Osteoporosis Nanoparticules

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Life Sciences 147 (2016) 46–58

Contents lists available at ScienceDirect

Life Sciences

journal homepage: www.elsevier.com/locate/lifescie

Natural products for treatment of osteoporosis: The effects and


mechanisms on promoting osteoblast-mediated bone formation
Jing An ⁎, Hao Yang, Qian Zhang, Cuicui Liu, Jingjing Zhao, Lingling Zhang, Bo Chen
Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China

a r t i c l e i n f o a b s t r a c t

Article history: Osteoporosis is a systemic metabolic bone disease characterized by a reduction in bone mass, bone quality, and
Received 31 August 2015 microarchitectural deterioration. An imbalance in bone remodeling that is caused by more osteoclast-mediated
Received in revised form 4 January 2016 bone resorption than osteoblast-mediated bone formation results in such pathologic bone disorder. Traditional
Accepted 13 January 2016
Chinese medicines (TCM) have long been used to prevent and treat osteoporosis and have received extensive at-
Available online 18 January 2016
tentions and researches at home and abroad, because they have fewer adverse reactions and are more suitable for
Chemical compounds studied in this article:
long-term use compared with chemically synthesized medicines. Here, we put the emphasis on osteoblasts, sum-
Icariin (PubChem CID): 5,318,997 marized the detailed research progress on the active compounds derived from TCM with potential anti-
Naringin (PubChem CID): 442,428 osteoporosis effects and their molecular mechanisms on promoting osteoblast-mediated bone formation. It
Genistein (PubChem CID): 5,280,961 could be concluded that TCM with kidney-tonifying, spleen-tonifying, and stasis-removing effects all have the
Puerarin (PubChem CID): 5,281,807 potential effects on treating osteoporosis. The active ingredients derived from TCM that possess effects on pro-
Psoralen (PubChem CID): 6199 moting osteoblasts proliferation and differentiation include flavonoids, glycosides, coumarins, terpenoids
Osthole (PubChem CID): 10,228 (sesquiterpenoids, monoterpenoids, diterpenoids), phenolic acids, phenols and others (tetrameric stilbene, an-
Costunolide (PubChem CID): 5,281,437
thraquinones, diarylheptanoids). And it was confirmed that the bone formation effect induced by the above nat-
Kirenol (PubChem CID): 15,736,732
ural products was regulated by the expressions of bone specific matrix proteins (ALP, BSP, OCN, OPN, COL I),
Vanillic acid (PubChem CID): 8468
Honokiol (PubChem CID): 72,303 transcription factor (Runx2, Cbfa1, Osx), signal pathways (MAPK, BMP), local factors (ROS, NO), OPG/RANKL sys-
tem of osteoblasts and estrogen-like biological activities. All the studies provided theoretical basis for clinical ap-
Keywords: plication, as well as new drug research and development on treating osteoporosis.
Osteoporosis © 2016 Elsevier Inc. All rights reserved.
Osteoblasts
Traditional Chinese medicines
Natural products
Molecular mechanism

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
2. Osteoblasts-mediated bone formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3. Bone specific matrix proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
4. Modulatory effects of natural products on transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
4.1. Transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
4.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5. Modulatory effects of natural products on OPG/RANKL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.1. OPG/RANKL system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49

Abbreviations: ALP, alkaline phosphatase; OCN, osteocalcin (bone gamma-carboxyglutamic acid protein, BGLAP); OPN, osteopontin (“bone-bridging” protein, secreted phosphoprotein
1, SPP1); BSP, bone sialoprotein; COL I, collagen type I; COL1A1, collagen type I, alpha 1; Runx2, runt-related transcription factor 2; Cbfa1, core binding factor α1; Osx, osterix; OPG, oste-
oprotegerin; RANK, receptor activator of NF-κB; RANKL, receptor activator of NF-κB ligand; MSCs, mesenchymal stem cells; BMSCs, bone marrow stromal cells; BMMSCs, bone marrow
mesenchymal stem cells; MAPKs, mitogen-activated protein kinases; ERK, extracellular signal-regulated kinase; JNK, c-Jun N terminal kinase; BMPs, bone morphogenetic proteins;
PKC, protein kinase C; BMD, bone mineral density; ERs, estrogen receptors; EREs, estrogen response elements; ROS, reactive oxygen species; C/EBP-β, CCAAT-enhancer-binding
protein-β; MDA, malondialdehyde; NO, nitric oxide; NOS, nitric oxide synthase; sGC, soluble guanylyl cyclase; cGMP, cyclic guanosine monophosphate; PKG, protein kinase-G; PI3K, phos-
phatidylinositol 3-kinase.
⁎ Corresponding author.
E-mail address: anjing198812@126.com (J. An).

http://dx.doi.org/10.1016/j.lfs.2016.01.024
0024-3205/© 2016 Elsevier Inc. All rights reserved.
J. An et al. / Life Sciences 147 (2016) 46–58 47

6. Modulatory effects of natural products on MAPK pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49


6.1. MAPK signaling pathways. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
6.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
7. Modulatory effects of natural products on BMP pathways. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
7.1. BMP signaling pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
7.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
8. Modulatory effects of natural products on ERs-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
8.1. ERs-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
8.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
9. Modulatory effects of natural products on oxidative stress-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
9.1. Oxidative stress-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
9.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
10. Modulatory effects of natural products on NO-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
10.1. NO-mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
10.2. Natural products from TCM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
11. Conclusions and future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56

1. Introduction The ideal strategy for treating osteoporosis is to inhibit bone resorp-
tion by osteoclasts and/or increase bone formation by osteoblasts.
Bone, a highly mineralized connective tissue, is continuously However, most of the current therapies for treating osteoporosis
broken-down and re-formed in a process of turnover known as bone re- focus on inhibiting bone resorption, and there are only few agents
modeling, which occurs through the interaction and balance between available that promote bone formation. Major treatments currently
bone forming cells, called osteoblasts, and bone resorbing cells, called used for osteoporosis include hormone-replacement therapy (HRT)
osteoclasts [1]. Bone remodeling is regulated by several systemic [13], bisphosphonates [14], calcitonin, selective estrogen receptor
hormones (e.g., parathyroid hormone, 1,25-dihydroxyvitamin D3, sex modulators (SERMs) like raloxifene and droloxifen [15], recombinant
hormones and calcitonin) and local factors (e.g., nitric oxide, prosta- human parathyroid hormone (rhPTH), Vitamin D analogs, and
glandins, growth factors and cytokines) [2,3] This remodeling is respon- ipriflavone [16,17]. The effect of these drugs in increasing bone mass
sible for normal skeletal formation, skeletal functions and mineral or recovering bone loss is relatively minor, probably no more than 2%
homeostasis. Deregulation in one of these processes can lead to bone per year [18]. However, conventional drug therapies have both pros
diseases such as osteoporosis. and cons. For example, while estrogen replacement has produced posi-
Osteoporosis (OP) is a systemic metabolic bone disease that is char- tive results with respect to improved bone mineral density (BMD) and
acterized by microarchitectural deterioration, low bone mass, and in- reduced fracture incidence in early menopause, its prolonged use is re-
creased risk of fractures, which is a prevalent disease that results from stricted because of potential complications such as breast cancer, uter-
an increase in bone breakdown relative to bone formation. The clinical ine bleeding, and cardiovascular events. One concern related to the
complications include fractures, disability and chronic pain. The causes usage of bisphosphates is the complications of osteonecrosis of the
of osteoporosis are very complicated, and the disease itself is closely re- jaw (ONJ). The incidence of ONJ disease seems relatively low in patients
lated to aging, endocrine disorders, calcium malabsorption, and limb receiving oral bisphosphates for osteoporosis or Paget's disease and
disuse, as well as immune, nutritional, and genetic factors [4]. Osteopo- considerably higher in patients with malignancy receiving high doses
rosis falls into two categories: Postmenopausal osteoporosis (Type 1 os- of intravenous bisphosphates [19]. Despite an excellent safety profile
teoporosis) is the most common disease in women after menopause, for parathyroid hormone (PTH), concerns do arise from its persistence
which is linked to an estrogen deficiency. Senile osteoporosis (Type 2 after discontinuation without sequential use of antiresorptive drugs
osteoporosis) is related to aging (N75 years) and results from a deficien- [20]. In addition, PTH is contraindicated for patients at risk for osteosar-
cy in dietary calcium, Vitamin D decline, or increased activity of the coma. The belief that combined use of both types of drugs may have a
parathyroid glands [5]. synergistic effect on BMD is not fully supported by some observational
Osteoporosis is a major global public health problem that is consid- studies [21,22]. To date, most of the effective osteoporosis therapies re-
ered by the World Health Organization (WHO) to be an important duce bone loss but do not restore lost bone mass and strength. It is de-
health issue secondary to coronary heart disease. It can occur at any sirable, therefore, to have satisfactory bone building (anabolic) agents
age and in any racial or ethnic group, affecting millions of people all that stimulate new bone formation and correct the imbalance of trabec-
over the world, especially the aging population [6]. In America, it threats ular microarchitecture characteristic of established osteoporosis, which
an estimated 44 million people, or 55% of the people of 50 years of age would create a new alternative for treating osteoporosis [23].
and older [7]. In Europe, osteoporosis related fractures account for In recent years, there is a growing interest in the treatment of oste-
more disability-adjusted life years (DALYs) lost than common cancers oporosis with plant-based therapies including traditional Chinese med-
except for lung cancer [8]. In China, it is estimated that over 90 million icines (TCM), for which extensive experience has been accumulated
people are suffering from osteoporosis [9]. In addition, about 18% of over thousands of years [24]. TCM have been widely used in clinical
women and 6% of men worldwide will experience hip fragility fractures practice to prevent and treat bone diseases in many countries of the
resulting from osteoporosis [10]. Most cases of osteoporosis occur in world, because they have fewer adverse reactions and are more suitable
postmenopausal women due to the dramatic estrogen levels decline as- for long-term use compared with chemically synthesized medicines.
sociated with menopause. About 40% of postmenopausal women are af- TCM contain numerous chemical constituents, which usually exert
fected by osteoporosis, and with an aging population, this number is their therapeutic effects through multi-pathways and multi-targets,
expected to steadily increase in the near future [11]. The economic bur- these properties are in correspondence with the multi-factorial patho-
den of osteoporosis is markedly increased with the expansion of aging genesis of osteoporosis [25]. Natural products that derived from
world population [12]. TCM have been shown to be excellent and reliable sources for the
48 J. An et al. / Life Sciences 147 (2016) 46–58

development of new drugs, that through a combination of effective sin- bone gamma-carboxyglutamic acid protein, BGLAP) appears on the ter-
gle compounds or in combination with existing commercial drugs, such minal of osteoblastic differentiation. Ducy et al. (1996) indicated that
as estrogen or growth hormone products used to prevent bone loss [26]. OCN can bind to Ca2 + to regulate calcium ion homeostasis and
For centuries, TCM have been used all over the world for the treatment bone mineralization. Contradictorily, bone formation is even increased
of illness and disease, and it will undoubtedly continue to be used as a in OCN-deficient mice [35]. These reports may imply that OCN is a
cost-effective alternative to commercial pharmaceutical products. late osteoblastic marker but doesn't symbolize a degree of bone miner-
There are a number of TCM that possess functions to ease the joints, alization. The expression of BSP and OCN is representative of middle and
strengthen bones and muscles, and have been used for treatment of late stages of osteoblast differentiation phenotypic markers, respective-
bone injuries and bone related diseases, such as osteoporosis. As the ly. Osteopontin (OPN, “bone-bridging” protein, secreted phosphopro-
most common metabolic bone disease, the cause of osteoporosis is an tein 1, SPP1), is one of the more abundant non-collagenous proteins in
imbalance between the formation and resorption of the bone. At the cel- bone matrix that is produced by osteoblasts and osteoclasts. Evidence
lular level, communication and coupling between the main bone-cell has suggested that OPN is a potent stimulator of the osteoclastogenesis
types, the bone-forming osteoblasts and the bone-degrading osteo- and resorptive activity of mature osteoclasts [36]. Although OPN does
clasts, constitute the smallest functional unit on bone metabolism. In not appear to be required for normal development of bones [37], the ab-
this paper, we focused on the osteoblasts, summarized the effect and sence of OPN makes the animal less sensitive to ovariectomy- and
molecular mechanism on bone formation induced by natural products unloading-induced bone loss [38,39]. In addition, collagen type I (COL
derived from TCM. Many studies have shown that natural products I), an extracellular matrix protein, stimulates osteoblast adhesion and
can improve osteoblasts proliferation and differentiation by regulating differentiation [40]. The expressions of all these differentiation-related
the expression of bone specific matrix proteins, transcription factor, cy- genes can be regulated by several local and systemic factors, including
tokines, signal pathways, OPG/RANKL system and hormone-like biolog- estrogen, growth factors and cytokines [41].
ical activities. This paper will provide the readers with the detailed
research progress on the active compounds from TCM with potential
of anti-osteoporosis effect and the pharmacological mechanism on pro- 4. Modulatory effects of natural products on transcription factors
moting osteoblasts-mediated bone formation from the above aspects.
4.1. Transcription factors
2. Osteoblasts-mediated bone formation
Osteogenic differentiation of mesenchymal pluripotent cells is regu-
Osteoblast cells play an important role in the bone formation, which lated by various transcriptional factors, such as Runt-related transcrip-
is composed of a three-step process: (i) proliferation—the increase of tion factor 2/core binding factor α1 (Runx2/Cbfa1) and osterix (Osx).
pre-osteoblast number, (ii) differentiation—the process by which a Runx2/Cbfa1 and Osx are crucial for the regulation of genes responsible
pre-osteoblast becomes a mature osteoblast, and (iii) matrix for the production of bone specific matrix proteins, including ALP, COL I,
mineralization—the formation of new bone matrix by the mature oste- BSP, OCN, OPN and eventually stimulate mineralization of bone nodules
oblasts [27]. [42].
The transcriptional factor, core binding factor α1 (Cbfa1), is essential
3. Bone specific matrix proteins for bone formation during embryogenesis and is a key mediator of bone
differentiation. Recent studies have shown that the expression of Cbfa1
The osteoblast phenotype is specifically acquired in two stages. In correlates with the transition of osteoblasts from the proliferative to the
the first stage, cells proliferate and the matrix matures. Specific proteins differentiated phenotype [43]. However, the over-expression of Cbfa1 in
associated with the bone cell phenotype, such as ALP, can be detected cells of the osteoblastic lineage does not necessarily induce a substantial
during cell proliferation and matrix maturation from the period increase in bone formation in the adult skeleton, but does express a pos-
of 10–15 days. In the second stage of osteoblast phenotype itive effect on osteoclast differentiation in vitro and can dramatically en-
acquisition, the matrix becomes mineralized and late bone markers, hance bone resorption in vivo through increased RANKL expression
such as osteocalcin (OCN), are produced after 10–15 days and up to [44].
25–30 days. Finally, multiple anabolic signaling pathways are positively Runx2 (Runt-related transcription factor 2) is the central control
involved in controlling bone formation, such as the BMP, Wnt, and gene of the osteoblast phenotype. Runx2 is an important osteogenic
Runx2 pathways [28]. transcription factor that can bind to the osteoblast-specific cis-acting el-
Alkaline phosphatase (ALP) activity is the most widely recognized ement (OSE) 2 in the promoter region of osteogenic genes [45]. The im-
biochemical marker for osteoblastic activity. It is the typical protein portance of Runx2 in osteoblast differentiation has been highlighted by
product of an osteoblast phenotype and of osteoblast differentiation. the evidence that disruption of one copy of the Runx2 gene can result in
Composed of homodimeric metalloenzymes which contain zinc at the cleidocranial dysplasia and that Runx2 gene knockout in mice prevents
active site [29], ALP hydrolyzes a variety of phosphate compounds, osteoblast development [46].
such as ATP, releasing the products, specifically inorganic phosphate Osterix (Osx, also known as Sp7) is a novel zinc finger-containing
(Pi), into the extracellular matrix [30]. As a result, ALP is associated osteoblast-specific transcription factor that is essential for osteoblast
with Pi homeostasis. Also containing other Ca-dependent proteins, the proliferation, differentiation and bone formation. Osx proteins regulate
matrix vesicle uses the Pi to initiate crystal nucleation of extracellular the expression of many specific osteoblast differentiation markers in-
matrix calcium deposits, leading to the formation of hydroxyapatite cluding Runx2 and Osteonectin (ON). Genetic studies have indicated
[31]. In general, the appearance of ALP activity is represented as an that Osx acts as a downstream gene of Runx2 in the osteoblast differen-
early phenotypic marker for osteoblastogenesis [32]. Wennberg et al. tiation signaling pathways [47].
have demonstrated that osteoblasts obtained from ALP-deficient mice Mesenchymal stem cells differentiate into osteoblasts, adipocytes,
differentiate normally but can't form mineralized nodules in vitro [33], chondroblasts, myoblasts, and fibroblasts [48]. The nuclear receptor
supporting the crucial role of ALP in this particular process. PPAR-γ is the dominant regulator of adipogenesis, and CCAAT-
A complicated network of various proteins is involved in the regula- enhancer-binding protein-β (C/EBP-β) is the primary transcription
tion of osteogenesis. Bone sialoprotein (BSP) appears following ALP ex- factor activated upon adipogenic induction. Activation of PPAR-γ and
pression and localizes to the matrix of mineralization. BSP can promote C/EBP-β favors differentiation of mesenchymal stem cells to adipocytes
the nucleation of hydroxyapatite mineralization in vitro and increases rather than to osteoblasts. In addition, PPAR-γ insufficiency leads to in-
calcium incorporation and nodule formation [34]. Osteocalcin (OCN, creased osteoblast differentiation and higher trabecular bone volume
J. An et al. / Life Sciences 147 (2016) 46–58 49

[49]. Thus, inhibition of adipocyte differentiation might induce mesen- rat osteoblast-like UMR 106 cells [61]. Kirenol is a natural diterpenoid
chymal stem cells to form osteoblasts. compound isolated from Herba Siegesbeckia, including S. orientalis,
S. glabrescens, and S. pubescens. Kim's study demonstrated that kirenol
4.2. Natural products from TCM not only increased the expression of osteoblast differentiation markers,
such as ALP, COL I, and OPN, but also increased the expression of OPG/
Icariin, the main active flavonoid glucoside isolated from Epimedii RANKL ratio [62]. Studies have shown that honokiol [63], apocynin
Folium. Icariin treatment (0.1–100 nM) significantly increased COL I [64], puerarin [65] icariin [66], baohuoside-I, epimedin B, and
and ALP mRNA expression of osteoblastic MC3T3-E1 cells in a dose- sagittatoside A [67] significantly increased OPG release and decreased
dependent manner until 10 nM concentration whereas OPN expression the production of RANKL (P b 0.05).
was marginally but significantly increased from 1 nM concentration on-
ward [50]. Naringin, dihydro-flavonoid, the main effective compound of 6. Modulatory effects of natural products on MAPK pathways
Drynariae Rhizoma. Neoeriocitrin, dihydro-flavonoid, a new compound
isolated from Drynariae Rhizoma. Neoeriocitrin more significantly im- 6.1. MAPK signaling pathways
proved proliferation and ALP activity as well as up-regulated Runx2,
COL I and OCN expression in MC3T3-E1 cells by 56%, 37% and 14% re- The mitogen-activated protein kinases (MAPKs) are the family of
spectively than naringin [51]. Poncirin, flavanone glycoside, isolated secondary messengers that convey signals from the cell surface to the
from the fruit of Poncirus trifoliata. Poncirin inhibited cytoplasm lipid nucleus in response to a wide range of stimuli, including hormones,
droplet accumulation and PPAR-γ and C/EBP-β mRNA expression in chemicals and stress. Three major families of MAPKs are extracellular
the murine pluripotent mesenchymal cell line C3H10T1/2 cells. By con- signal-regulated kinase (ERK), p38 kinase, and c-Jun N terminal kinase
trast, poncirin enhanced the expression of Runx2 and transcriptional co- (JNK) and each of these consists of their own subfamilies. MAPKs in-
activator with PDZ-binding motif (TAZ). It also enhanced ALP and OCN volve in the regulation of many cellular physiological functions such
mRNA expression, and increased mineral nodule formation in primary as proliferation, differentiation, inflammation, and apoptosis. Among
bone marrow mesenchymal stem cells (BMMSCs) [52]. Quercetin and numerous transcription factors activated by MAPKs signaling, well rec-
Rutin are common flavonoids in fruits and vegetables. Both of them ognized factors include c-fos, c-Jun, AP-1, CREB and NFAT. Many of these
were found to up-regulate the osteoblast differentiation of BMMSCs in transcription factors are directly involved in cellular proliferation in nu-
dose dependent manner. Quercetin and rutin increased ALP activity by merous cell types [68].
about 150 and 240% and demonstrated mineralization up to 110 and Osteoblasts and adipocytes differentiate from a common precursor,
200% respectively as compared to control. Further, both of them were the pluripotent mesenchymal stem cells in bone marrow and regulation
also found to increase the expression of OPN, Osx, Runx-2, OPG and of PPARγ2 activity has been shown to control the fate of these cells to-
OCN [53]. Satue et al. reported that the certain doses of quercitrin and wards osteogenesis or adipogenesis. Suppression of PPARγ2 activity is
taxifolin induced BSP and OCN mRNA expression, and decreased associated with enhanced osteogenesis [69]. It has been reported that
RANKL gene expression in MC3T3-E1 cells [54]. Sweroside is a bioactive ERK can directly result in the phosphorylation of PPARγ2 and reduces
herbal ingredient isolated from Fructus Corni. Sweroside significantly in- the levels of PPARγ2 [70]. p38 MAPK plays a negative role in regulating
creased the proliferation of human osteoblast-like MG-63 cells and rat PPARγ2 transcriptional activities, inhibition or disruption of p38 leads
osteoblasts. Sweroside increased the ALP activity and OCN expression. to increased PPARγ2 expression and transactivation [71].
It also attenuated and inhibited apoptosis [55]. Recently, many studies discussed the relationship between MAPK
family and Runx2/Osx. For instance, phosphorylation of p38 MAPK
5. Modulatory effects of natural products on OPG/RANKL induced the activation of Runx2 via TAK1 and MEK3 signal pathway
during early osteoblastic differentiation [72]. Furthermore, ERK1/2 me-
5.1. OPG/RANKL system diated Runx2 phosphorylation and transcriptional activity in bone [73].
Previous studies also reported that p38 MAPK regulated osteoblastic dif-
Bone formation/resorption balance is governed by a wide variety of ferentiation through Osx [74].
cell signals, hormones, and growth factors that are essentially regulated
by the receptor activator of NF-κB ligand (RANKL) and its two receptors, 6.2. Natural products from TCM
receptor activator of NF-κB (RANK) and osteoprotegerin (OPG) [56,57].
RANKL, a member of the TNF ligand family [57], is highly expressed on Icariin enhanced MC3T3-E1 cell differentiation and mineralization.
the surface of bone marrow stromal cells (BMSCs), preosteoblasts and Icariin treatment rapidly induced ERK and JNK activation but showed
T cells. When RANKL binds to RANK, osteoclast differentiation and func- no effect on activation of p38 kinase. Furthermore, Icariin-mediated ef-
tion are enhanced [58]. OPG, produced by BMSCs and osteoblasts, is a fects on osteoblasts were dramatically attenuated by treatment with
soluble decoy receptor for RANKL and prevents RANKL from binding specific inhibitors of MAPKs, U0126 (ERK inhibitor) and SP600125
to, and activating, RANK on the surface of osteoclastic lineage cells [56, (JNK inhibitor) [50]. Ugonin K is a flavonoid isolated from the roots
57]. The relative ratio of RANKL to OPG is considered the critical deter- of Helminthostachys zeylanica. Ugonin K significantly increased the ALP
minant and final step in the regulation of osteoclast biology and bone activity, expressions of BSP, OCN, and mineralization of MC3T3-E1 oste-
resorption, and this ratio is modulated by various osteotropic hormones, oblasts. The mRNA expressions of Runx2 and Osx were also up-
cytokines, and drugs [59]. regulated. Both p38 and ERK participated in regulating ugonin K evoked
osteogenesis but p38 seemed to play a more important role [75].
5.2. Natural products from TCM Neobavaisoflavone (NBIF) is an isoflavone isolated from Psoralea
corylifolia. NBIF concentration-dependently promoted osteogenesis in
Echinacoside (ECH), a phenylethanoid glycosides isolated from MC3T3-E1 cells. NBIF increased the phosphorylated level of p38. Appli-
Cistanches Herba. ECH caused a significant increase in cell proliferation, cation of p38 inhibitor SB203580 repressed NBIF-induced activation of
ALP activity, COL I contents, OCN levels and an enhancement of miner- ALP, the expression of COL I, OCN and BSP, and the matrix proteins min-
alization in osteoblastic MC3T3-E1 cells at the concentration range eralization, as well as abolished the stimulatory effect of NBIF on the ex-
from 0.01 to 10 nmol L−1. In addition, ECH enhanced the ratio of OPG/ pression of Runx2 and Osx [76]. Salvianolic acid B (Sal B), a phenolic acid
RANKL [60]. Vanillic acid, a phenolic acid isolated from Sambucus isolated from Salvia miltiorrhiza. Xu's study indicated that Sal B promot-
williamsii Hance, could stimulate proliferation, ALP activity as well as in- ed osteogenesis of human mesenchymal stem cells (hMSCs) through
creased Runx2, OCN and the ratio of OPG/RANKL mRNA expressions in activating ERK signaling pathway. Sal B supplementation (5 μM)
50 J. An et al. / Life Sciences 147 (2016) 46–58

increased the ALP activity, OPN, Runx2 and Osx expression and promot- subsequently regulating Cbfa1/Runx2, OPG/RANKL gene expressions.
ed mineralization in hMSCs. Sal B promoted the phosphorylation of Concurrent treatment involving the BMP antagonist (Noggin) or the
ERK1/2, which in turn up-regulated the expression of Runx2, and when NOS inhibitor (L-NAME) diminished the icariin-induced osteoblasts pro-
the ERK signaling pathway was blocked, the anabolic effects of Sal B liferation, ALP activity, NO production, as well as the BMP-2, SMAD4,
were diminished [77]. Costunolide, one of the active sesquiterpene lac- Cbfa1/Runx2, OPG, RANKL gene expressions [66]. Liang's research indi-
tones isolated from the roots of Saussurea lappa. Lee et al. reported that cated that icariin can modulate bone formation process via the BMP-2/
the effect of costunolide in increasing cell growth might be partly medi- SMAD4 signal transduction pathway in human osteoblastic hFOB 1.19
ated from estrogen receptor (ER), PI3K, ERK, protein kinase C (PKC) and cells [89]. Naringin, a polymethoxylated flavonoid, was shown to in-
mitochondrial ATP-sensitive K + channel; the effect on ALP activity crease BMP-2 expression and enhance osteogenic response via the
might be mediated from ER, ERK, p38, and PKC; the effect on collagen PI3K, Akt, c-Fos/c-Jun and AP-1-dependent signaling pathway. Naringin
synthesis might be mediated from PI3K, ERK, p38, JNK, and PKC; and enhanced ALP activity, OCN level, OPN synthesis and cell proliferation
the induction of mineralization by costunolide is associated with in- in primary cultured osteoblasts. Naringin mediated the transcriptional
creased activation of ER and PI3K [78]. Imperatorin and Bergapten, two regulation of BMP-2 via phosphorylation of Akt and activation of the ac-
furanocoumarins, isolated from Cnidium monnieri. Both of them en- tivator protein-1 (AP-1) components c-Fos and c-Jun. It also enhanced
hanced the phosphorylation of SMAD (transcription factors activated the binding of c-Fos and c-Jun to the AP-1 element on the BMP-2 pro-
by TGF-β) 1/5/8, p38 and ERK. Pretreatment of osteoblasts with p38 in- moter. Transfection with dominant-negative mutant of p85 and Akt or
hibitor (SB203580) or MAPK inhibitor (PD98059) or transfected with c-Fos and c-Jun antisense oligonucleotide inhibited the potentiating ac-
dominant negative mutant of p38 or ERK antagonized the elevation of tion of naringin on BMP-2 production [90]. Psoralen, a coumarin-like de-
BMP-2 expression and ALP activity induced by imperatorin and rivative extracted from Psoralea corylifolia. Psoralen up-regulated the
bergapten [79]. Kobophenol A, a tetrameric stilbene, is one of the major expression of BMP-2 and BMP-4 genes, increased the protein level of
active compounds from Caragana sinica Rhed. Kwak's study demonstrat- phospho-SMAD1/5/8, and activated BMP reporter (12xSBE-OC-Luc) ac-
ed that Kobophenol A enhances proliferation and ALP activity of human tivity in a dose-dependent manner, as well as enhanced the expression
osteoblast-like cells with activation of the p38 pathway [80]. of Osx, the direct target gene of BMP signaling. Deletion of the BMP-2
and BMP-4 genes abolished the stimulatory effect of psoralen on the ex-
7. Modulatory effects of natural products on BMP pathways pression of COL I, ALP, OCN and BSP in primary mouse calvarial osteo-
blasts [91]. Osthole, a coumarin-like derivative extracted from Fructus
7.1. BMP signaling pathways Cnidii. Osthole activated Wnt/β-catenin signaling, increased BMP-2 ex-
pression, and stimulated osteoblast differentiation. Targeted deletion of
Bone morphogenetic proteins (BMPs), with more than 20 members, the β-catenin and BMP-2 genes abolished the stimulatory effect of
belong to the TGF-β superfamily and are originally identified by their osthole on osteoblast differentiation [92]. Imperatorin and bergapten,
unique ability to induce ectopic cartilage and bone formation in vivo. two coumarin derivatives, were shown to enhance ALP activity, COL I
BMPs play important roles in bone formation and bone cell differentia- synthesis and bone nodule formation in primary cultured osteoblasts.
tion by stimulating ALP activity and synthesis of proteoglycan, collagen Both imperatorin and bergapten increased mRNA levels of BMP-2, and
type I, fibronectin and osteocalcin [2]. The action of BMPs is mediated by the BMP-2 antagonist noggin attenuated the increase of ALP activity
heterotetrameric serine/threonine kinase receptors and the down- [79]. Kim's study demonstrated that kirenol is capable of promoting os-
stream transcription factors SMAD1/5/8 [81]. Once these transcription teoblast differentiation in MC3T3-E1 cells through activation of the
factors are phosphorylated on serine residues, they form a complex BMP and Wnt/β-catenin signaling pathways. Kirenol treatment activated
with a common mediator, SMAD4, and the complex is translocated the mRNA expression of BMP-2, Runx2, and Osx in MC3T3-E1 cells, as
into the nucleus to activate the transcription of target genes and then well as up-regulated the mRNA expression of genes in the Wnt/β-
stimulates bone formation [82]. A genetic study using a combined link- catenin signaling pathway related regulators, including LRP5, DVL2, β-
age and association analyses has linked the BMP-2 gene to a phenotype catenin, cyclin D1, and phosphorylated GSK3β [62]. Emodin is a naturally
of low bone mineral density (BMD) and high fracture risk [83]. Several occurring anthraquinone present in the roots and bark of numerous
lines of evidence have demonstrated that BMP-2 induces or promotes plants of the genus Rhamnus. Lee et al. reported that the low concentra-
the expression of Runx2 and Osx [84], which are essential transcription tion (5 μM) of emodin highly induced the mRNA expression of BMP-2 in
factors for osteoblast differentiation and bone formation [47]. MC3T3-E1 cells. The up-regulation of BMP-2 could be mediated through
Recent studies have demonstrated that BMP expression is activated the activation of both Akt and MAPK by activating PI3K, because those in-
by canonical Wnt/β-catenin signaling in osteoblast cells and in bone ductions by emodin were completely inhibited by the PI3K inhibitor,
marrow stromal cells. It has indicated that the Wnt/β-catenin pathway LY294002 [93]. Salidroside (SAL), the glucoside-containing component
plays an important role in bone biology, particularly in bone mass acqui- of Rhodiola rosea L. SAL increased the mRNA expressions of ALP, OPN,
sition, remodeling, differentiation, and maintenance [85]. Cyclin D1, a Runx2 and Osx in either C3H10T1/2 or MC3T3-E1 cells. SAL also in-
target gene of Wnt pathway, was reported to be up-regulated during creased the mRNA level of BMP-2, BMP-6 and BMP-7 and enhanced
Wnt/β-catenin activation [86]. the phosphorylation of SMAD 1/5/8 and ERK1/2. The osteogenic effect
In addition, Ras activation has been shown to integrate the PI3K sig- of SAL was abolished by BMP antagonist noggin or by BMP receptor ki-
nal to Akt and MAPK for BMP-2 expression in osteoblast differentiation, nase inhibitor dorsomorphin [94]. Acerogenin A, a diarylheptanoids iso-
Ras is immediately activated by stimulus and then it subsequently in- lated from Acer nikoense Maxim. Acerogenin A treatment increased BMP-
duces the activation of PI3K resulting in the activation of Akt and 2, BMP-4, and BMP-7 mRNA expression levels in MC3T3-E1 cells. Noggin
MAPK (especially ERK) that are required for BMP-2 expression [87]. (a BMP antagonist) inhibited the acerogenin A-induced increase in the
The mechanism by which Akt regulates BMP-2 transcription is not OCN, Osx and Runx2 mRNA expression levels [95].
clear, but one possibility may include the activation of NF-κB that has
been shown to regulate BMP-2 gene expression in chondrocytes [88]. 8. Modulatory effects of natural products on ERs-mediated
pathways
7.2. Natural products from TCM
8.1. ERs-mediated pathways
Icariin has been reported to enhance bone healing and reduce osteo-
porosis occurrence. Hsieh's study demonstrated that icariin may exert its Estrogens exert their physiological effects on target tissues by
osteogenic effects through the induction of BMP-2 and NO synthesis, interacting with estrogen receptors, which are members of the
J. An et al. / Life Sciences 147 (2016) 46–58 51

superfamily of ligand regulated nuclear transcription factors. Estrogen induced increases in ALP activity, expressions of BSP, OCN, Runx2 and
receptors (ERs) are a family of intracellular receptors, including estro- Osx, and subsequent bone nodule formation in MC3T3-E1 cells were
gen receptor α (ERα) and estrogen receptor β (ERβ) [96]. Both recep- inhibited by ICI 182,780. The raised c-Src phosphorylated level induced
tors have been identified in osteoblasts and osteoclasts as well as in by ugonin K was significantly attenuated by ICI 182,780. Application of
their precursors. Estrogens play an important role in coordinating activ- PP2 (c-Src specific inhibitor) repressed ugonin K-induced osteogenesis.
ities of osteoblasts and osteoclasts in bone homeostasis and preventing Ugonin K also increased the nuclear level of ERα protein. The results
postmenopausal bone loss [97]. demonstrated that ugonin K stimulated osteogenesis might act through
ERs stimulate classical effects by acting as nuclear transcription fac- an ER dependent activation of a non-classical signaling pathway medi-
tors as well as non-classical effects by activating distinct cytoplasmic ated by phosphorylation of c-Src. Moreover, a transactivation potential
protein kinase cascades. Typically, the majority of ER α and β are towards ERα through a classical pathway might not be precluded
found in the cytoplasm and nucleus, although small amounts are [105]. Genistein enhanced the phosphorylation of ERK 1/2, p38 MAPK
expressed on cell membrane. In the classical pathway, ERs are activated and JNK 1/2 in MC3T3-E1 cells. Sequentially, it increased the transloca-
by directly binding to estrogen and subsequently by stimulating gene tion of NF-κB and c-Jun from the cytoplasm to the nucleus. Genistein in-
transcription via interacting with estrogen response elements (EREs) creased ERα mRNA expression and the amounts of cytosolic and nuclear
in the promoters of target genes [98]. Other transcription factors in nu- ERα in MC3T3-E1 cells and rat calvarial osteoblasts. And there are
cleus, that include NF-κB, AP-1 and SP-1, interact with the ligand–recep- several ERα-specific DNA-binding elements in the 5′-promoter regions
tor complex to influence gene transcription [99]. Such nuclear canonical of the BMP-6, COL I and OCN genes. The results indicated that genistein
pathway mediates the long term actions of estrogen. Alternatively, es- can induce ERα gene expression via the activation of MAPK/NF-κB/AP-1
trogen can induce rapid effects through membrane-bound ERs. ERs and accordingly stimulates differentiation-related gene expressions and
can be activated by MAPK via phosphorylation upon growth factor re- osteoblast mineralisation [106]. Puerarin, the main isoflavone glycoside
ceptors activation in a ligand-independent manner. MAPK signaling found in the radix of Pueraria lobata (Willd.) Ohwi. Wang's study
pathways involve the activation of membrane-associated receptor tyro- showed that treatment with ICI 182,780 markedly abolished puerarin-
sine kinases (e.g., IGF-IR and EGFR), Ras protein kinase, Raf mitogen- increased proliferation, ALP activity and COL I secretion in human oste-
activated protein kinase kinase (MEK/MAPKK) as well as downstream oblastic MG-63 cells (P b 0.05). In addition, protective effect of puerarin
extracellular signal-regulated protein kinase (ERK). [100]. upon cisplatin-induced apoptosis was also significantly blocked by ICI
ERα has been reported to be the major regulator mediating the ef- 182,780 (P b 0.05). Using siRNA technology, it was demonstrated that
fects of estrogen on bone metabolism. According to the classical mech- the effects of puerarin on proliferation, differentiation as well as
anism, estrogen binds to ERα to form a receptor dimer, and then this cisplatin-induced cell death are mediated by both ERα and ERβ [107].
dimer is translocated from the cytoplasm to the nucleus. After binding Tiyasatkulkovit's study also showed that after exposure to ICI 182,780,
to its specific ERE, this ER-estrogen complex can initiate certain gene the puerarin-induced upregulation of ALP expression in UMR106 cells
expressions and control osteoblast differentiation [101]. Serine 118 is was completely abolished [65]. Xiao et al. reported that co-treatment
reported to be the major phosphorylation site in ERα for ligand- of UMR 106 cells with ICI 182,780 abolished the stimulatory effects of
independent activation by the MAPK-mediated pathway. [100]. Multi- vanillic acid (VA) on osteoblast proliferation and ALP activity, but VA
ple lines of evidence suggest that activation of the tyrosine kinase c- failed to bind to ERα or ERβ and did not activate ERE-luciferase activity
Src represents one of the initial steps in ERα-mediated cell signaling via ERα or ERβ in UMR 106 cells. In contrast, VA induced the phosphor-
[102]. ylation of MEK 1/2, ERK 1/2 and ERα at serine 118 residue in UMR 106
Estrogen has an important role in the development and growth of cells. The results indicated that the bone sparing effects of VA involved a
bones and later in the maintenance of bone mass. It is believed that ligand independent, ERE-independent and MAPK-mediated
the major action of estrogen on the skeleton in vivo is through the inhi- nongenomic ER signaling pathway [61]. Resveratrol (RSVL), a polyphe-
bition of bone resorption. Although some of the anti-resorptive effects nol mainly existing in red grapes and berries. Dai's study suggested that
of estrogen are via direct actions on bone-resorbing osteoclasts, estro- treatment of human BMMSCs with ICI 182,780 attenuated the stimula-
gen has also been shown to have indirect effects by regulating bone- tory effects of RSVL on cell growth, ALP activity, calcium deposition, and
forming osteoblasts and bone marrow stromal cells. As one of these ef- the mRNA expressions of Runx2/Cbfa1, Osx and OCN, associated with
fects, 17β-estradiol has been suggested to protect osteoblasts from apo- suppression of ERK 1/2 phosphorylation [108].
ptosis. Estrogen has been shown to protect calvaria-derived osteoblastic
cells from etoposide-induced apoptosis as well as to protect osteoblasts 9. Modulatory effects of natural products on oxidative
from ethanol-induced bone loss [103,104]. stress-mediated pathways

8.2. Natural products from TCM 9.1. Oxidative stress-mediated pathways

Icariin, baohuoside-I, epimedin B and sagittatoside A are four major Oxidative stress, which is caused by excessive production of reactive
flavonoids isolated from Herba epimedii (HEP). Xiao's study showed oxygen species (ROS) and insufficient antioxidant defense mechanisms,
that all the four compounds significantly stimulated the cell prolifera- has been shown to be a crucial pathogenic factor of osteoporosis [109].
tion rate, ALP activity and OPG/RANKL mRNA expression in rat Osteoporotic postmenopausal women exhibit a decrease in bone miner-
osteoblastic-like UMR-106 cells and their effects could be abolished by al density (BMD) that is related to higher oxidation of plasma lipids and
co-incubation with 10−6 M ICI 182,780 (ER antagonist). None of the fla- lower superoxide dismutase and catalase efficacies. In rat femurs, ovari-
vonoids exhibited binding affinities towards ERα and ERβ. However, ectomy (OVX) results in oxidative damage and decreases the capacity of
sagittatoside A selectively activated ERE-luciferase activity via ERα. In antioxidant defense mechanisms. Thus, oxidative stress might serve as a
addition, icariin and sagittatoside A induced ERα phosphorylation at major contributor to postmenopausal osteoporosis [110]. Both aging
serine 118 residue. The results indicated that all four flavonoids from and estrogen deficiency elevate the levels of ROS, and there is strong ev-
HEP stimulated ER-dependent osteoblastic functions in UMR-106 cells, idence to indicate that the damage on bone mass due to estrogen loss
but only icariin and sagittatoside A appeared to exert their actions by might be prevented by antioxidants.
ligand-independent activation of ERα [67]. Song's study suggested Oxidative stress, results from deregulation of the intracellular reduc-
that treatment of osteoblasts with ICI 182,780 attenuated the stimulato- tion–oxidation balance, which is mainly manifested by superfluous su-
ry effects of icariin on COL I, ALP and OPN mRNA expressions, associated peroxide radical anion (O2−), H2O2 and the hydroxyl radical (OH.) in
with suppression of ERK and JNK phosphorylation [50]. Ugonin K cells. ROS, mostly derived from mitochondrial activity. A certain amount
52 J. An et al. / Life Sciences 147 (2016) 46–58

of oxidative stress is required by the mitochondrial respiratory chain generator [63]. Apocynin is a naturally occurring methoxy-substituted
during the metabolic process, however, an over-generation of ROS catechol, experimentally used as an inhibitor of NADPH-oxidase.
would cause lipid and protein oxidation and alter mitochondrial Apocynin could protect osteoblastic MC3T3-E1 cells from mitochondrial
and nuclear DNA integrity, which would lead to tissue damage. Under dysfunction-induced toxicity. Apocynin significantly (P b 0.05) in-
certain pathological conditions, the dynamic balance between the gen- creased cell survival, calcium deposition, and OPG release and decreased
eration and elimination of ROS may be broken and thus the intracellular the production of ROS and osteoclast differentiation inducing factors
ROS levels increase significantly. It can be suggested that ROS produc- such as TNF-α, IL-6, and RANKL in the presence of antimycin A [64].
tion is particularly involved in mineral tissue homeostasis and contrib- Ginsenoside-Rb2 (Rb2), a 20(S)-protopanaxadiol glycoside extracted
utes mostly to bone remodeling by reducing bone formation and from ginseng, is a potent antioxidant. Huang's study indicated that the
promoting bone resorption [111,112]. potential anti-osteoporosis effects of Rb2 were linked to a reduction of
Mitochondria not only produce ROS but are also the principal target oxidative damage and bone-resorbing cytokines. Rb2 (0.1 to 10 μM)
of ROS attacks. Impaired mitochondrial function can lead to increased promoted the proliferation, ALP activity, calcium mineralization and
ROS generation and may increase oxidative stress if the antioxidant de- mRNA expressions of ALP, COL1A1, OCN and OPN against oxidative
fense mechanisms of the cells are overwhelmed. Increased oxidative damage induced by H2O2 in MC3T3-E1 cells. Importantly, Rb2 reduced
stress caused by mitochondrial dysfunction is considered the causal the expression levels of RANKL and IL-6 and inhibited the H2O2-
link between elevated ROS and the major biochemical pathways postu- induced production of ROS. The in vivo study indicated that the Rb2 ad-
lated to be involved in the pathogenesis of senile disorders. There are ministered for 12 weeks partially decreased blood MDA activity and el-
two major sites of superoxide release in the respiratory chain, the first evated the GSH activity in ovariectomized (OVX) mice [117].
being complex I, the second complex III. Antimycin A (AMA) is an inhib- Ophiopogonin D (OP-D), the chief active component isolated from
itor of mitochondrial electron transport via its binding to complex III, Radix Ophiopogon japonicus. OP-D increased osteogenic differentiation
has been used as a ROS generator in biological systems [113]. by promoting calcium mineralization, ALP activity of MC3T3-E1 cells
Hydrogen peroxide (H2O2) is favorable to serve as both an extra- and improved the mRNA expressions of OCN, COL1A1 and OPN. OP-D
and an intercellular signal molecule. H2O2 is generated from nearly suppressed ROS generation in both MC3T3-E1 and RAW264.7 cells. Fur-
all sources of oxidative stress. Because of its long half-life and the ther research showed that the protective effects of OP-D against osteo-
ability to cross biological membranes, H2O2 has the characteristics porosis are linked to a reduction in oxidative stress via the FoxO3a-β-
most suited to act both as an intra- and an intercellular signal. It catenin signaling pathway [118]. In addition, studies have shown that
has been demonstrated that it plays a crucial role in postmenopausal treatment of osteoblast cells with kobophenol A resulted in improve-
osteoporosis. ment of ROS scavenging activity [80].

9.2. Natural products from TCM


10. Modulatory effects of natural products on
NO-mediated pathways
2,3,5,40-Tetrahydroxystil bene-2-O-b-D-glucoside (TSG), a
polyhydrostilbenes derived from Polygonum multiflorum Thunb. TSG
10.1. NO-mediated pathways
might be effective in providing protection against osteoporosis associat-
ed with oxidative stress. TSG significantly increased cell survival, ALP ac-
Nitric oxide (NO) is a short-lived signaling molecule generated from
tivity, calcium deposition, and the mRNA expressions of ALP, COL I and
L-arginine by nitric oxide synthase (NOS) isoenzymes. An accumulating
OCN in the presence of H2O2 (P b 0.05). In addition, TSG decreased the
body of evidence has emerged to suggest that NO plays an important
production of RANKL, IL-6, intracellular ROS and malondialdehyde
role as a paracrine and autocrine mediator of bone metabolism and
(MDA) of osteoblastic MC3T3-E1 cells induced by H2O2 [114].
bone cell activity in response to diverse stimuli [119]. The NO–sGC–
Gastrodin, a glucoside isolated from Gastrodia elata, is a potent antioxi-
cGMP–PKG pathway, begins with activation of NOS, the enzyme that
dant. Gastrodin inhibited H2O2-induced cytotoxicity in human BMMSCs,
synthesizes NO in cells. The increased NO levels activate the soluble
attenuated H2O2-induced hBMMSC dysfunction, reduced lipid genera-
guanylyl cyclase (sGC), thus increasing the level of cyclic guanosine
tion and inhibited hBMMSC adipogenic differentiation under oxidative
monophosphate (cGMP) which acts to activate protein kinase-G
conditions. Gastrodin displayed good recovery effects on cellular ALP
(PKG), a cGMP dependent protein kinase [120].
activity, mRNA expressions of ALP, OCN, COL I, OPN and calcium miner-
Serine/threonine protein kinase Akt, also known as protein kinase B,
alization which was suppressed by H2O2. Gastrodin showed protective
is stimulated by a number of receptor tyrosine kinases and G protein-
effects against osteoporosis linking to a reduction in ROS [115].
coupled receptors through phosphatidylinositol 3-kinase (PI3K) [121].
Albiflorin, a monoterpene isolated from Paeoniae Radix, was investigat-
In addition, it has been reported that Akt can phosphorylate eNOS, one
ed for its ability to protect against antimycin A-induced osteoblast tox-
isoform of NOS [122]. Akt signaling has been reported to be involved
icity in the MC3T3-E1 cell line. The results suggested that albiflorin
in osteoblast proliferation and skeletal development [123], and Akt is
enhanced mitochondrial function to suppress antimycin A-induced ox-
upstream of NO pathway.
idative damage via the preservation of cytochrome c and cardiolipin.
Pretreatment with albiflorin before antimycin A reversed the loss of
cell viability, decreased apoptosis and lactate dehydrogenase release, 10.2. Natural products from TCM
decreased ROS/RNS levels, and increased mitochondrial function com-
pared to antimycin A-treated cultures. Albiflorin reduced antimycin A- Zhai et al. reported that the osteogenic effect of icariin involves the
induced mitochondrial cytochrome c loss and cardiolipin peroxidation, PI3K–Akt–eNOS–NO–cGMP–PKG signal pathway. Icariin increased ALP
conferring protection against ROS. In addition, albiflorin increased the activity, NOS activity, iNOS and eNOS expressions, NO production, sGC
mineralization reduced by antimycin A [116]. Honokiol is a phenolic and cGMP contents and PKG expression besides the phosphorylation
compound isolated from the bark of Magnolia officinalis. Honokiol of Akt. The addition of L-NAME, ODQ and PDE5 (the inhibitor of NOS,
caused a significant elevation of cell growth, ALP activity, collagen syn- sGC and cGMP) inhibited the icariin effects on above markers respec-
thesis, mineralization, glutathione content, and OPG release in the tively. The addition of LY294002 (the inhibitor of PI3K) decreased the
MC3T3-E1 cells (P b 0.05). Moreover, honokiol significantly (P b 0.05) p-Akt level, NOS activity, eNOS expression and NO production, but had
decreased the production of osteoclast differentiation inducing factors no significant effect on iNOS expression. The addition of any of the
such as TNF-α, IL-6, and RANKL in the presence of antimycin A, which four inhibitors also abolished the osteogenic effect of icariin on rat
inhibits mitochondrial electron transport and has been used as a ROS BMSCs as indicated by ALP activity, OCN synthesis, calcium deposition
J. An et al. / Life Sciences 147 (2016) 46–58 53

Table 1
The category, source, structure, dose, cell lines and mechanism of 36 natural compoundsa on osteoblast-mediated bone formation.

Compound name Category Source Structure Dose Cell lines used Mechanism References
for in vitro
studiesb

Icariin Flavonoid glucoside Herba epimedii 10−7, 10−6, MC3T3-E1 cells ↑(ALP, COL I, bone nodule); [50]
10−5, 10−4, ER-mediated ERK and JNK
10−3 M signal pathway
10−8 M Primary ↑(BMP-2, NO, Cbfa1/Runx2, [66]
osteoblast cells OPG/RANKL)
10−5 M rBMSCs PI3K–AKT–eNOS–NO–cGMP– [124]
PKG signal pathway
10−12, 10−9, hFOB 1.19 cells ↑(BMP-2, SMAD4, Cbfa1/ [89]
10−6 M Runx2, OPG/RANKL)
10−8 M UMR-106 cells ↑(ALP, OPG/RANKL); ligand- [67]
independent activation of ERα
−10
Baohuoside-I, epimedin B Flavonoid glucoside Herba epimedii 10 , UMR-106 cells ↑(ALP, OPG/RANKL); [67]
Sagittatoside A 10−8 M ER-dependent osteoblastic
functions; Sagittatoside A:
ligand-independent activation
of ER α

Neoeriocitrin Dihydro-flavonoid Drynaria Rhizome 2 μg mL−1 MC3T3-E1 cells ↑(ALP, Runx2, COL I, OCN) [51]

Naringin Dihydro-flavonoid Drynaria Rhizome 2 μg mL−1 MC3T3-E1 cells ↑(ALP, Runx2, COL I, OCN) [51]
Citrus fruits 0.3, 1, 3, MC3T3-E1 cells; BMP-2; PI3K, Akt, c-Fos/c-Jun [90]
10 μM hOB; primary and AP-1 pathway
osteoblastic cells
Poncirin Flavanone glycoside Poncirus trifoliata 0.1, 1, C3H10T1/2 ↓(PPAR-γ, C/EBP-β); ↑(Runx2, [52]
10 μM cells; BMMSCs TAZ, ALP, OCN)

Rutin Flavonoid Fruits and vegetables 25, 50, BMMSCs ↑(ALP, OPN, Osx, Runx2, [53]
100 μM OPG, OCN)

Quercetin Flavonoid Fruits and vegetables 25, 50, BMMSCs ↑(ALP, OPN, Osx, Runx2, [53]
100 μM OPG, OCN)
200, 500 μM MC3T3-E1 cells ↑(BSP, OCN); ↓(RANKL) [54]

Taxifolin Flavonoid Fruits and vegetables 100, MC3T3-E1 cells ↑(BSP, OCN); ↓(RANKL) [54]
200 μM

Ugonin K Flavonoid Helminthostachys 0.5, 1, 5, MC3T3-E1 cells ↑(ALP, BSP, OCN, Runx2, Osx); [75,105]
zeylanica 10 μM p38 MAPK- and ERK- mediated
pathway; ER-dependent
non-classical Src pathway and
classical pathway
Neobavaisoflavone Isoflavone Psoralea corylifolia 1, 5, 10, MC3T3-E1 cells ↑(ALP, COL I, OCN, BSP, Runx2 [76]
15, 20 μM and Osx); p38-dependent
pathway
Genistein Flavonoid Sophora japonica L. 10 μM MC3T3-E1 cells; Induce ERα expression [106]
primary rat through MAPK/NF-κB/AP-1
osteoblast cells pathway
Puerarin Isoflavone glycoside Pueraria mirifica 0.1 μM MG-63 cells ER-dependent MEK/ERK and [107]
PI3K/Akt activation
0.1, 10, UMR 106 cells ↑(ALP, OPG/RANKL); [65]
1000 nM ER-dependent

Echinacoside Phenylethanoid Cistanche tubulosa 0.01, 0.1, 1, MC3T3-E1 cells ↑(ALP, COL I, OCN, [60]
glycoside (Schrenk) R. Wight 10 nM OPG/RANKL)
stems

(continued on next page)


54 J. An et al. / Life Sciences 147 (2016) 46–58

Table 1 (continued)

Compound name Category Source Structure Dose Cell lines used Mechanism References
for in vitro
studiesb

Sweroside Iridoid glycoside Fructus Corni 10−6– MG-63 cells; rat ↑(ALP, OCN); inhibit apoptosis [55]
10−1 g mL−1 osteoblast cells

Salidroside Phenylpropanoid Rhodiola rosea L. 0.5, 1, 5, 10, C3H10T1/2 ↑(ALP, OPN, Runx2, Osx, [94]
glycoside 50 μM cells; MC3T3-E1 BMP-2, BMP-6, BMP-7,
cells phosphorylation of SMAD
1/5/8 and ERK1/2); BMP
signaling

Gastrodin Phenolic glycoside Gastrodia elata 0.1, 1, 10 μM hBMMSCs ↑(ALP, OCN, COL I, OPN); ↓ROS [115]

Ophiopogonin D Steroidal saponin Radix Ophiopogon 1, 10, MC3T3-E1 ↑(ALP, OCN, COL1A1, OPN); [118]
japonicus 100 μM subclone 4 cells ↓ROS; the FoxO3a-β-catenin
signaling pathway

Ginsenoside-Rb2 20 Ginseng 0.1, 1, 10 μM MC3T3-E1 cells ↑(ALP, COL I, OCN, OPN); [117]
(S)-protopanaxadiol ↓(RANKL, IL-6, ROS)
glycoside

2,3,5,4′-Tetrahydroxystil Polyhydrostilbenes Polygonum 0.1, 1, 10 μM MC3T3-E1 cells ↑(ALP, COL I, OCN); ↓(RANKL, [114]
bene-2-O-β-D-glucoside (glycoside) multiflorum Thunb IL-6, ROS, MDA)
(TSG)

Imperatorin Furanocoumarins Cnidium monnieri 0.3, 1, 3, Primary ↑(ALP, COL I, bone nodule, [79]
(L.) Cuss 10 μM osteoblastic BMP-2, phosphorylation of
cells SMAD 1/5/8); p38 and
ERK-dependent pathway

Bergapten

Psoralen Coumarin-like Psoralea corylifolia 1, 10, Primary mouse ↑(ALP, COL I, OCN, BSP, Osx, [91]
derivative 100 μM calvarial BMP-2, BMP-4, phosphorylation
osteoblasts of SMAD 1/5/8); BMP signaling
Osthole Coumarin-like Cnidium monnieri 10, 50, Primary ↑BMP-2; Wnt/β-catenin [92]
derivative (L.) Cusson 100 μM calvarial signaling
osteoblasts

Costunolide Sesquiterpene Saussurea lappa 10 μg mL−1 MC3T3-E1 cells ER, PI3K, ERK, PKC, p38, JNK, [78]
lactones and mitochondrial
ATP-sensitive K+ channel

Albiflorin Monoterpene Paeonia albiflora Pall 0.01, 0.1, MC3T3-E1 cells protect against oxidative [116]
1 μM stress-mediated toxicity

Kirenol Diterpenoid Herba Siegesbeckia 40 μM MC3T3-E1 cells ↑(ALP, COL I, OPN, OPG/ [62]
RANKL, BMP-2, Runx2, Osx;
LRP5, DVL2, β-catenin,
CCND1); BMP and
Wnt/β-catenin pathways
Vanillic acid Phenolic acid Sambucus williamsii 10−10, UMR 106 cells ↑(ALP, Runx2, OCN, [61]
Hance 10−8 M OPG/RANKL); MAPK
(MEK/ERK)-mediated ER
signaling pathway
J. An et al. / Life Sciences 147 (2016) 46–58 55

Table 1 (continued)

Compound name Category Source Structure Dose Cell lines used Mechanism References
for in vitro
studiesb

Salvianolic acid B Phenolic acid Salvia miltiorrhiza 5 μM hMSCs ↑(ALP, OPN, Runx2, Osx); ERK [77]
signaling pathway

Honokiol Phenolic Magnolia officinalis 0.01, 0.1, MC3T3-E1 cells ↑(ALP, COL I, OPG/RANKL, [63]
1 μM glutathione); ↓(TNF-α, IL-6,
mitochondrial electron
transport)

Resveratrol Polyphenol Red grapes and 10−8, 10−6, hBMMSCs ↑(ALP, Runx2/Cbfa1, Osx, [108]
berries 10−5 M OCN); ER-dependent ERK1/2
pathway

Apocynin Methoxy-substituted Picrorrhiza kurrooa 0.01, 0.1, MC3T3-E1 cells ↑(ALP, COL I, OPG/RANKL); [64]
catechol Royle ex Benth 1 μM ↓(ROS, TNF-α, IL-6)

Kobophenol A Tetrameric stilbene Caragana sinica Rhed 25, MG-63 cells ↓(NO-induced cell death); [125]
50 μg mL−1 regulate JNK, NF-κΒ and AP-1
signaling pathways
Human ↑(ALP); ↓(ROS); p38 pathway [80]
osteoblast-like
cells (MG-63,
HOS, SaOS-2)
Emodin Anthraquinone The genus Rhamnus 1, 2, 5 μM MC3T3-E1 ↑(BMP-2, ALP); PI3K, Akt and [93]
subclone 4 cells MAPK pathways

Acerogenin A Diarylheptanoids Acer nikoense Maxim 30 μM MC3T3-E1 cells; ↑(OCN, Osx, Runx2, BMP-2, [95]
RD-C6 cells; BMP-4, BMP-7)
primary
osteoblastic
cells
a
The 36 natural compounds include 14 flavonoids, 7 glycosides, 4 coumarins, 3 terpenoids, 2 phenolic acids, 3 phenols and 3 others.
b
Bone marrow mesenchymal stem cells (BMMSCs): multipotent stromal cells; C3H10T1/2: mesenchymal stem cells; hFOB 1.19: human osteoblastic cell line; MC3T3-E1 cells: oste-
oblast-like cell line; MG-63 cells: osteoblast-like cell line; UMR106 cells: osteoblast-like cell line.

and the number and areas of calcified nodules [124]. Hsieh's study this paper, it could be concluded that herbs with kidney-tonifying,
showed that 10− 8 M icariin increased the proliferation and matrix spleen-tonifying, and stasis-removing effects all have the potential ef-
mineralization of osteoblasts and promoted NO synthesis. L-NAME di- fect on treating osteoporosis. As shown in Table 1, the active ingredients
minished the icariin-induced cell proliferation, ALP activity, NO produc- from TCM that possess effects on promoting osteoblasts proliferation
tion, as well as the BMP-2, SMAD4, Cbfa1/Runx2, OPG, RANKL gene and differentiation including flavonoids, glycosides, coumarins, terpe-
expressions [66]. Kobophenol A (kob A), a tetrameric stilbene, is one noids (sesquiterpenoids, monoterpenoids, diterpenoids), phenolic
of the major active compounds from Caragana sinica Rhed. Sodium ni- acids, phenols and others (tetrameric stilbene, anthraquinones,
troprusside (SNP), a direct NO donor, can induce cell death and NO diarylheptanoids). And it is confirmed that the bone formation effect in-
production in MG-63 cells. Lee's study suggested that co-incubation of duced by natural products was regulated by the expressions of bone
kob A in SNP-treated MG-63 cells resulted in a significant protection specific matrix proteins (ALP, BSP, OCN, OPN, COL I), transcription fac-
against NO-induced cell death. Furthermore, kob A inhibited SNP- tors (Runx2, Cbfa1, Osx), signal pathways (MAPK, BMP), local factors
induced phosphorylation of JNK and c-Jun, and SNP-induced reduction (ROS, NO), OPG/RANKL system of osteoblasts and estrogen-like biolog-
in NF-κΒ and AP-1 activities, implicated that protective effect of kob A ical activities. It provided theoretical basis for clinical application, as
might occur through the regulation of JNK, NF-κΒ and AP-1 signaling well as new drug research and development on treating osteoporosis.
pathways [125]. Combined with modern scientific technologies, new developments
will be achieved in the fields of pharmacodynamic material basis, effect
11. Conclusions and future prospects targets and signaling pathways of TCM on the function of osteoblasts-
mediated bone formation in the future. Natural products on prevention
Cell molecular research on the pharmacological mechanisms behind and treatment of osteoporosis possess the whole regulative effects and
osteoblasts-mediated bone formation induced by natural products from the fewer adverse reactions, have received extensive attentions and re-
TCM has made great progress. According to the information collected in searches at home and abroad. It is desirable to have satisfactory anabolic
56 J. An et al. / Life Sciences 147 (2016) 46–58

agents that stimulate new bone formation and regulate the bone me- [30] J.R. Farley, S.L. Hall, M.A. Tanner, J.E. Wergedal, Specific activity of skeletal alkaline
phosphatase in human osteoblast-line cells regulated by phosphate, phosphate es-
tabolism balance, which would create a new alternative for treating ters, and phosphate analogs and release of alkaline phosphatase activity inversely
osteoporosis. regulated by calcium, J. Bone Miner. Res. 9 (1994) 497–508.
[31] M. Balcerzak, E. Hamade, L. Zhang, S. Pikula, G. Azzar, J. Radisson, et al., The roles of
annexins and alkaline phosphatase in mineralization process, Acta Biochim. Pol. 50
Conflict of interest statement
(2003) 1019–1038.
[32] G.S. Stein, J.B. Lian, T.A. Owen, Relationship of cell growth to the regulation of
The authors declare that there are no conflicts of interest. tissue-specific gene expression during osteoblast differentiation, FASEB J. 4
(1990) 3111–3123.
Acknowledgments [33] C. Wennberg, L. Hessle, P. Lundberg, S. Mauro, S. Narisawa, U.H. Lerner, et al., Func-
tional characterization of osteoblasts and osteoclasts from alkaline phosphatase
knockout mice, J. Bone Miner. Res. 15 (2000) 1879–1888.
The authors acknowledge financial support from the Youth Fund of [34] J.A.R. Gordon, C.E. Tye, A.V. Sampaio, T.M. Underhill, G.K. Hunter, H.A. Goldberg,
National Natural Science Foundation of China (No. 81403278). Bone sialoprotein expression enhances osteoblast differentiation and matrix min-
eralization in vitro, Bone 41 (2007) 462–473.
[35] P. Ducy, C. Desbois, B. Boyce, G. Pinero, B. Story, C. Dunstan, et al., Increased bone
References formation in osteocalcin-deficient mice, Nature 382 (1996) 448–452.
[36] K. Merry, R. Dodds, A. Littlewood, M. Gowen, Expression of osteopontin mRNA by
[1] G. Karsenty, The complexities of skeletal biology, Nature 423 (2003) 316–318. osteoclasts and osteoblasts in modelling adult human bone, J. Cell Sci. 104 (1993)
[2] M.R. Urist, Bone: formation by autoinduction, Science 150 (1965) 893–899. 1013–1020.
[3] D.J. Hadjidakis, I. Androulakis, Bone remodeling, Ann. N. Y. Acad. Sci. 1092 (2006) [37] S.R. Rittling, H.N. Matsumoto, M.D. Mckee, A. Nanci, X.R. An, K.E. Novick, et al., Mice
385–396. lacking osteopontin show normal development and bone structure but display al-
[4] L.G. Raisz, Pathogenesis of osteoporosis: concepts, conflicts, and prospects, J. Clin. tered osteoclast formation in vitro, J. Bone Miner. Res. 13 (1998) 1101–1111.
Investig. 115 (2005) 3318–3325. [38] H. Yoshitake, S.R. Rittling, D.T. Denhardt, M. Noda, Osteopontin-deficient mice are
[5] B.L. Riggs, H. Wahner, E. Seeman, K. Offord, W. Dunn, R. Mazess, et al., Changes in resistant to ovariectomy-induced bone resorption, Proc. Natl. Acad. Sci. U. S. A. 96
bone mineral density of the proximal femur and spine with aging, J. Clin. Investig. (1999) 8156–8160.
70 (1982) 716–723. [39] M. Ishijima, K. Tsuji, S.R. Rittling, T. Yamashita, H. Kurosawa, D.T. Denhardt, et al.,
[6] A. Leboime, C. Confavreux, N. Mehsen, J. Paccou, C. David, C. Roux, Osteoporosis Resistance to unloading-induced three-dimensional bone loss in osteopontin-
and mortality, Joint Bone Spine 77 (Suppl. 2) (2010) S107–S112. deficient mice, J. Bone Miner. Res. 17 (2002) 661–667.
[7] M.C.A. Buencamino, A.L. Sikon, A. Jain, H.L. Thacker, An observational study on the [40] M. Mizuno, Y. Kuboki, Osteoblast-related gene expression of bone marrow cells
adherence to treatment guidelines of osteopenia, J. Women's Health 18 (2009) during the osteoblastic differentiation induced by type I collagen, J. Biochem. 129
873–881. (2001) 133–138.
[8] O. Johnell, J.A. Kanis, An estimate of the worldwide prevalence and disability asso- [41] S. Mathews, R. Bhonde, P.K. Gupta, S. Totey, Extracellular matrix protein mediated
ciated with osteoporotic fractures, Osteoporos. Int. 17 (2006) 1726–1733. regulation of the osteoblast differentiation of bone marrow derived human mesen-
[9] C. Hu, C. Wang, L. Mo, C. Lin, L. Sun, A review on antiosteoporotic drugs, Chin. J. chymal stem cells, Differentiation 84 (2012) 185–192.
Gerontol. 11 (2008) 2178–2180. [42] E. Hinoi, S. Fujimori, L. Wang, H. Hojo, K. Uno, Y. Yoneda, Nrf2 negatively regulates
[10] P. Piscitelli, G. Iolascon, F. Gimigliano, M. Muratore, P. Camboa, O. Borgia, et al., In- osteoblast differentiation via interfering with Runx2-dependent transcriptional ac-
cidence and costs of hip fractures compared to acute myocardial infarction in the tivation, J. Biol. Chem. 281 (2006) 18015–18024.
Italian population: a 4-year survey, Osteoporos. Int. 18 (2007) 211–219. [43] J. Pratap, M. Galindo, S.K. Zaid, D. Vradii, B.M. Bhat, J.A. Robinson, et al., Cell growth
[11] L. Melton, E. Chrischilles, C. Cooper, A. Lane, B. Riggs, Perspective. How many regulatory role of Runx2 during proliferative expansion of preosteoblasts, Cancer
women have osteoporosis? J. Bone Miner. Res. 7 (1992) 1005–1010. Res. 63 (2003) 5357–5362.
[12] R. Burge, B. Dawson-Hughes, D. Solomon, J. Wong, A. King, A. Tosteson, Incidence [44] V. Geoffroy, M. Kneissel, B. Fournier, A. Boyde, P. Matthias, High bone resorption in
and economic burden of osteoporosis related fractures in the United States, adult aging transgenic mice overexpressing cbfa1/runx2 in cells of the osteoblastic
2005–2025, J. Bone Miner. Res. 22 (2007) 465–475. lineage, Mol. Cell. Biol. 22 (2002) 6222–6233.
[13] V. Beral, Million women study collaborators, breast cancer and hormone- [45] R.T. Franceschi, G. Xiao, D. Jiang, R. Gopalakrishnan, S. Yang, E. Reith, Multiple sig-
replacement therapy in the million women study, Lancet 362 (2003) 419–427. naling pathways converge on the Cbfa1/Runx2 transcription factor to regulate os-
[14] M.B. Zanchetta, M. Diehl, M. Buttazzoni, A. Galich, F. Silveira, C.E. Bogado, et al., As- teoblast differentiation, Connect. Tissue Res. 44 (2003) 109–116.
sessment of bone microarchitecture in postmenopausal women on long term bis- [46] P. Ducy, R. Zhang, V. Geoffroy, A.L. Ridall, G. Karsenty, Osf2/Cbfa1: a transcriptional
phosphonate therapy with atypical fractures of the femur, J. Bone Miner. Res. 29 activator of osteoblast differentiation, Cell 89 (1997) 747–754.
(2013) 999–1004. [47] K. Nakashima, X. Zhou, G. Kunkel, Z. Zhang, J.M. Deng, R.R. Behringer, et al., The
[15] J. Pinkerton, S. Thomas, Use of SERMs for treatment in postmenopausal women, novel zinc finger-containing transcription factor osterix is required for osteoblast
J. Steroid Biochem. Mol. Biol. 142 (2014) 142–154. differentiation and bone formation, Cell 108 (2002) 17–29.
[16] K. Cappuzzo, J. Delafuente, Teriparatide for severe osteoporosis, Ann. [48] M.F. Pittenger, A.M. Mackay, S.C. Beck, R.K. Jaiswal, R. Douglas, J.D. Mosca, et al.,
Pharmacother. 38 (2004) 294–302. Multilineage potential of adult human mesenchymal stem cells, Science 284
[17] T. Ponnapakkam, R. Katikaneni, J. Sakon, R. Stratford, R. Gensure, Treating osteopo- (1999) 143–147.
rosis by targeting parathyroid hormone to bone, Drug Discov. Today 19 (2013) [49] D. Prusty, B.H. Park, K.E. Davis, S.R. Farmer, Activation of MEK/ERK signaling pro-
204–208. motes adipogenesis by enhancing peroxisome proliferator-activated receptor
[18] G.A. Rodan, T.J. Martin, Therapeutic approaches to bone diseases, Science 289 gamma (PPARgamma) and C/EBPalpha gene expression during the differentiation
(2000) 1508–1514. of 3T3-L1 preadipocytes, J. Biol. Chem. 277 (2002) 46226–46232.
[19] S.K. Chair, D. Burr, J. Cauley, D.W. Dempster, P.R. Ebeling, D. Felsenberg, et al., [50] L. Song, J. Zhao, X. Zhang, H. Li, Y. Zhou, Icariin induces osteoblast proliferation, dif-
Bisphosphonate-associated osteonecrosis of the jaw: report of a task force of the ferentiation and mineralization through estrogenre ceptor-mediated ERK and JNK
American society for bone and mineral research, J. Bone Miner. Res. 22 (2007) signal activation, Eur. J. Pharmacol. 714 (2013) 15–22.
1479–1491. [51] L. Li, Z. Zeng, G. Cai, Comparison of neoeriocitrin and naringin on proliferation and
[20] J.P. Bilezikian, Combination anabolic and antiresorptive therapy for osteoporosis: osteogenic differentiation in MC3T3-E1, Phytomedicine 18 (2011) 985–989.
opening the anabolic window, Curr. Osteoporos. Rep. 6 (2008) 24–30. [52] H.Y. Yoon, S.I. Yun, B.Y. Kim, Q. Jin, E.R. Woo, S.Y. Jeong, et al., Poncirin promotes
[21] D.M. Black, S.L. Greenspan, K.E. Ensrud, L. Palermo, J.A. Mcgowan, T.F. Lang, et al., osteoblast differentiation but inhibits adipocyte differentiation in mesenchymal
The effects of parathyroid hormone and alendronate alone or in combination in stem cells, Eur. J. Pharmacol. 664 (2011) 54–59.
postmenopausal osteoporosis, N. Engl. J. Med. 349 (2003) 1207–1215. [53] S. Srivastava, R. Bankar, P. Roy, Assessment of the role of flavonoids for inducing os-
[22] J.S. Finkelstein, A. Hayes, J.L. Hunzelman, J.J. Wyland, H. Lee, A.R.M. Neer, The effects teoblast differentiation in isolated mouse bone marrow derived mesenchymal
of parathyroid hormone, alendronate, or both in men with osteoporosis, N. Engl. J. stem cells, Phytomedicine 20 (2013) 683–690.
Med. 349 (2003) 1216–1226. [54] M.A. Satue´, M.D.M. Arriero, M. Monjo, J.M. Ramis, Quercitrin and taxifolin stimu-
[23] A.V. Uihlein, B.Z. Leder, Anabolic therapies for osteoporosis, Endocrinol. Metab. late osteoblast differentiation in MC3T3-E1 cells and inhibit osteoclastogenesis in
Clin. N. Am. 41 (2012) 507–525. RAW 264.7 cells, Biochem. Pharmacol. 86 (2013) 1476–1486.
[24] Y. Liu, J. Liu, Y. Xia, Chinese herbal medicines for treating osteoporosis, Cochrane [55] H. Sun, L. Li, A. Zhang, N. Zhang, H. Lv, W. Sun, et al., Protective effects of sweroside
Database Syst. Rev. (2014), http://dx.doi.org/10.1002/14651858.CD005467.pub2. on human MG-63 cells and rat osteoblasts, Fitoterapia 84 (2013) 174–179.
[25] H. Xu, D. Lawson, A. Kras, D. Ryan, The use of preventive strategies for bone loss, [56] W.S. Simonet, D.L. Lacey, C.R. Dunstan, M. Kelley, M.S. Chang, R. Lüthy, et al.,
Am. J. Chin. Med. (2005) 299–306. Osteoprotegerin: a novel secreted protein involved in the regulation of bone den-
[26] A. Maurice, Drug combination strategies for osteoporosis, Joint Bone Spine 73 sity, Cell 89 (1997) 309–319.
(2006) 374–378. [57] D.L. Lacey, E. Timms, H.L. Tan, M.J. Kelley, C.R. Dunstan, T. Burgess, et al., Osteopro-
[27] R. Marcus, S. Majumdar, The nature of osteoporosis, in: R. Marcus, D. Feldman, J. tegerin ligand is a cytokine that regulates osteoclast differentiation and activation,
Kelsey (Eds.), Osteoporosis, Seconded, Academic Press, San Diego 2001, pp. 3–17. Cell 93 (1998) 165–176.
[28] R.T. Franceschi, C. Ge, G. Xiao, H. Roca, D. Jiang, Transcriptional regulation of oste- [58] T. Ikeda, M. Utsuyama, K. Hirokawa, Expression profiles of receptor activator of nu-
oblasts, Ann. N. Y. Acad. Sci. 1116 (2007) 196–207. clear factor κB ligand, receptor activator of nuclear factor κB, and osteoprotegerin
[29] I. Bremner, J. Beattie, Copper and zinc metabolism in health and disease: speciation messenger RNA in aged and ovariectomized rat bones, J. Bone Miner. Res. 16
and interactions, Proc. Nutr. Soc. 54 (1995) 489–499. (2001) 1416–1425.
J. An et al. / Life Sciences 147 (2016) 46–58 57

[59] K. S, The OPG/RANKL/RANK system, Endocrinology 142 (2001) 5050–5055. [88] J.Q. Feng, L. Xing, J.H. Zhang, M. Zhao, D. Horn, J. Chan, et al., NF-κB specifically ac-
[60] F. Li, Y. Yang, P. Zhu, W. Chen, D. Qi, X. Shi, et al., Echinacoside promotes bone re- tivates BMP-2 gene expression in growth plate chondrocytes in vivo and in a chon-
generation by increasing OPG/RANKL ratio in MC3T3-E1 cells, Fitoterapia 83 drocyte cell line in vitro, J. Biol. Chem. 278 (2003) 29130–29135.
(2012) 1443–1450. [89] W. Liang, M. Lin, X. Li, C. Li, B. Gao, H. Gan, et al., Icariin promotes bone formation
[61] H. Xiao, Q. Gao, Y. Zhang, K.C. Wong, Y. Dai, X. Yao, et al., Vanillic acid exerts via the BMP-2/Smad4 signal transduction pathway in the hFOB 1.19 human oste-
oestrogen-like activities in osteoblast-like UMR 106 cells through MAP kinase oblastic cell line, Int. J. Mol. Med. 30 (2012) 889–895.
(MEK/ERK)-mediated ER signaling pathway, J. Steroid Biochem. Mol. Biol. 144 [90] J.B. Wu, Y.C. Fong, H.Y. Tsai, Y.F. Chen, M. Tsuzuki, C.H. Tang, Naringin-induced
(2014) 382–391. bone morphogenetic protein-2 expression via PI3K, Akt, c-Fos/c-Jun and AP-1
[62] M.B. Kim, Y. Song, J.K. Hwang, Kirenol stimulates osteoblast differentiation through pathway in osteoblasts, Eur. J. Pharmacol. 588 (2008) 333–341.
activation of the BMP and Wnt/β-catenin signaling pathways in MC3T3-E1 cells, [91] D.Z. Tang, F. Yang, Z. Yang, J. Huang, Q. Shi, D. Chen, et al., Psoralen stimulates os-
Fitoterapia 98 (2014) 59–65. teoblast differentiation through activation of BMP signaling, Biochem. Biophys. Res.
[63] E.M. Choi, Honokiol isolated from Magnolia officinalis stimulates osteoblast func- Commun. 405 (2011) 256–261.
tion and inhibits the release of bone-resorbing mediators, Int. Immunopharmacol. [92] D.Z. Tang, W. Hou, Q. Zhou, M. Zhang, J. Holz, T.J. Sheu, et al., Osthole stimulates os-
11 (2011) 1541–1545. teoblast differentiation and bone formation by activation of b-catenin-BMP signal-
[64] Y.S. Lee, E.M. Choi, Apocynin stimulates osteoblast differentiation and inhibits ing, J. Bone Miner. Res. 25 (2010) 1234–1245.
bone-resorbing mediators in MC3T3-E1 cells, Cell. Immunol. 270 (2011) 224–229. [93] S.U. Lee, H.K. Shin, Y.K. Min, S.H. Kim, Emodin accelerates osteoblast differentiation
[65] W. Tiyasatkulkovit, N. Charoenphandhu, K. Wongdee, J. Thongbunchoo, N. through phosphatidylinositol 3-kinase activation and bone morphogenetic
Krishnamra, S. Malaivijitnondc, Upregulation of osteoblastic differentiation marker protein-2 gene expression, Int. Immunopharmacol. 8 (2008) 741–747.
mRNA expression in osteoblast-like UMR106 cells by puerarin and phytoestrogens [94] J. Chen, N. Zhang, G. Mao, X. He, Y. Zhan, H. Deng, et al., Salidroside stimulates os-
from Pueraria mirifica, Phytomedicine 19 (2012) 1147–1155. teoblast differentiation through BMP signaling pathway, Food Chem. Toxicol. 62
[66] T.P. Hsieh, S.Y. Sheu, J.S. Sun, M.H. Chen, M.H. Liu, Icariin isolated from epimedium (2013) 499–505.
pubescens regulates osteoblasts anabolism through BMP-2, SMAD4, and Cbfa1 ex- [95] T. Kihara, S. Ichikawa, T. Yonezawa, J.-W. Lee, T. Akihisa, J.T. Woo, et al., Acerogenin
pression, Phytomedicine 17 (2010) 414–423. A, a natural compound isolated from Acer nikoense Maxim, stimulates osteoblast
[67] H.H. Xiao, C.Y. Fung, S.K. Mok, K.C. Wong, M.X. Ho, X.L. Wang, et al., Flavonoids differentiation through bone morphogenetic protein action, Biochem. Biophys.
from herba epimedii selectively activate estrogen receptor alpha (ERα) Res. Commun. 406 (2011) 211–217.
andstimulate ER-dependent osteoblastic functionsin UMR-106 cells, J. Steroid [96] K.D. Wright, V. Cavailles, S.A. Fuqua, V.C. Jordan, J.A. Katzenellenbogen, K.S. Korach,
Biochem. Mol. Biol. 143 (2014) 141–151. et al., International union of pharmacology. LXIV. Estrogen receptors, Pharmacol.
[68] L. Chang, M. Karin, Mammalian MAP kinase signalling cascades, Nature 410 (2001) Rev. 58 (2006) 773–781.
37–40. [97] R.T. Turner, B.L. Riggs, T.C. Spelsberg, Skeletal effects of estrogen, Endocr. Rev. 15
[69] T. Akune, S. Ohba, S. Kamekura, M. Yamaguchi, U. Chung, N. Kubota, et al., PPARγ (1994) 275–300.
insufficiency enhances osteogenesis through osteoblast formation from bone mar- [98] B.J. Cheskis, J.G. Greger, S. Nagpal, L.P. Freedman, Signaling by estrogens, J. Cell.
row progenitors, J. Clin. Investig. 113 (2004) 846–855. Physiol. 213 (2007) 610–617.
[70] E. Hu, J. Kim, P. Sarraf, B. Spiegelman, Inhibition of adipogenesis through [99] M. Marino, M. Pellegrini, P.L. Rosa, F. Acconcia, Susceptibility of estrogen receptor
MAP kinase-mediated phosphorylation of PPARgamma, Science 274 (1996) rapid responses to xenoestrogens: physiological outcomes, Steroids 77 (2012)
2100–2103. 910–917.
[71] M. Aouadi, K. Laurent, M. Prot, Y.L.M. Brustel, B. Binétruy, F. Bost, Inhibition of p38 [100] S. Kato, H. Endoh, Y. Masuhiro, T. Kitamoto, S. Uchiyama, H. Sasaki, et al., Activation
MAPK increases adipogenesis from embryonic to adult stages, Diabetes 55 (2006) of the estrogen receptor through phosphorylation by mitogen-activated protein ki-
281–289. nase, Science 270 (1995) 1491–1494.
[72] M.B. Greenblatt, J.H. Shim, W. Zou, D. Sitara, M. Schweitzer, D. Hu, et al., The p38 [101] C.J. Gruberemail, D.M. Gruber, I.M.L. Gruber, F. Wieser, J.C. Huber, Anatomy of the
MAPK pathway is essential for skeletogenesis and bone homeostasis in mice, estrogen response element, Trends Endocrinol. Metab. 15 (2004) 73–78.
J. Clin. Investig. 120 (2010) 2457–2473. [102] A. Migliaccio, G. Castoria, M. Domenico, A. Falco, A. Bilancio, F. Auricchio, Src is an
[73] C. Ge, G. Xiao, D. Jiang, R.T. Franceschi, Critical role of the extracellular signal- initial target of sex steroid hormone action, Ann. N. Y. Acad. Sci. 963 (2002)
regulated kinase-MAPK pathway in osteoblast differentiation and skeletal devel- 185–190.
opment, J. Cell Biol. 176 (2007) 709–718. [103] A. Zallone, Direct and indirect estrogen actions on osteoblasts and osteoclasts, Ann.
[74] X. Wang, C.H. Goh, B. Li, p38 Mitogen-activated protein kinase regulates osteoblast N. Y. Acad. Sci. 1068 (2006) 173–179.
differentiation through osterix, Endocrinology 148 (2007) 1629–1637. [104] S. Kousteni, T. Bellido, L.I. Plotkin, C.A. O'brien, D.L. Bodenner, L. Han, et al.,
[75] C.H. Lee, Y.L. Huang, J.F. Liao, W.F. Chiou, Ugonin K promotes osteoblastic differen- Nongenotropic, sex-nonspecific signaling through the estrogen or androgen recep-
tiation and mineralization by activation of p38 MAPK- and ERK-mediated expres- tors: dissociation from transcriptional activity, Cell 104 (2001) 719–730.
sion of Runx2 and osterix, Eur. J. Pharmacol. 668 (2011) 383–389. [105] C.H. Lee, Y.L. Huang, J.F. Liao, W.F. Chiou, Ugonin K-stimulated osteogenesis in-
[76] M.J. Don, L.C. Lin, W.F. Chiou, Neobavaisoflavone stimulates osteogenesis via p38- volves estrogen receptor-dependent activation of non-classical Src signaling path-
mediated up-regulation of transcription factors and osteoid genes expression in way and classical pathway, Eur. J. Pharmacol. 676 (2012) 26–33.
MC3T3-E1 cells, Phytomedicine 19 (2012) 551–561. [106] M.H. Liao, Y.T. Tai, Y.G. Cherng, S.H. Liu, Y.A. Chang, P.I. Lin, et al., Genistein induces
[77] D. Xu, L. Xu, C. Zhou, W.Y.W. Lee, T. Wu, L. Cui, et al., Salvianolic acid B promotes oestrogen receptor-α gene expression in osteoblasts through the activation of
osteogenesis of human mesenchymalstem cells through activating ERK signaling mitogen-activated protein kinases/NF-κB/activator protein-1 and promotes cell
pathway, Int. J. Biochem. Cell Biol. 51 (2014) 1–9. mineralisation, Br. J. Nutr. 111 (2014) 55–63.
[78] Y.S. Lee, E.M. Choi, Costunolide stimulates the function of osteoblastic MC3T3-E1 [107] Y. Wang, W.L. Wang, W.L. Xie, L.Z. Li, J. Sun, W.J. Sun, et al., Puerarin stimulates pro-
cells, Int. Immunopharmacol. 11 (2011) 712–718. liferation and differentiation and protects against celldeath in human osteoblastic
[79] C.H. Tang, R.S. Yang, M.Y. Chien, C.C. Chen, W.M. Fu, Enhancement of bone MG-63 cells via ER-dependent MEK/ERK and PI3K/Akt activation, Phytomedicine
morphogenetic protein-2 expression and bone formation by coumarin derivatives 20 (2013) 787–796.
via p38 and ERK-dependent pathway in osteoblasts, Eur. J. Pharmacol. 579 (2008) [108] Z. Dai, Y. Li, L.D. Quarles, T. Song, W. Pan, H. Zhou, et al., Resveratrol enhances pro-
40–49. liferation and osteoblastic differentiation in human mesenchymal stem cells via
[80] J.H. Kwak, S.R. Lee, H.J. Park, H.E. Byun, E.H. Sohn, B.O. Kim, et al., Kobophenol A en- ER-dependent ERK1/2 activation, Phytomedicine 14 (2007) 806–814.
hances proliferation of human osteoblast-like cells with activation of the p38 path- [109] S.C. Manolagas, From oestrogen-centric to aging and oxidative stress: a revised
way, Int. Immunopharmacol. 17 (2013) 704–713. perspective of the pathogenesis of osteoporosis, Endocr. Rev. 31 (2010) 266–300.
[81] T. Sakou, T. Onishi, T. Yamamoto, T. Nagamine, T.K. Sampath, P.T. Dijke, Localization [110] O.F. Sendur, Y. Turan, E. Tastaban, M. Serter, Antioxidant status in patients with os-
of Smads, the TGF-β family intracellular signaling components during endochon- teoporosis: a controlled study, Joint Bone Spine 76 (2009) 514–518.
dral ossification, J. Bone Miner. Res. 14 (1999) 1145–1152. [111] X. Bai, D. Lu, A. Liu, Z. Zhang, X. Li, Z. Zou, et al., Reactive oxygen species stimulates
[82] A. Nohe, E. Keating, P. Knaus, N.O. Petersen, Signal transduction of bone morphoge- receptor activator of NF-kappaB ligand expression in osteoblast, J. Biol. Chem. 280
netic protein receptors, Cell. Signal. 16 (2004) 291–299. (2005) 17497–17506.
[83] U. Styrkarsdottir, J.B. Cazier, A. Kong, O. Rolfsson, H. Larsen, E. Bjarnadottir, et al., [112] H. Ha, H.B. Kwak, S.W. Lee, H.M. Jin, H.M. Kim, H.H. Kim, et al., Reactive oxygen
Linkage of osteoporosis to chromosome 20p12 and association to BMP2, PLoS species mediate RANK signaling in osteoclasts, Exp. Cell Res. 301 (2004)
Biol. 1 (2003), e69. 119–127.
[84] M.H. Lee, Y.J. Kim, H.J. Kim, H.D. Park, A.R. Kang, H.M. Kyung, et al., BMP-2-induced [113] K.H. Baek, K.W. Oh, W.Y. Lee, S.S. Lee, M.K. Kim, H.S. Kwon, et al., Association of ox-
Runx2 expression is mediated by Dlx5, and TGF-beta 1 opposes the BMP-2- idative stress with postmenopausal osteoporosis and the effects of hydrogen per-
induced osteoblast differentiation by suppression of Dlx5 expression, J. Biol. oxide on osteoclast formation in human bone marrow cell cultures, Calcif. Tissue
Chem. 278 (2003) 34387–34394. Int. 87 (2010) 226–235.
[85] T.F. Day, X. Guo, L.G. Beal, Y. Yang, Wnt/β-catenin signaling in mesenchymal pro- [114] J.K. Zhang, L. Yang, G.L. Meng, J. Fan, J.Z. Chen, Q.Z. He, et al., Protective effect of
genitors controls osteoblast and chondrocyte differentiation during vertebrate tetrahydroxystilbene glucoside against hydrogen peroxide-induced dysfunction
skeletogenesis, Dev. Cell 8 (2005) 739–750. and oxidative stress in osteoblastic MC3T3-E1 cells, Eur. J. Pharmacol. 689 (2012)
[86] M. Shtutman, J. Zhurinsky, I. Simcha, C. Albanese, M. D'amico, R. Pestell, et al., The cy- 31–37.
clin D1 gene is a target of the β-catenin/LEF-1 pathway, Proc. Natl. Acad. Sci. U. S. A. [115] Q. Huang, J. Shi, B. Gao, H.Y. Zhang, J. Fan, X.J. Li, et al., Gastrodin: an ancient
96 (1999) 5522–5527. Chinese herbal medicine as a source for anti-osteoporosis agents via reducing re-
[87] N. Ghosh-Choudhury, C.C. Mandal, G.G. Choudhury, Statin-induced Ras activation active oxygen species, Bone 73 (2015) 132–144.
integrates the phosphatidylinositol 3-kinase signal to Akt and MAPK for bone mor- [116] K.S. Suh, E.M. Choi, Y.S. Lee, Y.S. Kim, Protective effect of albiflorin against
phogenetic protein-2 expression in osteoblast differentiation, J. Biol. Chem. 282 oxidative-stress-mediated toxicity in osteoblast-like MC3T3-E1 cells, Fitoterapia
(2007) 4983–4993. 89 (2013) 33–41.
58 J. An et al. / Life Sciences 147 (2016) 46–58

[117] Q. Huang, B. Gao, Q. Jie, B.Y. Wei, J. Fan, H.Y. Zhang, et al., Ginsenoside-Rb2 displays [122] S. Dimmeler, I. Fleming, B. Fisslthaler, C. Hermann, R. Busse, A.M. Zeiher, Activation
anti-osteoporosis effects through reducing oxidative damage and bone-resorbing of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation,
cytokines during osteogenesis, Bone 66 (2014) 306–314. Nature 399 (1999) 601–605.
[118] Q. Huang, B. Gao, L. Wang, H.Y. Zhang, X.J. Li, J. Shi, et al., Ophiopogonin D: a new [123] S. Rokutanda, T. Fujita, N. Kanatani, C.A. Yoshida, H. Komori, W. Liu, et al., Akt reg-
herbal agent against osteoporosis, Bone 74 (2015) 18–28. ulates skeletal development through GSK3, mTOR, and FoxOs, Dev. Biol. 328
[119] S.J. Wimalawansa, Nitric oxide: novel therapy for osteoporosis, Expert. Opin. (2009) 78–93.
Pharmacother. 9 (2008) 3025–3044. [124] Y.K. Zhai, X.Y. Guo, B.F. Ge, P. Zhen, X.N. Ma, J. Zhou, et al., Icariin stimulates the os-
[120] S. Gutiérrez, J.P. Petiti, L.D.V. Sosa, L. Fozzatti, A.L.D. Paul, A.M. Masini-Repiso, et al., teogenic differentiation of rat bone marrow stromal cells via activating the PI3K–
17β-Oestradiol acts as a negative modulator of insulin-induced lactotroph cell pro- AKT–eNOS–NO–cGMP–PKG, Bone 66 (2014) 189–198.
liferation through oestrogen receptor alpha, via nitric oxide/guanylyl cyclase/ [125] S.R. Lee, J.H. Kwak, D.S. Park, S. Pyo, Protective effect of kobophenol A on nitric
cGMP, Cell Prolif. 43 (2010) 505–514. oxide-induced cell apoptosis in human osteoblast-like MG-63 cells: involvement
[121] L.C. Cantley, The phosphoinositide 3-kinase pathway, Science 296 (2002) of JNK, NF-κB and AP-1 pathways, Int. Immunopharmacol. 11 (2011) 1251–1259.
1655–1657.

You might also like