1
|
Cheng X, Bai X, Shang WY, Wei L, Jia JY, Yan TK, Gu QH. Profiling dendritic cells subsets in renal tissue of patients with crescentic glomerulonephritis. Int Urol Nephrol 2025; 57:263-273. [PMID: 39069601 DOI: 10.1007/s11255-024-04175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Dendritic cells (DCs) have been speculated to be involved in the pathogenesis of glomerular diseases. However, the numbers and distribution of DC subsets in the kidneys of patients with crescentic glomerulonephritis (CrGN) have not been clearly elucidated. METHODS A total of 26 patients with biopsy-proven CrGN were enrolled. Indirect immunofluorescence staining was used to quantify DC subsets in renal specimens. Double staining of HLA with CD11C, BDCA2 and CD209 respectively was performed to detect DC subsets. The correlation between DC subsets infiltrated in the kidney and clinical and pathological parameters was investigated. RESULTS DC subsets were predominantly present in the kidney interstitium, particularly in the peri-glomerular area. The numbers of CD11C+DCs, BDCA2+DCs and CD209+DCs increased in the patients with CrGN and varied among different types of CrGN. Though significant correlation between DC subsets and the percentage of crescents had not been identified, a notable increase in the number of CD11C+DCs were observed with the chronic development of crescents. Furthermore, patients with severe tubulointerstitial injury exhibited significantly more infiltrations of CD11C+DCs, BDCA2+DCs and CD209+DCs. Moreover, the numbers of CD11C+DCs and BDCA2+DCs were found to correlate with the level of serum C3. CONCLUSIONS Patients with CrGN showed increased kidney infiltration of DC subsets, primarily localized in the renal interstitium and peri-glomerular region. The correlation between DC subsets and fibrosis of crescent and severe tubulointerstitial injury implied a potential involvement of DCs in the development of CrGN.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xue Bai
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wen-Ya Shang
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Li Wei
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jun-Ya Jia
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tie-Kun Yan
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiu-Hua Gu
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
2
|
Fonceca AM, Lauzon-Joset J, Scott N, Stumbles PA, Strickland D, Everard ML. In Vivo Evidence of Respiratory Syncytial Virus Persistence in a Subset of Pulmonary Dendritic Cells Following a Primary Infection. Viral Immunol 2023; 36:466-474. [PMID: 37523237 DOI: 10.1089/vim.2023.0007] [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] [Indexed: 08/02/2023] Open
Abstract
Respiratory syncytial virus (RSV) causes annual epidemics of infections affecting the whole population. In vitro, it has been shown to infect and persist in human dendritic cells (DCs) for prolonged periods. Initially persistence is associated with low levels of replication before the virus becomes dormant. Reactivation of viral replication can be triggered many months later. Infection of DCs is likely to influence the host's ability to generate effective long-term memory responses. A well-established animal was utilized to confirm that RSV both infects and persists in pulmonary DCs in vivo. Mice were infected with a modified strain of RSV expressing red fluorescent protein (RSV-RFP) when replicating. Clinical symptoms of infection were monitored using weight change and inflammatory cell counts from bronchoalveolar lavage, which correlated with the RSV viral titer (quantitative polymerase chain reaction). Lung tissues were collected at 3, 5, 7, and 21 days postinfection (dpi) to assess leukocyte populations by flow cytometry. Clinical symptoms and RSV viral load peaked at 5 dpi. RSV-RFP was most prevalent in macrophages at 3 dpi and also observed in B cells and DCs. At 21 dpi, RSV-RFP remained evident in a subset of conventional DCs (CD103+CD11b+) even though both clinical symptoms and pulmonary inflammation had resolved. These results confirm that in this well-established mouse model, RSV persists in lung conventional DCs following resolution of the acute infection. Further work is required to explore whether the virus continues with low-level replication before becoming dormant in vivo, as has been described in vitro.
Collapse
Affiliation(s)
- Angela M Fonceca
- School of Biomedical Sciences, University of Western Australia, Nedlands, Australia
| | | | - Naomi Scott
- Telethon Kids Institute, Nedlands, Australia
| | - Philip A Stumbles
- School of Biomedical Sciences, University of Western Australia, Nedlands, Australia
- Telethon Kids Institute, Nedlands, Australia
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Australia
| | | | - Mark L Everard
- Division of Paediatrics, School of Medicine, University of Western Australia, Nedlands, Australia
| |
Collapse
|
3
|
Varghese PM, Kishore U, Rajkumari R. Human C1q Regulates Influenza A Virus Infection and Inflammatory Response via Its Globular Domain. Int J Mol Sci 2022; 23:3045. [PMID: 35328462 PMCID: PMC8949502 DOI: 10.3390/ijms23063045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
The Influenza A virus (IAV) is a severe respiratory pathogen. C1q is the first subcomponent of the complement system's classical pathway. C1q is composed of 18 polypeptide chains. Each of these chains contains a collagen-like region located at the N terminus, and a C-terminal globular head region organized as a heterotrimeric structure (ghA, ghB and ghC). This study was aimed at investigating the complement activation-independent modulation by C1q and its individual recombinant globular heads against IAV infection. The interaction of C1q and its recombinant globular heads with IAV and its purified glycoproteins was examined using direct ELISA and far-Western blotting analysis. The effect of the complement proteins on IAV replication kinetics and immune modulation was assessed by qPCR. The IAV entry inhibitory properties of C1q and its recombinant globular heads were confirmed using cell binding and luciferase reporter assays. C1q bound IAV virions via HA, NA and M1 IAV proteins, and suppressed replication in H1N1, while promoting replication in H3N2-infected A549 cells. C1q treatment further triggered an anti-inflammatory response in H1N1 and pro-inflammatory response in H3N2-infected cells as evident from differential expression of TNF-α, NF-κB, IFN-α, IFN-β, IL-6, IL-12 and RANTES. Furthermore, C1q treatment was found to reduce luciferase reporter activity of MDCK cells transfected with H1N1 pseudotyped lentiviral particles, indicative of an entry inhibitory role of C1q against infectivity of IAV. These data appear to demonstrate the complement-independent subtype specific modulation of IAV infection by locally produced C1q.
Collapse
Affiliation(s)
- Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London UB8 3PH, UK;
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London UB8 3PH, UK;
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
4
|
Varghese PM, Mukherjee S, Al-Mohanna FA, Saleh SM, Almajhdi FN, Beirag N, Alkahtani SH, Rajkumari R, Nal Rogier B, Sim RB, Idicula-Thomas S, Madan T, Murugaiah V, Kishore U. Human Properdin Released By Infiltrating Neutrophils Can Modulate Influenza A Virus Infection. Front Immunol 2021; 12:747654. [PMID: 34956182 PMCID: PMC8695448 DOI: 10.3389/fimmu.2021.747654] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The complement system is designed to recognise and eliminate invading pathogens via activation of classical, alternative and lectin pathways. Human properdin stabilises the alternative pathway C3 convertase, resulting in an amplification loop that leads to the formation of C5 convertase, thereby acting as a positive regulator of the alternative pathway. It has been noted that human properdin on its own can operate as a pattern recognition receptor and exert immune functions outside its involvement in complement activation. Properdin can bind directly to microbial targets via DNA, sulfatides and glycosaminoglycans, apoptotic cells, nanoparticles, and well-known viral virulence factors. This study was aimed at investigating the complement-independent role of properdin against Influenza A virus infection. As one of the first immune cells to arrive at the site of IAV infection, we show here that IAV challenged neutrophils released properdin in a time-dependent manner. Properdin was found to directly interact with haemagglutinin, neuraminidase and matrix 1 protein Influenza A virus proteins in ELISA and western blot. Furthermore, modelling studies revealed that properdin could bind HA and NA of the H1N1 subtype with higher affinity compared to that of H3N2 due to the presence of an HA cleavage site in H1N1. In an infection assay using A549 cells, properdin suppressed viral replication in pH1N1 subtype while promoting replication of H3N2 subtype, as revealed by qPCR analysis of M1 transcripts. Properdin treatment triggered an anti-inflammatory response in H1N1-challenged A549 cells and a pro-inflammatory response in H3N2-infected cells, as evident from differential mRNA expression of TNF-α, NF-κB, IFN-α, IFN-β, IL-6, IL-12 and RANTES. Properdin treatment also reduced luciferase reporter activity in MDCK cells transduced with H1N1 pseudotyped lentiviral particles; however, it was increased in the case of pseudotyped H3N2 particles. Collectively, we conclude that infiltrating neutrophils at the site of IAV infection can release properdin, which then acts as an entry inhibitor for pandemic H1N1 subtype while suppressing viral replication and inducing an anti-inflammatory response. H3N2 subtype can escape this immune restriction due to altered haemagglutinin and neuraminindase, leading to enhanced viral entry, replication and pro-inflammatory response. Thus, depending on the subtype, properdin can either limit or aggravate IAV infection in the host.
Collapse
Affiliation(s)
- Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Shuvechha Mukherjee
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Souad M Saleh
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fahad N Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Saad H Alkahtani
- Department of Zoology, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Beatrice Nal Rogier
- INSERM U1104 Centre d'immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Robert B Sim
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Taruna Madan
- Department of Innate Immunity, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
5
|
Tsao LC, Force J, Hartman ZC. Mechanisms of Therapeutic Antitumor Monoclonal Antibodies. Cancer Res 2021; 81:4641-4651. [PMID: 34145037 PMCID: PMC8448950 DOI: 10.1158/0008-5472.can-21-1109] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/24/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies (mAb) are a major component of cancer therapy. In this review, we summarize the different therapeutic mAbs that have been successfully developed against various tumor-expressed antigens and examine our current understanding of their different mechanisms of antitumor action. These mechanisms of action (MOA) largely center on the stimulation of different innate immune effector processes, which appear to be principally responsible for the efficacy of most unconjugated mAb therapies against cancer. This is evident in studies of mAbs targeting antigens for hematologic cancers, with emerging data also demonstrating the critical nature of innate immune-mediated mechanisms in the efficacy of anti-HER2 mAbs against solid HER2+ cancers. Although HER2-targeted mAbs were originally described as inhibitors of HER2-mediated signaling, multiple studies have since demonstrated these mAbs function largely through their engagement with Fc receptors to activate innate immune effector functions as well as complement activity. Next-generation mAbs are capitalizing on these MOAs through improvements to enhance Fc-activity, although regulation of these mechanisms may vary in different tumor microenvironments. In addition, novel antibody-drug conjugates have emerged as an important means to activate different MOAs. Although many unknowns remain, an improved understanding of these immunologic MOAs will be essential for the future of mAb therapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Li-Chung Tsao
- Department of Surgery, Duke University, Durham, North Carolina
| | - Jeremy Force
- Department of Medicine, Duke University, Durham, North Carolina
| | - Zachary C Hartman
- Department of Surgery, Duke University, Durham, North Carolina.
- Department of Pathology, Duke University, Durham, North Carolina
| |
Collapse
|
6
|
Jenkins MM, Bachus H, Botta D, Schultz MD, Rosenberg AF, León B, Ballesteros-Tato A. Lung dendritic cells migrate to the spleen to prime long-lived TCF1 hi memory CD8 + T cell precursors after influenza infection. Sci Immunol 2021; 6:eabg6895. [PMID: 34516781 DOI: 10.1126/sciimmunol.abg6895] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Meagan M Jenkins
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Holly Bachus
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Davide Botta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael D Schultz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - André Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
7
|
Complement Decay-Accelerating Factor is a modulator of influenza A virus lung immunopathology. PLoS Pathog 2021; 17:e1009381. [PMID: 34197564 PMCID: PMC8248730 DOI: 10.1371/journal.ppat.1009381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Clearance of viral infections, such as SARS-CoV-2 and influenza A virus (IAV), must be fine-tuned to eliminate the pathogen without causing immunopathology. As such, an aggressive initial innate immune response favors the host in contrast to a detrimental prolonged inflammation. The complement pathway bridges innate and adaptive immune system and contributes to the response by directly clearing pathogens or infected cells, as well as recruiting proinflammatory immune cells and regulating inflammation. However, the impact of modulating complement activation in viral infections is still unclear. In this work, we targeted the complement decay-accelerating factor (DAF/CD55), a surface protein that protects cells from non-specific complement attack, and analyzed its role in IAV infections. We found that DAF modulates IAV infection in vivo, via an interplay with the antigenic viral proteins hemagglutinin (HA) and neuraminidase (NA), in a strain specific manner. Our results reveal that, contrary to what could be expected, DAF potentiates complement activation, increasing the recruitment of neutrophils, monocytes and T cells. We also show that viral NA acts on the heavily sialylated DAF and propose that the NA-dependent DAF removal of sialic acids exacerbates complement activation, leading to lung immunopathology. Remarkably, this mechanism has no impact on viral loads, but rather on the host resilience to infection, and may have direct implications in zoonotic influenza transmissions. Exacerbated complement activation and immune deregulation are at the basis of several pathologies induced by respiratory viruses. Here, we report that complement decay-accelerating factor (DAF), which inhibits complement activation in healthy cells, increases disease severity upon influenza A virus (IAV) infection. Remarkably, DAF interaction with IAV proteins, hemagglutinin (HA) and neuraminidase (NA), resulted in excessive complement activation and recruitment of innate and adaptive immune cells, without affecting viral loads. Furthermore, we observed that viral NA directly cleaves DAF and promotes complement activation, providing a possible link between IAV-DAF interaction and pathology. Therefore, our results unveil a novel pathway that could modulate disease severity, which may help to understand the increased pathogenicity of zoonotic and pandemic IAV infections.
Collapse
|
8
|
Kohn M, Lanfermann C, Laudeley R, Glage S, Rheinheimer C, Klos A. Complement and Chlamydia psittaci: Early Complement-Dependent Events Are Important for DC Migration and Protection During Mouse Lung Infection. Front Immunol 2021; 12:580594. [PMID: 33767691 PMCID: PMC7986412 DOI: 10.3389/fimmu.2021.580594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/10/2021] [Indexed: 11/24/2022] Open
Abstract
The zoonotic intracellular bacterium Chlamydia psittaci causes life-threatening pneumonia in humans. During mouse lung infection, complement factor C3 and the anaphylatoxin C3a augment protection against C. psittaci by a so far unknown mechanism. To clarify how complement contributes to the early, innate and the late, specific immune response and resulting protection, this study addresses the amount of C3, the timing when its presence is required as well as the anaphylatoxin receptor(s) mediating its effects and the complement-dependent migration of dendritic cells. Challenge experiments with C. psittaci on various complement KO mice were combined with transient decomplementation by pharmacological treatment, as well as the analysis of in vivo dendritic cells migration. Our findings reveal that a plasma concentration of C3 close to wildtype levels was required to achieve full protection. The diminished levels of C3 of heterozygote C3+/- mice permitted already relative effective protection and improved survival as compared to C3-/- mice, but overall recovery of these animals was delayed. Complement was in particular required during the first days of infection. However, additionally, it seems to support protection at later stages. Migration of CD103+ dendritic cells from the infected lung to the draining lymph node-as prerequisite of antigen presentation-depended on C3 and C3aR and/or C5aR. Our results provide unique mechanistic insight in various aspects of complement-dependent immune responses under almost identical, rather physiological experimental conditions. Our study contributes to an improved understanding of the role of complement, and C3a in particular, in infections by intracellular bacteria.
Collapse
Affiliation(s)
- Martin Kohn
- Medical School Hannover, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Christian Lanfermann
- Medical School Hannover, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Robert Laudeley
- Medical School Hannover, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Silke Glage
- Medical School Hannover, Institute for Laboratory Animal Science, Hannover, Germany
| | - Claudia Rheinheimer
- Medical School Hannover, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Andreas Klos
- Medical School Hannover, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| |
Collapse
|
9
|
Kohn M, Lanfermann C, Laudeley R, Glage S, Rheinheimer C, Klos A. Complement and Chlamydia psittaci: Non-Myeloid-Derived C3 Predominantly Induces Protective Adaptive Immune Responses in Mouse Lung Infection. Front Immunol 2021; 12:626627. [PMID: 33746963 PMCID: PMC7969653 DOI: 10.3389/fimmu.2021.626627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Recent advances in complement research have revolutionized our understanding of its role in immune responses. The immunomodulatory features of complement in infections by intracellular pathogens, e.g., viruses, are attracting increasing attention. Thereby, local production and activation of complement by myeloid-derived cells seem to be crucial. We could recently show that C3, a key player of the complement cascade, is required for effective defense against the intracellular bacterium Chlamydia psittaci. Avian zoonotic strains of this pathogen cause life-threatening pneumonia with systemic spread in humans; closely related non-avian strains are responsible for less severe diseases of domestic animals with economic loss. To clarify how far myeloid- and non-myeloid cell-derived complement contributes to immune response and resulting protection against C. psittaci, adoptive bone marrow transfer experiments focusing on C3 were combined with challenge experiments using a non-avian (BSL 2) strain of this intracellular bacterium. Surprisingly, our data prove that for C. psittaci-induced pneumonia in mice, non-myeloid-derived, circulating/systemic C3 has a leading role in protection, in particular on the development of pathogen-specific T- and B- cell responses. In contrast, myeloid-derived and most likely locally produced C3 plays only a minor, mainly fine-tuning role. The work we present here describes authentic, although less pronounced, antigen directed immune responses.
Collapse
Affiliation(s)
- Martin Kohn
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, Hannover, Germany
| | - Christian Lanfermann
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, Hannover, Germany
| | - Robert Laudeley
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Medical School Hannover, Hannover, Germany
| | - Claudia Rheinheimer
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, Hannover, Germany
| | - Andreas Klos
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
10
|
Lu Y, Tan CTY, Gwee X, Yap KB, Fulop T, Pan F, Larbi A, Ng TP. Pathogen Burden, Blood Biomarkers and Functional Ageing in Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2021; 76:1864-1873. [PMID: 33640984 DOI: 10.1093/gerona/glab057] [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: 11/13/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Lifelong accumulation of latent or persistent or repeated infections may be a contributing factor to the deterioration of physical and cognitive function associated with functional ageing, but the evidence is limited and the biological underpinnings are unclear. METHODS We profiled the seropositivity for common viral, bacterial and plasmodial pathogens of local importance in community-living older adults in two studies involving 745 older adults (mean age 67.0, SD: 7.7 years), and 142 older adults (mean age 72.7, SD: 8.3 years). Pathogen load was related to different sets of age-related physical and cognitive measures of functional ageing and the frailty index, and plasma levels of biomarkers of inflammation, innate and adaptive immunity, and other physiological functions. RESULTS High pathogen load was associated with impaired gait speed (GS), (p<0.015), functional mobility (POMA) (p<0.029), cognitive function (MMSE) (p<<0.05), and increased frailty index (FI) (p<<0.05). High pathogen load was significantly associated with C3a complement activity (p<0.001), matrix metalloproteinase-7, macrophage inflammatory protein-1α (p<0.05), and monocyte chemoattractant protein 2 (p=0.028). Blood biomarkers did not fully explain the observed association between pathogen load and functional ageing measures. CONCLUSIONS The present study provides novel evidence linking lifelong cumulated numbers of latent, persistent or repeated infection to functional ageing, plausibly via inflammatory and immune and other biological factors.
Collapse
Affiliation(s)
- Yanxia Lu
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Shandong University, China
| | - Crystal Tze Ying Tan
- Biology of Ageing Laboratory, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Xinyi Gwee
- Gerontology Research Programme, Department of Psychological Medicine, National University Health System, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Keng Bee Yap
- Geriatric Medicine and Palliative Medicine Department, Ng Teng Fong General Hospital
| | - Tamas Fulop
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Fang Pan
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Shandong University, China
| | - Anis Larbi
- Biology of Ageing Laboratory, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.,Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Tze Pin Ng
- Gerontology Research Programme, Department of Psychological Medicine, National University Health System, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
11
|
Aggio JB, Krmeská V, Ferguson BJ, Wowk PF, Rothfuchs AG. Vaccinia Virus Infection Inhibits Skin Dendritic Cell Migration to the Draining Lymph Node. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:776-784. [PMID: 33419767 PMCID: PMC7851745 DOI: 10.4049/jimmunol.2000928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
There is a paucity of information on dendritic cell (DC) responses to vaccinia virus (VACV), including the traffic of DCs to the draining lymph node (dLN). In this study, using a mouse model of infection, we studied skin DC migration in response to VACV and compared it with the tuberculosis vaccine Mycobacterium bovis bacille Calmette-Guérin (BCG), another live attenuated vaccine administered via the skin. In stark contrast to BCG, skin DCs did not relocate to the dLN in response to VACV. Infection with UV-inactivated VACV or modified VACV Ankara promoted DC movement to the dLN, indicating that interference with skin DC migration requires replication-competent VACV. This suppressive effect of VACV was capable of mitigating responses to a secondary challenge with BCG in the skin, ablating DC migration, reducing BCG transport, and delaying CD4+ T cell priming in the dLN. Expression of inflammatory mediators associated with BCG-triggered DC migration were absent from virus-injected skin, suggesting that other pathways invoke DC movement in response to replication-deficient VACV. Despite adamant suppression of DC migration, VACV was still detected early in the dLN and primed Ag-specific CD4+ T cells. In summary, VACV blocks skin DC mobilization from the site of infection while retaining the ability to access the dLN to prime CD4+ T cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | - Veronika Krmeská
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Brian J Ferguson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Pryscilla Fanini Wowk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | | |
Collapse
|
12
|
Alon R, Sportiello M, Kozlovski S, Kumar A, Reilly EC, Zarbock A, Garbi N, Topham DJ. Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19. Nat Rev Immunol 2021; 21:49-64. [PMID: 33214719 PMCID: PMC7675406 DOI: 10.1038/s41577-020-00470-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). Understanding of the fundamental processes underlying the versatile clinical manifestations of COVID-19 is incomplete without comprehension of how different immune cells are recruited to various compartments of virus-infected lungs, and how this recruitment differs among individuals with different levels of disease severity. As in other respiratory infections, leukocyte recruitment to the respiratory system in people with COVID-19 is orchestrated by specific leukocyte trafficking molecules, and when uncontrolled and excessive it results in various pathological complications, both in the lungs and in other organs. In the absence of experimental data from physiologically relevant animal models, our knowledge of the trafficking signals displayed by distinct vascular beds and epithelial cell layers in response to infection by SARS-CoV-2 is still incomplete. However, SARS-CoV-2 and influenza virus elicit partially conserved inflammatory responses in the different respiratory epithelial cells encountered early in infection and may trigger partially overlapping combinations of trafficking signals in nearby blood vessels. Here, we review the molecular signals orchestrating leukocyte trafficking to airway and lung compartments during primary pneumotropic influenza virus infections and discuss potential similarities to distinct courses of primary SARS-CoV-2 infections. We also discuss how an imbalance in vascular activation by leukocytes outside the airways and lungs may contribute to extrapulmonary inflammatory complications in subsets of patients with COVID-19. These multiple molecular pathways are potential targets for therapeutic interventions in patients with severe COVID-19.
Collapse
Affiliation(s)
- Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Stav Kozlovski
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ashwin Kumar
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Emma C Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alexander Zarbock
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Natalio Garbi
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
13
|
Agrawal P, Sharma S, Pal P, Ojha H, Mullick J, Sahu A. The imitation game: a viral strategy to subvert the complement system. FEBS Lett 2020; 594:2518-2542. [DOI: 10.1002/1873-3468.13856] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/10/2020] [Accepted: 05/23/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Palak Agrawal
- Complement Biology Laboratory National Centre for Cell Science S. P. Pune University Campus Ganeshkhind Pune 411007 India
| | - Samriddhi Sharma
- Complement Biology Laboratory National Centre for Cell Science S. P. Pune University Campus Ganeshkhind Pune 411007 India
| | - Pradipta Pal
- Complement Biology Laboratory National Centre for Cell Science S. P. Pune University Campus Ganeshkhind Pune 411007 India
| | - Hina Ojha
- Complement Biology Laboratory National Centre for Cell Science S. P. Pune University Campus Ganeshkhind Pune 411007 India
| | - Jayati Mullick
- Microbial Containment Complex ICMR‐National Institute of Virology Pune 411021 India
| | - Arvind Sahu
- Complement Biology Laboratory National Centre for Cell Science S. P. Pune University Campus Ganeshkhind Pune 411007 India
| |
Collapse
|
14
|
Murugaiah V, Varghese PM, Saleh SM, Tsolaki AG, Alrokayan SH, Khan HA, Collison KS, Sim RB, Nal B, Al-Mohanna FA, Kishore U. Complement-Independent Modulation of Influenza A Virus Infection by Factor H. Front Immunol 2020; 11:355. [PMID: 32269562 PMCID: PMC7109256 DOI: 10.3389/fimmu.2020.00355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/13/2020] [Indexed: 02/05/2023] Open
Abstract
The complement system is an ancient innate immune defense mechanism that can recognize molecular patterns on the invading pathogens. Factor H, as an inhibitor of the alternative pathway, down-regulates complement activation on the host cell surface. Locally synthesized factor H at the site of infection/injury, including lungs, can act as a pattern recognition molecule without involving complement activation. Here, we report that factor H, a sialic acid binder, interacts with influenza A virus (IAV) and modulates IAV entry, as evident from down-regulation of matrix protein 1 (M1) in H1N1 subtype-infected cells and up-regulation of M1 expression in H3N2-infected A549 cells. Far-western blot revealed that factor H binds hemagglutinin (HA, ~70 kDa), neuraminidase (NA, ~60 kDa), and M1 (~25 kDa). IAV-induced transcriptional levels of IFN-α, TNF-α, IL-12, IL-6, IFN-α, and RANTES were reduced following factor H treatment for the H1N1 subtype at 6 h post-infection. However, for the H3N2 subtype, mRNA levels of these pro-inflammatory cytokines were enhanced. A recombinant form of vaccinia virus complement control protein (VCP), which like factor H, contains CCP modules and has complement-regulatory activity, mirrored the results obtained with factor H. Both factor H (25%), and VCP (45%) were found to reduce luciferase reporter activity in MDCK cells transduced with H1N1 pseudotyped lentiviral particles. Factor H (50%) and VCP (30%) enhanced the luciferase reporter activity for H3N2, suggesting an entry inhibitory role of factor H and VCP against H1N1, but not H3N2. Thus, factor H can modulate IAV infection and inflammatory responses, independent of its complement-related functions.
Collapse
Affiliation(s)
- Valarmathy Murugaiah
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Praveen M. Varghese
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Soad M. Saleh
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Anthony G. Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Salman H. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haseeb A. Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kate S. Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Robert B. Sim
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Béatrice Nal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Futwan A. Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
15
|
León B, Lund FE. Compartmentalization of dendritic cell and T-cell interactions in the lymph node: Anatomy of T-cell fate decisions. Immunol Rev 2019; 289:84-100. [PMID: 30977197 PMCID: PMC6464380 DOI: 10.1111/imr.12758] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022]
Abstract
Upon receiving cognate and co-stimulatory priming signals from antigen (Ag)-presenting dendritic cells (DCs) in secondary lymphoid tissues, naïve CD4+ T cells differentiate into distinct effector and memory populations. These alternate cell fate decisions, which ultimately control the T-cell functional attributes, are dictated by programming signals provided by Ag-bearing DCs and by other cells that are present in the microenvironment in which T-cell priming occurs. We know that DCs can be subdivided into multiple populations and that the various DC subsets exhibit differential capacities to initiate development of the different CD4+ T-helper populations. What is less well understood is why different subanatomic regions of secondary lymphoid tissues are colonized by distinct populations of Ag-presenting DCs and how the location of these DCs influences the type of T-cell response that will be generated. Here we review how chemokine receptors and their ligands, which position allergen and nematode-activated DCs within different microdomains of secondary lymphoid tissues, contribute to the establishment of IL-4 committed follicular helper T and type 2 helper cell responses.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frances E. Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
16
|
Inhibition of the alternative complement pathway accelerates repair processes in the murine model of choroidal neovascularization. Mol Immunol 2019; 108:8-12. [PMID: 30763805 DOI: 10.1016/j.molimm.2019.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/10/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in the US. Polymorphisms in complement components are associated with increased AMD risk, and it has been hypothesized that an overactive complement system is partially responsible for AMD pathology. Choroidal neovascularization (CNV) has two phases, injury/angiogenesis and repair/fibrosis. Complement activation has been shown to be involved in the angiogenesis phase of murine CNV, but has not been investigated during repair. Anaphylatoxin (C3a and C5a) signaling in particular has been shown to be involved in both tissue injury and repair in other models. CNV was triggered by laser-induced photocoagulation in C57BL/6 J mice, and lesion sizes measured by optical coherence tomography. Alternative pathway (AP) activation or C3a-receptor (C3aR) and C5a-receptor (C5aR) engagement was inhibited during the repair phase only of CNV with the AP-inhibitor CR2-fH, a C3aR antagonist (N2-[(2,2-diphenylethoxy)acetyl]-l-arginine, TFA), or a C5a blocking antibody (CLS026), respectively. Repair after CNV was also investigated in C3aR/C5aR double knockout mice. CR2-fH treatment normalized anaphylatoxin levels in the eye and accelerated regression of CNV lesions. In contrast, blockade of anaphylatoxin-receptor signaling pharmacologically or genetically did not significantly alter the course of lesion repair. These results suggest that continued complement activation prevents fibrotic scar resolution, and emphasizes the importance of reducing anaphylatoxins to homeostatic levels. This duality of complement, playing a role in injury and repair, will need to be considered when selecting a complement inhibitory strategy for AMD.
Collapse
|
17
|
Ng SL, Teo YJ, Setiagani YA, Karjalainen K, Ruedl C. Type 1 Conventional CD103 + Dendritic Cells Control Effector CD8 + T Cell Migration, Survival, and Memory Responses During Influenza Infection. Front Immunol 2018; 9:3043. [PMID: 30622538 PMCID: PMC6308161 DOI: 10.3389/fimmu.2018.03043] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022] Open
Abstract
Type 1 conventional CD103+ dendritic cells (cDC1) contribute significantly to the cytotoxic T lymphocyte (CTL) response during influenza virus infection; however, the mechanisms by which cDC1s promote CTL recruitment and viral clearance are unclear. We demonstrate that cDC1 ablation leads to a deficient influenza-specific primary CD8+ T cell response alongside severe pulmonary inflammation, intensifying susceptibility to infection. The diminished pulmonary CTL population is not only a consequence of reduced priming in the lymph node (LN), but also of dysregulated CD8+ T cell egression from the LN and reduced CD8+ T cell viability in the lungs. cDC1s promote S1PR expression on CTLs, a key chemokine receptor facilitating CTL LN egress, and express high levels of the T cell survival cytokine, IL-15, to support CTL viability at the site of infection. Moreover, cDC1 ablation leads to severe impairment of CD8+ T cell memory recall and cross-reactive protection, suggesting that cDC1 are not only involved in primary T cell activation, but also in supporting the development of effective memory CD8+ T cell precursors. Our findings demonstrate a previously unappreciated and multifaceted role of CD103+ DCs in controlling pulmonary T cell-mediated immune responses.
Collapse
Affiliation(s)
- See Liang Ng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yi Juan Teo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Klaus Karjalainen
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
18
|
Zha H, Wang X, Zhu Y, Chen D, Han X, Yang F, Gao J, Hu C, Shu C, Feng Y, Tan Y, Zhang J, Li Y, Wan YY, Guo B, Zhu B. Intracellular Activation of Complement C3 Leads to PD-L1 Antibody Treatment Resistance by Modulating Tumor-Associated Macrophages. Cancer Immunol Res 2018; 7:193-207. [PMID: 30514794 DOI: 10.1158/2326-6066.cir-18-0272] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/27/2018] [Accepted: 11/29/2018] [Indexed: 11/16/2022]
Abstract
Complement aids in the construction of an immunosuppressive tumor microenvironment. Tumor cell-derived C3 has been previously reported, but whether and how it acts on antitumor immunity remains to be elucidated. Here, we describe a mechanism for tumor cell-derived C3 in suppressing antitumor immunity. Tumor cell-derived C3 was activated intracellularly, which results in generation of C3a. C3a modulated tumor-associated macrophages via C3a-C3aR-PI3Kγ signaling, thereby repressing antitumor immunity. Deletion of C3 in tumor cells that had high C3 expression enhanced efficacy of anti-PD-L1 treatment. Collectively, our results suggest tumor cell-derived C3 may be a useful target for cancer immunotherapy and that targeting C3 in tumor cells may enhance antitumor immunity.
Collapse
Affiliation(s)
- Haoran Zha
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China.,Department of Oncology, The General Hospital of the PLA Rocket Force, Beijing, P.R. China
| | - Xinxin Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Ying Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Diangang Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Xiao Han
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Fei Yang
- Department of Immunology, Third Military Medical University, Chongqing, P.R. China
| | - Jianbao Gao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Chunyan Hu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Chi Shu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Yi Feng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| | - Yulong Tan
- Institute of Tropical Medicine, Third Military Medical University, Chongqing, P.R. China
| | - Jinyu Zhang
- Department of Immunology, Third Military Medical University, Chongqing, P.R. China
| | - Yongsheng Li
- Clinical Medicine Research Center and Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Yisong Y Wan
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Centre, University of North Carolina at Chapel Hill, Chapel Hill, North California
| | - Bo Guo
- Maternal and Child Health Research Institute, Baoan Women's and Children's Hospital, Jinan University, Shenzhen, P.R. China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China. .,Chongqing Key Laboratory of Immunotherapy, Chongqing, P.R. China
| |
Collapse
|
19
|
Gujar R, Sen P. Transforming growth factor-β1 impairs lymph node homing of dendritic cells by downregulating C-type lectin receptor-2 expression. Cytokine 2018; 110:39-43. [DOI: 10.1016/j.cyto.2018.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022]
|
20
|
Complement C3 Plays a Key Role in Inducing Humoral and Cellular Immune Responses to Influenza Virus Strain-Specific Hemagglutinin-Based or Cross-Protective M2 Extracellular Domain-Based Vaccination. J Virol 2018; 92:JVI.00969-18. [PMID: 30068650 DOI: 10.1128/jvi.00969-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
The complement pathway is involved in eliminating antigen immune complexes. However, the role of the C3 complement system remains largely unknown in influenza virus M2 extracellular (M2e) domain or hemagglutinin (HA) vaccine-mediated protection after vaccination. Using a C3 knockout (C3 KO) mouse model, we found that complement protein C3 was required for effective induction of immune responses to vaccination with M2e-based or HA-based vaccines, which include isotype class-switched antibodies and effector CD4 and CD8 T cell responses. C3 KO mice after active immunization with cross-protective nonneutralizing M2e-based vaccine were not protected against influenza virus, although low levels of M2e-specific antibodies were protective after passive coadministration with virus in wild-type mice. In contrast, C3 KO mice that were immunized with strain-specific neutralizing HA-based vaccine were protected against homologous virus challenge despite lower levels of HA antibody responses. C3 KO mice showed impaired maintenance of innate immune cells and a defect in innate immune responses upon exposure to antigens. The findings in this study suggest that C3 is required for effective induction of humoral and cellular adaptive immune responses as well as protective immunity after nonneutralizing influenza M2e vaccination.IMPORTANCE Complement is the well-known innate immune defense system involved in the opsonization and lysis of pathogens but is less studied in establishing adaptive immunity after vaccination. Influenza virus HA-based vaccination confers protection via strain-specific neutralizing antibodies, whereas M2e-based vaccination induces a broad spectrum of protection by immunity against the conserved M2e epitopes. This study revealed the critical roles of C3 complement in inducing humoral and cellular immune responses after immunization with M2e or HA vaccines. C3 was found to be required for protection by M2e-based but not by HA-based active vaccination as well as for maintaining innate antigen-presenting cells. Findings in this study have insight into better understanding the roles of C3 complement in inducing effective innate and adaptive immunity as well as in conferring protection by cross-protective conserved M2e vaccination.
Collapse
|
21
|
Zhang K, Xu WW, Zhang Z, Liu J, Li J, Sun L, Sun W, Jiao P, Sang X, Ren Z, Yu Z, Li Y, Feng N, Wang T, Wang H, Yang S, Zhao Y, Zhang X, Wilker PR, Liu W, Liao M, Chen H, Gao Y, Xia X. The innate immunity of guinea pigs against highly pathogenic avian influenza virus infection. Oncotarget 2018; 8:30422-30437. [PMID: 28418930 PMCID: PMC5444753 DOI: 10.18632/oncotarget.16503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022] Open
Abstract
H5N1 avian influenza viruses are a major pandemic concern. In contrast to the highly virulent phenotype of H5N1 in humans and many animal models, guinea pigs do not typically display signs of severe disease in response to H5N1 virus infection. Here, proteomic and transcriptional profiling were applied to identify host factors that account for the observed attenuation of A/Tiger/Harbin/01/2002 (H5N1) virulence in guinea pigs. RIG-I and numerous interferon stimulated genes were among host proteins with altered expression in guinea pig lungs during H5N1 infection. Overexpression of RIG-I or the RIG-I adaptor protein MAVS in guinea pig cell lines inhibited H5N1 replication. Endogenous GBP-1 expression was required for RIG-I mediated inhibition of viral replication upstream of the activity of MAVS. Furthermore, we show that guinea pig complement is involved in viral clearance, the regulation of inflammation, and cellular apoptosis during influenza virus infection of guinea pigs. This work uncovers features of the guinea pig innate immune response to influenza that may render guinea pigs resistant to highly pathogenic influenza viruses.
Collapse
Affiliation(s)
- Kun Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China.,Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298, USA
| | - Wei Wei Xu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Zhaowei Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Jing Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lijuan Sun
- Department of Influenza Vaccine, Changchun Institute of Biological Product, Changchun, 130062, PR China
| | - Weiyang Sun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Peirong Jiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, PR China
| | - Xiaoyu Sang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Zhiguang Ren
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Zhijun Yu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Yuanguo Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Xuemei Zhang
- Department of Influenza Vaccine, Changchun Institute of Biological Product, Changchun, 130062, PR China
| | - Peter R Wilker
- Department of Microbiology, University of Wisconsin La Crosse, La Crosse, Wisconsin, 54601, USA
| | - WenJun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, PR China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, PR China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, The Military Veterinary Institute, Academy of Military Medical Science of PLA, Changchun, 130122, PR China
| |
Collapse
|
22
|
Lee DCP, Tay NQ, Thian M, Prabhu N, Furuhashi K, Kemeny DM. Prior exposure to inhaled allergen enhances anti-viral immunity and T cell priming by dendritic cells. PLoS One 2018; 13:e0190063. [PMID: 29293541 PMCID: PMC5749744 DOI: 10.1371/journal.pone.0190063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/07/2017] [Indexed: 01/12/2023] Open
Abstract
Influenza and asthma are two of the major public health concerns in the world today. During the 2009 influenza pandemic asthma was found to be the commonest comorbid illness of patients admitted to hospital. Unexpectedly, it was also observed that asthmatic patients admitted to hospital with influenza infection were less likely to die or require admission to intensive care compared with non-asthmatics. Using an in vivo model of asthma and influenza infection we demonstrate that prior exposure to Blomia tropicalis extract (BTE) leads to an altered immune response to influenza infection, comprised of less severe weight loss and faster recovery following infection. This protection was associated with significant increases in T cell numbers in the lungs of BTE sensitised and infected mice, as well as increased IFN-γ production from these cells. In addition, elevated numbers of CD11b+ dendritic cells (DCs) were found in the lung draining lymph nodes following infection of BTE sensitised mice compared to infected PBS treated mice. These CD11b+ DCs appeared to be better at priming CD8 specific T cells both in vivo and ex vivo, a function not normally attributed to CD11b+ DCs. We propose that this alteration in cross-presentation and more efficient T cell priming seen in BTE sensitised mice, led to the earlier increase in T cells in the lungs and subsequently faster clearance of the virus and reduced influenza induced pathology. We believe this data provides a novel mechanism that explains why asthmatic patients may present with less severe disease when infected with influenza.
Collapse
Affiliation(s)
- Debbie C. P. Lee
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
- * E-mail:
| | - Neil Q. Tay
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
| | - Marini Thian
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
| | - Nayana Prabhu
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
| | - Kazuki Furuhashi
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
| | - David M. Kemeny
- Immunology Programme, Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore
| |
Collapse
|
23
|
Killick J, Morisse G, Sieger D, Astier AL. Complement as a regulator of adaptive immunity. Semin Immunopathol 2018; 40:37-48. [PMID: 28842749 PMCID: PMC5794818 DOI: 10.1007/s00281-017-0644-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
The complement system is an ancient and evolutionarily conserved effector system comprising in mammals over 50 circulating and membrane bound proteins. Complement has long been described as belonging to the innate immune system; however, a number of recent studies have demonstrated its key role in the modulation of the adaptive immune response. This review does not set out to be an exhaustive list of the numerous interactions of the many complement components with adaptive immunity; rather, we will focus more precisely on the role of some complement molecules in the regulation of antigen presenting cells, as well as on their direct effect on the activation of the core adaptive immune cells, B and T lymphocytes. Recent reports on the local production and activation of complement proteins also suggest a major role in the control of effector responses. The crucial role of complement in adaptive immunity is further highlighted by several examples of dysregulation of these pathways in human diseases.
Collapse
Affiliation(s)
- Justin Killick
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Gregoire Morisse
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Dirk Sieger
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Anne L Astier
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK.
- Inserm U1043, CNRS U5282, Université de Toulouse, Centre de Physiopathologie Toulouse-Purpan (CPTP), F-31300, Toulouse, France.
| |
Collapse
|
24
|
Multiplex peptide-MHC tetramer staining using mass cytometry for deep analysis of the influenza-specific T-cell response in mice. J Immunol Methods 2017; 453:30-36. [PMID: 28974367 DOI: 10.1016/j.jim.2017.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/19/2017] [Accepted: 09/29/2017] [Indexed: 11/24/2022]
Abstract
Antigen-specific T cells play a crucial role for the host protective immunity against viruses and other diseases. The use of mass cytometry together with a combinatorial multiplex tetramer staining has successfully been applied for probing and characterization of multiple antigen-specific CD8+ T cells in human blood samples. The present study shows that this approach can also be used to rapidly assess the magnitude of influenza-specific CD8+ T cell epitope dominance across lymph nodes and lungs in a murine model of a highly pathological influenza infection. Moreover, we show feasibility of extending this approach to include concurrent identification of virus-specific CD4+ T cells. By using a double coding approach, we probed for five influenza-specific MHCI-peptide complexes as well as one influenza-specific MHCII-peptide complex in the presence of irrelevant control peptides and show that this approach is capable of tracking antigen-specific T cells across individual lymph nodes and lungs. The simultaneous staining with 26 surface maker molecules further facilitated an in-depth characterization of T cells reacting with influenza epitopes and revealed tissue specific phenotypic differences between CD4+ T cells targeting the same pathogenic epitope. In conclusion, this approach provides the possibility for a rapid and comprehensive analysis of antigen-specific CD8+ and CD4+ T cells in different disease settings that might be advantageous for subsequent vaccine formulation strategies.
Collapse
|
25
|
Freeley S, Kemper C, Le Friec G. The "ins and outs" of complement-driven immune responses. Immunol Rev 2017; 274:16-32. [PMID: 27782335 DOI: 10.1111/imr.12472] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The complement system represents an evolutionary old and critical component of innate immunity where it forms the first line of defense against invading pathogens. Originally described as a heat-labile fraction of the serum responsible for the opsonization and subsequent lytic killing of bacteria, work over the last century firmly established complement as a key mediator of the general inflammatory response but also as an acknowledged vital bridge between innate and adaptive immunity. However, recent studies particularly spanning the last decade have provided new insights into the novel modes and locations of complement activation and highlighted unexpected additional biological functions for this ancient system, for example, in regulating basic processes of the cell. In this review, we will cover the current knowledge about complement's established and novel roles in innate and adaptive immunity with a focus on the functional differences between serum circulating and intracellularly active complement and will describe and discuss the newly discovered cross-talks of complement with other cell effector systems particularly during T-cell induction and contraction.
Collapse
Affiliation(s)
- Simon Freeley
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK. .,Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Gaëlle Le Friec
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
26
|
Novel insights into the expression pattern of anaphylatoxin receptors in mice and men. Mol Immunol 2017; 89:44-58. [PMID: 28600003 DOI: 10.1016/j.molimm.2017.05.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
Abstract
The anaphylatoxins (AT) C3a and C5a play important roles as mediators of inflammation. Further, they regulate and control multiple innate and adaptive immune responses through binding and activation of their cognate G protein-coupled receptors, i.e. C3a receptor (C3aR), C5a receptor 1 (C5aR1) and C5a receptor 2 (C5aR2), although the latter lacks important sequence motifs for G protein-coupling. Based on their pleiotropic functions, they contribute not only to tissue homeostasis but drive, perpetuate and resolve immune responses in many inflammatory diseases including infections, malignancies, autoimmune as well as allergic diseases. During the past few years, transcriptome expression data provided detailed insights into AT receptor tissue mRNA expression. In contrast, our understanding of cellular AT receptor expression in human and mouse tissues under steady and inflammatory conditions is still sketchy. Ligand binding studies, flow cytometric and immunohistochemical analyses convincingly demonstrated tissue-specific C5aR1 expression in various cells of myeloid origin. However, a detailed map for C3aR or C5aR2 expression in human or mouse tissue cells is still lacking. Also, reports about AT expression in lymphoid cells is still controversial. To understand the multiple roles of the ATs in the innate and adaptive immune networks, a detailed understanding of their receptor expression in health and disease is required. Recent findings obtained with novel GFP or tdTomato AT-receptor knock-in mice provide detailed insights into their expression pattern in tissue immune and stroma cells. Here, we will provide an update about our current knowledge of AT receptor expression pattern in humans and mice.
Collapse
|
27
|
Rattan A, Pawar SD, Nawadkar R, Kulkarni N, Lal G, Mullick J, Sahu A. Synergy between the classical and alternative pathways of complement is essential for conferring effective protection against the pandemic influenza A(H1N1) 2009 virus infection. PLoS Pathog 2017; 13:e1006248. [PMID: 28301559 PMCID: PMC5354441 DOI: 10.1371/journal.ppat.1006248] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 02/17/2017] [Indexed: 12/22/2022] Open
Abstract
The pandemic influenza A(H1N1) 2009 virus caused significant morbidity and mortality worldwide thus necessitating the need to understand the host factors that influence its control. Previously, the complement system has been shown to provide protection during the seasonal influenza virus infection, however, the role of individual complement pathways is not yet clear. Here, we have dissected the role of intact complement as well as of its individual activation pathways during the pandemic influenza virus infection using mouse strains deficient in various complement components. We show that the virus infection in C3-/- mice results in increased viral load and 100% mortality, which can be reversed by adoptive transfer of naïve wild-type (WT) splenocytes, purified splenic B cells, or passive transfer of immune sera from WT, but not C3-/- mice. Blocking of C3a and/or C5a receptor signaling in WT mice using receptor antagonists and use of C3aR-/- and C5aR-/- mice showed significant mortality after blocking/ablation of C3aR, with little or no effect after blocking/ablation of C5aR. Intriguingly, deficiency of C4 and FB in mice resulted in only partial mortality (24%-32%) suggesting a necessary cross-talk between the classical/lectin and alternative pathways for providing effective protection. In vitro virus neutralization experiments performed to probe the cross-talk between the various pathways indicated that activation of the classical and alternative pathways in concert, owing to coating of viral surface by antibodies, is needed for its efficient neutralization. Examination of the virus-specific complement-binding antibodies in virus positive subjects showed that their levels vary among individuals. Together these results indicate that cooperation between the classical and alternative pathways not only result in efficient direct neutralization of the pandemic influenza virus, but also lead to the optimum generation of C3a, which when sensed by the immune cells along with the antigen culminates in generation of effective protective immune responses. The pandemic influenza A(H1N1) 2009 virus is now circulating seasonally and causing a significant disease burden worldwide. Hence, it is important to delineate the immune components required for protection against its infection. Here we demonstrate that presence of intact complement is essential for clearing the pandemic influenza virus infection, wherein complement synthesized by B cells plays a major role. Further, we show that activation of the classical as well as alternative pathways is a requisite for efficient neutralization of the virus as well as the optimum generation of C3a, which is necessary for boosting the protective immune responses. Our results thus reveal that deficiencies of components of the classical and alternative pathways enhance the susceptibility to and severity of the pandemic influenza virus infection.
Collapse
Affiliation(s)
- Ajitanuj Rattan
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, India
| | - Shailesh D. Pawar
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - Renuka Nawadkar
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, India
| | - Neeraja Kulkarni
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, India
| | - Girdhari Lal
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, India
| | - Jayati Mullick
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - Arvind Sahu
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, India
- * E-mail:
| |
Collapse
|
28
|
Khameneh HJ, Ho AWS, Laudisi F, Derks H, Kandasamy M, Sivasankar B, Teng GG, Mortellaro A. C5a Regulates IL-1β Production and Leukocyte Recruitment in a Murine Model of Monosodium Urate Crystal-Induced Peritonitis. Front Pharmacol 2017; 8:10. [PMID: 28167912 PMCID: PMC5253373 DOI: 10.3389/fphar.2017.00010] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/04/2017] [Indexed: 12/31/2022] Open
Abstract
Gouty arthritis results from the generation of monosodium urate (MSU) crystals within joints. These MSU crystals elicit acute inflammation characterized by massive infiltration of neutrophils and monocytes that are mobilized by the pro-inflammatory cytokine IL-1β. MSU crystals also activate the complement system, which regulates the inflammatory response; however, it is unclear whether or how MSU-mediated complement activation is linked to IL-1β release in vivo, and the various roles that might be played by individual components of the complement cascade. Here we show that exposure to MSU crystals in vivo triggers the complement cascade, leading to the generation of the biologically active complement proteins C3a and C5a. C5a, but not C3a, potentiated IL-1β and IL-1α release from LPS–primed MSU-exposed peritoneal macrophages and human monocytic cells in vitro; while in vivo MSU–induced C5a mediated murine neutrophil recruitment as well as IL-1β production at the site of inflammation. These effects were significantly ameliorated by treatment of mice with a C5a receptor antagonist. Mechanistic studies revealed that C5a most likely increased NLRP3 inflammasome activation via production of reactive oxygen species (ROS), and not through increased transcription of inflammasome components. Therefore we conclude that C5a generated upon MSU-induced complement activation increases neutrophil recruitment in vivo by promoting IL-1 production via the generation of ROS, which activate the NLRP3 inflammasome. Identification of the C5a receptor as a key determinant of IL-1-mediated recruitment of inflammatory cells provides a novel potential target for therapeutic intervention to mitigate gouty arthritis.
