Thyroid hormone and cardiac disease: from basic concepts to clinical application

Iordanis Mourouzis, Francesca Forini, Constantinos Pantos, Giorgio Iervasi, Iordanis Mourouzis, Francesca Forini, Constantinos Pantos, Giorgio Iervasi

Abstract

Nature's models of regeneration provide substantial evidence that a natural healing process may exist in the heart. Analogies existing between the damaged myocardium and the developing heart strongly indicate that regulatory factors which drive embryonic heart development may also control aspects of heart regeneration. In this context, thyroid hormone (TH) which is critical in heart maturation during development appears to have a reparative role in adult life. Thus, changes in TH -thyroid hormone receptor (TR) homeostasis are shown to govern the return of the damaged myocardium to the fetal phenotype. Accordingly, thyroid hormone treatment preferentially rebuilds the injured myocardium by reactivating developmental gene programming. Clinical data provide further support to this experimental evidence and changes in TH levels and in particular a reduction of biologically active triiodothyronine (T3) in plasma after myocardial infarction or during evolution of heart failure, are strongly correlated with patients morbidity and mortality. The potential of TH to regenerate a diseased heart has now been testing in patients with acute myocardial infarction in a phase II, randomized, double blind, placebo-controlled study (the THiRST study).

Figures

Figure 1
Figure 1
Schematic of the sequence of biological events occurring in response to environmental stimuli and lead to tissue restoration. In the case of intense and sustained stressful stimuli redifferentiation “deficit” occurs and results in a disease state (e.g., heart failure, cancer, etc.). Exogenous TH can enhance redifferentiation and restore damage.
Figure 2
Figure 2
Cardiac cells are de-differentiated upon exposure to progrowth stimuli such as phenylephrine (PE). This response is mediated via ERK/TRα1 and requires an intact mTOR signaling. Inhibition of mTOR signaling with rapamycin not only abolishes PE-induced nuclear TRα1 overexpression but results in marked TRα1 decrease with cell atrophy. De-differentiated cells retain the ability to re-differentiate when T3 is added to the medium. TRα1 by its dual action (liganded versus unliganded) seems to act as a regulator of the cell dedifferentiation/redifferentiation process.
Figure 3
Figure 3
Schematic of molecular, structural, and functional changes during post-ischaemic cardiac remodeling in untreated (a) and TH-treated hearts (b). TH shortly after myocardial infarction induces favorable changes in left ventricular chamber remodeling in a time-dependent manner; TH treatment accelerates the development of cardiac hypertrophy which normalizes wall tension and reshapes left ventricular chamber towards a more ellipsoidal shape at later stages [20].
Figure 4
Figure 4
Structural and phenotypic effects of T3 supplementation. After 10 days of culture, T3-supplemented myocardial fragments presented better preserved cardiomyocytes both at histological (a) and immunohistochemical examination (c). Weaker staining and progressive atrophy was observed in the corresponding untreated samples ((b) and (d), resp.). Myocardial sections were stained hematoxylin eosin (a), (b) for total protein content evaluation or immunostained for α-sarcomeric actinin (c), (d) for sarcomeric specif protein content. Images were acquired with a CCD camera and optical density was measured (scale bar = 50 μm). (e) Quantification of the histological results obtained from hematoxylin-eosin and α-sarcomeric-actinin staining of 10-day-old human myocardium in culture. P was determined using the two-tailed unpaired Student's t-test; data are expressed as mean ± SEM (experimental data derived from [28]).

References

    1. Taegtmeyer H, Sen S, Vela D. Return to the fetal gene program: a suggested metabolic link to gene expression in the heart. Annals of the New York Academy of Sciences. 2010;1188:191–198.
    1. Pantos C, Mourouzis I, Cokkinos DV. New insights into the role of thyroid hormone in cardiac remodeling: time to reconsider? Heart Failure Reviews. 2011;16(1):79–96.
    1. Lee Y-K, Ng K-M, Chan Y-C, et al. Triiodothyronine promotes cardiac differentiation and maturation of embryonic stem cells via the classical genomic pathway. Molecular Endocrinology. 2010;24(9):1728–1736.
