Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation - PubMed (original) (raw)

Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation

Matthew J Christopher et al. J Bone Miner Res. 2008 Nov.

Abstract

Long-term treatment of mice or humans with granulocyte colony-stimulating factor (G-CSF) is associated with a clinically significant osteopenia characterized by increased osteoclast activity and number. In addition, recent reports have observed a decrease in number of mature osteoblasts during G-CSF administration. However, neither the extent of G-CSF's suppressive effect on the osteoblast compartment nor its mechanisms are well understood. Herein, we show that short-term G-CSF treatment in mice leads to decreased numbers of endosteal and trabecular osteoblasts. The effect is specific to mature osteoblasts, because bone-lining cells, osteocytes, and periosteal osteoblasts are unaffected. G-CSF treatment accelerates osteoblast turnover in the bone marrow by inducing osteoblast apoptosis. In addition, whereas G-CSF treatment sharply increases osteoprogenitor number, differentiation of mature osteoblasts is impaired. Bone marrow transplantation studies show that G-CSF acts through a hematopoietic intermediary to suppress osteoblasts. Finally, G-CSF treatment, through suppression of mature osteoblasts, also leads to a marked decrease in osteoprotegerin expression in the bone marrow, whereas expression of RANKL remains relatively constant, suggesting a novel mechanism contributing to the increased osteoclastogenesis seen with long-term G-CSF treatment. In sum, these findings suggest that the hematopoietic system may play a novel role in regulating osteoblast differentiation and apoptosis during G-CSF treatment.

PubMed Disclaimer

Figures

FIG. 1

FIG. 1

Loss of osteoblast number and function during G-CSF treatment. (A) Mice were treated with G-CSF (250 μg/kg/d) for 5 days, and osteoblast surface per bone surface (Ob.S/BS) was determined. (B) Immunohistochemistry showing GFP+ (brown) osteoblasts (arrows), bone-lining cells (arrowheads), and osteocytes in untreated or day 5 G-CSF treated pOBCol2.3-GFP transgenic mouse femurs. Insets show enlargement of area enclosed by dotted line. Original magnification ×100. (C) Quantification of mature osteoblasts, bone-lining cells, and osteocytes in transgenic mice treated with G-CSF for 5 days or untreated (n = 4 each group). (D) Representative scatter plots showing GFP expression (bottom panels) in the stromal (CD45 negative, Ter119 negative) cell population (top panels) isolated from nontransgenic and pOBCol2.3-GFP mice (left and right, respectively). (E) Number of GFP+ cells recovered from the femurs of transgenic mice after treatment with G-CSF (n = 2–10 each time point). Data represent the mean ± SE. a p < 0.01 vs. all other groups; b p < 0.05.

FIG. 2

FIG. 2

Loss of endosteal and trabecular, but not periosteal, osteoblast activity during G-CSF treatment. Osteoid and mineralization were measured in untreated mice or mice treated for 7 days with G-CSF (n = 2–3 each group). (A) Percent osteoid surface was calculated in Masson trichrome–stained tibial sections from untreated and treated wildtype mice. (B) Mineral apposition rate (MAR) and percent mineralizing surface (Md.S/BS) were calculated on endosteal and periosteal surfaces from calcein-labeled calvaria. (C) Osteocalcin RNA in situ hybridization of long bones harvested from untreated mice or mice treated for 5 days with G-CSF. Shown are representative photomicrographs of three independent experiments. Periosteal surfaces (arrows), endosteal surfaces (arrowheads), bone (B), and bone marrow (BM) are indicated. Original magnification ×100. Data represent the mean ± SE. a p = 0.057; b p = 0.10.

FIG. 3

FIG. 3

G-CSF receptor–deficient bone marrow chimeras. (A) G-CSFR−/− CD45+ cKit+ lineage− hematopoietic cells (KL) cells were transplanted into wildtype recipients (n = 4–5 each group). After hematopoietic reconstitution (6–8 wk), chimeric mice were treated with G-CSF (or left untreated), and osteocalcin mRNA expression in the bone marrow was measured by real-time RT-PCR. (B) Wildtype KL cells were transplanted into irradiated G-CSFR−/− recipients (n = 6–7, each group) and analyzed in a similar fashion. Data represent the mean ± SE. a p < 0.05.

FIG. 4

FIG. 4

Osteoblast turnover during G-CSF treatment. (A) Transgenic pOBCol2.3-GFP mice (n = 5–6, each group) were administered BrdU for 14 days and either treated for 5 days with G-CSF or left untreated. Mice were analyzed just before G-CSF treatment, after 5 days of G-CSF treatment, or after a 5 day recovery period (arrowheads). Shown is the percent of GFP+ cells in the bone marrow that were labeled with BrdU. (B) Representative scatter plots showing activated caspase 3 staining in the GFP+ cell population from untreated (left) or G-CSF–treated pOBCol2.3-GFP mice (right). (C) Shown is the percentage of GFP+ cells that express activated caspase 3 from untreated and day 3 G-CSF–treated mice (n = 4 each group). Data represent the mean ± SE. a p < 0.01, b p < 0.05.

FIG. 5

FIG. 5

Analysis of early osteoblast lineage cells during G-CSF treatment. (A) Real-time RT-PCR for the indicated genes (BSP, bone sialoprotein; ALP, alkaline phosphatase; OC, osteocalcin) was performed on total bone marrow RNA isolated after 5 days of G-CSF treatment. RNA expression relative to β-actin mRNA was calculated and compared with untreated bone marrow (assigned a value of 1; n = 5–12). (B) Shown is the number of alkaline phosphatase negative (AP negative, left) and positive (AP positive, right) CFU-F generated from the bone marrow of untreated or G-CSF–treated mice (n = 5–6 each group). (C) Mice (n = 2–4 each time point) were treated with G-CSF for the indicated period up to 22 days. Mice were killed at the indicated time points and analyzed for osteocalcin mRNA by real-time RT-PCR. (D) Mice (n = 6, each group) were treated 5 days with G-CSF or left untreated and administered BrdU for 5 days during the recovery period. Shown is the percent of GFP+ cells in the bone marrow that were labeled with BrdU. Data represent the mean ± SE. a p < 0.05 vs. untreated; b p < 0.01 vs. untreated, p < 0.05 vs. runx2 and OC; c p < 0.0001 vs. untreated, p < 0.05 vs. other groups; d p < 0.01; e p < 0.05 vs. untreated.

FIG. 6

FIG. 6

Osteoclastogenesis during G CSF treatment. (A and B) Wildtype mice (n = 2–6 each group) were treated with G-CSF for the indicated time or left untreated. Osteoclast number (A) and surface (B) were estimated by enumerating TRACP+ cells in paraffin-embedded sections of mouse long bones. (C) RANKL and OPG mRNA expression in the bone marrow of untreated or 5-day G-CSF–treated mice (N = 5–8 each group) was measured by real-time RT-PCR. (D) GFP+ cells were sorted from G-CSF–treated pOBCol2.3-GFP mice. RANKL and OPG mRNA was measured within this fraction. Data represent the mean ± SE. a p < 0.01 vs. other groups; b p < 0.001; c p < 0.05.

Similar articles

Cited by

References

    1. Taichman RS. Blood and bone: Two tissues whose fates are intertwined to create the hematopoietic stem-cell niche. Blood. 2005;105:2631–2639. - PubMed
    1. Lorenzo J. Interactions between immune and bone cells: New insights with many remaining questions. J Clin Invest. 2000;106:749–752. - PMC - PubMed
    1. Dale DC, Cottle TE, Fier CJ, Bolyard AA, Bonilla MA, Boxer LA, Cham B, Freedman MH, Kannourakis G, Kinsey SE, Davis R, Scarlata D, Schwinzer B, Zeidler C, Welte K. Severe chronic neutropenia: Treatment and follow-up of patients in the Severe Chronic Neutropenia International Registry. Am J Hematol. 2003;72:82–93. - PubMed
    1. Lee MY, Fukunaga R, Lee TJ, Lottsfeldt JL, Nagata S. Bone modulation in sustained hematopoietic stimulation in mice. Blood. 1991;77:2135–2141. - PubMed
    1. Takahashi T, Wada T, Mori M, Kokai Y, Ishii S. Overexpression of the granulocyte colony-stimulating factor gene leads to osteoporosis in mice. Lab Invest. 1996;74:827–834. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources