Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 2.
Published in final edited form as: Regen Med. 2012 Jul;7(4):535–549. doi: 10.2217/rme.12.33

The role of small molecules in musculoskeletal regeneration

Kevin W-H Lo 1,2, Keshia M Ashe 1,3,4, Ho Man Kan 1,3, Cato T Laurencin 1,3,4,*
PMCID: PMC4698169  NIHMSID: NIHMS744474  PMID: 22817627

Abstract

The uses of bone morphogenetic proteins and parathyroid hormone therapeutics are fraught with several fundamental problems, such as cost, protein stability, immunogenicity, contamination and supraphysiological dosage. These downsides may effectively limit their more universal use. Therefore, there is a clear need for alternative forms of biofactors to obviate the drawbacks of protein-based inductive factors for bone repair and regeneration. Our group has studied small molecules with the capacity to regulate osteoblast differentiation and mineralization because their inherent physical properties minimize limitations observed in protein growth factors. For instance, in general, small molecule inducers are usually more stable, highly soluble, nonimmunogenic, more affordable and require lower dosages. Small molecules with the ability to induce osteoblastic differentiation may represent the next generation of bone regenerative medicine. This review describes efforts to develop small molecule-based biofactors for induction, paying specific attention to their novel roles in bone regeneration.

Keywords: bone regeneration, growth factor, orthopedic, osteogenesis, small molecule, tissue engineering

Bone growth factors

Bone growth factors are usually proteins secreted by cells that provide a driving force for osteoblast functions including proliferation and maturation. The mechanism of action of bone protein growth factors is to interact with a membrane receptor on a target cell and trigger an intracellular signal transduction system that ultimately induces the expression of bone-specific genes in the nucleus and protein production in the cytoplasm (Figure 1A) [1].

Figure 1. Different forms of bone growth factors involved in osteoblast differentiation.

Figure 1

(A) The protein ligand interacts with the extracellular domain of the membrane receptor, and in turn the intracellular domain of the receptor activates the target signal cascade. (B) The small molecules pass through the cell membrane and directly activate the intracellular target signaling cascade. Activation of the signaling cascade will lead to modification of the transcription factor. The modified transcription factor will then translocate from the cytoplasm to the nucleus and induce the expression of the osteoblast-associated gene.

Stimulation of bone formation using bone protein growth factors is an established approach to treating musculoskeletal problems due to trauma, congenital deformities and specific bone diseases such as osteoporosis [1]. Particularly, bone morphogenetic proteins (BMPs) have been extensively used and studied due to their osteoinductive capacity – that is, the ability to induce ectopic bone formation [2,3]. BMP-2 and BMP-7 have obtained US FDA clearance for specific clinical applications such as spinal fusions and long-bone nonunions [46]. More recently, PTH 1–34 was approved by the FDA as an anabolic drug for the treatment of osteoporosis [710]. While successful, the uses of protein-based therapeutics have their drawbacks. The current recombinant BMPs and PTH medications are produced by Escherichia coli using recombinant DNA technology [8,11]. One of the major concerns of this technology is that the recombinant protein produced from E. coli is not glycosylated, as is normal in the body, and thus may present reduced stability and biological activity [12]. Additionally, since trace amounts of biologically active impurities from E. coli can jeopardize therapeutic applications, contamination can be another issue [13]. Additionally, the costly processes of recombinant protein production and purification make protein-based therapy very expensive [8,14,15]. Furthermore, the immunogeneticity of PTH and BMPs have been reported as potentially eliciting undesirable immune responses in patients [12,1618]. Although other protein growth factors such as FGF, IGFs, TGF-β and PDGF have shown potential for use in bone repair and regeneration [1,1922], they too have specific drawbacks. In general, high manufacturing cost, protein instability, unwanted immune response and contamination are the common limitations in protein-based therapeutic strategies (Table 1) [2325]. Therefore, there is clear need for an alternative and effective therapeutic strategy for treating bone diseases.

Table 1.

Comparison of therapeutic proteins and small molecules.

Advantages Disadvantages
Protein Specific Unstable
Impurities
High dose requirement
High cost
Immunogenic
Given by injection
Small molecule Low cost Nonspecific
Stable Short half-life
Nonimmunogenic
Can be given by oral route

Adapted with permission from [24].

Small molecules with the ability to induce osteoblast differentiation are appealing because their inherent properties can minimize or even overcome the drawbacks observed with protein-based growth factors (Table 1) [24,25]. For example, small molecules are unlikely to have any issues with immunogenicity because they are too small in size to stimulate an immune response [26]. Moreover, unlike protein growth factors, the bioactivities of small molecules usually do not require structural integrity [23,2729]. Interestingly, many small molecule drugs can be taken through the oral route, and therefore avoiding painful multiple injections [30]. More importantly, compared with recombinant proteins, many small molecule drugs are inexpensive organic compounds [15,31]. Considering a growing number of small molecules have been recently discovered with osteoblastic differentiation capability, they may represent the next generation of bone regenerative medicine. This review article will focus on recent developments and the prospective future of small molecules associated with bone regeneration.

Small molecules with osteoinductive potential

A small molecule is a low molecular weight organic compound (i.e., molecular weight is usually less than 1000 Da) that can diffuse across the cell membrane to reach intracellular targets, and in certain cases, act as a signaling molecule in a number of cell signaling pathways [15,32]. Typically, the small molecules trigger intracellular signaling pathways by specifically binding to a protein receptor in the cytoplasm, which in turn leads to particular gene transcription and eventually protein expression, including osteoblast-associated genes (Figure 1b) [23,33]. Signal transduction systems such as cAMP/PKA [23,34], Hh [35,36], BMP/Smad [37], MAPK [38,39] and Wnt have well-known roles in regulating osteogenesis [40,41]. Therefore, identification of the small molecules that regulate these signaling cascades is the current drug discovery strategy in the area of bone repair and regeneration [15]. Using high-throughput screens and/or cell-based functional assays, a large number of small molecules have been recently discovered based on their capability of regulating osteoblast-associated gene expression and inducing the osteogenic phenotype through the activity of the osteogenic-related signaling pathways. Table 2 summarizes these small molecules and their target signaling mechanisms.

Table 2.

Small molecules that promote osteoblast differentiation with known signaling targets.

Small molecule In vitro effective dosage (μM) Target signaling In vitro activity (cell types) In vivo activity Ref.
6-Bnz-cAMP 100 PKA/CREB MC3T3-E1; rat and human MSCs Not reported [23]
XT-611 0.1 PKA/CREB Rat and mouse bone marrow cells Not reported [80]
AICAR 1–2 PKA Human amnion MSCs; rabbit MSCs Not reported [141]
AMG0892, AMG0309 0.01–0.2 CREB MC3T3-E1 Not reported [81]
Statins§ 0.0001–5 BMP/Smad MC3T3-E1; MG63; rat and human bone marrow stromal cells; mouse embryonic stem cells Subcutaneous injection over the calvaria of mice; ovariectomized rats treated systemically [49,53, 142144]
Phenamil 10 BMP/Smad Rat MSCs Not reported [71]
FK506§ 0.05–1 BMP/Smad Rat bone marrow cells; rat MSCs; MC3T3-E1; ST-2; C2C12; C17 Subcutaneous implantation in rats [63,65,145]
Rapamycin§ 0.05 BMP/Smad ROS 17/2.8; human embryonic stem cells Subcutaneous implantation in mice [69,70,77,146]
TH 1 BMP/Smad MC3T3-E1; mouse primary osteoblast; mouse embryonic stem cells Calvarial critical-sized defect in mouse [60,72]
Icariin BMP MC3T3-E1; mouse primary osteoblasts Ovariectomized mice treated systemically; calvarial defect model in mouse [66,67,147]
Purmorphamine 1 Hh C3H10T1/2 cells Not reported [82]
WAY-316606 0.03 Wnt Ex vivo mouse calvarial organ culture Not reported [95]
TAK-778 10 ER dependent Human bone marrow cells Not reported [89]
Oxysterols 2.5 Hh and Wnt M2-10B4 Critical-sized rat calvarial defect model [100,148]
Herbal medicines
Puerarin 0.1 PI3K/Akt Rat calvaria osteoblasts Intramuscular implantation in rats; calvarial parietal bone defect model in rabbit [149,150]
Genistin 1 ER dependent Human bone marrow stromal cells Calvarial parietal bone defect model in rabbit [151,152]
Psoralen 100 BMP Mouse calvarial osteoblasts Calvarial parietal bone defect model in rabbit [153,154]
Osthole 100 β-catenin/BMP Mouse calvarial osteoblasts Ovariectomized rats treated systemically [155,156]
Maohuoside A 80 BMP/MAPK Rat MSCs Not reported [157]
Berberine ~150 MAPK C3H10T1/2; MC3T3-E1 Not reported [158]
Resveratrol 10 ER dependent Human MSCs Not reported [159]

Unpublished observations.

Currently under investigation.

§

Osteoinductive small molecules.

ER: Estrogen receptor; MSC: Mescenchymal stem cell.

Osteoinduction was originally proposed by Friedenstein in the late 1960s as “the induction of undifferentiated inducible osteoprogenitor cells that are not yet committed to the osteogenic lineage to form osteoprogenitor cells” [42]. More recently, there has been generally accepted conclusive evidence that osteoinduction can only be claimed by ectopic bone formation in vivo, or in other words, “bone formation in the tissues or organs where bone does not naturally grow” [43,44]. Therefore, the term ‘osteoinductive small molecule’ would be used when the small molecule gives rise to ectopic bone formation after implantation in an animal. Preferably, the implantation site is subcutaneous or intramuscular [3,4547].

Statins are classified as osteoinductive small molecules because they were able to induce ectopic bone formation in a rat subcutaneous implantation model [48]. To date, statins are the most studied small molecules for bone regeneration because a number of studies have robustly demonstrated their bone regenerative capability in various cell and animal models. For instance, studies showed that cerivastatin, fluvastatin, lovastatin and simvastatin induced osteogenesis in vitro and in vivo by increasing the expression of BMP-2 through the BMP/Smad signaling mechanism [4957]. It is also worth noting that statins are the FDA-approved drugs for treating cardiovascular disease and they have been safely used in patients for decades [58]. This facilitated entering statins into clinical studies as a new bone regenerative therapeutic for treating various bone disorders [59].

Similar to statins, several small molecules have been discovered with osteogenic activity through the BMP/Smad signaling mechanism [20]. These include a helioxanthin derivative (TH) [60], tacrolimus hydrate (FK506) [6165], icariin [6668], rapamycin [69,70] and phenamil [71]. TH and icariin promoted osteoblastic differentiation in mouse osteoblast-like MC3T3-E1 cells, mouse primary osteoblasts and mouse embryonic stem cells [60,66]. TH’s in vivo osteogenic potential was further evaluated by implanting a sustained-release carrier containing TH into mouse calvarial defects, where new bone formation occurred within 4 weeks [72]. FK506 and rapamycin are immunosuppressants that have been used for reducing the activity of patients’ immune systems after organ transplantations in order to lower the risk of organ rejections [7375]. Intriguingly, these two small molecules were recently found to be osteoinductive, as evidenced by the ectopic bone formation in subcutaneous sites [76,77]. Phenamil is a derivative of the diuretic amiloride, which has been recently shown to induce osteogenesis of mouse mesenchymal stem cells (MSCs) in vitro [71]. However, in vivo data were not reported. Interestingly, the small molecule phenamil activates the BMP/Smad signaling pathway through the induction of Trb3 [71]. Trb3 is a novel inducer of the BMP signaling pathway that acts by downregulating the BMP antagonist Smurf1 [78]. The expression of Trb3 will thus stabilize the Smad family signaling transducers, which in turn induce the osteoblast-associated gene transcriptions [71]. This study also suggested that Trb3 might represent a new therapeutic target for bone repair and regeneration.

6-Bnz-cAMP is a PKA-specific cAMP analog that is well-validated as a molecular tool to study signaling mediated exclusively by PKA signaling [79]. Our group recently demonstrated that stimulation of the PKA signaling pathway by administrating the small molecule 6-Bnz-cAMP continuously resulted in vitro osteogenesis in several cell types such as mouse osteoblast-like MC3T3-E1 cells [23] and human MSCs (Figure 2). We further demonstrated that phosphorylation of CREB by PKA is involved in 6-Bnz-cAMP-induced osteogenesis [23]. We are now studying in vivo bone regeneration by 6-Bnz-cAMP in animals including rats and rabbits. Consistent with our observation of 6-Bnz-cAMP, Miyamoto et al. demonstrated that the phosphodiesterase type 4 small molecule inhibitor XT-611 potentiated the osteoblastic differentiation of rodent bone marrow cells through the cAMP/PKA signaling pathway [80]. Similarly, recent data from the Babij group at Amgen Inc. suggested that the CREB pathway activity is involved in the differentiation of the MC3T3-E1 cell line in response to their osteogenic small molecules (i.e., AMG0892 [benzothiazole core] and AMG0309 [naphthyl amide core]) [81].

Figure 2. 6-Bnz-cAMP at 100 μM promoted matrix calcification at day 20 of human mesenchymal stem cells on a biodegradable polymeric scaffold of poly(lactic-co-glycolic) acid thin films.

Figure 2

Briefly, for the qualitative determination of calcified matrix mineralization, at day 20, media were removed and cells were rinsed with calcium-free phosphate-buffered saline three-times. The calcium level was then measured with a calcium assay kit (Calcium LiquiColor®, Stanbio Laboratory, TX, USA) according to the manufacturer’s instructions [23]. Cells cultured in osteogenic medium served as a positive control, while cells cultured in control medium served as the baseline. To facilitate the comparison of different experimental settings, the ‘control’ values were set to one relative unit. *Statistical analysis was performed using the Student’s t-test at a significance of p < 0.05.

The small molecule purmorphamine, a purine derivative, has been shown by the Schultz and the Rosa groups to induce in vitro osteogenesis in mouse embryonic mesoderm fibroblasts and human osteoblasts [82,83]. The mechanism study of purmorphamine demonstrated that it functions as an agonist of Smoothened, a 7-transmembrane receptor of the Hh signaling cascade [84,85]. The Rosa group also investigated the mechanism of the previously shown osteogenic small molecule TAK-778 [8688], a derivative of ipriflavone, and their studies revealed that small molecule TAK-778 promoted osteoblast differentiation of human bone marrow cells via an estrogen receptor-dependent signaling pathway [89].

It is a well-known fact that the Wnt signaling cascade plays a key role in osteogenesis by directly stimulating the expression of Runx2, an essential transcription factor for the regulation of osteoblast differentiation [40,9094]. The small molecule inhibitor diphenylsulfonyl sulfonamide (WAY-316606) of the Wnt antagonist sFRP1 has been shown to activate the Wnt signaling pathway in order to enhance bone formation in an ex vivo organ culture assay [9597]. While various signaling pathways have been implicated in the regulation of osteoblast differentiation, several recent studies attempted to explore the possibility of integrating multiple osteogenic signaling pathways in osteoblast differentiation [98,99]. For instance, the small molecule oxysterols, cholesterol oxidation products, have been reported to execute their osteogenesis activity by activating both Hh and noncanonical Wnt signaling cascades [100], suggesting that signaling mechanism cross-talk between Wnt and Hh is important in osteogenesis.

It is important to point out that a large number of small molecules with osteogenic potential have been recently identified with unknown underlying signaling mechanisms. Table 3 summarizes these osteogenic small molecules. The Burdick group at the University of Pennsylvania (PA, USA) utilized a high-throughput screening of a small molecule library, and their results revealed that small molecule inducers encompass a wide range of medications including antibiotics (e.g., cephalexin), pain relievers (e.g., aspirin), psychotropic drugs (e.g., zolpidem) and steroids (e.g., cholecalciferol) [101]. Similarly, Chung’s group at the University of Tokyo (Japan) developed a high-throughput screen system for osteogenic drugs and identified a novel osteogenic small molecule: glabrisoflavone (isoflavone derivative) [102]. Deng’s group in China found a potent osteogenic small molecule (OIC-A006) using a high-throughput screen technology and demonstrated its osteoinductive potential in vitro and in vivo [103]. The signaling mechanisms underlying these osteogenic small molecules are currently unknown and await further investigation. It is also interesting to note that a number of research groups recently examined small molecule components extracted from herbal medicine products and claimed osteogenic effects in different cell and animal models. These osteogenic small molecules are summarized in Tables 2 & 3. Importantly, these interesting observations open a new area in bone regenerative medicine by introducing herbal medicines, including traditional Chinese medicine.

Table 3.

Small molecules that promote osteoblast differentiation with unknown signaling targets.

Small molecule In vitro effective dosage (μM) In vitro activity (cell types) In vivo activity Ref.
Antibiotics or antivirals: cephalexin, doxycycline hydrochloride, gentamicin sulfate, minocycline hydrochloride, valacyclovir hydrochloride, chloroguanide hydrochloride Pain relievers: aspirin, acetanilide, antipyrine, nalbuphine hydrochloride
Steroids: cholecalciferol, fludrocortisone acetate, fluorometholone, halcinonide, medrysone, triamcinolone diacetate, medroxyprogesterone acetate
Psychotropic drugs: zolpidem, spiperone, olanzapine, inositol, isopropamide iodide, clopamide
Miscellaneous: avobenzone, aminohippuric acid, tolaxamide, protoveratrine A, ropinirole tolaxamide, trimebutine maleate, ebselen, tuaminoheptane sulfate
0.1 Human MSCs Not reported [101]
Decalpenic acid 1 C3H10T1/2 Not reported [160]
Glabrisoflavone 30 MC3T3-E1; mouse primary osteoblasts Not reported [102]
β-cryptoxanthin 10 MC3T3-E1 Not reported [161]
Prostaglandins 0.01–10 Adipose tissue-derived MSCs Systemic treatment of ovariectomized rats [162,163]
OIC-A006 6.25 Bone marrow stromal cells Skull defect model in rabbit [103]
Herbal medicines
Naringin 0.1 UMR 106; osteoblastic cells Systemic treatment of ovariectomized mice [164,165]
Quercetin 5 Human adipose stromal cells Calvarial parietal bone defect model in rabbit [166,167]

MSC: Mescenchymal stem cell.

Osteogenic/osteoinductive small molecules with the potential to directly promote angiogenesis and/or inhibit osteoclast function

Successful approaches to bone regeneration and bone graft repair also require other phenomena such as neovascularization and osteoclastic bone resorption. Neovascularization is particularly important because this facilitates the transport of oxygen and nutrients to cells transplanted in the bone grafts, as well as migration from the host tissue [104]. Insufficient neovascularization of implanted bone grafts often impedes recovery and may lead to hypoxia-induced cell death [29]. Several protein growth factors have been investigated to promote angiogenesis during bone repair and regeneration [21]. For instance, VEGF has been extensively researched for this purpose [105]. While promising, as discussed in the previous section, protein-based therapeutic strategies are generally limited in their universal use (Table 1) [15,24,25,106]. The Botchwey group has proposed that small molecules with angiogenic potential would serve as options to combat the issues with protein-based factors [29,106,107].

Osteoclasts are well known for bone resorption, which is associated with many bone-related disorders such as osteoporosis. Therapeutic strategies have targeted this cell type for developing antiresorptive agents [15]. A large number of small molecule drugs have been discovered and developed with the capability of inhibiting osteoclast activity [15]. For instance, bisphosphonate (BPP) is a synthetic small molecule that is one of the major antiresorptive drugs currently available on the market to treat osteoporosis and related bone diseases [108]. However, similar to most anti-resorptive drugs, BPP may not meet the demand for osteoporosis because it is unable to promote the regeneration of damaged osteoporotic bone tissue. In order to regenerate and repair bone efficiently, novel therapeutic approaches should be developed in order to promote osteogenesis, promote angiogenesis and inhibit osteoclastogenesis. More specifically, multifunctional small molecules, in which one functionality is capable of inducing osteogenesis and angiogenesis while another functionality is capable of inhibiting osteoclast formation, should be investigated. In this review, in addition to focusing on osteo-inductive potential, we also summarized a number of osteogenic/osteoinductive small molecules with the potential to promote angiogenesis (Table 4) and/or inhibit osteoclast function (Table 5). According to Tables 25, it is interesting to note that statins, icariin and prostaglandins are the small molecules that contain all three properties that are important for bone regeneration.

Table 4.

Osteogenic/osteoinductive small molecules that directly promote angiogenesis.

Small molecule Study summary Ref.
Statins Both in vitro and in vivo angiogenesis were enhanced at low concentrations of statins (0.005–0.01 μmol/l). Pravastatin at 0.01 μM induced proangiogenic effects via the PI3K/Akt pathway [168170]
Icariin Icariin stimulated in vitro angiogenesis by activating the MEK/ERK- and PI3K/Akt/eNOS-dependent signal pathways in human endothelial cells [171]
Purmorphamine Purmorphamine induced vascular lumen formation in vitro in the coculture of hMSCs and human umbilical vein endothelial cells [172]
Puerarin Puerarin (120 mg/kg) induced angiogenesis in the nonischemic and ischemic myocardium of rats [173]
Prostaglandins Prostaglandin induced angiogenesis in the myocardium of patients with ischemic heart disease [174]
Olanzapine Olanzapine treatment increased VEGF levels and angiogenesis in the rat hippocampus [175]

hMSC: Human mesenchymal stem cell.

Table 5.

Osteogenic/osteoinductive small molecules that inhibit osteoclast function.

Small molecule Study summary Ref.
Icariin Icariin (10 μM) inhibited the formation and bone resorption activity of osteoclasts in a mouse bone marrow culture [176]
XT-611 XT-611 inhibited osteoclast formation in coculture of mouse bone marrow stromal cell line ST2 and adherent cell-depleted BMCs [177]
Statins Cerivastatin inhibited parathyroid hormone-stimulated bone resorption in vitro. Simvastatin inhibited osteoclastogenesis through suppression of ROS-mediated signaling pathways [178,179]
FK506 FK506 induced osteoclast apoptosis in mouse BMCs [180]
Rapamycin Rapamycin inhibited osteoclast formation and bone resorption in giant cell tumors of bone [181]
Osthole Osthole inhibited bone resorption by decreasing the formation, differentiation and TRAP activity of osteoclasts derived from rat marrow cells [182]
Berberine Berberine inhibited osteoclast formation and survival through suppression of NF-κB and Akt activation [183]
Resveratrol Resveratrol inhibited osteoclastogenesis through suppression of ROS-mediated signaling pathways [184]
Prostaglandins Prostaglandin E2 inhibited human osteoclast formation [185]
Quercetin Quercetin (0.1–10 μM) decreased osteoclastogenesis via a mechanism involving NF-κB and AP-1 [186]
Naringin Naringin inhibited osteoclastogenesis and bone resorption via the inhibition of RANKL-induced NF-κB and ERK activation [187]
β-cryptoxanthin β-cryptoxanthin induced osteoclast apoptosis and suppressed cell function in osteoclastic cells [188]

BMC: Bone marrow cell; ROS: Reactive oxygen species.

Future perspective & challenges

Strategies employing protein-based growth factors have been successfully used in bone repair and regeneration; however, as discussed, they have had a number of drawbacks. The incidence of skeletal complications will continue to grow as the life expectancy of the population increases. Therefore, there is a clear need for an alternative, yet effective, therapeutic strategy. It is believed that using bone regenerative small molecules is an attractive alternative strategy. In addition, recent advances in phenotypic high-throughput screens of small molecule libraries play a remarkable role in accelerating the process of drug discovery. It is expected that the value of small molecules for musculoskeletal complications over the next 5–10 years is enormous.

While promising, it is important to point out that a common issue with small molecule drugs is their off-target cell effects (Table 1) [24]. To overcome the aforementioned issues, small molecules with osteoinductive potential can be delivered to bone defect sites locally via controlled release systems [24,109]. Biodegradable polymeric-based matrix systems have already shown great promise as novel drug delivery vehicles in the controlled delivery of vaccines, chemotherapeutics and various growth factors [24,110115]. In fact, research studies have shown that small molecules can be easily incorporated into biodegradable polymer systems by a variety of methods for bone regenerative engineering applications [29,116,117]. Therefore, polymeric controlled release systems have great potential to be utilized in the delivery of osteoinductive small molecules for treating musculoskeletal complications through bone regenerative engineering methods [24,118]. In addition, it is worth noting that despite the significant evidence of potential for bone regeneration in various cell types and animal models, further investigation will be needed to define variables such as dosage, specificity and general toxicity.

Specificity & dosage

Although oral administration is feasible and effective for many small molecule drugs due to their ease of absorption into the bloodstream by the digestive system, the bioavailability of small molecule drugs is low for bone tissue, thus the same effectiveness is not expected for treating bone disorders [119]. Interestingly, specificity and effectiveness can be also achieved by delivering small molecule drugs locally to the bone site via a controllable, sustained-release polymeric-based delivery vehicle as mentioned above [120]. Such local delivery systems are advantageous for two main reasons: they reduce systemic side effects normally caused by supraphysiological dose requirements and increase therapeutic effectiveness by providing targeted delivery and improved drug stability against chemical/ enzymatic degradation [121]. Additionally, delivery vehicles can be engineered so they not only provide a matrix for local drug delivery, but also a physical substrate for cell attachment and growth. Particularly, polymeric biodegradable scaffolds have the capability to provide such support while releasing loaded agents through hydrolytic degradation of the matrix into non-toxic components. Once released, drug delivery is typically mediated by passive diffusion, but also impeded by renal clearance [122]. As depicted in Figure 3, there is an extended therapeutic window of drug efficiency when delivered from a delivery vehicle, as opposed to more traditionally systemic methods of delivery [123]. Despite the many advantages, local delivery carriers have some disadvantages. For instance, dosage cannot be adjusted after injection or implantation, and the rate of drug release typically decreases with time. Furthermore, repeated implantation may be required for long-term release and the implantation surgery is invasive [124]. While systemic drug delivery can be advantageous because repeated administration allows for dosage adjustments, the possible toxic effects due to high systemic exposure to the drug are avoidable. Since both drug delivery systems have advantages and disadvantages, the selection of the appropriate drug delivery carrier is dependent on the nature and location of the disease. However, in this section, we will focus on local delivery methods for the treatment of musculoskeletal injuries and diseases.

Figure 3. Controlled release strategies (solid line) allow delivered drugs to stay within the ‘therapeutic window’ for a longer time in contrast to systemic, one-off delivery (dashed line).

Figure 3

The extended period of time (teff) of controlled release strategies not only permits the drug to diffuse from the scaffold and be present in the tissue at a more sustained rate, but also avoids being present in high concentrations that could be toxic to the local environment.

teff: Effective time.

Adapted with permission from [123].

There are many delivery vehicle systems that have been investigated in order to achieve local delivery. For example, a recent report suggested the use of an injectable hydrogel drug delivery system to deliver statins (e.g., fluvastatin) for bone regeneration [119]. Hydrogels are cross-liked networks of insoluble hydrophilic polymers that can absorb large amounts of water. While their soft and rubbery consistency makes these constructs attractive for soft tissue applications [125], hydrogels have also been used for bone tissue engineering [126]. Small molecules entrapped within the polymeric chains of the porous matrix are distributed heterogeneously throughout the biomaterial, and as the hydrogel degrades, they diffuse from the gel to the surrounding environment [127]. As previously reviewed, several natural and synthetic polymers are used to fabricate hydrogels, and these exhibit many different properties such as charge, mechanical and structural features and physical structures, among others [128]. One large advantage of hydrogels is their ability to form matrices in situ, thus overcoming the aforementioned issue of intrusive implantation surgery. Such injectable hydrogels can gel via thermoresponsiveness, photopolymerization and other environmental stimuli [129]. On the other hand, hydrogels can be disadvantageous because they lack the physical integrity to bear heavy mechanical loads, and can exhibit rapid release profiles of drugs without physical and/or chemical modifications [126].

Another biomaterial frequently used for bone regeneration is calcium phosphate (CaP). Researchers have been recently investigating CaP matrices as targeted drug-delivery vehicles to deliver small molecule BPP specifically to osteoporotic bone tissue [130132]. CaP delivery vehicles of various forms (ceramics, cements and composite coatings) have been investigated extensively in bone tissue engineering due to their marked similarities in mineral composition, properties and microarchitecture to human cancellous bone. Further basis is on the strong and specific chelation between BPP and CaP crystals. There are three main categories of CaP materials with varying chemical and thus physical properties: hydroxyapatite (HAp), β-tricalcium phosphate and biphasic CaP (combination of β-tricalcium phosphate and HAp). Of these categories, HAp is highly attractive for BPP encapsulation and release since its high affinity for BPP does not hamper BPP pharmacological ability [133]. On the other hand, BPP incorporated in CaP cements are advantageous because cements can harden in situ, easily matching the shape of bone cavities or defects [134]. Pristine or combinatorial uses of these biomaterials with natural or synthetic biodegradable polymers result in a wide range of applications with varying mechanical strength, bioresorbabilty and osteoconductivity.

Another popular class of local drug carriers is biodegradable synthetic polymers. Several types of polymeric scaffolds are used extensively in bone tissue engineering due to their biocompatibility, hydrolytic biodegradability, formability and ease of use [135]. There are a number of ways to deliver small molecules from biodegradable polymers, but they are typically either physically encapsulated within the polymeric matrix or immobilized to the surface, in addition to undergoing ionic, hydrophobic and/or hydrogen bonding interactions with the polymer. Our group is currently engineering biodegradable poly(lactide-co-glycolic) acid (PLAGA)-based microsphere scaffolds encapsulated with 6-Bnz-cAMP for bone regenerative engineering applications. Other than its demonstrated biocompatibility, PLAGA has been shown to degrade into nontoxic components, and has thus been FDA approved for several clinical applications [25].

In addition to employing local delivery vehicles, lowering the dosages of the drugs can further minimize potential nonspecific side effects due to small molecule drugs; thus, the dose–response relationship of small molecules on cell and tissue cultures should be evaluated carefully in order to determine the lowest possible effective concentration [136].

It should be noted that the osteogenic activities of many small molecules described here were studied in rodent models only. In the field of orthopedic research, the human MSC is the appropriate cell model to investigate the osteogenesis of a molecule or biomaterial because: they have demonstrated the ability to differentiate into the osteogenic lineages [101,137139]; they offer an alternative cell source for bone tissue engineering procedures that involves a less invasive harvest technique [140]; and it is believed that the use of human MSCs for studying osteogenesis will bring basic science concepts closer to clinical studies [34]. Thus, further studies are needed to investigate the interspecies differences of small molecules’ effects between rodents and humans in terms of osteoinductivity, dosages and pharmacokinetic properties. In addition, since a number of small molecules listed in this review were not tested for bone-forming activity in animals, further investigation using appropriate animal model is therefore needed to determine their in vivo bone-forming activity.

Many small molecules discussed in this review are primarily focused on osteoinductive potential. Further investigation is needed to determine whether the small molecules have the ability to promote neovascularization and to control bone remodeling, including osteoclastic resorption, because these two events are important to bone regeneration and bone graft repair success.

Small molecules are a useful tool for studying bone cell biology

The signaling mechanism underlying osteogenesis is still unclear in some cell types, including MSCs. Many small molecules described here could offer the unique opportunity to study osteogenic signaling pathways in cells and tissues because they can be utilized as molecular probes. For instance, cAMP analogs have been developed for decades and routinely used as biological tools for investigating various cAMP-mediated signal transduction pathways [23,33,79]. Therefore, the focus of the small molecule research here is not only on the development of new bone regenerative medicine, but also the development of an improved understanding of the mechanistic pathways that drive regeneration of cells and tissues [71].

Executive summary.

Bone growth factors

  • Bone growth factors are usually polypeptide-based macromolecules such as BMP, FGF or IGF.

  • Protein-based therapeutic agents usually carry a number of limitations including protein instability, high cost, contamination and immunogenicity.

  • Small molecules with the ability to induce bone formation have recently gained a more prominent reputation as therapeutics because their inherent properties can minimize the limitations observed with protein growth factors.

Small molecules with bone regenerative potential

  • In general, small molecules are low molecular weight organic compounds that can freely diffuse across the cell membrane and trigger a signal transduction system by activating a specific intracellullar protein sensor.

  • Identification of the small molecule that modulates the activity of the osteogenic signaling pathways (e.g., cAMP/PKA, BMP/Smad or Wnt) is the current drug discovery strategy for bone regenerative medicine.

  • A large number of small molecules potentially with bone regeneration ability have been discovered in the last decade.

Future perspective

  • There exists the potential for treatment of various musculoskeletal defects and disorders by combining osteoinductive small molecules with scaffold-based bone regenerative engineering.

  • Future investigation will be needed to define the dosage, specificity, general toxicity and in vivo activity of small molecule drugs.

  • It would be of great interest to explore whether the small molecules also have the ability to promote neovascularization and inhibit osteoclastogenesis.

Acknowledgments

The authors would like to thank C Nelson for comments on this manuscript.

Footnotes

Financial & competing interests disclosure

CT Laurencin was the recipient of a Presidential Faculty Fellowship Award from the National Science Foundation. KW-H Lo thanks the JoAnne Smith, MD Research and Education Foundation for their funded support of his research. The authors gratefully acknowledge funding from NSF-EFRI 0736002 and NIH-R21AR060480. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

▪ of interest

▪ ▪ of considerable interest

  • 1▪.Lieberman JR, Daluiski A, Einhorn TA. The role of growth factors in the repair of bone. Biology and clinical applications. J Bone Joint Surg Am. 2002;84-A(6):1032–1044. doi: 10.2106/00004623-200206000-00022. Provides a comprehensive look at various protein-based growth factors for bone repair and potential clinical applications. [DOI] [PubMed] [Google Scholar]
  • 2.Wang EA, Rosen V, D’Alessandro JS, et al. Recombinant human bone morphogenetic protein induces bone formation. Proc Natl Acad Sci USA. 1990;87(6):2220–2224. doi: 10.1073/pnas.87.6.2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Urist MR. Bone: formation by autoinduction. Science. 1965;150(698):893–899. doi: 10.1126/science.150.3698.893. [DOI] [PubMed] [Google Scholar]
  • 4.White AP, Vaccaro AR, Hall JA, Whang PG, Friel BC, McKee MD. Clinical applications of BMP-7/OP-1 in fractures, nonunions and spinal fusion. Int Orthop. 2007;31(6):735–741. doi: 10.1007/s00264-007-0422-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.McKay WF, Peckham SM, Badura JM. A comprehensive clinical review of recombinant human bone morphogenetic protein-2 (INFUSE Bone Graft) Int Orthop. 2007;31(6):729–734. doi: 10.1007/s00264-007-0418-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from laboratory to clinic, part II (BMP delivery) J Tissue Eng Regen Med. 2008;2(2–3):81–96. doi: 10.1002/term.74. [DOI] [PubMed] [Google Scholar]
  • 7.Rubin MR, Bilezikian JP. The anabolic effects of parathyroid hormone therapy. Clin Geriatr Med. 2003;19(2):415–432. doi: 10.1016/s0749-0690(02)00074-5. [DOI] [PubMed] [Google Scholar]
  • 8.Deal C, Gideon J. Recombinant human PTH 1–34 (Forteo): an anabolic drug for osteoporosis. Cleve Clin J Med. 2003;70(7):585–586. 589–590, 592–594. doi: 10.3949/ccjm.70.7.585. passim. [DOI] [PubMed] [Google Scholar]
  • 9.Holick MF. PTH (1–34): a novel anabolic drug for the treatment of osteoporosis. South Med J. 2005;98(11):1114–1117. doi: 10.1097/01.smj.0000140864.78760.8d. [DOI] [PubMed] [Google Scholar]
  • 10.Garrett IR. Anabolic agents and the bone morphogenetic protein pathway. Curr Top Dev Biol. 2007;78:127–171. doi: 10.1016/S0070-2153(06)78004-8. [DOI] [PubMed] [Google Scholar]
  • 11.Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from the laboratory to the clinic, part I (basic concepts) J Tissue Eng Regen Med. 2008;2(1):1–13. doi: 10.1002/term.63. [DOI] [PubMed] [Google Scholar]
  • 12.Dingermann T. Recombinant therapeutic proteins: production platforms and challenges. Biotechnol J. 2008;3(1):90–97. doi: 10.1002/biot.200700214. [DOI] [PubMed] [Google Scholar]
  • 13.Ratelade J, Miot MC, Johnson E, Betton JM, Mazodier P, Benaroudj N. Production of recombinant proteins in the lon-deficient BL21(DE3) strain of Escherichia coli in the absence of the DnaK chaperone. Appl Environ Microbiol. 2009;75(11):3803–3807. doi: 10.1128/AEM.00255-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schein CH. Production of soluble recombinant proteins in bacteria. Biotechnology. 1989;7:1141–1149. [Google Scholar]
  • 15.Egusa H, Saeki M, Doi M, et al. A small-molecule approach to bone regenerative medicine in dentistry. J Oral Biosci. 2010;52(2):107–118. [Google Scholar]
  • 16.Walker DH, Wright NM. Bone morphogenetic proteins and spinal fusion. Neurosurg Focus. 2002;13(6):e3. doi: 10.3171/foc.2002.13.6.4. [DOI] [PubMed] [Google Scholar]
  • 17.Hanley DA, Watson PH, Hodsman AB, Dempster DW. Pharmacological mechanisms of therapeutics: parathyroid hormone. Prin Bone Biol. 2008;2:1661–1695. [Google Scholar]
  • 18.Hwang CJ, Vaccaro AR, Lawrence JP, et al. Immunogenicity of bone morphogenetic proteins. J Neurosurg Spine. 2009;10(5):443–451. doi: 10.3171/2009.1.SPINE08473. [DOI] [PubMed] [Google Scholar]
  • 19.Gong Z, Zhou S, Cao J, Gu X. Effects of recombinant human basic fibroblast growth factor on cell proliferation during mandibular fracture healing in rabbits. Chin J Traumatol. 2001;4(2):110–112. [PubMed] [Google Scholar]
  • 20.Hollinger JO, Hart CE, Hirsch SN, Lynch S, Friedlaender GE. Recombinant human platelet-derived growth factor: biology and clinical applications. J Bone Joint Surg Am. 2008;90(Suppl 1):48–54. doi: 10.2106/JBJS.G.01231. [DOI] [PubMed] [Google Scholar]
  • 21.Devescovi V, Leonardi E, Ciapetti G, Cenni E. Growth factors in bone repair. Chir Organi Mov. 2008;92(3):161–168. doi: 10.1007/s12306-008-0064-1. [DOI] [PubMed] [Google Scholar]
  • 22.Ripamonti U, Ferretti C, Teare J, Blann L. Transforming growth factor-beta isoforms and the induction of bone formation: implications for reconstructive craniofacial surgery. J Craniofac Surg. 2009;20(5):1544–1555. doi: 10.1097/SCS.0b013e3181b09ca6. [DOI] [PubMed] [Google Scholar]
  • 23.Lo KW-H, Kan HM, Ashe KM, Laurencin CT. The small molecule PKA-selective cyclic AMP analogue as an inducer of osteoblast-like cells differentiation and mineralization. J Tissue Eng Regen Med. 2012;6:40–48. doi: 10.1002/term.395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lo KW, Ulery BD, Deng M, Ashe KM, Laurencin CT. Current patents on osteoinductive molecules for bone tissue engineering. Recent Patents Biomed Eng. 2012;4(3):153–167. [Google Scholar]
  • 25▪ ▪.Lo KW-H, Ulery BD, Ashe KM, Laurencin CT. Studies of bone morphogenetic protein based surgical repair. Adv Drug Deliv Rev. 2012 doi: 10.1016/j.addr.03.014. (Epub ahead of print). Provides the most updated information on the current development of bone morphogenetic proteins and their limitations. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Blaich G, Janssen B, Roth G, Salfeld J. Handbook of Pharmaceutical Biotechnology. John Wiley & Sons Inc; NJ, USA: 2006. Overview: differentiating issues in the development of macromolecules compared with small molecules; pp. 89–123. [Google Scholar]
  • 27.van de Weert M, Hennink WE, Jiskoot W. Protein instability in poly(lactic-co-glycolic acid) microparticles. Pharm Res. 2000;17(10):1159–1167. doi: 10.1023/a:1026498209874. [DOI] [PubMed] [Google Scholar]
  • 28.Fu K, Klibanov AM, Langer R. Protein stability in controlled-release systems. Nat Biotechnol. 2000;18(1):24–25. doi: 10.1038/71875. [DOI] [PubMed] [Google Scholar]
  • 29.Wieghaus KA, Capitosti SM, Anderson CR, et al. Small molecule inducers of angiogenesis for tissue engineering. Tissue Eng. 2006;12(7):1903–1913. doi: 10.1089/ten.2006.12.1903. [DOI] [PubMed] [Google Scholar]
  • 30.Pyeritz RE. A small molecule for a large disease. N Engl J Med. 2008;358(26):2829–2831. doi: 10.1056/NEJMe0804008. [DOI] [PubMed] [Google Scholar]
  • 31.Le GT, Abbenante G. Inhibitors of TACE and caspase-1 as anti-inflammatory drugs. Curr Med Chem. 2005;12(25):2963–2977. doi: 10.2174/092986705774462851. [DOI] [PubMed] [Google Scholar]
  • 32.Carroll SF. Discovery and Nonclinical Development. Springer; NY, USA: 2007. pp. 84–106. [Google Scholar]
  • 33.Lo KWH, Ashe KM, Kan HM, Lee DA, Laurencin CT. Activation of cyclic AMP/ protein kinase A signaling pathway enhances osteoblast cell adhesion on biomaterials for regenerative engineering. J Orthop Res. 2011;29(4):602–608. doi: 10.1002/jor.21276. [DOI] [PubMed] [Google Scholar]
  • 34.Siddappa R, Doorn J, Liu J, et al. Timing, rather than the concentration of cyclic AMP, correlates to osteogenic differentiation of human mesenchymal stem cells. J Tissue Eng Regen Med. 2010;4(5):356–365. doi: 10.1002/term.246. [DOI] [PubMed] [Google Scholar]
  • 35.Kinto N, Iwamoto M, Enomoto-Iwamoto M, et al. Fibroblasts expressing sonic hedgehog induce osteoblast differentiation and ectopic bone formation. FEBS Lett. 1997;404(2–3):319–323. doi: 10.1016/s0014-5793(97)00014-8. [DOI] [PubMed] [Google Scholar]
  • 36.Nakamura T, Aikawa T, Iwamoto-Enomoto M, et al. Induction of osteogenic differentiation by hedgehog proteins. Biochem Biophys Res Commun. 1997;237(2):465–469. doi: 10.1006/bbrc.1997.7156. [DOI] [PubMed] [Google Scholar]
  • 37.Zwijsen A, Verschueren K, Huylebroeck D. New intracellular components of bone morphogenetic protein/Smad signaling cascades. FEBS Lett. 2003;546(1):133–139. doi: 10.1016/s0014-5793(03)00566-0. [DOI] [PubMed] [Google Scholar]
  • 38.Hu Y, Chan E, Wang SX, Li B. Activation of p38 mitogen-activated protein kinase is required for osteoblast differentiation. Endocrinology. 2003;144(5):2068–2074. doi: 10.1210/en.2002-220863. [DOI] [PubMed] [Google Scholar]
  • 39.Lin FH, Chang JB, Brigman BE. Role of mitogen-activated protein kinase in osteoblast differentiation. J Orthop Res. 2011;29(2):204–210. doi: 10.1002/jor.21222. [DOI] [PubMed] [Google Scholar]
  • 40.Gaur T, Lengner CJ, Hovhannisyan H, et al. Canonical Wnt signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005;280(39):33132–33140. doi: 10.1074/jbc.M500608200. [DOI] [PubMed] [Google Scholar]
  • 41.Liu G, Vijayakumar S, Grumolato L, et al. Canonical Wnts function as potent regulators of osteogenesis by human mesenchymal stem cells. J Cell Biol. 2009;185(1):67–75. doi: 10.1083/jcb.200810137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Friedenstein AY. Induction of bone tissue by transitional epithelium. Clin Orthop Relat Res. 1968;59:21–37. [PubMed] [Google Scholar]
  • 43.Wilson-Hench J. Osteoinduction. In: Williams D, editor. Progress in Biomedical Engineering: Definitions in Biomaterials. Elsevier; The Netherlands: 1987. p. 29. [Google Scholar]
  • 44.Barradas AM, Yuan H, van Blitterswijk CA, Habibovic P. Osteoinductive biomaterials: current knowledge of properties, experimental models and biological mechanisms. Eur Cell Mater. 2011;21:407–429. doi: 10.22203/ecm.v021a31. discussion 429. [DOI] [PubMed] [Google Scholar]
  • 45.Bentz H, Nathan RM, Rosen DM, et al. Purification and characterization of a unique osteoinductive factor from bovine bone. J Biol Chem. 1989;264:20805–20810. [PubMed] [Google Scholar]
  • 46.Yoshida K, Bessho K, Fujimura K, et al. Osteoinduction capability of recombinant human bone morphogenetic protein-2 in intramuscular and subcutaneous sites: an experimental study. J Craniomaxillofac Surg. 1998;26(2):112–115. doi: 10.1016/s1010-5182(98)80050-4. [DOI] [PubMed] [Google Scholar]
  • 47.Habibovic P, Sees TM, van den Doel MA, van Blitterswijk CA, de Groot K. Osteoinduction by biomaterials – physicochemical and structural influences. J Biomed Mater Res A. 2006;77(4):747–762. doi: 10.1002/jbm.a.30712. [DOI] [PubMed] [Google Scholar]
  • 48.Houshmand B, Hassanizade R, Eslami B, et al. Simuvastatin and lovastatin induce ectopic bone formation in rat subcutaneous tissue. J Periodontol Implant Dent. 2011;2(1):12–16. [Google Scholar]
  • 49.Mundy G, Garrett R, Harris S, et al. Stimulation of bone formation in vitro and in rodents by statins. Science. 1999;286(5446):1946–1949. doi: 10.1126/science.286.5446.1946. [DOI] [PubMed] [Google Scholar]
  • 50.Sugiyama M, Kodama T, Konishi K, Abe K, Asami S, Oikawa S. Compactin and simvastatin, but not pravastatin, induce bone morphogenetic protein-2 in human osteosarcoma cells. Biochem Biophys Res Commun. 2000;271(3):688–692. doi: 10.1006/bbrc.2000.2697. [DOI] [PubMed] [Google Scholar]
  • 51.Gutierrez G, Garrett IR, Rossini G. Dermal application of lovastatin to rats causes greater increases in bone formation and plasma concentrations that when administered by oral gavage. J Bone Miner Res. 2000;15:S427. [Google Scholar]
  • 52.Whang K, Zhao M, Qiao M, et al. Administration of lovastatin locally in low doses in a novel delivery system induces prolonged bone formation. J Bone Miner Res. 2000;15:S225. [Google Scholar]
  • 53.Maeda T, Matsunuma A, Kawane T, Horiuchi N. Simvastatin promotes osteoblast differentiation and mineralization in MC3T3-E1 cells. Biochem Biophys Res Commun. 2001;280(3):874–877. doi: 10.1006/bbrc.2000.4232. [DOI] [PubMed] [Google Scholar]
  • 54.Garrett IR, Gutierrez G, Mundy GR. Statins and bone formation. Curr Pharm Des. 2001;7(8):715–736. doi: 10.2174/1381612013397762. [DOI] [PubMed] [Google Scholar]
  • 55.Maritz FJ, Conradie MM, Hulley PA, Gopal R, Hough S. Effect of statins on bone mineral density and bone histomorphometry in rodents. Arterioscler Thromb Vasc Biol. 2001;21(10):1636–1641. doi: 10.1161/hq1001.097781. [DOI] [PubMed] [Google Scholar]
  • 56.Edwards CJ, Spector TD. Statins as modulators of bone formation. Arthritis Res. 2002;4(3):151–153. doi: 10.1186/ar399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chen PY, Sun JS, Tsuang YH, Chen MH, Weng PW, Lin FH. Simvastatin promotes osteoblast viability and differentiation via Ras/Smad/Erk/BMP-2 signaling pathway. Nutr Res. 2010;30(3):191–199. doi: 10.1016/j.nutres.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 58.Eichler K, Puhan MA, Bachmann LM. The role of statins in primary prevention of cardiovascular disease. Arch Intern Med. 2007;167(10):1100. doi: 10.1001/archinte.167.10.1100-a. author reply 1100–1101. [DOI] [PubMed] [Google Scholar]
  • 59.Cushenberry L, Debittner MR. Potential use of HMG-CoA reductase inhibitors for osteoporosis. Ann Pharmacother. 2002;36:671–678. doi: 10.1345/aph.1A072. [DOI] [PubMed] [Google Scholar]
  • 60.Ohba S, Nakajima K, Komiyama Y, et al. A novel osteogenic helioxanthin-derivative acts in a BMP-dependent manner. Biochem Biophys Res Commun. 2007;357(4):854–860. doi: 10.1016/j.bbrc.2007.03.173. [DOI] [PubMed] [Google Scholar]
  • 61.Yoshikawa T, Nakajima H, Yamada E, et al. In vivo osteogeneic capability of cultured allogeneic bone in porous hydroxvapatite: immunosuppressive and osteogeneic potential of FK506 in vivo. J Bone Miner Res. 2000;15:1147–1157. doi: 10.1359/jbmr.2000.15.6.1147. [DOI] [PubMed] [Google Scholar]
  • 62.Voggenreiter G, Assenmacher S, Kreuzfelder E, et al. Immunosuppression with FK506 increases bone induction in demineralized isogeneic and xenogeneic bone matrix in the rat. J Bone Miner Res. 2000;15(9):1825–1834. doi: 10.1359/jbmr.2000.15.9.1825. [DOI] [PubMed] [Google Scholar]
  • 63.Tang L, Ebara S, Kawasaki S, Wakabayashi S, Nikaido T, Takaoka K. FK506 enhanced osteoblastic differentiation in mesenchymal cells. Cell Biol Int. 2002;26(1):75–84. doi: 10.1006/cbir.2001.0812. [DOI] [PubMed] [Google Scholar]
  • 64.Kugimiya F, Yano F, Ohba S, et al. Mechanism of osteogenic induction by FK506 via BMP/ Smad pathways. Biochem Biophys Res Commun. 2005;338(2):872–879. doi: 10.1016/j.bbrc.2005.10.024. [DOI] [PubMed] [Google Scholar]
  • 65.Yoshikawa T, Nakajima H, Uemura T, et al. In vitro bone formation induced by immunosuppressive agent tacrolimus hydrate (FK506) Tissue Eng. 2005;11(3–4):609–617. doi: 10.1089/ten.2005.11.609. [DOI] [PubMed] [Google Scholar]
  • 66.Zhao J, Ohba S, Shinkai M, Chung UI, Nagamune T. Icariin induces osteogenic differentiation in vitro in a BMP- and Runx2-dependent manner. Biochem Biophys Res Commun. 2008;369(2):444–448. doi: 10.1016/j.bbrc.2008.02.054. [DOI] [PubMed] [Google Scholar]
  • 67.Zhao J, Ohba S, Komiyama Y, Shinkai M, Chung UI, Nagamune T. Icariin: a potential osteoinductive compound for bone tissue engineering. Tissue Eng Part A. 2010;16(1):233–243. doi: 10.1089/ten.TEA.2009.0165. [DOI] [PubMed] [Google Scholar]
  • 68.Wei H, Zili L, Yuanlu C, et al. Effect of icariin on bone formation during distraction osteogenesis in the rabbit mandible. Int J Oral Maxillofac Surg. 2011;40(4):413–418. doi: 10.1016/j.ijom.2010.10.015. [DOI] [PubMed] [Google Scholar]
  • 69.Ogawa T, Tokuda M, Tomizawa K, et al. Osteoblastic differentiation is enhanced by rapamycin in rat osteoblast like osteosarcoma (ROS 17/2.8) cells. Biochem Biophys Res Commun. 1998;249:226–230. doi: 10.1006/bbrc.1998.9118. [DOI] [PubMed] [Google Scholar]
  • 70.Lee KW, Yook JY, Son MY, et al. Rapamycin promotes the osteoblastic differentiation of human embryonic stem cells by blocking the mTOR pathway and stimulating the BMP/Smad pathway. Stem Cells Dev. 2010;19(4):557–568. doi: 10.1089/scd.2009.0147. [DOI] [PubMed] [Google Scholar]
  • 71.Park KW, Waki H, Kim WK. The small molecule phenamil induces osteoblast differentiation and mineralization. Mol Cell Biol. 2009;29(14):3905–3914. doi: 10.1128/MCB.00002-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nakajima K, Komiyama Y, Hojo H, et al. Enhancement of bone formation ex vivo and in vivo by a helioxanthin-derivative. Biochem Biophys Res Commun. 2010;395(4):502–508. doi: 10.1016/j.bbrc.2010.04.041. [DOI] [PubMed] [Google Scholar]
  • 73.Sawada S, Suzuki G, Kawase Y, Takaku F. Novel immunosuppressive agent, FK506. In vitro effects on the cloned T cell activation. J Immunol. 1987;139(6):1797–1803. [PubMed] [Google Scholar]
  • 74.Thomson AW. FK-506 – how much potential? Immunol Today. 1989;10(1):6–9. doi: 10.1016/0167-5699(89)90057-1. [DOI] [PubMed] [Google Scholar]
  • 75.Chang JY, Sehgal SN, Bansbach CC. FK506 and rapamycin: novel pharmacological probes of the immune response. Trends Pharmacol Sci. 1991;12(6):218–223. doi: 10.1016/0165-6147(91)90555-7. [DOI] [PubMed] [Google Scholar]
  • 76.Kotobuki N, Katsube Y, Katou Y, Tadokoro M, Hirose M, Ohgushi H. In vivo survival and osteogenic differentiation of allogeneic rat bone marrow mesenchymal stem cells (MSCs) Cell Transplant. 2008;17(6):705–712. doi: 10.3727/096368908786092793. [DOI] [PubMed] [Google Scholar]
  • 77.Lim DJ, Park BK, Jang WG, et al. Regulation of bone formation using rapamycin-induced BMP2 expression system: influence of implanted cell number. Mol Cell Toxicol. 2010;6(2):187–194. [Google Scholar]
  • 78.Chan MC, Nguyen PH, Davis BN, et al. A novel regulatory mechanism of the bone morphogenetic protein (BMP) signaling pathway involving the carboxyl-terminal tail domain of BMP type II receptor. Mol Cell Biol. 2007;27(16):5776–5789. doi: 10.1128/MCB.00218-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Poppe H, Rybalkin SD, Rehmann H, et al. Cyclic nucleotide analogs as probes of signaling pathways. Nat Methods. 2008;5(4):277–278. doi: 10.1038/nmeth0408-277. [DOI] [PubMed] [Google Scholar]
  • 80.Miyamoto K, Suzuki H, Yamamoto S, et al. Prostaglandin E2-mediated anabolic effect of a novel inhibitor of phosphodiesterase 4, XT-611, in the in vitro bone marrow culture. J Bone Miner Res. 2003;18(8):1471–1477. doi: 10.1359/jbmr.2003.18.8.1471. [DOI] [PubMed] [Google Scholar]
  • 81.Han CY, Wang Y, Yu L, et al. Small molecules with potent osteogenic-inducing activity in osteoblast cells. Bioorg Med Chem Lett. 2009;19(5):1442–1445. doi: 10.1016/j.bmcl.2009.01.025. [DOI] [PubMed] [Google Scholar]
  • 82.Wu X, Ding S, Ding Q, Gray NS, Schultz PG. A small molecule with osteogenesis-inducing activity in multipotent mesenchymal progenitor cells. J Am Chem Soc. 2002;124(49):14520–14521. doi: 10.1021/ja0283908. [DOI] [PubMed] [Google Scholar]
  • 83.Beloti MM, Bellesini LS, Rosa AL. Purmorphamine enhances osteogenic activity of human osteoblasts derived from bone marrow mesenchymal cells. Cell Biol Int. 2005;29(7):537–541. doi: 10.1016/j.cellbi.2005.02.007. [DOI] [PubMed] [Google Scholar]
  • 84.Wu X, Walker J, Zhang J, Ding S, Schultz PG. Purmorphamine induces osteogenesis by activation of the hedgehog signaling pathway. Chem Biol. 2004;11(9):1229–1238. doi: 10.1016/j.chembiol.2004.06.010. [DOI] [PubMed] [Google Scholar]
  • 85.Sinha S, Chen JK. Purmorphamine activates the Hedgehog pathway by targeting Smoothened. Nat Chem Biol. 2006;2(1):29–30. doi: 10.1038/nchembio753. [DOI] [PubMed] [Google Scholar]
  • 86.Oda T, Notoya K, Gotoh M, et al. Synthesis of novel 2-benzothiopyran and 3-benzothiepin derivatives and their stimulatory effect on bone formation. J Med Chem. 1999;42:751–760. doi: 10.1021/jm980583b. [DOI] [PubMed] [Google Scholar]
  • 87.Hoshino T, Muranishi H, Saito K, et al. Enhancement of fracture repair in rats with streptozotocin-induced diabetes by a single injection of biodegradable microcapsules containing a bone formation stimulant, TAK-778. J Biomed Mater Res. 2000;51(3):299–306. doi: 10.1002/1097-4636(20000905)51:3<299::aid-jbm2>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 88.Rosa AL, Beloti MM. TAK-778 enhances osteoblast differentiation of human bone marrow cells. J Cell Biochem. 2003;89(6):1148–1153. doi: 10.1002/jcb.10582. [DOI] [PubMed] [Google Scholar]
  • 89.Rosa AL, Beloti MM. TAK-778 enhances osteoblast differentiation of human bone marrow cells via an estrogen-receptor-dependent pathway. J Cell Biochem. 2004;91(4):749–755. doi: 10.1002/jcb.10773. [DOI] [PubMed] [Google Scholar]
  • 90.Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997;89(5):747–754. doi: 10.1016/s0092-8674(00)80257-3. [DOI] [PubMed] [Google Scholar]
  • 91.Schroeder TM, Jensen ED, Westendorf JJ. Runx2: a master organizer of gene transcription in developing and maturing osteoblasts. Birth Defects Res C Embryo Today. 2005;75(3):213–225. doi: 10.1002/bdrc.20043. [DOI] [PubMed] [Google Scholar]
  • 92.Hoeppner LH, Secreto FJ, Westendorf JJ. Wnt signaling as a therapeutic target for bone diseases. Expert Opin Ther Targets. 2009;13(4):485–496. doi: 10.1517/14728220902841961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Milat F, Ng KW. Is Wnt signalling the final common pathway leading to bone formation? Mol Cell Endocrinol. 2009;310(1–2):52–62. doi: 10.1016/j.mce.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 94.Agholme F, Aspenberg P. Wnt signaling and orthopedics, an overview. Acta Orthop. 2011;82(2):125–130. doi: 10.3109/17453674.2011.572252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Bodine PV, Stauffer B, Ponce-de-Leon H, et al. A small molecule inhibitor of the Wnt antagonist secreted frizzled-related protein-1 stimulates bone formation. Bone. 2009;44(6):1063–1068. doi: 10.1016/j.bone.2009.02.013. [DOI] [PubMed] [Google Scholar]
  • 96.Moore WJ, Kern JC, Bhat R, et al. Modulation of Wnt signaling through inhibition of secreted frizzled-related protein I (sFRP-1) with N-substituted piperidinyl diphenylsulfonyl sulfonamides. J Med Chem. 2009;52(1):105–116. doi: 10.1021/jm801144h. [DOI] [PubMed] [Google Scholar]
  • 97.Moore WJ, Kern JC, Bhat R, et al. Modulation of Wnt signaling through inhibition of secreted frizzled-related protein I (sFRP-1) with N-substituted piperidinyl diphenylsulfonyl sulfonamides: part II. Bioorg Med Chem. 2010;18(1):190–201. doi: 10.1016/j.bmc.2009.11.002. [DOI] [PubMed] [Google Scholar]
  • 98.Baksh D, Boland GM, Tuan RS. Cross-talk between Wnt signaling pathways in human mesenchymal stem cells leads to functional antagonism during osteogenic differentiation. J Cell Biochem. 2007;101(5):1109–1124. doi: 10.1002/jcb.21097. [DOI] [PubMed] [Google Scholar]
  • 99.Lin GL, Hankenson KD. Integration of BMP, Wnt, and Notch signaling pathways in osteoblast differentiation. J Cell Biochem. 2011;112(12):3491–3501. doi: 10.1002/jcb.23287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Amantea CM, Kim WK, Meliton V, Tetradis S, Parhami F. Oxysterol-induced osteogenic differentiation of marrow stromal cells is regulated by Dkk-1 inhibitable and PI3-kinase mediated signaling. J Cell Biochem. 2008;105(2):424–436. doi: 10.1002/jcb.21840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Brey DM, Motlekar NA, Diamond SL, Mauck RL, Garino JP, Burdick JA. High-throughput screening of a small molecule library for promoters and inhibitors of mesenchymal stem cell osteogenic differentiation. Biotechnol Bioeng. 2011;108(1):163–174. doi: 10.1002/bit.22925. [DOI] [PubMed] [Google Scholar]
  • 102.Hojo H, Igawa K, Ohba S, et al. Development of high-throughput screening system for osteogenic drugs using a cell-based sensor. Biochem Biophys Res Commun. 2008;376(2):375–379. doi: 10.1016/j.bbrc.2008.08.167. [DOI] [PubMed] [Google Scholar]
  • 103.Cai M, Liu X, Shao J, et al. OIC-A006 promotes osteogenesis in vitro and in vivo. Pharmazie. 2008;63(10):751–756. [PubMed] [Google Scholar]
  • 104.Jabbarzadeh E, Starnes T, Khan YM, et al. Induction of angiogenesis in tissue-engineered scaffolds designed for bone repair: a combined gene therapy-cell transplantation approach. Proc Natl Acad Sci USA. 2008;105(32):11099–11104. doi: 10.1073/pnas.0800069105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Keramaris NC, Calori GM, Nikolaou VS, Schemitsch EH, Giannoudis PV. Fracture vascularity and bone healing: a systematic review of the role of VEGF. Injury. 2008;39(Suppl 2):S45–S57. doi: 10.1016/S0020-1383(08)70015-9. [DOI] [PubMed] [Google Scholar]
  • 106.Sefcik LS, Petrie Aronin CE, Botchwey EA. Engineering vascularized tissues using natural and synthetic small molecules. Organogenesis. 2008;4(4):215–227. doi: 10.4161/org.4.4.6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sefcik LS, Petrie Aronin CE, Wieghaus KA, Botchwey EA. Sustained release of sphingosine 1-phosphate for therapeutic arteriogenesis and bone tissue engineering. Biomaterials. 2008;29(19):2869–2877. doi: 10.1016/j.biomaterials.2008.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Papapoulos SE, Landman JO, Bijvoet OL, et al. The use of bisphosphonates in the treatment of osteoporosis. Bone. 1992;13(Suppl 1):S41–S49. doi: 10.1016/s8756-3282(09)80009-4. [DOI] [PubMed] [Google Scholar]
  • 109.Tayalia P, Mooney DJ. Controlled growth factor delivery for tissue engineering. Adv Mater. 2009;21:3269–3285. doi: 10.1002/adma.200900241. [DOI] [PubMed] [Google Scholar]
  • 110.Hans ML, Lowman AM. Biodegradable nanoparticles for drug delivery and targeting. Curr Opin Solid State Mater Sci. 2002;6(4):319–327. [Google Scholar]
  • 111.Pridgen EM, Langer R, Farokhzad OC. Biodegradable, polymeric nanoparticle delivery systems for cancer therapy. Nanomedicine (Lond ) 2007;2(5):669–680. doi: 10.2217/17435889.2.5.669. [DOI] [PubMed] [Google Scholar]
  • 112.Wischke C, Schwendeman SP. Principles of encapsulating hydrophobic drugs in PLA/PLGA microparticles. Int J Pharm. 2008;364(2):298–327. doi: 10.1016/j.ijpharm.2008.04.042. [DOI] [PubMed] [Google Scholar]
  • 113.Oredein-McCoy O, Krogman NR, Weikel AL, Hindenlang MD, Allcock HR, Laurencin CT. Novel factor-loaded polyphosphazene matrices: potential for driving angiogenesis. J Microencapsul. 2009;26(6):544–555. doi: 10.1080/02652040802500473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Peach MS, Kumbar SG, Brown JL, Laurencin CT. Recent developments in nanoparticle based targeted delivery of chemotherapeutics. Curr Bioactive Compunds. 2009;5:170–184. [Google Scholar]
  • 115.Cushnie EK, Khan YM, Laurencin CT. Tissue-engineered matrices as functional delivery systems: adsorption and release of bioactive proteins from degradable composite scaffolds. J Biomed Mater Res A. 2011;94(2):568–575. doi: 10.1002/jbm.a.32722. [DOI] [PubMed] [Google Scholar]
  • 116.Siddappa R, Martens A, Doorn J, et al. cAMP/PKA pathway activation in human mesenchymal stem cells in vitro results in robust bone formation in vivo. Proc Natl Acad Sci USA. 2008;105(20):7281–7286. doi: 10.1073/pnas.0711190105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Morozowich NL, Weikel AL, Nichol JL, et al. Polyphosphazene containing vitamin substituents: synthesis, characterization and hydrolytic sensitivity. Macromolecules. 2011;44:1355–1365. [Google Scholar]
  • 118.Laurencin CT. Musculoskeletal regenerative engineering: taking on the grand challenges. Rejuvenation Res. 2008;10:37S. [Google Scholar]
  • 119.Benoit DS, Nuttelman CR, Collins SD, Anseth KS. Synthesis and characterization of a fluvastatin-releasing hydrogel delivery system to modulate hMSC differentiation and function for bone regeneration. Biomaterials. 2006;27(36):6102–6110. doi: 10.1016/j.biomaterials.2006.06.031. [DOI] [PubMed] [Google Scholar]
  • 120.Leach JK, Mooney DJ. Bone engineering by controlled delivery of osteoinductive molecules and cells. Expert Opin Biol Ther. 2004;4(7):1015–1027. doi: 10.1517/14712598.4.7.1015. [DOI] [PubMed] [Google Scholar]
  • 121.Hamidi M, Azadi A, Rafiei P. Hydrogel nanoparticles in drug delivery. Adv Drug Deliv Rev. 2008;60(15):1638–1649. doi: 10.1016/j.addr.2008.08.002. [DOI] [PubMed] [Google Scholar]
  • 122▪.Haller MF, Saltzman WM. Nerve growth factor delivery systems. J Control Release. 1998;53(1–3):1–6. doi: 10.1016/s0168-3659(97)00232-0. Comprehensive review of hydrogels for drug delivery. [DOI] [PubMed] [Google Scholar]
  • 123▪ ▪.Ladewig K. Drug delivery in soft tissue engineering. Expert Opin Drug Deliv. 2011;8(9):1175–1188. doi: 10.1517/17425247.2011.588698. Insightful article describing many methods under current investigation for drug delivery. [DOI] [PubMed] [Google Scholar]
  • 124.Saltzman WM, Olbricht WL. Building drug delivery into tissue engineering. Nat Rev Drug Discov. 2002;1(3):177–186. doi: 10.1038/nrd744. [DOI] [PubMed] [Google Scholar]
  • 125.Tsang VL, Bhatia SN. Three-dimensional tissue fabrication. Adv Drug Deliv Rev. 2004;56(11):1635–1647. doi: 10.1016/j.addr.2004.05.001. [DOI] [PubMed] [Google Scholar]
  • 126.Gkioni K, Leeuwenburgh SC, Douglas TE, Mikos AG, Jansen JA. Mineralization of hydrogels for bone regeneration. Tissue Eng Part B Rev. 2010;16(6):577–585. doi: 10.1089/ten.TEB.2010.0462. [DOI] [PubMed] [Google Scholar]
  • 127.Cheng F, Choy YB, Choi H, Kim KK. Modeling of small-molecule release from crosslinked hydrogel microspheres: effect of crosslinking and enzymatic degradation of hydrogel matrix. Int J Pharm. 2011;403(1–2):90–95. doi: 10.1016/j.ijpharm.2010.10.029. [DOI] [PubMed] [Google Scholar]
  • 128.Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev Med Devices. 2011;8(5):607–626. doi: 10.1586/erd.11.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Van Tomme SR, Storm G, Hennink WE. In situ gelling hydrogels for pharmaceutical and biomedical applications. Int J Pharm. 2008;355(1–2):1–18. doi: 10.1016/j.ijpharm.2008.01.057. [DOI] [PubMed] [Google Scholar]
  • 130.Su K, Shi X, Varshney RR, Wang DA. Transplantable delivery systems for in situ controlled release of bisphosphonate in orthopedic therapy. Expert Opin Drug Deliv. 2011;8(1):113–126. doi: 10.1517/17425247.2011.541438. [DOI] [PubMed] [Google Scholar]
  • 131.Verron E, Gauthier O, Janvier P, et al. In vivo bone augmentation in an osteoporotic environment using bisphosphonate-loaded calcium deficient apatite. Biomaterials. 2010;31(30):7776–7784. doi: 10.1016/j.biomaterials.2010.06.047. [DOI] [PubMed] [Google Scholar]
  • 132.Lalatonne Y, Monteil M, Jouni H, et al. Superparamagnetic bifunctional bisphosphonates nanoparticles: a potential MRI contrast agent for osteoporosis therapy and diagnostic. J Osteoporos. 2010;2010:747852. doi: 10.4061/2010/747852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133▪.Seshima H, Yoshinari M, Takemoto S, et al. Control of bisphosphonate release using hydroxyapatite granules. J Biomed Mater Res B Appl Biomater. 2006;78(2):215–221. doi: 10.1002/jbm.b.30446. Interesting article highlighting the controlled release of bisphosphonate from hydroxyapatite. [DOI] [PubMed] [Google Scholar]
  • 134.Panzavolta S, Torricelli P, Bracci B, Fini M, Bigi A. Alendronate and pamidronate calcium phosphate bone cements: setting properties and in vitro response of osteoblast and osteoclast cells. J Inorg Biochem. 2009;103(1):101–106. doi: 10.1016/j.jinorgbio.2008.09.012. [DOI] [PubMed] [Google Scholar]
  • 135▪ ▪.Nair SL, Laurencin CT. Polymers as biomaterials for tissue engineering and controlled drug delivery. Adv Biochem Eng Biotechnol. 2006;102:47–90. doi: 10.1007/b137240. Comprehensive review providing an in-depth look at polymeric scaffolds for bone tissue engineering. [DOI] [PubMed] [Google Scholar]
  • 136.Weiss WA, Taylor SS, Shokat KM. Recognizing and exploiting differences between RNAi and small-molecule inhibitors. Nat Chem Biol. 2007;3(12):739–744. doi: 10.1038/nchembio1207-739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J Cell Biochem. 2006;98(3):538–554. doi: 10.1002/jcb.20719. [DOI] [PubMed] [Google Scholar]
  • 138.Sumanasinghe RD, Bernacki SH, Loboa EG. Osteogenic differentiation of human mesenchymal stem cells in collagen matrices: effect of uniaxial cyclic tensile strain on bone morphogenetic protein (BMP-2) mRNA expression. Tissue Eng. 2006;12(12):3459–3465. doi: 10.1089/ten.2006.12.3459. [DOI] [PubMed] [Google Scholar]
  • 139.Marie PJ, Fromigue O. Osteogenic differentiation of human marrow-derived mesenchymal stem cells. Regen Med. 2006;1(4):539–548. doi: 10.2217/17460751.1.4.539. [DOI] [PubMed] [Google Scholar]
  • 140.Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 141.Wu W, Ye Z, Zhou Y, Tan WS. AICAR, a small chemical molecule, primes osteogenic differentiation of adult mesenchymal stem cells. Int J Artif Organs. 2012;34(12):1128–1136. doi: 10.5301/ijao.5000007. [DOI] [PubMed] [Google Scholar]
  • 142.Baek KH, Lee WY, Oh KW, et al. The effect of simvastatin on the proliferation and differentiation of human bone marrow stromal cells. J Korean Med Sci. 2005;20(3):438–444. doi: 10.3346/jkms.2005.20.3.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Ruiz-Gaspa S, Nogues X, Enjuanes A, et al. Simvastatin and atorvastatin enhance gene expression of collagen type 1 and osteocalcin in primary human osteoblasts and MG-63 cultures. J Cell Biochem. 2007;101(6):1430–1438. doi: 10.1002/jcb.21259. [DOI] [PubMed] [Google Scholar]
  • 144.Pagkalos J, Cha JM, Kang Y, Heliotis M, Tsiridis E, Mantalaris A. Simvastatin induces osteogenic differentiation of murine embryonic stem cells. J Bone Miner Res. 2010;25(11):2470–2478. doi: 10.1002/jbmr.163. [DOI] [PubMed] [Google Scholar]
  • 145.Dai W, Dong J, Fang T, Uemura T. Stimulation of osteogenic activity in mesenchymal stem cells by FK506. J Biomed Mater Res A. 2008;86(1):235–243. doi: 10.1002/jbm.a.31685. [DOI] [PubMed] [Google Scholar]
  • 146.Koh JT, Ge C, Zhao M, et al. Use of a stringent dimerizer-regulated gene expression system for controlled BMP2 delivery. Mol Ther. 2006;14:684–691. doi: 10.1016/j.ymthe.2006.05.021. [DOI] [PubMed] [Google Scholar]
  • 147.Mok SK, Chen WF, Lai WP, et al. Icariin protects against bone loss induced by oestrogen deficiency and activates oestrogen receptor-dependent osteoblastic functions in UMR 106 cells. Br J Pharmacol. 2010;159(4):939–949. doi: 10.1111/j.1476-5381.2009.00593.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Aghaloo TL, Amantea CM, Cowan CM, et al. Oxysterols enhance osteoblast differentiation in vitro and bone healing in vivo. J Orthop Res. 2007;25(11):1488–1497. doi: 10.1002/jor.20437. [DOI] [PubMed] [Google Scholar]
  • 149.Zhang Y, Zeng X, Zhang L, Zheng X. Stimulatory effect of puerarin on bone formation through activation of PI3K/Akt pathway in rat calvaria osteoblasts. Planta Med. 2007;73(4):341–347. doi: 10.1055/s-2007-967168. [DOI] [PubMed] [Google Scholar]
  • 150.Wong R, Rabie B. Effect of puerarin on bone formation. Osteoarthritis Cartilage. 2007;15(8):894–899. doi: 10.1016/j.joca.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 151.Heim M, Frank O, Kampmann G, et al. The phytoestrogen genistein enhances osteogenesis and represses adipogenic differentiation of human primary bone marrow stromal cells. Endocrinology. 2004;145(2):848–859. doi: 10.1210/en.2003-1014. [DOI] [PubMed] [Google Scholar]
  • 152.Wong RW, Rabie AB. Effect of genistin on bone formation. Front Biosci (Elite Ed ) 2010;2:764–770. doi: 10.2741/e136. [DOI] [PubMed] [Google Scholar]
  • 153.Tang DZ, Yang F, Yang Z. Psoralen stimulates osteoblast differentiation through activation of BMP signaling. Biochem Biophys Res Commun. 2011;405(2):256–261. doi: 10.1016/j.bbrc.2011.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Wong RW, Rabie AB. Effect of psoralen on bone formation. J Orthop Res. 2011;29(2):158–164. doi: 10.1002/jor.21124. [DOI] [PubMed] [Google Scholar]
  • 155.Tang DZ, Hou W, Zhou Q, et al. Osthole stimulates osteoblast differentiation and bone formation by activation of beta-catenin–BMP signaling. J Bone Miner Res. 2010;25(6):1234–1245. doi: 10.1002/jbmr.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Li CY, Wu T, Li QN, et al. Quantitative study on the effect of osthole on proximal tibiae in ovariectomized (OVX) rats. Yao Xue Xue Bao. 1996;31(5):327–332. [PubMed] [Google Scholar]
  • 157.Yang L, Wang NL, Cai GP. Maohuoside A promotes osteogenesis of rat mesenchymal stem cells via BMP and MAPK signaling pathways. Mol Cell Biochem. 2011;358(1–2):37–44. doi: 10.1007/s11010-011-0918-y. [DOI] [PubMed] [Google Scholar]
  • 158.Lee HW, Suh JH, Kim HN, et al. Berberine promotes osteoblast differentiation by Runx2 activation with p38 MAPK. J Bone Miner Res. 2008;23(8):1227–1237. doi: 10.1359/jbmr.080325. [DOI] [PubMed] [Google Scholar]
  • 159.Dai Z, Li Y, Quarles LD, et al. Resveratrol enhances proliferation and osteoblastic differentiation in human mesenchymal stem cells via ER-dependent ERK1/2 activation. Phytomedicine. 2007;14(12):806–814. doi: 10.1016/j.phymed.2007.04.003. [DOI] [PubMed] [Google Scholar]
  • 160.Sakamoto S, Kojima F, Igarashi M, et al. Decalpenic acid, a novel small molecule from Penicillium verruculosum CR37010, induces early osteoblastic markers in pluripotent mesenchymal cells. J Antibiot (Tokyo) 2010;63(12):703–708. doi: 10.1038/ja.2010.114. [DOI] [PubMed] [Google Scholar]
  • 161.Uchiyama S, Yamaguchi M. Beta-cryptoxanthin stimulates cell differentiation and mineralization in osteoblastic MC3T3-E1 cells. J Cell Biochem. 2005;95(6):1224–1234. doi: 10.1002/jcb.20496. [DOI] [PubMed] [Google Scholar]
  • 162.Harada SI, Balena R, Rodan GA, Rodan SB. The role of prostaglandins in bone formation. Connect Tissue Res. 1995;31(4):279–282. doi: 10.3109/03008209509010823. [DOI] [PubMed] [Google Scholar]
  • 163.Knippenberg M, Helder MN, de Blieck-Hogervorst JM, Wuisman PI, Klein-Nulend J. Prostaglandins differentially affect osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. Tissue Eng. 2007;13(10):2495–2503. doi: 10.1089/ten.2006.0420. [DOI] [PubMed] [Google Scholar]
  • 164.Wong RW, Rabie AB. Effect of naringin on bone cells. J Orthop Res. 2006;24(11):2045–2050. doi: 10.1002/jor.20279. [DOI] [PubMed] [Google Scholar]
  • 165.Pang WY, Wang XL, Mok SK, et al. Naringin improves bone properties in ovariectomized mice and exerts oestrogen-like activities in rat osteoblast-like (UMR-106) cells. Br J Pharmacol. 2010;159(8):1693–1703. doi: 10.1111/j.1476-5381.2010.00664.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Kim YJ, Bae YC, Suh KT, Jung JS. Quercetin, a flavonoid, inhibits proliferation and increases osteogenic differentiation in human adipose stromal cells. Biochem Pharmacol. 2006;72(10):1268–1278. doi: 10.1016/j.bcp.2006.08.021. [DOI] [PubMed] [Google Scholar]
  • 167.Wong RW, Rabie AB. Effect of quercetin on preosteoblasts and bone defects. Open Orthop J. 2008;2:27–32. doi: 10.2174/1874325000802010027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have biphasic effects on angiogenesis. Circulation. 2002;105(6):739–745. doi: 10.1161/hc0602.103393. [DOI] [PubMed] [Google Scholar]
  • 169.Shiota M, Hikita Y, Kawamoto Y, et al. Pravastatin-induced proangiogenic effects depend upon extracellular FGF-2. J Cell Mol Med. 2011 doi: 10.1111/j.1582-4934.2011.01494.x. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Llevadot J, Asahara T. Effects of statins on angiogenesis and vasculogenesis. Rev Esp Cardiol. 2002;55(8):838–844. doi: 10.1016/s0300-8932(02)76713-4. [DOI] [PubMed] [Google Scholar]
  • 171.Chung BH, Kim JD, Kim CK, et al. Icariin stimulates angiogenesis by activating the MEK/ERK- and PI3K/Akt/eNOS-dependent signal pathways in human endothelial cells. Biochem Biophys Res Commun. 2008;376(2):404–408. doi: 10.1016/j.bbrc.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 172.Rivron NC, Raiss CC, Liu J, et al. Sonic Hedgehog-activated engineered blood vessels enhance bone tissue formation. Proc Natl Acad Sci USA. 2012;109(12):4413–4418. doi: 10.1073/pnas.1117627109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Zhang S, Chen S, Shen Y, et al. Puerarin induces angiogenesis in myocardium of rat with myocardial infarction. Biol Pharm Bull. 2006;29(5):945–950. doi: 10.1248/bpb.29.945. [DOI] [PubMed] [Google Scholar]
  • 174.Mehrabi MR, Serbecic N, Tamaddon F, et al. Clinical and experimental evidence of prostaglandin E1-induced angiogenesis in the myocardium of patients with ischemic heart disease. Cardiovasc Res. 2002;56(2):214–224. doi: 10.1016/s0008-6363(02)00591-6. [DOI] [PubMed] [Google Scholar]
  • 175.Pillai A, Mahadik SP. Differential effects of haloperidol and olanzapine on levels of vascular endothelial growth factor and angiogenesis in rat hippocampus. Schizophr Res. 2006;87(1–3):48–59. doi: 10.1016/j.schres.2006.06.017. [DOI] [PubMed] [Google Scholar]
  • 176.Chen KM, Ge BF, Liu XY, et al. Icariin inhibits the osteoclast formation induced by RANKL and macrophage-colony stimulating factor in mouse bone marrow culture. Pharmazie. 2007;62(5):388–391. [PubMed] [Google Scholar]
  • 177.Yamagami H, Nishioka T, Ochiai E, et al. Inhibition of osteoclastogenesis by a phosphodiesterase 4 inhibitor XT-611 through synergistic action with endogenous prostaglandin E2. Biochem Pharmacol. 2003;66(5):801–807. doi: 10.1016/s0006-2952(03)00409-x. [DOI] [PubMed] [Google Scholar]
  • 178.Staal A, Frith JC, French MH, et al. The ability of statins to inhibit bone resorption is directly related to their inhibitory effect on HMG-CoA reductase activity. J Bone Miner Res. 2003;18(1):88–96. doi: 10.1359/jbmr.2003.18.1.88. [DOI] [PubMed] [Google Scholar]
  • 179.Moon HJ, Kim SE, Yun YP, et al. Simvastatin inhibits osteoclast differentiation by scavenging reactive oxygen species. Exp Mol Med. 2011;43(11):605–612. doi: 10.3858/emm.2011.43.11.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Igarashi K, Hirotani H, Woo JT, Stern PH. Cyclosporine A and FK506 induce osteoclast apoptosis in mouse bone marrow cell cultures. Bone. 2004;35(1):47–56. doi: 10.1016/j.bone.2004.02.009. [DOI] [PubMed] [Google Scholar]
  • 181.Smink JJ, Tunn P-U, Leutz A. Rapamycin inhibits osteoclast formation in giant cell tumor of bone through the C/EBP{beta}–MafB axis. J Mol Med. 2011;90(1):25–30. doi: 10.1007/s00109-011-0823-6. [DOI] [PubMed] [Google Scholar]
  • 182.Zhang Q, Qin L, He W, et al. Coumarins from Cnidium monnieri and their antiosteoporotic activity. Planta Med. 2007;73(1):13–19. doi: 10.1055/s-2006-951724. [DOI] [PubMed] [Google Scholar]
  • 183.Hu JP, Nishishita KS, Yoshida H, et al. Berberine inhibits RANKL-induced osteoclast formation and survival through suppressing the NF-kappaB and Akt pathways. Eur J Pharmacol. 2008;580(1–2):70–79. doi: 10.1016/j.ejphar.2007.11.013. [DOI] [PubMed] [Google Scholar]
  • 184.He X, Andersson G, Lindgren U, Li Y. Resveratrol prevents RANKL-induced osteoclast differentiation of murine osteoclast progenitor RAW 264.7 cells through inhibition of ROS production. Biochem Biophys Res Commun. 2010;401(3):356–362. doi: 10.1016/j.bbrc.2010.09.053. [DOI] [PubMed] [Google Scholar]
  • 185.Take I, Kobayashi Y, Yamamoto Y, et al. Prostaglandin E2 strongly inhibits human osteoclast formation. Endocrinology. 2005;146(12):5204–5214. doi: 10.1210/en.2005-0451. [DOI] [PubMed] [Google Scholar]
  • 186.Wattel A, Kamel S, Prouillet C, et al. Flavonoid quercetin decreases osteoclastic differentiation induced by RANKL via a mechanism involving NF kappa B and AP-1. J Cell Biochem. 2004;92(2):285–295. doi: 10.1002/jcb.20071. [DOI] [PubMed] [Google Scholar]
  • 187.Ang ES, Yang X, Chen H, Liu Q, Zheng MH, Xu J. Naringin abrogates osteoclastogenesis and bone resorption via the inhibition of RANKL-induced NF-kappaB and ERK activation. FEBS Lett. 2011;585(17):2755–2762. doi: 10.1016/j.febslet.2011.07.046. [DOI] [PubMed] [Google Scholar]
  • 188.Uchiyama S, Yamaguchi M. Beta-cryptoxanthin stimulates apoptotic cell death and suppresses cell function in osteoclastic cells: change in their related gene expression. J Cell Biochem. 2006;98(5):1185–1195. doi: 10.1002/jcb.20824. [DOI] [PubMed] [Google Scholar]

RESOURCES