Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
EMBO J ; 40(5): e106228, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258165

RESUMEN

Nucleoprotein (N) is an immunodominant antigen in many enveloped virus infections. While the diagnostic value of anti-N antibodies is clear, their role in immunity is not. This is because while they are non-neutralising, they somehow clear infection by coronavirus, influenza and LCMV in vivo. Here, we show that anti-N immune protection is mediated by the cytosolic Fc receptor and E3 ubiquitin ligase TRIM21. Exploiting LCMV as a model system, we demonstrate that TRIM21 uses anti-N antibodies to target N for cytosolic degradation and generate cytotoxic T cells (CTLs) against N peptide. These CTLs rapidly eliminate N-peptide-displaying cells and drive efficient viral clearance. These results reveal a new mechanism of immune synergy between antibodies and T cells and highlights N as an important vaccine target.


Asunto(s)
Anticuerpos Antivirales/inmunología , Inmunidad Celular , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas de la Nucleocápside/inmunología , Ribonucleoproteínas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Noqueados , Proteínas de la Nucleocápside/genética , Ribonucleoproteínas/genética , Vacunas Virales/genética , Vacunas Virales/inmunología
2.
EMBO J ; 38(21): e101365, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31468569

RESUMEN

Inflammasomes are potent innate immune signalling complexes that couple cytokine release with pro-inflammatory cell death. However, pathogens have evolved strategies to evade this cell autonomous system. Here, we show how antibodies combine with innate sensors in primary human macrophages to detect viral infection and activate the inflammasome. Our data demonstrate that antibody opsonisation of virions can activate macrophages in multiple ways. In the first, antibody binding of adenovirus causes lysosomal damage, activating NLRP3 to drive inflammasome formation and IL-1ß release. Importantly, this mechanism enhances virion capture but not infection and is accompanied by cell death, denying the opportunity for viral replication. Unexpectedly, we also find that antibody-coated viruses, which successfully escape into the cytosol, trigger a second system of inflammasome activation. These viruses are intercepted by the cytosolic antibody receptor TRIM21 and the DNA sensor cGAS. Together, these sensors stimulate both NLRP3 inflammasome formation and NFκB activation, driving dose-dependent IL-1ß and TNF secretion, without inducing cell death. Our data highlight the importance of cooperativity between multiple sensing networks to expose viruses to the inflammasome pathway, which is particularly important for how our innate immune system responds to infection in the presence of pre-existing immunity.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Anticuerpos Antivirales/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nucleotidiltransferasas/metabolismo , Ribonucleoproteínas/metabolismo , Replicación Viral/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/virología , Animales , Células Cultivadas , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nucleotidiltransferasas/genética , Ribonucleoproteínas/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
PLoS Pathog ; 16(8): e1008732, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32750093

RESUMEN

Rotavirus is a major cause of gastroenteritis in children, with infection typically inducing high levels of protective antibodies. Antibodies targeting the middle capsid protein VP6 are particularly abundant, and as VP6 is only exposed inside cells, neutralisation must be post-entry. However, while a system of poly immune globulin receptor (pIgR) transcytosis has been proposed for anti-VP6 IgAs, the mechanism by which VP6-specific IgG mediates protection remains less clear. We have developed an intracellular neutralisation assay to examine how antibodies neutralise rotavirus inside cells, enabling comparison between IgG and IgA isotypes. Unexpectedly we found that neutralisation by VP6-specific IgG was much more efficient than by VP6-specific IgA. This observation was highly dependent on the activity of the cytosolic antibody receptor TRIM21 and was confirmed using an in vivo model of murine rotavirus infection. Furthermore, mice deficient in only IgG and not other antibody isotypes had a serious deficit in intracellular antibody-mediated protection. The finding that VP6-specific IgG protect mice against rotavirus infection has important implications for rotavirus vaccination. Current assays determine protection in humans predominantly by measuring rotavirus-specific IgA titres. Measurements of VP6-specific IgG may add to existing mechanistic correlates of protection.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Proteínas de la Cápside/inmunología , Inmunoglobulina G/inmunología , Infecciones por Rotavirus/inmunología , Rotavirus/fisiología , Animales , Antígenos Virales/genética , Proteínas de la Cápside/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Rotavirus/genética , Infecciones por Rotavirus/virología , Especificidad de la Especie
4.
Proc Natl Acad Sci U S A ; 115(41): 10440-10445, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30209217

RESUMEN

Adenovirus has enormous potential as a gene-therapy vector, but preexisting immunity limits its widespread application. What is responsible for this immune block is unclear because antibodies potently inhibit transgene expression without impeding gene transfer into target cells. Here we show that antibody prevention of adenoviral gene delivery in vivo is mediated by the cytosolic antibody receptor TRIM21. Genetic KO of TRIM21 or a single-antibody point mutation is sufficient to restore transgene expression to near-naïve immune levels. TRIM21 is also responsible for blocking cytotoxic T cell induction by vaccine vectors, preventing a protective response against subsequent influenza infection and an engrafted tumor. Furthermore, adenoviral preexisting immunity can lead to an augmented immune response upon i.v. administration of the vector. Transcriptomic analysis of vector-transduced tissue reveals that TRIM21 is responsible for the specific up-regulation of hundreds of immune genes, the majority of which are components of the intrinsic or innate response. Together, these data define a major mechanism underlying the preimmune block to adenovirus gene therapy and demonstrate that TRIM21 efficiently blocks gene delivery in vivo while simultaneously inducing a rapid program of immune transcription.


Asunto(s)
Infecciones por Adenoviridae/terapia , Adenoviridae/inmunología , Anticuerpos/inmunología , Fibrosarcoma/terapia , Terapia Genética , Ribonucleoproteínas/fisiología , Vacunación , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Animales , Fibrosarcoma/genética , Fibrosarcoma/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transgenes , Células Tumorales Cultivadas
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Nat Commun ; 14(1): 3109, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37253747

RESUMEN

Antibody-based blocking of vascular endothelial growth factor (VEGF) reduces choroidal neovascularization (CNV) and retinal edema, rescuing vision in patients with neovascular age-related macular degeneration (nAMD). However, poor response and resistance to anti-VEGF treatment occurs. We report that targeting the Notch ligand Jagged1 by a monoclonal antibody reduces neovascular lesion size, number of activated phagocytes and inflammatory markers and vascular leakage in an experimental CNV mouse model. Additionally, we demonstrate that Jagged1 is expressed in mouse and human eyes, and that Jagged1 expression is independent of VEGF signaling in human endothelial cells. When anti-Jagged1 was combined with anti-VEGF in mice, the decrease in lesion size exceeded that of either antibody alone. The therapeutic effect was solely dependent on blocking, as engineering antibodies to abolish effector functions did not impair the therapeutic effect. Targeting of Jagged1 alone or in combination with anti-VEGF may thus be an attractive strategy to attenuate CNV-bearing diseases.


Asunto(s)
Neovascularización Coroidal , Factor A de Crecimiento Endotelial Vascular , Humanos , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales/metabolismo , Neovascularización Coroidal/patología , Anticuerpos Bloqueadores/uso terapéutico , Transducción de Señal/fisiología , Modelos Animales de Enfermedad , Inhibidores de la Angiogénesis/uso terapéutico
13.
Int J Retina Vitreous ; 9(1): 65, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37936232

RESUMEN

BACKGROUND: Intravitreal injection (IVI) of antibody biologics is a key treatment approach in ophthalmology. Pharmaceutical compounding and storage of prefilled syringes for IVI must take place without impairing the structure and function of the biologics. This study investigated the effect of withdrawing and storing the therapeutic antibody faricimab (Vabysmo, Roche, Basel, Switzerland) in the Zero Residual silicone oil-free, 0.2-mL syringe (SJJ Solutions, The Hague, the Netherlands). METHODS: To assess the effect of syringe withdrawal on faricimab, we compared samples from syringes prepared at day 0 with samples taken directly from faricimab vials. To assess the effect of syringe storage on faricimab, we kept prefilled syringes in the dark at 4 oC for 7, 14, or 37 days and compared samples from these syringes with day 0. We measured protein concentration (with spectrophotometry), stability and integrity (with sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), size-exclusion chromatography (SEC), and melting temperature (Tm)), as well as binding of faricimab to its cognate antigens: vascular endothelial growth factor A (VEGF-A) and angiopoietin-2 (Ang-2) (with enzyme-linked immunosorbent assay (ELISA)). RESULTS: Faricimab migrated in line with its expected molecular mass under both reducing and non-reducing conditions for all time points when analyzed with SDS-PAGE, without any sign of degradation products or aggregation. The SEC elution profiles were identical for all time points. There were slight variations in Tm for different time points compared to day 0 but without consistent relationship with storage time. ELISA did not detect differences in VEGF-A or Ang-2 binding between time points, and faricimab did not bind the neonatal Fc receptor. CONCLUSIONS: Withdrawal and storage of faricimab in syringes for up to day 37 did not impair the structure and bi-specific binding properties of the therapeutic antibody.

14.
Front Immunol ; 14: 1178817, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37346044

RESUMEN

Upregulation of surface expressed sialoglycans on tumor cells is one of the mechanisms which promote tumor growth and progression. Specifically, the interactions of sialic acids with sialic acid-binding immunoglobulin-like lectins (Siglecs) on lymphoid or myeloid cells transmit inhibitory signals and lead to suppression of anti-tumor responses. Here, we show that neutrophils express among others Siglec-9, and that EGFR and HER2 positive breast tumor cells express ligands for Siglec-9. Treatment of tumor cells with neuraminidases or a sialyl transferase inhibitor significantly reduced binding of a soluble recombinant Siglec-9-Fc fusion protein, while EGFR and HER2 expression remained unchanged. Importantly, the cytotoxic activity of neutrophils driven by therapeutic EGFR or HER2 antibodies in vitro was increased by blocking the sialic acid/Siglec interaction, either by reducing tumor cell sialylation or by a Siglec-9 blocking antibody containing an effector silenced Fc domain. In vivo a short-term xenograft mouse model confirmed the improved therapeutic efficacy of EGFR antibodies against sialic acid depleted, by a sialyltransferase inhibitor, tumor cells compared to untreated cells. Our studies demonstrate that sialic acid/Siglec interactions between tumor cells and myeloid cells can impair antibody dependent tumor cell killing, and that Siglec-9 on polymorphonuclear cells (PMN) is critically involved. Considering that PMN are often a highly abundant cell population in the tumor microenvironment, Siglec-9 constitutes a promising target for myeloid checkpoint blockade to improve antibody-based tumor immunotherapy.


Asunto(s)
Ácido N-Acetilneuramínico , Neoplasias , Humanos , Ratones , Animales , Ácido N-Acetilneuramínico/metabolismo , Neutrófilos/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Anticuerpos , Ácidos Siálicos/metabolismo , Receptores ErbB , Microambiente Tumoral
15.
Science ; 379(6639): 1336-1341, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36996217

RESUMEN

Aggregates of the protein tau are proposed to drive pathogenesis in neurodegenerative diseases. Tau can be targeted by using passively transferred antibodies (Abs), but the mechanisms of Ab protection are incompletely understood. In this work, we used a variety of cell and animal model systems and showed that the cytosolic Ab receptor and E3 ligase TRIM21 (T21) could play a role in Ab protection against tau pathology. Tau-Ab complexes were internalized to the cytosol of neurons, which enabled T21 engagement and protection against seeded aggregation. Ab-mediated protection against tau pathology was lost in mice that lacked T21. Thus, the cytosolic compartment provides a site of immunotherapeutic protection, which may help in the design of Ab-based therapies in neurodegenerative disease.


Asunto(s)
Anticuerpos Monoclonales , Inmunización Pasiva , Ribonucleoproteínas , Tauopatías , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas tau , Animales , Ratones , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Citosol/metabolismo , Modelos Animales de Enfermedad , Receptores Fc , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Proteínas tau/inmunología , Tauopatías/terapia , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
Sci Immunol ; 7(70): eabj1640, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35486676

RESUMEN

Humans have four IgG antibody subclasses that selectively or differentially engage immune effector molecules to protect against infections. Although IgG1 has been studied in detail and is the subclass of most approved antibody therapeutics, increasing evidence indicates that IgG3 is associated with enhanced protection against pathogens. Here, we report that IgG3 has superior capacity to mediate intracellular antiviral immunity compared with the other subclasses due to its uniquely extended and flexible hinge region, which facilitates improved recruitment of the cytosolic Fc receptor TRIM21, independently of Fc binding affinity. TRIM21 may also synergize with complement C1/C4-mediated lysosomal degradation via capsid inactivation. We demonstrate that this process is potentiated by IgG3 in a hinge-dependent manner. Our findings reveal differences in how the four IgG subclasses mediate intracellular immunity, knowledge that may guide IgG subclass selection and engineering of antiviral antibodies for prophylaxis and therapy.


Asunto(s)
Antivirales , Inmunoglobulina G , Anticuerpos Antivirales , Proteínas del Sistema Complemento , Humanos , Receptores Fc
17.
Commun Biol ; 5(1): 832, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35982144

RESUMEN

Antibody-based therapeutics (ABTs) are used to treat a range of diseases. Most ABTs are either full-length IgG1 antibodies or fusions between for instance antigen (Ag)-binding receptor domains and the IgG1 Fc fragment. Interestingly, their plasma half-life varies considerably, which may relate to how they engage the neonatal Fc receptor (FcRn). As such, there is a need for an in-depth understanding of how different features of ABTs affect FcRn-binding and transport behavior. Here, we report on how FcRn-engagement of the IgG1 Fc fragment compare to clinically relevant IgGs and receptor domain Fc fusions, binding to VEGF or TNF-α. The results reveal FcRn-dependent intracellular accumulation of the Fc, which is in line with shorter plasma half-life than that of full-length IgG1 in human FcRn-expressing mice. Receptor domain fusion to the Fc increases its half-life, but not to the extent of IgG1. This is mirrored by a reduced cellular recycling capacity of the Fc-fusions. In addition, binding of cognate Ag to ABTs show that complexes of similar size undergo cellular transport at different rates, which could be explained by the biophysical properties of each ABT. Thus, the study provides knowledge that should guide tailoring of ABTs regarding optimal cellular sorting and plasma half-life.


Asunto(s)
Inmunoglobulina G , Receptores Fc , Animales , Semivida , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Ratones , Receptores Fc/genética
18.
Cells ; 11(6)2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35326398

RESUMEN

Pemphigus vulgaris is an autoimmune blistering disease of the epidermis, caused by autoantibodies against desmosomal proteins, mainly desmogleins 1 and 3, which induce an impairment of desmosomal adhesion and blister formation. Recent findings have shown that inhibition of immunoglobulin G binding on the neonatal Fc receptor, FcRn, results in reduced autoantibody recycling and shortens their half-life, providing a valid treatment option for PV. We have here analyzed the role of FcRn in human keratinocytes treated with antibodies isolated from pemphigus vulgaris patient or with recombinant anti-desmoglein-3 antibodies that induce pathogenic changes in desmosomes, such as loss of monolayer integrity, aberrant desmoglein-3 localization and degradation of desmoglein-3. We show that blocking IgG binding on FcRn by efgartigimod, a recombinant Fc fragment undergoing clinical studies for pemphigus, stabilizes the keratinocyte monolayer, whereas the loss of desmoglein-3 is not prevented by efgartigimod. Our data show that FcRn may play a direct role in the pathogenesis of pemphigus at the level of the autoantibody target cells, the epidermal keratinocytes. Our data suggest that in keratinocytes, FcRn may have functions different from its known function in IgG recycling. Therefore, stabilization of keratinocyte adhesion by FcRn blocking entities may provide a novel treatment paradigm for pemphigus.


Asunto(s)
Enfermedades Autoinmunes , Pénfigo , Autoanticuerpos , Enfermedades Autoinmunes/metabolismo , Desmogleína 3/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Recién Nacido , Queratinocitos/metabolismo , Pénfigo/tratamiento farmacológico , Pénfigo/metabolismo
20.
Immunohorizons ; 6(1): 90-103, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35074850

RESUMEN

Maternal alloantibodies toward paternally inherited Ags on fetal platelets can cause thrombocytopenia and bleeding complications in the fetus or neonate, referred to as fetal and neonatal alloimmune thrombocytopenia (FNAIT). This is most commonly caused by Abs against the human platelet Ag (HPA)-1a in Caucasians, and a prophylactic regimen to reduce the risk for alloimmunization to women at risk would be beneficial. We therefore aimed to examine the prophylactic potential of a fully human anti-HPA-1a IgG1 (mAb 26.4) with modified Fc region or altered N-glycan structures. The mAb 26.4 wild-type (WT) variants all showed efficient platelet clearance capacity and ability to mediate phagocytosis independent of their N-glycan structure, compared with an effector silent variant (26.4.AAAG), although the modified N-glycan variants showed differential binding to FcγRs measured in vitro. In an in vivo model, female mice were transfused with platelets from transgenic mice harboring an engineered integrin ß3 containing the HPA-1a epitope. When these preimmunized mice were bred with transgenic males, Abs against the introduced epitope induced thrombocytopenia in the offspring, mimicking FNAIT. Prophylactic administration of the mAb 26.4.WT, and to some extent the mAb 26.4.AAAG, prior to platelet transfusion resulted in reduced alloimmunization in challenged mice and normal platelet counts in neonates. The notion that the effector silent variant hampered alloimmunization demonstrates that rapid platelet clearance, as seen with mAb 26.4.WT, is not the sole mechanism in action. Our data thus successfully demonstrate efficient Ab-mediated immunosuppression and prevention of FNAIT by anti-HPA-1a monoclonal variants, providing support for potential use in humans.


Asunto(s)
Antígenos de Plaqueta Humana/inmunología , Integrina beta3/inmunología , Isoanticuerpos/sangre , Trombocitopenia Neonatal Aloinmune/inmunología , Trombocitopenia Neonatal Aloinmune/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Isoformas de Proteínas , Células THP-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA