1
|
Thomsen ME, Bennike TB, Christiansen G, Emmersen J, Laursen NS, Zarantonello A, Andersen GR, Liu L, Dueholm MKD, Opstrup KV, Stensballe A, Birkelund S. Differences in complement activation of serum-resistant and serum-sensitive Klebsiella pneumoniae isolates. Mol Immunol 2025; 183:274-285. [PMID: 40424786 DOI: 10.1016/j.molimm.2025.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/27/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
The gram-negative bacteria Klebsiella pneumoniae are genetically heterogeneous and a common cause of sepsis and bacteremia in humans. The complement system is the first line of defence against bacteria when they invade the body. We previously investigated K. pneumoniae isolates from sepsis patients. We found that complement factor (C) 3 is deposited on all isolates independent of serum sensitivity, but the membrane attack complex (MAC) was only formed on the serum-sensitive isolates. To investigate the mechanism for serum resistance, we incubated one serum-sensitive and one serum-resistant isolate in human serum and identified bound complement factors by mass spectrometry. The serum-sensitive isolate had all expected complement factors bound, including C4, while the serum-resistant isolate had only C3 bound. The serum resistance was caused by a fast cleavage of C3b to iC3b. Thereby, the C5 convertase, and thus MAC, cannot be formed. To confirm the role of C4 in serum sensitivity, C4 was inhibited by the nanobody hC4Nb8, resulting in the survival of the serum-sensitive isolate. This suggests that C4 is indispensable for MAC formation through the classical and lectin pathways. In contrast, when activated selectively, the alternative pathway primarily leads to the generation of iC3b, thereby enabling serum resistance by bypassing MAC assembly.
Collapse
Affiliation(s)
- Mikkel Eggert Thomsen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| | - Gunna Christiansen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| | - Jeppe Emmersen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| | | | - Alessandra Zarantonello
- Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen DK-2200, Denmark.
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, Aarhus C 8000, Denmark.
| | - Lei Liu
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg Ø 9220, Denmark.
| | - Morten Kam Dahl Dueholm
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg Ø 9220, Denmark.
| | - Katharina V Opstrup
- Danish Veterinary and Food Administration, Stationsparken 31-33, Glostrup 2600, Denmark.
| | - Allan Stensballe
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| | - Svend Birkelund
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Selma Lagerløfs Vej 249, Gistrup 9260, Denmark.
| |
Collapse
|
2
|
Gadeberg TAF, Jørgensen MH, Olesen HG, Lorentzen J, Harwood SL, Almeida AV, Fruergaard MU, Jensen RK, Kanis P, Pedersen H, Tranchant E, Petersen SV, Thøgersen IB, Kragelund BB, Lyons JA, Enghild JJ, Andersen GR. Cryo-EM analysis of complement C3 reveals a reversible major opening of the macroglobulin ring. Nat Struct Mol Biol 2025; 32:884-895. [PMID: 39849196 DOI: 10.1038/s41594-024-01467-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/03/2024] [Indexed: 01/25/2025]
Abstract
The C3 protein is the central molecule within the complement system and undergoes proteolytic activation to C3b in the presence of pathogens. Pattern-independent activation of C3 also occurs via hydrolysis, resulting in C3(H2O), but the structural details of C3 hydrolysis remain elusive. Here we show that the conformation of the C3(H2O) analog, C3MA, is indistinguishable from C3b. In contrast, the reaction intermediate C3* adopts a conformation dramatically different from both C3 and C3MA. In C3*, unlocking of the macroglobulin (MG) 3 domain creates a large opening in the MG ring through which the anaphylatoxin (ANA) domain translocates through a transient opening. C3MA formation is inhibited by an MG3-specific nanobody and prevented by linking the ANA domain to the C3 β-chain. Our study reveals an unexpected dynamic behavior of C3 and forms the basis for elucidation of the in vivo contribution of C3 hydrolysis and for controlling complement upon intravascular hemolysis and surface-contact-induced activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Philipp Kanis
- Department of Molecular Biology and Genetics, Aarhus, Denmark
| | - Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus, Denmark
| | - Emil Tranchant
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Birthe Brandt Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Joseph Anthony Lyons
- Department of Molecular Biology and Genetics, Aarhus, Denmark
- Interdisciplinary Nanoscience Center (iNANO), Aarhus, Denmark
| | | | | |
Collapse
|
3
|
Butler AE, Moin ASM, Begam HH, Waris S, Azeez JM, Sathyapalan T, Atkin SL, Brennan E. Association of Complement Proteins with C Reactive Protein in Non-Obese Women with and Without Polycystic Ovary Syndrome. Int J Mol Sci 2025; 26:3008. [PMID: 40243681 PMCID: PMC11989124 DOI: 10.3390/ijms26073008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Complement proteins are increased in polycystic ovary syndrome (PCOS), as are markers of inflammation, such as the C-reactive protein (CRP); however, both may be increased in obesity. We hypothesised that body mass index (BMI)-driven CRP would comparably associate with an increase in complement proteins when obesity was accounted for in non-obese women with and without PCOS. In a non-obese, non-insulin-resistant population without inflammation (24 with PCOS and 24 control women), plasma CRP was measured by immunoassay. Slow Off-rate Modified Aptamer (SOMA)-scan plasma proteomic analysis of the classical, lectin, and alternative pathway complement proteins was undertaken. BMI, insulin resistance, and CRP did not differ (p < 0.05) between the cohorts. The alternative pathway of the complement protein system was overexpressed in PCOS (p < 0.05). CRP correlated positively (p < 0.05) with alternate pathway parameters in women with and without PCOS for C3a, iC3b, Factor B, Factor H, and Factor I; in PCOS alone for C3, C3adesArg, and C3d; and in women without PCOS, for properdin. CRP did not correlate with lectin pathway C2 or MBL (p > 0.05). CRP correlated positively (p < 0.05) with C4 of the classical pathways in women with PCOS alone. Hyperandrogenemia did not correlate with CRP or complement in non-obese PCOS. BMI correlated positively with C3, C3adesArg, C3a, iC3b, Factor B, Factor H, and properdin: classical pathway proteins; C1q, C4, C5 and C5a in PCOS women; BMI only correlated negatively with C1q in non-PCOS women. Upregulation of complement proteins occur in non-obese PCOS, and CRP is positively associated with complement protein changes in both women with and without PCOS. This indicates that BMI induces changes in CRP that lead to changes in the complement pathways, particularly the alternate pathway, with increases in CRP (though still within the reference laboratory normal range) leading to upregulation of complement proteins in PCOS. This suggests an enhanced set point for CRP-induced complement protein dysregulation in PCOS.
Collapse
Affiliation(s)
- Alexandra E. Butler
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Abu Saleh Md Moin
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Hamna H. Begam
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Sana Waris
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Juberiya M. Azeez
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, University of Hull, Hull HU6 7RU, UK;
| | - Stephen L. Atkin
- School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain; (A.E.B.); (A.S.M.M.); (H.H.B.); (S.W.); (J.M.A.); (S.L.A.)
| | - Edwina Brennan
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain
| |
Collapse
|
4
|
Duan H, Zhang Y, Otis MR, Drolet DW, Geisbrecht BV. The Inhibitory Effects of a Factor B-Binding DNA Aptamer Family Supersede the Gain of Function of Factor B Variants Associated with Atypical Hemolytic Uremic Syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1691-1702. [PMID: 39431879 PMCID: PMC11573645 DOI: 10.4049/jimmunol.2400420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
Aptamers are short, single-stranded oligonucleotides that selectively bind to target biomolecules. Although they generally exhibit good binding specificity, their affinities are often limited because of the relative lack of hydrophobic groups in nucleic acids. Chemically modified nucleotides incorporating hydrophobic structures into uracil have been synthesized to address this obstacle. Modified DNA aptamers containing such nonstandard nucleotides have been developed for >20 different complement proteins. These modified aptamers show increased affinity and enhanced serum stability and have potential value as therapeutic agents. We recently conducted a structure/function study on a family of modified DNA aptamers that bind specifically to complement Factor B (FB). This work revealed that these aptamers selectively inhibit the complement alternative pathway (AP) by preventing the formation of the AP complement component C3 (C3) proconvertase complex, C3bB. Certain patients with atypical hemolytic uremic syndrome express gain-of-function variants of FB that enhance the formation of the proconvertase complex and/or decrease the efficacy of endogenous regulators against the C3 convertases they form. To investigate whether these FB-binding aptamers could override the effects of disease-causing mutations in FB, we examined how they interacted with several FB variants, including D279G, F286L, K323E, and K350N, in various assays of complement function. We found that the inhibitory effect of the FB-binding aptamers superseded the gain-of-function mutations in FB, although the aptamers could not dissociate preformed C3 convertases. These findings suggest that FB-binding aptamers could be further developed as a potential treatment for certain atypical hemolytic uremic syndrome patients or those with other diseases characterized by excessive complement activity.
Collapse
Affiliation(s)
- Huiquan Duan
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | - Ying Zhang
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | | | | | - Brian V. Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| |
Collapse
|
5
|
Li L, Ding P, Dong Y, Shen S, Lv X, Yu J, Li L, Chen J, Wang P, Han B, Xu T, Hu W. CG001, a C3b-targeted complement inhibitor, blocks 3 complement pathways: development and preclinical evaluation. Blood Adv 2024; 8:4181-4193. [PMID: 38865712 PMCID: PMC11334799 DOI: 10.1182/bloodadvances.2024012874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024] Open
Abstract
ABSTRACT Excessively activated or dysregulated complement activation may contribute to the pathogenesis of a wide range of human diseases, thus leading to a surge in complement inhibitors. Herein, we developed a human-derived and antibody-like C3b-targeted fusion protein (CRIg-FH-Fc) x2, termed CG001, that could potently block all 3 complement pathways. Complement receptor of the immunoglobulin superfamily (CRIg) and factor H (FH) bind to distinct sites in C3b and synergistically inhibit complement activation. CRIg occupancy in C3b prevents the recruitment of C3 and C5 substrates, whereas FH occupancy in C3b accelerates the decay of C3/C5 convertases and promotes the factor I-mediated degradation and inactivation of C3b. CG001 also showed therapeutic effects in alternative pathways-induced hemolytic mouse and classical pathways-induced mesangial proliferative glomerulonephritis rat models. In the pharmacological/toxicological evaluation in rats and cynomolgus monkeys, CG001 displayed an antibody-like pharmacokinetic profile, a convincing complement inhibitory effect, and no observable toxic effects. Therefore, CG001 holds substantial potential for human clinical studies.
Collapse
Affiliation(s)
- Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | | | - Shupei Shen
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Xinyue Lv
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jie Yu
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Luying Li
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pilin Wang
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Ting Xu
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Reeve J. De-stabilizing innate immunity in COVID-19: effects of its own positive feedback and erratic viraemia on the alternative pathway of complement. ROYAL SOCIETY OPEN SCIENCE 2024; 11:221597. [PMID: 38234438 PMCID: PMC10791537 DOI: 10.1098/rsos.221597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Complement provides powerful, fast responses in the human circulation to SARS-CoV-2 (COVID-19 virus) infection of the lower respiratory tract. COVID-19 effects were investigated in a revised human in silico Mass Action model of complement's alternative pathway (AP) responses. Bursts of newly circulating virions increased the fission of Complement protein C3 into C3a and C3b via stimulation of the lectin pathway or inhibited complement factor H. Viral reproduction sub-models incorporated smoothly exponential or step-wise exponential growth. Starting complement protein concentrations were drawn randomly from published normal male or female ranges and each infection model run for 10 days. C3 and factor B (FB) syntheses driven by Lectin Pathway stimulation led to declining plasma C3 and increasing FB concentrations. The C3-convertase concentration, a driver of viral elimination, could match viral growth over three orders of magnitude but near-complete exhaustion of circulating C3 was more prevalent with step-wise than with 'smooth' increases in viral stimulation. C3 exhaustion could be prolonged. Type 2 Diabetes and hypertension led to greatly increased peak C3-convertase concentrations, as did short-term variability of COVID-19 viraemia, pulmonary capillary clotting and secondary acidosis. Positive feedback in the AP greatly extends its response range at the expense of stability.
Collapse
Affiliation(s)
- Jonathan Reeve
- Senior Research Fellow, Nuffield Department of Orthopaedics, Rheumatological and Musculoskeletal Sciences, University of Oxford Botnar Research Centre, Windmill Road, Oxford OX3 7LD, UK
| |
Collapse
|
7
|
Dani R, Oroszlán G, Martinusz R, Farkas B, Dobos B, Vadas E, Závodszky P, Gál P, Dobó J. Quantification of the zymogenicity and the substrate-induced activity enhancement of complement factor D. Front Immunol 2023; 14:1197023. [PMID: 37283768 PMCID: PMC10239819 DOI: 10.3389/fimmu.2023.1197023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Complement factor D (FD) is a serine protease present predominantly in the active form in circulation. It is synthesized as a zymogen (pro-FD), but it is continuously converted to FD by circulating active MASP-3. FD is a unique, self-inhibited protease. It has an extremely low activity toward free factor B (FB), while it is a highly efficient enzyme toward FB complexed with C3b (C3bB). The structural basis of this phenomenon is known; however, the rate enhancement was not yet quantified. It has also been unknown whether pro-FD has any enzymatic activity. In this study, we aimed to measure the activity of human FD and pro-FD toward uncomplexed FB and C3bB in order to quantitatively characterize the substrate-induced activity enhancement and zymogenicity of FD. Pro-FD was stabilized in the proenzyme form by replacing Arg25 (precursor numbering) with Gln (pro-FD-R/Q). Activated MASP-1 and MASP-3 catalytic fragments were also included in the study for comparison. We found that the complex formation with C3b enhanced the cleavage rate of FB by FD approximately 20 million-fold. C3bB was also a better substrate for MASP-1, approximately 100-fold, than free FB, showing that binding to C3b renders the scissile Arg-Lys bond in FB to become more accessible for proteolysis. Though easily measurable, this cleavage by MASP-1 is not relevant physiologically. Our approach provides quantitative data for the two-step mechanism characterized by the enhanced susceptibility of FB for cleavage upon complex formation with C3b and the substrate-induced activity enhancement of FD upon its binding to C3bB. Earlier MASP-3 was also implicated as a potential FB activator; however, MASP-3 does not cleave C3bB (or FB) at an appreciable rate. Finally, pro-FD cleaves C3bB at a rate that could be physiologically significant. The zymogenicity of FD is approximately 800, i.e., the cleavage rate of C3bB by pro-FD-R/Q was found to be approximately 800-fold lower than that by FD. Moreover, pro-FD-R/Q at approximately 50-fold of the physiological FD concentration could restore half-maximal AP activity of FD-depleted human serum on zymosan. The observed zymogen activity of pro-FD might be relevant in MASP-3 deficiency cases or during therapeutic MASP-3 inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
8
|
Turner NA, Moake JL. Heat-inactivated Factor B inhibits alternative pathway fluid-phase activation and convertase formation on endothelial cell-secreted ultra-large von Willebrand factor strings. Sci Rep 2023; 13:5764. [PMID: 37031266 PMCID: PMC10082794 DOI: 10.1038/s41598-023-33007-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023] Open
Abstract
Defective regulation of the alternative complement pathway (AP) causes excessive activation and promotes the inflammation and renal injury observed in atypical hemolytic-uremic syndrome (aHUS). The usefulness of heat-inactivated Factor B (HFB) in reducing AP activation was evaluated in: fluid-phase reactions, using purified complement proteins and Factor H (FH)-depleted serum; and in surface-activated reactions using human endothelial cells (ECs). C3a and Ba levels, measured by quantitative Western blots, determined the extent of fluid-phase activation. In reactions using C3, FB, and Factor D proteins, HFB addition (2.5-fold FB levels), reduced C3a levels by 60% and Ba levels by 45%. In reactions using FH-depleted serum (supplemented with FH at 12.5% normal levels), Ba levels were reduced by 40% with HFB added at 3.5-fold FB levels. The effectiveness of HFB in limiting AP convertase formation on activated surfaces was evaluated using stimulated ECs. Fluorescent microscopy was used to quantify endogenously released C3, FB, and C5 attached to EC-secreted ultra-large VWF strings. HFB addition reduced attachment of C3b by 2.7-fold, FB by 1.5-fold and C5 by fourfold. Our data indicate that HFB may be of therapeutic value in preventing AP-mediated generation of C3a and C5a, and the associated inflammation caused by an overactive AP.
Collapse
Affiliation(s)
- Nancy A Turner
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Joel L Moake
- Department of Bioengineering, Rice University, Houston, TX, USA
| |
Collapse
|
9
|
Pisarenka S, Meyer NC, Xiao X, Goodfellow R, Nester CM, Zhang Y, Smith RJH. Modeling C3 glomerulopathies: C3 convertase regulation on an extracellular matrix surface. Front Immunol 2023; 13:1073802. [PMID: 36846022 PMCID: PMC9947773 DOI: 10.3389/fimmu.2022.1073802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction C3 glomerulopathies (C3G) are ultra-rare complement-mediated diseases that lead to end-stage renal disease (ESRD) within 10 years of diagnosis in ~50% of patients. Overactivation of the alternative pathway (AP) of complement in the fluid phase and on the surface of the glomerular endothelial glycomatrix is the underlying cause of C3G. Although there are animal models for C3G that focus on genetic drivers of disease, in vivo studies of the impact of acquired drivers are not yet possible. Methods Here we present an in vitro model of AP activation and regulation on a glycomatrix surface. We use an extracellular matrix substitute (MaxGel) as a base upon which we reconstitute AP C3 convertase. We validated this method using properdin and Factor H (FH) and then assessed the effects of genetic and acquired drivers of C3G on C3 convertase. Results We show that C3 convertase readily forms on MaxGel and that this formation was positively regulated by properdin and negatively regulated by FH. Additionally, Factor B (FB) and FH mutants impaired complement regulation when compared to wild type counterparts. We also show the effects of C3 nephritic factors (C3Nefs) on convertase stability over time and provide evidence for a novel mechanism of C3Nef-mediated C3G pathogenesis. Discussion We conclude that this ECM-based model of C3G offers a replicable method by which to evaluate the variable activity of the complement system in C3G, thereby offering an improved understanding of the different factors driving this disease process.
Collapse
Affiliation(s)
- Sofiya Pisarenka
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
- Molecular Medicine Graduate Program, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Nicole C. Meyer
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Xue Xiao
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Renee Goodfellow
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Carla M. Nester
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Richard J. H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
- Molecular Medicine Graduate Program, Caver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
10
|
Schubart A, Flohr S, Junt T, Eder J. Low-molecular weight inhibitors of the alternative complement pathway. Immunol Rev 2023; 313:339-357. [PMID: 36217774 PMCID: PMC10092480 DOI: 10.1111/imr.13143] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dysregulation of the alternative complement pathway predisposes individuals to a number of diseases. It can either be evoked by genetic alterations in or by stabilizing antibodies to important pathway components and typically leads to severe diseases such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, C3 glomerulopathy, and age-related macular degeneration. In addition, the alternative pathway may also be involved in many other diseases where its amplifying function for all complement pathways might play a role. To identify specific alternative pathway inhibitors that qualify as therapeutics for these diseases, drug discovery efforts have focused on the two central proteases of the pathway, factor B and factor D. Although drug discovery has been challenging for a number of reasons, potent and selective low-molecular weight (LMW) oral inhibitors have now been discovered for both proteases and several molecules are in clinical development for multiple complement-mediated diseases. While the clinical development of these inhibitors initially focuses on diseases with systemic and/or peripheral tissue complement activation, the availability of LMW inhibitors may also open up the prospect of inhibiting complement in the central nervous system where its activation may also play an important role in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Schubart
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stefanie Flohr
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jörg Eder
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
11
|
Complement C3 Regulates Inflammatory Response and Monocyte/Macrophage Phagocytosis of Streptococcus agalactiae in a Teleost Fish. Int J Mol Sci 2022; 23:ijms232415586. [PMID: 36555227 PMCID: PMC9779060 DOI: 10.3390/ijms232415586] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 12/14/2022] Open
Abstract
The complement system is composed of a complex protein network and is pivotal to innate immunity. Complement 3 (C3) is a critical protein in the complement cascade and participates in complement activation and immune defense. In this study, C3 from Nile tilapia (Oreochromis niloticus) was cloned and its function in resisting pathogen infection was characterized. The full length of OnC3 open reading frame is 4974 bp, encoding 1657 aa, and the predicted protein mass weight is 185.93 kDa. The OnC3 amino acid sequence contains macroglobulin domains. The expression pattern of OnC3 mRNA in the tissues of healthy fish was detected, with the highest in the liver and the lowest in the muscle. After challenged with Streptococcus agalactiae and Aeromonas hydrophila, the expression of OnC3 mRNA was significantly up-regulated in the liver, spleen, and head kidney. Further, the recombinant OnC3 protein alleviated the inflammatory response and pathological damage of tissues after infected with S. agalactiae. Moreover, the OnC3 promoted the phagocytosis of monocytes/macrophages to S. agalactiae. The data obtained in this study provide a theoretical reference for in-depth understanding of C3 in host defense against bacterial infection and the immunomodulatory roles in teleost fish.
Collapse
|
12
|
Components of the Complement Cascade Differ in Polycystic Ovary Syndrome. Int J Mol Sci 2022; 23:ijms232012232. [PMID: 36293087 PMCID: PMC9603248 DOI: 10.3390/ijms232012232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Complement pathway proteins are reported to be increased in polycystic ovary syndrome (PCOS) and may be affected by obesity and insulin resistance. To investigate this, a proteomic analysis of the complement system was undertaken, including inhibitory proteins. In this cohort study, plasma was collected from 234 women (137 with PCOS and 97 controls). SOMALogic proteomic analysis was undertaken for the following complement system proteins: C1q, C1r, C2, C3, C3a, iC3b, C3b, C3d, C3adesArg, C4, C4a, C4b, C5, C5a, C5b-6 complex, C8, properdin, factor B, factor D, factor H, factor I, mannose-binding protein C (MBL), complement decay-accelerating factor (DAF) and complement factor H-related protein 5 (CFHR5). The alternative pathway of the complement system was primarily overexpressed in PCOS, with increased C3 (p < 0.05), properdin and factor B (p < 0.01). In addition, inhibition of this pathway was also seen in PCOS, with an increase in CFHR5, factor H and factor I (p < 0.01). Downstream complement factors iC3b and C3d, associated with an enhanced B cell response, and C5a, associated with an inflammatory cytokine release, were increased (p < 0.01). Hyperandrogenemia correlated positively with properdin and iC3b, whilst insulin resistance (HOMA-IR) correlated with iC3b and factor H (p < 0.05) in PCOS. BMI correlated positively with C3d, factor B, factor D, factor I, CFHR5 and C5a (p < 0.05). This comprehensive evaluation of the complement system in PCOS revealed the upregulation of components of the complement system, which appears to be offset by the concurrent upregulation of its inhibitors, with these changes accounted for in part by BMI, hyperandrogenemia and insulin resistance.
Collapse
|
13
|
Schäfer N, Grässel S. Involvement of complement peptides C3a and C5a in osteoarthritis pathology. Peptides 2022; 154:170815. [PMID: 35598724 DOI: 10.1016/j.peptides.2022.170815] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) affects more than 500 million people worldwide and is among the five diseases in Germany causing the highest suffering of the patients and cost for the society. The quality of life of OA patients is severely compromised, and adequate therapy is lacking owing to a knowledge gap that acts as a major barrier to finding safe and effective solutions. Chronic, low-grade inflammation plays a central role in OA pathogenesis and is associated with both OA pain and disease progression. Innate immune pathways, such as the complement- and pattern-recognition receptor pathways, are pivotal to the inflammation in OA and key components of the innate immune system implicated in OA include DAMP-TLR signaling, the complement system, carboxypeptidase B (CPB), and mononuclear cells. Anaphylatoxins C3a and C5a are small polypeptides (77 and 74 amino acids, respectively) which are released by proteolytic cleavage of the complement components C3 and C5. The alternative complement pathway seems to play a crucial role in OA pathogenesis as these complement components, mostly C3 and its activation peptide C3a, were detected at high levels in osteoarthritic cartilage, synovial membrane, and cultured chondrocytes. Targeting the complement system by using anti-complement drugs as a therapeutic option bears the risk of major side effects such as increasing the risk of infection, interfering with cell regeneration and metabolism, and suppressing the clearance of immune complexes. Despite those adverse effects, several synthetic complement peptide antagonists show promising effects in ameliorating inflammatory cell responses also in joint tissues.
Collapse
Affiliation(s)
- Nicole Schäfer
- Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany
| | - Susanne Grässel
- Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany; Department of Orthopaedic Surgery, University of Regensburg, Germany.
| |
Collapse
|
14
|
Bansal L, Nichols EM, Howsmon DP, Neisen J, Bessant CM, Cunningham F, Petit-Frere S, Ludbrook S, Damian V. Mathematical Modeling of Complement Pathway Dynamics for Target Validation and Selection of Drug Modalities for Complement Therapies. Front Pharmacol 2022; 13:855743. [PMID: 35517827 PMCID: PMC9061988 DOI: 10.3389/fphar.2022.855743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Motivation: The complement pathway plays a critical role in innate immune defense against infections. Dysregulation between activation and regulation of the complement pathway is widely known to contribute to several diseases. Nevertheless, very few drugs that target complement proteins have made it to the final regulatory approval because of factors such as high concentrations and dosing requirements for complement proteins and serious side effects from complement inhibition. Methods: A quantitative systems pharmacology (QSP) model of the complement pathway has been developed to evaluate potential drug targets to inhibit complement activation in autoimmune diseases. The model describes complement activation via the alternative and terminal pathways as well as the dynamics of several regulatory proteins. The QSP model has been used to evaluate the effect of inhibiting complement targets on reducing pathway activation caused by deficiency in factor H and CD59. The model also informed the feasibility of developing small-molecule or large-molecule antibody drugs by predicting the drug dosing and affinity requirements for potential complement targets. Results: Inhibition of several complement proteins was predicted to lead to a significant reduction in complement activation and cell lysis. The complement proteins that are present in very high concentrations or have high turnover rates (C3, factor B, factor D, and C6) were predicted to be challenging to engage with feasible doses of large-molecule antibody compounds (≤20 mg/kg). Alternatively, complement fragments that have a short half-life (C3b, C3bB, and C3bBb) were predicted to be challenging or infeasible to engage with small-molecule compounds because of high drug affinity requirements (>1 nM) for the inhibition of downstream processes. The drug affinity requirements for disease severity reduction were predicted to differ more than one to two orders of magnitude than affinities needed for the conventional 90% target engagement (TE) for several proteins. Thus, the QSP model analyses indicate the importance for accounting for TE requirements for achieving reduction in disease severity endpoints during the lead optimization stage.
Collapse
Affiliation(s)
- Loveleena Bansal
- Systems Modeling and Translational Biology, Computational Sciences, GSK, Upper Providence, Collegeville, PA, United States
| | | | - Daniel P Howsmon
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Jessica Neisen
- Immunology Research Unit, GSK, Stevenage, United Kingdom
| | | | | | | | - Steve Ludbrook
- Immunology Research Unit, GSK, Stevenage, United Kingdom
| | - Valeriu Damian
- Systems Modeling and Translational Biology, Computational Sciences, GSK, Upper Providence, Collegeville, PA, United States
| |
Collapse
|
15
|
De la O Becerra KI, Oosterheert W, van den Bos RM, Xenaki KT, Lorent JH, Ruyken M, Schouten A, Rooijakkers SHM, van Bergen En Henegouwen PMP, Gros P. Multifaceted Activities of Seven Nanobodies against Complement C4b. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2207-2219. [PMID: 35428691 PMCID: PMC9047069 DOI: 10.4049/jimmunol.2100647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 02/14/2022] [Indexed: 11/20/2022]
Abstract
Cleavage of the mammalian plasma protein C4 into C4b initiates opsonization, lysis, and clearance of microbes and damaged host cells by the classical and lectin pathways of the complement system. Dysregulated activation of C4 and other initial components of the classical pathway may cause or aggravate pathologies, such as systemic lupus erythematosus, Alzheimer disease, and schizophrenia. Modulating the activity of C4b by small-molecule or protein-based inhibitors may represent a promising therapeutic approach for preventing excessive inflammation and damage to host cells and tissue. Here, we present seven nanobodies, derived from llama (Lama glama) immunization, that bind to human C4b (Homo sapiens) with high affinities ranging from 3.2 nM to 14 pM. The activity of the nanobodies varies from no to complete inhibition of the classical pathway. The inhibiting nanobodies affect different steps in complement activation, in line with blocking sites for proconvertase formation, C3 substrate binding to the convertase, and regulator-mediated inactivation of C4b. For four nanobodies, we determined single-particle cryo-electron microscopy structures in complex with C4b at 3.4-4 Å resolution. The structures rationalize the observed functional effects of the nanobodies and define their mode of action during complement activation. Thus, we characterized seven anti-C4b nanobodies with diverse effects on the classical pathway of complement activation that may be explored for imaging, diagnostic, or therapeutic applications.
Collapse
Affiliation(s)
- Karla I De la O Becerra
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Wout Oosterheert
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Ramon M van den Bos
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Katerina T Xenaki
- Cell Biology, Neurobiology & Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Joseph H Lorent
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands; and
| | - Maartje Ruyken
- Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Arie Schouten
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | | | | | - Piet Gros
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands;
| |
Collapse
|
16
|
Complement component C3: A structural perspective and potential therapeutic implications. Semin Immunol 2022; 59:101627. [PMID: 35760703 PMCID: PMC9842190 DOI: 10.1016/j.smim.2022.101627] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/18/2023]
Abstract
As the most abundant component of the complement system, C3 and its proteolytic derivatives serve essential roles in the function of all three complement pathways. Central to this is a network of protein-protein interactions made possible by the sequential proteolysis and far-reaching structural changes that accompany C3 activation. Beginning with the crystal structures of C3, C3b, and C3c nearly twenty years ago, the physical transformations underlying C3 function that had long been suspected were finally revealed. In the years that followed, a compendium of crystallographic information on C3 derivatives bound to various enzymes, regulators, receptors, and inhibitors generated new levels of insight into the structure and function of the C3 molecule. This Review provides a concise classification, summary, and interpretation of the more than 50 unique crystal structure determinations for human C3. It also highlights other salient features of C3 structure that were made possible through solution-based methods, including Hydrogen/Deuterium Exchange and Small Angle X-ray Scattering. At this pivotal time when the first C3-targeted therapeutics begin to see use in the clinic, some perspectives are also offered on how this continually growing body of structural information might be leveraged for future development of next-generation C3 inhibitors.
Collapse
|
17
|
Michels MAHM, Maas RJF, van der Velden TJAM, van de Kar NCAJ, van den Heuvel LPWJ, Volokhina EB. The Role of Properdin in C5 Convertase Activity and C5b-9 Formation in the Complement Alternative Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2465-2472. [PMID: 34635587 DOI: 10.4049/jimmunol.2100238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022]
Abstract
The complement system is an important part of innate immunity. Complement activation leads to formation of convertase enzymes, switch of their specificity from C3 to C5 cleavage, and generation of lytic membrane attack complexes (C5b-9) on surfaces of pathogens. Most C5 cleavage occurs via the complement alternative pathway (AP). The regulator properdin promotes generation and stabilization of AP convertases. However, its role in C5 activation is not yet understood. In this work, we showed that serum properdin is essential for LPS- and zymosan-induced C5b-9 generation and C5b-9-mediated lysis of rabbit erythrocytes. Furthermore, we demonstrated its essential role in C5 cleavage by AP convertases. To this end, we developed a hemolytic assay in which AP convertases were generated on rabbit erythrocytes by using properdin-depleted serum in the presence of C5 inhibitor (step 1), followed by washing and addition of purified C5-C9 components to allow C5b-9 formation (step 2). In this assay, addition of purified properdin to properdin-depleted serum during convertase formation (step 1) was required to restore C5 cleavage and C5b-9-mediated hemolysis. Importantly, C5 convertase activity was also fully restored when properdin was added together with C5b-9 components (step 2), thus after convertase formation. Moreover, with C3-depleted serum, not capable of forming new convertases but containing properdin, in step 2 of the assay, again full C5b-9 formation was observed and blocked by addition of properdin inhibitor Salp20. Thus, properdin is essential for the convertase specificity switch toward C5, and this function is independent of properdin's role in new convertase formation.
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, Nijmegen, the Netherlands;
| | - Rianne J F Maas
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Thea J A M van der Velden
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, 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, 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, Nijmegen, the Netherlands.,Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Pediatrics/Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium; and.,Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Elena B Volokhina
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | |
Collapse
|
18
|
Functional Identification of Complement Factor D and Analysis of Its Expression during GCRV Infection in Grass Carp ( Ctenopharyngodon idella). Int J Mol Sci 2021; 22:ijms222112011. [PMID: 34769442 PMCID: PMC8584590 DOI: 10.3390/ijms222112011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Complement factor D (Df) is a serine protease well known for activating the alternative pathway (AP) in mammals by promoting the cleavage of complement component 3 (C3), thus becoming involved in innate defense. In teleost fish, however, the functional mechanisms of Df in the AP and against pathogen infection are far from clear. In the present study, we cloned and characterized the Df gene, CiDf, from grass carp (Ctenopharyngodon idella) and analyzed its function in promoting C3 cleavage and expression changes after grass carp reovirus (GCRV) infection. The open reading frame of CiDf was found to be 753 bp, encoding 250 amino acids with a molecular mass of 27.06 kDa. CiDf harbors a conserved Tryp_SPc domain, with three conserved residues representing the catalytic triad and three conserved binding sites in the substrate specificity pocket. Pairwise alignment showed that CiDf shares the highest identity (96%) and similarity (98%) with Df from Anabarilius grahami. Phylogenetic analysis indicated that CiDf and other fish Dfs formed a distinct evolutionary branch. Similar to most Dfs from other vertebrates, the CiDf gene structure is characterized by four introns and five exons. The incubation of recombinant CiDf protein with grass carp serum significantly increased the C3b content, demonstrating the conserved function of CiDf in the AP in promoting C3 cleavage, similar to Dfs in mammals. CiDf mRNA expression was widely detected in various tissues and levels were relatively higher in the liver, spleen, and intestine of grass carp. During GCRV infection over a 168-hour period, a high level of CiDf mRNA expression in the liver, spleen, and intestine was maintained at 144 and 168 h, suggesting AP activity at the late stage of GCRV infection. Collectively, the above results reveal the conserved structure and function of CiDf and its distinct expression patterns after GCRV infection, which provide a key basis for studying the roles of Df and AP during GCRV infection in the grass carp C. idella.
Collapse
|
19
|
Kim BJ, Mastellos DC, Li Y, Dunaief JL, Lambris JD. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions. Prog Retin Eye Res 2021; 83:100936. [PMID: 33321207 PMCID: PMC8197769 DOI: 10.1016/j.preteyeres.2020.100936] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration (AMD) remains a major cause of legal blindness, and treatment for the geographic atrophy form of AMD is a significant unmet need. Dysregulation of the complement cascade is thought to be instrumental for AMD pathophysiology. In particular, C3 and C5 are pivotal components of the complement cascade and have become leading therapeutic targets for AMD. In this article, we discuss C3 and C5 in detail, including their roles in AMD, biochemical and structural aspects, locations of expression, and the functions of C3 and C5 fragments. Further, the article critically reviews developing therapeutics aimed at C3 and C5, underscoring the potential effects of broad inhibition of complement at the level of C3 versus more specific inhibition at C5. The relationships of complement biology to the inflammasome and microglia/macrophage activity are highlighted. Concepts of C3 and C5 biology will be emphasized, while we point out questions that need to be settled and directions for future investigations.
