Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 942
Filter
Add more filters

Publication year range
1.
Anal Chem ; 90(16): 9667-9672, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30071161

ABSTRACT

In conventional competitive immunoassays for small molecules (SM), antibodies are either immobilized to solid phases or labeled with magnetic particles or probes. The former involves laborious blocking and washing steps, whereas the latter requires complicated labeling and purification steps. To circumvent these limitations, we describe here a new type of molecular beacon, termed antibody-bridged beacon (AbB), enabling homogeneous detection of SM without any immobilization or labeling of the antibody. The AbB is formed by the binding of an antibody to a pair of SM-labeled oligonucleotide probes that each comprise a stem sequence conjugated by either a fluorophore or a quencher. Competitive binding of the SM target to the antibody destructs the stem-loop structure of AbB, restoring the quenched fluorescence. A minimum binding energy of stem sequences is required for efficient formation of the desired stem-loop structure of AbB. A systematic study of the impact of stem sequences on the fluorescence background and quenching efficiency provided useful benchmarks, e.g., binding energy of -11 kcal/mol, for the construction of AbB. The optimized AbB showed fast signal responses, as demonstrated in the analyses of two small molecule targets, biotin and digoxin. Low nanomolar limits of detection were achieved. The novel AbB strategy, along with the guidelines established for the construction and application of AbB, offers a promising approach for homogeneous detection of small molecules, obviating immobilization or labeling of antibodies as required by other competitive immunoassays.


Subject(s)
Antibodies, Monoclonal/immunology , DNA Probes/chemistry , DNA/chemistry , Fluorescent Dyes/chemistry , Immunoassay/methods , Binding, Competitive/immunology , Biotin/analysis , Biotin/immunology , DNA/genetics , DNA Probes/genetics , Digoxigenin/chemistry , Digoxin/analysis , Digoxin/immunology , Fluorescence , Limit of Detection , Nucleic Acid Hybridization
2.
Cancer Immunol Immunother ; 66(11): 1411-1424, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28634816

ABSTRACT

T-cell receptor (TCR)-pMHC affinity has been generally accepted to be the most important factor dictating antigen recognition in gene-modified T-cells. As such, there is great interest in optimizing TCR-based immunotherapies by enhancing TCR affinity to augment the therapeutic benefit of TCR gene-modified T-cells in cancer patients. However, recent clinical trials using affinity-enhanced TCRs in adoptive cell transfer (ACT) have observed unintended and serious adverse events, including death, attributed to unpredicted off-tumor or off-target cross-reactivity. It is critical to re-evaluate the importance of other biophysical, structural, or cellular factors that drive the reactivity of TCR gene-modified T-cells. Using a model for altered antigen recognition, we determined how TCR-pMHC affinity influenced the reactivity of hepatitis C virus (HCV) TCR gene-modified T-cells against a panel of naturally occurring HCV peptides and HCV-expressing tumor targets. The impact of other factors, such as TCR-pMHC stabilization and signaling contributions by the CD8 co-receptor, as well as antigen and TCR density were also evaluated. We found that changes in TCR-pMHC affinity did not always predict or dictate IFNγ release or degranulation by TCR gene-modified T-cells, suggesting that less emphasis might need to be placed on TCR-pMHC affinity as a means of predicting or augmenting the therapeutic potential of TCR gene-modified T-cells used in ACT. A more complete understanding of antigen recognition by gene-modified T-cells and a more rational approach to improve the design and implementation of novel TCR-based immunotherapies is necessary to enhance efficacy and maximize safety in patients.


Subject(s)
Adoptive Transfer/methods , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , Binding, Competitive/immunology , Cell Line , Cell Line, Tumor , Coculture Techniques , Flow Cytometry , HEK293 Cells , Hep G2 Cells , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , Mice , Peptides/genetics , Peptides/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 111(48): 17248-53, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25411315

ABSTRACT

T-cell receptor affinity for self-antigen has an important role in establishing self-tolerance. Three transgenic mouse strains expressing antigens of variable affinity for the OVA transgenic-I T-cell receptor were generated to address how TCR affinity affects the efficiency of negative selection, the ability to prime an autoimmune response, and the elimination of the relevant target cell. Mice expressing antigens with an affinity just above the negative selection threshold exhibited the highest risk of developing experimental autoimmune diabetes. The data demonstrate that close to the affinity threshold for negative selection, sufficient numbers of self-reactive T cells escape deletion and create an increased risk for the development of autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/metabolism , Autoimmunity/genetics , Binding, Competitive/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Flow Cytometry , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism , Self Tolerance/genetics , Self Tolerance/immunology , T-Lymphocytes/metabolism
4.
J Immunoassay Immunochem ; 38(1): 72-81, 2017.
Article in English | MEDLINE | ID: mdl-27624975

ABSTRACT

Human cardiac troponin I (cTni) is the gold marker for early diagnosis of myocardial infarction. In this regard, four immune-dominant epitopes of cTni were predicted and their 3D structures were determined. Thereafter, the competitive performance of the peptides was monitored with the developed polyclonal antibody-based indirect competitive ELISA; a half-maximal inhibitory concentration (IC50) of 0.49 (µg/mL) and detection limit of 0.037 (µg/mL) were achieved for recombinant cTni. The competitive ELISA determined sensitivity levels of 0.306, 0.141, 0.960, and 0.155 (µg/mL), respectively, for each peptide as competitor. We indicated that two of the selected epitopes have significant sensitivity scales and inhibition ability.


Subject(s)
Binding, Competitive/immunology , Enzyme-Linked Immunosorbent Assay/methods , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Immunosorbents/chemistry , Peptides/immunology , Troponin I/analysis , Troponin I/immunology , Dose-Response Relationship, Drug , Humans , Molecular Dynamics Simulation , Myocardial Infarction/diagnosis , Myocardial Infarction/immunology , Peptides/chemistry , Troponin I/chemistry
5.
Blood ; 124(19): 2937-47, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25202142

ABSTRACT

The glycosyltransferase gene, Ext1, is essential for heparan sulfate production. Induced deletion of Ext1 selectively in Mx1-expressing bone marrow (BM) stromal cells, a known population of skeletal stem/progenitor cells, in adult mice resulted in marked changes in hematopoietic stem and progenitor cell (HSPC) localization. HSPC egressed from BM to spleen after Ext1 deletion. This was associated with altered signaling in the stromal cells and with reduced vascular cell adhesion molecule 1 production by them. Further, pharmacologic inhibition of heparan sulfate mobilized qualitatively more potent and quantitatively more HSPC from the BM than granulocyte colony-stimulating factor alone, including in a setting of granulocyte colony-stimulating factor resistance. The reduced presence of endogenous HSPC after Ext1 deletion was associated with engraftment of transfused HSPC without any toxic conditioning of the host. Therefore, inhibiting heparan sulfate production may provide a means for avoiding the toxicities of radiation or chemotherapy in HSPC transplantation for nonmalignant conditions.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Heparitin Sulfate/biosynthesis , N-Acetylglucosaminyltransferases/metabolism , Stromal Cells/metabolism , Transplantation Conditioning , Animals , Anticoagulants/pharmacology , Binding, Competitive/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins/genetics , Heparin/pharmacology , Heparitin Sulfate/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , N-Acetylglucosaminyltransferases/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Stromal Cells/immunology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
6.
J Immunol ; 193(2): 571-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24943217

ABSTRACT

Autoreactive T cells infiltrating the target organ can possess a broad TCR affinity range. However, the extent to which such biophysical parameters contribute to T cell pathogenic potential remains unclear. In this study, we selected eight InsB9-23-specific TCRs cloned from CD4(+) islet-infiltrating T cells that possessed a relatively broad range of TCR affinity to generate NOD TCR retrogenic mice. These TCRs exhibited a range of two-dimensional affinities (∼ 10(-4)-10(-3) µm(4)) that correlated with functional readouts and responsiveness to activation in vivo. Surprisingly, both higher and lower affinity TCRs could mediate potent insulitis and autoimmune diabetes, suggesting that TCR affinity does not exclusively dictate or correlate with diabetogenic potential. Both central and peripheral tolerance mechanisms selectively impinge on the diabetogenic potential of high-affinity TCRs, mitigating their pathogenicity. Thus, TCR affinity and multiple tolerance mechanisms converge to shape and broaden the diabetogenic T cell repertoire, potentially complicating efforts to induce broad, long-term tolerance.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Immune Tolerance/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Binding, Competitive/immunology , Bone Marrow Transplantation/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Flow Cytometry , Insulin/deficiency , Insulin/genetics , Insulin/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, 129 Strain , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
7.
J Immunol ; 193(2): 783-96, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24928988

ABSTRACT

Human ficolin-2 (L-ficolin/p35) is a lectin-complement pathway activator that is present in normal human plasma and is associated with infectious diseases; however, little is known regarding the roles and mechanisms of ficolin-2 during chronic hepatitis C virus (HCV) infection. In this study, we found that ficolin-2 inhibits the entry of HCV at an early stage of viral infection, regardless of the viral genotype. Ficolin-2 neutralized and inhibited the initial attachment and infection of HCV by binding to the HCV envelope surface glycoproteins E1 and E2, blocking HCV attachment to low-density lipoprotein receptor (LDLR) and scavenger receptor B1, and weakly interfering with CD81 receptor attachment. However, no interference with claudin-1 and occludin receptor attachment was observed. The C-terminal fibrinogen domain (201-313 aa) of ficolin-2 was identified as the critical binding region for the HCV-E1-E2 N-glycans, playing a critical role in the anti-HCV activity. More importantly, we found that apolipoprotein E (ApoE)3, which is enriched in the low-density fractions of HCV RNA-containing particles, promotes HCV infection and inhibits ficolin-2-mediated antiviral activity. ApoE3, but not ApoE2 and ApoE4, blocked the interaction between ficolin-2 and HCV-E2. Our data suggest that the HCV entry inhibitor ficolin-2 is a novel and promising antiviral innate immune molecule, whereas ApoE3 blocks the effect of ficolin-2 and mediates an immune escape mechanism during chronic HCV infection. HCV may be neutralized using compounds directed against the lipoprotein moiety of the viral particle, and ApoE3 may be a new target to combat HCV infection.


Subject(s)
Apolipoprotein E3/immunology , Hepacivirus/immunology , Lectins/immunology , Tumor Escape/immunology , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Binding, Competitive/immunology , Blotting, Western , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Hepacivirus/genetics , Hepacivirus/physiology , Host-Pathogen Interactions/immunology , Humans , Lectins/genetics , Lectins/metabolism , Mannans/immunology , Mannans/metabolism , Microscopy, Confocal , Polysaccharides/immunology , Polysaccharides/metabolism , Protein Binding/immunology , RNA Interference , Receptors, LDL/genetics , Receptors, LDL/immunology , Receptors, LDL/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/immunology , Scavenger Receptors, Class B/metabolism , Tetraspanin 28/genetics , Tetraspanin 28/immunology , Tetraspanin 28/metabolism , Tumor Escape/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Ficolins
8.
J Immunol ; 192(7): 2984-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24591375

ABSTRACT

Allopurinol (ALP) hypersensitivity is a major cause of severe cutaneous adverse reactions and is strongly associated with the HLA-B*58:01 allele. However, it can occur in the absence of this allele with identical clinical manifestations. The immune mechanism of ALP-induced severe cutaneous adverse reactions is poorly understood, and the T cell-reactivity pattern in patients with or without the HLA-B*58:01 allele is not known. To understand the interactions among the drug, HLA, and TCR, we generated T cell lines that react to ALP or its metabolite oxypurinol (OXP) from HLA-B*58:01(+) and HLA-B*58:01(-) donors and assessed their reactivity. ALP/OXP-specific T cells reacted immediately to the addition of the drugs and bypassed intracellular Ag processing, which is consistent with the "pharmacological interaction with immune receptors" (p-i) concept. This direct activation occurred regardless of HLA-B*58:01 status. Although most OXP-specific T cells from HLA-B*58:01(+) donors were restricted by the HLA-B*58:01 molecule for drug recognition, ALP-specific T cells also were restricted to other MHC class I molecules. This can be explained by in silico docking data that suggest that OXP binds to the peptide-binding groove of HLA-B*58:01 with higher affinity. The ensuing T cell responses elicited by ALP or OXP were not limited to particular TCR Vß repertoires. We conclude that the drug-specific T cells are activated by OXP bound to HLA-B*58:01 through the p-i mechanism.


