Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 50(1): 142-145, 2020 01.
Article in English | MEDLINE | ID: mdl-31580480

ABSTRACT

The semi-public T-cell response towards the gluten epitope DQ2.5-glia-α2 uses a prototypic TCR encoded by the germline segments TRAV26-1 and TRBV7-2. Through mutagenesis experiments, we show that a TRAV26-1encoded recognition motif contacts the MHC ß-chain and the TCR CDR3ß loop underpinning this conserved T-cell response restricted to the prototypic TCRs.


Subject(s)
Celiac Disease/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Amino Acid Motifs/immunology , Epitopes, T-Lymphocyte/chemistry , Humans , Receptors, Antigen, T-Cell, alpha-beta/chemistry
2.
Gastroenterology ; 156(5): 1428-1439.e10, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30593798

ABSTRACT

BACKGROUND & AIMS: Development of celiac disease is believed to involve the transglutaminase-dependent response of CD4+ T cells toward deamidated gluten peptides in the intestinal mucosa of individuals with specific HLA-DQ haplotypes. We investigated the antigen presentation process during this mucosal immune response. METHODS: We generated monoclonal antibodies (mAbs) specific for the peptide-MHC (pMHC) complex of HLA-DQ2.5 and the immunodominant gluten epitope DQ2.5-glia-α1a using phage display. We used these mAbs to assess gluten peptide presentation and phenotypes of presenting cells by flow cytometry and enzyme-linked immune absorbent spot (ELISPOT) in freshly prepared single-cell suspensions from intestinal biopsies from 40 patients with celiac disease (35 untreated and 5 on a gluten-free diet) as well as 18 subjects with confirmed noninflamed gut mucosa (controls, 12 presumed healthy, 5 undergoing pancreatoduodenectomy, and 1 with potential celiac disease). RESULTS: Using the mAbs, we detected MHC complexes on cells from intestinal biopsies from patients with celiac disease who consume gluten, but not from patients on gluten-free diets. We found B cells and plasma cells to be the most abundant cells that present DQ2.5-glia-α1a in the inflamed mucosa. We identified a subset of plasma cells that expresses B-cell receptors (BCR) specific for gluten peptides or the autoantigen transglutaminase 2 (TG2). Expression of MHC class II (MHCII) was not restricted to these specific plasma cells in patients with celiac disease but was observed in an average 30% of gut plasma cells from patients and controls. CONCLUSIONS: A population of plasma cells from intestinal biopsies of patients with celiac disease express MHCII; this is the most abundant cell type presenting the immunodominant gluten peptide DQ2.5-glia-α1a in the tissues from these patients. These results indicate that plasma cells in the gut can function as antigen-presenting cells and might promote and maintain intestinal inflammation in patients with celiac disease or other inflammatory disorders.


Subject(s)
Antigen-Presenting Cells/immunology , Celiac Disease/immunology , Duodenum/immunology , Glutens/immunology , HLA-DQ Antigens/immunology , Immunity, Mucosal , Immunodominant Epitopes , Intestinal Mucosa/immunology , Peptide Fragments/immunology , Plasma Cells/immunology , Animals , Antigen-Presenting Cells/metabolism , Case-Control Studies , Celiac Disease/diagnosis , Celiac Disease/diet therapy , Celiac Disease/metabolism , Cell Line , Diet, Gluten-Free , Duodenum/metabolism , Duodenum/pathology , GTP-Binding Proteins/immunology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Phenotype , Plasma Cells/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/immunology
3.
PLoS One ; 13(4): e0195868, 2018.
Article in English | MEDLINE | ID: mdl-29649333

ABSTRACT

There is a quest for production of soluble protein of high quality for the study of T-cell receptors (TCRs), but expression often results in low yields of functional molecules. In this study, we used an E. coli chaperone-assisted periplasmic production system and compared expression of 4 different soluble TCR formats: single-chain TCR (scTCR), two different disulfide-linked TCR (dsTCR) formats, and chimeric Fab (cFab). A stabilized version of scTCR was also included. Additionally, we evaluated the influence of host (XL1-Blue or RosettaBlueTM) and the effect of IPTG induction on expression profiles. A celiac disease patient-derived TCR with specificity for gluten was used, and we achieved detectable expression for all formats and variants. We found that expression in RosettaBlueTM without IPTG induction resulted in the highest periplasmic yields. Moreover, after large-scale expression and protein purification, only the scTCR format was obtained in high yields. Importantly, stability engineering of the scTCR was a prerequisite for obtaining reliable biophysical characterization of the TCR-pMHC interaction. The scTCR format is readily compatible with high-throughput screening approaches that may enable both development of reagents allowing for defined peptide MHC (pMHC) characterization and discovery of potential novel therapeutic leads.


