The Role of Thyrotropin Receptor Activation in Adipogenesis and Modulation of Fat Phenotype

Mohd Shazli Draman, Michael Stechman, David Scott-Coombes, Colin M Dayan, Dafydd Aled Rees, Marian Ludgate, Lei Zhang, Mohd Shazli Draman, Michael Stechman, David Scott-Coombes, Colin M Dayan, Dafydd Aled Rees, Marian Ludgate, Lei Zhang

Abstract

Evidence from clinical and experimental data suggests that thyrotropin receptor (TSHR) signaling is involved in energy expenditure through its impact on white adipose tissue (WAT) and brown adipose tissue (BAT). TSHR expression increases during mesenchymal stem cell (MSC) differentiation into fat. We hypothesize that TSHR activation [TSHR*, elevated thyroid-stimulating hormone, thyroid-stimulating antibodies (TSAB), or activating mutation] influences MSC differentiation, which contributes to body composition changes seen in hypothyroidism or Graves' disease (GD). The role of TSHR activation on adipogenesis was first investigated using ex vivo samples. Neck fat (all euthyroid at surgery) was obtained from GD (n = 11, TSAB positive), toxic multinodular goiter (TMNG, TSAB negative) (n = 6), and control patients with benign euthyroid disease (n = 11, TSAB negative). The effect of TSHR activation was then analyzed using human primary abdominal subcutaneous preadipocytes (n = 16). Cells were cultured in complete medium (CM) or adipogenic medium [ADM, containing thiazolidinedione (TZD), PPARγ agonist, which is able to induce BAT formation] with or without TSHR activation (gain-of-function mutant) for 3 weeks. Adipogenesis was evaluated using oil red O (ORO), counting adipogenic foci, qPCR measurement of terminal differentiation marker (LPL). BAT [PGC-1α, uncoupling protein 1 (UCP1), and ZIC1], pre-BAT (PRDM16), BRITE- (CITED1), or WAT (LEPTIN) markers were analyzed by semiquantitative PCR or qPCR. In ex vivo analysis, there were no differences in the expression of UCP1, PGC-1α, and ZIC1. BRITE marker CITED1 levels were highest in GD followed by TMNG and control (p for trend = 0.009). This was associated with higher WAT marker LEPTIN level in GD than the other two groups (p < 0.001). In primary cell culture, TSHR activation substantially enhanced adipogenesis with 1.4 ± 0.07 (ORO), 8.6 ± 1.8 (foci), and 5.5 ± 1.6 (LPL) fold increases compared with controls. Surprisingly, TSHR activation in CM also significantly increased pre-BAT marker PRDM16; furthermore, TZD-ADM induced adipogenesis showed substantially increased BAT markers, PGC-1α and UCP1. Our study revealed that TSHR activation plays an important role in the adipogenesis process and BRITE/pre-BAT formation, which leads to WAT or BAT phenotype. It may contribute to weight loss as heat during hyperthyroidism and later transforms into WAT posttreatment of GD when patients gain excess weight.

Keywords: BRITE adipocytes; adipogenesis; body composition; brown adipose tissue; thyrotropin receptor; white adipose tissue.

