Skip to main content
Log in

To be or not to be: endothelial cell plasticity in development, repair, and disease

  • Review Paper
  • Published:
Angiogenesis Aims and scope Submit manuscript

Abstract

Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Dejana E, Hirschi KK, Simons M (2017) The molecular basis of endothelial cell plasticity. Nat Commun 8:14361. doi:https://doi.org/10.1038/ncomms14361

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tata PR, Rajagopal J (2016) Cellular plasticity: 1712 to the present day. Curr Opin Cell Biol 43:46–54. doi:https://doi.org/10.1016/j.ceb.2016.07.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yuan S, Norgard RJ, Stanger BZ (2019) Cellular plasticity in cancer. Cancer Discov 9(7):837–851. https://doi.org/10.1158/2159-8290.CD-19-0015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ellertsdottir E, Lenard A, Blum Y, Krudewig A, Herwig L, Affolter M, Belting HG (2010) Vascular morphogenesis in the zebrafish embryo. Dev Biol 341(1):56–65. doi:https://doi.org/10.1016/j.ydbio.2009.10.035

    Article  CAS  PubMed  Google Scholar 

  5. Gore AV, Monzo K, Cha YR, Pan W, Weinstein BM (2012) Vascular development in the zebrafish. Cold Spring Harb Perspect Med 2(5):a006684. doi:https://doi.org/10.1101/cshperspect.a006684

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hogan BM, Schulte-Merker S (2017) How to plumb a pisces: understanding vascular development and disease using zebrafish embryos. Dev Cell 42(6):567–583. https://doi.org/10.1016/j.devcel.2017.08.015

    Article  CAS  PubMed  Google Scholar 

  7. Isogai S, Horiguchi M, Weinstein BM (2001) The vascular anatomy of the developing zebrafish: an atlas of embryonic and early larval development. Dev Biol 230(2):278–301. doi:https://doi.org/10.1006/dbio.2000.9995

    Article  CAS  PubMed  Google Scholar 

  8. Lawson ND, Weinstein BM (2002) In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol 248(2):307–318. doi:https://doi.org/10.1006/dbio.2002.0711

    Article  CAS  PubMed  Google Scholar 

  9. Isogai S, Lawson ND, Torrealday S, Horiguchi M, Weinstein BM (2003) Angiogenic network formation in the developing vertebrate trunk. Development 130(21):5281–5290. doi:https://doi.org/10.1242/dev.00733

    Article  CAS  PubMed  Google Scholar 

  10. Stainier DY, Weinstein BM, Detrich HW 3rd, Zon LI, Fishman MC (1995) Cloche, an early acting zebrafish gene, is required by both the endothelial and hematopoietic lineages. Development 121(10):3141–3150

    Article  CAS  Google Scholar 

  11. Das RN, Yaniv K (2020) Discovering new progenitor cell populations through lineage tracing and in vivo imaging. Cold Spring Harb Perspect Biol. https://doi.org/10.1101/cshperspect.a035618

    Article  PubMed  Google Scholar 

  12. Bertrand JY, Chi NC, Santoso B, Teng S, Stainier DY, Traver D (2010) Haematopoietic stem cells derive directly from aortic endothelium during development. Nature 464(7285):108–111. doi:https://doi.org/10.1038/nature08738

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gunawan F, Gentile A, Gauvrit S, Stainier DYR, Bensimon-Brito A (2020) Nfatc1 promotes interstitial cell formation during cardiac valve development in zebrafish. Circ Res 126(8):968–984. https://doi.org/10.1161/CIRCRESAHA.119.315992

    Article  CAS  PubMed  Google Scholar 

  14. Kissa K, Herbomel P (2010) Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature 464(7285):112–115. doi:https://doi.org/10.1038/nature08761

    Article  CAS  PubMed  Google Scholar 

  15. Lam EY, Hall CJ, Crosier PS, Crosier KE, Flores MV (2010) Live imaging of Runx1 expression in the dorsal aorta tracks the emergence of blood progenitors from endothelial cells. Blood 116(6):909–914. doi:https://doi.org/10.1182/blood-2010-01-264382

    Article  CAS  PubMed  Google Scholar 

  16. Weijts B, Gutierrez E, Saikin SK, Ablooglu AJ, Traver D, Groisman A, Tkachenko E (2018) Blood flow-induced Notch activation and endothelial migration enable vascular remodeling in zebrafish embryos. Nat Commun 9(1):5314. doi:https://doi.org/10.1038/s41467-018-07732-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yaniv K, Isogai S, Castranova D, Dye L, Hitomi J, Weinstein BM (2006) Live imaging of lymphatic development in the zebrafish. Nat Med 12(6):711–716. doi:https://doi.org/10.1038/nm1427

    Article  CAS  PubMed  Google Scholar 

  18. Xu C, Hasan SS, Schmidt I, Rocha SF, Pitulescu ME, Bussmann J, Meyen D, Raz E, Adams RH, Siekmann AF (2014) Arteries are formed by vein-derived endothelial tip cells. Nat Commun 5:5758. doi:https://doi.org/10.1038/ncomms6758

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gurevich DB, Severn CE, Twomey C, Greenhough A, Cash J, Toye AM, Mellor H, Martin P (2018) Live imaging of wound angiogenesis reveals macrophage orchestrated vessel sprouting and regression. EMBO J. https://doi.org/10.15252/embj.201797786

  20. Marin-Juez R, Marass M, Gauvrit S, Rossi A, Lai SL, Materna SC, Black BL, Stainier DY (2016) Fast revascularization of the injured area is essential to support zebrafish heart regeneration. Proc Natl Acad Sci USA 113(40):11237–11242. https://doi.org/10.1073/pnas.1605431113

    Article  CAS  PubMed  Google Scholar 

  21. Noishiki C, Yuge S, Ando K, Wakayama Y, Mochizuki N, Ogawa R, Fukuhara S (2019) Live imaging of angiogenesis during cutaneous wound healing in adult zebrafish. Angiogenesis 22(2):341–354. doi:https://doi.org/10.1007/s10456-018-09660-y

    Article  PubMed  Google Scholar 

  22. De Val S, Chi NC, Meadows SM, Minovitsky S, Anderson JP, Harris IS, Ehlers ML, Agarwal P, Visel A, Xu SM, Pennacchio LA, Dubchak I, Krieg PA, Stainier DY, Black BL (2008) Combinatorial regulation of endothelial gene expression by ets and forkhead transcription factors. Cell 135(6):1053–1064. doi:https://doi.org/10.1016/j.cell.2008.10.049

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lee D, Park C, Lee H, Lugus JJ, Kim SH, Arentson E, Chung YS, Gomez G, Kyba M, Lin S, Janknecht R, Lim DS, Choi K (2008) ER71 acts downstream of BMP, Notch, and Wnt signaling in blood and vessel progenitor specification. Cell Stem Cell 2(5):497–507. doi:https://doi.org/10.1016/j.stem.2008.03.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pham VN, Lawson ND, Mugford JW, Dye L, Castranova D, Lo B, Weinstein BM (2007) Combinatorial function of ETS transcription factors in the developing vasculature. Dev Biol 303(2):772–783. doi:https://doi.org/10.1016/j.ydbio.2006.10.030

    Article  CAS  PubMed  Google Scholar 

  25. Sumanas S, Lin S (2006) Ets1-related protein is a key regulator of vasculogenesis in zebrafish. PLoS Biol 4(1):e10. doi:https://doi.org/10.1371/journal.pbio.0040010

    Article  CAS  PubMed  Google Scholar 

  26. Veldman MB, Zhao C, Gomez GA, Lindgren AG, Huang H, Yang H, Yao S, Martin BL, Kimelman D, Lin S (2013) Transdifferentiation of fast skeletal muscle into functional endothelium in vivo by transcription factor Etv2. PLoS Biol 11(6):e1001590. doi:https://doi.org/10.1371/journal.pbio.1001590

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Reischauer S, Stone OA, Villasenor A, Chi N, Jin SW, Martin M, Lee MT, Fukuda N, Marass M, Witty A, Fiddes I, Kuo T, Chung WS, Salek S, Lerrigo R, Alsio J, Luo S, Tworus D, Augustine SM, Mucenieks S, Nystedt B, Giraldez AJ, Schroth GP, Andersson O, Stainier DY (2016) Cloche is a bHLH-PAS transcription factor that drives haemato-vascular specification. Nature 535(7611):294–298. doi:https://doi.org/10.1038/nature18614

    Article  CAS  PubMed  Google Scholar 

  28. Garcia-Cardena G, Slegtenhorst BR (2016) Hemodynamic control of endothelial cell fates in development. Annu Rev Cell Dev Biol 32:633–648. https://doi.org/10.1146/annurev-cellbio-100814-125610

    Article  CAS  PubMed  Google Scholar 

  29. Wang HU, Chen ZF, Anderson DJ (1998) Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 93(5):741–753. doi:https://doi.org/10.1016/s0092-8674(00)81436-1

    Article  CAS  PubMed  Google Scholar 

  30. Herzog Y, Guttmann-Raviv N, Neufeld G (2005) Segregation of arterial and venous markers in subpopulations of blood islands before vessel formation. Dev Dyn 232(4):1047–1055. doi:https://doi.org/10.1002/dvdy.20257

    Article  CAS  PubMed  Google Scholar 

  31. Lawson ND, Scheer N, Pham VN, Kim CH, Chitnis AB, Campos-Ortega JA, Weinstein BM (2001) Notch signaling is required for arterial-venous differentiation during embryonic vascular development. Development 128(19):3675–3683

    Article  CAS  Google Scholar 

  32. Lawson ND, Vogel AM, Weinstein BM (2002) sonic hedgehog and vascular endothelial growth factor act upstream of the Notch pathway during arterial endothelial differentiation. Dev Cell 3(1):127–136. doi:https://doi.org/10.1016/s1534-5807(02)00198-3

    Article  CAS  PubMed  Google Scholar 

  33. Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M (2004) The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev 18(8):901–911. doi:https://doi.org/10.1101/gad.291004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela DM, Murphy AJ, Adams NC, Lin HC, Holash J, Thurston G, Yancopoulos GD (2004) Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci USA 101(45):15949–15954. https://doi.org/10.1073/pnas.0407290101

    Article  CAS  PubMed  Google Scholar 

  35. Krebs LT, Xue Y, Norton CR, Shutter JR, Maguire M, Sundberg JP, Gallahan D, Closson V, Kitajewski J, Callahan R, Smith GH, Stark KL, Gridley T (2000) Notch signaling is essential for vascular morphogenesis in mice. Genes Dev 14(11):1343–1352

    CAS  PubMed  PubMed Central  Google Scholar 

  36. You LR, Lin FJ, Lee CT, DeMayo FJ, Tsai MJ, Tsai SY (2005) Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity. Nature 435(7038):98–104. doi:https://doi.org/10.1038/nature03511

    Article  CAS  PubMed  Google Scholar 

  37. Swift MR, Pham VN, Castranova D, Bell K, Poole RJ, Weinstein BM (2014) SoxF factors and Notch regulate nr2f2 gene expression during venous differentiation in zebrafish. Dev Biol 390(2):116–125. doi:https://doi.org/10.1016/j.ydbio.2014.03.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Neal A, Nornes S, Payne S, Wallace MD, Fritzsche M, Louphrasitthiphol P, Wilkinson RN, Chouliaras KM, Liu K, Plant K, Sholapurkar R, Ratnayaka I, Herzog W, Bond G, Chico T, Bou-Gharios G, De Val S (2019) Venous identity requires BMP signalling through ALK3. Nat Commun 10(1):453. doi:https://doi.org/10.1038/s41467-019-08315-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bai H, Wang Z, Li M, Sun P, Wei S, Wang Z, Xing Y, Dardik A (2020) Adult human vein grafts retain plasticity of vessel identity. Ann Vasc Surg. https://doi.org/10.1016/j.avsg.2020.04.046

    Article  PubMed  Google Scholar 

  40. Moyon D, Pardanaud L, Yuan L, Breant C, Eichmann A (2001) Plasticity of endothelial cells during arterial-venous differentiation in the avian embryo. Development 128(17):3359–3370

    Article  CAS  Google Scholar 

  41. Claxton S, Fruttiger M (2005) Oxygen modifies artery differentiation and network morphogenesis in the retinal vasculature. Dev Dyn 233(3):822–828. doi:https://doi.org/10.1002/dvdy.20407

    Article  CAS  PubMed  Google Scholar 

  42. le Noble F, Moyon D, Pardanaud L, Yuan L, Djonov V, Matthijsen R, Breant C, Fleury V, Eichmann A (2004) Flow regulates arterial-venous differentiation in the chick embryo yolk sac. Development 131(2):361–375. doi:https://doi.org/10.1242/dev.00929

    Article  CAS  PubMed  Google Scholar 

  43. Fang JS, Coon BG, Gillis N, Chen Z, Qiu J, Chittenden TW, Burt JM, Schwartz MA, Hirschi KK (2017) Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat Commun 8(1):2149. doi:https://doi.org/10.1038/s41467-017-01742-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mack JJ, Mosqueiro TS, Archer BJ, Jones WM, Sunshine H, Faas GC, Briot A, Aragon RL, Su T, Romay MC, McDonald AI, Kuo CH, Lizama CO, Lane TF, Zovein AC, Fang Y, Tarling EJ, de A Vallim, Navab TQ, Fogelman M, Bouchard AM, Iruela-Arispe LS ML (2017) NOTCH1 is a mechanosensor in adult arteries. Nat Commun 8(1):1620. doi:https://doi.org/10.1038/s41467-017-01741-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Garcia MD, Larina IV (2014) Vascular development and hemodynamic force in the mouse yolk sac. Front Physiol 5:308. doi:https://doi.org/10.3389/fphys.2014.00308

    Article  PubMed  PubMed Central  Google Scholar 

  46. Gore AV, Pillay LM, Venero Galanternik M, Weinstein BM (2018) The zebrafish: A fintastic model for hematopoietic development and disease. Wiley Interdiscip Rev Dev Biol 7(3):e312. doi:https://doi.org/10.1002/wdev.312

