Skip to main content

β-Adrenergic Receptor Signaling in Heart Failure

  • Chapter
  • First Online:
Cardiac Remodeling

Part of the book series: Advances in Biochemistry in Health and Disease ((ABHD,volume 5))

Abstract

Acute activation of the sympathetic system and resultant β-adrenergic receptor (β-AR) signaling are required to maintain homeostasis, providing inotropic support in times of need, as in “fight or flight” or response to any stress, such as cardiac dysfunction and heart failure. For most of the twentieth century, it was reasoned that sympathetic stimulation of β-ARs through administration of naturally occurring catecholamines or synthetic sympathomimetic amines could provide inotropic support and should be used in heart failure therapy. However, in heart failure, sympathetic drive to the heart is excessively increased, and chronic sympathetic stimulation is deleterious, since it increases \( \text{M}\text{ {0.05em}}\stackrel{·}{\text{V}}\text{ {0.05em}}{\text{O}}_{2} \), which cannot be met by appropriate increases in coronary blood flow, thereby creating subendocardial ischemia and intensifying the cardiac dysfunction. Furthermore, continued stimulation of the β-ARs also becomes problematic because it can activate multiple cellular processes including those involved in pathological remodeling seen in the development of cardiomyopathy. However, this reasoning took a diametrically opposite turn in the latter twentieth century when the adverse effects of chronic β-AR stimulation became apparent from experimental studies in transgenic mice with cardiac-specific overexpression of G and β-ARs and also from clinical studies with poor outcomes for patients on chronic sympathomimetic amine therapy. At this time it was also found that internal compensatory physiological processes countering continued β-AR stimulation in the heart were cleverer than physicians. As a protective response, β-AR desensitize, which reduces the effectiveness of β-AR stimulation and the consequent increases in myocardial oxygen demands. Taken together, these factors were fundamental to the change in course from β-AR stimulation to β-AR blockade in the treatment of heart failure.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Cannon WB, Rosenbueth A (1933) Studies on conditions of activity in endocrine organs: XXIX. Sympathin E and sympathin I. Am J Physiol 104:557–574

    CAS  Google Scholar 

  2. Ahlquist RP (1948) A study of the adrenotropic receptors. Am J Physiol 153:586–600

    PubMed  CAS  Google Scholar 

  3. Dale HH (1906) On some physiological actions of ergot. J Physiol 34:163–206

    PubMed  Google Scholar 

  4. Dale H (1943) Modes of drug action. General introductory address. Trans Faraday Soc 39:319b–322b

    Google Scholar 

  5. De Jongh DK (1964) Some introductory remarks on the conception of receptors. In: Ariëns EJ (ed) Molecular pharmacology: the mode of action of biologically active compounds, vol 1. Academic, New York, NY, pp 13–15

    Google Scholar 

  6. Ahlquist RP (1967) Development of the concept of alpha and beta adrenotropic receptors. Ann N Y Acad Sci 139:549–552

    PubMed  CAS  Google Scholar 

  7. Lands AM, Arnold A, McAuliff JP et al (1967) Differentiation of receptor systems activated by sympathomimetic amines. Nature 214:597–598

    PubMed  CAS  Google Scholar 

  8. Carlsson E, Ablad B, Brandstrom A et al (1972) Differentiated blockage of the chronotropic effects of various adrenergic stimuli in the cat heart. Life Sci 11:953–958

    CAS  Google Scholar 

  9. Alexander RW, Williams LT, Lefkowitz RJ (1975) Identification of cardiac beta-adrenergic receptors by (-)[3H]alprenolol binding. Proc Natl Acad Sci USA 72:1564–1568

    PubMed  CAS  Google Scholar 

  10. Stiles GL, Taylor S, Lefkowitz RJ (1983) Human cardiac beta-adrenergic receptors: subtype heterogeneity delineated by direct radioligand binding. Life Sci 33:467–473

    PubMed  CAS  Google Scholar 

  11. Kent RS, De Lean A, Lefkowitz RJ (1980) A quantitative analysis of beta adrenergic receptor interactions: resolution of high and low affinity states of the receptor by computer modeling of ligand binding data. Mol Pharmacol 17:14–23

    PubMed  CAS  Google Scholar 

  12. Rodbell M (1980) The role of hormone receptors and GTP-regulating proteins in membrane transduction. Nature 284:17–22

    PubMed  CAS  Google Scholar 

  13. Limbird LE, Gill DM, Lefkowitz RJ (1980) Agonist promoted coupling of the beta-adrenergic receptor with the guanine nucleotide regulatory protein of the adenylate cyclase system. Proc Natl Acad Sci USA 77:775–779

    PubMed  CAS  Google Scholar 

  14. Dixon RA, Kobilka BK, Strader DJ et al (1986) Cloning of the gene and cDNA for mammalian beta-adrenergic receptor and homology with rhodopsin. Nature 321:75–79

    PubMed  CAS  Google Scholar 

  15. Bylund DB (1992) Subtypes of alpha 1- and alpha 2-adrenergic receptors. FASEB J 6:832–839

    PubMed  CAS  Google Scholar 

  16. Graham RM, Perez DM, Hwa J et al (1996) Alpha 1-adrenergic receptor subtypes. Molecular structure, function and signaling. Circ Res 78:737–749

    PubMed  CAS  Google Scholar 

  17. Frielle T, Collins S, Daniel KW et al (1987) Cloning of the cDNA for the human beta 1-adrenergic receptor. Proc Natl Acad Sci USA 84:7920–7924

    PubMed  CAS  Google Scholar 

  18. Emorine LJ, Marullo S, Delavier-Klutchko C et al (1987) Structure of the gene for human beta 2-adrenergic receptor: expression and promoter characterization. Proc Natl Acad Sci USA 84:6995–6999

    PubMed  CAS  Google Scholar 

  19. Emorine LJ, Marullo S, Briend-Sutren MM et al (1989) Molecular characterization of the human beta 3-adrenergic receptor. Science 245:1118–1121

    PubMed  CAS  Google Scholar 

  20. Lowell BB (1998) Using gene knockout and transgenic techniques to study the physiology and pharmacology of beta3-adrenergic receptors. Endocr J 45(Suppl):S9–S13

