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 (5.9 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

Super enhancers targeting ZBTB16 in osteogenesis protect against osteoporosis

Wenhui Yu1,Zhongyu Xie1,2,Jinteng Li1Jiajie Lin1Zepeng Su1Yunshu Che1Feng Ye3Zhaoqiang Zhang1Peitao Xu1Yipeng Zeng1Xiaojun Xu1Zhikun Li1Pei Feng4Rujia Mi4Yanfeng Wu2,4( )Huiyong Shen1,2 ( )
Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518003, PR China
Shenzhen Key Laboratory of Ankylosing Spondylitis, Shenzhen 518003, PR China
Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518003, PR China

These authors contributed equally: Wenhui Yu, Zhongyu Xie

Show Author Information

Abstract

As the major cell precursors in osteogenesis, mesenchymal stem cells (MSCs) are indispensable for bone homeostasis and development. However, the primary mechanisms regulating osteogenic differentiation are controversial. Composed of multiple constituent enhancers, super enhancers (SEs) are powerful cis-regulatory elements that identify genes that ensure sequential differentiation. The present study demonstrated that SEs were indispensable for MSC osteogenesis and involved in osteoporosis development. Through integrated analysis, we identified the most common SE-targeted and osteoporosis-related osteogenic gene, ZBTB16. ZBTB16, positively regulated by SEs, promoted MSC osteogenesis but was expressed at lower levels in osteoporosis. Mechanistically, SEs recruited bromodomain containing 4 (BRD4) at the site of ZBTB16, which then bound to RNA polymerase Ⅱ-associated protein 2 (RPAP2) that transported RNA polymerase Ⅱ (POL Ⅱ) into the nucleus. The subsequent synergistic regulation of POL Ⅱ carboxyterminal domain (CTD) phosphorylation by BRD4 and RPAP2 initiated ZBTB16 transcriptional elongation, which facilitated MSC osteogenesis via the key osteogenic transcription factor SP7. Bone-targeting ZBTB16 overexpression had a therapeutic effect on the decreased bone density and remodeling capacity of Brd4fl/fl Prx1-cre mice and osteoporosis (OP) models. Therefore, our study shows that SEs orchestrate the osteogenesis of MSCs by targeting ZBTB16 expression, which provides an attractive focus and therapeutic target for osteoporosis.Without SEs located on osteogenic genes, BRD4 is not able to bind to osteogenic identity genes due to its closed structure before osteogenesis. During osteogenesis, histones on osteogenic identity genes are acetylated, and OB-gain SEs appear, enabling the binding of BRD4 to the osteogenic identity gene ZBTB16. RPAP2 transports RNA Pol Ⅱ from the cytoplasm to the nucleus and guides Pol Ⅱ to target ZBTB16 via recognition of the navigator BRD4 on SEs. After the binding of the RPAP2-Pol Ⅱ complex to BRD4 on SEs, RPAP2 dephosphorylates Ser5 at the Pol Ⅱ CTD to terminate the transcriptional pause, and BRD4 phosphorylates Ser2 at the Pol Ⅱ CTD to initiate transcriptional elongation, which synergistically drives efficient transcription of ZBTB16, ensuring proper osteogenesis. Dysregulation of SE-mediated ZBTB16 expression leads to osteoporosis, and bone-targeting ZBTB16 overexpression is efficient in accelerating bone repair and treating osteoporosis.

References

1

Salhotra, A., Shah, H. N., Levi, B. & Longaker, M. T. Mechanisms of bone development and repair. Nat. Rev. Mol. Cell Biol. 21, 696–711 (2020).

2

Wang, R., Wang, Y., Zhu, L., Liu, Y. & Li, W. Epigenetic regulation in mesenchymal stem cell aging and differentiation and osteoporosis. Stem Cells Int. 2020, 8836258 (2020).

3

Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

4

Brown, J. D. et al. BET bromodomain proteins regulate enhancer function during adipogenesis. Proc. Natl Acad. Sci. USA 115, 2144–2149 (2018).

5

Zhao, Y. et al. MyoD induced enhancer RNA interacts with hnRNPL to activate target gene transcription during myogenic differentiation. Nat. Commun. 10, 5787 (2019).

6

Chen, Z. et al. Fusion between a novel Krüppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia. Embo J. 12, 1161–1167 (1993).

7

Vincent-Fabert, C. et al. PLZF mutation alters mouse hematopoietic stem cell function and cell cycle progression. Blood 127, 1881–1885 (2016).

8

Hosokawa, H. et al. Bcl11b sets pro-T cell fate by site-specific cofactor recruitment and by repressing Id2 and Zbtb16. Nat. Immunol. 19, 1427–1440 (2018).

9

Wasim, M. et al. PLZF/ZBTB16, a glucocorticoid response gene in acute lymphoblastic leukemia, interferes with glucocorticoid-induced apoptosis. J. Steroid Biochem. Mol. Biol. 120, 218–227 (2010).

10

Sharma, M. et al. Identification of EOMES-expressing spermatogonial stem cells and their regulation by PLZF. Elife 8, e43352 (2019).

11

Barna, M., Hawe, N., Niswander, L. & Pandolfi, P. P. Plzf regulates limb and axial skeletal patterning. Nat. Genet. 25, 166–172 (2000).

12

Onizuka, S. et al. ZBTB16 as a downstream target gene of osterix regulates osteoblastogenesis of human multipotent mesenchymal stromal cells. J. Cell Biochem. 117, 2423–2434 (2016).

13

Felthaus, O., Gosau, M. & Morsczeck, C. ZBTB16 induces osteogenic differentiation marker genes in dental follicle cells independent from RUNX2. J. Periodontol. 85, e144–151 (2014).

14

Rauch, A. et al. Osteogenesis depends on commissioning of a network of stem cell transcription factors that act as repressors of adipogenesis. Nat. Genet. 51, 716–727 (2019).

15

Najafova, Z. et al. BRD4 localization to lineage-specific enhancers is associated with a distinct transcription factor repertoire. Nucleic Acids Res. 45, 127–141 (2017).

16

Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

17

Loven, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

18

Alghamdi, S. et al. BET protein inhibitor JQ1 inhibits growth and modulates WNT signaling in mesenchymal stem cells. Stem Cell Res. Ther. 7, 22 (2016).

19

Geng, Y. et al. Systematic analysis of mRNAs and ncRNAs in BMSCs of senile osteoporosis patients. Front. Genet. 12, 776984 (2021).

20

Forget, D. et al. Nuclear import of RNA polymerase Ⅱ is coupled with nucleocytoplasmic shuttling of the RNA polymerase Ⅱ-associated protein 2. Nucleic Acids Res. 41, 6881–6891 (2013).

21

Egloff, S. & Murphy, S. Cracking the RNA polymerase Ⅱ CTD code. Trends Genet. 24, 280–288 (2008).

22

Ni, Z. et al. RPRD1A and RPRD1B are human RNA polymerase Ⅱ C-terminal domain scaffolds for Ser5 dephosphorylation. Nat. Struct. Mol. Biol. 21, 686–695 (2014).

23

Devaiah, B. N. et al. BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase Ⅱ carboxy-terminal domain. Proc. Natl Acad. Sci. USA 109, 6927–6932 (2012).

24

Yang, Y. S. et al. Bone-targeting AAV-mediated silencing of Schnurri-3 prevents bone loss in osteoporosis. Nat. Commun. 10, 2958 (2019).

25

Gao, J. et al. SIRT3/SOD2 maintains osteoblast differentiation and bone formation by regulating mitochondrial stress. Cell Death Differ. 25, 229–240 (2018).

26

Pal, S., Porwal, K., Rajak, S., Sinha, R. A. & Chattopadhyay, N. Selective dietary polyphenols induce differentiation of human osteoblasts by adiponectin receptor 1-mediated reprogramming of mitochondrial energy metabolism. Biomed. Pharmacother. 127, 110207 (2020).

27

Chen, X. et al. Regulatory role of RNA N(6)-methyladenosine modification in bone biology and osteoporosis. Front. Endocrinol. 10, 911 (2019).

28

Zhang, W. et al. Differential long noncoding RNA/mRNA expression profiling and functional network analysis during osteogenic differentiation of human bone marrow mesenchymal stem cells. Stem Cell Res. Ther. 8, 30 (2017).

29

Liu, Z. et al. Myeloma cells shift osteoblastogenesis to adipogenesis by inhibiting the ubiquitin ligase MURF1 in mesenchymal stem cells. Sci. Signal 13, eaay8203 (2020).

30

Pott, S. & Lieb, J. D. What are super-enhancers? Nat. Genet. 47, 8–12 (2015).

31

Siersbaek, R. et al. Transcription factor cooperativity in early adipogenic hotspots and super-enhancers. Cell Rep. 7, 1443–1455 (2014).

32

Lee, B. K. et al. Super-enhancer-guided mapping of regulatory networks controlling mouse trophoblast stem cells. Nat. Commun. 10, 4749 (2019).

33

Paradise, C. R. et al. The epigenetic reader Brd4 is required for osteoblast differentiation. J. Cell Physiol. 235, 5293–5304 (2020).

34

Paradise, C. R. et al. Brd4 is required for chondrocyte differentiation and endochondral ossification. Bone 154, 116234 (2022).

35

Lin, L. et al. Super-enhancer-associated MEIS1 promotes transcriptional dysregulation in Ewing sarcoma in co-operation with EWS-FLI1. Nucleic Acids Res. 47, 1255–1267 (2019).

36

Shin, H. Y. et al. Hierarchy within the mammary STAT5-driven Wap super-enhancer. Nat. Genet. 48, 904–911 (2016).

37

Marofi, F. et al. Gene expression of TWIST1 and ZBTB16 is regulated by methylation modifications during the osteoblastic differentiation of mesenchymal stem cells. J. Cell Physiol. 234, 6230–6243 (2019).

38

Hall, D. D., Spitler, K. M. & Grueter, C. E. Disruption of cardiac Med1 inhibits RNA polymerase Ⅱ promoter occupancy and promotes chromatin remodeling. Am. J. Physiol. Heart Circ. Physiol. 316, H314–h325 (2019).

39

Chen, F. X., Smith, E. R. & Shilatifard, A. Born to run: control of transcription elongation by RNA polymerase Ⅱ. Nat. Rev. Mol. Cell Biol. 19, 464–478 (2018).

40

Adelman, K. & Lis, J. T. Promoter-proximal pausing of RNA polymerase Ⅱ: emerging roles in metazoans. Nat. Rev. Genet. 13, 720–731 (2012).

41

Harlen, K. M. & Churchman, L. S. The code and beyond: transcription regulation by the RNA polymerase Ⅱ carboxy-terminal domain. Nat. Rev. Mol. Cell Biol. 18, 263–273 (2017).

42

Egloff, S., Zaborowska, J., Laitem, C., Kiss, T. & Murphy, S. Ser7 phosphorylation of the CTD recruits the RPAP2 Ser5 phosphatase to snRNA genes. Mol. Cell 45, 111–122 (2012).

43

Reid, I. R. & Billington, E. O. Drug therapy for osteoporosis in older adults. Lancet 399, 1080–1092 (2022).

44

Cheng, C., Wentworth, K. & Shoback, D. M. New frontiers in osteoporosis therapy. Annu. Rev. Med. 71, 277–288 (2020).

45

Ma, Y. et al. Autophagy controls mesenchymal stem cell properties and senescence during bone aging. Aging cell 17, e12709 (2018).

46

Guo, Y. et al. Sirt3-mediated mitophagy regulates AGEs-induced BMSCs senescence and senile osteoporosis. Redox Biol. 41, 101915 (2021).

47

Colella, P., Ronzitti, G. & Mingozzi, F. Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Mol. Ther. Methods Clin. Dev. 8, 87–104 (2018).

48

Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

49

Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

50

Li, R., Li, Y., Kristiansen, K. & Wang, J. SOAP: short oligonucleotide alignment program. Bioinformatics 24, 713–714 (2008).

51

Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

52

Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

53

Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 12, 323 (2011).

54

Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

55

Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

56

Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–191 (2014).

Bone Research
Article number: 30
Cite this article:
Yu W, Xie Z, Li J, et al. Super enhancers targeting ZBTB16 in osteogenesis protect against osteoporosis. Bone Research, 2023, 11: 30. https://doi.org/10.1038/s41413-023-00267-8

138

Views

2

Downloads

2

Crossref

2

Web of Science

3

Scopus

Altmetrics

Received: 25 December 2022
Revised: 20 March 2023
Accepted: 18 April 2023
Published: 07 June 2023
© The Author(s) 2023, corrected publication 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