Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 50(1): 142-145, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31580480

RESUMEN

The semi-public T-cell response towards the gluten epitope DQ2.5-glia-α2 uses a prototypic TCR encoded by the germline segments TRAV26-1 and TRBV7-2. Through mutagenesis experiments, we show that a TRAV26-1encoded recognition motif contacts the MHC ß-chain and the TCR CDR3ß loop underpinning this conserved T-cell response restricted to the prototypic TCRs.


Asunto(s)
Enfermedad Celíaca/inmunología , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/inmunología , Secuencias de Aminoácidos/inmunología , Epítopos de Linfocito T/química , Humanos , Receptores de Antígenos de Linfocitos T alfa-beta/química
2.
Scand J Immunol ; 93(2): e13017, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33351196

RESUMEN

The neonatal Fc receptor (FcRn) was first recognized for its role in transfer of maternal IgG to the foetus or newborn, providing passive immunity early in life. However, it has become clear that the receptor is versatile, widely expressed and plays an indispensable role in both immunological and non-immunological processes throughout life. The receptor rescues immunoglobulin G (IgG) and albumin from intracellular degradation and shuttles the ligands across polarized cell barriers, in all cases via a pH-dependent binding-and-release mechanism. These processes secure distribution and high levels of both IgG and albumin throughout the body. At mucosal sites, FcRn transports IgG across polarized epithelial cells where it retrieves IgG in complex with luminal antigens that is delivered to tissue-localized immune cells. In dendritic cells (DCs), FcRn orchestrates processing of IgG-opsonized immune complexes (ICs) in concert with classical Fcγ receptors, which results in antigen presentation and cross-presentation of antigenic peptides on MHC class II and I to CD4+ and CD8+ T cells, respectively. Hence, FcRn regulates transport of the ligands within and across different types of cells, but also processing of IgG-ICs by immune cells. As such, the receptor is involved in immune surveillance and protection against infections. In this brief review, we highlight how FcRn expressed by hematopoietic and non-hematopoietic cells contributes to immune regulation at mucosal barriers-biology that can be utilized in development of biologics and subunit vaccines for non-invasive delivery.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Membrana Mucosa/inmunología , Receptores Fc/inmunología , Animales , Presentación de Antígeno/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Inmunoglobulina G/inmunología , Factores Inmunológicos/inmunología
3.
Gastroenterology ; 156(5): 1428-1439.e10, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30593798

RESUMEN

BACKGROUND & AIMS: Development of celiac disease is believed to involve the transglutaminase-dependent response of CD4+ T cells toward deamidated gluten peptides in the intestinal mucosa of individuals with specific HLA-DQ haplotypes. We investigated the antigen presentation process during this mucosal immune response. METHODS: We generated monoclonal antibodies (mAbs) specific for the peptide-MHC (pMHC) complex of HLA-DQ2.5 and the immunodominant gluten epitope DQ2.5-glia-α1a using phage display. We used these mAbs to assess gluten peptide presentation and phenotypes of presenting cells by flow cytometry and enzyme-linked immune absorbent spot (ELISPOT) in freshly prepared single-cell suspensions from intestinal biopsies from 40 patients with celiac disease (35 untreated and 5 on a gluten-free diet) as well as 18 subjects with confirmed noninflamed gut mucosa (controls, 12 presumed healthy, 5 undergoing pancreatoduodenectomy, and 1 with potential celiac disease). RESULTS: Using the mAbs, we detected MHC complexes on cells from intestinal biopsies from patients with celiac disease who consume gluten, but not from patients on gluten-free diets. We found B cells and plasma cells to be the most abundant cells that present DQ2.5-glia-α1a in the inflamed mucosa. We identified a subset of plasma cells that expresses B-cell receptors (BCR) specific for gluten peptides or the autoantigen transglutaminase 2 (TG2). Expression of MHC class II (MHCII) was not restricted to these specific plasma cells in patients with celiac disease but was observed in an average 30% of gut plasma cells from patients and controls. CONCLUSIONS: A population of plasma cells from intestinal biopsies of patients with celiac disease express MHCII; this is the most abundant cell type presenting the immunodominant gluten peptide DQ2.5-glia-α1a in the tissues from these patients. These results indicate that plasma cells in the gut can function as antigen-presenting cells and might promote and maintain intestinal inflammation in patients with celiac disease or other inflammatory disorders.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Enfermedad Celíaca/inmunología , Duodeno/inmunología , Glútenes/inmunología , Antígenos HLA-DQ/inmunología , Inmunidad Mucosa , Epítopos Inmunodominantes , Mucosa Intestinal/inmunología , Fragmentos de Péptidos/inmunología , Células Plasmáticas/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Estudios de Casos y Controles , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/metabolismo , Línea Celular , Dieta Sin Gluten , Duodeno/metabolismo , Duodeno/patología , Proteínas de Unión al GTP/inmunología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Fenotipo , Células Plasmáticas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/inmunología
4.
Proc Natl Acad Sci U S A ; 114(14): E2862-E2871, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28330995

RESUMEN

The neonatal crystallizable fragment receptor (FcRn) is responsible for maintaining the long half-life and high levels of the two most abundant circulating proteins, albumin and IgG. In the latter case, the protective mechanism derives from FcRn binding to IgG in the weakly acidic environment contained within endosomes of hematopoietic and parenchymal cells, whereupon IgG is diverted from degradation in lysosomes and is recycled. The cellular location and mechanism by which FcRn protects albumin are partially understood. Here we demonstrate that mice with global or liver-specific FcRn deletion exhibit hypoalbuminemia, albumin loss into the bile, and increased albumin levels in the hepatocyte. In vitro models with polarized cells illustrate that FcRn mediates basal recycling and bidirectional transcytosis of albumin and uniquely determines the physiologic release of newly synthesized albumin into the basal milieu. These properties allow hepatic FcRn to mediate albumin delivery and maintenance in the circulation, but they also enhance sensitivity to the albumin-bound hepatotoxin, acetaminophen (APAP). As such, global or liver-specific deletion of FcRn results in resistance to APAP-induced liver injury through increased albumin loss into the bile and increased intracellular albumin scavenging of reactive oxygen species. Further, protection from injury is achieved by pharmacologic blockade of FcRn-albumin interactions with monoclonal antibodies or peptide mimetics, which cause hypoalbuminemia, biliary loss of albumin, and increased intracellular accumulation of albumin in the hepatocyte. Together, these studies demonstrate that the main function of hepatic FcRn is to direct albumin into the circulation, thereby also increasing hepatocyte sensitivity to toxicity.


Asunto(s)
Albúminas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Acetaminofén/efectos adversos , Acetaminofén/metabolismo , Animales , Bilis/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Perros , Femenino , Hepatocitos/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Homeostasis , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores Fc/genética , Albúmina Sérica Humana/genética , Albúmina Sérica Humana/metabolismo , Transcitosis/genética
5.
Immunol Rev ; 268(1): 328-39, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497531

RESUMEN

Antibodies are key molecules in the fight against infections. Although previously thought to mediate protection solely in the extracellular environment, recent research has revealed that antibody-mediated protection extends to the cytosolic compartment of cells. This postentry viral defense mechanism requires binding of the antibody to a cytosolic Fc receptor named tripartite motif containing 21 (TRIM21). In contrast to other Fc receptors, TRIM21 shows remarkably broad isotype specificity as it does not only bind IgG but also IgM and IgA. When viral pathogens coated with these antibody isotypes enter the cytosol, TRIM21 is rapidly recruited and efficient neutralization occurs before the virus has had the time to replicate. In addition, inflammatory signaling is induced. As such, TRIM21 acts as a cytosolic sensor that engages antibodies that have failed to protect against infection in the extracellular environment. Here, we summarize our current understanding of how TRIM21 orchestrates humoral immunity in the cytosolic environment.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Isotipos de Inmunoglobulinas/inmunología , Isotipos de Inmunoglobulinas/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Citosol/metabolismo , Activación Enzimática , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Isotipos de Inmunoglobulinas/química , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Ribonucleoproteínas/química , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Immunol ; 196(8): 3452-3459, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26962230

RESUMEN

Ab-coated viruses can be detected in the cytosol by the FcR tripartite motif-containing 21 (TRIM21), which rapidly recruits the proteasomal machinery and triggers induction of immune signaling. As such, TRIM21 plays a key role in intracellular protection by targeting invading viruses for destruction and alerting the immune system. A hallmark of immunity is elicitation of a balanced response that is proportionate to the threat, to avoid unnecessary inflammation. In this article, we show how Ab affinity modulates TRIM21 immune function. We constructed a humanized monoclonal IgG1 against human adenovirus type 5 (AdV5) and a panel of Fc-engineered variants with a wide range of affinities for TRIM21. We found that IgG1-coated viral particles were neutralized via TRIM21, even when affinity was reduced by as much as 100-fold. In contrast, induction of NF-κB signaling was more sensitive to reduced affinity between TRIM21 and the Ab variants. Thus, TRIM21 mediates neutralization under suboptimal conditions, whereas induction of immune signaling is balanced according to the functional affinity for the incoming immune stimuli. Our findings have implications for engineering of antiviral IgG therapeutics with tailored effector functions.


Asunto(s)
Adenovirus Humanos/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/inmunología , Inmunoglobulina G/inmunología , Ribonucleoproteínas/inmunología , Animales , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/inmunología , Pruebas de Neutralización , Ribonucleoproteínas/genética , Transducción de Señal/inmunología , Resonancia por Plasmón de Superficie
7.
J Immunol ; 194(11): 5497-508, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25904551

RESUMEN

Engineering of the constant Fc part of monoclonal human IgG1 (hIgG1) Abs is an approach to improve effector functions and clinical efficacy of next-generation IgG1-based therapeutics. A main focus in such development is tailoring of in vivo half-life and transport properties by engineering the pH-dependent interaction between IgG and the neonatal Fc receptor (FcRn), as FcRn is the main homeostatic regulator of hIgG1 half-life. However, whether such engineering affects binding to other Fc-binding molecules, such as the classical FcγRs and complement factor C1q, has not been studied in detail. These effector molecules bind to IgG1 in the lower hinge-CH2 region, structurally distant from the binding site for FcRn at the CH2-CH3 elbow region. However, alterations of the structural composition of the Fc may have long-distance effects. Indeed, in this study we show that Fc engineering of hIgG1 for altered binding to FcRn also influences binding to both the classical FcγRs and complement factor C1q, which ultimately results in alterations of cellular mechanisms such as Ab-dependent cell-mediated cytotoxicity, Ab-dependent cellular phagocytosis, and Ab-dependent complement-mediated cell lysis. Thus, engineering of the FcRn-IgG1 interaction may greatly influence effector functions, which has implications for the therapeutic efficacy and use of Fc-engineered hIgG1 variants.


Asunto(s)
Anticuerpos Monoclonales/genética , Complemento C1q/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/genética , Receptores Fc/inmunología , Receptores de IgG/inmunología , Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/genética , Afinidad de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular , Células HEK293 , Exones de la Región Bisagra/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunoglobulina G/inmunología , Nitrohidroxiyodofenilacetato/inmunología , Fagocitosis/inmunología , Ingeniería de Proteínas , Receptores Fc/genética , Receptores de IgG/genética , Resonancia por Plasmón de Superficie
8.
J Biol Chem ; 289(50): 34583-94, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25344603

RESUMEN

Albumin is an abundant blood protein that acts as a transporter of a plethora of small molecules like fatty acids, hormones, toxins, and drugs. In addition, it has an unusual long serum half-life in humans of nearly 3 weeks, which is attributed to its interaction with the neonatal Fc receptor (FcRn). FcRn protects albumin from intracellular degradation via a pH-dependent cellular recycling mechanism. To understand how FcRn impacts the role of albumin as a distributor, it is of importance to unravel the structural mechanism that determines pH-dependent binding. Here, we show that although the C-terminal domain III (DIII) of human serum albumin (HSA) contains the principal binding site, the N-terminal domain I (DI) is important for optimal FcRn binding. Specifically, structural inspection of human FcRn (hFcRn) in complex with HSA revealed that two exposed loops of DI were in proximity with the receptor. To investigate to what extent these contacts affected hFcRn binding, we targeted selected amino acid residues of the loops by mutagenesis. Screening by in vitro interaction assays revealed that several of the engineered HSA variants showed decreased binding to hFcRn, which was also the case for two missense variants with mutations within these loops. In addition, four of the variants showed improved binding. Our findings demonstrate that both DI and DIII are required for optimal binding to FcRn, which has implications for our understanding of the FcRn-albumin relationship and how albumin acts as a distributor. Such knowledge may inspire development of novel HSA-based diagnostics and therapeutics.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Albúmina Sérica/química , Albúmina Sérica/metabolismo , Sustitución de Aminoácidos , Unión Competitiva , Humanos , Concentración de Iones de Hidrógeno , Cinética , Modelos Moleculares , Unión Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Albúmina Sérica/genética
9.
J Biol Chem ; 289(19): 13492-502, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24652290

RESUMEN

A major challenge for the therapeutic use of many peptides and proteins is their short circulatory half-life. Albumin has an extended serum half-life of 3 weeks because of its size and FcRn-mediated recycling that prevents intracellular degradation, properties shared with IgG antibodies. Engineering the strictly pH-dependent IgG-FcRn interaction is known to extend IgG half-life. However, this principle has not been extensively explored for albumin. We have engineered human albumin by introducing single point mutations in the C-terminal end that generated a panel of variants with greatly improved affinities for FcRn. One variant (K573P) with 12-fold improved affinity showed extended serum half-life in normal mice, mice transgenic for human FcRn, and cynomolgus monkeys. Importantly, favorable binding to FcRn was maintained when a single-chain fragment variable antibody was genetically fused to either the N- or the C-terminal end. The engineered albumin variants may be attractive for improving the serum half-life of biopharmaceuticals.


Asunto(s)
Albúminas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Albúminas/genética , Albúminas/farmacología , Sustitución de Aminoácidos , Animales , Femenino , Semivida , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/farmacología , Humanos , Macaca fascicularis , Ratones , Mutación Missense , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
10.
J Biol Chem ; 289(24): 17228-39, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24764301

RESUMEN

Albumin is the most abundant protein in blood and plays a pivotal role as a multitransporter of a wide range of molecules such as fatty acids, metabolites, hormones, and toxins. In addition, it binds a variety of drugs. Its role as distributor is supported by its extraordinary serum half-life of 3 weeks. This is related to its size and binding to the cellular receptor FcRn, which rescues albumin from intracellular degradation. Furthermore, the long half-life has fostered a great and increasing interest in utilization of albumin as a carrier of protein therapeutics and chemical drugs. However, to fully understand how FcRn acts as a regulator of albumin homeostasis and to take advantage of the FcRn-albumin interaction in drug design, the interaction interface needs to be dissected. Here, we used a panel of monoclonal antibodies directed towards human FcRn in combination with site-directed mutagenesis and structural modeling to unmask the binding sites for albumin blocking antibodies and albumin on the receptor, which revealed that the interaction is not only strictly pH-dependent, but predominantly hydrophobic in nature. Specifically, we provide mechanistic evidence for a crucial role of a cluster of conserved tryptophan residues that expose a pH-sensitive loop of FcRn, and identify structural differences in proximity to these hot spot residues that explain divergent cross-species binding properties of FcRn. Our findings expand our knowledge of how FcRn is controlling albumin homeostasis at a molecular level, which will guide design and engineering of novel albumin variants with altered transport properties.


Asunto(s)
Albúminas/metabolismo , Antígenos de Histocompatibilidad Clase I/química , Receptores Fc/química , Secuencia de Aminoácidos , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo
11.
Blood ; 122(7): 1174-81, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23843496

RESUMEN

The neonatal Fc receptor (FcRn) directs the transfer of maternal immunoglobulin G (IgG) antibodies across the placenta and thus provides the fetus and newborn with passive protective humoral immunity. Pathogenic maternal IgG antibodies will also be delivered via the placenta and can cause alloimmunity, which may be lethal. A novel strategy to control pathogenic antibodies would be administration of a nondestructive IgG antibody blocking antigen binding while retaining binding to FcRn. We report on 2 human IgG3 antibodies with a hinge deletion and a C131S point mutation (IgG3ΔHinge) that eliminate complement activation and binding to all classical Fcγ receptors (FcγRs) and to C1q while binding to FcRn is retained. Additionally, 1 of the antibodies has a single point mutation in the Fc (R435H) at the binding site for FcRn (IgG3ΔHinge:R435H). We compared transplacental transport with wild-type IgG1 and IgG3, and found transport across trophoblast-derived BeWo cells and ex vivo placenta perfusions with hierarchies as follows: IgG3ΔHinge:R435H>wild-type IgG1≥IgG3ΔHinge and IgG3ΔHinge:R435H=wild-type IgG1=wild-type IgG3>>>IgG3ΔHinge, respectively. Collectively, IgG3ΔHinge:R435H was transported efficiently from the maternal to the fetal placental compartment. Thus, IgG3ΔHinge:R435H may be a good candidate for transplacental delivery of a nondestructive antibody to the fetus to combat pathogenic antibodies.


Asunto(s)
Anticuerpos/inmunología , Feto/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/inmunología , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Receptores Fc/inmunología , Proteínas Recombinantes/inmunología , Anticuerpos/metabolismo , Sitios de Unión , Transporte Biológico , Coriocarcinoma/inmunología , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Recién Nacido , Placenta/metabolismo , Embarazo , Receptores Fc/metabolismo , Proteínas Recombinantes/metabolismo , Neoplasias Uterinas/inmunología , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología
12.
J Immunol ; 191(9): 4769-77, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24062486

RESUMEN

CD14 is a key recognition molecule of innate immune responses, interacting with several TLRs. TLR signaling cross-talks extensively with the complement system, and combined CD14 and complement inhibition has been proved effective in attenuating inflammatory responses. Pig models of human diseases have emerged as valuable tools to study therapeutic intervention, but suitable neutralizing Abs are rare. Undesired Fc-mediated functions, such as platelet activation and IL-8 release induced by the porcine CD14-specific clone Mil2, limit further studies. Therefore, an inert human IgG2/IgG4 hybrid C region was chosen for an rMil2. As revealed in ex vivo and in vivo pig experiments, rMil2 inhibited the CD14-mediated proinflammatory cytokine response similar to the original clone, but lacked the undesired Fc-effects, and inflammation was attenuated further by simultaneous complement inhibition. Moreover, rMil2 bound porcine FcRn, a regulator of t1/2 and biodistribution. Thus, rMil2, particularly combined with complement inhibitors, should be well suited for in vivo studies using porcine models of diseases, such as sepsis and ischemia-reperfusion injury. Similarly, the recombinant anti-human CD14 IgG2/4 Ab, r18D11, was generated with greatly reduced Fc-mediated effects and preserved inhibitory function ex vivo. Such Abs might be drug candidates for the treatment of innate immunity-mediated human diseases.


Asunto(s)
Inmunoglobulina G/uso terapéutico , Inmunoterapia , Inflamación/inmunología , Inflamación/terapia , Receptores de Lipopolisacáridos/inmunología , Animales , Anticuerpos , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Antiidiotipos/uso terapéutico , Antígenos de Diferenciación/inmunología , Línea Celular , Activación de Complemento/inmunología , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Receptores de IgG/inmunología , Sus scrofa
13.
Biochemistry ; 53(13): 2091-100, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24625295

RESUMEN

In eukaryotes, different chromatin states facilitate or repress gene expression and restrict the activity of transposable elements. Post-translational modifications (PTMs) of amino acid residues on the N-terminal tails of histones are suggested to define such states. The histone lysine methyltransferase (HKMTase) SU(VAR)3-9 RELATED4 (SUVR4) of Arabidopsis thaliana functions as a repressor of transposon activity. Binding of ubiquitin by the WIYLD domain facilitates the addition of two methyl groups to monomethylated lysine 9 of histone H3. By using nuclear magnetic resonance (NMR) spectroscopy, we identified SUVR4 WIYLD (S4WIYLD) as a domain with a four-helix bundle structure, in contrast to three-helix bundles of other ubiquitin binding domains. NMR titration analyses showed that residues of helix α1 (Q38, L39, and D40) and helix α4 (N68, T70, A71, V73, D74, I76, S78, and E82) of S4WIYLD and residues between the first and second ß-strands (T9 and G10) and on ß-strands 3 (R42, G47, K48, and Q49) and 4 (H68, R72, and L73) undergo significant chemical shift changes when the two proteins interact. A model of the complex, generated using HADDOCK, suggests that the N-terminal and C-terminal parts of S4WIYLD constitute a surface that interacts with charged residues close to the hydrophobic patch of ubiquitin. The WIYLD domains of the closely related SUVR1 and SUVR2 Arabidopsis proteins also bind ubiquitin, indicating that this is a general feature of this domain. The question of whether SUVR proteins act as both readers of monoubiquitinated H2B and writers of histone PTMs is discussed.


Asunto(s)
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Ensayo de Inmunoadsorción Enzimática , Histonas/química , Histonas/metabolismo , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Secundaria de Proteína , Alineación de Secuencia , Ubiquitina/química
14.
J Biol Chem ; 288(33): 24277-85, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818524

RESUMEN

Albumin has a serum half-life of 3 weeks in humans. This has been utilized to extend the serum persistence of biopharmaceuticals that are fused to albumin. In light of the fact that the neonatal Fc receptor (FcRn) is a key regulator of albumin homeostasis, it is crucial to address how fusion of therapeutics to albumin impacts binding to FcRn. Here, we report on a detailed molecular investigation on how genetic fusion of a short peptide or an single-chain variable fragment (scFv) fragment to human serum albumin (HSA) influences pH-dependent binding to FcRn from mouse, rat, monkey, and human. We have found that fusion to the N- or C-terminal end of HSA only slightly reduces receptor binding, where the most noticeable effect is seen after fusion to the C-terminal end. Furthermore, in contrast to the observed strong binding to human and monkey FcRn, HSA and all HSA fusions bound very poorly to mouse and rat versions of the receptor. Thus, we demonstrate that conventional rodents are limited as preclinical models for analysis of serum half-life of HSA-based biopharmaceuticals. This finding is explained by cross-species differences mainly found within domain III (DIII) of albumin. Our data demonstrate that although fusion, particularly to the C-terminal end, may slightly reduce the affinity for FcRn, HSA is versatile as a carrier of biopharmaceuticals.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Albúmina Sérica/metabolismo , Albúmina Sérica/uso terapéutico , Anticuerpos de Cadena Única/metabolismo , Animales , Unión Competitiva , Semivida , Humanos , Cinética , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Especificidad de la Especie
15.
Nucleic Acids Res ; 40(16): e120, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22539265

RESUMEN

Phage display has been instrumental in discovery of novel binding peptides and folded domains for the past two decades. We recently reported a novel pIX phagemid display system that is characterized by a strong preference for phagemid packaging combined with low display levels, two key features that support highly efficient affinity selection. However, high diversity in selected repertoires are intimately coupled to high display levels during initial selection rounds. To incorporate this additional feature into the pIX display system, we have developed a novel helper phage termed DeltaPhage that allows for high-valence display on pIX. This was obtained by inserting two amber mutations close to the pIX start codon, but after the pVII translational stop, conditionally inactivating the helper phage encoded pIX. Until now, the general notion has been that display on pIX is dependent on wild-type complementation, making high-valence display unachievable. However, we found that DeltaPhage does facilitate high-valence pIX display when used with a non-suppressor host. Here, we report a side-by-side comparison with pIII display, and we find that this novel helper phage complements existing pIX phagemid display systems to allow both low and high-valence display, making pIX display a complete and efficient alternative to existing pIII phagemid display systems.


Asunto(s)
Bacteriófagos/genética , Virus Helper/genética , Biblioteca de Péptidos , Animales , Antígenos/análisis , Bacteriófagos/fisiología , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Virus Helper/fisiología , Ratones , Receptores de Antígenos de Linfocitos T/análisis
16.
Proc Natl Acad Sci U S A ; 108(24): 9927-32, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21628593

RESUMEN

Cross-presentation of IgG-containing immune complexes (ICs) is an important means by which dendritic cells (DCs) activate CD8(+) T cells, yet it proceeds by an incompletely understood mechanism. We show that monocyte-derived CD8(-)CD11b(+) DCs require the neonatal Fc receptor for IgG (FcRn) to conduct cross-presentation of IgG ICs. Consequently, in the absence of FcRn, Fcγ receptor (FcγR)-mediated antigen uptake fails to initiate cross-presentation. FcRn is shown to regulate the intracellular sorting of IgG ICs to the proper destination for such cross-presentation to occur. We demonstrate that FcRn traps antigen and protects it from degradation within an acidic loading compartment in association with the rapid recruitment of key components of the phagosome-to-cytosol cross-presentation machinery. This unique mechanism thus enables cross-presentation to evolve from an atypically acidic loading compartment. FcRn-driven cross-presentation is further shown to control cross-priming of CD8(+) T-cell responses in vivo such that during chronic inflammation, FcRn deficiency results in inadequate induction of CD8(+) T cells. These studies thus demonstrate that cross-presentation in CD8(-)CD11b(+) DCs requires a two-step mechanism that involves FcγR-mediated internalization and FcRn-directed intracellular sorting of IgG ICs. Given the centrality of FcRn in controlling cross-presentation, these studies lay the foundation for a unique means to therapeutically manipulate CD8(+) T-cell responses.


Asunto(s)
Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/inmunología , Receptores Fc/inmunología , Animales , Antígenos/inmunología , Western Blotting , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Citosol/inmunología , Citosol/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextran , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Concentración de Iones de Hidrógeno , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , NADPH Oxidasa 2 , NADPH Oxidasas/inmunología , NADPH Oxidasas/metabolismo , Fagosomas/inmunología , Fagosomas/metabolismo , Unión Proteica , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , ATPasas de Translocación de Protón Vacuolares/inmunología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Proteínas de Unión al GTP rab/inmunología , Proteínas de Unión al GTP rab/metabolismo , Proteínas rab27 de Unión a GTP
17.
Nat Commun ; 15(1): 2007, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453922

RESUMEN

Monoclonal IgG antibodies constitute the fastest growing class of therapeutics. Thus, there is an intense interest to design more potent antibody formats, where long plasma half-life is a commercially competitive differentiator affecting dosing, frequency of administration and thereby potentially patient compliance. Here, we report on an Fc-engineered variant with three amino acid substitutions Q311R/M428E/N434W (REW), that enhances plasma half-life and mucosal distribution, as well as allows for needle-free delivery across respiratory epithelial barriers in human FcRn transgenic mice. In addition, the Fc-engineered variant improves on-target complement-mediated killing of cancer cells as well as both gram-positive and gram-negative bacteria. Hence, this versatile Fc technology should be broadly applicable in antibody design aiming for long-acting prophylactic or therapeutic interventions.


Asunto(s)
Neoplasias , Receptores Fc , Ratones , Animales , Humanos , Inmunoglobulina G , Semivida , Antibacterianos/uso terapéutico , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Ratones Transgénicos , Anticuerpos Monoclonales , Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias/terapia , Neoplasias/tratamiento farmacológico
18.
J Biol Chem ; 287(27): 22927-37, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22570488

RESUMEN

Serum half-life of IgG is controlled by the neonatal Fc receptor (FcRn) that interacts with the IgG Fc region and may be increased or decreased as a function of altered FcRn binding. Preclinical evaluations of modified IgGs are frequently carried out in mice, but such IgGs may bind differently to mouse and human FcRn (mFcRn and hFcRn). Here, we report a detailed characterization of a matched set of mouse-human chimeric T84.66 scFv-Fc variants with specificity for the tumor carcinoembryonic antigen and mutations in the FcRn-binding site. Binding to soluble mFcRn and hFcRn was measured using in vitro assays, and the results were compared with blood clearance in vivo in normal (mFcRn bearing) and hFcRn transgenic mice. All variants bound better to mFcRn than to hFcRn. The loss of affinity varied among the mutants, however, and also the hierarchy of binding differed depending on the receptor. The mutations had no major impact on binding to the classical Fcγ receptors. Importantly, the trend of blood clearance in both strains of mice correlated with the hierarchy of binding obtained using soluble FcRn. Consequently, in vitro interaction analysis of engineered IgGs regarding their cross-species FcRn binding ability provides information for prediction of in vivo pharmacokinetics.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/inmunología , Ingeniería de Proteínas/métodos , Receptores Fc/inmunología , Receptores de IgG/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Especificidad de Anticuerpos , Reacciones Cruzadas/inmunología , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/sangre , Radioisótopos de Yodo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Anticuerpos de Cadena Única/farmacocinética , Especificidad de la Especie
19.
J Immunol ; 187(6): 3064-71, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21849672

RESUMEN

Posttranslational modification of Ag is implicated in several autoimmune diseases. In celiac disease, a cereal gluten-induced enteropathy with several autoimmune features, T cell recognition of the gluten Ag is heavily dependent on the posttranslational conversion of Gln to Glu residues. Evidence suggests that the enhanced recognition of deamidated gluten peptides results from improved peptide binding to the MHC and TCR interaction with the peptide-MHC complex. In this study, we report that there is a biased usage of TCR Vß6.7 chain among TCRs reactive to the immunodominant DQ2-α-II gliadin epitope. We isolated Vß6.7 and DQ2-αII tetramer-positive CD4(+) T cells from peripheral blood of gluten-challenged celiac patients and sequenced the TCRs of a large number of single T cells. TCR sequence analysis revealed in vivo clonal expansion, convergent recombination, semipublic response, and the notable conservation of a non-germline-encoded Arg residue in the CDR3ß loop. Functional testing of a prototype DQ2-α-II-reactive TCR by analysis of TCR transfectants and soluble single-chain TCRs indicate that the deamidated residue in the DQ2-α-II peptide poses constraints on the TCR structure in which the conserved Arg residue is a critical element. The findings have implications for understanding T cell responses to posttranslationally modified Ags.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad Celíaca/genética , Glútenes/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Receptores de Antígenos de Linfocitos T/genética , Secuencia de Bases , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Gliadina/inmunología , Glútenes/genética , Glútenes/metabolismo , Humanos , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Resonancia por Plasmón de Superficie
20.
Methods ; 58(1): 40-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22819858

RESUMEN

Phage display technology has evolved to become an extremely versatile and powerful platform for protein engineering. The robustness of the phage particle, its ease of handling and its ability to tolerate a range of different capsid fusions are key features that explain the dominance of phage display in combinatorial engineering. Implementation of new technology is likely to ensure the continuation of its success, but has also revealed important short comings inherent to current phage display systems. This is in particular related to the biology of the two most popular display capsids, namely pIII and pVIII. Recent findings using two alternative capsids, pVII and pIX, located to the phage tip opposite that of pIII, suggest how they may be exploited to alleviate or circumvent many of these short comings. This review addresses important aspects of the current phage display standard and then discusses the use of pVII and pIX. These may both complement current systems and be used as alternative scaffolds for display and selection to further improve phage display as the ultimate combinatorial engineering platform.


Asunto(s)
Proteínas de la Cápside/biosíntesis , Técnicas de Visualización de Superficie Celular , Proteínas Recombinantes de Fusión/biosíntesis , Bacteriófagos , Proteínas de la Cápside/genética , Evolución Molecular Dirigida , Escherichia coli , Humanos , Biblioteca de Péptidos , Ingeniería de Proteínas , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA