Mesenchymal Stem Cells Can Be Mobilized Into Peripheral Blood by Short-Term Hypoxia: a Possible Role of HIF-1α.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1450-1450
Author(s):  
Lizhen Liu ◽  
Qin Yu ◽  
Jie Lin ◽  
Weijie Cao ◽  
Xiaoyu Lai ◽  
...  

Abstract Abstract 1450 Background: Mesenchymal stem cells (MSCs) constitute a population of multipotential cells giving rise to adipocytes, osteoblasts and chondrocytes. Combining with their engraftment promoting capacity and immunosuppressive property, MSCs may be therapeutically useful for haematopoietic stem cell transplantation. There is growing evidence that these cells can, under the right circumstances, enter peripheral circulation. Previous study revealed that MSCs are mobilized into peripheral blood (PB) by 3 weeks of chronic hypoxia, but the mobilization effect of short-term hypoxia and the underlying mechanisms are currently unknown. In this study, we used rat model to determine whether short-term hypoxia can mobilize MSCs into PB and investigated the related factors which may regulate the mobilization process. Design and Methods: Rats were housed in a hypoxic chamber (FiO2=10%) for 1, 2, 3, 5, 7 and 14 days, respectively, while control ones were housed in normoxic environment for equal periods. To quantify the number of MSCs and evaluate mobilization efficiency, PB and bone barrow (BM) samples of each group were collected and colony-forming unit fibroblast (CFU-F) assays were performed. Mobilized PB derived MSCs were identified by immunophenotype and trilineage differentiation. Since BM is the main reservoir and typical microenvironment of MSCs, we investigated the response of BM environment exposed to hypoxia which may potentially facilitate MSCs mobilization. Hypoxia-inducible factor 1α (HIF-1α) expression of BM cells was detected by Western blot; vascular endothelial growth factor (VEGF) in BM was qualified by ELISA and Immunohistochemistry (IHC). To evaluate the change of BM sinusoid vessels (BMSVs), VEGFR3 was stained by IHC and positive vessels were counted. The levels of stromal cell-derived factor-1α (SDF-1α) and VEGF in PB were tested by ELISA. Moreover, we compared migration capacity of MSCs in hypoxic condition (PO2=1% or 8%) with normal condition (PO2=21%) in vitro using Transwell assay. Results: We found that MSCs were mobilized into PB by exposing to short-term hypoxia (2d) and the CFU-F frequency was 5.80±0.58 vs. 1.40±0.24 CFU-Fs per 3×106 cells (p<0.05, n=5). From 2d to 14d of hypoxic exposure, the number of CFU-Fs mobilized in PB of hypoxic group was gradually increased in a time dependent manner. However, no significant differences were observed in bone marrow CFU-Fs among varies groups (P>0.05). Mobilized PB derived adherent cells were positive for CD90, CD29 and CD44, but negative for CD34 and CD45 and they could differentiate into adipocyte, osteoblast, and chondrocyte, which indicated that mobilized PB-derived cells are bona fide MSCs. What's more, we showed here that during hypoxic exposure, HIF-1α was stabilized and expressed continuously in BM of rats which is a main niche of MSCs. Stabilization and up-regulation of HIF-1α suggested that BM is hypoxia-sensitive and during hypoxic exposure it became a lower oxygen environment (PO2<1%). Previous studies have proved that VEGF and SDF-1α are directly regulated by the transcription factor HIF-1α. Our results showed that, induced by HIF-1α, VEGF was elevated from 2d to 7d in the BM of hypoxic rats which may increase BM vascular permeability and induce vasodilatation; VEGFR3(+) BMSVs increased in 7d and 14d hypoxic BM which may further facilitate the egress of MSCs. SDF-1α in PB increased from 2d to 14d, especially 7d of hypoxia (1976.7±148.1 vs. 663.6±56.7pg/ml, P<0.01). In addition, exposure of MSCs to low oxygen (8% PO2) significantly promoted their in vitro migration and a further increase was observed under lower oxygen condition (1% PO2). MSCs migrated more rapidly in response to SDF-1α exposed to hypoxia. Conclusion: Taken together, we show here that MSCs can be mobilized into PB by short-term hypoxia and the mobilization efficacy increased in a time dependent manner. Our results suggest the mechanisms of hypoxia inducing MSCs mobilization relate to the lower oxygen milieu of BM and stabilization of HIF-1α may play a pivotal role in MSCs mobilization. Our data provide meaningful clues to clarify the mechanisms of MSCs mobilization and important evidence for further exploring the exact agents that of clinical use. Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Hong Kyung Lee ◽  
Eun Young Kim ◽  
Hyung Sook Kim ◽  
Eun Jae Park ◽  
Hye Jin Lee ◽  
...  

