scholarly journals Reparative effect of mesenchymal stromal cells on endothelial cells after hypoxic and inflammatory injury

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Jesus M. Sierra-Parraga ◽  
Ana Merino ◽  
Marco Eijken ◽  
Henri Leuvenink ◽  
Rutger Ploeg ◽  
...  
Cytotherapy ◽  
2020 ◽  
Vol 22 (5) ◽  
pp. S171-S172
Author(s):  
J.M. Sierra Parraga ◽  
A. Merino ◽  
M. Eijken ◽  
R. Ploeg ◽  
B. Møller ◽  
...  

Stroke ◽  
2018 ◽  
Vol 49 (Suppl_1) ◽  
Author(s):  
Kaavya Giridhar ◽  
Nikunj Satani ◽  
Bing Yang ◽  
Songmi Lee ◽  
Xiaopei Xi ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (8) ◽  
pp. 1063-1075 ◽  
Author(s):  
Douaa Dhahri ◽  
Kaori Sato-Kusubata ◽  
Makiko Ohki-Koizumi ◽  
Chiemi Nishida ◽  
Yoshihiko Tashiro ◽  
...  

Key Points tPA expands mesenchymal stromal cells (MSCs) in the bone marrow by a cytokine (KitL and PDGF-BB) crosstalk with endothelial cells. Pharmacologic inhibition of receptor tyrosine kinases (c-Kit and PDGFRα) impairs tPA-mediated MSC proliferation.


2020 ◽  
Author(s):  
Jesus Maria Sierra-Parraga ◽  
Ana Merino ◽  
Marco Eijken ◽  
Henri Leuvenink ◽  
Rutger J. Ploeg ◽  
...  

Abstract Background The renal endothelium is a prime target for ischemia reperfusion injury (IRI) during donation and transplantation procedures. Mesenchymal stromal cells (MSC) have been shown to ameliorate kidney function after IRI. However, whether this involves repair of the endothelium is not clear. Therefore, our objective is to study potential regenerative effects of MSC on injured endothelial cells and to identify the molecular mechanisms involved. Methods Human umbilical vein endothelial cells (HUVEC) were submitted to hypoxia and reoxygenation and TNF-a treatment. To determine whether physical interaction or soluble factors released by MSC were responsible for the potential regenerative effects of MSC on endothelial cells, dose-response experiments were performed in co-culture and transwell conditions and with secretome deficient MSC. Results MSC showed increased migration and adhesion to injured HUVEC, mediated by CD29 and CD44 on the MSC membrane. MSC decreased membrane injury marker expression, oxidative stress levels and monolayer permeability of injured HUVEC, which was observed only when allowing both physical and paracrine interaction between MSC and HUVEC. Furthermore, viable MSC in direct contact with injured HUVEC improved wound healing capacity by 45% and completely restored their angiogenic capacity. In addition, MSC exhibited an increased ability to migrate through an injured HUVEC monolayer compared to non-injured HUVEC in vitro. Conclusions These results show that MSC have regenerative effects on injured HUVEC via a mechanism which requires both physical and paracrine interaction. The identification of specific effector molecules involved in MSC-HUVEC interaction will allow targeted modification of MSC to apply and enhance the therapeutic effects of MSC in IRI.


2021 ◽  
Vol 12 ◽  
Author(s):  
Ana Merino ◽  
Marta Sablik ◽  
Sander S. Korevaar ◽  
Carmen López-Iglesias ◽  
Maitane Ortiz-Virumbrales ◽  
...  

Proinflammatory stimuli lead to endothelial injury, which results in pathologies such as cardiovascular diseases, autoimmune diseases, and contributes to alloimmune responses after organ transplantation. Both mesenchymal stromal cells (MSC) and the extracellular vesicles (EV) released by them are widely studied as regenerative therapy for the endothelium. However, for therapeutic application, the manipulation of living MSC and large-scale production of EV are major challenges. Membrane particles (MP) generated from MSC may be an alternative to the use of whole MSC or EV. MP are nanovesicles artificially generated from the membranes of MSC and possess some of the therapeutic properties of MSC. In the present study we investigated whether MP conserve the beneficial MSC effects on endothelial cell repair processes under inflammatory conditions. MP were generated by hypotonic shock and extrusion of MSC membranes. The average size of MP was 120 nm, and they showed a spherical shape. The effects of two ratios of MP (50,000; 100,000 MP per target cell) on human umbilical vein endothelial cells (HUVEC) were tested in a model of inflammation induced by TNFα. Confocal microscopy and flow cytometry showed that within 24 hours >90% of HUVEC had taken up MP. Moreover, MP ended up in the lysosomes of the HUVEC. In a co-culture system of monocytes and TNFα activated HUVEC, MP did not affect monocyte adherence to HUVEC, but reduced the transmigration of monocytes across the endothelial layer from 138 ± 61 monocytes per microscopic field in TNFα activated HUVEC to 61 ± 45 monocytes. TNFα stimulation induced a 2-fold increase in the permeability of the HUVEC monolayer measured by the translocation of FITC-dextran to the lower compartment of a transwell system. At a dose of 1:100,000 MP significantly decreased endothelial permeability (1.5-fold) respect to TNFα Stimulated HUVEC. Finally, MP enhanced the angiogenic potential of HUVEC in an in vitro Matrigel assay by stimulating the formation of angiogenic structures, such as percentage of covered area, total tube length, total branching points, total loops. In conclusion, MP show regenerative effects on endothelial cells, opening a new avenue for treatment of vascular diseases where inflammatory processes damage the endothelium.


2020 ◽  
Vol 2020 ◽  
pp. 1-8
Author(s):  
Hataiwan Kunkanjanawan ◽  
Tanut Kunkanjanawan ◽  
Veerapol Khemarangsan ◽  
Rungrueang Yodsheewan ◽  
Kasem Theerakittayakorn ◽  
...  

Coimplantation of endothelial cells (ECs) and mesenchymal stromal cells (MSCs) into the transplantation site could be a feasible option to achieve a sufficient level of graft-host vascularization. To find a suitable source of tissue that provides a large number of high-quality ECs and MSCs suited for future clinical application, we developed a simplified xeno-free strategy for isolation of human umbilical vein endothelial cells (HUVECs) and Wharton’s jelly-derived mesenchymal stromal cells (WJ-MSCs) from the same umbilical cord. We also assessed whether the coculture of HUVECs and WJ-MSCs derived from the same umbilical cord (autogenic cell source) or from different umbilical cords (allogenic cell sources) had an impact on in vitro angiogenic capacity. We found that HUVECs grown in 5 ng/ml epidermal growth factor (EGF) supplemented xeno-free condition showed higher proliferation potential compared to other conditions. HUVECs and WJ-MSCs obtained from this technic show an endothelial lineage (CD31 and von Willebrand factor) and MSC (CD73, CD90, and CD105) immunophenotype characteristic with high purity, respectively. It was also found that only the coculture of HUVEC/WJ-MSC, but not HUVEC or WJ-MSC mono-culture, provides a positive effect on vessel-like structure (VLS) formation, in vitro. Further investigations are needed to clarify the pros and cons of using autogenic or allogenic source of EC/MSC in tissue engineering applications. To the best of our knowledge, this study offers a simple, but reliable, xeno-free strategy to establish ECs and MSCs from the same umbilical cord, a new opportunity to facilitate the development of personal cell-based therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2589-2589
Author(s):  
Olga Nilmaer ◽  
Melanie Giesen ◽  
Erika Deak ◽  
Victoria Lang ◽  
Johannes Leibacher ◽  
...  

Abstract Abstract 2589 Mesenchymal Stromal Cells (MSCs) are increasingly used in patients e.g. to combat Graft-versus-Host-Disease or to support hematopoietic regeneration after allogeneic Hematopoietic Stem Cell Transplantation. However, both the dynamics and the mechanisms by which intravenously transplanted MSCs (n=5 donors) interact with the vessel wall are incompletely characterized. We compared the ability of MSCs and blood leukocytes (PBMCs; n=5) to interact with known endothelial ligands using a flow chamber system and transplantation into immunodeficient mice. Although recombinant P- and E-selectin reduced the flow speed of MSCs significantly by 30±3% (means ± SD), which was reversible in the presence of function-blocking anti P- or E-selectin antibodies MSCs were induced to roll at speeds of 1–5 μm/s at much lower efficiency as PBMCs at shear stresses of 0.35 – 8 dyn/cm2 (MSCs, 2±1%; PBMCs, 21±4% of interacting cells; means ± SD). In contrast, immobilized recombinant VCAM-1 Ig under these conditions dose-dependently induced interactions of MSCs with a comparable efficiency as of PBMCs, leading to stable arrest. Whereas 33 ± 8% (means ± SD) of PBMCs rolled on 20 μg/ml VCAM-1 Ig, MSCs howerver lacked rolling behaviour but always directly stopped. Analysis of the velocity vectors of individual MSCs revealed an staggering interaction of MSCs during the stopping process with prolonged distances until they reached stable arrest (58±10 vs 14±3 μm for MSCs and PBMCs, respectively (means ± SD, p=0.004). Whereas PBMCs were induced to both roll and arrest on the b2 integrin ligand ICAM-1 and on the a4b7 integrin ligand MAdCAM-1 at 0.35–2 dyn/cm2, MSCs were incapable to roll or arrest under these conditions. Analysis of MSC arrest behaviour on HUVEC endothelial cells pretreated with TNF-a that expressed both VCAM-1 and ICAM-1 and coated with chemokines CXCL12 or CCL19 showed that MSCs arrested with only 14±5% of the efficiency of PBMCs at 0.35dyn/cm2. Function blocking antibodies confirmed the pivotal role of a4b1/VCAM-1 dependent adhesion in stable arrest of MSCs. We found that MSCs bound to the HUVECs more rigidly than PBMCs since, when arrested cells were exposed to increased shear stress of 5 dyn/cm2 only 3±4% of MSCs de-adhered, in contrast to PBMCs which 74±4% de-adhered. Chemokine-induced signals were instrumental in strengthening adhesion of MSCs since for CXCL12-induced MSCs to endothelial cells adhesion was counteracted (i) pretreatment with pertussis toxin, (ii) by application of the CXCR4 inhibitor (AMD3100) and (iii) after siRNA mediated downregulation of b arrestins 1 or 2, or the GPCR kinase (GRK)-6, which known to be involved in chemokine receptor signalling. All above treatments lead to significantly decreased numbers of MSCs that remained adherent at shear stresses between 2 and 15 dyn/cm2. Since cDNA microarray analises using MSCs exposed to shear stress for 0–12h indicated, among others, upregulation of the GTPase Rap1A and its activator, PDZ-GEF1, we analyzed activation of Rap1 proteins using a pulldown assay in a CXCL12-induced manner. MSCs depleted of Rap1A and B proteins by transfection with siRNA showed a significant reduction of stably arresting cells (>10s) on HUVEC precoated with CCL19 to 66±4% of controls (p=0.04). Moreover, siRNA mediated depletion of Rap1A and B in MSCs resulted in a >60% reduction in numbers of transplanted MSCs accumulating in the lungs of NOD/SCID mice, and concomitant significant increases of numbers of MSCs detected in blood (8.9±0.4 fold; p<0.05) and other tissues such as liver (2.2±0.8 fold, p<0.05) and spleen (2.1±0.7 fold, p<0.05). Together, MSCs display major deficits in adhesion dynamics and adhesion receptor usage compared with normal leukocytes. Modulation of the GTPase Rap1 may serve as a strategy to counteract the strong activation of b1 integrins and improve the circulation behaviour of transplanted MSCs. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document