Next Article in Journal
Breast Cancer MCF-7 Cells Acquire Heterogeneity during Successive Co-Culture with Hematopoietic and Bone Marrow-Derived Mesenchymal Stem/Stromal Cells
Next Article in Special Issue
Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882
Previous Article in Journal
MicroRNAs in Cancer and Cardiovascular Disease
Previous Article in Special Issue
Current Status and Prospects of Targeted Therapy for Osteosarcoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases

Microsurgery & Reconstruction Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(22), 3552; https://doi.org/10.3390/cells11223552
Submission received: 5 September 2022 / Revised: 16 October 2022 / Accepted: 7 November 2022 / Published: 10 November 2022
(This article belongs to the Special Issue Advances in Bone Metabolism)

Abstract

:
In the physiological condition, the skeletal system’s bone resorption and formation are in dynamic balance, called bone homeostasis. However, bone homeostasis is destroyed under pathological conditions, leading to the occurrence of bone metabolism diseases. The expression of hypoxia-inducible factor-1α (HIF-1α) is regulated by oxygen concentration. It affects energy metabolism, which plays a vital role in preventing bone metabolic diseases. This review focuses on the HIF-1α pathway and describes in detail the possible mechanism of its involvement in the regulation of bone homeostasis and angiogenesis, as well as the current experimental studies on the use of HIF-1α in the prevention of bone metabolic diseases. HIF-1α/RANKL/Notch1 pathway bidirectionally regulates the differentiation of macrophages into osteoclasts under different conditions. In addition, HIF-1α is also regulated by many factors, including hypoxia, cofactor activity, non-coding RNA, trace elements, etc. As a pivotal pathway for coupling angiogenesis and osteogenesis, HIF-1α has been widely studied in bone metabolic diseases such as bone defect, osteoporosis, osteonecrosis of the femoral head, fracture, and nonunion. The wide application of biomaterials in bone metabolism also provides a reasonable basis for the experimental study of HIF-1α in preventing bone metabolic diseases.

1. Introduction

The skeletal system is an essential part of the human body. Under physiological conditions, osteoclasts and osteoblasts play a role in maintaining bone resorption and formation in dynamic balance [1]. Bone marrow mesenchymal stem cells (BMSCs) are important precursor cells of the osteoblast line [2]. Under appropriate conditions, BMSCs can differentiate into mesenchymal-derived cell types, such as chondrocytes, osteoblasts, and adipocytes, which play a crucial role in bone remodeling [3]. Osteoclasts derived from bone marrow-derived macrophages (BMMs) clear old or damaged bone under the trigger of osteocytes while releasing growth factors from the bone matrix, thus inducing BMSCs recruitment and migration to the repair area, resulting in BMSCs differentiation into osteoblasts and formation of new bone at specific sites [4,5]. However, under the pathological conditions of estrogen deficiency, abnormal mechanical stress, and drug side effects, the homeostasis of bone formation and bone resorption is disrupted, leading to osteoporosis, fracture, nonunion, or osteonecrosis of the femoral head [2]. Therefore, the regulation of bone formation and bone resorption is the basic strategy for preventing and treating bone metabolism-related diseases, in which osteoclasts and osteoblasts play a crucial role.
Hypoxia-inducible factor (HIF-1) is a pivotal transcriptional regulator of cell response to hypoxia, which is composed of HIF-1α and HIF-1β subunits, interacting with the hypoxia-responsive element (HRE) and further regulating the expression of target genes [6]. Under the normoxic condition, the proline residue of HIF-1α protein is hydroxylated by proline hydroxylase (PHD) in the cytoplasm with the participation of cofactors oxygen (O2), ferrous iron (Fe2+), and α-ketoglutaric acid. Then, it binds to the von Hippel–Lindau protein (VHL) and is rapidly degraded by the ubiquitin-proteasome system (UPS) [7,8,9]. However, hypoxia inhibits the hydroxylation of HIF-1α and induces HIF-1α to enter the nucleus, bind to HIF-1β, and initiate the transcription of hypoxia-related genes, such as vascular endothelial growth factor (VEGF) [10,11,12,13]. The biological function of HIF-1α has been widely studied in orthopedics fields. It participates in the occurrence and development of osteosarcoma, osteoarthritis, osteoporosis, fracture, osteonecrosis, and other diseases by affecting the osteoclast differentiation of BMMs and the osteogenic differentiation of BMSCs [14,15,16,17,18,19].

2. The Relationship between HIF-1α and Osteoclasts

Ferroptosis is a new form of regulatory cell death driven by iron-dependent lipid peroxidation, which participates in osteoclast differentiation [20,21]. Under normoxic conditions, ferroptosis could be induced due to the iron-starvation response (increased transferrin receptor 1, decreased ferritin) followed by the Receptor Activator of Nuclear Factor-κB Ligand (RANKL) stimulation [22]. Under hypoxia, HIF-1α upregulation inhibits ferroptosis by preventing BMMs autophagosome formation and promotes the expression of RANKL in osteoblasts by activating Janus kinase 2 (JAK2)/Signal Transducers and Activators of Transcription 3 (STAT3) pathway, which supports BMMs differentiating into osteoclasts, leading to osteoporosis [16,21,23,24]. However, Hulley et al. [25] believe that HIF-1α mainly acts as a regulator of osteoclast-mediated bone resorption and has almost no effect on osteoclast differentiation. In the inflammatory environment of high-energy metabolism, lysine-specific demethylase 1 (LSD1) induced by RANKL in a rapamycin-dependent manner stabilizes the expression of HIF-1α, thus promoting glycolysis and leads to pathological bone resorption [26]. Such an interaction between HIF-1α and RANKL plays a vital role in osteoclast differentiation and physiological/pathological bone resorption mediated by osteoclast.
Studies have shown that HIF-1α promotes osteocyte apoptosis by activating the c-Jun N-terminal kinase (JNK)/caspase-3 pathway, stimulates surrounding surviving osteocytes to synthesize cytokines such as RANKL and VEGF, and RANKL triggers Notch1 signal pathway necessary for BMMs to differentiate into osteoclasts, thus recruiting osteoclasts to clear dead cells and initiating remodeling of the surrounding matrix [27,28,29,30,31]. On the contrary, osteoprotegerin (OPG), a RANKL secreted non-signaling decoy receptor secreted by osteoblasts, can also be upregulated by HIF-1α and inhibit osteoclast-mediated physiological and pathological bone resorption [32,33]. In addition, the activation of the HIF-1α pathway in osteoblasts also promotes interleukin 33 (IL-33) expression. Then, IL-33 upregulates miR-34a-5p in a dose-dependent manner, followed by downregulation of osteoclast formation-related genes via the RANKL/Notch1 signal pathway, such as tartrate-resistant acid phosphatase (Trap), capthekin-K, nuclear factor of activated T cells c1 (NFATc1), and C-fos [34]. In addition, HIF-1α stimulates osteoclasts secreting Cardiotrophin-1 (CT-1), a gp130-signaling cytokine of the IL-6 superfamily, inducing BMSCs differentiating into osteoblast [35,36,37,38]. Chen et al. [39] stably expressed HIF-1α by knocking out the VHL gene in osteocytes and found that it could boost the osteogenic differentiation of BMSCs and damage the osteoclast differentiation of BMMs.
These results suggest that HIF-1α plays a dual role in the regulation of osteoclasts, which may be attributed to the maturity of osteoblasts (Figure 1). Mature osteoblasts reduce the destructive effect of osteoclasts around the sites where the bone is being removed by secreting OPG. In contrast, more immature osteoblasts promote bone resorption by expressing RANKL and a small amount of OPG [32].

3. Hypoxia Affects the Expression of HIF-1α and Regulates the Osteogenic Activity of BMSCs

3.1. Hypoxia Dual-Directionally Regulates the Expression of HIF-1α, Affecting the Proliferation and Osteogenic Differentiation of BMSCs

As oxygen concentration from the lungs to organs and tissues gradually decreases, the oxygen content around BMSCs becomes very low [40,41,42]. Related studies discussed the effects of oxygen concentration and hypoxia duration on the characteristics of human BMSCs. The results showed that the senescence of BMSCs cultured under the hypoxia condition of 5% O2 was inhibited, and the efficiency of osteogenic differentiation was improved [43,44,45,46,47]. In addition, HIF-1α enhanced the proliferation and stemness of peripheral blood-derived mesenchymal stromal cells (PBMSCs) under 5% O2 hypoxia conditions, accompanied by the multidirectional differentiation potential, including chondrogenesis, osteogenesis, and adipogenesis [48]. However, the expression of HIF-1α in BMSCs decreased under 1–2% O2 hypoxia conditions, activating the Notch1 and extracellular regulated protein kinase 1/2 (ERK1/2)/p38 mitogen-activated protein kinase (MAPK) signal pathways, which inhibited the osteogenic differentiation of BMSCs [49,50,51]. Furthermore, extreme hypoxia (0.2% O2) stimulates adipocyte differentiation rather than osteogenic differentiation of BMSCs by upregulating CCAAT enhancer-binding proteins (C/EBPs) and HIF-1α [52]. However, with the prolonged duration of hypoxia, the expression of HIF-1α decreases gradually, accompanied by core binding factor α1 (Cbfα1) increases, promoting human BMSCs differentiating into chondrocytes and osteoclasts [53,54,55]. In addition, hypoxia also activates HIF-1α through epigenetic mechanisms such as histone modification, DNA methylation, and regulation of non-coding RNA-related genes [56].
TWIST, as a transcriptional inhibitor of runt-related transcription factor 2 (RUNX2), is one of the downstream targets of HIF-1α, inhibiting BMSCs osteogenic differentiation by downregulating bone morphogenetic protein 2 (BMP2) and RUNX2 [57,58,59,60,61,62]. Mechano growth factor (MGF) is a splicing variant of insulin-like growth factor 1 (IGF-1), which can be used for autocrine tissue repair. Studies have shown that it promotes the growth and osteogenic differentiation of MSCs through the Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway [63,64]. Furthermore, the MGF E peptide reversed the low expression of HIF-1α under severe hypoxia via MEK-ERK1/2 and PI3K-Akt signal pathways, promoting BMSCs proliferation and osteogenic differentiation [65]. These results suggest that hypoxia degree and duration affect HIF-1α expression, regulating the biological behavior of BMSCs (Figure 2). It is unclear whether TWIST, as a critical factor in connecting HIF-1α and osteogenic genes, results in the above contradictory results, but the mechanism of MGF E peptide reversing HIF-1α low expression induced by severe hypoxia has been elucidated.

3.2. HIF-1α Regulates Bone Homeostasis by Affecting Energy Metabolism

Hypoxia leads to the insufficient energy production of mitochondria and excessive reactive oxygen species (ROS), which damages mitochondria. The high expression of HIF-1α transforms energy metabolism from oxidative phosphorylation to glycolysis, reduces ROS production, and enhances mitochondria’s tolerance to hypoxia injury, suggesting that hypoxia plays a regulatory role in energy metabolism by inducing adaptive high expression of HIF-1α [66,67]. HIF-1α is associated with various diseases by affecting the glycolysis pathways of tumors, inflammation, and immune cells and regulating biological processes such as cell proliferation, differentiation, migration, chemotaxis, phagocytosis, and apoptosis.
Studies have shown that energy metabolic genes, including glucose transporter 1 (GLUT1), pyruvate dehydrogenase kinase 1 (PDK-1), lactate dehydrogenase (LDH), and monocarboxylate transporter-4 (MCT-4), upregulate in a hypoxia environment [68,69]. HIF-1α promotes glycolysis in the bone marrow microenvironment by upregulating the key enzyme PDK1 and then induces osteoblast differentiation, stimulating bone formation [70,71]. The enhancement of glycolysis in VHL-deficient osteoblasts increases bone mass, suggesting that HIF-1α affects bone homeostasis by regulating energy metabolism [72]. In addition, HIF-1α up-regulates in the early stage of BMSCs differentiation, promoting glycolysis to provide energy for BMSCs proliferation, but significantly downregulates in the late stage of BMSCs differentiation, enhancing oxidative phosphorylation in both osteogenic and chondrogenic-induced BMSCs [73,74,75]. These results somewhat explain the opposite effects of HIF-1α in bone energy metabolism in different studies. Some studies have pointed out that HIF-1α inhibits load-induced bone formation by activating the AKT/mammalian target of rapamycin (mTOR) pathway and inhibiting the Wnt/β-catenin pathway, changing osteoblast and osteocyte sensitivity to mechanical signals, which plays a negative regulatory role in the anabolism induced by bone mechanical load [71,76,77,78].

4. Inactivation of PHD/VHL Stabilizes HIF-1α, Promoting Angiogenesis and BMSCs Osteogenic Differentiation

4.1. PHD

Cobalt ions (Co2+) inhibit HIF-1α degradation by inactivating PHD and preventing VHL proline residues from being bound to HIF-1α [79,80]. Therefore, Co2+ stabilizes HIF-1α expression and upregulates VEGF by inducing ROS production and activating HRE [81]. Co2+ stimulates STAT3 phosphorylation and HIF-1α expression in BMSCs, inducing BMSCs migration to the bone repair zone [82,83]. Moreover, Co2+ also downregulates proliferation genes in BMSCs, including cyclin D1, while upregulating HIF-1α target genes, including erythropoietin (EPO), VEGF, and p21 [84]. Based on these studies, Co2+, as a chemical inducer of HIF-1α, plays a regulatory role in BMSCs migration and proliferation. Furthermore, Co2+ promotes osteogenesis and angiogenesis by stabilizing HIF-1α, which supports its application and development in the basic research of bone metabolism-related diseases [83,85,86,87].
Deferoxamine (DFO), a chelating agent, has been widely used in the clinical treatment of iron overload diseases [88,89,90]. Like Co2+, DFO chelates with non-protein bound iron and inhibits Fe2+-dependent PHD activity to simulate hypoxia, thus inhibiting the hydroxylation and degradation of HIF-1α [91,92,93]. DFO reduces ROS production in mitochondria and cell apoptosis in a HIF-1α dependent manner and promotes tubular formation, proliferation, and migration of endothelial cells (ECs) by promoting VEGF expression [94,95,96,97,98,99,100]. Short-term DFO treatment significantly changes the BMSCs glycolysis pathway, reduces mitochondrial oxidative stress, and upregulates nuclear protein1 (NUPR1) in a concentration-dependent manner, promoting BMSCs survival and cell protective autophagy [91]. In addition, DFO also affects histone acetylation and DNA methylation-related genes in osteoblasts through epigenetic mechanisms [101]. Previous studies have shown that DFO reduced the adverse effects of radiation on angiogenesis and distraction osteogenesis [102,103]. To go a step further, DFO promotes mineralization, angiogenesis, and osteogenic differentiation of BMSCs by stabilizing and enhancing HIF-1α [104]. As a HIF-1α stabilizer, DFO has been widely studied in promoting osteoblast activity and inhibiting osteoclast differentiation. However, its clinical application in bone metabolism diseases needs to be further explored [105,106,107,108,109,110].
Ethyl 3,4-dihydroxybenzoate (EDHB) is a small molecule antioxidant, which inhibits PHD activity, stabilizes HIF-1α, and promotes angiogenesis by upregulating VEGF, thus preventing steroid-induced osteonecrosis of the femoral head [18,111]. As a PHD inhibitor, Roxadustat promotes bone formation and inhibits bone resorption by activating HIF-1α and Wnt/β-catenin signal pathways, improving bone microstructure deterioration and reducing bone loss in ovariectomized rats [16,112]. IOX2 and dimethyloxalylglycine (DMOG), as inhibitors of PHD, reduce the degradation of HIF-1α, regulate the expression of downstream target genes of HIF-1α, prompting angiogenesis and the proliferation, migration, and osteogenic differentiation of BMSCs, as well as inhibiting osteoblast apoptosis [17,113,114,115,116,117,118]. However, the long-term duration activation of HIF-1α by DMOG and baicalein inhibits the osteogenic differentiation and enhances angiogenesis of adipose-derived stem cells (ASCs), while the stem cell characteristics are preserved, which still shows superiority in repairing rat skull defects [119,120] (Figure 3).

4.2. VHL

VHL affects HIF-1α expression by binding and degrading the hydroxylated HIF-1α protein. Inactivated VHL gene stimulates osteochondral progenitor cells to differentiate to osteoblasts via upregulating HIF-1α/VEGF and Smad1/5/8 signal pathways in newborn mice, accompanied by increased differentiation markers Runx2 and osteocalcin (OCN), showing significant cancellous bone accumulation, microvascular density, and bone formation increase [121]. BMP9, expressed in the developing mouse liver, induces MSCs to express HIF-1α through the Smad1/5/8 signal pathway and plays a synergistic role with HIF-1α in the process of inducing osteogenic differentiation and angiogenesis [122,123,124,125,126]. These results suggest that VHL inactivation may upregulate HIF-1α by inducing the Smad1/5/8 signal pathway, which may be a new mechanism by which VHL participates in HIF-1α regulation (Figure 3).
Selective knockout of the VHL gene up-regulates HIF-1α in mouse osteoblasts. It promotes bone marrow angiogenesis, showing a significant increase in trabecular volume, which effectively reverses bone loss caused by estrogen deficiency and promotes long bone formation with high bone density and rich blood supply [127,128,129]. Zuo et al. [130] found that the cortical bone area of VHL-deficient mice increased significantly, which was attributed to the proliferation and osteogenic differentiation of BMSCs promoted by the VHL/HIF-1α/β-catenin pathway. In addition, VHL deficiency of osteocytes increases bone mass and bone marrow hematopoiesis by regulating HIF-1α/Wnt signal pathway [131]. These studies suggest that VHL/HIF-1α pathway is a pivotal mediator in regulating angiogenesis and osteogenic differentiation of BMSCs. In addition, VHL also plays a vital role in regulating bone morphogenesis. Although the absence of VHL does not change the differentiation direction of mesenchymal progenitor cells into chondrocytes during development, it damages the proliferation ability of chondrocytes, resulting in the structural collapse of the growth plate and affecting bone morphogenesis [132,133].

5. HIF-1α/VEGF Pathway Regulates Type H Vessels, Coupling of Angiogenesis and Osteogenesis

5.1. HIF-1α/VEGF Pathway Connects Type H ECs and Osteoblasts in the Skeletal System

As the target gene of HIF-1α, VEGF plays an essential role in the regulation of angiogenesis [10,11,12,13,134,135]. Studies have shown that HIF-1α/VEGF pathway is involved in tumor immunity, inflammation, ischemia-reperfusion injury, oxidative stress, and other angiogenesis or vascular remodeling pathophysiological processes [136,137,138,139,140,141,142,143]. The mechanism of the HIF-1α/VEGF pathway involved in the regulation of bone homeostasis has also been widely studied, including angiogenesis–osteogenesis coupling. Under the mutual regulation of angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), HIF-1α stimulates BMSCs to secrete VEGF and inhibits the expression of tissue inhibitor of metalloproteinase-3 (TIMP-3), an endogenous competitive inhibitor of VEGF receptor, which mediates osteogenesis and angiogenesis. This mutual regulation relationship is known as the “HIF-1α-VEGF-Ang-1 axis” [144,145,146,147,148].
Type H ECs are a subtype of vascular endothelial cell with high expression of CD31 and Endomucin (EMCN) in the metaphyseal, distributing many bone progenitor cells around, which can differentiate into osteoblasts and osteocytes [149,150]. Two consecutive studies in Nature by the same team in 2014 found that Type H ECs mediate local vascular growth via HIF-1α/VEGF and provide clear signals for perivascular osteoblasts through the Notch signal pathway [150,151]. HIF-1α is highly expressed in Type H ECs of young rats, decreasing with aging, and is related to age-dependent bone loss, which can be reversed by DFO [152]. EC-specific inactivated VHL gene enhances Type H vessels angiogenesis and increases the number of bone progenitor cells by stabilizing HIF-1α in ECs [8]. In addition, hypoxia signals increase the number of Type H vessels and enhance endochondral angiogenesis and osteogenesis [150]. Osteostatin (OST), mainly expressed in bone marrow, induces BMSCs osteogenic differentiation and promotes the proliferation, migration, and angiogenesis of Type H ECs via the HIF-1α/VEGF pathway under hypoxia conditions [153,154]. These results suggest that HIF-1α/VEGF pathway regulates angiogenesis and osteogenesis by connecting Type H ECs and osteoblasts in the skeletal system (Figure 4).

5.2. HIF-1α/VEGF Pathway Regulates Type H Vessels in Various Bone Metabolic Disease Models

Studies have shown that the number of Type H vessels in the long bone of aging mice decreased significantly, accompanied by bone progenitor cells decrease. The osteoporosis model of ovariectomized mice showed the same phenotype [152]. Local injection of tetramethylpyrazine into the bone marrow of aging mice directly induces the formation of Type H vessels. It improves bone homeostasis by regulating the AMPK/mTOR and HIF-1α/VEGF signal pathways, preventing and treating glucocorticoid-induced osteoporosis [155]. In addition, miR-210 significantly promotes HIF-1α and VEGF expression in BMSCs dose-and-time-dependently, upregulates alkaline phosphatase (ALP) and Osterix, inhibits peroxisome proliferators-activated receptors (PPARγ), and induces BMSCs differentiating into osteoblasts, improving osteoporosis caused by estrogen deficiency [156]. Similarly, miR-497~195 cluster improves senile osteoporosis by inducing Type H vessel angiogenesis coupling with osteogenesis via Notch and HIF-1α pathways [157]. In addition, matrix metalloproteinase-2 inhibitor 1 (MMP2-I1) could induce BMSCs osteogenesis differentiation and promote Type H vessel angiogenesis through the HIF-1α signal pathway, rescuing osteonecrosis, bone defect, as well as osteoporosis [158,159] (Figure 4).
Figure 4. HIF-1α couples angiogenesis and osteogenesis. HIF-1α/VEGF and Notch signal pathways are pivotal in coupling Type H vessels angiogenesis and BMSCs osteogenic differentiation, in which matrix metalloproteinase-2 inhibitor 1 (MMP2-I1) and Osteostatin (OST) play a positive regulatory role. In addition, the HIF-1α/VEGF pathway also promotes alkaline phosphatase (ALP) and Osterix, inhibits peroxisome proliferators-activated receptors (PPARγ), promotes BMSCs osteogenic differentiation, and participates in the occurrence of osteoporosis, osteonecrosis, and bone defect.
Figure 4. HIF-1α couples angiogenesis and osteogenesis. HIF-1α/VEGF and Notch signal pathways are pivotal in coupling Type H vessels angiogenesis and BMSCs osteogenic differentiation, in which matrix metalloproteinase-2 inhibitor 1 (MMP2-I1) and Osteostatin (OST) play a positive regulatory role. In addition, the HIF-1α/VEGF pathway also promotes alkaline phosphatase (ALP) and Osterix, inhibits peroxisome proliferators-activated receptors (PPARγ), promotes BMSCs osteogenic differentiation, and participates in the occurrence of osteoporosis, osteonecrosis, and bone defect.
Cells 11 03552 g004
Low-level laser therapy (LLLT) has a positive photobiological stimulation effect on cell proliferation, angiogenesis, osteogenic differentiation, bone regeneration, and fracture healing [160,161,162]. The relevant evidence shows that laser irradiation changes the mitochondrial membrane potential, promotes the oxidation of ferrous iron by inducing ROS production, and then inhibits HIF-1α degradation by inactivating the PHD, accompanied by VEGF and transforming growth factor-β (TGF-β) expression upregulation, promoting Type H vessel formation and BMSCs osteogenic differentiation, coupling angiogenesis and osteogenesis [163,164,165] (Figure 4).

6. The Role of Non-Coding RNA (ncRNA) in HIF-1α Regulating Bone Homeostasis

6.1. Interaction between miRNA and HIF-1α Promotes Angiogenesis and BMSCs Osteogenic Differentiation, Preventing Bone Metabolism Diseases

MicroRNA (miRNA) is a group of single-stranded endogenous ncRNA with a length of 20–24 nucleotides. It plays a role in maintaining bone homeostasis by regulating angiogenesis and the BMSCs differentiation direction and may be used as a diagnostic marker for bone metabolism diseases [166]. Studies have shown that miRNA-21 promotes BMSCs expressing the osteogenic protein dose-dependently, including BMP-2, Runx2, OCN, and OPN, which may be related to phosphate and tension homology deleted on chromosome ten (PTEN)/PI3K/Akt/HIF-1α pathway [167]. MiRNA-675-5p upregulates Wnt/β-catenin and HIF-1α/VEGF signal pathways under hypoxia conditions, stimulating MSCs stemness markers (CD44, CD90, and CD73) downregulation and early osteoblast markers (ALP and COL1A1) upregulation, and then induces MSCs osteogenic and chondrogenic differentiation and promotes angiogenesis [15,168]. Low-intensity pulsed ultrasound (LIPUS) upregulates miR-31-5p, then promotes HIF-1α expression and Ras Homolog Family Member A (RhoA) activation through both VHL-dependent and VHL-independent pathways, upregulating RUNX-2, ALP, OCN, and secreted phosphoprotein 1 (SPP1), inducing human MSCs migration and promoting angiogenesis and bone regeneration [169]. Under hypoxia conditions, miR-210-3p in MSCs-derived extracellular vesicles (EVs) was regulated by HIF-1α, which blocked tyrosine kinase ligand ephrin-A3 (EFNA3) expression, then significantly promoted the angiogenesis by activating the PI3K/AKT pathway [23]. In addition, miR-26a-5p in EVs derived from urine-derived stem cells (USCs) activates HIF-1α/VEGF pathway by inhibiting histone deacetylase 4 (HDAC4), then promotes the differentiation of osteoblast progenitor cells and inhibits the activity of osteoclasts, thus preventing the occurrence of diabetic osteoporosis [170] (Figure 3).

6.2. Interaction between lncRNA and HIF-1α Regulates Apoptosis, Angiogenesis, and BMSCs Osteogenic Differentiation Involved in Bone Development and Regeneration

Long non-coding RNA (lncRNA) plays a vital role in cell proliferation, invasion, metabolism, apoptosis, and stem cell differentiation. LncRNA participates in bone development and regeneration by regulating apoptosis, angiogenesis, and BMSCs differentiation [171]. The gene sequencing results showed that 95 lncRNAs were differentially expressed in osteoclasts knocked out of HIF-1α, suggesting that lncRNA may play a role in bone homeostasis regulation by HIF-1α [172]. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), as a highly conserved lncRNA, induces angiogenesis through the mTOR/HIF-1α pathway [173]. Enhancer of zeste homolog 2 (EZH2) is a catalytic subunit of histone methyltransferase, which catalyzes the histone H3 lysine 27 trimethylation (H3K27me3) [174]. Under hypoxia conditions, HIF-1α binds to the HRE of the EZH2 promoter, then suppresses the expression of LncRNA-Tmem235 and downregulates miR-34a-3p/baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5), resulting in anoxic apoptosis of BMSCs [175]. In addition, TGF-β in BMSCs induces deacetylase sirtuin 1 (SIRT1) overexpression, downregulates lncRNA-HIF-1α-AS1 and blocks homeobox (HOX) D10 expression to interfere with acetylation, thus inhibiting osteoblast differentiation [176] (Figure 3).

7. HIF-1α Involvement in the Bone Repair of Biomaterials

7.1. Biomaterials Doped with Trace Elements Upregulate HIF-1α, Promoting Angiogenesis and Osteogenesis

7.1.1. Copper

Biomaterials doped with trace elements as scaffolds have attracted much attention in bone regeneration. The most common trace elements are copper (Cu), lithium (Li), cobalt (Co), zinc (Zn), magnesium (Mg), titanium (Ti), silicon (Si), etc. Studies have shown that continuous release of Cu2+ and Li2+ from organic or inorganic scaffolds causes crosstalk between Wnt and HIF-1α signal pathways, coupling angiogenesis and BMSCs osteogenic differentiation [177]. Hydroxyapatite (HA), as a commonly used inorganic biomaterial, loaded with co-overexpressed osteogenic factor semaphoring 3A (Sema3A) and HIF-1α modified MSCs, promotes osteogenic differentiation [178,179]. Furthermore, Nano-HA stimulates ECs to produce HIF-1α dose-dependently and upregulate osteogenic genes via the ERK1/2 signal pathway, increasing mineralized nodules and ALP [180]. Cu-Li-doped Nano-HA (Cu-Li-nHA) upregulates HIF-1α and stromal cell-derived factor-1 (SDF-1) by steadily releasing Cu2+, then promotes angiogenesis and induces BMSCs homing to necrotic areas, stimulating BMSCs to differentiate into osteoblasts through Wnt/GSK-3β/β-catenin pathway [181,182,183].
Titanium dioxide (TiO2) coating containing Cu not only improves biomedical implants’ antibacterial ability but also promotes HIF-1α/VEGF and osteogenic markers expression in BMSCs, thus achieving antibacterial properties, osteogenesis, and angiogenesis [184]. Water-soluble graphene oxide-Cu nanocomposite (GO-Cu) stably releases Cu2+, activating the ERK1/2 signal pathway and upregulating HIF-1α/VEGF and BMP-2 in BMSCs [185]. Furthermore, porous calcium phosphate (CaP) scaffold coated on GO-Cu significantly promoted angiogenesis and osteogenesis, repairing the bone defect of the rat skull. Collagen-coated Cu-doped calcium polyphosphate (CCPP) scaffolds stably release Cu2+ and collagen coating, induce BMSCs proliferation and migration to the bone defect area, then upregulate HIF-1α/VEGF and osteogenesis-related genes, stimulating BMSCs osteogenic differentiation [186].
PH-neutral bioactive glass (PSC) induces angiogenesis and BMSCs osteogenic differentiation through the PI3K/AKT/HIF-1α pathway. Compared with traditional 45S5 bioactive glass (BG) and β-tricalcium phosphate (β-TCP), PSC has more advantages in promoting BMSCs proliferation, migration, mineralization, and angiogenesis [187]. Cu-containing BG (Cu-BG) not only supports BMSCs attachment and expansion but also continuously activates HIF-1α and tumor necrosis factor-α (TNF-α) by stably releasing Cu2+, promoting downstream vascular-related growth factors and inflammation-related factors expression, directly inducing BMSCs differentiating into osteoblasts [188,189].

7.1.2. Cobalt

Co2+ stabilizes HIF-1α by inactivating PHD in a normoxic environment, commonly used as a hypoxia inducer [80]. It has been confirmed that BMSCs treated with Co2+ promote osteogenesis and angiogenesis in bone defect areas [85]. Co-doped HA significantly upregulates HIF-1α and VEGF in MG-63 cells, then promotes cell cycle progression and proliferation and induces osteocyte differentiation [190]. HA as a prosthesis coating triggers survival osteogenic gene signals, rescues the inhibition of osteoblasts and osteoclasts activity caused by Co2+ and chromium (Cr) ions concentration elevation around the prosthesis through regulating the HIF-1α signal pathway and endocytic/cytoskeletal gene [191]. Co-doped BG scaffolds with different mesoporous or chemical modifications of the upregulation of VEGF, HIF-1α, and osteogenic genes in BMSCs, promote angiogenesis and osteogenic differentiation, as well as inhibit chondrogenic differentiation [86,192,193]. The hydrogel fiber scaffold composed of collagen and alginate achieves a more substantial bone repair by carrying Co2+ and BMP2 [194]. In addition, combined organic and inorganic biomaterials doped with Co2+, such as collagen glycosaminoglycan (CG)-BG and calcium alginate (CA)-Nano-HA, also have an excellent ability to promote bone tissue regeneration [195,196].

7.1.3. Other Trace Elements

Zn commonly presents in all tissues, body fluids, and organs of the human body—more than 95% of which are present in cells and involved in the transcription of DNA [197]. Under 1% O2 conditions, Zn2+ inhibits HIF-1α expression, promoting migration and proliferation and delaying the senescence of BMSCs [198]. Ti scaffolds coated on Mg significantly stimulate the proliferation, adhesion, extracellular matrix mineralization, and ALP activity of MC3T3-E1 cells via the JAK1/STAT1/HIF-1α pathway, as well as improve the proliferation, adhesion, tubular formation, scratch healing and Transwell ability of human umbilical vein endothelial cells (HUVECs) through HIF-1α/VEGF pathway [199,200]. Zn/Mg co-implanted Ti scaffold (Zn/Mg-PIII) upregulates magnesium transporter 1 (MAGT1) in HUVECs, promotes Mg2+ influx, and activates the HIF-1α/VEGF pathway, inducing angiogenesis. In addition, Zn/Mg-PIII upregulates integrin α1 and integrin β1 to promote rat BMSCs adhesion and spread, promoting Runx2, ALP, and OCN expression in BMSCs by inducing Zn2+ and Mg2+ to recruit into cells. Compared with traditional Ti scaffolds, the Zn/Mg-PIII enhanced bone formation, angiogenesis, and antibacterial activity [201]. The ions released from Si-based biomaterials upregulate HIF-1α. The porous TiO2 coating doped with a small amount of Si significantly enhances HUVECs angiogenesis, while excessive Si doping will impair the vascular response [202]. Monte et al. [203] confirmed that 0.5mM, as an appropriate concentration of Si4+, enhanced HUVECs viability by alleviating oxidative stress damage and promoted HUVECs proliferation, migration, and angiogenesis through upregulating HIF-1α. Furthermore, mesoporous silica nanoparticles (MSN) loaded with platelet-derived growth factor BB (PDGF-BB) significantly stimulate the “HIF-1α-VEGF-Ang-1 axis” and up-regulate osteogenesis-related genes in BMSCs, inducing osteogenesis and angiogenesis [204].
These results suggest that suitable biomaterials induce BMSCs migration and attachment to the bone repair zone by stably releasing trace elements such as Cu2+ and Co2+ and upregulate HIF-1α/VEGF and osteogenic genes, coupling angiogenesis and osteogenesis, achieving bone regeneration (Figure 5).

7.2. Biomaterials Loaded with PHD Inhibitors Stabilize HIF-1α, Promoting Bone Defect Repair

7.2.1. DFO

As an iron chelating agent, DFO stabilizes HIF-1α by inhibiting PHD activity. Poly (lactic-co-glycolic acid) (PLGA) loaded with DFO significantly stimulates angiogenesis and BMSCs osteogenic differentiation, promoting osteoporotic bone defect repair [205]. DFO also effectively induces bone regeneration in true bone ceramic (TBC) scaffolds, promoting angiogenesis and segmental bone defect repair [206,207]. Titania nanotube (TNT) diameter dependently enhances BMSCs proliferation and mineralization ability. As a drug carrier loaded with DFO, TNT promotes angiogenesis and BMSCs osteogenic gene expression by activating the HIF-1α signal pathway by continuously and stably releasing DFO, then stimulates BMSCs adhesion and proliferation and affecting HUVECs growth behavior [61,208]. DFO also significantly improves the surface roughness and hydrophilicity of the polydopamine (PDOPA) membrane, which is more conducive to the attachment, proliferation, and spread of MC3T3-E1 cells and HUVECs, thus achieving bone repair [209] (Figure 5).

7.2.2. DMOG

DMOG, as an inhibitor of PHD, reduces HIF-1α degradation. β-TCP loaded with DMOG significantly enhances the angiogenic activity of BMSCs, thus repairing skull defects [210]. Implantation of ASCs on porous β-TCP-alginate-gelatin scaffolds containing DMOG induces angiogenesis and bone repair in rat skulls [211]. MSN slowly releases Si4+ to activate ALP, OCN, RUNX2, and OPN expression in human BMSCs and to stimulate BMSCs osteogenic differentiation, while MSN loaded with DMOG upregulates angiogenic genes in BMSCs by stabilizing HIF-1α, coupling osteogenesis and angiogenesis, which play a role in repairing bone defects [212]. Similarly, mesoporous bioactive glass (MBG) doped with DMOG significantly promotes osteogenesis and angiogenesis [213,214,215]. In addition, different mesoporous MBGs loaded with DMOG stabilize HIF-1α continuously by controlling DMOG release, promoting VEGF and osteogenic gene expression in BMSCs, which achieves more obvious angiogenesis and osteogenic effect [216] (Figure 5).

7.3. Application of Tissue Engineering Combined with Gene Mutation Technique to HIF-1α in Bone Defects Repair

Angiogenic and osteogenic gene upregulation in normoxic conditions can be achieved by transplanting HIF-1α-modified BMSCs into bone defect areas [217]. Studies have shown that gene mutation technology effectively improves HIF-1α activity, promoting the application in bone metabolic diseases. Compared with wild-type HIF-1α, the EVs derived from BMSCs modified by mutant HIF-1α achieve a better osteogenic and angiogenic ability, thus rescuing steroid-induced osteonecrosis of the femoral head [218]. Furthermore, β-TCP scaffold loaded the rat BMSCs-derived EVs carrying mutant HIF-1α promotes angiogenesis and BMSCs proliferation and osteogenic differentiation [219]. Ca-Mg phosphate cement (CMPC) scaffold carrying the structural active form (CA5) of HIF-1αachieves higher osteogenic activity in repairing bone defects [220,221]. The gelatin sponge (GS) loaded the BMSCs transfecting CA5 upregulates angiogenic genes in BMSCs, promoting angiogenesis in the bone defect area [222]. These results suggest that tissue engineering combined with gene mutation effectively promotes bone regeneration and may be used as an effective treatment for bone metabolism diseases.

8. The Role of Other Factors in Bone Homeostasis Regulation by HIF-1α

Toll-like receptor 2 (TLR2) is essential in regulating the immune response. Previous studies have shown that TLR2 activation promotes tissue angiogenesis and wound healing [223]. Furthermore, TLR2 significantly enhances HIF-1α and BMP-2 expression in BMSCs, upregulating the downstream osteogenic and angiogenic genes [224]. Epidermal growth factor (EGF) participates in proliferation, differentiation, adhesion, and survival processes. As a ligand of EGF, betacellulin (BTC) stabilizes HIF-1α, stimulating osteogenic progenitor cell proliferation while inhibiting differentiation [225,226]. As a demethylase, fat mass and obesity-associated protein (FTO) regulates the balance between adipogenic and osteogenic differentiation of BMSCs [227]. However, FTO induces BMSCs osteogenic differentiation by upregulating HIF-1α under mechanical stress conditions [228].
Icariin (ICA) is the main active component of Epimedium, which induces BMSCs migration by activating the SDF-1α/HIF-1α/CXCR4 pathway [229]. Safflower yellow (SY), as the main component of the traditional Chinese medicine safflower, promotes blood circulation and removes blood stasis [230]. In addition, SY also plays a pivotal role in osteogenesis and angiogenesis via the VHL/HIF-1α/VEGF signal pathway [231]. Curcumin, which exists in turmeric, significantly rescues hypoxia and reoxygenation (H/R) damage of BMSC through inhibiting mitochondrial ROS accumulation, which may be related to HIF-1α instability, the exchange protein activated by cAMP-1 (Epac1) and Akt activation and ERK1/2 and p38 inactivation [232]. Salidroside (SAL), as the main bioactive component of Rhodiola, significantly promotes the proliferation, migration, and angiogenesis of HUVECs through the HIF-1α/VEGF pathway and induces EC germination from the metatarsal [233]. Statins are traditional lipid-lowering drugs that promote fracture healing and bone defect repair [234,235,236]. Furthermore, simvastatin may induce BMSCs and endothelial progenitor cell migration via the HIF-1α/BMP-2 pathway, promoting bone defect healing [237].

9. Conclusions

Currently, the clinical treatment of bone metabolic diseases includes surgery and drug therapy. Bone transplantation in treating bone defects, joint prosthesis replacement in treating osteonecrosis of the femoral head, and plates and screws in treating fractures have achieved good results. However, it is difficult for drugs to reverse osteoporosis and prevent complications. Therefore, it is urgent to study the mechanism of bone metabolic disorder and implement effective intervention measures. With the effects of HIF-1α having been widely studied, its role in angiogenesis and bone homeostasis regulation has aroused the interest of researchers. HIF-1α regulates angiogenesis and osteoclasts and osteoblasts differentiation in various mechanisms, in which the epigenetic regulation of ncRNA also plays an important role. Based on the beneficial effects of HIF-1α coupling angiogenesis and osteogenesis and the application of biomaterials in bone metabolic diseases, new scaffold materials loaded with trace elements or drugs involved in the HIF-1α pathway may reach a better bone regeneration effect. Although HIF-1α has been widely studied in bone metabolism, there are still some problems to be explored. The bidirectional regulation of hypoxia on HIF-1α expression and the effect of HIF-1α on osteoclast differentiation have not been fully elucidated. It is necessary to study the mechanism of these opposite effects further and explore the effectiveness of HIF-1α in preventing bone metabolic diseases, laying a foundation for further clinical application.

Author Contributions

Conceptualization, L.Q. and J.T.; data curation, G.L.; writing—original draft preparation, W.C.; writing—review and editing, W.C., P.W. and L.Q.; supervision, F.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the General project of the National Natural Science Foundation of China, grant number 81871577.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Palumbo, C.; Ferretti, M. The Osteocyte: From “Prisoner” to “Orchestrator”. J. Funct. Morphol. Kinesiol. 2021, 6, 28. [Google Scholar] [CrossRef] [PubMed]
  2. Zhou, X.; Cao, H.; Guo, J.; Yuan, Y.; Ni, G. Effects of BMSC-Derived EVs on Bone Metabolism. Pharmaceutics 2022, 14, 1012. [Google Scholar] [CrossRef] [PubMed]
  3. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage Potential of Adult Human Mesenchymal Stem Cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Liu, M.; Sun, Y.; Zhang, Q. Emerging Role of Extracellular Vesicles in Bone Remodeling. J. Dent. Res. 2018, 97, 859–868. [Google Scholar] [CrossRef] [PubMed]
  5. Zhou, X.; Cao, H.; Yuan, Y.; Wu, W. Biochemical Signals Mediate the Crosstalk between Cartilage and Bone in Osteoarthritis. BioMed. Res. Int. 2020, 2020, 5720360. [Google Scholar] [CrossRef] [PubMed]
  6. Heyman, S.N.; Rosen, S.; Rosenberger, C. Hypoxia-inducible factors and the prevention of acute organ injury. Crit. Care 2011, 15, 209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Maxwell, P.H.; Wiesener, M.S.; Chang, G.-W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.; Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275. [Google Scholar] [CrossRef]
  8. Jaakkola, P.; Mole, D.R.; Tian, Y.-M.; Wilson, M.I.; Gielbert, J.; Gaskell, S.J.; von Kriegsheim, A.; Hebestreit, H.F.; Mukherji, M.; Schofield, C.J.; et al. Targeting of HIF-alpha to the von Hippel-Lindau Ubiquitylation Complex by O2-Regulated Prolyl Hydroxylation. Science 2001, 292, 468–472. [Google Scholar] [CrossRef]
  9. Ohh, M.; Park, C.W.; Ivan, M.; Hoffman, M.A.; Kim, T.Y.; Huang, L.E.; Pavletich, N.; Chau, V.; Kaelin, W.G. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat. Cell Biol. 2000, 2, 423–427. [Google Scholar] [CrossRef]
  10. Semenza, G.L. HIF-1 and mechanisms of hypoxia sensing. Curr. Opin. Cell Biol. 2001, 13, 167–171. [Google Scholar] [CrossRef]
  11. Campochiaro, P.A. Ocular neovascularization. J. Mol. Med. 2013, 9, 311–321. [Google Scholar] [CrossRef]
  12. Park, S.-Y.; Jang, W.-J.; Yi, E.-Y.; Jang, J.-Y.; Jung, Y.; Jeong, J.-W.; Kim, Y.-J. Melatonin suppresses tumor angiogenesis by inhibiting HIF-1alpha stabilization under hypoxia. J. Pineal Res. 2010, 48, 178–184. [Google Scholar] [CrossRef]
  13. Wu, Y.-C.; Chang, C.-Y.; Kao, A.; Hsi, B.; Lee, S.-H.; Chen, Y.-H.; Wang, I.-J. Hypoxia-Induced Retinal Neovascularization in Zebrafish Embryos: A Potential Model of Retinopathy of Prematurity. PLoS ONE 2015, 10, e0126750. [Google Scholar] [CrossRef] [Green Version]
  14. Ouyang, Y.; Li, H.; Bu, J.; Li, X.; Chen, Z.; Xiao, T. Hypoxia-inducible factor-1 expression predicts osteosarcoma patients’ survival: A meta-analysis. Int. J. Biol. Markers 2016, 31, e229–e234. [Google Scholar] [CrossRef]
  15. Zhang, F.-J.; Luo, W.; Lei, G.-H. Role of HIF-1α and HIF-2α in osteoarthritis. Jt. Bone Spine 2015, 82, 144–147. [Google Scholar] [CrossRef]
  16. Fayed, H.A.; Barakat, B.M.; Elshaer, S.S.; Abdel-Naim, A.B.; Menze, E.T. Antiosteoporotic activities of isoquercitrin in ovariec-tomized rats: Role of inhibiting hypoxia inducible factor-1 alpha. Eur. J. Pharmacol. 2019, 865, 172785. [Google Scholar] [CrossRef]
  17. Chen, C.; Yan, S.; Geng, Z.; Wang, Z. Fracture repair by IOX2: Regulation of the hypoxia inducible factor-1α signaling path-way and BMSCs. Eur. J. Pharmacol. 2022, 921, 174864. [Google Scholar] [CrossRef]
  18. Li, D.; Hu, Q.; Tan, G.; Xie, X.; Yang, Z.; Kang, P. Erythropoietin Enhances Bone Repair Effects via the Hypoxia-Inducible Factor Signal Pathway in Glucocorticoid-Induced Osteonecrosis of the Femoral Head. Am. J. Med. Sci. 2018, 355, 597–606. [Google Scholar] [CrossRef]
  19. Park, I.-H.; Kim, K.-H.; Choi, H.-K.; Shim, J.-S.; Whang, S.-Y.; Hahn, S.J.; Kwon, O.-J.; Oh, I.-H. Constitutive stabilization of hypoxia-inducible factor alpha selectively promotes the self-renewal of mesenchymal progenitors and maintains mesenchymal stromal cells in an undifferentiated state. Exp. Mol. Med. 2013, 45, e44. [Google Scholar] [CrossRef] [Green Version]
  20. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  21. Arnett, T.R.; Gibbons, D.C.; Utting, J.C.; Orriss, I.R.; Hoebertz, A.; Rosendaal, M.; Meghji, S. Hypoxia is a major stimulator of osteoclast formation and bone resorption. J. Cell. Physiol. 2003, 196, 2–8. [Google Scholar] [CrossRef] [PubMed]
  22. Ni, S.; Yuan, Y.; Qian, Z.; Zhong, Z.; Lv, T.; Kuang, Y.; Yu, B. Hypoxia inhibits RANKL-induced ferritinophagy and protects osteoclasts from ferroptosis. Free Radic. Biol. Med. 2021, 169, 271–282. [Google Scholar] [CrossRef]
  23. Zhuang, Y.; Cheng, M.; Li, M.; Cui, J.; Huang, J.; Zhang, C.; Si, J.; Lin, K.; Yu, H. Small extracellular vesicles derived from hypoxic mesenchymal stem cells promote vascularized bone regeneration through the miR-210-3p/EFNA3/PI3K pathway. Acta Biomater. 2022, 150, 413–426. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, J.; Tang, Y.; Wu, Q.; Ji, Y.-C.; Feng, Z.-F.; Kang, F.-W. HIF-1α facilitates osteocyte-mediated osteoclastogenesis by activating JAK2/STAT3 pathway in vitro. J. Cell. Physiol. 2019, 234, 21182–21192. [Google Scholar] [CrossRef] [PubMed]
  25. Hulley, P.A.; Bishop, T.; Vernet, A.; Schneider, J.E.; Edwards, J.R.; Athanasou, N.A.; Knowles, H.J. Hypoxia-inducible factor 1-alpha does not regulate osteoclastogenesis but enhances bone resorption activity via prolyl-4-hydroxylase 2. J. Pathol. 2017, 242, 322–333. [Google Scholar] [CrossRef] [Green Version]
  26. Doi, K.; Murata, K.; Ito, S.; Suzuki, A.; Terao, C.; Ishie, S.; Umemoto, A.; Murotani, Y.; Nishitani, K.; Yoshitomi, H.; et al. Role of Lysine-Specific Demethylase 1 in Metabolically Integrating Osteoclast Differentiation and Inflammatory Bone Resorption Through Hypoxia-Inducible Factor 1α and E2F1. Arthritis Rheumatol. 2022, 74, 948–960. [Google Scholar] [CrossRef]
  27. Song, X.; Tang, Y.; Zhu, J.; Tian, Y.; Song, Z.; Hu, X.; Hong, C.; Cai, Y.; Kang, F. HIF-1α induces hypoxic apoptosis of MLO-Y4 osteocytes via JNK/caspase-3 pathway and the apoptotic-osteocyte-mediated osteoclastogenesis in vitro. Tissue Cell 2020, 67, 101402. [Google Scholar] [CrossRef]
  28. Cheung, W.Y.; Fritton, J.C.; Morgan, S.A.; Seref-Ferlengez, Z.; Basta-Pljakic, J.; Thi, M.M.; Suadicani, S.O.; Spray, D.C.; Majeska, R.J.; Schaffler, M.B. Pannexin-1 and P2X7-Receptor Are Required for Apoptotic Osteocytes in Fatigued Bone to Trigger RANKL Production in Neighboring Bystander Osteocytes. J. Bone Miner. Res. 2016, 31, 890–899. [Google Scholar] [CrossRef] [Green Version]
  29. Bondar, C.; Ormazabal, M.; Crivaro, A.; Ferreyra-Compagnucci, M.; Delpino, M.V.; Rozenfeld, P.A.; Mucci, J.M. Osteocyte Al-terations Induce Osteoclastogenesis in an In Vitro Model of Gaucher Disease. Int. J. Mol. Sci. 2017, 18, 112. [Google Scholar] [CrossRef] [Green Version]
  30. Boyce, B.F. Advances in the Regulation of Osteoclasts and Osteoclast Functions. J. Dent. Res. 2013, 92, 860–867. [Google Scholar] [CrossRef]
  31. Choi, Y.-H.; Ann, E.-J.; Yoon, J.-H.; Mo, J.-S.; Kim, M.-Y.; Park, H.-S. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) enhances osteoclast differentiation via the up-regulation of Notch1 protein stability. Biochim. Biophys. Acta 2013, 1833, 69–79. [Google Scholar] [CrossRef] [Green Version]
  32. Boyce, B.F.; Xing, L.; Chen, D. Osteoprotegerin, the bone protector, is a surprising target for beta-catenin signaling. Cell Metab. 2005, 2, 344–345. [Google Scholar] [CrossRef] [Green Version]
  33. Shao, J.; Zhang, Y.; Yang, T.; Qi, J.; Zhang, L.; Deng, L. HIF-1α disturbs osteoblasts and osteoclasts coupling in bone remodeling by up-regulating OPG expression. In Vitro Cell. Dev. Biol.-Anim. 2015, 51, 808–814. [Google Scholar] [CrossRef]
  34. Kang, H.; Yang, K.; Xiao, L.; Guo, L.; Guo, C.; Yan, Y.; Qi, J.; Wang, F.; Ryffel, B.; Li, C.; et al. Osteoblast Hypoxia-Inducible Factor-1α Pathway Activation Restrains Osteoclastogenesis via the Interleukin-33-MicroRNA-34a-Notch1 Pathway. Front. Immunol. 2017, 8, 1312. [Google Scholar] [CrossRef] [Green Version]
  35. Walker, E.C.; McGregor, N.E.; Poulton, I.J.; Pompolo, S.; Allan, E.H.; Quinn, J.M.W.; Gillespie, M.T.; Martin, T.J.; Sims, N.A. Car-diotrophin-1 is an osteoclast-derived stimulus of bone formation required for normal bone remodeling. J. Bone Miner. Res. 2008, 23, 2025–2032. [Google Scholar] [CrossRef]
  36. Richards, C.D.; Langdon, C.; Deschamps, P.; Pennica, D.; Shaughnessy, S.G. Stimulation of osteoclast differentiation in vitro by mouse oncostatin M, leukaemia inhibitory factor, cardiotrophin-1 and interleukin 6: Synergy with dexamethasone. Cytokine 2000, 12, 613–621. [Google Scholar] [CrossRef]
  37. Walker, E.C.; McGregor, N.E.; Poulton, I.J.; Solano, M.; Pompolo, S.; Fernandes, T.J.; Constable, M.J.; Nicholson, G.C.; Zhang, J.-G.; Nicola, N.A.; et al. Oncostatin M promotes bone formation independently of resorption when signaling through leukemia inhibitory factor receptor in mice. J. Clin. Investig. 2010, 120, 582–592. [Google Scholar] [CrossRef] [Green Version]
  38. Tian, Y.; Shao, Q.; Tang, Y.; Li, X.; Qi, X.; Jiang, R.; Liang, Y.; Kang, F. HIF-1α regulates osteoclast activation and mediates osteogenesis during mandibular bone repair via CT-1. Oral Dis. 2020, 28, 428–441. [Google Scholar] [CrossRef]
  39. Chen, K.; Zhao, J.; Qiu, M.; Zhang, L.; Yang, K.; Chang, L.; Jia, P.; Qi, J.; Deng, L.; Li, C. Osteocytic HIF-1α Pathway Manipulates Bone Micro-structure and Remodeling via Regulating Osteocyte Terminal Differentiation. Front. Cell Dev. Biol. 2022, 9, 721561. [Google Scholar] [CrossRef]
  40. Pasarica, M.; Sereda, O.R.; Redman, L.M.; Albarado, D.C.; Hymel, D.T.; Roan, L.E.; Rood, J.C.; Burk, D.H.; Smith, S.R. Reduced adipose tissue oxygenation in human obesity: Evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 2009, 58, 718–725. [Google Scholar] [CrossRef]
  41. Yamaguchi, T.; Kawamoto, E.; Gaowa, A.; Park, E.J.; Shimaoka, M. Remodeling of Bone Marrow Niches and Roles of Exosomes in Leukemia. Int. J. Mol. Sci. 2021, 22, 1881. [Google Scholar] [CrossRef] [PubMed]
  42. Adolfsson, E.; Helenius, G.; Friberg, Ö.; Samano, N.; Frøbert, O.; Johansson, K. Bone marrow- and adipose tissue-derived mes-enchymal stem cells from donors with coronary artery disease; growth, yield, gene expression and the effect of oxygen con-centration. Scand. J. Clin. Lab. Investig. 2020, 80, 318–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kwon, S.Y.; Chun, S.Y.; Ha, Y.-S.; Kim, D.H.; Kim, J.; Song, P.H.; Kim, H.T.; Yoo, E.S.; Kim, B.S.; Kwon, T.G. Hypoxia Enhances Cell Properties of Human Mesenchymal Stem Cells. Tissue Eng. Regen. Med. 2017, 14, 595–604. [Google Scholar] [CrossRef] [PubMed]
  44. Weijers, E.M.; Van Den Broek, L.J.; Waaijman, T.; Van Hinsbergh, V.W.M.; Gibbs, S.; Koolwijk, P. The Influence of Hypoxia and Fibrinogen Variants on the Expansion and Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cells. Tissue Eng. Part A 2011, 17, 2675–2685. [Google Scholar] [CrossRef] [PubMed]
  45. Santos, F.D.; Andrade, P.Z.; Boura, J.S.; Abecasis, M.M.; da Silva, C.L.; Cabral, J.M.S. Ex vivo expansion of human mesen-chymal stem cells: A more effective cell proliferation kinetics and metabolism under hypoxia. J. Cell. Physiol. 2010, 223, 27–35. [Google Scholar]
  46. Pattappa, G.; Thorpe, S.D.; Jegard, N.C.; Heywood, H.K.; de Bruijn, J.D.; Lee, D.A. Continuous and uninterrupted oxygen ten-sion influences the colony formation and oxidative metabolism of human mesenchymal stem cells. Tissue Eng Part C Methods 2013, 19, 68–79. [Google Scholar] [CrossRef] [Green Version]
  47. Yasui, Y.; Chijimatsu, R.; Hart, D.A.; Koizumi, K.; Sugita, N.; Shimomura, K.; Myoui, A.; Yoshikawa, H.; Nakamura, N. Prepara-tion of Scaffold-Free Tissue-Engineered Constructs Derived from Human Synovial Mesenchymal Stem Cells Under Low Oxy-gen Tension Enhances Their Chondrogenic Differentiation Capacity. Tissue Eng. Part A 2016, 22, 490–500. [Google Scholar] [CrossRef]
  48. Wang, P.; Zhu, P.; Yu, C.; Wu, J. The Proliferation and Stemness of Peripheral Blood-Derived Mesenchymal Stromal Cells Were Enhanced by Hypoxia. Front. Endocrinol. 2022, 13, 873662. [Google Scholar] [CrossRef]
  49. Yang, M.; Liu, H.; Wang, Y.; Wu, G.; Qiu, S.; Liu, C.; Tan, Z.; Guo, J.; Zhu, L. Hypoxia reduces the osteogenic differentiation of peripheral blood mesenchymal stem cells by upregulating Notch-1 expression. Connect. Tissue Res. 2019, 60, 583–596. [Google Scholar] [CrossRef]
  50. Antebi, B.; Ii, L.A.R.; Walker, K.P.; Asher, A.M.; Kamucheka, R.M.; Alvarado, L.; Mohammadipoor, A.; Cancio, L.C. Short-term physiological hypoxia potentiates the therapeutic function of mesenchymal stem cells. Stem Cell Res. Ther. 2018, 9, 265. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, P.; Ha, N.; Dai, Q.; Zhou, S.; Yu, C.; Jiang, L. Hypoxia suppresses osteogenesis of bone mesenchymal stem cells via the extracellular signal-regulated 1/2 and p38-mitogen activated protein kinase signaling pathways. Mol. Med. Rep. 2017, 16, 5515–5522. [Google Scholar] [CrossRef]
  52. Jiang, C.; Sun, J.; Dai, Y.; Cao, P.; Zhang, L.; Peng, S.; Zhou, Y.; Li, G.; Tang, J.; Xiang, J. HIF-1A and C/EBPs transcriptionally reg-ulate adipogenic differentiation of bone marrow-derived MSCs in hypoxia. Stem Cell Res. Ther. 2015, 6, 21. [Google Scholar] [CrossRef] [Green Version]
  53. Rabie, A.M.; Tang, G.H.; Hägg, U. Cbfa1 couples chondrocytes maturation and endochondral ossification in rat mandibular condylar cartilage. Arch. Oral Biol. 2003, 49, 109–118. [Google Scholar] [CrossRef] [PubMed]
  54. Augello, A.; de Bari, C. The regulation of differentiation in mesenchymal stem cells. Hum. Gene Ther. 2010, 21, 1226–1238. [Google Scholar] [CrossRef]
  55. Huang, J.; Deng, F.; Wang, L.; Xiang, X.R.; Zhou, W.W.; Hu, N.; Xu, L. Hypoxia induces osteogenesis-related activities and ex-pression of core binding factor alpha1 in mesenchymal stem cells. Tohoku J. Exp. Med. 2011, 224, 7–12. [Google Scholar] [CrossRef] [Green Version]
  56. Kindrick, J.; Mole, D. Hypoxic Regulation of Gene Transcription and Chromatin: Cause and Effect. Int. J. Mol. Sci. 2020, 21, 8320. [Google Scholar] [CrossRef]
  57. Chen, Z.F.; Behringer, R.R. Twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev. 1995, 9, 686–699. [Google Scholar] [CrossRef] [Green Version]
  58. Isenmann, S.; Arthur, A.; Zannettino, A.C.; Turner, J.L.; Shi, S.; Glackin, C.A.; Gronthos, S. TWIST Family of Basic Helix-Loop-Helix Transcription Factors Mediate Human Mesenchymal Stem Cell Growth and Commitment. Stem Cells 2009, 27, 2457–2468. [Google Scholar] [CrossRef]
  59. Yang, D.-C.; Yang, M.-H.; Tsai, C.-C.; Huang, T.-F.; Chen, Y.-H.; Hung, S.-C. Hypoxia Inhibits Osteogenesis in Human Mesenchymal Stem Cells through Direct Regulation of RUNX2 by TWIST. PLoS ONE 2011, 6, e23965. [Google Scholar] [CrossRef] [Green Version]
  60. Yu, H.; Yu, W.; Liu, Y.; Yuan, X.; Yuan, R.; Guo, Q. Expression of HIF-1α in cycling stretch-induced osteogenic differentiation of bone mesenchymal stem cells. Mol. Med. Rep. 2019, 20, 4489–4498. [Google Scholar] [CrossRef] [Green Version]
  61. Meng, Y.; Chen, J.; Wu, X. Effect of Hypoxia-Inducible Factor-1α on Osteogenesis of Titanium Dioxide Nanotube Bone Marrow Mesenchymal Stem Cells with Different Diameters Under Periodic Tensile Stress. J. Biomed. Nanotechnol. 2022, 18, 112–119. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.; Huang, X.; Yu, H.; Yang, J.; Li, Y.; Yuan, X.; Guo, Q. HIF-1α-TWIST pathway restrains cyclic mechanical stretch-induced osteogenic differentiation of bone marrow mesenchymal stem cells. Connect. Tissue Res. 2019, 60, 544–554. [Google Scholar] [CrossRef] [PubMed]
  63. Hill, M.; Goldspink, G. Expression and splicing of the insulin-like growth factor gene in rodent muscle is associated with mus-cle satellite (stem) cell activation following local tissue damage. J. Physiol. 2003, 549, 409–418. [Google Scholar] [CrossRef] [PubMed]
  64. Tong, Y.; Feng, W.; Wu, Y.; Lv, H.; Jia, Y.; Jiang, D. Mechano-growth factor accelerates the proliferation and osteogenic differen-tiation of rabbit mesenchymal stem cells through the PI3K/AKT pathway. BMC Biochem. 2015, 16, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sha, Y.; Lv, Y.; Xu, Z.; Yang, L.; Hao, X.; Afandi, R. MGF E peptide pretreatment improves the proliferation and osteogenic dif-ferentiation of BMSCs via MEK-ERK1/2 and PI3K-Akt pathway under severe hypoxia. Life Sci. 2017, 189, 52–62. [Google Scholar] [CrossRef]
  66. Wang, X.; Hou, Y.; Li, Q.; Li, X.; Wang, W.; Ai, X.; Kuang, T.; Chen, X.; Zhang, Y.; Zhang, J.; et al. Rhodiola crenulata attenuates apoptosis and mitochondrial energy metabolism disorder in rats with hypobaric hypoxia-induced brain injury by regulating the HIF-1α/microRNA 210/ISCU1/2(COX10) signaling pathway. J. Ethnopharmacol. 2019, 241, 111801. [Google Scholar] [CrossRef]
  67. Hu, J.; Nie, Y.; Chen, S.; Xie, C.; Fan, Q.; Wang, Z.; Long, B.; Yan, G.; Zhong, Q.; Yan, X. Leucine reduces reactive oxygen species levels via an energy metabolism switch by activation of the mTOR-HIF-1α pathway in porcine intestinal epithelial cells. Int. J. Biochem. Cell Biol. 2017, 89, 42–56. [Google Scholar] [CrossRef]
  68. Saraswati, S.; Guo, Y.; Atkinson, J.; Young, P.P. Prolonged hypoxia induces monocarboxylate transporter-4 expression in mes-enchymal stem cells resulting in a secretome that is deleterious to cardiovascular repair. Stem Cells 2015, 33, 1333–1344. [Google Scholar] [CrossRef] [Green Version]
  69. Lavrentieva, A.; Majore, I.; Kasper, C.; Hass, R. Effects of hypoxic culture conditions on umbilical cord-derived human mesen-chymal stem cells. Cell Commun. Signal. 2010, 8, 18. [Google Scholar] [CrossRef] [Green Version]
  70. Regan, J.N.; Lim, J.; Shi, Y.; Joeng, K.S.; Arbeit, J.M.; Shohet, R.V.; Long, F. Up-regulation of glycolytic metabolism is required for HIF1α-driven bone formation. Proc. Natl. Acad. Sci. USA 2014, 111, 8673–8678. [Google Scholar] [CrossRef] [Green Version]
  71. Xu, W.N.; Zheng, H.L.; Yang, R.Z.; Jiang, L.S.; Jiang, S.D. HIF-1alpha Regulates Glucocorticoid-Induced Osteoporosis Through PDK1/AKT/mTOR Signaling Pathway. Front. Endocrinol. 2019, 10, 922. [Google Scholar] [CrossRef]
  72. Dirckx, N.; Tower, R.J.; Mercken, E.M.; Vangoitsenhoven, R.; Moreau-Triby, C.; Breugelmans, T.; Nefyodova, E.; Cardoen, R.; Mathieu, C.; Van der Schueren, B.; et al. Vhl deletion in osteoblasts boosts cellular glycolysis and improves global glucose metabolism. J. Clin. Investig. 2018, 128, 1087–1105. [Google Scholar] [CrossRef] [Green Version]
  73. Shum, L.C.; White, N.S.; Mills, B.N.; Bentley, K.L.; Eliseev, R.A. Energy Metabolism in Mesenchymal Stem Cells During Osteo-genic Differentiation. Stem Cells Dev. 2016, 25, 114–122. [Google Scholar] [CrossRef] [Green Version]
  74. Ke, W.; Ma, L.; Wang, B.; Song, Y.; Luo, R.; Li, G.; Liao, Z.; Shi, Y.; Wang, K.; Feng, X.; et al. N-cadherin mimetic hydrogel enhances MSC chondrogenesis through cell metabolism. Acta Biomater. 2022, 150, 83–95. [Google Scholar] [CrossRef]
  75. Mas-Bargues, C.; Sanz-Ros, J.; Román-Domínguez, A.; Gimeno-Mallench, L.; Inglés, M.; Viña, J.; Borrás, C. Extracellular Vesicles from Healthy Cells Improves Cell Function and Stemness in Premature Senescent Stem Cells by miR-302b and HIF-1α Acti-vation. Biomolecules 2020, 10, 957. [Google Scholar] [CrossRef]
  76. Riddle, R.C.; Leslie, J.M.; Gross, T.S.; Clemens, T.L. Hypoxia-inducible Factor-1α Protein Negatively Regulates Load-induced Bone Formation*. J. Biol. Chem. 2011, 286, 44449–44456. [Google Scholar] [CrossRef] [Green Version]
  77. Zhao, H.; Yeersheng, R.; Xia, Y.; Kang, P.; Wang, W. Hypoxia Enhanced Bone Regeneration Through the HIF-1alpha/beta-Catenin Pathway in Femoral Head Osteonecrosis. Am. J. Med. Sci. 2021, 362, 78–91. [Google Scholar] [CrossRef]
  78. Sheng, H.; Lao, Y.; Zhang, S.; Ding, W.; Lu, D.; Xu, B. Combined Pharmacotherapy with Alendronate and Desferoxamine Regu-late the Bone Resorption and Bone Regeneration for Preventing Glucocorticoids-Induced Osteonecrosis of the Femoral Head. Biomed. Res. Int. 2020, 2020, 3120458. [Google Scholar] [CrossRef]
  79. Yuan, Y.; Hilliard, G.; Ferguson, T.; Millhorn, D.E. Cobalt Inhibits the Interaction between Hypoxia-inducible Factor-α and von Hippel-Lindau Protein by Direct Binding to Hypoxia-inducible Factor-α. J. Biol. Chem. 2003, 278, 15911–15916. [Google Scholar] [CrossRef] [Green Version]
  80. Hewitson, K.S.; McNeill, L.A.; Riordan, M.V.; Tian, Y.-M.; Bullock, A.N.; Welford, R.W.; Elkins, J.M.; Oldham, N.J.; Bhattacharya, S.; Gleadle, J.M.; et al. Hypoxia-inducible Factor (HIF) Asparagine Hydroxylase Is Identical to Factor Inhibiting HIF (FIH) and Is Related to the Cupin Structural Family. J. Biol. Chem. 2002, 277, 26351–26355. [Google Scholar] [CrossRef] [Green Version]
  81. Loboda, A.; Jazwa, A.; Wegiel, B.; Jozkowicz, A.; Dulak, J. Heme oxygenase-1-dependent and -independent regulation of angi-ogenic genes expression: Effect of cobalt protoporphyrin and cobalt chloride on VEGF and IL-8 synthesis in human microvas-cular endothelial cells. Cell Mol. Biol. 2005, 51, 347–355. [Google Scholar] [PubMed]
  82. Yu, X.; Wan, Q.; Cheng, G.; Cheng, X.; Zhang, J.; Pathak, J.L.; Li, Z. CoCl, a mimic of hypoxia, enhances bone marrow mesen-chymal stem cells migration and osteogenic differentiation via STAT3 signaling pathway. Cell Biol. Int. 2018, 42, 1321–1329. [Google Scholar] [CrossRef] [PubMed]
  83. Yu, X.; Wan, Q.; Ye, X.; Cheng, Y.; Pathak, J.L.; Li, Z. Cellular hypoxia promotes osteogenic differentiation of mesenchymal stem cells and bone defect healing via STAT3 signaling. Cell. Mol. Biol. Lett. 2019, 24, 64. [Google Scholar] [CrossRef] [PubMed]
  84. Pacary, E.; Legros, H.; Valable, S.; Duchatelle, P.; Lecocq, M.; Petit, E.; Nicole, O.; Bernaudin, M. Synergistic effects of CoCl2 and ROCK inhibition on mesenchymal stem cell differentiation into neuron-like cells. J. Cell Sci. 2006, 119, 2667–2678. [Google Scholar] [CrossRef] [Green Version]
  85. Fan, W.; Crawford, R.; Xiao, Y. Enhancing in vivo vascularized bone formation by cobalt chloride-treated bone marrow stro-mal cells in a tissue engineered periosteum model. Biomaterials 2010, 31, 3580–3589. [Google Scholar] [CrossRef] [Green Version]
  86. Wu, C.; Zhou, Y.; Fan, W.; Han, P.; Chang, J.; Yuen, J.; Zhang, M.; Xiao, Y. Hypoxia-mimicking mesoporous bioactive glass scaf-folds with controllable cobalt ion release for bone tissue engineering. Biomaterials 2012, 33, 2076–2085. [Google Scholar] [CrossRef] [Green Version]
  87. Azevedo, M.M.; Jell, G.; O’Donnell, M.D.; Law, R.V.; Hill, R.G.; Stevens, M.M. Synthesis and characterization of hypoxia-mimicking bioactive glasses for skeletal regeneration. J. Mater. Chem. 2010, 20, 8854–8864. [Google Scholar] [CrossRef] [Green Version]
  88. Badat, M.; Kaya, B.; Telfer, P. Combination-therapy with concurrent deferoxamine and deferiprone is effective in treating re-sistant cardiac iron-loading in aceruloplasminaemia. Br. J. Haematol. 2015, 171, 430–432. [Google Scholar] [CrossRef] [Green Version]
  89. Smith, G.C.; Alpendurada, F.; Carpenter, J.P.; Alam, M.H.; Berdoukas, V.; Karagiorga, M.; Ladis, V.; Piga, A.; Aessopos, A.; Gotsis, E.D.; et al. Effect of deferiprone or deferoxamine on right ventricular function in thalassemia major patients with myocardial iron overload. J. Cardiovasc. Magn. Reson. 2011, 13, 34. [Google Scholar] [CrossRef] [Green Version]
  90. Sorond, F.A.; Tan, C.O.; LaRose, S.; Monk, A.D.; Fichorova, R.; Ryan, S.; Lipsitz, L.A. Deferoxamine, Cerebrovascular Hemody-namics, and Vascular Aging: Potential Role for Hypoxia-Inducible Transcription Factor-1-Regulated Pathways. Stroke 2015, 46, 2576–2583. [Google Scholar] [CrossRef] [Green Version]
  91. Matsunaga, K.; Fujisawa, K.; Takami, T.; Burganova, G.; Sasai, N.; Matsumoto, T.; Yamamoto, N.; Sakaida, I. NUPR1 acts as a pro-survival factor in human bone marrow-derived mesenchymal stem cells and is induced by the hypoxia mimetic reagent deferoxamine. J. Clin. Biochem. Nutr. 2019, 64, 209–216. [Google Scholar] [CrossRef]
  92. Shadid, M.; Buonocore, G.; Groenendaal, F.; Moison, R.; Ferrali, M.; Berger, H.M.; van Bel, F. Effect of deferoxamine and allopu-rinol on non-protein-bound iron concentrations in plasma and cortical brain tissue of newborn lambs following hypoxia-ischemia. Neurosci. Lett. 1998, 248, 5–8. [Google Scholar] [CrossRef]
  93. Bartolome, S.; Dhillon, N.K.; Buch, S.; Casillan, A.J.; Wood, J.G.; O’Brien-Ladner, A.R. Deferoxamine mimics the pattern of hy-poxia-related injury at the microvasculature. Shock 2009, 31, 481–485. [Google Scholar] [CrossRef]
  94. Hou, Z.; Nie, C.; Si, Z.; Ma, Y. Deferoxamine enhances neovascularization and accelerates wound healing in diabetic rats via the accumulation of hypoxia-inducible factor-1α. Diabetes Res. Clin. Pract. 2013, 101, 62–71. [Google Scholar] [CrossRef]
  95. Xie, P.; Yang, L.; Talaiti, A.; Wu, J.J.; Yu, J.; Yu, T.; Wang, H.Y.; Huang, B.; Wu, Q.; Maimaitili, Y.; et al. Deferoxamine-activated hypoxia-inducible factor-1 restores cardioprotective effects of sevoflurane postconditioning in diabetic rats. Acta Physiol. 2017, 221, 98–114. [Google Scholar] [CrossRef]
  96. Choi, C.W.; Lee, J.; Lee, H.J.; Park, H.-S.; Chun, Y.-S.; Kim, B.I. Deferoxamine Improves Alveolar and Pulmonary Vascular De-velopment by Upregulating Hypoxia-inducible Factor-1α in a Rat Model of Bronchopulmonary Dysplasia. J. Korean Med. Sci. 2015, 30, 1295–1301. [Google Scholar] [CrossRef] [Green Version]
  97. Du, R.; Zhao, J.; Wen, Y.; Zhu, Y.; Jiang, L. Deferoxamine enhances the migration of dental pulp cells via hypoxia-inducible factor 1α. Am. J. Transl. Res. 2021, 13, 4780–4787. [Google Scholar]
  98. Oses, C.; Olivares, B.; Ezquer, M.; Acosta, C.; Bosch, P.; Donoso, M.; Léniz, P.; Ezquer, F. Preconditioning of adipose tissue-derived mesenchymal stem cells with deferoxamine increases the production of pro-angiogenic, neuroprotective and anti-inflammatory factors: Potential application in the treatment of diabetic neuropathy. PLoS ONE 2017, 12, e0178011. [Google Scholar] [CrossRef] [Green Version]
  99. Nouri, F.; Salehinejad, P.; Nematollahi-Mahani, S.N.; Kamarul, T.; Zarrindast, M.R.; Sharifi, A.M. Deferoxamine Precondition-ing of Neural-Like Cells Derived from Human Wharton’s Jelly Mesenchymal Stem Cells as a Strategy to Promote Their Toler-ance and Therapeutic Potential: An In Vitro Study. Cell Mol. Neurobiol. 2016, 36, 689–700. [Google Scholar] [CrossRef]
  100. Groenendaal, F.; Shadid, M.; McGowan, J.E.; Mishra, O.P.; van Bel, F. Effects of deferoxamine, a chelator of free iron, on NA(+), K(+)-ATPase activity of cortical brain cell membrane during early reperfusion after hypoxia-ischemia in newborn lambs. Pediatr. Res. 2000, 48, 560–564. [Google Scholar] [CrossRef] [Green Version]
  101. Vrtačnik, P.; Marc, J.; Ostanek, B. Hypoxia mimetic deferoxamine influences the expression of histone acetylation- and DNA methylation-associated genes in osteoblasts. Connect. Tissue Res. 2015, 56, 228–235. [Google Scholar] [CrossRef] [PubMed]
  102. Donneys, A.; Weiss, D.M.; Deshpande, S.S.; Ahsan, S.; Tchanque-Fossuo, C.N.; Sarhaddi, D.; Levi, B.; Goldstein, S.A.; Buchman, S.R. Localized deferoxamine injection augments vascularity and improves bony union in pathologic fracture healing after radiotherapy. Bone 2013, 52, 318–325. [Google Scholar] [CrossRef] [PubMed]
  103. Farberg, A.S.; Jing, X.L.; Monson, L.A.; Donneys, A.; Tchanque-Fossuo, C.N.; Deshpande, S.S.; Buchman, S.R. Deferoxamine reverses radiation induced hypovascularity during bone regeneration and repair in the murine mandible. Bone 2012, 50, 1184–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lang, A.; Stefanowski, J.; Pfeiffenberger, M.; Wolter, A.; Damerau, A.; Hemmati-Sadeghi, S.; Haag, R.; Hauser, A.E.; Lohning, M.; Duda, G.N.; et al. MIF does only marginally enhance the pro-regenerative capaci-ties of DFO in a mouse-osteotomy-model of compromised bone healing conditions. Bone 2022, 154, 116247. [Google Scholar] [CrossRef] [PubMed]
  105. Messer, J.G.; Cooney, P.T.; Kipp, D.E. Iron chelator deferoxamine alters iron-regulatory genes and proteins and suppresses osteoblast phenotype in fetal rat calvaria cells. Bone 2010, 46, 1408–1415. [Google Scholar] [CrossRef] [Green Version]
  106. Chen, B.; Yan, Y.-L.; Liu, C.; Bo, L.; Li, G.; Wang, H.; Xu, Y.-J. Therapeutic Effect of Deferoxamine on Iron Overload-Induced Inhibition of Osteogenesis in a Zebrafish Model. Calcif. Tissue Res. 2014, 94, 353–360. [Google Scholar] [CrossRef]
  107. Chung, J.H.; Kim, Y.S.; Noh, K.; Lee, Y.M.; Chang, S.W.; Kim, E.C. Deferoxamine promotes osteoblastic differentiation in hu-man periodontal ligament cells via the nuclear factor erythroid 2-related factor-mediated antioxidant signaling pathway. J. Periodontal Res. 2014, 49, 563–573. [Google Scholar] [CrossRef]
  108. Lee, K.E.; Mo, S.; Lee, H.-S.; Jeon, M.; Song, J.S.; Choi, H.-J.; Cho, H.; Kang, C.-M. Deferoxamine Reduces Inflammation and Os-teoclastogenesis in Avulsed Teeth. Int. J. Mol. Sci. 2021, 22, 8225. [Google Scholar]
  109. Guo, C.; Yang, K.; Yan, Y.; Yan, D.; Cheng, Y.; Yan, X.; Qian, N.; Zhou, Q.; Chen, B.; Jiang, M.; et al. SF-deferoxamine, a bone-seeking angiogenic drug, prevents bone loss in estrogen-deficient mice. Bone 2019, 120, 156–165. [Google Scholar] [CrossRef]
  110. Zhang, J.; Hu, W.; Ding, C.; Yao, G.; Zhao, H.; Wu, S. Deferoxamine inhibits iron-uptake stimulated osteoclast differentiation by suppressing electron transport chain and MAPKs signaling. Toxicol. Lett. 2019, 313, 50–59. [Google Scholar] [CrossRef]
  111. Fan, L.; Li, J.; Yu, Z.; Dang, X.; Wang, K. Hypoxia-inducible factor prolyl hydroxylase inhibitor prevents steroid-associated oste-onecrosis of the femoral head in rabbits by promoting angiogenesis and inhibiting apoptosis. PLoS ONE 2014, 9, e107774. [Google Scholar] [CrossRef]
  112. Li, L.; Li, A.; Zhu, L.; Gan, L.; Zuo, L. Roxadustat promotes osteoblast differentiation and prevents estrogen deficiency-induced bone loss by stabilizing HIF-1α and activating the Wnt/β-catenin signaling pathway. J. Orthop. Surg. Res. 2022, 17, 286. [Google Scholar] [CrossRef]
  113. Deppe, J.; Popp, T.; Egea, V.; Steinritz, D.; Schmidt, A.; Thiermann, H.; Weber, C.; Ries, C. Impairment of hypoxia-induced HIF-1α signaling in keratinocytes and fibroblasts by sulfur mustard is counteracted by a selective PHD-2 inhibitor. Arch. Toxicol. 2015, 90, 1141–1150. [Google Scholar] [CrossRef]
  114. Peng, J.; Lai, Z.G.; Fang, Z.L.; Xing, S.; Hui, K.; Hao, C.; Jin, Q.; Qi, Z.; Shen, W.J.; Dong, Q.N.; et al. Dimethyloxalyl-glycine prevents bone loss in ovariectomized C57BL/6J mice through enhanced angiogenesis and osteogenesis. PLoS ONE 2014, 9, e112744. [Google Scholar] [CrossRef] [Green Version]
  115. Zhou, B.; Ge, T.; Zhou, L.; Jiang, L.; Zhu, L.; Yao, P.; Yu, Q. Dimethyloxalyl Glycine Regulates the HIF-1 Signaling Pathway in Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2020, 16, 702–710. [Google Scholar] [CrossRef]
  116. Woo, K.M.; Jung, H.-M.; Oh, J.-H.; Rahman, S.U.; Kim, S.M.; Baek, J.-H.; Ryoo, H.-M. Synergistic effects of dimethyloxalylglycine and butyrate incorporated into α-calcium sulfate on bone regeneration. Biomaterials 2015, 39, 1–14. [Google Scholar] [CrossRef]
  117. Zhang, J.; Feng, Z.; Wei, J.; Yu, Y.; Luo, J.; Zhou, J.; Li, Y.; Zheng, X.; Tang, W.; Liu, L.; et al. Repair of Critical-Sized Mandible Defects in Aged Rat Using Hypoxia Preconditioned BMSCs with Up-regulation of Hif-1α. Int. J. Biol. Sci. 2018, 14, 449–460. [Google Scholar] [CrossRef] [Green Version]
  118. Wang, X.; Wei, L.; Li, Q.; Lai, Y. HIF-1α protects osteoblasts from ROS-induced apoptosis. Free Radic. Res. 2022, 56, 143–153. [Google Scholar] [CrossRef]
  119. Abu-Shahba, A.G.; Gebraad, A.; Kaur, S.; Paananen, R.O.; Peltoniemi, H.; Seppanen-Kaijansinkko, R.; Mannerstrom, B. Proan-giogenic Hypoxia-Mimicking Agents Attenuate Osteogenic Potential of Adipose Stem/Stromal Cells. Tissue Eng. Regen. Med. 2020, 17, 477–493. [Google Scholar] [CrossRef]
  120. Ding, H.; Gao, Y.-S.; Wang, Y.; Hu, C.; Sun, Y.; Zhang, C. Dimethyloxaloylglycine Increases the Bone Healing Capacity of Adipose-Derived Stem Cells by Promoting Osteogenic Differentiation and Angiogenic Potential. Stem Cells Dev. 2014, 23, 990–1000. [Google Scholar] [CrossRef] [Green Version]
  121. Weng, T.; Xie, Y.; Huang, J.; Luo, F.; Yi, L.; He, Q.; Chen, D.; Chen, L. Inactivation of Vhl in osteochondral progenitor cells causes high bone mass phenotype and protects against age-related bone loss in adult mice. J. Bone Miner. Res. 2014, 29, 820–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Cheng, H.; Jiang, W.; Phillips, F.M.; Haydon, R.C.; Peng, Y.; Zhou, L.; Luu, H.H.; An, N.; Breyer, B.; Vanichakarn, P.; et al. Osteogenic Activity of the Fourteen Types of Human Bone Morphogenetic Proteins (BMPS). J. Bone Jt. Surg. 2003, 85, 1544–1552. [Google Scholar] [CrossRef] [PubMed]
  123. Song, J.J.; Celeste, A.J.; Kong, F.M.; Jirtle, R.L.; Rosen, V.; Thies, R.S. Bone morphogenetic protein-9 binds to liver cells and stimu-lates proliferation. Endocrinology 1995, 136, 4293–4297. [Google Scholar] [CrossRef] [PubMed]
  124. Castonguay, R.; Werner, E.D.; Matthews, R.G.; Presman, E.; Mulivor, A.W.; Solban, N.; Sako, D.; Pearsall, R.S.; Underwood, K.W.; Seehra, J.; et al. Soluble Endoglin Specifically Binds Bone Morphogenetic Proteins 9 and 10 via Its Orphan Domain, Inhibits Blood Vessel Formation, and Suppresses Tumor Growth. J. Biol. Chem. 2011, 286, 30034–30046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Cunha, S.I.; Pardali, E.; Thorikay, M.; Anderberg, C.; Hawinkels, L.; Goumans, M.-J.; Seehra, J.; Heldin, C.-H.; Dijke, P.T.; Pietras, K. Genetic and pharmacological targeting of activin receptor-like kinase 1 impairs tumor growth and angiogenesis. J. Exp. Med. 2010, 207, 85–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hu, N.; Jiang, D.; Huang, E.; Liu, X.; Li, R.; Liang, X.; Kim, S.H.; Chen, X.; Gao, J.L.; Zhang, H.; et al. BMP9-regulated angiogenic signaling plays an important role in the osteogenic differentiation of mesenchymal pro-genitor cells. J. Cell Sci. 2013, 126, 532–541. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, Y.; Wan, C.; Deng, L.; Liu, X.; Cao, X.; Gilbert, S.R.; Bouxsein, M.L.; Faugere, M.-C.; Guldberg, R.E.; Gerstenfeld, L.C.; et al. The hypoxia-inducible factor alpha pathway couples angiogenesis to osteo-genesis during skeletal development. J. Clin. Investig. 2007, 117, 1616–1626. [Google Scholar] [CrossRef]
  128. Zhang, L.-F.; Qi, J.; Zuo, G.; Jia, P.; Shen, X.; Shao, J.; Kang, H.; Yang, H.; Deng, L. Osteoblast-secreted factors promote prolifera-tion and osteogenic differentiation of bone marrow stromal cells via VEGF/heme-oxygenase-1 pathway. PLoS ONE 2014, 9, e99946. [Google Scholar]
  129. Zhao, Q.; Shen, X.; Zhang, W.; Zhu, G.; Qi, J.; Deng, L. Mice with increased angiogenesis and osteogenesis due to conditional activation of HIF pathway in osteoblasts are protected from ovariectomy induced bone loss. Bone 2012, 50, 763–770. [Google Scholar] [CrossRef]
  130. Zuo, G.-L.; Zhang, L.-F.; Qi, J.; Kang, H.; Jia, P.; Chen, H.; Shen, X.; Guo, L.; Zhou, H.-B.; Wang, J.-S.; et al. Activation of HIFa Pathway in Mature Osteoblasts Disrupts the Integrity of the Osteocyte/Canalicular Network. PLoS ONE 2015, 10, e0121266. [Google Scholar] [CrossRef] [Green Version]
  131. Loots, G.G.; Robling, A.G.; Chang, J.C.; Murugesh, D.K.; Bajwa, J.; Carlisle, C.; Manilay, J.O.; Wong, A.; Yellowley, C.E.; Genetos, D.C. Vhl deficiency in osteocytes produces high bone mass and hematopoietic defects. Bone 2018, 116, 307–314. [Google Scholar] [CrossRef] [Green Version]
  132. Mangiavini, L.; Merceron, C.; Araldi, E.; Khatri, R.; Gerard-O’Riley, R.; Wilson, T.L.; Rankin, E.B.; Giaccia, A.J.; Schipani, E. Loss of VHL in mesenchymal progenitors of the limb bud alters multiple steps of endochondral bone development. Dev. Biol. 2014, 393, 124–136. [Google Scholar] [CrossRef] [Green Version]
  133. Pfander, D.; Kobayashi, T.; Knight, M.C.; Zelzer, E.; Chan, D.A.; Olsen, B.R.; Giaccia, A.J.; Johnson, R.S.; Haase, V.H.; Schipani, E. Deletion of Vhlh in chondrocytes reduces cell proliferation and increases matrix deposition during growth plate development. Development 2004, 131, 2497–2508. [Google Scholar] [CrossRef] [Green Version]
  134. Rivera, J.C.; Madaan, A.; Zhou, T.E.; Chemtob, S. Review of the mechanisms and therapeutic avenues for retinal and choroidal vascular dysfunctions in retinopathy of prematurity. Acta Paediatr. 2016, 105, 1421–1433. [Google Scholar] [CrossRef]
  135. Rubio, R.G.; Adamis, A.P. Ocular Angiogenesis: Vascular Endothelial Growth Factor and Other Factors. Basic Sci. Retin. 2015, 55, 28–37. [Google Scholar] [CrossRef]
  136. Palazon, A.; Tyrakis, P.A.; Macias, D.; Veliça, P.; Rundqvist, H.; Fitzpatrick, S.; Vojnovic, N.; Phan, A.T.; Loman, N.; Hedenfalk, I.; et al. An HIF-1α/VEGF-A Axis in Cytotoxic T Cells Regulates Tumor Progression. Cancer Cell 2017, 32, 669–683.e5. [Google Scholar] [CrossRef] [Green Version]
  137. Xu, Y.; Lu, X.; Hu, Y.; Yang, B.; Tsui, C.-K.; Yu, S.; Lu, L.; Liang, X. Melatonin attenuated retinal neovascularization and neu-roglial dysfunction by inhibition of HIF-1α-VEGF pathway in oxygen-induced retinopathy mice. J. Pineal Res. 2018, 64, e12473. [Google Scholar] [CrossRef]
  138. Liu, J.; Wang, W.; Wang, L.; Chen, S.; Tian, B.; Huang, K.; Corrigan, C.J.; Ying, S.; Wang, W.; Wang, C. IL-33 Initiates Vascular Remodelling in Hypoxic Pulmonary Hypertension by up-Regulating HIF-1α and VEGF Expression in Vascular Endothelial Cells. eBioMedicine 2018, 33, 196–210. [Google Scholar] [CrossRef]
  139. Xu, Z.; Zhu, C.; Chen, C.; Zong, Y.; Feng, H.; Liu, D.; Feng, W.; Zhao, J.; Lu, A. CCL19 suppresses angiogenesis through promot-ing miR-206 and inhibiting Met/ERK/Elk-1/HIF-1α/VEGF-A pathway in colorectal cancer. Cell Death Dis. 2018, 9, 974. [Google Scholar] [CrossRef] [Green Version]
  140. Fan, J.; Lv, H.; Li, J.; Che, Y.; Xu, B.; Tao, Z.; Jiang, W. Roles of Nrf2/HO-1 and HIF-1α/VEGF in lung tissue injury and repair following cerebral ischemia/reperfusion injury. J. Cell. Physiol. 2019, 234, 7695–7707. [Google Scholar] [CrossRef]
  141. Lin, C.-J.; Lan, Y.-M.; Ou, M.-Q.; Ji, L.-Q.; Lin, S.-D. Expression of miR-217 and HIF-1α/VEGF pathway in patients with diabetic foot ulcer and its effect on angiogenesis of diabetic foot ulcer rats. J. Endocrinol. Investig. 2019, 42, 1307–1317. [Google Scholar] [CrossRef] [PubMed]
  142. Mao, H.; Jiang, C.; Xu, L.; Chen, D.; Liu, H.; Xu, Y.; Ma, K.; Wang, M. Ginsenoside protects against AKI via activation of HIF-1α and VEGF-A in the kidney-brain axis. Int. J. Mol. Med. 2020, 45, 939–946. [Google Scholar] [CrossRef]
  143. Hepp, M.; Werion, A.; De Greef, A.; Goyet, C.D.V.D.; de Bournonville, M.; Behets, C.; Lengelé, B.; Daumerie, C.; Mourad, M.; Ludgate, M.; et al. Oxidative Stress-Induced Sirtuin1 Downregulation Correlates to HIF-1α, GLUT-1, and VEGF-A Upregulation in Th1 Autoimmune Hashimoto’s Thyroiditis. Int. J. Mol. Sci. 2021, 22, 3806. [Google Scholar] [CrossRef] [PubMed]
  144. Feng, Z.-T.; Yang, T.; Hou, X.-Q.; Wu, H.-Y.; Feng, J.-T.; Ou, B.-J.; Cai, S.-J.; Li, J.; Mei, Z.-G. Sinomenine mitigates collagen-induced arthritis mice by inhibiting angiogenesis. Biomed. Pharmacother. 2019, 113, 108759. [Google Scholar] [CrossRef] [PubMed]
  145. Westra, J.; Molema, G.; Kallenberg, C.G.M. Hypoxia-Inducible Factor-1 as Regulator of Angiogenesis in Rheumatoid Arthritis—Therapeutic Implications. Curr. Med. Chem. 2010, 17, 254–263. [Google Scholar] [CrossRef]
  146. Veale, D.J.; Orr, C.; Fearon, U. Cellular and molecular perspectives in rheumatoid arthritis. Semin. Immunopathol. 2017, 39, 343–354. [Google Scholar] [CrossRef]
  147. Chen, Y.-Y.; Brown, N.J.; Jones, R.; Lewis, C.E.; Mujamammi, A.H.; Muthana, M.; Seed, M.P.; Barker, M.D. A peptide derived from TIMP-3 inhibits multiple angiogenic growth factor receptors and tumour growth and inflammatory arthritis in mice. Angiogenesis 2013, 17, 207–219. [Google Scholar] [CrossRef] [Green Version]
  148. Chai, M.; Gu, C.; Shen, Q.; Liu, J.; Zhou, Y.; Jin, Z.; Xiong, W.; Zhou, Y.; Tan, W. Hypoxia alleviates dexamethasone-induced inhi-bition of angiogenesis in cocultures of HUVECs and rBMSCs via HIF-1alpha. Stem Cell Res. Ther. 2020, 11, 343. [Google Scholar] [CrossRef]
  149. Nakashima, K.; Zhou, X.; Kunkel, G.; Zhang, Z.; Deng, J.M.; Behringer, R.R.; de Crombrugghe, B. The Novel Zinc Finger-Containing Transcription Factor Osterix Is Required for Osteoblast Differentiation and Bone Formation. Cell 2002, 108, 17–29. [Google Scholar] [CrossRef] [Green Version]
  150. Kusumbe, A.P.; Ramasamy, S.K.; Adams, R.H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014, 507, 323–328. [Google Scholar] [CrossRef] [Green Version]
  151. Ramasamy, S.K.; Kusumbe, A.P.; Wang, L.; Adams, R.H. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature 2014, 507, 376–380. [Google Scholar] [CrossRef] [Green Version]
  152. Wang, L.; Zhou, F.; Zhang, P.; Wang, H.; Qu, Z.; Jia, P.; Yao, Z.; Shen, G.; Li, G.; Zhao, G.; et al. Human type H vessels are a sensitive biomarker of bone mass. Cell Death Dis. 2017, 8, e2760. [Google Scholar] [CrossRef] [Green Version]
  153. García-Martín, A.; Acitores, A.; Maycas, M.; Villanueva-Peñacarrillo, M.L.; Esbrit, P. Src kinases mediate VEGFR2 transactiva-tion by the osteostatin domain of PTHrP to modulate osteoblastic function. J. Cell. Biochem. 2013, 114, 1404–1413. [Google Scholar] [CrossRef]
  154. Wu, D.; Liu, L.; Fu, S.; Zhang, J. Osteostatin improves the Osteogenic differentiation of mesenchymal stem cells and enhances angiogenesis through HIF-1α under hypoxia conditions in vitro. Biochem. Biophys. Res. Commun. 2022, 606, 100–107. [Google Scholar] [CrossRef]
  155. Gao, B.; Lin, X.; Jing, H.; Fan, J.; Ji, C.; Jie, Q.; Zheng, C.; Wang, D.; Xu, X.; Hu, Y.; et al. Local delivery of tetra-methylpyrazine eliminates the senescent phenotype of bone marrow mesenchymal stromal cells and creates an anti-inflammatory and angiogenic environment in aging mice. Aging Cell 2018, 17, e12741. [Google Scholar] [CrossRef]
  156. Liu, X.-D.; Cai, F.; Liu, L.; Zhang, Y.; Yang, A.-L. microRNA-210 is involved in the regulation of postmenopausal osteoporosis through promotion of VEGF expression and osteoblast differentiation. Biol. Chem. 2015, 396, 339–347. [Google Scholar] [CrossRef]
  157. Yang, M.; Li, C.-J.; Sun, X.; Guo, Q.; Xiao, Y.; Su, T.; Tu, M.-L.; Peng, H.; Lu, Q.; Liu, Q.; et al. MiR-497∼195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch and HIF-1α activity. Nat. Commun. 2017, 8, 16003. [Google Scholar] [CrossRef] [Green Version]
  158. Weinstein, R.S.; Hogan, E.A.; Borrelli, M.J.; Liachenko, S.; O’Brien, C.A.; Manolagas, S.C. The Pathophysiological Sequence of Glucocorticoid-Induced Osteonecrosis of the Femoral Head in Male Mice. Endocrinology 2017, 158, 3817–3831. [Google Scholar] [CrossRef] [Green Version]
  159. Jiang, L.; Sheng, K.; Wang, C.; Xue, D.; Pan, Z. The Effect of MMP-2 Inhibitor 1 on Osteogenesis and Angiogenesis During Bone Regeneration. Front. Cell Dev. Biol. 2021, 8, 596783. [Google Scholar] [CrossRef]
  160. Yamauchi, N.; Taguchi, Y.; Kato, H.; Umeda, M. High-power, red-light-emitting diode irradiation enhances proliferation, osteogenic differentiation, and mineralization of human periodontal ligament stem cells via ERK signaling pathway. J. Periodontol. 2018, 89, 351–360. [Google Scholar] [CrossRef]
  161. Sarvestani, F.K.; Dehno, N.S.; Nazhvani, S.D.; Bagheri, M.H.; Abbasi, S.; Khademolhosseini, Y.; Gorji, E. Effect of low-level laser therapy on fracture healing in rabbits. Laser Ther. 2017, 26, 189–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Escudero, J.S.B.; Perez, M.G.B.; Rosso, M.P.D.O.; Buchaim, D.V.; Pomini, K.T.; Campos, L.M.G.; Audi, M.; Buchaim, R.L. Photobiomodulation therapy (PBMT) in bone repair: A systematic review. Injury 2019, 50, 1853–1867. [Google Scholar] [CrossRef]
  163. Chen, C.; Yan, S.; Qiu, S.; Geng, Z.; Wang, Z. HIF/Ca2+/NO/ROS is critical in roxadustat treating bone fracture by stimulating the proliferation and migration of BMSCs. Life Sci. 2021, 264, 118684. [Google Scholar] [CrossRef] [PubMed]
  164. Migliario, M.; Pittarella, P.; Fanuli, M.; Rizzi, M.; Renò, F. Laser-induced osteoblast proliferation is mediated by ROS production. Lasers Med. Sci. 2014, 29, 1463–1467. [Google Scholar] [CrossRef] [PubMed]
  165. Bai, J.; Li, L.; Kou, N.; Bai, Y.; Zhang, Y.; Lu, Y.; Gao, L.; Wang, F. Low level laser therapy promotes bone regeneration by cou-pling angiogenesis and osteogenesis. Stem Cell Res. Ther. 2021, 12, 432. [Google Scholar] [CrossRef]
  166. Li, Z.; Yang, B.; Weng, X.; Tse, G.; Chan, M.T.V.; Wu, W.K.K. Emerging roles of MicroRNAs in osteonecrosis of the femoral head. Cell Prolif. 2017, 51, e12405. [Google Scholar] [CrossRef] [Green Version]
  167. Yang, C.; Liu, X.; Zhao, K.; Zhu, Y.; Hu, B.; Zhou, Y.; Wang, M.; Wu, Y.; Zhang, C.; Xu, J.; et al. miRNA-21 promotes osteogenesis via the PTEN/PI3K/Akt/HIF-1α pathway and enhances bone regeneration in critical size defects. Stem Cell Res. Ther. 2019, 10, 65. [Google Scholar] [CrossRef]
  168. Costa, V.; Raimondi, L.; Conigliaro, A.; Salamanna, F.; Carina, V.; de Luca, A.; Bellavia, D.; Alessandro, R.; Fini, M.; Giavaresi, G. Hypoxia-inducible factor 1A may regulate the commitment of mesenchymal stromal cells toward angio-osteogenesis by mir-na-675-5P. Cytotherapy 2017, 19, 1412–1425. [Google Scholar] [CrossRef]
  169. Costa, V.; Carina, V.; Conigliaro, A.; Raimondi, L.; De Luca, A.; Bellavia, D.; Salamanna, F.; Setti, S.; Alessandro, R.; Fini, M.; et al. miR-31-5p Is a LIPUS-Mechanosensitive MicroRNA that Targets HIF-1α Signaling and Cytoskeletal Proteins. Int. J. Mol. Sci. 2019, 20, 1569. [Google Scholar] [CrossRef] [Green Version]
  170. Zhang, D.; Du, J.; Yu, M.; Suo, L. Urine-derived stem cells-extracellular vesicles ameliorate diabetic osteoporosis through HDAC4/HIF-1α/VEGFA axis by delivering microRNA-26a-5p. Cell Biol. Toxicol. 2022. [Google Scholar] [CrossRef]
  171. Li, Z.; Huang, C.; Yang, B.; Hu, W.; Chan, M.T.; Wu, W.K.K. Emerging roles of long non-coding RNAs in osteonecrosis of the femoral head. Am. J. Transl. Res. 2020, 12, 5984–5991. [Google Scholar]
  172. Tian, Y.; Shao, Q.; Gu, J.; Tang, Y.; Bie, M.; Zhou, Y.; Cheng, C.; Liang, Y.; Zhang, Q.; Kang, F. LncRNA-mRNA Expression Pro-files of Osteoclast After Conditional Knockout HIF-1α. Front. Genet. 2022, 13, 909095. [Google Scholar] [CrossRef]
  173. Zhang, Z.-C.; Tang, C.; Dong, Y.; Zhang, J.; Yuan, T.; Tao, S.-C.; Li, X.-L. Targeting the long noncoding RNA MALAT1 blocks the pro-angiogenic effects of osteosarcoma and suppresses tumour growth. Int. J. Biol. Sci. 2017, 13, 1398–1408. [Google Scholar] [CrossRef]
  174. Saigusa, N.; Hirai, H.; Tada, Y.; Kawakita, D.; Nakaguro, M.; Tsukahara, K.; Kano, S.; Ozawa, H.; Kondo, T.; Okami, K.; et al. The Role of the EZH2 and H3K27me3 Expression as a Predictor of Clinical Outcomes in Salivary Duct Carcinoma Patients: A Large-Series Study with Emphasis on the Relevance to the Combined Androgen Blockade and HER2-Targeted Therapy. Front. Oncol. 2022, 11, 779882. [Google Scholar] [CrossRef]
  175. Zhang, F.; Luo, H.; Peng, W.; Wang, L.; Wang, T.; Xie, Z.; Zhang, J.; Dong, W.; Zheng, X.; Liu, G.; et al. Hy-poxic condition induced H3K27me3 modification of the LncRNA Tmem235 promoter thus supporting apoptosis of BMSCs. Apoptosis 2022, 27, 762–777. [Google Scholar] [CrossRef]
  176. Xu, Y.; Wang, S.; Tang, C.; Chen, W. Upregulation of long non-coding RNA HIF 1α-anti-sense 1 induced by transforming growth factor-β-mediated targeting of sirtuin 1 promotes osteoblastic differentiation of human bone marrow stromal cells. Mol. Med. Rep. 2015, 12, 7233–7238. [Google Scholar] [CrossRef] [Green Version]
  177. Tan, Z.; Zhou, B.; Zheng, J.; Huang, Y.; Zeng, H.; Xue, L.; Wang, D. Lithium and Copper Induce the Osteogenesis-Angiogenesis Coupling of Bone Marrow Mesenchymal Stem Cells via Crosstalk between Canonical Wnt and HIF-1alpha Signaling Path-ways. Stem Cells Int. 2021, 2021, 6662164. [Google Scholar] [CrossRef]
  178. Li, J.-Y.; Wang, T.-T.; Li, C.; Wang, Z.-F.; Li, S.; Ma, L.; Zheng, L.-L. Semaphorin 3A-hypoxia inducible factor 1 subunit alpha co-overexpression enhances the osteogenic differentiation of induced pluripotent stem cells-derived mesenchymal stem cells in vitro. Chin. Med. J. 2020, 133, 301–309. [Google Scholar] [CrossRef]
  179. Li, J.; Wang, T.; Li, C.; Wang, Z.; Wang, P.; Zheng, L. Sema3A and HIF1alpha co-overexpressed iPSC-MSCs/HA scaffold facili-tates the repair of calvarial defect in a mouse model. J. Cell Physiol. 2020, 235, 6754–6766. [Google Scholar] [CrossRef]
  180. Wang, Z.; Han, T.; Zhu, H.; Tang, J.; Guo, Y.; Jin, Y.; Wang, Y.; Chen, G.; Gu, N.; Wang, C. Potential Osteoinductive Effects of Hydroxyapatite Nanoparticles on Mesenchymal Stem Cells by Endothelial Cell Interaction. Nanoscale Res. Lett. 2021, 16, 67. [Google Scholar] [CrossRef]
  181. Li, Q.; Yang, Z.; Wei, Z.; Li, D.; Luo, Y.; Kang, P. Copper-Lithium-Doped Nanohydroxyapatite Modulates Mesenchymal Stem Cells Homing to Treat Glucocorticoids-Related Osteonecrosis of the Femoral Head. Front. Bioeng. Biotechnol. 2022, 10, 916562. [Google Scholar] [CrossRef] [PubMed]
  182. Li, B.; Lei, Y.; Hu, Q.; Li, D.; Zhao, H.; Kang, P. Porous copper- and lithium-doped nano-hydroxyapatite composite scaffold promotes angiogenesis and bone regeneration in the repair of glucocorticoids-induced osteonecrosis of the femoral head. Biomed. Mater. 2021, 16, 065012. [Google Scholar] [CrossRef] [PubMed]
  183. Li, D.; Huifang, L.; Zhao, J.; Yang, Z.; Xie, X.; Wei, Z.; Li, D.; Kang, P. Porous lithium-doped hydroxyapatite scaffold seeded with hypoxia-preconditioned bone-marrow mesenchymal stem cells for bone-tissue regeneration. Biomed. Mater. 2018, 13, 055002. [Google Scholar] [CrossRef] [PubMed]
  184. Wu, Q.; Li, J.; Zhang, W.; Qian, H.; She, W.; Pan, H.; Wen, J.; Zhang, X.; Liu, X.; Jiang, X. Antibacterial property, angiogenic and osteogenic activity of Cu-incorporated TiO2 coating. J. Mater. Chem. B 2014, 2, 6738–6748. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, W.; Chang, Q.; Xu, L.; Li, G.; Yang, G.; Ding, X.; Wang, X.; Cui, D.; Jiang, X. Graphene Oxide-Copper Nanocomposite-Coated Porous CaP Scaffold for Vascularized Bone Regeneration via Activation of Hif-1α. Adv. Healthc. Mater. 2016, 5, 1299–1309. [Google Scholar] [CrossRef]
  186. Wang, X.; Wu, C.; Qi, H.; Tian, M.; Xie, H.; Wang, Y.; Gu, Z.; Peng, X.; Yu, X. Introducing copper and collagen (via poly(DOPA)) coating to activate inert ceramic scaffolds for excellent angiogenic and osteogenic capacity. RSC Adv. 2018, 8, 15575–15586. [Google Scholar] [CrossRef] [Green Version]
  187. Zhao, H.; Liang, G.; Liang, W.; Li, Q.; Huang, B.; Li, A.; Qiu, D.; Jin, D. In vitro and in vivo evaluation of the pH-neutral bioac-tive glass as high performance bone grafts. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 116, 111249. [Google Scholar] [CrossRef]
  188. Dai, Q.; Li, Q.; Gao, H.; Yao, L.; Lin, Z.; Li, D.; Zhu, S.; Liu, C.; Yang, Z.; Wang, G.; et al. 3D printing of Cu-doped bioactive glass composite scaffolds promotes bone regeneration through activating the HIF-1α and TNF-α pathway of hUVECs. Biomater. Sci. 2021, 9, 5519–5532. [Google Scholar] [CrossRef]
  189. Li, H.; Li, J.; Jiang, J.; Lv, F.; Chang, J.; Chen, S.; Wu, C. An osteogenesis/angiogenesis-stimulation artificial ligament for anterior cruciate ligament reconstruction. Acta Biomater. 2017, 54, 399–410. [Google Scholar] [CrossRef]
  190. Kulanthaivel, S.; Roy, B.; Agarwal, T.; Giri, S.; Pramanik, K.; Pal, K.; Ray, S.S.; Maiti, T.K.; Banerjee, I. Cobalt doped proangiogen-ic hydroxyapatite for bone tissue engineering application. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 58, 648–658. [Google Scholar] [CrossRef]
  191. Shah, K.; Dunning, M.; Gartland, A.; Wilkinson, J. Distinct Concentration-Dependent Molecular Pathways Regulate Bone Cell Responses to Cobalt and Chromium Exposure from Joint Replacement Prostheses. Int. J. Mol. Sci. 2021, 22, 5225. [Google Scholar] [CrossRef]
  192. Deng, Z.; Lin, B.; Jiang, Z.; Huang, W.; Li, J.; Zeng, X.; Wang, H.; Wang, D.; Zhang, Y. Hypoxia-Mimicking Cobalt-Doped Borosil-icate Bioactive Glass Scaffolds with Enhanced Angiogenic and Osteogenic Capacity for Bone Regeneration. Int. J. Biol. Sci. 2019, 15, 1113–1124. [Google Scholar] [CrossRef] [Green Version]
  193. Littmann, E.; Autefage, H.; Solanki, A.K.; Kallepitis, C.; Jones, J.R.; Alini, M.; Peroglio, M.; Stevens, M.M. Cobalt-containing bio-active glasses reduce human mesenchymal stem cell chondrogenic differentiation despite HIF-1α stabilization. J. Eur. Ceram. Soc. 2018, 38, 877–886. [Google Scholar] [CrossRef]
  194. Perez, R.; Kim, J.-H.; Buitrago, J.O.; Wall, I.B.; Kim, H.-W. Novel therapeutic core–shell hydrogel scaffolds with sequential delivery of cobalt and bone morphogenetic protein-2 for synergistic bone regeneration. Acta Biomater. 2015, 23, 295–308. [Google Scholar] [CrossRef] [PubMed]
  195. Quinlan, E.; Partap, S.; Azevedo, M.M.; Jell, G.; Stevens, M.M.; O’Brien, F.J. Hypoxia-mimicking bioactive glass/collagen gly-cosaminoglycan composite scaffolds to enhance angiogenesis and bone repair. Biomaterials 2015, 52, 358–366. [Google Scholar] [CrossRef]
  196. Kulanthaivel, S.; Agarwal, T.; Rathnam, V.S.; Pal, K.; Banerjee, I. Cobalt doped nano-hydroxyapatite incorporated gum tragacanth-alginate beads as angiogenic-osteogenic cell encapsulation system for mesenchymal stem cell based bone tissue engineering. Int. J. Biol. Macromol. 2021, 179, 101–115. [Google Scholar] [CrossRef]
  197. Plum, L.M.; Rink, L.; Haase, H. The Essential Toxin: Impact of Zinc on Human Health. Int. J. Environ. Res. Public Health 2010, 7, 1342–1365. [Google Scholar] [CrossRef] [Green Version]
  198. Rizvi, S.F.A.; Wasim, B.; Usman, S.; Borges, K.J.J.; Sahibdad, I.; Salim, A.; Khan, I. Zinc and hypoxic preconditioning: A strategy to enhance the functionality and therapeutic potential of bone marrow-derived mesenchymal stem cells. Mol. Cell. Biochem. 2022. [Google Scholar] [CrossRef]
  199. Gao, P.; Fan, B.; Yu, X.; Liu, W.; Wu, J.; Shi, L.; Yang, D.; Tan, L.; Wan, P.; Hao, Y.; et al. Biofunc-tional magnesium coated Ti6Al4V scaffold enhances osteogenesis and angiogenesis and for orthopedic application. Bioact. Mater. 2020, 5, 680–693. [Google Scholar] [CrossRef]
  200. Wang, Y.; Gan, Z.; Lu, H.; Liu, Z.; Shang, P.; Zhang, J.; Yin, W.; Chu, H.; Yuan, R.; Ye, Y.; et al. Impact of High-Altitude Hypoxia on Early Osseointegration With Bioactive Titanium. Front. Physiol. 2021, 12, 689807. [Google Scholar] [CrossRef]
  201. Yu, Y.; Jin, G.; Xue, Y.; Wang, D.; Liu, X.; Sun, J. Multifunctions of dual Zn/Mg ion co-implanted titanium on osteogenesis, angiogenesis and bacteria inhibition for dental implants. Acta Biomater. 2017, 49, 590–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Ding, Z.; Qiao, Y.; Peng, F.; Xia, C.; Qian, S.; Wang, T.; Sun, J.; Liu, X. Si-doped porous TiO2 coatings enhanced in vitro angiogenic behavior of human umbilical vein endothelial cells. Colloids Surf. B Biointerfaces 2017, 159, 493–500. [Google Scholar] [CrossRef] [PubMed]
  203. Monte, F.; Cebe, T.; Ripperger, D.; Ighani, F.; Kojouharov, H.; Chen, B.M.; Kim, H.K.W.; Aswath, P.B.; Varanasi, V.G. Ionic silicon improves endothelial cells’ survival under toxic oxidative stress by overexpressing angiogenic markers and antioxidant enzymes. J. Tissue Eng. Regen. Med. 2018, 12, 2203–2220. [Google Scholar] [CrossRef] [PubMed]
  204. Ma, C.; Wei, Q.; Cao, B.; Cheng, X.; Tian, J.; Pu, H.; Yusufu, A.; Cao, L. A multifunctional bioactive material that stimulates oste-ogenesis and promotes the vascularization bone marrow stem cells and their resistance to bacterial infection. PLoS ONE 2017, 12, e0172499. [Google Scholar]
  205. Jia, P.; Chen, H.; Kang, H.; Qi, J.; Zhao, P.; Jiang, M.; Guo, L.; Zhou, Q.; Qian, N.D.; Zhou, H.B.; et al. Deferoxamine released from poly(lactic-co-glycolic acid) promotes healing of osteoporotic bone defect via enhanced angio-genesis and osteogenesis. J. Biomed. Mater. Res. A 2016, 104, 2515–2527. [Google Scholar] [CrossRef]
  206. Zhang, W.; Li, G.; Deng, L.; Qiu, S.; Deng, R. New bone formation in a true bone ceramic scaffold loaded with desferrioxamine in the treatment of segmental bone defect: A preliminary study. J. Orthop. Sci. 2012, 17, 289–298. [Google Scholar] [CrossRef]
  207. Stewart, R.; Goldstein, J.; Eberhardt, A.; Chu, G.T.-M.G.; Gilbert, S. Increasing Vascularity to Improve Healing of a Segmental Defect of the Rat Femur. J. Orthop. Trauma 2011, 25, 472–476. [Google Scholar] [CrossRef] [Green Version]
  208. Ran, Q.; Yu, Y.; Chen, W.; Shen, X.; Mu, C.; Yuan, Z.; Tao, B.; Hu, Y.; Yang, W.; Cai, K. Deferoxamine loaded titania nanotubes substrates regulate osteogenic and angiogenic differentiation of MSCs via activation of HIF-1α signaling. Mater. Sci. Eng. C 2018, 91, 44–54. [Google Scholar] [CrossRef]
  209. Li, H.; Luo, B.; Wen, W.; Zhou, C.; Tian, L.; Ramakrishna, S. Deferoxamine immobilized poly(D,L-lactide) membrane via poly-dopamine adhesive coating: The influence on mouse embryo osteoblast precursor cells and human umbilical vein endothelial cells. Mater. Sci. Engineering. C Mater. Biol. Appl. 2017, 70, 701–709. [Google Scholar] [CrossRef]
  210. Ding, H.; Chen, S.; Song, W.-Q.; Gao, Y.-S.; Guan, J.-J.; Wang, Y.; Sun, Y.; Zhang, C.-Q. Dimethyloxaloylglycine improves angio-genic activity of bone marrow stromal cells in the tissue-engineered bone. Int. J. Biol. Sci. 2014, 10, 746–756. [Google Scholar] [CrossRef] [Green Version]
  211. Jahangir, S.; Hosseini, S.; Mostafaei, F.; Sayahpour, F.A.; Eslaminejad, M.B. 3D-porous β-tricalcium phosphate–alginate–gelatin scaffold with DMOG delivery promotes angiogenesis and bone formation in rat calvarial defects. J. Mater. Sci. Mater. Med. 2018, 30, 1. [Google Scholar] [CrossRef]
  212. Shi, M.; Zhou, Y.; Shao, J.; Chen, Z.; Song, B.; Chang, J.; Wu, C.; Xiao, Y. Stimulation of osteogenesis and angiogenesis of hBMSCs by delivering Si ions and functional drug from mesoporous silica nanospheres. Acta Biomater. 2015, 21, 178–189. [Google Scholar] [CrossRef]
  213. Qi, X.; Liu, Y.; Ding, Z.-Y.; Cao, J.-Q.; Huang, J.-H.; Zhang, J.-Y.; Jia, W.-T.; Wang, J.; Liu, C.-S.; Li, X.-L. Synergistic effects of di-methyloxallyl glycine and recombinant human bone morphogenetic protein-2 on repair of critical-sized bone defects in rats. Sci. Rep. 2017, 7, 42820. [Google Scholar] [CrossRef] [Green Version]
  214. Min, Z.; Shichang, Z.; Chen, X.; Yufang, Z.; Changqing, Z. 3D-printed dimethyloxallyl glycine delivery scaffolds to improve angiogenesis and osteogenesis. Biomater. Sci. 2015, 3, 1236–1244. [Google Scholar] [CrossRef]
  215. Jin, X.; Han, D.; Tao, J.; Huang, Y.; Zhou, Z.; Zhang, Z.; Qi, X.; Jia, W. Dimethyloxallyl Glycine-Incorporated Borosilicate Bioac-tive Glass Scaffolds for Improving Angiogenesis and Osteogenesis in Critical-Sized Calvarial Defects. Curr. Drug Deliv. 2019, 16, 565–576. [Google Scholar] [CrossRef]
  216. Wu, C.; Zhou, Y.; Chang, J.; Xiao, Y. Delivery of dimethyloxallyl glycine in mesoporous bioactive glass scaffolds to improve angiogenesis and osteogenesis of human bone marrow stromal cells. Acta Biomater. 2013, 9, 9159–9168. [Google Scholar] [CrossRef]
  217. Zou, D.; Han, W.; You, S.; Ye, D.; Wang, L.; Wang, S.; Zhao, J.; Zhang, W.; Jiang, X.; Zhang, X.; et al. In vitro study of en-hanced osteogenesis induced by HIF-1alpha-transduced bone marrow stem cells. Cell Prolif. 2011, 44, 234–243. [Google Scholar] [CrossRef]
  218. Li, H.; Liu, D.; Li, C.; Zhou, S.; Tian, D.; Xiao, D.; Zhang, H.; Gao, F.; Huang, J. Exosomes secreted from mutant-HIF-1α-modified bone-marrow-derived mesenchymal stem cells attenuate early steroid-induced avascular necrosis of femoral head in rabbit. Cell Biol. Int. 2017, 41, 1379–1390. [Google Scholar] [CrossRef]
  219. Ying, C.; Wang, R.; Wang, Z.; Tao, J.; Yin, W.; Zhang, J.; Yi, C.; Qi, X.; Han, D. BMSC-Exosomes Carry Mutant HIF-1α for Improving Angiogenesis and Osteogenesis in Critical-Sized Calvarial Defects. Front. Bioeng. Biotechnol. 2020, 8, 565561. [Google Scholar] [CrossRef]
  220. Zou, D.; Zhang, Z.; He, J.; Zhu, S.; Wang, S.; Zhang, W.; Zhou, J.; Xu, Y.; Huang, Y.; Wang, Y.; et al. Repairing critical-sized calvarial defects with BMSCs modified by a constitutively active form of hypoxia-inducible factor-1α and a phosphate cement scaffold. Biomaterials 2011, 32, 9707–9718. [Google Scholar] [CrossRef]
  221. Zou, D.; Zhang, Z.; Ye, D.; Tang, A.; Deng, L.; Han, W.; Zhao, J.; Wang, S.; Zhang, W.; Zhu, C.; et al. Repair of Critical-Sized Rat Calvarial Defects Using Genetically Engineered BMSCs Overexpressing HIF-1α. Stem Cells 2011, 29, 1380–1390. [Google Scholar] [CrossRef] [PubMed]
  222. Zou, D.; Zhang, Z.; He, J.; Zhang, K.; Ye, D.; Han, W.; Zhou, J.; Wang, Y.; Li, Q.; Liu, X.; et al. Blood vessel formation in the tissue-engineered bone with the constitutively active form of HIF-1α mediated BMSCs. Biomaterials 2011, 33, 2097–2108. [Google Scholar] [CrossRef]
  223. West, X.Z.; Malinin, N.L.; Merkulova, A.A.; Tischenko, M.; Kerr, B.A.; Borden, E.C.; Podrez, E.A.; Salomon, R.G.; Byzova, T.V. Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands. Nature 2010, 467, 972–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Zhou, Q.; Gu, X.; Dong, J.; Zhu, C.; Cai, Z.; He, D.; Yang, C.; Xu, L.; Zheng, J. The use of TLR2 modified BMSCs for enhanced bone regeneration in the inflammatory micro-environment. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3329–3337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Citri, A.; Yarden, Y. EGF–ERBB signalling: Towards the systems level. Nat. Rev. Mol. Cell Biol. 2006, 7, 505–516. [Google Scholar] [CrossRef]
  226. Genetos, D.C.; Rao, R.R.; Vidal, M.A. Betacellulin inhibits osteogenic differentiation and stimulates proliferation through HIF-1α. Cell Tissue Res. 2010, 340, 81–89. [Google Scholar] [CrossRef]
  227. Li, Y.; Yang, F.; Gao, M.; Gong, R.; Jin, M.; Liu, T.; Sun, Y.; Fu, Y.; Huang, Q.; Zhang, W.; et al. miR-149-3p Regulates the Switch between Adipogenic and Os-teogenic Differentiation of BMSCs by Targeting FTO, Molecular Therapy. Nucleic Acids 2019, 17, 590–600. [Google Scholar] [CrossRef] [Green Version]
  228. Sun, R.; Zhang, C.; Liu, Y.; Chen, Z.; Liu, W.; Yang, F.; Zeng, F.; Guo, Q. Demethylase FTO promotes mechanical stress induced osteogenic differentiation of BMSCs with up-regulation of HIF-1α. Mol. Biol. Rep. 2022, 49, 2777–2784. [Google Scholar] [CrossRef]
  229. Zhu, H.; Wang, X.; Han, Y.; Zhang, W.; Xin, W.; Zheng, X.; Zhang, J. Icariin promotes the migration of bone marrow stromal cells via the SDF-1α/HIF-1α/CXCR4 pathway. Drug Des. Dev. Ther. 2018, 12, 4023–4031. [Google Scholar] [CrossRef]
  230. Zhang, L.-L.; Tian, K.; Tang, Z.-H.; Chen, X.-J.; Bian, Z.-X.; Wang, Y.-T.; Lu, J.-J. Phytochemistry and Pharmacology of Cartha-mus tinctorius L. Am. J. Chin. Med. 2016, 44, 197–226. [Google Scholar] [CrossRef]
  231. Tang, Z.; Xie, H.; Jiang, S.; Cao, S.; Pu, Y.; Zhou, B.; Zhang, X.; Xiong, H. Safflower yellow promotes angiogenesis through p-VHL/ HIF-1alpha/VEGF signaling pathway in the process of osteogenic differentiation. Biomed. Pharmacother. 2018, 107, 1736–1743. [Google Scholar] [CrossRef]
  232. Wang, X.; Zhang, Y.; Yang, Y.; Zhang, W.; Luo, L.; Han, F.; Guan, H.; Tao, K.; Hu, D. Curcumin pretreatment protects against hypoxia/reoxgenation injury via improvement of mitochondrial function, destabilization of HIF-1α and activation of Epac1-Akt pathway in rat bone marrow mesenchymal stem cells. Biomed. Pharmacother. 2018, 109, 1268–1275. [Google Scholar] [CrossRef]
  233. Guo, Q.; Yang, J.; Chen, Y.; Jin, X.; Li, Z.; Wen, X.; Xia, Q.; Wang, Y. Salidroside improves angiogenesis-osteogenesis coupling by regulating the HIF-1α/VEGF signalling pathway in the bone environment. Eur. J. Pharmacol. 2020, 884, 173394. [Google Scholar] [CrossRef]
  234. Pauly, S.; Luttosch, F.; Morawski, M.; Haas, N.P.; Schmidmaier, G.; Wildemann, B. Simvastatin locally applied from a biode-gradable coating of osteosynthetic implants improves fracture healing comparable to BMP-2 application. Bone 2009, 45, 505–511. [Google Scholar] [CrossRef]
  235. Fukui, T.; Ii, M.; Shoji, T.; Matsumoto, T.; Mifune, Y.; Kawakami, Y.; Akimaru, H.; Kawamoto, A.; Kuroda, T.; Saito, T.; et al. Therapeutic effect of local administration of low-dose simvastatin-conjugated gelatin hydrogel for fracture healing. J. Bone Miner. Res. 2012, 27, 1118–1131. [Google Scholar] [CrossRef]
  236. Rojbani, H.; Nyan, M.; Ohya, K.; Kasugai, S. Evaluation of the osteoconductivity of α-tricalcium phosphate, β-tricalcium phosphate, and hydroxyapatite combined with or without simvastatin in rat calvarial defect. J. Biomed. Mater. Res. Part A 2011, 98, 488–498. [Google Scholar] [CrossRef] [PubMed]
  237. Yueyi, C.; Xiaoguang, H.; Jingying, W.; Quansheng, S.; Jie, T.; Xin, F.; Yingsheng, X.; Chunli, S. Calvarial defect healing by re-cruitment of autogenous osteogenic stem cells using locally applied simvastatin. Biomaterials 2013, 34, 9373–9380. [Google Scholar] [CrossRef]
Figure 1. Hypoxia-inducible factor-1α (HIF-1α) plays a bi-directional regulatory role in differentiating macrophages into osteoclasts. Nuclear Factor-κB Ligand (RANKL)/Notch1 is a crucial signal pathway for inducing macrophages to differentiate into osteoclasts. Hypoxia upregulates HIF-1α, inhibiting autophagosomes formation in macrophages, promoting the expression of RANKL by activating the Janus kinase 2 (JAK2)/Signal Transducers and Activators of Transcription 3 (STAT3) pathway in osteoblast. In addition, HIF-1α also activates the c-Jun N-terminal kinase (JNK)/caspase-3 pathway in osteocytes and stimulates the RANKL/Notch1 signal pathway. However, miR-34a-5p inhibits RANKL/Notch1 signal pathway under HIF-1α/interleukin 33 (IL-33) stimulation.
Figure 1. Hypoxia-inducible factor-1α (HIF-1α) plays a bi-directional regulatory role in differentiating macrophages into osteoclasts. Nuclear Factor-κB Ligand (RANKL)/Notch1 is a crucial signal pathway for inducing macrophages to differentiate into osteoclasts. Hypoxia upregulates HIF-1α, inhibiting autophagosomes formation in macrophages, promoting the expression of RANKL by activating the Janus kinase 2 (JAK2)/Signal Transducers and Activators of Transcription 3 (STAT3) pathway in osteoblast. In addition, HIF-1α also activates the c-Jun N-terminal kinase (JNK)/caspase-3 pathway in osteocytes and stimulates the RANKL/Notch1 signal pathway. However, miR-34a-5p inhibits RANKL/Notch1 signal pathway under HIF-1α/interleukin 33 (IL-33) stimulation.
Cells 11 03552 g001
Figure 2. Hypoxia affects the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts by regulating HIF-1α expression. An amount of 5% O2 up-regulates HIF-1α; 1% O2 downregulates HIF-1α. In the process of HIF-1α affecting BMSCs osteogenic differentiation, core binding factor α1 (Cbfα1), bone morphogenetic protein 2 (BMP2), runt-related transcription factor 2 (RUNX2), mechano growth factor (MGF), phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), MEK-extracellular regulated protein kinase 1/2 (ERK1/2), pyruvate dehydrogenase kinase 1 (PDK-1), and Wnt/β-catenin play a positive regulatory role, while Notch1, C/EBP, TWIST, and AKT/mTOR play a negative regulatory role.
Figure 2. Hypoxia affects the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts by regulating HIF-1α expression. An amount of 5% O2 up-regulates HIF-1α; 1% O2 downregulates HIF-1α. In the process of HIF-1α affecting BMSCs osteogenic differentiation, core binding factor α1 (Cbfα1), bone morphogenetic protein 2 (BMP2), runt-related transcription factor 2 (RUNX2), mechano growth factor (MGF), phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), MEK-extracellular regulated protein kinase 1/2 (ERK1/2), pyruvate dehydrogenase kinase 1 (PDK-1), and Wnt/β-catenin play a positive regulatory role, while Notch1, C/EBP, TWIST, and AKT/mTOR play a negative regulatory role.
Cells 11 03552 g002
Figure 3. The intracellular regulation mechanism of HIF-1α. Cobalt (Co) ion, deferoxamine (DFO), and other chemicals affect HIF-1α gene expression and hinder HIF-1α degradation by inhibiting the activity of proline hydroxylase (PHD). MiRNAs and lncRNAs participate in the regulation of HIF-1α through a variety of mechanisms, such as Wnt/β-catenin, phosphate, and tension homology deleted on chromosome ten (PTEN)/PI3K/Akt, Smad1/5/8 signal pathways, extracellular vesicles, and epigenetic mechanisms, and exert biological functions such as osteogenesis, angiogenesis, and hematopoiesis.
Figure 3. The intracellular regulation mechanism of HIF-1α. Cobalt (Co) ion, deferoxamine (DFO), and other chemicals affect HIF-1α gene expression and hinder HIF-1α degradation by inhibiting the activity of proline hydroxylase (PHD). MiRNAs and lncRNAs participate in the regulation of HIF-1α through a variety of mechanisms, such as Wnt/β-catenin, phosphate, and tension homology deleted on chromosome ten (PTEN)/PI3K/Akt, Smad1/5/8 signal pathways, extracellular vesicles, and epigenetic mechanisms, and exert biological functions such as osteogenesis, angiogenesis, and hematopoiesis.
Cells 11 03552 g003
Figure 5. Application of HIF-1α in biomaterials. Organic/inorganic biomaterials can stabilize HIF-1α by doping trace elements such as copper (Cu) and Co or carrying chemical drugs such as dimethyloxalylglycine (DMOG) and DFO. The signal pathways involved include Wnt/β-catenin, ERK1/2, PI3K/AKT, and JAK1/STAT1, which upregulate downstream osteogenic and angiogenic genes.
Figure 5. Application of HIF-1α in biomaterials. Organic/inorganic biomaterials can stabilize HIF-1α by doping trace elements such as copper (Cu) and Co or carrying chemical drugs such as dimethyloxalylglycine (DMOG) and DFO. The signal pathways involved include Wnt/β-catenin, ERK1/2, PI3K/AKT, and JAK1/STAT1, which upregulate downstream osteogenic and angiogenic genes.
Cells 11 03552 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chen, W.; Wu, P.; Yu, F.; Luo, G.; Qing, L.; Tang, J. HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases. Cells 2022, 11, 3552. https://doi.org/10.3390/cells11223552

AMA Style

Chen W, Wu P, Yu F, Luo G, Qing L, Tang J. HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases. Cells. 2022; 11(22):3552. https://doi.org/10.3390/cells11223552

Chicago/Turabian Style

Chen, Wei, Panfeng Wu, Fang Yu, Gaojie Luo, Liming Qing, and Juyu Tang. 2022. "HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases" Cells 11, no. 22: 3552. https://doi.org/10.3390/cells11223552

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

Article Metrics

Back to TopTop