    1. Swynghedauw B. Molecular mechanisms of myocardial remodeling. Physiological Reviews. 1999;79(1):215–262.
    1. Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: a strong hypothesis. Heart Failure Reviews. 2007;12(3-4):331–343.
    1. Pantos C, Mourouzis I, Dimopoulos A, et al. Enhanced tolerance of the rat myocardium to ischemia and reperfusion injury earlyafter acute myocardial infarction. Basic Research in Cardiology. 2007;102(4):327–333.
    1. Pantos C, Mourouzis I, Saranteas T, et al. Thyroid hormone receptors α1 and β1 are downregulated in the post-infarcted rat heart: consequences on the response to ischaemia-reperfusion. Basic Research in Cardiology. 2005;100(5):422–432.
    1. Odelberg SJ. Inducing cellular dedifferentiation: a potential method for enhancing endogenous regeneration in mammals. Seminars in Cell and Developmental Biology. 2002;13(5):335–343.
    1. Sánchez Alvarado A. Regeneration in the metazoans: why does it happen? BioEssays. 2000;22(6):578–590.
    1. Porrello ER, Mahmoud AI, Simpson E, et al. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331(6020):1078–1080.
    1. Oviedo NJ, Beane WS. Regeneration: the origin of cancer or a possible cure? Seminars in Cell and Developmental Biology. 2009;20(5):557–564.
    1. Gudernatsch JF. Feeding experiments on tadpoles—I: the influence of specific organs given as food on growth and differentiation. A contribution to the knowledge of organs with internal secretion. Development Genes and Evolution. 1912;35(3):457–483.
    1. Paris M, Brunet F, Markov GV, Schubert M, Laudet V. The amphioxus genome enlightens the evolution of the thyroid hormone signaling pathway. Development Genes and Evolution. 2008;218(11-12):667–680.
    1. Slack JMW, Lin G, Chen Y. Molecular and cellular basis of regeneration and tissue repair: the Xenopus tadpole: a new model for regeneration research. Cellular and Molecular Life Sciences. 2008;65(1):54–63.
    1. Chassande O. Do unliganded thyroid hormone receptors have physiological functions? Journal of Molecular Endocrinology. 2003;31(1):9–20.
    1. Sato Y, Buchholz DR, Paul BD, Shi YB. A role of unliganded thyroid hormone receptor in postembryonic development in Xenopus laevis. Mechanisms of Development. 2007;124(6):476–488.
    1. Furlow JD, Neff ES. A developmental switch induced by thyroid hormone: Xenopus laevis metamorphosis. Trends in Endocrinology and Metabolism. 2006;17(2):40–47.
    1. Rolfe M, McLeod LE, Pratt PF, Proud CG. Activation of protein synthesis in cardiomyocytes by the hypertrophic agent phenylephrine requires the activation of ERK and involves phosphorylation of tuberous sclerosis complex 2 (TSC2) Biochemical Journal. 2005;388(3):973–984.
    1. Pantos C, Xinaris C, Mourouzis I, et al. Thyroid hormone receptor α1: a switch to cardiac cell “metamorphosis”? Journal of Physiology and Pharmacology. 2008;59(2):253–269.
    1. Pantos C, Mourouzis I, Galanopoulos G, et al. Thyroid hormone receptor 1 downregulation in postischemic heart failure progression: the potential role of tissue hypothyroidism. Hormone and Metabolic Research. 2010;42(10):718–724.
    1. Kinugawa K, Jeong MY, Bristow MR, Long CS. Thyroid hormone induces cardiac myocyte hypertrophy in a thyroid hormone receptor α1-specific manner that requires TAK1 and p38 mitogen-activated protein kinase. Molecular Endocrinology. 2005;19(6):1618–1628.
    1. Tavi P, Sjögren M, Lunde PK, et al. Impaired Ca2+ handling and contraction in cardiomyocytes from mice with a dominant negative thyroid hormone receptor α. Journal of Molecular and Cellular Cardiology. 2005;38(4):655–663.
    1. Pantos C, Mourouzis I, Cokkinos DV. Thyroid hormone as a therapeutic option for treating ischaemic heart disease: from early reperfusion to late remodelling. Vascular Pharmacology. 2010;52(3-4):157–165.
    1. Henderson KK, Danzi S, Paul JT, Leya G, Klein I, Samarel AM. Physiological replacement of T3 improves left ventricular function in an animal model of myocardial infarction-induced congestive heart failure. Circulation: Heart Failure. 2009;2(3):243–252.
    1. Chen YF, Kobayashi S, Chen J, et al. Short term triiodo-L-thyronine treatment inhibits cardiac myocyte apoptosis in border area after myocardial infarction in rats. Journal of Molecular and Cellular Cardiology. 2008;44(1):180–187.
    1. Pantos C, Mourouzis I, Markakis K, et al. Thyroid hormone attenuates cardiac remodeling and improves hemodynamics early after acute myocardial infarction in rats. European Journal of Cardio-thoracic Surgery. 2007;32(2):333–339.
    1. Pantos C, Mourouzis I, Markakis K, Tsagoulis N, Panagiotou M, Cokkinos DV. Long-term thyroid hormone administration reshapes left ventricular chamber and improves cardiac function after myocardial infarction in rats. Basic Research in Cardiology. 2008;103(4):308–318.
    1. Forini F, Lionetti V, Ardehali H, et al. Early long-term L-T3 replacement rescues mitochondria and prevents ischemic cardiac remodelling in rats. Journal of Cellular and Molecular Medicine. 2011;15(3):514–524.
    1. Kalofoutis C, Mourouzis I, Galanopoulos G, et al. Thyroid hormone can favorably remodel the diabetic myocardium after acute myocardial infarction. Molecular and Cellular Biochemistry. 2010;345(1-2):161–169.
    1. Pantos C, Mourouzis I, Tsagoulis N, et al. Thyroid hormone at supra-physiological dose optimizes cardiac geometry and improves cardiac function in rats with old myocardial infarction. Journal of Physiology and Pharmacology. 2009;60(3):49–56.
    1. Linnane AW, Eastwood H. Cellular redox regulation and prooxidant signaling systems: a new perspective on the free radical theory of aging. Annals of the New York Academy of Sciences. 2006;1067(1):47–55.
    1. Araujo ASR, Schenkel P, Enzveiler AT, et al. The role of redox signaling in cardiac hypertrophy induced by experimental hyperthyroidism. Journal of Molecular Endocrinology. 2008;41(5-6):423–430.
    1. Pantos C, Xinaris Ch, Mourouzis I, Malliopoulou V, Kardami E, Cokkinos DV. Thyroid hormone changes cardiomyocyte shape and geometry via ERK signaling pathway: potential therapeutic implications in reversing cardiac remodeling? Molecular and Cellular Biochemistry. 2007;297(1-2):65–72.
    1. Pantos C, Malliopoulou V, Mourouzis I, et al. Thyroxine pretreatment increases basal myocardial heat-shock protein 27 expression and accelerates translocation and phosphorylation of this protein upon ischaemia. European Journal of Pharmacology. 2003;478(1):53–60.
    1. Pantos C, Malliopoulou V, Mourouzis I, et al. Hyperthyroid hearts display a phenotype of cardioprotection against ischemic stress: a possible involvement of heat shock protein 70. Hormone and Metabolic Research. 2006;38(5):308–313.
    1. Pantos CI, Malliopoulou VA, Mourouzis IS, et al. Long-term thyroxine administration protects the heart in a pattern similar to ischemic preconditioning. Thyroid. 2002;12(4):325–329.
    1. Shulga A, Blaesse A, Kysenius K, et al. Thyroxin regulates BDNF expression to promote survival of injured neurons. Molecular and Cellular Neuroscience. 2009;42(4):408–418.
    1. Kahaly GJ, Dillmann WH. Thyroid hormone action in the heart. Endocrine Reviews. 2005;26(5):704–728.
    1. Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocrine Reviews. 2008;29(1):76–131.
    1. Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system. The New England Journal of Medicine. 2001;344(7):501–509.
    1. Gomberg-Maitland M, Frishman WH. Thyroid hormone and cardiovascular disease. American Heart Journal. 1998;135(2):187–196.
    1. Hamilton MA, Stevenson LW, Luu M, Walden JA. Altered thyroid hormone metabolism in advanced heart failure. Journal of the American College of Cardiology. 1990;16(1):91–95.
    1. Opasich C, Pacini F, Ambrosino N, et al. Sick euthyroid syndrome in patients with moderate-to-severe chronic heart failure. European Heart Journal. 1996;17(12):1860–1866.
    1. Pingitore A, Landi P, Taddei MC, Ripoli A, L’Abbate A, Iervasi G. Triiodothyronine levels for risk stratification of patients with chronic heart failure. American Journal of Medicine. 2005;118(2):132–136.
    1. Ascheim DD, Hryniewicz K. Thyroid hormone metabolism in patients with congestive heart failure: the low triiodothyronine state. Thyroid. 2002;12(6):511–515.
    1. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the “euthyroid sick syndrome”. Endocrine Reviews. 1982;3(2):164–217.
    1. Utiger RD. Altered thyroid function in nonthyroidal illness and surgery. To treat or not to treat? The New England Journal of Medicine. 1995;333(23):1562–1563.
    1. Forini F, Paolicchi A, Pizzorusso T, et al. 3,5,3′-triiodothyronine deprivation affects phenotype and intracellular [Ca2+]i of human cardiomyocytes in culture. Cardiovascular Research. 2001;51(2):322–330.
    1. Friberg L, Werner S, Eggertsen G, Ahnve S. Rapid down-regulation of thyroid hormones in acute myocardial infarction: is it cardioprotective in patients with angina? Archives of Internal Medicine. 2002;162(12):1388–1394.
    1. Galli E, Pingitore A, Iervasi G. The role of thyroid hormone in the pathophysiology of heart failure: clinical evidence. Heart Failure Reviews. 2010;15(2):155–169.
    1. Ceremuzyński L, Górecki A, Czerwosz L, et al. Low serum triiodothyronine in acute myocardial infarction indicates major heart injury. Kardiologia Polska. 2004;60(5):468–480.
    1. Kozdag G, Ural D, Vural A, et al. Relation between free triiodothyronine/free thyroxine ratio, echocardiographic parameters and mortality in dilated cardiomyopathy. European Journal of Heart Failure. 2005;7(1):113–118.
    1. Iervasi G, Pingitore A, Landi P, et al. Low-T3 syndrome: a strong prognostic predictor of death in patients with heart disease. Circulation. 2003;107(5):708–713.
    1. Iervasi G, Molinaro S, Landi P, et al. Association between increased mortality and mild thyroid dysfunction in cardiac patients. Archives of Internal Medicine. 2007;167(14):1526–1532.
    1. Ladenson PW, Sherman SI, Baughman KL, Ray PE, Feldman AM. Reversible alterations in myocardial gene expression in a young man with dilated cardiomyopathy and hypothyroidism. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(12):5251–5255.
    1. Colucci WS. Molecular and cellular mechanisms of myocardial failure. The American Journal of Cardiology. 1997;80(11A):15L–25L.
    1. Khalife WI, Tang YID, Kuzman JA, et al. Treatment of subclinical hypothyroidism reverses ischemia and prevents myocyte loss and progressive LV dysfunction in hamsters with dilated cardiomyopathy. American Journal of Physiology. 2005;289(6):H2409–H2415.
    1. Tribulova N, Knezl V, Shainberg A, Seki S, Soukup T. Thyroid hormones and cardiac arrhythmias. Vascular Pharmacology. 2010;52(3-4):102–112.
    1. Klein I, Danzi S. Thyroid disease and the heart. Circulation. 2007;116(15):1725–1735.
    1. Kahaly GJ, Kampmann C, Mohr-Kahaly S. Cardiovascular hemodynamics and exercise tolerance in thyroid disease. Thyroid. 2002;12(6):473–481.
    1. Biondi B, Palmieri EA, Fazio S, et al. Endogenous subclinical hyperthyroidism affects quality of life and cardiac morphology and function in young and middle-aged patients. Journal of Clinical Endocrinology and Metabolism. 2000;85(12):4701–4705.
    1. Kahaly GJ, Nieswandt J, Wagner S, Schlegel J, Mohr-Kahaly S, Hommel G. Ineffective cardiorespiratory function in hyperthyroidism. Journal of Clinical Endocrinology and Metabolism. 1998;83(11):4075–4078.
    1. Hu LW, Benvenuti LA, Liberti EA, Carneiro-Ramos MS, Barreto-Chaves MLM. Thyroxine-induced cardiac hypertrophy: influence of adrenergic nervous system versus renin-angiotensin system on myocyte remodeling. American Journal of Physiology. 2003;285(6):R1473–R1480.
    1. Barry SP, Davidson SM, Townsend PA. Molecular regulation of cardiac hypertrophy. International Journal of Biochemistry and Cell Biology. 2008;40(10):2023–2039.
    1. Kaminski G, Michalkiewicz D, Makowski K, et al. Prospective echocardiographic evaluation of patients with endogenous subclinical hyperthyroidism and after restoring euthyroidism. Clinical Endocrinology. 2011;74(4):501–507.
    1. Abdulrahman RM, Delgado V, Ng ACT, et al. Abnormal cardiac contractility in long-term exogenous subclinical hyperthyroid patients as demonstrated by two-dimensional echocardiography speckle tracking imaging. European Journal of Endocrinology. 2010;163(3):435–441.
    1. Biondi B. Invited commentary: cardiovascular mortality in subclinical hyperthyroidism: an ongoing dilemma. European Journal of Endocrinology. 2010;162(3):587–589.
    1. Vadiveloo T, Donnan PT, Cochrane L, Leese GP. The thyroid epidemiology, audit, and research study (TEARS): the natural history of endogenous subclinical hyperthyroidism. Journal of Clinical Endocrinology and Metabolism. 2011;96(1):E1–E8.
    1. Parle JV, Maisonneuve P, Sheppard MC, Boyle P, Franklyn JA. Prediction of all-cause and cardiovascular mortality in elderly people from one low serum thyrotropin result: a 10-year cohort study. The Lancet. 2001;358(9285):861–865.
    1. Osman F, Gammage MD, Franklyn JA. Hyperthyroidism and cardiovascular morbidity and mortality. Thyroid. 2002;12(6):483–487.
    1. The Coronary Drug Project Research Group. The coronary drug project. Findings leading to further modifications of its protocol with respect to dextrothyroxine. Journal of the American Medical Association. 1972;220(7):996–1008.
    1. Young WF, Jr., Gorman CA, Jiang NS, Machacek D, Hay ID. L-thyroxine contamination of pharmaceutical D-thyroxine: probable cause of therapeutic effect. Clinical Pharmacology and Therapeutics. 1984;36(6):781–787.
    1. Portman MA, Slee A, Olson AK, et al. Triiodothyronine supplementation in infants and children undergoing cardiopulmonary bypass (TRICC): a multicenter placebo-controlled randomized trial: age analysis. Circulation. 2010;122(11, supplement 1):S224–S233.
    1. Klemperer JD, Klein I, Gomez M, et al. Thyroid hormone treatment after coronary-artery bypass surgery. The New England Journal of Medicine. 1995;333(23):1522–1527.
    1. Klemperer JD, Zelano J, Helm RE, et al. Triiodothyronine improves left ventricular function without oxygen wasting effects after global hypothermic ischemia. Journal of Thoracic and Cardiovascular Surgery. 1995;109(3):457–465.
    1. Pingitore A, Galli E, Barison A, et al. Acute effects of triiodothyronine (T3) replacement therapy in patients with chronic heart failure and low-T3 syndrome: a randomized, placebo-controlled study. Journal of Clinical Endocrinology and Metabolism. 2008;93(4):1351–1358.
    1. Malik FS, Mehra MR, Uber PA, Park MH, Scott RL, van Meter CH. Intravenous thyroid hormone supplementation in heart failure with cardiogenic shock. Journal of Cardiac Failure. 1999;5(1):31–37.
    1. Hamilton MA, Stevenson LW, Fonarow GC, et al. Safety and hemodynamic effects of intravenous triiodothyronine in advanced congestive heart failure. The American Journal of Cardiology. 1998;81(4):443–447.
    1. Moruzzi P, Doria E, Agostoni PG. Medium-term effectiveness of L-thyroxine treatment in idiopathic dilated cardiomyopathy. American Journal of Medicine. 1996;101(5):461–467.
    1. Iervasi G, Emdin M, Colzani RMP, et al. Beneficial effects of long-term triiodothyronine (T3) infusion in patients with advanced heart failure and low T3 syndrome. In: Kimchi A, editor. Proceedings of the 2nd International Congress on Heart Disease—New Trends in Research, Diagnosis and Treatment. Washington, DC, USA: Medimond Medical Publications; 2001. pp. 549–553.
    1. Trivieri MG, Oudit GY, Sah R, et al. Cardiac-specific elevations in thyroid hormone enhance contractility and prevent pressure overload-induced cardiac dysfunction. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(15):6043–6048.
    1. Belke DD, Gloss B, Swanson EA, Dillmann WH. Adeno-associated virus-mediated expression of thyroid hormone receptor isoforms-α1 and -β1 improves contractile function in pressure overload-induced cardiac hypertrophy. Endocrinology. 2007;148(6):2870–2877.
    1. Moruzzi P, Doria E, Agostoni PG, Capacchione V, Sganzerla P. Usefulness of L-thyroxine to improve cardiac and exercise performance in idiopathic dilated cardiomyopathy. The American Journal of Cardiology. 1994;73(5):374–378.
    1. Degens H, Gilde AJ, Lindhout M, Willemsen PHM, van der Vusse GJ, van Bilsen M. Functional and metabolic adaptation of the heart to prolonged thyroid hormone treatment. American Journal of Physiology. 2003;284(1):H108–H115.
    1. Boehm EA, Jones BE, Radda GK, Veech RL, Clarke K. Increased uncoupling proteins and decreased efficiency in palmitate-perfused hyperthyroid rat heart. American Journal of Physiology. 2001;280(3):H977–H983.
    1. Pennock GD, Raya TE, Bahl JJ, Goldman S, Morkin E. Cardiac effects of 3,5-diiodothyropropionic acid, a thyroid hormone analog with inotropic selectivity. Journal of Pharmacology and Experimental Therapeutics. 1992;263(1):163–169.
    1. Wang X, Zheng W, Christensen LP, Tomanek RJ. DITPA stimulates bFGF, VEGF, angiopoietin, and Tie-2 and facilitates coronary arteriolar growth. American Journal of Physiology. 2003;284(2):H613–H618.
    1. Morkin E, Pennock G, Spooner PH, Bahl JJ, Underhill Fox K, Goldman S. Pilot studies on the use of 3,5-diiodothyropropionic acid, a thyroid hormone analog, in the treatment of congestive heart failure. Cardiology. 2002;97(4):218–225.
    1. Goldman S, McCarren M, Morkin E, et al. DITPA (3,5-diiodothyropropionic acid), a thyroid hormone analog to treat heart failure: phase II trial veterans affairs cooperative study. Circulation. 2009;119(24):3093–3100.
    1. Pingitore A, Iervasi G, Gerdes MA. Letter by Pingitore et al regarding article, “DITPA (3,5-diiodothyropropionic acid), a thyroid hormone analog to treat heart failure: phase II trial Veterans Affairs cooperative study“. Circulation. 2010;121(10, e240)
    1. Mizuma H, Murakami M, Mori M. Thyroid hormone activation in human vascular smooth muscle cells: expression of type II iodothyronine deiodinase. Circulation Research. 2001;88(3):313–318.
    1. Colantuoni A, Marchiafava PL, Lapi D, Forini FS, Iervasi G. Effects of tetraiodothyronine and triiodothyronine on hamster cheek pouch microcirculation. American Journal of Physiology. 2005;288(4):H1931–H1936.
    1. Gerdes AM, Iervasi G. Thyroid replacement therapy and heart failure. Circulation. 2010;122(4):385–393.
    1. Ezekowitz JA, Kaul P, Bakal JA, Armstrong PW, Welsh RC, McAlister FA. Declining in-hospital mortality and increasing heart failure incidence in elderly patients with first myocardial infarction. Journal of the American College of Cardiology. 2009;53(1):13–20.
    1. Khumri TM, Reid KJ, Kosiborod M, Spertus JA, Main ML. Usefulness of left ventricular diastolic dysfunction as a predictor of one-year rehospitalization in survivors of acute myocardial infarction. The American Journal of Cardiology. 2009;103(1):17–21.

Source: PubMed

3
Suscribir