Collapse
Affiliation(s)
- Hanif J Khameneh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Adrian W S Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Federica Laudisi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Heidi Derks
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Matheswaran Kandasamy
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Baalasubramanian Sivasankar
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| | - Gim Gee Teng
- Division of Rheumatology, University Medicine Cluster, National University Health System (NUHS)Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore (NUS) and National University Health System (NUHS)Singapore, Singapore
| | - Alessandra Mortellaro
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) Singapore, Singapore
| |
Collapse
|
29
|
Lee TY, Kim CU, Bae EH, Seo SH, Jeong DG, Yoon SW, Chang KT, Kim YS, Kim SH, Kim DJ. Outer membrane vesicles harboring modified lipid A moiety augment the efficacy of an influenza vaccine exhibiting reduced endotoxicity in a mouse model. Vaccine 2016; 35:586-595. [PMID: 28024958 PMCID: PMC7115551 DOI: 10.1016/j.vaccine.2016.12.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/24/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
Influenza is an acute respiratory disease and a major health problem worldwide. Since mucosal immunity plays a critical role in protection against influenza virus infection, mucosal immunization is considered a promising vaccination route. However, except for live-attenuated vaccines, there are no effective killed or recombinant mucosal influenza vaccines to date. Outer membrane vesicles (OMVs) are nano-sized vesicles produced by gram-negative bacteria, and contain various bacterial components capable of stimulating the immune system of the host. We generated an OMV with low endotoxicity (fmOMV) by modifying the structure of the lipid A moiety of lipopolysaccharide and investigated its effect as an intranasal vaccine adjuvant in an influenza vaccine model. In this model, fmOMV exhibited reduced toll-like receptor 4-stimulating activity and attenuated endotoxicity compared to that of native OMV. Intranasal injection of the vaccine antigen with fmOMV significantly increased systemic antibody and T cell responses, mucosal IgA levels, and the frequency of lung-resident influenza-specific T cells. In addition, the number of antigen-bearing CD103+ dendritic cells in the mediastinal lymph nodes was significantly increased after fmOMV co-administration. Notably, the mice co-immunized with fmOMV showed a significantly higher protection rate against challenge with a lethal dose of homologous or heterologous influenza viruses without adverse effects. These results show the potential of fmOMV as an effective mucosal adjuvant for intranasal vaccines.
Collapse
Affiliation(s)
- Tae-Young Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Eun-Hye Bae
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Sang-Hwan Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Dae Gwin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Sun-Woo Yoon
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Kyu-Tae Chang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young Sang Kim
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Sang-Hyun Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea.
| |
Collapse
|
30
|
Koh VHQ, Ng SL, Ang MLT, Lin W, Ruedl C, Alonso S. Role and contribution of pulmonary CD103 + dendritic cells in the adaptive immune response to Mycobacterium tuberculosis. Tuberculosis (Edinb) 2016; 102:34-46. [PMID: 28061951 DOI: 10.1016/j.tube.2016.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/24/2016] [Accepted: 12/05/2016] [Indexed: 01/17/2023]
Abstract
Despite international control programmes, the global burden of tuberculosis remains enormous. Efforts to discover novel drugs have largely focused on targeting the bacterium directly. Alternatively, manipulating the host immune response may represent a valuable approach to enhance immunological clearance of the bacilli, but necessitates a deeper understanding of the immune mechanisms associated with protection against Mycobacterium tuberculosis infection. Here, we examined the various dendritic cells (DC) subsets present in the lung and draining lymph nodes (LN) from mice intra-tracheally infected with M. tuberculosis. We showed that although limited in number, pulmonary CD103+ DCs appeared to be involved in the initial transport of mycobacteria to the draining mediastinal LN and subsequent activation of T cells. Using CLEC9A-DTR transgenic mice enabling the inducible depletion of CD103+ DCs, we established that this DC subset contributes to the control of mycobacterial burden and plays a role in the early activation of T cells, in particular CD8+ T cells. Our findings thus support a previously unidentified role for pulmonary CD103+ DCs in the rapid mobilization of mycobacteria from the lungs to the draining LN soon after exposure to M. tuberculosis, which is a critical step for the development of the host adaptive immune response.
Collapse
Affiliation(s)
- Vanessa Hui Qi Koh
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Immunology Programme, Life Sciences Institute, NUS, Singapore
| | - See Liang Ng
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Michelle Lay Teng Ang
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Immunology Programme, Life Sciences Institute, NUS, Singapore
| | - Wenwei Lin
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Immunology Programme, Life Sciences Institute, NUS, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore.
| | - Sylvie Alonso
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Immunology Programme, Life Sciences Institute, NUS, Singapore.
| |
Collapse
|
31
|
Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2016; 17:30-48. [PMID: 27890914 DOI: 10.1038/nri.2016.116] [Citation(s) in RCA: 584] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are potent and versatile antigen-presenting cells, and their ability to migrate is key for the initiation of protective pro-inflammatory as well as tolerogenic immune responses. Recent comprehensive studies have highlighted the importance of DC migration in the maintenance of immune surveillance and tissue homeostasis, and also in the pathogenesis of a range of diseases. In this Review, we summarize the anatomical, cellular and molecular factors that regulate the migration of different DC subsets in health and disease. In particular, we focus on new insights concerning the role of migratory DCs in the pathogenesis of diseases of the skin, intestine, lung, and brain, as well as in autoimmunity and atherosclerosis.
Collapse
Affiliation(s)
- Tim Worbs
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Swantje I Hammerschmidt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
32
|
Abstract
The respiratory immune response consists of multiple tiers of cellular responses that are engaged in a sequential manner in order to control infections. The stepwise engagement of effector functions with progressively increasing host fitness costs limits tissue damage. In addition, specific mechanisms are in place to promote disease tolerance in response to respiratory infections. Environmental factors, obesity and the ageing process can alter the efficiency and regulation of this tiered response, increasing pathology and mortality as a result. In this Review, we describe the cell types that coordinate pathogen clearance and tissue repair through the serial secretion of cytokines, and discuss how the environment and comorbidity influence this response.
Collapse
|
33
|
Deficiency of the Complement Component 3 but Not Factor B Aggravates Staphylococcus aureus Septic Arthritis in Mice. Infect Immun 2016; 84:930-939. [PMID: 26787717 PMCID: PMC4807474 DOI: 10.1128/iai.01520-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/11/2016] [Indexed: 12/23/2022] Open
Abstract
The complement system plays an essential role in the innate immune response and protection against bacterial infections. However, detailed knowledge regarding the role of complement in Staphylococcus aureus septic arthritis is still largely missing. In this study, we elucidated the roles of selected complement proteins in S. aureus septic arthritis. Mice lacking the complement component 3 (C3(-/-)), complement factor B (fB(-/-)), and receptor for C3-derived anaphylatoxin C3a (C3aR(-/-)) and wild-type (WT) control mice were intravenously or intra-articularly inoculated with S. aureus strain Newman. The clinical course of septic arthritis, as well as histopathological and radiological changes in joints, was assessed. After intravenous inoculation, arthritis severity and frequency were significantly higher in C3(-/-)mice than in WT controls, whereas fB(-/-)mice displayed intermediate arthritis severity and frequency. This was in accordance with both histopathological and radiological findings. C3, but not fB, deficiency was associated with greater weight loss, more frequent kidney abscesses, and higher bacterial burden in kidneys. S. aureus opsonized with C3(-/-)sera displayed decreased uptake by mouse peritoneal macrophages compared with bacteria opsonized with WT or fB(-/-)sera. C3aR deficiency had no effect on the course of hematogenous S. aureus septic arthritis. We conclude that C3 deficiency increases susceptibility to hematogenous S. aureus septic arthritis and impairs host bacterial clearance, conceivably due to diminished opsonization and phagocytosis of S. aureus.
Collapse
|
34
|
Givi ME, Folkerts G, Wagenaar GTM, Redegeld FA, Mortaz E. Cigarette smoke differentially modulates dendritic cell maturation and function in time. Respir Res 2015; 16:131. [PMID: 26498483 PMCID: PMC4619524 DOI: 10.1186/s12931-015-0291-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 10/13/2015] [Indexed: 12/26/2022] Open
Abstract
Background Dendritic cells (DCs) as professional antigen presenting cells (APCs) play a critical role in the regulation of host immune responses. DCs evolve from immature, antigen-capturing cells, to mature antigen-presenting cells. The relative contribution of DCs to cigarette smoke-induced inflammation is not well documented. In the current study, we investigated a modulatory effect of cigarette smoke extract (CSE) on differentiation, maturation and function of DCs. Methods Primary murine DCs were grown from bone marrow cells with GM-CSF. Development of DC was analyzed by expression of CD11c, MHCII, CD86, CD40 and CD83 using flow cytometry. Murine DC’s and human L428 cells were co-cultured with CSE for various periods of time. Functional activity was analyzed by measuring FITC-dextran uptake, cytokine production and the ability to stimulate T cell activation in a mixed lymphocyte reaction. Results Our results show that short-term CSE stimulation (~24 h) influence the maturation status of newly differentiated and immature DCs towards more mature cells as revealed by upregulation of MHCII, CD83, CD86, CD40, reduction in antigen up-take capacity and enhanced secretion of pro-inflammatory (IL-12, IL-6 and TNF-α) cytokines. Interestingly, long-term CSE exposure, time- and concentration-dependently, suppressed the development of functional DCs. This suppression was demonstrated by a decline in CD11c/MHCII, CD83, CD86 and CD40 expression, the production of cytokines and ability to stimulate T lymphocytes. Moreover, CSE significantly suppressed the endocytosis function of mouse DCs which was not due to diminished DC viability. Similar to mouse DCs, long-term co-culturing of the human L428 DC cell line with CSE time-dependently suppressed the expression of CD54. Conclusions The present study provides evidence that CSE modulates DC-mediated immune responses via affecting both the function and maturation of DCs. The suppressive effects of cigarette smoke on DC function might lead to impaired immune responses to various infections. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0291-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masoumeh Ezzati Givi
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO BOX 80082, 3508, TB, Utrecht, The Netherlands.,Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Shahid Chamran University, Ahvaz, Iran
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO BOX 80082, 3508, TB, Utrecht, The Netherlands
| | - Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO BOX 80082, 3508, TB, Utrecht, The Netherlands.
