Cargando…

Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening condition of critically-ill patients, characterized by overwhelming inflammatory responses in the lung. Multiple lines of evidence suggest that the excessive activation of Toll-like receptor 4 (TLR4) plays an im...

Descripción completa

Detalles Bibliográficos
Autores principales: Gao, Wei, Wang, Yulu, Xiong, Ye, Sun, Liya, Wang, Lu, Wang, Kun, Lu, Henry Y., Bao, Aihua, Turvey, Stuart E., Li, Qiang, Yang, Hong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Acta Materialia Inc. Published by Elsevier Ltd. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8960115/
https://www.ncbi.nlm.nih.gov/pubmed/30597258
http://dx.doi.org/10.1016/j.actbio.2018.12.046
_version_ 1784677318501859328
author Gao, Wei
Wang, Yulu
Xiong, Ye
Sun, Liya
Wang, Lu
Wang, Kun
Lu, Henry Y.
Bao, Aihua
Turvey, Stuart E.
Li, Qiang
Yang, Hong
author_facet Gao, Wei
Wang, Yulu
Xiong, Ye
Sun, Liya
Wang, Lu
Wang, Kun
Lu, Henry Y.
Bao, Aihua
Turvey, Stuart E.
Li, Qiang
Yang, Hong
author_sort Gao, Wei
collection PubMed
description Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening condition of critically-ill patients, characterized by overwhelming inflammatory responses in the lung. Multiple lines of evidence suggest that the excessive activation of Toll-like receptor 4 (TLR4) plays an important role in this detrimental lung inflammation. Recently, we developed a unique class of peptide-gold nanoparticle (GNP) hybrids that act as potent nano-inhibitors of TLR4 signaling by modulating the process of endosomal acidification. In this study, we aimed to identify the key physiochemical factors that could further strengthen the anti-inflammatory activity of these nano-inhibitors, including the nanoparticle size, the density of peptides coating the nanoparticle surface, as well as the number of the effective amino acid phenylalanine (F) residues in the peptide sequence. Among these factors, we found that the nanoparticle size could significantly affect the TLR4 inhibition. Specifically, the peptide-GNP hybrids with a GNP core of 20 nm (P12(G20)) exhibited the most potent inhibitory activity on TLR4 activation and its downstream cytokine production among those with a GNP core of 13 nm (P12(G13)) and 5 nm (P12(G5)) in THP-1 cell-derived macrophages. This size-dependent anti-inflammatory effect of the hybrid P12 was also observed in a lipopolysaccharide (LPS)-induced mouse model of ALI. It was found that P12(G20) was superior to P12(G13) in prolonging the survival of mice experiencing lethal LPS challenge, decreasing the acute lung inflammation, and alleviating diffuse alveolar damage in the lungs. Interestingly, P12(G20) could also promote long-term tolerance to endotoxin. Detailed mechanistic studies demonstrated that when compared to the smaller P12(G13), the larger P12(G20) had higher cellular uptake and a stronger endosomal pH buffering capacity, contributing to its enhanced therapeutic effects on reducing TLR4 activation in vitro and in vivo. Overall, this study suggests that nanoparticle size is one key factor determining the anti-inflammatory potency of the peptide-GNP hybrids, and the hybrid P12 may serve as a promising, novel class of nanotherapeutics for modulating TLR signaling to treat ALI/ARDS. STATEMENT OF SIGNIFICANCE: We have developed a new class of nanoparticle-based inhibitors (i.e., peptide-GNP hybrids) targeting TLR4 signaling in macrophages. Through evidence-based engineering of the nanoparticle size, surface peptide ligand density and effective amino acid (phenylalanine, F) chain length, we identified a peptide-GNP hybrid, P12(G20), with enhanced anti-inflammatory activity. Specifically, P12(G20) was more potent in reducing inflammation in THP-1 cell-derived macrophages and in a LPS-induced ALI mouse model. More interestingly, P12(G20) facilitated long-term protection against lethal LPS challenge in vivo and induced endotoxin tolerance in vitro. We anticipate that these new hybrids would serve as the next generation anti-inflammatory nano-therapeutics for the treatment of ALI/ARDS or other acute and chronic inflammatory diseases.
format Online
Article
Text
id pubmed-8960115
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Acta Materialia Inc. Published by Elsevier Ltd.
record_format MEDLINE/PubMed
spelling pubmed-89601152022-03-29 Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury Gao, Wei Wang, Yulu Xiong, Ye Sun, Liya Wang, Lu Wang, Kun Lu, Henry Y. Bao, Aihua Turvey, Stuart E. Li, Qiang Yang, Hong Acta Biomater Full Length Article Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening condition of critically-ill patients, characterized by overwhelming inflammatory responses in the lung. Multiple lines of evidence suggest that the excessive activation of Toll-like receptor 4 (TLR4) plays an important role in this detrimental lung inflammation. Recently, we developed a unique class of peptide-gold nanoparticle (GNP) hybrids that act as potent nano-inhibitors of TLR4 signaling by modulating the process of endosomal acidification. In this study, we aimed to identify the key physiochemical factors that could further strengthen the anti-inflammatory activity of these nano-inhibitors, including the nanoparticle size, the density of peptides coating the nanoparticle surface, as well as the number of the effective amino acid phenylalanine (F) residues in the peptide sequence. Among these factors, we found that the nanoparticle size could significantly affect the TLR4 inhibition. Specifically, the peptide-GNP hybrids with a GNP core of 20 nm (P12(G20)) exhibited the most potent inhibitory activity on TLR4 activation and its downstream cytokine production among those with a GNP core of 13 nm (P12(G13)) and 5 nm (P12(G5)) in THP-1 cell-derived macrophages. This size-dependent anti-inflammatory effect of the hybrid P12 was also observed in a lipopolysaccharide (LPS)-induced mouse model of ALI. It was found that P12(G20) was superior to P12(G13) in prolonging the survival of mice experiencing lethal LPS challenge, decreasing the acute lung inflammation, and alleviating diffuse alveolar damage in the lungs. Interestingly, P12(G20) could also promote long-term tolerance to endotoxin. Detailed mechanistic studies demonstrated that when compared to the smaller P12(G13), the larger P12(G20) had higher cellular uptake and a stronger endosomal pH buffering capacity, contributing to its enhanced therapeutic effects on reducing TLR4 activation in vitro and in vivo. Overall, this study suggests that nanoparticle size is one key factor determining the anti-inflammatory potency of the peptide-GNP hybrids, and the hybrid P12 may serve as a promising, novel class of nanotherapeutics for modulating TLR signaling to treat ALI/ARDS. STATEMENT OF SIGNIFICANCE: We have developed a new class of nanoparticle-based inhibitors (i.e., peptide-GNP hybrids) targeting TLR4 signaling in macrophages. Through evidence-based engineering of the nanoparticle size, surface peptide ligand density and effective amino acid (phenylalanine, F) chain length, we identified a peptide-GNP hybrid, P12(G20), with enhanced anti-inflammatory activity. Specifically, P12(G20) was more potent in reducing inflammation in THP-1 cell-derived macrophages and in a LPS-induced ALI mouse model. More interestingly, P12(G20) facilitated long-term protection against lethal LPS challenge in vivo and induced endotoxin tolerance in vitro. We anticipate that these new hybrids would serve as the next generation anti-inflammatory nano-therapeutics for the treatment of ALI/ARDS or other acute and chronic inflammatory diseases. Acta Materialia Inc. Published by Elsevier Ltd. 2019-02 2018-12-28 /pmc/articles/PMC8960115/ /pubmed/30597258 http://dx.doi.org/10.1016/j.actbio.2018.12.046 Text en © 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.
spellingShingle Full Length Article
Gao, Wei
Wang, Yulu
Xiong, Ye
Sun, Liya
Wang, Lu
Wang, Kun
Lu, Henry Y.
Bao, Aihua
Turvey, Stuart E.
Li, Qiang
Yang, Hong
Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title_full Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title_fullStr Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title_full_unstemmed Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title_short Size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
title_sort size-dependent anti-inflammatory activity of a peptide-gold nanoparticle hybrid in vitro and in a mouse model of acute lung injury
topic Full Length Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8960115/
https://www.ncbi.nlm.nih.gov/pubmed/30597258
http://dx.doi.org/10.1016/j.actbio.2018.12.046
work_keys_str_mv AT gaowei sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT wangyulu sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT xiongye sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT sunliya sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT wanglu sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT wangkun sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT luhenryy sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT baoaihua sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT turveystuarte sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT liqiang sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury
AT yanghong sizedependentantiinflammatoryactivityofapeptidegoldnanoparticlehybridinvitroandinamousemodelofacutelunginjury