Skip to main content

Optimization of seeding density of OP9 cells to improve hematopoietic differentiation efficiency

Abstract

Background

OP9 mouse stromal cell line has been widely used to induce differentiation of human embryonic stem cells (hESCs) into hematopoietic stem/progenitor cells (HSPCs). However, the whole co-culture procedure usually needs 14–18 days, including preparing OP9 cells at least 4 days. Therefore, the inefficient differentiation system is not appreciated. We aimed to optimize the culture conditions to improve differentiation efficiency.

Methods

In the experimental group, we set six different densities of OP9 cells and just cultured them for 24 h before co-culture, and in the control group, OP9 cells were cultured for 4 days to reach an overgrown state before co-culture. Then we compared the hematopoietic differentiation efficiency among them.

Results

OP9 cells were randomly assigned into two groups. In the experimental group, six different plated numbers of OP9 cells were cultured for 1 day before co-culture with hESCs. In contrast, in the control group, OP9 cells were cultured for 4 days at a total number of 3.1 × 104 cells/cm2 in a 6-well plate to reach an overgrown state before co-culture. Hematopoietic differentiation was evaluated with CD34 immunostaining, and compared between these two groups. We could not influence the differentiation efficiency of OP9 cells with a total number of 10.4 × 104 cells/cm2 in a 6-well plate which was cultured just for 1 day, followed by co-culture with hESCs. It reached the same differentiation efficiency 5 days earlier than the control group.

Conclusion

The peak of CD34 + cells appeared 2 days earlier compared to the control group. A total number of 1.0 × 106 cells in a 6-well plate for OP9 cells was appropriate to have high differentiation efficiency.

Peer Review reports

Background

Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), have an inexhaustible capacity for self-renewal and differentiation into cells of the three germ layers. Therefore, PSCs are an attractive source for generating hematopoietic stem cells (HSCs) [1, 2], which are the core of hematopoiesis for their capability of cell proliferation and differentiation into different blood and immune cells [3, 4]. To date, allo-genetic HSC transplantation (allo-HSCT) is the only definitive cure for many malignant blood diseases, cancers, and genetic immune disorders. Primary HSCs are exclusively obtained from bone marrow, mobilized peripheral blood, and cord blood, however, the major problems are the limitation of HLA-matched donor sources and the difficulty of obtaining sufficient HSCs for transplantation, and hinder the use of stem cell therapy in the clinic. For the past several decades, methods have been developed to direct the differentiation of hPSCs to hematopoietic stem/progenitor cells (HSPCs) in vitro using either formation of the embryonic body (EB) [5], or co-culture with stromal cell lines that imitate hematopoietic microenvironment, such as OP9, S17, and AGM [6,7,8].

OP9 mouse stromal cell line has been widely used to induce hPSCs differentiation to HSPCs and multi-lineage hematopoietic cells [9,10,11]. One advantage of this co-culture system to induce hESCs differentiation into HSPCs is that it does not need additional cytokines. However, the disadvantage of this system is its low differentiation efficiency. The whole co-culture procedure usually needs 14–18 days, including preparing OP9 cells for at least 4 days. Previous studies optimized the OP9 co-culture system with a focus on adding extrinsic cytokines [10, 12] and stromal cell gene editing [9].

Traditional differentiation system does not set a precise density of OP9 cells for co-culture, i.e. overgrown OP9 cells were prepared by feeding and prolonged culture of confluent OP9 cells, followed by induction of hESCs differentiation into HSPCs for 10 days [6, 13]. We hypothesized that the density of stromal cells in a co-culture system can influence the efficiency of hematopoietic differentiation. In the present study, we examined the effects of different densities of OP9 cells in co-culture systems and determined the optimal density for efficient differentiation. We hope that this optimized system can be used as a new induction method for in vitro HSPC differentiation.

Methods

Cell culture

The hESCs line (HN14, 23–30 passages) was obtained from the Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research and maintained on Matrigel-coated plates with hPSCs medium mTeSRâ„¢1 (Stem Cell Technologies, CAN) and passaged every 3 days at a 1:3 ratio using 0.5 mM EDTA, and changed fresh medium every day. Cells were maintained at 37 Â°C and 5% CO2 [14].

OP9 stromal cells (3–15 passages) were purchased from the Shanghai Cell Bank of the Chinese Academy of Sciences and cultured on 0.1% gelatinized plates in α-MEM (Gibco, USA) supplemented with 15% fetal bovine serum (FBS; Gibco, USA) until confluence. When passaged, these cells dissociated with 0.25% trypsin (Gibco) for 1 min at 37 Â°C. For cryopreservation, OP9 cells were suspended in 90% FBS added 10% Dimethyl Sulfoxide (Gibco) and stored in a deep freezer at -80 Â°C overnight, then transferred to liquid nitrogen.

OP9 cells and HN14 cells were co-cultured in a differentiation medium consisting of α-MEM medium supplemented with 10% defined fetal bovine serum (Hyclone, USA) and 100 Î¼mol/L monothioglycerol (MTG; Sigma, USA) [9].

Different co-culture strategies

OP9 cells were randomly assigned into two groups, experimental and control groups. In the experimental group, six different seeding numbers of OP9 cells were designed for each subgroup: 3.1 × 104cells/cm2, 5.2 × 104cells/cm2, 7.3 × 104cells/cm2, 10.4 × 104cells/cm2, 13.5 × 104cells/cm2 and 16.6 × 104cells/cm2 in 6-well plates. hESCs were plated on the related stromal cell layer (0.7–1.0 × 106 cells in 6-well plate) [9]. These cells were cultured for 1 day and co-cultured with hESCs. In the control group, OP9 cells at a seeding number of 3.1 × 104cells/cm2 in 6-well plates were cultured for 4 days to reach a confluent state followed by co-culture with hESCs. Undifferentiated hESCs were harvested by treatment with 2 mg/ml dispase enzyme (Gibco, USA) and scraped to maintain the cells in small clumps. The hESCs/OP9 cocultures were incubated for up to 10 days at 37 Â°C in normoxic conditions and 5% CO2 with a whole-medium change on day 1 and a half-medium change on days 4, 6, 8, and 10.

Immunofluorescence staining

To determine the identity of cell line OP9, we examined the immunophenotype of OP9. They were negative for CD34 and uniformly positive for Calponin [15]. They were fixed with methanol-acetone (1:1) for 30 min at -20 Â°C. After washing in phosphate-buffered saline (PBS), cells were fixed with 0.05% Tween-20 and 10% goat serum in PBS for 1 h at room temperature, then incubated with primary antibodies against Calponin (Abcam, USA) and CD34 (Abcam, USA) overnight at 4 Â°C. Then they were washed 3 times and incubated with Donkey Anti-Rabbit IgG (H&L) (Abcam, USA) for an additional 60 min at room temperature. Finally, the cells were incubated with 4′6-diamidino-2-phenylindole (DAPI) for 20 min at room temperature.

Flow cytometry analysis

To analyze the differentiation efficiency of the coculture system, hESCs/OP9 coculture cells were treated with 0.25% trypsin (Gibco, USA) for 20 min at 37 Â°C on days 8, 10 and 12. Trypsinization was terminated by adding a coculture medium. Co-culture cells were harvested by centrifugation, and washed twice by washing buffer (PBS supplemented by 5% FBS) [16]. These cells were immediately incubated with APC-human TRA-1–85 (R&D, USA), PE-Cy5.5 mouse anti-human CD34 (Becton Dickinson Company, USA) for 30 min at 4 Â°C. After staining, these cells were washed with washing buffer, centrifuged, and resuspended with moderate washing buffer, followed by flow cytometric analysis (BD FACSAriaâ„¢ II sorter (BD Biosciences, USA). Data were analyzed with FlowJo version 7.6.1 (FlowJo, LLC). For negative control, APC mouse anti-human CD34 (Becton Dickinson Company, USA) was used to HN14, while OP9 cells labeled anti-CD34 [EP373Y] (Abcam, USA) and used Donkey Anti-Rabbit IgG (H&L) (Abcam, USA) as the secondary antibody.

Magnetic sorting for CD34 positive cells

Single-cell suspension from day 10 of hESCs/OP9 co-cultures was labeled with CD34 Microbeads Kit (MACS) as recommended by the manufacturer and went through an LS separation column attached to a Midi-MACS Separator to obtain CD34+ cells. The purity of isolated CD34 + cells was always higher than 95% as evaluated by trypan blue exclusion.

RNA isolation, RT-PCR, and quantitative RT-PCR

Total RNA was extracted from cultured cells using TRIzol reagent (Invitrogen, USA). cDNA was synthesized using 1 Î¼g of RNA from each sample with iScriptâ„¢ cDNA Synthesis Kit (Promega, USA). RT-PCR cycling conditions were as follows: initial denaturation at 95 Â°C for 10 min followed by 40 denaturation cycles at 95 Â°C for 15 s, primer annealing at 60 Â°C for 60 s, primer extension phase at 72 Â°C 15 s and final extension step at 72 Â°C for 7 min (Agilent Stratagene Mx3000P, Agilent Technologies, USA). GAPDH was used as an internal control. All experiments were performed three times. Sequences of primers used in this study are shown in Table 1.

Table 1 Primer sequences for RT-PCR

Colony-forming assay

Sorted CD34+ cells were plated at a density of 2.5 × 104 cells/dish and suspended in 1 ml methylcellulose medium (MethoCult H4434, Stem Cell Technologies) in 35 mm low adherent dishes at 37 Â°C in a humidified atmosphere of 5% CO2 for 14–16 days. All clonogenic progenitor assays were performed in duplicate. Colonies of more than 50 cells were scored as positive clones and defined as macrophage (M), granulocyte and macrophage (GM), granulocyte (G), erythroid (E) according to their colony morphology.

Statistical analysis

Data were analyzed with Prism 5 software (GraphPad, USA), and are presented shown as the mean ± SD. Significant differences were considered at the P < 0.05 level.

Results

Identification of OP9 cells and differentiation of hESCs

The OP9 cells were bipolar fibroblast-like in morphology. They were morphologically homogeneous populations. Immunofluorescence staining was used to assess the molecular characteristics of the OP9 (Fig. 1A), the OP9 cells were negative for CD34 and expressed a marker of Calponin. As shown in Fig. 1D, OP9 cells were cultured for 1 day with six different seeding numbers in 6-well plates: a to f were the morphology of different seeding numbers,3.1 × 104cells/cm2,5.2 × 104cells/cm2,7.3 × 104cells/cm2,10.4 × 104cells/cm2,13.5 × 104cells/cm2and 16.6 × 104cells/cm2, respectively. OP9 cells plated at the numbers of 1.0–1.3 × 106 cells reached nearly 100% confluency after 1 day of culture (d, e). When the seeding number of OP9 cells could reach 16.6 × 104cells/cm2, the cultures reached confluence. In the control group, overgrown OP9 cultures were prepared for 4 days (Fig. 1C). HN14 cells were plated on the stromal cell layer when they reached 80% confluency. During the differentiation process, the morphology of cells started to change on days 8, 10, and 12 and hESCs started differentiating through the morphologic observation (Fig. 1B). As shown in Fig. 2, HN14 cells seeded on six different numbers of OP9 cells, 3.1 × 104 cells, 5.2 × 104 cells, 7.3 × 104 cells, 10.4 × 104 cells, 13.5 × 104 cells, and 16.6 × 104 cells per square centimeter, respectively, on days 8, 10 or 12 (bar = 100 Î¼m).

Fig. 1
figure 1

Identification and morphology of OP9 cells and hESCs before co-culture. A Immunophenotype of OP9 cells. Immunofluorescence staining of Calponin and CD34 in the OP9. Original magnification: × 40. B The morphology of hESCs (HN14) before co-culture (bar = 100 Î¼m). hESCs monolayers at 80% confluence and co-cultured with OP9 cells. C The morphology of OP9 cells in the control group. OP9 cells reached confluence for culturing for 4 days at a seeding number of 3.1 × 104 cells/cm2 in 6-well plates (bar = 100 Î¼m). D The morphology of OP9 cells in the experimental group. OP9 cells were cultured for 1 day with six different seeding numbers in 6-well plates: a to f show the morphology of different seeding densities, i.e. 3.1 × 104 cells, 5.2 × 104 cells, 7.3 × 104 cells, 10.4 × 104 cells, 13.5 × 104 cells, and 16.6 × 104 cells per square centimeter, respectively (bar = 100 Î¼m). When the seeding number of OP9 cells could reach 16.6 × 104cells/cm2, the cultures reached confluence

Fig. 2
figure 2

The morphology of ES cells on OP9 stromal cells during differentiation on days 8, 10 and 12. (A) a to f show the morphological changes of HN14 cells on six different seeding numbers of OP9 cells, i.e. 3.1 × 104 cells, 5.2 × 104 cells, 7.3 × 104 cells, 10.4 × 104 cells, 13.5 × 104 cells, and 16.6 × 104 cells/cm2, respectively, on days 8, 10 or 12 (bar = 100 Î¼m). B The morphologies of differentiated HN14 colonies during co-culture on days 8, 10 and 12 in the control group (bar = 100 Î¼m). In all groups, the presence of rounded cells at the edge of the colonies

Effects of seeding density on the expression of CD34, CD43, and CD31

Using flow cytometry analysis, we could easily distinguish human cells from OP9 because of APC-human TRA-1–85. CD34 is an important surface marker for HSPCs and can define HSPCs in the human hESCs/OP9 co-culture system [15]. CD34 was not expressed in original cells including HN14 and OP9 cells (Fig. 3A). During differentiation, we harvested co-culture cells on days 8, 10 and 12, and analyzed the expression of CD34 by flow cytometry. As shown in Fig. 3C, a to f were the six different seeding numbers of OP9 cells from 3.1 × 104 cells to 16.6 × 104 cells per square centimeter in 6-well plates. With the increasing density of OP9 cells, the efficiency of hematopoietic differentiation was improved from 2.47% to 6.27%. The group of seeding number of 10.4 × 104cells resulted in 10% of CD34+ cells or even more on day 10. Except for the seeding number of 3 × 105cells in a 6-well plate, all other groups had a peak of CD34+ cells on day 10 during hematopoietic differentiation. However, the control group had a peak of CD34 + cells on day 12 or later (Fig. 3B). These findings suggested that optimizing the seeding density of OP9 cells improves hESCs hematopoietic differentiation. Consistent with the results of flow cytometry, the expression of CD34 mRNA between 10,4 × 104cells, 13.5 × 104 cells, and 16.6 × 104 cells/cm2 groups was comparable (Fig. 3D).

Fig. 3
figure 3

Determination of differentiation efficiency in the hESCs/OP9 co-culture system. A The expression of CD34 in undifferentiated human embryonic stem cells (HN14) and OP9 cells, separately. B Percentage of CD34+ cells by flow cytometry (x-axis) in the control group. The peak of hematopoietic differentiation on D12. C Percentage of CD34+ cells by flow cytometry (x-axis) in the experimental group. a to f show six different seeding numbers in 6-well plates, 3.1 × 104 cells, 5.2 × 104 cells, 7.3 × 104 cells, 10.4 × 104 cells, 13.5 × 104 cells, and 16.6 × 104 cells/cm2, respectively. Single-cell suspension from hESCs/OP9 co-culture obtained at the indicated time points was labeled with CD34 (x-axis). The number indicates the percentage of positive cells in the corresponding group. Except for (a) group, the peak of hematopoietic differentiation on D10. D Evaluation of the expression of hematopoietic-related genes on day 10 in the experimental group. RNA was extracted, and gene expression was quantified by qRT-PCR. Statistical results of phase-specific genes are shown as the mean ± SD, n = 3. ## P < 0.01 and # P < 0.05 compared with the 16.6 × 104 cells/cm2 group. ▲▲ P < 0.01 and ▲ P < 0.05 compared with the 13.5 × 104 cells/cm2 group. ★★ P < 0.01 and ★ P < 0.05 compared with the 10.4 × 104 cells/cm2 group

We also investigated the expression of CD31 and CD43 in these cells on day 10 (Fig. 3C) by qRT-PCR. CD43 and CD31 were also markers of HSPCs [3]. When the seeding number was up to 16.6 × 104 cells per square centimeter in a 6-well plate, the expression of CD43 was increasing. We also found that when the seeding number was 10.4 × 104 cells per square centimeter in a 6-well plate, CD31 expression increased more obviously than other densities. Likewise, the expression level of CD43 and CD31 reached a peak on day 10 in the experimental group.

No effect of seeding density on the differentiation of HSPCs

The purpose of this study was to optimize the co-culture system. We compared the expression of CD34, CD31, and CD43 between the experimental group on day 10 and the control group on day 12. The expression of CD34, CD31 and CD43 had no significant difference (Fig. 4A). We enriched CD34 + cells with magnetic cell sorting (Fig. 4B and C) and promoted them to differentiate into different CFUs, such as macrophage (M), granulocyte and macrophage (GM), granulocyte (G), erythroid (E). Consistent with the observations from flow cytometry and RT-qPCR, the proficiency of further differentiation had no difference between the two groups when the seeding density reached 5.0 × 105cells/ml.

Fig. 4
figure 4

Improving the initial seeding number of OP9 cells shortened the whole time of co-culture system without changing the efficiency of hematopoietic differentiation. A Comparison of the expression of hematopoietic-related genes during differentiation between the experimental subgroup (10.4 × 104 cells, 13.5 × 104 cells, and 16.6 × 104 cells/cm2 in a six-well plate) and the control group. Data are presented as means ± SD (n = 3). These results were consistent with flow cytometry. B Cell distribution in CFU from sorted CD34 positive cells for culturing 14–16 days in a Semi-solid medium. E, erythroid; G, granulocyte; GM, granulocyte/macrophage; M, macrophage. C Morphology of different colony-forming unit types, including M, GM, G and E. Scale bar = 100 mm

Discussion

Stromal cells are one of the most important cell types in bone marrow that contain the hematopoietic niche, and hematopoiesis is regulated by microenvironment or niche [17, 18]. Some studies attempted to derive robustly induced HPCs from PSCs in vitro by simulating hematopoietic niches [18,19,20], and others tried to improve techniques to mimic the hematopoietic niche. The objective of these studies was to improve HSC differentiation to a large scale so that it may be used in regenerative medicine applications.

The co-culture technique was used for many applications, and the efficiency of differentiation was closely related to the condition of the stromal cells. Stromal cells producing M-CSF induce the differentiation of embryonic stem cells (ESC) down the monocyte-macrophage lineage [20, 21]. Nevertheless, OP9 stromal cells lacking M-CSF promote diferentiation down other haematopoietic lineages [6, 22, 23].

OP9 cells are one of the stromal cells that have been widely used because they provide more efficient support of hematopoiesis than other stromal cell lines probably due to that OP9 could release paracrine mediated signaling [24, 25]. In previous studies, hPSCs seeded onto overconfluent OP9 stroma, after 12 days of co-culture, with OP9 cells, the percentage of CD34 positive cells reached 10.3 ± 1.8%, but did not define the seed density and culture time before co-culture [26]. As the efficiency of co-culture was not stable, most of the studies focused on modification of gene expression, the function of OP9 stromal cells, or add hematopoietic-related cytokines to improve differentiation efficiency, previous studies showed that over-expressed transcription factor Lhx2 onto OP9 cells [9] or modified extrinsic factors directly on OP9 cells [27] enhanced hPSCs hematopoietic differentiation. With an in-depth understanding of hematopoietic differentiation, OP9 cells were modified with Notch ligand delta-like 1 to develop T lymphocytes [28] and NK cells [29]. Importantly, OP9 cells secreted factors and signaling molecules to induce hematopoietic differentiation [30, 31]. One study used single-cell q-PCR to track the dynamic gene expression of OP9-hESCs co-culture system at different stages and transiently over-expressed specific transcription factors (TFs) at different stages [32] and showed that over-expression of these TFs mainly promoted differentiation at the initial stage. Another recent study used 3D cell culture by co-culture of OP9 cells, and provided a minimal, scalable, biomimetic in vitro model of hematopoietic differentiation in the absence of exogenous cytokines [33]. Although OP9 stromal cells are universally used in hematopoietic differentiation. The efficiency of HSPC generation is related to the density of OP9 cells, however, no study explored the association between the density of OP9 cells with hematopoietic differentiation. We suggest that the longer OP9 cells were cultured, the more aging OP9 cells would become, and aging OP9 cells are prone to fat degeneration and insufficiency of secreting cytokines.

In the present study, to define the density of OP9 cells, we established an optimized OP9 co-culture system without adding any recombinant cytokines (Fig. 5). We focused on the seeding density of OP9 cells in the co-culture system. A seeding number of 16.6 × 104 cells per square centimeter in a 6-well plate of OP9 cells was able to reduce culture time without significantly affecting the differentiation efficiency compared to previous study [26]. For example, the initial seeding density of OP9 cells at 10.4 × 104 cells/cm2 in a 6-well plate had 10% percent of CD34+ cells at day 10, compared with day 12 in the control group. Increasing the density of OP9 cells further increased the percentage of CD34+ but showed no significant difference between the control and experimental groups. In other words, we reduced the differentiation time by 2 days with the same differentiation of efficiency. In addition, in our optimized OP9 co-culture system, HSPCs successfully differentiated into different CFUs (colony-forming units) with comparable efficiency, which meant that this system had the potential to produce multipotent hematopoietic progenitors. Thus, we have optimized the co-culture system to reduce the differentiation time for at least 5 days without changing differentiation efficiency.

Fig. 5
figure 5

Schematic diagram showing two different differentiation systems. As shown in the schematic diagram, the optimized method could shorten the whole differentiation procedure by nearly 5 days

Conclusions

Our optimized approach is a novel, faster, and low-cost method to induce hematopoietic differentiation in vitro. However, the underlying mechanisms remain unclear. Future studies are needed to identify the possible factors or signaling molecules secreted by OP9 cells at different seeding densities and determine which is responsible for the improved differentiation observed in the present study. In the future, we will test related factors to induce differentiation and further optimize the differentiation system in the absence of OP9 cells.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

hESC:

Human embryonic stem cells

HSPC:

Hematopoietic stem/progenitor cell

HPSC:

Human pluripotent stem cells

iPSC:

Induced pluripotent stem cell

EB:

Embryonic body

TF:

Transcription factors

CFU:

Colony-forming units

References

  1. Palacios R, Golunski E, Samaridis J. In vitro generation of hematopoietic stem cells from an embryonic stem cell line. Proc Natl Acad Sci U S A. 1995;92:7530–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Niwa A, Umeda K, Chang H, Saito M, Okita K, et al. Orderly hematopoietic development of induced pluripotent stem cells via Flk-1(+) hemoangiogenic progenitors. J Cell Physiol. 2009;221:367–77.

    Article  CAS  PubMed  Google Scholar 

  3. Morrison SJUN, Weissman IL. The biology of hematopoietic stem cells. Annu Rev Cell Dev Biol. 1995;11:35–71.

    Article  CAS  PubMed  Google Scholar 

  4. Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell. 2000;100:157–68.

    Article  CAS  PubMed  Google Scholar 

  5. Song B, Fan Y, He W, Zhu D, Niu X, et al. Improved hematopoietic differentiation efficiency of gene-corrected beta-thalassemia induced pluripotent stem cells by CRISPR/Cas9 system. Stem Cells Dev. 2015;24:1053–65.

    Article  CAS  PubMed  Google Scholar 

  6. Vodyanik MA, Bork JA, Thomson JA, Slukvin II. Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood. 2005;105:617–26.

    Article  CAS  PubMed  Google Scholar 

  7. Tian XWP, Morris JK, Linehan JL, Kaufman DS. Hematopoietic engraftment of human embryonic stem cell derived cells is regulated by recipient innate immunity. Stem Cells. 2006;24:1370–80.

    Article  CAS  PubMed  Google Scholar 

  8. Ledran MH, Krassowska A, Armstrong L, Dimmick I, Renstrom J, et al. Efficient hematopoietic differentiation of human embryonic stem cells on stromal cells derived from hematopoietic niches. Cell Stem Cell. 2008;3:85–98.

    Article  CAS  PubMed  Google Scholar 

  9. Chen XZQ, Li C, Geng Y, Huang K, Zhang J, Wang X, Yang J, Wang T, Xia C, Liu X, Meng M, Yang D, Zheng Y, Du J, Zhang X, Chen J, Pan G, Wang J. OP9-Lhx2 stromal cells facilitate derivation of hematopoietic progenitors both in vitro and in vivo. Stem Cell Reseach. 2015;15:395–402.

    Article  CAS  Google Scholar 

  10. Kennedy M, Awong G, Sturgeon CM, Ditadi A, LaMotte-Mohs R, et al. T lymphocyte potential marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures. Cell Rep. 2012;2:1722–35.

    Article  CAS  PubMed  Google Scholar 

  11. Yang CTFA, Goh PA, Pagnamenta A, Mettananda S, Taylor J, Knight S, Nathwani A, Roberts DJ, Watt SM, Carpenter L. Human induced pluripotent stem cell derived erythroblasts can undergo definitive erythropoiesis and co-express gamma and beta globins. Br J Haematol. 2014;166:435–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Choi KD, Vodyanik MA, Togarrati PP, Suknuntha K, Kumar A, et al. Identification of the hemogenic endothelial progenitor and its direct precursor in human pluripotent stem cell differentiation cultures. Cell Rep. 2012;2:553–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Vodyanik MA, Thomson JA, Slukvin II. Leukosialin (CD43) defines hematopoietic progenitors in human embryonic stem cell differentiation cultures. Blood. 2006;108:2095–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang K, Zhu Y, Ma Y, et al. BMI1 enables interspecies chimerism with human pluripotent stem cells. Nat Commun. 2018;9(1):4649.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Gao J, Yan XL, et al. Characterization of OP9 as authentic mesenchymal stem cell line. J Genet Genomics. 2010;37(7):475–82.

    Article  CAS  PubMed  Google Scholar 

  16. Fathi E, Valipour B, et al. Targeting the proliferation inhibition of chronic myeloid leukemia cells by bone marrow derived-mesenchymal stem cells via ERK pathway as a therapeutic strategy. Acta Med Iran. 2020;58(5):199.

    Google Scholar 

  17. Chadwick K, Wang L, Li L, Menendez P, Murdoch B, et al. Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood. 2003;102:906–15.

    Article  CAS  PubMed  Google Scholar 

  18. Morrison SJSD. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505:327–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Crane GM, Jeffery E, Morrison SJ. Adult haematopoietic stem cell niches. Nat Rev Immunol. 2017;17:573–90.

    Article  CAS  PubMed  Google Scholar 

  20. Klimchenko O, Mori M, Distefano A, Langlois T, Larbret F, et al. A common bipotent progenitor generates the erythroid and megakaryocyte lineages in embryonic stem cell-derived primitive hematopoiesis. Blood. 2009;114:1506–17.

    Article  CAS  PubMed  Google Scholar 

  21. Nakano T, Kodama H, Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science. 1994;265:1098–101.

    Article  CAS  PubMed  Google Scholar 

  22. Dias J, et al. Generation of red blood cells from human induced pluripotent stem cells. Stem Cells Dev. 2011;20:1639–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Trakarnsanga K, et al. Qualitative and quantitative comparison of the proteome of erythroid cells diferentiated from human iPSCs and adult erythroid cells by multiplex TMT labelling and nanoLC-MS/MS. PLoS ONE. 2014;9:e100874.

    Article  PubMed  PubMed Central  Google Scholar 

  24. McKinney-Freeman SL, Naveiras O. et al. Isolation of hematopoietic stem cells from mouse embryonic stem cells. Curr Protoc Stem Cell Biol Chapter 1: Unit 1F. 2008;3. https://doi.org/10.1002/9780470151808.sc01f03s4.

  25. Ji J, Vijayaragavan K, et al. OP9 stroma augments survival of hematopoietic precursors and progenitors during hematopoietic differentiation from human embryonic stem cells. Stem Cells. 2008;26(10):2485–95.

    Article  CAS  PubMed  Google Scholar 

  26. French A, Yang CT, Taylor S, Watt SM, Carpenter L. Human induced pluripotent stem cell-derived B lymphocytes express sIgM and can be generated via a hemogenic endothelium intermediate. Stem Cells Dev. 2015;24(9):1082–95.

    Article  CAS  PubMed  Google Scholar 

  27. Ichii M, Frank MB, Iozzo RV, Kincade PW. The canonical Wnt pathway shapes niches supportive of hematopoietic stem/progenitor cells. Blood. 2012;119:1683–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kaneko S. In vitro generation of antigen-specific T cells from induced pluripotent stem cells of antigen-specific T cell origin. Methods Mol Bio. 2016;1393:67.

    Article  Google Scholar 

  29. Kyoizumi S, Kubo Y, Kajimura J, Yoshida K, Imai K, Hayashi T, et al. Age- associated changes in the differentiation potentials of human circulating hematopoietic progenitors to T- or NK-lineage cells. J Immunol. 2013;190(12):6164.

    Article  CAS  PubMed  Google Scholar 

  30. Figueiredo LM, Costa EB, Orellana MD, Picanco-Castro V, Covas DT. OP9 Stromal Cells Proteins Involved in Hematoendothelial Differentiation from Human Embryonic Stem Cells. Cell Reprogram. 2015;17:338–46.

    Article  CAS  PubMed  Google Scholar 

  31. Trakarnsanga K, Wilson MC, Heesom KJ, Andrienko TN, Srisawat C, Frayne J. Secretory factors from OP9 stromal cells delay differentiation and increase the expansion potential of adult erythroid cells in vitro. Sci Rep. 2018;8:1983.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Chen H, Jiang M, Xiao L, Huang H. Single-cell qPCR facilitates the optimization of hematopoietic differentiation in hPSCs/OP9 coculture system. Braz J Med Biol Res. 2018;51:e7183.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tavakol DN, Tratwal J, Bonini F, Genta M, Campos V, et al. Injectable, scalable 3D tissue-engineered model of marrow hematopoiesis. Biomaterials. 2020;232:119665.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all staff in the Department of Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University (especially Dr. Fei Sun) for their help and support.

Funding

This work was supported in part by grants from the Major Science and Technology Program of Hainan Province (ZDK2021037), and Hainan Province Science and Technology Special Fund (ZDYF2020117, ZDYF2022SHFZ312), Hainan Province Science and Technology Project (LCXY202102, LCYX202203, LCYX202301) the project supported by Hainan Province Clinical Medical Center, the specific research fund of The Innovation Platform for Academicians of Hainan Province.

Author information

Authors and Affiliations

Authors

Contributions

J.X., M.Y., L.Q. and H.Y. initiated and designed the project. J.X., M.Y. and L.Q. wrote the manuscript. W.Y. and J.X. performed most experiments and analyzed the result data. H.Y., and M.Y. supervise the designed structure of the manuscript, review and help revise this manuscript.

Corresponding authors

Correspondence to Yuan-hua Huang or Yan-lin Ma.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Research Ethics Committee of the First Affiliated Hospital of Hainan Medical University.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

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

Jiang, Xx., Song, My., Li, Q. et al. Optimization of seeding density of OP9 cells to improve hematopoietic differentiation efficiency. BMC Mol and Cell Biol 25, 10 (2024). https://doi.org/10.1186/s12860-024-00503-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12860-024-00503-x

Keywords