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:

Arteriolar niches maintain haematopoietic stem cell quiescence

Abstract

Cell cycle quiescence is a critical feature contributing to haematopoietic stem cell (HSC) maintenance. Although various candidate stromal cells have been identified as potential HSC niches, the spatial localization of quiescent HSCs in the bone marrow remains unclear. Here, using a novel approach that combines whole-mount confocal immunofluorescence imaging techniques and computational modelling to analyse significant three-dimensional associations in the mouse bone marrow among vascular structures, stromal cells and HSCs, we show that quiescent HSCs associate specifically with small arterioles that are preferentially found in endosteal bone marrow. These arterioles are ensheathed exclusively by rare NG2 (also known as CSPG4)+ pericytes, distinct from sinusoid-associated leptin receptor (LEPR)+ cells. Pharmacological or genetic activation of the HSC cell cycle alters the distribution of HSCs from NG2+ periarteriolar niches to LEPR+ perisinusoidal niches. Conditional depletion of NG2+ cells induces HSC cycling and reduces functional long-term repopulating HSCs in the bone marrow. These results thus indicate that arteriolar niches are indispensable for maintaining 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: Spatial relationships between HSCs and the bone marrow vasculature.
Figure 2: Nestin+ cell subsets define distinct vascular structures.
Figure 3: Quiescent arteriolar niche cells are protected from myeloablation.
Figure 4: Spatial relationship between arterioles and quiescent HSCs.
Figure 5: NG2+ periarteriolar cells are an essential constituent of the HSC niche promoting HSC quiescence.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Data deposits

The RNA sequencing data have been deposited in Gene Expression Omnibus under accession number GSE48764.

References

  1. 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 

  2. Li, L. & Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science 327, 542–545 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hsu, Y. C., Pasolli, H. A. & Fuchs, E. Dynamics between stem cells, niche, and progeny in the hair follicle. Cell 144, 92–105 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Orford, K. W. & Scadden, D. T. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nature Rev. Genet. 9, 115–128 (2008)

    Article  CAS  PubMed  Google Scholar 

  5. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1 . Science 287, 1804–1808 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Ito, K. et al. PML targeting eradicates quiescent leukaemia-initiating cells. Nature 453, 1072–1078 (2008)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Essers, M. A. et al. IFNα activates dormant haematopoietic stem cells in vivo . Nature 458, 904–908 (2009)

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Sato, T. et al. Interferon regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferon-dependent exhaustion. Nature Med. 15, 696–700 (2009)

    Article  CAS  PubMed  Google Scholar 

  9. Passegué, E., Wagers, A. J., Giuriato, S., Anderson, W. C. & Weissman, I. L. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J. Exp. Med. 202, 1599–1611 (2005)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008)

    Article  CAS  PubMed  Google Scholar 

  11. Baldridge, M. T., King, K. Y., Boles, N. C., Weksberg, D. C. & Goodell, M. A. Quiescent haematopoietic stem cells are activated by IFN-γ in response to chronic infection. Nature 465, 793–797 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836–841 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Sugimura, R. et al. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell 150, 351–365 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Arai, F. et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118, 149–161 (2004)

    Article  CAS  PubMed  Google Scholar 

  16. Nilsson, S. K. et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood 106, 1232–1239 (2005)

    Article  CAS  PubMed  Google Scholar 

  17. Stier, S. et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J. Exp. Med. 201, 1781–1791 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kiel, M. J., Yilmaz, O. H., Iwashita, T., Terhorst, C. & Morrison, S. J. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121 (2005)

    Article  CAS  PubMed  Google Scholar 

  19. Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 25, 977–988 (2006)

    Article  CAS  PubMed  Google Scholar 

  20. Omatsu, Y. et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 33, 387–399 (2010)

    Article  CAS  PubMed  Google Scholar 

  21. Méndez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010)

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  22. Ding, L., Saunders, T. L., Enikolopov, G. & Morrison, S. J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ema, H. & Suda, T. Two anatomically distinct niches regulate stem cell activity. Blood 120, 2174–2181 (2012)

    Article  CAS  PubMed  Google Scholar 

  24. Hooper, A. T. et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 4, 263–274 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nombela-Arrieta, C. et al. Quantitative imaging of haematopoietic stem and progenitor cell localization and hypoxic status in the bone marrow microenvironment. Nature Cell Biol. 15, 533–543 (2013)

    Article  CAS  PubMed  Google Scholar 

  26. Li, X. M., Hu, Z., Jorgenson, M. L. & Slayton, W. B. High levels of acetylated low-density lipoprotein uptake and low tyrosine kinase with immunoglobulin and epidermal growth factor homology domains-2 (Tie2) promoter activity distinguish sinusoids from other vessel types in murine bone marrow. Circulation 120, 1910–1918 (2009)

    Article  CAS  PubMed  Google Scholar 

  27. Shen, Z., Lu, Z., Chhatbar, P. Y., O'Herron, P. & Kara, P. An artery-specific fluorescent dye for studying neurovascular coupling. Nature Methods 9, 273–276 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Takaku, T. et al. Hematopoiesis in 3 dimensions: human and murine bone marrow architecture visualized by confocal microscopy. Blood 116, e41–e55 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Naveiras, O. et al. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature 460, 259–263 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Katayama, Y. et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 124, 407–421 (2006)

    Article  CAS  PubMed  Google Scholar 

  31. Méndez-Ferrer, S., Lucas, D., Battista, M. & Frenette, P. S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447 (2008)

    Article  ADS  PubMed  CAS  Google Scholar 

  32. Yamazaki, S. et al. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 147, 1146–1158 (2011)

    Article  CAS  PubMed  Google Scholar 

  33. Bhattacharya, D. et al. Niche recycling through division-independent egress of hematopoietic stem cells. J. Exp. Med. 206, 2837–2850 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kalajzic, Z. et al. Directing the expression of a green fluorescent protein transgene in differentiated osteoblasts: comparison between rat type I collagen and rat osteocalcin promoters. Bone 31, 654–660 (2002)

    Article  CAS  PubMed  Google Scholar 

  35. Liu, F. et al. Csf3r mutations in mice confer a strong clonal HSC advantage via activation of Stat5. J. Clin. Invest. 118, 946–955 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Greenbaum, A. M. & Link, D. C. Mechanisms of G-CSF-mediated hematopoietic stem and progenitor mobilization. Leukemia 25, 211–217 (2011)

    Article  CAS  PubMed  Google Scholar 

  37. Birbrair, A. et al. Skeletal muscle pericyte subtypes differ in their differentiation potential. Stem Cell Res. 10, 67–84 (2013)

    Article  CAS  PubMed  Google Scholar 

  38. Zhu, X. et al. Age-dependent fate and lineage restriction of single NG2 cells. Development 138, 745–753 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chi, J. T. et al. Endothelial cell diversity revealed by global expression profiling. Proc. Natl Acad. Sci. USA 100, 10623–10628 (2003)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Frenette, P. S., Pinho, S., Lucas, D. & Scheiermann, C. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu. Rev. Immunol. 31, 285–316 (2013)

    Article  PubMed  Google Scholar 

  41. Greenbaum, A. et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature 495, 227–230 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ding, L. & Morrison, S. J. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495, 231–235 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sipkins, D. A. et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 435, 969–973 (2005)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Winkler, I. G. et al. Vascular niche E-selectin regulates hematopoietic stem cell dormancy, self renewal and chemoresistance. Nature Med. 18, 1651–1657 (2012)

    Article  CAS  PubMed  Google Scholar 

  45. Mignone, J. L., Kukekov, V., Chiang, A. S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004)

    Article  CAS  PubMed  Google Scholar 

  46. Kawamoto, T. Use of a new adhesive film for the preparation of multi-purpose fresh-frozen sections from hard tissues, whole-animals, insects and plants. Arch. Histol. Cytol. 66, 123–143 (2003)

    Article  PubMed  Google Scholar 

  47. Chiang, E. Y., Hidalgo, A., Chang, J. & Frenette, P. S. Imaging receptor microdomains on leukocyte subsets in live mice. Nature Methods 4, 219–222 (2007)

    Article  CAS  PubMed  Google Scholar 

  48. Miller, C. L., Dykstra, B. & Eaves, C. J. Characterization of mouse hematopoietic stem and progenitor cells. Curr. Protoc. Immunol. 22, 22B.2.1–22B.2.31 (2008)

    Google Scholar 

Download references

Acknowledgements

We thank P. P. Pandolfi for providing Pml−/− mice and D. Rowe, J. Butler and S. Rafii for providing Col2.3–GFP mice. We are grateful to L. Tesfa and O. Uche for technical assistance with sorting. S.P. is supported by a New York Stem Cell Foundation-Druckenmiller Fellowship and C.S. by the German Research Foundation (DFG) (Emmy-Noether-Program). J.A. was supported by Training Grant T32 063754. This work was enabled by the National Institutes of Health (R01 grants DK056638, HL069438, HL097700) to P.S.F.

Author information

Authors and Affiliations

Authors

Contributions

Y.K. and C.S. performed the whole-mount imaging experiments and analysed the data; I.B. and S.P. performed FACS sorting and qPCR analyses; J.A. and A.B. performed computational modelling and statistical analysis of the data; D.Z. analysed NG2-creERTM/iDTR mice; T.M. performed the imaging experiments in bone marrow sections; Q.W. and J.C.M. analysed the RNA-seq data; D.L. performed the LTC-IC assay and characterization of endothelial cells by FACS; K.I. provided mice and interpreted data; P.S.F. initiated and directed the study. Y.K. and P.S.F. wrote the manuscript. All of the authors contributed to the design of experiments, discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Paul S. Frenette.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Whole-mount immunofluorescence imaging techniques of the sternal and long-bone bone marrow using in vivo immunofluorescence staining of vessels.

a, Whole-mount immunofluorescence techniques of the sternal bone marrow. Tissue preparation and representative images of the vasculature and haematopoietic cells. b, Analysis of the localization of sinusoids, arterioles and HSCs in the femoral bone marrow of transverse-shaved whole-mount immunofluorescence images. The central vein was identified and localization of sinusoids, arterioles and HSCs were plotted on the axis between central vein and bone as a ratio from 0 to 1. c, Representative FACS plots of bone marrow CD45 Ter119 stromal cells of three independent experiments. Anti-Sca-1 antibody administered intravenously stains a fraction of CD31+ endothelial cells whereas CD31 cells are not stained. d, e, Haematopoietic (d) or Nes–GFPbright mesenchymal (e) progenitor cells are not stained by intravenously injected anti-Sca-1 antibody. f, Average distances between individual sinusoidal vessels in the femoral bone marrow. n = 6 mice.

Extended Data Figure 2 Identification of bone marrow arterioles.

a, FACS plots of bone marrow endothelial cells. Bone marrow endothelial cells are identified as a VEGFR2+ CD31+ population. Representative data of 3 mice. 90% of bone marrow endothelial cells are VEGFR2+ VEGFR3+ Sca-1lo (sinusoidal) and 10% are VEGFR2+ VEGFR3 Sca-1hi (arteriolar). b, Whole-mount images of femoral bone marrow from TIE2–GFP mice stained with anti-VEGFR3, anti-Sca-1, anti-VE-cadherin and anti-PECAM1 antibodies. Scale bar, 25 μm. c, Whole-mount images of the sternal bone marrow stained with Alexa Fluor 633 and Dil-Ac-LDL in vivo. Alexa Fluor 633+ arteriolar vessels do not take up Dil-Ac-LDL. Scale bar, 50 μm. d, e, Whole-mount images of sternal (d) and transverse-shaved femoral (e) bone marrow from Nes–GFP mice stained with Alexa Fluor 633 in vivo (d, e) and anti-PECAM1, anti-VE-cadherin antibodies (e). Alexa Fluor 633 specifically stains vessels accompanied by Nes–GFPbright cells (arterioles). Scale bar, 50 μm. f, Intravital imaging of the mouse calvarial bone marrow stained with intravenously injected Rhodamine 6G and Alexa Fluor 633. Sinusoidal vessels identified by Rhodamine 6G are not stained with Alexa Fluor633. Scale bars, 100 μm.

Extended Data Figure 3 Three-dimensional analysis of sinusoids, arterioles and HSCs by the whole-mount immunofluorescence imaging technique of the bone marrow.

a, Illustrative example of transverse-shaved femoral bone marrow. Arrowheads denote HSCs. Scale bar, 100 μm. b, c, Strategy to identify phenotypic CD150+ CD41 CD48 Lineage HSCs. Megakaryocytes are distinguished by their size and CD41 expression. b, Two representative areas highlighted in dashed squares in Fig. 1f are shown in high magnification. Arrowheads denote HSCs, arrows show CD150+ Lin/CD48/CD41+ cells. Scale bar, 50 μm. c, Three-dimensionally reconstructed images. Grid, 50 μm. d, Estimated HSC number per sternal segment measured by FACS and whole-mount image analysis. e, f, Distances of HSCs to Nes–GFPbright cells, Nes–GFPdim(n = 98 HSCs from 5 mice), arterioles or sinusoids (n = 119 HSCs from 5 mice) shown in absolute numbers (e) and absolute numbers of adjacent HSCs to those structures (f) per sternal segment (75-μm thickness). Similar distribution patterns were obtained when plotting distances of HSCs from Nes–GFPperi cells or arterioles (two-sample Kolmogorov–Smirnov test; P = 0.97), and from Nes–GFPdim cells or sinusoids (two-sample Kolmogorov–Smirnov test; P = 0.45).

Extended Data Figure 4 A null modelling of the spatial relationship between HSCs and arteriolar or sinusoidal vessels.

a, Computational simulation of randomly distributed HSCs on images of whole-mount prepared sterna. To establish the null-model, binary spatial maps of the sinusoids, arterioles (marked by Nes–GFPbright cells, details in Fig. 2a, b and Extended Data Figs 2d, e and 5a–h) were defined from the images of whole-mount prepared sterna. To simulate a null model in which HSCs are not preferentially localized in the marrow, we randomly placed 20 HSCs (to reflect the mean HSCs/sternum observed in situ) on the unoccupied regions of the spatial maps and measured the Euclidean distance of HSCs to the nearest vascular structure. The means of 1,000 simulations defined a distribution of mean distances one would observe for non-preferentially localized HSCs in relation to the respective structures. If the in situ distance measurements were not statistically significantly different from those obtained by a random placement of HSCs on the same structures in silica, this would indicate a non-preferential spatial HSC distribution. The cumulative probability P(X ≤ μ) of observing the in situ mean X was calculated based on the normal distribution, N(μ,σ2 ), with mean (μ) ± s.d. (σ) obtained from our simulation on a map of each bone marrow structure. b, c, Illustrative examples depicting measurements of randomly placed HSCs (b), and distributions of actual (white) and randomly placed (red) HSCs (c) on the image in Fig. 1f.

Extended Data Figure 5 Distinct vascular structures are associated with two types of Nestin+ cells.

a, Quantification of GFP fluorescence intensity of Nes–GFP+ cells by whole-mount imaging of the femoral bone marrow. Two distinct populations, Nes–GFPbright and Nes–GFPdim, can be distinguished based on fluorescence intensity. n = 423 cells from 4 mice. b, The absolute numbers of Nes–GFPbright (Nesperi) and Nes–GFPdim (Nesretic) cells in the femoral bone marrow analysed by FACS. n = 3 mice. cf, Whole-mount images of longitudinally shaved femoral bone marrow from Nes–GFP mice stained with anti-PECAM1, anti-VE-cadherin and anti-Sca-1 antibodies in vivo. Low-power overview (c) and enlarged images (df). Scale bars, 100 μm (c), 50 μm (d), 25 μm (e, f). g, Whole-mount images of sternum from Nes–GFP mice stained with anti-PECAM1 and anti-VE-cadherin, showing morphological differences of arterioles (arrows) and sinusoids (arrowheads). Scale bar, 25 μm. h, Whole-mount images of transverse-shaved femoral bone marrow from Nes–GFP mice, stained with anti-PECAM1 and anti-VE-cadherin in vivo. Scale bar, 10 μm. i, Whole-mount images of the Nes–GFP mouse sternum stained with anti-VE-cadherin and anti-tyrosine hydroxylase (TH) antibodies. Scale bar, 50 μm. j, Quantification of CFU-F content of Nes CD31, Nesretic and Nesperi stromal cells. n = 4 mice per group. k, Quantitative PCR analysis of HSC niche-related genes within Nesperi, Nesretic and Nes–GFP CD31 stromal cells. n = 8, 9, 10, 9, 6 independent experiments from left to right. *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Figure 6 Correlation between cell cycle and localization of HSCs.

a, FACS plots of HSC cell cycle analysed by staining with anti-Ki-67 antibody and Hoechst 33342. Representative data of 5 mice. b, c, Percentages of cells adjacent to Nesperi cells in total or EdU+ HSCs in the long-bone bone marrow (b; n = 244, 52 HSCs from 3 mice) and in Ki-67 or Ki-67+ HSCs in the sternal bone marrow (c; n = 64, 116 HSCs from 7 mice). d, Localization of Ki-67 and Ki-67+ HSCs relative to arterioles in the sternal bone marrow shown as number per sternum segment (75 μm thickness). n = 64, 116 HSCs from 7 mice. e, Distributions of distances of HSCs from arterioles, bone or Col2.3–GFP+ osteoblasts. n = 109, 109, 121 HSCs from 5, 5, 3 mice, respectively. f, Probability distribution of the mean distances from 1,000 simulations of 20 randomly positioned HSCs from Col2.3–GFP+ cells (Fig. 4e). A dashed line depicts the actual mean distance observed in situ. A solid line and dotted lines show the mean ± 2 s.d. (95% confidential interval) calculated by the simulation. The observed mean distance of HSCs was not significantly different from that predicted by the simulation (P = 0.33). g, Localization of total, Ki-67 and Ki-67+ HSCs relative to Col2.3–GFP+ osteoblasts shown in absolute numbers per sternum segment (75 μm thickness). n = 121, 80, 41 HSCs from 3 mice. h, i, Localization of HSCs relative to Nesperi cells after 5-FU treatment shown in number per sternum segment (75-μm thickness). Distribution of HSCs (h), kinetics of the number of HSCs located within 20 μm from Nesperi cells compared to total HSCs (h, inset) and adjacent cells (i). n = 98, 39, 70, 55, 112 HSCs from 5, 9, 7, 4, 4 mice per control, day 3, day 7, day 14 and day 21 groups, respectively.

Extended Data Figure 7 Induction of HSC cell cycle alters their localization.

a, FACS analysis for HSC (CD150+ CD48 Sca-1+ c-kit+ Lineage gated) cell cycle by using Ki-67 and Hoechst 33342 staining after Poly(I:C) injection. n = 4, 6 mice for control and treatment groups, respectively. b, HSC localization relative to Nesperi cells after Poly(I:C) treatment. n = 106, 123 HSCs from 9, 4 mice for control and treatment groups, respectively. Two-sample Kolmogorov–Smirnov test; P = 0.007. c, Altered distances of HSCs from arterioles in Pml−/− sternal bone marrow. n = 118, 146 HSCs from 5, 4 mice for wild-type control and Pml−/− groups, respectively. Wild type/Pml−/−: 32.9 ± 5.4%/11.5 ± 4.2% in 0–20-μm proximity, 25.4 ± 3.4%/51.1 ± 6.8% > 80-μm distance. Two-sample Kolmogorov–Smirnov test; P = 1.9 × 10−6. d, e, Quantification of distances of Ki-67 quiescent (d; n = 68, 73 HSCs from 5, 4 mice per wild-type control and Pml−/− groups, respectively) and Ki-67+ non-quiescent (e; n = 53, 73 HSCs from 5, 4 mice per wild-type control and Pml−/− groups, respectively). HSCs from arteriolar niches in Pml−/− bone marrow. Two-sample Kolmogorov–Smirnov test; P = 9.6 × 10−8 (d), P = 0.48 (e). f, Localization of HSCs relative to Nesperi cells in the sternal bone marrow after G-CSF treatment. n = 98, 73, 122, 94 HSCs from 6, 3, 4, 5 mice for control, day 1, day 2 and day 4 groups, respectively. Two-sample Kolmogorov–Smirnov test; day 1, P = 0.026; day 2, P = 0.012; day 4, P = 0.23 compared to control. g, Absolute numbers of HSCs that located within 20-μm distance from Nesperi cells in the sternal bone marrow after G-CSF treatment. n = 6, 3, 4, 5 mice for control, day 1, day 2 and day 4 groups, respectively. *P < 0.05, **P < 0.01.

Extended Data Figure 8 Peri-sinusoidal Nesretic cells, not periarteriolar Nesperi, overlap with LEPR+ cells.

ac, Analyses of the bone marrow from Lepr-cre/loxp-tdTomato/Nes–GFP transgenic mice. Whole-mount immunofluorescence images of sternum (a, b) and FACS analysis of the femoral bone marrow (c). Representative data of 3 mice. Scale bars, 50 μm.

Extended Data Figure 9 Periarteriolar Nesperi cells are NG2+ and α-smooth muscle actin+ pericytes.

a, b, Immunofluorescence images of the bone marrow from wild-type mice stained with anti-NG2 antibody in a sectioned femur (a) and a whole-mount sternum (b). cf, Whole-mount immunofluorescence (ce) and FACS (f) analyses of the bone marrow from Nes–GFP mice stained with anti-α-smooth muscle actin (αSMA) (c, d) or anti-NG2 (e, f) antibodies. gi, Analysis of the bone marrow from NG2-creERTM/loxp-tdTomato/Nes–GFP transgenic mice. Whole-mount images of sternal bone marrow (g, i) and recombination efficiency for tdTomato protein expression on Nesperi cells analysed by whole-mount imaging after tamoxifen treatment (h). Arrowheads denote Nesretic cells, arrows show Nesperi cells. n = 6 mice. Scale bars, 50 μm.

Extended Data Figure 10 Periarteriolar NG2+ cells are essential for HSC maintenance in the bone marrow and spleen.

Functional analyses of NG2+ cells using NG2-creERTM/iDTR mice. ac, Quantification of Nesperi cells (a) and vascular volume (b, c), accessed by whole-mount imaging of the sternal bone marrow from NG2-creERTM/iDTR/Nes–GFP mice. Scale bars, 100 μm. n = 3 mice per group. d, e, Cxcl12, Kitl and Angpt1 gene expressions assessed by quantitative PCR in sorted Sca-1hi arteriolar (d) and Sca-1lo sinusoidal (e) endothelial (CD45 Ter119 CD31+) cells after NG2+ cell depletion. n = 4 mice per group. f, HSC localization relative to sinusoids in the sternal bone marrow. n = 69, 71 HSCs from 3, 4 mice per control and NG2-creERTM/iDTR groups, respectively. Two-sample Kolmogorov–Smirnov test, P = 0.29. g, Quantification of bone marrow cellularity, frequency and number of phenotypic CD150+ CD48 Sca-1+ c-kit+ Lineage HSCs in spleen. n = 6 mice per group. h, i, Quantification of long-term reconstituting HSCs by LTC-IC assays. n = 3 mice per group. j, Numbers of total leukocytes and phenotypic CD150+ CD48 Sca-1+ c-kit+ Lineage HSCs in blood. n = 3 mice per group. *P < 0.05, **P < 0.01.

Supplementary information

Supplementary Tables

This file contains Supplementary Tables 1-3. (PDF 815 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kunisaki, Y., Bruns, I., Scheiermann, C. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013). https://doi.org/10.1038/nature12612

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12612

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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