Sclerotic zone in femoral head necrosis: from pathophysiology to therapeutic implications

in EFORT Open Reviews
Authors:
Pengqiang Lou Liaoning University of Traditional Chinese Medicine, Shenyang, China

Search for other papers by Pengqiang Lou in
Current site
Google Scholar
PubMed
Close
,
Guangzhi Zhou Liaoning University of Traditional Chinese Medicine, Shenyang, China

Search for other papers by Guangzhi Zhou in
Current site
Google Scholar
PubMed
Close
,
Bo Wei Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China

Search for other papers by Bo Wei in
Current site
Google Scholar
PubMed
Close
,
Xiaolei Deng Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China

Search for other papers by Xiaolei Deng in
Current site
Google Scholar
PubMed
Close
, and
Decai Hou Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China

Search for other papers by Decai Hou in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-1461-2134

Correspondence should be addressed to D Hou; Email: lutcmhdc@163.com

*(P Lou and G Zhou contributed equally and are co-first authors)

Open access

  • This review summarizes the sclerotic zone's pathophysiology, characterization, formation process, and impact on femoral head necrosis.

  • The sclerotic zone is a reaction interface formed during the repair of femoral head necrosis.

  • Compared with normal bone tissue, the mechanical properties of the sclerotic zone are significantly enhanced.

  • Many factors influence the formation of the sclerotic zone, including mechanics, bone metabolism, angiogenesis, and other biological processes.

  • The sclerotic zone plays an essential role in preventing the collapse of the femoral head and can predict the risk of the collapse of the femoral head.

  • Regulating the formation of the sclerotic zone of the femoral head has become a direction worthy of study in treating femoral head necrosis.

Abstract

  • This review summarizes the sclerotic zone's pathophysiology, characterization, formation process, and impact on femoral head necrosis.

  • The sclerotic zone is a reaction interface formed during the repair of femoral head necrosis.

  • Compared with normal bone tissue, the mechanical properties of the sclerotic zone are significantly enhanced.

  • Many factors influence the formation of the sclerotic zone, including mechanics, bone metabolism, angiogenesis, and other biological processes.

  • The sclerotic zone plays an essential role in preventing the collapse of the femoral head and can predict the risk of the collapse of the femoral head.

  • Regulating the formation of the sclerotic zone of the femoral head has become a direction worthy of study in treating femoral head necrosis.

Introduction

Osteonecrosis of the femoral head (ONFH) is a joint and difficult-to-treat disease, often leading to claudication and pain in the hip joint, limited mobility (1), and eventually leading to the collapse of the femoral head and hip osteoarthritis within 1–4 years of onset (2). The disease is common in young adults, and its prevalence is rising, bringing a heavy burden to families and society (3). Major risk factors for non-traumatic femoral head osteonecrosis include the application of glucocorticoids (4, 5) and alcohol consumption (6). The mechanism of femoral head necrosis may be due to the interruption of blood circulation in the femoral head due to various causes, such as damage to the vascular endothelium and the formation of microvessels leading to reduced perfusion in the femoral head, as well as intramedullary hypertension in the femoral head, which leads to arteriovenous stagnation and occlusion (7, 8, 9, 10). At the same time, osteocyte apoptosis, osteoblast reduction, osteoclast increase, and bone remodeling imbalance (11) lead to the accumulation of a large number of microfractures in the femoral head, involving the subchondral bone, which eventually manifests as the collapse of the subchondral bone (12). In addition, the concentration of local shear stresses between the femoral head's necrotic area and common bone tissue areas may cause the femoral head to collapse (13). A sclerotic zone exists in the transition area between the necrotic and common bone tissue in the femoral head (14), also known as a sclerotic rim, which is a dense bone band of higher strength than the normal cancellous bone of the femoral head produced during the repair process of femoral head necrosis (15). This article provides a review of the pathophysiology of the sclerotic zone in femoral head necrosis, the formation process, and the impact on femoral head necrosis.

Pathology of femoral head necrosis and sclerotic zone formation

The death of bone marrow hematopoietic cells occurs within 6–12 h in the pathological process of bone necrosis (16), while osteoblasts (osteocytes, osteoblasts, and osteoclasts) can survive for 12–48 h (16, 17) and adipocytes for 2–5 days (16). Then, within the tissue adjacent to the necrotic area, there is angiogenesis and bone marrow reactivity changes with increased inflammatory fibrovascular infiltration. A reactive interface of continuous repair ensues to isolate and repair the necrotic area that appears as a border on the femoral head sample (18). Over time, this reactive interface matures and progresses, and its essence is vascularized granulation tissue to repair the necrotic area, known as the creeping substitution zone (19). Because this area is richly vascularized, osteoblasts and osteoclasts are supported, allowing osteoblasts to deposit on necrotic bone trabeculae and osteoclasts to resorb the necrotic bone so that the bone weakening caused by the reactive interface at the edge of the creeping substitution zone is compensated, bone strengthening occurs, and eventually a sclerotic zone is formed (19). However, in the vast majority of cases, the repair interface cannot replace or repair areas of osteonecrosis (20). Besides, increased bone resorption at the junction of the reaction zone and the subchondral bone, as well as the synergistic effect of the weight on the femoral head, leads to subchondral bone fracture, and eventually to femoral head collapse and hip osteoarthritis (21).

Characterization of the sclerotic zone

Irregular dense bone tissue is seen between the area of femoral head necrosis and normal tissue, called sclerotic zone (13); it is the repair interface between the two during the repair of femoral head necrosis (Fig. 1).

Figure 1
Figure 1

The femoral head section (black arrow: the sclerotic zone).

Citation: EFORT Open Reviews 8, 6; 10.1530/EOR-22-0092

In the x-ray and the CT images (Fig. 2A, B and C) (19, 22, 23), the sclerotic zone appears as an irregular patchy or striated hyperdense shadow at the junction of necrotic tissue and normal tissue, while CT shows necrotic bone fractures and granulation tissue on the side of the sclerotic zone near the necrotic area (24). Furthermore, in the MRI images (19, 25), the sclerotic zone is not evident and exhibits a band-like, ring-like low signal rim on the T1 and T2 signal (Fig. 2D), suggesting a change in the structure and bone mineral salt content of the sclerotic zone reparative granulation tissue at the reaction interface. When micro-CT was applied to the necrotic femoral head (12, 26, 27), it was observed that the bone trabeculae in the sclerotic zone were disorganized and anisotropically arranged, and the trabeculae were thickened and fused. In contrast, the bone mineral density (BMD), bone volume fraction (BV/TV), trabecular thickness (Tb.Th), and trabecular number (Tb.N) values in the sclerotic zone of the femoral head increased compared with the normal femoral head, while the trabecular separation (Tb.Sp) values decreased. It indicates that in the osteosclerotic zone, as the BMD increases, the trabecular structure become dense, the trabecular hypertrophy, the number of trabeculae increase, and the trabecular separation become narrow. Also, the quantitative parameters of bone marrow perfusion on dynamic contrast-enhanced MRI and Raman spectroscopy suggest the presence of active bone repair in the bone tissue of the sclerotic zone (28).

Figure 2
Figure 2

The radiology of sclerotic zone in femoral head necrosis. (A) X-ray image (white arrow: sclerotic zone); (B–C) CT images (white arrow: sclerotic zone); (D) T1 signal in MRI (white arrow: sclerotic zone).

Citation: EFORT Open Reviews 8, 6; 10.1530/EOR-22-0092

The histology showed that the bone trabeculae in the sclerotic zone were thickened and increased in number (12), the arrangement was disorganized, and microfractures were observed (26). There were more bone lacunas in the trabeculae (29) and a large number of blood vessels in the sclerotic zone (30), woven bone, cartilage-like bone, and disordered new bone formation near the sclerotic zone (31). There was a significant increase in the number of alkaline phosphatase (ALP)-stained positive osteoblasts in the sclerotic zone, whereas there was no significant difference in the number of tartrate-resistant acid phosphatase (TRAP)-stained positive osteoclasts (12, 27). Increased apoptosis of osteoblasts was seen (31). However, the composition of the sclerotic zone did not differ significantly from the normal zone (31).

As a result of the earlier changes within the sclerotic zone of the femoral head, the mechanical parameters of the sclerotic zone also changed. Compared with the normal femoral head, the elastic modulus of the sclerotic zone increased by 58%, the yield strength by 42%, and the ultimate strength by 45%, indicating that the biomechanical strength of the sclerotic zone of the femoral head was significantly increased (32).

In addition, a study compared the sclerotic zone formed by femoral head necrosis with the osteoarthritic sclerotic zone of the hip and suggests that the thickness of bone trabeculae in the sclerotic zone formed by femoral head necrosis was higher than that in the osteoarthritic sclerotic zone of the hip. The bone marrow perfusion and bone repair in the sclerotic zone were more substantial than those in the osteoarthritic sclerotic zone of the hip (28), revealing the difference between the sclerotic zone of the femoral head necrosis and the osteoarthritic sclerotic zone of the hip.

Factors affecting the formation of the sclerotic zone

Mechanics

According to Wolff's law (33), the human or animal skeleton remodels itself to adapt to changes in load. Some researchers applied finite element analysis to study the stress changes in the femoral head during early femoral head necrosis, and found that stresses in the femoral head, such as shear stress and equivalent stresses, were redistributed (34), and the stresses focus on the repair interface between the necrotic area and normal tissue (35, 36). In order to accommodate the redistributed stress, bone tissue below the interface undergoes reconstruction: cancellous bone is realigned, bone trabeculae are thickened, and biomechanical properties are enhanced (26). Furthermore, later in order to accommodate the stress concentration at the junction of the thinner bone trabeculae in the necrotic area and the thicker bone trabeculae in the repaired area, eventually leading to the formation of a sclerotic zone (18), and later to accommodate the stress concentration at the junction of the thinner bone trabeculae in the necrotic area and the thicker bone trabeculae in the repaired area, eventually leading to the formation of a sclerotic zone (18). With the progression of the disease, the trabecular bone thickened, the degree of hardening of the sclerosing zone changed (13), and the stresses changed (22). Remodel of bone tissue is influenced by various mechanical factors (37); among the relevant stresses, compressive stress can enhance osteoblast activity and promote bone regeneration (38), and shear stress can stimulate osteoblast proliferation (39).

Bone metabolism

Cytokines and their pathways play an essential role in sclerotic zone formation by regulating cell differentiation, proliferation, apoptosis, and promoting angiogenesis. Immunohistochemistry showed (27) that bone morphogenetic protein 2 (BMP2), bone morphogenetic protein 7 (BMP7), Runt-associated transcription factor 2 (Runx2), and osteoprotegerin (OPG) were expressed in the trabecular matrix of the sclerotic zone of the femoral head. Furthermore, RT-PCR results show that (27) the expression of Runx2 and BMP2 in the sclerotic zone was higher than in other parts of the necrotic femoral head, as well as the expression of nuclear factor receptor activator kB (RANK) was higher than that in normal tissue and lower than that in necrotic tissue and subchondral bone area. The nuclear factor receptor activator ligand (RANKL) expression and the ratio of RANKL/OPG were significantly lower than in other areas (12). Besides this, transforming growth factor beta 1 (TGFβ-1) expression was increased in the femoral head necrosis area (40). While western blot showed that (27) the expression of BMP2, BMP7, Runx2, and OPG was higher than in other sites, the expression of RANK and RANKL was higher than the normal tissue and subchondral bone area but lower than the necrotic tissue. The earlier results illustrate the increased activity of osteoblasts and osteoclasts in the sclerotic zone and active bone reconstruction. The skeletal system maintains homeostasis and function by regulating osteoblasts and osteoclasts. Among them, the OPG/RANKL/RANK signaling pathway is an important pathway that regulates bone metabolism (41). Bone remodel depends on the balance of OPG and RANKL (42); when the ratio of RANKL/OPG increases, the number and activity of osteoclasts increase. TGFβ-1, BMP2, and BMP7 belong to the TGF-β superfamily and play an essential role in bone formation through the TGF-β/BMP signaling pathway (43, 44). BMP2 and BMP7 have intense osteoinductive activity (45, 46), inducing differentiation of undifferentiated mesenchymal cells into osteoblasts, thereby inducing bone formation. TGFβ-1 is an important osteogenic regulator that promotes osteoblast proliferation and apoptosis of osteoclasts (47) and bone reconstruction (48). RUNX2 is a specific osteogenic differentiation transcription factor that promotes the transcription of genes related to osteoblast and chondrocyte mineralization and their differentiation into osteoblasts. RUNX2 also plays a vital role in bone growth and regulates cell differentiation and osteoblasts, osteoclasts, and various cytokines related to bone metabolism (49, 50).

Angiogenesis

Some studies have shown that vascular damage precedes changes in bone microstructure during femoral head necrosis (32). Femoral head microcirculation disorders may be a cause of osteonecrosis (51). Many microvessels provide blood supply for the formation of the sclerotic zone. Various cytokines and pathways are involved in vascular formation, including the hypoxia-inducible factor-1 (HIF-1α) signaling pathway and its downstream target, vascular endothelial growth factor (VEGF). HIF-1α is a critical transcription factor that mediates cellular adaptation to hypoxia (52) and plays an essential role in the coupled regulation of angiogenesis–osteogenesis during bone repair and regeneration (53). Angiogenesis and osteogenesis genes are coupled at specific vascular subtypes in the skeleton (54). Thus, blood vessels are essential for bone reconstruction, and this process is dependent on the HIF-1α pathway (55). Sustained activation of the HIF-1α pathway in osteoblasts upregulates VEGF and stimulates bone reconstruction (56). It has been shown that activation of the HIF-1α signaling pathway by HIF-1α molecules and modulation of its downstream target VEGF can intervene in femoral head necrosis (57). The sclerotic zone is a repair area located between the necrotic area and normal tissue of the femoral head, which is active in angiogenesis and other repair activities, but its specific mechanisms have been poorly studied.

Apoptosis and autophagy

Apoptosis and autophagy affect the formation of the femoral sclerotic area. Apoptosis is a programmed cell death to protect the overall homeostasis of the muscle by a complex mechanism (58). During femoral head necrosis, glucocorticosteroid and other factors can directly cause apoptosis of osteoblasts (59) and osteoclasts (60) and inhibit apoptosis of osteoclasts (61), thus affecting bone transformation and bone formation, as well as leading to apoptosis of vascular endothelial cells and affecting the blood supply within the femoral head (62). It has been reported that regulation of various cytokines and pathways, including AMPK pathway (63), mTOR pathway (64), PI3K/Akt pathway50, Wnt/β-catenin pathway (65), endoplasmic reticulum stress, and mitochondrial pathway-related pathways (66, 67), miRNA (68), exosomes (69, 70), and their interactions can interfere with apoptosis and prevent the development and progression of femoral necrosis. Autophagy is a protective mechanism for cells under conditions of external stimulation or starvation, providing nutrients to cells through the degradation of intracellular proteins and damaged organelles to maintain intracellular homeostasis and internal environmental homeostasis (71) and to save cells from apoptosis. It has been shown that hormone-induced autophagy can reduce the apoptosis rate of osteoblasts and osteocytes (72, 73). Therefore, apoptosis and autophagy interact with each other to jointly condition the pathological process of femoral head necrosis and are closely related to the formation of the sclerotic zone.

The role of the sclerotic zone in ONFH

It has been reported that sclerotic bands can slow down the collapse process of the femoral head during ONFH repair (74, 75). However, this is related to the location, morphology, and integrity of the sclerotic zone formation, and the mechanism may be that the high-density bone trabeculae within the sclerotic zone maintain the mechanical strength and, under certain conditions, form a closed sclerotic rim to reduce the original high stress in the subchondral bone (75). Some studies have shown that patients with a continuous sclerotic zone would not experience femoral head collapse, while patients with a discontinuous sclerotic zone had a 63% femoral head collapse rate. In contrast, all patients without a sclerotic zone eventually experienced femoral head collapse (76). Moreover, ARCO stage II ONFH with a ring-shaped sclerotic zone usually has an ideal prognosis (77). When the ring-shaped sclerotic zone is located in the weight-bearing area, femoral head collapse progresses slowly, and hip symptoms are not obvious, which has no significant impact on the patient's daily life. When the sclerotic zone is located in the non-weight-bearing area, the patient's symptoms disappear spontaneously gradually (77). Besides this, Zhang et al. (78) found that the sclerotic band followed specific shapes that determined the mechanical conduction's stability after examining plenty of images of the femoral head with sclerotic bands and proposed a ‘sclerotic band’ type of classification system. The classification system divided the sclerotic band into ‘U’, ‘inverted U’, ‘X’, and ‘S’ types according to the sclerotic band morphology as well as the stability of mechanical conduction. Based on this, the classification system expends more subtypes to assess the femoral head's stability. By scoring the stability of the femoral head, the classification system can provide additional information for clinical determination of the risk of femoral head collapse.

However, some research suggests that the formation of the sclerotic area hinders the growth of new vessels and bone tissue into the necrotic zone, which is detrimental to bone repair (79), but there is currently insufficient evidence to support this view. Besides this, finite element analysis of different types of the sclerotic zone indicates that the concentrated shear stress and strain at the junction between the sclerotic and necrotic area will lead to an intra-femoral microfracture (13) and eventually extends to the subchondral bone, resulting in a subchondral bone fracture and the femoral head collapse (18, 36). Therefore, using CT images, Huang et al. (22) studied the dynamic evolution of bone structure and the risk of dynamic collapse in femoral head necrosis and showed that soft tissue volume in the sclerotic zone decreased while the volume and bone density of sclerotic bone increased as the disease progressed, suggesting that sclerotic bone volume and bone density could be used as indicators to assess the risk of femoral head collapse.

Many studies on ONFH are currently hoping to find a valid method to accurately predict the prognosis and risk of collapse of ONFH (80, 81, 82). Most studies concluded that the femoral head is prone to collapse when the extent of necrosis is large and the necrotic area overlaps with the weight-bearing area. In contrast, Yu et al. (83) proposed the proportion of sclerotic bands to predict the risk of femoral head collapse. The final results showed that the percentage of the sclerotic zone was significant in predicting a femoral head collapse in ONFH, and the risk of collapse was low when the percentage of sclerosis was more significant than 30% and higher when it was less than 30%, and adequate mechanical support should be provided. However, as the article mentioned, the method has some limitations: the method could not be applied to predict the prognosis of ONFH at ARCO stages 0 and I because CT images do not reveal the sclerotic rim. Besides this, measurement of the proportion of the proximal sclerotic rim would be difficult to perform if the boundary of the focus was not clearly displayed for various reasons. We considered that combining the approach and the ‘sclerotic band’ type of classification system mentioned above can improve the performance of predicting collapse based on the sclerotic band.

Discussion

The specific role and influence of the sclerotic zone in the process of femoral head necrosis have not been thoroughly studied yet, and the specific molecular mechanism of its formation needs to be further investigated. However, the formation of the sclerotic zone and the formation site and morphology play a crucial role in the collapse of the femoral head and its prognosis in ONFH pathology. Compared with normal tissue, the mechanical parameters of the sclerotic zone appear to be enhanced with the thickening of bone trabeculae. When the sclerotic zone provides sufficient mechanical support to the subchondral bone, it can prevent the femoral head from collapsing. Based on this theory, the clinical prediction model was proposed to predict the risk of femoral head collapse by applying the ratio of the sclerotic zone with high accuracy.

On the other hand, the sclerotic zone also obstructs or interrupts the repair of the necrotic area, making treating femoral head necrosis difficult. Therefore, how to regulate the formation of the femoral head sclerosis band becomes the direction of further research. The formation of sclerotized bands is influenced by a variety of cytokines and mechanical factors. By intervening in related pathways such as bone metabolism, bone differentiation, angiogenesis, apoptosis, and autophagy, we can intervene in cell differentiation and bone trabecular formation at different stages of necrotic repair, so as to regulate the form and location of sclerotic zone formation. In need of mechanical support, part of the subchondral bone is formed through the strength of sclerotic zone in order to prevent the collapse of the femoral head. To prevent the crawling replacement of of a thick sclerotic zone that would hinder the repair of femoral head necrosis area, or for patients with high-risk factors such as application of glucocorticoid, early control of the formation of the sclerotic zone is required. In addition, the necrotic area is limited in order to treat the femoral head necrosis. The possibility of drug therapy for femoral head necrosis should also be discussed.

Conclusion

The sclerotic zone is a reaction interface formed during the repair of femoral head necrosis. Compared with normal bone tissue, the mechanical properties of the sclerotic zone are significantly enhanced. Many factors influence the formation of the sclerotic zone, including mechanics, bone metabolism, angiogenesis, and other biological processes. The sclerotic zone plays an essential role in preventing the collapse of the femoral head and can predict the risk of the collapse of the femoral head. Regulating the formation of the sclerotic zone of the femoral head has become a direction worthy of study in treating femoral head necrosis.

ICMJE conflict of interest statement

The authors declare that they have no competing interests

Funding

This study is financially supported by Shenyang Science and Technology Bureau Technology Talent Application Technology Research Project (grant no. 21-174-9-03); Department of Science and Technology of Liaoning Province (grant No.2017020223-201).

Ethical review statement

This research did not require approval by the local ethics board.

Consent to participate

The participants approved the x-ray, CT, MRI, and the femoral head section images used in this study and signed an informed consent form.

Author contribution statement

P-qL, G-zZ, and D-cH conceived and designed the study. P-qL, G-zZ, BW, and X-LD performed literature search. P-qL, G-Zz, BW, and X-LD organized related papers. P-qL and G-zZ wrote the manuscript. All authors contributed to the article and approved the submitted version.

References

  • 1.

    Zalavras CG, & Lieberman Jr. Osteonecrosis of the femoral head: evaluation and treatment. Journal of the American Academy of Orthopaedic Surgeons 2014 22 455464. (https://doi.org/10.5435/JAAOS-22-07-455)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    Yoon BH, Jones LC, Chen CH, Cheng EY, Cui Q, Drescher W, Fukushima W, Gangji V, Goodman SB, Ha YC, et al.Etiologic classification criteria of ARCO on femoral head osteonecrosis Part 1: Glucocorticoid-associated osteonecrosis. Journal of Arthroplasty 2019 34 163168.e1. (https://doi.org/10.1016/j.arth.2018.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Cui L, Zhuang Q, Lin J, Jin J, Zhang K, Cao L, Lin J, Yan S, Guo W, He W, et al.Multicentric epidemiologic study on six thousand three hundred and ninety five cases of femoral head osteonecrosis in China. International Orthopaedics 2016 40 267276. (https://doi.org/10.1007/s00264-015-3061-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Mont MA, Pivec R, Banerjee S, Issa K, Elmallah RK, & Jones LC. High-dose corticosteroid use and risk of hip osteonecrosis: meta-analysis and systematic literature review. Journal of Arthroplasty 2015 30 15061512.e5. (https://doi.org/10.1016/j.arth.2015.03.036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5.

