In the skeletal system, blood vessels not only function as a conduit system for transporting gases, nutrients, metabolic waste, or cells but also provide multifunctional signal molecules regulating bone development, regeneration, and remodeling. Endothelial cells (ECs) in bone tissues, unlike in other organ tissues, are in direct contact with the pericytes of blood vessels, resulting in a closer connection with peripheral connective tissues. Close-contact ECs contribute to osteogenesis and osteoclastogenesis by secreting various cytokines in the paracrine or juxtacrine pathways. An increasing number of studies have revealed that extracellular vesicles (EVs) derived from ECs can directly regulate maturation process of osteoblasts and osteoclasts. The different pathways focus on targets at different distances, forming the basis of the intimate spatial and temporal link between bone tissue and blood vessels. Here, we provide a systematic review to elaborate on the function of ECs in bone biology and its underlying mechanisms based on three aspects: paracrine, EVs, and juxtacrine. This review proposes the possibility of a therapeutic strategy targeting blood vessels, as an adjuvant treatment for bone disorders.

Both bone resorption and bone formation are important aspects of having strong and well-functioning bones throughout life [1]. Among many direct and indirect factors influencing osteogenesis and bone resorption, blood vessels in bone tissue are a notable and significant factor for coordinating these two activities. In addition to taking on the role of transportation and substance exchange, blood vessels also participate in various processes of bone development, reconstruction, and repair. During both endochondral and intramembranous ossification, blood vessels are the pioneers, leading to subsequent osteogenesis [2,3]. In other words, angiogenesis is one of the essential elements in bone defect healing [4,5].

The interaction between cells can be divided into three types: paracrine, extracellular vesicles (EVs), and juxtacrine. Paracrine activity involves the secretion of proteins/peptides and hormones, which are usually diluted during diffusion [6]. The different concentrations of paracrine factors affect different responses in target cells. EVs comprise exosomes, microvesicles, and apoptosomes containing growth factors, proteins, bioactive lipids, mRNA/miRNA, and DNA, and are perceived as mediators for intercellular communication even across distant tissues [7]. Juxtacrine interaction relies on the interaction between transmembrane ligands on one cell and receptors on the cell membrane of a neighboring cell. In bone tissue, most of the blood vessels are capillaries that make direct contact with pericytes, without the basement membrane between them; this makes the interaction between endothelial cells (ECs) and their pericytes tighter [8]. As a result, ECs in bone tissue have abundant opportunities to communicate with surrounding cells through juxtacrine, paracrine, and EVs. This review is based on these three aspects and describes the effects of microvascular ECs on surrounding cells in bone tissue, which finally affect osteogenic metabolism and bone resorption. We believe that clarifying the molecular mechanisms that underlie ECs participating in bone biology will not only further update our recognition of the pathogenesis of bone disorders but also afford new potential targets for treating bone diseases.

Bone formation and resorption are complicated processes. Hence, it is not straightforward to elucidate the function of ECs at the stages of bone development, remodeling, and regeneration. Both osteogenesis and osteoclastogenesis are closely coupled with angiogenesis. However, in the past, the coupling between angiogenesis and osteogenesis has attracted more attention. Numerous studies have evaluated the various functions and regulations of blood vessels in osteogenesis, which provide an opportunity to understand their role in bone biology more comprehensively. During endochondral bone formation, new blood vessels grow and transport osteoclast and osteoblast progenitors into the center of the future bone, as a first step in the process of bone formation [2]. Although intramembranous osteogenesis is poorly understood compared with endochondral bone formation, current studies conclude that small-bore capillaries invade into the initial ossification site at the initial stage of intramembranous osteogenesis and endochondral bone formation [3]. Furthermore, osteodistraction models have also shown that angiogenesis predominantly occurs before osteogenesis [9]. Blood vessels can lead to longitudinal growth of long bone by regulating cartilage resorption [10]. Numerous studies have shown the importance of angiogenesis in the process of bone, which is accompanied by the various signaling factors in the hematoma or blood clot [4,5,11]. Additionally, confocal microscopy revealed that mature osteoblasts gather around blood vessels that invade into the cartilaginous callus tissues during fracture healing [2]. Furthermore, evidence indicated the presence of signaling factor cross-talk from ECs to osteoclast lineage cells to promote migration from the circulatory system to bone tissue and osteoclastic differentiation. After recognition by ECs, monocytes can pass through endothelial gaps into bone tissue.

Based on recent reports, bone microvessels are divided into three subtypes, including type H, type L, and type E [8,12]. Based on the protein level of CD31 and endomucin (Emcn) in ECs, capillary vessels in bone tissue are defined as type H (CD31high Emcnhigh) and type L (CD31low Emcnlow) blood vessels [8]. The former is known to play a vital role in bone development, inducing ossification. Another study proposed that type E blood vessels appearing in the embryonic and early postnatal bone, as the third EC subtype in bone tissue, supported osteoblast lineage cells more strongly than type H blood vessels and could transform into other EC subpopulations [12].

Additionally, another kind of cell—mesenchymal stem cells (MSCs)—that is indicated to be the same type of cell as pericytes [13] can pass through endothelial gaps. MSCs dwell in the perivascular niche of almost all mature tissues and will mobilize and migrate into damaged tissues to promote tissue healing [13–15]. Migration of MSCs from other tissues into bone is critical for bone repair [16].

In summary, blood vessels in bone tissue perform multiple functions, mostly because of EC-derived signaling molecules. This review elaborates the role of these molecules on bone biology including paracrine, juxtacrine, and secreted protein or other substances in EVs.

A number of previous studies have shown that ECs secrete several signaling molecules by paracrine interaction, such as platelet-derived growth factor (PDGF)-BB, vascular endothelial growth factor (VEGF), bone morphogenetic protein (BMP) 2, matrix Gla protein (MGP), receptor activator of nuclear factor-κB ligand (RANKL), and osteoprotegerin (OPG), which play important and critical roles in osteogenesis and bone resorption (Figure 1). The concentration of these cytokines are reduced as they move away from the blood vessels, resulting in limited effect. As reported by Francis and Palsson, the maximal distance a solitary cell in vitro can effectively communicate is approximately 250 μm by soluble cyto- and chemokines [17]. This distance can be further regulated if the cytokines bind to the extracellular matrix (ECM). Another study pointed out that this distance also depended on the strength of the enhanced-release time and rate [18]. Meanwhile, the degradation of released molecules would further limit the distance [19]. These factors determine the paracrine molecules secreted by ECs focus on the target cells close to blood vessels.

The effects of EC-secreted paracrine factors on osteogenesis and osteoclastogenesis

Figure 1
The effects of EC-secreted paracrine factors on osteogenesis and osteoclastogenesis

In bone tissue, PDGF can recruit pericytes/MSCs, promote their growth, and inhibit their osteogenesis, and antagonize VEGF. BMP2 can promote osteogenesis, antagonize MGP, and Noggin. Besides, BMP2 can attract monocytes to adhere to ECs by antagonizing Noggin. VEGF can induce osteoclasts to migrate and affect osteoblastic differentiation. Furthermore, RANKL can induce osteoclastic differentiation by antagonizing OPG. As for inflammatory cytokines, CCLs and CXCLs can induce monocytes to migrate into bone tissue and differentiate into osteoclasts. MMPs can promote osteoclastic differentiation. Meanwhile, BMP2, VEGF, and CCL2 can prevent osteocyte apoptosis.

Figure 1
The effects of EC-secreted paracrine factors on osteogenesis and osteoclastogenesis

In bone tissue, PDGF can recruit pericytes/MSCs, promote their growth, and inhibit their osteogenesis, and antagonize VEGF. BMP2 can promote osteogenesis, antagonize MGP, and Noggin. Besides, BMP2 can attract monocytes to adhere to ECs by antagonizing Noggin. VEGF can induce osteoclasts to migrate and affect osteoblastic differentiation. Furthermore, RANKL can induce osteoclastic differentiation by antagonizing OPG. As for inflammatory cytokines, CCLs and CXCLs can induce monocytes to migrate into bone tissue and differentiate into osteoclasts. MMPs can promote osteoclastic differentiation. Meanwhile, BMP2, VEGF, and CCL2 can prevent osteocyte apoptosis.

Close modal

Several studies have shown a comprehensive perspective of PDGF in bone tissue. A recent study estimated that PDGF-BB in bone marrow was predominantly from TRAP+ cells (72.6%), while 12.6% were from ECs and 14.8% from other bone marrow cells [20]. Previous studies have shown that that PDGF-BB from macrophage-lineage TRAP+ cells could recruit Nestin+ and LepR+ periosteum-derived cells to the periosteal surface for periosteal bone formation [21]. Meanwhile, EC-derived PDGF-BB could recruit PDGFR-β-expressing pericyte progenitors into the new bone area [22,23]. The attached pericytes could stabilize the structure of newly formed blood vessels [24,25]. On the one hand, PDGF-BB can induce MSCs proliferation through PI3K signaling, while on the other, PDGF-BB can regulate the differentiation of MSCs via Erk signaling [26]. Another in vitro experiment also revealed that PDGF-BB can promote the proliferation of pericytes [27]. Besides, other studies also pointed out that PDGF-BB/PDGFR-β can increase the migratory response and proliferative capacity of MSCs but strongly inhibit osteogenic differentiation of MSCs [28–32]. After secretion from the ECs, PDGF binds to ECM via heparan sulfate proteoglycans and is confined to specific sites [33]. The interaction between PDGF and ECM promotes PDGF to retain itself close to ECs and form a chemoattracting gradient [34]. Simultaneously, it attenuates the effects of secretion rate and degradation of PDGF to the chemoattracting gradient. To sum up, the chemoattracting gradient of PDGF contributes to MSCs aggregation and benefits MSCs to maintain self-renewal and proliferation. When MSCs are far away from the blood vessels, the inhibition of osteogenesis from ECs-derived PDGF-BB will be weakened. Such a phenomenon may also occur in other secretory factors.

VEGF in bone tissue is mainly produced by hypertrophic chondrocytes, and some VEGF is secreted by newly formed ECs [35]. Quiescent ECs in vitro did not express VEGF [35]. However, stimulated by FGF2, quiescent ECs can be activated to form new capillaries and express both VEGF mRNA and protein. Additionally, hypoxia can also stimulate ECs to secrete VEGF [36], which occurs in fractured hematomas. Additionally, VEGF can inhibit the migration and proliferation of MSCs through PDGF receptors [37]. This is in line with a study wherein VEGF can antagonize PDGF-stimulated pericyte recruitment to regenerate blood vessels during angiogenesis [38]. In other words, in the progress of neovascularization, VEGF reduces vascular pericyte coverage and causes vessel destabilization [38]. Based on the retinal angiogenesis model, it was found that the signal across the angiogenic front was up-regulated with the loss of pericyte coverage [39]. This could likely be because reduction in vascular pericyte coverage caused by VEGF benefits blood vessels to sprout more easily during angiogenesis. Apart from strong regulation of angiogenesis, VEGF also plays an influential role in recruiting monocytes and osteoclasts, as well as regulating osteoclast differentiation [40–44]. VEGF can also regulate the fate of cartilage and inhibition of VEGF benefits cartilage fates [45,46], which play a crucial role in bone development when blood vessels invade the cartilage. In terms of osteogenesis, a previous study showed that VEGF could promote bone mesenchymal stromal cells to proliferate and show osteogenic differentiation [47]. This study showed that appropriate VEGF could promote osteogenesis, while a high dose of VEGF could inhibit osteogenesis [48]. The deletion of VEGF receptor 2 in osteoblastic lineage cells increased the maturation of osteoblast and mineralization in intramembranous ossification-mediated bone repair. However, an in vitro experiment showed contrary effects of VEGFR2 in that its activation promoted the survival of osteocytes [49]. Taken together, VEGF, as a paracrine factor, can work on a variety of cells and play a complicated role at an early stage of bone development. EC-derived VEGF can affect the pericytes surrounding ECs to a certain degree, especially during the period of angiogenesis. As for the effect of EC-derived VEGF on the whole bone tissue, a previous study shows that Vegfafl/fl VE-cadherin-Cre mice do not show significant differences in bone healing of a tibial monocortical defect model, which contrasted with findings from the littermate controls [48].

As a member of the TGF-β superfamily, BMPs can stimulate MSCs and osteogenic lineage cells to undergo osteoblast differentiation through the canonical BMP-SMAD pathway [50], which is vital in the earliest step of fracture healing [51]. The BMP-SMAD pathway can also prevent osteocytes, the terminal stage of osteogenic differentiation, from apoptosis [52]. Among the BMP family, BMP2 and BMP4 play a role in the interaction between ECs and pericytes. Under pathological stimuli such as the inflammatory microenvironment, the expression of BMP2 and BMP4 will have a strong response in ECs [53,54]. Nonetheless, a previous study showed that targeted deletion of Bmp2 in vascular ECs did not impact fracture healing in any way [55]. Recent immunohistological studies of BMP2 expression showed that BMP2 was most strongly expressed in periosteal cells and hypertrophic chondrocytes [56]. BMP2 can also be released by pre-hypertrophic chondrocytes, osteoblasts, and osteocytes during the progression of endochondral healing [56,57]. Taken together, we can conclude that EC-derived BMP2 plays an insignificant role in fracture healing. With respect to bone development, EC-derived BMP2 has little effect on postnatal skeletal growth, structure, and strength [58]. BMP4 is a weaker stimulator of osteogenesis than BMP2 [59], and it is not required for limb skeletogenesis, bone formation, and fracture healing [60]. On the other hand, ECs can also secrete BMP antagonists such as MGP, follistatin, and Noggin through exocytosis. In healthy bone tissue, EC-derived MGP is supposed to interact with BMP2 to inhibit ossification [61], which regulates the differentiation of pericytes around ECs. Another article showed that Noggin was the main BMP antagonist secreted by ECs in bone tissue, regulating the differentiation of pericytes and thereby osteogenesis and promoting chondrocyte maturation [62]. If the balance of BMPs was broken, vascular calcification or tibial dyschondroplasia would occur [61,63]. Interestingly, BMP2 also plays a part in the adhesion of monocytes to ECs [64], ultimately affecting osteoclast formation. At the same time, Noggin or other BMP antagonist can interfere with monocyte migration by inhibition of BMP2 signaling [64], thereby decreasing the number of osteoclasts.

During osteoclast differentiation, RANKL and macrophage-colony stimulating factor (M-CSF) both played important roles. The latter could not be secreted by vascular ECs, rather only lymphatic ECs [65]. Patricia et al. first revealed that microvascular ECs can express both mRNAs of RANKL and OPG [66]. In a later study, it was shown that under pathological conditions, ECs stimulated by TGF-β could increase the expression of RANKL to promote osteoclastogenesis to benefit bone remodeling [67]. EC-derived RANKL plays a role in the differentiation of osteoclasts, which can be demonstrated by a phenomenon that the absence of EC-derived RANKL reduced the number of osteoclasts around ECs along with the total number of osteoclasts [10]. As for OPG which can bind to RANKL to block the interaction of the latter with RANK on the osteoclast membrane, it was noted that OPG could also be synthesized by vascular ECs [68]. Malyankar et al. found that at least some EC-derived OPG were associated with the surface of ECs such as a juxtacrine factor; normally, OPG does not contain any transmembrane domain [68]. However, this study did not prove whether the OPG binding to the surface of ECs still had the capacity to interact with RANKL. Another in vitro study showed that ECs from various tissues could secrete OPG to inhibit the differentiation of osteoclasts [69]. Lekesiz et al. revealed that, in ECs, the expression level of OPG was negatively correlated with RANKL [70]. Taken together, it may be concluded that vascular ECs normally inhibit osteoclast differentiation, which is consistent with a phenomenon that, in a healthy state, osteoclasts are typically found around the trabecular bone rather than blood vessels. Osteocytes strongly express OPG and are the major source of RANKL [71,72]. Besides, Kehmia et al. pointed out that OPG was mainly derived from B cells in the bone microenvironment [73]. Those two pieces of evidence further prove the limitation of RANKL and OPG deriving form ECs.

In pathological conditions, ECs are an important source of inflammatory cytokines. After undergoing ionizing radiation, ECs overexpressed CX3CL1 that attracted circulating CX3CR1+/CD11b+ cells and induced the latter to undergo osteoclast differentiation [74]. Additionally, CX3CL1 stimulated ECs to secrete other inflammatory chemokines like CXCL2 and CXCL12 in the form of autocrine signaling. A recent study showed that CXCL10, CCL2, and CCL5 have similar osteoclastogenic effects, with the latter especially possessing the largest chemotactic effect on osteoclast progenitors [75]. In another study, under hypoxic or ischemic conditions, cardiac microvascular ECs significantly increased the production of CXCL10 [76]. There is a study comparing the chemokine secretion ability of ECs from different vascular beds [77]. This study determined that CXCL8 and CCL2 could be constitutively produced by human saphenous vein endothelium, lung and dermal microvascular ECs, human umbilical vein ECs (HUVECs), and a bone marrow EC (BMEC) line. Besides, CCL5 and CXCL10 were secreted only after those cells were stimulated by tumor necrosis factor-α (TNF-α) or interferon-γ (IFN-γ). A related phenomenon showed that human BMECs under the stimulation of parathyroid hormone-related protein can secrete CCL2 to promote the differentiation of osteoclast in vitro [78]. Interestingly, Kitase et al. reported that CCL2, at a low dose, can prevent apoptosis of osteocytes [79]. Additionally, an immunostaining result demonstrated that HUVECs stimulated by lipopolysaccharide or atorvastatin could secrete CCL19 and CCL21 [80]. In this study, it was also proven that CCL19 and CCL21 could induce monocytes to adhere and migrate to HUVECs. Furthermore, another study demonstrated that CCL19 and CCL21 could promote osteoclast’s capacity of resorption and migration [81]. Taken together, it can be stated that ECs can secrete some of the chemokines in a healthy state and mostly in an inflammatory state, inducing monocytes to migrate into bone tissue and differentiate into osteoclasts.

MMPs are a type of collagenases capable of regulating the progression of the embryo and physiological remodeling tissue as well as disease development [82]. A recent study showed that ECs from bone tissue could produce more MMPs such as MMP9 and MMP14 than those produced from osteoclasts. In the present study, the absence of EC-derived MMP9 resulted in the reduction in growth plate size [10]. This supports ECs-derived MMP9 as contributing to the process of blood vessels invading and degrading cartilage tissue during bone development. Meanwhile, the degraded cartilage matrix released large amounts of VEGF, inducing angiogenesis. Hence, MMPs can recruit osteoclasts in bone development [83]. Engsig et al. demonstrated that MMP inhibitors could completely prevent TRAP+ cells’ migration. Subsequently, another study confirmed that the deficiency of gelatinase B/MMP9 in mice caused delayed osteoclast recruitment, which affected early bone development [44]. As for MMP14, which is also called membrane-type 1 MMP, it can maintain osteoblasts and osteocytes survival through activating TGF-β [84]. However, another study revealed that MMP14 increased soluble RANKL production, thereby stimulating osteoclast formation and bone resorption [85].

Besides size, EV populations can be categorized by additional qualifiers of identity-differential biogenesis, including exosomes (30–150 nm in diameter), microvesicles (50–1000 nm in diameter), and apoptosomes (50–5000 nm in diameter) [86]. Compared with the paracrine pathway, EVs can protect their contents such as sequestered proteins and mRNA from degradation and enable cell communication across tissues. Studies about EC-derived EVs are insufficient. The studies reporting the effect of ECs-derived EVs on bone tissue are even fewer. Alique et al. found that the number of total microvesicles secreted from senescent HUVECs was greater than that from young cells [87]. In senescent ECs, the secretion of EVs containing miR-31 is up-regulated [88]. Later, it was shown that miR-31 could regulate osteogenesis by targeting Osx, Runx2, and SATB2 [89,90]. Then, a recent study further reported that EC-derived EVs containing miR-31 could be taken up by bone MSCs (BMSCs), which inhibits the differentiation of osteogenesis through down-regulating the expression of FZD3, a Wnt5A receptor [91]. EVs contain a variety of substances. A previous study showed that under TNF-α stimulation, HUVECs could release more endothelial microparticles, one kind of EVs [92]; interestingly, the endothelial microparticles contained significant BMP2 which could promote osteogenic differentiation and was normally secreted by exocytosis [93]. The association between EC-derived EVs and osteoclasts was revealed gradually. Vítková et al. confirmed that EC-derived EVs can directly bind to monocytes, resulting in increasing transendothelial migration of monocytes [94]. Another study showed that EVs from quiescent ECs could regulate the inflammatory responses of monocytes; more importantly, these EVs inhibited monocyte/macrophage activation by transferring miR-10a into monocytic cells and targeting some components of the NF-κB pathway, such as IRAK4 [95]. On the other hand, Zhan et al. showed that under the induction of oxidative low-density lipoprotein and homocysteine, ECs effectively increased the release of EVs which contained HSP70; and HSP70 could activate monocytes and induce them to adhere to ECs [96]. A recent study further confirmed the relation between ECs and osteoclasts through EVs [97]. This study showed that bone marrow-derived macrophages could internalize EC-exosomes that contained a large amount of miR-155, which then altered their morphology; and miR-155 could suppress osteoclastic differentiation. Additionally, it was found that EC-exosomes showed more effective bone targeting than exosomes derived from osteoblasts or BMSCs, which was confirmed with biophotonic imaging after injecting intravenous Dil-labeled exosomes. Another study revealed that ECs might direct the function of heterotypic cell types, systemically and specifically, via the contents contained within their EVs (Figure 2) [98].

EC-derived EVs with different contents will have different effects in bone tissues

Figure 2
EC-derived EVs with different contents will have different effects in bone tissues

EVs containing miR-31 can inhibit the osteogenesis of BMSCs. And EVs within HSP70 can promote the adhesion of monocytes. EVs within miR-10a will inhibit monocyte/macrophage activation. Besides, EVs containing miR-155 can suppress osteoclastic differentiation.

Figure 2
EC-derived EVs with different contents will have different effects in bone tissues

EVs containing miR-31 can inhibit the osteogenesis of BMSCs. And EVs within HSP70 can promote the adhesion of monocytes. EVs within miR-10a will inhibit monocyte/macrophage activation. Besides, EVs containing miR-155 can suppress osteoclastic differentiation.

Close modal

Juxtacrine interaction is a cell–cell signaling mediated by the direct interaction between adjacent cells and plays a key role in cell growth, proliferation, and metabolism. Among various juxtacrine factors, the most studied signaling pathways are the ephrin-Eph and Notch. Some of them are involved in osteogenesis and osteoclastogenesis. As early as 1995, a juxtacrine model was reported in a study that bovine bone ECs had a certain adhesion to preosteoclastic cells by direct contact in vitro [99].

Direct and indirect evidence from extensive studies showed the potential of pericytes to differentiate into osteoblasts, chondrocytes, adipocytes, smooth muscle cells, fibroblasts, and macrophages [100–105]. A recent study reported the role of direct interaction between ECs and pericytes in maintaining differentiation capacity [106]. This study showed that ECs promoted human BMSCs (HBMSCs) to maintain its self-renewal and stemness by the juxtacrine pathway—ephrin-Eph signaling (Figure 3). As signaling factors of bidirectional juxtacrine signaling, ephrins and Ephs can be ligands and receptors for each other, giving rise to bidirectional signal transduction and have been researched extensively. In the last century, studies have shown the interaction between ECs and surrounding mesenchymal cells through ephrin-Eph signaling [107]. In bone tissue, ephrins and Ephs are widely expressed in various cells (Table 1), and ephrinB2 is known to bind to EphB1-4, EphB6, and EphA4, while EphB4 can only bind to ephrinB2. Through the activation of bidirectional signaling pathways, the ephrin-Eph signaling pathway produces diverse effects in tissues. For example, the EphB4-ephrinB2 signaling pathway has been shown to repress osteoclast precursors from maturing through inhibiting c-Fos-NFATc1 cascade [108], which is consistent with another study that showed inhibition of ephrin-Eph signaling in osteoclastic differentiation [109]. On the contrary, ephrinB2 enhanced osteogenic differentiation through EphB4 of osteoblasts, which is a forward signaling [108,110]. Osteocytes with the absence of ephrinB2 increased more autophagosomes both in vivo and in vitro [111]. Besides, the stimulation of ephrinB2 affected the production of collagen type II in osteoarthritic chondrocytes, indicating its role in controlling chondrocyte differentiation [112]. Given all that, a reasonable conclusion may be reached in that arterial or venous ECs in bone tissue may interact with osteocytes, osteoblasts, and chondrocytes through ephrin-Eph signaling. However, relevant data are lacking.

ECs interact with surrounding cells via juxtacrine factors on the membrane

Figure 3
ECs interact with surrounding cells via juxtacrine factors on the membrane

ECs-derived Dll4 can control the differentiation of monocyte/macrophage. In bone tissue, ECs promote HBMSCs to maintain self-renewal and proliferation by ephrinB2-EphB4 signaling.

Figure 3
ECs interact with surrounding cells via juxtacrine factors on the membrane

ECs-derived Dll4 can control the differentiation of monocyte/macrophage. In bone tissue, ECs promote HBMSCs to maintain self-renewal and proliferation by ephrinB2-EphB4 signaling.

Close modal
Table 1
Cell distribution of components of ephrin-Eph signaling
CellLigandReceptorReferences
EC ephrinB2 EphB4 [106,107,159,160
BMSC ephrinB2  [106
Osteoblast ephrinB2 EphB2, -B4 [108,110
Osteocyte ephrinB2 EphB4 [111,161
Osteoclast precursor and osteoclast ephrinB2  [108,109
Chondrocyte ephrinB2 EphB4 [109
CellLigandReceptorReferences
EC ephrinB2 EphB4 [106,107,159,160
BMSC ephrinB2  [106
Osteoblast ephrinB2 EphB2, -B4 [108,110
Osteocyte ephrinB2 EphB4 [111,161
Osteoclast precursor and osteoclast ephrinB2  [108,109
Chondrocyte ephrinB2 EphB4 [109

As a vital parameter of the mammalian vascular system, the Notch pathway works by direct contact between two adjacent cells. In bone tissue, Notch ligands and receptors exist in various cells (Table 2). The Notch pathway can regulate MSC behavior as well as embryological bone formation [113–117]. Deleting Notch proteins in osteoclast precursors can enhance their osteoclastic differentiation and bone resorption [118]. As for mesenchymal progenitor cells, Notch2 can inhibit their differentiation that did not bias lineage allocation [115]. An in vitro experiment showed that inhibition of Notch signaling significantly decreased the expression of cyclin D1 in human MSCs, which indicated that the proliferation of human MSC was weakened [119]. A relevant conclusion was drawn in another research that the activation of Notch promoted the expansion of mesenchymal progenitor cells [120]. Further, this research revealed that Notch1 and -2 could control the fate of MSCs and their differentiation towards osteoblastic lineage cells, while Notch2 could suppress osteoblast differentiation [120]. However, another study showed Jagged1 could induce BMSCs to osteoblastic differentiation through Notch2 [121]. Thus, some studies support that Notch promotes osteogenesis [117,122,123], while others do not [118,120,124,125]. Besides, an in vitro experiment showed that mice with conditional deletion of Rbpj (a downstream molecule of Notch) in osteoblastic cells did not have an appreciable defect in osteoprogenitor cells [62]. These controversial results implied the complex role of the Notch pathway in regulating osteoblastic differentiation. A series of studies have shown that Jagged1 was up-regulated highly during the reparation of bone fracture and could induce HBMSCs to differentiate into osteoblasts [121,126,127]. In another study, while both Notch ligands, Jagged1 and Dll4, had opposite effects on angiogenesis in the skin [128], the latter could block endothelial activation and reduce angiogenesis through Notch1 signaling, and the former could block Dll4-Notch1 signaling to allow endothelial activation by VEGF and endothelial layer growth. Taken together, the complex role of the Notch pathway in bone tissue may be considered from the perspective of diverse Notch ligands with opposite effects. Notch1 has been shown to play a more important role than Notch2 and Notch4 in vascular angiogenesis [62,129–131]. The pleiotropy effects of Notch signaling was also noted in another phenomenon Notch1 activated by Dll4 from adjacent ECs promoted EC proliferation, and vessel growth in bone tissue and indirectly promoted osteogenesis through secretion of Noggin [62], which was contrary to the phenomenon discovered in other organs and tumors that Notch in ECs represses vascular angiogenesis [128,132,133]. Given the existence of an interaction between ECs and other cells, it is likely that EC-derived Notch ligands may act upon Notch proteins on the cell membrane around blood vessels in bone tissue. A previously conducted review suggested the possibility of this hypothesis [134]. Additionally, a study showed that EC-derived Dll1 can transform Ly6Chi monocyte into Ly6Clo monocyte by Notch2, both in vivo and in vitro [135], which indicated that ECs can regulate the fate of monocytes through the Notch pathway and also likely act on the process of osteoclastic differentiation through the Notch pathway. Furthermore, some studies showed that EC-derived Dll4 controlled the differentiation of monocyte/macrophage in the heart (Figure 3) [136,137].

Table 2
Cell distribution of components of Notch signaling
CellLigandReceptorReferences
EC Dll1, -4 and Jagged1 Notch1, -2, -4 [62,131,135,162
Osteoclast precursor  Notch1, -2, -3 [118
BMSC and osteoblastic lineage cell  Notch1, -2 [120
Mesenchymal progenitor cell Dll1, -4 and Jagged1 Notch1, -2 Notch1, -2 [115,120
Chondrocyte  Notch2 [120
CellLigandReceptorReferences
EC Dll1, -4 and Jagged1 Notch1, -2, -4 [62,131,135,162
Osteoclast precursor  Notch1, -2, -3 [118
BMSC and osteoblastic lineage cell  Notch1, -2 [120
Mesenchymal progenitor cell Dll1, -4 and Jagged1 Notch1, -2 Notch1, -2 [115,120
Chondrocyte  Notch2 [120

There are other juxtacrine signaling pathways between ECs and the cells around the blood vessels, such as the semaphorin signaling system [138]. A previous in vitro study showed that HUVECs can express Plexin-B1, which is a membrane receptor and can bind with semaphorin 4D, a membrane protein similar to the αVβ3 integrin heterodimer [27,139]. Semaphorin 4D is found expressed in premature and mature osteoclasts [140]. Under the stimulation of semaphorin 4D, ECs can make PDGF-B, restrain stem cells from transforming into pericytes [27].

In addition to the above signal pathways, some researchers have revealed other forms of contact between ECs and their surrounding cells. An in vitro experiment showed that direct contact of HBMSCs with HUVECs increased the expression of type I collagen and alkaline phosphatase activity [141]. In this study, both HBMSCs and HUVECs could express connexin43, which constituted a gap junctional channel and benefited cell-to-cell communication, as confirmed by immunocytochemistry. Interestingly, confocal microscopy has revealed the mitochondria transferred from MSCs to rat lung microvascular ECs through juxtacrine interaction [142], consistent with previous reports [143,144].

Osteoporosis has become a major public health issue with a high risk of fractures, especially for postmenopausal women. Histological results showed that primary osteoporosis is characterized by decreased numbers of sinuses and arterial capillaries in bone tissue [145]. The same phenomenon occurs in postmenopausal osteoporosis [146]. Later, in the research about age-related bone loss, it was found that type L capillaries did not change significantly, while the number of type H vessels decreased in old mice [8]. Bisphosphonate is a commonly used medicine to treat osteoporosis, which is considered to play a function in osteogenesis. Recently and interestingly, it was found that bisphosphonate could enhance blood flow and angiogenesis in old mice [147]. This study further revealed that treatment with bisphosphonate in aged mice activated the Notch pathway in ECs and increased type H capillaries. Bisphosphonate’s effect on osteoporosis is partly achieved by its action on blood vessels. Another study also showed that the administration of recombinant SLIT3 could be a vascular-targeted medicine to treat osteoporosis, which promoted angiogenesis of type H vessels [148].

ECM remodeling is an important process of bone fracture healing. A previous experiment has shown that bone healing was delayed in Mmp9−/− mice [149]. MMP9 is mainly produced by type H blood vessels [10]. Xu et al. found recombinant SLIT3 can also play a function in fracture healing by expanding type H vessels [148]. Therefore, the mechanism may be that ECs secrete MMPs to degrade the ECM and promote callus mineralization.

Radiation osteomyelitis usually occurs after head and neck radiation therapy, as a common and serious complication [150]. In an irradiated murine model, ECs overexpressed CX3CL1, resulting in recruiting circulating osteoclast precursor cells to the irradiated bone, which induced osteoclastic differentiation and bone loss [74]. Subsequent experiments have confirmed that knockout of CX3CL1 or using its antagonist could prevent osteoclastogenesis and ameliorate bone resorption. This finding provides a theoretical basis for targeted vascular therapy of bone resorption, beyond just radiation osteomyelitis.

Lung, prostate, and breast cancer, as the most common human cancers, are prone to bone metastasis, in which damage to bones is not negligible [151,152]. During bone metastasis of prostate cancer, osteoblasts and BMECs stimulated by prostate-derived parathyroid hormone-related protein increase the secretion of CCL2 that could enhance osteoclastic activity and prostate cancer growth in bone [78,153]. Meanwhile, anti-CCL2 treatment in vivo effectively suppressed the process. It has been reported that ECs played a major role in the secretion of CCL2 during bone metastasis of prostate cancer [154]. Therefore, it is feasible to target the secretion of inflammatory factors from blood vessels for osteoclastic diseases and bone metastasis, which may also apply to other diseases causing systemic inflammation.

In the treatment of bone tissue diseases, just as with osteoporosis and bone fractures, most attention was paid to promoting osteogenesis in the past. However, during such treatment, the drug targeting blood vessels was not thoroughly studied and widely used in the past. In the process of drug ingestion, blood vessels are the first stop for drugs to enter bone tissue. On the other hand, in addition to transporting nutrients, blood vessels, as mentioned above, also have multiple other functions that can affect bone tissue. Therefore, we put forward that a potential strategy to treat bone disease is using drugs targeting blood vessels in bone tissue.

During the transportation of drugs in vivo, EV can become an ideal carrier, which is an effective communication medium between cells even from different organs, both locally and remotely [155]. EC-derived EVs can target bone tissue [97]. A similar phenomenon was also found wherein EVs derived from osteoclasts could also target bone tissue [156]. The mechanism behind is worth studying in particular and has significant applications in the field of targeted therapy, which improves the curative effects of drugs in bone tissue and reduces the side effects of the drugs in other organs and tissues. Compared with the EVs derived from other cells, research about EVs released from ECs in bone tissue still has broad prospects and the related mechanisms are not completely understood, which is worthy of further in-depth exploration.

Most information pertaining to the role of ECs in bone tissue comes from research focusing on the coupling of angiogenesis and osteogenesis. However, our description of the ECs’ role in osteoclastogenesis is inadequate and incomplete. Therapeutic strategies that target inflammatory factors secreted from blood vessels such as CCL2 and CX3CL1 in some animal models of bone disease have proven successful and effective. The potential mechanism of regulation of osteoclastogenesis by ECs remains to be explored and studied. Similar to this, there are likely to be mechanisms that have not yet been elucidated that address the process of MSCs and osteoclast progenitors in peripheral blood entering into bone tissue, especially the mechanism of juxtacrine pathways.

The role of ECs in bone biology is still insufficiently clarified. For instance, osteocytes comprising approximately 95% of total bone cells have shown the role of osteocytes in hematopoiesis, myeloid differentiation, and lymphopoiesis [157,158]. Nevertheless, the ECs’ effects on osteocytes need further study. Additionally, there is another direction worth studying that the stimulation of a specific flow pattern generated by endothelial buds is important for bone tissue which is sensitive to mechanical stimulation [147]. Besides, ECs are not the main source of various cytokines in the bone matrix, which limits the influence of ECs on bone tissue and is contrary to the coupling between angiogenesis and osteogenesis. Additionally, research about these three EC subtypes is not comprehensive enough. It would be very interesting to further study the interconversion of these three EC subtypes.

Many of the above experimental results were found in mice, which means that they may likely not exist in an identical manner in the human body. Despite some of these limitations, we have the opportunity to further understand the detailed mechanisms by which ECs affect bone tissue and thereby target ECs as a potential therapeutic strategy for treating numerous bone diseases.

The data are available from the corresponding author upon reasonable request.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the National Natural Science Foundation of China for Distinguished Young Scholars (to L.C.).

Ying Yin and Qingming Tang wrote the original draft. Lili Chen and Li Hu revising and editing the manuscript. Mengru Xie contributed to the figures. All authors have read and agreed to the published version of the manuscript.

BMEC

bone marrow endothelial cell

BMP

bone morphogenetic protein

BMSC

bone mesenchymal stem cell

CCL

C-C motif chemokine ligand

CXCL

C-X-C motif chemokine ligand

EC

endothelial cell

ECM

extracellular matrix

Emcn

endomucin

EV

extracellular vesicle

FGF

fibroblast growth factor

HBMSC

human bone mesenchymal stem cell

HUVEC

human umbilical vein EC

MGP

matrix Gla protein

MSC

mesenchymal stem cell

OPG

osteoprotegerin

PDGF

platelet-derived growth factor

RANKL

receptor activator of nuclear factor-κB ligand

TNF-α

tumor necrosis factor-α

VEGF

vascular endothelial growth factor

1.
Zhu
L.
,
Tang
Y.
,
Li
X.Y.
,
Keller
E.T.
,
Yang
J.
,
Cho
J.S.
et al.
(
2020
)
Osteoclast-mediated bone resorption is controlled by a compensatory network of secreted and membrane-tethered metalloproteinases
.
Sci. Transl. Med.
12
,
eaaw6143
2.
Maes
C.
,
Kobayashi
T.
,
Selig
M.K.
,
Torrekens
S.
,
Roth
S.I.
,
Mackem
S.
et al.
(
2010
)
Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels
.
Dev. Cell
19
,
329
344
[PubMed]
3.
Percival
C.J.
and
Richtsmeier
J.T.
(
2013
)
Angiogenesis and intramembranous osteogenesis
.
Dev. Dyn.
242
,
909
922
[PubMed]
4.
Saran
U.
,
Gemini Piperni
S.
and
Chatterjee
S.
(
2014
)
Role of angiogenesis in bone repair
.
Arch. Biochem. Biophys.
561
,
109
117
[PubMed]
5.
Sivaraj
K.K.
and
Adams
R.H.
(
2016
)
Blood vessel formation and function in bone
.
Development
143
,
2706
2715
[PubMed]
6.
Ansorge
M.
and
Pompe
T.
(
2018
)
Systems for localized release to mimic paracrine cell communication in vitro
.
J. Control. Release
278
,
24
36
7.
Shao
H.
,
Im
H.
,
Castro
C.M.
,
Breakefield
X.
,
Weissleder
R.
and
Lee
H.
(
2018
)
New technologies for analysis of extracellular vesicles
.
Chem. Rev.
118
,
1917
1950
[PubMed]
8.
Kusumbe
A.P.
,
Ramasamy
S.K.
and
Adams
R.H.
(
2014
)
Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone
.
Nature
507
,
323
328
[PubMed]
9.
Brandi
M.L.
and
Collin-Osdoby
P.
(
2006
)
Vascular biology and the skeleton
.
J. Bone Miner. Res.
21
,
183
192
10.
Romeo
S.G.
,
Alawi
K.M.
,
Rodrigues
J.
,
Singh
A.
,
Kusumbe
A.P.
and
Ramasamy
S.K.
(
2019
)
Endothelial proteolytic activity and interaction with non-resorbing osteoclasts mediate bone elongation
.
Nat. Cell Biol.
21
,
430
441
[PubMed]
11.
Lu
C.
,
Marcucio
R.
and
Miclau
T.
(
2006
)
Assessing angiogenesis during fracture healing
.
Iowa Orthop. J.
26
,
17
26
[PubMed]
12.
Langen
U.H.
,
Pitulescu
M.E.
,
Kim
J.M.
,
Enriquez-Gasca
R.
,
Sivaraj
K.K.
,
Kusumbe
A.P.
et al.
(
2017
)
Cell-matrix signals specify bone endothelial cells during developmental osteogenesis
.
Nat. Cell Biol.
19
,
189
201
[PubMed]
13.
Crisan
M.
,
Yap
S.
,
Casteilla
L.
,
Chen
C.W.
,
Corselli
M.
,
Park
T.S.
et al.
(
2008
)
A perivascular origin for mesenchymal stem cells in multiple human organs
.
Cell Stem Cell
3
,
301
313
[PubMed]
14.
da Silva Meirelles
L.
,
Chagastelles
P.C.
and
Nardi
N.B.
(
2006
)
Mesenchymal stem cells reside in virtually all post-natal organs and tissues
.
J. Cell Sci.
119
,
2204
2213
[PubMed]
15.
Shi
S.
and
Gronthos
S.
(
2003
)
Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp
.
J. Bone Miner. Res.
18
,
696
704
16.
Su
P.
,
Tian
Y.
,
Yang
C.
,
Ma
X.
,
Wang
X.
,
Pei
J.
et al.
(
2018
)
Mesenchymal stem cell migration during bone formation and bone diseases therapy
.
Int. J. Mol. Sci.
19
,
2343
17.
Francis
K.
and
Palsson
B.O.
(
1997
)
Effective intercellular communication distances are determined by the relative time constants for cyto/chemokine secretion and diffusion
.
Proc. Natl. Acad. Sci. U.S.A.
94
,
12258
12262
[PubMed]
18.
Yoshida
F.
and
Horiike
K.
(
2002
)
Dependence of effective communication distance and characteristic time on the secretion rate in intercellular signaling
.
Jpn. J. Physiol.
52
,
399
401
[PubMed]
19.
Gupta
V.K.
(
2017
)
Effect of cyto/chemokine degradation in effective intercellular communication distances
.
Phys. A
468
,
244
251
[PubMed]
20.
Xie
H.
,
Cui
Z.
,
Wang
L.
,
Xia
Z.
,
Hu
Y.
,
Xian
L.
et al.
(
2014
)
PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis
.
Nat. Med.
20
,
1270
1278
[PubMed]
21.
Gao
B.
,
Deng
R.
,
Chai
Y.
,
Chen
H.
,
Hu
B.
,
Wang
X.
et al.
(
2019
)
Macrophage-lineage TRAP+ cells recruit periosteum-derived cells for periosteal osteogenesis and regeneration
.
J. Clin. Invest.
129
,
2578
2594
[PubMed]
22.
Enge
M.
,
Bjarnegård
M.
,
Gerhardt
H.
,
Gustafsson
E.
,
Kalén
M.
,
Asker
N.
et al.
(
2002
)
Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy
.
EMBO J.
21
,
4307
4316
[PubMed]
23.
Gerhardt
H.
and
Betsholtz
C.
(
2003
)
Endothelial-pericyte interactions in angiogenesis
.
Cell Tissue Res.
314
,
15
23
[PubMed]
24.
Sanz
L.
,
Santos-Valle
P.
,
Alonso-Camino
V.
,
Salas
C.
,
Serrano
A.
,
Vicario
J.L.
et al.
(
2008
)
Long-term in vivo imaging of human angiogenesis: critical role of bone marrow-derived mesenchymal stem cells for the generation of durable blood vessels
.
Microvasc. Res.
75
,
308
314
[PubMed]
25.
Sorrell
J.M.
,
Baber
M.A.
and
Caplan
A.I.
(
2009
)
Influence of adult mesenchymal stem cells on in vitro vascular formation
.
Tissue Eng. Part A
15
,
1751
1761
[PubMed]
26.
Gharibi
B.
,
Ghuman
M.S.
and
Hughes
F.J.
(
2012
)
Akt- and Erk-mediated regulation of proliferation and differentiation during PDGFRbeta-induced MSC self-renewal
.
J. Cell. Mol. Med.
16
,
2789
2801
[PubMed]
27.
Zhou
H.
,
Yang
Y.H.
and
Basile
J.R.
(
2014
)
The Semaphorin 4D-Plexin-B1-RhoA signaling axis recruits pericytes and regulates vascular permeability through endothelial production of PDGF-B and ANGPTL4
.
Angiogenesis
17
,
261
274
[PubMed]
28.
Tokunaga
A.
,
Oya
T.
,
Ishii
Y.
,
Motomura
H.
,
Nakamura
C.
,
Ishizawa
S.
et al.
(
2008
)
PDGF receptor beta is a potent regulator of mesenchymal stromal cell function
.
J. Bone Miner. Res.
23
,
1519
1528
[PubMed]
29.
Mishima
Y.
and
Lotz
M.
(
2008
)
Chemotaxis of human articular chondrocytes and mesenchymal stem cells
.
J. Orthop. Res.
26
,
1407
1412
[PubMed]
30.
Ng
F.
,
Boucher
S.
,
Koh
S.
,
Sastry
K.S.
,
Chase
L.
,
Lakshmipathy
U.
et al.
(
2008
)
PDGF, TGF-beta, and FGF signaling is important for differentiation and growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages
.
Blood
112
,
295
307
[PubMed]
31.
Gruber
R.
,
Karreth
F.
,
Kandler
B.
,
Fuerst
G.
,
Rot
A.
,
Fischer
M.B.
et al.
(
2004
)
Platelet-released supernatants increase migration and proliferation, and decrease osteogenic differentiation of bone marrow-derived mesenchymal progenitor cells under in vitro conditions
.
Platelets
15
,
29
35
[PubMed]
32.
Kratchmarova
I.
,
Blagoev
B.
,
Haack-Sorensen
M.
,
Kassem
M.
and
Mann
M.
(
2005
)
Mechanism of divergent growth factor effects in mesenchymal stem cell differentiation
.
Science
308
,
1472
1477
[PubMed]
33.
Raines
E.W.
and
Ross
R.
(
1992
)
Compartmentalization of PDGF on extracellular binding sites dependent on exon-6-encoded sequences
.
J. Cell Biol.
116
,
533
543
[PubMed]
34.
Abramsson
A.
,
Lindblom
P.
and
Betsholtz
C.
(
2003
)
Endothelial and nonendothelial sources of PDGF-B regulate pericyte recruitment and influence vascular pattern formation in tumors
.
J. Clin. Invest.
112
,
1142
1151
[PubMed]
35.
Seghezzi
G.
,
Patel
S.
,
Ren
C.J.
,
Gualandris
A.
,
Pintucci
G.
,
Robbins
E.S.
et al.
(
1998
)
Fibroblast growth factor-2 (FGF-2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: an autocrine mechanism contributing to angiogenesis
.
J. Cell Biol.
141
,
1659
1673
[PubMed]
36.
Tang
N.
,
Wang
L.
,
Esko
J.
,
Giordano
F.J.
,
Huang
Y.
,
Gerber
H.-P.
et al.
(
2004
)
Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis
.
Cancer Cell
6
,
485
495
[PubMed]
37.
Ball
S.G.
,
Shuttleworth
C.A.
and
Kielty
C.M.
(
2007
)
Vascular endothelial growth factor can signal through platelet-derived growth factor receptors
.
J. Cell Biol.
177
,
489
500
[PubMed]
38.
Greenberg
J.I.
,
Shields
D.J.
,
Barillas
S.G.
,
Acevedo
L.M.
,
Murphy
E.
,
Huang
J.
et al.
(
2008
)
A role for VEGF as a negative regulator of pericyte function and vessel maturation
.
Nature
456
,
809
813
[PubMed]
39.
Walpole
J.
,
Mac Gabhann
F.
,
Peirce
S.M.
and
Chappell
J.C.
(
2017
)
Agent-based computational model of retinal angiogenesis simulates microvascular network morphology as a function of pericyte coverage
.
Microcirculation
24
,
[PubMed]
40.
Barleon
B.
,
Sozzani
S.
,
Zhou
D.
,
Weich
H.A.
,
Mantovani
A.
and
Marmé
D.
(
1996
)
Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1
.
Blood
87
,
3336
3343
[PubMed]
41.
Leek
R.D.
,
Hunt
N.C.
,
Landers
R.J.
,
Lewis
C.E.
,
Royds
J.A.
and
Harris
A.L.
(
2000
)
Macrophage infiltration is associated with VEGF and EGFR expression in breast cancer
.
J. Pathol.
190
,
430
436
[PubMed]
42.
Nakagawa
M.
,
Kaneda
T.
,
Arakawa
T.
,
Morita
S.
,
Sato
T.
,
Yomada
T.
et al.
(
2000
)
Vascular endothelial growth factor (VEGF) directly enhances osteoclastic bone resorption and survival of mature osteoclasts
.
FEBS Lett.
473
,
161
164
[PubMed]
43.
Aldridge
S.E.
,
Lennard
T.W.J.
,
Williams
J.R.
and
Birch
M.A.
(
2005
)
Vascular endothelial growth factor receptors in osteoclast differentiation and function
.
Biochem. Biophys. Res. Commun.
335
,
793
798
[PubMed]
44.
Engsig
M.T.
,
Chen
Q.J.
,
Vu
T.H.
,
Pedersen
A.C.
,
Therkidsen
B.
,
Lund
L.R.
et al.
(
2000
)
Matrix metalloproteinase 9 and vascular endothelial growth factor are essential for osteoclast recruitment into developing long bones
.
J. Cell Biol.
151
,
879
889
[PubMed]
45.
Chan
C.K.
,
Seo
E.Y.
,
Chen
J.Y.
,
Lo
D.
,
McArdle
A.
,
Sinha
R.
et al.
(
2015
)
Identification and specification of the mouse skeletal stem cell
.
Cell
160
,
285
298
[PubMed]
46.
Carlevaro
M.F.
,
Cermelli
S.
,
Cancedda
R.
and
Descalzi Cancedda
F.
(
2000
)
Vascular endothelial growth factor (VEGF) in cartilage neovascularization and chondrocyte differentiation: auto-paracrine role during endochondral bone formation
.
J. Cell Sci.
113
,
59
69
[PubMed]
47.
Zhang
L.F.
,
Qi
J.
,
Zuo
G.
,
Jia
P.
,
Shen
X.
,
Shao
J.
et al.
(
2014
)
Osteoblast-secreted factors promote proliferation and osteogenic differentiation of bone marrow stromal cells via VEGF/heme-oxygenase-1 pathway
.
PLoS ONE
9
,
e99946
[PubMed]
48.
Hu
K.
and
Olsen
B.R.
(
2016
)
Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair
.
J. Clin. Invest.
126
,
509
526
[PubMed]
49.
de Castro
L.F.
,
Maycas
M.
,
Bravo
B.
,
Esbrit
P.
and
Gortazar
A.
(
2015
)
VEGF receptor 2 (VEGFR2) activation is essential for osteocyte survival induced by mechanotransduction
.
J. Cell. Physiol.
230
,
278
285
[PubMed]
50.
Yoon
B.S.
,
Ovchinnikov
D.A.
,
Yoshii
I.
,
Mishina
Y.
,
Behringer
R.R.
and
Lyons
K.M.
(
2005
)
Bmpr1a and Bmpr1b have overlapping functions and are essential for chondrogenesis in vivo
.
Proc. Natl. Acad. Sci. U.S.A.
102
,
5062
5067
[PubMed]
51.
Tsuji
K.
,
Bandyopadhyay
A.
,
Harfe
B.D.
,
Cox
K.
,
Kakar
S.
,
Gerstenfeld
L.
et al.
(
2006
)
BMP2 activity, although dispensable for bone formation, is required for the initiation of fracture healing
.
Nat. Genet.
38
,
1424
1429
[PubMed]
52.
Chai
S.
,
Wan
L.
,
Wang
J.L.
,
Huang
J.C.
and
Huang
H.X.
(
2019
)
Gushukang inhibits osteocyte apoptosis and enhances BMP-2/Smads signaling pathway in ovariectomized rats
.
Phytomedicine
64
,
153063
[PubMed]
53.
Bostrom
K.I.
,
Rajamannan
N.M.
and
Towler
D.A.
(
2011
)
The regulation of valvular and vascular sclerosis by osteogenic morphogens
.
Circ. Res.
109
,
564
577
[PubMed]
54.
Csiszar
A.
,
Lehoux
S.
and
Ungvari
Z.
(
2009
)
Hemodynamic forces, vascular oxidative stress, and regulation of BMP-2/4 expression
.
Antioxid. Redox Signal.
11
,
1683
1697
55.
McBride-Gagyi
S.H.
,
McKenzie
J.A.
,
Buettmann
E.G.
,
Gardner
M.J.
and
Silva
M.J.
(
2015
)
Bmp2 conditional knockout in osteoblasts and endothelial cells does not impair bone formation after injury or mechanical loading in adult mice
.
Bone
81
,
533
543
[PubMed]
56.
Yu
Y.Y.
,
Lieu
S.
,
Lu
C.
,
Miclau
T.
,
Marcucio
R.S.
and
Colnot
C.
(
2010
)
Immunolocalization of BMPs, BMP antagonists, receptors, and effectors during fracture repair
.
Bone
46
,
841
851
[PubMed]
57.
Kloen
P.
,
Lauzier
D.
and
Hamdy
R.C.
(
2012
)
Co-expression of BMPs and BMP-inhibitors in human fractures and non-unions
.
Bone
51
,
59
68
[PubMed]
58.
McBride
S.H.
,
McKenzie
J.A.
,
Bedrick
B.S.
,
Kuhlmann
P.
,
Pasteris
J.D.
,
Rosen
V.
et al.
(
2014
)
Long bone structure and strength depend on BMP2 from osteoblasts and osteocytes, but not vascular endothelial cells
.
PLoS ONE
9
,
e96862
[PubMed]
59.
Bostrom
K.I.
,
Jumabay
M.
,
Matveyenko
A.
,
Nicholas
S.B.
and
Yao
Y.
(
2011
)
Activation of vascular bone morphogenetic protein signaling in diabetes mellitus
.
Circ. Res.
108
,
446
457
[PubMed]
60.
Tsuji
K.
,
Cox
K.
,
Bandyopadhyay
A.
,
Harfe
B.D.
,
Tabin
C.J.
and
Rosen
V.
(
2008
)
BMP4 is dispensable for skeletogenesis and fracture-healing in the limb
.
J. Bone. Joint Surg. Am.
90
,
14
18
[PubMed]
61.
Dan
H.
,
Simsa-Maziel
S.
,
Reich
A.
,
Sela-Donenfeld
D.
and
Monsonego-Ornan
E.
(
2012
)
The role of matrix gla protein in ossification and recovery of the avian growth plate
.
Front. Endocrinol. (Lausanne)
3
,
79
[PubMed]
62.
Ramasamy
S.K.
,
Kusumbe
A.P.
,
Wang
L.
and
Adams
R.H.
(
2014
)
Endothelial Notch activity promotes angiogenesis and osteogenesis in bone
.
Nature
507
,
376
380
[PubMed]
63.
Yao
Y.
,
Shahbazian
A.
and
Boström
K.I.
(
2008
)
Proline and γ-carboxylated glutamate residues in matrix Gla protein are critical for binding of bone morphogenetic protein-4
.
Circ. Res.
102
,
1065
1074
[PubMed]
64.
Pardali
E.
,
Makowski
L.M.
,
Leffers
M.
,
Borgscheiper
A.
and
Waltenberger
J.
(
2018
)
BMP-2 induces human mononuclear cell chemotaxis and adhesion and modulates monocyte-to-macrophage differentiation
.
J. Cell. Mol. Med.
22
,
5429
5438
[PubMed]
65.
Wang
W.
,
Wang
H.
,
Zhou
X.
,
Li
X.
,
Sun
W.
,
Dellinger
M.
et al.
(
2017
)
Lymphatic endothelial cells produce M-CSF, causing massive bone loss in mice
.
J. Bone Miner. Res.
32
,
939
950
[PubMed]
66.
Collin-Osdoby
P.
,
Rothe
L.
,
Anderson
F.
,
Nelson
M.
,
Maloney
W.
and
Osdoby
P.
(
2001
)
Receptor activator of NF-kappa B and osteoprotegerin expression by human microvascular endothelial cells, regulation by inflammatory cytokines, and role in human osteoclastogenesis
.
J. Biol. Chem.
276
,
20659
20672
[PubMed]
67.
Ishida
A.
,
Fujita
N.
,
Kitazawa
R.
and
Tsuruo
T.
(
2002
)
Transforming growth factor-beta induces expression of receptor activator of NF-kappa B ligand in vascular endothelial cells derived from bone
.
J. Biol. Chem.
277
,
26217
26224
[PubMed]
68.
Malyankar
U.M.
,
Scatena
M.
,
Suchland
K.L.
,
Yun
T.J.
,
Clark
E.A.
and
Giachelli
C.M.
(
2000
)
Osteoprotegerin is an alpha vbeta 3-induced, NF-kappa B-dependent survival factor for endothelial cells
.
J. Biol. Chem.
275
,
20959
20962
[PubMed]
69.
Chikatsu
N.
,
Takeuchi
Y.
,
Fukumoto
S.
,
Yano
K.
,
Fujita
N.
,
Tsuruo
T.
et al.
(
2002
)
Clonal endothelial cells produce humoral factors that inhibit osteoclast-like cell formation in vitro
.
Endocr. J.
49
,
439
447
[PubMed]
70.
Lekesiz
K.
,
Naumnik
B.
,
Borysewicz-Sanczyk
H.
,
Koc-Zurawska
E.
and
Mysliwiec
M.
(
2013
)
Effect of unfractionated heparin, enoxaparin and sulodexide on the relations between secretion and expression of OPG, RANKL and vWF in HUVEC
.
Folia Histochem. Cytobiol.
51
,
156
163
[PubMed]
71.
Nakashima
T.
,
Hayashi
M.
,
Fukunaga
T.
,
Kurata
K.
,
Oh-Hora
M.
,
Feng
J.Q.
et al.
(
2011
)
Evidence for osteocyte regulation of bone homeostasis through RANKL expression
.
Nat. Med.
17
,
1231
1234
[PubMed]
72.
Moriishi
T.
,
Fukuyama
R.
,
Ito
M.
,
Miyazaki
T.
,
Maeno
T.
,
Kawai
Y.
et al.
(
2012
)
Osteocyte network; a negative regulatory system for bone mass augmented by the induction of Rankl in osteoblasts and Sost in osteocytes at unloading
.
PLoS ONE
7
,
e40143
[PubMed]
73.
Li
Y.
,
Toraldo
G.
,
Li
A.
,
Yang
X.
,
Zhang
H.
,
Qian
W.P.
et al.
(
2007
)
B cells and T cells are critical for the preservation of bone homeostasis and attainment of peak bone mass in vivo
.
Blood
109
,
3839
3848
[PubMed]
74.
Han
K.H.
,
Ryu
J.W.
,
Lim
K.E.
,
Lee
S.H.
,
Kim
Y.
,
Hwang
C.S.
et al.
(
2014
)
Vascular expression of the chemokine CX3CL1 promotes osteoclast recruitment and exacerbates bone resorption in an irradiated murine model
.
Bone
61
,
91
101
[PubMed]
75.
Sucur
A.
,
Jajic
Z.
,
Artukovic
M.
,
Matijasevic
M.I.
,
Anic
B.
,
Flegar
D.
et al.
(
2017
)
Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells
.
Arthritis Res. Ther.
19
,
142
[PubMed]
76.
Xia
J.-B.
,
Liu
G.-H.
,
Chen
Z.-Y.
,
Mao
C.-Z.
,
Zhou
D.-C.
,
Wu
H.-Y.
et al.
(
2016
)
Hypoxia/ischemia promotes CXCL10 expression in cardiac microvascular endothelial cells by NFkB activation
.
Cytokine
81
,
63
70
[PubMed]
77.
Hillyer
P.
,
Mordelet
E.
,
Flynn
G.
and
Male
D.
(
2003
)
Chemokines, chemokine receptors and adhesion molecules on different human endothelia: discriminating the tissue-specific functions that affect leucocyte migration
.
Clin. Exp. Immunol.
134
,
431
441
[PubMed]
78.
Lu
Y.
,
Xiao
G.
,
Galson
D.L.
,
Nishio
Y.
,
Mizokami
A.
,
Keller
E.T.
et al.
(
2007
)
PTHrP-induced MCP-1 production by human bone marrow endothelial cells and osteoblasts promotes osteoclast differentiation and prostate cancer cell proliferation and invasion in vitro
.
Int. J. Cancer
121
,
724
733
[PubMed]
79.
Kitase
Y.
,
Lee
S.
,
Gluhak-Heinrich
J.
,
Johnson
M.L.
,
Harris
S.E.
and
Bonewald
L.F.
(
2014
)
CCL7 is a protective factor secreted by mechanically loaded osteocytes
.
J. Dent. Res.
93
,
1108
1115
[PubMed]
80.
Cai
W.
,
Tao
J.
,
Zhang
X.
,
Tian
X.
,
Liu
T.
,
Feng
X.
et al.
(
2014
)
Contribution of homeostatic chemokines CCL19 and CCL21 and their receptor CCR7 to coronary artery disease
.
Arterioscler. Thromb. Vasc. Biol.
34
,
1933
1941
[PubMed]
81.
Lee
J.
,
Park
C.
,
Kim
H.J.
,
Lee
Y.D.
,
Lee
Z.H.
,
Song
Y.W.
et al.
(
2017
)
Stimulation of osteoclast migration and bone resorption by C-C chemokine ligands 19 and 21
.
Exp. Mol. Med.
49
,
e358
[PubMed]
82.
Cui
N.
,
Hu
M.
and
Khalil
R.A.
(
2017
)
Biochemical and biological attributes of matrix metalloproteinases
.
Prog. Mol. Biol. Transl. Sci.
147
,
1
73
[PubMed]
83.
Blavier
L.
and
Delaissé
J.M.
(
1995
)
Matrix metalloproteinases are obligatory for the migration of preosteoclasts to the developing marrow cavity of primitive long bones
.
J. Cell Sci.
108
,
3649
3659
[PubMed]
84.
Karsdal
M.A.
,
Andersen
T.A.
,
Bonewald
L.
and
Christiansen
C.
(
2004
)
Matrix metalloproteinases (MMPs) safeguard osteoblasts from apoptosis during transdifferentiation into osteocytes: MT1-MMP maintains osteocyte viability
.
DNA Cell Biol.
23
,
155
165
[PubMed]
85.
Delgado-Calle
J.
,
Hancock
B.
,
Likine
E.F.
,
Sato
A.Y.
,
McAndrews
K.
,
Sanudo
C.
et al.
(
2018
)
MMP14 is a novel target of PTH signaling in osteocytes that controls resorption by regulating soluble RANKL production
.
FASEB J.
32
,
2878
2890
[PubMed]
86.
Barile
L.
,
Moccetti
T.
,
Marban
E.
and
Vassalli
G.
(
2017
)
Roles of exosomes in cardioprotection
.
Eur. Heart J.
38
,
1372
1379
[PubMed]
87.
Alique
M.
,
Ruíz-Torres
M.P.
,
Bodega
G.
,
Noci
M.V.
,
Troyano
N.
,
Bohórquez
L.
et al.
(
2017
)
Microvesicles from the plasma of elderly subjects and from senescent endothelial cells promote vascular calcification
.
Aging (Albany N.Y.)
9
,
778
789
[PubMed]
88.
Hackl
M.
,
Brunner
S.
,
Fortschegger
K.
,
Schreiner
C.
,
Micutkova
L.
,
Mück
C.
et al.
(
2010
)
miR-17, miR-19b, miR-20a, and miR-106a are down-regulated in human aging
.
Aging Cell
9
,
291
296
[PubMed]
89.
Deng
Y.
,
Wu
S.
,
Zhou
H.
,
Bi
X.
,
Wang
Y.
,
Hu
Y.
et al.
(
2013
)
Effects of a miR-31, Runx2, and Satb2 regulatory loop on the osteogenic differentiation of bone mesenchymal stem cells
.
Stem Cells Dev.
22
,
2278
2286
[PubMed]
90.
Deng
Y.
,
Zhou
H.
,
Zou
D.
,
Xie
Q.
,
Bi
X.
,
Gu
P.
et al.
(
2013
)
The role of miR-31-modified adipose tissue-derived stem cells in repairing rat critical-sized calvarial defects
.
Biomaterials
34
,
6717
6728
[PubMed]
91.
Weilner
S.
,
Schraml
E.
,
Wieser
M.
,
Messner
P.
,
Schneider
K.
,
Wassermann
K.
et al.
(
2016
)
Secreted microvesicular miR-31 inhibits osteogenic differentiation of mesenchymal stem cells
.
Aging Cell
15
,
744
754
[PubMed]
92.
Curtis
A.M.
,
Edelberg
J.
,
Jonas
R.
,
Rogers
W.T.
,
Moore
J.S.
,
Syed
W.
et al.
(
2013
)
Endothelial microparticles: sophisticated vesicles modulating vascular function
.
Vasc. Med.
18
,
204
214
[PubMed]
93.
Buendía
P.
,
Montes de Oca
A.
,
Madueño
J.A.
,
Merino
A.
,
Martín-Malo
A.
,
Aljama
P.
et al.
(
2015
)
Endothelial microparticles mediate inflammation-induced vascular calcification
.
FASEB J.
29
,
173
181
[PubMed]
94.
Vítková
V.
,
Živný
J.
and
Janota
J.
(
2018
)
Endothelial cell-derived microvesicles: potential mediators and biomarkers of pathologic processes
.
Biomark. Med.
12
,
161
175
[PubMed]
95.
Njock
M.S.
,
Cheng
H.S.
,
Dang
L.T.
,
Nazari-Jahantigh
M.
,
Lau
A.C.
,
Boudreau
E.
et al.
(
2015
)
Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing antiinflammatory microRNAs
.
Blood
125
,
3202
3212
[PubMed]
96.
Zhan
R.
,
Leng
X.
,
Liu
X.
,
Wang
X.
,
Gong
J.
,
Yan
L.
et al.
(
2009
)
Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes
.
Biochem. Biophys. Res. Commun.
387
,
229
233
[PubMed]
97.
Song
H.
,
Li
X.
,
Zhao
Z.
,
Qian
J.
,
Wang
Y.
,
Cui
J.
et al.
(
2019
)
Reversal of osteoporotic activity by endothelial cell-secreted bone targeting and biocompatible exosomes
.
Nano Lett.
19
,
3040
3048
[PubMed]
98.
McCann
J.V.
,
Liu
A.
,
Musante
L.
,
Erdbrugger
U.
,
Lannigan
J.
and
Dudley
A.C.
(
2019
)
A miRNA signature in endothelial cell-derived extracellular vesicles in tumor-bearing mice
.
Sci. Rep.
9
,
16743
[PubMed]
99.
Formigli
L.
,
Orlandini
S.Z.
,
Benvenuti
S.
,
Masi
L.
,
Pinto
A.
,
Gattei
V.
et al.
(
1995
)
In vitro structural and functional relationships between preosteoclastic and bone endothelial cells: a juxtacrine model for migration and adhesion of osteoclast precursors
.
J. Cell. Physiol.
162
,
199
212
[PubMed]
100.
Sims
D.E.
(
1991
)
Recent advances in pericyte biology–implications for health and disease
.
Can. J. Cardiol.
7
,
431
443
[PubMed]
101.
Díaz-Flores
L.
,
Gutiérrez
R.
,
Varela
H.
,
Rancel
N.
and
Valladares
F.
(
1991
)
Microvascular pericytes: a review of their morphological and functional characteristics
.
Histol. Histopathol.
6
,
269
286
[PubMed]
102.
Doherty
M.J.
and
Canfield
A.E.
(
1999
)
Gene expression during vascular pericyte differentiation
.
Crit. Rev. Eukaryot. Gene Expr.
9
,
1
17
[PubMed]
103.
Farrington-Rock
C.
,
Crofts
N.J.
,
Doherty
M.J.
,
Ashton
B.A.
,
Griffin-Jones
C.
and
Canfield
A.E.
(
2004
)
Chondrogenic and adipogenic potential of microvascular pericytes
.
Circulation
110
,
2226
2232
[PubMed]
104.
Doherty
M.J.
,
Ashton
B.A.
,
Walsh
S.
,
Beresford
J.N.
,
Grant
M.E.
and
Canfield
A.E.
(
1998
)
Vascular pericytes express osteogenic potential in vitro and in vivo
.
J. Bone Miner. Res.
13
,
828
838
105.
Schor
A.M.
,
Allen
T.D.
,
Canfield
A.E.
,
Sloan
P.
and
Schor
S.L.
(
1990
)
Pericytes derived from the retinal microvasculature undergo calcification in vitro
.
J. Cell Sci.
97
,
449
461
[PubMed]
106.
Cao
C.
,
Huang
Y.
,
Tang
Q.
,
Zhang
C.
,
Shi
L.
,
Zhao
J.
et al.
(
2018
)
Bidirectional juxtacrine ephrinB2/Ephs signaling promotes angiogenesis of ECs and maintains self-renewal of MSCs
.
Biomaterials
172
,
1
13
[PubMed]
107.
Adams
R.H.
,
Wilkinson
G.A.
,
Weiss
C.
,
Diella
F.
,
Gale
N.W.
,
Deutsch
U.
et al.
(
1999
)
Roles of ephrinB ligands and EphB receptors in cardiovascular development: demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis
.
Genes Dev.
13
,
295
306
[PubMed]
108.
Zhao
C.
,
Irie
N.
,
Takada
Y.
,
Shimoda
K.
,
Miyamoto
T.
,
Nishiwaki
T.
et al.
(
2006
)
Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis
.
Cell Metab.
4
,
111
121
[PubMed]
109.
Tonna
S.
,
Poulton
I.J.
,
Taykar
F.
,
Ho
P.W.
,
Tonkin
B.
,
Crimeen-Irwin
B.
et al.
(
2017
)
Correction: Chondrocytic ephrin B2 promotes cartilage destruction by osteoclasts in endochondral ossification
.
Development
144
,
530
[PubMed]
110.
Allan
E.H.
,
Hausler
K.D.
,
Wei
T.
,
Gooi
J.H.
,
Quinn
J.M.
,
Crimeen-Irwin
B.
et al.
(
2008
)
EphrinB2 regulation by PTH and PTHrP revealed by molecular profiling in differentiating osteoblasts
.
J. Bone Miner. Res.
23
,
1170
1181
[PubMed]
111.
Vrahnas
C.
,
Blank
M.
,
Dite
T.A.
,
Tatarczuch
L.
,
Ansari
N.
,
Crimeen-Irwin
B.
et al.
(
2019
)
Increased autophagy in EphrinB2-deficient osteocytes is associated with elevated secondary mineralization and brittle bone
.
Nat. Commun.
10
,
3436
[PubMed]
112.
Kwan Tat
S.
,
Pelletier
J.-P.
,
Amiable
N.
,
Boileau
C.
,
Lavigne
M.
and
Martel-Pelletier
J.
(
2009
)
Treatment with ephrin B2 positively impacts the abnormal metabolism of human osteoarthritic chondrocytes
.
Arthritis Res. Ther.
11
,
R119
[PubMed]
113.
Kohn
A.
,
Dong
Y.
,
Mirando
A.J.
,
Jesse
A.M.
,
Honjo
T.
,
Zuscik
M.J.
et al.
(
2012
)
Cartilage-specific RBPjkappa-dependent and -independent Notch signals regulate cartilage and bone development
.
Development
139
,
1198
1212
[PubMed]
114.
Tao
J.
,
Chen
S.
,
Yang
T.
,
Dawson
B.
,
Munivez
E.
,
Bertin
T.
et al.
(
2010
)
Osteosclerosis owing to Notch gain of function is solely Rbpj-dependent
.
J. Bone Miner. Res.
25
,
2175
2183
[PubMed]
115.
Dong
Y.
,
Jesse
A.M.
,
Kohn
A.
,
Gunnell
L.M.
,
Honjo
T.
,
Zuscik
M.J.
et al.
(
2010
)
RBPjkappa-dependent Notch signaling regulates mesenchymal progenitor cell proliferation and differentiation during skeletal development
.
Development
137
,
1461
1471
[PubMed]
116.
Zanotti
S.
,
Smerdel-Ramoya
A.
,
Stadmeyer
L.
,
Durant
D.
,
Radtke
F.
and
Canalis
E.
(
2008
)
Notch inhibits osteoblast differentiation and causes osteopenia
.
Endocrinology
149
,
3890
3899
[PubMed]
117.
Engin
F.
,
Yao
Z.
,
Yang
T.
,
Zhou
G.
,
Bertin
T.
,
Jiang
M.M.
et al.
(
2008
)
Dimorphic effects of Notch signaling in bone homeostasis
.
Nat. Med.
14
,
299
305
[PubMed]
118.
Bai
S.
,
Kopan
R.
,
Zou
W.
,
Hilton
M.J.
,
Ong
C.T.
,
Long
F.
et al.
(
2008
)
NOTCH1 regulates osteoclastogenesis directly in osteoclast precursors and indirectly via osteoblast lineage cells
.
J. Biol. Chem.
283
,
6509
6518
[PubMed]
119.
Liang
T.
,
Zhu
L.
,
Gao
W.
,
Gong
M.
,
Ren
J.
,
Yao
H.
et al.
(
2017
)
Coculture of endothelial progenitor cells and mesenchymal stem cells enhanced their proliferation and angiogenesis through PDGF and Notch signaling
.
FEBS Open Bio
7
,
1722
1736
[PubMed]
120.
Hilton
M.J.
,
Tu
X.
,
Wu
X.
,
Bai
S.
,
Zhao
H.
,
Kobayashi
T.
et al.
(
2008
)
Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation
.
Nat. Med.
14
,
306
314
[PubMed]
121.
Zhu
F.
,
Sweetwyne
M.T.
and
Hankenson
K.D.
(
2013
)
PKCdelta is required for Jagged-1 induction of human mesenchymal stem cell osteogenic differentiation
.
Stem Cells
31
,
1181
1192
[PubMed]
122.
Nobta
M.
,
Tsukazaki
T.
,
Shibata
Y.
,
Xin
C.
,
Moriishi
T.
,
Sakano
S.
et al.
(
2005
)
Critical regulation of bone morphogenetic protein-induced osteoblastic differentiation by Delta1/Jagged1-activated Notch1 signaling
.
J. Biol. Chem.
280
,
15842
15848
[PubMed]
123.
Tezuka
K.-I.
,
Yasuda
M.
,
Watanabe
N.
,
Morimura
N.
,
Kuroda
K.
,
Miyatani
S.
et al.
(
2002
)
Stimulation of osteoblastic cell differentiation by Notch
.
J. Bone Miner. Res.
17
,
231
239
124.
Zamurovic
N.
,
Cappellen
D.
,
Rohner
D.
and
Susa
M.
(
2004
)
Coordinated activation of notch, Wnt, and transforming growth factor-beta signaling pathways in bone morphogenic protein 2-induced osteogenesis. Notch target gene Hey1 inhibits mineralization and Runx2 transcriptional activity
.
J. Biol. Chem.
279
,
37704
37715
[PubMed]
125.
Deregowski
V.
,
Gazzerro
E.
,
Priest
L.
,
Rydziel
S.
and
Canalis
E.
(
2006
)
Notch 1 overexpression inhibits osteoblastogenesis by suppressing Wnt/beta-catenin but not bone morphogenetic protein signaling
.
J. Biol. Chem.
281
,
6203
6210
[PubMed]
126.
Dishowitz
M.I.
,
Terkhorn
S.P.
,
Bostic
S.A.
and
Hankenson
K.D.
(
2012
)
Notch signaling components are upregulated during both endochondral and intramembranous bone regeneration
.
J. Orthop. Res.
30
,
296
303
[PubMed]
127.
Dishowitz
M.I.
,
Zhu
F.
,
Sundararaghavan
H.G.
,
Ifkovits
J.L.
,
Burdick
J.A.
and
Hankenson
K.D.
(
2014
)
Jagged1 immobilization to an osteoconductive polymer activates the Notch signaling pathway and induces osteogenesis
.
J. Biomed. Mater. Res. A
102
,
1558
1567
[PubMed]
128.
Pedrosa
A.R.
,
Trindade
A.
,
Fernandes
A.C.
,
Carvalho
C.
,
Gigante
J.
,
Tavares
A.T.
et al.
(
2015
)
Endothelial Jagged1 antagonizes Dll4 regulation of endothelial branching and promotes vascular maturation downstream of Dll4/Notch1
.
Arterioscler. Thromb. Vasc. Biol.
35
,
1134
1146
[PubMed]
129.
Limbourg
F.P.
,
Takeshita
K.
,
Radtke
F.
,
Bronson
R.T.
,
Chin
M.T.
and
Liao
J.K.
(
2005
)
Essential role of endothelial Notch1 in angiogenesis
.
Circulation
111
,
1826
1832
[PubMed]
130.
Krebs
L.T.
,
Xue
Y.
,
Norton
C.R.
,
Shutter
J.R.
,
Maguire
M.
,
Sundberg
J.P.
et al.
(
2000
)
Notch signaling is essential for vascular morphogenesis in mice
.
Genes Dev.
14
,
1343
1352
[PubMed]
131.
Kusumbe
A.P.
,
Ramasamy
S.K.
,
Itkin
T.
,
Mae
M.A.
,
Langen
U.H.
,
Betsholtz
C.
et al.
(
2016
)
Age-dependent modulation of vascular niches for haematopoietic stem cells
.
Nature
532
,
380
384
[PubMed]
132.
Benedito
R.
,
Roca
C.
,
Sorensen
I.
,
Adams
S.
,
Gossler
A.
,
Fruttiger
M.
et al.
(
2009
)
The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis
.
Cell
137
,
1124
1135
[PubMed]
133.
Noguera-Troise
I.
,
Daly
C.
,
Papadopoulos
N.J.
,
Coetzee
S.
,
Boland
P.
,
Gale
N.W.
et al.
(
2006
)
Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis
.
Nature
444
,
1032
1037
[PubMed]
134.
Saleh
F.A.
,
Whyte
M.
,
Ashton
P.
and
Genever
P.G.
(
2011
)
Regulation of mesenchymal stem cell activity by endothelial cells
.
Stem Cells Dev.
20
,
391
403
[PubMed]
135.
Gamrekelashvili
J.
,
Giagnorio
R.
,
Jussofie
J.
,
Soehnlein
O.
,
Duchene
J.
,
Briseno
C.G.
et al.
(
2016
)
Regulation of monocyte cell fate by blood vessels mediated by Notch signalling
.
Nat. Commun.
7
,
12597
[PubMed]
136.
Pagie
S.
,
Gerard
N.
and
Charreau
B.
(
2018
)
Notch signaling triggered via the ligand DLL4 impedes M2 macrophage differentiation and promotes their apoptosis
.
Cell Commun. Signal.
16
,
4
[PubMed]
137.
Mohtashami
M.
,
Shah
D.K.
,
Nakase
H.
,
Kianizad
K.
,
Petrie
H.T.
and
Zuniga-Pflucker
J.C.
(
2010
)
Direct comparison of Dll1- and Dll4-mediated Notch activation levels shows differential lymphomyeloid lineage commitment outcomes
.
J. Immunol.
185
,
867
876
[PubMed]
138.
Hota
P.K.
and
Buck
M.
(
2012
)
Plexin structures are coming: opportunities for multilevel investigations of semaphorin guidance receptors, their cell signaling mechanisms, and functions
.
Cell. Mol. Life Sci.
69
,
3765
3805
[PubMed]
139.
Love
C.A.
,
Harlos
K.
,
Mavaddat
N.
,
Davis
S.J.
,
Stuart
D.I.
,
Jones
E.Y.
et al.
(
2003
)
The ligand-binding face of the semaphorins revealed by the high-resolution crystal structure of SEMA4D
.
Nat. Struct. Biol.
10
,
843
848
[PubMed]
140.
Dacquin
R.
,
Domenget
C.
,
Kumanogoh
A.
,
Kikutani
H.
,
Jurdic
P.
and
Machuca-Gayet
I.
(
2011
)
Control of bone resorption by semaphorin 4D is dependent on ovarian function
.
PLoS ONE
6
,
e26627
[PubMed]
141.
Villars
F.
,
Guillotin
B.
,
Amédée
T.
,
Dutoya
S.
,
Bordenave
L.
,
Bareille
R.
et al.
(
2002
)
Effect of HUVEC on human osteoprogenitor cell differentiation needs heterotypic gap junction communication
.
Am. J. Physiol. Cell Physiol.
282
,
C775
C785
[PubMed]
142.
Otsu
K.
,
Das
S.
,
Houser
S.D.
,
Quadri
S.K.
,
Bhattacharya
S.
and
Bhattacharya
J.
(
2009
)
Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells
.
Blood
113
,
4197
4205
[PubMed]
143.
Spees
J.L.
,
Olson
S.D.
,
Whitney
M.J.
and
Prockop
D.J.
(
2006
)
Mitochondrial transfer between cells can rescue aerobic respiration
.
Proc. Natl. Acad. Sci. U.S.A.
103
,
1283
1288
[PubMed]
144.
Koyanagi
M.
,
Brandes
R.P.
,
Haendeler
J.
,
Zeiher
A.M.
and
Dimmeler
S.
(
2005
)
Cell-to-cell connection of endothelial progenitor cells with cardiac myocytes by nanotubes: a novel mechanism for cell fate changes?
Circ. Res.
96
,
1039
1041
[PubMed]
145.
Burkhardt
R.
,
Kettner
G.
,
Böhm
W.
,
Schmidmeier
M.
,
Schlag
R.
,
Frisch
B.
et al.
(
1987
)
Changes in trabecular bone, hematopoiesis and bone marrow vessels in aplastic anemia, primary osteoporosis, and old age: a comparative histomorphometric study
.
Bone
8
,
157
164
[PubMed]
146.
Zhao
Q.
,
Shen
X.
,
Zhang
W.
,
Zhu
G.
,
Qi
J.
and
Deng
L.
(
2012
)
Mice with increased angiogenesis and osteogenesis due to conditional activation of HIF pathway in osteoblasts are protected from ovariectomy induced bone loss
.
Bone
50
,
763
770
[PubMed]
147.
Ramasamy
S.K.
,
Kusumbe
A.P.
,
Schiller
M.
,
Zeuschner
D.
,
Bixel
M.G.
,
Milia
C.
et al.
(
2016
)
Blood flow controls bone vascular function and osteogenesis
.
Nat. Commun.
7
,
13601
[PubMed]
148.
Xu
R.
,
Yallowitz
A.
,
Qin
A.
,
Wu
Z.
,
Shin
D.Y.
,
Kim
J.M.
et al.
(
2018
)
Targeting skeletal endothelium to ameliorate bone loss
.
Nat. Med.
24
,
823
833
[PubMed]
149.
Colnot
C.
,
Thompson
Z.
,
Miclau
T.
,
Werb
Z.
and
Helms
J.A.
(
2003
)
Altered fracture repair in the absence of MMP9
.
Development
130
,
4123
4133
[PubMed]
150.
Marx
R.E.
(
1983
)
Osteoradionecrosis: a new concept of its pathophysiology
.
J. Oral Maxillofac. Surg.
41
,
283
288
[PubMed]
151.
Mundy
G.R.
(
2002
)
Metastasis to bone: causes, consequences and therapeutic opportunities
.
Nat. Rev. Cancer
2
,
584
593
[PubMed]
152.
Singh
A.
,
Veeriah
V.
,
Xi
P.
,
Labella
R.
,
Chen
J.
,
Romeo
S.G.
et al.
(
2019
)
Angiocrine signals regulate quiescence and therapy resistance in bone metastasis
.
JCI Insight
4
,
e125679
153.
Li
X.
,
Loberg
R.
,
Liao
J.
,
Ying
C.
,
Snyder
L.A.
,
Pienta
K.J.
et al.
(
2009
)
A destructive cascade mediated by CCL2 facilitates prostate cancer growth in bone
.
Cancer Res.
69
,
1685
1692
[PubMed]
154.
Loberg
R.D.
,
Day
L.L.
,
Harwood
J.
,
Ying
C.
,
St John
L.N.
,
Giles
R.
et al.
(
2006
)
CCL2 is a potent regulator of prostate cancer cell migration and proliferation
.
Neoplasia
8
,
578
586
[PubMed]
155.
Pironti
G.
,
Strachan
R.T.
,
Abraham
D.
,
Mon-Wei Yu
S.
,
Chen
M.
,
Chen
W.
et al.
(
2015
)
Circulating exosomes induced by cardiac pressure overload contain functional angiotensin II type 1 receptors
.
Circulation
131
,
2120
2130
[PubMed]
156.
Li
D.
,
Liu
J.
,
Guo
B.
,
Liang
C.
,
Dang
L.
,
Lu
C.
et al.
(
2016
)
Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation
.
Nat. Commun.
7
,
10872
[PubMed]
157.
Divieti Pajevic
P.
and
Krause
D.S.
(
2019
)
Osteocyte regulation of bone and blood
.
Bone
119
,
13
18
[PubMed]
158.
Franz-Odendaal
T.A.
,
Hall
B.K.
and
Witten
P.E.
(
2006
)
Buried alive: how osteoblasts become osteocytes
.
Dev. Dyn.
235
,
176
190
[PubMed]
159.
Wang
H.U.
,
Chen
Z.F.
and
Anderson
D.J.
(
1998
)
Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4
.
Cell
93
,
741
753
[PubMed]
160.
Gerety
S.S.
,
Wang
H.U.
,
Chen
Z.F.
and
Anderson
D.J.
(
1999
)
Symmetrical mutant phenotypes of the receptor EphB4 and its specific transmembrane ligand ephrin-B2 in cardiovascular development
.
Mol. Cell
4
,
403
414
[PubMed]
161.
Wang
Y.
,
Menendez
A.
,
Fong
C.
,
ElAlieh
H.Z.
,
Chang
W.
and
Bikle
D.D.
(
2014
)
Ephrin B2/EphB4 mediates the actions of IGF-I signaling in regulating endochondral bone formation
.
J. Bone Miner. Res.
29
,
1900
1913
[PubMed]
162.
Iso
T.
,
Hamamori
Y.
and
Kedes
L.
(
2003
)
Notch signaling in vascular development
.
Arterioscler. Thromb. Vasc. Biol.
23
,
543
553
[PubMed]

Author notes

*

These authors contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).