Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884754

ABSTRACT

Chronic inflammation plays a critical role in the pathogenesis of atherosclerosis. Currently, the mechanism(s) by which inflammation contributes to this disease are not entirely understood. Inflammation is known to induce oxidative stress, which can lead to lipid peroxidation. Lipid peroxidation can result in the production of reactive by-products that can oxidatively modify macromolecules including DNA, proteins, and lipoproteins. A major reactive by-product of lipid peroxidation is malondialdehyde (MDA). MDA can subsequently break down to form acetaldehyde (AA). These two aldehydes can covalently interact with the epsilon (ε)-amino group of lysines within proteins and lipoproteins leading to the formation of extremely stable, highly immunogenic malondialdehyde/acetaldehyde adducts (MAA-adducts). The aim of this study was to investigate the inflammatory response to MAA-modified human serum albumin (HSA-MAA) and low-density lipoprotein (LDL-MAA). We found that animals injected with LDL-MAA generate antibodies specific to MAA-adducts. The level of anti-MAA antibodies were further increased in an animal model of atherosclerosis fed a Western diet. An animal model that combined both high fat diet and immunization of MAA-modified protein resulted in a dramatic increase in antibodies to MAA-adducts and vascular fat accumulation compared with controls. In vitro exposure of endothelial cells and macrophages to MAA-modified proteins resulted in increased fat accumulation as well as increased expression of adhesion molecules and pro-inflammatory cytokines. The expression of cytokines varied between the different cell lines and was unique to the individual modified proteins. The results of these studies demonstrate that different MAA-modified proteins elicit unique responses in different cell types. Additionally, the presence of MAA-modified proteins appears to modulate cellular metabolism leading to increased accumulation of triglycerides and further progression of the inflammatory response.


Subject(s)
Inflammation/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Protein Processing, Post-Translational , Serum Albumin, Human/immunology , Serum Albumin, Human/metabolism , Acetaldehyde/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Humans , Inflammation/etiology , Inflammation/immunology , Lipid Metabolism/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley
2.
Molecules ; 26(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34771115

ABSTRACT

Isoaspartate (isoAsp) is a damaging amino acid residue formed in proteins mostly as a result of spontaneous deamidation of asparaginyl residues. An association has been found between isoAsp in human serum albumin (HSA) and Alzheimer's disease (AD). Here we report on a novel monoclonal antibody (mAb) 1A3 with excellent specificity to isoAsp in the functionally important domain of HSA. Based on 1A3 mAb, an indirect enzyme-linked immunosorbent assay (ELISA) was developed, and the isoAsp occupancy in 100 healthy plasma samples was quantified for the first time, providing the average value of (0.74 ± 0.13)%. These results suggest potential of isoAsp measurements for supplementary AD diagnostics as well as for assessing the freshness of stored donor blood and its suitability for transfusion.


Subject(s)
Immunoassay/methods , Isoaspartic Acid/chemistry , Serum Albumin, Human/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Base Sequence , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Humans , Isoaspartic Acid/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Sensitivity and Specificity , Serum Albumin, Human/genetics , Serum Albumin, Human/immunology , Tandem Mass Spectrometry
3.
Eur J Pharmacol ; 904: 174152, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33964292

ABSTRACT

Antigen-binding fragments (Fabs) are preferred alternatives to antibodies for medical application, whereas their short half-lives limit therapeutic effectiveness. Albumin binding domain (ABD) with high affinity for albumin possesses a great potential in enhancing in vivo performance of biotherapeutics. In this study, to mitigate the poor pharmacokinetics of adalimumab Fab targeting tumor necrosis factor-α (TNFα), an ABD fusion strategy was applied innovatively using GA3, ABD035, ABD094 and ABDCon with high affinities for albumin. The prokaryotic expression, bioactivities and half-lives of those novel Fab-ABD fusions were investigated in vitro and in vivo. All Fab-ABD fusions were successfully purified, and they retained similar TNFα-binding activities with the unmodified Fab control, also presented high affinities for human/mouse serum albumin (HSA/MSA). Additionally, the simultaneous binding of the difunctional Fab-ABD fusions to TNFα and albumin was verified, and ABD fused to Fab neither interfered with Fab-TNFα binding nor impaired the association between Fc fragment of IgG receptor and transporter (FcRn) and albumin. Based on the highest binding affinity for HSA and maximal yield, Fab-ABDCon was selected for further evaluation. Fab-ABDCon showed similar thermostability with the Fab control and robust stability in human and mouse plasma. Most notably, the pharmacokinetics of Fab-ABDCon in mice was significantly improved with a 22-fold longer plasma half-life (28.2 h) compared with that of Fab control (1.31 h), which have contributed to its satisfactory therapeutic efficacy in murine TNFα-induced hepatonecrosis model. Thus, Fab-ABDCon could be a promising long-acting candidate suitable for drug development targeting TNFα-mediated inflammatory disease.


