Research article Special Issues

HB-EGF and ADAM 12S directed cellular reprogramming results in metabolically active brown adipose tissue-like cells

  • Brown adipose tissue (BAT) is considered a potential tool for the treatment of obesity and type 2 diabetes due to its ability to uncouple oxidative phosphorylation and stimulate non-shivering thermogenesis that utilizes glucose and lipids as its source of energy. Previous results from our lab demonstrated that co-expression of HB-EGF and ADAM 12S resulted in lipid accumulation and a BAT-like phenotype, including up-regulation of BAT genes, down-regulation of genes involved in formation of white adipose tissues, and increased mitochondrial staining in three cell lines including mouse fibroblasts, human epidermoid carcinoma cells, and human preadipocytes. Furthermore, qRT-PCR results demonstrated up-regulation of cellular reprogramming factors such as KLF4, KLF3, and FGF-2 and down-regulation of LMNA, a marker gene involved in differentiation, in the BAT-like reprogrammed cells. This study substantiates these finding using immunohistochemical analysis of reprogrammed BAT-like cells that demonstrate increased immunofluorescent detection of FGF-2, KLF3, and PGC-1α and decreased immunofluorescence for C/EBPα. Supportive evidence of cellular reprogramming involves the use of a stem-cell transcription factor RT-profiler array that results in enhanced expression of HOXA10 (3.04-fold) and HOXC5 (6.46-fold). In order to demonstrate that HB-EGF/ADAM 12S reprogrammed BAT-like cells function as BAT, oxygen consumption and extracellular acidification rates were measured using a Seahorse XFe24 Analyzer with and without catecholamine exposure followed by FCCP + Oligomycin exposure. HB-EGF/ADAM 12S reprogrammed BAT-like cells demonstrate a significant metabolic increase compared to MLC, HB-EGF, ADAM 12S. HB-EGF/ADAM12S reprogrammed BAT-like cells exhibit a metabolic profile similar to 3T3-L1 induced BAT cells. Collectively, these results demonstrate that HB-EGF/ADAM 12S co-expression stimulates cellular reprogramming into metabolically active BAT and may be a putative therapeutic tool to combat obesity and type 2 diabetes.

    Citation: Sean R. Taylor, Carly A. Gemma, Kayla M. Cartwright, Daniel C. Pfeil, Evan R. Miller, Charles E. Long, Paul A. Harding. HB-EGF and ADAM 12S directed cellular reprogramming results in metabolically active brown adipose tissue-like cells[J]. AIMS Cell and Tissue Engineering, 2018, 2(4): 203-219. doi: 10.3934/celltissue.2018.4.203

    Related Papers:

  • Brown adipose tissue (BAT) is considered a potential tool for the treatment of obesity and type 2 diabetes due to its ability to uncouple oxidative phosphorylation and stimulate non-shivering thermogenesis that utilizes glucose and lipids as its source of energy. Previous results from our lab demonstrated that co-expression of HB-EGF and ADAM 12S resulted in lipid accumulation and a BAT-like phenotype, including up-regulation of BAT genes, down-regulation of genes involved in formation of white adipose tissues, and increased mitochondrial staining in three cell lines including mouse fibroblasts, human epidermoid carcinoma cells, and human preadipocytes. Furthermore, qRT-PCR results demonstrated up-regulation of cellular reprogramming factors such as KLF4, KLF3, and FGF-2 and down-regulation of LMNA, a marker gene involved in differentiation, in the BAT-like reprogrammed cells. This study substantiates these finding using immunohistochemical analysis of reprogrammed BAT-like cells that demonstrate increased immunofluorescent detection of FGF-2, KLF3, and PGC-1α and decreased immunofluorescence for C/EBPα. Supportive evidence of cellular reprogramming involves the use of a stem-cell transcription factor RT-profiler array that results in enhanced expression of HOXA10 (3.04-fold) and HOXC5 (6.46-fold). In order to demonstrate that HB-EGF/ADAM 12S reprogrammed BAT-like cells function as BAT, oxygen consumption and extracellular acidification rates were measured using a Seahorse XFe24 Analyzer with and without catecholamine exposure followed by FCCP + Oligomycin exposure. HB-EGF/ADAM 12S reprogrammed BAT-like cells demonstrate a significant metabolic increase compared to MLC, HB-EGF, ADAM 12S. HB-EGF/ADAM12S reprogrammed BAT-like cells exhibit a metabolic profile similar to 3T3-L1 induced BAT cells. Collectively, these results demonstrate that HB-EGF/ADAM 12S co-expression stimulates cellular reprogramming into metabolically active BAT and may be a putative therapeutic tool to combat obesity and type 2 diabetes.


    加载中
    [1] Giordano A, Smorlesi A, Frontini A, et al. (2014) White, brown and pink adipocytes: the extraordinary plasticity of the adipose organ. Eur J Endocrinol 170: R159–171. doi: 10.1530/EJE-13-0945
    [2] Bauters D, Cobbaut M, Geys L, et al. (2017) Loss of ADAMTS5 enhances brown adipose tissue mass and promotes browning of white adipose tissue via CREB signaling. Mol Metab 6: 715–724. doi: 10.1016/j.molmet.2017.05.004
    [3] Cristancho AG, Lazar MA (2011) Forming functional fat: a growing understanding of adipocyte differentiation. Nat Rev Mol Cell Biol 12: 722–734. doi: 10.1038/nrm3198
    [4] Porter RK (2006) A new look at UCP 1. Biochim Biophys Acta 1757: 446–448. doi: 10.1016/j.bbabio.2006.04.012
    [5] Nedergaard J, Golozoubova V, Matthias A, et al. (2001) UCP1: the only protein able to mediate adaptive non-shivering thermogenesis and metabolic inefficiency. Biochim Biophys Acta 1504: 82–106. doi: 10.1016/S0005-2728(00)00247-4
    [6] Ghorbani M, Himms-Hagen J (1997) Appearance of brown adipocytes in white adipose tissue during CL 316,243-induced reversal of obesity and diabetes in Zucker fa/fa rats. Int J Obes Relat Metab Disord 21: 465–475. doi: 10.1038/sj.ijo.0800432
    [7] Zhou Z, Darwal MA, Cheng EA, et al. (2013) Cellular reprogramming into a brown adipose tissue-like phenotype by co-expression of HB-EGF and ADAM 12S. Growth Factors 31: 185–198. doi: 10.3109/08977194.2013.840297
    [8] Taylor SR, Klements JR, Markesbery MG, et al. (2014) Cellular transdifferentiation into brown adipose-like cells by adenoviral-directed expression or stable transfection of HB-EGF and ADAM 12S. J Cell Mol Biol 12: 55–62.
    [9] Christiaens V, Scroyen I, Lijnen HR (2008) Role of proteolysis in development of murine adipose tissue. Thromb Haemost 99: 290–294.
    [10] Sun K, Kusminski CM, Scherer PE (2011) Adipose tissue remodeling and obesity. J Clin Invest 121: 2094–2101. doi: 10.1172/JCI45887
    [11] Odaka M, Kohda D, Lax I, et al. (1997) Ligand-binding enhances the affinity of dimerization of the extracellular domain of the epidermal growth factor receptor. J Biochem 122: 116–121. doi: 10.1093/oxfordjournals.jbchem.a021718
    [12] Suzuki M, Raab G, Moses MA, et al. (1997) Matrix metalloproteinase-3 releases active heparin-binding epidermal growth factor by cleavage at a specific juxtamembrane site. J Biol Chem 272: 31730–31737. doi: 10.1074/jbc.272.50.31730
    [13] Asakura M, Kitakaze M, Takashima S, et al. (2002) Cardiac hyper-trophy is inhibited by antagonism of ADAM12 processing of HB-EGF: metalloproteinase inhibitors as a new therapy. Nat Med 8: 35–40. doi: 10.1038/nm0102-35
    [14] Nanba D, Mammoto A, Hashimoto K, et al. (2003) Proteolytic release of the carboxy-terminal fragment of pro-HB-EGF causes nuclear export of PLZF. J Cell Biol 163: 489–502.
    [15] Higashiyama S, Nanba D (2005) ADAM-mediated ectodomain shedding of HB-EGF in receptor cross-talk. Biochim Biophys Acta 1751: 110–117. doi: 10.1016/j.bbapap.2004.11.009
    [16] Kurisaki T, Masuda A, Sudo K, et al. (2003) Phenotypic analysis of Meltrin alpha (ADAM12)-deficient mice: involvement of Meltrin alpha in adipogenesis and myogenesis. Mol Cell Biol 23: 55–61. doi: 10.1128/MCB.23.1.55-61.2003
    [17] Taylor SR, Markesbery MG, Harding PA (2014) Heparin-binding epidermal growth factor-like growth factor (HB-EGF) and proteolytic processing by a disintegrin and metalloproteinases (ADAMs): a regulator of several pathways. Semin Cell Dev Biol 28: 22–30. doi: 10.1016/j.semcdb.2014.03.004
    [18] Asano H, Kanamori Y, Higurashi S, et al. (2014) Induction of beige-like adipocytes in 3T3-L1 cells. J Vet Med Sci 76: 57–64. doi: 10.1292/jvms.13-0359
    [19] Lowell BB, Spiegelman BM (2000) Towards a molecular understanding of adaptive thermogenesis. Nat 404: 652–660. doi: 10.1038/35007527
    [20] Vallier L, Touboul T, Brown S, et al. (2009) Signaling pathways controlling pluripotency and early cell fate decisions of human induced pluripotent stem cells. Stem Cells 27: 2655–2666. doi: 10.1002/stem.199
    [21] Bell-Anderson KS, Funnell AP, Williams H, et al. (2013) Loss of Krüppel-like factor 3 (KLF3/BKLF) leads to upregulation of the insulin-sensitizing factor adipolin (FAM132A/CTRP12/C1qdc2). Diabetes 62: 2728–2737. doi: 10.2337/db12-1745
    [22] Sue N, Jack BH, Eaton SA, et al. (2008) Targeted disruption of the basic Krüppel-like Factor Gene (Klf3) reveals a role in adipogenesis. Mol Cell Bio 28: 3967–3978. doi: 10.1128/MCB.01942-07
    [23] Linhart HG, Ishimura-Oka K, DeMayo F, et al. (2001) C/EBP alpha is required for differentiation of white, but not brown, adipose tissue. Proc Natl Acad Sci U S A 98: 12532–12537. doi: 10.1073/pnas.211416898
    [24] Shimizu Y, Shimazu T (2002) Thyroid hormone augments GLUT4 expression and insulin-sensitive glucose transport system in differentiating rat brown adipocytes in culture. J Vet Med Sci 64:677–681. doi: 10.1292/jvms.64.677
    [25] Myers P (2008) Hox genes in development: The Hox code. Nat Education 1: 1–4.
    [26] Cesar AS, Regitano LC, Koltes JE, et al. (2015) Putative regulatory factors associated with intramuscular fat content. PLoS One 10: e0128350. doi: 10.1371/journal.pone.0128350
    [27] Kawaguchi N, Xu X, Tajima R, et al. (2002) ADAM 12 protease induces adipogenesis in transgenic mice. Am J Pathol 160: 1895–1903. doi: 10.1016/S0002-9440(10)61136-4
    [28] Singh S, Rajput YS, Barui AK, et al. (2016) Fat accumulation in differentiated brown adipocytes is linked with expression of Hox genes. Gene Expr Patterns 20: 99–105. doi: 10.1016/j.gep.2016.01.002
    [29] Isidor MS, Winther S, Basse AL, et al. (2015) An siRNA-based method for efficient silencing of gene expression in mature brown adipocytes. Adipocyte 5: 175–185.
    [30] Caron A, Labbé SM, Carter S, et al. (2017) Loss of UCP2 impairs cold-induced non-shivering thermogenesis by promoting a shift toward glucose utilization in brown adipose tissue. Biochimie 134: 118–126. doi: 10.1016/j.biochi.2017.01.006
    [31] Pecqueur C, Bui T, Gelly C, et al. (2008) Uncoupling protein-2 controls proliferation by promoting fatty acid oxidation and limiting glycolysis-derived pyruvate utilization. FASEB J 22: 9–18. doi: 10.1096/fj.07-8945com
    [32] Pecqueur C, Alves-Guerra C, Ricquier D, et al. (2009) UCP2, a metabolic sensor coupling glucose oxidation to mitochondrial metabolism? IUBMB Life 61:762–767. doi: 10.1002/iub.188
    [33] Lee O, O'Brien PJ (2010) 1.19-Modifications of mitochondrial function by toxicants. Compr Toxicol 1:411–445.
    [34] Heytler PG, Prichard WW (1962) A new class of uncoupling agents-carbonyl cyanide phenylhydrazones. Biochem Biophys Res Commun 7: 272–275. doi: 10.1016/0006-291X(62)90189-4
    [35] Hyllienmark L, Brismar T (1996) Effect of metabolic inhibition on K+ channels in pyramidal cells of the hippocampal CA1 region in rat brain slices. J Physiol 496: 155–164. doi: 10.1113/jphysiol.1996.sp021673
    [36] Buckler KJ, Vaughan-Jones RD (1998) Effects of mitochondrial uncouplers on intracellular calcium, pH and membrane potential in rat carotid body type I cells. J Physiol 513:819–833. doi: 10.1111/j.1469-7793.1998.819ba.x
    [37] Park KS, Jo I, Pak K, et al. (2002) FCCP depolarizes plasma membrane potential by activating proton and Na+ currents in bovine aortic endothelial cells. Pflügers Arch 443: 344–352. doi: 10.1007/s004240100703
    [38] To MS, Aromataris EC, Castro J, et al. (2010) Mitochondrial uncoupler FCCP activates proton conductance but does not block store-operated Ca2+ current in liver cells. Arch Biochem Biophys 495: 152–158. doi: 10.1016/j.abb.2010.01.004
    [39] Prusiner SB, Cannon B, Ching TM et al. (1968) Oxidative metabolism in cells isolated from brown adipose tissue. 2. Catecholamine regulated respiratory control. Eur J Biochem 7: 51–57.
    [40] Reed N, Fain JN (1968) Stimulation of respiration in brown fat cells by epinephrine, dibutyryl-3',5'-adenosine monophosphate, and m-chloro (carbonyl cyanide) phenylhydrazone. J Biol Chem 243: 2843–2848.
    [41] Mohell N, Connolly E, Nedergaard J (1987) Distinction between mechanisms underlying alpha 1- and beta-adrenergic respiratory stimulation in brown fat cells. Am J Physiol 253:C301–308. doi: 10.1152/ajpcell.1987.253.2.C301
    [42] Flatmark T, Pedersen JI (1973) Studies on the energy state of isolated brown adipose tissue mitochondria. Effect of adenine nucleotides and oligomycin on the generation and dissipation of the "energy potential". Biochim Biophys Acta 292: 64–72.
    [43] Oropeza D, Jouvet N, Bouyakdan K, et al. (2015) PGC-1 coactivators in β-cells regulate lipid metabolism and are essential for insulin secretion coupled to fatty acids. Mol Metab 4: 811–822. doi: 10.1016/j.molmet.2015.08.001
    [44] Dlasková A, Clarke KJ, Porter RK (2010) The role of UCP 1 in production of reactive oxygen species by mitochondria isolated from brown adipose tissue. Biochim Biophys Acta 1797: 1470–1476. doi: 10.1016/j.bbabio.2010.04.008
  • Reader Comments
  • © 2018 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4761) PDF downloads(1836) Cited by(1)

Article outline

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog