1
|
Magnusen AF, Pandey MK. Complement System and Adhesion Molecule Skirmishes in Fabry Disease: Insights into Pathogenesis and Disease Mechanisms. Int J Mol Sci 2024; 25:12252. [PMID: 39596318 PMCID: PMC11594573 DOI: 10.3390/ijms252212252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal storage disorder caused by mutations in the galactosidase alpha (GLA) gene, resulting in the accumulation of globotriaosylceramide (Gb3) and its deacetylated form, globotriaosylsphingosine (Lyso-Gb3) in various tissues and fluids throughout the body. This pathological accumulation triggers a cascade of processes involving immune dysregulation and complement system activation. Elevated levels of complement 3a (C3a), C5a, and their precursor C3 are observed in the plasma, serum, and tissues of patients with Fabry disease, correlating with significant endothelial cell abnormalities and vascular dysfunction. This review elucidates how the complement system, particularly through the activation of C3a and C5a, exacerbates disease pathology. The activation of these pathways leads to the upregulation of adhesion molecules, including vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), platelet and endothelial cell adhesion molecule 1 (PECAM1), and complement receptor 3 (CR3) on leukocytes and endothelial cells. This upregulation promotes the excessive recruitment of leukocytes, which in turn exacerbates disease pathology. Targeting complement components C3a, C5a, or their respective receptors, C3aR (C3a receptor) and C5aR1 (C5a receptor 1), could potentially reduce inflammation, mitigate tissue damage, and improve clinical outcomes for individuals with Fabry disease.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
2
|
Vygonskaya M, Wu Y, Price TJ, Chen Z, Smith MT, Klyne DM, Han FY. The role and treatment potential of the complement pathway in chronic pain. THE JOURNAL OF PAIN 2024:104689. [PMID: 39362355 DOI: 10.1016/j.jpain.2024.104689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The role of the complement system in pain syndromes has garnered attention on the back of preclinical and clinical evidence supporting its potential as a target for new analgesic pharmacotherapies. Of the components that make up the complement system, component 5a (C5a) and component 3a (C3a) are most strongly and consistently associated with pain. Receptors for C5a are widely found in immune resident cells (microglia, astrocytes, sensory neuron-associated macrophages (sNAMs)) in the central nervous system (CNS) as well as hematogenous immune cells (mast cells, macrophages, T-lymphocytes, etc.). When active, as is often observed in chronic pain conditions, these cells produce various inflammatory mediators including pro-inflammatory cytokines. These events can trigger nervous tissue inflammation (neuroinflammation) which coexists with and potentially maintains peripheral and central sensitization. C5a has a likely critical role in initiating this process highlighting its potential as a promising non-opioid target for treating pain. This review summarizes the most up-to-date research on the role of the complement system in pain with emphasis on the C5 pathway in peripheral tissue, dorsal root ganglia (DRG) and the CNS, and explores advances in complement-targeted drug development and sex differences. A perspective on the optimal application of different C5a inhibitors for different types (e.g., neuropathic, post-surgical and chemotherapy-induced pain, osteoarthritis pain) and stages (e.g., acute, subacute, chronic) of pain is also provided to help guide future clinical trials. PERSPECTIVE: This review highlights the role and mechanisms of complement components and their receptors in physiological and pathological pain. The potential of complement-targeted therapeutics for the treatment of chronic pain is also explored with a focus on C5a inhibitors to help guide future clinical trials.
Collapse
Affiliation(s)
- Marina Vygonskaya
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Youzhi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Theodore J Price
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhuo Chen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David M Klyne
- NHMRC Centre of Clinical Research Excellence in Spinal Pain, Injury and Health, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Felicity Y Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
3
|
Dahmani M, Zhu JC, Cook JH, Riley SP. Anaphylatoxin signaling activates macrophages to control intracellular Rickettsia proliferation. Microbiol Spectr 2023; 11:e0253823. [PMID: 37855623 PMCID: PMC10714731 DOI: 10.1128/spectrum.02538-23] [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: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Pathogenic Rickettsia species are extremely dangerous bacteria that grow within the cytoplasm of host mammalian cells. In most cases, these bacteria are able to overpower the host cell and grow within the protected environment of the cytoplasm. However, a dramatic conflict occurs when Rickettsia encounter innate immune cells; the bacteria can "win" by taking over the host, or the bacteria can "lose" if the host cell efficiently fights the infection. This manuscript examines how the immune complement system is able to detect the presence of Rickettsia and alert nearby cells. Byproducts of complement activation called anaphylatoxins are signals that "activate" innate immune cells to mount an aggressive defensive strategy. This study enhances our collective understanding of the innate immune reaction to intracellular bacteria and will contribute to future efforts at controlling these dangerous infections.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jinyi C. Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jack H. Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Sean P. Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
4
|
Single-dose HPV vaccine immunity: is there a role for non-neutralizing antibodies? Trends Immunol 2022; 43:815-825. [PMID: 35995705 DOI: 10.1016/j.it.2022.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022]
Abstract
A single dose of human papillomavirus (HPV) vaccine against HPV infection (prerequisite for cervical cancer) appears to be as efficacious as two or three doses, despite inducing lower antibody titers. Neutralizing antibodies are thought to be the primary mediator of protection, but the threshold for protection is unknown. Antibody functions beyond neutralization have not been explored for HPV vaccines. Here, we discuss the immune mechanisms of HPV vaccines, with a focus on non-neutralizing antibody effector functions. In the context of single-dose HPV vaccination where antibody is limiting, we propose that non-neutralizing antibody functions may contribute to preventing HPV infection. Understanding the immunological basis of protection for single-dose HPV vaccination will provide a rationale for implementing single-dose HPV vaccine regimens.
Collapse
|
5
|
Zhang MF, Huang J, Zhang YM, Qu Z, Wang X, Wang F, Meng LQ, Cheng XY, Cui Z, Liu G, Zhao MH. Complement activation products in the circulation and urine of primary membranous nephropathy. BMC Nephrol 2019; 20:313. [PMID: 31399080 PMCID: PMC6688252 DOI: 10.1186/s12882-019-1509-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/31/2019] [Indexed: 12/19/2022] Open
Abstract
Background Complement activation plays a substantial role in the pathogenesis of primary membranous nephropathy (pMN). C5b-9, C3c, MBL, and factor B have been documented in the subepithelial immune deposits. However, the changing of complement activation products in circulation and urine is not clear. Methods We measured the circulating and urinary levels of C1q, MBL, C4d, Bb, properdin, C3a, C5a, and sC5b-9, in 134 patients with biopsy-proven pMN, by enzyme-linked immunosorbent assay. All the plasma values were corrected by eGFR and all the urinary values were corrected by urinary creatinine and urinary protein excretion. Anti-PLA2R antibodies were measured in all patients. Results The plasma complement activation products were elevated both in the patients with and without anti-PLA2R antibodies. C3a levels were remarkably increased in the circulation and urine, much higher than the elevated levels of C5a. C5b-9 was in normal range in plasma, but significantly higher in urine. The urinary C5a had a positive correlation with anti-PLA2R antibody levels and urinary protein. The plasma level of C4d was elevated, but C1q and MBL were comparable to healthy controls. Positive correlations were observed between plasma C4d/MBL and urinary protein, only in the patients with positive anti-PLA2R antibodies but not in those without. The plasma level of Bb was elevated and had positive correlation with urinary protein only in the patients without anti-PLA2R antibodies. Conclusion Complement activation products were remarkable increased in pMN and may serve as sensitive biomarkers of disease activity. The complement may be activated through lectin pathway with the existence of anti-PLA2R antibodies, while through alternative pathway in the absence of antibody.
Collapse
Affiliation(s)
- Mu-Fan Zhang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Jing Huang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Yi-Miao Zhang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Zhen Qu
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Xin Wang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Fang Wang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Li-Qiang Meng
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Xu-Yang Cheng
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Zhao Cui
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China. .,Institute of Nephrology, Peking University, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China.
| | - Gang Liu
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Ming-Hui Zhao
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Peking University, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
6
|
Berendam SJ, Koeppel AF, Godfrey NR, Rouhani SJ, Woods AN, Rodriguez AB, Peske JD, Cummings KL, Turner SD, Engelhard VH. Comparative Transcriptomic Analysis Identifies a Range of Immunologically Related Functional Elaborations of Lymph Node Associated Lymphatic and Blood Endothelial Cells. Front Immunol 2019; 10:816. [PMID: 31057546 PMCID: PMC6478037 DOI: 10.3389/fimmu.2019.00816] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/27/2019] [Indexed: 12/11/2022] Open
Abstract
Lymphatic and blood vessels are formed by specialized lymphatic endothelial cells (LEC) and blood endothelial cells (BEC), respectively. These endothelial populations not only form peripheral tissue vessels, but also critical supporting structures in secondary lymphoid organs, particularly the lymph node (LN). Lymph node LEC (LN-LEC) also have been shown to have important immunological functions that are not observed in LEC from tissue lymphatics. LN-LEC can maintain peripheral tolerance through direct presentation of self-antigen via MHC-I, leading to CD8 T cell deletion; and through transfer of self-antigen to dendritic cells for presentation via MHC-II, resulting in CD4 T cell anergy. LN-LEC also can capture and archive foreign antigens, transferring them to dendritic cells for maintenance of memory CD8 T cells. The molecular basis for these functional elaborations in LN-LEC remain largely unexplored, and it is also unclear whether blood endothelial cells in LN (LN-BEC) might express similar enhanced immunologic functionality. Here, we used RNA-Seq to compare the transcriptomic profiles of freshly isolated murine LEC and BEC from LN with one another and with freshly isolated LEC from the periphery (diaphragm). We show that LN-LEC, LN-BEC, and diaphragm LEC (D-LEC) are transcriptionally distinct from one another, demonstrating both lineage and tissue-specific functional specializations. Surprisingly, tissue microenvironment differences in gene expression profiles were more numerous than those determined by endothelial cell lineage specification. In this regard, both LN-localized endothelial cell populations show a variety of functional elaborations that suggest how they may function as antigen presenting cells, and also point to as yet unexplored roles in both positive and negative regulation of innate and adaptive immune responses. The present work has defined in depth gene expression differences that point to functional specializations of endothelial cell populations in different anatomical locations, but especially the LN. Beyond the analyses provided here, these data are a resource for future work to uncover mechanisms of endothelial cell functionality.
Collapse
Affiliation(s)
- Stella J. Berendam
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander F. Koeppel
- Department of Public Health Sciences and Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Nicole R. Godfrey
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sherin J. Rouhani
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Amber N. Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anthony B. Rodriguez
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - J. David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kara L. Cummings
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Stephen D. Turner
- Department of Public Health Sciences and Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Victor H. Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- *Correspondence: Victor H. Engelhard
| |
Collapse
|
7
|
Gour N, Smole U, Yong HM, Lewkowich IP, Yao N, Singh A, Gabrielson E, Wills-Karp M, Lajoie S. C3a is required for ILC2 function in allergic airway inflammation. Mucosal Immunol 2018; 11:1653-1662. [PMID: 30104625 PMCID: PMC6279480 DOI: 10.1038/s41385-018-0064-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/23/2018] [Indexed: 02/04/2023]
Abstract
Aberrant type 2 responses underlie the pathologies in allergic diseases like asthma, yet, our understanding of the mechanisms that drive them remains limited. Recent evidence suggests that dysregulated innate immune factors can perpetuate asthma pathogenesis. In susceptible individuals, allergen exposure triggers the activation of complement, a major arm of innate immunity, leading to the aberrant generation of the C3a anaphylatoxin. C3 and C3a have been shown to be important for the development of Th2 responses, yet remarkably, the mechanisms by which C3a regulates type 2 immunity are relatively unknown. We demonstrate a central role for C3a in driving type 2 innate lymphoid cells (ILC2)-mediated inflammation in response to allergen and IL-33. Our data suggests that ILC2 recruitment is C3a-dependent. Further, we show that ILC2s directly respond to C3a, promoting type 2 responses by specifically: (1) inducing IL-13 and granulocyte-macrophage colony-stimulating factor, whereas inhibiting IL-10 production from ILC2; and (2) enhancing their antigen-presenting capability during ILC-T-cell cross-talk. In summary, we identify a novel mechanism by which C3a can mediate aberrant type 2 responses to aeroallergen exposure, which involves a yet unrecognized cross-talk between two major innate immune components-complement and group 2 innate lymphoid cells.
Collapse
Affiliation(s)
- Naina Gour
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD,Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD
| | - Ursula Smole
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD,Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hwan-Mee Yong
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Ian P. Lewkowich
- Department of Immunobiology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH
| | - Nu Yao
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Anju Singh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Stephane Lajoie
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
8
|
Thomas MA, Kleist AB, Volkman BF. Decoding the chemotactic signal. J Leukoc Biol 2018; 104:359-374. [PMID: 29873835 PMCID: PMC6099250 DOI: 10.1002/jlb.1mr0218-044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/25/2018] [Indexed: 12/20/2022] Open
Abstract
From an individual bacterium to the cells that compose the human immune system, cellular chemotaxis plays a fundamental role in allowing cells to navigate, interpret, and respond to their environments. While many features of cellular chemotaxis are shared among systems as diverse as bacteria and human immune cells, the machinery that guides the migration of these model organisms varies widely. In this article, we review current literature on the diversity of chemoattractant ligands, the cell surface receptors that detect and process chemotactic gradients, and the link between signal recognition and the regulation of cellular machinery that allow for efficient directed cellular movement. These facets of cellular chemotaxis are compared among E. coli, Dictyostelium discoideum, and mammalian neutrophils to derive organizational principles by which diverse cell systems sense and respond to chemotactic gradients to initiate cellular migration.
Collapse
Affiliation(s)
- Monica A. Thomas
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Andrew B. Kleist
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Brian F. Volkman
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
9
|
Mommert S, Aslan D, Ratz L, Stark H, Gutzmer R, Werfel T. The Anaphylatoxin C3a Receptor Expression on Human M2 Macrophages Is Down-Regulated by Stimulating the Histamine H4 Receptor and the IL-4 Receptor. J Innate Immun 2018; 10:349-362. [PMID: 30032131 DOI: 10.1159/000490426] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/28/2018] [Indexed: 12/19/2022] Open
Abstract
The anaphylatoxin C3a triggers inflammation by binding to its specific G-protein-coupled C3a receptor (C3aR). Since the number of C3aR, which is expressed on the cell surface, affects the response to C3a, we investigated the expression levels of C3aR on human M2 macrophages in allergic situations where high levels of the Th2 cytokine IL-4 and histamine are present in a local microenvironment. The histamine H1 receptor (H1R), H2R and the H4R mRNA expressions were induced or up-regulated during the differentiation process of M2 macrophages. The presence of histamine or agonists targeting the H1R, H2R and, in particular, the H4R during in vitro differentiation from monocytes to macrophages modified the M2 phenotype by regulating the macrophage differentiation marker CD68 and CD163 expressions. In -addition, the C3aR expression was also down-regulated by -ST-1006 during this process. Histamine and ST-1006 down-regulated the expression of C3aR with different time kinetics on fully differentiated M2 macrophages. By analysing C3a-induced IL-6 mRNA expression, we observed a diminished response to C3a in ST-1006-treated M2 macrophages when compared to un-treated cells. Expression of C3 was not affected by histamine, whereas IL-4 strongly down-regulated C3aR and C3 expressions. Our data suggests that down-regulation of C3aR expression by mediators present in allergic situations such as IL-4 or histamine has an anti-inflammatory impact by reducing the sensitivity to C3a-induced down-stream signaling, thereby contributing to the regulation of local inflammatory responses in the skin.
Collapse
Affiliation(s)
- Susanne Mommert
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Derya Aslan
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Lisanne Ratz
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Duesseldorf, Germany
| | - Ralf Gutzmer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Núñez K, Thevenot P, Alfadhli A, Cohen A. Complement Activation in Liver Transplantation: Role of Donor Macrosteatosis and Implications in Delayed Graft Function. Int J Mol Sci 2018; 19:ijms19061750. [PMID: 29899265 PMCID: PMC6032339 DOI: 10.3390/ijms19061750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/28/2018] [Accepted: 06/08/2018] [Indexed: 12/16/2022] Open
Abstract
The complement system anchors the innate inflammatory response by triggering both cell-mediated and antibody-mediated immune responses against pathogens. The complement system also plays a critical role in sterile tissue injury by responding to damage-associated molecular patterns. The degree and duration of complement activation may be a critical variable controlling the balance between regenerative and destructive inflammation following sterile injury. Recent studies in kidney transplantation suggest that aberrant complement activation may play a significant role in delayed graft function following transplantation, confirming results obtained from rodent models of renal ischemia/reperfusion (I/R) injury. Deactivating the complement cascade through targeting anaphylatoxins (C3a/C5a) might be an effective clinical strategy to dampen reperfusion injury and reduce delayed graft function in liver transplantation. Targeting the complement cascade may be critical in donor livers with mild to moderate steatosis, where elevated lipid burden amplifies stress responses and increases hepatocyte turnover. Steatosis-driven complement activation in the donor liver may also have implications in rejection and thrombolytic complications following transplantation. This review focuses on the roles of complement activation in liver I/R injury, strategies to target complement activation in liver I/R, and potential opportunities to translate these strategies to transplanting donor livers with mild to moderate steatosis.
Collapse
Affiliation(s)
- Kelley Núñez
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Paul Thevenot
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Abeer Alfadhli
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Ari Cohen
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| |
Collapse
|
11
|
Quell KM, Karsten CM, Kordowski A, Almeida LN, Briukhovetska D, Wiese AV, Sun J, Ender F, Antoniou K, Schröder T, Schmudde I, Berger JL, König P, Vollbrandt T, Laumonnier Y, Köhl J. Monitoring C3aR Expression Using a Floxed tdTomato-C3aR Reporter Knock-in Mouse. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28626064 DOI: 10.4049/jimmunol.1700318] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
C3a exerts multiple biologic functions through activation of its cognate C3a receptor. C3-/- and C3aR-/- mice have been instrumental in defining important roles of the C3a/C3aR axis in the regulation of acute and chronic inflammatory diseases, including ischemia/reperfusion injury, allergic asthma, autoimmune nephritis, and rheumatoid arthritis. Surprisingly little is known about C3aR expression and function in immune and stromal cells. To close this gap, we generated a floxed tandem-dye Tomato (tdTomato)-C3aR reporter knock-in mouse, which we used to monitor C3aR expression in cells residing in the lung, airways, lamina propria (LP) of the small intestine, brain, visceral adipose tissue, bone marrow (BM), spleen, and the circulation. We found a strong expression of tdTomato-C3aR in the brain, lung, LP, and visceral adipose tissue, whereas it was minor in the spleen, blood, BM, and the airways. Most macrophage and eosinophil populations were tdTomato-C3aR+ Interestingly, most tissue eosinophils and some macrophage populations expressed C3aR intracellularly. BM-derived dendritic cells (DCs), lung-resident cluster of differentiation (CD) 11b+ conventional DCs (cDCs) and monocyte-derived DCs, LP CD103+, and CD11b+ cDCs but not pulmonary CD103+ cDCs and splenic DCs were tdTomato-C3aR+ Surprisingly, neither BM, blood, lung neutrophils, nor mast cells expressed C3aR. Similarly, all lymphoid-derived cells were tdTomato-C3aR-, except some LP-derived type 3 innate lymphoid cells. Pulmonary and LP-derived epithelial cells expressed at best minor levels of C3aR. In summary, we provide novel insights into the expression pattern of C3aR in mice. The floxed C3aR knock-in mouse will help to reliably track and conditionally delete C3aR expression in experimental models of inflammation.
Collapse
Affiliation(s)
- Katharina M Quell
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | | | - Daria Briukhovetska
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Jing Sun
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Konstantina Antoniou
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Torsten Schröder
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Inken Schmudde
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Johann L Berger
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Tillman Vollbrandt
- Cell Analysis Core Facility, University of Lübeck, Lübeck 23562, Germany; and
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany;
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany; .,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
12
|
Novel insights into the expression pattern of anaphylatoxin receptors in mice and men. Mol Immunol 2017; 89:44-58. [PMID: 28600003 DOI: 10.1016/j.molimm.2017.05.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
Abstract
The anaphylatoxins (AT) C3a and C5a play important roles as mediators of inflammation. Further, they regulate and control multiple innate and adaptive immune responses through binding and activation of their cognate G protein-coupled receptors, i.e. C3a receptor (C3aR), C5a receptor 1 (C5aR1) and C5a receptor 2 (C5aR2), although the latter lacks important sequence motifs for G protein-coupling. Based on their pleiotropic functions, they contribute not only to tissue homeostasis but drive, perpetuate and resolve immune responses in many inflammatory diseases including infections, malignancies, autoimmune as well as allergic diseases. During the past few years, transcriptome expression data provided detailed insights into AT receptor tissue mRNA expression. In contrast, our understanding of cellular AT receptor expression in human and mouse tissues under steady and inflammatory conditions is still sketchy. Ligand binding studies, flow cytometric and immunohistochemical analyses convincingly demonstrated tissue-specific C5aR1 expression in various cells of myeloid origin. However, a detailed map for C3aR or C5aR2 expression in human or mouse tissue cells is still lacking. Also, reports about AT expression in lymphoid cells is still controversial. To understand the multiple roles of the ATs in the innate and adaptive immune networks, a detailed understanding of their receptor expression in health and disease is required. Recent findings obtained with novel GFP or tdTomato AT-receptor knock-in mice provide detailed insights into their expression pattern in tissue immune and stroma cells. Here, we will provide an update about our current knowledge of AT receptor expression pattern in humans and mice.
Collapse
|
13
|
Abdelbaset-Ismail A, Borkowska-Rzeszotek S, Kubis E, Bujko K, Brzeźniakiewicz-Janus K, Bolkun L, Kloczko J, Moniuszko M, Basak GW, Wiktor-Jedrzejczak W, Ratajczak MZ. Activation of the complement cascade enhances motility of leukemic cells by downregulating expression of HO-1. Leukemia 2017; 31:446-458. [PMID: 27451975 PMCID: PMC5288274 DOI: 10.1038/leu.2016.198] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023]
Abstract
As a crucial arm of innate immunity, the complement cascade (ComC) is involved both in mobilization of normal hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood and in their homing to BM. Despite the fact that ComC cleavage fragments alone do not chemoattract normal HSPCs, we found that leukemia cell lines as well as clonogenic blasts from chronic myeloid leukemia and acute myeloid leukemia patients respond robustly to C3 and C5 cleavage fragments by chemotaxis and increased adhesion. This finding was supported by the detection of C3a and C5a receptors in cells from human malignant hematopoietic cell lines and patient blasts at the mRNA (reverse transcriptase-polymerase chain reaction) and protein level (fluorescence-activated cell sorting), and by the demonstration that these receptors respond to stimulation by C3a and C5a by phosphorylation of p42/44 and p38 mitogen-activated protein kinases (MAPK), and protein kinase B (PKB/AKT). We also found that inducible heme oxygenase 1 (HO-1) is a negative regulator of ComC-mediated trafficking of leukemic cells, and that stimulation of leukemic cells by C3 or C5 cleavage fragments activates p38 MAPK, which downregulates HO-1 expression, rendering cells more mobile. We conclude that activation of the ComC in leukemia/lymphoma patients (for example, as a result of accompanying infections) enhances the motility of malignant cells and contributes to their spread in a p38 MAPK-HO-1-dependent manner. Therefore, inhibition of p38 MAPK or upregulation of HO-1 by small-molecule modulators would have a beneficial effect on ameliorating cell migration-mediated expansion of leukemia/lymphoma cells when the ComC becomes activated.
Collapse
Affiliation(s)
- A Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | | | - E Kubis
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - K Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | | | - L Bolkun
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - J Kloczko
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - M Moniuszko
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
| | - G W Basak
- Department of Hematology, Warsaw Medical University, Warsaw, Poland
| | | | - M Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
14
|
Dima E, Koltsida O, Katsaounou P, Vakali S, Koutsoukou A, Koulouris NG, Rovina N. Implication of Interleukin (IL)-18 in the pathogenesis of chronic obstructive pulmonary disease (COPD). Cytokine 2015; 74:313-7. [PMID: 25922275 DOI: 10.1016/j.cyto.2015.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/13/2015] [Accepted: 04/15/2015] [Indexed: 12/15/2022]
Abstract
Interleukin (IL)-18 is a pro-inflammatory cytokine that was firstly described as an interferon (IFN)-γ-inducing factor. Similar to IL-1β, IL-18 is synthesized as an inactive precursor requiring processing by caspase-1 into an active cytokine. The platform for activating caspase-1 is known as the inflammasome, a multiple protein complex. Macrophages and dendritic cells are the primary sources for the release of active IL-18, whereas the inactive precursor remains in the intracellular compartment of mesenchymal cells. Finally, the IL-18 precursor is released from dying cells and processed extracellularly. IL-18 has crucial host defense and antitumor activities, and gene therapy to increase IL-18 levels in tissues protects experimental animals from infection and tumor growth and metastasis. Moreover, multiple studies in experimental animal models have shown that IL-18 over-expression results to emphysematous lesions in mice. The published data prompt to the hypothesis that IL-18 induces a broad spectrum of COPD-like inflammatory and remodeling responses in the murine lung and also induces a mixed type 1, type 2, and type 17 cytokine responses. The majority of studies identify IL-18 as a potential target for future COPD therapeutics to limit both the destructive and remodeling processes occurring in COPD lungs.
Collapse
Affiliation(s)
- Efrossini Dima
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece
| | - Ourania Koltsida
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece
| | - Paraskevi Katsaounou
- Pumonary Department, Intensive Care Medicine, Evaggelismos Hospital, Medical School, University of Athens, Greece
| | - Sofia Vakali
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece
| | - Antonia Koutsoukou
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece
| | - Nikolaos G Koulouris
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece
| | - Nikoletta Rovina
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and "Sotiria" Chest Disease Hospital, 11527 Athens, Greece.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Lim H, Kim YU, Drouin SM, Mueller-Ortiz S, Yun K, Morschl E, Wetsel RA, Chung Y. Negative regulation of pulmonary Th17 responses by C3a anaphylatoxin during allergic inflammation in mice. PLoS One 2012; 7:e52666. [PMID: 23285141 PMCID: PMC3527591 DOI: 10.1371/journal.pone.0052666] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 11/20/2012] [Indexed: 12/14/2022] Open
Abstract
Activation of complement is one of the earliest immune responses to exogenous threats, resulting in various cleavage products including anaphylatoxin C3a. In addition to its contribution to host defense, C3a has been shown to mediate Th2 responses in animal models of asthma. However, the role of C3a on pulmonary Th17 responses during allergic inflammation remains unclear. Here, we show that mice deficient in C3a receptor (C3aR) exhibited (i) higher percentages of endogenous IL-17-producing CD4+ T cells in the lungs, (ii) higher amounts of IL-17 in the bronchoalveolar lavage fluid, and (iii) more neutrophils in the lungs than wild-type mice when challenged with intranasal allergens. Moreover, adoptive transfer experiments showed that the frequencies of antigen-specific IL-17-producing CD4+ T cells were significantly higher in the lungs and bronchial lymph nodes of C3aR-deficient recipients than those of wild-types recipients. Bone-marrow reconstitution study indicated that C3aR-deficiency on hematopoietic cells was required for the increased Th17 responses. Furthermore, C3aR-deficient mice exhibited increased percentages of Foxp3+ regulatory T cells; however, depletion of these cells minimally affected the induction of antigen-specific Th17 cell population in the lungs. Neutralization of IL-17 significantly reduced the number of neutrophils in bronchoalveolar lavage fluid of C3aR-deficient mice. Our findings demonstrate that C3a signals negatively regulate antigen-specific Th17 responses during allergic lung inflammation and the size of Foxp3+ regulatory T cell population in the periphery.
