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 (6.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Full Length Article | Open Access

Mapk7 deletion in chondrocytes causes vertebral defects by reducing MEF2C/PTEN/AKT signaling

Chengzhi Wua,1,Hengyu Liua,1Dongmei Zhongc,1Xiaoming YangdZhiheng LiaoaYuyu ChenaShun ZhangaDeying SueBaolin ZhangaChuan LibLiru TianbCaixia Xub( )Peiqiang Sua( )
Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
Research Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
Guangdong Provincial Key Laboratory of Proteomics and State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China

1 These authors contributed equally to this work.

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

Mutation of the MAPK7 gene was related to human scoliosis. Mapk7 regulated the development of limb bones and skulls in mice. However, the role of MAPK7 in vertebral development is still unclear. In this study, we constructed Col2a1-cre; Mapk7f/f transgenic mouse model to delete Mapk7 in cartilage, which displayed kyphosis and osteopenia. Mechanistically, Mapk7 loss decreased MEF2C expression and thus activated PTEN to oppose PI3K/AKT signaling in vertebral growth plate chondrocytes, which impaired chondrocyte hypertrophy and attenuated vertebral ossification. In vivo, systemic pharmacological activation of AKT rescued impaired chondrocyte hypertrophy and alleviated mouse vertebral defects caused by Mapk7 deficiency. Our study firstly clarified the mechanism by which MAPK7 was involved in vertebral development, which might contribute to understanding the pathology of spinal deformity and provide a basis for the treatment of developmental disorders of the spine.

References

1

Scaal M. Early development of the vertebral column. Semin Cell Dev Biol. 2016;49:83-91.

2

Williams S, Alkhatib B, Serra R. Development of the axial skeleton and intervertebral disc. Curr Top Dev Biol. 2019;133:49-90.

3

Mackie EJ, Ahmed YA, Tatarczuch L, et al. Endochondral ossification: how cartilage is converted into bone in the developing skeleton. Int J Biochem Cell Biol. 2008;40(1):46-62.

4

Long F, Ornitz DM. Development of the endochondral skeleton. Cold Spring Harbor Perspect Biol. 2013;5(1):a008334.

5

Kronenberg HM. Developmental regulation of the growth plate. Nature. 2003;423(6937):332-336.

6

Yang G, Zhu L, Hou N, et al. Osteogenic fate of hypertrophic chondrocytes. Cell Res. 2014;24(10):1266-1269.

7

Zhou X, von der Mark K, Henry S, et al. Chondrocytes transdifferentiate into osteoblasts in endochondral bone during development, postnatal growth and fracture healing in mice. PLoS Genet. 2014;10(12):e1004820.

8

Yang L, Tsang KY, Tang HC, et al. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc Natl Acad Sci U S A. 2014;111(33):12097-12102.

9

Hallett SA, Ono W, Ono N. Growth plate chondrocytes: skeletal development, growth and beyond. Int J Mol Sci. 2019;20(23):6009.

10

Takeda S, Bonnamy JP, Owen MJ, et al. Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev. 2001;15(4):467-481.

11

Nithianandarajah-Jones GN, Wilm B, Goldring CEP, et al. ERK5: structure, regulation and function. Cell Signal. 2012;24(11):2187-2196.

12

Paudel R, Fusi L, Schmidt M. The MEK5/ERK5 pathway in health and disease. Int J Mol Sci. 2021;22(14):7594.

13

Zhou T, Chen C, Xu C, et al. Mutant MAPK7-induced idiopathic scoliosis is linked to impaired osteogenesis. Cell Physiol Biochem. 2018;48(3):880-890.

14

Gao W, Chen C, Zhou T, et al. Rare coding variants in MAPK7 predispose to adolescent idiopathic scoliosis. Hum Mutat. 2017;38(11):1500-1510.

15

Iezaki T, Fukasawa K, Horie T, et al. The MAPK Erk5 is necessary for proper skeletogenesis involving a Smurf-Smad-Sox9 molecular axis. Development. 2018;145(14):dev164004.

16

Yang X, Zhong D, Gao W, et al. Conditional ablation of MAPK7 expression in chondrocytes impairs endochondral bone formation in limbs and adaptation of chondrocytes to hypoxia. Cell Biosci. 2020;10:103.

17

Horie T, Fukasawa K, Yamada T, et al. Erk5 in bone marrow mesenchymal stem cells regulates bone homeostasis by preventing osteogenesis in adulthood. Stem Cell. 2022;40(4):411-422.

18

Hall KC, Hill D, Otero M, et al. ADAM17 controls endochondral ossification by regulating terminal differentiation of chondrocytes. Mol Cell Biol. 2013;33(16):3077-3090.

19

Rigueur D, Lyons KM. Whole-mount skeletal staining. Methods Mol Biol. 2014;1130:113-121.

20

Laws N, Hoey A. Progression of kyphosis in mdx mice. J Appl Physiol (1985). 2004;97(5):1970-1977.

21

Huang DW, Sherman BT, Tan Q, et al. DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 2007;35(Web Server issue):W169-W175.

22

Ran FA, Hsu PD, Wright J, et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8(11):2281-2308.

23

Fleming A, Kishida MG, Kimmel CB, et al. Building the backbone: the development and evolution of vertebral patterning. Development. 2015;142(10):1733-1744.

24

Ikegami D, Akiyama H, Suzuki A, et al. Sox9 sustains chondrocyte survival and hypertrophy in part through Pik3ca-Akt pathways. Development. 2011;138(8):1507-1519.

25

Worby CA, Dixon JE. Pten. Annu Rev Biochem. 2014;83:641-669.

26

Yang G, Sun Q, Teng Y, et al. PTEN deficiency causes dyschondroplasia in mice by enhanced hypoxia-inducible factor 1α signaling and endoplasmic reticulum stress. Development. 2008;135(21):3587-3597.

27

Kato Y, Kravchenko VV, Tapping RI, et al. BMK1/ERK5 regulates serum-induced early gene expression through transcription factor MEF2C. EMBO J. 1997;16(23):7054-7066.

28

Dy P, Wang W, Bhattaram P, et al. Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes. Dev Cell. 2012;22(3):597-609.

29

Arnold MA, Kim Y, Czubryt MP, et al. MEF2C transcription factor controls chondrocyte hypertrophy and bone development. Dev Cell. 2007;12(3):377-389.

30

Scheuermann HW. The classic: kyphosis dorsalis juvenilis. Clin Orthop Relat Res. 1977;(128):5-7.

31

Lowe TG, Line BG. Evidence based medicine: analysis of Scheuermann kyphosis. Spine. 2007;32(19 Suppl):S115-S119.

32

Bradford DS, Brown DM, Moe JH, et al. Scheuermann’s kyphosis: a form of osteoporosis? Clin Orthop Relat Res. 1976(118):10-15.

33

Damborg F, Engell V, Andersen M, et al. Prevalence, concordance, and heritability of Scheuermann kyphosis based on a study of twins. J Bone Joint Surg Am. 2006;88(10):2133-2136.

34

Palazzo C, Sailhan F, Revel M. Scheuermann’s disease: an update. Jt Bone Spine. 2014;81(3):209-214.

35

Ueta C, Iwamoto M, Kanatani N, et al. Skeletal malformations caused by overexpression of Cbfa1 or its dominant negative form in chondrocytes. J Cell Biol. 2001;153(1):87-100.

36

Ulici V, Hoenselaar KD, Gillespie JR, et al. The PI3K pathway regulates endochondral bone growth through control of hypertrophic chondrocyte differentiation. BMC Dev Biol. 2008;8:40.

37

Hsieh SC, Chen NT, Lo SH. Conditional loss of PTEN leads to skeletal abnormalities and lipoma formation. Mol Carcinog. 2009;48(6):545-552.

38

Kim JM, Yang YS, Park KH, et al. A RUNX2 stabilization pathway mediates physiologic and pathologic bone formation. Nat Commun. 2020;11:2289.

39

Yoshida CA, Yamamoto H, Fujita T, et al. Runx2 and Runx3 are essential for chondrocyte maturation, and Runx2 regulates limb growth through induction of Indian hedgehog. Genes Dev. 2004;18(8):952-963.

40

Fujita T, Azuma Y, Fukuyama R, et al. Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J Cell Biol. 2004;166(1):85-95.

41

Salhotra A, Shah HN, Levi B, et al. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol. 2020;21(11):696-711.

42

Loveridge CJ, van ’t Hof RJ, Charlesworth G, et al. Analysis of Nkx3.1:Cre-driven Erk5 deletion reveals a profound spinal deformity which is linked to increased osteoclast activity. Sci Rep. 2017;7:13241.

43

Shu HS, Liu YL, Tang XT, et al. Tracing the skeletal progenitor transition during postnatal bone formation. Cell Stem Cell. 2021;28(12):2122-2136.e3.

44

Ono N, Ono W, Nagasawa T, et al. A subset of chondrogenic cells provides early mesenchymal progenitors in growing bones. Nat Cell Biol. 2014;16(12):1157-1167.

45

Mizuhashi K, Ono W, Matsushita Y, et al. Resting zone of the growth plate houses a unique class of skeletal stem cells. Nature. 2018;563(7730):254-258.

46

Chan CF, Seo E, Chen J, et al. Identification and specification of the mouse skeletal stem cell. Cell. 2015;160(1-2):285-298.

47

Bobick BE, Matsche AI, Chen FH, et al. The ERK5 and ERK1/2 signaling pathways play opposing regulatory roles during chondrogenesis of adult human bone marrow-derived multipotent progenitor cells. J Cell Physiol. 2010;224(1):178-186.

48

Stokes IA, Mente PL, Iatridis JC, et al. Enlargement of growth plate chondrocytes modulated by sustained mechanical loading. J Bone Joint Surg Am. 2002;84(10):1842-1848.

49

Lee D, Erickson A, Dudley AT, et al. Mechanical stimulation of growth plate chondrocytes: previous approaches and future directions. Exp Mech. 2019;59(9):1261-1274.

Genes & Diseases
Pages 964-977
Cite this article:
Wu C, Liu H, Zhong D, et al. Mapk7 deletion in chondrocytes causes vertebral defects by reducing MEF2C/PTEN/AKT signaling. Genes & Diseases, 2024, 11(2): 964-977. https://doi.org/10.1016/j.gendis.2023.02.012

261

Views

3

Downloads

4

Crossref

3

Web of Science

3

Scopus

0

CSCD

Altmetrics

Received: 07 November 2022
Revised: 17 January 2023
Accepted: 07 February 2023
Published: 24 March 2023
© 2023 The Authors.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return