Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 165(4): 813-26, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27114034

ABSTRACT

The HIV-1-envelope (Env) trimer is covered by a glycan shield of ∼90 N-linked oligosaccharides, which comprises roughly half its mass and is a key component of HIV evasion from humoral immunity. To understand how antibodies can overcome the barriers imposed by the glycan shield, we crystallized fully glycosylated Env trimers from clades A, B, and G, visualizing the shield at 3.4-3.7 Å resolution. These structures reveal the HIV-1-glycan shield to comprise a network of interlocking oligosaccharides, substantially ordered by glycan crowding, that encase the protein component of Env and enable HIV-1 to avoid most antibody-mediated neutralization. The revealed features delineate a taxonomy of N-linked glycan-glycan interactions. Crowded and dispersed glycans are differently ordered, conserved, processed, and recognized by antibody. The structures, along with glycan-array binding and molecular dynamics, reveal a diversity in oligosaccharide affinity and a requirement for accommodating glycans among known broadly neutralizing antibodies that target the glycan-shielded trimer.


Subject(s)
HIV-1/chemistry , env Gene Products, Human Immunodeficiency Virus/chemistry , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Crystallography, X-Ray , Glycosylation , HIV-1/classification , HIV-1/immunology , Immune Evasion , Models, Molecular , Molecular Dynamics Simulation , Polysaccharides/analysis , Polysaccharides/metabolism
2.
Immunity ; 48(3): 500-513.e6, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29548671

ABSTRACT

Virtually the entire surface of the HIV-1-envelope trimer is recognized by neutralizing antibodies, except for a highly glycosylated region at the center of the "silent face" on the gp120 subunit. From an HIV-1-infected donor, #74, we identified antibody VRC-PG05, which neutralized 27% of HIV-1 strains. The crystal structure of the antigen-binding fragment of VRC-PG05 in complex with gp120 revealed an epitope comprised primarily of N-linked glycans from N262, N295, and N448 at the silent face center. Somatic hypermutation occurred preferentially at antibody residues that interacted with these glycans, suggesting somatic development of glycan recognition. Resistance to VRC-PG05 in donor #74 involved shifting of glycan-N448 to N446 or mutation of glycan-proximal residue E293. HIV-1 neutralization can thus be achieved at the silent face center by glycan-recognizing antibody; along with other known epitopes, the VRC-PG05 epitope completes coverage by neutralizing antibody of all major exposed regions of the prefusion closed trimer.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV-1/immunology , Polysaccharides/immunology , Amino Acid Sequence , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Antigens, Viral/chemistry , Antigens, Viral/immunology , Binding Sites , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Glycopeptides/chemistry , Glycopeptides/immunology , Glycosylation , HIV Antibodies/chemistry , HIV Antibodies/genetics , HIV Antibodies/metabolism , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , Humans , Models, Molecular , Molecular Conformation , Polysaccharides/chemistry , Protein Binding/immunology , Somatic Hypermutation, Immunoglobulin/immunology , Structure-Activity Relationship
3.
Proc Natl Acad Sci U S A ; 121(5): e2313397121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38252815

ABSTRACT

Non-small cell lung cancer (NSCLC), a major life-threatening disease accounting for 85% of all lung cancer cases, has been treated with tyrosine kinase inhibitors (TKIs), but often resulted in drug resistance, and approximately 60% of TKI-resistant cases are due to acquired secondary (epithelial growth factor receptor) EGFR-T790M mutation. To identify alternative targets for TKI-resistant NSCLC with EGFR-T790M mutation, we found that the three globo-series glycosphingolipids are increasingly expressed on this type of NSCLC cell lines, and among them, the increase of stage-specific embryonic antigen-4 (SSEA-4) expression is the most significant. Compared to TKI-sensitive cell lines, SSEA-4 and the key enzyme ß3GalT5 responsible for the synthesis of SSEA3 are more expressed in TKI-resistant NSCLC cell lines with EGFR-T790M mutation, and the expression levels strongly correlate with poor survival in patients with EGFR mutation. In addition, we demonstrated that a SSEA-4 targeted monoclonal antibody, especially the homogeneous glycoform with well-defined Fc glycan designed to improve effective functions, is highly effective against this subpopulation of NSCLC in cell-based and animal studies. These findings provide a direction for the prediction of tumor recurrence and treatment of TKI-resistant NSCLC with EGFR-T790M mutation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Stage-Specific Embryonic Antigens , Animals , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , ErbB Receptors/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Neoplasm Recurrence, Local
4.
Immunity ; 47(3): 524-537.e3, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28916265

ABSTRACT

Apex broadly neutralizing HIV antibodies (bnAbs) recognize glycans and protein surface close to the 3-fold axis of the envelope (Env) trimer and are among the most potent and broad Abs described. The evolution of apex bnAbs from one donor (CAP256) has been studied in detail and many Abs at different stages of maturation have been described. Using diverse engineering tools, we investigated the involvement of glycan recognition in the development of the CAP256.VRC26 Ab lineage. We found that sialic acid-bearing glycans were recognized by germline-encoded and somatically mutated residues on the Ab heavy chain. This recognition provided an "anchor" for the Abs as the core protein epitope varies, prevented complete neutralization escape, and eventually led to broadening of the response. These findings illustrate how glycan-specific maturation enables a human Ab to cope with pathogen escape mechanisms and will aid in optimization of immunization strategies to induce V2 apex bnAb responses.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , Polysaccharides/metabolism , Amino Acid Sequence , Antibody Affinity/immunology , Antibody Formation/immunology , Binding Sites , Epitopes/immunology , HIV Antibodies/chemistry , HIV Antibodies/classification , HIV Antibodies/genetics , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Infections/virology , Humans , Immunoglobulin Heavy Chains/genetics , Models, Molecular , N-Acetylneuraminic Acid/metabolism , Neutralization Tests , Peptide Fragments/immunology , Phylogeny , Protein Binding/immunology , Protein Conformation , Protein Multimerization
5.
Proc Natl Acad Sci U S A ; 120(49): e2314392120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011546

ABSTRACT

Since the outbreak of Severe Acute Respiratory Syndrome Virus-2 (SARS-CoV-2) in 2019, more than 15 million spike protein sequences have been identified, raising a new challenge for the development of a broadly protective vaccine against the various emerging variants. We found that the virus, like most other human viruses, depends on host-made glycans to shield the conserved epitopes on spike protein from immune response and demonstrated that deletion of the glycan shields exposed highly conserved epitopes and elicited broadly protective immune responses. In this study, we identified 17 conserved epitopes from 14 million spike protein sequences and 11 of the conserved epitopes are in the S2 domain, including the six most conserved epitopes in the stem region. We also demonstrated that deletion of the glycosites in the spike messenger RNA (mRNA) S2 domain or the stem region exposed the highly conserved epitopes and elicited broadly protective immune responses, particularly CD-8+ T cell response against various SARS-CoV-2 variants, and other human coronaviruses including MERS, SARS viruses, and those causing common cold.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/prevention & control , Sugars , RNA, Messenger/genetics , Spike Glycoprotein, Coronavirus/genetics , Vaccines, Synthetic , Epitopes , Antibodies, Viral , mRNA Vaccines
6.
Immunity ; 45(1): 31-45, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27438765

ABSTRACT

The dense patch of high-mannose-type glycans surrounding the N332 glycan on the HIV envelope glycoprotein (Env) is targeted by multiple broadly neutralizing antibodies (bnAbs). This region is relatively conserved, implying functional importance, the origins of which are not well understood. Here we describe the isolation of new bnAbs targeting this region. Examination of these and previously described antibodies to Env revealed that four different bnAb families targeted the (324)GDIR(327) peptide stretch at the base of the gp120 V3 loop and its nearby glycans. We found that this peptide stretch constitutes part of the CCR5 co-receptor binding site, with the high-mannose patch glycans serving to camouflage it from most antibodies. GDIR-glycan bnAbs, in contrast, bound both (324)GDIR(327) peptide residues and high-mannose patch glycans, which enabled broad reactivity against diverse HIV isolates. Thus, as for the CD4 binding site, bnAb effectiveness relies on circumventing the defenses of a critical functional region on Env.


Subject(s)
Antibodies, Neutralizing/immunology , Binding Sites, Antibody/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/immunology , HIV-1/immunology , Polysaccharides/metabolism , Amino Acid Motifs , CD4 Antigens/metabolism , Epitope Mapping , Epitopes/metabolism , Genetic Engineering , HEK293 Cells , HIV Envelope Protein gp120/immunology , Humans , Immunity, Humoral , Immunologic Memory , Peptide Fragments/metabolism , Polysaccharides/immunology , Protein Binding , Receptors, CCR5/metabolism
7.
Immunity ; 44(5): 1215-26, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27192579

ABSTRACT

The high-mannose patch on HIV Env is a preferred target for broadly neutralizing antibodies (bnAbs), but to date, no vaccination regimen has elicited bnAbs against this region. Here, we present the development of a bnAb lineage targeting the high-mannose patch in an HIV-1 subtype-C-infected donor from sub-Saharan Africa. The Abs first acquired autologous neutralization, then gradually matured to achieve breadth. One Ab neutralized >47% of HIV-1 strains with only ∼11% somatic hypermutation and no insertions or deletions. By sequencing autologous env, we determined key residues that triggered the lineage and participated in Ab-Env coevolution. Next-generation sequencing of the Ab repertoire showed an early expansive diversification of the lineage followed by independent maturation of individual limbs, several of them developing notable breadth and potency. Overall, the findings are encouraging from a vaccine standpoint and suggest immunization strategies mimicking the evolution of the entire high-mannose patch and promoting maturation of multiple diverse Ab pathways.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Africa South of the Sahara , Antibody Diversity/genetics , Biological Evolution , Cell Differentiation , Complementarity Determining Regions/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunodominant Epitopes/immunology , Lymphocyte Activation , Mannose/immunology , Mannose/metabolism , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism
8.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35149556

ABSTRACT

Development of the messenger RNA (mRNA) vaccine has emerged as an effective and speedy strategy to control the spread of new pathogens. After vaccination, the mRNA is translated into the real protein vaccine, and there is no need to manufacture the protein in vitro. However, the fate of mRNA and its posttranslational modification inside the cell may affect immune response. Here, we showed that the mRNA vaccine of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein with deletion of glycosites in the receptor-binding domain (RBD) or especially the subunit 2 (S2) domain to expose more conserved epitopes elicited stronger antibody and CD8+ T cell responses with broader protection against the alpha, beta, gamma, delta, and omicron variants, compared to the unmodified mRNA. Immunization of such mRNA resulted in accumulation of misfolded spike protein in the endoplasmic reticulum, causing the up-regulation of BiP/GRP78, XBP1, and p-eIF2α to induce cell apoptosis and strong CD8+ T cell response. In addition, dendritic cells (DCs) incubated with S2-glysosite deleted mRNA vaccine increased class I major histocompatibility complex (MHC I) expression. This study provides a direction for the development of broad-spectrum mRNA vaccines which may not be achieved with the use of expressed proteins as antigens.


Subject(s)
COVID-19 Vaccines/immunology , Spike Glycoprotein, Coronavirus/genetics , Animals , Antibodies, Viral/immunology , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Glycosylation , HEK293 Cells , Histocompatibility Antigens/metabolism , Humans , Immunity , Mice, Inbred BALB C , Unfolded Protein Response , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology
9.
Immunity ; 43(5): 959-73, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588781

ABSTRACT

Broadly neutralizing antibodies (bnAbs) directed to the V2 apex of the HIV envelope (Env) trimer isolated from individual HIV-infected donors potently neutralize diverse HIV strains, but strategies for designing immunogens to elicit bnAbs have not been identified. Here, we compared four prototypes (PG9, CH01, PGT145, and CAP256.VRC26.09) of V2 apex bnAbs and showed that all recognized a core epitope of basic V2 residues and the glycan-N160. Two prototype bnAbs were derived from VH-germlines that were 99% identical and used a common germline D-gene encoded YYD-motif to interact with the V2-epitope. We identified isolates that were neutralized by inferred germline (iGL) versions of three of the prototype bnAbs. Soluble Env derived from one of these isolates was shown to form a well-ordered Env trimer that could serve as an immunogen to initiate a V2-apex bnAb response. These studies illustrate a strategy to transition from panels of bnAbs to vaccine candidates.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV-1/immunology , Vaccines/immunology , Viral Envelope Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Epitopes/immunology , HEK293 Cells , HIV Infections/immunology , Humans , Molecular Sequence Data
10.
Glycobiology ; 33(5): 423-431, 2023 06 03.
Article in English | MEDLINE | ID: mdl-36988396

ABSTRACT

A novel Gal-binding lectin from mussels (Crenomytilus grayanus, CGL) with 6 binding sites in the dimeric structure has been previously shown to have antifungal, anticancer, and antibacterial activities. In this study, a glycan array was used to confirm that CGL recognizes a range of non-reducing end α- or ß-linked Gal glycans on normal cells but not sialic acid-capped glycans. This finding suggests that CGL has potential in the tumor detection due to the hyper-sialylation present in cell surface glycans from cancer cells. To evaluate the feasibility of this possibility, we labeled CGL with biotin and then mixed it with streptavidin-horseradish peroxidase (HRP) to create a CGL-biotin-SP complex as a probe for use in enzyme-linked lectin assays. CGL-biotin-SP successfully distinguished not only HeLa cells and de-sialylated HeLa cells that mimic normal cell surface glycans but also lung and breast cancer cells with different metastatic abilities. This work provides the insights into a new Gal-binding lectin by establishing its specificity and also demonstrates practical applications in cancer diagnosis greater than other reported lectins.


Subject(s)
Lectins , Mytilidae , Animals , Humans , Lectins/chemistry , HeLa Cells , Biotin , Mytilidae/metabolism , Polysaccharides/metabolism
11.
J Am Chem Soc ; 145(17): 9840-9849, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37089019

ABSTRACT

Polysaccharides have been successfully used as immunogens for the development of vaccines against bacterial infection; however, there are no oligosaccharide-based vaccines available to date and no previous studies of their processing and presentation. We reported here the intracellular enzymatic processing and antigen presentation of an oligosaccharide-conjugate cancer vaccine prepared from the glycan of Globo-H (GH), a globo-series glycosphingolipid (GSL). This oligosaccharide-conjugate vaccine was shown to elicit antibodies against the glycan moieties of all three globo-series GSLs that are exclusively expressed on many types of cancer and their stem cells. To understand the specificity and origin of cross-reactivity of the antibodies elicited by the vaccine, we found that the vaccine is first processed by fucosidase 1 in the early endosome of dendritic cells to generate a common glycan antigen of the GSLs along with GH for MHC class II presentation. This work represents the first study of oligosaccharide processing and presentation and is expected to facilitate the design and development of glycoconjugate vaccines based on oligosaccharide antigens.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Vaccines, Conjugate , Antigen Presentation , Antibodies , Polysaccharides , Oligosaccharides
12.
PLoS Pathog ; 17(3): e1009407, 2021 03.
Article in English | MEDLINE | ID: mdl-33750987

ABSTRACT

Incessant antigenic evolution enables the persistence and spread of influenza virus in the human population. As the principal target of the immune response, the hemagglutinin (HA) surface antigen on influenza viruses continuously acquires and replaces N-linked glycosylation sites to shield immunogenic protein epitopes using host-derived glycans. Anti-glycan antibodies, such as 2G12, target the HIV-1 envelope protein (Env), which is even more extensively glycosylated and contains under-processed oligomannose-type clusters on its dense glycan shield. Here, we illustrate that 2G12 can also neutralize human seasonal influenza A H3N2 viruses that have evolved to present similar oligomannose-type clusters on their HAs from around 20 years after the 1968 pandemic. Using structural biology and mass spectrometric approaches, we find that two N-glycosylation sites close to the receptor binding site (RBS) on influenza hemagglutinin represent the oligomannose cluster recognized by 2G12. One of these glycan sites is highly conserved in all human H3N2 strains and the other emerged during virus evolution. These two N-glycosylation sites have also become crucial for fitness of recent H3N2 strains. These findings shed light on the evolution of the glycan shield on influenza virus and suggest 2G12-like antibodies can potentially act as broad neutralizers to target human enveloped viruses.


Subject(s)
Antibodies, Viral/immunology , HIV-1/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/immunology , Broadly Neutralizing Antibodies , Cross Reactions , HIV Infections/immunology , Humans , Influenza, Human/immunology
13.
Immunity ; 40(5): 657-68, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24768347

ABSTRACT

Broadly neutralizing HIV antibodies are much sought after (a) to guide vaccine design, both as templates and as indicators of the authenticity of vaccine candidates, (b) to assist in structural studies, and (c) to serve as potential therapeutics. However, the number of targets on the viral envelope spike for such antibodies has been limited. Here, we describe a set of human monoclonal antibodies that define what is, to the best of our knowledge, a previously undefined target on HIV Env. The antibodies recognize a glycan-dependent epitope on the prefusion conformation of gp41 and unambiguously distinguish cleaved from uncleaved Env trimers, an important property given increasing evidence that cleavage is required for vaccine candidates that seek to mimic the functional HIV envelope spike. The availability of this set of antibodies expands the number of vaccine targets on HIV and provides reagents to characterize the native envelope spike.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/immunology , Cell Line , Epitopes/immunology , HEK293 Cells , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/metabolism , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Humans , Molecular Sequence Data , Polysaccharides/immunology
14.
J Biomed Sci ; 29(1): 9, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35130876

ABSTRACT

BACKGROUND: K1 capsular polysaccharide (CPS)-associated Klebsiella pneumoniae is the primary cause of pyogenic liver abscesses (PLA) in Asia. Patients with PLA often have serious complications, ultimately leading to a mortality of ~ 5%. This K1 CPS has been reported as a promising target for development of glycoconjugate vaccines against K. pneumoniae infection. The pyruvylation and O-acetylation modifications on the K1 CPS are essential to the immune response induced by the CPS. To date, however, obtaining the fragments of K1 CPS that contain the pyruvylation and O-acetylation for generating glycoconjugate vaccines still remains a challenge. METHODS: We analyzed the digested CPS products with NMR spectroscopy and mass spectrometry to reveal a bacteriophage-derived polysaccharide depolymerase specific to K1 CPS. The biochemical and biophysical properties of the enzyme were characterized and its crystal structures containing bound CPS products were determined. We also performed site-directed mutagenesis, enzyme kinetic analysis, phage absorption and infectivity studies, and treatment of the K. pneumoniae-infected mice with the wild-type and mutant enzymes. RESULTS: We found a bacteriophage-derived polysaccharide lyase that depolymerizes the K1 CPS into fragments of 1-3 repeating trisaccharide units with the retention of the pyruvylation and O-acetylation, and thus the important antigenic determinants of intact K1 CPS. We also determined the 1.46-Å-resolution, product-bound crystal structure of the enzyme, revealing two distinct carbohydrate-binding sites in a trimeric ß-helix architecture, which provide the first direct evidence for a second, non-catalytic, carbohydrate-binding site in bacteriophage-derived polysaccharide depolymerases. We demonstrate the tight interaction between the pyruvate moiety of K1 CPS and the enzyme in this second carbohydrate-binding site to be crucial to CPS depolymerization of the enzyme as well as phage absorption and infectivity. We also demonstrate that the enzyme is capable of protecting mice from K1 K. pneumoniae infection, even against a high challenge dose. CONCLUSIONS: Our results provide insights into how the enzyme recognizes and depolymerizes the K1 CPS, and demonstrate the potential use of the protein not only as a therapeutic agent against K. pneumoniae, but also as a tool to prepare structurally-defined oligosaccharides for the generation of glycoconjugate vaccines against infections caused by this organism.


Subject(s)
Bacteriophages , Klebsiella Infections , Lyases , Animals , Bacterial Capsules/genetics , Bacteriophages/genetics , Humans , Kinetics , Klebsiella pneumoniae , Mice
15.
Proc Natl Acad Sci U S A ; 116(9): 3518-3523, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808745

ABSTRACT

The globo-series glycosphingolipids (GSLs) SSEA3, SSEA4, and Globo-H specifically expressed on cancer cells are found to correlate with tumor progression and metastasis, but the functional roles of these GSLs and the key enzyme ß1,3-galactosyltransferase V (ß3GalT5) that converts Gb4 to SSEA3 remain largely unclear. Here we show that the expression of ß3GalT5 significantly correlates with tumor progression and poor survival in patients, and the globo-series GSLs in breast cancer cells form a complex in membrane lipid raft with caveolin-1 (CAV1) and focal adhesion kinase (FAK) which then interact with AKT and receptor-interacting protein kinase (RIP), respectively. Knockdown of ß3GalT5 disrupts the complex and induces apoptosis through dissociation of RIP from the complex to interact with the Fas death domain (FADD) and trigger the Fas-dependent pathway. This finding provides a link between SSEA3/SSEA4/Globo-H and the FAK/CAV1/AKT/RIP complex in tumor progression and apoptosis and suggests a direction for the treatment of breast cancer, as demonstrated by the combined use of antibodies against Globo-H and SSEA4.


Subject(s)
Breast Neoplasms/genetics , Galactosyltransferases/genetics , Glycosphingolipids/genetics , Membrane Microdomains/genetics , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Apoptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Disease Progression , Fas-Associated Death Domain Protein/genetics , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Neoplastic/genetics , Glycosphingolipids/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Membrane Microdomains/metabolism , Middle Aged , Proto-Oncogene Proteins c-akt/genetics , Saporins/genetics , Signal Transduction/genetics , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism
17.
Bioorg Chem ; 112: 104863, 2021 07.
Article in English | MEDLINE | ID: mdl-33823405

ABSTRACT

The chemoenzymatic remodeled monoclonal antidodies with well-defined glycan structure at the Fc domain display improved biological activities, such as ADCC and ADCP, and are more likely to yield a better safety profile by eliminating the non-human glycans derived from CHO cell culture. We covalently immobilize wild type endoglycosidase S (EndoS), fucosidase, and EndoS2 mutant on magnetic beads through a linker to efficiently generate homogeneous antibody glycoforms without additional purification step to remove endoglycosidase and fucosidase. We also used the biotinylated wild type EndoS2 and EndoS2 mutant in combination with covalently immobilized fucosidase on magnetic beads to allow the sequential removal of endoglycosidases and fucosidase for efficient glyco-engineering and isolation of antibodies without purifying deglycosylated antibody intermediate. Notably, the relatively expensive fucosidase can be recovered to reduce the cost, and the strong affinity of streptavidin to biotin would complete the isolation of biotinylated enzymes. We used Trastuzumab as a model to demonstrate both approaches were reliable for the large-scale production and isolation of antibodies without the residual contamination of endoglycosidase to avoid deglycosylation over storage time.


Subject(s)
Anti-Bacterial Agents/metabolism , Drug Development , Glycoside Hydrolases/metabolism , Trastuzumab/metabolism , alpha-L-Fucosidase/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Biotinylation , Dose-Response Relationship, Drug , Enzymes, Immobilized/genetics , Enzymes, Immobilized/metabolism , Glycoside Hydrolases/genetics , Magnetic Phenomena , Molecular Structure , Mutation , Structure-Activity Relationship , Trastuzumab/chemistry , Trastuzumab/isolation & purification , alpha-L-Fucosidase/genetics
18.
Proc Natl Acad Sci U S A ; 115(4): 720-725, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311294

ABSTRACT

Monoclonal antibodies (mAbs) have been developed as therapeutics, especially for the treatment of cancer, inflammation, and infectious diseases. Because the glycosylation of mAbs in the Fc region influences their interaction with effector cells that kill antibody-targeted cells, and the current method of antibody production is relatively expensive, efforts have been directed toward the development of alternative expressing systems capable of large-scale production of mAbs with desirable glycoforms. In this study, we demonstrate that the mAb trastuzumab expressed in glycoengineered P. pastoris can be remodeled through deglycosylation by endoglycosidases identified from the Carbohydrate Active Enzymes database and through transglycosylation using glycans with a stable leaving group to generate a homogeneous antibody designed to optimize the effector functions. The 10 newly identified recombinant bacterial endoglycosidases are complementary to existing endoglycosidases (EndoA, EndoH, EndoS), two of which can even accept sialylated tri- and tetraantennary glycans as substrates.


Subject(s)
Glycoproteins/pharmacology , Glycoside Hydrolases/metabolism , Trastuzumab/chemistry , Antibodies, Monoclonal/metabolism , Antibody-Dependent Cell Cytotoxicity/physiology , Glycoproteins/metabolism , Glycoside Hydrolases/pharmacology , Glycosylation , Humans , Pichia/metabolism , Polysaccharides/metabolism , Saccharomyces cerevisiae/metabolism , Trastuzumab/metabolism
19.
Proc Natl Acad Sci U S A ; 115(28): 7302-7307, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29941599

ABSTRACT

Protein O-glycosylation by attachment of ß-N-acetylglucosamine (GlcNAc) to the Ser or Thr residue is a major posttranslational glycosylation event and is often associated with protein folding, stability, and activity. The methylation of histone H3 at Lys-27 catalyzed by the methyltransferase EZH2 was known to suppress gene expression and cancer development, and we previously reported that the O-GlcNAcylation of EZH2 at S76 stabilized EZH2 and facilitated the formation of H3K27me3 to inhibit tumor suppression. In this study, we employed a fluorescence-based method of sugar labeling combined with mass spectrometry to investigate EZH2 glycosylation and identified five O-GlcNAcylation sites. We also find that mutation of one or more of the O-GlcNAcylation sites S73A, S76A, S84A, and T313A in the N-terminal region decreases the stability of EZH2, but does not affect its association with the PRC2 components SUZ12 and EED. Mutation of the C-terminal O-GlcNAcylation site (S729A) in the catalytic domain of EZH2 abolishes the di- and trimethylation activities, but not the monomethylation of H3K27, nor the integrity of the PRC2/EZH2 core complex. Our results show the effect of individual O-GlcNAcylation sites on the function of EZH2 and suggest an alternative approach to tumor suppression through selective inhibition of EZH2 O-GlcNAcylation.


Subject(s)
Acetylglucosamine/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Mutation, Missense , Acetylglucosamine/chemistry , Acetylglucosamine/genetics , Amino Acid Substitution , Cell Line , Enhancer of Zeste Homolog 2 Protein/chemistry , Enhancer of Zeste Homolog 2 Protein/genetics , Enzyme Stability , Glycosylation , Humans , Protein Domains
20.
Proc Natl Acad Sci U S A ; 115(10): E2386-E2392, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463703

ABSTRACT

Epidemics and pandemics of influenza are characterized by rapid global spread mediated by non-mutually exclusive transmission modes. The relative significance between contact, droplet, and airborne transmission is yet to be defined, a knowledge gap for implementing evidence-based infection control measures. We devised a transmission chamber that separates virus-laden particles by size and determined the particle sizes mediating transmission of influenza among ferrets through the air. Ferret-to-ferret transmission was mediated by airborne particles larger than 1.5 µm, consistent with the quantity and size of virus-laden particles released by the donors. Onward transmission by donors was most efficient before fever onset and may continue for 5 days after inoculation. Multiple virus gene segments enhanced the transmissibility of a swine influenza virus among ferrets by increasing the release of virus-laden particles into the air. We provide direct experimental evidence of influenza transmission via droplets and fine droplet nuclei, albeit at different efficiency.


Subject(s)
Air/analysis , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/transmission , Influenza, Human/virology , Air Microbiology , Animals , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Male , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL