Cargando…

Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice

BACKGROUND: Progressive β-cell dysfunction, a major characteristic of type 2 diabetes (T2D), is closely related to the infiltration of inflammatory macrophages within islets. Mesenchymal stem cells (MSCs) have been identified to alleviate β-cell dysfunction by modulating macrophage phenotype in T2D,...

Descripción completa

Detalles Bibliográficos
Autores principales: Xue, Jing, Cheng, Yu, Hao, Haojie, Gao, Jieqing, Yin, Yaqi, Yu, Songyan, Zou, Junyan, Liu, Jiejie, Zhang, Qi, Mu, Yiming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7157805/
https://www.ncbi.nlm.nih.gov/pubmed/32322278
http://dx.doi.org/10.1155/2020/4689798
_version_ 1783522425775849472
author Xue, Jing
Cheng, Yu
Hao, Haojie
Gao, Jieqing
Yin, Yaqi
Yu, Songyan
Zou, Junyan
Liu, Jiejie
Zhang, Qi
Mu, Yiming
author_facet Xue, Jing
Cheng, Yu
Hao, Haojie
Gao, Jieqing
Yin, Yaqi
Yu, Songyan
Zou, Junyan
Liu, Jiejie
Zhang, Qi
Mu, Yiming
author_sort Xue, Jing
collection PubMed
description BACKGROUND: Progressive β-cell dysfunction, a major characteristic of type 2 diabetes (T2D), is closely related to the infiltration of inflammatory macrophages within islets. Mesenchymal stem cells (MSCs) have been identified to alleviate β-cell dysfunction by modulating macrophage phenotype in T2D, but the restoration of β-cells by a single MSC infusion is relatively transient. Decitabine (DAC) has been reported to polarize macrophages towards the anti-inflammatory phenotype at low doses. We therefore investigated whether low-dose decitabine could enhance the antidiabetic effect of MSCs and further promote the restoration of β-cell function. METHODS: We induced a T2D mice model by high-fat diets and streptozotocin (STZ) injection. Mice were divided into five groups: the normal group, the T2D group, the DAC group, the MSC group, and the MSC plus DAC group (MD group). We examined the blood glucose and serum insulin levels of mice 1, 2, and 4 weeks after MSC and/or DAC treatment. Dynamic changes in islets and the phenotype of intraislet macrophages were detected via immunofluorescence. In vitro, we explored the effect of MSCs and DAC on macrophage polarization. RESULTS: The blood glucose and serum insulin levels revealed that DAC prolonged the antidiabetic effect of MSCs to 4 weeks in T2D mice. Immunofluorescence staining demonstrated more sustainable morphological and structural amelioration in islets of the MD group than in the MSC group. Interestingly, further analysis showed more alternatively activated macrophages (M2, anti-inflammatory) and fewer classically activated macrophages (M1, proinflammatory) in islets of the MD group 4 weeks after treatment. An in vitro study demonstrated that DAC together with MSCs further polarized macrophages from the M1 to M2 phenotype via the PI3K/AKT pathway. CONCLUSION: These data unveiled that DAC prolonged the antidiabetic effect of MSCs and promoted sustainable β-cell restoration, possibly by modulating the macrophage phenotype. Our results offer a preferable therapeutic strategy for T2D.
format Online
Article
Text
id pubmed-7157805
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-71578052020-04-22 Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice Xue, Jing Cheng, Yu Hao, Haojie Gao, Jieqing Yin, Yaqi Yu, Songyan Zou, Junyan Liu, Jiejie Zhang, Qi Mu, Yiming Stem Cells Int Research Article BACKGROUND: Progressive β-cell dysfunction, a major characteristic of type 2 diabetes (T2D), is closely related to the infiltration of inflammatory macrophages within islets. Mesenchymal stem cells (MSCs) have been identified to alleviate β-cell dysfunction by modulating macrophage phenotype in T2D, but the restoration of β-cells by a single MSC infusion is relatively transient. Decitabine (DAC) has been reported to polarize macrophages towards the anti-inflammatory phenotype at low doses. We therefore investigated whether low-dose decitabine could enhance the antidiabetic effect of MSCs and further promote the restoration of β-cell function. METHODS: We induced a T2D mice model by high-fat diets and streptozotocin (STZ) injection. Mice were divided into five groups: the normal group, the T2D group, the DAC group, the MSC group, and the MSC plus DAC group (MD group). We examined the blood glucose and serum insulin levels of mice 1, 2, and 4 weeks after MSC and/or DAC treatment. Dynamic changes in islets and the phenotype of intraislet macrophages were detected via immunofluorescence. In vitro, we explored the effect of MSCs and DAC on macrophage polarization. RESULTS: The blood glucose and serum insulin levels revealed that DAC prolonged the antidiabetic effect of MSCs to 4 weeks in T2D mice. Immunofluorescence staining demonstrated more sustainable morphological and structural amelioration in islets of the MD group than in the MSC group. Interestingly, further analysis showed more alternatively activated macrophages (M2, anti-inflammatory) and fewer classically activated macrophages (M1, proinflammatory) in islets of the MD group 4 weeks after treatment. An in vitro study demonstrated that DAC together with MSCs further polarized macrophages from the M1 to M2 phenotype via the PI3K/AKT pathway. CONCLUSION: These data unveiled that DAC prolonged the antidiabetic effect of MSCs and promoted sustainable β-cell restoration, possibly by modulating the macrophage phenotype. Our results offer a preferable therapeutic strategy for T2D. Hindawi 2020-04-03 /pmc/articles/PMC7157805/ /pubmed/32322278 http://dx.doi.org/10.1155/2020/4689798 Text en Copyright © 2020 Jing Xue et al. http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Xue, Jing
Cheng, Yu
Hao, Haojie
Gao, Jieqing
Yin, Yaqi
Yu, Songyan
Zou, Junyan
Liu, Jiejie
Zhang, Qi
Mu, Yiming
Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title_full Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title_fullStr Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title_full_unstemmed Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title_short Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice
title_sort low-dose decitabine assists human umbilical cord-derived mesenchymal stem cells in protecting β cells via the modulation of the macrophage phenotype in type 2 diabetic mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7157805/
https://www.ncbi.nlm.nih.gov/pubmed/32322278
http://dx.doi.org/10.1155/2020/4689798
work_keys_str_mv AT xuejing lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT chengyu lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT haohaojie lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT gaojieqing lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT yinyaqi lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT yusongyan lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT zoujunyan lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT liujiejie lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT zhangqi lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice
AT muyiming lowdosedecitabineassistshumanumbilicalcordderivedmesenchymalstemcellsinprotectingbcellsviathemodulationofthemacrophagephenotypeintype2diabeticmice