Subject(s)
HLA-B Antigens/immunology , Lymphocyte Activation/immunology , Oxypurinol/immunology , T-Lymphocytes/immunology , Allopurinol/chemistry , Allopurinol/immunology , Allopurinol/pharmacology , Binding, Competitive/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cells, Cultured , Flow Cytometry , HLA-B Antigens/chemistry , HLA-B Antigens/genetics , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Models, Molecular , Molecular Structure , Oxypurinol/chemistry , Oxypurinol/pharmacology , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
9.
Cell Mol Life Sci ; 72(7): 1405-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25287047

ABSTRACT

Affinity proteins have advanced the field of targeted therapeutics due to their generally higher specificity compared to small molecular compounds. However, side effects caused by on-target binding in healthy tissues are still an issue. Here, we design and investigate a prodrug strategy for improving tissue specificity of Affibody molecules in future in vivo studies. The prodrug Affibody (pro-Affibody) against the HER2 receptor was constructed by fusing a HER2-specific Affibody (ZHER2) to an anti-idiotypic Affibody (anti-ZHER2). The linker was engineered to comprise a substrate peptide for the cancer-associated matrix metalloprotease 1 (MMP-1). The hypothesis was that the binding surface of ZHER2 would thereby be blocked from interacting with HER2 until the substrate peptide was specifically hydrolyzed by MMP-1. Binding should thereby only occur where MMP-1 is overexpressed, potentially decreasing on-target toxicities in normal tissues. The pro-Affibody was engineered to find a suitable linker and substrate peptide, and the different constructs were evaluated with a new bacterial display assay. HER2-binding of the pro-Affibody was efficiently masked and proteolytic activation of the best variant yielded over 1,000-fold increase in apparent binding affinity. Biosensor analysis revealed that blocking of the pro-Affibody primarily affected the association phase. In a cell-binding assay, the activated pro-Affibody targeted native HER2 on cancer cells as opposed to the non-activated pro-Affibody. We believe this prodrug approach with proteolytic activation is promising for improving tissue specificity in future in vivo targeting applications and can hopefully be extended to other Affibody molecules and similar affinity proteins as well.


Subject(s)
Neoplasms/metabolism , Peptide Hydrolases/metabolism , Prodrugs/metabolism , Recombinant Fusion Proteins/metabolism , Binding, Competitive/immunology , Cell Line, Tumor , Flow Cytometry , Humans , Matrix Metalloproteinase 1/metabolism , Neoplasms/enzymology , Neoplasms/immunology , Prodrugs/chemistry , Protein Binding/immunology , Proteolysis , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Surface Plasmon Resonance
10.
J Biol Chem ; 289(2): 594-9, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24302742

ABSTRACT

Immunotherapies and vaccines based on the induction of broadly neutralizing monoclonal antibodies (bNAbs) have become outstanding strategies against HIV-1. Diverse bNAbs recognizing different regions of the HIV-1 envelope have been identified and extensively studied. However, there is little information about the thermodynamics of binding of these bNAbs and their epitopes. We used isothermal titration calorimetry to characterize thermodynamically the interactions between bNAb2F5 (in both the IgG and Fab forms) and its functional and core epitope peptides. We found that these interactions are enthalpically driven and opposed by a negative entropy change. The highest affinity was found for 2F5 IgG for its functional epitope, indicating that additional interactions involving residues flanking the core epitope contribute strongly to higher affinity. In addition, the strong influence of the Fc region on the binding affinity suggests long-range allosteric effects within IgG. Our results provide useful information for developing new therapeutics against HIV-1 and, in a broader scope, contribute to a better understanding of antigen-antibody recognition.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Epitopes/metabolism , Immunoglobulin Fc Fragments/metabolism , Thermodynamics , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibody Affinity/immunology , Binding, Competitive/immunology , Calorimetry/methods , Epitopes/chemistry , Epitopes/immunology , HIV Envelope Protein gp41/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Molecular Sequence Data , Protein Binding/immunology
11.
J Biol Chem ; 289(36): 25374-81, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25053417

ABSTRACT

Shiga toxin Stx2e is the major known agent that causes edema disease in newly weaned pigs. This severe disease is characterized by neurological disorders, hemorrhagic lesions, and frequent fatal outcomes. Stx2e consists of an enzymatically active A subunit and five B subunits that bind to a specific glycolipid receptor on host cells. It is evident that antibodies binding to the A subunit or the B subunits of Shiga toxin variants may have the capability to inhibit their cytotoxicity. Here, we report the discovery and characterization of a VHH single domain antibody (nanobody) isolated from a llama phage display library that confers potent neutralizing capacity against Stx2e toxin. We further present the crystal structure of the complex formed between the nanobody (NbStx2e1) and the Stx2e toxoid, determined at 2.8 Å resolution. Structural analysis revealed that for each B subunit of Stx2e, one NbStx2e1 is interacting in a head-to-head orientation and directly competing with the glycolipid receptor binding site on the surface of the B subunit. The neutralizing NbStx2e1 can in the future be used to prevent or treat edema disease.


Subject(s)
Antibodies, Neutralizing/chemistry , Protein Structure, Tertiary , Shiga Toxin 2/chemistry , Single-Domain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Binding Sites/genetics , Binding Sites/immunology , Binding, Competitive/immunology , Camelids, New World/immunology , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Binding/immunology , Protein Subunits/chemistry , Protein Subunits/immunology , Protein Subunits/metabolism , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Shiga Toxin 2/immunology , Shiga Toxin 2/metabolism , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
12.
Eur J Immunol ; 44(4): 1058-68, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24510524

ABSTRACT

A key component of immunity against viruses, CD4(+) T cells expand and differentiate into functional subsets upon primary infection, where effector (Teff) cells facilitate infection control and regulatory (Treg) cells mitigate immunopathology. After secondary infection, Teff cells mount a robust response from the memory pool. Here, we show that Treg-cell responses are diminished upon secondary infection, and Treg-cell response dynamics are associated more with T-cell receptors (TCRs) repertoire and avidity than with epitope specificity. In the murine model, the IA(b) M209 epitope of respiratory syncytial virus is recognized by both CD4(+) Treg and Teff cells, while the IA(b) M226 epitope is recognized almost exclusively by CD4(+) Teff cells expressing high avidity TCR Vß8.1/8.2 and dominating the CD4(+) T-cell response during primary and secondary infections. IA(b) M209 -Teff cells express relatively low avidity TCRs during early primary infection, but high avidity TCR Vß7-expressing IA(b) M209 -Teff cells emerge during the late phase, and become dominant after secondary infection. The emerging high avidity IA(b) M209 -Teff cells outcompete IA(b) M209 -Treg cells that share the same epitope, but have low avidity and are restricted to TCR Vß2 and Vß6 subpopulations. These data indicate that MHC-peptide-TCR interactions can produce different kinetic and functional profiles in CD4(+) T-cell populations even when responding to the same epitope.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Binding, Competitive/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/virology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , Host-Pathogen Interactions/immunology , Humans , Kinetics , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology
13.
Blood ; 121(22): 4484-92, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23610371

ABSTRACT

Interleukin-7 (IL-7) is essential to T-cell survival as well as homeostatic proliferation, and clinical trials that exploit the mitogenic effects of IL-7 have achieved success in treating human diseases. In mice, the in vivo potency of IL-7 improves dramatically when it is administered as a complex with the anti-IL-7 neutralizing monoclonal antibody clone M25. However, the mechanism whereby M25 augments IL-7 potency is unknown. We have analyzed the discrete contributions of the antibody constant (Fc) and IL-7-binding (Fab) domains to the mechanism. By engaging the neonatal Fc receptor the Fc domain extends the in vivo lifespan of IL-7/M25 complexes and accounts for the majority of their activity. Unexpectedly, the IL-7-neutralizing Fab domain provides an additional, albeit smaller, contribution, possibly by serving as a cytokine depot. This study is the first to demonstrate that the neutralizing aspect of the monoclonal antibody is directly involved in enhancing the potency of a cytokine with a single form of receptor. Lessons from the mechanism of IL-7/M25 complexes inform the design of next-generation cytokine therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Benzofurans , Binding, Competitive/immunology , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Interleukin-7/metabolism , Male , Mice , Mice, Transgenic , Quinolines , Receptors, Fc/immunology , Receptors, Fc/metabolism , Receptors, Interleukin-7/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
J Biol Chem ; 288(28): 20520-31, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720782

ABSTRACT

Members of a family of collagen-binding microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) from Gram-positive bacteria are established virulence factors in several infectious diseases models. Here, we report that these adhesins also can bind C1q and act as inhibitors of the classical complement pathway. Molecular analyses of Cna from Staphylococcus aureus suggested that this prototype MSCRAMM bound to the collagenous domain of C1q and interfered with the interactions of C1r with C1q. As a result, C1r2C1s2 was displaced from C1q, and the C1 complex was deactivated. This novel function of the Cna-like MSCRAMMs represents a potential immune evasion strategy that could be used by numerous Gram-positive pathogens.


Subject(s)
Adhesins, Bacterial/immunology , Complement Activation/immunology , Complement Pathway, Classical/immunology , Gram-Positive Bacteria/immunology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Binding, Competitive/immunology , Collagen/immunology , Collagen/metabolism , Complement C1q/immunology , Complement C1q/metabolism , Complement C1r/immunology , Complement C1r/metabolism , Enzyme-Linked Immunosorbent Assay , Gram-Positive Bacteria/metabolism , History, 18th Century , Humans , Immunoblotting , Kinetics , Models, Molecular , Protein Binding/immunology , Protein Structure, Tertiary , Surface Plasmon Resonance
15.
J Biol Chem ; 288(30): 21569-80, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23754290

ABSTRACT

The staphylococcal superantigen-like protein (SSL) family is composed of 14 exoproteins sharing structural similarity with superantigens but no superantigenic activity. Target proteins of four SSLs have been identified to be involved in host immune responses. However, the counterparts of other SSLs have been functionally uncharacterized. In this study, we have identified porcine plasma prothrombin as SSL10-binding protein by affinity purification using SSL10-conjugated Sepharose. The resin recovered the prodomain of prothrombin (fragment 1 + 2) as well as factor Xa in pull-down analysis. The equilibrium dissociation constant between SSL10 and prothrombin was 1.36 × 10(-7) M in surface plasmon resonance analysis. On the other hand, the resin failed to recover γ-carboxyglutamic acid (Gla) domain-less coagulation factors and prothrombin from warfarin-treated mice, suggesting that the Gla domain of the coagulation factors is essential for the interaction. SSL10 prolonged plasma clotting induced by the addition of Ca(2+) and factor Xa. SSL10 did not affect the protease activity of thrombin but inhibited the generation of thrombin activity in recalcified plasma. S. aureus produces coagulase that non-enzymatically activates prothrombin. SSL10 attenuated clotting induced by coagulase, but the inhibitory effect was weaker than that on physiological clotting, and SSL10 did not inhibit protease activity of staphylothrombin, the complex of prothrombin with coagulase. These results indicate that SSL10 inhibits blood coagulation by interfering with activation of coagulation cascade via binding to the Gla domain of coagulation factor but not by directly inhibiting thrombin activity. This is the first finding that the bacterial protein inhibits blood coagulation via targeting the Gla domain of coagulation factors.


Subject(s)
1-Carboxyglutamic Acid/immunology , Bacterial Proteins/immunology , Factor Xa/immunology , Prothrombin/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , 1-Carboxyglutamic Acid/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/metabolism , Binding Sites/immunology , Binding, Competitive/immunology , Blood Coagulation/immunology , Calcium/immunology , Calcium/metabolism , Coagulase/immunology , Coagulase/metabolism , Electrophoresis, Polyacrylamide Gel , Factor Xa/metabolism , Humans , Immune Sera/immunology , Immune Sera/metabolism , Mice , Molecular Sequence Data , Protein Binding/immunology , Prothrombin/metabolism , Staphylococcus aureus/metabolism , Superantigens/metabolism , Surface Plasmon Resonance , Swine , Thrombin/immunology , Thrombin/metabolism
16.
J Biol Chem ; 288(41): 29562-72, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23979133

ABSTRACT

The chemokine receptor CXCR7, belonging to the membrane-bound G protein-coupled receptor superfamily, is expressed in several tumor types. Inhibition of CXCR7 with either small molecules or small interference (si)RNA has shown promising therapeutic benefits in several tumor models. With the increased interest and effectiveness of biologicals inhibiting membrane-bound receptors we made use of the "Nanobody platform" to target CXCR7. Previously we showed that Nanobodies, i.e. immunoglobulin single variable domains derived from naturally occurring heavy chain-only camelids antibodies, represent new biological tools to efficiently tackle difficult drug targets such as G protein-coupled receptors. In this study we developed and characterized highly selective and potent Nanobodies against CXCR7. Interestingly, the CXCR7-targeting Nanobodies displayed antagonistic properties in contrast with previously reported CXCR7-targeting agents. Several high affinity CXCR7-specific Nanobodies potently inhibited CXCL12-induced ß-arrestin2 recruitment in vitro. A wide variety of tumor biopsies was profiled, showing for the first time high expression of CXCR7 in head and neck cancer. Using a patient-derived CXCR7-expressing head and neck cancer xenograft model in nude mice, tumor growth was inhibited by CXCR7-targeting Nanobody therapy. Mechanistically, CXCR7-targeting Nanobodies did not inhibit cell cycle progression but instead reduced secretion of the angiogenic chemokine CXCL1 from head and neck cancer cells in vitro, thus acting here as inverse agonists, and subsequent angiogenesis in vivo. Hence, with this novel class of CXCR7 inhibitors, we further substantiate the therapeutic relevance of targeting CXCR7 in head and neck cancer.


Subject(s)
Head and Neck Neoplasms/immunology , Receptors, CXCR/immunology , Single-Domain Antibodies/immunology , Xenograft Model Antitumor Assays , Animals , Arrestins/immunology , Arrestins/metabolism , Binding, Competitive/immunology , Camelids, New World/immunology , Cell Line, Tumor , Chemokine CXCL12/pharmacology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/prevention & control , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Radioligand Assay , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Domain Antibodies/pharmacology , Tumor Burden/drug effects , Tumor Burden/immunology , beta-Arrestins
17.
FASEB J ; 27(8): 3272-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23671272

ABSTRACT

Dendritic cells (DCs) initiate host immune responses by presenting captured antigens to naive T cells. Hence, DC-binding peptides may be used for antigen targeting to boost naive and memory immune responses. By biopanning peptide phage libraries on human monocyte-derived DCs, we identified novel DC-binding peptides. One of the selected phages, displaying the NW peptide (NWYLPWLGTNDW), bound DCs with high affinity, and its binding was inhibited by the corresponding synthetic peptide. Antigenic peptides or proteins conjugated to the NW peptide bound to DCs and were internalized without negative effects on DC phenotype and function. Ex vivo targeted delivery of CMV-pp65 peptides to DCs via the NW peptide increased T-cell responses in HLA-A2(+)/CMV(+) donors compared to untargeted peptides (P<0.001). Stimulation of CD45RO-depleted peripheral blood mononuclear cells from CMV(-) donors with the NW-pp65 fusion peptides expanded pp65-specific precursor T cells. Moreover, the NW peptide mediated small interfering RNA delivery to DCs, and a significant gene silencing was obtained. Collectively, the data reveal that proteins and nucleic acids can be directed to DCs through the NW peptide, enabling effective uptake and functional effects such as T-cell activation in the context of MHC class I and II molecules.


Subject(s)
Antigens/immunology , Dendritic Cells/immunology , Nucleic Acids/immunology , Oligopeptides/immunology , Amino Acid Sequence , Antigen Presentation/immunology , Antigens/metabolism , Binding, Competitive/immunology , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Flow Cytometry , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Microscopy, Fluorescence , Monocytes/immunology , Monocytes/metabolism , Nucleic Acids/metabolism , Oligopeptides/metabolism , Phosphoproteins/immunology , Phosphoproteins/metabolism , Protein Binding/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism
18.
J Immunol ; 188(5): 2455-63, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22271652

ABSTRACT

The phenotype of B cells responsible for the production of anti-pneumococcal polysaccharide Ab has been unclear. Although individuals that respond poorly to the 23-valent pneumococcal polysaccharide (PPS) vaccine, Pneumovax, such as children <2 y, the asplenic, and a subset of common variable immunodeficiency patients, are profoundly deficient or lack IgM memory cells (CD27(+)IgM(+)), they are also deficient in the switched memory (CD27(+)IgM(-)) compartment. Direct characterization of PPS-specific B cells has not been performed. In this study, we labeled PPS14 and PPS23F with fluorescent markers. Fluorescently labeled PPS were used in FACSAria flow cytometry to characterize the phenotype of PPS-specific B cells obtained from 18 young adults pre- and postimmunization with Pneumovax. The labeled PPS were capable of inhibiting binding of Ab to the native PPS. Similarly, the native PPS were able to inhibit binding of PPS-specific B cells in a flow cytometric assay demonstrating specificity and functionality. Phenotypic analysis of unselected B cells, pre- and postimmunization, demonstrated a predominance of naive CD27(-)IgM(+) cells accounting for 61.5% of B cells. Likewise, the PPS-specific B cells obtained preimmunization consisted primarily of naive, CD27(-) B cells, 55.4-63.8%. In contrast, the PPS-specific B cells obtained postimmunization were predominantly IgM memory cells displaying the CD27(+)IgM(+), 54.2% for PPS14 and 66% for PPS23F, significantly higher than both unselected B cells and PPS-specific B cells. There was no significant difference in switched memory B cell populations (CD27(+)IgM(-)) between groups. These results suggest a dominant role of IgM memory cells in the immune response to pneumococcal polysaccharides.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Immunophenotyping/methods , Pneumococcal Vaccines/immunology , Adolescent , Adult , Animals , Bacterial Capsules/immunology , Bacterial Capsules/metabolism , Binding Sites, Antibody , Binding, Competitive/immunology , Humans , Hybridomas , Immunoglobulin A/biosynthesis , Immunoglobulin A/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/metabolism , Immunoglobulin M/biosynthesis , Immunoglobulin M/metabolism , Immunologic Memory , Mice , Pneumococcal Vaccines/metabolism , Young Adult
19.
J Immunol ; 188(11): 5283-92, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544926

ABSTRACT

Vascular endothelial cells (EC) are an exposed tissue with intimate contact with circulating Ag-specific CTL. Experimental in vitro and clinical data suggested that endothelial cells present a different repertoire of MHC class I-restricted peptides compared with syngeneic leukocytes or epithelial cells. This endothelial-specific peptide repertoire might protect EC from CTL-mediated cell death. The HLA-A*02-restricted peptide profile of human EC and syngeneic B lymphoblastoid cells was biochemically analyzed and compared. For EC selective peptides, source protein expression, peptide binding affinity, and peptide-HLA-A*02 turnover were measured. The significance of abundant peptide presentation for target cell recognition by immunodominant CTL was tested by small interfering RNA treatment of EC to knock down the source proteins. High amounts of two peptides, PTRF(56-64) and CD59(106-114), were consistently detected in EC. This predominance of two endothelial peptides was explained by cell type-specific source protein expression that compensated for poor HLA-A*02 binding affinity and short half-live of peptide/HLA-A*02 complexes. Knocking down the source proteins containing the abundant endothelial peptide motifs led to a nearly 100-fold increase of surface expression of SMCY(311-319), an immunodominant minor histocompatibility Ag, as detected by cytotoxicity assays using SMCY(311-319)-specific CTL. We conclude that EC express and present preferentially two distinct HLA-A*02-restricted peptides at extraordinary high levels. These abundant self-peptides may protect EC from CTL-mediated lysis by competing for HLA-A*02 binding sites with immunodominant scarcely expressed antigenic peptides.


Subject(s)
Endothelium, Vascular/immunology , HLA-A2 Antigen/physiology , T-Lymphocytes, Cytotoxic/immunology , Binding, Competitive/immunology , Cell Line, Tumor , Cells, Cultured , Cytotoxicity Tests, Immunologic , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , HLA-A2 Antigen/biosynthesis , HLA-A2 Antigen/metabolism , Humans , Immunodominant Epitopes/biosynthesis , Immunodominant Epitopes/metabolism , Immunodominant Epitopes/physiology , Peptide Fragments/biosynthesis , Peptide Fragments/metabolism , Peptide Fragments/physiology , Protein Binding/immunology , Spectrometry, Mass, Electrospray Ionization , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology
20.
J Immunol ; 189(8): 3983-94, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22966084

ABSTRACT

HLA-DM (DM) is a nonclassical MHC class II (MHC II) protein that acts as a peptide editor to mediate the exchange of peptides loaded onto MHC II during Ag presentation. Although the ability of DM to promote peptide exchange in vitro and in vivo is well established, the role of DM in epitope selection is still unclear, especially in human response to infectious disease. In this study, we addressed this question in the context of the human CD4 T cell response to vaccinia virus. We measured the IC(50), intrinsic dissociation t(1/2), and DM-mediated dissociation t(1/2) for a large set of peptides derived from the major core protein A10L and other known vaccinia epitopes bound to HLA-DR1 and compared these properties to the presence and magnitude of peptide-specific CD4(+) T cell responses. We found that MHC II-peptide complex kinetic stability in the presence of DM distinguishes T cell epitopes from nonrecognized peptides in A10L peptides and also in a set of predicted tight binders from the entire vaccinia genome. Taken together, these analyses demonstrate that DM-mediated dissociation t(1/2) is a strong and independent factor governing peptide immunogenicity by favoring the presentation of peptides with greater kinetic stability in the presence of DM.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/immunology , HLA-D Antigens/physiology , Peptide Fragments/metabolism , Vaccinia virus/immunology , Amino Acid Sequence , Binding, Competitive/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Epitopes, T-Lymphocyte/metabolism , HLA-D Antigens/chemistry , Half-Life , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Binding/immunology , Viral Proteins/chemistry , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL