Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The mitochondrial respiratory chain is essential for haematopoietic stem cell function

Abstract

Adult and fetal haematopoietic stem cells (HSCs) display a glycolytic phenotype, which is required for maintenance of stemness; however, whether mitochondrial respiration is required to maintain HSC function is not known. Here we report that loss of the mitochondrial complex III subunit Rieske iron-sulfur protein (RISP) in fetal mouse HSCs allows them to proliferate but impairs their differentiation, resulting in anaemia and prenatal death. RISP-null fetal HSCs displayed impaired respiration resulting in a decreased NAD+/NADH ratio. RISP-null fetal HSCs and progenitors exhibited an increase in both DNA and histone methylation associated with increases in 2-hydroxyglutarate (2HG), a metabolite known to inhibit DNA and histone demethylases. RISP inactivation in adult HSCs also impaired respiration resulting in loss of quiescence concomitant with severe pancytopenia and lethality. Thus, respiration is dispensable for adult or fetal HSC proliferation, but essential for fetal HSC differentiation and maintenance of adult HSC quiescence.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Loss of RISP in haematopoietic cells impairs mitochondrial oxidative metabolism and results in severe fetal anaemia.
Figure 2: RISP is essential for fetal HSC differentiation.
Figure 3: Ablation of RISP depletes fetal myeloid progenitors.
Figure 4: RISP deficiency impairs fetal HSC competitive repopulation capacity.
Figure 5: Loss of RISP in fetal HSCs leads to deregulation of gene expression, DNA and histone hypermethylation, and histone hypoacetylation.
Figure 6: RISP deficiency decreases NAD+/NADH ratio and increases 2HG.
Figure 7: Loss of RISP in adult haematopoietic stem cells impairs mitochondrial oxidative metabolism and leads to a severe pancytopenia phenotype.
Figure 8: RISP deficiency in adult HSCs leads to impaired differentiation, loss of quiescence, increased apoptosis and increased DNA damage.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Chandel, N. S., Jasper, H., Ho, T. T. & Passegue, E. Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing. Nat. Cell Biol. 18, 823–832 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243–256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Takubo, K. et al. Regulation of glycolysis by pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Adelman, D. M., Maltepe, E. & Simon, M. C. Multilineage embryonic hematopoiesis requires hypoxic ARNT activity. Genes Dev. 13, 2478–2483 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Norddahl, G. L. et al. Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell 8, 499–510 (2011).

    Article  CAS  PubMed  Google Scholar 

  7. Parmar, K., Mauch, P., Vergilio, J.-A., Sackstein, R. & Down, J. D. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc. Natl Acad. Sci. USA 104, 5431–5436 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Spencer, J. A. et al. Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 508, 269–273 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12, 446–451 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Chen, C. et al. TSC–mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J. Experim. Med. 205, 2397–2408 (2008).

    Article  CAS  Google Scholar 

  12. Jung, H. et al. TXNIP maintains the hematopoietic cell pool by switching the function of p53 under oxidative stress. Cell Metab. 18, 75–85 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Ito, K. et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431, 997–1002 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Maryanovich, M. et al. The ATM-BID pathway regulates quiescence and survival of haematopoietic stem cells. Nat. Cell Biol. 14, 535–541 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Ahlqvist, K. J. et al. Somatic progenitor cell vulnerability to mitochondrial DNA mutagenesis underlies progeroid phenotypes in Polg mutator mice. Cell Metab. 15, 100–109 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Maryanovich, M. et al. An MTCH2 pathway repressing mitochondria metabolism regulates haematopoietic stem cell fate. Nat. Commun. 6, 7901 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Yu, W.-M. et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12, 62–74 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gan, B. et al. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature 468, 701–704 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gurumurthy, S. et al. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature 468, 659–663 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nakada, D., Saunders, T. L. & Morrison, S. J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 468, 653–658 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ema, H. & Nakauchi, H. Expansion of hematopoietic stem cells in the developing liver of a mouse embryo. Blood 95, 2284–2288 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Larsson, N. G. et al. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat. Genet. 18, 231–236 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. Martinez-Reyes, I. et al. TCA cycle and mitochondrial membrane potential are necessary for diverse biological functions. Mol. Cell 61, 199–209 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. McDonnell, E. et al. Lipids reprogram metabolism to become a major carbon source for histone acetylation. Cell Rep. 17, 1463–1472 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Losman, J. A. & Kaelin, W. G. Jr What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev. 27, 836–852 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Xiao, M. et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sullivan, L. B. et al. The proto-oncometabolite fumarate binds glutathione to amplify ROS-dependent signaling. Mol. Cell 51, 236–248 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sciacovelli, M. et al. Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature 537, 544–547 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Engqvist, M. K., Esser, C., Maier, A., Lercher, M. J. & Maurino, V. G. Mitochondrial 2-hydroxyglutarate metabolism. Mitochondrion 19B, 275–281 (2014).

    Article  CAS  Google Scholar 

  33. Oldham, W. M., Clish, C. B., Yang, Y. & Loscalzo, J. Hypoxia-mediated increases in l-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab. 22, 291–303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chowdhury, R. et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12, 463–469 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chandel, N. S. Evolution of mitochondria as signaling organelles. Cell Metab. 22, 204–206 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Tormos, K. V. et al. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 14, 537–544 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Owusu-Ansah, E. & Banerjee, U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature 461, 537–541 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, J. et al. UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells. EMBO J. 30, 4860–4873 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mullen, A. R. et al. Oxidation of α-ketoglutarate is required for reductive carboxylation in cancer cells with mitochondrial defects. Cell Rep. 7, 1679–1690 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shim, E. H. et al. L-2-Hydroxyglutarate: an epigenetic modifier and putative oncometabolite in renal cancer. Cancer Discov. 4, 1290–1298 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Weinberg, F. et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl Acad. Sci. USA 107, 8788–8793 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wheaton, W. W. et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. eLife 3, e02242 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Evans, D. R. & Guy, H. I. Mammalian pyrimidine biosynthesis: fresh insights into an ancient pathway. J. Biol. Chem. 279, 33035–33038 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Mullen, A. R. et al. Reductive carboxylation supports growth in tumour cells with defective mitochondria. Nature 481, 385–388 (2012).

    Article  CAS  Google Scholar 

  48. Liu, X. et al. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nat. Cell Biol. http://dx.doi.org/10.1038/ncb3527 (2017).

  49. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  52. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lu, W. et al. Metabolomic analysis via reversed-phase ion-pairing liquid chromatography coupled to a stand alone orbitrap mass spectrometer. Anal. Chem. 82, 3212–3221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Garcia, B. A. et al. Chemical derivatization of histones for facilitated analysis by mass spectrometry. Nat. Protoc. 2, 933–938 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zheng, Y., Tipton, J. D., Thomas, P. M., Kelleher, N. L. & Sweet, S. M. Site-specific human histone H3 methylation stability: fast K4me3 turnover. Proteomics 14, 2190–2199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zheng, Y., Thomas, P. M. & Kelleher, N. L. Measurement of acetylation turnover at distinct lysines in human histones identifies long-lived acetylation sites. Nat. Commun. 4, 2203 (2013).

    Article  PubMed  CAS  Google Scholar 

  57. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the NIH (R35CA197532) to N.S.C., NIH (T32 GM008061) to L.P.D., NIH (T32 T32HL076139) to S.E.W. J.X. is supported by the NIH/NIDDK grants K01DK093543 and R01DK111430 and the Cancer Prevention and Research Institute of Texas (CPRIT) New Investigator award (RR140025). We thank Robert H. Lurie Cancer Center Flow Cytometry facility supported by NCI CCSG P30 CA060553 for their invaluable assistance. Proteomics services were performed by the Northwestern Proteomics Core Facility, generously supported by NCI CCSG P30 CA060553 awarded to the Robert H. Lurie Comprehensive Cancer Center and the National Resource for Translational and Developmental Proteomics supported by P41 GM108569.

Author information

Authors and Affiliations

Authors

Contributions

E.A., S.E.W. and L.P.D. carried out most of the experiments in the paper. B.J.T. and S.M. provided technical expertise and carried out some of the initial experiments. X.L., Y.Z. and Z.S. performed RNA sequence analysis. M.S. and K.M.M. conducted and analysed metabolomics. P.T.S. generated the RISP-KO mice. E.A., S.E.W., J.X., L.P.D., J.D.C. and N.S.C. provided intellectual input and wrote the paper.

Corresponding author

Correspondence to Navdeep S. Chandel.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Loss of TFAM in hematopoietic cells results in perinatal death due to severe fetal anemia.

(a) Genotypes from crosses between Tfamfl/fl (TFAM WT) and Tfamfl/fl Vav-iCre (TFAM KO) mice. (b) Protein Expression of TFAM in WT, Heterozygous (HET) and KO lineage negative (Lin) FL cells analyzed by Western blot. Experiment performed 3 times and figure displayed is one representative experiment. Image of the whole blot is displayed in Supplementary Fig. 8. (c) Pictures at E15.5 and E18.5 days of TFAM WT and KO fetuses. Images are representative of 10 animals of each genotype isolated on at least three separate days. (d) Total cell number per FL at E15.5 days in TFAM WT and KO fetuses (WT n = 17; HET n = 10; KO n = 10). (e) Peripheral blood cell parameters at E18.5 days fetuses, including RBC (red blood cells), HB (hemoglobin), HCT (hematocrit), WBC (white blood cells) and PLT (platelets) in TFAM WT and KO fetuses. (WT n = 11; HET n = 4; KO n = 8). All data in this figure represent mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test.

Supplementary Figure 2 Fetal HSC and progenitors gating strategy.

(a) Within the lineage negative population: LSKs were defined as Sca-1high, CD48low. Within the LSK population: HSCs (hematopoietic stem cells) were defined as Mac-1high, CD150high, and MPPs (Multipotent progenitors) were defined as Mac-1low, CD150low. (b) Within the lineage negative population: MPs (myeloid progenitors) were defined as c-kithigh, Sca-1low. Within the MP population: MEPs (Megakaryocyte-erythroid progenitors) were defined as CD34low, FcγRIIIlow, CMPs (common myeloid progenitors) were defined as CD34high, FcγRIIIlow, and GMPs (granulocyte-monocyte progenitors) were defined as CD34high, FcγRIIIhigh. (c) Fetal liver hematopoiesis populations described in manuscript and gating definitions.

Supplementary Figure 3 Defective hematopoiesis due to loss of RISP is independent of ROS.

(a) MitoSOX mean fluorescence intensity (MFI) of RISP-WT FL and RISP-KO FL MPs. Data are shown as relative to WT (WT n = 9; KO n = 8). (b) TMRE MFI of RISP-WT FL and RISP-KO FL MPs. Results are CCCP corrected. Data are shown as relative to WT (WT n = 9; KO n = 7). (c) Cell frequency and cell number of erythroid populations defined by staining against surface markers CD71 and Ter119 in RISP-WT FL and RISP-KO FL mice treated with NAC (1 mg ml−1) in vivo (WT n = 8; KO n = 5). (d) Quantification of CFU-GM, BFU-E and CFU-GEMM colonies from Methocult cultures of 105 RISP WT or KO FL cells treated with NAC (+) in vivo (1 mg ml−1) and in vitro (500 μM) (WT n = 8; KO n = 5). (e) Cell frequency and cell number of erythroid populations defined by staining against surface markers CD71 and Ter119 in RISP-WT FL and RISP-KO FL mice treated with BSO (2 mM) in vivo (WT n = 7; KO n = 4). (f) Quantification of CFU-GM, BFU-E and CFU-GEMM colonies from Methocult cultures of 105 FL cells from RISP WT or KO fetuses treated with BSO in vivo (2 mM) and in vitro (500 μM) (WT n = 7; KO n = 4). (g) Quantitative RT-PCR analysis of p16INK4a in RISP WT and KO FL Lin cells, and WT FL Lin cells treated with different doses of BSO for two days. Results are expressed as relative fold change of WT untreated cells. Experiment was performed twice and three experimental replicates were done per experiment. (WT n = 3; KO n = 3; Different BSO doses on WT cells n = 3). (h) Cell frequency and cell number of erythroid populations defined by staining against surface markers CD71 and Ter119 in RISP-WT FL and RISP-KO FL mice treated with BSO (20 mM) in vivo (WT n = 6; KO n = 4). All experiments were done with FL cells from E15.5 day fetuses. All data in this figure represent mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test.

Supplementary Figure 4 Ablation of RISP in the HSCs impaired long-term repopulation activity and competitive repopulation capacity.

(A) Design of the competitive repopulation assay. (B) The contribution of CD45.2 FL donor cells to reconstitute the peripheral blood of previously irradiated CD45.1 recipient mice in competitive transplantation assays. (C) The contribution of FL donor cells to peripheral multilineage reconstitution including B-cell lineage (B220+), T-cell lineage (CD3+) and myeloid lineage (Mac-1+, Gr-1+). Data represent the average contribution for n = 6 recipient mice per genotype in Supplementary Fig. 4b, c. Data presented in Supplementary Fig. 4b, c are mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All p values calculated using Student’s t-test. (d) The total contribution of CD45.2 fetal liver (FL) donor cells to the peripheral blood of irradiated CD45.1 recipient mice in a competitive transplantation assay following engraftment and subsequent Poly (I:C) treatment. Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. (e) The contribution of CD45.2 FL donor cells to specific lineages in the peripheral blood of irradiated CD45.1 recipient mice in a competitive transplantation assay following engraftment and subsequent Poly (I:C) treatment. B-cell lineage (B220+), T-cell lineage (CD3+) and myeloid lineage (Mac-1+, Gr-1+). Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. Data represent the average contribution for Rispfl/fl (RISP-WT FL; n = 9 mice) and Rispfl/fl Mx1-Cre (RISP-KO FL; n = 6 mice) in Supplementary Fig. 4D and S4E. Data in Supplementary Fig. 4D and S4E are mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test.

Supplementary Figure 5 Metabolite levels in RISP deficient fetal liver lineage negative cells.

Relative abundance of glycolysis (a), TCA cycle (b), urea cycle metabolites (c), redox (d), nucleotides (e) and amino acids (f) extracted from RISP-WT FL and-KO FL Lin FL cells. Peak areas of each metabolite were normalized to cell number. Metabolites were measured from WT n = 7 mice; KO n = 5 mice. Raw metabolite data is available in Supplementary Table 3. Data in this figure represent mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test.

Supplementary Figure 6 RISP deficiency impairs adult HSC competitive repopulation capacity.

(a) The contribution of CD45.2 BM donor cells to specific lineages in the peripheral blood of irradiated CD45.1 recipient mice in a competitive transplantation assay following engraftment and subsequent Poly (I:C) treatment. B-cell lineage (B220+), T-cell lineage (CD3+) and myeloid lineage (Mac-1+, Gr-1+). Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. (b) The contribution of CD45.2 BM donor cells to specific lineages (LSK, ST-HSC, LT-HSC, MPP, MP, CMP, GMP, and MEP) in the bone marrow of irradiated CD45.1 recipient mice in a competitive transplantation assay after engraftment and subsequent Poly (I:C) treatment. Bone marrow was assayed four months post-Poly (I:C) treatment. Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. Data represent the average contribution for Rispfl/fl (RISP-WT BM; n = 9 mice) and Rispfl/fl Mx1-Cre (RISP-KO BM; n = 4 mice) in Supplementary Fig. 7a, b. Data are mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test. Supplementary Fig. 7c, d represent the 2nd independent experiment. (c) The total contribution of CD45.2 BM donor cells to the peripheral blood of irradiated CD45.1 recipient mice in a competitive transplantation assay following engraftment and subsequent Poly (I:C) treatment. Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. (d) The contribution of CD45.2 BM donor cells to specific lineages in the peripheral blood of irradiated CD45.1 recipient mice in a competitive transplantation assay following engraftment and subsequent Poly (I:C) treatment. B-cell lineage (B220+), T-cell lineage (CD3+) and myeloid lineage (Mac-1+, Gr-1+). Data represent the percentage of CD45.2 donor cells relative to the initial chimerism. Data represent the average contribution for Rispfl/fl (RISP-WT BM; n = 10 mice) and Rispfl/fl Mx1-Cre (RISP-KO BM; n = 9 mice) in Supplementary Fig. 7c, d. Data are mean ± s.e.m. P ≤ 0.05; P ≤ 0.01. All P values calculated using Student’s t-test.

Supplementary Figure 7 Adult HSC and progenitors gating strategy.

(a) Within the lineage negative population: MPs (myeloid progenitors) were defined as c-kithigh, Sca-1low. Within the MP population: MEPs (Megakaryocyte-erythroid progenitors) were defined as CD34low, FcγRlow, CMPs (common myeloid progenitors) were defined as CD34high, FcγRlow, and GMPs (granulocyte-monocyte progenitors) were defined as CD34high, FcγRhigh. (b) Within the lineage negative population: LSKs were defined as Sca-1high, c-kithigh. Within the LSK population: ST-HSCs (short-term hematopoietic stem cells) were defined as CD48low, CD150low, LT-HSCs (long-term hematopoietic stem cells) were defined as CD48low, CD150high, and MPPs (Multipotent progenitors) were defined as CD48high, CD150low. (c) Adult bone marrow hematopoiesis populations studied, and gating definitions.

Supplementary Figure 8 Full western blot images.

The original western blot images with ladder. Cropped sections are shown in Fig. 1b and Supplementary Fig. 1b.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2505 kb)

Supplementary Table 1

Supplementary Information (XLSX 11 kb)

Supplementary Table 2

Supplementary Information (XLSX 10 kb)

Supplementary Table 3

Supplementary Information (XLSX 69 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ansó, E., Weinberg, S., Diebold, L. et al. The mitochondrial respiratory chain is essential for haematopoietic stem cell function. Nat Cell Biol 19, 614–625 (2017). https://doi.org/10.1038/ncb3529

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3529

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing