IMMUNOLOGY

ORIGINAL ARTICLE

Human monoclonal Fab and human plasma antibodies to carbamylepitopes cross-react with malondialdehyde-adducts Outi Kummu,1,2,3 S. Pauliina Turunen,1,2,3 Piotr Prus,4,5 Jaakko Lehtim€aki,1,2 Marja Veneskoski,1,2 Chunguang Wang1,2 and Sohvi H€ orkk€ o1,2,3 1 Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Oulu, 2Medical Research Center Oulu, Oulu, 3NordLab Oulu, Oulu University Hospital, Oulu, 4Department of Biochemistry, University of Oulu, Oulu, and 5 Biocenter Oulu, University of Oulu, Oulu, Finland

doi:10.1111/imm.12204 Received 4 June 2013; revised 23 October 2013; accepted 23 October 2013. Correspondence: Outi Kummu, Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, PO Box 5000, 90014 University of Oulu, Oulu, Finland. Email: [email protected] Senior author: Sohvi Ho¨rkko¨, email: [email protected]

Summary Oxidized low-density lipoprotein (OxLDL) plays a crucial role in the development of atherosclerosis. Carbamylated LDL has been suggested to promote atherogenesis in patients with chronic kidney disease. Here we observed that plasma IgG and IgM antibodies to carbamylated epitopes were associated with IgG and IgM antibodies to oxidation-specific epitopes (q = 065–086, P < 0001) in healthy adults, suggesting a crossreaction between antibodies recognizing carbamyl-epitopes and malondialdehyde (MDA)/malondialdehyde acetaldehyde (MAA) -adducts. We used a phage display technique to clone a human Fab antibody that bound to carbamylated LDL and other carbamylated proteins. Anti-carbamyl-Fab (Fab106) cross-reacted with oxidation-specific epitopes, especially with MDA-LDL and MAA-LDL. We showed that Fab106 bound to apoptotic Jurkat cells known to contain these oxidation-specific epitopes, and the binding was competed with soluble carbamylated and MDA-/MAA-modified LDL and BSA. In addition, Fab106 was able to block the uptake of carbamyl-LDL and MDA-LDL by macrophages and stained mouse atherosclerotic lesions. The observed cross-reaction between carbamylated and MDA-/MAA-modified LDL and its contribution to enhanced atherogenesis in uraemic patients require further investigation. Keywords: antibody; atherosclerosis; carbamylation; cross-reaction; lowdensity lipoprotein.

Introduction Modified low-density lipoprotein (LDL) is a significant source of various antigens in atherosclerosis as well as in a number of other diseases characterized by increased oxidative stress. Oxidized LDL (OxLDL) is highly immunogenic and elicits antibody formation against oxidationspecific epitopes originating from various end-products of lipid peroxidation, including reactive aldehydes, such as malondialdehyde (MDA), 4-hydroxynonenal and 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine.1 Both IgM and IgG antibodies to OxLDL can be detected in the plasma of healthy human subjects as well as in animal models of atherosclerosis, and their role as potential markers and functional components in human disease has been of interest. It has been demonstrated in humans that

low levels of IgM antibodies to epitopes of OxLDL associate with increased carotid artery atherosclerosis.2 High levels of IgM antibodies against phosphocholine (PC), OxLDL and MDA-LDL predict a decreased rate of progression of atherosclerosis in hypertensive patients3 and low IgM antibody levels against PC predict the risk of death in haemodialysis patients.4 Recently, IgG and IgM antibodies to oxidation-specific epitopes were shown to predict cardiovascular disease and stroke in a 15-year follow-up study; IgG to copper oxidized LDL (CuOxLDL) predicted a higher cardiovascular disease event rate, whereas IgM to MDA-LDL predicted a lower event rate.5 The recent general view has been that B-1 cell activation and production of natural IgM are associated with atheroprotection.6 Animal studies have implied that the IgM antibodies to OxLDL are natural antibodies of the innate immune system,

Abbreviations: BHT, butylated hydroxytoluene; CPM, counts per minute; CuOxLDL, copper oxidized LDL; DAPI, 4′,6-diamidino-2-phenylindole; KD, steady state affinity; LDL, low-density lipoprotein; MAA, malondialdehyde acetaldehyde; MDA, malondialdehyde; OxLDL, oxidized low-density lipoprotein; PC, phosphocholine; PI, propidium iodide; RLU, relative light unit; RU, resonance unit 416

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope and they cross-react with a wide variety of common epitopes shared by microbes7 and apoptotic cells.8 One of the key characteristics of natural antibodies to OxLDL implicated in atherogenesis is their ability to inhibit the uptake of OxLDL by macrophage scavenger receptors.9 So far, the roles of the adaptive humoral immune response and IgG antibodies to OxLDL in atherogenesis have been less studied and the data remain inconclusive, even though some early studies suggested a pro-atherogenic role for IgG antibodies to OxLDL.10 Chronic kidney disease and uraemia are known to be associated with enhanced atherosclerosis, and one important mechanism suggested is post-translational modification of LDL by carbamylation (homocitrullination).11–13 This reaction is based on covalent binding of isocyanic acid, a decomposition product of urea, to proteins, lipids, amino acids and peptides.14–17 A minimal degree of carbamylated plasma proteins and haemoglobin is found in healthy subjects, whereas patients with renal insufficiency have increased levels of carbamylation of various plasma proteins due to elevated plasma urea levels.18 Higher levels of carbamylated proteins have recently been associated with higher mortality in patients with end-stage renal disease.19,20 Carbamylation in vivo may also occur by myeloperoxidase-catalysed oxidation of thiocyanate. Elevated thiocyanate levels are especially found in smokers. Myeloperoxidase-catalysed carbamylation has been linked to inflammation and atherosclerosis.21 We have previously shown in mice that carbamyl-LDL immunization induces a specific IgG immune response, which is cross-reactive with MDA-LDL.22 We have also shown that levels of IgG antibodies to carbamylated proteins are elevated in conditions known to induce enhanced carbamylation, such as uraemia and smoking.22 The aim of the current study was to investigate humoral antibody cross-reaction between carbamylated LDL and OxLDL in humans. Both carbamyl- and MDA-epitopes are found in humans and associated with increased atherosclerosis,19,20,23–26 which prompted us to investigate the association of human plasma antibodies binding to carbamyl-epitopes and oxidation-specific MDA-epitopes. An additional aim was to clone human monoclonal anticarbamyl-Fab antibody by phage display technique and investigate the binding properties and cross-reactivity to carbamyl- and oxidation-specific epitopes. Cross-reactive antibodies may provide important new knowledge concerning the enhanced atherogenesis in uraemic patients.

Materials and methods Human samples Human blood samples (n = 42 for antibody measurements, n = 4 for phage display library construction) were collected from healthy volunteers in the Clinical Research ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Center of Oulu University Hospital and informed written consent was obtained from each participant. The studies were approved by the Ethics Committee of Oulu University Hospital, Oulu, Finland (21/2006 and 159/2001) and followed the Declaration of Helsinki.

LDL isolation and modifications Low-density lipoprotein fraction (density 1019–1063 g/ ml) was isolated by sequential density gradient ultracentrifugation from pooled plasma of healthy donors.27 The LDL was carbamylated in vitro with potassium cyanate as previously described.22 First, 20 lM butylated hydroxytoluene (BHT) and 027 mM EDTA were added into freshly isolated LDL to minimize oxidation. Then, 2 mg LDL was diluted to 15 times the original volume with 03 M Na2B4O7, pH 80 buffer and 20 mg potassium cyanate was added per mg of LDL. The LDL was carbamylated for 6 hr at 37°. In addition, carbamyl-LDL preparation was further tested for the absence of thiobarbituric acid reactive substances, and tested with monoclonal antibodies for the absence of oxidized phospholipids. Carbamylated albumin (using BSA) was prepared similarly with 24 hr incubation. The extent of lysine modification was determined with the 2,4,6-trinitrobenzene sulphonic acid method28 and the amount of homocitrulline (carbamyl-lysine) was verified by amino acid analysis.22 Malondialdehyde-modified LDL (MDA-LDL) and malondialdehyde acetaldehyde-modified LDL (MAA-LDL) were prepared as described previously.29 Freshly prepared 05 M MDA was used to modify LDL with EDTA and BHT for 3 hr at + 37°: 05 M MDA was prepared from 1,1,3,3-tetramethoxypropane malonaldehyde-bis(dimethyl acetal) in 06% HCl and incubated at + 37° for 10 min. The pH was adjusted to 60–70 with NaOH and sterile water was added to a final volume of 4 ml. Then, 150 ll of 05 M MDA solution was used for MDA conjugation of 1 mg LDL protein. For MAA-modification, 05 M MDA pH 48 was prepared; 310 ll PBS, 140 ll 20% acetaldehyde, 5 mg LDL and 300 ll 05 M MDA were mixed in order. The pH was re-adjusted to 48 and the mixture was incubated at + 37° for 2 hr. MDA-BSA and MAABSA were prepared similarly. The extent of lysine modification was verified with the 2,4,6-trinitrobenzene sulphonic acid method28 and the absence of homocitrulline (carbamyl-lysine) in native, MDA- and MAA-modified proteins was verified by amino acid analysis as described previously.22 For copper oxidation, LDL without BHT was first extensively dialysed to remove EDTA and then oxidized by incubating LDL 1 mg/ml in PBS with 4 mM CuSO4 at 37° for 24 hr. The reaction was stopped by addition of EDTA to a 200 lM final concentration. Modified LDL and BSA preparations were dialysed against PBS with 027 mM EDTA and sterile filtered. 417

O. Kummu et al. Construction of phage display library Total RNA was isolated from peripheral blood lymphocytes with the RNeasy Mini Kit (Qiagen, Hilden, Germany) and used to synthesize cDNA with Moloney murine leukaemia virus reverse transcriptase and oligo (dT)18 primers included in the First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, Waltham, MA). The cDNAs were used for generation of antibody libraries. The phage display library was constructed in three rounds of PCR with human Fab primers.30 Heavy chain variable regions, j-light chain variable regions and k-light chain variable regions were first amplified separately using cDNA as a template. The constant regions of heavy chain and j- and k-light chains were amplified from the cloned human Fab template plasmids pComb3XTT and pComb3Xk obtained from Dr C.F. Barbas III at the Scripps Research Institute, La Jolla, CA. In the secondround PCR the heavy and light chain overlap products were generated separately from the pooled first-round PCR products. The final full-length Fab-coding fragments were assembled in the third PCR from the second-round products. The Fab-coding fragments were digested with SfiI and cloned into the pComb3X phagemid vector. The precipitated and resuspended ligation mixtures were transformed into XL1-Blue Escherichia coli (Agilent Technologies, Santa Clara, CA) by electroporation (Gene Pulser electroporator and cuvette with 02-cm gap; Bio-Rad, Hercules, CA). After transformation, the bacteria were amplified and infected with the VCSM13 helper phage (Agilent Technologies). Phage particles were obtained from the overnight culture medium by 4% (weight/volume) polyethylene glycol-8000/3% (weight/volume) sodium chloride precipitation and centrifugation at 15 000 g for 15 min. Five rounds of panning against carbamyl-LDL were performed, and individual clones binding to carbamyl-LDL were selected with chemiluminescence immunoassay. The phagemid DNAs were isolated with the QIAprep Spin Miniprep kit (Qiagen, Hilden, Germany). The nucleotide sequences were confirmed and aligned to the germline genes with the IMGT/V-QUEST sequence alignment tool (http://www.imgt.org).31,32

Chemiluminescence immunoassay method Chemiluminescence immunoassays to detect plasma antibodies binding to specific antigens, or to test human Fab antibody binding to antigens were performed as previously described.22,29,33 Antigens were immobilized on microtitre plates (Nunc Microfluor2, Thermo Fisher Scientific, Waltham, MA) overnight at + 4° in PBS with 027 mM EDTA. The plates were blocked with 05% gelatine in PBS with 027 mM EDTA for 30 min. The primary antibodies or human plasma samples (1 : 500) were diluted in 05% gelatine in PBS with 027 mM 418

EDTA and incubated for 1 hr at room temperature or overnight at + 4°. The amount of bound molecules was measured with alkaline phosphatase-conjugated secondary antibodies, or alkaline phosphatase-conjugated Neutravidin (for biotinylated antibodies), LumiPhos 530 (Lumigen, Southfield, MI) as a substrate and a Wallac Victor3 (Perkin Elmer, Waltham, MA). The results are expressed as relative light units measured in 100 ms (RLU/100 ms). The specificity of human plasma antibodies and Fab106 antibody to carbamyl-LDL was tested using liquid-phase competition immunoassay. Plasma or antibody dilutions were incubated overnight at + 4° in the presence and absence of competitors (0–200 lg/ml). The immunocomplexes were pelleted by centrifugation for 30 min at 16 000 g, + 4° and antibodies remaining in the liquid phase were analysed using the chemiluminescence immunoassay described above.

Analysis of modified BSA on SDS–PAGE and Western blot Native BSA, carbamyl-BSA, MDA-BSA and MAA-BSA (2 lg each) were reduced with b-mercaptoethanol and analysed on 10% SDS–PAGE and by Western blotting. Precision Plus ProteinTM All Blue standard (Bio-Rad) was used as the molecular weight marker. The samples were transferred from the gel onto nitrocellulose membrane and blocked with 3% gelatine in PBS. The membrane was incubated with Fab106 antibody (2 lg/ml) for 1 hr at room temperature and the binding was detected with Fab-specific goat anti-human IgG (Sigma, St Louis, MO) and AlexaFluor 680-labelled donkey anti-goat (Invitrogen, Carlsbad, CA) antibodies (05 lg/ml 1 hr and 02 lg/ml 45 min at room temperature, respectively). The membrane was scanned with an Odyssey infrared scanner (LI-COR Biosciences, Lincoln, NE).

Biacore interaction analysis Studies were performed with a Biacore T200 instrument at 25° using a standard CM5 sensor chip (GE Healthcare, Uppsala, Sweden). Dulbecco’s PBS with 027 mM EDTA was used as a running buffer. The Biacore T200 instrument was cleaned according to the manufacturer’s recommended methods before each measurement. Fab106 was immobilized on a Biacore CM5 chip. First, Fab106 was diluted into Biacore immobilization buffer (10 mM sodium acetate pH 50) to obtain a final concentration of 38 lg/ml. The new inserted CM5 chip was primed with immobilization buffer and equilibrated to 25°. The chip surface was activated by a 7-min injection of an earlier prepared mixture of 01 M N-hydroxysuccinimide and 04 M N-ethyl-N-(3-dimethylaminopropyl)carbodiimide in water with a flow rate of 10 ll/min. The Fab106 sample was then injected during 60 seconds, and residual ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope activated groups on the chip surface were blocked by a 7min injection of 1 M ethanolamine (pH 85). On average, 1500 resonance units (RU) were immobilized. The channel Fc2 was used for modification, channel Fc1 was used as a reference and left unmodified. Native BSA, carbamylBSA, MDA-BSA, MAA-BSA, native LDL, carbamyl-LDL, MDA-LDL and MAA-LDL were diluted into running buffer to obtain a final concentration within the range of 20 nM to 50 lM. Interaction analysis was performed by injecting each sample for 3 min with the flow rate of 30 ll/min. The chip was regenerated with a 30-second pulse of 1 M sodium chloride and a 30-second pulse of 005% digitonin. Data were analysed using BIACORE T200 evaluation software v1.0.

Binding of Fab106 to apoptotic human T cells Apoptosis was induced in human Jurkat T cells on a Petri dish by UV irradiation 51 mJ/cm2 (UV Stratalinker; Stratagene, Santa Clara, CA) followed by overnight incubation in a humidified atmosphere with 5% CO2 at + 37°. The cells were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum, 10 mM HEPES, 1 mM sodium pyruvate, 2 mM L-glutamine, 100 U/ ml penicillin and 100 lg/ml streptomycin. Then, 5 9 105 cells were stained with anti-carbamyl-Fab106 antibody (5 lg/ml) and the binding was detected with FITClabelled anti-human IgG (Fab-specific) (1 lg/ml, Sigma). Jurkat cells were also counterstained with 4′,6-diamidino2-phenylindole (DAPI) nucleic acid stain according to the manufacturer’s protocol (Invitrogen). Cells were imaged using a Zeiss Axio Imager.D2 microscope with a Colibri LED light source attached to an AxioCam MRm camera and AXIOVISION 4.8 software. Fab106 binding to apoptotic cells (1 9 106 cells) was verified by flow cytometry analysis. Direct staining with antibody (25 lg/ml) and competitive assay to investigate the binding specificity were performed. Fab106 in a final concentration of 3 lg/ml was incubated with or without competitors (250 lg/ml) carbamyl-LDL, MDA-LDL, native LDL, carbamyl-BSA, MDA-BSA, MAA-BSA and native BSA diluted in FACS buffer (01% BSA in PBS) overnight at + 4°. Samples were prepared in triplicates. Apoptotic Jurkat cells were washed with FACS buffer and centrifuged at 1800 g for 5 min. The competition samples were added and incubated for 40 min at + 4°. Cells were washed and incubated with FITC-labelled secondary antihuman IgG (Fab-specific) antibody (05 lg/ml, Sigma) for 40 min at + 4°. The washing was repeated and apoptotic cells were identified with propidium iodide (PI) staining (1 lg/ml). Binding of Fab106 to apoptotic T cells was analysed with a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA) and the data were analysed using FCS EXPRESS V3 software (De Novo Software, Los Angeles, CA). ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Modified LDL uptake into J774A.1 mouse macrophages Macrophage uptake assay was performed with 3H-labelled carbamyl-LDL and IRDye800-conjugated MDA-LDL as described earlier34 with slight modifications. To prepare 3 H-carbamyl-LDL, 40 mCi of tritium-labelled cholesteryl oleate [cholesteryl-1,2–3H(N)] (Perkin Elmer) per mg carbamyl-LDL was dried under nitrogen flow. Dimethylsulphoxide (10% of final volume) was added and vortexed for 1 min, mixed with carbamyl-LDL and incubated for 2 hr at 40° in a water bath followed by dialysis against 150 mM NaCl at + 4°. J774A.1 mouse macrophages were plated in 96-well plates (1 9 105 cells/ well) in Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum, 100 U/ml penicillin and 100 lg/ ml streptomycin and grown overnight at + 37° with 5% CO2. The cells were washed and incubated at + 37° for 2 hr with 3H-carbamyl-LDL (25 lg/ml) in the presence or absence of unlabelled carbamyl-LDL as a competitor (75 lg/ml) or Fab106 antibody (305 lg/ml) mixed with labelled carbamyl-LDL. The cells were washed and lysed with 01 M NaOH for 2 hr in a plate shaker (1000 rpm) at room temperature. Cell lysate (50 ll or 25 ll) and 175 ll OptiPhase supermix scintillation liquid (Perkin Elmer) per well were added to a Wallac 96-well sample plate, shaked for 30 min (1000 rpm) and incubated for 15 min at room temperature. Cellular radioactivity was measured using a Wallac MicroBeta TriLux 1450 LSC & Luminescence Counter (Perkin Elmer). The cell protein concentrations were determined and radioactivity in cells (counts per minute, CPM)/lg cell protein was calculated. The MDA-LDL was labelled with IRDye 800CW (LICOR Biosciences) according to the manufacturer’s protocol. IRDye800-conjugated MDA-LDL (1 lg/ml) in serum-free Dulbecco’s modified Eagle’s medium was incubated with macrophages (2 9 105 cells/well) in the presence (50 lg/ml) or absence of the anti-carbamyl Fab106 for 3 hr at + 37°. A 50-fold excess of unlabelled MDA-LDL in separate wells was used as a control. The cells were washed three times with PBS and analysed with an Odyssey infrared imager (LI-COR Biosciences). After analysis the cells were lysed with 01 M sodium hydroxide overnight at room temperature, the amount of cell protein was measured, and the results were calculated as an infrared signal normalized with the protein contents of each well.

Immunohistochemical staining of mouse atherosclerotic lesions Formalin-fixed and paraffin-embedded heart cross-sections of aortic origin from LDLR / mice fed with a high-fat diet were stained with Fab106. As a control, aortic origin heart cross-sections from normal C57BL/6 mice without atherosclerosis were stained similarly. Antigen 419

O. Kummu et al. retrieval was performed by a 10-minute treatment with 10 mM sodium citrate (pH 60) near its boiling point. The cross-sections were stained with the Goat-on-Rodent HRP-polymer kit (Biocare Medical, Concord, CA) according to the kit protocol using DAB+ (3,3′-diaminobenzidine) chromogen (Dako, Glostrup, Denmark). Briefly, the endogenous peroxidase activity was quenched with the Peroxidase Block. Fab106 (5 lg/ml) or PBS was applied onto separate sections for 1 hr, followed by secondary goat anti-human IgG (Fab-specific) antibody (2 lg/ml, Sigma). HRP-labelled polymer conjugated with anti-goat antibody and DAB+ chromogen were used for detection. The cross-sections were further counterstained with Mayer’s haematoxylin. Images were acquired with a Leica DM 3000 microscope and Leica Application Suite (LAS) V4.1.0 (Leica microsystems).

Statistical analysis Data analysis was performed with SPSS Statistics 19.0. All the results for continuous variables are presented as mean  SD. The differences between the groups were analysed by Student’s t-test or Mann–Whitney U-test, as appropriate. Associations were analysed by Spearman’s correlation coefficient. P-value < 005 was regarded as statistically significant. P-values are marked with asterisks in the figures as follows: *P < 005; **P < 001; ***P < 0001.

Results Human plasma contains antibodies binding to carbamyl-epitope that associate with antibodies to MDA-/MAA-adducts Human plasma IgG and IgM binding to carbamyl-LDL, CuOxLDL, MDA-LDL and MAA-LDL and to carbamylBSA, MDA-BSA and MAA-BSA were measured from 42 healthy adults aged between 24 and 59 years (mean 36  9 years). Statistically significant, strong positive associations were observed between plasma antibody levels, both IgG and IgM, to carbamylated and oxidationspecific epitopes in LDL (Fig. 1a) and BSA (Fig. 1b). Competitive immunoassays were then performed to study the specificity of human plasma antibodies bound to carbamyl-epitope. Carbamyl-LDL and MDA-LDL competed for binding of plasma IgG and IgM antibodies to carbamyl-LDL (Fig. 2a) and also to MDA-LDL (Fig. 2b). Carbamyl-BSA, MDA-BSA and MAA-BSA also competed for plasma IgG binding to carbamyl-BSA (Fig. 2c) and

MDA-BSA (Fig. 2d). MDA- and MAA-adducts were more efficient soluble competitors than carbamylated proteins when measuring polyclonal plasma IgG and IgM binding to immobilized carbamylated antigens (Fig. 2a,c). Plasma IgM antibody levels to carbamyl-BSA and MDA-BSA were very low, and therefore competition assay was not performed.

Human monoclonal anti-carbamyl Fab antibody (Fab106) cross-reacts with oxidation-specific epitopes The origin and specificity of the human antibodies to carbamylated proteins were further investigated by constructing a phage display Fab-antibody library from peripheral blood lymphocytes of four healthy humans. After five rounds of panning against carbamyl-LDL, several phage clones were randomly selected (n = 19) from both VH/Vj and VH/Vk libraries, all demonstrating binding to carbamyl-LDL. The characteristics of 10 human monoclonal anti-carbamyl Fab antibody clones including VH/DH/JH and VL/JL gene use are presented in Table 1. Nine out of the ten clones originated from the large VH3 family (six from VH3-33, two from VH3-23 and one from VH3-66 germline gene), and only one clone originated from the VH4 family (VH4-39 germline gene). The DH motifs were very variable, but all clones demonstrated an almost exclusive use of JH4. In addition, the VH sequences demonstrated a surprisingly high degree of identity to the corresponding germline genes (between 86% and 100%). One clone (IFP-02_106) was chosen for the production of soluble Fab antibody (Fab106). The binding of Fab106 to native and modified LDL and BSA was tested with chemiluminescence immunoassay (Fig. 3a). Fab106 showed a dose-dependent increase in binding to carbamyl-LDL, but also to MDA-LDL, MAA-LDL and CuOxLDL (Fig. 3a). Similar binding was observed to carbamylated BSA and MDA-/MAA-modified BSA. Fab106 did not, however, bind to PC conjugated to BSA (PC-BSA), which suggests that binding to CuOxLDL was a result of the small amount of MDA-epitopes generated during preparation of CuOxLDL1 (Fig. 3a). The binding specificity of Fab106 to carbamyl-modification was determined in liquid-phase competition immunoassay (Fig. 3b). Anticarbamyl-Fab binding to carbamyl-LDL was competed out by adding an increasing amount of carbamyl-BSA and carbamyl-LDL, but not by native LDL or native BSA (Fig. 3b). This suggests that the antibody was specific for the carbamyl-epitope, and not the carrier protein. Fab106 binding to carbamyl-LDL was also competed with MDALDL and MAA-LDL (Fig. 3b) demonstrating cross-reactive

Figure 1. Association of human plasma anti-carbamyl antibodies with antibodies to oxidation-specific epitopes. (a) Plasma samples of healthy adult humans (n = 42) were analysed for IgG and IgM binding to carbamyl-low-density lipoprotein (LDL), copper oxidized LDL (CuOxLDL), malondialdehyde (MDA) -LDL and malondialdehyde acetaldehyde (MAA) -LDL. (b) Assays were also performed using carbamylated and MDAor MAA-modified BSA. Associations were determined using Spearman rank correlation coefficient. RLU = relative light unit.

420

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope (a)

45 000

35 000 30 000 25 000

ρ = 0·759 P < 0·001

20 000 15 000 10 000

Plasma IgM to CuOxLDL (RLU/100 ms)

Plasma IgG to CuOxLDL (RLU/100 ms)

25 000 40 000

20 000

15 000

ρ = 0·833 P < 0·001

10 000

5000

5000 40 000 50 000

10 000

20 000

30 000

40 000

15 000

25 000

20 000

35 000

40 000 35 000 30 000

ρ = 0·857

25 000

P < 0·001

20 000 15 000

Plasma IgM to MDA-LDL (RLU/100 ms)

45 000

Plasma IgG to MDA-LDL (RLU/100 ms)

10 000

5000

50 000

30 000 25 000

ρ = 0·738 P < 0·001

20 000 15 000 10 000

10 000 5000 5000 10 000

20 000

30 000

40 000

50 000

90 000 80 000 70 000 60 000

ρ = 0·696

50 000

P < 0·001

40 000

60 000

Plasma IgM to MAA-LDL (RLU/100 ms)

Plasma IgG to MAA-LDL (RLU/100 ms)

100 000

5000

20 000

25 000

50 000

40 000

30 000

ρ = 0·769 P < 0·001

20 000

20 000

30 000

40 000

5000

50 000

Plasma IgG to carbamyl-LDL (RLU/100 ms)

10 000

15 000

20 000

25 000

Plasma IgM to carbamyl-LDL (RLU/100 ms)

14 000

20 000

12 000 10 000

ρ = 0·829

8000

P < 0·001

Plasma IgM to MDA-BSA (RLU/100 ms)

Plasma IgG to MDA-BSA (RLU/100 ms)

20 000

0 10 000

6000 4000 2000 35 000

15 000

ρ = 0·820 10 000

P < 0·001

5000

0 25 000

1000

2000

3000

4000

5000

6000

7000

8000

1000

1500

2000

2500

3000

3500

4000

2000

2500

3000

3500

4000

30 000 20 000

Plasma IgM to MAA-BSA (RLU/100 ms)

Plasma IgG to MAA-BSA (RLU/100 ms)

15 000

10 000

30 000

(b)

10 000

25 000 20 000

ρ = 0·647 P < 0·001

15 000 10 000 5000 0

15 000

ρ = 0·735 P < 0·001

10 000

5000

0 1000

2000

3000

4000

5000

6000

7000

Plasma IgG to carbamyl-BSA (RLU/100 ms)

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

8000

1000

1500

Plasma IgM to carbamyl-BSA (RLU/100 ms)

421

O. Kummu et al. MDA-LDL

Carbamyl-LDL

(b)

Carbamyl-LDL

MAA-BSA

MDA-LDL

1·2

1·0

1·0

0·8

0·8

0·6

0·6

0·4

0·4

0·2

0·2

0·0

0·0

1·2

1·2

1·0

1·0

0·8

0·8

0·6

0·6

0·4

0·4

0·2

0·2 0·0

0·0 0

20

(c)

40

60

80

100

0

20

(d)

Carbamyl-BSA

1·2

40

60

80

100

MDA-BSA

1·2

1·0

1·0

0·8

0·8

0·6

0·6

0·4

0·4

0·2

0·2

0·0

IgG to MDA-BSA (B/B0)

IgG to carbamyl-BSA (B/B0)

IgM to MDA-LDL (B/B0)

IgM to carbamyl-LDL (B/B0)

(a)

MDA-BSA

IgG to MDA-LDL (B/B0)

IgG to carbamyl-LDL (B/B0)

1·2

Carbamyl-BSA

0·0 0

20

40

60

80

100

Competitor (µg/ml)

0

20

40

60

80

100

Competitor (µg/ml)

Figure 2. Specificity of human plasma antibodies to carbamyl epitopes. Specific binding of human plasma IgM and IgG to immobilized (a) carbamyl-low-density lipoprotein (LDL) and (b) malondialdehyde (MDA) -LDL was analysed using carbamyl-LDL and MDA-LDL as competitors in a liquid-phase competition immunoassay. Specificity of human plasma IgG to (c) carbamyl-BSA and (d) MDA-BSA was analysed using carbamyl-BSA, MDA- and malondialdehyde acetaldehyde (MAA) -BSA as soluble competitors. Mean (SD) of four samples is shown.

epitopes in carbamylated and MDA-/MAA-modified proteins. In addition, CuOxLDL was shown to compete for Fab106 binding to carbamyl-LDL but no competition for PC-BSA was observed, verifying that the MDA-epitopes in CuOxLDL were recognized by Fab106 antibody (Fig. 3b). Native BSA, carbamyl-BSA, MDA-BSA and MAA-BSA were analysed on reducing SDS–PAGE (Fig. 3c) and Western blotted with Fab106 antibody (Fig. 3d). Fab106 bound to carbamyl-BSA and also to MDA-BSA and MAA-BSA, but not to native BSA in Western blot analysis (Fig. 3d). 422

Fab106 binds with high affinity to carbamyl- and MDA-/MAA-adducts The steady-state binding affinity of monoclonal Fab106 antibody to carbamyl- and MDA-/MAA-modified antigens (BSA, Fig. 4a and LDL, Fig. 4b) was tested by surface plasmon resonance analysis. Fab106 bound most strongly to MDA-LDL and MAA-LDL with steady-state affinity (KD) values 524 9 10 6 M and 407 9 10 6 M, respectively. Binding to carbamyl-LDL was also high compared with native LDL, with the KD value ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope Table 1. Characteristics of 10 human monoclonal anti-carbamyl-Fab clones

Clone

VDJ-usage

Identity of V-region to germline (%)

1

VH3-33*01/DH6-25*01/JH4*02 Vj3-20*01/Jj1*01 VH3-33*01/DH6-25*01/JH4*02 Vj3-20*01/Jj1*01 VH3-33*01/DH3-10*01/JH4*02 Vj1-39*01/Jj1*01 VH3-23*01/DH3-3*01/JH4*02 Vj1-39*01/Jj3*01 VH4-39*01/DH1-26*01/JH4*03 Vj1-5*03/Jj1*01 VH3-33*01/DH3-3*01/JH4*02 Vk3-1*01/Jk3*02 VH3-33*01/DH2-02*01/JH4*02 Vk2-14*03/Jk2*01 VH3-23*01/DH5-18*01/JH4*02 Vk3-1*01/Jk2*01 VH3-33*01/DH2-8*01/JH4*02 Vk10-54*01/Jk3*02 VH3-66*02/DH3-3*01/JH4*03 Vk2-28*01/Jk2*01

1000 973 1000 918 861 907 1000 899 911 906 992 572 932 736 970 932 935 907 935 1000

21 3 4 5 6 7 8 9 10

Fab106 binds to apoptotic Jurkat cells and binding can be inhibited with carbamyl-, MDA- and MAAmodified proteins To study further the cross-reactivity between oxidationspecific epitopes and carbamyl-epitopes, the binding of Fab106 to apoptotic cells was investigated. Previously, apoptotic cells have been shown to contain oxidationspecific epitopes.8 Fab106 demonstrated binding to apoptotic cells in immunofluorescence microscopy (Fig. 5a, b). Fab106 binding to Jurkat T cells with UVinduced apoptosis was further investigated in a flow cytometry assay (Fig. 5c–i). Fab106 did not bind to non-apoptotic, PI-negative cells (Gate 1, Fig. 5d–f), but bound to PI-positive late apoptotic cells (Gate 2,

1

Clone IFP-02_106 (Fab106).

200 000

213 9 10 5 M. Fab106 also bound modified BSA antigens, but with a slightly weaker affinity than modified LDL. KD values for MAA-BSA, MDA-BSA and carbamyl509 9 10 4 M and BSA were 328 9 10 5 M, 4 158 9 10 M, respectively. Binding affinity to native LDL and native BSA was very weak, with KD values 808 9 10 1 M for LDL and 970 9 10 2 M for BSA. Steady-state affinity KD (M) values are shown in Fig. 4(c).

Native LDL

Carbamyl-LDL

MDA-LDL

MAA-LDL

CuOxLDL

Native BSA

Carbamyl-BSA

MDA-BSA

MAA-BSA

PC-BSA

LDL antigens

(a)

(b)

LDL competitors

1·4

kDa

1·2

160 000

0·8

80 000

0·6 0·4

40 000

0·2 0 0

5

120 000

10 15 BSA antigens

20

0

100 BSA competitors

200

0·0 1·6 1·4 1·2 1·0

80 000

0·8 0·6 40 000

Fab106 bound to carbamyl-LDL (B/B0)

Fab106 bound to antigen (RLU/100 ms)

1·0 120 000

1

2

3

4

5

250 150 100 75 50 37

(c)

250 150 100 75 50

0·4 0·2

0

37

(d)

0·0 0

5

10

15

20

0

Fab (µg/ml)

100

200

Competitor (µg/ml)

Figure 3. Characterization of binding properties of Fab106. (a) Direct binding assay for anti-carbamyl-Fab (Fab106). Increasing amount of Fab106 (0–20 lg/ml) bound to native low-density lipoprotein (LDL), carbamyl-LDL, malondialdehyde (MDA) -LDL, malondialdehyde acetaldehyde (MAA) -LDL and copper oxidized LDL (CuOxLDL) (upper panel) or to native BSA, carbamyl-BSA, MDA-BSA, MAA-BSA and phosphocholine (PC) -BSA (lower panel). (b) Competitive liquid phase immunoassay to demonstrate Fab106 binding specificity. Fab106 binding to immobilized carbamyl-LDL was tested when native LDL, carbamyl-LDL, MDA-LDL, MAA-LDL and CuOxLDL and also native BSA, carbamyl-BSA, MDA-BSA, MAA-BSA and PC-BSA were used as competitors. RLU = relative light unit. (c) SDS–PAGE of modified BSA and (d) Western blot with Fab106 antibody. Molecular weight marker (lane 1), native BSA (lane 2), carbamyl-BSA (lane 3), MDA-BSA (lane 4) and MAA-BSA (lane 5).

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

423

O. Kummu et al. (a) 30

2·5

Response (RU)

Native BSA

25

2·0

Carbamyl-BSA

20

1·5

15 1·0 10 0·5

5

0·0

0

0·0

Response (RU)

2·0E-7 4·0E-7 6·0E-7 8·0E-7 1·0E-6 1·2E-6 1·4E-6

0·0

3·0E-6 6·0E-6 9·0E-6 1·2E-5 1·5E-5 1·8E-5

80

50

MDA-BSA

70

MAA-BSA

(c)

60 30

50

20

30 10

9·70e-2

Native LDL

8·08e-1

Carbamyl-BSA

1·58e-4

Carbamyl-LDL

2·13e-5

MDA-BSA

5·09e-4

MDA-LDL

5·24e-6

MAA-BSA

3·28e-5

MAA-LDL

4·07e-6

20 10 0

1E-5

2E-5

3E-5

4E-5

5E-5

6E-5

0 0·0

5·0E-6

Concentration (M)

1·0E-5

1·5E-5

2·0E-5

2·5E-5

Concentration (M)

(b) 350

Response (RU)

Native BSA

40

0

300

Native LDL

70

50

200

40

150

30

100

20

50

10

0

0

200

2·0E-6 4·0E-6 6·0E-6 8·0E-6 1·0E-5 1·2E-5

MDA-LDL

Carbamyl-LDL

60

250

0·0

Response (RU)

Affinity-KD (M)

40

0·0 300

4·0E-7

8·0E-7

1·2E-6

1·6E-6

2·0E-6

MAA-LDL

250

160

200 120 150 80 100 40 0 0·0

50

5·0E-7 1·0E-6 1·5E-6 2·0E-6 2·5E-6 3·0E-6

Concentration (M)

0 0·0

5·0E-7 1·0E-6 1·5E-6 2·0E-6 2·5E-6 3·0E-6

Concentration (M)

Figure 4. Biacore interaction analysis. Steady-state affinity curves for immobilized Fab106 bound to soluble antigens (a) native BSA, carbamylBSA, malondialdehyde (MDA) -BSA and malondialdehyde acetaldehyde (MAA) -BSA and also to (b) native low-density lipoprotein (LDL), carbamyl-LDL, MDA-LDL and MAA-LDL. (c) Steady-state affinity KD (M) values.

Fig. 5g–i). Fab106 bound approximately 30% of the apoptotic cell population, as shown in Fig. 5(h). The binding of Fab106 to apoptotic cells was reduced to 18% when carbamyl-LDL was added or to 8% when carbamyl-BSA was added as a competitor (Fig. 6). Fab106 bound < 4% of the apoptotic cells when MDALDL, MDA-BSA or MAA-BSA was used as competitor (Fig. 6). This provides further evidence for the Fab106 cross-reaction between carbamylated and MDA-/MAAepitopes. MAA-LDL was not used as a competitor in this flow cytometry assay, because of its tendency to form aggregates when added to apoptotic cells. Native LDL and native BSA did not compete for Fab106 binding to apoptotic Jurkat cells (Fig. 6). The binding of the negative control Fab antibody (clone IFUp-08_109) to 424

Jurkat cells was also tested as described in the Supplementary material (Data S1 and Fig. S1).

Fab106 blocks the uptake of carbamyl-LDL and MDA-LDL by macrophages and binds to epitopes in mouse atherosclerotic lesion Uptake of modified LDL into macrophages is a key step in the pathogenesis of atherosclerosis. The functional role of antibodies to modified lipid and protein epitopes in vivo is not fully understood. The role of Fab106 antibody was investigated in macrophage uptake assay (Fig. 7). Fab106 was able to significantly inhibit the uptake of 3Hcarbamyl-LDL by J774A.1 mouse macrophages, a 63% decrease (P < 0001) was observed when Fab106 antibody ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope (a)

Fab106

(b)

(c)

2Ab

1024

SSC-H

768 512 Gate 1

Gate 2 256 0 0

Cells only

256 512 768 1024 FSC-H

Gate 1 Non-apoptotic Jurkat cells 0·04%

0·66%

(e) 104

FL1-H

102

100

(f) 299

103

101 4·22%

102

95·08%

4·05%

31·77%

0·00%

10 FL1-H

10

2

10

101

0·00%

0·00%

98·48%

0

FSC-H

2 Ab

Cells only Cells + 2 Ab Cells + Fab106

188

102

126 63

68·23%

100

256 512 768 1024

101 102 FL1-H

(i) 251

3

101

100

0 100

Count

3

95·74%

256 512 768 1024 FSC-H

Gate 2 Apoptotic Jurkat cells (g) 104 (h) 104 1·52%

150 75

100 256 512 768 1024 0 Fab106 FSC-H

2 Ab

Cells only Cells + 2 Ab Cells + Fab106

224

101

0

FL1-H

Figure 5. Fab106 binding to apoptotic Jurkat T cells. Immunofluorescence microscopy image of Fab106 bound to apoptotic Jurkat cells (a) and secondary antibody control (b). Fab antibody bound to apoptotic cells is shown in green (FITC) and nuclear DNA staining in blue (DAPI). (c–i) Flow cytometry assay for non-apoptotic, propidium iodide (PI) -negative (Gate 1, d-f) and apoptotic, PI-positive (Gate 2, g-i) Jurkat cells after UV irradiation of 51 mJ/cm2. Density plots demonstrate secondary antibody (d, g) and Fab106 antibody (e, h) bound to Jurkat cells and histograms (f, i) illustrate apoptotic cells only (black), secondary antibody binding (red) and Fab106 antibody binding (blue) to Jurkat cells.

FL1-H

103

0·19%

0·01%

Count

(d) 104

0

0·00%

256 512 768 1024

0 100

FSC-H

Fab106

101

102

FL1-H

Figure 6. Flow cytometry competition assay. Fab106 binding to apoptotic Jurkat cells was analysed with flow cytometry using soluble competitors carbamyl-low-density lipoprotein (LDL), malondialdehyde (MDA) -LDL, native LDL, carbamyl-BSA, MDA-BSA, malondialdehyde acetaldehyde (MAA) -BSA and native BSA. Propidium iodide (PI) -positive apoptotic Jurkat cells were gated and cells in the upper left (UL) quadrant of forward scatter versus FL-1 density plots were analysed (see Fig. 5g and 5h). Samples were analysed in triplicates.

Fab106 bound to apoptotic cells (% of cells in UL quadrant)

40

30

–40% 20

–89%

–74%

–87%

–87%

**

**

**

**

10

** 0 2 Ab only

was added together with labelled carbamyl-LDL (Fig. 7a). A similar assay was performed using IRDye800-labelled MDA-LDL. Fab106 was able to inhibit 33% (P < 0001) of the IRDye800-MDA-LDL uptake by J774A.1 macroª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Fab106

Native Carbamyl- MDALDL LDL LDL

Native Carbamyl- MDABSA BSA BSA

MAABSA

Competitor

phages (Fig. 7b). Fab106 was also used for staining mouse heart aortic origin cross-sections. The Fab106 antibody bound to epitopes found in advanced atherosclerotic lesions that develop within intima in LDLR / mice 425

O. Kummu et al. (a)

***

***

1·2

***

1·0

1·0

0·8

0·8

0·6

0·6

0·4

0·4

0·2

0·2

0·0 No competitor

unlabelled carbamyl-LDL

Fab106

No competitor

Competitor

(a)

unlabelled MDA-LDL

Fab106

Competitor

0·0

Relative IRDye800-MDA-LDL uptake by J774A.1 macrophages

Relative 3H-carbamyl-LDL uptake by J774A.1 macrophages

1·2

(b)

***

Figure 7. Carbamyl-low-density lipoprotein (LDL) and malondialdehyde (MDA) -LDL uptake by mouse J774A.1 macrophages. Uptake of (a) 3H-labelled carbamyl-LDL and (b) IRDye800-labelled MDA-LDL was tested in the presence and absence of Fab106. 309 or 509 excess of unlabelled carbamyl- or MDA-LDL was used as an assay control.

(b)

(c)

Figure 8. Immunohistochemical staining of atherosclerotic lesions in mouse heart aortic origin cross-section with the anti-carbamyl Fab106 antibody. Secondary antibody control (a) and Fab106 staining (b, c). Magnification 59 (a, b) and 109 (c), scale bar = 400 lm.

(Fig. 8). No similar staining was observed in normal aortic origin cross-sections from C57BL/6 mice without atherosclerotic lesions (see Supplementary material, Fig. S2).

Discussion Both carbamyl-epitopes and oxidation-specific MDA-LDL epitopes are found in humans23,24,35 and have been shown to contribute to the development of atherosclerosis.19,20,25,26 Furthermore, both epitopes are immunogenic and they induce antibody production.22,36–38 Previously, we showed that carbamyl-LDL immunization induces a 426

specific IgG immune response that cross-reacted with MDA-adducts in mice.22 This prompted us to study further if cross-reactive antibodies to carbamyl-adducts and MDA-adducts exist in humans. Carbamyl-adducts are generated in vivo in reaction with isocyanate formed by dissociation from urea or by myeloperoxidase catalysed oxidation of thiocyanate,21 and the chemistry has been well described.39,40 Carbamylation of LDL is suggested to contribute to a higher prevalence of atherosclerosis in uraemic patients11–13 and protein carbamylation has been linked with mortality in patients with end-stage renal disease in two separate studies.19,20 ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope Protein-bound homocitrulline19 and carbamylated serum albumin20 were detected in patients with end-stage renal disease, and the amount of carbamylated protein was associated with mortality in these patients during the follow-up period. Carbamyl-epitopes are found in atherosclerotic plaque and have been shown to co-localize with myeloperoxidase.21 Other epitopes found in atherosclerotic lesions include oxidation-specific lipid epitopes such as MDA-/MAA-adducts. In this study we showed a specific cross-reactivity between carbamyl- and MDA-/MAA-adducts with the monoclonal antibody Fab106. This is an interesting finding because there seems to be no similar chemistry in the modifications. During atherogenesis LDL is accumulated in the arterial wall and exposed to oxidation, leading to formation of OxLDL. Oxidation-specific epitopes are generated when highly reactive lipid peroxidation end-products modify proteins and lipids.1 One of the abundant dialdehydes generated during lipid peroxidation is MDA, which reacts widely with primary amines to create MDAadducts.41 Acetaldehyde, another oxidation product, forms MAA-adducts in the presence of MDA.42,43 CuOxLDL is commonly used in laboratory experiments and contains a wide range of different epitopes, including MDA-adducts and, as the major epitope, PC-head groups of oxidized phospholipids, e.g. 1-palmitoyl-2-(5′-oxovaleroyl)-sn-glycero-3-phosphocholine.44 These epitopes are also found on apoptotic cells8 and atherosclerotic lesions,44 and have been suggested to participate in the immune modulation of atherogenesis. The wide diversity of epitopes on OxLDL and apoptotic cells is recognized by antibodies that may have various effects on atherogenesis. The human Fab106 monoclonal antibody was cloned against carbamyl-LDL and cross-reacted with oxidationspecific MDA-/MAA-epitopes, but not with PC-epitopes. Also, Fab106 bound to epitopes found on apoptotic cells and in the atherosclerotic lesion. Whether Fab106 recognizes oxidation-specific epitopes or carbamyl-epitopes on apoptotic cells remains to be studied. Published data concerning the antibodies to carbamylated LDL or carbamylated proteins are sparse. Here we demonstrated that healthy humans have antibodies to carbamylated LDL and carbamylated proteins, and they are associated with antibodies to oxidation-specific epitopes. We showed an association on both IgM and IgG isotypes, suggesting that the roles of these antibodies could be both natural and adaptive in vivo. We and others have also shown that IgG antibodies to carbamylated proteins are associated with end-stage renal disease22 and rheumatoid arthritis.45,46 In both of these chronic diseases, the patients have a higher incidence of atherosclerotic cardiovascular disease that cannot be explained only by traditional risk factors.47–50 In our previous study we showed IgG and IgM antibodies to carbamylated LDL and carbamylated proteins in patients with end-stage ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

renal disease as well as in healthy subjects.22 The levels of IgG antibodies were significantly higher in uraemic patients than in healthy controls, and also higher in smokers than in non-smokers within the study groups.22 Human IgG and IgA antibodies recognizing carbamylated proteins in the sera of patients with rheumatoid arthritis are shown to predict a more severe disease course in patients that tested negative for antibodies against citrullinated protein antigens, which are used in the prognosis and diagnosis of rheumatoid arthritis.45 Anti-carbamyl antibodies have also been found in patients with arthralgia and are shown to predict the development of rheumatoid arthritis.46 Citrulline and homocitrulline (carbamyllysine) have similar structures; they differ only by one methylene (-CH2) group, homocitrulline being longer.51 A recent study revealed that also citrulline-epitopes are present in atherosclerotic plaque and could be a target for anti-citrullinated protein antibodies in patients with rheumatoid arthritis, and the resulting immune complexes could promote atherogenesis in these patients.52 Antibodies to both citrullinated and carbamylated proteins exist in patients with rheumatoid arthritis.45,46 An interesting question is whether these two chronic diseases, namely rheumatoid arthritis and chronic kidney disease, share some common factors that have an impact on atherogenesis. Both diseases demonstrate antibodies to carbamylated proteins and increased risk for atherosclerotic cardiovascular disease. Our present data suggest that human antibody cross-reactivity between carbamylated and oxidation-specific epitopes may be involved in enhanced atherogenesis in these patients. The role of antibodies in the development of atherosclerosis remains inconclusive. The plasma antibodies and the relation to atherosclerosis have been investigated in several studies that have used different detection methods and different target antigens in the assays, and the results are difficult to compare. Technical issues related to unstandardized methods of measuring these antigen-specific antibodies might lead to divergent results and interpretation. Some early studies have shown that IgG antibodies to OxLDL predict atherosclerotic disease.10 Antibody levels to OxLDL, when measured as a ratio of anti-OxLDL and anti-native LDL, have been shown to be elevated in patients with chronic kidney disease and uraemia.53–56 The anti-OxLDL antibodies could be markers of oxidative stress during atherogenesis. Also, an inverse association of IgG antibodies to OxLDL and coronary artery disease has been shown, and the study suggested that the IgG immunocomplexes formed affect the measurement of free IgG antibodies.57 The IgM antibodies, however, have been shown to be associated with diminished atherosclerosis in several studies.2,3,5 Natural IgM antibodies to oxidation-specific epitopes have been cloned from mice and shown to have atheroprotective properties, e.g. to block the uptake of OxLDL by macrophages.9,58 427

O. Kummu et al. In this study, we cloned from healthy humans a monoclonal Fab antibody with high germline homology and cross-reactivity between carbamylated and oxidation-specific epitopes, and apoptotic cells. These data suggest a natural origin of the Fab106 antibody. Natural antibodies are usually an IgM isotype and share high identity with germline genes. The isotype origin of Fab106 remains unknown because of the technique used in constructing the phage display Fab library. Only Fab variable regions were amplified from the cDNA, whereas antibody isotype-determining constant regions were amplified from the template cloned into the pComb3X vector. Natural IgM antibodies have low affinity, but they are polyreactive and can successfully bind previously unencountered antigens.59,60 IgM antibodies also prime the adaptive IgG immune response; the primary response after antigen encounter is low-affinity IgM followed by the secondary response of high-affinity IgG.59 Additionally, one important property of natural IgM antibodies is to maintain cellular homeostasis and participate in the clearance of apoptotic cells.61 It has been shown that IgM, but not IgG, antibodies bind to late apoptotic PIpositive human Jurkat T cells via the antibody’s Fab domain after trypsin digestion.62 The Fab antibody cloned in this study was originally selected for binding to carbamyl-LDL but was observed to also bind carbamyl-adducts in other proteins and to crossreact with oxidation-specific epitopes, especially with MDA- and MAA-modified proteins. Previously published human monoclonal Fab antibodies binding to MDA-LDL63 and OxLDL64 have been cloned from phage display libraries constructed from atherosclerotic patients. Sequence analysis of these human Fabs against OxLDL shows high homology with germline genes, ability to block the uptake of OxLDL by macrophages and binding to epitopes found in atherosclerotic lesions. The Fab106 antibody to carbamyl-LDL, although cloned from healthy humans, showed similar functional properties to the previously published monoclonal Fab antibodies to OxLDL. The Fab106 shared homology with germline genes, bound to epitopes in atherosclerotic plaque and inhibited the uptake of modified LDL by macrophages, which is a crucial step in foam cell formation and atherogenesis. Nevertheless, Fab106 can be distinguished from the previously characterized human Fabs to OxLDL in its ability to cross-react with carbamylated and MDA-/ MAA-modified oxidation-specific epitopes but not with PC-epitopes. The polyclonal repertoire of human plasma antibodies also recognized the cross-reactivity between carbamylated proteins and MDA-/MAA-adducts supporting the presence of natural antibodies that possess features equivalent with the Fab described in this study.

Conclusions We demonstrated an association between antibodies binding to carbamyl- and oxidation-specific malondialdehyde428

derived adducts in human plasma, and also cloned a human monoclonal Fab antibody with characteristics of a natural antibody and ability to bind both carbamyl- and malondialdehyde-derived epitopes. This cross-reactivity between antibodies binding to carbamylated and oxidation-specific epitopes, and their possible role in explaining the link between atherogenesis and kidney disease will require additional studies.

Acknowledgements We thank Ms Sirpa Rannikko for her excellent technical assistance. This study was supported by the Academy of Finland, the Finnish Foundation for Cardiovascular Research, The Sigrid Juselius Foundation, the Sohlberg Foundation and the Aarne Koskelo Foundation.

Author contributions OK, SPT, PP, JL, MV, CW and SH designed the study, OK, SPT, PP, JL, MV and CW performed the experiments, OK, SPT, PP, JL, MV, CW and SH wrote the paper.

Disclosures The authors declare no conflicting interests.

References 1 H€ orkk€ o S, Binder CJ, Shaw PX, Chang MK, Silverman G, Palinski W, Witztum JL. Immunological responses to oxidized LDL. Free Radic Biol Med 2000; 28: 1771–9. 2 Karvonen J, P€aiv€ansalo M, Kes€aniemi YA, H€ orkk€ o S. Immunoglobulin M type of autoantibodies to oxidized low-density lipoprotein has an inverse relation to carotid artery atherosclerosis. Circulation 2003; 108:2107–12. 3 Su J, Georgiades A, Wu R, Thulin T, de Faire U, Frostegard J. Antibodies of IgM subclass to phosphorylcholine and oxidized LDL are protective factors for atherosclerosis in patients with hypertension. Atherosclerosis 2006; 188:160–6. 4 Carrero JJ, Hua X, Stenvinkel P, Qureshi AR, Heimburger O, Barany P, Lindholm B, Frostegard J. Low levels of IgM antibodies against phosphorylcholine-A increase mortality risk in patients undergoing haemodialysis. Nephrol Dial Transplant 2009; 24:3454– 60. 5 Tsimikas S, Willeit P, Willeit J et al. Oxidation-specific biomarkers, prospective 15-year cardiovascular and stroke outcomes, and net reclassification of cardiovascular events. J Am Coll Cardiol 2012; 60:2218–29. 6 Kyaw T, Tay C, Krishnamurthi S, Kanellakis P, Agrotis A, Tipping P, Bobik A, Toh BH. B1a B lymphocytes are atheroprotective by secreting natural IgM that increases IgM deposits and reduces necrotic cores in atherosclerotic lesions. Circ Res 2011; 109:830–40. 7 Binder CJ, H€ orkk€ o S, Dewan A et al. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL. Nat Med 2003; 9:736–43. 8 Chang MK, Bergmark C, Laurila A, H€ orkk€ o S, Han KH, Friedman P, Dennis EA, Witztum JL. Monoclonal antibodies against oxidized low-density lipoprotein bind to apoptotic cells and inhibit their phagocytosis by elicited macrophages: evidence that oxidation-specific epitopes mediate macrophage recognition. Proc Natl Acad Sci USA 1999; 96:6353–8. 9 H€ orkk€ o S, Bird DA, Miller E et al. Monoclonal autoantibodies specific for oxidized phospholipids or oxidized phospholipid-protein adducts inhibit macrophage uptake of oxidized low-density lipoproteins. J Clin Invest 1999; 103:117–28. 10 Salonen JT, Yl€a-Herttuala S, Yamamoto R et al. Autoantibody against oxidised LDL and progression of carotid atherosclerosis. Lancet 1992; 339:883–7.

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Cross-reactivity of human antibodies to carbamyl-epitope 11 H€ orkk€ o S, Savolainen MJ, Kervinen K, Kes€aniemi YA. Carbamylation-induced alterations in low-density lipoprotein metabolism. Kidney Int 1992; 41:1175–81. 12 H€ orkk€ o S, Huttunen K, Kervinen K, Kes€aniemi YA. Decreased clearance of uraemic and mildly carbamylated low-density lipoprotein. Eur J Clin Invest 1994; 24:105–13. 13 Apostolov EO, Shah SV, Ok E, Basnakian AG. Carbamylated low-density lipoprotein induces monocyte adhesion to endothelial cells through intercellular adhesion mole-

38 Freigang S, H€ orkk€ o S, Miller E, Witztum JL, Palinski W. Immunization of LDL receptor-deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high titers of antibodies to oxidative neoepitopes. Arterioscler Thromb Vasc Biol 1998; 18:1972–82. 39 Stark GR, Stein WH, Moore S. Reactions of the cyanate present in aqueous urea with

cule-1 and vascular cell adhesion molecule-1. Arterioscler Thromb Vasc Biol 2007; 27:826–32. 14 Trepanier DJ, Thibert RJ. Carbamylation of erythrocyte membrane aminophospholipids: an in vitro and in vivo study. Clin Biochem 1996; 29:333–45. 15 Smith WG, Holden M, Benton M, Brown CB. Carbamylated haemoglobin in chronic renal failure. Clin Chim Acta 1988; 178:297–303.

amino acids and proteins. J Biol Chem 1960; 235:3177–81. 40 Stark GR. Reactions of cyanate with functional groups of proteins. 3. Reactions with amino and carboxyl groups. Biochemistry 1965; 4:1030–6. 41 Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 1991; 11:81–128. 42 Tuma DJ, Thiele GM, Xu D, Klassen LW, Sorrell MF. Acetaldehyde and malondialde-

16 Oimomi M, Hatanaka H, Yoshimura Y, Yokono K, Baba S, Taketomi Y. Carbamylation of insulin and its biological activity. Nephron 1987; 46:63–6. 17 Kraus LM, Kraus AP Jr. Carbamoylation of amino acids and proteins in uremia. Kidney Int Suppl 2001; 78:S102–7. 18 Berlyne GM. Carbamylated proteins and peptides in health and in uremia. Nephron 1998; 79:125–30. 19 Koeth RA, Kalantar-Zadeh K, Wang Z, Fu X, Tang WH, Hazen SL. Protein carbamyla-

hyde react together to generate distinct protein adducts in the liver during long-term ethanol administration. Hepatology 1996; 23:872–80. 43 Tuma DJ, Kearley ML, Thiele GM, Worrall S, Haver A, Klassen LW, Sorrell MF. Elucidation of reaction scheme describing malondialdehyde-acetaldehyde-protein adduct formation. Chem Res Toxicol 2001; 14:822–32. orkk€ o S, Miller E, Steinbrecher UP, Powell HC, Curtiss LK, Witztum JL. 44 Palinski W, H€ Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apoli-

tion predicts mortality in ESRD. J Am Soc Nephrol 2013; 24:853–61. 20 Berg AH, Drechsler C, Wenger J et al. Carbamylation of serum albumin as a risk factor for mortality in patients with kidney failure. Sci Transl Med 2013; 5:175ra29. 21 Wang Z, Nicholls SJ, Rodriguez ER et al. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 2007; 13:1176–84. 22 Kummu O, Turunen SP, Wang C et al. Carbamyl adducts on low-density lipoprotein

poprotein E-deficient mice. Demonstration of epitopes of oxidized low density lipoprotein in human plasma. J Clin Invest 1996; 98:800–14. 45 Shi J, Knevel R, Suwannalai P et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc Natl Acad Sci USA 2011; 108:17372–7. 46 Shi J, van de Stadt LA, Levarht EW, Huizinga TW, Toes RE, Trouw LA, van Schaarden-

induce IgG response in LDLR–/– mice and bind plasma autoantibodies in humans under enhanced carbamylation. Antioxid Redox Signal 2013; 19:1047–62. 23 Apostolov EO, Shah SV, Ok E, Basnakian AG. Quantification of carbamylated LDL in human sera by a new sandwich ELISA. Clin Chem 2005; 51:719–28. 24 Bevan RJ, Durand MF, Hickenbotham PT et al. Validation of a novel ELISA for measurement of MDA-LDL in human plasma. Free Radic Biol Med 2003; 35:517–27.

burg D. Anti-carbamylated protein antibodies are present in arthralgia patients and predict the development of rheumatoid arthritis. Arthritis Rheum 2013; 65:911–5. 47 Lindner A, Charra B, Sherrard DJ, Scribner BH. Accelerated atherosclerosis in prolonged maintenance hemodialysis. N Engl J Med 1974; 290:697–701. 48 Apostolov EO, Basnakian AG, Ok E, Shah SV. Carbamylated low-density lipoprotein: nontraditional risk factor for cardiovascular events in patients with chronic kidney dis-

25 Yamazaki K, Bujo H, Taira K, Itou N, Shibasaki M, Takahashi K, Saito Y. Increased circulating malondialdehyde-modified LDL in the patients with familial combined hyperlipidemia and its relation with the hepatic lipase activity. Atherosclerosis 2004; 172:181–7. 26 Miyazaki T, Shimada K, Sato O et al. Circulating malondialdehyde-modified LDL and atherogenic lipoprotein profiles measured by nuclear magnetic resonance spectroscopy in patients with coronary artery disease. Atherosclerosis 2005; 179:139–45.

ease. J Ren Nutr 2012; 22:134–8. 49 del Rincon ID, Williams K, Stern MP, Freeman GL, Escalante A. High incidence of cardiovascular events in a rheumatoid arthritis cohort not explained by traditional cardiac risk factors. Arthritis Rheum 2001; 44:2737–45. 50 Solomon DH, Goodson NJ, Katz JN, Weinblatt ME, Avorn J, Setoguchi S, Canning C, Schneeweiss S. Patterns of cardiovascular risk in rheumatoid arthritis. Ann Rheum Dis 2006; 65:1608–12.

27 Havel RJ, Eder HA, Bragdon JH. The distribution and chemical composition of ultracentrifugally separated lipoproteins in human serum. J Clin Invest 1955; 34:1345–53. 28 Habeeb AF. Determination of free amino groups in proteins by trinitrobenzenesulfonic acid. Anal Biochem 1966; 14:328–36. 29 Turunen SP, Kummu O, Harila K, Veneskoski M, Soliymani R, Baumann M, Pussinen PJ, H€ orkk€ o S. Recognition of Porphyromonas gingivalis gingipain epitopes by natural

51 Trouw LA, Huizinga TW, Toes RE. Autoimmunity in rheumatoid arthritis: different antigens – common principles. Ann Rheum Dis 2013; 72(Suppl. 2):ii132–6. 52 Sokolove J, Sharpe O, Brennan M et al. Citrullination within the atherosclerotic plaque: a potential target for the anti-citrullinated protein antibody response in rheumatoid arthritis. Arthritis Rheum 2013; 65:1719–24. 53 Bellazzi R, Maggi E, Bellomo G et al. Low-density lipoprotein oxidation and antioxi-

IgM binding to malondialdehyde modified low-density lipoprotein. PLoS ONE 2012; 7: e34910. 30 Andris-Widhopf J, Steinberger P, Fuller R, Rader C, Barbas CF 3rd. Generation of antibody libraries: PCR amplification and assembly of light- and heavy-chain coding sequences. In: Barbas CF3rd, Burton DR, Scott JK, Silverman GJ, eds. Phage Display: A Laboratory Manual. New York: Cold Spring Harbor Laboratory Press, 2001:9.1–9.113. 31 Brochet X, Lefranc MP, Giudicelli V. IMGT/V-QUEST: the highly customized and inte-

dized LDL antibodies in peritoneal dialysis patients. Adv Perit Dial 1993; 9:312–7. 54 Maggi E, Bellazzi R, Falaschi F et al. Enhanced LDL oxidation in uremic patients: an additional mechanism for accelerated atherosclerosis? Kidney Int 1994; 45:876–83. 55 Maggi E, Bellazzi R, Gazo A, Seccia M, Bellomo G. Autoantibodies against oxidativelymodified LDL in uremic patients undergoing dialysis. Kidney Int 1994; 46:869–76. 56 Maggi E, Chiesa R, Melissano G, Castellano R, Astore D, Grossi A, Finardi G, Bellomo G. LDL oxidation in patients with severe carotid atherosclerosis. A study of in vitro and

grated system for IG and TR standardized V-J and V-D-J sequence analysis. Nucleic Acids Res 2008; 36:W503–8. 32 Giudicelli V, Brochet X, Lefranc MP. IMGT/V-QUEST: IMGT standardized analysis of the immunoglobulin (IG) and T cell receptor (TR) nucleotide sequences. Cold Spring Harb Protoc 2011; 2011:695–715. o S. 33 Wang C, Turunen SP, Kummu O, Veneskoski M, Lehtim€aki J, Nissinen AE, H€ orkk€

in vivo oxidation markers. Arterioscler Thromb 1994; 14:1892–9. 57 Lopes-Virella MF, Virella G, Orchard TJ, Koskinen S, Evans RW, Becker DJ, Forrest KY. Antibodies to oxidized LDL and LDL-containing immune complexes as risk factors for coronary artery disease in diabetes mellitus. Clin Immunol 1999; 90:165–72. 58 Tuominen A, Miller YI, Hansen LF, Kes€aniemi YA, Witztum JL, H€ orkk€ o S. A natural antibody to oxidized cardiolipin binds to oxidized low-density lipoprotein, apoptotic

Natural antibodies of newborns recognize oxidative stress-related malondialdehyde acetaldehyde adducts on apoptotic cells and atherosclerotic plaques. Int Immunol 2013; 25:575–87. 34 Veneskoski M, Turunen SP, Kummu O, Nissinen A, Rannikko S, Levonen AL, H€ orkk€ o S. Specific recognition of malondialdehyde and malondialdehyde acetaldehyde adducts on oxidized LDL and apoptotic cells by complement anaphylatoxin C3a. Free Radic Biol Med 2011; 51:834–43.

cells, and atherosclerotic lesions. Arterioscler Thromb Vasc Biol 2006; 26:2096–102. 59 Manson JJ, Mauri C, Ehrenstein MR. Natural serum IgM maintains immunological homeostasis and prevents autoimmunity. Springer Semin Immunopathol 2005; 26:425–32. 60 Binder CJ. Natural IgM antibodies against oxidation-specific epitopes. J Clin Immunol 2010; 30(Suppl. 1):S56–60. 61 Kaveri SV, Silverman GJ, Bayry J. Natural IgM in immune equilibrium and harnessing their therapeutic potential. J Immunol 2012; 188:939–45.

35 Yamaguchi Y, Kunitomo M, Haginaka J. Assay methods of modified lipoproteins in plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 781:313–30. 36 Steinbrecher UP, Fisher M, Witztum JL, Curtiss LK. Immunogenicity of homologous low density lipoprotein after methylation, ethylation, acetylation, or carbamylation: generation of antibodies specific for derivatized lysine. J Lipid Res 1984; 25:1109–16. 37 Gonen B, Fallon JJ, Baker SA. Immunogenicity of malondialdehyde-modified low

62 Kim SJ, Gershov D, Ma X, Brot N, Elkon KB. I-PLA(2) activation during apoptosis promotes the exposure of membrane lysophosphatidylcholine leading to binding by natural immunoglobulin M antibodies and complement activation. J Exp Med 2002; 196:655–65. 63 Shaw PX, H€ orkk€ o S, Tsimikas S, Chang MK, Palinski W, Silverman GJ, Chen PP, Witztum JL. Human-derived anti-oxidized LDL autoantibody blocks uptake of oxidized

density lipoproteins. studies with monoclonal antibodies. Atherosclerosis 1987; 65:265–72.

LDL by macrophages and localizes to atherosclerotic lesions in vivo. Arterioscler Thromb Vasc Biol 2001; 21:1333–9.

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

429

O. Kummu et al. 64 Jeon YE, Seo CW, Yu ES, Lee CJ, Park SG, Jang YJ. Characterization of human monoclonal autoantibody Fab fragments against oxidized LDL. Mol Immunol 2007; 44:827– 36.

Supporting Information

Data S1. Methods. Figure S1. Fab binding to apoptotic Jurkat T cells. Figure S2. Immunohistochemical staining of normal C57BL/6 mouse heart aortic origin cross-section with the anti-carbamyl Fab106 antibody.

Additional Supporting Information may be found in the online version of this article:

430

ª 2013 John Wiley & Sons Ltd, Immunology, 141, 416–430

Human monoclonal Fab and human plasma antibodies to carbamyl-epitopes cross-react with malondialdehyde-adducts.

Oxidized low-density lipoprotein (OxLDL) plays a crucial role in the development of atherosclerosis. Carbamylated LDL has been suggested to promote at...
692KB Sizes 0 Downloads 0 Views