Você está na página 1de 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/260119004

Dexamethasone Use During Pregnancy: Potential


Adverse Effects on Embryonic Skeletogenesis

Article in Current pharmaceutical design · February 2014


DOI: 10.2174/1381612820666140205144534 · Source: PubMed

CITATIONS READS

4 316

4 authors:

Xin Cheng Guang Wang


University of Jinan (Jinan, China) Jinan University (Guangzhou, China)
46 PUBLICATIONS 126 CITATIONS 72 PUBLICATIONS 174 CITATIONS

SEE PROFILE SEE PROFILE

Kenneth Ka-Ho Lee Xuesong Yang


The Chinese University of Hong Kong Jinan University (Guangzhou, China)
198 PUBLICATIONS 2,152 CITATIONS 136 PUBLICATIONS 2,101 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

biomedicine View project

The mechanism of birth defects derived from early embryonic development View project

All content following this page was uploaded by Kenneth Ka-Ho Lee on 11 March 2014.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.net
Current Pharmaceutical Design, 2014, 20, 000-000 1

Dexamethasone Use During Pregnancy: Potential Adverse Effects on Embryonic


Skeletogenesis

Xin Cheng1*, Guang Wang1, Kenneth Ka Ho Lee2 and Xuesong Yang1,3*

1
Department of Histology & Embryology, Joint Lab for Brain Function and Health, School of Medicine, Jinan University, Guangzhou
510632, China; 2Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong,
Shatin, Hong Kong; 3Institute of Fetal-Preterm Labor Medicine, Jinan University, Guangzhou 510632, China

Abstract: Glucocorticoids are important regulators of cell differentiation and mesenchymal cell lineage commitment during skeletogene-
sis. In clinical practice, it has been difficult to study the effects of glucocorticoids on target tissues because patients taking glucocorticoids
often suffer from adverse skeletal effects. Dexamethasone (Dex) is a long-acting synthetic corticosteroid hormone that ranks amongst the
most widely used prescribed drugs, and it is a powerful medication that is increasingly employed during the perinatal and neonatal peri-
ods. However, Dex is a potential teratogen. In particular, it has been claimed that Dex exposure during pregnancy can affect osteogenesis
in the developing embryo, although this claim remains highly controversial. In this review, we summarize the published data from nu-
merous clinical follow-up, animal-based and in vitro studies on the effects of Dex exposure on embryonic skeletogenesis. These studies
indicate that Dex may adversely affect skeletal progenitor cells during development. In addition, Dex can exert a number of effects on
bone growth at different developmental stages. We also discuss how glucocorticoids influence the BMP, FGF, Hedgehog and Wnt signal-
ing pathways, which are key regulators of skeletogenesis in the embryo. A fuller understanding of the negative, and perhaps teratogenic,
effects of Dex on skeletogenesis will have important implications for the routine use of Dex in clinical practice.
Keywords: Glucocorticoids, dexamethasone (Dex), chondrogenesis, osteogenesis, signal transduction pathways.

INTRODUCTION increasing. Dex has also been used to reduce the risk of neonatal
Glucocorticoids are considered important regulators of os- respiratory distress syndrome and as a medication for several types
teogenic cell differentiation and mesenchymal cell lineage com- of severe pregnancy disorders, including congenital adrenal hyper-
mitment. However, the functional effects of glucocorticoids on plasia, premature delivery caused by pregnancy-induced hyperten-
target tissues and organs are still not fully understood, and patients sion (PIH), placenta previa and multiple pregnancies [6, 9-12].
taking glucocorticoids often suffer from harmful side effects. For However, certain data suggest that human fetal exposure to Dex has
example, children that are given glucocorticoids show reduced bone detrimental effects on birth outcome, childhood cognition and long-
quality (also observed in adults), which can impact their growth and term behavior, the ramifications of which may be permanent. In
stature [1]. It has been reported that the treatment of childhood addition, the prenatal administration of Dex can affect lifespan and
asthma, autoimmune diseases and pediatric cancers with the long- predisposition to chronic diseases [13]. Therefore, opinions differ
term administration of anti-inflammatory glucocorticoids can result on whether glucocorticoids should be used therapeutically in pre-
in growth retardation, bone loss and potentially premature or severe mature infants to prevent prematurity-related complications and to
osteoporosis. In addition, there is a growing body of evidence treat neonatal respiratory distress syndrome. Despite this, Dex re-
showing that even short-term administration of glucocorticoids can mains widely used in primary hospitals as one of the most effective,
alter bone growth and turnover and that these effects can vary de- long-term glucocorticoids.
pending on how frequently (e.g., single or repeated dose) glucocor- As the use of Dex has been extensive, it is extremely important
ticoids are administered [2-7]. On the other hand, a fetus is nor- that we understand any potentially harmful side effects it may have
mally exposed to elevated levels of endogenous glucocorticoids on embryonic development. The effects of Dex on adults and ado-
during the late gestational period, which is essential for the adaption lescents have been much better studied compared with its effects on
and maturation of developing bone during postnatal life. embryos, and we know relatively little of how Dex exposure affects
Dexamethasone (Dex), a synthetic long-acting corticosteroid signaling molecules and pathways during development. Moreover,
hormone, is one of the most widely prescribed drugs for the treat- the majority of current studies address the neurotoxic effects of Dex
ment of inflammatory disorders such as arthritis, swelling, redness during development, rather than discussing the merits and demerits
of the skin, adrenal hormone insufficiency, resistance to adrenal of Dex treatment during the perinatal period [7, 14-19]. In addition,
hormones, and asthma and kidney disorders. Furthermore, Dex there are relatively few reports on the consequences of Dex expo-
induces the expression of the xenobiotic-metabolizing enzyme CYP sure on fetal bone growth. Therefore, the aim of this review is to
in the liver, and it has therefore also been used to amplify the ef- provide information on the effects of Dex usage on skeletogenesis
fects of anti-cancer drugs [8] and as an illicit enhancer of athletic during the prenatal period and to highlight previously published
performance. clinical follow-up, animal-based and in vitro studies.
In the clinical setting, Dex is one of the most powerful drugs VERTEBRATE SKELETOGENESIS
prescribed during the perinatal and neonatal periods, and its use is
Bones are normally formed through either intramembranous
ossification or endochondral ossification, which are distinct but
*Address correspondence to these authors at the Division of Histology & related morphogenetic processes. During intramembranous ossifica-
Embryology, Medical College, Jinan University, No. 601 Huangpu Road tion – which is how the bones in the skull, jaw and collarbones are
West, Guangzhou 510632, PR China; Tel: +86-20-85228316, E-mail: formed – richly vascularized mesenchymal cells condense to form
yang_xuesong@126.com nodules and flat plates that then directly differentiate into os-
Tel: +86-20-85220254, E-mail: chengxin633@gmail.com.cn
teoblasts. However, the vertebrate skeleton primarily develops via

1381-6128/14 $58.00+.00 © 2014 Bentham Science Publishers


2 Current Pharmaceutical Design, 2014, Vol. 20, No. 00 Cheng et al.

endochondral ossification, which is a well-orchestrated and intri- stages. At the tailbud stage, GR is expressed in the anterior portion
cately controlled set of processes that includes the condensation, of the embryo, including the somites. Furthermore, when zygotes
proliferation, migration, differentiation and activation of multiple over-expressing GR were treated with Dex at the blastula stage,
cell types [20-22]. In particular, endochondral ossification is how early differentiation was significantly inhibited.
long bones and the growth plates within long bones develop during
the embryonic and postnatal periods, respectively. In addition, os- EMBRYONIC SOMITES
teogenesis occurs not only in growing bones but also throughout Hansen et al. (1994) examined the toxicity of Dex in mouse and
adult life, and it is involved in the formation of primary and secon- rat embryos, and they observed a significant reduction in the num-
dary ossification centers as well as in bone absorption. Indeed, bone ber of somite pairs produced when embryos were treated with Dex.
turnover is very active in young children and can occur up to 200 As somites differentiate to form the sclerotome, which will eventu-
times as quickly as it does in adults. Nevertheless, the most crucial ally form the vertebrae, it was therefore not surprising that Dex
and active period of osteogenesis is during embryogenesis. treatment led to malformations of the axial skeleton. The crown-
rump and head lengths of these embryos were also significantly
THE EFFECTS OF DEXAMETHASONE ON PROGENITOR reduced. In addition, these researchers reported that mouse embryos
CELLS DURING SKELETAL DEVELOPMENT IN THE cultured in vitro were more adversely affected by Dex than were rat
EMBRYO embryos, even at lower concentrations [30]. This difference in the
The vertebrate skeleton is derived from three cell populations sensitivity of these two species to Dex may be attributed to inherent
during embryonic development. Neural crest cells give rise to the genetic differences or to minor differences in the developmental
craniofacial bones [23], the sclerotome compartments of somites staging of the embryos used in the culturing experiments.
form the axial skeleton [24], and lateral plate mesoderm cells form Glucocorticoids exert their effects primarily through the GR,
the limb bones [25]. and these receptors are extensively expressed in mammalian tissues,
including bone growth plates [31, 32]. Many studies have been
NEURAL CREST CELLS
conducted to examine how glucocorticoids regulate skeletogenesis,
During the early stages of embryonic development, neural crest and these studies indicate that glucocorticoids affect the formation
cell derivatives become responsive to glucocorticoids once the of hyaline cartilage, which forms the prerequisite limb scaffolding
sympathetic ganglia are formed, and Dex plays a crucial role in the necessary for endochondral ossification in the long bones. Studies
migration and differentiation of these neural crest cells. Several involving the use of a GR morpholino on developing embryos
retrospective studies have been carried out on infants born to showed that it had the ability to delay somitogenesis, induce somite
women who had received multiple courses of glucocorticoids dur- and tail malformations, and reduce embryo size. A key finding of
ing pregnancy, and these studies reported reduced body weight and this study was the 70-90% reduction in bone morphogenetic pro-
head circumference at birth in these children. Furthermore, a 3-year teins (BMPs) expression observed following GR-morpholino treat-
follow-up study showed similar findings for children exposed to ment. Further molecular analysis identified multiple putative gluco-
only a single course of glucocorticoids during development [6]. corticoid response elements upstream of the BMP genes, and it was
Indeed, a single course of Dex is considered sufficient for prevent- shown that Exposure to the GR morpholino reduced expression of
ing prematurity-related complications. It has been reported that the BMP-regulated genes eve1 and pax3 [33].
exposure to Dex during early gestational stages can cause minor
The release of cells and tissues from growth-inhibitory condi-
cranial-skeletal abnormalities, including encephalocele and menin-
tions generally results in compensatory growth in mammals. In
gocele, which was also observed in rhesus macaques [26]. These
piglets, it has been shown that prenatal exposure to Dex during the
findings suggest that the development and migration of neural crest
last 24 days of the fetal period resulted in a dramatic reduction in
cells may be affected by Dex exposure during early gestation. Neu-
bone mineral density and content. In addition, they observed de-
ral crest cells also contribute to the formation of the palate, and
creases in blood serum osteocalcin levels as well as in various bone
animal models have been developed that use hydrocortisone, pred-
geometric parameters, ultimately increasing the risk of these piglets
nisolone and Dex on pregnant to produce embryos with cleft pal-
for developing bone fractures [34, 35]. Although prenatal Dex
ates. From these studies, it was shown that the teratogenicity of Dex
treatment did not reduce birth weight or growth rate during the first
is 300 times greater than that of hydrocortisone for inducing cleft
30 days post-birth, no compensatory catch-up growth in terms of
palate [27]. For this reason, Dex has been widely employed in ani-
bone mineralization was observed in these suckling piglets [34].
mal models for inducing cleft palate in vivo, although the precise
mechanism through which Dex induces this abnormality is not EMBRYONIC LIMB BUDS
clear. It is known that the adhesion and fusion of the palatal shelves
are necessary for the proper development of the secondary palate, The limb develops as an outgrowth, or limb bud, from the flank
and failures in these processes can lead to cleft palate. In the fetuses of the embryo consisting of the lateral plate mesoderm. The limb
of mothers exposed to Dex, the elevation of the palatal shelves is bud is composed of mesenchymal cells covered by a single layer of
delayed and the descent of the tongue is interrupted, which impedes ectoderm. At the distal end of the bud, the ectoderm thickens and
the rotation of the shelves, leading to a failure in the fusion of the forms a multilayered cellular structure called the apical ectodermal
shelves. Furthermore, the cell death and proliferation that normally ridge (AER) [36]. The AER secretes signaling molecules that
occur at the medial edge of the palatal epithelium can also be inhib- stimulate the underlying mesenchymal cells to rapidly proliferate
ited by Dex exposure during development, leading to altered cell while maintaining a multipotent state. This mesenchymal region is
fates, small palate size, delayed shelf elevation and unfused palates, known as the progress zone, and these cells eventually differentiate
which may be the most important mechanism underlying the failure into cartilage, perichondrium, tendon, muscle connective tissue and
of palatal shelf fusion [28]. dermis. The AER directs the mesenchyme in the progress zone to
differentiate into chondrogenic and non-chondrogenic precursors.
As Dex is a glucocorticoid receptor (GR) agonist, its effects on The limb bud forms a chondrogenic core and is surrounded periph-
embryonic development can be elucidated by studying the GR. The erally by soft connective tissue. Limb myogenic cells, which origi-
spatial-temporal expression pattern of GR in the early Xenopus nate from the somites, migrate into the limb bud and differentiate
embryo has been systematically mapped [29], and it was found that into skeletal muscles in a pattern that is directed by the connective
GR mRNAs are localized to the dorsal ectoderm during the gastrula tissues. Highly coordinated signaling between the AER and the
stage and to the notoplate during the early neurula stage; GR ex- underlying mesenchymal cells specifies the distal structures of the
pression is lost sometime between the middle and late neurula limb and also maintains the survival of the mesenchymal cells. In
Dexamethasone Use During Pregnancy Current Pharmaceutical Design, 2014, Vol. 20, No. 00 3

particular, the signaling genes involved in this process are Shh, M) of Dex. It appears that Dex acts on cartilage by regulating mul-
Hox, Fgf10, Fgf8, Tbx4, Tbx5 and the BMPs, which must be ex- tiple classes of genes to promote the accumulation of ECM and the
pressed in a precise spatiotemporal fashion for the limbs to develop activation of metabolic-related genes to maintain the chondrocytic
and pattern properly [37-40]. However, there are few reports in the phenotype. At the same time, Dex repressed cytokine and growth
literature of the effects of glucocorticoids on limb bud develop- factor synthesis, which can accelerate the shift from cartilage to
ment. bone phenotypes. In addition, Dex-induced gene expression data
have been compared with gene expression data from various devel-
THE REGULATORY EFFECTS OF DEXAMETHASONE ON opmental phases of micromass cultures. These results indicate that
DIFFERENT STAGES OF BONE DEVELOPMENT Dex maintained the expression of various chondrocytic marker
Endochondral ossification is composed of two crucial morpho- genes while inhibiting factors that promote the vascularization and
genetic stages. First, the mesenchymal progenitor cells condense ossification of the cartilaginous anlagen [48]. These studies suggest
into a nodule, which is then followed by the differentiation of these that Dex exerts its effects in a tissue-specific manner, and determin-
cells into chondroblasts and perichondrium in a process called ing the precise network of genes influenced by Dex treatment
chondrogenesis. Next, the cartilage scaffolding is replaced by mi- should allow us to understand the mechanisms through which Dex
crovasculature, osteoblasts, osteoclasts, osteocytes and bone matrix, affects vertebrate skeletogenesis.
ultimately giving rise to bone. Crucially, bone growth depends on
the tight coordination between these two morphogenetic processes. THE INFLUENCE OF DEXAMETHASONE ON SIGNAL
Glucocorticoids have been implicated in the regulation of chondro- TRANSDUCTION AND SKELETOGENESIS
genesis and osteoblast differentiation, as well as in maintaining the A large number of studies have implicated several signaling
homeostasis of cartilage and bone at a physiological level. molecules, including growth factors and hormones, in the regula-
tion of skeletogenesis. Chondrocytes and osteoblasts constantly
CHONDROGENESIS receive signals from surrounding cells and tissues, and even from
Pharmacological doses of glucocorticoids have been shown to distant organs, that regulate their proliferation, biological activity
inhibit chondrocyte proliferation within the growth plate, and this is and survival. Two of the fundamental questions of interest to devel-
one of the major side effects of glucocorticoids that suppress longi- opmental biologists are how undifferentiated precursor cells acquire
tudinal bone growth. More specifically, Dex exposure significantly spatial patterning information, and in turn, how these cells differen-
reduces cell number, cell proliferation, and proteoglycan synthesis tiate and respond to the information provided. The developing ver-
and increases alkaline phosphatase (ALP) activity during the chon- tebrate limb has been extensively studied as a model for addressing
drogenesis in the ATDC5 cell line, which mimics the in vivo proc- these questions, and as a consequence, the body of knowledge gen-
ess of longitudinal bone growth; apoptosis was unaltered in this erated by such studies has provided novel insights into the mecha-
model [41]. On the other hand, Dex has been used in combination nisms governing the development of skeletal phenotypes and the
with transforming growth factor beta-1 (TGF-1) to induce the signaling pathways involved. Nevertheless, the global regulatory
differentiation of mesenchymal stem cells (MSCs) from a variety of effects of glucocorticoids on the expression of key skeletal-related
tissues into chondrocytes [42-44]. In fact, the effects of Dex on the genes has not been comprehensively evaluated, and in particular,
chondrogenic differentiation of MSCs is influenced by microenvi- how these signaling pathways are spatiotemporally regulated to
ronment and tissue source, as well as by the nature of the particular orchestrate growth and differentiation during embryogenesis re-
growth factor used [45], suggesting that Dex is necessary for MSC mains unclear. Ultimately, we wish to know how the disruption of
chondrogenesis. Some of the regulatory targets of glucocorticoids these signaling pathways, through either genetic mutations and/or
during chondrogenesis are now known [46], and they include ex- environmental factors, leads to skeletal malformations. The impor-
tracellular matrix (ECM)-related, metabolic, cytokine and growth- tance of the precise regulation of signal transduction for normal
factor genes. However, the global effects of glucocorticoids (at development is highlighted by the observation that these pathways
pharmacological doses) on chondrocyte gene expression have not are often misregulated in adult diseases and congenital malforma-
yet been comprehensively evaluated. tions. These findings are summarized here, and the important fea-
tures of the primary signaling pathways will be discussed as they
OSSIFICATION relate to glucocorticoids and Dex-mediated skeletogenesis.
During endochondral bone formation, terminal differentiation
of chondrocytes involves a number of steps: the rate of cell prolif- BONE MORPHOGENETIC PROTEINS
eration decreases; the chondrocytes become hypertrophic; the ECM The BMPs are a family of secreted proteins that have multi-
surrounding the hypertrophic chondrocytes becomes progressively functional roles in specifying the dorsal-ventral axis embryos and in
mineralized; and finally, the mineralized matrix serves as a tem- determining cell fate at the neural plate and during limb bud devel-
plate for bone deposition. This chondrocyte maturation process is opment [49]. BMPs induce a variety of biological activities in an
precisely controlled, with chondrocyte proliferation and terminal assortment of cell types at various stages of differentiation, al-
differentiation being tightly balanced, and indeed, imbalances can though their roles during normal physiology are still not completely
result in an abnormal bone development. In general, the degree of clear. Nevertheless, there is no doubt that BMPs play an essential
chondrocyte maturation is evaluated by measuring the levels of role in endochondral bone development [50].
ALP expression and the presence of type X collagen within the When mesenchymal progenitor cells are treated with Dex, it
mineralized matrix. induces these cells to differentiate into chondroblasts or osteoblasts.
Leclerc et al. (2004) revealed that Dex could arrest osteoblast However, the effect of Dex on cell differentiation is enhanced in the
development in MC3T3-E1 cell cultures [47]. In contrast, Dex had presence of BMP-2 [51, 52]. In addition, the decision made by pro-
little effect on terminal differentiation in the chondrocyte cell line genitor cells to differentiate into either chondroblasts or osteoblasts
ATDC-5 [41]. It appears that the ability of Dex to inhibit osteoblast is dependent on the microenvironment and whether other inducers
development is strongly dependent on concentration and the timing are also present [53-55]. It has also been reported that BMP-7 is
of administration, which suggests that there is a specific period of expressed by proliferating chondrocytes [56].
skeletogenesis during which that tissue is sensitive to Dex expo- As described above, chondrocyte maturation is precisely con-
sure. trolled to yield a balance between proliferation and terminal differ-
A comparative microarray analysis was conducted on mouse entiation during endochondral ossification. BMPs exert profound
embryonic chondrocytes treated with a pharmacological dose (10-7 effects on chondrocyte maturation, and BMP-2 and BMP-6 are
4 Current Pharmaceutical Design, 2014, Vol. 20, No. 00 Cheng et al.

expressed in hypertrophic chondrocytes. Microarray analysis of FIBROBLAST GROWTH FACTORS


Dex-arrested osteoblasts in MC3T3-E1 cells revealed that BMP The fibroblast growth factor (FGF) family is composed of 22
biological activity was inhibited as a result of the strong repression distinct FGF genes and 4 FGF receptor genes. These molecules are
of Krox20/Egr2. Furthermore, when Follistatin (a BMP antagonist) expressed at nearly every stage of bone formation, and they un-
was over-expressed in cells, it negatively regulated by Krox20 [47]. doubtedly play key roles in the regulation of bone development.
When ascorbate and BMP-2 inducers are combined and added to During the earliest stages of bone development, FGF receptor-1
sternal pre-hypertrophic chondrocytes, they induced a threefold (FGFR1) is expressed in prehypertrophic/hypertrophic chondro-
increase in ALP levels compared with controls [57]. Minina 2001 cytes and in the perichondrium. FGFR2 is expressed in the chon-
reported that the addition of BMP-2 to mouse and chick limb ex- drogenic condensation, perichondrium, periosteum and primary
plant cultures dramatically increased chondrocyte proliferation and spongiosa. FGFR3 is expressed in proliferating chondrocytes. Fi-
delayed terminal differentiation (i.e., chondrocyte hypertrophy) nally, FGF-7, -8 -17 and -18 are expressed in the perichondrium
[58]. It is possible that these different observed effects for inducers [60]. Each of these growth factors appears to play distinct and im-
containing the same signaling molecules is partially due to the dif- portant roles in bone development, although their exact roles have
ferent drug concentrations used or the specific culturing systems not been fully defined. FGF-2, platelet-derived growth factors
and manipulation protocols. (PDGFs) and TGF-1 have long been recognized as inducers of
chondrogenic phenotypes in MSCs, and they have been used to
INDIAN HEDGEHOG AND PARATHYROID HORMONE-
generate cartilage grafts [53, 61-64]. This process requires specific
RELATED PROTEINS
culture conditions that permit rapid the rapid expansion of MSCs
During endochondral ossification, the Indian hedgehog (Ihh) while maintaining the potential of these cells for further differentia-
and parathyroid hormone-related (PTHrP) proteins have been iden- tion. Dex is routinely added to MSC cultures to promote cell prolif-
tified as components of the negative feedback loop that regulates eration and cell pellet expansion, and these effects can be enhanced
chondrocyte hypertrophic differentiation. Ihh is a dominant regula- when combined with a low concentration of FGF-2 (1 ng/mL) [65-
tor of bone development and is involved in coordinating chondro- 67]. Dex enhances chondrogenesis and osteogenesis in pellet cul-
cyte proliferation and differentiation, as well as osteoblast differen- tures by increasing nodule formation, glycosaminoglycan deposi-
tiation. This protein directly stimulates chondrocyte proliferation tion, collagen type II synthesis and SOX9 expression. It also posi-
and determines the length of the proliferating chondrocyte columns tively promotes cell maturation by increasing expression of the
through the stimulation of PTHrP synthesis. Furthermore, Ihh ex- developmental markers STRO-1 and ALP [67, 68]. MSCs ex-
pression also defines the sites where hypertrophic differentiation panded with Dex/FGF-2 display lower ALP levels than Dex-
begins. In the cartilage growth plate, the biological activities of expanded cells but significantly higher ALP levels than FGF-2-
mature chondrocytes and early hypertrophic chondrocytes are pre- expanded cells [67]. These results suggest that both Dex and FGF-2
cisely regulated by Ihh and PTHrP, which is essential for normal are crucial components in the maintenance of osteogenic potential,
longitudinal growth. although their exact roles are still obscure.
Ihh is closely related to Sonic hedgehog (Shh), one of the main Among the FGF receptors, the function of FGFR3 is most fully
regulators of limb bud outgrowth. During endochondral bone de- understood. FGF signaling is able to inhibit cell proliferation
velopment, Ihh is synthesized by early hypertrophic chondrocytes through FGFR3. When Fgfr3 is knocked out in mice, it leads to
and by chondrocytes just leaving the proliferative zone, which are severe and progressive bone dysplasia with enhanced and pro-
known as prehypertrophic chondrocytes. When MSCs are exposed longed endochondral bone growth. This phenotype is accompanied
to low doses of Dex (10-8 M), they differentiate into osteoblasts, by the expansion of proliferating chondrocyte columns and hyper-
which is indicated by enhanced ALP activity and the production of trophic chondrocytes within the cartilaginous growth plate [69]. In
collagen type I. Dex can also enhance Shh expression via a Gli1- humans, Fgfr3 mutations cause achondroplasia, which is the most
independent mechanism during osteoblast differentiation in MSCs. common type of dwarfism [70, 71]. Knocking out Fgf-18 in mice
The expression of both Ihh and Gli1 is down-regulated during this also leads to increased chondrocyte proliferation and delayed ossi-
process [59]. These findings partially support the observations of fication, similar to what is observed in Fgfr3-knockout mice [72].
Heine and Rowitch [14] that chronic treatment of mouse pups with Therefore, FGF18 is thought to play a role in FGFR3 signaling.
glucocorticoids inhibits neonatal cerebellar growth and neuronal
D8-SBMC is a cell line established from rat bone marrow that
proliferation by promoting early cell cycle exit and premature dif-
is capable of producing bone-like nodules and robust mineralization
ferentiation. In vitro analysis revealed that glucocorticoids re-
when induced with FGF-2 and Dex. When AJ18 (a regulator of
pressed Gli1 (a target of Shh) expression, suggesting the existence
bone formation that is up-regulated by BMP-7) is over-expressed in
of a mutual antagonism between glucocorticoids and Shh and that
D8-SBMC cells, it increases cell proliferation and mineralization.
different patterns of hedgehog signaling are involved in Dex-
Expression of bone-related gene markers is also up-regulated by
induced development.
AJ18 over-expression [73]. Furthermore, the biphasic modulatory
Studies involving transgenic mice that specifically over- effects of AJ18 on cell proliferation and mineralization are associ-
expressed Ihh in chondrocytes revealed that PTHrP expression was ated with BMP-7.
up-regulated in these cells and that there was a delay in the onset of
Each of the signaling systems mentioned above is essential,
hypertrophic differentiation. In general, it is thought that BMPs are
although not sufficient, for bone formation; in other words, loss of
potential integrators of the Ihh/PTHrP feedback loop. Treatment of
any of these systems can prevent bone from developing normally.
the limbs obtained from these transgenic mice with Noggin (an
These signaling systems influence and interact with one another to
inhibitor of both BMP and Ihh/PTHrP signaling) revealed that this
provide the developing bone with precise patterning information for
molecule did not antagonize the effects of Ihh overexpression.
further growth. FGF signaling shortens chondrogenic proliferative
Conversely, the enhancement of chondrocyte maturation induced
columns in the cartilage scaffolding by directly inhibiting chondro-
by cyclopamine (an inhibitor of Ihh signaling) could not be re-
cyte proliferation and suppressing Ihh expression. When Fgfr3 is
versed by BMP-2. These observations suggest that BMP signaling
knocked out, it increases Ihh expression, whereas activation of
alone delays hypertrophic differentiation, independent of the
FGFR3 decreases Ihh expression. FGF signaling can also accelerate
Ihh/PTHrP signaling pathway. Or in other words, BMP signaling
the terminal differentiation of hypertrophic chondrocytes independ-
does not act as a downstream signal of Ihh/PTHrP to delay the on-
ently of Ihh and PTHrP. BMPs can oppose the effects of FGF sig-
set of hypertrophic differentiation [58].
naling by increasing chondrocyte proliferation and inducing Ihh
expression. BMPs and FGFs have opposing effects on terminal
Dexamethasone Use During Pregnancy Current Pharmaceutical Design, 2014, Vol. 20, No. 00 5

hypertrophic chondrocyte differentiation, and these pathways can cultures. Moreover, treatment of the transgenic cultures with Wnt3a
be considered as antagonistic at several levels [74]. In this context, was shown to stimulate osteoblastogenesis and inhibit adipogenesis
chondrocyte proliferation, osteoblast differentiation and hypertro- [80]. Taken together, these results demonstrate a central role for
phic differentiation are the result of antagonistic interactions be- osteoblasts in the regulation of early lineage commitment in pro-
tween several signaling pathways. Therefore, simply increasing or genitor cells, which can be blocked by the loss of Wnt signaling.
decreasing one of these signaling systems cannot account for the Cells that express Wnt3a often show enhanced proliferation and cell
variety of phenotypes observed during skeletogenesis. survival. Wnt3a can also up-regulate ALP expression without influ-
encing matrix mineralization but represses most of the other genes
WNT PROTEIN LIGAND associated with osteogenic differentiation, which can be partially
In humans, the Wnt family is composed of 19 members. They reversed by BMP-2 [81]. Furthermore, the abnormalities observed
are involved cell-cell communication and are essential for normal in Col2.3-11HSD2 mice can be rescued by supracalvarial injection
embryonic development and differentiation. The Wnt signaling of Wnt3a protein [78], demonstrating the role of Wnt3a in os-
pathway is activated when Wnt protein ligands bind to a Frizzled teoblastogenetic potential. The up-regulation of ALP expression
family receptor protein. These signaling pathways are activated in a induced by Wnt-3a could be effectively suppressed through the
highly coordinated manner to provide positional information to combined activity of osteogenic supplements, Dex and 1,25-
cells in order to determine their proper identity and developmental dihydroxyvitamin D (1,25(OH)2D3) [78, 81].
fate and ensure proper embryonic patterning. Wnts affect many Glucocorticoids exert both anabolic and catabolic effects on
diverse processes, including embryonic induction, generation of cell bone, which are partly mediated by Wnt signaling molecules and
polarity and cell fate specification [75]. During early craniofacial their inhibitors in mature osteoblasts [82]. The mRNA levels of
development in mouse embryos, Wnt family members are ex- Wnt2, Wnt2b, Wnt4, Wnt5a, Wnt10b, and Wnt11 are significantly
pressed in a variety of locations, including the tooth initiation sites higher in osteoblasts than in their progenitor cells. In particular, the
of the oral epithelium, the mesenchyme of the elevating palatal expression of Wnt7b and Wnt10b in osteoblasts is modulated by
shelves, and the prospective sites of the maxilla and mandible glucocorticoids in a biphasic manner, with both genes being up-
bones. The Wnt ligands function by activating both Wnt/-Catenin- regulated at low corticosterone concentrations and down-regulated
dependent and -Catenin-independent pathways. It has been re- at high concentrations. High concentrations of glucocorticoids also
ported that Wnt/-Catenin signaling was down-regulated in a Dex- increase the expression of the Wnt inhibitors sFRP-1 and DKK-1.
induced cleft palate mouse embryo model and that the downstream
molecules of Wnt/-catenin signaling, including -catenin, Lef-1, c- GROWTH HORMONE AND INSULIN-LIKE GROWTH
Jun and phospho-c-Jun, were completely inhibited by Dex treat- FACTOR-I
ment. Indeed, altered signaling in the affected cells directly im- The growth inhibition caused by glucocorticoid therapy during
paired their ability to form a normal palate [28]. In the limb bud, development is thought to be mediated by the somatotropic hor-
Wnt ligands secreted by the ectoderm promote the underlying mes- mone axis and by direct local effects on growth plate chondrocytes.
enchymal cells to proliferate via the activity of Nmyc and inhibit Therefore, changes in the secretion of growth hormone (GH) could
chondrogenic differentiation via the repression of Sox9 [37]. impair the longitudinal growth of the chondrocyte plate. The inter-
Col2.3-11HSD2 transgenic mice, which are characterized by actions between glucocorticoids, GH and parathyroid hormone
osteoblast-targeted disruption of glucocorticoid signaling, have (PTH) have recently been investigated in detail. In growing chon-
been used as a model to study the relationship between skeleto- drocytes cultures, the introduction of GH, PTH and 1,25(OH)2D3
genesis and glucocorticoids [76-78]. Micro-CT scans of Col2.3- increases chondrocyte proliferation by stimulating insulin-like
11HSD2 fetuses and neonates revealed they had hypoplasia and growth factor-1 (IGF-1) secretion in a paracrine fashion. Long-term
osteopenia of the skull bones, disorganized frontal and parietal high-doses of glucocorticoids decreased GH secretion, whereas
bones, increased suture patency, ectopic cartilage in the sagittal PTH and 1,25(OH)2D3 stimulated cell growth in a dose-dependent
suture, and disrupted postnatal removal of parietal cartilage. These manner [83]. Glucocorticoids at high doses reduced GH receptor
transgenic mice showed markedly reduced levels of Mmp14, an and IGF type-1 receptor (IGF-1R) expression. However, the pri-
enzyme essential for calvarial cartilage removal. Wnt9a and mary anti-proliferative effect of the glucocorticoids was to reduce
Wnt10b expression was also significantly reduced in osteoblasts the basal levels of hormone-stimulated IGF-1 secretion by human
with disrupted glucocorticoid signaling, which in turn caused a osteoblast-like cells [84]. These in vitro results are consistent with
reduction in -catenin (an upstream regulator of Mmp14) accumu- observations of animal models and children treated with glucocorti-
lation within osteoblasts, chondrocytes and mesenchymal progeni- coids showing that the inhibitory effects of glucocorticoids on bone
tors [78]. These observations suggest that intramembranous ossifi- formation in humans are mediated via a reduction in the
cation is tightly regulated by glucocorticoids and that this hormone autocrine/paracrine expression of IGF-1, which can be compensated
initiates and controls cartilage dissolution after birth. These findings for by supra-physiological levels of GH or IGF-1 [85].
agree with experiments conducted on transgenic mice lacking Axin2 Treatment with Dex, either continuously or on alternating days,
(a negative regulator of Wnts) and mutant for Fgfr1, which leads to induced the same degree of growth retardation in fetal metatarsal
the development of craniosynostosis. It appears that switching mes- cultures. Dex inhibited cell proliferation within the growing bone,
enchymal cell fate from osteoblasts to chondroblasts resulted in whereas IGF-1, either alone or in combination with Dex, induced
suture abnormalities, suggesting that ectopic endochondral ossifica- an increase in linear bone growth. GH alone at 100 ng/mL exerted
tion may be a mechanism for producing craniosynostosis [79]. no beneficial effects on metatarsal growth. However, IGF-1 signifi-
Therefore, the balance between Wnt and FGF signaling influences cantly increased the length of hypertrophic zone, whereas Dex
the developmental fate of mesenchymal cells during skeletogenesis. alone had no significant effect [86]. Treatment of osteogenic pre-
It has been reported that calvarial cell cultures produced from cursors with IGF-1, either alone or in combination with Dex, had no
Col2.3-11HSD2 mice were more prone to undergo adipogenesis significant effects on colony formation or the expression of ALP
than osteoblastogenesis. This change in cell fate commitment by and the developmental marker STRO-1. In contrast, exposing these
Col2.3-11HSD2 progenitor cells was associated with reductions in same cells to FGF-2 increased colony formation, cell proliferation
Wnt7b, Wnt10b and -catenin expression. However, co-culturing and expression of STRO-1/ALP. Normally, the  and  subunits of
these transgenic cells with wild-type osteoblasts restored the ten- IGF-1R are expressed in the majority of osteoprogenitor cells. Dex
dency of these cells to form osteoblasts. This effect could be treatment did not affect IGF-1R expression, although it did increase
blocked by the introduction sFRP1, a Wnt inhibitor, to the co- the number of cells co-expressing IGF-1 and ALP [87]. Taken to-
6 Current Pharmaceutical Design, 2014, Vol. 20, No. 00 Cheng et al.

Fig. (1). Dex exerts distinct effects at every stage of embryonic skeletogenesis. The vertebrate skeleton is derived from three cell populations: neural crest
cells, somites and limb buds. Dex reduces body weight and head circumference, causes cranial skeletal abnormalities and cleft palate, reduces somite pair
number and embryo size, delays somitogenesis, and induces somite and tail malformations; the precise effects of Dex on limb buds remains unclear.
Dex regulates proliferation and differentiation in chondrocytesand the process of bone ossification , although the effects are variable and may be de-
pendent on micro-environment. Such changes in proliferation and differentiation may account for the cellular mechanisms underlying the phenotypes caused
by Dex treatment. The global effects of Dex on key skeletal-related signaling pathways have not been comprehensively evaluated. In general, FGFs act to
decrease chondrocyte proliferation, increase expression of Ihh/PTHrP, and accelerate terminal differentiation in hypertrophic chondrocytes (solid-line arrow).
BMPs antagonize FGFs at several steps (broken line). Wnt/-catenin and GH/IGF-1 signaling play vital roles in promoting cell proliferation and inhibiting
chondrogenic differentiation (broken-line arrows).

gether, the effects of IGF-1 on skeletogenesis are not mediated cological doses) on vertebrate skeletogenesis and have established
through osteoprogenitor cells, which is consistent with the hypothe- the foundation for future functional studies. It is worth noting that
sis that IGF-1 exerts its effects on more mature cells of the os- the majority of the studies that have been reported, particularly
teoblast lineage. those relating to signal transduction and bone growth, have been
It has been reported that treatment of cultured chondrocytes conducted in vitro, which should be considered when interpreting
with Dex and IGF-1 repressed IGF-binding protein (IGFBP)-2 ex- the data. Undoubtedly, elucidation of the molecular pathways in-
pression without affecting expression of IGFBP-4 and -5. These volved in this process has been greatly enhanced by the availability
chondrocytes proliferated more rapidly than cells treated with IGF- of cell culture models. However, we should not neglect the fact that
1 alone. Untreated chondrocytes normally expressed IGFBP-2 Dex exerts a biphasic influence on chondrogenesis and osteoblast
through -6, IGF-1 and -2, and their corresponding IGF receptors. function in vitro and that it can both stimulate and suppress biologi-
Dex treatment up-regulated expression of IGFBP-5 and IGF-1R and cal activity in culture, depending on the Dex concentration used
down-regulated expression of IGFBP-2. However, the inhibitory [53]. There are also other disadvantages of using cell culture sys-
effects could be prevented by the addition of the GR antagonist tems. For example, the genetic background of a cell line may have
Org34116 [88]. Therefore, at pharmacological doses, Dex can in- been transformed such that the results produced may no longer
hibit cell proliferation and alter IGFBP-2, -5 and IGF-1R expres- accurately reflect the in vivo system. Another possibility is that a
sion, suggesting a role for the IGF axis in glucocorticoid-induced cell line may be heterogeneous, among others. Therefore, results
growth retardation. and conclusions drawn from in vitro studies must also be validated
using animal models and/or human bone biopsy studies, particularly
FUTURE PROSPECTS when trying to confirm whether Dex can adversely affect bone
In summary, Dex exerts different effects on every stage of em- growth. At present, more research will be required to establish a
bryonic skeletogenesis, the challenge of uncovering the link be- link between the multiple observed effects of Dex on bone growth.
tween Dex and skeletogenesis is now centered on determining the In addition, it will be useful to discover methods to preserve the
mechanisms and molecular signals involved in the activity of Dex effectiveness of Dex while minimizing its side effects during the
in vitro and in vivo (see Fig. 1). Previously published studies have prenatal period.
provided us with novel insights into the effects of Dex (at pharma-
Dexamethasone Use During Pregnancy Current Pharmaceutical Design, 2014, Vol. 20, No. 00 7

CONFLICT OF INTEREST [21] Stanton LA, Underhill TM, Beier F. MAP kinases in chondrocyte
differentiation. Dev Biol 2003; 263: 165-75.
The authors confirm that this article content has no conflicts of [22] Kanczler JM, Oreffo RO. Osteogenesis and angiogenesis: the po-
interest. tential for engineering bone. Eur Cell Mater 2008; 15: 100-14.
[23] Couly GF, Coltey PM, Le Douarin NM. The triple origin of skull in
ACKNOWLEDGEMENTS higher vertebrates: a study in quail-chick chimeras. Development
This study was supported by “973 Project” (2010CB529703); 1993; 117: 409-29.
NSFC grant (31071054, 30971493) and Guangdong Natural Sci- [24] Olivera-Martinez I, Coltey M, Dhouailly D, Pourquie O. Medio-
ence Foundation (S2011010001593, S2013010013392) to X Yang. lateral somitic origin of ribs and dermis determined by quail-chick
chimeras. Development 2000; 127: 4611-7.
REFERENCES [25] Pearse RV, 2nd, Scherz PJ, Campbell JK, Tabin CJ. A cellular
lineage analysis of the chick limb bud. Dev Biol 2007; 310: 388-
[1] Avioli LV. Glucocorticoid effects on statural growth. Br J Rheuma- 400.
tol 1993; 32 Suppl 2: 27-30. [26] Jerome CP, Hendrickx AG. Comparative teratogenicity of triamci-
[2] Ahmed SF, Wallace WH, Crofton PM, et al. Short-term changes in nolone acetonide and dexamethasone in the rhesus monkey
lower leg length in children treated for acute lymphoblastic leu- (Macaca mulatta). J Med Primatol 1988; 17: 195-203.
kaemia. J Pediatr Endocrinol Metab 1999; 12: 75-80. [27] Pinsky L, Digeorge AM. Cleft Palate in the Mouse: a Teratogenic
[3] Ahmed SF, Tucker P, Mushtaq T, et al. Short-term effects on linear Index of Glucocorticoid Potency. Science 1965; 147: 402-3.
growth and bone turnover in children randomized to receive pred- [28] Hu X, Gao JH, Liao YJ, et al. Dexamethasone alters epithelium
nisolone or dexamethasone. Clin Endocrinol (Oxf) 2002; 57: 185- proliferation and survival and suppresses Wnt/beta-catenin signal-
91. ing in developing cleft palate. Food Chem Toxicol 2013; 56: 67-74.
[4] Crofton PM, Ahmed SF, Wade JC, et al. Effects of intensive che- [29] Gao X, Stegeman BI, Lanser P, et al. GR transcripts are localized
motherapy on bone and collagen turnover and the growth hormone during early Xenopus laevis embryogenesis and overexpression of
axis in children with acute lymphoblastic leukemia. J Clin Endocri- GR inhibits differentiation after dexamethasone treatment. Bio-
nol Metab 1998; 83: 3121-9. chem Biophys Res Commun 1994; 199: 734-41.
[5] Antenatal betamethasone: reassuring long-term data. Prescrire Int [30] Hansen DK, Grafton TF. Comparison of dexamethasone-induced
2008; 17: 73. embryotoxicity in vitro in mouse and rat embryos. Teratog Car-
[6] Senat MV. [Corticosteroid for fetal lung maturation: indication and cinog Mutagen 1994; 14: 281-9.
treatment protocols]. J Gynecol Obstet Biol Reprod (Paris) 2002; [31] van der Eerden BC, Karperien M, Wit JM. Systemic and local
31: 5S105-13. regulation of the growth plate. Endocr Rev 2003; 24: 782-801.
[7] Rajadurai VS, Tan KH. The use and abuse of steroids in perinatal [32] Cole TJ, Blendy JA, Monaghan AP, et al. Targeted disruption of
medicine. Ann Acad Med Singapore 2003; 32: 324-34. the glucocorticoid receptor gene blocks adrenergic chromaffin cell
[8] Celander M, Hahn ME, Stegeman JJ. Cytochromes P450 (CYP) in development and severely retards lung maturation. Genes Dev
the Poeciliopsis lucida hepatocellular carcinoma cell line (PLHC- 1995; 9: 1608-21.
1): dose- and time-dependent glucocorticoid potentiation of [33] Nesan D, Kamkar M, Burrows J, et al. Glucocorticoid receptor
CYP1A induction without induction of CYP3A. Arch Biochem signaling is essential for mesoderm formation and muscle devel-
Biophys 1996; 329: 113-22. opment in zebrafish. Endocrinology 2012; 153: 1288-300.
[9] Ballabh P, Lo ES, Kumari J, et al. Pharmacokinetics of betametha- [34] Sliwa E, Dobrowolski P, Piersiak T. Bone development of suckling
sone in twin and singleton pregnancy. Clin Pharmacol Ther 2002; piglets after prenatal, neonatal or perinatal treatment with dex-
71: 39-45. amethasone. J Anim Physiol Anim Nutr (Berl) 2010; 94: 293-306.
[10] Liggins GC, Howie RN. A controlled trial of antepartum glucocor- [35] Sliwa E, Tatara MR, Nowakowski H, et al. Effect of maternal
ticoid treatment for prevention of the respiratory distress syndrome dexamethasone and alpha-ketoglutarate administration on skeletal
in premature infants. Pediatrics 1972; 50: 515-25. development during the last three weeks of prenatal life in pigs. J
[11] Young BK, Klein SA, Katz M, et al. Intravenous dexamethasone Matern Fetal Neonatal Med 2006; 19: 489-93.
for prevention of neonatal respiratory distress: A prospective con- [36] Capdevila J, Izpisua Belmonte JC. Patterning mechanisms control-
trolled study. Am J Obstet Gynecol 1980; 138: 203-9. ling vertebrate limb development. Annu Rev Cell Dev Biol 2001;
[12] Morales WJ, Diebel ND, Lazar AJ, Zadrozny D. The effect of 17: 87-132.
antenatal dexamethasone administration on the prevention of respi- [37] ten Berge D, Brugmann SA, Helms JA, Nusse R. Wnt and FGF
ratory distress syndrome in preterm gestations with premature rup- signals interact to coordinate growth with cell fate specification
ture of membranes. Am J Obstet Gynecol 1986; 154: 591-5. during limb development. Development 2008; 135: 3247-57.
[13] Sloboda DM, Challis JR, Moss TJ, Newnham JP. Synthetic gluco- [38] Litingtung Y, Dahn RD, Li Y, et al. Shh and Gli3 are dispensable
corticoids: antenatal administration and long-term implications. for limb skeleton formation but regulate digit number and identity.
Curr Pharm Des 2005; 11: 1459-72. Nature 2002; 418: 979-83.
[14] Heine VM, Rowitch DH. Hedgehog signaling has a protective [39] Williamson I, Eskeland R, Lettice LA, et al. Anterior-posterior
effect in glucocorticoid-induced mouse neonatal brain injury differences in HoxD chromatin topology in limb development. De-
through an 11betaHSD2-dependent mechanism. J Clin Invest 2009; velopment 2012; 139: 3157-67.
119: 267-77. [40] Duboc V, Logan MP. Pitx1 is necessary for normal initiation of
[15] Bodensteiner JB, Johnsen SD. Cerebellar injury in the extremely hindlimb outgrowth through regulation of Tbx4 expression and
premature infant: newly recognized but relatively common out- shapes hindlimb morphologies via targeted growth control. Devel-
come. J Child Neurol 2005; 20: 139-42. opment 2011; 138: 5301-9.
[16] Allin MP, Salaria S, Nosarti C, et al. Vermis and lateral lobes of [41] Mushtaq T, Farquharson C, Seawright E, Ahmed SF. Glucocorti-
the cerebellum in adolescents born very preterm. Neuroreport coid effects on chondrogenesis, differentiation and apoptosis in the
2005; 16: 1821-4. murine ATDC5 chondrocyte cell line. J Endocrinol 2002; 175: 705-
[17] Heine VM, Griveau A, Chapin C, et al. A small-molecule smooth- 13.
ened agonist prevents glucocorticoid-induced neonatal cerebellar [42] Johnstone B, Hering TM, Caplan AI, et al. In vitro chondrogenesis
injury. Sci Transl Med 2011; 3: 105ra104. of bone marrow-derived mesenchymal progenitor cells. Exp Cell
[18] Gulino A, De Smaele E, Ferretti E. Glucocorticoids and neonatal Res 1998; 238: 265-72.
brain injury: the hedgehog connection. J Clin Invest 2009; 119: [43] Yoo JU, Barthel TS, Nishimura K, et al. The chondrogenic poten-
243-6. tial of human bone-marrow-derived mesenchymal progenitor cells.
[19] Uno H, Lohmiller L, Thieme C, et al. Brain damage induced by J Bone Joint Surg Am 1998; 80: 1745-57.
prenatal exposure to dexamethasone in fetal rhesus macaques. I. [44] Tezval M, Tezval H, Dresing K, et al. Differentiation dependent
Hippocampus. Brain Res Dev Brain Res 1990; 53: 157-67. expression of urocortin's mRNA and peptide in human osteopro-
[20] Cancedda R, Castagnola P, Cancedda FD, et al. Developmental genitor cells: influence of BMP-2, TGF-beta-1 and dexamethasone.
control of chondrogenesis and osteogenesis. Int J Dev Biol 2000; J Mol Histol 2009; 40: 331-41.
44: 707-14. [45] Shintani N, Hunziker EB. Differential effects of dexamethasone on
the chondrogenesis of mesenchymal stromal cells: influence of mi-
8 Current Pharmaceutical Design, 2014, Vol. 20, No. 00 Cheng et al.

croenvironment, tissue origin and growth factor. Eur Cell Mater [67] Muraglia A, Martin I, Cancedda R, Quarto R. A nude mouse model
2011; 22: 302-19; discussion 319-20. for human bone formation in unloaded conditions. Bone 1998; 22:
[46] Kalak R, Zhou H, Street J, et al. Endogenous glucocorticoid signal- 131S-134S.
ling in osteoblasts is necessary to maintain normal bone structure in [68] Walsh S, Jefferiss C, Stewart K, et al. Expression of the develop-
mice. Bone 2009; 45: 61-7. mental markers STRO-1 and alkaline phosphatase in cultures of
[47] Leclerc N, Luppen CA, Ho VV, et al. Gene expression profiling of human marrow stromal cells: regulation by fibroblast growth factor
glucocorticoid-inhibited osteoblasts. J Mol Endocrinol 2004; 33: (FGF)-2 and relationship to the expression of FGF receptors 1-4.
175-93. Bone 2000; 27: 185-95.
[48] James CG, Ulici V, Tuckermann J, et al. Expression profiling of [69] Deng C, Wynshaw-Boris A, Zhou F, et al. Fibroblast growth factor
Dexamethasone-treated primary chondrocytes identifies targets of receptor 3 is a negative regulator of bone growth. Cell 1996; 84:
glucocorticoid signalling in endochondral bone development. BMC 911-21.
Genomics 2007; 8: 205. [70] Naski MC, Wang Q, Xu J, Ornitz DM. Graded activation of fibro-
[49] Ramel MC, Hill CS. Spatial regulation of BMP activity. FEBS Lett blast growth factor receptor 3 by mutations causing achondroplasia
2012; 586: 1929-41. and thanatophoric dysplasia. Nat Genet 1996; 13: 233-7.
[50] Hogan BL. Bone morphogenetic proteins: multifunctional regula- [71] Naski MC, Colvin JS, Coffin JD, Ornitz DM. Repression of hedge-
tors of vertebrate development. Genes Dev 1996; 10: 1580-94. hog signaling and BMP4 expression in growth plate cartilage by fi-
[51] Lai CH, Chen SC, Chiu LH, et al. Effects of low-intensity pulsed broblast growth factor receptor 3. Development 1998; 125: 4977-
ultrasound, dexamethasone/TGF-beta1 and/or BMP-2 on the tran- 88.
scriptional expression of genes in human mesenchymal stem cells: [72] Ohbayashi N, Shibayama M, Kurotaki Y, et al. FGF18 is required
chondrogenic vs. osteogenic differentiation. Ultrasound Med Biol for normal cell proliferation and differentiation during osteogenesis
2010; 36: 1022-33. and chondrogenesis. Genes Dev 2002; 16: 870-9.
[52] Rickard DJ, Sullivan TA, Shenker BJ, et al. Induction of rapid [73] Jheon A, Bansal AK, Zhu B, et al. Characterisation of the constitu-
osteoblast differentiation in rat bone marrow stromal cell cultures tive over-expression of AJ18 in a novel rat stromal bone marrow
by dexamethasone and BMP-2. Dev Biol 1994; 161: 218-28. cell line (D8-SBMC). Arch Oral Biol 2009; 54: 705-16.
[53] Diekman BO, Estes BT, Guilak F. The effects of BMP6 overex- [74] Minina E, Kreschel C, Naski MC, et al. Interaction of FGF,
pression on adipose stem cell chondrogenesis: Interactions with Ihh/Pthlh, and BMP signaling integrates chondrocyte proliferation
dexamethasone and exogenous growth factors. J Biomed Mater Res and hypertrophic differentiation. Dev Cell 2002; 3: 439-49.
A 2010; 93: 994-1003. [75] Mikels AJ, Nusse R. Wnts as ligands: processing, secretion and
[54] Oshina H, Sotome S, Yoshii T, et al. Effects of continuous dex- reception. Oncogene 2006; 25: 7461-8.
amethasone treatment on differentiation capabilities of bone mar- [76] Kalajzic Z, Liu P, Kalajzic I, et al. Directing the expression of a
row-derived mesenchymal cells. Bone 2007; 41: 575-83. green fluorescent protein transgene in differentiated osteoblasts:
[55] Ochi K, Derfoul A, Tuan RS. A predominantly articular cartilage- comparison between rat type I collagen and rat osteocalcin promot-
associated gene, SCRG1, is induced by glucocorticoid and stimu- ers. Bone 2002; 31: 654-60.
lates chondrogenesis in vitro. Osteoarthritis Cartilage 2006; 14: 30- [77] Sher LB, Harrison JR, Adams DJ, Kream BE. Impaired cortical
8. bone acquisition and osteoblast differentiation in mice with os-
[56] Houston B, Thorp BH, Burt DW. Molecular cloning and expression teoblast-targeted disruption of glucocorticoid signaling. Calcif Tis-
of bone morphogenetic protein-7 in the chick epiphyseal growth sue Int 2006; 79: 118-25.
plate. J Mol Endocrinol 1994; 13: 289-301. [78] Zhou H, Mak W, Kalak R, et al. Glucocorticoid-dependent Wnt
[57] Leboy PS, Sullivan TA, Nooreyazdan M, Venezian RA. Rapid signaling by mature osteoblasts is a key regulator of cranial skeletal
chondrocyte maturation by serum-free culture with BMP-2 and development in mice. Development 2009; 136: 427-36.
ascorbic acid. J Cell Biochem 1997; 66: 394-403. [79] Maruyama T, Mirando AJ, Deng CX, Hsu W. The balance of WNT
[58] Minina E, Wenzel HM, Kreschel C, et al. BMP and Ihh/PTHrP and FGF signaling influences mesenchymal stem cell fate during
signaling interact to coordinate chondrocyte proliferation and dif- skeletal development. Sci Signal 2011; 3: ra40.
ferentiation. Development 2001; 128: 4523-34. [80] Zhou H, Mak W, Zheng Y, et al. Osteoblasts directly control line-
[59] Ma X, Zhang X, Jia Y, et al. Dexamethasone induces osteogenesis age commitment of mesenchymal progenitor cells through Wnt
via regulation of hedgehog signalling molecules in rat mesenchy- signaling. J Biol Chem 2008; 283: 1936-45.
mal stem cells. Int Orthop 2013; 37: 1399-404. [81] Derfoul A, Carlberg AL, Tuan RS, Hall DJ. Differential regulation
[60] Kronenberg HM. Developmental regulation of the growth plate. of osteogenic marker gene expression by Wnt-3a in embryonic
Nature 2003; 423: 332-6. mesenchymal multipotential progenitor cells. Differentiation 2004;
[61] Tay AG, Farhadi J, Suetterlin R, et al. Cell yield, proliferation, and 72: 209-23.
postexpansion differentiation capacity of human ear, nasal, and rib [82] Mak W, Shao X, Dunstan CR, et al. Biphasic glucocorticoid-
chondrocytes. Tissue Eng 2004; 10: 762-70. dependent regulation of Wnt expression and its inhibitors in mature
[62] Quarto N, Longaker MT. FGF-2 inhibits osteogenesis in mouse osteoblastic cells. Calcif Tissue Int 2009; 85: 538-45.
adipose tissue-derived stromal cells and sustains their proliferative [83] Wehrenberg WB, Janowski BA, Piering AW, et al. Glucocorti-
and osteogenic potential state. Tissue Eng 2006; 12: 1405-18. coids: potent inhibitors and stimulators of growth hormone secre-
[63] Jakob M, Demarteau O, Schafer D, et al. Specific growth factors tion. Endocrinology 1990; 126: 3200-3.
during the expansion and redifferentiation of adult human articular [84] Swolin D, Brantsing C, Matejka G, Ohlsson C. Cortisol decreases
chondrocytes enhance chondrogenesis and cartilaginous tissue for- IGF-I mRNA levels in human osteoblast-like cells. J Endocrinol
mation in vitro. J Cell Biochem 2001; 81: 368-77. 1996; 149: 397-403.
[64] Quarto R, Campanile G, Cancedda R, Dozin B. Modulation of [85] Klaus G, Jux C, Fernandez P, et al. Suppression of growth plate
commitment, proliferation, and differentiation of chondrogenic chondrocyte proliferation by corticosteroids. Pediatr Nephrol 2000;
cells in defined culture medium. Endocrinology 1997; 138: 4966- 14: 612-5.
76. [86] Mushtaq T, Bijman P, Ahmed SF, Farquharson C. Insulin-like
[65] Liu Y, Wagner DR. Effect of expansion media containing fibro- growth factor-I augments chondrocyte hypertrophy and reverses
blast growth factor-2 and dexamethasone on the chondrogenic po- glucocorticoid-mediated growth retardation in fetal mice metatarsal
tential of human adipose-derived stromal cells. Cell Biol Int 2012; cultures. Endocrinology 2004; 145: 2478-86.
36: 611-5. [87] Walsh S, Jefferiss CM, Stewart K, Beresford JN. IGF-I does not
[66] Lee SY, Lim J, Khang G, et al. Enhanced ex vivo expansion of affect the proliferation or early osteogenic differentiation of human
human adipose tissue-derived mesenchymal stromal cells by fibro- marrow stromal cells. Bone 2003; 33: 80-9.
blast growth factor-2 and dexamethasone. Tissue Eng Part A 2009; [88] Smink JJ, Koedam JA, Koster JG, van Buul-Offers SC. Dex-
15: 2491-9. amethasone-induced growth inhibition of porcine growth plate
chondrocytes is accompanied by changes in levels of IGF axis
components. J Endocrinol 2002; 174: 343-52.

Received: December 11, 2013 Accepted: February 4, 2014

View publication stats

Você também pode gostar