Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
PLoS Pathog ; 20(8): e1012453, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146376

ABSTRACT

Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by human sera is a strong correlate of protection against symptomatic and severe Coronavirus Disease 2019 (COVID-19). The emergence of antigenically distinct SARS-CoV-2 variants of concern (VOCs) and the relatively rapid waning of serum antibody titers, however, raises questions about the sustainability of serum protection. In addition to serum neutralization, other antibody functionalities and the memory B cell (MBC) response are suggested to help maintaining this protection. In this study, we investigate the breadth of spike (S) protein-specific serum antibodies that mediate effector functions by interacting with Fc-gamma receptor IIa (FcγRIIa) and FcγRIIIa, and of the receptor binding domain (RBD)-specific MBCs, following a primary SARS-CoV-2 infection with the D614G, Alpha, Beta, Gamma, Delta, Omicron BA.1 or BA.2 variant. Irrespectively of the variant causing the infection, the breadth of S protein-specific serum antibodies that interact with FcγRIIa and FcγRIIIa and the RBD-specific MBC responses exceeded the breadth of serum neutralization, although the Alpha-induced B cell response seemed more strain-specific. Between VOC groups, both quantitative and qualitative differences in the immune responses were observed, suggesting differences in immunogenicity. Overall, this study contributes to the understanding of protective humoral and B cell responses in the light of emerging antigenically distinct VOCs, and highlights the need to study the immune system beyond serum neutralization to gain a better understanding of the protection against emerging variants.


Subject(s)
Antibodies, Viral , COVID-19 , Memory B Cells , Receptors, IgG , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/immunology , Receptors, IgG/immunology , Memory B Cells/immunology , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology
2.
Blood ; 131(1): 131-143, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29061569

ABSTRACT

Most patients with acute myeloid leukemia (AML) can only be cured when allogeneic hematopoietic stem-cell transplantation induces a graft-versus-leukemia immune response (GVL). Although the role of T cells and natural killer cells in tumor immunology has been established, less is known about the contribution of B cells. From B cells of high-risk patients with AML with potent and lasting GVL responses, we isolated monoclonal antibodies directed against antigens expressed on the cell surface of AML cells but not on normal hematopoietic and nonhematopoietic cells. A number of these donor-derived antibodies recognized the U5 snRNP200 complex, a component of the spliceosome that in normal cells is found in the cell. In AML however, the U5 snRNP200 complex is exposed on the cell membrane of leukemic blasts. U5 snRNP200 complex-specific antibodies induced death of AML cells in an Fc receptor-dependent way in the absence of cytotoxic leukocytes or complement. In an AML mouse model, treatment with U5 snRNP200 complex-specific antibodies led to significant tumor growth inhibition. Thus, donor-derived U5 snRNP200 complex-recognizing AML-specific antibodies may contribute to antitumor responses.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/immunology , Graft vs Leukemia Effect/immunology , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Ribonucleoprotein, U5 Small Nuclear/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Animals , Combined Modality Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Male , Mice, SCID , Middle Aged , Prognosis
3.
Immunol Rev ; 270(1): 65-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864105

ABSTRACT

Monoclonal antibodies are essential therapeutics and diagnostics in a large number of diseases. Moreover, they are essential tools in all sectors of life sciences. Although the great majority of monoclonal antibodies currently in use are of mouse origin, the use of human B cells to generate monoclonal antibodies is increasing as new techniques to tap the human B cell repertoire are rapidly emerging. Cloned lines of immortalized human B cells are ideal sources of monoclonal antibodies. In this review, we summarize our studies to the regulation of the replicative life span, differentiation, and maturation of B cells that led to the development of a platform that uses immortalization of human B cells by in vitro genetic modification for antibody development. We describe a number of human antibodies that were isolated using this platform and the application of the technique in other species. We also discuss the use of immortalized B cells as antigen-presenting cells for the discovery of tumor neoantigens.


Subject(s)
Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Self Renewal , Animals , Antibodies, Monoclonal/biosynthesis , Antibody Formation/immunology , Antibody-Producing Cells/cytology , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/cytology , Cell Culture Techniques , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Drug Discovery , Gene Expression Regulation , Germinal Center/cytology , Germinal Center/physiology , Humans , Immunologic Memory , Interleukins/metabolism , Phenotype , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Vaccines/immunology , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
J Hepatol ; 71(1): 14-24, 2019 07.
Article in English | MEDLINE | ID: mdl-30797052

ABSTRACT

BACKGROUND & AIMS: In order to design an effective vaccine against hepatitis C virus (HCV) infection, it is necessary to understand immune protection. A number of broadly reactive neutralizing antibodies have been isolated from B cells of HCV-infected patients. However, it remains unclear whether B cells producing such antibodies contribute to HCV clearance and long-term immune protection against HCV. METHODS: We analysed the B cell repertoire of 13 injecting drug users from the Amsterdam Cohort Study, who were followed up for a median of 17.5 years after primary infection. Individuals were classified into 2 groups based on the outcome of HCV infection: 5 who became chronically infected either after primary infection or after reinfection, and 8 who were HCV RNA negative following spontaneous clearance of ≥1 HCV infection(s). From each individual, 10,000 CD27+IgG+B cells, collected 0.75 year after HCV infection, were cultured to characterize the antibody repertoire. RESULTS: Using a multiplex flow cytometry-based assay to study the antibody binding to E1E2 from genotype 1 to 6, we found that a high frequency of cross-genotype antibodies was associated with spontaneous clearance of 1 or multiple infections (p = 0.03). Epitope specificity of these cross-genotype antibodies was determined by alanine mutant scanning in 4 individuals who were HCV RNA negative following spontaneous clearance of 1 or multiple infections. Interestingly, the cross-genotype antibodies were mainly antigenic region 3 (AR3)-specific and showed cross-neutralizing activity against HCV. In addition to AR3 antibodies, 3 individuals developed antibodies recognizing antigenic region 4, of which 1 monoclonal antibody showed cross-neutralizing capacity. CONCLUSIONS: Together, these data suggest that a strong B cell response producing cross-genotype and neutralizing antibodies, especially targeting AR3, contributes to HCV clearance and long-term immune protection against HCV. LAY SUMMARY: Although effective treatments against hepatitis C virus (HCV) are available, 500,000 people die from liver disease caused by HCV each year and approximately 1.75 million people are newly infected. This could be prevented by a vaccine. To design a vaccine against HCV, more insight into the role of antibodies in the protection against HCV infection is needed. In a cohort of injecting drug users, we found that antibodies interfering with virus cell entry, and recognizing multiple HCV genotypes, conferred long-term protection against chronic HCV infection.


Subject(s)
Antibodies, Neutralizing , Epitopes, B-Lymphocyte/immunology , Hepacivirus , Hepatitis C Antibodies , Hepatitis C, Chronic , Substance Abuse, Intravenous/virology , Viral Hepatitis Vaccines/pharmacology , Adaptive Immunity/immunology , Adult , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/blood , Female , Hepacivirus/genetics , Hepacivirus/immunology , Hepacivirus/isolation & purification , Hepatitis C Antibodies/biosynthesis , Hepatitis C Antibodies/blood , Hepatitis C, Chronic/etiology , Hepatitis C, Chronic/immunology , Humans , Immunologic Memory , Male , RNA, Viral/isolation & purification , Substance Abuse, Intravenous/complications , Viral Envelope Proteins/immunology
5.
Eur J Immunol ; 48(11): 1796-1809, 2018 11.
Article in English | MEDLINE | ID: mdl-30184252

ABSTRACT

Type I and type III interferons (IFNs) are fundamental for antiviral immunity, but prolonged expression is also detrimental to the host. Therefore, upon viral infection high levels of type I and III IFNs are followed by a strong and rapid decline. However, the mechanisms responsible for this suppression are still largely unknown. Here, we show that IgG opsonization of model viruses influenza and respiratory syncytial virus (RSV) strongly and selectively suppressed type I and III IFN production by various human antigen-presenting cells. This suppression was induced by selective inhibition of TLR, RIG-I-like receptor, and STING-dependent type I and III IFN gene transcription. Surprisingly, type I and III IFN suppression was mediated by Syk and PI3K independent inhibitory signaling via FcγRIIa, thereby identifying a novel non-canonical FcγRIIa pathway in myeloid cells. Together, these results indicate that IgG opsonization of viruses functions as a novel negative feedback mechanism in humans, which may play a role in the selective suppression of type I and III IFN responses during the late-phase of viral infections. In addition, activation of this pathway may be used as a tool to limit type I IFN-associated pathology.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Myeloid Cells/immunology , Receptors, IgG/immunology , Animals , Antigen-Presenting Cells/immunology , Cells, Cultured , Female , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Respiratory Syncytial Viruses/immunology , Signal Transduction/immunology , Syk Kinase/immunology , Transcription, Genetic/immunology , Virus Diseases/immunology , Interferon Lambda
6.
PLoS Pathog ; 11(7): e1005035, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26161532

ABSTRACT

Prevention efforts for respiratory syncytial virus (RSV) have been advanced due to the recent isolation and characterization of antibodies that specifically recognize the prefusion conformation of the RSV fusion (F) glycoprotein. These potently neutralizing antibodies are in clinical development for passive prophylaxis and have also aided the design of vaccine antigens that display prefusion-specific epitopes. To date, prefusion-specific antibodies have been shown to target two antigenic sites on RSV F, but both of these sites are also present on monomeric forms of F. Here we present a structural and functional characterization of human antibody AM14, which potently neutralized laboratory strains and clinical isolates of RSV from both A and B subtypes. The crystal structure and location of escape mutations revealed that AM14 recognizes a quaternary epitope that spans two protomers and includes a region that undergoes extensive conformational changes in the pre- to postfusion F transition. Binding assays demonstrated that AM14 is unique in its specific recognition of trimeric furin-cleaved prefusion F, which is the mature form of F on infectious virions. These results demonstrate that the prefusion F trimer contains potent neutralizing epitopes not present on monomers and that AM14 should be particularly useful for characterizing the conformational state of RSV F-based vaccine antigens.


Subject(s)
Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/ultrastructure , Epitopes, B-Lymphocyte/ultrastructure , Respiratory Syncytial Viruses/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cell Line , Chromatography, Gel , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Flow Cytometry , Glycoproteins/chemistry , Glycoproteins/immunology , Glycoproteins/ultrastructure , Humans , Protein Structure, Quaternary , Surface Plasmon Resonance
7.
Proc Natl Acad Sci U S A ; 111(47): 16820-5, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385586

ABSTRACT

Bispecific antibodies have therapeutic potential by expanding the functions of conventional antibodies. Many different formats of bispecific antibodies have meanwhile been developed. Most are genetic modifications of the antibody backbone to facilitate incorporation of two different variable domains into a single molecule. Here, we present a bispecific format where we have fused two full-sized IgG antibodies via their C termini using sortase transpeptidation and click chemistry to create a covalently linked IgG antibody heterodimer. By linking two potent anti-influenza A antibodies together, we have generated a full antibody dimer with bispecific activity that retains the activity and stability of the two fusion partners.


Subject(s)
Antibodies, Bispecific/biosynthesis , Click Chemistry , Influenza A virus/immunology , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , B-Lymphocytes/virology , Blotting, Western , Cells, Cultured , Dimerization , Electrophoresis, Polyacrylamide Gel , Humans , Influenza A virus/classification , Surface Plasmon Resonance
8.
Proc Natl Acad Sci U S A ; 111(1): 445-50, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24335589

ABSTRACT

The discovery and characterization of broadly neutralizing antibodies (bnAbs) against influenza viruses have raised hopes for the development of monoclonal antibody (mAb)-based immunotherapy and the design of universal influenza vaccines. Only one human bnAb (CR8020) specifically recognizing group 2 influenza A viruses has been previously characterized that binds to a highly conserved epitope at the base of the hemagglutinin (HA) stem and has neutralizing activity against H3, H7, and H10 viruses. Here, we report a second group 2 bnAb, CR8043, which was derived from a different germ-line gene encoding a highly divergent amino acid sequence. CR8043 has in vitro neutralizing activity against H3 and H10 viruses and protects mice against challenge with a lethal dose of H3N2 and H7N7 viruses. The crystal structure and EM reconstructions of the CR8043-H3 HA complex revealed that CR8043 binds to a site similar to the CR8020 epitope but uses an alternative angle of approach and a distinct set of interactions. The identification of another antibody against the group 2 stem epitope suggests that this conserved site of vulnerability has great potential for design of therapeutics and vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Influenza A virus/chemistry , Animals , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Immunologic Memory , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Kinetics , Mice , Mice, Inbred BALB C , Microscopy, Electron , Models, Molecular , Molecular Conformation , Species Specificity
9.
J Virol ; 89(18): 9571-80, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26157123

ABSTRACT

UNLABELLED: Since it was first recognized in 2004 that human parechoviruses (HPeV) are a significant cause of central nervous system and neonatal sepsis, their clinical importance, primarily in children, has started to emerge. Intravenous immunoglobulin treatment is the only treatment available in such life-threatening cases and has given moderate success. Direct inhibition of parechovirus infection using monoclonal antibodies is a potential treatment. We have developed two neutralizing monoclonal antibodies against HPeV1 and HPeV2, namely, AM18 and AM28, which also cross-neutralize other viruses. Here, we present the mapping of their epitopes using peptide scanning, surface plasmon resonance, fluorescence-based thermal shift assays, electron cryomicroscopy, and image reconstruction. We determined by peptide scanning and surface plasmon resonance that AM18 recognizes a linear epitope motif including the arginine-glycine-aspartic acid on the C terminus of capsid protein VP1. This epitope is normally used by the virus to attach to host cell surface integrins during entry and is found in 3 other viruses that AM18 neutralizes. Therefore, AM18 is likely to cause virus neutralization by aggregation and by blocking integrin binding to the capsid. Further, we show by electron cryomicroscopy, three-dimensional reconstruction, and pseudoatomic model fitting that ordered RNA interacts with HPeV1 VP1 and VP3. AM28 recognizes quaternary epitopes on the capsid composed of VP0 and VP3 loops from neighboring pentamers, thereby increasing the RNA accessibility temperature for the virus-AM28 complex compared to the virus alone. Thus, inhibition of RNA uncoating probably contributes to neutralization by AM28. IMPORTANCE: Human parechoviruses can cause mild infections to severe diseases in young children, such as neonatal sepsis, encephalitis, and cardiomyopathy. Intravenous immunoglobulin treatment is the only treatment available in such life-threatening cases. In order to develop more targeted treatment, we have searched for human monoclonal antibodies that would neutralize human parechoviruses 1 and 2, associated with mild infections such as gastroenteritis and severe infections of the central nervous system, and thus allow safe treatment. In the current study, we show how two such promising antibodies interact with the virus, modeling the atomic interactions between the virus and the antibody to propose how neutralization occurs. Both antibodies can cause aggregation; in addition, one antibody interferes with the virus recognizing its target cell, while the other, recognizing only the whole virus, inhibits the genome uncoating and replication in the cell.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Capsid Proteins/chemistry , Models, Molecular , Parechovirus/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid Proteins/immunology , Cell Line, Tumor , Cross Reactions , Humans , Parechovirus/immunology , Protein Structure, Secondary , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL