Cargando…

Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells

Adipose tissue (AT) is used extensively in reconstructive and regenerative therapies, but transplanted fat often undergoes cell death, leading to inflammation, calcification, and requirement for further revision surgery. Previously, we have found that mesenchymal progenitor cells within human AT can...

Descripción completa

Detalles Bibliográficos
Autores principales: Rojas-Rodriguez, Raziel, Lujan-Hernandez, Jorge, Min, So Yun, DeSouza, Tiffany, Teebagy, Patrick, Desai, Anand, Tessier, Heather, Slamin, Robert, Siegel-Reamer, Leah, Berg, Cara, Baez, Angel, Lalikos, Janice, Corvera, Silvia
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Mary Ann Liebert, Inc., publishers 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6590775/
https://www.ncbi.nlm.nih.gov/pubmed/30306830
http://dx.doi.org/10.1089/ten.tea.2018.0067
_version_ 1783429623717036032
author Rojas-Rodriguez, Raziel
Lujan-Hernandez, Jorge
Min, So Yun
DeSouza, Tiffany
Teebagy, Patrick
Desai, Anand
Tessier, Heather
Slamin, Robert
Siegel-Reamer, Leah
Berg, Cara
Baez, Angel
Lalikos, Janice
Corvera, Silvia
author_facet Rojas-Rodriguez, Raziel
Lujan-Hernandez, Jorge
Min, So Yun
DeSouza, Tiffany
Teebagy, Patrick
Desai, Anand
Tessier, Heather
Slamin, Robert
Siegel-Reamer, Leah
Berg, Cara
Baez, Angel
Lalikos, Janice
Corvera, Silvia
author_sort Rojas-Rodriguez, Raziel
collection PubMed
description Adipose tissue (AT) is used extensively in reconstructive and regenerative therapies, but transplanted fat often undergoes cell death, leading to inflammation, calcification, and requirement for further revision surgery. Previously, we have found that mesenchymal progenitor cells within human AT can proliferate in three-dimensional culture under proangiogenic conditions. These cells (primed ADipose progenitor cells, PADS) robustly differentiate into adipocytes in vitro (ad-PADS). The goal of this study is to determine whether ad-PADS can form structured AT in vivo, with potential for use in surgical applications. Grafts formed from ad-PADS were compared to grafts formed from AT obtained by liposuction after implantation into nude mice. Graft volume was measured by microcomputed tomography scanning, and the functionality of cells within the graft was assessed by quantifying circulating human adiponectin. The degree of graft vascularization by donor or host vessels and the content of human or mouse adipocytes within the graft were measured using species-specific endothelial and adipocyte-specific quantitative real time polymerase chain reaction probes, and histochemistry with mouse and human-specific lectins. Our results show that ad-PADS grafted subcutaneously into nude mice induce robust vascularization from the host, continue to increase in volume over time, express the human adipocyte marker PLIN1 at levels comparable to human AT, and secrete increasing amounts of human adiponectin into the mouse circulation. In contrast, grafts composed of AT fragments obtained by liposuction become less vascularized, develop regions of calcification and decreased content of PLIN1, and secrete lower amounts of adiponectin per unit volume. Enrichment of liposuction tissue with ad-PADS improves vascularization, indicating that ad-PADS may be proangiogenic. Mechanistically, ad-PADS express an extracellular matrix gene signature that includes elements previously associated with small vessel development (COL4A1). Thus, through the formation of a proangiogenic environment, ad-PADS can form functional AT with capacity for long-term survival, and can potentially be used to improve outcomes in reconstructive and regenerative medicine. IMPACT STATEMENT: This research describes the use of human mesenchymal progenitor cells for generating functional adipose tissue in vivo in a nude mouse model. Further preclinical development of the methods and insights described in this article can lead to therapeutic use of these cells in regenerative and reconstructive medicine.
format Online
Article
Text
id pubmed-6590775
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Mary Ann Liebert, Inc., publishers
record_format MEDLINE/PubMed
spelling pubmed-65907752019-06-24 Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells Rojas-Rodriguez, Raziel Lujan-Hernandez, Jorge Min, So Yun DeSouza, Tiffany Teebagy, Patrick Desai, Anand Tessier, Heather Slamin, Robert Siegel-Reamer, Leah Berg, Cara Baez, Angel Lalikos, Janice Corvera, Silvia Tissue Eng Part A Original Articles Adipose tissue (AT) is used extensively in reconstructive and regenerative therapies, but transplanted fat often undergoes cell death, leading to inflammation, calcification, and requirement for further revision surgery. Previously, we have found that mesenchymal progenitor cells within human AT can proliferate in three-dimensional culture under proangiogenic conditions. These cells (primed ADipose progenitor cells, PADS) robustly differentiate into adipocytes in vitro (ad-PADS). The goal of this study is to determine whether ad-PADS can form structured AT in vivo, with potential for use in surgical applications. Grafts formed from ad-PADS were compared to grafts formed from AT obtained by liposuction after implantation into nude mice. Graft volume was measured by microcomputed tomography scanning, and the functionality of cells within the graft was assessed by quantifying circulating human adiponectin. The degree of graft vascularization by donor or host vessels and the content of human or mouse adipocytes within the graft were measured using species-specific endothelial and adipocyte-specific quantitative real time polymerase chain reaction probes, and histochemistry with mouse and human-specific lectins. Our results show that ad-PADS grafted subcutaneously into nude mice induce robust vascularization from the host, continue to increase in volume over time, express the human adipocyte marker PLIN1 at levels comparable to human AT, and secrete increasing amounts of human adiponectin into the mouse circulation. In contrast, grafts composed of AT fragments obtained by liposuction become less vascularized, develop regions of calcification and decreased content of PLIN1, and secrete lower amounts of adiponectin per unit volume. Enrichment of liposuction tissue with ad-PADS improves vascularization, indicating that ad-PADS may be proangiogenic. Mechanistically, ad-PADS express an extracellular matrix gene signature that includes elements previously associated with small vessel development (COL4A1). Thus, through the formation of a proangiogenic environment, ad-PADS can form functional AT with capacity for long-term survival, and can potentially be used to improve outcomes in reconstructive and regenerative medicine. IMPACT STATEMENT: This research describes the use of human mesenchymal progenitor cells for generating functional adipose tissue in vivo in a nude mouse model. Further preclinical development of the methods and insights described in this article can lead to therapeutic use of these cells in regenerative and reconstructive medicine. Mary Ann Liebert, Inc., publishers 2019-06-01 2019-06-12 /pmc/articles/PMC6590775/ /pubmed/30306830 http://dx.doi.org/10.1089/ten.tea.2018.0067 Text en © Raziel Rojas-Rodriguez et al., 2019; Published by Mary Ann Liebert, Inc. This Open Access article is distributed under the terms of the Creative Commons Attribution Noncommercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are cited.
spellingShingle Original Articles
Rojas-Rodriguez, Raziel
Lujan-Hernandez, Jorge
Min, So Yun
DeSouza, Tiffany
Teebagy, Patrick
Desai, Anand
Tessier, Heather
Slamin, Robert
Siegel-Reamer, Leah
Berg, Cara
Baez, Angel
Lalikos, Janice
Corvera, Silvia
Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title_full Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title_fullStr Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title_full_unstemmed Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title_short Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells
title_sort generation of functional human adipose tissue in mice from primed progenitor cells
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6590775/
https://www.ncbi.nlm.nih.gov/pubmed/30306830
http://dx.doi.org/10.1089/ten.tea.2018.0067
work_keys_str_mv AT rojasrodriguezraziel generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT lujanhernandezjorge generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT minsoyun generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT desouzatiffany generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT teebagypatrick generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT desaianand generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT tessierheather generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT slaminrobert generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT siegelreamerleah generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT bergcara generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT baezangel generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT lalikosjanice generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells
AT corverasilvia generationoffunctionalhumanadiposetissueinmicefromprimedprogenitorcells