Российский физиологический журнал им. И.М. Сеченова, 2023, T. 109, № 6, стр. 703-722

Механизмы физиологического ангиогенеза

А. Н. Иванов 1, Ю. Р. Чаббаров 1*

1 Саратовский государственный медицинский университет им. В.И. Разумовского
Саратов, Россия

* E-mail: chabbarov@bk.ru

Поступила в редакцию 12.03.2023
После доработки 05.05.2023
Принята к публикации 07.05.2023

Аннотация

Ангиогенез представляет собой образование новых кровеносных сосудов из уже существующих для удовлетворения потребности ткани в перфузии. Во взрослом организме это часто реализуется в процессе роста и регенерации организма. Ангиогенез привлекает внимание исследователей с позиции его терапевтической регуляции: усиление может способствовать существенному прогрессу в терапии ишемических заболеваний, а ингибирование активно изучается для лечения опухолевых заболеваний. Регуляция ангиогенеза невозможна без точных знаний его механизмов. Ангиогенез осуществляется либо путем миграции эндотелиальных клеток существующего сосуда во внеклеточный матрикс для формирования сосуда по направлению ангиогенных стимулов, либо путем разделения сформированного сосуда на два новых в результате образования перегородки, отграничивающей два самостоятельных просвета. В данном обзоре рассматриваются основные этапы каждого из видов физиологического ангиогенеза, их механизмы и регуляция.

Ключевые слова: ангиогенез, эндотелиальные клетки, факторы роста, микроциркуляция, кровеносный сосуд, регуляция ангиогенеза, механизмы ангиогенеза

Список литературы

  1. Oliveira de Oliveira LB, Faccin Bampi V, Ferreira Gomes C, Braga da Silva JL, Encarnação Fiala Rechsteiner SM (2014) Morphological characterization of sprouting and intussusceptive angiogenesis by SEM in oral squamous cell carcinoma: Sprouting and intussusceptive angiogenesis in oral cancer. Scanning 36: 293–300. https://doi.org/10.1002/sca.21104

  2. Deev R, Plaksa I, Bozo I, Isaev A (2017) Results of an International Postmarketing Surveillance Study of pl-VEGF165 Safety and Efficacy in 210 Patients with Peripheral Arterial Disease. Am J Cardiovasc Drugs 17: 235–242. https://doi.org/10.1007/s40256-016-0210-3

  3. Lheureux S, Oaknin A, Garg S, Bruce JP, Madariaga A, Dhani NC, Bowering V, White J, Accardi S, Tan Q, Braunstein M, Karakasis K, Cirlan I, Pedersen S, Li T, Fariñas-Madrid L, Lee YC, Liu ZA, Pugh TJ, Oza AM (2020) EVOLVE: A Multicenter Open-Label Single-Arm Clinical and Translational Phase II Trial of Cediranib Plus Olaparib for Ovarian Cancer after PARP Inhibition Progression. Clin Cancer Res 26: 4206–4215. https://doi.org/10.1158/1078-0432.CCR-19-4121

  4. Kosolapov VP, Yarmonova MV (2021) The analysis of high cardiovascular morbidity and mortality in the adult population as a medical and social problem and the search for ways to solve it. Jour 20: 58–64. https://doi.org/10.52420/2071-5943-2021-20-1-58-64

  5. Hasin Y, Seldin M, Lusis A (2017) Multi-omics approaches to disease. Genome Biol 18: 83. https://doi.org/10.1186/s13059-017-1215-1

  6. Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS (2022) Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 14(4): e1550. https://doi.org/10.1002/wsbm.1550

  7. Wang J-C, Li G-Y, Li P-P, Sun X, Li W-M, Li Y, Lu S-Y, Liu P-J (2017) Suppression of hypoxia-induced excessive angiogenesis by metformin via elevating tumor blood perfusion. Oncotarget 8: 73892–73904. https://doi.org/10.18632/oncotarget.18029

  8. Apte RS, Chen DS, Ferrara N (2019) VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 176: 1248–1264. https://doi.org/10.1016/j.cell.2019.01.021

  9. Zhang Y, Zhong Y, Liu W, Zheng F, Zhao Y, Zou L, Liu X (2022) PFKFB3-mediated glycometabolism reprogramming modulates endothelial differentiation and angiogenic capacity of placenta-derived mesenchymal stem cells. Stem Cell Res Ther 13: 391. https://doi.org/10.1186/s13287-022-03089-3

  10. Melincovici CS, Boşca AB, Şuşman S, Mărginean M, Mihu C, Istrate M, Moldovan IM, Roman AL, Mihu CM (2018) Vascular endothelial growth factor (VEGF) – key factor in normal and pathological angiogenesis. Rom J Morphol Embryol 59: 455–467.

  11. Noren DP, Chou WH, Lee SH, Qutub AA, Warmflash A, Wagner DS, Popel AS, Levchenko A (2016) Endothelial cells decode VEGF-mediated Ca2+ signaling patterns to produce distinct functional responses. Sci Signal 9: ra20. https://doi.org/10.1126/scisignal.aad3188

  12. Balberova OV, Bykov EV, Shnayder NA, Petrova MM, Gavrilyuk OA, Kaskaeva DS, Soloveva IA, Petrov KV, Mozheyko EY, Medvedev GV, Nasyrova RF (2021) The “Angiogenic Switch” and Functional Resources in Cyclic Sports Athletes. Int J Mol Sci 22: 6496. https://doi.org/10.3390/ijms22126496

  13. Mamer SB, Wittenkeller A, Imoukhuede PI (2020) VEGF-A splice variants bind VEGFRs with differential affinities. Sci Rep 10: 14413. https://doi.org/10.1038/s41598-020-71484-y

  14. Teran M, Nugent MA (2015) Synergistic Binding of Vascular Endothelial Growth Factor-A and Its Receptors to Heparin Selectively Modulates Complex Affinity. J Biol Chem 290: 16451–16462. https://doi.org/10.1074/jbc.M114.627372

  15. Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, Crncec I, Kramer N, Unger C, Hengstschläger M, Eferl R, Moriggl R, Sommergruber W, Gerner C, Dolznig H (2020) Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis 23: 159–177. https://doi.org/10.1007/s10456-019-09688-8

  16. Zhao C, Popel AS (2015) Computational Model of MicroRNA Control of HIF-VEGF Pathway: Insights into the Pathophysiology of Ischemic Vascular Disease and Cancer. PLoS Comput Biol 11: e1004612. https://doi.org/10.1371/journal.pcbi.1004612

  17. Naik MU, Chatterjee S, Naik UP (2020) Fibroblast Growth Factor-2-Induced Endothelial Cell Migration Is Regulated by Junctional Adhesion Molecule-A Through Its Tyrosine Phosphorylation and Interaction With Afadin. The FASEB J 34: 1–1. https://doi.org/10.1096/fasebj.2020.34.s1.06430

  18. Weinstein N, Mendoza L, Gitler I, Klapp J (2017) A Network Model to Explore the Effect of the Micro-environment on Endothelial Cell Behavior during Angiogenesis. Front Physiol 8: 960. https://doi.org/10.3389/fphys.2017.00960

  19. Vasuri F, Fittipaldi S, Abualhin M, Degiovanni A, Gargiulo M, Stella A, Pasquinelli G (2014) Biochemical and immunomorphological evaluation of hepatocyte growth factor and c-Met pathway in patients with critical limb ischemia. Eur J Vasc Endovasc Surg 48: 430–437. https://doi.org/10.1016/j.ejvs.2014.05.002

  20. Koudstaal S, Bastings MMC, Feyen DAM, Waring CD, van Slochteren FJ, Dankers PYW, Torella D, Sluijter JPG, Nadal-Ginard B, Doevendans PA, Ellison GM, Chamuleau SAJ (2014) Sustained delivery of insulin-like growth factor-1/hepatocyte growth factor stimulates endogenous cardiac repair in the chronic infarcted pig heart. J Cardiovasc Transl Res 7: 232–241. https://doi.org/10.1007/s12265-013-9518-4

  21. Salabarria A-C, Braun G, Heykants M, Koch M, Reuten R, Mahabir E, Cursiefen C, Bock F (2019) Local VEGF-A blockade modulates the microenvironment of the corneal graft bed. Am J Transplant 19: 2446–2456. https://doi.org/10.1111/ajt.15331

  22. Yazdani S, Kasajima A, Tamaki K, Nakamura Y, Fujishima F, Ohtsuka H, Motoi F, Unno M, Watanabe M, Sato Y, Sasano H (2014) Angiogenesis and vascular maturation in neuroendocrine tumors. Human Pathol 45: 866–874. https://doi.org/10.1016/j.humpath.2013.09.024

  23. Michalczyk ER, Chen L, Fine D, Zhao Y, Mascarinas E, Grippo PJ, DiPietro LA (2018) Pigment Epithelium-Derived Factor (PEDF) as a Regulator of Wound Angiogenesis. Sci Rep 8: 11142. https://doi.org/10.1038/s41598-018-29465-9

  24. Nagai T, Sato M, Kobayashi M, Yokoyama M, Tani Y, Mochida J (2014) Bevacizumab, an anti-vascular endothelial growth factor antibody, inhibits osteoarthritis. Arthritis Res Ther 16: 427. https://doi.org/10.1186/s13075-014-0427-y

  25. Yadav L, Puri N, Rastogi V, Satpute P, Sharma V (2015) Tumour Angiogenesis and Angiogenic Inhibitors: A Review. J Clin Diagn Res 9: XE01–XE05. https://doi.org/10.7860/JCDR/2015/12016.6135

  26. El-Kenawi AE, El-Remessy AB (2013) Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol 170: 712–729. https://doi.org/10.1111/bph.12344

  27. Zhang X, Crawford R, Xiao Y (2014) Anti-angiogenic factors are essential regulators in cartilage homeostasis and osteoarthritis. Osteoarthritis and Cartilage 22: S132. https://doi.org/10.1016/j.joca.2014.02.243

  28. Granstam E, Aurell S, Sjövall K, Paul A (2021) Switching anti-VEGF agent for wet AMD: evaluation of impact on visual acuity, treatment frequency and retinal morphology in a real-world clinical setting. Graefes Arch Clin Exp Ophthalmol 259: 2085–2093. https://doi.org/10.1007/s00417-020-05059-y

  29. Mabeta P, Hull R, Dlamini Z (2022) LncRNAs and the Angiogenic Switch in Cancer: Clinical Significance and Therapeutic Opportunities. Genes (Basel) 13: 152. https://doi.org/10.3390/genes13010152

  30. Luo H, Shen Y, Liao W, Li Q, Wu N, Zhong J, Xiao C, Gan J, Yang Y, Dong E, Zhang G, Liu B, Yue X, Xu L, Liu Y, Zhao C, Zhong Q, Yang H (2022) The inhibition of protein translation promotes tumor angiogenic switch. Mol Biomed 3: 18. https://doi.org/10.1186/s43556-022-00081-4

  31. Mentzer SJ, Konerding MA (2014) Intussusceptive angiogenesis: expansion and remodeling of microvascular networks. Angiogenesis 17: 499–509. https://doi.org/10.1007/s10456-014-9428-3

  32. Kangsamaksin T, Murtomaki A, Kofler NM, Cuervo H, Chaudhri RA, Tattersall IW, Rosenstiel PE, Shawber CJ, Kitajewski J (2015) NOTCH decoys that selectively block DLL/NOTCH or JAG/NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor growth. Cancer Discov 5: 182–197. https://doi.org/10.1158/2159-8290.CD-14-0650

  33. Zalpoor H, Aziziyan F, Liaghat M, Bakhtiyari M, Akbari A, Nabi-Afjadi M, Forghaniesfidvajani R, Rezaei N (2022) The roles of metabolic profiles and intracellular signaling pathways of tumor microenvironment cells in angiogenesis of solid tumors. Cell Commun Signal 20: 186. https://doi.org/10.1186/s12964-022-00951-y

  34. Travisano SI, Oliveira VL, Prados B, Grego-Bessa J, Piñeiro-Sabarís R, Bou V, Gómez MJ, Sánchez-Cabo F, MacGrogan D, de la Pompa JL (2019) Coronary arterial development is regulated by a Dll4-Jag1-EphrinB2 signaling cascade. eLife 8: e49977. https://doi.org/10.7554/eLife.49977

  35. Kasprzak A (2020) Angiogenesis-Related Functions of Wnt Signaling in Colorectal Carcinogenesis. Cancers (Basel) 12: E3601. https://doi.org/10.3390/cancers12123601

  36. Santos-Oliveira P, Correia A, Rodrigues T, Ribeiro-Rodrigues TM, Matafome P, Rodríguez-Manzaneque JC, Seiça R, Girão H, Travasso RDM (2015) The Force at the Tip–Modelling Tension and Proliferation in Sprouting Angiogenesis. PLoS Comput Biol 11: e1004436. https://doi.org/10.1371/journal.pcbi.1004436

  37. Neve A, Cantatore FP, Maruotti N, Corrado A, Ribatti D (2014) Extracellular matrix modulates angiogenesis in physiological and pathological conditions. Biomed Res Int 2014: 756078. https://doi.org/10.1155/2014/756078

  38. Ruehle MA, Eastburn EA, LaBelle SA, Krishnan L, Weiss JA, Boerckel JD, Wood LB, Guldberg RE, Willett NJ (2020) Extracellular matrix compression temporally regulates microvascular angiogenesis. Sci Adv 6: eabb6351. https://doi.org/10.1126/sciadv.abb6351

  39. Conway RE, Joiner K, Patterson A, Bourgeois D, Rampp R, Hannah BC, McReynolds S, Elder JM, Gilfilen H, Shapiro LH (2013) Prostate specific membrane antigen produces pro-angiogenic laminin peptides downstream of matrix metalloprotease-2. Angiogenesis 16: 847–860. https://doi.org/10.1007/s10456-013-9360-y

  40. Lai K-C, Lu C-C, Tang Y-J, Chiang J-H, Kuo D-H, Chen F-A, Chen I-L, Yang J-S (2014) Allyl isothiocyanate inhibits cell metastasis through suppression of the MAPK pathways in epidermal growth factor‑stimulated HT29 human colorectal adenocarcinoma cells. Oncol Rep 31: 189–196. https://doi.org/10.3892/or.2013.2865

  41. Chambers SEJ, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, Medina RJ (2021) Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl Med 10 Suppl 2: S54–S61. https://doi.org/10.1002/sctm.21-0022

  42. Charpentier MS, Conlon FL (2014) Cellular and molecular mechanisms underlying blood vessel lumen formation. Bioessays 36: 251–259. https://doi.org/10.1002/bies.201300133

  43. Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL (2015) A Distinct Mechanism of Vascular Lumen Formation in Xenopus Requires EGFL7. PLoS One 10: e0116086. https://doi.org/10.1371/journal.pone.0116086

  44. Davis GE, Stratman AN, Sacharidou A, Koh W (2011) Molecular basis for endothelial lumen formation and tubulogenesis during vasculogenesis and angiogenic sprouting. Int Rev Cell Mol Biol 288: 101–165. https://doi.org/10.1016/B978-0-12-386041-5.00003-0

  45. Boas SEM, Merks RMH (2014) Synergy of cell-cell repulsion and vacuolation in a computational model of lumen formation. J R Soc Interface 11: 20131049. https://doi.org/10.1098/rsif.2013.1049

  46. Lenard A, Daetwyler S, Betz C, Ellertsdottir E, Belting H-G, Huisken J, Affolter M (2015) Endothelial Cell Self-fusion during Vascular Pruning. PLoS Biol 13: e1002126. https://doi.org/10.1371/journal.pbio.1002126

  47. Lenard A, Ellertsdottir E, Herwig L, Krudewig A, Sauteur L, Belting H-G, Affolter M (2013) In vivo analysis reveals a highly stereotypic morphogenetic pathway of vascular anastomosis. Dev Cell 25: 492–506. https://doi.org/10.1016/j.devcel.2013.05.010

  48. Zarkada G, Heinolainen K, Makinen T, Kubota Y, Alitalo K (2015) VEGFR3 does not sustain retinal angiogenesis without VEGFR2. Proc Natl Acad Sci U S A 112: 761–766. https://doi.org/10.1073/pnas.1423278112

  49. Zhang Z, Warner KA, Mantesso A, Nör JE (2022) PDGF-BB signaling via PDGFR-β regulates the maturation of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Front Cell Dev Biol 10: 977725. https://doi.org/10.3389/fcell.2022.977725

  50. Gianni-Barrera R, Bartolomeo M, Vollmar B, Djonov V, Banfi A (2014) Split for the cure: VEGF, PDGF-BB and intussusception in therapeutic angiogenesis. Biochem Soc Trans 42: 1637–1642. https://doi.org/10.1042/BST20140234

  51. Khan M, Aziz AA, Shafi NA, Abbas T, Khanani AM (2020) Targeting Angiopoietin in Retinal Vascular Diseases: A Literature Review and Summary of Clinical Trials Involving Faricimab. Cells 9: E1869. https://doi.org/10.3390/cells9081869

  52. Chu H, Sun Y, Gao Y, Guan X, Yan H, Cui X, Zhang X, Li X, Li H, Cheng M (2019) Function of Kruppel‑like factor 2 in the shear stress‑induced cell differentiation of endothelial progenitor cells to endothelial cells. Mol Med Report 19(3): 1739–1746.https://doi.org/10.3892/mmr.2019.9819

  53. Dekker RJ, van Thienen JV, Rohlena J, de Jager SC, Elderkamp YW, Seppen J, de Vries CJM, Biessen EAL, van Berkel TJC, Pannekoek H, Horrevoets AJG (2005) Endothelial KLF2 Links Local Arterial Shear Stress Levels to the Expression of Vascular Tone-Regulating Genes. Am J Pathol 167: 609–618. https://doi.org/10.1016/S0002-9440(10)63002-7

  54. Korn C, Augustin HG (2015) Mechanisms of Vessel Pruning and Regression. Dev Cell 34: 5–17. https://doi.org/10.1016/j.devcel.2015.06.004

  55. Shahik SMD, Salauddin A, Hossain MDS, Noyon SH, Moin AT, Mizan S, Raza MDT (2021) Screening of novel alkaloid inhibitors for vascular endothelial growth factor in cancer cells: an integrated computational approach. Genomics Inform 19: e6. https://doi.org/10.5808/gi.20068

  56. Díaz-Flores L, Gutiérrez R, García MP, Gayoso S, Carrasco JL, Díaz-Flores Lucio, González-Gómez M, Madrid JF (2020) Intussusceptive Angiogenesis and Peg–Socket Junctions between Endothelial Cells and Smooth Muscle Cells in Early Arterial Intimal Thickening. IJMS 21: 8049. https://doi.org/10.3390/ijms21218049

  57. Ackermann M, Tsuda A, Secomb TW, Mentzer SJ, Konerding MA (2013) Intussusceptive remodeling of vascular branch angles in chemically-induced murine colitis. Microvasc Res 87: 75–82. https://doi.org/10.1016/j.mvr.2013.02.002

  58. Nitzsche B, Rong WW, Goede A, Hoffmann B, Scarpa F, Kuebler WM, Secomb TW, Pries AR (2022) Coalescent angiogenesis-evidence for a novel concept of vascular network maturation. Angiogenesis 25: 35–45. https://doi.org/10.1007/s10456-021-09824-3

  59. Gifre-Renom L, Jones EAV (2021) Vessel Enlargement in Development and Pathophysiology. Front Physiol 12: 639645. https://doi.org/10.3389/fphys.2021.639645

Дополнительные материалы отсутствуют.