|
Abstract
Otitis media (OM) is a common cause of childhood hearing loss. The large medical costs involved in treating this condition have meant that research to understand the pathology of this disease and identify new therapeutic interventions is important. There is evidence that susceptibility to OM has a significant genetic component, although little is known about the key genetic pathways involved. Mouse models for disease have become an important resource to understand a variety of human pathologies, including OM, due to the ability to easily manipulate their genetic components. This has enabled researchers to create models of acute OM, and has aided in the identification of a number of new genes associated with chronic disease, through the use of mutagenesis programs. The use of mouse models has identified a number of key molecular signalling pathways involved in the development of this condition, with genes identified from models shown to be associated with human OM.
Collapse
|
18
|
Short KR, von Köckritz-Blickwede M, Langereis JD, Chew KY, Job ER, Armitage CW, Hatcher B, Fujihashi K, Reading PC, Hermans PW, Wijburg OL, Diavatopoulos DA. Antibodies mediate formation of neutrophil extracellular traps in the middle ear and facilitate secondary pneumococcal otitis media. Infect Immun 2014; 82:364-70. [PMID: 24191297 PMCID: PMC3911859 DOI: 10.1128/iai.01104-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 10/14/2013] [Indexed: 12/16/2022] Open
Abstract
Otitis media (OM) (a middle ear infection) is a common childhood illness that can leave some children with permanent hearing loss. OM can arise following infection with a variety of different pathogens, including a coinfection with influenza A virus (IAV) and Streptococcus pneumoniae (the pneumococcus). We and others have demonstrated that coinfection with IAV facilitates the replication of pneumococci in the middle ear. Specifically, we used a mouse model of OM to show that IAV facilitates the outgrowth of S. pneumoniae in the middle ear by inducing middle ear inflammation. Here, we seek to understand how the host inflammatory response facilitates bacterial outgrowth in the middle ear. Using B cell-deficient infant mice, we show that antibodies play a crucial role in facilitating pneumococcal replication. We subsequently show that this is due to antibody-dependent neutrophil extracellular trap (NET) formation in the middle ear, which, instead of clearing the infection, allows the bacteria to replicate. We further demonstrate the importance of these NETs as a potential therapeutic target through the transtympanic administration of a DNase, which effectively reduces the bacterial load in the middle ear. Taken together, these data provide novel insight into how pneumococci are able to replicate in the middle ear cavity and induce disease.
Collapse
Affiliation(s)
- Kirsty R. Short
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | | | - Jeroen D. Langereis
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Keng Yih Chew
- Department of Zoology, The University of Melbourne, Melbourne, Australia
| | - Emma R. Job
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Charles W. Armitage
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Brandon Hatcher
- Department of Pediatric Dentistry and Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry and Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Patrick C. Reading
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Parkville, Victoria, Australia
| | - Peter W. Hermans
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Odilia L. Wijburg
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Dimitri A. Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Boué S, De León H, Schlage WK, Peck MJ, Weiler H, Berges A, Vuillaume G, Martin F, Friedrichs B, Lebrun S, Meurrens K, Schracke N, Moehring M, Steffen Y, Schueller J, Vanscheeuwijck P, Peitsch MC, Hoeng J. Cigarette smoke induces molecular responses in respiratory tissues of ApoE−/− mice that are progressively deactivated upon cessation. Toxicology 2013; 314:112-24. [DOI: 10.1016/j.tox.2013.09.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/24/2013] [Accepted: 09/24/2013] [Indexed: 12/25/2022]
|
20
|
Christaki E, Giamarellos-Bourboulis EJ. The complex pathogenesis of bacteremia: from antimicrobial clearance mechanisms to the genetic background of the host. Virulence 2013; 5:57-65. [PMID: 24067507 PMCID: PMC3916384 DOI: 10.4161/viru.26514] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bacteremia develops when bacteria manage to escape the host immune mechanisms or when the otherwise well-orchestrated immune response fails to control bacterial spread due to inherent or acquired immune defects that are associated with susceptibility to infection. The pathogenesis of bacteremia has some characteristic features that are influenced by the genetic signature of the host. In this review, the host defense mechanisms that help prevent bacteremia will be described and the populations who are at risk because of congenital or acquired deficiencies in such mechanisms will be defined. A special mention will be made to novel insights regarding host immune defense against the most commonly isolated organisms from patients with community-acquired bloodstream infections.
Collapse
Affiliation(s)
- Eirini Christaki
- Third Department of Internal Medicine; Aristotle University of Thessaloniki; Papageorgiou General Hospital; Thessaloniki, Greece; Infectious Diseases Division; Warren Alpert Medical School of Brown University; Providence, RI USA
| | - Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine; Medical School; University of Athens; Athens, Greece; Integrated Research and Treatment Center; Center for Sepsis Control and Care; Jena University Hospital; Jena, Germany
| |
Collapse
|
21
|
Murphy TF, Chonmaitree T, Barenkamp S, Kyd J, Nokso-Koivisto J, Patel JA, Heikkinen T, Yamanaka N, Ogra P, Swords WE, Sih T, Pettigrew MM. Panel 5: Microbiology and immunology panel. Otolaryngol Head Neck Surg 2013; 148:E64-89. [PMID: 23536533 DOI: 10.1177/0194599812459636] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The objective is to perform a comprehensive review of the literature from January 2007 through June 2011 on the virology, bacteriology, and immunology related to otitis media. DATA SOURCES PubMed database of the National Library of Medicine. REVIEW METHODS Three subpanels with co-chairs comprising experts in the virology, bacteriology, and immunology of otitis media were formed. Each of the panels reviewed the literature in their respective fields and wrote draft reviews. The reviews were shared with all panel members, and a second draft was created. The entire panel met at the 10th International Symposium on Recent Advances in Otitis Media in June 2011 and discussed the review and refined the content further. A final draft was created, circulated, and approved by the panel. CONCLUSION Excellent progress has been made in the past 4 years in advancing an understanding of the microbiology and immunology of otitis media. Advances include laboratory-based basic studies, cell-based assays, work in animal models, and clinical studies. IMPLICATIONS FOR PRACTICE The advances of the past 4 years formed the basis of a series of short-term and long-term research goals in an effort to guide the field. Accomplishing these goals will provide opportunities for the development of novel interventions, including new ways to better treat and prevent otitis media.
Collapse
Affiliation(s)
- Timothy F Murphy
- Clinical and Translational Research Center, University at Buffalo, State University of New York, Buffalo, New York 14203, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Li JD, Hermansson A, Ryan AF, Bakaletz LO, Brown SD, Cheeseman MT, Juhn SK, Jung TTK, Lim DJ, Lim JH, Lin J, Moon SK, Post JC. Panel 4: Recent advances in otitis media in molecular biology, biochemistry, genetics, and animal models. Otolaryngol Head Neck Surg 2013; 148:E52-63. [PMID: 23536532 DOI: 10.1177/0194599813479772] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Otitis media (OM) is the most common childhood bacterial infection and also the leading cause of conductive hearing loss in children. Currently, there is an urgent need for developing novel therapeutic agents for treating OM based on full understanding of molecular pathogenesis in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. OBJECTIVE To provide a state-of-the-art review concerning recent advances in OM in the areas of molecular biology, biochemistry, genetics, and animal model studies and to discuss the future directions of OM studies in these areas. DATA SOURCES AND REVIEW METHODS A structured search of the current literature (since June 2007). The authors searched PubMed for published literature in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. RESULTS Over the past 4 years, significant progress has been made in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. These studies brought new insights into our understanding of the molecular and biochemical mechanisms underlying the molecular pathogenesis of OM and helped identify novel therapeutic targets for OM. CONCLUSIONS AND IMPLICATIONS FOR PRACTICE Our understanding of the molecular pathogenesis of OM has been significantly advanced, particularly in the areas of inflammation, innate immunity, mucus overproduction, mucosal hyperplasia, middle ear and inner ear interaction, genetics, genome sequencing, and animal model studies. Although these studies are still in their experimental stages, they help identify new potential therapeutic targets. Future preclinical and clinical studies will help to translate these exciting experimental research findings into clinical applications.
Collapse
Affiliation(s)
- Jian-Dong Li
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Biofilm formation avoids complement immunity and phagocytosis of Streptococcus pneumoniae. Infect Immun 2013; 81:2606-15. [PMID: 23649097 DOI: 10.1128/iai.00491-13] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Streptococcus pneumoniae is a frequent member of the microbiota of the human nasopharynx. Colonization of the nasopharyngeal tract is a first and necessary step in the infectious process and often involves the formation of sessile microbial communities by this human pathogen. The ability to grow and persist as biofilms is an advantage for many microorganisms, because biofilm-grown bacteria show reduced susceptibility to antimicrobial agents and hinder recognition by the immune system. The extent of host protection against biofilm-related pneumococcal disease has not been determined yet. Using pneumococcal strains growing as planktonic cultures or as biofilms, we have investigated the recognition of S. pneumoniae by the complement system and its interactions with human neutrophils. Deposition of C3b, the key complement component, was impaired on S. pneumoniae biofilms. In addition, binding of C-reactive protein and the complement component C1q to the pneumococcal surface was reduced in biofilm bacteria, demonstrating that pneumococcal biofilms avoid the activation of the classical complement pathway. In addition, recruitment of factor H, the downregulator of the alternative pathway, was enhanced by S. pneumoniae growing as biofilms. Our results also show that biofilm formation diverts the alternative complement pathway activation by a PspC-mediated mechanism. Furthermore, phagocytosis of pneumococcal biofilms was also impaired. The present study confirms that biofilm formation in S. pneumoniae is an efficient means of evading both the classical and the PspC-dependent alternative complement pathways the host immune system.
Collapse
|
24
|
Li Q, Li YX, Douthitt K, Stahl GL, Thurman JM, Tong HH. Role of the alternative and classical complement activation pathway in complement mediated killing against Streptococcus pneumoniae colony opacity variants during acute pneumococcal otitis media in mice. Microbes Infect 2012; 14:1308-18. [PMID: 22975410 PMCID: PMC3511655 DOI: 10.1016/j.micinf.2012.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 07/30/2012] [Accepted: 08/10/2012] [Indexed: 01/24/2023]
Abstract
There is considerable evidence that phase variation among transparent and opaque colony phenotypes of Streptococcus pneumoniae (Spn) plays an important role in the pneumococcal adherence and invasion. The current study was designed to investigate the interactions of the opacity phenotype variants of Spn with specific complement pathway activation in a mouse model of acute otitis media (AOM). Although the opaque colony phenotype was expected to be more resistant to complement mediated killing compared to the transparent Spn variant, we discovered that C3b deposition on the transparent Spn is, in large part, dependent on the alternative pathway activation. There were no significant differences in resistance to complement mediated opsonophagocytosis between the two variants in factor B deficient mice. In addition, an in vitro study demonstrated that significantly more C4b-binding protein (C4BP) (the classical pathway inhibitor) and factor H (FH) (the alternative pathway inhibitor) bound to the transparent strain compared with the opaque one. Our data suggest that the difference in the relative virulence of Spn opacity phenotypes is associated with its ability to evade complement-mediated opsonophagocytosis in a mouse model of pneumococcal AOM.
Collapse
Affiliation(s)
- Qian Li
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
- Department of Infectious Diseases, FuDan University affiliated Hua Shan Hospital, Shanghai, China
| | - Yong Xing Li
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | - Kelsey Douthitt
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | - Gregory L. Stahl
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine,, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua M. Thurman
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Hua Hua Tong
- Department of Otolaryngology, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
25
|
Abstract
Pneumococcal meningitis continues to be associated with high rates of mortality and long-term neurological sequelae. The most common route of infection starts by nasopharyngeal colonization by Streptococcus pneumoniae, which must avoid mucosal entrapment and evade the host immune system after local activation. During invasive disease, pneumococcal epithelial adhesion is followed by bloodstream invasion and activation of the complement and coagulation systems. The release of inflammatory mediators facilitates pneumococcal crossing of the blood-brain barrier into the brain, where the bacteria multiply freely and trigger activation of circulating antigen-presenting cells and resident microglial cells. The resulting massive inflammation leads to further neutrophil recruitment and inflammation, resulting in the well-known features of bacterial meningitis, including cerebrospinal fluid pleocytosis, cochlear damage, cerebral edema, hydrocephalus, and cerebrovascular complications. Experimental animal models continue to further our understanding of the pathophysiology of pneumococcal meningitis and provide the platform for the development of new adjuvant treatments and antimicrobial therapy. This review discusses the most recent views on the pathophysiology of pneumococcal meningitis, as well as potential targets for (adjunctive) therapy.
Collapse
|
26
|
Essential role of factor B of the alternative complement pathway in complement activation and opsonophagocytosis during acute pneumococcal otitis media in mice. Infect Immun 2011; 79:2578-85. [PMID: 21502587 DOI: 10.1128/iai.00168-11] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We recently reported that the complement system plays a pivotal role in innate immune defense against Streptococcus pneumoniae during acute otitis media (OM) in mice. The current study was designed to determine which of the complement pathways are activated during acute pneumococcal OM and whether components of complement are expressed in the middle ear epithelium. Gene expression was determined by quantitative PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence staining. We found that S. pneumoniae induced increased gene expression of factor B of the alternative complement pathway and C3 in mouse middle ear epithelium. Activation of factor B and C3 in the middle ear lavage fluids was significantly greater than in simultaneously obtained serum samples as determined by Western blotting. Using mice deficient in complement C1qa, factor B, and factor B/C2, we found that complement C3 activation and opsonophagocytosis of S. pneumoniae were greatly attenuated in factor B- and factor B/C2-deficient mice. These findings support the concept that local complement activation is an important host innate immune response and that activation of the alternative complement pathway represents one of the innate immune defense mechanisms against pneumococcal infection during the early stage of acute OM.
Collapse
|