Bmi1 confers resistance to oxidative stress on hematopoietic stem cells

Shunsuke Nakamura, Motohiko Oshima, Jin Yuan, Atsunori Saraya, Satoru Miyagi, Takaaki Konuma, Satoshi Yamazaki, Mitsujiro Osawa, Hiromitsu Nakauchi, Haruhiko Koseki, Atsushi Iwama, Shunsuke Nakamura, Motohiko Oshima, Jin Yuan, Atsunori Saraya, Satoru Miyagi, Takaaki Konuma, Satoshi Yamazaki, Mitsujiro Osawa, Hiromitsu Nakauchi, Haruhiko Koseki, Atsushi Iwama

Abstract

Background: The polycomb-group (PcG) proteins function as general regulators of stem cells. We previously reported that retrovirus-mediated overexpression of Bmi1, a gene encoding a core component of polycomb repressive complex (PRC) 1, maintained self-renewing hematopoietic stem cells (HSCs) during long-term culture. However, the effects of overexpression of Bmi1 on HSCs in vivo remained to be precisely addressed.

Methodology/principal findings: In this study, we generated a mouse line where Bmi1 can be conditionally overexpressed under the control of the endogenous Rosa26 promoter in a hematopoietic cell-specific fashion (Tie2-Cre;R26Stop(FL)Bmi1). Although overexpression of Bmi1 did not significantly affect steady state hematopoiesis, it promoted expansion of functional HSCs during ex vivo culture and efficiently protected HSCs against loss of self-renewal capacity during serial transplantation. Overexpression of Bmi1 had no effect on DNA damage response triggered by ionizing radiation. In contrast, Tie2-Cre;R26Stop(FL)Bmi1 HSCs under oxidative stress maintained a multipotent state and generally tolerated oxidative stress better than the control. Unexpectedly, overexpression of Bmi1 had no impact on the level of intracellular reactive oxygen species (ROS).

Conclusions/significance: Our findings demonstrate that overexpression of Bmi1 confers resistance to stresses, particularly oxidative stress, onto HSCs. This thereby enhances their regenerative capacity and suggests that Bmi1 is located downstream of ROS signaling and negatively regulated by it.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Generation of mice overexpressing Bmi1…
Figure 1. Generation of mice overexpressing Bmi1 in hematopoietic cells.
(A) Strategy for making a knock-in allele for Bmi1 downstream of the Rosa26 promoter. A loxP-flanked neor-stop cassette followed by Flag-tagged Bmi1, an frt-flanked IRES-eGFP cassette, and a bovine polyadenylation sequence was knocked-in the Rosa26 locus. (B) Quantitative RT-PCR analysis of Bmi1 in BM LSK cells from Tie2-Cre and Tie2-Cre;R26StopFLBmi1 mice. mRNA levels were normalized to Hprt1 expression. Expression levels relative to that in Tie2-Cre LSK cells are shown as the mean ± S.D. (n = 3). (C) Western blotting analysis of Bmi1 in c-Kit+ BM cells from Tie2-Cre and Tie2-Cre;R26StopFLBmi1 mice. α-tubulin was used as the loading control. (D) Hematopoietic analysis of 10-week-old Tie2-Cre and Tie2-Cre;R26StopFLBmi1 mice. Absolute numbers of BM cells, CD34-LSK cells, and LSK cells in bilateral femurs and tibiae are presented as the mean ± S.D. (upper panels, Tie2-Cre; n = 7, Tie2-Cre;R26StopFLBmi1; n = 8). PB analysis of 10-week-old Tie2-Cre and Tie2-Cre;R26StopFLBmi1 mice. White blood cell (WBC) counts and lineage contribution of myeloid, B, and T cells are shown as the mean ± S.D. (lower panels, Tie2-Cre; n = 7, Tie2-Cre;R26StopFLBmi1; n = 8). ** p<0.01.
Figure 2. Effects of overexpression of Bmi1…
Figure 2. Effects of overexpression of Bmi1 on HSCs in vitro.
(A) Colony formation by HSCs isolated from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice. Single CD34-LSK cells were sorted into 96-well microtiter plates containing the SF-O3 medium supplemented with 10% FBS and multiple cytokines (10 ng/ml SCF, 10 ng/ml TPO, 10 ng/ml IL-3, and 3 u/ml EPO) and allowed to form colonies. At day 14 of culture, the colonies were counted and individually collected for morphological examination. Absolute numbers of LPP and HPP-CFCs which gave rise to colonies with a diameter less and greater than 1 mm, respectively are shown as the mean ± S.D. for triplicate cultures (left panel). Absolute numbers of each colony types were defined by the composition of colonies (right panel). Colonies were recovered and examined by microscopy to determine colony types. Composition of colonies is depicted as n, neutrophils; m, macrophages; E, erythroblasts; and M, megakaryocytes. (B) Colony formation by HSCs cultured for 10 days. CD34-LSK cells from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml of SCF and TPO. At day 10 of culture, the cells were counted (left panel) and plated in methylcellulose medium to allow formation of colonies in the presence of 20 ng/ml SCF, 20 ng/ml TPO, 20 ng/ml IL-3, and 3 u/ml EPO. Absolute numbers of LPP and HPP-CFCs (middle panel) are shown as the mean ± S.D. for triplicate cultures. Absolute numbers of each colony type are shown in the right panel. (C) Flow cytometric analysis of CD34-LSK HSCs at day14 of culture. Representative flow cytometric profiles of LSK cells in cultures of CD34-LSK HSCs from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice are depicted. The proportion of Lin- and LSK cells in total cells are indicated. (D) Quantitative RT-PCR analysis of the expression of p19Arf, and Bmi1 in Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) LSK cells. LSK cells were purified by cell sorting from CD34-LSK cultures in (C) at day 14 of culture. Each value was normalized to Hprt1 expression and the expression level of each gene in control cells was arbitrarily set to 1. Data are shown as the mean ± S.D. for triplicate analyses. * p<0.05, **p<0.01.
Figure 3. Overexpression of Bmi1 enhances expansion…
Figure 3. Overexpression of Bmi1 enhances expansion of HSCs ex vivo.
Competitive repopulating unit (CRU) assays using limiting numbers of CD34-LSK cells from Tie2-Cre (Control) mice and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice. Freshly isolated CD34-LSK cells were immediately used for BM transplantation, or CD34-LSK cells were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml SCF and TPO for 10 days, and then a fraction of the culture cells corresponding to the indicated number (0.5∼10) of initial CD34-LSK cells was subjected to BM transplantation. The test cells (CD45.2) were transplanted along with 2×105 competitor BM cells (CD45.1) into CD45.1 recipient mice lethally irradiated at a dose of 9.5 Gy. Percent chimerism of donor cells in the recipient PB was determined at 16 weeks after transplantation. The mice with chimerism more than 1% in all three lineages (myeloid, B, and T cells) were considered successfully engrafted and the others were defined as negative mice. The frequency of HSCs was calculated using L-Calc software. The proportion of engrafted mice, frequency of functional HSCs, and the 95% confidence interval (CI) are summarized in the table and each data is plotted in the bottom panel. *** p<0.001.
Figure 4. Overexpression of Bmi1 protects HSCs…
Figure 4. Overexpression of Bmi1 protects HSCs during serial transplantation.
(A) Serial transplantation of BM cells. BM cells (5×105) from Tie2-Cre (denoted as “C”) and Tie2-Cre;R26StopFLBmi1 (denoted as “B”) mice (CD45.2) along with 5×105 competitor BM cells (CD45.2) were transplanted into CD45.1 recipient mice lethally irradiated at a dose of 9.5 Gy. For serial transplantation, BM cells were collected from all recipient mice at 12–20 weeks after transplantation and pooled together. Then, 5×106 BM cells were transplanted into lethally irradiated recipient mice without competitor cells. Third and fourth transplantation were similarly performed using 5×106 pooled BM cells. Percent chimerism of donor cells in the recipient PB and BM LSK cells was determined at 16 weeks post-transplantation. Results are shown as the mean ± S.D. (n = 6, 3rd transplantation; n = 4). (B) Serial transplantation of cultured CD34-LSK cells. CD34-LSK cells were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml of SCF and TPO for 10 days. Then, the cells in culture corresponding to the 20 initial CD34-LSK cells were injected into a recipient mouse along with 2×105 competitor BM cells (CD45.2) as described in (A) (n = 6, 4th transplantation; n = 5). * P<0.05, ** P<0.01.
Figure 5. DNA damage response of Tie2-Cre;R26Stop…
Figure 5. DNA damage response of Tie2-Cre;R26StopFLBmi1 HSCs.
(A) DNA damage response of CD34-LSK cells from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice in vitro. Purified CD34-LSK cells were irradiated (IR) at a dose of 2 Gy. At 2 and 4 hours after irradiation, cells were stained with anti-γH2AX. Numbers of γH2AX foci expressed per cell are depicted. (B) DNA damage response of CD34-LSK cells from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice in vivo. LSK cells were purified from the recipients of tertiary transplantation and stained with anti-γH2AX. Numbers of γH2AX foci expressed per cell are depicted as the mean ± S.D. (n = 3).
Figure 6. Overexpression of Bmi1 confers oxidative…
Figure 6. Overexpression of Bmi1 confers oxidative stress on HSCs.
(A) Colony formation by HSCs cultured for 3 days. CD34-LSK cells from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml SCF, TPO and 0.05 mM of BSO. At day 3 of culture, the cells were plated in methylcellulose medium to allow formation of colonies in the presence of 20 ng/ml SCF, 20 ng/ml TPO, 20 ng/ml IL-3, and 3 u/ml EPO. Absolute numbers of LPP and HPP-CFCs (left panel) are shown as the mean ± S.D. for triplicate cultures. Absolute numbers of each colony types are shown in the right panel. Data are shown as the mean ± S.D. for triplicate analyses. Statistical analyses were performed on the total colony numbers (left panel) and nmEM colony numbers (right panel), respectively. **p<0.01. (B) Levels of ROS in cells overexpressing Bmi1. CD34-LSK cells from Tie2-Cre (Control) and Tie2-Cre;R26StopFLBmi1 (Bmi1) mice were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml SCF and TPO. Representative flow cytometric profiles of LSK and Lineage marker-Sca-1low/−c-Kit+ cells in cultures at day 14 are depicted. (C) Effects of NAC on Bmi1 culture. CD34-LSK cells from Tie2-Cre and Tie2-Cre;R26StopFLBmi1 mice were cultured in the SF-O3 serum-free medium supplemented with 50 ng/ml SCF and TPO in the presence and absence of 150 µM NAC. Representative flow cytometric profiles of LSK cells in cultures at day 14 are depicted. The proportion of Lin- and LSK cells in total cells are indicated.

References

    1. Simon JA, Kingston RE. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol. 2009;10:697–708.
    1. Iwama A, Oguro H, Negishi M, Kato Y, Nakauchi H. Epigenetic regulation of hematopoietic stem cell self-renewal by polycomb group genes. Int J Hematol. 2005;81:294–300.
    1. Konuma T, Oguro H, Iwama A. Role of the polycomb group proteins in hematopoietic stem cells. Dev Growth Differ. 2010;52:505–516.
    1. Sauvageau M, Sauvageau G. Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell. 2010;7:299–313.
    1. Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423:255–260.
    1. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423:302–305.
    1. Iwama A, Oguro H, Negishi M, Kato Y, Morita Y, et al. Enhanced self-renewal of hematopoietic stem cells mediated by the polycomb gene product Bmi-1. Immunity. 2004;21:843–851.
    1. Oguro H, Iwama A, Morita Y, Kamijo T, van Lohuizen M, et al. Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice. J Exp Med. 2006;203:2247–2253.
    1. Oguro H, Yuan J, Ichikawa H, Ikawa T, Yamazaki S, et al. Poised lineage specification in multipotent hematopoietic stem and progenitor cells by the polycomb protein Bmi1. Cell Stem Cell. 2010;6:279–286.
    1. Mihara K, Chowdhury M, Nakaju N, Hidani S, Ihara A, et al. Bmi-1 is useful as a novel molecular marker for predicting progression of myelodysplastic syndrome and patient prognosis. Blood. 2006;107:305–308.
    1. Rizo A, Horton SJ, Olthof S, Dontje B, Ausema A, et al. BMI1 collaborates with BCR-ABL in leukemic transformation of human CD34+ cells. Blood. 2010;116:4621–4630.
    1. Kisanuki YY, Hammer RE, Miyazaki J, Williams SC, Richardson JA, et al. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev Biol. 2001;230:230–242.
    1. Takano H, Ema H, Sudo K, Nakauchi H. Asymmetric division and lineage commitment at the level of hematopoietic stem cells: inference from differentiation in daughter cell and granddaughter cell pairs. J Exp Med. 2004;199:295–302.
    1. Ema H, Takano H, Sudo K, Nakauchi H. In vitro self-renewal division of hematopoietic stem cells. J Exp Med. 2000;192:1281–1288.
    1. Shima H, Takubo K, Iwasaki H, Yoshihara H, Gomei Y, et al. Reconstitution activity of hypoxic cultured human cord blood CD34-positive cells in NOG mice. Biochem Biophys Res Commun. 2009;378:467–472.
    1. Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12:446–451.
    1. Yahata T, Takanashi T, Muguruma Y, Ibrahim AA, Matsuzawa H, et al. Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood. 2011;118:2941–2950.
    1. Rossi DJ, Jamieson CH, Weissman IL. Stems cells and the pathways to aging and cancer. Cell. 2008;132:681–696.
    1. Chagraoui J, Hébert J, Girard S, Sauvageau G. An anticlastogenic function for the Polycomb Group gene Bmi1. Proc Natl Acad Sci USA. 2011;108:5284–5289.
    1. Ginjala V, Nacerddine K, Kulkarni A, Oza J, Hill SJ, et al. BMI1 is recruited to DNA breaks and contributes to DNA damage-induced H2A ubiquitination and repair. Mol Cell Biol. 2011;31:1972–1982.
    1. Facchino S, Abdouh M, Chatoo W, Bernier G. BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. J Neurosci. 2010;30:10096–10111.
    1. Shao L, Li H, Pazhanisamy SK, Meng A, Wang Y, et al. Reactive oxygen species and hematopoietic stem cell senescence. Int J Hematol. 2010;94:24–32.
    1. Jang YY, Sharkis SJ. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood. 2007;110:3056–3063.
    1. Liu J, Liu C, Chen J, Song S, Lee IH, et al. Bmi1 regulates mitochondrial function and the DNA damage response pathway. Nature. 2009;459:387–392.
    1. Rizo A, Olthof S, Han L, Vellenga E, de Haan G, et al. Repression of BMI1 in normal and leukemic human CD34+ cells impairs self-renewal and induces apoptosis. Blood. 2009;114:1498–1505.
    1. Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009;461:537–541.
    1. Bracken AP, Kleine-Kohlbrecher D, Dietrich N, Pasini D, Gargiulo G, et al. The polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 2007;21:525–530.
    1. Negishi M, Saraya A, Mochizuki S, Helin K, Koseki H, et al. A novel zinc finger protein Zfp277 mediates transcriptional repression of the Ink4a/Arf locus through polycomb repressive complex 1. PLoS One. 2010;5:e12373.
    1. Kamminga LM, Bystrykh LV, de Boer A, Houwer S, Douma J, et al. The Polycomb group gene Ezh2 prevents hematopoietic stem cell exhaustion. Blood. 2006;107:2170–2179.
    1. Stapels M, Piper C, Yang T, Li M, Stowell C, et al. Polycomb group proteins as epigenetic mediators of neuroprotection in ischemic tolerance. Sci Signal. 2010;3:ra15.
    1. Bracken AP, Helin K. Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat Rev Cancer. 2009;9:773–784.
    1. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458:780–783.
    1. Zhou J, Bi C, Cheong LL, Mahara S, Liu SC, et al. The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood. 2011;118:2830–2839.
    1. Sasaki Y, Derudder E, Hobeika E, Pelanda R, Reth M, et al. Canonical NF-kappaB activity, dispensable for B cell development, replaces BAFF-receptor signals and promotes B cell proliferation upon activation. Immunity. 2006;24:729–739.
    1. Fukamachi H, Fukuda K, Suzuki M, Furumoto T, Ichinose M, et al. Mesenchymal transcription factor Fkh6 is essential for the development and differentiation of parietal cells. Biochem Biophys Res Commun. 2001;280:1069–1076.

Source: PubMed

3
Abonneren