Systemic lupus erythematosus (SLE) is an autoimmune disease, which is characterized by hyperactivation of T and B cells. Human mesenchymal stem cells (hMSCs) ameliorate the progression of SLE in preclinical studies using lupus-prone MRL.Faslpr mice. However, whether hMSCs inhibit the functions of xenogeneic mouse T and B cells is not clear. To address this issue, we examined the in vitro effects of hMSCs on T and B cells isolated from MRL.Faslpr mice. Naïve hMSCs inhibited the functions of T cells but not B cells. hMSCs preconditioned with IFN-γ (i) inhibited the proliferation of and IgM production by B cells, (ii) attracted B cells for cell–cell interactions in a CXCL10-dependent manner, and (iii) inhibited B cells by producing indoleamine 2,3-dioxygenase. In summary, our data demonstrate that hMSCs exert therapeutic activity in mice in three steps: first, naïve hMSCs inhibit the functions of T cells, hMSCs are then activated by IFN-γ, and finally, they inhibit B cells.


2019 ◽  
Vol 2019 ◽  
pp. 1-14 ◽  
Author(s):  
Barbara Hersant ◽  
Mounia Sid-Ahmed ◽  
Laura Braud ◽  
Maud Jourdan ◽  
Yasmine Baba-Amer ◽  
...  

Chronic and acute nonhealing wounds represent a major public health problem, and replacement of cutaneous lesions by the newly regenerated skin is challenging. Mesenchymal stem cells (MSC) and platelet-rich plasma (PRP) were separately tested in the attempt to regenerate the lost skin. However, these treatments often remained inefficient to achieve complete wound healing. Additional studies suggested that PRP could be used in combination with MSC to improve the cell therapy efficacy for tissue repair. However, systematic studies related to the effects of PRP on MSC properties and their ability to rebuild skin barrier are lacking. We evaluated in a mouse exhibiting 4 full-thickness wounds, the skin repair ability of a treatment combining human adipose-derived MSC and human PRP by comparison to treatment with saline solution, PRP alone, or MSC alone. Wound healing in these animals was measured at day 3, day 7, and day 10. In addition, we examined in vitro and in vivo whether PRP alters in MSC their proangiogenic properties, their survival, and their proliferation. We showed that PRP improved the efficacy of engrafted MSC to replace lost skin in mice by accelerating the wound healing processes and ameliorating the elasticity of the newly regenerated skin. In addition, we found that PRP treatment stimulated in vitro, in a dose-dependent manner, the proangiogenic potential of MSC through enhanced secretion of soluble factors like VEGF and SDF-1. Moreover, PRP treatment ameliorated the survival and activated the proliferation of in vitro cultured MSC and that these effects were accompanied by an alteration of the MSC energetic metabolism including oxygen consumption rate and mitochondrial ATP production. Similar observations were found in vivo following combined administration of PRP and MSC into mouse wounds. In conclusion, our study strengthens that the use of PRP in combination with MSC might be a safe alternative to aid wound healing.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2563-2563
Author(s):  
Fernando Fierro ◽  
Thomas Illmer ◽  
Duhoui Jing ◽  
Philip Le Coutre ◽  
Gerhard Ehninger ◽  
...  

Abstract Recent data show that the tyrosine kinase inhibitor Imatinib mesylate (IM) also affects normal hematopoietic stem cells (HSC), T lymphocyte activation and dendritic cell function not relying on the specific inhibition of bcr-abl activity. Mesenchymal stem cells (MSC) have been identified in the bone marrow (BM) as multipotent non-hematopoietic progenitor cells that differentiate into osteoblasts, adipocytes, chondrocytes, tenocytes, skeletal myocytes, and cells of visceral mesoderm. MSC interact with HSC, influencing their homing and differentiation through cell-cell contact and the production of factors including chemokines We evaluated possible effects of IM in vitro on human bone marrow-derived MSC. Screening the activity of fourty-two receptor tyrosine kinases by a phospho-receptor tyrosine kinase (RTK)-array revealed an exclusive inhibition of platelet-derived growth factor receptor (PDGFRβ) by IM which consequently affects downstream targets of PDGFRβ as Akt and Erk1/2 signalling pathways in a concentration and time dependent manner. Furthermore, perinuclear multivesicular bodies harbouring PDGFRβ were found within 18–20 hours culture of MSC in the presence of 5 μM IM. Cell proliferation and clonogenicity (evaluated as the capability to form colony forming units - fibroblasts (CFU-F)) of MSC were significantly inhibited by IM in a concentration dependent fashion. IM inhibits significantly the differentiation process of MSC into osteoblasts as evaluated by decreased alkaline phosphatase activity and reduced calcium phosphate precipitates. In contrary, differentiation of MSC into adipocytes was strongly favoured in presence of IM. All these functional deficits described, probably contribute to an observed 50% reduction in the support of clonogenic hematopoietic stem cells, as evaluated by a long term culture-initiating cells (LTC-IC)-based assay. In summary our experiments show that IM inhibits the capacity of human MSC to proliferate and to differentiate into the osteogenic lineage, favouring adipogenesis. This effect is mainly mediated by an inhibition of PDGFRβ autophosphorylation leading to a more pronounced inhibition of PI3K/Akt compared to Erk1/2 signalling. This work confirms the role of PDGFRβ recently described for the proliferation and differentiation potential of MSC and provides a first possible explanation for the altered bone metabolism found in certain patients treated with IM.


Author(s):  
Ying Zhang ◽  
Jing Zhu ◽  
Hao Xu ◽  
Qin Yi ◽  
Liang Yan ◽  
...  

Mesenchymal stem cells (MSCs) are promising tools for cancer therapy, but there is a risk of malignant transformation in their clinical application. Our previous work revealed that the paracrine protein S100B in the glioma microenvironment induces malignant transformation of MSCs and upregulates intracellular S100B, which could affect cell homeostasis by interfering with p53. The purpose of this study was to investigate whether extracellular S100B can be internalized by MSCs and the specific endocytic pathway involved in S100B internalization. By using real-time confocal microscopy and structured illumination microscopy (SIM), we visualized the uptake of fluorescently labeled S100B protein (S100B-Alexa488) and monitored the intracellular trafficking of internalized vesicles. The results showed that S100B-Alexa488 was efficiently internalized into MSCs in a time-dependent manner and transported through endolysosomal pathways. After that, we used chemical inhibitors and RNA interference approaches to investigate possible mechanisms involved in S100B-Alexa488 uptake. The internalization of S100B-Alexa488 was inhibited by pitstop-2 or dyngo-4a treatment or RNA-mediated silencing of clathrin or dynamin, and the lipid raft-mediated endocytosis inhibitors nystatin and MβCD. In conclusion, our findings show that clathrin and lipid rafts contribute to the internalization of S100B-Alexa488, which provides promising interventions for the safe application of MSCs in glioma therapy.


2021 ◽  
Vol 21 ◽  
Author(s):  
Qiu-Yun Li ◽  
Juan Chen ◽  
Yong-Heng Luo ◽  
Wei Zhang ◽  
En-Hua Xiao

Objective: The treatment of liver failure by stem cell transplantation has attracted growing interest. Herein, we aim to explore the role of sodium butyrate (NaB) in the hepatic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) under liver-specific factors induction in vitro and vivo. Materials & Methods: We isolated BM-MSCs from the mononuclear cell fraction of rabbit bone marrow samples, and identified the cells by Immunophenotypic analysis. We investigated the effects of different concentrations and induction conditions. The histone deacetylase inhibitor NaB induced hepatic differentiation of BM-MSCs under liver-specific factors induction in vitro. Morphological features, liver-specific gene and protein expression, and functional analyses in vitro and vivo were performed to evaluate the hepatic differentiation of BM-MSCs. Results: Our results showed that pre-treated NaB inhibited the expression of liver-specific protein in a dose-dependent manner. The induction efficiency of NaB with 24h pre-treatment was higher than that of NaB continuous intervention. 0.5 mM 24h NaB pre-treated cells can improve liver tissue damage in vivo. And the liver ALB, AAT and the serum TP were significantly increased, while the serum ALT was significantly reduced. Conclusion: Continuous NaB treatment can inhibit BM-MSCs proliferation in a dose-dependent manner at a certain concentration range. 0.5 mM 24h pre-treatment of NaB enhanced differentiation of BM-MSCs into hepatocytes and improves liver injury in vitro and vivo.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3186-3186
Author(s):  
Andrew W. Lee ◽  
Michael D. Milsom ◽  
Yi Zheng ◽  
Jose A. Cancelas ◽  
David A. Williams

Abstract Fanconi anemia (FA) is a genetic syndrome characterized by development of progressive bone marrow failure and cancer predisposition. The difficulty in harvesting FA HSC and their fragility during subsequent in vitro manipulations has proven a confounding factor in attempted gene therapy of this disease. Mobilization of HSC/P in FA patients is poor (Croop et al. Blood, 2001) probably due to HSC deficiency. We have previously shown that the genetic deletion of the Rac GTPases 1 and 2 results in an increase in circulating hematopoietic stem cells and progenitors (HSC/P) (Gu et al. Science 2003). In addition, administration of a single dose of a small molecule inhibitor of Rac GTPases, NSC23766, results in a transient mobilization of engraftable stem cells (Cancelas et al. Nat. Med., 2005). Here, we analyzed the role of NSC23766 in mobilizing HSC/P in FA A (Fanca −/ −) mice (Cheng et al., Hum. Mol. Genet., 2000; kindly provided by M. Grompe, OHSU). First, we validated that this murine model of FA demonstrated a stem cell phenotype by a competitive repopulation assay of BM HSC. We found that Fanca −/ − HSC contribute decreased chimerism in short-term engraftment (52.6 ± 2.6% donor engraftment) compared to wild-type (WT) controls (63.8 ± 1.0%, respectively, p < 0.005). BM and spleen homing of Fanca −/ − HSC/P at sixteen-hours post infusion was not impaired (7.0% in BM and 6.1% in spleen) compared to WT mice (7.8% in BM and 5.4% in spleen) and there was no difference in expression of CXCR4, a4-integrin, a5-integrin and L-selectin between Lin−/c-kit+/Sca-1+ BMC and mobilized PBC derived from Fanca −/ − and WT mice, also supporting an intrinsic HSC defect. We then analyzed the ability of NSC23766, alone or in combination with G-CSF, to mobilize HSC. We observed that Fanca −/ − mice also show an impaired mobilization response to G-CSF administration (200 mcg/Kg/day for five days), which can be partially rescued by administering a single dose of NSC23766, 6 hours before peripheral blood harvest (Table1). We additionally demonstrated the impaired engraftment of in vitro manipulated Fanca −/ − BMC in a competitive transplant assay. This engraftment defect could be completely ameliorated by treatment with Diprotin A (5.9±2.0% donor engraftment untreated vs. 12.0±4.4% treated; p value = 0.01). Diprotin A is an inhibitor of CD26 peptidase which has been shown to cleave SDF1alpha/CXCL12. The combined use of G-CSF and NSC23766 may constitute a future novel approach to induce mobilization of Fanconi anemia HSC and, when coupled with Diprotin A treatment, could act to enhance the engraftment of cells undergoing genetic correction. Table 1. Competitive Repopulating Units (normalized to WT GCSF) Short Term Engraftment (+1 month) Long Term Engraftment (+4 months) * p value < 0.05, compared to WT GCSF; ** p value < 0.001, compared to WT GCSF, *** p value < 0.05 compared to Fanca −/ − GCSF WT GCSF (%) 100 ± 34.08 100 ± 52.34 WT GCSF + NSC 23766 (%) 77.43 ± 37.11 53.34 ± 54.61 Fanca −/− GCSF (%) 11.25 ± 5.92 * 2.54 ± 2.59 ** Fanca −/− GCSF + NSC 23766 (%) 24.45 ± 1.89 39.01 ± 29.47 ***


2015 ◽  
Vol 2015 ◽  
pp. 1-9 ◽  
Author(s):  
Isaac Perea-Gil ◽  
Marta Monguió-Tortajada ◽  
Carolina Gálvez-Montón ◽  
Antoni Bayes-Genis ◽  
Francesc E. Borràs ◽  
...  

Cell-based strategies to regenerate injured myocardial tissue have emerged over the past decade, but the optimum cell type is still under scrutiny. In this context, human adult epicardial fat surrounding the heart has been characterized as a reservoir of mesenchymal-like progenitor cells (cardiac ATDPCs) with potential clinical benefits. However, additional data on the possibility that these cells could trigger a deleterious immune response following implantation are needed. Thus, in the presented study, we took advantage of the well-established low immunogenicity of umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) to comparatively assess the immunomodulatory properties of cardiac ATDPCs in anin vitroallostimulatory assay using allogeneic mature monocyte-derived dendritic cells (MDDCs). Similar to UCBMSCs, increasing amounts of seeded cardiac ATDPCs suppressed the alloproliferation of T cells in a dose-dependent manner. Secretion of proinflammatory cytokines (IL6, TNFα, and IFNγ) was also specifically modulated by the different numbers of cardiac ATDPCs cocultured. In summary, we show that cardiac ATDPCs abrogate T cell alloproliferation upon stimulation with allogeneic mature MDDCs, suggesting that they could further regulate a possible harmful immune responsein vivo. Additionally, UCBMSCs can be considered as valuable tools to preclinically predict the immunogenicity of prospective regenerative cells.


2018 ◽  
Vol 128 (1) ◽  
pp. 287-295 ◽  
Author(s):  
Jonathan G. Thomas ◽  
Brittany C. Parker Kerrigan ◽  
Anwar Hossain ◽  
Joy Gumin ◽  
Naoki Shinojima ◽  
...  

OBJECTIVEMesenchymal stem cells (MSCs) have been shown to localize to gliomas after intravascular delivery. Because these cells home to areas of tissue injury, the authors hypothesized that the administration of ionizing radiation (IR) to tumor would enhance the tropism of MSCs to gliomas. Additionally, they sought to identify which radiation-induced factors might attract MSCs.METHODSTo assess the effect of IR on MSC migration in vitro, transwell assays using conditioned medium (CM) from an irradiated commercially available glioma cell line (U87) and from irradiated patient-derived glioma stem-like cells (GSCs; GSC7-2 and GSC11) were employed. For in vivo testing, green fluorescent protein (GFP)-labeled MSCs were injected into the carotid artery of nude mice harboring orthotopic U87, GSC7-2, or GSC17 xenografts that were treated with either 0 or 10 Gy of IR, and brain sections were quantitatively analyzed by immunofluorescence for GFP-positive cells. These GSCs were used because GSC7-2 is a weak attractor of MSCs at baseline, whereas GSC17 is a strong attractor. To determine the factors implicated in IR-induced tropism, CM from irradiated GSC7-2 and from GSC11 was assayed with a cytokine array and quantitative ELISA.RESULTSTranswell migration assays revealed statistically significant enhanced MSC migration to CM from irradiated U87, GSC7-2, and GSC11 compared with nonirradiated controls and in a dose-dependent manner. After their intravascular delivery into nude mice harboring orthotopic gliomas, MSCs engrafted more successfully in irradiated U87 (p = 0.036), compared with nonirradiated controls. IR also significantly increased the tropism of MSCs to GSC7-2 xenografts (p = 0.043), which are known to attract MSCs only poorly at baseline (weak-attractor GSCs). Ionizing radiation also increased the engraftment of MSCs in strong-attractor GSC17 xenografts, but these increases did not reach statistical significance. The chemokine CCL2 was released by GSC7-2 and GSC11 after irradiation in a dose-dependent manner and mediated in vitro transwell migration of MSCs. Immunohistochemistry revealed increased CCL2 in irradiated GSC7-2 gliomas near the site of MSC engraftment.CONCLUSIONSAdministering IR to gliomas enhances MSC localization, particularly in GSCs that attract MSCs poorly at baseline. The chemokine CCL2 appears to play a crucial role in the IR-induced tropism of MSCs to gliomas.


Sign in / Sign up

Export Citation Format

Share Document