- Research
- Open access
- Published:
HES1 is required for mouse fetal hematopoiesis
Stem Cell Research & Therapy volume 15, Article number: 235 (2024)
Abstract
Background
Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling.
Methods
In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay.
Results
We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo.
Conclusion
Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.
Introduction
Hairy Enhancer of Split 1 (HES1) is a basic helix-loop-helix (HLH) transcriptional repressor [1] and an evolutionarily conserved target of Notch signaling [2, 3]. It plays an essential role in the development of many organs by promoting the maintenance of stem/progenitor cells, controlling the reversibility of cellular quiescence, and regulating both cell fate decisions and the timing of several developmental events [4, 5]. HES1 is more than a repressor. For example, HES1 amplifies RUNX2 (Runt-related transcription factor 2) expression by cooperating with retinoblastoma protein (pRb) through its WRPW domain [6]. In addition, HES1 uses basic helix-loop-helix (bHLH) domain and orange domain to bind with STAT3 and activates STAT3 phosphorylation [7]. Other unknown function of HES1 remains to be elucidated.
Blood cells are continuously produced from a pool of progenitors that derived from HSCs. Balanced stem cell self-renewal and initiation of lineage specification program is essential for the development and homeostasis of the hematopoietic system [8]. Embryonic hematopoiesis begins in the yolk sac and transitions to definitive hematopoiesis in the fetal liver [9]. On embryonic day E9.5 ∼ 10.5 in murine development, hematopoietic stem cells (HSCs) migrate from their origin sites to the fetal liver for expansion [10,11,12], before terminal migrating to the bone marrow (BM) on E16.5 to 17.5 [10,11,12,13]. This transient residence in the fetal liver is essential for the maturation of adult HSCs and functional adult hematopoiesis [12,13,14]. It has been shown that fetal hematopoietic progenitors are functionally distinct from adult progenitors [15]. For example, murine fetal progenitors exhibit greater engraftment efficiency, biased lineage differentiation, and altered susceptibility to transformation compared with adult counterparts [16,17,18,19]. Analogous studies in humans showed that fetal and cord blood CD34+ cells are more proliferative and have a greater propensity to form myeloid colonies in methylcellulose culture than adult cells [20, 21]. Although fetal liver of Hes1-deficient embryos had been previously analyzed [22], neither HSC phenotype nor function were investigated. A recent study showed that cumulative deletion of Hes1 and Hes5 alleles leads to the generation of nonfunctional hematopoietic progenitors in the developing aorta-gonad-mesonephros (AGM) and the total absence of HSC activity [23]. Our previous studies have demonstrated that HES1 is dispensable for steady state adult hematopoiesis [4]. However, the precise role of HES1 in regulating embryonic hematopoiesis remains elusive.
Both p27 (cyclin-dependent kinase inhibitor 1B; also known as p27KIP1) and PTEN (phosphatase and tension homolog deleted on chromosome 10) are known HES1 targets [24, 25]. p27KIP1 regulates the G1 to S-phase transition by binding to and regulating the activity of cyclin-dependent kinases [26]. Tumor suppressor, PTEN acts as a phosphatase for PIP3 and negatively regulates the PI3K/AKT pathway. However, the connection between HES1 and downstream factors, p27 and PTEN in fetal hematopoietic tissues/system are not known. Using a fetal hematopoietic Hes1 specific knockout strain, here we show that loss of Hes1 affects fetal hematopoiesis possibly through de-repressing the two HES1 downstream factors, therefore subsequently suppressing HSC proliferation and increasing HSC death. Together, our studies reveal a novel role for HES1 in regulating hematopoiesis during embryonic development, and add another layer of understanding the distinct HES1 functions in modulating gene expression at different stages of development.
Materials and methods
Mice and treatment
Heterozygous Hes1fl/+ mice [4, 27] were interbred with Flt3Cre mice [28, 29] to generate Hes1fl/flFlt3Cre and Hes1fl/fl littermates. This Flt3Cre strain allows reliable deletion of Hes1 in multipotent progenitors (MPPs) at E10.5. 8 ∼ 12-week-old BoyJ mice were used as bone marrow transplant (BMT) recipients. Animals including BoyJ recipient mice were maintained in the animal barrier facility at Division of Laboratory Animal Resources (DLAR) at University of Pittsburgh. All animal experiments were carried out in accordance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee (IACUC) of University of Pittsburgh.
Fetal liver cells isolated from Hes1fl/flFlt3Cre or Hes1fl/fl embryo were cultured in stem cell culture medium (IMEM supplemented with 1% glutamine, 20% FBS, 50 ng/ml m-SCF, 20 ng/ml mTPO, and 1% Pen-Strep; [30]) in the presence or absence of 2 μM of p27 inhibitor, SJ403 (full name SJ572403; MedChemExpress; [31, 32]); or 100 μmol/L of PTEN inhibitor (bpV(HOpic) from Sigma-Aldrich; [33, 34]), for 24 h followed by flow cytometry analysis, CFU or bone marrow transplant (BMT).
Flow cytometry analysis
Single-cell suspensions were prepared from the fetal livers and cells were stained with saturating concentrations of Pacific Blue, FITC, PE, PE-Cy7, PerCP, APC, APC-Cy7 or e-Fluor780 conjugated antibodies in PBS containing 0.1% BSA and 0.05% NaN3 at 4 ºC for 30 min, then washed with PBS and subsequently analyzed using a BD Biosciences FACS Fortessa and FCS express software. For the analysis of hematopoietic stem and progenitor (HSPC) populations, 1 × 106 fetal liver cells were stained with a biotinylated antibody cocktail against lineage markers CD3, CD8a, CD19, B220, Gr1, CD11b, CD11c, Ter119 (BD Bioscience, San Diego, CA; Cat: # 559971) followed by streptavidin PerCP-Cy5.5 (BioLegend, San Diego, CA; Cat: # 405214) antibody staining. In addition to the lineage markers for the analysis of LSK (Lin−Sca1+c-kit+) and LK (Lin−c-kit+) populations, fetal liver cells were stained with antibodies against Pacific Blue Pacific Blue-Sca1 (BioLegend, San Diego, CA; Cat #: 108120; Clone: D7), PeCy7-CD150 (BioLegend, San Diego, CA; Cat # 115914; Clone: TC15-12F12.2), APC-CD48 (eBioscience, San Diego, CA; Cat #: 17-0481-82; Clone: HM48-1) and eFluor780-c-Kit (eBioscience, San Diego, CA; Cat #: 47-1171-82; Clone: 2B8). Fetal liver cells were stained with PeCy7-CD16/32 (eBioscience; Cat #: 25-0161-82; Clone: 93) and Alexa Fluor 647 anti-CD34 (BD Biosciences, San Diego, CA; Cat #: 560230; Clone: RAM34) monoclonal antibodies to obtain GMP, CMP and MEP populations and with PE-IL7Ra (eBioscience, San Diego, CA; Cat #: 12-1271-82; Clone: A7R34) and CD135 PECy7 (eBioscience, San Diego, CA; Cat #: 12-1351-82; Clone: A2F10) to obtain CLP cells. Cell sorting was performed on MoFlo machines (Dako Cytomation) or FACS AriaII (Becton Dickinson, San Jose, CA).
For cell cycle analysis, surface marker-stained cells were fixed and permeabilized using Cytofix/Cytoperm buffer (BD Pharmingen, 554722) followed by intensive wash using Perm/Wash Buffer (BD Pharmingen, 554723). Cells were then labeled with Hoechst 33342 and Pyronin Y staining buffer (10 mg/mL Hoechst 33342 and 150 ng/mL Pyronin Y in Perm/Wash buffer; MilliporeSigma, 92-32-0) at 37˚C for 1 h at room temperature for 30 min followed by flow cytometry analysis on CD45.2+ SLAM-gated population.
For BrdU incorporation assay, Bromodeoxyuridine (BrdU, 150 μl of 10 mg/ml) were intraperitoneally (i.p.) injected to the indicated pregnant mice followed by embryo isolation 14 h later. BrdU incorporated cells (S phase) were analyzed with the BrdU Flow Kit (BD Biosciences, San Jose, CA; Cat #: 556029; Clone: 3D4), following the manufacturer’s instructions. Briefly, cells were surface stained then fixed and permeabilized using BD Cytofix/Cytoperm Buffer. After 1 h incubation with DNase at 37 °C, cells were stained with anti-BrdU monoclonal antibody. DAPI was added to each sample right before Flow Cytometry analysis (BD Biosciences, San Jose, CA).
For apoptosis staining, surface marker-stained cells were incubated with Annexin V and 7AAD using the BD ApoAlert Annexin V Kit (BD Pharmingen, San Jose, CA; Cat #: 550475) in accordance with the manufacturer’s instruction. Flow cytometry analysis was then performed to determine the proportion of Annexin V-positive cells.
For donor-derived chimera analysis, peripheral blood (PB) from the recipient mice were subjected to staining using PE-anti-CD45.1 and APC-anti-CD45.2 (both from BD Biosciences; Cat #: 553776 & 558702; Clone: A20 & 104) antibodies followed by flow cytometry analysis.
Clonogenic assays
Single cell suspension from E12.5 fetal liver cells (1 × 105) were plated in a 35-mm tissue culture dish in 3 mL of MethoCult M3134 (Stem Cell Technologies, Vancouver, BC, Canada) supplemented with the following growth factors: 100 ng/ml SCF, 10 ng/ml IL-3, 100 ng/ml GM-CSF, and 4 units/mL erythropoietin (Peprotech, Burlington, NC). On day 7 after plating, erythroid and myeloid colonies were enumerated. For serial plating, cells from primary CFU assays were pooled and re-plated to evaluate secondary CFUs [35]. Hematopoietic clonal growth results were expressed as means (of triplicate plates) ± SD of three experiments.
Transplantations
8 ∼ 12-week-old BoyJ mice were used for all transplant experiments. For non-competitive BMT, 5 × 105 fetal liver cells were injected via tail vein to lethally irradiated (11.75 Gy) BoyJ recipients. For competitive BMT, 5 × 105 fetal liver cells, along with 5 × 105 congenic BoyJ WBMCs were injected via tail vein. For secondary transplantations 3 ∼ 5 × 106 BM cells from the primary recipients were injected to sublethally irradiated (7.0 Gy) BoyJ recipients.
Immunohistochemistry and apoptosis analysis
Fetal livers were fixed overnight in 4% buffered formalin. Four-micron sections were produced from paraffin embedded samples and immunohistochemistry was performed according to antibody specification. Proliferation cells were detected with anti-ki67 antibody. Apoptotic cells were detected by cleaved caspase 3 antibody.
Animal euthanasia and anesthesia
The method of euthanasia used in this study is CO2 narcosis before cervical dislocation. For all experiments described in this study, including tail biopsies, and tail bleeding, bleeding and pain is minimal, anesthesia is not required. This work has been reported in line with the ARRIVE guidelines 2.0.
Results
Loss of Hes1 affects fetal development
Previous study has confirmed Hes1 expression in mouse embryonic liver and spleen [36]. To study the role of HES1 in fetal hematopoiesis, we crossed the published Hes1 conditional knockout mice (Hes1fl/fl; [4, 27]) with Flt3Cre strain, in which robust Flt3Cre activity begins in multipotent progenitors (MPPs) at E10.5 and is subsequently observed in more than 90% of mature leukocytes [29, 37]. We consistently observed a slightly reduced litter size from Hes1fl/flFlt3Cre breeding compared to the control breeding (8.43 ± 1.5 vs. 6.43 ± 0.99; p = 0.0467), indicating that a small portion of embryos may have died during embryonic development. Due to the extremely low recombination activity of Flt3Cre in female mice [28, 37], Hes1fl/flFlt3Cre females were born far below the expected Mendelian ratios (data not shown). Therefore, we decided to mate Hes1fl/flFlt3Cre male with Hes1fl/fl female for embryonic analysis (Fig S1A). Since Flt3 is generally considered to be absent from the surface of HSCs in the adult mice [38,39,40], Hes1fl/flFlt3Cre male do not exhibit obvious phenotype (data not shown), which is consistent with our previous findings that HES1 is dispensable from adult hematopoiesis under steady state condition [4].
Embryo genotypes determined by PCR analysis definitively identified Hes1fl/fl (control) and Hes1fl/flFlt3Cre (knockout; KO) embryos (Fig S1B). Hes1 deletion in the fetal liver was confirmed by both immunohistochemistry (IHC) staining using fetal liver isolated on E13.5 (Fig S1C) and intracellular flow cytometry using fetal liver cells (Fig S1D). Gross examination of Hes1fl/flFlt3Cre mutant embryos revealed that the most obvious consequence of homozygous knockout of Hes1 gene was an impairment of embryonic development, evidenced by the reduction in the weight of Hes1fl/flFlt3Cre embryos compared to the control embryos (Fig. 1A). It should be noted that hemizygous embryos appeared normal, consistent with the fact that healthy live births of hemizygous Hes1fl/+Flt3Cre males were obtained. Dissection of mutant embryos revealed that Hes1 knockout results in smaller, anemic fetal livers, while the control fetal liver showed a bright red appearance (Fig. 1B, Left). Hes1fl/flFlt3Cre fetal livers weighted significantly less than the control fetal livers (Fig. 1B, Middle), even after normalizing to total embryo weight (Fig. 1B, Right). In addition, the total cellularity of Hes1fl/flFlt3Cre fetal liver was also slightly reduced (Fig. 1C). Together, these findings indicate that loss of Hes1 affects fetal liver development.
Loss of Hes1 reduces HSPC pool and alters hematopoietic lineage differentiation
We next performed flow cytometry analysis to examine different hematopoietic cell subpopulations in the mouse fetal livers. We found that Hes1 deficiency caused a reduction in the frequencies of hematopoietic stem and progenitor cells (HSPCs, Lin−Sca1+c-kit+; LSK). Importantly, this reduction was even more profound in the phenotypic hematopoietic stem cell (HSC, Lin−Sca1+c-kit+CD150+CD48−; Signaling lymphocyte activation molecules; SLAM; 41) compartment (Fig. 2A). We also measured the frequences of other progenitor cells in the fetal liver [42, 43], namely CLP (common lymphoid progenitor), CMP (common myeloid progenitor), MEP (Megakaryocyte/erythrocyte progenitor), and GMP (granulocyte/macrophage progenitor). Flow cytometry analysis revealed that loss of Hes1 resulted in increased CMP at the expense of CLP in Hes1fl/flFlt3Cre fetal liver (Fig. 2B). Among the myeloid progenitors, we found that the frequencies of CMP and GMP were significantly higher, whereas MEP frequency was notably lower in Hes1fl/flFlt3Cre fetal livers compared to those in the control fetal livers. These data suggest that deletion of Hes1 in fetal hematopoietic system alters HSPC pool.
Quiescence is an important feature of HSC homeostasis [44,45,46]. Next, we analyzed the cell cycle status of fetal HSCs. Hochest 333342/Pyronin staining revealed a moderate increase in the proportion of quiescent (G0) SLAM cells in Hes1fl/flFlt3Cre fetal liver compared to those in Hes1fl/fl control livers (Fig. 2C, Left). Consistently, percentage of proliferating SLAM cells was notably lower in Hes1fl/flFlt3Cre fetal liver than Hes1fl/fl fetal livers as shown by a well-established BrdU incorporation assay (Fig. 2C, Right). In addition, we observed a slightly increased spontaneous apoptosis in SLAM cells isolated from Hes1fl/flFlt3Cre fetal liver as analyzed by Annexin V/DAPI staining (Fig. 2D). These results suggest that HES1 may be required for fetal HSC homeostasis.
Since we observed anemic livers and increased MEP, indicative of a defective erythroid differentiation, in Hes1fl/flFlt3Cre fetal livers, we then measured erythroid differentiation using the previously described CD71/Ter119 staining protocol [47,48,49,50]. We found that loss of Hes1 led to an increase in S0 (CD71intermediateTer119low) and decreases in S3 (CD71highTer119high) and S4-5 (CD71intermediateTer119high; Fig. 2E), indicating a blockage of S0 to S1 transition and an erythroid maturation defect. These data suggest that HES1 is required for lineage commitment during embryonic development.
Loss of Hes1 impairs HSPC function
Decreased cycling and increased apoptosis of hematopoietic cells in Hes1fl/flFlt3Cre fetal liver may affect HSPC function [45]. By using an in vitro colony formation unit (CFU) assay [46, 51], we found that fetal liver cells from E12.5 Hes1fl/flFlt3Cre embryos produced significantly fewer colonies than those from Hes1fl/fl embryos when plated in methylcellulose supplemented with hematopoietic cytokines (Fig. 3A, Left). Significantly, Hes1fl/flFlt3Cre cells showed a marked decrease in serial replating activity compared to Hes1fl/fl cells, indicative of replicative exhaustion in the absence of stromal support (Fig. 3A, Right).
We then measured the in vivo hematopoietic repopulating capacity of Hes1fl/flFlt3Cre HSCs by a non-competitive bone marrow transplantation (BMT) assay (Fig. 3B), where 5 × 105 E14.5 fetal liver cells from Hes1fl/fl or Hes1fl/flFlt3Cre embryos (CD45.2+) were injected into lethally irradiated BoyJ recipients (CD45.1+). Although we observed a significant reduction in donor-derived chimera in Hes1fl/flFlt3Cre cell transplanted recipients, all the primary recipients survived 4-month observation period (Fig. 3C), albeit that the bones from the recipient mice transplanted with Hes1-KO fetal liver cells were paler than those transplanted with control cells (Fig S2A). Further analysis revealed that this reduction was not resulted from homing defects since Hes1-KO fetal liver cells gave rise to comparable CFSE+ cells in the bone marrow (BM) of the recipients 16 h post-transplant in a previously established homing assay (Fig S2B; [51, 52]). However, the secondary recipients transplanted with whole bone marrow cells (WBMCs) from Hes1fl/flFlt3Cre fetal liver cells transplanted primary recipients died significantly earlier than those transplanted with WBMCs from the mice transplanted with control Hes1fl/fl fetal liver cells (Fig. 3D), suggesting a premature HSC exhaustion.
To further substantiate the findings, we then performed competitive BMT (Fig. 3E) by transplanting 5 × 105 E14.5 fetal liver cells from Hes1fl/fl or Hes1fl/flFlt3Cre fetal livers (CD45.2+), along with 5 × 105 congenic WBMCs from the BoyJ mice into lethally irradiated BoyJ recipients (CD45.1+; 51). Flow cytometry analysis demonstrated a reduced donor-derived chimera (CD45.2+) in the peripheral blood (PB) of the recipients transplanted with Hes1fl/flFlt3Cre fetal liver cells compared to those transplanted with Hes1fl/fl cells (Fig. 3F). Hes1 deletion also altered lineage differentiation (Fig. 3G), as evidenced by the slightly increased myeloid cells and decreased T cells in the recipients transplanted with Hes1fl/flFlt3Cre fetal liver cells. Furthermore, complete blood count (CBC) revealed significant decreases in red blood cell (RBC) count, hemoglobin (Hb) and platelet count in the PB of Hes1fl/flFlt3Cre fetal liver cells transplanted recipient mice compared with Hes1fl/fl cells transplanted recipients (Fig S2C). Additionally, serial BMT confirmed a long-term repopulation defect of Hes1fl/flFlt3Cre HSCs in the secondary transplanted recipients (Fig. 3H). Surprisingly, different from that Hes1-deficient adult HSCs undergo exhaustion under replicative stress through augmented fatty acid oxidative (FAO; 4), fetal LSK cells deficient for Hes1 only exhibited marginally increased FAO (Fig S3A). Consistently, FAO related genes were scarcely upregulated (Fig S3B). Taken together, these data suggest that HES1 is required for fetal HSC maintenance, possibly through a distinct mechanism from their adult counterparts.
Depletion of Hes1 leads to decreased proliferation and increased apoptosis of fetal liver
Since Hes1-KO fetal livers appear to be smaller and anemic, we then performed histopathology analysis of fetal livers from Hes1fl/flFlt3Cre or Hes1fl/fl embryos. H&E-stained sagittal sections revealed fragmented pyknotic nuclei, which is the characteristic of cells undergoing apoptosis [53], in the fetal livers isolated from Hes1fl/flFlt3Cre embryos (Fig. 4A). Moreover, Hes1fl/flFlt3Cre fetal livers exhibited an increase in cells stained positive for cleaved caspase 3, a hallmark of both intrinsic and extrinsic apoptosis pathways ([54, 55]; Fig. 4B). These data support the notion that increased apoptosis contributes to the impaired development of Hes1fl/flFlt3Cre fetal livers.
To determine whether the decrease in cell proliferation also contributes to the reduced size of Hes1fl/flFlt3Cre fetal livers, we examined the level of cellular proliferation within the embryonic livers using the cell proliferation marker Ki67. Ki67 immunoreactivity of Hes1fl/fl fetal livers showed that almost all cells were proliferating, consistent with rapid growth of the fetal liver at this stage of development (Fig. 4C, Upper; 9). However, Ki67+ cells in Hes1fl/flFlt3Cre fetal livers were remarkably lower (Fig. 4C, Lower). Together, these results indicate that decreased fetal liver size in E13.5 Hes1fl/flFlt3Cre embryos may be due to, at least in part, increased cell death and decreased proliferation.
HES1 suppresses p27 and PTEN expression in the fetal liver
Previous studies have shown that HES1 directly controls cell proliferation through the transcriptional repression of p27Kip1 [24], a cyclin-dependent kinase (Cdk) inhibitor 1B that regulates cell proliferation, cell motility and apoptosis [56]. HES1 also opposes a PTEN-dependent check on survival, differentiation, and proliferation of TCRb-selected mouse thymocytes [25]. Since we observed reduced cell proliferation and increased cell death in Hes1-KO fetal liver, we then measured Pten and p27 levels in HSPCs isolated from Hes1fl/flFlt3Cre fetal liver. Indeed, we found that both mRNA and protein levels of PTEN and p27 were elevated in Hes1-deficient fetal HSPCs compared to those in the control HSPCs (Fig S4). Concomitantly, pharmacological inhibition of p27 by SJ572403 (SJ403; [31, 32]), or PTEN by bpV [33, 34], significantly improved HSC proliferation (Fig. 5A), and ameliorated apoptosis of mouse fetal HSPCs (Fig. 5B). Functionally, p27 and PTEN inhibition notably improved Hes1-KO progenitor activity in CFU assay (Fig. 5C and D), hematopoietic repopulating in the lethally irradiated recipient mice (Fig. 5E and F). Together, our data indicate that HES1 regulates fetal hematopoiesis through repressing p27 and PTEN, thereby promoting fetal cell proliferation and suppressing cell death.
Discussion
Fetal hematopoiesis exhibits distinct features from adult hematopoiesis [9, 15, 57]. Although previous studies have shown the role of HES1 in regulating adult hematopoiesis, how HES1 functions in fetal hematopoiesis remains to be elucidated. By using a novel fetal hematopoietic specific Hes1 knockout mouse model (Hes1fl/flFlt3Cre), the study presented here provides evidence that HES1 is a critical regulator of fetal hematopoiesis in a cell autonomous manner. There are several findings that highlight the significance of our study: (1) HES1 is indispensable for fetal hematopoiesis; (2) Hes1-deficient mouse fetal HSCs are more quiescent and exhibit increased spontaneous apoptosis; (3) Hes1 loss in fetal hematopoietic system affects lineage differentiation; (4) Deletion of Hes1 de-represses p27 and Pten and leads to elevated p27 and PTEN expression in fetal liver hematopoietic cells; (5) Pharmacological inhibition of p27 and PTEN improves Hes1-deficient fetal liver HSPC function.
It is known that fetal progenitors are highly proliferative, and have greater engraftment potential and higher propensity to form myeloid colonies compared with adult counterparts [16,17,18,19,20,21]. Here we show that HES1 plays distinct roles in fetal hematopoiesis and loss of Hes1 in embryonic hematopoietic progenitor cells alters fetal liver HSPC subpopulation and compromises their function (Figs. 2, 3 and 4). While dispensable for steady state adult hematopoiesis, HES1 is required for stress hematopoiesis through regulating fatty acid oxidation (FAO; 4). One of the potential explanations on these disparities is that fetal HSPCs cope with highly proliferative status of fetal organ development, thereby are consistently under replication stress. Moreover, inflammatory signals play critical roles in fetal HSPC emergence, specification, and maturation [58]. In fact, each stage of pregnancy is characterized by a unique inflammatory environment [59]. A recent study demonstrated that HES1 acts in concert with a Fanconi anemia protein, FANCD2 to suppress inflammation-induced PPARg to prevent HSC exhaustion [60]. Our current data add another layer to the understanding of how HES1 regulates hematopoiesis during embryonic development, suggesting a distinct role for HES1 in modulating HSC maintenance at different stages of development.
In addition to previously described T cell development defects, here we show that Flt3Cre-mediated Hes1 deletion impairs definitive hematopoiesis, particularly in erythroid lineage commitment (Fig. 2E and S2C). The haemopoietic composition of fetal liver during gestation shifts from being predominantly erythroid, accompanied by a parallel change in differentiation potential of HSC in human [61]. Similarly, three major waves of hematopoietic potential emerge sequentially during murine embryogenesis. First of all, primitive hematopoiesis emerges in the yolk sac and primitive erythroid cells subsequently mature in the bloodstream. Secondly, erythro-myeloid progenitors (EMP) emerge in the yolk sac then seed the fetal liver, where they rapidly differentiate and give rise to the first circulating definitive erythrocytes. These two waves of hematopoiesis also contain megakaryocyte and myeloid potential. Finally, adult repopulating HSCs emerge from major arteries and seed the fetal liver, and eventually the BM [62]. Mouse fetal liver between E11 and E15 is primarily an erythropoietic tissue [63]. CD71/Ter119 staining pattern in the fetal liver is dependent on embryonic age [48,49,50]. At the onset of definitive erythropoiesis on embryonic day 11 (E11) in the fetal liver, cells are concentrated in subsets S0 and S1, and are mostly erythroid colony-forming cells (CFU-e). With embryonic development, CFU-e cells differentiate into proerythroblasts (CD71highTer119intermediate) and maturing erythroblasts and gradually populate subsets S2 to S5 [47,48,49]. Our current studies demonstrate that Hes1 deficiency blocks fetal erythroid differentiation during S0 to S1 transition (Fig. 2E), which marks a switch from CFUe self-renewal to differentiation. Therefore, our findings lend support to the most recent study showing that another member of HES family member, HES6 regulates human erythropoiesis in a GATA1 and GATA2-independent manner [64, 65], and highlight a previously unappreciated role for HES family members in regulating erythropoiesis.
Another interesting finding of the present study is that loss of Hes1 in fetal liver dampens HSC cycling and increases spontaneous cell death. We did not observe significant differences in HSC quiescence and apoptosis in adult hematopoiesis in Hes1fl/flVav1Cre mice [4]. The current findings suggest divergent roles for HES1 in regulating fetal versus adult hematopoiesis, which might be due to the higher expression of HES1 in fetal liver than in adult tissues [4, 36]. Mechanistically, we show that HES1 promotes proliferation and suppresses cell death through repressing p27 and PTEN expression in fetal HSPCs. Cyclin-dependent kinase inhibitor 1B, p27 is known as a mediator of the G1-S phase block [66]. Indeed, BrdU incorporation analysis demonstrates a decreased proportion of cells in S phase and increased proportion of cells stuck in G1 phase in Hes1-KO fetal liver (Fig. 2C), associated with the increased p27 in Hes1fl/flFlt3Cre HSPCs (Fig S3). It has been shown that pharmacological inhibition of PTEN leads to PI3K/AKT/mTOR upregulation, and improves cell proliferation and migration [34, 67], thereby has been considered as a promising strategy for supporting tissue repair [68, 69]. In agreement with these findings, we show that PTEN inhibition significantly improves Hes1-deficient fetal HSPC function both in vitro and in vivo (Fig. 5). It is in this context, our study demonstrates that HES1 controls fetal hematopoiesis by two potential and synergistic cell autonomous mechanisms, subsequently modulates fetal HSPC viability and proliferation.
Hepatic and hematopoietic lineages develop simultaneously during the process of fetal liver hematopoiesis [9]. For example, macrophage-released oncostatin M (OSM) is crucial for hepatocyte differentiation during fetal hematopoietic development [70]. Similarly, megakaryocyte progenitors are known to promote hepatoepithelial cell development into hepatocytes in E11.5 fetal liver through a cell-to-cell contact manner [71]. Opposingly, hepatic stromal cells also affect fetal liver hematopoiesis [72]. In line with these previous reports, we observed fetal liver developmental defects in the newly established fetal hematopoietic specific Hes1-KO mice (Figs. 1 and 4). Although further analysis is needed, our present study highlights a bidirectional crosstalk between hepatocytes and hematopoietic cells during the embryonic development, emphasizing the role of embryonic hematopoietic cells in modulating fetal liver development.
Conclusions
Our study identifies a previously undescribed role for HES1 in regulating definitive fetal hematopoiesis, and provide mechanistic insight into the function of HES1 in fetal HSC maintenance.
Availability of supporting data
All data generated or analyzed during this study are included in this published article.
References
Kageyama R, Ohtsuka T, Kobayashi T. The hes gene family: repressors and oscillators that orchestrate embryogenesis. Development. 2007;134:1243–51.
Jarriault S, Brou C, Logeat F, Schroeter EH, Kopan R, Israel A. Signalling downstream of activated mammalian notch. Nature. 1995;377:355–8.
Nishimura M, Isaka F, Ishibashi M, Tomita K, Tsuda H, Nakanishi S, Kageyama R. Structure, chromosomal locus, and promoter of mouse Hes2 gene, a homologue of Drosophila hairy and enhancer of split. Genomics. 1998;49:69–75.
Ma Z, Xu J, Wu L, Wang J, Lin Q, Chowdhury FA, Mazumder MHH, Hu G, Li X, Du W. Hes1 deficiency causes hematopoietic stem cell exhaustion. Stem Cells. 2020;38(6):756–68.
Burns CE, Traver D, Mayhall E, Shepard JL, Zon LI. Hematopoietic stem cell fate is established by the Notch-Runx pathway. Genes Dev. 2005;19(19):2331–42.
Lee JS, Thomas DM, Gutierrez G, Carty SA, Yanagawa S, Hinds PW. HES1 cooperates with pRb to activate RUNX2-dependent transcription. J Bone Mineral Res. 2006;21(6):921–33.
Suh JH, Lee HW, Lee JW, Kim JB. Hes1 stimulates transcriptional activity of Runx2 by increasing protein stabilization during osteoblast differentiation. Biochem Biophys Res Commun. 2008;367(1):97–102.
Xu J, Fei P, Simon DW, Morowitz MJ, Mehta PA, Du W. Crosstalk between DNA damage repair and metabolic regulation in hematopoietic stem cells. Cells. 2024;13(9):733.
Lewis K, Yoshimoto M, Takebe T. Fetal liver hematopoiesis: from development to delivery. Stem Cell Res Ther. 2021;12(1):139.
Ciriza J, Thompson H, Petrosian R, Manilay JO, Garcia-Ojeda ME. The migration of hematopoietic progenitors from the fetal liver to the fetal bone marrow: lessons learned and possible clinical applications. Exp Hematol. 2013;41:411–23.
Zhou Y, Lewallen M, Xie T. Stem cells’ exodus: a journey to immortality. Dev Cell. 2013;24:113–4.
Coskun S, Chao H, Vasavada H, et al. Development of the fetal bone marrow niche and regulation of HSC quiescence and homing ability by emerging osteolineage cells. Cell Rep. 2014;9:581–90.
Zanjani E, Ascensao J, Tavassoli M. Liver-derived fetal hematopoietic stem cells selectively and preferentially home to the fetal bone marrow. Blood. 1993;81:399–404.
Ara T, Tokoyoda K, Sugiyama T, Egawa T, Kawabata K, Nagasawa T. Long-term hematopoietic stem cells require stromal cell-derived factor-1 for colonizing bone marrow during ontogeny. Immunity. 2003;19:257–67.
Liu Y, Chen Q, Jeong HW, Koh BI, Watson EC, Xu C, Stehling M, Zhou B, Adams RH. A specialized bone marrow microenvironment for fetal haematopoiesis. Nat Comm. 2022;13(1):1327.
Copley MR, Babovic S, Benz C, Knapp DJ, Beer PA, Kent DG, Wohrer S, Treloar DQ, Day C, Rowe K, Mader H, Kuchenbauer F, Humphries RK, Eaves CJ. The Lin28b-let-7-Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells. Nat Cell Biol. 2013;15(8):916–25.
Yuan J, Nguyen CK, Liu X, Kanellopoulou C, Muljo SA. Lin28b reprograms adult bone marrow hematopoietic progenitors to mediate fetal-like lymphopoiesis. Science. 2012;335(6073):1195–200.
Porter SN, Cluster AS, Yang W, Busken KA, Patel RM, Ryoo J, Magee JA. Fetal and neonatal hematopoietic progenitors are functionally and transcriptionally resistant to Flt3-ITD mutations. Elife. 2016;5:e18882.
Bowie MB, McKnight KD, Kent DG, McCaffrey L, Hoodless PA, Eaves CJ. Hematopoietic stem cells proliferate until after birth and show a reversible phase-specific engraftment defect. J Clin Invest. 2006;116(10):2808–16.
Lansdorp PM, Dragowska W, Mayani H. Ontogeny-related changes in proliferative potential of human hematopoietic cells. J Exp Med. 1993;178(3):787–91.
Broxmeyer HE, Hangoc G, Cooper S, Ribeiro RC, Graves V, Yoder M, Wagner J, Vadhan-Raj S, Benninger L, Rubinstein P. Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults. Proc Natl Acad Sci USA. 1992;89(9):4109–13.
Tomita K, Hattori M, Nakamura E, Nakanishi S, Minato N, Kageyama R. The bHLH gene Hes1 is essential for expansion of early T cell precursors. Genes Dev. 1999;13:1203–10.
Guiu J, Shimuzu R, D’Altri T, Fraser ST, Hatakeyama J, Brensnick EH, et al. Hes repressors are essential regulators of hematopoietic stem cell development downstream of notch signaling. J Exp Med. 2013;210(1):71–84.
Murata K, Hattori M, Hirai N, Shinozuka Y, Hirata H, Kageyama R, Sakai T, Minato N. Hes1 directly controls cell proliferation through the transcriptional repression of p27Kip1. Mol Cell Biol. 2005;25(10):4262–71.
Wong GW, Knowles GC, Mak TW, Ferrando AA, Zuniga-Pflucker JC. HES1 opposes a PTEN-dependent check on survival, differentiation, and proliferation of TCRβ-selected mouse thymocytes. Blood. 2017;120(7):1439–48.
Shao J, Li S, Palmqvist L, Fogelstrand L, Wei SY, Busayavalasa K, Liu K, Liu VM. p27KIP1 and PTEN cooperate in myeloproliferative neoplasm tumor suppression in mice. Exp Hematol Onco. 2016;5:17.
Imayoshi I, Shimogori T, Ohtsuka T, Kageyama R. Hes genes and neurogenin regulate non-neural versus neural fate specification in the dorsal telencephalic midline. Development. 2008;135(15):2531–41.
Benz C, Martins VC, Radtke F, Bleul CC. The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development. J Exp Med. 2008;205(5):1187–99.
Tarnawsky SP, Kobayashi M, Chan RJ, Yoder MC. Mice expressing KrasG12D in hematopoietic multipotent progenitor cells develop neonatal myeloid leukemia. J Clin Invest. 2017;127(10):3652–6.
Li X, Chatla S, Wilson AF, Wu L, Atale N, Du W. Persistent DNA damage and oncogenic stress-induced Trem1 promotes leukemia in mice. Haematologica. 2022;107(11):2576–88.
Iconaru LI, Ban D, Bharatham K, Ramanathan A, Zhang W, Shelat AA, Zuo J, Kriwacki RW. Discovery of small molecules that inhibit the disordered protein, p27Kip1. Sci Rep. 2015;5:15686.
Zhou X, Li TM, Luo JZ, et al. CYP2C8 suppress proliferation, migration, invasion and sorafenib resistance of hepatocellular carcinoma via PI3K/Akt/p27kip1 axis. J Hepatocell Carcinoma. 2021;8:1323–38.
Chauhan A, Kumar Sah D, Kumari N, Kalra N, Soni R, Bhatt AN. PTEN inhibitor bpV(HOpic) confers protection against ionizing radiation. Sci Rep. 2021;11:1720.
Borges GA, Webber LP, Marques AE, Guerra ENS, Castilho RM. Pharmacological PTEN inhibition: potential clinical applications and effects in tissue regeneration. Regen Med. 2020;15(2):1329–44.
Karamitros D, Patmanidi AL, Kotantaki P, Potocnik AJ, Bähr-Ivacevic T, Benes V, Lygerou Z, Kioussis D, Taraviras S. Geminin deletion increases the number of fetal hematopoietic stem cells by affecting the expression of key transcription factors. Development. 2015;142(1):70–81.
Sasai Y, Kageyama R, Tagawa Y, Shigemoto R, Nakanishi S. Two mammalian helix-loop-helix factors structurally related to Drosophila hairy and enhancer of split. Genes Dev. 1992;6:2620–34.
Mooney CJ, Cunningham A, Tsapogas P, Toellner KM, Brown G. Selective expression of Flt3 within the mouse hematopoietic stem cell compartment. Int J Mol Sci. 2017;18(5):1037.
Adolfsson J, Borge OJ, Bryder D, Theilgaard-Mönch K, Astrand-Grundström I, Sitnicka E, Sasaki Y, Jacobsen SE. Upregulation of flt3 expression within the bone marrow Lin–Sca1+c-kit+ stem cell compartment is accompanied by loss of self-renewal capacity. Immunity. 2001;15:659–69.
Christensen JL, Weissman IL. Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells. Proc Natl Acad Sci USA. 2001;98:14541–6.
Sitnicka E, Bryder D, Theilgaard-Mönch K, Buza-Vidas N, Adolfsson J, Jacobsen SE. Key role of Flt3 ligand in regulation of the common lymphoid progenitor but not in maintenance of the hematopoietic stem cell pool. Immunity. 2002;17:463–72.
Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121(7):1109–21.
Niizuma H, Searleman AC, Takeda S, Armstrong SA, Park CY, Cheng EH, Hsieh JJ. Taspase1 orchestrates fetal liver hematopoietic stem cell and vertebrae fates by cleaving TFIIA. JCI Insight. 2021;6(15):e149382.
Kayvanjoo AH, Splichalova I, Bejarano DA, Huang H, Mauel K, Makdissi N, et al. Fetal liver macrophages contribute to the hematopoietic. eLife. 2024;13:e86493.
Kobayashi M, Srour EF. Regulation of murine hematopoietic stem cells quiescence by Dmtf1. Blood. 2011;118(25):6562–71.
Lin Q, Wu L, Chatla S, Chowdhury FA, Atale N, Joseph J, Du W. Hematopoietic stem cell regeneration through paracrine regulation of the Wnt5a/Prox1 signaling axis. J Clin Invest. 2022;132(12):e155914.
Wu L, Lin Q, Chatla S, Amarachintha S, Wilson AF, Atale N, Gao ZJ, Jesph J, Wolff EV, Du W. LepR + niche cell-derived AREG compromises hematopoietic stem cell maintenance under conditions of DNA repair deficiency and aging. Blood. 2023;142(18):1529–42.
Raman R, Kumar RS, Hinge A, Kumar S, Nayak R, Szczur K, Cancelas JA, Filippi MD. p190-B RhoGAP regulates the functional composition of the mesenchymal microenvironment. Leukemia. 2013;27(11):2209–19.
Zhang J, Socolovsky M, Gross AW, Lodish HF. Role of Ras signaling in erythroid differentiation of mouse fetal liver cells: functional analysis by a flow cytometry based novel culture system. Blood. 2003;102:3938–46.
Koulnis M, Pop R, Porpiglia E, Shearstne JR, Hidalgo D. Socolovsky. Identification and analysis of mouse erythroid progenitors using the CD71/TER119 Flow-cytometric assay. J Vis Exp. 2011;54:2809.
Socolovsky M. Negative autoregulation by FAS mediates robust fetal erythropoiesis. PLoS Biol. 2007;5:e252–252.
Du W, Amarachintha S, Erden O, Wilson A, Meetei AR, Andreasson PR, Namekawa SH, Pang Q. Fancb deficiency impairs hematopoietic stem cell function. Sci Rep. 2015;5:18127.
Celso CL, Fleing HE, Wu JW, et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457:92–6.
Lee JA, Wang Z, Sambo D, Bunting KD, Pallas DC. Global loss of leucin carboxy;l methyltransferase-1 causes severe defects in fetal liver hematopoiesis. J Biol Chem. 2018;293(25):9636–50.
Beck L, Leroy C, Beck-Cormier S, Forand A, Salaun C, Paris N, et al. The phosphotate transporter PiT1 (Slc20a1) revealed as a new essential gene for mouse liver development. PLoS ONE. 2010;5(2):e9148.
Wang L, Yu H, Cheng H, He K, Fang Z, Ge L, Cheng T, Jin Y. Deletion of Stk40 impairs definitive erythropoiesis in the mouse fetal liver. Cell Death Dis. 2017;8(3):e2772.
Chu IM, Hengst L, Slingerland JM. The cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat Rev Cancer. 2008;8(4):253–67.
Lopez DA, Apostol AC, Lebish EJ, Valencia CH, Romero-Mulero MC, Pavlovich PV, Hernandez GE, Forsberg EC, Cabezas-Wallscheid N, Beaudin AE. Prenatal inflammation perturbs murine fetal hematopoietic development and causes persistent changes to postnatal immunity. Cell Rep. 2022;41(8):111677.
Collins A, Mitchell CA, Passegue E. Inflammatory signaling regulates hematopoietic stem and progenitor cell development and homeostasis. J Exp Med. 2021;218(7):e20201545.
Mor G, Cardenas I, Abrahams V, Guller S. Inflammation and pregnancy: the role of the immune system at the implantation site. Ann NY Acad Sci. 2011;1221(1):80–7.
Wu L, Li X, Lin Q, Chowdhury F, Mazumder MH, Du W. FANCD2 and HES1 suppress inflammation-induced PPARɣ to prevent haematopoietic stem cell exhaustion. Br J Haematol. 2021;192(3):652–63.
Popescu DM, Botting RA, Stephenson E, et al. Decoding human fetal liver haematopoiesis. Nature. 2019;574(7778):365–71.
Frame JM, McGrath KE, Palis J, Erythro-Myeloid, Progenitors. Definitive hematopoiesis in the conceptus prior to the emergence of hematopoietic stem cells. Blood Cells Mol Dis. 2013;51(4):220–5.
Pop R, Shearstone JR, Shen Q, Liu Y, Hallstrom K, Koulnis M, Gribnau J, Socolovsky M. A key commitment step in erythropoiesis is synchronized with the cell cycle clock through mutual inhibition between PU.1 and S-Phase progression. PLoS Biol. 2010;8(9):e1000484.
De Vos T, Oatman N, Boehme L, Putteman T, Velghe I, Van Droogenbroeck Y et al. HES6 knockdown in human hematopoietic precursor cells reduces their in vivo engraftment potential and their capacity to differentiate into erythroid cells, B cells, T cells and plasmacytoid dendritic cells. Haematologica. 2024. Apr 4.
Xu J, Du W. HES6: an emerging factor for human hematopoiesis. Haematologica. 2024. May 9.
Wang QM, Jones JB, Studzinski GP. Cyclin-dependent kinase inhibitor p27 as a mediator of the G1-S phase block induced by 1,25-dihydroxyvitamin D3 in HL60 cells. Cancer Res. 1996;56(2):264–7.
Pulido R. PTEN inhibition in human disease therapy. Molecules. 2018;23(2):285.
Zagni C, Almeida LO, Balan T, Martins MT, Rosselli-Murai LK, Papagerakis P, Castillo RM, Squarize CH. PTEN mediates activation of core clock protein BMAL1 and accumulation of epidermal stem cells. Stem Cell Rep. 2017;9(1):304–14.
Squarize CH, Castilho RM, Bugge TH, Gutkind JS. Accelerated wound healing by mTOR activation in genetically defined mouse models. PLoS ONE. 2010;5(5):e10643.
Kamiya A, Kinoshita T, Ito Y, Matsui Y, Senba E, Nakashima K, Taga T, Yoshida K, Kishimoto T, Miyajima A. Fetal liver development requires a paracrine action of oncostatin M through the gp130 signal transducer. EMBO J. 1999;18(8):2127–36.
Serrano N, Cortegano I, Ruiz C, Alia M, de Andres B, Rejas MT, Marcos MA, Gaspar ML. Megakaryocytes promote hepatoepithelial liver cell development in E11.5 mouse embryos by cell-to-cell contact and by vascular endothelial growth factor A signaling. Hepatology. 2012;56(5):1934–45.
Solanki A, Lau C-I, Saldaña JI, Ross S, Crompton T. The transcription factor Gli3 promotes B cell development in fetal liver through repression of shh. J Exp Med. 2017;214(7):2041–58.
Acknowledgements
We thank Drs. Ryoichiro Kageyama (Kyoto University) for Hes1fl/fl mice and Paul Love (NICHD/NIH) for Flt3Cre mice. This work is supported by an NIH/National Heart, Lung, and Blood Institute grant (R01HL151390 to W.D.), and a Children’s Leukemia Research Association (CLRA) Research Grant (to W.D.). This project used the Hillman Animal Facilit supported in part by NIH/National Cancer Institute grant (P30CA047904).
Author information
Authors and Affiliations
Contributions
A.Z.Z. performed the research and analyzed the data; Z.M., E.V.W., Z.L., Z.J.G. and X. L. performed some of the research, assist data analysis. W.D. designed the research, analyzed the data, and wrote the paper.
Corresponding author
Ethics declarations
Ethical approval and consent to participate
No human data is used. Title of the approved project: “Role of FA proteins in hematopoiesis”. Name of the institutional approval committee or unit: Institutional Animal Care and Use Committee, University of Pittsburgh (Approval number: #24044702; Date of approval: 4/1/2024).
Consent for publication
All authors confirm their consent for publication.
Conflict of interest
The authors declare no conflicts of interest.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
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.
About this article
Cite this article
Zhu, A.Z., Ma, Z., Wolff, E.V. et al. HES1 is required for mouse fetal hematopoiesis. Stem Cell Res Ther 15, 235 (2024). https://doi.org/10.1186/s13287-024-03836-8
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s13287-024-03836-8