Notch signal pathway - therapeutic target for regulation of reparative processes in the heart


Cite item

Full Text

Abstract

Notch signaling pathway is a universal regulator of cell fate in embryogenesis and in maintaining the cell homeostasis of adult tissue. Through local cell-cell interactions, he controls neighboring cells behavior and determines their capacity for self-renewal, growth, survival, differentiation, and apoptosis. Recent studies have shown that the control of regenerative processes in the heart is also carried out with the participation of Notch system. At the heart of Notch regulates migration bone marrow progenitors and stimulates the proliferation of cardiomyocytes, cardiac progenitor cell activity, limits cardiomyocyte hypertrophy and fibrosis progression and stimulates angiogenesis. Notch signaling pathway may be regarded as a very promising target for the development of drugs for the stimulation of regeneration in the myocardium.

About the authors

K V Dergilev

National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation

Email: doctorkote@gmail.com
к.м.н., в.н.с. лаб. ангиогенеза НМИЦ кардиологии Moscow, Russia

E S Zubkova

National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation

н.с. лаб. ангиогенеза НМИЦ кардиологии Moscow, Russia

I B Beloglazova

National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation

к.б.н., с.н.с. лаб. ангиогенеза НМИЦ кардиологии Moscow, Russia

M Yu Menshikov

National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation

к.б.н., в.н.с. лаб. ангиогенеза НМИЦ кардиологии Moscow, Russia

E V Parfyonova

National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation; M.V. Lomonosov Moscow State Universit

д.м.н., проф., директор Института экспериментальной кардиологии НМИЦ кардиологии, руководитель лаб. постгеномных технологий в медицине фак-та фундаментальной медицины МГУ им. М.В. Ломоносова Moscow, Russia

References

  1. Porrello E.R, Mahmoud A.I, Simpson E, Johnson B.A, Grinsfelder D, Canseco D, Mammen P.P, Rothermel B.A, Olson E.N, Sadek H.A. Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family. Proc Natl Acad Sci USA. 1996;110(1):187-92. doi: 10.1073/pnas.1208863110
  2. De la Pompa J.L, Epstein J.A. Coordinating tissue interactions: Notch signaling in cardiac development and disease. Dev Cell. 2012;22(2):244-54. doi: 10.1016/j.devcel.2012.01.014
  3. Luxán G, D'Amato G, Mac Grogan D, de la Pompa J.L. Endocardial Notch Signaling in Cardiac Development and Disease. Circ Res. 2016;118(1):1-18. doi: 10.1161/CIRCRESAHA.115.305350
  4. High F.A, Epstein J.A. The multifaceted role of Notch in cardiac development and disease. Nat Rev Genet. 2008;9(1):49-61. doi: 10.1038/nrg2279
  5. Weinmaster G, Roberts V.J, Lemke G. Notch2: a second mammalian Notch gene. Development. 1992;116(4):931-41.
  6. Del Amo F.F, Smith D.E, Swiatek P.J, Gendron-Maguire M, Greenspan R.J, McMahon A.P, Gridley T. Expression pattern of Motch, a mouse homolog of Drosophila Notch, suggests an important role in early postimplantation mouse development. Development. 1992;115:737-44.
  7. Lardelli M, Dahlstrand J, Lendahl U. The novel Notch homologue mouse Notch3 lacks specific epidermal growth factor repeats and is expressed in proliferating neuroepithelium. Mech Dev. 1994;46(2):123-36.
  8. Uyttendaele H, Marazzi G, Wu G, Yan Q, Sassoon D, Kitajewski J. Notch4/int-3, a mammary proto oncogene, is an endothelial cell - specific mammalian Notch gene. Development. 1996;122(7):2251-9.
  9. Bettenhausen B, Hrabe de Angelis M, Simon D, Guenet D, Gossler A. Transient and restricted expression during mouse embryogenesis of Dll1, a murine gene closely related to Drosophila delta. Development. 1995;121(8):2407-18.
  10. Dunwoodie S.L, Henrique D, Harrison S.M, Beddington R.S. Mouse Dll3: a novel divergent Delta gene which may complement the function of other Delta homologues during early pattern formation in the mouse embryo. Development. 1997;124(16):3065-76.
  11. Krebs L.T, Xue Y, Norton C.R, Shutter J.R, Maguire M, Sundberg J.P, Gallahan D, Closson V, Kitajewski J, Callahan R, Smith G.H, Stark K.L, Gridley T. Notch signaling is essential for vascular morphogenesis in mice. Genes Dev. 2000;14:1343-52. doi: 10.1101/gad.14.11.1343
  12. Lindsell C.E, Shawber C.J, Boulter J, Weinmaster G. Jagged: a mammalian ligand that activates Notch1. Cell. 1995;80(6):909-17.
  13. Shawber C, Boulter J, Lindsell C.E, Weinmaster G. Jagged2: a serrate like gene expressed during rat embryogenesis. Dev Biol. 1996;180(1):370-6. doi: 10.1006/dbio.1996.0310
  14. D'Souza B, Meloty-Kapella L, Weinmaster G. Canonical and Non-Canonical Notch Ligands. Curr Top Develop Biol. 2010;92:73-129. doi: 10.1016/S0070-2153(10)92003-6
  15. Hori K, Sen A, Artavanis-Tsakonas S. Notch Signaling at a Glance. J Cell Sci. 2013;126(10):2135-40. doi: 10.1242/jcs.127308
  16. Tamura K, Taniguchi Y, Minoguchi S, Sakai T, Tun T, Furukawa T, Honjo T. Physical interaction between a novel domain of the receptor Notch and the transcription factor RBP-J kappa/Su(H). Curr Biol. 1995;5(12):1416-23.
  17. Blank V, Kourilsky P, Israel A. NF-kappa B and related proteins: Rel/dorsal homologies meet ankyrin - like repeats. Trends Biochem Sci. 1992;17(4):135-40. doi: 10.1016/0968-0004(92)90321-Y
  18. Rechsteiner M. Regulation of enzyme levels by proteolysis: the role of pest regions. Adv Enzyme Regul. 1988;27:135-51.
  19. Chillakuri C.R, Sheppard D, Le S.M, Handford P.A. Notch Receptor - ligand Binding and Activation: Insights from Molecular Studies. Semin Cell Develop Biol. 2012;23(4):421-8. doi: 10.1016/j.semcdb.2012.01.009
  20. Komatsu H, Chao M.Y, Larkins-Ford J, Corkins M.E, Somers G.A, Tucey T, Dionne H.M, White J.Q, Wani K, Boxem M, Hart A.C. OSM-11 facilitates LIN-12 Notch signaling during Caenorhabditis elegans vulval development. PLoS Biol. 2008;6(8):196-8. doi: 10.1371/journal.pbio.0060196
  21. La Foya B, Munroe J.A, Mia M.M, Detweiler M.A, Crow J.J, Wood T, Roth S, Sharma B, Albig A.R. Notch: A multi - functional integrating system of microenvironmental signals. Dev Biol. 2016;418(2):227-41. doi: 10.1016/j.ydbio.2016.08.023
  22. Guruharsha K.G, Kankel M.W, Artavanis-Tsakonas S. The Notch Signalling System: Recent Insights into the Complexity of a Conserved Pathway. Nat Rev Genet. 2012;13(9):654-66. doi: 10.1038/nrg3272
  23. Sprinzak D, Lakhanpal A, Lebon L, Santat L.A, Fontes M.E, Anderson G.A, Garcia-Ojalvo J, Elowitz M.B. Cis Interactions between Notch and Delta Generate Mutually Exclusive Signaling States. Nature. 2010;465(7294):86-90. doi: 10.1038/nature08959
  24. Wang M.M. Notch Signaling and Notch Signaling Modifiers. Int J Biochem Cell Biol. 2011;43(11):1550-62. doi: 10.1016/j.biocel.2011.08.005
  25. Mumm J.S, Schroeter E.H, Saxena M.T, Griesemer A, Tian X, Pan D.J, Ray W.J, Kopan R. A ligand - induced extracellular cleavage regulates gamma - secretase - like proteolytic activation of Notch1. Mol Cell. 2000;5(2):197-206. doi: 10.1016/S1097-2765(00)80416-5
  26. Toonen J.A, Ronchetti A, Sidjanin D.J. A Disintegrin and Metalloproteinase10 (ADAM10) Regulates NOTCH Signaling during Early Retinal Development. PLoS One. 2016;11(5):e0156184. doi: 10.1371/journal.pone.0156184
  27. De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, Mumm J.S, Schroeter E.H, Schrijvers V, Wolfe M.S, Ray W.J, Goate A, Kopan R. A presenilin-1-dependent gamma - secretase - like protease mediates release of Notch intracellular domain. Nature. 1999;398(6727):518-22. doi: 10.1038/19083
  28. Wu L, Aster J.C, Blacklow S.C, Lake R, Artavanis-Tsakonas S, Griffin J.D. MAML1, a human homologue of Drosophila mastermind, is a transcriptional coactivator for NOTCH receptors. Nat Genet. 2000;26:484-9. doi: 10.1038/82644
  29. Kurooka H, Honjo T. Functional interaction between the mouse Notch1 intracellular region and histone acetyltransferases PCAF and GCN5. J Biol Chem. 2000;275:17211-20.
  30. Kokubo H, Miyagawa-Tomita S, Johnson R.L. Hesr, a mediator of the notch signaling functions in heart and vessel development. Trends Cardiovasc Med. 2005;15:190-4.
  31. Sanalkumar R, Dhanesh S.B, James J. Non - canonical activation of Notch signaling/target genes in vertebrates. Cell Mol Life Sci. 2010;67(17):2957-68. doi: 10.1007/s00018-010-0391-x
  32. Ladi E, Nichols J.T, Ge W, Miyamoto A, Yao C, Yang L.T, Boulter J, Sun Y.E, Kintner C, Weinmaster G. The divergent DSL ligand Dll3 does not activate Notch signaling but cell autonomously attenuates signaling induced by other DSL ligands. J Cell Biol. 2005;170:983-92. doi: 10.1083/jcb.200503113
  33. Lee S.F, Srinivasan B, Sephton C.F, Dries D.R, Wang B, Yu C, Wang Y, Dewey C.M, Shah S, Jiang J, Yu G. Gamma - secretase - regulated proteolysis of the Notch receptor by mitochondrial intermediate peptidase. J Biol Chem. 2011;286(31):27447-53. doi: 10.1074/jbc.M111.243154
  34. Ayaz F, Osborne B.A. Non-Canonical Notch Signaling in Cancer and Immunity. Front Oncol. 2014;4(4):345. doi: 10.3389/fonc.2014.00345
  35. Forrester J.S, Price M.J, Makkar R.R. Stem cell repair of infarcted myocardium an overview for clinicians. Circulation. 2003;108:1139-45. doi: 10.1161/01.CIR.0000085305.82019.65
  36. Li Y, Hiroi Y, Ngoy S, Okamoto R, Noma K, Wang C.Y, Wang H.W, Zhou Q, Radtke F, Liao R, Liao J.K. Notch1 in bone marrow - derived cells mediates cardiac repair after myocardial infarction. Circulation. 2011;123(8):866-76. doi: 10.1161/CIRCULATIONAHA.110.947531
  37. Swiatek P.J, Lindsell C.E, del Amo F.F, Weinmaster G, Gridley T. Notch1 is essential for postimplantation development in mice. Genes Dev. 1994;8:707-19.
  38. Li Y, Fukuda N, Yokoyama S, Kusumi Y, Hagikura K, Kawano T, Takayama T, Matsumoto T, Satomi A, Honye J, Mugishima H, Mitsumata M, Saito S. Effects of G-CSF on cardiac remodeling and arterial hyperplasia in rats. Eur J Pharmacol. 2006;549:98-106. doi: 10.1016/j.ejphar.2006.08.006
  39. Da Silva Meirelles L, Caplan A.I, Nardi N.B. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008;26:2287-99. doi: 10.1634/stemcells.2007-1122
