HIF‑1α: Its notable role in the maintenance of oxygen, bone and iron homeostasis (Review)

  • Authors:
    • Xinyi Huang
    • Yili Zhang
    • Baoyu Qi
    • Kai Sun
    • Ning Liu
    • Bin Tang
    • Shengjie Fang
    • Liguo Zhu
    • Xu Wei
  • View Affiliations

  • Published online on: October 27, 2022     https://doi.org/10.3892/ijmm.2022.5197
  • Article Number: 141
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxia is a characteristic feature of numerous diseases, including metabolic bone disease, solid tumors, cardiovascular diseases, neurodegeneration and inflammation. It is also a risk factor for a poor prognosis in various diseases. Hypoxia‑inducible factor‑1α (HIF‑1α) is activated by hypoxia to regulate a series of pathophysiological pathways, which is of utmost significance for maintaining body homeostasis. The present review highlights the role of the HIF‑1α in oxygen, bone and iron homeostasis, and alludes on the biological complexity and dual functions of HIF‑1α regulation. In addition, the pathophysiological significance of HIF‑1α in bone formation, bone absorption, angiogenesis, erythropoiesis, oxidative stress, energy metabolism, iron death, etc., is discussed An accurate understanding of all these processes may aid in the identification of possible therapeutic targets that may then be used in the treatment of related diseases. However, further studies are required to unravel the extensive complexity of HIF‑1α regulation and to develop more precise treatment strategies.

Introduction

Hypoxia-inducible factor (HIF) heterodimers consist of one of three α-subunits (HIF-1α, HIF-2α and HIF-3α) and one β-subunit. HIF-1α, a 120 kDa polypeptide subunit that heterodimerizes with HIF-1β (a 91 to 94 kDa polypeptide subunit), is a transcription factor regulated by hypoxia (1). Under normoxic conditions, HIF-1α is hydroxylated to interact with von Hippel-Lindau (VHL) protein for ubiquitination and proteasomal degradation. HIF-1a is expressed in almost all cell types, whereas HIF-2a has a more limited distribution. Under hypoxic conditions, HIF-1α plays a crucial role in the body's metabolic and functional adaptation to these conditions. All these observations have allowed the identification of HIF-1a as a critical factor in the regulation of homeostasis. It is worth noting that in the field of integrative physiology, research on baroreflex, chemoreflex, glucose regulation and temperature regulation is essentially the study of a series of homeostasis (24). Among these, oxygen, bone and iron homeostasis are involved in various critical functions of the body, including bone resorption and formation, mesenchymal stem cell (MSC) homing, angiogenesis, erythropoiesis, oxidative stress, iron metabolism and ferroptosis.

Bone homeostasis is maintained by a balance between osteoblast-mediated bone formation and osteoclast-driven bone resorption (5). Under hypoxic conditions, HIF-1α exerts a series of direct and indirect effects on this balance (6). Further studies have indicated its critical role in the manipulation of bone mass accrual, bone material properties as well as micro-structures, including bone mineralization, bone collagen fiber formation and bone remodeling (7). Moreover, HIF-1α is a master regulator of oxygen homeostasis in the body, which can induce the expression of angiogenic factors, promote glycolysis, increase the delivery of oxygen and nutrients (8). HIF-1α also plays a key role in iron homeostasis by activating the transcription of iron metabolism genes, such as transferrin (Tf), transferrin receptor (TFR), ceruloplasmin and heme oxygenase 1 (HO-1) (9,10). Roxadustat, a HIF-prolyl hydroxylase inhibitor, has been shown to improve iron metabolism in phase 3 trials (11,12).

However, over the years, although the association between HIF-1α and oxygen, bone and iron homeostasis has been the subject of increasing attention, no consensus has yet been reached on the role of HIF-1α, at least to the best of our knowledge. Research into its effects on osteocyte apoptosis and osteocyte-mediated osteoclasts has also yielded non-univocal results (1315). In addition, the local activation of HIF-1α is required for chondrocyte survival in the center of the expanding growth plate; however, the cellular-intrinsic mechanisms remain unclear (16) The expression of the majority of HIF-1α-dependent genes contributes to the adaption of hypoxic environments in the human body. For example, the increase in the delivery of oxygen to hypoxic tissues is associated with the expression of erythropoietin (EPO) and glycolytic enzymes, which allows for the increased conversion of glucose to produce energy (17). However, HIF-1α can also play a negative role in the hypoxic process.

Overall, considering the numerous processes in which HIF-1α is involved and the yet not fully defined underlying mechanisms, the present review focused on the intimate association between HIF-1α and bone homeostasis, oxygen homeostasis, as well as iron homeostasis. In addition, the pathophysiological significance of HIF-1α in bone formation, bone absorption, angiogenesis, erythropoiesis, oxidative stress, energy metabolism, iron death, etc. is also discussed (Fig. 1). HIF-1α is a promising target for the treatment of related diseases, and further information is required to determine the clinical utility of this factor.

Oxygen homeostasis

HIF-1α, mediating the expression of a series of genes, has been strongly established as a critical factor for maintaining oxygen homeostasis. The regulation of oxygen homeostasis is considered to be achieved by oxygen delivery and oxygen. Oxygen delivery is involved in the control of erythropoiesis, angiogenesis and vascular remodeling. Oxygen utilization is implicated in the regulation of glucose metabolism and redox homeostasis (18).

Oxygen delivery

Vascular endothelial growth factor (VEGF) is the most potent proangiogenic factor. EPO, a glycoprotein, is considered as the principal stimulator for erythropoiesis primarily. The expression of HIF-1α is induced by a hypoxic environment, and it subsequently upregulates downstream key factors, such as EPO and VEGF, which promote angiogenesis to adapt to the environment and recover the oxygen content.

The primary cause of the ectopic overexpression of VEGF in tumors is the dysregulated expression of HIF-1α involving the c-X-c chemokine receptor type 4 (CXCR4)/stromal-derived cell factor-1 (SDF-1) axis (19). The study by Li et al (20) conducted on cerebral ischemic rats, found that HIF-1α attenuated neuronal apoptosis, partially by upregulating EPO expression. There is a novel molecular mechanism for the anti-angiogenic effects of peroxisome proliferator-activated receptor α, which are achieved by inhibiting ischemia-induced EPC mobilization and homing through the inhibition of the HIF-1α/SDF-1 pathway (21). Rankin et al (22) found that osterix-VHL mice with a deficiency in VHL in osteoblasts exhibited overexpressed HIFs, accompanied by a significant increase in circulating red blood cells. Gerri et al (23) reported that HIF-1α regulated hematopoietic stem cells upstream of the Notch signaling pathway.

Oxygen utilization

HIF-1α, in response to hypoxic irritation, participates in the regulation of glucose transporters and glycolytic enzymes, which are key genes in energy metabolism and exert critical effects on cell survival (24). Moreover, HIF-1α inhibits pyruvate dehydrogenase by activating pyruvate dehydrogenase kinase 1 (PDK1), and thereby, pyruvate is redirected from the tricarboxylic acid (TCA) cycle and converted into lactate (25).

The overexpression of constitutive cardiac-specific HIF-1α leads to changes in cellular metabolism and increased glucose utilization, subsequently resulting in cardiomyopathy in aging mice (26). On the other hand, the deletion of HIF-1α in cardiomyocytes results in decreased ATP, lactate and phosphocreatine levels, and inn an impaired myocardial contractility (27). Chondrocytes maintain an optimal energy balance during endochondral ossification, which is achieved by confined HIF-1α signaling (28). However, it is only under hypoxic conditions that glucose uptake and bone resorption can be affected by HIF-1α knockdown. HIF-1α promotes glycolysis during hypoxia; however, it also affects metabolism under normoxic conditions. A decreased HIF-1α activity also has effects on mitochondrial metabolism that results in mitochondrial loss and lipid accumulation, along with reduced oxidative phosphorylation and fatty acid metabolism (26,29). In addition, studies have demonstrated that HIF-dependent metabolic processes can also modulated by dimethyloxalylglycine, desferrioxamine, prolyl hydroxylase (PHD) and other small molecules (30,31).

Oxidative stress

HIF-1α is an endogenous anti-oxidative stress modulator. The oxidative stress pathway induces the activation of HIF-1α, and increases the production of mitochondrial complex II-mediated reactive oxygen species (ROS) (32,33). Moreover, it has been demonstrated that increased superoxide anion radicals induce PHD inactivation, resulting in the stabilization and accumulation of HIF-1α (34). Under hypoxic conditions, HIF-1α dynamically regulates glucose flux through the glycolytic pathway to resist the increased risk of ROS production and confers protection against apoptosis and renal injury in diabetes (35,36).

In recent years, increasing evidence has indicated that HIF-1α can enhance antioxidant activity and neuroprotection (37,38). HIF-1α has the ability to mitigate this toxicity or regulate redox homeostasis by limiting TCA activity, regulating the levels of NADPH and glutathione (GSH), and reducing mitochondrial mass through the upregulation of the mitochondrial proteins, PDK1 and Bcl-2 interacting protein 3 (BNIP3) (39). Furthermore, HIF-1α may be an indirect player in the promotion of mitochondrial-selective autophagy and may subsequently lower the mitochondrial mass, which inhibits the oxidation of both glucose and fatty acids, and reduces mitochondrial ROS production under hypoxic conditions (40). A previous study revealed that HIF-1α can activate the nuclear factor erythroid 2-related factor 2 (Nrf2)/ARE pathway to protect against ischemia-reperfusion cardiac and skeletal muscle injuries (41).

Bone homeostasis

Bone formation

Previous research has indicated that HIF-1α may affect the osteogenesis of osteoblasts through the prevention of chondrocyte cell death in the growth plate, and also via direct or indirect actions on the delivery of oxygen and nutrients, together with metabolic adaptations (8). It has been reported that the overexpression of HIF-1α, through its downstream marker, BNIP3, reduces the inhibitory effects of dexamethasone on hypoxia-induced mitophagy and protects osteocytes from apoptosis (42). There is also evidence to suggest that miRNA-21, by upregulating the activation of HIF-1α and p-Akt, can promote the osteogenic ability of bone MSCs (BMSCs) (43). HIF-1α does not only promote osteogenesis, but also has repairing effects on bone (44). Moreover, it has been demonstrated that prolonged HIF-1α signaling in chondrocytes, interfering with cellular bioenergetics and biosynthesis, results in skeletal dysplasia by collagen overmodification (27).

Bone resorption

The delicate balance between osteoblastic bone formation and osteoclastic bone resorption is a key factor in the regulation of mature bone tissue formation (45). Nevertheless, no consensus has yet been reached on the role of HIF-1α in regulating osteoclast differentiation, at least to the best of our knowledge.

Promotion of osteoclast differentiation

HIF-1α expression has been proposed to increase bone erosion in rheumatoid arthritis (46). HIF-1α is involved in the increase of osteoclastogenesis and bone resorption, since it has recently been shown to enhance the osteoclast-mediated stimulation of BMSC differentiation by secreting cardiotrophin-1 (47). Moreover, HIF-1α functions by activating the JAK2/STAT3 pathway, promoting the expression of RANKL, and thus enhancing the differentiation of osteocyte-mediated osteoclastic in vitro (48). HIF-1α also plays a pro-apoptotic role in JNK/caspase-3 signaling pathway activation. Osteocyte-mediated osteoclastogenesis has been shown to be reduced with a concomitant decrease in HIF-1α and caspase-3 expression (49). A previous study demonstrated that the deceleration of osteoclastogenesis occurred under conditions of HIF-1α deficiency, by inhibiting AMPK signaling under anoxic conditions (50). Of note, HIF-1α knockdown reduces bone resorption under both normoxic and hypoxic conditions. Thus, the targeting of HIF-1α may prove to be of value in th treatment of osteoporosis (13).

Inhibition of osteoclast differentiation

Both the VHL/HIF and PHD/HIF signaling pathways in osteoblasts have been shown to reduce osteoclastogenesis by increasing osteoprotegerin expression and inhibiting sclerostin expression, resulting in increased bone formation and decreased resorption (14,51). In addition, it has been suggested that the activation of osteoblast HIF-1α contributes to the inhibition of osteoclastogenesis, by increasing IL-33 expression (52).

Angiogenesis

A vital role in bone remodeling and vascularization is attributed to the HIF-1α/VEGF signaling pathway (53). The study by Kusumbe et al (54) demonstrated endothelial HIF-1α as a critical promoter of type H vessel formation in the metaphysis. HIF-1α has also been implicated in the increased number of type-H vessels and the restoration of bone mass (5557). The miR-497B195 cluster has been proposed to regulate angiogenesis during coupling with osteogenesis, by maintaining endothelial Notch and HIF-1α activity (58). Furthermore, HIF-1α may have a dual function in the regulation of osteogenesis-angiogenesis coupling of long bone via the ROS-HIF1α/p53 axis (59).

MSC migration

HIF-1α has also been demonstrated to trigger wound healing and functional recovery by regulating corresponding stem cells (60,61). BMSCs, a class of heterogeneous cells, have a series of feasible and diverse clinical values for generating stroma which can support hematopoiesis, bones, adipocytes and cartilage (62).

HIF-1α regulates the expression levels of surface molecules, such as SDF-1, a downstream gene of HIF-1α, which binds to its specific receptor, CXCR4, forming a pair of coupling molecules and promoting stem cell migration to ischemic and hypoxic sites (63). Guo et al (62) demonstrated that the HIF-1α/SDF-1/CXCR4 axis enhanced BMSC migration, and alleviated neuronal damage and apoptosis. Moreover, there is evidence to suggest that the increased secretion of HIF-1α induced by the hypoxic conditions of surrounding brain tissue accelerates the fracture repair process via chemotaxis due to the SDF-1/CXCR4 axis. In addition, the silencing of HIF-1α has been shown to decrease MSC migration, as well as the mRNA and protein levels of SDF-1 and CXCR4 in MSCs (64).

Iron homeostasis

Iron metabolism

HIF-1α is a vital factor in iron metabolism by regulating the expression of iron-related proteins, such as divalent metal transporter 1, ferroportin 1, duodenal cytochrome B and TFR (65,66). An overload of iron has been found to be related to the dysfunction of MSCs and to the damage of the microenvironment that may be involved in the pathogenesis of myelodysplastic syndromes, and which may be achieved by the regulation of cytokine of MSCs through the ROS/HIF-1α pathway (67). It has been demonstrated that HIF-1α induces TFR1 expression, thereby increasing iron uptake (68). In addition, it has been suggested that HO-1, induced by HIF-1α, degrades heme into biliverdin, carbon monoxide and iron (69). Weinreb et al (70) and Guo et al (71) demonstrated that in various brain regions of adult mice, the upregulation of HIF-1α by the iron chelator, M30, results in differentially induced levels of TFR, tyrosine hydroxylase and EPO (72).

Ferroptosis

Ferroptosis is a new form of regulated cell death as a result of iron-dependent lipid peroxidation (73). Moreover, HIF-1α downregulation also promotes ferroptosis by inducing ferritin heavy chain degradation in RANKL-stimulated bone marrow-derived macrophages. A previous in vitro study demonstrated that the overexpression of inhibitor of apoptosis-stimulating protein of p53 inhibited ferroptosis through the Nrf2/HIF-1α/TF signaling pathway (74). In another study, following pre-treatment with roxadustat (an inhibitor of HIF prolyl hydroxylase), the risk of ferroptosis was correspondingly reduced, along with increased levels of antioxidant enzymes and GSH, and decreased iron accumulation (75).

Summary and future perspectives

Constant oxygen supply is essential for proper tissue function, development and homeostasis. Hypoxia is a distinctive feature of diseases, including solid tumors, metabolic bone disease, cardiovascular diseases, neurodegeneration, inflammation and other chronic diseases (76,77). It is also a risk factor for a poor prognosis in various diseases. For example, hypoxia is responsible for the failure of the majority of solid tumors to respond to treatment, and promotes drug resistance (78). Under normoxic conditions, HIF is rapidly degraded by the high activity of PHDs. However, under hypoxic conditions, the shortage of oxygen results in the dimerization of non-hydroxylated and non-degradable HIF-1α with HIF-1β, which binds to hypoxia-responsive elements in the regulatory regions of oxygen-sensitive genes. Given the ubiquitous localization of HIF-1α, HIF-1α acts as a main regulator in the expression of several thousand genes coding, including growth factors, enzymes, transcription factors, cytokines, hormones, receptors, solute transporters, ion channels and other essential regulators, which are involved in almost every cell function or dysfunction (79).

There is no doubt that bone, oxygen and iron homeostasis are of utmost significance to the human body, and the role of HIF-1α in the maintenance of homeostasis cannot be ignored. Although HIF-1α plays a beneficial role in the regulation of bone homeostasis, the degree of HIF-1α pathway activation must be fine-tuned to avoid the disruption of homeostasis (8,80). Previous studies have confirmed that osteoblastic HIF-1α affects bone formation (8183); however, its role in osteoclasts remains controversial. An interesting aspect is that HIF-1α has minimal effects on osteoclast differentiation, although it predominantly functions as a regulator of osteoclast-mediated bone resorption (84). Similarly, the knockdown of HIF-1α does not affect the process of osteoclast differentiation, although it prevents the increased bone resorption under hypoxic conditions (6). Moreover, HIF-1α has been demonstrated to regulate osteogenesis-angiogenesis coupling bidirectionally, and the effect is age-related (56). There is also evidence to suggest that HIF-1α functions by increasing EPO levels directly or indirectly, inducing the expression and processing of fibroblast growth factor23 (FGF23), and thus affecting mineral homeostasis and vitamin D metabolism (8). Of note, increased serum FGF23 levels have been reported to be associated with the reduction of serum phosphate or 1,25(OH)2D, which in turn may alter bone homeostasis, although further confirmation is required (85,86).

Taken together, HIF-1α expression is mainly induced by hypoxic stress and is common during the development of various diseases. The present review mainly focused on the role of HIF-1α in regulating oxygen, bone and iron homeostasis. Although significant progress has been made in the understanding of the pathogenesis of diseases, such as atherosclerosis and emerging drug treatments, the current treatment options continue to have a number of deficiencies. Regulating the expression and signaling pathways of HIF-1α may be a promising strategy for the treatment of diseases involving the pathophysiology of hypoxia (Table I). To date, a number of active ingredients of traditional Chinese medicine and natural products have been found to regulate the HIF-1α content (87). At present, HIF-1α inhibitors have been used to treat various diseases, such as tumors, leukemia, diabetes, ischemic cardiovascular and brain diseases, etc. Manassantin A and Manassantin B exert potent inhibitory effects on the secretion of hypoxia-induced VEGF, cyclin-dependent kinase inhibitor 1 and GLUT-1 genes (88,89). Lificiguat (YC-1) is a targeted HIF-1α inhibitor, which can reduce HIF-1α protein expression and is associated with the enhancement of EGFR degradation, thereby exerting antitumor effects (90). The benefit of S-nitrosoglutathione on traumatic brain injury is mediated by S-nitrosylation to stabilize HIF-1α (91).

Table I.

Mechanisms and effects of HIF-1α on various diseases.

Table I.

Mechanisms and effects of HIF-1α on various diseases.

Differential expression at protein/gene levelsMechanismsEffects(Refs.)
VEGFChemokine receptor 4/stromal-derived cell factor 1 (CXCR4/SDF-1) axisPromotes tumor angiogenesis(19)
EPOUpregulating EPOAttenuates neuronal apoptosis(20)
SDF-1HIF-1a/SDF-1 pathwayAnti-angiogenic effect(21)
PDK1Inactivates pyruvate dehydrogenase (PDH)Increases ATP levels and prevents toxic ROS production(39)
NADPH and GSHSwitches cells from oxidative to glycolytic metabolism, to reduce mitochondrial superoxide generationMaintains redox homeostasis(40)
Nrf2Nrf2/ARE pathwayProtects against ischemia-reperfusion cardiac and skeletal muscle injuries(41)
BNIP3Reduces the inhibitory effects of DEX on hypoxia-induced mitophagyProtects bone cells from apoptosis(42)
CT-1Enhances the osteoclast-mediated stimulation of BMSC differentiationBone resorption(47)
RANKLJAK2/STAT3 pathwayEnhances the differentiation of osteocyte-mediated osteoclastic(48)
IL-33Acts on bone marrow-derived monocytesContributes to osteoclastogenesis inhibition(52)
SDF-1HIF-1α/SDF-1/CXCR4 axisAlleviates neuronal damage and apoptosis(62)

[i] VEGF, vascular endothelial growth factor; EPO, erythropoietin; PDK1, pyruvate dehydrogenase kinase 1; DEX, dexamethasone; CT-1, cardiotrophin-1; IL-33, interleukin-33; SDF-1, stromal cell-derived factor-1; CXCR4, c-X-c chemokine receptor type 4; RANKL, receptor activator of nuclear factor kappa-B ligand; GSH, glutathione; BNIP3, Bcl-2 interacting protein 3; Nrf2, nuclear factor erythroid 2-related factor 2; RANKL, receptor activator of nuclear factor κΒ ligand.

Notably, immense efforts and resources have been invested in identifying possible effective and specific small-molecules inhibitors of HIF-1α. HIF-1α, as a common pathophysiological mechanism of numerous diseases, plays an exploratory role in the treatment of comorbidities. However, there are several questions and challenges involved in translating the findings from mechanobiological studies into novel HIF-1α-targeted therapeutics. The potential interaction network of multiple molecules regulates the expression of important genes. Important interactions between NF-κB and HIF-1α (9294) have been described recently. In addition, efficacy needs to be supported by high-quality clinical trial evidence.

HIF-1α is a master regulator of homeostasis, and plays critical roles in physiological and pathological processes. Understanding the roles and regulation mechanisms of HIF-1α in bone, oxygen and iron homeostasis may open a new era in the development of therapeutic strategies against a variety of pathologic conditions, such as metabolic bone disease, ischemic/hypoxic injuries, tumor growth, wound healing and cardiovascular remodeling.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 82174416 and 82205140), the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (grant no. ZYYCXTD-C-202003), the Basic Research Program of Jiangsu Province (Natural Science Foundation; grant no. BK20220468), the National Natural Science Foundation of China (NSFC) Matching Project of Nanjing University of Chinese Medicine (grant no. XPT82205140), and the Fundamental Research Funds for the Central Public Welfare Research Institutes (grant no. ZZ13-YQ-039).

Availability of data and materials

Not applicable.

Authors' contributions

XW and LZ were responsible for the conceptualization of the study. XH was responsible for the research design. XH and YZ were responsible for the determination of the research design. XH and YZ wrote the manuscript. BQ, KS and NL were responsible for the literature search. BT and SF completed the references and were involved in document management and preparation. YZ and XH prepared the original draft. XW and LZ reviewed and edited the manuscript. All authors contributed to the article and have read and approved the submitted version. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Tf

transferrin

TFR

transferrin receptor

HO-1

heme oxygenase 1

VEGF

vascular endothelial growth factor

EPO

erythropoietin

PDK1

pyruvate dehydrogenase kinase 1

TCA

tricarboxylic acid

PHD

prolyl hydroxylase

BMSCs

bone mesenchymal stem cells

SDF-1

stromal cell-derived factor-1

CXCR4

c-X-c chemokine receptor type 4

MSCs

mesenchymal stem cells

GSH

glutathione

FGF23

fibroblast growth factor 23

GSNO

S-nitrosoglutathione

References

1 

Greijer AE, van der Groep P, Kemming D, Shvarts A, Semenza GL, Meijer GA, van de Wiel MA, Belien JA, van Diest PJ and van der Wall E: Up-regulation of gene expression by hypoxia is mediated predominantly by hypoxia-inducible factor 1 (HIF-1). J Pathol. 206:291–304. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Bentley ER and Little SR: Local delivery strategies to restore immune homeostasis in the context of inflammation. Adv Drug Deliv Rev. 178:1139712021. View Article : Google Scholar : PubMed/NCBI

3 

Goldstein DS: How does homeostasis happen? Integrative physiological, systems biological, and evolutionary perspectives. Am J Physiol Regul Integr Comp Physiol. 316:R301–R317. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Suciadi LP, Henrina J, Putra ICS, Cahyadi I and Gunawan HFH: Chronic heart failure: Clinical implications of iron homeostasis disturbances revisited. Cureus. 14:e212242022.PubMed/NCBI

5 

Lee SY, Park KH, Yu HG, Kook E, Song WH, Lee G, Koh JT, Shin HI, Choi JY, Huh YH and Ryu JH: Controlling hypoxia-inducible factor-2α is critical for maintaining bone homeostasis in mice. Bone Res. 7:142019. View Article : Google Scholar : PubMed/NCBI

6 

Knowles HJ: Distinct roles for the hypoxia-inducible transcription factors HIF-1α and HIF-2α in human osteoclast formation and function. Sci Rep. 10:210722020. View Article : Google Scholar : PubMed/NCBI

7 

Chen S, Xiao L, Li Y, Qiu M, Yuan Y, Zhou R, Li C, Zhang L, Jiang ZX, Liu M and Zhou X: Osteocytic HIF-1α pathway manipulates bone micro-structure and remodeling via regulating osteocyte terminal differentiation. Front Cell Dev Biol. 9:7215612021. View Article : Google Scholar : PubMed/NCBI

8 

Stegen S and Carmeliet G: Hypoxia, hypoxia-inducible transcription factors and oxygen-sensing prolyl hydroxylases in bone development and homeostasis. Curr Opin Nephrol Hypertens. 28:328–335. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Das NK, Schwartz AJ, Barthel G, Inohara N, Liu Q, Sankar A, Hill DR, Ma X, Lamberg O, Schnizlein MK, et al: Microbial metabolite signaling is required for systemic iron homeostasis. Cell Metab. 31:115–130. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Galaris D, Barbouti A and Pantopoulos K: Iron homeostasis and oxidative stress: An intimate relationship. Biochim Biophys Acta Mol Cell Res. 1866:1185352019. View Article : Google Scholar : PubMed/NCBI

11 

Ikeda Y: Novel roles of HIF-PHIs in chronic kidney disease: The link between iron metabolism, kidney function, and FGF23. Kidney Int. 100:14–16. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Chen N, Hao C, Peng X, Lin H, Yin A, Hao L, Tao Y, Liang X, Liu Z, Xing C, et al: Roxadustat for anemia in patients with kidney disease not receiving dialysis. N Engl J Med. 381:1001–1010. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Ni S, Yuan Y, Qian Z, Zhong Z, Lv T, Kuang Y and Yu B: Hypoxia inhibits RANKL-induced ferritinophagy and protects osteoclasts from ferroptosis. Free Radic Biol Med. 169:271–282. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Shao J, Zhang Y, Yang T, Qi J, Zhang L and Deng L: HIF-1α disturbs osteoblasts and osteoclasts coupling in bone remodeling by up-regulating OPG expression. In Vitro Cell Dev Biol Anim. 51:808–814. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Meng X, Wielockx B, Rauner M and Bozec A: Hypoxia-inducible factors regulate osteoclasts in health and disease. Front Cell Dev Biol. 9:6588932021. View Article : Google Scholar : PubMed/NCBI

16 

Maes C, Araldi E, Haigh K, Khatri R, Van Looveren R, Giaccia AJ, Haigh JJ, Carmeliet G and Schipani E: VEGF-independent cell-autonomous functions of HIF-1α regulating oxygen consumption in fetal cartilage are critical for chondrocyte survival. J Bone Miner Res. 27:596–609. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Papandreou I, Cairns RA, Fontana L, Lim AL and Denko NC: HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 3:187–197. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Semenza GL: Hypoxia-inducible factor 1 and cardiovascular disease. Annu Rev Physiol. 76:39–56. 2014. View Article : Google Scholar : PubMed/NCBI

19 

de Nigris F, Crudele V, Giovane A, Casamassimi A, Giordano A, Garban HJ, Cacciatore F, Pentimalli F, Marquez-Garban DC, Petrillo A, et al: CXCR4/YY1 inhibition impairs VEGF network and angiogenesis during malignancy. Proc Natl Acad Sci USA. 107:14484–14489. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Li J, Tao T, Xu J, Liu Z, Zou Z and Jin M: HIF-1α attenuates neuronal apoptosis by upregulating EPO expression following cerebral ischemia-reperfusion injury in a rat MCAO model. Int J Mol Med. 45:1027–1036. 2020.PubMed/NCBI

21 

Wang Z, Moran E, Ding L, Cheng R, Xu X and Ma JX: PPARα regulates mobilization and homing of endothelial progenitor cells through the HIF-1α/SDF-1 pathway. Invest Ophthalmol Vis Sci. 55:3820–3832. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Rankin EB, Wu C, Khatri R, Wilson TL, Andersen R, Araldi E, Rankin AL, Yuan J, Kuo CJ, Schipani E and Giaccia AJ: The HIF signaling pathway in osteoblasts directly modulates erythropoiesis through the production of EPO. Cell. 149:63–74. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Gerri C, Marass M, Rossi A and Stainier DYR: Hif-1α and Hif-2α regulate hemogenic endothelium and hematopoietic stem cell formation in zebrafish. Blood. 131:963–973. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Jimenez-Blasco D, Busquets-Garcia A, Hebert-Chatelain E, Serrat R, Vicente-Gutierrez C, Ioannidou C, Gómez-Sotres P, Lopez-Fabuel I, Resch-Beusher M, Resel E, et al: Glucose metabolism links astroglial mitochondria to cannabinoid effects. Nature. 583:603–608. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Cerychova R and Pavlinkova G: HIF-1, Metabolism, and diabetes in the embryonic and adult heart. Front Endocrinol (Lausanne). 9:4602018. View Article : Google Scholar : PubMed/NCBI

26 

Hölscher M, Schäfer K, Krull S, Farhat K, Hesse A, Silter M, Lin Y, Pichler BJ, Thistlethwaite P, El-Armouche A, et al: Unfavourable consequences of chronic cardiac HIF-1α stabilization. Cardiovasc Res. 94:77–86. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Huang Y, Hickey RP, Yeh JL, Liu D, Dadak A, Young LH, Johnson RS and Giordano FJ: Cardiac myocyte-specific HIF-1alpha deletion alters vascularization, energy availability, calcium flux, and contractility in the normoxic heart. FASEB J. 18:1138–1140. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Stegen S, Laperre K, Eelen G, Rinaldi G, Fraisl P, Torrekens S, Van Looveren R, Loopmans S, Bultynck G, Vinckier S, et al: HIF-1α metabolically controls collagen synthesis and modification in chondrocytes. Nature. 565:511–515. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Ambrose LJ, Abd-Jamil AH, Gomes RS, Carter EE, Carr CA, Clarke K and Heather LC: Investigating mitochondrial metabolism in contracting HL-1 cardiomyocytes following hypoxia and pharmacological HIF activation identifies HIF-dependent and independent mechanisms of regulation. J Cardiovasc Pharmacol Ther. 19:574–585. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Semenza GL: Pharmacologic targeting of hypoxia-inducible factors. Annu Rev Pharmacol Toxicol. 59:379–403. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Knutson AK, Williams AL, Boisvert WA and Shohet RV: HIF in the heart: Development, metabolism, ischemia, and atherosclerosis. J Clin Invest. 131:e1375572021. View Article : Google Scholar : PubMed/NCBI

32 

Jiang L, Zeng H, Ni L, Qi L, Xu Y, Xia L, Yu Y, Liu B, Yang H, Hao H and Li P: HIF-1α preconditioning potentiates antioxidant activity in ischemic injury: The role of sequential administration of Dihydrotanshinone I and Protocatechuic aldehyde in Cardioprotection. Antioxid Redox Signal. 31:227–242. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Li X, Zhang Q, Nasser MI, Xu L, Zhang X, Zhu P, He Q and Zhao M: Oxygen homeostasis and cardiovascular disease: A role for HIF? Biomed Pharmacother. 128:1103382020. View Article : Google Scholar : PubMed/NCBI

34 

Wu LY, He YL and Zhu LL: Possible Role of PHD Inhibitors as Hypoxia-mimicking agents in the maintenance of neural stem cells' self-renewal properties. Front Cell Dev Biol. 6:1692018. View Article : Google Scholar : PubMed/NCBI

35 

Zheng X, Narayanan S, Xu C, Eliasson Angelstig S, Grünler J, Zhao A, Di Toro A, Bernardi L, Mazzone M, Carmeliet P, et al: Repression of hypoxia-inducible factor-1 contributes to increased mitochondrial reactive oxygen species production in diabetes. Elife. 11:e707142022. View Article : Google Scholar : PubMed/NCBI

36 

Semenza GL: Hypoxia-inducible factors: Coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J. 36:252–259. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Wu K, Zhou K, Wang Y, Zhou Y, Tian N, Wu Y, Chen D, Zhang D, Wang X, Xu H and Zhang X: Stabilization of HIF-1α by FG-4592 promotes functional recovery and neural protection in experimental spinal cord injury. Brain Res. 1632:19–26. 2016. View Article : Google Scholar : PubMed/NCBI

38 

He Q, Ma Y, Liu J, Zhang D, Ren J, Zhao R, Chang J, Guo ZN and Yang Y: Biological functions and regulatory mechanisms of hypoxia-inducible factor-1α in Ischemic Stroke. Front Immunol. 12:8019852021. View Article : Google Scholar : PubMed/NCBI

39 

Kim JW, Tchernyshyov I, Semenza GL and Dang CV: HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 3:177–185. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Samanta D and Semenza GL: Maintenance of redox homeostasis by hypoxia-inducible factors. Redox Biol. 13:331–335. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Ji W, Wang L, He S, Yan L, Li T, Wang J, Kong AT, Yu S and Zhang Y: Effects of acute hypoxia exposure with different durations on activation of Nrf2-ARE pathway in mouse skeletal muscle. PLoS One. 13:e02084742018. View Article : Google Scholar : PubMed/NCBI

42 

Xu K, Lu C, Ren X, Wang J, Xu P and Zhang Y: Overexpression of HIF-1α enhances the protective effect of mitophagy on steroid-induced osteocytes apoptosis. Environ Toxicol. 36:2123–2137. 2021. View Article : Google Scholar : PubMed/NCBI

43 

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. 10:652019. View Article : Google Scholar : PubMed/NCBI

44 

Yu Y, Ma L, Zhang H, Sun W, Zheng L, Liu C and Miao L: EPO could be regulated by HIF-1 and promote osteogenesis and accelerate bone repair. Artif Cells Nanomed Biotechnol. 48:206–217. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Nakashima T, Hayashi M and Takayanagi H: New insights into osteoclastogenic signaling mechanisms. Trends Endocrinol Metab. 23:582–590. 2012. View Article : Google Scholar : PubMed/NCBI

46 

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. 74:948–960. 2022. View Article : Google Scholar : PubMed/NCBI

47 

Tian Y, Shao Q, Tang Y, Li X, Qi X, Jiang R, Liang Y and Kang F: HIF-1α regulates osteoclast activation and mediates osteogenesis during mandibular bone repair via CT-1. Oral Dis. 28:428–441. 2022. View Article : Google Scholar : PubMed/NCBI

48 

Zhu J, Tang Y, Wu Q, Ji YC, Feng ZF and Kang FW: HIF-1α facilitates osteocyte-mediated osteoclastogenesis by activating JAK2/STAT3 pathway in vitro. J Cell Physiol. 234:21182–21192. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Song X, Tang Y, Zhu J, Tian Y, Song Z, Hu X, Hong C, Cai Y and 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. 67:1014022020. View Article : Google Scholar : PubMed/NCBI

50 

Tang Y, Hong C, Cai Y, Zhu J, Hu X, Tian Y, Song X, Song Z, Jiang R and Kang F: HIF-1α mediates osteoclast-induced mandibular condyle growth via AMPK signaling. J Dent Res. 99:1377–1386. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Wu C, Rankin EB, Castellini L, Alcudia JF, LaGory EL, Andersen R, Rhodes SD, Wilson TL, Mohammad KS, Castillo AB, et al: Oxygen-sensing PHDs regulate bone homeostasis through the modulation of osteoprotegerin. Genes Dev. 29:817–831. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Kang H, Yang K, Xiao L, Guo L, Guo C, Yan Y, Qi J, Wang F, Ryffel B, Li C and Deng L: Osteoblast Hypoxia-inducible Factor-1α pathway activation restrains osteoclastogenesis via the interleukin-33-MicroRNA-34a-Notch1 pathway. Front Immunol. 8:13122017. View Article : Google Scholar : PubMed/NCBI

53 

Zou D, Han W, You S, Ye D, Wang L, Wang S, Zhao J, Zhang W, Jiang X, Zhang X and Huang Y: In vitro study of enhanced osteogenesis induced by HIF-1α-transduced bone marrow stem cells. Cell Prolif. 44:234–243. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Kusumbe AP, Ramasamy SK and Adams RH: Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature. 507:323–328. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Peng Y, Wu S, Li Y and Crane JL: Type H blood vessels in bone modeling and remodeling. Theranostics. 10:426–436. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Ding W, Xu C, Zhang Y and Chen H: Advances in the understanding of the role of type-H vessels in the pathogenesis of osteoporosis. Arch Osteoporos. 15:52020. View Article : Google Scholar : PubMed/NCBI

57 

Ramasamy SK, Kusumbe AP, Wang L and Adams RH: Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature. 507:376–380. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Yang M, Li CJ, Sun X, Guo Q, Xiao Y, Su T, Tu ML, 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. 8:160032017. View Article : Google Scholar : PubMed/NCBI

59 

Shao J, Liu S, Zhang M, Chen S, Gan S, Chen C, Chen W, Li L and Zhu Z: A dual role of HIF1α in regulating osteogenesis-angiogenesis coupling. Stem Cell Res Ther. 13:592022. View Article : Google Scholar : PubMed/NCBI

60 

Tao L, Li D, Liu H, Jiang F, Xu Y, Cao Y, Gao R and Chen G: Neuroprotective effects of metformin on traumatic brain injury in rats associated with NF-κB and MAPK signaling pathway. Brain Res Bull. 140:154–161. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Yao R, Hou W and Bao J: Complete oxidative conversion of lignocellulose derived non-glucose sugars to sugar acids by Gluconobacter oxydans. Bioresour Technol. 244:1188–1192. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Guo K, Yao X, Wu W, Yu Z, Li Z, Ma Z and Liu D: HIF-1α/SDF-1/CXCR4 axis reduces neuronal apoptosis via enhancing the bone marrow-derived mesenchymal stromal cell migration in rats with traumatic brain injury. Exp Mol Pathol. 114:1044162020. View Article : Google Scholar : PubMed/NCBI

63 

Knerlich-Lukoschus F, von der Ropp-Brenner B, Lucius R, Mehdorn HM and Held-Feindt J: Spatiotemporal CCR1, CCL3(MIP-1α), CXCR4, CXCL12(SDF-1α) expression patterns in a rat spinal cord injury model of posttraumatic neuropathic pain. J Neurosurg Spine. 14:583–597. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Xue Y, Li Z, Wang Y, Zhu X, Hu R and Xu W: Role of the HIF-1α/SDF-1/CXCR4 signaling axis in accelerated fracture healing after craniocerebral injury. Mol Med Rep. 22:2767–2774. 2020.PubMed/NCBI

65 

Tacchini L, Bianchi L, Bernelli-Zazzera A and Cairo G: Transferrin receptor induction by hypoxia. HIF-1-mediated transcriptional activation and cell-specific post-transcriptional regulation. J Biol Chem. 274:24142–24146. 1999. View Article : Google Scholar : PubMed/NCBI

66 

Yang L, Fan M, Du F, Gong Q, Bi ZG, Zhu ZJ, Zhu LL and Ke Y: Hypoxic preconditioning increases iron transport rate in astrocytes. Biochim Biophys Acta. 1822:500–508. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Hu J, Meng F, Hu X, Huang L, Liu H, Liu Z and Li L: Iron overload regulate the cytokine of mesenchymal stromal cells through ROS/HIF-1α pathway in Myelodysplastic syndromes. Leuk Res. 93:1063542020. View Article : Google Scholar : PubMed/NCBI

68 

Lok CN and Ponka P: Identification of a hypoxia response element in the transferrin receptor gene. J Biol Chem. 274:24147–24152. 1999. View Article : Google Scholar : PubMed/NCBI

69 

Lee PJ, Jiang BH, Chin BY, Iyer NV, Alam J, Semenza GL and Choi AM: Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J Biol Chem. 272:5375–5381. 1997. View Article : Google Scholar : PubMed/NCBI

70 

Weinreb O, Mandel S, Youdim MB and Amit T: Targeting dysregulation of brain iron homeostasis in Parkinson's disease by iron chelators. Free Radic Biol Med. 62:52–64. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Guo C, Hao LJ, Yang ZH, Chai R, Zhang S, Gu Y, Gao HL, Zhong ML, Wang T, Li JY and Wang ZY: Deferoxamine-mediated up-regulation of HIF-1α prevents dopaminergic neuronal death via the activation of MAPK family proteins in MPTP-treated mice. Exp Neurol. 280:13–23. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Lim J, Kim HI, Bang Y, Seol W, Choi HS and Choi HJ: Hypoxia-inducible factor-1α upregulates tyrosine hydroxylase and dopamine transporter by nuclear receptor ERRγ in SH-SY5Y cells. Neuroreport. 26:380–386. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Li Y, Cao Y, Xiao J, Shang J, Tan Q, Ping F, Huang W, Wu F, Zhang H and Zhang X: Inhibitor of apoptosis-stimulating protein of p53 inhibits ferroptosis and alleviates intestinal ischemia/reperfusion-induced acute lung injury. Cell Death Differ. 27:2635–2650. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Li X, Zou Y, Xing J, Fu YY, Wang KY, Wan PZ and Zhai XY: Pretreatment with Roxadustat (FG-4592) attenuates folic acid-induced kidney injury through Antiferroptosis via Akt/GSK-3β/Nrf2 Pathway. Oxid Med Cell Longev. 2020:62869842020.PubMed/NCBI

76 

Nakazawa MS, Keith B and Simon MC: Oxygen availability and metabolic adaptations. Nat Rev Cancer. 16:663–673. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Piccoli C, D'Aprile A, Ripoli M, Scrima R, Boffoli D, Tabilio A and Capitanio N: The hypoxia-inducible factor is stabilized in circulating hematopoietic stem cells under normoxic conditions. FEBS Lett. 581:3111–3119. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Lequeux A, Noman MZ, Xiao M, Van Moer K, Hasmim M, Benoit A, Bosseler M, Viry E, Arakelian T, Berchem G, et al: Targeting HIF-1 alpha transcriptional activity drives cytotoxic immune effector cells into melanoma and improves combination immunotherapy. Oncogene. 40:4725–4735. 2021. View Article : Google Scholar : PubMed/NCBI

79 

López-Barneo J and Simon MC: Cellular adaptation to oxygen deficiency beyond the Nobel award. Nat Commun. 11:6072020. View Article : Google Scholar : PubMed/NCBI

80 

Loots GG, Robling AG, Chang JC, Murugesh DK, Bajwa J, Carlisle C, Manilay JO, Wong A, Yellowley CE and Genetos DC: Vhl deficiency in osteocytes produces high bone mass and hematopoietic defects. Bone. 116:307–314. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Lappin KM, Mills KI and Lappin TR: Erythropoietin in bone homeostasis-Implications for efficacious anemia therapy. Stem Cells Transl Med. 10:836–843. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Johnson RW, Schipani E and Giaccia AJ: HIF targets in bone remodeling and metastatic disease. Pharmacol Ther. 150:169–177. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Tao J, Miao R, Liu G, Qiu X, Yang B, Tan X, Liu L, Long J, Tang W and Jing W: Spatiotemporal correlation between HIF-1α and bone regeneration. FASEB J. 36:e225202022. View Article : Google Scholar : PubMed/NCBI

84 

Hulley PA, Bishop T, Vernet A, Schneider JE, Edwards JR, Athanasou NA and Knowles HJ: Hypoxia-inducible factor 1-alpha does not regulate osteoclastogenesis but enhances bone resorption activity via prolyl-4-hydroxylase 2. J Pathol. 242:322–333. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Clinkenbeard EL, Hanudel MR, Stayrook KR, Appaiah HN, Farrow EG, Cass TA, Summers LJ, Ip CS, Hum JM, Thomas JC, et al: Erythropoietin stimulates murine and human fibroblast growth factor-23, revealing novel roles for bone and bone marrow. Haematologica. 102:e427–e430. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Daryadel A, Bettoni C, Haider T, Imenez Silva PH, Schnitzbauer U, Pastor-Arroyo EM, Wenger RH, Gassmann M and Wagner CA: Erythropoietin stimulates fibroblast growth factor 23 (FGF23) in mice and men. Pflugers Arch. 470:1569–1582. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Li RL, He LY, Zhang Q, Liu J, Lu F, Duan HX, Fan LH, Peng W, Huang YL and Wu CJ: HIF-1α is a potential molecular target for herbal medicine to treat diseases. Drug Des Devel Ther. 14:4915–4949. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Kasper AC, Moon EJ, Hu X, Park Y, Wooten CM, Kim H, Yang W, Dewhirst MW and Hong J: Analysis of HIF-1 inhibition by manassantin A and analogues with modified tetrahydrofuran configurations. Bioorg Med Chem Lett. 19:3783–3786. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Kwak SH, Stephenson TN, Lee HE, Ge Y, Lee H, Min SM, Kim JH, Kwon DY, Lee YM and Hong J: Evaluation of Manassantin A tetrahydrofuran core region analogues and cooperative therapeutic effects with EGFR inhibition. J Med Chem. 63:6821–6833. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Hu H, Miao XK, Li JY, Zhang XW, Xu JJ, Zhang JY, Zhou TX, Hu MN, Yang WL and Mou LY: YC-1 potentiates the antitumor activity of gefitinib by inhibiting HIF-1α and promoting the endocytic trafficking and degradation of EGFR in gefitinib-resistant non-small-cell lung cancer cells. Eur J Pharmacol. 874:1729612020. View Article : Google Scholar : PubMed/NCBI

91 

Khan M, Dhammu TS, Baarine M, Kim J, Paintlia MK, Singh I and Singh AK: GSNO promotes functional recovery in experimental TBI by stabilizing HIF-1α. Behav Brain Res. 340:63–70. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Lei R, Li J, Liu F, Li W, Zhang S, Wang Y, Chu X and Xu J: HIF-1α promotes the keloid development through the activation of TGF-β/Smad and TLR4/MyD88/NF-κB pathways. Cell Cycle. 18:3239–3250. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Feng S, Bowden N, Fragiadaki M, Souilhol C, Hsiao S, Mahmoud M, Allen S, Pirri D, Ayllon BT, Akhtar S, et al: Mechanical activation of hypoxia-inducible Factor 1α drives endothelial dysfunction at atheroprone sites. Arterioscler Thromb Vasc Biol. 37:2087–2101. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Wu D, Huang RT, Hamanaka RB, Krause M, Oh MJ, Kuo CH, Nigdelioglu R, Meliton AY, Witt L, Dai G, et al: HIF-1α is required for disturbed flow-induced metabolic reprogramming in human and porcine vascular endothelium. Elife. 6:e252172017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2022
Volume 50 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang X, Zhang Y, Qi B, Sun K, Liu N, Tang B, Fang S, Zhu L and Wei X: HIF‑1α: Its notable role in the maintenance of oxygen, bone and iron homeostasis (Review). Int J Mol Med 50: 141, 2022
APA
Huang, X., Zhang, Y., Qi, B., Sun, K., Liu, N., Tang, B. ... Wei, X. (2022). HIF‑1α: Its notable role in the maintenance of oxygen, bone and iron homeostasis (Review). International Journal of Molecular Medicine, 50, 141. https://doi.org/10.3892/ijmm.2022.5197
MLA
Huang, X., Zhang, Y., Qi, B., Sun, K., Liu, N., Tang, B., Fang, S., Zhu, L., Wei, X."HIF‑1α: Its notable role in the maintenance of oxygen, bone and iron homeostasis (Review)". International Journal of Molecular Medicine 50.6 (2022): 141.
Chicago
Huang, X., Zhang, Y., Qi, B., Sun, K., Liu, N., Tang, B., Fang, S., Zhu, L., Wei, X."HIF‑1α: Its notable role in the maintenance of oxygen, bone and iron homeostasis (Review)". International Journal of Molecular Medicine 50, no. 6 (2022): 141. https://doi.org/10.3892/ijmm.2022.5197