Publikationsserver der Universitätsbibliothek Marburg

Titel:Bone Morphogenetic Protein 9 and Bone Morphogenetic Protein 10 are Vital Factors in Maintaining Adult Vascular Homeostasis and Cardiac Function
Autor:Rice, Megan
Weitere Beteiligte: Braun, Thomas (Prof. Dr.)
Veröffentlicht:2017
URI:https://archiv.ub.uni-marburg.de/diss/z2017/0517
URN: urn:nbn:de:hebis:04-z2017-05176
DOI: https://doi.org/10.17192/z2017.0517
DDC:570 Biowissenschaften, Biologie
Titel (trans.):Knochenmorphogenetisches Protein 9 und Knochenmorphogenetisches Protein 10 sind wichtige Faktoren bei der Erhaltung der erwachsenen vaskulären Homöostase und Herzfunktion
Publikationsdatum:2019-08-05
Lizenz:https://creativecommons.org/licenses/by-nc-sa/4.0

Dokument

Schlagwörter:

Summary:
Bone Morphogenetic Proteins (BMPs) are members of the well known Transforming Growth Factor (TGF) superfamily, consisting of TGF β proteins, Activins and Growth Differentiation Factors (GDFs). These factors play an essential role in numerous different aspects of embryonic development and physiological organ function. BMP9 and BMP10 are members of this superfamily; however their role, especially in the cardiovascular system, is still poorly characterised. BMP9 is produced by and is secreted from the liver and is proposed to act as a major circulating vascular quiescence factor. BMP10, however, has a more limited expression where it is expressed during embryonic development in the ventricles from embryonic day (E) 9.0 to 13.5. After E 14.5, expression of BMP10 declines in the ventricles, but is maintained in the right atria of postnatal and adult hearts. BMP9 and BMP10 bind with high affinity to ALK1, an endothelial specific receptor as well as to endoglin (ENG), a TGF β co-receptor. Since both of these ligands bind with very high affinity to the ALK1 receptor and soluble ENG receptor, it was suggested that BMP9 and BMP10 are important mediators of cardiovascular development and homeostasis. So far it has been shown that mutations in ALK1, ENG and SMAD4 genes can result in hereditary haemorrhagic telangiectasias (HHT), which is a disease that results in vascular abnormalities. BMP10 along with ENG has been shown to be involved in pre-eclampsia, a disease resulting in high blood pressure (BP), thus strengthening the argument that these BMP family members are involved in cardio vascular homeostasis. The aim of this project was to elucidate the role of BMP9 and BMP10 in the adult heart and the ir role in cardiovascular homeostasis. Constitutive BMP9 knockout mice were generated, with a conditional heart-specific BMP10 deletion under the control of ANF Cre. Results demonstrated that the loss of BMP9 and BMP10 leads to vessel defects which caused disruption in vascular tone and altered cardiac function. In summary, these results have identified an essential role for BMP9 and BMP10 in postnatal cardiac remodelling and vascular homeostasis.

Bibliographie / References

  1. 166. Van de Peer, Y. Calculate and draw custom Venn diagrams. at <http://bioinformatics.psb.ugent.be/webtools/Venn/>
  2. 209. College, M. T. The Cardiovascular System: Blood Vessels. (2012). at <http://classes.midlandstech.edu/carterp/Courses/bio211/chap19/chap19.html>
  3. 177. Ruiz, S. et al. A mouse model of hereditary hemorrhagic telangiectasia generated by transmammary-delivered immunoblocking of BMP9 and BMP10. Sci. Rep. (2016). doi:10.1038/srep37366
  4. 258. Montecucco, F., Carbone, F. & Schindler, T. H. Pathophysiology of ST-segment elevation myocardial infarction: novel mechanisms and treatments. Eur. Heart J. ehv592 (2015). doi:10.1093/eurheartj/ehv592
  5. 271. Kolk, M. V. V et al. LAD-ligation: a murine model of myocardial infarction. J. Vis. Exp. e1438 (2009). doi:10.3791/1438
  6. 275. Hu, N. et al. BMP9-regulated angiogenic signaling plays an important role in the osteogenic differentiation of mesenchymal progenitor cells. J Cell Sci (2012). doi:jcs.114231 [pii]\r10.1242/jcs.114231
  7. 259. Mendis, S., Puska, P. & Norrving, B. Global atlas on cardiovascular disease prevention and control. World Heal. Organ. 2-14 (2011). doi:NLM classification: WG 120
  8. 196. Soriano, P. Abnormal kidney development and hematological disorders in PDGF beta-receptor mutant mice. Genes Dev. 8, 1888-1896 (1994).
  9. 147. Gallione, C. J. et al. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet 363, 852-859 (2004).
  10. 194. Boettger, T. et al. Acquistion of the contractile phenotype by murine arterial smooth muscle cells depends on the miR-143/145 gene cluster. J. Clin. Invest. 119, 2634-2647 (2009).
  11. 135. Oh, S. P. et al. Activin receptor-like kinase 1 modulates transforming growth factor- beta 1 signaling in the regulation of angiogenesis. Proc Natl Acad Sci U S A 97, 2626-2631 (2000).
  12. 265. Goumans, M. J. et al. Activin receptor-like kinase (ALK)1 is an antagonistic mediator of lateral TGFβ/ALK5 signaling. Mol. Cell 12, 817-828 (2003).
  13. 153. Sharff, K. a et al. Hey1 basic helix-loop-helix protein plays an important role in mediating BMP9-induced osteogenic differentiation of mesenchymal progenitor cells. J. Biol. Chem. 284, 649-59 (2009).
  14. 208. Cunha, S. I. & Pietras, K. ALK1 as an emerging target for antiangiogenic therapy of cancer. Blood 117, 6999-7006 (2011).
  15. 140. Park, S. O. et al. ALK5- and TGFBR2-independent role of ALK1 in the pathogenesis of hereditary hemorrhagic telangiectasia type 2. Blood 111, 633-642 (2008).
  16. 101. Tzima, E. et al. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 437, 426-431 (2005).
  17. 144. Bourdeau, A., Dumont, D. J. & Letarte, M. A murine model of hereditary hemorrhagic telangiectasia. J. Clin. Invest. 104, 1343-51 (1999).
  18. Porrello, E. R. & Olson, E. N. A neonatal blueprint for cardiac regeneration. Stem Cell Res. 13, 556-70 (2014).
  19. 244. Iivanainen, E. et al. Angiopoietin-regulated recruitment of vascular smooth muscle cells by endothelial-derived heparin binding EGF-like growth factor. FASEB J. 17, 1609-21 (2003).
  20. 245. Andersen, C. U., Hilberg, O., Mellemkjaer, S., Nielsen-Kudsk, J. E. & Simonsen, U. Apelin and pulmonary hypertension. Pulm. Circ. 1, 334-46 (2013).
  21. 198. Wang, C. et al. Apelin induces vascular smooth muscle cells migration via a PI3K/Akt/FoxO3a/MMP-2 pathway. Int. J. Biochem. Cell Biol. 69, 173-182 (2015).
  22. 197. Zhang, H. et al. Apelin inhibits the proliferation and migration of rat PASMCs via the activation of PI3K/Akt/mTOR signal and the inhibition of autophagy under hypoxia. J. Cell. Mol. Med. 18, 542-553 (2014).
  23. 281. Greenberg, J. I. et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature 456, 809-13 (2008).
  24. 86. Paszkowiak, J. J. & Dardik, A. Arterial wall shear stress: observations from the bench to the bedside. Vasc. Endovascular Surg. 37, 47-57 (2003).
  25. 132. Peacock, H., Caolo, V. & Jones, E. Arteriovenous malformations in Hereditary Haemorrhagic Telangiectasia: looking beyond ALK1-NOTCH interactions. Cardiovasc. Res. (2015).
  26. 134. Urness, L. D., Sorensen, L. K. & Li, D. Y. Arteriovenous malformations in mice lacking activin receptor-like kinase-1. Nat. Genet. 26, 328-31 (2000).
  27. 220. Cox, E. J. & Marsh, S. A. A systematic review of fetal genes as biomarkers of cardiac hypertrophy in rodent models of diabetes. PLoS ONE 9, (2014).
  28. 168. De Lange, F. J., Moorman, A. F. M. & Christoffels, V. M. Atrial cardiomyocyte-specific expression of Cre recombinase driven by an Nppa gene fragment. Genesis 37, 1-4 (2003).
  29. 222. Franco, V. et al. Atrial natriuretic peptide dose-dependently inhibits pressure overload-induced cardiac remodeling. Hypertension 44, 746-750 (2004).
  30. Potente, M., Gerhardt, H. & Carmeliet, P. Basic and therapeutic aspects of angiogenesis. Cell 146, 873-887 (2011).
  31. 118. Chen, H. et al. BMP10 is essential for maintaining cardiac growth during murine cardiogenesis. Development 131, 2219-2231 (2004).
  32. 120. Ricard, N. et al. BMP9 and BMP10 are critical for postnatal retinal vascular remodeling. Blood 119, 6162-6171 (2012).
  33. 239. Kim, J. H., Peacock, M. R., George, S. C. & Hughes, C. C. W. BMP9 induces EphrinB2 expression in endothelial cells through an Alk1-BMPRII/ActRII-ID1/ID3-dependent pathway: Implications for hereditary hemorrhagic telangiectasia type II. Angiogenesis 15, 497-509 (2012).
  34. 112. Bidart, M. et al. BMP9 is produced by hepatocytes and circulates mainly in an active mature form complexed to its prodomain. Cell. Mol. life Sci. 69, 313-24 (2012).
  35. 130. Wooderchak-Donahue, W. L. et al. BMP9 Mutations Cause a Vascular-Anomaly Syndrome with Phenotypic Overlap with Hereditary Hemorrhagic Telangiectasia. Am. J. Hum. Genet. 93, 530-537 (2013).
  36. 121. Scharpfenecker, M. et al. BMP-9 signals via ALK1 and inhibits bFGF-induced endothelial cell proliferation and VEGF-stimulated angiogenesis. J. Cell Sci. 120, 964-972 (2007).
  37. 131. Rigelsky, C. M. et al. BMPR2 mutation in a patient with pulmonary arterial hypertension and suspected hereditary hemorrhagic telangiectasia. Am. J. Med. Genet. A 146A, 2551-6 (2008).
  38. 254. Beppu, H. et al. BMPR-II heterozygous mice have mild pulmonary hypertension and an impaired pulmonary vascular remodeling response to prolonged hypoxia. Am. J. Physiol. Lung Cell. Mol. Physiol. 287, L1241-L1247 (2004).
  39. 148. Beppu, H. et al. BMP type II receptor is required for gastrulation and early development of mouse embryos. Dev. Biol. 221, 249-258 (2000).
  40. 167. Bücker, S. BMP und SMAD signale im kardiovaskulären system. (JUSTUS-LIEBIGUNIVERSITÄT, GIESSEN, 2011).
  41. 116. Miller, A. F., Harvey, S. A., Thies, R. S. & Olson, M. S. Bone morphogenetic protein-9. An autocrine/paracrine cytokine in the liver. J. Biol. Chem. 275, 17937-45 (2000).
  42. 119. David, L. et al. Bone morphogenetic protein-9 is a circulating vascular quiescence factor. Circ. Res. 102, 914-922 (2008).
  43. 126. Rider, C. C. & Mulloy, B. Bone morphogenetic protein and growth differentiation factor cytokine families and their protein antagonists. Biochem. J. 429, 1-12 (2010).
  44. 155. Upton, P. D., Davies, R. J., Trembath, R. C. & Morrell, N. W. Bone morphogenetic protein (BMP) and activin type II receptors balance BMP9 signals mediated by activin receptor-like kinase-1 in human pulmonary artery endothelial cells. J. Biol. Chem. 284, 15794-804 (2009).
  45. 205. Wang, R. N. et al. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis. 1, 87-105 (2014).
  46. 221. Rosenkranz, A. C., Hood, S. G., Woods, R. L., Dusting, G. J. & Ritchie, R. H. B-type natriuretic peptide prevents acute hypertrophic responses in the diabetic rat heart: Importance of cyclic GMP. Diabetes 52, 2389-2395 (2003).
  47. Caprioli, a et al. Hemangioblast commitment in the avian allantois: cellular and molecular aspects. Dev. Biol. 238, 64-78 (2001).
  48. 157. Shou, W. et al. Cardiac defects and altered ryanodine receptor function in mice lacking FKBP12. Nature 391, 489-492 (1998).
  49. 217. Senyo, S. E., Lee, R. T. & Kühn, B. Cardiac regeneration based on mechanisms of cardiomyocyte proliferation and differentiation. Stem Cell Research 13, 532-541 (2014).
  50. Kennedy-Lydon, T. & Rosenthal, N. Cardiac regeneration: epicardial mediated repair. Proc Biol Sci. 282, (2015).
  51. 216. Mihl, C., Dassen, W. R. . & Kuipers, H. Cardiac remodelling: concentric versus eccentric hypertrophy in strength and endurance athletes. Neth Hear. J 16, 129-133 (2008).
  52. 238. Park, S. et al. Cardiac, skeletal, and smooth muscle mitochondrial respiration: are all mitochondria created equal? Am J Physiol Hear. Circ Physiol 307, H346-H352 (2014).
  53. Townsend, N., Nichols, M., Scarborough, P. & Rayner, M. Cardiovascular disease in Europe 2014: Epidemiological update. Eur. Heart J. 35, 2950-2959 (2014).
  54. Townsend, N., Nichols, M., Scarborough, P. & Rayner, M. Cardiovascular disease in Europe: Epidemiological update. Eur. Heart J. 34, 3028-3034 (2013).
  55. Townsend, N., Nichols, M., Scarborough, P. & Rayner, M. Cardiovascular disease in Europe - epidemiological update 2015. Eur. Heart J. 36, 2696-2705 (2015).
  56. 247. Tirziu, D., Giordano, F. & Simons, M. Cell Communications in the Heart. Circulation 122, 928- 937 (2010).
  57. 129. Storkebaum, E., Quaegebeur, A., Vikkula, M. & Carmeliet, P. Cerebrovascular disorders: molecular insights and therapeutic opportunities. Nat. Neurosci. 14, 1390-1397 (2011).
  58. 262. Ren, G., Michael, L. H., Entman, M. L. & Frangogiannis, N. G. Morphological Characteristics of the Microvasculature in Healing Myocardial Infarcts. J. Histochem. Cytochem. 50, 71-79 (2002).
  59. 156. Laux, D. W. et al. Circulating Bmp10 acts through endothelial Alk1 to mediate flow-dependent arterial quiescence. Development 140, 3403-12 (2013).
  60. 280. Cao, R. et al. Comparative Evaluation of FGF-2-, VEGF-A-, and VEGF-C-Induced Angiogenesis Lymphangiogenesis, Vascular Fenestrations, and Permeability. Circ. Res. 94, 664-670 (2004).
  61. 180. Sidney, L. E., Branch, M. J., Dunphy, S. E., Dua, H. S. & Hopkinson, A. Concise review: Evidence for CD34 as a common marker for diverse progenitors. Stem Cells 32, 1380-1389 (2014).
  62. 114. Chen, H. et al. Context-dependent signaling defines roles of BMP9 and BMP10 in embryonic and postnatal development. Proc. Natl. Acad. Sci. U. S. A. 110, 11887-92 (2013).
  63. 242. Geudens, I. & Gerhardt, H. Coordinating cell behaviour during blood vessel formation. Development 138, 4569-4583 (2011).
  64. 143. Li, D. Y. et al. Defective angiogenesis in mice lacking endoglin. Science 284, 1534-1537 (1999).
  65. 170. Hershberger, R. E., Hedges, D. J. & Morales, A. Dilated cardiomyopathy: the complexity of a diverse genetic architecture. Nat. Rev. Cardiol. 10, 531-47 (2013).
  66. Bai, H. & Wang, Z. Z. Directing human embryonic stem cells to generate vascular progenitor cells. Gene Ther. 15, 89-95 (2008).
  67. 225. Roman, B. L. et al. Disruption of acvrl1 increases endothelial cell number in zebrafish cranial vessels. Development 129, 3009-3019 (2002).
  68. 149. Liu, D. et al. Dosage-dependent requirement of BMP type II receptor for maintenance of vascular integrity. Blood 110, 1502-10 (2007).
  69. 276. Liehn, E. A. et al. Double-edged role of the CXCL12/CXCR4 axis in experimental myocardial infarction. J. Am. Coll. Cardiol. 58, 2415-2423 (2011).
  70. 85. Franco, C. A. et al. Dynamic endothelial cell rearrangements drive developmental vessel regression. PLoS Biol. 13, e1002125 (2015).
  71. 278. Lee, S. H. et al. Early expression of angiogenesis factors in acute myocardial ischemia and infarction. N. Engl. J. Med. 342, 626-33 (2000).
  72. 152. Jane, J. a et al. Ectopic osteogenesis using adenoviral bone morphogenetic protein (BMP)-4 and BMP-6 gene transfer. Mol. Ther. 6, 464-470 (2002).
  73. 223. Wang, D. et al. Effects of pressure overload on extracellular matrix expression in the heart of the atrial natriuretic peptide-null mouse. Hypertension 42, 88-95 (2003).
  74. 227. Siddiqui, A. Effects of Vasodilation and Arterial Resistance on Cardiac Output. J. Clin. Exp. Cardiolog. 2, 170 (2011).
  75. 191. Chng, S., Ho, L., Tian, J. & Reversade, B. ELABELA: A hormone essential for heart development signals via the apelin receptor. Dev. Cell 27, 672-680 (2013).
  76. 190. Xie, F., Lv, D. & Chen, L. ELABELA: a novel hormone in cardiac development acting as a new endogenous ligand for the APJ receptor. Acta Biochim Biophys Sin 46, 620-622 (2014).
  77. Drake, C. J. Embryonic and adult vasculogenesis. Birth Defects Res. Part C - Embryo Today Rev. 69, 73-82 (2003).
  78. Pardanaud, L. & Dieterlen-Lièvre, F. Emergence of endothelial and hemopoietic cells in the avian embryo. Anat. Embryol. (Berl). 187, 107-14 (1993).
  79. 110. Tillet, E. & Bailly, S. Emerging roles of BMP9 and BMP10 in hereditary hemorrhagic telangiectasia. Front. Genet. 5, 456 (2014).
  80. 142. McAllister, K. et al. Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat. Genet. 8, 345-351 (1994).
  81. 264. Morris, E. et al. Endoglin promotes TGF-??/Smad1 signaling in scleroderma fibroblasts. J. Cell. Physiol. 226, 3340-3348 (2011).
  82. 270. Docherty, N. G. et al. Endoglin regulates renal ischaemia-reperfusion injury. Nephrol. Dial. Transplant. 21, 2106-2119 (2006).
  83. 83. Dimmeler, S. & Zeiher, a M. Endothelial cell apoptosis in angiogenesis and vessel regression. Circ. Res. 87, 434-439 (2000).
  84. 89. Korn, C. et al. Endothelial cell-derived non-canonical Wnt ligands control vascular pruning in angiogenesis. Development 141, 1757-1766 (2014).
  85. 106. Wang, C., Baker, B. M., Chen, C. S. & Schwartz, M. A. Endothelial cell sensing of flow direction. Arterioscler. Thromb. Vasc. Biol. 33, 2130-2136 (2013).
  86. 105. Davies, P. & Barbee, K. Endothelial cell surface imaging: insights into hemodynamic force transduction. Physiology (Bethesda). 153-157 (1994).
  87. 248. Budhiraja, R., Tuder, R. M. & Hassoun, P. M. Endothelial dysfunction in pulmonary hypertension. Am. Hear. Assoc. 109, 159-65 (2004).
  88. 87. Dekker, R. J. et al. Endothelial KLF2 Links Local Arterial Shear Stress Levels to the Expression of Vascular Tone-Regulating Genes. Am. J. Pathol. 167, 609-618 (2005).
  89. 92. Gaengel, K., Genové, G., Armulik, A. & Betsholtz, C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 29, 630-638 (2009).
  90. Eguchi, M., Masuda, H. & Asahara, T. Endothelial progenitor cells for postnatal vasculogenesis. Clin. Exp. Nephrol. 11, 18-25 (2007).
  91. 187. Stow, L. R., Jacobs, M. E., Wingo, C. S. & Cain, B. D. Endothelin-1 gene regulation. FASEB J. 25, 16-28 (2011).
  92. 94. Pitulescu, M. E. & Adams, R. H. Eph/ephrin molecules - A hub for signaling and endocytosis. Genes Dev. 24, 2480-2492 (2010).
  93. 90. Salvucci, O. et al. EphrinB2 controls vessel pruning through STAT1-JNK3 signalling. Nat. Commun. 6, 6576 (2015).
  94. 202. Sciences, M. et al. Epiregulin is a potent vascular smooth muscle cell-derived mitogen induced by angiotensin II, endothelin-1, and thrombin. Proc. Natl. Acad. Sci. U. S. A. 96, 1633-1638 (1999).
  95. 96. Arroyo, A. G. & Iruela-Arispe, M. L. Extracellular matrix, inflammation, and the angiogenic response. Cardiovasc. Res. 86, 226-235 (2010).
  96. 188. Kardami, E. et al. Fibroblast growth factor 2 isoforms and cardiac hypertrophy. Cardiovasc. Res. 63, 458-466 (2004).
  97. 103. Conway, D. E. & Schwartz, M. a. Flow-dependent cellular mechanotransduction in atherosclerosis. J. Cell Sci. 126, 5101-9 (2013).
  98. 98. Davies, P. F. Flow-mediated endothelial mechanotransduction. Physiol. Rev. 75, 519-60 (1995).
  99. 104. Nigro, P., Abe, J.-I. & Berk, B. C. Flow shear stress and atherosclerosis: a matter of site specificity. Antioxid. Redox Signal. 15, 1405-14 (2011).
  100. 233. Wilhelm, K. et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature 529, 1-18 (2016).
  101. 111. Susan-Resiga, D. et al. Furin is the major processing enzyme of the cardiac-specific growth factor bone morphogenetic protein 10. J. Biol. Chem. 286, 22785-94 (2011).
  102. 165. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U. S. A. 102, 15545-50 (2005).
  103. 253. Soubrier, F. et al. Genetics and genomics of pulmonary arterial hypertension. in Journal of the American College of Cardiology 62, (2013).
  104. 163. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).
  105. 249. Kreymborg, K. grosse et al. Identification of right heart-enriched genes in a murine model of chronic outflow tract obstruction. J. Mol. Cell. Cardiol. 49, 598-605 (2010).
  106. 235. Ruhrberg, C. Growing and shaping the vascular tree: Multiple roles for VEGF. BioEssays 25, 1052-1060 (2003).
  107. 117. Neuhaus, H., Rosen, V. & Thies, R. S. Heart specific expression of mouse BMP-10 a novel member of the TGF-beta superfamily. Mech. Dev. 80, 181-4 (1999).
  108. 199. England, J. & Loughna, S. Heavy and light roles: Myosin in the morphogenesis of the heart. Cellular and Molecular Life Sciences 70, 1221-1239 (2013).
  109. 173. Frey, N., Katus, H. A., Olson, E. N. & Hill, J. A. Hypertrophy of the Heart: A New Therapeutic Target? Circulation 109, 1580-1589 (2004).
  110. 274. Kido, M. et al. Hypoxia-inducible factor 1-alpha reduces infarction and attenuates progression of cardiac dysfunction after myocardial infarction in the mouse. J. Am. Coll. Cardiol. 46, 2116- 2124 (2005).
  111. 232. Del Toro, R. et al. Identification and functional analysis of endothelial tip cell-enriched genes. Blood 116, 4025-4033 (2010).
  112. 113. David, L., Mallet, C., Mazerbourg, S., Feige, J. J. & Bailly, S. Identification of BMP9 and BMP10 as functional activators of the orphan activin receptor-like kinase 1 (ALK1) in endothelial cells. Blood 109, 1953-1961 (2007).
  113. 172. Muñoz-Félix JM et al. Identification of bone morphogenetic protein 9 (BMP9) as a novel profibrotic factor in vitro. Cell Signal 28, 1252-61 (2016).
  114. Hazebroek, M., Dennert, R. & Heymans, S. Idiopathic dilated cardiomyopathy: possible triggers and treatment strategies. Netherlands Hear. J. 20, 332-335 (2012).
  115. 169. Kuba, K. et al. Impaired heart contractility in Apelin gene-deficient mice associated with aging and pressure overload. Circ. Res. 101, (2007).
  116. 108. Laughlin, M. H., Newcomer, S. C. & Bender, S. B. Importance of hemodynamic forces as signals for exercise-induced changes in endothelial cell phenotype. J. Appl. Physiol. 104, 588- 600 (2008).
  117. 158. Schierling, W. et al. Increased intravascular flow rate triggers cerebral arteriogenesis. J. Cereb. Blood Flow Metab. 29, 726-737 (2009).
  118. 154. López-Coviella, I., Berse, B., Krauss, R., Thies, R. S. & Blusztajn, J. K. Induction and maintenance of the neuronal cholinergic phenotype in the central nervous system by BMP-9. Science 289, 313-316 (2000).
  119. 181. Kasai, A. et al. Inhibition of apelin expression switches endothelial cells from proliferative to mature state in pathological retinal angiogenesis. Angiogenesis 16, 723-734 (2013).
  120. 224. Corti, P. et al. Interaction between alk1 and blood flow in the development of arteriovenous malformations. Development 138, 1573-1582 (2011).
  121. 138. Bacharach, E., Itin, A. & Keshet, E. In vivo patterns of expression of urokinase and its inhibitor PAI-1 suggest a concerted role in regulating physiological angiogenesis. Proc Natl Acad Sci U S A 89, 10686-90 (1992).
  122. 228. Jackson, W. Ion Channels and Vascular Tone. Hypertension 35, 173-178 (2000).
  123. 161. O'Connell, T. D., Rodrigo, M. C. & Simpson, P. C. Isolation and culture of adult mouse cardiac myocytes. Methods Mol. Biol. 357, 271-296 (2007).
  124. 252. Gore, B. et al. Key role of the endothelial TGF-β/ALK1/endoglin signaling pathway in humans and rodents pulmonary hypertension. PLoS One 9, (2014).
  125. 263. Velasco, S. et al. L- and S-endoglin differentially modulate TGFbeta1 signaling mediated by ALK1 and ALK5 in L6E9 myoblasts. J. Cell Sci. 121, 913-9 (2008).
  126. 229. Schreier, B. et al. Loss of epidermal growth factor receptor in vascular smooth muscle cells and cardiomyocytes causes arterial hypotension and cardiac hypertrophy. Hypertension 61, 333-340 (2013).
  127. Rhee, J. M. & Iannaccone, P. M. Mapping mouse hemangioblast maturation from headfold stages. Dev. Biol. 365, 1-13 (2012).
  128. 193. Halayko, A. J., Salari, H., Ma, X. & Stephens, N. L. Markers of airway smooth muscle cell phenotype. Am.J.Physiol 270, L1040--L1051 (1996).
  129. 219. Dietz, J. Mechanisms of atrial natriuretic peptide secretion from the atrium. Cardiovasc Res 68, 8-17 (2005).
  130. 122. Shi, Y. & Massagué, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113, 685-700 (2003).
  131. 185. Gerthoffer, W. T. Mechanisms of vascular smooth muscle cell migration. Circulation Research 100, 607-621 (2007).
  132. 84. Wietecha, M. S., Cerny, W. L. & DiPietro, L. A. Mechanisms of vessel regression: toward an understanding of the resolution of angiogenesis. Curr Top Microbiol Immunol 367, 3-32 (2013).
  133. 99. Hahn, C. & Schwartz, M. A. Mechanotransduction in vascular physiology and atherogenesis. Nat. Rev. Mol. Cell Biol. 10, 53-62 (2009).
  134. 174. Razeghi, P. et al. Metabolic gene expression in fetal and failing human heart. Circulation 104, 2923-2931 (2001).
  135. 159. Schürmann, C., Gremse, F., Jo, H., Kiessling, F. & Brandes, R. Micro-CT Technique Is Well Suited for Documentation of Remodeling Processes in Murine Carotid Arteries. PLoS One 10, (2015).
  136. 218. Cameron, V. & Ellmers, L. Mini review: natriuretic peptides during development of the fetal heart and circulation. Endocrinology 144, 2191-2194 (2003).
  137. 107. Haga, J. H., Li, Y.-S. J. & Chien, S. Molecular basis of the effects of mechanical stretch on vascular smooth muscle cells. J. Biomech. 40, 947-960 (2007).
  138. 231. Herbert, S. P. & Stainier, D. Y. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat Rev Mol Cell Biol 12, 551-564 (2011).
  139. 214. Creemers, E. E. & Pinto, Y. M. Molecular mechanisms that control interstitial fibrosis in the pressure-overloaded heart. Cardiovasc. Res. 89, 265-272 (2011).
  140. 268. Fernandez, A. et al. Mutation study of Spanish patients with hereditary hemorrhagic telangiectasia and expression analysis of Endoglin and ALK1. Hum.Mutat. 27, 295 (2006).
  141. 171. Conrad, C. H. et al. Myocardial fibrosis and stiffness with hypertrophy and heart failure in the spontaneously hypertensive rat. Circulation 91, 161-170 (1995).
  142. 215. Ho, C. Y. et al. Myocardial Fibrosis as an Early Manifestation of Hypertrophic Cardiomyopathy. N. Engl. J. Med. 363, 552-563 (2010).
  143. 251. Lörchner, H. et al. Myocardial healing requires Reg3β-dependent accumulation of macrophages in the ischemic heart. Nat. Med. 21, 1-21 (2015).
  144. 125. Itoh, S. & ten Dijke, P. Negative regulation of TGF-beta receptor/Smad signal transduction. Curr. Opin. Cell Biol. 19, 176-84 (2007).
  145. 241. Seki, T., Hong, K.-H. & Oh, S. P. Nonoverlapping expression patterns of ALK1 and ALK5 reveal distinct roles of each receptor in vascular development. Lab. Invest. 86, 116-29 (2006).
  146. 95. Gridley, T. Notch signaling in the vasculature. Curr. Top. Dev. Biol. 92, 277-309 (2010).
  147. 250. Kubin, T. et al. Oncostatin M is a major mediator of cardiomyocyte dedifferentiation and remodeling. Cell Stem Cell 9, 420-432 (2011).
  148. 128. Garg, N., Khunger, M., Gupta, A. & Kumar, N. Optimal management of hereditary hemorrhagic telangiectasia. J. Blood Med. 5, 191-206 (2014).
  149. 189. Chen, H. et al. Overexpression of bone morphogenetic protein 10 in myocardium disrupts cardiac postnatal hypertrophic growth. J. Biol. Chem. 281, 27481-27491 (2006).
  150. 151. Gallione, C. et al. Overlapping spectra of SMAD4 mutations in juvenile polyposis (JP) and JPHHT syndrome. Am. J. Med. Genet. A 152A, 333-9 (2010).
  151. 236. Claxton, S. & Fruttiger, M. Oxygen modifies artery differentiation and network morphogenesis in the retinal vasculature. Dev. Dyn. 233, 822-828 (2005).
  152. 145. Mahmoud, M. et al. Pathogenesis of arteriovenous malformations in the absence of endoglin. Circ. Res. 106, 1425-1433 (2010).
  153. 195. Lindahl, P. Pericyte Loss and Microaneurysm Formation in PDGF-B-Deficient Mice. Science 277, 242-245 (1997).
  154. 273. Jürgensen, J. S. et al. Persistent induction of HIF-1alpha and -2alpha in cardiomyocytes and stromal cells of ischemic myocardium. FASEB J. 18, 1415-7 (2004).
  155. 164. Mootha, V. K. et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267-273 (2003).
  156. 176. Ola, R. et al. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun. 7, (2016).
  157. 284. Iwasaki, H. et al. PLGF repairs myocardial ischemia through mechanisms of angiogenesis, cardioprotection and recruitment of myo-angiogenic competent marrow progenitors. PLoS One 6, (2011).
  158. 201. Kim, Y. M. et al. Proteomic identification of adam12 as a regulator for tgf-β1-induced differentiation of human mesenchymal stem cells to smooth muscle cells. PLoS One 7, (2012).
  159. 255. Jerkic, M. et al. Pulmonary hypertension in adult Alk1 heterozygous mice due to oxidative stress. Cardiovasc. Res. 92, 375-384 (2011).
  160. 277. Hashimoto, E. et al. Rapid induction of vascular endothelial growth factor expression by transient ischemia in rat heart. Am J Physiol 267, H1948-54 (1994).
  161. 141. Park, S. O. et al. Real-time imaging of de novo arteriovenous malformation in a mouse model of hereditary hemorrhagic telangiectasia. J. Clin. Invest. 119, 3487-96 (2009).
  162. 267. Kapur, N. et al. Reduced endoglin activity limits cardiac fibrosis and improves survival in heart failure. Circulation 125, 2728-2738 (2012).
  163. 82. Mitchell, C. A., Risau, W. & Drexler, H. C. Regression of vessels in the tunica vasculosa lentis is initiated by coordinated endothelial apoptosis: a role for vascular endothelial growth factor as a survival factor for endothelium. Dev. Dyn. 213, 322-33 (1998).
  164. 136. Tkachuk, V., Stepanova, V., Little, P. J. & Bobik, A. Regulation and role of urokinase plasminogen activator in vascular remodelling. Clin Exp Pharmacol Physiol 23, 759-765 (1996).
  165. 272. Pugh, C. W. & Ratcliffe, P. J. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat. Med. 9, 677-684 (2003).
  166. 260. Frangogiannis, N. G. Regulation of the inflammatory response in cardiac repair. Circulation Research 110, 159-173 (2012).
  167. Pardanaud, L., Yassine, F. & Dieterlen-Lievre, F. Relationship between vasculogenesis, angiogenesis and haemopoiesis during avian ontogeny. Development 105, 473-485 (1989). dependent ocular tissue remodelling. Development 120, 3395-403 (1994).
  168. 213. Cioffi, G., de Simone, G., Mureddu, G., Tarantini, L. & Stefenelli, C. Right atrial size and function in patients with pulmonary hypertension associated with disorders of respiratory system or hypoxemia. Eur J Echocardiogr. 8, 322-331 (2007).
  169. 175. Sato, Y. Role of ETS family transcription factors in vascular development and angiogenesis. Cell Struct. Funct. 26, 19-24 (2001).
  170. 243. Hellström, M., Kalén, M., Lindahl, P., Abramsson, A. & Betsholtz, C. Role of PDGF-B and PDGFR-β in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 126, 3047-3055 (1999).
  171. 283. Autiero, M. et al. Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1. Nat. Med. 9, 936-943 (2003).
  172. 203. Gordon, K. J. & Blobe, G. C. Role of transforming growth factor-β superfamily signaling pathways in human disease. Biochim. Biophys. Acta - Mol. Basis Dis. 1782, 197-228 (2008).
  173. 257. Long, L. et al. Selective enhancement of endothelial BMPR-II with BMP9 reverses pulmonary arterial hypertension. Nat. Med. 21, 777-85 (2015).
  174. 230. Wöltje, K., Jabs, M. & Fischer, A. Serum Induces transcription of Hey1 and Hey2 genes by Alk1 but not notch signaling in endothelial cells. PLoS One 10, (2015).
  175. 91. Pardali, E., Goumans, M. J. & ten Dijke, P. Signaling by members of the TGF-β family in vascular morphogenesis and disease. Trends in Cell Biology 20, 556-567 (2010).
  176. 206. Guo, X. & Wang, X.-F. Signaling cross-talk between TGF-beta/BMP and other pathways. Cell Res. 19, 71-88 (2009).
  177. Patel-Hett, S. & D'Amore, P. A. Signal transduction in vasculogenesis and developmental angiogenesis. Int. J. Dev. Biol. 55, 353-363 (2011).
  178. 146. Gallione, C. J. et al. SMAD4 mutations found in unselected HHT patients. J. Med. Genet. 43, 793-7 (2006).
  179. 162. Owens, G. K., Rabinovitch, P. S. & Schwartz, S. M. Smooth muscle cell hypertrophy versus hyperplasia in hypertension. Proc. Natl. Acad. Sci. 78, 7759-7763 (1981).
  180. 124. Castonguay, R. et al. Soluble endoglin specifically binds bone morphogenetic proteins 9 and 10 via its orphan domain, inhibits blood vessel formation, and suppresses tumor growth. J. Biol. Chem. 286, 30034-30046 (2011).
  181. 123. Townson, S. a et al. Specificity and structure of a high affinity activin receptor-like kinase 1 (ALK1) signaling complex. J. Biol. Chem. 287, 27313-25 (2012).
  182. 256. Toporsian, M. et al. Spontaneous adult-onset pulmonary arterial hypertension attributable to increased endothelial oxidative stress in a murine model of hereditary hemorrhagic telangiectasia. Arterioscler. Thromb. Vasc. Biol. 30, 509-517 (2010).
  183. 183. Moya, I. M. et al. Stalk Cell Phenotype Depends on Integration of Notch and Smad1/5 Signaling Cascades. Dev. Cell 22, 501-514 (2012).
  184. Segers, V. F. M. & Lee, R. T. Stem-cell therapy for cardiac disease. Nature 451, 937-942 (2008).
  185. 279. Li, J., Hampton, T., Morgan, J. P. & Simons, M. Stretch-induced VEGF expression in the heart. J. Clin. Invest. 100, 18-24 (1997).
  186. Sabin, F. Studies on the origin of blood-vessels and of red blood-corpuscles as seen in the living blastoderm of chicks during the second day of incubation. Contrib Embryol 36, 213-259 (1920).
  187. 93. Huang, H., Bhat, A., Woodnutt, G. & Lappe, R. Targeting the ANGPT-TIE2 pathway in malignancy. Nat. Rev. Cancer 10, 575-585 (2010).
  188. 212. Wilcox, J. E. et al. 'Targeting the Heart' in Heart Failure: Myocardial Recovery in Heart Failure With Reduced Ejection Fraction. JACC: Heart Failure 3, 661-669 (2015).
  189. 204. Akhurst, R. J. & Hata, A. Targeting the TGFβ signalling pathway in disease. Nat. Rev. Drug Discov. 11, 790-811 (2012).
  190. 182. Goumans, M.-J., Liu, Z., ten Dijke, P. & Dijke, P. TGF-beta signaling in vascular biology and dysfunction. Cell Res. 19, 116-27 (2009).
  191. 127. Schmierer, B. & Hill, C. S. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat. Rev. Mol. Cell Biol. 8, 970-982 (2007).
  192. 192. González-Núñez, M., Muñoz-Félix, J. M. & López-Novoa, J. M. The ALK-1/Smad1 pathway in cardiovascular physiopathology. A new target for therapy? Biochimica et Biophysica Acta - Molecular Basis of Disease 1832, 1492-1510 (2013).
  193. 246. O'Carroll, A. M., Lolait, S. J., Harris, L. E. & Pope, G. R. The apelin receptor APJ: journey from an orphan to a multifaceted regulator of homeostasis. J Endocrinol 219, (2013).
  194. 88. Lobov, I. B. et al. The DII4/notch pathway controls postangiogenic blood vessel remodeling and regression by modulating vasoconstriction and blood flow. Blood 117, 6728-6737 (2011).
  195. Frangogiannis, N. G. The immune system and cardiac repair. Pharmacol. Res. 58, 88-111 (2008).
  196. 160. Stahl, A. et al. The mouse retina as an angiogenesis model. Investigative Ophthalmology and Visual Science 51, 2813-2826 (2010).
  197. 237. Saint-Geniez, M., Argence, C. B., Knibiehler, B. & Audigier, Y. The msr/apj gene encoding the apelin receptor is an early and specific marker of the venous phenotype in the retinal vasculature. Gene Expr. Patterns 3, 467-472 (2003).
  198. 179. Fischer, A., Schumacher, N., Maier, M., Sendtner, M. & Gessler, M. The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev. 18, 901-911 (2004).
  199. 115. Jiang, H. et al. The Prodomain-Bound Form of Bone Morphogenetic Protein 10 is Biologically Active on Endothelial Cells. JBC (2015).
  200. 109. Weiss, A. & Attisano, L. The TGFbeta superfamily signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 2, 47-63 (2013).
  201. 150. Sirard, C. et al. The tumor suppressor gene Smad4/Dpc4 is required for gastrulation and later for anterior development of the mouse embryo. Genes Dev. 12, 107-119 (1998).
  202. 226. Lacolley, P., Regnault, V., Nicoletti, A., Li, Z. & Michel, J. B. The vascular smooth muscle cell in arterial pathology: A cell that can take on multiple roles. Cardiovascular Research 95, 194- 204 (2012).
  203. 184. Somekawa, S. et al. Tmem100, an ALK1 receptor signaling-dependent gene essential for arterial endothelium differentiation and vascular morphogenesis. Proc. Natl. Acad. Sci. U. S. A. 109, 12064-9 (2012).
  204. 200. Guo, X. & Chen, S.-Y. Transforming growth factor-β and smooth muscle differentiation. World J. Biol. Chem. 3, 41-52 (2012).
  205. 240. Fràter-Schröder, M., Müller, G., Birchmeier, W. & Böhlen, P. Transforming growth factor-beta inhibits endothelial cell proliferation. Biochem. Biophys. Res. Commun. 137, 295-302 (1986).
  206. 178. Daly, A., Randall, R. & Hill, C. Transforming Growth Factor β-Induced Smad1/5 Phosphorylation in Epithelial Cells Is Mediated by Novel Receptor Complexes and Is Essential for Anchorage-Independent Growth. Mol. Cell. Biol 28, 6889-6902 (2008).
  207. 266. DeAlmeida, A., van Oort, R. & Wehrens, X. Transverse Aortic Constriction in Mice. J Vis Exp. 3, 1729 (2010).
  208. 269. Panchenko, M., Williams, M., Brody, K. & Yu, Q. Type I receptor serine-threonine kinase preferentially expressed in pulmonary blood vessels. Am. J. Physiol. 270, L547-L558 (1996).
  209. 100. Osawa, M., Masuda, M., Harada, N., Lopes, R. B. & Fujiwara, K. Tyrosine phosphorylation of platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) in mechanically stimulated vascular endothelial cells. Eur. J. Cell Biol. 72, 229-237 (1997).
  210. 97. Blasi, F. & Carmeliet, P. uPAR: a versatile signalling orchestrator. Nat. Rev. Mol. Cell Biol. 3, 932-943 (2002).
  211. 137. Pepper, M. S., Vassalli, J. D., Montesano, R. & Orci, L. Urokinase-type plasminogen activator is induced in migrating capillary endothelial cells. J Cell Biol 105, 2535-2541 (1987).
  212. 139. Mandriota, S. J. et al. Vascular endothelial growth factor increases urokinase receptor expression in vascular endothelial cells. J. Biol. Chem. 270, 9709-9716 (1995).
  213. 282. Zhao, T., Zhao, W., Chen, Y., Ahokas, R. A. & Sun, Y. Vascular endothelial growth factor (VEGF)-A: Role on cardiac angiogenesis following myocardial infarction. Microvasc. Res. 80, 188-194 (2010).
  214. Reagan, F. Vascularization phenomena in fragments of embryonic bodies completely isolated from yolk-sac blastoderm. Anat. Rec. 9, 329-341 (1915).
  215. 186. Louis, S. F. & Zahradka, P. Vascular smooth muscle cell motility: From migration to invasion. Experimental and Clinical Cardiology 15, (2010).
  216. 234. Gerhardt, H. VEGF and endothelial guidance in angiogenic sprouting. Organogenesis 4, 241- 246 (2008).
  217. 261. Li, J. et al. VEGF, flk-1, and flt-1 expression in a rat myocardial infarction model of angiogenesis. Am. J. Physiol. 270, H1803-11 (1996).
  218. 102. Shay-Salit, A. et al. VEGF receptor 2 and the adherens junction as a mechanical transducer in vascular endothelial cells. Proc. Natl. Acad. Sci. U. S. A. 99, 9462-9467 (2002).
  219. 210. Klabunde, R. Regulation of Stroke Volume. Cardiovascular Physiology Concepts (2015). at <http://www.cvphysiology.com/Cardiac Function/CF002.htm>
  220. Mayo Clinic Staff. Heart Disease. (2014). at <http://www.mayoclinic.org/diseasesconditions/heart-disease/basics/definition/con-20034056> Reinecke, H., Minami, E., Zhu, W.-Z. & Laflamme, M. A. Cardiogenic differentiation and transdifferentiation of progenitor cells. Circ. Res. 103, 1058-71 (2008).
  221. Kelvinsong - Own work, C. B.-S. 3. . Diagram of blood vessel structures. at <https://commons.wikimedia.org/w/index.php?curid=25165240> Risau, W. et al. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development 102, 471-478 (1988).
  222. 133. Strizhak, A. Arteriovenous Malformations (AVM). (2016). at <https://www.doctorstrizhak.com/Arteriovenous_Malformation.php>


* Das Dokument ist im Internet frei zugänglich - Hinweise zu den Nutzungsrechten