Subject(s)
Adalimumab/biosynthesis , Adalimumab/pharmacology , Albumins/metabolism , Anti-Inflammatory Agents/pharmacology , Immunoglobulin Fab Fragments/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Albumins/immunology , Animals , Anti-Inflammatory Agents/immunology , Cell Death/drug effects , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/prevention & control , Drug Design , Female , Galactosamine/administration & dosage , Galactosamine/toxicity , Half-Life , Humans , Hydrogen-Ion Concentration , Immunoglobulin Fab Fragments/pharmacology , Injections, Intraperitoneal , Mice, Inbred BALB C , Necrosis/chemically induced , Necrosis/prevention & control , Protein Binding/genetics , Protein Domains/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Serum Albumin, Human/immunology , Serum Albumin, Human/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/toxicity
4.
Inflammation ; 44(4): 1620-1628, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33751358

ABSTRACT

The currently used anti-cytokine therapeutic antibodies cannot selectively neutralize pathogenic cytokine signalling that cause collateral damage to protective signalling cascades. The single domain chain firstly discovered in Camelidae displays fully functional ability in antigen-binding against variable targets, which has been seemed as attractive candidates for the next-generation biologic drug study. In this study, we established a simple prokaryotic expression system for a dual target-directed single domain-based fusion protein against the interleukin-6 receptor and human serum, albumin, the recombinant anti-IL-6R fusion protein (VHH-0031). VHH-0031 exhibited potent anti-inflammatory effects produced by LPS on cell RAW264.7, where the major cytokines and NO production were downregulated after 24 h incubation with VHH-0031 in a dose-dependent manner. In vivo, VHH-0031 presented significant effects on the degree reduction of joint swelling in the adjuvant-induced arthritis (AIA) rat, having a healthier appearance compared with the dexamethasone. The expression level of JNK protein in the VHH-0031 group was significantly decreased, demonstrating that VHH-0031 provides a low-cost and desirable effect in the treatment of more widely patients.


Subject(s)
Anti-Inflammatory Agents/immunology , Arthritis, Experimental/drug therapy , Interleukin-6/antagonists & inhibitors , Serum Albumin, Human/antagonists & inhibitors , Single-Domain Antibodies/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Antibody Specificity , Arthritis, Experimental/immunology , Cytokines/metabolism , DNA, Complementary/genetics , Dexamethasone/therapeutic use , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Humans , Interleukin-6/immunology , Lipopolysaccharides/toxicity , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Kinase 4/genetics , Mice , Models, Molecular , Molecular Targeted Therapy , Nitric Oxide/metabolism , Protein Conformation , RAW 264.7 Cells , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Serum Albumin, Human/immunology , Single-Domain Antibodies/genetics
5.
MAbs ; 13(1): 1893888, 2021.
Article in English | MEDLINE | ID: mdl-33691596

ABSTRACT

Albumin has a serum half-life of 3 weeks in humans. This feature can be used to improve the pharmacokinetics of shorter-lived biologics. For instance, an albumin-binding domain (ABD) can be used to recruit albumin. A prerequisite for such design is that the ABD-albumin interaction does not interfere with pH-dependent binding of albumin to the human neonatal Fc receptor (FcRn), as FcRn acts as the principal regulator of the half-life of albumin. Thus, there is a need to know how ABDs act in the context of fusion partners and human FcRn. Here, we studied the binding and transport properties of human immunoglobulin A1 (IgA1), fused to a Streptococcus protein G-derived engineered ABD, in in vitro and in vivo systems harboring human FcRn. IgA has great potential as a therapeutic protein, but its short half-life is a major drawback. We demonstrate that ABD-fused IgA1 binds human FcRn pH-dependently and is rescued from cellular degradation in a receptor-specific manner in the presence of albumin. This occurs when ABD is fused to either the light or the heavy chain. In human FcRn transgenic mice, IgA1-ABD in complex with human albumin, gave 4-6-fold extended half-life compared to unmodified IgA1, where the light chain fusion showed the longest half-life. When the heavy chain-fused protein was pre-incubated with an engineered human albumin with improved FcRn binding, cellular rescue and half-life was further enhanced. Our study reveals how an ABD, which does not interfere with albumin binding to human FcRn, may be used to extend the half-life of IgA.Abbreviations: ABD - Albumin binding domain, ADA - anti-drug-antibodies, ADCC - Antibody-dependent cellular cytotoxicity, ELISA - Enzyme-linked Immunosorbent assay, FcαRI - Fcα receptor, FcγR - Fcγ receptor, FcRn - The neonatal Fc receptor, GST - Glutathione S-transferase, HC - Heavy chain, HERA - Human endothelial cell-based recycling assay, Her2 - Human epidermal growth factor 2, HMEC - Human microvascular endothelial cells, IgG - Immunoglobulin G, IgA - Immunoglobulin A, LC - Light chain, QMP - E505Q/T527M/K573P, WT - Wild type.


Subject(s)
Bacterial Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoglobulin A/metabolism , Receptors, Fc/metabolism , Serum Albumin, Human/metabolism , Animals , Antibody Specificity , Bacterial Proteins/genetics , Bacterial Proteins/immunology , HEK293 Cells , Half-Life , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Mice, Transgenic , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Receptors, Fc/genetics , Receptors, Fc/immunology , Recombinant Fusion Proteins/metabolism , Serum Albumin, Human/genetics , Serum Albumin, Human/immunology
6.
Medicine (Baltimore) ; 100(10): e24925, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33725853

ABSTRACT

ABSTRACT: Ileocolonoscopy is currently recognized as the gold standard for evaluating mucosal healing in patients with Crohn disease (CD). However, the ideal noninvasive marker to assess mucosal healing instead of invasive ileocolonoscopy is not available. This study aimed to determine the correlations between the mucosal healing and serological optimizing markers in CD.This retrospective study consecutively included 62 CD patients with 137 hospitalizations between March 2014 and March 2020. On the basis of the Simple Endoscopic Score for Crohn's disease (SES-CD), the CD patients were divided into mucosal healing group (SES-CD ≤ 2) and nonmucosal healing group (SES-CD > 2). We collected the results of ileocolonoscopy examination and inflammatory markers and then serological optimizing markers, including C-reactive protein/albumin ratio (CRP/ALB), platelet/albumin ratio (PLT/ALB), neutrophil-lymphocyte ratio (NLR), and platelet-lymphocyte ratio (PLR) were calculated. The control group consisted of 50 healthy volunteers in the corresponding period.We found that CRP/ALB, PLT/ALB, NLR, and PLR were correlated with the mucosal healing of CD, and the correlation of CRP/ALB with the mucosal healing was the highest (r = -0.64). Receiver operating characteristic (ROC) analysis showed that the area under the curve (AUC) of CRP/ALB (0.87) was higher than NLR (0.69), PLR (0.72), and PLT/ALB (0.81). In the efficacy of assessing the mucosal healing in CD, the sensitivity of CRP/ALB, NLR, PLR, and PLT/ALB were 91.1%, 83.9%, 73.2%, and 73.2%, respectively, and the specificity was 76.5%, 46.9%, 64.2%, and 75.3%, respectively.CRP/ALB was the most appropriate marker to assess CD mucosal healing among the serological optimizing markers.


Subject(s)
C-Reactive Protein/analysis , Crohn Disease/diagnosis , Intestinal Mucosa/immunology , Serum Albumin, Human/analysis , Adult , Biomarkers/blood , C-Reactive Protein/immunology , Colon/diagnostic imaging , Colon/immunology , Colonoscopy , Crohn Disease/blood , Crohn Disease/immunology , Female , Humans , Ileum/diagnostic imaging , Ileum/immunology , Intestinal Mucosa/diagnostic imaging , Male , Retrospective Studies , Serum Albumin, Human/immunology , Severity of Illness Index , Young Adult
7.
Medicine (Baltimore) ; 100(10): e25119, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33725909

ABSTRACT

ABSTRACT: The purpose of the present study was to investigate the efficacy of perioperative oral managements (POMs) on perioperative nutritional conditions in patients undergoing surgery with general anesthesia. Medical records were retrospectively reviewed and the effects of POMs were investigated based on a large number of cases using a multicenter analysis. The profile of serum albumin levels was assessed and compared between patients with and without POMs using the multivariate analysis. Seventeen Eleven thousand and one hundred sixty patients (4,873 males and 6,287 females) were reviewed. Of these, 2710 patients (24.3%) had undergone POMs. The results of a multivariate analysis revealed the significant positive effect of POMs on perioperative serum albumin level (change between at admission and discharge, (Estimate: 0.022, standard error: 0.012, P < .0001). Patient gender, age, surgical site, performance status, the American Society of Anesthesiologists (ASA) physical status classification, operation time, amount of blood loss, and serum albumin level at admission were also significant predictors. Adjusted multivariate analysis of the effects of POMs on perioperative change of serum albumin level in all subjects reveled the significance of POMs intervention (estimate: 0.022, standard error: 0.012, P < .0001). These results suggest that POMs exerts significant positive effects on perioperative serum albumin levels in patients underwent surgery under general anesthesia.


Subject(s)
Anesthesia, General/adverse effects , Oral Hygiene , Perioperative Care/methods , Serum Albumin, Human/analysis , Surgical Procedures, Operative/adverse effects , Adult , Female , Humans , Japan , Male , Middle Aged , Perioperative Period , Postoperative Complications/immunology , Postoperative Complications/prevention & control , Retrospective Studies , Risk Factors , Serum Albumin, Human/immunology , Treatment Outcome
8.
Front Immunol ; 11: 507092, 2020.
Article in English | MEDLINE | ID: mdl-33363530

ABSTRACT

The pathogenicity of group A Streptococcus (GAS) is mediated by direct bacterial invasivity and toxin-associated damage. Among the extracellular products, the exotoxin streptolysin O (SLO) is produced by almost all GAS strains. SLO is a pore forming toxin (PFT) hemolitically active and extremely toxic in vivo. Recent evidence suggests that human serum albumin (HSA), the most abundant protein in plasma, is a player in the innate immunity "orchestra." We previously demonstrated that HSA acts as a physiological buffer, partially neutralizing Clostridioides difficile toxins that reach the bloodstream after being produced in the colon. Here, we report the in vitro and ex vivo capability of HSA to neutralize the cytotoxic and hemolytic effects of SLO. HSA binds SLO with high affinity at a non-conventional site located in domain II, which was previously reported to interact also with C. difficile toxins. HSA:SLO recognition protects HEp-2 and A549 cells from cytotoxic effects and cell membrane permeabilization induced by SLO. Moreover, HSA inhibits the SLO-dependent hemolytic effect in red blood cells isolated from healthy human donors. The recognition of SLO by HSA may have a significant protective role in human serum and sustains the emerging hypothesis that HSA is an important constituent of the innate immunity system.


Subject(s)
Erythrocytes/immunology , Hemolysis/immunology , Immunity, Innate , Serum Albumin, Human/immunology , Streptococcus pyogenes/immunology , Streptolysins/immunology , A549 Cells , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Humans , Serum Albumin, Human/chemistry , Streptococcus pyogenes/chemistry , Streptolysins/chemistry
9.
Int J Mol Sci ; 21(19)2020 Oct 03.
Article in English | MEDLINE | ID: mdl-33023043

ABSTRACT

In some instances, when chemicals bind to proteins, they have the potential to induce a conformational change in the macromolecule that may misfold in such a way that makes it similar to the various target sites or act as a neoantigen without conformational change. Cross-reactivity then can occur if epitopes of the protein share surface topology to similar binding sites. Alteration of peptides that share topological equivalence with alternating side chains can lead to the formation of binding surfaces that may mimic the antigenic structure of a variant peptide or protein. We investigated how antibodies made against thyroid target sites may bind to various chemical-albumin compounds where binding of the chemical has induced human serum albumin (HSA) misfolding. We found that specific monoclonal or polyclonal antibodies developed against thyroid-stimulating hormone (TSH) receptor, 5'-deiodinase, thyroid peroxidase, thyroglobulin, thyroxine-binding globulin (TBG), thyroxine (T4), and triiodothyronine (T3) bound to various chemical HSA compounds. Our study identified a new mechanism through which chemicals bound to circulating serum proteins lead to structural protein misfolding that creates neoantigens, resulting in the development of antibodies that bind to key target proteins of the thyroid axis through protein misfolding. For demonstration of specificity of thyroid antibody binding to various haptenic chemicals bound to HSA, both serial dilution and inhibition studies were performed and proportioned to the dilution. A significant decline in these reactions was observed. This laboratory analysis of immune reactivity between thyroid target sites and chemicals bound to HSA antibodies identifies a new mechanism by which chemicals can disrupt thyroid function.


Subject(s)
Antibodies/immunology , Blood Proteins/immunology , Haptens/immunology , Serum Albumin, Human/immunology , Antibodies/genetics , Antibody Specificity/immunology , Blood Proteins/chemistry , Epitopes/immunology , Haptens/genetics , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/immunology , Protein Binding/immunology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Serum Albumin, Human/chemistry , Thyroid Gland/immunology , Triiodothyronine/genetics , Triiodothyronine/immunology
10.
MAbs ; 12(1): 1832861, 2020.
Article in English | MEDLINE | ID: mdl-33073698

ABSTRACT

Two-pore physiologically-based pharmacokinetics (PBPK) for biologics describes the tissue distribution and elimination kinetics of soluble proteins as a function of their hydrodynamic radius and the physiological properties of the organs. Whilst many studies have been performed in rodents to parameterize the PBPK framework in terms of organ-specific lymph flow rates, similar validation in humans has been limited. This is mainly due to the paucity of the tissue distribution time course data for biologics that is not distorted by target-related binding. Here, we demonstrate that a PBPK model based on rodent data provided good to satisfactory extrapolation to the tissue distribution time course of 89Zr-labeled albumin-binding domain antibody (AlbudAb™) GSK3128349 in healthy human volunteers, including correct prediction of albumin-like plasma half-life, volume of distribution, and extravasation half-life. The AlbudAb™ used only binds albumin, and hence it also provides information about the tissue distribution kinetics and turnover of that ubiquitous and multifunctional plasma protein.


Subject(s)
Antibodies, Monoclonal , Models, Biological , Radioisotopes , Serum Albumin, Human/immunology , Zirconium , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Humans , Radioisotopes/chemistry , Radioisotopes/pharmacokinetics , Radioisotopes/pharmacology , Tissue Distribution , Zirconium/chemistry , Zirconium/pharmacokinetics , Zirconium/pharmacology
11.
Int J Mol Sci ; 21(17)2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32825512

ABSTRACT

Osteoarthritis (OA) is associated with cartilage breakdown, brought about by ADAMTS-5 mediated aggrecan degradation followed by MMP-derived aggrecan and type II collagen degradation. We investigated a novel anti-ADAMTS-5 inhibiting Nanobody® (M6495) on cartilage turnover ex vivo. Bovine cartilage (BEX, n = 4), human osteoarthritic - (HEX, n = 8) and healthy-cartilage (hHEX, n = 1) explants and bovine synovium and cartilage were cultured up to 21 days in medium alone (w/o), with pro-inflammatory cytokines (oncostatin M (10 ng/mL) + TNFα (20 ng/mL) (O + T), IL-1α (10 ng/mL) or oncostatin M (50 ng/mL) + IL-1ß (10 ng/mL)) with or without M6495 (1000-0.46 nM). Cartilage turnover was assessed in conditioned medium by GAG (glycosaminoglycan) and biomarkers of ADAMTS-5 driven aggrecan degradation (huARGS and exAGNxI) and type II collagen degradation (C2M) and formation (PRO-C2). HuARGS, exAGNxI and GAG peaked within the first culture week in pro-inflammatory stimulated explants. C2M peaked from day 14 by O + T and day 21 in co-culture experiments. M6495 dose dependently decreased huARGS, exAGNxI and GAG after pro-inflammatory stimulation. In HEX C2M was dose-dependently reduced by M6495. M6495 showed no effect on PRO-C2. M6495 showed cartilage protective effects by dose-dependently inhibiting ADAMTS-5 mediated cartilage degradation and inhibiting overall cartilage deterioration in ex vivo cartilage cultures.


Subject(s)
ADAMTS5 Protein/antagonists & inhibitors , Cartilage, Articular/drug effects , Cartilage, Articular/physiopathology , Single-Domain Antibodies/pharmacology , ADAMTS5 Protein/immunology , ADAMTS5 Protein/metabolism , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Cattle , Coculture Techniques , Collagen Type II/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Humans , Male , Middle Aged , Oncostatin M/pharmacology , Organ Culture Techniques , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Serum Albumin, Human/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Synovial Membrane/cytology
12.
FASEB J ; 34(6): 8574-8595, 2020 06.
Article in English | MEDLINE | ID: mdl-32369236

ABSTRACT

Although effective immunological diagnostic systems for autoimmune bullous skin diseases (AIBD) have been established, there are still unidentified cutaneous autoantigens. The purpose of this study is to investigative whether anti-human serum albumin (HSA) autoantibodies exist in AIBD sera and their potential pathogenesis. By immunoprecipitation-immunoblotting, immunofluorescence assay, anti-HSA autoantibodies could be detected in AIBD sera; by ELISAs, positive rates of AIBD sera for IgG and IgA anti-HSA autoantibodies were 29% and 34%, respectively. The IgG anti-HSA autoantibodies in ABID sera recognized a number of HSA antigen epitopes and therefore a polyclonal antibody against HSA were next employed to study its pathogenesis. In vitro cell and tissue culture models, anti-HSA antibody could influence DNA damage-related signaling proteins, via activation of phospho-p38 signaling pathway. This is the first report that an autoantibody may influence DNA damage-related signaling proteins. Statistical analyses also proved that anti-HSA autoantibodies were positively correlated with various known autoantibodies and clinical features of ABID patients. In summary, IgG and IgA autoantibodies to HSA may have diagnosis values for AIBD. DNA damage-related signaling proteins might be involved in the pathogenic role of anti-HSA autoantibodies in AIBD. Phospho-p38 signaling pathway is a potential target for treatment of AIBD positive for serum anti-HSA autoantibodies.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Serum Albumin, Human/immunology , Skin Diseases, Vesiculobullous/immunology , Adult , Aged , Aged, 80 and over , Autoantigens/immunology , Cell Line , Cell Line, Tumor , Child , DNA Damage/immunology , Epitopes/immunology , Female , Humans , Immunoblotting/methods , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Male , Middle Aged , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/immunology
13.
PLoS One ; 15(4): e0231101, 2020.
Article in English | MEDLINE | ID: mdl-32302339

ABSTRACT

Mast cells and basophils are central players in allergic reactions triggered by immunoglobulin E (IgE). They have intracellular granules containing allergic mediators (e.g., histamine, serotonin, inflammatory cytokines, proteases and ß-hexosaminidase), and stimulation by IgE-allergen complex leads to the release of such allergic mediators from the granules, that is, degranulation. Mast cells are residents of mucosal surfaces, including those of nasal and oral cavities, and play an important role in the innate defense system. Members of the mitis group streptococci such as Streptococcus oralis, are primary colonizers of the human oral cavity. They produce hydrogen peroxide (H2O2) as a by-product of sugar metabolism. In this study, we investigated the effects of streptococcal infection on RBL-2H3 mast cell/basophil cell line. Infection by oral streptococci did not induce degranulation of the cells. Stimulation of the RBL-2H3 cells with anti-dinitrophenol (DNP) IgE and DNP-conjugated human serum albumin triggers degranulation with the release of ß-hexosaminidase. We found that S. oralis and other mitis group streptococci inhibited the IgE-triggered degranulation of RBL-2H3 cells. Since mitis group streptococci produce H2O2, we examined the effect of S. oralis mutant strain deficient in producing H2O2, and found that they lost the ability to suppress the degranulation. Moreover, H2O2 alone inhibited the IgE-induced degranulation. Subsequent analysis suggested that the inhibition of degranulation was related to the cytotoxicity of streptococcal H2O2. Activated RBL-2H3 cells produce interleukin-4 (IL-4); however, IL-4 production was not induced by streptococcal H2O2. Furthermore, an in vivo study using the murine pollen-induced allergic rhinitis model suggested that the streptococcal H2O2 reduces nasal allergic reaction. These findings reveal that H2O2 produced by oral mitis group streptococci inhibits IgE-stimulated degranulation by inducing cell death. Consequently, streptococcal H2O2 can be considered to modulate the allergic reaction in mucosal surfaces.


Subject(s)
Allergens/metabolism , Hypersensitivity/immunology , Immunoglobulin E/immunology , Streptococcal Infections/drug therapy , Allergens/immunology , Animals , Basophils/immunology , Basophils/microbiology , Basophils/pathology , Cell Degranulation/immunology , Cell Survival/immunology , Dinitrophenols/pharmacology , Humans , Hydrogen Peroxide/metabolism , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Immunoglobulin E/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Mast Cells/immunology , Mast Cells/microbiology , Mast Cells/pathology , Mice , Plant Extracts/chemistry , Plant Extracts/pharmacology , Serum Albumin, Human/immunology , Serum Albumin, Human/metabolism , Streptococcal Infections/immunology , Streptococcus oralis/immunology , Streptococcus oralis/pathogenicity , Sugars/metabolism
14.
Biochem Biophys Res Commun ; 526(4): 941-946, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32284170

ABSTRACT

Human serum albumin (HSA) has been used to extend the serum half-lives of various protein therapeutics through genetic fusion because HSA exhibits an exceptionally long circulation time as a result of neonatal Fc receptor (FcRn)-mediated recycling. As another serum half-life extender, the human antibody Fab SL335 that strongly binds HSA was developed. When SL335 was fused to a protein therapeutic, SL335 was shown to prolong the half-life of the drug. Despite the significance of SL335-HSA binding in the extension of drug circulation time, it remains unclear how SL335 interacts with HSA at a molecular structural level. To reveal the structural basis of HSA recognition by SL335, we determined the crystal structure of the SL335-HSA complex at a resolution of 2.95 Å. SL335 binds HSA at a 1:1 stoichiometry. SL335 uses the exposed loops of its heavy and light chains to specifically recognize the IIa and IIb subdomains of HSA. The SL335 epitope is located on the opposite side of the FcRn-binding site and does not overlap with it, suggesting that SL335 extends the serum half-lives of itself and its fusion partner through an FcRn-dependent recycling mechanism.


Subject(s)
Antibodies/immunology , Immunoglobulin Fab Fragments/immunology , Recombinant Proteins/blood , Recombinant Proteins/therapeutic use , Serum Albumin, Human/chemistry , Serum Albumin, Human/immunology , Antibodies/chemistry , Cross Reactions , Half-Life , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Models, Molecular , Protein Binding , Receptors, Fc/metabolism
15.
Int Immunopharmacol ; 83: 106433, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32224441

ABSTRACT

BACKGROUND/OBJECTIVE: Malondialdehyde-acetaldehyde adducts (MAA) act as potent immune adjuvants and co-localize with citrullinated antigens in tissues effected by rheumatoid arthritis (RA). We sought to examine the role of MAA-adducts in promoting RA-related autoimmunity and inflammation. METHODS: DBA/J1 mice were immunized with human serum albumin (HSA), HSA-MAA, citrullinated HSA (HSA-Cit), or HSA-MAA-Cit with subsequent measurement of serum anti-citrullinated protein antibody (ACPA) and anti-Cit T cell responses. Cellular binding of the same antigens was examined using THP-1 monocytes and Chinese Hamster Ovary (CHO) cells transfected with specific scavenger receptors (SRs: TLR4, SR-B2, SREC-1). The effects of these antigens on THP-1 activation were then examined by quantifying plate adherence, pro-inflammatory (TNFα, IL-1ß, IL-10) cytokine release, and SR (CD14, SR-B2)/co-stimulatory molecule (CD80, HLA-DR) expression. Comparisons were completed using one-way ANOVA with Tukey's post-hoc test. RESULTS: Mice immunized with co-modified HSA produced significantly higher ACPA concentrations than all other groups whereas T cell responses to citrullinated proteins were highest following immunization with HSA-MAA. Both transfected CHO and THP-1 cells demonstrated significantly higher binding of HSA-MAA-Cit vs. HSA or HSA-Cit. THP-1 cells exposed to HSA-MAA-Cit expressed significantly higher concentrations of TNFα, IL-1ß, and IL-10 vs. all other groups. Furthermore, THP-1 cells demonstrated significantly increased plate adherence and higher expression of CD14, SR-B2, and HLA-DR following incubation with HSA-MAA-Cit vs. HSA or HSA-Cit. CONCLUSION: These studies demonstrate that MAA-adduction of citrullinated antigen greatly enhances immune and cellular responses, potentially acting as a key co-factor in RA pathogenesis.


Subject(s)
Acetaldehyde/immunology , Anti-Citrullinated Protein Antibodies/blood , Citrullination/immunology , Malondialdehyde/immunology , Acetaldehyde/chemistry , Adjuvants, Immunologic/chemistry , Animals , Anti-Citrullinated Protein Antibodies/immunology , Arthritis, Rheumatoid/immunology , CHO Cells , Cricetulus , Cytokines/metabolism , Humans , Immunogenicity, Vaccine , Inflammation/metabolism , Male , Malondialdehyde/chemistry , Mice, Inbred DBA , Monocytes/metabolism , Receptors, Scavenger/metabolism , Serum Albumin, Human/chemistry , Serum Albumin, Human/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , THP-1 Cells
16.
Cell Immunol ; 347: 104020, 2020 01.
Article in English | MEDLINE | ID: mdl-31767118

ABSTRACT

High-mobility group box 1 (HMGB1) concentration in serum or plasma has been proposed as an important biological marker in various inflammation-related pathologies. We previously showed that low titer autoantibodies against HMGB1 could emerge during the course of sepsis. Importantly their presence was positively related with patients' survival. In this study, we focused on plasma samples from 2 patients who survived sepsis and exhibited high titer antibodies to HMGB1. These antibodies were proved to be specific for HMGB1 since they did not bind to HMGB2 or to human serum albumin. Following IgG purification, it has shown that both patients secreted HMGB1-hydrolyzing autoantibodies in vitro. These findings suggested that proteolytic antibodies directed against HMGB1 can be produced in patients surviving septic shock.


Subject(s)
Autoantibodies/immunology , HMGB1 Protein/immunology , Shock, Septic/immunology , Autoantibodies/blood , HMGB2 Protein/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Proteolysis , Serum Albumin, Human/immunology , Shock, Septic/mortality , Shock, Septic/pathology
17.
Anal Biochem ; 589: 113491, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31676284

ABSTRACT

Among the biological approaches to therapeutics, are the cells, such as CAR-T cells engineered or not, the antibodies armed or not, and the smaller protein scaffolds that can be modified to render them specific of other proteins, à la façon of antibodies. For several years, we explored ways to substitute antibodies by nanobodies (also known as VHHs), the smallest recognizing part of camelids' heavy-chain antibodies: production of those small proteins in host microorganisms, minute analyses, characterization, and qualification of their affinity towards designed targets. Here, we present three standard VHHs described in the literature: anti-albumin, anti-EGF receptor and anti-HER2, a typical cancer cell surface -associated protein. Because they differ slightly in global structure, they are good models to assess our body of analytical methodologies. The VHHs were expressed in several bacteria strains in order to identify and overcome the bottlenecks to obtain homogeneous preparations of this protein. A large panel of biophysical tools, ranging from spectroscopy to mass spectrometry, was here combined to assess VHH structural features and the impact of the disulfide bond. The routes are now ready to move to more complex VHHs raised against specific targets in numerous areas including oncology.


Subject(s)
Camelids, New World/immunology , Immunoglobulin Heavy Chains , Receptor, ErbB-2/immunology , Serum Albumin, Human/immunology , Single-Domain Antibodies , Animals , Antigens/immunology , Cloning, Molecular , ErbB Receptors/immunology , Escherichia coli/genetics , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/isolation & purification , Recombinant Proteins/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/isolation & purification
18.
Methods Mol Biol ; 2033: 25-37, 2019.
Article in English | MEDLINE | ID: mdl-31332745

ABSTRACT

This protocol details a novel bioconjugation strategy that uses a methanesulfonyl acrylate reagent that is directed to the most reactive lysine on human serum albumin, which enables the construction of chemically defined and stable bioconjugates. The reaction proceeds rapidly and a regioselective modification is achieved using a single molar equivalent of the reagent under biocompatible conditions (37 °C, pH 8.0). Importantly, the bioconjugate retains both the secondary structural content and function of the unmodified protein. During the reaction of the amino group of lysine and the sulfonyl acrylate reagent, methanesulfinic acid is released after the conjugate addition, which then generates an electrophilic acrylate moiety on the protein. This acrylate can be further used for site-specific protein labeling using a synthetic molecule bearing a reactive amine under biocompatible conditions (21 °C, pH 8.0).


Subject(s)
Lysine/chemistry , Protein Engineering/methods , Proteins/chemistry , Serum Albumin, Human/chemistry , Acrylates/chemistry , Humans , Lysine/immunology , Proteins/immunology , Serum Albumin, Human/immunology
19.
J Immunotoxicol ; 16(1): 82-86, 2019 12.
Article in English | MEDLINE | ID: mdl-31271074

ABSTRACT

Measurements of complement-bound circulating immune complexes (cCICs) in pre-clinical studies may provide important information about the etiology of certain pathology findings suggestive of being immune complex mediated. This article describes the development and qualification of a universal methodology to measure cCIC in mice after dosing with species foreign proteins. The assay is a sandwich enzyme-linked immunosorbent assay - exclusively based on commercially available reagents - that could detect mouse IgG bound to complement C3 independent of the test-substance present in the plasma sample. Heat-aggregated serum was used as positive control. The assay was qualified by assessment of acceptance criteria, stability of positive control, precision, and specificity. Finally, the performance of the assay was tested using plasma from mice administered either of three different proteins, i.e bovine serum albumin (BSA), a fully human monoclonal antibody, and a humanized monoclonal antibody.


Subject(s)
Antigen-Antibody Complex/isolation & purification , Complement C3/isolation & purification , Immunoglobulin G/isolation & purification , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/isolation & purification , Antigen-Antibody Complex/blood , Antigen-Antibody Complex/immunology , Complement C3/immunology , Enzyme-Linked Immunosorbent Assay/methods , Female , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Serum Albumin, Human/administration & dosage , Serum Albumin, Human/immunology , Serum Albumin, Human/isolation & purification
20.
PLoS One ; 14(5): e0216736, 2019.
Article in English | MEDLINE | ID: mdl-31116779

ABSTRACT

Over consumption of fructose may lead to obesity and dyslipidemia and cause fructosylation-induced alterations in the structure and function of proteins. The aim of this study was to investigate the role of fructosylated-HSA-AGE in the pathogenesis of fatty liver (NAFLD and NASH) by biochemical, immunological and histological studies. Immunogenicity of fructosylated-HSA-AGE was probed by inducing antibodies in rabbits. Fructosylated-HSA-AGE was found to be highly immunogenic. Furthermore, fructosylated-HSA-AGE caused mild fibrosis with steatosis and portal inflammation of hepatocytes in experimental animals. Liver function test and dyslipidemic parameters in immunized animals were also found to be raised. Ultrasonography, which should form part of the assessment of chronically raised transaminases, shows fatty infiltration. Interestingly, alanine aminotransferase (ALT), aspartate aminotransferase (AST), bilirubin, total cholesterol (TC) and triglyceride (TG) profiles confirms USG images of overweight, obese patients. Thus, present study demonstrates that fructosylated-HSA-AGE is hepatotoxic, immunologically active and may cause dyslipidemia.


Subject(s)
Advanced Oxidation Protein Products/blood , Autoantibodies/blood , Fructose/blood , Obesity/blood , Overweight/blood , Serum Albumin, Human/immunology , Adult , Advanced Oxidation Protein Products/immunology , Animals , Antibody Specificity , Case-Control Studies , Dyslipidemias/blood , Dyslipidemias/etiology , Dyslipidemias/immunology , Female , Fructose/immunology , Humans , Liver/metabolism , Liver/pathology , Microscopy, Electron, Scanning , Obesity/immunology , Obesity/pathology , Overweight/immunology , Overweight/pathology , Rabbits , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...