Vol 28, No 3 (2021)
Review Article
Published online: 2020-04-17

open access

Page views 1419
Article views/downloads 1381
Get Citation

Connect on Social Media

Connect on Social Media

Role of extracellular signal-regulated kinase 1/2 signaling underlying cardiac hypertrophy

Zhi-Peng Yan12, Jie-Ting Li2, Ni Zeng2, Guo-Xin Ni1
Pubmed: 32329039
Cardiol J 2021;28(3):473-482.

Abstract

Cardiac hypertrophy is the result of increased myocardial cell size responding to an increased workload and developmental signals. These extrinsic and intrinsic stimuli as key drivers of cardiac hypertrophy have spurred efforts to target their associated signaling pathways. The extracellular signal-regulated kinases 1/2 (ERK1/2), as an essential member of mitogen-activated protein kinases (MAPKs), has been widely recognized for promoting cardiac growth. Several modified transgenic mouse models have been generated through either affecting the upstream kinase to change ERK1/2 activity, manipulating the direct role of ERK1/2 in the heart, or targeting phosphatases or MAPK scaffold proteins to alter total ERK1/2 activity in response to an increased workload. Using these models, both regulation of the upstream events and modulation of each isoform and indirect effector could provide important insights into how ERK1/2 modulates cardiomyocyte biology. Furthermore, a plethora of compounds, inhibitors, and regulators have emerged in consideration of ERK, or its MAPK kinases, are possible therapeutic targets against cardiac hypertrophic diseases. Herein, is a review of the available evidence regarding the exact role of ERK1/2 in regulating cardiac hypertrophy and a discussion of pharmacological strategy for treatment of cardiac hypertrophy.

Article available in PDF format

View PDF Download PDF file

References

  1. Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol. 2016; 97: 245–262.
  2. Nakamura M, Sadoshima J. Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol. 2018; 15(7): 387–407.
  3. Rauen KA. The RASopathies. Annu Rev Genomics Hum Genet. 2013; 14: 355–369.
  4. Post GR, Goldstein D, Thuerauf DJ, et al. Dissociation of p44 and p42 mitogen-activated protein kinase activation from receptor-induced hypertrophy in neonatal rat ventricular myocytes. J Biol Chem. 1996; 271(14): 8452–8457.
  5. Yamazaki T, Tobe K, Hoh E, et al. Mechanical loading activates mitogen-activated protein kinase and S6 peptide kinase in cultured rat cardiac myocytes. J Biol Chem. 1993; 268(16): 12069–12076.
  6. Ueyama T, Kawashima S, Sakoda T, et al. Requirement of activation of the extracellular signal-regulated kinase cascade in myocardial cell hypertrophy. J Mol Cell Cardiol. 2000; 32(6): 947–960.
  7. Liang F, Lu S, Gardner DG. Endothelin-dependent and -independent components of strain-activated brain natriuretic peptide gene transcription require extracellular signal regulated kinase and p38 mitogen-activated protein kinase. Hypertension. 2000; 35(1 Pt 2): 188–192.
  8. Liang Q, Wiese RJ, Bueno OF, et al. The transcription factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes. Mol Cell Biol. 2001; 21(21): 7460–7469.
  9. Glennon PE, Kaddoura S, Sale EM, et al. Depletion of mitogen-activated protein kinase using an antisense oligodeoxynucleotide approach downregulates the phenylephrine-induced hypertrophic response in rat cardiac myocytes. Circ Res. 1996; 78(6): 954–961.
  10. Ferguson BS, Harrison BC, Jeong MY, et al. Signal-dependent repression of DUSP5 by class I HDACs controls nuclear ERK activity and cardiomyocyte hypertrophy. Proc Natl Acad Sci U S A. 2013; 110(24): 9806–9811.
  11. Lorenz K, Schmitt JP, Schmitteckert EM, et al. A new type of ERK1/2 autophosphorylation causes cardiac hypertrophy. Nat Med. 2009; 15(1): 75–83.
  12. Thorburn J, Frost JA, Thorburn A. Mitogen-activated protein kinases mediate changes in gene expression, but not cytoskeletal organization associated with cardiac muscle cell hypertrophy. J Cell Biol. 1994; 126(6): 1565–1572.
  13. Thorburn J, McMahon M, Thorburn A. Raf-1 kinase activity is necessary and sufficient for gene expression changes but not sufficient for cellular morphology changes associated with cardiac myocyte hypertrophy. J Biol Chem. 1994; 269(48): 30580–30586.
  14. Silberbach M, Gorenc T, Hershberger RE, et al. Extracellular signal-regulated protein kinase activation is required for the anti-hypertrophic effect of atrial natriuretic factor in neonatal rat ventricular myocytes. J Biol Chem. 1999; 274(35): 24858–24864.
  15. Mathur A, Ma Z, Loskill P, et al. In vitro cardiac tissue models: Current status and future prospects. Adv Drug Deliv Rev. 2016; 96: 203–213.
  16. Vozzi F, Logrand F, Cabiati M, et al. Biomimetic engineering of the cardiac tissue through processing, functionalization, and biological characterization of polyester urethanes. Biomed Mater. 2018; 13(5): 055006.
  17. Ghosh LD, Jain A, Sundaresan NR, et al. Elucidating molecular events underlying topography mediated cardiomyogenesis of stem cells on 3D nanofibrous scaffolds. Mater Sci Eng C Mater Biol Appl. 2018; 88: 104–114.
  18. Molkentin JD, Robbins J. With great power comes great responsibility: using mouse genetics to study cardiac hypertrophy and failure. J Mol Cell Cardiol. 2009; 46(2): 130–136.
  19. Breitenbach T, Lorenz K, Dandekar T. How to steer and control ERK and the ERK signaling cascade exemplified by looking at cardiac insufficiency. Int J Mol Sci. 2019; 20(9).
  20. Lloyd AC. Distinct functions for ERKs? J Biol. 2006; 5(5): 13.
  21. Gerits N, Kostenko S, Moens U. In vivo functions of mitogen-activated protein kinases: conclusions from knock-in and knock-out mice. Transgenic Res. 2007; 16(3): 281–314.
  22. Wang Y. Mitogen-activated protein kinases in heart development and diseases. Circulation. 2007; 116(12): 1413–1423.
  23. Shaul YD, Seger R. The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta. 2007; 1773(8): 1213–1226.
  24. García-Niño WR, Correa F, Rodríguez-Barrena JI, et al. Cardioprotective kinase signaling to subsarcolemmal and interfibrillar mitochondria is mediated by caveolar structures. Basic Res Cardiol. 2017; 112(2): 15.
  25. Nomura S, Satoh M, Fujita T, et al. Cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure. Nat Commun. 2018; 9(1): 4435.
  26. Lu YT, Li LZ, Yang YL, et al. Succinate induces aberrant mitochondrial fission in cardiomyocytes through GPR91 signaling. Cell Death Dis. 2018; 9(6): 672.
  27. Huang CY, Chen JY, Kuo CH, et al. Mitochondrial ROS-induced ERK1/2 activation and HSF2-mediated AT R upregulation are required for doxorubicin-induced cardiotoxicity. J Cell Physiol. 2018; 233(1): 463–475.
  28. Gartz M, Darlington A, Afzal MZ, et al. Exosomes exert cardioprotection in dystrophin-deficient cardiomyocytes via ERK1/2-p38/MAPK signaling. Sci Rep. 2018; 8(1): 16519.
  29. Liu R, Molkentin JD. Regulation of cardiac hypertrophy and remodeling through the dual-specificity MAPK phosphatases (DUSPs). J Mol Cell Cardiol. 2016; 101: 44–49.
  30. Junttila MR, Li SP, Westermarck J. Phosphatase-mediated crosstalk between MAPK signaling pathways in the regulation of cell survival. FASEB J. 2008; 22(4): 954–965.
  31. Hunter JJ, Tanaka N, Rockman HA, et al. Ventricular expression of a MLC-2v-ras fusion gene induces cardiac hypertrophy and selective diastolic dysfunction in transgenic mice. J Biol Chem. 1995; 270(39): 23173–23178.
  32. Zheng M, Dilly K, Dos Santos Cruz J, et al. Sarcoplasmic reticulum calcium defect in Ras-induced hypertrophic cardiomyopathy heart. Am J Physiol Heart Circ Physiol. 2004; 286(1): H424–H433.
  33. Mitchell S, Ota A, Foster W, et al. Distinct gene expression profiles in adult mouse heart following targeted MAP kinase activation. Physiol Genomics. 2006; 25(1): 50–59.
  34. Wei BR, Martin PL, Hoover SB, et al. Capacity for resolution of Ras-MAPK-initiated early pathogenic myocardial hypertrophy modeled in mice. Comp Med. 2011; 61(2): 109–118.
  35. Ramos-Kuri M, Rapti K, Mehel H, et al. Dominant negative Ras attenuates pathological ventricular remodeling in pressure overload cardiac hypertrophy. Biochim Biophys Acta. 2015; 1853(11 Pt A): 2870–2884.
  36. Ho PD, Zechner DK, He H, et al. The Raf-MEK-ERK cascade represents a common pathway for alteration of intracellular calcium by Ras and protein kinase C in cardiac myocytes. J Biol Chem. 1998; 273(34): 21730–21735.
  37. Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. Physiol Rev. 2010; 90(4): 1507–1546.
  38. Wu X, Simpson J, Hong JH, et al. MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1(L613V) mutation. J Clin Invest. 2011; 121(3): 1009–1025.
  39. Passariello CL, Martinez EC, Thakur H, et al. RSK3 is required for concentric myocyte hypertrophy in an activated Raf1 model for Noonan syndrome. J Mol Cell Cardiol. 2016; 93: 98–105.
  40. Harris IS, Zhang S, Treskov I, et al. Raf-1 kinase is required for cardiac hypertrophy and cardiomyocyte survival in response to pressure overload. Circulation. 2004; 110(6): 718–723.
  41. Yin JC, Platt MJ, Tian X, et al. Cellular interplay via cytokine hierarchy causes pathological cardiac hypertrophy in RAF1-mutant Noonan syndrome. Nat Commun. 2017; 8: 15518.
  42. Bueno OF, De Windt LJ, Tymitz KM, et al. The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J. 2000; 19(23): 6341–6350.
  43. Mutlak M, Schlesinger-Laufer M, Haas T, et al. Extracellular signal-regulated kinase (ERK) activation preserves cardiac function in pressure overload induced hypertrophy. Int J Cardiol. 2018; 270: 204–213.
  44. Jochmann S, Elkenani M, Mohamed BA, et al. Assessing the role of extracellular signal-regulated kinases 1 and 2 in volume overload-induced cardiac remodelling. ESC Heart Fail. 2019; 6(5): 1015–1026.
  45. Smorodinsky-Atias K, Goshen-Lago T, Goldberg-Carp A, et al. Intrinsically active variants of Erk oncogenically transform cells and disclose unexpected autophosphorylation capability that is independent of TEY phosphorylation. Mol Biol Cell. 2016; 27(6): 1026–1039.
  46. Lai S, Pelech S. Regulatory roles of conserved phosphorylation sites in the activation T-loop of the MAP kinase ERK1. Mol Biol Cell. 2016; 27(6): 1040–1050.
  47. Ruppert C, Deiss K, Herrmann S, et al. Interference with ERK(Thr188) phosphorylation impairs pathological but not physiological cardiac hypertrophy. Proc Natl Acad Sci U S A. 2013; 110(18): 7440–7445.
  48. Purcell NH, Wilkins BJ, York A, et al. Genetic inhibition of cardiac ERK1/2 promotes stress-induced apoptosis and heart failure but has no effect on hypertrophy in vivo. Proc Natl Acad Sci U S A. 2007; 104(35): 14074–14079.
  49. Kehat I, Davis J, Tiburcy M, et al. Extracellular signal-regulated kinases 1 and 2 regulate the balance between eccentric and concentric cardiac growth. Circ Res. 2011; 108(2): 176–183.
  50. Ulm S, Liu W, Zi M, et al. Targeted deletion of ERK2 in cardiomyocytes attenuates hypertrophic response but provokes pathological stress induced cardiac dysfunction. J Mol Cell Cardiol. 2014; 72: 104–116.
  51. Maillet M, Purcell NH, Sargent MA, et al. DUSP6 (MKP3) null mice show enhanced ERK1/2 phosphorylation at baseline and increased myocyte proliferation in the heart affecting disease susceptibility. J Biol Chem. 2008; 283(45): 31246–31255.
  52. Missinato MA, Saydmohammed M, Zuppo DA, et al. Dusp6 attenuates Ras/MAPK signaling to limit zebrafish heart regeneration. Development. 2018; 145(5).
  53. Liu R, van Berlo JH, York AJ, et al. DUSP8 regulates cardiac ventricular remodeling by altering ERK1/2 signaling. Circ Res. 2016; 119(2): 249–260.
  54. Owens DM, Keyse SM. Differential regulation of MAP kinase signalling by dual-specificity protein phosphatases. Oncogene. 2007; 26(22): 3203–3213.
  55. Casar B, Crespo P. ERK signals: scaffolding scaffolds? Front Cell Dev Biol. 2016; 4: 49.
  56. Song Y, Xu J, Li Y, et al. Cardiac ankyrin repeat protein attenuates cardiac hypertrophy by inhibition of ERK1/2 and TGF-β signaling pathways. PLoS One. 2012; 7(12): e50436.
  57. Zhong L, Chiusa M, Cadar AG, et al. Targeted inhibition of ANKRD1 disrupts sarcomeric ERK-GATA4 signal transduction and abrogates phenylephrine-induced cardiomyocyte hypertrophy. Cardiovasc Res. 2015; 106(2): 261–271.
  58. Sbroggiò M, Carnevale D, Bertero A, et al. IQGAP1 regulates ERK1/2 and AKT signalling in the heart and sustains functional remodelling upon pressure overload. Cardiovasc Res. 2011; 91(3): 456–464.
  59. Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol. 2005; 6(11): 827–837.
  60. Himpel S, Tegge W, Frank R, et al. Specificity determinants of substrate recognition by the protein kinase DYRK1A. J Biol Chem. 2000; 275(4): 2431–2438.
  61. Liang Y, Sheikh F. Scaffold proteins regulating extracellular regulated kinase function in cardiac hypertrophy and disease. Front Pharmacol. 2016; 7: 37.
  62. Tarone G, Sbroggiò M, Brancaccio M. Key role of ERK1/2 molecular scaffolds in heart pathology. Cell Mol Life Sci. 2013; 70(21): 4047–4054.
  63. Zong J, Zhang Dp, Zhou H, et al. Baicalein protects against cardiac hypertrophy through blocking MEK-ERK1/2 signaling. J Cell Biochem. 2013; 114(5): 1058–1065.
  64. Xiong X, Yang X, Duan L, et al. Traditional Chinese medicine suppresses left ventricular hypertrophy by targeting extracellular signal-regulated kinases signaling pathway in spontaneously hypertensive rats. Sci Rep. 2017; 7: 42965.
  65. Sun S, Kee HJ, Jin Li, et al. Gentisic acid attenuates pressure overload-induced cardiac hypertrophy and fibrosis in mice through inhibition of the ERK1/2 pathway. J Cell Mol Med. 2018; 22(12): 5964–5977.
  66. Li C, Chen Z, Yang H, et al. Selumetinib, an Oral Anti-Neoplastic Drug, May Attenuate Cardiac Hypertrophy via Targeting the ERK Pathway. PLoS One. 2016; 11(7): e0159079.
  67. Sala V, Gallo S, Gatti S, et al. Cardiac concentric hypertrophy promoted by activated Met receptor is mitigated in vivo by inhibition of Erk1,2 signalling with Pimasertib. J Mol Cell Cardiol. 2016; 93: 84–97.
  68. Kodama H, Fukuda K, Pan J, et al. Significance of ERK cascade compared with JAK/STAT and PI3-K pathway in gp130-mediated cardiac hypertrophy. Am J Physiol Heart Circ Physiol. 2000; 279(4): H1635–H1644.
  69. Yue TL, Gu JL, Wang C, et al. Extracellular signal-regulated kinase plays an essential role in hypertrophic agonists, endothelin-1 and phenylephrine-induced cardiomyocyte hypertrophy. J Biol Chem. 2000; 275(48): 37895–37901.
  70. Lee CY, Lee J, Seo HH, et al. TAK733 attenuates adrenergic receptor-mediated cardiomyocyte hypertrophy via inhibiting ErkThr188 phosphorylation. Clin Hemorheol Microcirc. 2019; 72(2): 179–187.
  71. Woodard GE, Jardín I, Berna-Erro A, et al. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. Int Rev Cell Mol Biol. 2015; 317: 97–183.
  72. Liu Yu, Huang He, Zhang Y, et al. Regulator of G protein signaling 3 protects against cardiac hypertrophy in mice. J Cell Biochem. 2014; 115(5): 977–986.
  73. Li H, He C, Feng J, et al. Regulator of G protein signaling 5 protects against cardiac hypertrophy and fibrosis during biomechanical stress of pressure overload. Proc Natl Acad Sci U S A. 2010; 107(31): 13818–13823.
  74. Miao R, Lu Y, Xing X, et al. Regulator of G-protein signaling 10 negatively regulates cardiac remodeling by blocking mitogen-activated protein kinase-extracellular signal-regulated protein kinase 1/2 signaling. Hypertension. 2016; 67(1): 86–98.
  75. Huang J, Chen L, Yao Y, et al. Pivotal role of regulator of G-protein signaling 12 in pathological cardiac hypertrophy. Hypertension. 2016; 67(6): 1228–1236.
  76. Li Y, Tang XH, Li XH, et al. Regulator of G protein signalling 14 attenuates cardiac remodelling through the MEK-ERK1/2 signalling pathway. Basic Res Cardiol. 2016; 111(4): 47.