    Fukushima W, Fujioka M, Kubo T, Tamakoshi A, Nagai M, & Hirota Y. Nationwide epidemiologic survey of idiopathic osteonecrosis of the femoral head. Clinical Orthopaedics and Related Research 2010 468 27152724. (https://doi.org/10.1007/s11999-010-1292-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Gold EW, & Cangemi PJ. Incidence and pathogenesis of alcohol-induced osteonecrosis of the femoral head. Clinical Orthopaedics and Related Research 1979 143 222226. (https://doi.org/10.1097/00003086-197909000-00037)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Anderson PA, Jeray KJ, Lane JM, & Binkley NC. Bone health optimization: beyond own the bone: AOA critical issues. Journal of Bone and Joint Surgery. American Volume 2019 101 14131419. (https://doi.org/10.2106/JBJS.18.01229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Gaddini GW, Grant KA, Woodall A, Stull C, Maddalozzo GF, Zhang B, Turner RT, & Iwaniec UT. Twelve months of voluntary heavy alcohol consumption in male rhesus macaques suppresses intracortical bone remodeling. Bone 2015 71 227236. (https://doi.org/10.1016/j.bone.2014.10.025)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Kahler-Quesada AM, Grant KA, Walter NAR, Newman N, Allen MR, Burr DB, Branscum AJ, Maddalozzo GF, Turner RT, & Iwaniec UT. Voluntary chronic heavy alcohol consumption in male rhesus macaques suppresses cancellous bone formation and increases bone marrow adiposity. Alcoholism, Clinical and Experimental Research 2019 43 24942503. (https://doi.org/10.1111/acer.14202)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Mont MA, Salem HS, Piuzzi NS, Goodman SB, & Jones LC. Nontraumatic osteonecrosis of the femoral head: where do we stand today? A 5-year update. Journal of Bone and Joint Surgery. American Volume 2020 102 10841099. (https://doi.org/10.2106/JBJS.19.01271)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Houdek MT, Wyles CC, Packard BD, Terzic A, Behfar A, & Sierra RJ. Decreased osteogenic activity of mesenchymal stem cells in patients with corticosteroid-induced osteonecrosis of the femoral head. Journal of Arthroplasty 2016 31 893898. (https://doi.org/10.1016/j.arth.2015.08.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Wang C, Meng H, Wang Y, Zhao B, Zhao C, Sun W, Zhu Y, Han B, Yuan X, Liu R, et al.Analysis of early stage osteonecrosis of the human femoral head and the mechanism of femoral head collapse. International Journal of Biological Sciences 2018 14 156164. (https://doi.org/10.7150/ijbs.18334)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Karasuyama K, Yamamoto T, Motomura G, Sonoda K, Kubo Y, & Iwamoto Y. The role of sclerotic changes in the starting mechanisms of collapse: a histomorphometric and FEM study on the femoral head of osteonecrosis. Bone 2015 81 644648. (https://doi.org/10.1016/j.bone.2015.09.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Malizos KN, Karantanas AH, Varitimidis SE, Dailiana ZH, Bargiotas K, & Maris T. Osteonecrosis of the femoral head: etiology, imaging and treatment. European Journal of Radiology 2007 63 1628. (https://doi.org/10.1016/j.ejrad.2007.03.019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Wen PF, Guo WS, Zhang QD, Gao FQ, Yue JA, Liu ZH, Cheng LM, & Li ZR. Significance of lateral pillar in osteonecrosis of femoral head: a finite element analysis. Chinese Medical Journal 2017 130 25692574. (https://doi.org/10.4103/0366-6999.217077)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Imhof H, Breitenseher M, Trattnig S, Kramer J, Hofmann S, Plenk H, Schneider W, & Engel A. Imaging of avascular necrosis of bone. European Radiology 1997 7 180186. (https://doi.org/10.1007/s003300050131)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Utting JC, Robins SP, Brandao-Burch A, Orriss IR, Behar J, & Arnett TR. Hypoxia inhibits the growth, differentiation and bone-forming capacity of rat osteoblasts. Experimental Cell Research 2006 312 16931702. (https://doi.org/10.1016/j.yexcr.2006.02.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Bullough PG, & DiCarlo EF. Subchondral avascular necrosis: a common cause of arthritis. Annals of the Rheumatic Diseases 1990 49 412420. (https://doi.org/10.1136/ard.49.6.412)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Murphey MD, Foreman KL, Klassen-Fischer MK, Fox MG, Chung EM, & Kransdorf MJ. From the radiologic pathology archives imaging of osteonecrosis: radiologic-pathologic correlation. RadioGraphics 2014 34 10031028. (https://doi.org/10.1148/rg.344140019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Plenk H Jr, Gstettner M, Grossschmidt K, Breitenseher M, Urban M, & Hofmann S. Magnetic resonance imaging and histology of repair in femoral head osteonecrosis. Clinical Orthopaedics and Related Research 2001 386 4253. (https://doi.org/10.1097/00003086-200105000-00006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Chang C, Greenspan A, & Gershwin ME. The pathogenesis, diagnosis and clinical manifestations of steroid-induced osteonecrosis. Journal of Autoimmunity 2020 110 102460. (https://doi.org/10.1016/j.jaut.2020.102460)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Huang Z, Tan B, Ye H, Fu F, Wang R, & Chen W. Dynamic evolution of osseous structure in osteonecrosis of the femoral head and dynamic collapse risks: a preliminary CT image study. Journal of Orthopaedic Surgery and Research 2020 15 539. (https://doi.org/10.1186/s13018-020-02069-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Wang C, Wang J, Zhang Y, Yuan C, Liu D, Pei Y, Li X, Wu Z, Li Y, & Guo Z. A canine model of femoral head osteonecrosis induced by an ethanol injection navigated by a novel template. International Journal of Medical Sciences 2013 10 14511458. (https://doi.org/10.7150/ijms.6314)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Hamada H, Takao M, Sakai T, & Sugano N. Subchondral fracture begins from the bone resorption area in osteonecrosis of the femoral head: a micro-computerised tomography study. International Orthopaedics 2018 42 14791484. (https://doi.org/10.1007/s00264-018-3879-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Pascart T, Paccou J, Colard T, Norberciak L, Girard J, Delattre J, Marchandise P, Legrand J, Penel G, Coursier R, et al.T1-weighted MRI images accurately represent the volume and surface of architectural mineral damage of osteonecrosis of the femoral head: comparison with high-resolution computed tomography. Bone 2020 130 115099. (https://doi.org/10.1016/j.bone.2019.115099)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Ma JX, He WW, Zhao J, Kuang MJ, Bai HH, Sun L, Lu B, Tian AX, Wang Y, Dong BC, et al.Bone microarchitecture and biomechanics of the necrotic femoral head. Scientific Reports 2017 7 13345. (https://doi.org/10.1038/s41598-017-13643-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Wang C, Wang X, Xu XL, Yuan XL, Gou WL, Wang AY, Guo QY, Peng J, & Lu SB. Bone microstructure and regional distribution of osteoblast and osteoclast activity in the osteonecrotic femoral head. PLoS One 2014 9 e96361. (https://doi.org/10.1371/journal.pone.0096361)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Pascart T, Falgayrac G, Cortet B, Paccou J, Bleuse M, Coursier R, Putman S, Quinchon JF, Bertheaume N, Delattre J, et al.Subchondral involvement in osteonecrosis of the femoral head: insight on local composition, microstructure and vascularization. Osteoarthritis and Cartilage 2022 30 11031115. (https://doi.org/10.1016/j.joca.2022.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Shi SH, Li ZR, Wang BL, Sun W, Cheng LM, Pan L, & Wang RD. Study on the relationship between sclerosis rim and bone morphogenetic proteins of osteonecrosis of the femoral head. Zhonghua Wai Ke Za Zhi [Chinese Journal of Surgery] 2010 48 13051308. (https://doi.org/10.3760/cma.j.issn.0529-5815.2010.17.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Vicaş RM, Bodog FD, Fugaru FO, Grosu F, Badea O, Lazăr L, Cevei ML, Nistor-Cseppento CD, Beiuşanu GC, Holt G, et al.Histopathological and immunohistochemical aspects of bone tissue in aseptic necrosis of the femoral head. Romanian Journal of Morphology and Embryology 2020 61 12491258. (https://doi.org/10.47162/RJME.61.4.26)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Pascart T, Falgayrac G, Migaud H, Quinchon JF, Norberciak L, Budzik JF, Paccou J, Cotten A, Penel G, & Cortet B. Region specific Raman spectroscopy analysis of the femoral head reveals that trabecular bone is unlikely to contribute to non-traumatic osteonecrosis. Scientific Reports 2017 7 97. (https://doi.org/10.1038/s41598-017-00162-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Weinstein RS, Hogan EA, Borrelli MJ, Liachenko S, O'Brien CA, & Manolagas SC. The pathophysiological sequence of glucocorticoid-induced osteonecrosis of the femoral head in male mice. Endocrinology 2017 158 38173831. (https://doi.org/10.1210/en.2017-00662)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Wolff J The Law of Bone Remodelling: Springer Science & Business Media 2012. (https://doi.org/10.1007/978-3-642-71031-5)

  • 34.

    Brown TD, & Hild GL. Pre-collapse stress redistributions in femoral head osteonecrosis--a three-dimensional finite element analysis. Journal of Biomechanical Engineering 1983 105 171176. (https://doi.org/10.1115/1.3138402)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Ueo T, Tsutsumi S, Yamamuro T, Okumura H, Shimizu A, & Nakamura T. Biomechanical aspects of the development of aseptic necrosis of the femoral head. Archives of Orthopaedic and Traumatic Surgery. Archiv Fur Orthopadische und Unfall-Chirurgie 1985 104 145149. (https://doi.org/10.1007/BF00454690)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Utsunomiya T, Motomura G, Ikemura S, Kubo Y, Sonoda K, Hatanaka H, Baba S, Kawano K, Yamamoto T, & Nakashima Y. Effects of sclerotic changes on stress concentration in early-stage osteonecrosis: a patient-specific, 3D finite element analysis. Journal of Orthopaedic Research 2018 36 31693177. (https://doi.org/10.1002/jor.24124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Lee YK, Yoo JJ, Koo KH, Yoon KS, Min BW, & Kim HJ. Collapsed subchondral fatigue fracture of the femoral head. Orthopedic Clinics of North America 2009 40 259265. (https://doi.org/10.1016/j.ocl.2008.10.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Zhang YG, Yang Z, Zhang H, Liu M, Qiu Y, & Guo X. Negative pressure technology enhances bone regeneration in rabbit skull defects. BMC Musculoskeletal Disorders 2013 14 76. (https://doi.org/10.1186/1471-2474-14-76)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Li P, Ma YC, Sheng XY, Dong HT, Han H, Wang J, & Xia YY. Cyclic fluid shear stress promotes osteoblastic cells proliferation through ERK5 signaling pathway. Molecular and Cellular Biochemistry 2012 364 321327. (https://doi.org/10.1007/s11010-012-1233-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Tingart M, Beckmann J, Opolka A, Matsuura M, Wiech O, Grifka J, & Grässel S. Influence of factors regulating bone formation and remodeling on bone quality in osteonecrosis of the femoral head. Calcified Tissue International 2008 82 300308. (https://doi.org/10.1007/s00223-008-9111-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Sung B, Prasad S, Yadav VR, Gupta SC, Reuter S, Yamamoto N, Murakami A, & Aggarwal BB. RANKL signaling and osteoclastogenesis is negatively regulated by cardamonin. PLoS One 2013 8 e64118. (https://doi.org/10.1371/journal.pone.0064118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Mochizuki A, Takami M, Miyamoto Y, Nakamaki T, Tomoyasu S, Kadono Y, Tanaka S, Inoue T, & Kamijo R. Cell adhesion signaling regulates RANK expression in osteoclast precursors. PLoS One 2012 7 e48795. (https://doi.org/10.1371/journal.pone.0048795)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Chen G, Deng C, & Li YP. TGF-β and BMP signaling in osteoblast differentiation and bone formation. International Journal of Biological Sciences 2012 8 272–288. (https://doi.org/10.7150/ijbs.2929)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Wu M, Chen G, & Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Research 2016 4 16009. (https://doi.org/10.1038/boneres.2016.9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Medici M, van Meurs JB, Rivadeneira F, Zhao H, Arp PP, Hofman A, Pols HA, & Uitterlinden AG. BMP-2 gene polymorphisms and osteoporosis: the Rotterdam Study. Journal of Bone and Mineral Research 2006 21 845854. (https://doi.org/10.1359/jbmr.060306)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Phimphilai M, Zhao Z, Boules H, Roca H, & Franceschi RT. BMP signaling is required for RUNX2-dependent induction of the osteoblast phenotype. Journal of Bone and Mineral Research 2006 21 637646. (https://doi.org/10.1359/jbmr.060109)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Shen J, Li S, & Chen D. TGF-β signaling and the development of osteoarthritis. Bone Research 2014 2 14002–. (https://doi.org/10.1038/boneres.2014.2)

  • 48.

    Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin FB, Frassica FJ, Chang W, Yao J, et al.Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nature Medicine 2013 19 704712. (https://doi.org/10.1038/nm.3143)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Takarada T, Hinoi E, Nakazato R, Ochi H, Xu C, Tsuchikane A, Takeda S, Karsenty G, Abe T, Kiyonari H, et al.An analysis of skeletal development in osteoblast-specific and chondrocyte-specific runt-related transcription factor-2 (Runx2) knockout mice. Journal of Bone and Mineral Research 2013 28 20642069. (https://doi.org/10.1002/jbmr.1945)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Chen J, Yuan K, Mao X, Miano JM, Wu H, & Chen Y. Serum response factor regulates bone formation via IGF-1 and Runx2 signals. Journal of Bone and Mineral Research 2012 27 16591668. (https://doi.org/10.1002/jbmr.1607)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51.

    Shigemura T, Nakamura J, Kishida S, Harada Y, Ohtori S, Kamikawa K, Ochiai N, & Takahashi K. Incidence of osteonecrosis associated with corticosteroid therapy among different underlying diseases: prospective MRI study. Rheumatology (Oxford) 2011 50 20232028. (https://doi.org/10.1093/rheumatology/ker277)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Choudhry H, & Harris AL. Advances in hypoxia-inducible factor biology. Cell Metabolism 2018 27 281298. (https://doi.org/10.1016/j.cmet.2017.10.005)

  • 53.

    Schipani E, Maes C, Carmeliet G, & Semenza GL. Regulation of osteogenesis-angiogenesis coupling by HIFs and VEGF. Journal of Bone and Mineral Research 2009 24 13471353. (https://doi.org/10.1359/jbmr.090602)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Kusumbe AP, Ramasamy SK, & Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014 507 323328. (https://doi.org/10.1038/nature13145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55.

    Wan C, Gilbert SR, Wang Y, Cao X, Shen X, Ramaswamy G, Jacobsen KA, Alaql ZS, Eberhardt AW, Gerstenfeld LC, et al.Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proceedings of the National Academy of Sciences of the United States of America 2008 105 686691. (https://doi.org/10.1073/pnas.0708474105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Wang Y, Wan C, Deng L, Liu X, Cao X, Gilbert SR, Bouxsein ML, Faugere MC, Guldberg RE, Gerstenfeld LC, et al.The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development. Journal of Clinical Investigation 2007 117 16161626. (https://doi.org/10.1172/JCI31581)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Jing X, Du T, Yang X, Zhang W, Wang G, Liu X, Li T, & Jiang Z. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1α expression. Journal of Cellular Physiology 2020 235 98649875. (https://doi.org/10.1002/jcp.29799)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Plotkin LI, Manolagas SC, & Bellido T. Glucocorticoids induce osteocyte apoptosis by blocking focal adhesion kinase-mediated survival. Evidence for inside-out signaling leading to anoikis. Journal of Biological Chemistry 2007 282 2412024130. (https://doi.org/10.1074/jbc.M611435200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59.

    Feng Z, Zheng W, Tang Q, Cheng L, Li H, Ni W, & Pan X. Fludarabine inhibits STAT1-mediated up-regulation of caspase-3 expression in dexamethasone-induced osteoblasts apoptosis and slows the progression of steroid-induced avascular necrosis of the femoral head in rats. Apoptosis 2017 22 10011012. (https://doi.org/10.1007/s10495-017-1383-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60.

    Cheng X, & Ferrell JE Jr. Apoptosis propagates through the cytoplasm as trigger waves. Science 2018 361 607612. (https://doi.org/10.1126/science.aah4065)

  • 61.

    Chotiyarnwong P, & McCloskey EV. Pathogenesis of glucocorticoid-induced osteoporosis and options for treatment. Nature Reviews. Endocrinology 2020 16 437447. (https://doi.org/10.1038/s41574-020-0341-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62.

    Weinstein RS. Clinical practice. Glucocorticoid-induced bone disease. New England Journal of Medicine 2011 365 6270. (https://doi.org/10.1056/NEJMcp1012926)

  • 63.

    She C, Zhu LQ, Zhen YF, Wang XD, & Dong QR. Activation of AMPK protects against hydrogen peroxide-induced osteoblast apoptosis through autophagy induction and NADPH maintenance: new implications for osteonecrosis treatment? Cellular Signalling 2014 26 18. (https://doi.org/10.1016/j.cellsig.2013.08.046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64.

    Wang XY, Gong LJ, Huang JM, Jiang C, & Yan ZQ. Pinocembrin alleviates glucocorticoid-induced apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway in osteocytes. European Journal of Pharmacology 2020 880 173212. (https://doi.org/10.1016/j.ejphar.2020.173212)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65.

    Pei J, Fan L, Nan K, Li J, Shi Z, Dang X, & Wang K. Excessive activation of TLR4/NF-κB interactively suppresses the canonical Wnt/β-catenin pathway and induces SANFH in SD rats. Scientific Reports 2017 7 11928. (https://doi.org/10.1038/s41598-017-12196-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66.

    Gao Y, Zhu H, Wang Q, Feng Y, & Zhang C. Inhibition of PERK signaling prevents against glucocorticoid-induced endotheliocyte apoptosis and osteonecrosis of the femoral head. International Journal of Biological Sciences 2020 16 543552. (https://doi.org/10.7150/ijbs.35256)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67.

    Yao X, Yu S, Jing X, Guo J, Sun K, Guo F, & Ye Y. PTEN inhibitor VO-OHpic attenuates GC-associated endothelial progenitor cell dysfunction and osteonecrosis of the femoral head via activating Nrf2 signaling and inhibiting mitochondrial apoptosis pathway. Stem Cell Research and Therapy 2020 11 140. (https://doi.org/10.1186/s13287-020-01658-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 68.

    Tian L, Sun S, Li W, Yuan L, & Wang X. Down-regulated microRNA-141 facilitates osteoblast activity and inhibits osteoclast activity to ameliorate osteonecrosis of the femoral head via up-regulating TGF-β2. Cell Cycle 2020 19 772786. (https://doi.org/10.1080/15384101.2020.1731053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69.

    Tao SC, Yuan T, Rui BY, Zhu ZZ, Guo SC, & Zhang CQ. Exosomes derived from human platelet-rich plasma prevent apoptosis induced by glucocorticoid-associated endoplasmic reticulum stress in rat osteonecrosis of the femoral head via the Akt/Bad/Bcl-2 signal pathway. Theranostics 2017 7 733750. (https://doi.org/10.7150/thno.17450)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 70.

    Kuang MJ, Huang Y, Zhao XG, Zhang R, Ma JX, Wang DC, & Ma XL. Exosomes derived from Wharton's jelly of human umbilical cord mesenchymal stem cells reduce osteocyte apoptosis in glucocorticoid-induced osteonecrosis of the femoral head in rats via the miR-21-PTEN-AKT signalling pathway. International Journal of Biological Sciences 2019 15 18611871. (https://doi.org/10.7150/ijbs.32262)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71.

    Levine B, & Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell 2019 176 1142. (https://doi.org/10.1016/j.cell.2018.09.048)

  • 72.

    Han Y, Zhang L, Xing Y, Zhang L, Chen X, Tang P, & Chen Z. Autophagy relieves the function inhibition and apoptosispromoting effects on osteoblast induced by glucocorticoid. International Journal of Molecular Medicine 2018 41 800808. (https://doi.org/10.3892/ijmm.2017.3270)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73.

    Zhu L, Chen J, Zhang J, Guo C, Fan W, Wang YM, & Yan Z. Parathyroid hormone (PTH) induces autophagy to protect osteocyte cell survival from dexamethasone damage. Medical Science Monitor 2017 23 40344040. (https://doi.org/10.12659/msm.903432)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 74.

    Yu T, Xie L, & Chu F. A sclerotic rim provides mechanical support for the femoral head in osteonecrosis. Orthopedics 2015 38 e374e379. (https://doi.org/10.3928/01477447-20150504-53)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75.

    Ito H, Matsuno T, & Kaneda K. Prognosis of early stage avascular necrosis of the femoral head. Clinical Orthopaedics and Related Research 1999 358 149157. (https://doi.org/10.1097/00003086-199901000-00018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 76.

    Ficat RP. Idiopathic bone necrosis of the femoral head. Early diagnosis and treatment. Journal of Bone and Joint Surgery. British Volume 1985 67 39. (https://doi.org/10.1302/0301-620X.67B1.3155745)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77.

    Wu ZS, Hong G, Yang P, Yang F, Chen ZQ, He W, & Wei QS. The survival of non-traumatic osteonecrosis of femoral head at ARCO II with ring-shaped sclerotic zone: a mid-term follow-up retrospective study. Journal of Hip Preservation Surgery 2020 7 705712. (https://doi.org/10.1093/jhps/hnab013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78.

    Zhang Y, Liu YW, Sun RB, Fan YN, Shi QY, Zhang LL, Li PF, & Li WY. "Sclerotic Band" type of classification system and measurement of necrotic area for osteonecrosis of the femoral head. Medicine (Baltimore) 2020 99 e22106. (https://doi.org/10.1097/MD.0000000000022106)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 79.

    Castro FP Jr, & Barrack RL. Core decompression and conservative treatment for avascular necrosis of the femoral head: a meta-analysis. American Journal of Orthopedics (Belle Mead, NJ) 2000 29 187194.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 80.

    Chen L, Hong G, Fang B, Zhou G, Han X, Guan T, & He W. Predicting the collapse of the femoral head due to osteonecrosis: from basic methods to application prospects. Journal of Orthopaedic Translation 2017 11 6272. (https://doi.org/10.1016/j.jot.2016.11.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 81.

    Li TX, Huang ZQ, Li Y, Xue ZP, Sun JG, Gao HH, He HJ, & Chen WH. Prediction of collapse using patient-specific finite element analysis of osteonecrosis of the femoral head. Orthopaedic Surgery 2019 11 794800. (https://doi.org/10.1111/os.12520)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 82.

    Cheng W, Xian H, Wang L, Luo D, Huang Z, Lian K, & Lin D. Frog leg lateral view is a reliable predictor of the prognosis in osteonecrosis of the femoral head. Journal of Orthopaedic Research 2021 39 950958. (https://doi.org/10.1002/jor.24825)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 83.

    Yu T, Xie L, Zhang Z, Ke X, & Liu Y. Prediction of osteonecrosis collapse of the femoral head based on the proportion of the proximal sclerotic rim. International Orthopaedics 2015 39 10451050. (https://doi.org/10.1007/s00264-014-2602-9)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    The femoral head section (black arrow: the sclerotic zone).

  • Figure 2

    The radiology of sclerotic zone in femoral head necrosis. (A) X-ray image (white arrow: sclerotic zone); (B–C) CT images (white arrow: sclerotic zone); (D) T1 signal in MRI (white arrow: sclerotic zone).

  • 1.

    Zalavras CG, & Lieberman Jr. Osteonecrosis of the femoral head: evaluation and treatment. Journal of the American Academy of Orthopaedic Surgeons 2014 22 455464. (https://doi.org/10.5435/JAAOS-22-07-455)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    Yoon BH, Jones LC, Chen CH, Cheng EY, Cui Q, Drescher W, Fukushima W, Gangji V, Goodman SB, Ha YC, et al.Etiologic classification criteria of ARCO on femoral head osteonecrosis Part 1: Glucocorticoid-associated osteonecrosis. Journal of Arthroplasty 2019 34 163168.e1. (https://doi.org/10.1016/j.arth.2018.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Cui L, Zhuang Q, Lin J, Jin J, Zhang K, Cao L, Lin J, Yan S, Guo W, He W, et al.Multicentric epidemiologic study on six thousand three hundred and ninety five cases of femoral head osteonecrosis in China. International Orthopaedics 2016 40 267276. (https://doi.org/10.1007/s00264-015-3061-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Mont MA, Pivec R, Banerjee S, Issa K, Elmallah RK, & Jones LC. High-dose corticosteroid use and risk of hip osteonecrosis: meta-analysis and systematic literature review. Journal of Arthroplasty 2015 30 15061512.e5. (https://doi.org/10.1016/j.arth.2015.03.036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5.

    Fukushima W, Fujioka M, Kubo T, Tamakoshi A, Nagai M, & Hirota Y. Nationwide epidemiologic survey of idiopathic osteonecrosis of the femoral head. Clinical Orthopaedics and Related Research 2010 468 27152724. (https://doi.org/10.1007/s11999-010-1292-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Gold EW, & Cangemi PJ. Incidence and pathogenesis of alcohol-induced osteonecrosis of the femoral head. Clinical Orthopaedics and Related Research 1979 143 222226. (https://doi.org/10.1097/00003086-197909000-00037)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Anderson PA, Jeray KJ, Lane JM, & Binkley NC. Bone health optimization: beyond own the bone: AOA critical issues. Journal of Bone and Joint Surgery. American Volume 2019 101 14131419. (https://doi.org/10.2106/JBJS.18.01229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Gaddini GW, Grant KA, Woodall A, Stull C, Maddalozzo GF, Zhang B, Turner RT, & Iwaniec UT. Twelve months of voluntary heavy alcohol consumption in male rhesus macaques suppresses intracortical bone remodeling. Bone 2015 71 227236. (https://doi.org/10.1016/j.bone.2014.10.025)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Kahler-Quesada AM, Grant KA, Walter NAR, Newman N, Allen MR, Burr DB, Branscum AJ, Maddalozzo GF, Turner RT, & Iwaniec UT. Voluntary chronic heavy alcohol consumption in male rhesus macaques suppresses cancellous bone formation and increases bone marrow adiposity. Alcoholism, Clinical and Experimental Research 2019 43 24942503. (https://doi.org/10.1111/acer.14202)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Mont MA, Salem HS, Piuzzi NS, Goodman SB, & Jones LC. Nontraumatic osteonecrosis of the femoral head: where do we stand today? A 5-year update. Journal of Bone and Joint Surgery. American Volume 2020 102 10841099. (https://doi.org/10.2106/JBJS.19.01271)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Houdek MT, Wyles CC, Packard BD, Terzic A, Behfar A, & Sierra RJ. Decreased osteogenic activity of mesenchymal stem cells in patients with corticosteroid-induced osteonecrosis of the femoral head. Journal of Arthroplasty 2016 31 893898. (https://doi.org/10.1016/j.arth.2015.08.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Wang C, Meng H, Wang Y, Zhao B, Zhao C, Sun W, Zhu Y, Han B, Yuan X, Liu R, et al.Analysis of early stage osteonecrosis of the human femoral head and the mechanism of femoral head collapse. International Journal of Biological Sciences 2018 14 156164. (https://doi.org/10.7150/ijbs.18334)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Karasuyama K, Yamamoto T, Motomura G, Sonoda K, Kubo Y, & Iwamoto Y. The role of sclerotic changes in the starting mechanisms of collapse: a histomorphometric and FEM study on the femoral head of osteonecrosis. Bone 2015 81 644648. (https://doi.org/10.1016/j.bone.2015.09.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Malizos KN, Karantanas AH, Varitimidis SE, Dailiana ZH, Bargiotas K, & Maris T. Osteonecrosis of the femoral head: etiology, imaging and treatment. European Journal of Radiology 2007 63 1628. (https://doi.org/10.1016/j.ejrad.2007.03.019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Wen PF, Guo WS, Zhang QD, Gao FQ, Yue JA, Liu ZH, Cheng LM, & Li ZR. Significance of lateral pillar in osteonecrosis of femoral head: a finite element analysis. Chinese Medical Journal 2017 130 25692574. (https://doi.org/10.4103/0366-6999.217077)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Imhof H, Breitenseher M, Trattnig S, Kramer J, Hofmann S, Plenk H, Schneider W, & Engel A. Imaging of avascular necrosis of bone. European Radiology 1997 7 180186. (https://doi.org/10.1007/s003300050131)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Utting JC, Robins SP, Brandao-Burch A, Orriss IR, Behar J, & Arnett TR. Hypoxia inhibits the growth, differentiation and bone-forming capacity of rat osteoblasts. Experimental Cell Research 2006 312 16931702. (https://doi.org/10.1016/j.yexcr.2006.02.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Bullough PG, & DiCarlo EF. Subchondral avascular necrosis: a common cause of arthritis. Annals of the Rheumatic Diseases 1990 49 412420. (https://doi.org/10.1136/ard.49.6.412)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Murphey MD, Foreman KL, Klassen-Fischer MK, Fox MG, Chung EM, & Kransdorf MJ. From the radiologic pathology archives imaging of osteonecrosis: radiologic-pathologic correlation. RadioGraphics 2014 34 10031028. (https://doi.org/10.1148/rg.344140019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Plenk H Jr, Gstettner M, Grossschmidt K, Breitenseher M, Urban M, & Hofmann S. Magnetic resonance imaging and histology of repair in femoral head osteonecrosis. Clinical Orthopaedics and Related Research 2001 386 4253. (https://doi.org/10.1097/00003086-200105000-00006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Chang C, Greenspan A, & Gershwin ME. The pathogenesis, diagnosis and clinical manifestations of steroid-induced osteonecrosis. Journal of Autoimmunity 2020 110 102460. (https://doi.org/10.1016/j.jaut.2020.102460)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Huang Z, Tan B, Ye H, Fu F, Wang R, & Chen W. Dynamic evolution of osseous structure in osteonecrosis of the femoral head and dynamic collapse risks: a preliminary CT image study. Journal of Orthopaedic Surgery and Research 2020 15 539. (https://doi.org/10.1186/s13018-020-02069-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Wang C, Wang J, Zhang Y, Yuan C, Liu D, Pei Y, Li X, Wu Z, Li Y, & Guo Z. A canine model of femoral head osteonecrosis induced by an ethanol injection navigated by a novel template. International Journal of Medical Sciences 2013 10 14511458. (https://doi.org/10.7150/ijms.6314)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Hamada H, Takao M, Sakai T, & Sugano N. Subchondral fracture begins from the bone resorption area in osteonecrosis of the femoral head: a micro-computerised tomography study. International Orthopaedics 2018 42 14791484. (https://doi.org/10.1007/s00264-018-3879-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Pascart T, Paccou J, Colard T, Norberciak L, Girard J, Delattre J, Marchandise P, Legrand J, Penel G, Coursier R, et al.T1-weighted MRI images accurately represent the volume and surface of architectural mineral damage of osteonecrosis of the femoral head: comparison with high-resolution computed tomography. Bone 2020 130 115099. (https://doi.org/10.1016/j.bone.2019.115099)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Ma JX, He WW, Zhao J, Kuang MJ, Bai HH, Sun L, Lu B, Tian AX, Wang Y, Dong BC, et al.Bone microarchitecture and biomechanics of the necrotic femoral head. Scientific Reports 2017 7 13345. (https://doi.org/10.1038/s41598-017-13643-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Wang C, Wang X, Xu XL, Yuan XL, Gou WL, Wang AY, Guo QY, Peng J, & Lu SB. Bone microstructure and regional distribution of osteoblast and osteoclast activity in the osteonecrotic femoral head. PLoS One 2014 9 e96361. (https://doi.org/10.1371/journal.pone.0096361)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Pascart T, Falgayrac G, Cortet B, Paccou J, Bleuse M, Coursier R, Putman S, Quinchon JF, Bertheaume N, Delattre J, et al.Subchondral involvement in osteonecrosis of the femoral head: insight on local composition, microstructure and vascularization. Osteoarthritis and Cartilage 2022 30 11031115. (https://doi.org/10.1016/j.joca.2022.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Shi SH, Li ZR, Wang BL, Sun W, Cheng LM, Pan L, & Wang RD. Study on the relationship between sclerosis rim and bone morphogenetic proteins of osteonecrosis of the femoral head. Zhonghua Wai Ke Za Zhi [Chinese Journal of Surgery] 2010 48 13051308. (https://doi.org/10.3760/cma.j.issn.0529-5815.2010.17.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Vicaş RM, Bodog FD, Fugaru FO, Grosu F, Badea O, Lazăr L, Cevei ML, Nistor-Cseppento CD, Beiuşanu GC, Holt G, et al.Histopathological and immunohistochemical aspects of bone tissue in aseptic necrosis of the femoral head. Romanian Journal of Morphology and Embryology 2020 61 12491258. (https://doi.org/10.47162/RJME.61.4.26)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Pascart T, Falgayrac G, Migaud H, Quinchon JF, Norberciak L, Budzik JF, Paccou J, Cotten A, Penel G, & Cortet B. Region specific Raman spectroscopy analysis of the femoral head reveals that trabecular bone is unlikely to contribute to non-traumatic osteonecrosis. Scientific Reports 2017 7 97. (https://doi.org/10.1038/s41598-017-00162-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Weinstein RS, Hogan EA, Borrelli MJ, Liachenko S, O'Brien CA, & Manolagas SC. The pathophysiological sequence of glucocorticoid-induced osteonecrosis of the femoral head in male mice. Endocrinology 2017 158 38173831. (https://doi.org/10.1210/en.2017-00662)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Wolff J The Law of Bone Remodelling: Springer Science & Business Media 2012. (https://doi.org/10.1007/978-3-642-71031-5)

  • 34.

    Brown TD, & Hild GL. Pre-collapse stress redistributions in femoral head osteonecrosis--a three-dimensional finite element analysis. Journal of Biomechanical Engineering 1983 105 171176. (https://doi.org/10.1115/1.3138402)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Ueo T, Tsutsumi S, Yamamuro T, Okumura H, Shimizu A, & Nakamura T. Biomechanical aspects of the development of aseptic necrosis of the femoral head. Archives of Orthopaedic and Traumatic Surgery. Archiv Fur Orthopadische und Unfall-Chirurgie 1985 104 145149. (https://doi.org/10.1007/BF00454690)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Utsunomiya T, Motomura G, Ikemura S, Kubo Y, Sonoda K, Hatanaka H, Baba S, Kawano K, Yamamoto T, & Nakashima Y. Effects of sclerotic changes on stress concentration in early-stage osteonecrosis: a patient-specific, 3D finite element analysis. Journal of Orthopaedic Research 2018 36 31693177. (https://doi.org/10.1002/jor.24124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Lee YK, Yoo JJ, Koo KH, Yoon KS, Min BW, & Kim HJ. Collapsed subchondral fatigue fracture of the femoral head. Orthopedic Clinics of North America 2009 40 259265. (https://doi.org/10.1016/j.ocl.2008.10.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Zhang YG, Yang Z, Zhang H, Liu M, Qiu Y, & Guo X. Negative pressure technology enhances bone regeneration in rabbit skull defects. BMC Musculoskeletal Disorders 2013 14 76. (https://doi.org/10.1186/1471-2474-14-76)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Li P, Ma YC, Sheng XY, Dong HT, Han H, Wang J, & Xia YY. Cyclic fluid shear stress promotes osteoblastic cells proliferation through ERK5 signaling pathway. Molecular and Cellular Biochemistry 2012 364 321327. (https://doi.org/10.1007/s11010-012-1233-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Tingart M, Beckmann J, Opolka A, Matsuura M, Wiech O, Grifka J, & Grässel S. Influence of factors regulating bone formation and remodeling on bone quality in osteonecrosis of the femoral head. Calcified Tissue International 2008 82 300308. (https://doi.org/10.1007/s00223-008-9111-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Sung B, Prasad S, Yadav VR, Gupta SC, Reuter S, Yamamoto N, Murakami A, & Aggarwal BB. RANKL signaling and osteoclastogenesis is negatively regulated by cardamonin. PLoS One 2013 8 e64118. (https://doi.org/10.1371/journal.pone.0064118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Mochizuki A, Takami M, Miyamoto Y, Nakamaki T, Tomoyasu S, Kadono Y, Tanaka S, Inoue T, & Kamijo R. Cell adhesion signaling regulates RANK expression in osteoclast precursors. PLoS One 2012 7 e48795. (https://doi.org/10.1371/journal.pone.0048795)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Chen G, Deng C, & Li YP. TGF-β and BMP signaling in osteoblast differentiation and bone formation. International Journal of Biological Sciences 2012 8 272–288. (https://doi.org/10.7150/ijbs.2929)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Wu M, Chen G, & Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Research 2016 4 16009. (https://doi.org/10.1038/boneres.2016.9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Medici M, van Meurs JB, Rivadeneira F, Zhao H, Arp PP, Hofman A, Pols HA, & Uitterlinden AG. BMP-2 gene polymorphisms and osteoporosis: the Rotterdam Study. Journal of Bone and Mineral Research 2006 21 845854. (https://doi.org/10.1359/jbmr.060306)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Phimphilai M, Zhao Z, Boules H, Roca H, & Franceschi RT. BMP signaling is required for RUNX2-dependent induction of the osteoblast phenotype. Journal of Bone and Mineral Research 2006 21 637646. (https://doi.org/10.1359/jbmr.060109)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Shen J, Li S, & Chen D. TGF-β signaling and the development of osteoarthritis. Bone Research 2014 2 14002–. (https://doi.org/10.1038/boneres.2014.2)

  • 48.

    Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin FB, Frassica FJ, Chang W, Yao J, et al.Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nature Medicine 2013 19 704712. (https://doi.org/10.1038/nm.3143)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Takarada T, Hinoi E, Nakazato R, Ochi H, Xu C, Tsuchikane A, Takeda S, Karsenty G, Abe T, Kiyonari H, et al.An analysis of skeletal development in osteoblast-specific and chondrocyte-specific runt-related transcription factor-2 (Runx2) knockout mice. Journal of Bone and Mineral Research 2013 28 20642069. (https://doi.org/10.1002/jbmr.1945)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Chen J, Yuan K, Mao X, Miano JM, Wu H, & Chen Y. Serum response factor regulates bone formation via IGF-1 and Runx2 signals. Journal of Bone and Mineral Research 2012 27 16591668. (https://doi.org/10.1002/jbmr.1607)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51.

    Shigemura T, Nakamura J, Kishida S, Harada Y, Ohtori S, Kamikawa K, Ochiai N, & Takahashi K. Incidence of osteonecrosis associated with corticosteroid therapy among different underlying diseases: prospective MRI study. Rheumatology (Oxford) 2011 50 20232028. (https://doi.org/10.1093/rheumatology/ker277)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Choudhry H, & Harris AL. Advances in hypoxia-inducible factor biology. Cell Metabolism 2018 27 281298. (https://doi.org/10.1016/j.cmet.2017.10.005)

  • 53.

    Schipani E, Maes C, Carmeliet G, & Semenza GL. Regulation of osteogenesis-angiogenesis coupling by HIFs and VEGF. Journal of Bone and Mineral Research 2009 24 13471353. (https://doi.org/10.1359/jbmr.090602)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Kusumbe AP, Ramasamy SK, & Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014 507 323328. (https://doi.org/10.1038/nature13145)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55.

    Wan C, Gilbert SR, Wang Y, Cao X, Shen X, Ramaswamy G, Jacobsen KA, Alaql ZS, Eberhardt AW, Gerstenfeld LC, et al.Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proceedings of the National Academy of Sciences of the United States of America 2008 105 686691. (https://doi.org/10.1073/pnas.0708474105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Wang Y, Wan C, Deng L, Liu X, Cao X, Gilbert SR, Bouxsein ML, Faugere MC, Guldberg RE, Gerstenfeld LC, et al.The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development. Journal of Clinical Investigation 2007 117 16161626. (https://doi.org/10.1172/JCI31581)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Jing X, Du T, Yang X, Zhang W, Wang G, Liu X, Li T, & Jiang Z. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1α expression. Journal of Cellular Physiology 2020 235 98649875. (https://doi.org/10.1002/jcp.29799)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Plotkin LI, Manolagas SC, & Bellido T. Glucocorticoids induce osteocyte apoptosis by blocking focal adhesion kinase-mediated survival. Evidence for inside-out signaling leading to anoikis. Journal of Biological Chemistry 2007 282 2412024130. (https://doi.org/10.1074/jbc.M611435200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59.

    Feng Z, Zheng W, Tang Q, Cheng L, Li H, Ni W, & Pan X. Fludarabine inhibits STAT1-mediated up-regulation of caspase-3 expression in dexamethasone-induced osteoblasts apoptosis and slows the progression of steroid-induced avascular necrosis of the femoral head in rats. Apoptosis 2017 22 10011012. (https://doi.org/10.1007/s10495-017-1383-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60.

    Cheng X, & Ferrell JE Jr. Apoptosis propagates through the cytoplasm as trigger waves. Science 2018 361 607612. (https://doi.org/10.1126/science.aah4065)

  • 61.

    Chotiyarnwong P, & McCloskey EV. Pathogenesis of glucocorticoid-induced osteoporosis and options for treatment. Nature Reviews. Endocrinology 2020 16 437447. (https://doi.org/10.1038/s41574-020-0341-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62.

    Weinstein RS. Clinical practice. Glucocorticoid-induced bone disease. New England Journal of Medicine 2011 365 6270. (https://doi.org/10.1056/NEJMcp1012926)

  • 63.

    She C, Zhu LQ, Zhen YF, Wang XD, & Dong QR. Activation of AMPK protects against hydrogen peroxide-induced osteoblast apoptosis through autophagy induction and NADPH maintenance: new implications for osteonecrosis treatment? Cellular Signalling 2014 26 18. (https://doi.org/10.1016/j.cellsig.2013.08.046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64.

    Wang XY, Gong LJ, Huang JM, Jiang C, & Yan ZQ. Pinocembrin alleviates glucocorticoid-induced apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway in osteocytes. European Journal of Pharmacology 2020 880 173212. (https://doi.org/10.1016/j.ejphar.2020.173212)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65.

    Pei J, Fan L, Nan K, Li J, Shi Z, Dang X, & Wang K. Excessive activation of TLR4/NF-κB interactively suppresses the canonical Wnt/β-catenin pathway and induces SANFH in SD rats. Scientific Reports 2017 7 11928. (https://doi.org/10.1038/s41598-017-12196-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66.

    Gao Y, Zhu H, Wang Q, Feng Y, & Zhang C. Inhibition of PERK signaling prevents against glucocorticoid-induced endotheliocyte apoptosis and osteonecrosis of the femoral head. International Journal of Biological Sciences 2020 16 543552. (https://doi.org/10.7150/ijbs.35256)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67.

    Yao X, Yu S, Jing X, Guo J, Sun K, Guo F, & Ye Y. PTEN inhibitor VO-OHpic attenuates GC-associated endothelial progenitor cell dysfunction and osteonecrosis of the femoral head via activating Nrf2 signaling and inhibiting mitochondrial apoptosis pathway. Stem Cell Research and Therapy 2020 11 140. (https://doi.org/10.1186/s13287-020-01658-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 68.

    Tian L, Sun S, Li W, Yuan L, & Wang X. Down-regulated microRNA-141 facilitates osteoblast activity and inhibits osteoclast activity to ameliorate osteonecrosis of the femoral head via up-regulating TGF-β2. Cell Cycle 2020 19 772786. (https://doi.org/10.1080/15384101.2020.1731053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69.

    Tao SC, Yuan T, Rui BY, Zhu ZZ, Guo SC, & Zhang CQ. Exosomes derived from human platelet-rich plasma prevent apoptosis induced by glucocorticoid-associated endoplasmic reticulum stress in rat osteonecrosis of the femoral head via the Akt/Bad/Bcl-2 signal pathway. Theranostics 2017 7 733750. (https://doi.org/10.7150/thno.17450)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 70.

    Kuang MJ, Huang Y, Zhao XG, Zhang R, Ma JX, Wang DC, & Ma XL. Exosomes derived from Wharton's jelly of human umbilical cord mesenchymal stem cells reduce osteocyte apoptosis in glucocorticoid-induced osteonecrosis of the femoral head in rats via the miR-21-PTEN-AKT signalling pathway. International Journal of Biological Sciences 2019 15 18611871. (https://doi.org/10.7150/ijbs.32262)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71.

    Levine B, & Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell 2019 176 1142. (https://doi.org/10.1016/j.cell.2018.09.048)

  • 72.

    Han Y, Zhang L, Xing Y, Zhang L, Chen X, Tang P, & Chen Z. Autophagy relieves the function inhibition and apoptosispromoting effects on osteoblast induced by glucocorticoid. International Journal of Molecular Medicine 2018 41 800808. (https://doi.org/10.3892/ijmm.2017.3270)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73.

    Zhu L, Chen J, Zhang J, Guo C, Fan W, Wang YM, & Yan Z. Parathyroid hormone (PTH) induces autophagy to protect osteocyte cell survival from dexamethasone damage. Medical Science Monitor 2017 23 40344040. (https://doi.org/10.12659/msm.903432)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 74.

    Yu T, Xie L, & Chu F. A sclerotic rim provides mechanical support for the femoral head in osteonecrosis. Orthopedics 2015 38 e374e379. (https://doi.org/10.3928/01477447-20150504-53)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75.

    Ito H, Matsuno T, & Kaneda K. Prognosis of early stage avascular necrosis of the femoral head. Clinical Orthopaedics and Related Research 1999 358 149157. (https://doi.org/10.1097/00003086-199901000-00018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 76.

    Ficat RP. Idiopathic bone necrosis of the femoral head. Early diagnosis and treatment. Journal of Bone and Joint Surgery. British Volume 1985 67 39. (https://doi.org/10.1302/0301-620X.67B1.3155745)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77.

    Wu ZS, Hong G, Yang P, Yang F, Chen ZQ, He W, & Wei QS. The survival of non-traumatic osteonecrosis of femoral head at ARCO II with ring-shaped sclerotic zone: a mid-term follow-up retrospective study. Journal of Hip Preservation Surgery 2020 7 705712. (https://doi.org/10.1093/jhps/hnab013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78.

    Zhang Y, Liu YW, Sun RB, Fan YN, Shi QY, Zhang LL, Li PF, & Li WY. "Sclerotic Band" type of classification system and measurement of necrotic area for osteonecrosis of the femoral head. Medicine (Baltimore) 2020 99 e22106. (https://doi.org/10.1097/MD.0000000000022106)

    • PubMed
    • Search Google Scholar