Skip to main content
Tissue Engineering. Part B, Reviews logoLink to Tissue Engineering. Part B, Reviews
. 2021 Aug 11;27(4):313–329. doi: 10.1089/ten.teb.2020.0154

Bone Microvasculature: Stimulus for Tissue Function and Regeneration

Eun-Jin Lee 1, Mahim Jain 2, Stella Alimperti 1,
PMCID: PMC8390780  PMID: 32940150

Abstract

Bone is a highly vascularized organ, providing structural support to the body, and its development, regeneration, and remodeling depend on the microvascular homeostasis. Loss or impairment of vascular function can develop diseases, such as large bone defects, avascular necrosis, osteoporosis, osteoarthritis, and osteopetrosis. In this review, we summarize how vasculature controls bone development and homeostasis in normal and disease cases. A better understanding of this process will facilitate the development of novel disease treatments that promote bone regeneration and remodeling. Specifically, approaches based on tissue engineering components, such as stem cells and growth factors, have demonstrated the capacity to induce bone microvasculature regeneration and mineralization. This knowledge will have relevant clinical implications for the treatment of bone disorders by developing novel pharmaceutical approaches and bone grafts. Finally, the tissue engineering approaches incorporating vascular components may widely be applied to treat other organ diseases by enhancing their regeneration capacity.

Impact statement

Bone vasculature is imperative in the process of bone development, regeneration, and remodeling. Alterations or disruption of the bone vasculature leads to loss of bone homeostasis and the development of bone diseases. In this study, we review the role of vasculature on bone diseases and how vascular tissue engineering strategies, with a detailed emphasis on the role of stem cells and growth factors, will contribute to bone therapeutics.

Keywords: bone vasculature, angiogenesis, osteogenesis, tissue engineering, diseases, growth factors, stem cells

Introduction

The skeletal system, including bones and joints, supports body structure and movement. Bone has a complex structure with multiple intricate hierarchical architectures preserving functionality throughout the lifetime.1–3 Several risk factors, such as injuries, age, and genetic diseases, may impair bone homeostasis. Under these circumstances, the bone may be either nonfunctional or incompletely repaired with limited functionality.4,5

Surgical procedures using bone grafting are considered the major strategy to repair and rebuild damaged bones. These methods are often associated with complications, such as reduced integration with the host tissue, high risk of infections, and rejections by the immune system.6,7 Thus, the demand of developing novel strategies for treatments is necessary. The emerging field of tissue engineering holds promises for developing novel scaffolds, bioactive molecules, and stem cell technologies to promote bone regeneration.8

Successful bone regeneration requires neovessel formation between the host and the graft without development of fibrous tissue at the bone-implant interface and eventually, complete replacement of the scaffold with new bone. Given the significance of vasculature in bone functionality, we will describe the importance of bone vasculature during bone development, regeneration, and remodeling under normal and diseased cases. We will then address how stem cells and growth factors as therapeutic tissue engineering approaches are able to promote bone microvasculature and mineral regeneration.

Bone Structure

The blood vessels supply the bone with nutrients, oxygen, growth factors, hormones, and osteoprogenitor cells. The largest vessel, named nutrient artery, supplies more than 50% of total blood volume into the bone. It branches from the circulatory system and enters through the cortex into the innermost part of bone, named medullar cavity. It sprouts further into medullar cavity by forming microvascular branches, which anastomose with metaphyseal and epiphyseal blood vessels. Within the cortex, the arterial sprouts are extending longitudinally to bone, while others proceed radially and ultimately form capillaries within the Haversian system. Some of these arterioles anastomose with periosteal vascular network. Specifically, periosteum has been described as a thin highly vascularized membrane lined with osteoblast precursor cells, which envelopes the bone structure. Beneath the periosteum, the microvessels divide into branches, thereby entering the Volkmann's canals to supply the Haversian canal in the cortex (Fig. 1).3,9,10

FIG. 1.

FIG. 1.

Schematic of bone vasculature. (A) Longitudinal section shows vascular system in the epiphysis, metaphysis, and diaphysis areas of long bone. Boxed regions are magnified in (B). (B) Diagram shows arrangement of arteries and veins, and the blood supply into bone, demonstrating the connection between cortical and medullary blood flow. Periosteal arteries are connected intermittently with cortical blood vessels. Color images are available online.

Vascularization in Bone Development

The bone development begins by the sixth or seventh week of embryonic life. The process of bone development from hyaline cartilage is named endochondral ossification, and from fibrous membranes, it is named intramembranous ossification.11 The latter one is involved in the development of flat bones of the skull, mandibles, and clavicles. All the other bone types, including femur and tibia, are formed through the process of endochondral ossification. It has been reported that both intramembranous and endochondral angiogenesis participate in similar molecular mechanisms.12 Specifically, in intramembranous ossification, mesenchymal cells directly differentiate into osteoprogenitors and osteoblasts. These cells are further differentiated to osteocytes and they secrete proangiogenic factors, that is, vascular endothelial growth factor (VEGF), extracellular matrix proteins, and osteoids, which stimulate the formation of blood vessel in the ossification center. As flat bone is further developing, vascularization induces osteogenesis.13

Similar to intramembranous ossification, the process of endochondral angiogenesis involves a series of events. At embryonic period (∼E13), mesenchymal cells aggregate and differentiate into chondrocytes, which cease proliferation and become hypertrophic. During the embryonic period (∼E15), the hypertrophic chondrocytes, osteoblasts, and osteoprogenitor cells, which are located at the primary ossification center, secrete proangiogenic factors, that is, VEGF.14 The high levels of VEGF expression stimulate the invasion of blood vessels into the hypertrophic cartilage, which is accompanied by ossification processes.15,13 On postnatal day 1 (P1), the blood vessels sprout throughout the diaphysis. By postnatal day 6 (P6), these vessels invade the epiphyseal chondrocytes at the two distal ends of the long bone and initiate secondary ossification center formation. Also, it has been reported that distinguishable metaphyseal and diaphyseal capillary networks have been formed.13 Finally, on postnatal day 21 (P21), the vascular and bone elements are established (Fig. 2).16

FIG. 2.

FIG. 2.

An illustration of the role of microvasculature in bone development. MSCs aggregate and differentiate into chondrocytes to form a cartilage at ∼E13. The hypertrophic chondrocytes in the POC serve as template and stimulate angiogenesis at ∼E15. Blood vessels extend toward the epiphysis at P1 and subsequently invade into them to form the SOCs at P6. Bone development and blood vessel formation are built at P21. MSCs, mesenchymal stromal cells; OBs, osteoblasts; POC, primary ossification center; SOCs, secondary ossification centers. Color images are available online.

These osteogenic and angiogenic mechanisms that occur during bone development also take place in adult bone repair under normal cases.17,18 If these events are blocked or impaired due to injury, bone may not be having the regeneration capacity to be healed by itself. Overall, a better understanding of how vasculature participates in bone development may facilitate the detection of osteogenic and angiogenic molecular targets, which may be applied to enhance adult bone repair under disease conditions.

Role of Vasculature in Bone Diseases

Bone has a substantial capacity to be self-repaired in response to injury.19 During healing process, vasculature plays a significant role to restore bone tissue function.20,21 It actively participates in the four stages of bone healing process, named (i) inflammation and hematoma, (ii) cartilaginous callus formation and periosteal response, (iii) bony callus, and (iv) bone formation and remodeling.22,23 Specifically, upon tissue damage, bone microvasculature is disrupted and a hematoma is formed around the damaged site.24 The inflammatory cascade is activated by releasing growth factors and cytokines, and by recruiting osteoprogenitor cells and endothelial precursor cells. This initial reaction stimulates the formation of new vascular networks, while macrophages and monocyte-derived osteoclasts remove necrotic bone tissue.5 During the initial soft callus phase, the fracture callus remains avascular. However, as the chondrocytes mature and become hypertrophic, they secrete proangiogenic factors, which stimulate vascular ingrowth from vessel in the periosteum.25 Next, during the bony callus formation, the cartilage matrix is calcified. Specifically, chondrocytes undergo apoptosis, osteoclasts reabsorb the cartilage, mesenchymal stem cells differentiate to osteoblasts, and neovessels are formed. These events initiate the remodeling process which reestablishes the vascularized osteon structures.19,21

Bone diseases are often accompanied by alterations or disruptions of bone microvasculature during bone healing process. Improper neovessel formation and loss of microvascular function during bone repair process may lead to disease development.26–29 Although some diseases such as osteosarcoma and Paget's disease are caused by excessive vascularization, most pathogenic conditions, including fractures, avascular necrosis (osteonecrosis), osteopetrosis, osteoporosis, and osteoarthritis (OA), are related to inadequate microvascular functionality. Below, we report the role of vasculature on these diseases and state existing substantial treatments and surgical procedures that are being widely used.

Large bone defects, nonunions, and bone contusions

Large bone defects (critical-size defects) caused by trauma, congenital condition, or tumor resection often require complicated reconstructive approaches. Bone cannot be healed spontaneously, since it presents limited regeneration capacity due to inhibition of re-ossification and neovascularization.30,31 Common treatments for large bone defects require surgical operations of autologous, allologous, xenologous, or synthetic bone grafts. These approaches present limitations due to laborious procedures, high risk for infections and rejection by the immune system.6,10,32 Also, it has been shown that during the postsurgical restorative period, complications may appear, such as nonunions.33

Nonunions are considered the interruption of reparative process of bone healing.34 Major reason for these nonunions is the absence of proper vascular network to supply the fracture site with oxygen and nutrients. Several factors, such as age, nutrition, and systematic diseases (i.e., diabetes mellitus, anemia, and endocrine disorders) increase the risk of nonunions. Surgical treatment approaches for nonunions require removal of dead bone or poorly vascularized tissue from the fracture site.35,36 Recently, the use of pedicled vascular grafts for nonunion treatments has been demonstrated.37

Although large defects and nonunions require complicated reconstructive surgical approaches, there is no proper treatment for lower extremity injuries, such as bone contusions. Repeated forces and mechanical stress damage bone and vasculature without causing an overt fracture. When trauma occurs, the blood vessels are widened and stagnate the flow underneath of periosteal bone, causing subperiosteal hematoma. Similarly, loss of vasculature functionality in bone medulla can cause internal bleeding and the formation of interosseous bruise.28,38–42 Existing therapeutics for bone contusions and microfractures are conservative, and include rest, refraining from the injury-inducing activity, possible immobilization, ice, and anti-inflammatory medication. Finally, physical therapy is, also, recommended to improve healing and prevent injury reoccurrence.28

Avascular necrosis

Avascular necrosis is a clinical finding with twenty thousand new diagnoses per year, which often progresses to surgical treatment, such as hip replacements.43,44 The femoral head is a common site of occurrence, resulting in significant pain and impaired mobility.45 The underlying mechanism involves disruption of vascular circulation in femoral head, tissue necrosis, and eventually, bone collapse.46 Secondary pathogenic factors involve the development of high intraosseous pressures, cell cytotoxicity, and intravascular coagulation. The risk of avascular necrosis is increased with environmental exposure in glucocorticoids, alcohol, trauma, systemic lupus erythematosus, sickle cell disease, and underlying genetic susceptibility.47 Early stages of avascular necrosis can be treated by medication, physical therapy, and joint-sparing surgical interventions.48 Although surgical procedures of femoral head replacement is necessary for later stages, they have demonstrated high failure rates.49 Recent innovative procedures implicate the use of vascularized fibular graft. This approach has shown to benefit the graft viability, induce mineralization, and promote vascularization.27,50–52

Osteopetrosis

Osteopetrosis is a rare inherited disorder. Mutations in the Chloride channel 7 (CLCN7) gene are responsible for about 75% cases of autosomal dominant osteopetrosis with mild symptoms.53 The symptoms of osteopetrosis include fracture, low production of blood cells, poor blood supply, and loss of cranial nerve system. It is characterized by thickening cortical bone region and narrowing bone marrow space. It results from osteoblast–osteoclast imbalanced function. Despite the normal osteoblast activity, the reduced functionality of osteoclast leads to the accumulation of bone deposition. Thus, osteopetrotic bones are becoming brittle with abnormal architecture and susceptible to fracture.54 In addition, obliteration of the medullary cavities causes limited perfusion to the bone, which results in the formation of necrotic site with excessive anaerobic bacteria growth. The poor blood flow restricts the response of antimicrobial agents and host immune cells to access this bacteria area.55 Despite the absence of proper treatment, recent studies demonstrate hematopoietic stem cell transplantation as curative treatment for infantile malignant osteopetrosis.29

Osteoporosis

Osteoporosis is the most common type of metabolic bone disease characterized by the loss of bone density and strength, resulting in high risks of fracture formation.56 Age, postmenopausal state, and diabetes are major risk factors associated with the development of osteoporosis.57,58 In general, osteoporosis is characterized by reduced imbalanced activity between osteoblasts and osteoclasts, resulting in abnormal bone remodeling.4,59 Also, osteoporosis is associated with bone endothelial dysfunction. Specifically, postmenopausal osteoporosis is accompanied by a decreased number of sinusoidal and arterial capillaries in the bone marrow, leading to limited perfusion. In addition, it has been reported that elderly women with osteoporosis demonstrated reduced bone blood flow in femur.60–62 Osteoporotic patients are usually treated with anabolic agents that stimulate bone formation, such as parathyroid hormone (PTH) or antiresorptive drugs that inhibit bone resorption, such as bisphosphonates.63 Finally, Vidyadhara et al. have demonstrated the use of vascularized fibular graft for the treatment of osteoporotic humeral shaft nonunions.64

Osteoarthritis

OA, known as degenerative joint disease, is the most common type of arthritis.65 The etiology of OA includes joint injury, obesity, and aging.66–68 Inflammation and injury to the joint trigger bony changes, deterioration of tendons and ligaments, and a breakdown of cartilage, resulting in pain, swelling, and deformity of the joint.65,66 Although OA is characterized by progressive degenerative damage to articular cartilage, the initiation, and progression of OA are closely associated with reduced blood flow into the small vessels of the subchondral bone.69 Blood flow may be decreased by venous occlusion and stasis or by the development of microemboli in the subchondral vessels. Thus, the articular cartilage may not be supplied with sufficient nutrients and oxygen, which induce the apoptosis of osteocytes, bone resorption, and inflammation in subchondral bone.69 Treatments for OA are pain relief medications such as acetaminophen and nonsteroidal anti-inflammatory drugs and joint replacement.70

Osteosarcoma

Osteosarcoma is the most common primary malignant bone cancer with five per million per year estimated incidence rate. Despite its rarity, osteosarcoma represents ∼4% of all childhood cancers and 56% of malignant bone tumors in children. Although it is diagnosed and defined through the observation of malignant osteoblasts, its origin remains undefined. These cells exhibited high proliferation and mineralization activity and increased bone turnover in the metaphysis.71 Animal studies have demonstrated that tumor angiogenesis process accelerate its formation, growth, and metastasis.72 Specifically, aggressive osteosarcoma cells induce formation of neovessels, facilitating tumor perfusion and metastasis to the other organs (i.e., lung) through the main bloodstream.73 Typical treatments for patients with localized osteosarcoma demonstrate high survival rate (60–70%) and involve surgical removal of the tumor, chemotherapy, and radiation therapy. However, treatment of metastatic and recurrent osteosarcoma exhibits 5-year survival rate up to 20%, since the treatment is limited without efficacy.74 Postsurgical effects of these treatments include the development of pathological fracture causing pain, immobility, and local recurrence.75 Although skeletal reconstruction following tumor resection remains challenging, the free vascularized fibular graft is a promising technique for the reconstruction of large femoral defects in children with osteosarcoma.26

Paget's disease

Paget's disease (or Osteitis Deformans) is a chronic skeletal disorder, which causes skull and lower extremity deformities. Unlike other systemic bone loss diseases, such as osteoporosis, Paget's disease affects one or several bones.76 It has been reported that mutations in the sequestosome 1 gene produce susceptibility to develop Paget's disease.77 The initial stage of Paget's disease, named osteolytic phase, is characterized by the presence of numerous hyperactive osteoclasts. The hematopoietic bone marrow is replaced by fibrous connective tissue and numerous large blood vessels. This distorted osteoclast resorption is accompanied with an irregular osteoblastic response. This excessive bone turnover rate results in a disorganized, mosaic pattern of woven and lamellar bone, which is associated with mechanical weakness and fracture formation.78 Rare complications of Paget's disease involve heart failure and the development of a malignant bone tumor such as osteosarcoma.77–79 The most common treatment for Paget's disease is the use of drugs for osteoporosis, such as bisphosphonates.80

Taken together, impaired or excessive vasculature in bone reduces bone functionality, leading to structural and movement problems of the body. Thus, promising strategies based on vascularized bone tissue engineering might enhance the bone remodeling under disease cases. Below, we report the role of stem cells and growth factors on promoting osteogenesis and angiogenesis and overall bone regeneration.

Vasculature and Bone Cells

During bone repair, stem cells and osteogenic and vasculogenic precursor cells move to the injured site. They differentiate into mature osteoblasts and endothelial cells, which are actively involved in ossification and angiogenesis processes.81,82 The mechanisms, involved in these events, require the intracellular communication of endothelial cells with other cell types, including mesenchymal stromal cells (MSCs), osteoblast precursor cells, macrophages, pericytes, and endothelial progenitor cells (EPCs) (Fig. 3).83–85 Finally, this process is supplemented by secretion of osteogenic and angiogenic growth factors, which promote angiogenesis and mineralization.86 Thus, a better understanding of these cellular events will advance the development of co-culture and growth factor approaches for enhancing bone regeneration. Next, we will report predifferentiated osteoblastic and endothelial cell sources and growth factors, which have been applied to treat bone diseases by inducing osteogenesis and vascularization.

FIG. 3.

FIG. 3.

Schematic for different cell types involved in bone regeneration. Bone and vascular cells are differentiated from MSC types, such as bone marrow (BM-MSCs), adipose (ADSCs), umbilical cord (UCMSCs), urine (USCs), periosteum (PDPCs), induced pluripotent stem cells (iPSC-MSCs), and EPCs, respectively. Macrophages, neutrophils, and monocytes are implicated in the process. BM-MSCs, bone marrow-derived mesenchymal stromal cells; ADSCs, adipose-derived stromal cells; UCMSCs, umbilical cord blood-derived mesenchymal stromal cells; USCs, urine-derived stromal cells; PDPCs, periosteum-derived progenitor cells; iPSC-MSCs, induced pluripotent stem cell-derived mesenchymal stromal cells; EPCs, endothelial progenitor cells. Color images are available online.

Mesenchymal stromal cells

MSCs are adult progenitor cells, which are lying in the bone marrow, in adipose tissue,87 and in periosteum.88,89 They have the capacity to regenerate bone, cartilage, and fat by being differentiated into osteoblasts, chondrocytes, and adipose cells, respectively.86,87,90 The MSC to osteoblast differentiation process is initiated by transcriptional regulation of runt-related transcription factor-2 (Runx2) and nuclear localization of β-catenin through canonical WNT signaling, which further activate osterix, osteopontin, and osteocalcin.91–93 Apart from their osteogenic capability, MSCs play a critical role in stimulating immune microenvironment. Specifically, they play a critical role in proliferation, migration, and differentiation of progenitor cells and macrophage polarization by secreting transforming growth factor beta-1 (TGF-β1), insulin-like growth factor-1 (IGF-1) epidermal growth factor, stem cell factor, and granulocyte and macrophage colony-stimulating factors.94,95 MSCs promote neovascularization through their communication with EPCs by releasing proangiogenic factors such as VEGF, platelet-derived growth factor (PDGF), fibroblast growth factor-2 (FGF-2), IGF-1, and angiopoietin-1 (Ang-1).96–98 These factors induce the EPC proliferation and differentiation, which in turn lead EPCs to release osteogenic growth factors that promote MSC differentiation.99,100 Their differentiation potential, their ability to secrete bioactive factors that can modulate the immune system and promote tissue repair, characterizes MSCs as unique cell source for bone regeneration applications. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) reside in native bone and they are the most common and longest utilized source for bone regeneration and repair. In addition, MSCs isolated from different anatomic locations with similar osteogenic capacity as BM-MSCs, such as adipose-derived stromal cells (ADSCs), periosteum-derived progenitor cells (PDPCs), and umbilical cord blood-derived mesenchymal stromal cells (UCMSCs), have widely been used. Apart from these cell sources, induced pluripotent stem cell (iPSC)-derived MSCs (iPSC-MSCs) is another cell source that has been used by engineers and researchers to attempt mimicking the in vivo bone microenvironment. Below, we report these MSC sources and their application in bone tissue engineering (Table 1).

Table 1.

Applications of Cell Types to Bone Regeneration by Inducing Osteogenesis and Angiogenesis

Cell Characteristic Application Refs.
BM-MSCs Isolated from bone marrow
High osteogenic capacity
Support vascularization and mineralization
To repair craniofacial defect
Effective as osteoblastic precursors
in critical-sized defect reconstruct
Transplanted into avascular necrosis
101,102,105
PDPCs Isolated from periosteum
Greater clonogenicity and differentiation capacity than BM-MSCs
Suitable to chondrogenic/osteogenic differentiation
To heal critical-sized tibial defect
To repair a distal femur atrophic nonunion
88,89,108–111
ADSCs Isolated from adipose tissue
Abundant, less painful to harvest, and easily expandable
Promising alternative cell source to BM-MSCs
Similar osteogenicity to BM-MSCs
To repair avascular necrosis of femoral head
To reconstruct the calvarial defects
To ameliorate large bone defects
112,113,115–118
UCMSCs Isolated from umbilical cord
Limited by finite supply and morbidity
No invasive method
Low immunogenicity
Differentiated toward osteogenic lineage
To heal critical-sized alveolar cleft defects
To treat avascular necrosis of the femoral head
119–123
USCs Obtained from voided urine
No invasive procedures
Promising alternative stem cell source
Similar osteogenic potential to ADSCs
Compatible with both calcium sulfate/PLGA composite and β-TCP scaffolds
To repair large bone defects
124,125
iPSC-MSCs Engineered patient-specific cells
Noninvasive
Differentiated into MSCs/ECs
Tumorigenic potential
Implant various skeletal defects
To apply critical-sized calvarial defects
126–129
EPCs Isolated from peripheral blood
Secrete VEGF
Proangiogenic features
Differentiate to ECs
Combine with MSCs to repair calvarial defects
To treat avascular necrosis of the femoral head
EPC utilization for osteopetrosis
131–138

BM-MSC, bone marrow-derived mesenchymal stromal cell; PDPC, periosteum-derived progenitor cell; ADSC, adipose-derived stromal cell; UCMSC, umbilical cord blood-derived mesenchymal stromal cell; USC, urine-derived stromal cell, iPSC-MSCs, induced pluripotent stem cell-derived mesenchymal stromal cells; EPC, endothelial progenitor cell; EC, endothelial cell; VEGF, vascular endothelial growth factor; PLGA, poly(lactic-co-glycolic acid); β-TCP, β-tricalcium phosphate.

Bone marrow-derived mesenchymal stromal cells

BM-MSCs are the most common MSC type with capacity to differentiate into bone, cartilage, and adipose lineage.86 Studies have reported their regeneration capacity to large defect bone reconstruction upon craniofacial trauma and fractures.101 Also, it has been shown that BM-MSC transplantation in avascular necrosis model promotes bone regeneration.102 Although BM-MSCs have been used in bone transplantation, major limitations have been raised. For example, after prolonged in vitro expansion, BM-MSCs inevitably obtain a senescent phenotype. Also, their osteogenic potential is decreased due to donor age and in presence of systematic diseases. These factors limit the actual amount and the quality of BM-MSCs that may be obtained for clinical application.103,104 Also, it is unclear whether their capacity to regenerate bone is resulted in their osteogenic potential or is mediated through their trophic activities.102,105 Finally, there is, as yet, no clear, report regarding the long-term safety of BM-MSCs, since their tumorigenic and profibrogenic potential have been reported as major obstacles in their therapeutic use.106,107

Periosteum-derived progenitor cells

PDPCs reside in periosteum and they act as major players in bone development and fracture healing process.8,108 Although PDPCs and BM-MSCs are derived from a common mesenchymal lineage, postnatally, PDPCs exhibit greater clonogenicity and osteogenic differentiation capacity than BM-MSCs.109 These characteristics make PDPCs suitable cell source for bone tissue engineering applications.108 Specifically, autologous PDPCs cultured on a three-dimensional matrix are responsible to promote the healing of a distal femur atrophic nonunion.110 Finally, Bolander et al. has demonstrated that the combination of implanted PDPCs with exogenous bone morphogenic protein-2 (BMP-2) induces fracture healing of a critical-size tibial defect in mice.111

Adipose-derived stromal cells

ADSCs are obtained from abundant adipose tissue by a minimally invasive procedure, which results in isolation of a high number of cells. In addition, ADSCs have shown similar osteogenic potential to BM-MSCs with certain subpopulations, demonstrating enhanced osteoblastic potential.112,113 These features make them a promising source for bone transplantation.114,115 Specifically, it has been demonstrated that direct injection of autologous ADSCs into avascular necrotic femoral head promoted the osteogenesis.116 Also, Pak et al. reported a case of avascular necrosis of the femoral head treated with ADSCs and platelet-rich plasma, providing significant improvement of the patient's severe hip pain.117 Finally, autologous ADSCs and spongy cells from the iliac crest have been transplanted to reconstruct the bone defects after severe head injury, resulting in new calvarial bone formation.118

Other stromal cell types

Apart from BM-MSCs and ADSCs, UCMSCs and urine-derived stromal cells (USCs) have been used as cell sources for bone applications. The isolation of UCMSCs from umbilical cord is an easier, less expensive, and noninvasive method than collecting MSCs from bone marrow aspirates. UCMSCs demonstrate the capacity to retain low immunogenicity effect, to be propagated in vitro, and to be differentiated toward osteogenic lineage.119,120 Specifically, UCMSCs embedded in poly(lactic-co-glycolic acid) (PLGA) scaffold have been applied for treatment of large alveolar cleft defects in a swine model.121 In addition, studies have demonstrated the allogeneic UCMSC potential to treat avascular necrosis of the femoral head. Although no obvious side-effects have been issued, extended studies with large number of patients are needed to further evaluate the efficiency and safety of allogeneic UCMSC application in treating avascular necrosis of the femoral head.122,123

Another promising stem cell source is USCs, which hold advantages of easily isolation without invasive and laborious processes, demonstrating similar osteogenic potential to ADSCs.124 These cells combined with PLGA and tricalcium phosphate composites have been applied in large bone defect repairing.124,125

Induced pluripotent stem cell-derived mesenchymal stromal cells

iPSCs are an alternative source to engineer high-quality patient-specific MSCs without the limitations of laborious isolation and propagation, which are required to obtain tissue-derived MSCs.126 The iPSC-MSCs have been characterized by the expression of major MSC markers, named CD24−/CD105+, and by their high proliferative and osteogenic differentiation potential.127 Several studies have demonstrated their application on bone regeneration. Specifically, transplantation of Runx2 genetically modified iPSCs into skeletal defects accelerates the alveolar bone regeneration and cementum and periodontal ligaments formation.128 Ye et al., demonstrate enhanced bone regeneration in critical-sized calvarial defects after transduction of special AT-rich sequence-binding protein 2 genetically modified iPSCs.129 Despite these promising studies, concerns regarding the tumorigenic capacity of iPSC-MSCs have eliminated their clinical applications. Finally, iPSCs have showed the capacity to be differentiated toward endothelial cells without the ability to generate bone-specific endothelial cells.130

Endothelial progenitor cells

EPCs are hematopoietic stem cells that reside in the bone marrow and peripheral blood. EPCs have proangiogenic features by promoting neovascularization during in vivo bone regeneration.131–135 In addition, it has been reported that EPCs in-vitro differentiate to endothelial cells and participate in vascular network formation.83 Sun et al., reported that EPC transplantation in avascular necrotic femoral head enhances the neovascularization and bone regeneration in rabbit models.136 EPCs regulate bone regeneration and angiogenesis by promoting the migratory and osteoclast differentiation potential of macrophages in vitro and in mouse fracture model through Talin-1.137 Disruption of this process, such as in osteopetrosis cases, decreases macrophage differentiation potential toward osteoclast precursor cells, resulting in production of osteoclasts with low or no resorption activity.138

Inflammatory cells: macrophages and neutrophils

Macrophages are heterogeneous immune cells and they demonstrate either proinflammatory M1 or anti-inflammatory M2 phenotype in different microenvironments.139 M1 macrophages secrete proinflammatory cytokines and participate in the early stage of fracture healing such as hematoma formation and angiogenesis. M2 macrophages contribute to bone repair by the resolution of inflammation.86 Dysfunction of macrophage polarization is associated with delays in bone regeneration.138 Neutrophils are also key factors in the systemic immune response. Their interactions with monocytes/macrophages enable the host to efficiently defend against inflammation and contribute to the early fracture hematoma.140 However, the recent studies have shown that neutrophils have a negative effect on fracture healing in animal model.141 Thus, despite their significant role in bone functionality, macrophages and neutrophils have not been directly utilized as sources for bone transplantation.

Overall, different MSC sources along with EPCs may be promising transplant sources for bone repair and regeneration by inducing neovascularization and bone formation (Fig. 3).

Angiogenic and Osteogenic Growth Factors

Growth factors are key components participating in bone repair by regulating angiogenesis and osteogenesis.22 Specifically, we report the in vitro and in vivo role of VEGF, Ang-1, Sonic hedgehog (SHH), FGF-2, PDGF, IGF-1, and BMP-2 in regulating angiogenesis, osteogenesis, and endothelial bone communication upon bone healing (Table 2).86,142,143

Table 2.

Contribution of Angiogenic and Osteogenic Growth Factors in Bone Regeneration

Growth factor Role in bone repair Angiogenic effect Osteogenic effect Intracellular communication Cell source Refs.
VEGF Hematoma formation
Cartilaginous callus
Bony callus
Bone remodeling
In vivo: promotes blood vessel formation in avascular necrosis rabbit and femoral fractures, increases vascular invasion In vivo: improves ossification in fracture mice, bone formation in nonunion and avascular necrosis rabbit, induces stem cell recruiting In vivo: paracrine VEGF secreted by osteoblasts binds to endothelial VEGF receptors
In vitro: VEGF levels are upregulated in presence of ADSC-endothelial co-culture system
EPCs/ECs
MSCs/OBs
Macrophages
Neutrophils
142–156
Ang-1 Cartilaginous callus In vivo: induces angiogenesis in fracture repair and ischemic necrosis rat
In vitro: promotes blood vessel remodeling and maturation
In vivo: ameliorates bone remodeling in ischemic necrosis rat In vitro: co-culture system of osteodifferentiating ADSCs or MSCs with ECs increase angiogenesis by secreting Ang-1 MSCs 150,160–166
SHH Callus formation In vivo: induces neovascularization by activating VEGF, Ang-1, PDGF, and TGF-β1 in ischemic mouse In vivo: increases bone regeneration in calvarial defect rabbit
In vitro: stimulates Runx2 and OCN, increases alkaline phosphatase and mineralization
In vitro: endothelial cell and MSCs co-culture systems increase mineralization capacity and the formation of capillary-like structures MSCs 167–174
FGF-2 Hematoma formation
Cartilaginous callus
In vivo: promotes vascularization in segmental defect rat
In vitro: induces neovessel formation and maturation
In vivo: increases callus formation in diabetic rat, increases callus, bone mineral density, and biomechanical stability in canine tibial fracture dog
In vitro: stimulates mitogenesis of mesenchymal cells and OBs
In vitro: endothelial-ADSCs co-culture systems demonstrated the upregulation of FGF-2 MSCs/OBs
ECs
Macrophages
150,175–185
PDGF Hematoma formation
Cartilaginous callus
In vitro: increases blood vessel formation by regulation of VEGF in ECs In vivo: increases bone density and strength, and callus in proximal tibial osteotomy rabbit, improves alveolar bone healing in periodontal lesions
In vitro: induces OBs migration, proliferation, and differentiation
In vivo: secreted by endothelial cells and stimulates the recruitment, proliferation, and differentiation of osteoblast precursor cells
In vitro: upregulated in co-cultures by promoting osteogenic differentiation and blood vessel sprouting
ECs
Macrophages
150,186–192
IGF-1 Cartilaginous callus
Bone remodeling
In vitro: increases EC migration and induces neovascularization In vivo: stimulates ossification and bone growth in mice, induces fracture repair by PTH secretion In vitro: promotes angiogenesis in endothelial-ADSC co-culture system MSCs/OBs 193–195,199–203
BMP-2 Hematoma formation
Callus formation
Bone remodeling
In vitro: promotes angiogenesis by VEGF secretion In vivo: induces bone repair in large craniofacial defects
In vitro: induces osteogenesis through Smad signaling
In vitro: co-culture systems of MSCs/ECs in presence of BMP-2 have demonstrated enhanced osteogenesis and angiogenesis MSCs/OBs 111,204–213

Ang-1, angiopoietin-1; SHH, Sonic hedgehog; FGF-2, fibroblast growth factor-2; PDGF, platelet-derived growth factor; IGF-1, insulin-like growth factor-1; BMP-2, bone morphogenic protein-2; OB, osteoblast; Runx2, Runt-related transcription factor-2; OCN, osteocalcin; TGF-β1, transforming growth factor beta-1; PTH, parathyroid hormone.

Vascular endothelial growth factor

VEGF controls endothelial cell differentiation and proliferation in bone. Upon injury, VEGF is produced by inflammatory cells and progenitor cells.144,145 The release of VEGF initiates angiogenesis within the hematoma and fibrin-rich granulation tissue begins to develop.142,146 It was shown that inhibition of VEGF impaired bone healing by disrupting conversion of the cartilaginous callus to bony callus.147 In endochondral bone formation during bone callus stage, VEGF secreted by osteoblast precursors in the perichondrium and hypertrophic chondrocytes promotes recruitment of osteoblastic cells.15 Furthermore, in vivo and in vitro studies have demonstrated that paracrine VEGF is a key molecule participating in the bone-endothelial interaction.146 Specifically, osteoprogenitor cells and osteoblasts secrete paracrine VEGF, which subsequently binds to endothelial VEGF receptors, and eventually promotes endothelial migration and tubing formation.148,149 Also, in vitro MSC and endothelial cell co-culture systems have demonstrated upregulation in VEGF levels.150,151

Given the importance of VEGF in bone repair, treatment with exogenous VEGF to damaged bone tissue area would potentially enhance angiogenesis and blood flow.147,152,153 It has been reported that VEGF improves ossification and callus maturation in a mouse femur fracture model,147 and enhances bone healing in a rabbit nonunion fracture model.154 Also, VEGF gene delivery into the femoral head necrotic area in rabbits demonstrated high bone formation 8 weeks after treatment.155 Finally, it has been reported that combination of VEGF and BMP-2 promoted blood vessel regeneration in avascular necrosis.156 Despite its significance, excessive VEGF may have reverse results, since it increases neovessel formation over osteogenesis, resulting in tumor development. Thus, anti-VEGF treatments could be an alternative efficient treatment against osteosarcoma.157–159

Angiopoietin-1

The role of Ang in fracture repair is not as well understood as the VEGF pathway. Although VEGF has a major role in blood vessel development and growth, Ang-1 induces blood vessel remodeling and maturation.160 Also, Ang-2 has double role as inducer of neovascularization in presence of VEGF or as suppressor of vascular degeneration in absence of VEGF by segregating the endothelial cells from the perivascular cells.161–163 The expression of Ang-1 is induced during the initial stages of fracture repair, suggesting that the vascular growth in periosteum plays an important role in the repair process.164 In addition, in vitro studies have demonstrated that co-culture system of osteodifferentiating ADSCs increases angiogenesis by secreting Ang-1.150,165 Finally, it has been used in surgery-induced ischemic necrosis of the femoral head rat model. Intraosseous infusion of Ang-1 ameliorates angiogenesis and bone remodeling.166

Sonic hedgehog

Hedgehog signaling is involved in fracture healing.167 In the early phase of fracture repair, the expression of SHH is detected in proliferating callus-forming cells in the periosteum.168 SHH has an important role to facilitate both angiogenesis and osteogenesis in vivo and in vitro. It is reported that hedgehog proteins directly act on osteogenic precursor cells and osteoblasts to stimulate osteogenic differentiation.169 Specifically, it stimulates the expression of Runx2 and OCN and induces alkaline phosphatase activity. Studies with endothelial cell and MSC co-culture systems demonstrated that SHH increases mineralization capacity and the formation of capillary-like structures.170–172 Also, it has been shown that it induces neovascularization and vascular stabilization in ischemic hind limb aged mouse model indirectly by stimulating VEGF, PDGF, TGF-β1, and the Ang-1/2.173 Finally, the implantation of SHH-transduced cells increased the bone regeneration in a rabbit model of calvarial defects.174

Fibroblast growth factor-2

Even though the role of FGFs is not clear in bone fracture healing process, FGF-1 and FGF-2 receptors participate in regulation of bone regeneration.175–178 FGF-2 induces endothelial cell proliferation and migration and neovessel formation and maturation.179–181 It also stimulates mitogenesis of mesenchymal progenitors and osteoblasts.182 In vitro endothelial-ADSC co-culture systems demonstrated the upregulation of FGF-2, upon osteogenic differentiation.150 Exogenous FGF-2 delivery increased the callus formation and structural stability of fibular fractures in diabetic rats.183 Also, administration of FGF-2 on hydroxyapatite graft increased vascularization 2 weeks following segmental defect in a rat.184 Finally, in a canine tibial fracture model, FGF-2 injection at the fracture site increased callus area, mineral content, and mechanical strength.185

Platelet-derived growth factor

During the inflammatory stage of bone healing process, PDGF released from degranulating platelets and macrophages.186 PDGF promotes blood vessel formation by regulation of VEGF in endothelial cells.187 It is secreted by endothelial cells and stimulates the recruitment, proliferation, and differentiation of osteoblast precursor cells.188 Also, it has been demonstrated that it was also upregulated in long-term in vitro co-cultures by promoting osteogenic differentiation and blood vessel sprouting.150 Animal studies have evaluated the role of exogenous recombinant PDGF on fracture healing.189,190 Specifically, it has been demonstrated that PDGF increased callus density and volume, and improved mechanical strength following proximal tibial osteotomy in a rabbit model.190 Finally, clinical trials have shown that recombinant human PDGF-BB combined with allograft bone matrix was delivered in periodontal lesions, resulting in accelerating alveolar bone regeneration.191,192

Insulin-like growth factor-1

Insulin-like growth factor-1 (IGF-1) plays an important role during bone healing by controlling key osteogenic and angiogenic events. Specifically, conditional IGF-1 knockout mice revealed low bone mineral density, reduction of hypertrophic chondrocytes in the growth plate area, and limited periosteal expansion. These results demonstrated that IGF-1 regulates both endochondral ossification and bone formation.193 In addition, in vivo studies showed that IGF-1 promotes fracture repair194,195 by inducing the secretion of PTH.196–198 In contrast, it has been showed that osteocyte-derived IGF-1 may have an inhibitory function in fracture healing progression.199 In vitro studies have demonstrated the key role of IGF-1 in osteogenesis and in angiogenesis formation.199–201 Specifically, IGF-1 promotes angiogenesis in endothelial-ADSC co-culture system through AKT pathway.202 Also, BM-MSCs promote EPC proliferation by secreting IGF-1 in the in vitro co-culture systems.203

Bone morphogenic protein-2

BMPs play an important role during fracture repair. They are produced by MSCs and osteoblasts and they stimulated MSC proliferation, differentiation, angiogenesis, and matrix synthesis.204,205 BMP-2 plays a role in initiating the repair cascade and is genetically associated with the maintenance of normal bone mass.206,207 It has the capacity to induce bone repair in large craniofacial defects, such as alveolar ridge.208–210 Also, β-tricalcium phosphate (β-TCP) graft combined with ADSCs and BMP-2 ameliorate large bone defect repair.211 In general, BMP-2 stimulates osteoblast differentiation through Smad signaling pathway and promotes angiogenesis by inducing VEGF secretion from osteoblasts. Similar to VEGF, BMP-2 is an important molecule participating in MSCs-EPCs crosstalk.212 Co-culture systems of MSCs/ECs in presence of BMP-2 have demonstrated enhanced osteogenesis and angiogenesis.213 Finally, BMP-7 and BMP-9 play an important role in cartilaginous callus and bony callus formation, respectively. BMP-7 promotes angiogenesis, while BMP-9 enhances in vitro and in vivo osteoblastogenesis of MSCs.32,214–221

In vivo and in vitro experiments demonstrated that osteogenic and angiogenic growth factors contribute to bone repair process. Given the importance of these factors on bone healing, the application of stem cells and exogenous growth factors may pave the way to engineer promising tissue engineering strategies in promoting bone healing.

Conclusions and Future Directions

This review demonstrated the crucial role of vasculature in bone development, regeneration, and remodeling. The functional regulation and molecular crosstalk between endothelial cells and osteoblasts play a vital role in angiogenesis and osteogenesis in normal and disease conditions. Vascularization is indispensable for bone formation or remodeling, and thus, it can be the key factor in the development of in vivo and ex vivo bone tissue engineering strategies.

Novel approaches based on bone microvasculature regeneration may be integrated into the current treatments. Specifically, bone in vitro models may integrate vascular components, which will be advantageous to understand the pathobiology of bone diseases and identify key players involved in endothelial-bone cell interaction. A better understanding of the underlying physiological mechanisms linking vascular function to bone function has the potential to develop promising personalized vascularized bone regeneration techniques. Finally, strategies that combine vascular components, growth factors, and progenitor cells on bone grafts may promote faster and proper host-graft integration, avoiding the limitations of existing bone grafts.

Overall, an understanding of bone microvasculature functionality can be utilized for the development of pharmaceutical and clinical treatments for improving or impeding bone vascularization in avascular necrosis or osteosarcoma, respectively. It also suggests that these approaches may be widely adopted to other organ diseases to promote their regeneration capacity. We anticipate that the knowledge gained from bone microvasculature may be applied to highly vascularized organs such as lung and kidney. Upon injury, diseases related to these organs, such as pulmonary arterial hypertension and acute kidney injury, demonstrated limited regeneration capacity, which is related to their impaired microvascular functionality.222,223 Thus, the tissue engineering approaches of stem cells and growth factors may be applied as potential treatments to improve tissue microvascular regeneration capacity.224,225

Acknowledgment

We thank Ruth Ann Cho for her kind assistance with illustrations in figures.

Disclosure Statement

No competing financial interests exist.

Funding Information

This work was supported by the American Dental Association Science and Research Institute and American Dental Association Foundation grant (97700142) and the National Institute of Arthritis And Musculoskeletal And Skin Diseases of the National Institutes of Health (R21AR076497).

References

  • 1.Clarke, B.Normal bone anatomy and physiology. Clin J Am Soc Nephrol 3(Suppl 3), S131, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lopes, D., Martins-Cruz, C., Oliveira, M.B., and Mano, J.F.. Bone physiology as inspiration for tissue regenerative therapies. Biomaterials 185,240, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mercado-Pagan, A.E., Stahl, A.M., Shanjani, Y., and Yang, Y.. Vascularization in bone tissue engineering constructs. Ann Biomed Eng 43,718, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Feng, X., and McDonald, J.M.. Disorders of bone remodeling. Annu Rev Pathol 6,121, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Loi, F., Cordova, L.A., Pajarinen, J., Lin, T.H., Yao, Z., and Goodman, S.B.. Inflammation, fracture and bone repair. Bone 86,119, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Oryan, A., Alidadi, S., Moshiri, A., and Maffulli, N.. Bone regenerative medicine: classic options, novel strategies, and future directions. J Orthop Surg Res 9,18, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ehrler, D.M., and Vaccaro, A.R. The use of allograft bone in lumbar spine surgery. Clin Orthop Relat Res 38, 2000 [DOI] [PubMed] [Google Scholar]
  • 8.Ho-Shui-Ling, A., Bolander, J., Rustom, L.E., Johnson, A.W., Luyten, F.P., and Picart, C.. Bone regeneration strategies: engineered scaffolds, bioactive molecules and stem cells current stage and future perspectives. Biomaterials 180,143, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Marrella, A., Lee, T.Y., Lee, D.H., et al. Engineering vascularized and innervated bone biomaterials for improved skeletal tissue regeneration. Mater Today 21,362, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nguyen, L.H., Annabi, N., Nikkhah, M., et al. Vascularized bone tissue engineering: approaches for potential improvement. Tissue Eng Part B Rev 18,363, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kronenberg, H.M.Developmental regulation of the growth plate. Nature 423,332, 2003 [DOI] [PubMed] [Google Scholar]
  • 12.Percival, C.J., and Richtsmeier, J.T.. Angiogenesis and intramembranous osteogenesis. Dev Dyn 242,909, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sivaraj, K.K., and Adams, R.H.. Blood vessel formation and function in bone. Development 143,2706, 2016 [DOI] [PubMed] [Google Scholar]
  • 14.Nakashima, K., and de Crombrugghe, B.. Transcriptional mechanisms in osteoblast differentiation and bone formation. Trends Genet 19,458, 2003 [DOI] [PubMed] [Google Scholar]
  • 15.Maes, C., Kobayashi, T., Selig, M.K., et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev Cell 19,329, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Brandi, M.L., and Collin-Osdoby, P.. Vascular biology and the skeleton. J Bone Miner Res 21,183, 2006 [DOI] [PubMed] [Google Scholar]
  • 17.Kanczler, J.M., and Oreffo, R.O.. Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cells Mater 15,100, 2008 [DOI] [PubMed] [Google Scholar]
  • 18.Yang, Y.-Q., Tan, Y.-Y., Wong, R., Wenden, A., Zhang, L.-K., and Rabie, A.B.M.. The role of vascular endothelial growth factor in ossification. Int J Oral Sci 4,64, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Walmsley, G.G., Ransom, R.C., Zielins, E.R., et al. Stem cells in bone regeneration. Stem Cell Rev Rep 12,524, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Carano, R.A., and Filvaroff, E.H.. Angiogenesis and bone repair. Drug Discov Today 8,980, 2003 [DOI] [PubMed] [Google Scholar]
  • 21.Das, A., and Botchwey, E.. Evaluation of angiogenesis and osteogenesis. Tissue Eng Part B Rev 17,403, 2011 [DOI] [PubMed] [Google Scholar]
  • 22.Ai-Aql, Z.S., Alagl, A.S., Graves, D.T., Gerstenfeld, L.C., and Einhorn, T.A.. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res 87,107, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Einhorn, T.A., and Gerstenfeld, L.C.. Fracture healing: mechanisms and interventions. Nat Rev Rheumatol 11,45, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kolar, P., Schmidt-Bleek, K., Schell, H., et al. The early fracture hematoma and its potential role in fracture healing. Tissue Eng Part B Rev 16,427, 2010 [DOI] [PubMed] [Google Scholar]
  • 25.Keramaris, N.C., Calori, G.M., Nikolaou, V.S., Schemitsch, E.H., and Giannoudis, P.V.. Fracture vascularity and bone healing: a systematic review of the role of VEGF. Injury 39(Suppl 2), S45, 2008 [DOI] [PubMed] [Google Scholar]
  • 26.Cashin, M., Coombs, C., and Torode, I.. A-frame free vascularized fibular graft and femoral lengthening for osteosarcoma pediatric patients. J Pediatr Orthop 38,e83, 2018 [DOI] [PubMed] [Google Scholar]
  • 27.Fang, T., Zhang, E.W., Sailes, F.C., McGuire, R.A., Lineaweaver, W.C., and Zhang, F.. Vascularized fibular grafts in patients with avascular necrosis of femoral head: a systematic review and meta-analysis. Arch Orthop Trauma Surg 133,1, 2013 [DOI] [PubMed] [Google Scholar]
  • 28.Patel, D.S., Roth, M., and Kapil, N.. Stress fractures: diagnosis, treatment, and prevention. Am Fam Physician 83,39, 2011 [PubMed] [Google Scholar]
  • 29.Shadur, B., Zaidman, I., NaserEddin, A., et al. Successful hematopoietic stem cell transplantation for osteopetrosis using reduced intensity conditioning. Pediatr Blood Cancer 65,e27010, 2018 [DOI] [PubMed] [Google Scholar]
  • 30.Engstrand, T., Kihlstrom, L., Neovius, E., et al. Development of a bioactive implant for repair and potential healing of cranial defects. J Neurosurg 120,273, 2014 [DOI] [PubMed] [Google Scholar]
  • 31.Schmitz, J.P., and Hollinger, J.O. The critical size defect as an experimental model for craniomandibulofacial nonunions. Clin Orthop Relat Res 299, 1986 [PubMed] [Google Scholar]
  • 32.Fishero, B.A., Kohli, N., Das, A., Christophel, J.J., and Cui, Q.. Current concepts of bone tissue engineering for craniofacial bone defect repair. Craniomaxillofac Trauma Reconstr 8,23, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stewart, S.K.Fracture non-union: a review of clinical challenges and future research needs. Malays Orthop J 13,1, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Crenshaw, A.H., Daugherty, K., and Campbell, W.C.. Campbell's Operative Orthopaedics. St. Louis, MO: Mosby Year Book, 1992 [Google Scholar]
  • 35.Ding, Z.C., Lin, Y.K., Gan, Y.K., and Tang, T.T.. Molecular pathogenesis of fracture nonunion. J Orthop Transl 14,45, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Miclau, K.R., Brazina, S.A., Bahney, C.S., et al. Stimulating fracture healing in ischemic environments: does oxygen direct stem cell fate during fracture healing? Front Cell Dev Biol 5, 45, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Karaismailoglu, B., Fatih Guven, M., Erenler, M., and Botanlioglu, H.. The use of pedicled vascularized bone grafts in the treatment of scaphoid nonunion: clinical results, graft options and indications. EFORT Open Rev 5,1, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fleming, J.L.One-year prevalence of lower extremity injuries among active duty military soldiers. Mil Med 153,476, 1988 [PubMed] [Google Scholar]
  • 39.Hauret, K.G., Jones, B.H., Bullock, S.H., Canham-Chervak, M., and Canada, S.. Musculoskeletal injuries description of an under-recognized injury problem among military personnel. Am J Prev Med 38,S61, 2010 [DOI] [PubMed] [Google Scholar]
  • 40.Kilcoyne, K.G., Dickens, J.F., and Rue, J.P.. Tibial stress fractures in an active duty population: long-term outcomes. J Surg Orthop Adv 22,50, 2013 [DOI] [PubMed] [Google Scholar]
  • 41.Milgrom, C., Giladi, M., Stein, M., et al. Stress fractures in military recruits. A prospective study showing an unusually high incidence. J Bone Joint Surg Br 67,732, 1985 [DOI] [PubMed] [Google Scholar]
  • 42.Wentz, L., Liu, P.Y., Haymes, E., and Ilich, J.Z.. Females have a greater incidence of stress fractures than males in both military and athletic populations: a systemic review. Mil Med 176,420, 2011 [DOI] [PubMed] [Google Scholar]
  • 43.Mankin, H.J.Nontraumatic necrosis of bone (osteonecrosis). N Engl J Med 326,1473, 1992 [DOI] [PubMed] [Google Scholar]
  • 44.Moya-Angeler, J., Gianakos, A.L., Villa, J.C., Ni, A., and Lane, J.M.. Current concepts on osteonecrosis of the femoral head. World J Orthop 6,590, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zizic, T.M., Marcoux, C., Hungerford, D.S., and Stevens, M.B.. The early diagnosis of ischemic necrosis of bone. Arthritis Rheum 29,1177, 1986 [DOI] [PubMed] [Google Scholar]
  • 46.Chang, C.C., Greenspan, A., and Gershwin, M.E.. Osteonecrosis: current perspectives on pathogenesis and treatment. Semin Arthritis Rheum 23,47, 1993 [DOI] [PubMed] [Google Scholar]
  • 47.Shah, K.N., Racine, J., Jones, L.C., and Aaron, R.K.. Pathophysiology and risk factors for osteonecrosis. Curr Rev Musculoskelet Med 8,201, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Urbaniak, J.R., Coogan, P.G., Gunneson, E.B., and Nunley, J.A.. Treatment of osteonecrosis of the femoral head with free vascularized fibular grafting. A long-term follow-up study of one hundred and three hips. J Bone Joint Surg Am 77,681, 1995 [DOI] [PubMed] [Google Scholar]
  • 49.Johannson, H.R., Zywiel, M.G., Marker, D.R., Jones, L.C., McGrath, M.S., and Mont, M.A.. Osteonecrosis is not a predictor of poor outcomes in primary total hip arthroplasty: a systematic literature review. Int Orthop 35,465, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gonzalez Della Valle, A., Slullitel, G., Piccaluga, F., and Salvati, E.A.. The precision and usefulness of preoperative planning for cemented and hybrid primary total hip arthroplasty. J Arthroplast 20,51, 2005 [DOI] [PubMed] [Google Scholar]
  • 51.Wang, G., Zhang, C.Q., Sun, Y., et al. Changes in femoral head blood supply and vascular endothelial growth factor in rabbits with steroid-induced osteonecrosis. J Int Med Res 38,1060, 2010 [DOI] [PubMed] [Google Scholar]
  • 52.Yoo, M.C., Kim, K.I., Hahn, C.S., and Parvizi, J.. Long-term followup of vascularized fibular grafting for femoral head necrosis. Clin Orthop Relat Res 466,1133, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Frattini, A., Pangrazio, A., Susani, L., et al. Chloride channel ClCN7 mutations are responsible for severe recessive, dominant, and intermediate osteopetrosis. J Bone Miner Res 18,1740, 2003 [DOI] [PubMed] [Google Scholar]
  • 54.Coudert, A.E., de Vernejoul, M.C., Muraca, M., and Del Fattore, A.. Osteopetrosis and its relevance for the discovery of new functions associated with the skeleton. Int J Endocrinol 2015,372156, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tolar, J., Teitelbaum, S.L., and Orchard, P.J.. Osteopetrosis. N Engl J Med 351,2839, 2004 [DOI] [PubMed] [Google Scholar]
  • 56.Kanis, J.A.Diagnosis of osteoporosis and assessment of fracture risk. Lancet 359,1929, 2002 [DOI] [PubMed] [Google Scholar]
  • 57.Hyassat, D., Alyan, T., Jaddou, H., and Ajlouni, K.M.. Prevalence and risk factors of osteoporosis among Jordanian postmenopausal women attending the national center for diabetes, endocrinology and genetics in Jordan. BioRes Open Access 6,85, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jackuliak, P., and Payer, J.. Osteoporosis, fractures, and diabetes. Int J Endocrinol 2014,820615, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kenkre, J.S., and Bassett, J.. The bone remodelling cycle. Ann Clin Biochem 55,308, 2018 [DOI] [PubMed] [Google Scholar]
  • 60.Alagiakrishnan, K., Juby, A., Hanley, D., Tymchak, W., and Sclater, A.. Role of vascular factors in osteoporosis. J Gerontol Ser A Biol Sci Med Sci 58,362, 2003 [DOI] [PubMed] [Google Scholar]
  • 61.Marenzana, M., and Arnett, T.R.. The key role of the blood supply to bone. Bone Res 1,203, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rhinelander, F.W.The normal microcirculation of diaphyseal cortex and its response to fracture. J Bone Joint Surg Am 50,784, 1968 [DOI] [PubMed] [Google Scholar]
  • 63.Black, D.M., and Rosen, C.J.. Clinical practice. Postmenopausal osteoporosis. N Engl J Med 374,254, 2016 [DOI] [PubMed] [Google Scholar]
  • 64.Vidyadhara, S., Vamsi, K., Rao, S.K., Gnanadoss, J.J., and Pandian, S.. Use of intramedullary fibular strut graft: a novel adjunct to plating in the treatment of osteoporotic humeral shaft nonunion. Int Orthop 33,1009, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chen, D., Shen, J., Zhao, W., et al. Osteoarthritis: toward a comprehensive understanding of pathological mechanism. Bone Res 5,16044, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Felson, D.T.Clinical practice. Osteoarthritis of the knee. N Engl J Med 354,841, 2006 [DOI] [PubMed] [Google Scholar]
  • 67.Loeser, R.F.Aging and osteoarthritis: the role of chondrocyte senescence and aging changes in the cartilage matrix. Osteoarthritis Cartilage 17,971, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Loeser, R.F., Goldring, S.R., Scanzello, C.R., and Goldring, M.B.. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 64,1697, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Findlay, D.M.Vascular pathology and osteoarthritis. Rheumatology 46,1763, 2007 [DOI] [PubMed] [Google Scholar]
  • 70.Mora, J.C., Przkora, R., and Cruz-Almeida, Y.. Knee osteoarthritis: pathophysiology and current treatment modalities. J Pain Res 11,2189, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Meyers, P.A., and Gorlick, R.. Osteosarcoma. Pediatr Clin North Am 44,973, 1997 [DOI] [PubMed] [Google Scholar]
  • 72.Gorlick, R., Anderson, P., Andrulis, I., et al. Biology of childhood osteogenic sarcoma and potential targets for therapeutic development: meeting summary. Clin Cancer Res 9,5442, 2003 [PubMed] [Google Scholar]
  • 73.Ando, K., Mori, K., Verrecchia, F., Marc, B., Redini, F., and Heymann, D.. Molecular alterations associated with osteosarcoma development. Sarcoma 2012,523432, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bielack, S.S., Kempf-Bielack, B., Delling, G., et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 20,776, 2002 [DOI] [PubMed] [Google Scholar]
  • 75.Scully, S.P., Ghert, M.A., Zurakowski, D., Thompson, R.C., and Gebhardt, M.C.. Pathologic fracture in osteosarcoma: prognostic importance and treatment implications. J Bone Joint Surg Am 84,49, 2002 [PubMed] [Google Scholar]
  • 76.Singer, F.R. Paget's disease of bone. In: Feingold K.R., Anawalt B., Boyce A., Chrousos G., Dungan K., Grossman A., et al., eds. Endotext. South Dartmouth MA, 2000. PMID: [Google Scholar]
  • 77.Galson, D.L., and Roodman, G.D.. Pathobiology of Paget's disease of bone. J Bone Metab 21,85, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shaker, J.L.Paget's disease of bone: a review of epidemiology, pathophysiology and management. Ther Adv Musculoskelet Dis 1,107, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hansen, M.F., Seton, M., and Merchant, A.. Osteosarcoma in Paget's disease of bone. J Bone Miner Res 21(Suppl 2), P58, 2006 [DOI] [PubMed] [Google Scholar]
  • 80.Kravets, I.Paget's disease of bone: diagnosis and treatment. Am J Med 131,1298, 2018 [DOI] [PubMed] [Google Scholar]
  • 81.McAllister, B.S., and Haghighat, K.. Bone augmentation techniques. J Periodontol 78,377, 2007 [DOI] [PubMed] [Google Scholar]
  • 82.Meijer, G.J., de Bruijn, J.D., Koole, R., and van Blitterswijk, C.A.. Cell based bone tissue engineering in jaw defects. Biomaterials 29,3053, 2008 [DOI] [PubMed] [Google Scholar]
  • 83.Asahara, T., Masuda, H., Takahashi, T., et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 85,221, 1999 [DOI] [PubMed] [Google Scholar]
  • 84.Shi, Y., Kramer, G., Schroder, A., et al. Early endothelial progenitor cells as a source of myeloid cells to improve the pre-vascularisation of bone constructs. Eur Cells Mater 27,64, 2014 [DOI] [PubMed] [Google Scholar]
  • 85.Yoder, M.C., Mead, L.E., Prater, D., et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood 109,1801, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Almubarak, S., Nethercott, H., Freeberg, M., et al. Tissue engineering strategies for promoting vascularized bone regeneration. Bone 83,197, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hass, R., Kasper, C., Bohm, S., and Jacobs, R.. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 9,12, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Duchamp de Lageneste, O., Julien, A., Abou-Khalil, R., et al. Periosteum contains skeletal stem cells with high bone regenerative potential controlled by periostin. Nat Commun 9,773, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.De Bari, C., Dell'Accio, F., Vanlauwe, J., et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum 54,1209, 2006 [DOI] [PubMed] [Google Scholar]
  • 90.Pittenger, M.F., Mackay, A.M., Beck, S.C., et al. Multilineage potential of adult human mesenchymal stem cells. Science 284,143, 1999 [DOI] [PubMed] [Google Scholar]
  • 91.Clevers, H.Wnt/beta-catenin signaling in development and disease. Cell 127,469, 2006 [DOI] [PubMed] [Google Scholar]
  • 92.Gaur, T., Lengner, C.J., Hovhannisyan, H., et al. Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem 280,33132, 2005 [DOI] [PubMed] [Google Scholar]
  • 93.Nakashima, K., Zhou, X., Kunkel, G., et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108,17, 2002 [DOI] [PubMed] [Google Scholar]
  • 94.Haynesworth, S.E., Baber, M.A., and Caplan, A.I.. Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell Physiol 166,585, 1996 [DOI] [PubMed] [Google Scholar]
  • 95.Bahney, C.S., Hu, D.P., Miclau, T., 3rd, and Marcucio, R.S.. The multifaceted role of the vasculature in endochondral fracture repair. Front Endocrinol 6,4, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gershovich, J.G., Dahlin, R.L., Kasper, F.K., and Mikos, A.G.. Enhanced osteogenesis in cocultures with human mesenchymal stem cells and endothelial cells on polymeric microfiber scaffolds. Tissue Eng Part A 19,2565, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kolbe, M., Xiang, Z., Dohle, E., Tonak, M., Kirkpatrick, C.J., and Fuchs, S.. Paracrine effects influenced by cell culture medium and consequences on microvessel-like structures in cocultures of mesenchymal stem cells and outgrowth endothelial cells. Tissue Eng Part A 17,2199, 2011 [DOI] [PubMed] [Google Scholar]
  • 98.Rehman, J., Li, J., Orschell, C.M., and March, K.L.. Peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation 107,1164, 2003 [DOI] [PubMed] [Google Scholar]
  • 99.Ern, C., Krump-Konvalinkova, V., Docheva, D., et al. Interactions of human endothelial and multipotent mesenchymal stem cells in cocultures. Open Biomed Eng J 4,190, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Guerrero, J., Catros, S., Derkaoui, S.M., et al. Cell interactions between human progenitor-derived endothelial cells and human mesenchymal stem cells in a three-dimensional macroporous polysaccharide-based scaffold promote osteogenesis. Acta Biomater 9,8200, 2013 [DOI] [PubMed] [Google Scholar]
  • 101.Ueda, M., Yamada, Y., Ozawa, R., and Okazaki, Y.. Clinical case reports of injectable tissue-engineered bone for alveolar augmentation with simultaneous implant placement. Int J Periodontics Restorative Dent 25,129, 2005 [PubMed] [Google Scholar]
  • 102.Yan, Z., Hang, D., Guo, C., and Chen, Z.. Fate of mesenchymal stem cells transplanted to osteonecrosis of femoral head. J Orthop Res 27,442, 2009 [DOI] [PubMed] [Google Scholar]
  • 103.Dimmeler, S., and Leri, A.. Aging and disease as modifiers of efficacy of cell therapy. Circ Res 102,1319, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Li, Y., Wu, Q., Wang, Y., Li, L., Bu, H., and Bao, J.. Senescence of mesenchymal stem cells (review). Int J Mol Med 39,775, 2017 [DOI] [PubMed] [Google Scholar]
  • 105.Caplan, A.I., and Dennis, J.E.. Mesenchymal stem cells as trophic mediators. J Cell Biochem 98,1076, 2006 [DOI] [PubMed] [Google Scholar]
  • 106.Qiao, B., Shui, W., Cai, L., Guo, S., and Jiang, D.. Human mesenchymal stem cells as delivery of osteoprotegerin gene: homing and therapeutic effect for osteosarcoma. Drug Des Devel Ther 9,969, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Warner, K.E., and Murt, H.A.. Economic incentives for health. Annu Rev Public Health 5,107, 1984 [DOI] [PubMed] [Google Scholar]
  • 108.Colnot, C.Skeletal cell fate decisions within periosteum and bone marrow during bone regeneration. J Bone Miner Res 24,274, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lambrechts, T., Papantoniou, I., Rice, B., Schrooten, J., Luyten, F.P., and Aerts, J.M.. Large-scale progenitor cell expansion for multiple donors in a monitored hollow fibre bioreactor. Cytotherapy 18,1219, 2016 [DOI] [PubMed] [Google Scholar]
  • 110.Funk, J.F., Matziolis, G., Krocker, D., and Perka, C. [Promotion of bone healing through clinical application of autologous periosteum derived stem cells in a case of atrophic non-union]. Z Orthop Unfall 145, 790, 2007 (Article in German) [DOI] [PubMed] [Google Scholar]
  • 111.Bolander, J., Ji, W., Leijten, J., et al. Healing of a large long-bone defect through serum-free in vitro priming of human periosteum-derived cells. Stem Cell Rep 8,758, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chung, M.T., Liu, C., Hyun, J.S., et al. CD90 (Thy-1)-positive selection enhances osteogenic capacity of human adipose-derived stromal cells. Tissue Eng Part A 19,989, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Liao, Y.H., Chang, Y.H., Sung, L.Y., et al. Osteogenic differentiation of adipose-derived stem cells and calvarial defect repair using baculovirus-mediated co-expression of BMP-2 and miR-148b. Biomaterials 35,4901, 2014 [DOI] [PubMed] [Google Scholar]
  • 114.Levi, B., James, A.W., Nelson, E.R., et al. Human adipose derived stromal cells heal critical size mouse calvarial defects. PLoS One 5,e11177, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Strem, B.M., Hicok, K.C., Zhu, M., et al. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med 54,132, 2005 [DOI] [PubMed] [Google Scholar]
  • 116.Abudusaimi, A., Aihemaitijiang, Y., Wang, Y.H., Cui, L., Maimaitiming, S., and Abulikemu, M.. Adipose-derived stem cells enhance bone regeneration in vascular necrosis of the femoral head in the rabbit. J Int Med Res 39,1852, 2011 [DOI] [PubMed] [Google Scholar]
  • 117.Pak, J., Lee, J.H., Jeon, J.H., and Lee, S.H.. Complete resolution of avascular necrosis of the human femoral head treated with adipose tissue-derived stem cells and platelet-rich plasma. J Int Med Res 42,1353, 2014 [DOI] [PubMed] [Google Scholar]
  • 118.Lendeckel, S., Jodicke, A., Christophis, P., et al. Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: case report. J Craniomaxillofac Surg 32,370, 2004 [DOI] [PubMed] [Google Scholar]
  • 119.Divya, M.S., Roshin, G.E., Divya, T.S., et al. Umbilical cord blood-derived mesenchymal stem cells consist of a unique population of progenitors co-expressing mesenchymal stem cell and neuronal markers capable of instantaneous neuronal differentiation. Stem Cell Res Ther 3,57, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Gang, E.J., Jeong, J.A., Hong, S.H., et al. Skeletal myogenic differentiation of mesenchymal stem cells isolated from human umbilical cord blood. Stem Cells 22,617, 2004 [DOI] [PubMed] [Google Scholar]
  • 121.Caballero, M., Morse, J.C., Halevi, A.E., et al. Juvenile swine surgical alveolar cleft model to test novel autologous stem cell therapies. Tissue Eng Part C Methods 21,898, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cai, J., Wu, Z., Huang, L., et al. Cotransplantation of bone marrow mononuclear cells and umbilical cord mesenchymal stem cells in avascular necrosis of the femoral head. Transplant Proc 46,151, 2014 [DOI] [PubMed] [Google Scholar]
  • 123.Chen, C., Qu, Z., Yin, X., et al. Efficacy of umbilical cord-derived mesenchymal stem cell-based therapy for osteonecrosis of the femoral head: a three-year follow-up study. Mol Med Rep 14,4209, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Guan, J., Zhang, J., Guo, S., et al. Human urine-derived stem cells can be induced into osteogenic lineage by silicate bioceramics via activation of the Wnt/beta-catenin signaling pathway. Biomaterials 55,1, 2015 [DOI] [PubMed] [Google Scholar]
  • 125.Guan, J., Zhang, J., Li, H., et al. Human urine derived stem cells in combination with beta-TCP can be applied for bone regeneration. PLoS One 10,e0125253, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhao, C., and Ikeya, M.. Generation and applications of induced pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Int 2018,9601623, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lian, Q., Zhang, Y., Zhang, J., et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation 121,1113, 2010 [DOI] [PubMed] [Google Scholar]
  • 128.Duan, X., Tu, Q., Zhang, J., et al. Application of induced pluripotent stem (iPS) cells in periodontal tissue regeneration. J Cell Physiol 226,150, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ye, J.H., Xu, Y.J., Gao, J., et al. Critical-size calvarial bone defects healing in a mouse model with silk scaffolds and SATB2-modified iPSCs. Biomaterials 32,5065, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Williams, I.M., and Wu, J.C.. Generation of endothelial cells from human pluripotent stem cells. Arterioscler Thromb Vasc Biol 39,1317, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.De Palma, M., Murdoch, C., Venneri, M.A., Naldini, L., and Lewis, C.E.. Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic implications. Trends Immunol 28,519, 2007 [DOI] [PubMed] [Google Scholar]
  • 132.Okuno, Y., Nakamura-Ishizu, A., Kishi, K., Suda, T., and Kubota, Y.. Bone marrow-derived cells serve as proangiogenic macrophages but not endothelial cells in wound healing. Blood 117,5264, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.van der Pouw Kraan, T.C., van der Laan, A.M., Piek, J.J., and Horrevoets, A.J.. Surfing the data tsunami, a bioinformatic dissection of the proangiogenic monocyte. Vascul Pharmacol 56,297, 2012 [DOI] [PubMed] [Google Scholar]
  • 134.Kudo, F.A., Nishibe, T., Nishibe, M., and Yasuda, K.. Autologous transplantation of peripheral blood endothelial progenitor cells (CD34+) for therapeutic angiogenesis in patients with critical limb ischemia. Int Angiol J 22,344, 2003 [PubMed] [Google Scholar]
  • 135.Taguchi, A., Soma, T., Tanaka, H., et al. Administration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse model. J Clin Invest 114,330, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sun, Y., Feng, Y., Zhang, C., et al. Beneficial effect of autologous transplantation of endothelial progenitor cells on steroid-induced femoral head osteonecrosis in rabbits. Cell Transplant 20,233, 2011 [DOI] [PubMed] [Google Scholar]
  • 137.Cui, Y., Fu, S., Hou, T., and Wu, X.. Endothelial progenitor cells enhance the migration and osteoclastic differentiation of bone marrow-derived macrophages in vitro and in a mouse femur fracture model through talin-1. Cell Physiol Biochem 49,555, 2018 [DOI] [PubMed] [Google Scholar]
  • 138.Slade Shantz, J.A., Yu, Y.Y., Andres, W., Miclau, T., 3rd, and Marcucio, R.. Modulation of macrophage activity during fracture repair has differential effects in young adult and elderly mice. J Orthop Trauma 28(Suppl 1), S10, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wynn, T.A., and Vannella, K.M.. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44,450, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bastian, O.W., Koenderman, L., Alblas, J., Leenen, L.P., and Blokhuis, T.J.. Neutrophils contribute to fracture healing by synthesizing fibronectin+ extracellular matrix rapidly after injury. Clin Immunol 164,78, 2016 [DOI] [PubMed] [Google Scholar]
  • 141.Chung, R., Cool, J.C., Scherer, M.A., Foster, B.K., and Xian, C.J.. Roles of neutrophil-mediated inflammatory response in the bony repair of injured growth plate cartilage in young rats. J Leukocyte Biol 80,1272, 2006 [DOI] [PubMed] [Google Scholar]
  • 142.Devescovi, V., Leonardi, E., Ciapetti, G., and Cenni, E.. Growth factors in bone repair. Chi Organi Mov 92,161, 2008 [DOI] [PubMed] [Google Scholar]
  • 143.Malizos, K.N., and Papatheodorou, L.K.. The healing potential of the periosteum molecular aspects. Injury 36(Suppl 3), S13, 2005 [DOI] [PubMed] [Google Scholar]
  • 144.Broxmeyer, H.E., Cooper, S., Li, Z.H., et al. Myeloid progenitor cell regulatory effects of vascular endothelial cell growth factor. Int J Hematol 62,203, 1995 [DOI] [PubMed] [Google Scholar]
  • 145.Goad, D.L., Rubin, J., Wang, H., Tashjian, A.H.Jr., and Patterson, C.. Enhanced expression of vascular endothelial growth factor in human SaOS-2 osteoblast-like cells and murine osteoblasts induced by insulin-like growth factor I. Endocrinology 137,2262, 1996 [DOI] [PubMed] [Google Scholar]
  • 146.Hu, K., and Olsen, B.R.. Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J Clin Invest 126,509, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Street, J., Bao, M., deGuzman, L., et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci U S A 99,9656, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Asahara, T., Takahashi, T., Masuda, H., et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J 18,3964, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Santos, M.I., and Reis, R.L.. Vascularization in bone tissue engineering: physiology, current strategies, major hurdles and future challenges. Macromol Biosci 10,12, 2010 [DOI] [PubMed] [Google Scholar]
  • 150.Genova, T., Petrillo, S., Zicola, E., et al. The crosstalk between osteodifferentiating stem cells and endothelial cells promotes angiogenesis and bone formation. Front Physiol 10,1291, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Herzog, D.P., Dohle, E., Bischoff, I., and Kirkpatrick, C.J.. Cell communication in a coculture system consisting of outgrowth endothelial cells and primary osteoblasts. BioMed Res Int 2014,320123, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Eckardt, H., Bundgaard, K.G., Christensen, K.S., Lind, M., Hansen, E.S., and Hvid, I.. Effects of locally applied vascular endothelial growth factor (VEGF) and VEGF-inhibitor to the rabbit tibia during distraction osteogenesis. J Orthop Res 21,335, 2003 [DOI] [PubMed] [Google Scholar]
  • 153.Li, R., Stewart, D.J., von Schroeder, H.P., Mackinnon, E.S., and Schemitsch, E.H.. Effect of cell-based VEGF gene therapy on healing of a segmental bone defect. J Orthop Res 27,8, 2009 [DOI] [PubMed] [Google Scholar]
  • 154.Eckardt, H., Ding, M., Lind, M., Hansen, E.S., Christensen, K.S., and Hvid, I.. Recombinant human vascular endothelial growth factor enhances bone healing in an experimental nonunion model. J Bone Joint Surg Br 87,1434, 2005 [DOI] [PubMed] [Google Scholar]
  • 155.Yang, C., Yang, S., Du, J., Li, J., Xu, W., and Xiong, Y.. Vascular endothelial growth factor gene transfection to enhance the repair of avascular necrosis of the femoral head of rabbit. Chin Med J 116,1544, 2003 [PubMed] [Google Scholar]
  • 156.Ma, X.W., Cui, D.P., and Zhao, D.W.. Vascular endothelial growth factor/bone morphogenetic protein-2 bone marrow combined modification of the mesenchymal stem cells to repair the avascular necrosis of the femoral head. Int J Clin Exp Med 8,15528, 2015 [PMC free article] [PubMed] [Google Scholar]
  • 157.Garcia, P., Pieruschka, A., Klein, M., et al. Temporal and spatial vascularization patterns of unions and nonunions: role of vascular endothelial growth factor and bone morphogenetic proteins. J Bone Joint Surg Am 94,49, 2012 [DOI] [PubMed] [Google Scholar]
  • 158.Kaigler, D., Wang, Z., Horger, K., Mooney, D.J., and Krebsbach, P.H.. VEGF scaffolds enhance angiogenesis and bone regeneration in irradiated osseous defects. J Bone Miner Res 21,735, 2006 [DOI] [PubMed] [Google Scholar]
  • 159.Peng, H., Usas, A., Olshanski, A., et al. VEGF improves, whereas sFlt1 inhibits, BMP2-induced bone formation and bone healing through modulation of angiogenesis. J Bone Miner Res 20,2017, 2005 [DOI] [PubMed] [Google Scholar]
  • 160.Fagiani, E., and Christofori, G.. Angiopoietins in angiogenesis. Cancer Lett 328,18, 2013 [DOI] [PubMed] [Google Scholar]
  • 161.Fujii, T., and Kuwano, H.. Regulation of the expression balance of angiopoietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cell Dev Biol Anim 46,487, 2010 [DOI] [PubMed] [Google Scholar]
  • 162.Holash, J., Maisonpierre, P.C., Compton, D., et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 284,1994, 1999 [DOI] [PubMed] [Google Scholar]
  • 163.Visconti, R.P., Richardson, C.D., and Sato, T.N.. Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF). Proc Natl Acad Sci U S A 99,8219, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Lehmann, W., Edgar, C.M., Wang, K., et al. Tumor necrosis factor alpha (TNF-alpha) coordinately regulates the expression of specific matrix metalloproteinases (MMPS) and angiogenic factors during fracture healing. Bone 36,300, 2005 [DOI] [PubMed] [Google Scholar]
  • 165.Pedersen, T.O., Blois, A.L., Xue, Y., et al. Osteogenic stimulatory conditions enhance growth and maturation of endothelial cell microvascular networks in culture with mesenchymal stem cells. J Tissue Eng 3,2041731412443236, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Park, B.H., Jang, K.Y., Kim, K.H., et al. COMP-angiopoietin-1 ameliorates surgery-induced ischemic necrosis of the femoral head in rats. Bone 44,886, 2009 [DOI] [PubMed] [Google Scholar]
  • 167.Takebe, H., Shalehin, N., Hosoya, A., Shimo, T., and Irie, K.. Sonic hedgehog regulates bone fracture healing. Int J Mol Sci 21,677, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Miyaji, T., Nakase, T., Iwasaki, M., et al. Expression and distribution of transcripts for sonic hedgehog in the early phase of fracture repair. Histochem Cell Biol 119,233, 2003 [DOI] [PubMed] [Google Scholar]
  • 169.Nakamura, T., Aikawa, T., Iwamoto-Enomoto, M., et al. Induction of osteogenic differentiation by hedgehog proteins. Biochem Biophys Res Commun 237,465, 1997 [DOI] [PubMed] [Google Scholar]
  • 170.Dohle, E., Fuchs, S., Kolbe, M., Hofmann, A., Schmidt, H., and Kirkpatrick, C.J.. Sonic hedgehog promotes angiogenesis and osteogenesis in a coculture system consisting of primary osteoblasts and outgrowth endothelial cells. Tissue Eng Part A 16,1235, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Dohle, E., Fuchs, S., Kolbe, M., Hofmann, A., Schmidt, H., and Kirkpatrick, C.J.. Comparative study assessing effects of sonic hedgehog and VEGF in a human co-culture model for bone vascularisation strategies. Eur Cells Mater 21,144, 2011 [DOI] [PubMed] [Google Scholar]
  • 172.Ho, J.E., Chung, E.H., Wall, S., Schaffer, D.V., and Healy, K.E.. Immobilized sonic hedgehog N-terminal signaling domain enhances differentiation of bone marrow-derived mesenchymal stem cells. J Biomed Mater Res Part A 83,1200, 2007 [DOI] [PubMed] [Google Scholar]
  • 173.Pola, R., Ling, L.E., Silver, M., et al. The morphogen Sonic hedgehog is an indirect angiogenic agent upregulating two families of angiogenic growth factors. Nat Med 7,706, 2001 [DOI] [PubMed] [Google Scholar]
  • 174.Edwards, P.C., Ruggiero, S., Fantasia, J., et al. Sonic hedgehog gene-enhanced tissue engineering for bone regeneration. Gene Ther 12,75, 2005 [DOI] [PubMed] [Google Scholar]
  • 175.Haque, T., Amako, M., Nakada, S., Lauzier, D., and Hamdy, R.C.. An immunohistochemical analysis of the temporal and spatial expression of growth factors FGF 1, 2 and 18, IGF 1 and 2, and TGFbeta1 during distraction osteogenesis. Histol Histopathol 22,119, 2007 [DOI] [PubMed] [Google Scholar]
  • 176.Pacicca, D.M., Patel, N., Lee, C., et al. Expression of angiogenic factors during distraction osteogenesis. Bone 33,889, 2003 [DOI] [PubMed] [Google Scholar]
  • 177.Schmid, G.J., Kobayashi, C., Sandell, L.J., and Ornitz, D.M.. Fibroblast growth factor expression during skeletal fracture healing in mice. Dev Dyn 238,766, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Su, N., Sun, Q., Li, C., et al. Gain-of-function mutation in FGFR3 in mice leads to decreased bone mass by affecting both osteoblastogenesis and osteoclastogenesis. Hum Mol Genet 19,1199, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Collin-Osdoby, P., Rothe, L., Bekker, S., Anderson, F., Huang, Y., and Osdoby, P.. Basic fibroblast growth factor stimulates osteoclast recruitment, development, and bone pit resorption in association with angiogenesis in vivo on the chick chorioallantoic membrane and activates isolated avian osteoclast resorption in vitro. J Bone Miner Res 17,1859, 2002 [DOI] [PubMed] [Google Scholar]
  • 180.Hayek, A., Culler, F.L., Beattie, G.M., Lopez, A.D., Cuevas, P., and Baird, A.. An in vivo model for study of the angiogenic effects of basic fibroblast growth factor. Biochem Biophys Res Commun 147,876, 1987 [DOI] [PubMed] [Google Scholar]
  • 181.Montesano, R., Vassalli, J.D., Baird, A., Guillemin, R., and Orci, L.. Basic fibroblast growth factor induces angiogenesis in vitro. Proc Natl Acad Sci U S A 83,7297, 1986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Globus, R.K., Patterson-Buckendahl, P., and Gospodarowicz, D.. Regulation of bovine bone cell proliferation by fibroblast growth factor and transforming growth factor beta. Endocrinology 123,98, 1988 [DOI] [PubMed] [Google Scholar]
  • 183.Kawaguchi, H., Kurokawa, T., Hanada, K., et al. Stimulation of fracture repair by recombinant human basic fibroblast growth factor in normal and streptozotocin-diabetic rats. Endocrinology 135,774, 1994 [DOI] [PubMed] [Google Scholar]
  • 184.Andreshak, J.L., Rabin, S.I., Patwardhan, A.G., and Wezeman, F.H.. Tibial segmental defect repair: chondrogenesis and biomechanical strength modulated by basic fibroblast growth factor. Anat Rec 248,198, 1997 [DOI] [PubMed] [Google Scholar]
  • 185.Nakamura, T., Hara, Y., Tagawa, M., et al. Recombinant human basic fibroblast growth factor accelerates fracture healing by enhancing callus remodeling in experimental dog tibial fracture. J Bone Miner Res 13,942, 1998 [DOI] [PubMed] [Google Scholar]
  • 186.Barrientos, S., Stojadinovic, O., Golinko, M.S., Brem, H., and Tomic-Canic, M.. Growth factors and cytokines in wound healing. Wound Repair Regen 16,585, 2008 [DOI] [PubMed] [Google Scholar]
  • 187.Guo, P., Hu, B., Gu, W., et al. Platelet-derived growth factor-B enhances glioma angiogenesis by stimulating vascular endothelial growth factor expression in tumor endothelia and by promoting pericyte recruitment. Am J Pathol 162,1083, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Fiedler, J., Roderer, G., Gunther, K.P., and Brenner, R.E.. BMP-2, BMP-4, and PDGF-bb stimulate chemotactic migration of primary human mesenchymal progenitor cells. J Cell Biochem 87,305, 2002 [DOI] [PubMed] [Google Scholar]
  • 189.Hollinger, J.O., Onikepe, A.O., MacKrell, J., et al. Accelerated fracture healing in the geriatric, osteoporotic rat with recombinant human platelet-derived growth factor-BB and an injectable beta-tricalcium phosphate/collagen matrix. J Orthop Res 26,83, 2008 [DOI] [PubMed] [Google Scholar]
  • 190.Nash, T.J., Howlett, C.R., Martin, C., Steele, J., Johnson, K.A., and Hicklin, D.J.. Effect of platelet-derived growth factor on tibial osteotomies in rabbits. Bone 15,203, 1994 [DOI] [PubMed] [Google Scholar]
  • 191.Camelo, M., Nevins, M.L., Schenk, R.K., Lynch, S.E., and Nevins, M.. Periodontal regeneration in human class II furcations using purified recombinant human platelet-derived growth factor-BB (rhPDGF-BB) with bone allograft. Int J Periodontics Restorative Dent 23,213, 2003 [PubMed] [Google Scholar]
  • 192.Nevins, M., Camelo, M., Nevins, M.L., Schenk, R.K., and Lynch, S.E.. Periodontal regeneration in humans using recombinant human platelet-derived growth factor-BB (rhPDGF-BB) and allogenic bone. J Periodontol 74,1282, 2003 [DOI] [PubMed] [Google Scholar]
  • 193.Sheng, M.H., Zhou, X.D., Bonewald, L.F., Baylink, D.J., and Lau, K.H.. Disruption of the insulin-like growth factor-1 gene in osteocytes impairs developmental bone growth in mice. Bone 52,133, 2013 [DOI] [PubMed] [Google Scholar]
  • 194.Meinel, L., Zoidis, E., Zapf, J., et al. Localized insulin-like growth factor I delivery to enhance new bone formation. Bone 33,660, 2003 [DOI] [PubMed] [Google Scholar]
  • 195.Schmidmaier, G., Wildemann, B., Heeger, J., et al. Improvement of fracture healing by systemic administration of growth hormone and local application of insulin-like growth factor-1 and transforming growth factor-beta1. Bone 31,165, 2002 [DOI] [PubMed] [Google Scholar]
  • 196.Ellegaard, M., Kringelbach, T., Syberg, S., et al. The effect of PTH(1–34) on fracture healing during different loading conditions. J Bone Miner Res 28,2145, 2013 [DOI] [PubMed] [Google Scholar]
  • 197.Nakajima, A., Shimoji, N., Shiomi, K., et al. Mechanisms for the enhancement of fracture healing in rats treated with intermittent low-dose human parathyroid hormone (1–34). J Bone Miner Res 17,2038, 2002 [DOI] [PubMed] [Google Scholar]
  • 198.Rhee, Y., Allen, M.R., Condon, K., et al. PTH receptor signaling in osteocytes governs periosteal bone formation and intracortical remodeling. J Bone Miner Res 26,1035, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Sheng, M.H., Lau, K.H., and Baylink, D.J.. Role of osteocyte-derived insulin-like growth factor I in developmental growth, modeling, remodeling, and regeneration of the bone. J Bone Metab 21,41, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Bach, L.A.Endothelial cells and the IGF system. J Mol Endocrinol 54,R1, 2015 [DOI] [PubMed] [Google Scholar]
  • 201.Shigematsu, S., Yamauchi, K., Nakajima, K., Iijima, S., Aizawa, T., and Hashizume, K.. IGF-1 regulates migration and angiogenesis of human endothelial cells. Endocr J 46(Suppl), S59, 1999 [DOI] [PubMed] [Google Scholar]
  • 202.Lin, S., Zhang, Q., Shao, X., et al. IGF-1 promotes angiogenesis in endothelial cells/adipose-derived stem cells co-culture system with activation of PI3K/Akt signal pathway. Cell Prolif 50,e12390, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Hou, J., Peng, X., Wang, J., et al. Mesenchymal stem cells promote endothelial progenitor cell proliferation by secreting insulinlike growth factor. Mol Med Rep 16,1502, 2017 [DOI] [PubMed] [Google Scholar]
  • 204.Reddi, A.H.Bone morphogenetic proteins: from basic science to clinical applications. J Bone Joint Surg Am 83-A(Suppl 1), S1, 2001 [DOI] [PubMed] [Google Scholar]
  • 205.Sakou, T.Bone morphogenetic proteins: from basic studies to clinical approaches. Bone 22,591, 1998 [DOI] [PubMed] [Google Scholar]
  • 206.Tsuji, K., Bandyopadhyay, A., Harfe, B.D., et al. BMP2 activity, although dispensable for bone formation, is required for the initiation of fracture healing. Nat Genet 38,1424, 2006 [DOI] [PubMed] [Google Scholar]
  • 207.Xiong, D.H., Shen, H., Zhao, L.J., et al. Robust and comprehensive analysis of 20 osteoporosis candidate genes by very high-density single-nucleotide polymorphism screen among 405 white nuclear families identified significant association and gene-gene interaction. J Bone Miner Res 21,1678, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Dickinson, B.P., Ashley, R.K., Wasson, K.L., et al. Reduced morbidity and improved healing with bone morphogenic protein-2 in older patients with alveolar cleft defects. Plast Reconstr Surg 121,209, 2008 [DOI] [PubMed] [Google Scholar]
  • 209.Hong, P., Boyd, D., Beyea, S.D., and Bezuhly, M.. Enhancement of bone consolidation in mandibular distraction osteogenesis: a contemporary review of experimental studies involving adjuvant therapies. J Plast Reconstr Aesth Surg 66,883, 2013 [DOI] [PubMed] [Google Scholar]
  • 210.Yuan, J., Cao, Y., and Liu, W.. Biomimetic scaffolds: implications for craniofacial regeneration. J Craniofac Surg 23,294, 2012 [DOI] [PubMed] [Google Scholar]
  • 211.Li, H., Dai, K., Tang, T., Zhang, X., Yan, M., and Lou, J.. Bone regeneration by implantation of adipose-derived stromal cells expressing BMP-2. Biochem Biophys Res Commun 356,836, 2007 [DOI] [PubMed] [Google Scholar]
  • 212.Raida, M., Heymann, A.C., Gunther, C., and Niederwieser, D.. Role of bone morphogenetic protein 2 in the crosstalk between endothelial progenitor cells and mesenchymal stem cells. Int J Mol Med 18,735, 2006 [DOI] [PubMed] [Google Scholar]
  • 213.Zhang, Y., Yang, W., Devit, A., and van den Beucken, J.. Efficiency of coculture with angiogenic cells or physiological BMP-2 administration on improving osteogenic differentiation and bone formation of MSCs. J Biomed Mater Res Part A 107,643, 2019 [DOI] [PubMed] [Google Scholar]
  • 214.Bostrom, M.P., and Camacho, N.P. Potential role of bone morphogenetic proteins in fracture healing. Clin Orthop Relat Res S274, 1998 [DOI] [PubMed] [Google Scholar]
  • 215.Cheng, S.L., Lou, J., Wright, N.M., Lai, C.F., Avioli, L.V., and Riew, K.D.. In vitro and in vivo induction of bone formation using a recombinant adenoviral vector carrying the human BMP-2 gene. Calcif Tissue Int 68,87, 2001 [PubMed] [Google Scholar]
  • 216.Kang, Q., Song, W.X., Luo, Q., et al. A comprehensive analysis of the dual roles of BMPs in regulating adipogenic and osteogenic differentiation of mesenchymal progenitor cells. Stem Cells Dev 18,545, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Krebsbach, P.H., Gu, K., Franceschi, R.T., and Rutherford, R.B.. Gene therapy-directed osteogenesis: BMP-7-transduced human fibroblasts form bone in vivo. Hum Gene Ther 11,1201, 2000 [DOI] [PubMed] [Google Scholar]
  • 218.Lamplot, J.D., Qin, J., Nan, G., et al. BMP9 signaling in stem cell differentiation and osteogenesis. Am J Stem Cells 2,1, 2013 [PMC free article] [PubMed] [Google Scholar]
  • 219.Luu, H.H., Song, W.X., Luo, X., et al. Distinct roles of bone morphogenetic proteins in osteogenic differentiation of mesenchymal stem cells. J Orthop Res 25,665, 2007 [DOI] [PubMed] [Google Scholar]
  • 220.Riew, K.D., Wright, N.M., Cheng, S., Avioli, L.V., and Lou, J.. Induction of bone formation using a recombinant adenoviral vector carrying the human BMP-2 gene in a rabbit spinal fusion model. Calcif Tissue Int 63,357, 1998 [DOI] [PubMed] [Google Scholar]
  • 221.Varady, P., Li, J.Z., Cunningham, M., et al. Morphologic analysis of BMP-9 gene therapy-induced osteogenesis. Hum Gene Ther 12,697, 2001 [DOI] [PubMed] [Google Scholar]
  • 222.Beers, M.F., and Morrisey, E.E.. The three R's of lung health and disease: repair, remodeling, and regeneration. J Clin Invest 121,2065, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Humphreys, B.D.Kidney injury, stem cells and regeneration. Curr Opin Nephrol Hypertens 23,25, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Farkas, L., and Kolb, M.. Vascular repair and regeneration as a therapeutic target for pulmonary arterial hypertension. Respiration 85,355, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Humphreys, B.D., and Bonventre, J.V.. Mesenchymal stem cells in acute kidney injury. Annu Rev Med 59,311, 2008 [DOI] [PubMed] [Google Scholar]

Articles from Tissue Engineering. Part B, Reviews are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES