Cargando…

Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model

The use of carbon nanotubes has increased in the past few decades. Carbon nanotubes are implicated in the pathogenesis of pulmonary sarcoidosis, a chronic granulomatous inflammatory condition. We developed a murine model of chronic granulomatous inflammation using multiwall carbon nanotubes (MWCNT)...

Descripción completa

Detalles Bibliográficos
Autores principales: Soliman, Eman, Bhalla, Sophia, Elhassanny, Ahmed E. M., Malur, Anagha, Ogburn, David, Leffler, Nancy, Malur, Achut G., Thomassen, Mary Jane
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8744594/
https://www.ncbi.nlm.nih.gov/pubmed/35008476
http://dx.doi.org/10.3390/ijms23010047
_version_ 1784630142031626240
author Soliman, Eman
Bhalla, Sophia
Elhassanny, Ahmed E. M.
Malur, Anagha
Ogburn, David
Leffler, Nancy
Malur, Achut G.
Thomassen, Mary Jane
author_facet Soliman, Eman
Bhalla, Sophia
Elhassanny, Ahmed E. M.
Malur, Anagha
Ogburn, David
Leffler, Nancy
Malur, Achut G.
Thomassen, Mary Jane
author_sort Soliman, Eman
collection PubMed
description The use of carbon nanotubes has increased in the past few decades. Carbon nanotubes are implicated in the pathogenesis of pulmonary sarcoidosis, a chronic granulomatous inflammatory condition. We developed a murine model of chronic granulomatous inflammation using multiwall carbon nanotubes (MWCNT) to investigate mechanisms of granuloma formation. Using this model, we demonstrated that myeloid deficiency of ATP-binding cassette (ABC) cholesterol transporter (ABCG1) promotes granuloma formation and fibrosis with MWCNT instillation; however, the mechanism remains unclear. Our previous studies showed that MWCNT induced apoptosis in bronchoalveolar lavage (BAL) cells of wild-type (C57BL/6) mice. Given that continual apoptosis causes persistent severe lung inflammation, we hypothesized that ABCG1 deficiency would increase MWCNT-induced apoptosis thereby promoting granulomatous inflammation and fibrosis. To test our hypothesis, we utilized myeloid-specific ABCG1 knockout (ABCG1 KO) mice. Our results demonstrate that MWCNT instillation enhances pulmonary fibrosis in ABCG1 KO mice compared to wild-type controls. Enhanced fibrosis is indicated by increased trichrome staining and transforming growth factor-beta (TGF-β) expression in lungs, together with an increased expression of TGF-β related signaling molecules, interleukin-13 (IL-13) and Smad-3. MWCNT induced more apoptosis in BAL cells of ABCG1 KO mice. Initiation of apoptosis is most likely mediated by the extrinsic pathway since caspase 8 activity and Fas expression are significantly higher in MWCNT instilled ABCG1 KO mice compared to the wild type. In addition, TUNEL staining shows that ABCG1 KO mice instilled with MWCNT have a higher percentage of TUNEL positive BAL cells and more efferocytosis than the WT control. Furthermore, BAL cells of ABCG1 KO mice instilled with MWCNT exhibit an increase in efferocytosis markers, milk fat globule-EGF factor 8 (MFG-E8) and integrin β3. Therefore, our observations suggest that ABCG1 deficiency promotes pulmonary fibrosis by MWCNT, and this effect may be due to an increase in apoptosis and efferocytosis in BAL cells.
format Online
Article
Text
id pubmed-8744594
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-87445942022-01-11 Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model Soliman, Eman Bhalla, Sophia Elhassanny, Ahmed E. M. Malur, Anagha Ogburn, David Leffler, Nancy Malur, Achut G. Thomassen, Mary Jane Int J Mol Sci Article The use of carbon nanotubes has increased in the past few decades. Carbon nanotubes are implicated in the pathogenesis of pulmonary sarcoidosis, a chronic granulomatous inflammatory condition. We developed a murine model of chronic granulomatous inflammation using multiwall carbon nanotubes (MWCNT) to investigate mechanisms of granuloma formation. Using this model, we demonstrated that myeloid deficiency of ATP-binding cassette (ABC) cholesterol transporter (ABCG1) promotes granuloma formation and fibrosis with MWCNT instillation; however, the mechanism remains unclear. Our previous studies showed that MWCNT induced apoptosis in bronchoalveolar lavage (BAL) cells of wild-type (C57BL/6) mice. Given that continual apoptosis causes persistent severe lung inflammation, we hypothesized that ABCG1 deficiency would increase MWCNT-induced apoptosis thereby promoting granulomatous inflammation and fibrosis. To test our hypothesis, we utilized myeloid-specific ABCG1 knockout (ABCG1 KO) mice. Our results demonstrate that MWCNT instillation enhances pulmonary fibrosis in ABCG1 KO mice compared to wild-type controls. Enhanced fibrosis is indicated by increased trichrome staining and transforming growth factor-beta (TGF-β) expression in lungs, together with an increased expression of TGF-β related signaling molecules, interleukin-13 (IL-13) and Smad-3. MWCNT induced more apoptosis in BAL cells of ABCG1 KO mice. Initiation of apoptosis is most likely mediated by the extrinsic pathway since caspase 8 activity and Fas expression are significantly higher in MWCNT instilled ABCG1 KO mice compared to the wild type. In addition, TUNEL staining shows that ABCG1 KO mice instilled with MWCNT have a higher percentage of TUNEL positive BAL cells and more efferocytosis than the WT control. Furthermore, BAL cells of ABCG1 KO mice instilled with MWCNT exhibit an increase in efferocytosis markers, milk fat globule-EGF factor 8 (MFG-E8) and integrin β3. Therefore, our observations suggest that ABCG1 deficiency promotes pulmonary fibrosis by MWCNT, and this effect may be due to an increase in apoptosis and efferocytosis in BAL cells. MDPI 2021-12-21 /pmc/articles/PMC8744594/ /pubmed/35008476 http://dx.doi.org/10.3390/ijms23010047 Text en © 2021 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Soliman, Eman
Bhalla, Sophia
Elhassanny, Ahmed E. M.
Malur, Anagha
Ogburn, David
Leffler, Nancy
Malur, Achut G.
Thomassen, Mary Jane
Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title_full Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title_fullStr Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title_full_unstemmed Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title_short Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model
title_sort myeloid abcg1 deficiency enhances apoptosis and initiates efferocytosis in bronchoalveolar lavage cells of murine multi-walled carbon nanotube-induced granuloma model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8744594/
https://www.ncbi.nlm.nih.gov/pubmed/35008476
http://dx.doi.org/10.3390/ijms23010047
work_keys_str_mv AT solimaneman myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT bhallasophia myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT elhassannyahmedem myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT maluranagha myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT ogburndavid myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT lefflernancy myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT malurachutg myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel
AT thomassenmaryjane myeloidabcg1deficiencyenhancesapoptosisandinitiatesefferocytosisinbronchoalveolarlavagecellsofmurinemultiwalledcarbonnanotubeinducedgranulomamodel