Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1170462, 2023.
Article in English | MEDLINE | ID: mdl-37207206

ABSTRACT

MHC class I "single-chain trimer" molecules, coupling MHC heavy chain, ß2-microglobulin, and a specific peptide into a single polypeptide chain, are widely used in research. To more fully understand caveats associated with this design that may affect its use for basic and translational studies, we evaluated a set of engineered single-chain trimers with combinations of stabilizing mutations across eight different classical and non-classical human class I alleles with 44 different peptides, including a novel human/murine chimeric design. While, overall, single-chain trimers accurately recapitulate native molecules, care was needed in selecting designs for studying peptides longer or shorter than 9-mers, as single-chain trimer design could affect peptide conformation. In the process, we observed that predictions of peptide binding were often discordant with experiment and that yields and stabilities varied widely with construct design. We also developed novel reagents to improve the crystallizability of these proteins and confirmed novel modes of peptide presentation.


Subject(s)
Histocompatibility Antigens Class I , Peptides , Humans , Mice , Animals , Histocompatibility Antigens Class I/genetics , Peptides/metabolism , Epitopes/chemistry
2.
Commun Biol ; 6(1): 528, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37193826

ABSTRACT

The discovery and characterization of antigen-specific CD8+ T cell clonotypes typically involves the labor-intensive synthesis and construction of peptide-MHC tetramers. We adapt single-chain trimer (SCT) technologies into a high throughput platform for pMHC library generation, showing that hundreds can be rapidly prepared across multiple Class I HLA alleles. We use this platform to explore the impact of peptide and SCT template mutations on protein expression yield, thermal stability, and functionality. SCT libraries were an efficient tool for identifying T cells recognizing commonly reported viral epitopes. We then construct SCT libraries to capture SARS-CoV-2 specific CD8+ T cells from COVID-19 participants and healthy donors. The immunogenicity of these epitopes is validated by functional assays of T cells with cloned TCRs captured using SCT libraries. These technologies should enable the rapid analyses of peptide-based T cell responses across several contexts, including autoimmunity, cancer, or infectious disease.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Humans , SARS-CoV-2/genetics , Antigens , Epitopes , Peptides/genetics
3.
Res Sq ; 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36415462

ABSTRACT

CD8 + cytotoxic T cell responses against viral infection represent a major element of the adaptive immune response. We describe the development of a peptide antigen - major histompatibility complex (pMHC) library representing the full SARS-CoV-2 viral proteome, and comprised of 634 pMHC multimers representing the A*02.01, A*24.02, and B*07.02 HLA alleles, as well as specific antigens associated with the cytomegalovirus (CMV). These libraries were used to capture non-expanded CD8 + T cells from blood samples collected from 64 infected individuals, and then analyzed using single cell RNA-seq. The discovery and characterization of antigen-specific CD8 + T cell clonotypes typically involves the labor-intensive synthesis and construction of peptide-MHC tetramers. We adapted single-chain trimer (SCT) technologies into a high throughput platform for pMHC library generation, showing that hundreds can be rapidly prepared across multiple Class I HLA alleles. We used this platform to explore the impact of peptide and SCT template mutations on protein expression yield, thermal stability, and functionality. SCT libraries were an efficient tool for identifying T cells recognizing commonly reported viral epitopes. We then constructed SCT libraries designed to capture SARS-CoV-2 specific CD8 + T cells from COVID-19 participants and healthy donors. The immunogenicity of these epitopes was validated by functional assays of T cells with cloned TCRs captured using SCT libraries. These technologies should enable the rapid analyses of peptide-based T cell responses across several contexts, including autoimmunity, cancer, or infectious disease.

4.
Proc Natl Acad Sci U S A ; 119(31): e2203410119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35878026

ABSTRACT

Tissue-specific antigens can serve as targets for adoptive T cell transfer-based cancer immunotherapy. Recognition of tumor by T cells is mediated by interaction between peptide-major histocompatibility complexes (pMHCs) and T cell receptors (TCRs). Revealing the identity of peptides bound to MHC is critical in discovering cognate TCRs and predicting potential toxicity. We performed multimodal immunopeptidomic analyses for human prostatic acid phosphatase (PAP), a well-recognized tissue antigen. Three physical methods, including mild acid elution, coimmunoprecipitation, and secreted MHC precipitation, were used to capture a thorough signature of PAP on HLA-A*02:01. Eleven PAP peptides that are potentially A*02:01-restricted were identified, including five predicted strong binders by NetMHCpan 4.0. Peripheral blood mononuclear cells (PBMCs) from more than 20 healthy donors were screened with the PAP peptides. Seven cognate TCRs were isolated which can recognize three distinct epitopes when expressed in PBMCs. One TCR shows reactivity toward cell lines expressing both full-length PAP and HLA-A*02:01. Our results show that a combined multimodal immunopeptidomic approach is productive in revealing target peptides and defining the cloned TCR sequences reactive with prostatic acid phosphatase epitopes.


Subject(s)
Acid Phosphatase , Antigens, Neoplasm , Receptors, Antigen, T-Cell , Acid Phosphatase/metabolism , Antigens, Neoplasm/metabolism , Epitopes , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Leukocytes, Mononuclear , Neoplasms/immunology , Peptides , Receptors, Antigen, T-Cell/metabolism
5.
Front Immunol ; 12: 658372, 2021.
Article in English | MEDLINE | ID: mdl-33986749

ABSTRACT

Conventional immunoprecipitation/mass spectroscopy identification of HLA-restricted peptides remains the purview of specializing laboratories, due to the complexity of the methodology, and requires computational post-analysis to assign peptides to individual alleles when using pan-HLA antibodies. We have addressed these limitations with ARTEMIS: a simple, robust, and flexible platform for peptide discovery across ligandomes, optionally including specific proteins-of-interest, that combines novel, secreted HLA-I discovery reagents spanning multiple alleles, optimized lentiviral transduction, and streamlined affinity-tag purification to improve upon conventional methods. This platform fills a middle ground between existing techniques: sensitive and adaptable, but easy and affordable enough to be widely employed by general laboratories. We used ARTEMIS to catalog allele-specific ligandomes from HEK293 cells for seven classical HLA alleles and compared results across replicates, against computational predictions, and against high-quality conventional datasets. We also applied ARTEMIS to identify potentially useful, novel HLA-restricted peptide targets from oncovirus oncoproteins and tumor-associated antigens.


Subject(s)
Epitope Mapping/methods , Mass Spectrometry/methods , Peptides/chemistry , Peptides/immunology , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Mice , Models, Molecular , Protein Binding , Reproducibility of Results , Structure-Activity Relationship , Workflow
6.
J Mol Biol ; 432(14): 3989-4009, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32304700

ABSTRACT

The impenetrability of the blood-brain barrier (BBB) to most conventional drugs impedes the treatment of central nervous system (CNS) disorders. Interventions for diseases like brain cancer, neurodegeneration, or age-associated inflammatory processes require varied approaches to CNS drug delivery. Cystine-dense peptides (CDPs) have drawn recent interest as drugs or drug-delivery vehicles. Found throughout the phylogenetic tree, often in drug-like roles, their size, stability, and protein interaction capabilities make CDPs an attractive mid-size biologic scaffold to complement conventional antibody-based drugs. Here, we describe the identification, maturation, characterization, and utilization of a CDP that binds to the transferrin receptor (TfR), a native receptor and BBB transporter for the iron chaperone transferrin. We developed variants with varying binding affinities (KD as low as 216 pM), co-crystallized it with the receptor, and confirmed murine cross-reactivity. It accumulates in the mouse CNS at ~25% of blood levels (CNS blood content is only ~1%-6%) and delivers neurotensin, an otherwise non-BBB-penetrant neuropeptide, at levels capable of modulating CREB signaling in the mouse brain. Our work highlights the utility of CDPs as a diverse, easy-to-screen scaffold family worthy of inclusion in modern drug discovery strategies, demonstrated by the discovery of a candidate CNS drug delivery vehicle ready for further optimization and preclinical development.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System Diseases/drug therapy , Drug Delivery Systems , Peptides/pharmacology , Animals , Antigens, CD/chemistry , Antigens, CD/drug effects , Antigens, CD/genetics , Antigens, CD/pharmacology , Central Nervous System/drug effects , Cystine/chemistry , Cystine/genetics , Humans , Inflammation/drug therapy , Inflammation/pathology , Mice , Neuropeptides/chemistry , Neuropeptides/pharmacology , Neurotensin/chemistry , Neurotensin/pharmacology , Peptides/chemistry , Protein Binding/drug effects , Receptors, Transferrin/chemistry , Receptors, Transferrin/drug effects , Receptors, Transferrin/genetics
7.
PLoS One ; 11(9): e0163297, 2016.
Article in English | MEDLINE | ID: mdl-27649529

ABSTRACT

Based on previous findings supporting HLA-F as a ligand for KIR3DL2 and KIR2DS4, we investigated the potential for MHC-I open conformers (OCs) as ligands for KIR3DS1 and KIR3DL1 through interactions measured by surface plasmon resonance. These measurements showed physical binding of KIR3DS1 but not KIR3DL1 with HLA-F and other MHC-I OC while also confirming the allotype specific binding of KIR3DL1 with MHC-I peptide complex. Concordant results were obtained with biochemical pull-down from cell lines and biochemical heterodimerization experiments with recombinant proteins. In addition, surface binding of HLA-F and KIR3DS1 to native and activated NK and T cells was coincident with specific expression of the putative ligand or receptor. A functional response of KIR3DS1 was indicated by increased granule exocytosis in activated cells incubated with HLA-F bound to surfaces. The data extend a model for interaction between MHC-I open conformers and activating KIR receptors expressed during an inflammatory response, potentially contributing to communication between the innate and adaptive immune response.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/metabolism , Receptors, KIR3DS1/metabolism , Humans , Killer Cells, Natural/cytology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Models, Molecular , Protein Binding , Protein Folding
9.
PLoS Pathog ; 10(9): e1004403, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25254371

ABSTRACT

The process of antibody ontogeny typically improves affinity, on-rate, and thermostability, narrows polyspecificity, and rigidifies the combining site to the conformer optimal for binding from the broader ensemble accessible to the precursor. However, many broadly-neutralizing anti-HIV antibodies incorporate unusual structural elements and recognition specificities or properties that often lead to autoreactivity. The ontogeny of 4E10, an autoreactive antibody with unexpected combining site flexibility, was delineated through structural and biophysical comparisons of the mature antibody with multiple potential precursors. 4E10 gained affinity primarily by off-rate enhancement through a small number of mutations to a highly conserved recognition surface. Controverting the conventional paradigm, the combining site gained flexibility and autoreactivity during ontogeny, while losing thermostability, though polyspecificity was unaffected. Details of the recognition mechanism, including inferred global effects due to 4E10 binding, suggest that neutralization by 4E10 may involve mechanisms beyond simply binding, also requiring the ability of the antibody to induce conformational changes distant from its binding site. 4E10 is, therefore, unlikely to be re-elicited by conventional vaccination strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibody Specificity , HIV Antibodies/immunology , HIV Antibodies/metabolism , HIV Infections/immunology , HIV-1/immunology , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Broadly Neutralizing Antibodies , Crystallography, X-Ray , HIV Antibodies/chemistry , HIV Infections/virology , Humans , Molecular Sequence Data , Neutralization Tests , Sequence Homology, Amino Acid , Surface Plasmon Resonance
10.
PLoS Pathog ; 9(9): e1003639, 2013.
Article in English | MEDLINE | ID: mdl-24086134

ABSTRACT

The broadly-neutralizing anti-HIV antibody 4E10 recognizes an epitope in the membrane-proximal external region of the HIV envelope protein gp41. Previous attempts to elicit 4E10 by vaccination with envelope-derived or reverse-engineered immunogens have failed. It was presumed that the ontogeny of 4E10-equivalent responses was blocked by inherent autoreactivity and exceptional polyreactivity. We generated 4E10 heavy-chain knock-in mice, which displayed significant B cell dysregulation, consistent with recognition of autoantigen/s by 4E10 and the presumption that tolerance mechanisms may hinder the elicitation of 4E10 or 4E10-equivalent responses. Previously proposed candidate 4E10 autoantigens include the mitochondrial lipid cardiolipin and a nuclear splicing factor, 3B3. However, using carefully-controlled assays, 4E10 bound only weakly to cardiolipin-containing liposomes, but also bound negatively-charged, non-cardiolipin-containing liposomes comparably poorly. 4E10/liposome binding was predominantly mediated by electrostatic interactions rather than presumed hydrophobic interactions. The crystal structure of 4E10 free of bound ligands showed a dramatic restructuring of the combining site, occluding the HIV epitope binding site and revealing profound flexibility, but creating an electropositive pocket consistent with non-specific binding of phospholipid headgroups. These results strongly suggested that antigens other than cardiolipin mediate 4E10 autoreactivity. Using a synthetic peptide library spanning the human proteome, we determined that 4E10 displays limited and focused, but unexceptional, polyspecificity. We also identified a novel autoepitope shared by three ER-resident inositol trisphosphate receptors, validated through binding studies and immunohistochemistry. Tissue staining with 4E10 demonstrated reactivity consistent with the type 1 inositol trisphosphate receptor as the most likely candidate autoantigen, but is inconsistent with splicing factor 3B3. These results demonstrate that 4E10 recognition of liposomes competes with MPER recognition and that HIV antigen and autoepitope recognition may be distinct enough to permit eliciting 4E10-like antibodies, evading autoimmunity through directed engineering. However, 4E10 combining site flexibility, exceptional for a highly-matured antibody, may preclude eliciting 4E10 by conventional immunization strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Autoantibodies/immunology , Autoantigens/immunology , Complementarity Determining Regions/immunology , Epitopes/immunology , HIV Antibodies/immunology , HIV-1/immunology , Immunoglobulin Heavy Chains/immunology , Inositol 1,4,5-Trisphosphate Receptors/immunology , Animals , Antibodies, Monoclonal/genetics , Autoantibodies/genetics , Autoantigens/genetics , Broadly Neutralizing Antibodies , Cardiolipins/genetics , Cardiolipins/immunology , Complementarity Determining Regions/genetics , Epitopes/genetics , HIV Antibodies/genetics , Humans , Immunoglobulin Heavy Chains/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Transgenic , Proteome/genetics , Proteome/immunology
11.
Immunol Rev ; 250(1): 239-57, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23046134

ABSTRACT

Natural killer (NK) cells are key components of innate immune responses, providing surveillance against cells undergoing tumorigenesis or infection, by viruses or internal pathogens. NK cells can directly eliminate compromised cells and regulate downstream responses of the innate and acquired immune systems through the release of immune modulators (cytokines, interferons). The importance of the role NK cells play in immune defense was demonstrated originally in herpes viral infections, usually mild or localized, which become severe and life threatening in NK-deficient patients . NK cell effector functions are governed by balancing opposing signals from a diverse array of activating and inhibitory receptors. Many NK receptors occur in paired activating and inhibitory isoforms and recognize major histocompatibility complex (MHC) class I proteins with varying degrees of peptide specificity. Structural studies have made considerable inroads into understanding the molecular mechanisms employed to broadly recognize multiple MHC ligands or specific pathogen-associated antigens and the strategies employed by viruses to thwart these defenses. Although many details of NK development, signaling, and integration remain mysterious, it is clear that NK receptors are key components of a system exquisitely tuned to sense any dysregulation in MHC class I expression, or the expression of certain viral antigens, resulting in the elimination of affected cells.


Subject(s)
Antigens, Viral/chemistry , Histocompatibility Antigens Class I/chemistry , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/chemistry , Virus Diseases/immunology , Viruses/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Cytokines/immunology , Gene Expression , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immune Evasion , Immunity, Innate , Killer Cells, Natural/virology , Models, Molecular , Protein Binding , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Signal Transduction , T-Cell Antigen Receptor Specificity , Virus Diseases/virology
12.
Ann Biomed Eng ; 37(7): 1464-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19365729

ABSTRACT

In the present study, Raman spectroscopy is employed to assess the potential toxicity of chemical substances. Having several advantages compared to other traditional methods, Raman spectroscopy is an ideal solution for investigating cells in their natural environment. In the present work, we combine the power of spectral resolution of Raman with one of the most widely used machine learning techniques. Support vector machines (SVMs) are used in the context of classification on a well established database. The database is constructed on three different classes: healthy cells, Triton X-100 (necrotic death), and etoposide (apoptotic death). SVM classifiers successfully assess the potential effect of the test toxins (Triton X-100, etoposide). The cells that are exposed to heat (45 degrees C) are tested using the classification rules obtained. It is shown that the heat effect results in apoptotic death, which is in agreement with existing literature.


Subject(s)
Algorithms , Apoptosis/physiology , Artificial Intelligence , Epithelial Cells/cytology , Epithelial Cells/physiology , Pattern Recognition, Automated/methods , Spectrum Analysis, Raman/methods , Cells, Cultured , Humans
13.
Antimicrob Agents Chemother ; 51(10): 3688-98, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17682095

ABSTRACT

The bacterial type II topoisomerases DNA gyrase and topoisomerase IV are validated targets for clinically useful quinolone antimicrobial drugs. A significant limitation to widely utilized quinolone inhibitors is the emergence of drug-resistant bacteria due to an altered DNA gyrase. To address this problem, we have used structure-based molecular docking to identify novel drug-like small molecules that target sites distinct from those targeted by quinolone inhibitors. A chemical ligand database containing approximately 140,000 small molecules (molecular weight, <500) was molecularly docked onto two sites of Escherichia coli DNA gyrase targeting (i) a previously unexplored structural pocket formed at the dimer interface of subunit A and (ii) a small region of the ATP binding pocket on subunit B overlapping the site targeted by coumarin and cyclothialidine drugs. This approach identified several small-molecule compounds that inhibited the DNA supercoiling activity of purified E. coli DNA gyrase. These compounds are structurally unrelated to previously identified gyrase inhibitors and represent potential scaffolds for the optimization of novel antibacterial agents that act on fluoroquinolone-resistant strains.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Computer Simulation , Crystallization , DNA Gyrase/chemistry , DNA Helicases/chemistry , DNA, Superhelical/drug effects , Databases, Genetic , Drug Design , Drug Evaluation, Preclinical , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/growth & development , Indicators and Reagents , Molecular Conformation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...