1
|
Shaughnessy J, Chabeda A, Lewis LA, Ram S. Alternative pathway amplification and infections. Immunol Rev 2023; 313:162-180. [PMID: 36336911 DOI: 10.1111/imr.13160] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The alternative pathway (AP) is the phylogenetically oldest arm of the complement system and may have evolved to mark pathogens for elimination by phagocytes. Studies using purified AP proteins or AP-specific serum showed that C3b amplification on bacteria commenced following a lag phase of about 5 min and was highly dependent on the concentration of complement. Most pathogens have evolved several elegant mechanisms to evade complement, including expressing proteases that degrade AP proteins and secreting proteins that block function of C3 convertases. In an example of convergent evolution, many microbes recruit the AP inhibitor factor H (FH) using molecular mechanisms that mimic FH interactions with host cells. In most instances, the AP serves to amplify C3b deposited on microbes by the classical pathway (CP). The role of properdin on microbes appears to be restricted to stabilization of C3 convertases; scant evidence exists for its role as an initiator of the AP on pathogens in the context of serum. Therapeutic complement inhibition carries with it an increased risk of infection. Antibody (Ab)-dependent AP activation may be critical for complement activation by vaccine-elicited Ab when the CP is blocked, and its molecular mechanism is discussed.
Collapse
Affiliation(s)
- Jutamas Shaughnessy
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Aleyo Chabeda
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Lisa A Lewis
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
2
|
Hahn DL, Webley W. U-BIOPRED/BIOAIR proteins: inflammation or infection? Eur Respir J 2022; 60:2200571. [PMID: 36202417 DOI: 10.1183/13993003.00571-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Affiliation(s)
| | - Wilmore Webley
- University of Massachusetts Amherst, Department of Microbiology, Amherst, MA, USA
| |
Collapse
|
3
|
Cortes C, Desler C, Mazzoli A, Chen JY, Ferreira VP. The role of properdin and Factor H in disease. Adv Immunol 2022; 153:1-90. [PMID: 35469595 DOI: 10.1016/bs.ai.2021.12.001] [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] [Indexed: 01/26/2023]
Abstract
The complement system consists of three pathways (alternative, classical, and lectin) that play a fundamental role in immunity and homeostasis. The multifunctional role of the complement system includes direct lysis of pathogens, tagging pathogens for phagocytosis, promotion of inflammatory responses to control infection, regulation of adaptive cellular immune responses, and removal of apoptotic/dead cells and immune complexes from circulation. A tight regulation of the complement system is essential to avoid unwanted complement-mediated damage to the host. This regulation is ensured by a set of proteins called complement regulatory proteins. Deficiencies or malfunction of these regulatory proteins may lead to pro-thrombotic hematological diseases, renal and ocular diseases, and autoimmune diseases, among others. This review focuses on the importance of two complement regulatory proteins of the alternative pathway, Factor H and properdin, and their role in human diseases with an emphasis on: (a) characterizing the main mechanism of action of Factor H and properdin in regulating the complement system and protecting the host from complement-mediated attack, (b) describing the dysregulation of the alternative pathway as a result of deficiencies, or mutations, in Factor H and properdin, (c) outlining the clinical findings, management and treatment of diseases associated with mutations and deficiencies in Factor H, and (d) defining the unwanted and inadequate functioning of properdin in disease, through a discussion of various experimental research findings utilizing in vitro, mouse and human models.
Collapse
Affiliation(s)
- Claudio Cortes
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States.
| | - Caroline Desler
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Amanda Mazzoli
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Jin Y Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States.
| |
Collapse
|
4
|
Essen MF, Schlagwein N, den Hoven EM, Gijlswijk‐Janssen DJ, Lubbers R, den Bos RM, den Born J, Ruben JM, Trouw LA, Kooten C. Initial properdin binding contributes to alternative pathway activation at the surface of viable and necrotic cells. Eur J Immunol 2022; 52:597-608. [PMID: 35092629 PMCID: PMC9303752 DOI: 10.1002/eji.202149259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022]
Abstract
Properdin, the only known positive regulator of the complement system, stabilizes the C3 convertase, thereby increasing its half‐life. In contrast to most other complement factors, properdin is mainly produced extrahepatically by myeloid cells. Recent data suggest a role for properdin as a pattern recognition molecule. Here, we confirmed previous findings of properdin binding to different necrotic cells including Jurkat T cells. Binding can occur independent of C3, as demonstrated by HAP‐1 C3 KO cells, excluding a role for endogenous C3. In view of the cellular source of properdin, interaction with myeloid cells was examined. Properdin bound to the surface of viable monocyte‐derived pro‐ and anti‐inflammatory macrophages, but not to DCs. Binding was demonstrated for purified properdin as well as fractionated P2, P3, and P4 properdin oligomers. Binding contributed to local complement activation as determined by C3 and C5b‐9 deposition on the cell surfaces and seems a prerequisite for alternative pathway activation. Interaction of properdin with cell surfaces could be inhibited with the tick protein Salp20 and by different polysaccharides, depending on sulfation and chain length. These data identify properdin as a factor interacting with different cell surfaces, being either dead or alive, contributing to the local stimulation of complement activation.
Collapse
Affiliation(s)
- Mieke F. Essen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Nicole Schlagwein
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Elisa M.P. den Hoven
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Daniëlle J. Gijlswijk‐Janssen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Rosalie Lubbers
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| | - Ramon M. den Bos
- Crystal and Structural Chemistry Bijvoet Center for Biomolecular Research Department of Chemistry Faculty of Science Utrecht University Utrecht The Netherlands
| | - Jacob den Born
- Department of Nephrology University Medical Center Groningen Groningen The Netherlands
| | - Jurjen M. Ruben
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Leendert A. Trouw
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
- Department of Immunology Leiden University Medical Center Leiden The Netherlands
| | - Cees Kooten
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | | |
Collapse
|
5
|
Read BJ, Won L, Kraft JC, Sappington I, Aung A, Wu S, Bals J, Chen C, Lee KK, Lingwood D, King NP, Irvine DJ. Mannose-binding lectin and complement mediate follicular localization and enhanced immunogenicity of diverse protein nanoparticle immunogens. Cell Rep 2022; 38:110217. [PMID: 35021101 PMCID: PMC8805147 DOI: 10.1016/j.celrep.2021.110217] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/03/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
Nanoparticle (NP) vaccine formulations promote immune responses through multiple mechanisms. We recently reported that mannose-binding lectin (MBL) triggers trafficking of glycosylated HIV Env-immunogen NPs to lymph node follicles. Here, we investigate effects of MBL and complement on NP forms of HIV and other viral antigens. MBL recognition of oligomannose on gp120 nanoparticles significantly increases antigen accumulation in lymph nodes and antigen-specific germinal center (GC) responses. MBL and complement also mediate follicular trafficking and enhance GC responses to influenza, HBV, and HPV particulate antigens. Using model protein nanoparticles bearing titrated levels of glycosylation, we determine that mannose patches at a minimal density of 2.1 × 10-3 mannose patches/nm2 are required to trigger follicular targeting, which increases with increasing glycan density up to at least ∼8.2 × 10-3 patches/nm2. Thus, innate immune recognition of glycans has a significant impact on humoral immunity, and these findings provide a framework for engineering glycan recognition to optimize vaccine efficacy.
Collapse
Affiliation(s)
- Benjamin J Read
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lori Won
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John C Kraft
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Isaac Sappington
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shengwei Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Julia Bals
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA
| | - Chengbo Chen
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; Biological Physics Structure and Design Program, University of Washington, Seattle, WA 98195, USA
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; Biological Physics Structure and Design Program, University of Washington, Seattle, WA 98195, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
6
|
Varghese PM, Mukherjee S, Al-Mohanna FA, Saleh SM, Almajhdi FN, Beirag N, Alkahtani SH, Rajkumari R, Nal Rogier B, Sim RB, Idicula-Thomas S, Madan T, Murugaiah V, Kishore U. Human Properdin Released By Infiltrating Neutrophils Can Modulate Influenza A Virus Infection. Front Immunol 2021; 12:747654. [PMID: 34956182 PMCID: PMC8695448 DOI: 10.3389/fimmu.2021.747654] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The complement system is designed to recognise and eliminate invading pathogens via activation of classical, alternative and lectin pathways. Human properdin stabilises the alternative pathway C3 convertase, resulting in an amplification loop that leads to the formation of C5 convertase, thereby acting as a positive regulator of the alternative pathway. It has been noted that human properdin on its own can operate as a pattern recognition receptor and exert immune functions outside its involvement in complement activation. Properdin can bind directly to microbial targets via DNA, sulfatides and glycosaminoglycans, apoptotic cells, nanoparticles, and well-known viral virulence factors. This study was aimed at investigating the complement-independent role of properdin against Influenza A virus infection. As one of the first immune cells to arrive at the site of IAV infection, we show here that IAV challenged neutrophils released properdin in a time-dependent manner. Properdin was found to directly interact with haemagglutinin, neuraminidase and matrix 1 protein Influenza A virus proteins in ELISA and western blot. Furthermore, modelling studies revealed that properdin could bind HA and NA of the H1N1 subtype with higher affinity compared to that of H3N2 due to the presence of an HA cleavage site in H1N1. In an infection assay using A549 cells, properdin suppressed viral replication in pH1N1 subtype while promoting replication of H3N2 subtype, as revealed by qPCR analysis of M1 transcripts. Properdin treatment triggered an anti-inflammatory response in H1N1-challenged A549 cells and a pro-inflammatory response in H3N2-infected cells, as evident from differential mRNA expression of TNF-α, NF-κB, IFN-α, IFN-β, IL-6, IL-12 and RANTES. Properdin treatment also reduced luciferase reporter activity in MDCK cells transduced with H1N1 pseudotyped lentiviral particles; however, it was increased in the case of pseudotyped H3N2 particles. Collectively, we conclude that infiltrating neutrophils at the site of IAV infection can release properdin, which then acts as an entry inhibitor for pandemic H1N1 subtype while suppressing viral replication and inducing an anti-inflammatory response. H3N2 subtype can escape this immune restriction due to altered haemagglutinin and neuraminindase, leading to enhanced viral entry, replication and pro-inflammatory response. Thus, depending on the subtype, properdin can either limit or aggravate IAV infection in the host.
Collapse
Affiliation(s)
- Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Shuvechha Mukherjee
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Souad M Saleh
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fahad N Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Saad H Alkahtani
- Department of Zoology, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Beatrice Nal Rogier
- INSERM U1104 Centre d'immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Robert B Sim
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Taruna Madan
- Department of Innate Immunity, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
7
|
Zewde NT, Hsu RV, Morikis D, Palermo G. Systems Biology Modeling of the Complement System Under Immune Susceptible Pathogens. FRONTIERS IN PHYSICS 2021; 9:603704. [PMID: 35145963 PMCID: PMC8827490 DOI: 10.3389/fphy.2021.603704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The complement system is assembled from a network of proteins that function to bring about the first line of defense of the body against invading pathogens. However, complement deficiencies or invasive pathogens can hijack complement to subsequently increase susceptibility of the body to infections. Moreover, invasive pathogens are increasingly becoming resistant to the currently available therapies. Hence, it is important to gain insights into the highly dynamic interaction between complement and invading microbes in the frontlines of immunity. Here, we developed a mathematical model of the complement system composed of 670 ordinary differential equations with 328 kinetic parameters, which describes all three complement pathways (alternative, classical, and lectin) and includes description of mannose-binding lectin, collectins, ficolins, factor H-related proteins, immunoglobulin M, and pentraxins. Additionally, we incorporate two pathogens: (type 1) complement susceptible pathogen and (type 2) Neisseria meningitidis located in either nasopharynx or bloodstream. In both cases, we generate time profiles of the pathogen surface occupied by complement components and the membrane attack complex (MAC). Our model shows both pathogen types in bloodstream are saturated by complement proteins, whereas MACs occupy <<1.0% of the pathogen surface. Conversely, the MAC production in nasopharynx occupies about 1.5-10% of the total N. meningitidis surface, thus making nasal MAC levels at least about eight orders of magnitude higher. Altogether, we predict complement-imbalance, favoring overactivation, is associated with nasopharynx homeostasis. Conversely, orientating toward complement-balance may cause disruption to the nasopharynx homeostasis. Thus, for sporadic meningococcal disease, our model predicts rising nasal levels of complement regulators as early infection biomarkers.
Collapse
Affiliation(s)
- Nehemiah T. Zewde
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| | - Rohaine V. Hsu
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
- Correspondence: Giulia Palermo, , Dimitrios Morikis,
| | - Giulia Palermo
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
- Correspondence: Giulia Palermo, , Dimitrios Morikis,
| |
Collapse
|
8
|
Lausen M, Thomsen ME, Christiansen G, Karred N, Stensballe A, Bennike TB, Birkelund S. Analysis of complement deposition and processing on Chlamydia trachomatis. Med Microbiol Immunol 2020; 210:13-32. [PMID: 33206237 DOI: 10.1007/s00430-020-00695-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022]
Abstract
Chlamydia trachomatis (C. trachomatis) is the leading cause of sexually transmitted bacterial infections worldwide, with over 120 million annual cases. C. trachomatis infections are associated with severe reproductive complications in women such as extrauterine pregnancy and tubal infertility. The infections are often long lasting, associated with immunopathology, and fail to elicit protective immunity which makes recurrent infections common. The immunological mechanisms involved in C. trachomatis infections are only partially understood. Murine infection models suggest that the complement system plays a significant role in both protective immunity and immunopathology during primary Chlamydia infections. However, only limited structural and mechanistic evidence exists on complement-mediated immunity against C. trachomatis. To expand our current knowledge on this topic, we analyzed global complement deposition on C. trachomatis using comprehensive in-depth mass spectrometry-based proteomics. We show that factor B, properdin, and C4b bind to C. trachomatis demonstrating that C. trachomatis-induced complement activation proceeds through at least two activation pathways. Complement activation leads to cleavage and deposition of C3 and C5 activation products, causing initiation of the terminal complement pathway and deposition of C5b, C6, C7, C8, C9 on C. trachomatis. Interestingly, using immunoelectron microscopy, we show that C5b-9 deposition occurred sporadically and only in rare cases formed complete lytic terminal complexes, possibly caused by the presence of the negative regulators vitronectin and clusterin. Finally, cleavage analysis of C3 demonstrated that deposited C3b is degraded to the opsonins iC3b and C3dg and that this complement opsonization facilitates C. trachomatis binding to human B-cells.
Collapse
Affiliation(s)
- Mads Lausen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark.
| | - Mikkel Eggert Thomsen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Gunna Christiansen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark.,Department of Biomedicine, Aarhus University, Wilhelms Meyers Allé 4, 8000, Aarhus, Denmark
| | - Nichlas Karred
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| |
Collapse
|
9
|
Martinez APG, Abreu PAE, de Arruda Vasconcellos S, Ho PL, Ferreira VP, Saggu G, Barbosa AS, Isaac L. The Role of Properdin in Killing of Non-Pathogenic Leptospira biflexa. Front Immunol 2020; 11:572562. [PMID: 33240263 PMCID: PMC7683387 DOI: 10.3389/fimmu.2020.572562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/05/2020] [Indexed: 11/25/2022] Open
Abstract
Properdin (P) is a positive regulatory protein that stabilizes the C3 convertase and C5 convertase of the complement alternative pathway (AP). Several studies have suggested that properdin can bind directly to the surface of certain pathogens regardless of the presence of C3bBb. Saprophytic Leptospira are susceptible to complement-mediated killing, but the interaction of properdin with Leptospira spp. has not been evaluated so far. In this work, we demonstrate that properdin present in normal human serum, purified properdin, as well as properdin oligomers P2, P3, and P4, interact with Leptospira. Properdin can bind directly to the bacterial surface even in the absence of C3b. In line with our previous findings, AP activation was shown to be important for killing non-pathogenic L. biflexa, and properdin plays a key role in this process since this microorganism survives in P-depleted human serum and the addition of purified properdin to P-depleted human serum decreases the number of viable leptospires. A panel of pathogenic L.interrogans recombinant proteins was used to identify putative properdin targets. Lsa30, an outer membrane protein from L. interrogans, binds to unfractionated properdin and to a lesser extent to P2-P4 properdin oligomers. In conclusion, properdin plays an important role in limiting bacterial proliferation of non-pathogenic Leptospira species. Once bound to the leptospiral surface, this positive complement regulatory protein of the AP contributes to the formation of the C3 convertase on the leptospire surface even in the absence of prior addition of C3b.
Collapse
Affiliation(s)
| | | | - Silvio de Arruda Vasconcellos
- Laboratory of Bacterial Zoonoses, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Paulo Lee Ho
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Viviana P. Ferreira
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Gurpanna Saggu
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | | | - Lourdes Isaac
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Zhang J, Song L, Pedersen DV, Li A, Lambris JD, Andersen GR, Mollnes TE, Ma YJ, Garred P. Soluble collectin-12 mediates C3-independent docking of properdin that activates the alternative pathway of complement. eLife 2020; 9:60908. [PMID: 32909942 PMCID: PMC7511233 DOI: 10.7554/elife.60908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/09/2020] [Indexed: 01/11/2023] Open
Abstract
Properdin stabilizes the alternative C3 convertase (C3bBb), whereas its role as pattern-recognition molecule mediating complement activation is disputed for decades. Previously, we have found that soluble collectin-12 (sCL-12) synergizes complement alternative pathway (AP) activation. However, whether this observation is C3 dependent is unknown. By application of the C3-inhibitor Cp40, we found that properdin in normal human serum bound to Aspergillus fumigatus solely in a C3b-dependent manner. Cp40 also prevented properdin binding when properdin-depleted serum reconstituted with purified properdin was applied, in analogy with the findings achieved by C3-depleted serum. However, when opsonized with sCL-12, properdin bound in a C3-independent manner exclusively via its tetrameric structure and directed in situ C3bBb assembly. In conclusion, a prerequisite for properdin binding and in situ C3bBb assembly was the initial docking of sCL-12. This implies a new important function of properdin in host defense bridging pattern recognition and specific AP activation.
Collapse
Affiliation(s)
- Jie Zhang
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lihong Song
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pharmaceutical Science, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dennis V Pedersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Anna Li
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, and University of Oslo, Oslo, Norway.,Research Laboratory, Nordland Hospital, K. G. Jebsen TREC, University of Tromsø, Bodø, Norway.,Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ying Jie Ma
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Chen JY, Galwankar NS, Emch HN, Menon SS, Cortes C, Thurman JM, Merrill SA, Brodsky RA, Ferreira VP. Properdin Is a Key Player in Lysis of Red Blood Cells and Complement Activation on Endothelial Cells in Hemolytic Anemias Caused by Complement Dysregulation. Front Immunol 2020; 11:1460. [PMID: 32793201 PMCID: PMC7387411 DOI: 10.3389/fimmu.2020.01460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
The complement system alternative pathway (AP) can be activated excessively in inflammatory diseases, particularly when there is defective complement regulation. For instance, deficiency in complement regulators CD55 and CD59, leads to paroxysmal nocturnal hemoglobinuria (PNH), whereas Factor H mutations predispose to atypical hemolytic uremic syndrome (aHUS), both causing severe thrombohemolysis. Despite eculizumab being the treatment for these diseases, benefits vary considerably among patients. Understanding the molecular mechanisms involved in complement regulation is essential for developing new treatments. Properdin, the positive AP regulator, is essential for complement amplification by stabilizing enzymatic convertases. In this study, the role of properdin in red blood cell (RBC) lysis and endothelial cell opsonization in these AP-mediated diseases was addressed by developing in vitro assays using PNH patient RBCs and human primary endothelial cells, where the effects of inhibiting properdin, using novel monoclonal antibodies (MoAbs) that we generated and characterized, were compared to other complement inhibitors. In in vitro models of PNH, properdin inhibition prevented hemolysis of patient PNH type II and III RBCs more than inhibition of Factor B, C3, and C5 (>17-fold, or >81-fold, or >12-fold lower molar IC90 values, respectively). When tested in an in vitro aHUS hemolysis model, the anti-properdin MoAbs had 11-fold, and 86-fold lower molar IC90 values than inhibition of Factor B, or C3, respectively (P < 0.0001). When comparing target/inhibitor ratios in all hemolysis assays, inhibiting properdin was at least as efficient as the other complement inhibitors in most cases. In addition, using in vitro endothelial cell assays, the data indicate a critical novel role for properdin in promoting complement activation on human endothelial cells exposed to heme (a hemolysis by-product) and rH19-20 (to inhibit Factor H cell-surface protection), as occurs in aHUS. Inhibition of properdin or C3 in this system significantly reduced C3 fragment deposition by 75%. Altogether, the data indicate properdin is key in promoting RBC lysis and complement activation on human endothelial cells, contributing to the understanding of PNH and aHUS pathogenesis. Further studies to determine therapeutic values of inhibiting properdin in complement-mediated diseases, in particular those that are characterized by AP dysregulation, are warranted.
Collapse
Affiliation(s)
- Jin Y Chen
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Neeti S Galwankar
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Heather N Emch
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Smrithi S Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Claudio Cortes
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Joshua M Thurman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Samuel A Merrill
- Section of Hematology/Oncology, Department of Medicine, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Robert A Brodsky
- Division of Hematology, Department of Medicine, John Hopkins University School of Medicine, Baltimore, MD, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
12
|
Keb G, Fields KA. An Ancient Molecular Arms Race: Chlamydia vs. Membrane Attack Complex/Perforin (MACPF) Domain Proteins. Front Immunol 2020; 11:1490. [PMID: 32760406 PMCID: PMC7371996 DOI: 10.3389/fimmu.2020.01490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022] Open
Abstract
Dynamic interactions that govern the balance between host and pathogen determine the outcome of infection and are shaped by evolutionary pressures. Eukaryotic hosts have evolved elaborate and formidable defense mechanisms that provide the basis for innate and adaptive immunity. Proteins containing a membrane attack complex/Perforin (MACPF) domain represent an important class of immune effectors. These pore-forming proteins induce cell killing by targeting microbial or host membranes. Intracellular bacteria can be shielded from MACPF-mediated killing, and Chlamydia spp. represent a successful paradigm of obligate intracellular parasitism. Ancestors of present-day Chlamydia likely originated at evolutionary times that correlated with or preceded many host defense pathways. We discuss the current knowledge regarding how chlamydiae interact with the MACPF proteins Complement C9, Perforin-1, and Perforin-2. Current evidence indicates a degree of resistance by Chlamydia to MACPF effector mechanisms. In fact, chlamydiae have acquired and adapted their own MACPF-domain protein to facilitate infection.
Collapse
Affiliation(s)
- Gabrielle Keb
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Kenneth A Fields
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States
| |
Collapse
|
13
|
Pedersen DV, Rösner T, Hansen AG, Andersen KR, Thiel S, Andersen GR, Valerius T, Laursen NS. Recruitment of properdin by bi-specific nanobodies activates the alternative pathway of complement. Mol Immunol 2020; 124:200-210. [PMID: 32599335 DOI: 10.1016/j.molimm.2020.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/28/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
The complement system represents a powerful part of the innate immune system capable of removing pathogens and damaged host cells. Nevertheless, only a subset of therapeutic antibodies are capable of inducing complement dependent cytotoxicity, which has fuelled the search for new strategies to potentiate complement activation. Properdin (FP) functions as a positive complement regulator by stabilizing the alternative pathway C3 convertase. Here, we explore a novel strategy for direct activation of the alternative pathway of complement using bi-specific single domain antibodies (nanobodies) that recruit endogenous FP to a cell surface. As a proof-of-principle, we generated bi-specific nanobodies with specificity toward FP and the validated cancer antigen epidermal growth factor receptor (EGFR) and tested their ability to activate complement onto cancer cell lines expressing EGFR. Treatment led to recruitment of FP, complement activation and significant deposition of C3 fragments on the cells in a manner sensitive to the geometry of FP recruitment. The bi-specific nanobodies induced complement dependent lysis of baby hamster kidney cells expressing human EGFR but were unable to lyse human tumour cells due to the presence of complement regulators. Our results confirm that FP can function as a surface bound focal point for initiation of complement activation independent of prior C3b deposition. However, recruitment of FP by bi-specific nanobodies appears insufficient for overcoming the inhibitory action of the negative complement regulators overexpressed by many human tumour cell lines. Our data provide general information on the efficacy of properdin as an initiator of complement but suggest that properdin recruitment on its own may have limited utility as a platform for potent complement activation on regulated cell surfaces.
Collapse
Affiliation(s)
- Dennis V Pedersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Gustav Wieds vej 10 C, 8000 Aarhus C, Denmark
| | - Thies Rösner
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University, Rosalind-Franklin-Straße 12, 24103 Kiel, Germany
| | - Annette G Hansen
- Department of Biomedicine, Aarhus University, Høgh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Kasper R Andersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Gustav Wieds vej 10 C, 8000 Aarhus C, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Høgh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Gustav Wieds vej 10 C, 8000 Aarhus C, Denmark
| | - Thomas Valerius
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University, Rosalind-Franklin-Straße 12, 24103 Kiel, Germany
| | - Nick S Laursen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Gustav Wieds vej 10 C, 8000 Aarhus C, Denmark.
| |
Collapse
|
14
|
Opsonophagocytosis of Chlamydia pneumoniae by Human Monocytes and Neutrophils. Infect Immun 2020; 88:IAI.00087-20. [PMID: 32284372 DOI: 10.1128/iai.00087-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 01/08/2023] Open
Abstract
The human respiratory tract pathogen Chlamydia pneumoniae, which causes mild to severe infections, has been associated with the development of chronic inflammatory diseases. To understand the biology of C. pneumoniae infections, several studies have investigated the interaction between C. pneumoniae and professional phagocytes. However, these studies have been conducted under nonopsonizing conditions, making the role of opsonization in C. pneumoniae infections elusive. Thus, we analyzed complement and antibody opsonization of C. pneumoniae and evaluated how opsonization affects chlamydial infectivity and phagocytosis in human monocytes and neutrophils. We demonstrated that IgG antibodies and activation products of complement C3 and C4 are deposited on the surface of C. pneumoniae elementary bodies when incubated in human serum. Complement activation limits C. pneumoniae infectivity in vitro and has the potential to induce bacterial lysis by the formation of the membrane attack complex. Coculture of C. pneumoniae and freshly isolated human leukocytes showed that complement opsonization is superior to IgG opsonization for efficient opsonophagocytosis of C. pneumoniae in monocytes and neutrophils. Neutrophil-mediated phagocytosis of C. pneumoniae was crucially dependent on opsonization, while monocytes retained minor phagocytic potential under nonopsonizing conditions. Complement opsonization significantly enhanced the intracellular neutralization of C. pneumoniae in peripheral blood mononuclear cells and neutrophils and almost abrogated the infectious potential of C. pneumoniae In conclusion, we demonstrated that complements limit C. pneumoniae infection in vitro by interfering with C. pneumoniae entry into permissive cells by direct complement-induced lysis and by tagging bacteria for efficient phagocytosis in both monocytes and neutrophils.
Collapse
|
15
|
Kaposi's Sarcoma-Associated Herpesvirus and Host Interaction by the Complement System. Pathogens 2020; 9:pathogens9040260. [PMID: 32260199 PMCID: PMC7237997 DOI: 10.3390/pathogens9040260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/19/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) modulates the immune response to allow the virus to establish persistent infection in the host and facilitate the development of KSHV-associated cancer. The complement system has a central role in the defense against pathogens. Hence, KSHV has adopted an evasion strategy for complement attack using the viral protein encoded by KSHV open reading frame 4. However, despite this defense mechanism, the complement system appears to become activated in KSHV-infected cells as well as in the region surrounding Kaposi’s sarcoma tumors. Given that the complement system can affect cell fate as well as the inflammatory microenvironment, complement activation is likely associated with KSHV pathogenesis. A better understanding of the interplay between KSHV and the complement system may, therefore, translate into the development of novel therapeutic interventions for KSHV-associated tumors. In this review, the mechanisms and functions of complement activation in KSHV-infected cells are discussed.
Collapse
|
16
|
Michels MAHM, Volokhina EB, van de Kar NCAJ, van den Heuvel LPWJ. The role of properdin in complement-mediated renal diseases: a new player in complement-inhibiting therapy? Pediatr Nephrol 2019; 34:1349-1367. [PMID: 30141176 PMCID: PMC6579773 DOI: 10.1007/s00467-018-4042-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/16/2022]
Abstract
Properdin is known as the only positive regulator of the complement system. Properdin promotes the activity of this defense system by stabilizing its key enzymatic complexes: the complement alternative pathway (AP) convertases. Besides, some studies have indicated a role for properdin as an initiator of complement activity. Though the AP is a powerful activation route of the complement system, it is also involved in a wide variety of autoimmune and inflammatory diseases, many of which affect the kidneys. The role of properdin in regulating complement in health and disease has not received as much appraisal as the many negative AP regulators, such as factor H. Historically, properdin deficiency has been strongly associated with an increased risk for meningococcal disease. Yet only recently had studies begun to link properdin to other complement-related diseases, including renal diseases. In the light of the upcoming complement-inhibiting therapies, it is interesting whether properdin can be a therapeutic target to attenuate AP-mediated injury. A full understanding of the basic concepts of properdin biology is therefore needed. Here, we first provide an overview of the function of properdin in health and disease. Then, we explore its potential as a therapeutic target for the AP-associated renal diseases C3 glomerulopathy, atypical hemolytic uremic syndrome, and proteinuria-induced tubulointerstitial injury. Considering current knowledge, properdin-inhibiting therapy seems promising in certain cases. However, knowing the complexity of properdin's role in renal pathologies in vivo, further research is required to clarify the exact potential of properdin-targeted therapy in complement-mediated renal diseases.
Collapse
Affiliation(s)
- Marloes A. H. M. Michels
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands
| | - Elena B. Volokhina
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands ,Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands
| | - Nicole C. A. J. van de Kar
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands
| | - Lambertus P. W. J. van den Heuvel
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands ,Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6525 GA Nijmegen, The Netherlands ,Department of Pediatrics/Pediatric Nephrology and Department of Development & Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
17
|
Heesterbeek DAC, Angelier ML, Harrison RA, Rooijakkers SHM. Complement and Bacterial Infections: From Molecular Mechanisms to Therapeutic Applications. J Innate Immun 2018; 10:455-464. [PMID: 30149378 PMCID: PMC6784045 DOI: 10.1159/000491439] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/21/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022] Open
Abstract
Complement is a complex protein network of plasma, and an integral part of the innate immune system. Complement activation results in the rapid clearance of bacteria by immune cells, and direct bacterial killing via large pore-forming complexes. Here we review important recent discoveries in the complement field, focusing on interactions relevant for the defense against bacteria. Understanding the molecular interplay between complement and bacteria is of great importance for future therapies for infectious and inflammatory diseases. Antibodies that support complement-dependent bacterial killing are of interest for the development of alternative therapies to treat infections with antibiotic-resistant bacteria. Furthermore, a variety of novel therapeutic complement inhibitors have been developed to prevent unwanted complement activation in autoimmune inflammatory diseases. A better understanding of how such inhibitors may increase the risk of bacterial infections is essential if such therapies are to be successful.
Collapse
Affiliation(s)
- Dani A C Heesterbeek
- Department of Medical Microbiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Mathieu L Angelier
- Department of Medical Microbiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Richard A Harrison
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
18
|
Chen JY, Cortes C, Ferreira VP. Properdin: A multifaceted molecule involved in inflammation and diseases. Mol Immunol 2018; 102:58-72. [PMID: 29954621 DOI: 10.1016/j.molimm.2018.05.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/16/2018] [Accepted: 05/23/2018] [Indexed: 01/17/2023]
Abstract
Properdin, the widely known positive regulator of the alternative pathway (AP), has undergone significant investigation over the last decade to define its function in inflammation and disease, including its role in arthritis, asthma, and kidney and cardiovascular diseases. Properdin is a glycoprotein found in plasma that is mainly produced by leukocytes and can positively regulate AP activity by stabilizing C3 and C5 convertases and initiating the AP. Promotion of complement activity by properdin results in changes in the cellular microenvironment that contribute to innate and adaptive immune responses, including pro-inflammatory cytokine production, immune cell infiltration, antigen presenting cell maturation, and tissue damage. The use of properdin-deficient mouse models and neutralizing antibodies has contributed to the understanding of the mechanisms by which properdin contributes to promoting or preventing disease pathology. This review mainly focusses on the multifaceted roles of properdin in inflammation and diseases, and how understanding these roles is contributing to the development of new disease therapies.
Collapse
Affiliation(s)
- Jin Y Chen
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States.
| | - Claudio Cortes
- Department of Biomedical Sciences, University of Oakland University School of Medicine, Rochester, MI, United States.
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States.
| |
Collapse
|
19
|
Al-Mozaini MA, Tsolaki AG, Abdul-Aziz M, Abozaid SM, Al-Ahdal MN, Pathan AA, Murugaiah V, Makarov EM, Kaur A, Sim RB, Kishore U, Kouser L. Human Properdin Modulates Macrophage: Mycobacterium bovis BCG Interaction via Thrombospondin Repeats 4 and 5. Front Immunol 2018; 9:533. [PMID: 29867915 PMCID: PMC5951972 DOI: 10.3389/fimmu.2018.00533] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/01/2018] [Indexed: 02/01/2023] Open
Abstract
Mycobacterium tuberculosis can proficiently enter macrophages and diminish complement activation on its cell surface. Within macrophages, the mycobacterium can suppress macrophage apoptosis and survive within the intracellular environment. Previously, we have shown that complement regulatory proteins such as factor H may interfere with pathogen–macrophage interactions during tuberculosis infection. In this study, we show that Mycobacterium bovis BCG binds properdin, an upregulator of the complement alternative pathway. TSR4+5, a recombinant form of thrombospondin repeats 4 and 5 of human properdin expressed in tandem, which is an inhibitor of the alternative pathway, was also able to bind to M. bovis BCG. Properdin and TSR4+5 were found to inhibit uptake of M. bovis BCG by THP-1 macrophage cells in a dose-dependent manner. Quantitative real-time PCR revealed elevated pro-inflammatory responses (TNF-α, IL-1β, and IL-6) in the presence of properdin or TSR4+5, which gradually decreased over 6 h. Correspondingly, anti-inflammatory responses (IL-10 and TGF-β) showed suppressed levels of expression in the presence of properdin, which gradually increased over 6 h. Multiplex cytokine array analysis also revealed that properdin and TSR4+5 significantly enhanced the pro-inflammatory response (TNF-α, IL-1β, and IL-1α) at 24 h, which declined at 48 h, whereas the anti-inflammatory response (IL-10) was suppressed. Our results suggest that properdin may interfere with mycobacterial entry into macrophages via TSR4 and TSR5, particularly during the initial stages of infection, thus affecting the extracellular survival of the pathogen. This study offers novel insights into the non-complement related functions of properdin during host–pathogen interactions in tuberculosis.
Collapse
Affiliation(s)
- Maha Ahmed Al-Mozaini
- College of Health and Life Sciences, Brunel University London, London, United Kingdom.,Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Anthony G Tsolaki
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Munirah Abdul-Aziz
- College of Health and Life Sciences, Brunel University London, London, United Kingdom.,Department of Biochemistry, Oxford University, Oxford, United Kingdom
| | - Suhair M Abozaid
- College of Health and Life Sciences, Brunel University London, London, United Kingdom.,Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohammed N Al-Ahdal
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ansar A Pathan
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Valarmathy Murugaiah
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Evgeny M Makarov
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Anuvinder Kaur
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Robert B Sim
- Department of Biochemistry, Oxford University, Oxford, United Kingdom
| | - Uday Kishore
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Lubna Kouser
- College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
20
|
Kouser L, Paudyal B, Kaur A, Stenbeck G, Jones LA, Abozaid SM, Stover CM, Flahaut E, Sim RB, Kishore U. Human Properdin Opsonizes Nanoparticles and Triggers a Potent Pro-inflammatory Response by Macrophages without Involving Complement Activation. Front Immunol 2018; 9:131. [PMID: 29483907 PMCID: PMC5816341 DOI: 10.3389/fimmu.2018.00131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/16/2018] [Indexed: 12/28/2022] Open
Abstract
Development of nanoparticles as tissue-specific drug delivery platforms can be considerably influenced by the complement system because of their inherent pro-inflammatory and tumorigenic consequences. The complement activation pathways, and its recognition subcomponents, can modulate clearance of the nanoparticles and subsequent inflammatory response and thus alter the intended translational applications. Here, we report, for the first time, that human properdin, an upregulator of the complement alternative pathway, can opsonize functionalized carbon nanotubes (CNTs) via its thrombospondin type I repeat (TSR) 4 and 5. Binding of properdin and TSR4+5 is likely to involve charge pattern/polarity recognition of the CNT surface since both carboxymethyl cellulose-coated carbon nanotubes (CMC-CNT) and oxidized (Ox-CNT) bound these proteins well. Properdin enhanced the uptake of CMC-CNTs by a macrophage cell line, THP-1, mounting a robust pro-inflammatory immune response, as revealed by qRT-PCR, multiplex cytokine array, and NF-κB nuclear translocation analyses. Properdin can be locally synthesized by immune cells in an inflammatory microenvironment, and thus, its interaction with nanoparticles is of considerable importance. In addition, recombinant TSR4+5 coated on the CMC-CNTs inhibited complement consumption by CMC-CNTs, suggesting that nanoparticle decoration with TSR4+5, can be potentially used as a complement inhibitor in a number of pathological contexts arising due to exaggerated complement activation.
Collapse
Affiliation(s)
- Lubna Kouser
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Basudev Paudyal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Faculty of Science, Engineering and Computing, Kingston University, Kingston upon Thames, Surrey, United Kingdom
| | - Anuvinder Kaur
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Gudrun Stenbeck
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Lucy A. Jones
- Faculty of Science, Engineering and Computing, Kingston University, Kingston upon Thames, Surrey, United Kingdom
| | - Suhair M. Abozaid
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Cordula M. Stover
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Emmanuel Flahaut
- Université de Toulouse, CNRS, INPT, UPS, UMR CNRS-UPS-INP N°5085, 3 Paul Sabatier, Bât. CIRIMAT, Toulouse, France
| | - Robert B. Sim
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
21
|
Harrison RA. The properdin pathway: an "alternative activation pathway" or a "critical amplification loop" for C3 and C5 activation? Semin Immunopathol 2017; 40:15-35. [PMID: 29167939 DOI: 10.1007/s00281-017-0661-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/30/2017] [Indexed: 11/28/2022]
Abstract
This review is not intended to cover in detail all aspects of the discovery and evolution of our understanding of the "alternative pathway" of complement activation, there are many excellent reviews that do this (see Fearon (CRC Crit Rev Immunol 1:1-32, 1979), Pangburn and Müller-Eberhard (Springer Semin Immunopathol 7:163-192, 1984)), but instead to give sufficient background for current concepts to be put in context. The prevailing textbook view, of components having a primary role as an alternative "pathway" for C3 activation, is challenged, with an argument developed for the primary role of the system being that of providing a surface-dependent amplification loop for both C3 and C5 activation. Whatever the mechanism by which the initial C3b molecule is generated, deposition onto a surface has the potential to target that surface for elimination. Elimination or escape from initial targeting is determined by a sophisticated and highly regulated amplification loop for C3 activation. This viewpoint of the system is then briefly developed to provide a context for therapeutic treatment of disease caused, at least in part, by dysregulated amplification of C3 activation, and to highlight some of the challenges that such therapies will face and need to address.
Collapse
Affiliation(s)
- Richard A Harrison
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
22
|
Ramírez-Toloza G, Ferreira A. Trypanosoma cruzi Evades the Complement System as an Efficient Strategy to Survive in the Mammalian Host: The Specific Roles of Host/Parasite Molecules and Trypanosoma cruzi Calreticulin. Front Microbiol 2017; 8:1667. [PMID: 28919885 PMCID: PMC5585158 DOI: 10.3389/fmicb.2017.01667] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/17/2017] [Indexed: 12/20/2022] Open
Abstract
American Trypanosomiasis is an important neglected reemerging tropical parasitism, infecting about 8 million people worldwide. Its agent, Trypanosoma cruzi, exhibits multiple mechanisms to evade the host immune response and infect host cells. An important immune evasion strategy of T. cruzi infective stages is its capacity to inhibit the complement system activation on the parasite surface, avoiding opsonizing, immune stimulating and lytic effects. Epimastigotes, the non-infective form of the parasite, present in triatomine arthropod vectors, are highly susceptible to complement-mediated lysis while trypomastigotes, the infective form, present in host bloodstream, are resistant. Thus T. cruzi susceptibility to complement varies depending on the parasite stage (amastigote, trypomastigotes or epimastigote) and on the T. cruzi strain. To avoid complement-mediated lysis, T. cruzi trypomastigotes express on the parasite surface a variety of complement regulatory proteins, such as glycoprotein 58/68 (gp58/68), T. cruzi complement regulatory protein (TcCRP), trypomastigote decay-accelerating factor (T-DAF), C2 receptor inhibitor trispanning (CRIT) and T. cruzi calreticulin (TcCRT). Alternatively, or concomitantly, the parasite captures components with complement regulatory activity from the host bloodstream, such as factor H (FH) and plasma membrane-derived vesicles (PMVs). All these proteins inhibit different steps of the classical (CP), alternative (AP) or lectin pathways (LP). Thus, TcCRP inhibits the CP C3 convertase assembling, gp58/68 inhibits the AP C3 convertase, T-DAF interferes with the CP and AP convertases assembling, TcCRT inhibits the CP and LP, CRIT confers ability to resist the CP and LP, FH is used by trypomastigotes to inhibit the AP convertases and PMVs inhibit the CP and LP C3 convertases. Many of these proteins have similar molecular inhibitory mechanisms. Our laboratory has contributed to elucidate the role of TcCRT in the host-parasite interplay. Thus, we have proposed that TcCRT is a pleiotropic molecule, present not only in the parasite endoplasmic reticulum, but also on the trypomastigote surface, participating in key processes to establish T. cruzi infection, such as inhibition of the complement system and serving as an important virulence factor. Additionally, TcCRT interaction with key complement components, participates as an anti-angiogenic and anti-tumor molecule, inhibiting at least in important part, tumor growth in infected animals.
Collapse
Affiliation(s)
- Galia Ramírez-Toloza
- Laboratory of Parasitology, Department of Animal Preventive Medicine, Faculty of Veterinary Medicine and Livestock Sciences, University of ChileSantiago, Chile
| | - Arturo Ferreira
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of ChileSantiago, Chile
| |
Collapse
|
23
|
Schatz-Jakobsen JA, Pedersen DV, Andersen GR. Structural insight into proteolytic activation and regulation of the complement system. Immunol Rev 2017; 274:59-73. [PMID: 27782336 DOI: 10.1111/imr.12465] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The complement system is a highly complex and carefully regulated proteolytic cascade activated through three different pathways depending on the activator recognized. The structural knowledge regarding the intricate proteolytic enzymes that activate and control complement has increased dramatically over the last decade. This development has been pivotal for understanding how mutations within complement proteins might contribute to pathogenesis and has spurred new strategies for development of complement therapeutics. Here we describe and discuss the complement system from a structural perspective and integrate the most recent findings obtained by crystallography, small-angle X-ray scattering, and electron microscopy. In particular, we focus on the proteolytic enzymes governing activation and their products carrying the biological effector functions. Additionally, we present the structural basis for some of the best known complement inhibitors. The large number of accumulated molecular structures enables us to visualize the relative size, position, and overall orientation of many of the most interesting complement proteins and assembled complexes on activator surfaces and in membranes.
Collapse
Affiliation(s)
| | - Dennis V Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
24
|
Blatt AZ, Pathan S, Ferreira VP. Properdin: a tightly regulated critical inflammatory modulator. Immunol Rev 2017; 274:172-190. [PMID: 27782331 PMCID: PMC5096056 DOI: 10.1111/imr.12466] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The complement alternative pathway is a powerful arm of the innate immune system that enhances diverse inflammatory responses in the human host. Key to the effects of the alternative pathway is properdin, a serum glycoprotein that can both initiate and positively regulate alternative pathway activity. Properdin is produced by many different leukocyte subsets and circulates as cyclic oligomers of monomeric subunits. While the formation of non‐physiological aggregates in purified properdin preparations and the presence of potential properdin inhibitors in serum have complicated studies of its function, properdin has, regardless, emerged as a key player in various inflammatory disease models. Here, we review basic properdin biology, emphasizing the major hurdles that have complicated the interpretation of results from properdin‐centered studies. In addition, we elaborate on an emerging role for properdin in thromboinflammation and discuss the potential utility of properdin inhibitors as long‐term therapeutic options to treat diseases marked by increased formation of platelet/granulocyte aggregates. Finally, we describe the interplay between properdin and the alternative pathway negative regulator, Factor H, and how aiming to understand these interactions can provide scientists with the most effective ways to manipulate alternative pathway activation in complex systems.
Collapse
Affiliation(s)
- Adam Z Blatt
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sabina Pathan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
25
|
Pedersen DV, Roumenina L, Jensen RK, Gadeberg TA, Marinozzi C, Picard C, Rybkine T, Thiel S, Sørensen UB, Stover C, Fremeaux-Bacchi V, Andersen GR. Functional and structural insight into properdin control of complement alternative pathway amplification. EMBO J 2017; 36:1084-1099. [PMID: 28264884 DOI: 10.15252/embj.201696173] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/16/2017] [Accepted: 01/31/2017] [Indexed: 12/22/2022] Open
Abstract
Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.
Collapse
Affiliation(s)
- Dennis V Pedersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Lubka Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Rasmus K Jensen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Trine Af Gadeberg
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Chiara Marinozzi
- Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Capucine Picard
- Centre d'études des déficits immunitaires, CHU Paris - Hôpital Necker-Enfants Malades, Paris, France
| | - Tania Rybkine
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Cordula Stover
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Veronique Fremeaux-Bacchi
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Properdin binding to complement activating surfaces depends on initial C3b deposition. Proc Natl Acad Sci U S A 2017; 114:E534-E539. [PMID: 28069958 DOI: 10.1073/pnas.1612385114] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Two functions have been assigned to properdin; stabilization of the alternative convertase, C3bBb, is well accepted, whereas the role of properdin as pattern recognition molecule is controversial. The presence of nonphysiological aggregates in purified properdin preparations and experimental models that do not allow discrimination between the initial binding of properdin and binding secondary to C3b deposition is a critical factor contributing to this controversy. In previous work, by inhibiting C3, we showed that properdin binding to zymosan and Escherichia coli is not a primary event, but rather is solely dependent on initial C3 deposition. In the present study, we found that properdin in human serum bound dose-dependently to solid-phase myeloperoxidase. This binding was dependent on C3 activation, as demonstrated by the lack of binding in human serum with the C3-inhibitor compstatin Cp40, in C3-depleted human serum, or when purified properdin is applied in buffer. Similarly, binding of properdin to the surface of human umbilical vein endothelial cells or Neisseria meningitidis after incubation with human serum was completely C3-dependent, as detected by flow cytometry. Properdin, which lacks the structural homology shared by other complement pattern recognition molecules and has its major function in stabilizing the C3bBb convertase, was found to bind both exogenous and endogenous molecular patterns in a completely C3-dependent manner. We therefore challenge the view of properdin as a pattern recognition molecule, and argue that the experimental conditions used to test this hypothesis should be carefully considered, with emphasis on controlling initial C3 activation under physiological conditions.
Collapse
|
27
|
Kouser L, Abdul-Aziz M, Tsolaki AG, Singhal D, Schwaeble WJ, Urban BC, Khan HA, Sim RB, Kishore U. A recombinant two-module form of human properdin is an inhibitor of the complement alternative pathway. Mol Immunol 2016; 73:76-87. [PMID: 27060503 DOI: 10.1016/j.molimm.2016.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 02/08/2023]
Abstract
Properdin upregulates the alternative complement pathway by binding and stabilising the C3 convertase complex (C3bBb). Properdin is a soluble glycoprotein and its flexible rod-like 53kDa monomers form cyclic polymers (dimers, trimers, tetramers and pentamers). The properdin monomer consists of seven thrombospondin type I repeats (TSR 0-6), which are similar and homologous to domains found in circumsporozoite and thrombospondin-related anonymous proteins of Plasmodium species, ETP100 of Eimeria tenella, various complement components C6-C9, and thrombospondin I and II. Using deletion constructs, TSR4 and TSR5 of human properdin were implicated in C3b binding and stabilising C3 convertase. However, individually expressed TSR4 or TSR5 failed to bind properdin ligands. Here, we have expressed and characterized biologically active TSR4 and TSR5 together (TSR4+5) in tandem in Escherichia coli, fused to maltose-binding protein. MBP-TSR4+5 bind solid-phase C3b, sulfatides and glycosaminoglycans. In addition, functionally active recombinant TSR4+5 modules inhibit the alternative pathway of complement.
Collapse
Affiliation(s)
- Lubna Kouser
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Munirah Abdul-Aziz
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Anthony G Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Dipti Singhal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Wilhelm J Schwaeble
- Department of Infection, Immunity and Inflammation, Maurice Shock Medical Sciences Building, University Road, Leicester, LE1 9HN, UK
| | - Britta C Urban
- Liverpool School of Tropical Medicine, Pembroke Place Liverpool, L3 5QA, UK
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Robert B Sim
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; Department of Infection, Immunity and Inflammation, Maurice Shock Medical Sciences Building, University Road, Leicester, LE1 9HN, UK
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
28
|
Cagliani R, Forni D, Filippi G, Mozzi A, De Gioia L, Pontremoli C, Pozzoli U, Bresolin N, Clerici M, Sironi M. The mammalian complement system as an epitome of host-pathogen genetic conflicts. Mol Ecol 2016; 25:1324-39. [PMID: 26836579 DOI: 10.1111/mec.13558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/29/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022]
Abstract
The complement system is an innate immunity effector mechanism; its action is antagonized by a wide array of pathogens and complement evasion determines the virulence of several infections. We investigated the evolutionary history of the complement system and of bacterial-encoded complement-interacting proteins. Complement components targeted by several pathogens evolved under strong selective pressure in primates, with selection acting on residues at the contact interface with microbial/viral proteins. Positively selected sites in CFH and C4BPA account for the human specificity of gonococcal infection. Bacterial interactors, evolved adaptively as well, with selected sites located at interaction surfaces with primate complement proteins. These results epitomize the expectation under a genetic conflict scenario whereby the host's and the pathogen's genes evolve within binding avoidance-binding seeking dynamics. In silico mutagenesis and protein-protein docking analyses supported this by showing that positively selected sites, both in the host's and in the pathogen's interacting partner, modulate binding.
Collapse
Affiliation(s)
- Rachele Cagliani
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| | - Diego Forni
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| | - Giulia Filippi
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126, Milan, Italy
| | - Alessandra Mozzi
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126, Milan, Italy
| | - Chiara Pontremoli
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| | - Uberto Pozzoli
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| | - Nereo Bresolin
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy.,Dino Ferrari Centre, Department of Physiopathology and Transplantation, University of Milan, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Mario Clerici
- Department of Physiopathology and Transplantation, University of Milan, 20090, Milan, Italy.,Don C. Gnocchi Foundation ONLUS, IRCCS, 20148, Milan, Italy
| | - Manuela Sironi
- Bioinformatics, Scientific Institute IRCCS E. MEDEA, 23842, Bosisio Parini, Italy
| |
Collapse
|
29
|
Zewde N, Gorham RD, Dorado A, Morikis D. Quantitative Modeling of the Alternative Pathway of the Complement System. PLoS One 2016; 11:e0152337. [PMID: 27031863 PMCID: PMC4816337 DOI: 10.1371/journal.pone.0152337] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/11/2016] [Indexed: 12/26/2022] Open
Abstract
The complement system is an integral part of innate immunity that detects and eliminates invading pathogens through a cascade of reactions. The destructive effects of the complement activation on host cells are inhibited through versatile regulators that are present in plasma and bound to membranes. Impairment in the capacity of these regulators to function in the proper manner results in autoimmune diseases. To better understand the delicate balance between complement activation and regulation, we have developed a comprehensive quantitative model of the alternative pathway. Our model incorporates a system of ordinary differential equations that describes the dynamics of the four steps of the alternative pathway under physiological conditions: (i) initiation (fluid phase), (ii) amplification (surfaces), (iii) termination (pathogen), and (iv) regulation (host cell and fluid phase). We have examined complement activation and regulation on different surfaces, using the cellular dimensions of a characteristic bacterium (E. coli) and host cell (human erythrocyte). In addition, we have incorporated neutrophil-secreted properdin into the model highlighting the cross talk of neutrophils with the alternative pathway in coordinating innate immunity. Our study yields a series of time-dependent response data for all alternative pathway proteins, fragments, and complexes. We demonstrate the robustness of alternative pathway on the surface of pathogens in which complement components were able to saturate the entire region in about 54 minutes, while occupying less than one percent on host cells at the same time period. Our model reveals that tight regulation of complement starts in fluid phase in which propagation of the alternative pathway was inhibited through the dismantlement of fluid phase convertases. Our model also depicts the intricate role that properdin released from neutrophils plays in initiating and propagating the alternative pathway during bacterial infection.
Collapse
Affiliation(s)
- Nehemiah Zewde
- Department of Bioengineering, University of California Riverside, Riverside, California, United States of America
| | - Ronald D. Gorham
- Department of Bioengineering, University of California Riverside, Riverside, California, United States of America
| | - Angel Dorado
- Department of Mechanical Engineering, University of California Riverside, Riverside, California, United States of America
| | - Dimitrios Morikis
- Department of Bioengineering, University of California Riverside, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Wang Y, Miwa T, Ducka-Kokalari B, Redai IG, Sato S, Gullipalli D, Zangrilli JG, Haczku A, Song WC. Properdin Contributes to Allergic Airway Inflammation through Local C3a Generation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1171-81. [PMID: 26116506 DOI: 10.4049/jimmunol.1401819] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 05/23/2015] [Indexed: 01/04/2023]
Abstract
Complement is implicated in asthma pathogenesis, but its mechanism of action in this disease remains incompletely understood. In this study, we investigated the role of properdin (P), a positive alternative pathway complement regulator, in allergen-induced airway inflammation. Allergen challenge stimulated P release into the airways of asthmatic patients, and P levels positively correlated with proinflammatory cytokines in human bronchoalveolar lavage (BAL). High levels of P were also detected in the BAL of OVA-sensitized and challenged but not naive mice. Compared with wild-type (WT) mice, P-deficient (P(-/-)) mice had markedly reduced total and eosinophil cell counts in BAL and significantly attenuated airway hyperresponsiveness to methacholine. Ab blocking of P at both sensitization and challenge phases or at challenge phase alone, but not at sensitization phase alone, reduced airway inflammation. Conversely, intranasal reconstitution of P to P(-/-) mice at the challenge phase restored airway inflammation to wild-type levels. Notably, C3a levels in the BAL of OVA-challenged P(-/-) mice were significantly lower than in wild-type mice, and intranasal coadministration of an anti-C3a mAb with P to P(-/-) mice prevented restoration of airway inflammation. These results show that P plays a key role in allergen-induced airway inflammation and represents a potential therapeutic target for human asthma.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Blerina Ducka-Kokalari
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Imre G Redai
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Sayaka Sato
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Angela Haczku
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
31
|
Swe PM, Reynolds SL, Fischer K. Parasitic scabies mites and associated bacteria joining forces against host complement defence. Parasite Immunol 2015; 36:585-93. [PMID: 25081184 DOI: 10.1111/pim.12133] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/25/2014] [Indexed: 02/06/2023]
Abstract
Scabies is a ubiquitous and contagious skin disease caused by the parasitic mite Sarcoptes scabiei Epidemiological studies have identified scabies as a causative agent for secondary skin infections caused by Staphylococcus aureus and Streptococcus pyogenes. This is an important notion, as such bacterial infections can lead to serious downstream life-threatening complications. As the complement system is the first line of host defence that confronts invading pathogens, both the mite and bacteria produce a large array of molecules that inhibit the complement cascades. It is hypothesised that scabies mite complement inhibitors may play an important role in providing a favourable micro-environment for the establishment of secondary bacterial infections. This review aims to bring together the current literature on complement inhibition by scabies mites and bacteria associated with scabies and to discuss the proposed molecular link between scabies and bacterial co-infections.
Collapse
Affiliation(s)
- P M Swe
- Biology Department, QIMR Berghofer Medical Research Institute, Infectious Diseases Program, Brisbane, Qld, Australia
| | | | | |
Collapse
|
32
|
Berends ETM, Kuipers A, Ravesloot MM, Urbanus RT, Rooijakkers SHM. Bacteria under stress by complement and coagulation. FEMS Microbiol Rev 2014; 38:1146-71. [PMID: 25065463 DOI: 10.1111/1574-6976.12080] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 02/03/2023] Open
Abstract
The complement and coagulation systems are two related protein cascades in plasma that serve important roles in host defense and hemostasis, respectively. Complement activation on bacteria supports cellular immune responses and leads to direct killing of bacteria via assembly of the Membrane Attack Complex (MAC). Recent studies have indicated that the coagulation system also contributes to mammalian innate defense since coagulation factors can entrap bacteria inside clots and generate small antibacterial peptides. In this review, we will provide detailed insights into the molecular interplay between these protein cascades and bacteria. We take a closer look at how these pathways are activated on bacterial surfaces and discuss the mechanisms by which they directly cause stress to bacterial cells. The poorly understood mechanism for bacterial killing by the MAC will be reevaluated in light of recent structural insights. Finally, we highlight the strategies used by pathogenic bacteria to modulate these protein networks. Overall, these insights will contribute to a better understanding of the host defense roles of complement and coagulation against bacteria.
Collapse
Affiliation(s)
- Evelien T M Berends
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
A scabies mite serpin interferes with complement-mediated neutrophil functions and promotes staphylococcal growth. PLoS Negl Trop Dis 2014; 8:e2928. [PMID: 24945501 PMCID: PMC4063749 DOI: 10.1371/journal.pntd.0002928] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/21/2014] [Indexed: 01/30/2023] Open
Abstract
Background Scabies is a contagious skin disease caused by the parasitic mite Sarcoptes scabiei. The disease is highly prevalent worldwide and known to predispose to secondary bacterial infections, in particular by Streptococcus pyogenes and Staphylococcus aureus. Reports of scabies patients co-infected with methicillin resistant S. aureus (MRSA) pose a major concern for serious down-stream complications. We previously reported that a range of complement inhibitors secreted by the mites promoted the growth of S. pyogenes. Here, we show that a recently characterized mite serine protease inhibitor (SMSB4) inhibits the complement-mediated blood killing of S. aureus. Methodology/Principal Findings Blood killing of S. aureus was measured in whole blood bactericidal assays, counting viable bacteria recovered after treatment in fresh blood containing active complement and phagocytes, treated with recombinant SMSB4. SMSB4 inhibited the blood killing of various strains of S. aureus including methicillin-resistant and methicillin-sensitive isolates. Staphylococcal growth was promoted in a dose-dependent manner. We investigated the effect of SMSB4 on the complement-mediated neutrophil functions, namely phagocytosis, opsonization and anaphylatoxin release, by flow cytometry and in enzyme linked immuno sorbent assays (ELISA). SMSB4 reduced phagocytosis of S. aureus by neutrophils. It inhibited the deposition of C3b, C4b and properdin on the bacteria surface, but did not affect the depositions of C1q and MBL. SMSB4 also inhibited C5 cleavage as indicated by a reduced C5b-9 deposition. Conclusions/Significance We postulate that SMSB4 interferes with the activation of all three complement pathways by reducing the amount of C3 convertase formed. We conclude that SMSB4 interferes with the complement-dependent killing function of neutrophils, thereby reducing opsonization, phagocytosis and further recruitment of neutrophils to the site of infection. As a consequence secreted scabies mites complement inhibitors, such as SMSB4, provide favorable conditions for the onset of S. aureus co-infection in the scabies-infected microenvironment by suppressing the immediate host immune response. There is increasing evidence that in the tropics bacterial pyoderma and life-threatening downstream complications caused by Staphylococcus aureus and Streptococcus pyogenes are often linked with scabies infestation. The bacteria were commonly thought to find easy entry into the skin, damaged by scabies mites. However, there may be more to it. Our study aimed to identify the molecular mechanisms underlying the link between mites and bacteria. We recently characterized a molecule (SMSB4) that the mite produces for self-protection from the immediate host defense system in the skin. We show here that SMSB4 reduces the uptake of S. aureus by neutrophils, which are the killer immune cells, first to arrive at the infection site. These cells are guided by a number of complement factors. SMSB4 reduces the deposition of several complement components on the bacteria surface, thereby interrupting the essential defense cascades and inhibiting further neutrophil recruitment to the site. In summary, a mite molecule that likely originally evolved to protect the mite against the host defense plays a further role in the pathogenesis of secondary infections: when secreted into the damaged skin it promotes the onset of S. aureus infection thereby increasing the prevalence of debilitating disease associated with scabies.
Collapse
|
34
|
Dutow P, Fehlhaber B, Bode J, Laudeley R, Rheinheimer C, Glage S, Wetsel RA, Pabst O, Klos A. The complement C3a receptor is critical in defense against Chlamydia psittaci in mouse lung infection and required for antibody and optimal T cell response. J Infect Dis 2013; 209:1269-78. [PMID: 24273177 DOI: 10.1093/infdis/jit640] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The complement system protects against extracellular pathogens and links innate and adaptive immunity. In this study, we investigated the anaphylatoxin C3a receptor (C3aR) in Chlamydia psittaci lung infection and elucidated C3a-dependent adaptive immune mechanisms. METHODS Survival, body weight, and clinical score were monitored in primary mouse infection and after serum transfer. Bacterial load, histology, cellular distribution, cytokines, antibodies, and lymphocytes were analyzed. RESULTS C3aR(-/-) mice showed prolonged pneumonia with decreased survival, lower weight, and higher clinical score. Compared to wild-type mice bacterial clearance was impaired, and inflammatory parameters were increased. In lung-draining lymph nodes of C3aR(-/-) mice the total number of B cells, CD4(+) T cells, and Chlamydia-specific IFN-γ(+) (CD4(+) or CD8(+)) cells was reduced upon infection, and the mice were incapable of Chlamydia-specific immunoglobulin M or immunoglobulin G production. Performed before infection, transfer of hyperimmune serum prolonged survival of C3aR(-/-) mice. CONCLUSIONS C3a and its receptor are critical for defense against C. psittaci in mouse lung infection. In this model, C3a acts via its receptor as immune modulator. Enhancement of specific B and T cell responses upon infection with an intracellular bacterium were identified as hitherto unknown features of C3a/C3aR. These new functions might be of general immunological importance.
Collapse
Affiliation(s)
- Pavel Dutow
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH)
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Despite considerable advances in the understanding of the pathogenesis of meningococcal disease, this infection remains a major cause of morbidity and mortality globally. The role of the complement system in innate immune defenses against invasive meningococcal disease is well established. Individuals deficient in components of the alternative and terminal complement pathways are highly predisposed to invasive, often recurrent meningococcal infections. Genome-wide analysis studies also point to a central role for complement in disease pathogenesis. Here we review the pathophysiologic events pertinent to the complement system that accompany meningococcal sepsis in humans. Meningococci use several often redundant mechanisms to evade killing by human complement. Capsular polysaccharide and lipooligosaccharide glycan composition play critical roles in complement evasion. Some of the newly described protein vaccine antigens interact with complement components and have sparked considerable research interest.
Collapse
Affiliation(s)
- Lisa A Lewis
- Division of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester, MA USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester, MA USA
| |
Collapse
|
36
|
Lesher AM, Nilsson B, Song WC. Properdin in complement activation and tissue injury. Mol Immunol 2013; 56:191-8. [PMID: 23816404 DOI: 10.1016/j.molimm.2013.06.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 05/22/2013] [Indexed: 12/22/2022]
Abstract
The plasma protein properdin is the only known positive regulator of complement activation. Although regarded as an initiator of the alternative pathway of complement activation at the time of its discovery more than a half century ago, the role and mechanism of action of properdin in the complement cascade has undergone significant conceptual evolution since then. Despite the long history of research on properdin, however, new insight and unexpected findings on the role of properdin in complement activation, pathogen infection and host tissue injury are still being revealed by ongoing investigations. In this article, we provide a brief review on recent studies that shed new light on properdin biology, focusing on the following three topics: (1) its role as a pattern recognition molecule to direct and trigger complement activation, (2) its context-dependent requirement in complement activation on foreign and host cell surfaces, and (3) its involvement in alternative pathway complement-mediated immune disorders and considerations of properdin as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Allison M Lesher
- Department of Pharmacology and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
37
|
Saggu G, Cortes C, Emch HN, Ramirez G, Worth RG, Ferreira VP. Identification of a novel mode of complement activation on stimulated platelets mediated by properdin and C3(H2O). JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:6457-67. [PMID: 23677468 PMCID: PMC3784323 DOI: 10.4049/jimmunol.1300610] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Elevated numbers of activated platelets circulate in patients with chronic inflammatory diseases, including atherosclerosis and coronary disease. Activated platelets can activate the complement system. Although complement activation is essential for immune responses and removal of spent cells from circulation, it also contributes to inflammation and thrombosis, especially in patients with defective complement regulation. Proinflammatory activated leukocytes, which interact directly with platelets in response to vascular injury, are among the main sources of properdin, a positive regulator of the alternative pathway. The role of properdin in complement activation on stimulated platelets is unknown. Our data show that physiological forms of human properdin bind directly to human platelets after activation by strong agonists in the absence of C3, and bind nonproportionally to surface CD62P expression. Activation of the alternative pathway on activated platelets occurs when properdin is on the surface and recruits C3b or C3(H2O) to form C3b,Bb or a novel cell-bound C3 convertase [C3(H2O),Bb], which normally is present only in the fluid phase. Alternatively, properdin can be recruited by C3(H2O) on the platelet surface, promoting complement activation. Inhibition of factor H-mediated cell surface complement regulation significantly increases complement deposition on activated platelets with surface properdin. Finally, properdin released by activated neutrophils binds to activated platelets. Altogether, these data suggest novel molecular mechanisms for alternative pathway activation on stimulated platelets that may contribute to localization of inflammation at sites of vascular injury and thrombosis.
Collapse
Affiliation(s)
- Gurpanna Saggu
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
| | - Claudio Cortes
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
- Department of Molecular Sciences, Medical University of the Americas, Charlestown, Nevis, West Indies
| | - Heather N. Emch
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
| | - Galia Ramirez
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
- Department of Preventive Animal Medicine, Faculty of Veterinary Medicine, University of Chile, Santiago, Chile
| | - Randall G. Worth
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
| | - Viviana P. Ferreira
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH. 43614
| |
Collapse
|
38
|
Kouser L, Abdul-Aziz M, Nayak A, Stover CM, Sim RB, Kishore U. Properdin and factor h: opposing players on the alternative complement pathway "see-saw". Front Immunol 2013; 4:93. [PMID: 23630525 PMCID: PMC3632793 DOI: 10.3389/fimmu.2013.00093] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/05/2013] [Indexed: 12/16/2022] Open
Abstract
Properdin and factor H are two key regulatory proteins having opposite functions in the alternative complement pathway. Properdin up-regulates the alternative pathway by stabilizing the C3bBb complex, whereas factor H downregulates the pathway by promoting proteolytic degradation of C3b. While factor H is mainly produced in the liver, there are several extrahepatic sources. In addition to the liver, factor H is also synthesized in fetal tubuli, keratinocytes, skin fibroblasts, ocular tissue, adipose tissue, brain, lungs, heart, spleen, pancreas, kidney, muscle, and placenta. Neutrophils are the major source of properdin, and it is also produced by monocytes, T cells and bone marrow progenitor cell line. Properdin is released by neutrophils from intracellular stores following stimulation by N-formyl-methionine-leucine-phenylalanine (fMLP) and tumor necrosis factor alpha (TNF-α). The HEP G2 cells derived from human liver has been found to produce functional properdin. Endothelial cells also produce properdin when induced by shear stress, thus is a physiological source for plasma properdin. The diverse range of extrahepatic sites for synthesis of these two complement regulators suggests the importance and need for local availability of the proteins. Here, we discuss the significance of the local synthesis of properdin and factor H. This assumes greater importance in view of recently identified unexpected and novel roles of properdin and factor H that are potentially independent of their involvement in complement regulation.
Collapse
Affiliation(s)
- Lubna Kouser
- Centre for Infection, Immunity and Disease Mechanisms, Biosciences, School of Health Sciences and Social Care, Brunel University London, UK
| | | | | | | | | | | |
Collapse
|
39
|
Barratt-Due A, Pischke SE, Brekke OL, Thorgersen EB, Nielsen EW, Espevik T, Huber-Lang M, Mollnes TE. Bride and groom in systemic inflammation--the bells ring for complement and Toll in cooperation. Immunobiology 2013; 217:1047-56. [PMID: 22964230 DOI: 10.1016/j.imbio.2012.07.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 07/19/2012] [Accepted: 07/19/2012] [Indexed: 01/08/2023]
Abstract
Attenuating the sepsis-induced systemic inflammatory response, with subsequent homeostatic imbalance, has for years been one of the main tasks in sepsis related research. Complement and the TLR family constitute two important upstream sensor and effector-systems of innate immunity. Although they act as partly independent branches of pattern recognition, recent evidence indicate a considerable cross-talk implying that they can either compensate, synergize or antagonize each other. Combined inhibition of these pathways is therefore a particularly interesting approach with a profound anti-inflammatory potential. In previous preclinical studies, we demonstrated that targeting the key molecules C3 or C5 of complement and CD14 of the TLR family had a vast anti-inflammatory effect on Gram-negative bacteria-induced inflammation and sepsis. In this review, we elucidate the significance of these key molecules as important targets for intervention in sepsis and systemic inflammatory response syndrome. Finally, we argue that a combined inhibition of complement and CD14 represent a potential general treatment regimen, beyond the limit of sepsis, including non-infectious systemic inflammation and ischemia reperfusion injury.
Collapse
Affiliation(s)
- Andreas Barratt-Due
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Cortes C, Ohtola JA, Saggu G, Ferreira VP. Local release of properdin in the cellular microenvironment: role in pattern recognition and amplification of the alternative pathway of complement. Front Immunol 2013; 3:412. [PMID: 23335922 PMCID: PMC3547370 DOI: 10.3389/fimmu.2012.00412] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 12/18/2012] [Indexed: 12/24/2022] Open
Abstract
Properdin, the only positive regulatory protein of the complement system, acts as both a stabilizer of the alternative pathway (AP) convertases and as a selective pattern recognition molecule of certain microorganisms and host cells (i.e., apoptotic/necrotic cells) by serving as a platform for de novo C3b,Bb assembly. Properdin, a highly positively charged protein, normally exists as cyclic dimers (P(2)), trimers (P(3)), and tetramers (P(4)) of head-to-tail associations of monomeric 53 kDa subunits. While most complement proteins are produced mainly in the liver, properdin is synthesized primarily by various cell types, including neutrophils, monocytes, primary T cells, and shear-stressed endothelial cells resulting in properdin serum levels of 4-25 μg/ml. Multiple inflammatory agonists stimulate the release of properdin from stimulated leukocytes into the cellular microenvironment. Concentrated, focused increases in properdin levels may lead to stabilization and initiation of AP convertases, thus greatly amplifying the complement response to a local stimulus. This review highlights current knowledge related to these properties and discusses the implications of properdin production in a pro-inflammatory microenvironment.
Collapse
Affiliation(s)
- Claudio Cortes
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo Toledo, OH, USA ; Department Medical Immunology and Microbiology, Medical University of the Americas West Indies, Nevis
| | | | | | | |
Collapse
|
41
|
Bode J, Dutow P, Sommer K, Janik K, Glage S, Tümmler B, Munder A, Laudeley R, Sachse KW, Klos A. A new role of the complement system: C3 provides protection in a mouse model of lung infection with intracellular Chlamydia psittaci. PLoS One 2012. [PMID: 23189195 PMCID: PMC3506576 DOI: 10.1371/journal.pone.0050327] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The complement system modulates the intensity of innate and specific immunity. While it protects against infections by extracellular bacteria its role in infection with obligate intracellular bacteria, such as the avian and human pathogen Chlamydia (C.) psittaci, is still unknown. In the present study, knockout mice lacking C3 and thus all main complement effector functions were intranasally infected with C. psittaci strain DC15. Clinical parameters, lung histology, and cytokine levels were determined. A subset of infections was additionally performed with mice lacking C5 or C5a receptors. Complement activation occurred before symptoms of pneumonia appeared. Mice lacking C3 were ∼100 times more susceptible to the intracellular bacteria compared to wild-type mice, with all C3−/− mice succumbing to infection after day 9. At a low infective dose, C3−/− mice became severely ill after an even longer delay, the kinetics suggesting a so far unknown link of complement to the adaptive, protective immune response against chlamydiae. The lethal phenotype of C3−/− mice is not based on differences in the anti-chlamydial IgG response (which is slightly delayed) as demonstrated by serum transfer experiments. In addition, during the first week of infection, the absence of C3 was associated with partial protection characterized by reduced weight loss, better clinical score and lower bacterial burden, which might be explained by a different mechanism. Lack of complement functions downstream of C5 had little effect. This study demonstrates for the first time a strong and complex influence of complement effector functions, downstream of C3 and upstream of C5, on the outcome of an infection with intracellular bacteria, such as C. psittaci.
Collapse
Affiliation(s)
- Jenny Bode
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
| | - Pavel Dutow
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
| | - Kirsten Sommer
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
| | - Katrin Janik
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Hannover Medical School (MHH), Hannover, Germany
| | - Burkhard Tümmler
- Clinical Research Group, Pediatric Pulmonology and Neonatology, Hannover Medical School (MHH), Hannover, Germany
| | - Antje Munder
- Clinical Research Group, Pediatric Pulmonology and Neonatology, Hannover Medical School (MHH), Hannover, Germany
| | - Robert Laudeley
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
| | - Konrad W. Sachse
- Friedrich-Loeffler-Institut (Federal Research Institute for Animal Health), Jena, Germany
| | - Andreas Klos
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School (MHH), Hannover, Germany
- * E-mail:
| |
Collapse
|
42
|
Khandhadia S, Foster S, Cree A, Griffiths H, Osmond C, Goverdhan S, Lotery A. Chlamydia infection status, genotype, and age-related macular degeneration. Mol Vis 2012; 18:29-37. [PMID: 22259222 PMCID: PMC3258520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 01/05/2012] [Indexed: 11/05/2022] Open
Abstract
PURPOSE To evaluate whether Chlamydia (C.) infections are associated with age-related macular degeneration (AMD) and to assess if this association is influenced by the complement factor H (CFH) Y402H or the high temperature requirement A serine peptidase 1 (HTRA1) rs11200638 risk genotypes. METHODS One hundred ninety-nine AMD patients with early and late forms of the disease and 100 unaffected controls, at least 50 years old were included in the study. Patients in the AMD and control groups were selected based on known CFH Y402H variant genotype status (one third homozygous CC, one third heterozygous CT, and one third wild-type TT). Plasma from all patients and controls was tested for C. pneumoniae, C. trachomatis, and C. psittaci IgG seropositivity using a micro-immunofluorescent assay to establish previous infection status. Assays were conducted blind to risk genotypes and the results analyzed using univariate and multivariate (logistic regression) analysis. RESULTS IgG seropositivity to C. pneumoniae was most prevalent (69.2%, n=207), followed by C. trachomatis (7.4%, n=22) and C. psittaci (3.3%, n=10). No association was found between each of the three Chlamydia species IgG seropositivity and AMD status or severity (early/late). There was also no significant association between Chlamydia species IgG seropositivity and AMD status or severity, in patients carrying at least one CFH Y402H risk allele (C) or HTRA1 rs11200638 risk allele (A), with univariate or logistic regression analysis. CONCLUSIONS Chlamydia infection status does not appear to be associated with AMD status or severity. The presence of CFH Y402H and HTRA1 rs11200638 risk genotypes does not alter this negative association.
Collapse
Affiliation(s)
- Sam Khandhadia
- Division of Clinical Neurosciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | | | | | | | | | | | | |
Collapse
|
43
|
Carter C. Alzheimer's Disease: APP, Gamma Secretase, APOE, CLU, CR1, PICALM, ABCA7, BIN1, CD2AP, CD33, EPHA1, and MS4A2, and Their Relationships with Herpes Simplex, C. Pneumoniae, Other Suspect Pathogens, and the Immune System. Int J Alzheimers Dis 2011; 2011:501862. [PMID: 22254144 PMCID: PMC3255168 DOI: 10.4061/2011/501862] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 09/02/2011] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease susceptibility genes, APP and gamma-secretase, are involved in the herpes simplex life cycle, and that of other suspect pathogens (C. pneumoniae, H. pylori, C. neoformans, B. burgdorferri, P. gingivalis) or immune defence. Such pathogens promote beta-amyloid deposition and tau phosphorylation and may thus be causative agents, whose effects are conditioned by genes. The antimicrobial effects of beta-amyloid, the localisation of APP/gamma-secretase in immunocompetent dendritic cells, and gamma secretase cleavage of numerous pathogen receptors suggest that this network is concerned with pathogen disposal, effects which may be abrogated by the presence of beta-amyloid autoantibodies in the elderly. These autoantibodies, as well as those to nerve growth factor and tau, also observed in Alzheimer's disease, may well be antibodies to pathogens, due to homology between human autoantigens and pathogen proteins. NGF or tau antibodies promote beta-amyloid deposition, neurofibrillary tangles, or cholinergic neuronal loss, and, with other autoantibodies, such as anti-ATPase, are potential agents of destruction, whose formation is dictated by sequence homology between pathogen and human proteins, and thus by pathogen strain and human genes. Pathogen elimination in the ageing population and removal of culpable autoantibodies might reduce the incidence and offer hope for a cure in this affliction.
Collapse
Affiliation(s)
- Chris Carter
- PolygenicPathways, Flat 2, 40 Baldslow Road, Hastings, East Sussex TN34 2EY, UK
| |
Collapse
|