scholarly journals TBX20 Contributes to Balancing the Differentiation of Perivascular Adipose-Derived Stem Cells to Vascular Lineages and Neointimal Hyperplasia

Author(s):  
Yongli Ji ◽  
Yuankun Ma ◽  
Jian Shen ◽  
Hui Ni ◽  
Yunrui Lu ◽  
...  

BackgroundPerivascular adipose-derived stem cells (PVASCs) can contribute to vascular remodeling, which are also capable of differentiating into multiple cell lineages. The present study aims to investigate the mechanism of PVASC differentiation toward smooth muscle cells (SMCs) and endothelial cells (ECs) as well as its function in neointimal hyperplasia.MethodsSingle-cell sequencing and bulk mRNA sequencing were applied for searching key genes in PVASC regarding its role in vascular remodeling. PVASCs were induced to differentiate toward SMCs and ECs in vitro, which was quantitatively evaluated using immunofluorescence, quantitative real-time PCR (QPCR), and Western blot. Lentivirus transfections were performed in PVASCs to knock down or overexpress TBX20. In vivo, PVASCs transfected with lentivirus were transplanted around the guidewire injured femoral artery. Hematoxylin–eosin (H&E) staining was performed to examine their effects on neointimal hyperplasia.ResultsBulk mRNA sequencing and single-cell sequencing revealed a unique expression of TBX20 in PVASCs. TBX20 expression markedly decreased during smooth muscle differentiation while it increased during endothelial differentiation of PVASCs. TBX20 knockdown resulted in the upregulation of SMC-specific marker expression and activated Smad2/3 signaling, while inhibiting endothelial differentiation. In contrast, TBX20 overexpression repressed the differentiation of PVASCs toward smooth muscle cells but promoted endothelial differentiation in vitro. Transplantation of PVASCs transfected with TBX20 overexpression lentivirus inhibited neointimal hyperplasia in a murine femoral artery guidewire injury model. On the contrary, neointimal hyperplasia significantly increased in the TBX20 knockdown group.ConclusionA subpopulation of PVASCs uniquely expressed TBX20. TBX20 could regulate SMC and EC differentiation of PVASCs in vitro. Transplantation of PVASCs after vascular injury suggested that PVASCs participated in neointimal hyperplasia via TBX20.

Biomedicines ◽  
2021 ◽  
Vol 9 (7) ◽  
pp. 797
Author(s):  
Alvaro Yogi ◽  
Marina Rukhlova ◽  
Claudie Charlebois ◽  
Ganghong Tian ◽  
Danica B. Stanimirovic ◽  
...  

Synthetic grafts have been developed for vascular bypass surgery, however, the risks of thrombosis and neointimal hyperplasia still limit their use. Tissue engineering with the use of adipose-derived stem cells (ASCs) has shown promise in addressing these limitations. Here we further characterized and optimized the ASC differentiation into smooth muscle cells (VSMCs) induced by TGF-β and BMP-4. TGF-β and BMP-4 induced a time-dependent expression of SMC markers in ASC. Shortening the differentiation period from 7 to 4 days did not impair the functional property of contraction in these cells. Stability of the process was demonstrated by switching cells to regular growth media for up to 14 days. The role of IGFBP7, a downstream effector of TGF-β, was also examined. Finally, topographic and surface patterning of a substrate is recognized as a powerful tool for regulating cell differentiation. Here we provide evidence that a non-woven PET structure does not affect the differentiation of ASC. Taken together, our results indicate that VSMCs differentiated from ASCs are a suitable candidate to populate a PET-based vascular scaffolds. By employing an autologous source of cells we provide a novel alternative to address major issues that reduces long-term patency of currently vascular grafts.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Yongli Ji ◽  
Yunrui Lu ◽  
Jian Shen ◽  
Meixiang Xiang ◽  
Yao Xie

Introduction: Recent researches identified the existence of perivascular adipose-derived stem cells (ADSCs) which could differentiate into vascular lineages and participate in vascular remodeling. Single-cell mRNA analysis revealed cellular heterogeneity of subcutaneous ADSCs in respect to cell clustering and cell differentiation. However, such analysis of perivascular ADSCs has not been investigated at a single-cell level. Hypothesis: There is a significant difference among perivascular ADSCs subpopulations in respect to vascular-lineage differentiation. Methods: We performed droplet-based single-cell profiling of subcutaneous and perivascular adipose stromal cells and compared ADSCs regarding their heterogeneity, gene ontology, and cell fate trajectory by applying single-cell analysis as well as in vitro and in vivo assays. Results: Single-cell analysis uncovered 4 perivascular ADSCs subpopulations including Dpp4+ , Col4a2+ / Icam1+ , Clec11a+ / Cpe+ and Sult1e1+ cells. Notably, the Clec11a + subpopulation comprised the bulk of perivascular ADSCs, while was hardly presented in subcutaneous ADSCs. Further gene-set enrichment analysis suggested Clec11a + ADSCs were potentially involved with TGF-β signaling pathways and pseudotemporal analysis predicted that Clec11a + subpopulation lay at the end of the differential trajectory towards smooth muscle cells (SMCs). In vitro assays displayed that perivascular ADSCs could differentiate into SMCs via CLEC11A regulation when treated by TGF-β1. To further elucidate the role of the Clec11a + subpopulation in SMCs differentiation, we labeled CLEC11A+ and CLEC11A- perivascular ADSCs by lentivirus transfection and isolated them by FACS assay. CLEC11A+ cells showed the greater capability of SMCs differentiation in response to TGF-β1 in vitro and enhanced neointima formation when transplanted to the adventitial side of guidewire injured arteries. Conclusions: The present study depicted the unique heterogeneity of perivascular ADSCs and the novel role of the Clec11a + subpopulation, providing a supplement for the relationship between perivascular ADSCs and vascular SMCs.


2015 ◽  
Vol 61 (6) ◽  
pp. 172S
Author(s):  
Spencer Brown ◽  
Francis J. Caputo ◽  
Telisha Ortiz ◽  
Ping Zhang ◽  
William M. Harris ◽  
...  

2021 ◽  
Author(s):  
Yao Xie ◽  
Yongli Ji ◽  
Yunrui Lu ◽  
Yuankun Ma ◽  
Hui Ni ◽  
...  

Adipose derived stem cells (ADSCs) can differentiate into vascular lineages and participate in vascular remodeling. Perivascular ADSCs (PV-ADSCs) draw attention due to their unique location. The heterogeneity of subcutaneous (SUB-) and abdominal ADSCs were well addressed, but PV-ADSCs’ heterogeneity hasn’t been investigated. In the present study, we applied single-cell analysis to compare SUB-ADSCs and PV-ADSCs respectively regarding their subpopulations, functions, and cell fates. We uncovered 4 subpopulations of PV-ADSCs including <i>Dpp4+</i>, <i>Col4a2+</i>/<i>Icam1+</i>, <i>Clec11a+</i>/<i>Cpe+</i> and <i>Sult1e1+</i> cells, among which <a></a><a><i>Clec11a</i>+ subpopulation</a> potentially participated in and regulated the PV-ADSCs differentiation towards a smooth muscle cell (SMC) phenotype. The present study revealed the <a></a><a>distinct characteristics </a>between PV-ADSCs and SUB-ADSCs.


2021 ◽  
Author(s):  
Yao Xie ◽  
Yongli Ji ◽  
Yunrui Lu ◽  
Yuankun Ma ◽  
Hui Ni ◽  
...  

Adipose derived stem cells (ADSCs) can differentiate into vascular lineages and participate in vascular remodeling. Perivascular ADSCs (PV-ADSCs) draw attention due to their unique location. The heterogeneity of subcutaneous (SUB-) and abdominal ADSCs were well addressed, but PV-ADSCs’ heterogeneity hasn’t been investigated. In the present study, we applied single-cell analysis to compare SUB-ADSCs and PV-ADSCs respectively regarding their subpopulations, functions, and cell fates. We uncovered 4 subpopulations of PV-ADSCs including <i>Dpp4+</i>, <i>Col4a2+</i>/<i>Icam1+</i>, <i>Clec11a+</i>/<i>Cpe+</i> and <i>Sult1e1+</i> cells, among which <a></a><a><i>Clec11a</i>+ subpopulation</a> potentially participated in and regulated the PV-ADSCs differentiation towards a smooth muscle cell (SMC) phenotype. The present study revealed the <a></a><a>distinct characteristics </a>between PV-ADSCs and SUB-ADSCs.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Pegah Nammian ◽  
Seyedeh-Leili Asadi-Yousefabad ◽  
Sajad Daneshi ◽  
Mohammad Hasan Sheikhha ◽  
Seyed Mohammad Bagher Tabei ◽  
...  

Abstract Introduction Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. Methods For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. Results Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. Conclusions Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.


2021 ◽  
Vol 30 ◽  
pp. 096368972110354
Author(s):  
Eun-Jung Yoon ◽  
Hye Rim Seong ◽  
Jangbeen Kyung ◽  
Dajeong Kim ◽  
Sangryong Park ◽  
...  

Stamina-enhancing effects of human adipose derived stem cells (hADSCs) were investigated in young Sprague-Dawley rats. Ten-day-old male rats were transplanted intravenously (IV) or intracerebroventricularly (ICV) with hADSCs (1 × 106 cells/rat), and physical activity was measured by locomotor activity and rota-rod performance at post-natal day (PND) 14, 20, 30, and 40, as well as a forced swimming test at PND 41. hADSCs injection increased the moving time in locomotor activity, the latency in rota-rod performance, and the maximum swimming time. For the improvement of physical activity, ICV transplantation was superior to IV injection. In biochemical analyses, ICV transplantation of hADSCs markedly reduced serum creatine phosphokinase, lactate dehydrogenase, alanine transaminase, and muscular lipid peroxidation, the markers for muscular and hepatic injuries, despite the reduction in muscular glycogen and serum triglycerides as energy sources. Notably, hADSCs secreted brain-derived neurotrophic factor (BDNF) and nerve growth factor in vitro, and increased the level of BDNF in the brain and muscles in vivo. The results indicate that hADSCs enhance physical activity including stamina not only by attenuating tissue injury, but also by strengthening the muscles via production of BDNF.


2019 ◽  
Vol 103 (3) ◽  
pp. 925-934
Author(s):  
Eкaterina Vackova ◽  
Darko Bosnakovski ◽  
Bodil Bjørndal ◽  
Penka Yonkova ◽  
Natalia Grigorova ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document