1
|
Alibrandi S, Clemens A, Chun N. Complement and T cell activation in transplantation. Transplant Rev (Orlando) 2025; 39:100898. [PMID: 39615218 PMCID: PMC11710966 DOI: 10.1016/j.trre.2024.100898] [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: 08/14/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
The complement system plays a critical role in modulating adaptive T cell responses. Coordination of the proinflammatory signaling cascade and complement regulators permits efficient T cell priming and survival, while minimizing off-target damage to healthy host cells. In the context of transplantation, anti-donor T cell immunity remains a barrier to long term graft health and complement-targeted therapies have shown the potential to significantly improve patient outcomes. Here we will review our current understanding of complement-mediated T cell function and how these findings may be harnessed in organ transplantation.
Collapse
Affiliation(s)
- Sara Alibrandi
- Translational Transplant Research Center and Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, NY, NY, USA; Department of Medicine and Surgery, University of Parma, Parma, Italy; Nephrology Unit, University Hospital of Parma, Parma, Italy
| | - Angela Clemens
- Translational Transplant Research Center and Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Nicholas Chun
- Translational Transplant Research Center and Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, NY, NY, USA.
| |
Collapse
|
2
|
Jo H, Jeoung J, Kim W, Jeoung D. Regulating Immune Responses Induced by PEGylated Messenger RNA-Lipid Nanoparticle Vaccine. Vaccines (Basel) 2024; 13:14. [PMID: 39852793 PMCID: PMC11768904 DOI: 10.3390/vaccines13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Messenger RNA (mRNA)-based therapeutics have shown remarkable progress in the treatment and prevention of diseases. Lipid nanoparticles (LNPs) have shown great successes in delivering mRNAs. After an mRNA-LNP vaccine enters a cell via an endosome, mRNA is translated into an antigen, which can activate adaptive immunity. mRNAs can bind to various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), and increase the production of inflammatory cytokines. This review summarizes mechanisms of innate immunity induced by mRNAs. Polyethylene glycol (PEG) has been employed as a component of the mRNA-LNP vaccine. PEGylated nanoparticles display enhanced stability by preventing aggregation of particles. However, PEGylation can cause adverse reactions, including blood clearance (ABC) of nanoparticles via complement activation and anaphylaxis. Mechanisms of PEG-induced ABC phenomenon and anaphylaxis are presented and discussed. There have been studies aimed at reducing immune responses associated with PEG to make safe and effective vaccines. Effects of modifying or replacing PEG in reducing immune responses associated with PEGylated nanoparticles are also discussed. Modifying mRNA can induce immune tolerance, which can prevent hypersensitivity reactions induced by PEGylated mRNA-LNP vaccines. Current progress of immune tolerance induction in association with mRNA-LNP is also summarized. This review might be helpful for developing safe and effective PEGylated mRNA-LNP vaccines.
Collapse
Affiliation(s)
| | | | | | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (H.J.); (J.J.); (W.K.)
| |
Collapse
|
3
|
Song J, Huan X, Chen Y, Luo Y, Zhong H, Wang Y, Yang L, Xi C, Yang Y, Xi J, Zheng J, Wu Z, Zhao C, Luo S. The safety and efficacy profile of eculizumab in myasthenic crisis: a prospective small case series. Ther Adv Neurol Disord 2024; 17:17562864241261602. [PMID: 39072008 PMCID: PMC11282533 DOI: 10.1177/17562864241261602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024] Open
Abstract
Eculizumab has improved recovery from ventilatory support in myasthenic crisis (MC) cases. However, the safety and efficacy profiles from prospective studies are still lacking. This study aimed to explore eculizumab's safety and efficacy in a prospective case series of patients with refractory MC. We followed a series of anti-acetylcholine receptor (AChR) antibody-positive myasthenia gravis (MG) patients who received eculizumab as an add-on therapy for 12 weeks during MC to facilitate the weaning process and reduced disease activity. Serum anti-AChR antibodies and peripheral immune molecules associated with the complement pathway were evaluated before and after eculizumab administration. Compared to the baseline Myasthenia Gravis Foundation of America (MGFA)-quantitative MG test (QMG) scores (22.25 ± 4.92) and MG-activities of daily living (MG-ADL; 18.25 ± 2.5) scores at crisis, improvements were observed from 4 weeks (14.5 ± 10.47 and 7.5 ± 7.59, respectively) through 12 weeks (7.5 ± 5.74 and 2.25 ± 3.86, respectively) post-treatment. Muscle strength consistently improved across ocular, bulbar, respiratory, and limb/gross domain groups. One patient died of cardiac failure at 16 weeks. Three cases remained in remission at 24 weeks, with a mean QMG score of 2.67 ± 2.89 and ADL score of 0.33 ± 0.58. No significant side effects were reported. Serum CH50 and soluble C5b-9 levels significantly declined, while there were no significant changes in serum anti-AChR antibody levels, C1q, C5a levels, or peripheral lymphocyte proportions. Eculizumab was well tolerated and showed efficacy in this case series. Large prospective cohort studies with extended follow-up periods are needed to further explore the safety and efficacy profile in real-world practice.
Collapse
Affiliation(s)
- Jie Song
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Xiao Huan
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Yuanyi Chen
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Yeting Luo
- Department of Neurology, Ganzhou People’s Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Huahua Zhong
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Yuan Wang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Yang
- Department of Neurosurgery & Neurocritical care, Huashan Hospital Fudan University, Shanghai, China
| | - Caihua Xi
- Department of Neurosurgery & Neurocritical care, Huashan Hospital Fudan University, Shanghai, China
| | - Yu Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianying Xi
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Jianming Zheng
- Department of Infectious Diseases, Huashan Hospital, National Medical Center for Infectious Diseases, Fudan University, Shanghai, China
| | - Zongtai Wu
- Faculty of Biology, University of Cambridge, Cambridge, UK
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
4
|
Laumonnier Y, Korkmaz RÜ, Nowacka AA, Köhl J. Complement-mediated immune mechanisms in allergy. Eur J Immunol 2023; 53:e2249979. [PMID: 37381711 DOI: 10.1002/eji.202249979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 06/30/2023]
Abstract
Allergic conditions are associated with canonical and noncanonical activation of the complement system leading to the release of several bioactive mediators with inflammatory and immunoregulatory properties that regulate the immune response in response to allergens during the sensitization and/or the effector phase of allergic diseases. Further, immune sensors of complement and regulator proteins of the cascade impact on the development of allergies. These bioactive mediators comprise the small and large cleavage fragments of C3 and C5. Here, we provide an update on the multiple roles of immune sensors, regulators, and bioactive mediators of complement in allergic airway diseases, food allergies, and anaphylaxis. A particular emphasis is on the anaphylatoxins C3a and C5a and their receptors, which are expressed on many of the effector cells in allergy such as mast cells, eosinophils, basophils, macrophages, and neutrophils. Also, we will discuss the multiple pathways, by which the anaphylatoxins initiate and control the development of maladaptive type 2 immunity including their impact on innate lymphoid cell recruitment and activation. Finally, we briefly comment on the potential to therapeutically target the complement system in different allergic conditions.
Collapse
Affiliation(s)
- Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Institute for Nutritional Medicine, University of Lübeck, Lübeck, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Rabia Ülkü Korkmaz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Alicja A Nowacka
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, USA
| |
Collapse
|
5
|
Nascimento CM, Casaro MC, Perez ER, Ribeiro WR, Mayer MPA, Ishikawa KH, Lino-dos-Santos-Franco A, Pereira JNB, Ferreira CM. Experimental allergic airway inflammation impacts gut homeostasis in mice. Heliyon 2023; 9:e16429. [PMID: 37484240 PMCID: PMC10360590 DOI: 10.1016/j.heliyon.2023.e16429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/30/2023] [Accepted: 05/16/2023] [Indexed: 07/25/2023] Open
Abstract
Background /Aims: Epidemiological data show that there is an important relationship between respiratory and intestinal diseases. To improve our understanding on the interconnectedness between the lung and intestinal mucosa and the overlap between respiratory and intestinal diseases, our aim was to investigate the influence of ovalbumin (OVA)-induced allergic airway inflammation on gut homeostasis. Methods A/J mice were sensitized and challenged with OVA. The animals were euthanized 24 h after the last challenge, lung inflammation was determined by evaluating cells in Bronchoalveolar lavage fluid, serum anti-OVA IgG titers and colon morphology, inflammation and integrity of the intestinal mucosa were investigated. IL-4 and IL-13 levels and myeloperoxidase activity were determined in the colon samples. The expression of genes involved in inflammation and mucin production at the gut mucosa was also evaluated. Results OVA challenge resulted not only in lung inflammation but also in macroscopic alterations in the gut such as colon shortening, increased myeloperoxidase activity and loss of integrity in the colonic mucosal. Neutral mucin intensity was lower in the OVA group, which was followed by down-regulation of transcription of ATOH1 and up-regulation of TJP1 and MUC2. In addition, the OVA group had higher levels of IL-13 and IL-4 in the colon. Ova-specific IgG1 and OVA-specific IgG2a titers were higher in the serum of the OVA group than in controls. Conclusions Our data using the OVA experimental model suggested that challenges in the respiratory system may result not only in allergic airway inflammation but also in the loss of gut homeostasis.
Collapse
Affiliation(s)
- Carolina Martins Nascimento
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Mateus Campos Casaro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Evelyn Roxana Perez
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Willian Rodrigues Ribeiro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Karin Hitomi Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | | | - Caroline Marcantonio Ferreira
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| |
Collapse
|
6
|
Messelodi D, Giuliani C, Cipriani F, Armuzzi S, di Palmo E, Garagnani P, Bertelli L, Astolfi A, Luiselli D, Ricci G, Pession A. C5 and SRGAP3 Polymorphisms Are Linked to Paediatric Allergic Asthma in the Italian Population. Genes (Basel) 2022; 13:214. [PMID: 35205259 PMCID: PMC8871526 DOI: 10.3390/genes13020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Asthma is a complex and heterogeneous disease, caused by the interaction between genetic and environmental factors with a predominant allergic background in children. The role of specific genes in asthmatic bronchial reactivity is still not clear, probably because of the many common pathways shared with other allergic disorders. This study is focused on 11 SNPs possibly related to asthma that were previously identified in a GWAS study. The genetic variability of these SNPs has been analysed in a population of 773 Italian healthy controls, and the presence of an association between the polymorphisms and the asthma onset was evaluated performing genotyping analysis on 108 children affected with asthma compared with the controls. Moreover, a pool of 171 patients with only allergic rhinoconjunctivitis has been included in the case-control analysis. The comparison of allele frequencies in asthmatic patients versus healthy controls identified two SNPs-rs1162394 (p = 0.019) and rs25681 (p = 0.044)-associated with the asthmatic condition, which were not differentially distributed in the rhinoconjunctivitis group. The rs25681 SNP, together with three other SNPs, also resulted in not being homogenously distributed in the Italian population. The significantly higher frequency of the rs25681 and rs1162394 SNPs (located, respectively, in the C5 and SRGAP3 genes) in the asthmatic population suggests an involvement of these genes in the asthmatic context, playing a role in increasing the inflammatory condition that may influence asthma onset and clinical course.
Collapse
Affiliation(s)
- Daria Messelodi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Cristina Giuliani
- Laboratory of Molecular Anthropology, Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Francesca Cipriani
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (E.d.P.); (L.B.); (A.P.)
| | - Silvia Armuzzi
- Institute of Hematology “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy; (P.G.); (A.A.)
| | - Emanuela di Palmo
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (E.d.P.); (L.B.); (A.P.)
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy; (P.G.); (A.A.)
| | - Luca Bertelli
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (E.d.P.); (L.B.); (A.P.)
| | - Annalisa Astolfi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy; (P.G.); (A.A.)
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Donata Luiselli
- Laboratory of Ancient DNA (aDNALab), Department of Cultural Heritage (DBC), Ravenna Campus, University of Bologna, 40126 Bologna, Italy;
| | - Giampaolo Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (E.d.P.); (L.B.); (A.P.)
| |
Collapse
|
7
|
Freiwald T, Afzali B. Renal diseases and the role of complement: Linking complement to immune effector pathways and therapeutics. Adv Immunol 2021; 152:1-81. [PMID: 34844708 PMCID: PMC8905641 DOI: 10.1016/bs.ai.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The complement system is an ancient and phylogenetically conserved key danger sensing system that is critical for host defense against pathogens. Activation of the complement system is a vital component of innate immunity required for the detection and removal of pathogens. It is also a central orchestrator of adaptive immune responses and a constituent of normal tissue homeostasis. Once complement activation occurs, this system deposits indiscriminately on any cell surface in the vicinity and has the potential to cause unwanted and excessive tissue injury. Deposition of complement components is recognized as a hallmark of a variety of kidney diseases, where it is indeed associated with damage to the self. The provenance and the pathophysiological role(s) played by complement in each kidney disease is not fully understood. However, in recent years there has been a renaissance in the study of complement, with greater appreciation of its intracellular roles as a cell-intrinsic system and its interplay with immune effector pathways. This has been paired with a profusion of novel therapeutic agents antagonizing complement components, including approved inhibitors against complement components (C)1, C3, C5 and C5aR1. A number of clinical trials have investigated the use of these more targeted approaches for the management of kidney diseases. In this review we present and summarize the evidence for the roles of complement in kidney diseases and discuss the available clinical evidence for complement inhibition.
Collapse
Affiliation(s)
- Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD, United States; Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Behdad Afzali
- Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
8
|
Polack FP, Alvarez-Paggi D, Libster R, Caballero MT, Blair RV, Hijano DR, de la Iglesia Niveyro PX, Menendez DR, Gladwell W, Avendano LM, Velozo L, Wanek A, Bergel E, Prince GA, Kleeberger SR, Johnson J, Pociask D, Kolls JK. Fatal enhanced respiratory syncytial virus disease in toddlers. Sci Transl Med 2021; 13:eabj7843. [PMID: 34669442 PMCID: PMC10712289 DOI: 10.1126/scitranslmed.abj7843] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In 1967, two toddlers immunized with a formalin-inactivated vaccine against respiratory syncytial virus (FIRSV) in the United States died from enhanced RSV disease (ERD), a severe form of illness resulting from aberrant priming of the antiviral immune response during vaccination. Up to 80% of immunized children subsequently exposed to wild-type virus were hospitalized. These events hampered RSV vaccine development for decades. Here, we provide a characterization of the clinical, immunopathological, and transcriptional signature of fatal human ERD, outlining evidence for safety evaluation of RSV vaccines and a framework for understanding disease enhancement for pathogens in general.
Collapse
Affiliation(s)
- Fernando P. Polack
- Fundación INFANT, Buenos Aires, Argentina
- Vanderbilt University, Nashville, TN, USA
| | - Damián Alvarez-Paggi
- Fundación INFANT, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Romina Libster
- Fundación INFANT, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Mauricio T. Caballero
- Fundación INFANT, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | | | - Diego R. Hijano
- Fundación INFANT, Buenos Aires, Argentina
- St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Daniel R. Menendez
- National Institute of Environmental Health Sciences (NIEHS), Raleigh, NC, USA
| | - Wes Gladwell
- National Institute of Environmental Health Sciences (NIEHS), Raleigh, NC, USA
| | | | - Luis Velozo
- Universidad Nacional de Chile, Santiago, Chile
| | | | - Eduardo Bergel
- Instituto de Efectividad Clinica y Sanitaria, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
9
|
Nicholas TP, Boyes WK, Scoville DK, Workman TW, Kavanagh TJ, Altemeier WA, Faustman EM. The effects of gene × environment interactions on silver nanoparticle toxicity in the respiratory system: An adverse outcome pathway. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1708. [PMID: 33768701 DOI: 10.1002/wnan.1708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/07/2021] [Accepted: 01/30/2021] [Indexed: 11/07/2022]
Abstract
The Adverse Outcome Pathway (AOP) framework is serving as a basis to integrate new data streams in order to enhance the power of predictive toxicology. AOP development for engineered nanomaterials (ENM), including silver nanoparticles (AgNP), is currently lagging behind other chemicals of regulatory interest due to our limited understanding of the mechanism by which underlying genetics or diseases directly modify host response to AgNP exposures. This also highlights the importance of considering the Aggregate Exposure Pathway (AEP) framework, which precedes the AOP framework and outlines source to target site exposure. The AEP and AOP frameworks interface at the target site, where a molecular initiating event (MIE) occurs and is followed by key events (KE) for adverse cellular and organ responses along a biological pathway and ends with the adverse organism response. The primary goal of this study is to use AgNP to interrogate the AEP-AOP framework by organizing and integrating in vitro dose-response data and in vivo exposure-response data from previous studies to evaluate the effects of interactions between host genetic and acquired factors, or gene × environment interactions (G × E), on AgNP toxicity in the respiratory system. Using this framework will help us to identify plausible key event relationships (KER) between MIE and adverse organism responses when KE are not measured using the same assay in order to derive future predictive models, guide research, and support development of tools for making risk-based, regulatory decisions on ENM. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Tyler P Nicholas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - William K Boyes
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - David K Scoville
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Tomomi W Workman
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - William A Altemeier
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - Elaine M Faustman
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Li Y, Yi JS, Howard JF, Chopra M, Russo MA, Guptill JT. Cellular changes in eculizumab early responders with generalized myasthenia gravis. Clin Immunol 2021; 231:108830. [PMID: 34450290 DOI: 10.1016/j.clim.2021.108830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 11/19/2022]
Abstract
Eculizumab (ECU), a C5 complement inhibitor, is approved to treat acetylcholine receptor autoantibody positive generalized myasthenia gravis (AChR MG). The clinical effect of ECU relies on inhibition of the terminal complement complex; however, the effect of ECU on lymphocytes is largely unknown. We evaluated innate and adaptive immunity among AChR MG patients (N = 3) before ECU and ≥3 months later while on stable therapy, and found reduced activation markers in memory CD4+ T cell subsets, increased regulatory T cell populations, and reduced frequencies of CXCR5+HLA-DR+CCR7+ Tfh subsets and CD11b+ migratory memory B cells. We observed increases within CD8+ T cell subsets that were terminally differentiated and senescent. Our data suggest complement inhibition with ECU modulates the adaptive immunity in patients with MG, consistent with preclinical data showing changes in complement-mediated signaling by T- and antigen-presenting cells. These findings extend our understanding of ECU's mechanism of action when treating patients with MG.
Collapse
Affiliation(s)
- Yingkai Li
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - John S Yi
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - James F Howard
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Manisha Chopra
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa A Russo
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey T Guptill
- Department of Neurology, Duke University Medical Center, Durham, NC, USA; Duke Clinical Research Institute, Durham, NC, USA.
| |
Collapse
|
11
|
Jahns H, Taneja N, Willoughby JLS, Akabane-Nakata M, Brown CR, Nguyen T, Bisbe A, Matsuda S, Hettinger M, Manoharan RM, Rajeev KG, Maier MA, Zlatev I, Charisse K, Egli M, Manoharan M. Chirality matters: stereo-defined phosphorothioate linkages at the termini of small interfering RNAs improve pharmacology in vivo. Nucleic Acids Res 2021; 50:1221-1240. [PMID: 34268578 PMCID: PMC8860597 DOI: 10.1093/nar/gkab544] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/29/2021] [Accepted: 06/30/2021] [Indexed: 12/04/2022] Open
Abstract
A critical challenge for the successful development of RNA interference-based therapeutics therapeutics has been the enhancement of their in vivo metabolic stability. In therapeutically relevant, fully chemically modified small interfering RNAs (siRNAs), modification of the two terminal phosphodiester linkages in each strand of the siRNA duplex with phosphorothioate (PS) is generally sufficient to protect against exonuclease degradation in vivo. Since PS linkages are chiral, we systematically studied the properties of siRNAs containing single chiral PS linkages at each strand terminus. We report an efficient and simple method to introduce chiral PS linkages and demonstrate that Rp diastereomers at the 5′ end and Sp diastereomers at the 3′ end of the antisense siRNA strand improved pharmacokinetic and pharmacodynamic properties in a mouse model. In silico modeling studies provide mechanistic insights into how the Rp isomer at the 5′ end and Sp isomer at the 3′ end of the antisense siRNA enhance Argonaute 2 (Ago2) loading and metabolic stability of siRNAs in a concerted manner.
Collapse
Affiliation(s)
- Hartmut Jahns
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Nate Taneja
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | | | | | | | - Tuyen Nguyen
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Anna Bisbe
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Shigeo Matsuda
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Matt Hettinger
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Rajar M Manoharan
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | | | - Martin A Maier
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Ivan Zlatev
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Klaus Charisse
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| | - Martin Egli
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Muthiah Manoharan
- Alnylam Pharmaceuticals, 675 W. Kendall St, Cambridge, MA 02142, USA
| |
Collapse
|
12
|
Fan KH, Devos KM, Schliekelman P. Strategies for eQTL mapping in allopolyploid organisms. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2020; 133:2477-2497. [PMID: 32462429 DOI: 10.1007/s00122-020-03612-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
KEY MESSAGE This study uses simulations to explore statistical power and false-positive rates for eQTL mapping in allopolyploid organisms and provides guidelines to apply eQTL mapping in these organisms. In recent years, RNA-seq has become the dominant technology for eQTL studies. However, most work has been in diploid organisms. Many species of economic and environmental importance are polyploid, and approaches for eQTL mapping in polyploids are not well developed. High similarity between duplicated genes in polyploids will cause misassignment of sequence reads and may cause false-positive results and/or lack of power to detect eQTL. In this paper, we first explore the similarity of homoeologous transcripts in polyploid organisms. We find that 5-20% of genes (varying with organism) in important agricultural plants such as wheat, soybean, and switchgrass are not sufficiently diverged between duplicated genomes to allow unambiguous assignment of reads. Second, we examine the impact of misassigned reads on eQTL mapping and show that both false-positive and false-negative rates can be greatly inflated. Third, we compare four strategies for dealing with ambiguous reads: (1) dividing ambiguous reads evenly between homoeologous transcripts, (2) assigning them proportionally, (3) using all reads for all genes, and (4) discarding ambiguous reads. We find that the strategy of discarding ambiguous reads gives the best balance of false-positive and false-negative rates for most genes. However, for genes that are very similar between genomes, using all reads is the only choice. This leads to reduced power, but false-positive rates will be maintained. We also discuss QTL mapping in polyploids using allele-specific expression (ASE) and show how the proportion of ASE-informative reads varies according to the divergence between homoeologous genes.
Collapse
Affiliation(s)
- Kang-Hsien Fan
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Katrien M Devos
- Department of Crop and Soil Sciences, Institute of Plant Breeding, Genetics and Genomics, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
13
|
Briukhovetska D, Ohm B, Mey FT, Aliberti J, Kleingarn M, Huber-Lang M, Karsten CM, Köhl J. C5aR1 Activation Drives Early IFN-γ Production to Control Experimental Toxoplasma gondii Infection. Front Immunol 2020; 11:1397. [PMID: 32733463 PMCID: PMC7362728 DOI: 10.3389/fimmu.2020.01397] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a parasite infecting animals and humans. In intermediate hosts, such as humans or rodents, rapidly replicating tachyzoites drive vigorous innate and adaptive immune responses resulting in bradyzoites that survive within tissue cysts. Activation of the innate immune system is critical during the early phase of infection to limit pathogen growth and to instruct parasite-specific adaptive immunity. In rodents, dendritic cells (DCs) sense T. gondii through TLR11/12, leading to IL-12 production, which activates NK cells to produce IFN-γ as an essential mechanism for early parasite control. Further, C3 can bind to T. gondii resulting in limited complement activation. Here, we determined the role of C5a/C5aR1 axis activation for the early innate immune response in a mouse model of peritoneal T. gondii infection. We found that C5ar1−/− animals suffered from significantly higher weight loss, disease severity, mortality, and parasite burden in the brain than wild type control animals. Severe infection in C5ar1−/− mice was associated with diminished serum concentrations of IL-12, IL-27, and IFN-γ. Importantly, the serum levels of pro-inflammatory cytokines, including IL-1α, IL-6, and TNF-α, as well as several CXC and CC chemokines, were decreased in comparison to wt animals, whereas anti-inflammatory IL-10 was elevated. The defect in IFN-γ production was associated with diminished Ifng mRNA expression in the spleen and the brain, reduced frequency of IFN-γ+ NK cells in the spleen, and decreased Nos2 expression in the brain of C5ar1−/− mice. Mechanistically, DCs from the spleen of C5ar1−/− mice produced significantly less IL-12 in response to soluble tachyzoite antigen (STAg) stimulation in vivo and in vitro. Our findings suggest a model in which the C5a/C5aR1 axis promotes IL-12 induction in splenic DCs that is critical for IFN-γ production from NK cells and subsequent iNOS expression in the brain as a critical mechanism to control acute T. gondii infection.
Collapse
Affiliation(s)
- Daria Briukhovetska
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Birte Ohm
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Fabian T Mey
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Julio Aliberti
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Marie Kleingarn
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital and College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
14
|
Antoniou K, Ender F, Vollbrandt T, Laumonnier Y, Rathmann F, Pasare C, Singh H, Köhl J. Allergen-Induced C5a/C5aR1 Axis Activation in Pulmonary CD11b + cDCs Promotes Pulmonary Tolerance through Downregulation of CD40. Cells 2020; 9:cells9020300. [PMID: 31991941 PMCID: PMC7072238 DOI: 10.3390/cells9020300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022] Open
Abstract
Activation of the C5/C5a/C5a receptor 1 (C5aR1) axis during allergen sensitization protects from maladaptive T cell activation. To explore the underlying regulatory mechanisms, we analyzed the impact of C5aR1 activation on pulmonary CD11b+ conventional dendritic cells (cDCs) in the context of house-dust-mite (HDM) exposure. BALB/c mice were intratracheally immunized with an HDM/ovalbumin (OVA) mixture. After 24 h, we detected two CD11b+ cDC populations that could be distinguished on the basis of C5aR1 expression. C5aR1− but not C5aR1+ cDCs strongly induced T cell proliferation of OVA-reactive transgenic CD4+ T cells after re-exposure to antigen in vitro. C5aR1− cDCs expressed higher levels of MHC-II and CD40 than their C5aR1+ counterparts, which correlated directly with a higher frequency of interactions with cognate CD4+ T cells. Priming of OVA-specific T cells by C5aR1+ cDCs could be markedly increased by in vitro blockade of C5aR1 and this was associated with increased CD40 expression. Simultaneous blockade of C5aR1 and CD40L on C5aR1+ cDCs decreased T cell proliferation. Finally, pulsing with OVA-induced C5 production and its cleavage into C5a by both populations of CD11b+ cDCs. Thus, we propose a model in which allergen-induced autocrine C5a generation and subsequent C5aR1 activation in pulmonary CD11b+ cDCs promotes tolerance towards aeroallergens through downregulation of CD40.
Collapse
Affiliation(s)
- Konstantina Antoniou
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; (K.A.); (F.E.); (Y.L.); (F.R.)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; (K.A.); (F.E.); (Y.L.); (F.R.)
| | | | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; (K.A.); (F.E.); (Y.L.); (F.R.)
- Airway Research Center North, Member of the German Center for Lung Research (DZL), 23562 Lübeck, Germany
| | - Franziska Rathmann
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; (K.A.); (F.E.); (Y.L.); (F.R.)
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA (H.S.)
| | - Harinder Singh
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA (H.S.)
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; (K.A.); (F.E.); (Y.L.); (F.R.)
- Cell Analysis Core, University of Lübeck, 23562 Lübeck, Germany;
- Airway Research Center North, Member of the German Center for Lung Research (DZL), 23562 Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA (H.S.)
- Correspondence: ; Tel.: +49-451-500-51400
| |
Collapse
|
15
|
Nicholas TP, Haick AK, Bammler TK, Workman TW, Kavanagh TJ, Faustman EM, Gharib SA, Altemeier WA. The Effects of Genotype × Phenotype Interactions on Transcriptional Response to Silver Nanoparticle Toxicity in Organotypic Cultures of Murine Tracheal Epithelial Cells. Toxicol Sci 2020; 173:131-143. [PMID: 31562762 PMCID: PMC6944213 DOI: 10.1093/toxsci/kfz209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The airway epithelium is critical for maintaining innate and adaptive immune responses, and occupational exposures that disrupt its immune homeostasis may initiate and amplify airway inflammation. In our previous study, we demonstrated that silver nanoparticles (AgNP), which are engineered nanomaterials used in multiple applications but primarily in the manufacturing of many antimicrobial products, induce toxicity in organotypic cultures derived from murine tracheal epithelial cells (MTEC), and those differentiated toward a "Type 2 [T2]-Skewed" phenotype experienced an increased sensitivity to AgNP toxicity, suggesting that asthmatics could be a sensitive population to AgNP exposures in occupational settings. However, the mechanistic basis for this genotype × phenotype (G × P) interaction has yet to be defined. In this study, we conducted transcriptional profiling using RNA-sequencing to predict the enrichment of specific canonical pathways and upstream transcriptional regulators to assist in defining a mechanistic basis for G × P effects on AgNP toxicity. Organotypic cultures were derived from MTEC across 2 genetically inbred mouse strains (A/J and C57BL/6J mice), 2 phenotypes ("Normal" and "T2-Skewed"), and 1 AgNP exposure (an acute 24 h exposure) to characterize G × P effects on transcriptional response to AgNP toxicity. The "T2-Skewed" phenotype was marked by increased pro-inflammatory T17 responses to AgNP toxicity, which are significant predictors of neutrophilic/difficult-to-control asthma and suggests that asthmatics could be a sensitive population to AgNP exposures in occupational settings. This study highlights the importance of considering G × P effects when identifying these sensitive populations, whose underlying genetics or diseases could directly modify their response to AgNP exposures.
Collapse
Affiliation(s)
- Tyler P Nicholas
- Department of Environmental and Occupational Health Sciences
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Anoria K Haick
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences
| | | | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Sina A Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Altemeier
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
16
|
Association of TRAF1/C5 Locus Polymorphisms with Epilepsy and Clinical Traits in Mexican Patients with Neurocysticercosis. Infect Immun 2019; 87:IAI.00347-19. [PMID: 31570557 DOI: 10.1128/iai.00347-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/20/2019] [Indexed: 02/04/2023] Open
Abstract
Neurocysticercosis is caused by the establishment of Taenia solium cysts in the central nervous system. Murine cysticercosis by Taenia crassiceps is a useful model of cysticercosis in which the complement component 5 (C5) has been linked to infection resistance/permissiveness. This work aimed to study the possible relevance for human neurocysticercosis of single nucleotide polymorphisms (SNPs) in the C5-TRAF1 region (rs17611 C/T, rs992670 G/A, rs25681 G/A, rs10818488 A/G, and rs3761847 G/A) in a Mexican population and associated with clinical and radiological traits related to neurocysticercosis severity (cell count in the cerebrospinal fluid [CSF cellularity], parasite location and parasite load in the brain, parasite degenerating stage, and epilepsy). The AG genotype of the rs3761847 SNP showed a tendency to associate with multiple brain parasites, while the CT and GG genotypes of the rs17611 and rs3761847 SNPs, respectively, showed a tendency to associate with low CSF cellularity. The rs3761847 SNP was associated with epilepsy under a dominant model, whereas rs10818488 was associated with CSF cellularity and parasite load under dominant and recessive models, respectively. For haplotypes, C5- and the TRAF1-associated SNPs were, respectively, in strong linkage disequilibrium with each other; thus, these haplotypes were studied independently. For C5 SNPs, carrying the CAA haplotype increases the risk of showing high CSF cellularity 3-fold and the risk of having extraparenchymal parasites 4-fold, two conditions that are related to severe disease. For TRAF1 SNPs, the GA and AG haplotypes were associated with CSF cellularity, and the AG haplotype was associated with epilepsy. Overall, these findings support the clear participation of C5 and TRAF1 in the risk of developing severe neurocysticercosis in the Mexican population.
Collapse
|
17
|
Fukumori C, Casaro MB, Thomas AM, Mendes E, Ribeiro WR, Crisma AR, Murata GM, Bizzarro B, Dias-Neto E, Setubal JC, Oliveira MA, Tavares-de-Lima W, Curi R, Bordin S, Sartorelli P, Ferreira CM. Maternal supplementation with a synbiotic has distinct outcomes on offspring gut microbiota formation in A/J and C57BL/6 mice, differentially affecting airway inflammatory cell infiltration and mucus production. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
18
|
Effect of complement component 5 polymorphisms on mastitis resistance in Egyptian buffalo and cattle. Res Vet Sci 2018; 119:45-51. [PMID: 29857245 DOI: 10.1016/j.rvsc.2018.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/16/2018] [Accepted: 05/23/2018] [Indexed: 11/20/2022]
Abstract
Mastitis is one of the costliest diseases affecting the world's dairy industry. The important contribution of complement Component 5 (C5) to phagocytosis, which plays a major role in the defence of the bovine mammary gland against infection, makes this component of innate immunity a potential contributor in defending udder against mastitis. The objectives of this study were to sequence and analyse the whole coding region of the C5 gene in Egyptian buffalo and cattle, to detect any nucleotide variations (polymorphisms) and to investigate their associations with milk somatic cell score (SCS) as an indicator of mastitis in dairy animals. We sequenced a buffalo C5 cDNA fragment of 5336 bp (KP221293) and a cattle C5 cDNA fragment of 5303 bp (KP221294), which included the whole coding region and 3-UTR. Buffalo and cattle C5 cDNA shared sequence identity of 99%. The predicted complement C5 proteins consist of 1677 amino acid residues in both animals, one amino acid less than in humans and three amino acids more than in mouse C5 protein. Comparing cDNA sequences of different animals revealed nine novel SNPs in buffalo and seven SNPs in cattle, with two of them being novel. The association analysis revealed that five SNPs in buffalo are highly associated with SCS; indicating the contribution of complement C5 variants in buffalo mastitis resistance. No significant associations were detected between C5 variants and SCS in cattle. This is the first report about C5 variants in buffalo and its association with SCS.
Collapse
|
19
|
Abstract
The complement system is an evolutionarily ancient key component of innate immunity required for the detection and removal of invading pathogens. It was discovered more than 100 years ago and was originally defined as a liver-derived, blood-circulating sentinel system that classically mediates the opsonization and lytic killing of dangerous microbes and the initiation of the general inflammatory reaction. More recently, complement has also emerged as a critical player in adaptive immunity via its ability to instruct both B and T cell responses. In particular, work on the impact of complement on T cell responses led to the surprising discoveries that the complement system also functions within cells and is involved in regulating basic cellular processes, predominantly those of metabolic nature. Here, we review current knowledge about complement's role in T cell biology, with a focus on the novel intracellular and noncanonical activities of this ancient system.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
| | - Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
20
|
West EE, Afzali B, Kemper C. Unexpected Roles for Intracellular Complement in the Regulation of Th1 Responses. Adv Immunol 2018; 138:35-70. [PMID: 29731006 DOI: 10.1016/bs.ai.2018.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The complement system is generally recognized as an evolutionarily ancient and critical part of innate immunity required for the removal of pathogens that have breached the protective host barriers. It was originally defined as a liver-derived serum surveillance system that induces the opsonization and killing of invading microbes and amplifies the general inflammatory reactions. However, studies spanning the last four decades have established complement also as a vital bridge between innate and adaptive immunity. Furthermore, recent work on complement, and in particular its impact on human T helper 1 (Th1) responses, has led to the unexpected findings that the complement system also functions within cells and that it participates in the regulation of basic processes of the cell, including metabolism. These recent new insights into the unanticipated noncanonical activities of this ancient system suggest that the functions of complement extend well beyond mere host protection and into cellular physiology.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Behdad Afzali
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States; Lymphocyte Cell Biology Section (Molecular Immunology and Inflammation Branch), National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States; Division of Transplant Immunology and Mucosal Biology, King's College London, London, United Kingdom; Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
21
|
Padmanabhan S, Joe B. Towards Precision Medicine for Hypertension: A Review of Genomic, Epigenomic, and Microbiomic Effects on Blood Pressure in Experimental Rat Models and Humans. Physiol Rev 2017; 97:1469-1528. [PMID: 28931564 PMCID: PMC6347103 DOI: 10.1152/physrev.00035.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 12/11/2022] Open
Abstract
Compelling evidence for the inherited nature of essential hypertension has led to extensive research in rats and humans. Rats have served as the primary model for research on the genetics of hypertension resulting in identification of genomic regions that are causally associated with hypertension. In more recent times, genome-wide studies in humans have also begun to improve our understanding of the inheritance of polygenic forms of hypertension. Based on the chronological progression of research into the genetics of hypertension as the "structural backbone," this review catalogs and discusses the rat and human genetic elements mapped and implicated in blood pressure regulation. Furthermore, the knowledge gained from these genetic studies that provide evidence to suggest that much of the genetic influence on hypertension residing within noncoding elements of our DNA and operating through pervasive epistasis or gene-gene interactions is highlighted. Lastly, perspectives on current thinking that the more complex "triad" of the genome, epigenome, and the microbiome operating to influence the inheritance of hypertension, is documented. Overall, the collective knowledge gained from rats and humans is disappointing in the sense that major hypertension-causing genes as targets for clinical management of essential hypertension may not be a clinical reality. On the other hand, the realization that the polygenic nature of hypertension prevents any single locus from being a relevant clinical target for all humans directs future studies on the genetics of hypertension towards an individualized genomic approach.
Collapse
Affiliation(s)
- Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and Center for Hypertension and Personalized Medicine; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Bina Joe
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and Center for Hypertension and Personalized Medicine; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
22
|
Laumonnier Y, Wiese AV, Figge J, Karsten C. Regulation and function of anaphylatoxins and their receptors in allergic asthma. Mol Immunol 2017; 84:51-56. [PMID: 27916272 DOI: 10.1016/j.molimm.2016.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023]
Abstract
Allergic asthma is a disease of the airways driven by maladaptive T helper 2 (Th2) and Th17 immune response against harmless, airborne substances. The hallmarks of this disease are airway hyperresponsiveness (AHR), eosinophilic and neutrophilic airway inflammation and mucus overproduction. Distinct dendric cell (DC) subsets together with airway epithelial and pulmonary vascular endothelial cells play critical roles in allergen sensing and in driving T cell differentiation towards Th2 and Th17 effector or regulatory T cells (Treg). Previous studies suggested already a pivotal role for the anaphylatoxins (C5a, C3a) in the pathogenesis of allergic asthma. During sensitization for example it is described, that C3a promotes, whereas C5a protects from the development of maladaptive immunity during allergen sensitization. Here we will discuss the role of the anaphylatoxins (C3a, C5a) and their receptors during the pathogenesis of allergic asthma, and specifically in lung DC biology. We will also have a look on canonical and non-canonical complement activation and we will discuss novel concepts on how the adaptive immune system can regulate the function of ATRs also in the context of allergic asthma.
Collapse
Affiliation(s)
- Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany.
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Christian Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
23
|
Verschoor A, Karsten CM, Broadley SP, Laumonnier Y, Köhl J. Old dogs-new tricks: immunoregulatory properties of C3 and C5 cleavage fragments. Immunol Rev 2017; 274:112-126. [PMID: 27782330 DOI: 10.1111/imr.12473] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The activation of the complement system by canonical and non-canonical mechanisms results in the generation of multiple C3 and C5 cleavage fragments including anaphylatoxins C3a and C5a as well as opsonizing C3b/iC3b. It is now well appreciated that anaphylatoxins not only act as pro-inflammatory mediators but as immunoregulatory molecules that control the activation status of cells and tissue at several levels. Likewise, C3b/iC3b is more than the opsonizing fragment that facilitates engulfment and destruction of targets by phagocytes. In the circulation, it also facilitates the transport and delivery of bacteria and immune complexes to phagocytes, through a process known as immune adherence, with consequences for adaptive immunity. Here, we will discuss non-classical immunoregulatory properties of C3 and C5 cleavage fragments. We highlight the influence of anaphylatoxins on Th2 and Th17 cell development during allergic asthma with a particular emphasis on their role in the modulation of CD11b+ conventional dendritic cells and monocyte-derived dendritic cells. Furthermore, we discuss the control of anaphylatoxin-mediated activation of dendritic cells and allergic effector cells by adaptive immune mechanisms that involve allergen-specific IgG1 antibodies and plasma or regulatory T cell-derived IL-10 production. Finally, we take a fresh look at immune adherence with a particular focus on the development of antibacterial cytotoxic T-cell responses.
Collapse
Affiliation(s)
- Admar Verschoor
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Steven P Broadley
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany. .,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
24
|
Ender F, Wiese AV, Schmudde I, Sun J, Vollbrandt T, König P, Laumonnier Y, Köhl J. Differential regulation of C5a receptor 1 in innate immune cells during the allergic asthma effector phase. PLoS One 2017; 12:e0172446. [PMID: 28231307 PMCID: PMC5322932 DOI: 10.1371/journal.pone.0172446] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022] Open
Abstract
C5a drives airway constriction and inflammation during the effector phase of allergic asthma, mainly through the activation of C5a receptor 1 (C5aR1). Yet, C5aR1 expression on myeloid and lymphoid cells during the allergic effector phase is ill-defined. Recently, we generated and characterized a floxed green fluorescent protein (GFP)-C5aR1 knock-in mouse. Here, we used this reporter strain to monitor C5aR1 expression in airway, pulmonary and lymph node cells during the effector phase of OVA-driven allergic asthma. C5aR1 reporter and wildtype mice developed a similar allergic phenotype with comparable airway resistance, mucus production, eosinophilic/neutrophilic airway inflammation and Th2/Th17 cytokine production. During the allergic effector phase, C5aR1 expression increased in lung tissue eosinophils but decreased in airway and pulmonary macrophages as well as in pulmonary CD11b+ conventional dendritic cells (cDCs) and monocyte-derived DCs (moDCs). Surprisingly, expression in neutrophils was not affected. Of note, moDCs but not CD11b+ cDCs from mediastinal lymph nodes (mLN) expressed less C5aR1 than DCs residing in the lung after OVA challenge. Finally, neither CD103+ cDCs nor cells of the lymphoid lineage such as Th2 or Th17-differentiated CD4+ T cells, B cells or type 2 innate lymphoid cells (ILC2) expressed C5aR1 under allergic conditions. Our findings demonstrate a complex regulation pattern of C5aR1 in the airways, lung tissue and mLN of mice, suggesting that the C5a/C5aR1 axis controls airway constriction and inflammation through activation of myeloid cells in all three compartments in an experimental model of allergic asthma.
Collapse
Affiliation(s)
- Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Anna V. Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Jing Sun
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | | | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- * E-mail: (JK); (YL)
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail: (JK); (YL)
| |
Collapse
|
25
|
Carroll AM, Cheng R, Collie-Duguid ESR, Meharg C, Scholz ME, Fiering S, Fields JL, Palmer AA, Lionikas A. Fine-mapping of genes determining extrafusal fiber properties in murine soleus muscle. Physiol Genomics 2017; 49:141-150. [PMID: 28087756 PMCID: PMC5374456 DOI: 10.1152/physiolgenomics.00092.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 02/08/2023] Open
Abstract
Muscle fiber cross-sectional area (CSA) and proportion of different fiber types are important determinants of muscle function and overall metabolism. Genetic variation plays a substantial role in phenotypic variation of these traits; however, the underlying genes remain poorly understood. This study aimed to map quantitative trait loci (QTL) affecting differences in soleus muscle fiber traits between the LG/J and SM/J mouse strains. Fiber number, CSA, and proportion of oxidative type I fibers were assessed in the soleus of 334 genotyped female and male mice of the F34 generation of advanced intercross lines (AIL) derived from the LG/J and SM/J strains. To increase the QTL detection power, these data were combined with 94 soleus samples from the F2 intercross of the same strains. Transcriptome of the soleus muscle of LG/J and SM/J females was analyzed by microarray. Genome-wide association analysis mapped four QTL (genome-wide P < 0.05) affecting the properties of muscle fibers to chromosome 2, 3, 4, and 11. A 1.5-LOD QTL support interval ranged between 2.36 and 4.67 Mb. On the basis of the genomic sequence information and functional and transcriptome data, we identified candidate genes for each of these QTL. The combination of analyses in F2 and F34 AIL populations with transcriptome and genomic sequence data in the parental strains is an effective strategy for refining QTL and nomination of the candidate genes.
Collapse
Affiliation(s)
- A M Carroll
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill Aberdeen, United Kingdom.,The New Zealand Institute for Plant & Food Research Limited, Palmerston North, New Zealand
| | - R Cheng
- Research School of Biology, Australian National University, Acton, Australia
| | - E S R Collie-Duguid
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill Aberdeen, United Kingdom.,Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Old Aberdeen, Aberdeen, United Kingdom
| | - C Meharg
- Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| | - M E Scholz
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill Aberdeen, United Kingdom
| | - S Fiering
- Department of Microbiology/Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire; and
| | - J L Fields
- Department of Microbiology/Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire; and
| | - A A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - A Lionikas
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill Aberdeen, United Kingdom;
| |
Collapse
|
26
|
Revealing the acute asthma ignorome: characterization and validation of uninvestigated gene networks. Sci Rep 2016; 6:24647. [PMID: 27097888 PMCID: PMC4838989 DOI: 10.1038/srep24647] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/01/2016] [Indexed: 01/12/2023] Open
Abstract
Systems biology provides opportunities to fully understand the genes and pathways in disease pathogenesis. We used literature knowledge and unbiased multiple data meta-analysis paradigms to analyze microarray datasets across different mouse strains and acute allergic asthma models. Our combined gene-driven and pathway-driven strategies generated a stringent signature list totaling 933 genes with 41% (440) asthma-annotated genes and 59% (493) ignorome genes, not previously associated with asthma. Within the list, we identified inflammation, circadian rhythm, lung-specific insult response, stem cell proliferation domains, hubs, peripheral genes, and super-connectors that link the biological domains (Il6, Il1ß, Cd4, Cd44, Stat1, Traf6, Rela, Cadm1, Nr3c1, Prkcd, Vwf, Erbb2). In conclusion, this novel bioinformatics approach will be a powerful strategy for clinical and across species data analysis that allows for the validation of experimental models and might lead to the discovery of novel mechanistic insights in asthma.
Collapse
|
27
|
Moreno-Fernandez ME, Aliberti J, Groeneweg S, Köhl J, Chougnet CA. A Novel Role for the Receptor of the Complement Cleavage Fragment C5a, C5aR1, in CCR5-Mediated Entry of HIV into Macrophages. AIDS Res Hum Retroviruses 2016; 32:399-408. [PMID: 26537334 DOI: 10.1089/aid.2015.0099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The complement system is an ancient pattern recognition system that becomes activated during all stages of HIV infection. Previous studies have shown that C5a can enhance the infection of monocyte-derived macrophages and T cells indirectly through the production of interleukin (IL)-6 and tumor necrosis factor (TNF)-α and the attraction of dendritic cells. C5a exerts its multiple biologic functions mainly through activation of C5a receptor 1 (C5aR1). Here, we assessed the role of C5aR1 as an enhancer of CCR5-mediated HIV infection. We determined CCR5 and C5aR1 heterodimer formation in myeloid cells and the impact of C5aR1 blockade on HIV entry and genomic integration. C5aR1/CCR5 heterodimer formation was identified by immunoprecipitation and western blotting. THP-1 cells and monocyte-derived macrophages (MDM) were infected by R5 laboratory strains or HIV pseudotyped for the vesicular stomatitis virus (VSV) envelope. Levels of integrated HIV were measured by quantitative PCR after targeting of C5aR1 by a C5aR antagonist, neutralizing C5aR1 monoclonal antibody (mAb) or hC5a. C5aR1 was also silenced by specific siRNA prior to viral entry. We found that C5aR1 forms heterodimers with the HIV coreceptor CCR5 in myeloid cells. Targeting C5aR1 significantly decreased integration by R5 viruses but not by VSV-pseudotyped viruses, suggesting that C5aR1 is critical for viral entry. The level of inhibition achieved with C5aR1-blocking reagents was comparable to that of CCR5 antagonists. Mechanistically, C5aR1 targeting decreased CCR5 expression. MDM from CCR5Δ32 homozygous subjects expressed levels of C5aR1 similar to CCR5 WT individuals, suggesting that mere C5aR1 expression is not sufficient for HIV infection. HIV appeared to preferentially enter THP-1 cells expressing high levels of both C5aR1 and CCR5. Targeted reduction of C5aR1 expression in such cells reduced HIV infection by ~50%. Our data thus suggest that C5aR1 acts as an enhancer of CCR5-mediated HIV entry into macrophages, the targeting of which may prove useful to reduce HIV infection by R5 strains.
Collapse
Affiliation(s)
- Maria E. Moreno-Fernandez
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Julio Aliberti
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Sander Groeneweg
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati, College of Medicine, Cincinnati, Ohio
| |
Collapse
|
28
|
Impaired Cell Cycle Regulation in a Natural Equine Model of Asthma. PLoS One 2015; 10:e0136103. [PMID: 26292153 PMCID: PMC4546272 DOI: 10.1371/journal.pone.0136103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 07/29/2015] [Indexed: 12/14/2022] Open
Abstract
Recurrent airway obstruction (RAO) is a common and potentially debilitating lower airway disease in horses, which shares many similarities with human asthma. In susceptible horses RAO exacerbation is caused by environmental allergens and irritants present in hay dust. The objective of this study was the identification of genes and pathways involved in the pathology of RAO by global transcriptome analyses in stimulated peripheral blood mononuclear cells (PBMCs). We performed RNA-seq on PBMCs derived from 40 RAO affected and 45 control horses belonging to three cohorts of Warmblood horses: two half-sib families and one group of unrelated horses. PBMCs were stimulated with hay dust extract, lipopolysaccharides, a recombinant parasite antigen, or left unstimulated. The total dataset consisted of 561 individual samples. We detected significant differences in the expression profiles between RAO and control horses. Differential expression (DE) was most marked upon stimulation with hay dust extract. An important novel finding was a strong upregulation of CXCL13 together with many genes involved in cell cycle regulation in stimulated samples from RAO affected horses, in addition to changes in the expression of several HIF-1 transcription factor target genes. The RAO condition alters systemic changes observed as differential expression profiles of PBMCs. Those changes also depended on the cohort and stimulation of the samples and were dominated by genes involved in immune cell trafficking, development, and cell cycle regulation. Our findings indicate an important role of CXCL13, likely macrophage or Th17 derived, and the cell cycle regulator CDC20 in the immune response in RAO.
Collapse
|
29
|
Wang Y, Miwa T, Ducka-Kokalari B, Redai IG, Sato S, Gullipalli D, Zangrilli JG, Haczku A, Song WC. Properdin Contributes to Allergic Airway Inflammation through Local C3a Generation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1171-81. [PMID: 26116506 DOI: 10.4049/jimmunol.1401819] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 05/23/2015] [Indexed: 01/04/2023]
Abstract
Complement is implicated in asthma pathogenesis, but its mechanism of action in this disease remains incompletely understood. In this study, we investigated the role of properdin (P), a positive alternative pathway complement regulator, in allergen-induced airway inflammation. Allergen challenge stimulated P release into the airways of asthmatic patients, and P levels positively correlated with proinflammatory cytokines in human bronchoalveolar lavage (BAL). High levels of P were also detected in the BAL of OVA-sensitized and challenged but not naive mice. Compared with wild-type (WT) mice, P-deficient (P(-/-)) mice had markedly reduced total and eosinophil cell counts in BAL and significantly attenuated airway hyperresponsiveness to methacholine. Ab blocking of P at both sensitization and challenge phases or at challenge phase alone, but not at sensitization phase alone, reduced airway inflammation. Conversely, intranasal reconstitution of P to P(-/-) mice at the challenge phase restored airway inflammation to wild-type levels. Notably, C3a levels in the BAL of OVA-challenged P(-/-) mice were significantly lower than in wild-type mice, and intranasal coadministration of an anti-C3a mAb with P to P(-/-) mice prevented restoration of airway inflammation. These results show that P plays a key role in allergen-induced airway inflammation and represents a potential therapeutic target for human asthma.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Blerina Ducka-Kokalari
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Imre G Redai
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Sayaka Sato
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Angela Haczku
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
30
|
Park CS. Role of murine asthma model in discovering asthma susceptible genes. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2014; 6:475-7. [PMID: 25374745 PMCID: PMC4214966 DOI: 10.4168/aair.2014.6.6.475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Choon-Sik Park
- Division of Allergy & Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
31
|
Engelke C, Wiese AV, Schmudde I, Ender F, Ströver HA, Vollbrandt T, König P, Laumonnier Y, Köhl J. Distinct roles of the anaphylatoxins C3a and C5a in dendritic cell-mediated allergic asthma. THE JOURNAL OF IMMUNOLOGY 2014; 193:5387-401. [PMID: 25355927 DOI: 10.4049/jimmunol.1400080] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conventional dendritic cells (cDC) are necessary and sufficient to drive mixed maladaptive Th2/Th17 immune responses toward aeroallergens in experimental allergy models. Previous studies suggest that the anaphylatoxin C3a promotes, whereas C5a protects from the development of maladaptive immunity during allergen sensitization. However, only limited evidence exists that such effects are directly mediated through anaphylatoxin-receptor signaling in cDCs. In this study, we assessed the impact of C3a and C5a on cDC-mediated induction pulmonary allergy by adoptively transferring house dust mite (HDM)-pulsed bone marrow-derived DCs (BMDC) from wild-type (WT) C3aR(-/-), C5aR1(-/-), or C3aR(-/-)/C5aR1(-/-) into WT mice. Transfer of HDM-pulsed WT BMDCs promoted a strong asthmatic phenotype characterized by marked airway resistance, strong Th2 cytokine, and mucus production, as well as mixed eosinophilic and neurophilic airway inflammation. Surprisingly, C3aR(-/-) cDCs induced a strong allergic phenotype, but no IL-17A production, whereas HDM-pulsed C5aR1(-/-) cDCs failed to drive pulmonary allergy. Transfer of C3aR(-/-)/C5aR1(-/-) cDCs resulted in a slightly reduced allergic phenotype associated with increased IFN-γ production. Mechanistically, C3aR and C5aR1 signaling is required for IL-23 production from HDM-pulsed BMDCs in vitro. Furthermore, C3aR(-/-) BMDCs produced less IL-1β. The mechanisms underlying the failure of C5aR1(-/-) BMDCs to induce experimental allergy include a reduced capability to migrate into the lung tissue and a decreased potency to direct pulmonary homing of effector T cells. Thus, we uncovered a crucial role for C5a, but only a minor role for C3a in BMDC-mediated pulmonary allergy, suggesting that BMDCs inappropriately reflect the impact of complement on lung cDC-mediated allergic asthma development.
Collapse
Affiliation(s)
- Carsten Engelke
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | - Heike A Ströver
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany
| | | | - Peter König
- Institute for Anatomy, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany; and
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany;
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck and Airway Research Center North, member of the German Center for Lung Research, 23538 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| |
Collapse
|
32
|
Schmitz V, Almeida LN, Svensjö E, Monteiro AC, Köhl J, Scharfstein J. C5a and Bradykinin Receptor Cross-Talk Regulates Innate and Adaptive Immunity inTrypanosoma cruziInfection. THE JOURNAL OF IMMUNOLOGY 2014; 193:3613-23. [DOI: 10.4049/jimmunol.1302417] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
33
|
Temesi G, Virág V, Hadadi E, Ungvári I, Fodor LE, Bikov A, Nagy A, Gálffy G, Tamási L, Horváth I, Kiss A, Hullám G, Gézsi A, Sárközy P, Antal P, Buzás E, Szalai C. Novel genes in Human Asthma Based on a Mouse Model of Allergic Airway Inflammation and Human Investigations. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2014; 6:496-503. [PMID: 25374748 PMCID: PMC4214969 DOI: 10.4168/aair.2014.6.6.496] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/10/2014] [Accepted: 04/14/2014] [Indexed: 12/03/2022]
Abstract
Purpose Based on a previous gene expression study in a mouse model of asthma, we selected 60 candidate genes and investigated their possible roles in human asthma. Methods In these candidate genes, 90 SNPs were genotyped using MassARRAY technology from 311 asthmatic children and 360 healthy controls of the Hungarian (Caucasian) population. Moreover, gene expression levels were measured by RT PCR in the induced sputum of 13 asthmatics and 10 control individuals. t-tests, chi-square tests, and logistic regression were carried out in order to assess associations of SNP frequency and expression level with asthma. Permutation tests were performed to account for multiple hypothesis testing. Results The frequency of 4 SNPs in 2 genes differed significantly between asthmatic and control subjects: SNPs rs2240572, rs2240571, rs3735222 in gene SCIN, and rs32588 in gene PPARGC1B. Carriers of the minor alleles had reduced risk of asthma with an odds ratio of 0.64 (0.51-0.80; P=7×10-5) in SCIN and 0.56 (0.42-0.76; P=1.2×10-4) in PPARGC1B. The expression levels of SCIN, PPARGC1B and ITLN1 genes were significantly lower in the sputum of asthmatics. Conclusions Three potentially novel asthma-associated genes were identified based on mouse experiments and human studies.
Collapse
Affiliation(s)
- Gergely Temesi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Viktor Virág
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Eva Hadadi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary. ; Department of Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - Ildikó Ungvári
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Lili E Fodor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - András Bikov
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | | | - Gabriella Gálffy
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Lilla Tamási
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary. ; Ministry of National Resources, Budapest, Hungary
| | - András Kiss
- Heim, Pal Children Hospital, Budapest, Hungary
| | - Gábor Hullám
- Department of Measurement and Information Systems, University of Technology and Economics, Budapest, Hungary
| | - András Gézsi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Péter Sárközy
- Department of Measurement and Information Systems, University of Technology and Economics, Budapest, Hungary
| | - Péter Antal
- Department of Measurement and Information Systems, University of Technology and Economics, Budapest, Hungary
| | - Edit Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Csaba Szalai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary. ; Heim, Pal Children Hospital, Budapest, Hungary. ; Csertex Research Laboratory, Budapest, Hungary
| |
Collapse
|
34
|
Abstract
Allergic asthma is a complex disease characterized in part by granulocytic inflammation of the airways. In addition to eosinophils, neutrophils (PMN) are also present, particularly in cases of severe asthma. We sought to identify the genetic determinants of neutrophilic inflammation in a mouse model of house dust mite (HDM)-induced asthma. We applied an HDM model of allergic asthma to the eight founder strains of the Collaborative Cross (CC) and 151 incipient lines of the CC (preCC). Lung lavage fluid was analyzed for PMN count and the concentration of CXCL1, a hallmark PMN chemokine. PMN and CXCL1 were strongly correlated in preCC mice. We used quantitative trait locus (QTL) mapping to identify three variants affecting PMN, one of which colocalized with a QTL for CXCL1 on chromosome (Chr) 7. We used lung eQTL data to implicate a variant in the gene Zfp30 in the CXCL1/PMN response. This genetic variant regulates both CXCL1 and PMN by altering Zfp30 expression, and we model the relationships between the QTL and these three endophenotypes. We show that Zfp30 is expressed in airway epithelia in the normal mouse lung and that altering Zfp30 expression in vitro affects CXCL1 responses to an immune stimulus. Our results provide strong evidence that Zfp30 is a novel regulator of neutrophilic airway inflammation.
Collapse
|
35
|
Xu GL, Chen J, Yang F, Li GQ, Zheng LX, Wu YZ. C5a/C5aR pathway is essential for the pathogenesis of murine viral fulminant hepatitis by way of potentiating Fgl2/fibroleukin expression. Hepatology 2014; 60:114-24. [PMID: 24604562 DOI: 10.1002/hep.27114] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 03/03/2014] [Indexed: 12/17/2022]
Abstract
UNLABELLED Viral fulminant hepatitis (FH) remains a serious clinical problem with very high mortality. Lacking understanding of FH pathogenesis has in essence hindered efficient clinical treatment. Inferring from a correlation observed between the genetic differences in the complement component 5 (C5) and the susceptibility of mouse strains to murine hepatitis virus strain-3 (MHV-3) infections, we propose that excessive complement activation plays a critical role in the development of FH. We show that MHV-3 infection causes massive complement activation, along with a rapid increase in serum C5a levels and quick development of FH in susceptible strains. Mice deficient in the C5a receptor (C5aR) or the susceptible strains treated with C5aR antagonists (C5aRa) exhibit significant attenuation of the disease, accompanied by a remarkable reduction of hepatic fibrinogen-like protein 2 (Fgl2), a hallmark protein that causes necrosis of infected livers. In accordance, biopsy of FH patients shows a dramatic increase of Fgl2 expression, which correlates with C5aR up-regulation in the liver. In vitro C5a administration accelerates MHV-3-induced Fgl2 secretion by macrophages. Furthermore, inhibiting ERK1/2 and p38 efficiently blocks C5a-mediated Fgl2 production during viral infections. CONCLUSION These data provide evidence that mouse susceptibility to MHV-3-induced FH may rely on C5a/C5aR interactions, for which ERK1/2 and p38 pathways participate in up-regulating Fgl2 expression. Inhibition of C5a/C5aR interactions is expected to be beneficial in the clinical treatment of FH patients.
Collapse
Affiliation(s)
- Gui-lian Xu
- Institute of Immunology, PLA, College of Basic Medical Sciences, Third Military Medical University, Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
36
|
Treatment with the C5a receptor/CD88 antagonist PMX205 reduces inflammation in a murine model of allergic asthma. Int Immunopharmacol 2014; 21:293-300. [PMID: 24859057 DOI: 10.1016/j.intimp.2014.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 02/03/2023]
Abstract
Allergic asthma is a chronic inflammatory airway disease arising from an aberrant immune response following exposure to environmental stimuli in genetically susceptible persons. The complement component 5 (C5)/C5a Receptor (C5aR/CD88) signaling pathway has been implicated in both experimental allergic asthma and human asthmatic disease. Targeting the C5a/C5aR signaling pathway in rodent models has been shown to either enhance or reduce allergic asthma consequences. Treatment with a recombinant humanized monoclonal antibody directed against C5 has shown unclear results in patients with asthma. The objective of this proof-of-concept animal study was to determine whether the low molecular weight C5aR peptidomimetic antagonist, PMX205, would reduce experimental allergic asthma consequences in mice. PMX205 or vehicle control was administered subcutaneously to BALB/c mice prior to and during standard ovalbumin (OVA) allergen sensitization and aerosolized challenge phases. PMX205 substantially reduced OVA-induced total cell (60%), neutrophil (66%) and eosinophil (65%) influxes in lavage fluid sampling. There were also significant reductions in OVA-induced lavage fluid IL-13 protein and lung Th2 cytokine gene expression with PMX205 administration. PMX205 treatment also diminished OVA-induced lung parenchyma cellular infiltration. PMX205 administration did not reduce OVA-induced serum IgE levels or epithelial mucous/goblet cell generation. There was no evidence of toxicity observed with PMX205 treatment in saline or OVA-challenged animals. These data provide evidence that pharmacologic blockade of C5aR by a low molecular weight antagonist (PMX205) reduces airway inflammatory cell and cytokine responses in experimental allergic asthma, and suggests that PMX205 might represent a novel therapeutic agent for reducing asthmatic outcomes.
Collapse
|
37
|
Sayegh ET, Bloch O, Parsa AT. Complement anaphylatoxins as immune regulators in cancer. Cancer Med 2014; 3:747-58. [PMID: 24711204 PMCID: PMC4303144 DOI: 10.1002/cam4.241] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/10/2014] [Accepted: 02/26/2014] [Indexed: 12/31/2022] Open
Abstract
The role of the complement system in innate immunity is well characterized. However, a recent body of research implicates the complement anaphylatoxins C3a and C5a as insidious propagators of tumor growth and progression. It is now recognized that certain tumors elaborate C3a and C5a and that complement, as a mediator of chronic inflammation and regulator of immune function, may in fact foster rather than defend against tumor growth. A putative mechanism for this function is complement-mediated suppression of immune effector cells responsible for immunosurveillance within the tumor microenvironment. This paradigm accords with models of immune dysregulation, such as autoimmunity and infectious disease, which have defined a pathophysiological role for abnormal complement signaling. Several types of immune cells express the cognate receptors for the complement anaphylatoxins, C3aR and C5aR, and demonstrate functional modulation in response to complement stimulation. In turn, impairment of antitumor immunity has been intimately tied to tumor progression in animal models of cancer. In this article, the literature was systematically reviewed to identify studies that have characterized the effects of the complement anaphylatoxins on the composition and function of immune cells within the tumor microenvironment. The search identified six studies based upon models of lymphoma and ovarian, cervical, lung, breast, and mammary cancer, which collectively support the paradigm of complement as an immune regulator in the tumor microenvironment.
Collapse
Affiliation(s)
- Eli T Sayegh
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | |
Collapse
|
38
|
Boussommier-Calleja A, Overby DR. The influence of genetic background on conventional outflow facility in mice. Invest Ophthalmol Vis Sci 2013; 54:8251-8. [PMID: 24235015 DOI: 10.1167/iovs.13-13025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Intraocular pressure (IOP) varies between genetically distinct strains of mice. The purpose was to test the hypothesis that strain-dependent differences in IOP are attributable to differences in conventional outflow facility (C). METHODS The IOP was measured by rebound tonometry in conscious or anesthetized BALB/cJ, C57BL/6J, and CBA/J mice (N = 6-10 per strain). Conventional outflow facility was measured by ex vivo perfusion of enucleated eyes (N = 9-10 per strain). RESULTS Conscious IOP varied between strains, being highest in CBA/J (14.5 ± 0.9 mm Hg, mean ± SD), intermediate in C57BL/6J (12.3 ± 1.0 mm Hg), and lowest in BALB/cJ (10.6 ± 1.8 mm Hg) mice. Anesthesia reduced IOP and eliminated any detectable differences between strains. Conventional outflow facility also varied between strains, but, in contrast to IOP, C was lowest in CBA/J (0.0113 ± 0.0031 μL/min/mm Hg) and highest in BALB/cJ (0.0164 ± 0.0059 μL/min/mm Hg). Like IOP, C was intermediate in C57BL/6J (0.0147 ± 0.0029 μL/min/mm Hg). There was a strong correlation between conscious IOP and outflow resistance (1/C) from individual eyes across all three strains, revealing that 70% of the variation in IOP was attributable to variation in outflow resistance. CONCLUSIONS Differences in IOP among three genetically distinct murine strains are attributable largely to differences in conventional outflow facility. These results motivate further studies using mice to identify the morphologic and genetic factors that underlie IOP regulation within the conventional outflow pathway.
Collapse
|
39
|
Seow V, Lim J, Iyer A, Suen JY, Ariffin JK, Hohenhaus DM, Sweet MJ, Fairlie DP. Inflammatory responses induced by lipopolysaccharide are amplified in primary human monocytes but suppressed in macrophages by complement protein C5a. THE JOURNAL OF IMMUNOLOGY 2013; 191:4308-16. [PMID: 24043889 DOI: 10.4049/jimmunol.1301355] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Monocytes and macrophages are important innate immune cells equipped with danger-sensing receptors, including complement and Toll-like receptors. Complement protein C5a, acting via C5aR, is shown in this study to differentially modulate LPS-induced inflammatory responses in primary human monocytes versus macrophages. Whereas C5a enhanced secretion of LPS-induced IL-6 and TNF from primary human monocytes, C5a inhibited these responses while increasing IL-10 secretion in donor-matched human monocyte-derived macrophages differentiated by GM-CSF or M-CSF. Gαi/c-Raf/MEK/ERK signaling induced by C5a was amplified in macrophages but not in monocytes by LPS. Accordingly, the Gαi inhibitor pertussis toxin and MEK inhibitor U0126 blocked C5a inhibition of LPS-induced IL-6 and TNF production from macrophages. This synergy was independent of IL-10, PI3K, p38, JNK, and the differentiating agent. Furthermore, C5a did not inhibit IL-6 production from macrophages induced by other TLR agonists that are selective for Toll/IL-1R domain-containing adapter inducing IFN-β (polyinosinic-polycytidylic acid) or MyD88 (imiquimod), demonstrating selectivity for C5a regulation of LPS responses. Finally, suppression of proinflammatory cytokines IL-6 and TNF in macrophages did not compromise antimicrobial activity; instead, C5a enhanced clearance of the Gram-negative bacterial pathogen Salmonella enterica serovar Typhimurium from macrophages. C5aR is thus a regulatory switch that modulates TLR4 signaling via the Gαi/c-Raf/MEK/ERK signaling axis in human macrophages but not monocytes. The differential effects of C5a are consistent with amplifying monocyte proinflammatory responses to systemic danger signals, but attenuating macrophage cytokine responses (without compromising microbicidal activity), thereby restraining inflammatory responses to localized infections.
Collapse
Affiliation(s)
- Vernon Seow
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
40
|
C5a receptor signalling in dendritic cells controls the development of maladaptive Th2 and Th17 immunity in experimental allergic asthma. Mucosal Immunol 2013; 6:807-25. [PMID: 23212198 DOI: 10.1038/mi.2012.119] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The pathways underlying dendritic cell (DC) activation in allergic asthma are incompletely understood. Here we demonstrate that adoptive transfer of ovalbumin-pulsed wild-type (wt) but not of C5a receptor-deficient (C5aR⁻/⁻) bone marrow (BM)-derived DCs (BMDCs) induced mixed T helper type 2 (Th2)/Th17 maladaptive immunity, associated with severe airway hyperresponsiveness, mucus production, and mixed eosinophilic/neutrophilic inflammation. Mechanistically, antigen uptake, processing, and CD11b expression were reduced in C5aR⁻/⁻ BMDCs. Further, interleukin (IL)-1β, -6, and -23 production were impaired resulting in reduced Th17 cell differentiation, associated with accelerated activated T-cell death in vitro and in vivo. Surprisingly, we found an increased frequency of CD11b(hi)CD11c(int)Gr1⁺F4/80⁺ cells, expressing arginase and nitric oxide synthase in C5aR⁻/⁻ BM preparations. Intratracheal administration of ovalbumin-pulsed wt DCs and sorted CD11b(hi)CD11c(int)Gr1⁺F4/80⁺ C5aR⁻/⁻ cells reduced Th2 immune responses in vivo. Together, we uncover novel roles for C5aR in Th17 differentiation, T-cell survival, and differentiation of a DC-suppressor population controlling Th2 immunity in experimental allergic asthma.
Collapse
|
41
|
Kemper C, Köhl J. Novel roles for complement receptors in T cell regulation and beyond. Mol Immunol 2013; 56:181-90. [PMID: 23796748 DOI: 10.1016/j.molimm.2013.05.223] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 05/20/2013] [Indexed: 12/16/2022]
Abstract
Complement receptors are expressed on cells of the innate and the adaptive immune system. They play important roles in pathogen and danger sensing as they translate the information gathered by complement fluid phase sensors into cellular responses. Further, they control complement activation on viable and apoptotic host cells, clearance of immune complexes and mediate opsonophagocytosis. More recently, evidence has accumulated that complement receptors form a complex network with other innate receptors systems such as the Toll-like receptors, the Notch signaling system, IgG Fc receptors and C-type lectin receptors contributing to the benefit and burden of innate and adaptive immune responses in autoimmune and allergic diseases as well as in cancer and transplantation. Here, we will discuss recent developments and emerging concepts of complement receptor activation and regulation with a particular focus on the differentiation, maintenance and contraction of effector and regulatory T cells.
Collapse
Affiliation(s)
- Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.
| | | |
Collapse
|
42
|
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
|
43
|
Abstract
Immunotoxicity can take the form of an enhanced immune response or hypersensitivity. Asthma is one possible consequence of hypersensitivity in the lung, with characteristics that include reversible airway obstruction, eosinophil infiltration into the lung, and airway hyperresponsiveness to agonists such as methacholine. In toxicology, two primary areas of investigation prompt the measurement of the asthmatic response in an animal: (1) identification of chemicals or proteins that cause asthma, i.e., respiratory allergens, and (2) identification of exposures that will exacerbate existing asthma. An ovalbumin-induced asthma model can be used to identify exposures that exacerbate existing asthma. A protocol for the sensitization and challenge of mice with ovalbumin is described; it leads to the asthma symptoms of airway hyperresponsiveness and eosinophil infiltration. Assessment of airway hyperresponsiveness to methacholine uses whole body plethysmography in conscious unrestrained mice. Bronchoalveolar lavage of the mouse determines the extent of cellular infiltration into the airspace. Removal of lung lobes and assay of eosinophil peroxidase and myeloperoxidase provides a measure of the numbers of eosinophils and neutrophils, respectively, in the lung. Depending on the experimental goals, bronchoalveolar lavage fluid and lung tissue can also be used for isolation of RNA, and measurement of cytokines, chemokines, antibodies, and inflammatory mediators.
Collapse
Affiliation(s)
- Jean F Regal
- School of Medicine, University of Minnesota, Duluth, MN, USA
| |
Collapse
|
44
|
Myers JM, Fairweather D, Huber SA, Cunningham MW. Autoimmune myocarditis, valvulitis, and cardiomyopathy. CURRENT PROTOCOLS IN IMMUNOLOGY 2013; Chapter 15:Unit 15.14.1-51. [PMID: 23564686 PMCID: PMC3672855 DOI: 10.1002/0471142735.im1514s101] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Myocarditis and valvulitis are inflammatory diseases affecting myocardium and valve. Myocarditis, a viral-induced disease of myocardium, may lead to dilated cardiomyopathy and loss of heart function. Valvulitis leads to deformed heart valves and altered blood flow in rheumatic heart disease. Animal models recapitulating these diseases are important in understanding the human condition. Cardiac myosin is a major autoantigen in heart, and antibodies and T cells to cardiac myosin are evident in inflammatory heart diseases. This unit is a practical guide to induction and evaluation of experimental autoimmune myocarditis (EAM) in several mouse strains and the Lewis rat. Purification protocols for cardiac myosin and protocols for induction of EAM by cardiac myosin and its myocarditis-producing peptides, and coxsackievirus CVB3, are defined. Protocols for assessment of myocarditis and valvulitis in humans and animal models provide methods to define functional autoantibodies targeting cardiac myosin, β-adrenergic, and muscarinic receptors, and their deposition in tissues.
Collapse
Affiliation(s)
- Jennifer M Myers
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | | | | |
Collapse
|
45
|
Brenner M, Gulko PS. The arthritis severity locus Cia5a regulates the expression of inflammatory mediators including Syk pathway genes and proteases in pristane-induced arthritis. BMC Genomics 2012; 13:710. [PMID: 23249408 PMCID: PMC3548698 DOI: 10.1186/1471-2164-13-710] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 12/07/2012] [Indexed: 02/02/2023] Open
Abstract
Background Cia5a is a locus on rat chromosome 10 that regulates disease severity and joint damage in two models of rheumatoid arthritis, collagen- and pristane-induced arthritis (PIA). In this study, we aimed to identify cellular and molecular processes regulated by Cia5a using microarray-based gene expression analysis of synovial tissues from MHC identical DA (severe erosive disease) and DA.F344(Cia5a) congenics (mild non-erosive disease) rats. Results Synovial tissues from six DA and eight DA.F344(Cia5a) rats were analyzed 21 days after the induction of PIA using the Illumina RatRef-12 BeadChip (21,922 genes) and selected data confirmed with qPCR. There was a significantly increased expression of pro-inflammatory mediators such as Il1b (5-fold), Il18 (3.9-fold), Cxcl1 (10-fold), Cxcl13 (7.5-fold) and Ccl7 (7.9-fold), and proteases like Mmp3 (23-fold), Mmp9 (32-fold), Mmp14 (4.4-fold) and cathepsins in synovial tissues from DA, with reciprocally reduced levels in congenics. mRNA levels of 47 members of the Spleen Tyrosine Kinase (Syk) pathway were significantly increased in DA synovial tissues compared with DA.F344(Cia5a), and included Syk (5.4-fold), Syk-activating receptors and interacting proteins, and genes regulated by Syk such as NFkB, and NAPDH oxidase complex genes. Nuclear receptors (NR) such as Rxrg, Pparg and Rev-erba were increased in the protected congenics, and so was the anti-inflammatory NR-target gene Scd1 (54-fold increase). Tnn (72-fold decrease) was the gene most significantly increased in DA. Conclusions Analyses of gene expression in synovial tissues revealed that the arthritis severity locus Cia5a regulates the expression of key mediators of inflammation and joint damage, as well as the expression of members of the Syk pathway. This expression pattern correlates with disease severity and joint damage and along with the gene accounting for Cia5a could become a useful biomarker to identify patients at increased risk for severe and erosive disease. The identification of the gene accounting for Cia5a has the potential to generate a new and important target for therapy and prognosis.
Collapse
Affiliation(s)
- Max Brenner
- Laboratory of Experimental Rheumatology, Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive Room 1240, Manhasset, NY 11030, USA
| | | |
Collapse
|
46
|
Lionikas A, Meharg C, Derry JM, Ratkevicius A, Carroll AM, Vandenbergh DJ, Blizard DA. Resolving candidate genes of mouse skeletal muscle QTL via RNA-Seq and expression network analyses. BMC Genomics 2012; 13:592. [PMID: 23126637 PMCID: PMC3505184 DOI: 10.1186/1471-2164-13-592] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 10/26/2012] [Indexed: 11/28/2022] Open
Abstract
Background We have recently identified a number of Quantitative Trait Loci (QTL) contributing to the 2-fold muscle weight difference between the LG/J and SM/J mouse strains and refined their confidence intervals. To facilitate nomination of the candidate genes responsible for these differences we examined the transcriptome of the tibialis anterior (TA) muscle of each strain by RNA-Seq. Results 13,726 genes were expressed in mouse skeletal muscle. Intersection of a set of 1061 differentially expressed transcripts with a mouse muscle Bayesian Network identified a coherent set of differentially expressed genes that we term the LG/J and SM/J Regulatory Network (LSRN). The integration of the QTL, transcriptome and the network analyses identified eight key drivers of the LSRN (Kdr, Plbd1, Mgp, Fah, Prss23, 2310014F06Rik, Grtp1, Stk10) residing within five QTL regions, which were either polymorphic or differentially expressed between the two strains and are strong candidates for quantitative trait genes (QTGs) underlying muscle mass. The insight gained from network analysis including the ability to make testable predictions is illustrated by annotating the LSRN with knowledge-based signatures and showing that the SM/J state of the network corresponds to a more oxidative state. We validated this prediction by NADH tetrazolium reductase staining in the TA muscle revealing higher oxidative potential of the SM/J compared to the LG/J strain (p<0.03). Conclusion Thus, integration of fine resolution QTL mapping, RNA-Seq transcriptome information and mouse muscle Bayesian Network analysis provides a novel and unbiased strategy for nomination of muscle QTGs.
Collapse
Affiliation(s)
- Arimantas Lionikas
- School of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| | | | | | | | | | | | | |
Collapse
|
47
|
Regal JF, Greene AL, Regal RR. Mechanisms of occupational asthma: Not all allergens are equal. Environ Health Prev Med 2012; 12:165-71. [PMID: 21432060 DOI: 10.1007/bf02897986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 03/09/2007] [Indexed: 01/17/2023] Open
Abstract
Asthma is a heterogeneous lung disorder characterized by airway obstruction, inflammation and eosinophil infiltration into the lung. Both genetics and environmental factors influence the expression of asthma, and not all asthma is the result of a specific immune response to allergen. Numerous asthma phenotypes have been described, including occupational asthma, and therapeutic strategies for asthma control are similar regardless of phenotype. We hypothesized that mechanistic pathways leading to asthma symptoms in the effector phase of the disorder differ with the inciting allergen. Since route of allergen exposure can influence mechanistic pathways, mice were sensitized by identical routes with a high molecular weight occupational allergen ovalbumin and a low molecular weight occupational allergen trimellitic anhydride (TMA). Different statistical methods with varying selection criteria resulted in identification of similar candidate genes. Array data are intended to provide candidate genes for hypothesis generation and further experimentation. Continued studies focused on genes showing minimal changes in the TMA-induced model but with clear up-regulation in the ovalbumin model. Two of these genes, arginase 1 and eotaxin 1 are the focus of continuing investigations in mouse models of asthma regarding differences in mechanistic pathways depending on the allergen. Microarray data from the ovalbumin and TMA model of asthma were also compared to previous data usingAspergillus as allergen to identify putative asthma 'signature genes', i.e. genes up-regulated with all 3 allergens. Array studies provide candidate genes to identify common mechanistic pathways in the effector phase, as well as mechanistic pathways unique to individual allergens.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Biochemistry & Molecular Biology, University of Minnesota Medical School Duluth, 1035 University Drive, 55812, Duluth, MN, USA,
| | | | | |
Collapse
|
48
|
Regal JF. Immunologic effector mechanisms in animal models of occupational asthma. J Immunotoxicol 2012; 1:25-37. [PMID: 18958638 DOI: 10.1080/15476910490438351] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Occupational asthma is a form of immunotoxicity resulting from an exaggerated immune response to substances encountered in the workplace. Symptoms include reversible airway obstruction, airway hyperresponsiveness, airway remodeling, mucus production and cellular infiltration into the lung, particularly eosinophilia. The asthmatic response is divided into the induction phase, occurring after initial exposure to allergen, followed by the effector phase where a subsequent exposure to the allergen results in the respiratory symptoms. Animal models have been used to investigate the asthmatic response and this review will focus on mechanistic studies of the effector phase. Variables that may impact the effector phase include strain and species of animal, dose of allergen, route of exposure, and developmental stage of the animal. Both trimellitic anhydride (TMA) and ovalbumin are known causes of occupational asthma. Ovalbumin is also a reference protein allergen in immunology, and TMA is used as a prototype of a low molecular weight respiratory allergen. Differences in effector mechanisms for TMA and ovalbumin have been noted in different animal models. Studies in the guinea pig provide the most direct comparisons of effector mechanisms of TMA and ovalbumin, with differences in the role of the complement system and arachidonate metabolites being noted. Besides the guinea pig, the Brown Norway rat, and various mouse strains provide useful asthma models for TMA and ovalbumin. However, studies of effector mechanisms are somewhat lacking in either of these species using TMA as the allergen. Continued studies are indicated to determine if unique effector mechanisms can be identified for the many different causes of occupational asthma.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Pharmacology, School of Medicine, University of Minnesota, Duluth, Minnesota, USA
| |
Collapse
|
49
|
Moghimi SM, Wibroe PP, Helvig SY, Farhangrazi ZS, Hunter AC. Genomic perspectives in inter-individual adverse responses following nanomedicine administration: The way forward. Adv Drug Deliv Rev 2012; 64:1385-93. [PMID: 22634158 DOI: 10.1016/j.addr.2012.05.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 05/17/2012] [Indexed: 01/18/2023]
Abstract
The underlying mechanism of intravenous infusion-related adverse reactions inherent to regulatory-approved nanomedicines still remains elusive. There are substantial inter-individual differences in observed adverse reactions, which may include cardiovascular, broncho-pulmonary, muco-cutaneous, neuro-psychosomatic and autonomic manifestations. Although nanomedicine-mediated triggering of complement activation has been suggested to be a significant contributing factor to these adverse events, complement activation may still proceed in non-responders. Whether these reactions share similar immunological mechanisms and underpinning genetic factors with drug hypersensitivity syndrome remains to be investigated. Genetic association studies could be a powerful tool to dissect causative factors and reveal the multiple molecular pathways that induce infusion related adverse reactions. It is envisaged that such research may lead to the design of reliable in vitro profiling tests for risk assessment and treatment decisions, thereby revolutionizing the practice of medicine with nanopharmaceuticals. Such procedures may further improve regulatory approval processes for nanomedicines currently in the pipeline and decrease the overall cost of health care. Here we discuss some key innate immunity genes and their polymorphisms in relation to nanomedicine infusion-mediated symptomatic responses.
Collapse
Affiliation(s)
- S Moein Moghimi
- Centre for Pharmaceutical Nanotechnology and Nanotoxicology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark.
| | | | | | | | | |
Collapse
|
50
|
Wright FA, Shabalin AA, Rusyn I. Computational tools for discovery and interpretation of expression quantitative trait loci. Pharmacogenomics 2012; 13:343-52. [PMID: 22304583 DOI: 10.2217/pgs.11.185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Expression quantitative trait locus (eQTL) analysis is rapidly moving from a cutting-edge concept in genomics to a mature area of investigation, with important connections to genome-wide association studies for human disease, pharmacogenomics and toxicogenomics. Despite the importance of the topic, many investigators must develop their own code or use tools not specifically suited for eQTL analysis. Convenient computational tools are becoming available, but they are not widely publicized, and investigators who are interested in discovery or eQTL, or in using them to interpret genome-wide association study results may have difficulty navigating the available resources. The purpose of this review is to help investigators find appropriate programs for eQTL analysis and interpretation.
Collapse
Affiliation(s)
- Fred A Wright
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|