  40. Nemir M, Metrich M, Plaisance I, Lepore M, Cruchet S, Berthonneche C, Sarre A, Radtke F, Pedrazzini T. The Notch pathway controls fibrotic and regenerative repair in the adult heart. Eur Heart J. 2014;35(32):2174-85. doi: 10.1093/eurheartj/ehs269
  41. Abbott J.D, Huang Y, Liu D, Hickey R, Krause D, Giordano F. Stromal cell - derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 2004;110:3300-5. doi: 10.1161/01.CIR.0000147780.30124.CF
  42. Wang Y.C, Hu X.B, He F, Feng F, Wang L, Li W, Zhang P, Li D, Jia Z.S, Liang Y.M, Han H. Lipopolysaccharide - induced maturation of bone marrow - derived dendritic cells is regulated by notch signaling through the up - regulation of CXCR4. J Biol Chem. 2009;284:15993-6003. doi: 10.1074/jbc.M901144200
  43. Xie J.I, Wang W, Si J.W, Miao X.Y, Li J.C, Wang Y.C, Wang Z.R, Ma J, Zhao X.C, Li Z, Yi H, Han H. Notch signaling regulates CXCR4 expression and the migration of mesenchymal stem cells. Cell Immunol. 2013;281(1):68-75. doi: 10.1016/j.cellimm.2013.02.001
  44. Li Q, Turdi S, Thomas D.P, Zhou T, Ren J. Intra - myocardial delivery of mesenchymal stem cells ameliorates left ventricular and cardiomyocyte contractile dysfunction following myocardial infarction. Toxicol Lett. 2010;195(23):119-26. doi: 10.1016/j.toxlet.2010.03.009
  45. Yu X.Y, Geng Y.J, Li X.H, Lin Q.X, Shan Z.X, Lin S.G, Song Y.H, Li Y. The effects of mesenchymal stem cells on c - kit up - regulation and cell - cycle re - entry of neonatal cardiomyocytes are mediated by activation of insulin - like growth factor 1 receptor. Mol Cell Biochem. 2009;332(1-2):25-32. doi: 10.1007/s11010-009-0170-x
  46. Sassoli C, Pini A, Mazzanti B, Quercioli F, Nistri S, Saccardi R, Zecchi-Orlandini S, Bani D, Formigli L. Mesenchymal stromal cells affect cardiomyocyte growth through juxtacrine Notch-1/Jagged-1 signaling and paracrine mechanisms: clues for cardiac regeneration. J Mol Cell Cardiol. 2011;51(3):399-408. doi: 10.1016/j.yjmcc.2011.06.004
  47. Kassner N, Krueger M, Yagita H, Dzionek A, Hutloff A, Kroczek R, Scheffold A, Rutz S. Plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta - like-4/Notch axis. J Immunol. 2010;184(2):550-4. doi: 10.4049/jimmunol.0903152
  48. Woollard K.J, Geissmann F. Monocytes in atherosclerosis: Subsets and functions. Nat Rev Cardiol. 2010;7(2):77-86. doi: 10.1038/nrcardio.2009.228
  49. Piggott K, Deng J, Warrington K, Younge B, Kubo J.T, Desai M, Goronzy J.J, Weyand C.M. Blocking the NOTCH pathway inhibits vascular inflammation in large - vessel vasculitis. Circulation. 2011;123(3):309-18. doi: 10.1161/CIRCULATIONAHA.110.936203
  50. Chow J.C, Young D.W, Golenbock D.T, Christ W.J, Gusovsky F. Toll - like receptor-4 mediates lipopolysaccharide - induced signal transduction. J Biol Chem. 1999;274:10689-92. doi: 10.1074/jbc.274.16.10689
  51. Mosser D.M, Edwards J.P. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958-69. doi: 10.1038/nri2448
  52. Nathan C.F, Murray H.W, Wiebe M.E, Rubin B.Y. Identification of interferon - gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983;158:670-89. doi: 10.1084/jem.158.3.670
  53. Loke P, Gallagher I, Nair M.G, Zang X, Brombacher F, Mohrs M, Allison J.P, Allen J.E. Alternative activation is an innate response to injury that requires CD4+ T cells to be sustained during chronic infection. J Immunol. 2007;179:3926-36. doi: 10.4049/jimmunol.179.6.3926
  54. Paliard X, de Waal Malefijt R, Yssel H, Blanchard D, Chretien I, Abrams J, de Vries J, Spits H. Simultaneous production of IL-2, IL-4, and IFN-gamma by activated human CD4+ and CD8+ T cell clones. J Immunol. 1988;141:849-55.
  55. Wang Y.C, He F, Feng F, Liu X.W, Dong G.Y, Qin H.Y, Hu X.B, Zheng M.H, Liang L, Feng L, Liang Y.M, Han H. Notch signaling determines the M1 versus M2 polarization of macrophages in antitumor immune responses. Cancer Res. 2010;70:4840-9. doi: 10.1158/0008-5472.CAN-10-0269
  56. Singla R.D, Wang J, Singla D.K. Regulation of Notch 1 signaling in THP-1 cells enhances M2 macrophage differentiation. Am J Physiol Heart Circ Physiol. 2014;307(11):1634-42. doi: 10.1152/ajpheart.00896.2013
  57. Ahmad H.R, Hashmi S. Is biological repair of heart on the horizon? Pak J Med Sci. 2017;33(4):1042-46. doi: 10.12669/pjms.334.12938
  58. Urbanek K, Cabral da Silva M.C, Ide-Iwata N, Maestroni S, Delucchi F, Zheng H, Ferreira-Martins J, Ogorek B, D’Amario D, Bauer M, Zerbini G, Rota M, Hosoda T, Liao R, Anversa P, Kajstura J, Leri A. Inhibition of Notch1-dependent cardiomyogenesis leads to a dilated myopathy in the neonatal heart. Circ Res. 2010;107:429-41. doi: 10.1161/CIRCRESAHA.110.218487
  59. Collesi C, Felician G, Secco I, Gutierrez M.I, Martelletti E, Ali H, Zentilin L, Myers M.P, Giacca M. Reversible Notch1 acetylation tunes proliferative signalling in cardiomyocytes. Cardiovasc Res. 2018;114(1):103-22. doi: 10.1093/cvr/cvx228
  60. Ge W, Ren J. mTOR-STAT3-Notch signalling contributes to ALDH2-induced protection against cardiac contractile dysfunction and autophagy under alcoholism. J Cell Mol Med. 2012;16(3):616-26. doi: 10.1111/j.1582-4934.2011.01347.x
  61. Lee J.H, Suk J, Park J, Kim S.B, Kwak S.S, Kim J.W, Lee C.H, Byun B, Ahn J.K, Joe C.O. Notch signal activates hypoxia pathway through hes1-dependent src/signal transducers and activators of transcription pathway. Mol Cancer Res. 2009;7:1663-71. doi: 10.1158/1541-7786.MCR-09-0191
  62. Gude N.A, Emmanuel G, Wu W, Cottage C.T, Fischer K., Quijada P, Muraski J.A, Alvarez R, Rubio M, Schaefer E, Sussman M.A. Activation of Notch - mediated protective signaling in the myocardium. Circ Res. 2008;102:1025-35. doi: 10.1161/CIRCRESAHA.107.164749
  63. Kratsios P, Catela C, Salimova E, Huth M, Berno V, Rosenthal N, Mourkioti F. Distinct roles for cell - autonomous Notch signaling in cardiomyocytes of the embryonic and adult heart. Circ Res. 2010;106:559-72. doi: 10.1161/CIRCRESAHA.109.203034
  64. Patel N.S, Li J.L, Generali D, Poulsom R, Cranston D.W, Harris A.L. Up - regulation of δ - like 4 ligand in human tumor vasculature and the role of basal expression in endothelial cell function. Cancer Res. 2005;65(19):8690-7. doi: 10.1158/0008-5472.CAN-05-1208
  65. Mailhos C, Modlich U, Lewis J, Harris A, Bicknell R, Ish-Horowicz D. Delta4, an endothelial specific notch ligand expressed at sites of physiological and tumor angiogenesis. Differentiation. 2001;69:135-44. doi: 10.1046/j.1432-0436.2001.690207.x
  66. Gustafsson M.V, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, Ruas J.L, Poellinger L, Lendahl U, Bondesson M. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell. 2005;9:617-28. doi: 10.1016/j.devcel.2005.09.010
  67. Croquelois A, Domenighetti A.A, Nemir M, Lepore M, Rosenblatt-Velin N, Radtke F, Pedrazzini T. Control of the adaptive response of the heart to stress via the Notch1 receptor pathway. J Exp Med. 2008;205:3173-85. doi: 10.1084/jem.20081427
  68. Yu H.C, Qin H.Y, He F, Wang L, Fu W, Liu D, Guo F.C, Liang L, Dou K.F, Han H. Canonical notch pathway protects hepatocytes from ischemia/reperfusion injury in mice by repressing reactive oxygen species production through JAK2/STAT3 signaling. Hepatology. 2011;54:979-88. doi: 10.1002/hep.24469
  69. Pei H, Yu Q, Xue Q, Guo Y, Sun L, Hong Z, Han H, Gao E, Qu Y, Tao L. Notch1 cardioprotection in myocardial ischemia/reperfusion involves reduction of oxidative/nitrative stress. Basic Res Cardiol. 2013;108(5):373. doi: 10.1007/s00395-013-0373-x
  70. Liu Y, Wang T, Yan J, Jiagbogu N, Heideman D.A, Canfield A.E, Alexander M.Y. HGF/c-Met signalling promotes Notch3 activation and human vascular smooth muscle cell osteogenic differentiation in vitro. Atherosclerosis. 2011;219(2):440-7. doi: 10.1016/j.atherosclerosis.2011.08.033
  71. Kochegura T.N, Makarevich P.I, Ovchinnikov A.G, Zhigunova L.V, Lahova E.L, Shestakova M.V, Ageev F.T, Parfenova E.V. Circulating hepatocyte growth factor (hgf) in patients with comorbidity of chronic heart failure, type 2 diabetes mellitus and impaired lipid metabolism. Diabetes Mellitus. 2013;16(2):17-25. doi: 10.14341/2072-0351-3752
  72. Farzaneh M, Rahimi F, Alishahi M, Khoshnam S.E. Paracrine mechanisms involved in mesenchymal stem cell differentiation into cardiomyocytes. Curr Stem Cell Res Ther. 2018;1(2):34-45. doi: 10.2174/1574888X13666180821160421
  73. Campa V.M, Gutierrez-Lanza R, Cerignoli F, Diaz-Trelles R, Nelson B, Tsuji T, Barcova M, Jiang W, Mercola M. Notch activates cell cycle reentry and progression in quiescent cardiomyocytes. J Cell Biol. 2008;183(1):129-41. doi: 10.1083/jcb.200806104
  74. Beltrami A.P, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763-8. doi: 10.1016/S0092-8674(03)00687-1
  75. Dergilev K.V, Tsokolaeva Z.I, Makarevich P.I, Boldyreva M.A, Beloglazova I.B, Zubkova E.S, Rubina K.A, Sysoeva V.Y, Sharonov G.V, Akchurin R.S, Parfenova E.V. Isolation and characterization of cardiac progenitor cells from myocardial right atrial appendage tissue. Cell Tiss Biol. 2016;10(5):349-56.
  76. Дергилев К.В., Рубина К.А., Парфенова Е.В. Резидентные стволовые клетки сердца. Кардиология. 2011;51(4):84-92.
  77. Dergilev K.V, Makarevich P.I, Tsokolaeva Z.I, Boldyreva M.A, Beloglazova I.B, Zubkova E.S, Menshikov M.Y, Parfenova E.V. Comparison of cardiac stem cell sheeys detached from versene solution and from thermoresponsive dishes reveals similar properties of constructs. Tiss Cell. 2017;49(1):64-71. doi: 10.1016/j.tice.2016.12.001
  78. Messina E, Giacomello A. Diabetic cardiomyopathy: a "cardiac stem cell disease" involving p66Shc, an attractive novel molecular target for heart failure therapy. Circ Res. 2006;99(1):1-2. doi: 10.1161/01.RES.0000233141.65522.3e
  79. Urbanek K, Torella D, Sheikh F, De Angelis A, Nurzynska D, Silvestri F, Beltrami CA, Bussani R, Beltrami A.P, Quaini F, Bolli R, Leri A, Kajstura J, Anversa P. Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc Natl Acad Sci USA. 2005;102(24):8692-7. doi: 10.1073/pnas.0500169102
  80. Linke A, Muller P, Nurzynska D, Casarsa C, Torella D, Nascimbene A, Castaldo C, Cascapera S, Böhm M, Quaini F, Urbanek K, Leri A, Hintze T.H, Kajstura J, Anversa P. Stem cells in the dog heart are self - renewing, clonogenic, and multipotent and regenerate infarcted myocardium, improving cardiac function. Proc Natl Acad Sci USA. 2005;102(25):8966-71. doi: 10.1073/pnas.0502678102
  81. Johnston P.V, Sasano T, Mills K, Evers R, Lee S.T, Smith R.R, Lardo A.C, Lai S, Steenbergen C, Gerstenblith G, Lange R, Marbán E. Engraftment, differentiation, and functional benefits of autologous cardiosphere - derived cells in porcine ischemic cardiomyopathy. Circulation. 2009;120(12):1075-83. doi: 10.1161/CIRCULATIONAHA.108.816058
  82. Urbanek K, Quaini F, Tasca G, Torella D, Castaldo C, Nadal-Ginard B, Leri A, Kajstura J, Quaini E, Anversa P. Intense myocyte formation from cardiac stem cells in human cardiac hypertrophy. Proc Natl Acad Sci USA. 2003;100(18):10440-5. doi: 10.1073/pnas.1832855100
  83. Smith R.R, Barile L, Cho H.C, Leppo M.K, Hare J.M, Messina E, Giacomello A, Abraham M.R, Marbán E. Regenerative potential of cardiosphere - derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115(7):896-908. doi: 10.1161/CIRCULATIONAHA.106.655209
  84. Bearzi C, Rota M, Hosoda T, Tillmanns J, Nascimbene A, De Angelis A, Yasuzawa-Amano S, Trofimova I, Siggins R.W, Lecapitaine N, Cascapera S, Beltrami A.P, D'Alessandro D.A, Zias E, Quaini F, Urbanek K, Michler R.E, Bolli R, Kajstura J, Leri A, Anversa P. Human cardiac stem cells. Proc Natl Acad Sci USA. 2007;104(35):14068-73. doi: 10.1073/pnas.0706760104
  85. Wang Y, Haider H.Kh, Ahmad N, Zhang D, Ashraf M. Evidence for ischemia induced host - derived bone marrow cell mobilization into cardiac allografts. J Mol Cell Cardiol. 2006;41(3):478-87. doi: 10.1016/j.yjmcc.2006.06.074
  86. Dergilev K, Tsokolaeva Z, Boldyreva M, Beloglazova I, Zubkova E, Parfyonova Ye. Notch Activation Enchances Vascular Lineage Commitment of Cardiac Stem Cells. Mol Ther. 2016;24(1):177-8.
