Cargando…

The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD

Although the positive effects of recombinant fibroblast growth factor-2 (rFGF-2) in chronic obstructive pulmonary disease (COPD) have been implicated in previous studies, knowledge of its role in COPD remains limited. The mechanism of FGF2 in a COPD mouse model and the therapeutic potential of rFGF-...

Descripción completa

Detalles Bibliográficos
Autores principales: Kim, You-Sun, Hong, Goohyeon, Kim, Doh Hyung, Kim, Young Min, Kim, Yoon-Keun, Oh, Yeon-Mok, Jee, Young-Koo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6235987/
https://www.ncbi.nlm.nih.gov/pubmed/30429461
http://dx.doi.org/10.1038/s12276-018-0178-y
_version_ 1783370947809509376
author Kim, You-Sun
Hong, Goohyeon
Kim, Doh Hyung
Kim, Young Min
Kim, Yoon-Keun
Oh, Yeon-Mok
Jee, Young-Koo
author_facet Kim, You-Sun
Hong, Goohyeon
Kim, Doh Hyung
Kim, Young Min
Kim, Yoon-Keun
Oh, Yeon-Mok
Jee, Young-Koo
author_sort Kim, You-Sun
collection PubMed
description Although the positive effects of recombinant fibroblast growth factor-2 (rFGF-2) in chronic obstructive pulmonary disease (COPD) have been implicated in previous studies, knowledge of its role in COPD remains limited. The mechanism of FGF2 in a COPD mouse model and the therapeutic potential of rFGF-2 were investigated in COPD. The mechanism and protective effects of rFGF-2 were evaluated in cigarette smoke-exposed or elastase-induced COPD animal models. Inflammation was assessed in alveolar cells and lung tissues from mice. FGF-2 was decreased in the lungs of cigarette smoke-exposed mice. Intranasal use of rFGF-2 significantly reduced macrophage-dominant inflammation and alveolar destruction in the lungs. In the elastase-induced emphysema model, rFGF-2 improved regeneration of the lungs. In humans, plasma FGF-2 was decreased significantly in COPD compared with normal subjects (10 subjects, P = 0.037). The safety and efficacy of inhaled rFGF-2 use was examined in COPD patients, along with changes in respiratory symptoms and pulmonary function. A 2-week treatment with inhaled rFGF-2 in COPD (n = 6) resulted in significantly improved respiratory symptoms compared with baseline levels (P < 0.05); however, the results were not significant compared with the placebo. The pulmonary function test results of COPD improved numerically compared with those in the placebo, but the difference was not statistically significant. No serious adverse events occurred during treatment with inhaled rFGF-2. The loss of FGF-2 production is an important mechanism in the development of COPD. Inhaling rFGF-2 may be a new therapeutic option for patients with COPD because rFGF-2 decreases inflammation in lungs exposed to cigarette smoke.
format Online
Article
Text
id pubmed-6235987
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-62359872018-11-16 The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD Kim, You-Sun Hong, Goohyeon Kim, Doh Hyung Kim, Young Min Kim, Yoon-Keun Oh, Yeon-Mok Jee, Young-Koo Exp Mol Med Article Although the positive effects of recombinant fibroblast growth factor-2 (rFGF-2) in chronic obstructive pulmonary disease (COPD) have been implicated in previous studies, knowledge of its role in COPD remains limited. The mechanism of FGF2 in a COPD mouse model and the therapeutic potential of rFGF-2 were investigated in COPD. The mechanism and protective effects of rFGF-2 were evaluated in cigarette smoke-exposed or elastase-induced COPD animal models. Inflammation was assessed in alveolar cells and lung tissues from mice. FGF-2 was decreased in the lungs of cigarette smoke-exposed mice. Intranasal use of rFGF-2 significantly reduced macrophage-dominant inflammation and alveolar destruction in the lungs. In the elastase-induced emphysema model, rFGF-2 improved regeneration of the lungs. In humans, plasma FGF-2 was decreased significantly in COPD compared with normal subjects (10 subjects, P = 0.037). The safety and efficacy of inhaled rFGF-2 use was examined in COPD patients, along with changes in respiratory symptoms and pulmonary function. A 2-week treatment with inhaled rFGF-2 in COPD (n = 6) resulted in significantly improved respiratory symptoms compared with baseline levels (P < 0.05); however, the results were not significant compared with the placebo. The pulmonary function test results of COPD improved numerically compared with those in the placebo, but the difference was not statistically significant. No serious adverse events occurred during treatment with inhaled rFGF-2. The loss of FGF-2 production is an important mechanism in the development of COPD. Inhaling rFGF-2 may be a new therapeutic option for patients with COPD because rFGF-2 decreases inflammation in lungs exposed to cigarette smoke. Nature Publishing Group UK 2018-11-14 /pmc/articles/PMC6235987/ /pubmed/30429461 http://dx.doi.org/10.1038/s12276-018-0178-y Text en © The Author(s) 2018 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Kim, You-Sun
Hong, Goohyeon
Kim, Doh Hyung
Kim, Young Min
Kim, Yoon-Keun
Oh, Yeon-Mok
Jee, Young-Koo
The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title_full The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title_fullStr The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title_full_unstemmed The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title_short The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD
title_sort role of fgf-2 in smoke-induced emphysema and the therapeutic potential of recombinant fgf-2 in patients with copd
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6235987/
https://www.ncbi.nlm.nih.gov/pubmed/30429461
http://dx.doi.org/10.1038/s12276-018-0178-y
work_keys_str_mv AT kimyousun theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT honggoohyeon theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimdohhyung theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimyoungmin theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimyoonkeun theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT ohyeonmok theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT jeeyoungkoo theroleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimyousun roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT honggoohyeon roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimdohhyung roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimyoungmin roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT kimyoonkeun roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT ohyeonmok roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd
AT jeeyoungkoo roleoffgf2insmokeinducedemphysemaandthetherapeuticpotentialofrecombinantfgf2inpatientswithcopd