Collapse
Affiliation(s)
- Hoyong Lim
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Young Uk Kim
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, the University of Texas Health Science Center, Houston, Houston, Texas, United States of America
| | - Scott M. Drouin
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, the University of Texas Health Science Center, Houston, Houston, Texas, United States of America
| | - Stacey Mueller-Ortiz
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Kyoungah Yun
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
- Daejon Health Sciences College, Daejon, South Korea
| | - Eva Morschl
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Rick A. Wetsel
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, the University of Texas Health Science Center, Houston, Houston, Texas, United States of America
| | - Yeonseok Chung
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, the University of Texas Medical School at Houston, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, the University of Texas Health Science Center, Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
18
|
Niebuhr M, Bäumer W, Kietzmann M, Wichmann K, Heratizadeh A, Werfel T. Participation of complement 3a receptor (C3aR) in the sensitization phase of Th2 mediated allergic contact dermatitis. Exp Dermatol 2012; 21:52-6. [PMID: 22151392 DOI: 10.1111/j.1600-0625.2011.01403.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The complement system has emerged as a bridge between innate and adaptive immune responses. An involvement of C3aR has been described during skin inflammation. The aim of the study was to investigate the role of C3a in a mouse model of allergic skin inflammation, such as allergic contact dermatitis (ACD) which is a clinical manifestation of contact sensitivity (CS). The sensitization phase was studied using the local lymph node test: Mice were sensitized on three consecutive days by application of non-irritant concentrations of toluene-2,4-diisocyanate (TDI; 0.5%) onto the ear skin. On day 5, auricular draining lymph nodes were obtained. The elicitation phase was investigated by sensitization with TDI on the depilated and tape-stripped abdominal skin and challenge with TDI on the ear skin and measuring of ear swelling in vivo and cytokine secretion in activated splenocytes in vitro respectively. Complement 3a receptor deficient (C3aRKO) mice showed increased cytokine responses (interleukin[IL]-5, IL-6, IL-17, granulocyte macrophage-colony stimulating factor [GM-CSF]) in the sensitization phase of ACD to TDI. However, no differences in CS responses to TDI were observed in C3aR KO mice compared with WT controls in the elicitation phase of ACD as assessed by measuring of ear swelling in vivo and cytokines in skin and in activated splenocytes in vitro, namely IL-1α, IL-2, IL-4, IL-5, IL-6, IL-10, IL-17, interferon-γ (IFN-γ), GM-CSF and tumor necrosis factor (TNF)-α. These findings provide a new insight into the participation of C3a in the sensitization phase of CS immune responses.
Collapse
Affiliation(s)
- Margarete Niebuhr
- Department of Dermatology and Allergy, Division of Immunodermatology and Allergy Research, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
19
|
Zhou W. The new face of anaphylatoxins in immune regulation. Immunobiology 2012; 217:225-34. [DOI: 10.1016/j.imbio.2011.07.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 06/22/2011] [Accepted: 07/17/2011] [Indexed: 11/26/2022]
|
20
|
Li K, Fazekasova H, Wang N, Peng Q, Sacks SH, Lombardi G, Zhou W. Functional modulation of human monocytes derived DCs by anaphylatoxins C3a and C5a. Immunobiology 2011; 217:65-73. [PMID: 21855168 PMCID: PMC3234345 DOI: 10.1016/j.imbio.2011.07.033] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 07/29/2011] [Indexed: 10/24/2022]
Abstract
Anaphylatoxins C3a and C5a are important modulators for dendritic cell activation and function in mice. In order to verify the significance of these observations in man, we have investigated the functional modulation of human monocytes derived DCs by C3a and C5a. Here we report that engagement of C3aR or C5aR on human monocytes derived DCs (moDCs) enhances the cell activation and their capacity for allostimulation. In addition, we show that intracellular production of cAMP is reduced and PI3K/AKT, ERK and NF-κB signalling is increased following stimulation with C3a or C5a, identifying intracellular signalling pathways that could convert cell surface C3aR and C5aR engagement into changes in moDC functions. Our data provide evidence that human DCs are equipped to react to C3a/C5a and undergo phenotypic change as well as functional modulation. Complement offers a potential route to modulate human DC function and regulate T cell mediated immunity.
Collapse
Affiliation(s)
- Ke Li
- King's College London, MRC Centre for Transplantation, NIHR Comprehensive Biomedical Research Centre, Guy's Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
21
|
Purwar R, Bäumer W, Niebuhr M, Tschernig T, Kietzmann M, Werfel T. A protective role of complement component 3 in T cell-mediated skin inflammation. Exp Dermatol 2011; 20:709-14. [PMID: 21569105 DOI: 10.1111/j.1600-0625.2011.01295.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Keratinocytes synthesize complement component 3 (C3) constitutively, and increased expression of C3 has been described during skin inflammation. In this study, we investigated the role of C3 in T cell-mediated allergic contact dermatitis, which is a clinical manifestation of contact sensitivity (CS). C3-deficient mice (C3KO) showed substantial higher CS responses to haptens, inducing a Th1 cytokine-mediated skin inflammation (2,4-dinitrofluorobenzene and dinitrochlorobenzene), and to haptens known to induce a Th2-polarized inflammatory response (fluoro-isothiocynate and toluene-2,4-diisocyanate) as compared to their wild-type (WT) controls. There was a higher influx of GR-1(+) , CD4(+) , and CD8(+) cells into the skin of hapten-treated C3KO mice compared with WT mice. Activated splenocytes from C3KO mice immunized with DNCB secreted higher amounts of IFN-γ compared with WT controls but not of Th2 (IL-4, IL-5, and IL-10) cytokines or IL-17. A higher secretion of IL-12 from splenocytes of C3KO mice as compared with WT mice was observed after TLR-4 ligand (LPS) or TLR-2 ligand (peptidoglycan) stimulation. Thus, an increased expression of IL-12 and of IFN-γ may be responsible for the increased hapten-induced inflammation in C3 deficiency. Finally, we demonstrated that C3KO mice developed oral tolerance to haptens to a lower degree than WT mice. Our findings provide a new insight into a novel anti-inflammatory role of C3 in skin inflammation.
Collapse
Affiliation(s)
- Rahul Purwar
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Ricklinger Strasse 5, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Wiederin JL, Donahoe RM, Anderson JR, Yu F, Fox HS, Gendelman HE, Ciborowski PS. Plasma proteomic analysis of simian immunodeficiency virus infection of rhesus macaques. J Proteome Res 2011; 9:4721-31. [PMID: 20677826 DOI: 10.1021/pr1004345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lentiviral replication in its target cells affects a delicate balance between cellular cofactors required for virus propagation and immunoregulation for host defense. To better elucidate cellular proteins linked to viral infection, we tested plasma from rhesus macaques infected with the simian immunodeficiency viral strain SIVsmm9, prior to, 10 days (acute), and 49 weeks (chronic) after viral infection. Changes in plasma protein content were measured by quantitative mass spectrometry by isobaric tags for absolute and relative quantitation (iTRAQ) methods. An 81 and 232% increase in SERPINA1 was seen during acute and chronic infection, respectively. Interestingly, gelsolin, vitamin D binding protein and histidine rich glycoprotein were decreased by 45% in acute conditions but returned to baseline during chronic infection. When compared to uninfected controls, a 48-103% increase in leucine rich alpha 2-glycoprotein, vitronectin, and ceruloplasmin was observed during chronic viral infection. Observed changes in plasma proteins expression likely represent a compensatory host response to persistent viral infection.
Collapse
Affiliation(s)
- Jayme L Wiederin
- University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Belgorosky D, Sander VA, Yorio MPD, Faletti AG, Motta AB. Hyperandrogenism alters intraovarian parameters during early folliculogenesis in mice. Reprod Biomed Online 2010; 20:797-807. [PMID: 20362510 DOI: 10.1016/j.rbmo.2010.02.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 11/23/2009] [Accepted: 01/21/2010] [Indexed: 01/31/2023]
Abstract
This study aimed to investigate how hyperandrogenism affects early folliculogenesis. Hyperandrogenism was induced in prepuberal female BALB/c mice by daily s.c. injection of dehydroepiandrosterone (60 mg/kg body weight in 0.1 ml sesame oil) for 10 consecutive days. Although hyperandrogenism increased the growth rate of primary follicles, it also increased ovarian oxidative stress (evaluated by the increase in lipid peroxidation, the decrease in superoxide dismutase activity and the fact that glutathione content was not modified). By using the annexin V/cytometry assay it was found that the excess of androgens decreased viable ovarian cells and increased early apoptotic ones. The increased lipid peroxidation induced enhanced ovarian prostaglandin E production. In addition, hyperandrogenism increased the number of T lymphocytes that infiltrate ovarian tissue and modified their phenotype (decreased CD4+ or helper and increased the suppressor/cytotoxic CD8+). The excess of androgens decreased the ovarian expression of the long isoform of leptin receptor (Ob-Rb, the only isoform expressed in the ovarian tissue) when compared with controls. All these alterations increased serum concentrations of oestradiol, a pro-apoptotic agent. It is concluded that the excess of androgens impairs early follicular development by modulating some endocrine and immune parameters that are either directly or indirectly related to follicular atresia.
Collapse
Affiliation(s)
- Denise Belgorosky
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Departamento de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 1121ABG Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
24
|
Ali H. Regulation of human mast cell and basophil function by anaphylatoxins C3a and C5a. Immunol Lett 2009; 128:36-45. [PMID: 19895849 DOI: 10.1016/j.imlet.2009.10.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 10/21/2009] [Accepted: 10/23/2009] [Indexed: 12/18/2022]
Abstract
Allergic diseases such as asthma result from inappropriate immunologic responses to common environmental allergens in genetically susceptible individuals. Following allergen exposure, interaction of dendritic cells (DC) with CD4(+) T cells leads to the production of Th2 cytokines, which induce B cells to synthesize IgE molecules (sensitization phase). These IgE molecules bind to their high affinity receptors (FcvarepsilonRI) on the surface of mast cells and basophils and their subsequent cross-linking by allergen results in the release of preformed and newly synthesized mediators, which cause bronchoconstriction, lung inflammation and airway hyperresponsiveness (AHR) in asthma (effector phase). The complement components C3a and C5a levels are increased in the lungs of patients with asthma and are likely generated via the actions of both allergen and mast cell proteases. In vivo studies with rodents have shown that while C3a facilitates allergen sensitization in some models C5a inhibits this response. Despite this difference, both anaphylatoxins promote lung inflammation and AHR in vivo indicating that cells other than DC and T cells likely mediate the functional effects of C3a and C5a in asthma. This review focuses on the contribution of C3a and C5a in the pathogenesis of asthma with a particular emphasis on mast cells and basophils. It discusses the mechanisms by which anaphylatoxins activate mast cells and basophils and the associated signaling pathways via which their receptors are regulated by priming and desensitization.
Collapse
Affiliation(s)
- Hydar Ali
- Department of Pathology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104-6030, USA.
| |
Collapse
|
25
|
Schmalstieg FC, Goldman AS. Jules Bordet (1870-1961): a bridge between early and modern immunology. JOURNAL OF MEDICAL BIOGRAPHY 2009; 17:217-224. [PMID: 20029083 DOI: 10.1258/jmb.2009.009061] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Jules Bordet, a pioneering immunologist, lived until the dawn of molecular immunology. He was born in Belgium in 1870, obtained a medical degree in 1892, worked at l'Institut Pasteur in Paris from 1894 to 1901 and then established the Pasteur Institute of Brabant in Brussels. Before World War I, Bordet found that complement binds to antibody-antigen complexes regardless of the antigen or antibodies involved. Subsequently he developed the complement fixation test that was of diagnostic importance for several decades. For his research concerning complement he was awarded the 1919 Nobel Prize in Physiology or Medicine. During that period he also discovered anaphylatoxin, conglutinin, and the cause of whooping cough (Bordetella pertussis). After World War I he found how thrombin forms, how platelets participate in clotting, lysozyme in human milk and much of the biology of bacteriophages. In addition, Bordet worked fervently to limit weapons of mass destruction and promote peace until his death in 1961.
Collapse
|
26
|
Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Köhl J. The role of the anaphylatoxins in health and disease. Mol Immunol 2009; 46:2753-66. [PMID: 19477527 DOI: 10.1016/j.molimm.2009.04.027] [Citation(s) in RCA: 522] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 04/28/2009] [Indexed: 12/16/2022]
Abstract
The anaphylatoxin (AT) C3a, C5a and C5a-desArg are generally considered pro-inflammatory polypeptides generated after proteolytic cleavage of C3 and C5 in response to complement activation. Their well-appreciated effector functions include chemotaxis and activation of granulocytes, mast cells and macrophages. Recent evidence suggests that ATs are also generated locally within tissues by pathogen-, cell-, or contact system-derived proteases. This local generation of ATs is important for their pleiotropic biologic effects beyond inflammation. The ATs exert most of the biologic activities through ligation of three cognate receptors, i.e. the C3a receptor, the C5a receptor and the C5a receptor-like, C5L2. Here, we will discuss recent findings suggesting that ATs regulate cell apoptosis, lipid metabolism as well as innate and adaptive immune responses through their impact on antigen-presenting cells and T cells. As we will outline, such regulatory functions of ATs and their receptors play important roles in the pathogenesis of allergy, autoimmunity, neurodegenerative diseases, cancer and infections with intracellular pathogens.
Collapse
Affiliation(s)
- Andreas Klos
- Institute of Medical Microbiology and Hospital Epidemiology, Medical School Hannover, MHH, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Buzzeo MP, Yang J, Casella G, Reddy V. A preliminary gene expression profile of acute graft-versus-host disease. Cell Transplant 2009; 17:489-94. [PMID: 18714668 DOI: 10.3727/096368908785096042] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice for high-risk hematological malignancies, yet a major complication associated with this therapy is acute graft-versus-host disease (GVHD). Despite a well-defined pathophysiological mechanism, there are no definitive markers for predicting acute GVHD development or progression to advanced stages. In the current study, we enrolled four acute GVHD and four acute GVHD-free recipients of allogeneic HSCT and collected peripheral blood just prior to onset of clinical acute GVHD for analysis on Affymetrix GeneChip Human Genome U133 Plus 2.0 microarrays. We noted significant differences in expression of 1,658 genes between control and acute GVHD patients, based on an analysis of covariance (ANCOVA) by type of transplant, a pooled error estimate, and a false discovery rate (FDR) of 10%. In conclusion, we offer the first report of a preliminary molecular signature of acute GVHD in allogeneic HSCT patients.
Collapse
Affiliation(s)
- Matthew P Buzzeo
- Department of Medicine, Division of Hematology/Oncology, University of Florida, Gainesville, FL, USA
| | | | | | | |
Collapse
|
28
|
Rynkowski MA, Kim GH, Garrett MC, Zacharia BE, Otten ML, Sosunov SA, Komotar RJ, Hassid BG, Ducruet AF, Lambris JD, Connolly ES. C3a receptor antagonist attenuates brain injury after intracerebral hemorrhage. J Cereb Blood Flow Metab 2009; 29:98-107. [PMID: 18728680 PMCID: PMC3731073 DOI: 10.1038/jcbfm.2008.95] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroprotective therapy targeting the complement cascade may reduce injury associated with intracerebral hemorrhage (ICH). We investigated the role of C3a-receptor antagonist (C3aRA) after ICH in mice. Autologous whole blood was infused into the right striatum of mice that were treated with C3aRA or vehicle, using both a pre- and postinjury dosing regimen. Hematoma volume, brain water content, and inflammatory cell profile were assessed at 72 h post-ICH. Neurologic dysfunction was assessed by evaluating both spatial memory and sensorimotor capacity. Animals pretreated with C3aRA showed significantly improved neurologic function, brain water content, and granulocyte infiltration relative to vehicle-treated animals when assessed at 72 h. There was no significant difference in hemorrhagic/nonhemorrhagic ratio of microglial activation among all groups. Hematoma volumes were also not significantly different between C3aRA-treated and vehicle-treated animals. Administration of C3aRA beginning 6 h postinjury afforded significant amelioration of neurologic dysfunction as well as a reduction in brain water content. Treatment with C3aRA improved neurologic outcome while reducing inflammatory cell infiltration and brain edema formation after experimental ICH in mice. Results of this study suggest that the C3a receptor may be a promising target for therapeutic intervention in hemorrhagic stroke.
Collapse
Affiliation(s)
- Michal A Rynkowski
- Department of Neurological Surgery, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zaidi AK, Ali H. C3a receptors signaling in mast cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 598:126-40. [PMID: 17892209 DOI: 10.1007/978-0-387-71767-8_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Asifa K Zaidi
- University of Pennsylvania School of Dental Medicine, Department of Pathology, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
30
|
Yoon SY, Kwon YB, Kim HW, Roh DH, Seo HS, Han HJ, Lee HJ, Beitz AJ, Hwang SW, Lee JH. Peripheral bee venom's anti-inflammatory effect involves activation of the coeruleospinal pathway and sympathetic preganglionic neurons. Neurosci Res 2007; 59:51-9. [PMID: 17588699 DOI: 10.1016/j.neures.2007.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/18/2007] [Accepted: 05/25/2007] [Indexed: 01/26/2023]
Abstract
There are several reports indicating that the locus coeruleus (LC) is capable of altering immune responses. Moreover, it is well established that the LC is the major source of descending noradrenergic system. Recently we have demonstrated that subcutaneous bee venom (BV) injection dramatically suppressed peripheral inflammation through activation of sympathetic preganglionic neurons (SPNs) leading to release of adreno-medullary catecholamines. Importantly, this 'BV-induced anti-inflammatory effect' (BVAI) is also associated with an increase of the activity of LC. Based on these data, present study examined whether BV-induced LC activation increased the activity of SPNs and this pathway played a role in BVAI using a zymosan-induced inflammatory air pouch model in mice. Unilateral BV injection into left hind limb produced anti-inflammation and specifically increased Fos expression in SPNs of the T7-T11 (which mainly project to adrenal medulla), but not those of the T1-T6 or T12-L2 spinal cord. 6-Hydroxydopamine-induced unilateral lesion of the contralateral, but not ipsilateral (to the BV injection site) LC significantly blocked BVAI and BV-induced Fos expression in SPNs. Additionally, intrathecal administration of idazoxan (alpha2-adrenoceptor antagonist), blocked BVAI. These results indicate that BV-induced activation of the contralateral LC-descending noradrenergic pathway increased the activity of SPNs that project to the adrenal medulla and this pathway is necessary for BVAI.
Collapse
Affiliation(s)
- Seo-Yeon Yoon
- Biotherapy Human Resources Center, College of Veterinary Medicine, Chonnam National University, Gwang-ju, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
DiScipio RG, Schraufstatter IU. The role of the complement anaphylatoxins in the recruitment of eosinophils. Int Immunopharmacol 2007; 7:1909-23. [PMID: 18039528 DOI: 10.1016/j.intimp.2007.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 07/09/2007] [Indexed: 01/21/2023]
Abstract
Eosinophils are blood and tissue immune cells that participate in a diverse range of activities normally beneficial for the host defense, but in circumstances of untoward inflammatory conditions these cells can be responsible for pathological responses. Accordingly the transit of eosinophils from the blood to tissues is a subject of considerable importance in immunology. In this article we review how the complement anaphylatoxins, C3a and C5a bring about eosinophil extravasation. These mediators do not merely provide a chemotactic or haptotactic gradient but are responsible for orchestrating innumerable responses by other cells types, including of endothelial cells, mast cells, and basophils in order to create an environment that is conducive for eosinophil infiltration. C5a has the capacity to prime the endothelium directly to present P-selectin, and C5a stimulated generation of eosinophil hydrogen peroxide and other oxidants can cause additional upregulation of endothelial P-selectin and ICAM-1. Moreover, the anaphylatoxins have the ability to recruit mast cells and basophils and can stimulate these cells to release IL-4 and IL-13, which by augmenting endothelial VCAM-1, convey some selectivity for eosinophils. The anaphylatoxins also have the capability to evoke the release and activation of eosinophil MMP-9, which is employed by this cell type to digest its way past the subendothelial matrix. Finally, because C3a and C5a can stimulate the generation of nitric oxide along with the secretion of histamine and LTC4 from several cell types, the anaphylatoxins can bring about an increase in vascular permeability that facilitates eosinophil accumulation at sites of allergic inflammation.
Collapse
Affiliation(s)
- Richard G DiScipio
- La Jolla Institute for Molecular Medicine, 4570 Executive Dr. #100, San Diego, CA 92122, USA.
| | | |
Collapse
|
32
|
Abstract
The complement system is an important component of the innate immune system and a modulator of adaptive immunity. The entire complement system is focused on C3 and C5. Thus, there are proteins that activate C3 and C5, those that regulate this activation, and those that transduce the effects of C3 and C5 activation products; each can affect the kidney in renal injury. The normal kidney has the inherent capacity to protect itself from complement activation through cellular expression of decay-accelerating factor, membrane cofactor protein (in human beings), and Crry (in rodents). In addition, plasma factor H protects vascular spaces in the kidney. Although the main function of these proteins is to limit complement activation, there is now considerable evidence that they can transduce signals on engagement in immune cells. The G-protein-coupled 7-span transmembrane receptors for C3a and C5a, and the integral membrane complement receptors (CR) for C3b, iC3b, and C3dg, are expressed outside the kidney, particularly in cells of hematopoietic and immune lineage. These are important in renal injury through their infiltration of the kidney and/or by affecting kidney-directed immune responses. There is mounting evidence that intrinsic glomerular and tubular cell C3aR and C5aR expression and activation also can affect renal injury. CR1 on podocytes and the beta2 integrins CR3 and CR4 in kidney dendritic cells have functions that remain poorly defined. Cells of the kidney also have the capacity to produce and activate their own complement proteins. Thus, intrinsic renal cells express decay-accelerating factor, membrane cofactor protein, Crry, C3aR, C5aR, CR1, CR3, and CR4. These can be engaged by C3 and C5 activation products derived from systemic and local pools in renal injury. Given their capacity to provide signals that influence kidney cellular behavior, their activation can have substantial effects in renal injury. Defining these in a cell- and disease-specific fashion is an exciting challenge for future research.
Collapse
Affiliation(s)
- Tipu S Puri
- Section of Nephrology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
33
|
Reis ES, Barbuto JAM, Isaac L. Complement components, regulators and receptors are produced by human monocyte-derived dendritic cells. Immunobiology 2007; 212:151-7. [PMID: 17412282 DOI: 10.1016/j.imbio.2006.11.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 11/14/2006] [Accepted: 11/27/2006] [Indexed: 01/19/2023]
Abstract
Complement and dendritic cells (DCs) are essential components of innate immunity. Both participate in local inflammation and moreover have roles in the initiation of the acquired immunity response and in the maintenance of tolerance. Recent studies have demonstrated the ability of DCs to synthesize C1q, C3, Factor I, Factor B and complement receptors 3 and 4. In this study, we demonstrate that human DCs are a source of other soluble complement proteins including C1q, C4b binding protein (C4BP), C7 and C8. Complement receptors (CR)1 and the CD18 chain (common for CR3 and CR4) were also present on DCs while CR2 was not detected.
Collapse
Affiliation(s)
- Edimara S Reis
- Laboratório de Complemento, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1730, 05508-900 São Paulo, Brazil
| | | | | |
Collapse
|
34
|
Purwar R, Wittmann M, Zwirner J, Oppermann M, Kracht M, Dittrich-Breiholz O, Gutzmer R, Werfel T. Induction of C3 and CCL2 by C3a in keratinocytes: a novel autocrine amplification loop of inflammatory skin reactions. THE JOURNAL OF IMMUNOLOGY 2006; 177:4444-50. [PMID: 16982879 DOI: 10.4049/jimmunol.177.7.4444] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The complement fragment-3a (C3a) acts via a G protein-coupled C3aR and is of importance in allergic and inflammatory diseases. Recent studies suggest the presence of complement proteins in the epidermal compartment and synthesis of some of these proteins (C3, factor B, and factor H) by human primary keratinocytes (KCs) during inflammation. However, expression of C3aR and its role in human KCs is not elucidated thus far. In this study, we demonstrate the expression of C3aR on KCs as detected by quantitative real-time RT-PCR and flow cytometry. IFN-gamma and IFN-alpha strongly up-regulated the surface expression of C3aR on KCs among all other cytokines tested. After up-regulation of C3aR by IFN-gamma and IFN-alpha, we observed the induction of five genes (CCL2, CCL5, CXCL8, CXCL10, and C3) after stimulation of KCs with C3a in microarray analysis. We confirmed the induction of C3 and CCL2 at RNA and protein levels. Furthermore, incubation of C3 with skin mast cells tryptase resulted in the generation of C3 fragments with C3a activity. In conclusion, our data illustrate that epidermal KCs express functional C3aR. The increases of C3 and CCL2 synthesis by C3a and C3 activation by skin mast cell tryptase delineates a novel amplification loop of complement activation and inflammatory responses that may influence the pathogenesis of allergic/inflammatory skin diseases.
Collapse
Affiliation(s)
- Rahul Purwar
- Department of Dermatology and Allergology, Hannover Medical University, Ricklinger Strasse 05, D-30449 Hannover, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Clingan JM, Yanagawa Y, Iwabuchi K, Onoé K. Effect of T helper 1 (Th1)/Th2 cytokine on chemokine-induced dendritic cell functions. Cell Immunol 2006; 242:72-9. [PMID: 17098215 DOI: 10.1016/j.cellimm.2006.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 08/25/2006] [Accepted: 09/21/2006] [Indexed: 10/23/2022]
Abstract
Effects of T helper 2 (Th2) and Th1 cytokines, interleukin (IL)-4, and interferon (IFN)-gamma, respectively, on chemokine-induced DC migration and endocytosis are not well understood. We investigated herein the effects of these cytokines on chemokine-induced functions of murine myeloid DCs. As expected, immature DCs markedly migrated to CCL3 but not CCL19, while mature DCs showed vigorous migration in response to CCL19 but not CCL3. Both IL-4 and IFN-gamma significantly decreased CCL3-induced migration of immature DCs. In contrast, these cytokines exerted no significant effects on CCL19-induced migration of mature DCs. Of note, both IL-4 and IFN-gamma markedly enhanced CCL3-induced endocytosis of immature DCs. The messenger RNA level of CCR5, a CCL3 receptor, in immature DCs was slightly increased by IL-4 or IFN-gamma treatment. These results demonstrate that these Th1/Th2 cytokines can act to either inhibit or enhance chemokine-mediated DC functions, and may play a role in increasing antigen uptake by immature DCs at inflammatory sites.
Collapse
Affiliation(s)
- Jonathan M Clingan
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan
| | | | | | | |
Collapse
|
36
|
Zhou W, Peng Q, Li K, Sacks SH. Role of dendritic cell synthesis of complement in the allospecific T cell response. Mol Immunol 2006; 44:57-63. [PMID: 16870256 DOI: 10.1016/j.molimm.2006.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 06/27/2006] [Accepted: 06/27/2006] [Indexed: 11/26/2022]
Abstract
Although extrahepatic synthesis of complement and particularly C3 has been widely studied in most cells and tissues, new information is emerging on dendritic cells (DCs). This research has shown that mouse bone marrow (BM) derived DCs are able to synthesise C3 and this synthesis has a substantial impact on DC activation, affecting the diverse range of DC functions relevant to the allospecific T cell response. Thus, local production of C3 appears to regulate the capacity of DCs to trigger the primary T cell response against donor alloantigen. Understanding of the key mechanisms by which complement activation modulates DC maturation could lead to the development of therapeutic strategies to down regulate DC activation thus reduce allograft rejection.
Collapse
Affiliation(s)
- Wuding Zhou
- King's College London School of Medicine at Guy's, King's College and St. Thomas' Hospitals, Department of Nephrology and Transplantation, London, SE1 9RT, UK.
| | | | | | | |
Collapse
|
37
|
Cecic I, Sun J, Korbelik M. Role of complement anaphylatoxin C3a in photodynamic therapy-elicited engagement of host neutrophils and other immune cells. Photochem Photobiol 2006; 82:558-62. [PMID: 16613513 DOI: 10.1562/2005-09-09-ra-681] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor treatment by photodynamic therapy (PDT) provokes a host-protective inflammatory and acute-phase response and an immune reaction. Neutrophilia manifested in this context is driven by multiple mediators of neutrophil chemotaxis orchestrated by an activated complement system. Mouse FsaR fibrosarcoma was used in this study to further investigate neutrophilia induced by Photofrin-based PDT. The complement anaphylatoxin C3a was identified as a major chemoattractant in the advanced phase of PDT-induced neutrophilia, because injecting mice with antibodies blocking its receptor C3aR significantly inhibited the increase in neutrophil levels 8 h after PDT. At the same time point, an increased C3aR expression was detected in neutrophils, monocytes and B lymphocytes in the blood of host mice. Peritoneal macrophages and mast cells harvested from treatment-naive mice exhibited elevated C3aR expression after coincubation in vitro for 8 h with PDT-treated FsaR cells. Thus, C3a emerges as one of the key effector molecules engaged in PDT-induced host response.
Collapse
Affiliation(s)
- Ivana Cecic
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | | |
Collapse
|
38
|
Gutzmer R, Köther B, Zwirner J, Dijkstra D, Purwar R, Wittmann M, Werfel T. Human plasmacytoid dendritic cells express receptors for anaphylatoxins C3a and C5a and are chemoattracted to C3a and C5a. J Invest Dermatol 2006; 126:2422-9. [PMID: 16778800 DOI: 10.1038/sj.jid.5700416] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The presence of plasmacytoid dendritic cells (pDC) was recently demonstrated in lesions of inflammatory skin diseases. Since anaphylatoxins or their precursors were also found in such lesions, we investigated a possible interaction between pDC and anaphylatoxins C3a and C5a. pDC precursors isolated from peripheral blood did not express the receptors for C3a and C5a, complement C3a receptor (C3aR) and complement C3a receptor (C5aR). If these pDC precursors were cultured with IL-3, the resultant immature pDC expressed both receptors. Expression of C3aR and C5aR could also be demonstrated on pDC in lesions of cutaneous lupus erythematosus and allergic contact dermatitis. Such pDC were immature since they lacked the expression of the maturation marker CD83. Blood-derived pDC matured with CpG oligonucleotides downregulated the receptors. Immature pDC responded to C3a and C5a (but not C3adesArg) stimulation with increased F-actin polymerization and chemotactic migration. In contrast, interferon alpha production, surface molecule expression, and T-cell stimulatory capacity were not significantly modulated by C3a or C5a. Thus, immature pDC represent another type of antigen-presenting cell that express C3aR and C5aR, and respond to anaphylatoxins with chemotaxis. This might be relevant in the direction of pDC to cutaneous lesions of inflammation, for example, in lupus erythematosus or contact dermatitis.
Collapse
Affiliation(s)
- Ralf Gutzmer
- Department of Dermatology and Allergology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
39
|
Bao L, Osawe I, Haas M, Quigg RJ. Signaling through up-regulated C3a receptor is key to the development of experimental lupus nephritis. THE JOURNAL OF IMMUNOLOGY 2005; 175:1947-55. [PMID: 16034139 DOI: 10.4049/jimmunol.175.3.1947] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Signaling of the C3a anaphylatoxin through its G protein-coupled receptor, C3aR, is relevant in a variety of inflammatory diseases, but its role in lupus nephritis is undefined. In this study, we show that expression of C3aR was significantly increased in prediseased and diseased kidneys of MRL/lpr lupus mice compared with MRL/+ controls. To investigate the role of C3aR in experimental lupus, a small molecule antagonist of C3aR (C3aRa) was administered continuously to MRL/lpr mice from 13 to 19 wk of age. All 13 C3aRa-treated mice survived during the 6-wk treatment compared with 9 of 14 (64.3%) control animals given vehicle (p = 0.019). Relative to controls, C3aRa-treated animals were protected from renal disease as measured by albuminuria (p = 0.040) and blood urea nitrogen (p = 0.021). In addition, there were fewer neutrophils, monocytes, and apoptotic cells in the kidneys of C3aRa-treated mice. C3aRa treatment also led to reduced renal IL-1beta and RANTES mRNA and phosphorylated phosphatase and tensin homologue deleted on chromosome 10 protein, whereas the mass of phosphorylated protein kinase B/Akt was increased by C3aRa. Thus, C3aR antagonism significantly reduces renal disease in MRL/lpr mice, which further translates into prolonged survival. These data illustrate that C3aR is relevant in experimental lupus nephritis and may be a target for therapeutic intervention in the human disease.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/biosynthesis
- Apoptosis/drug effects
- Apoptosis/immunology
- Arginine/administration & dosage
- Arginine/analogs & derivatives
- Benzhydryl Compounds/administration & dosage
- Complement C3a/metabolism
- Complement C3a/physiology
- Complement Inactivator Proteins/administration & dosage
- Fluorescent Antibody Technique, Indirect
- Inflammation Mediators/metabolism
- Kidney/drug effects
- Kidney/immunology
- Kidney/metabolism
- Kidney/pathology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/mortality
- Lupus Erythematosus, Systemic/pathology
- Male
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred MRL lpr
- PTEN Phosphohydrolase
- Phosphoric Monoester Hydrolases/metabolism
- Phosphorylation/drug effects
- RNA, Messenger/biosynthesis
- Receptors, Complement/antagonists & inhibitors
- Receptors, Complement/biosynthesis
- Receptors, Complement/genetics
- Receptors, Complement/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Tumor Suppressor Proteins/metabolism
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Lihua Bao
- Section of Nephrology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
40
|
Sunyer JO, Boshra H, Li J. Evolution of anaphylatoxins, their diversity and novel roles in innate immunity: Insights from the study of fish complement. Vet Immunol Immunopathol 2005; 108:77-89. [PMID: 16112742 DOI: 10.1016/j.vetimm.2005.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anaphylatoxins are small molecules ( approximately 9 kDa) that are generated as a result of the activation of the complement system. These molecules play an important role in inflammation, and they are responsible for the activation of various innate and adaptive immune processes. The study of these important inflammatory molecules has been restricted to mammalian species so far. Recent studies have shown that teleost fish, unlike any other known animal species, contain multiple forms of the C3a anaphylatoxin, all of which are functionally active and play a prominent role in inducing superoxide production in fish leukocytes. The C5a anaphylatoxin has also been characterized in these animals, and like in mammals, it plays an important role in leukocyte chemotaxis and in triggering the respiratory burst of leukocytes. Interestingly, it has been shown that rainbow trout anaphylatoxins play an unexpected role in enhancing phagocytosis of particles. C5a and C3a receptors have recently been cloned and characterized in rainbow trout, suggesting that the duplication of these receptors from a common ancestor occurred before the emergence of teleosts. The studies derived from these molecules in teleost fish indicate that the basic structure and function of anaphylatoxins and their receptors, have been conserved for more than 300 million years.
Collapse
Affiliation(s)
- J O Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 413 Rosenthal, 3800 Spruce St., Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
41
|
Boshra H, Wang T, Hove-Madsen L, Hansen J, Li J, Matlapudi A, Secombes CJ, Tort L, Sunyer JO. Characterization of a C3a Receptor in Rainbow Trout andXenopus: The First Identification of C3a Receptors in Nonmammalian Species. THE JOURNAL OF IMMUNOLOGY 2005; 175:2427-37. [PMID: 16081814 DOI: 10.4049/jimmunol.175.4.2427] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Virtually nothing is known about the structure, function, and evolutionary origins of the C3aR in nonmammalian species. Because C3aR and C5aR are thought to have arisen from the same common ancestor, the recent characterization of a C5aR in teleost fish implied the presence of a C3aR in this animal group. In this study we report the cloning of a trout cDNA encoding a 364-aa molecule (TC3aR) that shows a high degree of sequence homology and a strong phylogenetic relationship with mammalian C3aRs. Northern blotting demonstrated that TC3aR was expressed primarily in blood leukocytes. Flow cytometric analysis and immunofluorescence microscopy showed that Abs raised against TC3aR stained to a high degree all blood B lymphocytes and, to a lesser extent, all granulocytes. More importantly, these Abs inhibited trout C3a-mediated intracellular calcium mobilization in trout leukocytes. A fascinating structural feature of TC3aR is the lack of a significant portion of the second extracellular loop (ECL2). In all C3aR molecules characterized to date, the ECL2 is exceptionally large when compared with the same region of C5aR. However, the exact function of the extra portion of ECL2 is unknown. The lack of this segment in TC3aR suggests that the extra piece of ECL2 was not necessary for the interaction of the ancestral C3aR with its ligand. Our findings represent the first C3aR characterized in nonmammalian species and support the hypothesis that if C3aR and C5aR diverged from a common ancestor, this event occurred before the emergence of teleost fish.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Blocking/chemistry
- Binding Sites, Antibody
- Blotting, Northern
- Blotting, Southern
- Calcium/antagonists & inhibitors
- Calcium/metabolism
- Complement C3a/antagonists & inhibitors
- Complement C3a/physiology
- Complement Inactivator Proteins/physiology
- DNA, Complementary/isolation & purification
- Fluorescent Antibody Technique, Indirect
- Guinea Pigs
- Humans
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Leukocytes/immunology
- Leukocytes/metabolism
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/immunology
- Membrane Proteins/isolation & purification
- Membrane Proteins/metabolism
- Mice
- Molecular Sequence Data
- Oncorhynchus mykiss
- Rats
- Receptors, Complement/antagonists & inhibitors
- Receptors, Complement/immunology
- Receptors, Complement/isolation & purification
- Receptors, Complement/metabolism
- Sequence Analysis, DNA
- Xenopus
Collapse
Affiliation(s)
- Hani Boshra
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Luchetti CG, Solano ME, Sander V, Arcos MLB, Gonzalez C, Di Girolamo G, Chiocchio S, Cremaschi G, Motta AB. Effects of dehydroepiandrosterone on ovarian cystogenesis and immune function. J Reprod Immunol 2005; 64:59-74. [PMID: 15596227 DOI: 10.1016/j.jri.2004.04.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/22/2004] [Accepted: 04/23/2004] [Indexed: 10/26/2022]
Abstract
The purpose of the present report was to study the possible relationship between ovarian functionality and the immune response during cystogenesis induced by androgenization with dehydroepiandrosterone (DHEA). Daily injection of DHEA (6 mg/kg body weight) for 20 consecutive days induced ovarian cysts in BALB/c mice. As markers of ovarian function, serum estradiol (E) and progesterone (P) and the ovarian inmunomodulator prostaglandin E (PGE) were analyzed. In order to know how the integrity of the tissue was altered after induction of cystogenesis, the oxidative status was also evaluated. Serum E and P levels, and ovarian PGE concentration, were increased in animals with cysts compared with healthy controls. The oxidant status (quantified by malondialdehyde (MDA) formed after the breakdown of the cellular membrane by free radical mechanisms) was augmented, meanwhile the antioxidant (evaluated by the glutathione (GSH) content) diminished during the induction of cystogenesis. Both immunohistochemical and flow cytometry assays demonstrated that DHEA treatment increased the number of T lymphocytes infiltrating ovarian tissue. Therefore, while ovarian controls showed equivalent expression of CD4+ and CD8+ T cell subsets, injection of DHEA yielded a selective ovarian T cell infiltration as demonstrated by enhanced CD8+ and diminished CD4+ T lymphocyte expression. These results show that the development of cysts involves changes in ovarian function and an imbalance in the oxidant-antioxidant equilibrium. We observed also both an increased and selective T lymphocyte infiltration.
Collapse
Affiliation(s)
- Carolina Griselda Luchetti
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Serrano 669, C1414DEM Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ali H, Panettieri RA. Anaphylatoxin C3a receptors in asthma. Respir Res 2005; 6:19. [PMID: 15723703 PMCID: PMC551592 DOI: 10.1186/1465-9921-6-19] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 02/21/2005] [Indexed: 11/30/2022] Open
Abstract
The complement system forms the central core of innate immunity but also mediates a variety of inflammatory responses. Anaphylatoxin C3a, which is generated as a byproduct of complement activation, has long been known to activate mast cells, basophils and eosinophils and to cause smooth muscle contraction. However, the role of C3a in the pathogenesis of allergic asthma remains unclear. In this review, we examine the role of C3a in promoting asthma. Following allergen challenge, C3a is generated in the lung of subjects with asthma but not healthy subjects. Furthermore, deficiency in C3a generation or in G protein coupled receptor for C3a abrogates allergen-induced responses in murine models of pulmonary inflammation and airway hyperresponsiveness. In addition, inhibition of complement activation or administration of small molecule inhibitors of C3a receptor after sensitization but before allergen challenge inhibits airway responses. At a cellular level, C3a stimulates robust mast cell degranulation that is greatly enhanced following cell-cell contact with airway smooth muscle (ASM) cells. Therefore, C3a likely plays an important role in asthma primarily by regulating mast cell-ASM cell interaction.
Collapse
Affiliation(s)
- Hydar Ali
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 South 40Street, Philadelphia, PA, 19104, USA
| | - Reynold A Panettieri
- Pulmonary Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, BRBII/III, 421 Curie Boulevard, Philadelphia PA 19104, USA
| |
Collapse
|
44
|
Boos L, Campbell IL, Ames R, Wetsel RA, Barnum SR. Deletion of the complement anaphylatoxin C3a receptor attenuates, whereas ectopic expression of C3a in the brain exacerbates, experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2004; 173:4708-14. [PMID: 15383607 DOI: 10.4049/jimmunol.173.7.4708] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The C3aR is expressed throughout the CNS and is increased in expression on glial cells during CNS inflammation. However, the role that C3a and the C3aR play in chronic inflammation, such as in the demyelinating disease experimental autoimmune encephalomyelitis (EAE), remains unclear. We show in this study that deletion of the C3aR is protective in myelin oligodendrocyte glycoprotein-induced EAE in C57BL/6 mice. C3aR-deficient (C3aR(-/-)) mice had a significantly attenuated course of EAE compared with control mice during the chronic phase of the disease. Immunohistochemical analysis demonstrated modestly reduced macrophage and T cell infiltration in the spinal cords of C3aR(-/-) mice. To examine the role of C3a in EAE, we developed a transgenic mouse that expresses C3a exclusively in the CNS using the glial fibrillary acidic protein (GFAP) promoter. We observed that C3a/GFAP mice had exacerbated EAE during the chronic phase of the disease, with significant mortality compared with nontransgenic littermates. C3a/GFAP mice had massive meningeal and perivascular infiltration of macrophages and CD4(+) T cells. These studies indicate that C3a may contribute to the pathogenesis of demyelinating disease by directly or indirectly chemoattracting encephalitogenic cells to the CNS.
Collapse
MESH Headings
- Animals
- Brain/immunology
- Brain/metabolism
- Cell Movement/genetics
- Cell Movement/immunology
- Cells, Cultured
- Complement C3a/biosynthesis
- Complement C3a/genetics
- Complement C3a/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Gene Deletion
- Glial Fibrillary Acidic Protein/genetics
- Glycoproteins/administration & dosage
- Humans
- Membrane Proteins/biosynthesis
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin Sheath/pathology
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/administration & dosage
- Receptors, Complement/biosynthesis
- Receptors, Complement/deficiency
- Receptors, Complement/genetics
- Receptors, Complement/physiology
Collapse
Affiliation(s)
- Laura Boos
- Department of Microbiology, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
45
|
Kawamoto S, Yalcindag A, Laouini D, Brodeur S, Bryce P, Lu B, Humbles AA, Oettgen H, Gerard C, Geha RS. The anaphylatoxin C3a downregulates the Th2 response to epicutaneously introduced antigen. J Clin Invest 2004; 114:399-407. [PMID: 15286806 PMCID: PMC484971 DOI: 10.1172/jci19082] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2003] [Accepted: 06/01/2004] [Indexed: 12/16/2022] Open
Abstract
Mechanical injury to the skin results in activation of the complement component C3 and release of the anaphylatoxin C3a. C3a binds to a seven-transmembrane G protein-coupled receptor, C3aR. We used C3aR(-/-) mice to examine the role of C3a in a mouse model of allergic inflammation induced by epicutaneous sensitization with OVA. C3aR(-/-) mice exhibited an exaggerated Th2 response to epicutaneous but not to intraperitoneal sensitization with OVA, as evidenced by significantly elevated levels of serum OVA-specific IgG1 and significantly increased secretion of the Th2 cytokines IL-4, IL-5, and IL-10 by antigen-stimulated splenocytes. Presentation of OVA peptide by C3aR(-/-) APCs caused significantly more IL-4 and IL-5 secretion by T cells from OVA-T cell receptor (OVA-TCR) transgenic mice compared with presentation by WT APCs. C3a inhibited the ability of splenocytes, but not of highly purified T cells, to secrete Th2 cytokines in response to TCR ligation. This inhibition was mediated by IL-12 secreted by APCs in response to C3a. These results suggest that C3a-C3aR interactions inhibit the ability of APCs to drive Th2 cell differentiation in response to epicutaneously introduced antigen and may have important implications for allergic skin diseases.
Collapse
Affiliation(s)
- Seiji Kawamoto
- Division of Immunology, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kawamoto S, Yalcindag A, Laouini D, Brodeur S, Bryce P, Lu B, Humbles AA, Oettgen H, Gerard C, Geha RS. The anaphylatoxin C3a downregulates the Th2 response to epicutaneously introduced antigen. J Clin Invest 2004. [DOI: 10.1172/jci200419082] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|