Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
ACS Chem Biol ; 19(2): 483-496, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38321945

RESUMEN

Human sialic-acid-binding immunoglobulin-like lectin-9 (Siglec-9) is a glycoimmune checkpoint receptor expressed on several immune cells. Binding of Siglec-9 to sialic acid containing glycans (sialoglycans) is well documented to modulate its functions as an inhibitory receptor. Here, we first assigned the amino acid backbone of the Siglec-9 V-set domain (Siglec-9d1), using well-established triple resonance three-dimensional nuclear magnetic resonance (NMR) methods. Then, we combined solution NMR and molecular dynamic simulation methods to decipher the molecular details of the interaction of Siglec-9 with the natural ligands α2,3 and α2,6 sialyl lactosamines (SLN), sialyl Lewis X (sLeX), and 6-O sulfated sLeX and with two synthetically modified sialoglycans that bind with high affinity. As expected, Neu5Ac is accommodated between the F and G ß-strands at the canonical sialic acid binding site. Addition of a heteroaromatic scaffold 9N-5-(2-methylthiazol-4-yl)thiophene sulfonamide (MTTS) at the C9 position of Neu5Ac generates new interactions with the hydrophobic residues located at the G-G' loop and the N-terminal region of Siglec-9. Similarly, the addition of the aromatic substituent (5-N-(1-benzhydryl-1H-1,2,3-triazol-4-yl)methyl (BTC)) at the C5 position of Neu5Ac stabilizes the conformation of the long and flexible B'-C loop present in Siglec-9. These results expose the underlying mechanism responsible for the enhanced affinity and specificity for Siglec-9 for these two modified sialoglycans and sheds light on the rational design of the next generation of modified sialoglycans targeting Siglec-9.


Asunto(s)
Simulación de Dinámica Molecular , Ácido N-Acetilneuramínico , Humanos , Antígenos de Diferenciación Mielomonocítica/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Polisacáridos/metabolismo , Espectroscopía de Resonancia Magnética , Ligandos
2.
Cell Host Microbe ; 32(2): 261-275.e4, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38307019

RESUMEN

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.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A , Gripe Humana , Animales , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Receptores Virales/química , Ácidos Siálicos/metabolismo , Polisacáridos/metabolismo , Pollos , Glicoproteínas Hemaglutininas del Virus de la Influenza
3.
JACS Au ; 3(3): 868-878, 2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37006776

RESUMEN

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.

4.
JACS Au ; 3(1): 204-215, 2023 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-36711084

RESUMEN

Human sialic acid binding immunoglobulin-like lectin-8 (Siglec-8) is an inhibitory receptor that triggers eosinophil apoptosis and can inhibit mast cell degranulation when engaged by specific monoclonal antibodies (mAbs) or sialylated ligands. Thus, Siglec-8 has emerged as a critical negative regulator of inflammatory responses in diverse diseases, such as allergic airway inflammation. Herein, we have deciphered the molecular recognition features of the interaction of Siglec-8 with the mAb lirentelimab (2C4, under clinical development) and with a sialoside mimetic with the potential to suppress mast cell degranulation. The three-dimensional structure of Siglec-8 and the fragment antigen binding (Fab) portion of the anti-Siglec-8 mAb 2C4, solved by X-ray crystallography, reveal that 2C4 binds close to the carbohydrate recognition domain (V-type Ig domain) on Siglec-8. We have also deduced the binding mode of a high-affinity analogue of its sialic acid ligand (9-N-napthylsufonimide-Neu5Ac, NSANeuAc) using a combination of NMR spectroscopy and X-ray crystallography. Our results show that the sialoside ring of NSANeuAc binds to the canonical sialyl binding pocket of the Siglec receptor family and that the high affinity arises from the accommodation of the NSA aromatic group in a nearby hydrophobic patch formed by the N-terminal tail and the unique G-G' loop. The results reveal the basis for the observed high affinity of this ligand and provide clues for the rational design of the next generation of Siglec-8 inhibitors. Additionally, the specific interactions between Siglec-8 and the N-linked glycans present on the high-affinity receptor FcεRIα have also been explored by NMR.

5.
J Allergy Clin Immunol ; 150(6): 1476-1485.e4, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35839842

RESUMEN

BACKGROUND: Circulating IgE and subsequent severe allergic reactions to peanut are sustained and propagated by recall of peanut allergen-specific memory B cells. OBJECTIVES: This study aimed to determine whether targeting mouse and human CD22 on peanut-specific memory B cells induces tolerance to peanut allergens. METHODS: Siglec-engaging tolerance-inducing antigenic liposomes (STALs) codisplaying peanut allergens (Ara h 1, Ara h 2, or Ara h 3) and high-affinity CD22 ligand (CD22L-STALs) were employed in various mouse models (BALB/cJ, C57BL/6, human CD22 transgenic, and NSG) of peanut allergy. To investigate memory B cells, a conferred memory model was used in which splenocytes from peanut-sensitized mice were transferred into naive animals. Reconstituted mice received either CD22L-STALs or an immunogenic liposome control, followed by a peanut allergen boost and later a challenge with individual peanut allergens. To assess the effects of CD22L-STALs on human B cells, PBMCs were injected into NSG mice, followed by administration of human CD22L-STALs (hCD22L-STALs) and later a whole peanut extract boost. Blood was collected to quantify WPE- and Ara h 1-, 2-, and 3-specific immunoglobulins. RESULTS: Mouse CD22L-STALs (mCD22L-STALs) significantly suppressed systemic memory to Ara h 1, Ara h 2, and Ara h 3 in BALB/cJ and C57BL/6 mice, as demonstrated by reduced allergen-specific IgE, IgG1, and anaphylaxis on challenge. Importantly, 2 doses of mCD22L-STALs led to prolonged tolerance for at least 3 months. hCD22L-STALs displayed similar suppression in mice expressing human CD22 on B cells. Finally, human B cells were tolerized in vivo in NSG mice by hCD22L-STALs. CONCLUSIONS: Antigen-specific exploitation of CD22 on memory B cells can induce systemic immune tolerance.


Asunto(s)
Alérgenos , Arachis , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Células B de Memoria , Tolerancia Inmunológica , Lectina 2 Similar a Ig de Unión al Ácido Siálico
6.
ACS Chem Biol ; 17(2): 292-298, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35020348

RESUMEN

Extrahepatic targeted delivery of oligonucleotides, such as small interfering RNA (siRNA) and antisense oligonucleotides (ASOs), is an attractive technology for the development of nucleic acid-based medicines. To target CD22-expressing B cells, several drug platforms have shown promise, including antibodies, antibody-drug conjugates, and nanoparticles, but to date CD22-targeted delivery of oligonucleotide therapeutics has not been reported. Here we report the uptake and enhancement of siRNA gene expression knockdown in CD22-expressing B cells using a chemically stabilized and modified CD22 glycan ligand-conjugated siRNA. This finding has the potential to broaden the use of siRNA technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of siRNAs to B cell lymphomas.


Asunto(s)
Oligonucleótidos Antisentido , Polisacáridos , Técnicas de Silenciamiento del Gen , Ligandos , Polisacáridos/metabolismo , ARN Interferente Pequeño/genética
7.
Nat Chem Biol ; 17(7): 806-816, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958792

RESUMEN

The central dogma of biology does not allow for the study of glycans using DNA sequencing. We report a liquid glycan array (LiGA) platform comprising a library of DNA 'barcoded' M13 virions that display 30-1,500 copies of glycans per phage. A LiGA is synthesized by acylation of the phage pVIII protein with a dibenzocyclooctyne, followed by ligation of azido-modified glycans. Pulldown of the LiGA with lectins followed by deep sequencing of the barcodes in the bound phage decodes the optimal structure and density of the recognized glycans. The LiGA is target agnostic and can measure the glycan-binding profile of lectins, such as CD22, on cells in vitro and immune cells in a live mouse. From a mixture of multivalent glycan probes, LiGAs identify the glycoconjugates with optimal avidity necessary for binding to lectins on living cells in vitro and in vivo.


Asunto(s)
Bacteriófago M13/química , Análisis por Micromatrices , Polisacáridos/química , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteriófago M13/genética , Bacteriófago M13/metabolismo , Ratones , Polisacáridos/genética , Polisacáridos/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33893239

RESUMEN

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.


Asunto(s)
Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Análisis de Matrices Tisulares/métodos , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Mucina-1 , Polisacáridos/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/metabolismo
9.
J Immunol ; 206(10): 2290-2300, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33911007

RESUMEN

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.


Asunto(s)
Alérgenos/administración & dosificación , Anafilaxia/tratamiento farmacológico , Anafilaxia/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Desensibilización Inmunológica/métodos , Sistemas de Liberación de Medicamentos/métodos , Inmunoglobulina E/metabolismo , Lectinas/metabolismo , Mastocitos/inmunología , Nanopartículas/química , Polisacáridos/administración & dosificación , Receptores de IgE/metabolismo , Anafilaxia/inmunología , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/genética , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Lectinas/genética , Ligandos , Liposomas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Polisacáridos/metabolismo , Ratas , Receptores de IgE/genética , Resultado del Tratamiento
10.
Nat Commun ; 11(1): 1233, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144244

RESUMEN

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.


Asunto(s)
Antígenos Virales/genética , Evolución Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Receptores de Superficie Celular/metabolismo , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Sitios de Unión/genética , Cristalografía por Rayos X , Análisis Mutacional de ADN , Perros , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Células de Riñón Canino Madin Darby , Mutación , Dominios Proteicos/genética , Dominios Proteicos/inmunología , ARN Viral/genética , ARN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
11.
J Am Chem Soc ; 141(36): 14032-14037, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31460762

RESUMEN

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.


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/metabolismo , Linfocitos B/efectos de los fármacos , Lectinas/antagonistas & inhibidores , Ácidos Siálicos/farmacología , Sulfonamidas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Células CHO , Cricetulus , Humanos , Lectinas/metabolismo , Liposomas/química , Liposomas/metabolismo , Ratones , Conformación Molecular , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Ácidos Siálicos/química , Sulfonamidas/química , Linfocitos T/metabolismo
12.
Cell Host Microbe ; 25(6): 836-844.e5, 2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31151913

RESUMEN

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.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Mutación Missense , Adaptación Biológica , Animales , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Sitios de Unión , Embrión de Pollo , Cristalografía por Rayos X , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Subtipo H3N2 del Virus de la Influenza A/genética , Conformación Proteica , Tecnología Farmacéutica/métodos , Cultivo de Virus/métodos
13.
ACS Chem Biol ; 14(4): 644-654, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30835424

RESUMEN

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.


Asunto(s)
Artritis Reumatoide/metabolismo , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Estudios de Casos y Controles , Citrulina/metabolismo , Humanos , Memoria Inmunológica , Ratones
14.
J Clin Invest ; 129(3): 1387-1401, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30645205

RESUMEN

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.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/prevención & control , Desensibilización Inmunológica , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Anafilaxia/genética , Anafilaxia/inmunología , Anafilaxia/patología , Animales , Broncoconstricción/genética , Broncoconstricción/inmunología , Humanos , Inmunoglobulina E/genética , Mastocitos/patología , Ratones , Ratones Transgénicos , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética
15.
Cell Chem Biol ; 26(1): 131-136.e4, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30393066

RESUMEN

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.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Macrófagos/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética
16.
Nat Commun ; 9(1): 1264, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593268

RESUMEN

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.


Asunto(s)
Sitios de Unión , Epistasis Genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Gripe Humana/virología , Mutación , Receptores Virales/química , Antígenos Virales/química , Células HEK293 , Humanos , Enlace de Hidrógeno , Sistema Inmunológico , Subtipo H3N2 del Virus de la Influenza A/genética , Oligosacáridos/química , Unión Proteica
17.
J Am Chem Soc ; 140(10): 3592-3602, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29446631

RESUMEN

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.


Asunto(s)
Polisacáridos/química , Células HeLa , Humanos , Liposomas/química , Liposomas/metabolismo , Polisacáridos/metabolismo
18.
J Immunol ; 200(3): 949-956, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29288201

RESUMEN

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.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Eritrocitos/inmunología , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/inmunología , Animales , Apoptosis/inmunología , Linfocitos B/metabolismo , Células CHO , Línea Celular , Cricetulus , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Muramidasa/genética , Muramidasa/inmunología , Muramidasa/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Ácidos Siálicos/inmunología
19.
PLoS Pathog ; 13(10): e1006682, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29059230

RESUMEN

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.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Sustitución de Aminoácidos , Antígenos Virales/química , Antígenos Virales/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...