    Article  PubMed  PubMed Central  Google Scholar 

  47. Zovein AC, Hofmann JJ, Lynch M, French WJ, Turlo KA, Yang Y, Becker MS, Zanetta L, Dejana E, Gasson JC, Tallquist MD, Iruela-Arispe ML (2008) Fate tracing reveals the endothelial origin of hematopoietic stem cells. Cell Stem Cell 3(6):625–636. doi:https://doi.org/10.1016/j.stem.2008.09.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chanda B, Ditadi A, Iscove NN, Keller G (2013) Retinoic acid signaling is essential for embryonic hematopoietic stem cell development. Cell 155(1):215–227. doi:https://doi.org/10.1016/j.cell.2013.08.055

    Article  CAS  PubMed  Google Scholar 

  49. Goldie LC, Lucitti JL, Dickinson ME, Hirschi KK (2008) Cell signaling directing the formation and function of hemogenic endothelium during murine embryogenesis. Blood 112(8):3194–3204. doi:https://doi.org/10.1182/blood-2008-02-139055

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Marcelo KL, Sills TM, Coskun S, Vasavada H, Sanglikar S, Goldie LC, Hirschi KK (2013) Hemogenic endothelial cell specification requires c-Kit, Notch signaling, and p27-mediated cell-cycle control. Dev Cell 27(5):504–515. doi:https://doi.org/10.1016/j.devcel.2013.11.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Burns CE, Traver D, Mayhall E, Shepard JL, Zon LI (2005) Hematopoietic stem cell fate is established by the Notch-Runx pathway. Genes Dev 19(19):2331–2342. doi:https://doi.org/10.1101/gad.1337005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kumano K, Chiba S, Kunisato A, Sata M, Saito T, Nakagami-Yamaguchi E, Yamaguchi T, Masuda S, Shimizu K, Takahashi T, Ogawa S, Hamada Y, Hirai H (2003) Notch1 but not Notch2 is essential for generating hematopoietic stem cells from endothelial cells. Immunity 18(5):699–711. doi:https://doi.org/10.1016/s1074-7613(03)00117-1

    Article  CAS  PubMed  Google Scholar 

  53. Robert-Moreno A, Guiu J, Ruiz-Herguido C, Lopez ME, Ingles-Esteve J, Riera L, Tipping A, Enver T, Dzierzak E, Gridley T, Espinosa L, Bigas A (2008) Impaired embryonic haematopoiesis yet normal arterial development in the absence of the Notch ligand Jagged1. EMBO J 27(13):1886–1895. doi:https://doi.org/10.1038/emboj.2008.113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gama-Norton L, Ferrando E, Ruiz-Herguido C, Liu Z, Guiu J, Islam AB, Lee SU, Yan M, Guidos CJ, Lopez-Bigas N, Maeda T, Espinosa L, Kopan R, Bigas A (2015) Notch signal strength controls cell fate in the haemogenic endothelium. Nat Commun 6:8510. doi:https://doi.org/10.1038/ncomms9510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen MJ, Yokomizo T, Zeigler BM, Dzierzak E, Speck NA (2009) Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature 457(7231):887–891. doi:https://doi.org/10.1038/nature07619

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lieu YK, Reddy EP (2009) Conditional c-myb knockout in adult hematopoietic stem cells leads to loss of self-renewal due to impaired proliferation and accelerated differentiation. Proc Natl Acad Sci USA 106(51):21689–21694. https://doi.org/10.1073/pnas.0907623106

    Article  PubMed  Google Scholar 

  57. Soza-Ried C, Hess I, Netuschil N, Schorpp M, Boehm T (2010) Essential role of c-myb in definitive hematopoiesis is evolutionarily conserved. Proc Natl Acad Sci U S A 107(40):17304–17308. doi:https://doi.org/10.1073/pnas.1004640107

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhang Y, Jin H, Li L, Qin FX, Wen Z (2011) cMyb regulates hematopoietic stem/progenitor cell mobilization during zebrafish hematopoiesis. Blood 118(15):4093–4101. doi:https://doi.org/10.1182/blood-2011-03-342501

    Article  CAS  PubMed  Google Scholar 

  59. Gore AV, Athans B, Iben JR, Johnson K, Russanova V, Castranova D, Pham VN, Butler MG, Williams-Simons L, Nichols JT, Bresciani E, Feldman B, Kimmel CB, Liu PP, Weinstein BM (2016) Epigenetic regulation of hematopoiesis by DNA methylation. Elife 5:e11813. doi:https://doi.org/10.7554/eLife.11813

    Article  PubMed  PubMed Central  Google Scholar 

  60. Yvernogeau L, Gautier R, Petit L, Khoury H, Relaix F, Ribes V, Sang H, Charbord P, Souyri M, Robin C, Jaffredo T (2019) In vivo generation of haematopoietic stem/progenitor cells from bone marrow-derived haemogenic endothelium. Nat Cell Biol 21(11):1334–1345. doi:https://doi.org/10.1038/s41556-019-0410-6

    Article  CAS  PubMed  Google Scholar 

  61. Srinivasan RS, Dillard ME, Lagutin OV, Lin FJ, Tsai S, Tsai MJ, Samokhvalov IM, Oliver G (2007) Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev 21(19):2422–2432. doi:https://doi.org/10.1101/gad.1588407

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hong YK, Harvey N, Noh YH, Schacht V, Hirakawa S, Detmar M, Oliver G (2002) Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate. Dev Dyn 225(3):351–357. doi:https://doi.org/10.1002/dvdy.10163

    Article  CAS  PubMed  Google Scholar 

  63. Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Yla-Herttuala S, Alitalo K (2002) Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. EMBO J 21(17):4593–4599. doi:https://doi.org/10.1093/emboj/cdf470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wigle JT, Harvey N, Detmar M, Lagutina I, Grosveld G, Gunn MD, Jackson DG, Oliver G (2002) An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J 21(7):1505–1513. doi:https://doi.org/10.1093/emboj/21.7.1505

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wigle JT, Oliver G (1999) Prox1 function is required for the development of the murine lymphatic system. Cell 98(6):769–778. doi:https://doi.org/10.1016/s0092-8674(00)81511-1

    Article  CAS  PubMed  Google Scholar 

  66. Koltowska K, Lagendijk AK, Pichol-Thievend C, Fischer JC, Francois M, Ober EA, Yap AS, Hogan BM (2015) Vegfc regulates bipotential precursor division and Prox1 expression to promote lymphatic identity in zebrafish. Cell Rep 13(9):1828–1841. https://doi.org/10.1016/j.celrep.2015.10.055

    Article  CAS  PubMed  Google Scholar 

  67. Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, Jeltsch M, Jackson DG, Talikka M, Rauvala H, Betsholtz C, Alitalo K (2004) Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5(1):74–80. doi:https://doi.org/10.1038/ni1013

    Article  CAS  PubMed  Google Scholar 

  68. Srinivasan RS, Escobedo N, Yang Y, Interiano A, Dillard ME, Finkelstein D, Mukatira S, Gil HJ, Nurmi H, Alitalo K, Oliver G (2014) The Prox1-Vegfr3 feedback loop maintains the identity and the number of lymphatic endothelial cell progenitors. Genes Dev 28(19):2175–2187. doi:https://doi.org/10.1101/gad.216226.113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Johnson NC, Dillard ME, Baluk P, McDonald DM, Harvey NL, Frase SL, Oliver G (2008) Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity. Genes Dev 22(23):3282–3291. doi:https://doi.org/10.1101/gad.1727208

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chen CY, Bertozzi C, Zou Z, Yuan L, Lee JS, Lu M, Stachelek SJ, Srinivasan S, Guo L, Vicente A, Mericko P, Levy RJ, Makinen T, Oliver G, Kahn ML (2012) Blood flow reprograms lymphatic vessels to blood vessels. J Clin Invest 122(6):2006–2017. doi:https://doi.org/10.1172/JCI57513

    Article  PubMed  PubMed Central  Google Scholar 

  71. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K (2015) A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 212(7):991–999. doi:https://doi.org/10.1084/jem.20142290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J (2015) Structural and functional features of central nervous system lymphatic vessels. Nature 523(7560):337–341. doi:https://doi.org/10.1038/nature14432

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Castranova D, Samasa B, Venero Galanternik M, Jung HM, Pham VN, Weinstein BM (2020) Live Imaging of Intracranial Lymphatics in the Zebrafish. Circ Res. doi:https://doi.org/10.1161/CIRCRESAHA.120.317372

    Article  PubMed  Google Scholar 

  74. Okuda KS, Astin JW, Misa JP, Flores MV, Crosier KE, Crosier PS (2012) lyve1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish. Development 139(13):2381–2391. doi:https://doi.org/10.1242/dev.077701

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Bower NI, Koltowska K, Pichol-Thievend C, Virshup I, Paterson S, Lagendijk AK, Wang W, Lindsey BW, Bent SJ, Baek S, Rondon-Galeano M, Hurley DG, Mochizuki N, Simons C, Francois M, Wells CA, Kaslin J, Hogan BM (2017) Mural lymphatic endothelial cells regulate meningeal angiogenesis in the zebrafish. Nat Neurosci 20(6):774–783. doi:https://doi.org/10.1038/nn.4558

    Article  CAS  PubMed  Google Scholar 

  76. van Lessen M, Shibata-Germanos S, van Impel A, Hawkins TA, Rihel J, Schulte-Merker S (2017) Intracellular uptake of macromolecules by brain lymphatic endothelial cells during zebrafish embryonic development. Elife. https://doi.org/10.7554/eLife.25932

    Article  PubMed  PubMed Central  Google Scholar 

  77. Venero Galanternik M, Castranova D, Gore AV, Blewett NH, Jung HM, Stratman AN, Kirby MR, Iben J, Miller MF, Kawakami K, Maraia RJ, Weinstein BM (2017) A novel perivascular cell population in the zebrafish brain. Elife. https://doi.org/10.7554/eLife.24369

    Article  PubMed  Google Scholar 

  78. Beis D, Bartman T, Jin SW, Scott IC, D’Amico LA, Ober EA, Verkade H, Frantsve J, Field HA, Wehman A, Baier H, Tallafuss A, Bally-Cuif L, Chen JN, Stainier DY, Jungblut B (2005) Genetic and cellular analyses of zebrafish atrioventricular cushion and valve development. Development 132(18):4193–4204. doi:https://doi.org/10.1242/dev.01970

    Article  CAS  PubMed  Google Scholar 

  79. Chen IH, Wang HH, Hsieh YS, Huang WC, Yeh HI, Chuang YJ (2013) PRSS23 is essential for the Snail-dependent endothelial-to-mesenchymal transition during valvulogenesis in zebrafish. Cardiovasc Res 97(3):443–453. doi:https://doi.org/10.1093/cvr/cvs355

    Article  CAS  PubMed  Google Scholar 

  80. de Lange FJ, Moorman AF, Anderson RH, Manner J, Soufan AT, de Gier-de Vries C, Schneider MD, Webb S, van den Hoff MJ, Christoffels VM (2004) Lineage and morphogenetic analysis of the cardiac valves. Circ Res 95(6):645–654. doi:https://doi.org/10.1161/01.RES.0000141429.13560.cb

    Article  CAS  PubMed  Google Scholar 

  81. Lincoln J, Alfieri CM, Yutzey KE (2004) Development of heart valve leaflets and supporting apparatus in chicken and mouse embryos. Dev Dyn 230(2):239–250. doi:https://doi.org/10.1002/dvdy.20051

    Article  CAS  PubMed  Google Scholar 

  82. Garside VC, Chang AC, Karsan A, Hoodless PA (2013) Co-ordinating Notch, BMP, and TGF-beta signaling during heart valve development. Cell Mol Life Sci 70(16):2899–2917. doi:https://doi.org/10.1007/s00018-012-1197-9

    Article  CAS  PubMed  Google Scholar 

  83. Ma L, Lu MF, Schwartz RJ, Martin JF (2005) Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterning. Development 132(24):5601–5611. doi:https://doi.org/10.1242/dev.02156

    Article  CAS  PubMed  Google Scholar 

  84. Azhar M, Runyan RB, Gard C, Sanford LP, Miller ML, Andringa A, Pawlowski S, Rajan S, Doetschman T (2009) Ligand-specific function of transforming growth factor beta in epithelial-mesenchymal transition in heart development. Dev Dyn 238(2):431–442. doi:https://doi.org/10.1002/dvdy.21854

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Timmerman LA, Grego-Bessa J, Raya A, Bertran E, Perez-Pomares JM, Diez J, Aranda S, Palomo S, McCormick F, Izpisua-Belmonte JC, de la Pompa JL (2004) Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev 18(1):99–115. doi:https://doi.org/10.1101/gad.276304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chang CP, Neilson JR, Bayle JH, Gestwicki JE, Kuo A, Stankunas K, Graef IA, Crabtree GR (2004) A field of myocardial-endocardial NFAT signaling underlies heart valve morphogenesis. Cell 118(5):649–663. doi:https://doi.org/10.1016/j.cell.2004.08.010

    Article  CAS  PubMed  Google Scholar 

  87. Wu B, Wang Y, Lui W, Langworthy M, Tompkins KL, Hatzopoulos AK, Baldwin HS, Zhou B (2011) Nfatc1 coordinates valve endocardial cell lineage development required for heart valve formation. Circ Res 109(2):183–192. doi:https://doi.org/10.1161/CIRCRESAHA.111.245035

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Bensimon-Brito A, Ramkumar S, Boezio GLM, Guenther S, Kuenne C, Helker CSM, Sanchez-Iranzo H, Iloska D, Piesker J, Pullamsetti S, Mercader N, Beis D, Stainier DYR (2020) TGF-beta signaling promotes tissue formation during cardiac valve regeneration in adult zebrafish. Dev Cell 52(1):9-20 e27. https://doi.org/10.1016/j.devcel.2019.10.027

    Article  CAS  PubMed  Google Scholar 

  89. Bischoff J, Aikawa E (2011) Progenitor cells confer plasticity to cardiac valve endothelium. J Cardiovasc Transl Res 4(6):710–719. doi:https://doi.org/10.1007/s12265-011-9312-0

    Article  PubMed  Google Scholar 

  90. Kametani Y, Chi NC, Stainier DY, Takada S (2015) Notch signaling regulates venous arterialization during zebrafish fin regeneration. Genes Cells 20(5):427–438. doi:https://doi.org/10.1111/gtc.12234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Red-Horse K, Siekmann AF (2019) Veins and arteries build hierarchical branching patterns differently: bottom-up versus top-down. Bioessays 41(3):e1800198. https://doi.org/10.1002/bies.201800198

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kudo FA, Muto A, Maloney SP, Pimiento JM, Bergaya S, Fitzgerald TN, Westvik TS, Frattini JC, Breuer CK, Cha CH, Nishibe T, Tellides G, Sessa WC, Dardik A (2007) Venous identity is lost but arterial identity is not gained during vein graft adaptation. Arterioscler Thromb Vasc Biol 27(7):1562–1571. doi:https://doi.org/10.1161/ATVBAHA.107.143032

    Article  CAS  PubMed  Google Scholar 

  93. Harrison MR, Bussmann J, Huang Y, Zhao L, Osorio A, Burns CG, Burns CE, Sucov HM, Siekmann AF, Lien CL (2015) Chemokine-guided angiogenesis directs coronary vasculature formation in zebrafish. Dev Cell 33(4):442–454. doi:https://doi.org/10.1016/j.devcel.2015.04.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Marin-Juez R, El-Sammak H, Helker CSM, Kamezaki A, Mullapuli ST, Bibli SI, Foglia MJ, Fleming I, Poss KD, Stainier DYR (2019) Coronary revascularization during heart regeneration is regulated by epicardial and endocardial cues and forms a scaffold for cardiomyocyte repopulation. Dev Cell 51(4):503-515 e504. https://doi.org/10.1016/j.devcel.2019.10.019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhao L, Borikova AL, Ben-Yair R, Guner-Ataman B, MacRae CA, Lee RT, Burns CG, Burns CE (2014) Notch signaling regulates cardiomyocyte proliferation during zebrafish heart regeneration. Proc Natl Acad Sci U S A 111(4):1403–1408. doi:https://doi.org/10.1073/pnas.1311705111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Miquerol L, Thireau J, Bideaux P, Sturny R, Richard S, Kelly RG (2015) Endothelial plasticity drives arterial remodeling within the endocardium after myocardial infarction. Circ Res 116(11):1765–1771. doi:https://doi.org/10.1161/CIRCRESAHA.116.306476

    Article  CAS  PubMed  Google Scholar 

  97. Ubil E, Duan J, Pillai IC, Rosa-Garrido M, Wu Y, Bargiacchi F, Lu Y, Stanbouly S, Huang J, Rojas M, Vondriska TM, Stefani E, Deb A (2014) Mesenchymal-endothelial transition contributes to cardiac neovascularization. Nature 514(7524):585–590. doi:https://doi.org/10.1038/nature13839

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, Pu W, Liu Q, Zhang H, Tian X, Zhao H, Liu X, Zhang S, Nie Y, Hu S, Miao X, Wang QD, Wang F, Chen T, Xu Q, Lui KO, Molkentin JD, Zhou B (2017) Preexisting endothelial cells mediate cardiac neovascularization after injury. J Clin Invest 127(8):2968–2981. doi:https://doi.org/10.1172/JCI93868

    Article  PubMed  PubMed Central  Google Scholar 

  99. Tang J, Zhang H, He L, Huang X, Li Y, Pu W, Yu W, Zhang L, Cai D, Lui KO, Zhou B (2018) Genetic Fate Mapping Defines the Vascular Potential of Endocardial Cells in the Adult Heart. Circ Res 122(7):984–993. doi:https://doi.org/10.1161/CIRCRESAHA.117.312354

    Article  CAS  PubMed  Google Scholar 

  100. Chen HI, Sharma B, Akerberg BN, Numi HJ, Kivela R, Saharinen P, Aghajanian H, McKay AS, Bogard PE, Chang AH, Jacobs AH, Epstein JA, Stankunas K, Alitalo K, Red-Horse K (2014) The sinus venosus contributes to coronary vasculature through VEGFC-stimulated angiogenesis. Development 141(23):4500–4512. doi:https://doi.org/10.1242/dev.113639

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M, Magner M, Isner JM (1999) Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 85(3):221–228. doi:https://doi.org/10.1161/01.res.85.3.221

    Article  CAS  PubMed  Google Scholar 

  102. Crosby JR, Kaminski WE, Schatteman G, Martin PJ, Raines EW, Seifert RA, Bowen-Pope DF (2000) Endothelial cells of hematopoietic origin make a significant contribution to adult blood vessel formation. Circ Res 87(9):728–730. doi:https://doi.org/10.1161/01.res.87.9.728

    Article  CAS  PubMed  Google Scholar 

  103. Guo X, Liu L, Zhang M, Bergeron A, Cui Z, Dong JF, Zhang J (2009) Correlation of CD34 + cells with tissue angiogenesis after traumatic brain injury in a rat model. J Neurotrauma 26(8):1337–1344. doi:https://doi.org/10.1089/neu.2008-073310.1089/neu.2008.0733

    Article  PubMed  PubMed Central  Google Scholar 

  104. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S (2001) Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 7(4):430–436. doi:https://doi.org/10.1038/86498

    Article  CAS  PubMed  Google Scholar 

  105. Urbich C, Dimmeler S (2004) Endothelial progenitor cells: characterization and role in vascular biology. Circ Res 95(4):343–353. doi:https://doi.org/10.1161/01.RES.0000137877.89448.78

    Article  CAS  PubMed  Google Scholar 

  106. Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Jung S, Chimenti S, Landsman L, Abramovitch R, Keshet E (2006) VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 124(1):175–189. doi:https://doi.org/10.1016/j.cell.2005.10.036

    Article  CAS  PubMed  Google Scholar 

  107. Ziegelhoeffer T, Fernandez B, Kostin S, Heil M, Voswinckel R, Helisch A, Schaper W (2004) Bone marrow-derived cells do not incorporate into the adult growing vasculature. Circ Res 94(2):230–238. doi:https://doi.org/10.1161/01.RES.0000110419.50982.1C

    Article  CAS  PubMed  Google Scholar 

  108. Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs K, Ghaeni L, Milosevic M, Bohm M, Nickenig G (2003) Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation. Circulation 107(24):3059–3065. doi:https://doi.org/10.1161/01.CIR.0000077911.81151.30

    Article  CAS  PubMed  Google Scholar 

  109. Hagensen MK, Raarup MK, Mortensen MB, Thim T, Nyengaard JR, Falk E, Bentzon JF (2012) Circulating endothelial progenitor cells do not contribute to regeneration of endothelium after murine arterial injury. Cardiovasc Res 93(2):223–231. doi:https://doi.org/10.1093/cvr/cvr278

    Article  CAS  PubMed  Google Scholar 

  110. Tsuzuki M (2009) Bone marrow-derived cells are not involved in reendothelialized endothelium as endothelial cells after simple endothelial denudation in mice. Basic Res Cardiol 104(5):601–611. doi:https://doi.org/10.1007/s00395-009-0021-7

    Article  PubMed  Google Scholar 

  111. Fadini GP, Agostini C, Avogaro A (2010) Autologous stem cell therapy for peripheral arterial disease meta-analysis and systematic review of the literature. Atherosclerosis 209(1):10–17. doi:https://doi.org/10.1016/j.atherosclerosis.2009.08.033

    Article  CAS  PubMed  Google Scholar 

  112. Gyongyosi M, Haller PM, Blake DJ, Martin Rendon E (2018) Meta-analysis of cell therapy studies in heart failure and acute myocardial infarction. Circ Res 123(2):301–308. https://doi.org/10.1161/CIRCRESAHA.117.311302

    Article  CAS  PubMed  Google Scholar 

  113. Gancz D, Raftrey BC, Perlmoter G, Marin-Juez R, Semo J, Matsuoka RL, Karra R, Raviv H, Moshe N, Addadi Y, Golani O, Poss KD, Red-Horse K, Stainier DY, Yaniv K (2019) Distinct origins and molecular mechanisms contribute to lymphatic formation during cardiac growth and regeneration. Elife 8. doi:https://doi.org/10.7554/eLife.44153

  114. Harrison MR, Feng X, Mo G, Aguayo A, Villafuerte J, Yoshida T, Pearson CA, Schulte-Merker S, Lien CL (2019) Late developing cardiac lymphatic vasculature supports adult zebrafish heart function and regeneration. Elife 8. doi:https://doi.org/10.7554/eLife.42762

  115. Henri O, Pouehe C, Houssari M, Galas L, Nicol L, Edwards-Levy F, Henry JP, Dumesnil A, Boukhalfa I, Banquet S, Schapman D, Thuillez C, Richard V, Mulder P, Brakenhielm E (2016) Selective Stimulation of Cardiac Lymphangiogenesis Reduces Myocardial Edema and Fibrosis Leading to Improved Cardiac Function Following Myocardial Infarction. Circulation 133(15):1484–1497. doi:https://doi.org/10.1161/CIRCULATIONAHA.115.020143 discussion 1497.

    Article  CAS  PubMed  Google Scholar 

  116. Klotz L, Norman S, Vieira JM, Masters M, Rohling M, Dube KN, Bollini S, Matsuzaki F, Carr CA, Riley PR (2015) Cardiac lymphatics are heterogeneous in origin and respond to injury. Nature 522(7554):62–67. doi:https://doi.org/10.1038/nature14483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Chen J, He J, Ni R, Yang Q, Zhang Y, Luo L (2019) Cerebrovascular Injuries Induce Lymphatic Invasion into Brain Parenchyma to Guide Vascular Regeneration in Zebrafish. Dev Cell 49(5):697–710 e695. doi:https://doi.org/10.1016/j.devcel.2019.03.022

    Article  CAS  PubMed  Google Scholar 

  118. Yanev P, Poinsatte K, Hominick D, Khurana N, Zuurbier KR, Berndt M, Plautz EJ, Dellinger MT, Stowe AM (2020) Impaired meningeal lymphatic vessel development worsens stroke outcome. J Cereb Blood Flow Metab 40(2):263–275. doi:https://doi.org/10.1177/0271678X18822921

    Article  PubMed  Google Scholar 

  119. Dal-Bianco JP, Aikawa E, Bischoff J, Guerrero JL, Handschumacher MD, Sullivan S, Johnson B, Titus JS, Iwamoto Y, Wylie-Sears J, Levine RA, Carpentier A (2009) Active adaptation of the tethered mitral valve: insights into a compensatory mechanism for functional mitral regurgitation. Circulation 120(4):334–342. doi:https://doi.org/10.1161/CIRCULATIONAHA.108.846782

    Article  PubMed  PubMed Central  Google Scholar 

  120. Fang J, Hirschi K (2019) Molecular regulation of arteriovenous endothelial cell specification. F1000Res 8. doi:https://doi.org/10.12688/f1000research.16701.1

  121. Krebs LT, Starling C, Chervonsky AV, Gridley T (2010) Notch1 activation in mice causes arteriovenous malformations phenocopied by ephrinB2 and EphB4 mutants. Genesis 48(3):146–150. doi:https://doi.org/10.1002/dvg.20599

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Murphy PA, Kim TN, Huang L, Nielsen CM, Lawton MT, Adams RH, Schaffer CB, Wang RA (2014) Constitutively active Notch4 receptor elicits brain arteriovenous malformations through enlargement of capillary-like vessels. Proc Natl Acad Sci U S A 111(50):18007–18012. doi:https://doi.org/10.1073/pnas.1415316111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Amyere M, Revencu N, Helaers R, Pairet E, Baselga E, Cordisco M, Chung W, Dubois J, Lacour JP, Martorell L, Mazereeuw-Hautier J, Pyeritz RE, Amor DJ, Bisdorff A, Blei F, Bombei H, Dompmartin A, Brooks D, Dupont J, Gonzalez-Ensenat MA, Frieden I, Gerard M, Kvarnung M, Hanson-Kahn AK, Hudgins L, Leaute-Labreze C, McCuaig C, Metry D, Parent P, Paul C, Petit F, Phan A, Quere I, Salhi A, Turner A, Vabres P, Vicente A, Wargon O, Watanabe S, Weibel L, Wilson A, Willing M, Mulliken JB, Boon LM, Vikkula M (2017) Germline loss-of-function mutations in EPHB4 cause a second form of capillary malformation-arteriovenous malformation (CM-AVM2) deregulating RAS-MAPK signaling. Circulation 136(11):1037–1048. https://doi.org/10.1161/CIRCULATIONAHA.116.026886

    Article  CAS  PubMed  Google Scholar 

  124. Whitehead KJ, Smith MC, Li DY (2013) Arteriovenous malformations and other vascular malformation syndromes. Cold Spring Harb Perspect Med 3(2):a006635. doi:https://doi.org/10.1101/cshperspect.a006635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Sorensen LK, Brooke BS, Li DY, Urness LD (2003) Loss of distinct arterial and venous boundaries in mice lacking endoglin, a vascular-specific TGFbeta coreceptor. Dev Biol 261(1):235–250. doi:https://doi.org/10.1016/s0012-1606(03)00158-1

    Article  CAS  PubMed  Google Scholar 

  126. Piera-Velazquez S, Jimenez SA (2019) Endothelial to mesenchymal transition: role in physiology and in the pathogenesis of human diseases. Physiol Rev 99(2):1281–1324. https://doi.org/10.1152/physrev.00021.2018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Manetti M, Romano E, Rosa I, Guiducci S, Bellando-Randone S, De Paulis A, Ibba-Manneschi L, Matucci-Cerinic M (2017) Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann Rheum Dis 76(5):924–934. doi:https://doi.org/10.1136/annrheumdis-2016-210229

    Article  CAS  PubMed  Google Scholar 

  128. Mendoza FA, Piera-Velazquez S, Farber JL, Feghali-Bostwick C, Jimenez SA (2016) Endothelial cells expressing endothelial and mesenchymal cell gene products in lung tissue from patients with systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol 68(1):210–217. https://doi.org/10.1002/art.39421

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Maddaluno L, Rudini N, Cuttano R, Bravi L, Giampietro C, Corada M, Ferrarini L, Orsenigo F, Papa E, Boulday G, Tournier-Lasserve E, Chapon F, Richichi C, Retta SF, Lampugnani MG, Dejana E (2013) EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature 498(7455):492–496. doi:https://doi.org/10.1038/nature12207

    Article  CAS  PubMed  Google Scholar 

  130. Bravi L, Malinverno M, Pisati F, Rudini N, Cuttano R, Pallini R, Martini M, Larocca LM, Locatelli M, Levi V, Bertani GA, Dejana E, Lampugnani MG (2016) Endothelial cells lining sporadic cerebral cavernous malformation cavernomas undergo endothelial-to-mesenchymal transition. Stroke 47(3):886–890. https://doi.org/10.1161/STROKEAHA.115.011867

    Article  PubMed  Google Scholar 

  131. Zheng X, Xu C, Di Lorenzo A, Kleaveland B, Zou Z, Seiler C, Chen M, Cheng L, Xiao J, He J, Pack MA, Sessa WC, Kahn ML (2010) CCM3 signaling through sterile 20-like kinases plays an essential role during zebrafish cardiovascular development and cerebral cavernous malformations. J Clin Invest 120(8):2795–2804. doi:https://doi.org/10.1172/JCI39679

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Otten C, Knox J, Boulday G, Eymery M, Haniszewski M, Neuenschwander M, Radetzki S, Vogt I, Hahn K, De Luca C, Cardoso C, Hamad S, Igual Gil C, Roy P, Albiges-Rizo C, Faurobert E, von Kries JP, Campillos M, Tournier-Lasserve E, Derry WB, Abdelilah-Seyfried S (2018) Systematic pharmacological screens uncover novel pathways involved in cerebral cavernous malformations. EMBO Mol Med. https://doi.org/10.15252/emmm.201809155

  133. Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, Tellides G, Schwartz MA, Simons M (2015) Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest 125(12):4514–4528. doi:https://doi.org/10.1172/JCI82719

    Article  PubMed  PubMed Central  Google Scholar 

  134. Evrard SM, Lecce L, Michelis KC, Nomura-Kitabayashi A, Pandey G, Purushothaman KR, d’Escamard V, Li JR, Hadri L, Fujitani K, Moreno PR, Benard L, Rimmele P, Cohain A, Mecham B, Randolph GJ, Nabel EG, Hajjar R, Fuster V, Boehm M, Kovacic JC (2016) Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat Commun 7:11853. doi:https://doi.org/10.1038/ncomms11853

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Li Y, Zhong C, Liu D, Yu W, Chen W, Wang Y, Shi S, Yuan Y (2018) Evidence for kaposi sarcoma originating from mesenchymal stem cell through KSHV-induced mesenchymal-to-endothelial transition. Cancer Res 78(1):230–245. https://doi.org/10.1158/0008-5472.CAN-17-1961

    Article  CAS  PubMed  Google Scholar 

  136. Wang HW, Trotter MW, Lagos D, Bourboulia D, Henderson S, Makinen T, Elliman S, Flanagan AM, Alitalo K, Boshoff C (2004) Kaposi sarcoma herpesvirus-induced cellular reprogramming contributes to the lymphatic endothelial gene expression in Kaposi sarcoma. Nat Genet 36(7):687–693. doi:https://doi.org/10.1038/ng1384

    Article  CAS  PubMed  Google Scholar 

  137. Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K, Kerjaschki D (1999) Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol 154(2):385–394. doi:https://doi.org/10.1016/S0002-9440(10)65285-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Morris VA, Punjabi AS, Lagunoff M (2008) Activation of Akt through gp130 receptor signaling is required for Kaposi’s sarcoma-associated herpesvirus-induced lymphatic reprogramming of endothelial cells. J Virol 82(17):8771–8779. doi:https://doi.org/10.1128/JVI.00766-08

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Flamini V, Jiang WG, Lane J, Cui YX (2016) Significance and therapeutic implications of endothelial progenitor cells in angiogenic-mediated tumour metastasis. Crit Rev Oncol Hematol 100:177–189. doi:https://doi.org/10.1016/j.critrevonc.2016.02.010

    Article  PubMed  Google Scholar 

  140. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A, Heissig B, Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett NR, Crystal RG, Moore MA, Hajjar KA, Manova K, Benezra R, Rafii S (2001) Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 7(11):1194–1201. doi:https://doi.org/10.1038/nm1101-1194

    Article  CAS  PubMed  Google Scholar 

  141. Yu D, Sun X, Qiu Y, Zhou J, Wu Y, Zhuang L, Chen J, Ding Y (2007) Identification and clinical significance of mobilized endothelial progenitor cells in tumor vasculogenesis of hepatocellular carcinoma. Clin Cancer Res 13(13):3814–3824. doi:https://doi.org/10.1158/1078-0432.CCR-06-2594

    Article  CAS  PubMed  Google Scholar 

  142. Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, Trent JM, Meltzer PS, Hendrix MJ (1999) Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155(3):739–752. doi:https://doi.org/10.1016/S0002-9440(10)65173-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Sun B, Zhang D, Zhao N, Zhao X (2017) Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 8(18):30502–30510. doi:https://doi.org/10.18632/oncotarget.8461

    Article  PubMed  Google Scholar 

  144. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R (2007) Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res 67(21):10123–10128. doi:https://doi.org/10.1158/0008-5472.CAN-07-3127

    Article  CAS  PubMed  Google Scholar 

  145. Madar S, Goldstein I, Rotter V (2013) ’Cancer associated fibroblasts’—more than meets the eye. Trends Mol Med 19(8):447–453. https://doi.org/10.1016/j.molmed.2013.05.004

    Article  CAS  PubMed  Google Scholar 

  146. Platel V, Faure S, Corre I, Clere N (2019) Endothelial-to-Mesenchymal Transition (EndoMT): Roles in Tumorigenesis, Metastatic Extravasation and Therapy Resistance. J Oncol 2019:8361945. doi:https://doi.org/10.1155/2019/8361945

  147. Jung HM, Castranova D, Swift MR, Pham VN, Venero Galanternik M, Isogai S, Butler MG, Mulligan TS, Weinstein BM (2017) Development of the larval lymphatic system in zebrafish. Development 144(11):2070–2081. doi:https://doi.org/10.1242/dev.145755

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Herwig L, Blum Y, Krudewig A, Ellertsdottir E, Lenard A, Belting HG, Affolter M (2011) Distinct cellular mechanisms of blood vessel fusion in the zebrafish embryo. Curr Biol 21(22):1942–1948. doi:https://doi.org/10.1016/j.cub.2011.10.016

    Article  CAS  PubMed  Google Scholar 

  149. Bussmann J, Wolfe SA, Siekmann AF (2011) Arterial-venous network formation during brain vascularization involves hemodynamic regulation of chemokine signaling. Development 138(9):1717–1726. doi:https://doi.org/10.1242/dev.059881

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. van Impel A, Zhao Z, Hermkens DM, Roukens MG, Fischer JC, Peterson-Maduro J, Duckers H, Ober EA, Ingham PW, Schulte-Merker S (2014) Divergence of zebrafish and mouse lymphatic cell fate specification pathways. Development 141(6):1228–1238. doi:https://doi.org/10.1242/dev.105031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Butko E, Distel M, Pouget C, Weijts B, Kobayashi I, Ng K, Mosimann C, Poulain FE, McPherson A, Ni CW, Stachura DL, Del Cid N, Espin-Palazon R, Lawson ND, Dorsky R, Clements WK, Traver D (2015) Gata2b is a restricted early regulator of hemogenic endothelium in the zebrafish embryo. Development 142(6):1050–1061. doi:https://doi.org/10.1242/dev.119180

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Nicenboim J, Malkinson G, Lupo T, Asaf L, Sela Y, Mayseless O, Gibbs-Bar L, Senderovich N, Hashimshony T, Shin M, Jerafi-Vider A, Avraham-Davidi I, Krupalnik V, Hofi R, Almog G, Astin JW, Golani O, Ben-Dor S, Crosier PS, Herzog W, Lawson ND, Hanna JH, Yanai I, Yaniv K (2015) Lymphatic vessels arise from specialized angioblasts within a venous niche. Nature 522(7554):56–61. doi:https://doi.org/10.1038/nature14425

    Article  CAS  PubMed  Google Scholar 

  153. Tian Y, Xu J, Feng S, He S, Zhao S, Zhu L, Jin W, Dai Y, Luo L, Qu JY, Wen Z (2017) The first wave of T lymphopoiesis in zebrafish arises from aorta endothelium independent of hematopoietic stem cells. J Exp Med 214(11):3347–3360. doi:https://doi.org/10.1084/jem.20170488

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Hatta K, Tsujii H, Omura T (2006) Cell tracking using a photoconvertible fluorescent protein. Nat Protoc 1(2):960–967. doi:https://doi.org/10.1038/nprot.2006.96

    Article  CAS  PubMed  Google Scholar 

  155. Mosimann C, Kaufman CK, Li P, Pugach EK, Tamplin OJ, Zon LI (2011) Ubiquitous transgene expression and Cre-based recombination driven by the ubiquitin promoter in zebrafish. Development 138(1):169–177. doi:https://doi.org/10.1242/dev.059345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Pan YA, Freundlich T, Weissman TA, Schoppik D, Wang XC, Zimmerman S, Ciruna B, Sanes JR, Lichtman JW, Schier AF (2013) Zebrabow: multispectral cell labeling for cell tracing and lineage analysis in zebrafish. Development 140(13):2835–2846. doi:https://doi.org/10.1242/dev.094631

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Corti P, Young S, Chen CY, Patrick MJ, Rochon ER, Pekkan K, Roman BL (2011) Interaction between alk1 and blood flow in the development of arteriovenous malformations. Development 138(8):1573–1582. doi:https://doi.org/10.1242/dev.060467

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Sugden WW, Meissner R, Aegerter-Wilmsen T, Tsaryk R, Leonard EV, Bussmann J, Hamm MJ, Herzog W, Jin Y, Jakobsson L, Denz C, Siekmann AF (2017) Endoglin controls blood vessel diameter through endothelial cell shape changes in response to haemodynamic cues. Nat Cell Biol 19(6):653–665. doi:https://doi.org/10.1038/ncb3528

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Mably JD, Chuang LP, Serluca FC, Mohideen MA, Chen JN, Fishman MC (2006) santa and valentine pattern concentric growth of cardiac myocardium in the zebrafish. Development 133(16):3139–3146. doi:https://doi.org/10.1242/dev.02469

    Article  CAS  PubMed  Google Scholar 

  160. Yoruk B, Gillers BS, Chi NC, Scott IC (2012) Ccm3 functions in a manner distinct from Ccm1 and Ccm2 in a zebrafish model of CCM vascular disease. Dev Biol 362(2):121–131. doi:https://doi.org/10.1016/j.ydbio.2011.12.006

    Article  CAS  PubMed  Google Scholar 

  161. Stoletov K, Fang L, Choi SH, Hartvigsen K, Hansen LF, Hall C, Pattison J, Juliano J, Miller ER, Almazan F, Crosier P, Witztum JL, Klemke RL, Miller YI (2009) Vascular lipid accumulation, lipoprotein oxidation, and macrophage lipid uptake in hypercholesterolemic zebrafish. Circ Res 104(8):952–960. doi:https://doi.org/10.1161/CIRCRESAHA.108.189803

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Liu C, Gates KP, Fang L, Amar MJ, Schneider DA, Geng H, Huang W, Kim J, Pattison J, Zhang J, Witztum JL, Remaley AT, Dong PD, Miller YI (2015) Apoc2 loss-of-function zebrafish mutant as a genetic model of hyperlipidemia. Dis Model Mech 8(8):989–998. doi:https://doi.org/10.1242/dmm.019836

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Liu C, Kim YS, Kim J, Pattison J, Kamaid A, Miller YI (2018) Modeling hypercholesterolemia and vascular lipid accumulation in LDL receptor mutant zebrafish. J Lipid Res 59(2):391–399. doi:https://doi.org/10.1194/jlr.D081521

    Article  CAS  PubMed  Google Scholar 

  164. Pringle ES, Wertman J, Melong N, Coombs AJ, Young AL, O’Leary D, Veinotte C, Robinson CA, Ha MN, Dellaire G, Druley TE, McCormick C, Berman JN (2019) The zebrafish xenograft platform—a novel tool for modeling KSHV-associated diseases. Viruses. https://doi.org/10.3390/v12010012

  165. Patton EE, Widlund HR, Kutok JL, Kopani KR, Amatruda JF, Murphey RD, Berghmans S, Mayhall EA, Traver D, Fletcher CD, Aster JC, Granter SR, Look AT, Lee C, Fisher DE, Zon LI (2005) BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma. Curr Biol 15(3):249–254. doi:https://doi.org/10.1016/j.cub.2005.01.031

    Article  CAS  PubMed  Google Scholar 

  166. Park SW, Davison JM, Rhee J, Hruban RH, Maitra A, Leach SD (2008) Oncogenic KRAS induces progenitor cell expansion and malignant transformation in zebrafish exocrine pancreas. Gastroenterology 134(7):2080–2090. doi:https://doi.org/10.1053/j.gastro.2008.02.084

    Article  PubMed  PubMed Central  Google Scholar 

  167. Yang HW, Kutok JL, Lee NH, Piao HY, Fletcher CD, Kanki JP, Look AT (2004) Targeted expression of human MYCN selectively causes pancreatic neuroendocrine tumors in transgenic zebrafish. Cancer Res 64(20):7256–7262. doi:https://doi.org/10.1158/0008-5472.CAN-04-0931

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank members of the Weinstein lab for their help and support. We apologize to authors whose work we could not cite due to space limitations.

Funding

This paper was supported by the intramural program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (ZIA-HD008915, ZIA-HD008808, and ZIA-HD001011, to BMW).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Brant M. Weinstein.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Greenspan, L.J., Weinstein, B.M. To be or not to be: endothelial cell plasticity in development, repair, and disease. Angiogenesis 24, 251–269 (2021). https://doi.org/10.1007/s10456-020-09761-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10456-020-09761-7

Keywords

Navigation