Mesenchymal Stromal Cells (MSCs) Lack Adhesion to Endothelial Cells through Selectins and b2 Integrins and Interact Dominantly Using Integrin a4b1 Activated by GTPase Rap1.

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.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4358-4358
Author(s):  
Manal Alsheikh ◽  
Roya Pasha ◽  
Nicolas Pineault

Abstract Osteoblasts (OST) found within the endosteal niche are important regulators of Hematopoietic Stem and Progenitor Cells (HSPC) under steady state and during hematopoietic reconstitution. OST are derived from mesenchymal stromal cells (MSC) following osteogenic differentiation. MSC and OST secrete a wide array of soluble factors that sustain hematopoiesis. Recently, we showed that media conditioned with OST derived from MSC (referred as M-OST) after 6 days of osteogenic differentiation were superior to MSC conditioned media (CM) for the expansion of cord blood (CB) progenitors, and CB cells expanded with M-OST CM supported a more robust engraftment of platelets in NSG mice after transplantation. These findings raised the possibility that M-OST could be superior to MSC for the ex vivoexpansion HSPC. In this study, we set out to test the hypothesis that the growth modulatory activity of M-OST would vary as a function of their maturation status. The objectives were to first monitor the impact of M-OST differentiation and maturation status on the expression of soluble factors that promote HSPC expansion and in second, to investigate the capacity of M-OST CMs prepared from M-OST at distinct stages of differentiation to support the expansion and differentiation of HSPCs in culture. M-OST at distinct stages of differentiation were derived by culturing bone marrow MSC in osteogenic medium for various length of time (3 to 21 days). All CB CD34+ enriched (92±7% purity) cell cultures were done with serum free media conditioned or not with MSC or M-OST and supplemented with cytokines SCF, TPO and FL. We first confirmed the progressive differentiation and maturation of M-OST as a function of osteogenic culture length, which was evident by the induction of the osteogenic transcription factors Osterix, Msx2 and Runx2 mRNAs, the gradual increase in osteopontin and alkaline phosphatase positive cells and quantitative increases in calcium deposit. Next, we investigated the expression in MSC and M-OSTs of genes known to collaborate for the expansion of HSPCs by Q-PCR. Transcript copy numbers for IGFBP-2 increased swiftly during osteogenic differentiation, peaking at day-3 (˃100-fold vs MSC, n=2) and returning below MSC level by day-21. In contrast, ANGPTL members (ANGPTL-1, -2, -3 and -5) remained superior in M-OSTs throughout osteogenic differentiation with expression levels peaking around day 6 (n=2). Next, we tested the capacity of media conditioned with primitive (day-3, -6), semi-mature (day-10, -14) and mature M-OST (day-21) to support the growth of CB cells. All M-OST CMs increased (p˂0.03) the growth of total nucleated cells (TNC) after 6 days of culture compared to non-conditioned medium used as control (mean 2.0-fold, n=4). Moreover, there was a positive correlation between cell growth and M-OST maturation status though differences between the different M-OST CMs tested were not significant. The capacity of M-OST CMs to increase (mean 2-fold, n=4) the expansion of CD34+ cells was also shared by all M-OST CMs (p˂0.05), as supported by significant increases with immature day-3 (mean ± SD of 18 ± 6, p˂0.02) and mature day-21 M-OST CMs (14 ± 5, p˂0.05) vs. control (8 ± 3, n=4). Conversely, expansions of TNC and CD34+ cells in MSC CM cultures were in-between that of control and M-OST CMs cultures. Interestingly, M-OST CMs also modulated the expansion of the HSPC compartment. Indeed, while the expansion of multipotent progenitors defined as CD34+CD45RA+ was promoted in control culture (ratio of 4.5 for CD34+CD45RA+/CD34+CD45RA- cells), M-OST CMs supported greater expansion of the more primitive CD34+CD45RA- HSPC subpopulation reducing the ratio to 3.3±0.4 for M-OST cultures (cumulative mean of 10 cultures, n=2). Moreover, the expansions of CD34+CD38- cells and of the long term HSC-enriched subpopulation (CD34+CD38-CD45RA-Thy1+) in M-OST CM cultures were respectively 2.7- and 2.8-fold greater than those measured in control cultures (n=2-4). Finally, the impact of M-OST CMs on the expansion of myeloid progenitors was investigated using a colony forming assay; expansion of myeloid progenitors were superior in all M-OST CM cultures (1.6±0.2 fold, n=2). In conclusion, our results demonstrate that M-OST rapidly acquire the expression of growth factors known to promote HSPC expansion. Moreover, the capacity of M-OST CMs to support the expansion of HSPCs appears to be a property shared by M-OST at various stages of maturation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1587-1587
Author(s):  
Il-Hoan Oh ◽  
Hyun-Kyung Choi

Abstract Mesenchymal stromal cells (MSCs) are characterized by heterogeneity in the proliferation/self-renewal potentials and hematopoietic supporting activity among subpopulations. Numerous studies have suggested that a primitive state of MSC subpopulation are correlated to its niche function to support hematopoietic stem cells (HSCs), but the mechanisms regulating primitive state of MSCs remains poorly understood. In the present study, we examined the role of a chromatin remodeling enzyme, chd1 in the maintenance of open chromatin and undifferentiated state of MSCs. We analyzed for expression in MSCs, the expression level of chd1 progressively decreased during in-vitro subculture (from 7 to 18 passages) in a manner proportional to the passage numbers. Moreover, chd1 expression was down regulated in the MSCs during their differentiation into adipogenic or osteogenic lineages, compared to proliferative state, indicating the correlations between MSC proliferation potentials and expression level of chd1. Next, we transduced human bone marrow-derived MSCs with shRNAs against chd1 and found that chd1 knock down MSCs (chd1-KD) exhibit significant loss of colony forming activity (CFU-F), decrease of cell proliferation and loss of multi-lineage differentiation towards osteogenic or adipogenic lineages. Moreover, chd1-KD MSCs exhibited lower level expression of pluripotency-related genes, oct-4, sox-2 and nanog, with concomitant increase of H3K9me3 on the promoters and decreased chromatin accessibility in the oct-4 promoter, suggesting that chd1 regulate open chromatin and multi-lineage potential of MSCs. However, KD of chd1 in MSCs did not affect the HSC-supporting activity of MSCs; human cord blood-derived CD34+ cells co-cultured on chd1-KD MSCs exhibited rather higher maintenance of primitive phenotype (CD34+90+) and higher repopulating activity in NOD/SCID-ɤC KO mice compared to those co-cultured on control group MSCs. Together, these results show that, while primitive state of MSCs are regulated by chromatin remodeling complex,chd1, the hematopoietic niche activity of MSCs is not directly influenced by the primitive state of MSCs, raising a questions on the prevailing notion that undifferentiated MSCs can better support hematopoietic function. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol Volume 12 ◽  
pp. 517-528
Author(s):  
Emilia Barreto-Duran ◽  
Claudia Camila Mejia-Cruz ◽  
Luis Fernando Jaramillo-Garcia ◽  
Efrain Leal-Garcia ◽  
Alfonso Barreto-Prieto ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1443-1443
Author(s):  
Duohui Jing ◽  
Nael Alakel ◽  
Martin Bornhauser ◽  
Gerhard Ehninger ◽  
Rainer Ordemann

Abstract Abstract 1443 Poster Board I-466 Background Hematopoietic stem cells (HSCs) are located mainly in the bone marrow interacting with a specific microenvironment called “stem cell niche”. The niche has been proven to be critical for stem cell regulation. Coculture with mesenchymal stromal cells (MSCs) has been used as an in vitro model to investigate the interaction between HSCs and MSCs. In our study we investigated the impact of normoxia and hypoxia on the distribution of HSC subsets with regard to their spatial localization in cocultures during ex-vivo expansion. Design and Methods Three HSC subsets are defined: (i) cells in supernatant (non-adherent cells); (ii) cells adhering on the MSC layer surface (phase-bright cells); (iii) cells beneath the MSC layer (phase-dim cells). Using pimonidazole binding we investigated the spatial distribution of hypoxic cells in various cell subsets. Cell cycle, cell division, immunophenotype and migratory capacity of the three HSC subsets under distinct oxygen tension were studied. In addition the impact of oxygen tension on HSCs via VEGF-A and SDF-1 were analyzed by ELISA and gene knockdown with siRNA. Results First we could show that phase-bright cells contained the highest proportion of cycling progenitors. In contrast, phase-dim cells divided much more slowly and retained a more immature phenotype. Next pimonidazole binding revealed that the most hypoxic area in the coculture is the compartment beneath MSC layer. Then we investigated the impact of hypoxia conditions on HSCs in cocultures. We could demonstrate that under hypoxic conditions phase-bright cells were significantly diminished and phase-dim cells were increased. Interestingly, the migratory capacity of phase-bright cells from cocultures performed under hypoxic conditions was consistently enhanced in comparison to normoxia (32.7 ±2.2.0% vs 17.6 ±2.6%, p<0.01). Surprisingly, the SDF-1 concentration was lower after hypoxic coculture (189 ±33μg/ml vs 352 ±40μg/ml, p<0.05). In contrast, the VEGF-A concentration was significantly increased compared to normoxic conditions (7.7 ±1.2ng/ml vs 4.5 ±1.0ng/ml, p<0.01). In addition we could demonstrate that the lower adhesion and higher migratory capacity of HSCs under hypoxia can be partially inversed by silencing VEGF-A with siRNA in MSCs. Conclusions Our data indicate that under our experimental conditions, the MSC surface is the dominant location where HSCs proliferate, whereas the compartment beneath the MSC layer seems to be a hypoxic niche dedicated to the maintenance of HSC stemness. The lower levels of SDF-1 in the supernatant may be explained by the increased internalization of SDF-1 by MSCs when cultured together with HSC. This hypothesis will require the concomitant analysis of protein and SDF-1 mRNA in MSC. In addition our data suggest that low oxygen tension facilitates HSC migration into the in-vitro niche provided by MSCs which preserves immaturity of HSCs and modifies the cytokine profile of MSCs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2401-2401
Author(s):  
Jingzhu Zhang ◽  
Daniel C. Link

The bone marrow microenvironment contains hematopoietic niches that regulate the proliferation, differentiation, and trafficking of hematopoietic stem/progenitors cells (HSPCs). These hematopoietic niches are comprised of a heterogeneous population of stromal cells that include, endothelial cells, osteoblasts, CXCL12-abundant reticular (CAR) cells, mesenchymal stem cells (MSCs), arteriolar pericytes, and sympathetic nerves. Emerging data suggest that specific stromal populations may regulate distinct types of HPSCs. Thus, it is important to have validated approaches to interrogate and target specific stromal cell populations. Prior studies have shown that Prx1-Cre, Osx-Cre, Lepr-Cre, and Nes-Cre broadly target mesenchymal stromal cells in the bone marrow. Here, we rigorously define the stromal cell populations targeted by two Cre-transgenes that are commonly used to target osteolineage cells (Ocn-Cre, and Dmp1-Cre) and introduce a new Cre-transgene (Tagln-Cre) that efficiently targets bone marrow pericytes. For each Cre-transgene, we performed lineage mapping using ROSA26Ai9/Ai9 mice, in which cells that have undergone Cre-mediated recombination express tdTomato. In some cases, we further crossed these mice to introduce the Cxcl12gfp transgene, which can be used to define GFP-bright CAR cells. Immunostaining of bone sections and flow cytometry were used to define the target stromal cell population(s) in these mice. Osteocalcin (Bglap, Ocn) is primarily expressed in mature osteoblasts. Accordingly, Ocn-Cre is widely used to specifically target osteoblasts. However, our lineage mapping studies show that Ocn-Cre targets not only all osteoblasts, but also 72 ± 4.0% of CAR cells. Ocn-Cre also targets a subset of NG2+ arteriolar pericytes. Dentin matrix acidic phosphoprotein 1 (Dmp1) is expressed primarily in osteocytes, and Dmp1-Cre has been widely used to specifically target osteocytes. However, we show that Dmp1-Cre also efficiently targets endosteal osteoblasts and approximately 40% of CAR cells. To target bone marrow pericytes, we tested several Cre-transgenes, ultimately focusing on Tagln-Cre. Transgelin (Tagln, SM22a) is broadly expressed in pericytes, smooth muscle cells, and cardiomyocytes. Lineage-mapping studies show that Tagln-Cre targets all arteriolar and venous sinusoidal pericytes in the bone marrow. It also targets osteoblasts and 75 ± 5.2% of CAR cells. There are several recent studies that have ascribed specific functions to osteoblasts or osteocytes based on targeting using Ocn-Cre or Dmp1-Cre, respectively. In light of our data, these conclusions need to be re-evaluated. Ocn-Cre, Dmp1-Cre, and Tagln-Cre each target a subset of CAR cells. Studies are underway to determine whether these CAR subsets have unique expression profiles and functions. Finally, Talgn-Cre represents a new tool for investigators in the field to efficiently target bone marrow pericytes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5516-5516
Author(s):  
Salar Abbas ◽  
Aparna Venkatraman ◽  
Sanjay Kumar ◽  
Archana Kini ◽  
Marie Therese Manipadam ◽  
...  

Abstract Introduction:The myelodysplastic syndromes (MDS) are clonal disorders characterized by cytopenias and abnormal hematopoiesis. Though there are reports of perturbations in the hematopoietic stem cells (HSC) and the mesenchymal stromal cells (MSC) as well as other elements of the bone marrow (BM) niche in an instructive or permissive manner leading to the genesis of ineffective hematopoiesis in this condition, most studies have evaluated single elements. Here we demonstrate altered HSC, MSC and the vascular niche elements in patients with MDS - Refractory cytopenia with multilineage dysplasia (RCMD). Methods and results: Bone marrow aspirates from patients with RCMD (n=12, mean age 54.33±14.51 years) were compared with those from age-matched controls (n=18, mean age 47.78±18.60 years) who had a 'normal' marrow obtained for other diagnostic purposes. Cytogenetic analysis showed abnormal karyotypes in seven patients and normal karyotypes in five. The abnormalities seen were as follows: deletion 7q/monosomy 7 (four patients), deletion 5q (three patients), loss of Y (two patients), monosomy 5, deletion 20q (one each), complex karyotypes (four patients). Phenotypic enumeration of HSPCs revealed a marked decrease in the frequency of highly purified HSCs (Lin-CD34+CD38-CD90+CD45RA-) in RCMD (0.04135±0.01748%, n=11) when compared to controls (0.3168±0.05266%, n=13, p<0.0001) (Figure 1). Patients with RCMD also had increased common myeloid progenitors (CMP) (3.221 ± 0.7478%, n=8) compared to control (1.243±0.4463%, n=10, p<0.0302) and loss of granulocyte-macrophage progenitors (GMP) (0.4863±0.1638%, n=8) compared to controls (2.047±0.5422%, n=10, p<0.0242) (Figure 2). Assessment of the frequency of de novo MSCs (CD31-CD45/CD71- population) expressing CD271 and/or CD146, indicating the more primitive population, in total nucleated cells showed increased CD271+CD146-MSCs (0.632±0.2, n=5) compared to controls (0.2000±0.05158, n=6, p<0.0401) as also the CD271+CD146+ MSCs (0.2900±0.09803, n=5) in RCMD patients when compared to controls (0.02861±0.01354, n=6, p<0.0172) (Figure 3). We also evaluated in vitro cultured MSCs (P4) in these patients. CD271+CD146+ MSCs within total cultured MSCs were higher (0.1900±0.06429%, n=3) than in controls (0.0040±0.0040%, n=3, p<0.0447). RCMD MSCs had significantly lower proliferation index (32±3.7%, n=3) compared to controls (60±9.2%, n=3, p<0.0479). Cell cycle analysis of MSC showed significantly lower numbers in G0 in RCMD (0.1567 ± 0.07881 %, n=3) compared to control (0.6400 ± 0.1007 %, n=3; p<0.0194). Apoptosis was much higher in RCMD MSCs (2.700±0.8007%, n=3) compared to controls (0.03633±0.01802%, n=3, P<0.0292). No significant differences in expression profile of stem cell maintenance related cytokines and growth factors (CXCL12a, SCF, VEGF, ANGPT and LIF ) or components of Notch (Notch1, Notch3, Jagged-1, Delta like-1 and Hes1) and Wnt (Dkk1 and Dkk-2) pathways were found between RCMD and control MSCs within the limited numbers evaluated. Interestingly, unlike controls, immunofluorescence imaging of bone marrow trephine from MDS-RCMD revealed CD271+CD146+ MSCs co-localized with sinusoids and in direct contact with CD34+ (<5% blast) HSC/progenitor cells (HSPCs). Conclusion: Our data shows that in patients with MDS-RCMD, both qualitative and quantitative abnormalities exist in the HSC, HSPC and niche elements. We also show that the cytopenia could be related to decreased numbers of primitive HSCs and a differentiation arrest at the CMP stage. Primitive MSCs are reduced, and those that exist show poorer proliferative and survival features. Further studies are needed to understand the cause and effect relationship of these changes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2425-2425
Author(s):  
Felicia Ciuculescu ◽  
Melanie Giesen ◽  
Erika Deak ◽  
Erhard Seifried ◽  
Tanja Nicole Hartmann ◽  
...  

Abstract Mesenchymal Stromal Cells (MSCs) are being developed as a cellular therapeutic, and used clinically used for induction of immunomodulation and suppression of inflammation after application via the intravenous route. However, relatively little is known on how MSCs interact with the vessel wall to induce tissue-specific egress. To elucidate potentially underlying mechanisms, we analyzed human bone-marrow derived MSCs in parallel plate flow chambers and characterized their interaction with endothelial cells or immobilized endothelial ligands as to tethering and rolling, sudden arrest, adhesion strengthening, and transendothelial migration. Flow cytometric analysis of MSCs confirmed the known expression predominantly of integrins α4, α5 and β1, and revealed only low detectable levels of a range of chemokine receptors including CCR1, 5, 6, 7 and CXCR1, 4 and 5. To investigate a potential role of chemokines in the adhesion process of MSCs, we first seeded MSCs without shear onto immobilized recombinant (r) VCAM-Fc fusion protein without or with co-immobilized chemokines in parallel plate flow chambers for 3 min, and subsequently started flow by stepwise increasing shear stress from 0.35 to 15 dynes/cmE2. Co-immobilization of various chemokines including CCL15/HCC-2, a known activator of CCR1 and 3, CCL20/LARC (of CCR6), CCL19/ELC (of CCR7), CXCL8/IL-8 (of CXCR1/2), CXCL12/SDF-1 (of CXCR4) and CXCL13/BCA-1 (of CXCR5) with rVCAM-1 increased shear-resistant binding of MSCs by up to four-fold compared to controls on rVCAM alone, with only moderate increases through CXCL8/IL-8 and CXCL12/SDF-1 and strongest effects through CCL19/ELC and CCL20/LARC. The reverse was observed in hematopoetic progenitor cells, which responded best to CXCL12/SDF-1. No or little adhesion of MSCs to rICAM-1 was observed under analogous conditions. We next chose to study CXCL12/SDF-1 and CCL19/ELC in more detail on intact endothelial cells. MSCs were flushed through parallel plate flow chambers on Human Umbilical Vein Endothelial Cells (HUVECs) at an initial shear stress of 0.35 or 0.5 dyn/cmE2 for 1 min followed by an elevated shear stress of 5 dyn/cmE2 for further 10 min. We found that rolling of MSCs was increased after pre-treatment of HUVECs with TNF-α but only few spontaneous arrests were obsereved under these conditions. However, when HUVECs were additionally overlaid with CXCL12/SDF-1 or CCL19/ELC, MSCs arrested firmly on the endothelium. Pretreatment of MSCs with function-blocking anti-CD44 or anti-VLA-4 antibodies, or pre-treatment of HUVECs with anti-E-selectin or anti-VCAM-1 antibodies partially (anti-CD44, -E-selectin) or completely (anti-VLA-4, -VCAM-1) suppressed arrest of MSCs. Analysis of adhesion strengthening on HUVECs as well as on immobilized rVCAM-1 confirmed the mediation of shear-resistant binding of MSCs by CXCL12/SDF-1 and CCL19/ELC. Pre-incubation of MSCs with pertussis toxin also suppressed both, sudden arrest and firm adhesion, indicating a role of signalling through G protein coupled receptor via Gαi. Finally, MSCs were able to transmigrate chemokine-coated HUVECs but not untreated. We conclude that MSCs respond to individual chemokines with different efficiencies to induce arrest and firm adhesion before their transendothelial migration. MSCs were found highly responsive to chemokines which physiologically act on lymphocytes to enter secondary lymphoid organs or sites of inflammation. This points to a preference of MSCs to follow lymphocyte egress pathways and reach sites of inflammation to induce immunomodulation and –suppression.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4086-4086
Author(s):  
Peilong Lai ◽  
Jianyu Weng ◽  
Chenwei Luo ◽  
Rong Guo ◽  
Zesheng Lu ◽  
...  

Abstract Abstract 4086 Background: Cutaneous chronic graft versus host disease (cGVHD) is an increasingly common complication in long term survivors of hematopoietic stem cell transplantation, but currently available therapies have demonstrated limited efficacy. Furthermore, Mesenchymal Stromal Cells (MSCs) have been reported to be effective in various immune-mediated disease models, but their therapeutic potentials versus cutaneous cGVHD have not been determined. Methods: We enrolled 23 patients suffering from cutaneous cGVHD who had failed to respond to conventional immunosuppressive therapy or relapsed after reduction of prednisone. They received intravenous in vitro expanded bone marrow (BM)-derived MSCs. The median follow-up period was 90 days (range 60–146 days).The dermal manifestations of cGVHD were monitored, and a score was given to the cutaneous response by the physician. B lymphocyte subsets and ratio of CD4/CD8 were detected by flow cytometry before the first and after the last MSC infusion. The plasmatic levels of TNF-α, sICAM-1 and Th1 or Th2 factors, including IL-2 and IL-4, were measured by enzyme-linked immunosorbent assay (ELISA). Results: Nineteen patients (82.61%) had skin symptom abatement after MSCs treatment with the skin scores improved according to the NIH criteria. Most patients had healing of skin ulceration, regression on skin findings and increased flexibility of involved joint. Steroid sparing or discontinuation of immunosuppressive medications was possible in all of the patients. Clinical improvement was accompanied by increasing levels of CD19+CD5+ B cells and elevated ratio of CD4/CD8. In addition, after the last MSCs treatment, the patients had significantly higher levels of IL-2 compared to before the first MSC infusion, while they had lower levels of TNF-α, sICAM-1 and IL-4. Conclusion: MSCs infusion represents an effective treatment option for cutaneous chronic GVHD. This is of high interest in patients suffering from cutanous cGVHD resistant to conventional immunosuppressive therapy. MSCs alter allo-reactivity by affecting CD19+CD5+ B cells, possibly normalize the ratio of CD4/CD8 and Th1/Th2 and decreased the level of proinflammatory cytokine TNF-α and proadhesive cytokine sICAM-1. Our study demonstrates CD19+CD5+ B cells could be a possible target for therapeutic intervention in cutaneous cGVHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2249-2249 ◽  
Author(s):  
Igor B Resnick ◽  
Polina Stepensky ◽  
Michael Y. Shapira ◽  
Claudine Barkatz ◽  
Gregory Elkin ◽  
...  

Abstract Abstract 2249 Poster Board II-226 BACKGROUND. Allogeneic hematopoietic stem cell transplantation (HSCT) is being used to treat a range malignant and nonmalignant conditions. However, it often causes potentially lethal Graft-versus-Host Disease (GVHD). Several studies revealed that mesenchymal stromal cells (MSCs) from human bone marrow can down regulate GVHD after HSCT. METHODS. MSCs were obtained from BM, expanded and characterized by their morphology, immunophenotype, immunosuppressive potential for autologous, partially and fully mismatched lymphocytes. Twenty five patients (pts) got 39 (range, 1 to 4) MSCs infusions for 27 episodes of acute GVHD, which was defined as steroid resistant grade IV aGVHD in all but one patient. RESULTS. GvHD characteristics. Acute GVHD started from day +6 to d +46 from HSCT (median d+18) excluding single pt in whom it occurred on d+150. In 2 pts disease manifested in a hyper acute form before the engraftment (on d+6 and d+8, respectively). In all but one pt GI GVHD was defined as grade IV with full clinical picture of it. Skin GVHD accompanied GI symptoms in 17 pts and 13 was > grade II, in 4 grade IV. Liver symptoms presented in 13 pts, one pt was determinate as VOD. In 3 pts liver involvement was defined as grade IV. In all 24 of 25 pts had GVHD grade IV 4 and one grade III. Previous conventional anti GVHD therapies included: MP in all pts, MTX, various calcineurin inhibitors, MMF, ECP, serotherapy. Only 3 pts showed temporary limited partial response. Treatment with MSCs. The 1st infusion with MSC was given on day +32.5 (range, 8 to 74); d+50 (range, +28 to +180) post diagnosis of aGVHD and HSCT HSCT, respectively. In 22 evaluable pts we treated 24 separate episodes of GVHD: 22 first episodes and in 2 pts relapse of GVHD. In 24 of 39 cases treatment was performed with frozen MSC and in 15 with fresh cultured cells. In 37/39 cases passages 1 to 3 were used. Median number of infused cells was 1 (range, 0.3 to 3.1) x10 6 per kg of pt body weight in each treatment. Initial response was seen in 17/24 episodes of aGVHD (70.8%), in 8 partial (PR) and in 9 complete (CR). Two pts experienced GVHD relapse after achieving CR and very good PR: one case was treated successfully while another was resistant to MSCs. The latest pt with grade IV aggressive liver GVHD received MSCs injections intra hepatic arteries under radiological control following IRB approval with no anti GVHD effect. The procedure was uneventful with no evidence of microembolisation, no changes of blood flow or deterioration of liver tests (cytolysis). We observed a trend for better effect with higher MSC cell dose: total and per first infusion MSCs dose (1.93±1.28 vs. 1.23±0.50, p=0.078; 1.28±0.79 vs. 0.88±0.28, p=0.086). In all cases effect was seen after the first procedure. No difference was noted in the anti GVHD activity between fresh vs. frozen MSCs. There were no immediate or late toxicity or side effects. Overall survival. 14/25 patients are alive up to 20 months follow up. 8/11 pts, died from GVHD (4 within 1-10 d from MSCs infusion), and 3 from other unrelated causes including TTP-1, DAH-1 and disease progression-1. CONCLUSION. Treatment with MSCs seems to be a new novel type of therapy for steroids resistant GVHD with promising preliminary results and low toxicity. Obviously multicenter 2 arm randomized study is in need to confirm these encouraging. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5410-5410
Author(s):  
Mark van der Garde ◽  
Melissa Van Pel ◽  
Jose Millan Rivero ◽  
Alice de Graaf-Dijkstra ◽  
Manon Slot ◽  
...  

Abstract Co-transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) and mesenchymal stromal cells (MSC) enhances HSPC engraftment. For these applications, MSC are mostly obtained from bone marrow. However, MSC can also be sourced from Wharton's jelly (WJ) of the human umbilical cord, which as a 'waste product', is cheaper to acquire and without significant burden to the donor. Here, we evaluated the ability of WJ MSC to enhance HSPC engraftment. First, we compared cultured human WJ MSC with human bone marrow-derived MSC (BM MSC) for in vitro marker expression, immunomodulatory capacity and differentiation into three mesenchymal lineages. Although we confirmed that WJ MSC have a more restricted differentiation capacity, both WJ MSC and BM MSC expressed similar levels of surface markers and exhibited similar immune inhibitory capacities. Co-transplantation of either WJ MSC or BM MSC with CB CD34+ cells into NOD-SCID mice showed faster recovery of human platelets and CD45+ cells in the peripheral blood and a 3-fold higher engraftment in the BM, blood and spleen six weeks after transplantation when compared to transplantation of CD34+ cells alone. Upon co-incubation, both MSC sources increased the expression of adhesion molecules on CD34+ cells, although SDF-1-induced migration of CD34+ cells remained unaltered. Interestingly, there was an increase in CFU-GEMM when CB CD34+ cells were cultured on monolayers of WJ MSC in the presence of exogenous thrombopoietin, and an increase in BFU-E when BM MSC replaced WJ MSC in such cultures. Our results suggest that WJ MSC is likely to be a practical alternative for BM MSC to enhance CB CD34+ cell engraftment. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document