Figures

Figure 1
Figure 1
Ex vivo analysis of deep neck fat. Samples were snap frozen, and total RNA was isolated. Gene transcripts were measured by qPCR, (A) thyrotropin receptor (TSHR); (B)ZIC1 (brown adipose tissue marker), shown as transcript copy number (TCN) per 1,000 copies of housekeeper gene APRT (adenosine phosphoribosyl transferase); standard PCR was used to analyze (C)LEPTIN (white adipose tissue marker), (D)CITED1 (BRITE marker), densitometry were measured and corrected to housekeeping gene (GAPDH). Representative photos were shown (gels with all samples had been included in Figure S1 in Supplementary Material). Post-ANOVA test for linear trend of CITED1 was performed (p = 0.009). Results expressed as mean ± SD of all samples studied (each performed in duplicate) (**p ≤ 0.01; ***p < 0.001).
Figure 2
Figure 2
Adipogenesis in subcutaneous preadipocytes was assessed by foci counting (n = 11, representative photos were shown with arrows indicating differentiating adipocytes), LPL transcripts measured by qPCR (n = 9) and oil red O (ORO, n = 4). Result presented as fold increase in TSHR* populations relative to empty vector controls. The table reports raw data for qPCR results expressed as transcript copy number per 1,000 copies of housekeeper gene APRT (adenosine phosphoribosyl transferase), together with foci numbers and ORO optical density values (mean ± SEM). Histograms = mean ± SEM of all samples studied (each performed in duplicate) (*p < 0.05; **p ≤ 0.01; ***p < 0.005).
Figure 3
Figure 3
Preadipocytes/fibroblasts from subcutaneous fat (n = 6) expressing activating thyrotropin receptor (TSHR) mutant L629F (TSHR*) and equivalent empty vector controls were cultured until confluent (day 0) before changing to differentiation medium for 22 days. Total RNA was isolated before or after adipogenesis. PRDM16 (relative expression ratio) (A), PGC-1α (B), and uncoupling protein 1 (UCP1) (C) transcripts were measured by qPCR. Results are expressed as comparative qPCR (relative ratio to APRT) of PRDM16 or absolute qPCR (PGC-1α and UCP1) of transcript copy number (TCN) per 1,000 copies of housekeeper gene APRT (adenosine phosphoribosyl transferase). Histograms = mean ± SEM of all samples studied (each performed in duplicate). Two-tailed t-test or Mann–Whitney test used for statistic analysis (*p < 0.03; **p = 0.01).

References

    1. Paschke R, Ludgate M. The thyrotropin receptor in thyroid diseases. N Engl J Med (1997) 337:1675–81.10.1056/NEJM199712043372307
    1. Seppel T, Kosel A, Schlaghecke R. Bioelectrical impedance assessment of body composition in thyroid disease. Eur J Endocrinol (1997) 136:493–8.10.1530/eje.0.1360493
    1. Lahesmaa M, Orava J, Schalin-Jantti C, Soinio M, Hannukainen JC, Noponen T, et al. Hyperthyroidism increases brown fat metabolism in humans. J Clin Endocrinol Metab (2014) 99:E28–35.10.1210/jc.2013-2312
    1. Obregon MJ. Adipose tissues and thyroid hormones. Front Physiol (2014) 5:479.10.3389/fphys.2014.00479
    1. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med (2009) 360:1509–17.10.1056/NEJMoa0810780
    1. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med (2009) 360:1500–8.10.1056/NEJMoa0808718
    1. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional brown adipose tissue in healthy adults. N Engl J Med (2009) 360:1518–25.10.1056/NEJMoa0808949
    1. Tigas S, Idiculla J, Beckett G, Toft A. Is excessive weight gain after ablative treatment of hyperthyroidism due to inadequate thyroid hormone therapy? Thyroid (2000) 10:1107–11.10.1089/thy.2000.10.1107
    1. Jansson S, Berg G, Lindstedt G, Michanek A, Nystrom E. Overweight – a common problem among women treated for hyperthyroidism. Postgrad Med J (1993) 69:107–11.10.1136/pgmj.69.808.107
    1. Dale J, Daykin J, Holder R, Sheppard MC, Franklyn JA. Weight gain following treatment of hyperthyroidism. Clin Endocrinol (2001) 55:233–9.10.1046/j.1365-2265.2001.01329.x
    1. Pears J, Jung RT, Gunn A. Long-term weight changes in treated hyperthyroid and hypothyroid patients. Scott Med J (1990) 35:180–2.10.1177/003693309003500609
    1. Weinreb JT, Yang Y, Braunstein GD. Do patients gain weight after thyroidectomy for thyroid cancer? Thyroid (2011) 21:1339–42.10.1089/thy.2010.0393
    1. Laurberg P, Wallin G, Tallstedt L, Abraham-Nordling M, Lundell G, Torring O. TSH-receptor autoimmunity in Graves’ disease after therapy with anti-thyroid drugs, surgery, or radioiodine: a 5-year prospective randomized study. Eur J Endocrinol (2008) 158:69–75.10.1530/EJE-07-0450
    1. Marcus C, Ehren H, Bolme P, Arner P. Regulation of lipolysis during the neonatal period. Importance of thyrotropin. J Clin Invest (1988) 82:1793–7.10.1172/JCI113793
    1. Crisp MS, Lane C, Halliwell M, Wynford-Thomas D, Ludgate M. Thyrotropin receptor transcripts in human adipose tissue. J Clin Endocrinol Metab (1997) 82:2003–5.
    1. Zhang L, Baker G, Janus D, Paddon CA, Fuhrer D, Ludgate M. Biological effects of thyrotropin receptor activation on human orbital preadipocytes. Invest Ophthalmol Vis Sci (2006) 47:5197–203.10.1167/iovs.06-0596
    1. Abe E, Marians RC, Yu W, Wu XB, Ando T, Li Y, et al. TSH is a negative regulator of skeletal remodeling. Cell (2003) 115:151–62.10.1016/S0092-8674(03)00771-2
    1. Elgadi A, Zemack H, Marcus C, Norgren S. Tissue-specific knockout of TSHR in white adipose tissue increases adipocyte size and decreases TSH-induced lipolysis. Biochem Biophys Res Commun (2010) 393:526–30.10.1016/j.bbrc.2010.02.042
    1. Lu S, Guan Q, Liu Y, Wang H, Xu W, Li X, et al. Role of extrathyroidal TSHR expression in adipocyte differentiation and its association with obesity. Lipids Health Dis (2012) 11:17.10.1186/1476-511X-11-17
    1. Ma S, Jing F, Xu C, Zhou L, Song Y, Yu C, et al. Thyrotropin and obesity: increased adipose triglyceride content through glycerol-3-phosphate acyltransferase 3. Sci Rep (2015) 5:7633.10.1038/srep07633
    1. Muscogiuri G, Sorice GP, Mezza T, Prioletta A, Lassandro AP, Pirronti T, et al. High-normal TSH values in obesity: is it insulin resistance or adipose tissue’s guilt? Obesity (Silver Spring) (2013) 21:101–6.10.1002/oby.20240
    1. Endo T, Kobayashi T. Thyroid-stimulating hormone receptor in brown adipose tissue is involved in the regulation of thermogenesis. Am J Physiol Endocrinol Metab (2008) 295:E514–8.10.1152/ajpendo.90433.2008
    1. Endo T, Kobayashi T. Expression of functional TSH receptor in white adipose tissues of TSH receptor mutant mice induces lipolysis in vivo. Am J Physiol Endocrinol Metab (2012) 302:E1569–75.10.1152/ajpendo.00572.2011
    1. Martinez-deMena R, Anedda A, Cadenas S, Obregon MJ. TSH effects on thermogenesis in rat brown adipocytes. Mol Cell Endocrinol (2015) 404:151–8.10.1016/j.mce.2015.01.028
    1. de Lloyd A, Bursell J, Gregory JW, Rees DA, Ludgate M. TSH receptor activation and body composition. J Endocrinol (2010) 204:13–20.10.1677/JOE-09-0262
    1. Chondronikola M, Volpi E, Borsheim E, Porter C, Annamalai P, Enerback S, et al. Brown adipose tissue improves whole-body glucose homeostasis and insulin sensitivity in humans. Diabetes (2014) 63:4089–99.10.2337/db14-0746
    1. Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature (2008) 454:961–7.10.1038/nature07182
    1. Stephens M, Ludgate M, Rees DA. Brown fat and obesity: the next big thing? Clin Endocrinol (2011) 74:661–70.10.1111/j.1365-2265.2011.04018.x
    1. Wu J, Cohen P, Spiegelman BM. Adaptive thermogenesis in adipocytes: is beige the new brown? Genes Dev (2013) 27:234–50.10.1101/gad.211649.112
    1. Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell (2012) 150:366–76.10.1016/j.cell.2012.05.016
    1. Jespersen NZ, Larsen TJ, Peijs L, Daugaard S, Homoe P, Loft A, et al. A classical brown adipose tissue mRNA signature partly overlaps with brite in the supraclavicular region of adult humans. Cell Metab (2013) 17:798–805.10.1016/j.cmet.2013.04.011
    1. Cohen P, Levy JD, Zhang Y, Frontini A, Kolodin DP, Svensson KJ, et al. Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch. Cell (2014) 156:304–16.10.1016/j.cell.2013.12.021
    1. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell (2002) 13:4279–95.10.1091/mbc.E02-02-0105
    1. Starkey KJ, Janezic A, Jones G, Jordan N, Baker G, Ludgate M. Adipose thyrotrophin receptor expression is elevated in Graves’ and thyroid eye diseases ex vivo and indicates adipogenesis in progress in vivo. J Mol Endocrinol (2003) 30:369–80.10.1677/jme.0.0300369
    1. Evans C, Morgenthaler NG, Lee S, Llewellyn DH, Clifton-Bligh R, John R, et al. Development of a luminescent bioassay for thyroid stimulating antibodies. J Clin Endocrinol Metab (1999) 84:374–7.10.1210/jcem.84.1.5532
    1. Rice SP, Zhang L, Grennan-Jones F, Agarwal N, Lewis MD, Rees DA, et al. Dehydroepiandrosterone (DHEA) treatment in vitro inhibits adipogenesis in human omental but not subcutaneous adipose tissue. Mol Cell Endocrinol (2010) 320:51–7.10.1016/j.mce.2010.02.017
    1. Fuhrer D, Lewis MD, Alkhafaji F, Starkey K, Paschke R, Wynford-Thomas D, et al. Biological activity of activating thyroid-stimulating hormone receptor mutants depends on the cellular context. Endocrinology (2003) 144:4018–30.10.1210/en.2003-0438
    1. Ussar S, Lee KY, Dankel SN, Boucher J, Haering MF, Kleinridders A, et al. ASC-1, PAT2, and P2RX5 are cell surface markers for white, beige, and brown adipocytes. Sci Transl Med (2014) 6:247ra103.10.1126/scitranslmed.3008490
    1. Sharp LZ, Shinoda K, Ohno H, Scheel DW, Tomoda E, Ruiz L, et al. Human BAT possesses molecular signatures that resemble beige/brite cells. PLoS One (2012) 7:e49452.10.1371/journal.pone.0049452
    1. Ohno H, Shinoda K, Spiegelman BM, Kajimura S. PPARgamma agonists induce a white-to-brown fat conversion through stabilization of PRDM16 protein. Cell Metab (2012) 15:395–404.10.1016/j.cmet.2012.01.019
    1. Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass CA, et al. Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat Med (2013) 19:635–9.10.1038/nm.3112
    1. Nedergaard J, Cannon B. How brown is brown fat? It depends where you look. Nat Med (2013) 19:540–1.10.1038/nm.3187
    1. Rosenwald M, Perdikari A, Rulicke T, Wolfrum C. Bi-directional interconversion of brite and white adipocytes. Nat Cell Biol (2013) 15:659–67.10.1038/ncb2740
    1. Lee YH, Petkova AP, Mottillo EP, Granneman JG. In vivo identification of bipotential adipocyte progenitors recruited by beta3-adrenoceptor activation and high-fat feeding. Cell Metab (2012) 15:480–91.10.1016/j.cmet.2012.03.009
    1. Lee JY, Takahashi N, Yasubuchi M, Kim YI, Hashizaki H, Kim MJ, et al. Triiodothyronine induces UCP-1 expression and mitochondrial biogenesis in human adipocytes. Am J Physiol Cell Physiol (2012) 302:C463–72.10.1152/ajpcell.00010.2011
    1. Zhang L, Grennan-Jones F, Lane C, Rees DA, Dayan CM, Ludgate M. Adipose tissue depot-specific differences in the regulation of hyaluronan production of relevance to Graves’ orbitopathy. J Clin Endocrinol Metab (2012) 97:653–62.10.1210/jc.2011-1299
    1. Zhang L, Ji QH, Ruge F, Lane C, Morris D, Tee AR, et al. Reversal of pathological features of Graves’ orbitopathy by activation of forkhead transcription factors, FOXOs. J Clin Endocrinol Metab (2016) 101:114–22.10.1210/jc.2015-2932
    1. Peirce V, Carobbio S, Vidal-Puig A. The different shades of fat. Nature (2014) 510:76–83.10.1038/nature13477

Source: PubMed

3
Abonner