#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Rho-associated kinase and zipper-interacting protein kinase, but not myosin light chain kinase, are involved in the regulation of myosin phosphorylation in serum-stimulated human arterial smooth muscle cells


Autoři: Jing-Ti Deng aff001;  Sabreena Bhaidani aff001;  Cindy Sutherland aff001;  Justin A. MacDonald aff001;  Michael P. Walsh aff001
Působiště autorů: Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada aff001
Vyšlo v časopise: PLoS ONE 14(12)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0226406

Souhrn

Myosin regulatory light chain (LC20) phosphorylation plays an important role in vascular smooth muscle contraction and cell migration. Ca2+/calmodulin-dependent myosin light chain kinase (MLCK) phosphorylates LC20 (its only known substrate) exclusively at S19. Rho-associated kinase (ROCK) and zipper-interacting protein kinase (ZIPK) have been implicated in the regulation of LC20 phosphorylation via direct phosphorylation of LC20 at T18 and S19 and indirectly via phosphorylation of MYPT1 (the myosin targeting subunit of myosin light chain phosphatase, MLCP) and Par-4 (prostate-apoptosis response-4). Phosphorylation of MYPT1 at T696 and T853 inhibits MLCP activity whereas phosphorylation of Par-4 at T163 disrupts its interaction with MYPT1, exposing the sites of phosphorylation in MYPT1 and leading to MLCP inhibition. To evaluate the roles of MLCK, ROCK and ZIPK in these phosphorylation events, we investigated the time courses of phosphorylation of LC20, MYPT1 and Par-4 in serum-stimulated human vascular smooth muscle cells (from coronary and umbilical arteries), and examined the effects of siRNA-mediated MLCK, ROCK and ZIPK knockdown and pharmacological inhibition on these phosphorylation events. Serum stimulation induced rapid phosphorylation of LC20 at T18 and S19, MYPT1 at T696 and T853, and Par-4 at T163, peaking within 30–120 s. MLCK knockdown or inhibition, or Ca2+ chelation with EGTA, had no effect on serum-induced LC20 phosphorylation. ROCK knockdown decreased the levels of phosphorylation of LC20 at T18 and S19, of MYPT1 at T696 and T853, and of Par-4 at T163, whereas ZIPK knockdown decreased LC20 diphosphorylation, but increased phosphorylation of MYPT1 at T696 and T853 and of Par-4 at T163. ROCK inhibition with GSK429286A reduced serum-induced phosphorylation of LC20 at T18 and S19, MYPT1 at T853 and Par-4 at T163, while ZIPK inhibition by HS38 reduced only LC20 diphosphorylation. We also demonstrated that serum stimulation induced phosphorylation (activation) of ZIPK, which was inhibited by ROCK and ZIPK down-regulation and inhibition. Finally, basal phosphorylation of LC20 in the absence of serum stimulation was unaffected by MLCK, ROCK or ZIPK knockdown or inhibition. We conclude that: (i) serum stimulation of cultured human arterial smooth muscle cells results in rapid phosphorylation of LC20, MYPT1, Par-4 and ZIPK, in contrast to the slower phosphorylation of kinases and other proteins involved in other signaling pathways (Akt, ERK1/2, p38 MAPK and HSP27), (ii) ROCK and ZIPK, but not MLCK, are involved in serum-induced phosphorylation of LC20, (iii) ROCK, but not ZIPK, directly phosphorylates MYPT1 at T853 and Par-4 at T163 in response to serum stimulation, (iv) ZIPK phosphorylation is enhanced by serum stimulation and involves phosphorylation by ROCK and autophosphorylation, and (v) basal phosphorylation of LC20 under serum-free conditions is not attributable to MLCK, ROCK or ZIPK.

Klíčová slova:

MAPK signaling cascades – Myosins – Phosphorylation – Small interfering RNAs – Smooth muscle cells – Smooth muscles – Stoichiometry – AKT signaling cascade


Zdroje

1. Hartshorne DJ. Biochemistry of the contractile process in smooth muscle. In: Johnson LR, editor. Physiology of the gastrointestinal tract. New York: Raven Press; 1987. pp. 423–482.

2. Allen BG, Walsh MP (1994) The biochemical basis of the regulation of smooth-muscle contraction. Trends Biochem Sci 19: 362–368. doi: 10.1016/0968-0004(94)90112-0 7985229

3. Gallagher PJ, Herring PB, Stull JT (1997) Myosin light chain kinases. J Muscle Res Cell Motil 18: 1–16. doi: 10.1023/a:1018616814417 9147985

4. Kamm KE, Stull JT (2001) Dedicated myosin light chain kinases with diverse cellular functions. J Biol Chem 276: 4527–4530. doi: 10.1074/jbc.R000028200 11096123

5. Ikebe M, Hartshorne DJ (1985) Phosphorylation of smooth muscle myosin at two distinct sites by myosin light chain kinase. J Biol Chem. 260 10027–10031. 3839510

6. Ikebe M, Hartshorne DJ, Elzinga M (1986) Identification, phosphorylation, and dephosphorylation of a second site for myosin light chain kinase on the 20,000-Dalton light chain of smooth muscle myosin. J Biol Chem 261: 36–39. 3079756

7. Mita M, Yanagihara H, Hishinuma S, Saito M, Walsh MP (2002) Membrane depolarization-induced contraction of rat caudal arterial smooth muscle involves Rho-associated kinase. Biochem J 364: 431–440. doi: 10.1042/BJ20020191 12023886

8. Wilson DP, Sutherland C, Walsh, MP (2002) Ca2+ activation of smooth muscle contraction. Evidence for the involvement of calmodulin that is bound to the Triton-insoluble fraction even in the absence of Ca2+. J Biol Chem 277: 2186–2192. doi: 10.1074/jbc.M110056200 11707462

9. Takeya K, Wang X, Kathol I, Loutzenhiser K, Loutzenhiser R, et al. (2014) Endothelin-1, but not angiotensin II, induces afferent arteriolar myosin diphosphorylation as a potential contributor to prolonged vasoconstriction. Kidney Int 87: 370–381. doi: 10.1038/ki.2014.284 25140913

10. Sutherland C, Walsh MP (2012) Myosin regulatory light chain diphosphorylation slows relaxation of arterial smooth muscle. J Biol Chem 287: 24064–24076. doi: 10.1074/jbc.M112.371609 22661704

11. Sasaki Y, Iwata K, Sasaki Y (1990) Concanavalin A- and fetal-calf-serum-induced rounding and myosin light chain phosphorylation in cultured smooth muscle cells. J Cell Physiol 144: 183–189. doi: 10.1002/jcp.1041440202 2116423

12. Sakurada K, Seto M, Sasaki Y (1998) Dynamics of myosin light chain phosphorylation at Ser19 and Thr18/Ser19 in smooth muscle cells in culture. Am J Physiol Cell Physiol 274: C1563–C1572.

13. Hirano M, Hirano K (2016) Myosin di-phosphorylation and peripheral actin bundle formation as initial events during endothelial barrier disruption. Sci Reports 6: 20989.

14. Weber LP, Van Lierop JE, Walsh MP (1999) Ca2+-independent phosphorylation of myosin in rat caudal artery and chicken gizzard myofilaments. J Physiol 516: 805–824. doi: 10.1111/j.1469-7793.1999.0805u.x 10200427

15. Amano M, Ito M, Kimura K, Fukata Y, Chihara K, et al. (1996) Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase). J Biol Chem 271: 20246–20249. doi: 10.1074/jbc.271.34.20246 8702756

16. Murata-Hori M, Suizu F, Iwasaki T, Kikuchi A, Hosoya H (1999) ZIP kinase identified as a novel myosin regulatory light chain kinase in HeLa cells. FEBS Lett 451: 81–84. doi: 10.1016/s0014-5793(99)00550-5 10356987

17. Murata-Hori M, Fukuta Y, Ueda K, Iwasaki T, Hosoya H (2001) HeLa ZIP kinase induces diphosphorylation of myosin II regulatory light chain and reorganization of actin filaments in nonmuscle cells. Oncogene 20: 8175–8183. doi: 10.1038/sj.onc.1205055 11781833

18. Niiro N, Ikebe M (2001) Zipper-interacting protein kinase induces Ca2+ free smooth muscle contraction via myosin light chain phosphorylation. J Biol Chem 276: 29567–29574. doi: 10.1074/jbc.M102753200 11384979

19. Komatsu S, Ikebe M (2004) ZIP kinase is responsible for the phosphorylation of myosin II and necessary for cell motility in mammalian fibroblasts. J Cell Biol 165: 243–254. doi: 10.1083/jcb.200309056 15096528

20. Moffat L, Brown S, Grassie M, Ulke-Lemée A, Williamson L, et al. (2011) Chemical genetics of zipper-interacting protein kinase reveal myosin light chain as a bone fide substrate in permeabilized arterial smooth muscle. J Biol Chem 286: 36978–36991. doi: 10.1074/jbc.M111.257949 21880706

21. Deng JT, Van Lierop JE, Sutherland C, Walsh MP (2001) Ca2+-independent smooth muscle contraction. A novel function for integrin-linked kinase. J Biol Chem 276: 16365–16373. doi: 10.1074/jbc.M011634200 11278951

22. Wilson DP, Sutherland C, Borman MA, Deng JT, MacDonald JA, et al. (2005a) Integrin-linked kinase is responsible for Ca2+-independent myosin diphosphorylation and contraction of vascular smooth muscle. Biochem J 392: 641–648. doi: 10.1042/BJ20051173 16201970

23. Yamashiro S, Totsukawa G, Yamakita Y, Sasaki Y, Madaule P, et al. (2003) Citron kinase, a Rho-dependent kinase, induces di-phosphorylation of regulatory light chain of myosin II. Mol Biol Cell 14: 1745–1756. doi: 10.1091/mbc.E02-07-0427 12802051

24. Tamura M, Nakao H, Yoshizaki H, Shiratsuchi M, Shigyo H, et al. (2005) Development of specific Rho-kinase inhibitors and their clinical application. Biochim Biophys Acta 1754: 245–252. doi: 10.1016/j.bbapap.2005.06.015 16213195

25. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, et al. (2007) The selectivity of protein kinase inhibitors: a further update. Biochem J 408: 297–315. doi: 10.1042/BJ20070797 17850214

26. Nichols RJ, Dzamko N, Hutti JE, Cantley LC, Deak M, et al. (2009) Substrate specificity and inhibitors of LRRK2, a protein kinase mutated in Parkinson’s disease. Biochem J 424: 47–60. doi: 10.1042/BJ20091035 19740074

27. Carlson DA, Franke AS, Weitzel DH, Speer BL, Hughes PF, et al. (2013) Fluorescence linked enzyme chemoproteomic strategy for discovery of a potent and selective DAPK1 and ZIPK inhibitor. ACS Chem Biol 8: 2715–2723. doi: 10.1021/cb400407c 24070067

28. MacDonald JA, Sutherland C, Carlson DA, Bhaidani S, Al-Ghabkari A, et al. (2016) A small molecule pyrazolo[3,4-d]pyrimidinone inhibitor of zipper-interacting protein kinase suppresses calcium sensitization of vascular smooth muscle. Mol Pharmacol 89: 105–117. doi: 10.1124/mol.115.100529 26464323

29. Al-Ghabkari A, Deng JT, McDonald PC, Dedhar S, Alshehri M, et al. (2016) A novel inhibitory effect of oxazol-5-one compounds on ROCKII signaling in human coronary artery vascular smooth muscle cells. Sci Rep 6: 32118. doi: 10.1038/srep32118 27573465

30. Okamoto M, Takayama K, Shimizu T, Ishida K, Takahashi O, et al. (2009) Identification of death-associated protein kinase inhibitors using structure-based virtual screening. J Med Chem 52: 7323–7327. doi: 10.1021/jm901191q 19877644

31. Davies SP, Reddy H, Caivano M, Cohen P (2000) Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351: 95–105. doi: 10.1042/0264-6021:3510095 10998351

32. Bain J, McLauchlan H, Elliott M, Cohen P (2003) The specificities of protein kinase inhibitors: an update. Biochem J 371: 199–204. doi: 10.1042/BJ20021535 12534346

33. Deng JT, Wang XL, Chen YX, O’Brien ER, Gui Y, et al. (2015) The effects of knockdown of Rho-associated kinase 1 and zipper-interacting protein kinase in gene expression and function in cultured human arterial smooth muscle cells. PLoS ONE 10(2): e0116969. doi: 10.1371/journal.pone.0116969 25723491

34. Paul ER, Ngai PK, Walsh MP, Gröschel-Stewart U (1995) Embryonic chicken gizzard: expression of the smooth muscle regulatory proteins caldesmon and myosin light chain kinase. Cell Tissue Res 279: 331–337. doi: 10.1007/bf00318489 7895272

35. Kinoshita E, Kinoshika-Kikuta E, Takiyama K, Koike T (2006) Phosphate-binding tag, a new tool to visualize phosphorylated proteins. Mol Cell Proteomics 5: 749–757. doi: 10.1074/mcp.T500024-MCP200 16340016

36. Shiojima I, Walsh K (2002) Role of Akt signaling in vascular homeostasis and angiogenesis. Circ Res 90: 1243–1250. doi: 10.1161/01.res.0000022200.71892.9f 12089061

37. Morello F, Perino A, Hirsch E (2009) Phosphoinositide 3-kinase signalling in the vascular system. Cardiovasc Res 82: 261–271. doi: 10.1093/cvr/cvn325 19038971

38. Risso G, Blaustein M, Pozzi B, Mammi P, Srebrow A (2015) Akt/PKB: one kinase, many modifications. Biochem J 468: 203–214. doi: 10.1042/BJ20150041 25997832

39. Johnson GL, Lapadat R (2002) Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science 298: 1911–1912. doi: 10.1126/science.1072682 12471242

40. Huang C, Jacobson K, Schaller MD (2004) MAP kinases and cell migration. J Cell Sci 117: 4619–4628. doi: 10.1242/jcs.01481 15371522

41. Herlaar E, Brown Z (1999) p38 MAPK signaling cascades in inflammatory disease. Mol Med Today 5: 439–447. doi: 10.1016/s1357-4310(99)01544-0 10498912

42. Kostenko S, Moens U (2009) Heat shock protein 27 phosphorylation: kinases, phosphatases, functions and pathology. Cell Mol Life Sci 66: 3289–3307. doi: 10.1007/s00018-009-0086-3 19593530

43. Salinthone S, Tyagi M, Gerthoffer WT (2008) Small heat shock proteins in smooth muscle. Pharmacol Ther 119: 44–54. doi: 10.1016/j.pharmthera.2008.04.005 18579210

44. Wilson DP, Susnjar M, Kiss E, Sutherland C, Walsh MP (2005b) Thromboxane A2-induced contraction of rat caudal arterial smooth muscle involves activation of Ca2+ entry and Ca2+ sensitization: Rho-associated kinase-mediated phosphorylation of MYPT1 at Thr-855 but not Thr-697. Biochem J 389: 763–774. doi: 10.1042/BJ20050237 15823093

45. Graves PR, Winkfield KM, Haystead TAJ (2005) Regulation of zipper-interacting protein kinase activity in vitro and in vivo by multisite phosphorylation. J Biol Chem 280: 9363–9374. doi: 10.1074/jbc.M412538200 15611134

46. Somlyo AP, Somlyo AV (2000) Signal transduction by G-proteins, Rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J Physiol 522: 177–185. doi: 10.1111/j.1469-7793.2000.t01-2-00177.x 10639096

47. MacDonald JA, Walsh MP, Cole WC. Calcium sensitization in smooth muscle involving regulation of myosin light chain phosphatase activity. In: Trebak M and Earley S, editors. Signal transduction and smooth muscle. Boca Raton, Florida: CRC Press; 2018. pp. 123–154.

48. Fukata Y, Amano M, Kaibuchi K (2001) Rho-Rho-kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci 22: 32–39. doi: 10.1016/s0165-6147(00)01596-0 11165670

49. Riento K, Ridley AJ (2003) ROCKs: Multifunctional kinases in cell behaviour. Nat Rev Cell Biol 4: 446–456.

50. Haystead TA (2005) ZIP kinase, a key regulator of myosin protein phosphatase 1. Cell Signal 17: 1313–1322. doi: 10.1016/j.cellsig.2005.05.008 16005610

51. Ihara E, MacDonald JA (2007) The regulation of smooth muscle contractility by zipper-interacting protein kinase. Can J Physiol Pharmacol 85: 79–87. doi: 10.1139/y06-103 17487247

52. Usui T, Okada M, Yamawaki H (2013) Zipper-interacting protein kinase (ZIPK): function and signaling. Apoptosis 19: 387–391.

53. Kureishi Y, Kobayashi S, Amano M, Kimura K, Kanaide H, et al. (1997) Rho-associated kinase directly induces smooth muscle contraction through myosin light chain phosphorylation. J Biol Chem 272: 12257–12260. doi: 10.1074/jbc.272.19.12257 9139666

54. Borman MA, MacDonald JA, Murányi A, Hartshorne DJ, Haystead TAJ. (2002) Smooth muscle myosin phosphatase-associated kinase induces Ca2+ sensitization via myosin phosphatase inhibition. J Biol Chem 277: 23441–23446. doi: 10.1074/jbc.M201597200 11976330

55. Al-Ghabkari A, Moffat LD, Walsh MP, MacDonald JA (2018) Validation of chemical genetics for the study of zipper-interacting protein kinase signaling. Proteins 86: 1211–1217. doi: 10.1002/prot.25607 30381843

56. Kitazawa T, Kobayashi S, Horiuti K, Somlyo AV, Somlyo AP (1989) Receptor-coupled, permeabilized smooth muscle. Role of the phosphatidylinositol cascade, G-proteins, and modulation of the contractile response to Ca2+. J Biol Chem 264: 5339–5342. 2494163

57. Kitazawa T, Somlyo AP (1991) Modulation of Ca2+ sensitivity by agonists in smooth muscle. Adv Exp Biol Med 304: 97–109.

58. Kitazawa T, Masuo M, Somlyo AP (1991) G protein-mediated inhibition of myosin light-chain phosphatase in vascular smooth muscle. Proc Natl Acad Sci USA 88: 9307–9310. doi: 10.1073/pnas.88.20.9307 1656467

59. Kubota Y, Nomura M, Kamm KE, Mumby MC, Stull JT (1992) GTP gamma S-dependent regulation of smooth muscle contractile elements. Am J Physiol Cell Physiol 262: C405–C410.

60. Masuo M, Reardon S, Ikebe M, Kitazawa T (1994) A novel mechanism for the Ca2+-sensitizing effect of protein kinase C on vascular smooth muscle: Inhibition of myosin light chain phosphatase. J Gen Physiol 104: 265–286. doi: 10.1085/jgp.104.2.265 7807049

61. Iizuka Y, Yoshii A, Samizo K, Tsukagoshi H, Ishizuka T, et al. (1999) A major role for the Rho-associated coiled coil forming protein kinase in G-protein-mediated Ca2+ sensitization through inhibition of myosin phosphatase in rabbit trachea. Br J Pharmacol 128: 925–933. doi: 10.1038/sj.bjp.0702864 10556927

62. Swärd K, Dreja K, Susnjar M, Hellstrand P, Hartshorne DJ, et al. (2000) Inhibition of Rho-associated kinase blocks agonist-induced Ca2+ sensitization of myosin phosphorylation and force in guinea pig ileum. J Physiol 522: 33–49. doi: 10.1111/j.1469-7793.2000.0033m.x 10618150

63. Totsukawa G, Yamakita Y, Yamashiro S, Hartshorne DJ, Sasaki Y, et al. (2000) Distinct roles of ROCK (Rho-kinase) and MLCK in spatial regulation of MLC phosphorylation for assembly of stress fibers and focal adhesions in 3T3 fibroblasts. J Cell Biol 150: 797–806. doi: 10.1083/jcb.150.4.797 10953004

64. Kassianidou E, Hughes JH, Kumar S (2017) Activation of ROCK and MLCK tunes regional stress fiber formation and mechanics via preferential myosin light chain phosphorylation. Mol Biol Cell 28: 3832–3843. doi: 10.1091/mbc.E17-06-0401 29046396

65. Kawano Y, Fukata Y, Oshiro N, Amano M, Nakamura T, et al. (1999) Phosphorylation of myosin-binding subunit (MBS) of myosin phosphatase by Rho-kinase in vivo. J Cell Biol 147: 1023–1038. doi: 10.1083/jcb.147.5.1023 10579722

66. Feng J, Ito M, Ichikawa K, Isaka N, Nishikawa N, et al. (1999) Inhibitory phosphorylation site for Rho-associated kinase on smooth muscle myosin phosphatase. J Biol Chem 274: 37385–37390. doi: 10.1074/jbc.274.52.37385 10601309

67. Katoh K, Kano Y, Amano M, Onishi H, Kaibuchi K, et al. (2001) Rho kinase-mediated contraction of isolated stress fibers. J Cell Biol 153: 569–584. doi: 10.1083/jcb.153.3.569 11331307

68. Velasco G, Armstrong C, Morrice N, Frame S, Cohen P (2002) Phosphorylation of the regulatory subunit of smooth muscle protein phosphatase 1M at Thr850 induces its dissociation from myosin. FEBS Lett 527: 101–104. doi: 10.1016/s0014-5793(02)03175-7 12220642

69. Ito M, Nakano T, Erdődi F, Hartshorne DJ (2004) Myosin phosphatase: structure, regulation and function. Mol Cell Biochem 259: 197–209. doi: 10.1023/b:mcbi.0000021373.14288.00 15124925

70. Walsh MP, MacDonald JA (2018) Regulation of smooth muscle myosin light chain phosphatase by multisite phosphorylation of the myosin targeting subunit, MYPT1. Cardiovasc Hematol Disord Drug Targets 18: 4–13. doi: 10.2174/1871529X18666180326120638 29577868

71. Ren X-D, Wang R, Li Q, Kahek LAF, Kaibuchi K, et al. (2004) Disruption of Rho signal transduction upon cell detachment. J Cell Sci 117: 3511–3518. doi: 10.1242/jcs.01205 15226371

72. Vetterkind S, Lee E, Sundberg E, Poythress RH, Tao TC, et al. (2010) Par-4: a new activator of myosin phosphatase. Mol Biol Cell 21: 1214–1224. doi: 10.1091/mbc.E09-08-0711 20130087

73. MacDonald JA, Moffat LD, Al-Ghabkari A, Sutherland C, Walsh MP (2013) Prostate-apoptosis response-4 phosphorylation in vascular smooth muscle. Arch Biochem Biophys 535: 84–90. doi: 10.1016/j.abb.2012.11.009 23219599

74. Kawai T, Matsunoto M, Takeda K, Sanjo H, Akira S (1998) ZIPK, a novel serine/threonine kinase which mediates apoptosis. Mol Cell Biol 18: 1642–1651. doi: 10.1128/mcb.18.3.1642 9488481

75. Shani G, Marash L, Gozuacik D, Bialik S, Teitelbaum L, et al. (2004) Death-associated protein kinase phosphorylates ZIP kinase, forming a unique kinase hierarchy to activate its cell death functions. Mol Cell Biol 24: 8611–8626. doi: 10.1128/MCB.24.19.8611-8626.2004 15367680

76. Usui T, Sakatsume T, Nijima R, Otani K, Kazama K, et al. (2014) Death-associated protein kinase 3 mediates vascular structural remodelling via stimulating smooth muscle cell proliferation and migration. Clin Sci 127: 539–548. doi: 10.1042/CS20130591 24814693

77. Sato N, Kamada N, Muromoto R, Kawai T, Sugiyama K, et al. (2006) Phosphorylation of threonine-265 in zipper-interacting protein kinase plays an important role in its activity and is induced by IL-6 family cytokines. Immunol Lett 103: 127–134. doi: 10.1016/j.imlet.2005.10.015 16325270

78. Hagerty L, Weitzel DH, Chambers J, Fortner CN, Brush MH, et al. (2007) ROCK1 phosphorylates and activates zipper-interacting protein kinase. J Biol Chem 282: 4884–4893. doi: 10.1074/jbc.M609990200 17158456

79. Hamao K, Ono T, Matsushita M, Hosoya H (2019) ZIP kinase phosphorylated and activated by Rho kinase/ROCK contributes to cytokinesis in mammalian cultures cells. Exp Cell Res doi: 10.1016/j.yexcr.2019.111707 31693874

80. Chamley-Campbell J, Campbell GR, Ross R (1979) The smooth muscle cell in culture. Physiol Rev 59: 1–61. doi: 10.1152/physrev.1979.59.1.1 108688

81. Gomez D, Owens GK (2016) Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res 95: 156–164.

82. Grassie ME, Sutherland C, Ulke-Lemée A, Chappellaz M, Kiss E, et al. (2012) Cross-talk between Rho-associated kinase and cyclic nucleotide-dependent kinase signaling pathways in the regulation of smooth muscle myosin light chain phosphatase. J Biol Chem 287: 36356–36369. doi: 10.1074/jbc.M112.398479 22948155

83. Walsh M. P. (2011) Vascular smooth muscle myosin light chain diphosphorylation: mechanism, function and pathological implications. IUBMB Life 63: 987–1000. doi: 10.1002/iub.527 21990256

84. Sutherland C, MacDonald JA, Walsh MP (2016) Analysis of phosphorylation of the myosin-targeting subunit of myosin light chain phosphatase by Phos-tag SDS-PAGE. Am J Physiol Cell Physiol 310: C681–C691. doi: 10.1152/ajpcell.00327.2015 26864694


Článek vyšel v časopise

PLOS One


2019 Číslo 12
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#