Cargando…

Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury

BACKGROUND: Despite advances in neonatal care, bronchopulmonary dysplasia (BPD) remains a significant contributor to infant mortality and morbidity. While human amnion epithelial cells (hAECs) have shown promise in small and large animal models of BPD, there is scarce information on long-term benefi...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhu, Dandan, Tan, Jean, Maleken, Amina S., Muljadi, Ruth, Chan, Siow T., Lau, Sin N., Elgass, Kirstin, Leaw, Bryan, Mockler, Joanne, Chambers, Daniel, Leeman, Kristen T., Kim, Carla F., Wallace, Euan M., Lim, Rebecca
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5681809/
https://www.ncbi.nlm.nih.gov/pubmed/29126435
http://dx.doi.org/10.1186/s13287-017-0689-9
_version_ 1783277986001190912
author Zhu, Dandan
Tan, Jean
Maleken, Amina S.
Muljadi, Ruth
Chan, Siow T.
Lau, Sin N.
Elgass, Kirstin
Leaw, Bryan
Mockler, Joanne
Chambers, Daniel
Leeman, Kristen T.
Kim, Carla F.
Wallace, Euan M.
Lim, Rebecca
author_facet Zhu, Dandan
Tan, Jean
Maleken, Amina S.
Muljadi, Ruth
Chan, Siow T.
Lau, Sin N.
Elgass, Kirstin
Leaw, Bryan
Mockler, Joanne
Chambers, Daniel
Leeman, Kristen T.
Kim, Carla F.
Wallace, Euan M.
Lim, Rebecca
author_sort Zhu, Dandan
collection PubMed
description BACKGROUND: Despite advances in neonatal care, bronchopulmonary dysplasia (BPD) remains a significant contributor to infant mortality and morbidity. While human amnion epithelial cells (hAECs) have shown promise in small and large animal models of BPD, there is scarce information on long-term benefit and clinically relevant questions surrounding administration strategy remain unanswered. In assessing the therapeutic potential of hAECs, we investigated the impact of cell dosage, administration routes and timing of treatment in a pre-clinical model of BPD. METHODS: Lipopolysaccharide was introduced intra-amniotically at day 16 of pregnancy prior to exposure to 65% oxygen (hyperoxia) at birth. hAECs were administered either 12 hours (early) or 4 days (late) after hyperoxia commenced. Collective lung tissues were subjected to histological analysis, multikine ELISA for inflammatory cytokines, FACS for immune cell populations and 3D lung stem cell culture at neonatal stage (postnatal day 7 and 14). Invasive lung function test and echocardiography were applied at 6 and 10 weeks of age. RESULTS: hAECs improved the tissue-to-airspace ratio and septal crest density in a dose-dependent manner, regardless of administration route. Early administration of hAECs, coinciding with the commencement of postnatal hyperoxia, was associated with reduced macrophages, dendritic cells and natural killer cells. This was not the case if hAECs were administered when lung injury was established. Fittingly, early hAEC treatment was more efficacious in reducing interleukin-1β, tumour necrosis factor alpha and monocyte chemoattractant protein-1 levels. Early hAEC treatment was also associated with reduced airway hyper-responsiveness and normalisation of pressure–volume loops. Pulmonary hypertension and right ventricle hypertrophy were also prevented in the early hAEC treatment group, and this persisted until 10 weeks of age. CONCLUSIONS: Early hAEC treatment appears to be advantageous over late treatment. There was no difference in efficacy between intravenous and intratracheal administration. The benefits of hAEC administration resulted in long-term improvements in cardiorespiratory function. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13287-017-0689-9) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5681809
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-56818092017-11-17 Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury Zhu, Dandan Tan, Jean Maleken, Amina S. Muljadi, Ruth Chan, Siow T. Lau, Sin N. Elgass, Kirstin Leaw, Bryan Mockler, Joanne Chambers, Daniel Leeman, Kristen T. Kim, Carla F. Wallace, Euan M. Lim, Rebecca Stem Cell Res Ther Research BACKGROUND: Despite advances in neonatal care, bronchopulmonary dysplasia (BPD) remains a significant contributor to infant mortality and morbidity. While human amnion epithelial cells (hAECs) have shown promise in small and large animal models of BPD, there is scarce information on long-term benefit and clinically relevant questions surrounding administration strategy remain unanswered. In assessing the therapeutic potential of hAECs, we investigated the impact of cell dosage, administration routes and timing of treatment in a pre-clinical model of BPD. METHODS: Lipopolysaccharide was introduced intra-amniotically at day 16 of pregnancy prior to exposure to 65% oxygen (hyperoxia) at birth. hAECs were administered either 12 hours (early) or 4 days (late) after hyperoxia commenced. Collective lung tissues were subjected to histological analysis, multikine ELISA for inflammatory cytokines, FACS for immune cell populations and 3D lung stem cell culture at neonatal stage (postnatal day 7 and 14). Invasive lung function test and echocardiography were applied at 6 and 10 weeks of age. RESULTS: hAECs improved the tissue-to-airspace ratio and septal crest density in a dose-dependent manner, regardless of administration route. Early administration of hAECs, coinciding with the commencement of postnatal hyperoxia, was associated with reduced macrophages, dendritic cells and natural killer cells. This was not the case if hAECs were administered when lung injury was established. Fittingly, early hAEC treatment was more efficacious in reducing interleukin-1β, tumour necrosis factor alpha and monocyte chemoattractant protein-1 levels. Early hAEC treatment was also associated with reduced airway hyper-responsiveness and normalisation of pressure–volume loops. Pulmonary hypertension and right ventricle hypertrophy were also prevented in the early hAEC treatment group, and this persisted until 10 weeks of age. CONCLUSIONS: Early hAEC treatment appears to be advantageous over late treatment. There was no difference in efficacy between intravenous and intratracheal administration. The benefits of hAEC administration resulted in long-term improvements in cardiorespiratory function. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13287-017-0689-9) contains supplementary material, which is available to authorized users. BioMed Central 2017-11-10 /pmc/articles/PMC5681809/ /pubmed/29126435 http://dx.doi.org/10.1186/s13287-017-0689-9 Text en © The Author(s). 2017 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided 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 Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Zhu, Dandan
Tan, Jean
Maleken, Amina S.
Muljadi, Ruth
Chan, Siow T.
Lau, Sin N.
Elgass, Kirstin
Leaw, Bryan
Mockler, Joanne
Chambers, Daniel
Leeman, Kristen T.
Kim, Carla F.
Wallace, Euan M.
Lim, Rebecca
Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title_full Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title_fullStr Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title_full_unstemmed Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title_short Human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
title_sort human amnion cells reverse acute and chronic pulmonary damage in experimental neonatal lung injury
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5681809/
https://www.ncbi.nlm.nih.gov/pubmed/29126435
http://dx.doi.org/10.1186/s13287-017-0689-9
work_keys_str_mv AT zhudandan humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT tanjean humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT malekenaminas humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT muljadiruth humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT chansiowt humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT lausinn humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT elgasskirstin humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT leawbryan humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT mocklerjoanne humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT chambersdaniel humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT leemankristent humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT kimcarlaf humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT wallaceeuanm humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury
AT limrebecca humanamnioncellsreverseacuteandchronicpulmonarydamageinexperimentalneonatallunginjury