Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(31): e2203410119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35878026

RESUMEN

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.


Asunto(s)
Fosfatasa Ácida , Antígenos de Neoplasias , Receptores de Antígenos de Linfocitos T , Fosfatasa Ácida/metabolismo , Antígenos de Neoplasias/metabolismo , Epítopos , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Leucocitos Mononucleares , Neoplasias/inmunología , Péptidos , Receptores de Antígenos de Linfocitos T/metabolismo
2.
J Clin Microbiol ; 59(10): e0052721, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34288726

RESUMEN

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.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Chlorocebus aethiops , Células HEK293 , Humanos , Pruebas de Neutralización , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero
3.
Nature ; 507(7491): 201-6, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24499818

RESUMEN

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.


Asunto(s)
Diseño de Fármacos , Epítopos/química , Epítopos/inmunología , Estabilidad Proteica , Vacunas contra Virus Sincitial Respiratorio/química , Vacunas contra Virus Sincitial Respiratorio/inmunología , Secuencias de Aminoácidos , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/análisis , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Cristalografía por Rayos X , Ensayo de Inmunoadsorción Enzimática , Macaca mulatta/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Pruebas de Neutralización , Conformación Proteica , Virus Sincitiales Respiratorios/química , Virus Sincitiales Respiratorios/inmunología
4.
Proc Natl Acad Sci U S A ; 112(33): 10342-7, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26240330

RESUMEN

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.


Asunto(s)
Elementos de Series Actinoides/química , Proteínas Portadoras/química , Proteínas Portadoras/fisiología , Proteínas/química , Elementos de Series Actinoides/farmacocinética , Quelantes/química , Cristalografía por Rayos X , Humanos , Concentración de Iones de Hidrógeno , Iones , Cinética , Elementos de la Serie de los Lantanoides , Ligandos , Lipocalina 2 , Luminiscencia , Metales/química , Conformación Molecular , Plantas de Energía Nuclear , Fotoquímica , Unión Proteica , Liberación de Radiactividad Peligrosa , Espectrofotometría , Electricidad Estática , Difracción de Rayos X
5.
PLoS Pathog ; 11(5): e1004857, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25974133

RESUMEN

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.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Anticuerpos Monoclonales/inmunología , Adhesión Bacteriana , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Escherichia coli Uropatógena/metabolismo , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Modelos Moleculares
6.
PLoS Pathog ; 10(9): e1004403, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25254371

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Anticuerpos ampliamente neutralizantes , Cristalografía por Rayos X , Anticuerpos Anti-VIH/química , Infecciones por VIH/virología , Humanos , Datos de Secuencia Molecular , Pruebas de Neutralización , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
7.
Inorg Chem ; 55(22): 11930-11936, 2016 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-27802058

RESUMEN

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.


Asunto(s)
Quelantes/química , Complejos de Coordinación/química , Proteínas/química , Radioterapia/métodos , Torio/química , Circonio/química , Animales , Complejos de Coordinación/farmacocinética , Femenino , Ligandos , Ratones , Modelos Químicos , Distribución Tisular
8.
J Immunol ; 192(4): 1587-96, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24442437

RESUMEN

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.


Asunto(s)
Flagelina/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Receptor Toll-Like 5/metabolismo , Animales , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Células Cultivadas , Flagelina/genética , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Proteína Inhibidora de la Apoptosis Neuronal/deficiencia , Proteína Inhibidora de la Apoptosis Neuronal/genética , Ovalbúmina , Receptores de IgG/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-18/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética , Receptor Toll-Like 5/deficiencia , Receptor Toll-Like 5/genética
9.
J Immunol ; 193(12): 6081-9, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25398327

RESUMEN

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.


Asunto(s)
Proteínas de Fase Aguda/genética , Infecciones Bacterianas/genética , Lipocalinas/genética , Proteínas Proto-Oncogénicas/genética , Vejiga Urinaria/metabolismo , Vejiga Urinaria/microbiología , Infecciones Urinarias/genética , Infecciones Urinarias/microbiología , Proteínas de Fase Aguda/metabolismo , Adolescente , Adulto , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Carga Bacteriana , Cistitis/genética , Cistitis/inmunología , Cistitis/metabolismo , Cistitis/microbiología , Modelos Animales de Enfermedad , Escherichia coli , Femenino , Expresión Génica , Humanos , Hierro/metabolismo , Lipocalina 2 , Lipocalinas/metabolismo , Ratones , Persona de Mediana Edad , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Proteínas Proto-Oncogénicas/metabolismo , Sideróforos/metabolismo , Vejiga Urinaria/patología , Infecciones Urinarias/inmunología , Infecciones Urinarias/patología , Adulto Joven
10.
Immunol Rev ; 250(1): 239-57, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23046134

RESUMEN

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.


Asunto(s)
Antígenos Virales/química , Antígenos de Histocompatibilidad Clase I/química , Células Asesinas Naturales/inmunología , Receptores de Células Asesinas Naturales/química , Virosis/inmunología , Virus/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Citocinas/inmunología , Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Evasión Inmune , Inmunidad Innata , Células Asesinas Naturales/virología , Modelos Moleculares , Unión Proteica , Receptores de Células Asesinas Naturales/genética , Receptores de Células Asesinas Naturales/inmunología , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T , Virosis/virología
11.
PLoS Pathog ; 9(9): e1003639, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086134

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Regiones Determinantes de Complementariedad/inmunología , Epítopos/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Receptores de Inositol 1,4,5-Trifosfato/inmunología , Animales , Anticuerpos Monoclonales/genética , Autoanticuerpos/genética , Autoantígenos/genética , Anticuerpos ampliamente neutralizantes , Cardiolipinas/genética , Cardiolipinas/inmunología , Regiones Determinantes de Complementariedad/genética , Epítopos/genética , Anticuerpos Anti-VIH/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Ratones Transgénicos , Proteoma/genética , Proteoma/inmunología
12.
PLoS Pathog ; 9(1): e1003106, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23300456

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos CD4/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/genética , VIH-1/inmunología , Vacunas contra el SIDA/inmunología , Anticuerpos Antiidiotipos/inmunología , Afinidad de Anticuerpos/inmunología , Antígenos Virales/inmunología , Linfocitos B/inmunología , Línea Celular , Epítopos/inmunología , Células HEK293 , Infecciones por VIH/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/inmunología , Activación de Linfocitos , Pruebas de Neutralización , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
13.
FASEB J ; 28(4): 1555-67, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24361577

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Glicoproteínas de Membrana/genética , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Alquenos/química , Alquenos/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ergosterol/química , Ergosterol/metabolismo , Herbicidas/farmacología , Peróxido de Hidrógeno/farmacología , Ligandos , Lipocalinas/genética , Lipocalinas/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Oxidantes/farmacología , Paraquat/farmacología , Mutación Puntual , Unión Proteica/genética , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Tretinoina/química , Tretinoina/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(6): 2414-9, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262824

RESUMEN

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.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/química , Receptores de Antígenos de Linfocitos T gamma-delta/química , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/inmunología , Cristalografía por Rayos X , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad Innata/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Neoplasias/inmunología , Estructura Cuaternaria de Proteína , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/química , Linfocitos T/inmunología , Virosis/inmunología
15.
Structure ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38703776

RESUMEN

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.

16.
Heliyon ; 10(7): e28583, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38586421

RESUMEN

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.

17.
J Biol Chem ; 287(17): 13524-31, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22389496

RESUMEN

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.


Asunto(s)
Hierro/química , Lipocalinas/química , Sideróforos/química , Animales , Antibacterianos/química , Apoptosis , Transporte Biológico , Proteínas Portadoras/química , Catecoles/química , Quelantes/farmacología , Humanos , Concentración de Iones de Hidrógeno , Inmunidad Innata , Cinética , Lipocalina 2 , Modelos Biológicos , Modelos Químicos
18.
J Exp Med ; 204(2): 393-403, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17283206

RESUMEN

The molecular basis for Toll-like receptor (TLR) recognition of microbial ligands is unknown. We demonstrate that mouse and human TLR5 discriminate between different flagellins, and we use this difference to map the flagellin recognition site on TLR5 to 228 amino acids of the extracellular domain. Through molecular modeling of the TLR5 ectodomain, we identify two conserved surface-exposed regions. Mutagenesis studies demonstrate that naturally occurring amino acid variation in TLR5 residue 268 is responsible for human and mouse discrimination between flagellin molecules. Mutations within one conserved surface identify residues D295 and D367 as important for flagellin recognition. These studies localize flagellin recognition to a conserved surface on the modeled TLR5 structure, providing detailed analysis of the interaction of a TLR with its ligand. These findings suggest that ligand binding at the beta sheets results in TLR activation and provide a new framework for understanding TLR-agonist interactions.


Asunto(s)
Bacterias/química , Flagelina/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína , Receptor Toll-Like 5/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Células CHO , Cricetinae , Cricetulus , Flagelina/aislamiento & purificación , Variación Genética , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Mutagénesis , FN-kappa B/metabolismo , Unión Proteica , Alineación de Secuencia , Especificidad de la Especie , Receptor Toll-Like 5/metabolismo
19.
Nature ; 447(7143): 482-6, 2007 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-17495932

RESUMEN

Tumour-associated ligands of the activating NKG2D (natural killer group 2, member D; also called KLRK1) receptor-which are induced by genotoxic or cellular stress-trigger activation of natural killer cells and co-stimulation of effector T cells, and may thus promote resistance to cancer. However, many progressing tumours in humans counter this anti-tumour activity by shedding the soluble major histocompatibility complex class-I-related ligand MICA, which induces internalization and degradation of NKG2D and stimulates population expansions of normally rare NKG2D+CD4+ T cells with negative regulatory functions. Here we show that on the surface of tumour cells, MICA associates with endoplasmic reticulum protein 5 (ERp5; also called PDIA6 or P5), which, similar to protein disulphide isomerase, usually assists in the folding of nascent proteins inside cells. Pharmacological inhibition of thioreductase activity and ERp5 gene silencing revealed that cell-surface ERp5 function is required for MICA shedding. ERp5 and membrane-anchored MICA form transitory mixed disulphide complexes from which soluble MICA is released after proteolytic cleavage near the cell membrane. Reduction of the seemingly inaccessible disulphide bond in the membrane-proximal alpha3 domain of MICA must involve a large conformational change that enables proteolytic cleavage. These results uncover a molecular mechanism whereby domain-specific deconstruction regulates MICA protein shedding, thereby promoting tumour immune evasion, and identify surface ERp5 as a strategic target for therapeutic intervention.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Receptores Inmunológicos/metabolismo , Línea Celular Tumoral , Disulfuros/química , Disulfuros/metabolismo , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Antígenos de Histocompatibilidad Clase I/química , Humanos , Ligandos , Chaperonas Moleculares/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias/enzimología , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Células Asesinas Naturales , Especificidad por Sustrato
20.
Nucleic Acids Res ; 39(21): e143, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21911364

RESUMEN

A key challenge for the academic and biopharmaceutical communities is the rapid and scalable production of recombinant proteins for supporting downstream applications ranging from therapeutic trials to structural genomics efforts. Here, we describe a novel system for the production of recombinant mammalian proteins, including immune receptors, cytokines and antibodies, in a human cell line culture system, often requiring <3 weeks to achieve stable, high-level expression: Daedalus. The inclusion of minimized ubiquitous chromatin opening elements in the transduction vectors is key for preventing genomic silencing and maintaining the stability of decigram levels of expression. This system can bypass the tedious and time-consuming steps of conventional protein production methods by employing the secretion pathway of serum-free adapted human suspension cell lines, such as 293 Freestyle. Using optimized lentiviral vectors, yields of 20-100 mg/l of correctly folded and post-translationally modified, endotoxin-free protein of up to ~70 kDa in size, can be achieved in conventional, small-scale (100 ml) culture. At these yields, most proteins can be purified using a single size-exclusion chromatography step, immediately appropriate for use in structural, biophysical or therapeutic applications.


Asunto(s)
Lentivirus/genética , Proteínas Recombinantes/biosíntesis , Proteínas de Fase Aguda/biosíntesis , Proteínas de Fase Aguda/química , Proteínas de Fase Aguda/genética , Animales , Línea Celular , Cristalografía , Citocinas/genética , Citocinas/metabolismo , Técnicas Genéticas , Vectores Genéticos , Humanos , Lipocalina 2 , Lipocalinas/biosíntesis , Lipocalinas/química , Lipocalinas/genética , Ratones , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA