1
|
Zheng G, Wu L, Bouamar H, Cserhati M, Chiu YC, Hinck CS, Wieteska Ł, Zeballos Torrez CR, Hu R, Easley A, Chen Y, Hinck AP, Cigarroa FG, Sun LZ. Ficolin-3 induces apoptosis and suppresses malignant property of hepatocellular carcinoma cells via the complement pathway. Life Sci 2024; 357:123103. [PMID: 39357793 DOI: 10.1016/j.lfs.2024.123103] [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: 06/07/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
AIMS Ficolin 3 (FCN3) has the highest complement-activating capacity through the lectin pathway and is synthesized mainly in the liver and lung. Yet, its potential molecular mechanism in hepatocarcinogenesis is not fully understood. MATERIALS AND METHODS The expression of FCN3 in hepatocellular carcinoma (HCC) tumor and non-tumor tissues was analyzed by RT-qPCR, Western blotting and immunofluorescence staining assays. Lentivector-mediated ectopic overexpression was performed to explore the role of FCN3 in vitro and in vivo. Whether FCN3 inhibited HCC cell growth and survival via complement pathway was determined with immunocytochemical staining for C3b, membrane attack complex (MAC) formation and complement killing assay using recombinant FCN3 (rFCN3) in combination with human serum with or without heat inactivation, and with C6 blocking antibody. KEY FINDINGS The transcript and protein of FCN3 were found to be remarkably down-regulated in HCC tumor tissues. FCN3 expression was found to be associated with better survival of HCC patients. Restoration of FCN3 expression significantly inhibited proliferation, migration and anchorage independent growth of HCC cell lines, and xenograft tumor growth. FCN3 expression induced apoptosis of HCC cells. C3 and MAC formation was stimulated by FCN3 overexpression or rFCN3 treatment. rFCN3 enhanced human serum-induced complement activation and cell death. C6 blocking antibody significantly attenuated complement-mediated cell death and restored the growth of FCN3-overexpressing HCC cells. SIGNIFICANCE FCN3 has a malignant suppressor role in HCC cells. Our study provides new insights into the molecular mechanisms that drive HCC progression and potential therapeutic targets for treating HCC.
Collapse
Affiliation(s)
- Guixi Zheng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America; Department of Clinical Laboratory, Qilu Hospital of Shandong University, China
| | - Lianqiu Wu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Hakim Bouamar
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Matyas Cserhati
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Yu-Chiao Chiu
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Cinthia S Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, PA, United States of America
| | - Łukasz Wieteska
- Department of Structural Biology, University of Pittsburgh School of Medicine, PA, United States of America
| | - Carla R Zeballos Torrez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Ruolei Hu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Acarizia Easley
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Yidong Chen
- Department of Structural Biology, University of Pittsburgh School of Medicine, PA, United States of America; Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, TX, United States of America
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, PA, United States of America
| | - Francisco G Cigarroa
- Transplant Center, University of Texas Health Science Center at San Antonio, TX, United States of America.
| | - Lu-Zhe Sun
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, United States of America.
| |
Collapse
|
2
|
Riedelová Z, de Los Santos Pereira A, Dorado Daza DF, Májek P, Dyčka F, Riedel T. Mass-Spectrometric Identification of Proteins and Pathways Responsible for Fouling on Poly(ethylene glycol) Methacrylate Polymer Brushes. Macromol Biosci 2024; 24:e2300558. [PMID: 38350051 DOI: 10.1002/mabi.202300558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/09/2024] [Indexed: 02/15/2024]
Abstract
Prevention of fouling from proteins in blood plasma attracts significant efforts, and great progress is made in identifying surface coatings that display antifouling properties. In particular, poly(ethylene glycol) (PEG) is widely used and dense PEG-like cylindrical brushes of poly[oligo(ethylene glycol) methacrylate] (poly(OEGMA)) can drastically reduce blood plasma fouling. Herein, a comprehensive study of the variation of blood plasma fouling on this surface, including the analysis of the composition of protein deposits on poly(OEGMA) coatings after contact with blood plasma from many different donors, is reported. Correlation between the plasma fouling behavior and protein deposit composition points to the activation of the complement system as the main culprit of dramatically increased and accelerated deposition of blood plasma proteins on this type of antifouling coating, specifically through the classical pathway. These findings are consistent with observations on PEGylated drug carriers and highlight the importance of understanding the potential interactions between antifouling coatings and their environment.
Collapse
Affiliation(s)
- Zuzana Riedelová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Andres de Los Santos Pereira
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Diego Fernando Dorado Daza
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Pavel Májek
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague, 128 00, Czech Republic
| | - Filip Dyčka
- Faculty of Science, University of South Bohemia, Branišovská 1760, České Budějovice, 370 05, Czech Republic
| | - Tomáš Riedel
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| |
Collapse
|
3
|
Götz MP, Duque Villegas MA, Fageräng B, Kerfin A, Skjoedt MO, Garred P, Rosbjerg A. Transient Binding Dynamics of Complement System Pattern Recognition Molecules on Pathogens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1493-1503. [PMID: 38488502 DOI: 10.4049/jimmunol.2300768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 04/17/2024]
Abstract
Previous studies of pattern recognition molecules (PRMs) of the complement system have revealed difficulties in observing binding on pathogens such as Aspergillus fumigatus and Escherichia coli, despite complement deposition indicative of classical and lectin pathway activation. Thus, we investigated the binding dynamics of PRMs of the complement system, specifically C1q of the classical pathway and mannose-binding lectin (MBL) of the lectin pathway. We observed consistently increasing deposition of essential complement components such as C4b, C3b, and the terminal complement complex on A. fumigatus and E. coli. However, C1q and MBL binding to the surface rapidly declined during incubation after just 2-4 min in 10% plasma. The detachment of C1q and MBL can be linked to complement cascade activation, as the PRMs remain bound in the absence of plasma. The dissociation and the fate of C1q and MBL seem to have different mechanistic functions. Notably, C1q dynamics were associated with local C1 complex activation. When C1s was inhibited in plasma, C1q binding not only remained high but further increased over time. In contrast, MBL binding was inversely correlated with total and early complement activation due to MBL binding being partially retained by complement inhibition. Results indicate that detached MBL might be able to functionally rebind to A. fumigatus. In conclusion, these results reveal a (to our knowledge) novel "hit-and-run" complement-dependent PRM dynamic mechanism on pathogens. These dynamics may have profound implications for host defense and may help increase the functionality and longevity of complement-dependent PRMs in circulation.
Collapse
Affiliation(s)
- Maximilian Peter Götz
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Immunology and Infectious Diseases, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute for Systemic Inflammation Research, Medicine Section, University of Lübeck, Lübeck, Germany
| | - Mario Alejandro Duque Villegas
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Infection Immunology, Research Center Borstel, Borstel, Germany
- Molecular and Experimental Mycobacteriology, Research Center Borstel, Borstel, Germany
| | - Beatrice Fageräng
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Aileen Kerfin
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Institute for Systemic Inflammation Research, Medicine Section, University of Lübeck, Lübeck, Germany
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Gabriela R, Vera V, Pavel R, Helena R, Igor S, Marie D, Marketa M, Alena MF, Ales T. Discovering the Radiation Biomarkers in the Plasma of Total-Body Irradiated Leukemia Patients. Radiat Res 2024; 201:418-428. [PMID: 38315067 DOI: 10.1667/rade-23-00137.1] [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: 07/21/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
The increased risk of acute large-scale radiological exposure for the world's population underlines the need for optimal radiation biomarkers. Ionizing radiation triggers a complex response by the genome, proteome, and metabolome, all of which have been reported as suitable indicators of radiation-induced damage in vivo. This study analyzed peripheral blood samples from total-body irradiation (TBI) leukemia patients through mass spectrometry (MS) to identify and quantify differentially regulated proteins in plasma before and after irradiation. In brief, samples were taken from 16 leukemic patients prior to and 24 h after TBI (2 × 2.0 Gy), processed with Tandem Mass Tag isobaric labelling kit (TMTpro-16-plex), and analyzed by MS. In parallel, label-free relative quantification was performed with a RP-nanoLC-ESI-MS/MS system in a Q-Exactive mass spectrometer. Protein identification was done in Proteome Discoverer v.2.2 platform (Thermo). Data is available via ProteomeXchange with identifier PXD043516. Using two different methods, we acquired two datasets of up-regulated (ratio ≥ 1.2) or down-regulated (ratio ≤ 0.83) plasmatic proteins 24 h after irradiation, identifying 356 and 346 proteins in the TMT-16plex and 285 and 308 label-free analyses, respectively (P ≤ 0.05). Combining the two datasets yielded 15 candidates with significant relation to gamma-radiation exposure. The majority of these proteins were associated with the inflammatory response and lipid metabolism. Subsequently, from these, five proteins showed the strongest potential as radiation biomarkers in humans (C-reactive protein, Alpha amylase 1A, Mannose-binding protein C, Phospholipid transfer protein, and Complement C5). These candidate biomarkers might have implications for practical biological dosimetry.
Collapse
Affiliation(s)
- Rydlova Gabriela
- Department of Radiobiology
- Department of Biology, Faculty of Natural Sciences, University of Hradec Králové, Czech Republic, Hradec Králové, Czech Republic
| | | | | | - Rehulkova Helena
- Department of Toxicology, Faculty of Military Health Sciences, University of Defence, Hradec Králové, Czech Republic
| | - Sirak Igor
- Department of Oncology and Radiotherapy and 4th Department of Internal Medicine - Haematology, University Hospital, Hradec Kralove, Czech Republic
| | - Davidkova Marie
- Department of Radiation Dosimetry, Nuclear Physics Institute of the Czech Academy of Sciences, Prague, Czech Republic
| | - Markova Marketa
- Department of Haematology and Blood Transfusion, University Hospital Na Bulovce, Prague, Czech Republic
| | - Myslivcova-Fucikova Alena
- Department of Biology, Faculty of Natural Sciences, University of Hradec Králové, Czech Republic, Hradec Králové, Czech Republic
| | | |
Collapse
|
5
|
Lindelöf L, Rantapää-Dahlqvist S, Lundtoft C, Sandling JK, Leonard D, Sayadi A, Rönnblom L, Enocsson H, Sjöwall C, Jönsen A, Bengtsson AA, Hong MG, Diaz-Gallo LM, Bianchi M, Kozyrev SV, Lindblad-Toh K, Nilsson Ekdahl K, Nilsson B, Gunnarsson I, Svenungsson E, Eriksson O. A survey of ficolin-3 activity in Systemic Lupus Erythematosus reveals a link to hematological disease manifestations and autoantibody profile. J Autoimmun 2024; 143:103166. [PMID: 38219652 DOI: 10.1016/j.jaut.2023.103166] [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: 10/26/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
The complement system plays a central role in the pathogenesis of Systemic Lupus Erythematosus (SLE), but most studies have focused on the classical pathway. Ficolin-3 is the main initiator of the lectin pathway of complement in humans, but its role in systemic autoimmune disease has not been conclusively determined. Here, we combined biochemical and genetic approaches to assess the contribution of ficolin-3 to SLE risk and disease manifestations. Ficolin-3 activity was measured by a functional assay in serum or plasma samples from Swedish SLE patients (n = 786) and controls matched for age and sex (n = 566). Genetic variants in an extended 300 kb genomic region spanning the FCN3 locus were analyzed for their association with ficolin-3 activity and SLE manifestations in a Swedish multicenter cohort (n = 985). Patients with ficolin-3 activity in the highest tertile showed a strong enrichment in an SLE cluster defined by anti-Sm/DNA/nucleosome antibodies (OR 3.0, p < 0.001) and had increased rates of hematological disease (OR 1.4, p = 0.078) and lymphopenia (OR = 1.6, p = 0.039). Genetic variants associated with low ficolin-3 activity mapped to an extended haplotype in high linkage disequilibrium upstream of the FCN3 gene. Patients carrying the lead genetic variant associated with low ficolin-3 activity had a lower frequency of hematological disease (OR 0.67, p = 0.018) and lymphopenia (OR 0.63, p = 0.031) and fewer autoantibodies (p = 0.0019). Loss-of-function variants in the FCN3 gene were not associated with SLE, but four (0.5 %) SLE patients developed acquired ficolin-3 deficiency where ficolin-3 activity in serum was depleted following diagnosis of SLE. Taken together, our results provide genetic and biochemical evidence that implicate the lectin pathway in hematological SLE manifestations. We also identify lectin pathway activation through ficolin-3 as a factor that contributes to the autoantibody response in SLE.
Collapse
Affiliation(s)
- Linnea Lindelöf
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Christian Lundtoft
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Johanna K Sandling
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Dag Leonard
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Ahmed Sayadi
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Helena Enocsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Christopher Sjöwall
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Andreas Jönsen
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Mun-Gwan Hong
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lina-Marcela Diaz-Gallo
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Matteo Bianchi
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sergey V Kozyrev
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kristina Nilsson Ekdahl
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; Linnaeus Center for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Elisabet Svenungsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Oskar Eriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
6
|
DIA-based proteomics analysis of serum-derived exosomal proteins as potential candidate biomarkers for intrahepatic cholestasis in pregnancy. Arch Gynecol Obstet 2022; 308:79-89. [PMID: 35849169 DOI: 10.1007/s00404-022-06703-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/03/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Data-independent acquisition (DIA) is one of the most powerful and reproducible proteomic technologies for large-scale digital qualitative and quantitative research. The aim of this study was to use proteomic methodologies for the identification of biomarkers that are over or underexpressed in women with intrahepatic cholestasis of pregnancy (ICP) compared with controls and discover a potential biomarker panel for ICP detection. METHODS The participants included 11 ICP patients and 11 healthy pregnant women as controls. The clinical characteristic data and the laboratory biochemical data were collected at the time of recruitment. Then, a data-independent acquisition (DIA)-based proteomics approach was used to identify differentially expressed proteins (DEPs) in serum exosomes between ICP patients and controls. Finally, bioinformatics analysis was used to identify the relevant processes in which these DEPs were involved. RESULTS The proteomics results showed that there were 162 DEPs in serum exosomes between pregnant women with ICP and healthy pregnant women, of which 106 were upregulated and 56 were downregulated in ICP. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the identified proteins were functionally related to specific cell processes including apoptosis, lipid metabolism, immune response and cell proliferation, and metabolic disorders, suggesting that these may be primary causative factors in ICP pathogenesis. Meanwhile, complement and coagulation cascades may be closely related to the development of ICP. Receiver operating characteristic curve (ROC) analysis showed that the area under the curve values of Elongation factor 1-alpha 1, Beta-2-glycoprotein I, Zinc finger protein 238, CP protein and Ficolin-3 were all approximately 0.9, indicating the promising diagnostic value of these proteins. CONCLUSIONS This preliminary work provides a better understanding of the proteomic alterations in the serum exosomes of pregnant women with ICP.
Collapse
|
7
|
Caron B, Patin E, Rotival M, Charbit B, Albert ML, Quintana-Murci L, Duffy D, Rausell A. Integrative genetic and immune cell analysis of plasma proteins in healthy donors identifies novel associations involving primary immune deficiency genes. Genome Med 2022; 14:28. [PMID: 35264221 PMCID: PMC8905727 DOI: 10.1186/s13073-022-01032-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Blood plasma proteins play an important role in immune defense against pathogens, including cytokine signaling, the complement system, and the acute-phase response. Recent large-scale studies have reported genetic (i.e., protein quantitative trait loci, pQTLs) and non-genetic factors, such as age and sex, as major determinants to inter-individual variability in immune response variation. However, the contribution of blood-cell composition to plasma protein heterogeneity has not been fully characterized and may act as a mediating factor in association studies. METHODS Here, we evaluated plasma protein levels from 400 unrelated healthy individuals of western European ancestry, who were stratified by sex and two decades of life (20-29 and 60-69 years), from the Milieu Intérieur cohort. We quantified 229 proteins by Luminex in a clinically certified laboratory and their levels of variation were analyzed together with 5.2 million single-nucleotide polymorphisms. With respect to non-genetic variables, we included 254 lifestyle and biochemical factors, as well as counts of seven circulating immune cell populations measured by hemogram and standardized flow cytometry. RESULTS Collectively, we found 152 significant associations involving 49 proteins and 20 non-genetic variables. Consistent with previous studies, age and sex showed a global, pervasive impact on plasma protein heterogeneity, while body mass index and other health status variables were among the non-genetic factors with the highest number of associations. After controlling for these covariates, we identified 100 and 12 pQTLs acting in cis and trans, respectively, collectively associated with 87 plasma proteins and including 19 novel genetic associations. Genetic factors explained the largest fraction of the variability of plasma protein levels, as compared to non-genetic factors. In addition, blood-cell fractions, including leukocytes, lymphocytes, monocytes, neutrophils, eosinophils, basophils, and platelets, had a larger contribution to inter-individual variability than age and sex and appeared as confounders of specific genetic associations. Finally, we identified new genetic associations with plasma protein levels of five monogenic Mendelian disease genes including two primary immunodeficiency genes (Ficolin-3 and FAS). CONCLUSIONS Our study identified novel genetic and non-genetic factors associated to plasma protein levels which may inform health status and disease management.
Collapse
Affiliation(s)
- Barthelemy Caron
- Université de Paris, INSERM UMR1163, Imagine Institute, Clinical Bioinformatics Laboratory, F-75006, Paris, France
| | - Etienne Patin
- Human Evolutionary Genetics Unit, Institut Pasteur, UMR2000, CNRS, Université de Paris, F-75015, Paris, France
| | - Maxime Rotival
- Human Evolutionary Genetics Unit, Institut Pasteur, UMR2000, CNRS, Université de Paris, F-75015, Paris, France
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université de Paris, F-75015, Paris, France
| | | | - Lluis Quintana-Murci
- Human Evolutionary Genetics Unit, Institut Pasteur, UMR2000, CNRS, Université de Paris, F-75015, Paris, France
- Human Genomics and Evolution, Collège de France, F-75005, Paris, France
| | - Darragh Duffy
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université de Paris, F-75015, Paris, France.
- Translational Immunology Unit, Institut Pasteur, Université de Paris, F-75015, Paris, France.
| | - Antonio Rausell
- Université de Paris, INSERM UMR1163, Imagine Institute, Clinical Bioinformatics Laboratory, F-75006, Paris, France.
- Service de Médecine Génomique des Maladies Rares, AP-HP, Necker Hospital for Sick Children, F-75015, Paris, France.
| |
Collapse
|
8
|
Nielsen TL, Pilely K, Lund KP, Warming PE, Plesner LL, Iversen KK, Garred P. Hemodialysis leads to plasma depletion of lectin complement pathway initiator molecule ficolin-2. Hemodial Int 2021; 25:479-488. [PMID: 34132045 DOI: 10.1111/hdi.12948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/22/2021] [Accepted: 05/23/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION This study aimed to investigate changes in complement system-related molecules in patients undergoing hemodialysis. METHODS Patients >18 years of age on maintenance hemodialysis were included. Using enzyme-linked immunosorbent assays (ELISA) methods complement related molecules ficolin-1, ficolin-2, ficolin-3 mannose-binding lectin, long pentraxin 3, complement activation products C3c, and complement activation potentials were measured before and after a single hemodialysis treatment. All patients were dialyzed with synthetic high flux filters >1.6 m2 , respectively, Polyamix and Polysulfone, and the Kt/V was maintained >1.3. FINDINGS Three hundred and four patients were included. There was a modest decrease in plasma level of ficolin-1 (p < 0.001). Ficolin-2 was virtually depleted with median 3.9 (interquartile range [IQR]: 2.6-6.1, range 0.3-13.5) μg/ml before dialysis to median 0.0 (IQR: 0.0-0.5, range 0.0-5.5) μg/ml after dialysis (p < 0.001). No significant difference before and after hemodialysis was seen for mannose-binding lectin and long pentraxin 3 (p > 0.05). In a random subgroup of 160 patients ficolin-2-binding, ficolin-3-mediated lectin pathway capacity and classical pathway capacity were significantly decreased due to hemodialysis. The complement capacity of the alternative pathway was increased after hemodialysis (p = 0.0101), while mannose-binding lectin-mediated lectin pathway capacity was unaltered (p = 0.79). There was an increase in the complement activation product C3c (p < 0.0001), while the concentration of total C4 and C3 did not change (p > 0.158). Multivariate Cox proportional hazard analyses showed an increased risk for all-cause mortality with increasing ficolin-2 (p = 0.002) after hemodialysis. DISCUSSION Plasma ficolin-2 was virtually depleted from the circulation after hemodialysis. However, elevated plasma ficolin-2 levels after hemodialysis was independently associated with increased mortality.
Collapse
Affiliation(s)
- Ture Lange Nielsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Herlev Hospital, Herlev, Denmark
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Kit P Lund
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Galbraith MD, Kinning KT, Sullivan KD, Baxter R, Araya P, Jordan KR, Russell S, Smith KP, Granrath RE, Shaw JR, Dzieciatkowska M, Ghosh T, Monte AA, D'Alessandro A, Hansen KC, Benett TD, Hsieh EWY, Espinosa JM. Seroconversion stages COVID19 into distinct pathophysiological states. eLife 2021; 10:e65508. [PMID: 33724185 PMCID: PMC7963480 DOI: 10.7554/elife.65508] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
COVID19 is a heterogeneous medical condition involving diverse underlying pathophysiological processes including hyperinflammation, endothelial damage, thrombotic microangiopathy, and end-organ damage. Limited knowledge about the molecular mechanisms driving these processes and lack of staging biomarkers hamper the ability to stratify patients for targeted therapeutics. We report here the results of a cross-sectional multi-omics analysis of hospitalized COVID19 patients revealing that seroconversion status associates with distinct underlying pathophysiological states. Low antibody titers associate with hyperactive T cells and NK cells, high levels of IFN alpha, gamma and lambda ligands, markers of systemic complement activation, and depletion of lymphocytes, neutrophils, and platelets. Upon seroconversion, all of these processes are attenuated, observing instead increases in B cell subsets, emergency hematopoiesis, increased D-dimer, and hypoalbuminemia. We propose that seroconversion status could potentially be used as a biosignature to stratify patients for therapeutic intervention and to inform analysis of clinical trial results in heterogenous patient populations.
Collapse
Affiliation(s)
- Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pediatrics, Division of Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Ryan Baxter
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kimberly R Jordan
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Seth Russell
- Data Science to Patient Value, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Jessica R Shaw
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public HealthAuroraUnited States
| | - Andrew A Monte
- Department of Emergency Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Tellen D Benett
- Department of Pediatrics, Sections of Informatics and Data Science and Critical Care Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Elena WY Hsieh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pediatrics, Division of Allergy/Immunology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
10
|
Galbraith MD, Kinning KT, Sullivan KD, Baxter R, Araya P, Jordan KR, Russell S, Smith KP, Granrath RE, Shaw J, Dzieciatkowska M, Ghosh T, Monte AA, D’Alessandro A, Hansen KC, Bennett TD, Hsieh EW, Espinosa JM. Seroconversion stages COVID19 into distinct pathophysiological states. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.05.20244442. [PMID: 33330890 PMCID: PMC7743101 DOI: 10.1101/2020.12.05.20244442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
COVID19 is a heterogeneous medical condition involving a suite of underlying pathophysiological processes including hyperinflammation, endothelial damage, thrombotic microangiopathy, and end-organ damage. Limited knowledge about the molecular mechanisms driving these processes and lack of staging biomarkers hamper the ability to stratify patients for targeted therapeutics. We report here the results of a cross-sectional multi-omics analysis of hospitalized COVID19 patients revealing that seroconversion status associates with distinct underlying pathophysiological states. Seronegative COVID19 patients harbor hyperactive T cells and NK cells, high levels of IFN alpha, gamma and lambda ligands, markers of systemic complement activation, neutropenia, lymphopenia and thrombocytopenia. In seropositive patients, all of these processes are attenuated, observing instead increases in B cell subsets, emergency hematopoiesis, increased markers of platelet activation, and hypoalbuminemia. We propose that seroconversion status could potentially be used as a biosignature to stratify patients for therapeutic intervention and to inform analysis of clinical trial results in heterogenous patient populations.
Collapse
Affiliation(s)
- Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T. Kinning
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Division of Developmental Biology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan Baxter
- Department of Immunology and Microbiology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Seth Russell
- Data Science to Patient Value; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P. Smith
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E. Granrath
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Shaw
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew A. Monte
- Department of Emergency Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tellen D. Bennett
- Department of Pediatrics, Sections of Informatics and Data Science and Critical Care Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena W.Y. Hsieh
- Department of Immunology and Microbiology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Division of Allergy/Immunology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology; University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Correspondence to:
| |
Collapse
|
11
|
Kjældgaard AL, Pilely K, Olsen KS, Lauritsen AØ, Pedersen SW, Møller K, Garred P. Amyotrophic lateral sclerosis and the innate immune system: protocol for establishing a biobank and statistical analysis plan. BMJ Open 2020; 10:e037753. [PMID: 32759248 PMCID: PMC7409992 DOI: 10.1136/bmjopen-2020-037753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a devastating, progressive disease that causes degeneration of the motor neurons leading to paresis of the bulbar and the skeletal musculature. The pathogenesis of ALS remains unknown. We will test the hypothesis that the complement system is involved in the pathophysiology of ALS. This protocol article describes our efforts to establish a national Danish ALS biobank. The primary aim is to obtain biological material from patients with ALS for the current study as well as for future studies. METHODS AND ANALYSIS We intend to establish an observational ALS biobank; some of the material from this biobank will be used for a prospective, observational case-control study. The participants are patients with ALS, neurologically healthy controls and non-ALS neurological controls. Each participant consents to be interviewed and to donate blood and cerebrospinal fluid to the biobank. Analysis of the complement system will be carried out on the three groups of patients and compared. ETHICS AND DISSEMINATION The project has been approved by the Committees on Health Research Ethics in the Capital Region of Denmark (Approval number H-16017145) and the Danish Data Protection Agency (file number 2012-58-0004). All results will be published in peer-reviewed, medical journals and presented at scientific conferences. TRIAL REGISTRATION NUMBER NCT02869048.
Collapse
Affiliation(s)
- Anne-Lene Kjældgaard
- Neuroanaesthesiology, The Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Diagnostic Centre, Rigshospitalet, Copenhagen, Denmark
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Diagnostic Centre, Rigshospitalet, Copenhagen, Denmark
| | | | - Anne Øberg Lauritsen
- Neuroanaesthesiology, The Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
| | | | - Kirsten Møller
- Neuroanaesthesiology, The Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Diagnostic Centre, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Miles AM, Huson HJ. Time- and population-dependent genetic patterns underlie bovine milk somatic cell count. J Dairy Sci 2020; 103:8292-8304. [PMID: 32622601 DOI: 10.3168/jds.2020-18322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
The objective of this study was to determine whether genetic regulation of bovine milk somatic cell count (SCC) varied throughout the course of an individual lactation and to identify quantitative trait loci (QTL) that may differentiate populations of chronically mastitic and robustly healthy cows. Milk SCC has long been a proxy for clinical mastitis diagnosis in management and genetic improvement strategies to control the disease. Cows (n = 471) were genotyped on the Illumina BovineHD 777K BeadChip (Illumina Inc., San Diego, CA), and composite milk samples were collected for SCC at 0-1 d in milk (DIM), 3-5 DIM, 10-14 DIM, 90-110 DIM, and 210-230 DIM, with each time span representing key physiological transitions for the cow. Median lactation somatic cell score (SCS) and area under the SCS curve were calculated from farm test data. A total of 8 genome-wide associations were performed and 167 SNP spanning the genome were significantly associated (false discovery rate <0.05). Of these associated regions, 27 of 48 associated QTL were novel for clinical mastitis or SCC. The linkage disequilibrium block surrounding the associated QTL or a 1-Mb window in the absence of linkage disequilibrium was interrogated for candidate genes, and many of those identified were related to multiple arms of the immune system, including toll-like receptor signaling, macrophage activation, B-cell maturation, T-cell recruitment, and the complement pathway. These genes included EXOC4, BAMBI, ITSN2, IL34, FCN3, CD8A, and CD8B. In addition, we identified populations of robustly healthy (SCS ≤4 from 10-14 DIM until study end), chronically mastitic (SCS >4 from 10-14 DIM until study end), and average cows with fluctuating SCS, and calculated fixation indices to identify regions of the genome differentiating these 3 populations. A total of 12 SNP were identified that showed moderate allelic differentiation (Wright's F statistic, FST ≥ 0.4) between the "chronic," "healthy," and "average" populations of cows. Candidate genes in the region surrounding differentiated QTL were related to cell signaling and immune response, such as JAKMIP1 and MADCAM1. The wide range of significantly associated QTL spanning the genome and the diversity of gene functions reinforces that mastitis is a complex trait and suggests that selection based on lactation stage-specific SCS rather than a generalized score may lead to greater success in breeding mastitis-resistant cows.
Collapse
Affiliation(s)
- Asha M Miles
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - Heather J Huson
- Department of Animal Science, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
13
|
Talsma DT, Poppelaars F, Dam W, Meter-Arkema AH, Vivès RR, Gál P, Boons GJ, Chopra P, Naggi A, Seelen MA, Berger SP, Daha MR, Stegeman CA, van den Born J. MASP-2 Is a Heparin-Binding Protease; Identification of Blocking Oligosaccharides. Front Immunol 2020; 11:732. [PMID: 32425936 PMCID: PMC7212410 DOI: 10.3389/fimmu.2020.00732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/31/2020] [Indexed: 12/30/2022] Open
Abstract
It is well-known that heparin and other glycosaminoglycans (GAGs) inhibit complement activation. It is however not known whether fractionation and/or modification of GAGs might deliver pathway-specific inhibition of the complement system. Therefore, we evaluated a library of GAGs and their derivatives for their functional pathway specific complement inhibition, including the MASP-specific C4 deposition assay. Interaction of human MASP-2 with heparan sulfate/heparin was evaluated by surface plasmon resonance, ELISA and in renal tissue. In vitro pathway-specific complement assays showed that highly sulfated GAGs inhibited all three pathways of complement. Small heparin- and heparan sulfate-derived oligosaccharides were selective inhibitors of the lectin pathway (LP). These small oligosaccharides showed identical inhibition of the ficolin-3 mediated LP activation, failed to inhibit the binding of MBL to mannan, but inhibited C4 cleavage by MASPs. Hexa- and pentasulfated tetrasaccharides represent the smallest MASP inhibitors both in the functional LP assay as well in the MASP-mediated C4 assay. Surface plasmon resonance showed MASP-2 binding with heparin and heparan sulfate, revealing high Kon and Koff rates resulted in a Kd of ~2 μM and confirmed inhibition by heparin-derived tetrasaccharide. In renal tissue, MASP-2 partially colocalized with agrin and heparan sulfate, but not with activated C3, suggesting docking, storage, and potential inactivation of MASP-2 by heparan sulfate in basement membranes. Our data show that highly sulfated GAGs mediated inhibition of all three complement pathways, whereas short heparin- and heparan sulfate-derived oligosaccharides selectively blocked the lectin pathway via MASP-2 inhibition. Binding of MASP-2 to immobilized heparan sulfate/heparin and partial co-localization of agrin/heparan sulfate with MASP, but not C3b, might suggest that in vivo heparan sulfate proteoglycans act as a docking platform for MASP-2 and possibly prevent the lectin pathway from activation.
Collapse
Affiliation(s)
- Ditmer T Talsma
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Felix Poppelaars
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Wendy Dam
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Anita H Meter-Arkema
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Peter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands.,Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Pradeep Chopra
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | | | - Marc A Seelen
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Stephan P Berger
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Mohamed R Daha
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Coen A Stegeman
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Jacob van den Born
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | | |
Collapse
|
14
|
Kirketerp-Møller N, Bayarri-Olmos R, Krogfelt KA, Garred P. C1q/TNF-Related Protein 6 Is a Pattern Recognition Molecule That Recruits Collectin-11 from the Complement System to Ligands. THE JOURNAL OF IMMUNOLOGY 2020; 204:1598-1606. [PMID: 32041782 DOI: 10.4049/jimmunol.1901316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/06/2020] [Indexed: 11/19/2022]
Abstract
C1q/TNF-related protein (CTRP) 6 is a member of the CTRP protein family associated with the regulation of cellular and endocrine processes. CTRP6 contains collagen and globular structures, resembling the pattern recognition molecules (PRMs) of the classical and lectin complement pathways. We expressed human CTRP6 in Chinese hamster ovary cells and investigated the binding to different putative ligands (acetylated BSA [AcBSA], zymosan, mannan, and LPS from Escherichia coli and Salmonella as well as to the monosaccharides l-fucose, d-mannose, N-acetylglucosamine, N-acetylgalactosamine, and galactose). Furthermore, we investigated the binding of CTRP6 to various Gram-negative bacteria as well as PRMs and enzymes of the lectin complement pathway. We found that CTRP6 bound to AcBSA and to a lesser extent to zymosan. Using EDTA as chelating agent, we observed an increased binding to AcBSA, zymosan and the two strains of LPS. We detected no binding to mannan and BSA. We identified l-fucose as a ligand for CTRP6 and that it bound to certain enteroaggregative Escherichia coli and Pseudomonas aeruginosa isolates, whereas to other bacterial isolates, no binding was observed. CTRP6 did not appear to interact directly with the activating enzymes of the lectin pathway; however, we could show the specific recruitment of collectin-11 and subsequent initiation of the complement cascade through deposition of C4. In conclusion, our results demonstrate the binding of CTRP6 to a variety of microbial and endogenous ligands identifying CTRP6 as a novel human lectin and PRM of importance for complement recognition and innate immunity.
Collapse
Affiliation(s)
- Nikolaj Kirketerp-Møller
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Karen Angeliki Krogfelt
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, 2300 Copenhagen, Denmark; and.,Department of Science and Environment, Molecular and Medical Biology, Roskilde University, 4000 Roskilde, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark;
| |
Collapse
|
15
|
Liu X, Wang G. The Effect of High-Intensity Interval Training on Physical Parameters, Metabolomic Indexes and Serum Ficolin-3 Levels in Patients with Prediabetes and Type 2 Diabetes. Exp Clin Endocrinol Diabetes 2020; 129:740-749. [PMID: 31931532 DOI: 10.1055/a-1027-6511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Exercise benefits people with abnormal glucose metabolism, and serum ficolin-3 levels have been reported to be associated with diabetes. However, no relevant study has discussed the relationship between high-intensity interval training (HIIT) and ficolin-3 in a Chinese population. OBJECTIVE To compare the effect of HIIT and moderate-intensity continuous training (MICT) on blood pressure, glucose control, the lipid profile and the serum ficolin-3 level in patients with prediabetes and type 2 diabetes (T2D). METHODS We recruited 145 patients with prediabetes and 196 T2D patients from March to June 2018. All participants were randomly grouped into HIIT and MICT groups. HIIT consisted of progressing to twelve 1-min bouts at 90% maximal aerobic capacity (1 min recovery), and MICT consisted of progressing to 20 min at 65% maximal aerobic capacity. ˙VO2peak, body composition, blood pressure, glucose, the lipid profile and the serum ficolin-3 level were measured before and after three weeks of training. RESULTS After 3 weeks of training, participants in both the HIIT and MICT groups had significantly lower SBP, BMI, waist circumference, % body fat, and serum levels of FPG, TC, TGs, UA and ficolin-3, as well as increased vital capacity and VOmax. Additionally, the patients in the HIIT group still had significantly lower levels of 2hPG and LDL-C, regardless of prediabetes or diabetes status. After comparing the differences in the variation in parameters between the HIIT and MICT groups, we found that HIIT could help patients with prediabetes or diabetes acquire better effects of treatment in regard to anthropometry, blood pressure, glucose control, UA and ficolin-3 levels than MICT. Finally, the patients in the HIIT group had a lower rate of loss to follow-up and a higher rate of session attendance. CONCLUSIONS Both HIIT and MICT were beneficial exercise strategies for health in patients with prediabetes or T2D. However, HIIT is a more time-efficient strategy and could lower the serum level of ficolin-3 in patients after 3 weeks of training.
Collapse
Affiliation(s)
- Xiaochen Liu
- Department of Physical Education, Henan Finance University, Zhengzhou, Henan, China
| | - Gaifeng Wang
- Encephalopathy Ward, Henan Province Hospital of TCM, Zhengzhou, Henan, China
| |
Collapse
|
16
|
Skattum L. Clinical Complement Analysis-An Overview. Transfus Med Rev 2019; 33:207-216. [PMID: 31672339 DOI: 10.1016/j.tmrv.2019.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
The complement system plays an important role in varying types of disease, ranging from inflammatory and autoimmune disorders to immune deficiency states. In addition, new settings have emerged where complement analysis is of interest to monitor complement-directed therapy and aid identification of transplant complications. Therefore, it is critical that clinical laboratories offer optimized and timely complement analysis. This review presents a comprehensive overview of the most important complement analysis methods that are currently used. It also points to some areas within complement diagnostics where development is needed, for example, regarding certain analytes for which practical methods suitable for the routine laboratory are lacking. Furthermore, it contains a more detailed discussion on complement autoantibody assessment. The list of analyses providing clinically valuable information includes analysis of complement function, quantification of individual complement components and complement activation fragments, identification of autoantibodies to complement, as well as genetic complement analyses. There is still a shortage of commercially available methods suitable for high-throughput screening of complement deficiency and for assessment of complement activation, but development is under way. There is also ongoing work within the complement community to improve standardization of measurements, and recently, an extensive quality assurance program has been initiated.
Collapse
Affiliation(s)
- Lillemor Skattum
- Department of Laboratory Medicine, Section of Microbiology, Immunology and Glycobiology, Lund University, and Clinical Immunology and Transfusion Medicine, Region Skåne, Lund, Sweden.
| |
Collapse
|
17
|
Pérez-Alós L, Bayarri-Olmos R, Skjoedt MO, Garred P. Combining MAP-1:CD35 or MAP-1:CD55 fusion proteins with pattern-recognition molecules as novel targeted modulators of the complement cascade. FASEB J 2019; 33:12723-12734. [PMID: 31469600 DOI: 10.1096/fj.201901643r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dysregulation of the complement system is involved in the pathogenesis of several diseases, and its inhibition has been shown to be a feasible therapeutic option. Therefore, there is an interest in the development of complement modulators to treat complement activation-related inflammatory pathologies. Mannose-binding lectin (MBL)/ficolin/collectin-associated protein-1 (MAP-1) is a regulatory molecule of the lectin pathway (LP), whereas complement receptor 1 (CD35) and decay-accelerating factor (CD55) are membrane-anchored regulators with effects on the central effector molecule C3. In this study, we developed 2 novel soluble chimeric inhibitors by fusing MAP-1 to the 3 first domains of CD35 (CD351-3) or the 4 domains of CD55 (CD551-4) to modulate the complement cascade at 2 different stages. The constructs showed biologic properties similar to those of the parent molecules. In functional complement activation assays, the constructs were very efficient in inhibiting LP activation at the level of C3 and in the formation of terminal complement complex. This activity was enhanced when coincubated with recombinant LP recognition molecules MBL and ficolin-3. Recombinant MAP-1 fusion proteins, combined with recombinant LP recognition molecules to target sites of inflammation, represent a novel and effective therapeutic approach involving the initiation and the central and terminal effector functions of the complement cascade.-Pérez-Alós, L., Bayarri-Olmos, R., Skjoedt, M.-O., Garred, P. Combining MAP-1:CD35 or MAP-1:CD55 fusion proteins with pattern-recognition molecules as novel targeted modulators of the complement cascade.
Collapse
Affiliation(s)
- Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
18
|
Szakács D, Kocsis A, Szász R, Gál P, Pál G. Novel MASP-2 inhibitors developed via directed evolution of human TFPI1 are potent lectin pathway inhibitors. J Biol Chem 2019; 294:8227-8237. [PMID: 30952698 PMCID: PMC6527154 DOI: 10.1074/jbc.ra119.008315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/28/2019] [Indexed: 12/28/2022] Open
Abstract
The lectin pathway (LP) of the complement system is an important antimicrobial defense mechanism, but it also contributes significantly to ischemia reperfusion injury (IRI) associated with myocardial infarct, stroke, and several other clinical conditions. Mannan-binding lectin-associated serine proteinase 2 (MASP-2) is essential for LP activation, and therefore, it is a potential drug target. We have previously developed the first two generations of MASP-2 inhibitors by in vitro evolution of two unrelated canonical serine proteinase inhibitors. These inhibitors were selective LP inhibitors, but their nonhuman origin rendered them suboptimal lead molecules for drug development. Here, we present our third-generation MASP-2 inhibitors that were developed based on a human inhibitor scaffold. We subjected the second Kunitz domain of human tissue factor pathway inhibitor 1 (TFPI1 D2) to directed evolution using phage display to yield inhibitors against human and rat MASP-2. These novel TFPI1-based MASP-2 inhibitor (TFMI-2) variants are potent and selective LP inhibitors in both human and rat serum. Directed evolution of the first Kunitz domain of TFPI1 had already yielded the potent kallikrein inhibitor, Kalbitor® (ecallantide), which is an FDA-approved drug to treat acute attacks of hereditary angioedema. Like hereditary angioedema, acute IRI is also related to the uncontrolled activation of a specific plasma serine proteinase. Therefore, TFMI-2 variants are promising lead molecules for drug development against IRI.
Collapse
Affiliation(s)
- Dávid Szakács
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest
| | - Andrea Kocsis
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest
| | - Róbert Szász
- Department of Hematology, Institute of Internal Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest
| | - Gábor Pál
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest; EvolVeritas Biotechnology Ltd., Somogyi Béla u. 17, H-6600 Szentes, Hungary.
| |
Collapse
|
19
|
Larsen JB, Andersen AS, Hvas CL, Thiel S, Lassen MR, Hvas AM, Hansen AT. Lectin pathway proteins of the complement system in normotensive pregnancy and pre-eclampsia. Am J Reprod Immunol 2019; 81:e13092. [PMID: 30672631 DOI: 10.1111/aji.13092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 01/11/2019] [Indexed: 12/27/2022] Open
Abstract
PROBLEM The lectin pathway of the complement system may be involved in the pathogenesis of pre-eclampsia. We aimed to investigate changes in serum concentrations of a broad range of lectin pathway proteins during normal pregnancy and their association with pre-eclampsia, placental infarctions and intrauterine growth restriction (IUGR). METHOD OF STUDY We included 51 women with normotensive pregnancies and 54 women with pregnancies complicated by pre-eclampsia. Blood samples were obtained at gestational weeks 16, 33, 37, and after delivery for the normotensive pregnant women and before and after delivery for women with pre-eclampsia. Mannose-binding lectin (MBL), H- and M-ficolin, collectin liver-1 (CL-L1), MBL-associated serine protease (MASP)-1, MASP-2 and MASP-3 and MBL-associated proteins of 19 (MAp19) and 44 (MAp44) kDa were analysed. Clinical information was obtained from medical records. The placentae were examined by two experienced perinatal pathologists. RESULTS Lectin pathway protein concentrations generally increased during normal pregnancy and decreased after delivery in both normotensive pregnant women and women with pre-eclampsia. Exceptions were MASP-3 which increased after delivery in both groups (P < 0.0001) and H-ficolin which increased after delivery in pre-eclampsia (P < 0.0001). H-ficolin (P < 0.0001), M-ficolin (P = 0.005) and MASP-3 (P = 0.03) concentrations were lower in women with pre-eclampsia than in normotensive pregnant women. Low MASP-3 concentrations were associated with placental infarction (P = 0.03) and IUGR (P = 0.04). Low H-ficolin concentrations were associated with IUGR (P < 0.01). CONCLUSION In general, lectin pathway protein serum concentrations increased during normal pregnancy. H-ficolin and MASP-3 may be involved in the pathophysiology of pre-eclampsia and IUGR and could be potential future pre-eclampsia biomarkers.
Collapse
Affiliation(s)
| | | | | | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anette Tarp Hansen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
20
|
Jalal PJ, Urbanowicz RA, Horncastle E, Pathak M, Goddard C, Saeed A, Mason CP, Ball JK, Irving WL, McClure CP, King BJ, Tarr AW. Expression of human ficolin-2 in hepatocytes confers resistance to infection by diverse hepatotropic viruses. J Med Microbiol 2019; 68:642-648. [PMID: 30747617 DOI: 10.1099/jmm.0.000935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The liver-expressed pattern recognition receptors mannose-binding lectin (MBL), ficolin-2 and ficolin-3 contribute to the innate immune response by activating complement. Binding of soluble ficolin-2 to viral pathogens can directly neutralize virus entry. We observed that the human hepatoma cell line HuH7.5, which is routinely used for the study of hepatotropic viruses, is deficient in expression of MBL, ficolin-2 and ficolin-3. We generated a cell line that expressed and secreted ficolin-2. This cell line (HuH7.5 [FCN2]) was more resistant to infection with hepatitis C virus (HCV), ebolavirus and vesicular stomatitis virus, but surprisingly was more susceptible to infection with rabies virus. Cell-to-cell spread of HCV was also inhibited in ficolin-2 expressing cells. This illustrates that ficolin-2 expression in hepatocytes contributes to innate resistance to virus infection, but some viruses might utilize ficolin-2 to facilitate entry.
Collapse
Affiliation(s)
- Paywast J Jalal
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Biology Department, Faculty of Science, University of Sulaimani, Sulaimani, Iraq
| | - Richard A Urbanowicz
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Emma Horncastle
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Monika Pathak
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Chun Goddard
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Amanj Saeed
- Biology Department, Faculty of Science, University of Sulaimani, Sulaimani, Iraq
| | - Christopher P Mason
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Jonathan K Ball
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - William L Irving
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - C Patrick McClure
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Barnabas J King
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Alexander W Tarr
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| |
Collapse
|
21
|
Eriksson O, Chiu J, Hogg PJ, Atkinson JP, Liszewski MK, Flaumenhaft R, Furie B. Thiol isomerase ERp57 targets and modulates the lectin pathway of complement activation. J Biol Chem 2019; 294:4878-4888. [PMID: 30670593 DOI: 10.1074/jbc.ra118.006792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/13/2019] [Indexed: 11/06/2022] Open
Abstract
ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.
Collapse
Affiliation(s)
- Oskar Eriksson
- From the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115
| | - Joyce Chiu
- the Centenary Institute, National Health and Medical Research Council Clinical Trials Centre, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia, and
| | - Philip J Hogg
- the Centenary Institute, National Health and Medical Research Council Clinical Trials Centre, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia, and
| | - John P Atkinson
- the Department of Medicine/Rheumatology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - M Kathryn Liszewski
- the Department of Medicine/Rheumatology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Robert Flaumenhaft
- From the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115
| | - Bruce Furie
- From the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115,
| |
Collapse
|
22
|
Eppa Ł, Pągowska-Klimek I, Świerzko AS, Moll M, Krajewski WR, Cedzyński M. Deposition of mannose-binding lectin and ficolins and activation of the lectin pathway of complement on the surface of polyurethane tubing used for cardiopulmonary bypass. J Biomed Mater Res B Appl Biomater 2018; 106:1202-1208. [PMID: 28561998 DOI: 10.1002/jbm.b.33933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/05/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2023]
Abstract
The artificial surface used for cardiopulmonary bypass (CPB) is a crucial factor activating the complement system and thus contributing to the generation of a systemic inflammatory response. The activation of classical and alternative pathways on this artificial surface is well known. In contrast, lectin pathway (LP) activation has not been fully investigated, although noted during CPB in several studies. Moreover, we have recently proved the contribution of the LP to the generation of the systemic inflammatory response syndrome after pediatric cardiac surgery. The aim of this study was to assess LP-mediated complement activation on the surface of polyurethane CPB circuit tubing (noncoated Chalice ® ), used for CPB procedures in children with congenital heart disease. We found deposition of mannose-binding lectin, ficolin-1, -2, and -3 on the surface of unused tubing and on tubing used for CPB from a small minority of patients. Furthermore, we observed deposition of complement C4 activation products on tubing used for CPB and previously unused tubing after incubation with normal serum. The latter finding indicates LP activation in vitro on the polyurethane surface. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1202-1208, 2018.
Collapse
Affiliation(s)
- Łukasz Eppa
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Izabela Pągowska-Klimek
- Department of Anesthesiology and Intensive Care, Polish Mother's Memorial Hospital Research Institute, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Anna S Świerzko
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Maciej Moll
- Department of Cardiac Surgery, Polish Mother's Memorial Hospital Research Institute, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Wojciech R Krajewski
- Department of Anesthesiology and Intensive Care, Polish Mother's Memorial Hospital Research Institute, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Maciej Cedzyński
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| |
Collapse
|
23
|
Genster N, Østrup O, Schjalm C, Eirik Mollnes T, Cowland JB, Garred P. Ficolins do not alter host immune responses to lipopolysaccharide-induced inflammation in vivo. Sci Rep 2017; 7:3852. [PMID: 28634324 PMCID: PMC5478672 DOI: 10.1038/s41598-017-04121-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/09/2017] [Indexed: 11/17/2022] Open
Abstract
Ficolins are a family of pattern recognition molecules that are capable of activating the lectin pathway of complement. A limited number of reports have demonstrated a protective role of ficolins in animal models of infection. In addition, an immune modulatory role of ficolins has been suggested. Yet, the contribution of ficolins to inflammatory disease processes remains elusive. To address this, we investigated ficolin deficient mice during a lipopolysaccharide (LPS)-induced model of systemic inflammation. Although murine serum ficolin was shown to bind LPS in vitro, there was no difference between wildtype and ficolin deficient mice in morbidity and mortality by LPS-induced inflammation. Moreover, there was no difference between wildtype and ficolin deficient mice in the inflammatory cytokine profiles after LPS challenge. These findings were substantiated by microarray analysis revealing an unaltered spleen transcriptome profile in ficolin deficient mice compared to wildtype mice. Collectively, results from this study demonstrate that ficolins are not involved in host response to LPS-induced systemic inflammation.
Collapse
Affiliation(s)
- Ninette Genster
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Olga Østrup
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Camilla Schjalm
- Department of Immunology, Oslo University Hospital, Rikshospitalet, 0424, Oslo, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, 0424, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, and K.J. Jebsen TREC, University of Tromsø, Tromsø, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jack B Cowland
- The Granulocyte Research Laboratory, Department of Hematology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
24
|
Horváth Z, Csuka D, Vargova K, Leé S, Varga L, Garred P, Préda I, Zsámboki ET, Prohászka Z, Kiss RG. Association of Low Ficolin-Lectin Pathway Parameters with Cardiac Syndrome X. Scand J Immunol 2016; 84:174-81. [PMID: 27312152 DOI: 10.1111/sji.12454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022]
Abstract
In patients with typical angina pectoris, inducible myocardial ischaemia and macroscopically normal coronaries (cardiac syndrome X (CSX)), a significantly elevated plasma level of terminal complement complex (TCC), the common end product of complement activation, has been observed without accompanying activation of the classical or the alternative pathways. Therefore, our aim was to clarify the role of the ficolin-lectin pathway in CSX. Eighteen patients with CSX, 37 stable angina patients with significant coronary stenosis (CHD) and 54 healthy volunteers (HC) were enrolled. Serum levels of ficolin-2 and ficolin-3, ficolin-3/MASP-2 complex and ficolin-3-mediated TCC deposition (FCN3-TCC) were determined. Plasma level of TCC was significantly higher in the CSX than in the HC or CHD group (5.45 versus 1.30 versus 2.04 AU/ml, P < 0.001). Serum levels of ficolin-2 and ficolin-3 were significantly lower in the CSX compared to the HC or CHD group (3.60 versus 5.80 or 5.20 μg/ml, P < 0.05; 17.80 versus 24.10 or 26.80 μg/ml, P < 0.05). The ficolin-3/MASP-2 complex was significantly lower in the CSX group compared to the HC group (92.90 versus 144.90 AU/ml, P = 0.006). FCN3-TCC deposition was significantly lower in the CSX group compared to the HC and CHD groups (67.8% versus 143.3% or 159.7%, P < 0.05). In the CSX group, a significant correlation was found between TCC and FCN3-TCC level (r = 0.507, P = 0.032) and between ficolin-3/MASP-2 complex level and FCN3-TCC deposition (r = 0.651, P = 0.003). In conclusion, in patients with typical angina and myocardial ischaemia despite macroscopically normal coronary arteries, low levels of several lectin pathway parameters were observed, indicating complement activation and consumption. Complement activation through the ficolin-lectin pathway might play a role in the complex pathomechanism of CSX.
Collapse
Affiliation(s)
- Z Horváth
- Research Group for Inflammation Biology and Immunogenomics of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| | - D Csuka
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - K Vargova
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| | - S Leé
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| | - L Varga
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - P Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - I Préda
- Research Group for Inflammation Biology and Immunogenomics of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| | - E T Zsámboki
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| | - Z Prohászka
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - R G Kiss
- Research Group for Inflammation Biology and Immunogenomics of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
- Department of Cardiology, Hungarian Defence Forces Medical Centre, Budapest, Hungary
| |
Collapse
|
25
|
Hansen MB, Rasmussen LS, Pilely K, Hellemann D, Hein E, Madsen MB, Hyldegaard O, Garred P. The Lectin Complement Pathway in Patients with Necrotizing Soft Tissue Infection. J Innate Immun 2016; 8:507-16. [PMID: 27355483 PMCID: PMC6738884 DOI: 10.1159/000447327] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mannose-binding lectin (MBL) and ficolins are pattern recognition molecules (PRMs) that play an important role during infection through activation of the lectin complement pathway. We assessed whether plasma PRM levels were associated with mortality in patients with necrotizing soft tissue infection (NSTI). METHODS We conducted a prospective, observational study over 25 months involving 135 NSTI patients with a maximum follow-up of 2.7 years. Blood samples were taken upon admission. Non-infected patients served as controls. RESULTS PRM levels were significantly lower compared with controls. A baseline Ficolin-2 level below the median was associated with mortality at the end of follow-up (p = 0.007). No significant association was found for MBL, Ficolin-1 and Ficolin-3. A Ficolin-2 level below the median had a negative predictive value of 0.94 for 28-day mortality, and a level below the optimal cut-off was independently associated with 28-day mortality when adjusted for age, sex and chronicity [hazard ratio 6.27 (95% confidence interval 2.28-17.21), p < 0.0001], also when Simplified Acute Physiology Score II was included in the analysis [hazard ratio 3.16 (95% confidence interval 1.03-9.73), p = 0.045]. CONCLUSIONS All PRMs were significantly lower in patients with NSTI than in controls. Only baseline Ficolin-2 was associated with short- and long-term mortality. A high baseline Ficolin-2 level indicated a 94% chance of surviving the first 28 days after admission.
Collapse
Affiliation(s)
- Marco B. Hansen
- Department of Anesthesia, Center of Head and Orthopedics, Slagelse, Denmark
- Hyperbaric Unit, Department of Anesthesia, Center of Head and Orthopedics, Slagelse, Denmark
| | - Lars S. Rasmussen
- Department of Anesthesia, Center of Head and Orthopedics, Slagelse, Denmark
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Slagelse, Denmark
| | - Dorthe Hellemann
- Department of Anesthesia and Intensive Care, Slagelse Hospital, Slagelse, Denmark
| | - Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Slagelse, Denmark
| | - Martin B. Madsen
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, Copenhagen, Slagelse, Denmark
| | - Ole Hyldegaard
- Department of Anesthesia, Center of Head and Orthopedics, Slagelse, Denmark
- Hyperbaric Unit, Department of Anesthesia, Center of Head and Orthopedics, Slagelse, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Slagelse, Denmark
| |
Collapse
|
26
|
Pilely K, Rosbjerg A, Genster N, Gal P, Pál G, Halvorsen B, Holm S, Aukrust P, Bakke SS, Sporsheim B, Nervik I, Niyonzima N, Bartels ED, Stahl GL, Mollnes TE, Espevik T, Garred P. Cholesterol Crystals Activate the Lectin Complement Pathway via Ficolin-2 and Mannose-Binding Lectin: Implications for the Progression of Atherosclerosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:5064-74. [PMID: 27183610 DOI: 10.4049/jimmunol.1502595] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/13/2016] [Indexed: 12/16/2023]
Abstract
Cholesterol crystals (CC) play an essential role in the formation of atherosclerotic plaques. CC activate the classical and the alternative complement pathways, but the role of the lectin pathway is unknown. We hypothesized that the pattern recognition molecules (PRMs) from the lectin pathway bind CC and function as an upstream innate inflammatory signal in the pathophysiology of atherosclerosis. We investigated the binding of the PRMs mannose-binding lectin (MBL), ficolin-1, ficolin-2, and ficolin-3, the associated serine proteases, and complement activation products to CC in vitro using recombinant proteins, specific inhibitors, as well as deficient and normal sera. Additionally, we examined the deposition of ficolin-2 and MBL in human carotid plaques by immunohistochemistry and fluorescence microscopy. The results showed that the lectin pathway was activated on CC by binding of ficolin-2 and MBL in vitro, resulting in activation and deposition of complement activation products. MBL bound to CC in a calcium-dependent manner whereas ficolin-2 binding was calcium-independent. No binding was observed for ficolin-1 or ficolin-3. MBL and ficolin-2 were present in human carotid plaques, and binding of MBL to CC was confirmed in vivo by immunohistochemistry, showing localization of MBL around CC clefts. Moreover, we demonstrated that IgM, but not IgG, bound to CC in vitro and that C1q binding was facilitated by IgM. In conclusion, our study demonstrates that PRMs from the lectin pathway recognize CC and provides evidence for an important role for this pathway in the inflammatory response induced by CC in the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Ninette Genster
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Peter Gal
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, 1113 Budapest, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; K.G. Jebsen Inflammation Research Center, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Hospital for Rheumatic Diseases, 2609 Lillehammer, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; K.G. Jebsen Inflammation Research Center, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Siril Skaret Bakke
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Bjørnar Sporsheim
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Ingunn Nervik
- Section for Children's and Women's Health, Department of Laboratory Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Nathalie Niyonzima
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Emil D Bartels
- Department of Clinical Biochemistry, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen O, Denmark
| | - Gregory L Stahl
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Tom Eirik Mollnes
- K.G. Jebsen Inflammation Research Center, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Laboratory, Nordland Hospital, 8038 Bodø, Norway; and K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9019 Tromsø, Norway
| | - Terje Espevik
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen O, Denmark;
| |
Collapse
|
27
|
Man-Kupisinska A, Michalski M, Maciejewska A, Swierzko AS, Cedzynski M, Lugowski C, Lukasiewicz J. A New Ligand-Based Method for Purifying Active Human Plasma-Derived Ficolin-3 Complexes Supports the Phenomenon of Crosstalk between Pattern-Recognition Molecules and Immunoglobulins. PLoS One 2016; 11:e0156691. [PMID: 27232184 PMCID: PMC4883783 DOI: 10.1371/journal.pone.0156691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/18/2016] [Indexed: 11/18/2022] Open
Abstract
Despite recombinant protein technology development, proteins isolated from natural sources remain important for structure and activity determination. Ficolins represent a class of proteins that are difficult to isolate. To date, three methods for purifying ficolin-3 from plasma/serum have been proposed, defined by most critical step: (i) hydroxyapatite absorption chromatography (ii) N-acetylated human serum albumin affinity chromatography and (iii) anti-ficolin-3 monoclonal antibody-based affinity chromatography. We present a new protocol for purifying ficolin-3 complexes from human plasma that is based on an exclusive ligand: the O-specific polysaccharide of Hafnia alvei PCM 1200 LPS (O-PS 1200). The protocol includes (i) poly(ethylene glycol) precipitation; (ii) yeast and l-fucose incubation, for depletion of mannose-binding lectin; (iii) affinity chromatography using O-PS 1200-Sepharose; (iv) size-exclusion chromatography. Application of this protocol yielded average 2.2 mg of ficolin-3 preparation free of mannose-binding lectin (MBL), ficolin-1 and -2 from 500 ml of plasma. The protein was complexed with MBL-associated serine proteases (MASPs) and was able to activate the complement in vitro. In-process monitoring of MBL, ficolins, and total protein content revealed the presence of difficult-to-remove immunoglobulin G, M and A, in some extent in agreement with recent findings suggesting crosstalk between IgG and ficolin-3. We demonstrated that recombinant ficolin-3 interacts with IgG and IgM in a concentration-dependent manner. Although this association does not appear to influence ficolin-3-ligand interactions in vitro, it may have numerous consequences in vivo. Thus our purification procedure provides Ig-ficolin-3/MASP complexes that might be useful for gaining further insight into the crosstalk and biological activity of ficolin-3.
Collapse
Affiliation(s)
- Aleksandra Man-Kupisinska
- Department of Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Mateusz Michalski
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- Institute of Microbiology, Immunology and Biotechnology, University of Lodz, Lodz, Poland
| | - Anna Maciejewska
- Department of Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Anna S. Swierzko
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Maciej Cedzynski
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Czeslaw Lugowski
- Department of Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Jolanta Lukasiewicz
- Department of Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
28
|
Zhang X, Hu Y, Shen J, Zeng H, Lu J, Li L, Bao Y, Liu F, Jia W. Low levels of ficolin-3 are associated with diabetic peripheral neuropathy. Acta Diabetol 2016; 53:295-302. [PMID: 26116288 DOI: 10.1007/s00592-015-0780-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/26/2015] [Indexed: 01/08/2023]
Abstract
AIMS Ficolin-3, a soluble molecule of the innate immune system, has a primary role in the activation of the lectin pathway in the complement system. Considering that inflammatory mechanisms are involved in complement activation and take part in the pathophysiology of diabetic peripheral neuropathy (DPN), we conducted this study to explore the link between serum ficolin-3 and DPN in diabetic patients. METHODS A total of 466 diabetic patients were enrolled in this cross-sectional study. DPN was evaluated by neurological symptoms, neurological signs, neurothesiometer and electromyogram. The concentration of serum ficolin-3 was determined by enzyme-linked immunosorbent assay. RESULTS The concentration of serum ficolin-3 was lower in DPN patients compared with non-DPN patients (18.73 ± 4.75 vs. 26.69 ± 5.68 ng/mL, P < 0.001). In addition, it was found negatively correlated to the vibration perception threshold (r = -0.158; P = 0.025). The results of multiple regression analysis of DPN indicated that age, diabetes duration, serum ficolin-3 were all independent impact factors for DPN (P < 0.05). Patients were then assigned into quartiles according to the serum ficolin-3 levels, and the prevalence of DPN ascended as the concentration of ficolin-3 descended (Trend analysis, P < 0.001). Compared with ficolin-3 Quartile 1 (referent), the risk of DPN was significantly greater in Quartile 2 (OR 2.76; 95 % CI 1.56-4.88; P < 0.001), Quartile 3 (OR 3.02; 95 % CI 1.69-5.40; P < 0.001) and Quartile 4 (OR 6.84; 95 % CI 3.39-13.80; P < 0.001). CONCLUSIONS Lower ficolin-3 level is independently associated with DPN, and it may be a potential biomarker for DPN.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Yanyun Hu
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Jing Shen
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Hui Zeng
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Junxi Lu
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Lianxi Li
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China.
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute of Diabetes, Shanghai Key Laboratory of Diabetes, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
29
|
Kozarcanin H, Lood C, Munthe-Fog L, Sandholm K, Hamad OA, Bengtsson AA, Skjoedt MO, Huber-Lang M, Garred P, Ekdahl KN, Nilsson B. The lectin complement pathway serine proteases (MASPs) represent a possible crossroad between the coagulation and complement systems in thromboinflammation. J Thromb Haemost 2016; 14:531-45. [PMID: 26614707 DOI: 10.1111/jth.13208] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Indexed: 12/26/2022]
Abstract
UNLABELLED ESSENTIALS: The lectin pathway's MASP-1/2 activates coagulation factors but the trigger of the activation is unknown. MASP-1/2 activation was assessed by quantifying complexes between MASPs and antithrombin/C1-inhibitor. Activated platelets and fibrin were demonstrated to activate MASP-1 and MASP-2 both in vitro and in vivo. These findings may represent a crossroad between the complement and the coagulation systems. SUMMARY BACKGROUND The activated forms of the complement lectin pathway (LP) proteases MASP-1 and MASP-2 are able to cleave the coagulation factors prothrombin, fibrinogen, factor XIII and thrombin-activatable fibrinolysis inhibitor in vitro. In vivo studies also show that MASP-1 is involved in thrombogenesis. OBJECTIVES To clarify the not yet identified mechanisms involved in triggering activation of the LP during thrombotic reactions. METHODS Novel sandwich-ELISAs for detection of complexes between MASP-1 or MASP-2 and the serpins C1 inhibitor (C1-INH) or antithrombin (AT), were used to specifically detect and quantify the activated forms of MASP-1 and MASP-2. RESULTS Activated platelets were shown by flow cytometry to bind Ficolin-1, -2 and -3 but not MBL, which was associated with activation of MASP-1 and MASP-2. We also demonstrated that fibrin and the plasmin-generated fibrin fragment DD in plasma, bind and activate MASP-1 and MASP-2. As demonstrated by the ELISA and SDS-PAGE/Western blotting, the fibrin-associated activation was reflected in a specific inactivation by AT during clotting without the assistance of heparin. In all other cases the MASPs were, as previously reported, inactivated by C1-INH. In systemic lupus erythematosus patients with thrombotic disease and in polytrauma patients, the levels of activated MASP-1 and MASP-2 in complex with both AT and C1-INH were associated with markers of thrombotic disease and contact/coagulation system activation. CONCLUSIONS MASP-1 and MASP-2 are activated during blood clotting. This activation is triggered by activated platelets and by the generation of fibrin during thrombotic reactions in vitro and in vivo, and may represent a novel activation/amplification mechanism in thromboinflammation.
Collapse
Affiliation(s)
- H Kozarcanin
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - C Lood
- Section of Rheumatology, Department of Clinical Sciences Lund, Skåne University Hospital and Lund University, Lund, Sweden
| | - L Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - K Sandholm
- Linnaeus Center for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - O A Hamad
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - A A Bengtsson
- Section of Rheumatology, Department of Clinical Sciences Lund, Skåne University Hospital and Lund University, Lund, Sweden
| | - M-O Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Huber-Lang
- Department of Traumatology, Hand, Plastic, Reconstructive Surgery, University Hospital of Ulm, Ulm, Germany
| | - P Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - K N Ekdahl
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
- Linnaeus Center for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - B Nilsson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| |
Collapse
|
30
|
Pan JW, Gao XW, Jiang H, Li YF, Xiao F, Zhan RY. Low serum ficolin-3 levels are associated with severity and poor outcome in traumatic brain injury. J Neuroinflammation 2015; 12:226. [PMID: 26627059 PMCID: PMC4666053 DOI: 10.1186/s12974-015-0444-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/23/2015] [Indexed: 11/24/2022] Open
Abstract
Background Ficolin-mediated activation of the lectin pathway of complement contributes to the complement-independent inflammatory processes of traumatic brain injury. Lower serum ficolin-3 levels have been demonstrated to be highly associated with unfavorable outcome after ischemic stroke. This prospective observatory study was designed to investigate the relationships between serum ficolin-3 levels and injury severity and clinical outcomes after severe traumatic brain injury. Methods Serum ficolin-3 levels of 128 patients and 128 healthy controls were measured by sandwich immunoassays. An unfavorable outcome was defined as Glasgow Outcome Scale score of 1–3. Study endpoints included mortality at 1 week and 6 months and unfavorable outcome at 6 months after head trauma. Injury severity was assessed by Glasgow Coma Scale score. Multivariate logistic models were structured to evaluate the relationships between serum ficolin-3 levels and study endpoints and injury severity. Results Compared with the healthy controls, serum ficolin-3 levels on admission were statistically decreased in patients with severe traumatic brain injury. Serum ficolin-3 levels were independently correlated with Glasgow Coma Scale scores. Ficolin-3 was also identified as an independent prognostic predictor for 1-week mortality, 6-month mortality, and 6-month unfavorable outcome. Under receiver operating characteristics curves, ficolin-3 has similar prognostic predictive values for all study endpoints compared with Glasgow Coma Scale scores. Conclusions It was proposed that lower serum ficolin-3 levels, correlated with injury severity, had the potential to be the useful, complementary tool to predict short- or long-term clinical outcomes after severe traumatic brain injury.
Collapse
Affiliation(s)
- Jian-Wei Pan
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.
| | - Xiong-Wei Gao
- Department of Neurosurgery, Sanmen People's Hospital, 171 Renmin Road, Sanmen, 317100, People's Republic of China.
| | - Hao Jiang
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.
| | - Ya-Feng Li
- Department of Neurosurgery, Sanmen People's Hospital, 171 Renmin Road, Sanmen, 317100, People's Republic of China.
| | - Feng Xiao
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.
| | - Ren-Ya Zhan
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
31
|
Hansen CB, Csuka D, Munthe-Fog L, Varga L, Farkas H, Hansen KM, Koch C, Skjødt K, Garred P, Skjoedt MO. The Levels of the Lectin Pathway Serine Protease MASP-1 and Its Complex Formation with C1 Inhibitor Are Linked to the Severity of Hereditary Angioedema. THE JOURNAL OF IMMUNOLOGY 2015; 195:3596-604. [DOI: 10.4049/jimmunol.1402838] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
|
32
|
Nordmaj MA, Munthe-Fog L, Hein E, Skjoedt MO, Garred P. Genetically engineered fusion of MAP-1 and factor H domains 1-5 generates a potent dual upstream inhibitor of both the lectin and alternative complement pathways. FASEB J 2015; 29:4945-55. [PMID: 26260032 DOI: 10.1096/fj.15-277103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/03/2015] [Indexed: 01/01/2023]
Abstract
Inhibition of the complement cascade has emerged as an option for treatment of a range of diseases. Mannose-binding lectin/ficolin/collectin-associated protein (MAP-1) is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway. The central regulator of the alternative pathway (AP) is complement factor H (FH). Our aim was to design a dual upstream inhibitor of both human lectin and APs by fusing MAP-1 with a part of FH. There were 2 different recombinant chimeric proteins comprising full-length human MAP-1 and the first 5 N-terminal domains of human FH designed. The FH domains were orientated either in the N- or C-terminal part of MAP-1. The complement inhibition potential in human serum was assessed. Both chimeric constructs displayed the characteristics of the native molecules and bound to the PRMs with an EC50 of ∼ 2 nM. However, when added to serum diluted 1:4 in a solid-phase functional assay, only the first 5 N-terminal domains of complement FH fused to the C-terminal part of full-length MAP-1 chimeric construct were able to combine inhibition of lectin and AP activation with an half maximal inhibitory concentration of ∼ 100 and 20 nM, respectively. No effect was seen on the classical pathway. Fusion of MAP-1 with FH domains represents a novel therapeutic approach for selective targeting upstream and central complement activation at sites of inflammation.
Collapse
Affiliation(s)
- Mie Anemone Nordmaj
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Jusko M, Potempa J, Mizgalska D, Bielecka E, Ksiazek M, Riesbeck K, Garred P, Eick S, Blom AM. A Metalloproteinase Mirolysin of Tannerella forsythia Inhibits All Pathways of the Complement System. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26209620 DOI: 10.4049/jimmunol.1402892] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Recent reports focusing on virulence factors of periodontal pathogens implicated proteinases as major determinants of remarkable pathogenicity of these species, with special emphasis on their capacity to modulate complement activity. In particular, bacteria-mediated cleavage of C5 and subsequent release of C5a seems to be an important phenomenon in the manipulation of the local inflammatory response in periodontitis. In this study, we present mirolysin, a novel metalloproteinase secreted by Tannerella forsythia, a well-recognized pathogen strongly associated with periodontitis. Mirolysin exhibited a strong effect on all complement pathways. It inhibited the classical and lectin complement pathways due to efficient degradation of mannose-binding lectin, ficolin-2, ficolin-3, and C4, whereas inhibition of the alternative pathway was caused by degradation of C5. This specificity toward complement largely resembled the activity of a previously characterized metalloproteinase of T. forsythia, karilysin. Interestingly, mirolysin released the biologically active C5a peptide in human plasma and induced migration of neutrophils. Importantly, we demonstrated that combination of mirolysin with karilysin, as well as a cysteine proteinase of another periodontal pathogen, Prevotella intermedia, resulted in a strong synergistic effect on complement. Furthermore, mutant strains of T. forsythia, devoid of either mirolysin or karilysin, showed diminished survival in human serum, providing further evidence for the synergistic inactivation of complement by these metalloproteinases. Taken together, our findings on interactions of mirolysin with complement significantly add to the understanding of immune evasion strategies of T. forsythia and expand the knowledge on molecular mechanisms driving pathogenic events in the infected periodontium.
Collapse
Affiliation(s)
- Monika Jusko
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 205 02 Malmö, Sweden
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30 387 Krakow, Poland; Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202
| | - Danuta Mizgalska
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30 387 Krakow, Poland
| | - Ewa Bielecka
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 205 02 Malmö, Sweden; Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30 387 Krakow, Poland
| | - Miroslaw Ksiazek
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30 387 Krakow, Poland
| | - Kristian Riesbeck
- Division of Clinical Microbiology, Department of Translational Medicine, Lund University, 205 02 Malmö, Sweden
| | - Peter Garred
- Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; and
| | - Sigrun Eick
- Department of Periodontology, Laboratory of Oral Microbiology, University of Bern, 3010 Bern, Switzerland
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 205 02 Malmö, Sweden;
| |
Collapse
|
34
|
Hein E, Munthe-Fog L, Thiara AS, Fiane AE, Mollnes TE, Garred P. Heparin-coated cardiopulmonary bypass circuits selectively deplete the pattern recognition molecule ficolin-2 of the lectin complement pathway in vivo. Clin Exp Immunol 2015; 179:294-9. [PMID: 25174443 PMCID: PMC4298406 DOI: 10.1111/cei.12446] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2014] [Indexed: 12/28/2022] Open
Abstract
The complement system can be activated via the lectin pathway by the recognition molecules mannose-binding lectin (MBL) and the ficolins. Ficolin-2 exhibits binding against a broad range of ligands, including biomaterials in vitro, and low ficolin-2 levels are associated with increased risk of infections. Thus, we investigated the biocompatibility of the recognition molecules of the lectin pathway in two different types of cardiopulmonary bypass circuits. Bloods were drawn at five time-points before, during and postoperatively from 30 patients undergoing elective cardiac surgery. Patients were randomized into two groups using different coatings of cardiopulmonary bypass circuits, Phisio® (phosphorylcholine polymer coating) and Bioline® (albumin-heparin coating). Concentrations of MBL, ficolin-1, -2 and -3 and soluble C3a and terminal complement complex (TCC) in plasma samples were measured. Ficolin-3-mediated complement activation potential was evaluated with C4, C3 and TCC as output. There was no significant difference between the two circuit materials regarding MBL, ficolin-1 and -3. In the Bioline® group the ficolin-2 levels decreased significantly after initiation of surgery (P < 0.0001) and remained reduced throughout the sampling period. This was not seen for Phisio®-coated circuits. Ficolin-3-mediated complement activation potential was reduced significantly in both groups after start of operation (P < 0.0001), whereas soluble C3a and TCC in the samples were increased (P < 0.0001). Ficolin-2 was depleted from plasma during cardiac surgery when using heparin-coated bypass circuits and did not reach baseline level 24 h postoperation. These findings may have implications for the postoperative susceptibility to infections in patients undergoing extracorporeal circulation procedures.
Collapse
Affiliation(s)
- E Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of CopenhagenCopenhagen, Norway
| | - L Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of CopenhagenCopenhagen, Norway
| | - A S Thiara
- Department of Cardiothoracic Surgery, Oslo University HospitalOslo, Norway
| | - A E Fiane
- Department of Cardiothoracic Surgery, Oslo University HospitalOslo, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and TechnologyTrondheim, Norway
| | - T E Mollnes
- Department of Immunology, Oslo University Hospital Rikshospitalet, K.G.Jebsen IRC, University of OsloOslo, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and TechnologyTrondheim, Norway
- Research Laboratory, Nordland Hospital, Bodø, and Faculty of Health Sciences, K.G. Jebsen TREC, University of TromsøTromsø, Norway
| | - P Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of CopenhagenCopenhagen, Norway
| |
Collapse
|
35
|
Hein E, Nielsen LA, Nielsen CT, Munthe-Fog L, Skjoedt MO, Jacobsen S, Garred P. Ficolins and the lectin pathway of complement in patients with systemic lupus erythematosus. Mol Immunol 2015; 63:209-14. [PMID: 25069872 DOI: 10.1016/j.molimm.2014.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 12/15/2022]
Abstract
The complement system plays a pathophysiological role in systemic lupus erythematosus (SLE). This study aims to investigate whether an association exists between the ficolins that are part of the lectin complement pathway and SLE. EDTA plasma samples from 68 Danish SLE patients and 29 healthy donors were included in the study. Plasma concentrations of Ficolin-1, -2, and -3 were determined in specific sandwich ELISAs. Lectin pathway activity via Ficolin-3 was measured in ELISA on acetylated bovine serum albumin (acBSA) and measured as Ficolin-3 binding and deposition of C4, C3 and the terminal complement complex (TCC). SLE patients had increased levels of Ficolin-3, 21.6μg/ml as compared to 17.0μg/ml in healthy controls (P=0.0098). The Ficolin-1 plasma concentration was negatively correlated with SLE Disease Activity Index (SLEDAI) (Rho=-0.29, P=0.015) and positively correlated to the [Systemic Lupus International Collaborating Clinics (SLICC)/American College of Rheumatology (ACR) Damage Index] (SDI) (Rho=0.27, P=0.026). The Ficolin-1 concentration was also associated with the occurrence of arterial (P=0.0053) but not venous thrombosis (P=0.42). Finally, deposition of C4, C3 and TCC in the Ficolin-3 pathway were all correlated to SLEDAI, respectively (P<0.0076). The Ficolin-1 association to SLEDAI and SDI as well as arterial thrombosis shown in this study suggests that Ficolin-1 may be a potential new biomarker for patients with SLE. Furthermore, Ficolin-3 mediated complement activation may be valuable in monitoring disease activity in SLE patients due to the high sensitivity for complement consumption in the assay independent of the Ficolin-3 concentration.
Collapse
Affiliation(s)
- Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louise Aas Nielsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer T Nielsen
- Department of Clinical Biochemistry, Statens Serum Institut, Copenhagen, Denmark; Department of Infectious Diseases and Rheumatology, Rigshospitalet, Copenhagen, Denmark
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Jacobsen
- Department of Infectious Diseases and Rheumatology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Hein E, Garred P. The Lectin Pathway of Complement and Biocompatibility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 865:77-92. [PMID: 26306444 DOI: 10.1007/978-3-319-18603-0_5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In modern health technologies the use of biomaterials in the form of stents, haemodialysis tubes, artificial implants, bypass circuits etc. is rapidly expanding. The exposure of synthetic, foreign surfaces to the blood and tissue of the host, calls for strict biocompatibility in respect to contact activation, the coagulation system and the complement system. The complement system is an important part of the initial immune response and consists of fluid phase molecules in the blood stream. Three different activation pathways can initiate the complement system, the lectin, the classical and the alternative pathway, all converging in an amplification loop of the cascade system and downstream reactions. Thus, when exposed to foreign substances complement components will be activated and lead to a powerful inflammatory response. Biosurface induced complement activation is a recognised issue that has been broadly documented. However, the specific role of lectin pathway and the pattern recognition molecules initiating the pathway has only been transiently investigated. Here we review the current data on the field.
Collapse
Affiliation(s)
- Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen O, Denmark
| | | |
Collapse
|
37
|
Csuka D, Munthe-Fog L, Hein E, Zotter Z, Prohászka Z, Farkas H, Varga L, Garred P. Activation of the ficolin-lectin pathway during attacks of hereditary angioedema. J Allergy Clin Immunol 2014; 134:1388-1393.e1. [PMID: 25042985 DOI: 10.1016/j.jaci.2014.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 04/26/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND The activation of plasma enzyme systems is insufficiently controlled in hereditary angioedema due to the deficiency of C1-inhibitor (C1-INH) (HAE-C1-INH). Recently, it was suggested that the ficolin-lectin pathway (ficolin-LP) might play a more dominant role than the mannose-binding lectin-lectin pathway in the pathomechanism of HAE-C1-INH. OBJECTIVE Because the role of the ficolin-LP during edematous attacks is still enigmatic, we analyzed its activity during such episodes. METHODS Thirty-five patients with HAE-C1-INH, who have experienced severe attacks on 106 occasions, were enrolled. We analyzed blood samples drawn during attacks, and obtained 35 samples from the same patients during symptom-free periods. The serum levels of ficolin-2, ficolin-3, MASP-2, ficolin-3/MASP-2 complex, C1-INH, and C4, as well as the extent of ficolin-3-mediated terminal complement complex (FCN3-TCC) deposition, were measured using ELISA-based methods. RESULTS Levels of MASP-2 and of the ficolin-3/MASP-2 complex were elevated (P < .0001 and .033, respectively), whereas that of FCN3-TCC was lower (P < .0001) during attacks than during the symptom-free period. During symptom-free periods, FCN3-TCC deposition was significantly related to concentrations of ficolin-3 (R = 0.2778; P = .0022), antigenic C1-INH (R = 0.3152; P = .0006), and C4 (R = 0.5307; P < .0001). Both ficolin-3 and MASP-2 levels correlated inversely with the time from the onset of the attack until blood sampling. CONCLUSIONS There is a marked heterogeneity of the pathomechanism and development of hereditary angioedema attacks in different patients. Our results suggest that the activation of the ficolin-LP may deplete the innately low level of C1-INH and thus, it may contribute to the uncontrolled activation of plasma cascade systems, and thereby to edema formation.
Collapse
Affiliation(s)
- Dorottya Csuka
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary; Laboratory of Molecular Medicine, Faculty of Health Sciences, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Faculty of Health Sciences, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Hein
- Laboratory of Molecular Medicine, Faculty of Health Sciences, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Zsuzsanna Zotter
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltán Prohászka
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Henriette Farkas
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Lilian Varga
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Garred
- Laboratory of Molecular Medicine, Faculty of Health Sciences, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Zanier ER, Zangari R, Munthe-Fog L, Hein E, Zoerle T, Conte V, Orsini F, Tettamanti M, Stocchetti N, Garred P, De Simoni MG. Ficolin-3-mediated lectin complement pathway activation in patients with subarachnoid hemorrhage. Neurology 2014; 82:126-34. [PMID: 24336142 DOI: 10.1212/wnl.0000000000000020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES To assess the involvement of ficolin-3, the main initiator of the lectin complement pathway (LCP), in subarachnoid hemorrhage (SAH) pathology and outcome. METHODS In this preliminary exploratory study, plasma concentration of ficolin-3 and of ficolin-3-mediated functional LCP activity was measured, along with that of other LCP initiators (mannose-binding lectin, ficolin-2, and ficolin-1), C3 activation products, and soluble C5b-9 terminal complex, in a prospective cohort of 39 patients with SAH and 20 healthy controls. The following parameters were recorded: SAH severity, assessed using the World Federation of Neurosurgical Societies grading scale; vasospasm, defined as neuro-worsening with angiographic confirmation of vessel narrowing; cerebral ischemia, defined as hypodense lesion on CT scan performed before discharge; and 6-month outcome, assessed using the Glasgow Outcome Scale. RESULTS In patients, no changes were detected for ficolin-3 compared with controls. Notably, however, ficolin-3-mediated functional LCP activity was reduced. Low levels of plasma ficolin-3 and ficolin-3-mediated functional LCP activity were related to SAH severity, vasospasm, and cerebral ischemia. Moreover, ficolin-3 functional LCP activity was decreased in patients with unfavorable outcome. CONCLUSION Our data provide evidence that LCP is activated after SAH and that the actual plasma concentrations of ficolin-3 reflect the severity of brain injury as evaluated by clinical and structural parameters. These results support the idea that ficolin-3-mediated functional LCP activity may be targeted to control injury progression in SAH.
Collapse
Affiliation(s)
- Elisa R Zanier
- From the IRCCS-Istituto di Ricerche Farmacologiche Mario Negri (E.R.Z., R.Z., F.O., M.T., M.-G.D.S.), Department of Neuroscience, Milan; Department of Physiopathology and Transplant, Milan University and Neuro ICU (R.Z., T.Z., V.C., N.S.), Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; and Laboratory of Molecular Medicine (L.M.-F., E.H., P.G.), Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Matsushita M, Kilpatrick D, Shiraki H, Liu Y, Tateishi K, Tsujimura M, Endo Y, Fujita T. Purification, measurement of concentration, and functional complement assay of human ficolins. Methods Mol Biol 2014; 1100:141-59. [PMID: 24218257 DOI: 10.1007/978-1-62703-724-2_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Ficolins constitute a group of lectins involved in innate immunity. L-Ficolin, H-ficolin, and M-ficolin are present in human serum. The human ficolins differ in carbohydrate-binding specificity, but they have in common the ability to recognize the acetyl group. L-Ficolin and H-ficolin are associated with serine proteases termed MASPs (MBL-associated serine proteases) and their truncated proteins, and the complexes (L/H-ficolin-MASP) activate the lectin pathway of complement upon binding to their ligands. Recombinant M-ficolin is also able to form a complex with MASP, resulting in complement activation. L-Ficolin and H-ficolin can be purified as a complex with MASP from serum by utilizing their binding specificities. These ficolin-MASP complexes have an ability to activate C4. Human ficolins are quantified by ELISA using specific antibodies or ligands.
Collapse
Affiliation(s)
- Misao Matsushita
- Department of Applied Biochemistry, Tokai University, Hiratsuka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Evaluation of total complement function in human serum is an essential component of laboratory diagnostics of the human complement system. During recent years, established hemolytic assays for classical pathway and alternative pathway function, CH50 and AP50 assays, respectively, have been replaced in many diagnostic laboratories by ELISA assays. Next to an improved standardization, this assay platform also allows for functional analysis of the lectin pathway of complement. The present chapter describes the methodology of ELISA assays for assessment of the classical pathway, the alternative pathway, the MBL-dependent lectin pathway, and the Ficolin-3-dependent lectin pathway of complement in clinical laboratory diagnostics.
Collapse
Affiliation(s)
- Anja Roos
- Wieslab B.V., Eurodiagnostica, Nijmegen, The Netherlands
| | | |
Collapse
|
41
|
Hein E, Bay JT, Munthe-Fog L, Garred P. Ficolin-2 reveals different analytical and biological properties dependent on different sample handling procedures. Mol Immunol 2013; 56:406-12. [PMID: 23911396 DOI: 10.1016/j.molimm.2013.05.233] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 05/23/2013] [Indexed: 11/21/2022]
Abstract
Ficolin-2 (L-ficolin) is a germ line encoded pattern recognition molecule circulating in the blood, and functions as a recognition molecule in the lectin complement pathway. However, consistent and reliable measurements of Ficolin-2 concentration and activity have been difficult to achieve. After recurrent observations of deviations in Ficolin-2 properties between different blood sample procedures, we decided to investigate this closer. Blood samples from ten healthy donors were collected in various serum and plasma tubes and Ficolin-2 properties were evaluated by different ELISA setups. We found that serum prepared from tubes containing the clot activator silica used as a standard technique in many routine laboratories held a significantly lower concentration of Ficolin-2 as compared to the other sample types. Furthermore, Ficolin-2 binding and complement activation potential in this type of serum was impaired when using an acetylated compound as matrix. On the other hand, Ficolin-2 in serum made without clot activator and in plasma irrespective of additive used, had the same concentration and was capable of initiating the lectin pathway measured as C4 and C3 deposition on the ligand. No Ficolin-2 mediated formation of the terminal complement complex was observed under the applied assay conditions. In conclusion, our results show that Ficolin-2 is a promiscuous molecule and that care should be taken during sampling, handling and matrix chosen for measurement of Ficolin-2 levels and activity.
Collapse
Affiliation(s)
- Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
42
|
Matsushita M. Ficolins in complement activation. Mol Immunol 2013; 55:22-6. [PMID: 22959617 DOI: 10.1016/j.molimm.2012.08.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 08/06/2012] [Accepted: 08/07/2012] [Indexed: 11/30/2022]
Abstract
Ficolins are a group of multimeric lectins made up of single subunits each of which is composed of a collagen-like domain and a fibrinogen-like domain. Most of the ficolins identified to date bind to acetylated compounds such as N-acetylglucosamine (GlcNAc) and N-acetylgalactosamine (GalNAc). Ficolins in serum are complexed with MBL-associated serine proteases (MASPs) and their truncated proteins. These lectins play an important role in innate immunity. Binding of the ficolin-MASP complex to carbohydrates present on the surface of microbes initiates complement activation via the lectin pathway.
Collapse
Affiliation(s)
- Misao Matsushita
- Department of Applied Biochemistry, Tokai University, Hiratsuka, Kanagawa, Japan.
| |
Collapse
|
43
|
Matsushita M, Endo Y, Fujita T. Structural and functional overview of the lectin complement pathway: its molecular basis and physiological implication. Arch Immunol Ther Exp (Warsz) 2013; 61:273-83. [PMID: 23563865 DOI: 10.1007/s00005-013-0229-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 03/25/2013] [Indexed: 01/19/2023]
Abstract
The complement system is an effector mechanism in immunity. It is activated in three ways, the classical, alternative and lectin pathways. The lectin pathway is initiated by the binding of mannose-binding lectin (MBL) or ficolins to carbohydrates on the surfaces of pathogens. In humans, MBL and three types of ficolins (L-ficolin, H-ficolin, and M-ficolin) are present in plasma. Of these lectins, at least, MBL, L-ficolin, and H-ficolin are complexed with three types of MBL-associated serine proteases (MASPs), MASP-1, MASP-2, and MASP-3 and their truncated proteins (MAp44 and sMAP). In the lectin pathway, the lectin-MASP complex (i.e., a complex of lectin, MASPs and their truncated proteins) binds to pathogens, resulting in the activation of C4 and C2 to generate a C3 convertase capable of activating C3. MASP-2 is involved in the activation of C4 and C2. MASP-1 activates C2 and MASP-2. The functions of MASP-3, sMAP, and MAp44 in the lectin pathway remain unknown. MASP-1 and MASP-3 also have a role in the alternative pathway. MBL and ficolins are able to bind to a variety of pathogens depending on their carbohydrate binding specificity, resulting in the activation of the lectin pathway. Deficiencies of the components of the lectin pathway are associated to susceptibility to infection, indicating an important role of the lectin pathway in innate immunity. The lectin-MASP complex is also involved in innate immunity by activating the coagulation system. Recent findings suggest a crucial role of MASP-3 in development.
Collapse
Affiliation(s)
- Misao Matsushita
- Department of Applied Biochemistry, Tokai University, Hiratsuka, Kanagawa 259-1292, Japan
| | | | | |
Collapse
|
44
|
Jusko M, Potempa J, Kantyka T, Bielecka E, Miller HK, Kalinska M, Dubin G, Garred P, Shaw LN, Blom AM. Staphylococcal proteases aid in evasion of the human complement system. J Innate Immun 2013; 6:31-46. [PMID: 23838186 DOI: 10.1159/000351458] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/15/2013] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that presents severe health care concerns due to the prevalence of multiple antibiotic-resistant strains. New treatment strategies are urgently needed, which requires an understanding of disease causation mechanisms. Complement is one of the first lines of defense against bacterial pathogens, and S. aureus expresses several specific complement inhibitors. The effect of extracellular proteases from this bacterium on complement, however, has been the subject of limited investigation, except for a recent report regarding cleavage of the C3 component by aureolysin (Aur). We demonstrate here that four major extracellular proteases of S. aureus are potent complement inhibitors. Incubation of human serum with the cysteine proteases staphopain A and staphopain B, the serine protease V8 and the metalloproteinase Aur resulted in a drastic decrease in the hemolytic activity of serum, whereas two staphylococcal serine proteases D and E, had no effect. These four proteases were found to inhibit all pathways of complement due to the efficient degradation of several crucial components. Furthermore, S. aureus mutants lacking proteolytic enzymes were found to be more efficiently killed in human blood. Taken together, the major proteases of S. aureus appear to be important for pathogen-mediated evasion of the human complement system.
Collapse
Affiliation(s)
- Monika Jusko
- Section of Medical Protein Chemistry, Department of Laboratory Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
The C-type lectin of the aggrecan G3 domain activates complement. PLoS One 2013; 8:e61407. [PMID: 23596522 PMCID: PMC3626604 DOI: 10.1371/journal.pone.0061407] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 03/10/2013] [Indexed: 12/18/2022] Open
Abstract
Excessive complement activation contributes to joint diseases such as rheumatoid arthritis and osteoarthritis during which cartilage proteins are fragmented and released into the synovial fluid. Some of these proteins and fragments activate complement, which may sustain inflammation. The G3 domain of large cartilage proteoglycan aggrecan interacts with other extracellular matrix proteins, fibulins and tenascins, via its C-type lectin domain (CLD) and has important functions in matrix organization. Fragments containing G3 domain are released during normal aggrecan turnover, but increasingly so in disease. We now show that the aggrecan CLD part of the G3 domain activates the classical and to a lesser extent the alternative pathway of complement, via binding of C1q and C3, respectively. The complement control protein (CCP) domain adjacent to the CLD showed no effect on complement initiation. The binding of C1q to G3 depended on ionic interactions and was decreased in D2267N mutant G3. However, the observed complement activation was attenuated due to binding of complement inhibitor factor H to CLD and CCP domains. This was most apparent at the level of deposition of terminal complement components. Taken together our observations indicate aggrecan CLD as one factor involved in the sustained inflammation of the joint.
Collapse
|
46
|
Prohászka Z, Munthe-Fog L, Ueland T, Gombos T, Yndestad A, Förhécz Z, Skjoedt MO, Pozsonyi Z, Gustavsen A, Jánoskuti L, Karádi I, Gullestad L, Dahl CP, Askevold ET, Füst G, Aukrust P, Mollnes TE, Garred P. Association of ficolin-3 with severity and outcome of chronic heart failure. PLoS One 2013; 8:e60976. [PMID: 23596511 PMCID: PMC3626638 DOI: 10.1371/journal.pone.0060976] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/05/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Inflammatory mechanisms involving complement activation has been shown to take part in the pathophysiology of congestive heart failure, but the initiating mechanisms are unknown. We hypothesized that the main initiator molecules of the lectin complement pathway mannose-binding lectin (MBL), ficolin-2 and ficolin-3 were related to disease severity and outcome in chronic heart failure. METHODS AND RESULTS MBL, ficolin-2 and ficolin-3 plasma concentrations were determined in two consecutive cohorts comprising 190 patients from Hungary and 183 patients from Norway as well as controls. Disease severity and clinical parameters were determined at baseline, and all-cause mortality was registered after 5-years follow-up. In univariate analysis a low level of ficolin-3, but not that of MBL or ficolin-2, was significantly associated with advanced heart failure (New York Heart Association Class IV, p<0.001 for both cohorts) and showed inverse correlation with B- type natriuretic peptide (BNP) levels (r = -0.609, p<0.001 and r = -0.467, p<0.001, respectively). In multivariable Cox regression analysis, adjusted for age, gender and BNP, decreased plasma ficolin-3 was a significant predictor of mortality (HR 1.368, 95% CI 1.052-6.210; and HR 1.426, 95% CI 1.013-2.008, respectively). Low ficolin-3 levels were associated with increased complement activation product C3a and correspondingly decreased concentrations of complement factor C3. CONCLUSIONS This study provides evidence for an association of low ficolin-3 levels with advanced heart failure. Concordant results from two cohorts show that low levels of ficolin-3 are associated with advanced heart failure and outcome. The decrease of ficolin-3 was associated with increased complement activation.
Collapse
Affiliation(s)
- Zoltán Prohászka
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Thor Ueland
- The Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Timea Gombos
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Arne Yndestad
- The Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Zsolt Förhécz
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Zoltan Pozsonyi
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Alice Gustavsen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Lívia Jánoskuti
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - István Karádi
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Lars Gullestad
- Center for Heart Failure Research, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Christen P. Dahl
- The Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Erik T. Askevold
- The Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - George Füst
- IIIrd Department of Internal Medicine, Semmelweis University, and Research Group of Inflammation Biology and Immunogenomics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Pål Aukrust
- The Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital Rikshospitalet, Medical Faculty, University of Oslo, Norway
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
47
|
Pre-transplant levels of ficolin-3 are associated with kidney graft survival. Clin Immunol 2013; 146:240-7. [DOI: 10.1016/j.clim.2013.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 01/29/2023]
|
48
|
Csuka D, Munthe-Fog L, Skjoedt MO, Hein E, Bay JT, Varga L, Füst G, Garred P. A novel assay to quantitate MASP-2/ficolin-3 complexes in serum. J Immunol Methods 2013; 387:237-44. [DOI: 10.1016/j.jim.2012.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/23/2012] [Accepted: 10/31/2012] [Indexed: 10/27/2022]
|
49
|
Complement defects in patients with chronic rhinosinusitis. PLoS One 2012; 7:e47383. [PMID: 23144819 PMCID: PMC3492390 DOI: 10.1371/journal.pone.0047383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/12/2012] [Indexed: 11/19/2022] Open
Abstract
The complement system is an important part of our immune system, and complement defects lead generally to increased susceptibility to infections and autoimmune diseases. We have studied the role of complement activity in relation with chronic rhinosinusitis (CRS), and more specifically studied whether complement defects collectively predispose individuals for CRS or affect CRS severity. The participants comprised 87 CRS patients randomly selected from the general population, and a control group of 150 healthy blood donors. The CRS patients were diagnosed according to the European Position Paper on Rhinosinusitis and nasal Polyps criteria, and severity was evaluated by the Sino-nasal Outcome Test-22. Serum samples were analysed by ELISA for activity of the respective pathways of complement, and subsequently for serum levels of relevant components. We found that the frequency of complement defects was significantly higher among CRS patients than among healthy control subjects. A majority of Mannan-binding lectin deficient CRS patients was observed. The presence of complement defects had no influence on the severity of subjective symptoms. Our studies show that defects in the complement system collectively may play an immunological role related to the development of CRS. However, an association between severity of symptoms and presence of complement defects could not be demonstrated.
Collapse
|
50
|
Harboe M, Garred P, Lindstad JK, Pharo A, Müller F, Stahl GL, Lambris JD, Mollnes TE. The role of properdin in zymosan- and Escherichia coli-induced complement activation. THE JOURNAL OF IMMUNOLOGY 2012; 189:2606-13. [PMID: 22851705 DOI: 10.4049/jimmunol.1200269] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Properdin is well known as an enhancer of the alternative complement amplification loop when C3 is activated, whereas its role as a recognition molecule of exogenous pathogen-associated molecular patterns and initiator of complement activation is less understood. We therefore studied the role of properdin in activation of complement in normal human serum by zymosan and various Escherichia coli strains. In ELISA, microtiter plates coated with zymosan induced efficient complement activation with deposition of C4b and terminal complement complex on the solid phase. Virtually no deposition of C4b or terminal complement complex was observed with mannose-binding lectin (MBL)-deficient serum. Reconstitution with purified MBL showed distinct activation in both readouts. In ELISA, normal human serum-induced deposition of properdin by zymosan was abolished by the C3-inhibiting peptide compstatin. Flow cytometry was used to further explore whether properdin acts as an initial recognition molecule reacting directly with zymosan and three E. coli strains. Experiments reported by other authors were made with EGTA Mg²⁺ buffer, permitting autoactivation of C3. We found inhibition by compstatin on these substrates, indicating that properdin deposition depended on initial C3b deposition followed by properdin in a second step. Properdin released from human polymorphonuclear cells stimulated with PMA did not bind to zymosan or E. coli, but when incubated in properdin-depleted serum this form of properdin bound efficiently to both substrates in a strictly C3-dependent manner, as the binding was abolished by compstatin. Collectively, these data indicate that properdin in serum as well as polymorphonuclear-released properdin is unable to bind and initiate direct alternative pathway activation on these substrates.
Collapse
Affiliation(s)
- Morten Harboe
- Institute of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, NO-0027 Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|