Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Mar;591(7850):438-444.
doi: 10.1038/s41586-021-03298-5. Epub 2021 Feb 24.

A mechanosensitive peri-arteriolar niche for osteogenesis and lymphopoiesis

Affiliations

A mechanosensitive peri-arteriolar niche for osteogenesis and lymphopoiesis

Bo Shen et al. Nature. 2021 Mar.

Abstract

Stromal cells in adult bone marrow that express leptin receptor (LEPR) are a critical source of growth factors, including stem cell factor (SCF), for the maintenance of haematopoietic stem cells and early restricted progenitors1-6. LEPR+ cells are heterogeneous, including skeletal stem cells and osteogenic and adipogenic progenitors7-12, although few markers have been available to distinguish these subsets or to compare their functions. Here we show that expression of an osteogenic growth factor, osteolectin13,14, distinguishes peri-arteriolar LEPR+ cells poised to undergo osteogenesis from peri-sinusoidal LEPR+ cells poised to undergo adipogenesis (but retaining osteogenic potential). Peri-arteriolar LEPR+osteolectin+ cells are rapidly dividing, short-lived osteogenic progenitors that increase in number after fracture and are depleted during ageing. Deletion of Scf from adult osteolectin+ cells did not affect the maintenance of haematopoietic stem cells or most restricted progenitors but depleted common lymphoid progenitors, impairing lymphopoiesis, bacterial clearance, and survival after acute bacterial infection. Peri-arteriolar osteolectin+ cell maintenance required mechanical stimulation. Voluntary running increased, whereas hindlimb unloading decreased, the frequencies of peri-arteriolar osteolectin+ cells and common lymphoid progenitors. Deletion of the mechanosensitive ion channel PIEZO1 from osteolectin+ cells depleted osteolectin+ cells and common lymphoid progenitors. These results show that a peri-arteriolar niche for osteogenesis and lymphopoiesis in bone marrow is maintained by mechanical stimulation and depleted during ageing.

PubMed Disclaimer

Conflict of interest statement

COMPETING INTERESTS

The authors declare no competing interests.

Figures

Extended Data Figure 1.
Extended Data Figure 1.. OlnmT reporter mice showed that Osteolectin is expressed by osteoblasts, osteocytes, and chondrocytes in addition to peri-arteriolar LepR+ stromal cells.
(a-c) The mouse Osteolectin gene was modified to insert an mTomato-WPRE-pA cassette between the 5’ untranslated region and exon 3 to generate the targeting vector. These sites were selected to avoid disrupting conserved intronic sequences. Open boxes indicate untranslated regions and black boxes indicate translated regions of Osteolectin. The targeted founder mouse (F0) was identified by southern blotting (b) using 5’ and WPRE probes (black bars). (c) PCR genotyping of genomic DNA confirmed germline transmission of the OlnmT allele. Mice were backcrossed at least three times onto a C57BL/Ka background before analysis. (d) Osteolectin transcript levels by qRT-PCR in Oln-Tomato+ and Oln-Tomato bone marrow cells (3 mice analyzed in 3 independent experiments). (e) Flow cytometry gates to distinguish Oln-mTomato+LepR+ from Oln-mTomatoLepR+ stromal cells in enzymatically dissociated bone marrow. (f) Osteolectin transcript levels by qRT-PCR in Oln-Tomato+LepR+ and Oln-TomatoLepR+ bone marrow cells (4 mice analyzed in 4 independent experiments). (g) We did not detect Oln-Tomato in VE-cadherin-expressing bone marrow endothelial cells (4 mice per genotype analyzed in 4 independent experiments). (h and i) Femur epiphysis (h) and diaphysis (i) from OlnmT/+; Col1a1*2.3-EGFP mice (images are representative of 3 independent experiments from 8–10-week-old mice). In panel h, the arrowhead points to Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts in trabecular bone and the arrow points to Oln-Tomato+Col1a1*2.3-EGFP hypertrophic chondrocytes in the growth plate (scale bar = 100μm). Panel i shows Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts at the endosteum (arrow), Oln-Tomato+ osteocytes (arrowhead), and Oln-Tomato+ periosteal cells (asterisk) (scale bars = 40μm). All data represent mean ± SD.
Extended Data Figure 2.
Extended Data Figure 2.. Bone marrow Oln-mTomato+ cells localized mainly around arterioles in the diaphysis.
(a) A low magnification view of the femur diaphysis showed Oln-Tomato+ stromal cells associated with arterioles enriched in the center of the marrow. In this image, arteriolar and sinusoidal blood vessels were distinguished based on size, morphology, and continuity of the basal lamina, visualized using anti-laminin antibody staining as previously described (images are representative of 3 independent experiments; scale bar = 200μm). (b) OlnmT/+ femur bone marrow showing that, in contrast to the diaphysis (Fig. 1h), most Sca-1+ arterioles were not surrounded by Oln-Tomato+ stromal cells in the metaphysis. The Oln-Tomato+ cells in this panel were osteoblasts and osteocytes associated with trabecular bone (scale bar = 500μm). (c, d) Gene set enrichment analysis showing significant enrichment of genes associated with osteogenesis in CD45Ter119CD31Scf-GFP+Oln-mTomato+ stromal cells and adipogenesis in CD45Ter119CD31Scf-GFP+Oln-mTomato stromal cells (FDR, false discovery rate; NES, normalized enrichment score; 4 mice analyzed in 4 independent experiments). (e-f) The mouse Osteolectin gene was modified by inserting an iCreER-WPRE-pA cassette between the 5’ untranslated region and exon 3 to generate the targeting vector. These sites were selected to avoid disrupting conserved intronic sequences. Open boxes indicate untranslated regions and black boxes indicate translated regions of Osteolectin. The targeted founder mouse (F0) was identified by Southern blotting (f) using 5’ and WPRE probes (black bars). (g) PCR genotyping of genomic DNA confirmed germline transmission of the OlniCreER allele. Mice were backcrossed at least three times onto a C57BL/Ka background before analysis. (h) Deep imaging of OlniCreER/+; Rosa26loxp-tdTomato/+ femur bone marrow 3 days after tamoxifen administration at 2 months of age, showing that Oln-Tomato+ cells were exclusively peri-arteriolar in the diaphysis (images are representative of 3 independent experiments; scale bar = 200μm). (i and j) Deep imaging of the femur epiphysis (i) and diaphysis (j) one month after tamoxifen administration at 2 months of age (images are representative of 3 independent experiments). Panel i shows Oln-Tomato+ hypertrophic chondrocytes in the growth plate (arrow) and Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts in trabecular bone (arrowhead; scale bar = 60mm). Panel j shows Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts at the endosteum (arrow), an Oln-Tomato+ osteocyte (arrowhead), and Oln-Tomato+ periosteal cells (asterisk; scale bars = 30μm). (k) Col1a1-CreER; Rosa26loxp-tdTomato/+; Col1a1*2.3-EGFP mice were treated with tamoxifen at 2 months of age and the percentage of Col1a1*2.3-EGFP+ osteoblasts that were Tomato+ was assessed 3 days to 1 month later (3 mice per time point analyzed in 3 independent experiments). All data represent mean ± SD.
Extended Data Figure 3.
Extended Data Figure 3.. Osteolectin+ cells create a niche for CLPs but not other hematopoietic stem/progenitor cells.
(a) 2-month-old OlniCreER/+; Rosa26loxp-tdTomato/+ mice were sublethally irradiated 3 days after tamoxifen administration. Two weeks later, none of the adipocytes in the bone marrow were Tomato+ (image representative of 3 independent experiments; scale bar = 60μm). (b) Representative H&E stained sections from ossicles formed by Oln-Tomato+ (left) or Oln-Tomato (right) stromal cells sorted from OlnmT/+ mice showing bone (arrowheads), hematopoietic cells (arrow), and adipocytes (asterisk; images are representative of 5 independent experiments; scale bar = 100μm). (c-e) OlniCreER/+; Scffl/fl and Scffl/fl littermate controls were treated with tamoxifen at 2-months of age. One month later, blood from OlniCreER/+; Scffl/fl mice showed normal white blood cell (c), red blood cell (d) and platelet (e) counts (6 mice per genotype analyzed in 3 independent experiments). (f-k) OlniCreER/+; Scffl/fl mice and littermate controls exhibited no significant differences in the frequencies of B220+ B cells (f), CD3+ T cells (g), Gr-1+Mac-1+ myeloid cells (h), CD41+ megakaryocyte lineage cells (i), CD71+/Ter119+ erythroid lineage cells (j), HSCs or MPPs in the spleen (k; 6 mice per genotype analyzed in 3 independent experiments). (l) Bone marrow cells from OlniCreER/+; Scffl/fl mice and littermate controls gave similar levels of donor cell reconstitution upon competitive transplantation into irradiated mice (5 donor mice per genotype analyzed in 3 independent experiments). The differences between OlniCreER/+; Scffl/fl and Scffl/fl mice were not statistically significant in c-l. (m) OlnmT/+; ScfGFP/+ femur bone marrow showing Oln-Tomato+ osteoblasts at the endosteum were negative for Scf-GFP, while peri-arteriolar Oln-Tomato+ stromal cells were positive for Scf-GFP (representative of 3 independent experiments; scale bar = 80μm). (n) OlnmT/+; Col1a1*2.3-EGFP femur bone marrow showing Col1a1*2.3-EGFP+ osteoblasts at the endosteum were Oln-Tomato+ (representative of 3 independent experiments; scale bar = 100μm). All data represent mean ± SD. Statistical significance was assessed using matched samples two-way ANOVAs followed by Sidak’s (c-e, and k-l) or Tukey’s (f-j) multiple comparisons tests.
Extended Data Figure 4.
Extended Data Figure 4.. Flow cytometry gating strategy for the isolation of hematopoietic stem and progenitor cell populations.
(a-d) Representative flow cytometry gates used to identify the hematopoietic stem and progenitor cell populations in the bone marrow (a, b), and T lineage progenitors in the thymus (c, d). The markers used to identify each of the cell populations characterized in this study are listed in Supplementary Table 1. (e) Representative flow cytometry gates showing that more than 50% of IL7Rα+Lineage cells were LinSca1lowc-kitlowIL7Ra+Flt3+ CLPs and most of the non-CLP IL7Rα+Lineage (Flt3) cells were CD19+, likely to be other early B lineage progenitors (4 mice analyzed in 3 independent experiments). All data represent mean ± SD.
Extended Data Figure 5.
Extended Data Figure 5.. Oln-mTomato+ osteoblasts and osteocytes in the metaphysis do not express Scf-GFP and Oln-mTomato+ cells in the diaphysis expand during fracture healing.
The differences among treatments in cell frequencies were also evident in absolute numbers. (a-c) Epiphysis (a) and metaphysis (b, c) of OlnmT/+; ScfGFP/+ femur bone marrow showing that hypertrophic chondrocytes (arrowhead in a; scale bars = 30μm) as well as osteoblasts and osteocytes associated with trabecular bone (arrowhead in c) were negative for Scf-GFP (images are representative of 3 independent experiments; scale bars = 400μm (b) and 100μm (c)). Most of the Oln-Tomato staining in the metaphysis reflects Col1a1*2.3-GFP+ osteoblasts and osteocytes associated with trabecular bone, as shown in Fig. 1d and Extended Data Fig. 2b. The boxed area in panel b is magnified in panel c. (d) Image of the metaphysis of ScfGFP/+ femur bone marrow showing limited non-specific staining by anti-tdTomato antibody (scale bar = 100mm). (e-g) OlniCreER/+; Scffl/fl and Scffl/fl littermate control mice were fed tamoxifen chow from 2–4 months of age. They exhibited no significant differences in the frequencies of HSCs, MPPs, GMPs, MEPs, or CMPs in the bone marrow (e), or B220+ B cells (f), or CD3+ T cells (g) in the bone marrow or spleen. The differences between OlniCreER/+; Scffl/fl and Scffl/fl mice were not statistically significant in e-g (5 mice per genotype analyzed in 3 independent experiments). (h, i) Three days after tamoxifen, femurs were fractured in 2-month-old OlnmT/iCreER; Scffl/fl mice and OlnmT/+; Scffl/fl littermate controls then the bone marrow was analyzed two weeks later. HSC and MPP frequencies did not significantly change during fracture healing. In contrast, CLP frequencies significantly increased in OlnmT/+; Scffl/fl control but not in OlnmT/iCreER; Scffl/fl mice (h). The frequency of Osteolectin+ cells significantly increased in both OlnmT/iCreER; Scffl/fl mice and OlnmT/+; Scffl/fl controls (I; 5 mice per genotype analyzed in 3 independent experiments). (j) Localization of IL7Rα+Lineage lymphoid progenitors in the marrow of the fractured as compared to control (CON) femur (4 mice per treatment analyzed in 4 independent experiments). All data represent mean ± SD. Statistical significance was assessed using matched samples two-way ANOVAs followed by Sidak’s (e-i) or Tukey’s multiple comparisons tests (h), or Cochran-Mantel-Haenszel test (j).
Extended Data Figure 6.
Extended Data Figure 6.. CLPs and peri-arteriolar Osteolectin+ cells are depleted during aging but most other hematopoietic stem and progenitor cell populations are not.
The differences among treatments in cell frequencies were also evident in absolute numbers. (a-d) OlnmT/+ mice received daily subcutaneous injections with PBS or 40 μg/kg human PTH for 28 days. PTH treated mice exhibited significantly thicker cortical bone (a, b) and significant reductions in the frequencies of Osteolectin+ cells (c) and CLPs (d; 8 mice per treatment analyzed in 4 independent experiments). Micro CT images (b, scale bar = 800μm) of cortical bone are representative of 8 independent experiments. (e, f) The frequency of LepR+ cells (e) and the percentage of LepR+ cells that were Oln-mTomato+ (f) in OlnmT/+ bone marrow at 2 to 18 months of age (8 mice per time point analyzed in 4 independent experiments). (g) Femur bone marrow from an 18-month-old OlnmT/+ mouse showing an arteriole surrounded by Oln-mTomato+ cells (arrowhead) and an arteriole lacking Oln-mTomato+ cells (arrow) in the diaphysis (image is representative of 4 independent experiments; scale bar = 100μm). (h) The frequency of CLPs in OlnmT/+ mice at 2 to 18 months of age. (i-m) During aging, the frequencies of HSCs (i) and MPPs (j) in the bone marrow increased while the frequencies of GMPs (k), CMPs (l), and MEPs (m) did not significantly change (8 mice per time point analyzed in 4 independent experiments). (n) Localization of IL7Rα+Lineage cells in the bone marrow of 2- and 18-month old mice (7 mice per time point analyzed in 3 independent experiments). (o) Aging significantly depleted Osteolectin+ cells associated with arterioles in both endosteal and non-endosteal regions of diaphysis bone marrow (8 mice per time point analyzed in 4 independent experiments). (p) Number of IL7Rα+Lineage cells per 100 μm of Osteolectin+ or Osteolectin arteriole at 2 and 18 months of age (7 mice per time point analyzed in 3 independent experiments). All data represent mean ± SD. Statistical significance was assessed using paired t-tests (a, c, d, o, p), one-way ANOVA followed by Dunnett’s multiple comparisons test (e), matched samples two-way ANOVAs followed by Dunnett’s multiple comparisons tests (f, h-m), Cochran-Mantel-Haenszel test (n), or Mann–Whitney tests followed by Holm-Sidak’s multiple comparisons adjustments (o).
Extended Data Figure 7.
Extended Data Figure 7.. OlniCreER/+; Scffl/fl mice exhibited reduced lymphopoiesis and impaired bacterial clearance after Listeria infection.
The same differences that were evident among treatments in the frequencies of cell populations were also evident in absolute numbers. (a-c) OlniCreER/+; Scffl/fl mice and littermate controls were treated with tamoxifen at 2 months of age and then 3 days later were administered Listeria orally. Relative to Scffl/fl littermate controls, OlniCreER/+; Scffl/fl mice had decreased B (a) and T (b) cell counts in the spleen 5 days after Listeria infection and increased bacterial CFUs in the spleen 10 days after infection (c; 11–16 mice per genotype analyzed in 3 independent experiments). (d-k) OlniCreER/+; Scffl/fl and Scffl/fl littermate controls were fed tamoxifen from 2 to 4-months of age then 3 days later were administered Listeria intraperitoneally. Relative to Scffl/fl littermate controls, OlniCreER/+; Scffl/fl mice exhibited reduced frequencies of Pre-proB cells, but not ProB or PreB cells, in the bone marrow (d) as well as ETPs and DN1 cells in the thymus (e) at 5 and/or 10 days after infection. OlniCreER/+; Scffl/fl mice also exhibited reduced numbers of NK cells in the spleen (f) as well as B cells (g), T cells (h), and NK cells (i) in mesenteric lymph nodes (mLN) at 5 and 10 days after infection. (j) OlniCreER/+; Scffl/fl mice exhibited increased bacterial CFUs in mLN at 5 and 10 days after infection. (k) The percentage of CD3+ T cells that were IFN-γ+ did not significantly differ between the spleens of OlniCreER/+; Scffl/fl mice and littermate controls (18–27 mice per genotype analyzed in 3 independent experiments). (l-n) Compared to Scffl/fl controls, OlniCreER/+; Scffl/fl mice had decreased spleen B (l) and T (m) cell counts, and increased spleen bacterial CFUs (n) at 5 and 10 days after infection with Listeria (21–27 mice per genotype analyzed in 3 independent experiments). All data represent mean ± SD. Statistical significance was assessed using matched samples two-way ANOVAs followed by Sidak’s multiple comparisons tests (a, b, d-n) or Wilcoxon matched-pairs test (c).
Extended Data Figure 8.
Extended Data Figure 8.. Mechanical stimulation is required for the maintenance of peri-arteriolar, but not peri-sinusoidal, niches for lymphoid progenitors in the bone marrow.
(a) The effect of voluntary running for 4 weeks on femoral cortical bone mineral density (6 mice per treatment analyzed in 3 independent experiments). (b) Voluntary running for 4 weeks did not significantly affect the frequencies of HSCs, MPPs, GMPs, MEPs, CMPs, or CLPs in calvarium bone marrow (5 mice per treatment analyzed in 3 independent experiments). (c-h) Hindlimb unloading (c) for 2 weeks did not significantly affect forelimb cortical bone mineral density (d), cortical thickness (e), the percentage of LepR+ cells that were Oln-mTomato+ (f), or the frequencies of HSCs, MPPs, GMPs, MEPs, CMPs, or CLPs in humerus bone marrow (g). (h) Hindlimb unloading for 2 weeks did significantly reduce hindlimb (femur) cortical bone mineral density (5 mice per treatment analyzed in 3 independent experiments). (i) All point current amplitude histograms of the electrical recordings in Fig. 4h. The single channel conductance was 15 ± 1 pS from the Gaussian fits to the amplitude histograms. The y-axis shows the number of events. (j, k) Single channel current recordings and corresponding all point current histograms of Piezo1 deficient Oln-mTomato+ cells isolated from OlnmT/iCreER; Piezo1fl/fl mice. The data were collected at −60 mm Hg applied pressure and at the holding potential of +60 mV, without (j) or with (k) 40 μM Yoda1 in the pipette. (l) Piezo1 transcript levels by qRT-PCR in Oln-Tomato+ cells from OlnmT/iCreER; Piezo1fl/fl and OlnmT/+; Piezo1fl/fl control mice (6 mice per genotype analyzed in 3 independent experiments). (m and n) The effect of Piezo1 deletion in Osteolectin+ cells on femoral cortical bone mineral density (m) and the percentage of Osteolectin+ cells that incorporated a 48 hour pulse of BrdU (n; 6 mice per genotype analyzed in 3 independent experiments). (o) Location of IL7Rα+Lineage cells in the bone marrow of OlnmT/iCreER; Piezo1fl/fl and OlnmT/+; Piezo1fl/fl control mice (6 mice per genotype analyzed in 3 independent experiments). (p-s) We treated Col1a1-creER; Piezo1fl/fl; OlnmT/+ mice and littermate controls with tamoxifen at 2 months of age and analyzed them 1 month later. Piezo1 deletion in osteoblasts significantly reduced femur bone mineral density (p) and cortical thickness (q) but not the frequencies of Osteolectin+ cells (r) or HSCs, MPPs, GMPs, MEPs, or CLPs in the bone marrow (s; 5–6 mice per genotype analyzed in 3 independent experiments). (t-w) We analyzed the femurs of Leprcre/+; Piezo1fl/fl; OlnmT/+ mice and littermate controls at 4 months of age and found that Piezo1 deletion in LepR+ cells did not significantly reduce bone mineral density (t) but did reduce cortical thickness (u). Piezo1 deletion in LepR+ cells also significantly reduced the frequencies of Osteolectin+ cells (v) and CLPs (w) without affecting the frequencies of HSCs, MPPs, GMPs, MEPs, or CMPs (w) in the bone marrow (5 mice per genotype analyzed in 3 independent experiments). (x-z) Compared to Scffl/fl littermate controls, OlniCreER/+; Piezo1fl/fl mice had decreased B (x) and T (y) cell counts in the spleen 5 days after oral Listeria infection and increased bacterial CFUs in the spleen 10 days after infection (z; 11–16 mice per genotype analyzed in 3 independent experiments). All data represent mean ± SD. Statistical significance was assessed using paired t-tests (a, l, m, n, p-r, t-v, z), matched samples two-way ANOVAs followed by Sidak’s (b, d-f, h, s, w-y) or Holm-Sidak’s multiple comparisons adjustment (g), or Cochran-Mantel-Haenszel test (o).
Figure 1.
Figure 1.. Osteolectin is expressed by peri-arteriolar LepR+ cells.
Images are representative of 3–5 experiments with 8–10-week-old mice. (a-c) Flow cytometric analysis of enzymatically dissociated bone marrow from OlnmT/+ mice (4 mice per genotype in 4 independent experiments). (b) Most Oln-Tomato+ cells were LepR+. (c) Only a small minority of LepR+ cells were Oln-Tomato+. (d and e) Representative femur sections showing the epiphysis (d) and diaphysis (e) from 2–4-month-old OlnmT/+; Col1a1*2.3-EGFP mice. Panel d shows Oln-Tomato+ hypertrophic chondrocytes (arrow) and Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts in trabecular bone (arrowhead; scale bar = 30μm). Panel e shows Oln-Tomato+Col1a1*2.3-EGFP+ osteoblasts in the endosteum (arrow) and Oln-Tomato+ osteocytes (arrowhead; scale bars = 25μm). (f and g) Deep imaging of OlnmT/+ femur bone marrow. The boxed area in panel f is magnified in panel g. Panel f shows peri-arteriolar Oln-Tomato+ cells (arrowhead) as well as Oln-Tomato+ cells on the endosteal surface (arrow; scale bar = 100μm). Panel g shows Oln-Tomato+LepR+ peri-arteriolar stromal cells (arrowhead) and Oln-TomatoLepR+ peri-sinusoidal stromal cells (arrow; scale bar = 40μm). (h) Oln-Tomato+ cells around Sca-1+ arterioles (arrow) but not Endomucinhigh sinusoids (scale bar = 200μm). All data represent mean ± SD.
Figure 2.
Figure 2.. Osteolectin+ cells are short-lived osteogenic progenitors.
(a) Percentage of CFU-F colonies that were Tomato+ when formed by bone marrow cells from 2 month-old Leprcre/+; Rosa26loxp-tdTomato/+ or OlniCreER/+; Rosa26loxp-tdTomato/+ mice, 3 days after tamoxifen administration (4 independent experiments). (b) Percentage of LepR+, LepR+Oln-mTomato+, and LepR+Oln-mTomato bone marrow stromal cells that formed CFU-F colonies. (c and d) Osteogenic (c) and adipogenic (d) differentiation of adherent cells cultured from LepR+Oln-mTomato+ or LepR+Oln-mTomato stromal cells (6 independent experiments in b-d). (e) The average numbers of perilipin+ adipocytes or alkaline phosphatase+ (ALP+) osteogenic cells that spontaneously differentiated per CFU-F colony after 1 week of culture (4 independent experiments). (f) Percentage of LepR+, LepR+Oln-mTomato+ and LepR+Oln-mTomato bone marrow stromal cells that incorporated a 24-hour pulse of BrdU in vivo (4 independent experiments). (g) OlniCreER/mT; Rosa26loxp-EGFP/+ mice were treated with tamoxifen at 2 months of age and the percentage of Oln-Tomato+ cells that were EGFP+ was assessed (3 independent experiments). (h and i) OlniCreER/+; Rosa26loxp-tdTomato/+; Col1a1*2.3-EGFP mice were treated with tamoxifen at 2 months of age and the percentages of LepR+ stromal cells (h) and Col1a1*2.3-EGFP+ osteoblasts (i) that were Tomato+ was assessed (5 independent experiments). (j) Bone, adipocytes, and hematopoiesis in ossicles formed by CFU-F cultured from LepR+Oln-mTomato+ or LepR+Oln-mTomato stromal cells (80 ossicles per cell population derived from 20 mice in 5 independent experiments). (k-l) Femurs were fractured 3 days after tamoxifen administration to OlniCreER/+; Rosa26loxp-tdTomato/+; Col1a1*2.3-EGFP mice. Two weeks later the number of LepR+ cells per femur (k) and the percentage of LepR+ cells that were Tomato+ (l) were assessed (5 independent experiments). All data represent mean ± SD. Statistical significance was assessed using an unpaired t-test (a), paired t-tests (c, k, l), matched samples one-way ANOVAs followed by Tukey’s multiple comparisons tests (b, f), Wilcoxon test (d), Wilcoxon test followed by Holm-Sidak’s multiple comparisons adjustment (e), or Cochran-Mantel-Haenszel test (j).
Figure 3.
Figure 3.. Osteolectin+ peri-arteriolar stromal cells maintain early lymphoid progenitors by synthesizing SCF.
Images are representative of 3 independent experiments. The same differences that were evident among the frequencies of cell populations were also evident in absolute numbers. (a, b) In femur bone marrow from 2-month-old ScfGFP/+; OlnmT/+ mice, Scf-GFP was expressed by both Oln-mTomato+ peri-arteriolar cells (arrowhead in a) and Oln-mTomato peri-sinusoidal cells (arrow in a; scale bar = 20μm). (c-d) OlniCreER/+; Scffl/fl and Scffl/fl littermate control mice were treated with tamoxifen at 2-months of age then bone marrow and spleen cellularity (c), and hematopoietic stem and progenitor cell frequencies in the bone marrow (d) were analyzed one month later (3 independent experiments). (e, f) Localization of IL7Rα+Lineage lymphoid progenitors (arrows) adjacent to peri-arteriolar Osteolectin+ cells (scale bar = 15μm; 9 mice in 3 independent experiments). (g) Localization of IL7Rα+Lineage cells in the bone marrow of OlniCreER/+; Scffl/fl and Scffl/fl littermate control mice (6 mice per genotype in 3 independent experiments). (h-j) OlniCreER/+; Scffl/fl mice and littermate controls were fed tamoxifen from 2–4 months of age and the bone marrow (h) and thymus (i, j) were analyzed (3 independent experiments). (k and l) Compared to Scffl/fl controls, OlniCreER/+; Scffl/fl mice had decreased CLP frequency in the bone marrow (k) and worse survival (l) after Listeria infection (17–27 mice per genotype in 3 independent experiments). All data represent mean ± SD. Statistical significance was assessed using matched samples two-way ANOVAs followed by Sidak’s multiple comparisons tests (c, d, h-k), Cochran-Mantel-Haenszel tests (f, g), or Mantel-Cox test (l).
Figure 4.
Figure 4.. Mechanical stimulation is required for the maintenance of peri-arteriolar niches for lymphoid progenitors in the bone marrow.
The same differences that were evident among the frequencies of cell populations were also evident in absolute numbers. (a-c) The effect of voluntary running for 4 weeks on femoral cortical thickness (a), the percentage of LepR+ cells that were Oln-mTomato+ (b), and the frequencies of hematopoietic stem and progenitor cells in the bone marrow (c; 6–10 mice per treatment in 3–5 independent experiments). (d-g) The effect of hindlimb unloading for 2 weeks on femur cortical thickness (d), the percentage of LepR+ cells that were Oln-mTomato+ (e), the frequencies of stem and progenitors cells in the bone marrow (f), and the percentage of Osteolectin+ cells that incorporated a 48 hour pulse of BrdU (g; 5 mice per treatment in 3 independent experiments). (h) Single channel current recordings in Oln-mTomato+ cells at the indicated applied pressures. O and C denote the open and closed states of the channels. (i) Normalized open probability–pressure relationship from Oln-mTomato+ cells recorded with (blue) or without (white) 40 μM Yoda1 in the patch pipette (6 mice in 6 independent experiments). (j) Yoda1 mediated Ca++ response in Oln-mTomato+ cells isolated from OlnmT/iCreER; Piezo1fl/fl or OlnmT/+; Piezo1fl/fl littermate control mice (5 mice per genotype in 3 independent experiments). (k) Western blot of Osteolectin+ cells from OlnmT/iCreER; Piezo1fl/fl or OlnmT/+; Piezo1fl/fl mice (representative of three independent experiments). (l-n) The effect of Piezo1 deletion in Osteolectin+ cells on cortical thickness (l), the percentage of LepR+ cells that were Oln-mTomato+ (m), and the frequencies of stem and progenitor cells in bone marrow (n; 6 mice per genotype in 3 independent experiments). All data represent mean ± SD. Statistical significance was assessed using paired t-tests (a, b, g, l, m) and matched samples two-way ANOVAs followed by Sidak’s (c-e, j, n) or Holm-Sidak’s (f) multiple comparisons adjustments.

Comment in

Similar articles

Cited by

References

    1. Ding L, Saunders TL, Enikolopov G & Morrison SJ Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462, (2012). - PMC - PubMed
    1. Ding L & Morrison SJ Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495, 231–235, (2013). - PMC - PubMed
    1. Oguro H, Ding L & Morrison SJ SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell 13, 102–116, (2013). - PMC - PubMed
    1. Cordeiro Gomes A et al. Hematopoietic Stem Cell Niches Produce Lineage-Instructive Signals to Control Multipotent Progenitor Differentiation. Immunity 45, 1219–1231, (2016). - PMC - PubMed
    1. Himburg HA et al. Distinct Bone Marrow Sources of Pleiotrophin Control Hematopoietic Stem Cell Maintenance and Regeneration. Cell Stem Cell 23, 370–381 e375, (2018). - PMC - PubMed

Publication types

LinkOut - more resources