Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 38(21): e101365, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31468569

ABSTRACT

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.


Subject(s)
Adenoviridae Infections/immunology , Antibodies, Viral/immunology , Inflammasomes/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotidyltransferases/metabolism , Ribonucleoproteins/metabolism , Virus Replication/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Animals , Cells, Cultured , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nucleotidyltransferases/genetics , Ribonucleoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
J Neurol Neurosurg Psychiatry ; 94(1): 19-22, 2023 01.
Article in English | MEDLINE | ID: mdl-34670844

ABSTRACT

INTRODUCTION: The effect of disease-modifying therapies (DMT) on vaccine responses is largely unknown. Understanding the development of protective immunity is of paramount importance to fight the COVID-19 pandemic. OBJECTIVE: To characterise humoral immunity after mRNA-COVID-19 vaccination of people with multiple sclerosis (pwMS). METHODS: All pwMS in Norway fully vaccinated against SARS-CoV-2 were invited to a national screening study. Humoral immunity was assessed by measuring anti-SARS-CoV-2 SPIKE RBD IgG response 3-12 weeks after full vaccination, and compared with healthy subjects. RESULTS: 528 pwMS and 627 healthy subjects were included. Reduced humoral immunity (anti-SARS-CoV-2 IgG <70 arbitrary units) was present in 82% and 80% of all pwMS treated with fingolimod and rituximab, respectively, while patients treated with other DMT showed similar rates as healthy subjects and untreated pwMS. We found a significant correlation between time since the last rituximab dose and the development of humoral immunity. Revaccination in two seronegative patients induced a weak antibody response. CONCLUSIONS: Patients treated with fingolimod or rituximab should be informed about the risk of reduced humoral immunity and vaccinations should be timed carefully in rituximab patients. Our results identify the need for studies regarding the durability of vaccine responses, the role of cellular immunity and revaccinations.


Subject(s)
COVID-19 , Multiple Sclerosis , Humans , Immunization, Secondary , Immunity, Humoral , Rituximab/therapeutic use , Multiple Sclerosis/drug therapy , Fingolimod Hydrochloride/therapeutic use , COVID-19 Vaccines/therapeutic use , Pandemics , SARS-CoV-2 , COVID-19/prevention & control , Vaccination , Antibodies, Viral , Immunoglobulin G , RNA, Messenger
3.
Proc Natl Acad Sci U S A ; 117(40): 25018-25025, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32943538

ABSTRACT

Respiratory failure in the acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is hypothesized to be driven by an overreacting innate immune response, where the complement system is a key player. In this prospective cohort study of 39 hospitalized coronavirus disease COVID-19 patients, we describe systemic complement activation and its association with development of respiratory failure. Clinical data and biological samples were obtained at admission, days 3 to 5, and days 7 to 10. Respiratory failure was defined as PO2/FiO2 ratio of ≤40 kPa. Complement activation products covering the classical/lectin (C4d), alternative (C3bBbP) and common pathway (C3bc, C5a, and sC5b-9), the lectin pathway recognition molecule MBL, and antibody serology were analyzed by enzyme-immunoassays; viral load by PCR. Controls comprised healthy blood donors. Consistently increased systemic complement activation was observed in the majority of COVID-19 patients during hospital stay. At admission, sC5b-9 and C4d were significantly higher in patients with than without respiratory failure (P = 0.008 and P = 0.034). Logistic regression showed increasing odds of respiratory failure with sC5b-9 (odds ratio 31.9, 95% CI 1.4 to 746, P = 0.03) and need for oxygen therapy with C4d (11.7, 1.1 to 130, P = 0.045). Admission sC5b-9 and C4d correlated significantly to ferritin (r = 0.64, P < 0.001; r = 0.69, P < 0.001). C4d, sC5b-9, and C5a correlated with antiviral antibodies, but not with viral load. Systemic complement activation is associated with respiratory failure in COVID-19 patients and provides a rationale for investigating complement inhibitors in future clinical trials.


Subject(s)
Betacoronavirus/immunology , Complement Activation , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Respiratory Insufficiency/immunology , Aged , Biomarkers/blood , COVID-19 , Case-Control Studies , Coronavirus Infections/blood , Coronavirus Infections/complications , Female , Host-Pathogen Interactions/immunology , Humans , Male , Mannose-Binding Lectin/blood , Middle Aged , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/complications , Respiratory Insufficiency/virology , SARS-CoV-2 , Viral Load
4.
PLoS Pathog ; 16(8): e1008732, 2020 08.
Article in English | MEDLINE | ID: mdl-32750093

ABSTRACT

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.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Capsid Proteins/immunology , Immunoglobulin G/immunology , Rotavirus Infections/immunology , Rotavirus/physiology , Animals , Antigens, Viral/genetics , Capsid Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rotavirus/genetics , Rotavirus Infections/virology , Species Specificity
5.
Bioconjug Chem ; 33(8): 1494-1504, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35875886

ABSTRACT

Recombinantly produced biotherapeutics hold promise for improving the current standard of care for snakebite envenoming over conventional serotherapy. Nanobodies have performed well in the clinic, and in the context of antivenom, they have shown the ability to neutralize long α-neurotoxins in vivo. Here, we showcase a protein engineering approach to increase the valence and hydrodynamic size of neutralizing nanobodies raised against a long α-neurotoxin (α-cobratoxin) from the venom of the monocled cobraNaja kaouthia. Based on the p53 tetramerization domain, a panel of anti-α-cobratoxin nanobody-p53 fusion proteins, termed Quads, were produced with different valences, inclusion or exclusion of Fc regions for endosomal recycling purposes, hydrodynamic sizes, and spatial arrangements, comprising up to 16 binding sites. Measurements of binding affinity and stoichiometry showed that the nanobody binding affinity was retained when incorporated into the Quad scaffold, and all nanobody domains were accessible for toxin binding, subsequently displaying increased blocking potency in vitro compared to the monomeric format. Moreover, functional assessment using automated patch-clamp assays demonstrated that the nanobody and Quads displayed neutralizing effects against long α-neurotoxins from both N. kaouthia and the forest cobra N. melanoleuca. This engineering approach offers a means of altering the valence, endosomal recyclability, and hydrodynamic size of existing nanobody-based therapeutics in a simple plug-and-play fashion and can thus serve as a technology for researchers tailoring therapeutic properties for improved neutralization of soluble targets such as snake toxins.


Subject(s)
Elapidae , Single-Domain Antibodies , Animals , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Elapidae/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Single-Domain Antibodies/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Ann Intern Med ; 174(9): 1261-1269, 2021 09.
Article in English | MEDLINE | ID: mdl-34251903

ABSTRACT

BACKGROUND: New treatment modalities are urgently needed for patients with COVID-19. The World Health Organization (WHO) Solidarity trial showed no effect of remdesivir or hydroxychloroquine (HCQ) on mortality, but the antiviral effects of these drugs are not known. OBJECTIVE: To evaluate the effects of remdesivir and HCQ on all-cause, in-hospital mortality; the degree of respiratory failure and inflammation; and viral clearance in the oropharynx. DESIGN: NOR-Solidarity is an independent, add-on, randomized controlled trial to the WHO Solidarity trial that included biobanking and 3 months of clinical follow-up (ClinicalTrials.gov: NCT04321616). SETTING: 23 hospitals in Norway. PATIENTS: Eligible patients were adults hospitalized with confirmed SARS-CoV-2 infection. INTERVENTION: Between 28 March and 4 October 2020, a total of 185 patients were randomly assigned and 181 were included in the full analysis set. Patients received remdesivir (n = 42), HCQ (n = 52), or standard of care (SoC) (n = 87). MEASUREMENTS: In addition to the primary end point of WHO Solidarity, study-specific outcomes were viral clearance in oropharyngeal specimens, the degree of respiratory failure, and inflammatory variables. RESULTS: No significant differences were seen between treatment groups in mortality during hospitalization. There was a marked decrease in SARS-CoV-2 load in the oropharynx during the first week overall, with similar decreases and 10-day viral loads among the remdesivir, HCQ, and SoC groups. Remdesivir and HCQ did not affect the degree of respiratory failure or inflammatory variables in plasma or serum. The lack of antiviral effect was not associated with symptom duration, level of viral load, degree of inflammation, or presence of antibodies against SARS-CoV-2 at hospital admittance. LIMITATION: The trial had no placebo group. CONCLUSION: Neither remdesivir nor HCQ affected viral clearance in hospitalized patients with COVID-19. PRIMARY FUNDING SOURCE: National Clinical Therapy Research in the Specialist Health Services, Norway.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , COVID-19/virology , Hydroxychloroquine/therapeutic use , Viral Load/drug effects , Adenosine Monophosphate/therapeutic use , Alanine/therapeutic use , Antibodies, Viral/blood , Biomarkers/blood , COVID-19/complications , COVID-19/mortality , Cause of Death , Female , Hospital Mortality , Humans , Inflammation/virology , Male , Middle Aged , Norway/epidemiology , Oropharynx/virology , Respiratory Insufficiency/virology , SARS-CoV-2/immunology , Severity of Illness Index , Standard of Care , Treatment Outcome
7.
Scand J Immunol ; 93(2): e13017, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33351196

ABSTRACT

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.


Subject(s)
Histocompatibility Antigens Class I/immunology , Mucous Membrane/immunology , Receptors, Fc/immunology , Animals , Antigen Presentation/immunology , Antigen-Antibody Complex/immunology , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunoglobulin G/immunology , Immunologic Factors/immunology
8.
Proc Natl Acad Sci U S A ; 115(41): 10440-10445, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30209217

ABSTRACT

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.


Subject(s)
Adenoviridae Infections/therapy , Adenoviridae/immunology , Antibodies/immunology , Fibrosarcoma/therapy , Genetic Therapy , Ribonucleoproteins/physiology , Vaccination , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Animals , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Gene Transfer Techniques , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , Transgenes , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 114(14): E2862-E2871, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28330995

ABSTRACT

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.


Subject(s)
Albumins/metabolism , Chemical and Drug Induced Liver Injury/genetics , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Acetaminophen/adverse effects , Acetaminophen/metabolism , Animals , Bile/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Dogs , Female , Hepatocytes/metabolism , Histocompatibility Antigens Class I/genetics , Homeostasis , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Fc/genetics , Serum Albumin, Human/genetics , Serum Albumin, Human/metabolism , Transcytosis/genetics
10.
Eur J Nucl Med Mol Imaging ; 46(11): 2311-2321, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31309259

ABSTRACT

PURPOSE: The aim of this study was to explore the ß-emitting lutetium-177 labelled anti-CD37 antibody NNV003 (177Lu-NNV003, Humalutin®) for the treatment of non-Hodgkin's lymphoma in in vitro studies and in animal models. METHODS: Cytotoxicity of 177Lu-NNV003 was measured in REC-1 (mantle cell lymphoma) and DOHH-2 (diffuse large B cell lymphoma) cell lines. Biodistribution was studied in mice bearing subcutaneous DOHH-2 or MEC-2 (chronic lymphocytic leukaemia) xenografts. The therapeutic effect of a single injection of 177Lu-NNV003 was measured in mice intravenously or subcutaneously injected with REC-1 cells. Haematological and histopathological assessments were used to evaluate the toxic effect of 177Lu-NNV003. The immunotherapeutic effect of NNV003 was assessed by measuring binding to Fcγ receptors, activation of ADCC and ADCP. NNV003's immunogenicity potential was assessed using in silico immunogenicity prediction tools. RESULTS: 177Lu-NNV003 showed an activity dependent antiproliferative effect in all cell lines. Maximum tumour uptake in vivo was 45% of injected activity/g in MEC-2 tumours and 15% injected activity/g in DOHH-2 tumours. In mice injected intravenously with REC-1 cells, 177Lu-NNV003 (50-100 MBq/kg) improved survival compared to control groups (p < 0.02). In mice with subcutaneous REC-1 xenografts, 500 MBq/kg 177Lu-NNV003 extended survival compared to the control treatments (p < 0.005). Transient haematological toxicity was observed in all mice treated with radioactivity. NNV003 induced ADCC and ADCP and was predicted to have a lower immunogenicity potential than its murine counterpart. CONCLUSION: 177Lu-NNV003 had a significant anti-tumour effect and a favourable toxicity profile. These results warrant further clinical testing in patients with CD37-expressing B cell malignancies.


Subject(s)
Antigens, Neoplasm/chemistry , Immunoconjugates/therapeutic use , Lutetium/chemistry , Lymphoma, Non-Hodgkin/therapy , Radioisotopes/chemistry , Tetraspanins/chemistry , Animals , Antibodies/chemistry , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Female , Humans , Immunotherapy , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Radioimmunotherapy , Radiometry , Tissue Distribution
11.
Immunol Rev ; 268(1): 328-39, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26497531

ABSTRACT

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.


Subject(s)
Antibody Specificity/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/metabolism , Ribonucleoproteins/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Cytosol/metabolism , Enzyme Activation , Host-Pathogen Interactions/immunology , Humans , Immunity , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/chemistry , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Ribonucleoproteins/chemistry , Ubiquitin-Protein Ligases/metabolism
12.
J Immunol ; 196(8): 3452-3459, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26962230

ABSTRACT

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.


Subject(s)
Adenoviruses, Human/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Antibody Affinity/immunology , Immunoglobulin G/immunology , Ribonucleoproteins/immunology , Animals , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Neutralization Tests , Ribonucleoproteins/genetics , Signal Transduction/immunology , Surface Plasmon Resonance
13.
Amino Acids ; 49(3): 489-500, 2017 03.
Article in English | MEDLINE | ID: mdl-27394141

ABSTRACT

In the extracellular environment, the enzyme transglutaminase 2 (TG2) is involved in cell-matrix interactions through association with the extracellular matrix protein, fibronectin (FN). The 45 kDa gelatin-binding domain of FN (45FN) is responsible for the binding to TG2. Previous studies have demonstrated that the FN-binding site of TG2 is located in the N-terminal domain of the enzyme although with conflicting results regarding the specific residues involved. Here we have mapped the FN interaction site of human TG2 by use of hydrogen/deuterium exchange coupled with mass spectrometry, and we confirm that the FN-binding site is located in the N-terminal domain of TG2. Furthermore, by combination of site-directed mutagenesis and surface plasmon resonance analysis we have identified the TG2 residues K30, R116 and H134 as crucial to maintain the high affinity interaction with FN. Mutation of all three residues simultaneously reduced binding to 45FN by more than 2000-fold. We also identified residues in the catalytic core domain of TG2 that contributed to FN binding, hence extending the binding interface between TG2 and FN. This study provides new insights into the high affinity interaction between TG2 and FN.


Subject(s)
Fibronectins/chemistry , GTP-Binding Proteins/chemistry , Protein Interaction Domains and Motifs , Transglutaminases/chemistry , Amino Acid Sequence , Antibodies/chemistry , Antibodies/isolation & purification , Catalytic Domain , Cloning, Molecular , Deuterium Exchange Measurement , Escherichia coli/genetics , Escherichia coli/metabolism , Fibronectins/genetics , Fibronectins/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Transglutaminases/genetics , Transglutaminases/metabolism
14.
J Immunol ; 194(11): 5497-508, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25904551

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/genetics , Complement C1q/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/genetics , Receptors, Fc/immunology , Receptors, IgG/immunology , Antibodies, Monoclonal/immunology , Antibody Affinity/genetics , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , HEK293 Cells , Hinge Exons/genetics , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/immunology , Nitrohydroxyiodophenylacetate/immunology , Phagocytosis/immunology , Protein Engineering , Receptors, Fc/genetics , Receptors, IgG/genetics , Surface Plasmon Resonance
15.
J Biol Chem ; 290(35): 21365-75, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26160175

ABSTRACT

Antibodies to the autoantigen transglutaminase 2 (TG2) are a hallmark of celiac disease. We have studied the interaction between TG2 and an anti-TG2 antibody (679-14-E06) derived from a single gut IgA plasma cell of a celiac disease patient. The antibody recognizes one of four identified epitopes targeted by antibodies of plasma cells of the disease lesion. The binding interface was identified by small angle x-ray scattering, ab initio and rigid body modeling using the known crystal structure of TG2 and the crystal structure of the antibody Fab fragment, which was solved at 2.4 Å resolution. The result was confirmed by testing binding of the antibody to TG2 mutants by ELISA and surface plasmon resonance. TG2 residues Arg-116 and His-134 were identified to be critical for binding of 679-14-E06 as well as other epitope 1 antibodies. In contrast, antibodies directed toward the two other main epitopes (epitopes 2 and 3) were not affected by these mutations. Molecular dynamics simulations suggest interactions of 679-14-E06 with the N-terminal domain of TG2 via the CDR2 and CDR3 loops of the heavy chain and the CDR2 loop of the light chain. In addition there were contacts of the framework 3 region of the heavy chain with the catalytic domain of TG2. The results provide an explanation for the biased usage of certain heavy and light chain gene segments by epitope 1-specific antibodies in celiac disease.


Subject(s)
Autoantibodies/immunology , Celiac Disease/enzymology , Celiac Disease/immunology , GTP-Binding Proteins/immunology , Immunoglobulin Fab Fragments/immunology , Transglutaminases/immunology , Autoantibodies/chemistry , Celiac Disease/genetics , Epitopes/chemistry , Epitopes/immunology , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Humans , Immunoglobulin Fab Fragments/chemistry , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Conformation , Protein Glutamine gamma Glutamyltransferase 2 , Scattering, Small Angle , Surface Plasmon Resonance , Transglutaminases/chemistry , Transglutaminases/genetics , X-Ray Diffraction
17.
J Biol Chem ; 289(50): 34583-94, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25344603

ABSTRACT

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.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Amino Acid Substitution , Binding, Competitive , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Protein Binding , Protein Stability , Protein Structure, Tertiary , Serum Albumin/genetics
18.
J Biol Chem ; 289(19): 13492-502, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24652290

ABSTRACT

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.


Subject(s)
Albumins/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Albumins/genetics , Albumins/pharmacology , Amino Acid Substitution , Animals , Female , Half-Life , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/pharmacology , Humans , Macaca fascicularis , Mice , Mutation, Missense , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
19.
J Biol Chem ; 289(24): 17228-39, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24764301

ABSTRACT

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.


Subject(s)
Albumins/metabolism , Histocompatibility Antigens Class I/chemistry , Receptors, Fc/chemistry , Amino Acid Sequence , Antibodies, Blocking/immunology , Antibodies, Monoclonal/immunology , Binding Sites , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism
20.
Blood ; 122(7): 1174-81, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23843496

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Fetus/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Maternal-Fetal Exchange/immunology , Placenta/immunology , Receptors, Fc/immunology , Recombinant Proteins/immunology , Antibodies/metabolism , Binding Sites , Biological Transport , Choriocarcinoma/immunology , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fetus/metabolism , Flow Cytometry , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/metabolism , Infant, Newborn , Placenta/metabolism , Pregnancy , Receptors, Fc/metabolism , Recombinant Proteins/metabolism , Uterine Neoplasms/immunology , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL