Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Feb 26, 2024; 16(2): 102-113
Published online Feb 26, 2024. doi: 10.4252/wjsc.v16.i2.102
Crosstalk between Wnt and bone morphogenetic protein signaling during osteogenic differentiation
Pakkath Narayanan Arya, Iyyappan Saranya, Nagarajan Selvamurugan, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India
ORCID number: Nagarajan Selvamurugan (0000-0003-3713-1920).
Author contributions: Arya PN and Saranya I conceptualized and wrote the manuscript; Selvamurugan N reviewed and edited the manuscript and secured funding for this work.
Supported by Indian Council of Medical Research, 2020-0282/SCR/ADHOC-BMS; and Department of Science and Technology, India, DST/INSPIRE Fellowship: 2021/IF210073.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nagarajan Selvamurugan, PhD, Professor, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, Kattankulathur 603203, India. selvamun@srmist.edu.in
Received: October 22, 2023
Peer-review started: October 22, 2023
First decision: December 25, 2023
Revised: January 4, 2024
Accepted: January 22, 2024
Article in press: January 22, 2024
Published online: February 26, 2024

Abstract

Mesenchymal stem cells (MSCs) originate from many sources, including the bone marrow and adipose tissue, and differentiate into various cell types, such as osteoblasts and adipocytes. Recent studies on MSCs have revealed that many transcription factors and signaling pathways control osteogenic development. Osteogenesis is the process by which new bones are formed; it also aids in bone remodeling. Wnt/β-catenin and bone morphogenetic protein (BMP) signaling pathways are involved in many cellular processes and considered to be essential for life. Wnt/β-catenin and BMPs are important for bone formation in mammalian development and various regulatory activities in the body. Recent studies have indicated that these two signaling pathways contribute to osteogenic differentiation. Active Wnt signaling pathway promotes osteogenesis by activating the downstream targets of the BMP signaling pathway. Here, we briefly review the molecular processes underlying the crosstalk between these two pathways and explain their participation in osteogenic differentiation, emphasizing the canonical pathways. This review also discusses the crosstalk mechanisms of Wnt/BMP signaling with Notch- and extracellular-regulated kinases in osteogenic differentiation and bone development.

Key Words: Bone, Mesenchymal stem cells, Osteogenic differentiation, Wnt/β-catenin, Bone morphogenetic proteins

Core Tip: Osteogenesis is the process of new bone formation that is influenced by various signaling pathways, such as the Wnt/β-catenin and bone morphogenetic protein (BMP) pathways. These pathways are crucial for bone formation and regulatory activities during osteogenesis. This review explores the molecular processes and mechanisms of the crosstalk between the Wnt and BMP signaling pathways in osteogenic differentiation and bone development.



INTRODUCTION

Bone is a mineralized connective tissue composed of four cell types: Osteoblasts, osteocytes, bone-lining cells, and osteoclasts. Despite its apparent immobility, bone is an extremely dynamic organ that is constantly resorbed by osteoclasts and neoformed by osteoblasts. The functions of bone-lining cells are key for balancing bone resorption and formation[1]. Bone remodeling is essential for the bone to adapt to the constant mechanical modifications necessary for skeletal functions under various environmental conditions[2]. Bone develops during early fetal development depending on the interactions between the two cell lineages. The primary cells involved in remodeling are osteoblasts and osteoclasts, which are responsible for bone formation and resorption, respectively[3]. Osteoblasts arise from mesenchymal stem cells (MSCs) and aid in bone formation. Osteoclasts are responsible for bone resorption and are derived from a hematopoietic lineage that comprises various osteogenically differentiated cell types in the bone marrow[4]. Modifications may occur during the commitment or differentiation of MSCs to the osteogenic lineage, resulting in calcification or bone loss under various conditions[5]. Osteogenesis or bone formation is a process by which the pre-existing mesenchymal tissue is transformed into bone tissue[6].

Several signaling mechanisms stimulate osteogenesis in stem cells. For example, Wnt signaling is a well-established pathway of osteogenic differentiation[7]. Wnt proteins are crucial for several biological functions, including organogenesis, tissue regeneration, and cancer. The Wnt system comprises two independent intracellular cascades: Canonical (β-catenin-mediated) and non-canonical Wnt pathways. Neither of these factors induce osteogenic differentiation. Wnt/β-catenin signaling is the conventional (or classic) Wnt pathway[8]. Canonical Wnt signaling pathway results in the nuclear translocation of β-catenin protein and regulation of its target genes. In the absence of Wnt ligands, β-catenin is destroyed by an intracellular complex composed of glycogen synthase kinase 3 (GSK-3). Canonical Wnt activation induces the expression of disheveled 1 protein and suppresses GSK-3[9]. Although non-canonical Wnt signaling is less understood compared to the β-catenin-mediated Wnt pathway, it may also contribute to bone tissue development[10]. In Wnt signaling, a set of co-receptors, known as low-density lipoprotein receptor-related proteins 5 or 6 (LRP5/6), enhance the binding of Wnt ligands to frizzled proteins (FZDs). The binding of LRP5/6 inhibitors [known as dickkopf (DKK)] to LRP5/6 coreceptors disrupts Wnt ligand-FZD binding and suppresses Wnt signaling[11].

Another key osteogenic signaling pathway is the bone morphogenetic protein (BMP)/small mothers against decapentaplegic (Smad) signaling pathway, which is also a pro-osteogenic and pro-adipogenic signaling system[12]. This pathway becomes active when transforming growth factor-beta (TGF-β) and superfamily member BMPs bind to the heterodimeric type I/II BMP transmembrane serine/threonine kinase receptors[13]. A Smad-dependent or-independent intracellular cascade is activated depending on the context. The Smad-dependent pathway becomes active when the BMP ligand binds to its specific receptor, inducing the phosphorylation and binding of receptor Smad (Smad1/5/8) to common Smad (Smad4)[14]. The translocation of this complex to the nucleus subsequently modulates the BMP target genes and osteogenic differentiation.

Wnt and BMP pathways are essential signaling pathways in osteogenesis[15]. Activation of Smad proteins initiates the BMP signaling pathway, stimulating the Runt-related transcription factor 2 (Runx2) gene. Runx2 is a transcription factor that promotes the differentiation of MSCs toward the osteogenic lineage[16]. It functions upstream of the osteoblast-specific transcription factor, osterix, and other specialized osteoblastic genes, including Sparc (osteonectin), SPP1 (osteopontin), and collagen type I alpha 1[17]. Wnt signaling, which is important for bone formation, regulates Runx2 expression.

This review outlines the importance of the Wnt and BMP signaling pathways and their individual impact and crosstalk in osteogenic differentiation. We also discuss the currently available techniques and clinical studies on the regulation of Wnt and BMP signaling to promote bone osteogenesis and the limitations of these strategies. In addition, we highlight the potential therapeutic effects of targeting the Wnt-BMP interactions in bone and bone-related ailments.

OSTEOGENIC DIFFERENTIATION

Osteogenesis is a sophisticated multistep mechanism that involves the differentiation of MSCs into osteoblast precursor cells, pre-osteoblasts, osteoblasts, and osteocytes, along with the various ways in which these cells communicate with each other for the formation and remodeling of bones. Most studies have focused on the basic characteristics of MSCs, signaling pathways, and variables promoting the osteogenic differentiation of MSCs[18]. The first phase of the osteogenic differentiation of MSCs, termed the proliferative phase, includes the acquisition and proliferation of osteoprogenitor cells, the second phase involves cell maturation in pre-osteoblasts after extracellular matrix (ECM) development, and the final phase involves matrix mineralization. Various signaling pathways influence each of these actions. TGF-β/BMP and Wnt/β-catenin cascades play key roles in bone healing and have been extensively explored for therapeutic applications[19]. Although distinct factors control them, both cascades involve Runx2, the principal transcription factor involved in osteogenesis[20].

Intramembranous and endochondral ossification

Bone is a dynamic and robust connective tissue that constantly remodels throughout life. There are two basic types of bone formation, called osteogenesis[21], which entails the transformation of pre-existing mesenchymal tissue into bone tissue (Figure 1). Intramembranous ossification refers to the direct conversion of mesenchymal tissue into bone. This process occurs mainly in the skull bones, flat bones of the neurocranium and viscerocranium, and a portion of the clavicle, which is characterized by the condensation of MSCs into osteoblasts upon commitment as osteoprogenitors[22]. Osteoblasts build a collagen-proteoglycan matrix that binds to calcium salts. This interaction causes calcification of the pre-bone (osteoid) matrix. Generally, a layer of the osteoid matrix constituted by osteoblasts is separated from the calcification region. However, osteoblasts become entangled in the calcified matrix and transform into bone cells, called osteocytes. As calcification advances, bony spicules extend from the location where ossification begins[23]. Furthermore, dense mesenchymal cells comprising the periosteum (protective barrier of bone) surround the entire calcified spicule region. Osteoblastic cells on the inner surface of the periosteum lay down the osteoid matrix parallel to the previously existing spicules. Multiple layers of bone develop during this process. Mature osteoblasts differentiate into bone-lining cells and osteocytes or cause cell death. Osteocytes can act as mechanical sensors, modify the perilacunar environment, and contribute to bone function by interacting with organic and inorganic compounds in response to mechanical stimuli[24].

Figure 1
Figure 1 Schematic representation of intramembranous and endochondral ossification. A: Undifferentiated mesenchymal cells develop into osteoprogenitor cells (osteoblasts), which lay down the osteoid matrix and mineralize to form ossification centers. Osteoblasts die due to apoptosis or become trapped in the matrix, developing into osteocytes; B: Condensed mesenchymal cells commit to become the cartilage and undergo chondrogenic differentiation. Chondrocytes at the primordium core establish a growth plate and undergo hypertrophy. Hypertrophic chondrocytes calcify and are penetrated by microvessels, resulting in primary ossification. Vessels infiltrate the epiphyses and produce secondary ossification centers in conjunction with osteoblasts and bone marrow. The growth plate aids in long bone formation. MSCs: Mesenchymal stem cells.

In contrast, MSCs initially develop into cartilage and are eventually replaced by bone via endochondral ossification (indirect ossification) in the long bones, vertebrae, and base and posterior regions of the skull. Endochondral ossification is divided into five phases. First, mesenchymal cells commit to becoming cartilage cells[25]. Second, cartilage calcification occurs when chondrocytes move toward the center of the cartilage model and change their ECM content. Following the establishment of the main ossification center, the cartilage model gradually became vascularized via vascular bud penetration. Through the vascular buds, osteoprogenitor cells enter the cartilage model and develop into osteoblasts[26]. The creation of the main ossification center in the diaphysis (central section of the bone) occurs during this process. Following the establishment of a secondary ossification center, the bone gradually replaces the cartilage in the diaphysis via osteoblast proliferation, while the cartilage continues to grow at the ends of the bone (epiphysis), increasing the bone length[27]. This is where the secondary ossification center developed. Finally, mature and compact bones are formed in the cartilage model (Figure 1).

Roles and regulation of Wnt signaling in osteogenesis

Wnt signaling is an immensely conserved pathway that is crucial for the development of several tissues and organs. Wnt signaling regulates cellular activities, including cell fate, differentiation, migration, and proliferation, are regulated by Wnt signaling[28]. A subset of extracellular Wnt ligands, including 19 secreted glycoproteins, activate the canonical Wnt pathway. Wnt ligands bind to the dual receptor complex of FZD and either LRP5 or LRP6 to initiate this signaling. To avoid β-catenin phosphorylation and subsequent proteasomal breakdown, the multiprotein “destruction complex” for β-catenin is deactivated. After that, β-catenin builds up in the cytoplasm and translocates into the nucleus, where it acts with transcription factors to control the transcription of the target gene. Numerous studies have emphasized the crucial role that canonical Wnt signaling plays in maintaining bone homeostasis; when this pathway is activated, bone mass and bone production strength increase[29-31].

Non-canonical Wnt signaling routes function without the assistance of β-catenin, whereas the canonical pathway facilitates signaling through the stability of β-catenin[32]. Wnt signaling promotes MSC development toward the osteoblast lineage and inhibits differentiation toward the adipocyte and chondrocyte lineages[33]. Abnormal changes in the Wnt signaling pathway markers are linked to changes in bone metabolism and MSCs’ osteogenic ability of MSCs[34]. Activation of the Wnt signaling pathway can boost Wnt-related gene expression, restore the osteogenic capacity of bone marrow stem cells (BMSCs), and reduce bone loss. DKK-1 diminishes the osteogenic capability of BMSCs, resulting in bone loss. Earlier studies stated that the reduced expression and osteogenic ability of β-catenin, phospho-GSK-3β, and lymphoid enhancing factor 1 (LEF1) in BMSCs obtained during osteoporosis (OP-BMSCs) were upregulated by transducing OP-BMSCs with methyltransferase-like 3 (METTL3). Overexpression of METTL3 partially restored the osteogenic ability of OP-BMSCs, activated the Wnt signaling pathway, and elevated the expression of osteopontin, Runx2, phospho-GSK-3β, β-catenin, and LEF1[35].

Lysine-specific demethylase 4A (KDM4A) inhibited the canonical Wnt signaling pathway. Overexpression of KDM4A increased the expression of secreted FZD-related protein 4 (SFRP4) and CCAAT/enhancer-binding protein α, resulting in the promotion of adipogenesis by inhibiting canonical Wnt signaling and decreasing osteogenesis from marrow stromal progenitor cells. These results demonstrated the existence of a network between KDM4A, SFRP4, and Wnt/β-catenin signaling that balances osteogenic and adipogenic development. These findings show that KDM4A may be an appealing prospective target for novel therapeutics targeting metabolic diseases, such as osteoporosis[36]. Recent studies have reported the involvement of various molecules, such as transcription factors and protein-coding genes, in the regulation of osteogenesis via the Wnt signaling pathway (Table 1).

Table 1 Molecules regulating osteogenesis via the Wnt/β-catenin signaling pathway.
No
Name
Experimental model(s)
Effect
Role
Ref.
1CRYABIn vitro and in vivoPromotes osteogenic activityActs as a regulator of the osteogenic differentiation of BMSCs and protects β-catenin from proteasomal degradation[37]
2LGR6In vitro and in vivoPromotes osteogenic activityLGR6 improves the signaling pathway mediated by Wnt/β-catenin during osteogenesis by stabilizing β-catenin within the cytoplasm[38]
3KLF14In vitro and in vivoInhibits osteogenic activityKLF14 and Wnt3A promoter interaction suppresses the expression of Wnt3A and downstream targets of Wnt signaling during osteogenesis[39]
4DLX2In vitroPromotes osteogenic activityDLX2 binds to WNT1, upregulating WNT1 transcription and activating the Wnt/β-catenin signaling pathway[40]
5FOXQ1In vitro and in vivoPromotes osteogenic activityFOXQ1 regulates the activities of Wnt/β-catenin pathway by binding to annexin A2[41]
6BRD4In vitroPromotes osteogenic activityBRD4 improves the expression of Wnt3a and β-catenin by binding to the promoter region of Wnt3a or β-catenin during osteogenic differentiation in BMSCs[42]
7LECT2In vitro and in vivoNegatively regulates osteogenic activityLECT2 knockdown triggers the Wnt/β-catenin pathway, promoting the osteoblast differentiation of MSCs[43]
Roles and regulation of BMP signaling in osteogenesis

BMPs correlate with TGFβ1 superfamily, over 30 known animals, and 14 distinct categories of human BMPs. Several signaling proteins, mainly BMP2, BMP6, BMP7, and BMP9, are effective osteogenic inducers both in vitro and in vivo[44]. They initiate a signaling cascade by interacting with dimeric complexes of type I and type II transmembrane serine-threonine kinase receptors. After activating the receptors, Smad proteins are phosphorylated and, along with Smad4, the Smad complex translocates to the nucleus to stimulate the transcription of several BMP-responsive genes. BMPs are essential for the osteogenic development of BMSCs. Several intracellular signaling proteins and cell membrane receptors enable BMPs to exert their biological effects. BMP/Smad signaling is a crucial pathway in this process[45].

The primary downstream components of BMPs that induce the osteogenic differentiation of BMSCs are Smad 1/5/8 and the transcription factors, Runx2 and Sp7[46]. Studies have shown that Runx2 is not only a downstream target of the BMP pathway but may also regulate BMP expression. The underlying mechanism between Runx2 and the BMP pathway in primary BMSCs from patients with craniofacial deformities patients was investigated in a recent study, which demonstrated that Runx2 controls the BMP4 pathway by repressing Chordin-like 1 (CHRDL1) transcription. Researchers have discovered an intriguing RUNX2/CHRDL1/BMP4 axis that promotes osteogenic differentiation, and hypothesized that BMP4 could serve as a possible treatment for bone disorders[47]. Previous studies demonstrated that BMP2 is a potent activator of osteoblastogenesis. Runx2 and SP7 are prominent BMP2-target genes, whereas DLX5 is crucial for SP7 expression during BMP signaling. Interaction of osteomodulin with BMP2 positively regulated osteogenesis[48].

Li et al[49] identified a novel mechanism for BMP2 in boosting osteogenic differentiation of MSCs; particularly, BMP2 modulated mitochondrial activity via regulating peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1) to enhance osteogenic differentiation of MSCs. Collectively, these results suggested that BMP2 and mitochondrial activity influenced osteogenic differentiation and bone development. This study reported that overexpression of BMP2 increased osteogenic differentiation and addressed the correlation between BMP2, mitochondrial activity, and PGC-1[49]. In recent studies, various molecules, such as protein-coding genes, cytokines, growth factors, and phytocompounds, have been shown to regulate osteogenesis via the BMP signaling pathway (Table 2).

Table 2 Molecules regulating osteogenesis via the bone morphogenetic protein/Smad-dependent pathway.
No
Name
Experimental model(s)
Effect
Role
Ref.
1LMCD1In vitro and in vivoPromotes osteogenic activityActs along with Smad-specific E3 ubiquitin protein ligase 1 to modulate the ubiquitination levels of Smad1 and Runx2[50]
2CarnosolIn vitroInduces osteogenic activityStimulates osteogenesis via the activation of the BMP signaling pathway and consequent upregulation of the expression of downstream target genes[51]
3IL-1βIn vitro and in vivoPromotes osteogenic activityEnhances the osteogenic differentiation potential of mouse bone marrow MSCs by activating the BMP/Smad osteogenic signaling pathway[52]
4NGFIn vitroPromotes osteogenic activityIncreases BMP2 expression and phosphorylation and nuclear translocation of Smad1/5/8[53]
5NobiletinIn vitroPromotes osteogenic activityStimulates osteogenic differentiation by promoting Runx2 and BMP2 signaling[54]
6SimvastatinIn vitro and in vivoPromotes osteogenic activityIncreases the expression of osteogenic differentiation- and BMP2/Smads pathway-associated genes[55]
7IsoquercitrinIn vitroPromotes osteogenic activityStimulates osteogenic differentiation via Runx2 expression in osteoblasts and BMP pathway in BMSCs[56]
CROSSTALK BETWEEN THE WNT AND BMP SIGNALING PATHWAYS IN OSTEOGENESIS

The Wnt and BMP signaling pathways play significant roles in osteogenesis. They can act independently of one another via separate ligands, receptors, and cytoplasmic and nuclear signal transducers without sharing any significant pathway aspects. However, in many biological circumstances, Wnt and BMP ligands happen spatially or across time in overlapping or complementary ways, as if they are “crosstalking” with one another[57]. Indeed, recent research has identified several instances in which these two pathways interact with or attenuate each other, resulting in outcomes that either alone cannot attain[58]. Recently, numerous studies have explored small-molecule compounds as osteogenic inducers[59] in cell-based therapies for bone regeneration, as shown in Figure 2[60]. The intense osteogenic effect of GD-A, a Ganoderma lucidum-derived tetracyclic triterpenoid molecule, on human amniotic MSCs has been previously reported. Key elements, including β-catenin, Wnt3, FZD4, BMP3, BMP4, Smad4, and Smad5, have significantly up-regulated in association with Wnt/β-catenin and BMP/SMAD signaling, which in turn stimulated human adipose-derived MSC differentiation into osteoblasts[61]. Recent studies have reported that albiflorin, an active component in Paeonia lactiflora, increased BMP-2/Smad and Wnt/β-catenin signaling and promoted Runx2 expression, a critical transcription factor for osteoblast differentiation to produce osteogenic genes. Furthermore, the potential of albiflorin to stimulate osteoblast differentiation improved fracture healing in a rat femoral fracture model[16]. The conditioned exosomes originated from human-exfoliated deciduous tooth stem cells (SHED) significantly promoted the osteogenic differentiation of periodontal ligament stem cells via the Wnt/β-catenin and BMP/Smad signaling pathways. Furthermore, crosstalk between the Wnt and BMP signaling pathways regulates the activity of dental stem cells. Increased phosphorylation of Smad1/5/8 and nuclear β-catenin expression improved both BMP/Smad signaling and Wnt/β-catenin signaling. SHED-exosomes exosomes have elevated levels of Wnt3a and BMP2. Silencing Wnt3a and BMP2 in SHED-exosomes slightly restored the increased osteogenic differentiation. These results provide new insights into the application of SHED exosomes in treating periodontitis-induced bone abnormalities[62]. Polydatin (PD), a natural resveratrol glucoside obtained from the roots of Polygonum cuspidatum, has been found to enhance the levels of the β-catenin and enable its nucleus translocation, resulting in increased expression of downstream target genes. PD also aided human bone marrow-derived MSCs (hBMSCs) osteogenesis by activating the BMP2-induced Wnt signaling pathway and increasing the accumulation and nuclear translocation of β-catenin[63]. Sclerostin small-molecule inhibitors (SMIs) significantly promoted Wnt/β-catenin and BMP signaling in vitro and in vivo, enhancing osteogenesis while significantly reducing bone resorption activity. Further findings imply that sclerostin inhibition by SMIs might occur in a multifrontal drive onto osteogenesis by blocking the receptor activator of nuclear factor-kappaB ligand-mediated osteoclastogenic response to endogenous BMP-2 despite increasing both Wnt and BMP signaling[64]. The extract of Juglans regia L (JRL) enhanced the expression level of osteogenic genes in hBMSCs. Meanwhile, JRL extract stimulated the differentiation of osteogenic cells and cell autophagy by activating the BMP2/Smad/Runx2 and Wnt/β-catenin signaling pathways[65].

Figure 2
Figure 2 Schematic illustration of small molecule compounds inducing the osteogenic differentiation of mesenchymal stem cells. Small molecule compounds stimulate the bone morphogenetic protein signaling pathway and promote β-catenin accumulation. Then, β-catenin migrates to the nucleus and forms a β-catenin-T-cell factor/lymphoid enhancer-binding factor complex to initiate the transcription of genes associated with osteogenesis. GSK-3β: Glycogen synthase kinase 3 beta; APC: Adenomatosis polyposis coli; CK1-α: Casein kinase 1α; DSH: Disheveled; TCF/LEF: T-cell factor/lymphoid enhancer-binding factor; DKK1: Dickkopf 1; LRP5/6: Lipoprotein receptor-related proteins 5 or 6; BMP: Bone morphogenetic protein.

Wang et al[66] reported that a cycloxygenase-2 specific inhibitor significantly decreased BMP9-induced osteogenic markers in MSCs, and this adverse effect was substantially ameliorated by the inclusion of all-trans-retinoic acid (ATRA), an important vitamin A derivative. Additional research revealed that ATRA’s reversal effect may be mediated by partially up-regulating the Wnt/β-catenin pathway. While committing progenitors toward osteoblastic lineage, BMP9 promoted the activity of the Wnt/β-catenin pathway, and silencing β-catenin diminished BMP9-induced osteogenic differentiation in MSCs[66]. A recent study has revealed the effects of a highly water-soluble curcuminoid-rich extract (CRE) on alkaline phosphatase activity. The CRE caused mouse pre-osteoblasts (MC3T3-E1) to become osteoblasts by Wnt/β-catenin and BMP signaling pathways[67]. T63, a new small molecular weight molecule discovered by Zhao et al[68], is a potent upregulator of Runx2 activity that promotes osteogenic differentiation and bone development. T63 was also reported to block adipogenic differentiation in C3H10T1/2 cells when osteogenic differentiation was induced and seemed to influence Runx2 activity indirectly via the BMPs/Smads and Wnt/β-catenin signaling. Recently, plant metabolites have been assigned to many natural medicinal compounds, particularly against metabolic disorders, such as diabetes, cardiovascular disease, and osteoporosis. 3,5-dicaffeoylepi-quinic acid (DCEQA), a bioactive derivative of caffeoylquinic acid, was recently found to exert anti-osteoporotic activity in hBMSCs. Treatment with DCEQA boosts both the mRNA and protein expression levels of osteogenic markers, which improves osteogenesis in osteo-induced hBMSCs via the initiation of BMP and Wnt-dependent pathways[69].

OTHER CROSSTALK PATHWAYS REGULATING OSTEOGENIC DIFFERENTIATION
Wnt/Notch pathway

Wnt and Notch pathways complement each other[70]. Notch1 intercellular domain (NICD) suppresses the activation of the Wnt/β-catenin pathway and expression of osteogenic factors, thereby suppressing osteogenic differentiation in human aortic valve interstitial cells (hAVICs). Reduced Notch1 expression and subsequently decreased NICD release in the nucleus of hAVICs are caused by Notch1 promoter methylation[71], which promotes Wnt/catenin pathway activation and the production of osteogenic-specific factors in hAVICs. According to Lee et al[72], competing Wnt and Notch signaling influences progenitor cell proliferation and their consequent differentiation into bone-forming osteoblasts. Wnt signaling stimulates early-stage osteoblast formation and inhibits terminal differentiation. Notch signaling maintains the bone marrow supply of intact stem cells and inhibits early progenitor differentiation. Consequently, low Notch activity is necessary to initiate early stage osteoblast differentiation.

BMP/Notch pathway

Notch signaling is a critical cell-to-cell communication route that regulates stem cell fate throughout embryonic development, including proliferation, self-renewal, and differentiation[73]. Numerous recent studies have shown that BMP and Notch signals interact to either promote or contradict each other during osteogenesis[74]. Activin-like kinase 2 overexpression may play a role in the ability of Notch signaling to increase the activity of BMP9 and promote osteogenic differentiation of MSCs[75]. Recent studies have revealed that BMP9 is the most effective inducer of osteogenic differentiation and is anticipated to play an integral role in tissue engineering. According to Cui et al[76], BMP9 increases the expression of Notch receptors and ligands during the intermediate stages of osteogenic differentiation. BMP9 also upregulated Hey1, a Notch downstream target gene, indicating that BMP9 may function upstream of Notch signaling. These findings suggest that Notch signaling may be critical in coordinating BMP9-induced osteogenic differentiation of MSCs.

Wnt/extracellular signal-regulated kinase 1/2 pathway

Extracellular signal-regulated kinase (ERK)1/2 signaling pathway is one of five traditional mitogen-activated protein kinase (MAPK) signaling pathways. MAPK pathways are found in the cytoplasm of most organisms and are involved in various physiological activities, including osteogenic differentiation of stem cells[77]. During osteoblast formation, activated ERK phosphorylates ELK-1 and stimulates various target proteins, such as the transcription factor complex activator protein 1 and osteoblast-specific transcription factors[78]. Runx2 is phosphorylated[79] and activated by ERK, thereby facilitating matrix mineralization and osteogenic differentiation. Interestingly, Wnt and ERK1/2 signaling pathways exhibit correlations with dihydroartemisinin (DHA) to induce the osteogenic development of MSCs. Inhibition of the ERK1/2 signaling pathway increases the osteogenic response in hMSCs, possibly because of the cell type or treatment. DHA barely affects the proliferation of hMSCs but improves osteogenic differentiation via the Wnt and ERK1/2 signaling pathways. However, the use of U0126 to inhibit the ERK1/2 signaling system decreases matrix mineralization and Runx2 protein expression[80], suggesting that the ERK1/2 pathway substantially influences DHA-induced osteogenic differentiation.

BMP/ERK1/2 pathway

ERK1/2 is among the most studied members of the MAPK family that is essential for the osteogenic differentiation of many distinct cell types, such as osteoblasts, BMSCs, and adipose-derived stem cells[81]. BMP-2 stimulates the p38, ERK1/2, and c-Jun N-terminal protein kinase 1/2 signaling pathways, further stimulating the expression and activation of the osteogenic-specific transcription factor, Runx2[82]. Daigang et al[83] reported that lipopolysaccharide-induced inflammation inhibits BMP-9-mediated Runx2 expression in vitro by activating the p38MAPK and ERK1/2 signaling. Inhibitory effect of lipopolysaccharide (LPS) on BMP-9-induced osteogenic differentiation may be reversed by blocking MAPK signaling using inhibitors. Additionally, MAPK mediates the negative regulatory functions of LPS in the BMP-9-induced activation of Smad signaling.

CONCLUSION

Wnt and BMP signaling pathways are known for their roles in bone development and homeostasis regulation. In this review, we discuss the interplay between both canonical Wnt and BMP pathways in stimulating the expression of downstream osteogenic target genes during osteogenic differentiation via accumulation and translocation of β-catenin to the nucleus and formation of the β-catenin–TCF/LEF complex (Figure 3). This functional interaction between the BMP and Wnt pathways is vital for combining the anabolic actions of both pathways in bone formation. The crosstalk between the Wnt/β-catenin/BMP and other signaling pathways, such as ERK and Notch pathways, implies that these pathways play critical regulatory roles in osteogenic differentiation and bone formation. However, further investigation is necessary to determine the interplay between Wnt/BMP signaling and other signaling pathways, such as the Hedgehog, fibroblast growth factor, Hippo, and TGF-β pathways, in the regulation of osteogenic development. Recent studies have highlighted the potential of small molecules for the treatment of common bone-related illnesses by targeting the Wnt and BMP signaling cascades. Future studies should focus on determining the specific mechanisms of small molecules and Wnt/BMP signaling in osteogenic differentiation.

Figure 3
Figure 3 Schematic illustration of the crosstalk between the Wnt and bone morphogenetic protein signaling pathways in osteogenesis. A: Binding of Wnt-specific ligand initiates the Wnt signaling pathway. In canonical pathway, activated disheveled 1 protein suppresses glycogen synthase kinase 3 and inhibits β-catenin degradation, thereby increasing the osteogenic gene expression. In non-canonical signaling, both Wnt Ca2+-dependent and Wnt planar cell polarity contribute to the upregulation of osteogenic gene expression; B: Bone morphogenetic proteins (BMPs) bind to the heterodimeric type I/II BMP transmembrane serine/threonine kinase receptors and activate the BMP signaling pathway. The canonical pathway becomes active when the BMP ligand binds to its specific receptor, inducing the phosphorylation and binding of receptor-Smads (Smad1/5/8) to the common Smad (Smad4). The translocation of this complex to the nucleus then modulates the expression of BMP target genes and osteogenic differentiation. In non-canonical pathway, activated mitogen-activated protein kinase induces extracellular signal-regulated kinase 1/2 and contributes to the upregulation of Runx2 expression; C: Upregulated Wnt and BMP signaling pathways induce the expression of Runx2/Osterix genes, followed by the expression of osteoblast differentiation genes, resulting in osteogenesis. This involves mesenchymal stem cell differentiation into osteogenic progenitors, pre-osteoblasts, and mature osteoblasts. GSK-3β: Glycogen synthase kinase 3 beta; APC: Adenomatosis polyposis coli; CK1-α: Casein kinase 1 alpha; PKC: Protein kinase C; NFAT: Nuclear factor of activated T cells; Rho: Ras homolog gene family; RAC: Ras-related C3 botulinum toxin substrate; JNK: Jun N-terminal kinase; DVL: Disheveled; TCF/LEF: T-cell factor/lymphoid enhancer-binding factor; MAPK: Mitogen-activated protein kinase; ERK1/2: Extracellular signal-regulated kinase 1/2; MSCs: Mesenchymal stem cells; BMP: Bone morphogenetic protein.
Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Li SC, United States; Li XM, China; Yan ZQ, China S-Editor: Wang JJ L-Editor: A P-Editor: Zhao S

References
1.  Black JD, Tadros BJ. Bone structure: from cortical to calcium. Orthop Trauma. 2020;34:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Zou ML, Chen ZH, Teng YY, Liu SY, Jia Y, Zhang KW, Sun ZL, Wu JJ, Yuan ZD, Feng Y, Li X, Xu RS, Yuan FL. The Smad Dependent TGF-β and BMP Signaling Pathway in Bone Remodeling and Therapies. Front Mol Biosci. 2021;8:593310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 78]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
3.  van Dijk Christiansen P, Andreasen CM, El-Masri BM, Laursen KS, Delaisse JM, Andersen TL. Osteoprogenitor recruitment and differentiation during intracortical bone remodeling of adolescent humans. Bone. 2023;177:116896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
4.  Bolamperti S, Villa I, Rubinacci A. Bone remodeling: an operational process ensuring survival and bone mechanical competence. Bone Res. 2022;10:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 72]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
5.  Zhang J, Jia G, Xue P, Li Z. Melatonin restores osteoporosis-impaired osteogenic potential of bone marrow mesenchymal stem cells and alleviates bone loss through the HGF/PTEN/Wnt/β-catenin axis. Ther Adv Chronic Dis. 2021;12:2040622321995685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
6.  Lin Z, Zhang X, Fritch MR, Li Z, Kuang B, Alexander PG, Hao T, Cao G, Tan S, Bruce KK, Lin H. Engineering pre-vascularized bone-like tissue from human mesenchymal stem cells through simulating endochondral ossification. Biomaterials. 2022;283:121451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
7.  Thomas S, Jaganathan BG. Signaling network regulating osteogenesis in mesenchymal stem cells. J Cell Commun Signal. 2022;16:47-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
8.  Jiang B, Xu J, Zhou Y, Mao J, Guan G, Xu X, Mei L. Estrogen Enhances Osteogenic Differentiation of Human Periodontal Ligament Stem Cells by Activating the Wnt/β-Catenin Signaling Pathway. J Craniofac Surg. 2020;31:583-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
9.  Wang B, Khan S, Wang P, Wang X, Liu Y, Chen J, Tu X. A Highly Selective GSK-3β Inhibitor CHIR99021 Promotes Osteogenesis by Activating Canonical and Autophagy-Mediated Wnt Signaling. Front Endocrinol (Lausanne). 2022;13:926622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
10.  Ben-Ghedalia-Peled N, Vago R. Wnt Signaling in the Development of Bone Metastasis. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
11.  Hong G, He X, Shen Y, Chen X, Yang F, Yang P, Pang F, Han X, He W, Wei Q. Chrysosplenetin promotes osteoblastogenesis of bone marrow stromal cells via Wnt/β-catenin pathway and enhances osteogenesis in estrogen deficiency-induced bone loss. Stem Cell Res Ther. 2019;10:277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
12.  Zhao B, Xing G, Wang A. The BMP signaling pathway enhances the osteoblastic differentiation of bone marrow mesenchymal stem cells in rats with osteoporosis. J Orthop Surg Res. 2019;14:462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
13.  Chen CN, Chang HI, Yen CK, Liu WL, Huang KY. Mechanical Stretch Induced Osteogenesis on Human Annulus Fibrosus Cells through Upregulation of BMP-2/6 Heterodimer and Activation of P38 and SMAD1/5/8 Signaling Pathways. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
14.  Li N, Liu J, Liu H, Wang S, Hu P, Zhou H, Xiao J, Liu C. Altered BMP-Smad4 signaling causes complete cleft palate by disturbing osteogenesis in palatal mesenchyme. J Mol Histol. 2021;52:45-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
15.  Wei XF, Chen QL, Fu Y, Zhang QK. Wnt and BMP signaling pathways co-operatively induce the differentiation of multiple myeloma mesenchymal stem cells into osteoblasts by upregulating EMX2. J Cell Biochem. 2019;120:6515-6527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
16.  Kim JH, Kim M, Hong S, Kim EY, Lee H, Jung HS, Sohn Y. Albiflorin Promotes Osteoblast Differentiation and Healing of Rat Femoral Fractures Through Enhancing BMP-2/Smad and Wnt/β-Catenin Signaling. Front Pharmacol. 2021;12:690113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
17.  Zhou L, Kuai F, Shi Q, Yang H. Doxorubicin restrains osteogenesis and promotes osteoclastogenesis in vitro. Am J Transl Res. 2020;12:5640-5654.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Tan Z, Zhou B, Zheng J, Huang Y, Zeng H, Xue L, Wang D. Lithium and Copper Induce the Osteogenesis-Angiogenesis Coupling of Bone Marrow Mesenchymal Stem Cells via Crosstalk between Canonical Wnt and HIF-1α Signaling Pathways. Stem Cells Int. 2021;2021:6662164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
19.  Hang K, Ye C, Xu J, Chen E, Wang C, Zhang W, Ni L, Kuang Z, Ying L, Xue D, Pan Z. Apelin enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly through Wnt/β-catenin signaling pathway. Stem Cell Res Ther. 2019;10:189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
20.  Liu DD, Zhang CY, Liu Y, Li J, Wang YX, Zheng SG. RUNX2 Regulates Osteoblast Differentiation via the BMP4 Signaling Pathway. J Dent Res. 2022;101:1227-1237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  Zhang J, Ma Z, Yan K, Wang Y, Yang Y, Wu X. Matrix Gla Protein Promotes the Bone Formation by Up-Regulating Wnt/β-Catenin Signaling Pathway. Front Endocrinol (Lausanne). 2019;10:891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
22.  Inoue S, Fujikawa K, Matsuki-Fukushima M, Nakamura M. Repair processes of flat bones formed via intramembranous versus endochondral ossification. J Oral Biosci. 2020;62:52-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
23.  Hartmann C, Yang Y. Molecular and cellular regulation of intramembranous and endochondral bone formation during embryogenesis. Principles Bone Biol. 2020;5-44.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Wang L, Huang J, Moore DC, Song Y, Ehrlich MG, Yang W. SHP2 regulates intramembranous ossification by modifying the TGFβ and BMP2 signaling pathway. Bone. 2019;120:327-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
25.  Galea GL, Zein MR, Allen S, Francis-West P. Making and shaping endochondral and intramembranous bones. Dev Dyn. 2021;250:414-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
26.  Prein C, Beier F. ECM signaling in cartilage development and endochondral ossification. Curr Top Dev Biol. 2019;133:25-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
27.  Fernández-Iglesias Á, Fuente R, Gil-Peña H, Alonso-Durán L, Santos F, López JM. The Formation of the Epiphyseal Bone Plate Occurs via Combined Endochondral and Intramembranous-Like Ossification. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
28.  Roa LA, Bloemen M, Carels CEL, Wagener FADTG, Von den Hoff JW. Retinoic acid disrupts osteogenesis in pre-osteoblasts by down-regulating WNT signaling. Int J Biochem Cell Biol. 2019;116:105597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
29.  Zhu B, Xue F, Zhang C, Li G. Ginkgolide B promotes osteoblast differentiation via activation of canonical Wnt signalling and alleviates osteoporosis through a bone anabolic way. J Cell Mol Med. 2019;23:5782-5793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
30.  Yang X, Wang G, Wang Y, Zhou J, Yuan H, Li X, Liu Y, Wang B. Histone demethylase KDM7A reciprocally regulates adipogenic and osteogenic differentiation via regulation of C/EBPα and canonical Wnt signalling. J Cell Mol Med. 2019;23:2149-2162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
31.  Lerner UH, Kindstedt E, Lundberg P. The critical interplay between bone resorbing and bone forming cells. J Clin Periodontol. 2019;46 Suppl 21:33-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
32.  Kim JH, Liu X, Wang J, Chen X, Zhang H, Kim SH, Cui J, Li R, Zhang W, Kong Y, Zhang J, Shui W, Lamplot J, Rogers MR, Zhao C, Wang N, Rajan P, Tomal J, Statz J, Wu N, Luu HH, Haydon RC, He TC. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Ther Adv Musculoskelet Dis. 2013;5:13-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 237]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
33.  Hung CC, Chaya A, Liu K, Verdelis K, Sfeir C. The role of magnesium ions in bone regeneration involves the canonical Wnt signaling pathway. Acta Biomater. 2019;98:246-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
34.  Peng S, Shi S, Tao G, Li Y, Xiao D, Wang L, He Q, Cai X, Xiao J. JKAMP inhibits the osteogenic capacity of adipose-derived stem cells in diabetic osteoporosis by modulating the Wnt signaling pathway through intragenic DNA methylation. Stem Cell Res Ther. 2021;12:120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
35.  Wu T, Tang H, Yang J, Yao Z, Bai L, Xie Y, Li Q, Xiao J. METTL3-m(6) A methylase regulates the osteogenic potential of bone marrow mesenchymal stem cells in osteoporotic rats via the Wnt signalling pathway. Cell Prolif. 2022;55:e13234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
36.  Qi Q, Wang Y, Wang X, Yang J, Xie Y, Zhou J, Li X, Wang B. Histone demethylase KDM4A regulates adipogenic and osteogenic differentiation via epigenetic regulation of C/EBPα and canonical Wnt signaling. Cell Mol Life Sci. 2020;77:2407-2421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
37.  Zhu B, Xue F, Li G, Zhang C. CRYAB promotes osteogenic differentiation of human bone marrow stem cells via stabilizing β-catenin and promoting the Wnt signalling. Cell Prolif. 2020;53:e12709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
38.  Liu SL, Zhou YM, Tang DB, Zhou N, Zheng WW, Tang ZH, Duan CW, Zheng L, Chen J. LGR6 promotes osteogenesis by activating the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 2019;519:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
39.  Weng J, Wu J, Chen W, Fan H, Liu H. KLF14 inhibits osteogenic differentiation of human bone marrow mesenchymal stem cells by downregulating WNT3A. Am J Transl Res. 2020;12:4445-4455.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Zeng X, Wang Y, Dong Q, Ma MX, Liu XD. DLX2 activates Wnt1 transcription and mediates Wnt/β-catenin signal to promote osteogenic differentiation of hBMSCs. Gene. 2020;744:144564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
41.  Xiang L, Zheng J, Zhang M, Ai T, Cai B. FOXQ1 promotes the osteogenic differentiation of bone mesenchymal stem cells via Wnt/β-catenin signaling by binding with ANXA2. Stem Cell Res Ther. 2020;11:403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
42.  Wang K, Zhao Z, Wang X, Zhang Y. BRD4 induces osteogenic differentiation of BMSCs via the Wnt/β-catenin signaling pathway. Tissue Cell. 2021;72:101555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
43.  Xu Z, He J, Zhou X, Zhang Y, Huang Y, Xu N, Yang H. Down-regulation of LECT2 promotes osteogenic differentiation of MSCs via activating Wnt/β-catenin pathway. Biomed Pharmacother. 2020;130:110593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
44.  Sun W, Li M, Zhang Y, Huang Y, Zhan Q, Ren Y, Dong H, Chen J, Li Z, Fan C, Huang F, Shen Z, Jiang Z. Total flavonoids of rhizoma drynariae ameliorates bone formation and mineralization in BMP-Smad signaling pathway induced large tibial defect rats. Biomed Pharmacother. 2021;138:111480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
45.  Huang Z, Wei H, Wang X, Xiao J, Li Z, Xie Y, Hu Y, Li X, Wang Z, Zhang S. Icariin promotes osteogenic differentiation of BMSCs by upregulating BMAL1 expression via BMP signaling. Mol Med Rep. 2020;21:1590-1596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
46.  Ahmadi A, Mazloomnejad R, Kasravi M, Gholamine B, Bahrami S, Sarzaeem MM, Niknejad H. Recent advances on small molecules in osteogenic differentiation of stem cells and the underlying signaling pathways. Stem Cell Res Ther. 2022;13:518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
47.  Liu T, Li B, Zheng XF, Jiang SD, Zhou ZZ, Xu WN, Zheng HL, Wang CD, Zhang XL, Jiang LS. Chordin-Like 1 Improves Osteogenesis of Bone Marrow Mesenchymal Stem Cells Through Enhancing BMP4-SMAD Pathway. Front Endocrinol (Lausanne). 2019;10:360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
48.  Lin W, Zhu X, Gao L, Mao M, Gao D, Huang Z. Osteomodulin positively regulates osteogenesis through interaction with BMP2. Cell Death Dis. 2021;12:147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
49.  Li Y, Fu G, Gong Y, Li B, Li W, Liu D, Yang X. BMP-2 promotes osteogenic differentiation of mesenchymal stem cells by enhancing mitochondrial activity. J Musculoskelet Neuronal Interact. 2022;22:123-131.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Zhu B, Xue F, Zhang C, Li G. LMCD1 promotes osteogenic differentiation of human bone marrow stem cells by regulating BMP signaling. Cell Death Dis. 2019;10:647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
51.  Abdallah BM. Carnosol induces the osteogenic differentiation of bone marrow-derived mesenchymal stem cells via activating BMP-signaling pathway. Korean J Physiol Pharmacol. 2021;25:197-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
52.  Wang H, Ni Z, Yang J, Li M, Liu L, Pan X, Xu L, Wang X, Fang S. IL-1β promotes osteogenic differentiation of mouse bone marrow mesenchymal stem cells via the BMP/Smad pathway within a certain concentration range. Exp Ther Med. 2020;20:3001-3008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
53.  Yang X, Mou D, Yu Q, Zhang J, Xiong Y, Zhang Z, Xing S. Nerve growth factor promotes osteogenic differentiation of MC3T3-E1 cells via BMP-2/Smads pathway. Ann Anat. 2022;239:151819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
54.  Pang Y, Liu L, Mu H, Priya Veeraraghavan V. Nobiletin promotes osteogenic differentiation of human osteoblastic cell line (MG-63) through activating the BMP-2/RUNX-2 signaling pathway. Saudi J Biol Sci. 2021;28:4916-4920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
55.  Feng C, Xiao L, Yu JC, Li DY, Tang TY, Liao W, Wang ZR, Lu AQ. Simvastatin promotes osteogenic differentiation of mesenchymal stem cells in rat model of osteoporosis through BMP-2/Smads signaling pathway. Eur Rev Med Pharmacol Sci. 2020;24:434-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
56.  Li M, Zhang C, Li X, Lv Z, Chen Y, Zhao J. Isoquercitrin promotes the osteogenic differentiation of osteoblasts and BMSCs via the RUNX2 or BMP pathway. Connect Tissue Res. 2019;60:189-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
57.  Chen L, Zhang M, Ding Y, Li M, Zhong J, Feng S. Fluoride induces hypomethylation of BMP2 and activates osteoblasts through the Wnt/β-catenin signaling pathway. Chem Biol Interact. 2022;356:109870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Wang J, Yang J, Tang Z, Yu Y, Chen H, Yu Q, Zhang D, Yan C. Curculigo orchioides polysaccharide COP70-1 stimulates osteogenic differentiation of MC3T3-E1 cells by activating the BMP and Wnt signaling pathways. Int J Biol Macromol. 2023;248:125879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
59.  Jiang H, Zhong J, Li W, Dong J, Xian CJ, Shen YK, Yao L, Wu Q, Wang L. Gentiopicroside promotes the osteogenesis of bone mesenchymal stem cells by modulation of β-catenin-BMP2 signalling pathway. J Cell Mol Med. 2021;25:10825-10836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
60.  Hu L, Cheng Z, Wu L, Luo L, Pan P, Li S, Jia Q, Yang N, Xu B. Histone methyltransferase SETDB1 promotes osteogenic differentiation in osteoporosis by activating OTX2-mediated BMP-Smad and Wnt/β-catenin pathways. Hum Cell. 2023;36:1373-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
61.  Wang YQ, Wang NX, Luo Y, Yu CY, Xiao JH. Ganoderal A effectively induces osteogenic differentiation of human amniotic mesenchymal stem cells via cross-talk between Wnt/β-catenin and BMP/SMAD signaling pathways. Biomed Pharmacother. 2020;123:109807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
62.  Wang M, Li J, Ye Y, He S, Song J. SHED-derived conditioned exosomes enhance the osteogenic differentiation of PDLSCs via Wnt and BMP signaling in vitro. Differentiation. 2020;111:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
63.  Chen XJ, Shen YS, He MC, Yang F, Yang P, Pang FX, He W, Cao YM, Wei QS. Polydatin promotes the osteogenic differentiation of human bone mesenchymal stem cells by activating the BMP2-Wnt/β-catenin signaling pathway. Biomed Pharmacother. 2019;112:108746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 87]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
64.  Sangadala S, Kim CH, Fernandes LM, Makkar P, Beck GR, Boden SD, Drissi H, Presciutti SM. Sclerostin small-molecule inhibitors promote osteogenesis by activating canonical Wnt and BMP pathways. Elife. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
65.  Pang X, Zhong Z, Jiang F, Yang J, Nie H. Juglans regia L. extract promotes osteogenesis of human bone marrow mesenchymal stem cells through BMP2/Smad/Runx2 and Wnt/β-catenin pathways. J Orthop Surg Res. 2022;17:88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
66.  Wang H, Hu Y, He F, Li L, Li PP, Deng Y, Li FS, Wu K, He BC. All-trans retinoic acid and COX-2 cross-talk to regulate BMP9-induced osteogenic differentiation via Wnt/β-catenin in mesenchymal stem cells. Biomed Pharmacother. 2019;118:109279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
67.  Pengjam Y, Syazwani N, Inchai J, Numit A, Yodthong T, Pitakpornpreecha T, Panichayupakaranant P. High water-soluble curcuminoids-rich extract regulates osteogenic differentiation of MC3T3-E1 cells: Involvement of Wnt/β-catenin and BMP signaling pathway. Chin Herb Med. 2021;13:534-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
68.  Zhao XL, Chen JJ, Zhang GN, Wang YC, Si SY, Chen LF, Wang Z. Small molecule T63 suppresses osteoporosis by modulating osteoblast differentiation via BMP and WNT signaling pathways. Sci Rep. 2017;7:10397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
69.  Karadeniz F, Oh JH, Lee JI, Seo Y, Kong CS. 3,5-dicaffeoylepi-quinic acid from Atriplex gmelinii enhances the osteoblast differentiation of bone marrow-derived human mesenchymal stromal cells via WnT/BMP signaling and suppresses adipocyte differentiation via AMPK activation. Phytomedicine. 2020;71:153225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
70.  Rong X, Kou Y, Zhang Y, Yang P, Tang R, Liu H, Li M. ED-71 Prevents Glucocorticoid-Induced Osteoporosis by Regulating Osteoblast Differentiation via Notch and Wnt/β-Catenin Pathways. Drug Des Devel Ther. 2022;16:3929-3946.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
71.  Zhou Y, Li J, Zhou K, Liao X, Zhou X, Shen K. The methylation of Notch1 promoter mediates the osteogenesis differentiation in human aortic valve interstitial cells through Wnt/β-catenin signaling. J Cell Physiol. 2019;234:20366-20376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
72.  Lee S, Remark LH, Josephson AM, Leclerc K, Lopez EM, Kirby DJ, Mehta D, Litwa HP, Wong MZ, Shin SY, Leucht P. Notch-Wnt signal crosstalk regulates proliferation and differentiation of osteoprogenitor cells during intramembranous bone healing. NPJ Regen Med. 2021;6:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
73.  Wagley Y, Chesi A, Acevedo PK, Lu S, Wells AD, Johnson ME, Grant SFA, Hankenson KD. Canonical Notch signaling is required for bone morphogenetic protein-mediated human osteoblast differentiation. Stem Cells. 2020;38:1332-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
74.  Seong CH, Chiba N, Kusuyama J, Subhan Amir M, Eiraku N, Yamashita S, Ohnishi T, Nakamura N, Matsuguchi T. Bone morphogenetic protein 9 (BMP9) directly induces Notch effector molecule Hes1 through the SMAD signaling pathway in osteoblasts. FEBS Lett. 2021;595:389-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
75.  Cao J, Wei Y, Lian J, Yang L, Zhang X, Xie J, Liu Q, Luo J, He B, Tang M. Notch signaling pathway promotes osteogenic differentiation of mesenchymal stem cells by enhancing BMP9/Smad signaling. Int J Mol Med. 2017;40:378-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
76.  Cui J, Zhang W, Huang E, Wang J, Liao J, Li R, Yu X, Zhao C, Zeng Z, Shu Y, Zhang R, Yan S, Lei J, Yang C, Wu K, Wu Y, Huang S, Ji X, Li A, Gong C, Yuan C, Zhang L, Liu W, Huang B, Feng Y, An L, Zhang B, Dai Z, Shen Y, Luo W, Wang X, Huang A, Luu HH, Reid RR, Wolf JM, Thinakaran G, Lee MJ, He TC. BMP9-induced osteoblastic differentiation requires functional Notch signaling in mesenchymal stem cells. Lab Invest. 2019;99:58-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
77.  Tian L, Xiao H, Li M, Wu X, Xie Y, Zhou J, Zhang X, Wang B. A novel Sprouty4-ERK1/2-Wnt/β-catenin regulatory loop in marrow stromal progenitor cells controls osteogenic and adipogenic differentiation. Metabolism. 2020;105:154189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
78.  Kuang Z, Bai J, Ni L, Hang K, Xu J, Ying L, Xue D, Pan Z. Withanolide B promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via ERK1/2 and Wnt/β-catenin signaling pathways. Int Immunopharmacol. 2020;88:106960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
79.  Arumugam B, Vairamani M, Partridge NC, Selvamurugan N. Characterization of Runx2 phosphorylation sites required for TGF-β1-mediated stimulation of matrix metalloproteinase-13 expression in osteoblastic cells. J Cell Physiol. 2018;233:1082-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
80.  Ni L, Kuang Z, Gong Z, Xue D, Zheng Q. Dihydroartemisinin Promotes the Osteogenesis of Human Mesenchymal Stem Cells via the ERK and Wnt/β-Catenin Signaling Pathways. Biomed Res Int. 2019;2019:3456719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
81.  Aimaiti A, Wahafu T, Keremu A, Yicheng L, Li C. Strontium Ameliorates Glucocorticoid Inhibition of Osteogenesis Via the ERK Signaling Pathway. Biol Trace Elem Res. 2020;197:591-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
82.  Heubel B, Nohe A. The Role of BMP Signaling in Osteoclast Regulation. J Dev Biol. 2021;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
83.  Daigang L, Jining Q, Jinlai L, Pengfei W, Chuan S, Liangku H, Ding T, Zhe S, Wei W, Zhong L, Kun Z. LPS-stimulated inflammation inhibits BMP-9-induced osteoblastic differentiation through crosstalk between BMP/MAPK and Smad signaling. Exp Cell Res. 2016;341:54-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]