Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition
Abstract
:1. Introduction
2. Genetic Mutations as the Cause of bAVMs
2.1. HHT Gene Mutations in bAVMs
2.2. Somatic Mutations in bAVMs
2.3. Phakomatoses and bAVMs
3. Endothelial Cells as the Major Player in bAVM Pathology
3.1. Molecular Signaling in ECs Relevant to bAVMs
3.1.1. TGFβ/BMP Signaling
3.1.2. Notch Signaling
3.1.3. RAF/MEK/ERK Signaling
3.1.4. VEGF Signaling
3.1.5. PI3K/AKT/mTOR Signaling
3.1.6. Targeted Therapies in the Treatment of bAVMs
3.2. EC Dysfunctions in bAVMs
3.2.1. Vascular Development and Arteriovenous Specification
3.2.2. EC Polarization and Migration
3.2.3. EC Junction Integrity and Vessel Stability
3.2.4. EC Dysfunctions in bAVMs
3.3. Endothelial-to-Mesenchymal Transition (EndMT) and bAVMs
3.3.1. General Definition of EndMT
3.3.2. Transcription Modulators of EndMT
3.3.3. TGFβ-Induced EndMT
3.3.4. Other Signaling Pathways Which Induce EndMT
3.3.5. EndMT as a Pathologic Event in bAVMs
3.3.6. Potential EndMT Inhibitors
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Al-Shahi, R.; Fang, J.S.; Lewis, S.C.; Warlow, C.P. Prevalence of adults with brain arteriovenous malformations: A community based study in Scotland using capture-recapture analysis. J. Neurol. Neurosurg. Psychiatry 2002, 73, 547–551. [Google Scholar] [CrossRef]
- Berman, M.F.; Sciacca, R.R.; Pile-Spellman, J.; Stapf, C.; Connolly, E.S., Jr.; Mohr, J.P.; Young, W.L. The epidemiology of brain arteriovenous malformations. Neurosurgery 2000, 47, 389–396; discussion 397. [Google Scholar] [CrossRef] [PubMed]
- Pan, P.; Weinsheimer, S.; Cooke, D.; Winkler, E.; Abla, A.; Kim, H.; Su, H. Review of treatment and therapeutic targets in brain arteriovenous malformation. J. Cereb. Blood Flow. Metab. 2021, 41, 3141–3156. [Google Scholar] [CrossRef] [PubMed]
- Shaligram, S.S.; Winkler, E.; Cooke, D.; Su, H. Risk factors for hemorrhage of brain arteriovenous malformation. CNS Neurosci. Ther. 2019, 25, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
- Stapf, C.; Mast, H.; Sciacca, R.R.; Choi, J.H.; Khaw, A.V.; Connolly, E.S.; Pile-Spellman, J.; Mohr, J.P. Predictors of hemorrhage in patients with untreated brain arteriovenous malformation. Neurology 2006, 66, 1350–1355. [Google Scholar] [CrossRef]
- Venugopal, V.; Sumi, S. Molecular Biomarkers and Drug Targets in Brain Arteriovenous and Cavernous Malformations: Where Are We? Stroke 2022, 53, 279–289. [Google Scholar] [CrossRef] [PubMed]
- Stefani, M.A.; Porter, P.J.; terBrugge, K.G.; Montanera, W.; Willinsky, R.A.; Wallace, M.C. Large and deep brain arteriovenous malformations are associated with risk of future hemorrhage. Stroke 2002, 33, 1220–1224. [Google Scholar] [CrossRef]
- Copelan, A.; Drocton, G.; Caton, M.T.; Smith, E.R.; Cooke, D.L.; Nelson, J.; Abla, A.A.; Fox, C.; Amans, M.R.; Dowd, C.F.; et al. Brain Arteriovenous Malformation Recurrence After Apparent Microsurgical Cure: Increased Risk in Children Who Present With Arteriovenous Malformation Rupture. Stroke 2020, 51, 2990–2996. [Google Scholar] [CrossRef]
- McDonald, J.; Wooderchak-Donahue, W.; VanSant Webb, C.; Whitehead, K.; Stevenson, D.A.; Bayrak-Toydemir, P. Hereditary hemorrhagic telangiectasia: Genetics and molecular diagnostics in a new era. Front. Genet. 2015, 6, 1. [Google Scholar] [CrossRef]
- Guttmacher, A.E.; Marchuk, D.A.; White, R.I., Jr. Hereditary hemorrhagic telangiectasia. N. Engl. J. Med. 1995, 333, 918–924. [Google Scholar] [CrossRef]
- Lam, S.; Guthrie, K.S.; Latif, M.A.; Weiss, C.R. Genetic counseling and testing for hereditary hemorrhagic telangiectasia. Clin. Genet. 2022, 101, 275–284. [Google Scholar] [CrossRef] [PubMed]
- Nishida, T.; Faughnan, M.E.; Krings, T.; Chakinala, M.; Gossage, J.R.; Young, W.L.; Kim, H.; Pourmohamad, T.; Henderson, K.J.; Schrum, S.D.; et al. Brain arteriovenous malformations associated with hereditary hemorrhagic telangiectasia: Gene-phenotype correlations. Am. J. Med. Genet. A 2012, 158A, 2829–2834. [Google Scholar] [CrossRef] [PubMed]
- Brinjikji, W.; Iyer, V.N.; Wood, C.P.; Lanzino, G. Prevalence and characteristics of brain arteriovenous malformations in hereditary hemorrhagic telangiectasia: A systematic review and meta-analysis. J. Neurosurg. 2017, 127, 302–310. [Google Scholar] [CrossRef]
- Johnson, D.W.; Berg, J.N.; Baldwin, M.A.; Gallione, C.J.; Marondel, I.; Yoon, S.J.; Stenzel, T.T.; Speer, M.; Pericak-Vance, M.A.; Diamond, A.; et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat. Genet. 1996, 13, 189–195. [Google Scholar] [CrossRef] [PubMed]
- McAllister, K.A.; Grogg, K.M.; Johnson, D.W.; Gallione, C.J.; Baldwin, M.A.; Jackson, C.E.; Helmbold, E.A.; Markel, D.S.; McKinnon, W.C.; Murrell, J.; et al. Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat. Genet. 1994, 8, 345–351. [Google Scholar] [CrossRef] [PubMed]
- Gallione, C.J.; Richards, J.A.; Letteboer, T.G.; Rushlow, D.; Prigoda, N.L.; Leedom, T.P.; Ganguly, A.; Castells, A.; Ploos van Amstel, J.K.; Westermann, C.J.; et al. SMAD4 mutations found in unselected HHT patients. J. Med. Genet. 2006, 43, 793–797. [Google Scholar] [CrossRef] [PubMed]
- Lesca, G.; Burnichon, N.; Raux, G.; Tosi, M.; Pinson, S.; Marion, M.J.; Babin, E.; Gilbert-Dussardier, B.; Riviere, S.; Goizet, C.; et al. Distribution of ENG and ACVRL1 (ALK1) mutations in French HHT patients. Hum. Mutat. 2006, 27, 598. [Google Scholar] [CrossRef] [PubMed]
- Prigoda, N.L.; Savas, S.; Abdalla, S.A.; Piovesan, B.; Rushlow, D.; Vandezande, K.; Zhang, E.; Ozcelik, H.; Gallie, B.L.; Letarte, M. Hereditary haemorrhagic telangiectasia: Mutation detection, test sensitivity and novel mutations. J. Med. Genet. 2006, 43, 722–728. [Google Scholar] [CrossRef] [PubMed]
- Gallione, C.J.; Repetto, G.M.; Legius, E.; Rustgi, A.K.; Schelley, S.L.; Tejpar, S.; Mitchell, G.; Drouin, E.; Westermann, C.J.; Marchuk, D.A. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet 2004, 363, 852–859. [Google Scholar] [CrossRef]
- Wooderchak-Donahue, W.L.; McDonald, J.; O’Fallon, B.; Upton, P.D.; Li, W.; Roman, B.L.; Young, S.; Plant, P.; Fulop, G.T.; Langa, C.; et al. BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am. J. Hum. Genet. 2013, 93, 530–537. [Google Scholar] [CrossRef]
- Aagaard, K.S.; Brusgaard, K.; Miceikaite, I.; Larsen, M.J.; Kjeldsen, A.D.; Lester, E.B.; Ousager, L.B.; Torring, P.M. Chromosomal translocation disrupting the SMAD4 gene resulting in the combined phenotype of Juvenile polyposis syndrome and Hereditary Hemorrhagic Telangiectasia. Mol. Genet. Genom. Med. 2020, 8, e1498. [Google Scholar] [CrossRef]
- Vorselaars, V.M.M.; Diederik, A.; Prabhudesai, V.; Velthuis, S.; Vos, J.A.; Snijder, R.J.; Westermann, C.J.J.; Mulder, B.J.; Ploos van Amstel, J.K.; Mager, J.J.; et al. SMAD4 gene mutation increases the risk of aortic dilation in patients with hereditary haemorrhagic telangiectasia. Int. J. Cardiol. 2017, 245, 114–118. [Google Scholar] [CrossRef]
- Gallione, C.; Aylsworth, A.S.; Beis, J.; Berk, T.; Bernhardt, B.; Clark, R.D.; Clericuzio, C.; Danesino, C.; Drautz, J.; Fahl, J.; et al. Overlapping spectra of SMAD4 mutations in juvenile polyposis (JP) and JP-HHT syndrome. Am. J. Med. Genet. A 2010, 152A, 333–339. [Google Scholar] [CrossRef]
- Balachandar, S.; Graves, T.J.; Shimonty, A.; Kerr, K.; Kilner, J.; Xiao, S.; Slade, R.; Sroya, M.; Alikian, M.; Curetean, E.; et al. Identification and validation of a novel pathogenic variant in GDF2 (BMP9) responsible for hereditary hemorrhagic telangiectasia and pulmonary arteriovenous malformations. Am. J. Med. Genet. A 2022, 188, 959–964. [Google Scholar] [CrossRef]
- Farhan, A.; Yuan, F.; Partan, E.; Weiss, C.R. Clinical manifestations of patients with GDF2 mutations associated with hereditary hemorrhagic telangiectasia type 5. Am. J. Med. Genet. A 2022, 188, 199–209. [Google Scholar] [CrossRef] [PubMed]
- Hodgson, J.; Ruiz-Llorente, L.; McDonald, J.; Quarrell, O.; Ugonna, K.; Bentham, J.; Mason, R.; Martin, J.; Moore, D.; Bergstrom, K.; et al. Homozygous GDF2 nonsense mutations result in a loss of circulating BMP9 and BMP10 and are associated with either PAH or an “HHT-like” syndrome in children. Mol. Genet. Genom. Med. 2021, 9, e1685. [Google Scholar] [CrossRef]
- Best, D.H.; Vaughn, C.; McDonald, J.; Damjanovich, K.; Runo, J.R.; Chibuk, J.M.; Bayrak-Toydemir, P. Mosaic ACVRL1 and ENG mutations in hereditary haemorrhagic telangiectasia patients. J. Med. Genet. 2011, 48, 358–360. [Google Scholar] [CrossRef]
- Clarke, J.M.; Alikian, M.; Xiao, S.; Kasperaviciute, D.; Thomas, E.; Turbin, I.; Olupona, K.; Cifra, E.; Curetean, E.; Ferguson, T.; et al. Low grade mosaicism in hereditary haemorrhagic telangiectasia identified by bidirectional whole genome sequencing reads through the 100,000 Genomes Project clinical diagnostic pipeline. J. Med. Genet. 2020, 57, 859–862. [Google Scholar] [CrossRef] [PubMed]
- Gedge, F.; McDonald, J.; Phansalkar, A.; Chou, L.S.; Calderon, F.; Mao, R.; Lyon, E.; Bayrak-Toydemir, P. Clinical and analytical sensitivities in hereditary hemorrhagic telangiectasia testing and a report of de novo mutations. J. Mol. Diagn. 2007, 9, 258–265. [Google Scholar] [CrossRef] [PubMed]
- McDonald, J.; Wooderchak-Donahue, W.L.; Henderson, K.; Paul, E.; Morris, A.; Bayrak-Toydemir, P. Tissue-specific mosaicism in hereditary hemorrhagic telangiectasia: Implications for genetic testing in families. Am. J. Med. Genet. A 2018, 176, 1618–1621. [Google Scholar] [CrossRef]
- Torring, P.M.; Kjeldsen, A.D.; Ousager, L.B.; Brusgaard, K. ENG mutational mosaicism in a family with hereditary hemorrhagic telangiectasia. Mol. Genet. Genom. Med. 2018, 6, 121–125. [Google Scholar] [CrossRef] [PubMed]
- Snellings, D.A.; Gallione, C.J.; Clark, D.S.; Vozoris, N.T.; Faughnan, M.E.; Marchuk, D.A. Somatic Mutations in Vascular Malformations of Hereditary Hemorrhagic Telangiectasia Result in Bi-allelic Loss of ENG or ACVRL1. Am. J. Hum. Genet. 2019, 105, 894–906. [Google Scholar] [CrossRef]
- Chen, W.; Young, W.L.; Su, H. Induction of brain arteriovenous malformation in the adult mouse. Methods Mol. Biol. 2014, 1135, 309–316. [Google Scholar] [CrossRef] [PubMed]
- Choi, E.J.; Chen, W.; Jun, K.; Arthur, H.M.; Young, W.L.; Su, H. Novel brain arteriovenous malformation mouse models for type 1 hereditary hemorrhagic telangiectasia. PLoS ONE 2014, 9, e88511. [Google Scholar] [CrossRef]
- Nikolaev, S.I.; Vetiska, S.; Bonilla, X.; Boudreau, E.; Jauhiainen, S.; Rezai Jahromi, B.; Khyzha, N.; DiStefano, P.V.; Suutarinen, S.; Kiehl, T.R.; et al. Somatic Activating KRAS Mutations in Arteriovenous Malformations of the Brain. N. Engl. J. Med. 2018, 378, 250–261. [Google Scholar] [CrossRef]
- Goss, J.A.; Huang, A.Y.; Smith, E.; Konczyk, D.J.; Smits, P.J.; Sudduth, C.L.; Stapleton, C.; Patel, A.; Alexandrescu, S.; Warman, M.L.; et al. Somatic mutations in intracranial arteriovenous malformations. PLoS ONE 2019, 14, e0226852. [Google Scholar] [CrossRef] [PubMed]
- Hong, T.; Yan, Y.; Li, J.; Radovanovic, I.; Ma, X.; Shao, Y.W.; Yu, J.; Ma, Y.; Zhang, P.; Ling, F.; et al. High prevalence of KRAS/BRAF somatic mutations in brain and spinal cord arteriovenous malformations. Brain 2019, 142, 23–34. [Google Scholar] [CrossRef]
- Li, H.; Nam, Y.; Huo, R.; Fu, W.; Jiang, B.; Zhou, Q.; Song, D.; Yang, Y.; Jiao, Y.; Weng, J.; et al. De Novo Germline and Somatic Variants Convergently Promote Endothelial-to-Mesenchymal Transition in Simplex Brain Arteriovenous Malformation. Circ. Res. 2021, 129, 825–839. [Google Scholar] [CrossRef]
- Oka, M.; Kushamae, M.; Aoki, T.; Yamaguchi, T.; Kitazato, K.; Abekura, Y.; Kawamata, T.; Mizutani, T.; Miyamoto, S.; Takagi, Y. KRAS G12D or G12V Mutation in Human Brain Arteriovenous Malformations. World Neurosurg. 2019, 126, e1365–e1373. [Google Scholar] [CrossRef]
- Priemer, D.S.; Vortmeyer, A.O.; Zhang, S.; Chang, H.Y.; Curless, K.L.; Cheng, L. Activating KRAS mutations in arteriovenous malformations of the brain: Frequency and clinicopathologic correlation. Hum. Pathol. 2019, 89, 33–39. [Google Scholar] [CrossRef]
- Al-Olabi, L.; Polubothu, S.; Dowsett, K.; Andrews, K.A.; Stadnik, P.; Joseph, A.P.; Knox, R.; Pittman, A.; Clark, G.; Baird, W.; et al. Mosaic RAS/MAPK variants cause sporadic vascular malformations which respond to targeted therapy. J. Clin. Investig. 2018, 128, 5185. [Google Scholar] [CrossRef]
- Fish, J.E.; Flores Suarez, C.P.; Boudreau, E.; Herman, A.M.; Gutierrez, M.C.; Gustafson, D.; DiStefano, P.V.; Cui, M.; Chen, Z.; De Ruiz, K.B.; et al. Somatic Gain of KRAS Function in the Endothelium Is Sufficient to Cause Vascular Malformations That Require MEK but Not PI3K Signaling. Circ. Res. 2020, 127, 727–743. [Google Scholar] [CrossRef]
- Park, E.S.; Kim, S.; Huang, S.; Yoo, J.Y.; Korbelin, J.; Lee, T.J.; Kaur, B.; Dash, P.K.; Chen, P.R.; Kim, E. Selective Endothelial Hyperactivation of Oncogenic KRAS Induces Brain Arteriovenous Malformations in Mice. Ann. Neurol. 2021, 89, 926–941. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.F.; Decker-Rockefeller, B.; Bajaj, A.; Pumiglia, K. Activation of Ras in the Vascular Endothelium Induces Brain Vascular Malformations and Hemorrhagic Stroke. Cell Rep. 2018, 24, 2869–2882. [Google Scholar] [CrossRef] [PubMed]
- Swarup, M.S.; Gupta, S.; Singh, S.; Prakash, A.; Mehndiratta, A.; Garg, A. Phakomatoses: A pictorial review. Indian. J. Radiol. Imaging 2020, 30, 195–205. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.K.; Keenaghan, M. Sturge-Weber Syndrome. In StatPearls; StatPearls: St. Petersburg, FL, USA, 2024. [Google Scholar]
- Sanchez-Espino, L.F.; Ivars, M.; Antonanzas, J.; Baselga, E. Sturge-Weber Syndrome: A Review of Pathophysiology, Genetics, Clinical Features, and Current Management Approache. Appl. Clin. Genet. 2023, 16, 63–81. [Google Scholar] [CrossRef]
- Shirley, M.D.; Tang, H.; Gallione, C.J.; Baugher, J.D.; Frelin, L.P.; Cohen, B.; North, P.E.; Marchuk, D.A.; Comi, A.M.; Pevsner, J. Sturge-Weber syndrome and port-wine stains caused by somatic mutation in GNAQ. N. Engl. J. Med. 2013, 368, 1971–1979. [Google Scholar] [CrossRef]
- Couto, J.A.; Huang, L.; Vivero, M.P.; Kamitaki, N.; Maclellan, R.A.; Mulliken, J.B.; Bischoff, J.; Warman, M.L.; Greene, A.K. Endothelial Cells from Capillary Malformations Are Enriched for Somatic GNAQ Mutations. Plast. Reconstr. Surg. 2016, 137, 77e–82e. [Google Scholar] [CrossRef]
- Martins, L.; Giovani, P.A.; Reboucas, P.D.; Brasil, D.M.; Haiter Neto, F.; Coletta, R.D.; Machado, R.A.; Puppin-Rontani, R.M.; Nociti, F.H., Jr.; Kantovitz, K.R. Computational analysis for GNAQ mutations: New insights on the molecular etiology of Sturge-Weber syndrome. J. Mol. Graph. Model. 2017, 76, 429–440. [Google Scholar] [CrossRef]
- Yu, F.X.; Luo, J.; Mo, J.S.; Liu, G.; Kim, Y.C.; Meng, Z.; Zhao, L.; Peyman, G.; Ouyang, H.; Jiang, W.; et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 2014, 25, 822–830. [Google Scholar] [CrossRef]
- Polubothu, S.; Al-Olabi, L.; Carmen Del Boente, M.; Chacko, A.; Eleftheriou, G.; Glover, M.; Jimenez-Gallo, D.; Jones, E.A.; Lomas, D.; Folster-Holst, R.; et al. GNA11 Mutation as a Cause of Sturge-Weber Syndrome: Expansion of the Phenotypic Spectrum of G(alpha/11) Mosaicism and the Associated Clinical Diagnoses. J. Investig. Dermatol. 2020, 140, 1110–1113. [Google Scholar] [CrossRef]
- Nathan, N.; Thaller, S.R. Sturge-Weber syndrome and associated congenital vascular disorders: A review. J. Craniofac Surg. 2006, 17, 724–728. [Google Scholar] [CrossRef]
- Harnarayan, P.; Harnanan, D. The Klippel-Trenaunay Syndrome in 2022: Unravelling Its Genetic and Molecular Profile and Its Link to the Limb Overgrowth Syndromes. Vasc Health Risk Manag 2022, 18, 201–209. [Google Scholar] [CrossRef]
- Flores Daboub, J.A.; Grimmer, J.F.; Frigerio, A.; Wooderchak-Donahue, W.; Arnold, R.; Szymanski, J.; Longo, N.; Bayrak-Toydemir, P. Parkes Weber syndrome associated with two somatic pathogenic variants in RASA1. Cold Spring Harb. Mol. Case Stud. 2020, 6, a005256. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.; Teng, J.M.; North, P.E.; Lapinski, P.E.; King, P.D. RASA1-dependent cellular export of collagen IV controls blood and lymphatic vascular development. J. Clin. Investig. 2019, 129, 3545–3561. [Google Scholar] [CrossRef] [PubMed]
- Eerola, I.; Boon, L.M.; Mulliken, J.B.; Burrows, P.E.; Dompmartin, A.; Watanabe, S.; Vanwijck, R.; Vikkula, M. Capillary malformation-arteriovenous malformation, a new clinical and genetic disorder caused by RASA1 mutations. Am. J. Hum. Genet. 2003, 73, 1240–1249. [Google Scholar] [CrossRef] [PubMed]
- So, J.M.; Mishra, C.; Holman, R.E. Wyburn-Mason Syndrome. In StatPearls; StatPearls: St. Petersburg, FL, USA, 2024. [Google Scholar]
- Bhattacharya, J.J.; Luo, C.B.; Suh, D.C.; Alvarez, H.; Rodesch, G.; Lasjaunias, P. Wyburn-Mason or Bonnet-Dechaume-Blanc as Cerebrofacial Arteriovenous Metameric Syndromes (CAMS). A New Concept and a New Classification. Interv. Neuroradiol. 2001, 7, 5–17. [Google Scholar] [CrossRef] [PubMed]
- Trimm, E.; Red-Horse, K. Vascular endothelial cell development and diversity. Nat. Rev. Cardiol. 2023, 20, 197–210. [Google Scholar] [CrossRef]
- Fang, J.; Hirschi, K. Molecular regulation of arteriovenous endothelial cell specification. F1000Res 2019, 8. [Google Scholar] [CrossRef]
- Liu, F.; Yao, Z.; Lu, P.; Jiao, Q.B.; Liu, Q.; Wu, H.X.; You, Y.; Minamisawa, S. Pathophysiologic Role of Molecules Determining Arteriovenous Differentiation in Adult Life. J. Vasc. Res. 2020, 57, 245–253. [Google Scholar] [CrossRef]
- Shabani, Z.; Schuerger, J.; Su, H. Cellular loci involved in the development of brain arteriovenous malformations. Front. Hum. Neurosci. 2022, 16, 968369. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Sun, Z.; Han, Z.; Jun, K.; Camus, M.; Wankhede, M.; Mao, L.; Arnold, T.; Young, W.L.; Su, H. De novo cerebrovascular malformation in the adult mouse after endothelial Alk1 deletion and angiogenic stimulation. Stroke 2014, 45, 900–902. [Google Scholar] [CrossRef] [PubMed]
- Shaligram, S.S.; Zhang, R.; Zhu, W.; Ma, L.; Luo, M.; Li, Q.; Weiss, M.; Arnold, T.; Santander, N.; Liang, R.; et al. Bone Marrow-Derived Alk1 Mutant Endothelial Cells and Clonally Expanded Somatic Alk1 Mutant Endothelial Cells Contribute to the Development of Brain Arteriovenous Malformations in Mice. Transl. Stroke Res. 2022, 13, 494–504. [Google Scholar] [CrossRef] [PubMed]
- Corti, P.; Young, S.; Chen, C.Y.; Patrick, M.J.; Rochon, E.R.; Pekkan, K.; Roman, B.L. Interaction between alk1 and blood flow in the development of arteriovenous malformations. Development 2011, 138, 1573–1582. [Google Scholar] [CrossRef] [PubMed]
- David, L.; Mallet, C.; Keramidas, M.; Lamande, N.; Gasc, J.M.; Dupuis-Girod, S.; Plauchu, H.; Feige, J.J.; Bailly, S. Bone morphogenetic protein-9 is a circulating vascular quiescence factor. Circ. Res. 2008, 102, 914–922. [Google Scholar] [CrossRef] [PubMed]
- Laux, D.W.; Young, S.; Donovan, J.P.; Mansfield, C.J.; Upton, P.D.; Roman, B.L. Circulating Bmp10 acts through endothelial Alk1 to mediate flow-dependent arterial quiescence. Development 2013, 140, 3403–3412. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Dimaio, T.A.; Liu, W.; Wang, S.; Sorenson, C.M.; Sheibani, N. Endoglin regulates the activation and quiescence of endothelium by participating in canonical and non-canonical TGF-beta signaling pathways. J. Cell Sci. 2013, 126, 1392–1405. [Google Scholar] [CrossRef] [PubMed]
- Rochon, E.R.; Menon, P.G.; Roman, B.L. Alk1 controls arterial endothelial cell migration in lumenized vessels. Development 2016, 143, 2593–2602. [Google Scholar] [CrossRef] [PubMed]
- Guo, X.; Wang, X.F. Signaling cross-talk between TGF-beta/BMP and other pathways. Cell Res. 2009, 19, 71–88. [Google Scholar] [CrossRef]
- Li, F.; Lan, Y.; Wang, Y.; Wang, J.; Yang, G.; Meng, F.; Han, H.; Meng, A.; Wang, Y.; Yang, X. Endothelial Smad4 maintains cerebrovascular integrity by activating N-cadherin through cooperation with Notch. Dev. Cell 2011, 20, 291–302. [Google Scholar] [CrossRef]
- Campioni, D.; Zauli, G.; Gambetti, S.; Campo, G.; Cuneo, A.; Ferrari, R.; Secchiero, P. In vitro characterization of circulating endothelial progenitor cells isolated from patients with acute coronary syndrome. PLoS ONE 2013, 8, e56377. [Google Scholar] [CrossRef] [PubMed]
- Gougos, A.; Letarte, M. Primary structure of endoglin, an RGD-containing glycoprotein of human endothelial cells. J. Biol. Chem. 1990, 265, 8361–8364. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Novoa, J.M.; Bernabeu, C. The physiological role of endoglin in the cardiovascular system. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H959–H974. [Google Scholar] [CrossRef] [PubMed]
- Alt, A.; Miguel-Romero, L.; Donderis, J.; Aristorena, M.; Blanco, F.J.; Round, A.; Rubio, V.; Bernabeu, C.; Marina, A. Structural and functional insights into endoglin ligand recognition and binding. PLoS ONE 2012, 7, e29948. [Google Scholar] [CrossRef] [PubMed]
- Gregory, A.L.; Xu, G.; Sotov, V.; Letarte, M. Review: The enigmatic role of endoglin in the placenta. Placenta 2014, 35, S93–S99. [Google Scholar] [CrossRef]
- Li, W.; Salmon, R.M.; Jiang, H.; Morrell, N.W. Regulation of the ALK1 ligands, BMP9 and BMP10. Biochem. Soc. Trans. 2016, 44, 1135–1141. [Google Scholar] [CrossRef] [PubMed]
- Rossi, E.; Bernabeu, C.; Smadja, D.M. Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function Beyond TGF-beta. Front. Med. (Lausanne) 2019, 6, 10. [Google Scholar] [CrossRef]
- Saito, T.; Bokhove, M.; Croci, R.; Zamora-Caballero, S.; Han, L.; Letarte, M.; de Sanctis, D.; Jovine, L. Structural Basis of the Human Endoglin-BMP9 Interaction: Insights into BMP Signaling and HHT1. Cell Rep. 2017, 19, 1917–1928. [Google Scholar] [CrossRef] [PubMed]
- Cunha, S.I.; Magnusson, P.U.; Dejana, E.; Lampugnani, M.G. Deregulated TGF-beta/BMP Signaling in Vascular Malformations. Circ. Res. 2017, 121, 981–999. [Google Scholar] [CrossRef]
- ten Dijke, P.; Arthur, H.M. Extracellular control of TGFbeta signalling in vascular development and disease. Nat. Rev. Mol. Cell Biol. 2007, 8, 857–869. [Google Scholar] [CrossRef]
- ten Dijke, P.; Goumans, M.J.; Pardali, E. Endoglin in angiogenesis and vascular diseases. Angiogenesis 2008, 11, 79–89. [Google Scholar] [CrossRef] [PubMed]
- Bourdeau, A.; Dumont, D.J.; Letarte, M. A murine model of hereditary hemorrhagic telangiectasia. J. Clin. Investig. 1999, 104, 1343–1351. [Google Scholar] [CrossRef]
- Li, D.Y.; Sorensen, L.K.; Brooke, B.S.; Urness, L.D.; Davis, E.C.; Taylor, D.G.; Boak, B.B.; Wendel, D.P. Defective angiogenesis in mice lacking endoglin. Science 1999, 284, 1534–1537. [Google Scholar] [CrossRef]
- Mahmoud, M.; Allinson, K.R.; Zhai, Z.; Oakenfull, R.; Ghandi, P.; Adams, R.H.; Fruttiger, M.; Arthur, H.M. Pathogenesis of arteriovenous malformations in the absence of endoglin. Circ. Res. 2010, 106, 1425–1433. [Google Scholar] [CrossRef] [PubMed]
- Lebrin, F.; Srun, S.; Raymond, K.; Martin, S.; van den Brink, S.; Freitas, C.; Breant, C.; Mathivet, T.; Larrivee, B.; Thomas, J.L.; et al. Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia. Nat. Med. 2010, 16, 420–428. [Google Scholar] [CrossRef] [PubMed]
- Hawinkels, L.J.; Kuiper, P.; Wiercinska, E.; Verspaget, H.W.; Liu, Z.; Pardali, E.; Sier, C.F.; ten Dijke, P. Matrix metalloproteinase-14 (MT1-MMP)-mediated endoglin shedding inhibits tumor angiogenesis. Cancer Res. 2010, 70, 4141–4150. [Google Scholar] [CrossRef] [PubMed]
- Valbuena-Diez, A.C.; Blanco, F.J.; Oujo, B.; Langa, C.; Gonzalez-Nunez, M.; Llano, E.; Pendas, A.M.; Diaz, M.; Castrillo, A.; Lopez-Novoa, J.M.; et al. Oxysterol-induced soluble endoglin release and its involvement in hypertension. Circulation 2012, 126, 2612–2624. [Google Scholar] [CrossRef]
- Rathouska, J.; Jezkova, K.; Nemeckova, I.; Nachtigal, P. Soluble endoglin, hypercholesterolemia and endothelial dysfunction. Atherosclerosis 2015, 243, 383–388. [Google Scholar] [CrossRef] [PubMed]
- Venkatesha, S.; Toporsian, M.; Lam, C.; Hanai, J.; Mammoto, T.; Kim, Y.M.; Bdolah, Y.; Lim, K.H.; Yuan, H.T.; Libermann, T.A.; et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat. Med. 2006, 12, 642–649. [Google Scholar] [CrossRef]
- Bernabeu, C.; Lopez-Novoa, J.M.; Quintanilla, M. The emerging role of TGF-beta superfamily coreceptors in cancer. Biochim. Biophys. Acta 2009, 1792, 954–973. [Google Scholar] [CrossRef]
- Blazquez-Medela, A.M.; Garcia-Ortiz, L.; Gomez-Marcos, M.A.; Recio-Rodriguez, J.I.; Sanchez-Rodriguez, A.; Lopez-Novoa, J.M.; Martinez-Salgado, C. Increased plasma soluble endoglin levels as an indicator of cardiovascular alterations in hypertensive and diabetic patients. BMC Med. 2010, 8, 86. [Google Scholar] [CrossRef] [PubMed]
- Ermini, L.; Ausman, J.; Melland-Smith, M.; Yeganeh, B.; Rolfo, A.; Litvack, M.L.; Todros, T.; Letarte, M.; Post, M.; Caniggia, I. A Single Sphingomyelin Species Promotes Exosomal Release of Endoglin into the Maternal Circulation in Preeclampsia. Sci. Rep. 2017, 7, 12172. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Hao, Q.; Kim, H.; Su, H.; Letarte, M.; Karumanchi, S.A.; Lawton, M.T.; Barbaro, N.M.; Yang, G.Y.; Young, W.L. Soluble endoglin modulates aberrant cerebral vascular remodeling. Ann. Neurol. 2009, 66, 19–27. [Google Scholar] [CrossRef] [PubMed]
- Park, E.S.; Kim, S.; Yao, D.C.; Savarraj, J.P.J.; Choi, H.A.; Chen, P.R.; Kim, E. Soluble Endoglin Stimulates Inflammatory and Angiogenic Responses in Microglia That Are Associated with Endothelial Dysfunction. Int. J. Mol. Sci. 2022, 23, 1225. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Nunez, M.; Munoz-Felix, J.M.; Lopez-Novoa, J.M. The ALK-1/Smad1 pathway in cardiovascular physiopathology. A new target for therapy? Biochim. Biophys. Acta 2013, 1832, 1492–1510. [Google Scholar] [CrossRef] [PubMed]
- Garrido-Martin, E.M.; Nguyen, H.L.; Cunningham, T.A.; Choe, S.W.; Jiang, Z.; Arthur, H.M.; Lee, Y.J.; Oh, S.P. Common and distinctive pathogenetic features of arteriovenous malformations in hereditary hemorrhagic telangiectasia 1 and hereditary hemorrhagic telangiectasia 2 animal models—brief report. Arter. Thromb. Vasc. Biol. 2014, 34, 2232–2236. [Google Scholar] [CrossRef] [PubMed]
- Park, S.O.; Wankhede, M.; Lee, Y.J.; Choi, E.J.; Fliess, N.; Choe, S.W.; Oh, S.H.; Walter, G.; Raizada, M.K.; Sorg, B.S.; et al. Real-time imaging of de novo arteriovenous malformation in a mouse model of hereditary hemorrhagic telangiectasia. J. Clin. Investig. 2009, 119, 3487–3496. [Google Scholar] [CrossRef] [PubMed]
- Seki, T.; Yun, J.; Oh, S.P. Arterial endothelium-specific activin receptor-like kinase 1 expression suggests its role in arterialization and vascular remodeling. Circ. Res. 2003, 93, 682–689. [Google Scholar] [CrossRef] [PubMed]
- Oh, S.P.; Seki, T.; Goss, K.A.; Imamura, T.; Yi, Y.; Donahoe, P.K.; Li, L.; Miyazono, K.; ten Dijke, P.; Kim, S.; et al. Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis. Proc. Natl. Acad. Sci. USA 2000, 97, 2626–2631. [Google Scholar] [CrossRef]
- Urness, L.D.; Sorensen, L.K.; Li, D.Y. Arteriovenous malformations in mice lacking activin receptor-like kinase-1. Nat. Genet. 2000, 26, 328–331. [Google Scholar] [CrossRef]
- Srinivasan, S.; Hanes, M.A.; Dickens, T.; Porteous, M.E.; Oh, S.P.; Hale, L.P.; Marchuk, D.A. A mouse model for hereditary hemorrhagic telangiectasia (HHT) type 2. Hum. Mol. Genet. 2003, 12, 473–482. [Google Scholar] [CrossRef]
- Roman, B.L.; Pham, V.N.; Lawson, N.D.; Kulik, M.; Childs, S.; Lekven, A.C.; Garrity, D.M.; Moon, R.T.; Fishman, M.C.; Lechleider, R.J.; et al. Disruption of acvrl1 increases endothelial cell number in zebrafish cranial vessels. Development 2002, 129, 3009–3019. [Google Scholar] [CrossRef] [PubMed]
- Hwan Kim, Y.; Vu, P.N.; Choe, S.W.; Jeon, C.J.; Arthur, H.M.; Vary, C.P.H.; Lee, Y.J.; Oh, S.P. Overexpression of Activin Receptor-Like Kinase 1 in Endothelial Cells Suppresses Development of Arteriovenous Malformations in Mouse Models of Hereditary Hemorrhagic Telangiectasia. Circ. Res. 2020, 127, 1122–1137. [Google Scholar] [CrossRef] [PubMed]
- Brown, M.A.; Zhao, Q.; Baker, K.A.; Naik, C.; Chen, C.; Pukac, L.; Singh, M.; Tsareva, T.; Parice, Y.; Mahoney, A.; et al. Crystal structure of BMP-9 and functional interactions with pro-region and receptors. J. Biol. Chem. 2005, 280, 25111–25118. [Google Scholar] [CrossRef] [PubMed]
- Nolan-Stevaux, O.; Zhong, W.; Culp, S.; Shaffer, K.; Hoover, J.; Wickramasinghe, D.; Ruefli-Brasse, A. Endoglin requirement for BMP9 signaling in endothelial cells reveals new mechanism of action for selective anti-endoglin antibodies. PLoS ONE 2012, 7, e50920. [Google Scholar] [CrossRef] [PubMed]
- Park, J.E.; Shao, D.; Upton, P.D.; Desouza, P.; Adcock, I.M.; Davies, R.J.; Morrell, N.W.; Griffiths, M.J.; Wort, S.J. BMP-9 induced endothelial cell tubule formation and inhibition of migration involves Smad1 driven endothelin-1 production. PLoS ONE 2012, 7, e30075. [Google Scholar] [CrossRef] [PubMed]
- van Meeteren, L.A.; Thorikay, M.; Bergqvist, S.; Pardali, E.; Stampino, C.G.; Hu-Lowe, D.; Goumans, M.J.; ten Dijke, P. Anti-human activin receptor-like kinase 1 (ALK1) antibody attenuates bone morphogenetic protein 9 (BMP9)-induced ALK1 signaling and interferes with endothelial cell sprouting. J. Biol. Chem. 2012, 287, 18551–18561. [Google Scholar] [CrossRef] [PubMed]
- Tillet, E.; Bailly, S. Emerging roles of BMP9 and BMP10 in hereditary hemorrhagic telangiectasia. Front. Genet. 2014, 5, 456. [Google Scholar] [CrossRef]
- Salmon, R.M.; Guo, J.; Wood, J.H.; Tong, Z.; Beech, J.S.; Lawera, A.; Yu, M.; Grainger, D.J.; Reckless, J.; Morrell, N.W.; et al. Molecular basis of ALK1-mediated signalling by BMP9/BMP10 and their prodomain-bound forms. Nat. Commun. 2020, 11, 1621. [Google Scholar] [CrossRef]
- Atri, D.; Larrivee, B.; Eichmann, A.; Simons, M. Endothelial signaling and the molecular basis of arteriovenous malformation. Cell Mol. Life Sci. 2013, 71, 867–883. [Google Scholar] [CrossRef]
- David, L.; Feige, J.J.; Bailly, S. Emerging role of bone morphogenetic proteins in angiogenesis. Cytokine Growth Factor. Rev. 2009, 20, 203–212. [Google Scholar] [CrossRef] [PubMed]
- 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 2007, 109, 1953–1961. [Google Scholar] [CrossRef] [PubMed]
- Garcia de Vinuesa, A.; Abdelilah-Seyfried, S.; Knaus, P.; Zwijsen, A.; Bailly, S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor. Rev. 2016, 27, 65–79. [Google Scholar] [CrossRef] [PubMed]
- Scharpfenecker, M.; van Dinther, M.; Liu, Z.; van Bezooijen, R.L.; Zhao, Q.; Pukac, L.; Lowik, C.W.; ten Dijke, P. BMP-9 signals via ALK1 and inhibits bFGF-induced endothelial cell proliferation and VEGF-stimulated angiogenesis. J. Cell Sci. 2007, 120, 964–972. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, Y.; Ohga, N.; Morishita, Y.; Hida, K.; Miyazono, K.; Watabe, T. BMP-9 induces proliferation of multiple types of endothelial cells in vitro and in vivo. J. Cell Sci. 2010, 123, 1684–1692. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Brady Ridgway, J.; Sai, T.; Lai, J.; Warming, S.; Chen, H.; Roose-Girma, M.; Zhang, G.; Shou, W.; Yan, M. Context-dependent signaling defines roles of BMP9 and BMP10 in embryonic and postnatal development. Proc. Natl. Acad. Sci. USA 2013, 110, 11887–11892. [Google Scholar] [CrossRef] [PubMed]
- Levet, S.; Ciais, D.; Merdzhanova, G.; Mallet, C.; Zimmers, T.A.; Lee, S.J.; Navarro, F.P.; Texier, I.; Feige, J.J.; Bailly, S.; et al. Bone morphogenetic protein 9 (BMP9) controls lymphatic vessel maturation and valve formation. Blood 2013, 122, 598–607. [Google Scholar] [CrossRef] [PubMed]
- Ricard, N.; Ciais, D.; Levet, S.; Subileau, M.; Mallet, C.; Zimmers, T.A.; Lee, S.J.; Bidart, M.; Feige, J.J.; Bailly, S. BMP9 and BMP10 are critical for postnatal retinal vascular remodeling. Blood 2012, 119, 6162–6171. [Google Scholar] [CrossRef]
- Yoshimatsu, Y.; Lee, Y.G.; Akatsu, Y.; Taguchi, L.; Suzuki, H.I.; Cunha, S.I.; Maruyama, K.; Suzuki, Y.; Yamazaki, T.; Katsura, A.; et al. Bone morphogenetic protein-9 inhibits lymphatic vessel formation via activin receptor-like kinase 1 during development and cancer progression. Proc. Natl. Acad. Sci. USA 2013, 110, 18940–18945. [Google Scholar] [CrossRef]
- Chen, H.; Shi, S.; Acosta, L.; Li, W.; Lu, J.; Bao, S.; Chen, Z.; Yang, Z.; Schneider, M.D.; Chien, K.R.; et al. BMP10 is essential for maintaining cardiac growth during murine cardiogenesis. Development 2004, 131, 2219–2231. [Google Scholar] [CrossRef]
- Bray, S.J. Notch signalling: A simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 2006, 7, 678–689. [Google Scholar] [CrossRef] [PubMed]
- Kopan, R. Notch signaling. Cold Spring Harb. Perspect. Biol. 2012, 4, a011213. [Google Scholar] [CrossRef] [PubMed]
- D‘Souza, B.; Meloty-Kapella, L.; Weinmaster, G. Canonical and non-canonical Notch ligands. Curr. Top. Dev. Biol. 2010, 92, 73–129. [Google Scholar] [CrossRef]
- Benedito, R.; Trindade, A.; Hirashima, M.; Henrique, D.; da Costa, L.L.; Rossant, J.; Gill, P.S.; Duarte, A. Loss of Notch signalling induced by Dll4 causes arterial calibre reduction by increasing endothelial cell response to angiogenic stimuli. BMC Dev. Biol. 2008, 8, 117. [Google Scholar] [CrossRef] [PubMed]
- Duarte, A.; Hirashima, M.; Benedito, R.; Trindade, A.; Diniz, P.; Bekman, E.; Costa, L.; Henrique, D.; Rossant, J. Dosage-sensitive requirement for mouse Dll4 in artery development. Genes. Dev. 2004, 18, 2474–2478. [Google Scholar] [CrossRef]
- Gridley, T. Notch signaling in the vasculature. Curr. Top. Dev. Biol. 2010, 92, 277–309. [Google Scholar] [CrossRef]
- Trindade, A.; Kumar, S.R.; Scehnet, J.S.; Lopes-da-Costa, L.; Becker, J.; Jiang, W.; Liu, R.; Gill, P.S.; Duarte, A. Overexpression of delta-like 4 induces arterialization and attenuates vessel formation in developing mouse embryos. Blood 2008, 112, 1720–1729. [Google Scholar] [CrossRef]
- Fang, J.S.; Coon, B.G.; Gillis, N.; Chen, Z.; Qiu, J.; Chittenden, T.W.; Burt, J.M.; Schwartz, M.A.; Hirschi, K.K. Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat. Commun. 2017, 8, 2149. [Google Scholar] [CrossRef]
- Marcelo, K.L.; Sills, T.M.; Coskun, S.; Vasavada, H.; Sanglikar, S.; Goldie, L.C.; Hirschi, K.K. Hemogenic endothelial cell specification requires c-Kit, Notch signaling, and p27-mediated cell-cycle control. Dev. Cell 2013, 27, 504–515. [Google Scholar] [CrossRef]
- Murphy, P.A.; Kim, T.N.; Huang, L.; Nielsen, C.M.; Lawton, M.T.; Adams, R.H.; Schaffer, C.B.; Wang, R.A. Constitutively active Notch4 receptor elicits brain arteriovenous malformations through enlargement of capillary-like vessels. Proc. Natl. Acad. Sci. USA 2014, 111, 18007–18012. [Google Scholar] [CrossRef]
- Krebs, L.T.; Xue, Y.; Norton, C.R.; Shutter, J.R.; Maguire, M.; Sundberg, J.P.; Gallahan, D.; Closson, V.; Kitajewski, J.; Callahan, R.; et al. Notch signaling is essential for vascular morphogenesis in mice. Genes. Dev. 2000, 14, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
- Swiatek, P.J.; Lindsell, C.E.; del Amo, F.F.; Weinmaster, G.; Gridley, T. Notch1 is essential for postimplantation development in mice. Genes. Dev. 1994, 8, 707–719. [Google Scholar] [CrossRef]
- Lawson, N.D.; Scheer, N.; Pham, V.N.; Kim, C.H.; Chitnis, A.B.; Campos-Ortega, J.A.; Weinstein, B.M. Notch signaling is required for arterial-venous differentiation during embryonic vascular development. Development 2001, 128, 3675–3683. [Google Scholar] [CrossRef] [PubMed]
- Carlson, T.R.; Yan, Y.; Wu, X.; Lam, M.T.; Tang, G.L.; Beverly, L.J.; Messina, L.M.; Capobianco, A.J.; Werb, Z.; Wang, R. Endothelial expression of constitutively active Notch4 elicits reversible arteriovenous malformations in adult mice. Proc. Natl. Acad. Sci. USA 2005, 102, 9884–9889. [Google Scholar] [CrossRef]
- Krebs, L.T.; Starling, C.; Chervonsky, A.V.; Gridley, T. Notch1 activation in mice causes arteriovenous malformations phenocopied by ephrinB2 and EphB4 mutants. Genesis 2010, 48, 146–150. [Google Scholar] [CrossRef]
- Murphy, P.A.; Kim, T.N.; Lu, G.; Bollen, A.W.; Schaffer, C.B.; Wang, R.A. Notch4 normalization reduces blood vessel size in arteriovenous malformations. Sci. Transl. Med. 2012, 4, 117ra118. [Google Scholar] [CrossRef]
- Murphy, P.A.; Lu, G.; Shiah, S.; Bollen, A.W.; Wang, R.A. Endothelial Notch signaling is upregulated in human brain arteriovenous malformations and a mouse model of the disease. Lab. Investig. 2009, 89, 971–982. [Google Scholar] [CrossRef]
- Murphy, P.A.; Lam, M.T.; Wu, X.; Kim, T.N.; Vartanian, S.M.; Bollen, A.W.; Carlson, T.R.; Wang, R.A. Endothelial Notch4 signaling induces hallmarks of brain arteriovenous malformations in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 10901–10906. [Google Scholar] [CrossRef] [PubMed]
- Esser, J.S.; Steiner, R.E.; Deckler, M.; Schmitt, H.; Engert, B.; Link, S.; Charlet, A.; Patterson, C.; Bode, C.; Zhou, Q.; et al. Extracellular bone morphogenetic protein modulator BMPER and twisted gastrulation homolog 1 preserve arterial-venous specification in zebrafish blood vessel development and regulate Notch signaling in endothelial cells. FEBS J. 2018, 285, 1419–1436. [Google Scholar] [CrossRef]
- Mouillesseaux, K.P.; Wiley, D.S.; Saunders, L.M.; Wylie, L.A.; Kushner, E.J.; Chong, D.C.; Citrin, K.M.; Barber, A.T.; Park, Y.; Kim, J.D.; et al. Notch regulates BMP responsiveness and lateral branching in vessel networks via SMAD6. Nat. Commun. 2016, 7, 13247. [Google Scholar] [CrossRef]
- Larrivee, B.; Prahst, C.; Gordon, E.; del Toro, R.; Mathivet, T.; Duarte, A.; Simons, M.; Eichmann, A. ALK1 signaling inhibits angiogenesis by cooperating with the Notch pathway. Dev. Cell 2012, 22, 489–500. [Google Scholar] [CrossRef]
- Somekawa, S.; Imagawa, K.; Hayashi, H.; Sakabe, M.; Ioka, T.; Sato, G.E.; Inada, K.; Iwamoto, T.; Mori, T.; Uemura, S.; et al. Tmem100, an ALK1 receptor signaling-dependent gene essential for arterial endothelium differentiation and vascular morphogenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 12064–12069. [Google Scholar] [CrossRef]
- Colicelli, J. Human RAS superfamily proteins and related GTPases. Sci. STKE 2004, 2004, RE13. [Google Scholar] [CrossRef]
- Bos, J.L.; Rehmann, H.; Wittinghofer, A. GEFs and GAPs: Critical elements in the control of small G proteins. Cell 2007, 129, 865–877. [Google Scholar] [CrossRef]
- Pao, W.; Girard, N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011, 12, 175–180. [Google Scholar] [CrossRef]
- De Luca, A.; Maiello, M.R.; D‘Alessio, A.; Pergameno, M.; Normanno, N. The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: Role in cancer pathogenesis and implications for therapeutic approaches. Expert. Opin. Ther. Targets 2012, 16 (Suppl. S2), S17–S27. [Google Scholar] [CrossRef]
- Prior, I.A.; Hood, F.E.; Hartley, J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020, 80, 2969–2974. [Google Scholar] [CrossRef]
- Coffman, S.A.; Peterson, K.; Contillo, N.; Fargen, K.M.; Wolfe, S.Q. A comprehensive review on the development of sporadic cerebral arteriovenous malformations: From Padget to next-generation sequencing. J. Neurosurg. 2024, 1, 1–10. [Google Scholar] [CrossRef]
- Nguyen, H.L.; Boon, L.M.; Vikkula, M. Trametinib as a promising therapeutic option in alleviating vascular defects in an endothelial KRAS-induced mouse model. Hum. Mol. Genet. 2023, 32, 276–289. [Google Scholar] [CrossRef]
- Edwards, E.A.; Phelps, A.S.; Cooke, D.; Frieden, I.J.; Zapala, M.A.; Fullerton, H.J.; Shimano, K.A. Monitoring Arteriovenous Malformation Response to Genotype-Targeted Therapy. Pediatrics 2020, 146. [Google Scholar] [CrossRef]
- Lekwuttikarn, R.; Lim, Y.H.; Admani, S.; Choate, K.A.; Teng, J.M.C. Genotype-Guided Medical Treatment of an Arteriovenous Malformation in a Child. JAMA Dermatol. 2019, 155, 256–257. [Google Scholar] [CrossRef]
- Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef]
- Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol. 2007, 8, 464–478. [Google Scholar] [CrossRef]
- McColl, B.K.; Stacker, S.A.; Achen, M.G. Molecular regulation of the VEGF family–inducers of angiogenesis and lymphangiogenesis. Apmis 2004, 112, 463–480. [Google Scholar] [CrossRef]
- Hashimoto, T.; Wu, Y.; Lawton, M.T.; Yang, G.Y.; Barbaro, N.M.; Young, W.L. Coexpression of angiogenic factors in brain arteriovenous malformations. Neurosurgery 2005, 56, 1058–1065; discussion 1058–1065. [Google Scholar]
- Hatva, E.; Jaaskelainen, J.; Hirvonen, H.; Alitalo, K.; Haltia, M. Tie endothelial cell-specific receptor tyrosine kinase is upregulated in the vasculature of arteriovenous malformations. J. Neuropathol. Exp. Neurol. 1996, 55, 1124–1133. [Google Scholar] [CrossRef]
- Kilic, T.; Pamir, M.N.; Kullu, S.; Eren, F.; Ozek, M.M.; Black, P.M. Expression of structural proteins and angiogenic factors in cerebrovascular anomalies. Neurosurgery 2000, 46, 1179–1191; discussion 1191–1172. [Google Scholar] [CrossRef]
- Koizumi, T.; Shiraishi, T.; Hagihara, N.; Tabuchi, K.; Hayashi, T.; Kawano, T. Expression of vascular endothelial growth factors and their receptors in and around intracranial arteriovenous malformations. Neurosurgery 2002, 50, 117–124; discussion 124–116. [Google Scholar] [CrossRef]
- Sandalcioglu, I.E.; Wende, D.; Eggert, A.; Muller, D.; Roggenbuck, U.; Gasser, T.; Wiedemayer, H.; Stolke, D. Vascular endothelial growth factor plasma levels are significantly elevated in patients with cerebral arteriovenous malformations. Cerebrovasc. Dis. 2006, 21, 154–158. [Google Scholar] [CrossRef]
- Han, C.; Nguyen, C.L.; Scherschinski, L.; Schriber, T.D.; Arthur, H.M.; Lawton, M.T.; Oh, S.P. VEGFR2 Expression Correlates with Postnatal Development of Brain Arteriovenous Malformations in a Mouse Model of Type I Hereditary Hemorrhagic Telangiectasia. Biomedicines 2023, 11, 3153. [Google Scholar] [CrossRef]
- Chen, W.; Guo, Y.; Walker, E.J.; Shen, F.; Jun, K.; Oh, S.P.; Degos, V.; Lawton, M.T.; Tihan, T.; Davalos, D.; et al. Reduced mural cell coverage and impaired vessel integrity after angiogenic stimulation in the Alk1-deficient brain. Arter. Thromb. Vasc. Biol. 2013, 33, 305–310. [Google Scholar] [CrossRef]
- Choi, E.J.; Walker, E.J.; Shen, F.; Oh, S.P.; Arthur, H.M.; Young, W.L.; Su, H. Minimal homozygous endothelial deletion of Eng with VEGF stimulation is sufficient to cause cerebrovascular dysplasia in the adult mouse. Cerebrovasc. Dis. 2012, 33, 540–547. [Google Scholar] [CrossRef]
- Walker, E.J.; Su, H.; Shen, F.; Choi, E.J.; Oh, S.P.; Chen, G.; Lawton, M.T.; Kim, H.; Chen, Y.; Chen, W.; et al. Arteriovenous malformation in the adult mouse brain resembling the human disease. Ann. Neurol. 2011, 69, 954–962. [Google Scholar] [CrossRef]
- Han, C.; Choe, S.W.; Kim, Y.H.; Acharya, A.P.; Keselowsky, B.G.; Sorg, B.S.; Lee, Y.J.; Oh, S.P. VEGF neutralization can prevent and normalize arteriovenous malformations in an animal model for hereditary hemorrhagic telangiectasia 2. Angiogenesis 2014, 17, 823–830. [Google Scholar] [CrossRef]
- Walker, E.J.; Su, H.; Shen, F.; Degos, V.; Amend, G.; Jun, K.; Young, W.L. Bevacizumab attenuates VEGF-induced angiogenesis and vascular malformations in the adult mouse brain. Stroke 2012, 43, 1925–1930. [Google Scholar] [CrossRef]
- Fang, L.; Li, Y.; Wang, S.; Li, Y.; Chang, H.M.; Yi, Y.; Yan, Y.; Thakur, A.; Leung, P.C.K.; Cheng, J.C.; et al. TGF-beta1 induces VEGF expression in human granulosa-lutein cells: A potential mechanism for the pathogenesis of ovarian hyperstimulation syndrome. Exp. Mol. Med. 2020, 52, 450–460. [Google Scholar] [CrossRef]
- Minet, E.; Arnould, T.; Michel, G.; Roland, I.; Mottet, D.; Raes, M.; Remacle, J.; Michiels, C. ERK activation upon hypoxia: Involvement in HIF-1 activation. FEBS Lett. 2000, 468, 53–58. [Google Scholar] [CrossRef]
- Sodhi, A.; Montaner, S.; Miyazaki, H.; Gutkind, J.S. MAPK and Akt act cooperatively but independently on hypoxia inducible factor-1alpha in rasV12 upregulation of VEGF. Biochem. Biophys. Res. Commun. 2001, 287, 292–300. [Google Scholar] [CrossRef]
- Sun, H.; Miao, C.; Liu, W.; Qiao, X.; Yang, W.; Li, L.; Li, C. TGF-beta1/TbetaRII/Smad3 signaling pathway promotes VEGF expression in oral squamous cell carcinoma tumor-associated macrophages. Biochem. Biophys. Res. Commun. 2018, 497, 583–590. [Google Scholar] [CrossRef]
- Zhao, L.; Yang, Y.F.; Gao, Y.B.; Wang, S.M.; Wang, L.F.; Zuo, H.Y.; Dong, J.; Xu, X.P.; Su, Z.T.; Zhou, H.M.; et al. Upregulation of HIF-1alpha via activation of ERK and PI3K pathway mediated protective response to microwave-induced mitochondrial injury in neuron-like cells. Mol. Neurobiol. 2014, 50, 1024–1034. [Google Scholar] [CrossRef]
- Doanes, A.M.; Hegland, D.D.; Sethi, R.; Kovesdi, I.; Bruder, J.T.; Finkel, T. VEGF stimulates MAPK through a pathway that is unique for receptor tyrosine kinases. Biochem. Biophys. Res. Commun. 1999, 255, 545–548. [Google Scholar] [CrossRef]
- Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR Signaling in Cancer. Front. Oncol. 2014, 4, 64. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Thakkar, H.; Tyan, F.; Gim, S.; Robinson, H.; Lee, C.; Pandey, S.K.; Nwokorie, C.; Onwudiwe, N.; Srivastava, R.K. Constitutively active Akt is an important regulator of TRAIL sensitivity in prostate cancer. Oncogene 2001, 20, 6073–6083. [Google Scholar] [CrossRef]
- Hussain, A.R.; Ahmed, S.O.; Ahmed, M.; Khan, O.S.; Al Abdulmohsen, S.; Platanias, L.C.; Al-Kuraya, K.S.; Uddin, S. Cross-talk between NFkB and the PI3-kinase/AKT pathway can be targeted in primary effusion lymphoma (PEL) cell lines for efficient apoptosis. PLoS ONE 2012, 7, e39945. [Google Scholar] [CrossRef] [PubMed]
- Pawson, T.; Nash, P. Protein-protein interactions define specificity in signal transduction. Genes. Dev. 2000, 14, 1027–1047. [Google Scholar] [CrossRef] [PubMed]
- Testa, J.R.; Bellacosa, A. AKT plays a central role in tumorigenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 10983–10985. [Google Scholar] [CrossRef]
- Memmott, R.M.; Dennis, P.A. Akt-dependent and -independent mechanisms of mTOR regulation in cancer. Cell Signal 2009, 21, 656–664. [Google Scholar] [CrossRef]
- Fu, W.; Hall, M.N. Regulation of mTORC2 Signaling. Genes 2020, 11, 1045. [Google Scholar] [CrossRef]
- Zhao, Y.; Adjei, A.A. The clinical development of MEK inhibitors. Nat. Rev. Clin. Oncol. 2014, 11, 385–400. [Google Scholar] [CrossRef]
- Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS mutation: From undruggable to druggable in cancer. Signal Transduct. Target. Ther. 2021, 6, 386. [Google Scholar] [CrossRef]
- Yan, D.; Hao, Q.; Chen, Y.; Li, Z.; Zhang, H.; Yuan, K.; Li, R.; Li, R.; Zhao, Y.; Wang, K.; et al. mTOR-FABP4 signal is activated in brain arteriovenous malformations in humans. J. Mol. Med. 2022, 100, 1287–1297. [Google Scholar] [CrossRef] [PubMed]
- Gautam, A.; Sun, Z.; Winkler, E.; Su, H.; McCalmont, T.H.; Kim, H.; Tihan, T.; Hoffman, W.Y.; Dowd, C.F.; Frieden, I.J.; et al. Concurrent presentation of brain arteriovenous malformation, peripheral arteriovenous malformation, and cerebellar astrocytoma: Case report. Interdiscip. Neurosurg. 2020, 20, 100689. [Google Scholar] [CrossRef] [PubMed]
- Soblet, J.; Limaye, N.; Uebelhoer, M.; Boon, L.M.; Vikkula, M. Variable Somatic TIE2 Mutations in Half of Sporadic Venous Malformations. Mol. Syndr. 2013, 4, 179–183. [Google Scholar] [CrossRef] [PubMed]
- Saharinen, P.; Eklund, L.; Alitalo, K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat. Rev. Drug Discov. 2017, 16, 635–661. [Google Scholar] [CrossRef] [PubMed]
- Soon, K.; Li, M.; Wu, R.; Zhou, A.; Khosraviani, N.; Turner, W.D.; Wythe, J.D.; Fish, J.E.; Nunes, S.S. A human model of arteriovenous malformation (AVM)-on-a-chip reproduces key disease hallmarks and enables drug testing in perfused human vessel networks. Biomaterials 2022, 288, 121729. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One drug, many effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef] [PubMed]
- Seront, E.; Van Damme, A.; Legrand, C.; Bisdorff-Bresson, A.; Orcel, P.; Funck-Brentano, T.; Sevestre, M.A.; Dompmartin, A.; Quere, I.; Brouillard, P.; et al. Preliminary results of the European multicentric phase III trial regarding sirolimus in slow-flow vascular malformations. JCI Insight 2023, 8. [Google Scholar] [CrossRef] [PubMed]
- Sebold, A.J.; Day, A.M.; Ewen, J.; Adamek, J.; Byars, A.; Cohen, B.; Kossoff, E.H.; Mizuno, T.; Ryan, M.; Sievers, J.; et al. Sirolimus Treatment in Sturge-Weber Syndrome. Pediatr. Neurol. 2021, 115, 29–40. [Google Scholar] [CrossRef]
- Govindarajan, V.; Burks, J.D.; Luther, E.M.; Thompson, J.W.; Starke, R.M. Medical Adjuvants in the Treatment of Surgically Refractory Arteriovenous Malformations of the Head and Face: Case Report and Review of Literature. Cerebrovasc. Dis. 2021, 50, 493–499. [Google Scholar] [CrossRef]
- Dredge, K.; Horsfall, R.; Robinson, S.P.; Zhang, L.H.; Lu, L.; Tang, Y.; Shirley, M.A.; Muller, G.; Schafer, P.; Stirling, D.; et al. Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Microvasc. Res. 2005, 69, 56–63. [Google Scholar] [CrossRef]
- Zhu, W.; Chen, W.; Zou, D.; Wang, L.; Bao, C.; Zhan, L.; Saw, D.; Wang, S.; Winkler, E.; Li, Z.; et al. Thalidomide Reduces Hemorrhage of Brain Arteriovenous Malformations in a Mouse Model. Stroke 2018, 49, 1232–1240. [Google Scholar] [CrossRef] [PubMed]
- Kenyon, B.M.; Browne, F.; D’Amato, R.J. Effects of thalidomide and related metabolites in a mouse corneal model of neovascularization. Exp. Eye Res. 1997, 64, 971–978. [Google Scholar] [CrossRef] [PubMed]
- Invernizzi, R.; Quaglia, F.; Klersy, C.; Pagella, F.; Ornati, F.; Chu, F.; Matti, E.; Spinozzi, G.; Plumitallo, S.; Grignani, P.; et al. Efficacy and safety of thalidomide for the treatment of severe recurrent epistaxis in hereditary haemorrhagic telangiectasia: Results of a non-randomised, single-centre, phase 2 study. Lancet Haematol. 2015, 2, e465–e473. [Google Scholar] [CrossRef] [PubMed]
- Muster, R.; Ko, N.; Smith, W.; Su, H.; Dickey, M.A.; Nelson, J.; McCulloch, C.E.; Sneed, P.K.; Clarke, J.L.; Saloner, D.A.; et al. Proof-of-concept single-arm trial of bevacizumab therapy for brain arteriovenous malformation. BMJ Neurol. Open 2021, 3, e000114. [Google Scholar] [CrossRef] [PubMed]
- Al-Samkari, H. Hereditary hemorrhagic telangiectasia: Systemic therapies, guidelines, and an evolving standard of care. Blood 2021, 137, 888–895. [Google Scholar] [CrossRef] [PubMed]
- Al-Samkari, H.; Kasthuri, R.S.; Parambil, J.G.; Albitar, H.A.; Almodallal, Y.A.; Vazquez, C.; Serra, M.M.; Dupuis-Girod, S.; Wilsen, C.B.; McWilliams, J.P.; et al. An international, multicenter study of intravenous bevacizumab for bleeding in hereditary hemorrhagic telangiectasia: The InHIBIT-Bleed study. Haematologica 2021, 106, 2161–2169. [Google Scholar] [CrossRef]
- Seebauer, C.T.; Wiens, B.; Hintschich, C.A.; Platz Batista da Silva, N.; Evert, K.; Haubner, F.; Kapp, F.G.; Wendl, C.; Renner, K.; Bohr, C.; et al. Targeting the microenvironment in the treatment of arteriovenous malformations. Angiogenesis 2024, 27, 91–103. [Google Scholar] [CrossRef] [PubMed]
- Zivi, A.; Cerbone, L.; Recine, F.; Sternberg, C.N. Safety and tolerability of pazopanib in the treatment of renal cell carcinoma. Expert. Opin. Drug Saf. 2012, 11, 851–859. [Google Scholar] [CrossRef] [PubMed]
- Jonasch, E.; McCutcheon, I.E.; Gombos, D.S.; Ahrar, K.; Perrier, N.D.; Liu, D.; Robichaux, C.C.; Villarreal, M.F.; Weldon, J.A.; Woodson, A.H.; et al. Pazopanib in patients with von Hippel-Lindau disease: A single-arm, single-centre, phase 2 trial. Lancet Oncol. 2018, 19, 1351–1359. [Google Scholar] [CrossRef]
- Abe, H.; Kikuchi, S.; Hayakawa, K.; Iida, T.; Nagahashi, N.; Maeda, K.; Sakamoto, J.; Matsumoto, N.; Miura, T.; Matsumura, K.; et al. Discovery of a Highly Potent and Selective MEK Inhibitor: GSK1120212 (JTP-74057 DMSO Solvate). ACS Med. Chem. Lett. 2011, 2, 320–324. [Google Scholar] [CrossRef]
- Maruani, A.; Tavernier, E.; Boccara, O.; Mazereeuw-Hautier, J.; Leducq, S.; Bessis, D.; Guibaud, L.; Vabres, P.; Carmignac, V.; Mallet, S.; et al. Sirolimus (Rapamycin) for Slow-Flow Malformations in Children: The Observational-Phase Randomized Clinical PERFORMUS Trial. JAMA Dermatol. 2021, 157, 1289–1298. [Google Scholar] [CrossRef]
- Raper, D.M.S.; Winkler, E.A.; Rutledge, W.C.; Cooke, D.L.; Abla, A.A. An Update on Medications for Brain Arteriovenous Malformations. Neurosurgery 2020, 87, 871–878. [Google Scholar] [CrossRef] [PubMed]
- Ferrara, N.; Hillan, K.J.; Gerber, H.P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400. [Google Scholar] [CrossRef]
- Dupuis-Girod, S.; Riviere, S.; Lavigne, C.; Fargeton, A.E.; Gilbert-Dussardier, B.; Grobost, V.; Leguy-Seguin, V.; Maillard, H.; Mohamed, S.; Decullier, E.; et al. Efficacy and safety of intravenous bevacizumab on severe bleeding associated with hemorrhagic hereditary telangiectasia: A national, randomized multicenter trial. J. Intern. Med. 2023, 294, 761–774. [Google Scholar] [CrossRef]
- Derdeyn, C.P.; Zipfel, G.J.; Albuquerque, F.C.; Cooke, D.L.; Feldmann, E.; Sheehan, J.P.; Torner, J.C.; American Heart Association Stroke, C. Management of Brain Arteriovenous Malformations: A Scientific Statement for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke 2017, 48, e200–e224. [Google Scholar] [CrossRef]
- Kato, Y.; Dong, V.H.; Chaddad, F.; Takizawa, K.; Izumo, T.; Fukuda, H.; Hara, T.; Kikuta, K.; Nakai, Y.; Endo, T.; et al. Expert Consensus on the Management of Brain Arteriovenous Malformations. Asian J. Neurosurg. 2019, 14, 1074–1081. [Google Scholar] [CrossRef] [PubMed]
- Naranbhai, N.; Perez, R. Management of Brain Arteriovenous Malformations: A Review. Cureus 2023, 15, e34053. [Google Scholar] [CrossRef] [PubMed]
- Corada, M.; Morini, M.F.; Dejana, E. Signaling pathways in the specification of arteries and veins. Arter. Thromb. Vasc. Biol. 2014, 34, 2372–2377. [Google Scholar] [CrossRef]
- Fish, J.E.; Wythe, J.D. The molecular regulation of arteriovenous specification and maintenance. Dev. Dyn. 2015, 244, 391–409. [Google Scholar] [CrossRef]
- Naito, H.; Iba, T.; Takakura, N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int. Immunol. 2020, 32, 295–305. [Google Scholar] [CrossRef]
- Marcelo, K.L.; Goldie, L.C.; Hirschi, K.K. Regulation of endothelial cell differentiation and specification. Circ. Res. 2013, 112, 1272–1287. [Google Scholar] [CrossRef] [PubMed]
- Risau, W. Mechanisms of angiogenesis. Nature 1997, 386, 671–674. [Google Scholar] [CrossRef] [PubMed]
- Casie Chetty, S.; Rost, M.S.; Enriquez, J.R.; Schumacher, J.A.; Baltrunaite, K.; Rossi, A.; Stainier, D.Y.; Sumanas, S. Vegf signaling promotes vascular endothelial differentiation by modulating etv2 expression. Dev. Biol. 2017, 424, 147–161. [Google Scholar] [CrossRef]
- Kohli, V.; Schumacher, J.A.; Desai, S.P.; Rehn, K.; Sumanas, S. Arterial and venous progenitors of the major axial vessels originate at distinct locations. Dev. Cell 2013, 25, 196–206. [Google Scholar] [CrossRef] [PubMed]
- Lawson, N.D.; Vogel, A.M.; Weinstein, B.M. sonic hedgehog and vascular endothelial growth factor act upstream of the Notch pathway during arterial endothelial differentiation. Dev. Cell 2002, 3, 127–136. [Google Scholar] [CrossRef] [PubMed]
- Phng, L.K.; Gerhardt, H. Angiogenesis: A team effort coordinated by notch. Dev. Cell 2009, 16, 196–208. [Google Scholar] [CrossRef] [PubMed]
- Blanco, R.; Gerhardt, H. VEGF and Notch in tip and stalk cell selection. Cold Spring Harb. Perspect. Med. 2013, 3, a006569. [Google Scholar] [CrossRef] [PubMed]
- Pitulescu, M.E.; Schmidt, I.; Giaimo, B.D.; Antoine, T.; Berkenfeld, F.; Ferrante, F.; Park, H.; Ehling, M.; Biljes, D.; Rocha, S.F.; et al. Dll4 and Notch signalling couples sprouting angiogenesis and artery formation. Nat. Cell Biol. 2017, 19, 915–927. [Google Scholar] [CrossRef] [PubMed]
- Tian, X.; Hu, T.; Zhang, H.; He, L.; Huang, X.; Liu, Q.; Yu, W.; He, L.; Yang, Z.; Yan, Y.; et al. Vessel formation. De novo formation of a distinct coronary vascular population in neonatal heart. Science 2014, 345, 90–94. [Google Scholar] [CrossRef]
- You, L.R.; Lin, F.J.; Lee, C.T.; DeMayo, F.J.; Tsai, M.J.; Tsai, S.Y. Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity. Nature 2005, 435, 98–104. [Google Scholar] [CrossRef]
- Neal, A.; Nornes, S.; Payne, S.; Wallace, M.D.; Fritzsche, M.; Louphrasitthiphol, P.; Wilkinson, R.N.; Chouliaras, K.M.; Liu, K.; Plant, K.; et al. Venous identity requires BMP signalling through ALK3. Nat. Commun. 2019, 10, 453. [Google Scholar] [CrossRef] [PubMed]
- Payne, S.; Gunadasa-Rohling, M.; Neal, A.; Redpath, A.N.; Patel, J.; Chouliaras, K.M.; Ratnayaka, I.; Smart, N.; De Val, S. Regulatory pathways governing murine coronary vessel formation are dysregulated in the injured adult heart. Nat. Commun. 2019, 10, 3276. [Google Scholar] [CrossRef] [PubMed]
- Bahary, N.; Goishi, K.; Stuckenholz, C.; Weber, G.; Leblanc, J.; Schafer, C.A.; Berman, S.S.; Klagsbrun, M.; Zon, L.I. Duplicate VegfA genes and orthologues of the KDR receptor tyrosine kinase family mediate vascular development in the zebrafish. Blood 2007, 110, 3627–3636. [Google Scholar] [CrossRef] [PubMed]
- Benedito, R.; Hellstrom, M. Notch as a hub for signaling in angiogenesis. Exp. Cell Res. 2013, 319, 1281–1288. [Google Scholar] [CrossRef] [PubMed]
- Bussmann, J.; Wolfe, S.A.; Siekmann, A.F. Arterial-venous network formation during brain vascularization involves hemodynamic regulation of chemokine signaling. Development 2011, 138, 1717–1726. [Google Scholar] [CrossRef] [PubMed]
- Covassin, L.D.; Villefranc, J.A.; Kacergis, M.C.; Weinstein, B.M.; Lawson, N.D. Distinct genetic interactions between multiple Vegf receptors are required for development of different blood vessel types in zebrafish. Proc. Natl. Acad. Sci. USA 2006, 103, 6554–6559. [Google Scholar] [CrossRef] [PubMed]
- Ehling, M.; Adams, S.; Benedito, R.; Adams, R.H. Notch controls retinal blood vessel maturation and quiescence. Development 2013, 140, 3051–3061. [Google Scholar] [CrossRef] [PubMed]
- Hellstrom, M.; Phng, L.K.; Hofmann, J.J.; Wallgard, E.; Coultas, L.; Lindblom, P.; Alva, J.; Nilsson, A.K.; Karlsson, L.; Gaiano, N.; et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 2007, 445, 776–780. [Google Scholar] [CrossRef] [PubMed]
- Leslie, J.D.; Ariza-McNaughton, L.; Bermange, A.L.; McAdow, R.; Johnson, S.L.; Lewis, J. Endothelial signalling by the Notch ligand Delta-like 4 restricts angiogenesis. Development 2007, 134, 839–844. [Google Scholar] [CrossRef]
- Red-Horse, K.; Siekmann, A.F. Veins and Arteries Build Hierarchical Branching Patterns Differently: Bottom-Up versus Top-Down. Bioessays 2019, 41, e1800198. [Google Scholar] [CrossRef]
- Siekmann, A.F.; Covassin, L.; Lawson, N.D. Modulation of VEGF signalling output by the Notch pathway. Bioessays 2008, 30, 303–313. [Google Scholar] [CrossRef]
- Suchting, S.; Freitas, C.; le Noble, F.; Benedito, R.; Breant, C.; Duarte, A.; Eichmann, A. The Notch ligand Delta-like 4 negatively regulates endothelial tip cell formation and vessel branching. Proc. Natl. Acad. Sci. USA 2007, 104, 3225–3230. [Google Scholar] [CrossRef]
- Zygmunt, T.; Trzaska, S.; Edelstein, L.; Walls, J.; Rajamani, S.; Gale, N.; Daroles, L.; Ramirez, C.; Ulrich, F.; Torres-Vazquez, J. ‘In parallel’ interconnectivity of the dorsal longitudinal anastomotic vessels requires both VEGF signaling and circulatory flow. J. Cell Sci. 2012, 125, 5159–5167. [Google Scholar] [CrossRef] [PubMed]
- Chappell, J.C.; Taylor, S.M.; Ferrara, N.; Bautch, V.L. Local guidance of emerging vessel sprouts requires soluble Flt-1. Dev. Cell 2009, 17, 377–386. [Google Scholar] [CrossRef] [PubMed]
- Jakobsson, L.; Franco, C.A.; Bentley, K.; Collins, R.T.; Ponsioen, B.; Aspalter, I.M.; Rosewell, I.; Busse, M.; Thurston, G.; Medvinsky, A.; et al. Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat. Cell Biol. 2010, 12, 943–953. [Google Scholar] [CrossRef] [PubMed]
- Carmeliet, P.; Ferreira, V.; Breier, G.; Pollefeyt, S.; Kieckens, L.; Gertsenstein, M.; Fahrig, M.; Vandenhoeck, A.; Harpal, K.; Eberhardt, C.; et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 1996, 380, 435–439. [Google Scholar] [CrossRef]
- Ferrara, N.; Carver-Moore, K.; Chen, H.; Dowd, M.; Lu, L.; O’Shea, K.S.; Powell-Braxton, L.; Hillan, K.J.; Moore, M.W. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature 1996, 380, 439–442. [Google Scholar] [CrossRef]
- Shalaby, F.; Rossant, J.; Yamaguchi, T.P.; Gertsenstein, M.; Wu, X.F.; Breitman, M.L.; Schuh, A.C. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 1995, 376, 62–66. [Google Scholar] [CrossRef]
- Fong, G.H.; Rossant, J.; Gertsenstein, M.; Breitman, M.L. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 1995, 376, 66–70. [Google Scholar] [CrossRef]
- Winkler, E.A.; Kim, C.N.; Ross, J.M.; Garcia, J.H.; Gil, E.; Oh, I.; Chen, L.Q.; Wu, D.; Catapano, J.S.; Raygor, K.; et al. A single-cell atlas of the normal and malformed human brain vasculature. Science 2022, 375, eabi7377. [Google Scholar] [CrossRef]
- Lee, H.W.; Xu, Y.; He, L.; Choi, W.; Gonzalez, D.; Jin, S.W.; Simons, M. Role of Venous Endothelial Cells in Developmental and Pathologic Angiogenesis. Circulation 2021, 144, 1308–1322. [Google Scholar] [CrossRef]
- McDonald, A.I.; Shirali, A.S.; Aragon, R.; Ma, F.; Hernandez, G.; Vaughn, D.A.; Mack, J.J.; Lim, T.Y.; Sunshine, H.; Zhao, P.; et al. Endothelial Regeneration of Large Vessels Is a Biphasic Process Driven by Local Cells with Distinct Proliferative Capacities. Cell Stem Cell 2018, 23, 210–225 e216. [Google Scholar] [CrossRef]
- Hou, S.; Li, Z.; Dong, J.; Gao, Y.; Chang, Z.; Ding, X.; Li, S.; Li, Y.; Zeng, Y.; Xin, Q.; et al. Heterogeneity in endothelial cells and widespread venous arterialization during early vascular development in mammals. Cell Res. 2022, 32, 333–348. [Google Scholar] [CrossRef]
- Park, H.; Furtado, J.; Poulet, M.; Chung, M.; Yun, S.; Lee, S.; Sessa, W.C.; Franco, C.A.; Schwartz, M.A.; Eichmann, A. Defective Flow-Migration Coupling Causes Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia. Circulation 2021, 144, 805–822. [Google Scholar] [CrossRef]
- Lee, H.W.; Shin, J.H.; Simons, M. Flow goes forward and cells step backward: Endothelial migration. Exp. Mol. Med. 2022, 54, 711–719. [Google Scholar] [CrossRef] [PubMed]
- Roman, B.L.; Hinck, A.P. ALK1 signaling in development and disease: New paradigms. Cell Mol. Life Sci. 2017, 74, 4539–4560. [Google Scholar] [CrossRef] [PubMed]
- Jin, Y.; Muhl, L.; Burmakin, M.; Wang, Y.; Duchez, A.C.; Betsholtz, C.; Arthur, H.M.; Jakobsson, L. Endoglin prevents vascular malformation by regulating flow-induced cell migration and specification through VEGFR2 signalling. Nat. Cell Biol. 2017, 19, 639–652. [Google Scholar] [CrossRef] [PubMed]
- Poduri, A.; Chang, A.H.; Raftrey, B.; Rhee, S.; Van, M.; Red-Horse, K. Endothelial cells respond to the direction of mechanical stimuli through SMAD signaling to regulate coronary artery size. Development 2017, 144, 3241–3252. [Google Scholar] [CrossRef]
- Chang, A.H.; Raftrey, B.C.; D’Amato, G.; Surya, V.N.; Poduri, A.; Chen, H.I.; Goldstone, A.B.; Woo, J.; Fuller, G.G.; Dunn, A.R.; et al. DACH1 stimulates shear stress-guided endothelial cell migration and coronary artery growth through the CXCL12-CXCR4 signaling axis. Genes. Dev. 2017, 31, 1308–1324. [Google Scholar] [CrossRef]
- Raftrey, B.; Williams, M.; Rios Coronado, P.E.; Fan, X.; Chang, A.H.; Zhao, M.; Roth, R.; Trimm, E.; Racelis, R.; D’Amato, G.; et al. Dach1 Extends Artery Networks and Protects Against Cardiac Injury. Circ. Res. 2021, 129, 702–716. [Google Scholar] [CrossRef]
- Franco, C.A.; Jones, M.L.; Bernabeu, M.O.; Vion, A.C.; Barbacena, P.; Fan, J.; Mathivet, T.; Fonseca, C.G.; Ragab, A.; Yamaguchi, T.P.; et al. Non-canonical Wnt signalling modulates the endothelial shear stress flow sensor in vascular remodelling. Elife 2016, 5, e07727. [Google Scholar] [CrossRef]
- Dubrac, A.; Genet, G.; Ola, R.; Zhang, F.; Pibouin-Fragner, L.; Han, J.; Zhang, J.; Thomas, J.L.; Chedotal, A.; Schwartz, M.A.; et al. Targeting NCK-Mediated Endothelial Cell Front-Rear Polarity Inhibits Neovascularization. Circulation 2016, 133, 409–421. [Google Scholar] [CrossRef]
- Genet, G.; Boye, K.; Mathivet, T.; Ola, R.; Zhang, F.; Dubrac, A.; Li, J.; Genet, N.; Henrique Geraldo, L.; Benedetti, L.; et al. Endophilin-A2 dependent VEGFR2 endocytosis promotes sprouting angiogenesis. Nat. Commun. 2019, 10, 2350. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.H.; Choi, J.; Yang, M.J.; Hong, S.P.; Lee, C.K.; Kubota, Y.; Lim, D.S.; Koh, G.Y. A MST1-FOXO1 cascade establishes endothelial tip cell polarity and facilitates sprouting angiogenesis. Nat. Commun. 2019, 10, 838. [Google Scholar] [CrossRef] [PubMed]
- Lavina, B.; Castro, M.; Niaudet, C.; Cruys, B.; Alvarez-Aznar, A.; Carmeliet, P.; Bentley, K.; Brakebusch, C.; Betsholtz, C.; Gaengel, K. Defective endothelial cell migration in the absence of Cdc42 leads to capillary-venous malformations. Development 2018, 145. [Google Scholar] [CrossRef] [PubMed]
- Geudens, I.; Coxam, B.; Alt, S.; Gebala, V.; Vion, A.C.; Meier, K.; Rosa, A.; Gerhardt, H. Artery-vein specification in the zebrafish trunk is pre-patterned by heterogeneous Notch activity and balanced by flow-mediated fine-tuning. Development 2019, 146. [Google Scholar] [CrossRef] [PubMed]
- Rosa, A.; Giese, W.; Meier, K.; Alt, S.; Klaus-Bergmann, A.; Edgar, L.T.; Bartels-Klein, E.; Collins, R.T.; Szymborska, A.; Coxam, B.; et al. WASp controls oriented migration of endothelial cells to achieve functional vascular patterning. Development 2022, 149. [Google Scholar] [CrossRef]
- Weijts, B.; Gutierrez, E.; Saikin, S.K.; Ablooglu, A.J.; Traver, D.; Groisman, A.; Tkachenko, E. Blood flow-induced Notch activation and endothelial migration enable vascular remodeling in zebrafish embryos. Nat. Commun. 2018, 9, 5314. [Google Scholar] [CrossRef]
- Franco, C.A.; Jones, M.L.; Bernabeu, M.O.; Geudens, I.; Mathivet, T.; Rosa, A.; Lopes, F.M.; Lima, A.P.; Ragab, A.; Collins, R.T.; et al. Dynamic endothelial cell rearrangements drive developmental vessel regression. PLoS Biol. 2015, 13, e1002125. [Google Scholar] [CrossRef]
- Aird, W.C. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ. Res. 2007, 100, 158–173. [Google Scholar] [CrossRef]
- Komarova, Y.A.; Kruse, K.; Mehta, D.; Malik, A.B. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ. Res. 2017, 120, 179–206. [Google Scholar] [CrossRef]
- Stamatovic, S.M.; Johnson, A.M.; Keep, R.F.; Andjelkovic, A.V. Junctional proteins of the blood-brain barrier: New insights into function and dysfunction. Tissue Barriers 2016, 4, e1154641. [Google Scholar] [CrossRef] [PubMed]
- Duong, C.N.; Vestweber, D. Mechanisms Ensuring Endothelial Junction Integrity Beyond VE-Cadherin. Front. Physiol. 2020, 11, 519. [Google Scholar] [CrossRef] [PubMed]
- Abbruscato, T.J.; Davis, T.P. Protein expression of brain endothelial cell E-cadherin after hypoxia/aglycemia: Influence of astrocyte contact. Brain Res. 1999, 842, 277–286. [Google Scholar] [CrossRef]
- Luo, Y.; Radice, G.L. N-cadherin acts upstream of VE-cadherin in controlling vascular morphogenesis. J. Cell Biol. 2005, 169, 29–34. [Google Scholar] [CrossRef] [PubMed]
- Orsenigo, F.; Giampietro, C.; Ferrari, A.; Corada, M.; Galaup, A.; Sigismund, S.; Ristagno, G.; Maddaluno, L.; Koh, G.Y.; Franco, D.; et al. Phosphorylation of VE-cadherin is modulated by haemodynamic forces and contributes to the regulation of vascular permeability in vivo. Nat. Commun. 2012, 3, 1208. [Google Scholar] [CrossRef] [PubMed]
- Carmeliet, P.; Lampugnani, M.G.; Moons, L.; Breviario, F.; Compernolle, V.; Bono, F.; Balconi, G.; Spagnuolo, R.; Oosthuyse, B.; Dewerchin, M.; et al. Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 1999, 98, 147–157. [Google Scholar] [CrossRef]
- Gulino, D.; Delachanal, E.; Concord, E.; Genoux, Y.; Morand, B.; Valiron, M.O.; Sulpice, E.; Scaife, R.; Alemany, M.; Vernet, T. Alteration of endothelial cell monolayer integrity triggers resynthesis of vascular endothelium cadherin. J. Biol. Chem. 1998, 273, 29786–29793. [Google Scholar] [CrossRef]
- Corada, M.; Mariotti, M.; Thurston, G.; Smith, K.; Kunkel, R.; Brockhaus, M.; Lampugnani, M.G.; Martin-Padura, I.; Stoppacciaro, A.; Ruco, L.; et al. Vascular endothelial-cadherin is an important determinant of microvascular integrity in vivo. Proc. Natl. Acad. Sci. USA 1999, 96, 9815–9820. [Google Scholar] [CrossRef]
- Frye, M.; Dierkes, M.; Kuppers, V.; Vockel, M.; Tomm, J.; Zeuschner, D.; Rossaint, J.; Zarbock, A.; Koh, G.Y.; Peters, K.; et al. Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin. J. Exp. Med. 2015, 212, 2267–2287. [Google Scholar] [CrossRef]
- Gotsch, U.; Borges, E.; Bosse, R.; Boggemeyer, E.; Simon, M.; Mossmann, H.; Vestweber, D. VE-cadherin antibody accelerates neutrophil recruitment in vivo. J. Cell Sci. 1997, 110 Pt 5, 583–588. [Google Scholar] [CrossRef] [PubMed]
- Matsuyoshi, N.; Toda, K.; Horiguchi, Y.; Tanaka, T.; Nakagawa, S.; Takeichi, M.; Imamura, S. In vivo evidence of the critical role of cadherin-5 in murine vascular integrity. Proc. Assoc. Am. Physicians 1997, 109, 362–371. [Google Scholar] [PubMed]
- Boulday, G.; Rudini, N.; Maddaluno, L.; Blecon, A.; Arnould, M.; Gaudric, A.; Chapon, F.; Adams, R.H.; Dejana, E.; Tournier-Lasserve, E. Developmental timing of CCM2 loss influences cerebral cavernous malformations in mice. J. Exp. Med. 2011, 208, 1835–1847. [Google Scholar] [CrossRef]
- Bravi, L.; Rudini, N.; Cuttano, R.; Giampietro, C.; Maddaluno, L.; Ferrarini, L.; Adams, R.H.; Corada, M.; Boulday, G.; Tournier-Lasserve, E.; et al. Sulindac metabolites decrease cerebrovascular malformations in CCM3-knockout mice. Proc. Natl. Acad. Sci. USA 2015, 112, 8421–8426. [Google Scholar] [CrossRef]
- Glading, A.; Han, J.; Stockton, R.A.; Ginsberg, M.H. KRIT-1/CCM1 is a Rap1 effector that regulates endothelial cell cell junctions. J. Cell Biol. 2007, 179, 247–254. [Google Scholar] [CrossRef] [PubMed]
- Maddaluno, L.; Rudini, N.; Cuttano, R.; Bravi, L.; Giampietro, C.; Corada, M.; Ferrarini, L.; Orsenigo, F.; Papa, E.; Boulday, G.; et al. EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature 2013, 498, 492–496. [Google Scholar] [CrossRef]
- Xu, H.; Huo, R.; Li, H.; Jiao, Y.; Weng, J.; Wang, J.; Yan, Z.; Zhang, J.; Zhao, S.; He, Q.; et al. KRAS mutation-induced EndMT of brain arteriovenous malformation is mediated through the TGF-beta/BMP-SMAD4 pathway. Stroke Vasc. Neurol. 2023, 8, 197–206. [Google Scholar] [CrossRef]
- Hermans, D.; Rodriguez-Mogeda, C.; Kemps, H.; Bronckaers, A.; de Vries, H.E.; Broux, B. Nectins and Nectin-like molecules drive vascular development and barrier function. Angiogenesis 2023, 26, 349–362. [Google Scholar] [CrossRef]
- Takai, Y.; Irie, K.; Shimizu, K.; Sakisaka, T.; Ikeda, W. Nectins and nectin-like molecules: Roles in cell adhesion, migration, and polarization. Cancer Sci. 2003, 94, 655–667. [Google Scholar] [CrossRef]
- Bekes, I.; Lob, S.; Holzheu, I.; Janni, W.; Baumann, L.; Wockel, A.; Wulff, C. Nectin-2 in ovarian cancer: How is it expressed and what might be its functional role? Cancer Sci. 2019, 110, 1872–1882. [Google Scholar] [CrossRef]
- Martin, T.A.; Lane, J.; Harrison, G.M.; Jiang, W.G. The expression of the Nectin complex in human breast cancer and the role of Nectin-3 in the control of tight junctions during metastasis. PLoS ONE 2013, 8, e82696. [Google Scholar] [CrossRef] [PubMed]
- Reymond, N.; Imbert, A.M.; Devilard, E.; Fabre, S.; Chabannon, C.; Xerri, L.; Farnarier, C.; Cantoni, C.; Bottino, C.; Moretta, A.; et al. DNAM-1 and PVR regulate monocyte migration through endothelial junctions. J. Exp. Med. 2004, 199, 1331–1341. [Google Scholar] [CrossRef] [PubMed]
- Son, Y.; Lee, B.; Choi, Y.J.; Jeon, S.A.; Kim, J.H.; Lee, H.K.; Kwon, S.M.; Cho, J.Y. Nectin-2 (CD112) Is Expressed on Outgrowth Endothelial Cells and Regulates Cell Proliferation and Angiogenic Function. PLoS ONE 2016, 11, e0163301. [Google Scholar] [CrossRef] [PubMed]
- Lampugnani, M.G.; Dejana, E.; Giampietro, C. Vascular Endothelial (VE)-Cadherin, Endothelial Adherens Junctions, and Vascular Disease. Cold Spring Harb. Perspect. Biol. 2018, 10, a029322. [Google Scholar] [CrossRef] [PubMed]
- Simionescu, M.; Simionescu, N.; Palade, G.E. Segmental differentiations of cell junctions in the vascular endothelium. Arteries and veins. J. Cell Biol. 1976, 68, 705–723. [Google Scholar] [CrossRef] [PubMed]
- Tietz, S.; Engelhardt, B. Brain barriers: Crosstalk between complex tight junctions and adherens junctions. J. Cell Biol. 2015, 209, 493–506. [Google Scholar] [CrossRef] [PubMed]
- Morita, K.; Sasaki, H.; Furuse, M.; Tsukita, S. Endothelial claudin: Claudin-5/TMVCF constitutes tight junction strands in endothelial cells. J. Cell Biol. 1999, 147, 185–194. [Google Scholar] [CrossRef] [PubMed]
- Krause, G.; Winkler, L.; Mueller, S.L.; Haseloff, R.F.; Piontek, J.; Blasig, I.E. Structure and function of claudins. Biochim. Biophys. Acta 2008, 1778, 631–645. [Google Scholar] [CrossRef] [PubMed]
- Daneman, R.; Zhou, L.; Agalliu, D.; Cahoy, J.D.; Kaushal, A.; Barres, B.A. The mouse blood-brain barrier transcriptome: A new resource for understanding the development and function of brain endothelial cells. PLoS ONE 2010, 5, e13741. [Google Scholar] [CrossRef]
- Nitta, T.; Hata, M.; Gotoh, S.; Seo, Y.; Sasaki, H.; Hashimoto, N.; Furuse, M.; Tsukita, S. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J. Cell Biol. 2003, 161, 653–660. [Google Scholar] [CrossRef]
- Ohtsuki, S.; Yamaguchi, H.; Katsukura, Y.; Asashima, T.; Terasaki, T. mRNA expression levels of tight junction protein genes in mouse brain capillary endothelial cells highly purified by magnetic cell sorting. J. Neurochem. 2008, 104, 147–154. [Google Scholar] [CrossRef] [PubMed]
- Liebner, S.; Corada, M.; Bangsow, T.; Babbage, J.; Taddei, A.; Czupalla, C.J.; Reis, M.; Felici, A.; Wolburg, H.; Fruttiger, M.; et al. Wnt/beta-catenin signaling controls development of the blood-brain barrier. J. Cell Biol. 2008, 183, 409–417. [Google Scholar] [CrossRef] [PubMed]
- Sadowska, G.B.; Ahmedli, N.; Chen, X.; Stonestreet, B.S. Ontogeny of tight junction protein expression in the ovine cerebral cortex during development. Neuroscience 2015, 310, 422–429. [Google Scholar] [CrossRef] [PubMed]
- Saitou, M.; Furuse, M.; Sasaki, H.; Schulzke, J.D.; Fromm, M.; Takano, H.; Noda, T.; Tsukita, S. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol. Biol. Cell 2000, 11, 4131–4142. [Google Scholar] [CrossRef] [PubMed]
- Bellmann, C.; Schreivogel, S.; Gunther, R.; Dabrowski, S.; Schumann, M.; Wolburg, H.; Blasig, I.E. Highly conserved cysteines are involved in the oligomerization of occludin-redox dependency of the second extracellular loop. Antioxid. Redox Signal 2014, 20, 855–867. [Google Scholar] [CrossRef] [PubMed]
- Buschmann, M.M.; Shen, L.; Rajapakse, H.; Raleigh, D.R.; Wang, Y.; Wang, Y.; Lingaraju, A.; Zha, J.; Abbott, E.; McAuley, E.M.; et al. Occludin OCEL-domain interactions are required for maintenance and regulation of the tight junction barrier to macromolecular flux. Mol. Biol. Cell 2013, 24, 3056–3068. [Google Scholar] [CrossRef] [PubMed]
- Cording, J.; Berg, J.; Kading, N.; Bellmann, C.; Tscheik, C.; Westphal, J.K.; Milatz, S.; Gunzel, D.; Wolburg, H.; Piontek, J.; et al. In tight junctions, claudins regulate the interactions between occludin, tricellulin and marvelD3, which, inversely, modulate claudin oligomerization. J. Cell Sci. 2013, 126, 554–564. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Fanning, A.S.; Anderson, J.M.; Lavie, A. Structure of the conserved cytoplasmic C-terminal domain of occludin: Identification of the ZO-1 binding surface. J. Mol. Biol. 2005, 352, 151–164. [Google Scholar] [CrossRef] [PubMed]
- Nusrat, A.; Brown, G.T.; Tom, J.; Drake, A.; Bui, T.T.; Quan, C.; Mrsny, R.J. Multiple protein interactions involving proposed extracellular loop domains of the tight junction protein occludin. Mol. Biol. Cell 2005, 16, 1725–1734. [Google Scholar] [CrossRef]
- Schmidt, A.; Utepbergenov, D.I.; Mueller, S.L.; Beyermann, M.; Schneider-Mergener, J.; Krause, G.; Blasig, I.E. Occludin binds to the SH3-hinge-GuK unit of zonula occludens protein 1: Potential mechanism of tight junction regulation. Cell Mol. Life Sci. 2004, 61, 1354–1365. [Google Scholar] [CrossRef]
- Bazzoni, G.; Dejana, E. Endothelial cell-to-cell junctions: Molecular organization and role in vascular homeostasis. Physiol. Rev. 2004, 84, 869–901. [Google Scholar] [CrossRef] [PubMed]
- Bradfield, P.F.; Nourshargh, S.; Aurrand-Lions, M.; Imhof, B.A. JAM family and related proteins in leukocyte migration (Vestweber series). Arter. Thromb. Vasc. Biol. 2007, 27, 2104–2112. [Google Scholar] [CrossRef] [PubMed]
- Sobocki, T.; Sobocka, M.B.; Babinska, A.; Ehrlich, Y.H.; Banerjee, P.; Kornecki, E. Genomic structure, organization and promoter analysis of the human F11R/F11 receptor/junctional adhesion molecule-1/JAM-A. Gene 2006, 366, 128–144. [Google Scholar] [CrossRef] [PubMed]
- Williams, L.A.; Martin-Padura, I.; Dejana, E.; Hogg, N.; Simmons, D.L. Identification and characterisation of human Junctional Adhesion Molecule (JAM). Mol. Immunol. 1999, 36, 1175–1188. [Google Scholar] [CrossRef] [PubMed]
- Ebnet, K.; Aurrand-Lions, M.; Kuhn, A.; Kiefer, F.; Butz, S.; Zander, K.; Meyer zu Brickwedde, M.K.; Suzuki, A.; Imhof, B.A.; Vestweber, D. The junctional adhesion molecule (JAM) family members JAM-2 and JAM-3 associate with the cell polarity protein PAR-3: A possible role for JAMs in endothelial cell polarity. J. Cell Sci. 2003, 116, 3879–3891. [Google Scholar] [CrossRef] [PubMed]
- Itoh, M.; Sasaki, H.; Furuse, M.; Ozaki, H.; Kita, T.; Tsukita, S. Junctional adhesion molecule (JAM) binds to PAR-3: A possible mechanism for the recruitment of PAR-3 to tight junctions. J. Cell Biol. 2001, 154, 491–497. [Google Scholar] [CrossRef] [PubMed]
- Ostermann, G.; Weber, K.S.; Zernecke, A.; Schroder, A.; Weber, C. JAM-1 is a ligand of the beta(2) integrin LFA-1 involved in transendothelial migration of leukocytes. Nat. Immunol. 2002, 3, 151–158. [Google Scholar] [CrossRef] [PubMed]
- Sladojevic, N.; Stamatovic, S.M.; Keep, R.F.; Grailer, J.J.; Sarma, J.V.; Ward, P.A.; Andjelkovic, A.V. Inhibition of junctional adhesion molecule-A/LFA interaction attenuates leukocyte trafficking and inflammation in brain ischemia/reperfusion injury. Neurobiol. Dis. 2014, 67, 57–70. [Google Scholar] [CrossRef] [PubMed]
- Wojcikiewicz, E.P.; Koenen, R.R.; Fraemohs, L.; Minkiewicz, J.; Azad, H.; Weber, C.; Moy, V.T. LFA-1 binding destabilizes the JAM-A homophilic interaction during leukocyte transmigration. Biophys. J. 2009, 96, 285–293. [Google Scholar] [CrossRef]
- Nasdala, I.; Wolburg-Buchholz, K.; Wolburg, H.; Kuhn, A.; Ebnet, K.; Brachtendorf, G.; Samulowitz, U.; Kuster, B.; Engelhardt, B.; Vestweber, D.; et al. A transmembrane tight junction protein selectively expressed on endothelial cells and platelets. J. Biol. Chem. 2002, 277, 16294–16303. [Google Scholar] [CrossRef]
- Suzu, S.; Hayashi, Y.; Harumi, T.; Nomaguchi, K.; Yamada, M.; Hayasawa, H.; Motoyoshi, K. Molecular cloning of a novel immunoglobulin superfamily gene preferentially expressed by brain and testis. Biochem. Biophys. Res. Commun. 2002, 296, 1215–1221. [Google Scholar] [CrossRef] [PubMed]
- Wegmann, F.; Ebnet, K.; Du Pasquier, L.; Vestweber, D.; Butz, S. Endothelial adhesion molecule ESAM binds directly to the multidomain adaptor MAGI-1 and recruits it to cell contacts. Exp. Cell Res. 2004, 300, 121–133. [Google Scholar] [CrossRef] [PubMed]
- Ooi, A.G.; Karsunky, H.; Majeti, R.; Butz, S.; Vestweber, D.; Ishida, T.; Quertermous, T.; Weissman, I.L.; Forsberg, E.C. The adhesion molecule esam1 is a novel hematopoietic stem cell marker. Stem Cells 2009, 27, 653–661. [Google Scholar] [CrossRef]
- Yokota, T.; Oritani, K.; Butz, S.; Kokame, K.; Kincade, P.W.; Miyata, T.; Vestweber, D.; Kanakura, Y. The endothelial antigen ESAM marks primitive hematopoietic progenitors throughout life in mice. Blood 2009, 113, 2914–2923. [Google Scholar] [CrossRef]
- Itoh, M.; Furuse, M.; Morita, K.; Kubota, K.; Saitou, M.; Tsukita, S. Direct binding of three tight junction-associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J. Cell Biol. 1999, 147, 1351–1363. [Google Scholar] [CrossRef]
- Itoh, M.; Morita, K.; Tsukita, S. Characterization of ZO-2 as a MAGUK family member associated with tight as well as adherens junctions with a binding affinity to occludin and alpha catenin. J. Biol. Chem. 1999, 274, 5981–5986. [Google Scholar] [CrossRef]
- Fanning, A.S.; Little, B.P.; Rahner, C.; Utepbergenov, D.; Walther, Z.; Anderson, J.M. The unique-5 and -6 motifs of ZO-1 regulate tight junction strand localization and scaffolding properties. Mol. Biol. Cell 2007, 18, 721–731. [Google Scholar] [CrossRef] [PubMed]
- Utepbergenov, D.I.; Fanning, A.S.; Anderson, J.M. Dimerization of the scaffolding protein ZO-1 through the second PDZ domain. J. Biol. Chem. 2006, 281, 24671–24677. [Google Scholar] [CrossRef]
- Goodenough, D.A.; Goliger, J.A.; Paul, D.L. Connexins, connexons, and intercellular communication. Annu. Rev. Biochem. 1996, 65, 475–502. [Google Scholar] [CrossRef]
- Nagasawa, K.; Chiba, H.; Fujita, H.; Kojima, T.; Saito, T.; Endo, T.; Sawada, N. Possible involvement of gap junctions in the barrier function of tight junctions of brain and lung endothelial cells. J. Cell Physiol. 2006, 208, 123–132. [Google Scholar] [CrossRef]
- Nagy, J.I.; Dudek, F.E.; Rash, J.E. Update on connexins and gap junctions in neurons and glia in the mammalian nervous system. Brain Res. Brain Res. Rev. 2004, 47, 191–215. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Xin, Y.; He, Z.; Hu, W. Function of Connexins in the Interaction between Glial and Vascular Cells in the Central Nervous System and Related Neurological Diseases. Neural Plast. 2018, 2018, 6323901. [Google Scholar] [CrossRef] [PubMed]
- Danesh-Meyer, H.V.; Kerr, N.M.; Zhang, J.; Eady, E.K.; O’Carroll, S.J.; Nicholson, L.F.; Johnson, C.S.; Green, C.R. Connexin43 mimetic peptide reduces vascular leak and retinal ganglion cell death following retinal ischaemia. Brain 2012, 135, 506–520. [Google Scholar] [CrossRef] [PubMed]
- Johnson, A.M.; Roach, J.P.; Hu, A.; Stamatovic, S.M.; Zochowski, M.R.; Keep, R.F.; Andjelkovic, A.V. Connexin 43 gap junctions contribute to brain endothelial barrier hyperpermeability in familial cerebral cavernous malformations type III by modulating tight junction structure. FASEB J. 2018, 32, 2615–2629. [Google Scholar] [CrossRef] [PubMed]
- Phillips, C.M.; Johnson, A.M.; Stamatovic, S.M.; Keep, R.F.; Andjelkovic, A.V. 20 kDa isoform of connexin-43 augments spatial reorganization of the brain endothelial junctional complex and lesion leakage in cerebral cavernous malformation type-3. Neurobiol. Dis. 2023, 186, 106277. [Google Scholar] [CrossRef] [PubMed]
- Gkatzis, K.; Thalgott, J.; Dos-Santos-Luis, D.; Martin, S.; Lamande, N.; Carette, M.F.; Disch, F.; Snijder, R.J.; Westermann, C.J.; Mager, J.J.; et al. Interaction Between ALK1 Signaling and Connexin40 in the Development of Arteriovenous Malformations. Arter. Thromb. Vasc. Biol. 2016, 36, 707–717. [Google Scholar] [CrossRef] [PubMed]
- Bai, J.; Wang, Y.J.; Liu, L.; Zhao, Y.L. Ephrin B2 and EphB4 selectively mark arterial and venous vessels in cerebral arteriovenous malformation. J. Int. Med. Res. 2014, 42, 405–415. [Google Scholar] [CrossRef] [PubMed]
- Fu, W.; Huo, R.; Yan, Z.; Xu, H.; Li, H.; Jiao, Y.; Wang, L.; Weng, J.; Wang, J.; Wang, S.; et al. Mesenchymal Behavior of the Endothelium Promoted by SMAD6 Downregulation Is Associated With Brain Arteriovenous Malformation Microhemorrhage. Stroke 2020, 51, 2197–2207. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.G.; Lee, A.; Chang, W.; Lee, M.S.; Kim, J. Endothelial to Mesenchymal Transition Represents a Key Link in the Interaction between Inflammation and Endothelial Dysfunction. Front. Immunol. 2018, 9, 294. [Google Scholar] [CrossRef]
- Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [CrossRef]
- Piera-Velazquez, S.; Jimenez, S.A. Endothelial to Mesenchymal Transition: Role in Physiology and in the Pathogenesis of Human Diseases. Physiol. Rev. 2019, 99, 1281–1324. [Google Scholar] [CrossRef]
- Arciniegas, E.; Sutton, A.B.; Allen, T.D.; Schor, A.M. Transforming growth factor beta 1 promotes the differentiation of endothelial cells into smooth muscle-like cells in vitro. J. Cell Sci. 1992, 103 Pt 2, 521–529. [Google Scholar] [CrossRef]
- Welch-Reardon, K.M.; Wu, N.; Hughes, C.C. A role for partial endothelial-mesenchymal transitions in angiogenesis? Arter. Thromb. Vasc. Biol. 2015, 35, 303–308. [Google Scholar] [CrossRef] [PubMed]
- Xiao, L.; Dudley, A.C. Fine-tuning vascular fate during endothelial-mesenchymal transition. J. Pathol. 2017, 241, 25–35. [Google Scholar] [CrossRef]
- Sun, J.X.; Chang, T.F.; Li, M.H.; Sun, L.J.; Yan, X.C.; Yang, Z.Y.; Liu, Y.; Xu, W.Q.; Lv, Y.; Su, J.B.; et al. SNAI1, an endothelial-mesenchymal transition transcription factor, promotes the early phase of ocular neovascularization. Angiogenesis 2018, 21, 635–652. [Google Scholar] [CrossRef] [PubMed]
- Medici, D.; Potenta, S.; Kalluri, R. Transforming growth factor-beta2 promotes Snail-mediated endothelial-mesenchymal transition through convergence of Smad-dependent and Smad-independent signalling. Biochem. J. 2011, 437, 515–520. [Google Scholar] [CrossRef]
- Kokudo, T.; Suzuki, Y.; Yoshimatsu, Y.; Yamazaki, T.; Watabe, T.; Miyazono, K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J. Cell Sci. 2008, 121, 3317–3324. [Google Scholar] [CrossRef] [PubMed]
- Garcia, J.; Sandi, M.J.; Cordelier, P.; Binetruy, B.; Pouyssegur, J.; Iovanna, J.L.; Tournaire, R. Tie1 deficiency induces endothelial-mesenchymal transition. EMBO Rep. 2012, 13, 431–439. [Google Scholar] [CrossRef]
- Lopez, D.; Niu, G.; Huber, P.; Carter, W.B. Tumor-induced upregulation of Twist, Snail, and Slug represses the activity of the human VE-cadherin promoter. Arch. Biochem. Biophys. 2009, 482, 77–82. [Google Scholar] [CrossRef]
- Kurahara, H.; Takao, S.; Maemura, K.; Mataki, Y.; Kuwahata, T.; Maeda, K.; Ding, Q.; Sakoda, M.; Iino, S.; Ishigami, S.; et al. Epithelial-mesenchymal transition and mesenchymal-epithelial transition via regulation of ZEB-1 and ZEB-2 expression in pancreatic cancer. J. Surg. Oncol. 2012, 105, 655–661. [Google Scholar] [CrossRef]
- Lee, J.; Jung, E.; Gestoso, K.; Heur, M. ZEB1 Mediates Fibrosis in Corneal Endothelial Mesenchymal Transition Through SP1 and SP3. Investig. Ophthalmol. Vis. Sci. 2020, 61, 41. [Google Scholar] [CrossRef] [PubMed]
- Franco, H.L.; Casasnovas, J.; Rodriguez-Medina, J.R.; Cadilla, C.L. Redundant or separate entities?—roles of Twist1 and Twist2 as molecular switches during gene transcription. Nucleic Acids Res. 2011, 39, 1177–1186. [Google Scholar] [CrossRef] [PubMed]
- Mammoto, T.; Muyleart, M.; Konduri, G.G.; Mammoto, A. Twist1 in Hypoxia-induced Pulmonary Hypertension through Transforming Growth Factor-beta-Smad Signaling. Am. J. Respir. Cell Mol. Biol. 2018, 58, 194–207. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Niu, W.; Dong, H.Y.; Liu, M.L.; Luo, Y.; Li, Z.C. Hypoxia induces endothelial-mesenchymal transition in pulmonary vascular remodeling. Int. J. Mol. Med. 2018, 42, 270–278. [Google Scholar] [CrossRef]
- Ning, X.; Zhang, K.; Wu, Q.; Liu, M.; Sun, S. Emerging role of Twist1 in fibrotic diseases. J. Cell Mol. Med. 2018, 22, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
- Palumbo-Zerr, K.; Soare, A.; Zerr, P.; Liebl, A.; Mancuso, R.; Tomcik, M.; Sumova, B.; Dees, C.; Chen, C.W.; Wohlfahrt, T.; et al. Composition of TWIST1 dimers regulates fibroblast activation and tissue fibrosis. Ann. Rheum. Dis. 2017, 76, 244–251. [Google Scholar] [CrossRef]
- Ciszewski, W.M.; Sobierajska, K.; Wawro, M.E.; Klopocka, W.; Chefczynska, N.; Muzyczuk, A.; Siekacz, K.; Wujkowska, A.; Niewiarowska, J. The ILK-MMP9-MRTF axis is crucial for EndMT differentiation of endothelial cells in a tumor microenvironment. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2283–2296. [Google Scholar] [CrossRef]
- Dufton, N.P.; Peghaire, C.R.; Osuna-Almagro, L.; Raimondi, C.; Kalna, V.; Chauhan, A.; Webb, G.; Yang, Y.; Birdsey, G.M.; Lalor, P.; et al. Dynamic regulation of canonical TGFbeta signalling by endothelial transcription factor ERG protects from liver fibrogenesis. Nat. Commun. 2017, 8, 895. [Google Scholar] [CrossRef]
- Mihira, H.; Suzuki, H.I.; Akatsu, Y.; Yoshimatsu, Y.; Igarashi, T.; Miyazono, K.; Watabe, T. TGF-beta-induced mesenchymal transition of MS-1 endothelial cells requires Smad-dependent cooperative activation of Rho signals and MRTF-A. J. Biochem. 2012, 151, 145–156. [Google Scholar] [CrossRef]
- Yao, J.; Wu, X.; Zhang, D.; Wang, L.; Zhang, L.; Reynolds, E.X.; Hernandez, C.; Bostrom, K.I.; Yao, Y. Elevated endothelial Sox2 causes lumen disruption and cerebral arteriovenous malformations. J. Clin. Investig. 2019, 129, 3121–3133. [Google Scholar] [CrossRef]
- Ghosh, A.K.; Nagpal, V.; Covington, J.W.; Michaels, M.A.; Vaughan, D.E. Molecular basis of cardiac endothelial-to-mesenchymal transition (EndMT): Differential expression of microRNAs during EndMT. Cell Signal 2012, 24, 1031–1036. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Li, P.; Bledsoe, G.; Yang, Z.R.; Chao, L.; Chao, J. Kallistatin inhibits TGF-beta-induced endothelial-mesenchymal transition by differential regulation of microRNA-21 and eNOS expression. Exp. Cell Res. 2015, 337, 103–110. [Google Scholar] [CrossRef] [PubMed]
- Katsura, A.; Suzuki, H.I.; Ueno, T.; Mihira, H.; Yamazaki, T.; Yasuda, T.; Watabe, T.; Mano, H.; Yamada, Y.; Miyazono, K. MicroRNA-31 is a positive modulator of endothelial-mesenchymal transition and associated secretory phenotype induced by TGF-beta. Genes. Cells 2016, 21, 99–116. [Google Scholar] [CrossRef] [PubMed]
- Lagendijk, A.K.; Goumans, M.J.; Burkhard, S.B.; Bakkers, J. MicroRNA-23 restricts cardiac valve formation by inhibiting Has2 and extracellular hyaluronic acid production. Circ. Res. 2011, 109, 649–657. [Google Scholar] [CrossRef] [PubMed]
- Kumarswamy, R.; Volkmann, I.; Jazbutyte, V.; Dangwal, S.; Park, D.H.; Thum, T. Transforming growth factor-beta-induced endothelial-to-mesenchymal transition is partly mediated by microRNA-21. Arter. Thromb. Vasc. Biol. 2012, 32, 361–369. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, H.I.; Katsura, A.; Mihira, H.; Horie, M.; Saito, A.; Miyazono, K. Regulation of TGF-beta-mediated endothelial-mesenchymal transition by microRNA-27. J. Biochem. 2017, 161, 417–420. [Google Scholar] [CrossRef] [PubMed]
- Bai, Y.; Zhang, Y.; Han, B.; Yang, L.; Chen, X.; Huang, R.; Wu, F.; Chao, J.; Liu, P.; Hu, G.; et al. Circular RNA DLGAP4 Ameliorates Ischemic Stroke Outcomes by Targeting miR-143 to Regulate Endothelial-Mesenchymal Transition Associated with Blood-Brain Barrier Integrity. J. Neurosci. 2018, 38, 32–50. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.P.; He, Q.; Shen, Z.; Shu, X.L.; Wang, C.H.; Zhu, J.J.; Shi, L.P.; Du, L.Z. MiR-126a-5p is involved in the hypoxia-induced endothelial-to-mesenchymal transition of neonatal pulmonary hypertension. Hypertens. Res. 2017, 40, 552–561. [Google Scholar] [CrossRef] [PubMed]
- Bijkerk, R.; de Bruin, R.G.; van Solingen, C.; Duijs, J.M.; Kobayashi, K.; van der Veer, E.P.; ten Dijke, P.; Rabelink, T.J.; Goumans, M.J.; van Zonneveld, A.J. MicroRNA-155 functions as a negative regulator of RhoA signaling in TGF-beta-induced endothelial to mesenchymal transition. Microrna 2012, 1, 2–10. [Google Scholar] [CrossRef]
- Geng, H.; Guan, J. MiR-18a-5p inhibits endothelial-mesenchymal transition and cardiac fibrosis through the Notch2 pathway. Biochem. Biophys. Res. Commun. 2017, 491, 329–336. [Google Scholar] [CrossRef]
- Miscianinov, V.; Martello, A.; Rose, L.; Parish, E.; Cathcart, B.; Mitic, T.; Gray, G.A.; Meloni, M.; Al Haj Zen, A.; Caporali, A. MicroRNA-148b Targets the TGF-beta Pathway to Regulate Angiogenesis and Endothelial-to-Mesenchymal Transition during Skin Wound Healing. Mol. Ther. 2018, 26, 1996–2007. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Hu, J.; Liu, L. MiR-200a modulates TGF-beta1-induced endothelial-to-mesenchymal shift via suppression of GRB2 in HAECs. Biomed. Pharmacother. 2017, 95, 215–222. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Zhang, Z.; Zhang, D.Y.; Zhu, J.; Zhang, T.; Wang, C. microRNA 126 inhibits the transition of endothelial progenitor cells to mesenchymal cells via the PIK3R2-PI3K/Akt signalling pathway. PLoS ONE 2013, 8, e83294. [Google Scholar] [CrossRef] [PubMed]
- Fang, S.; Guo, H.; Cheng, Y.; Zhou, Z.; Zhang, W.; Han, B.; Luo, W.; Wang, J.; Xie, W.; Chao, J. circHECTD1 promotes the silica-induced pulmonary endothelial-mesenchymal transition via HECTD1. Cell Death Dis. 2018, 9, 396. [Google Scholar] [CrossRef]
- Huang, X.; Chen, Y.; Xiao, J.; Huang, Z.; He, L.; Xu, D.; Peng, J. Identification of differentially expressed circular RNAs during TGF-ss1-induced endothelial-to-mesenchymal transition in rat coronary artery endothelial cells. Anatol. J. Cardiol. 2018, 19, 192–197. [Google Scholar] [CrossRef] [PubMed]
- Neumann, P.; Jae, N.; Knau, A.; Glaser, S.F.; Fouani, Y.; Rossbach, O.; Kruger, M.; John, D.; Bindereif, A.; Grote, P.; et al. The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2. Nat. Commun. 2018, 9, 237. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Han, B.; Zhang, Y.; Bai, Y.; Chao, J.; Hu, G.; Yao, H. Engagement of circular RNA HECW2 in the nonautophagic role of ATG5 implicated in the endothelial-mesenchymal transition. Autophagy 2018, 14, 404–418. [Google Scholar] [CrossRef] [PubMed]
- Goumans, M.J.; van Zonneveld, A.J.; ten Dijke, P. Transforming growth factor beta-induced endothelial-to-mesenchymal transition: A switch to cardiac fibrosis? Trends Cardiovasc. Med. 2008, 18, 293–298. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Sanchez-Duffhues, G.; Goumans, M.J.; Ten Dijke, P. TGF-beta-Induced Endothelial to Mesenchymal Transition in Disease and Tissue Engineering. Front. Cell Dev. Biol. 2020, 8, 260. [Google Scholar] [CrossRef]
- Markwald, R.R.; Fitzharris, T.P.; Manasek, F.J. Structural development of endocardial cushions. Am. J. Anat. 1977, 148, 85–119. [Google Scholar] [CrossRef]
- van Meeteren, L.A.; ten Dijke, P. Regulation of endothelial cell plasticity by TGF-beta. Cell Tissue Res. 2012, 347, 177–186. [Google Scholar] [CrossRef] [PubMed]
- Frid, M.G.; Kale, V.A.; Stenmark, K.R. Mature vascular endothelium can give rise to smooth muscle cells via endothelial-mesenchymal transdifferentiation: In vitro analysis. Circ. Res. 2002, 90, 1189–1196. [Google Scholar] [CrossRef]
- Paruchuri, S.; Yang, J.H.; Aikawa, E.; Melero-Martin, J.M.; Khan, Z.A.; Loukogeorgakis, S.; Schoen, F.J.; Bischoff, J. Human pulmonary valve progenitor cells exhibit endothelial/mesenchymal plasticity in response to vascular endothelial growth factor-A and transforming growth factor-beta2. Circ. Res. 2006, 99, 861–869. [Google Scholar] [CrossRef]
- Camenisch, T.D.; Molin, D.G.; Person, A.; Runyan, R.B.; Gittenberger-de Groot, A.C.; McDonald, J.A.; Klewer, S.E. Temporal and distinct TGFbeta ligand requirements during mouse and avian endocardial cushion morphogenesis. Dev. Biol. 2002, 248, 170–181. [Google Scholar] [CrossRef]
- Sabbineni, H.; Verma, A.; Somanath, P.R. Isoform-specific effects of transforming growth factor beta on endothelial-to-mesenchymal transition. J. Cell Physiol. 2018, 233, 8418–8428. [Google Scholar] [CrossRef]
- Azhar, M.; Runyan, R.B.; Gard, C.; Sanford, L.P.; Miller, M.L.; Andringa, A.; Pawlowski, S.; Rajan, S.; Doetschman, T. Ligand-specific function of transforming growth factor beta in epithelial-mesenchymal transition in heart development. Dev. Dyn. 2009, 238, 431–442. [Google Scholar] [CrossRef] [PubMed]
- Brown, C.B.; Boyer, A.S.; Runyan, R.B.; Barnett, J.V. Requirement of type III TGF-beta receptor for endocardial cell transformation in the heart. Science 1999, 283, 2080–2082. [Google Scholar] [CrossRef]
- Romano, L.A.; Runyan, R.B. Slug is an essential target of TGFbeta2 signaling in the developing chicken heart. Dev. Biol. 2000, 223, 91–102. [Google Scholar] [CrossRef] [PubMed]
- Medici, D.; Shore, E.M.; Lounev, V.Y.; Kaplan, F.S.; Kalluri, R.; Olsen, B.R. Conversion of vascular endothelial cells into multipotent stem-like cells. Nat. Med. 2010, 16, 1400–1406. [Google Scholar] [CrossRef]
- Ma, L.; Lu, M.F.; Schwartz, R.J.; Martin, J.F. Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterning. Development 2005, 132, 5601–5611. [Google Scholar] [CrossRef]
- Kaneko, K.; Li, X.; Zhang, X.; Lamberti, J.J.; Jamieson, S.W.; Thistlethwaite, P.A. Endothelial expression of bone morphogenetic protein receptor type 1a is required for atrioventricular valve formation. Ann. Thorac. Surg. 2008, 85, 2090–2098. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Sridurongrit, S.; Dudas, M.; Thomas, P.; Nagy, A.; Schneider, M.D.; Epstein, J.A.; Kaartinen, V. Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heart. Dev. Biol. 2005, 286, 299–310. [Google Scholar] [CrossRef] [PubMed]
- Budi, E.H.; Mamai, O.; Hoffman, S.; Akhurst, R.J.; Derynck, R. Enhanced TGF-beta Signaling Contributes to the Insulin-Induced Angiogenic Responses of Endothelial Cells. iScience 2019, 11, 474–491. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.Y.; Qin, L.; Li, G.; Wang, Z.; Dahlman, J.E.; Malagon-Lopez, J.; Gujja, S.; Cilfone, N.A.; Kauffman, K.J.; Sun, L.; et al. Endothelial TGF-beta signalling drives vascular inflammation and atherosclerosis. Nat. Metab. 2019, 1, 912–926. [Google Scholar] [CrossRef]
- Madri, J.A.; Pratt, B.M.; Tucker, A.M. Phenotypic modulation of endothelial cells by transforming growth factor-beta depends upon the composition and organization of the extracellular matrix. J. Cell Biol. 1988, 106, 1375–1384. [Google Scholar] [CrossRef]
- Heldin, C.H.; Miyazono, K.; ten Dijke, P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997, 390, 465–471. [Google Scholar] [CrossRef]
- Heldin, C.H.; Moustakas, A. Role of Smads in TGFbeta signaling. Cell Tissue Res. 2012, 347, 21–36. [Google Scholar] [CrossRef]
- Fu, Y.; Chang, A.; Chang, L.; Niessen, K.; Eapen, S.; Setiadi, A.; Karsan, A. Differential regulation of transforming growth factor beta signaling pathways by Notch in human endothelial cells. J. Biol. Chem. 2009, 284, 19452–19462. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Hu, Z.F.; Liao, H.H.; Liu, W.; Liu, J.; Ma, Z.G.; Wu, Q.Q.; Xu, M.; Zhang, N.; Zhang, Y.; et al. Toll-like receptor 5 deficiency attenuates interstitial cardiac fibrosis and dysfunction induced by pressure overload by inhibiting inflammation and the endothelial-mesenchymal transition. Biochim. Biophys. Acta 2015, 1852, 2456–2466. [Google Scholar] [CrossRef]
- Imamura, T.; Takase, M.; Nishihara, A.; Oeda, E.; Hanai, J.; Kawabata, M.; Miyazono, K. Smad6 inhibits signalling by the TGF-beta superfamily. Nature 1997, 389, 622–626. [Google Scholar] [CrossRef]
- Nakao, A.; Afrakhte, M.; Moren, A.; Nakayama, T.; Christian, J.L.; Heuchel, R.; Itoh, S.; Kawabata, M.; Heldin, N.E.; Heldin, C.H.; et al. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature 1997, 389, 631–635. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Wang, F.; Zha, S.; Cao, Q.; Sheng, J.; Chen, S. SIRT1 inhibits TGF-beta-induced endothelial-mesenchymal transition in human endothelial cells with Smad4 deacetylation. J. Cell Physiol. 2018, 233, 9007–9014. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.R.; Zheng, Y.J.; Zhang, Z.B.; Shen, W.L.; Li, X.D.; Wei, T.; Ruan, C.C.; Chen, X.H.; Zhu, D.L.; Gao, P.J. Suppression of Endothelial-to-Mesenchymal Transition by SIRT (Sirtuin) 3 Alleviated the Development of Hypertensive Renal Injury. Hypertension 2018, 72, 350–360. [Google Scholar] [CrossRef] [PubMed]
- Stawski, L.; Marden, G.; Trojanowska, M. The Activation of Human Dermal Microvascular Cells by Poly(I:C), Lipopolysaccharide, Imiquimod, and ODN2395 Is Mediated by the Fli1/FOXO3A Pathway. J. Immunol. 2018, 200, 248–259. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Jimenez, S.A. Protein kinase Cdelta and c-Abl kinase are required for transforming growth factor beta induction of endothelial-mesenchymal transition in vitro. Arthritis Rheum. 2011, 63, 2473–2483. [Google Scholar] [CrossRef] [PubMed]
- Milara, J.; Ballester, B.; Morell, A.; Ortiz, J.L.; Escriva, J.; Fernandez, E.; Perez-Vizcaino, F.; Cogolludo, A.; Pastor, E.; Artigues, E.; et al. JAK2 mediates lung fibrosis, pulmonary vascular remodelling and hypertension in idiopathic pulmonary fibrosis: An experimental study. Thorax 2018, 73, 519–529. [Google Scholar] [CrossRef] [PubMed]
- Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.E. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009, 19, 128–139. [Google Scholar] [CrossRef]
- Zhang, Y.E. Non-Smad Signaling Pathways of the TGF-beta Family. Cold Spring Harb. Perspect. Biol. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
- Noseda, M.; McLean, G.; Niessen, K.; Chang, L.; Pollet, I.; Montpetit, R.; Shahidi, R.; Dorovini-Zis, K.; Li, L.; Beckstead, B.; et al. Notch activation results in phenotypic and functional changes consistent with endothelial-to-mesenchymal transformation. Circ. Res. 2004, 94, 910–917. [Google Scholar] [CrossRef]
- Liu, J.; Dong, F.; Jeong, J.; Masuda, T.; Lobe, C.G. Constitutively active Notch1 signaling promotes endothelial-mesenchymal transition in a conditional transgenic mouse model. Int. J. Mol. Med. 2014, 34, 669–676. [Google Scholar] [CrossRef] [PubMed]
- Fu, Y.; Chang, A.C.; Fournier, M.; Chang, L.; Niessen, K.; Karsan, A. RUNX3 maintains the mesenchymal phenotype after termination of the Notch signal. J. Biol. Chem. 2011, 286, 11803–11813. [Google Scholar] [CrossRef]
- Blokzijl, A.; Dahlqvist, C.; Reissmann, E.; Falk, A.; Moliner, A.; Lendahl, U.; Ibanez, C.F. Cross-talk between the Notch and TGF-beta signaling pathways mediated by interaction of the Notch intracellular domain with Smad3. J. Cell Biol. 2003, 163, 723–728. [Google Scholar] [CrossRef]
- Niessen, K.; Fu, Y.; Chang, L.; Hoodless, P.A.; McFadden, D.; Karsan, A. Slug is a direct Notch target required for initiation of cardiac cushion cellularization. J. Cell Biol. 2008, 182, 315–325. [Google Scholar] [CrossRef] [PubMed]
- Guan, S.; Zhou, J. CXCR7 attenuates the TGF-beta-induced endothelial-to-mesenchymal transition and pulmonary fibrosis. Mol. Biosyst. 2017, 13, 2116–2124. [Google Scholar] [CrossRef]
- Chang, A.C.; Garside, V.C.; Fournier, M.; Smrz, J.; Vrljicak, P.; Umlandt, P.; Fuller, M.; Robertson, G.; Zhao, Y.; Tam, A.; et al. A Notch-dependent transcriptional hierarchy promotes mesenchymal transdifferentiation in the cardiac cushion. Dev. Dyn. 2014, 243, 894–905. [Google Scholar] [CrossRef]
- Chang, A.C.; Fu, Y.; Garside, V.C.; Niessen, K.; Chang, L.; Fuller, M.; Setiadi, A.; Smrz, J.; Kyle, A.; Minchinton, A.; et al. Notch initiates the endothelial-to-mesenchymal transition in the atrioventricular canal through autocrine activation of soluble guanylyl cyclase. Dev. Cell 2011, 21, 288–300. [Google Scholar] [CrossRef] [PubMed]
- Yin, Q.; Wang, W.; Cui, G.; Nan, H.; Yan, L.; Zhang, W.; Zhang, S.; Wei, J. The expression levels of Notch-related signaling molecules in pulmonary microvascular endothelial cells in bleomycin-induced rat pulmonary fibrosis. Physiol. Res. 2017, 66, 305–315. [Google Scholar] [CrossRef]
- Patel, J.; Baz, B.; Wong, H.Y.; Lee, J.S.; Khosrotehrani, K. Accelerated Endothelial to Mesenchymal Transition Increased Fibrosis via Deleting Notch Signaling in Wound Vasculature. J. Investig. Dermatol. 2018, 138, 1166–1175. [Google Scholar] [CrossRef]
- Cadigan, K.M.; Nusse, R. Wnt signaling: A common theme in animal development. Genes. Dev. 1997, 11, 3286–3305. [Google Scholar] [CrossRef]
- He, X.; Semenov, M.; Tamai, K.; Zeng, X. LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: Arrows point the way. Development 2004, 131, 1663–1677. [Google Scholar] [CrossRef] [PubMed]
- Komiya, Y.; Habas, R. Wnt signal transduction pathways. Organogenesis 2008, 4, 68–75. [Google Scholar] [CrossRef] [PubMed]
- Nusse, R. Wnt signaling in disease and in development. Cell Res. 2005, 15, 28–32. [Google Scholar] [CrossRef] [PubMed]
- Pfister, A.S.; Kuhl, M. Of Wnts and Ribosomes. Prog. Mol. Biol. Transl. Sci. 2018, 153, 131–155. [Google Scholar] [CrossRef] [PubMed]
- Habas, R.; Dawid, I.B. Dishevelled and Wnt signaling: Is the nucleus the final frontier? J. Biol. 2005, 4, 2. [Google Scholar] [CrossRef] [PubMed]
- Aisagbonhi, O.; Rai, M.; Ryzhov, S.; Atria, N.; Feoktistov, I.; Hatzopoulos, A.K. Experimental myocardial infarction triggers canonical Wnt signaling and endothelial-to-mesenchymal transition. Dis. Model. Mech. 2011, 4, 469–483. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, C.; Distler, J.H. Canonical Wnt signaling in systemic sclerosis. Lab. Investig. 2016, 96, 151–155. [Google Scholar] [CrossRef]
- Foulquier, S.; Daskalopoulos, E.P.; Lluri, G.; Hermans, K.C.M.; Deb, A.; Blankesteijn, W.M. WNT Signaling in Cardiac and Vascular Disease. Pharmacol. Rev. 2018, 70, 68–141. [Google Scholar] [CrossRef] [PubMed]
- Niehrs, C. The complex world of WNT receptor signalling. Nat. Rev. Mol. Cell Biol. 2012, 13, 767–779. [Google Scholar] [CrossRef]
- Nusse, R.; Clevers, H. Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef]
- Li, L.; Chen, L.; Zang, J.; Tang, X.; Liu, Y.; Zhang, J.; Bai, L.; Yin, Q.; Lu, Y.; Cheng, J.; et al. C3a and C5a receptor antagonists ameliorate endothelial-myofibroblast transition via the Wnt/beta-catenin signaling pathway in diabetic kidney disease. Metabolism 2015, 64, 597–610. [Google Scholar] [CrossRef] [PubMed]
- Alvandi, Z.; Nagata, Y.; Passos, L.S.A.; Hashemi Gheinani, A.; Guerrero, J.L.; Wylie-Sears, J.; Romero, D.C.; Morris, B.A.; Sullivan, S.M.; Yaghoubian, K.M.; et al. Wnt Site Signaling Inhibitor Secreted Frizzled-Related Protein 3 Protects Mitral Valve Endothelium From Myocardial Infarction-Induced Endothelial-to-Mesenchymal Transition. J. Am. Heart Assoc. 2022, 11, e023695. [Google Scholar] [CrossRef] [PubMed]
- Hall, I.F.; Kishta, F.; Xu, Y.; Baker, A.H.; Kovacic, J.C. Endothelial to mesenchymal transition: At the axis of cardiovascular health and disease. Cardiovasc. Res. 2024, 120, 223–236. [Google Scholar] [CrossRef]
- Lipphardt, M.; Dihazi, H.; Jeon, N.L.; Dadafarin, S.; Ratliff, B.B.; Rowe, D.W.; Muller, G.A.; Goligorsky, M.S. Dickkopf-3 in aberrant endothelial secretome triggers renal fibroblast activation and endothelial-mesenchymal transition. Nephrol Dial Transpl. 2019, 34, 49–62. [Google Scholar] [CrossRef] [PubMed]
- Cheng, S.L.; Shao, J.S.; Behrmann, A.; Krchma, K.; Towler, D.A. Dkk1 and MSX2-Wnt7b signaling reciprocally regulate the endothelial-mesenchymal transition in aortic endothelial cells. Arter. Thromb. Vasc. Biol. 2013, 33, 1679–1689. [Google Scholar] [CrossRef] [PubMed]
- Chester, A.H.; Yacoub, M.H. The role of endothelin-1 in pulmonary arterial hypertension. Glob Cardiol Sci Pr. 2014, 2014, 62–78. [Google Scholar] [CrossRef] [PubMed]
- Kawanabe, Y.; Nauli, S.M. Endothelin. Cell Mol. Life Sci. 2011, 68, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Masaki, T. The discovery, the present state, and the future prospects of endothelin. J. Cardiovasc. Pharmacol. 1989; 13, (Suppl. S5), S1–S4; discussion S18. [Google Scholar] [CrossRef]
- Shao, D.; Park, J.E.; Wort, S.J. The role of endothelin-1 in the pathogenesis of pulmonary arterial hypertension. Pharmacol. Res. 2011, 63, 504–511. [Google Scholar] [CrossRef] [PubMed]
- Abraham, D.; Ponticos, M.; Nagase, H. Connective tissue remodeling: Cross-talk between endothelins and matrix metalloproteinases. Curr. Vasc. Pharmacol. 2005, 3, 369–379. [Google Scholar] [CrossRef]
- Jain, R.; Shaul, P.W.; Borok, Z.; Willis, B.C. Endothelin-1 induces alveolar epithelial-mesenchymal transition through endothelin type A receptor-mediated production of TGF-beta1. Am. J. Respir. Cell Mol. Biol. 2007, 37, 38–47. [Google Scholar] [CrossRef]
- Recchia, A.G.; Filice, E.; Pellegrino, D.; Dobrina, A.; Cerra, M.C.; Maggiolini, M. Endothelin-1 induces connective tissue growth factor expression in cardiomyocytes. J. Mol. Cell Cardiol. 2009, 46, 352–359. [Google Scholar] [CrossRef]
- Rodriguez-Pascual, F.; Busnadiego, O.; Gonzalez-Santamaria, J. The profibrotic role of endothelin-1: Is the door still open for the treatment of fibrotic diseases? Life Sci. 2014, 118, 156–164. [Google Scholar] [CrossRef]
- Shi-Wen, X.; Chen, Y.; Denton, C.P.; Eastwood, M.; Renzoni, E.A.; Bou-Gharios, G.; Pearson, J.D.; Dashwood, M.; du Bois, R.M.; Black, C.M.; et al. Endothelin-1 promotes myofibroblast induction through the ETA receptor via a rac/phosphoinositide 3-kinase/Akt-dependent pathway and is essential for the enhanced contractile phenotype of fibrotic fibroblasts. Mol. Biol. Cell 2004, 15, 2707–2719. [Google Scholar] [CrossRef] [PubMed]
- Shi-Wen, X.; Renzoni, E.A.; Kennedy, L.; Howat, S.; Chen, Y.; Pearson, J.D.; Bou-Gharios, G.; Dashwood, M.R.; du Bois, R.M.; Black, C.M.; et al. Endogenous endothelin-1 signaling contributes to type I collagen and CCN2 overexpression in fibrotic fibroblasts. Matrix Biol. 2007, 26, 625–632. [Google Scholar] [CrossRef] [PubMed]
- Simonson, M.S.; Ismail-Beigi, F. Endothelin-1 increases collagen accumulation in renal mesangial cells by stimulating a chemokine and cytokine autocrine signaling loop. J. Biol. Chem. 2011, 286, 11003–11008. [Google Scholar] [CrossRef] [PubMed]
- Swigris, J.J.; Brown, K.K. The role of endothelin-1 in the pathogenesis of idiopathic pulmonary fibrosis. BioDrugs 2010, 24, 49–54. [Google Scholar] [CrossRef] [PubMed]
- Weng, C.M.; Yu, C.C.; Kuo, M.L.; Chen, B.C.; Lin, C.H. Endothelin-1 induces connective tissue growth factor expression in human lung fibroblasts by ETAR-dependent JNK/AP-1 pathway. Biochem. Pharmacol. 2014, 88, 402–411. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.W.; Howat, S.L.; Renzoni, E.A.; Holmes, A.; Pearson, J.D.; Dashwood, M.R.; Bou-Gharios, G.; Denton, C.P.; du Bois, R.M.; Black, C.M.; et al. Endothelin-1 induces expression of matrix-associated genes in lung fibroblasts through MEK/ERK. J. Biol. Chem. 2004, 279, 23098–23103. [Google Scholar] [CrossRef]
- Widyantoro, B.; Emoto, N.; Nakayama, K.; Anggrahini, D.W.; Adiarto, S.; Iwasa, N.; Yagi, K.; Miyagawa, K.; Rikitake, Y.; Suzuki, T.; et al. Endothelial cell-derived endothelin-1 promotes cardiac fibrosis in diabetic hearts through stimulation of endothelial-to-mesenchymal transition. Circulation 2010, 121, 2407–2418. [Google Scholar] [CrossRef]
- Cipriani, P.; Di Benedetto, P.; Ruscitti, P.; Capece, D.; Zazzeroni, F.; Liakouli, V.; Pantano, I.; Berardicurti, O.; Carubbi, F.; Pecetti, G.; et al. The Endothelial-mesenchymal Transition in Systemic Sclerosis Is Induced by Endothelin-1 and Transforming Growth Factor-beta and May Be Blocked by Macitentan, a Dual Endothelin-1 Receptor Antagonist. J. Rheumatol. 2015, 42, 1808–1816. [Google Scholar] [CrossRef]
- Wermuth, P.J.; Li, Z.; Mendoza, F.A.; Jimenez, S.A. Stimulation of Transforming Growth Factor-beta1-Induced Endothelial-To-Mesenchymal Transition and Tissue Fibrosis by Endothelin-1 (ET-1): A Novel Profibrotic Effect of ET-1. PLoS ONE 2016, 11, e0161988. [Google Scholar] [CrossRef] [PubMed]
- Hojo, H.; Takei, T.; Asahina, Y.; Okumura, N.; Takao, T.; So, M.; Suetake, I.; Sato, T.; Kawamoto, A.; Hirabayashi, Y. Total Synthesis and Structural Characterization of Caveolin-1. Angew. Chem. Int. Ed. Engl. 2021, 60, 13900–13905. [Google Scholar] [CrossRef]
- Shu, Y.; Jin, S. Caveolin-1 in endothelial cells: A potential therapeutic target for atherosclerosis. Heliyon 2023, 9, e18653. [Google Scholar] [CrossRef] [PubMed]
- Del Galdo, F.; Lisanti, M.P.; Jimenez, S.A. Caveolin-1, transforming growth factor-beta receptor internalization, and the pathogenesis of systemic sclerosis. Curr. Opin. Rheumatol. 2008, 20, 713–719. [Google Scholar] [CrossRef] [PubMed]
- Gvaramia, D.; Blaauboer, M.E.; Hanemaaijer, R.; Everts, V. Role of caveolin-1 in fibrotic diseases. Matrix Biol. 2013, 32, 307–315. [Google Scholar] [CrossRef] [PubMed]
- Razani, B.; Zhang, X.L.; Bitzer, M.; von Gersdorff, G.; Bottinger, E.P.; Lisanti, M.P. Caveolin-1 regulates transforming growth factor (TGF)-beta/SMAD signaling through an interaction with the TGF-beta type I receptor. J. Biol. Chem. 2001, 276, 6727–6738. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Wermuth, P.J.; Benn, B.S.; Lisanti, M.P.; Jimenez, S.A. Caveolin-1 deficiency induces spontaneous endothelial-to-mesenchymal transition in murine pulmonary endothelial cells in vitro. Am. J. Pathol. 2013, 182, 325–331. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Pan, L.; Pu, H.; Wang, Y.; Zhang, X.; Li, C.; Yang, Z. Loss of caveolin-1 promotes endothelial-mesenchymal transition during sepsis: A membrane proteomic study. Int. J. Mol. Med. 2013, 32, 585–592. [Google Scholar] [CrossRef] [PubMed]
- Dejana, E.; Hirschi, K.K.; Simons, M. The molecular basis of endothelial cell plasticity. Nat. Commun. 2017, 8, 14361. [Google Scholar] [CrossRef]
- Fang, J.S.; Hultgren, N.W.; Hughes, C.C.W. Regulation of Partial and Reversible Endothelial-to-Mesenchymal Transition in Angiogenesis. Front. Cell Dev. Biol. 2021, 9, 702021. [Google Scholar] [CrossRef] [PubMed]
- Bravi, L.; Malinverno, M.; Pisati, F.; Rudini, N.; Cuttano, R.; Pallini, R.; Martini, M.; Larocca, L.M.; Locatelli, M.; Levi, V.; et al. Endothelial Cells Lining Sporadic Cerebral Cavernous Malformation Cavernomas Undergo Endothelial-to-Mesenchymal Transition. Stroke 2016, 47, 886–890. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.Y.; Qin, L.; Barnes, C.; Charisse, K.; Yi, T.; Zhang, X.; Ali, R.; Medina, P.P.; Yu, J.; Slack, F.J.; et al. FGF regulates TGF-beta signaling and endothelial-to-mesenchymal transition via control of let-7 miRNA expression. Cell Rep. 2012, 2, 1684–1696. [Google Scholar] [CrossRef] [PubMed]
- Malinverno, M.; Maderna, C.; Abu Taha, A.; Corada, M.; Orsenigo, F.; Valentino, M.; Pisati, F.; Fusco, C.; Graziano, P.; Giannotta, M.; et al. Endothelial cell clonal expansion in the development of cerebral cavernous malformations. Nat. Commun. 2019, 10, 2761. [Google Scholar] [CrossRef] [PubMed]
- Medici, D.; Kalluri, R. Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype. Semin. Cancer Biol. 2012, 22, 379–384. [Google Scholar] [CrossRef] [PubMed]
- Shoemaker, L.D.; McCormick, A.K.; Allen, B.M.; Chang, S.D. Evidence for endothelial-to-mesenchymal transition in human brain arteriovenous malformations. Clin. Transl. Med. 2020, 10, e99. [Google Scholar] [CrossRef] [PubMed]
- He, Q.; Huo, R.; Wang, J.; Xu, H.; Zhao, S.; Zhang, J.; Sun, Y.; Jiao, Y.; Weng, J.; Zhao, J.; et al. Exosomal miR-3131 derived from endothelial cells with KRAS mutation promotes EndMT by targeting PICK1 in brain arteriovenous malformations. CNS Neurosci. Ther. 2023, 29, 1312–1324. [Google Scholar] [CrossRef] [PubMed]
- Jackson, A.O.; Zhang, J.; Jiang, Z.; Yin, K. Endothelial-to-mesenchymal transition: A novel therapeutic target for cardiovascular diseases. Trends Cardiovasc. Med. 2017, 27, 383–393. [Google Scholar] [CrossRef] [PubMed]
- Gao, Q.; Guan, L.; Hu, S.; Yao, Y.; Ren, X.; Zhang, Z.; Cheng, C.; Liu, Y.; Zhang, C.; Huang, J.; et al. Study on the mechanism of HIF1a-SOX9 in glucose-induced cardiomyocyte hypertrophy. Biomed. Pharmacother. 2015, 74, 57–62. [Google Scholar] [CrossRef] [PubMed]
- Jin, Y.G.; Yuan, Y.; Wu, Q.Q.; Zhang, N.; Fan, D.; Che, Y.; Wang, Z.P.; Xiao, Y.; Wang, S.S.; Tang, Q.Z. Puerarin Protects against Cardiac Fibrosis Associated with the Inhibition of TGF-beta1/Smad2-Mediated Endothelial-to-Mesenchymal Transition. PPAR Res. 2017, 2017, 2647129. [Google Scholar] [CrossRef]
- Wei, W.Y.; Zhang, N.; Li, L.L.; Ma, Z.G.; Xu, M.; Yuan, Y.P.; Deng, W.; Tang, Q.Z. Pioglitazone Alleviates Cardiac Fibrosis and Inhibits Endothelial to Mesenchymal Transition Induced by Pressure Overload. Cell Physiol. Biochem. 2018, 45, 26–36. [Google Scholar] [CrossRef]
- Wu, Q.Q.; Xiao, Y.; Jiang, X.H.; Yuan, Y.; Yang, Z.; Chang, W.; Bian, Z.Y.; Tang, Q.Z. Evodiamine attenuates TGF-beta1-induced fibroblast activation and endothelial to mesenchymal transition. Mol. Cell Biochem. 2017, 430, 81–90. [Google Scholar] [CrossRef] [PubMed]
- Zou, J.; Liu, Y.; Li, B.; Zheng, Z.; Ke, X.; Hao, Y.; Li, X.; Li, X.; Liu, F.; Zhang, Z. Autophagy attenuates endothelial-to-mesenchymal transition by promoting Snail degradation in human cardiac microvascular endothelial cells. Biosci. Rep. 2017, 37, BSR20171049. [Google Scholar] [CrossRef] [PubMed]
- Cai, J.; Chen, X.; Chen, X.; Chen, L.; Zheng, G.; Zhou, H.; Zhou, X. Anti-Fibrosis Effect of Relaxin and Spironolactone Combined on Isoprenaline-Induced Myocardial Fibrosis in Rats via Inhibition of Endothelial-Mesenchymal Transition. Cell Physiol. Biochem. 2017, 41, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Cai, J.; Zhou, X.; Chen, L.; Gong, Y.; Gao, Z.; Zhang, H.; Huang, W.; Zhou, H. Protective Effect of Spironolactone on Endothelial-to-Mesenchymal Transition in HUVECs via Notch Pathway. Cell Physiol. Biochem. 2015, 36, 191–200. [Google Scholar] [CrossRef] [PubMed]
- Zhou, H.; Chen, X.; Chen, L.; Zhou, X.; Zheng, G.; Zhang, H.; Huang, W.; Cai, J. Anti-fibrosis effect of scutellarin via inhibition of endothelial-mesenchymal transition on isoprenaline-induced myocardial fibrosis in rats. Molecules 2014, 19, 15611–15623. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Chen, X.; Cai, J.J.; Chen, L.Z.; Gong, Y.S.; Wang, L.X.; Gao, Z.; Zhang, H.Q.; Huang, W.J.; Zhou, H. Relaxin inhibits cardiac fibrosis and endothelial-mesenchymal transition via the Notch pathway. Drug Des. Devel Ther. 2015, 9, 4599–4611. [Google Scholar] [CrossRef] [PubMed]
- Milan, M.; Pace, V.; Maiullari, F.; Chirivi, M.; Baci, D.; Maiullari, S.; Madaro, L.; Maccari, S.; Stati, T.; Marano, G.; et al. Givinostat reduces adverse cardiac remodeling through regulating fibroblasts activation. Cell Death Dis. 2018, 9, 108. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Peng, Z.; Zu, C.; Ma, J.; Lu, S.; Zhong, J.; Zhang, S. Losartan Attenuates Myocardial Endothelial-To-Mesenchymal Transition in Spontaneous Hypertensive Rats via Inhibiting TGF-beta/Smad Signaling. PLoS ONE 2016, 11, e0155730. [Google Scholar] [CrossRef]
- Wylie-Sears, J.; Aikawa, E.; Levine, R.A.; Yang, J.H.; Bischoff, J. Mitral valve endothelial cells with osteogenic differentiation potential. Arter. Thromb. Vasc. Biol. 2011, 31, 598–607. [Google Scholar] [CrossRef]
- Ma, Z.; Zhu, L.; Liu, Y.; Wang, Z.; Yang, Y.; Chen, L.; Lu, Q. Lovastatin Alleviates Endothelial-to-Mesenchymal Transition in Glomeruli via Suppression of Oxidative Stress and TGF-beta1 Signaling. Front. Pharmacol. 2017, 8, 473. [Google Scholar] [CrossRef]
- Suzuki, T.; Tada, Y.; Gladson, S.; Nishimura, R.; Shimomura, I.; Karasawa, S.; Tatsumi, K.; West, J. Vildagliptin ameliorates pulmonary fibrosis in lipopolysaccharide-induced lung injury by inhibiting endothelial-to-mesenchymal transition. Respir. Res. 2017, 18, 177. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Tang, R.N.; Liu, H.; Xu, M.; Pan, M.M.; Liu, B.C. Cinacalcet attenuates the renal endothelial-to-mesenchymal transition in rats with adenine-induced renal failure. Am. J. Physiol. Ren. Physiol. 2014, 306, F138–F146. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Wang, J.; Cheng, Y.; Li, X.; He, M.; Zhu, J.; Han, H.; Wei, G.; Kong, H.; Xie, W.; et al. Glucagon-Like Peptide-1 Mediates the Protective Effect of the Dipeptidyl Peptidase IV Inhibitor on Renal Fibrosis via Reducing the Phenotypic Conversion of Renal Microvascular Cells in Monocrotaline-Treated Rats. Biomed. Res. Int. 2018, 2018, 1864107. [Google Scholar] [CrossRef] [PubMed]
- Song, Y.; Lee, S.Y.; Kim, A.R.; Kim, S.; Heo, J.; Shum, D.; Kim, S.H.; Choi, I.; Lee, Y.J.; Seo, H.R. Identification of radiation-induced EndMT inhibitors through cell-based phenomic screening. FEBS Open Bio 2019, 9, 82–91. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhang, Y.X.; Ning, D.S.; Chen, J.; Li, S.X.; Mo, Z.W.; Peng, Y.M.; He, S.H.; Chen, Y.T.; Zheng, C.J.; et al. Simvastatin inhibits POVPC-mediated induction of endothelial-to-mesenchymal cell transition. J. Lipid Res. 2021, 62, 100066. [Google Scholar] [CrossRef] [PubMed]
- Tuuminen, R.; Syrjala, S.; Krebs, R.; Keranen, M.A.; Koli, K.; Abo-Ramadan, U.; Neuvonen, P.J.; Tikkanen, J.M.; Nykanen, A.I.; Lemstrom, K.B. Donor simvastatin treatment abolishes rat cardiac allograft ischemia/reperfusion injury and chronic rejection through microvascular protection. Circulation 2011, 124, 1138–1150. [Google Scholar] [CrossRef] [PubMed]
- Goldmann, J.M.; Veltman, J.A.; Gilissen, C. De Novo Mutations Reflect Development and Aging of the Human Germline. Trends Genet. 2019, 35, 828–839. [Google Scholar] [CrossRef] [PubMed]
- Winkler, E.A.; Birk, H.; Burkhardt, J.K.; Chen, X.; Yue, J.K.; Guo, D.; Rutledge, W.C.; Lasker, G.F.; Partow, C.; Tihan, T.; et al. Reductions in brain pericytes are associated with arteriovenous malformation vascular instability. J. Neurosurg. 2018, 129, 1464–1474. [Google Scholar] [CrossRef] [PubMed]
- Ricciardelli, A.R.; Robledo, A.; Fish, J.E.; Kan, P.T.; Harris, T.H.; Wythe, J.D. The Role and Therapeutic Implications of Inflammation in the Pathogenesis of Brain Arteriovenous Malformations. Biomedicines 2023, 11, 2876. [Google Scholar] [CrossRef]
- Zhang, J.; Cai, L.; Song, Y.; Shan, B.; He, M.; Ren, Q.; Chen, C.; Liu, Z.; Zeng, Y.; Xu, J. Prognostic role of neutrophil lymphocyte ratio in patients with spontaneous intracerebral hemorrhage. Oncotarget 2017, 8, 77752–77760. [Google Scholar] [CrossRef]
- Bonaventura, A.; Vecchie, A.; Abbate, A.; Montecucco, F. Neutrophil Extracellular Traps and Cardiovascular Diseases: An Update. Cells 2020, 9, 231. [Google Scholar] [CrossRef] [PubMed]
- Massena, S.; Christoffersson, G.; Vagesjo, E.; Seignez, C.; Gustafsson, K.; Binet, F.; Herrera Hidalgo, C.; Giraud, A.; Lomei, J.; Westrom, S.; et al. Identification and characterization of VEGF-A-responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans. Blood 2015, 126, 2016–2026. [Google Scholar] [CrossRef] [PubMed]
- Shimada, K.; Yamaguchi, I.; Ishihara, M.; Miyamoto, T.; Sogabe, S.; Miyake, K.; Tada, Y.; Kitazato, K.T.; Kanematsu, Y.; Takagi, Y. Involvement of Neutrophil Extracellular Traps in Cerebral Arteriovenous Malformations. World Neurosurg. 2021, 155, e630–e636. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Hossain, M.; Thanabalasuriar, A.; Gunzer, M.; Meininger, C.; Kubes, P. Visualizing the function and fate of neutrophils in sterile injury and repair. Science 2017, 358, 111–116. [Google Scholar] [CrossRef]
- Thomas, J.M.; Sasankan, D.; Surendran, S.; Abraham, M.; Rajavelu, A.; Kartha, C.C. Aberrant regulation of retinoic acid signaling genes in cerebral arterio venous malformation nidus and neighboring astrocytes. J. Neuroinflammation 2021, 18, 61. [Google Scholar] [CrossRef]
- Rochfort, K.D.; Collins, L.E.; Murphy, R.P.; Cummins, P.M. Downregulation of blood-brain barrier phenotype by proinflammatory cytokines involves NADPH oxidase-dependent ROS generation: Consequences for interendothelial adherens and tight junctions. PLoS ONE 2014, 9, e101815. [Google Scholar] [CrossRef]
- Davies, P.F. Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat. Clin. Pr. Cardiovasc. Med. 2009, 6, 16–26. [Google Scholar] [CrossRef]
- Campinho, P.; Vilfan, A.; Vermot, J. Blood Flow Forces in Shaping the Vascular System: A Focus on Endothelial Cell Behavior. Front. Physiol. 2020, 11, 552. [Google Scholar] [CrossRef] [PubMed]
- Buschmann, I.; Pries, A.; Styp-Rekowska, B.; Hillmeister, P.; Loufrani, L.; Henrion, D.; Shi, Y.; Duelsner, A.; Hoefer, I.; Gatzke, N.; et al. Pulsatile shear and Gja5 modulate arterial identity and remodeling events during flow-driven arteriogenesis. Development 2010, 137, 2187–2196. [Google Scholar] [CrossRef]
- Hwa, J.J.; Beckouche, N.; Huang, L.; Kram, Y.; Lindskog, H.; Wang, R.A. Abnormal arterial-venous fusions and fate specification in mouse embryos lacking blood flow. Sci. Rep. 2017, 7, 11965. [Google Scholar] [CrossRef]
- le Noble, F.; Moyon, D.; Pardanaud, L.; Yuan, L.; Djonov, V.; Matthijsen, R.; Breant, C.; Fleury, V.; Eichmann, A. Flow regulates arterial-venous differentiation in the chick embryo yolk sac. Development 2004, 131, 361–375. [Google Scholar] [CrossRef] [PubMed]
- Lucitti, J.L.; Jones, E.A.; Huang, C.; Chen, J.; Fraser, S.E.; Dickinson, M.E. Vascular remodeling of the mouse yolk sac requires hemodynamic force. Development 2007, 134, 3317–3326. [Google Scholar] [CrossRef] [PubMed]
Agent | Target/Mechanism(s) | Stage of Investigation |
---|---|---|
Rapamycin (sirolimus) | mTOR pathway/ Allosteric inhibitor of mTORC1 [188] | |
Thalidomide | BBB impairment and angiogenesis/ Immunomodulation, increased mural cell coverage, vessel maturation, and PDGFB expression in ECs [87,204] VEGF antagonism via metabolites [194] |
|
Bevacizumab | VEGF overexpression/ Monoclonal antibody targeting VEGF [205] |
|
Pazopanib | Angiogenesis/ Antagonism of VEGF, PDGF, & FGF receptors, as well as c-Kit [200] |
|
Trametinib | MAPK/ERK pathway upregulation/ Allosteric inhibitor of MEK 1 & 2 [202] |
|
Indicated Pathway | Agent(s) | Context of Investigation | Clinically Related Outcomes |
---|---|---|---|
TGF-β/Smad | Evodiamine [464], Givinostat [470], Losartan [471], MEK inhibitor (U0126) [279], Pioglitazone [463], Prazosin [471], Puerarin [462], Rapamycin [465], Relaxin [466], Scutellarin [468], Simvastatin [478,479], Spironolactone [467] | Cardiac Fibrosis | Reduction in fibroblast activation, type I & III collagen production, pro-fibrotic cytokines production, and increased microvascular density |
Lovastatin [279,473] | KRASG12D-mutated HUVECs [279], Diabetic Nephropathy [473] | Suppression of oxidative stress and glomerular dysfunction | |
Liraglutide and Sitagliptin [476] | Renal Fibrosis | Inhibition of glomerular tuft hypertrophy, glomerular mesangial expansion, and microvascular thrombosis | |
Vildagliptin and linagliptin [474] | Pulmonary Fibrosis | Inhibited production of pro-fibrotic markers (α-SMA or S100A4) in pulmonary vascular ECs and EndMT cell activity | |
Undetermined | Cinacalcet [475] | Renal Fibrosis | Prevention of kidney dysfunction, proteinuria, and a reduction in circulating PTH in rodents |
Compound CHIR-99021 [477] | Pulmonary Fibrosis and Chemosensitivity in Irradiated Nonsmall cell lung cancer (NSCLC) cells | Inhibited stress-fiber production and EndMT marker (α-SMA) expression in irradiated cells |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jeong, J.Y.; Bafor, A.E.; Freeman, B.H.; Chen, P.R.; Park, E.S.; Kim, E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024, 12, 1795. https://doi.org/10.3390/biomedicines12081795
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines. 2024; 12(8):1795. https://doi.org/10.3390/biomedicines12081795
Chicago/Turabian StyleJeong, Jae Yeong, Adrian E. Bafor, Bridger H. Freeman, Peng R. Chen, Eun S. Park, and Eunhee Kim. 2024. "Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition" Biomedicines 12, no. 8: 1795. https://doi.org/10.3390/biomedicines12081795
APA StyleJeong, J. Y., Bafor, A. E., Freeman, B. H., Chen, P. R., Park, E. S., & Kim, E. (2024). Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines, 12(8), 1795. https://doi.org/10.3390/biomedicines12081795