Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Publication year range
1.
Immunity ; 56(11): 2463-2465, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37967527

ABSTRACT

The influenza neuraminidase has historically been understudied compared to its surface protein counterpart, hemagglutinin. In two recent Immunity papers, Hansen et al. and Lei et al. bolster resurging interest in neuraminidase-targeting antibodies and their implications for therapy and "universal" vaccines.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Humans , Neuraminidase , Antibodies, Viral , Hemagglutinins , Hemagglutinin Glycoproteins, Influenza Virus
3.
Immunity ; 52(6): 1022-1038.e7, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32454024

ABSTRACT

Class-switched antibodies to double-stranded DNA (dsDNA) are prevalent and pathogenic in systemic lupus erythematosus (SLE), yet mechanisms of their development remain poorly understood. Humans and mice lacking secreted DNase DNASE1L3 develop rapid anti-dsDNA antibody responses and SLE-like disease. We report that anti-DNA responses in Dnase1l3-/- mice require CD40L-mediated T cell help, but proceed independently of germinal center formation via short-lived antibody-forming cells (AFCs) localized to extrafollicular regions. Type I interferon (IFN-I) signaling and IFN-I-producing plasmacytoid dendritic cells (pDCs) facilitate the differentiation of DNA-reactive AFCs in vivo and in vitro and are required for downstream manifestations of autoimmunity. Moreover, the endosomal DNA sensor TLR9 promotes anti-dsDNA responses and SLE-like disease in Dnase1l3-/- mice redundantly with another nucleic acid-sensing receptor, TLR7. These results establish extrafollicular B cell differentiation into short-lived AFCs as a key mechanism of anti-DNA autoreactivity and reveal a major contribution of pDCs, endosomal Toll-like receptors (TLRs), and IFN-I to this pathway.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Communication , DNA/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon Type I/metabolism , Animals , Antibodies, Antinuclear/immunology , Autoantigens/immunology , Autoimmunity , Biomarkers , CD40 Ligand/deficiency , Cell Communication/genetics , Cell Communication/immunology , Disease Models, Animal , Disease Susceptibility , Endodeoxyribonucleases/deficiency , Fluorescent Antibody Technique , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/pathology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Knockout , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism
4.
Immunity ; 50(6): 1530-1541.e8, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31216462

ABSTRACT

Rapidly evolving RNA viruses, such as the GII.4 strain of human norovirus (HuNoV), and their vaccines elicit complex serological responses associated with previous exposure. Specific correlates of protection, moreover, remain poorly understood. Here, we report the GII.4-serological antibody repertoire-pre- and post-vaccination-and select several antibody clonotypes for epitope and structural analysis. The humoral response was dominated by GII.4-specific antibodies that blocked ancestral strains or by antibodies that bound to divergent genotypes and did not block viral-entry-ligand interactions. However, one antibody, A1431, showed broad blockade toward tested GII.4 strains and neutralized the pandemic GII.P16-GII.4 Sydney strain. Structural mapping revealed conserved epitopes, which were occluded on the virion or partially exposed, allowing for broad blockade with neutralizing activity. Overall, our results provide high-resolution molecular information on humoral immune responses after HuNoV vaccination and demonstrate that infection-derived and vaccine-elicited antibodies can exhibit broad blockade and neutralization against this prevalent human pathogen.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Caliciviridae Infections/immunology , Caliciviridae Infections/prevention & control , Norovirus/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/chemistry , Caliciviridae Infections/epidemiology , Caliciviridae Infections/virology , Cell Line , Conserved Sequence , Epitopes/chemistry , Epitopes/immunology , Humans , Immunoglobulin G/immunology , Models, Molecular , Norovirus/classification , Protein Binding , Protein Conformation , Recombinant Proteins/immunology , Vaccination
5.
Cell ; 147(3): 653-65, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036571

ABSTRACT

Sensory-motor circuits in the spinal cord are constructed with a fine specificity that coordinates motor behavior, but the mechanisms that direct sensory connections with their motor neuron partners remain unclear. The dorsoventral settling position of motor pools in the spinal cord is known to match the distal-to-proximal position of their muscle targets in the limb, but the significance of invariant motor neuron positioning is unknown. An analysis of sensory-motor connectivity patterns in FoxP1 mutant mice, where motor neuron position has been scrambled, shows that the final pattern of sensory-motor connections is initiated by the projection of sensory axons to discrete dorsoventral domains of the spinal cord without regard for motor neuron subtype or, indeed, the presence of motor neurons. By implication, the clustering and dorsoventral settling position of motor neuron pools serve as a determinant of the pattern of sensory input specificity and thus motor coordination.


Subject(s)
Body Patterning , Motor Neurons/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Electromyography , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hindlimb/innervation , Mice , Mutation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sensory Receptor Cells/metabolism
6.
J Proteome Res ; 21(7): 1616-1627, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35653804

ABSTRACT

In this study, we used multiple enzyme digestions, coupled with higher-energy collisional dissociation (HCD) and electron-transfer/higher-energy collision dissociation (EThcD) fragmentation to develop a mass-spectrometric (MS) method for determining the complete protein sequence of monoclonal antibodies (mAbs). The method was refined on an mAb of a known sequence, a SARS-CoV-1 antireceptor binding domain (RBD) spike monoclonal antibody. The data were searched using Supernovo to generate a complete template-assisted de novo sequence for this and two SARS-CoV-2 mAbs of known sequences resulting in correct sequences for the variable regions and correct distinction of Ile and Leu residues. We then used the method on a set of 25 antihemagglutinin (HA) influenza antibodies of unknown sequences and determined high confidence sequences for >99% of the complementarity determining regions (CDRs). The heavy-chain and light-chain genes were cloned and transfected into cells for recombinant expression followed by affinity purification. The recombinant mAbs displayed binding curves matching the original mAbs with specificity to the HA influenza antigen. Our findings indicate that this methodology results in almost complete antibody sequence coverage with high confidence results for CDR regions on diverse mAb sequences.


Subject(s)
COVID-19 , Influenza, Human , Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , COVID-19/diagnosis , Humans , Mass Spectrometry , SARS-CoV-2/genetics
7.
Am J Pathol ; 190(8): 1680-1690, 2020 08.
Article in English | MEDLINE | ID: mdl-32473109

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has spread globally, and no proven treatments are available. Convalescent plasma therapy has been used with varying degrees of success to treat severe microbial infections for >100 years. Patients (n = 25) with severe and/or life-threatening COVID-19 disease were enrolled at the Houston Methodist hospitals from March 28, 2020, to April 14, 2020. Patients were transfused with convalescent plasma, obtained from donors with confirmed severe acute respiratory syndrome coronavirus 2 infection who had recovered. The primary study outcome was safety, and the secondary outcome was clinical status at day 14 after transfusion. Clinical improvement was assessed on the basis of a modified World Health Organization six-point ordinal scale and laboratory parameters. Viral genome sequencing was performed on donor and recipient strains. At day 7 after transfusion with convalescent plasma, nine patients had at least a one-point improvement in clinical scale, and seven of those were discharged. By day 14 after transfusion, 19 (76%) patients had at least a one-point improvement in clinical status, and 11 were discharged. No adverse events as a result of plasma transfusion were observed. Whole genome sequencing data did not identify a strain genotype-disease severity correlation. The data indicate that administration of convalescent plasma is a safe treatment option for those with severe COVID-19 disease.


Subject(s)
Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Adult , Aged , Betacoronavirus/genetics , COVID-19 , Female , Humans , Immunization, Passive , Investigational New Drug Application , Male , Middle Aged , Pandemics , SARS-CoV-2 , Texas , Whole Genome Sequencing , Young Adult , COVID-19 Serotherapy
8.
J Immunol ; 200(12): 3962-3969, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29703863

ABSTRACT

FCRL4, a low-affinity IgA Ab receptor with strong immunoregulatory potential, is an identifying feature of a tissue-based population of memory B cells (Bmem). We used two independent approaches to perform a comparative analysis of the Ag receptor repertoires of FCRL4+ and FCRL4- Bmem in human tonsils. We determined that FCRL4+ Bmem displayed lower levels of somatic mutations in their Ag receptors compared with FCRL4- Bmem but had similar frequencies of variable gene family usage. Importantly, Abs with reactivity to commensal microbiota were enriched in FCRL4+ cells, a phenotype not due to polyreactive binding characteristics. Our study links expression of the immunoregulatory FCRL4 molecule with increased recognition of commensal microbial Ags.


Subject(s)
Antibodies/immunology , Antigens/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Microbiota/immunology , Receptors, Fc/immunology , Cell Line , Gene Expression/immunology , HEK293 Cells , Humans , Immunoglobulin A/immunology , Lymphocyte Activation/immunology , Phenotype
9.
Proc Natl Acad Sci U S A ; 113(19): E2636-45, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114511

ABSTRACT

Elucidating how antigen exposure and selection shape the human antibody repertoire is fundamental to our understanding of B-cell immunity. We sequenced the paired heavy- and light-chain variable regions (VH and VL, respectively) from large populations of single B cells combined with computational modeling of antibody structures to evaluate sequence and structural features of human antibody repertoires at unprecedented depth. Analysis of a dataset comprising 55,000 antibody clusters from CD19(+)CD20(+)CD27(-) IgM-naive B cells, >120,000 antibody clusters from CD19(+)CD20(+)CD27(+) antigen-experienced B cells, and >2,000 RosettaAntibody-predicted structural models across three healthy donors led to a number of key findings: (i) VH and VL gene sequences pair in a combinatorial fashion without detectable pairing restrictions at the population level; (ii) certain VH:VL gene pairs were significantly enriched or depleted in the antigen-experienced repertoire relative to the naive repertoire; (iii) antigen selection increased antibody paratope net charge and solvent-accessible surface area; and (iv) public heavy-chain third complementarity-determining region (CDR-H3) antibodies in the antigen-experienced repertoire showed signs of convergent paired light-chain genetic signatures, including shared light-chain third complementarity-determining region (CDR-L3) amino acid sequences and/or Vκ,λ-Jκ,λ genes. The data reported here address several longstanding questions regarding antibody repertoire selection and development and provide a benchmark for future repertoire-scale analyses of antibody responses to vaccination and disease.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , High-Throughput Nucleotide Sequencing/methods , Sequence Alignment/methods , Amino Acid Sequence , Antibodies/genetics , Antigen-Antibody Complex/genetics , Base Sequence , Computer Simulation , High-Throughput Screening Assays/methods , Humans , Models, Chemical , Models, Genetic , Models, Immunological , Sequence Homology, Amino Acid
10.
Proc Natl Acad Sci U S A ; 113(5): E577-86, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787899

ABSTRACT

High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however, remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional enforcement of ABC-DLBCL hallmarks, including the classical NF-κB and MYD88 (myeloid differentiation primary response gene 88) pathways. FOXP1 promoted gene expression underlying transition of the GCB cell to the plasmablast--the transient B-cell stage targeted in ABC-DLBCL transformation--by antagonizing pathways distinctive of GCB-DLBCL, including that of the GCB "master regulator," BCL6 (B-cell lymphoma 6). Cell-line derived FOXP1 target genes that were highly correlated with FOXP1 expression in primary DLBCL accurately segregated the corresponding clinical subtypes of a large cohort of primary DLBCL isolates and identified conserved pathways associated with ABC-DLBCL pathology.


Subject(s)
B-Lymphocytes/immunology , Forkhead Transcription Factors/physiology , Lymphoma, Large B-Cell, Diffuse/immunology , Repressor Proteins/physiology , Cell Differentiation , Cell Line, Tumor , Humans , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Transcription, Genetic
11.
Cancer Immunol Immunother ; 67(5): 729-738, 2018 May.
Article in English | MEDLINE | ID: mdl-29427082

ABSTRACT

A better understanding of antitumor immune responses is the key to advancing the field of cancer immunotherapy. Endogenous immunity in cancer patients, such as circulating anticancer antibodies or tumor-reactive B cells, has been historically yet incompletely described. Here, we demonstrate that tumor-draining (sentinel) lymph node (SN) is a rich source for tumor-reactive B cells that give rise to systemic IgG anticancer antibodies circulating in the bloodstream of breast cancer patients. Using a synergistic combination of high-throughput B-cell sequencing and quantitative immunoproteomics, we describe the prospective identification of tumor-reactive SN B cells (based on clonal frequency) and also demonstrate an unequivocal link between affinity-matured expanded B-cell clones in the SN and antitumor IgG in the blood. This technology could facilitate the discovery of antitumor antibody therapeutics and conceivably identify novel tumor antigens. Lastly, these findings highlight the unique and specialized niche the SN can fill in the advancement of cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Breast Neoplasms/immunology , Clone Cells/immunology , Immunoglobulin G/immunology , Sentinel Lymph Node/immunology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured , Female , Humans , Sequence Homology
12.
Hepatology ; 66(3): 885-895, 2017 09.
Article in English | MEDLINE | ID: mdl-28470667

ABSTRACT

A major problem in autoimmunity has been identification of the earliest events that lead to breach of tolerance. Although there have been major advances in dissecting effector pathways and the multilineage immune responses to mitochondrial self-antigens in primary biliary cholangitis, the critical links between environmental factors and tolerance remain elusive. We hypothesized that environmental xenobiotic modification of the E2 subunit of the pyruvate dehydrogenase (PDC-E2) inner lipoyl domain can lead to loss of tolerance to genetically susceptible hosts. Previously we demonstrated that serum anti-PDC-E2 autoantibodies cross-react with the chemical xenobiotics 2-octynoic acid and 6,8-bis (acetylthio) octanoic acid and further that there is a high frequency of PDC-E2-specific peripheral plasmablasts. Herein we generated 104 recombinant monoclonal antibodies (mAbs) based on paired heavy-chain and light-chain variable regions of individual plasmablasts derived from primary biliary cholangitis patients. We identified 32 mAbs reactive with native PDC-E2, including 20 specific for PDC-E2 and 12 cross-reactive with both PDC-E2 and 2-octynoic acid and 6,8-bis (acetylthio) octanoic acid. A lower frequency of replacement somatic hypermutations, indicating a lower level of affinity maturation, was observed in the complementarity-determining regions of the cross-reactive mAbs in comparison to mAbs exclusively recognizing PDC-E2 or those for irrelevant antigens. In particular, when the highly mutated heavy-chain gene of a cross-reactive mAb was reverted to the germline sequence, the PDC-E2 reactivity was reduced dramatically, whereas the xenobiotic reactivity was retained. Importantly, cross-reactive mAbs also recognized lipoic acid, a mitochondrial fatty acid that is covalently bound to PDC-E2. CONCLUSION: Our data reflect that chemically modified lipoic acid or lipoic acid itself, through molecular mimicry, is the initial target that leads to the development of primary biliary cholangitis. (Hepatology 2017;66:885-895).


Subject(s)
Antibodies, Monoclonal/immunology , Autoantigens/immunology , Autoimmunity/genetics , Cholangitis/immunology , Cholangitis/pathology , Xenobiotics/immunology , Antibodies, Monoclonal/metabolism , Autoantigens/genetics , Autoimmunity/immunology , Female , Gene Amplification , Humans , Immunoblotting , Male , Molecular Mimicry/genetics , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Thioctic Acid/immunology , Thioctic Acid/metabolism
13.
Biologicals ; 46: 81-87, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28131552

ABSTRACT

Next-Generation Sequencing combined with bioinformatics is a powerful tool for analyzing the large number of DNA sequences present in the expressed antibody repertoire and these data sets can be used to advance a number of research areas including antibody discovery and engineering. The accurate measurement of the immune repertoire sequence composition, diversity and abundance is important for understanding the repertoire response in infections, vaccinations and cancer immunology and could also be useful for elucidating novel molecular targets. In this study 4 individual domestic cats (Felis catus) were subjected to antibody repertoire sequencing with total number of sequences generated 1079863 for VH for IgG, 1050824 VH for IgM, 569518 for VK and 450195 for VL. Our analysis suggests that a similar VDJ expression patterns exists across all cats. Similar to the canine repertoire, the feline repertoire is dominated by a single subgroup, namely VH3. The antibody paratope of felines showed similar amino acid variation when compared to human, mouse and canine counterparts. All animals show a similarly skewed VH CDR-H3 profile and, when compared to canine, human and mouse, distinct differences are observed. Our study represents the first attempt to characterize sequence diversity in the expressed feline antibody repertoire and this demonstrates the utility of using NGS to elucidate entire antibody repertoires from individual animals. These data provide significant insight into understanding the feline immune system function.


Subject(s)
Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Amino Acid Sequence , Animals , Antibodies/genetics , Cats , Complementarity Determining Regions/genetics , Dogs , Gene Expression Profiling/methods , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction , VDJ Exons/genetics
14.
Proc Natl Acad Sci U S A ; 111(11): E998-1006, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591644

ABSTRACT

The plasmacytoid dendritic cell (pDC) is vital to the coordinated action of innate and adaptive immunity. pDC development has not been unequivocally traced, nor has its transcriptional regulatory network been fully clarified. Here we confirm an essential requirement for the BCL11A transcription factor in fetal pDC development, and demonstrate this lineage-specific requirement in the adult organism. Furthermore, we identify BCL11A gene targets and provide a molecular mechanism for its action in pDC commitment. Embryonic germ-line deletion of Bcl11a revealed an absolute cellular, molecular, and functional absence of pDCs in fetal mice. In adults, deletion of Bcl11a in hematopoietic stem cells resulted in perturbed yet continued generation of progenitors, loss of downstream pDC and B-cell lineages, and persisting myeloid, conventional dendritic, and T-cell lineages. Challenge with virus resulted in a marked reduction of antiviral response in conditionally deleted adults. Genome-wide analyses of BCL11A DNA binding and expression revealed that BCL11A regulates transcription of E2-2 and other pDC differentiation modulators, including ID2 and MTG16. Our results identify BCL11A as an essential, lineage-specific factor that regulates pDC development, supporting a model wherein differentiation into pDCs represents a primed "default" pathway for common dendritic cell progenitors.


Subject(s)
Carrier Proteins/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Nuclear Proteins/immunology , Animals , B-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Knockout , Mice, Mutant Strains , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction
15.
Proc Natl Acad Sci U S A ; 111(6): 2259-64, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469811

ABSTRACT

Most vaccines confer protection via the elicitation of serum antibodies, yet more than 100 y after the discovery of antibodies, the molecular composition of the human serum antibody repertoire to an antigen remains unknown. Using high-resolution liquid chromatography tandem MS proteomic analyses of serum antibodies coupled with next-generation sequencing of the V gene repertoire in peripheral B cells, we have delineated the human serum IgG and B-cell receptor repertoires following tetanus toxoid (TT) booster vaccination. We show that the TT(+) serum IgG repertoire comprises ∼100 antibody clonotypes, with three clonotypes accounting for >40% of the response. All 13 recombinant IgGs examined bound to vaccine antigen with Kd ∼ 10(-8)-10(-10) M. Five of 13 IgGs recognized the same linear epitope on TT, occluding the binding site used by the toxin for cell entry, suggesting a possible explanation for the mechanism of protection conferred by the vaccine. Importantly, only a small fraction (<5%) of peripheral blood plasmablast clonotypes (CD3(-)CD14(-)CD19(+)CD27(++)CD38(++)CD20(-)TT(+)) at the peak of the response (day 7), and an even smaller fraction of memory B cells, were found to encode antibodies that could be detected in the serological memory response 9 mo postvaccination. This suggests that only a small fraction of responding peripheral B cells give rise to the bone marrow long-lived plasma cells responsible for the production of biologically relevant amounts of vaccine-specific antibodies (near or above the Kd). Collectively, our results reveal the nature and dynamics of the serological response to vaccination with direct implications for vaccine design and evaluation.


Subject(s)
Antibodies, Bacterial/biosynthesis , Tetanus Toxoid/administration & dosage , Amino Acid Sequence , Antibodies, Bacterial/chemistry , B-Lymphocytes/immunology , Chromatography, Liquid , Humans , Immunophenotyping , Molecular Sequence Data , Tandem Mass Spectrometry
16.
Nature ; 460(7259): 1093-7, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19657335

ABSTRACT

The contribution of changes in cis-regulatory elements or trans-acting factors to interspecies differences in gene expression is not well understood. The mammalian beta-globin loci have served as a model for gene regulation during development. Transgenic mice containing the human beta-globin locus, consisting of the linked embryonic (epsilon), fetal (gamma) and adult (beta) genes, have been used as a system to investigate the temporal switch from fetal to adult haemoglobin, as occurs in humans. Here we show that the human gamma-globin (HBG) genes in these mice behave as murine embryonic globin genes, revealing a limitation of the model and demonstrating that critical differences in the trans-acting milieu have arisen during mammalian evolution. We show that the expression of BCL11A, a repressor of human gamma-globin expression identified by genome-wide association studies, differs between mouse and human. Developmental silencing of the mouse embryonic globin and human gamma-globin genes fails to occur in mice in the absence of BCL11A. Thus, BCL11A is a critical mediator of species-divergent globin switching. By comparing the ontogeny of beta-globin gene regulation in mice and humans, we have shown that alterations in the expression of a trans-acting factor constitute a critical driver of gene expression changes during evolution.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Developmental , Globins/genetics , Nuclear Proteins/metabolism , Animals , Carrier Proteins/genetics , DNA-Binding Proteins , Embryo, Mammalian/metabolism , Evolution, Molecular , Fetus/metabolism , Gene Silencing , Hematopoiesis , Humans , Mice , Nuclear Proteins/genetics , Repressor Proteins , Species Specificity , beta-Globins/genetics , gamma-Globins/genetics
17.
Curr Opin Biotechnol ; 86: 103082, 2024 04.
Article in English | MEDLINE | ID: mdl-38428225

ABSTRACT

Monoclonal antibodies have revolutionized the treatment of human diseases, which has made them the fastest-growing class of therapeutics, with global sales expected to reach $346.6 billion USD by 2028. Advances in antibody engineering and development have led to the creation of increasingly sophisticated antibody-based therapeutics (e.g. bispecific antibodies and chimeric antigen receptor T cells). However, approaches for antibody discovery have remained comparatively grounded in conventional yet reliable in vitro assays. Breakthrough developments in high-throughput single B-cell sequencing and immunoglobulin proteomic serology, however, have enabled the identification of high-affinity antibodies directly from endogenous B cells or circulating immunoglobulin produced in vivo. Moreover, advances in artificial intelligence offer vast potential for antibody discovery and design with large-scale repertoire datasets positioned as the optimal source of training data for such applications. We highlight advances and recent trends in how these technologies are being applied to antibody repertoire analysis.


Subject(s)
Antibodies, Bispecific , Proteomics , Humans , Artificial Intelligence , Antibodies, Monoclonal
18.
Int J Biol Macromol ; 257(Pt 1): 128362, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38029898

ABSTRACT

N-glycosylation at the antibody variable domain has emerged as an important modification influencing antibody function. Despite its significance, information regarding its role and regulation remains limited. To address this gap, we comprehensively explored antibody structures housing N-glycosylation within the Protein Data Bank, yielding fresh insights into this intricate landscape. Our findings revealed that among 208 structures, N-glycosylation was more prevalent in human and mouse antibodies containing IGHV1-8 and IGHV2-2 germline genes, respectively. Moreover, our research highlights the potential for somatic hypermutation to introduce N-glycosylation sites by substituting polar residues (Ser or Thr) in germline variable genes with asparagine. Notably, our study underscores the prevalence of N-glycosylation in antiviral antibodies, especially anti-HIV. Besides antigen-antibody interaction, our findings suggest that N-glycosylation may impact antibody specificity, affinity, and avidity by influencing Fab dimer formation and complementary-determining region orientation. We also identified different glycan structures in HIV and SARS-CoV-2 antibody proteomic datasets, highlighting disparities from the N-glycan structures between PDB antibodies and biological repertoires further highlighting the complexity of N-glycosylation patterns. Our findings significantly enrich our understanding of the N-glycosylation's multifaceted characteristics within the antibody variable domain. Additionally, they underscore the pressing imperative for a more comprehensive characterization of its impact on antibody function.


Subject(s)
Antibodies, Viral , Proteomics , Humans , Mice , Animals , Glycosylation , Antibodies, Viral/metabolism , Polysaccharides/metabolism
19.
bioRxiv ; 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38545622

ABSTRACT

We used plasma IgG proteomics to study the molecular composition and temporal durability of polyclonal IgG antibodies triggered by ancestral SARS-CoV-2 infection, vaccination, or their combination ("hybrid immunity"). Infection, whether primary or post-vaccination, mainly triggered an anti-spike antibody response to the S2 domain, while vaccination predominantly induced anti-RBD antibodies. Immunological imprinting persisted after a secondary (hybrid) exposure, with >60% of the ensuing serological response originating from the initial antibodies generated during the first exposure. We highlight one instance where hybrid immunity arising from breakthrough infection resulted in a marked increase in the breadth and affinity of a highly abundant vaccination-elicited plasma IgG antibody, SC27. With an intrinsic binding affinity surpassing a theoretical maximum (K D < 5 pM), SC27 demonstrated potent neutralization of various SARS-CoV-2 variants and SARS-like zoonotic viruses (IC 50 ∼0.1-1.75 nM) and provided robust protection in vivo . Cryo-EM structural analysis unveiled that SC27 binds to the RBD class 1/4 epitope, with both VH and VL significantly contributing to the binding interface. These findings suggest that exceptionally broad and potent antibodies can be prevalent in plasma and can largely dictate the nature of serological neutralization. HIGHLIGHTS: ▪ Infection and vaccination elicit unique IgG antibody profiles at the molecular level▪ Immunological imprinting varies between infection (S2/NTD) and vaccination (RBD)▪ Hybrid immunity maintains the imprint of first infection or first vaccination▪ Hybrid immune IgG plasma mAbs have superior neutralization potency and breadth.

20.
EMBO J ; 28(6): 711-24, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19214191

ABSTRACT

Regulation of BCR signalling strength is crucial for B-cell development and function. Bright is a B-cell-restricted factor that complexes with Bruton's tyrosine kinase (Btk) and its substrate, transcription initiation factor-I (TFII-I), to activate immunoglobulin heavy chain gene transcription in the nucleus. Here we show that a palmitoylated pool of Bright is diverted to lipid rafts of resting B cells where it associates with signalosome components. After BCR ligation, Bright transiently interacts with sumoylation enzymes, blocks calcium flux and phosphorylation of Btk and TFII-I and is then discharged from lipid rafts as a Sumo-I-modified form. The resulting lipid raft concentration of Bright contributes to the signalling threshold of B cells, as their sensitivity to BCR stimulation decreases as the levels of Bright increase. Bright regulates signalling independent of its role in IgH transcription, as shown by specific dominant-negative titration of rafts-specific forms. This study identifies a BCR tuning mechanism in lipid rafts that is regulated by differential post-translational modification of a transcription factor with implications for B-cell tolerance and autoimmunity.


Subject(s)
Membrane Microdomains/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Antigens/metabolism , B-Lymphocytes/enzymology , DNA-Binding Proteins , Humans , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Lipoylation , Lymphocyte Activation , Membrane Microdomains/enzymology , Mice , Mutation/genetics , Oncogenes , Phosphorylation , Protein Binding , Protein Transport , Protein-Tyrosine Kinases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors, TFII/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL