Next Article in Journal
Obesity as a Risk Factor for Severe COVID-19 and Complications: A Review
Next Article in Special Issue
Extracellular Vesicles from Mesenchymal Stem Cells as Potential Treatments for Osteoarthritis
Previous Article in Journal
A Propagated Skeleton Approach to High Throughput Screening of Neurite Outgrowth for In Vitro Parkinson’s Disease Modelling
Previous Article in Special Issue
TNFAIP8 Regulates Intestinal Epithelial Cell Differentiation and May Alter Terminal Differentiation of Secretory Progenitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review

by
Anita Novais
1,2,
Eirini Chatzopoulou
1,2,3,
Catherine Chaussain
1,2 and
Caroline Gorin
1,2,*
1
Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France
2
AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
3
Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
*
Author to whom correspondence should be addressed.
Cells 2021, 10(4), 932; https://doi.org/10.3390/cells10040932
Submission received: 5 March 2021 / Revised: 10 April 2021 / Accepted: 15 April 2021 / Published: 17 April 2021
(This article belongs to the Collection Stem Cells in Tissue Engineering and Regeneration)

Abstract

:
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The “gold standard” for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.

1. Introduction

The skeletal system is dynamic, metabolically active and functionally diverse. As well as a structural function, it has a metabolic role. It supports and protects the vital internal organs and is the site of synthesis of the hematopoietic marrow and provides sites of muscle attachment for locomotion. Bone is involved in both mineral metabolism, via calcium and phosphate homeostasis, and acid–base balance, via the buffering of hydrogen ions [1]. Moreover, it has been suggested that bone has other important endocrine functions in fertility, glucose metabolism, appetite regulation and muscle function [2,3]. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. Throughout life, the craniofacial area is at risk of complex injuries that require bone grafting to restore function. The etiology of such injuries may be accidental (e.g., acute trauma), congenital (e.g., birth defects or deformities), pathological (e.g., maxillofacial tumors, such as ameloblastoma, or infection) or surgical. Whenever the lesions are extensive, they cause large bone defects that cannot self-repair because they exceed the body’s natural regenerative capacity. The “gold standard” for treating these large defects is autologous bone grafting. Since the defects are extensive, harvesting of the necessary bone at the donor site can cause major morbidity, including bone deformity, as well as pain and occasionally continuous progressive resorption. Tissue engineering has made it possible to approach these issues from another angle. Before talking about tissue engineering; however, it is necessary to describe the tissue of interest. This review is, therefore, organized into three sections, the first describing general bone physiology (e.g., its composition, functioning and signaling pathways), the second the implication of FGF-2 in bone physiology, and the third highlighting the interesting characteristics of FGF-2 for craniofacial bone engineering and its various current potential uses for promoting bone repair.

2. Background on Bone Physiology

In the skeleton, there are two types of bone differentiated by their structure—cortical and trabecular. Although both types have an identical chemical composition, they differ both macroscopically and microscopically [4] (Appendix Figure A1).

2.1. Bone Composition

Bone is a mineralized connective tissue composed of bone cells, vessels, and an extracellular matrix (ECM), which is produced by the bone cells. The proportion of these components varies with bone type (long or flat bone) and anatomical site [5]. The mineralized component of bone tissue gives rigidity and hardness to the material, while the organic components of the ECM provide flexibility [6].

2.1.1. Bone Cells

The osteoblast, a specialized bone-forming cell, mostly differentiated from mesenchymal stem cells (MSCs) [7,8], produces and secretes the major bone matrix protein essential for matrix mineralization, namely, type I collagen [9,10]. These cells work in clusters on the bone surface [5], becoming committed to one of various possible fates, thanks to some key proteins and pathway signaling, such as runt-related transcription factor 2 (Runx-2) and Osterix (Figure 1).
Osteocytes, the most abundant cell type, are found in mature bone and are long-lived [11]. They are distributed throughout the bone matrix and can interact with other osteocytes or osteoblasts on the bone surface, vasculature and bone cells on the surface of bones, in a complex intercellular network [6,12,13]. Osteocyte-transduced signals seem to orchestrate bone response by regulating the synchronized action of osteoblasts and osteoclasts [14,15,16] (Figure 1).
The osteoclasts, multinucleated cells formed by the fusion of precursors derived from the hematopoietic cells of the mononuclear lineage [17,18], are the only cells known to be capable of resorbing bone (Figure 1).
Other types of cells, such as chondrocytes are found within the bone. These are derived from pluripotent MSCs, and secrete type II collagen, participating in the endochondral ossification process [19].

2.1.2. Bone Extracellular Matrix

Bone ECM contains both a mineral and an organic phase, the latter representing approximately 90% of the organic content of bone tissue. The mineral portion is largely calcium phosphate in the form of hydroxyapatite crystals deposited in an osteoid matrix and is responsible for bone’s mechanical rigidity and load-bearing strength. The organic portion, which contains water, non-collagenous proteins, lipids and specialized bone cells, gives flexibility and elasticity to bone tissue [1,6].
Non-collagenous proteins constitute 10% to 15% of total bone protein content. Almost 25% of non-collagenous proteins are adsorbed from serum and due to their acidic properties are able to bind to the matrix [20], which allows strengthening of the collagen structure and regulating its mineralization [4]. Osteocalcin is the major non-collagenous protein, and is involved in calcium binding, stabilization of hydroxyapatite in the matrix and the regulation of bone formation, acting as a negative regulator, inhibiting premature or inappropriate mineralization [21,22].

2.2. Bone Formation

Bones form through two different processes: bone modeling and bone remodeling (Figure 2). Bone modeling occurs primarily during growth and development in childhood, although it can also appear after the skeleton has matured [23,24].
In contrast, bone remodeling occurs after the skeleton has reached maturity, during adulthood, involving resorption of old or damaged bone, and its replacement by newly formed bone. Here, we focus on bone remodeling.

2.2.1. Bone Remodeling Process

Bone remodeling is essential for structural integrity, biomechanical stability, bone volume and calcium/phosphate homeostasis [1,4,25]. In normal adult bone, there is a homeostatic balance in which bone resorption is followed by bone formation for maintaining bone strength and mineral homeostasis, keeping the overall bone volume and structure unchanged [26,27]. By this process, about 10% of the skeleton may be renewed every year [28]. That is, bone remodeling only takes place when it is required, either because the specific area is damaged and/or old.
The bone remodeling cycle has several phases: cell activation, bone resorption, reversal, bone formation and termination (Figure 2).

2.2.2. Regulation of Bone Remodeling

The remodeling cycle is tightly regulated to achieve a balance between bone formation and resorption. Considering that remodeling can occur in several locations simultaneously, local regulation is critical to achieving this balance.
  • Major signaling pathways
  • RANKL/RANK/OPG
One of the major signaling pathways that regulates bone remodeling involves three proteins: RANKL, receptor activator of nuclear factor kappa-B (RANK) and osteoprotegerin (OPG). The interaction between these proteins determines whether, at a specific location, bone resorption or bone formation occur. RANKL is a cytokine expressed on the surface of osteoblasts, osteocytes and chondrocytes. It activates nuclear factor kappa-B (NF-kB) and other signaling pathways through the interaction with its receptor, RANK, located on osteoclast precursors. RANKL/RANK activation has an important role in osteoclast differentiation, allowing osteoclast formation, activation and survival [13,29,30,31,32]. OPG, a soluble decoy receptor for RANKL expressed by osteoblasts and osteocytes, binds to RANKL with high affinity, preventing it from binding to its receptor RANK. Thus, OPG is a natural inhibitor of RANKL. The RANKL/OPG expression rate regulates the extent of osteoclast formation and activity [33,34,35] (Figure 1).
2.
Wnt signaling
Wingless-related integration site (Wnt) molecules are cysteine-rich glycoproteins involved in controlling cell proliferation, cell-fate specification, gene expression and cell survival. Wnt signaling pathways are involved in bone formation, having an anabolic effect, and increasing bone density and strength, by regulation of osteoblast differentiation and function [36,37] (Figure 1).
Wnt pathways also play a major role in osteoclast differentiation. Specifically, Wnt canonical signaling up-regulates OPG and downregulates RANKL, which inhibits osteoclast formation and therefore bone resorption [36,38]. In contrast, activation of the noncanonical pathway in osteoclast precursors enhances RANKL-induced osteoclastic differentiation [36].
Wnt signaling is inhibited by secreted proteins such as sclerostin and Dickkopf-related protein 1–4 (DKK-3/4) synthesized by osteocytes [39,40] (Figure 1). Nevertheless, during bone remodeling, osteocytes decrease the expression of sclerostin and Dickkopf-related protein 1 and 2 (DKK-1/2), allowing osteoblast bone formation to occur after bone resorption. After the completion of remodeling, newly formed osteocytes become entombed within the bone matrix and start expressing Wnt inhibitors, stopping further bone formation [12].
  • Endocrine regulation
Bone turnover by osteoblasts and osteoclasts is essential for the maintenance of bone strength and morphology. Due to its importance, this process must be thoroughly regulated to prevent malfunction of the remodeling process at any stage of the cycle. Several hormonal agents are involved in this regulation, including PTH [10], 1,25 (OH) Vitamin D [4], calcitonin [41], thyroid hormone [42], growth hormone [43], glucocorticoids [44] and sex hormones [45].
  • Paracrine regulation
Some cytokines may have stimulatory and inhibitory effects on bone metabolism. Cytokines like interleukin 1 (IL-1), IL-6, and tumor necrosis factor (TNFα) can increase osteoclastic resorption, whereas others, such as interleukin 4 (IL-4) and gamma interferon, decrease osteoclast proliferation and differentiation [46,47].
Prostaglandins may also influence bone formation, although their exact role remains unclear. Prostaglandin E2 (PGE2) is a major inducer of bone resorption and is thought to increase the RANKL/OPG ratio to improve osteoclastogenesis (Figure 1). At the same time, it has been hypothesized to stimulate osteoblast proliferation and differentiation thereby enhancing bone formation [48]. Inside the bone matrix, there are growth factors that affect bone metabolism. The main families involved are the transforming growth factor β (TGFβ) family (TGFβ and BMPs) and the fibroblast growth factor (FGF) family.

2.3. Bone Vascularization

There is an intimate link between osteogenesis and angiogenesis, both processes needing to be tightly coupled for optimal physiological bone function [49]. In the event of a critical bone defect, early vascularization is necessary for osteogenic reconstruction, to allow the nutritional support for the bone grafts [50,51,52]. The close relationship between blood vessels and bone cells is also well illustrated by abnormalities resulting from inappropriate vascularization, these leading to the appearance of skeletal malformation, such as craniofacial dysmorphology [53].
In the close connection between these two processes, several factors have been described as being both angiogenic and osteogenic. Notably, it has been demonstrated that hypoxia (oxygen tension) and the vascular endothelial growth factor (VEGF) family affect endochondral angiogenesis as well as cells from the bone lineage [54,55,56].
Indeed, several pro-angiogenic factors are involved in bone repair. Some of these factors have direct effects, both having angiogenic properties and regulating osteogenic molecules, like BMPs, angiopoietin (Ang), platelet-derived growth factor (PDGF) and insulin-like growth factor (IGF) family members (Appendix Table A2).
Others are well known to indirectly enhance bone repair using their pro-angiogenic properties. VEGF, an endothelial cell (EC)-specific mitogen, is secreted by cells involved in skeletal development and repair, such as hypertrophic chondrocytes or differentiating mesenchymal cells, osteoblasts, and ECs [57,58]. It can be a chemoattractant molecule, engaging ECs into bone tissue and tightly controlling the differentiation and functions of osteoblasts and osteoclasts [55,59,60,61,62]. It is also involved both in endochondral ossification promoting vessel invasion and cell recruitment [59,63], and in intramembranous ossification, by affecting bone cell activity [52,53,64]. It has been reported that VEGF upregulates the RANK receptor in ECs and strongly stimulates angiogenesis [65]. In turn, RANKL may have an important role in enabling EC survival via the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway [66,67], one of the major signaling pathways triggered by activated VEGF receptor [68] (Figure 1).
The hypoxic signaling pathway has been reported to directly enhance VEGF expression, being described as a major regulator of VEGF expression [69,70]. Indeed, hypoxia-inducible transcription factors (HIFs) are expressed in osteogenic cells, notably osteoblasts, and hence, hypoxia upregulates VEGF expression, thereby promoting angiogenesis and osteogenesis. Thus, hypoxia and VEGF signaling are involved in the coupling of angiogenesis and osteogenesis [54,55,71].
This review focuses on the FGF family and specifically fibroblast growth factor 2 (FGF-2) and its potential usefulness in bone tissue engineering.

3. FGF-2 in Bone Homeostasis

3.1. FGF/FGFR Signaling in Bone

Various growth factors act within the bone matrix and influence bone metabolism. The main families involved are the TGFβ (TGFβ and BMPs) and FGF families. The latter is also involved in angiogenesis, which makes it interesting to study more in detail. In this review, we focus on the great potential of FGF-2 in bone metabolism and discuss the interest in its use in bone tissue engineering.
Members of the FGF family are single-chain polypeptide growth factors of approximately 20–35 kDa. At least 23 members of this family have been described in mammals [72], from FGF-1 to FGF-23. The secreted FGFs are differentially expressed in almost all tissues of the developing embryo, functioning as essential regulators of the earliest stages of embryonic development. They are also expressed in postnatal and adult tissues, fulfilling essential roles in tissue homeostasis, repair, regeneration, angiogenesis and bone metabolism [61,73,74,75,76,77,78]. There are four FGF receptors: FGFR-1 to FGFR-4 [79,80].
Given the ubiquitous roles of FGF signals in development, homeostasis, and disease, tight regulation of the pathways is essential through cofactors that participate in the affinity and specificity of FGFR-binding, and also in specifying signaling activities [81,82]. The endocrine regulation of FGFs, including FGF-19, FGF-21 and FGF-23, is mediated by their binding with the Klotho protein family (α or βKlotho or KLPH) [83,84,85], which allows them to circulate through the matrix without being trapped and stored [82], thereby acting in an endocrine-like fashion.
Canonical FGFs exert their pleiotropic effects by binding and activating the FGFR subfamily of receptor tyrosine kinases that are coded by four genes (FGFR-1, FGFR-2, FGFR-3, and FGFR-4) in mammals. [86]. The four FGFRs have distinct ligand specificity and are expressed in a tissue-specific manner [87]. Cofactors such as heparan sulfate and klotho are low-affinity receptors that do not induce a biological signal but rather are used as auxiliary proteins to regulate FGF binding and subsequent phosphorylation of adaptor proteins for four major intracellular pathways [88,89,90,91]: rat sarcoma-mitogen-activated protein kinase-extracellular signal-regulated kinase 1 and 2 (RAS-MAPK-ERK1/2), PI3K-AKT-glycogen synthase kinase 3 (GSK3), phospholipase Cγ-protein kinase C (PLCγ-PKC), and signal transducer and activator of transcription proteins-Janus kinase (STAT-Jak) [81,86,92,93]. The main regulation pathway remains the paracrine one, involving FGF-2, which binds FGFRs through a heparan sulphate glycosaminoglycan binding site, limiting their diffusion through the ECM [88,90,94]. Upon binding of FGF to its receptor, receptor dimerization and transautophosphorylation of the kinase domain take place [95] (Figure 3).
FGFs can activate several MAPKs such as C-Jun N-terminal kinase (JNK), ERK, and p38MAPK [96], that share many structural similarities, while inducing different responses [97,98]. The ERK1/2 branch, one of the major routes for FGF signaling [99], promotes a mitogenic response and is observed in all cell types, while p38 and JNK kinase are usually associated with inflammatory and stress responses [98,100] and are mainly involved in the cell cycle, cytoskeleton and cell migration [101]. For instance, FGF-2 regulates mesenchymal stem cell migration [102] via the PI3K-AKT pathway, neural progenitor cell proliferation via PI3K/GSK3 signaling [103], and fibroblast migration via PI3 kinase, Ras-related C3 botulinum toxin substrate 1 (Rac1) and JNK [104], as well as promoting bone marrow MSC proliferation through the ERK1/2 pathway [105]. Activation of the MAPK pathway, in response to FGF-2 signaling, is key in determining the activity of RUNX-2, a master transcription factor of bone formation [106]. FGF-2 promotes osteoblast proliferation and differentiation [107], through the activation of the ERK1/2 pathway [108,109,110] and a crucial role of this pathway has been identified in the differentiation of osteoblasts and chondrocytes [107,111,112,113]. Specifically, MAPK phosphorylates Runx-2 Ser/Thr residues, a critical step for Runx2 acetylation and stabilization against degradation [114]. Sprouty (SPRY) and Sprouty related (SPRED) are two antagonists of this pathway [115,116] that act as intracellular antagonists of FGFR signaling, the first by suppressing ERK1/2 activation [117] and the second by suppressing rapidly accelerated fibrosarcoma (Raf) activation [118] (Figure 3).
The PI3K/AKT pathway is implicated in cell polarization, migration, cell fate determination and apoptosis. For instance, FGF-2 enhances the migratory activity of periodontal ligament cells (PDLSCs) through this pathway. In addition, FGF-2 induced Akt phosphorylation promotes proliferation in neural progenitor cells [103] and MSCs [105]. Sprouty2 has also been associated with the PI3K/Akt pathway, suppressing Akt phosphorylation [101].
The FGFR tyrosine kinase domain can also directly phosphorylate PLCγ, which leads to the hydrolysis of phosphatidylinositol 4,5-bisphosphate to produce inositol triphosphate (IP3) and diacylglycerol. Subsequently, IP3 increases intracellular calcium ion levels, while diacylglycerol activates PKC. Furthermore, it has been shown that FGFs can induce expression of receptor of activated protein C kinase 1 (RACK-1), a protein that further stabilizes activated PKC [119]. It has been shown that FGFR-1 and FGFR-2 can directly bind to activate PLCγ1 [120]. In fact, FGF-2-induced activation of the PLCγ pathway is involved both in increased expression and transcriptional activity of RUNX2 [121]. In dental tissue-derived mesenchymal stem cells, it has been shown that neurogenic differentiation in human dental pulp stem cells (DPSCs) is induced by FGF-2 via the PLCγ signaling pathway [122], which is already known for its role in the differentiation of neuronal cells [123].
Other pathways such as STAT [96] and ribosomal S6 kinase 2 (RSK2) pathways [124] have been shown to be involved in FGF-2 signaling. The activated FGFR also phosphorylates and activates STAT1, STAT3, and STAT5, to regulate STAT pathway target gene expression [79]. These pathways control the steps of osteoblastogenesis resulting in the modulation of bone cell proliferation, differentiation, and apoptosis, depending on the stage of cell differentiation [125,126]. Therefore, normal FGFR activity is critical for the development of numerous types of tissue, including the craniofacial skeleton, as observed in several genetic diseases causing abnormalities in bone and cartilage formation due to mutations in the genes encoding FGFs or their receptors [127,128].

3.2. FGF-2: An Essential Regulator in Skeletal Tissue

FGF-2, also known as basic fibroblast growth factor, is a canonical FGF that belongs to the FGF-1 subfamily. It is encoded by FGF-2, on chromosome 4 [129], and is a wide-spectrum angiogenic, mitogenic and neurotrophic factor expressed by many types of cells in both adult and developmental stages [130]. It is a regulator of proliferation [131], migration [132], differentiation [122,129,133], cell survival [134], and stemness in human stem cells [135,136]. Apart from its key role in angiogenesis, FGF-2 is a major player in skeletal development, bone formation, and fracture repair [137,138,139]. During embryogenesis, it is a strong mesodermal inducer, and its receptors are strongly expressed in developing bones [140,141]. Throughout life, it is constantly expressed in osteoblasts and stored in the ECM [142]. Taken together, these properties make FGF-2 an attractive molecule for clinical and pharmaceutical applications in bone regeneration.
FGF-2 is highly expressed in bone tissues, with several isoforms due to alternative start codons for FGF-2 mRNA translation initiation [143]. The high molecular weight FGF-2 (HMW FGF-2) isoforms (24, 23, and 22 kD) are localized in the nucleus, whereas low molecular weight FGF-2 (LMW FGF-2) (18 kD) is cytoplasmic, and membrane associated. This differential intracellular trafficking also reflects a difference in function. Specifically, the LMW FGF-2 promotes osteoblast differentiation and mineralization via the activation of the Wnt pathway [144] and via synergistic actions with bone morphogenetic protein 2 (BMP-2). Indeed, endogenous FGF/FGFR signaling is a positive upstream regulator of the BMP-2 gene in calvarial osteoblasts [145]. In contrast, HMW FGF-2 acts in the nucleus as a transcriptional factor that upregulates the expression of genes associated with impaired mineralization, such as SOST and FGF-23 [146].
Experimental in vitro evidence regarding FGFR-2 mutations associated with craniosynostosis syndromes highlights the major contribution of FGF-2 to bone cell fate. The genetic inactivation of FGFR-2 causes reduced osteoblast proliferation and increased osteopenia, while FGFR-1 has been associated with stage-dependent regulation of osteoblast proliferation and differentiation [147,148,149].
Exogenous FGF-2 rescued reduced bone nodule formation by upregulating the Wnt/β-catenin pathway in FGF-2-/- osteoblast cultures. [150,151]. Furthermore, when two FGFR-2 mutants from the Apert and Crouzon syndromes were expressed in immature osteoblasts, both inhibited osteoblastic differentiation but also increased apoptosis [152] downregulating many Wnt targets and inducing SRY-box 2 (SOX-2), a transcription factor that maintains the undifferentiated state of cells [153] (Figure 4). Furthermore, in a line of murine mesenchymal progenitor cells, induced overexpression of FGFR-2 led to increased cell proliferation and osteogenic differentiation through ERK1/2 by FGFR-2 signaling [154]. Inversely, primary calvarial osteoblasts from Fgf2-/- mice showed reduced BMP-2 induced periosteal bone formation [155]. In more mature cells, ERK1/2 activation by FGF-2 enhances acetylation and stabilization of RUNX-2, a key transcription factor involved in osteoblastogenesis and bone formation [114,156,157]. In addition, FGFR-2 signaling is crucial for the induction of apoptosis when osteoblasts are well differentiated [158,159], an important step for bone homeostasis. These results underline the dual temporal role of FGF-2 signaling in bone development, this protein promoting proliferation in immature osteoblasts and differentiation in mature ones. Furthermore, it should be underlined that since FGF-2 can bind equally to FGFR-1 and FGFR-2, differential activation of one of them may be responsible for signal transduction towards the proliferation of progenitors or towards osteogenic differentiation in post-proliferating cells [154].
The important dual role of FGF-2 in bone formation is saliently demonstrated by transgenic mouse models. Conditional deletion of fgf-2 in mice yields a skeletal dwarfism phenotype and reduced bone formation [147]. Furthermore, fgf-2 haploinsufficient mice are characterized by generalized osteopenia [151] and fgf-2 knockout mice display greatly reduced trabecular plate-like structures and loss of connecting rods [160]. This reduced bone formation is due to defective osteoblast differentiation and alteration of progenitor cell lineage commitment, FGF-2 deficiency resulting in increased bone marrow adipogenesis and reduced osteogenesis [161]. Nonetheless, overexpression of fgf-2 in mice also gives rise to skeletal defects, including a shortening and flattening of long bones, with a decrease in osteoblast differentiation, impaired bone formation, and moderate macrocephaly [162,163].

4. FGF-2 in Craniofacial Bone Tissue Engineering

In recent decades, tissue engineering has emerged as a new biomedical field with advanced approaches for tissue regeneration and healing [164]. It was initially defined as “the application of the principles of biology and engineering to the development of functional substitutes for damaged tissue” [165]. Bone tissue engineering needs to restore distinct functions: structural (e.g., bone, cartilage), barrier- and transport-related (e.g., blood vessels), and/or biochemical and secretory (e.g., hematopoiesis, calcium metabolism).
Tissue engineering is based on the combination of three basic tools: cells, biomaterials, and suitable biochemical and physical factors, seeking to mimic the physical and functional properties of the natural tissue creating a tissue-like structure [166,167]. The last factor to consider is the presence of exogenous chemical and mechanical stimuli, such as soluble growth and differentiation factors (e.g., BMP, FGF-2, VEGF and TGF-β), and mechanical forces. These factors can be incorporated into a construct during scaffold fabrication itself or included in the culture medium to facilitate the survival, proliferation, and differentiation of the implanted cells and their integration into the host. This section focuses on FGF-2 as a good candidate for craniofacial bone tissue engineering by acting on both angiogenic and osteogenic processes.

4.1. FGF-2: An Exogenous Factor In Vitro

Exogenous FGF-2 administration in vitro has multiple effects on bone cell fate. Firstly, it has been shown that FGF-2 maintains the osteoblast precursor proliferative state [142,168]. Its anabolic effect is evident through the stimulation of bone marrow stem cells, sustaining their osteogenic potential by maintaining the cells in an immature state with a fibroblast-like morphology expressing less alkaline phosphatase (ALP) [169]. DPSCs, stem cells from human exfoliated deciduous teeth (SHEDs) and stem cells from apical papilla (SCAPs) treated with FGF-2 express stemness-related markers including STRO-1 and CD-146 [170,171,172]. Recently, it has been reported that DPSCs/SHEDs displayed increased and prolonged proliferation upon FGF-2 treatment in vitro, together with delayed type 1 collagen expression [173]. On the other hand, human calvaria bone cells grown in mineralizing medium for several weeks with FGF-2 showed no stimulation of proliferation, suggesting that mature bone cells do not respond to FGFs’ mitogenic signal [174]. In fact, the decrease in intrinsic proliferation potential of human mesenchyme-derived progenitor cells with age was partially attributed to a reduction in FGF-2 expression, as observed in elderly humans [175]. In line with this, there is evidence that the efficacy of FGF-2 in inducing bone formation might be maximized if targeting younger cells, such as juvenile osteoblasts [176].
Concomitant with prolonged stemness, the abolition of mineralization by FGF-2 has been shown in various stem cell lines. Shimabukuro et al. [177] demonstrated that treatment of human DPSCs with FGF-2 increased their migration and proliferation ability but also impaired mineralization. On the contrary, if hDPSCs were only FGF-2 pretreated for a short period of time but left to differentiate under normal osteogenic conditions, ALP activity and nodule formation increased. Likewise, other studies confirm that pretreatment with FGF-2 during the proliferation phase leads to increased ALP activity, formation of mineralized nodules and expression of dentin sialoprotein and dentin matrix protein 1 (DMP-1) in hDPSCs [178]; dentin sialophosphoprotein (DSPP) and bone sialoprotein (BSP) expression in immature adult rat incisor dental pulp cells [179]; BSP, osteocalcin (OCN), osteopontin (OPN) and matrix Gla protein (MGP) in cementoblasts [180]; and hyaluronan in hDPSCs [181]. Nauman et al. [182] showed that in vitro administration of FGF-2 to rat osteoprogenitor cells accelerated the mineralization process through alkaline phosphatase and OCN expression and lowered the phosphate threshold needed for successful bone nodule formation. These observations suggest that FGF-2 enhances cell growth at early stages, a step that is crucial for accelerated differentiation at later time points. On the other hand, the spatiotemporal patterns of FGF signaling in vivo may differ from those found in culture conditions. Another possibility is that cell responses to FGF signaling in vivo are determined by, or are coordinated with, signaling from other cytokines in situ, such as BMPs and WNT [75,183].

4.2. FGF-2: An Exogenous Factor In Vivo

FGF-2 alone or in conjunction with other molecules and in combination with several types of scaffold has been assessed in various preclinical models for craniofacial bone regeneration. The rationale of FGF-2 administration is, first, its direct impact on bone cell proliferation and differentiation and, second, its pro-angiogenic action at the site of bone regeneration.
FGF-2 seems equally beneficial when tested in bone regeneration models of intramembranous or endochondral ossification. FGF-2 can be expressed by differentiating osteoblasts at sites of intramembranous ossification or by growth plate chondrocytes [128]. Its administration in vivo promotes regeneration of cranial [184,185], and periodontal [186,187,188] bone defects, as well as being associated with a shorter timeline of craniofacial bone repair [173]. Indeed, it has also been shown that FGF-2 was able to partially restore the lost cancellous bone mass in the ovariectomized rat [138]. The addition of FGF-2 significantly increased central defect bone filling in aged mice, leading to qualitatively superior bone formation. This suggests that FGF-2 is a good candidate for boosting bone regeneration in areas with impaired angiogenic potential or small numbers of native osteoprogenitor cells [189]. Similar benefits in angiogenesis and bone formation have been found in critical size defects in rat calvaria [190,191]. Furthermore, controlled delivery of FGF-2 in combination with a low dose of BMP-2 improved aged murine calvaria bone defect healing as compared to treatment with BMP-2 alone and the use of bone substitute impregnated with BMP-2 and FGF-2 promoted periodontal regeneration in non-human primates [189,192].
Vascularization plays a crucial role in bone tissue engineering seeking to replace large tissue losses due to trauma, surgery, or other clinical scenarios where spontaneous bone repair is not feasible. Indeed, using a radiotracer, Collignon et al. observed a correlation between early angiogenesis assessed by positron emission tomography and bone formation determined by micro-computed tomography within mouse calvarial bone critical size defects [193]. Recently, Novais et al. found that FGF-2 priming of DPSCs/SHEDs boosted intramembranous bone formation in critical size calvaria defects in immunodeficient mice [173]. Indeed, priming these cells with FGF-2 greatly enhanced stem cell early proliferation leading to increased bone regeneration concomitant with the expression of mineralization markers such as OPN, DMP1, or ALP.
These results suggest the importance of vascularization in bone regeneration. In fact, upon implantation in vivo, a major challenge is the maintenance of cell viability in the bone graft core, which critically depends on rapid invasion by host blood vessels. A functionally perfused vascular network will ensure oxygen and nutrient transport and waste removal. In this regard, endothelial cells play a key role in tissue regeneration and remodeling, since they can facilitate the recruitment of osteoprogenitors and immune cells, through the secretion of osteogenic factors, such as BMPs [194]. A recent study, with involvement of our research group, has shown implantation of a pre-vascularized scaffold network engineered in vitro to be a promising strategy for promoting blood supply deep into the graft, relying on inosculation with the host vasculature [195]. In particular, it has demonstrated the importance of grafting a mature microvascular network, displaying perivascular recruitment through the PDGF-BB pathway and basement membrane remodeling, taking advantage of the angiogenic properties of DPSCs/SHEDs and allowing self-assembly of endothelial cells into capillaries. Interestingly, we previously reported that the subcutaneous implantation of tissue-engineered constructs seeded with DPSCs/SHEDs primed with FGF-2, greatly enhanced vascularization within constructs thanks to the capacity of DPSCs/SHEDs to constitutively secrete hepatocyte growth factor (HGF) [170]. We have also demonstrated that FGF-2 is instrumental in promoting both VEGF and HGF secretion by DPSCs/SHEDs. Indeed, recruited stem cells participated in the deposition of vascular basement membrane and vessel maturation in athymic nude mice demonstrated the importance of in vitro production of mature microvasculature for improving cell-based therapies [195].

4.3. Human Clinical Applications of FGF-2 in the Craniofacial Area

The observations described above have been replicated in human clinical studies more recently.
Indeed, rhFGF-2 is already used in the clinical treatment of orofacial tissues. Kitamura et al. [196] performed a double-blind randomized controlled trial with 253 patients receiving rhFGF-2 0.2%, 0.3% or 0.4% or placebo during surgical management of periodontal intrabony defects. At 36 weeks, all FGF-2-treated groups demonstrated significantly higher radiographic bone fill than the placebo group, 0.3% being the best concentration. In addition, secondary analysis in a subgroup of patients showed very low levels of FGF-2 in serum and no adverse effects were reported. A randomized controlled trial of 30 patients showed an improvement in pocket depth reduction and more clinical attachment gain compared to control sites [197]. Application of FGF-2 for the treatment of intrabony defects has been studied in another randomized controlled trial, where various concentrations of FGF-2 were used mounted in β-tricalcium phosphate scaffolds. At 6 months, patients treated with 0.3% or 0.4% rhFGF-2 showed 71% success for the combined outcome of attachment gain of 1.5 mm and bone fill of 2.5 mm compared to 45% success in the 0.1% FGF-2 and control groups [198]. A meta-analysis of studies using recombinant human FGF-2 for the treatment of deep intrabony periodontal defects demonstrated a clinical benefit of FGF-2 in terms of bone fill [199]. A more recent meta-analysis of six randomized controlled trials shows that administration of 0.4% rhFGF-2 yielded 22% higher bone fill of periodontal defects than control treatment, though this result was not statistically significant. It also indicated that the impact of the treatment was dose dependent, with higher FGF-2 concentrations producing better bone regeneration outcomes [200]. To date, however, there is still no consensus on the optimal dose or delivery scaffolding method for the use of FGF-2 in the field of bone regeneration.

4.4. FGF-2 as an Exogenous Factor: A Synthesis of Current Knowledge

It is evident from animal models and human clinical application studies that exogenous administration of FGF-2 is a promising method for accelerating craniofacial bone regeneration. Given the spatiotemporal effect of FGF-2 on mineralization, a key challenge is to determine the optimal application strategy. Parameters such as dose, length of exposure, administration mode, and type of scaffolding, as well as the origin and differentiation state of the stem cells employed for craniofacial bone tissue engineering appear to be crucial. These parameters are discussed in the following sections and summarized in Table 1.

4.4.1. Dosage (In Vitro/In Vivo)

In vitro, FGF action is complex, and the biological effect of FGF-2 may depend on the dose, length and mode of exposure. Mouse bone chip outgrowth cells that were primed with FGF-2 (0, 0.0016, 0.016 or 0.16 ng/mL) demonstrated dose-dependent expression of mesenchymal markers, suggesting dose-dependent anabolic action of FGF-2 on proliferation [201]. Human mesenchyme-derived progenitor cells from cancellous bone were harvested from young and old patients and cultured under various FGF-2 concentrations (0.0016, 0.016, 0.16 and 1.6 ng/mL) for 4, 24, 28 and 72 h. Responsiveness regarding proliferation was dose and age dependent, with the proliferation rate diminishing with age [202]. Despite the inhibitory effect on differentiation and mineralization, the addition of FGF-2 to culture medium maintains stemness in SHEDs and embryonic stem cells [203], and it has been shown to be necessary to maintain the cells in a pluripotent state [204]. Indeed, continuous treatment of cultured osteoprogenitors with 10 ng/mL of rhFGF-2 over 2 days significantly reduced expression of alkaline phosphatase, osteopontin and collagen I expression, though RUNX-2 mRNA levels were not altered, indicating that cells treated with FGF-2 retain their osteogenic commitment [145]. Threshold doses vary between studies (e.g., Varkey et al. demonstrated that concentrations higher than 2 ng/mL inhibit proliferation and differentiation of bone marrow mesenchymal cells) [205].
In vivo, several studies have been conducted to assess the performance of FGF-2 in various administration modes in vivo with varying dosages depending on the animal model and bone defect configuration. A single local injection of FGF-2 directly at the site of interest has shown to increase periosteal bone formation in murine calvaria [206] and facilitate the healing of bone fracture and segmental bone defect in rats [137], rabbits [207,208], dogs [209], and non-human primates [210,211]. Kamo et al. compared the effect of a single local injection with that of cyclical injections of FGF-2 on a cancellous bone defect in the femoral condyle of rabbits. Only the high-dose single injection (1.2 μg/μL), and not the low-dose single injection (0.4 μg/μL) or cyclical injections (0.4 μg/μL, 3 times), significantly increased cancellous bone volume as measured by bone histomorphometry [212]. These results indicate that the effect of local injections of FGF-2 on cancellous bone regeneration is greater at the very early stage of bone healing. Thus, the positive effect on proliferation is useful, but only if it is temporary and regulated. A similar conclusion can be drawn from the results of Novais et al. with short FGF-2 priming of SHEDs/DPSCs before their implantation in the calvaria defect [173].
On the other hand, some animal studies have shown the efficacy of FGF-2 with a prolonged administration mode. The anabolic effect of FGF-2 was observed with daily systemic administration for 2 weeks in a rat bone defect model [213,214,215]. Lane et al. found that FGF-2 treatment for 14 days (1 mg/kg/rat) was associated with the development of new trabecular elements, but with the withdrawal of FGF-2 injections, the new trabeculae were rapidly lost thorough accelerated resorption [216]. Furthermore, it has been reported that continuous local infusion of bFGF using an osmotic pump is able to shorten the consolidation phase of limb lengthening in rabbits [217].

4.4.2. Scaffolding and Stabilization by Administration Mode

The dose is not the only parameter to consider. The administration mode influences the stability of FGF-2. Indeed, the inherent instability of this protein in aqueous solutions necessitates its delivery via biomimetic scaffolds that can stabilize and maximize its biological activity for a defined period of time [218]. FGF-2 exhibits a short half-life of 12 h in vivo due to degradation by proteolytic enzymes but also because of its instability as soon as it is thawed [219,220]. A crucial factor for the sustained release of FGF-2 is the resorption rate of the scaffold in which the growth factor is impregnated. A well-documented method is the use of gelatin hydrogels for the fabrication of FGF-2-loaded scaffolds for tissue regeneration applications since they can mimic the manner in which FGF-2 is stored in the ECM. Indeed, when FGF-2 in solution was directly injected ectopically in mice, the vascularization process remained unchanged, whereas the incorporation of FGF-2 into a gelatin hydrogel greatly enhanced neovascularization. Moreover, hydrogels with less water content (77.5% vs. 95.9%) were more efficacious in sustaining FGF-2 release due to their slower resorption rate [221]. Biodegradable gelatin hydrogel incorporating rhFGF-2 has been developed successfully in Japan and shown to restore bone [219,222,223].
FGF-2-mediated tissue regeneration has also been tested with chitosan/collagen scaffolding. FGF-2 controlled release by a chitosan/fucoidan complex hydrogel is presumed to immobilize it, prolong its biological half-life time and protect it from inactivation by heat or proteolysis. After injection of this hydrogel into an ectopic murine model, significant neovascularization was observed, attributed to both slow diffusion of FGF-2 and controlled biodegradation of the hydrogel [183]. Similar findings were reported with the use of heparin/protamine water-insoluble microparticles for FGF-2 delivery [224]. Various other heparin-mimicking molecules have been tested for FGF-2 administration, such as heparin mimetic peptide nanofibers [225], sulfated peptides [226], sulfonated dextrans [227] and polysulfonated polymers [228,229]. Combining poly (lactic-co-glycolic acid) (PLGA) and poly (vinyl alcohol) (PVA) to produce FGF-2-loaded microspheres has also shown to sustain the release and stability of FGF-2 for up to 4 days in a culture of human embryonic stem cells. Furthermore, FGF-2-loaded polycaprolactone (PCL) microspheres have been reported to enhance angiogenesis in vivo [230], and the use of microspheres constructed from combined alginate/collagen hydrogels yields a scaffold that provides controlled release of FGF-2 and enhances angiogenesis [231].
Radomsky et al. showed that a single local injection of FGF-2 in a hyaluronan gel, an ECM component, significantly promoted fracture healing of the fibulae in baboons, as evidenced by increased callus formation and mechanical strength [211]. Tabata et al. reported that FGF-2 incorporated into gelatin hydrogel induced bone formation at the site of a skull defect in non-human primates [184]. More recently, Murahashi et al. [232] have developed multi-layered FGF-2-loaded poly L-lactic acid nanosheets. Their subcutaneous application allowed the sustained release of loaded rhFGF-2 for about 2 weeks and enhanced bone regeneration upon implantation at critical size femoral murine defects. Controlling rhFGF-2 stability and delivery will make it possible to adapt the dose and length of exposure necessary to the bone defect to be repaired.

4.4.3. BMP-2: An Interesting Cytokine for Combined Treatments

FGF action is complex, and the biological effect of FGF-2 may depend on its interaction with other cytokines. The combination of FGF-2 and BMP-2 has already been tested at various concentrations and kinetics actions. A recent in vitro study suggests that sheep BMSCs supplemented with 20 ng/mL of FGF-2 and 100 ng/mL of BMP-2 may be a feasible cellular therapy for bone regeneration [233]. In vivo, the association of 10 μg of FGF-2 and 10 μg of BMP-2 in transfected human MSCs yielded a significantly increased bone regeneration of critical size calvarial defects of nude mice [234]. Indeed, implanted calcium phosphate ceramic tubes loaded with rat marrow MSCs preconditioned with both FGF-2 and BMP-2 yielded better bone formation than FGF-2 or BMP-2 treatment alone [235]. In addition, a novel biomimetic coating scaffold (calcium phosphate/polyelectrolyte multilayer (bCaP-PEM)) capable of sequential delivery indicated that FGF-2 delivery followed by BMP-2 increased bone regeneration in adult mouse calvarial bone defects more than delivery of BMP2 alone [201]. The combined action of FGF-2 and BMP-2 is also dose dependent. Notably, scaffolds loaded with 2 μg of BMP-2 on collagen disks enhanced osteoinduction when FGF-2 was in the range of 16–400 ng but inhibited with 10 or 50 μg of FGF-2 [236]. Similarly, using implants in rats, Takita et al. [237] found that 100 ng of FGF-2 was able to enhance BMP-2 (0.8 μg), inducing ectopic bone formation, whereas a higher dose of FGF-2 (10 μg) exerted an inhibitory effect. Higher doses may keep cells in an undifferentiated state for a longer time, this negatively impacting mineralization [173]. A possible mode of action is that high doses of FGF-2 do not increase the expression of BMP receptors, whereas low doses of FGF-2 strengthen bone formation via BMP-2 signaling as shown by increased Smad 1 expression, a major downstream effector in the BMP signaling pathway [238].

5. Discussion, Conclusions and Perspectives

Bone is a constantly evolving mineralized and vascularized connective tissue that can be repaired by simple immobilization in the case of non-displaced fractures. During major trauma, infection or cancer; however, bone loses its capacity to self-repair.
Tissue engineering is considered a therapy of the future, making it possible to clinically overcome the many limitations of current autologous graft therapies (notably, the limited quantity of tissue available and risk associated with several surgical sites on a single patient). The combination of biomaterials, cells and molecules of interest may allow great advances at the clinical level by stimulating the integration of grafts through vascularization and mineralization. The close relationship between blood vessels and bone cells has been demonstrated in studies on skeletal malformation, such as craniofacial dysmorphology.
FGF-2, by virtue of its proliferative, pro-angiogenic and pro-osteogenic properties, is one of the molecules studied in this line of research. This growth factor is a ubiquitous molecule present from the embryonic stage and throughout life and is involved in the formation of the ECM. It plays an important role in the homeostasis, repair and metabolism of bone tissue by regulating the proliferation and differentiation of osteoblasts, accelerating the healing of fractures and the repair of skeletal defects.
The effects of FGF-2 differ depending on the type and stage of cell differentiation. Some results appear to be contradictory but can be explained by differences in dose, mode of administration and lengths of exposure, as well as by the models used in in vitro or in vivo studies, each of which has certain biases.
It appears that high dose and/or long-term treatment inhibit bone regeneration. This could be explained by a positive effect on proliferation, which remains essential at the beginning, but must be temporary, and therefore regulated. Thus, low dose and/or short-term treatment may provide the best conditions for bone regeneration. There is a need to explore further the positive or negative effects on bone regeneration of other parameters, such as the type of culture medium used and any supplements added to boost the osteogenic effect (ascorbic acid, dexamethasone, β-GP, etc.). The way in which FGF-2 is delivered should also be considered, since it may affect bone repair, potentially leading to unwanted side effects. Moreover, the delivery mode seems to have a non-negligible impact on the stability of this cytokine, and therefore, must be carefully planned and tested before clinical use. Identifying the ideal dose and how to deliver it over a given time within a specific repair time frame remain the key challenges in this type of tissue-engineered therapy.

Author Contributions

A.N.: investigation, writing, and visualization; E.C.: Investigation, and writing; C.C.: supervision, and writing—review; and C.G.: investigation, and writing—original draft and review. All authors have read and agreed to the published version of the manuscript.

Funding

From Université de Paris, ANR PulpCell and Fondation Gueules Cassées for URP2496, and Fondation pour la Recherche Médicale (PhD grant to EC).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Figure A1. Morphology of cortical and trabecular bone—The cortical bone is dense and compact with penetrating vascular canals, that has slow turnover rate and high resistance to torsional and bending forces. It constitutes 80% of the skeleton, and it makes up the outer part of skeletal structures. This bone has an outer periosteal surface containing blood vessels, nerve endings, osteoblasts, and osteoclasts and an inner endosteal surface adjacent to the marrow. This tissue is arranged in osteons (A), which are concentric layers, composed of collagen fibers. These are made up of three helical chains and combine to form fibrils, which are interwoven and bound by crosslinks, providing bone elasticity, flexibility, and tensile strength. Cortical bone provides mechanical strength and protection, and it may also participate in metabolic responses, particularly when there is a long-lasting mineral deficit. In contrast, trabecular bone represents just 20% of the skeletal mass, but 80% of the bone surface. This type of bone, which is less dense and more elastic, has a higher turnover rate than cortical bone and has high resistance to compression. It provides mechanical support, helping to maintain skeletal strength and integrity with its rods and plates aligned in a pattern that provides maximal strength. It also exhibits greater metabolic activity than cortical bone, having a larger surface area for mineral exchange. These properties explain it being found inside the long bones, throughout the bodies of the vertebrae, and in the inner portions of the pelvis and other flat bones.
Figure A1. Morphology of cortical and trabecular bone—The cortical bone is dense and compact with penetrating vascular canals, that has slow turnover rate and high resistance to torsional and bending forces. It constitutes 80% of the skeleton, and it makes up the outer part of skeletal structures. This bone has an outer periosteal surface containing blood vessels, nerve endings, osteoblasts, and osteoclasts and an inner endosteal surface adjacent to the marrow. This tissue is arranged in osteons (A), which are concentric layers, composed of collagen fibers. These are made up of three helical chains and combine to form fibrils, which are interwoven and bound by crosslinks, providing bone elasticity, flexibility, and tensile strength. Cortical bone provides mechanical strength and protection, and it may also participate in metabolic responses, particularly when there is a long-lasting mineral deficit. In contrast, trabecular bone represents just 20% of the skeletal mass, but 80% of the bone surface. This type of bone, which is less dense and more elastic, has a higher turnover rate than cortical bone and has high resistance to compression. It provides mechanical support, helping to maintain skeletal strength and integrity with its rods and plates aligned in a pattern that provides maximal strength. It also exhibits greater metabolic activity than cortical bone, having a larger surface area for mineral exchange. These properties explain it being found inside the long bones, throughout the bodies of the vertebrae, and in the inner portions of the pelvis and other flat bones.
Cells 10 00932 g0a1
Table A1. Comparison of endochondral and intramembranous ossification
Table A1. Comparison of endochondral and intramembranous ossification
Endochondral OssificationIntramembranous Ossification
ProcessReplacement of cartilage
with bone
Direct conversion of
mesenchyme to bone
SiteMainly long bonesMainly flat bones
Cellular
embryonic origin
Neural crest-derived
mesenchymal cells
Mesoderm-derived
mesenchymal cells
Functional cellsChondrocytes that secrete
ECM to form cartilage
Osteoblasts that secrete
osteoid matrix
There are two different processes of bone modeling: endochondral ossification, involving cartilage as an intermediate stage, and intramembranous ossification, the chondrocyte-to-osteoblast trans-differentiation, which involves direct differentiation of MSCs into osteoblasts. The former is typical for long bones, while the latter is the main mechanism for the development of flat bones, such as those of the craniofacial skeleton [239,240].
Table A2. Molecules with angiogenic and osteogenic effects.
Table A2. Molecules with angiogenic and osteogenic effects.
Angiogenic EffectOsteogenic EffectFunction
BMPYes (indirectly) [241]Yes [242]Differentiation of osteoblast-like cells chemoattractant for neighboring
endothelial cells (ECs) [241,243,244]; proliferation and differentiation of mesenchymal osteoprogenitors [245]; BMP-2 and -7 enhance bone formation and repair, through induction of vascular endothelial growth
factor (VEGF) expression and angiogenesis stimulation [246,247]
TGF-βYes, but role not well understood [248]Yes [247]Chemoattractant for mesenchymal stem cells, differentiation of
osteoblasts [249,250,251], chondroblast, and osteoprogenitor cells and
matrix production stimulation in healing process [252,253]
PDGFYesYesChemoattractant for and mitogenic stimulation of osteoblasts [251,254]
MMPYesYesMatrix metallopeptidase 9 (MMP9) induces vascularization in bone
formation by the release of VEGF from ECM [255]; while
matrix metallopeptidase 13 (MMP13) induces osteogenesis [256]
Notch
signaling
YesYesNotch pathway modulates the angiogenic effect of VEGF in ECs.
Activation of Notch signaling in bone ECs promotes local
angiogenesis and osteogenesis [257].

References

  1. Clarke, B. Normal Bone Anatomy and Physiology. Clin. J. Am. Soc. Nephrol. 2008, 3, S131–S139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. DiGirolamo, D.J.; Clemens, T.L.; Kousteni, S. The skeleton as an endocrine organ. Nat. Rev. Rheumatol. 2012, 8, 674–683. [Google Scholar] [CrossRef] [PubMed]
  3. Oldknow, K.J.; Macrae, V.E.; Farquharson, C. Endocrine role of bone: Recent and emerging perspectives beyond osteocalcin. J. Endocrinol. 2015, 225, R1–R19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kenkre, J.S.; Bassett, J.H. The bone remodelling cycle. Ann. Clin. Biochem. Int. J. Lab. Med. 2018, 55, 308–327. [Google Scholar] [CrossRef] [PubMed]
  5. Hadjidakis, D.J.; Androulakis, I.I. Bone remodeling. Ann. N. Y. Acad. Sci. 2006, 1092, 385–396. [Google Scholar] [CrossRef] [PubMed]
  6. Alford, A.I.; Kozloff, K.M.; Hankenson, K.D. Extracellular matrix networks in bone remodeling. Int. J. Biochem. Cell Biol. 2015, 65, 20–31. [Google Scholar] [CrossRef]
  7. Yang, G.; Zhu, L.; Hou, N.; Lan, Y.; Wu, X.-M.; Zhou, B.; Teng, Y.; Yang, X. Osteogenic fate of hypertrophic chondrocytes. Cell Res. 2014, 24, 1266–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Matic, I.; Matthews, B.G.; Wang, X.; Dyment, N.A.; Worthley, D.L.; Rowe, D.W.; Grcevic, D.; Kalajzic, I. Quiescent Bone Lining Cells Are a Major Source of Osteoblasts During Adulthood. Stem Cells 2016, 34, 2930–2942. [Google Scholar] [CrossRef] [Green Version]
  9. Murshed, M.; Harmey, D.; Millán, J.L.; McKee, M.D.; Karsenty, G. Unique coexpression in osteoblasts of broadly expressed genes accounts for the spatial restriction of ECM mineralization to bone. Genes Dev. 2005, 19, 1093–1104. [Google Scholar] [CrossRef] [Green Version]
  10. Katsimbri, P. The biology of normal bone remodelling. Eur. J. Cancer Care 2017, 26, e12740. [Google Scholar] [CrossRef]
  11. Franz-Odendaal, T.A.; Hall, B.K.; Witten, P.E. Buried alive: How osteoblasts become osteocytes. Dev. Dyn. 2005, 235, 176–190. [Google Scholar] [CrossRef]
  12. Bonewald, L.F. The amazing osteocyte. J. Bone Miner. Res. 2011, 26, 229–238. [Google Scholar] [CrossRef]
  13. Xiao, W.; Wang, Y.; Pacios, S.; Li, S.; Graves, D.T. Cellular and Molecular Aspects of Bone Remodeling. Craniofac. Sutures 2015, 18, 9–16. [Google Scholar] [CrossRef]
  14. Bonewald, L.F.; Johnson, M.L. Osteocytes, mechanosensing and Wnt signaling. Bone 2008, 42, 606–615. [Google Scholar] [CrossRef] [Green Version]
  15. Dallas, S.L.; Prideaux, M.; Bonewald, L.F. The Osteocyte: An Endocrine Cell … and More. Endocr. Rev. 2013, 34, 658–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Lanyon, L.E. Osteocytes, strain detection, bone modeling and remodeling. Calcif. Tissue Int. 1993, 53, S102–S107. [Google Scholar] [CrossRef]
  17. Sims, A.N.; Martin, T.J. Coupling the activities of bone formation and resorption: A multitude of signals within the basic multicellular unit. Bonekey Rep. 2014, 3, 481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Teitelbaum, S.L. Bone Resorption by Osteoclasts. Science 2000, 289, 1504–1508. [Google Scholar] [CrossRef] [PubMed]
  19. Mackie, E.J.; Tatarczuch, L.; Mirams, M. The skeleton: A multi-functional complex organ. The growth plate chondrocyte and endochondral ossification. J. Endocrinol. 2011, 211, 109–121. [Google Scholar] [CrossRef] [PubMed]
  20. Brodsky, B.; Persikov, A.V. Molecular Structure of the Collagen Triple Helix. Adv. Protein Chem. 2005, 70, 301–339. [Google Scholar] [CrossRef] [PubMed]
  21. Ducy, P.; Desbois, C.; Boyce, B.; Pinero, G.; Story, B.; Dunstan, C.; Smith, E.; Bonadio, J.; Goldstein, S.; Gundberg, C.; et al. Increased bone formation in osteocalcin-deficient mice. Nat. Cell Biol. 1996, 382, 448–452. [Google Scholar] [CrossRef] [Green Version]
  22. Luo, G.; Ducy, P.; McKee, M.D.; Pinero, G.J.; Loyer, E.; Behringer, R.R.; Karsenty, G. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nat. Cell Biol. 1997, 386, 78–81. [Google Scholar] [CrossRef] [PubMed]
  23. Burr, D.; Schaffler, M.; Yang, K.; Wu, D.; Lukoschek, M.; Kandzari, D.; Sivaneri, N.; Blaha, J.; Radin, E. The effects of altered strain environments on bone tissue kinetics. Bone 1989, 10, 215–221. [Google Scholar] [CrossRef]
  24. Krahl, H.; Michaelis, U.; Pieper, H.G.; Quack, G.; Montag, M. Stimulation of bone growth through sports. A radiologic investigation of the upper extremities in professional tennis players. Am. J. Sports Med. 1994, 22, 751–757. [Google Scholar] [CrossRef] [PubMed]
  25. Feng, X.; McDonald, J.M. Disorders of Bone Remodeling. Annu. Rev. Pathol. Mech. Dis. 2011, 6, 121–145. [Google Scholar] [CrossRef] [Green Version]
  26. Kobayashi, S.; Takahashi, H.; Ito, A.; Saito, N.; Nawata, M.; Horiuchi, H.; Ohta, H.; Iorio, R.; Yamamoto, N.; Takaoka, K. Trabecular minimodeling in human iliac bone. Bone 2003, 32, 163–169. [Google Scholar] [CrossRef]
  27. Frost, H.M. Skeletal structural adaptations to mechanical usage (SATMU): Mechanical influences on intact fibrous tissues. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 1990, 226, 433–439. [Google Scholar] [CrossRef]
  28. Manolagas, S.C. Birth and death of bone cells: Basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr. Rev. 2000, 21, 115–137. [Google Scholar] [PubMed] [Green Version]
  29. Takayanagi, H. Osteoimmunology: Shared mechanisms and crosstalk between the immune and bone systems. Nat. Rev. Immunol. 2007, 7, 292–304. [Google Scholar] [CrossRef] [PubMed]
  30. Boyle, W.J.; Simonet, W.S.; Lacey, D.L. Osteoclast differentiation and activation. Nat. Cell Biol. 2003, 423, 337–342. [Google Scholar] [CrossRef] [PubMed]
  31. Hsu, H.; Lacey, D.L.; Dunstan, C.R.; Solovyev, I.; Colombero, A.; Timms, E.; Tan, H.-L.; Elliott, G.; Kelley, M.J.; Sarosi, I.; et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc. Natl. Acad. Sci. USA 1999, 96, 3540–3545. [Google Scholar] [CrossRef] [Green Version]
  32. Kong, Y.-Y.; Yoshida, H.; Sarosi, I.; Tan, H.-L.; Timms, E.; Capparelli, C.; Morony, S.; Oliveira-Dos-Santos, A.J.; Van, G.; Itie, A.; et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nat. Cell Biol. 1999, 397, 315–323. [Google Scholar] [CrossRef] [PubMed]
  33. Belibasakis, G.N.; Bostanci, N. The RANKL-OPG system in clinical periodontology. J. Clin. Periodontol. 2012, 39, 239–248. [Google Scholar] [CrossRef] [Green Version]
  34. Simonet, W.; Lacey, D.; Dunstan, C.; Kelley, M.; Chang, M.-S.; Lüthy, R.; Nguyen, H.; Wooden, S.; Bennett, L.; Boone, T.; et al. Osteoprotegerin: A Novel Secreted Protein Involved in the Regulation of Bone Density. Cell 1997, 89, 309–319. [Google Scholar] [CrossRef] [Green Version]
  35. Hofbauer, L.C.; Khosla, S.; Dunstan, C.R.; Lacey, D.L.; Boyle, W.J.; Riggs, B.L. The Roles of Osteoprotegerin and Osteoprotegerin Ligand in the Paracrine Regulation of Bone Resorption. J. Bone Miner. Res. 2000, 15, 2–12. [Google Scholar] [CrossRef] [PubMed]
  36. Takahashi, N.; Maeda, K.; Ishihara, A.; Uehara, S.; Kobayashi, Y. Regulatory mechanism of osteoclastogenesis by RANKL and Wnt signals. Front. Biosci. 2011, 16, 21–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Day, T.F.; Guo, X.; Garrett-Beal, L.; Yang, Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev. Cell 2005, 8, 739–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Glass, D.A., II.; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev. Cell 2005, 8, 751–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Daoussis, D.; Andonopoulos, A.P. The Emerging Role of Dickkopf-1 in Bone Biology: Is It the Main Switch Controlling Bone and Joint Remodeling? Semin. Arthritis Rheum. 2011, 41, 170–177. [Google Scholar] [CrossRef]
  40. Caetano-Lopes, J.; Canhão, H.; Fonseca, J.E. Osteoblasts and bone formation. Acta Reumatol. Port. 2007, 32, 103–110. [Google Scholar]
  41. Carter, P.H.; Schipan, E. The roles of parathyroid hormone and calcitonin in bone remodeling: Prospects for novel therapeutics. Endocr. Metab. Immune Disord. Drug Targets 2006, 6, 59–76. [Google Scholar] [CrossRef]
  42. Bassett, J.H.D.; Williams, G.R. Role of Thyroid Hormones in Skeletal Development and Bone Maintenance. Endocr. Rev. 2016, 37, 135–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Iglesias, L.; Yeh, J.K.; Castro-Magana, M.; Aloia, J.F. Effects of growth hormone on bone modeling and remodeling in hypophysectomized young female rats: A bone histomorphometric study. J. Bone Miner. Metab. 2010, 29, 159–167. [Google Scholar] [CrossRef] [PubMed]
  44. Weinstein, R.S.; Jilka, R.L.; Parfitt, A.M.; Manolagas, S.C. Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on bone. J. Clin. Investig. 1998, 102, 274–282. [Google Scholar] [CrossRef] [Green Version]
  45. Nakamura, T.; Imai, Y.; Matsumoto, T.; Sato, S.; Takeuchi, K.; Igarashi, K.; Harada, Y.; Azuma, Y.; Krust, A.; Yamamoto, Y.; et al. Estrogen Prevents Bone Loss via Estrogen Receptor α and Induction of Fas Ligand in Osteoclasts. Cell 2007, 130, 811–823. [Google Scholar] [CrossRef]
  46. Roodman, G.D. Role of cytokines in the regulation of bone resorption. Calcif. Tissue Int. 1993, 53, S94–S98. [Google Scholar] [CrossRef] [PubMed]
  47. Graves, D.T.; Oates, T.; Garlet, G.P. Review of osteoimmunology and the host response in endodontic and periodontal lesions. J. Oral Microbiol. 2011, 3, 3. [Google Scholar] [CrossRef] [PubMed]
  48. Raisz, L. Prostaglandins and bone: Physiology and pathophysiology. Osteoarthr. Cartil. 1999, 7, 419–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Brandi, M.L.; Collin-Osdoby, P. Vascular Biology and the Skeleton. J. Bone Miner. Res. 2006, 21, 183–192. [Google Scholar] [CrossRef]
  50. Fang, T.D.; Salim, A.; Xia, W.; Nacamuli, R.P.; Guccione, S.; Song, H.M.; Carano, R.A.; Filvaroff, E.H.; Bednarski, M.D.; Giaccia, A.J.; et al. Angiogenesis Is Required for Successful Bone Induction During Distraction Osteogenesis. J. Bone Miner. Res. 2005, 20, 1114–1124. [Google Scholar] [CrossRef] [PubMed]
  51. Seebach, C.; Henrich, D.; Wilhelm, K.; Barker, J.H.; Marzi, I. Endothelial Progenitor Cells Improve Directly and Indirectly Early Vascularization of Mesenchymal Stem Cell-Driven Bone Regeneration in a Critical Bone Defect in Rats. Cell Transplant. 2012, 21, 1667–1677. [Google Scholar] [CrossRef] [Green Version]
  52. Carvalho, R.; Einhorn, T.; Lehmann, W.; Edgar, C.; Al-Yamani, A.; Apazidis, A.; Pacicca, D.; Clemens, T.; Gerstenfeld, L. The role of angiogenesis in a murine tibial model of distraction osteogenesis. Bone 2004, 34, 849–861. [Google Scholar] [CrossRef]
  53. Percival, C.J.; Richtsmeier, J.T. Angiogenesis and intramembranous osteogenesis. Dev. Dyn. 2013, 242, 909–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Maes, C.; Clemens, T.L. Angiogenic–osteogenic coupling: The endothelial perspective. Bonekey Rep. 2014, 3, 578. [Google Scholar] [CrossRef] [Green Version]
  55. Schipani, E.; Maes, C.; Carmeliet, G.; Semenza, G.L. Regulation of Osteogenesis-Angiogenesis Coupling by HIFs and VEGF. J. Bone Miner. Res. 2009, 24, 1347–1353. [Google Scholar] [CrossRef] [PubMed]
  56. Sivaraj, K.K.; Adams, R.H. Blood vessel formation and function in bone. Development 2016, 143, 2706–2715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Jośko, J.; Gwóźdź, B.; Jedrzejowska-Szypułka, H.; Hendryk, S. Vascular endothelial growth factor (VEGF) and its effect on angiogenesis. Med. Sci. Monit. 2001, 6, 1047–1052. [Google Scholar]
  58. Bluteau, G.; Julien, M.; Magne, D.; Mallein-Gerin, F.; Weiss, P.; Daculsi, G.; Guicheux, J. VEGF and VEGF receptors are differentially expressed in chondrocytes. Bone 2007, 40, 568–576. [Google Scholar] [CrossRef] [PubMed]
  59. Hu, K.; Olsen, B.R. Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J. Clin. Investig. 2016, 126, 509–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Bouletreau, P.J.; Warren, S.M.; Spector, J.A.; Peled, Z.M.; Gerrets, R.P.; Greenwald, J.A.; Longaker, M.T. Hypoxia and VEGF Up-Regulate BMP-2 mRNA and Protein Expression in Microvascular Endothelial Cells: Implications for Fracture Healing. Plast. Reconstr. Surg. 2002, 109, 2384–2397. [Google Scholar] [CrossRef] [PubMed]
  61. Mayr-Wohlfart, U.; Waltenberger, J.; Hausser, H.; Kessler, S.; Günther, K.-P.; Dehio, C.; Puhl, W.; Brenner, R. Vascular endothelial growth factor stimulates chemotactic migration of primary human osteoblasts. Bone 2002, 30, 472–477. [Google Scholar] [CrossRef]
  62. Henriksen, K.; Karsdal, M.; Delaissé, J.M.; Engsig, M.T. RANKL and vascular endothelial growth factor (VEGF) induce osteoclast chemotaxis through an ERK1/2-dependent mechanism. J. Biol. Chem. 2003, 278, 48745–48753. [Google Scholar] [CrossRef] [Green Version]
  63. Carlevaro, M.F.; Cermelli, S.; Cancedda, R.; Cancedda, F.D. Vascular endothelial growth factor (VEGF) in cartilage neovascularization and chondrocyte differentiation: Auto-paracrine role during endochondral bone formation. J. Cell Sci. 2000, 113, 59–69. [Google Scholar]
  64. Street, J.; Bao, M.; DeGuzman, L.; Bunting, S.; Peale, F.V., Jr.; Ferrara, N.; Steinmetz, H.; Hoeffel, J.; Cleland, J.L.; Daugherty, A.; et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc. Natl. Acad. Sci. USA 2002, 99, 9656–9661. [Google Scholar] [CrossRef] [Green Version]
  65. Kim, Y.-M.; Lee, Y.M.; Kim, H.-S.; Kim, J.D.; Choi, Y.; Kim, K.-W.; Lee, S.-Y.; Kwon, Y.-G. TNF-related Activation-induced Cytokine (TRANCE) Induces Angiogenesis through the Activation of Src and Phospholipase C (PLC) in Human Endothelial Cells. J. Biol. Chem. 2002, 277, 6799–6805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Min, J.K.; Kim, Y.M.; Kim, Y.M.; Kim, E.C.; Gho, Y.S.; Kang, I.J.; Lee, S.Y.; Kong, Y.Y.; Kwon, Y.G. Vascular endothelial growth factor up-regulates expression of receptor activator of NF-kappa B (RANK) in endothelial cells. Concomitant increase of angiogenic responses to RANK ligand. J. Biol. Chem. 2003, 278, 39548–39557. [Google Scholar] [CrossRef] [Green Version]
  67. Kim, H.-H.; Shin, H.S.; Kwak, H.J.; Ahn, K.Y.; Kim, J.-H.; Lee, H.J.; Lee, M.-S.; Lee, Z.H.; Koh, G.Y. RANKL regulates endothelial cell survival through the phosphatidylinositol 3′-kinase/Akt signal transduction pathway. FASEB J. 2003, 17, 1–17. [Google Scholar] [CrossRef]
  68. Ulici, V.; Hoenselaar, K.D.; Agoston, H.; McErlain, D.D.; Umoh, J.; Chakrabarti, S.; Holdsworth, D.W.; Beier, F. The role of Akt1 in terminal stages of endochondral bone formation: Angiogenesis and ossification. Bone 2009, 45, 1133–1145. [Google Scholar] [CrossRef] [PubMed]
  69. Aragonés, J.; Fraisl, P.; Baes, M.; Carmeliet, P. Oxygen Sensors at the Crossroad of Metabolism. Cell Metab. 2009, 9, 11–22. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, Y.; Wan, C.; Deng, L.; Liu, X.; Cao, X.; Gilbert, S.R.; Bouxsein, M.L.; Faugere, M.-C.; Guldberg, R.E.; Gerstenfeld, L.C.; et al. The hypoxia-inducible factor α pathway couples angiogenesis to osteogenesis during skeletal development. J. Clin. Investig. 2007, 117, 1616–1626. [Google Scholar] [CrossRef]
  71. Riddle, R.C.; Khatri, R.; Schipani, E.; Clemens, T.L. Role of hypoxia-inducible factor-1α in angiogenic–osteogenic coupling. J. Mol. Med. 2009, 87, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ornitz, D.M. FGFs, heparan sulfate and FGFRs: Complex interactions essential for development. BioEssays 2000, 22, 108–112. [Google Scholar] [CrossRef]
  73. Collin-Osdoby, P.; Rothe, L.; Bekker, S.; Anderson, F.; Huang, Y.; Osdoby, P. Basic fibroblast growth factor stimulates osteoclast recruitment, development, and bone pit resorption in association with angiogenesis in vivo on the chick chorioallantoic membrane and activates isolated avian osteoclast resorption in vitro. J. Bone Miner. Res. 2002, 17, 1859–1871. [Google Scholar] [CrossRef]
  74. De Moerlooze, L.; Spencer-Dene, B.; Revest, J.M.; Hajihosseini, M.; Rosewell, I.; Dickson, C. An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Dev. 2000, 127, 483–492. [Google Scholar]
  75. Miraoui, H.; Marie, P.J. Fibroblast Growth Factor Receptor Signaling Crosstalk in Skeletogenesis. Sci. Signal. 2010, 3, re9. [Google Scholar] [CrossRef]
  76. Marie, P.J.; Miraoui, H.; Severe, N. FGF/FGFR signaling in bone formation: Progress and perspectives. Growth Factors 2012, 30, 117–123. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, J.; Park, J.-C.; Kim, S.-H.; Im, G.-I.; Kim, B.-S.; Lee, J.-B.; Choi, E.-Y.; Song, J.-S.; Cho, K.-S.; Kim, C.-S. Treatment of FGF-2 on stem cells from inflamed dental pulp tissue from human deciduous teeth. Oral Dis. 2014, 20, 191–204. [Google Scholar] [CrossRef]
  78. Hatch, N.E. FGF Signaling in Craniofacial Biological Control and Pathological Craniofacial Development. Crit. Rev. Eukaryot. Gene Expr. 2010, 20, 295–311. [Google Scholar] [CrossRef]
  79. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Turner, N.; Grose, R. Fibroblast growth factor signalling: From development to cancer. Nat. Rev. Cancer 2010, 10, 116–129. [Google Scholar] [CrossRef]
  81. Eswarakumar, V.; Lax, I.; Schlessinger, J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005, 16, 139–149. [Google Scholar] [CrossRef]
  82. Li, X.; Wang, C.; Xiao, J.; McKeehan, W.L.; Wang, F. Fibroblast growth factors, old kids on the new block. Semin. Cell Dev. Biol. 2016, 53, 155–167. [Google Scholar] [CrossRef] [Green Version]
  83. Potthoff, J.M.; Kliewer, S.A.; Mangelsdorf, D.J. Endocrine fibroblast growth factors 15/19 and 21: From feast to famine. Genes Dev. 2012, 26, 312–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tomlinson, E.; Fu, L.; John, L.; Hultgren, B.; Huang, X.; Renz, M.; Stephan, J.P.; Tsai, S.P.; Powell-Braxton, L.; French, D.; et al. Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology 2002, 143, 1741–1747. [Google Scholar] [CrossRef] [PubMed]
  85. Smith, E.R.; McMahon, L.P.; Holt, S.G. Fibroblast growth factor. Ann. Clin. Biochem. Int. J. Lab. Med. 2014, 51, 203–227. [Google Scholar] [CrossRef]
  86. Belov, A.A.; Mohammadi, M. Molecular Mechanisms of Fibroblast Growth Factor Signaling in Physiology and Pathology. Cold Spring Harb. Perspect. Biol. 2013, 5, a015958. [Google Scholar] [CrossRef]
  87. Ornitz, D.M.; Marie, P.J. Fibroblast growth factor signaling in skeletal development and disease. Genes Dev. 2015, 29, 1463–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Yayon, A.; Klagsbrun, M.; Esko, J.D.; Leder, P.; Ornitz, D.M. Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor. Cell 1991, 64, 841–848. [Google Scholar] [CrossRef]
  89. Rapraeger, C.A.; Krufka, A.; Olwin, B.B. Requirement of heparan sulfate for bFGF-mediated fibroblast growth and myoblast differentiation. Science 1991, 252, 1705–1708. [Google Scholar] [CrossRef] [Green Version]
  90. Ornitz, D.M.; Yayon, A.; Flanagan, J.G.; Svahn, C.M.; Levi, E.; Leder, P. Heparin is required for cell-free binding of basic fibroblast growth factor to a soluble receptor and for mitogenesis in whole cells. Mol. Cell. Biol. 1992, 12, 240–247. [Google Scholar] [CrossRef] [Green Version]
  91. Spivak-Kroizman, T.; Lemmon, M.; Dikic, I.; Ladbury, J.; Pinchasi, D.; Huang, J.; Jaye, M.; Crumley, G.; Schlessinger, J.; Lax, I. Heparin-induced oligomerization of FGF molecules is responsible for FGF receptor dimerization, activation, and cell proliferation. Cell 1994, 79, 1015–1024. [Google Scholar] [CrossRef]
  92. Beenken, A.; Mohammadi, M. The FGF family: Biology, pathophysiology and therapy. Nat. Rev. Drug Discov. 2009, 8, 235–253. [Google Scholar] [CrossRef] [Green Version]
  93. Matsuo, I.; Kimura-Yoshida, C. Extracellular modulation of Fibroblast Growth Factor signaling through heparan sulfate proteoglycans in mammalian development. Curr. Opin. Genet. Dev. 2013, 23, 399–407. [Google Scholar] [CrossRef] [PubMed]
  94. Xu, R.; Ori, A.; Rudd, T.R.; Uniewicz, K.A.; Ahmed, Y.A.; Guimond, S.E.; Skidmore, M.A.; Siligardi, G.; Yates, E.A.; Fernig, D.G. Diversification of the structural determinants of fibroblast growth factor-heparin interactions: Implications for binding specificity. J. Biol. Chem. 2012, 287, 40061–40073. [Google Scholar] [CrossRef] [Green Version]
  95. Goetz, R.; Mohammadi, M. Exploring mechanisms of FGF signalling through the lens of structural biology. Nat. Rev. Mol. Cell Biol. 2013, 14, 166–180. [Google Scholar] [CrossRef] [Green Version]
  96. Hart, K.C.; Robertson, S.C.; Kanemitsu, M.Y.; Meyer, A.N.; Tynan, J.A.; Donoghue, D.J. Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene 2000, 19, 3309–3320. [Google Scholar] [CrossRef] [Green Version]
  97. Shao, D.; Lazar, M.A. Modulating nuclear receptor function: May the phos be with you. J. Clin. Investig. 1999, 103, 1617–1618. [Google Scholar] [CrossRef] [Green Version]
  98. Yun, Y.-R.; Won, J.E.; Jeon, E.; Lee, S.; Kang, W.; Jo, H.; Jang, J.H.; Shin, U.S.; Kim, H.W. Fibroblast Growth Factors: Biology, Function, and Application for Tissue Regeneration. J. Tissue Eng. 2010, 2010, 218142. [Google Scholar] [CrossRef] [PubMed]
  99. Nugent, M.A.; Iozzo, R.V. Fibroblast growth factor-2. Int. J. Biochem. Cell Biol. 2000, 32, 115–120. [Google Scholar] [CrossRef]
  100. Johnson, L.G.; Lapadat, R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science 2002, 298, 1911–1912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Taketomi, T.; Onimura, T.; Yoshiga, D.; Muratsu, D.; Sanui, T.; Fukuda, T.; Kusukawa, J.; Nakamura, S. Sprouty2 is involved in the control of osteoblast proliferation and differentiation through the FGF and BMP signaling pathways. Cell Biol. Int. 2017, 42, 1106–1114. [Google Scholar] [CrossRef] [Green Version]
  102. Schmidt, A.; Ladage, D.; Schinköthe, T.; Klausmann, U.; Ulrichs, C.; Klinz, F.-J.; Brixius, K.; Arnhold, S.; Desai, B.; Mehlhorn, U.; et al. Basic Fibroblast Growth Factor Controls Migration in Human Mesenchymal Stem Cells. Stem Cells 2006, 24, 1750–1758. [Google Scholar] [CrossRef]
  103. Jin, L.; Hu, X.; Feng, L. NT3 inhibits FGF2-induced neural progenitor cell proliferation via the PI3K/GSK3 pathway. J. Neurochem. 2005, 93, 1251–1261. [Google Scholar] [CrossRef] [PubMed]
  104. Kanazawa, S.; Fujiwara, T.; Matsuzaki, S.; Shingaki, K.; Taniguchi, M.; Miyata, S.; Tohyama, M.; Sakai, Y.; Yano, K.; Hosokawa, K.; et al. bFGF Regulates PI3-Kinase-Rac1-JNK Pathway and Promotes Fibroblast Migration in Wound Healing. PLoS ONE 2010, 5, e12228. [Google Scholar] [CrossRef] [Green Version]
  105. Choi, S.C.; Kim, S.J.; Choi, J.H.; Park, C.Y.; Shim, W.J.; Lim, D.S. Fibroblast growth factor-2 and -4 promote the proliferation of bone marrow mesenchymal stem cells by the activation of the PI3K-Akt and ERK1/2 signaling pathways. Stem Cells Dev. 2008, 17, 725–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Yoon, H.; Kim, H.-L.; Chun, Y.-S.; Shin, D.H.; Lee, K.-H.; Shin, C.S.; Lee, D.Y.; Kim, H.-H.; Lee, Z.H.; Ryoo, H.-M.; et al. NAA10 controls osteoblast differentiation and bone formation as a feedback regulator of Runx2. Nat. Commun. 2014, 5, 5176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Lai, C.-F.; Chaudhary, L.; Fausto, A.; Halstead, L.R.; Ory, D.S.; Avioli, L.V.; Cheng, S.-L. Erk Is Essential for Growth, Differentiation, Integrin Expression, and Cell Function in Human Osteoblastic Cells. J. Biol. Chem. 2001, 276, 14443–14450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Marie, P. Fibroblast growth factor signaling controlling osteoblast differentiation. Gene 2003, 316, 23–32. [Google Scholar] [CrossRef]
  109. Chaudhary, L.R.; Avioli, L.V. Extracellular-signal regulated kinase signaling pathway mediates downregulation of type I procollagen gene expression by FGF-2, PDGF-BB, and okadaic acid in osteoblastic cells. J. Cell. Biochem. 2000, 76, 354–359. [Google Scholar] [CrossRef]
  110. Ge, C.; Xiao, G.; Jiang, D.; Franceschi, R.T. Critical role of the extracellular signal–regulated kinase–MAPK pathway in osteoblast differentiation and skeletal development. J. Cell Biol. 2007, 176, 709–718. [Google Scholar] [CrossRef]
  111. Marshak, D.R.; Jaiswal, R.K.; Jaiswal, N.; Bruder, S.P.; Mbalaviele, G.; Pittenger, M.F. Adult Human Mesenchymal Stem Cell Differentiation to the Osteogenic or Adipogenic Lineage Is Regulated by Mitogen-activated Protein Kinase. J. Biol. Chem. 2000, 275, 9645–9652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kratchmarova, I.; Blagoev, B.; Haack-Sorensen, M.; Kassem, M.; Mann, M. Mechanism of Divergent Growth Factor Effects in Mesenchymal Stem Cell Differentiation. Science 2005, 308, 1472–1477. [Google Scholar] [CrossRef] [Green Version]
  113. Matsushita, T.; Chan, Y.Y.; Kawanami, A.; Balmes, G.; Landreth, G.E.; Murakami, S. Extracellular Signal-Regulated Kinase 1 (ERK1) and ERK2 Play Essential Roles in Osteoblast Differentiation and in Supporting Osteoclastogenesis. Mol. Cell. Biol. 2009, 29, 5843–5857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Park, E.-S.; Lind, A.-K.; Dahm-Kähler, P.; Brännström, M.; Carletti, M.Z.; Christenson, L.K.; Curry, T.E.; Jo, M. RUNX2 Transcription Factor Regulates Gene Expression in Luteinizing Granulosa Cells of Rat Ovaries. Mol. Endocrinol. 2010, 24, 846–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Bundschu, K.; Knobeloch, K.-P.; Ullrich, M.; Schinke, T.; Amling, M.; Engelhardt, C.M.; Renné, T.; Walter, U.; Schuh, K. Gene Disruption of Spred-2 Causes Dwarfism. J. Biol. Chem. 2005, 280, 28572–28580. [Google Scholar] [CrossRef] [Green Version]
  116. Taniguchi, K.; Ayada, T.; Ichiyama, K.; Kohno, R.-I.; Yonemitsu, Y.; Minami, Y.; Kikuchi, A.; Maehara, Y.; Yoshimura, A. Sprouty2 and Sprouty4 are essential for embryonic morphogenesis and regulation of FGF signaling. Biochem. Biophys. Res. Commun. 2007, 352, 896–902. [Google Scholar] [CrossRef]
  117. Ozaki, K.-I.; Miyazaki, S.; Tanimura, S.; Kohno, M. Efficient suppression of FGF-2-induced ERK activation by the cooperative interaction among mammalian Sprouty isoforms. J. Cell Sci. 2005, 118, 5861–5871. [Google Scholar] [CrossRef] [Green Version]
  118. Yoshimura, A. Regulation of Cytokine Signaling by the SOCS and Spred Family Proteins. Keio J. Med. 2009, 58, 73–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Lu, H.C.; Swindell, E.C.; Sierralta, W.D.; Eichele, G.; Thaller, C. Evidence for a role of protein kinase C in FGF signal transduction in the developing chick limb bud. Development 2001, 128, 2451–2460. [Google Scholar]
  120. Mohammadi, M.; Schlessinger, J.; Hubbard, S.R. Structure of the FGF Receptor Tyrosine Kinase Domain Reveals a Novel Autoinhibitory Mechanism. Cell 1996, 86, 577–587. [Google Scholar] [CrossRef] [Green Version]
  121. Kim, H.-J.; Kim, J.-H.; Bae, S.-C.; Choi, J.-Y.; Ryoo, H.-M. The Protein Kinase C Pathway Plays a Central Role in the Fibroblast Growth Factor-stimulated Expression and Transactivation Activity of Runx2. J. Biol. Chem. 2003, 278, 319–326. [Google Scholar] [CrossRef] [Green Version]
  122. Osathanon, T.; Nowwarote, N.; Pavasant, P. Basic fibroblast growth factor inhibits mineralization but induces neuronal differentiation by human dental pulp stem cells through a FGFR and PLCγ signaling pathway. J. Cell. Biochem. 2011, 112, 1807–1816. [Google Scholar] [CrossRef] [PubMed]
  123. Spivak-Kroizman, T.; Mohammadi, M.; Hu, P.; Jaye, M.; Schlessinger, J.; Lax, I. Point mutation in the fibroblast growth factor receptor eliminates phosphatidylinositol hydrolysis without affecting neuronal differentiation of PC12 cells. J. Biol. Chem. 1994, 269, 14419–14423. [Google Scholar] [CrossRef]
  124. Kang, S.; Elf, S.; Dong, S.; Hitosugi, T.; Lythgoe, K.; Guo, A.; Ruan, H.; Lonial, S.; Khoury, H.J.; Williams, I.R.; et al. Fibroblast Growth Factor Receptor 3 Associates with and Tyrosine Phosphorylates p90 RSK2, Leading to RSK2 Activation That Mediates Hematopoietic Transformation. Mol. Cell. Biol. 2009, 29, 2105–2117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Dailey, L.; Ambrosetti, D.; Mansukhani, A.; Basilico, C. Mechanisms underlying differential responses to FGF signaling. Cytokine Growth Factor Rev. 2005, 16, 233–247. [Google Scholar] [CrossRef]
  126. Marie, P.; Coffin, J.; Hurley, M. FGF and FGFR signaling in chondrodysplasias and craniosynostosis. J. Cell. Biochem. 2005, 96, 888–896. [Google Scholar] [CrossRef] [PubMed]
  127. Muenke, M.; Schell, U. Fibroblast-growth-factor receptor mutations in human skeletal disorders. Trends Genet. 1995, 11, 308–313. [Google Scholar] [CrossRef]
  128. Ornitz, M.D.; Marie, P.J. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev. 2002, 16, 1446–1465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Ornitz, D.M.; Itoh, N. Fibroblast growth factors. Genome Biol. 2001, 2, 3005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Coffin, J.D.; Homer-Bouthiette, C.; Hurley, M.M. Fibroblast Growth Factor 2 and Its Receptors in Bone Biology and Disease. J. Endocr. Soc. 2018, 2, 657–671. [Google Scholar] [CrossRef] [PubMed]
  131. Du, E.; Xiao, L.; Hurley, M.M. FGF23 Neutralizing Antibody Ameliorates Hypophosphatemia and Impaired FGF Receptor Signaling in Kidneys of HMWFGF2 Transgenic Mice. J. Cell. Physiol. 2016, 232, 610–616. [Google Scholar] [CrossRef]
  132. Yang, J.; Meyer, M.; Müller, A.-K.; Böhm, F.; Grose, R.; Dauwalder, T.; Verrey, F.; Kopf, M.; Partanen, J.; Bloch, W.; et al. Fibroblast growth factor receptors 1 and 2 in keratinocytes control the epidermal barrier and cutaneous homeostasis. J. Cell Biol. 2010, 188, 935–952. [Google Scholar] [CrossRef] [Green Version]
  133. Sufen, G.; Xianghong, Y.; Yongxia, C.; Qian, P. bFGF and PDGF-BB have a synergistic effect on the proliferation, migration and VEGF release of endothelial progenitor cells. Cell Biol. Int. 2011, 35, 545–551. [Google Scholar] [CrossRef]
  134. Hill, P.A.; Tumber, A.; Meikle, M.C. Multiple Extracellular Signals Promote Osteoblast Survival and Apoptosis. Endocrinology 1997, 138, 3849–3858. [Google Scholar] [CrossRef]
  135. Nowwarote, N.; Sukarawan, W.; Pavasant, P.; Osathanon, T. Basic Fibroblast Growth Factor Regulates REX1 Expression Via IL-6 In Stem Cells Isolated From Human Exfoliated Deciduous Teeth. J. Cell. Biochem. 2017, 118, 1480–1488. [Google Scholar] [CrossRef]
  136. Wang, G.; Zhang, H.; Zhao, Y.; Li, J.; Cai, J.; Wang, P.; Meng, S.; Feng, J.; Miao, C.; Ding, M.; et al. Noggin and bFGF cooperate to maintain the pluripotency of human embryonic stem cells in the absence of feeder layers. Biochem. Biophys. Res. Commun. 2005, 330, 934–942. [Google Scholar] [CrossRef]
  137. Kawaguchi, H.; Kurokawa, T.; Hanada, K.; Hiyama, Y.; Tamura, M.; Ogata, E.; Matsumoto, T. Stimulation of fracture repair by recombinant human basic fibroblast growth factor in normal and streptozotocin-diabetic rats. Endocrinology 1994, 135, 774–781. [Google Scholar] [CrossRef]
  138. Liang, H.; Pun, S.; Wronski, T.J. Bone anabolic effects of basic fibroblast growth factor in ovariectomized rats. Endocrinology 1999, 140, 5780–5788. [Google Scholar] [CrossRef]
  139. Yamaguchi, T.P.; Rossant, J. Fibroblast growth factors in mammalian development. Curr. Opin. Genet. Dev. 1995, 5, 485–491. [Google Scholar] [CrossRef]
  140. Kimelman, D.; Kirschner, M. Synergistic induction of mesoderm by FGF and TGF-beta and the identification of an mRNA coding for FGF in the early Xenopus embryo. Cell 1987, 51, 869–877. [Google Scholar] [CrossRef]
  141. Peters, K.; Ornitz, D.; Werner, S.; Williams, L. Unique Expression Pattern of the FGF Receptor 3 Gene during Mouse Organogenesis. Dev. Biol. 1993, 155, 423–430. [Google Scholar] [CrossRef]
  142. Hurley, M.M.; Abreu, C.; Gronowicz, G.; Kawaguchi, H.; Lorenzo, J. Expression and regulation of basic fibroblast growth factor mRNA levels in mouse osteoblastic MC3T3-E1 cells. J. Biol. Chem. 1994, 269, 9392–9396. [Google Scholar] [CrossRef]
  143. Vagner, S.; Gensac, M.C.; Maret, A.; Bayard, F.; Amalric, F.; Prats, H.; Prats, A.C. Alternative translation of human fibroblast growth factor 2 mRNA occurs by internal entry of ribosomes. Mol. Cell. Biol. 1995, 15, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ding, V.M.; Ling, L.; Natarajan, S.; Yap, M.G.; Cool, S.M.; Choo, A.B. FGF-2 modulates Wnt signaling in undifferentiated hESC and iPS cells through activated PI3-K/GSK3beta signaling. J. Cell Physiol. 2010, 225, 417–428. [Google Scholar] [CrossRef] [PubMed]
  145. Fakhry, A.; Ratisoontorn, C.; Vedhachalam, C.; Salhab, I.; Koyama, E.; Leboy, P.; Pacifici, M.; Kirschner, R.E.; Nah, H.D. Effects of FGF-2/-9 in calvarial bone cell cultures: Differentiation stage-dependent mitogenic effect, inverse regulation of BMP-2 and noggin, and enhancement of osteogenic potential. Bone 2005, 36, 254–266. [Google Scholar] [CrossRef]
  146. Homer-Bouthiette, C.; Doetschman, T.; Xiao, L.; Hurley, M.M. Knockout of Nuclear High Molecular Weight FGF2 Isoforms in Mice Modulates Bone and Phosphate Homeostasis. J. Biol. Chem. 2014, 289, 36303–36314. [Google Scholar] [CrossRef] [Green Version]
  147. Yu, K.; Xu, J.; Liu, Z.; Sosic, D.; Shao, J.; Olson, E.N.; Towler, D.A.; Ornitz, D.M. Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development 2003, 130, 3063–3074. [Google Scholar] [CrossRef] [Green Version]
  148. Jacob, A.L.; Smith, C.; Partanen, J.; Ornitz, D.M. Fibroblast growth factor receptor 1 signaling in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev. Biol. 2006, 296, 315–328. [Google Scholar] [CrossRef] [Green Version]
  149. Lee, C.-H.; Su, S.-Y.; Sittampalam, K.; Chen, P.C.-H.; Petersson, F.; Kao, Y.-C.; Carpenter, T.O.; Hsieh, T.-H.; Konishi, E.; Tsai, J.-W.; et al. Frequent overexpression of klotho in fusion-negative phosphaturic mesenchymal tumors with tumorigenic implications. Mod. Pathol. 2019, 33, 858–870. [Google Scholar] [CrossRef]
  150. Fei, Y.; Xiao, L.; Doetschman, T.; Coffin, D.J.; Hurley, M.M. Fibroblast Growth Factor 2 Stimulation of Osteoblast Differentiation and Bone Formation Is Mediated by Modulation of the Wnt Signaling Pathway. J. Biol. Chem. 2011, 286, 40575–40583. [Google Scholar] [CrossRef] [Green Version]
  151. Naganawa, T.; Xiao, L.; Abogunde, E.; Sobue, T.; Kalajzic, I.; Sabbieti, M.; Agas, D.; Hurley, M. In vivo and in vitro comparison of the effects of FGF-2 null and haplo-insufficiency on bone formation in mice. Biochem. Biophys. Res. Commun. 2006, 339, 490–498. [Google Scholar] [CrossRef]
  152. Mansukhani, A.; Bellosta, P.; Sahni, M.; Basilico, C. Signaling by Fibroblast Growth Factors (Fgf) and Fibroblast Growth Factor Receptor 2 (Fgfr2)–Activating Mutations Blocks Mineralization and Induces Apoptosis in Osteoblasts. J. Cell Biol. 2000, 149, 1297–1308. [Google Scholar] [CrossRef]
  153. Mansukhani, A.; Ambrosetti, D.; Holmes, G.; Cornivelli, L.; Basilico, C. Sox2 induction by FGF and FGFR2 activating mutations inhibits Wnt signaling and osteoblast differentiation. J. Cell Biol. 2005, 168, 1065–1076. [Google Scholar] [CrossRef] [Green Version]
  154. Miraoui, H.; Oudina, K.; Petite, H.; Tanimoto, Y.; Moriyama, K.; Marie, P.J. Fibroblast growth factor receptor 2 promotes osteogenic differentiation in mesenchymal cells via ERK1/2 and protein kinase C signaling. J. Biol. Chem. 2009, 284, 4897–4904. [Google Scholar] [CrossRef] [Green Version]
  155. Naganawa, T.; Xiao, L.; Coffin, J.; Doetschman, T.; Sabbieti, M.G.; Agas, D.; Hurley, M.M. Reduced expression and function of bone morphogenetic protein-2 in bones of Fgf2 null mice. J. Cell. Biochem. 2008, 103, 1975–1988. [Google Scholar] [CrossRef]
  156. Xiao, G.; Jiang, D.; Gopalakrishnan, R.; Franceschi, R.T. Fibroblast Growth Factor 2 Induction of the Osteocalcin Gene Requires MAPK Activity and Phosphorylation of the Osteoblast Transcription Factor, Cbfa1/Runx2. J. Biol. Chem. 2002, 277, 36181–36187. [Google Scholar] [CrossRef] [Green Version]
  157. Ai-Aql, Z.S.; Alagl, A.S.; Graves, D.T.; Gerstenfeld, L.C.; Einhorn, T.A. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J. Dent Res. 2008, 87, 107–118. [Google Scholar] [CrossRef] [PubMed]
  158. Lemonnier, J.; Haÿ, E.; Delannoy, P.; Lomri, A.; Modrowski, D.; Caverzasio, J.; Marie, P.J. Role of N-Cadherin and Protein Kinase C in Osteoblast Gene Activation Induced by the S252W Fibroblast Growth Factor Receptor 2 Mutation in Apert Craniosynostosis. J. Bone Miner. Res. 2001, 16, 832–845. [Google Scholar] [CrossRef] [PubMed]
  159. Debiais, F.; Lefèvre, G.; Lemonnier, J.; Le Mée, S.; Lasmoles, F.; Mascarelli, F.; Marie, P. Fibroblast growth factor-2 induces osteoblast survival through a phosphatidylinositol 3-kinase-dependent, -β-catenin-independent signaling pathway. Exp. Cell Res. 2004, 297, 235–246. [Google Scholar] [CrossRef]
  160. Montero, A.; Okada, Y.; Tomita, M.; Ito, M.; Tsurukami, H.; Nakamura, T.; Doetschman, T.; Coffin, J.D.; Hurley, M.M. Disruption of the fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J. Clin. Investig. 2000, 105, 1085–1093. [Google Scholar] [CrossRef] [Green Version]
  161. Xiao, L.; Sobue, T.; Esliger, A.; Kronenberg, M.S.; Coffin, J.D.; Doetschman, T.; Hurley, M.M. Disruption of the Fgf2 gene activates the adipogenic and suppresses the osteogenic program in mesenchymal marrow stromal stem cells. Bone 2010, 47, 360–370. [Google Scholar] [CrossRef] [Green Version]
  162. Coffin, J.D.; Florkiewicz, R.Z.; Neumann, J.; Mort-Hopkins, T.; Dorn, G.W.; Lightfoot, P.; German, R.; Howles, P.N.; Kier, A.; O’Toole, B.A. Abnormal bone growth and selective translational regulation in basic fibroblast growth factor (FGF-2) transgenic mice. Mol. Biol. Cell 1995, 6, 1861–1873. [Google Scholar] [CrossRef]
  163. Sobue, T.; Naganawa, T.; Xiao, L.; Okada, Y.; Tanaka, Y.; Ito, M.; Okimoto, N.; Nakamura, T.; Coffin, J.; Hurley, M. Over-expression of fibroblast growth factor-2 causes defective bone mineralization and osteopenia in transgenic mice. J. Cell. Biochem. 2005, 95, 83–94. [Google Scholar] [CrossRef]
  164. Berthiaume, F.; Maguire, T.J.; Yarmush, M.L. Tissue Engineering and Regenerative Medicine: History, Progress, and Challenges. Annu. Rev. Chem. Biomol. Eng. 2011, 2, 403–430. [Google Scholar] [CrossRef]
  165. Langer, R.; Vacanti, J.P. Tissue engineering. Science 1993, 260, 920–926. [Google Scholar] [CrossRef] [Green Version]
  166. Black, C.R.M.; Goriainov, V.; Gibbs, D.; Kanczler, J.M.; Tare, R.S.; Oreffo, R.O.C. Bone Tissue Engineering. Curr. Mol. Biol. Rep. 2015, 1, 132–140. [Google Scholar] [CrossRef]
  167. Porter, J.R.; Ruckh, T.T.; Popat, K.C. Bone tissue engineering: A review in bone biomimetics and drug delivery strategies. Biotechnol. Prog. 2009, 25, 1539–1560. [Google Scholar] [CrossRef]
  168. McCarthy, T.L.; Centrella, M.; Canalis, E. Effects of Fibroblast Growth Factors on Deoxyribonucleic Acid and Collagen Synthesis in Rat Parietal Bone Cells. Endocrinology 1989, 125, 2118–2126. [Google Scholar] [CrossRef]
  169. Martin, I.; Muraglia, A.; Campanile, G.; Cancedda, R.; Quarto, R. Fibroblast growth factor-2 supports ex vivo expansion and maintenance of osteogenic precursors from human bone marrow. Endocrinology 1997, 138, 4456–4462. [Google Scholar] [CrossRef]
  170. Gorin, C.; Rochefort, G.Y.; Bascetin, R.; Ying, H.; Lesieur, J.; Sadoine, J.; Beckouche, N.; Berndt, S.; Novais, A.; Lesage, M.; et al. Priming Dental Pulp Stem Cells With Fibroblast Growth Factor-2 Increases Angiogenesis of Implanted Tissue-Engineered Constructs Through Hepatocyte Growth Factor and Vascular Endothelial Growth Factor Secretion. Stem Cells Transl. Med. 2016, 5, 392–404. [Google Scholar] [CrossRef] [PubMed]
  171. Hasegawa, T.; Chosa, N.; Asakawa, T.; Yoshimura, Y.; Asakawa, A.; Ishisaki, A.; Tanaka, M. Effect of fibroblast growth factor-2 on periodontal ligament cells derived from human deciduous teeth in vitro. Exp. Ther. Med. 2010, 1, 337–341. [Google Scholar] [CrossRef] [PubMed]
  172. Wu, J.; Huang, G.T.-J.; He, W.; Wang, P.; Tong, Z.; Jia, Q.; Dong, L.; Niu, Z.; Ni, L. Basic Fibroblast Growth Factor Enhances Stemness of Human Stem Cells from the Apical Papilla. J. Endod. 2012, 38, 614–622. [Google Scholar] [CrossRef] [Green Version]
  173. Novais, A.; Lesieur, J.; Sadoine, J.; Slimani, L.; Baroukh, B.; Saubaméa, B.; Schmitt, A.; Vital, S.; Poliard, A.; Hélary, C.; et al. Priming Dental Pulp Stem Cells from Human Exfoliated Deciduous Teeth with Fibroblast Growth Factor-2 Enhances Mineralization Within Tissue-Engineered Constructs Implanted in Craniofacial Bone Defects. Stem Cells Transl. Med. 2019, 8, 844–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Debiais, F.; Hott, M.; Graulet, A.M.; Marie, P.J. The Effects of Fibroblast Growth Factor-2 on Human Neonatal Calvaria Osteoblastic Cells Are Differentiation Stage Specific. J. Bone Miner. Res. 1998, 13, 645–654. [Google Scholar] [CrossRef] [PubMed]
  175. Hurley, M.M.; Adams, U.J.; Wang, L.; Jiang, X.; Burt, P.M.; Du, E.; Xiao, L. Accelerated fracture healing in transgenic mice overexpressing an anabolic isoform of fibroblast growth factor 2. J. Cell. Biochem. 2016, 117, 599–611. [Google Scholar] [CrossRef] [PubMed]
  176. Cowan, C.M.; Quarto, N.; Warren, S.M.; Salim, A.; Longaker, M.T. Age-related changes in the biomolecular mechanisms of calvarial osteoblast biology affect fibroblast growth factor-2 signaling and osteogenesis. J. Biol. Chem. 2003, 278, 45040. [Google Scholar] [CrossRef]
  177. Shimabukuro, Y.; Ueda, M.; Ozasa, M.; Anzai, J.; Takedachi, M.; Yanagita, M.; Ito, M.; Hashikawa, T.; Yamada, S.; Murakami, S. Fibroblast Growth Factor–2 Regulates the Cell Function of Human Dental Pulp Cells. J. Endod. 2009, 35, 1529–1535. [Google Scholar] [CrossRef] [PubMed]
  178. He, H.; Yu, J.; Liu, Y.; Lu, S.; Liu, H.; Shi, J.; Jin, Y. Effects of FGF2 and TGFbeta1 on the differentiation of human dental pulp stem cells in vitro. Cell Biol. Int. 2008, 32, 827–834. [Google Scholar] [CrossRef] [PubMed]
  179. Nakao, K.; Itoh, M.; Tomita, Y.; Tomooka, Y.; Tsuji, T. FGF-2 potently induces both proliferation and DSP expression in collagen type I gel cultures of adult incisor immature pulp cells. Biochem. Biophys. Res. Commun. 2004, 325, 1052–1059. [Google Scholar] [CrossRef] [PubMed]
  180. Hakki, S.S.; Hakki, E.E.; Nohutcu, R.M. Regulation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases by basic fibroblast growth factor and dexamethasone in periodontal ligament cells. J. Periodontal Res. 2009, 44, 794–802. [Google Scholar] [CrossRef]
  181. Shimabukuro, Y.; Ueda, M.; Ichikawa, T.; Terashi, Y.; Yamada, S.; Kusumoto, Y.; Takedachi, M.; Terakura, M.; Kohya, A.; Hashikawa, T.; et al. Fibroblast Growth Factor-2 Stimulates Hyaluronan Production by Human Dental Pulp Cells. J. Endod. 2005, 31, 805–808. [Google Scholar] [CrossRef]
  182. Nauman, E.A.; Sakata, T.; Keaveny, T.M.; Halloran, B.P.; Bikle, D.D. bFGF Administration Lowers the Phosphate Threshold for Mineralization in Bone Marrow Stromal Cells. Calcif. Tissue Int. 2003, 73, 147–152. [Google Scholar] [CrossRef]
  183. Nakamura, S.; Nambu, M.; Ishizuka, T.; Hattori, H.; Kanatani, Y.; Takase, B.; Kishimoto, S.; Amano, Y.; Aoki, H.; Kiyosawa, T.; et al. Effect of controlled release of fibroblast growth factor-2 from chitosan/fucoidan micro complex-hydrogel onin vitro andin vivo vascularization. J. Biomed. Mater. Res. Part. A 2007, 85, 619–627. [Google Scholar] [CrossRef]
  184. Tabata, Y.; Yamada, K.; Hong, L.; Miyamoto, S.; Hashimoto, N.; Ikada, Y. Skull bone regeneration in primates in response to basic fibroblast growth factor. J. Neurosurg. 1999, 91, 851–856. [Google Scholar] [CrossRef]
  185. Kwan, M.D.; Sellmyer, M.A.; Quarto, N.; Ho, A.M.; Wandless, T.J.; Longaker, M.T. Chemical Control of FGF-2 Release for Promoting Calvarial Healing with Adipose Stem Cells. J. Biol. Chem. 2011, 286, 11307–11313. [Google Scholar] [CrossRef] [Green Version]
  186. Nakahara, T.; Nakamura, T.; Kobayashi, E.; Inoue, M.; Shigeno, K.; Tabata, Y.; Eto, K.; Shimizu, Y. Novel Approach to Regeneration of Periodontal Tissues Based on in Situ Tissue Engineering: Effects of Controlled Release of Basic Fibroblast Growth Factor from a Sandwich Membrane. Tissue Eng. 2003, 9, 153–162. [Google Scholar] [CrossRef] [PubMed]
  187. Anzai, J.; Nagayasu-Tanaka, T.; Terashima, A.; Asano, T.; Yamada, S.; Nozaki, T.; Kitamura, M.; Murakami, S. Long-term Observation of Regenerated Periodontium Induced by FGF-2 in the Beagle Dog 2-Wall Periodontal Defect Model. PLoS ONE 2016, 11, e0158485. [Google Scholar] [CrossRef]
  188. Lee, J.-H.; Kang, K.-J.; Jang, Y.-J. Functional efficacy of human recombinant FGF-2s tagged with (His) 6 and (His-Asn) 6 at the N- and C-termini in human gingival fibroblast and periodontal ligament-derived cells. Protein Expr. Purif. 2017, 135, 37–44. [Google Scholar] [CrossRef] [PubMed]
  189. Charles, L.F.; Woodman, J.L.; Ueno, D.; Gronowicz, G.; Hurley, M.M.; Kuhn, L.T. Effects of low dose FGF-2 and BMP-2 on healing of calvarial defects in old mice. Exp. Gerontol. 2015, 64, 62–69. [Google Scholar] [CrossRef] [Green Version]
  190. Kigami, R.; Sato, S.; Tsuchiya, N.; Yoshimakai, T.; Arai, Y.; Ito, K. FGF-2 Angiogenesis in Bone Regeneration Within Critical-Sized Bone Defects in Rat Calvaria. Implant. Dent. 2013, 22, 422–427. [Google Scholar] [CrossRef] [PubMed]
  191. Song, K.; Rao, N.-J.; Chen, M.-L.; Huang, Z.-J.; Cao, Y.-G. Enhanced bone regeneration with sequential delivery of basic fibroblast growth factor and sonic hedgehog. Injury 2011, 42, 796–802. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, B.; Mastrogiacomo, S.; Yang, F.; Shao, J.; Ong, M.M.A.; Chanchareonsook, N.; Jansen, J.A.; Walboomers, X.F.; Yu, N. Application of BMP-Bone Cement and FGF-Gel on Periodontal Tissue Regeneration in Nonhuman Primates. Tissue Eng. Part C Methods 2019, 25, 748–756. [Google Scholar] [CrossRef] [PubMed]
  193. Collignon, A.-M.; Lesieur, J.; Anizan, N.; Ben Azzouna, R.; Poliard, A.; Gorin, C.; Letourneur, D.; Chaussain, C.; Rouzet, F.; Rochefort, G.Y. Early angiogenesis detected by PET imaging with 64Cu-NODAGA-RGD is predictive of bone critical defect repair. Acta Biomater. 2018, 82, 111–121. [Google Scholar] [CrossRef]
  194. Grosso, A.; Burger, M.G.; Lunger, A.; Schaefer, D.J.; Banfi, A.; Di Maggio, N. It Takes Two to Tango: Coupling of Angiogenesis and Osteogenesis for Bone Regeneration. Front. Bioeng. Biotechnol. 2017, 5, 68. [Google Scholar] [CrossRef]
  195. Atlas, Y.; Gorin, C.; Novais, A.; Marchand, M.F.; Chatzopoulou, E.; Lesieur, J.; Bascetin, R.; Binet-Moussy, C.; Sadoine, J.; Lesage, M.; et al. Microvascular maturation by mesenchymal stem cells in vitro improves blood perfusion in implanted tissue constructs. Biomaterials 2021, 268, 120594. [Google Scholar] [CrossRef]
  196. Kitamura, M.; Akamatsu, M.; Machigashira, M.; Hara, Y.; Sakagami, R.; Hirofuji, T.; Hamachi, T.; Maeda, K.; Yokota, M.; Kido, J.; et al. FGF-2 stimulates periodontal regeneration: Results of a multi-center randomized clinical trial. J. Dent. Res. 2011, 90, 35–40. [Google Scholar] [CrossRef]
  197. Santana, D.B.R.; de Santana, C.M. Human intrabony defect regeneration with rhFGF-2 and hyaluronic acid—A randomized controlled clinical trial. J. Clin. Periodontol. 2015, 42, 658–665. [Google Scholar] [CrossRef] [PubMed]
  198. Cochran, D.; Oh, T.-J.; Mills, M.; Clem, D.; McClain, P.; Schallhorn, R.; McGuire, M.; Scheyer, E.; Giannobile, W.; Reddy, M.; et al. A Randomized Clinical Trial Evaluating rh-FGF-2/β-TCP in Periodontal Defects. J. Dent. Res. 2016, 95, 523–530. [Google Scholar] [CrossRef] [PubMed]
  199. Khoshkam, V.; Chan, H.L.; Lin, G.H.; Mailoa, J.; Giannobile, W.V.; Wang, H.L.; Oh, T.J. Outcomes of regenerative treatment with rhPDGF-BB and rhFGF-2 for periodontal intra-bony defects: A systematic review and meta-analysis. J. Clin. Periodontol. 2015, 42, 272–280. [Google Scholar] [CrossRef] [PubMed]
  200. Li, F.; Yu, F.; Xu, X.; Li, C.; Huang, D.; Zhou, X.; Ye, L.; Zheng, L. Evaluation of Recombinant Human FGF-2 and PDGF-BB in Periodontal Regeneration: A Systematic Review and Meta-Analysis. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  201. Gronowicz, G.; Jacobs, E.; Peng, T.; Zhu, L.; Hurley, M.; Kuhn, L.T. Calvarial Bone Regeneration Is Enhanced by Sequential Delivery of FGF-2 and BMP-2 from Layer-by-Layer Coatings with a Biomimetic Calcium Phosphate Barrier Layer. Tissue Eng. Part. A 2017, 23, 1490–1501. [Google Scholar] [CrossRef]
  202. Ou, G.; Charles, L.; Matton, S.; Rodner, C.; Hurley, M.; Kuhn, L.; Gronowicz, G. Fibroblast Growth Factor-2 Stimulates the Proliferation of Mesenchyme-Derived Progenitor Cells From Aging Mouse and Human Bone. J. Gerontol. Ser. A Boil. Sci. Med. Sci. 2010, 65, 1051–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Sukarawan, W.; Nowwarote, N.; Kerdpon, P.; Pavasant, P.; Osathanon, T. Effect of basic fibroblast growth factor on pluripotent marker expression and colony forming unit capacity of stem cells isolated from human exfoliated deciduous teeth. Odontology 2013, 102, 160–166. [Google Scholar] [CrossRef]
  204. Ludwig, T.E.; Levenstein, M.E.; Jones, J.M.; Berggren, W.T.; Mitchen, E.R.; Frane, J.L.; Crandall, L.J.; Daigh, C.A.; Conard, K.R.; Piekarczyk, M.S.; et al. Derivation of human embryonic stem cells in defined conditions. Nat. Biotechnol. 2006, 24, 185–187. [Google Scholar] [CrossRef] [PubMed]
  205. Varkey, M.; Kucharski, C.; Haque, T.; Sebald, W.; Uludağ, H. In Vitro Osteogenic Response of Rat Bone Marrow Cells to bFGF and BMP-2 Treatments. Clin. Orthop. Relat. Res. 2006, 443, 113–123. [Google Scholar] [CrossRef] [Green Version]
  206. Dunstan, C.R.; Boyce, R.; Boyce, B.F.; Garrett, I.R.; Izbicka, E.; Burgess, W.H.; Mundy, G.R. Systemic Administration of Acidic Fibroblast Growth Factor (FGF-1) Prevents Bone Loss and Increases New Bone Formation in Ovariectomized Rats. J. Bone Miner. Res. 1999, 14, 953–959. [Google Scholar] [CrossRef] [PubMed]
  207. Kato, T.; Kawaguchi, H.; Hanada, K.; Aoyama, I.; Hiyama, Y.; Nakamura, T.; Kuzutani, K.; Tamura, M.; Kurokawa, T.; Nakamura, K. Single local injection of recombinant fibroblast growth factor-2 stimulates healing of segmental bone defects in rabbits. J. Orthop. Res. 1998, 16, 654–659. [Google Scholar] [CrossRef] [PubMed]
  208. Okazaki, H.; Kurokawa, T.; Nakamura, K.; Matsushita, T.; Mamada, K.; Kawaguchi, H. Stimulation of bone formation by recombinant fibroblast growth factor-2 in callotasis bone lengthening of rabbits. Calcif. Tissue Int. 1999, 64, 542–546. [Google Scholar] [CrossRef]
  209. Nakamura, T.; Hara, Y.; Tagawa, M.; Tamura, M.; Yuge, T.; Fukuda, H.; Nigi, H. Recombinant Human Basic Fibroblast Growth Factor Accelerates Fracture Healing by Enhancing Callus Remodeling in Experimental Dog Tibial Fracture. J. Bone Miner. Res. 1998, 13, 942–949. [Google Scholar] [CrossRef]
  210. Kawaguchi, H.; Nakamura, K.; Tabata, Y.; Ikada, Y.; Aoyama, I.; Anzai, J.; Nakamura, T.; Hiyama, Y.; Tamura, M. Acceleration of fracture healing in nonhuman primates by fibroblast growth factor-2. J. Clin. Endocrinol. Metab. 2001, 86, 875–880. [Google Scholar] [CrossRef]
  211. Radomsky, M.L.; Aufdemorte, T.B.; Swain, L.D.; Fox, W.C.; Spiro, R.C.; Poser, J.W. Novel formulation of fibroblast growth factor-2 in a hyaluronan gel accelerates fracture healing in nonhuman primates. J. Orthop. Res. 1999, 17, 607–614. [Google Scholar] [CrossRef] [PubMed]
  212. Kamo, K.; Miyakoshi, N.; Kasukawa, Y.; Sasaki, H.; Shimada, Y. Effects of single and cyclical local injections of basic fibroblast growth factor on cancellous bone defects in rabbits. J. Orthop. Sci. 2009, 14, 811–819. [Google Scholar] [CrossRef] [PubMed]
  213. Nakamura, T.; Hanada, K.; Tamura, M.; Shibanushi, T.; Nigi, H.; Tagawa, M.; Fukumoto, S.; Matsumoto, T. Stimulation of endosteal bone formation by systemic injections of recombinant basic fibroblast growth factor in rats. Endocrinology 1995, 136, 1276–1284. [Google Scholar] [CrossRef]
  214. Nagai, H.; Tsukuda, R.; Mayahara, H. Effects of basic fibroblast growth factor (bFGF) on bone formation in growing rats. Bone 1995, 16, 367–373. [Google Scholar] [CrossRef]
  215. Mayahara, H.; Ito, T.; Nagai, H.; Miyajima, H.; Tsukuda, R.; Taketomi, S.; Mizoguchi, J.; Kato, K. In VivoStimulation of Endosteal Bone Formation by Basic Fibroblast Growth Factor in Rats. Growth Factors 1993, 9, 73–80. [Google Scholar] [CrossRef] [PubMed]
  216. Lane, N.E.; Yao, W.; Kumer, J.; Breuning, T.; Wronski, T.; Modin, G.; Kinney, J.H. Basic fibroblast growth factor partially restores trabecular bone architecture in osteopenic ovariectomized rats. Osteoporos. Int. 2003, 4, 374–382. [Google Scholar]
  217. Abbaspour, A.; Takata, S.; Sairyo, K.; Katoh, S.; Yukata, K.; Yasui, N. Continuous local infusion of fibroblast growth factor-2 enhances consolidation of the bone segment lengthened by distraction osteogenesis in rabbit experiment. Bone 2008, 42, 98–106. [Google Scholar] [CrossRef]
  218. Benington, L.; Rajan, G.; Locher, C.; Lim, L.Y. Fibroblast Growth Factor 2—A Review of Stabilisation Approaches for Clinical Applications. Pharmaceutics 2020, 12, 508. [Google Scholar] [CrossRef]
  219. Kuroda, Y.; Kawai, T.; Goto, K.; Matsuda, S. Clinical application of injectable growth factor for bone regeneration: A systematic review. Inflamm. Regen. 2019, 39, 20. [Google Scholar] [CrossRef]
  220. Arakawa, T.; Prestrelski, S.J.; Kenney, W.C.; Carpenter, J.F. Factors affecting short-term and long-term stabilities of proteins. Adv. Drug Deliv. Rev. 2001, 46, 307–326. [Google Scholar] [CrossRef]
  221. Tabata, Y.; Hijikata, S.; Ikada, Y. Enhanced vascularization and tissue granulation by basic fibroblast growth factor impregnated in gelatin hydrogels. J. Control. Release 1994, 31, 189–199. [Google Scholar] [CrossRef]
  222. Kawaguchi, H.; Jingushi, S.; Izumi, T.; Fukunaga, M.; Matsushita, T.; Nakamura, T.; Mizuno, K.; Nakamura, T.; Nakamura, K. Local application of recombinant human fibroblast growth factor-2 on bone repair: A dose–escalation prospective trial on patients with osteotomy. J. Orthop. Res. 2007, 25, 480–487. [Google Scholar] [CrossRef] [PubMed]
  223. Kawaguchi, H.; Oka, H.; Jingushi, S.; Izumi, T.; Fukunaga, M.; Sato, K.; Matsushita, T.; Nakamura, K. For the TESK Group A local application of recombinant human fibroblast growth factor 2 for tibial shaft fractures: A randomized, placebo-controlled trial. J. Bone Miner. Res. 2010, 25, 2735–2743. [Google Scholar] [CrossRef]
  224. Nakamura, S.; Kanatani, Y.; Kishimoto, S.; Nakamura, S.I.; Ohno, C.; Horio, T.; Masanori, F.; Hattori, H.; Tanaka, Y.; Kiyosawa, T.; et al. Controlled release of FGF-2 using fragmin/protamine microparticles and effect on neovascularization. J. Biomed. Mater. Res. A 2009, 91, 814–823. [Google Scholar] [CrossRef]
  225. Mammadov, R.; Mammadov, B.; Guler, M.O.; Tekinay, A.B. Growth Factor Binding on Heparin Mimetic Peptide Nanofibers. Biomacromolecules 2012, 13, 3311–3319. [Google Scholar] [CrossRef] [PubMed]
  226. Kim, S.H.; Kiick, K.L. Heparin-mimetic sulfated peptides with modulated affinities for heparin-binding peptides and growth factors. Peptides 2007, 28, 2125–2136. [Google Scholar] [CrossRef] [Green Version]
  227. Tardieu, M.; Gamby, C.; Avramoglou, T.; Jozefonvicz, J.; Barritault, D. Derivatized dextrans mimic heparin as stabilizers, potentiators, and protectors of acidic or basic FGF. J. Cell. Physiol. 1992, 150, 194–203. [Google Scholar] [CrossRef]
  228. Liekens, S.; Leali, D.; Neyts, J.; Esnouf, R.; Rusnati, M.; Dell’Era, P.; Maudgal, P.C.; De Clercq, E.; Presta, M. Modulation of Fibroblast Growth Factor-2 Receptor Binding, Signaling, and Mitogenic Activity by Heparin-Mimicking Polysulfonated Compounds. Mol. Pharmacol. 1999, 56, 204–213. [Google Scholar] [CrossRef] [Green Version]
  229. Nguyen, T.H.; Paluck, S.J.; McGahran, A.J.; Maynard, H.D. Poly(vinyl sulfonate) Facilitates bFGF-Induced Cell Proliferation. Biomacromolecules 2015, 16, 2684–2692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Arunkumar, P.; Dougherty, J.A.; Weist, J.; Kumar, N.; Angelos, M.G.; Powell, H.M.; Khan, M. Sustained Release of Basic Fibroblast Growth Factor (bFGF) Encapsulated Polycaprolactone (PCL) Microspheres Promote Angiogenesis In Vivo. Nanomaterials 2019, 9, 1037. [Google Scholar] [CrossRef] [Green Version]
  231. Ali, Z.; Islam, A.; Sherrell, P.; Le-Moine, M.; Lolas, G.; Syrigos, K.; Rafat, M.; Jensen, L.D. Adjustable delivery of pro-angiogenic FGF-2 by alginate:collagen microspheres. Biol. Open 2018, 7, bio027060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Murahashi, Y.; Yano, F.; Nakamoto, H.; Maenohara, Y.; Iba, K.; Yamashita, T.; Tanaka, S.; Ishihara, K.; Okamura, Y.; Moro, T.; et al. Multi-layered PLLA-nanosheets loaded with FGF-2 induce robust bone regeneration with controlled release in critical-sized mouse femoral defects. Acta Biomater. 2019, 85, 172–179. [Google Scholar] [CrossRef]
  233. Gromolak, S.; Krawczenko, A.; Antończyk, A.; Buczak, K.; Kiełbowicz, Z.; Klimczak, A. Biological Characteristics and Osteogenic Differentiation of Ovine Bone Marrow Derived Mesenchymal Stem Cells Stimulated with FGF-2 and BMP-2. Int. J. Mol. Sci. 2020, 21, 9726. [Google Scholar] [CrossRef]
  234. Akita, S.; Fukui, M.; Nakagawa, H.; Fujii, T.; Akino, K. Cranial bone defect healing is accelerated by mesenchymal stem cells induced by coadministration of bone morphogenetic protein-2 and basic fibroblast growth factor. Wound Repair Regen. 2004, 12, 252–259. [Google Scholar] [CrossRef]
  235. Hanada, K.; Dennis, J.E.; Caplan, A.I. Stimulatory Effects of Basic Fibroblast Growth Factor and Bone Morphogenetic Protein-2 on Osteogenic Differentiation of Rat Bone Marrow-Derived Mesenchymal Stem Cells. J. Bone Miner. Res. 1997, 12, 1606–1614. [Google Scholar] [CrossRef]
  236. Fujimura, K.; Bessho, K.; Okubo, Y.; Kusumoto, K.; Segami, N.; Iizuka, T. The effect of fibroblast growth factor-2 on the osteoinductive activity of recombinant human bone morphogenetic protein-2 in rat muscle. Arch. Oral Biol. 2002, 47, 577–584. [Google Scholar] [CrossRef]
  237. Takita, H.; Tsuruga, E.; Ono, I.; Kuboki, Y. Enhancement by bFGF of osteogenesis induced by rhBMP-2 in rats. Eur. J. Oral Sci. 1997, 105, 588–592. [Google Scholar] [CrossRef]
  238. Nakamura, Y.; Tensho, K.; Nakaya, H.; Nawata, M.; Okabe, T.; Wakitani, S. Low dose fibroblast growth factor-2 (FGF-2) enhances bone morphogenetic protein-2 (BMP-2)-induced ectopic bone formation in mice. Bone 2005, 36, 399–407. [Google Scholar] [CrossRef] [PubMed]
  239. Berendsen, A.D.; Olsen, B.R. Bone development. Bone 2015, 80, 14–18. [Google Scholar] [CrossRef] [Green Version]
  240. Breeland, G.; Menezes, R.G. Embryology, Bone Ossification; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2019. [Google Scholar]
  241. Deckers, M.M.; Van Bezooijen, R.L.; Van Der Horst, G.; Hoogendam, J.; van Der Bent, C.; Papapoulos, S.E.; Löwik, C.W. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. Endocrinology 2002, 143, 1545–1553. [Google Scholar] [CrossRef] [PubMed]
  242. Wan, M.; Cao, X. BMP signaling in skeletal development. Biochem. Biophys. Res. Commun. 2005, 328, 651–657. [Google Scholar] [CrossRef] [PubMed]
  243. Peng, H.; Usas, A.; Olshanski, A.; Ho, A.M.; Gearhart, B.; Cooper, G.M.; Huard, J. VEGF Improves, Whereas sFlt1 Inhibits, BMP2-Induced Bone Formation and Bone Healing Through Modulation of Angiogenesis. J. Bone Miner. Res. 2005, 20, 2017–2027. [Google Scholar] [CrossRef]
  244. Valdimarsdottir, G.; Goumans, M.-J.; Rosendahl, A.; Brugman, M.; Itoh, S.; Lebrin, F.; Sideras, P.; Dijke, P.T. Stimulation of Id1 Expression by Bone Morphogenetic Protein Is Sufficient and Necessary for Bone Morphogenetic Protein–Induced Activation of Endothelial Cells. Circulation 2002, 106, 2263–2270. [Google Scholar] [CrossRef] [PubMed]
  245. Salazar, V.S.; Gamer, L.W.; Rosen, V. BMP signalling in skeletal development, disease and repair. Nat. Rev. Endocrinol. 2016, 12, 203–221. [Google Scholar] [CrossRef]
  246. Carano, R.A.; Filvaroff, E.H. Angiogenesis and bone repair. Drug Discov. Today 2003, 8, 980–989. [Google Scholar] [CrossRef]
  247. Chen, G.; Deng, C.; Li, Y.P. TGF-beta and BMP signaling in osteoblast differentiation and bone formation. Int. J. Biol. Sci. 2012, 8, 272–288. [Google Scholar] [CrossRef] [Green Version]
  248. Yang, E.Y.; Moses, H.L. Transforming growth factor beta 1-induced changes in cell migration, proliferation, and angiogenesis in the chicken chorioallantoic membrane. J. Cell Biol. 1990, 111, 731–741. [Google Scholar] [CrossRef] [PubMed]
  249. Sabbieti, M.G.; Marchetti, L.; Gabrielli, M.G.; Menghi, M.; Materazzi, S.; Menghi, G.; Raisz, L.G.; Hurley, M.M. Prostaglandins differently regulate FGF-2 and FGF receptor expression and induce nuclear translocation in osteoblasts via MAPK kinase. Cell Tissue Res. 2004, 319, 267–278. [Google Scholar] [CrossRef]
  250. Sobue, T.; Gravely, T.; Hand, A.; Min, Y.K.; Pilbeam, C.; Raisz, L.G.; Zhang, X.; Larocca, D.; Florkiewicz, R.; Hurley, M.M. Regulation of fibroblast growth factor 2 and fibroblast growth factor receptors by transforming growth factor beta in human osteoblastic MG-63 cells. J. Bone Miner. Res. 2002, 17, 502–512. [Google Scholar] [CrossRef]
  251. Fiedler, J.; Röderer, G.; Günther, K.-P.; Brenner, R.E. BMP-2, BMP-4, and PDGF-bb stimulate chemotactic migration of primary human mesenchymal progenitor cells. J. Cell. Biochem. 2002, 87, 305–312. [Google Scholar] [CrossRef]
  252. Edwards, J.R.; Nyman, J.S.; Lwin, S.T.; Moore, M.M.; Esparza, J.; O’Quinn, E.C.; Hart, A.J.; Biswas, S.; Patil, C.A.; Lonning, S.; et al. Inhibition of TGF-beta signaling by 1D11 antibody treatment increases bone mass and quality in vivo. J. Bone Miner. Res. 2010, 25, 2419–2426. [Google Scholar] [CrossRef] [PubMed]
  253. Tang, Y.; Wu, X.; Lei, W.; Pang, L.; Wan, C.; Shi, Z.; Zhao, L.; Nagy, T.R.; Peng, X.; Hu, J.; et al. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat. Med. 2009, 15, 757–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Hollinger, J.O.; Hart, C.E.; Hirsch, S.N.; Lynch, S.; Friedlaender, G.E. Recombinant Human Platelet-Derived Growth Factor: Biology and Clinical Applications. J. Bone Jt. Surg. Am. Vol. 2008, 90, 48–54. [Google Scholar] [CrossRef] [PubMed]
  255. Vu, T.H.; Shipley, J.; Bergers, G.; Berger, J.E.; Helms, J.A.; Hanahan, D.; Shapiro, S.D.; Senior, R.M.; Werb, Z. MMP-9/Gelatinase B Is a Key Regulator of Growth Plate Angiogenesis and Apoptosis of Hypertrophic Chondrocytes. Cell 1998, 93, 411–422. [Google Scholar] [CrossRef] [Green Version]
  256. Stickens, D.; Behonick, D.J.; Ortega, N.; Heyer, B.; Hartenstein, B.; Yu, Y.; Fosang, A.J.; Schorpp-Kistner, M.; Angel, P.; Werb, Z. Altered endochondral bone development in matrix metalloproteinase 13-deficient mice. Development 2004, 131, 5883–5895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Ramasamy, S.K.; Kusumbe, A.P.; Wang, L.; Adams, R.H. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature 2014, 507, 376–380. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Bone remodeling regulation can be paracrine or endocrine. Several factors participate in paracrine regulation including cytokines (IL-1, IL-6, TNF-alpha, IL-4 and interferon-gamma), PGE2, VEGF, and hypoxia, as well as bone cells. There are three main cell types involved: osteoblasts, osteocytes and osteoclasts. Osteoblasts, which differentiate from mesenchymal progenitors thanks to certain proteins (Runx2, Osx and Wnt) and FGF signaling pathways, are responsible for bone formation. They can also become osteocytes able to regulate osteoblastogenesis through production of inhibitors (DKK-1 and SOST), that inhibit Wnt signaling. Lastly, osteoclasts, involved in bone resorption are activated through RANK-RANKL-OPG signaling pathway cross-talk. Whenever there is a need for bone resorption, osteoblasts and osteocytes express RANKL on their surface, and this then binds to RANK in osteoclast precursors, activating their differentiation. OPG is then secreted to stop bone resorption binding to RANKL blocking the possibility of RANK-RANKL binding and preventing bone resorption. Once activated, mature osteoclasts bind to the bone matrix, becoming polarized. Their cytoskeleton organizes into actin rings forming the sealing zone, which provides an isolated acidic microenvironment, to dissolve minerals and digest the selected bone matrix thanks to the ruffle border (RB). After resorption, the osteoclasts endocytose the degraded collagen fragments, and the calcium and phosphate released are then transported through the cell and liberated at the functional secretory domain before being released into the bloodstream. Bone formation and resorption are also influenced by endocrine regulation. Various factors may be involved, for example, PTH, 1,25(OH) Vitamin D, calcitonin and thyroid hormone.
Figure 1. Bone remodeling regulation can be paracrine or endocrine. Several factors participate in paracrine regulation including cytokines (IL-1, IL-6, TNF-alpha, IL-4 and interferon-gamma), PGE2, VEGF, and hypoxia, as well as bone cells. There are three main cell types involved: osteoblasts, osteocytes and osteoclasts. Osteoblasts, which differentiate from mesenchymal progenitors thanks to certain proteins (Runx2, Osx and Wnt) and FGF signaling pathways, are responsible for bone formation. They can also become osteocytes able to regulate osteoblastogenesis through production of inhibitors (DKK-1 and SOST), that inhibit Wnt signaling. Lastly, osteoclasts, involved in bone resorption are activated through RANK-RANKL-OPG signaling pathway cross-talk. Whenever there is a need for bone resorption, osteoblasts and osteocytes express RANKL on their surface, and this then binds to RANK in osteoclast precursors, activating their differentiation. OPG is then secreted to stop bone resorption binding to RANKL blocking the possibility of RANK-RANKL binding and preventing bone resorption. Once activated, mature osteoclasts bind to the bone matrix, becoming polarized. Their cytoskeleton organizes into actin rings forming the sealing zone, which provides an isolated acidic microenvironment, to dissolve minerals and digest the selected bone matrix thanks to the ruffle border (RB). After resorption, the osteoclasts endocytose the degraded collagen fragments, and the calcium and phosphate released are then transported through the cell and liberated at the functional secretory domain before being released into the bloodstream. Bone formation and resorption are also influenced by endocrine regulation. Various factors may be involved, for example, PTH, 1,25(OH) Vitamin D, calcitonin and thyroid hormone.
Cells 10 00932 g001
Figure 2. Bone metabolism: Modeling vs. Remodeling—While bone modeling implies a change in bone shape or size since resorption and formation occur independently at distinct sites: osteoblasts secrete osteoid matrix in the opposite site where osteoclasts resorb bone. Bone remodeling involves the resorption and formation of bone, one after the other, at the same site to replace old and/or damaged bone by newly formed bone. An initiating remodeling signal, such as hormonal or mechanical signal, is detected by the bone, inducing the release of paracrine factors that lead to retraction of the bone lining cells which exposes the bone surface, allowing recruitment of osteoclast precursors from the capillaries directly into the basic multicellular unit. MSC-F and RANKL, secreted by osteocytes, induce recruitment of precursor cells of hematopoietic lineage, initiating their differentiation to multinucleated osteoclasts. The differentiated attached osteoclasts rearrange their cytoskeleton to adhere to the bone surface, decreasing the pH to as low as 4.5, this dissolving the bone mineral. Once resorption is finished, the osteoclasts go through apoptosis. After resorption, mononuclear cells are recruited to remove collagen fragments from the surface, and then new osteoblasts begin collagen deposition, forming what is known as osteoid matrix, until the cavities are filled. Osteoblasts produce new bone, and some of them become buried within the newly formed bone matrix turning into osteocytes with their extensive canalicular network connecting them to the bone surface lining cells, osteoblasts and other osteocytes. The osteoid mineralizes, and the bone enters into a quiescent phase.
Figure 2. Bone metabolism: Modeling vs. Remodeling—While bone modeling implies a change in bone shape or size since resorption and formation occur independently at distinct sites: osteoblasts secrete osteoid matrix in the opposite site where osteoclasts resorb bone. Bone remodeling involves the resorption and formation of bone, one after the other, at the same site to replace old and/or damaged bone by newly formed bone. An initiating remodeling signal, such as hormonal or mechanical signal, is detected by the bone, inducing the release of paracrine factors that lead to retraction of the bone lining cells which exposes the bone surface, allowing recruitment of osteoclast precursors from the capillaries directly into the basic multicellular unit. MSC-F and RANKL, secreted by osteocytes, induce recruitment of precursor cells of hematopoietic lineage, initiating their differentiation to multinucleated osteoclasts. The differentiated attached osteoclasts rearrange their cytoskeleton to adhere to the bone surface, decreasing the pH to as low as 4.5, this dissolving the bone mineral. Once resorption is finished, the osteoclasts go through apoptosis. After resorption, mononuclear cells are recruited to remove collagen fragments from the surface, and then new osteoblasts begin collagen deposition, forming what is known as osteoid matrix, until the cavities are filled. Osteoblasts produce new bone, and some of them become buried within the newly formed bone matrix turning into osteocytes with their extensive canalicular network connecting them to the bone surface lining cells, osteoblasts and other osteocytes. The osteoid mineralizes, and the bone enters into a quiescent phase.
Cells 10 00932 g002
Figure 3. FGF/FGFR signaling—FGFs can bind to FGFRs with the help of heparan sulfate, a co-factor, and thereby induce their biological effects through activation of four major signaling pathways: RAS-MAPK-ERK1/2, PI3K-AKT-GSK3, PLCγ-PKC, and STAT-Jak.
Figure 3. FGF/FGFR signaling—FGFs can bind to FGFRs with the help of heparan sulfate, a co-factor, and thereby induce their biological effects through activation of four major signaling pathways: RAS-MAPK-ERK1/2, PI3K-AKT-GSK3, PLCγ-PKC, and STAT-Jak.
Cells 10 00932 g003
Figure 4. FGF/FGFR signaling in bone—FGF-2 is highly expressed in bone tissues. There is a high molecular weight (HMW) form, located in the nucleus, that acts as a transcriptional factor, and upregulates the expression of SOST and FGF-23 responsible for inducing mineralization. The low molecular weight (LMW) form is cytoplasmic or membrane associated. The latter can promote osteoblast differentiation and mineralization through the Wnt pathway, BMP-2 signaling, or synergistic action with BMP-2. By activation of FGFR signaling, LMW FGF-2 also activates MAPK-ERK/2, which acts as a transcriptional factor that upregulates mineralization genes such as RUNX2.
Figure 4. FGF/FGFR signaling in bone—FGF-2 is highly expressed in bone tissues. There is a high molecular weight (HMW) form, located in the nucleus, that acts as a transcriptional factor, and upregulates the expression of SOST and FGF-23 responsible for inducing mineralization. The low molecular weight (LMW) form is cytoplasmic or membrane associated. The latter can promote osteoblast differentiation and mineralization through the Wnt pathway, BMP-2 signaling, or synergistic action with BMP-2. By activation of FGFR signaling, LMW FGF-2 also activates MAPK-ERK/2, which acts as a transcriptional factor that upregulates mineralization genes such as RUNX2.
Cells 10 00932 g004
Table 1. Overview of FGF-2 used in craniofacial studies arranged by research type: dose, timing, administration mode and scaffolding.
Table 1. Overview of FGF-2 used in craniofacial studies arranged by research type: dose, timing, administration mode and scaffolding.
StudyApplicationDosage and TimingAdministration and/or
Scaffolding
Results
in vitro studies
Gromolak et al. 2020Ovine bone marrow MSCsFGF-2 (20 ng/mL) alone
or
in combination with
BMP-2 (100 ng/mL)
In culture mediumOsteogenic differentiation
induced by BMP-2 is amplified with FGF-2 supplementation. FGF-2 alone boosted proliferation of smaller cells, but without osteoblast-like structures in culture and decreased expression of osteogenic genes.
Li et al. 2014Murine calvarial osteoblastsFGF-2 (1, 10, 20, 60 ng/mL)In culture mediumDoses ≤10 ng/mL yielded higher cell proliferation
Doses >10 ng/mL decreased proliferation
Increased mineralization at all doses
Sukarawan et al. 2014Stem cells from human
exfoliated deciduous teeth (SHEDs)
FGF-2 (10 ng/mL)In culture mediumFGF-2 maintains cell stemness
Ou et al. 2010Murine calvarial and femur osteoprogenitor cells
Human cancellous bone osteoprogenitor cells from young and old patients
rhFGF-2
(0.0016, 0.016, 0.16, or 1.6 ng/mL)
4, 24, 48, and 72 h
In culture mediumAccelerated proliferation
at all doses
FGF-2 induced proliferation
diminished with age
Varkey et al. 2006Rat bone marrow cellsFGF-2 (2, 10, 50 ng/mL)
and
BMP-2 (50, 150, 500 ng/mL) over 3 weeks
In culture mediumAccelerated mineralization
at 10 ng/mL but reduced
at 50 ng/mL of FGF-2
Synergistic role of FGF-2 and BMP-2 in old rat cells
Animal models
Novais et al. 2019Critical calvarial bone defects in nude miceFGF (10 ng/mL) over 72 h SHEDs in dense collagen matrices in osteogenic culture mediumEnhanced bone formation in calvarial critical size defect
Wang et al. 2019Mandibular defects in non-human primatesFGF-2 (0.25 μg/μL) Calcium phosphate cement for BMP-2 carrier
PGA gel for FGF-2 carrier
Promotion of periodontal
regeneration
Anzai et al. 20162-wall periodontal defects in Beagle dogsFGF-2 (3 mg/mL) vs. vehicleCellulose solution
Direct injection in
the defect
FGF-2 promoted regeneration
in alveolar bone, cementum
and periodontal ligament.
Charles et al. 2015Calvarial bone defects in old miceFGF-2 (5 ng) and BMP-2 (2 μg)Collagen
hydroxyapatite discs
Enhanced bone filling
in the central bone defect area when BMP-2 was
supplemented with FGF-2
Akita et al. 2004Calvarial defects in nude miceFGF-2 (2.5 ng/mL)
and BMP-2 (50 ng/mL)
vs.
FGF-2 alone, BMP-2 alone or vehicle
Transfected human MSCs in gelatin sponge carrierCombination of FGF-2 and BMP-2 showed the most
advanced bone formation within the defects
Clinical studies
Cochran et al. 2016Patients with periodontal
intrabony defects
rhFGF-2 (0.1%, 0.3%, 0.4%) or no applicationβ-TCPIncreased clinical attachment gain and bone fill at
concentrations of 0.4% and 0.3%
De Santana et al. 2015Patients with periodontal
intrabony defects
rhFGF-2 (4 mg/mL) vs. no application Sodium
hyaluronate gel
Enhanced clinical parameters of wound healing compared to negative control
Kitamura et al. 2001Patients with periodontal
intrabony defects
rhFGF-2 (0.2%, 0.3%, 0.4%) or placebo3% hydroxypropyl cellulose gelAt 36 weeks, all defects showed bone fill except placebo
0.3% dose had the best
radiographic outcomes
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Novais, A.; Chatzopoulou, E.; Chaussain, C.; Gorin, C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021, 10, 932. https://doi.org/10.3390/cells10040932

AMA Style

Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells. 2021; 10(4):932. https://doi.org/10.3390/cells10040932

Chicago/Turabian Style

Novais, Anita, Eirini Chatzopoulou, Catherine Chaussain, and Caroline Gorin. 2021. "The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review" Cells 10, no. 4: 932. https://doi.org/10.3390/cells10040932

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop