You are on page 1of 15

Connective Tissue Research

ISSN: 0300-8207 (Print) 1607-8438 (Online) Journal homepage: https://www.tandfonline.com/loi/icts20

Epigenetic regulation of bone cells

Kyung Hyun Park-Min

To cite this article: Kyung Hyun Park-Min (2017) Epigenetic regulation of bone cells, Connective
Tissue Research, 58:1, 76-89, DOI: 10.1080/03008207.2016.1177037

To link to this article: https://doi.org/10.1080/03008207.2016.1177037

Accepted author version posted online: 14


Apr 2016.
Published online: 17 May 2016.

Submit your article to this journal

Article views: 558

View related articles

View Crossmark data

Citing articles: 7 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=icts20
CONNECTIVE TISSUE RESEARCH
2017, VOL. 58, NO. 1, 76–89
http://dx.doi.org/10.1080/03008207.2016.1177037

REVIEW

Epigenetic regulation of bone cells


Kyung Hyun Park-Mina,b
a
Arthritis and Tissue Degeneration Program and David C. Rosensweig Center for Genomics Research, Hospital for Special Surgery, New York,
NY, USA; bDepartment of Medicine, Weill Cornell Medical College, New York, NY, USA

ABSTRACT ARTICLE HISTORY


Bone is a major organ in the skeletal system that supports and protects muscles and other organs, Received 19 February 2016
facilitates movement and hematopoiesis, and forms a reservoir of minerals including calcium. The Revised 5 April 2016
cells in the bone, such as osteoblasts, osteoclasts, and osteocytes, orchestrate sequential and Accepted 6 April 2016
Published online 17 May
balanced regulatory mechanisms to maintain bone and are capable of differentiating in bones.
2016
Bone development and remodeling require a precise regulation of gene expressions in bone cells,
a process governed by epigenetic mechanisms such as histone modification, DNA methylation, KEYWORDS
and chromatin structure. Importantly, lineage-specific transcription factors can determine the DNA methylation; epigenetic
epigenetic regulation of bone cells. Emerging data suggest that perturbation of epigenetic regulation; histone
programs can affect the function and activity of bone cells and contributes to pathogenesis of modification; osteoclasts;
bone diseases, including osteoporosis. Thus, understanding epigenetic regulations in bone cells osteoblasts
would be important for early diagnosis and future therapeutic approaches.

Introduction MSCs have been well characterized. Osteoblasts not only


play a central role in bone formation by synthesizing
Epigenetics refers to the changes in gene expression or
multiple bone matrix proteins, but they also regulate
phenotype without alterations in the DNA sequence (1).
osteoclast maturation and bone resorption by secreting
Epigenetic regulation controls gene expression by chro-
soluble factors and by direct cell–cell interactions.
matin regulators that bind to DNA, by modifications of
Osteoclasts are bone-resorbing cells important for bone
chromatin such as histone modification and DNA methy-
homeostasis and pathological bone resorption (4–8).
lation, or by non-coding RNAs. Increasing evidence sug-
Osteoclasts are differentiated from hematopoietic stem
gests that epigenetic regulation plays an important role
cells (HSCs) of the myeloid lineage under the influence
under physiological and pathological conditions. It has
of Macrophage-colony stimulating factor (M-CSF) and
become clear that epigenetic regulation is a key molecular
receptor activator of nuclear factor kappa B ligand
mechanism by which environmental influences and cues
(RANKL). Various autocrine and paracrine factors mod-
are imprinted on DNA/chromatin and patterns of gene
ulate bone cell differentiation and function. One such
expression, responses to environmental challenges, and
factor is RANKL which is produced by several cell types,
disease causation and pathogenesis are determined (2).
including osteoblasts, activated T cells, and osteocytes.
Bone is a dynamic tissue that undergoes constant
RANKL binds to its receptor, RANK that is expressed in
remodeling and controls skeletal development and regen-
osteoclast-lineage cells and the RANKL–RANK interac-
eration. Bone remodeling is a complex process whereby
tion activates signaling pathways that induce NFATc1, a
old, mature bone is removed by bone resorption and is
master transcription factor of osteoclast differentiation.
replaced by new bone formation. Bone remodeling is
Osteoprotegerin (OPG) is secreted by osteoblasts and
required for bone homeostasis including maintaining
other cell types and functions as a decoy receptor by
appropriate bone cell physiology and bone microarchitec-
binding to RANKL, subsequently preventing the activa-
ture. It is tightly regulated by a balance between bone
tion of RANK. Thus, OPG is a key negative regulator of
resorption by osteoclasts and bone formation by osteo-
osteoclastogenesis, a property that has already been ther-
blasts (Figure 1). Osteoblasts are bone-forming cells and
apeutically exploited. In addition to RANK/RANKL/
are derived from mesenchymal stem cells (MSCs) (3).
OPG, cytokines from immune cells also influence osteo-
Various extrinsic regulators and corresponding transcrip-
clast differentiation and function; and various proteins
tion factors important for osteogenic differentiation of
have been shown to control the bone integrity.

CONTACT Dr. Kyung Hyun Park-Min ParkminK@HSS.EDU Hospital for Special Surgery, Arthritis and Tissue Degeneration, 535 East 70th street, S
building 8th floor, New York, NY 10021-4898 USA.
© 2017 Taylor & Francis
EPIGENETIC REGULATION OF BONE CELLS 77

MSC
HSC
!"
pre-osteoblasts
RUNX2
OSX PU.1
HDAC1-8 FOS
ATF4
M-CSF NFATC1
TWIST1 DNMT1
DLX5 DNMT3B HDAC3
OCPs HDAC7
Bone-lining cells DNMT3A
RANKL
Osteoblasts

Osteocytes Osteoclasts Bone

Figure 1. The process of bone remodeling and epigenetic factors. Bone is maintained by bone remodeling which is a continuous cycle of
bone resorption and bone formation. Bone remodeling is a coupled and balanced process and takes place in basic multicellular units. In a
resting stage, the non-remodeling bone surface is covered by bone lining cells. Osteocytes sense bone damage and recruit osteoclast
precursor cells (OCPs), which further differentiate into mature osteoclasts. Osteoclasts resorb a damaged or old bone matrix and
subsequently osteoblasts are recruited into resorbed sites and form new bone. Osteoclasts are differentiated from HSCs. M-CSF and
RANKL are key drivers of osteoclastogenesis. M-CSF enables HSCs to differentiate into OCPs which express RANK, a receptor for RANKL.
RANKL further promotes OCPs into mature osteoclasts. Mesenchymal stem cells (MSCs) differentiate into osteoblasts and classical coupling
factors, which are released from the resorbed bone matrix or produced by osteoclasts, promote osteogenic differentiation through the
osteoblast lineage. Key transcription factors (orange boxes), epigenetic enzymes that participate in DNA methylation (blue boxes), and
histone deacetylases (yellow boxes) are indicated and described in the text.

Over the past years, substantive progress in epigenetic the organism. This diversity results from the capacity of
research on osteoclasts, osteoblasts, and their differentia- individual cell types to initiate and then maintain spe-
tion from HSCs and MSCs, respectively, has been made, cific gene expression patterns during development. To
highlighting the importance of epigenetic regulation in achieve this, cellular signaling events are thought to
bone remodeling. The impairment of bone remodeling is regulate the activity of cell type-specific DNA-binding
associated with many skeletal diseases such as osteoporo- transcription factors that function as master regulators
sis and, to date, anti-resorptive or osteoanabolic treat- of gene expression networks. Recently, epigenetic reg-
ments have been developed but all have significant side ulation has also been identified as an important factor
effects limiting their prolonged use. Investigating epige- that is involved in the regulation of gene expression
netic regulations may provide novel treatment targets for and cell fate decision.
promoting bone formation and suppressing excessive Due to their unique properties of self-renewal and
bone resorption. Understanding the epigenetic regulation their ability to undergo multilineage differentiation in
during physiological bone remodeling further provides response to appropriate signaling cues, pluripotent
insights into clinical and mechanistic characterization of embryonic stem cells (ESCs) are widely used to study
bone metabolism and into pathophysiology of bone dis- epigenetic mechanisms and show various characteristic
eases. Although osteocytes are recognized as important epigenetic phenotypes based on prevalence of different
regulators of bone turnover, this review summarizes cur- histone markers (9)]. A public research consortium
rent knowledge of epigenetic mechanisms in relation to called the Encyclopedia of DNA Elements (ENCODE)
the mechanisms regulating the differentiation of osteo- (10) and the Epigenomics Roadmap (11) carried out a
clasts and osteoblasts, and how these regulations can be project to identify all functional elements in the human
applied to therapies for bone diseases. genome sequence and generated an epigenomic map.
These data allow researchers to understand molecular
and cellular processes of particular cell types and to
Epigenetic regulation
elucidate fundamental processes of human diseases at a
A notable feature of multicellular organisms is their genomic level. Extensive epigenomic datasets of human
capacity to create functionally unique cell types from osteoblasts are currently available in the ENCODE
the same genome sequence that is shared by all cells in database and the chromatin landscape of human
78 K. H. PARK-MIN

osteoblasts has been shown by studies of the ENCODE (19). Other transcription factors have been identified
consortium. to control osteoblast differentiation, including Osterix
Beyond the sequences of the genome, a large regu- (OSX)(30), ATF4(31), twist homolog 1 (TWIST1)(32),
latory network controls cell specific identity and func- and distal-less homeobox 5 (DLX5)(33).
tion of cells. Each cell type has its own unique Various transcription factors coordinate a broad
chromatin structure and organization. Whereas spectrum of gene expression programs in osteoclasto-
mRNA expression profile indicates the current state of genesis (34). NFATc1 is a master transcriptional factor
a cell, epigenomic regulation can give perspectives of of osteoclastogenesis and drives the early stages of
history of cell states. Advanced high throughput osteoclast differentiation (35–37). NFATc1-deficient
sequencing makes the genome-wide analysis of chro- mice develop severe osteopetrosis in vivo (38), and
matin profiling feasible, resulting in generating a pro- NFATc1-deficient cells are incapable of differentiating
filing of the epigenome (12). As chromatin patterns are into osteoclasts in vitro (36,37,39). However, it has been
related to underlying regulatory processes, chromatin shown that NFATc1 forms a complex with osterix and
profiling enables identification of the regulatory net- both are required for osteoblastic bone formation (40).
work in bone cells. Increasing knowledge of regulatory These data suggest that NFATc1 cooperatively regulates
network in bone cells will reveal a cell-type specific osteoclastogenesis by interacting with other transcrip-
chromatin structure and organization leading to affect- tion factors. c-Fos is another essential transcription
ing cell-specific gene regulation. factor for osteoclastogenesis. c-Fos belongs to the AP-
1 family and c-Fos-deficient cells can differentiate into
macrophages but not into osteoclasts, suggesting the
Transcription factor network
role of c-Fos in osteoclastogenesis (41,42). The cis-
Transcription factors bind to specific DNA sequences regulatory elements enriched with binding sites of tran-
(called binding motifs) in promoter or enhancer scription factors and the lineage determining transcrip-
regions to regulate gene expression. Transcription fac- tion factors play an important role in pioneering the
tor networks and transcriptional processes help shape activity of transcription. The subsequent transcriptional
epigenetic regulation as well as the chromatin structure program sets the cell-specific gene expression program
(13). However, transcription factor binding to the pro- and establishes dynamic epigenetic states. Therefore,
moter is insufficient to fully regulate transcription and the complicated interaction between transcription fac-
the interaction between the promoter and the regula- tors and epigenetic regulation endow cell identity to
tory elements, called enhancers, provide another level bone cells.
of dynamic regulation of transcription (14). Unlike
other cells in mammals, interaction with bone and
Histone modification
bone marrow environments would be an important
factor to control the gene expression profile in bone The basic unit of chromatin is the nucleosome (43).
cells (15,16). Each nucleosome consists of a short (around 147 base
Molecular and biochemical studies reveal principal pair) DNA segment wrapped around a histone octamer
regulators of transcription in bone cells, while ChIP- that comprises two copies each of core histones: H2A,
based assays have mapped the binding sites of many H2B, H3, and H4, as shown in figure 2A. The
important transcription factors. During osteoblastogen- N-terminal tails of core histones are subject to various
esis, both the transcriptome and the epigenome of post-translational modifications by chromatin remode-
osteoblast lineage cells undergo dynamic changes (17– lers (44,45). More than eight different histone modifi-
21). Several transcription factors are known to control cations are identified, including acetylation,
bone development and osteoblast differentiation (22). methylation, phosphorylation, ubiquitination, and
Runt-related transcription factor 2 (RUNX2, Cbfa1) is a sumoylation. A large-scale mapping of histone modifi-
master transcription factor of osteoblasts and is indis- cations and related chromatin structure enables the
pensable in all stages of osteoblast differentiation (23). characterization and determination of functional con-
RUNX2 ablation causes bone defects in humans and sequences of changes in the chromatin structure
mice (24–26). RUNX2 regulates gene expression during (46,47). Currently, more than 14,000 datasets are
osteoblastogenesis and detailed epigenetic regulation deposited in the public domain such as Gene
has been identified for RUNX2-mediated gene expres- Expression Omnibus (GEO), allowing computational
sion (27–29). In MC3T3-E1 osteoblast-like cells, ChIP- integration of different datasets. These integrated
sequencing demonstrated that Runx2 occupancy is approaches can identify novel interactions between his-
actively regulated during osteoblast differentiation tone modification and genomic elements and supports
EPIGENETIC REGULATION OF BONE CELLS 79

A Histone complex Histone tails


H3K27Ac
(H2A, H2B, H3, and H4) H3K27Ac
H3K4me3
H3K4me3

Transcription
Factors

Transcription Factor
Binding Motifs

Nucleosome
Open chromatin
Promoters or Regulatory elements

B
CTCF CTCF

P GENE E P GENE
H3K27me3 H3K4me1 H3K4me3
H3K9me H3K27Ac H3/H4Ac

Figure 2. A. The core proteins of nucleosomes are two copies of H2A, H2B, H3, and H4 and 146 base pairs of DNA wraps around
these core histone octamer. Core histones are highly conserved and have amino-terminal tails which are subject to various post-
translational modifications. Histone modifications, such as methylation and acetylation, play an important role in gene expression
and active promoter regions are distinguished by specific histone modifications including H3K4me3 and H3K27Ac. Nucleosomes are
depleted in highly active regions, called “open chromatin”. Transcription factors and lineage determining factors such as NFAT and
RUNX2 bind to a specific binding motif in promoters or enhancers. Ac, acetylation; Me, methylation. B. Histone modifications of
functional elements including promoters, enhancers, and insulators. Insulator is a genetic boundary element that blocks the
interaction between enhancers and promoters and is defined by CCCTC-binding factor (CTCF) binding although CTCF has dual
effects on enhancers; either blocking or activating. Active promoters are enriched for H3K4me3 and H3/H4 acetylation. H3K9 me2/3
and H3K27me3 are associated with repressed promoter regions. Active enhancers are marked by H3Kme1 and H3K27Ac.

the idea that a different combination of histone mod- (reviewed (52)). Histone acetylation by HATs plays a
ifications serves as a “switch” to fine-tune genomic key role in transcriptional activation, whereas deacety-
elements (48,49). Unique patterns of histone modifica- lation of histones promotes transcriptional repression
tions are found in different genomic elements such as and silencing of genes. However, dysregulated acetyla-
promoters, gene bodies, enhancers, and chromatin tion of histone is often associated with the development
insulators (boundary elements) and reflect the status of a wide variety of human cancers. An excessive level
of transcription (Figure 2B). For example, active pro- of histone acetylation induces apoptotic cell death,
moters are enriched in histone H3 lysine 4 dimethyla- whereas extensive histone deacetylation has been linked
tion (H3K4me2), H3 lysine trimethylation (H3K4me3), to the repression of tumor regulatory genes promoting
histone acetylation (H3Ac and H4 Ac), and H2A.Z. cancer pathologies. Recruitment of HATs and subse-
Histone modifications are dynamically regulated and quent histone acetylation are dynamically regulated
have an influence on the chromatin structure and func- during the differentiation of bone cells (53,54) and
tion (45). Charting different combination of histone intervening histone acetylation by small molecule inhi-
modifications in the chromatin can predict the stage bitors results in altering the differentiation of bone
of transcription at a specific gene (49). Histone mod- cells.
ification is mediated by three components: writers, Due to the importance of histone acetylation, var-
erasers, and readers. Histone acetylation is an impor- ious small molecule inhibitors which target HATs or
tant determinant in multiple chromatin-dependent HDACs have been developed and contain therapeutic
processes, including gene expression, DNA replication, potential for various diseases. HDACs function as “era-
and DNA damage repair. Acetylation is generally asso- sers” which remove an acetyl group from histones and
ciated with elevated gene expression and open chroma- also have activity on non-histone proteins (55). HDACs
tin by reducing a positive charge of histone. The N-ε- are comprised of three classical classes (I, II, and IV)
lysine acetylation and deacetylation of histones are also that have 11 HDACs and class III (sirtuins) that con-
controlled by three groups of enzymes: histone acetyl- tain NAD-dependent catalytic sites. HDAC inhibitors
transferase (HAT, reviewed (50)), histone deacetylase (HDACi) cause an increase in the acetylated level of
(HDAC, reviewed (51)), and histone acetylation readers histones, which in turn promotes the re-expression of
80 K. H. PARK-MIN

the silenced regulatory genes in cancer cells and reverse proteins are readers of acetylated histones as well as
the malignant phenotype and thus HDAC inhibitors other acetylated proteins such as NF-kB and regulate
have recently emerged as potential cancer therapeutic gene expression (73). BET proteins are often deregu-
agents. Two HDAC inhibitors, vorinostat and romidep- lated in cancers leading to an aberrant expression of
sin, have received an approval from the US FDA for the proinflammatory cytokines and growth-promoting
treatment of cancer and certain neurological genes. Small molecule inhibitors of BET family proteins
conditions. have demonstrated efficacy in the treatment of cancers
Many studies testing the effect of HDAC inhibitors (73–79). We and others reported that BET inhibitors,
(HDACi; TSA, SAHA, MS-275, sodium butyrate, and I-BET151 and JQ1, effectively suppress osteoclastogen-
valproic acid) on osteoblasts and osteoclasts have been esis, attenuate inflammatory bone loss, and protect
documented, implicating that downregulation or dele- mice from ovariectomy-induced bone loss as well as
tion of HDACs may play a role in the differentiation of tumor metastasis induced osteolysis (54,80).
osteoclasts and osteoblasts. Histone acetylation of Interestingly, BET inhibitors also suppress osteoblast
RUNX2 is one of the key regulatory mechanisms for differentiation, but sensitivity to inhibitors of osteo-
RUNX2-mediated gene programs (56). HDAC inhibi- blasts is lower than in osteoclasts (54 80). These results
tors show positive effects on in vitro osteoblast differ- indicated that proper dosing of BET inhibitors can
entiation and maturation, in part, mediated by show beneficial effects on pathologic bone diseases,
promoting RUNX2 function. The treatment with a despite ubiquitous expression of BET proteins.
HDAC inhibitor promotes osteoblastogenesis (57–63) Histone methylation occurs on basic residues such as
and also increases genome-wide distribution of H4 lysine and arginine and plays an important role in
acetylation (64). A conditional deletion of HDAC3 in controlling gene expression. Histone methylation has
osteochondroprogenitor cells results in osteopenia and been shown to display differential effects on transcrip-
increases marrow adiposity (65). Resveratrol or isoni- tion (81). For example, trimethylation of H3K4,
cotinamide is an agonist of Sirtuin 1, class III HDAC, H3K36, and H3K79 positively controls gene expression.
and blocks spontaneous adipocyte differentiation and In contrast, trimethylation of H3K27, H3K9, and
thereby promotes osteoblast differentiation and bone H4K20 is associated with repression. The protein
mineralization (66). methylation process was thought to be irreversible
Although HDAC inhibitors suppress osteoclastogen- because the half-life of methylation is similar to that
esis (67), the deletion of individual HDAC shows dif- of histone. However, the discovery of histone demethy-
ferential effects on osteoclast differentiation depending lases had changed this paradigm to indicate that his-
on the class. The downregulation of HDAC3 by shRNA tone methylation is dynamically regulated. Two
suppresses osteoclastogenesis (68). However, Stemig families of lysine demethylases are identified: the
et al showed that a conditional deletion of HDAC7 in amine oxidases (LSD1 (82)) and jumonji C (JmjC)-
osteoclasts causes osteopenia (69), suggesting that domain-containing family (83). JMJD3 is a H3K27
HDAC7 functions as an inhibitor of osteoclastogenesis. demethylase (84). Yasui et al has shown that RANKL
Although 24% of 41 patients with epilepsy who stimulation removes trimethylation of H3K27, a repres-
received HDACi, valproate, experienced osteopenia, sive mark from the promoter of NFATC1 by, in part,
there was no significant correlation between HDACi inducing JMJD3, demonstrating the dynamic regula-
treatment and bone density, suggesting that bone meta- tion of histone methylation during osteoclast differen-
bolism may not be significantly affected by valproate tiation (85). JMJD3 expression is also induced during
monotherapy (70). Thus, a clinical association between osteoblast differentiation and regulates the expression
HDACi and bone metabolism is not clear yet. of bone-related genes including Runx2, osterix, osteo-
The readers of histone acetylation contain a bromo pontin, bone sialoprotein (BSP), and osteocalcin
domain (71), a tandem PHD domain (72) that recog- (OCN) (86). Consistently, Jmjd3−/− embryos exhibited
nizes acetylated lysines (52). The readers of histone open fontanelles, less-mineralized cranial bones, and
modification are regulated by non-coding RNAs, bind- hypoplastic clavicles compared to wild type littermates
ing partners, and conformational changes of readers. at E18.5, suggesting that JMJD3 is required for osteo-
After recognizing modified histones, the readers func- blast maturation during both intramembraneous and
tion as chromatin architectural proteins, chromatin endochondral bone formation (87).
remodelers, and chromatin modifiers and recruit The recruitment of histone demethylases is mediated
other components to regulate transcription, repair, by specific DNA sequences such as the polycomb
recombination, replication, and RNA processing (52). responsive element (PRE), cofactors, and DNA-binding
Bromodomain and extra terminal domain (BET) family factors. Polycomb proteins are transcriptional
EPIGENETIC REGULATION OF BONE CELLS 81

repressors essential for maintaining tissue specific gene across a variety of cell types (96,97) and quantifying
expression program and consist of two groups, poly- changes that lead to both activation and repression of
comb repressive complex 1 (PRC1), and polycomb gene expression. Open chromatin has been first identi-
repressive complex 2 (PRC2) (88). PRC1 and PRC2 fied by its sensitivity to cleavage by non-specific
target promoters are responsible for methylation of nucleases such as deoxyribonuclease I (DNase I) and
H3K27 that is known as a repressive epigenetic mark micrococcal nuclease (MNase) at a genome-wide level
and mediates transcriptional repression in cancer cells (98). Several methods to map open chromatin are devel-
(89). PRC1-type complexes also contain an ubiquitin oped including formaldehyde-assisted isolation of regu-
ligase activity for monoubiquitination of histone H2A latory elements (FAIRE) (99), and sonication of cross-
to promote H3K27 trimethylation (90). The enhancer linked chromatin sequencing (Sono-seq) (100). Assay for
of zeste 2 (EZH2) is a methyltransferase and the cata- transposase-accessible chromatin using sequencing
lytic subunit of the PRC2 and trimethylates H3K27 and (ATAC-seq) is a newly developed method to detect
plays an essential role in the epigenetic repression open chromatin faster and with a higher sensitivity and
(89,91). Wei et al reported that EZH2 suppresses applying the preferential integration of transposons into
mesenchymal stem cells (MSCs) from differentiating the nucleosome-depleted region to interrogate the chro-
into osteoblasts (92). Recently, Dudakovic et al demon- matin structure using Tn5 transposase (101). Measuring
strated the expression profiles of epigenetic regulators open chromatin is a powerful tool for identifying regu-
during osteogenic differentiation of human mesenchy- latory elements including transcription factors and
mal cells derived from the stromal vascular fraction of enhancers. Binding of proteins, such as transcription
adipose tissue (AMSCs) and show that EZH2 is down- factors to genomic DNA, protects the underlying
regulated during osteoblastogenesis (64)]. Whereas sequences from cleavage by DNase I. DNase I footprint-
EZH2 inhibition suppresses adipogenic differentiation, ing assay allows identification of transcription factor–
EZH2 functions as a repressor of osteoblastogenesis, DNA interaction at single base pair resolution and pro-
supported by experiments using the EZH2 inhibitor, vide information on regulatory elements (102–103).
siRNAs, and knock-out mice. WD repeat domain 5 Identification of open chromatin regions further allows
(WDR5) is another histone methyltransferase and is a predicting cell-type specific function by patterning of
component of the mixed lineage leukemia (MLL) com- chromatin accessibility (96).
plex, which methylates lysine 4 of histone H3. In con- Open chromatin regions have been identified in
trast to EZH2, WDR5 accelerates osteoblast osteoblasts and osteoclasts. Tai et al performed DNase
differentiation (93–95). Osteoblasts with reduced seq analysis during osteoblast differentiation including
expression of WDR5 exhibit persistent acceleration of the growth phase, matrix deposition stage, and miner-
osteoblast differentiation through the activation of the alization stage (GEO55046) and identified a role of a
canonical Wnt pathway. Altogether, dynamic regula- long-range interaction between bone specific RUNX2
tion of histone methylases and demethylases is required P1 promoter–Supt3h (suppressor of Ty 3 homolog)
for skeletal development and histone lysine methylation promoters in murine pre-osteoblastic MC3T3-E1 cells
plays an important role in bone remodeling by control- using DNase seq in combination with the ENCODE
ling the differentiation of both osteoclasts and osteo- dataset (104,105). Although these interactions are
blasts although their specific function needs to be detected in cells in which RUNX2 is silent, the interac-
further clarified. tion frequency between RUNX2 and Supt3h is signifi-
cantly increased during osteoblastogenesis as shown by
chromosome conformation capture (3C) analysis, sug-
Open chromatin
gesting Supt3h as a potential regulator of the bone
In order for transcripton factors to access and interact specific RUNX2 P1 promoter. DNase-seq analysis also
with genomic regulatory elements, local chromatin detected that RANKL facilitates the dynamic changes of
needs to undergo rearrangement. Nucleosomes are able chromatin accessibility in RAW 264.7 cells, a murine
to change their location and are depleted in highly active monocytic cell line that is capable of differentiating into
regulatory regions, called “open chromatin”(96). DNA in osteoclasts upon RANKL stimulation (106). Using
open chromatin regions has higher accessibility to pro- motif discovery analysis, Inoue et al further profiled
teins including transcription factors and open chromatin the transcription motifs in open chromatin regions
has a signature of active histone modification. Combined and identified novel players for osteoclastogenesis in
with next-generation sequencing, methods for searching addition to other well-defined osteoclastogenic tran-
genome-wide DNA accessibility have been proven to be scription factors. Therefore, linking DNase seq foot-
extremely effective in identifying regulatory elements printing analysis with cell-specific gene expression
82 K. H. PARK-MIN

profiles will expand our knowledge of a complicated cells and the importance of DNMTs in skeletal biol-
transcriptomic network of bone cells. ogy. Recently, Nishikawa et al have demonstrated that
DNMT3a is needed for osteoclast differentiation and
bone resorption by suppressing IRF8, a negative reg-
DNA methylation
ulator of osteoclast differentiation. DNMT3a sup-
DNA methylation is a reversible and covalent modifi- presses IRF8 via increased methylation on distal
cation of the 5’-carbon of the cytosine residue with regulatory elements of IRF8; methylation was pro-
addition of the methyl group from SAM and occurs moted by increased S-Adenosyl methionine (SAM)
mostly within CpG dinucleotides that cluster as a concentration. Both conditional osteoclast-specific
region called CpG islands in mammal (107,108). DNA DNMT3a deletion and treatment of mice with TF-3,
methylation can be mapped at base-pair resolution with a DNMT3 inhibitor, protected mice from ovariect-
bisulfite sequencing (109). In general, DNA methyla- omy-induced bone loss, supporting the potential role
tion establishes the pattern of a repressive state of gene of epigenetic therapy in bone diseases. 5-aza-cytidine,
expression by maintaining a stable condensed chroma- an another DNMT inhibitor, inhibits differentiation
tin configuration. DNA methylation is preserved dur- of MSCs into osteoblasts, adipocytes, and chondro-
ing DNA replication and mitosis and thus a repressive cytes (116), suggesting DNMT regulates the differen-
state of genome can be inherited into the next genera- tiation potential of MSCs. Consistently, oxidized
tion. Local deposition and removal of DNA methyla- adenosine (ADOX), an inhibitor of SAM-dependent
tion is tightly coupled with occupancy of transcription methyltransferases suppresses osteoblast differentia-
factors (110). It has been suggested that DNA methyla- tion (117). Bone mineral density and body weight
tion-mediated silencing plays a role in the expression of decrease with aging by reduced DNMT1 activity
important osteogenic transcriptional factors. During (118) and differentially methylated regions are
osteoblast differentiation, osteocalcin expression is enriched in genes associated with cell differentiation
increased but methylation in the promoter of osteocal- and skeletal embryogenesis in bone samples from
cin is significantly decreased. However, CpG methyla- patients with osteoporotic hip fracture (119). These
tion of the osteocalcin promoter does not alter binding data suggest that DNA methylation plays an impor-
of RUNX2, basal transcriptional activity, or vitamin-D tant role in aging (116) and changes of DNA methy-
mediated enhancement of OC genes, supporting an lation can be linked to disease pathogenesis of age-
indirect association between the OC promoter and associated bone diseases. In addition to DNMTs, the
CpG methylation (111)]. Hagh et al has shown that Ten-Eleven Translocation (TET) proteins are recently
the methylation status of promoters of RUNX2, discovered 5-methylcytosine hydroxylases that convert
DLX5, and BSP remains unchanged during osteoblastic 5-methylcytosine (5mc) to (5hmc). It has been sug-
differentiation. In contrast, the OSX promoter showed gested that 5-hydroxymethylcytosine (5hmC) plays a
a dynamic change in its methylation pattern (112). role as an early intermediate in active DNA demethy-
DNA methylation is also not overlapped with active lation (120). Although there is no direct evidence of
and open chromatin architecture, supporting a negative association of TET proteins with bone cells, the
correlation between H3K4me3 and DNA methyla- increase in AP activity and matrix mineralization in
tion (113). Ad-MSCs has been shown to be associated with an
Enzymes responsible for DNA methylation are increased presence of 5hmC as well as with an
DNA methyltransferases (DNMTs), which consist of increased TET2/3 gene expression (121). DNA methy-
DNMT1, DNMT3a, and DNMT3b. DNMT3a and 3b lation changes during osteoclast differentiation are
are de novo DNA methyltransferases and transfer also highly associated with TET2-mediated demethy-
methyl groups to unmethylated sites. Mice with lation and DNMT3b-mediated methylation (122).
DNMT deficiencies are embryonic lethal with reduced Although DNA methylation is important for the dif-
DNA methylation (114,115). These studies reveal that ferentiation of bone cells, the role of DNA methyla-
DNA methylation is essential for mammalian devel- tion in bone cells remains elusive and needs further
opment, suggesting methylation patterns are pre- investigation.
deposited during development. Genome-wide sequen-
cing in wild-type mouse ESCs reveals that most
Non-coding RNAs
methylated genes are differentiation-associated genes,
which are repressed in mouse ESCs. The cell-specific Only 1–2% of the human genome is translated into
roles of DNMT3a and 3b have been investigated, protein (123,124) and the remaining transcribed
showing that DNMTs have a large impact on bone RNAs are not annotated into protein-coding genes.
EPIGENETIC REGULATION OF BONE CELLS 83

The transcripts from non-coding regions of genomes have shown that osteoclast-specific DICER deletion by
are called non-coding RNAs (ncRNAs) (125,126). Non- cathepsin K cre also increases bone mass and, surpris-
coding RNAs have essential roles in regulating gene ingly, decreased osteoblastogenesis in addition to inhibi-
expression as well as other ncRNAs both in cis and in tion of osteoclastogenesis (142). These studies point to
trans. It has been shown that non-coding RNAs affect the potential functions of miRNAs from osteoclasts on
various biological processes during development and in osteoblast differentiation. Indeed, miRNAs secreted from
some pathological conditions. Importantly, ncRNAs are osteoclasts are delivered into osteoblasts and regulate
expressed in a stage specific and cell specific manner osteoblast differentiation. miRNAs are regulated by
and provide another view of the transcriptional circuit. important signaling pathways such as WNT, RANKL,
The major classes of ncRNAs are micro RNAs and BMP in osteoclasts and osteoblasts and inversely
(miRNAs) and long non-coding RNAs (lncRNAs). control and target multiple signaling pathways in osteo-
lncRNAs are non-coding transcripts length over 200 clasts and osteoblasts. In addition to the in vitro studies
nucleotides long (126) and are involved in many biolo- that identified the regulation and function of miRNAs in
gical functions (127). More than 28,000 distinct osteoblast and osteoclast differentiation (144–147), sev-
lncRNA transcripts are currently identified and eral studies have examined the expression of miRNAs
lncRNAs regulate gene expression by various mechan- during bone development in vivo and skeletal diseases.
isms (128). Large intergenic non-coding RNAs Extracellular miRNAs have recently been found in a
(lincRNAs) are the largest class of lncRNAs and other variety of body fluids including synovial fluids, plasma,
classes of lncRNAs are intronic lncRNA, sense lncRNA, serum, saliva, and urine. Emerging evidence shows that
and antisense lncRNA. Tong et al recently reported that miRNAs can be used a diagnostic tool to detect skeletal
lncRNA-DANCR expression is unregulated in circulat- diseases such as osteoporosis. Osteoporosis is character-
ing monocytes in patients with low-BMD and DANCR ized by low bone mass that results from imbalanced
promotes the expression of inflammatory cytokines bone remodeling: high osteoclast activity and low osteo-
such as IL-6 and TNF (129). LncRNA-DANCR directly blast activity (148). Bone fractures associated with osteo-
interacts with EZH2 and suppresses RUNX2 expression porosis can be predicted by measuring bone mineral
and osteoblast differentiation (130). Although lncRNAs density (BMD) using dual-energy X-ray absorptiometry
have been shown to engage in various biological pro- (DXA) and clinical history (149,150). However, current
cesses, the identity of lncRNAs in skeletal biology is not methods have a low detection rate and thus improved
well known (131). However, many studies about the fracture risk assessment methods are required. miRNAs
function and regulation of miRNAs in skeletal biology can be extruded into extracellular space, supporting the
have been reported (reviewed in (132,133)). Thus, this idea that miRNAs may function as biomarkers. Several
review briefly discusses the recent prospective view of miRNAs are detected in the blood of osteoporosis
miRNAs in bone diseases. patients, although the function of individual miRNAs is
MicroRNAs (miRNAs) are a class of short non-cod- not well characterized in the pathogenesis of osteoporo-
ing RNAs that mediate post-transcriptional silencing sis. A recent study shows that nine cell free miRNAs in
and are important for cell differentiation, embryonic the serum are significantly upregulated in patients with
organ development, and apoptosis (134–137). miRNAs osteoporotic fractures, opening up a possibility of using
control the expression of more than 30% of genes and miRNAs as a biomarker for diagnosing osteoporotic
have profound effects on physiological and pathological fracture (151). In addition, abnormal miRNA expres-
conditions. miRNAs are transcribed as primary miRNAs sions are often found in several diseases such as cancer,
(pri-miRNAs) containing a secondary loop structure, a leading to the first miRNA mimics reaching the clinical
5-cap structure and a poly(A) tail (138). Pri-mRNAs are phase I trial in patients with primary liver cancer in 2013
processed to pre-miRNA by Drosha (nuclear RNase III) (152). Since one miRNA can interact with many genes
and cofactor, DGCR8. Dicer (cytosolic RNase III) simultaneously, miRNA replacement therapy using syn-
sequentially processes pre-miRNAs to a mature form. thetic miRNA mimics would be an option to treat dis-
miRNAs are regulated during bone development (139) eases, if it can avoid off-target effects.
and Dicer inactivation, which results in defects on
miRNAs cleavage, regulates bone mass, suggesting that
Conclusions
Dicer-generated miRNAs play an important role in
osteoblastogenesis and osteoclastogenesis (140–143). The crucial role of bone cells in bone homeostasis and
Whereas DICER deficiency inhibits both in vitro and in skeletal diseases makes them potential therapeutic
in vivo osteoclastogenesis, CD11b-specific DICER null targets. For example, understanding the osteogenic
mice exhibit mild osteopetrosis (143). Mizoguchi et al. potential of MSCs can be used to treat bone defect
84 K. H. PARK-MIN

repair and bone diseases (153). Suppressing osteoclast osteoblast-specific precursors from stem cells. Specific
activity or differentiation allows one to block patholo- populations have been sorted using flow cytometry
gical bone resorption. Epigenetics regulations were techniques. However, single cell ChIP sequencing or
implicated in skeletal diseases by a few studies RNA sequencing would provide an advantage over
(119,151), suggesting that epigenetic changes can be isolating cells by flow cytometry and would allow
predisposited or used as a signature of skeletal diseases. investigation of a larger number of genes or epige-
Epigenetic regulation affects differentiation and activity netic regulation in a single cell which has the poten-
of bone cells and the response of bone cells to exogen- tial for differentiation into bone cells. Thus, single cell
ous stimulus. Understanding bone cell-specific epige- sequencing techniques will allow for identifying bone-
netic regulation using genome-wide sequencing cell specific epigenetic mechanisms and for detailed
technology will provide the foundation for cell-specific new information about cellular differentiation or dis-
therapeutic approaches. Recent drug development tar- ease in different biological contexts.
geting epigenetic mechanisms shows a promising pro-
spect for the treatment of cancers and inflammatory
conditions. The development of epigenetic drugs or Declaration of interest
therapies that changes epigenetic states will contribute The author reports no conflicts of interest. The author alone
to such treatments. In this review, we provide a brief is responsible for the content and writing of the article.
overview of epigenetic regulations in bone cells but the
understanding of chromatin landscapes in bone cell is
still incomplete. Acknowledgement
Over the past years, the study of histone post-trans-
I thank Drs. Lionel Ivashkiv, Mary-Beth Humphrey, and
lational modifications (PTMs) has made progress to Sungho Park for discussions and critical review of the
show the association of epigenetic modifications with manuscript.
fundamental biological processes and pathological con-
ditions. During the differentiation of bone cells, histone
PTMs are dynamically regulated. Recent advances in Funding
developing small molecules to modulate pharmacologi-
The research was supported by the National Institute of
cally epigenetic enzymes and interfere with these bio- Arthritis and Musculoskeletal and Skin Diseases (NIAMS,
chemical mechanisms offer great promise for therapy of R00AR061430).
bone diseases. In this article, we provide an overview of
modulating histone PTMs by small molecule inhibitors
including HDAC inhibitor and BET inhibitor which References
provides elucidation of the basic epigenetic regulation
1. Probst AV, Dunleavy E, Almouzni G. Epigenetic
of bone cells and devising future epigenetic therapy for
inheritance during the cell cycle. Nat Rev Mol Cell
bone diseases. Biol 2009;10:192–206.
Recent progress in genome-wide technologies 2. Jaenisch R, Bird A. Epigenetic regulation of gene
using small cell numbers has allowed researchers to expression: how the genome integrates intrinsic and
capture transcriptional and epigenetic snapshots of environmental signals. Nat Genet 2003;33
cells from small or rare biological samples. These Suppl:245–54.
3. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas
new methods allow identifying single-cell transcrip- R, Mosca JD, Moorman MA, Simonetti DW, Craig S,
tional and epigenomic variability across several cell Marshak DR. Multilineage potential of adult human
types and suggest that controlling single-cell variance mesenchymal stem cells. Science 1999;284:143–7.
is a fundamental characteristic of different biological 4. Takayanagi H. Osteoimmunology: shared mechanisms
states (154). Heterogeneity within cellular populations and crosstalk between the immune and bone systems.
Nat Rev Immunol 2007;7:292–304.
has been obvious since the first microscopic observa-
5. Lorenzo J, Horowitz M, Choi Y. Osteoimmunology:
tion of individual cells. Dissecting single-cell based interactions of the bone and immune system. Endocr
epigenetic heterogeneity and linked cis- and trans- Rev 2008;29:403–40.
effectors to variability in chromatin accessibility pro- 6. McInnes IB, Schett G. The pathogenesis of rheumatoid
vides new opportunities for studying multiple differ- arthritis. N Engl J Med 2011;365:2205–19.
ent layers of regulation and advances our 7. Novack DV, Teitelbaum SL. The osteoclast: friend or
foe? Annu Rev Pathol 2008;3:457–84.
understanding of physiological development and the 8. Weilbaecher KN, Guise TA, McCauley LK. Cancer
changes in pathological conditions and aging. Much to bone: a fatal attraction. Nat Rev Cancer
effort has been made to identify osteoclast-specific or 2011;11:411–25.
EPIGENETIC REGULATION OF BONE CELLS 85

9. Ng HH, Surani MA. The transcriptional and signalling Detection of novel skeletogenesis target genes by com-
networks of pluripotency. Nature cell biology prehensive analysis of a Runx2(-/-) mouse model. Gene
2011;13:490–6. Expression Patterns : GEP 2007;7:102–12.
10. Stamatoyannopoulos JA. What does our genome 26. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K,
encode? Genome Res 2012;22:1602–11. Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M,
11. Bernstein BE, Stamatoyannopoulos JA, Costello JF, Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto
Ren B, Milosavljevic A, Meissner M, Kellis M, Marra T. Targeted disruption of Cbfa1 results in a complete
MA, Beaudet AL, Ecker JR, Farnham PJ, Hirst M, lack of bone formation owing to maturational arrest of
Lander ES, Mikkelsen TS, Thomson JA. The NIH osteoblasts. Cell 1997;89:755–64.
Roadmap Epigenomics Mapping Consortium. Nat 27. Bradley EW, McGee-Lawrence ME, Westendorf JJ.
Biotechnol 2010;28:1045–8. Hdac-mediated control of endochondral and intra-
12. Zentner GE, Henikoff S. High-resolution digital profil- membranous ossification. Crit Rev Eukaryotic gene
ing of the epigenome. Nat Rev Genet 2014;15:814–27. Expression 2011;21:101–13.
13. Li B, Carey M, Workman JL. The role of chromatin 28. Pelletier N, Champagne N, Stifani S, Yang XJ. MOZ
during transcription. Cell 2007;128:707–19. and MORF histone acetyltransferases interact with the
14. Voss TC, Hager GL. Dynamic regulation of transcrip- Runt-domain transcription factor Runx2. Oncogene
tional states by chromatin and transcription factors. 2002;21:2729–40.
Nat Rev Genet 2014;15:69–81. 29. Villagra A, Cruzat F, Carvallo L, Paredes R, Olate J, van
15. Purdue PE, Crotti TN, Shen Z, Swantek J, Li J, Hill J, Wijnen AJ, Stein GS, Lian JB, Stein JL, Imbalzano AN,
Hanidu A, Dimock J, Nabozny G, Goldring SR, Montecino M. Chromatin remodeling and transcrip-
McHugh KP. Comprehensive profiling analysis of tional activity of the bone-specific osteocalcin gene
actively resorbing osteoclasts identifies critical signal- require CCAAT/enhancer-binding protein beta-depen-
ing pathways regulated by bone substrate. Sci Rep dent recruitment of SWI/SNF activity. J Biol Chem
2014;4:7595. 2006;281:22695–706.
16. Crane JL, Cao X. Bone marrow mesenchymal stem 30. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM,
cells and TGF-beta signaling in bone remodeling. J Behringer RR, de Crombrugghe B. The novel zinc
Clin Invest 2014;124:466–72. finger-containing transcription factor osterix is
17. Meyer MB, Benkusky NA, Lee CH, Pike JW. Genomic required for osteoblast differentiation and bone forma-
determinants of gene regulation by 1,25-dihydroxyvi- tion. Cell 2002;108:17–29.
tamin D3 during osteoblast-lineage cell differentiation. 31. Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC,
J Biol Chem 2014;289:19539–54. Schinke T, Li L, Brancorsini S, Sassone-Corsi P,
18. Meyer MB, Benkusky NA, Pike JW. The RUNX2 cis- Townes TM, Hanauer A, Karsenty G. ATF4 is a sub-
trome in osteoblasts: characterization, down-regulation strate of RSK2 and an essential regulator of osteoblast
following differentiation, and relationship to gene biology; implication for Coffin-Lowry Syndrome. Cell
expression. J Biol Chem 2014;289:16016–31. 2004;117:387–98.
19. Wu H, Whitfield TW, Gordon JA, Dobson JR, Tai PW, 32. Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N,
van Wijnen AJ, Stein JL, Stein GS, Lian JB. Genomic Wu H, Yu K, Ornitz DM, Olson EN, Justice MJ,
occupancy of Runx2 with global expression profiling Karsenty G. A twist code determines the onset of
identifies a novel dimension to control of osteoblasto- osteoblast differentiation. Dev cell 2004;6:423–35.
genesis. Genome Biol 2014;15:R52. 33. Acampora D, Merlo GR, Paleari L, Zerega B,
20. St John HC, Bishop KA, Meyer MB, Benkusky NA, Leng Postiglione MP, Mantero S, Bober E, Barbieri O,
N, Kendziorski C, Bonewald LF, Pike JW. The osteoblast Simeone A, Levi G. Craniofacial, vestibular and bone
to osteocyte transition: epigenetic changes and response defects in mice lacking the Distal-less-related gene
to the vitamin D3 hormone. Mol Endocrinol Dlx5. Dev 1999;126:3795–809.
2014;28:1150–65. 34. Takayanagi H, Kim S, Matsuo K, Suzuki H, Suzuki T,
21. Ramagopalan SV, Heger A, Berlanga AJ, Maugeri NJ, Sato K, Yokochi T, Oda H, Nakamura K, Ida N,
Lincoln MR, Burrell A, Handunnetthi L, Handel AE, Wagner EF, Taniguchi T. RANKL maintains bone
Disanto G, Orton SM, Watson CT, Morahan JM, homeostasis through c-Fos-dependent induction of
Giovannoni G, Ponting CP, Ebers GC, Knight JC. A interferon-beta. Nature 2002;416:744–9.
ChIP-seq defined genome-wide map of vitamin D 35. Negishi-Koga T, Takayanagi H. Ca2+-NFATc1 signal-
receptor binding: associations with disease and evolu- ing is an essential axis of osteoclast differentiation.
tion. Genome Res 2010;20:1352–60. Immunol Rev 2009;231:241–56.
22. Harada S, Rodan GA. Control of osteoblast function 36. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M,
and regulation of bone mass. Nature 2003;423:349–55. Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J,
23. Komori T. Regulation of osteoblast differentiation by Wagner EF, Mak TW, Kodama T, Taniguchi T.
transcription factors. J Cell Biochem 2006;99:1233–9. Induction and activation of the transcription factor
24. Otto F, Kanegane H, Mundlos S. Mutations in the NFATc1 (NFAT2) integrate RANKL signaling in term-
RUNX2 gene in patients with cleidocranial dysplasia. inal differentiation of osteoclasts. Dev Cell 2002;3:
Hum Mutat 2002;19:209–16. 889–901.
25. Hecht J, Seitz V, Urban M, Wagner F, Robinson PN, 37. Aliprantis AO, Ueki Y, Sulyanto R, Park A, Sigrist KS,
Stiege A, Dieterich C, Kornak U, Wilkening U, Brieske Sharma SM, Ostrowski MC, Olsen BR, Glimcher LH.
N, Zwingman C, Kidess A, Stricker S, Mundlos S. NFATc1 in mice represses osteoprotegerin during
86 K. H. PARK-MIN

osteoclastogenesis and dissociates systemic osteopenia 54. Park-Min KH, Lim E, Lee MJ, Park SH, Giannopoulou
from inflammation in cherubism. J Clin Invest E, Yarilina A, van der Meulen M, Zhao B, Smithers N,
2008;118:3775–89. Witherington J, Lee K, Tak PP, Prinjha RK, Ivashkiv
38. Endo-Munoz L, Cumming A, Rickwood D, Wilson D, LB. Inhibition of osteoclastogenesis and inflammatory
Cueva C, Ng C, Strutton G, Cassady AI, Evdokiou A, bone resorption by targeting BET proteins and epige-
Sommerville S, Dickinson I, Guminski A, Saunders netic regulation. Nat Commun 2014;5:5418.
NA. Loss of osteoclasts contributes to development of 55. Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation
osteosarcoma pulmonary metastases. Cancer Res and deacetylation of non-histone proteins. Gene
2010;70:7063–72. 2005;363:15–23.
39. Asagiri M, Sato K, Usami T, Ochi S, Nishina H, 56. Westendorf JJ. Histone deacetylases in control of ske-
Yoshida H, Morita I, Wagner EF, Mak TW, Serfling letogenesis. J Cell Biochem 2007;102:332–40.
E, Takayanagi H. Autoamplification of NFATc1 57. Jensen ED, Schroeder TM, Bailey J, Gopalakrishnan R,
expression determines its essential role in bone home- Westendorf JJ. Histone deacetylase 7 associates with
ostasis. J Exp Med 2005;202:1261–9. Runx2 and represses its activity during osteoblast
40. Koga T, Matsui Y, Asagiri M, Kodama T, de maturation in a deacetylation-independent manner. J
Crombrugghe B, Nakashima K, Takayanagi H. NFAT Bone Miner Res 2008;23:361–72.
and Osterix cooperatively regulate bone formation. Nat 58. Schroeder TM, Nair AK, Staggs R, Lamblin AF,
Med 2005;11:880–5. Westendorf JJ. Gene profile analysis of osteoblast
41. Grigoriadis AE, Wang ZQ, Cecchini MG, Hofstetter genes differentially regulated by histone deacetylase
W, Felix R, Fleisch HA, Wagner EF. c-Fos: a key inhibitors. BMC Genomics 2007;8:362.
regulator of osteoclast-macrophage lineage determina- 59. Iwami K, Moriyama T. Effects of short chain fatty acid,
tion and bone remodeling. Science 1994;266:443–8. sodium butyrate, on osteoblastic cells and osteoclastic
42. Wang ZQ, Ovitt C, Grigoriadis AE, Mohle-Steinlein U, cells. Int J Biochem 1993;25:1631–5.
Ruther U, Wagner EF. Bone and haematopoietic 60. Schroeder TM, Westendorf JJ. Histone deacetylase
defects in mice lacking c-fos. Nature 1992;360:741–5. inhibitors promote osteoblast maturation. J Bone
43. Richmond TJ, Davey CA. The structure of DNA in the Miner Res2005;20:2254–63.
nucleosome core. Nature 2003;423:145–50. 61. Di Bernardo G, Squillaro T, Dell’Aversana C, Miceli M,
44. Felsenfeld G, Groudine M. Controlling the double Cipollaro M, Cascino A, Altucci L, Galderisi U.
helix. Nature 2003;421:448–53. Histone deacetylase inhibitors promote apoptosis and
45. Kouzarides T. Chromatin modifications and their senescence in human mesenchymal stem cells. Stem
function. Cell 2007;128:693–705. Cells Dev 2009;18:573–81.
46. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang 62. Lee HW, Suh JH, Kim AY, Lee YS, Park SY, Kim JB.
Z, et al. High-resolution profiling of histone methylations Histone deacetylase 1-mediated histone modification
in the human genome. Cell 2007;129:823–37. regulates osteoblast differentiation. Mol Endocrinol
47. Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, 2006;20:2432–43.
Cuddapah S, Cui K, Roh TY, Peng W, Zhang MQ, 63. Schroeder TM, Kahler RA, Li X, Westendorf JJ.
Zhao K. Combinatorial patterns of histone acetylations Histone deacetylase 3 interacts with runx2 to repress
and methylations in the human genome. Nat Genet the osteocalcin promoter and regulate osteoblast differ-
2008;40:897–903. entiation. J Biol Chem 2004;279:41998–2007.
48. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward 64. Dudakovic A, Evans JM, Li Y, Middha S, McGee-
LD, Epstein CB, Zhang X, Wang L, Issner R, Coyne M, Lawrence ME, van Wijnen AJ, Westendorf JJ. Histone
Ku M, Durham T, Kellis M, Bernstein BE. Mapping deacetylase inhibition promotes osteoblast maturation
and analysis of chromatin state dynamics in nine by altering the histone H4 epigenome and reduces Akt
human cell types. Nature 2011;473:43–9. phosphorylation. J Biol Chem 2013;288:28783–91.
49. Zhou VW, Goren A, Bernstein BE. Charting histone 65. McGee-Lawrence ME, Carpio LR, Schulze RJ, Pierce JL,
modifications and the functional organization of mam- McNiven MA, Farr JN, Khosla S, Oursler MJ, Westendorf
malian genomes. Nat Rev Genet 2011;12:7–18. JJ. Hdac3 Deficiency Increases Marrow Adiposity and
50. Lee KK, Workman JL. Histone acetyltransferase com- Induces Lipid Storage and Glucocorticoid Metabolism
plexes: one size doesn’t fit all. Nat Rev Mol Cell Biol in Osteochondroprogenitor Cells. J Bone Miner Res
2007;8:284–95. 2015;21:993–1002.
51. Falkenberg KJ, Johnstone RW. Histone deacetylases 66. Backesjo CM, Li Y, Lindgren U, Haldosen LA.
and their inhibitors in cancer, neurological diseases Activation of Sirt1 decreases adipocyte formation dur-
and immune disorders. Nat Rev Drug Discovery ing osteoblast differentiation of mesenchymal stem
2014;13:673–91. cells. J Bone Miner Res 2006;21:993–1002.
52. Yun M, Wu J, Workman JL, Li B. Readers of histone 67. Rahman MM, Kukita A, Kukita T, Shobuike T,
modifications. Cell Res 2011;21:564–78. Nakamura T, Kohashi O. Two histone deacetylase
53. Kim S, Shevde NK, Pike JW. 1,25-Dihydroxyvitamin inhibitors, trichostatin A and sodium butyrate, sup-
D3 stimulates cyclic vitamin D receptor/retinoid X press differentiation into osteoclasts but not into
receptor DNA-binding, co-activator recruitment, and macrophages. Blood 2003;101:3451–9.
histone acetylation in intact osteoblasts. J Bone Miner 68. Pham L, Kaiser B, Romsa A, Schwarz T,
Res 2005;20:305–17. Gopalakrishnan R, Jensen ED, Mansky KC. HDAC3
EPIGENETIC REGULATION OF BONE CELLS 87

and HDAC7 have opposite effects on osteoclast differ- JE, Heymann D, Ory B. Selective inhibition of BET
entiation. J Biol Chem 2011;286:12056–65. bromodomain epigenetic signalling interferes with the
69. Stemig M, Astelford K, Emery A, Cho JJ, Allen B, bone-associated tumour vicious cycle. Nat Commun
Huang TH, Gopalakrishnan R, Mansky KC, Jensen 2014;5:3511.
ED. Deletion of histone deacetylase 7 in osteoclasts 81. Greer EL, Shi Y. Histone methylation: a dynamic mark
decreases bone mass in mice by interactions with in health, disease and inheritance. Nat Rev Genet
MITF. PloS One 2015;10:e0123843. 2012;13:343–57.
70. Triantafyllou N, Lambrinoudaki I, Armeni E, 82. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR,
Evangelopoulos EM, Boufidou F, Antoniou A, Cole PA, Casero RA, Shi Y. Histone demethylation
Tsivgoulis G. Effect of long-term valproate monother- mediated by the nuclear amine oxidase homolog
apy on bone mineral density in adults with epilepsy. J LSD1. Cell 2004;119:941–53.
Neurol Sci 2010;290:131–4. 83. Tsukada Y, Fang J, Erdjument-Bromage H, Warren
71. Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK, ME, Borchers CH, Tempst P, Zhang Y. Histone
Zhou MM. Structure and ligand of a histone acetyl- demethylation by a family of JmjC domain-containing
transferase bromodomain. Nature 1999;399:491–6. proteins. Nature 2006;439:811–6.
72. Zeng L, Zhang Q, Li S, Plotnikov AN, Walsh MJ, Zhou 84. De Santa F, Totaro MG, Prosperini E, Notarbartolo S,
MM. Mechanism and regulation of acetylated histone Testa G, Natoli G. The histone H3 lysine-27 demethy-
binding by the tandem PHD finger of DPF3b. Nature lase Jmjd3 links inflammation to inhibition of poly-
2010;466:258–62. comb-mediated gene silencing. Cell 2007;130:1083–94.
73. Belkina AC, Denis GV. BET domain co-regulators in 85. Yasui T, Hirose J, Tsutsumi S, Nakamura K, Aburatani
obesity, inflammation and cancer. Nat Rev Cancer H, Tanaka S. Epigenetic regulation of osteoclast differ-
2012;12:465–77. entiation: possible involvement of Jmjd3 in the histone
74. Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell demethylation of Nfatc1. J Bone Miner Res
S, Chung CW, Chandwani R, Marazzi I, Wilson P, 2011;26:2665–71.
Coste H, White J, Kirilovsky J, Rice CM, Lora JM, 86. Yang D, Okamura H, Nakashima Y, Haneji T. Histone
Prinjha RK, Lee K, Tarakhovsky A. Suppression of demethylase Jmjd3 regulates osteoblast differentiation
inflammation by a synthetic histone mimic. Nature via transcription factors Runx2 and osterix. J Biol
2010;468:1119–23. Chem 2013;288:33530–41.
75. Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, 87. Zhang F, Xu L, Xu L, Xu Q, Karsenty G, Chen CD.
Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf Histone demethylase JMJD3 is required for osteoblast
C, Savitski MM, Huthmacher C, Gudgin E, Lugo D, differentiation in mice. Sci Rep 2015;5:13418.
Beinke S, Chapman TD, Roberts EJ, Soden PE, Auger 88. Simon JA, Kingston RE. Mechanisms of polycomb
KR, Mirguet O, Doehner K, Delwel R, Burnett AK, gene silencing: knowns and unknowns. Nat Rev Mol
Jeffrey P, Drewes G, Lee K, Huntly BJ, Kouzarides T. Cell Biol 2009;10:697–708.
Inhibition of BET recruitment to chromatin as an 89. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage
effective treatment for MLL-fusion leukaemia. Nature H, Tempst P, Jones RS, Zhang Y. Role of histone H3
2010;478:529–33. lysine 27 methylation in Polycomb-group silencing.
76. Prinjha RK, Witherington J, Lee K. Place your BETs: Science 2002;298:1039–43.
the therapeutic potential of bromodomains. Trends 90. Kalb R, Latwiel S, Baymaz HI, Jansen PW, Muller CW,
Pharmacol Sci 2012;33:146–53. Vermeulen M, Muller J. Histone H2A monoubiquiti-
77. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, nation promotes histone H3 methylation in Polycomb
Fedorov O, Morse EM, Keates T, Hickman TT, Felletar repression. Nat Struct Mol Biol 2014;21:569–71.
I, Philpott M, Munro S, McKeown MR, Wang Y, 91. Vire E, Brenner C, Deplus R, Blanchon L, Fraga M,
Christie AL, West N, Cameron MJ, Schwartz B, Didelot C, Morey L, Van Eynde A, Bernard D,
Heightman TD, La Thangue N, French CA, Wiest O, Vanderwinden JM, Bollen M, Esteller M, Di Croce L,
Kung AL, Knapp S, Bradner JE. Selective inhibition of de Launoit Y, Fuks F. The Polycomb group protein
BET bromodomains. Nature 2010;468:1067–73. EZH2 directly controls DNA methylation. Nature
78. Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, 2006;439:871–4.
Sison EA, Magoon D, Qi J, Blatt K, Wunderlich M, 92. Wei Y, Chen YH, Li LY, Lang J, Yeh SP, Shi B, Yang
Taylor MJ, Johns C, Chicas A, Mulloy JC, Kogan SC, CC, Yang JY, Lin CY, Lai CC, Hung MC. CDK1-
Brown P, Valent P, Bradner JE, Lowe SW, Vakoc CR. dependent phosphorylation of EZH2 suppresses
RNAi screen identifies Brd4 as a therapeutic target in methylation of H3K27 and promotes osteogenic differ-
acute myeloid leukaemia. Nature 2011;478:524–8. entiation of human mesenchymal stem cells. Nat Cell
79. Asangani IA, Dommeti VL, Wang X, Malik R, Cieslik Biol 2011;13:87–94.
M, Yang R, Escara-Wilke J, Wilder-Romans K, 93. Gori F, Divieti P, Demay MB. Cloning and character-
Dhanireddy S, Engelke C, Iyer MK, Jing X, Wu YM, ization of a novel WD-40 repeat protein that dramati-
Cao X, Qin ZS, Wang S, Feng FY, Chinnaiyan AM. cally accelerates osteoblastic differentiation. J Biol
Therapeutic targeting of BET bromodomain proteins Chem 2001;276:46515–22.
in castration-resistant prostate cancer. Nature 94. Gori F, Friedman LG, Demay MB. Wdr5, a WD-40
2014;510:278–82. protein, regulates osteoblast differentiation during
80. Lamoureux F, Baud’huin M, Rodriguez Calleja L, embryonic bone development. Dev Biol
Jacques C, Berreur M, Redini F, Lecanda F, Bradner 2006;295:498–506.
88 K. H. PARK-MIN

95. Zhu ED, Demay MB, Gori F. Wdr5 is essential for distinguishes two distinct classes of promoters. Proc
osteoblast differentiation. J Biol Chem 2008;283:7361–7. Natl Acad Sci USA 2006;103:1412–7.
96. Thurman RE, Rynes E, Humbert R, Vierstra J, 108. Suzuki MM, Bird A. DNA methylation landscapes:
Maurano MT, et al. The accessible chromatin land- provocative insights from epigenomics. Nat Rev
scape of the human genome. Nature 2012;489:75–82. Genet 2008;9:465–76.
97. Song L, Zhang Z, Grasfeder LL, Boyle AP, Giresi PG, 109. Krueger F, Kreck B, Franke A, Andrews SR. DNA
Lee BK, Sheffield NC, Graf S, Huss M, Keefe D, Liu Z, methylome analysis using short bisulfite sequencing
London D, McDaniell RM, Shibata Y, Showers KA, data. Nat Methods 2012;9:145–51.
Simon JM, Vales T, Wang T, Winter D, Zhang Z, 110. Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong
Clarke ND, Birney E, Iyer VR, Crawford GE, Lieb JD, CT, Low HM, Kin Sung KW, Rigoutsos I, Loring J,
Furey TS. Open chromatin defined by DNaseI and Wei CL. Dynamic changes in the human methylome
FAIRE identifies regulatory elements that shape cell- during differentiation. Genome Res 2010;20:320–31.
type identity. Genome Res 2011;21:1757–67. 111. Villagra A, Gutierrez J, Paredes R, Sierra J, Puchi M,
98. Song L, Crawford GE. DNase-seq: a high-resolution Imschenetzky M, Wijnen Av A, Lian J, Stein G, Stein J,
technique for mapping active gene regulatory elements Montecino M. Reduced CpG methylation is associated
across the genome from mammalian cells. Cold Spring with transcriptional activation of the bone-specific rat
Harbor protocols 2010;2010:pdb prot5384. osteocalcin gene in osteoblasts. J Cell Biochem
99. Giresi PG, Kim J, McDaniell RM, Iyer VR, Lieb JD. 2002;85:112–22.
FAIRE (Formaldehyde-Assisted Isolation of Regulatory 112. Farshdousti Hagh M, Noruzinia M, Mortazavi Y,
Elements) isolates active regulatory elements from Soleimani M, Kaviani S, Abroun S, Dehghani Fard A,
human chromatin. Genome Res 2007;17:877–85. Mahmoodinia M. Different Methylation Patterns of
100. Auerbach RK, Euskirchen G, Rozowsky J, Lamarre- RUNX2, OSX, DLX5 and BSP in Osteoblastic
Vincent N, Moqtaderi Z, Lefrancois P, Struhl K, Differentiation of Mesenchymal Stem Cells. Cell J
Gerstein M, Snyder M. Mapping accessible chromatin 2015;17:71–82.
regions using Sono-Seq. Proc Natl Acad Sci USA 113. Guo X, Wang L, Li J, Ding Z, Xiao J, Yin X, He S, Shi P,
2009;106:14926–31. Dong L, Li G, Tian C, Wang J, Cong Y, Xu Y. Structural
101. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, insight into autoinhibition and histone H3-induced acti-
Greenleaf WJ. Transposition of native chromatin for vation of DNMT3A. Nature 2015;517:640–4.
fast and sensitive epigenomic profiling of open chro- 114. Li E, Bestor TH, Jaenisch R. Targeted mutation of the
matin, DNA-binding proteins and nucleosome posi- DNA methyltransferase gene results in embryonic leth-
tion. Nat Methods 2013;10:1213–8. ality. Cell 1992;69:915–26.
102. Neph S, Vierstra J, Stergachis AB, Reynolds AP, 115. Okano M, Bell DW, Haber DA, Li E. DNA methyl-
Haugen E, Vernot B, Thurman RE, John S, transferases Dnmt3a and Dnmt3b are essential for de
Sandstrom R, Johnson AK, Maurano MT, Humbert novo methylation and mammalian development. Cell
R, Rynes E, Wang H, Vong S, Lee K, Bates D, Diegel 1999;99:247–57.
M, Roach V, Dunn D, Neri J, Schafer A, Hansen RS, 116. Tsai CC, Su PF, Huang YF, Yew TL, Hung SC. Oct4
Kutyavin T, Giste E, Weaver M, Canfield T, Sabo P, and Nanog directly regulate Dnmt1 to maintain self-
Zhang M, Balasundaram G, Byron R, MacCoss MJ, renewal and undifferentiated state in mesenchymal
Akey JM, Bender MA, Groudine M, Kaul R, stem cells. Mol Cell 2012;47:169–82.
Stamatoyannopoulos JA. An expansive human regula- 117. Vaes BL, Lute C, van der Woning SP, Piek E, Vermeer
tory lexicon encoded in transcription factor footprints. J, Blom HJ, Mathers JC, Muller M, de Groot LC,
Nature 2012;489:83–90. Steegenga WT. Inhibition of methylation decreases
103. Boyle AP, Song L, Lee BK, London D, Keefe D, Birney E, osteoblast differentiation via a non-DNA-dependent
Iyer VR, Crawford GE, Furey TS. High-resolution gen- methylation mechanism. Bone 2010;46:514–23.
ome-wide in vivo footprinting of diverse transcription 118. Liu L, van Groen T, Kadish I, Li Y, Wang D, James SR,
factors in human cells. Genome Res 2011;21:456–64. Karpf AR, Tollefsbol TO. Insufficient DNA methyla-
104. Barutcu AR, Tai PW, Wu H, Gordon JA, Whitfield tion affects healthy aging and promotes age-related
TW, Dobson JR, Imbalzano AN, Lian JB, van Wijnen health problems. Clin Epigenet 2011;2:349–60.
AJ, Stein JL, Stein GS. The bone-specific Runx2-P1 119. Delgado-Calle J, Fernandez AF, Sainz J, Zarrabeitia
promoter displays conserved three-dimensional chro- MT, Sanudo C, Garcia-Renedo R, Perez-Nunez MI,
matin structure with the syntenic Supt3h promoter. Garcia-Ibarbia C, Fraga MF, Riancho JA. Genome-
Nucleic Acids Res 2014;42:10360–72. wide profiling of bone reveals differentially methylated
105. Zhou X, Lowdon RF, Li D, Lawson HA, Madden PA, regions in osteoporosis and osteoarthritis. Arthritis and
Costello JF, Wang T. Exploring long-range genome rheumatism 2013;65:197–205.
interactions using the WashU Epigenome Browser. 120. Pastor WA, Aravind L, Rao A. TETonic shift: biologi-
Nat Methods 2013;10:375–6. cal roles of TET proteins in DNA demethylation and
106. Inoue K, Imai Y. Identification of novel transcription transcription. Nat RevMol Cell Biol 2013;14:341–56.
factors in osteoclast differentiation using genome-wide 121. Yan X, Ehnert S, Culmes M, Bachmann A, Seeliger
analysis of open chromatin determined by DNase-seq. C, Schyschka L, Wang Z, Rahmanian-Schwarz A,
J Bone Miner Res 2014;29:1823–32. Stockle U, De Sousa PA, Pelisek J, Nussler AK. 5-
107. Saxonov S, Berg P, Brutlag DL. A genome-wide analy- azacytidine improves the osteogenic differentiation
sis of CpG dinucleotides in the human genome potential of aged human adipose-derived
EPIGENETIC REGULATION OF BONE CELLS 89

mesenchymal stem cells by DNA demethylation. PloS 139. Kapinas K, Delany AM. MicroRNA biogenesis and
One 2014;9:e90846. regulation of bone remodeling. Arthritis Res Ther
122. de la Rica L, Rodriguez-Ubreva J, Garcia M, Islam AB, 2011;13:220.
Urquiza JM, Hernando H, Christensen J, Helin K, 140. Harfe BD, McManus MT, Mansfield JH, Hornstein E,
Gomez-Vaquero C, Ballestar E. PU.1 target genes Tabin CJ. The RNaseIII enzyme Dicer is required for
undergo Tet2-coupled demethylation and DNMT3b- morphogenesis but not patterning of the vertebrate
mediated methylation in monocyte-to-osteoclast differ- limb. Proc Nat Acad Sci USA 2005;102:10898–903.
entiation. Genome Biol 2013;14:R99. 141. Gaur T, Hussain S, Mudhasani R, Parulkar I, Colby JL,
123. Lander ES, Linton LM, Birren B, Nusbaum C, Zody Frederick D, Kream BE, van Wijnen AJ, Stein JL, Stein
MC, et al. Initial sequencing and analysis of the human GS, Jones SN, Lian JB. Dicer inactivation in osteopro-
genome. Nature 2001;409:860–921. genitor cells compromises fetal survival and bone forma-
124. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, tion, while excision in differentiated osteoblasts increases
et al. The sequence of the human genome. Science bone mass in the adult mouse. Dev Biol 2010;340:10–21.
2001;291:1304–51. 142. Mizoguchi F, Izu Y, Hayata T, Hemmi H, Nakashima K,
125. Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Nakamura T, Kato S, Miyasaka N, Ezura Y, Noda M.
Regev A, Rinn JL. Integrative annotation of human large Osteoclast-specific Dicer gene deficiency suppresses osteo-
intergenic noncoding RNAs reveals global properties clastic bone resorption. J Cell Biochem 2010;109:866–75.
and specific subclasses. Genes Dev 2011;25:1915–27. 143. Sugatani T, Hruska KA. Impaired micro-RNA path-
126. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, ways diminish osteoclast differentiation and function. J
Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, Biol Chem 2009;284:4667–78.
Lagarde J, Veeravalli L, Ruan X, Ruan Y, Lassmann T, 144. Sugatani T, Vacher J, Hruska KA. A microRNA
Carninci P, Brown JB, Lipovich L, Gonzalez JM, Thomas expression signature of osteoclastogenesis. Blood
M, Davis CA, Shiekhattar R, Gingeras TR, Hubbard TJ, 2011;117:3648–57.
Notredame C, Harrow J, Guigo R. The GENCODE v7 145. Hassan MQ, Gordon JA, Beloti MM, Croce CM, van
catalog of human long noncoding RNAs: analysis of their Wijnen AJ, Stein JL, Stein GS, Lian JB. A network
gene structure, evolution, and expression. Genome Res connecting Runx2, SATB2, and the miR-23a~27a~24-2
2012;22:1775–89. cluster regulates the osteoblast differentiation program.
127. Quinn JJ, Chang HY. Unique features of long non- Proc Natl Acad Sci USA 2010;107:19879–84.
coding RNA biogenesis and function. Nat Rev Genet 146. Zeng Y, Qu X, Li H, Huang S, Wang S, Xu Q, Lin R,
2016;17:47–62. Han Q, Li J, Zhao RC. MicroRNA-100 regulates osteo-
128. Huarte M. The emerging role of lncRNAs in cancer. genic differentiation of human adipose-derived
Nat Med 2015;21:1253–61. mesenchymal stem cells by targeting BMPR2. FEBS
129. Tong X, Gu PC, Xu SZ, Lin XJ. Long non-coding Lett 2012;586:2375–81.
RNA-DANCR in human circulating monocytes: a 147. Zhang Y, Xie RL, Croce CM, Stein JL, Lian JB, van
potential biomarker associated with postmenopausal Wijnen AJ, Stein GS. A program of microRNAs con-
osteoporosis. Bioscience, biotechnology, and biochem- trols osteogenic lineage progression by targeting tran-
istry 2015;79:732–7. scription factor Runx2. Proc Natl Acad Sci USA
130. Zhu L, Xu PC. Downregulated LncRNA-ANCR pro- 2011;108:9863–8.
motes osteoblast differentiation by targeting EZH2 and 148. Kanis JA. Diagnosis of osteoporosis and assessment of
regulating Runx2 expression. Biochem Biophys Res fracture risk. Lancet 2002;359:1929–36.
Commun 2013;432:612–7. 149. Kanis JA, Johnell O, Oden A, De Laet C, Jonsson B,
131. Hassan MQ, Tye CE, Stein GS, Lian JB. Non-coding Dawson A. Ten-year risk of osteoporotic fracture and
RNAs: Epigenetic regulators of bone development and the effect of risk factors on screening strategies. Bone
homeostasis. Bone 2015;81:746–56. 2002;30:251–8.
132. Gamez B, Rodriguez-Carballo E, Ventura F. 150. Berry SD, Samelson EJ, Pencina MJ, McLean RR, Cupples
MicroRNAs and post-transcriptional regulation of ske- LA, Broe KE, Kiel DP. Repeat bone mineral density
letal development. J Mol Endocrinol 2014;52:R179–97. screening and prediction of hip and major osteoporotic
133. Lian JB, Stein GS, van Wijnen AJ, Stein JL, Hassan MQ, fracture. JAMA 2013;310:1256–62.
Gaur T, Zhang Y. MicroRNA control of bone formation 151. Seeliger C, Karpinski K, Haug AT, Vester H, Schmitt A,
and homeostasis. Nat Rev Endocrinol 2012;8:212–27. Bauer JS, van Griensven M. Five freely circulating miRNAs
134. Bartel DP. MicroRNAs: genomics, biogenesis, mechan- and bone tissue miRNAs are associated with osteoporotic
ism, and function. Cell 2004;116:281–97. fractures. J Bone Miner Res 2014;29:1718–28.
135. Plasterk RH. Micro RNAs in animal development. Cell 152. Bouchie A. First microRNA mimic enters clinic. Nat
2006;124:877–81. Biotech 2013;31:577.
136. Krol J, Loedige I, Filipowicz W. The widespread reg- 153. Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG,
ulation of microRNA biogenesis, function and decay. Simmons PJ, Wang CY. The meaning, the sense and the
Nat Rev Genet 2010;11:597–610. significance: translating the science of mesenchymal stem
137. Hobert O. Gene regulation by transcription factors and cells into medicine. Nat Med 2013;19:35–42.
microRNAs. Science 2008;319:1785–6. 154. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales
138. Starega-Roslan J, Koscianska E, Kozlowski P, Krzyzosiak ML, Snyder MP, Chang HY, Greenleaf WJ. Single-cell
WJ. The role of the precursor structure in the biogenesis chromatin accessibility reveals principles of regulatory var-
of microRNA. Cell Mol Life Sci 2011;68:2859–71. iation. Nature 2015;523:486–90.

You might also like