Vous êtes sur la page 1sur 9

From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

PHAGOCYTES, GRANULOCYTES, AND MYELOPOIESIS

Krox20/EGR2 deficiency accelerates cell growth and differentiation in the


monocytic lineage and decreases bone mass
Yankel Gabet,1,2 Sanjeev K. Baniwal,1,2 Nathalie Leclerc,1,2 Yunfan Shi,2 Alice E. Kohn-Gabet,2 Jon Cogan,2 Alexis Dixon,2
Marilyn Bachar,3 Lixin Guo,4 Jack E. Turman Jr,4 and Baruch Frenkel1,2,5
1Department of Biochemistry and Molecular Biology, Keck School of Medicine at the University of Southern California, Los Angeles, CA; 2Institute for Genetic

Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA; 3Bone Laboratory, Faculty of Dental Medicine, Hebrew University
of Jerusalem, Jerusalem, Israel; 4Center for Craniofacial Molecular Biology, Keck School of Medicine at the University of Southern California, Los Angeles, CA;
and 5Department of Orthopedic Surgery, Keck School of Medicine at the University of Southern California, Los Angeles, CA

Krox20/EGR2, one of the 4 early growth accelerated bone resorption as demon- silencing in preosteoclasts increased
response genes, is a highly conserved strated in vivo by increased osteoclast cFms expression and response to macro-
transcription factor implicated in hind- number and serum C-terminal telopep- phage colony-stimulating factor, leading
brain development, peripheral nerve tides, a marker for collagen degradation. to a cell-autonomous stimulation of
myelination, tumor suppression, and Krox20 haploinsufficiency did not reduce cell-cycle progression. Our data indicate
monocyte/macrophage cell fate determi- bone formation in vivo, nor did it compro- that the antimitogenic role of Krox20 in
nation. Here, we established a novel role mise osteoblast differentiation in vitro. In preosteoclasts is the predominant mecha-
for Krox20 in postnatal skeletal metabo- contrast, growth and differentiation were nism underlying the LBM phenotype of
lism. Microcomputed tomographic analy- significantly stimulated in preosteoclast Krox20-deficient mice. Stimulation of
sis of 4- and 8-week-old mice revealed a cultures derived from Krox20ⴙ/ⴚ spleno- Krox20 expression in preosteoclasts may
low bone mass phenotype (LBM) in both cytes, suggesting that the LBM is attribut- present a viable therapeutic strategy for
the distal femur and the vertebra of able to Krox20 haploinsufficiency in the high-turnover osteoporosis. (Blood. 2010;
Krox20ⴙ/ⴚ mice. This was attributable to monocytic lineage. Furthermore, Krox20 116(19):3964-3971)

Introduction
The family of early growth response (EGR) genes consists of and murine marrow progenitors at the expense of granulopoi-
4 members, EGR1/Krox24, EGR2/Krox20, EGR3, and EGR4.1 esis.27-30 Krox20 may also favor monocytic fate determination at
Their expression is rapidly induced by serum and growth factors, the expense of granulopoiesis by repressing transcription of Gfi-1,
such as nerve growth factor, epidermal growth factor, and platelet- a negative regulator of macrophage genes required for neutrophil
derived growth factor.2-4 The 4 EGR proteins are transcription cell differentiation.25 Krox24 and Krox20 have redundant roles in
factors that share a highly conserved DNA-binding domain com- monocytic maturation.25
posed of 3 zinc fingers, which recognize G:C-rich DNA motifs1,5-7 In vertebrates, bone mass and shape are determined by continu-
in the promoters of target genes such as Hox-1.4,8 Hoxa-2 and ous remodeling executed by the concerted action of osteoclasts, the
Hoxb-2,9 thymidine kinase,8,10 and synapsin I and II.11,12 The bone resorbing cells, and osteoblasts, the bone forming cells.
transactivation activity of EGR factors is modulated by co- Therefore, both decreased bone formation and increased bone
activators such as host cell factor C113 and corepressors such as resorption may result in bone loss. Little is known about the role of
NAB1,14 NAB2,15 and Ddx20.16 EGRs in bone. Krox24-deficient mice have low bone mass,
Early studies highlighted the role of EGR genes in cell attributable to increased expression of M-CSF in stromal cells and
proliferation.4 In cancer cells of various origins, Krox20 plays a increased bone turnover.31 In addition, retinoic acid and prostaglan-
role downstream of the PTEN tumor suppressor, and its deletion is din E2, which stimulate osteoblastic cell proliferation and differen-
associated with cancer cell proliferation.17,18 Krox20 knockout tiation, increase the expression of Krox24.32,33 Krox20 is largely
mice have severe abnormalities in hindbrain development19-21 and expressed in long bone at the cartilage-trabecular bone transition
impaired peripheral nerve myelination22 attributable to the role of line, and its absence results in failure of embryonic trabecular bone
Krox20 in regulating Schwann cells growth and differentiation.22-24 formation, which has been attributed to a differentiation block in
Krox20 has also been implicated in fate determination in the the chondroosteoblast lineage.34 Recently, we reported that Krox20
myeloid lineage. Specifically, it inhibits neutrophil- and activates expression was up-regulated in vitro during late stages of osteo-
macrophage-specific genes such as c-fms, encoding the macro- blast differentiation and was repressed by glucocorticoids.35 In vivo
phage colony-stimulating factor (M-CSF) receptor.25,26 Krox20 investigation of the role of Krox20 in postnatal bone metabolism is
interacts with PU.1, a master transcription factor critical in difficult because Krox20-null mice die within days after birth,
macrophage differentiation, to enhance transcription of c-fms.26 presumably due to neuronal defects.21 In this study, we employed
Krox24 stimulates monocytic differentiation in myeloid cell lines the viable mice haploinsufficient for Krox20 to assess its role in

Submitted January 12, 2010; accepted July 15, 2010. Prepublished online as payment. Therefore, and solely to indicate this fact, this article is hereby
Blood First Edition paper, August 17, 2010; DOI 10.1182/blood-2010-01-263830. marked ‘‘advertisement’’ in accordance with 18 USC section 1734.

The publication costs of this article were defrayed in part by page charge © 2010 by The American Society of Hematology

3964 BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19


From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19 KROX20 IN PREOSTEOCLASTS AND BONE MASS 3965

Table 1. Primers for genotyping and RT-qPCR (TRAP) staining and couterstaining with methyl green (Vector Inc).
Genotyping Sections were analyzed using a Zeiss Axioskop (Zeiss), with a 40⫻
Krox20 Fwd WT 5⬘ GCAGAAGGAACGGAAGAGCAG 3⬘ objective and a SPOT digital camera (Diagnostic Instruments). Metaphy-
Fwd Mutant 5⬘ GGCCGCTTTTCTGGATTCAT 3⬘ seal TRAP-labeled cells located at least 200 ␮m from the growth plate were
Rev 5⬘ ATCAAGGTCCTTTGCCCAGATC 3⬘ enumerated as either positive or negative for Krox20.
RT-qPCR
rpL10A Fwd 5⬘ CGCCGCAAGTTTCTGGAGAC 3⬘ CTX measurement
Rev 5⬘ CTTGCCAGCCTTGTTTAGGC 3⬘
Blood was drawn from 6-hour fasting animals at approximately 2 PM.
Krox20 Fwd 5⬘ TTGACCAGATGAACGGAGTG 3⬘
Levels of serum C-terminal telopeptides (CTX) were determined by
Rev 5⬘ CCAGAGAGGAGGTGGAAGTG 3⬘
enzyme-linked immunosorbent assay using RatLaps (Immunodiagnostic
Krox24 Fwd 5⬘ CCCTGACTATCTGTTTCC 3⬘
Systems) according to the manufacturer’s instructions.
Rev 5⬘ TCTGCTTTCTTGTCCTTC 3⬘
M-CSF Fwd 5⬘ AGTCTGTCTTCCACCTGCTG 3⬘
Rev 5⬘ TTCCACCTGTCTGTCCTCAT 3⬘
Tissue culture
cFms Fwd 5⬘ GGAGGTGGATTCCCTACCAT 3⬘ Splenocytes were extracted from 1-day-old pups, and preosteoclasts were
Rev 5⬘ GGCCCAGAACTGGTTGTAGA 3⬘ obtained after 3 days of culture in “standard” medium (␣-MEM, 10% FBS,
and 1% penicillin/streptomycin) supplemented with 100 ng/mL M-CSF.
For differentiation assay, adherent preosteoclasts were plated in 96-well
postnatal bone metabolism. We demonstrate that these mice have a plates with standard medium supplemented with 20 ng/mL M-CSF and
100 ng/mL receptor activator for nuclear factor ␬ B ligand (RANKL). On
low bone mass (LBM) phenotype attributable to increased bone
day 7, multinucleated osteoclasts were stained using a TRAP kit, and
resorption and that Krox20 attenuates osteoclast growth and
TRAP-positive surface was measured using ImageJ Version 1.41 software
differentiation in a cell autonomous manner. (National Institutes of Health). Alternatively, splenocytes were extracted,
plated in 96-well plates and treated from the first day with 20 ng/mL M-CSF
and 100 ng/mL RANKL for 8 days before TRAP staining. For proliferation
Methods assay, preosteoclasts were plated with standard medium supplemented with
100 ng/mL M-CSF, and cell number was assessed using the thiazolyl blue
Materials tetrazolium bromide (MTT) assay. Calvarial osteoblasts cultures were
prepared from 1-day-old pups as described previously.35 Cells were
All reagents were purchased from Sigma-Aldrich unless otherwise speci-
cultured on 12-well plates for reverse-transcription quantitative polymerase
fied. ␣-minimal essential medium and Dulbecco modified Eagle medium
chain reaction (RT-qPCR; see next paragraph) and mineralization assays.
were purchased from Gibco, fetal bovine serum (FBS) from Hyclone, and
For the latter, osteogenic medium containing ascorbic acid (50 ␮g/mL) and
culture plates from Corning. In splenocyte cultures, we used supernatant
␤-glycerophosphate (10mM) was initiated at confluence and alizarin red
from cardiomyogenic 14-12 cells, containing 1.3 ␮g/mL M-CSF.36
staining was performed at day 20. Mesenchymal stem cells were cultured
and analyzed as described previously.37
Animals

All experiments were approved by the Institutional Animal Care and Use mRNA and protein expression
Committee of the University of Southern California. Krox20⫹/⫺ female
RNA from cells was extracted and processed as described previously.37
mice were compared with their littermate female controls. The mice were
mRNA levels were corrected for ribosomal protein L10A (rpL10A) mRNA.
either on a pure C57BL/6 background (The Jackson Laboratory) or on a
Primers used for PCR are listed in Table 1. For Western blot analyses,
C57BL/6 ⫻ 129 background. Genotyping was performed as described
protein extraction, electrophoresis, and transfer were performed essentially
previously21 using primers listed in Table 1. To assess bone formation, mice
as previously described,35 and the Pierce Fast Western Blot kit (Thermo
were injected intraperitoneally with calcein 4 and 1 day before sacrifice.37
Scientifics) was used for antibody binding and chemiluminescence. Antibod-
ies were purchased from Cell Signaling (anti–phospho extracellular-signal-
Microcomputed tomography
regulated kinase [ERK]1/2 Thr202/Tyr204, anti-Akt, and anti–phospho Akt
Femora (1 per mouse) and third lumbar vertebrae were examined as Ser473), Promega (anti-ERK1/2), and Sigma-Aldrich (anti-cFms). The
reported previously37 using Scanco microcomputed tomography ␮CT40 mouse monoclonal anti-Tubulin antibody, developed by Dr Charles Walsh,
(Scanco Medical AG). Briefly, scans were performed at a 20-␮m resolution was obtained from the Developmental Studies Hybridoma Bank under the
in all 3 spatial dimensions. The mineralized tissues were differentially auspices of the National Institute of Child Health and Development and The
segmented by a global thresholding procedure.38 Trabecular parameters University of Iowa, Department of Biological Sciences, Iowa City, IA.
were measured in the secondary spongiosa of the distal femoral metaphysis
and in the entire trabecular bone compartment of the third lumbar vertebrae Krox20 silencing
body. Cortical parameters were determined in the mid-diaphyseal region.
We used Sigma Mission shRNA against Krox20 (shKrox20-1, Clone
567s1c1; shKrox20-2, Clone 788s1c1) or nonspecific (NS) shRNA as
Histomorphometry
control (SHC002). Lentiviral particles were generated by transfecting
Dynamic histomorphometric measurements and osteoclast counting were HEK293T cells with the plasmids carrying the shRNA sequence.40 For
performed as described previously using Media Cybernetic ImagePro Plus silencing, transduction of M-CSF–treated splenocytes was performed at
Version 6.0 software.37 The terminology and units used for these measure- approximately 30% confluence. Medium was changed after 16 hours, and
ments were according to the convention of standardized nomenclature.39 cells were allowed to recover for 48 hours before further analysis.

Assessment of Krox20 expression in osteoclasts in vivo FACS analyses

Both femora from 4 wild-type (WT) mice were removed, fixed in 4% For cell-cycle analysis, preosteoclasts were harvested by trypsinization
paraformaldehyde, and decalcified in 10% EDTA (ethylenediaminetetraace- 16 hours after medium change and analyzed with Fluorescence-activated
tic acid) before paraffin embedding. Five-micrometer sections separated by cell sorting (FACS) as previously described.41 To determine the monocytic
40 ␮m were incubated with Krox20 antibody (Covance) and labeled with cells distribution in bone marrow, 1 femur per animal was cut at the
Avidin-Biotin. This was followed by tartrate-resistant acid phosphatase mid-diaphysis and the epiphysis removed. Then, bone marrow cells were
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

3966 GABET et al BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19

flushed out from the distal cut end and red blood cells were lysed using a
10␮M Tris-HCl buffer containing 0.74% NH4Cl. Then, an aliquot of
1 ⫻ 106 cells was stained with allophycocyanin-conjugated anti–mouse
CD115 (eBiosciences). Stained cells were analyzed and sorted on a
FACSCalibur machine (Becton Dickinson). For analysis of apoptosis,
trypsinized cells were stained with fluorescein isothiocyanate (FITC)–
conjugated annexin V and propidium iodide according to the manufactur-
er’s instructions (Clontech). Cells positive for annexin V and/or propidium
iodide were recorded by flow cytometry.

Statistical analysis

Values are expressed as mean ⫾ SEM. Values beyond the mean ⫾ 1.5 ⫻ SD
range were excluded from the analysis. Difference between groups was
defined as P ⬍ .05 using Student t test and 1-way analysis of variance when
2 and 3 groups were compared, respectively.

Results
Low bone mass phenotype in Krox20-haploinsufficient mice

Although a role for Krox20 in embryonic endochondral bone


formation was suggested 2 decades ago,34 its potential postnatal
role in the skeleton has not been determined. Here, we investigated
the postnatal skeletal role of Krox20 using heterozygous mice,
which display no overt phenotype and whose total body weight is
similar to that of sex/age-matched WT controls. Microcomputed
tomographic analysis of 4- and 8-week-old Krox20-haploinsuffi-
cient female mice revealed a LBM phenotype compared with
littermate controls (Figure 1 and Table 2). The trabecular bone
volume density measured in the distal femur of Krox20⫹/⫺ mice
was 32% and 42% lower than the WT controls at 4 and 8 weeks of
age, respectively (Table 2 and Figure 1A). No difference was Figure 1. Low trabecular bone mass in Krox20ⴙ/ⴚ mice. (A) ␮CT analysis of the
observed in the femoral length, mid-diaphyseal diameter, and distal femoral trabecular bone of 10 Krox20⫹/⫹ (white) and 9 Krox20⫹/⫺ (gray)
cortical thickness (Table 3). The trabecular bone volume density 8-week-old female mice. BV/TV indicates trabecular bone volume density; Tb.N,
trabecular number; Tb.Th, trabecular thickness; and Conn.D, connectivity density.
was also lower in the body of the third lumbar vertebra in
Data are mean ⫾ SEM. *P ⬍ .05 compared with WT controls. (B) ␮CT images of
4-week-old mice (Table 2), suggesting a global positive role for distal femoral trabecular bone of Krox20⫹/⫹ (left) and Krox20⫹/⫺ (right) mice with
Krox20 in the postnatal regulation of trabecular bone mass. median BV/TV values.

Krox20 haploinsufficiency increases bone resorption in vivo


Figure 2E-G, the Krox20⫹/⫺ mice had normal mineralizing perim-
In pursuit of the cellular mechanism underlying the LBM pheno- eter (a surrogate for osteoblast number), normal mineral apposition
type of Krox20⫹/⫺ mice, we measured indicators of bone resorption rate (a surrogate for osteoblast activity), and therefore normal bone
and bone formation in vivo. First, we performed TRAP staining of formation rate. Thus, the LBM phenotype observed in Krox20-
histologic sections obtained from distal femoral metaphyses of heterozygous mice is attributable to increased bone resorption
8-week-old mice. As shown in Figure 2A-B, Krox20⫹/⫺ mice had a rather than decreased bone formation.
significant 32% higher osteoclast number compared with WT Krox20 haploinsufficiency results in increased preosteoclast
controls, suggesting that the Krox20⫹/⫺ LBM phenotype is attribut- proliferation in vitro
able to increased osteoclast activity. Further implicating bone
resorption in the LBM phenotype, Krox20⫹/⫺ mice exhibited a We next investigated the role of Krox20 in osteoclasts and
significant 44% increase in serum CTXs, a biochemical marker of osteoblasts in vitro. M-CSF–treated splenocytes were used to study
bone resorption (Figure 2C). Because osteoclasts derive from the effects of Krox20 on osteoclast growth and differentiation. The
monocytes,42 we used FACS to compare CD115⫹ monocytes in 2-fold decrease in Krox20 expression in the preosteoclast cultures
bone marrow of WT versus Krox20⫹/⫺ mice. However, the derived from the heterozygous animals (Figure 3A) was associated
monocyte count in the mutant mice was normal (Figure 2D), with a significant 70% increase in proliferation rate, as determined
suggesting that Krox20 haploinsufficiency does not exert its effect by MTT assay (Figure 3B). We then tested whether the increased
during early monocyte differentiation but rather later during the proliferation of the osteoclast precursors led to more differentiated
development of the osteoclast phenotype. osteoclasts. Splenocytes from WT and Krox20⫹/⫺ mice were plated
Because previous work with mouse embryos and cell culture and treated with M-CSF and RANKL to promote differentiation
models suggested a possible role for Krox20 in cells of the into preosteoclasts and then mature osteoclasts in the same culture.
osteoblast lineage,34,35,43 we also tested whether the Krox20⫹/⫺ Consistent with the increased proliferation (Figure 3B), the
LBM phenotype was associated with reduced bone formation. We Krox20⫹/⫺ splenocytes gave rise to significantly more osteoclasts
used vital double calcein labeling to assess bone matrix apposition than did their WT counterparts (Figure 3C). Next, we specifically
in vivo in the distal femur of 8-week-old mice, but, as shown in tested the role of Krox20 in the growth and differentiation of
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19 KROX20 IN PREOSTEOCLASTS AND BONE MASS 3967

Table 2. Trabecular bone microarchitecture in distal femoral metaphysis and third lumbar vertebral body from 4-week-old WT and
Krox20ⴙ/ⴚ mice
Distal femur L3 vertebral body
Krox20ⴙ/ⴙ (n ⴝ 5) Krox20ⴙ/ⴚ (n ⴝ 5) P Krox20ⴙ/ⴙ (n ⴝ 7) Krox20ⴙ/ⴚ (n ⴝ 5) P

BV/TV (%) 6.9 ⫾ 0.8 4.7 ⫾ 0.5 .034 13.6 ⫾ 1.0 10.6 ⫾ 0.9 .028
Tb.N (mm⫺1) 1.8 ⫾ 0.1 1.3 ⫾ 0.1 .017 3.8 ⫾ 0.2 3.2 ⫾ 0.2 .024
Tb.Th (␮m) 38 ⫾ 0.2 36 ⫾ 0.1 .225 35 ⫾ 0.1 32 ⫾ 0.1 .077
Conn.D (mm⫺3) 29.5 ⫾ 5.8 16.5 ⫾ 2.8 .046 109.7 ⫾ 10.8 71.1 ⫾ 10.6 .015

Data are mean ⫾ SEM.


BV/TV indicates trabecular bone volume density; Tb.N, trabecular number; Tb.Th, trabecular thickness; and Conn.D, connectivity density.

monocytes that have already committed to the osteoclast pheno- ingly increased in Krox20⫹/⫺ cultures (Figure 3I). Further cell
type. To this end, splenocytes were treated with M-CSF for 3 days, proliferation in the osteoblast lineage, assessed by the average size
and equal numbers of the resulting preosteoclasts from Krox20⫹/⫺ of the osteoblastic colonies, was unaffected by the Krox20 geno-
and WT mice were replated and induced to further differentiate by type (Figure 3J). Thus, consistent with the in vivo data (Figure 2),
the addition of RANKL. Following this protocol, we again the results of our ex vivo and culture assays indicate that the
observed significantly more TRAP⫹ osteoclasts in cultures derived Krox20⫹/⫺ LBM phenotype is attributable to increased bone
from Krox20-haploinsufficient mice (Figure 3D-E). These results resorption and not to decreased bone formation.
suggest that the in vivo Krox20⫹/⫺ LBM phenotype is attributable
to increased growth and differentiation of the osteoclast precursors. Krox20 attenuates preosteoclast cell cycle in a cell
The role of Krox20 in the osteoblast lineage was investigated autonomous manner
using osteoblast cultures derived from either newborn mouse
calvariae or adult bone marrow. As in preosteoclasts, Krox20 To determine the mechanism by which Krox20 attenuates preoste-
expression was reduced, by as much as 3.5-fold, in osteoblast oclasts proliferation, we initially profiled Krox20⫹/⫹, Krox20⫹/⫺,
cultures derived from calvariae of Krox20⫹/⫺ compared with and Krox20⫺/⫺ cells for their distribution among the phases of the
Krox20⫹/⫹ newborn mice (Figure 3F). However, this decrease was cell cycle. As shown in Figure 4A, reduction in the Krox20 gene
not associated with compromised mineralization as assessed by dosage was associated with a progressive, up to 3-fold, increase in
alizarin red staining of day-20 calvarial osteoblast cultures (Figure the percentage of cells in the S and G2/M phases, indicating a
3G-H). In mesenchymal stem cell cultures derived from the bone negative control of the preosteoclast cell cycle by Krox20. The role
marrow of Krox20 heterozygous mice, we again did not observe a of Krox20 in preosteoclast survival was then assessed based on
decrease in the potential for bone-like tissue formation. In fact, the annexin V staining and propidium iodide retention. As shown in
number of osteoblastic colony-forming units (CFU-Ob) was surpris- Figure 4B, reduction in the Krox20 gene dosage had a mild positive

Figure 2. Increased bone resorption in Krox20ⴙ/ⴚ


animals. (A) Histologic images showing increased oste-
oclast number and thinner trabeculae in the distal femur
of WT (left) and Krox20⫹/⫺ (right) animals. Arrows point at
TRAP⫹ lining osteoclasts. Images were acquired using a
Zeiss Axioskop, with a 40⫻ objective and a SPOT digital
camera. (B) Oc.N/BS, TRAP⫹ lining osteoclasts relative
to bone surface measured in histologic sections of the
distal femur from five 8-week-old mice per group.
(C) Serum CTX levels in 4 WT and 3 Krox20⫹/⫺ 6-week-
old mice. (D) FACS analysis of CD115⫹ monocytes in the
distal femoral metaphysis of 4 WT (left) and 5 Krox20⫹/⫺
(right) 6-week-old mice. Bars show the percentage
CD115⫹ monocytes from total red blood cell-free bone
marrow cells. (E-G) Parameters of bone formation were
measured in histologic sections of the distal femur from 5
8-week-old mice per group. (E) MAR indicates mineral
apposition rate. (F) Min.Peri indicates mineralizing perim-
eter. (G) BFR indicates bone formation rate. Bars are
mean ⫾ SEM with the Krox20⫹/⫺ values always on the
right. *P ⬍ .05 compared with WT controls.
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

3968 GABET et al BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19

Table 3. External and cortical dimensions of femora from 8-week-


old WT and Krox20ⴙ/ⴚ mice
Krox20ⴙ/ⴙ (n ⴝ 10) Krox20ⴙ/ⴚ (n ⴝ 9) P

Femoral length 14.76 ⫾ 0.23 14.65 ⫾ 0.27 .380


Dia.Dia 1.536 ⫾ 0.031 1.502 ⫾ 0.036 .235
Med.Dia 1.238 ⫾ 0.029 1.213 ⫾ 0.031 .279
Cort.Th 0.162 ⫾ 0.007 0.161 ⫾ 0.004 .492

Data are mean ⫾ SEM in mm.


Dia.Dia indicates mid-diaphyseal periosteal diameter; Med.Dia, mid-diaphyseal
medullary diameter; and Cort.Th, mid-diaphyseal cortical thickness.

effect on preosteoclast survival, attributable to decreased apoptosis,


measured by annexin V staining. Taken together, our results
indicate that Krox20 attenuates preosteoclast proliferation primar-
ily by restraining cell-cycle progression.
Although osteoclast growth and differentiation was increased in
the relatively pure splenocyte cultures from Krox20-deficient mice
(Figure 3B-D), these results do not necessarily indicate a cell
autonomous role of Krox20 because the splenocytes were extracted
from mice that were Krox20-compromised from conception and in
many cell types, including, for example, spleen stromal cells and
myeloid early precursors. To conclusively address the potential cell
autonomous effect of Krox20 in osteoclasts, we first confirmed the
expression of Krox20 in osteoclasts in vivo. Histologic sections of
mature bone were subjected to TRAP staining combined with
Krox20 immunohistochemistry as described in “Methods” (Figure
4C), and 263 TRAP⫹ cells were scored as either Krox20⫹ or
Krox20⫺. We found that 94% of the osteoclasts (248 cells)
expressed Krox20, indicating that Krox20 can have a cell autono-
mous role in osteoclasts in vivo.
To test whether the antiproliferative effect of Krox20 in
preosteoclasts is truly cell autonomous, we examined the effects of
Krox20 silencing in vitro. WT preosteoclasts were transduced with
lentiviruses encoding a shRNA against Krox20. To rule out off-target
effects, we used 2 different shRNA sequences, shKrox20-1 and Figure 3. Increased proliferation in osteoclast precursors derived from Krox20ⴙ/ⴚ
mice. (A) Splenocytes from WT and Krox20⫹/⫺ newborn mice were cultured for
shKrox20-2, which reduced Krox20 mRNA levels by 75% and 3 days with 100 ng/mL M-CSF. Krox20 and Krox24 mRNA expression in subconfluent
81%, respectively, compared with cells transduced with a lentivirus preosteoclasts cultures was assessed by RT-qPCR and corrected for the expression
encoding an NS shRNA (Figure 4D). As shown in Figure 4E, of rpL10A. (B) Spleen-derived preosteoclasts as in panel A were plated in 96-well
plates at 2000 cells per well and cultured with 100 ng/mL M-CSF. Medium was
Krox20 silencing by shKrox20-1 and shKrox20-2 resulted in 39%
changed on days 3 and 5, and proliferation was determined after 7 days using MTT
and 74% increase in preosteoclast proliferation, respectively, assay. (C) Splenocytes from WT and Krox20⫹/⫺ newborn mice were plated in 96-well
indicating that the antiproliferative effect of Krox20 does not plates at 150 000 cells per well and cultured from the first day with 20 ng/mL M-CSF
require involvement of other cells. Consistent with the cell-cycle and 100 ng/mL RANKL. Medium was changed on days 3 and 5, and TRAP staining
was performed on day 8. Bars represent relative TRAP⫹ area. (D-E) Spleen-derived
profiles of Krox20⫹/⫹, Krox20⫹/⫺, and Krox20⫺/⫺ preosteoclasts preosteoclasts pretreated with 100 ng/mL M-CSF alone as in panel A were plated in
(Figure 4A), the accelerated proliferation of the shKrox20- 96-well plates at 5000 cells per well and cultured with 20 ng/mL M-CSF and
transduced cells was associated with a 60% increase in the 100 ng/mL RANKL for 7 days. Medium was changed on days 3 and 5. Bars represent
relative TRAP⫹ area. (E) Representative images of TRAP-positive osteoclasts
percentage of cells in the S and G2/M phases of the cell cycle derived from WT (left) and Krox20⫹/⫺ (right) mice; bar ⫽ 0.5 mm. (F) Krox20 and
(Figure 4E-F). However, Krox20 silencing did not affect cell Krox24 mRNA in primary calvarial osteoblastic cultures from Krox20⫹/⫺ and control
survival (Figure 4G). Because the genetic approach suggested only mice was assessed by RT-qPCR and corrected for the expression of rpL10A.
(G-H) Primary calvarial osteoblast cultures from WT and Krox20⫹/⫺ mice were fixed
a minor role for Krox20 in preosteoclast apoptosis (Figure 4B), this
and stained with alizarin red 20 days after confluence. Bars represent percent alizarin
and the silencing approaches collectively support the conclusion red-positive area. (H) Representative images from WT- (left) and Krox20⫹/⫺-derived
that Krox20 inhibits cell proliferation primarily by attenuating (right) cultures. (I-J) Bone marrow–derived cultures from WT and Krox20⫹/⫺ mice.
preosteoclast cell-cycle progression in a cell autonomous manner. Number of alizarin red-positive CFU-Ob colonies (I) and mean colony size (J) were
determined in day-20 cultures after fixation and staining. Bars represent mean
⫾ SEM of Krox20⫹/⫹ (white) and Krox20⫹/⫺ (gray) cells. *P ⬍ .05 compared with
Krox20 attenuates cFms signaling in preosteoclasts
control littermates. Assays were performed in triplicate per animal, and the number of
animals is specified in parentheses inside each bar.
Preosteoclast proliferation is tightly regulated by binding of
M-CSF to its cFms receptor and the subsequent activation of the
phosphatidylinositol 3-kinase/Akt and MEK/ERK pathways.44 We As shown in Figure 5A-B, Krox20 silencing increased cFms
tested the hypothesis that Krox20 restrains cFms signaling in expression at both the mRNA and protein levels. Furthermore,
preosteoclasts by measuring cFms expression as well as M-CSF– analysis of Akt and ERK phosphorylation after M-CSF treatment
mediated activation of the phosphatidylinositol 3-kinase/Akt and demonstrated an augmented response in the Krox20-depleted cells
the MEK/ERK pathways in shKrox20-transduced preosteoclasts. (Figure 5C). These findings suggest that Krox20 attenuates the
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19 KROX20 IN PREOSTEOCLASTS AND BONE MASS 3969

Figure 4. Silencing of Krox20 in preosteoclasts accelerates cell-cycle progression. (A) Preosteoclasts derived from WT, Krox20⫹/⫺, and Krox20⫺/⫺ mice were plated in
12-well plates at 20 000 cells per well and cultured with 100 ng/mL M-CSF for 3 days before cell-cycle profiling. Bars represent percentage of cells in the S⫹G2/M phases.
(B) Apoptosis was assessed in preosteoclasts obtained as in panel A using annexin V and propidium iodide staining. Survival corresponds to the percentage of cells negative
for both FITC–annexin V labeling and propidium iodide. (A-B) Bars represent mean ⫾ SEM; n ⫽ 4; #P ⬍ .05, 1-way analysis of variance for the effect of gene dosage.
(C) Combined Krox20 immunohistochemistry (brown/black) and TRAP staining (purple) of a section from a WT distal femur, showing a representative Krox20-expressing
osteoclast. Arrow points at a brown/black granular immunohistochemical reaction product indicating nuclear Krox20. Arrowheads depict faint brown cytoplasmic Krox20
immunostaining. (D) Splenocytes from WT mice were cultured for 3 days with 100 ng/mL M-CSF, and Krox20 expression was silenced using 2 different shRNA-coding
lentiviruses (shKrox20-1 and shKrox20-2). Krox20 mRNA levels were determined by RT-qPCR and corrected for the corresponding expression of rpL10A. Bars represent
expression levels relative to cells transduced with a nonspecific (NS) shRNA lentivirus. (E) Preosteoclasts were plated in 48-well format at 6000 cells per well and cultured with
100 ng/mL M-CSF, and Krox20 expression was silenced as in panel D. Proliferation was determined between days 4 and 9 after transduction using MTT assay.
(F) Preosteoclasts were plated in 12-well plates at 20 000 cells per well and cultured with 100 ng/mL M-CSF. Cells were then transduced with lentiviruses encoding the
shKrox20-2 or the NS shRNA, and cell-cycle analysis was performed as in panel A. Representative cell-cycle profiles are shown on the right. (G) Apoptosis was assessed in
preosteoclasts obtained as in panel F. (D-G) Bars represent mean ⫾ SEM; n ⫽ 4; *P ⬍ .05 vs NS.

proliferation of preosteoclasts by down-regulating their cFms The resulting acceleration of bone resorption underlies the ob-
levels and thus blunting their response to M-CSF. served LBM phenotype.
By in large, macrophages and preosteoclasts are closely related
as they both differentiate from monocytes after M-CSF stimulation.
Discussion Cellular events triggered by RANKL can then induce M-CSF–
stimulated monocytes to further differentiate into mature oste-
For the first time, this work establishes a postnatal role for Krox20 oclasts. Here, we found that the number of bone marrow monocytes
in bone metabolism. Krox20-haploinsufficiency in preosteoclasts was not affected by the Krox20 gene dosage (Figure 2D). Krox20 is
results in accelerated proliferation and cell-cycle progression, also not critical for M-CSF–related differentiation of liver mono-
likely due to increased cFms expression and signaling. This leads to cytes into macrophages,45 likely because of the redundant role of
increased number of mature osteoclasts both in vitro and in vivo. Krox24 in monocyte maturation and macrophage differentiation.25
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

3970 GABET et al BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19

Figure 5. Silencing of Krox20 in preosteoclasts increases cFms


signaling. (A) Wild type splenocytes were cultured for 3 days with
100 ng/mL M-CSF, and Krox20 expression was silenced using shKrox20-2
lentiviruses. cFms mRNA levels were determined by RT-qPCR and
corrected for the corresponding expression of rpL10A. Bars represent
mean ⫾ SEM of expression levels relative to cells transduced with a
nonspecific (NS) shRNA lentivirus (n ⫽ 5; *P ⬍ .05 vs NS). (B) Western
blot analysis of cFms was performed on preosteoclasts obtained as in
panel A. Tubulin was used as loading control. (C) Preosteoclasts obtained
as in panel A were serum-starved for 2 hours and treated with M-CSF for
5 and 30 minutes as indicated, followed by Western blot analysis of
phospho- and total Akt and ERK. A vertical line has been inserted to
indicate a repositioned gel lane.

In addition, our data suggest that Krox20 does not play a significant (0.82 ⫾ 0.27 vs 1.00 ⫾ 0.25, respectively; n ⫽ 5; P ⫽ .64), total
role in terminal osteoclast differentiation, as the overall increase in M-CSF expression was elevated in RNA extracts from Krox20⫹/⫺
osteoclastogenesis in Krox20⫹/⫺ preosteoclasts cultures did not compared with WT femora (2.07 ⫾ 0.20 vs 1.00 ⫾ 0.18, respec-
exceed the increase in preosteoclast proliferation (Figure 3B-E). tively, n ⫽ 5, P ⬍ .05). Furthermore, any increase in osteoblast-
Therefore, our findings imply that the LBM phenotype observed in derived M-CSF in Krox20⫹/⫺ bones is expected to be amplified due
Krox20-haploinsufficient mice results from enhanced growth and to the cell-autonomous increase of cFms signaling in the Krox20⫹/⫺
differentiation of osteoclast precursors. preosteoclasts.
Unlike the inhibitory effect of Krox20 on the preosteoclast cell In contrast to our findings in preosteoclasts, Krox20 operates in
cycle, its role in apoptosis is more complex. We observed a mild concert with PU.1 to enhance cFms expression in myeloid progeni-
proapoptotic effect of Krox20 by comparing preosteoclasts from tors.25,26 Conceivably, the Krox20/PU.1-binding element at the
Krox20⫹/⫹, Krox20⫹/⫺, and Krox20⫺/⫺ mice. This effect is prob- cFms gene switches its function from a transcriptional enhancer to
ably indirect because apoptosis was not affected by Krox20 a transcriptional repressor as monocytes differentiate to preoste-
silencing in isolated WT cells. Interestingly, unlike our preoste- oclasts. Such switch can occur as the cellular milieu changes,
oclast cultures, Bradley et al46 reported a prosurvival role for resulting in replacement of Krox20 co-activators with
Krox20 in mature osteoclasts in vitro. Be that as it may, the corepressors.15,25
increased proliferation of Krox20-compromised preosteoclasts in In summary, we established Krox20 as a significant antiprolif-
vitro (Figures 3B and 4E) and the increased osteoclast number and erative transcription factor in preosteoclasts. Decreased levels of
bone resorption in Krox20⫹/⫺ mice in vivo (Figure 2A-C) demon- Krox20 led to increased osteoclast number, increased bone resorp-
strate that inhibition of cell-cycle progression (Figure 4A,F) is the tion, and low bone mass. Thus, maintenance or stimulation of
overall predominant effect of Krox20 in the osteoclast lineage. Krox20 expression in preosteoclasts may present a viable therapeu-
The present study demonstrates a major physiologic role for tic strategy for high-turnover osteoporosis.
osteoclastic Krox20 in postnatal bone mass control. However, we
cannot rule out a role for Krox20 in cells of the osteoblast lineage
as well. In fact, we report here that Krox20 haploinsufficiency Acknowledgments
increases bone marrow–derived CFU-Ob (Figure 3I). This could
reflect a bona fide effect of Krox20 on early osteoblast progenitors, We thank Dr Steve Teitelbaum and Dr Jennifer Reeve (Washington
similar to its effect on osteoclast progenitors. However, such a University, St Louis, MO) for sharing their expertise and for the
putative effect does not result in increased net bone formation in generous gifts of RANKL and the M-CSF–containing conditioned
Krox20⫹/⫺ mice (Figure 2E-G). On the other hand, because medium. We also thank Dr Debbie Johnson and Dr Sandra Johnson
preosteoblasts significantly contribute to the bone marrow M-CSF (University of Southern California) for sharing their expertise and
pool, the increased CFU-Ob may contribute to the elevated for the generous gifts of antibodies.
osteoclastogenesis observed in the Krox20-compromised animals. This work was supported by grant RO1 AR047052 from
Indeed, although RT-qPCR analysis of Krox20⫹/⫺ newborn cal- NIH/NIAMS. Y.G. was partly supported by a Meyer Young
varial osteoblast cultures demonstrated normal M-CSF expression Investigator Fellowship and S.K.B. by a postdoctoral Innovative
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

BLOOD, 11 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 19 KROX20 IN PREOSTEOCLASTS AND BONE MASS 3971

Chapter Research Award, both from the Arthritis Foundation S.K.B., N.L., J.E.T., and B.F. analyzed data; and Y.G. and B.F.
Southern California Chapter. B.F. is the holder of the J. Harold and wrote the paper.
Edna L. LaBriola Chair in Genetic Orthopaedic Research at the Conflict-of-interest disclosure: The authors declare no compet-
University of Southern California. ing financial interests.
Correspondence: Baruch Frenkel, DMD, PhD, Institute for
Genetic Medicine, University of Southern California Keck School
Authorship of Medicine, 2250 Alcazar St, IGM/CSC-240, Los Angeles, CA
90033; e-mail: frenkel@usc.edu; or Yankel Gabet, DMD, PhD,
Contribution: Y.G., N.L., and B.F. designed research; Y.G., S.K.B., Institute for Genetic Medicine, University of Southern California
N.L., Y.S., A.E.K.-G., J.C., A.D., M.B., and L.G. performed Keck School of Medicine, 2250 Alcazar St, IGM/CSC-240, Los
research; S.K.B. contributed new reagents/analytic tools; Y.G., Angeles, CA 90033; e-mail: gabet@usc.edu.

References
1. Gashler A, Sukhatme VP. Early growth response protein represses transcriptional activation by 32. Suva LJ, Ernst M, Rodan GA. Retinoic acid in-
protein 1 (Egr-1): prototype of a zinc-finger family Egr2/Krox-20. J Biol Chem. 2004;279:9056-9063. creases zif268 early gene expression in rat
of transcription factors. Prog Nucleic Acid Res 17. Unoki M, Nakamura Y. Growth-suppressive ef- preosteoblastic cells. Mol Cell Biol. 1991;11:
Mol Biol. 1995;50:191-224. fects of BPOZ and EGR2, two genes involved in 2503-2510.
2. Milbrandt J. A nerve growth factor-induced gene the PTEN signaling pathway. Oncogene. 2001; 33. Fang MA, Noguchi GM, McDougall S. Prosta-
encodes a possible transcriptional regulatory fac- 20:4457-4465. glandin E2 induces Egr-1 mRNA in MC3T3-E1
tor. Science. 1987;238:797-799. 18. Stanbridge EJ. Human tumor suppressor genes. osteoblastic cells by a protein kinase C-depen-
3. Christy BA, Lau LF, Nathans D. A gene activated Annu Rev Genet. 1990;24:615-657. dent pathway. Prostaglandins Leukot Essent
in mouse 3T3 cells by serum growth factors en- Fatty Acids. 1996;54:109-114.
19. Wilkinson DG, Bhatt S, Chavrier P, Bravo R,
codes a protein with “zinc finger” sequences. Charnay P. Segment-specific expression of a 34. Levi G, Topilko P, Schneider-Maunoury S, et al.
Proc Natl Acad Sci U S A. 1988;85:7857-7861. zinc-finger gene in the developing nervous sys- Defective bone formation in Krox-20 mutant mice.
4. Lemaire P, Revelant O, Bravo R, Charnay P. Two tem of the mouse. Nature. 1989;337:461-464. Development. 1996;122:113-120.
mouse genes encoding potential transcription 20. Schneider-Maunoury S, Topilko P, Seitandou T, et 35. Leclerc N, Noh T, Cogan J, Samarawickrama DB,
factors with identical DNA-binding domains are al. Disruption of Krox-20 results in alteration of Smith E, Frenkel B. Opposing effects of glucocor-
activated by growth factors in cultured cells. Proc rhombomeres 3 and 5 in the developing hind- ticoids and Wnt signaling on Krox20 and mineral
Natl Acad Sci U S A. 1988;85:4691-4695. brain. Cell. 1993;75:1199-1214. deposition in osteoblast cultures. J Cell Biochem.
5. Chavrier P, Zerial M, Lemaire P, Almendral J, 2008;103:1938-1951.
21. Swiatek PJ, Gridley T. Perinatal lethality and de-
Bravo R, Charnay P. A gene encoding a protein fects in hindbrain development in mice homozy- 36. Takeshita S, Kaji K, Kudo A. Identification and
with zinc fingers is activated during G0/G1 transi- gous for a targeted mutation of the zinc finger characterization of the new osteoclast progenitor
tion in cultured cells. Embo J. 1988;7:29-35. gene Krox20. Genes Dev. 1993;7:2071-2084. with macrophage phenotypes being able to differ-
6. Swirnoff AH, Milbrandt J. DNA-binding specificity entiate into mature osteoclasts. J Bone Miner
22. Topilko P, Schneider-Maunoury S, Levi G, et al.
of NGFI-A and related zinc finger transcription Res. 2000;15:1477-1488.
Krox-20 controls myelination in the peripheral
factors. Mol Cell Biol. 1995;15:2275-2287. nervous system. Nature. 1994;371:796-799. 37. Noh T, Gabet Y, Cogan J, et al. Lef1 haploinsuffi-
7. O’Donovan KJ, Tourtellotte WG, Millbrandt J, cient mice display a low turnover and low bone
23. Parkinson DB, Bhaskaran A, Droggiti A, et al.
Baraban JM. The EGR family of transcription- mass phenotype in a gender- and age-specific
Krox-20 inhibits Jun-NH2-terminal kinase/c-Jun
regulatory factors: progress at the interface of manner. PLoS One. 2009;4:e5438.
to control Schwann cell proliferation and death.
molecular and systems neuroscience. Trends J Cell Biol. 2004;164:385-394. 38. Ruegsegger P, Koller B, Muller R. A microtomo-
Neurosci. 1999;22:167-173. graphic system for the nondestructive evaluation of
24. Zorick TS, Syroid DE, Brown A, Gridley T, Lemke
8. Chavrier P, Vesque C, Galliot B, et al. The segment- G. Krox-20 controls SCIP expression, cell cycle bone architecture. Calcif Tissue Int. 1996;58:24-29.
specific gene Krox-20 encodes a transcription factor exit and susceptibility to apoptosis in developing 39. Parfitt AM, Drezner MK, Glorieux FH, et al. Bone
with binding sites in the promoter region of the Hox- myelinating Schwann cells. Development. 1999; histomorphometry: standardization of nomencla-
1.4 gene. EMBO J. 1990;9:1209-1218. 126:1397-1406. ture, symbols, and units. Report of the ASBMR
9. Nonchev S, Maconochie M, Vesque C, et al. The 25. Laslo P, Spooner CJ, Warmflash A, et al. Multilin- Histomorphometry Nomenclature Committee.
conserved role of Krox-20 in directing Hox gene eage transcriptional priming and determination of J Bone Miner Res. 1987;2:595-610.
expression during vertebrate hindbrain segmen- alternate hematopoietic cell fates. Cell. 2006;126: 40. Kim JH, Yang CK, Heo K, Roeder RG, An W,
tation. Proc Natl Acad Sci U S A. 1996;93:9339- 755-766. Stallcup MR. CCAR1, a key regulator of mediator
9345. complex recruitment to nuclear receptor tran-
26. Krysinska H, Hoogenkamp M, Ingram R, et al. A
10. Molnar G, Crozat A, Pardee AB. The immediate- two-step, PU. 1-dependent mechanism for devel- scription complexes. Mol Cell. 2008;31:510-519.
early gene Egr-1 regulates the activity of the thy- opmentally regulated chromatin remodeling and 41. Smith E, Redman RA, Logg CR, Coetzee GA,
midine kinase promoter at the G0-to-G1 transition transcription of the c-fms gene. Mol Cell Biol. Kasahara N, Frenkel B. Glucocorticoids inhibit
of the cell cycle. Mol Cell Biol. 1994;14:5242- 2007;27:878-887. developmental stage-specific osteoblast cell
5248. cycle. Dissociation of cyclin A-cyclin-dependent
27. Nguyen HQ, Hoffman-Liebermann B, Liebermann
11. Petersohn D, Schoch S, Brinkmann DR, Thiel G. DA. The zinc finger transcription factor Egr-1 is kinase 2 from E2F4-p130 complexes. J Biol
The human synapsin II gene promoter. Possible essential for and restricts differentiation along the Chem. 2000;275:19992-20001.
role for the transcription factor zif268/egr-1, poly- macrophage lineage. Cell. 1993;72:197-209. 42. Boyle WJ, Simonet WS, Lacey DL. Osteoclast
oma enhancer activator 3, and AP2. J Biol Chem. differentiation and activation. Nature. 2003;423:
28. Krishnaraju K, Hoffman B, Liebermann DA. The
1995;270:24361-24369. 337-342.
zinc finger transcription factor Egr-1 activates
12. Thiel G, Cibelli G. Regulation of life and death by macrophage differentiation in M1 myeloblastic 43. Levi G, Topilko P, Schneider-Maunoury S, et al.
the zinc finger transcription factor Egr-1. J Cell leukemia cells. Blood. 1998;92:1957-1966. Role of Krox-20 in endochondral bone formation.
Physiol. 2002;193:287-292. Ann N Y Acad Sci. 1996;785:288-291.
29. Krishnaraju K, Hoffman B, Liebermann DA. Early
13. Luciano RL, Wilson AC. HCF-1 functions as a growth response gene 1 stimulates development 44. Ross FP. M-CSF, c-Fms, and signaling in oste-
coactivator for the zinc finger protein Krox20. of hematopoietic progenitor cells along the mac- oclasts and their precursors. Ann N Y Acad Sci.
J Biol Chem. 2003;278:51116-51124. rophage lineage at the expense of the granulo- 2006;1068:110-116.
14. Russo MW, Sevetson BR, Milbrandt J. Identifica- cyte and erythroid lineages. Blood. 2001;97: 45. Carter JH, Tourtellotte WG. Early growth re-
tion of NAB1, a repressor of NGFI-A- and Krox20- 1298-1305. sponse transcriptional regulators are dispensable
mediated transcription. Proc Natl Acad Sci U S A. 30. Krishnaraju K, Nguyen HQ, Liebermann DA, for macrophage differentiation. J Immunol. 2007;
1995;92:6873-6877. Hoffman B. The zinc finger transcription factor Egr-1 178:3038-3047.
15. Svaren J, Sevetson BR, Apel ED, Zimonjic DB, potentiates macrophage differentiation of hematopoi- 46. Bradley EW, Ruan MM, Oursler MJ. Novel pro-
Popescu NC, Milbrandt J. NAB2, a corepressor of etic cells. Mol Cell Biol. 1995;15:5499-5507. survival functions of the Kruppel-like transcription
NGFI-A (Egr-1) and Krox20, is induced by prolif- 31. Cenci S, Weitzmann MN, Gentile MA, Aisa MC, factor Egr2 in promotion of macrophage colony-
erative and differentiative stimuli. Mol Cell Biol. Pacifici R. M-CSF neutralization and egr-1 defi- stimulating factor-mediated osteoclast survival
1996;16:3545-3553. ciency prevent ovariectomy-induced bone loss. downstream of the MEK/ERK pathway. J Biol
16. Gillian AL, Svaren J. The Ddx20/DP103 dead box J Clin Invest. 2000;105:1279-1287. Chem. 2008;283:8055-8064.
From www.bloodjournal.org by guest on May 23, 2018. For personal use only.

2010 116: 3964-3971


doi:10.1182/blood-2010-01-263830 originally published
online August 17, 2010

Krox20/EGR2 deficiency accelerates cell growth and differentiation in


the monocytic lineage and decreases bone mass
Yankel Gabet, Sanjeev K. Baniwal, Nathalie Leclerc, Yunfan Shi, Alice E. Kohn-Gabet, Jon Cogan,
Alexis Dixon, Marilyn Bachar, Lixin Guo, Jack E. Turman Jr and Baruch Frenkel

Updated information and services can be found at:


http://www.bloodjournal.org/content/116/19/3964.full.html
Articles on similar topics can be found in the following Blood collections
Phagocytes, Granulocytes, and Myelopoiesis (654 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

Vous aimerez peut-être aussi