The Role of Signaling Pathway on Osteoprogenitor Cell Behavior and Bone Formation

authors:

avatar Maedeh Hashemi 1 , avatar Vahid Shakeri 2 , avatar Haleh Mosallaei 3 , avatar Mona Kholdebarin 4 , avatar Mohamad Bakhtiari 5 , avatar Masoomeh Mohamadpour 6 , avatar Saeed Gholami 3 , avatar Seyed Amir Yazdanparast 7 , *

Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
Department of Orthopedic, Arak University of Medical Sciences, Arak, Iran
Iran University of Medical Sciences, Tehran, Iran
Science and Research Branch, Islamic Azad University, Tehran, Iran
Anatomy Department, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
Neuroscience Research Center, School of Advanced Medical Sciences and Technologies, Iran University of Medical Sciences, Tehran, Iran
Mycology Department, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran

how to cite: Hashemi M , Shakeri V , Mosallaei H , Kholdebarin M , Bakhtiari M , et al. The Role of Signaling Pathway on Osteoprogenitor Cell Behavior and Bone Formation. Thrita. 2018;7(1):e81485. https://doi.org/10.5812/thrita.81485.

Abstract

Different biomarkers and signaling pathways are involved in bone development. These factors lead to different osteoprogenitor cell reactions. Proliferation, migration, and differentiation of osteoprogenitor cells is induced by different intracellular and extracellular molecular signaling pathways. As mesenchymal stem cells (MSCs) and their differentiation during repair and development of bone processes have significant roles, it is believed that sensible molecular signaling pathways involvement is vital to the development of bone formation, bone substitute materials, and bone repair. This study reviews various keys of signaling pathway that terminate proliferation, migration, and differentiation of osteoprogenitor cells.

1. Context

Mesenchyme and stem cell have particular actions and function, such as proliferation, migration, and differentiation, independent of signaling pathways, biomarkers and etc. Researchers for the first time showed an origin of human mesenchyme stem cells, which is from the perivascular area (1-3). Furthermore, HMSC with special cluster of differentiation, such as CD37, CD90, and CD40 with little increased expression of CD14, CD34, CD45, and human leucocyte-DR (HLA-DR) can differentiate to osteoblast cells (4). Researchers exploit HMSC from different sources, such as dental tissue, endometria menstrual, blood, peripheral blood, placenta and fetal membrane salivary gland, skin, synovial fluid, foreskin, endometrium amniotic fluid, sub-amniotic umbilical cord lining membrane, and Wharton jelly (5).

Different types of bone cells have a significant effect in development and formation of bone (mineralization and matrix).

During life, bone is remodeled and osteoblast cells differentiate from mesenchyme progenitor cells that proliferate migration and differentiation, which dependents on balancing at bone mass and function (3). Good performance of osteoblast cells is required in the maintenance of regular bone homeostasis. This evolution is coordinated by multiple extracellular signals that are transduced by cell signaling pathways to monitor specialized changes in gene expression

Bone, as a specialized form of connective tissue, is the main component of the skeletal system. Bone formation is very complex and is controlled by multiple signaling factors that play a major role in the regulation of osteoblast and other bone lineage-specific differentiation (4, 5).

2. Evidence Acquisition

2.1. Effect of Ras-MAPK-Pathway on Osteoprogenitor Cells

Receptor tyrosine kinase, as a signaling pathway, has a key role in the development of osteoprogenitor cells.

Ras facilitates activation of mitogen activation protein kinase (MAPK), (phosphatidylinositol, tol 3’-kinase) PI3K and insulin-like growth factors (IGFs) by loss of the NF1 gene (6-8).

Ras-MAPK-pathway in osteoblast progress regulates EPK signaling to form the skeletal structure. Most times, EPK facilitates differentiation of osteoprogenitor cells without changing proliferation (9).

The PI3K pathway as well as EPK plays a key role in osteoprogenitor for proliferation, by inhibition of EPK signaling pathway (10).

FGFR3 has a negative regulation on function and proliferation of osteoprogenitor cells, and inhibits IHH signaling pathway to maintain PTHLH expression in reserve and articular chondrocyte (11).

2.2. Effect of BMP/ TGF-βs Pathway on Osteogenitor Cells

The transforming growth factor-β (TGF-β) superfamily is comprised of TGF-βs, activin, bone morphogenetic proteins (BMPs), and other related proteins. TGF-β acts through the heteromeric receptor complex, comprised of type I and type II receptors at the cell surface that transduces intracellular signals by Smad complex or MAPK cascade (12-17). At least 29 and probably up to 42 TGF-β, five type II receptors and seven type I receptors are encoded by the human genome (18, 19). Signals transduced by TGF-β superfamily members control the formation of tissue differentiation, through their effects on cell proliferation, differentiation, and migration (20-23).

Change of BMP and TGF-β as key factors of formation and development of osteoprogenitor cells leads to bone disorders, such as osteoarthritis, brachydactyl type A2, and tumor metastasis (24).

Furthermore, BMP activates Smad 1 and 5, as extracellular signals. TGF-β signaling pathway regulates proliferation, differentiation, and bone formation by activation of Smad2, 3. The MAPK cascade is involved in significant cellular action, such as movement, division, death and cell differentiation. To regenerate bone, P38/MAPK signaling activation has a significant effect for induction of osteoblast differentiation (25-32).

2.3. Effect of Ca2+ Signaling Pathway on Osteogenitor Cells

Ca2+, as a significant factor in osteoprogenitor cells, differentiates to osteoblast and regulates Runx 2. Furthermore, Runx 2 signaling pathway regulates ATP-dependent Ca2+ influx through calcium channels (33).

AP-1 is up-regulated by high levels of Ca2+ intracellular and leads to activation of growth factors, such as IGF1/2, VEGF, TGF-β, and BMP, and these factors cause high osteoblastic function and enhancement of mineralization capacity of osteoprogenitor cells. It has been established that Ca2+ results in significant enhancement of the proliferation level of hPDCs and MC3T3 and morphology alternation from fibroblastic to cuboidal, which is a prominent normal function of osteoblasts. On the other hand, revelation of the influence of Ca2+ signaling may also be profitable in the explanation of how CaP biomaterials can rise osteoblastic activation and high mineralization capacity of osteoprogenitor cells (34-37). The Ca2+ channels are actived by CaP crystals, which leads to high expression levels of the bone sialoprotein, osteopontin, and ALP (38). Furthermore, upon treatment of hMSCs with elevated Ca2+, enhancement in proliferation rate and expression of bone related genes, such as osteocalcin, bone sialoprotein and osteopontin, as well as BMP-2 was observed, indicating that Ca2+ is a potential osteoinductive trigger in hMSCs by interacting with the BMP2 signaling pathway (39).

2.4. Effect of Wnt Active Wnt/β Catenin Signaling Pathway on Osteogenitor Cells

Wnt, as a large family of ligands, for membrane-spooning frizzled (FZD) receptors, is involved in proliferation, growth, migration, polarity, differentiation, and cell death (40, 41). Wnt/Ca2+ and Wnt/planer facilitie osteoblast differentiation and mineralization processes. Wnt30 enhances proliferation of osteoprogenitor cells. Wnt-responding cells, which undergo a transient step of cell differentiation induced by local Wnt stimuli, regulate bone formation (42-44).

2.5. Effect of IGF Signaling Pathway on Osteogenitor Cells

These significant members of IGF, IGF1 and IGF2, are involved in proliferation and differentiation of osteoblast (45). IGF2 is the growth factor that is responsible for osteoblast differentiation. However, in contract to IGF1, IGF2 modulates and maintains bone mass. Both IGFs are expressed in osteoblasts and have similar biological function and properties (46, 47). Induction of osteoblast differentiation, stimulation of bone matrix deposition, expression of collagens, and non-collagenous proteins are among various effects of IGF1 and IGF2 in bone (48). Additionally, IGF-1 in combination with BMP2 has a stimulatory impression on the proliferation and osteogenic differentiation of stem cells derived from adipose tissue (49).

2.6. Effect of Panx3 Signaling Pathway on Osteoprogenitor Cells

Pannexins (Panxs) were recently recognized as a new gap junction protein family (50, 51). The Panx family consists of three members, Panx1, 2, and 3. Panx1 and 2 are ubiquitously expressed with particularly strong expression in the central nervous system (52). Panx3 is a member that was most recently identified by genome bioinformatic analysis (53). However, Panx 3 is expressed in certain soft tissues, such as skin and coronary arteries and developing hard tissues, including cartilage and bone. Panx3 has a negative impression on regulation proliferation and differentiation of osteoprogenitor cells. Panx signaling pathway inhibits proliferation by deduction of expression of cAmp/plcA signaling (54, 55).

3. Results

In summary, review of the literature indicates that a signaling pathway regulates proliferation, migration, differentiation of osteoprogenitor cells to osteoblast and bone formation. The current results indicated that different signaling pathways and different stages of cells have different actions.

4. Conclusions

This study reviewed how natural fracture restoration and regeneration take place following bone injuries. The different signal pathways in bone formation by MSCs have been discussed. Finally, this research discussed specifications and the involvement of diverse key molecular signaling pathways in special bone cells development. The current authors believe that a deeper understanding of the molecular signaling pathways involved in bone formation and structure can help bring novel therapies from the bench to the bedside in bone injury.

Acknowledgements

References

  • 1.

    Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res. 2015;48:62. [PubMed ID: 26530042]. [PubMed Central ID: PMC4630918]. https://doi.org/10.1186/s40659-015-0053-4.

  • 2.

    Zheng AQ, Xiao J, Xie J, Lu PP, Ding X. bFGF enhances activation of osteoblast differentiation and osteogenesis on titanium surfaces via PI3K/Akt signaling pathway. Int J Clin Exp Pathol. 2016;9(4):4680-92.

  • 3.

    Pogozhykh O, Pogozhykh D, Neehus AL, Hoffmann A, Blasczyk R, Muller T. Molecular and cellular characteristics of human and non-human primate multipotent stromal cells from the amnion and bone marrow during long term culture. Stem Cell Res Ther. 2015;6:150. [PubMed ID: 26297012]. [PubMed Central ID: PMC4546288]. https://doi.org/10.1186/s13287-015-0146-6.

  • 4.

    Anam K, Davis TA. Comparative analysis of gene transcripts for cell signaling receptors in bone marrow-derived hematopoietic stem/progenitor cell and mesenchymal stromal cell populations. Stem Cell Res Ther. 2013;4(5):112. [PubMed ID: 24405801]. [PubMed Central ID: PMC3854681]. https://doi.org/10.1186/scrt323.

  • 5.

    Alvarez-Viejo M, Menendez-Menendez Y, Otero-Hernandez J. CD271 as a marker to identify mesenchymal stem cells from diverse sources before culture. World J Stem Cells. 2015;7(2):470-6. [PubMed ID: 25815130]. [PubMed Central ID: PMC4369502]. https://doi.org/10.4252/wjsc.v7.i2.470.

  • 6.

    Campbell SL, Khosravi-Far R, Rossman KL, Clark GJ, Der CJ. Increasing complexity of Ras signaling. Oncogene. 1998;17(11 Reviews):1395-413. [PubMed ID: 9779987]. https://doi.org/10.1038/sj.onc.1202174.

  • 7.

    Vojtek AB, Der CJ. Increasing complexity of the ras signaling pathway. J Biol Chem. 1998;273(32):19925-8. https://doi.org/10.1074/jbc.273.32.19925.

  • 8.

    Mitin N, Rossman KL, Der CJ. Signaling interplay in Ras superfamily function. Curr Biol. 2005;15(14):R563-74. [PubMed ID: 16051167]. https://doi.org/10.1016/j.cub.2005.07.010.

  • 9.

    Ory S, Morrison DK. Signal transduction: implications for Ras-dependent ERK signaling. Curr Biol. 2004;14(7):R277-8. [PubMed ID: 15062121]. https://doi.org/10.1016/j.cub.2004.03.023.

  • 10.

    Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. 2003;4(5):373-84. [PubMed ID: 12728271]. https://doi.org/10.1038/nrm1105.

  • 11.

    Schindeler A, Little DG. Ras-MAPK signaling in osteogenic differentiation: friend or foe? J Bone Miner Res. 2006;21(9):1331-8. https://doi.org/10.1359/jbmr.060603.

  • 12.

    Rahman MS, Akhtar N, Jamil HM, Banik RS, Asaduzzaman SM. TGF-beta/BMP signaling and other molecular events: regulation of osteoblastogenesis and bone formation. Bone Res. 2015;3:15005. [PubMed ID: 26273537]. [PubMed Central ID: PMC4472151]. https://doi.org/10.1038/boneres.2015.5.

  • 13.

    Wu M, Chen G, Li YP. TGF-beta and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 2016;4:16009. [PubMed ID: 27563484]. [PubMed Central ID: PMC4985055]. https://doi.org/10.1038/boneres.2016.9.

  • 14.

    Beederman M, Lamplot JD, Nan G, Wang J, Liu X, Yin L, et al. BMP signaling in mesenchymal stem cell differentiation and bone formation. J Biomed Sci Eng. 2013;6(8A):32-52. [PubMed ID: 26819651]. [PubMed Central ID: PMC4725591]. https://doi.org/10.4236/jbise.2013.68A1004.

  • 15.

    Botchkarev VA. Bone morphogenetic proteins and their antagonists in skin and hair follicle biology. J Invest Dermatol. 2003;120(1):36-47. [PubMed ID: 12535196]. https://doi.org/10.1046/j.1523-1747.2003.12002.x.

  • 16.

    Lechleider RJ, Ryan JL, Garrett L, Eng C, Deng C, Wynshaw-Boris A, et al. Targeted mutagenesis of Smad1 reveals an essential role in chorioallantoic fusion. Dev Biol. 2001;240(1):157-67. [PubMed ID: 11784053]. https://doi.org/10.1006/dbio.2001.0469.

  • 17.

    Zhao GQ. Consequences of knocking out BMP signaling in the mouse. Genesis. 2003;35(1):43-56. [PubMed ID: 12481298]. https://doi.org/10.1002/gene.10167.

  • 18.

    Urist MR. Bone morphogenetic protein: the molecularization of skeletal system development. J Bone Miner Res. 1997;12(3):343-6. [PubMed ID: 9076576]. https://doi.org/10.1359/jbmr.1997.12.3.343.

  • 19.

    Carreira AC, Lojudice FH, Halcsik E, Navarro RD, Sogayar MC, Granjeiro JM. Bone morphogenetic proteins: facts, challenges, and future perspectives. J Dent Res. 2014;93(4):335-45. [PubMed ID: 24389809]. https://doi.org/10.1177/0022034513518561.

  • 20.

    Feng XH, Derynck R. Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol. 2005;21:659-93. [PubMed ID: 16212511]. https://doi.org/10.1146/annurev.cellbio.21.022404.142018.

  • 21.

    Balemans W, Van Hul W. Extracellular regulation of BMP signaling in vertebrates: a cocktail of modulators. Dev Biol. 2002;250(2):231-50. [PubMed ID: 12376100].

  • 22.

    Moustakas A, Heldin CH. The regulation of TGFbeta signal transduction. Development. 2009;136(22):3699-714. [PubMed ID: 19855013]. https://doi.org/10.1242/dev.030338.

  • 23.

    Schmierer B, Hill CS. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007;8(12):970-82. [PubMed ID: 18000526]. https://doi.org/10.1038/nrm2297.

  • 24.

    Guo L, Zhao RC, Wu Y. The role of microRNAs in self-renewal and differentiation of mesenchymal stem cells. Exp Hematol. 2011;39(6):608-16. [PubMed ID: 21288479]. https://doi.org/10.1016/j.exphem.2011.01.011.

  • 25.

    Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from the laboratory to the clinic, part I (basic concepts). J Tissue Eng Regen Med. 2008;2(1):1-13. [PubMed ID: 18293427]. https://doi.org/10.1002/term.63.

  • 26.

    Pan A, Chang L, Nguyen A, James AW. A review of hedgehog signaling in cranial bone development. Front Physiol. 2013;4:61. [PubMed ID: 23565096]. [PubMed Central ID: PMC3613593]. https://doi.org/10.3389/fphys.2013.00061.

  • 27.

    Horikiri Y, Shimo T, Kurio N, Okui T, Matsumoto K, Iwamoto M, et al. Sonic hedgehog regulates osteoblast function by focal adhesion kinase signaling in the process of fracture healing. PLoS One. 2013;8(10). e76785. [PubMed ID: 24124594]. [PubMed Central ID: PMC3790742]. https://doi.org/10.1371/journal.pone.0076785.

  • 28.

    Reichert JC, Schmalzl J, Prager P, Gilbert F, Quent VM, Steinert AF, et al. Synergistic effect of Indian hedgehog and bone morphogenetic protein-2 gene transfer to increase the osteogenic potential of human mesenchymal stem cells. Stem Cell Res Ther. 2013;4(5):105. [PubMed ID: 24004723]. [PubMed Central ID: PMC3854715]. https://doi.org/10.1186/scrt316.

  • 29.

    Kim JH, Liu X, Wang J, Chen X, Zhang H, Kim SH, et al. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Ther Adv Musculoskelet Dis. 2013;5(1):13-31. [PubMed ID: 23514963]. [PubMed Central ID: PMC3582304]. https://doi.org/10.1177/1759720X12466608.

  • 30.

    Issack PS, Helfet DL, Lane JM. Role of Wnt signaling in bone remodeling and repair. HSS J. 2008;4(1):66-70. [PubMed ID: 18751865]. [PubMed Central ID: PMC2504275]. https://doi.org/10.1007/s11420-007-9072-1.

  • 31.

    Terada K, Misao S, Katase N, Nishimatsu S, Nohno T. Interaction of Wnt Signaling with BMP/Smad Signaling during the Transition from Cell Proliferation to Myogenic Differentiation in Mouse Myoblast-Derived Cells. Int J Cell Biol. 2013;2013:616294. [PubMed ID: 23864860]. [PubMed Central ID: PMC3705783]. https://doi.org/10.1155/2013/616294.

  • 32.

    Lin GL, Hankenson KD. Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J Cell Biochem. 2011;112(12):3491-501. [PubMed ID: 21793042]. [PubMed Central ID: PMC3202082]. https://doi.org/10.1002/jcb.23287.

  • 33.

    Davies J, Warwick J, Totty N, Philp R, Helfrich M, Horton M. The osteoclast functional antigen, implicated in the regulation of bone resorption, is biochemically related to the vitronectin receptor. J Cell Biol. 1989;109(4 Pt 1):1817-26. [PubMed ID: 2477382]. [PubMed Central ID: PMC2115816].

  • 34.

    Di Capite J, Ng SW, Parekh AB. Decoding of cytoplasmic Ca(2+) oscillations through the spatial signature drives gene expression. Curr Biol. 2009;19(10):853-8. [PubMed ID: 19375314]. https://doi.org/10.1016/j.cub.2009.03.063.

  • 35.

    Hwang SY, Putney JW. Calcium signaling in osteoclasts. Mol Cell Res. 2011;1813(5):979-83. https://doi.org/10.1016/j.bbamcr.2010.11.002.

  • 36.

    Sun X, McLamore E, Kishore V, Fites K, Slipchenko M, Porterfield DM, et al. Mechanical stretch induced calcium efflux from bone matrix stimulates osteoblasts. Bone. 2012;50(3):581-91. [PubMed ID: 22227434]. https://doi.org/10.1016/j.bone.2011.12.015.

  • 37.

    Jung H, Best M, Akkus O. Microdamage induced calcium efflux from bone matrix activates intracellular calcium signaling in osteoblasts via L-type and T-type voltage-gated calcium channels. Bone. 2015;76:88-96. [PubMed ID: 25819792]. https://doi.org/10.1016/j.bone.2015.03.014.

  • 38.

    Kanno T, Takahashi T, Tsujisawa T, Ariyoshi W, Nishihara T. Mechanical stress-mediated Runx2 activation is dependent on Ras/ERK1/2 MAPK signaling in osteoblasts. J Cell Biochem. 2007;101(5):1266-77. [PubMed ID: 17265428]. https://doi.org/10.1002/jcb.21249.

  • 39.

    Jung H, Akkus O. Activation of intracellular calcium signaling in osteoblasts colocalizes with the formation of post-yield diffuse microdamage in bone matrix. Bonekey Rep. 2016;5:778. [PubMed ID: 26962448]. [PubMed Central ID: PMC4774084]. https://doi.org/10.1038/bonekey.2016.5.

  • 40.

    Gruber J, Yee Z, Tolwinski NS. Developmental drift and the role of wnt signaling in aging. Cancers (Basel). 2016;8(8). [PubMed ID: 27490570]. [PubMed Central ID: PMC4999782]. https://doi.org/10.3390/cancers8080073.

  • 41.

    Shi J, Chi S, Xue J, Yang J, Li F, Liu X. Emerging role and therapeutic implication of wnt signaling pathways in autoimmune diseases. J Immunol Res. 2016;2016:9392132. [PubMed ID: 27110577]. [PubMed Central ID: PMC4826689]. https://doi.org/10.1155/2016/9392132.

  • 42.

    Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling. J Clin Invest. 2006;116(5):1202-9. [PubMed ID: 16670761]. [PubMed Central ID: PMC1451219]. https://doi.org/10.1172/JCI28551.

  • 43.

    Ray S, Khassawna TE, Sommer U, Thormann U, Wijekoon ND, Lips K, et al. Differences in expression of Wnt antagonist Dkk1 in healthy versus pathological bone samples. J Microsc. 2017;265(1):111-20. [PubMed ID: 27580425]. https://doi.org/10.1111/jmi.12469.

  • 44.

    Ransom RC, Hunter DJ, Hyman S, Singh G, Ransom SC, Shen EZ, et al. Axin2-expressing cells execute regeneration after skeletal injury. Sci Rep. 2016;6:36524. [PubMed ID: 27853243]. [PubMed Central ID: PMC5113299]. https://doi.org/10.1038/srep36524.

  • 45.

    Kimura T, Kuwata T, Ashimine S, Yamazaki M, Yamauchi C, Nagai K, et al. Targeting of bone-derived insulin-like growth factor-II by a human neutralizing antibody suppresses the growth of prostate cancer cells in a human bone environment. Clin Cancer Res. 2010;16(1):121-9. [PubMed ID: 20028742]. [PubMed Central ID: PMC2802676]. https://doi.org/10.1158/1078-0432.CCR-09-0982.

  • 46.

    Arvidson K, Abdallah BM, Applegate LA, Baldini N, Cenni E, Gomez-Barrena E, et al. Bone regeneration and stem cells. J Cell Mol Med. 2011;15(4):718-46. [PubMed ID: 21129153]. [PubMed Central ID: PMC3922662]. https://doi.org/10.1111/j.1582-4934.2010.01224.x.

  • 47.

    Guo Y, Tang CY, Man XF, Tang HN, Tang J, Zhou CL, et al. Insulin-like growth factor-1 promotes osteogenic differentiation and collagen I alpha 2 synthesis via induction of mRNA-binding protein LARP6 expression. Dev Growth Differ. 2017;59(2):94-103. [PubMed ID: 28211947]. https://doi.org/10.1111/dgd.12342.

  • 48.

    Elias WY. Assessment of the osteogenic potential of morphogenetic protein-2 and insulin-like growth factor-I on adipose tissuederived stem cells. J Biomed Sci. 2016;5(1):1-6.

  • 49.

    Majidinia M, Sadeghpour A, Yousefi B. The roles of signaling pathways in bone repair and regeneration. J Cell Physiol. 2018;233(4):2937-48. [PubMed ID: 28590066]. https://doi.org/10.1002/jcp.26042.

  • 50.

    Iwamoto T, Nakamura T, Doyle A, Ishikawa M, de Vega S, Fukumoto S, et al. Pannexin 3 regulates intracellular ATP/cAMP levels and promotes chondrocyte differentiation. J Biol Chem. 2010;285(24):18948-58. [PubMed ID: 20404334]. [PubMed Central ID: PMC2881817]. https://doi.org/10.1074/jbc.M110.127027.

  • 51.

    Hung CT, Allen FD, Mansfield KD, Shapiro IM. Extracellular ATP modulates [Ca2+]i in retinoic acid-treated embryonic chondrocytes. Am J Physiol. 1997;272(5 Pt 1):C1611-7. [PubMed ID: 9176153]. https://doi.org/10.1152/ajpcell.1997.272.5.C1611.

  • 52.

    Ishikawa M, Iwamoto T, Nakamura T, Doyle A, Fukumoto S, Yamada Y. Pannexin 3 functions as an ER Ca(2+) channel, hemichannel, and gap junction to promote osteoblast differentiation. J Cell Biol. 2011;193(7):1257-74. [PubMed ID: 21690309]. [PubMed Central ID: PMC3216329]. https://doi.org/10.1083/jcb.201101050.

  • 53.

    Ishikawa M, Williams GL, Ikeuchi T, Sakai K, Fukumoto S, Yamada Y. Pannexin 3 and connexin 43 modulate skeletal development through their distinct functions and expression patterns. J Cell Sci. 2016;129(5):1018-30. [PubMed ID: 26759176]. [PubMed Central ID: PMC4813316]. https://doi.org/10.1242/jcs.176883.

  • 54.

    Koga T, Matsui Y, Asagiri M, Kodama T, de Crombrugghe B, Nakashima K, et al. NFAT and Osterix cooperatively regulate bone formation. Nat Med. 2005;11(8):880-5. [PubMed ID: 16041384]. https://doi.org/10.1038/nm1270.

  • 55.

    Ishikawa M, Yamada Y. The role of pannexin 3 in bone biology. J Dent Res. 2016;96(4):372-9. https://doi.org/10.1177/0022034516678203.