Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions. / Loke, Ian; Østergaard, Ole; Heegaard, Niels H H; Packer, Nicolle H; Thaysen-Andersen, Morten.

In: Molecular and Cellular Proteomics, Vol. 16, No. 8, 08.2017, p. 1507-1527.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Loke, I, Østergaard, O, Heegaard, NHH, Packer, NH & Thaysen-Andersen, M 2017, 'Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions', Molecular and Cellular Proteomics, vol. 16, no. 8, pp. 1507-1527. https://doi.org/10.1074/mcp.M116.066746

APA

Loke, I., Østergaard, O., Heegaard, N. H. H., Packer, N. H., & Thaysen-Andersen, M. (2017). Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions. Molecular and Cellular Proteomics, 16(8), 1507-1527. https://doi.org/10.1074/mcp.M116.066746

Vancouver

Loke I, Østergaard O, Heegaard NHH, Packer NH, Thaysen-Andersen M. Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions. Molecular and Cellular Proteomics. 2017 Aug;16(8):1507-1527. https://doi.org/10.1074/mcp.M116.066746

Author

Loke, Ian ; Østergaard, Ole ; Heegaard, Niels H H ; Packer, Nicolle H ; Thaysen-Andersen, Morten. / Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions. In: Molecular and Cellular Proteomics. 2017 ; Vol. 16, No. 8. pp. 1507-1527.

Bibtex

@article{86f18882edfa4843bc8f72a2c06d6416,
title = "Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions",
abstract = "Human neutrophil elastase (HNE) is an important N-glycosylated serine protease in the innate immune system, but the structure and immune-modulating functions of HNE N-glycosylation remain undescribed. Herein, LC-MS/MS-based glycan, glycopeptide and glycoprotein profiling were utilized to first determine the heterogeneous N-glycosylation of HNE purified from neutrophil lysates and then from isolated neutrophil granules of healthy individuals. The spatiotemporal expression of HNE during neutrophil activation and the biological importance of its N-glycosylation were also investigated using immunoblotting, cell surface capture, native MS, receptor interaction, protease inhibition, and bacteria growth assays. Site-specific HNE glycoprofiling demonstrated that unusual paucimannosidic N-glycans, particularly Manα1,6Manβ1,4GlcNAcβ1,4(Fucα1,6)GlcNAcβ, predominantly occupied Asn124 and Asn173. The equally unusual core fucosylated monoantenna complex-type N-sialoglycans also decorated these two fully occupied sites. In contrast, the mostly unoccupied Asn88 carried nonfucosylated paucimannosidic N-glycans probably resulting from low glycosylation site solvent accessibility. Asn185 was not glycosylated. Subcellular- and site-specific glycoprofiling showed highly uniform N-glycosylation of HNE residing in distinct neutrophil compartments. Stimulation-induced cell surface mobilization demonstrated a spatiotemporal regulation, but not cell surface-specific glycosylation signatures, of HNE in activated human neutrophils. The three glycosylation sites of HNE were located distal to the active site indicating glycan functions other than interference with HNE enzyme activity. Functionally, the paucimannosidic HNE glycoforms displayed preferential binding to human mannose binding lectin compared with the HNE sialoglycoforms, suggesting a glycoform-dependent involvement of HNE in complement activation. The heavily N-glycosylated HNE protease inhibitor, α1-antitrypsin, displayed concentration-dependent complex formation and preferred glycoform-glycoform interactions with HNE. Finally, both enzymatically active HNE and isolated HNE N-glycans demonstrated low micromolar concentration-dependent growth inhibition of clinically-relevant Pseudomonas aeruginosa, suggesting some bacteriostatic activity is conferred by the HNE N-glycans. Taken together, these observations support that the unusual HNE N-glycosylation, here reported for the first time, is involved in modulating multiple immune functions central to inflammation and infection.",
keywords = "Bacterial Infections/immunology, Catalytic Domain, Complement Activation, Glycopeptides/metabolism, Glycoproteins/metabolism, Glycosylation, Humans, Immunity, Innate, Inflammation/immunology, Lectins/metabolism, Leukocyte Elastase/antagonists & inhibitors, Male, Mannose/metabolism, Neutrophils/enzymology, Polysaccharides/metabolism, Pseudomonas Infections/immunology, Pseudomonas aeruginosa/immunology, alpha 1-Antitrypsin/pharmacology",
author = "Ian Loke and Ole {\O}stergaard and Heegaard, {Niels H H} and Packer, {Nicolle H} and Morten Thaysen-Andersen",
note = "{\textcopyright} 2017 by The American Society for Biochemistry and Molecular Biology, Inc.",
year = "2017",
month = aug,
doi = "10.1074/mcp.M116.066746",
language = "English",
volume = "16",
pages = "1507--1527",
journal = "Molecular and Cellular Proteomics",
issn = "1535-9476",
publisher = "American Society for Biochemistry and Molecular Biology",
number = "8",

}

RIS

TY - JOUR

T1 - Paucimannose-Rich N-glycosylation of Spatiotemporally Regulated Human Neutrophil Elastase Modulates Its Immune Functions

AU - Loke, Ian

AU - Østergaard, Ole

AU - Heegaard, Niels H H

AU - Packer, Nicolle H

AU - Thaysen-Andersen, Morten

N1 - © 2017 by The American Society for Biochemistry and Molecular Biology, Inc.

PY - 2017/8

Y1 - 2017/8

N2 - Human neutrophil elastase (HNE) is an important N-glycosylated serine protease in the innate immune system, but the structure and immune-modulating functions of HNE N-glycosylation remain undescribed. Herein, LC-MS/MS-based glycan, glycopeptide and glycoprotein profiling were utilized to first determine the heterogeneous N-glycosylation of HNE purified from neutrophil lysates and then from isolated neutrophil granules of healthy individuals. The spatiotemporal expression of HNE during neutrophil activation and the biological importance of its N-glycosylation were also investigated using immunoblotting, cell surface capture, native MS, receptor interaction, protease inhibition, and bacteria growth assays. Site-specific HNE glycoprofiling demonstrated that unusual paucimannosidic N-glycans, particularly Manα1,6Manβ1,4GlcNAcβ1,4(Fucα1,6)GlcNAcβ, predominantly occupied Asn124 and Asn173. The equally unusual core fucosylated monoantenna complex-type N-sialoglycans also decorated these two fully occupied sites. In contrast, the mostly unoccupied Asn88 carried nonfucosylated paucimannosidic N-glycans probably resulting from low glycosylation site solvent accessibility. Asn185 was not glycosylated. Subcellular- and site-specific glycoprofiling showed highly uniform N-glycosylation of HNE residing in distinct neutrophil compartments. Stimulation-induced cell surface mobilization demonstrated a spatiotemporal regulation, but not cell surface-specific glycosylation signatures, of HNE in activated human neutrophils. The three glycosylation sites of HNE were located distal to the active site indicating glycan functions other than interference with HNE enzyme activity. Functionally, the paucimannosidic HNE glycoforms displayed preferential binding to human mannose binding lectin compared with the HNE sialoglycoforms, suggesting a glycoform-dependent involvement of HNE in complement activation. The heavily N-glycosylated HNE protease inhibitor, α1-antitrypsin, displayed concentration-dependent complex formation and preferred glycoform-glycoform interactions with HNE. Finally, both enzymatically active HNE and isolated HNE N-glycans demonstrated low micromolar concentration-dependent growth inhibition of clinically-relevant Pseudomonas aeruginosa, suggesting some bacteriostatic activity is conferred by the HNE N-glycans. Taken together, these observations support that the unusual HNE N-glycosylation, here reported for the first time, is involved in modulating multiple immune functions central to inflammation and infection.

AB - Human neutrophil elastase (HNE) is an important N-glycosylated serine protease in the innate immune system, but the structure and immune-modulating functions of HNE N-glycosylation remain undescribed. Herein, LC-MS/MS-based glycan, glycopeptide and glycoprotein profiling were utilized to first determine the heterogeneous N-glycosylation of HNE purified from neutrophil lysates and then from isolated neutrophil granules of healthy individuals. The spatiotemporal expression of HNE during neutrophil activation and the biological importance of its N-glycosylation were also investigated using immunoblotting, cell surface capture, native MS, receptor interaction, protease inhibition, and bacteria growth assays. Site-specific HNE glycoprofiling demonstrated that unusual paucimannosidic N-glycans, particularly Manα1,6Manβ1,4GlcNAcβ1,4(Fucα1,6)GlcNAcβ, predominantly occupied Asn124 and Asn173. The equally unusual core fucosylated monoantenna complex-type N-sialoglycans also decorated these two fully occupied sites. In contrast, the mostly unoccupied Asn88 carried nonfucosylated paucimannosidic N-glycans probably resulting from low glycosylation site solvent accessibility. Asn185 was not glycosylated. Subcellular- and site-specific glycoprofiling showed highly uniform N-glycosylation of HNE residing in distinct neutrophil compartments. Stimulation-induced cell surface mobilization demonstrated a spatiotemporal regulation, but not cell surface-specific glycosylation signatures, of HNE in activated human neutrophils. The three glycosylation sites of HNE were located distal to the active site indicating glycan functions other than interference with HNE enzyme activity. Functionally, the paucimannosidic HNE glycoforms displayed preferential binding to human mannose binding lectin compared with the HNE sialoglycoforms, suggesting a glycoform-dependent involvement of HNE in complement activation. The heavily N-glycosylated HNE protease inhibitor, α1-antitrypsin, displayed concentration-dependent complex formation and preferred glycoform-glycoform interactions with HNE. Finally, both enzymatically active HNE and isolated HNE N-glycans demonstrated low micromolar concentration-dependent growth inhibition of clinically-relevant Pseudomonas aeruginosa, suggesting some bacteriostatic activity is conferred by the HNE N-glycans. Taken together, these observations support that the unusual HNE N-glycosylation, here reported for the first time, is involved in modulating multiple immune functions central to inflammation and infection.

KW - Bacterial Infections/immunology

KW - Catalytic Domain

KW - Complement Activation

KW - Glycopeptides/metabolism

KW - Glycoproteins/metabolism

KW - Glycosylation

KW - Humans

KW - Immunity, Innate

KW - Inflammation/immunology

KW - Lectins/metabolism

KW - Leukocyte Elastase/antagonists & inhibitors

KW - Male

KW - Mannose/metabolism

KW - Neutrophils/enzymology

KW - Polysaccharides/metabolism

KW - Pseudomonas Infections/immunology

KW - Pseudomonas aeruginosa/immunology

KW - alpha 1-Antitrypsin/pharmacology

U2 - 10.1074/mcp.M116.066746

DO - 10.1074/mcp.M116.066746

M3 - Journal article

C2 - 28630087

VL - 16

SP - 1507

EP - 1527

JO - Molecular and Cellular Proteomics

JF - Molecular and Cellular Proteomics

SN - 1535-9476

IS - 8

ER -

ID: 210473645