Cargando…

Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats

BACKGROUND: Mesenchymal stem cells (MSCs) exert anti-diabetic effects and improve long-term complications via secretory effects that regulate macrophage polarisation and attenuate inflammation. Enhancing the efficacy of MSCs needs to be explored further. The in vitro culture microenvironment influen...

Descripción completa

Detalles Bibliográficos
Autores principales: Su, Wanlu, Yu, Songyan, Yin, Yaqi, Li, Bing, Xue, Jing, Wang, Jie, Gu, Yulin, Zhang, Haixia, Lyu, Zhaohui, Mu, Yiming, Cheng, Yu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9389728/
https://www.ncbi.nlm.nih.gov/pubmed/35986406
http://dx.doi.org/10.1186/s13287-022-03114-5
_version_ 1784770520539987968
author Su, Wanlu
Yu, Songyan
Yin, Yaqi
Li, Bing
Xue, Jing
Wang, Jie
Gu, Yulin
Zhang, Haixia
Lyu, Zhaohui
Mu, Yiming
Cheng, Yu
author_facet Su, Wanlu
Yu, Songyan
Yin, Yaqi
Li, Bing
Xue, Jing
Wang, Jie
Gu, Yulin
Zhang, Haixia
Lyu, Zhaohui
Mu, Yiming
Cheng, Yu
author_sort Su, Wanlu
collection PubMed
description BACKGROUND: Mesenchymal stem cells (MSCs) exert anti-diabetic effects and improve long-term complications via secretory effects that regulate macrophage polarisation and attenuate inflammation. Enhancing the efficacy of MSCs needs to be explored further. The in vitro culture microenvironment influences the secretory profile of MSCs. Therefore, we hypothesised that a diabetic microenvironment would promote the secretion of cytokines responsible for macrophage polarisation, further attenuating systemic inflammation and enhancing the effects of MSCs on type 2 diabetes (T2D) and long-term diabetic complications. METHODS: Preconditioned adipose-derived mesenchymal stem cells (pre-ADSCs) were obtained after co-cultivating ADSCs in a diabetic metabolic environment (including high sugar, advanced glycation end-product, and lipopolysaccharides). The regulatory effects of pre-ADSCs on macrophages were observed in vitro. A T2D rat model was induced with a high-fat diet for 32 weeks combined with an intraperitoneal injection of streptozotocin. Sprague–Dawley (SD) rats were divided into four groups: normal group, diabetes without treatment group (PBS), ADSC treatment group, and pre-ADSC treatment group. ADSCs and pre-ADSCs were intravenously administered weekly to SD rats for 6 months, and then glucose homeostasis and long-term diabetic complications were evaluated in each group. RESULTS: The secretion of cytokines related to M2 macrophage polarisation (IL-6, MCP-1, etc.) was increased in the pre-ADSC group in the in vitro model. Pre-ADSC treatment significantly maintained blood glucose homeostasis, reduced insulin resistance, promoted islet regeneration, and ameliorated the complications related to diabetes in rats (chronic kidney disease, non-alcoholic steatohepatitis, lung fibrosis, and cataract) compared to the ADSC group (P < 0.05). Additionally, the number of anti-inflammatory M2 macrophage phenotypes was enhanced in tissues following pre-ADSC injections. Moreover, the expression of pro-inflammatory genes (iNOS, TNF-α, IL-1β) was reduced whereas that of anti-inflammatory genes (Arg1, CD206, and Il-10) was increased after cultivation with pre-ADSCs. CONCLUSION: Diabetic microenvironment-preconditioned ADSCs effectively strengthen the capacity against inflammation and modulate the progress of long-term T2D complications. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-022-03114-5.
format Online
Article
Text
id pubmed-9389728
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-93897282022-08-20 Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats Su, Wanlu Yu, Songyan Yin, Yaqi Li, Bing Xue, Jing Wang, Jie Gu, Yulin Zhang, Haixia Lyu, Zhaohui Mu, Yiming Cheng, Yu Stem Cell Res Ther Research BACKGROUND: Mesenchymal stem cells (MSCs) exert anti-diabetic effects and improve long-term complications via secretory effects that regulate macrophage polarisation and attenuate inflammation. Enhancing the efficacy of MSCs needs to be explored further. The in vitro culture microenvironment influences the secretory profile of MSCs. Therefore, we hypothesised that a diabetic microenvironment would promote the secretion of cytokines responsible for macrophage polarisation, further attenuating systemic inflammation and enhancing the effects of MSCs on type 2 diabetes (T2D) and long-term diabetic complications. METHODS: Preconditioned adipose-derived mesenchymal stem cells (pre-ADSCs) were obtained after co-cultivating ADSCs in a diabetic metabolic environment (including high sugar, advanced glycation end-product, and lipopolysaccharides). The regulatory effects of pre-ADSCs on macrophages were observed in vitro. A T2D rat model was induced with a high-fat diet for 32 weeks combined with an intraperitoneal injection of streptozotocin. Sprague–Dawley (SD) rats were divided into four groups: normal group, diabetes without treatment group (PBS), ADSC treatment group, and pre-ADSC treatment group. ADSCs and pre-ADSCs were intravenously administered weekly to SD rats for 6 months, and then glucose homeostasis and long-term diabetic complications were evaluated in each group. RESULTS: The secretion of cytokines related to M2 macrophage polarisation (IL-6, MCP-1, etc.) was increased in the pre-ADSC group in the in vitro model. Pre-ADSC treatment significantly maintained blood glucose homeostasis, reduced insulin resistance, promoted islet regeneration, and ameliorated the complications related to diabetes in rats (chronic kidney disease, non-alcoholic steatohepatitis, lung fibrosis, and cataract) compared to the ADSC group (P < 0.05). Additionally, the number of anti-inflammatory M2 macrophage phenotypes was enhanced in tissues following pre-ADSC injections. Moreover, the expression of pro-inflammatory genes (iNOS, TNF-α, IL-1β) was reduced whereas that of anti-inflammatory genes (Arg1, CD206, and Il-10) was increased after cultivation with pre-ADSCs. CONCLUSION: Diabetic microenvironment-preconditioned ADSCs effectively strengthen the capacity against inflammation and modulate the progress of long-term T2D complications. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-022-03114-5. BioMed Central 2022-08-19 /pmc/articles/PMC9389728/ /pubmed/35986406 http://dx.doi.org/10.1186/s13287-022-03114-5 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Su, Wanlu
Yu, Songyan
Yin, Yaqi
Li, Bing
Xue, Jing
Wang, Jie
Gu, Yulin
Zhang, Haixia
Lyu, Zhaohui
Mu, Yiming
Cheng, Yu
Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title_full Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title_fullStr Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title_full_unstemmed Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title_short Diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
title_sort diabetic microenvironment preconditioning of adipose tissue-derived mesenchymal stem cells enhances their anti-diabetic, anti-long-term complications, and anti-inflammatory effects in type 2 diabetic rats
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9389728/
https://www.ncbi.nlm.nih.gov/pubmed/35986406
http://dx.doi.org/10.1186/s13287-022-03114-5
work_keys_str_mv AT suwanlu diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT yusongyan diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT yinyaqi diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT libing diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT xuejing diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT wangjie diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT guyulin diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT zhanghaixia diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT lyuzhaohui diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT muyiming diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats
AT chengyu diabeticmicroenvironmentpreconditioningofadiposetissuederivedmesenchymalstemcellsenhancestheirantidiabeticantilongtermcomplicationsandantiinflammatoryeffectsintype2diabeticrats