Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue and its derived mesenchymal stromal cells.

Q4 Neuroscience Vascular Cell Pub Date : 2016-08-18 eCollection Date: 2016-01-01 DOI:10.1186/s13221-016-0037-3
Valentina Ceserani, Anna Ferri, Angiola Berenzi, Anna Benetti, Emilio Ciusani, Luisa Pascucci, Cinzia Bazzucchi, Valentina Coccè, Arianna Bonomi, Augusto Pessina, Erica Ghezzi, Offer Zeira, Piero Ceccarelli, Silvia Versari, Carlo Tremolada, Giulio Alessandri
{"title":"Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue and its derived mesenchymal stromal cells.","authors":"Valentina Ceserani, Anna Ferri, Angiola Berenzi, Anna Benetti, Emilio Ciusani, Luisa Pascucci, Cinzia Bazzucchi, Valentina Coccè, Arianna Bonomi, Augusto Pessina, Erica Ghezzi, Offer Zeira, Piero Ceccarelli, Silvia Versari, Carlo Tremolada, Giulio Alessandri","doi":"10.1186/s13221-016-0037-3","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for advanced cell-based therapies. They are routinely obtained enzymatically from fat lipoaspirate (LP) as SVF, and may undergo prolonged ex vivo expansion, with significant senescence and decline in multipotency. Besides, these techniques have complex regulatory issues, thus incurring in the compelling requirements of GMP guidelines. Hence, availability of a minimally manipulated, autologous adipose tissue would have remarkable biomedical and clinical relevance. For this reason, a new device, named Lipogems® (LG), has been developed. This ready-to-use adipose tissue cell derivate has been shown to have in vivo efficacy upon transplantation for ischemic and inflammatory diseases. To broaden our knowledge, we here investigated the angiogenic and anti-inflammatory properties of LG and its derived MSC (LG-MSCs) population.</p><p><strong>Methods: </strong>Human LG samples and their LG-MSCs were analyzed by immunohistochemistry for pericyte, endothelial and mesenchymal stromal cell marker expression. Angiogenesis was investigated testing the conditioned media (CM) of LG (LG-CM) and LG-MSCs (LG-MSCs-CM) on cultured endothelial cells (HUVECs), evaluating proliferation, cord formation, and the expression of the adhesion molecules (AM) VCAM-1 and ICAM-1. The macrophage cell line U937 was used to evaluate the anti-inflammatory properties, such as migration, adhesion on HUVECs, and release of RANTES and MCP-1.</p><p><strong>Results: </strong>Our results indicate that LG contained a very high number of mesenchymal cells expressing NG2 and CD146 (both pericyte markers) together with an abundant microvascular endothelial cell (mEC) population. Substantially, both LG-CM and LG-MSC-CM increased cord formation, inhibited endothelial ICAM-1 and VCAM-1 expression following TNFα stimulation, and slightly improved HUVEC proliferation. The addition of LG-CM and LG-MSC-CM strongly inhibited U937 migration upon stimulation with the chemokine MCP-1, reduced their adhesion on HUVECs and significantly suppressed the release of RANTES and MCP-1.</p><p><strong>Conclusions: </strong>Our data indicate that LG micro-fragmented adipose tissue retains either per se, or in its embedded MSCs content, the capacity to induce vascular stabilization and to inhibit several macrophage functions involved in inflammation.</p>","PeriodicalId":23948,"journal":{"name":"Vascular Cell","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2016-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1186/s13221-016-0037-3","citationCount":"59","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Vascular Cell","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13221-016-0037-3","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2016/1/1 0:00:00","PubModel":"eCollection","JCR":"Q4","JCRName":"Neuroscience","Score":null,"Total":0}
引用次数: 59

Abstract

Background: Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for advanced cell-based therapies. They are routinely obtained enzymatically from fat lipoaspirate (LP) as SVF, and may undergo prolonged ex vivo expansion, with significant senescence and decline in multipotency. Besides, these techniques have complex regulatory issues, thus incurring in the compelling requirements of GMP guidelines. Hence, availability of a minimally manipulated, autologous adipose tissue would have remarkable biomedical and clinical relevance. For this reason, a new device, named Lipogems® (LG), has been developed. This ready-to-use adipose tissue cell derivate has been shown to have in vivo efficacy upon transplantation for ischemic and inflammatory diseases. To broaden our knowledge, we here investigated the angiogenic and anti-inflammatory properties of LG and its derived MSC (LG-MSCs) population.

Methods: Human LG samples and their LG-MSCs were analyzed by immunohistochemistry for pericyte, endothelial and mesenchymal stromal cell marker expression. Angiogenesis was investigated testing the conditioned media (CM) of LG (LG-CM) and LG-MSCs (LG-MSCs-CM) on cultured endothelial cells (HUVECs), evaluating proliferation, cord formation, and the expression of the adhesion molecules (AM) VCAM-1 and ICAM-1. The macrophage cell line U937 was used to evaluate the anti-inflammatory properties, such as migration, adhesion on HUVECs, and release of RANTES and MCP-1.

Results: Our results indicate that LG contained a very high number of mesenchymal cells expressing NG2 and CD146 (both pericyte markers) together with an abundant microvascular endothelial cell (mEC) population. Substantially, both LG-CM and LG-MSC-CM increased cord formation, inhibited endothelial ICAM-1 and VCAM-1 expression following TNFα stimulation, and slightly improved HUVEC proliferation. The addition of LG-CM and LG-MSC-CM strongly inhibited U937 migration upon stimulation with the chemokine MCP-1, reduced their adhesion on HUVECs and significantly suppressed the release of RANTES and MCP-1.

Conclusions: Our data indicate that LG micro-fragmented adipose tissue retains either per se, or in its embedded MSCs content, the capacity to induce vascular stabilization and to inhibit several macrophage functions involved in inflammation.

Abstract Image

Abstract Image

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
微碎片脂肪组织及其衍生间充质间质细胞的血管生成和抗炎特性。
背景:脂肪源性间充质基质细胞(Ad-MSCs)是一种很有前途的先进细胞治疗工具。它们通常从抽脂(LP)中酶促获得SVF,并且可能经历长时间的体外扩增,具有明显的衰老和多能性下降。此外,这些技术有复杂的监管问题,因此在GMP指南的强制要求。因此,微创操作的自体脂肪组织的可用性将具有显著的生物医学和临床意义。为此,开发了一种名为Lipogems®(LG)的新设备。这种现成的脂肪组织细胞衍生物已被证明在体内对缺血性和炎症性疾病的移植有疗效。为了扩大我们的知识,我们在这里研究了LG及其衍生的MSC (LG- mscs)群体的血管生成和抗炎特性。方法:采用免疫组化方法对人LG样品及其LG- mscs进行周细胞、内皮细胞和间充质间质细胞标志物的表达分析。在培养的内皮细胞(HUVECs)上检测LG (LG-CM)和LG- mscs (LG- mscs -CM)的条件培养基(CM),评估其增殖、脐带形成以及粘附分子(AM) VCAM-1和ICAM-1的表达。采用巨噬细胞系U937评价其抗炎特性,如迁移、在HUVECs上的粘附、RANTES和MCP-1的释放。结果:我们的研究结果表明,LG含有大量表达NG2和CD146(两种周细胞标记物)的间充质细胞,以及丰富的微血管内皮细胞(mEC)群体。实质上,LG-CM和LG-MSC-CM都增加了脐带形成,抑制了TNFα刺激后内皮细胞ICAM-1和VCAM-1的表达,并略微改善了HUVEC的增殖。在趋化因子MCP-1刺激下,LG-CM和LG-MSC-CM的加入强烈抑制了U937的迁移,降低了它们在HUVECs上的粘附,并显著抑制了RANTES和MCP-1的释放。结论:我们的数据表明,LG微碎片脂肪组织本身或其嵌入的MSCs含量保留了诱导血管稳定和抑制参与炎症的巨噬细胞功能的能力。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Vascular Cell
Vascular Cell Neuroscience-Neurology
CiteScore
0.70
自引率
0.00%
发文量
0
期刊最新文献
A 3-Dimensional Hypothesis of Oxidative Phosphorylation The long and winding road: detecting and quantifying Notch activation in endothelial cells Vascular Tumors Result from Adeno-Associated Virus-9 Angiogenic Gene Therapy of Bone Allografts A clinically relevant model of stroke using aged rats The SARS-CoV2 - ACE2 link: a physiopathological analysis
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1