Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
MAbs ; 11(3): 516-531, 2019 04.
Article in English | MEDLINE | ID: mdl-30663541

ABSTRACT

We describe here the design, construction and validation of ALTHEA Gold Libraries™. These single-chain variable fragment (scFv), semisynthetic libraries are built on synthetic human well-known IGHV and IGKV germline genes combined with natural human complementarity-determining region (CDR)-H3/JH (H3J) fragments. One IGHV gene provided a universal VH scaffold and was paired with two IGKV scaffolds to furnish different topographies for binding distinct epitopes. The scaffolds were diversified at positions identified as in contact with antigens in the known antigen-antibody complex structures. The diversification regime consisted of high-usage amino acids found at those positions in human antibody sequences. Functionality, stability and diversity of the libraries were improved throughout a three-step construction process. In a first step, fully synthetic primary libraries were generated by combining the diversified scaffolds with a set of synthetic neutral H3J germline gene fragments. The second step consisted of selecting the primary libraries for enhanced thermostability based on the natural capacity of Protein A to bind the universal VH scaffold. In the third and final step, the resultant stable synthetic antibody fragments were combined with natural H3J fragments obtained from peripheral blood mononuclear cells of a large pool of 200 donors. Validation of ALTHEA Gold Libraries™ with seven targets yielded specific antibodies in all the cases. Further characterization of the isolated antibodies indicated KD values as human IgG1 molecules in the single-digit and sub-nM range. The thermal stability (Tm) of all the antigen-binding fragments was 75°C-80°C, demonstrating that ALTHEA Gold Libraries™ are a valuable source of specific, high affinity and highly stable antibodies.


Subject(s)
Complementarity Determining Regions , Gene Library , Immunoglobulin G , Single-Chain Antibodies , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Leukocytes, Mononuclear/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
2.
MAbs ; 9(5): 854-873, 2017 07.
Article in English | MEDLINE | ID: mdl-28379093

ABSTRACT

Amino acid sequence differences in the variable region of immunoglobulin (Ig) cause wide variations in secretion outputs. To address how a primary sequence difference comes to modulate Ig secretion, we investigated the biosynthetic process of 2 human IgG2κ monoclonal antibodies (mAbs) that differ only by one amino acid in the light chain complementarity-determining region 1 while showing ∼20-fold variance in secretion titer. Although poorly secreted, the lower-secreting mAb of the 2 was by no means defective in terms of its folding stability, antigen binding, and in vitro biologic activity. However, upon overexpression in HEK293 cells, the low-secreting mAb revealed a high propensity to aggregate into enlarged globular structures called Russell bodies (RBs) in the endoplasmic reticulum. While Golgi morphology was affected by the formation of RBs, secretory pathway membrane traffic remained operational in those cells. Importantly, cellular protein synthesis was severely suppressed in RB-positive cells through the phosphorylation of eIF2α. PERK-dependent signaling was implicated in this event, given the upregulation and nuclear accumulation of downstream effectors such as ATF4 and CHOP. These findings illustrated that the underlining process of poor Ig secretion in RB-positive cells was due to downregulation of Ig synthesis instead of a disruption or blockade of secretory pathway trafficking. Therefore, RB formation signifies an end of active Ig production at the protein translation level. Consequently, depending on how soon and how severely an antibody-expressing cell develops the RB phenotype, the productive window of Ig secretion can vary widely among the cells expressing different mAbs.


Subject(s)
Amino Acid Substitution , Complementarity Determining Regions/biosynthesis , Eukaryotic Initiation Factor-2/metabolism , Immunoglobulin G/biosynthesis , Protein Biosynthesis , Secretory Pathway , Animals , Complementarity Determining Regions/genetics , HEK293 Cells , Humans , Immunoglobulin G/genetics , Mice , Phosphorylation
3.
J Biol Chem ; 291(49): 25292-25305, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27707880

ABSTRACT

The pre-T cell receptor (pre-TCR) is a pTα-ß heterodimer functioning in early αß T cell development. Although once thought to be ligand-autonomous, recent studies show that pre-TCRs participate in thymic repertoire formation through recognition of peptides bound to major histocompatibility molecules (pMHC). Using optical tweezers, we probe pre-TCR bonding with pMHC at the single molecule level. Like the αßTCR, the pre-TCR is a mechanosensor undergoing force-based structural transitions that dynamically enhance bond lifetimes and exploiting allosteric control regulated via the Cß FG loop region. The pre-TCR structural transitions exhibit greater reversibility than TCRαß and ordered force-bond lifetime curves. Higher piconewton force requires binding through both complementarity determining region loops and hydrophobic Vß patch apposition. This patch functions in the pre-TCR as a surrogate Vα domain, fostering ligand promiscuity to favor development of ß chains with self-reactivity but is occluded by α subunit replacement of pTα upon αßTCR formation. At the double negative 3 thymocyte stage where the pre-TCR is first expressed, pre-TCR interaction with self-pMHC ligands imparts growth and survival advantages as revealed in thymic stromal cultures, imprinting fundamental self-reactivity in the T cell repertoire. Collectively, our data imply the existence of sequential mechanosensor αßTCR repertoire tuning via the pre-TCR.


Subject(s)
Complementarity Determining Regions , Gene Expression Regulation/physiology , Receptors, Antigen, T-Cell, alpha-beta , Thymocytes , Animals , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Mice , Mice, Knockout , Protein Structure, Secondary , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymocytes/chemistry , Thymocytes/cytology , Thymocytes/metabolism
4.
Monoclon Antib Immunodiagn Immunother ; 34(6): 404-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26683180

ABSTRACT

Single-chain variable antibody fragments (scFvs) are attractive candidates for targeted immunotherapy in several human diseases. In this study, a concise humanization strategy combined with an optimized production method for humanizing scFvs was successfully employed. Two antibody clones, one directed against the hemagglutinin of H5N1 influenza virus, the other against EpCAM, a cancer biomarker, were used to demonstrate the validity of the method. Heavy chain (VH) and light chain (VL) variable regions of immunoglobulin genes from mouse hybridoma cells were sequenced and subjected to the construction of mouse scFv 3-D structure. Based on in silico modeling, the humanized version of the scFv was designed via complementarity-determining region (CDR) grafting with the retention of mouse framework region (FR) residues identified by primary sequence analysis. Root-mean-square deviation (RMSD) value between mouse and humanized scFv structures was calculated to evaluate the preservation of CDR conformation. Mouse and humanized scFv genes were then constructed and expressed in Escherichia coli. Using this method, we successfully generated humanized scFvs that retained the targeting activity of their respective mouse scFv counterparts. In addition, the humanized scFvs were engineered with a C-terminal cysteine residue (hscFv-C) for site-directed conjugation for use in future targeting applications. The hscFv-C expression was extensively optimized to improve protein production yield. The protocol yielded a 20-fold increase in production of hscFv-Cs in E. coli periplasm. The strategy described in this study may be applicable in the humanization of other antibodies derived from mouse hybridoma.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Antigens/analysis , Complementarity Determining Regions/biosynthesis , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Light Chains/biosynthesis , Single-Chain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antigens/genetics , Antigens/immunology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cloning, Molecular , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Computer Simulation , Epithelial Cell Adhesion Molecule , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/analysis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Hybridomas/chemistry , Hybridomas/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Analysis, DNA , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Structural Homology, Protein
5.
MAbs ; 7(1): 180-91, 2015.
Article in English | MEDLINE | ID: mdl-25524068

ABSTRACT

The Wnt signaling pathway is of central importance in embryogenesis, development and adult tissue homeostasis, and dysregulation of this pathway is associated with cancer and other diseases. Despite the developmental and potential therapeutic significance of this pathway, many aspects of Wnt signaling, including the control of the master transcriptional co-activator ß-catenin, remain poorly understood. In order to explore this aspect, a diverse immune llama VHH phagemid library was constructed and panned against ß-catenin. VHH antibody fragments from the library were expressed intracellularly, and a number of antibodies were shown to possess function-modifying intracellular activity in a luciferase-based Wnt signaling HEK293 reporter bioassay. Further characterization of one such VHH (named LL3) confirmed that it bound endogenous ß-catenin, and that it inhibited the Wnt signaling pathway downstream of the destruction complex, while production of a control Ala-substituted complementarity-determining region (CDR)3 mutant demonstrated that the inhibition of ß-catenin activity by the parent intracellular antibody was dependent on the specific CDR sequence of the antibody.


Subject(s)
Complementarity Determining Regions/biosynthesis , Single-Chain Antibodies/biosynthesis , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , Binding Sites, Antibody , Complementarity Determining Regions/genetics , HEK293 Cells , Humans , Single-Chain Antibodies/genetics , beta Catenin/genetics , beta Catenin/metabolism
6.
Monoclon Antib Immunodiagn Immunother ; 33(2): 67-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24746146

ABSTRACT

Recombinant antibody technologies are rapidly becoming available and showing considerable clinical success. However, the immunogenicity of murine-derived monoclonal antibodies is restrictive in cancer immunotherapy. Humanized antibodies can overcome these problems and are considered to be a promising alternative therapeutic agent. There are several approaches for antibody humanization. In this article we review various methods used in the antibody humanization process.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Protein Engineering/methods , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Humans
7.
Methods ; 65(1): 68-76, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23816785

ABSTRACT

Antibodies are key components of the adaptive immune system and are well-established protein therapeutic agents. Typically high-affinity antibodies are obtained by immunization of rodent species that need to be humanized to reduce their immunogenicity. The complementarity-determining regions (CDRs) contain the residues in a defined loop structure that confer antigen binding, which must be retained in the humanized antibody. To design a humanized antibody, we graft the mature murine CDRs onto a germline human acceptor framework. Structural defects due to mismatches at the graft interface can be fixed by mutating some framework residues to murine, or by mutating some residues on the CDRs' backside to human or to a de novo designed sequence. The first approach, framework redesign, can yield an antibody with binding better than the CDR graft and one equivalent to the mature murine, and reduced immunogenicity. The second approach, CDR redesign, is presented here as a new approach, yielding an antibody with binding better than the CDR graft, and immunogenicity potentially less than that from framework redesign. Application of both approaches to the humanization of anti-α4 integrin antibody HP1/2 is presented and the concept of the hybrid humanization approach that retains "difficult to match" murine framework amino acids and uses de novo CDR design to minimize murine amino acid content and reduce cell-mediated cytotoxicity liabilities is discussed.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Complementarity Determining Regions/biosynthesis , Immunoglobulin Fab Fragments/biosynthesis , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibody Affinity , Binding Sites , Cloning, Molecular , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Hybridomas , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Jurkat Cells , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed
8.
Protein Eng Des Sel ; 26(2): 113-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23118340

ABSTRACT

We showed previously that humanization of 528, a murine anti-epidermal growth factor receptor (EGFR) antibody, causes reduced affinity for its target. Here, to improve the affinity of the humanized antibody for use in cancer immunotherapy, we constructed phage display libraries focused on the complementarity-determining regions (CDRs) of the antibody and carried out affinity selection. Two-step selections using libraries constructed in a stepwise manner enabled a 32-fold affinity enhancement of humanized 528 (h528). Thermodynamic analysis of the interactions between the variable domain fragment of h528 (h528Fv) mutants and the soluble extracellular domain of EGFR indicated that the h528Fv mutants obtained from the first selection showed a large increase in negative enthalpy change due to binding, resulting in affinity enhancement. Furthermore, mutants from the second selection showed a decrease in entropy loss, which led to further affinity maturation. These results suggest that a single mutation in the heavy chain variable domain (i.e. Tyr(52) to Trp) enthalpically contributed for overcoming the energetic barrier to the antigen-antibody interaction, which was a major hurdle for the in vitro affinity maturation of h528. We reported previously that the humanized bispecific diabody hEx3 Db, which targets EGFR and CD3, shows strong anti-tumor activity. hEx3 Db mutants, in which the variable domains of h528 were replaced with those of the affinity-enhanced mutants, were prepared and characterized. In a growth inhibition assay of tumor cells, the hEx3 Db mutants showed stronger anti-tumor activity than that of hEx3 Db, suggesting that affinity enhancement of h528Fv enhances the anti-tumor activity of the bispecific diabody.


Subject(s)
Antibody Affinity , Complementarity Determining Regions/biosynthesis , ErbB Receptors/immunology , Neoplasms/therapy , Single-Chain Antibodies/biosynthesis , Amino Acid Substitution , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Complementarity Determining Regions/genetics , Complementarity Determining Regions/pharmacology , Humans , Immunotherapy , Inhibitory Concentration 50 , Kinetics , Mutagenesis, Site-Directed , Peptide Library , Protein Binding , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Thermodynamics
9.
J Immunol ; 190(1): 447-57, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23203931

ABSTRACT

The optimum use of allogeneic blood and marrow transplantation (BMT) as a curative therapy for hematological malignancies lies in the successful separation of mature donor T cells that are host reactive and induce graft-versus-host disease (GVHD) from those that are tumor reactive and mediate graft-versus-leukemia (GVL) effects. To study whether this separation was possible in an MHC-matched murine BMT model (B10.BR→CBA) with a CBA-derived myeloid leukemia line, MMC6, we used TCR Vß CDR3-size spectratype analysis to first show that the Vß13 family was highly skewed in the B10.BR anti-MMC6 CD8(+) T cell response but not in the alloresponse against recipient cells alone. Transplantation of CD8(+)Vß13(+) T cells at the dose equivalent of their constituency in 1 × 10(7) CD8(+) T cells, a dose that had been shown to mediate lethal GVHD in recipient mice, induced a slight GVL response with no concomitant GVHD. Increasing doses of CD8(+)Vß13(+) T cells led to more significant GVL responses but also increased GVHD symptoms and associated mortality. Subsequent spectratype analysis of GVHD target tissues revealed involvement of gut-infiltrating CD8(+)Vß13(+) T cells accounting for the observed in vivo effects. When BMT recipients were given MMC6-presensitized CD8(+)Vß13(+) T cells, they displayed a significant GVL response with minimal GVHD. Spectratype analysis of tumor-presensitized, gut-infiltrating CD8(+)Vß13(+) T cells showed preferential usage of tumor-reactive CDR3-size lengths, and these cells expressed increased effector memory phenotype (CD44(+)CD62L(-/lo)). Thus, Vß spectratyping can identify T cells involved in antihost and antitumor reactivity and tumor presensitization can aid in the separation of GVHD and GVL responses.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Leukemia, Myeloid, Acute/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Animals , Bone Marrow Transplantation/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Complementarity Determining Regions/biosynthesis , Disease Models, Animal , Graft vs Host Disease/mortality , Graft vs Host Disease/therapy , Immunoglobulin Variable Region/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
10.
J Immunol ; 189(6): 3221-30, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22865917

ABSTRACT

To understand better how selection processes balance the benefits of Ig repertoire diversity with the risks of autoreactivity and nonfunctionality of highly variable IgH CDR3s, we collected millions of rearranged germline IgH CDR3 sequences by deep sequencing of DNA from mature human naive B cells purified from four individuals and analyzed the data with computational methods. Long HCDR3 regions, often components of HIV-neutralizing Abs, appear to derive not only from incorporation of long D genes and insertion of large N regions but also by usage of multiple D gene segments in tandem. However, comparison of productive and out-of-frame IgH rearrangements revealed a selection bias against long HCDR3 loops, suggesting these may be disproportionately either poorly functional or autoreactive. Our data suggest that developmental selection removes HCDR3 loops containing patches of hydrophobicity, which are commonly found in some auto-antibodies, and at least 69% of the initial productive IgH rearrangements are removed from the repertoire during B cell development. Additionally, we have demonstrated the potential utility of this new technology for vaccine development with the identification in all four individuals of related candidate germline IgH precursors of the HIV-neutralizing Ab 4E10.


Subject(s)
Antibodies, Neutralizing/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Gene Rearrangement, B-Lymphocyte/immunology , Immunoglobulin Heavy Chains/biosynthesis , Sequence Analysis, DNA , Antibodies, Neutralizing/genetics , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Computational Biology , Conserved Sequence/genetics , Conserved Sequence/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/biosynthesis , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Sequence Analysis, DNA/methods , Somatic Hypermutation, Immunoglobulin
11.
J Immunol ; 187(2): 879-86, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21677133

ABSTRACT

Anti-polysaccharide Ab responses in mice are often oligoclonal, and the mechanisms involved in Ag-specific clone production and selection remain poorly understood. We evaluated the relative contribution of D(H) germline content versus N nucleotide addition in a classic oligoclonal, T-independent Ab response (α 1→3 dextran [DEX]) by challenging adult TdT-sufficient (TdT(+/+)) and TdT-deficient (TdT(-/-)) gene-targeted mice, limited to the use of a single D(H) gene segment (D-limited mice), with Enterobacter cloacae. D-limited mice achieved anti-DEX-specific levels of Abs that were broadly comparable to those of wild-type (WT) BALB/c mice. Sequence analysis of the third CDR of the H chain intervals obtained by PCR amplification of V(H) domain DNA from DEX-specific plasmablasts revealed the near universal presence of an aspartic acid residue (D99) at the V-D junction, irrespective of the composition of the D(H) locus. Although WT mice were able to use germline D(H) (DQ52, DSP, or DST) gene segment sequence, TdT activity, or both to produce D99, all three D-limited mouse strains relied exclusively on N addition. Additionally, in the absence of TdT, D-limited mice failed to produce a DEX response. Coupled with previous studies demonstrating a reduced response to DEX in TdT(-/-) mice with a WT D(H) locus, we concluded that in the case of the anti-DEX repertoire, which uses a short third CDR of the H chain, the anti-DEX response relies more intensely on sequences created by postnatal N nucleotide addition than on the germline sequence of the D(H).


Subject(s)
Antibodies, Bacterial/biosynthesis , Antibody Diversity , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/immunology , Dextrans/immunology , Immunoglobulin Heavy Chains/biosynthesis , Polysaccharides, Bacterial/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Antibodies, Bacterial/genetics , Antibody Diversity/genetics , Base Sequence , Complementarity Determining Regions/genetics , Dextrans/administration & dosage , Dextrans/genetics , Enterobacter cloacae/immunology , Gene Rearrangement, B-Lymphocyte/immunology , Immunoglobulin Heavy Chains/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/genetics
12.
J Immunol ; 186(6): 3787-97, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21325620

ABSTRACT

Autoreactive T cells, responsible for the destruction of pancreatic ß cells in type 1 diabetes, are known to have a skewed TCR repertoire in the NOD mouse. To define the autoreactive T cell repertoire in human diabetes, we searched for intraislet monoclonal expansions from a recent onset in human pancreas to then trace them down to the patient's peripheral blood and spleen. Islet infiltration was diverse, but five monoclonal TCR ß-chain variable expansions were detected for Vß1, Vß7, Vß11, Vß17, and Vß22 families. To identify any sequence bias in the TCRs from intrapancreatic T cells, we analyzed 139 different CDR3 sequences. We observed amino acid preferences in the NDN region that suggested a skewed TCR repertoire within infiltrating T cells. The monoclonal expanded TCR sequences contained amino acid combinations that fit the observed bias. Using these CDR3 sequences as a marker, we traced some of these expansions in the spleen. There, we identified a Vß22 monoclonal expansion with identical CDR3 sequence to that found in the islets within a polyclonal TCR ß-chain variable repertoire. The same Vß22 TCR was detected in the patient's PBMCs, making a cross talk between the pancreas and spleen that was reflected in peripheral blood evident. No other pancreatic monoclonal expansions were found in peripheral blood or the spleen, suggesting that the Vß22 clone may have expanded or accumulated in situ by an autoantigen present in both the spleen and pancreas. Thus, the patient's spleen might be contributing to disease perpetuation by expanding or retaining some autoreactive T cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Receptors, Antigen, T-Cell/biosynthesis , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cell Movement/immunology , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/blood , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Humans , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/blood , Islets of Langerhans/pathology , Lymphocyte Activation/immunology , Molecular Sequence Data , Receptors, Antigen, T-Cell/blood , Spleen/pathology , T-Lymphocyte Subsets/pathology , Young Adult
13.
J Immunol Methods ; 353(1-2): 24-30, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-19961852

ABSTRACT

Phage display technology has emerged as a leading approach to select proteins with improved properties for many different types of applications. The selection typically selects not only for improved binding properties but also for other factors such as efficiency of protein production and folding in Escherichia coli, the host in which the proteins and the phage are produced. Furthermore, the selection methodology is likely to influence the character of retrieved variants. We have now defined the extent whereby the charge of the displayed proteins influence the selection process, resulting in an increased average positive charge among selected proteins in comparison to the proteins that are harbored in the library before selection. Implications of and possible routes to minimize this effect are discussed.


Subject(s)
Antibody Specificity , Complementarity Determining Regions/chemistry , Peptide Library , Amino Acid Sequence , Antibody Affinity , Biotinylation , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Models, Molecular , Molecular Sequence Data , Osmolar Concentration , Protein Conformation , Recombinant Proteins/chemistry , Sodium Chloride/chemistry , Surface Properties
14.
J Immunol ; 183(12): 8015-25, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933850

ABSTRACT

Patients chronically infected with Trypanosoma cruzi develop chronic Chagas' heart disease (cChHD). Their Ab response is suspected to be involved in the cardiac pathogenesis. Reactivity of serum Abs from these patients has been extensively studied but little is known about the diversity of the in vivo IgG repertoire. We analyzed 125 variable H chain (VH) genes and compared it to repertoires from healthy individuals, and patients with autoimmune processes and other infections. VH were from plasma cells isolated from heart tissue of three cChHD patients and from a Fab combinatorial library derived from bone marrow of another cChHD patient. The role of the parasite in shaping the Ab repertoire was assessed analyzing VH genes before and after panning against T. cruzi Ag. Among recovered VH genes, a significantly increased representation of VH4 was observed. Plasma cells at the site of cardiac infiltration showed an increased VH1 usage. CDR3 lengths were similar to the ones found in the healthy repertoire and significantly shorter than in other infections. VH derived from anti-T. cruzi Fab and plasma cells showed a higher proportion of hypermutated genes, 46.9% and 43.75%, respectively, vs 30.9% of the cChHD patient repertoire, pointing to the role of parasite Ags in the shaping of the humoral response in Chagas' disease. No histological evidence of germinal center-like structures was observed in heart tissue. In accordance, VH analysis of heart plasmocytes revealed no evidence of clonal B cell expansion, suggesting that they migrated into heart tissue from secondary lymphoid organs.


Subject(s)
Antibodies, Protozoan/genetics , Chagas Cardiomyopathy/immunology , Gene Rearrangement, B-Lymphocyte/genetics , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Adult , Amino Acid Sequence , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , B-Lymphocytes/pathology , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Chronic Disease , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Somatic Hypermutation, Immunoglobulin/genetics , Trypanosoma cruzi/immunology
15.
J Immunol ; 182(6): 3583-96, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265137

ABSTRACT

Developing autoreactive B cells may edit (change) their specificity by secondary H or L chain gene rearrangement. Recently, using mice hemizygous for a site-directed VDJH and VJkappa transgene (tg) encoding an autoreactive Ab, we reported ongoing L chain editing not only in bone marrow cells with a pre-B/immature B cell phenotype but also in immature/transitional splenic B cells. Using the same transgenic model, we report here that editing at the H chain locus appears to occur exclusively in bone marrow cells with a pro-B phenotype. H chain editing is shown to involve VH replacement at the tg allele or VH rearrangement at the wild-type (wt) allele when the tg is inactivated by nonproductive VH replacement. VH replacement/rearrangement at the tg/wt alleles was found to entail diverse usage of VH genes. Whereas the development of edited B cells expressing the wt allele was dependent on the lambda5 component of the surrogate L chain, the development of B cells expressing the tg allele, including those with VH replacement, appeared to be lambda5 independent. We suggest that the unique CDR3 region of the tg-encoded muH chain is responsible for the lambda5 independence of tg-expressing B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Immunoglobulin Light Chains, Surrogate/genetics , RNA Editing/immunology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Immunoglobulin Light Chains, Surrogate/biosynthesis , Immunoglobulin Light Chains, Surrogate/metabolism , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/biosynthesis , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/biosynthesis , Immunoglobulin lambda-Chains/genetics , Immunoglobulin mu-Chains/biosynthesis , Immunoglobulin mu-Chains/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , RNA Editing/genetics
16.
J Immunol ; 182(2): 784-92, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124721

ABSTRACT

There are well-characterized age-related changes in the peripheral repertoire of CD8 T cells characterized by reductions in the ratio of naive:memory T cells and the development of large clonal expansions in the memory pool. In addition, the TCR repertoire of naive T cells is reduced with aging. Because a diverse repertoire of naive T cells is essential for a vigorous response to new infections and vaccinations, there is much interest in understanding the mechanisms responsible for declining repertoire diversity. It has been proposed that one reason for declining repertoire diversity in the naive T cell pool is an increasing dependence on homeostatic proliferation in the absence of new thymic emigrants for maintenance of the naive peripheral pool. In this study, we have analyzed the naive CD8 T cell repertoire in young and aged mice by DNA spectratype and sequence analysis. Our data show that naive T cells from aged mice have perturbed spectratype profiles compared with the normally Gaussian spectratype profiles characteristic of naive CD8 T cells from young mice. In addition, DNA sequence analysis formally demonstrated a loss of diversity associated with skewed spectratype profiles. Unexpectedly, we found multiple repeats of the same sequence in naive T cells from aged but not young mice, consistent with clonal expansions previously described only in the memory T cell pool. Clonal expansions among naive T cells suggests dysregulation in the normal homeostatic proliferative mechanisms that operate in young mice to maintain diversity in the naive T cell repertoire.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Receptors, Antigen, T-Cell/biosynthesis , Aging/genetics , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Separation , Clone Cells , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Homeostasis/immunology , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology
17.
Biochem Biophys Res Commun ; 379(2): 314-8, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19103171

ABSTRACT

Humanization of nonhuman antibodies (Abs) has been carried out mainly for Abs which bind to antigen without catalytic activity. Here we report humanization of mouse-originated 3D8 (m3D8) mAbs (scFv, VH, and VL) with DNA hydrolyzing catalytic activity by grafting the complementarity determining regions (CDRs) into the corresponding regions of a fixed human framework scaffold, generating humanized 3D8 (h3D8) Abs in the respective format of scFv, VH, and VL. h3D8 Abs retained comparable DNA binding and hydrolyzing activities to those of the corresponding m3D8 Abs. Our results suggest that CDRs of anti-DNA hydrolyzing Abs might possess the intrinsic properties of DNA binding and hydrolyzing activities.


Subject(s)
Antibodies, Catalytic/biosynthesis , Antibodies, Monoclonal/biosynthesis , Complementarity Determining Regions/biosynthesis , DNA/immunology , Amino Acid Sequence , Animals , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Humans , Hydrolysis , Mice , Models, Molecular , Molecular Sequence Data
18.
Blood ; 112(8): 3517-25, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18541718

ABSTRACT

Following allogeneic blood and marrow transplantation (BMT), mature donor T cells can enhance engraftment, counteract opportunistic infections, and mount graft-versus-tumor (GVT) responses, but at the risk of developing graft-versus-host disease (GVHD). With the aim of separating the beneficial effects of donor T cells from GVHD, one approach would be to selectively deplete subsets of alloreactive T cells in the hematopoietic cell inoculum. In this regard, TCR Vbeta repertoire analysis by CDR3-size spectratyping can be a powerful tool for the characterization of alloreactive T-cell responses. We investigated the potential of this spectratype approach by comparing the donor T-cell alloresponses generated in vitro against patient peripheral blood lymphocytes (PBLs) with those detected in vivo posttransplantation. The results indicated that for most Vbeta families that exhibited alloreactive CDR3-size skewing, there was a robust overlap between the in vitro antipatient and in vivo spectratype histograms. Thus, in vitro spectratype analysis may be useful for determining the alloreactive T-cell response involved in GVHD development and, thereby, could serve to guide select Vbeta family depletion for designer transplants to improve outcomes.


Subject(s)
Bone Marrow Transplantation/methods , Hematologic Neoplasms/therapy , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Transplantation, Homologous/methods , Adult , Aged , Bone Marrow Cells/cytology , Bone Marrow Transplantation/instrumentation , Complementarity Determining Regions/biosynthesis , Female , Graft vs Host Disease , Hematologic Neoplasms/metabolism , Humans , Male , Middle Aged , Receptors, Antigen, T-Cell, alpha-beta/therapeutic use , T-Lymphocytes/metabolism , Transplantation, Homologous/instrumentation , Treatment Outcome
19.
J Immunol ; 180(4): 2347-56, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250444

ABSTRACT

Isolator piglets infected with porcine reproductive and respiratory syndrome virus (PRRSV), which is related to the lactate dehydrogenase-elevating virus of mice, develop severe hypergammaglobulinemia, lymph node adenopathy, and autoimmune disease. Many of the polyclonally activated B cell clones bear hydrophobic H chain CDR3s (HCDR3s) and are disseminated to most lymphoid tissues. We show in this study that B cells with identical hydrophobic HCDR3s are expressed with all major isotypes in PRRSV-infected piglets (PIPs), explaining why PRRSV-induced hypergammaglobulinemia is seen in all major isotypes. Up to one-third of randomly selected VDJ clones from the respiratory tract of PIPs have hydrophobic HCDR3s exclusively bearing VDJ rearrangements with CDR1, CDR2, and nearly intact DH segments in germline configuration. These HCDR3s are long and D(H)A and D(H)B are exclusively used in reading frame 3. A minimal tripeptide motif containing three hydrophobic amino acids (Leu, Val, and Ile) or any two plus alanine is common to this hydrophobic patch. We propose that PRRSV infection causes generalized Ag-independent B cell activation and hypergammaglobulinemia with biased expansion of a subpopulation of the preimmune repertoire with hydrophobic binding sites that normally disappears during Ag-driven repertoire diversification. Elevated Ig levels in PIP cannot be explained as antiviral Abs; some Igs can account for autoantibodies to dsDNA and Golgi, whereas those with hydrophobic binding sites may account for the Ig aggregates seen in PIPs and lactate dehydrogenase-elevating virus-infected mice. This diversion from normal repertoire development may explain the delayed immune response to PRRSV.


Subject(s)
B-Lymphocyte Subsets/cytology , Cell Differentiation/immunology , Cell Proliferation , Complementarity Determining Regions/biosynthesis , Germ Cells/cytology , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Isotypes/biosynthesis , Porcine respiratory and reproductive syndrome virus/immunology , Animals , Animals, Newborn , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/virology , Cell Differentiation/genetics , Clone Cells , Complementarity Determining Regions/genetics , Germ Cells/immunology , Germ Cells/metabolism , Germ Cells/virology , Hydrophobic and Hydrophilic Interactions , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/genetics , Molecular Sequence Data , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/pathology , Random Allocation , Somatic Hypermutation, Immunoglobulin , Swine
20.
J Immunol ; 179(6): 3841-50, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785821

ABSTRACT

Due to technical limitations, little knowledge exists on the composition of Ag-specific polyclonal Ab responses. Hence, we here present a molecular analysis of two representative human Ab repertoires isolated by using a novel single-cell cloning approach. The observed genetic diversity among tetanus toxoid-specific plasma cells indicate that human polyclonal repertoires are limited to the order of 100 B cell clones and hypermutated variants thereof. Affinity and kinetic binding constants are log-normally distributed, and median values are close to the proposed affinity ceilings for positive selection. Abs varied a million-fold in affinity but were restricted in their off-rates with an upper limit of 2 x 10(-3) s(-1). Identification of Abs of high affinity without hypermutations in combination with a modest effect of hypermutations on observed affinity increases indicate that Abs selected from the naive repertoire are not only of low affinity but cover a relatively large span in affinity, reaching into the subnanomolar range.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antibody Affinity , Antibody Diversity , Tetanus Toxoid/immunology , Adult , Amino Acid Sequence , Antibodies, Bacterial/genetics , Antibodies, Bacterial/isolation & purification , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibody Affinity/genetics , Antibody Diversity/genetics , Clostridium tetani/immunology , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , Immunoglobulin Joining Region/biosynthesis , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Kinetics , Male , Molecular Sequence Data , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tetanus Toxoid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...