scholarly journals PDGFRα and CD51 mark human Nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion

2013 ◽  
Vol 210 (7) ◽  
pp. 1351-1367 ◽  
Author(s):  
Sandra Pinho ◽  
Julie Lacombe ◽  
Maher Hanoun ◽  
Toshihide Mizoguchi ◽  
Ingmar Bruns ◽  
...  

The intermediate filament protein Nestin labels populations of stem/progenitor cells, including self-renewing mesenchymal stem cells (MSCs), a major constituent of the hematopoietic stem cell (HSC) niche. However, the intracellular location of Nestin prevents its use for prospective live cell isolation. Hence it is important to find surface markers specific for Nestin+ cells. In this study, we show that the expression of PDGFRα and CD51 among CD45− Ter119− CD31− mouse bone marrow (BM) stromal cells characterizes a large fraction of Nestin+ cells, containing most fibroblastic CFUs, mesenspheres, and self-renewal capacity after transplantation. The PDGFRα+ CD51+ subset of Nestin+ cells is also enriched in major HSC maintenance genes, supporting the notion that niche activity co-segregates with MSC activity. Furthermore, we show that PDGFRα+ CD51+ cells in the human fetal BM represent a small subset of CD146+ cells expressing Nestin and enriched for MSC and HSC niche activities. Importantly, cultured human PDGFRα+ CD51+ nonadherent mesenspheres can significantly expand multipotent hematopoietic progenitors able to engraft immunodeficient mice. These results thus indicate that the HSC niche is conserved between the murine and human species and suggest that highly purified nonadherent cultures of niche cells may represent a useful novel technology to culture human hematopoietic stem and progenitor cells.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4-4 ◽  
Author(s):  
Simon Mendez-Ferrer ◽  
Grigori N. Enikolopov ◽  
Sergio Lira ◽  
Paul S. Frenette

Abstract The identity of mesenchymal stem cells (MSCs) and their relationship to hematopoietic stem cells (HSCs) remain poorly defined. In addition, there are discrepancies regarding the cellular constituents of the HSC niche, with studies suggesting a role for bone-lining osteoblasts, and other data implicating sinusoidal endothelial and adventitial reticular cells. Previous work from our group has demonstrated that the sympathetic nervous system (SNS) is critical for both physiological and enforced egress of HSCs from the bone marrow (BM). HSC mobilization induced by G-CSF requires signals from the SNS (Katayama et al. 2006; Cell124:407–21). Physiological release of HSCs into the bloodstream follows circadian oscillations governed by the molecular clock and triggered by cyclical norepinephrine secretion by the SNS in the BM, activation of the β3-adrenergic receptor (encoded in Adrb3), degradation of Sp1 transcription factor and downregulation of Cxcl12 (Mendez-Ferrer et al. 2008; Nature452:442–7). Here, we have identified the cell targeted by the SNS in the BM as a perivascular stromal cell expressing Nestin, an intermediate filament protein characteristic of neuroectoderm-derived stem cells. Using transgenic mice expressing GFP under the regulatory elements of the Nestin promoter, we show that virtually all catecholaminergic fibers in the BM are associated with Nestin+ cells, which represent 4.0 ± 0.6% of BM CD45− cells and 0.08 ± 0.01% of total BM nucleated cells, as determined by FACS analyses. Quantitative real-time PCR (QPCR) analyses have revealed a ~30-fold higher expression of the gene encoding the chemokine CXCL12 in Nestin+ cells than in the rest of BM CD45− cells, whereas Adrb3 was exclusively expressed in Nestin+ cells and not detectable in Nestin− CD45− cells. Detailed immunofluorescence analyses of the spatial distribution of HSCs in longitudinal BM sections revealed that 60% of CD150+ CD48−/Lineage− cells were directly attached to Nestin+ cells, and 90% of HSCs were located within 5 cell diameters from Nestin+ cells in the endosteal or sinusoidal regions of the BM (n=30). In long-term BM cultures, Nestin+ cells were rare, but located near HSCs/progenitors-enriched cobblestone-forming areas. BM Nestin+ cells were associated with HSCs not only physically but also functionally, because core HSC retention signals (Cxcl12, Kitl, Vcam1, Angpt1, Il7) were highly expressed by Nestin+ cells and significantly downregulated during G-CSF-induced mobilization, whereas the expression of the same genes was significantly lower and was not downregulated by G-CSF in Nestin− CD45− cells, as measured by QPCR. A non-selective β- or a selective β3-adrenergic receptor agonist also downregulated these core HSC retention genes, underscoring the role of the SNS in regulating HSC adhesion in the BM niche. Cell sorting of Nestin+ CD45− and Nestin− CD45− cells revealed that all the mesenchymal progenitor activity of the bone marrow (CFU-F) was contained in the Nestin+ cell fraction. Further, Nestin+ cells could robustly differentiate into osteoblasts and adipocytes. Lineage-tracing studies using a Nestin-CRE transgenic line bred to R26R reporter mice have confirmed the contribution of Nestin+ cells to osteoblasts and chondrocytes during development. G-CSF, which induces proliferation of hematopoietic cells in the BM at the expense of non-hematopoietic lineages, significantly downregulated markers of osteoblastic and adipogenic differentiation in BM Nestin+ CD45− cells but not in Nestin− CD45− cells. By contrast, daily administration of parathyroid hormone over five weeks, a treatment previously shown to expand both the osteoblastic and HSC pools, induced proliferation of Nestin+ cells and favored their differentiation into Col1a1-LacZ+ osteoblasts. Finally, we have found that Nestin+ CD45− cells, but not Nestin− CD45− cells, can form self-renewing spheres in clonal density culture, with a frequency similar to other neural crest-derived stem cells. After two weeks in culture, clonal spheres showed spontaneous multilineage differentiation into adipocytes and Col1a1-LacZ+ osteoblasts. Altogether, these results suggest that the HSC niche is composed of a heterotypic MSC-HSC pairing that is tightly regulated by the SNS. This association may reconcile divergent views regarding the vascular and osteoblastic locations of the HSC niche, and its regulation by the SNS might explain the crosstalk between hematopoietic and mesenchymal lineages in the BM during health and disease.


2019 ◽  
Vol 3 (3) ◽  
pp. 419-431 ◽  
Author(s):  
Fang Dong ◽  
Haitao Bai ◽  
Xiaofang Wang ◽  
Shanshan Zhang ◽  
Zhao Wang ◽  
...  

Abstract The cell of origin, defined as the normal cell in which the transformation event first occurs, is poorly identified in leukemia, despite its importance in understanding of leukemogenesis and improving leukemia therapy. Although hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were used for leukemia models, whether their self-renewal and differentiation potentials influence the initiation and development of leukemia is largely unknown. In this study, the self-renewal and differentiation potentials in 2 distinct types of HSCs (HSC1 [CD150+CD41−CD34−Lineage−Sca-1+c-Kit+ cells] and HSC2 [CD150−CD41−CD34−Lineage−Sca-1+c-Kit+ cells]) and 3 distinct types of HPCs (HPC1 [CD150+CD41+CD34−Lineage−Sca-1+c-Kit+ cells], HPC2 [CD150+CD41+CD34+Lineage−Sca-1+c-Kit+ cells], and HPC3 [CD150−CD41−CD34+Lineage−Sca-1+c-Kit+ cells]) were isolated from adult mouse bone marrow, and examined by competitive repopulation assay. Then, cells from each population were retrovirally transduced to initiate MLL-AF9 acute myelogenous leukemia (AML) and the intracellular domain of NOTCH-1 T-cell acute lymphoblastic leukemia (T-ALL). AML and T-ALL similarly developed from all HSC and HPC populations, suggesting multiple cellular origins of leukemia. New leukemic stem cells (LSCs) were also identified in these AML and T-ALL models. Notably, switching between immunophenotypical immature and mature LSCs was observed, suggesting that heterogeneous LSCs play a role in the expansion and maintenance of leukemia. Based on this mouse model study, we propose that acute leukemia arises from multiple cells of origin independent of the self-renewal and differentiation potentials in hematopoietic stem and progenitor cells and is amplified by LSC switchover.


Cells ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 221
Author(s):  
Yu-Hee Kim ◽  
Kyung-Ah Cho ◽  
Hyun-Ji Lee ◽  
Minhwa Park ◽  
Sang-Jin Shin ◽  
...  

Cotransplantation of mesenchymal stem cells (MSCs) with hematopoietic stem cells (HSCs) has been widely reported to promote HSC engraftment and enhance marrow stromal regeneration. The present study aimed to define whether MSC conditioned medium could recapitulate the effects of MSC cotransplantation. Mouse bone marrow (BM) was partially ablated by the administration of a busulfan and cyclophosphamide (Bu–Cy)-conditioning regimen in BALB/c recipient mice. BM cells (BMCs) isolated from C57BL/6 mice were transplanted via tail vein with or without tonsil-derived MSC conditioned medium (T-MSC CM). Histological analysis of femurs showed increased BM cellularity when T-MSC CM or recombinant human pleiotrophin (rhPTN), a cytokine readily secreted from T-MSCs with a function in hematopoiesis, was injected with BMCs. Microstructural impairment in mesenteric and BM arteriole endothelial cells (ECs) were observed after treatment with Bu–Cy-conditioning regimen; however, T-MSC CM or rhPTN treatment restored the defects. These effects by T-MSC CM were disrupted in the presence of an anti-PTN antibody, indicating that PTN is a key mediator of EC restoration and enhanced BM engraftment. In conclusion, T-MSC CM administration enhances BM engraftment, in part by restoring vasculature via PTN production. These findings highlight the potential therapeutic relevance of T-MSC CM for increasing HSC transplantation efficacy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. SCI-26-SCI-26
Author(s):  
Simón Méndez-Ferrer

Hematopoietic stem cells (HSCs) traffic between bone marrow and circulation, what allows for life-saving clinical transplantation. Our previous work has shown that HSC numbers in blood follow circadian oscillations that are regulated by the central pacemaker in the brain, which reaches bone marrow nestin+ mesenchymal stem cells through peripheral sympathetic nerves. In the perinatal bone marrow, HSC-niche forming mesenchymal stem cells might be different from those that form the skeleton and some of them might be neural crest-derived, like peripheral neurons and supporting glial cells. Thus, tight regulation of the bone marrow stem-cell niche in vertebrates might build upon developmental relationships of its cellular components. We have found recently that cholinergic nerves regulate HSC maintenance, proliferation and migration in divergent niches. We will present unpublished evidence of how both branches of the autonomic nervous system cooperate to regulate HSC maintenance and function in spatially and temporally distinct niches. Moreover, we have shown recently that damage to this regulatory network is essential for the manifestation of myeloproliferative neoplasms. In these diseases, previously thought to be driven solely by mutated HSCs, protecting the HSC niche might represent a novel therapeutic strategy. Patients with myeloproliferative neoplasms have a higher risk of developing acute leukemia. However, at this stage, leukemic cells might be less sensitive to the normal control by the microenvironment and, instead, acute myelogenous leukemic cells might transform the bone marrow niches to support their own survival. We will discuss potential contributions of HSC niches to myeloproliferative neoplasms and MLL-AF9-driven acute myeloid leukemia. Disclosures Off Label Use: Potential use of selective estrogen receptor modulators and beta3-adrenergic agonists in myeloproliferative neoplasms.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5805-5805
Author(s):  
Lukun Zhou ◽  
Shuang Liu ◽  
Chuanyi M. Lu ◽  
Jianfeng Yao ◽  
Yuyan Shen ◽  
...  

Abstract Liver injury associated with veno-occlusive disease and graft-versus-host disease (GVHD) is a frequent and severe complication of hematopoietic stem cell transplantation, and remains an important cause of transplant-related mortality. Bone marrow derived mesenchymal stem cells (MSCs) have been evaluated for the prevention and treatment of refractory GVHD. However, poor cell viability has limited the therapeutic capacity of mesenchymal stromal cell therapy in vivo. In this study, we genetically engineered C57BL/6 mouse bone marrow MSCs using ex vivo retroviral transduction to overexpress Akt1, a serine threonine kinase and pro-survival signal protein, and tested the hypothesis that Akt1-expressing MSCs (Akt1-MSCs) are more resistant to apoptosis and can ameliorate acute liver injury induced by concanavalin A (ConA) in BALB/c mice. Cell proliferation and apoptosis analyses showed that, under both regular culture and high concentration IFN-γ (100 ng/mL) stimulation conditions, Akt1-GFP-MSCs had proliferation and survival (anti-apoptotic) advantages with down-regulated apoptosis pathways, compared to control GFP-MSCs. Twenty-four hours after receiving lethal dose of ConA (40 mg/kg, intravenous) (N=10 each group), no mouse survived, with or without 1x106 Akt1-MSCs or GFP-MSCs administration (intravenous); however, 3 and 1 survived in the 5×106 Akt1-MSCs group and 5×106 GFP-MSCs groups, respectively. In subsequent sub-lethal dose ConA (20 mg/kg) experiments, compared to GFP-MSCs, mice received Akt1-MSCs administration had significantly lower serum AST, ALT, TNF-α and IFN-γ levels and less histopathological abnormalities. In addition, Akt1-MSCs treated mice had significantly higher serum concentrations of IL-10, vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF). In vivo imaging showed that, hepatic fluorescence signal in sub-lethal ConA+Akt1-MSCs group was significantly stronger than ConA+GFP-MSCs group on day 0, and persisted up to 14 days, whereas the signal in ConA+GFP-MSCs, Akt1-MSCs and GFP-MSCs groups was negligible on both day 7 and day 14. Thus, bone marrow derived MSCs genetically enhanced with Akt1 had survival advantage in vitro and in vivo, and have the potential to be a potent therapy for prevention and amelioration of GVHD-associated liver impairment. Further translational pre-clinical studies are ongoing to further determine the efficacy, dosage and timing of administration of Akt1-MSCs in animal models. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 2011 ◽  
pp. 1-8 ◽  
Author(s):  
Tian Li ◽  
Yaojiong Wu

Hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) are both adult stem cells residing in the bone marrow. MSCs interact with HSCs, they stimulate and enhance the proliferation of HSCs by secreting regulatory molecules and cytokines, providing a specialized microenvironment for controlling the process of hematopoiesis. In this paper we discuss how MSCs contribute to HSC niche, maintain the stemness and proliferation of HSCs, and support HSC transplantation.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4045-4045
Author(s):  
Ferda Tekinturhan ◽  
Ludovic Zimmerlin ◽  
Vera S. Donnenberg ◽  
Melanie E. Pfeifer ◽  
Darlene A. Monlish ◽  
...  

Abstract Bone marrow (BM) contains hematopoietic stem cells (HSCs), which can give rise to all mature blood cells and marrow stromal cells as well. Recently, it has been shown that non-hematopoietic stem/progenitor cells which can differentiate into non-hematopoietic tissues also reside in the BM. Although culture expanded cells have been studied in great detail, little is known about the phenotype and quantity of these cells in freshly harvested adult human BM. The aim of this study is to isolate and characterize hematopoietic and non-hematopoietic stem/progenitor cells in adult human BM by comparing two different isolation techniques and their effects on the yield of hematopoietic, mesenchymal and endothelial stem/progenitor cell populations. BM samples were collected mechanically from isolated rib specimens obtained during lung resection (n=10), or from BM aspirates harvested from the humerus of orthopedic patients (n=17). BM mononuclear cells were purified on a Ficoll/Hypaque density gradient and stained simultaneously using CD105 FITC, CD73 PE, CD34 ECD, CD90 PE.Cy5, CD117 PE.Cy7, CD133 APC, CD45 APC.Cy7 and DAPI as a marker of nucleated cells. 2–15 million cells per sample were acquired on a Dako CyAn cytometer and the data were analyzed offline using prototype analytical software (Venturi, Applied Cytometry Systems). The significant difference in the percentage of the CD45 − singlets (non-hematopoietic cells) between BM aspirates and rib-derived samples indicates hemodilution in the bone marrow aspirates. Although we have observed a slight difference in the mean of hematopoietic stem cell content between samples, it was not statistically significant. According to our results, the quantity of mesenchymal stem cells was higher in rib-derived BM than BM aspirates (p value=0.028). The expression of some stem/progenitor cell markers, such as CD90 (Thy-1), CD117 (c-Kit) and CD133 remained similar for all cell types. Our results are shown in the table below. Surface Antigens RibBM (n=10)¥ BMA (n=17)¥ p Value % % Total Cells CD45- of nucleated cells 15.3 ± 7.9 5.7 ± 5.2 0.004 CD34+ Hematopoietic Stem Cells (HSCs)* CD34 of CD45+ 1.7 ± 1.48 2.6 ± 2.0 0.883 CD117 74.6 ± 31.3 53.3 ± 18.8 0.073 CD90 60.3 ± 44.5 35.9 ± 36.5 0.134 CD133 70.3 ± 31.8 62.3 ± 21.4 0.443 Endothelial Progenitor Cells (EPCs)* EPCs of nucleated cells 0.05 ± 0.03 0.12 ± 0.2 0.323 CD117 81.3 ± 29.8 78.1 ± 20.2 0.746 CD90 66.7 ± 39.7 53.7 ± 31.4 0.356 CD133 45.9 ± 32.7 33.9 ± 22.0 0.265 Mesenchymal Stem Cells (MSCs)* MSCs of nucleated cells 0.086 ± 0.14 0.008 ± 0.01 0.028 CD117 60.2 ± 36.8 49.8 ± 34.3 0.471 CD90 66.0 ± 27.7 65.7 ± 29.1 0.981 CD133 37.8 ± 27.4 39.9 ± 28.9 0.857 RibBM: Rib-derived BM, BMA: Bone Marrow Aspirate ¥Data are given as mean ± SD. *CD90, CD117 and CD133 expressions are shown for each stem/progenitor fraction: Hematopoietic stem cells (CD34 + CD45 + and light scatter properties according to the ISHAGE protocol), endothelial progenitor cells (CD34bright CD45 − CD105 +) and mesenchymal stem cells (CD34 − CD45 − CD73 + CD105 +).


Sign in / Sign up

Export Citation Format

Share Document