Collapse
Affiliation(s)
- Benjamin J Kim
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Yafeng Li
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Lambris
- Department of Laboratory Medicine and Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
20
|
Garland DL, Pierce EA, Fernandez-Godino R. Complement C5 is not critical for the formation of sub-RPE deposits in Efemp1 mutant mice. Sci Rep 2021; 11:10416. [PMID: 34001980 PMCID: PMC8128922 DOI: 10.1038/s41598-021-89978-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
The complement system plays a role in the formation of sub-retinal pigment epithelial (RPE) deposits in early stages of age-related macular degeneration (AMD). But the specific mechanisms that connect complement activation and deposit formation in AMD patients are unknown, which limits the development of efficient therapies to reduce or stop disease progression. We have previously demonstrated that C3 blockage prevents the formation of sub-RPE deposits in a mouse model of EFEMP1-associated macular degeneration. In this study, we have used double mutant Efemp1R345W/R345W:C5-/- mice to investigate the role of C5 in the formation of sub-RPE deposits in vivo and in vitro. The data revealed that the genetic ablation of C5 does not eliminate the formation of sub-RPE deposits. Contrarily, the absence of C5 in RPE cultures promotes complement dysregulation that results in increased activation of C3, which likely contributes to deposit formation even in the absence of EFEMP1-R345W mutant protein. The results also suggest that genetic ablation of C5 alters the extracellular matrix turnover through an effect on matrix metalloproteinases in RPE cell cultures. These results confirm that C3 rather than C5 could be an effective therapeutic target to treat early AMD.
Collapse
Affiliation(s)
- Donita L Garland
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, USA
| | - Eric A Pierce
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, USA
| | | |
Collapse
|
21
|
Willrich MAV, Braun KMP, Moyer AM, Jeffrey DH, Frazer-Abel A. Complement testing in the clinical laboratory. Crit Rev Clin Lab Sci 2021; 58:447-478. [PMID: 33962553 DOI: 10.1080/10408363.2021.1907297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The complement system is the human's first line of defense against microbial pathogens because of its important housekeeping and infection/inflammation roles. It is composed of a series of soluble and cell-bound proteins that are activated in a cascade effect, similar to the coagulation pathways. There are different pattern recognizing molecules that activate the complement system in response to stimuli or threats, acting through three initiation pathways: classical, lectin, and alternative. All three activation pathways converge at the C3 component and share the terminal pathway. The main outputs of the complement system action are lytic killing of microbes, the release of pro-inflammatory anaphylatoxins, and opsonization of targets. Laboratory testing is relevant in the setting of suspected complement deficiencies, as well as in the emerging number of diseases related to dysregulation (over-activation) of complement. Most common assays measure complement lytic activity and the different complement component concentrations. Specialized testing includes the evaluation of autoantibodies against complement components, activation fragments, and genetic studies. In this review, we cover laboratory testing for complement and the conditions with complement involvement, as well as current challenges in the field.
Collapse
Affiliation(s)
| | - Karin M P Braun
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - David H Jeffrey
- Exsera Biolabs, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ashley Frazer-Abel
- Exsera Biolabs, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
22
|
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
|
23
|
Zarantonello A, Pedersen H, Laursen NS, Andersen GR. Nanobodies Provide Insight into the Molecular Mechanisms of the Complement Cascade and Offer New Therapeutic Strategies. Biomolecules 2021; 11:biom11020298. [PMID: 33671302 PMCID: PMC7922070 DOI: 10.3390/biom11020298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 01/22/2023] Open
Abstract
The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer’s disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.
Collapse
Affiliation(s)
- Alessandra Zarantonello
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
| | - Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
| | - Nick S. Laursen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Gregers R. Andersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
- Correspondence: ; Tel.: +45-30256646
| |
Collapse
|
24
|
Granata F, Duca L, Brancaleoni V, Fustinoni S, De Luca G, Motta I, Graziadei G, Di Pierro E. Alternative Pathway Involvement in Protoporphyria Patients Related to Sun Exposure. Front Immunol 2021; 12:615620. [PMID: 33664746 PMCID: PMC7921788 DOI: 10.3389/fimmu.2021.615620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/25/2021] [Indexed: 11/17/2022] Open
Abstract
The homeostasis of tissues in a chronic disease is an essential function of the alternative pathway (AP) of the complement system (CS). However, if not controlled, it may also be detrimental to healthy cells with a consequent aggravation of symptoms. The protoporphyria (PP) is a rare chronic disease that causes phototoxicity in visible light with local skin pain and general malaise. In order to establish if there is a systemic involvement of the CS during sun exposure, we designed a non-invasive method with a serum collection in winter and summer from 19 PP and 13 controls to detect the levels of CS protein: Properdin, Factor H (FH), and C5. Moreover, the global radiation data were collected from the regional agency of environmental protection (ARPA). The results show growing values for every protein in patients with PP, compared to control, in both seasons, in particular in summer compared to winter. To reinforce the evidence, we have estimated the personal exposure of patients based on the global radiation data. The main factors of the AP increased over the season, confirming the involvement of the AP in relation to light exposure. The systemic response could justify the general malaise of patients after long light exposure and can be exploited to elucidate new therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Granata
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| | - Lorena Duca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| | - Valentina Brancaleoni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| | - Silvia Fustinoni
- EPIGET - Epidemiology, Epigenetics, and Toxicology Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Environmental and Industrial Toxicology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo De Luca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| | - Irene Motta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy.,Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giovanna Graziadei
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| | - Elena Di Pierro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, U.O.C. Medicina Generale, Milan, Italy
| |
Collapse
|
25
|
Jagatia H, Tsolaki AG. The Role of Complement System and the Immune Response to Tuberculosis Infection. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:84. [PMID: 33498555 PMCID: PMC7909539 DOI: 10.3390/medicina57020084] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/30/2022]
Abstract
The complement system orchestrates a multi-faceted immune response to the invading pathogen, Mycobacterium tuberculosis. Macrophages engulf the mycobacterial bacilli through bacterial cell surface proteins or secrete proteins, which activate the complement pathway. The classical pathway is activated by C1q, which binds to antibody antigen complexes. While the alternative pathway is constitutively active and regulated by properdin, the direct interaction of properdin is capable of complement activation. The lectin-binding pathway is activated in response to bacterial cell surface carbohydrates such as mannose, fucose, and N-acetyl-d-glucosamine. All three pathways contribute to mounting an immune response for the clearance of mycobacteria. However, the bacilli can reside, persist, and evade clearance by the immune system once inside the macrophages using a number of mechanisms. The immune system can compartmentalise the infection into a granulomatous structure, which contains heterogenous sub-populations of M. tuberculosis. The granuloma consists of many types of immune cells, which aim to clear and contain the infection whilst sacrificing the affected host tissue. The full extent of the involvement of the complement system during infection with M. tuberculosis is not fully understood. Therefore, we reviewed the available literature on M. tuberculosis and other mycobacterial literature to understand the contribution of the complement system during infection.
Collapse
Affiliation(s)
- Heena Jagatia
- Department for Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Anthony G. Tsolaki
- Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, Uxbridge UB8 3PN, UK;
| |
Collapse
|
26
|
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
|
27
|
Caruso A, Vollmer J, Machacek M, Kortvely E. Modeling the activation of the alternative complement pathway and its effects on hemolysis in health and disease. PLoS Comput Biol 2020; 16:e1008139. [PMID: 33006965 PMCID: PMC7531836 DOI: 10.1371/journal.pcbi.1008139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
The complement system is a powerful mechanism of innate immunity poised to eliminate foreign cells and pathogens. It is an intricate network of >35 proteins, which, once activated, leads to the tagging of the surface to be eliminated, produces potent chemoattractants to recruit immune cells, and inserts cytotoxic pores into nearby lipid surfaces. Although it can be triggered via different pathways, its net output is largely based on the direct or indirect activation of the alternative pathway. Complement dysregulation or deficiencies may cause severe pathologies, such as paroxysmal nocturnal hemoglobinuria (PNH), where a lack of complement control proteins leads to hemolysis and life-threatening anemia. The complexity of the system poses a challenge for the interpretation of experimental data and the design of effective pharmacological therapies. To address this issue, we developed a mathematical model of the alternative complement pathway building on previous modelling efforts. The model links complement activation to the hemolytic activity of the terminal alternative pathway, providing an accurate description of pathway activity as observed in vitro and in vivo, in health and disease. Through adjustment of the parameters describing experimental conditions, the model was capable of reproducing the results of an array of standard assays used in complement research. To demonstrate its clinical applicability, we compared model predictions with clinical observations of the recovery of hematological biomarkers in PNH patients treated with the complement inhibiting anti-C5 antibody eculizumab. In conclusion, the model can enhance the understanding of complement biology and its role in disease pathogenesis, help identifying promising targets for pharmacological intervention, and predict the outcome of complement-targeting pharmacological interventions.
Collapse
Affiliation(s)
- Antonello Caruso
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Elod Kortvely
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
28
|
Tille A, Lehnert T, Zipfel PF, Figge MT. Quantification of Factor H Mediated Self vs. Non-self Discrimination by Mathematical Modeling. Front Immunol 2020; 11:1911. [PMID: 33013842 PMCID: PMC7493836 DOI: 10.3389/fimmu.2020.01911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/16/2020] [Indexed: 11/13/2022] Open
Abstract
The complement system is part of the innate immune system and plays an important role in the host defense against infectious pathogens. One of the main effects is the opsonization of foreign invaders and subsequent uptake by phagocytosis. Due to the continuous default basal level of active complement molecules, a tight regulation is required to protect the body's own cells (self cells) from opsonization and from complement damage. A major complement regulator is Factor H, which is recruited from the fluid phase and attaches to cell surfaces where it effectively controls complement activation. Besides self cells, pathogens also have the ability to bind Factor H; they can thus escape opsonization and phagocytosis causing severe infections. In order to advance our understanding of the opsonization process at a quantitative level, we developed a mathematical model for the dynamics of the complement system-termed DynaCoSys model-that is based on ordinary differential equations for cell surface-bound molecules and on partial differential equations for concentration profiles of the fluid phase molecules in the environment of cells. This hybrid differential equation approach allows to model the complement cascade focusing on the role of active C3b in the fluid phase and on the cell surface as well as on its inactivation on the cell surface. The DynaCoSys model enables us to quantitatively predict the conditions under which Factor H mediated complement evasion occurs. Furthermore, investigating the quantitative impact of model parameters by a sensitivity analysis, we identify the driving processes of complement activation and regulation in both the self and non-self regime. The two regimes are defined by a critical Factor H concentration on the cell surface and we use the model to investigate the differential impact of complement model parameters on this threshold value. The dynamic modeling on the surface of pathogens are further relevant to understand pathophysiological situations where Factor H mutants and defective Factor H binding to target surfaces results in pathophysiology such as renal and retinal disease. In the future, this DynaCoSys model will be extended to also enable evaluating treatment strategies of complement-related diseases.
Collapse
Affiliation(s)
- Alexander Tille
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany.,Faculty of Biological Sciences, Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Teresa Lehnert
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
| | - Peter F Zipfel
- Faculty of Biological Sciences, Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany.,Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany.,Faculty of Biological Sciences, Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
29
|
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
|
30
|
Hew BE, Pangburn MK, Vogel CW, Fritzinger DC. Identification of intermolecular bonds between human factor B and Cobra Venom Factor important for C3 convertase stability. Toxicon 2020; 184:68-77. [PMID: 32526239 DOI: 10.1016/j.toxicon.2020.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 10/24/2022]
Abstract
Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF, Bb is an enzyme that cleaves C3 and C5. However, CVF, Bb exhibits significantly different functional properties from C3b,Bb. Whereas both, CVF, Bb and C3b, Bb exhibit spontaneous decay-dissociation into the respective subunits, thereby eliminating the enzymatic activity, the CVF, Bb convertase is physico-chemically far more stable, decaying with a half-life that is more than two orders of magnitude slower than that of C3b,Bb. In addition, CVF, Bb is completely resistant to inactivation by Factors H and I. These two properties of CVF, Bb allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the physico-chemical stability of CVF,Bb, we have created recombinant hybrid proteins of CVF and human C3, based on structural differences between CVF and human C3b in the C-terminal C345C domain. Here we describe three human C3/CVF hybrid proteins which differ in only one, two, or five amino acid residues from earlier described hybrid proteins. In all three cases, the hybrid proteins containing CVF residues form more stable convertases, and exhibit stronger complement-depletion activity than hybrid proteins with human C3 residues. Three bonds between CVF residues and Factor Bb residues could be identified by crystallographic modeling that contribute to the greater stability of the convertases.
Collapse
Affiliation(s)
- Brian E Hew
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Michael K Pangburn
- Biomedical Research Department, University of Texas Health Science Center, Tyler, TX, 75708, USA
| | - Carl-Wilhelm Vogel
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA; Department of Pathology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA.
| | - David C Fritzinger
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| |
Collapse
|
31
|
Fromell K, Adler A, Åman A, Manivel VA, Huang S, Dührkop C, Sandholm K, Ekdahl KN, Nilsson B. Assessment of the Role of C3(H 2O) in the Alternative Pathway. Front Immunol 2020; 11:530. [PMID: 32296436 PMCID: PMC7136553 DOI: 10.3389/fimmu.2020.00530] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
In this study we investigate the hydrolysis of C3 to C3(H2O) and its ability to initiate activation via the alternative pathway (AP) of the complement system. The internal thioester bond within C3 is hydrolyzed by water in plasma because of its inherent lability. This results in the formation of non-proteolytically activated C3(H2O) which is believed have C3b-like properties and be able to form an active initial fluid phase C3 convertase together with Factor B (FB). The generation of C3(H2O) occurs at a low but constant rate in blood, but the formation can be greatly accelerated by the interaction with various surfaces or nucleophilic and chaotropic agents. In order to more specifically elucidate the relevance of the C3(H2O) for AP activation, formation was induced in solution by repeated freeze/thawing, methylamine or KCSN treatment and named C3(x) where the x can be any of the reactive nucleophilic or chaotropic agents. Isolation and characterization of C3(x) showed that it exists in several forms with varying attributes, where some have more C3b-like properties and can be cleaved by Factor I in the presence of Factor H. However, in common for all these variants is that they are less active partners in initial formation of the AP convertase compared with the corresponding activity of C3b. These observations support the idea that formation of C3(x) in the fluid phase is not a strong initiator of the AP. It is rather likely that the AP mainly acts as an amplification mechanism of complement activation that is triggered by deposition of target-bound C3b molecules generated by other means.
Collapse
Affiliation(s)
- Karin Fromell
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Anna Adler
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Amanda Åman
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Vivek Anand Manivel
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Shan Huang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claudia Dührkop
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Kerstin Sandholm
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Kristina N Ekdahl
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden.,Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| |
Collapse
|
32
|
Mathematical Modelling of Alternative Pathway of Complement System. Bull Math Biol 2020; 82:33. [PMID: 32062771 PMCID: PMC7024062 DOI: 10.1007/s11538-020-00708-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 02/03/2020] [Indexed: 11/02/2022]
Abstract
The complement system (CS) is an integral part of innate immunity and can be activated via three different pathways. The alternative pathway (AP) has a central role in the function of the CS. The AP of complement system is implicated in several human disease pathologies. In the absence of triggers, the AP exists in a time-invariant resting state (physiological steady state). It is capable of rapid, potent and transient activation response upon challenge with a trigger. Previous models of AP have focused on the activation response. In order to understand the molecular machinery necessary for AP activation and regulation of a physiological steady state, we built parsimonious AP models using experimentally supported kinetic parameters. The models further allowed us to test quantitative roles played by negative and positive regulators of the pathway in order to test hypotheses regarding their mechanisms of action, thus providing more insight into the complex regulation of AP.
Collapse
|
33
|
Makou E, Bailey RG, Johnston H, Parkin JD, Hulme AN, Hähner G, Barlow PN. Combining SPR with atomic-force microscopy enables single-molecule insights into activation and suppression of the complement cascade. J Biol Chem 2019; 294:20148-20163. [PMID: 31719147 PMCID: PMC6937562 DOI: 10.1074/jbc.ra119.010913] [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: 09/01/2019] [Revised: 11/07/2019] [Indexed: 12/05/2022] Open
Abstract
Activation and suppression of the complement system compete on every serum-exposed surface, host or foreign. Potentially harmful outcomes of this competition depend on surface molecules through mechanisms that remain incompletely understood. Combining surface plasmon resonance (SPR) with atomic force microscopy (AFM), here we studied two complement system proteins at the single-molecule level: C3b, the proteolytically activated form of C3, and factor H (FH), the surface-sensing C3b-binding complement regulator. We used SPR to monitor complement initiation occurring through a positive-feedback loop wherein surface-deposited C3b participates in convertases that cleave C3, thereby depositing more C3b. Over multiple cycles of flowing factor B, factor D, and C3 over the SPR chip, we amplified C3b from ∼20 to ∼220 molecules·μm−2. AFM revealed C3b clusters of up to 20 molecules and solitary C3b molecules deposited up to 200 nm away from the clusters. A force of 0.17 ± 0.02 nanonewtons was needed to pull a single FH molecule, anchored to the AFM probe, from its complex with surface-attached C3b. The extent to which FH molecules stretched before detachment varied widely among complexes. Performing force-distance measurements with FH(D1119G), a variant lacking one of the C3b-binding sites and causing atypical hemolytic uremic syndrome, we found that it detached more uniformly and easily. In further SPR experiments, KD values between FH and C3b on a custom-made chip surface were 5-fold tighter than on commercial chips and similar to those on erythrocytes. These results suggest that the chemistry at the surface on which FH acts drives conformational adjustments that are functionally critical.
