1,217
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

SPRY4 promotes adipogenic differentiation of human mesenchymal stem cells through the MEK–ERK1/2 signaling pathway

, , , &
Pages 588-600 | Received 15 Jul 2022, Accepted 06 Sep 2022, Published online: 15 Sep 2022

References

  • van Dijk Sj, Molloy PL, Varinli H, et al. Epigenetics and human obesity [J]. Int J Obes (Lond). 2015;39(1):85–97.
  • Ng M, Fleming T, Robinson M, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the global burden of disease study 2013 [J]. Lancet. 2014;384(9945):766–781.
  • Malik VS, Willett WC, Hu FB. Global obesity: trends, risk factors and policy implications [J]. Nat Rev Endocrinol. 2013;9(1):13–27.
  • Zhang Y, Zheng Y, Fu Y, et al. Identification of biomarkers, pathways and potential therapeutic agents for white adipocyte insulin resistance using bioinformatics analysis [J]. Adipocyte. 2019;8(1):318–329.
  • Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity [J]. J Clin Invest. 2019;129(10):4022–4031.
  • Sim MO, Lee HJ, Jeong DE, et al. 6’-O-acetyl mangiferin from Iris rossii Baker inhibits lipid accumulation partly via AMPK activation in adipogenesis [J]. Chem Biol Interact. 2019;311:108755.
  • Wei Z, Chen N, Guo H, et al. Bone marrow mesenchymal stem cells from leukemia patients inhibit growth and apoptosis in serum-deprived K562 cells [J]. J Exp Clin Cancer Res. 2009;28(1):141.
  • Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells [J]. Differentiation. 2016;92(1–2):41–51.
  • Tang QQ, Lane MD. Adipogenesis: from stem cell to adipocyte [J]. Annu Rev Biochem. 2012;81(1):715–736.
  • Golpanian S, Wolf A, Hatzistergos KE, et al. Rebuilding the damaged heart: mesenchymal stem cells, cell-based therapy, and engineered heart tissue [J]. Physiol Rev. 2016;96(3):1127–1168.
  • Kim HJ, Bar-Sagi D. Modulation of signalling by Sprouty: a developing story [J]. Nat Rev Mol Cell Biol. 2004;5(6):441–450.
  • Mason JM, Morrison DJ, Basson MA, et al. Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling [J]. Trends Cell Biol. 2006;16(1):45–54.
  • Mandl M, Wagner SA, Hatzmann FM, et al. Sprouty1 is a weight-loss target gene in human adipose stem/progenitor cells that is mandatory for the initiation of adipogenesis [J]. Cell Death Dis. 2019;10(6):411.
  • Mandl M, Wagner SA, Hatzmann FM, et al. Sprouty1 prevents cellular senescence maintaining proliferation and differentiation capacity of human adipose stem/progenitor cells [J]. J Gerontol A Biol Sci Med Sci. 2020;75(12):2308–2319.
  • Urs S, Venkatesh D, Tang Y, et al. Sprouty1 is a critical regulatory switch of mesenchymal stem cell lineage allocation [J]. Faseb J. 2010;24(9):3264–3273.
  • Yang X, Pande S, Koza RA, et al. Sprouty1 regulates gonadal white adipose tissue growth through a PDGFRα/β-Akt pathway [J]. Adipocyte. 2021;10(1):574–586.
  • Park S, Arai Y, Kim BJ, et al. Suppression of SPRY4 promotes osteogenic differentiation and bone formation of mesenchymal stem cell [J]. Tissue Eng Part A. 2019;25(23–24):1646–1657.
  • Tian L, Xiao H, Li M, et al. A novel Sprouty4-ERK1/2-Wnt/β-catenin regulatory loop in marrow stromal progenitor cells controls osteogenic and adipogenic differentiation [J]. Metabolism. 2020;105:154189.
  • Li J, Li N, Chen Y, et al. SPRY4 is responsible for pathogenesis of adolescent idiopathic scoliosis by contributing to osteogenic differentiation and melatonin response of bone marrow-derived mesenchymal stem cells [J]. Cell Death Dis. 2019;10(11):805.
  • Huang S, Wang S, Bian C, et al. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression [J]. Stem Cells Dev. 2012;21(13):2531–2540.
  • Li H, Fan J, Fan L, et al. MiRNA-10b reciprocally stimulates osteogenesis and inhibits adipogenesis partly through the TGF-β/SMAD2 signaling pathway [J]. Aging Dis. 2018;9(6):1058–1073.
  • Sztalryd C, Brasaemle DL. The perilipin family of lipid droplet proteins: gatekeepers of intracellular lipolysis [J]. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862(10 Pt B):1221–1232.
  • Christofori G. Split personalities: the agonistic antagonist Sprouty [J]. Nat Cell Biol. 2003;5(5):377–379.
  • Aubert J, Belmonte N, Dani C. Role of pathways for signal transducers and activators of transcription, and mitogen-activated protein kinase in adipocyte differentiation [J]. Cell Mol Life Sci. 1999;56(5–6):538–542.
  • Bost F, Aouadi M, Caron L, et al. The role of MAPKs in adipocyte differentiation and obesity [J]. Biochimie. 2005;87(1):51–56.
  • Lee M, Sorn SR, Lee Y, et al. Salt induces adipogenesis/lipogenesis and inflammatory adipocytokines secretion in adipocytes [J]. Int J Mol Sci. 2019;20(1):160.
  • Ning X, He J, Shi X, et al. Regulation of adipogenesis by quinine through the ERK/S6 pathway [J]. Int J Mol Sci. 2016;17(4):504.
  • Gregoire FM, Smas CM, Sul HS. Understanding adipocyte differentiation [J]. Physiol Rev. 1998;78(3):783–809.
  • Zhu XY, Ma S, Eirin A, et al. Functional plasticity of adipose-derived stromal cells during development of obesity [J]. Stem Cells Transl Med. 2016;5(7):893–900.
  • Louwen F, Ritter A, Kreis NN, et al. Insight into the development of obesity: functional alterations of adipose-derived mesenchymal stem cells [J]. Obes Rev. 2018;19(7):888–904.
  • Matsushita K, Dzau VJ. Mesenchymal stem cells in obesity: insights for translational applications [J]. Lab Invest. 2017;97(10):1158–1166.
  • Hacohen N, Kramer S, Sutherland D, et al. sprouty encodes a novel antagonist of FGF signaling that patterns apical branching of the Drosophila airways [J]. Cell. 1998;92(2):253–263.
  • Casci T, Vinós J, Freeman M. Sprouty, an intracellular inhibitor of Ras signaling [J]. Cell. 1999;96(5):655–665.
  • Doriguzzi A, Haigl B, Gsur A, et al. The increased Sprouty4 expression in response to serum is transcriptionally controlled by Specific protein 1 [J]. Int J Biochem Cell Biol. 2015;64:220–228.
  • Joo A, Long R, Cheng Z, et al. Sprouty2 regulates endochondral bone formation by modulation of RTK and BMP signaling [J]. Bone. 2016;88:170–179.
  • Cabrita MA, Christofori G. Sprouty proteins, masterminds of receptor tyrosine kinase signaling [J]. Angiogenesis. 2008;11(1):53–62.
  • Masoumi-Moghaddam S, Amini A, Morris DL. The developing story of Sprouty and cancer [J]. Cancer Metastasis Rev. 2014;33(2–3):695–720.
  • Guy GR, Wong ES, Yusoff P, et al. Sprouty: how does the branch manager work? [J]. J Cell Sci. 2003;116(Pt 15):3061–3068.
  • Li X, Wheldon L, Heath JK. Sprouty: a controversial role in receptor tyrosine kinase signalling pathways [J]. Biochem Soc Trans. 2003;31(Pt 6):1445–1446.
  • Impagnatiello MA, Weitzer S, Gannon G, et al. Mammalian sprouty-1 and −2 are membrane-anchored phosphoprotein inhibitors of growth factor signaling in endothelial cells [J]. J Cell Biol. 2001;152(5):1087–1098.
  • Egan JE, Hall AB, Yatsula BA, et al. The bimodal regulation of epidermal growth factor signaling by human Sprouty proteins [J]. Proc Natl Acad Sci U S A. 2002;99(9):6041–6046.
  • Hall AB, Jura N, DaSilva J, et al. hSpry2 is targeted to the ubiquitin-dependent proteasome pathway by c-Cbl [J]. Curr Biol. 2003;13(4):308–314.
  • Rubin C, Litvak V, Medvedovsky H, et al. Sprouty fine-tunes EGF signaling through interlinked positive and negative feedback loops [J]. Curr Biol. 2003;13(4):297–307.
  • Cabrita MA, Christofori G. Sprouty proteins: antagonists of endothelial cell signaling and more [J]. Thromb Haemost. 2003;90(4):586–590.
  • Moseti D, Regassa A, Kim WK. Molecular regulation of adipogenesis and potential anti-adipogenic bioactive molecules [J]. Int J Mol Sci. 2016;17(1):124.
  • Wu WJ, Mo DL, Zhao CZ, et al. Knockdown of CTRP6 inhibits adipogenesis via lipogenic marker genes and Erk1/2 signalling pathway [J]. Cell Biol Int. 2015;39(5):554–562.
  • de Mora J F, Porras A, Ahn N, et al. Mitogen-activated protein kinase activation is not necessary for, but antagonizes, 3T3-L1 adipocytic differentiation [J]. Mol Cell Biol. 1997;17(10):6068–6075.
  • Takeno A, Kanazawa I, Notsu M, et al. phloretin promotes adipogenesis via mitogen-activated protein kinase pathways in mouse marrow stromal ST2 cells [J]. Int J Mol Sci. 2018;19(6):1772.
  • Liu GX, Zhu JC, Chen XY, et al. Inhibition of adipogenic differentiation of bone marrow mesenchymal stem cells by erythropoietin via activating ERK and P38 MAPK [J]. Genet Mol Res. 2015;14(2):6968–6977.
  • Kim S, Ahn C, Bong N, et al. Biphasic effects of FGF2 on adipogenesis [J]. PLoS One. 2015;10(3):e0120073.
  • Maurizi G, Petäistö T, Maurizi A, et al. Key-genes regulating the liposecretion process of mature adipocytes [J]. J Cell Physiol. 2018;233(5):3784–3793.