Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
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
2.
Mol Ther ; 31(10): 2901-2913, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37550965

ABSTRACT

Hematopoietic stem cell (HSC) gene therapy is currently performed on CD34+ hematopoietic stem and progenitor cells containing less than 1% true HSCs and requiring a highly specialized infrastructure for cell manufacturing and transplantation. We have previously identified the CD34+CD90+ subset to be exclusively responsible for short- and long-term engraftment. However, purification and enrichment of this subset is laborious and expensive. HSC-specific delivery agents for the direct modification of rare HSCs are currently lacking. Here, we developed novel targeted viral vectors to specifically transduce CD90-expressing HSCs. Anti-CD90 single chain variable fragments (scFvs) were engineered onto measles- and VSV-G-pseudotyped lentiviral vectors that were knocked out for native targeting. We further developed a custom hydrodynamic titration methodology to assess the loading of surface-engineered capsids, measure antigen recognition of the scFv, and predict the performance on cells. Engineered vectors formed with minimal impairment in the functional titer, maintained their ability to fuse with the target cells, and showed highly specific recognition of CD90 on cells ex vivo. Most important, targeted vectors selectively transduced human HSCs with secondary colony-forming potential. Our novel HSC-targeted viral vectors have the potential to significantly enhance the feasibility of ex vivo gene therapy and pave the way for future in vivo applications.


Subject(s)
Hematopoietic Stem Cell Transplantation , Humans , Antigens, CD34/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Hematopoietic Stem Cells
3.
J Clin Microbiol ; 59(10): e0052721, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34288726

ABSTRACT

Determinants of protective immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection require the development of well-standardized, reproducible antibody assays. This need has led to the emergence of a variety of neutralization assays. Head-to-head evaluation of different SARS-CoV-2 neutralization platforms could facilitate comparisons across studies and laboratories. Five neutralization assays were compared using 40 plasma samples from convalescent individuals with mild to moderate coronavirus disease 2019 (COVID-19): four cell-based systems using either live recombinant SARS-CoV-2 or pseudotyped viral particles created with lentivirus (LV) or vesicular stomatitis virus (VSV) packaging and one surrogate enzyme-linked immunosorbent assay (ELISA)-based test that measures inhibition of the spike protein receptor binding domain (RBD) binding its receptor human angiotensin converting enzyme 2 (hACE2). Vero cells, Vero E6 cells, HEK293T cells expressing hACE2, and TZM-bl cells expressing hACE2 and transmembrane serine protease 2 were tested. All cell-based assays showed 50% neutralizing dilution (ND50) geometric mean titers (GMTs) that were highly correlated (Pearson r = 0.81 to 0.89) and ranged within 3.4-fold. The live virus assay and LV pseudovirus assays with HEK293T/hACE2 cells showed very similar mean titers, 141 and 178, respectively. ND50 titers positively correlated with plasma IgG targeting SARS-CoV-2 spike protein and RBD (r = 0.63 to 0.89), but moderately correlated with nucleoprotein IgG (r = 0.46 to 0.73). ND80 GMTs mirrored ND50 data and showed similar correlation between assays and with IgG concentrations. The VSV pseudovirus assay and LV pseudovirus assay with HEK293T/hACE2 cells in low- and high-throughput versions were calibrated against the WHO SARS-CoV-2 IgG standard. High concordance between the outcomes of cell-based assays with live and pseudotyped virions enables valid cross-study comparison using these platforms.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Neutralizing , Antibodies, Viral , Chlorocebus aethiops , HEK293 Cells , Humans , Neutralization Tests , Spike Glycoprotein, Coronavirus/genetics , Vero Cells
4.
Nature ; 507(7491): 201-6, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24499818

ABSTRACT

Vaccines prevent infectious disease largely by inducing protective neutralizing antibodies against vulnerable epitopes. Several major pathogens have resisted traditional vaccine development, although vulnerable epitopes targeted by neutralizing antibodies have been identified for several such cases. Hence, new vaccine design methods to induce epitope-specific neutralizing antibodies are needed. Here we show, with a neutralization epitope from respiratory syncytial virus, that computational protein design can generate small, thermally and conformationally stable protein scaffolds that accurately mimic the viral epitope structure and induce potent neutralizing antibodies. These scaffolds represent promising leads for the research and development of a human respiratory syncytial virus vaccine needed to protect infants, young children and the elderly. More generally, the results provide proof of principle for epitope-focused and scaffold-based vaccine design, and encourage the evaluation and further development of these strategies for a variety of other vaccine targets, including antigenically highly variable pathogens such as human immunodeficiency virus and influenza.


Subject(s)
Drug Design , Epitopes/chemistry , Epitopes/immunology , Protein Stability , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Amino Acid Motifs , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Macaca mulatta/immunology , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Neutralization Tests , Protein Conformation , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/immunology
5.
Proc Natl Acad Sci U S A ; 112(33): 10342-7, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240330

ABSTRACT

Synthetic radionuclides, such as the transuranic actinides plutonium, americium, and curium, present severe health threats as contaminants, and understanding the scope of the biochemical interactions involved in actinide transport is instrumental in managing human contamination. Here we show that siderocalin, a mammalian siderophore-binding protein from the lipocalin family, specifically binds lanthanide and actinide complexes through molecular recognition of the ligands chelating the metal ions. Using crystallography, we structurally characterized the resulting siderocalin-transuranic actinide complexes, providing unprecedented insights into the biological coordination of heavy radioelements. In controlled in vitro assays, we found that intracellular plutonium uptake can occur through siderocalin-mediated endocytosis. We also demonstrated that siderocalin can act as a synergistic antenna to sensitize the luminescence of trivalent lanthanide and actinide ions in ternary protein-ligand complexes, dramatically increasing the brightness and efficiency of intramolecular energy transfer processes that give rise to metal luminescence. Our results identify siderocalin as a potential player in the biological trafficking of f elements, but through a secondary ligand-based metal sequestration mechanism. Beyond elucidating contamination pathways, this work is a starting point for the design of two-stage biomimetic platforms for photoluminescence, separation, and transport applications.


Subject(s)
Actinoid Series Elements/chemistry , Carrier Proteins/chemistry , Carrier Proteins/physiology , Proteins/chemistry , Actinoid Series Elements/pharmacokinetics , Chelating Agents/chemistry , Crystallography, X-Ray , Humans , Hydrogen-Ion Concentration , Ions , Kinetics , Lanthanoid Series Elements , Ligands , Lipocalin-2 , Luminescence , Metals/chemistry , Molecular Conformation , Nuclear Power Plants , Photochemistry , Protein Binding , Radioactive Hazard Release , Spectrophotometry , Static Electricity , X-Ray Diffraction
6.
PLoS Pathog ; 11(5): e1004857, 2015 May.
Article in English | MEDLINE | ID: mdl-25974133

ABSTRACT

Attachment proteins from the surface of eukaryotic cells, bacteria and viruses are critical receptors in cell adhesion or signaling and are primary targets for the development of vaccines and therapeutic antibodies. It is proposed that the ligand-binding pocket in receptor proteins can shift between inactive and active conformations with weak and strong ligand-binding capability, respectively. Here, using monoclonal antibodies against a vaccine target protein - fimbrial adhesin FimH of uropathogenic Escherichia coli, we demonstrate that unusually strong receptor inhibition can be achieved by antibody that binds within the binding pocket and displaces the ligand in a non-competitive way. The non-competitive antibody binds to a loop that interacts with the ligand in the active conformation of the pocket but is shifted away from ligand in the inactive conformation. We refer to this as a parasteric inhibition, where the inhibitor binds adjacent to the ligand in the binding pocket. We showed that the receptor-blocking mechanism of parasteric antibody differs from that of orthosteric inhibition, where the inhibitor replaces the ligand or allosteric inhibition where the inhibitor binds at a site distant from the ligand, and is very potent in blocking bacterial adhesion, dissolving surface-adherent biofilms and protecting mice from urinary bladder infection.


Subject(s)
Adhesins, Escherichia coli/metabolism , Antibodies, Monoclonal/immunology , Bacterial Adhesion , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Uropathogenic Escherichia coli/metabolism , Animals , Female , Male , Mice, Inbred C57BL , Models, Molecular
7.
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
8.
Inorg Chem ; 55(22): 11930-11936, 2016 Nov 21.
Article in English | MEDLINE | ID: mdl-27802058

ABSTRACT

Targeted α therapy holds tremendous potential as a cancer treatment: it offers the possibility of delivering a highly cytotoxic dose to targeted cells while minimizing damage to surrounding healthy tissue. The metallic α-generating radioisotopes 225Ac and 227Th are promising radionuclides for therapeutic use, provided adequate chelation and targeting. Here we demonstrate a new chelating platform composed of a multidentate high-affinity oxygen-donating ligand 3,4,3-LI(CAM) bound to the mammalian protein siderocalin. Respective stability constants log ß110 = 29.65 ± 0.65, 57.26 ± 0.20, and 47.71 ± 0.08, determined for the EuIII (a lanthanide surrogate for AcIII), ZrIV, and ThIV complexes of 3,4,3-LI(CAM) through spectrophotometric titrations, reveal this ligand to be one of the most powerful chelators for both trivalent and tetravalent metal ions at physiological pH. The resulting metal-ligand complexes are also recognized with extremely high affinity by the siderophore-binding protein siderocalin, with dissociation constants below 40 nM and tight electrostatic interactions, as evidenced by X-ray structures of the protein:ligand:metal adducts with ZrIV and ThIV. Finally, differences in biodistribution profiles between free and siderocalin-bound 238PuIV-3,4,3-LI(CAM) complexes confirm in vivo stability of the protein construct. The siderocalin:3,4,3-LI(CAM) assembly can therefore serve as a "lock" to consolidate binding to the therapeutic 225Ac and 227Th isotopes or to the positron emission tomography emitter 89Zr, independent of metal valence state.


Subject(s)
Chelating Agents/chemistry , Coordination Complexes/chemistry , Proteins/chemistry , Radiotherapy/methods , Thorium/chemistry , Zirconium/chemistry , Animals , Coordination Complexes/pharmacokinetics , Female , Ligands , Mice , Models, Chemical , Tissue Distribution
9.
J Immunol ; 192(4): 1587-96, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442437

ABSTRACT

Flagellin is a potent immunogen that activates the innate immune system via TLR5 and Naip5/6, and generates strong T and B cell responses. The adaptor protein MyD88 is critical for signaling by TLR5, as well as IL-1Rs and IL-18Rs, major downstream mediators of the Naip5/6 Nlrc4-inflammasome. In this study, we define roles of known flagellin receptors and MyD88 in Ab responses generated toward flagellin. We used mice genetically deficient in flagellin recognition pathways to characterize innate immune components that regulate isotype-specific Ab responses. Using purified flagellin from Salmonella, we dissected the contribution of innate flagellin recognition pathways to promote Ab responses toward flagellin and coadministered OVA in C57BL/6 mice. We demonstrate IgG2c responses toward flagellin were TLR5 and inflammasome dependent; IgG1 was the dominant isotype and partially TLR5 and inflammasome dependent. Our data indicate a substantial flagellin-specific IgG1 response was induced through a TLR5-, inflammasome-, and MyD88-independent pathway. IgA anti-FliC responses were TLR5 and MyD88 dependent and caspase-1 independent. Unlike C57BL/6 mice, flagellin-immunized A/J mice induced codominant IgG1 and IgG2a responses. Furthermore, MyD88-independent, flagellin-induced Ab responses were even more pronounced in A/J MyD88(-/-) mice, and IgA anti-FliC responses were suppressed by MyD88. Flagellin also worked as an adjuvant toward coadministered OVA, but it only promoted IgG1 anti-OVA responses. Our results demonstrate that a novel pathway for flagellin recognition contributes to Ab production. Characterization of this pathway will be useful for understanding immunity to flagellin and the rationale design of flagellin-based vaccines.


Subject(s)
Flagellin/immunology , Myeloid Differentiation Factor 88/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Toll-Like Receptor 5/metabolism , Animals , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , Flagellin/genetics , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Inflammasomes/metabolism , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Neuronal Apoptosis-Inhibitory Protein/deficiency , Neuronal Apoptosis-Inhibitory Protein/genetics , Ovalbumin , Receptors, IgG/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-18/metabolism , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics
10.
J Immunol ; 193(12): 6081-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25398327

ABSTRACT

Competition for iron is a critical component of successful bacterial infections, but the underlying in vivo mechanisms are poorly understood. We have previously demonstrated that lipocalin 2 (LCN2) is an innate immunity protein that binds to bacterial siderophores and starves them for iron, thus representing a novel host defense mechanism to infection. In the present study we show that LCN2 is secreted by the urinary tract mucosa and protects against urinary tract infection (UTI). We found that LCN2 was expressed in the bladder, ureters, and kidneys of mice subject to UTI. LCN2 was protective with higher bacterial numbers retrieved from bladders of Lcn2-deficient mice than from wild-type mice infected with the LCN2-sensitive Escherichia coli strain H9049. Uropathogenic E. coli mutants in siderophore receptors for salmochelin, aerobactin, or yersiniabactin displayed reduced fitness in wild-type mice, but not in mice deficient of LCN2, demonstrating that LCN2 imparts a selective pressure on bacterial growth in the bladder. In a human cohort of women with recurrent E. coli UTIs, urine LCN2 levels were associated with UTI episodes and with levels of bacteriuria. The number of siderophore systems was associated with increasing bacteriuria during cystitis. Our data demonstrate that LCN2 is secreted by the urinary tract mucosa in response to uropathogenic E. coli challenge and acts in innate immune defenses as a colonization barrier that pathogens must overcome to establish infection.


Subject(s)
Acute-Phase Proteins/genetics , Bacterial Infections/genetics , Lipocalins/genetics , Proto-Oncogene Proteins/genetics , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Acute-Phase Proteins/metabolism , Adolescent , Adult , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Bacterial Load , Cystitis/genetics , Cystitis/immunology , Cystitis/metabolism , Cystitis/microbiology , Disease Models, Animal , Escherichia coli , Female , Gene Expression , Humans , Iron/metabolism , Lipocalin-2 , Lipocalins/metabolism , Mice , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/pathology , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Proto-Oncogene Proteins/metabolism , Siderophores/metabolism , Urinary Bladder/pathology , Urinary Tract Infections/immunology , Urinary Tract Infections/pathology , Young Adult
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.
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
13.
PLoS Pathog ; 9(1): e1003106, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23300456

ABSTRACT

Vaccine candidates for HIV-1 so far have not been able to elicit broadly neutralizing antibodies (bNAbs) although they express the epitopes recognized by bNAbs to the HIV envelope glycoprotein (Env). To understand whether and how Env immunogens interact with the predicted germline versions of known bNAbs, we screened a large panel (N:56) of recombinant Envs (from clades A, B and C) for binding to the germline predecessors of the broadly neutralizing anti-CD4 binding site antibodies b12, NIH45-46 and 3BNC60. Although the mature antibodies reacted with diverse Envs, the corresponding germline antibodies did not display Env-reactivity. Experiments conducted with engineered chimeric antibodies combining the mature and germline heavy and light chains, respectively and vice-versa, revealed that both antibody chains are important for the known cross-reactivity of these antibodies. Our results also indicate that in order for b12 to display its broad cross-reactivity, multiple somatic mutations within its VH region are required. A consequence of the failure of the germline b12 to bind recombinant soluble Env is that Env-induced B-cell activation through the germline b12 BCR does not take place. Our study provides a new explanation for the difficulties in eliciting bNAbs with recombinant soluble Env immunogens. Our study also highlights the need for intense efforts to identify rare naturally occurring or engineered Envs that may engage the germline BCR versions of bNAbs.


Subject(s)
Antibodies, Neutralizing/immunology , CD4 Antigens/immunology , HIV Antibodies/immunology , HIV-1/genetics , HIV-1/immunology , AIDS Vaccines/immunology , Antibodies, Anti-Idiotypic/immunology , Antibody Affinity/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , Cell Line , Epitopes/immunology , HEK293 Cells , HIV Infections/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Lymphocyte Activation , Neutralization Tests , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
14.
FASEB J ; 28(4): 1555-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24361577

ABSTRACT

Lipocalins are small extracellular proteins mostly described as lipid carriers. The Drosophila lipocalin NLaz (neural Lazarillo) modulates the IIS pathway and regulates longevity, stress resistance, and behavior. Here, we test whether a native hydrophobic pocket structure is required for NLaz to perform its functions. We use a point mutation altering the binding pocket (NLaz(L130R)) and control mutations outside NLaz binding pocket. Tryptophan fluorescence titration reveals that NLaz(L130R) loses its ability to bind ergosterol and the pheromone 7(z)-tricosene but retains retinoic acid binding. Using site-directed transgenesis in Drosophila, we test the functionality of the ligand binding-altered lipocalin at the organism level. NLaz-dependent life span reduction, oxidative stress and starvation sensitivity, aging markers accumulation, and deficient courtship are rescued by overexpression of NLaz(WT), but not of NLaz(L130R). Transcriptional responses to aging and oxidative stress show a large set of age-responsive genes dependent on the integrity of NLaz binding pocket. Inhibition of IIS activity and modulation of oxidative stress and infection-responsive genes are binding pocket-dependent processes. Control of energy metabolites on starvation appears to be, however, insensitive to the modification of the NLaz binding pocket.


Subject(s)
Carrier Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Membrane Glycoproteins/genetics , Aging/drug effects , Aging/genetics , Alkenes/chemistry , Alkenes/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Ergosterol/chemistry , Ergosterol/metabolism , Herbicides/pharmacology , Hydrogen Peroxide/pharmacology , Ligands , Lipocalins/genetics , Lipocalins/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Oxidants/pharmacology , Paraquat/pharmacology , Point Mutation , Protein Binding/genetics , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/drug effects , Transcriptome/genetics , Tretinoin/chemistry , Tretinoin/metabolism
15.
Blood ; 120(26): 5153-62, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23018643

ABSTRACT

Immunotherapy with innate immune cells has recently evoked broad interest as a novel treatment option for cancer patients. γ9δ2T cells in particular are emerging as an innate cell population with high frequency and strong antitumor reactivity, which makes them and their receptors promising candidates for immune interventions. However, clinical trials have so far reported only limited tumor control by adoptively transferred γ9δ2T cells. As a potential explanation for this lack of efficacy, we found unexpectedly high variability in tumor recognition within the physiologic human γ9δ2T-cell repertoire, which is substantially regulated by the CDR3 domains of individual γ9δ2TCRs. In the present study, we demonstrate that the reported molecular requirements of CDR3 domains to interact with target cells shape the physiologic γ9δ2T-cell repertoire and, most likely, limit the protective and therapeutic antitumor efficacy of γ9δ2T cells. Based on these findings, we propose combinatorial-γδTCR-chain exchange as an efficient method for designing high-affinity γ9δ2TCRs that mediate improved antitumor responses when expressed in αßT cells both in vitro and in vivo in a humanized mouse model.


Subject(s)
Genes, T-Cell Receptor gamma/physiology , Immunoglobulin gamma-Chains/physiology , T-Cell Antigen Receptor Specificity , Adoptive Transfer , Animals , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/physiology , Genes, T-Cell Receptor gamma/genetics , Humans , Immunoglobulin gamma-Chains/chemistry , Immunoglobulin gamma-Chains/genetics , Immunotherapy, Adoptive/methods , K562 Cells , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Structure, Tertiary/physiology , T-Cell Antigen Receptor Specificity/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Proc Natl Acad Sci U S A ; 108(6): 2414-9, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262824

ABSTRACT

γδ T cells play important roles in bridging innate and adaptive immunity, but their recognition mechanisms remain poorly understood. Human γδ T cells of the V(δ)1 subset predominate in intestinal epithelia and respond to MICA and MICB (MHC class I chain-related, A and B; MIC) self-antigens, mediating responses to tumorigenesis or viral infection. The crystal structure of an MIC-reactive V(δ)1 γδ T-cell receptor (TCR) showed expected overall structural homology to antibodies, αß, and other γδ TCRs, but complementary determining region conformations and conservation of V(δ)1 use revealed an uncharacteristically flat potential binding surface. MIC, likewise, serves as a ligand for the activating immunoreceptor natural killer group 2, D (NKG2D), also expressed on γδ T cells. Although MIC recognition drives both the TCR-dependent stimulatory and NKG2D-dependent costimulatory signals necessary for activation, interaction analyses showed that MIC binding by the two receptors was mutually exclusive. Analysis of relative binding kinetics suggested sequential recognition, defining constraints for the temporal organization of γδ T-cell/target cell interfaces.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Receptors, Antigen, T-Cell, gamma-delta/chemistry , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Crystallography, X-Ray , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Innate/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms/immunology , Protein Structure, Quaternary , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology , Virus Diseases/immunology
17.
Structure ; 32(8): 1049-1054.e2, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38703776

ABSTRACT

Mesothelin (MSLN) is a cell-surface glycoprotein expressed at low levels on normal mesothelium but overexpressed in many cancers. Mesothelin has been implicated to play role/s in cell adhesion and multiple signaling pathways. Mucin-16/CA125 is an enormous cell-surface glycoprotein, also normally expressed on mesothelium and implicated in the progression and metastasis of several cancers, and directly binds mesothelin. However, the precise biological function/s of mesothelin and mucin-16/CA125 remain mysterious. We report protein engineering and recombinant production, qualitative and quantitative binding studies, and a crystal structure determination elucidating the molecular-level details governing recognition of mesothelin by mucin-16/CA125. The interface is small, consistent with the ∼micromolar binding constant and is free of glycan-mediated interactions. Sequence comparisons and modeling suggest that multiple mucin-16/CA125 modules can interact with mesothelin through comparable interactions, potentially generating a high degree of avidity at the cell surface to overcome the weak affinity, with implications for functioning and therapeutic interventions.


Subject(s)
CA-125 Antigen , GPI-Linked Proteins , Mesothelin , Models, Molecular , Protein Binding , Mesothelin/metabolism , Humans , CA-125 Antigen/metabolism , CA-125 Antigen/chemistry , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Crystallography, X-Ray , Binding Sites , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Amino Acid Sequence , Protein Engineering , Membrane Proteins
18.
Heliyon ; 10(7): e28583, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38586421

ABSTRACT

NKG2D and its ligands are critical regulators of protective immune responses controlling infections and cancer, defining a crucial immune signaling axis. Current therapeutic efforts targeting this axis almost exclusively aim at enhancing NKG2D-mediated effector functions. However, this axis can drive disease processes when dysregulated, in particular, driving stem-like cancer cell reprogramming and tumorigenesis through receptor/ligand self-stimulation on tumor cells. Despite complexities with its structure and biology, we developed multiple novel engineered proteins that functionally serve as axis-blocking NKG2D "decoys" and report biochemical, structural, in vitro, and in vivo evaluation of their functionality.

19.
Nat Commun ; 15(1): 5741, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009580

ABSTRACT

Targeted alpha therapy (TAT) pairs the specificity of antigen targeting with the lethality of alpha particles to eradicate cancerous cells. Actinium-225 [225Ac; t1/2 = 9.920(3) days] is an alpha-emitting radioisotope driving the next generation of TAT radiopharmaceuticals. Despite promising clinical results, a fundamental understanding of Ac coordination chemistry lags behind the rest of the Periodic Table due to its limited availability, lack of stable isotopes, and inadequate systems poised to probe the chemical behavior of this radionuclide. In this work, we demonstrate a platform that combines an 8-coordinate synthetic ligand and a mammalian protein to characterize the solution and solid-state behavior of the longest-lived Ac isotope, 227Ac [t1/2 = 21.772(3) years]. We expect these results to direct renewed efforts for 225Ac-TAT development, aid in understanding Ac coordination behavior relative to other +3 lanthanides and actinides, and more broadly inform this element's position on the Periodic Table.


Subject(s)
Actinium , Chelating Agents , Actinium/chemistry , Chelating Agents/chemistry , Crystallization , Radiopharmaceuticals/chemistry , Humans , Ligands
20.
J Biol Chem ; 287(17): 13524-31, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22389496

ABSTRACT

Bacteria use tight-binding, ferric-specific chelators called siderophores to acquire iron from the environment and from the host during infection; animals use proteins such as transferrin and ferritin to transport and store iron. Recently, candidate compounds that could serve endogenously as mammalian siderophore equivalents have been identified and characterized through associations with siderocalin, the only mammalian siderophore-binding protein currently known. Siderocalin, an antibacterial protein, acts by sequestering iron away from infecting bacteria as siderophore complexes. Candidate endogenous siderophores include compounds that only effectively transport iron as ternary complexes with siderocalin, explaining pleiotropic activities in normal cellular processes and specific disease states.


Subject(s)
Iron/chemistry , Lipocalins/chemistry , Siderophores/chemistry , Animals , Anti-Bacterial Agents/chemistry , Apoptosis , Biological Transport , Carrier Proteins/chemistry , Catechols/chemistry , Chelating Agents/pharmacology , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Kinetics , Lipocalin-2 , Models, Biological , Models, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL