You are on page 1of 4

Curr Osteoporos Rep (2013) 11:126–129

DOI 10.1007/s11914-013-0145-4

SKELETAL REGULATIONS (D GADDY, SECTION EDITOR)

Notch Signaling and Bone Remodeling


Jenna Regan & Fanxin Long

Published online: 22 March 2013


# Springer Science+Business Media New York 2013

Abstract Notch signaling plays context-dependent roles in Notch signaling in regulating bone physiology and discuss
the development and maintenance of many cell types and examples of skeletal diseases caused by genetic mutations in
tissues in mammals. In the skeleton, both osteoblasts and Notch pathway components.
osteoclasts require Notch signaling for proper differentiation
and function, and the specific roles of Notch are dependent
on the differentiation status of the cell. The recent discovery The Notch Signaling Pathway
of activating NOTCH2 mutations as the cause of Hajdu-
Cheney syndrome has highlighted the significance of Notch Notch signaling requires cell-cell contact, and is initiated
signaling in human bone physiology. when the single-pass transmembrane ligands (Jagged [JAG]
1 and 2 and Delta-like [DLL] 1, 3, and 4 in mammals) engage
Keywords Notch signaling . Bone remodeling . Notch (1–4 in mammals) receptors expressed on the surface of
Osteoblasts . Osteoclasts . Skeletal development . Skeletal a neighboring cell [1]. Ligand binding leads to proteolytic
diseases . Hajdu-Cheney syndrome . Alagille syndrome cleavage of the Notch receptor by the γ-secretase complex,
releasing the Notch intracellular domain (NICD). Within the
cell, the NICD translocates to the nucleus where it interacts with
Introduction the DNA-binding protein RBPjκ/CBF1, displacing co-
repressors and leading to the assembly of an activator complex
Following completion of skeletal development (also referred that also includes Mastermind-like (MAML) and transcription-
to as the modeling phase) the mammalian bones undergo a al co-activators. Notch target genes are thus activated, including
life-long process of remodeling that is mediated by the the transcriptional repressors hairy and enhancer of split (HES)
reciprocal actions of osteoblasts and osteoclasts. Both the and HES-related with YRPW motif (HEY) [2–4].
matrix-depositing activity of osteoblasts and the matrix- Notch signaling may interact with other signaling path-
resorbing activity of osteoclasts must be tightly regulated ways such as Wnt and bone morphogenetic protein (BMP)
in order to maintain the homeostasis of bone mass and to to regulate skeletal development and homeostasis. It should
prevent disease states. As one might expect, there are a be noted, however, that most studies to date have relied on
multitude of signaling pathways that control the differenti- overexpression of NICD from Notch1, which may not faith-
ation, function, and cross-talk of osteoblasts and osteoclasts. fully represent physiological Notch signaling. In cultured
This review will highlight recent insights into the role of osteoblast precursors, NICD overexpression opposed the
osteoblast-inducing activity of exogenous Wnt, an effect
mediated by Hes1 [5]. In vitro studies examining the rela-
J. Regan : F. Long
tionship between Notch and BMP signaling have provided
Department of Medicine, Washington University School of conflicting results. In one study, NICD overexpression
Medicine, St. Louis, MO 63110, USA blocked the differentiation of osteoblast precursors in re-
sponse to BMP2 [6•] but others showed that Notch signaling
F. Long (*)
enhanced [7], whereas inhibition of Notch impaired BMP2-
Department of Developmental Biology, Washington University
School of Medicine, St. Louis, MO 63110, USA induced osteoblast differentiation [8]. Future studies are
e-mail: flong@wustl.edu necessary to delineate the relationship between Notch and
Curr Osteoporos Rep (2013) 11:126–129 127

the other signaling pathways under physiological or patho- upon deletion of RBPjk [16•]. At the cellular level, NICD
physiological conditions. Nonetheless, it is expected that overexpression stimulated proliferation of early-stage
Notch signaling does not act in isolation, as cells almost osteoblast-lineage cells but prevented progression to fully
certainly receive and respond to multiple signals simulta- mature osteoblasts [15]. In contrast, when overexpressed
neously in vivo, resulting in an integrated phenotypic from the 3.6 kb Col1a1 promoter active in the osteoblast
response. precursors, NICD caused osteopenia [17]. Likewise, when
NICD was expressed from the R26-NICD allele [18] with
different Cre drivers targeting various stages of the oste-
Notch Signaling in the Osteoblast Lineage oblast lineage, varying bone phenotypes were observed
[19•]. Taken together, these studies support the notion that
Early mouse genetics studies suggested an important role for NICD overexpression suppresses differentiation from
Notch signaling in skeletal development. Axial skeletal de- early-stage precursors, promotes proliferation of interme-
fects were observed in global deletions of presenilin-1, a diate osteoblast-lineage cells, but impedes formation of
component of the γ-secretase complex [9, 10], and Notch mature osteoblasts.
ligand Dll3 [11]. However, elucidating the role of Notch Notch signaling in osteoblast-lineage cells also indirectly
signaling specifically in osteoblasts required tissue-specific regulates osteoclastogenesis by modulating the expression of
gene deletions. osteoclastogenic factors by those cells. In particular, removal
Genetic disruption of physiological Notch signaling has of Notch signaling in the osteoblast lineage notably reduced
revealed a critical role for this pathway in the regulation of the expression of Opg (also known as Tnfrsf11b), a known
osteoblastogenesis. Deletion of Notch1 and Notch2 in the anti-osteoclastogenic factor [12•, 15]. Conversely,
skeletogenic mesenchyme with Prx1-cre caused a massive overexpression of NICD in osteocytes suppressed bone re-
accumulation of bone within the marrow cavity by eight sorption [20]. Thus, Notch signaling in osteoblast-lineage
weeks of age [12•]. However, when mutant mice were cells regulates bone remodeling both cell-autonomously and
examined at 26 weeks, the trabecular bone mass was se- indirectly through regulation of the osteoclast.
verely reduced to only 10 % of control animals [12•]. Notch
signaling was found to play a crucial role in suppressing
differentiation of mesenchymal progenitors such that an Notch Signaling in the Osteoclast Lineage
early bolus of osteoblast differentiation in the Notch-
deficient mice produced the initially high bone mass but Notch signaling also directly regulates osteoclastogenesis.
also depleted the progenitor pool, preventing later renewal Osteoclasts differentiate from macrophage precursors in
of the osteoblast population [12•]. Subsequent studies re- response to the secreted factors RANKL and macrophage
vealed that Notch2, instead of Notch1, plays a predominant colony-stimulating factor (M-CSF), and the activation of
role in suppressing osteoblastogenesis, and that the regula- cell surface receptors DAP12 and FcRγ [21]. Deletion of
tion is largely mediated through RBPjk [13•]. Downstream Notch receptors in macrophages with Cre expressed from a
of RBPjK, the Hey family of transcriptional suppressors is viral vector enhanced osteoclast differentiation in response
an important mediator that both inhibits Runx2 activity and to M-CSF and RANKL in vitro [22•]. Conversely, bone
suppresses Nfatc1 transcription [12•]. The suppressive role marrow macrophages cultured with an immobilized Notch
of Hey proteins in osteoblastogenesis is also supported by ligand downregulated the receptor for M-CSF and exhibited
the finding that Hey1-overexpressing mice are osteopenic suppression of osteoclast differentiation [23]. However,
by 12 weeks of age [14]. Interestingly, disruption of RBPjK others reported that Notch2 and Hes1 were upregulated
with Osx-Cre or 2.3Col1-Cre (driven by the 2.3 kb promoter during RANKL-induced osteoclast differentiation in a mac-
of Col1a1) did not cause an obvious bone phenotype. Thus, rophage cell line, and that treatment with a γ-secretase
the suppressive role of Notch-RBPjk signaling in inhibitor blocked osteoclastogenesis in these cells [24].
osteoblastogenesis is limited to the early stages of osteoblast The seemingly contradictory results may reflect different
differentiation. requirements for Notch signaling at distinct stages of
Similarly, mouse transgenic studies employing osteoclast differentiation. Furthermore, the stage-specific
overexpression of NICD have revealed different responses regulation may involve different Notch receptors and
depending on the differentiation stage of osteoblast-lineage ligands, as it was reported that Jag1 and Notch1 blocked
cells. Overexpression of NICD under control of the 2.3 kb but Dll1 and Notch2 promoted osteoclastogenesis [25].
Col1a1 promoter active in committed osteoblastic cells Thus, the effect of Notch signaling on osteoclastogenesis
caused a high bone mass phenotype, characterized by ex- appears to depend both on the differentiation status of
cessive immature woven bone and fibrotic cells in the mar- the cell and the expression of particular ligands and
row cavity [15, 16•]. The effect of NICD was abolished receptors.
128 Curr Osteoporos Rep (2013) 11:126–129

Notch Signaling and Human Bone Diseases vitamin and mineral absorption caused by the cholestasis
[33, 41], recent evidence has implicated JAG1 in regulating
Hajdu-Cheney Syndrome bone mineral density. A genome-wide association study iden-
tified a single nucleotide polymorphism in the JAG1 gene
A series of recent studies identified mutations in Notch2 as correlated with higher BMD across multiple ethnic groups,
the underlying cause of Hajdu-Cheney Syndrome (HCS, and further analyses demonstrated a higher level of JAG1
OMIM# 102500), a rare disorder with an autosomal domi- transcript in individuals with the “high BMD” genotype
nant pattern of inheritance. The most prominent clinical [42•]. Given that ALGS1 results from haploinsufficiency of
features of HCS are craniofacial dysmorphology (including JAG1, compromised Notch signaling may contribute to skel-
hypertelorism, thick eyebrows, low-set ears, malar hypopla- etal fragility in young ALGS patients.
sia, prognathism, micrognathia, long philtrum, and abnor-
mal dentition) and radiographic abnormalities, particularly
Conclusions
acro-osteolysis, osteoporosis, and Wormian bones. Some
other features include short stature, hearing defects, cardio-
Mouse genetic studies in recent years have uncovered impor-
vascular abnormalities, and polycystic kidneys [26, 27].
tant roles for Notch signaling in both the osteoblast and oste-
Remarkably, all of the HCS-causing mutations identified
oclast lineages. A common feature for Notch regulation in both
to date occur within the final exon of the Notch2 gene [27,
cell lineages has emerged that the Notch effect is highly
28, 29•]. The mutated gene is predicted to encode a truncat-
dependent on the cell context represented by various differen-
ed Notch2 protein that contains all of the domains essential
tiation stages. Studies to date employing overexpression of
for receptor function, cleavage, and nuclear localization but
NICD have provided important clues to the potential effect of
lacks the domain containing conserved proline-glutamate-
hyperactive Notch signaling in pathophysiological conditions.
serine-threonine-rich motifs (PEST domain), which harbors
However, a mouse model recapitulating the Notch2 mutations
the degradation signal and controls the half-life of the pro-
causing HCS and SFPKS in humans is needed to elucidate the
tein [1]. Thus, HCS forms of Notch2 are expected to evade
cellular basis for the disease. The fact that physiological Notch
normal proteolytic degradation and consequently have in-
signaling suppresses osteoblast differentiation in the early pro-
creased signaling activity [30]. In addition, similar truncat-
genitors provides the hope that inhibition of the pathway may
ing mutations in Notch2 have been identified in serpentine
be explored as a means to stimulate bone formation. However,
fibula-polycystic kidney syndrome (SFPKS) [31, 32]. Thus,
given that Notch signaling affects both osteoblast and osteo-
HCS and SFPKS represent variable phenotypes of the same
clast lineages and that the effects are dependent on the cell
disease caused by gain-of-function mutations in Notch2.
context, it may be critical to develop therapeutic options that
can preferentially target the desired cell types.
Alagille Syndrome
Acknowledgements Work in the Long lab is supported by NIH grant
Alagille syndrome is an autosomal dominant disorder AR055923. J. Regan is a postdoctoral fellow supported by NIH T32
that occurs in at least 1 in 70,000 live births [33]. HL007873.
Haploinsufficiency in the JAG1 gene accounts for approxi-
Disclosure J. Regan declares no conflicts of interest. F. Long declares
mately 97 % of Alagille syndrome cases [34, 35], designated no conflicts of interest.
ALGS1 (OMIM# 118450). ALGS2 (OMIM# 610205) is
caused by mutations in NOTCH2 [36•] and accounts for
<1 % of total cases. Unlike HCS, the mutations causing
ALGS2 lead to Notch2 proteins that are predicted to exhibit References
compromised activity. The most common skeletal malforma-
tion found in ALGS patients is known as butterfly vertebrae, Papers of particular interest, published recently, have been
which is caused by incomplete fusion of the anterior arch [33, highlighted as:
37]. Other reported skeletal defects include hand and pelvic • Of importance
anomalies, shortened ulna, and absence of the 12th rib [38].
Characteristic facial dysmorphology can include a prominent
forehead, deep-set eyes with moderate hypertelorism, pointed 1. Kopan R, Ilagan MX. The canonical Notch signaling pathway:
chin, and straight nose with bulbous tip [39]. Although none unfolding the activation mechanism. Cell. 2009;137:216–33.
of the common skeletal malformations are functionally 2. Fortini ME. Notch signaling: the core pathway and its posttrans-
lational regulation. Dev Cell. 2009;16:633–47.
compromising, there have been reports of increased fracture 3. Andersson ER, Sandberg R, Lendahl U. Notch signaling: simplicity
incidence in young children with ALGS [40, 41]. While the in design, versatility in function. Development. 2011;138:3593–612.
increased fracture risk could be secondary to decreased 4. Canalis E. Notch signaling in osteoblasts. Sci Signal. 2008;1:17.
Curr Osteoporos Rep (2013) 11:126–129 129

5. Deregowski V, Gazzerro E, Priest L, Rydziel S, Canalis E. Notch 1 23. Yamada T, Yamazaki H, Yamane T, et al. Regulation of osteoclast
overexpression inhibits osteoblastogenesis by suppressing Wnt/ development by Notch signaling directed to osteoclast precursors
beta-catenin but not bone morphogenetic protein signaling. J Biol and through stromal cells. Blood. 2003;101:2227–34.
Chem. 2006;281:6203–10. 24. Fukushima H, Nakao A, Okamoto F, et al. The association of
6. • Sciaudone M, Gazzerro E, Priest L, Delany AM, Canalis E. Notch2 and NF-kappaB a ccelera tes R ANKL-induced
Notch 1 impairs osteoblastic cell differentiation. Endocrinology. osteoclastogenesis. Mol Cell Biol. 2008;28:6402–12.
2003;144:5631–9. An early in vitro study demonstrating a 25. Sekine C, Koyanagi A, Koyama N, Hozumi K, Chiba S, Yagita H.
suppresive role for Notch signaling in osteoblast differentiation. Differential regulation of osteoclastogenesis by Notch2/Delta-like
7. Tezuka K, Yasuda M, Watanabe N, et al. Stimulation of osteoblastic 1 and Notch1/Jagged1 axes. Arthritis Res Ther. 2012;14:R45.
cell differentiation by Notch. J Bone Miner Res. 2002;17:231–9. 26. Brennan AM, Pauli RM. Hajdu-Cheney syndrome: evolution of
8. Nobta M, Tsukazaki T, Shibata Y, et al. Critical regulation of bone phenotype and clinical problems. Am J Med Genet. 2001;100:
morphogenetic protein-induced osteoblastic differentiation by Del- 292–310.
ta1/Jagged1-activated Notch1 signaling. J Biol Chem. 2005;280: 27. Isidor B, Lindenbaum P, Pichon O, et al. Truncating mutations in
15842–8. the last exon of NOTCH2 cause a rare skeletal disorder with
9. Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ, Tonegawa S. osteoporosis. Nat Genet. 2011;43:306–8.
Skeletal and CNS defects in Presenilin-1-deficient mice. Cell. 28. Simpson MA, Irving MD, Asilmaz E, et al. Mutations in NOTCH2
1997;89:629–39. cause Hajdu-Cheney syndrome, a disorder of severe and progres-
10. Wong PC, Zheng H, Chen H, et al. Presenilin 1 is required for sive bone loss. Nat Genet. 2011;43:303–5.
Notch1 and DII1 expression in the paraxial mesoderm. Nature. 29. • Majewski J, Schwartzentruber JA, Caqueret A, et al. Mutations in
1997;387:288–92. NOTCH2 in families with Hajdu-Cheney syndrome. Hum Mutat.
11. Dunwoodie SL, Clements M, Sparrow DB, Sa X, Conlon RA, 2011;32:1114–7. These studies identify NOTCH2 mutations as the
Beddington RS. Axial skeletal defects caused by mutation in the cause for Hajdu-Cheney Syndrome.
spondylocostal dysplasia/pudgy gene Dll3 are associated with 30. Weng AP, Ferrando AA, Lee W, et al. Activating mutations of
disruption of the segmentation clock within the presomitic meso- NOTCH1 in human T cell acute lymphoblastic leukemia. Science.
derm. Development. 2002;129:1795–806. 2004;306:269–71.
12. • Hilton MJ, Tu X, Wu X, et al. Notch signaling maintains bone 31. Isidor B, Le Merrer M, Exner GU, et al. Serpentine fibula-
marrow mesenchymal progenitors by suppressing osteoblast dif- polycystic kidney syndrome caused by truncating mutations in
ferentiation. Nat Med. 2008;14:306–14. This mouse genetic study NOTCH2. Hum Mutat. 2011;32:1239–42.
establishes that physiological Notch singaling suppresses bone 32. Gray MJ, Kim CA, Bertola DR, et al. Serpentine fibula polycystic
formation in vivo. kidney syndrome is part of the phenotypic spectrum of Hajdu-
13. • Tu X, Chen J, Lim J, et al. Physiological notch signaling main- Cheney syndrome. Eur J Hum Genet. 2012;20:122–4.
tains bone homeostasis via RBPjk and Hey upstream of NFATc1. 33. Turnpenny PD, Ellard S. Alagille syndrome: pathogenesis, diag-
PLoS Genet. 2012;8:e1002577. This study delineates the mecha- nosis and management. Eur J Hum Genet. 2012;20:251–7.
nism through which physiological Notch signaling suppresses 34. Oda T, Elkahloun AG, Pike BL, et al. Mutations in the human
bone formation, and identifies Notch2 as a critical regulator. Jagged1 gene are responsible for Alagille syndrome. Nat Genet.
14. Salie R, Kneissel M, Vukevic M, et al. Ubiquitous overexpression 1997;16:235–42.
of Hey1 transcription factor leads to osteopenia and chondrocyte 35. Li L, Krantz ID, Deng Y, et al. Alagille syndrome is caused by
hypertrophy in bone. Bone. 2010;46:680–94. mutations in human Jagged1, which encodes a ligand for Notch1.
15. Engin F, Yao Z, Yang T, et al. Dimorphic effects of Notch signaling Nat Genet. 1997;16:243–51.
in bone homeostasis. Nat Med. 2008;14:299–305. 36. • McDaniell R, Warthen DM, Sanchez-Lara PA, et al. NOTCH2
16. • Tao J, Chen S, Yang T, et al. Osteosclerosis owing to Notch gain mutations cause Alagille syndrome, a heterogeneous disorder of
of function is solely Rbpj-dependent. J Bone Miner Res. the notch signaling pathway. Am J Hum Genet. 2006;79:169–73.
2010;25:2175–83. These two mouse genetic studies demonstrate These studies discovered the role of impaired Notch signaling in
that hyperactivation of Notch signaling through RBPj impairs Alagille syndrome.
bone homeostasis. 37. Sanderson E, Newman V, Haigh SF, Baker A, Sidhu PS.
17. Zanotti S, Smerdel-Ramoya A, Stadmeyer L, Durant D, Radtke F, Vertebral anomalies in children with Alagille syndrome: an
Canalis E. Notch inhibits osteoblast differentiation and causes analysis of 50 consecutive patients. Pediatr Radiol. 2002;32:
osteopenia. Endocrinology. 2008;149:3890–9. 114–9.
18. Murtaugh LC, Stanger BZ, Kwan KM, Melton DA. Notch signal- 38. Berrocal T, Gamo E, Navalón J, et al. Syndrome of Alagille:
ing controls multiple steps of pancreatic differentiation. Proc Natl radiological and sonographic findings. A review of 37 cases. Eur
Acad Sci U S A. 2003;100:14920–5. Radiol. 1997;7:115–8.
19. • Canalis E, Parker K, Feng JQ, Zanotti S. Osteoblast lineage- 39. Krantz ID, Piccoli DA, Spinner NB. Alagille syndrome. J Med
specific effects of Notch activation in the skeleton. Endocrinology. Genet. 1997;34:152–7.
2013;154(2):623–34. This study highlights the stage-specific 40. Hoffenberg EJ, Narkewicz MR, Sondheimer JM, Smith DJ,
effects of hyperactive Notch signaling in the osteoblast lineage. Silverman A, Sokol RJ. Outcome of syndromic paucity of
20. Canalis E, Parker K, Feng JQ, Zanotti S. Osteoblast lineage- interlobular bile ducts (Alagille syndrome) with onset of cholesta-
specific effects of notch activation in the skeleton. Endocrinology. sis in infancy. J Pediatr. 1995;127:220–4.
2013;154:623–34. 41. Bales CB, Kamath BM, Munoz PS, et al. Pathologic lower ex-
21. Novack DV, Teitelbaum SL. The osteoclast: friend or foe? Annu tremity fractures in children with Alagille syndrome. J Pediatr
Rev Pathol. 2008;3:457–84. Gastroenterol Nutr. 2010;51:66–70.
22. • Bai S, Kopan R, Zou W, et al. NOTCH1 regulates 42. • Kung AW, Xiao SM, Cherny S, et al. Association of JAG1 with
osteoclastogenesis directly in osteoclast precursors and indirectly bone mineral density and osteoporotic fractures: a genome-wide
via osteoblast lineage cells. J Biol Chem. 2008;283:6509–18. This association study and follow-up replication studies. Am J Hum
study demonstrates both direct and osteoblast-mediated regula- Genet. 2010;86:229–39. This study Links JAG1 polymorphism
tion of osteoclastogenesis by Notch. with bone mineral density in a diverse human population.

You might also like