Skip to main content

Co-administration of human MSC overexpressing HIF-1α increases human CD34+ cell engraftment in vivo

Abstract

Background

Poor graft function or graft failure after allogeneic stem cell transplantation is an unmet medical need, in which mesenchymal stromal cells (MSC) constitute an attractive potential therapeutic approach. Hypoxia-inducible factor-1α (HIF-1α) overexpression in MSC (HIF-MSC) potentiates the angiogenic and immunomodulatory properties of these cells, so we hypothesized that co-transplantation of MSC-HIF with CD34+ human cord blood cells would also enhance hematopoietic stem cell engraftment and function both in vitro and in vivo.

Methods

Human MSC were obtained from dental pulp. Lentiviral overexpression of HIF-1α was performed transducing cells with pWPI-green fluorescent protein (GFP) (MSC WT) or pWPI-HIF-1α-GFP (HIF-MSC) expression vectors. Human cord blood CD34+ cells were co-cultured with MSC WT or HIF-MSC (4:1) for 72 h. Then, viability (Annexin V and 7-AAD), cell cycle, ROS expression and immunophenotyping of key molecules involved in engraftment (CXCR4, CD34, ITGA4, c-KIT) were evaluated by flow cytometry in CD34+ cells. In addition, CD34+ cells clonal expansion was analyzed by clonogenic assays. Finally, in vivo engraftment was measured by flow cytometry 4-weeks after CD34+ cell transplantation with or without intrabone MSC WT or HIF-MSC in NOD/SCID mice.

Results

We did not observe significant differences in viability, cell cycle and ROS expression between CD34+ cells co-cultured with MSC WT or HIF-MSC. Nevertheless, a significant increase in CD34, CXCR4 and ITGA4 expression (p = 0.009; p = 0.001; p = 0.013, respectively) was observed in CD34+ cells co-cultured with HIF-MSC compared to MSC WT. In addition, CD34+ cells cultured with HIF-MSC displayed a higher CFU-GM clonogenic potential than those cultured with MSC WT (p = 0.048). We also observed a significant increase in CD34+ cells engraftment ability when they were co-transplanted with HIF-MSC compared to CD34+ co-transplanted with MSC WT (p = 0.016) or alone (p = 0.015) in both the injected and contralateral femurs (p = 0.024, p = 0.008 respectively).

Conclusions

Co-transplantation of human CD34+ cells with HIF-MSC enhances cell engraftment in vivo. This is probably due to the ability of HIF-MSC to increase clonogenic capacity of hematopoietic cells and to induce the expression of adhesion molecules involved in graft survival in the hematopoietic niche.

Background

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains as the only curative therapeutic approach for a variety of hematological diseases. An adequate hematopoietic function after an allo-HSCT is not only dependent on the number of hematopoietic stem cells (HSC) infused that is clearly an important factor for engraftment, but also on the functioning of the bone marrow (BM) microenvironment, a key regulator of the hematopoietic function. The latter is often damaged in this setting [1, 2]. Among the cell types in the BM microenvironment, probably Mesenchymal Stromal Cells (MSC) have attracted most of the attention for a number of reasons, including their regenerative potential within the BM niche and their immunomodulatory properties [3].

These properties have prompted their assessment in vivo for the treatment of important complications after allo-HSCT, especially graft-versus-host disease (GVHD) or poor graft function [4,5,6]. Human MSC can be isolated from several sources although the most common are BM and adipose tissue. Dental MSC (dental pulp, apical papilla, dental follicle and periodontal ligament) are also considered an attractive source of MSC due to their easy accessibility and in vitro expansion and their low immunogenicity [7]. We and others have shown that MSC are able to maintain in vitro hematopoiesis and that MSC co-infusion may modulate host alloreactivity and promote better engraftment of donor HSC [8, 9]. Some preliminary clinical experiences of the use of MSC to improve hematopoietic engraftment after allo-SCT [10, 11] are encouraging, but new strategies boosting these effects are needed.

In this regard, it has been described that preconditioning MSC with hypoxia or the overexpression of hypoxia-inducible factor-1α (HIF-1α) potentiates the therapeutic properties of MSC [12,13,14,15]. HIF-1α is a transcription factor that is activated in response to low oxygen levels, enabling cell adaptation to hypoxia modifying cell metabolism and cell survival. HIF-1α stabilization contributes to maintaining the undifferentiated and multipotent status of MSC [16], induces the activation of Notch and Akt signaling pathways, which regulate survival, migration and proliferation [17, 18], and activates the production of angiogenic factors [19].

We have previously demonstrated that the overexpression of HIF-1α in MSC improves migration, proliferation capacity, angiogenic and immunomodulatory properties of these cells leading that in turns potentiates their regenerative capacity in experimental models of ischemia [20,21,22]. Others reported that co-culture of human fetal liver stromal cells expressing HIF-1α improved the maintenance of self-renewal and pluripotency of human embryonic stem cells [23]. Indeed, MSC depend on HIF factors for maintenance and regulation of hematopoiesis [14].

In the current work, we show the ability of HIF-MSC to promote hematopoietic stem cell function and engraftment both in vitro and in vivo.

Methods

Cell culture and lentiviral transduction

Human MSC obtained from dental pulp were purchased from Inbiomed (Inbiobank, San Sebastian, Spain). Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM)-low glucose (Gibco, Life Technologies) supplemented with 10% fetal bovine serum (FBS; Gibco, Life Technologies) and 1% penicillin/streptomycin (Gibco) in a humidified atmosphere of 95% air and 5% CO2 at 37ºC.

Overexpression of HIF-1α was performed as previously described [22]. Briefly, MSC were transduced with a control pWPI-green fluorescent protein lentivirus (MSC wild type or MSC WT) or a HIF-1α-overexpressing pWPI-HIF-1α-GFP lentivirus (HIF-MSC) (http://addgene.org cat. #12254) daily for 3 days. Transduction efficiency was evaluated by flow cytometry (Coulter EPICS XL flow cytometer; Beckman Coulter) to determine the percentage of GFP-positive cells. The percentages of infection obtained were higher than 90% in all cases (Additional file 2: Figure S1). Cells were expanded and then cryopreserved until used.

For immunophenotypic characterization, MSC were incubated with the corresponding monoclonal antibodies (see Additional file 1). Then, they were acquired on a FACS Canto II device (BD Biosciences San Jose, CA) using the FACSDiva 6.1 software (BD Biosciences) and analyzed with the Infinicyt software.

CD34+ cell isolation

CD34+ cells were isolated from 24 fresh umbilical cord blood (UCB) units obtained in the Obstetrics Department of the University Hospital of Salamanca following standard procedures, after informed consent was obtained from the corresponding mothers (range 27–40 years). Mononucleated cells were isolated by Ficoll-Paque density gradient centrifugation, and then, CD34+ cells were purified by immunomagnetic sorting in an AutoMACS (MiltenyiBiotec GmbH) after labeling with the human CD34 MicroBead Kit (MiltenyiBiotec) according to the manufacturer’s recommendations. Purity of cells was confirmed by flow cytometry using FITC-CD34 (eBioscience), and the viability was evaluated by labeling cells with 7-amino-actinomycin D (7-AAD). Purity was superior to 90% in all cases and their viability was higher than 85%. All experimental procedures were approved by local Ethics Committee (code PI26/03/2018).

Co-culture of UCB CD34+ cells with MSC

For the in vitro experiments indicated below, 2 × 105 CD34+ cells from 10 UCB samples were incubated either with 5 × 104 MSC WT or HIF-MSC. Cultures were maintained for 72 h in a volume of 2.5 ml Roswell Park Memorial Institute Medium (RPMI; Gibco, Life Technologies) per well at 37ºC and 5% of CO2 (normoxic conditions) in a humidified atmosphere.

Apoptosis assays and cell cycle analysis

After 72 h of co-culture, CD34+ cells were collected and washed. For apoptosis assays, cells were stained with Annexin V and 7-AAD using the BD Pharmigen PE Annexin V Apoptosis Detection Kit I (BD Biosciences). For cell cycle analysis, cells were stained with propidium iodide using the kit FxCycletm PI/RNase Staining Solution (Life Technologies), according to manufacturer’s instructions. Samples were acquired on a FACS Canto II device (BD Biosciences San Jose, CA) using the FACSDiva 6.1 software (BD Biosciences). At least 1.5 × 105 events were recorded. For apoptosis assays, data were analyzed using Infinicyt (Cytognos). Cells were classified as early apoptotic, late apoptosis or dead if they were Annexin V+/7-AAD, Annexin V+/7-AAD+ or Annexin V/7-AAD+, respectively, as previously reported [2]. Ten samples were used for these experiments. For cell cycle analysis, ModFit LT version 5.0.9 (Verity Software) was used. Seven samples were analyzed. Data are represented as percentage of cells in each cell cycle phase.

Intracellular reactive oxygen species (ROS)

Intracellular ROS expression was assessed in both experimental groups as described [24]. Briefly, CD34+ cells suspended in phosphate-buffered saline (PBS; Gibco) were stained with the cell-permeant probe 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA; Sigma-Aldrich, St. Louis, MO, USA) at a final concentration of 10 µmol/l, at 37 °C in the dark for 30 min. Then, cells were labeled with 7-AAD and acquired on a FACSCanto II flow cytometer (BD Biosciences San Jose, CA) using the FACSDiva 6.1 software (BD Biosciences). Experiments were performed in ten samples. Data are represented as median fluorescence intensity (MFI).

Flow cytometric analysis of proteins involved in cell engraftment and hematopoiesis

CD34+ cells from 10 samples were collected after the co-culture with MSC WT or HIF-MSC, washed with PBS and incubated with the corresponding monoclonal antibodies (see Additional file 1). Then, they were acquired on a FACS Canto II device (BD Biosciences San Jose, CA) using the FACSDiva 6.1 software (BD Biosciences) and analyzed with the Infinicyt software. Data were represented as MFI.

Clonogenic assays

Clonal growth of CD34+ cells was evaluated after 72 h of co-culture with MSC WT or HIF-MSC. For this purpose, CD34+ cells were recovered and 2,500 cells were seeded in methylcellulose Stem MACS Media (HSC-CFU complete without Epo, human) (Miltenyi Biotec, Germany) that contains stem cell factor (SCF), granulomonocytic colony-stimulating factor (GM-CSF), granulocytic colony-stimulating factor (G-CSF), interleukin-3 and interleukin-6, to quantify progenitor cell colony-forming unit granulocyte/macrophage (CFU-GM), according to the manufacturer’s instructions. After 14 days at 37ºC in a fully humidified atmosphere of 5% CO2, colonies were scored using an inverted microscope. Ten experiments were performed.

Analysis of human hematopoietic engraftment in mice

For the in vivo studies, 6-week-old nonobese diabetic/severe combined immunodeficient mice (NOD/SCID; NOD.CB17-Prkdcscid/NcrCrl) were purchased from Charles River Laboratories (Barcelona, Spain) and maintained in the Animal Care Facility of the University of Salamanca. All procedures followed the Spanish and European Union guidelines (RD 1201/05 and 86/609/CEE) and were approved by the Bioethics Committee of the University of Salamanca (reg. 201100007924). The murine xenotransplant model was established as previously reported, with minor modifications [9, 25]. Thirty-six female mice were used establishing three experimental groups: (1) 1 × 105 UCB CD34+ cells, n = 8; (2) 1 × 105 UCB CD34+ cells + 5 × 105 MSC WT, n = 13; and (3) 1 × 105 UCB CD34+ cells + 5 × 105 HIF-MSC, n = 15. Before UCB transplantation, mice were irradiated with 350 cGy of total body irradiation using a cesium source (Gammacell-200, Nordion International). Then, animals were anesthetized with a mixture of ketamine (90 mg/kg; Imalgene 500) and xylazine (10 mg/kg; Rompun 2%, KVP Pharma) before MSC administration via intrafemorally, through the knee in a well-established method, described in prior works from our group and others [9, 25,26,27]. Four hours after MSC administration, CD34+ cells were injected intravenously. Finally, four weeks after transplantation, mice were killed, and hematopoietic engraftment was evaluated in the injected (right) and contralateral (left) femurs and spleen by flow cytometry. The information on the monoclonal antibodies and analysis is detailed in Additional file 1.

Statistical analysis

Values are summarized as median and range. The nonparametric Wilcoxon signed-rank test was used to compare the differences between paired results, and the nonparametric Mann–Whitney U test was used to compare the differences between unpaired results. Differences were considered to be statistically significant for values of p < 0.05. All statistical analyses were performed on the GraphPad Prism version 5.00 for Windows (GraphPad Software, San Diego, CA).

Results

Characterization of MSC WT and HIF-MSC

Both MSC WT and HIF-MSC displayed the characteristic fibroblastic-like morphology and were adherent to plastic surfaces. Immunophenotypic analysis showed that all samples expressed CD90, CD44, CD73, CD105 and CD166, while they were negative for hematopoietic markers (Additional file 3: Figure S2).

HIF-MSC do not alter the viability and proliferative capacity of CD34+ cells

We set an in vitro co-culture experiment to evaluate the effect of HIF-MSC in CD34+ cells. Cell viability assays of CD34+ cells were performed after 72 h of co-culture with MSC (n = 10). 78.74% [60.64%—81.66%] of CD34+ cells were viable when co-cultured with MSC WT, with no significant differences with the co-culture with HIF-MSC (75.78% [62.61–77.75%]) (Annexin V/7-AAD). There were also no significant differences in the percentage of early apoptotic cells (Annexin V+/7-AAD), late apoptotic cells (Annexin V+/7-AAD+) and dead cells (Annexin V/7-AAD+) between both groups (Fig. 1A).

Fig. 1
figure 1

Viability, proliferation and ROS expression in CD34+ cells after co-culture with MSC WT or HIF-MSC. Apoptosis assays in CD34+ cells previously co-cultured with MSC WT or HIF-MSC during 72 h. CD34+ cells were incubated with Annexin V, 7-AAD and CD34, and the expression of different cell surface markers was analyzed by flow cytometry. Cells were classified as viable (Annexin V/7-AAD), early apoptotic (Annexin V+/7-AAD), late apoptotic (Annexin V+/7-AAD+) or dead (Annexin V/7-AAD+). Data expressed as mean of the percentage of cells in the different conditions (A). Cell cycle profiling of CD34+ cells after 72 h of culture in each condition analyzed by flow cytometry. Data are represented as mean of the percentage of cells in each phase (B). Intracellular reactive oxygen species (ROS) expression of each population represented as median of fluorescence intensity (C). Ten experiments were done for each group. p:n.s means p > 0.05

Regarding cell cycle, both CD34+ cells co-cultured with MSC WT or HIF-MSC show similar profiles. We did not find significant differences in the percentage of cells in Phase G0/G1 (93.42% [92.11–94.63%] vs 93.97% [92.41–95.72%]), Phase G2/M (0.43% [0.17–0.51%] vs 0.37% [0.22–0.58%]) and S Phase (6.15% [5.30–7.27%] vs 5.44% [40.6–7.22%]) between MSC WT and HIF-MSC, respectively (n = 7) (Fig. 1B).

Intracellular ROS levels of CD34+ cells are not modified in co-culture with HIF-MSC

We also explored the possibility that HIF-MSC when co-cultured with CD34+ cells lowered their ROS, making them less vulnerable. When assessing the levels of intracellular ROS in CD34+ cells of both experimental conditions after 72 h of co-culture, we did not observe significant differences between them (n = 10) (Fig. 1C).

Culture of CD34+ cells with HIF-MSC increases their expression of surface molecules involved in engraftment

We next decided to test whether HIF-MSC when co-cultured with CD34+ cells could enhance their expression of proteins involved in hematopoietic engraftment and hematopoiesis maintenance. Surface expression in CD34+ cells of some key molecules involved in hematopoietic engraftment and hematopoiesis maintenance was also evaluated (n = 10). We found a significant increase in the expression of Hematopoietic Progenitor Cell Antigen (CD34), C-X-C Motif Chemokine Receptor 4 (CXCR4) and Integrin Subunit Alpha (ITGA4) in CD34+ cells that have been previously co-cultured with HIF-MSC compared to those co-cultured with MSC WT (p = 0.009, p = 0.001 and p = 0.013, respectively). We did not find significant changes in c-KIT expression (Fig. 2A).

Fig. 2
figure 2

Expression of surface proteins involved in hematopoiesis and engraftment in CD34+ cells and clonogenic capacity. Median fluorescence intensity of different surface proteins involved in hematopoiesis maintenance and engraftment as CD34, CXCR4, ITGA-4 and c-KIT was evaluated by flow cytometry. Samples were acquired on a FACS Canto flow cytometer (A). Total CFU-GM from CD34+ cells were scored after 14 days in methylcellulose medium. CD34+ cells were cultured with MSC WT or HIF -MSC for 72 h, and then, 1,500 cells were seeded into methylcellulose medium (B). Data are represented as mean of 10 experiments for each group. p:n.s means p > 0.05

Culture of CD34+ cells with HIF-MSC increases their clonogenic capacity in vitro

Regarding the evaluation of in vitro functional effects induced after co-culture, we observed that the capacity of CD34+ cells to form CFU-GM colonies was significantly higher when they were co-cultured with HIF-MSC in comparison with MSC WT (p = 0.048). There were no differences in the shape or size of the colonies between both groups (Fig. 2B). Clonogenic studies were performed in ten samples. Our data together suggested that MSC-HIF enhance molecules engraftment expression and clonogenic capacity of CD34+ cells fostering the usage of both cell types for in vivo therapeutic purposes.

Co-transplantation of CD34+ cells with HIF-MSC improves their engraftment ability in vivo

Finally, the in vivo engraftment ability of CD34+ cells was evaluated in a xenotransplantation murine model using NOD/SCID mice assessing the percentage of human CD45+ cells after 4 weeks in both femurs. We observed a significant increase in CD34+ cell engraftment ability when mice were co-transplanted with HIF-MSC compared to mice that received CD34+ or CD34+ co-transplanted with MSC WT in both the injected (right) (p = 0.016 and p = 0.015, respectively) and contralateral (left) femurs (p = 0.024 and p = 0.008, respectively) (Fig. 3A). Nevertheless, we did not observe significant differences among groups in the spleen (Additional file 4: Figure S3).

Fig. 3
figure 3

Analysis of human hematopoietic engraftment in vivo. Human hematopoietic engraftment was evaluated by flow cytometry after xenotrasplantation in NOD/SCID mice. The percentage of human CD45+ cells (donor chimerism) was analyzed in total bone marrow samples from both femurs, right (MSC injected site) and left (contralateral) 4 weeks after transplantation. Mice were transplanted intravenously with CD34+ cells alone or co-transplanted with MSC WT or HIF-MSC, that were administered in the right femur (A). Percentage of human hematopoietic subpopulations (CD34, CD14, CD13 and CD19) in the injected femur after 4 weeks was also analyzed (B). Data are represented as mean of 8 experiments for mice transplanted with CD34+ cells alone, 13 experiments for mice co-transplanted with CD34+ cells and WT MSC and 15 experiments for mice transplanted with CD34+ cells and HIF-MSC. p:n.s means p > 0.05

Cell engraftment was also evaluated at the subpopulation level assessing CD34+, CD14+, CD13+ and CD19+ cell subsets. We observed a significant increase in engraftment of CD34+, CD14+ and CD13+ cell subsets when mice were co-transplanted with HIF-MSC compared with the remaining groups (Fig. 3B). Although the engraftment of the CD19+ cell subset was similar to other cell populations, there was no significant differences among experimental groups in our study.

Discussion

The main originality of the current manuscript is that we have observed, for the first time, that co-transplantation of CD34+ cells with HIF-MSC increases their engraftment ability in vivo. In addition, we have described that this may be related to an increased expression of some key molecules involved in engraftment, including CXCR4 and ITGA4 and an improved clonogenic ability induced by MSC in CD34+ cells.

HIF-1α overexpression in MSC is not the only modification that has been implemented to enhance MSC properties in different experimental scenarios. For instance, it has been shown that modification of MSC by overexpression of key molecules involved in homing and immunosuppression (as CXCR4 and IL10) or by exofucosylation can improve MSC properties [28,29,30]. Preconditioning of these cells with pro-inflammatory cytokines, irradiation, pharmacological or chemical agents may have similar effects [31,32,33]. Our group was one of the firsts to describe that culturing MSC in hypoxia (with 5% O2) improved MSC proliferation and expansion [12]. Several studies have also reported that hypoxia preconditioning enhances the therapeutic potential of MSC [34,35,36,37,38]. However, maintaining in vitro the hypoxic conditions that MSC have physiologically in the human bone marrow niche is difficult to preserve only by regulating culture conditions, since the exposure to a normoxic environment rapidly reverses the gained features induced by hypoxia. There is extensive information about the key role of HIF-1α regulating the hypoxic metabolism [35, 37, 39, 40]. That is why in the current work, we have opted by overexpressing HIF-1α in MSC by lentiviral transduction in order to mimic some of the effects induced by hypoxia and maintain those effects over time.

Delving further into the mechanisms involved in the changes induced in MSC by low oxygen concentrations, it has been shown that the most remarkable is the increase in HIF-1α expression, promoting their regenerative and angiogenic capacity, activating Notch downstream genes, increasing cell proliferation and survival and the retention of stem cell properties as self-renewal and senescence inhibition [35, 37,38,39, 41]. In previous works, we have observed that most of these changes are also induced when transducing MSC with HIF-1α lentivirus vectors [17, 20,21,22].

Regarding the effects of HIF-1α expression in MSC on the hematopoietic cell compartment that is the focus of our work, Guarnerio et al. demonstrated that downregulation of HIF factors increases interferon expression, which induces expansion and differentiation of hematopoietic progenitors mediated by STAT-1. In fact, during systemic infections, where HSC proliferation and differentiation is required, HIF-1α expression is suppressed in MSC. However, when HIF-1α is overexpressed in MSC they observed a decrease of hematopoietic differentiation while no changes were detected in their proliferation capacity after 4 days of culture [14]. In agreement with these data, we have not seen significant differences regarding viability or cell cycle between CD34+ cells co-cultured with HIF-MSC or MSC WT after 3 days. Another report confirms the decrease in hematopoietic differentiation with HIF-MSC but, in contrast, notices an increase in CD34+ expansion after 10 days of co-culture [13]. This discordance regarding HSC proliferation may be related to the different experimental conditions used. It seems that HIF factors can regulate hematopoiesis altering MSC secretome, suppressing the release of some molecules that induce hematopoietic differentiation and increasing the release of other factors that induce HSC maintenance, as CRIPTO, a HIF regulated soluble factor [42] (Fig. 4).

Fig. 4
figure 4

Schematic representation of the potential mechanisms by which HIF-1α overexpression in MSC could enhance CD34+ cell engraftment. HIF-1α action may depend more on secreted soluble factors than on cell-to-cell contact. Interaction through ligands and receptors, cytokine release and extracellular vesicle release contribute to this process. Molecules that are increased in HIF-MSC are represented in green and molecules that are decreased are represented in red. HSC, hematopoietic stem cell; CXCR4, C-X-C chemokine receptor type 4; ITGA4, integrin subunit alpha 4; SDF-1, stromal-derived factor-1; SCF, stem cell factor; EPO, erythropoietin; PDGF, platelet-derived growth factor; ANGPT, angiopoietin; VEGF, vascular endothelial growth factor; ROS, reactive oxygen species; EV, extracellular vesicles; MSC WT: MSC wild type; HIF-MSC: MSC overexpressing HIF-1α

ROS signaling is obviously related to hypoxia [43]. In MSC, it has been observed a reduction in ROS when MSC are cultured at low oxygen concentration. Hypoxia induces a stabilization of HIF-1α that leads to anaerobic respiration where less oxygen is consumed and the production of ROS is lowered [35, 37]. We analyzed whether this antioxidant capacity of HIF-1α overexpressed in MSC could influence in any way the ROS status in CD34+ cells after co-culture, but we have not observed differences between CD34+ cells co-cultured with HIF-MSC or MSC WT.

Several interleukins and paracrine factors are upregulated in hypoxic MSC, together with some genes involved in hematopoiesis and engraftment including vascular endothelial growth factor (VEGF), erythropoietin, angiopoietin, platelet-derived growth factor, CXCR4 or SDF-1 [19, 37, 44, 45]. In this work, we have shown that the expression of CXCR4 in CD34+ cells co-cultured with HIF-MSC is significantly increased. This is in accordance with findings that SDF-1 expression, one of the most important ligands of CXCR4, is regulated by HIF-1α in stromal cells increasing homing of CXCR4 positive progenitor cells to reduced oxygen tension areas within the BM niche [46]. SDF-1 expression is directly proportional to tissue oxygen reduction. In fact, MSC overexpressing HIF-1α in our system have also increased expression of SDF-1 [21]. In addition, hypoxia induces an upregulation of CXCR4 in MSC that seems to be driven by HIF-1α [47]. We have not observed changes in c-kit expression in CD34+ cells co-cultured with HIF-MSC, although it has been described that the production of its ligand, SCF, is significantly increased when MSC overexpress HIF-1α [13]. SCF also causes upregulation of ITGA4 in CD34+ cells [48]. These data are consistent with our findings. ITGA4 expression in HSC co-cultured with HIF-MSC is significantly increased. ITGA4 is an important molecule involved in homing of HSC and is described to be upregulated in hypoxic environments as ischemic tissues or melanoma cells [49, 50]. Expression of other integrins and genes involved in cell adhesion is also upregulated in MSC overexpressing HIF-1α [22]. Additionally, we have detected an increase in CD34 expression in HSC cultured with HIF-MSC, and this could be related to a reduced HSC differentiation, as others have described [13, 14].

Moving to the functional implications of the changes induced in CD34+ cells after co-culture with HIF-MSC, we have seen an enhanced clonogenic capacity compared to those co-cultured with MSC WT. In accordance with these results, in another work where HIF-1α was overexpressed in MSC, they observed similar results with an increased number of CD34+ colonies after 5 days of co-culture. In this work, they also described that HIF-1α overexpression in MSC can augment the production of some hematopoietic growth factors increasing their hematopoietic supportive functions [13]. This effect could also be mediated by extracellular vesicles (EV). We have previously reported that the incorporation of EV from MSC into CD34+ cells increases their clonogenic capacity [25, 51] and other reports showed similar results [52,53,54]. In fact, HIF-1α action may depend more on secreted soluble factors than on cell-to-cell contact [23]. We have also described that in HIF-MSC, the release of EV is increased and their content is different from EV released by MSC WT [20].

The latest and most relevant result of our study is the significant better engraftment of human CD34+ cells co-transplanted with HIF-MSC in comparison with alone or co-transplanted with MSC WT in an established xenotransplantation model. These differences were observed both in the injected and in the contralateral femurs. Although there is no previous published data in this setting, there is an interesting study where extramedullary bone and bone marrow were developed in immmunodeficient mice after injecting subcutaneously human MSC and endothelial cells [55]. They demonstrate human HSC engraftment in this ectopic bone marrow niche, and when HIF-1α was knocked down in MSC, it was significantly reduced. These findings position HIF-1α expression in MSC as a key regulator of their role in favoring human HSC engraftment. We have also evaluated the engraftment of different cell subsets, observing a significant increase of human CD14+, CD13+ and CD34+ cell populations in mice co-transplanted with HIF-MSC. However, no significant improvement of engraftment was observed in the CD19+ cells. It has been described that HIF activity needs to be low at the immature B cell stages for a normal B cell development. HIF-1α activation could decrease BCR editing and cause arrest of immature B cells. Administration of HIF activators in clinical use reduce B cells [56], but the effects of co-transplantation of CD34+ cells with HIF-MSC on B cell development remains to be elucidated. Regarding potential mechanisms for these enhanced effects of modified MSC, in previous works we have described that overexpression of HIF-1α increases MSC migration ability and their cell adhesion capacity after intravenous administration, one of the limiting factors in the therapeutic potential of MSC [22]. We have also observed an improvement in their immunomodulatory capacity [57, 58]. Moreover, their osteogenic differentiation ability has been shown to be increased in MSC overexpressing HIF-1α [59]. All these data support the improvement of HSC engraftment after MSC modification. It would have been of interest the evaluation of CXCR4 and ITGA4 expression in the engrafted cells at 4 four weeks postinjection to examine if mice with higher human chimerism display a higher expression of these molecules. There is a study including patients undergoing HSCT where the authors found significant differences in CXCR4 expression between patients undergoing successful engraftment and those with poor or no engraftment at day 90 [60]. This evaluation would allow to assess, using the CXCR4 inhibitor AMD3100, if the effect of HIF-1α can be diminished through the inhibition of the SDF-1-CXCR4 axis.

Conclusions

In summary, we have described that overexpression of HIF-1α in MSC increases their favorable effects on supporting human CD34+ cell engraftment in vivo. This is also associated with an increase in the clonogenic capacity of hematopoietic cells and changes in adhesion molecules involved in these processes that are induced after the co-culture with modified MSC.

Availability of data and materials

The datasets supporting the conclusions of this article are available from the corresponding author upon reasonable request.

Abbreviations

allo-HSCT:

Allogeneic hematopoietic stem cell transplantation

HSC:

Hematopoietic stem cells

BM:

Bone marrow

MSC:

Mesenchymal stromal cells

GVHD:

Graft-versus-host disease

HIF-1α:

Hypoxia-inducible factor-1α

DMEM:

Dulbecco’s modified Eagle’s medium

FBS:

Fetal bovine serum

UCB:

Umbilical cord blood

7-AAD:

7-Amino-actinomycin D

MSC WT:

MSC wild type

HIF-MSC:

MSC overexpressing HIF-1α

RPMI:

Roswell Park Memorial Institute Medium

ROS:

Reactive oxygen species

DCFH-DA:

Dichlorodihydrofluorescein diacetate

MFI:

Median fluorescence intensity

PBS:

Phosphate-buffered saline

SCF:

Stem cell factor

GM-CSF:

Granulomonocytic colony-stimulating factor

G-CSF:

Granulocytic colony-stimulating factor

CFU-GM:

Colony-forming unit granulocyte/macrophage

NOD/SCID:

Nonobese diabetic/severe combined immunodeficient mice

CXCR4:

C-X-C motif chemokine receptor 4

ITGA4:

Integrin subunit alpha

VEGF:

Vascular endothelial growth factor

EV:

Extracellular vesicles

References

  1. Sanchez-Guijo FM, et al. Posttransplant hematopoiesis in patients undergoing sibling allogeneic stem cell transplantation reflects that of their respective donors although with a lower functional capability. Exp Hematol. 2005;33(8):935–43. https://doi.org/10.1016/j.exphem.2005.04.009.

    Article  CAS  PubMed  Google Scholar 

  2. Preciado S, et al. Mesenchymal stromal cell irradiation interferes with the adipogenic/osteogenic differentiation balance and improves their hematopoietic-supporting ability. Biol Blood Marrow Transplant. 2018;24(3):443–51. https://doi.org/10.1016/j.bbmt.2017.11.007.

    Article  PubMed  Google Scholar 

  3. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med. 2001;226(6):507–20. https://doi.org/10.1177/153537020122600603.

    Article  CAS  Google Scholar 

  4. Shi M, Liu ZW, Wang FS. Immunomodulatory properties and therapeutic application of mesenchymal stem cells. Clin Exp Immunol. 2011;164(1):1–8. https://doi.org/10.1111/j.1365-2249.2011.04327.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wong RSY. Mesenchymal stem cells: angels or demons? J Biomed Biotechnol. 2011. https://doi.org/10.1155/2011/459510.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Méndez-Ferrer S, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34. https://doi.org/10.1038/nature09262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ledesma-Martínez E, Mendoza-Núñez VM, Santiago-Osorio E. Mesenchymal stem cells derived from dental pulp: a review. Stem Cells Int. 2016. https://doi.org/10.1155/2016/4709572.

    Article  PubMed  Google Scholar 

  8. Carrancio S, et al. Bone marrow mesenchymal stem cells for improving hematopoietic function: an in vitro and in vivo model. Part 2: effect on bone marrow microenvironment. PLoS ONE. 2011. https://doi.org/10.1371/journal.pone.0026241.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Carrancio S, et al. Effects of MSC coadministration and route of delivery on cord blood hematopoietic stem cell engraftment. Cell Transplant. 2013;22(7):1171–83. https://doi.org/10.3727/096368912X657431.

    Article  CAS  PubMed  Google Scholar 

  10. Sánchez-Guijo FM, et al. Allogeneic mesenchymal stem cell therapy for refractory cytopenias after hematopoietic stem cell transplantation. Transfusion. 2012;52(5):1086–91. https://doi.org/10.1111/j.1537-2995.2011.03400.x.

    Article  PubMed  Google Scholar 

  11. Ball LM, et al. Cotransplantation of ex vivo-expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood. 2007;110(7):2764–7. https://doi.org/10.1182/blood-2007-04-087056.

    Article  CAS  PubMed  Google Scholar 

  12. Carrancio S, et al. Optimization of mesenchymal stem cell expansion procedures by cell separation and culture conditions modification. Exp Hematol. 2008;36(8):1014–21. https://doi.org/10.1016/j.exphem.2008.03.012.

    Article  CAS  PubMed  Google Scholar 

  13. Kiani AA, et al. Over expression of hif-1α in human mesenchymal stem cells increases their supportive functions for hematopoietic stem cells in an experimental co-culture model. Hematology. 2014;19(2):85–98. https://doi.org/10.1179/1607845413Y.0000000093.

    Article  CAS  PubMed  Google Scholar 

  14. Guarnerio J, Coltella N, Ala U, Tonon G, Pandolfi PP, Bernardi R. Bone marrow endosteal mesenchymal progenitors depend on HIF factors for maintenance and regulation of hematopoiesis. Stem Cell Rep. 2014;2(6):794–809. https://doi.org/10.1016/j.stemcr.2014.04.002.

    Article  CAS  Google Scholar 

  15. Yang C, Liu H, Liu D. Mutant hypoxia-inducible factor 1α modified bone marrow mesenchymal stem cells ameliorate cerebral ischemia. Int J Mol Med. 2014;34(6):1622–8. https://doi.org/10.3892/ijmm.2014.1953.

    Article  CAS  PubMed  Google Scholar 

  16. Basciano L, et al. Long term culture of mesenchymal stem cells in hypoxia promotes a genetic program maintaining their undifferentiated and multipotent status. BMC Cell Biol. 2011. https://doi.org/10.1186/1471-2121-12-12.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Ciria M, et al. Mesenchymal stem cell migration and proliferation are mediated by hypoxia-inducible factor-1α upstream of notch and SUMO pathways. Stem Cells Dev. 2017;26(13):973–85. https://doi.org/10.1089/scd.2016.0331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rosová I, Dao M, Capoccia B, Link D, Nolta JA. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells. 2008;26(8):2173–82. https://doi.org/10.1634/stemcells.2007-1104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hirota K, Semenza GL. Regulation of angiogenesis by hypoxia-inducible factor 1. Crit Rev Oncol Hematol. 2006;59(1):15–26. https://doi.org/10.1016/j.critrevonc.2005.12.003.

    Article  PubMed  Google Scholar 

  20. Gonzalez-King H, et al. Hypoxia inducible factor-1a potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells. 2017. https://doi.org/10.1002/stem.2618.

    Article  PubMed  Google Scholar 

  21. Martinez VG, et al. Overexpression of hypoxia-inducible factor 1 alpha improves immunomodulation by dental mesenchymal stem cells. Stem Cell Res Ther. 2017;8(1):1–12. https://doi.org/10.1186/s13287-017-0659-2.

    Article  CAS  Google Scholar 

  22. Cerrada I, et al. Hypoxia-inducible factor 1 alpha contributes to cardiac healing in mesenchymal stem cells-mediated cardiac repair. Stem Cells Dev. 2012;22(3):501–11. https://doi.org/10.1089/scd.2012.0340.

    Article  CAS  PubMed  Google Scholar 

  23. Ji L, et al. Self-renewal and pluripotency is maintained in human embryonic stem cells by co-culture with human fetal liver stromal cells expressing hypoxia inducible factor 1α. J Cell Physiol. 2009;221(1):54–66. https://doi.org/10.1002/jcp.21826.

    Article  CAS  PubMed  Google Scholar 

  24. Sardina JL, et al. P22phox-dependent NADPH oxidase activity is required for megakaryocytic differentiation. Cell Death Differ. 2010;17(12):1842–54. https://doi.org/10.1038/cdd.2010.67.

    Article  CAS  PubMed  Google Scholar 

  25. Preciado S, et al. The incorporation of extracellular vesicles from mesenchymal stromal cells into CD34+ cells increases their clonogenic capacity and bone marrow lodging ability. Stem Cells. 2019. https://doi.org/10.1002/stem.3032.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Yahata T, et al. A highly sensitive strategy for SCID-repopulating cell assay by direct injection of primitive human hematopoietic cells into NOD/SCID mice bone marrow. Blood. 2003;101(8):2905–13. https://doi.org/10.1182/blood-2002-07-1995.

    Article  CAS  PubMed  Google Scholar 

  27. Castello S, et al. Intra-bone marrow injection of bone marrow and cord blood cells: an alternative way of transplantation associated with a higher seeding efficiency. Exp Hematol. 2004;32(8):782–7. https://doi.org/10.1016/j.exphem.2004.05.026.

    Article  PubMed  Google Scholar 

  28. Hervás-Salcedo R, et al. Enhanced anti-inflammatory effects of mesenchymal stromal cells mediated by the transient ectopic expression of CXCR4 and IL10. Stem Cell Res Ther. 2021;12(1):1–20. https://doi.org/10.1186/s13287-021-02193-0.

    Article  CAS  Google Scholar 

  29. García-Bernal D, et al. Exofucosylation of adipose mesenchymal stromal cells alters their secretome profile. Front Cell Dev Biol. 2020;8(November):1–17. https://doi.org/10.3389/fcell.2020.584074.

    Article  Google Scholar 

  30. Chou KJ, et al. CD44 fucosylation on mesenchymal stem cell enhances homing and macrophage polarization in ischemic kidney injury. Exp Cell Res. 2017;350(1):91–102. https://doi.org/10.1016/j.yexcr.2016.11.010.

    Article  CAS  PubMed  Google Scholar 

  31. Hu C, Li L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J Cell Mol Med. 2018;22(3):1428–42. https://doi.org/10.1111/jcmm.13492.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Preciado S, et al. Mesenchymal stromal cell irradiation interferes with the adipogenic/osteogenic differentiation balance and improves their hematopoietic-supporting ability. Biol Blood Marrow Transplant. 2017. https://doi.org/10.1016/j.bbmt.2017.11.007.

    Article  PubMed  Google Scholar 

  33. Noronha NDC, et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 2019;10(1):1–21. https://doi.org/10.1186/s13287-019-1224-y.

    Article  CAS  Google Scholar 

  34. Liu H, et al. Hypoxic preconditioning advances CXCR4 and CXCR7 expression by activating HIF-1α in MSCs. Biochem Biophys Res Commun. 2010;401(4):509–15. https://doi.org/10.1016/j.bbrc.2010.09.076.

    Article  CAS  PubMed  Google Scholar 

  35. Nikolits I, Nebel S, Egger D, Kreß S, Kasper C. Towards physiologic culture approaches to improve standard cultivation of mesenchymal stem cells. Cells. 2021. https://doi.org/10.3390/cells10040886.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lin S, Zhu B, Huang G, Zeng Q, Wang C. Microvesicles derived from human bone marrow mesenchymal stem cells promote U2OS cell growth under hypoxia: the role of PI3K/AKT and HIF-1α. Hum Cell. 2019;32(1):64–74. https://doi.org/10.1007/s13577-018-0224-z.

    Article  CAS  PubMed  Google Scholar 

  37. Haque N, Rahman MT, Abu Kasim NH, Alabsi AM. Hypoxic culture conditions as a solution for mesenchymal stem cell based regenerative therapy. Sci World J. 2013. https://doi.org/10.1155/2013/632972.

    Article  Google Scholar 

  38. Kwon SY, et al. Hypoxia enhances cell properties of human mesenchymal stem cells. Tissue Eng Regen Med. 2017;14(5):595–604. https://doi.org/10.1007/s13770-017-0068-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chacko SM, Ahmed S, Selvendiran K, Kuppusamy ML, Khan M, Kuppusamy P. Hypoxic preconditioning induces the expression of prosurvival and proangiogenic markers in mesenchymal stem cells. Am J Physiol Cell Physiol. 2010;299(6):1562–70. https://doi.org/10.1152/ajpcell.00221.2010.

    Article  CAS  Google Scholar 

  40. Speth JM, Hoggatt J, Singh P, Pelus LM. Pharmacologic increase in HIF1α enhances hematopoietic stem and progenitor homing and engraftment. Blood. 2014;123(2):203–7. https://doi.org/10.1182/blood-2013-07-516336.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tsai C-C, Yew T-L, Yang D-C, Huang W-H, Hung S-C. Benefits of hypoxic culture on bone marrow multipotent stromal cells. Am J Blood Res. 2012;2(3):148–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Miharada K, et al. Cripto regulates hematopoietic stem cells as a hypoxic-niche-related factor through cell surface receptor GRP78. Cell Stem Cell. 2011;9(4):330–44. https://doi.org/10.1016/j.stem.2011.07.016.

    Article  CAS  PubMed  Google Scholar 

  43. Prabhakar NR, Kumar GK, Nanduri J, Semenza GL. ROS signaling in systemic and cellular responses to chronic intermittent hypoxia. Antioxidants Redox Signal. 2007;9(9):1397–403. https://doi.org/10.1089/ars.2007.1732.

    Article  CAS  Google Scholar 

  44. Hu C, Wu Z, Li L. Pre-treatments enhance the therapeutic effects of mesenchymal stem cells in liver diseases. J Cell Mol Med. 2020;24(1):40–9. https://doi.org/10.1111/jcmm.14788.

    Article  PubMed  Google Scholar 

  45. Pichiule P, Chavez JC, LaManna JC. Hypoxic regulation of angiopoietin-2 expression in endothelial cells. J Biol Chem. 2004;279(13):12171–80. https://doi.org/10.1074/jbc.M305146200.

    Article  CAS  PubMed  Google Scholar 

  46. Ceradini DJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10(8):858–64. https://doi.org/10.1038/nm1075.

    Article  CAS  PubMed  Google Scholar 

  47. Liu H, et al. The role of SDF-1-CXCR4/CXCR7 axis in the therapeutic effects of hypoxia-preconditioned mesenchymal stem cells for renal ischemia/reperfusion injury. PLoS ONE. 2012. https://doi.org/10.1371/journal.pone.0034608.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Imai Y, Shimaoka M, Kurokawa M. Essential roles of VLA-4 in the hematopoietic system. Int J Hematol. 2010;91(4):569–75. https://doi.org/10.1007/s12185-010-0555-3.

    Article  PubMed  Google Scholar 

  49. Olbryt M, Jarza̧b M, Jazowiecka-Rakus J, Simek K, Szala S, Sochanik A. Gene expression profile of B16(F10) murine melanoma cells exposed to hypoxic conditions in vitro. Gene Expr. 2006;13(3):191–203. https://doi.org/10.3727/000000006783991818.

    Article  CAS  PubMed  Google Scholar 

  50. Baczynska D, Michalowska D, Barc P, Skora J, Karczewski M, Sadakierska-Chudy A. The expression profile of angiogenic genes in critical limb ischemia popliteal arteries. J Physiol Pharmacol. 2016;67(3):353–62.

    CAS  PubMed  Google Scholar 

  51. Preciado S, Muntión S, Sánchez-Guijo F. Improving hematopoietic engraftment: potential role of mesenchymal stromal cell-derived extracellular vesicles. Stem Cells. 2021;39(1):26–32. https://doi.org/10.1002/stem.3278.

    Article  PubMed  Google Scholar 

  52. Xie H, et al. Mesenchymal stem cell-derived microvesicles support ex vivo expansion of cord blood-derived CD34+ cells. Stem Cells Int. 2016. https://doi.org/10.1155/2016/6493241.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Schoefinius JS, Brunswig-Spickenheier B, Speiseder T, Krebs S, Just U, Lange C. Mesenchymal stromal cell-derived extracellular vesicles provide long-term survival after total body irradiation without additional hematopoietic stem cell support. Stem Cells. 2017;35(12):2379–89. https://doi.org/10.1002/stem.2716.

    Article  CAS  PubMed  Google Scholar 

  54. Wen S, et al. Mesenchymal stromal cell-derived extracellular vesicles rescue radiation damage to murine marrow hematopoietic cells. Leukemia. 2016;30(11):2221–31. https://doi.org/10.1038/leu.2016.107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen Y, et al. Human extramedullary bone marrow in mice: A novel in vivo model of genetically controlled hematopoietic microenvironment. Blood. 2012;119(21):4971–80. https://doi.org/10.1182/blood-2011-11-389957.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Burrows N, et al. Dynamic regulation of hypoxia-inducible factor-1α activity is essential for normal B cell development. Nat Immunol. 2020;21(11):1408–20. https://doi.org/10.1038/s41590-020-0772-8.

    Article  CAS  PubMed  Google Scholar 

  57. Gómez-Ferrer M, et al. Hif-1α and pro-inflammatory signaling improves the immunomodulatory activity of MSC-derived extracellular vesicles. Int J Mol Sci. 2021. https://doi.org/10.3390/ijms22073416.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Contreras-Lopez R, et al. HIF1α-dependent metabolic reprogramming governs mesenchymal stem/stromal cell immunoregulatory functions. FASEB J. 2020;34(6):8250–64. https://doi.org/10.1096/fj.201902232R.

    Article  CAS  PubMed  Google Scholar 

  59. Zou D, et al. In vitro study of enhanced osteogenesis induced by HIF-1α-transduced bone marrow stem cells. Cell Prolif. 2011;44(3):234–43. https://doi.org/10.1111/j.1365-2184.2011.00747.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Asfour I, et al. CXCR4 (CD184) expression on stem cell harvest and CD34+ cells post-transplant. Hematol Oncol Stem Cell Ther. 2017;10(2):63–9. https://doi.org/10.1016/j.hemonc.2017.01.002.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Obstetrics Department of the University Hospital of Salamanca for provision of UCB samples.

Funding

This work was partially supported by Instituto de Salud Carlos III (ISCIII) (PI12/01775, PI16/01407, PI19/00245), Consejería de Educación (HUS308U14 and CAS079P17) and Consejería de Sanidad de Castilla y León (GRS1621/A/17, GRS1879/A/18, GRS1348/A/19), Junta de Castilla y León. SP is supported by Fundación Española de Hematología y Hemoterapia (FEHH). SM is supported by RETIC program of ISCIII-European Regional Development Fund (RD16/0011/0015, RD16/0011/0004), “A way to make Europe.” GJMC is supported by Ministerio de Ciencia e Innovación (FPU18/03533).

Author information

Authors and Affiliations

Authors

Contributions

SP did most of the experiments and statistical analysis, experiments with mice and manuscript writing; SS performed lentiviral transduction of MSC; SM helped with the experiments design, data interpretation and final approval of the manuscript; LO collaborated with flow cytometry experiments; GJM collaborated in culture and expansion of cells; AN helped with data interpretation and cytometry analysis; PS collaborated in design of the study, manuscript writing and final approval of the manuscript; and FSG designed the study, and collaborated in data interpretation, manuscript writing and final approval of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Fermín Sánchez-Guijo.

Ethics declarations

Ethics approval and consent to participate

CD34+ cells were isolated from 24 fresh UCB units obtained in the Obstetrics Department of the University Hospital of Salamanca following standard procedures, after informed consent was obtained from the corresponding mothers. For the in vivo studies, 6-week-old nonobese diabetic/severe combined immunodeficient mice (NOD/SCID; NOD.CB17-Prkdcscid/NcrCrl) were purchased from Charles River Laboratories (Barcelona, Spain) and maintained in the Animal Care Facility of the University of Salamanca. All procedures followed the Spanish and European Union guidelines (RD 1201/05 and 86/609/CEE) and were approved by the Bioethics Committee of the University of Salamanca (reg. 201600007333).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests regarding the current manuscript.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. Additional methods.

Additional file 2. Figure S1

. Transduction efficiency of MSC. Histogram of a representative sample, with transfection efficiency >90%. MSC WT: MSC wild type; HIF-MSC: MSC overexpressing HIF-1α; GFP: Green Fluorescent Protein

Additional file 3. Figure S2

. Immunophenotypic analysis of MSC WT and HIF-MSC. Histogram of the different surface molecules analyzed by flow cytometry. MSC WT: MSC wild type; HIF-MSC: MSC overexpressing HIF-1α

Additional file 4. Figure S3

. Analysis of human hematopoietic engraftment in spleen. Human hematopoietic engraftment was evaluated by flow cytometry after xenotrasplantation in NOD/SCID mice. The percentage of human CD45+ cells (donor chimerism) was analyzed in total spleen samples 4 weeks after transplantation. Mice were transplanted intravenously with CD34+ cells alone or co-transplanted with MSC WT or HIF-MSC, that were administered in the right femur. Data are represented as mean of 8 experiments for mice transplanted with CD34+ cells alone, 13 experiments for mice co-transplanted with CD34+ cells and WT MSC and 15 experiments for mice transplanted with CD34+ cells and HIF-MSC. p:n.s means p > 0.05

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Preciado, S., Sirerol-Piquer, M.S., Muntión, S. et al. Co-administration of human MSC overexpressing HIF-1α increases human CD34+ cell engraftment in vivo. Stem Cell Res Ther 12, 601 (2021). https://doi.org/10.1186/s13287-021-02669-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-021-02669-z

Keywords