AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (8.5 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

FAR591 promotes the pathogenesis and progression of SONFH by regulating Fos expression to mediate the apoptosis of bone microvascular endothelial cells

Fei Zhang1,2Lei Wei3Lei Wang2Tao Wang2Zhihong Xie2Hong Luo2Fanchao Li2Jian Zhang1,2Wentao Dong1,2Gang Liu1,2Qinglin Kang4Xuesong Zhu5Wuxun Peng1,2 ( )
Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, China
Department of Orthopedics, Rhode Island Hospital, Brown University, Providence, Rhode Island 02903, USA
Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
Show Author Information

Abstract

The specific pathogenesis of steroid-induced osteonecrosis of the femoral head (SONFH) is still not fully understood, and there is currently no effective early cure. Understanding the role and mechanism of long noncoding RNAs (lncRNAs) in the pathogenesis of SONFH will help reveal the pathogenesis of SONFH and provide new targets for its early prevention and treatment. In this study, we first confirmed that glucocorticoid (GC)-induced apoptosis of bone microvascular endothelial cells (BMECs) is a pre-event in the pathogenesis and progression of SONFH. Then, we identified a new lncRNA in BMECs via lncRNA/mRNA microarray, termed Fos-associated lincRNA ENSRNOT00000088059.1 (FAR591). FAR591 is highly expressed during GC-induced BMEC apoptosis and femoral head necrosis. Knockout of FAR591 effectively blocked the GC-induced apoptosis of BMECs, which then alleviated the damage of GCs to the femoral head microcirculation and inhibited the pathogenesis and progression of SONFH. In contrast, overexpression of FAR591 significantly promoted the GC-induced apoptosis of BMECs, which then aggravated the damage of GCs to the femoral head microcirculation and promoted the pathogenesis and progression of SONFH. Mechanistically, GCs activate the glucocorticoid receptor, which translocates to the nucleus and directly acts on the FAR591 gene promoter to induce FAR591 gene overexpression. Subsequently, FAR591 binds to the Fos gene promoter (–245~–51) to form a stable RNA:DNA triplet structure and then recruits TATA-box binding protein associated factor 15 and RNA polymerase Ⅱ to promote Fos expression through transcriptional activation. Fos activates the mitochondrial apoptotic pathway by regulating the expression of Bcl-2 interacting mediator of cell death (Bim) and P53 upregulated modulator of apoptosis (Puma) to mediate GC-induced apoptosis of BMECs, which leads to femoral head microcirculation dysfunction and femoral head necrosis. In conclusion, these results confirm the mechanistic link between lncRNAs and the pathogenesis of SONFH, which helps reveal the pathogenesis of SONFH and provides a new target for the early prevention and treatment of SONFH.

References

1

Fu, W., Liu, B., Wang, B. & Zhao, D. Early diagnosis and treatment of steroid-induced osteonecrosis of the femoral head. Int. Orthop. 43, 1083–1087 (2019).

2

Li, Z. et al. Circulating microRNA signature of steroid-induced osteonecrosis of the femoral head. Cell Prolif. 51, e12418 (2018).

3

Wang, A., Ren, M. & Wang, J. The pathogenesis of steroid-induced osteonecrosis of the femoral head: a systematic review of the literature. Gene 671, 103–109 (2018).

4

Maruyama, M. et al. The effects of a functionally-graded scaffold and bone marrow- derived mononuclear cells on steroid-induced femoral head osteonecrosis. Biomaterials 187, 39–46 (2018).

5

Li, T. et al. Discovery and validation an eight-biomarker serum gene signature for the diagnosis of steroid-induced osteonecrosis of the femoral head. Bone 122, 199–208 (2019).

6

Jiang, L. Y., Yu, X. & Pang, Q. J. Research in the precaution of recombinant human erythropoietin to steroid-induced osteonecrosis of the rat femoral head. J. Int. Med. Res. 45, 1324–1331 (2017).

7

Chang, C., Greenspan, A. & Gershwin, M. E. The pathogenesis, diagnosis and clinical manifestations of steroid-induced osteonecrosis. J. Autoimmun. 110, 102460 (2020).

8

Kerachian, M. A., Séguin, C. & Harvey, E. J. Glucocorticoids in osteonecrosis of the femoral head: a new understanding of the mechanisms of action. J. Steroid Biochem. Mol. Biol. 114, 121–128 (2009).

9

Zuo, R. et al. Exosomes derived from human CD34(+) stem cells transfected with miR-26a prevent glucocorticoid-induced osteonecrosis of the femoral head by promoting angiogenesis and osteogenesis. Stem Cell Res. Ther. 10, 321 (2019).

10

Ramasamy, S. K., Kusumbe, A. P., Wang, L. & Adams, R. H. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature 507, 376–380 (2014).

11

Yang, M. et al. MiR-497~195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch and HIF-1α activity. Nat. Commun. 8, 16003 (2017).

12

Zhu, S. et al. Endothelial cells produce angiocrine factors to regulate bone and cartilage via versatile mechanisms. Theranostics 10, 5957–5965 (2020).

13

Filipowska, J., Tomaszewski, K. A., Niedźwiedzki, Ł., Walocha, J. A. & Niedźwiedzki, T. The role of vasculature in bone development, regeneration and proper systemic functioning. Angiogenesis 20, 291–302 (2017).

14

Krüger-Genge, A., Blocki, A., Franke, R. P. & Jung, F. Vascular endothelial cell biology: an update. Int. J. Mol. Sci. 20, 4411 (2019).

15

Falkenberg, K. D., Rohlenova, K., Luo, Y. & Carmeliet, P. The metabolic engine of endothelial cells. Nat. Metab. 1, 937–946 (2019).

16

Hassanshahi, M., Hassanshahi, A., Khabbazi, S., Su, Y. W. & Xian, C. J. Bone marrow sinusoidal endothelium: damage and potential regeneration following cancer radiotherapy or chemotherapy. Angiogenesis 20, 427–442 (2017).

17

Zuo, W., Guo, W. S., Yu, H. C., Liu, P. & Zhang, Q. D. Role of junction-mediating and regulatory protein in the pathogenesis of glucocorticoid-induced endothelial cell lesions. Orthop. Surg. 12, 964–973 (2020).

18

Huang, Z., Wang, Q., Zhang, T., Fu, Y. & Wang, W. Hyper-activated platelet lysates prevent glucocorticoid-associated femoral head necrosis by regulating autophagy. Biomed. Pharmacother. 139, 111711 (2021).

19

Prisby, R. D. Bone marrow microvasculature. Compr. Physiol. 10, 1009–1046 (2020).

20

McCarthy, I. The physiology of bone blood flow: a review. J. Bone Jt. Surg. Am. 88(Suppl 3), 4–9 (2006).

21

Zhang, Q., Jin, L. V. & Jin, L. Role of coagulopathy in glucocorticoid-induced osteonecrosis of the femoral head. J. Int. Med. Res. 46, 2141–2148 (2018).

22

Zhang, Y., Yin, J., Ding, H., Zhang, C. & Gao, Y. S. Vitamin K2 ameliorates damage of blood vessels by glucocorticoid: a potential mechanism for its protective effects in glucocorticoid- induced osteonecrosis of the femoral head in a rat model. Int. J. Biol. Sci. 12, 776–785 (2016).

23

Clarisse, D., Offner, F. & De Bosscher, K. Latest perspectives on glucocorticoid-induced apoptosis and resistance in lymphoid malignancies. Biochim. Biophys. Acta Rev. Cancer 1874, 188430 (2020).

24

Dirks-Naylor, A. J. & Griffiths, C. L. Glucocorticoid-induced apoptosis and cellular mechanisms of myopathy. J. Steroid Biochem. Mol. Biol. 117, 1–7 (2009).

25

Schmidt, S. et al. Glucocorticoid-induced apoptosis and glucocorticoid resistance: molecular mechanisms and clinical relevance. Cell Death Differ. 11(Suppl 1), S45–S55 (2004).

26

Jing, D. et al. Opposing regulation of BIM and BCL2 controls glucocorticoid-induced apoptosis of pediatric acute lymphoblastic leukemia cells. Blood 125, 273–283 (2015).

27

Ploner, C. et al. The BCL2 rheostat in glucocorticoid-induced apoptosis of acute lymphoblastic leukemia. Leukemia 22, 370–377 (2008).

28

Zhan, J. et al. Allicin inhibits osteoblast apoptosis and steroid-induced necrosis of femoral head progression by activating the PI3K/AKT pathway. Food Funct. 11, 7830–7841 (2020).

29

Kopp, F. & Mendell, J. T. Functional classification and experimental dissection of long noncoding RNAs. Cell 172, 393–407 (2018).

30

Sanchez Calle, A., Kawamura, Y., Yamamoto, Y., Takeshita, F. & Ochiya, T. Emerging roles of long non-coding RNA in cancer. Cancer Sci. 109, 2093–2100 (2018).

31

Orom, U. A. & Shiekhattar, R. Noncoding RNAs and enhancers: complications of a long-distance relationship. Trends Genet. 27, 433–439 (2011).

32

Romero-Barrios, N., Legascue, M. F., Benhamed, M., Ariel, F. & Crespi, M. Splicing regulation by long noncoding RNAs. Nucleic Acids Res. 46, 2169–2184 (2018).

33

Ali, T. & Grote, P. Beyond the RNA-dependent function of LncRNA genes. Elife 9, e60583 (2020).

34

Kazimierczyk, M., Kasprowicz, M. K., Kasprzyk, M. E. & Wrzesinski, J. Human long noncoding RNA interactome: detection, characterization and function. Int. J. Mol. Sci. 21, 1027 (2020).

35

Noh, J. H., Kim, K. M., McClusky, W. G., Abdelmohsen, K. & Gorospe, M. Cytoplasmic functions of long noncoding RNAs. Wiley Interdiscip. Rev. RNA 9, e1471 (2018).

36

Shin, T. J., Lee, K. H. & Cho, J. Y. Epigenetic mechanisms of LncRNAs binding to protein in carcinogenesis. Cancers 12, 2925 (2020).

37

Zhang, H., Xu, H. B., Kurban, E. & Luo, H. W. LncRNA SNHG14 promotes hepatocellular carcinoma progression via H3K27 acetylation activated PABPC1 by PTEN signaling. Cell Death Dis. 11, 646 (2020).

38

Arab, K. et al. Long noncoding RNA TARID directs demethylation and activation of the tumor suppressor TCF21 via GADD45A. Mol. Cell 55, 604–614 (2014).

39

Lan, T. et al. KIAA1429 contributes to liver cancer progression through N6-methyladenosine-dependent post-transcriptional modification of GATA3. Mol. Cancer 18, 186 (2019).

40

Zhang, F. et al. LncRNA CRNDE attenuates chemoresistance in gastric cancer via SRSF6-regulated alternative splicing of PICALM. Mol. Cancer 20, 6 (2021).

41

Han, M. et al. Exosome-mediated lncRNA AFAP1-AS1 promotes trastuzumab resistance through binding with AUF1 and activating ERBB2 translation. Mol. Cancer 19, 26 (2020).

42

Wu, Z. et al. LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation. Mol. Neurobiol. 54, 7670–7685 (2017).

43

Li, L. et al. Bone marrow mesenchymal stem cell-derived exosomes promote plasminogen activator inhibitor 1 expression in vascular cells in the local microenvironment during rabbit osteonecrosis of the femoral head. Stem Cell Res Ther. 11, 480 (2020).

44

Chen, C. Y. et al. Extracellular vesicles from human urine-derived stem cells inhibit glucocorticoid-induced osteonecrosis of the femoral head by transporting and releasing pro-angiogenic DMBT1 and anti-apoptotic TIMP1. Acta Biomater. 111, 208–220 (2020).

45

Yao, X. et al. 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 Res Ther. 11, 140 (2020).

46

Chen, C. et al. Impairment of two types of circulating endothelial progenitor cells in patients with glucocorticoid-induced avascular osteonecrosis of the femoral head. Jt. Bone Spine 80, 70–76 (2013).

47

Yu, H. et al. Icariin promotes angiogenesis in glucocorticoid-induced osteonecrosis of femoral heads: in vitro and in vivo studies. J. Cell Mol. Med. 23, 7320–7330 (2019).

48

Jing, X. et al. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1alpha expression. J. Cell Physiol. 235, 9864–9875 (2020).

49

Dykes, I. M. & Emanueli, C. Transcriptional and post-transcriptional gene regulation by long non-coding RNA. Genom. Proteom. Bioinforma. 15, 177–186 (2017).

50

Zhang, X. et al. Mechanisms and functions of long non-coding RNAs at multiple regulatory levels. Int. J. Mol. Sci. 20, 5573 (2019).

51

Li, P. et al. Inhibition of long noncoding RNA HIF1A-AS2 confers protection against atherosclerosis via ATF2 downregulation. J. Adv. Res. 26, 123–135 (2020).

52

Cheng, D. et al. LncRNA HOTAIR epigenetically suppresses miR-122 expression in hepatocellular carcinoma via DNA methylation. EBioMedicine 36, 159–170 (2018).

53

Zhu, L. et al. Impaired autophagic degradation of lncRNA ARHGAP5-AS1 promotes chemoresistance in gastric cancer. Cell Death Dis. 10, 383 (2019).

54

Wang, F. W. et al. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J. Clin. Investig. 129, 727–743 (2019).

55

Malakar, P. et al. Long noncoding RNA MALAT1 regulates cancer glucose metabolism by enhancing mTOR-mediated translation of TCF7L2. Cancer Res. 79, 2480–2493 (2019).

56

Yu, Y. et al. LncRNA TUG1 promoted osteogenic differentiation through promoting bFGF ubiquitination. Vitr. Cell Dev. Biol. Anim. 56, 42–48 (2020).

57

Kong, L. et al. CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res. 35, W345–W349 (2007).

58

Wright, M. W. A short guide to long non-coding RNA gene nomenclature. Hum. Genom. 8, 7 (2014).

59

Akhter, R., Sanphui, P., Das, H., Saha, P. & Biswas, S. C. The regulation of p53 up-regulated modulator of apoptosis by JNK/c-Jun pathway in beta-amyloid-induced neuron death. J. Neurochem. 134, 1091–1103 (2015).

60

Eferl, R. & Wagner, E. F. AP-1: a double-edged sword in tumorigenesis. Nat. Rev. Cancer 3, 859–868 (2003).

61

Springhower, C. E., Rosen, M. K. & Chook, Y. M. Karyopherins and condensates. Curr. Opin. Cell Biol. 64, 112–123 (2020).

62

Janke, A. M. et al. Lysines in the RNA polymerase Ⅱ C-terminal domain contribute to TAF15 fibril recruitment. Biochemistry 57, 2549–2563 (2018).

63

Zhang, J., Pan, J. & Jing, W. Motivating role of type H vessels in bone regeneration. Cell Prolif. 53, e12874 (2020).

64

Statello, L., Guo, C. J., Chen, L. L. & Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 22, 96–118 (2021).

65

St Laurent, G., Wahlestedt, C. & Kapranov, P. The Landscape of long noncoding RNA classification. Trends Genet. 31, 239–251 (2015).

66

Chen, L. L. Linking long noncoding RNA localization and function. Trends Biochem. Sci. 41, 761–772 (2016).

67

Xu, J. et al. A comprehensive overview of lncRNA annotation resources. Brief. Bioinform 18, 236–249 (2017).

68

Zhao, X., Su, L., He, X., Zhao, B. & Miao, J. Long noncoding RNA CA7-4 promotes autophagy and apoptosis via sponging MIR877-3P and MIR5680 in high glucose-induced vascular endothelial cells. Autophagy 16, 70–85 (2020).

69

Leisegang, M. S. et al. Long noncoding RNA MANTIS facilitates endothelial angiogenic function. Circulation 136, 65–79 (2017).

70

Peng, W. X., Koirala, P. & Mo, Y. Y. LncRNA-mediated regulation of cell signaling in cancer. Oncogene 36, 5661–5667 (2017).

71

Long, Y., Wang, X., Youmans, D. T. & Cech, T. R. How do lncRNAs regulate transcription? Sci. Adv. 3, eaao2110 (2017).

72

Robinson, E. K., Covarrubias, S. & Carpenter, S. The how and why of lncRNA function: an innate immune perspective. Biochim Biophys. Acta Gene Regul. Mech. 1863, 194419 (2020).

73

Bridges, M. C., Daulagala, A. C. & Kourtidis, A. LNCcation: lncRNA localization and function. J. Cell Biol. 220, e202009045 (2021).

74

Ju, C. et al. Mesenchymal stem cell-associated lncRNA in osteogenic differentiation. Biomed. Pharmacother. 115, 108912 (2019).

75

Grossi, E. et al. A lncRNA-SWI/SNF complex crosstalk controls transcriptional activation at specific promoter regions. Nat. Commun. 11, 936 (2020).

76

Trembinski, D. J. et al. Aging-regulated anti-apoptotic long non-coding RNA Sarrah augments recovery from acute myocardial infarction. Nat. Commun. 11, 2039 (2020).

77

Kwon, I. et al. Phosphorylation-regulated binding of RNA polymerase Ⅱ to fibrous polymers of low-complexity domains. Cell 155, 1049–1060 (2013).

78

Singh, A. K., Kapoor, V., Thotala, D. & Hallahan, D. E. TAF15 contributes to the radiation- inducible stress response in cancer. Oncotarget 11, 2647–2659 (2020).

79

Kashyap, M., Ganguly, A. K. & Bhavesh, N. S. Structural delineation of stem-loop RNA binding by human TAF15 protein. Sci. Rep. 5, 17298 (2015).

80

Bonucci, A., Murrali, M. G., Banci, L. & Pierattelli, R. A combined NMR and EPR investigation on the effect of the disordered RGG regions in the structure and the activity of the RRM domain of FUS. Sci. Rep. 10, 20956 (2020).

Bone Research
Article number: 27
Cite this article:
Zhang F, Wei L, Wang L, et al. FAR591 promotes the pathogenesis and progression of SONFH by regulating Fos expression to mediate the apoptosis of bone microvascular endothelial cells. Bone Research, 2023, 11: 27. https://doi.org/10.1038/s41413-023-00259-8

121

Views

1

Downloads

7

Crossref

7

Web of Science

7

Scopus

Altmetrics

Received: 08 August 2022
Revised: 27 February 2023
Accepted: 09 March 2023
Published: 22 May 2023
© The Author(s) 2023

This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Return