Subject(s)
Escherichia coli/genetics , Gene Expression , Models, Molecular , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Escherichia coli/metabolism , Molecular Chaperones/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Antigen, T-Cell/isolation & purification , Receptors, Antigen, T-Cell/metabolism , Solubility , Structure-Activity Relationship
4.
JCI Insight ; 2(17)2017 09 07.
Article in English | MEDLINE | ID: mdl-28878121

ABSTRACT

Selection of biased T cell receptor (TCR) repertoires across individuals is seen in both infectious diseases and autoimmunity, but the underlying molecular basis leading to these shared repertoires remains unclear. Celiac disease (CD) occurs primarily in HLA-DQ2.5+ individuals and is characterized by a CD4+ T cell response against gluten epitopes dominated by DQ2.5-glia-α1a and DQ2.5-glia-α2. The DQ2.5-glia-α2 response recruits a highly biased TCR repertoire composed of TRAV26-1 paired with TRBV7-2 harboring a semipublic CDR3ß loop. We aimed to unravel the molecular basis for this signature. By variable gene segment exchange, directed mutagenesis, and cellular T cell activation studies, we found that TRBV7-3 can substitute for TRBV7-2, as both can contain the canonical CDR3ß loop. Furthermore, we identified a pivotal germline-encoded MHC recognition motif centered on framework residue Y40 in TRAV26-1 engaging both DQB1*02 and the canonical CDR3ß. This allowed prediction of expanded DQ2.5-glia-α2-reactive TCR repertoires, which were confirmed by single-cell sorting and TCR sequencing from CD patient samples. Our data refine our understanding of how HLA-dependent biased TCR repertoires are selected in the periphery due to germline-encoded residues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Codon , Complementarity Determining Regions/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/physiology , Celiac Disease/immunology , Clone Cells , Cloning, Molecular , Epitopes, T-Lymphocyte/immunology , Glutens/immunology , HLA-DQ Antigens/immunology , Humans , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/genetics
5.
J Immunol ; 194(11): 5497-508, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25904551

ABSTRACT

Engineering of the constant Fc part of monoclonal human IgG1 (hIgG1) Abs is an approach to improve effector functions and clinical efficacy of next-generation IgG1-based therapeutics. A main focus in such development is tailoring of in vivo half-life and transport properties by engineering the pH-dependent interaction between IgG and the neonatal Fc receptor (FcRn), as FcRn is the main homeostatic regulator of hIgG1 half-life. However, whether such engineering affects binding to other Fc-binding molecules, such as the classical FcγRs and complement factor C1q, has not been studied in detail. These effector molecules bind to IgG1 in the lower hinge-CH2 region, structurally distant from the binding site for FcRn at the CH2-CH3 elbow region. However, alterations of the structural composition of the Fc may have long-distance effects. Indeed, in this study we show that Fc engineering of hIgG1 for altered binding to FcRn also influences binding to both the classical FcγRs and complement factor C1q, which ultimately results in alterations of cellular mechanisms such as Ab-dependent cell-mediated cytotoxicity, Ab-dependent cellular phagocytosis, and Ab-dependent complement-mediated cell lysis. Thus, engineering of the FcRn-IgG1 interaction may greatly influence effector functions, which has implications for the therapeutic efficacy and use of Fc-engineered hIgG1 variants.


Subject(s)
Antibodies, Monoclonal/genetics , Complement C1q/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/genetics , Receptors, Fc/immunology , Receptors, IgG/immunology , Antibodies, Monoclonal/immunology , Antibody Affinity/genetics , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , HEK293 Cells , Hinge Exons/genetics , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/immunology , Nitrohydroxyiodophenylacetate/immunology , Phagocytosis/immunology , Protein Engineering , Receptors, Fc/genetics , Receptors, IgG/genetics , Surface Plasmon Resonance
6.
J Biol Chem ; 289(50): 34583-94, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25344603

ABSTRACT

Albumin is an abundant blood protein that acts as a transporter of a plethora of small molecules like fatty acids, hormones, toxins, and drugs. In addition, it has an unusual long serum half-life in humans of nearly 3 weeks, which is attributed to its interaction with the neonatal Fc receptor (FcRn). FcRn protects albumin from intracellular degradation via a pH-dependent cellular recycling mechanism. To understand how FcRn impacts the role of albumin as a distributor, it is of importance to unravel the structural mechanism that determines pH-dependent binding. Here, we show that although the C-terminal domain III (DIII) of human serum albumin (HSA) contains the principal binding site, the N-terminal domain I (DI) is important for optimal FcRn binding. Specifically, structural inspection of human FcRn (hFcRn) in complex with HSA revealed that two exposed loops of DI were in proximity with the receptor. To investigate to what extent these contacts affected hFcRn binding, we targeted selected amino acid residues of the loops by mutagenesis. Screening by in vitro interaction assays revealed that several of the engineered HSA variants showed decreased binding to hFcRn, which was also the case for two missense variants with mutations within these loops. In addition, four of the variants showed improved binding. Our findings demonstrate that both DI and DIII are required for optimal binding to FcRn, which has implications for our understanding of the FcRn-albumin relationship and how albumin acts as a distributor. Such knowledge may inspire development of novel HSA-based diagnostics and therapeutics.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Amino Acid Substitution , Binding, Competitive , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Protein Binding , Protein Stability , Protein Structure, Tertiary , Serum Albumin/genetics
7.
J Biol Chem ; 289(19): 13492-502, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24652290

ABSTRACT

A major challenge for the therapeutic use of many peptides and proteins is their short circulatory half-life. Albumin has an extended serum half-life of 3 weeks because of its size and FcRn-mediated recycling that prevents intracellular degradation, properties shared with IgG antibodies. Engineering the strictly pH-dependent IgG-FcRn interaction is known to extend IgG half-life. However, this principle has not been extensively explored for albumin. We have engineered human albumin by introducing single point mutations in the C-terminal end that generated a panel of variants with greatly improved affinities for FcRn. One variant (K573P) with 12-fold improved affinity showed extended serum half-life in normal mice, mice transgenic for human FcRn, and cynomolgus monkeys. Importantly, favorable binding to FcRn was maintained when a single-chain fragment variable antibody was genetically fused to either the N- or the C-terminal end. The engineered albumin variants may be attractive for improving the serum half-life of biopharmaceuticals.


Subject(s)
Albumins/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Albumins/genetics , Albumins/pharmacology , Amino Acid Substitution , Animals , Female , Half-Life , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/pharmacology , Humans , Macaca fascicularis , Mice , Mutation, Missense , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
8.
Nat Commun ; 3: 610, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22215085

ABSTRACT

Albumin is the most abundant protein in blood where it has a pivotal role as a transporter of fatty acids and drugs. Like IgG, albumin has long serum half-life, protected from degradation by pH-dependent recycling mediated by interaction with the neonatal Fc receptor, FcRn. Although the FcRn interaction with IgG is well characterized at the atomic level, its interaction with albumin is not. Here we present structure-based modelling of the FcRn-albumin complex, supported by binding analysis of site-specific mutants, providing mechanistic evidence for the presence of pH-sensitive ionic networks at the interaction interface. These networks involve conserved histidines in both FcRn and albumin domain III. Histidines also contribute to intramolecular interactions that stabilize the otherwise flexible loops at both the interacting surfaces. Molecular details of the FcRn-albumin complex may guide the development of novel albumin variants with altered serum half-life as carriers of drugs.


Subject(s)
Albumins/chemistry , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Mutagenesis , Receptors, Fc/chemistry , Receptors, Fc/genetics , Binding Sites , Escherichia coli/metabolism , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Ions , Models, Molecular , Molecular Conformation , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Serum Albumin/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...