Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Non-Lethal Ionizing Radiation Promotes Aging-Like Phenotypic Changes of Human Hematopoietic Stem and Progenitor Cells in Humanized Mice

  • Changshan Wang,

    Affiliations Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan, College of Life Sciences, Inner Mongolia University, Hohhot 010021, China

  • Motohiko Oshima,

    Affiliation Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan

  • Goro Sashida,

    Affiliations Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan, International Research Center for Medical Sciences, Kumamoto University, Kumamoto City 860–0811, Japan

  • Takahisa Tomioka,

    Affiliation Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan

  • Nagisa Hasegawa,

    Affiliations Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan, Department of Hematology, Chiba University Hospital, Chiba 260–8670, Japan

  • Makiko Mochizuki-Kashio,

    Affiliation Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan

  • Yaeko Nakajima-Takagi,

    Affiliation Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan

  • Yoichiro Kusunoki,

    Affiliation Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732–0815, Japan

  • Seishi Kyoizumi,

    Affiliation Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732–0815, Japan

  • Kazue Imai,

    Affiliation Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732–0815, Japan

  • Kei Nakachi,

    Affiliation Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732–0815, Japan

  • Atsushi Iwama

    aiwama@faculty.chiba-u.jp

    Affiliation Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba 260–8670, Japan

Abstract

Precise understanding of radiation effects is critical to develop new modalities for the prevention and treatment of radiation-induced damage. We previously reported that non-lethal doses of X-ray irradiation induce DNA damage in human hematopoietic stem and progenitor cells (HSPCs) reconstituted in NOD/Shi-scid IL2rγnull (NOG) immunodeficient mice and severely compromise their repopulating capacity. In this study, we analyzed in detail the functional changes in human HSPCs in NOG mice following non-lethal radiation. We transplanted cord blood CD34+ HSPCs into NOG mice. At 12 weeks post-transplantation, the recipients were irradiated with 0, 0.5, or 1.0 Gy. At 2 weeks post-irradiation, human CD34+ HSPCs recovered from the primary recipient mice were transplanted into secondary recipients. CD34+ HSPCs from irradiated mice showed severely impaired reconstitution capacity in the secondary recipient mice. Of interest, non-lethal radiation compromised contribution of HSPCs to the peripheral blood cells, particularly to CD19+ B lymphocytes, which resulted in myeloid-biased repopulation. Co-culture of limiting numbers of CD34+ HSPCs with stromal cells revealed that the frequency of B cell-producing CD34+ HSPCs at 2 weeks post-irradiation was reduced more than 10-fold. Furthermore, the key B-cell regulator genes such as IL-7R and EBF1 were downregulated in HSPCs upon 0.5 Gy irradiation. Given that compromised repopulating capacity and myeloid-biased differentiation are representative phenotypes of aged HSCs, our findings indicate that non-lethal ionizing radiation is one of the critical external stresses that promote aging of human HSPCs in the bone marrow niche.

Introduction

Human hematopoiesis is a critical physiological regenerative process that provisions mature blood cell lineages. All lineages of cells in the blood system are derived from hematopoietic stem cells (HSCs), which are capable of both self-renewal and multi-lineage differentiation. The self-renewal capacity is an essential feature of HSCs, generating at least one daughter cell that has stem-cell properties similar to the parental cell [1], [2]. Stem-cell exhaustion caused by impaired self-renewal capacity of stem cells determines the aging phenotypes of tissues and organs and thus has been recognized as one of the hallmarks of mammalian aging [3]. It also underlies certain degenerative diseases [4]. HSCs show several characteristic phenotypes with age, such as impaired homing and repopulating capacity and biased differentiation toward myeloid lineage at the expense of their lymphoid potential [511]. However, aging phenotypes have not been fully evaluated for human HSCs in vivo.

Genomic stability is extremely important for the normal physiology of hematopoietic system and for preventing hematopoietic failure. Ionizing radiation that induces DNA damage is a major risk factor for genomic instability. Accumulation of DNA damage and loss of DNA repair capacity are generally associated with apoptosis, cellular senescence, cancer and stem cell abnormalities [12]. Of note, accumulation of DNA damage in HSCs has been invoked to explain an age-related decline in stem-cell function [1317]. One mechanism through which ionizing radiation affects the human hematopoietic system, especially HSCs, is through inducing DNA damage in vivo; however, non-lethal radiation effects on human HSCs have not been well characterized in vivo due to lack of suitable animal models. Therefore, effects of ionizing radiation have thus far been evaluated mainly in mice or by irradiating human HSCs ex vivo [18]. In order to understand non-lethal radiation effects on human hematopoietic stem and progenitor cells (HSPCs) in vivo, we previously analyzed NOD/Shi-scid IL-2rγnull (NOG) immunodeficient mice reconstituted with human HSCs and preliminarily evaluated the effects of 0.5 and 1.0 Gy of total-body irradiation (TBI) on human HSCs [19], in which we observed that DNA damage inflicted by ionizing radiation restricts the self-renewal capacity of human HSPCs in vivo. This humanized mouse model is useful for the evaluation of radiation effects on human hematopoiesis, including DNA damage, particularly in the dose region where long-term effects on the hematopoietic system as well as other organs have been observed among atomic-bomb survivors [20].

In this study, we further evaluated the functional changes in human HSPCs in NOG mice and the effects of non-lethal radiation in this process. Our findings demonstrate that non-lethal doses of ionizing irradiation profoundly affect the function of HSPCs in humanized mice and promote part of the aging-related phenotypes of human HSPCs in humanized mice, including compromised repopulating capacity and myeloid-biased differentiation at the expense of B lymphopoiesis.

Materials and Methods

Ethics Statement

Experiments using mice were performed in accordance with institutional guidelines of the Graduate School of Medicine, Chiba University. This study was approved by the Institutional Review Committees of Chiba University (approval numbers 24–64 and 26–131).

Cord blood cells and transplantation

Human cord blood (hCB) was kindly provided by the Japanese Red Cross Kanto-Koshinetsu Cord Blood Bank (Tatsumi, Tokyo, Japan). CD34+ cells were immunomagnetically enriched using a magnetic-activated cell-sorting CD34 progenitor kit (Miltenyi Biotech, Auburn, CA, USA) and were kept frozen. The purity of hCB CD34+ cells was approximately 95%. Frozen stock of the CD34+ cells was thawed and plated at 5 x 105 cells/well in a 6-well plate pre-coated with 25 μg/mL fibronectin fragment CH-296 (Takara Shuzo, Otsu, Shiga, Japan) and cultured in a serum-free medium, StemSpan SFEM (Stem Cell Technologies, Vancouver, BC, Canada) in the presence of 100 ng/mL human stem cell factor (SCF) (PeproTech, Rocky Hill, NJ, USA) and 50 ng/mL human thrombopoietin (TPO) (PeproTech) for 16 hours. Subsequently, 5 x 104 CD34+ cells were injected intravenously into 8-week-old NOG mice which had been irradiated with 2.0 Gy of X-rays 2 hours before. At 2 weeks post-irradiation, human CD34+ HSPCs were purified from the bone marrow (BM) of primary recipient mice and 2 x 106 CD34+ HSPCs were transplanted into secondary NOG recipients which had been irradiated with 2.0 Gy of X-rays 2 hours before. NOG immunodeficient mice [21] were supplied by the Central Institute for Experimental Animals (Kawasaki, Kanagawa, Japan) and maintained in the Animal Research Facility of the Graduate School of Medicine, Chiba University, in accordance with institutional guidelines.

Irradiation

Mice were irradiated on a rotating platform of the Hitachi MBR1520R X-ray machine (250 kV; 15 mA; dose-rate: 1.5 Gy/min; 0.5 mm aluminum and 0.1 mm copper filters) (Hitachi, Tokyo, Japan). Irradiation doses were verified by performing dosimetry using TLD100 dosimeters provided by the University of Wisconsin-Madison Radiation Calibration Laboratory (Madison, WI, USA). For 4 weeks after irradiation, mice were fed with drinking water containing 0.17 mg/ml enrofloxacin (Bayer, Shawnee Mission, KS, USA).

Flow cytometry

Human cells in the BM and PB of recipient mice were stained with Lineage mixture (biotinylated anti-human CD14, CD3, CD56, CD19 and CD235a), PE-Cy7-conjugated anti-human CD34, APC-conjugated anti-human CD45, PE-conjugated anti-human CD38, biotinylated anti-human CD7, biotinylated anti-human CD10, PE-conjugated anti-human CD62L, APC-Cy7-conjugated anti-human CD3, PE-conjugated anti-human CD11b, or PE-Cy7-conjugated anti-human CD19 (BD Biosciences, San Jose, CA, USA). They were then analyzed on a FACS Canto II or sorted on a FACS Aria II (BD Biosciences).

Colony assay and differentiation of B cells in culture

Human CD34+ cells were plated in Methocult GF H4435 methylcellulose medium containing 50 ng/mL human SCF, 10 ng/mL human granulocyte-macrophage colony-stimulating factor (GM-CSF), 10 ng/mL human IL-3, and 3 U/mL human EPO (StemCell Technologies). After 12 to 14 days of culture, the colonies were counted under a microscope. For the detection of B cells, sorted cells were cultured for 4 weeks with TSt-4 stromal cells [22] in 48-well plates. The frequencies of B cell commitment were determined by limiting dilution assays and calculated with L-Calc software (STEMCELL Technologies, Vancouver, BC, Canada). All culture experiments were performed with RPMI 1640 medium supplemented with 10% fetal calf serum, 2 mM L-glutamine, 100 U/ml penicillin, 100 μg/ml streptomycin, 50 μM 2-β-mercaptoethanol, 1 mM sodium pyruvate, 0.1 mM non-essential amino acid solution (Invitrogen, Carlsbad, CA, USA), 20 ng/ml stem cell factor (SCF), 20 ng/ml Flt3 ligand (Flt3L), and 20 ng/ml IL-7 (Peprotech).

Quantitative real-time PCR analysis

Total RNA was isolated using TRIZOL LS solution (Invitrogen) and reverse transcribed by the ThermoScript RT-PCR system (Invitrogen) with an oligo-dT primer. Real-time quantitative PCR was performed with an ABI prism 7300 Thermal Cycler (Applied Biosystems, Foster City, CA) using FastStart Universal Probe Master (Roche Diagnostics, Branchburg, NJ, USA). GAPDH expression was used to calculate relative expression levels. Probe numbers and primer sequences were: Probe #65, 5’-AGGTGGGTAGAGGGTCTGC-3’ and 5’-TCCAATTCCCCTGCAAACT-3’ for p16INK4A; probe #66, 5’-CACCGCTTTTTCCGAGTG-3’ and CCCCATCGATCCCAATAA-3’ for HOXA9, probe 82, 5’-GAGAGGGAAGAGGGGGTGT-37 and 5’-CGCACACAGCTCATACCAAC for MEIS1, probe #9, 5’-AGGCACTTTACCTCCACGAG-3’ and GCTTTTGAGGACCCAGATGT-3’ for IL-7R, probe #25, 5’-AGGAGCTGCTGCAGTTGG-3’ and 5’-GCTGCCAACTCCCCCTAT-3’ for EBF1, probe #83, 5’-CGGCAGCGCTATAATAGTAGG-3’ and 5’-GGAGTGAGTTTTCCGGGAGT-3’ for PAX5 and probe #25, 5’-CTGACTTCAACAGCGACACC-3’ and 5’-TAGCCAAATTCGTTGTCATACC-3’ for GAPDH.

Statistical analysis

P-values for trend were calculated by nonparametric Jonckheere-Terpstra test. The frequencies of B cell-producing cells in non-irradiated and irradiated cells were compared by the χ2-test for a 2 x 2 contingency table. The t-test was used for comparison of mean values. Statistical analysis was performed using SPSS Statics (version 20, IBM Corp., Chicago, IL, USA).

Results

Experimental design to investigate radiation effects on human HSPCs in vivo

We previously reported that 0.5 or 1.0 Gy of X-ray TBI induced prolonged DNA damage in human HSPCs reconstituted in NOG mice and severely compromised their repopulating capacity [19]. To more deeply understand the effects of low-dose irradiation on human HSPCs, we analyzed the functional changes in human HSPCs in NOG mice following non-lethal irradiation in detail (Fig 1). We prepared a new cohort of humanized mice for this study. We transplanted cord blood CD34+ HSPC into NOG mice irradiated with 2.0 Gy. At 12 weeks after the primary transplantation, the NOG mice showed considerably high chimerism, i.e., the median percentage of human CD45+ hematopoietic cells in the peripheral blood (PB) was 57.2% (range: 32.0 to 80.6%, n = 42). The mice were randomly grouped into three categories and irradiated with 0, 0.5 or 1.0 Gy for each group (14 mice each), and the radiation effects on human HSPC in vivo were assessed (Fig 1).

thumbnail
Fig 1. Experimental design to detect the non-lethal radiation effects on human HSPCs in vivo.

Schema of experimental strategy. Cord blood CD34+ HSPCs (5 x 104) were transplanted via tail veins into 8-week-old NOG mice irradiated with 2.0 Gy (n = 42). In the twelfth week after transplantation, the mice were randomly grouped into three categories and irradiated with 0, 0.5 or 1.0 Gy for each group (14 mice each). At 2 weeks post-irradiation, part of the mice (9 mice each) were sacrificed to evaluate early effects of irradiation. At the same time, human CD34+ HSPCs were purified from the primary recipient mice (9 mice were pooled), and 2 x 106 CD34+ HSPCs were transplanted into the secondary NOG recipients. At 12 weeks post-secondary transplantation, mice were sacrificed to evaluate long-lasting effects of irradiation. The remaining primary recipient mice (5 mice each) were similarly analyzed at 12 weeks post-irradiation to evaluate long-lasting effects of irradiation.

https://doi.org/10.1371/journal.pone.0132041.g001

Early effects of radiation on human HSPCs in humanized mice

At 2 weeks post-irradiation, mice were sacrificed to evaluate early effects of radiation (9 mice each). In contrast to NOD/Rag2null IL2rγnull mice, hematopoietic cells of NOG mice are susceptible to irradiation due to the scid/scid background [23]. As expected, chimerism of mouse CD45+ hematopoietic cells declined in a radiation dose-dependent manner in the PB while that of human hematopoietic cells increased (Fig 2A). Among human cells in the PB, chimerism of B cells significantly decreased in a radiation dose-dependent manner while that of myeloid and T cells increased (Fig 2B). Of interest, chimerism of human CD45+ hematopoietic cells declined in a radiation dose-dependent manner in the BM while that of mouse hematopoietic cells increased probably because of inappropriate support of human HSPCs by mouse BM niche cells (Fig 2C). As a consequence, human cells were gradually outcompeted by mouse cells in the PB afterwards as we have reported previously (Figs 3 and 4) [19]. In the BM, radiation dose-dependent decreases in both proportion (data not shown) and absolute numbers were commonly observed in human CD34+CD38- HSCs, CD34+CD38+ HPCs, CD34+CD38+CD45RA-CD10-CD7-Lineage marker- common myeloid progenitors (CMPs) and CD34+CD38+CD45RA+CD62LhiLineage marker- lymphoid-primed multipotent progenitors (LMPPs) (Fig 2D). In addition, CD34+ HSPCs recovered from recipient mice at 2 weeks post-irradiation gave rise to fewer colonies compared with non-irradiated HSPCs (0 Gy vs. 0.5 Gy, P = 0.015 and 0 Gy vs. 1.0 Gy P = 0.003 by Dunnett t-test) (Fig 2E).

thumbnail
Fig 2. Early effects of irradiation on human HSPCs in humanized mice.

(A) Changes in chimerism of human hematopoietic cells in the PB after irradiation. At 12 weeks post-irradiation, the mice were randomly grouped into three categories and irradiated with 0, 0.5 or 1.0 Gy for each group. At 2 weeks post-irradiation, mice were sacrificed to evaluate early effects of irradiation. White blood cell (WBC) counts in the PB (left panel) and chimerism of CD45+ human (middle panel) and mouse (right panel) hematopoietic cells in the PB are presented as scatter diagrams with median values (bars) (n = 9 each). PB data of NOG mice just before the irradiation (Pre) are presented as controls (n = 27). (B) The percentages of CD11b+ myeloid cells, CD19+ B cells, and CD3+ T cells in CD45+ human hematopoietic cells in the PB of NOG mice just before the irradiation (Pre, n = 27) and at 2 weeks after irradiation (0–1.0 Gy, n = 9 each). (C) Total BM cell numbers (left panel) and chimerism of CD45+ human (middle) and mouse (right) hematopoietic cells in the BM just before the irradiation (Pre, n = 6) and at 2 weeks after irradiation (0–1.0 Gy, n = 9 each). (D) Absolute numbers of CD34+CD38- HSCs, CD34+CD38+ HPCs, CMPs and LMPPs in the BM (bilateral femurs and tibiae) just before the irradiation (Pre, n = 6) and 2 weeks after irradiation (0–1.0 Gy, n = 9 each). (E) Numbers of colony-forming cells included in 1,000 CD34+ human HSPCs recovered at 2 weeks after irradiation (0–1.0 Gy, n = 3 each). Bars in scatter diagrams indicate median values. P-values for trend were calculated by nonparametric Jonckheere-Terpstra test.

https://doi.org/10.1371/journal.pone.0132041.g002

thumbnail
Fig 3. Repopulation of hematopoiesis by irradiated human HSPCs in secondary recipients.

(A) Changes in chimerism of human hematopoietic cells in the PB of secondary recipients. At 2 weeks post-irradiation, CD34+ human HSPCs were purified from the primary recipient mice, and 2 x 106 CD34+ HSPCs were transplanted into secondary NOG recipients. Chimerism of CD45+ human and mouse hematopoietic cells in the PB are shown in upper panels. The percentages of CD11b+ myeloid cells, CD19+ B cells, and CD3+ T cells in CD45+ human hematopoietic cells in the PB are shown in lower panels. PB data of NOG mice just before the irradiation (Pre, n = 27) are also indicated at week 0. Data are presented as mean ± S.D. (0 Gy, n = 5; 0.5 Gy, n = 5; 1.0 Gy, n = 5). (B) Total BM cell numbers and chimerism of CD45+ human and mouse hematopoietic cells at 12 weeks after secondary transplantation. (C) Absolute numbers of CD34+CD38- HSCs, CD34+CD38+ HPCs, CMPs and LMPPs in the BM (bilateral femurs and tibiae) (0 Gy, n = 5; 0.5 Gy, n = 5; 1.0 Gy, n = 5). (D) Numbers of colony-forming cells included in 1,000 CD34+ human HSPCs recovered from the BM 12 weeks after secondary transplantation (0–1.0 Gy, n = 3 each). The data in Fig 3B-D are presented as scatter diagrams with median values (bars). P-values for trend were calculated by nonparametric Jonckheere-Terpstra test.

https://doi.org/10.1371/journal.pone.0132041.g003

thumbnail
Fig 4. Long-term radiation effects on human HSPCs in primary recipient mice.

Changes in chimerism of human hematopoietic cells in the PB of primary recipients at 12 weeks post-irradiation. Chimerism of CD45+ human and mouse hematopoietic cells in the PB are shown in upper panels. The percentages of CD11b+ myeloid cells, CD19+ B cells, and CD3+ T cells in CD45+ human hematopoietic cells in the PB are shown in lower panels. PB data of NOG mice just before the irradiation (Pre) are also indicated. Data are presented as scattering diagrams with medium values (bars) (Pre, n = 15; 0 Gy, n = 5; 0.5 Gy, n = 5; 1.0 Gy, n = 5). P-values for trend were calculated by nonparametric Jonckheere-Terpstra test. The t-test was used for comparison of mean values.

https://doi.org/10.1371/journal.pone.0132041.g004

Repopulation of hematopoiesis by irradiated human HSPCs in secondary recipients

To evaluate the effects of non-lethal radiation on HSPCs repopulated in NOG mice, we then purified CD34+ HSPCs from the primary recipient mice at 2 weeks post-irradiation (9 mice were pooled) and transplanted 2 x 106 CD34+ HSPCs into secondary NOG recipients. At 12 weeks post-secondary transplantation, we analyzed the contribution of human HSPCs to hematopoiesis (Fig 3). CD34+ HSPCs from irradiated mice showed severely impaired reconstitution capacity in the secondary recipient mice. Chimerism of human CD45+ hematopoietic cells in both the PB and BM declined in a radiation dose-dependent manner while that of mouse CD45+ hematopoietic cells increased (Fig 3A and 3B). Repopulation of human CD34+CD38- HSCs, CD34+CD38+ HPCs, CMPs and LMPPs in the BM was compromised in both proportion (data not shown) and absolute numbers in a radiation dose-dependent manner (Fig 3C). Of interest, non-lethal irradiation impaired contribution of human hematopoietic cells to the PB cells, particularly CD19+ B lymphocytes, which indicates the myeloid-biased repopulation of human hematopoietic cells (Fig 3A). CD34+ HSPCs from the secondary recipient mice again gave rise to fewer colonies in a manner dependent on radiation doses to which the cells had been exposed in the primary recipient mice (0 Gy vs. 0.5 Gy, P = 0.019 and 0 Gy vs. 1.0 Gy P = 0.002 by Dunnett t-test) (Fig 3D). Myeloid-biased hematopoiesis at the expense of B lymphopoiesis was also observed at 12 weeks post-irradiation in primary recipients (5 mice each) (Fig 4). Because impaired reconstituting capacity, myeloid-biased differentiation and inefficient production of B cells are hallmarks of aged HSCs [7], [10], [24], these data indicate that non-lethal doses of radiation accelerate part of the aging phenotypes of human HSPCs in humanized mice.

Little is known about the effects of non-lethal radiation on B-cell production from HSPCs. To understand how non-lethal irradiation impairs B lymphopoiesis, we purified CD34+ HSPCs at 2 weeks post-irradiation and co-cultured graded numbers of purified CD34+ HSPCs (50, 102, 5x102, 103, 5x103, 104 cells) with TSt-4 stromal cells in the presence of SCF, IL-7 and Flt3L (10 ng /ml each) for 4 weeks and analyzed the differentiation of CD19+ B cells. The frequency of B-cell-producing cells was 1/73 in control CD34+ HSPCs from the secondary recipients repopulated with non-irradiated CD34+ HSPCs. In contrast, the frequency of B cell-producing cells was 1/918 in CD34+ HSPCs from the secondary recipients repopulated with irradiated CD34+ HSPCs at a dose of 0.5 Gy in primary recipient mice. This frequency of irradiated CD34+ HSPCs was 12.5-fold lower than that of non-irradiated cells (Fig 5). A significant reduction in B cell-producing capacity of CD34+ HSPCs indicates that radiation damage affects the B cell commitment of CD34+ HSPCs.

thumbnail
Fig 5. In vitro differentiation of B cells from human HSPCs irradiated in humanized mice.

Frequency of B cell-producing CD34+ HSPCs irradiated in humanized mice. Human CD34+ HSPCs were recovered from the primary recipients at 2 weeks post-irradiation. Graded numbers of purified CD34+ HSPCs (50, 102, 5 x 102, 103, 5 x 103, 104 cells) were co-cultured with TSt-4 stromal cells in the presence of SCF, IL-7 and Flt3L (10 ng /ml each) for 4 weeks. Generation of CD19+ B cells was determined by flow cytometric analysis. The frequency of B–cell producing HSPCs was calculated using L-Calc software. The % of wells with B cell production and frequency of B-cell producing CD34+ HSPCs are summarized in the table and each data is plotted in the bottom panel. *** P < 0.001. The frequencies in non-irradiated and irradiated cells were compared by the χ2-test for a 2 x 2 contingency table.

https://doi.org/10.1371/journal.pone.0132041.g005

The molecular mechanisms underlying myeloid-biased differentiation of aged HSPCs remain controversial [6], [10]. However, reduced expression of EBF1 and PAX5, key B-cell regulator genes, has been implicated in this process [25].

To obtain an insight into the myeloid-biased differentiation of irradiated HSPCs, we compared the expression levels of each key B-cell regulator gene in CD34+ HSPCs from humanized mice at 2 weeks post-irradiation and those at 12 weeks post-secondary transplantation. Of note, expression levels of IL-7R, EBF1 and PAX5 encoding IL-7 receptor α and the EBF1 and PAX5 master transcription factors, respectively [26], [27] were significantly downregulated upon 0.5 Gy of irradiation (Fig 6A). This trend was conserved in the HSPCs recovered from the primary recipients at 2 weeks post-irradiation and the secondary recipients at 12 weeks post-transplantation (Fig 6A). In addition, the expression of cyclin-dependent kinase inhibitor 2A (CDKN2A) or p16INK4A, a hallmark of aging of HSCs [28], was upregulated in irradiated HSPCs as we observed in our previous study (Fig 6B) [19]. Among the transcriptional regulators implicated in HSC self-renewal [29], [30], MEIS1 was downregulated but HOXA9 was not (Fig 6B).

thumbnail
Fig 6. Impaired activation of B cell regulator genes in irradiated human HSPCs.

(A) Quantitative RT-PCR analysis of IL7R, EBF1 and PAX5 in CD34+ human HSPCs from humanized mice at 2 weeks post-irradiation and at 12 weeks post-secondary transplantation (n = 3 each). (B) Quantitative RT-PCR analysis of p16INKA, HOXA9 and MEIS1 in CD34+ human HSPCs from humanized mice at 2 weeks post-irradiation and at 12 weeks post-secondary transplantation (n = 3 each). Relative expression levels of each gene mRNA were adjusted in reference to GAPDH mRNA expression levels. Data are shown as the mean e adjust P < 0.05, **P < 0.01 and ***P < 0.001. The t-test was used for comparison of mean values.

https://doi.org/10.1371/journal.pone.0132041.g006

Discussion

Understanding the health effects of low levels of ionizing radiation remains an important issue. Although insufficient support of human HSPCs by the mouse BM niche cells is one of the limitations of xenotransplantation models, the humanized mouse model allows us to assess the long-term radiation effects as we previously reported [19].

In this study, non-lethal radiation doses of 0.5 and 1.0 Gy were shown to profoundly compromise the function of human HSPCs residing in the BM microenvironment. Human CD34+ HSPCs from irradiated humanized mice showed severely impaired reconstitution capacity in the secondary recipient mice and showed myeloid-biased repopulation at the expense of B-cell differentiation. Since these phenotypes are commonly observed in aged HSPCs, non-lethal ionizing irradiation could be one of the critical external stresses that promote aging of human HSPCs in the bone marrow niche.

Age-related decreases in regenerative capacity of HSCs have been, at least partly, attributed to accumulation of DNA damage [15], [16] or telomere shortening [31]. DNA damage triggers signaling cascades that activate tumor suppressor genes such as p53 and p16INK4A. Although its role in HSC aging is controversial [32], p16Ink4a expression increases with aging in HSCs and absence of p16Ink4a ameliorates some of the age-related phenotypes of HSCs in mice. We have previously demonstrated that DNA damage accumulated in HSCs upon non-lethal doses of irradiation in humanized mice and that expression of p16INK4A, a hallmark of aging of HSCs, was upregulated in HSPCs in irradiated mice [19]. In this study, p16INK4A was again significantly upregulated in CD34+ HSPCs which were exposed to 0.5 Gy irradiation in the BM niche of humanized mice; and this p16INK4A upregulation may account for the impaired HSC function. HOXA9 and MEIS1 are transcription factors essential for the self-renewal of HSCs [29], [30]. Of interest, while the expression of HOXA9 was not altered, that of MEIS1 was significantly downregulated in CD34+ HSPCs irradiated with 0.5 Gy, at both early and late time points post-irradiation (2 weeks post-irradiation and 3 months post-secondary transplantation, respectively). The transcription factor Meis1, belonging to the TALE family of non-Hox homeobox genes, protects and preserves HSCs by restricting oxidative metabolism [30]. These findings suggest that non-lethal doses of radiation could compromise, at least partly, transcriptional activation of genes crucial for the self-renewal capacity of HSCs.

Recently, HSCs have been demonstrated to be more heterogeneous in their capacity to repopulate hematopoiesis and produce a variety of progenies [3336]. Notably, there are higher numbers of myeloid-biased HSCs in the aged BM compared with the young BM [24], [37], [38]. As a consequence, granulocyte/macrophage progenitors increase while common lymphoid progenitors decrease in aged mice. Therefore, aged HSCs show a greater propensity for myeloid differentiation compared with younger HSCs [7], [24], [25]. In our humanized mouse model, non-lethal radiation profoundly impaired the capacity of CD34+ HSPCs to give rise to B cells both in vitro and in vivo. These recent findings suggest that radiation promotes clonal expansion of myeloid-biased HSCs at the expense of lymphoid-biased HSCs via inducing DNA damage. Key regulators of B cell differentiation, EBF1, PAX5 and IL-7R were all downregulated in CD34+ HSPCs upon 0.5 Gy of irradiation as was reported in human aged HSCs [25]. Downregulation of these B cell regulators could reflect the clonal expansion of myeloid-biased HSPCs in CD34+ HSPCs. Alternatively, radiation may alter transcriptional profiles of HSPCs crucial for their commitment and differentiation probably by inducing epigenetic changes at developmental regulator gene loci such as EBF1 and PAX5.

In this study, we clearly demonstrated that non-lethal doses of radiation caused profound functional changes in HSCs in humanized mice. However, irradiation of humanized mice affects not only human HSPCs but also mouse BM niche and hematopoietic cells. Damaged mouse cells, particularly niche cells, could indirectly affect the human HSPCs. This possibility needs to be experimentally addressed in the future. To this end, NOD/Rag2null IL2rγnull mice, which are more resistant to radiation compared with NOG mice, would be useful. Finally, continuous efforts are required to clarify the effects of much lower doses of irradiation on HSCs. Such efforts would be valuable in order to prevent and contend with health hazards caused by occupational or unexpected radiation exposures.

Acknowledgments

We thank the Japanese Red Cross Kanto-Koshinetsu Cord Blood Bank (Tatsumi, Tokyo, Japan) for providing us human cord blood, Y. Makiko for technical assistance and E. Douple and Ola Mohammed Kamel Rizq for critical review of the manuscript.

Author Contributions

Conceived and designed the experiments: CW YK SK KN AI. Performed the experiments: CW MO GS TT NH MM-K YN-T. Analyzed the data: CW KI AI. Wrote the paper: CW YK SK KN AI.

References

  1. 1. Zon LI (2008) Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature 453, 306–313. pmid:18480811
  2. 2. Doulatov S, Notta F, Laurenti E, Dick JE (2012) Hematopoiesis: a human perspective. Cell Stem Cell 10, 120–136. pmid:22305562
  3. 3. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153, 1194–217. pmid:23746838
  4. 4. Signer RA, Morrison SJ (2013) Mechanisms that regulate stem cell aging and life span. Cell Stem Cell 12, 152–165. pmid:23395443
  5. 5. Morrison SJ, Wandycz AM, Akashi K, Globerson A, Weissman IL (1996) The aging of hematopoietic stem cells. Nat Med 2, 1011–1016. pmid:8782459
  6. 6. Sudo K, Ema H, Morita Y, Nakauchi H (2000) Age-associated characteristics of murine hematopoietic stem cells. J Exp Med 192, 1273–1280. pmid:11067876
  7. 7. Rossi DJ, Bryder D, Zahn JM, Ahlenius H, Sonu R, Wagers AJ, et al. (2005) Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci USA 102, 9194–9199. pmid:15967997
  8. 8. Pang WW, Price EA, Sahoo D, Beerman I, Maloney WJ, Rossi DJ, et al. (2011) Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc Natl Acad Sci USA 108, 20012–20017. pmid:22123971
  9. 9. Kuranda K, Vargaftig J, de la Rochere P, Dosquet C, Charron D, Bardin F, et al. (2011) Age-related changes in human hematopoietic stem/progenitor cells. Aging Cell 10, 542–546. pmid:21418508
  10. 10. Geiger H, de Haan G, Florian MC (2013) The ageing haematopoietic stem cell compartment. Nat Rev Immunol 13, 376–389. pmid:23584423
  11. 11. Oshima M, Iwama A (2014) Epigenetics of hematopoietic stem cell aging and disease. Int J Hematol 100, 326–334. pmid:25082353
  12. 12. Hanahan D, Weinberg RA (2000) The Hallmarks of Cancer. Cell 100, 57–70 pmid:10647931
  13. 13. Shao L, Feng W, Li H, Gardner D, Luo Y, Wang Y, et al. (2014) Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent manner. Blood 123, 3105–3015. pmid:24622326
  14. 14. Van Zant G, Liang Y (2003) The role of stem cells in aging. Exp Hematol 31, 659–672. pmid:12901970
  15. 15. Nijnik A, Woodbine L, Marchetti C, Dawson S, Lambe T, Liu C, et al. (2007) DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690. pmid:17554302
  16. 16. Rossi DJ, Bryder D, Seita J, Nussenzweig A, Hoeijmakers J, Weissman IL (2007) Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729. pmid:17554309
  17. 17. Beerman I, Bock C, Garrison BS, Smith ZD, Gu H, Meissner A, et al. (2014) Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12, 413–425.
  18. 18. Rübe CE, Fricke A, Widmann TA, Fürst T, Madry H, Pfreundschuh M, et al. (2011) Accumulation of DNA damage in hematopoietic stem and progenitor cells during human aging. PLoS One 6, e17487. pmid:21408175
  19. 19. Wang C, Nakamura S, Oshima M, Mochizuki-Kashio M, Nakajima-Takagi Y, Osawa M, et al. (2013) Compromised hematopoiesis and increased DNA damage following non-lethal ionizing radiation of a human hematopoietic system reconstituted in immunodeficient mice. Int J Rad Biol 89, 132–137. pmid:23020858
  20. 20. Nakachi K, Hayashi T, Hamatani K, Eguchi H, Kusunoki Y (2008) Sixty years of follow-up of Hiroshima and Nagasaki survivors: current progress in molecular epidemiology studies. Mutat Res 659, 109–117. pmid:18406659
  21. 21. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, et al. (2002) NOD/SCID/gamma (c) (null) mouse: An excellent recipient mouse model for engraftment of human cells. Blood 100, 3175– 3182. pmid:12384415
  22. 22. Masuda K, Kubagawa H, Ikawa T, Chen CC, Kakugawa K, Hattori M, et al. (2005) Prethymic T-cell development defined by the expression of paired immunoglobulin-like receptors. EMBO J 24, 4052–4060. pmid:16292344
  23. 23. Fulop GM Phillips RA (1990) The scid mutation in mice causes a general defect in DNA repair. Nature 347, 479–482. pmid:2215662
  24. 24. Beerman I, Bhattacharya D, Zandi S, Sigvardsson M, Weissman IL, Bryder D, et al. (2010) Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion. Proc Natl Acad Sci USA 107, 5465–5470 pmid:20304793
  25. 25. Lescale C, Dias S, Maës J, Cumano A, Szabo P, Charron D, et al. (2010) Reduced EBF expression underlies loss of B-cell potential of hematopoietic progenitors with age. Aging Cell 9, 410–419. pmid:20331442
  26. 26. Von Freeden-Jeffry U, Vieira P, Lucian LA, McNeil T, Burdach SEG, Murray R (1995) Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine. J Exp Med 181, 1519–1526. pmid:7699333
  27. 27. Ramírez J, Lukin K, Hagman J (2010) From hematopoietic progenitors to B cells: mechanisms of lineage restriction and commitment. Curr Opin Immunol 22, 177–184. pmid:20207529
  28. 28. Janzen V, Forkert R, Fleming HE, Saito Y, Waring MT, Dombkowski DM, et al. (2006) Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443, 421–426. pmid:16957735
  29. 29. Lawrence HJ, Christensen J, Fong S, Hu YL, Weissman I, Sauvageau G, et al. (2005) Loss of expression of the Hoxa-9 homeobox gene impairs the proliferation and repopulating ability of hematopoietic stem cells. Blood 106, 3988–3994. pmid:16091451
  30. 30. Unnisa Z, Clark JP, Roychoudhury J, Thomas E, Tessarollo L, Copeland NG, et al. (2012) Meis1 preserves hematopoietic stem cells in mice by limiting oxidative stress. Blood 120, 4973–4981. pmid:23091297
  31. 31. Sperka T, Wang J, Rudolph KL (2012) DNA damage checkpoints in stem cells, ageing and cancer. Nat Rev Mol Cell Biol 13, 579–590. pmid:22914294
  32. 32. Attema LL, Pronk CJ, Norddahl GL, Nygren JM, Bryder D (2009) Hematopoietic stem cell ageing is uncoupled from p16INK4A-mediated senescence. Oncogene 28, 2238–2243. pmid:19398954
  33. 33. Dykstra B, Kent D, Bowie M, McCaffrey L, Hamilton M, Lyons K, et al. (2007) Long-term propagation of distinct hematopoietic differentiation programs in vivo. Cell Stem Cell 1, 218–229. pmid:18371352
  34. 34. Benz C, Copley MR, Kent DG, Wohrer S, Cortes A, Aghaeepour N, et al. (2012) Hematopoietic stem cell subtypes expand differentially during development and display distinct lymphopoietic programs. Cell Stem Cell 10, 273–283. pmid:22385655
  35. 35. Yamamoto R, Morita Y, Ooehara J, Hamanaka S, Onodera M, Rudolph KL, et al. (2013) Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell 154, 1112–1126. pmid:23993099
  36. 36. Ema H, Morita Y, Suda T (2014) Heterogeneity and hierarchy of hematopoietic stem cells. Exp Hematol 42, 74–82. pmid:24269919
  37. 37. Muller-Sieburg CE, Cho RH, Karlsson L, Huang JF, Sieburg HB (2004) Myeloid-biased hematopoietic stem cells have extensive self-renewal capacity but generate diminished lymphoid progeny with impaired IL-7 responsiveness. Blood 103, 4111–4118. pmid:14976059
  38. 38. Challen GA, Boles NC, Chambers SM, Goodell MA (2010) Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1. Cell Stem Cell 6, 265–278. pmid:20207229