Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell Host Microbe ; 32(2): 261-275.e4, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38307019

ABSTRACT

Hemagglutinins (HAs) from human influenza viruses descend from avian progenitors that bind α2-3-linked sialosides and must adapt to glycans with α2-6-linked sialic acids on human airway cells to transmit within the human population. Since their introduction during the 1968 pandemic, H3N2 viruses have evolved over the past five decades to preferentially recognize human α2-6-sialoside receptors that are elongated through addition of poly-LacNAc. We show that more recent H3N2 viruses now make increasingly complex interactions with elongated receptors while continuously selecting for strains maintaining this phenotype. This change in receptor engagement is accompanied by an extension of the traditional receptor-binding site to include residues in key antigenic sites on the surface of HA trimers. These results help explain the propensity for selection of antigenic variants, leading to vaccine mismatching, when H3N2 viruses are propagated in chicken eggs or cells that do not contain such receptors.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza, Human , Animals , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/metabolism , Receptors, Virus/chemistry , Sialic Acids/metabolism , Polysaccharides/metabolism , Chickens , Hemagglutinin Glycoproteins, Influenza Virus
2.
JACS Au ; 3(3): 868-878, 2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37006776

ABSTRACT

Influenza virus infection remains a threat to human health since viral hemagglutinins are constantly drifting, escaping infection and vaccine-induced antibody responses. Viral hemagglutinins from different viruses display variability in glycan recognition. In this context, recent H3N2 viruses have specificity for α2,6 sialylated branched N-glycans with at least three N-acetyllactosamine units (tri-LacNAc). In this work, we combined glycan arrays and tissue binding analyses with nuclear magnetic resonance experiments to characterize the glycan specificity of a family of H1 variants, including the one responsible for the 2009 pandemic outbreak. We also analyzed one engineered H6N1 mutant to understand if the preference for tri-LacNAc motifs could be a general trend in human-type receptor-adapted viruses. In addition, we developed a new NMR approach to perform competition experiments between glycans with similar compositions and different lengths. Our results point out that pandemic H1 viruses differ from previous seasonal H1 viruses by a strict preference for a minimum of di-LacNAc structural motifs.

3.
J Immunol ; 206(10): 2290-2300, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33911007

ABSTRACT

Siglec-8 is an inhibitory receptor expressed on eosinophils and mast cells. In this study, we took advantage of a novel Siglec-8 transgenic mouse model to assess the impact of modulating IgE-dependent mast cell degranulation and anaphylaxis using a liposomal platform to display an allergen with or without a synthetic glycan ligand for Siglec-8 (Sig8L). The hypothesis is that recruitment of Siglec-8 to the IgE-FcεRI receptor complex will inhibit allergen-induced mast cell degranulation. Codisplay of both allergen and Sig8L on liposomes profoundly suppresses IgE-mediated degranulation of mouse bone marrow-derived mast cells or rat basophilic leukemia cells expressing Siglec-8. In contrast, liposomes displaying only Sig8L have no significant suppression of antigenic liposome-induced degranulation, demonstrating that the inhibitory activity by Siglec-8 occurs only when Ag and Sig8L are on the same particle. In mouse models of anaphylaxis, display of Sig8L on antigenic liposomes completely suppresses IgE-mediated anaphylaxis in transgenic mice with mast cells expressing Siglec-8 but has no protection in mice that do not express Siglec-8. Furthermore, mice protected from anaphylaxis remain desensitized to subsequent allergen challenge because of loss of Ag-specific IgE from the cell surface and accelerated clearance of IgE from the blood. Thus, although expression of human Siglec-8 on murine mast cells does not by itself modulate IgE-FcεRI-mediated cell activation, the enforced recruitment of Siglec-8 to the FcεRI receptor by Sig8L-decorated antigenic liposomes results in inhibition of degranulation and desensitization to subsequent Ag exposure.


Subject(s)
Allergens/administration & dosage , Anaphylaxis/drug therapy , Anaphylaxis/genetics , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Desensitization, Immunologic/methods , Drug Delivery Systems/methods , Immunoglobulin E/metabolism , Lectins/metabolism , Mast Cells/immunology , Nanoparticles/chemistry , Polysaccharides/administration & dosage , Receptors, IgE/metabolism , Anaphylaxis/immunology , Animals , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Cell Degranulation/drug effects , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Line, Tumor , Disease Models, Animal , Humans , Lectins/genetics , Ligands , Liposomes , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polysaccharides/metabolism , Rats , Receptors, IgE/genetics , Treatment Outcome
4.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article in English | MEDLINE | ID: mdl-33893239

ABSTRACT

Siglecs are a family of sialic acid-binding receptors expressed by cells of the immune system and a few other cell types capable of modulating immune cell functions upon recognition of sialoglycan ligands. While human Siglecs primarily bind to sialic acid residues on diverse types of glycoproteins and glycolipids that constitute the sialome, their fine binding specificities for elaborated complex glycan structures and the contribution of the glycoconjugate and protein context for recognition of sialoglycans at the cell surface are not fully elucidated. Here, we generated a library of isogenic human HEK293 cells with combinatorial loss/gain of individual sialyltransferase genes and the introduction of sulfotransferases for display of the human sialome and to dissect Siglec interactions in the natural context of glycoconjugates at the cell surface. We found that Siglec-4/7/15 all have distinct binding preferences for sialylated GalNAc-type O-glycans but exhibit selectivity for patterns of O-glycans as presented on distinct protein sequences. We discovered that the sulfotransferase CHST1 drives sialoglycan binding of Siglec-3/8/7/15 and that sulfation can impact the preferences for binding to O-glycan patterns. In particular, the branched Neu5Acα2-3(6-O-sulfo)Galß1-4GlcNAc (6'-Su-SLacNAc) epitope was discovered as the binding epitope for Siglec-3 (CD33) implicated in late-onset Alzheimer's disease. The cell-based display of the human sialome provides a versatile discovery platform that enables dissection of the genetic and biosynthetic basis for the Siglec glycan interactome and other sialic acid-binding proteins.


Subject(s)
Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Tissue Array Analysis/methods , Gene Knockout Techniques , HEK293 Cells , Humans , Mucin-1 , Polysaccharides/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism
5.
Nat Commun ; 11(1): 1233, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144244

ABSTRACT

Antigenic drift of influenza virus hemagglutinin (HA) is enabled by facile evolvability. However, HA antigenic site B, which has become immunodominant in recent human H3N2 influenza viruses, is also evolutionarily constrained by its involvement in receptor binding. Here, we employ deep mutational scanning to probe the local fitness landscape of HA antigenic site B in six different human H3N2 strains spanning from 1968 to 2016. We observe that the fitness landscape of HA antigenic site B can be very different between strains. Sequence variants that exhibit high fitness in one strain can be deleterious in another, indicating that the evolutionary constraints of antigenic site B have changed over time. Structural analysis suggests that the local fitness landscape of antigenic site B can be reshaped by natural mutations via modulation of the receptor-binding mode. Overall, these findings elucidate how influenza virus continues to explore new antigenic space despite strong functional constraints.


Subject(s)
Antigens, Viral/genetics , Evolution, Molecular , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Receptors, Cell Surface/metabolism , Animals , Antigens, Viral/immunology , Antigens, Viral/metabolism , Binding Sites/genetics , Crystallography, X-Ray , DNA Mutational Analysis , Dogs , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/metabolism , Madin Darby Canine Kidney Cells , Mutation , Protein Domains/genetics , Protein Domains/immunology , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
6.
J Am Chem Soc ; 141(36): 14032-14037, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31460762

ABSTRACT

The Siglec family of cell surface receptors have emerged as attractive targets for cell-directed therapies due to their restricted expression on immune cells, endocytic properties, and ability to modulate receptor signaling. Human Siglec-8, for instance, has been identified as a therapeutic target for the treatment of eosinophil and mast cell disorders. A promising strategy to target Siglecs involves the use of liposomal nanoparticles with a multivalent display of Siglec ligands. A key challenge for this approach is the identification of a high affinity ligand for the target Siglec. Here, we report the development of a ligand of Siglec-8 and its closest murine functional orthologue Siglec-F that is capable of targeting liposomes to cells expressing Siglec-8 or -F. A glycan microarray library of synthetic 9-N-sulfonyl sialoside analogues was screened to identify potential lead compounds. The best ligand, 9-N-(2-naphthyl-sulfonyl)-Neu5Acα2-3-[6-O-sulfo]-Galß1-4GlcNAc (6'-O-sulfo NSANeu5Ac) combined the lead 2-naphthyl sulfonyl C-9 substituent with the preferred sulfated scaffold. The ligand 6'-O-sulfo NSANeu5Ac was conjugated to lipids for display on liposomes to evaluate targeted delivery to cells. Targeted liposomes showed strong in vitro binding/uptake and selectivity to cells expressing Siglec-8 or -F and, when administered to mice, exhibit in vivo targeting to Siglec-F+ eosinophils.


Subject(s)
Antigens, Differentiation, Myelomonocytic/metabolism , B-Lymphocytes/drug effects , Lectins/antagonists & inhibitors , Sialic Acids/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , Animals , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/metabolism , CHO Cells , Cricetulus , Humans , Lectins/metabolism , Liposomes/chemistry , Liposomes/metabolism , Mice , Molecular Conformation , Sialic Acid Binding Immunoglobulin-like Lectins , Sialic Acids/chemistry , Sulfonamides/chemistry , T-Lymphocytes/metabolism
7.
Cell Host Microbe ; 25(6): 836-844.e5, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31151913

ABSTRACT

Egg-based seasonal influenza vaccines are the major preventive countermeasure against influenza virus. However, their effectiveness can be compromised when antigenic changes arise from egg-adaptive mutations on influenza hemagglutinin (HA). The L194P mutation is commonly observed in egg-based H3N2 vaccine seed strains and significantly alters HA antigenicity. An approach to prevent L194P would therefore be beneficial. We show that emergence of L194P during egg passaging can be impeded by preexistence of a G186V mutation, revealing strong incompatibility between these mutations. X-ray structures illustrate that individual G186V and L194P mutations have opposing effects on the HA receptor-binding site (RBS), and when both G186V and L194P are present, the RBS is severely disrupted. Importantly, wild-type HA antigenicity is maintained with G186V, but not L194P. Our results demonstrate that these epistatic interactions can be used to prevent the emergence of mutations that adversely alter antigenicity during egg adaptation.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Mutation, Missense , Adaptation, Biological , Animals , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Binding Sites , Chick Embryo , Crystallography, X-Ray , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Protein Conformation , Technology, Pharmaceutical/methods , Virus Cultivation/methods
8.
ACS Chem Biol ; 14(4): 644-654, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30835424

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease that primarily affects the synovial joints and can lead to bone erosion and cartilage damage. One hallmark of RA is anticitrullinated protein autoantibodies (ACPA) and memory citrulline-specific B-cells, which have been implicated in RA pathogenesis. While depletion of B-cells with Rituximab improves clinical responses in RA patients, this treatment strategy leaves patients susceptible to infections. Here we use of Siglec-engaging Tolerance-inducing Antigenic Liposomes (STALs) to selectively target the citrulline-specific B-cells. ACPA production from purified human RA patients' B-cells in vitro was achieved through a set of stimulation conditions, which includes the following: BAFF, anti-CD40, IL-21, and LPS. In vivo generation of citrulline specific B-cells and ACPA production was accomplished by antigenic liposomes consisting of monophosphoryl lipid A (MPLA) and a cyclic citrullinated peptide (CCP) administered to SJL/J mice. We show that STALs that codisplay a high affinity CD22 glycan ligand and synthetic citrullinated antigen (CCP STALs) can prevent ACPA production from RA patients' memory B-cells in vitro. These CCP STALs were also effective in inducing tolerance to citrullinated antigens in SJL/J mice. The results demonstrate that tolerization of the B-cells responsible for ACPA can be achieved by exploiting the inhibitory receptor CD22 with high-affinity glycan ligands. Such a treatment strategy could be beneficial in the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , B-Lymphocytes/immunology , Sialic Acid Binding Ig-like Lectin 2/metabolism , Animals , Case-Control Studies , Citrulline/metabolism , Humans , Immunologic Memory , Mice
9.
J Clin Invest ; 129(3): 1387-1401, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30645205

ABSTRACT

Allergen immunotherapy for patients with allergies begins with weekly escalating doses of allergen under medical supervision to monitor and treat IgE mast cell-mediated anaphylaxis. There is currently no treatment to safely desensitize mast cells to enable robust allergen immunotherapy with therapeutic levels of allergen. Here, we demonstrated that liposomal nanoparticles bearing an allergen and a high-affinity glycan ligand of the inhibitory receptor CD33 profoundly suppressed IgE-mediated activation of mast cells, prevented anaphylaxis in Tg mice with mast cells expressing human CD33, and desensitized mice to subsequent allergen challenge for several days. We showed that high levels of CD33 were consistently expressed on human skin mast cells and that the antigenic liposomes with CD33 ligand prevented IgE-mediated bronchoconstriction in slices of human lung. The results demonstrated the potential of exploiting CD33 to desensitize mast cells to provide a therapeutic window for administering allergen immunotherapy without triggering anaphylaxis.


Subject(s)
Allergens/immunology , Anaphylaxis/prevention & control , Desensitization, Immunologic , Immunoglobulin E/immunology , Mast Cells/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Anaphylaxis/genetics , Anaphylaxis/immunology , Anaphylaxis/pathology , Animals , Bronchoconstriction/genetics , Bronchoconstriction/immunology , Humans , Immunoglobulin E/genetics , Mast Cells/pathology , Mice , Mice, Transgenic , Sialic Acid Binding Ig-like Lectin 3/genetics
10.
Cell Chem Biol ; 26(1): 131-136.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30393066

ABSTRACT

Macrophages (MØs) expressing the endocytic sialic acid-binding immunoglobulin-like lectin 1 (siglec-1, CD169, sialoadhesin) are known to be adept at antigen capture-primarily due to their strategic location within lymphatic tissues. Antigen concentrated in these cells can be harnessed to induce potent anti-tumor/anti-pathogen cytotoxic (CD8+) T cell responses. Here, we describe a chemical platform that exploits the CD169-mediated antigen capture pathway for biased priming of antigen-specific CD4+ or CD8+ T cells in vivo. In the absence of a toll-like receptor (TLR) agonist, antigen delivery through CD169 produced robust CD4+ T cell priming only. However, simultaneous treatment with targeted antigen and a TLR7 agonist induced CD8+ T cell priming, with concomitant suppression of the CD4+ T cell response. We exploited these observations to manipulate the activation ratio of CD4+/CD8+ T cells in the same animal. These findings represent a unique chemical strategy for targeting CD169+ macrophages to modulate antigen-specific T cell immunity.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Macrophages/immunology , Sialic Acid Binding Ig-like Lectin 1/immunology , Animals , Cells, Cultured , Humans , Male , Mice , Sialic Acid Binding Ig-like Lectin 1/genetics
11.
Nat Commun ; 9(1): 1264, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593268

ABSTRACT

The hemagglutinin (HA) receptor-binding site (RBS) in human influenza A viruses is critical for attachment to host cells, which imposes a functional constraint on its natural evolution. On the other hand, being part of the major antigenic sites, the HA RBS of human H3N2 viruses needs to constantly mutate to evade the immune system. From large-scale mutagenesis experiments, we here show that several of the natural RBS substitutions become integrated into an extensive epistatic network that prevents substitution reversion. X-ray structural analysis reveals the mechanistic consequences as well as changes in the mode of receptor binding. Further studies are necessary to elucidate whether such entrenchment limits future options for immune escape or adversely affect long-term viral fitness.


Subject(s)
Binding Sites , Epistasis, Genetic , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza, Human/virology , Mutation , Receptors, Virus/chemistry , Antigens, Viral/chemistry , HEK293 Cells , Humans , Hydrogen Bonding , Immune System , Influenza A Virus, H3N2 Subtype/genetics , Oligosaccharides/chemistry , Protein Binding
12.
J Am Chem Soc ; 140(10): 3592-3602, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29446631

ABSTRACT

Metabolic labeling of glycans with bioorthogonal reporters has been widely used for glycan imaging and glycoproteomic profiling. One of the intrinsic limitations of metabolic glycan labeling is the lack of cell-type selectivity. The recently developed liposome-assisted bioorthogonal reporter (LABOR) strategy provides a promising means to overcome this limitation, but the mechanism of LABOR has not been investigated in detail. In this work, we performed a mechanistic study on LABOR and explored its multiplexing capability. Our studies support an endocytosis-salvage mechanism. The ligand-targeted liposomes encapsulating azidosugars are internalized into the endosome via the receptor-mediated endocytosis. Unlike the conventional drug delivery, LABOR does not rely on the endosomal escape pathways. Rather, the liposomes are allowed to enter the lysosome, inside which the azidosugars are released from the liposomes. The released azidosugars then intercept the salvage pathways of monosaccharides and get transported into the cytosol by lysosomal sugar transporters. Based on this mechanism, we expanded the scope of LABOR by evaluating a series of ligand-receptor pairs for targeting sialoglycans in various cell types. Different ligand types including small molecules, antibodies, aptamers, and peptides could be easily implemented into LABOR. Finally, we demonstrated that the sialoglycans in two distinct cell populations in a co-cultured system could be selectively labeled with two distinct chemical reporters by performing a multiplexed LABOR labeling.


Subject(s)
Polysaccharides/chemistry , HeLa Cells , Humans , Liposomes/chemistry , Liposomes/metabolism , Polysaccharides/metabolism
13.
J Immunol ; 200(3): 949-956, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29288201

ABSTRACT

CD22 and sialic acid-binding Ig-like lectin (Siglec)-G are members of the Siglec family of inhibitory coreceptors expressed on B cells that participate in enforcement of peripheral B cell tolerance. We have shown previously that when a BCR engages its cognate Ag on a cell surface that also expresses Siglec ligands, B cell Siglecs are recruited to the immunological synapse, resulting in suppression of BCR signaling and B cell apoptosis. Because all cells display sialic acids, and CD22 and Siglec-G have distinct, yet overlapping, specificities for sialic acid-containing glycan ligands, any cell could, in principle, invoke this tolerogenic mechanism for cell surface Ags. However, we show in this article that C57BL/6J mouse RBCs are essentially devoid of CD22 and Siglec-G ligands. As a consequence, RBCs that display a cell surface Ag, membrane-bound hen egg lysozyme, strongly activate Ag-specific B cells. We reasoned that de novo introduction of CD22 ligands in RBCs should abolish B cell activation toward its cognate Ag on the surface of RBCs. Accordingly, we used a glyco-engineering approach wherein synthetic CD22 ligands linked to lipids are inserted into the membrane of RBCs. Indeed, insertion of CD22 ligands into the RBC cell surface strongly inhibited B cell activation, cytokine secretion, and proliferation. These results demonstrate that the lack of Siglec ligands on the surface of murine RBCs permits B cell responses to erythrocyte Ags and show that Siglec-mediated B cell tolerance is restricted to cell types that express glycan ligands for the B cell Siglecs.


Subject(s)
Antigens, Surface/immunology , B-Lymphocytes/immunology , Erythrocytes/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Animals , Apoptosis/immunology , B-Lymphocytes/metabolism , CHO Cells , Cell Line , Cricetulus , Mice , Mice, Inbred C57BL , Mice, Knockout , Muramidase/genetics , Muramidase/immunology , Muramidase/metabolism , Receptors, Antigen, B-Cell/immunology , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acids/immunology
14.
PLoS Pathog ; 13(10): e1006682, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29059230

ABSTRACT

The effectiveness of the annual influenza vaccine has declined in recent years, especially for the H3N2 component, and is a concern for global public health. A major cause for this lack in effectiveness has been attributed to the egg-based vaccine production process. Substitutions on the hemagglutinin glycoprotein (HA) often arise during virus passaging that change its antigenicity and hence vaccine effectiveness. Here, we characterize the effect of a prevalent substitution, L194P, in egg-passaged H3N2 viruses. X-ray structural analysis reveals that this substitution surprisingly increases the mobility of the 190-helix and neighboring regions in antigenic site B, which forms one side of the receptor binding site (RBS) and is immunodominant in recent human H3N2 viruses. Importantly, the L194P substitution decreases binding and neutralization by an RBS-targeted broadly neutralizing antibody by three orders of magnitude and significantly changes the HA antigenicity as measured by binding of human serum antibodies. The receptor binding mode and specificity are also altered to adapt to avian receptors during egg passaging. Overall, these findings help explain the low effectiveness of the seasonal vaccine against H3N2 viruses, and suggest that alternative approaches should be accelerated for producing influenza vaccines as well as isolating clinical isolates.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Amino Acid Substitution , Antigens, Viral/chemistry , Antigens, Viral/immunology , Humans
16.
Cell Host Microbe ; 21(6): 742-753.e8, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-28618270

ABSTRACT

Influenza A virus hemagglutinin (HA) initiates viral entry by engaging host receptor sialylated glycans via its receptor-binding site (RBS). The amino acid sequence of the RBS naturally varies across avian and human influenza virus subtypes and is also evolvable. However, functional sequence diversity in the RBS has not been fully explored. Here, we performed a large-scale mutational analysis of the RBS of A/WSN/33 (H1N1) and A/Hong Kong/1/1968 (H3N2) HAs. Many replication-competent mutants not yet observed in nature were identified, including some that could escape from an RBS-targeted broadly neutralizing antibody. This functional sequence diversity is made possible by pervasive epistasis in the RBS 220-loop and can be buffered by avidity in viral receptor binding. Overall, our study reveals that the HA RBS can accommodate a much greater range of sequence diversity than previously thought, which has significant implications for the complex evolutionary interrelationships between receptor specificity and immune escape.


Subject(s)
Binding Sites/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immune Evasion , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Receptors, Virus/genetics , Amino Acid Sequence , Antibodies, Viral/immunology , Antigenic Variation/genetics , Antigenic Variation/immunology , Antigens, Viral/genetics , Base Sequence , Binding Sites/immunology , Crystallography, X-Ray/instrumentation , Evolution, Molecular , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/virology , Mutation , Protein Conformation , Receptors, Virus/immunology , Sequence Analysis , Virus Internalization
17.
Glycobiology ; 27(7): 657-668, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28369504

ABSTRACT

Siglecs are transmembrane sialoglycan binding proteins, most of which are expressed on leukocyte subsets and have inhibitory motifs that translate cell surface ligation into immune suppression. In humans, Siglec-8 on eosinophils, mast cells and basophils and Siglec-9 on neutrophils, monocytes and some T-cells, mediate immune cell death, inhibition of immune mediator release and/or enhancement of anti-inflammatory mediator release. Endogenous sialoglycan ligands in tissues, mostly uncharacterized, engage siglecs on leukocytes to inhibit inflammation. Glycan array analyses demonstrated that Siglec-8, Siglec-9 and their mouse counterparts Siglec-F and Siglec-E (respectively) have distinct glycan binding specificities, with Siglec-8 more structurally restricted. Since siglecs are involved in lung inflammation, we studied Siglec-8 and Siglec-9 ligands in human lungs and airways. Siglec-8 ligands are in tracheal submucosal glands and cartilage but not airway epithelium or connective tissues, whereas Siglec-9 ligands are broadly distributed. Mouse airways do not have Siglec-8 ligands, whereas Siglec-9 ligands are on airways of both species. Extraction of human airways and lung followed by electrophoretic resolution and siglec blotting revealed Siglec-8 ligands in extracts of human trachea and cultured tracheal gland cells, but not parenchyma or cultured airway epithelial cells whereas Siglec-9 ligands were extracted from all airway and lung tissues and cells tested. Siglec-8 and Siglec-9 ligands in airways appear to be high molecular weight O-linked sialoglycoproteins. These data reveal differential glycan specificities of Siglec-8, Siglec-9 and their mouse counterparts Siglec-F and Siglec-E, and the tissue distributions and molecular characteristics of Siglec-8 and Siglec-9 sialoglycan ligands on human airways and lungs.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Lectins/metabolism , Respiratory Mucosa/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Adult , Antigens, CD/chemistry , Antigens, Differentiation, B-Lymphocyte/chemistry , Cells, Cultured , Female , Humans , Lectins/chemistry , Ligands , Lung/cytology , Lung/metabolism , Male , Middle Aged , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Protein Binding , Sialic Acid Binding Immunoglobulin-like Lectins/chemistry , Trachea/cytology , Trachea/metabolism
18.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28404848

ABSTRACT

Influenza A H15 viruses are members of a subgroup (H7-H10-H15) of group 2 hemagglutinin (HA) subtypes that include H7N9 and H10N8 viruses that were isolated from humans during 2013. The isolation of avian H15 viruses is, however, quite rare and, until recently, geographically restricted to wild shorebirds and waterfowl in Australia. The HAs of H15 viruses contain an insertion in the 150-loop (loop beginning at position 150) of the receptor-binding site common to this subgroup and a unique insertion in the 260-loop compared to any other subtype. Here, we show that the H15 HA has a high preference for avian receptor analogs by glycan array analyses. The H15 HA crystal structure reveals that it is structurally closest to H7N9 HA, but the head domain of the H15 trimer is wider than all other HAs due to a tilt and opening of the HA1 subunits of the head domain. The extended 150-loop of the H15 HA retains the conserved conformation as in H7 and H10 HAs. Furthermore, the elongated 260-loop increases the exposed HA surface and can contribute to antigenic variation in H15 HAs. Since avian-origin H15 HA viruses have been shown to cause enhanced disease in mammalian models, further characterization and immune surveillance of H15 viruses are warranted.IMPORTANCE In the last 2 decades, an apparent increase has been reported for cases of human infection by emerging avian influenza A virus subtypes, including H7N9 and H10N8 viruses isolated during 2013. H15 is the other member of the subgroup of influenza A virus group 2 hemagglutinins (HAs) that also include H7 and H10. H15 viruses have been restricted to Australia, but recent isolation of H15 viruses in western Siberia suggests that they could be spread more globally via the avian flyways that converge and emanate from this region. Here we report on characterization of the three-dimensional structure and receptor specificity of the H15 hemagglutinin, revealing distinct features and specificities that can aid in global surveillance of such viruses for potential spread and emerging threat to the human population.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A virus/chemistry , Animals , Australia , Binding Sites , Birds/virology , Crystallography, X-Ray , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H7N9 Subtype , Influenza A virus/classification , Influenza A virus/metabolism , Influenza, Human/virology , Models, Molecular , Polysaccharides , Protein Binding , Protein Conformation , Receptors, Virus/metabolism , Siberia
19.
Chembiochem ; 18(13): 1226-1233, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28231415

ABSTRACT

Unwanted antibody responses significantly impact human health, and current options for treating deleterious antibody responses largely rely on broad immunosuppressants that can compromise overall immunity. A desirable alternative is to induce antigen-specific immune tolerance. We have shown that co-presentation of antigen and ligands of B cell sialic acid-binding immunoglobulin-like lectins (Siglecs) on a liposomal nanoparticle induces antigen-specific tolerance. Although Siglec-engaging tolerance-inducing antigenic liposomes (STALs) induce robust B cell tolerance in naïve mice, the full potential of STALs requires long-term tolerance induction and suppression of an ongoing immune response. We hypothesized that STALs encapsulated with rapamycin (RAPA), an immunomodulator, could improve the efficacy of STALs and potentially enable their use in the context of immunological memory. Here, we showed that formulation of STALs with RAPA produced enhanced tolerance induction in naïve mice compared to STALs without RAPA but had minimal impact on inducing tolerance in previously sensitized mice. These findings indicate that the addition of immunomodulators to STALs could be beneficial in tolerance induction and support future development of STALs for the treatment of allergy and autoimmune diseases.


Subject(s)
Anti-Allergic Agents/pharmacology , Egg Hypersensitivity/therapy , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Liposomes/pharmacology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sirolimus/pharmacology , Animals , Anti-Allergic Agents/immunology , Antibodies/blood , Antibodies/drug effects , Antigens/immunology , Antigens/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Drug Compounding , Egg Hypersensitivity/genetics , Egg Hypersensitivity/immunology , Gene Expression , Humans , Immunosuppressive Agents/chemistry , Ligands , Liposomes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Ovalbumin , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sirolimus/chemistry
20.
Chem Sci ; 7(8): 4990-5001, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-30155149

ABSTRACT

P. aeruginosa causes a substantial number of nosocomial infections and is the leading cause of death of cystic fibrosis patients. This Gram-negative bacterium is highly resistant against antibiotics and further protects itself by forming a biofilm. Moreover, a high genomic variability among clinical isolates complicates therapy. Its lectin LecB is a virulence factor and necessary for adhesion and biofilm formation. We analyzed the sequence of LecB variants in a library of clinical isolates and demonstrate that it can serve as a marker for strain family classification. LecB from the highly virulent model strain PA14 presents 13% sequence divergence with LecB from the well characterized PAO1 strain. These differences might result in differing ligand binding specificities and ultimately in reduced efficacy of drugs directed towards LecB. Despite several amino acid variations at the carbohydrate binding site, glycan array analysis showed a comparable binding pattern for both variants. A common high affinity ligand could be identified and after its chemoenzymatic synthesis verified in a competitive binding assay: an N-glycan presenting two blood group O epitopes (H-type 2 antigen). Molecular modeling of the complex suggests a bivalent interaction of the ligand with the LecB tetramer by bridging two separate binding sites. This binding rationalizes the strong avidity (35 nM) of LecBPA14 to this human fucosylated N-glycan. Biochemical evaluation of a panel of glycan ligands revealed that LecBPA14 demonstrated higher glycan affinity compared to LecBPAO1 including the extraordinarily potent affinity of 70 nM towards the monovalent human antigen Lewisa. The structural basis of this unusual high affinity ligand binding for lectins was rationalized by solving the protein crystal structures of LecBPA14 with several glycans.

SELECTION OF CITATIONS
SEARCH DETAIL
...