Abstract 294: Expression of Matrix Metalloproteases during the Differentiation of Porcine Adopse-Drived Mesenchymal Stem Cells ro Endothelial Cells

2013 ◽  
Vol 33 (suppl_1) ◽  
Author(s):  
Sami G Almalki ◽  
Velidi Rao ◽  
Divya Pankajakshan ◽  
Devendra K Agrawal

Rationale Adipose-derived mesenchymal stem cells (ADMSCs) are multipotent cells that have the potential to differentiate into different cell linages, and represent promising tools in various clinical applications. However, the molecular mechanisms that control the ability of ADMSCs to remodel 3-dimensional extracellular matrix (ECM) barriers during differentiation are not clearly understood. Herein, we studied the expression of matrix metalloproteinases (MMPs) during the differentiation of ADMSCs to endothelial cells (ECs) in vitro . Methods MSCs were isolated from porcine abdominal adipose tissue, and characterized by positive staining for MSC markers, CD44, CD73, CD90, and negative staining for CD11b, CD34 and CD45. The plasticity of MSCs was detected by bi-lineage differentiation to osteocytes, and adipocytes. The mRNA transcripts for different MMPs and TIMPs and protein expression of EC markers were analyzed by RT-PCR and immunostaining. The enzyme activity and protein expression were also analyzed by gelatin zymography, ELISA, and Western blot. Results The differentiation of ADMSCs to ECs was confirmed by the positive staining and mRNA expression of the endothelial markers. The mRNA transcripts for MMP-2 and membrane type 1 MMP (MT1-MMP) was significantly increased by 2.5 and 2.0 fold, respectively, during the differentiation of MSCs into ECs. Western blot and ELISA showed an elevated MT1-MMP and MMP-2 expression. The enzyme activity of MMP-2 was also observed by gelatin zymography. Conclusion We demonstrated that porcine ADMSCs have the ability to differentiate into ECs, and this process involves the up-regulation of MMP-2 and MT1-MMP. The increase in the expression of MMP-2 and MT1-MMP may, at least partially, facilitate the change in morphology of MSCs by degrading the ECM barriers. These findings may provide a potential mechanism for the role of MMP2 and MT1-MMP in the differentiation of ADMSCs into ECs.

2021 ◽  
Vol 21 ◽  
Author(s):  
Xuping Niu ◽  
Qixin Han ◽  
Xiaofang Li ◽  
Juan Li ◽  
Yanmin Liu ◽  
...  

Objective: Psoriasis is a chronic inflammatory skin disease highly depending on angiogenesis. Our prior results showed that the mRNA and protein of Del-1 in dermal mesenchymal stem cells (dMSCs) was up-regulated from psoriasis. Our aim was further to investigate the role of Del-1 from dMSCs in the pathogenesis of psoriasis and confirm the effect of Del-1 on the pathogenesis of psoriasis. Methods: We conducted an immunohistochemistry experiment to further investigate the expression of Del-1in psoriatic lesions. In addition, dMSCs with over-expressed Del-1 via the lentiviral vector of Del-1 were co-cultured with ECs, and the protein expression of integrins (αvβ3, αvβ5 ,and α5β1) of ECs were detected by western blotting. Results: This research showed that Del-1 was significantly increased in lesions of patients with psoriasis (p< .05, 9.96 vs. 2.18), and Del-1 from dMSCs successfully induced up-regulation of integrins α5β1 and αvβ3 (all p < .05). Conclusion: This study demonstrated that Del-1 from dMSCs was involved in the pathogenesis of psoriasis through induced angiogenesis. And Del-1, αvβ3 and α5β1 may be potential new targets for inhibiting angiogenesis in psoriasis.


2015 ◽  
Vol 35 (suppl_1) ◽  
Author(s):  
Yovani Llamas ◽  
Sami Almalki ◽  
Devendra K Agrawal

Background: Cell-based therapy using adipose-derived mesenchymal stem cells (AMSCs) is an attractive option for re-endothelialization post-angioplasty procedures. Vitamin D levels in circulating blood may regulate AMSC-based re-endothelialization of injured arteries. To our knowledge, the role of vitamin D and vascular endothelial growth factor (VEGF) in the differentiation of adipose-derived AMSCs into endothelial cells (ECs) has not been examined. In this study, we investigated the effect of vitamin D on VEGF-induced differentiation of MSCs into ECs. Methods and Results: MSCs isolated from porcine adipose tissue were CD11b-CD34-CD44+CD73+CD90+ and showed characteristics of MSCs. The MSCs were stimulated and differentiated into ECs with endothelial growth media (EGM+50ng/ml of VEGF) and EGM media containing 10nM of calcitriol (EGM+50ng/ml of VEGF +10nM calcitriol) for 10 days. The EC differentiation was assessed by mRNA expression by qPCR and protein expression by flow cytometry for endothelial cell markers. Calcitriol enhanced EGM+VEGF-induced differentiation of MSCs into ECs, as revealed by 2-fold increase in mRNA and 4-fold increase in protein expression of EC markers. Angiogenesis assay and acetylated low density lipoprotein (LDL) uptake assay were used to assess the endothelial functionality that showed significant increase in capillary tube sprouting, and an increase in LDL uptake by the differentiated cells in response to EGM +VEGF+ calcitriol. Examination of the findings from Wnt Pathway array revealed a 3-fold decrease in β-catenin and 3-fold increase in KREMEN1 protein in the cells treated with EGM + VEGF+calcitriol. Conclusion: Vitamin D significantly enhanced VEGF-induced differentiation of MSCs into endothelial cells. Thus, vitamin D status of the patients undergoing coronary intervention might regulate the development of thrombosis and intimal hyperplasia.


Author(s):  
Francesca Pagani ◽  
Elisa Tratta ◽  
Patrizia Dell’Era ◽  
Manuela Cominelli ◽  
Pietro Luigi Poliani

AbstractEarly B-cell factor-1 (EBF1) is a transcription factor with an important role in cell lineage specification and commitment during the early stage of cell maturation. Originally described during B-cell maturation, EBF1 was subsequently identified as a crucial molecule for proper cell fate commitment of mesenchymal stem cells into adipocytes, osteoblasts and muscle cells. In vessels, EBF1 expression and function have never been documented. Our data indicate that EBF1 is highly expressed in peri-endothelial cells in both tumor vessels and in physiological conditions. Immunohistochemistry, quantitative reverse transcription polymerase chain reaction (RT-qPCR) and fluorescence-activated cell sorting (FACS) analysis suggest that EBF1-expressing peri-endothelial cells represent bona fide pericytes and selectively express well-recognized markers employed in the identification of the pericyte phenotype (SMA, PDGFRβ, CD146, NG2). This observation was also confirmed in vitro in human placenta-derived pericytes and in human brain vascular pericytes (HBVP). Of note, in accord with the key role of EBF1 in the cell lineage commitment of mesenchymal stem cells, EBF1-silenced HBVP cells showed a significant reduction in PDGFRβ and CD146, but not CD90, a marker mostly associated with a prominent mesenchymal phenotype. Moreover, the expression levels of VEGF, angiopoietin-1, NG2 and TGF-β, cytokines produced by pericytes during angiogenesis and linked to their differentiation and activation, were also significantly reduced. Overall, the data suggest a functional role of EBF1 in the cell fate commitment toward the pericyte phenotype.


RSC Advances ◽  
2021 ◽  
Vol 11 (30) ◽  
pp. 18685-18692
Author(s):  
Hiroki Masuda ◽  
Yoshinori Arisaka ◽  
Masahiro Hakariya ◽  
Takanori Iwata ◽  
Tetsuya Yoda ◽  
...  

Molecular mobility of polyrotaxane surfaces promoted mineralization in a co-culture system of mesenchymal stem cells and endothelial cells.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ji-wen Cheng ◽  
Li-xia Duan ◽  
Yang Yu ◽  
Pu Wang ◽  
Jia-le Feng ◽  
...  

Abstract Background Mesenchymal stem cells (MSCs) play a crucial role in cancer development and tumor resistance to therapy in prostate cancer, but the influence of MSCs on the stemness potential of PCa cells by cell–cell contact remains unclear. In this study, we investigated the effect of direct contact of PCa cells with MSCs on the stemness of PCa and its mechanisms. Methods First, the flow cytometry, colony formation, and sphere formation were performed to determine the stemness of PCaMSCs, and the expression of stemness-related molecules (Sox2, Oct4, and Nanog) was investigated by western blot analysis. Then, we used western blot and qPCR to determine the activity levels of two candidate pathways and their downstream stemness-associated pathway. Finally, we verified the role of the significantly changed pathway by assessing the key factors in this pathway via in vitro and in vivo experiments. Results We established that MSCs promoted the stemness of PCa cells by cell–cell contact. We here established that the enhanced stemness of PCaMSCs was independent of the CCL5/CCR5 pathway. We also found that PCaMSCs up-regulated the expression of Notch signaling-related genes, and inhibition of Jagged1-Notch1 signaling in PCaMSCs cells significantly inhibited MSCs-induced stemness and tumorigenesis in vitro and in vivo. Conclusions Our results reveal a novel interaction between MSCs and PCa cells in promoting tumorigenesis through activation of the Jagged1/Notch1 pathway, providing a new therapeutic target for the treatment of PCa.


Materials ◽  
2021 ◽  
Vol 14 (5) ◽  
pp. 1078
Author(s):  
Han Young Kim ◽  
Suk Ho Bhang

As a tissue regeneration strategy, the utilization of mesenchymal stem cells (MSCs) has drawn considerable attention. Comprehensive research using MSCs has led to significant preclinical or clinical outcomes; however, improving the survival rate, engraftment efficacy, and immunogenicity of implanted MSCs remains challenging. Although MSC-derived exosomes were recently introduced and reported to have great potential to replace conventional MSC-based therapeutics, the poor production yield and heterogeneity of exosomes are critical hurdles for their further applications. Herein, we report the fabrication of exosome-mimetic MSC-engineered nanovesicles (MSC-NVs) by subjecting cells to serial extrusion through filters. The fabricated MSC-NVs exhibit a hydrodynamic size of ~120 nm, which is considerably smaller than the size of MSCs (~30 μm). MSC-NVs contain both MSC markers and exosome markers. Importantly, various therapeutic growth factors originating from parent MSCs are encapsulated in the MSC-NVs. The MSC-NVs exerted various therapeutic effects comparable to those of MSCs. They also significantly induced the angiogenesis of endothelial cells and showed neuroprotective effects in damaged neuronal cells. The results collectively demonstrate that the fabricated MSC-NVs can serve as a nanosized therapeutic agent for tissue regeneration.


2011 ◽  
Vol 2011 ◽  
pp. 1-4 ◽  
Author(s):  
Zhaoxia Pu ◽  
Xiangdong You ◽  
Qiyuan Xu ◽  
Feng Gao ◽  
Xiaojie Xie ◽  
...  

Ultrasound-targeted microbubble destruction (UTMD) has been proposed as a new technique for organ-specific gene transfer and drug delivery. This study was performed to investigate the effect of UTMD on marrow mesenchymal stem cells (MSCs) transfected with pcDNA3.1−-hVEGF165.pcDNA3.1−-hVEGF165were transfected into the third passage of MSCs, with or without UTMD under different ultrasound conditions. Protein expression was quantified by hVEGF165-ELISA kit after transfection for 24, 48, and 72 hours. UTMD-mediated transfection of MSCs yielded a significant protein expression. UTMD of mechanic index (MI) 0.6 for 90 seconds led to the highest level of protein expression.


2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Sangho Lee ◽  
Min Kyung Lee ◽  
Hyunjoon Kong ◽  
Young-sup Yoon

Various hydrogels are used to create vascular structure in vitro or to improve cell engraftment to overcome low cell survival in vivo, a main hurdle for bare cell therapy Recently we developed a modified alginate hydrogel within which microchannels are aligned to guide the direction and spatial organization of loaded cells. We investigated whether these cell constructs in which HUVECs and human mesenchymal stem cells (hMSCs) are co-loaded in this novel microchanneled hydrogel facilitate formation of vessels in vitro and in vivo, and enhance recovery of hindlimb ischemia. We crafted a modified alginate hydrogel which has microchannels, incorporates a cell adhesion peptide RGD, and was encapsulated with VEGF. We then compared vascular structure formation between the HUVEC only (2 x 105 cells) group and the HUVEC plus hMSC group. In the HUVEC+hMSC group, we mixed HUVECs and hMSCs at the ratio of 3:1. For cell tracking, we labeled HUVECs with DiO, a green fluorescence dye. After loading cells into the microchannels of the hydrogel, these constructs were cultured for seven days and were examined by confocal microscopy. In the HUVEC only group, HUVECs stands as round shaped cells without forming tubular structures within the hydrogel. However, in the HUVEC+hMSC group, HUVECs were stretched out and connected with each other, and formed vessel-like structure following pre-designed microchannels. These results suggested that hMSCs play a critical role for vessel formation by HUVECs. We next determined their in vivo effects using a mouse hindlimb ischemia model. We found that engineered HUVEC+hMSC group showed significantly higher perfusion over 4 weeks compared to the engineered HUVEC only group or bare cell (HUVEC) group. Confocal microscopic analysis of harvested tissues showed more robust vessel formation within and outside of the cell constructs and longer term cell survival in HUVEC+hMSC group compared to the other groups. In conclusion, this novel microchanneled alginate hydrogel facilitates aligned vessel formation of endothelial cells when combined with MSCs. This vessel-embedded hydrogel constructs consisting of HUVECs and MSCs contribute to perfusable vessel formation, prolong cell survival in vivo, and are effective for recovering limb ischemia.


Sign in / Sign up

Export Citation Format

Share Document