    PubMed  CAS  Google Scholar 

  21. Rohrer DK, Desai KH, Jasper JR et al (1996) Targeted disruption of the mouse beta1-adrenergic receptor gene: developmental and cardiovascular effects. Proc Natl Acad Sci USA 93:7375–7380

    PubMed  CAS  Google Scholar 

  22. Rohrer DK, Bernstein D, Chruscinski A et al (1998) The developmental and physiological consequences of disrupting genes encoding beta 1 and beta 2 adrenoceptors. Adv Pharmacol 42:499–501

    PubMed  CAS  Google Scholar 

  23. Link RE, Stevens MS, Kulatunga M et al (1995) Targeted inactivation of the gene encoding the mouse alpha 2c-adrenoceptor homolog. Mol Pharmacol 48:48–55

    PubMed  CAS  Google Scholar 

  24. Triposkiadis F, Karayannis G, Giamouzis G et al (2009) The sympathetic nervous system in heart failure physiology, pathophysiology, and clinical implications. J Am Coll Cardiol 54:1747–1762

    PubMed  CAS  Google Scholar 

  25. Rockman HA, Koch WJ, Lefkowitz RJ (2002) Seven-transmembrane-spanning receptors and heart function. Nature 415:206–212

    PubMed  CAS  Google Scholar 

  26. Port JD, Bristow MR (2001) beta-Adrenergic receptors, transgenic mice, and pharmacological model systems. Mol Pharmacol 60:629–631

    PubMed  CAS  Google Scholar 

  27. Lohse MJ, Engelhardt S, Eschenhagen T (2003) What is the role of beta-adrenergic signaling in heart failure? Circ Res 93:896–906

    PubMed  CAS  Google Scholar 

  28. Brodde OE, Bruck H, Leineweber K (2006) Cardiac adrenoceptors: physiological and pathological relevance. J Pharmacol Sci 100:323–337

    PubMed  CAS  Google Scholar 

  29. Vatner SF, Franklin D, Higgins CB et al (1972) Left ventricular response to severe exertion in untethered dogs. J Clin Invest 51:3052–3060

    PubMed  CAS  Google Scholar 

  30. Gauthier C, Tavernier G, Charpentier F et al (1996) Functional beta3-adrenoceptor in the human heart. J Clin Invest 98:556–562

    PubMed  CAS  Google Scholar 

  31. Molinari C, Battaglia A, Grossini E et al (1999) The role of beta 2-adrenergic vascular receptors in the peripheral vasodilation caused by 17 beta-estradiol in anesthetized pigs. Life Sci 65:1545–1552

    PubMed  CAS  Google Scholar 

  32. Arch JS, Ainsworth AT, Cawthorne MA et al (1984) Atypical beta-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature 309:163–165

    PubMed  CAS  Google Scholar 

  33. Shen YT, Cervoni P, Claus T et al (1996) Differences in beta 3-adrenergic receptor cardiovascular regulation in conscious primates, rats, dogs. J Pharmacol Exp Ther 278:1435–1443

    PubMed  CAS  Google Scholar 

  34. Tavernier G, Toumaniantz G, Erfanian M et al (2003) beta3-Adrenergic stimulation produces a decrease of cardiac contractility ex vivo in mice overexpressing the human beta3-adrenergic receptor. Cardiovasc Res 59:288–296

    PubMed  CAS  Google Scholar 

  35. Gauthier C, Leblais V, Kobzik L et al (1998) The negative inotropic effect of beta3-adrenoceptor stimulation is mediated by activation of the nitric oxide synthase pathway in human ventricle. J Clin Invest 102:1377–1384

    PubMed  CAS  Google Scholar 

  36. Niu X, Watts VL, Cingolani OH et al (2012) Cardioprotective effect of beta-3 adrenergic receptor agonism: role of neuronal nitric oxide synthase. J Am Coll Cardiol 59:1979–1987

    PubMed  CAS  Google Scholar 

  37. Ishikawa Y, Homcy CJ (1997) The adenylyl cyclases as integrators of transmembrane signal transduction. Circ Res 80:297–304

    PubMed  CAS  Google Scholar 

  38. Gerhardstein BL, Puri TS, Chien AJ et al (1999) Identification of the sites phosphorylated by cyclic AMP-dependent protein kinase on the beta-2 subunit of the L-type voltage-dependent calcium channels. Biochemistry 38:10361–10370

    PubMed  CAS  Google Scholar 

  39. Marx SO, Reiken S, Hisamatsu Y et al (2000) PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell 101:365–376

    PubMed  CAS  Google Scholar 

  40. Simmerman HK, Jones LR (1998) Phospholamban: protein structure, mechanism of action, and role in cardiac function. Physiol Rev 78:921–947

    PubMed  CAS  Google Scholar 

  41. Sulakhe PV, Vo XT (1995) Regulation of phospholamban and troponin-I phosphorylation in the intact rat cardiomyocytes by adrenergic and cholinergic stimuli: roles of cyclic nucleotides, calcium, protein kinases and phosphatases and depolarization. Mol Cell Biochem 149–150:103–126

    PubMed  Google Scholar 

  42. De Jongh KS, Murphy BJ, Colvin AA et al (1996) Specific phosphorylation of a site in the full-length form of the alpha 1 subunit of the cardiac L-type calcium channel by adenosine 3′,5′-cyclic monophosphate-dependent protein kinase. Biochemistry 35:10392–10402

    PubMed  Google Scholar 

  43. Ginsburg KS, Bers DM (2004) Modulation of excitation-contraction coupling by ­isoproterenol in cardiomyocytes with controlled SR Ca2+ load and Ca2+ current trigger. J Physiol 556:463–480

    PubMed  CAS  Google Scholar 

  44. Shan J, Kushnir A, Betzenhauser MJ et al (2010) Phosphorylation of the ryanodine receptor mediates the cardiac fight or flight response in mice. J Clin Invest 120:4388–4398

    PubMed  CAS  Google Scholar 

  45. Luo W, Grupp IL, Harrer J et al (1994) Targeted ablation of the phospholamban gene is associated with markedly enhanced myocardial contractility and loss of beta-agonist stimulation. Circ Res 75:401–409

    PubMed  CAS  Google Scholar 

  46. MacLennan DH, Kranias EG (2003) Phospholamban: a crucial regulator of cardiac contractility. Nat Rev Mol Cell Biol 4:566–577

    PubMed  CAS  Google Scholar 

  47. Solaro RJ, Moir AJ, Perry SV (1976) Phosphorylation of troponin I and the inotropic effect of adrenaline in the perfused rabbit heart. Nature 262:615–617

    PubMed  CAS  Google Scholar 

  48. Zhang R, Zhao J, Mandveno A et al (1995) Cardiac troponin I phosphorylation increases the rate of cardiac muscle relaxation. Circ Res 76:1028–1035

    PubMed  CAS  Google Scholar 

  49. Kaumann A, Bartel S, Molenaar P (1999) Activation of beta2-adrenergic receptors hastens relaxation and mediates phosphorylation of phospholamban, troponin I, and C-protein in ventricular myocardium from patients with terminal heart failure. Circulation 99:65–72

    PubMed  CAS  Google Scholar 

  50. Xiao RP, Ji X, Lakatta EG (1995) Functional coupling of the β2-adrenoceptor to a pertussis toxin-sensitive G protein in cardiac myocytes. Mol Pharmacol 47:322–329

    PubMed  CAS  Google Scholar 

  51. Xiao RP, Avdonin P, Zhou YY et al (1999) Coupling of beta2-adrenoreceptor to Gi proteins and its physiological relevance in murine cardiac myocytes. Circ Res 84:43–52

    PubMed  CAS  Google Scholar 

  52. Lemoine H, Kaumann AJ (1991) Regional differences of beta1- and beta2-adrenoceptor-mediated functions in feline heart. A beta2-adrenoceptor mediated positive inotropic effects possibly unrelated to cyclic AMP. Naunyn Schmiedebergs Arch Pharmacol 334:56–69

    Google Scholar 

  53. McConville P, Fishbein KW, Lakatta EG et al (2003) Differences in the bioenergetic response of the isolated perfused rat heart to selective beta1- and beta2-adrenergic receptor stimulation. Circulation 107:2146–2152

    PubMed  Google Scholar 

  54. McConville P, Spencer RG, Lakatta EG (2005) Temporal dynamics of inotropic, chronotropic and metabolic responses during beta1- and beta2-AR stimulation in the isolated, perfused rat heart. Am J Physiol Endocrinol Metab 289:E412–E418

    PubMed  CAS  Google Scholar 

  55. Kilts JD, Gerhardt MA, Richardson MD et al (2000) Beta2-adrenergic and several other G protein-couple receptors in human atrial membranes activate both Gs and Gi. Circ Res 87:705–709

    PubMed  CAS  Google Scholar 

  56. Molenaar P, Savarimuthu SM, Sarsero D et al (2007) (-)-Adrenaline elicits positive inotropic, lusitropic, and biochemical effects through beta2-adrenoceptors in human atrial myocardium from nonfailing and failing hearts, consistent with Gs coupling but not with Gi coupling. Naunyn Schmiedebergs Arch Pharmacol 375:11–28

    PubMed  CAS  Google Scholar 

  57. Bristow MR, Ginsburg R, Minobe W et al (1982) Decreased catecholamine sensitivity and β-adrenergic-receptor density in failing human hearts. N Engl J Med 307:205–211

    PubMed  CAS  Google Scholar 

  58. Bristow MR, Ginsburg R, Umans V et al (1986) Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ Res 59:297–309

    PubMed  CAS  Google Scholar 

  59. Kiuchi K, Shannon RP, Komamura K et al (1993) Myocardial beta-adrenergic receptor function during the development of pacing-induced heart failure. J Clin Invest 91:907–914

    PubMed  CAS  Google Scholar 

  60. Hittinger L, Mirsky I, Shen YT et al (1995) Hemodynamic mechanisms responsible for reduced subendocardial coronary reserve in dogs with severe left ventricular hypertrophy. Circulation 92:978–986

    PubMed  CAS  Google Scholar 

  61. Nakano K, Corin WJ, Spann JF Jr et al (1989) Abnormal subendocardial blood flow in response to overload hypertrophy is associated with pacing-induced subendocardial dysfunction. Circ Res 65:1555–1564

    PubMed  CAS  Google Scholar 

  62. Goldberg LI, Bloodwell RD, Braunwald E et al (1960) The direct effects of norepinephrine, epinephrine, and methoxamine on myocardial contractile force in man. Circulation 22:1125–1132

    PubMed  CAS  Google Scholar 

  63. Smith HJ, Oriol A, Morch J et al (1967) Hemodynamic studies in cardiogenic shock. Treatment with isoproterenol and metaraminol. Circulation 35:1084–1091

    PubMed  CAS  Google Scholar 

  64. Maroko PR, Kjekshus JK, Sobel BE et al (1971) Factors influencing infarct size following experimental coronary artery occlusions. Circulation 43:67–82

    PubMed  CAS  Google Scholar 

  65. Hittinger L, Shannon RP, Kohin S et al (1989) Isoproterenol-induced alterations in myocardial blood flow, systolic and diastolic function in conscious dogs with heart failure. Circulation 80:658–668

    PubMed  CAS  Google Scholar 

  66. Lipp H, Fallicov RE, Resnekov L et al (1972) The effects of dopamine on depressed myocardial function following coronary embolization in the closed chest dog. Am Heart J 84:208–214

    PubMed  CAS  Google Scholar 

  67. Gunnar RM, Loeb HS (1972) Use of drugs in cardiogenic shock due to acute myocardial infarction. Circulation 45:1111–1124

    PubMed  CAS  Google Scholar 

  68. Tsai TH, Langer SZ, Trendelenburg U (1967) Effects of dopamine and alpha methyl-dopamine on smooth muscle and on the cardiac pacemaker. J Pharmacol Exp Ther 156:310–324

    PubMed  CAS  Google Scholar 

  69. Nash CW, Wolff SA, Ferguson BA (1968) Release of tritiated noradrenaline from perfused rat hearts by sympathomimetic amines. Can J Physiol Pharmacol 46:35–42

    PubMed  CAS  Google Scholar 

  70. Brodde OE, Inui J, Motomura S (1980) The mode of direct action of dopamine on the rabbit heart. J Cardiovasc Pharmacol 2:567–582

    PubMed  CAS  Google Scholar 

  71. Chidsey CA, Braunwald E, Morrow AG et al (1963) Myocardial norepinephrine concentration in man: effects of reserpine and of congestive heart failure. N Engl J Med 269:653–658

    PubMed  CAS  Google Scholar 

  72. Tuttle RR, Mills K (1975) Dobutamine. Development of a new catecholamine to selectively increase cardiac contractility. Circ Res 36:185–196

    PubMed  CAS  Google Scholar 

  73. McNay JL, McDonald RH Jr, Goldberg LI (1965) Direct renal vasodilation produced by dopamine in the dog. Circ Res 16:510–517

    PubMed  CAS  Google Scholar 

  74. Vatner SF, Millard RW, Higgins CB (1973) Coronary and myocardial effects of dopamine in the conscious dog: parasympatholytic augmentation of pressor and inotropic actions. J Pharmacol Exp Ther 187:280–295

    PubMed  CAS  Google Scholar 

  75. Vatner SF, McRitchie RJ, Braunwald E (1974) Effects of dobutamine on left ventricular ­performance, coronary dynamics, and redistribution of cardiac output. J Clin Invest 53:1265–1273

    PubMed  CAS  Google Scholar 

  76. Franciosa JA, Limas CJ, Guiha NH et al (1972) Improved left ventricular function during nitroprusside infusion in acute myocardial infarction. Lancet 1:650–654

    PubMed  CAS  Google Scholar 

  77. Guiha NJ, Cohn JN, Mikulic E et al (1974) Treatment of refractory heart failure with infusion of nitroprusside. N Engl J Med 291:587–592

    PubMed  CAS  Google Scholar 

  78. Miller RR, Vismara LA, Zelis R et al (1975) Clinical use of sodium nitroprusside in chronic ischemic heart disease. Effects on peripheral vascular resistance and venous tone and on ventricular volume, pump and mechanical performance. Circulation 51:328–336

    PubMed  CAS  Google Scholar 

  79. Capomolla S, Febo O, Opasich C et al (2001) Chronic infusion of dobutamine and nitroprusside in patients with end-stage heart failure awaiting heart transplantation: safety and clinical outcome. Eur J Heart Fail 3:601–610

    PubMed  CAS  Google Scholar 

  80. Elkayam U, Tasissa G, Brinanay C et al (2007) Use and impact of inotropes and vasodilator therapy in hospitalized patients with severe heart failure. Am Heart J 153:98–104

    PubMed  Google Scholar 

  81. Fitzpatrick D, Ikram H, Nicholls MG et al (1983) Hemodynamic, hormonal and electrolyte responses to prenalterol. Circulation 67:613–619

    PubMed  CAS  Google Scholar 

  82. Petch MC, Wisbey C, Ormerod O et al (1984) Acute haemodynamic effects of oral prenalterol in severe heart failure. Br Heart J 52:49–52

    PubMed  CAS  Google Scholar 

  83. Lambertz H, Meyer J, Erbel R (1984) Long-term hemodynamic effects of prenalterol in patients with severe congestive heart failure. Circulation 69:298–305

    PubMed  CAS  Google Scholar 

  84. Gollub SB, Elkayam U, Young JB et al (1991) Efficacy and safety of a short-term (6-h) intravenous infusion of dopexamine in patients with severe congestive heart failure: a randomized, double-blind, parallel, placebo-controlled multicenter study. J Am Coll Cardiol 18:383–390

    PubMed  CAS  Google Scholar 

  85. Asanoi H, Sasayama S, Sakurai T et al (1995) Intravenous dopexamine in the treatment of acute congestive heart failure: results of a multicenter, double-blind, placebo-controlled withdrawal study. Cardiovasc Drugs Ther 9:791–797

    PubMed  CAS  Google Scholar 

  86. Felker GM, Benza RL, Chandler AB et al (2003) Heart failure etiology and response to milrinone in decompensated heart failure: results from the OPTIME-CHF study. J Am Coll Cardiol 41:997–1003

    PubMed  CAS  Google Scholar 

  87. Installé E, Gonzales M, Jacquemart JL et al (1987) Comparative effects on hemodynamics of enoximone (MDL 17, 043), dobutamine and nitroprusside in severe congestive heart failure. Am J Cardiol 60:46C–52C

    PubMed  Google Scholar 

  88. Cowley AJ, Skene AM (1994) Treatment of severe heart failure: quantity or quality of life? A trial of enoximone. Enoximone investigators. Br Heart J 72:226–230

    PubMed  CAS  Google Scholar 

  89. Metra M, Eichhorn E, Abraham WT et al (2009) Effects of low-dose oral enoximone administration on mortality, morbidity, and exercise capacity in patients with advanced heart failure: the randomized, double-blind, placebo-controlled, parallel group ESSENTIAL trials. Eur Heart J 30:3015–3026

    PubMed  CAS  Google Scholar 

  90. Milano CA, Allen LF, Rockman HA et al (1994) Enhanced myocardial function in transgenic mice overexpressing the beta 2-adrenergic receptor. Science 264:582–586

    PubMed  CAS  Google Scholar 

  91. Liggett SB, Tepe NM, Lorenz JN et al (2000) Early and delayed consequences of beta(2)-adrenergic receptor overexpression in mouse hearts: critical role for expression level. Circulation 101:1707–1714

    PubMed  CAS  Google Scholar 

  92. Tevaearai HT, Eckhart AD, Walton GB et al (2002) Myocardial gene transfer and overexpression of beta2-adrenergic receptors potentiates the functional recovery of unloaded failing hearts. Circulation 106:124–129

    PubMed  CAS  Google Scholar 

  93. Iwase M, Bishop SP, Uechi M et al (1996) Adverse effects of chronic endogenous sympathetic drive induced by cardiac Gs alpha overexpression. Circ Res 78:517–524

    PubMed  CAS  Google Scholar 

  94. Asai K, Yang GP, Geng YJ et al (1999) Beta-adrenergic receptor blockade arrests myocyte damage and preserves cardiac function in the transgenic G(salpha) mouse. J Clin Invest 104:551–558

    PubMed  CAS  Google Scholar 

  95. Iwase M, Uechi M, Vatner DE et al (1997) Cardiomyopathy induced by cardiac Gs alpha overexpression. Am J Physiol 272:H585–H589

    PubMed  CAS  Google Scholar 

  96. Engelhardt S, Hein L, Wiesmann F et al (1999) Progressive hypertrophy and heart failure in beta1-adrenergic receptor transgenic mice. Proc Natl Acad Sci USA 96:7059–7064

    PubMed  CAS  Google Scholar 

  97. Peter P, Brady J, Yan L et al (2007) Inhibition of p38 alpha MAPK rescues cardiomyopathy induced by overexpressed beta 2-adrenergic receptor, but not beta 1-adrenergic receptor. J Clin Invest 117:1335–1343

    PubMed  CAS  Google Scholar 

  98. Engelhardt S, Hein L, Dyachenkow V et al (2004) Altered calcium handling is critically involved in the cardiotoxic effects of chronic beta-adrenergic stimulation. Circulation 109:1154–1160

    PubMed  CAS  Google Scholar 

  99. Du XJ, Gao XM, Wang B et al (2000) Age-dependent cardiomyopathy and heart failure phenotype in mice overexpressing beta(2)-adrenergic receptors in the heart. Cardiovasc Res 48:448–454

    PubMed  CAS  Google Scholar 

  100. Zaugg M, Xu W, Lucchinetti E et al (2000) Beta-adrenergic receptor subtypes differentially affect apoptosis in adult rat ventricular myocytes. Circulation 102:344–350

    PubMed  CAS  Google Scholar 

  101. Morisco C, Zebrowski DC, Vatner DE et al (2001) Beta-adrenergic cardiac hypertrophy is mediated primarily by the beta(1) subtype in the rat heart. J Mol Cell Cardiol 33:561–573

    PubMed  CAS  Google Scholar 

  102. Rona G, Chappel CI, Balazs T et al (1959) An infarct-like myocardial lesion and other toxic manifestations produced by isoproterenol in the rat. AMA Arch Pathol 67:443–445

    PubMed  CAS  Google Scholar 

  103. Rona G (1985) Catecholamine cardiotoxicity. J Mol Cell Cardiol 17:291–306

    PubMed  CAS  Google Scholar 

  104. Chappel CI, Rona G, Balaz T et al (1959) Severe myocardial necrosis produced by isoproterenol in the rat. Arch Int Pharmacodyn Ther 112:123–128

    Google Scholar 

  105. Jalil JE, Janicki JS, Pick R et al (1989) Fibrosis-induced reduction of endomyocardium in the rat after isoproterenol treatment. Circ Res 65:258–264

    PubMed  CAS  Google Scholar 

  106. Tisné-Versailles J, Constantin M, Lamar JC et al (1985) Cardiotoxicity of high doses of isoproterenol on cardiac haemodynamics and metabolism in SHR and WKY rats. Arch Int Pharmacodyn Ther 273:142–154

    PubMed  Google Scholar 

  107. Kudej RK, Iwase M, Uechi M et al (1997) Effects of chronic beta-adrenergic receptor stimulation in mice. J Mol Cell Cardiol 29:2735–2746

    PubMed  CAS  Google Scholar 

  108. Lai L, Yan L, Hu CL et al (2009) Abstract 3290: increased Type 5 adenylyl cyclase expression mediates chronic catecholamine stress via increases in oxidative stress and down-regulation of MnSOD. Circulation 120:S781

    Google Scholar 

  109. Balke CW, Shorofsky SR (1998) Alterations in calcium handling in cardiac hypertrophy and heart failure. Cardiovasc Res 37:290–299

    PubMed  CAS  Google Scholar 

  110. Gwathmey JK, Morgan JP (1985) Altered calcium handling in experimental pressure-overload hypertrophy in the ferret. Circ Res 57:836–843

    PubMed  CAS  Google Scholar 

  111. Gwathmey JK, Copelas L, MacKinnon R et al (1987) Abnormal intracellular calcium handling in myocardium from patients with end-stage heart failure. Circ Res 61:70–76

    PubMed  CAS  Google Scholar 

  112. Beuckelmann DJ, Näbauer M, Erdmann E (1992) Intracellular calcium handling in isolated ventricular myocytes from patients with terminal heart failure. Circulation 85:1046–1055

    PubMed  CAS  Google Scholar 

  113. Bartel S, Stein B, Eschenhagen T et al (1996) Protein phosphorylation in isolated trabeculae from nonfailing and failing human hearts. Mol Cell Biochem 157:171–179

    PubMed  CAS  Google Scholar 

  114. Schröder F, Handrock R, Beuckelmann DJ et al (1998) Increased availability and open probability of single L-type calcium channels from failing compared with nonfailing human ventricle. Circulation 98:969–976

    PubMed  Google Scholar 

  115. Molkentin JD, Lu JR, Antos CL et al (1998) A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell 93:215–228

    PubMed  CAS  Google Scholar 

  116. Zhang T, Johnson EN, Gu Y et al (2002) The cardiac-specific nuclear delta(B) isoform of Ca2+/calmodulin-dependent protein kinase II induces hypertrophy and dilated cardiomyopathy associated with increased protein phosphatase 2A activity. J Biol Chem 227:1261–1267

    Google Scholar 

  117. Bloom S, Davis DL (1972) Calcium as mediator of isoproterenol-induced myocardial necrosis. Am J Pathol 69:459–470

    PubMed  CAS  Google Scholar 

  118. Cheema Y, Sherrod JN, Zhao W et al (2011) Mitochondriocentric pathway to cardiomyocyte necrosis in aldosteronism: cardioprotective responses to carvedilol and nebivolol. J Cardiovasc Pharmacol 58:80–86

    PubMed  CAS  Google Scholar 

  119. Yan L, Vatner DE, O’Connor JP et al (2007) Type 5 adenylyl cyclase disruption increases longevity and protects against stress. Cell 2:247–258

    Google Scholar 

  120. Strohm C, Barancik T, Bruehl ML et al (2000) Inhibition of the ER-Kinase by PD98059 and UO126 counteracts ischemic preconditioning in pig myocardium. J Cardiovasc Pharmacol 36:218–229

    PubMed  CAS  Google Scholar 

  121. Ping P, Zhang J, Cao X et al (1999) PKC-dependent activation of p44/p42 MAPKs during myocardial ischemia-reperfusion in conscious rabbits. Am J Physiol Heart Circ Physiol 276:H1468–H1481

    CAS  Google Scholar 

  122. Fryer RM, Hsu AK, Gross GJ (1995) ERK and p38 MAP kinase activation are components of opioid-induced delayed cardioprotection. Basic Res Cardiol 96:136–142

    Google Scholar 

  123. Yue TL, Gu JL, Wang C et al (2000) Extracellular signal-regulated kinase plays an essential role in hypertrophic agonists, endothelin-1 and phenylephrine-induced cardiomyocyte hypertrophy. J Biol Chem 275:37895–37901

    PubMed  CAS  Google Scholar 

  124. Karoor V, Vatner SF, Takagi G et al (2004) Propranolol prevents enhanced stress signaling in Gs alpha cardiomyopathy: potential mechanism for beta-blockade in heart failure. J Mol Cell Cardiol 36:305–312

    PubMed  CAS  Google Scholar 

  125. Condorelli G, Drusco A, Stassi G et al (2002) Akt induces enhanced myocardial contractility and cell size in vivo in transgenic mice. Proc Natl Acad Sci USA 99:12333–12338

    PubMed  CAS  Google Scholar 

  126. Walsh K (2006) Akt signaling and growth of the heart. Circulation 113:2032–2034

    PubMed  Google Scholar 

  127. DeBosch B, Treskov I, Lupu TS et al (2006) Akt1 is required for physiological cardiac growth. Circulation 113:2097–2104

    PubMed  CAS  Google Scholar 

  128. Kim YK, Kim SJ, Yatani A et al (2003) Mechanism of enhanced cardiac function in mice with hypertrophy induced by overexpressed Akt. J Biol Chem 278:47622–47628

    PubMed  CAS  Google Scholar 

  129. Rota M, Boni A, Urbanek K (2005) Nuclear targeting of Akt enhances ventricular function and myocyte contractility. Circ Res 97:1332–1341

    PubMed  CAS  Google Scholar 

  130. Matsui T, Li L, del Monte F et al (1999) Adenoviral gene transfer of activated ­phosphatidylinositol 3′-kinase and Akt inhibits apoptosis of hypoxic cardiomyocytes in vitro. Circulation 100:2373–2379

    PubMed  CAS  Google Scholar 

  131. Fujio Y, Nguyen T, Wencker D et al (2000) Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. Circulation 101:660–667

    PubMed  CAS  Google Scholar 

  132. Matsui T, Tao J, del Monte F et al (2001) Akt activation preserves cardiac function and ­prevents injury after transient cardiac ischemia in vivo. Circulation 104:330–335

    PubMed  CAS  Google Scholar 

  133. Okumura S, Vatner DE, Kurotani R et al (2007) Disruption of type 5 adenylyl cyclase enhances desensitization of cyclic adenosine monophosphate signal and increases Akt signal with chronic catecholamine stress. Circulation 116:1776–1783

    PubMed  CAS  Google Scholar 

  134. Matsui T, Li L, Wu JC et al (2002) Phenotypic spectrum caused by transgenic overexpression of activated Akt in the heart. J Biol Chem 227:22896–22901

    Google Scholar 

  135. Nagoshi T, Matsui T, Aoyama T et al (2005) PI3K rescues the detrimental effects of chronic Akt activation in the heart during ischemia/reperfusion injury. J Clin Invest 115:2128–2138

    PubMed  CAS  Google Scholar 

  136. Packer M, Bristow MR, Cohn JN et al (1996) The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. carvedilol heart failure study group. N Engl J Med 334:1349–1355

    PubMed  CAS  Google Scholar 

  137. O’Connor CM, Gattis WA, Uretsky BF et al (1999) Continuous intravenous dobutamine is associated with an increased risk of death in patients with advanced heart failure: insights from the Flolan International Randomized Survival Trial (FIRST). Am Heart J 138:78–86

    PubMed  Google Scholar 

  138. Ginsburg R, Bristow MR, Billingham ME et al (1983) Study of the normal and failing isolated human heart: decreased response of failing heart to isoproterenol. Am Heart J 106:535–540

    PubMed  CAS  Google Scholar 

  139. Fowler MB, Laser JA, Hopkins GL et al (1986) Assessment of the beta-adrenergic receptor pathway in the intact failing human heart: progressive down-regulation and subsensitivity to agonist response. Circulation 74:1290–1302

    PubMed  CAS  Google Scholar 

  140. Bristow MR, Minobe WA, Raynolds MV et al (1993) Reduced beta 1 receptor messenger RNA abundance in the failing human heart. J Clin Invest 92:2737–2745

    PubMed  CAS  Google Scholar 

  141. Ungerer M, Böhm M, Elce JS et al (1993) Altered expression of beta-adrenergic receptor kinase and beta 1-adrenergic receptors in the failing human heart. Circulation 87:454–463

    PubMed  CAS  Google Scholar 

  142. Pende A, Tremmel KD, DeMaria CT et al (1996) Regulation of the mRNA-binding protein AUF1 by activation of the beta-adrenergic receptor signal transduction pathway. J Biol Chem 271:8493–8501

    PubMed  CAS  Google Scholar 

  143. Hausdorff WP, Caron MG, Lefkowitz RJ (1990) Turning off the signal: desensitization of beta-adrenergic receptor function. FASEB J 4:2881–2889

    PubMed  CAS  Google Scholar 

  144. Lohse MJ (1992) Mechanisms of b-adrenergic receptor desensitization. In: Hargrave PA, Hofmann KP, Kaupp UB (eds) Signal Transmission in Photoreceptor Systems. Berlin, Springer-Verlag, pp 160–171

    Google Scholar 

  145. Lohse MJ, Benovic JL, Caron MG et al (1990) Multiple pathways of rapid beta 2-adrenergic receptor desensitization. J Biol Chem 265:3202–3211

    PubMed  CAS  Google Scholar 

  146. Benovic JL, Mayor F Jr, Staniszewski C (1987) Purification and characterization of the beta-adrenergic receptor kinase. J Biol Chem 262:9026–9032

    PubMed  CAS  Google Scholar 

  147. Lohse MJ, Benovic JL, Codina J et al (1990) beta-Arrestin: a protein that regulates beta-adrenergic receptor function. Science 248:1547–1550

    PubMed  CAS  Google Scholar 

  148. Gerskowitch VP, Hull RAD, Shankley NP (1988) The pharmacological toolmakers rational approach to drug design—an appreciation of Sir James Black. Trends Pharmacol Sci 9:435–437

    PubMed  CAS  Google Scholar 

  149. Black JW (1976) Ahlquist and the development of beta-adrenoceptor antagonists. Postgrad Med J 52(4):11–13

    PubMed  Google Scholar 

  150. Black JW, Stephenson JS (1962) Pharmacology of a new adrenergic beta-receptor-blocking compound (Nethalide). Lancet 2:311–314

    PubMed  CAS  Google Scholar 

  151. Elij D, Medez R (1964) The modification of digitalis intoxication by excluding adrenergic influences on the heart. J Pharmacol Exp Ther 144:97–103

    Google Scholar 

  152. Williams EM, Sekiya A (1963) Prevention of arrhythmias due to cardiac glycosides by block of sympathetic beta-receptors. Lancet 1:420–421

    PubMed  CAS  Google Scholar 

  153. Somani P, Hardman HF, Lum BKB (1963) Antagonism of cardiac arrhythmias by nethalide. Phamacologist 5:260

    Google Scholar 

  154. Murray WJ, McKnight RL, Davis DA (1963) Antagonism of hydrocarbon anaesthetic-­epinephrine arrhythmias in dogs by nethalide, a dichloroisoproterenol analogue. Proc Soc Exp Biol med 113:439–441

    PubMed  CAS  Google Scholar 

  155. Prichard BN, Dickson CJ, Alleyne GA et al (1963) Effect of pronethalol in angina pectoris. Br Med J 2:1226–1229

    PubMed  CAS  Google Scholar 

  156. Black JW, Duncan WA, Shanks RG (1965) Comparison of some properties of pronethalol and propranolol. Br J Phamacol Chemother 25:577–591

    CAS  Google Scholar 

  157. Allin EF, Miller JM, Rowe GG et al (1974) Effects of intraperitoneal administration of propranolol on the mouse heart. Histochemical and electron microscopic observations. Am J Cardiol 33:639–642

    PubMed  CAS  Google Scholar 

  158. Fearson RE (1967) Propranolol in the prevention of ventricular fibrillation due to experimental coronary artery occlusion. Observation on its mode of action. Am J Cardiol 20:222–228

    Google Scholar 

  159. Khan MI, Hamilton JT, Manning GW (1972) Protective effect of beta adrenoceptor blockade in experimental coronary occlusion in conscious dogs. Am J Cardiol 30:832–837

    PubMed  CAS  Google Scholar 

  160. Mueller HS, Ayres SM, Religa A et al (1974) Propranolol in the treatment of acute myocardial infarction. Circulation 49:1078–1087

    PubMed  CAS  Google Scholar 

  161. Hawkins CM, Richardson DW, Vokonas PS (1983) Effect of propranolol in reducing mortality in older myocardial infarction patients. The Beta-Blocker Heart Attack Trial experience. Circulation 67:194–197

    Google Scholar 

  162. Hamer J, Grandjean T, Melendez L et al (1964) Effect of propranolol (Inderal) in angina pectoris: preliminary report. Br Med J 2:720–723

    PubMed  CAS  Google Scholar 

  163. Keelan P (1965) Double-blind trial of propranolol (Inderal) in angina pectoris. Br Med J 1:897–898

    PubMed  CAS  Google Scholar 

  164. Besterman EM, Friedlander DH (1965) Clinical experiences with propranolol. Postgrad Med J 41:526–535

    PubMed  CAS  Google Scholar 

  165. Dunlop D, Shanks RG (1968) Selective blockade of adrenoceptive beta receptors in the heart. Brit J Pharmacol Chemother 32:201

    CAS  Google Scholar 

  166. Kubota T, Yamazaki N, Sudo J et al (1990) Protective effects of adrenoceptor-blocking agents on myocardial injury induced by epinephrine in mice. J Toxicol Sci 15:1–13

    PubMed  CAS  Google Scholar 

  167. Waagstein F, Hjalmarson A, Varnauskas E et al (1975) Effect of chronic beta-adrenergic receptor blockade in congestive cardiomyopathy. Br Heart J 37:1022–1036

    PubMed  CAS  Google Scholar 

  168. Waagstein F, Bristow MR, Swedberg K et al (1993) Beneficial effects of metoprolol in idiopathic dilated cardiomyopathy. Metoprolol in dilated cardiomyopathy (MDC) trial study group. Lancet 342:1441–1446

    PubMed  CAS  Google Scholar 

  169. CIBIS investigators and committees (1994) A randomized trial of beta-blockade in heart failure. The Cardiac Insufficiency Bisoprolol Study (CIBIS). Circulation 90:1765–1773

    Google Scholar 

  170. CIBIS-II investigators and committees (1999) The cardiac insufficiency bisoprolol study II (CIBIS-II): a randomised trial. Lancet 353:9–13

    Google Scholar 

  171. Segev A, Mekori YA (1999) The cardiac insufficiency bisoprolol study II. Lancet 353:1361

    PubMed  CAS  Google Scholar 

  172. Drummond GA, Squire IB (1999) The cardiac insufficiency bisoprolol study II. Lancet 353:1361

    PubMed  CAS  Google Scholar 

  173. Poole-Wilson PA (1999) The cardiac insufficiency bisoprolol study II. Lancet 353:1360–1361

    PubMed  CAS  Google Scholar 

  174. Hjalmarson A, Goldstein S, Fagerberg B et al (2000) Effects of controlled-release metoprolol on total mortality, hospitalizations, and well-being in patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure (MERIT-HF). MERIT-HF Study Group. JAMA 283:1295–1302

    PubMed  CAS  Google Scholar 

  175. Aarons RD, Molinoff PB (1982) Changes in the density of beta adrenergic receptors in rat lymphocytes, heart and lung after chronic treatment with propranolol. J Pharmacol Exp Ther 221:439–443

    PubMed  CAS  Google Scholar 

  176. Gilbert EM, Abraham WT, Olsen S et al (1996) Comparative hemodynamic, left ventricular functional and antiadrenergic effects of chronic treatment with metoprolol versus carvedilol in the failing heart. Circulation 94:2817–2825

    PubMed  CAS  Google Scholar 

  177. Sigmund M, Jakob H, Becker H et al (1996) Effects of metoprolol on myocardial beta-­adrenoceptors and Gi alpha-proteins in patients with congestive heart failure. Eur J Clin Pharmacol 51:127–132

    PubMed  CAS  Google Scholar 

  178. Bristow MR (2000) beta-Adrenergic receptor blockade in chronic heart failure. Circulation 101:558–569

    PubMed  CAS  Google Scholar 

  179. Hata JA, Williams ML, Koch WJ (2004) Genetic manipulation of myocardial beta-adrenergic receptor activation and desensitization. J Mol Cell Cardiol 37:11–21

    PubMed  CAS  Google Scholar 

  180. Iaccarino G, Tomhave ED, Lefkowitz RJ et al (1998) Reciprocal in vivo regulation of myocardial G protein-coupled receptor kinase expression by beta-adrenergic receptor stimulation and blockade. Circulation 98:1783–1789

    PubMed  CAS  Google Scholar 

  181. Ping P, Gelzer-Bell R, Roth DA et al (1995) Reduced beta-adrenergic receptor activation decreases G-protein expression and beta-adrenergic receptor kinase activity in porcine heart. J Clin Invest 95:1271–1280

    PubMed  CAS  Google Scholar 

  182. Leineweber K, Roche P, Beilfuss A et al (2005) G-Protein-coupled receptor kinase activity in human heart failure: effects of beta-adrenoceptor blockade. Cardiovasc Res 66:512–519

    PubMed  CAS  Google Scholar 

  183. Brodde OE (2007) Beta-adrenoceptor blocker treatment and the cardiac beta-adrenoceptor-G-protein(s)-adenylyl cyclase system in chronic heart failure. Naunyn Schmiedebergs Arch Pharmacol 374:361–372

    PubMed  CAS  Google Scholar 

  184. Bodor GS, Oakeley AE, Allen PD et al (1997) Troponin I phosphorylation in the normal and failing adult human heart. Circulation 96:1495–1500

    PubMed  CAS  Google Scholar 

  185. Messer AE, Jacques AM, Marston SB (2007) Troponin phosphorylation and regulatory function in human heart muscle: dephosphorylation of Ser23/24 on troponin I could account for the contractile defect in end-stage heart failure. J Mol Cell Cardiol 42:247–259

    PubMed  CAS  Google Scholar 

  186. Reiken S, Wehrens XH, Vest JA et al (2003) Beta-blockers restore calcium release channel function and improve cardiac muscle performance in human heart failure. Circulation 107:2459–2466

    PubMed  CAS  Google Scholar 

  187. Lowes BD, Gilbert EM, Abraham WT et al (2002) Myocardial gene expression in dilated cardiomyopathy treated with beta-blocking agents. N Engl J Med 346:1357–1365

    PubMed  CAS  Google Scholar 

  188. Abraham WT, Gilbert EM, Lowes BD et al (2002) Coordinate changes in myosin heavy chain isoform gene expression are selectively associated with alterations in dilated cardiomyopathy phenotype. Mol Med 8:750–760

    PubMed  CAS  Google Scholar 

  189. Frankenstein L, Nelles M, Slavutsky M et al (2007) Beta-blockers influence the short-term and long-term prognostic information of natriuretic peptides and catecholamines in chronic heart failure independent from specific agents. J Heart Lung Transplant 26:1033–1039

    PubMed  Google Scholar 

  190. Iwatsubo K, Bravo C, Uechi M et al (2012) Prevention of heart failure in mice by an antiviral agent that inhibits type 5 cardiac adenylyl cyclase. Am J Physiol Heart Circ Physiol 302:H2622–H2628

    PubMed  CAS  Google Scholar 

  191. Shen TY, Malik FI, Zhao X et al (2010) Improvement of cardiac function by a cardiac Myosin activator in conscious dogs with systolic heart failure. Circ Heart Fail 3:522–527

    PubMed  Google Scholar 

  192. Malik FI, Hartman JJ, Elias KA et al (2011) Cardiac myosin activation: a potential therapeutic approach for systolic heart failure. Science 331:1439–1443

    PubMed  CAS  Google Scholar 

  193. Cleland JG, Teerlink JR, Senior R et al (2011) The effects of the cardiac myosin activator, omecamtiv mecarbil, on cardiac function in systolic heart failure: a double-blind, placebo-controlled, crossover, dose-ranging phase 2 trial. Lancet 378:676–683

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stephen F. Vatner M.D. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer Science+Business Media New York

About this chapter

Cite this chapter

Lee, G.J.A., Yan, L., Vatner, D.E., Vatner, S.F. (2013). β-Adrenergic Receptor Signaling in Heart Failure. In: Jugdutt, B., Dhalla, N. (eds) Cardiac Remodeling. Advances in Biochemistry in Health and Disease, vol 5. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-5930-9_1

Download citation

Publish with us

Policies and ethics