Collapse
Affiliation(s)
- Elisavet Makou
- EaStChem School of Chemistry, University of Edinburgh, Joseph Black Chemistry Building, Edinburgh, Scotland EH9 3FJ, United Kingdom
| | - Richard G Bailey
- EaStChem School of Chemistry, University of St. Andrews, St Andrews, Scotland KY16 9ST, United Kingdom
| | - Heather Johnston
- EaStChem School of Chemistry, University of Edinburgh, Joseph Black Chemistry Building, Edinburgh, Scotland EH9 3FJ, United Kingdom
| | - John D Parkin
- EaStChem School of Chemistry, University of St. Andrews, St Andrews, Scotland KY16 9ST, United Kingdom
| | - Alison N Hulme
- EaStChem School of Chemistry, University of Edinburgh, Joseph Black Chemistry Building, Edinburgh, Scotland EH9 3FJ, United Kingdom
| | - Georg Hähner
- EaStChem School of Chemistry, University of St. Andrews, St Andrews, Scotland KY16 9ST, United Kingdom
| | - Paul N Barlow
- EaStChem School of Chemistry, University of Edinburgh, Joseph Black Chemistry Building, Edinburgh, Scotland EH9 3FJ, United Kingdom .,School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland EH9 3JY, United Kingdom
| |
Collapse
|
34
|
Gupta P, Tripathy AS. Alternative pathway of complement activation has a beneficial role against Chandipura virus infection. Med Microbiol Immunol 2019; 209:109-124. [PMID: 31781935 PMCID: PMC7223837 DOI: 10.1007/s00430-019-00648-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/19/2019] [Indexed: 12/01/2022]
Abstract
The complement system is a critical component of both innate and adaptive immune responses. It has both protective and pathogenic roles in viral infections. There are no studies regarding the role of complement system in Chandipura virus (CHPV) infection. The current study has investigated the role of complement pathways in the in vitro neutralization of CHPV in Vero E6 cells. Using normal human serum (NHS), heat-inactivated serum (HIS), human serum deficient of complement factor, respective reconstituted serum, assays like in vitro neutralization, real-time PCR, and flow cytometry-based tissue culture-based limited dose assay (TC-LDA) were carried out for assessing the activation of different complement pathways. NHS from 9/10 donors showed complement dependent neutralization, reduction in viral load and decrease in percentage of CHPV-positive cells compared to their HIS counterparts. EGTA or EDTA pretreatment experiments indicated that CHPV neutralization proceeds through the alternative pathway of the complement activation. Our data showed a strong dependence on C3 for the in vitro neutralization of CHPV. Disparity in CHPV neutralization levels between factor B-deficient and reconstituted sera could be attributed to amplification loop/“tick-over” mechanism. Assays using C3, C5, and C8 deficient sera indicated that complement-mediated CHPV neutralization and suppression of CHPV infectivity are primarily through C3 and C5, and not dependent on downstream complement factor C8. With no specific anti-viral treatment/vaccine against Chandipura, the current data, elucidating role of human complement system in the neutralization of CHPV, may help in designing effective therapeutics.
Collapse
Affiliation(s)
- Pooja Gupta
- Hepatitis Group, ICMR-National Institute of Virology, Pune, 130/1, Sus Road, Pashan, Pune, Maharashtra 411021 India
| | - Anuradha S. Tripathy
- Hepatitis Group, ICMR-National Institute of Virology, Pune, 130/1, Sus Road, Pashan, Pune, Maharashtra 411021 India
| |
Collapse
|
35
|
Peng M, Li Z, Niu D, Liu X, Dong Z, Li J. Complement factor B/C2 in molluscs regulates agglutination and illuminates evolution of the Bf/C2 family. FASEB J 2019; 33:13323-13333. [PMID: 31550175 DOI: 10.1096/fj.201901142rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Complement factor B/C2 family (Bf/C2F) proteins are core complement system components in vertebrates that are absent in invertebrates and have been lost by numerous species, raising evolutionary questions. At least 3 duplication events have occurred from Cnidaria (ancestor) to mammals. Type II Bf/C2 genes appeared during separation of Proterostomia and Deuterostomes. The second event occurred during separation of vertebrates and invertebrates, yielding type II-2 Bf/C2. The third event occurred when jawed and jawless fish were separated, eventually producing Bf and C2 genes. Herein, we report the second mollusc Sinonovacula constricta Bf/C2-type gene (ScBf). ScBf is similar to Ruditapes decussatus Bf-like because both lack the first complement control protein module at the N terminus present in mammalian Bf/C2 proteins. Uniquely, the Ser protease (SP) module at the C terminus of ScBf is ∼50 aa longer than in other complement factor B/C2-type (Bf/C2T) proteins, and is Glu-rich. Bf/C2T proteins in molluscs lack the catalytic Ser in the SP module. Surprisingly, ScBf regulates rabbit erythrocyte agglutination, during which it is localized on the erythrocyte surface. Thus, ScBf may mediate the agglutination cascade and may be an upstream regulator of this process. Our findings provide new insight into the origin of the Bf/C2F.-Peng, M., Li, Z., Niu, D., Liu, X., Dong, Z., Li, J. Complement factor B/C2 in molluscs regulates agglutination and illuminates evolution of the Bf/C2 family.
Collapse
Affiliation(s)
- Maoxiao Peng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhi Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Donghong Niu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| | - Xiaojun Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhiguo Dong
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| | - Jiale Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Co-Innovation Centre of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China; and
| |
Collapse
|
36
|
van den Bos RM, Pearce NM, Granneman J, Brondijk THC, Gros P. Insights Into Enhanced Complement Activation by Structures of Properdin and Its Complex With the C-Terminal Domain of C3b. Front Immunol 2019; 10:2097. [PMID: 31552043 PMCID: PMC6736995 DOI: 10.3389/fimmu.2019.02097] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/20/2019] [Indexed: 11/13/2022] Open
Abstract
Properdin enhances complement-mediated opsonization of targeted cells and particles for immune clearance. Properdin occurs as dimers, trimers and tetramers in human plasma, which recognize C3b-deposited surfaces, promote formation, and prolong the lifetime of C3bBb-enzyme complexes that convert C3 into C3b, thereby enhancing the complement-amplification loop. Here, we report crystal structures of monomerized properdin, which was produced by co-expression of separate N- and C-terminal constructs that yielded monomer-sized properdin complexes that stabilized C3bBb. Consistent with previous low-resolution X-ray and EM data, the crystal structures revealed ring-shaped arrangements that are formed by interactions between thrombospondin type-I repeat (TSR) domains 4 and 6 of one protomer interacting with the N-terminal domain (which adopts a short transforming-growth factor B binding protein-like fold) and domain TSR1 of a second protomer, respectively. Next, a structure of monomerized properdin in complex with the C-terminal domain of C3b showed that properdin-domain TSR5 binds along the C-terminal α-helix of C3b, while two loops, one from domain TSR5 and one from TSR6, extend and fold around the C3b C-terminus like stirrups. This suggests a mechanistic model in which these TSR5 and TSR6 "stirrups" bridge interactions between C3b and factor B or its fragment Bb, and thereby enhance formation of C3bB pro-convertases and stabilize C3bBb convertases. In addition, properdin TSR6 would sterically block binding of the protease factor I to C3b, thus limiting C3b proteolytic degradation. The presence of a valine instead of a third tryptophan in the canonical Trp-ladder of TSR domains in TSR4 allows a remarkable ca. 60°-domain bending motion of TSR4. Together with variable positioning of TSR2 and, putatively, TSR3, this explains the conformational flexibility required for properdin to form dimers, trimers, and tetramers. In conclusion, the results indicate that binding avidity of oligomeric properdin is needed to distinguish surface-deposited C3b molecules from soluble C3b or C3 and suggest that properdin-mediated interactions bridging C3b-B and C3b-Bb enhance affinity, thus promoting convertase formation and stabilization. These mechanisms explain the enhancement of complement-mediated opsonization of targeted cells and particle for immune clearance.
Collapse
Affiliation(s)
- Ramon M van den Bos
- Crystal and Structural Chemistry, Department of Chemistry, Faculty of Science, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Nicholas M Pearce
- Crystal and Structural Chemistry, Department of Chemistry, Faculty of Science, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Joke Granneman
- Crystal and Structural Chemistry, Department of Chemistry, Faculty of Science, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - T Harma C Brondijk
- Crystal and Structural Chemistry, Department of Chemistry, Faculty of Science, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Piet Gros
- Crystal and Structural Chemistry, Department of Chemistry, Faculty of Science, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
Hew BE, Fritzinger DC, Pangburn MK, Vogel CW. Identification of functionally important amino acid sequences in cobra venom factor using human C3/Cobra venom factor hybrid proteins. Toxicon 2019; 167:106-116. [DOI: 10.1016/j.toxicon.2019.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/18/2019] [Accepted: 06/13/2019] [Indexed: 11/28/2022]
|
38
|
Ekdahl KN, Mohlin C, Adler A, Åman A, Manivel VA, Sandholm K, Huber-Lang M, Fromell K, Nilsson B. Is generation of C3(H 2O) necessary for activation of the alternative pathway in real life? Mol Immunol 2019; 114:353-361. [PMID: 31446306 DOI: 10.1016/j.molimm.2019.07.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/18/2019] [Accepted: 07/31/2019] [Indexed: 01/30/2023]
Abstract
In the alternative pathway (AP) an amplification loop is formed, which is strictly controlled by various fluid-phase and cell-bound regulators resulting in a state of homeostasis. Generation of the "C3b-like" C3(H2O) has been described as essential for AP activation, since it conveniently explains how the initial fluid-phase AP convertase of the amplification loop is generated. Also, the AP has a status of being an unspecific pathway despite thorough regulation at different surfaces. During complement attack in pathological conditions and inflammation, large amounts of C3b are formed by the classical/lectin pathway (CP/LP) convertases. After the discovery of LP´s recognition molecules and its tight interaction with the AP, it is increasingly likely that the AP acts in vivo mainly as a powerful amplification mechanism of complement activation that is triggered by previously generated C3b molecules initiated by the binding of specific recognition molecules. Also in many pathological conditions caused by a dysregulated AP amplification loop such as paroxysmal nocturnal hemoglobulinuria (PNH) and atypical hemolytic uremic syndrome (aHUS), C3b is available due to minute LP and CP activation and/or generated by non-complement proteases. Therefore, C3(H2O) generation in vivo may be less important for AP activation during specific attack or dysregulated homeostasis, but may be an important ligand for C3 receptors in cell-cell interactions and a source of C3 for the intracellular complement reservoir.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden; Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden.
| | - Camilla Mohlin
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Anna Adler
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Amanda Åman
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Vivek Anand Manivel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Kerstin Sandholm
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Karin Fromell
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|
39
|
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
|
40
|
Békássy ZD, Kristoffersson AC, Rebetz J, Tati R, Olin AI, Karpman D. Aliskiren inhibits renin-mediated complement activation. Kidney Int 2018; 94:689-700. [DOI: 10.1016/j.kint.2018.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 03/28/2018] [Accepted: 04/05/2018] [Indexed: 11/17/2022]
|
41
|
Dobó J, Kocsis A, Gál P. Be on Target: Strategies of Targeting Alternative and Lectin Pathway Components in Complement-Mediated Diseases. Front Immunol 2018; 9:1851. [PMID: 30135690 PMCID: PMC6092519 DOI: 10.3389/fimmu.2018.01851] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022] Open
Abstract
The complement system has moved into the focus of drug development efforts in the last decade, since its inappropriate or uncontrolled activation has been recognized in many diseases. Some of them are primarily complement-mediated rare diseases, such as paroxysmal nocturnal hemoglobinuria, C3 glomerulonephritis, and atypical hemolytic uremic syndrome. Complement also plays a role in various multifactorial diseases that affect millions of people worldwide, such as ischemia reperfusion injury (myocardial infarction, stroke), age-related macular degeneration, and several neurodegenerative disorders. In this review, we summarize the potential advantages of targeting various complement proteins with special emphasis on the components of the lectin (LP) and the alternative pathways (AP). The serine proteases (MASP-1/2/3, factor D, factor B), which are responsible for the activation of the cascade, are straightforward targets of inhibition, but the pattern recognition molecules (mannose-binding lectin, other collectins, and ficolins), the regulatory components (factor H, factor I, properdin), and C3 are also subjects of drug development. Recent discoveries about cross-talks between the LP and AP offer new approaches for clinical intervention. Mannan-binding lectin-associated serine proteases (MASPs) are not just responsible for LP activation, but they are also indispensable for efficient AP activation. Activated MASP-3 has recently been shown to be the enzyme that continuously supplies factor D (FD) for the AP by cleaving pro-factor D (pro-FD). In this aspect, MASP-3 emerges as a novel feasible target for the regulation of AP activity. MASP-1 was shown to be required for AP activity on various surfaces, first of all on LPS of Gram-negative bacteria.
Collapse
Affiliation(s)
- József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andrea Kocsis
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
42
|
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
|
43
|
Zewde N, Morikis D. A computational model for the evaluation of complement system regulation under homeostasis, disease, and drug intervention. PLoS One 2018; 13:e0198644. [PMID: 29874282 PMCID: PMC5991421 DOI: 10.1371/journal.pone.0198644] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022] Open
Abstract
The complement system is an intricate defense network that rapidly removes invading pathogens. Although many complement regulators are present to protect host cells under homeostasis, the impairment of Factor H (FH) regulatory mechanism has been associated with several autoimmune and inflammatory diseases. To understand the dynamics involved in the pivotal balance between activation and regulation, we have developed a comprehensive computational model of the alternative and classical pathways of the complement system. The model is composed of 290 ordinary differential equations with 142 kinetic parameters that describe the state of complement system under homeostasis and disorder through FH impairment. We have evaluated the state of the system by generating concentration-time profiles for the biomarkers C3, C3a-desArg, C5, C5a-desArg, Factor B (FB), Ba, Bb, and fC5b-9 that are influenced by complement dysregulation. We show that FH-mediated disorder induces substantial levels of complement activation compared to homeostasis, by generating reduced levels of C3 and FB, and to a lesser extent C5, and elevated levels of C3a-desArg, Ba, Bb, C5a-desArg, and fC5b-9. These trends are consistent with clinically observed biomarkers associated with complement-mediated diseases. Furthermore, we introduced therapy states by modeling known inhibitors of the complement system, a compstatin variant (C3 inhibitor) and eculizumab (C5 inhibitor). Compstatin demonstrates strong restorative effects for early-stage biomarkers, such as C3a-desArg, FB, Ba, and Bb, and milder restorative effects for late-stage biomarkers, such as C5a-desArg and fC5b-9, whereas eculizumab has strong restorative effects on late-stage biomarkers, and negligible effects on early-stage biomarkers. These results highlight the need for patient-tailored therapies that target early complement activation at the C3 level, or late-stage propagation of the terminal cascade at the C5 level, depending on the specific FH-mediated disease and the manifestations of a patient's genetic profile in complement regulatory function.
Collapse
Affiliation(s)
- Nehemiah Zewde
- Department of Bioengineering, University of California, Riverside, California, United States of America
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, California, United States of America
| |
Collapse
|
44
|
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
|
45
|
Anaphylatoxin Signaling in Retinal Pigment and Choroidal Endothelial Cells: Characteristics and Relevance to Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:45-51. [PMID: 29721926 DOI: 10.1007/978-3-319-75402-4_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in the USA. Polymorphisms in various complement components are associated with an increased risk for AMD, and it has been hypothesized that an overactive complement system is partially responsible for the pathology of AMD. AMD is classified as early, intermediate, or late AMD, depending on the degree of the associated pathologies. Late AMD can be characterized as either lesions associated with neovascular AMD or geographic atrophy. Both sets of lesions are associated with pathology at the RPE/choroid interface, which include a thickening of Bruch's membrane, presence of drusen, and pigmentary alterations, and deterioration of the blood-retina barrier has been reported. These changes can lead to the slow degeneration and atrophy of the photoreceptors in the macula in dry AMD, or progress to choroidal neovascularization (CNV) and leakage of these new vessels in wet AMD. It has been shown previously that complement anaphylatoxins C3a and C5a, signaling via their respective G-protein-coupled receptors, can alter RPE cell function and promote choroidal neovascularization. However, it is important to note these components also play a role in tissue repair. Here we discuss anaphylatoxin signaling in AMD-related target cells and the potential implications for the design of anti-complement therapeutics.
Collapse
|
46
|
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
|
47
|
de Jorge EG, Yebenes H, Serna M, Tortajada A, Llorca O, de Córdoba SR. How novel structures inform understanding of complement function. Semin Immunopathol 2017; 40:3-14. [PMID: 28808775 DOI: 10.1007/s00281-017-0643-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
During the last decade, the complement field has experienced outstanding advancements in the mechanistic understanding of how complement activators are recognized, what C3 activation means, how protein complexes like the C3 convertases and the membrane attack complex are assembled, and how positive and negative complement regulators perform their function. All of this has been made possible mostly because of the contributions of structural biology to the study of the complement components. The wealth of novel structural data has frequently provided support to previously held knowledge, but often has added alternative and unexpected insights into complement function. Here, we will review some of these findings focusing in the alternative and terminal complement pathways.
Collapse
Affiliation(s)
- Elena Goicoechea de Jorge
- Department of Microbiology I (Immunology), Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Hugo Yebenes
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Marina Serna
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Agustín Tortajada
- Department of Microbiology I (Immunology), Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Oscar Llorca
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain.,Structural Biology Programme, CNIO, C/ Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Santiago Rodríguez de Córdoba
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain. .,Ciber de Enfermedades Raras, Madrid, Spain.
| |
Collapse
|
48
|
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: 69] [Impact Index Per Article: 8.6] [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
|
49
|
Yorulmaz Avsar S, Jackman JA, Kim MC, Yoon BK, Hunziker W, Cho NJ. Immobilization Strategies for Functional Complement Convertase Assembly at Lipid Membrane Interfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:7332-7342. [PMID: 28683197 DOI: 10.1021/acs.langmuir.7b01465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The self-assembly formation of complement convertases-essential biomacromolecular complexes that amplify innate immune responses-is triggered by protein adsorption. Herein, a supported lipid bilayer platform was utilized to investigate the effects of covalent and noncovalent tethering strategies on the self-assembly of alternative pathway C3 convertase components, starting with C3b protein adsorption followed bythe addition of factors B and D. Quartz crystal microbalance-dissipation (QCM-D) experiments measured the real-time kinetics of convertase assembly onto supported lipid bilayers. The results demonstrate that the nature of C3b immobilization onto supported lipid bilayers is a key factor governing convertase assembly. The covalent attachment of C3b to maleimide-functionalized supported lipid bilayers promoted the self-assembly of functional C3 convertase in the membrane-associated state and further enabled successful evaluation of a clinically relevant complement inhibitor, compstatin. By contrast, noncovalent attachment of C3b to negatively charged supported lipid bilayers also permitted C3b protein uptake, albeit membrane-associated C3b did not support convertase assembly in this case. Taken together, the findings in this work demonstrate that the attachment scheme for immobilizing C3b protein at lipid membrane interfaces is critical for downstream C3 convertase assembly, thereby offering guidance for the design and evaluation of membrane-associated biomacromolecular complexes.
Collapse
Affiliation(s)
- Saziye Yorulmaz Avsar
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore 138673, Singapore
| | - Joshua A Jackman
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Min Chul Kim
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Bo Kyeong Yoon
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Walter Hunziker
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore 117599, Singapore
- Singapore Eye Research Institute , Singapore 169856, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University , 62 Nanyang Drive, Singapore 637459, Singapore
| |
Collapse
|
50
|
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
|