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 (11.6 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

Nutrient-regulated dynamics of chondroprogenitors in the postnatal murine growth plate

Takeshi Oichi1,2,3( )Joe Kodama1Kimberly Wilson1Hongying Tian1Yuka Imamura Kawasawa4Yu Usami5Yasushi Oshima2Taku Saito2Sakae Tanaka2Masahiro Iwamoto1Satoru Otsuru1Motomi Enomoto-Iwamoto1 ( )
Department of Orthopedics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
Sensory & Motor System Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 1138655, Japan
Department of Orthopedics, Teikyo University School of Medicine, Tokyo 1738608, Japan
Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
Department of Oral Pathology, Osaka University Graduate School of Dentistry, Suita, Osaka 5650871, Japan
Show Author Information

Abstract

Longitudinal bone growth relies on endochondral ossification in the cartilaginous growth plate, where chondrocytes accumulate and synthesize the matrix scaffold that is replaced by bone. The chondroprogenitors in the resting zone maintain the continuous turnover of chondrocytes in the growth plate. Malnutrition is a leading cause of growth retardation in children; however, after recovery from nutrient deprivation, bone growth is accelerated beyond the normal rate, a phenomenon termed catch-up growth. Although nutritional status is a known regulator of long bone growth, it is largely unknown whether and how chondroprogenitor cells respond to deviations in nutrient availability. Here, using fate-mapping analysis in Axin2CreERT2 mice, we showed that dietary restriction increased the number of Axin2+ chondroprogenitors in the resting zone and simultaneously inhibited their differentiation. Once nutrient deficiency was resolved, the accumulated chondroprogenitor cells immediately restarted differentiation and formed chondrocyte columns, contributing to accelerated growth. Furthermore, we showed that nutrient deprivation reduced the level of phosphorylated Akt in the resting zone and that exogenous IGF-1 restored the phosphorylated Akt level and stimulated differentiation of the pooled chondroprogenitors, decreasing their numbers. Our study of Axin2CreERT2 revealed that nutrient availability regulates the balance between accumulation and differentiation of chondroprogenitors in the growth plate and further demonstrated that IGF-1 partially mediates this regulation by promoting the committed differentiation of chondroprogenitor cells.

References

1

Kronenberg, H. M. Developmental regulation of the growth plate. Nature 423, 332–336 (2003).

2

Kember, N. F. Cell division in endochondral ossification. A study of cell proliferation in rat bones by the method of tritiated thymidine autoradiography. J. Bone Joint Surg. Br. 42B, 824–839 (1960).

3

Abad, V. et al. The role of the resting zone in growth plate chondrogenesis. Endocrinology 143, 1851–1857 (2002).

4

Mizuhashi, K. et al. Resting zone of the growth plate houses a unique class of skeletal stem cells. Nature 563, 254–258 (2018).

5

Newton, P. T. et al. A radical switch in clonality reveals a stem cell niche in the epiphyseal growth plate. Nature 567, 234–238 (2019).

6

Renthal, N. E., Nakka, P., Baronas, J. M., Kronenberg, H. M. & Hirschhorn, J. N. Genes with specificity for expression in the round cell layer of the growth plate are enriched in genomewide association study (GWAS) of human height. J. Bone Miner. Res. 36, 2300–2308 (2021).

7

Black, R. E. et al. Maternal and child undernutrition: global and regional exposures and health consequences. Lancet 371, 243–260 (2008).

8

Victora, C. G. et al. Maternal and child undernutrition: consequences for adult health and human capital. Lancet 371, 340–357 (2008).

9

Prader, A., Tanner, J. M. & von, H. G. Catch-up growth following illness or starvation. An example of developmental canalization in man. J. Pediatr. 62, 646–659 (1963).

10

Gafni, R. I. & Baron, J. Catch-up growth: possible mechanisms. Pediatr. Nephrol. 14, 616–619 (2000).

11

Clevers, H., Loh, K. M. & Nusse, R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science 346, 1248012 (2014).

12

Lustig, B. et al. Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors. Mol. Cell Biol. 22, 1184–1193 (2002).

13

van Amerongen, R., Bowman, A. N. & Nusse, R. Developmental stage and time dictate the fate of Wnt/beta-catenin-responsive stem cells in the mammary gland. Cell Stem Cell 11, 387–400 (2012).

14

Lim, X. et al. Interfollicular epidermal stem cells self-renew via autocrine Wnt signaling. Science 342, 1226–1230 (2013).

15

Wang, B., Zhao, L., Fish, M., Logan, C. Y. & Nusse, R. Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver. Nature 524, 180–185 (2015).

16

Maruyama, T., Jeong, J., Sheu, T. J. & Hsu, W. Stem cells of the suture mesenchyme in craniofacial bone development, repair and regeneration. Nat. Commun. 7, 10526 (2016).

17

Usami, Y. et al. Possible contribution of wnt-responsive chondroprogenitors to the postnatal murine growth plate. J. Bone Miner. Res. 34, 964–974 (2019).

18

Chan, C. K. et al. Identification and specification of the mouse skeletal stem cell. Cell 160, 285–298 (2015).

19

Muruganandan, S. et al. A FoxA2+ long-term stem cell population is necessary for growth plate cartilage regeneration after injury. Nat. Commun. 13, 2515 (2022).

20

Haseeb, A. et al. SOX9 keeps growth plates and articular cartilage healthy by inhibiting chondrocyte dedifferentiation/osteoblastic redifferentiation. Proc. Natl Acad. Sci. USA 118, e2019152118 (2021).

21

Hallett, S. A. et al. Chondrocytes in the resting zone of the growth plate are maintained in a Wnt-inhibitory environment. Elife 10, e64513 (2021).

22

Lui, J. C. et al. Differential aging of growth plate cartilage underlies differences in bone length and thus helps determine skeletal proportions. PLoS Biol. 16, e2005263 (2018).

23

St-Jacques, B., Hammerschmidt, M. & McMahon, A. P. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev. 13, 2072–2086 (1999).

24

Arnold, M. A. et al. MEF2C transcription factor controls chondrocyte hypertrophy and bone development. Dev. Cell 12, 377–389 (2007).

25

Hirai, T., Chagin, A. S., Kobayashi, T., Mackem, S. & Kronenberg, H. M. Parathyroid hormone/parathyroid hormone-related protein receptor signaling is required for maintenance of the growth plate in postnatal life. Proc. Natl. Acad. Sci. USA 108, 191–196 (2011).

26

Xie, L. et al. Effects of dietary calorie restriction or exercise on the PI3K and Ras signaling pathways in the skin of mice. J. Biol. Chem. 282, 28025–28035 (2007).

27

Peng, X. D. et al. Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. Genes Dev. 17, 1352–1365 (2003).

28

Brazil, D. P., Yang, Z. Z. & Hemmings, B. A. Advances in protein kinase B signalling: AKTion on multiple fronts. Trends Biochem. Sci. 29, 233–242 (2004).

29

Galvan, V., Logvinova, A., Sperandio, S., Ichijo, H. & Bredesen, D. E. Type 1 insulin-like growth factor receptor (IGF-IR) signaling inhibits apoptosis signal-regulating kinase 1 ASK1.J. Biol. Chem. 278, 13325–13332 (2003).

30

McMullen, J. R. et al. Deletion of ribosomal S6 kinases does not attenuate pathological, physiological, or insulin-like growth factor 1 receptor-phosphoinositide 3-kinase-induced cardiac hypertrophy. Mol. Cell Biol. 24, 6231–6240 (2004).

31

Radcliff, K. et al. Insulin-like growth factor-Ⅰ regulates proliferation and osteoblastic differentiation of calcifying vascular cells via extracellular signal-regulated protein kinase and phosphatidylinositol 3-kinase pathways. Circ. Res. 96, 398–400 (2005).

32

Kalluri, H. S., Vemuganti, R. & Dempsey, R. J. Mechanism of insulin-like growth factor Ⅰ-mediated proliferation of adult neural progenitor cells: role of Akt. Eur. J. Neurosci. 25, 1041–1048 (2007).

33

Underwood, L. E., Thissen, J. P., Lemozy, S., Ketelslegers, J. M. & Clemmons, D. R. Hormonal and nutritional regulation of IGF-Ⅰ and its binding proteins. Horm. Res. 42, 145–151 (1994).

34

Thissen, J. P., Ketelslegers, J. M. & Underwood, L. E. Nutritional regulation of the insulin-like growth factors. Endocr. Rev. 15, 80–101 (1994).

35

Cheng, C. W. et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression. Cell Stem Cell 14, 810–823 (2014).

36

Baron, J. et al. Catch-up growth after glucocorticoid excess: a mechanism intrinsic to the growth plate. Endocrinology 135, 1367–1371 (1994).

37

Yilmaz, O. H. et al. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 486, 490–495 (2012).

38

Chen, J., Astle, C. M. & Harrison, D. E. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp. Hematol. 31, 1097–1103 (2003).

39

Forni, M. F. et al. Caloric restriction promotes structural and metabolic changes in the skin. Cell Rep. 20, 2678–2692 (2017).

40

Lee, J., Seroogy, K. B. & Mattson, M. P. Dietary restriction enhances neurotrophin expression and neurogenesis in the hippocampus of adult mice. J. Neurochem. 80, 539–547 (2002).

41

Rosello-Diez, A. & Joyner, A. L. Regulation of long bone growth in vertebrates; it is time to catch up. Endocr. Rev. 36, 646–680 (2015).

42

Fattal-Valevski, A., Toledano-Alhadef, H., Golander, A., Leitner, Y. & Harel, S. Endocrine profile of children with intrauterine growth retardation. J. Pediatr. Endocrinol. Metab. 18, 671–676 (2005).

43

Kamei, H. et al. Role of IGF signaling in catch-up growth and accelerated temporal development in zebrafish embryos in response to oxygen availability. Development 138, 777–786 (2011).

44

Wang, J., Zhou, J. & Bondy, C. A. Igf1 promotes longitudinal bone growth by insulin-like actions augmenting chondrocyte hypertrophy. Faseb J. 13, 1985–1990 (1999).

45

Wang, Y. et al. IGF-1R signaling in chondrocytes modulates growth plate development by interacting with the PTHrP/Ihh pathway. J. Bone Miner. Res. 26, 1437–1446 (2011).

46

Cooper, K. L. et al. Multiple phases of chondrocyte enlargement underlie differences in skeletal proportions. Nature 495, 375–378 (2013).

47

Lui, J. C., Nilsson, O. & Baron, J. Recent research on the growth plate: recent insights into the regulation of the growth plate. J. Mol. Endocrinol. 53, T1–T9 (2014).

48

Lindahl, A., Nilsson, A. & Isaksson, O. G. Effects of growth hormone and insulin-like growth factor-Ⅰ on colony formation of rabbit epiphyseal chondrocytes at different stages of maturation. J. Endocrinol. 115, 263–271 (1987).

49

Hill, D. J. Stimulation of cartilage zones in the calf costochondral growth plate in vitro by growth hormone dependent plasma somatomedin activity. J. Endocrinol. 83, 219 (1979).

50

Makower, A. M., Wroblewski, J. & Pawlowski, A. Effects of IGF-Ⅰ, EGF, and FGF on proteoglycans synthesized by fractionated chondrocytes of rat rib growth plate. Exp. Cell Res. 179, 498–506 (1988).

51

Makower, A. M., Skottner, A. & Wroblewski, J. Binding of insulin-like growth factor-Ⅰ (IGF-Ⅰ) to primary cultures of chondrocytes from rat rib growth cartilage. Cell Biol. Int. Rep. 13, 655–665 (1989).

52

Ohlsson, C., Nilsson, A., Isaksson, O. & Lindahl, A. Growth hormone induces multiplication of the slowly cycling germinal cells of the rat tibial growth plate. Proc. Natl. Acad. Sci. USA 89, 9826–9830 (1992).

53

Hunziker, E. B., Wagner, J. & Zapf, J. Differential effects of insulin-like growth factor Ⅰ and growth hormone on developmental stages of rat growth plate chondrocytes in vivo. J. Clin. Invest. 93, 1078–1086 (1994).

54

Caselli, A. et al. IGF-1-mediated osteoblastic niche expansion enhances long-term hematopoietic stem cell engraftment after murine bone marrow transplantation. Stem Cells 31, 2193–2204 (2013).

55

Lui, J. C. et al. Cartilage-targeted IGF-1 treatment to promote longitudinal bone growth. Mol. Ther. 27, 673–680 (2019).

56

Mori, Y., Chung, U. I., Tanaka, S. & Saito, T. Determination of differential gene expression profiles in superficial and deeper zones of mature rat articular cartilage using RNA sequencing of laser microdissected tissue specimens. Biomed. Res. 35, 263–270 (2014).

Bone Research
Article number: 20
Cite this article:
Oichi T, Kodama J, Wilson K, et al. Nutrient-regulated dynamics of chondroprogenitors in the postnatal murine growth plate. Bone Research, 2023, 11: 20. https://doi.org/10.1038/s41413-023-00258-9

126

Views

1

Downloads

9

Crossref

3

Web of Science

3

Scopus

Altmetrics

Received: 03 April 2022
Revised: 03 March 2023
Accepted: 09 March 2023
Published: 21 April 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