| | - Esmaeil Mortaz
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO BOX 80082, 3508, TB, Utrecht, The Netherlands.,Chronic Respiratory Diseases Research Center and National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
35
|
Yuan X, Shan M, You R, Frazier MV, Hong MJ, Wetsel RA, Drouin S, Seryshev A, MD LZS, Cornwell L, Rossen RD, Corry DB, Kheradmand F. Activation of C3a receptor is required in cigarette smoke-mediated emphysema. Mucosal Immunol 2015; 8:874-85. [PMID: 25465103 PMCID: PMC4454642 DOI: 10.1038/mi.2014.118] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/23/2014] [Indexed: 02/04/2023]
Abstract
Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ming Shan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ran You
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Michael V. Frazier
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Monica Jeongsoo Hong
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Rick A. Wetsel
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Scott Drouin
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Alexander Seryshev
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Li-zhen Song MD
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | | | - Roger D Rossen
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - David B. Corry
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - Farrah Kheradmand
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| |
Collapse
|
36
|
Fukasaka M, Asari D, Kiyotoh E, Okazaki A, Gomi Y, Tanimoto T, Takeuchi O, Akira S, Hori M. A Lipopolysaccharide from Pantoea Agglomerans Is a Promising Adjuvant for Sublingual Vaccines to Induce Systemic and Mucosal Immune Responses in Mice via TLR4 Pathway. PLoS One 2015; 10:e0126849. [PMID: 25978818 PMCID: PMC4433252 DOI: 10.1371/journal.pone.0126849] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/08/2015] [Indexed: 12/30/2022] Open
Abstract
A lipopolysaccharide from Pantoea agglomerans (LPSpa) has been applied to various fields for human use as a Toll-like receptor 4 ligand and its safety has been confirmed. Here, we showed for the first time the application of LPSpa as an effective mucosal adjuvant for activating vaccine-induced antigen specific immune responses. Mice sublingually immunized with influenza vaccine (HA split vaccine) with LPSpa induced both HA-specific IgG (systemic) and IgA (mucosal) antibody responses, which led to a significant increase in survival rate against lethal influenza virus challenge compared with subcutaneous vaccination. After sublingual administration of ovalbumin with LPSpa, ovalbumin-specific mucosal IgA responses were induced at both mucosal surfaces close to the immunized site and at remote mucosal surfaces. Sublingual administration of LPSpa evoked local antigen-uptake by dendritic cells in cervical lymph nodes. LPSpa induced cytokine production and the maturation and proliferation of innate immune cells via Toll-like receptor 4 in dendritic cells. Collectively, these results suggest that LPSpa can be used as an effective mucosal adjuvant to stimulate and activate local innate immune cells to improve and enhance mucosal vaccine potency against various pathogens.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Administration, Sublingual
- Animals
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Immunity, Mucosal/drug effects
- Immunity, Mucosal/immunology
- Immunoglobulin A/immunology
- Immunoglobulin G/immunology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Lipopolysaccharides/administration & dosage
- Lipopolysaccharides/immunology
- Lipopolysaccharides/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Pantoea/immunology
- Toll-Like Receptor 4/physiology
Collapse
Affiliation(s)
- Masahiro Fukasaka
- Life Science Research Center, Corporate Research & Development Division, Nitto Denko Corporation, Ibaraki, Osaka, Japan
- * E-mail: (MF); (MH)
| | - Daisuke Asari
- Life Science Research Center, Corporate Research & Development Division, Nitto Denko Corporation, Ibaraki, Osaka, Japan
| | - Eiji Kiyotoh
- Life Science Research Center, Corporate Research & Development Division, Nitto Denko Corporation, Ibaraki, Osaka, Japan
| | - Arimichi Okazaki
- Life Science Research Center, Corporate Research & Development Division, Nitto Denko Corporation, Ibaraki, Osaka, Japan
| | - Yasuyuki Gomi
- Research and Production Technology Department, The Research Foundation for Microbial Diseases of Osaka University, Kanonji, Kagawa, Japan
| | - Takeshi Tanimoto
- Research and Production Technology Department, The Research Foundation for Microbial Diseases of Osaka University, Kanonji, Kagawa, Japan
| | - Osamu Takeuchi
- Laboratory of Infection and Prevention, Institute for Virus Research, Kyoto University, Shogoin Kawara-cho, Sakyo-ku, Kyoto, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Mitsuhiko Hori
- Life Science Research Center, Corporate Research & Development Division, Nitto Denko Corporation, Ibaraki, Osaka, Japan
- * E-mail: (MF); (MH)
| |
Collapse
|
37
|
Schlitzer A, McGovern N, Ginhoux F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin Cell Dev Biol 2015; 41:9-22. [DOI: 10.1016/j.semcdb.2015.03.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 12/23/2022]
|
38
|
Kopf M, Schneider C, Nobs SP. The development and function of lung-resident macrophages and dendritic cells. Nat Immunol 2015; 16:36-44. [PMID: 25521683 DOI: 10.1038/ni.3052] [Citation(s) in RCA: 402] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/10/2014] [Indexed: 12/12/2022]
Abstract
Gas exchange is the vital function of the lungs. It occurs in the alveoli, where oxygen and carbon dioxide diffuse across the alveolar epithelium and the capillary endothelium surrounding the alveoli, separated only by a fused basement membrane 0.2-0.5 μm in thickness. This tenuous barrier is exposed to dangerous or innocuous particles, toxins, allergens and infectious agents inhaled with the air or carried in the blood. The lung immune system has evolved to ward off pathogens and restrain inflammation-mediated damage to maintain gas exchange. Lung-resident macrophages and dendritic cells are located in close proximity to the epithelial surface of the respiratory system and the capillaries to sample and examine the air-borne and blood-borne material. In communication with alveolar epithelial cells, they set the threshold and the quality of the immune response.
Collapse
Affiliation(s)
- Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Christoph Schneider
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Samuel P Nobs
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Karsten CM, Laumonnier Y, Eurich B, Ender F, Bröker K, Roy S, Czabanska A, Vollbrandt T, Figge J, Köhl J. Monitoring and cell-specific deletion of C5aR1 using a novel floxed GFP-C5aR1 reporter knock-in mouse. THE JOURNAL OF IMMUNOLOGY 2015; 194:1841-55. [PMID: 25589074 DOI: 10.4049/jimmunol.1401401] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many of the biological properties of C5a are mediated through activation of its receptor (C5aR1), the expression of which has been demonstrated convincingly on myeloid cells, such as neutrophils, monocytes, and macrophages. In contrast, conflicting results exist regarding C5aR1 expression in dendritic cells (DCs) and lymphoid lineage cells. In this article, we report the generation of a floxed GFP-C5aR1 reporter knock-in mouse. Using this mouse strain, we confirmed strong C5aR1 expression in neutrophils from bone marrow, blood, lung, and spleen, as well as in peritoneal macrophages. Further, we show C5aR1 expression in lung eosinophils, lung- and lamina propria-resident and alveolar macrophages, bone marrow-derived DCs, and lung-resident CD11b(+) and monocyte-derived DCs, whereas intestinal and pulmonary CD103(+) DCs stained negative. Also, some splenic NKT cells expressed GFP, whereas naive NK cells and B2 cells lacked GFP expression. Finally, we did not observe any C5aR1 expression in naive or activated CD4(+) Th cells in vitro or in vivo. Mating the floxed GFP-C5aR1 mouse strain with LysMCre mice, we were able to specifically delete C5aR1 in neutrophils and macrophages, whereas C5aR1 expression was retained in DCs. In summary, our findings suggest that C5aR1 expression in mice is largely restricted to cells of the myeloid lineage. The novel floxed C5aR1 reporter knock-in mouse will prove useful to track C5aR1 expression in experimental models of acute and chronic inflammation and to conditionally delete C5aR1 in immune cells.
Collapse
Affiliation(s)
- Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Benjamin Eurich
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Katharina Bröker
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Sreeja Roy
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Anna Czabanska
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | | | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| |
Collapse
|
40
|
Lambrecht BN, Neyt K, van Helden MJ. The Mucosal Immune Response to Respiratory Viruses. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
41
|
|
42
|
Purnama C, Ng SL, Tetlak P, Setiagani YA, Kandasamy M, Baalasubramanian S, Karjalainen K, Ruedl C. Transient ablation of alveolar macrophages leads to massive pathology of influenza infection without affecting cellular adaptive immunity. Eur J Immunol 2014; 44:2003-12. [PMID: 24687623 DOI: 10.1002/eji.201344359] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/11/2014] [Accepted: 03/26/2014] [Indexed: 11/09/2022]
Abstract
Alveolar macrophages (AMs), localized at the pulmonary air-tissue interface, are one of the first lines of defense that interact with inhaled airborne pathogens such as influenza viruses. By using a new CD169-DTR transgenic mouse strain we demonstrate that specific and highly controlled in vivo ablation of this myeloid cell subset leads to severe impairment of the innate, but not adaptive, immune responses and critically affects the progression of the disease. In fact, AM-ablated mice, infected with a normally sublethal dose of PR8 influenza virus, showed dramatically increased virus load in the lungs, severe airway inflammation, pulmonary edema and vascular leakage, which caused the death of the infected animals. Our data highlight the possibilities for new therapeutic strategies focusing on modulation of AMs, which may efficiently boost innate responses to influenza infections.
Collapse
Affiliation(s)
- Christina Purnama
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Neyt K, Lambrecht BN. The role of lung dendritic cell subsets in immunity to respiratory viruses. Immunol Rev 2014; 255:57-67. [PMID: 23947347 DOI: 10.1111/imr.12100] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Viral infections are a common cause of acute respiratory disease. The clinical course of infection and symptoms depend on the viral strain, the health status of the host, and the immunological status of the host. Dendritic cells (DCs) play a crucial role in recognizing and presenting viral antigens and in inducing adaptive immune responses that clear the virus. Because the lung is continuously exposed to the air, the lung is equipped with an elaborate network of DCs to sense incoming foreign pathogens. Increasing knowledge on DC biology has informed us that DCs are not a single cell type. In the steady state lung, three DC subsets can be defined: CD11b(+) or CD103(+) conventional DCs and plasmacytoid DCs. Upon inflammation, inflammatory monocyte-derived DCs are recruited to the lung. It is only recently that tools became available to allow DC subsets to be clearly studied. This review focuses on the activation of DCs and the function of lung DCs in the context of respiratory virus infection and highlights some cautionary points for interpreting older experiments.
Collapse
Affiliation(s)
- Katrijn Neyt
- VIB Inflammation Research Center, Laboratory of Immunoregulation, Ghent, Belgium
| | | |
Collapse
|
44
|
Jin GB, Winans B, Martin KC, Paige Lawrence B. New insights into the role of the aryl hydrocarbon receptor in the function of CD11c⁺ cells during respiratory viral infection. Eur J Immunol 2014; 44:1685-1698. [PMID: 24519489 DOI: 10.1002/eji.201343980] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 01/28/2014] [Accepted: 02/06/2014] [Indexed: 01/06/2023]
Abstract
The aryl hydrocarbon receptor (AHR) has garnered considerable attention as a modulator of CD4(+) cell lineage development and function. It also regulates antiviral CD8(+) T-cell responses, but via indirect mechanisms that have yet to be determined. Here, we show that during acute influenza virus infection, AHR activation skews dendritic-cell (DC) subsets in the lung-draining lymph nodes, such that there are fewer conventional CD103(+) DCs and CD11b(+) DCs. Sorting DC subsets reveals AHR activation reduces immunostimulatory function of CD103(+) DCs in the mediastinal lymph nodes, and decreases their frequency in the lung. DNA-binding domain Ahr mutants demonstrate that alterations in DC subsets require the ligand-activated AHR to contain its inherent DNA-binding domain. To evaluate the intrinsic role of AHR in DCs, conditional knockouts were created using Cre-LoxP technology, which revealed that AHR in CD11c(+) cells plays a key role in controlling the acquisition of effector CD8(+) T cells in the infected lung. However, AHR within other leukocyte lineages contributes to diminished naïve CD8(+) T-cell activation in the draining lymphoid nodes. These findings indicate DCs are among the direct targets of AHR ligands in vivo, and AHR signaling modifies host responses to a common respiratory pathogen by affecting the complex interplay of multiple cell types.
Collapse
Affiliation(s)
- Guang-Bi Jin
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Bethany Winans
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Kyle C Martin
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.,Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
45
|
Abstract
Influenza virus infection induces a potent initial innate immune response, which serves to limit the extent of viral replication and virus spread. However, efficient (and eventual) viral clearance within the respiratory tract requires the subsequent activation, rapid proliferation, recruitment, and expression of effector activities by the adaptive immune system, consisting of antibody producing B cells and influenza-specific T lymphocytes with diverse functions. The ensuing effector activities of these T lymphocytes ultimately determine (along with antibodies) the capacity of the host to eliminate the viruses and the extent of tissue damage. In this review, we describe this effector T cell response to influenza virus infection. Based on information largely obtained in experimental settings (i.e., murine models), we will illustrate the factors regulating the induction of adaptive immune T cell responses to influenza, the effector activities displayed by these activated T cells, the mechanisms underlying the expression of these effector mechanisms, and the control of the activation/differentiation of these T cells, in situ, in the infected lungs.
Collapse
|
46
|
Prabhu N, Ho AW, Wong KHS, Hutchinson PE, Chua YL, Kandasamy M, Lee DCP, Sivasankar B, Kemeny DM. Gamma interferon regulates contraction of the influenza virus-specific CD8 T cell response and limits the size of the memory population. J Virol 2013; 87:12510-22. [PMID: 24027334 PMCID: PMC3838152 DOI: 10.1128/jvi.01776-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/06/2013] [Indexed: 11/20/2022] Open
Abstract
The factors that regulate the contraction of the CD8 T cell response and the magnitude of the memory cell population against localized mucosal infections such as influenza are important for generation of efficient vaccines but are currently undefined. In this study, we used a mouse model of influenza to demonstrate that the absence of gamma interferon (IFN-γ) or IFN-γ receptor 1 (IFN-γR1) leads to aberrant contraction of antigen-specific CD8 T cell responses. The increased accumulation of the effector CD8 T cell population was independent of viral load. Reduced contraction was associated with an increased fraction of CD8 T cells expressing the interleukin-7 receptor (IL-7R) at the peak of the response, resulting in enhanced numbers of memory/memory precursor cells in IFN-γ(-/-) and IFN-γR(-/-) compared to wild-type (WT) mice. Blockade of IL-7 within the lungs of IFN-γ(-/-) mice restored the contraction of influenza virus-specific CD8 T cells, indicating that IL-7R is important for survival and is not simply a consequence of the lack of IFN-γ signaling. Finally, enhanced CD8 T cell recall responses and accelerated viral clearance were observed in the IFN-γ(-/-) and IFN-γR(-/-) mice after rechallenge with a heterologous strain of influenza virus, confirming that higher frequencies of memory precursors are formed in the absence of IFN-γ signaling. In summary, we have identified IFN-γ as an important regulator of localized viral immunity that promotes the contraction of antigen-specific CD8 T cells and inhibits memory precursor formation, thereby limiting the size of the memory cell population after an influenza virus infection.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Female
- Humans
- Immunologic Memory
- Influenza A virus/genetics
- Influenza A virus/immunology
- Influenza, Human/genetics
- Influenza, Human/immunology
- Influenza, Human/virology
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Lymphocyte Count
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/immunology
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/immunology
- Species Specificity
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Nayana Prabhu
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
- NUS Graduate School for Integrated Sciences and Engineering, National University of Singapore, Singapore
| | - Adrian W. Ho
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Kenneth H. S. Wong
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Paul Edward Hutchinson
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Yen Leong Chua
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Matheswaran Kandasamy
- Infection & Immunity Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Debbie C. P. Lee
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Baalasubramanian Sivasankar
- Infection & Immunity Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - David Michael Kemeny
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
- NUS Graduate School for Integrated Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
47
|
Dutow P, Fehlhaber B, Bode J, Laudeley R, Rheinheimer C, Glage S, Wetsel RA, Pabst O, Klos A. The complement C3a receptor is critical in defense against Chlamydia psittaci in mouse lung infection and required for antibody and optimal T cell response. J Infect Dis 2013; 209:1269-78. [PMID: 24273177 DOI: 10.1093/infdis/jit640] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The complement system protects against extracellular pathogens and links innate and adaptive immunity. In this study, we investigated the anaphylatoxin C3a receptor (C3aR) in Chlamydia psittaci lung infection and elucidated C3a-dependent adaptive immune mechanisms. METHODS Survival, body weight, and clinical score were monitored in primary mouse infection and after serum transfer. Bacterial load, histology, cellular distribution, cytokines, antibodies, and lymphocytes were analyzed. RESULTS C3aR(-/-) mice showed prolonged pneumonia with decreased survival, lower weight, and higher clinical score. Compared to wild-type mice bacterial clearance was impaired, and inflammatory parameters were increased. In lung-draining lymph nodes of C3aR(-/-) mice the total number of B cells, CD4(+) T cells, and Chlamydia-specific IFN-γ(+) (CD4(+) or CD8(+)) cells was reduced upon infection, and the mice were incapable of Chlamydia-specific immunoglobulin M or immunoglobulin G production. Performed before infection, transfer of hyperimmune serum prolonged survival of C3aR(-/-) mice. CONCLUSIONS C3a and its receptor are critical for defense against C. psittaci in mouse lung infection. In this model, C3a acts via its receptor as immune modulator. Enhancement of specific B and T cell responses upon infection with an intracellular bacterium were identified as hitherto unknown features of C3a/C3aR. These new functions might be of general immunological importance.
Collapse
Affiliation(s)
- Pavel Dutow
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH)
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yoo JK, Kim TS, Hufford MM, Braciale TJ. Viral infection of the lung: host response and sequelae. J Allergy Clin Immunol 2013; 132:1263-76; quiz 1277. [PMID: 23915713 PMCID: PMC3844062 DOI: 10.1016/j.jaci.2013.06.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/21/2013] [Accepted: 06/05/2013] [Indexed: 02/06/2023]
Abstract
Because of its essential role in gas exchange and oxygen delivery, the lung has evolved a variety of strategies to control inflammation and maintain homeostasis. Invasion of the lung by pathogens (and in some instances exposure to certain noninfectious particulates) disrupts this equilibrium and triggers a cascade of events aimed at preventing or limiting colonization (and more importantly infection) by pathogenic microorganisms. In this review we focus on viral infection of the lung and summarize recent advances in our understanding of the triggering of innate and adaptive immune responses to viral respiratory tract infection, mechanisms of viral clearance, and the well-recognized consequences of acute viral infection complicating underlying lung diseases, such as asthma.
Collapse
Affiliation(s)
- Jae-Kwang Yoo
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
| | - Taeg S. Kim
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Pathology and Molecular Medicine, University of Virginia, Charlottesville, Va
| | - Matthew M. Hufford
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Va
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Va
- Department of Pathology and Molecular Medicine, University of Virginia, Charlottesville, Va
- Corresponding author: Thomas J. Braciale, MD, PhD, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908.
| |
Collapse
|
49
|
Schmudde I, Laumonnier Y, Köhl J. Anaphylatoxins coordinate innate and adaptive immune responses in allergic asthma. Semin Immunol 2013; 25:2-11. [PMID: 23694705 DOI: 10.1016/j.smim.2013.04.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/22/2013] [Indexed: 12/28/2022]
Abstract
Allergic asthma is a chronic disease of the airways in which maladaptive Th2 and Th17 immune responses drive airway hyperresponsiveness (AHR), eosinophilic and neutrophilic airway inflammation and mucus overproduction. Airway epithelial and pulmonary vascular endothelial cells in concert with different resident and monocyte-derived dendritic cells (DC) play critical roles in allergen sensing and consecutive activation of TH cells and their differentiation toward TH2 and TH17 effector or regulatory T cells (Treg). Further, myeloid-derived regulatory cells (MDRC) act on TH cells and either suppress or enhance their activation. The complement-derived anaphylatoxins (AT) C3a and C5a are generated during initial antigen encounter and regulate the development of maladaptive immunity at allergen sensitization. Here, we will review the complex role of ATs in activation and modulation of different DC populations, MDRCs and CD4⁺ TH cells. We will also discuss the potential impact of ATs on the regulation of the pulmonary stromal compartment as an important means to regulate DC functions.
Collapse
Affiliation(s)
- Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Germany
| | | | | |
Collapse
|