Cargando…

The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages

Sepsis is a major cause of death for hospitalized patients and is characterized by massive overreaction of immune responses to invading pathogens which is mediated by cytokines. For decades, there has been no effective treatment for sepsis. Sialic acid-binding, Ig-like lectin-9 (Siglec-9), is an imm...

Descripción completa

Detalles Bibliográficos
Autores principales: Chu, Sasa, Zhu, Xuhui, You, Na, Zhang, Wei, Zheng, Feng, Cai, Binggang, Zhou, Tingting, Wang, Yiwen, Sun, Qiannan, Yang, Zhiguo, Zhang, Xin, Wang, Changjun, Nie, Shinan, Zhu, Jin, Wang, Maorong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5183739/
https://www.ncbi.nlm.nih.gov/pubmed/28082984
http://dx.doi.org/10.3389/fimmu.2016.00649
_version_ 1782486105856147456
author Chu, Sasa
Zhu, Xuhui
You, Na
Zhang, Wei
Zheng, Feng
Cai, Binggang
Zhou, Tingting
Wang, Yiwen
Sun, Qiannan
Yang, Zhiguo
Zhang, Xin
Wang, Changjun
Nie, Shinan
Zhu, Jin
Wang, Maorong
author_facet Chu, Sasa
Zhu, Xuhui
You, Na
Zhang, Wei
Zheng, Feng
Cai, Binggang
Zhou, Tingting
Wang, Yiwen
Sun, Qiannan
Yang, Zhiguo
Zhang, Xin
Wang, Changjun
Nie, Shinan
Zhu, Jin
Wang, Maorong
author_sort Chu, Sasa
collection PubMed
description Sepsis is a major cause of death for hospitalized patients and is characterized by massive overreaction of immune responses to invading pathogens which is mediated by cytokines. For decades, there has been no effective treatment for sepsis. Sialic acid-binding, Ig-like lectin-9 (Siglec-9), is an immunomodulatory receptor expressed primarily on hematopoietic cells which is involved in various aspects of inflammatory responses and is a potential target for treatment of sepsis. The aim of the present study was to develop a human anti-Siglec-9 Fab fragment, which was named hS9-Fab03 and investigate its immune activity in human macrophages. We began by constructing the hS9-Fab03 prokaryotic expression vector from human antibody library and phage display. Then, we utilized a multitude of assays, including SDS-PAGE, Western blotting, ELISA, affinity, and kinetics assay to evaluate the binding affinity and specificity of hS9-Fab03. Results demonstrated that hS9-Fab03 specifically bind to Siglec-9 antigen with high affinity, and pretreatment with hS9-Fab03 could attenuate lipopolysaccharide (LPS)-induced TNF-α, IL-6, IL-1β, IL-8, and IFN-β production in human PBMC-derived macrophages, but slightly increased IL-10 production in an early time point. We also observed similar results in human THP-1-differentiated macrophages. Collectively, we prepared the hS9-Fab03 with efficient activity for blocking LPS-induced pro-inflammatory cytokines production in human macrophages. These results indicated that ligation of Siglec-9 with hS9-Fab03 might be a novel anti-inflammatory therapeutic strategy for sepsis.
format Online
Article
Text
id pubmed-5183739
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-51837392017-01-12 The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages Chu, Sasa Zhu, Xuhui You, Na Zhang, Wei Zheng, Feng Cai, Binggang Zhou, Tingting Wang, Yiwen Sun, Qiannan Yang, Zhiguo Zhang, Xin Wang, Changjun Nie, Shinan Zhu, Jin Wang, Maorong Front Immunol Immunology Sepsis is a major cause of death for hospitalized patients and is characterized by massive overreaction of immune responses to invading pathogens which is mediated by cytokines. For decades, there has been no effective treatment for sepsis. Sialic acid-binding, Ig-like lectin-9 (Siglec-9), is an immunomodulatory receptor expressed primarily on hematopoietic cells which is involved in various aspects of inflammatory responses and is a potential target for treatment of sepsis. The aim of the present study was to develop a human anti-Siglec-9 Fab fragment, which was named hS9-Fab03 and investigate its immune activity in human macrophages. We began by constructing the hS9-Fab03 prokaryotic expression vector from human antibody library and phage display. Then, we utilized a multitude of assays, including SDS-PAGE, Western blotting, ELISA, affinity, and kinetics assay to evaluate the binding affinity and specificity of hS9-Fab03. Results demonstrated that hS9-Fab03 specifically bind to Siglec-9 antigen with high affinity, and pretreatment with hS9-Fab03 could attenuate lipopolysaccharide (LPS)-induced TNF-α, IL-6, IL-1β, IL-8, and IFN-β production in human PBMC-derived macrophages, but slightly increased IL-10 production in an early time point. We also observed similar results in human THP-1-differentiated macrophages. Collectively, we prepared the hS9-Fab03 with efficient activity for blocking LPS-induced pro-inflammatory cytokines production in human macrophages. These results indicated that ligation of Siglec-9 with hS9-Fab03 might be a novel anti-inflammatory therapeutic strategy for sepsis. Frontiers Media S.A. 2016-12-26 /pmc/articles/PMC5183739/ /pubmed/28082984 http://dx.doi.org/10.3389/fimmu.2016.00649 Text en Copyright © 2016 Chu, Zhu, You, Zhang, Zheng, Cai, Zhou, Wang, Sun, Yang, Zhang, Wang, Nie, Zhu and Wang. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Chu, Sasa
Zhu, Xuhui
You, Na
Zhang, Wei
Zheng, Feng
Cai, Binggang
Zhou, Tingting
Wang, Yiwen
Sun, Qiannan
Yang, Zhiguo
Zhang, Xin
Wang, Changjun
Nie, Shinan
Zhu, Jin
Wang, Maorong
The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title_full The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title_fullStr The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title_full_unstemmed The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title_short The Fab Fragment of a Human Anti-Siglec-9 Monoclonal Antibody Suppresses LPS-Induced Inflammatory Responses in Human Macrophages
title_sort fab fragment of a human anti-siglec-9 monoclonal antibody suppresses lps-induced inflammatory responses in human macrophages
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5183739/
https://www.ncbi.nlm.nih.gov/pubmed/28082984
http://dx.doi.org/10.3389/fimmu.2016.00649
work_keys_str_mv AT chusasa thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhuxuhui thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT youna thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhangwei thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhengfeng thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT caibinggang thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhoutingting thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangyiwen thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT sunqiannan thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT yangzhiguo thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhangxin thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangchangjun thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT nieshinan thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhujin thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangmaorong thefabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT chusasa fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhuxuhui fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT youna fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhangwei fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhengfeng fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT caibinggang fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhoutingting fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangyiwen fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT sunqiannan fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT yangzhiguo fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhangxin fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangchangjun fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT nieshinan fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT zhujin fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages
AT wangmaorong fabfragmentofahumanantisiglec9monoclonalantibodysuppresseslpsinducedinflammatoryresponsesinhumanmacrophages