  87. Boni A, Urbanek K, Nascimbene A, Hosoda T, Zheng H, Delucchi F, Amano K, Gonzalez A, Vitale S, Ojaimi C, Rizzi R, Bolli R, Yutzey K.E, Rota M, Kajstura J, Anversa P, Leri A. Notch1 regulates the fate of cardiac progenitor cells. Proc Natl Acad Sci USA. 2008;105:15529-34. doi: 10.1073/pnas.0808357105
  88. Lawson N.D, Vogel A.M, Weinstein B.M. Sonic hedgehog and vascular endothelial growth factor act upstream of the Notch pathway during arterial endothelial differentiation. Dev Cell. 2002;3(1):127-36. doi: 10.1016/S1534-5807(02)00198-3
  89. Niessen K, Karsan A. Notch signaling in cardiac development. Circ Res. 2008;102(10):1169-81. doi: 10.1161/CIRCRESAHA.108.174318
  90. Espinosa L, Ingles-Esteve J, Aguilera C, Bigas A. Phosphorylation by glycogen synthase kinase-3 beta down - regulates Notch activity, a link for Notch and Wnt pathways. J Biol Chem. 2003;278(34):32227-35. doi: 10.1074/jbc.M304001200
  91. Bridges E, Oon C.E, Harris A. Notch regulation of tumor angiogenesis. Future Oncol. 2011;7(4):569-88. doi: 10.2217/fon.11.20
  92. Del Monte G, Casanova J.C, Guadix J.A, Mac Grogan D, Burch J.B, Perez-Pomares J.M, de la Pompa J.L. Differential Notch signaling in the epicardium is required for cardiac inflow development and coronary vessel morphogenesis. Circ Res. 2011;108:824-36. doi: 10.1161/CIRCRESAHA.110.229062
  93. Yang K, Doughman Y.Q, Karunamuni G, Gu S, Yang Y.C, Bader D.M, Watanabe M. Expression of active Notch1 in avian coronary development. Dev Dyn. 2009;238(1):162-70. doi: 10.1002/dvdy.21811
  94. Kwon S.M, Eguchi M, Wada M, Iwami Y, Hozumi K, Iwaguro H, Masuda H, Kawamoto A, Asahara T. Specific Jagged-1 signal from bone marrow microenvironment is required for endothelial progenitor cell development for neovascularization. Circulation. 2008;118(2):157-65. doi: 10.1161/CIRCULATIONAHA.107.754978
  95. Wang L, Wang Y.C, Hu X.B, Zhang B.F, Dou G.R, He F, Gao F, Feng F, Liang Y.M, Dou K.F, Han H. Notch-RBP-J signaling regulates the mobilization and function of endothelial progenitor cells by dynamic modulation of CXCR4 expression in mice. PLoS One. 2009;4(10):72-8. doi: 10.1371/journal.pone.0007572
  96. Tetzlaff F, Fischer A. Control of Blood Vessel Formation by Notch Signaling. Adv Exp Med Biol. 2018;1066:319-38. doi: 10.1007/978-3-319-89512-3_16
  97. Manderfield L.J, High F.A, Engleka K.A, Liu F, Li L, Rentschler S, Epstein J. Notch activation of Jagged1 contributes to the assembly of the arterial wall. Circulation. 2012;125:314-23. doi: 10.1161/CIRCULATIONAHA.111.047159
  98. High F.A, Zhang M, Proweller A, Tu L, Parmacek M.S, Pear W.S, Epstein J.A. An essential role for Notch in neural crest during cardiovascular development and smooth muscle differentiation. J Clin Invest. 2007;117:353-63. doi: 10.1172/JCI30070
  99. Grieskamp T, Rudat C, Ludtke T.H, Norden J, Kispert A. Notch signaling regulates smooth muscle differentiation of epicardium - derived cells. Circ Res. 2011;108:813-23. doi: 10.1161/CIRCRESAHA.110.228809
  100. Rentschler S, Harris B.S, Kuznekoff L, Jain R, Manderfield L, Lu M.M, Morley G.E, Patel V.V, Epstein J.A. Notch signaling regulates murine atrioventricular conduction and the formation of accessory pathways. J Clin Invest. 2011;121:525-33. doi: 10.1172/JCI44470
  101. Sciacca S, Pilato M, Mazzoccoli G, Pazienza V, Vinciguerra M. Anti - correlation between longevity gene SirT1 and Notch signaling in ascending aorta biopsies from patients with bicuspid aortic valve disease. Heart Vessels. 2013;28:268-75. doi: 10.1007/s00380-012-0238-5
  102. Acharya A, Hans C.P, Koenig S.N, Nichols H.A, Galindo C.L, Garner H.R, Merrill W.H, Hinton R.B, Garg V. Inhibitory role of Notch1 in calcific aortic valve disease. PloS One. 2011;6:277-83. doi: 10.1371/journal.pone.0027743
  103. Bin Hafeez B, Adhami V.M, Asim M, Siddiqui I.A, Bhat K.M, Zhong W, Saleem M, Din M, Setaluri V, Mukhtar H. Targeted knockdown of Notch1 inhibits invasion of human prostate cancer cells concomitant with inhibition of matrix metalloproteinase-9 and urokinase plasminogen activator. Clin Cancer Res. 2009;15(2):452-9. doi: 10.1158/1078-0432.CCR-08-1631
  104. Tkachuk V.A, Plekhanova O.S, Parfyonova Y.V. Regulation of arterial remodeling and angiogenesis by urokinase - type plasminogen activator. Can J Physiol Pharmacol. 2009;87(4):231-51. doi: 10.1139/Y08-113
  105. Loscalzo J. The macrophage and fibrinolysis. Semin Thromb Hemost. 1996;22(6):503-6. doi: 10.1055/s-2007-999051
  106. Binder B.R, Mihaly J, Prager G.W. uPAR-uPA-PAI-1 interactions and signaling: a vascular biologist's view. Thromb Haemost. 2007;97(3):336-42.
  107. Степанова В.В., Ткачук В.А. Урокиназа как мультидоменный белок и полифункциональный регулятор клеток. Биохимия (Москва). 2002;67(1):109-18.
  108. Парфенова Е.В., Плеханова О.С., Стапанова В.В., Меньшиков М.Ю., Цоколаева З.И., Талицкий К.А., Рахмат-Заде Т.М., Трактуев Д.О., Торосян Н.А., Рогунова Н.А., Ратнер Е.И., Ткачук В.А. Активатор плазминогена урокиназного типа: механизмы участия в ремоделировании сосудов и ангиогенезе, подходы генной терапии к ишемии. Российский физиологический журнал им. И.M. Сеченова. 2004;90(5):547-68.
  109. Aoyagi-Ikeda K, Maeno T, Matsui H, Ueno M, Hara K, Aoki Y, Aoki F, Shimizu T, Doi H, Kawai-Kowase K, Iso T, Suga T, Arai M, Kurabayashi M. Notch induces myofibroblast differentiation of alveolar epithelial cells via transforming growth factor-{beta}-Smad3 pathway. Am J Respir Cell Mol Biol. 2011;45(1):136-44. doi: 10.1165/rcmb.2010-0140OC
  110. Dees C, Zerr P, Tomcik M, Beyer C, Horn A, Akhmetshina A, Palumbo K, Reich N, Zwerina J. Inhibition of Notch signaling prevents experimental fibrosis and induces regression of established fibrosis. Arthritis Rheum. 2011;63:1396-404.
  111. Quillard T, Charreau B. Impact of Notch signaling on inflammatory responses in cardiovascular disorders. Int J Mol Sci. 2013;14(4):6863-88. doi: 10.3390/ijms14046863
  112. Felician G, Collesi C, Lusic M, Martinelli V, Ferro M.D, Zentilin L, Zacchigna S, Giacca M. Epigenetic modification at Notch responsive promoters blunts efficacy of inducing notch pathway reactivation after myocardial infarction. Circ Res. 2014;115(7):636-49. doi: 10.1161/CIRCRESAHA.115.304517
  113. Rodriguez P, Sassi Y, Troncone L, Benard L, Ishikawa K, Gordon R.E, Lamas S, Laborda J, Hajjar R.J, Lebeche D. Deletion of delta - like 1 homologue accelerates fibroblast - myofibroblast differentiation and induces myocardial fibrosis. Eur Heart J. 2018;1(2):23-42. doi: 10.1093/eurheartj/ehy188
  114. Boopathy A.V, Martinez M.D, Smith A.W, Brown M.E, García A.J, Davis M.E. Intramyocardial Delivery of Notch Ligand-Containing Hydrogels Improves Cardiac Function and Angiogenesis Following Infarction. Tissue Eng Part A. 2015;21(17-18):2315-22. doi: 10.1089/ten.TEA.2014.0622
  115. Zhou X.L, Zhu R.R, Liu S, Xu H, Xu X, Wu Q.C, Liu J.C. Notch signaling promotes angiogenesis and improves cardiac function after myocardial infarction. J Cell Biochem. 2018;119(8):7105-12. doi: 10.1002/jcb.27032
  116. Gude N, Joyo E, Toko H, Quijada P, Villanueva M, Hariharan N, Sacchi V, Truffa S, Joyo A, Voelkers M, Alvarez R, Sussman M.A. Notch activation enhances lineage commitment and protective signaling in cardiac progenitor cells. Basic Res Cardiol. 2015;110(3):29. doi: 10.1007/s00395-015-0488-3
  117. Li K, Li Y, Wu W, Gordon W.R, Chang D.W, Lu M, Scoggin S, Fu T, Vien L, Histen G, Zheng J, Martin-Hollister R, Duensing T, Singh S, Blacklow S.C, Yao Z, Aster J.C, Zhou B.B. Modulation of Notch signaling by antibodies specific for the extracellular negative regulatory region of NOTCH3. J Biol Chem. 2008;283(12):8046-54. doi: 10.1074/jbc.M800170200
  118. Li W, Lu Y, Han R, Yue Q, Song X, Wang F, Wu R, Hou F, Yang L, Xu L, Zhao R, Hu J. Gremlin2 Regulates the Differentiation and Function of Cardiac Progenitor Cells via the Notch Signaling Pathway. Cell Physiol Biochem. 2018;47(2):579-89. doi: 10.1159/000490012. Epub 2018 May 22.
  119. Mašek J, Andersson E.R. The developmental biology of genetic Notch disorders. Development. 2017;144(10):1743-63. doi: 10.1242/dev.148007

Copyright (c) 2018 Consilium Medicum

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.
 
 


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies