231
Views
9
CrossRef citations to date
0
Altmetric
Review

Genetic and Proteomic Contributions to the Pathophysiology of Moyamoya Angiopathy and Related Vascular Diseases

& ORCID Icon
Pages 145-171 | Published online: 18 Mar 2021

References

  • Scott RM, Smith ER. Moyamoya disease and moyamoya syndrome. N Engl J Med. 2009;360(12):1226–1237. doi:10.1056/NEJMra0804622
  • Smith ER. Moyamoya Biomarkers. J Korean Neurosurg Soc. 2015;57(6):415–421.
  • Roach ES, Golomb MR, Adams R, et al. Management of stroke in infants and children: a scientific statement from a Special Writing Group of the American Heart Association Stroke Council and the Council on Cardiovascular Disease in the Young. Stroke. 2008;39(9):2644–2691.
  • Guey S, Tournier-Lasserve E, Herve D, Kossorotoff M. Moyamoya disease and syndromes: from genetics to clinical management. Appl Clin Genet. 2015;8:49–68.
  • Bedini G, Blecharz KG, Nava S, et al. Vasculogenic and angiogenic pathways in moyamoya disease. Curr Med Chem. 2016;23(4):315–345.
  • Park YS. Single nucleotide polymorphism in patients with moyamoya disease. J Korean Neurosurg Soc. 2015;57(6):422–427.
  • Kuroda S, Houkin K. Moyamoya disease: current concepts and future perspectives. Lancet Neurol. 2008;7(11):1056–1066.
  • Scott RM, Smith JL, Robertson RL, Madsen JR, Soriano SG, Rockoff MA. Long-term outcome in children with moyamoya syndrome after cranial revascularization by pial synangiosis. J Neurosurg. 2004;100(2Suppl Pediatrics):142–149.
  • Hishikawa T, Sugiu K, Date I. Moyamoyadisease: a review of clinical research. Acta Med Okayama. 2016;70(4):229–236.
  • Hishikawa T, Tokunaga K, Sugiu K, Date I. Clinical and radiographic features of moyamoya disease in patients with both cerebral ischaemia and haemorrhage. Br J Neurosurg. 2013;27(2):198–201. doi:10.3109/02688697.2012.717983
  • Kang S, Liu X, Zhang D, et al. Natural course of moyamoya disease in patients with prior hemorrhagic stroke. Stroke. 2019;50(5):1060–1066.
  • Okada Y, Kawamata T, Kawashima A, Yamaguchi K, Ono Y, Hori T. The efficacy of superficial temporal artery-middle cerebral artery anastomosis in patients with moyamoya disease complaining of severe headache. J Neurosurg. 2012;116(3):672–679.
  • Takeuchi K, Shimizu K. Hypoplasia of the bilateral internal carotid arteries. Brain Nerve. 1957;9:37–43.
  • Suzuki J, Takaku A. Cerebrovascular “moyamoya” disease. Disease showing abnormal net-like vessels in base of brain. Arch Neurol. 1969;20(3):288–299.
  • Fujimura M, Sonobe S, Nishijima Y, et al. Genetics and biomarkers of moyamoya disease: significance of RNF213 as a susceptibility gene. J Stroke. 2014;16(2):65–72.
  • Bang OY, Fujimura M, Kim SK. The pathophysiology of moyamoya disease: an update. J Stroke. 2016;18(1):12–20.
  • Roder C, Peters V, Kasuya H, et al. Common genetic polymorphisms in moyamoya and atherosclerotic disease in Europeans. Childs Nerv Syst. 2011;27(2):245–252.
  • Lin R, Xie Z, Zhang J, et al. Clinical and immunopathological features of moyamoya disease. PLoS One. 2012;7(4):e36386.
  • Milewicz DM, Kwartler CS, Papke CL, Regalado ES, Cao J, Reid AJ. Genetic variants promoting smooth muscle cell proliferation can result in diffuse and diverse vascular diseases: evidence for a hyperplastic vasculomyopathy. Genet Med. 2010;12(4):196–203.
  • Kang HS, Kim JH, Phi JH, et al. Plasma matrix metalloproteinases, cytokines and angiogenic factors in moyamoya disease. J Neurol Neurosurg Psychiatry. 2010;81(6):673–678.
  • Sigdel TK, Shoemaker LD, Chen R, et al. Immune response profiling identifies autoantibodies specific to moyamoya patients. Orphanet J Rare Dis. 2013;8:45.
  • Tseng HC, Kao HW, Ho MR, et al. Cytoskeleton network and cellular migration modulated by nuclear-localized receptor tyrosine kinase ROR1. Anticancer Res. 2011;31(12):4239–4249.
  • Harada M, Miyoshi H, Uno M, et al. Neuronal impairment of adult moyamoya disease detected by quantified proton MRS and comparison with cerebral perfusion by SPECT with tc-99m HM-PAO: a trial of clinical quantification of metabolites. J Magn Reson Imaging. 1999;10(2):124–129.
  • Milam KE, Parikh SM. The angiopoietin-Tie2 signaling axis in the vascular leakage of systemic inflammation. Tissue Barriers. 2015;3(1–2):e957508.
  • Kamath BM, Bauer RC, Loomes KM, et al. NOTCH2 mutations in Alagille syndrome. J Med Genet. 2012;49(2):138–144.
  • Oda T, Elkahloun AG, Pike BL, et al. Mutations in the human Jagged1 gene are responsible for Alagille syndrome. Nat Genet. 1997;16(3):235–242.
  • Li L, Krantz ID, Deng Y, et al. Alagille syndrome is caused by mutations in human Jagged1, which encodes a ligand for Notch1. Nat Genet. 1997;16(3):243–251.
  • Connor SE, Hewes D, Ball C, Jarosz JM. Alagille syndrome associated with angiographic moyamoya. Childs Nerv Syst. 2002;18(3–4):186–190.
  • Emerick KM, Krantz ID, Kamath BM, et al. Intracranial vascular abnormalities in patients with Alagille syndrome. J Pediatr Gastroenterol Nutr. 2005;41(1):99–107.
  • Gaba RC, Shah RP, Muskovitz AA, Guzman G, Michals EA. Synchronous moyamoya syndrome and ruptured cerebral aneurysm in Alagille syndrome. J Clin Neurosci. 2008;15(12):1395–1398.
  • Kamath BM, Spinner NB, Emerick KM, et al. Vascular anomalies in Alagille syndrome: a significant cause of morbidity and mortality. Circulation. 2004;109(11):1354–1358.
  • Rocha R, Soro I, Leitao A, Silva ML, Leao M. Moyamoya vascular pattern in Alagille syndrome. Pediatr Neurol. 2012;47(2):125–128.
  • Turnpenny PD, Ellard S. Alagille syndrome: pathogenesis, diagnosis and management. Eur J Hum Genet. 2012;20(3):251–257.
  • Woolfenden AR, Albers GW, Steinberg GK, Hahn JS, Johnston DC, Farrell K. Moyamoya syndrome in children with Alagille syndrome: additional evidence of a vasculopathy. Pediatrics. 1999;103(2):505–508.
  • Roberts AE, Allanson JE, Tartaglia M, Gelb BD. Noonan syndrome. Lancet. 2013;381(9863):333–342.
  • Shoemaker LD, Clark MJ, Patwardhan A, et al. Disease variant landscape of a large multiethnic population of moyamoya patients by exome sequencing. G3 (Bethesda). 2015;6(1):41–49.
  • Duan L, Wei L, Tian Y, et al. Susceptibility loci for moyamoya disease revealed by a genome-wide association study. Stroke. 2018;49(1):11–18.
  • Araki Y, Yoshikawa K, Okamoto S, Sumitomo M, Maruwaka M, Wakabayashi T. Identification of novel biomarker candidates by proteomic analysis of cerebrospinal fluid from patients with moyamoya disease using SELDI-TOF-MS. BMC Neurol. 2010;10:112.
  • Lo EH, Dalkara T, Moskowitz MA. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4(5):399–415.
  • Ikeda H, Sasaki T, Yoshimoto T, Fukui M, Arinami T. Mapping of a familial moyamoya disease gene to chromosome 3p24.2-p26. Am J Hum Genet. 1999;64(2):533–537.
  • Bader HL, Systemic HT. VHL gene functions and the VHL disease. FEBS Lett. 2012;586(11):1562–1569.
  • Codd PJ, Scott RM, Smith ER. Seckel syndrome and moyamoya. J Neurosurg Pediatr. 2009;3(4):320–324.
  • Willems M, Genevieve D, Borck G, et al. Molecular analysis of pericentrin gene (PCNT) in a series of 24 Seckel/microcephalic osteodysplastic primordial dwarfism type II (MOPD II) families. J Med Genet. 2010;47(12):797–802.
  • Herve D, Philippi A, Belbouab R, et al. Loss of alpha1beta1 soluble guanylate cyclase, the major nitric oxide receptor, leads to moyamoya and achalasia. Am J Hum Genet. 2014;94(3):385–394.
  • Baffour R, Berman J, Garb JL, Rhee SW, Kaufman J, Friedmann P. Enhanced angiogenesis and growth of collaterals by in vivo administration of recombinant basic fibroblast growth factor in a rabbit model of acute lower limb ischemia: dose-response effect of basic fibroblast growth factor. J Vasc Surg. 1992;16(2):181–191.
  • Takagi Y, Kikuta K, Nozaki K, Hashimoto N. Histological features of middle cerebral arteries from patients treated for moyamoya disease. Neurol Med Chir (Tokyo). 2007;47(1):1–4.
  • Malek AM, Connors S, Robertson RL, Folkman J, Scott RM. Elevation of cerebrospinal fluid levels of basic fibroblast growth factor in moyamoya and central nervous system disorders. Pediatr Neurosurg. 1997;27(4):182–189.
  • Wang X, Zhang Z, Liu W, et al. Impacts and interactions of PDGFRB, MMP-3, TIMP-2, and RNF213 polymorphisms on the risk of moyamoya disease in Han Chinese human subjects. Gene. 2013;526(2):437–442.
  • Hidai C, Zupancic T, Penta K, et al. Cloning and characterization of developmental endothelial locus-1: an embryonic endothelial cell protein that binds the alphavbeta3 integrin receptor. Genes Dev. 1998;12(1):21–33.
  • Rezaee M, Penta K, Quertermous T. Del1 mediates VSMC adhesion, migration, and proliferation through interaction with integrin alpha(v)beta(3). Am J Physiol Heart Circ Physiol. 2002;282(5):H1924–H1932.
  • Inoue TK, Ikezaki K, Sasazuki T, Matsushima T, Fukui M. Linkage analysis of moyamoya disease on chromosome 6. J Child Neurol. 2000;15(3):179–182.
  • Weinberg DG, Arnaout OM, Rahme RJ, Aoun SG, Batjer HH, Bendok BR. Moyamoya disease: a review of histopathology, biochemistry, and genetics. Neurosurg Focus. 2011;30(6):E20.
  • Roder C, Nayak NR, Khan N, Tatagiba M, Inoue I, Krischek B. Genetics of moyamoya disease. J Hum Genet. 2010;55(11):711–716.
  • Young AM, Karri SK, Ogilvy CS, Zhao N. Is there a role for treating inflammation in moyamoya disease? A review of histopathology, genetics, and signaling cascades. Front Neurol. 2013;4:105.
  • Park YS, Jeon YJ, Kim HS, et al. The role of VEGF and KDR polymorphisms in moyamoya disease and collateral revascularization. PLoS One. 2012;7(10):e47158.
  • Hansen TM, Singh H, Tahir TA, Brindle NP. Effects of angiopoietins-1 and −2 on the receptor tyrosine kinase Tie2 are differentially regulated at the endothelial cell surface. Cell Signal. 2010;22(3):527–532.
  • Han H, Pyo CW, Yoo DS, Huh PW, Cho KS, Kim DS. Associations of moyamoya patients with HLA class I and class II alleles in the Korean population. J Korean Med Sci. 2003;18(6):876–880.
  • Hong SH, Wang KC, Kim SK, Cho BK, Park MH. Association of HLA-DR and -DQ genes with familial moyamoya disease in Koreans. J Korean Neurosurg Soc. 2009;46(6):558–563.
  • Takagi Y, Kikuta K, Nozaki K, et al. Expression of hypoxia-inducing factor-1 alpha and endoglin in intimal hyperplasia of the middle cerebral artery of patients with moyamoya disease. Neurosurgery. 2007;60(2):338–345.
  • Mouchtouris N, Jabbour PM, Starke RM, et al. Biology of cerebral arteriovenous malformations with a focus on inflammation. J Cereb Blood Flow Metab. 2015;35(2):167–175.
  • Gertz K, Kronenberg G, Kalin RE, et al. Essential role of interleukin-6 in post-stroke angiogenesis. Brain. 2012;135(Pt 6):1964–1980.
  • Hoffmann CJ, Harms U, Rex A, et al. Vascular signal transducer and activator of transcription-3 promotes angiogenesis and neuroplasticity long-term after stroke. Circulation. 2015;131(20):1772–1782.
  • Yamamoto M, Aoyagi M, Fukai N, Matsushima Y, Yamamoto K. Differences in cellular responses to mitogens in arterial smooth muscle cells derived from patients with moyamoya disease. Stroke. 1998;29(6):1188–1193.
  • Blecharz-Lang KG, Wagner J, Fries A, et al. Interleukin 6-mediated endothelial barrier disturbances can be attenuated by blockade of the IL6 receptor expressed in brain microvascular endothelial cells. Transl Stroke Res. 2018;9(6):631–642.
  • Pena-Silva RA, Chalouhi N, Wegman-Points L, et al. Novel role for endogenous hepatocyte growth factor in the pathogenesis of intracranial aneurysms. Hypertension. 2015;65(3):587–593.
  • Nakamura T, Mizuno S. The discovery of hepatocyte growth factor (HGF) and its significance for cell biology, life sciences and clinical medicine. Proc Jpn Acad Ser B Phys Biol Sci. 2010;86(6):588–610.
  • Nanba R, Kuroda S, Ishikawa T, Houkin K, Iwasaki Y. Increased expression of hepatocyte growth factor in cerebrospinal fluid and intracranial artery in moyamoya disease. Stroke. 2004;35(12):2837–2842.
  • Sakurai K, Horiuchi Y, Ikeda H, et al. A novel susceptibility locus for moyamoya disease on chromosome 8q23. J Hum Genet. 2004;49(5):278–281.
  • Felcht M, Luck R, Schering A, et al. Angiopoietin-2 differentially regulates angiogenesis through TIE2 and integrin signaling. J Clin Invest. 2012;122(6):1991–2005.
  • Jin KL, Mao XO, Nagayama T, Goldsmith PC, Greenberg DA. Induction of vascular endothelial growth factor and hypoxia-inducible factor-1alpha by global ischemia in rat brain. Neuroscience. 2000;99(3):577–585.
  • Kaelin WG. Von Hippel-Lindau disease. Annu Rev Pathol. 2007;2:145–173.
  • Comi AM. Pathophysiology of Sturge-Weber syndrome. J Child Neurol. 2003;18(8):509–516.
  • Guo DC, Papke CL, Tran-Fadulu V, et al. Mutations in smooth muscle alpha-actin (ACTA2) cause coronary artery disease, stroke, and moyamoya disease, along with thoracic aortic disease. Am J Hum Genet. 2009;84(5):617–627.
  • Munot P, Crow YJ, Ganesan V. Paediatric stroke: genetic insights into disease mechanisms and treatment targets. Lancet Neurol. 2011;10(3):264–274.
  • Munot P, Saunders DE, Milewicz DM, et al. A novel distinctive cerebrovascular phenotype is associated with heterozygous Arg179 ACTA2 mutations. Brain. 2012;135(Pt 8):2506–2514.
  • Sbaa E, Dewever J, Martinive P, et al. Caveolin plays a central role in endothelial progenitor cell mobilization and homing in SDF-1-driven postischemic vasculogenesis. Circ Res. 2006;98(9):1219–1227.
  • Ni G, Liu W, Huang X, et al. Increased levels of circulating SDF-1alpha and CD34+ CXCR4+ cells in patients with moyamoya disease. Eur J Neurol. 2011;18(11):1304–1309.
  • Ma J, You C. Association between matrix metalloproteinase-3 gene polymorphism and moyamoya disease. J Clin Neurosci. 2015;22(3):479–482.
  • Li H, Zhang ZS, Liu W, et al. Association of a functional polymorphism in the MMP-3 gene with moyamoya disease in the Chinese Han population. Cerebrovasc Dis. 2010;30(6):618–625.
  • Adam SS, Key NS, Greenberg CS. D-dimer antigen: current concepts and future prospects. Blood. 2009;113(13):2878–2887.
  • Dobson SR, Holden KR, Nietert PJ, et al. Moyamoya syndrome in childhood sickle cell disease: a predictive factor for recurrent cerebrovascular events. Blood. 2002;99(9):3144–3150.
  • Smith ER, McClain CD, Heeney M, Scott RM. Pial synangiosis in patients with moyamoya syndrome and sickle cell anemia: perioperative management and surgical outcome. Neurosurg Focus. 2009;26(4):E10.
  • Switzer JA, Hess DC, Nichols FT, Adams RJ. Pathophysiology and treatment of stroke in sickle-cell disease: present and future. Lancet Neurol. 2006;5(6):501–512.
  • Smith ER, Scott RM. Spontaneous occlusion of the circle of Willis in children: pediatric moyamoya summary with proposed evidence-based practice guidelines. A review. J Neurosurg Pediatr. 2012;9(4):353–360.
  • Schuhmacher AJ, Guerra C, Sauzeau V, Canamero M, Bustelo XR, Barbacid M. A mouse model for Costello syndrome reveals an Ang II-mediated hypertensive condition. J Clin Invest. 2008;118(6):2169–2179.
  • Wang S, Yu WM, Zhang W, McCrae KR, Neel BG, Qu CK. Noonan syndrome/leukemia-associated gain-of-function mutations in SHP-2 phosphatase (PTPN11) enhance cell migration and angiogenesis. J Biol Chem. 2009;284(2):913–920.
  • Korhonen EA, Lampinen A, Giri H, et al. Tie1 controls angiopoietin function in vascular remodeling and inflammation. J Clin Invest. 2016;126(9):3495–3510.
  • Wilhelm K, Happel K, Eelen G, et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature. 2016;529(7585):216–220.
  • Amano T, Inoha S, Wu CM, Matsushima T, Ikezaki K. Serum alpha1-antitrypsin level and phenotype associated with familial moyamoya disease. Childs Nerv Syst. 2003;19(9):655–658.
  • Lee JY, Moon YJ, Lee HO, et al. Deregulation of retinaldehyde dehydrogenase 2 leads to defective angiogenic function of endothelial colony-forming cells in pediatric moyamoya disease. Arterioscler Thromb Vasc Biol. 2015;35(7):1670–1677.
  • Kim SK, Yoo JI, Cho BK, et al. Elevation of CRABP-I in the cerebrospinal fluid of patients with moyamoya disease. Stroke. 2003;34(12):2835–2841.
  • Jeon JS, Ahn JH, Moon YJ, et al. Expression of cellular retinoic acid-binding protein-I (CRABP-I) in the cerebrospinal fluid of adult onset moyamoya disease and its association with clinical presentation and postoperative haemodynamic change. J Neurol Neurosurg Psychiatry. 2014;85(7):726–731.
  • Yamauchi T, Tada M, Houkin K, et al. Linkage of familial moyamoya disease (spontaneous occlusion of the circle of Willis) to chromosome 17q25. Stroke. 2000;31(4):930–935.
  • Xu J, Ismat FA, Wang T, Yang J, Epstein JA. NF1 regulates a Ras-dependent vascular smooth muscle proliferative injury response. Circulation. 2007;116(19):2148–2156.
  • Hamilton SJ, Friedman JM. Insights into the pathogenesis of neurofibromatosis 1 vasculopathy. Clin Genet. 2000;58(5):341–344.
  • Nanba R, Tada M, Kuroda S, Houkin K, Iwasaki Y. Sequence analysis and bioinformatics analysis of chromosome 17q25 in familial moyamoya disease. Childs Nerv Syst. 2005;21(1):62–68.
  • Mineharu Y, Liu W, Inoue K, et al. Autosomal dominant moyamoya disease maps to chromosome 17q25.3. Neurology. 2008;70(24 Pt 2):2357–2363.
  • Liu W, Hashikata H, Inoue K, et al. A rare Asian founder polymorphism of Raptor may explain the high prevalence of moyamoya disease among East Asians and its low prevalence among Caucasians. Environ Health Prev Med. 2010;15(2):94–104.
  • Kamada F, Aoki Y, Narisawa A, et al. A genome-wide association study identifies RNF213 as the first moyamoya disease gene. J Hum Genet. 2011;56(1):34–40.
  • Morito D, Nishikawa K, Hoseki J, et al. Moyamoya disease-associated protein mysterin/RNF213 is a novel AAA+ ATPase, which dynamically changes its oligomeric state. Sci Rep. 2015;4(1):4442. doi:10.1038/srep04442
  • Liu W, Morito D, Takashima S, et al. Identification of RNF213 as a susceptibility gene for moyamoya disease and its possible role in vascular development. PLoS One. 2011;6(7):e22542.
  • Miyatake S, Miyake N, Touho H, et al. Homozygous c.14576G>A variant of RNF213 predicts early-onset and severe form of moyamoya disease. Neurology. 2012;78(11):803–810.
  • Miyatake S, Touho H, Miyake N, et al. Sibling cases of moyamoya disease having homozygous and heterozygous c.14576G>A variant in RNF213 showed varying clinical course and severity. J Hum Genet. 2012;57(12):804–806.
  • Mineharu Y, Takagi Y, Takahashi JC, et al. Rapid progression of unilateral moyamoya disease in a patient with a family history and an RNF213 risk variant. Cerebrovasc Dis. 2013;36(2):155–157.
  • Itokawa K, Fukui M, Yamamoto T, Tamura N, Ishihara S, Araki N. Dural arteriovenous fistula as a possible cause of Tolosa-Hunt syndrome: a case report. J Neurol. 2010;257(5):846–847.
  • Yanagawa Y, Sugiura T, Suzuki K, Okada Y. Moyamoya disease associated with positive findings for rheumatoid factor and myeloperoxidase-anti-neutrophil cytoplasmic antibody. West Indian Med J. 2007;56(3):282–284.
  • Joutel A, Andreux F, Gaulis S, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000;105(5):597–605.
  • Roder C, Peters V, Kasuya H, et al. Polymorphisms in TGFB1 and PDGFRB are associated with moyamoya disease in European patients. Acta Neurochir (Wien). 2010;152(12):2153–2160.
  • Pepper MS. Transforming growth factor-beta: vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev. 1997;8(1):21–43.
  • McCartney-Francis N, Mizel D, Wong H, Wahl L, Wahl S. TGF-beta regulates production of growth factors and TGF-beta by human peripheral blood monocytes. Growth Factors. 1990;4(1):27–35.
  • Yamamoto M, Aoyagi M, Tajima S, et al. Increase in elastin gene expression and protein synthesis in arterial smooth muscle cells derived from patients with Moyamoya disease. Stroke. 1997;28(9):1733–1738.
  • Massague J. TGFbeta signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616–630.
  • Nakamura Y, Morishita R, Higaki J, et al. Expression of local hepatocyte growth factor system in vascular tissues. Biochem Biophys Res Commun. 1995;215(2):483–488.
  • Liu C, Roder C, Schulte C, et al. Analysis of TGFB1 in European and Japanese moyamoya disease patients. Eur J Med Genet. 2012;55(10):531–534.
  • Winkler DT, Bondolfi L, Herzig MC, et al. Spontaneous hemorrhagic stroke in a mouse model of cerebral amyloid angiopathy. J Neurosci. 2001;21(5):1619–1627.
  • Leno C, Mateo I, Cid C, Berciano J, Sedano C. Autoimmunity in Down’s syndrome: another possible mechanism of moyamoya disease. Stroke. 1998;29(4):868–869.
  • Aoyagi M, Fukai N, Sakamoto H, et al. Altered cellular responses to serum mitogens, including platelet-derived growth factor, in cultured smooth muscle cells derived from arteries of patients with moyamoya disease. J Cell Physiol. 1991;147(2):191–198.
  • Lim M, Cheshier S, Steinberg GK. New vessel formation in the central nervous system during tumor growth, vascular malformations, and moyamoya. Curr Neurovasc Res. 2006;3(3):237–245.
  • Herve D, Touraine P, Verloes A, et al. A hereditary moyamoya syndrome with multisystemic manifestations. Neurology. 2010;75(3):259–264.
  • Meyer S, Zanardo L, Kaminski WE, et al. Elastosis perforans serpiginosa-like pseudoxanthoma elasticum in a child with severe Moya Moya disease. Br J Dermatol. 2005;153(2):431–434.
  • Johnson C, Sung HJ, Lessner SM, Fini ME, Galis ZS. Matrix metalloproteinase-9 is required for adequate angiogenic revascularization of ischemic tissues: potential role in capillary branching. Circ Res. 2004;94(2):262–268.
  • Fujimura M, Watanabe M, Narisawa A, Shimizu H, Tominaga T. Increased expression of serum Matrix Metalloproteinase-9 in patients with moyamoya disease. Surg Neurol. 2009;72(5):476–480.
  • Sonobe S, Fujimura M, Niizuma K, et al. Increased vascular MMP-9 in mice lacking RNF213: moyamoya disease susceptibility gene. Neuroreport. 2014;25(18):1442–1446.
  • Asahara T, Chen D, Takahashi T, et al. Tie2 receptor ligands, angiopoietin-1 and angiopoietin-2, modulate VEGF-induced postnatal neovascularization. Circ Res. 1998;83(3):233–240.
  • Eklund L, Saharinen P. Angiopoietin signaling in the vasculature. Exp Cell Res. 2013;319(9):1271–1280.
  • Fagiani E, Christofori G. Angiopoietins in angiogenesis. Cancer Lett. 2013;328(1):18–26.
  • Kim M, Allen B, Korhonen EA, et al. Opposing actions of angiopoietin-2 on Tie2 signaling and FOXO1 activation. J Clin Invest. 2016;126(9):3511–3525.
  • Scheufler KM, Drevs J, van Velthoven V, et al. Implications of vascular endothelial growth factor, sFlt-1, and sTie-2 in plasma, serum and cerebrospinal fluid during cerebral ischemia in man. J Cereb Blood Flow Metab. 2003;23(1):99–110.
  • Tadros A, Hughes DP, Dunmore BJ, Brindle NP. ABIN-2 protects endothelial cells from death and has a role in the antiapoptotic effect of angiopoietin-1. Blood. 2003;102(13):4407–4409.
  • Jasmin JF, Malhotra S, Singh Dhallu M, Mercier I, Rosenbaum DM, Lisanti MP. Caveolin-1 deficiency increases cerebral ischemic injury. Circ Res. 2007;100(5):721–729.
  • Morais C, Ebrahem Q, Anand-Apte B, Parat MO. Altered angiogenesis in caveolin-1 gene-deficient mice is restored by ablation of endothelial nitric oxide synthase. Am J Pathol. 2012;180(4):1702–1714.
  • Sonveaux P, Martinive P, DeWever J, et al. Caveolin-1 expression is critical for vascular endothelial growth factor-induced ischemic hindlimb collateralization and nitric oxide-mediated angiogenesis. Circ Res. 2004;95(2):154–161.
  • Xu L, Guo R, Xie Y, Ma M, Ye R, Liu X. Caveolae: molecular insights and therapeutic targets for stroke. Expert Opin Ther Targets. 2015;19(5):633–650.
  • Jung KH, Chu K, Lee ST, et al. Circulating endothelial progenitor cells as a pathogenetic marker of moyamoya disease. J Cereb Blood Flow Metab. 2008;28(11):1795–1803.
  • Kang HS, Wang KC, Kim SK. Circulating vascular progenitor cells in moyamoya disease. J Korean Neurosurg Soc. 2015;57(6):428–431.
  • Kim JH, Jung JH, Phi JH, et al. Decreased level and defective function of circulating endothelial progenitor cells in children with moyamoya disease. J Neurosci Res. 2010;88(3):510–518.
  • Rafat N, Beck G, Pena-Tapia PG, Schmiedek P, Vajkoczy P. Increased levels of circulating endothelial progenitor cells in patients with moyamoya disease. Stroke. 2009;40(2):432–438.
  • Sugiyama T, Kuroda S, Nakayama N, Tanaka S, Houkin K. Bone marrow-derived endothelial progenitor cells participate in the initiation of moyamoya disease. Neurol Med Chir (Tokyo). 2011;51(11):767–773.
  • Yoshihara T, Taguchi A, Matsuyama T, et al. Increase in circulating CD34-positive cells in patients with angiographic evidence of moyamoya-like vessels. J Cereb Blood Flow Metab. 2008;28(6):1086–1089.
  • Zhang W, Wang X, Wang Z, Shang J, Wang H, Yin G. Effectiveness of superficial temporal artery-to-middle cerebral artery anastomosis in treating moyamoya disease by reducing endothelial progenitor cells. World Neurosurg. 2016;93:365–370.
  • Kang HS, Moon YJ, Kim YY, et al. Smooth-muscle progenitor cells isolated from patients with moyamoya disease: novel experimental cell model. J Neurosurg. 2014;120(2):415–425.
  • Bernard TJ, Fenton LZ, Apkon SD, et al. Biomarkers of hypercoagulability and inflammation in childhood-onset arterial ischemic stroke. J Pediatr. 2010;156(4):651–656.
  • Fujimura M, Fujimura T, Kakizaki A, et al. Increased serum production of soluble CD163 and CXCL5 in patients with moyamoya disease: involvement of intrinsic immune reaction in its pathogenesis. Brain Res. 2018;1679:39–44.
  • Phi JH, Suzuki N, Moon YJ, et al. Chemokine ligand 5 (CCL5) derived from endothelial colony-forming cells (ECFCs) mediates recruitment of smooth muscle progenitor cells (SPCs) toward critical vascular locations in moyamoya disease. PLoS One. 2017;12(1):e0169714.
  • Ogawa K, Nagahiro S, Arakaki R, Ishimaru N, Kobayashi M, Hayashi Y. Anti-alpha-fodrin autoantibodies in moyamoya disease. Stroke. 2003;34(12):e244–e246.
  • Ruggeberg S, Horn P, Li X, Vajkoczy P, Franz T. Detection of a gamma-carboxy-glutamate as novel post-translational modification of human transthyretin. Protein Pept Lett. 2008;15(1):43–46.
  • Koh EJ, Kim HN, Ma TZ, Choi HY, Kwak YG. Comparative analysis of serum proteomes of moyamoya disease and normal controls. J Korean Neurosurg Soc. 2010;48(1):8–13.
  • Kashiwazaki D, Uchino H, Kuroda S. Downregulation of apolipoprotein-E and apolipoprotein-J in moyamoya disease - a proteome analysis of cerebrospinal fluid. J Stroke Cerebrovasc Dis. 2017;26(12):2981–2987.
  • Maruwaka M, Yoshikawa K, Okamoto S, et al. Biomarker research for moyamoya disease in cerebrospinal fluid using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. J Stroke Cerebrovasc Dis. 2015;24(1):104–111.
  • Houkin K, Ito M, Sugiyama T, et al. Review of past research and current concepts on the etiology of moyamoya disease. Neurol Med Chir (Tokyo). 2012;52(5):267–277.
  • Chen JB, Liu Y, Zhou LX, Sun H, He M, You C. Increased prevalence of autoimmune disease in patients with unilateral compared with bilateral moyamoya disease. J Neurosurg. 2016;124(5):1215–1220.
  • Suzuki J, Kodama N. Moyamoya disease–a review. Stroke. 1983;14(1):104–109.
  • Hishikawa T, Tokunaga K, Sugiu K, Date I. Assessment of the difference in posterior circulation involvement between pediatric and adult patients with moyamoya disease. J Neurosurg. 2013;119(4):961–965.
  • Fujimura M, Tominaga T. Current status of revascularization surgery for moyamoya disease: special consideration for its ‘internal carotid-external carotid (IC-EC) conversion’ as the physiological reorganization system. Tohoku J Exp Med. 2015;236(1):45–53.
  • Bower RS, Mallory GW, Nwojo M, Kudva YC, Flemming KD, Meyer FB. Moyamoya disease in a primarily white, midwestern US population: increased prevalence of autoimmune disease. Stroke. 2013;44(7):1997–1999.
  • Chen JB, Liu Y, Zhou LX, Sun H, He M, You C. Prevalence of autoimmune disease in moyamoya disease patients in Western Chinese population. J Neurol Sci. 2015;351(1–2):184–186.
  • Suzuki S, Mitsuyama T, Horiba A, Fukushima S, Hashimoto N, Kawamata T. Moyamoya disease complicated by Graves’ disease and type 2 diabetes mellitus: report of two cases. Clin Neurol Neurosurg. 2011;113(4):325–329.
  • Mejia-Munne JC, Ellis JA, Feldstein NA, Meyers PM, Connolly ES. Moyamoya and inflammation. World Neurosurg. 2017;100:575–578.
  • Takagi Y, Hermanto Y, Takahashi JC, et al. Histopathological characteristics of distal middle cerebral artery in adult and pediatric patients with moyamoya disease. Neurol Med Chir (Tokyo). 2016;56(6):345–349.
  • Masuda J, Ogata J, Yutani C. Smooth muscle cell proliferation and localization of macrophages and T cells in the occlusive intracranial major arteries in moyamoya disease. Stroke. 1993;24(12):1960–1967.
  • Chalouhi N, Ali MS, Jabbour PM, et al. Biology of intracranial aneurysms: role of inflammation. J Cereb Blood Flow Metab. 2012;32(9):1659–1676.
  • Yu X, Li Z. MicroRNAs regulate vascular smooth muscle cell functions in atherosclerosis (review). Int J Mol Med. 2014;34(4):923–933.
  • Oka K, Yamashita M, Sadoshima S, Tanaka K. Cerebral haemorrhage in moyamoya disease at autopsy. Virchows Arch a Pathol Anat Histol. 1981;392(3):247–261.
  • Czabanka M, Pena-Tapia P, Schubert GA, Woitzik J, Vajkoczy P, Schmiedek P. Characterization of cortical microvascularization in adult moyamoya disease. Stroke. 2008;39(6):1703–1709.
  • Baltsavias G, Khan N, Valavanis A. The collateral circulation in pediatric moyamoya disease. Childs Nerv Syst. 2015;31(3):389–398.
  • Kim SJ, Son TO, Kim KH, et al. Neovascularization precedes occlusion in moyamoya disease: angiographic findings in 172 pediatric patients. Eur Neurol. 2014;72(5–6):299–305.
  • Hou K, Ji T, Guo Y, Xu K, Yu J. The coexistence of persistent primitive trigeminal artery, moyamoya disease, and multiple intracranial aneurysms: a case report and literature review. World Neurosurg. 2019;23.
  • Nishino K, Ito Y, Sorimachi T, Shimbo J, Fujii Y. Sturge-Weber syndrome associated with arteriovenous malformation in a patient presenting with progressive brain edema and cyst formation. J Neurosurg Pediatr. 2010;5(5):529–534.
  • Wu Z, Jiang H, Zhang L, et al. Molecular analysis of RNF213 gene for moyamoya disease in the Chinese Han population. PLoS One. 2012;7(10):e48179.
  • Mikami T, Suzuki H, Komatsu K, Mikuni N. Influence of inflammatory disease on the pathophysiology of moyamoya disease and quasi-moyamoya disease. Neurol Med Chir (Tokyo). 2019;59(10):361–370.
  • Choi EH, Lee H, Chung JW, et al. Ring finger protein 213 variant and plaque characteristics, vascular remodeling, and hemodynamics in patients with intracranial atherosclerotic stroke: a high-resolution magnetic resonance imaging and hemodynamic study. J Am Heart Assoc. 2019;8(20):e011996.
  • Zhu B, Liu X, Zhen X, et al. RNF213 gene polymorphism rs9916351 and rs8074015 significantly associated with moyamoya disease in Chinese population. Ann Transl Med. 2020;8(14):851.
  • Cecchi AC, Guo D, Ren Z, et al. RNF213 rare variants in an ethnically diverse population with moyamoya disease. Stroke. 2014;45(11):3200–3207.
  • Zhang Q, Liu Y, Zhang D, et al. RNF213 as the major susceptibility gene for Chinese patients with moyamoya disease and its clinical relevance. J Neurosurg. 2017;126(4):1106–1113.
  • Hitomi T, Habu T, Kobayashi H, et al. Downregulation of Securin by the variant RNF213 R4810K (rs112735431, G>A) reduces angiogenic activity of induced pluripotent stem cell-derived vascular endothelial cells from moyamoya patients. Biochem Biophys Res Commun. 2013;438(1):13–19.
  • Hitomi T, Habu T, Kobayashi H, et al. The moyamoya disease susceptibility variant RNF213 R4810K (rs112735431) induces genomic instability by mitotic abnormality. Biochem Biophys Res Commun. 2013;439(4):419–426.
  • Sonobe S, Fujimura M, Niizuma K, et al. Temporal profile of the vascular anatomy evaluated by 9.4-T magnetic resonance angiography and histopathological analysis in mice lacking RNF213: a susceptibility gene for moyamoya disease. Brain Res. 2014;1552:64–71.
  • Kanoke A, Fujimura M, Niizuma K, et al. Temporal profile of the vascular anatomy evaluated by 9.4-tesla magnetic resonance angiography and histological analysis in mice with the R4859K mutation of RNF213, the susceptibility gene for moyamoya disease. Brain Res. 2015;1624:497–505.
  • Kanoke A, Fujimura M, Niizuma K, et al. Temporal profile of magnetic resonance angiography and decreased ratio of regulatory T cells after immunological adjuvant administration to mice lacking RNF213, a susceptibility gene for moyamoya disease. Brain Res. 2016;1642:1–9.
  • Guo DC, Pannu H, Tran-Fadulu V, et al. Mutations in smooth muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat Genet. 2007;39(12):1488–1493.
  • Milewicz DM, Ostergaard JR, Ala-Kokko LM, et al. De novo ACTA2 mutation causes a novel syndrome of multisystemic smooth muscle dysfunction. Am J Med Genet A. 2010;152A(10):2437–2443.
  • Heyer GL, Dowling MM, Licht DJ, et al. The cerebral vasculopathy of PHACES syndrome. Stroke. 2008;39(2):308–316.
  • Siegel DH, Tefft KA, Kelly T, et al. Stroke in children with posterior fossa brain malformations, hemangiomas, arterial anomalies, coarctation of the aorta and cardiac defects, and eye abnormalities (PHACE) syndrome: a systematic review of the literature. Stroke. 2012;43(6):1672–1674.
  • Ganesan V, Kirkham FJ. Noonan syndrome and moyamoya. Pediatr Neurol. 1997;16(3):256–258.
  • Hinnant CA. Noonan syndrome associated with thromboembolic brain infarcts and posterior circulation abnormalities. Am J Med Genet. 1995;56(2):241–244.
  • Paez MT, Yamamoto T. Single nucleotide polymorphisms of tissue inhibitor of metalloproteinase genes in familial moyamoya disease. Neurosurgery. 2007;60(3):E582.
  • Bersano A, Guey S, Bedini G, et al. Research progresses in understanding the pathophysiology of moyamoya disease. Cerebrovasc Dis. 2016;41(3–4):105–118.
  • Aoyagi M, Fukai N, Matsushima Y, Yamamoto M, Yamamoto K. Kinetics of 125I-PDGF binding and down-regulation of PDGF receptor in arterial smooth muscle cells derived from patients with moyamoya disease. J Cell Physiol. 1993;154(2):281–288.
  • Saharinen P, Kerkela K, Ekman N, et al. Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2. J Cell Biol. 2005;169(2):239–243.
  • Blecharz KG, Frey D, Schenkel T, et al. Autocrine release of angiopoietin-2 mediates cerebrovascular disintegration in moyamoya disease. J Cereb Blood Flow Metab. 2016;5.
  • Yu J, Huang K, Pan J, Shen J, Zhan R. Significance of serum angiopoietin-2 in patients with hemorrhage in adult-onset moyamoya disease. Biomed Res Int. 2020;2020:8209313.
  • Bang OY, Chung JW, Kim SJ, et al. Caveolin-1, Ring finger protein 213, and endothelial function in moyamoya disease. Int J Stroke. 2016;11(9):999–1008.
  • Chung JW, Kim DH, Oh MJ, et al. Cav-1 (caveolin-1) and arterial remodeling in adult moyamoya disease. Stroke. 2018;49(11):2597–2604.
  • Taylor CT, Doherty G, Fallon PG, Cummins EP. Hypoxia-dependent regulation of inflammatory pathways in immune cells. J Clin Invest. 2016;126(10):3716–3724.
  • Zhang Z, Yao L, Yang J, Wang Z, Du G. PI3K/Akt and HIF1 signaling pathway in hypoxiaischemia (Review). Mol Med Rep. 2018;18(4):3547–3554.
  • Soriano SG, Cowan DB, Proctor MR, Scott RM. Levels of soluble adhesion molecules are elevated in the cerebrospinal fluid of children with moyamoya syndrome. Neurosurgery. 2002;50(3):544–549.
  • Hamauchi S, Shichinohe H, Uchino H, et al. Cellular functions and gene and protein expression profiles in endothelial cells derived from moyamoya disease-specific iPS cells. PLoS One. 2016;11(9):e0163561.
  • Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. Nov. 2010;5(6):463–466.
  • Bang OY, Goyal M, Liebeskind DS. Collateral circulation in ischemic stroke: assessment tools and therapeutic strategies. Stroke. 2015;46(11):3302–3309.
  • Navarro G, Borroto-Escuela DO, Fuxe K, Franco R. Potential of caveolae in the therapy of cardiovascular and neurological diseases. Front Physiol. 2014;5:370.
  • Lin N, Baird L, Koss M, et al. Discovery of asymptomatic moyamoya arteriopathy in pediatric syndromic populations: radiographic and clinical progression. Neurosurg Focus. 2011;31(6):E6.
  • Koss M, Scott RM, Irons MB, Smith ER, Ullrich NJ. Moyamoya syndrome associated with neurofibromatosis Type 1: perioperative and long-term outcome after surgical revascularization. J Neurosurg Pediatr. 2013;11(4):417–425.
  • Kuroda S, Hashimoto N, Yoshimoto T, Iwasaki Y. Research Committee on Moyamoya Disease in Japan. Radiological findings, clinical course, and outcome in asymptomatic moyamoya disease: results of multicenter survey in Japan. Stroke. 2007;38(5):1430–1435.
  • Kuroda S, Group AS. Asymptomatic moyamoya disease: literature review and ongoing AMORE study. Neurol Med Chir (Tokyo). 2015;55(3):194–198.
  • Lee JY, Kim SK, Cheon JE, et al. Posterior cerebral artery involvement in moyamoya disease: initial infarction and angle between PCA and basilar artery. Childs Nerv Syst. 2013;29(12):2263–2269.
  • Smith ER, Scott RM. Progression of disease in unilateral moyamoya syndrome. Neurosurg Focus. 2008;24(2):E17.