The Pathophysiology of Moyamoya Disease: An Update

Oh Young Bang, Miki Fujimura, Seung-Ki Kim, Oh Young Bang, Miki Fujimura, Seung-Ki Kim

Abstract

Moyamoya disease (MMD) is a unique cerebrovascular disease characterized by the progressive stenosis of large intracranial arteries and a hazy network of basal collaterals called moyamoya vessels. Because the etiology of MMD is unknown, its diagnosis is based on characteristic angiographic findings. Re-vascularization techniques (e.g., bypass surgery) are used to restore perfusion, and are the primary treatment for MMD. There is no specific treatment to prevent MMD progression. This review summarizes the recent advances in MMD pathophysiology, including the genetic and circulating factors related to disease development. Genetic and environmental factors may play important roles in the development of the vascular stenosis and aberrant angiogenesis in complex ways. These factors include the related changes in circulating endothelial/smooth muscle progenitor cells, cytokines related to vascular remodeling and angiogenesis, and endothelium, such as caveolin which is a plasma membrane protein. With a better understanding of MMD pathophysiology, nonsurgical approaches targeting MMD pathogenesis may be available to stop or slow the progression of this disease. The possible strategies include targeting growth factors, retinoic acid, caveolin-1, and stem cells.

Keywords: Angiogenesis; Caveolin; Endothelial progenitor cells; Growth factors; Moyamoya disease.

Conflict of interest statement

The authors have no financial conflicts of interest.

Figures

Figure 1.
Figure 1.
Potential mechanisms of moyamoya disease. The association between genetic, circulating, and environmental factors. RNF213, Ring finger 213; EPCs, endothelial progenitor cells; SPCs, smooth muscle progenitor cells; miRNAs, microRNAs; CRABP-1, cellular retinoic acid-binding protein-I.

References

    1. Scott RM, Smith ER. Moyamoya disease and moyamoya syndrome. N Engl J Med. 2009;360:1226–1237.
    1. Fukui M. Guidelines for the diagnosis and treatment of spontaneous occlusion of the circle of willis (‘moyamoya’ disease). Research committee on spontaneous occlusion of the circle of willis (moyamoya disease) of the ministry of health and welfare, japan. Clin Neurol Neurosurg. 1997;99 Suppl 2:S238–240.
    1. Bang OY, Ryoo S, Kim SJ, Yoon CH, Cha J, Yeon JY, et al. Adult moyamoya disease: a burden of intracranial stenosis in east asians? PLoS One. 2015;10:
    1. Suzuki J, Takaku A. Cerebrovascular “moyamoya” disease. Disease showing abnormal net-like vessels in base of brain. Arch Neurol. 1969;20:288–299.
    1. Kuroda S, Houkin K. Moyamoya disease: current concepts and future perspectives. Lancet Neurol. 2008;7:1056–1066.
    1. 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;47:1–4.
    1. Oka K, Yamashita M, Sadoshima S, Tanaka K. Cerebral haemorrhage in moyamoya disease at autopsy. Virchows Archiv. A, Pathological Anatomy and Histology. 1981;392:247–261.
    1. Chmelova J, Kolar Z, Prochazka V, Curik R, Dvorackova J, Sirucek P, et al. Moyamoya disease is associated with endothelial activity detected by anti-nestin antibody. Biomedical papers of the Medical Faculty of the University Palacky, Olomouc, Czechoslovakia. 2010;154:159–162.
    1. Lin R, Xie Z, Zhang J, Xu H, Su H, Tan X, et al. Clinical and immunopathological features of moyamoya disease. PLoS One. 2012;7:
    1. Takagi Y, Kikuta K, Nozaki K, Fujimoto M, Hayashi J, Imamura H, 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;60:338–345. discussion 345.
    1. Kaku Y, Morioka M, Ohmori Y, Kawano T, Kai Y, Fukuoka H, et al. Outer-diameter narrowing of the internal carotid and middle cerebral arteries in moyamoya disease detected on 3d constructive interference in steady-state mr image: is arterial constrictive remodeling a major pathogenesis? Acta Neurochir (Wien) 2012;154:2151–2157.
    1. Kim YJ, Lee DH, Kwon JY, Kang DW, Suh DC, Kim JS, et al. High resolution mri difference between moyamoya disease and intracranial atherosclerosis. Eur J Neurol. 2013;20:1311–1318.
    1. Ryoo S, Cha J, Kim SJ, Choi JW, Ki CS, Kim KH, et al. High-resolution magnetic resonance wall imaging findings of moyamoya disease. Stroke. 2014;45:2457–2460.
    1. Yuan M, Liu ZQ, Wang ZQ, Li B, Xu LJ, Xiao XL. High-resolution mr imaging of the arterial wall in moyamoya disease. Neurosci Lett. 2015;584:77–82.
    1. Guo DC, Papke CL, Tran-Fadulu V, Regalado ES, Avidan N, Johnson RJ, 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;84:617–627.
    1. Fukui M, Kono S, Sueishi K, Ikezaki K. Moyamoya disease. Neuropathology. 2000;20 Suppl:S61–64.
    1. Czabanka M, Pena-Tapia P, Schubert GA, Woitzik J, Vajkoczy P, Schmiedek P. Characterization of cortical microvascularization in adult moyamoya disease. Stroke. 2008;39:1703–1709.
    1. Kim SJ, Son TO, Kim KH, Jeon P, Hyun SH, Lee KH, et al. Neovascularization precedes occlusion in moyamoya disease: angiographic findings in 172 pediatric patients. Eur Neurol. 2014;72:299–305.
    1. 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;64:533–537.
    1. Inoue TK, Ikezaki K, Sasazuki T, Matsushima T, Fukui M. Linkage analysis of moyamoya disease on chromosome 6. J Child Neurol. 2000;15:179–182.
    1. Sakurai K, Horiuchi Y, Ikeda H, Ikezaki K, Yoshimoto T, Fukui M, et al. A novel susceptibility locus for moyamoya disease on chromosome 8q23. J Hum Genet. 2004;49:278–281.
    1. Yamauchi T, Tada M, Houkin K, Tanaka T, Nakamura Y, Kuroda S, et al. Linkage of familial moyamoya disease (spontaneous occlusion of the circle of willis) to chromosome 17q25. Stroke. 2000;31:930–935.
    1. Kamada F, Aoki Y, Narisawa A, Abe Y, Komatsuzaki S, Kikuchi A, et al. A genome-wide association study identifies rnf213 as the first moyamoya disease gene. J Hum Genet. 2011;56:34–40.
    1. Liu W, Morito D, Takashima S, Mineharu Y, Kobayashi H, Hitomi T, et al. Identification of rnf213 as a susceptibility gene for moyamoya disease and its possible role in vascular development. PLoS One. 2011;6:e22542.
    1. Miyatake S, Miyake N, Touho H, Nishimura-Tadaki A, Kondo Y, Okada I, et al. Homozygous c.14576g>a variant of rnf213 predicts early-onset and severe form of moyamoya disease. Neurology. 2012;78:803–810.
    1. Kim EH, Yum MS, Ra YS, Park JB, Ahn JS, Kim GH, et al. Importance of rnf213 polymorphism on clinical features and long-term outcome in moyamoya disease. J Neurosurg. 2015:1–7.
    1. Liu W, Hitomi T, Kobayashi H, Harada KH, Koizumi A. Distribution of moyamoya disease susceptibility polymorphism p.R4810k in rnf213 in east and southeast asian populations. Neurol Med Chir (Tokyo) 2012;52:299–303.
    1. Liu W, Hashikata H, Inoue K, Matsuura N, Mineharu Y, Kobayashi H, 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. Environmental Health and Preventive Medicine. 2010;15:94–104.
    1. Cecchi AC, Guo D, Ren Z, Flynn K, Santos-Cortez RL, Leal SM, et al. Rnf213 rare variants in an ethnically diverse population with moyamoya disease. Stroke. 2014;45:3200–3207.
    1. Wu Z, Jiang H, Zhang L, Xu X, Zhang X, Kang Z, et al. Molecular analysis of rnf213 gene for moyamoya disease in the chinese han population. PLoS One. 2012;7:
    1. Ma J, Liu Y, Ma L, Huang S, Li H, You C. Rnf213 polymorphism and moyamoya disease: a systematic review and meta-analysis. Neurology India. 2013;61:35–39.
    1. Kleinloog R, Regli L, Rinkel GJ, Klijn CJ. Regional differences in incidence and patient characteristics of moyamoya disease: a systematic review. J Neurol Neurosurg Psychiatry. 2012;83:531–536.
    1. Sonobe S, Fujimura M, Niizuma K, Nishijima Y, Ito A, Shimizu H, 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.
    1. Kanoke A, Fujimura M, Niizuma K, Ito A, Sakata H, Sato-Maeda M, 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 Research. 2015;1624:497–505.
    1. Fujimura M, Sonobe S, Nishijima Y, Niizuma K, Sakata H, Kure S, et al. Genetics and biomarkers of moyamoya disease: Significance of rnf213 as a susceptibility gene. Journal of Stroke. 2014;16:65–72.
    1. Ito A, Fujimura M, Niizuma K, Kanoke A, Sakata H, Morita-Fujimura Y, et al. Enhanced post-ischemic angiogenesis in mice lacking rnf213; a susceptibility gene for moyamoya disease. Brain Res. 2015;1594:310–320.
    1. Hitomi T, Habu T, Kobayashi H, Okuda H, Harada KH, Osafune K, 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;438:13–19.
    1. Kobayashi H, Matsuda Y, Hitomi T, Okuda H, Shioi H, Matsuda T, et al. Biochemical and functional characterization of rnf213 (mysterin) r4810k, a susceptibility mutation of moyamoya disease, in angiogenesis in vitro and in vivo. J Am Heart Assoc. 2015;4:
    1. Ohkubo K, Sakai Y, Inoue H, Akamine S, Ishizaki Y, Matsushita Y, et al. Moyamoya disease susceptibility gene rnf213 links inflammatory and angiogenic signals in endothelial cells. Sci Rep. 2015;5:13191.
    1. Kim SJ, Heo KG, Shin HY, Bang OY, Kim GM, Chung CS, et al. Association of thyroid autoantibodies with moyamoya-type cerebrovascular disease: A prospective study. Stroke. 2010;41:173–176.
    1. 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;44:1997–1999.
    1. Li H, Zhang ZS, Dong ZN, Ma MJ, Yang WZ, Han C, et al. Increased thyroid function and elevated thyroid autoantibodies in pediatric patients with moyamoya disease: a case-control study. Stroke. 2011;42:1138–1139.
    1. Koizumi A, Kobayashi H, Liu W, Fujii Y, Senevirathna ST, Nanayakkara S, et al. P.R4810k, a polymorphism of rnf213, the susceptibility gene for moyamoya disease, is associated with blood pressure. Environ Health Prev Med. 2013;18:121–129.
    1. Bartel DP. Micrornas: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297.
    1. Liu P, Zhao H, Wang R, Wang P, Tao Z, Gao L, et al. Microrna-424 protects against focal cerebral ischemia and reperfusion injury in mice by suppressing oxidative stress. Stroke. 2015;46:513–519.
    1. De Rosa S, Curcio A, Indolfi C. Emerging role of micrornas in cardiovascular diseases. Circulation Journal: Official Journal of the Japanese Circulation Society. 2014;78:567–575.
    1. Koutsis G, Siasos G, Spengos K. The emerging role of microrna in stroke. Current Topics in Medicinal Chemistry. 2013;13:1573–1588.
    1. Zeng L, He X, Wang Y, Tang Y, Zheng C, Cai H, et al. Microrna-210 overexpression induces angiogenesis and neurogenesis in the normal adult mouse brain. Gene Therapy. 2014;21:37–43.
    1. Dai D, Lu Q, Huang Q, Yang P, Hong B, Xu Y, et al. Serum mirna signature in moyamoya disease. PLoS One. 2014;9:
    1. Park YS, Jeon YJ, Lee BE, Kim TG, Choi JU, Kim DS, et al. Association of the mir-146ac>g, mir-196a2c>t, and mir-499a>g polymorphisms with moyamoya disease in the korean population. Neurosci Lett. 2012;521:71–75.
    1. Eberhard DA, Brown MD, VandenBerg SR. Alterations of annexin expression in pathological neuronal and glial reactions. Immunohistochemical localization of annexins i, ii (p36 and p11 subunits), iv, and vi in the human hippocampus. Am J Pathol. 1994;145:640–649.
    1. Herbert SP, Odell AF, Ponnambalam S, Walker JH. The confluence-dependent interaction of cytosolic phospholipase a2-alpha with annexin a1 regulates endothelial cell prostaglandin e2 generation. J Biol Chem. 2007;282:34468–34478.
    1. Kumar AH, Caplice NM. Clinical potential of adult vascular progenitor cells. Arterioscler Thromb Vasc Biol. 2010;30:1080–1087.
    1. Yoshihara T, Taguchi A, Matsuyama T, Shimizu Y, Kikuchi-Taura A, Soma T, et al. Increase in circulating cd34-positive cells in patients with angiographic evidence of moyamoya-like vessels. J Cereb Blood Flow Metab. 2008;28:1086–1089.
    1. 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;40:432–438.
    1. Kim JH, Jung JH, Phi JH, Kang HS, Kim JE, Chae JH, et al. Decreased level and defective function of circulating endothelial progenitor cells in children with moyamoya disease. J Neurosci Res. 2010;88:510–518.
    1. Jung KH, Chu K, Lee ST, Park HK, Kim DH, Kim JH, et al. Circulating endothelial progenitor cells as a pathogenetic marker of moyamoya disease. J Cereb Blood Flow Metab. 2008;28:1795–1803.
    1. Lee JY, Moon YJ, Lee HO, Park AK, Choi SA, Wang KC, 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;35:1670–1677.
    1. Kang HS, Moon YJ, Kim YY, Park WY, Park AK, Wang KC, et al. Smooth-muscle progenitor cells isolated from patients with moyamoya disease: Novel experimental cell model. J Neurosurg. 2014;120:415–425.
    1. Kang HS, Kim JH, Phi JH, Kim YY, Kim JE, Wang KC, et al. Plasma matrix metalloproteinases, cytokines and angiogenic factors in moyamoya disease. J Neurol Neurosurg Psychiatry. 2010;81:673–678.
    1. 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;35:2837–2842.
    1. Park YS, Jeon YJ, Kim HS, Chae KY, Oh SH, Han IB, et al. The role of vegf and kdr polymorphisms in moyamoya disease and collateral revascularization. PLoS One. 2012;7:
    1. Kim SK, Yoo JI, Cho BK, Hong SJ, Kim YK, Moon JA, et al. Elevation of crabp-i in the cerebrospinal fluid of patients with moyamoya disease. Stroke. 2003;34:2835–2841.
    1. 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;72:476–480. discussion 480.
    1. Kang HS, Kim SK, Cho BK, Kim YY, Hwang YS, Wang KC. Single nucleotide polymorphisms of tissue inhibitor of metalloproteinase genes in familial moyamoya disease. Neurosurgery. 2006;58:1074–1080. discussion 1074-1080.
    1. He J, Wang R, Zhang D, Zhang Y, Zhang Q, Zhao J. Expression of circulating vascular endothelial growth factor-antagonizing cytokines and vascular stabilizing factors prior to and following bypass surgery in patients with moyamoya disease. Exp Ther Med. 2014;8:302–308.
    1. Hoshimaru M, Takahashi JA, Kikuchi H, Nagata I, Hatanaka M. Possible roles of basic fibroblast growth factor in the pathogenesis of moyamoya disease: an immunohistochemical study. J Neurosurg. 1991;75:267–270.
    1. Park YS, Jeon YJ, Kim HS, Chae KY, Oh SH, Han IB, et al. The role of vegf and kdr polymorphisms in moyamoya disease and collateral revascularization. PLoS One. 2012;7:
    1. Wang X, Zhang Z, Liu W, Xiong Y, Sun W, Huang X, 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;526:437–442.
    1. 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. Frontiers in Neurology. 2013;4:105.
    1. Jeon JS, Ahn JH, Moon YJ, Cho WS, Son YJ, Kim SK, 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;85:726–731.
    1. 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;50:544–549.
    1. Noda A, Suzuki Y, Takayasu M, Watanabe K, Takagi T, Hara M, et al. Elevation of nitric oxide metabolites in the cerebrospinal fluid of patients with moyamoya disease. Acta Neurochirurgica. 2000;142:1275–1279. discussion 1279-1280.
    1. Park YS, Min KT, Kim TG, Lee YH, Cheong HJ, Yeom IS, et al. Age-specific enos polymorphisms in moyamoya disease. Childs Nerv Syst. 2011;27:1919–1926.
    1. Frank PG, Woodman SE, Park DS, Lisanti MP. Caveolin, caveolae, and endothelial cell function. Arterioscler Thromb Vasc Biol. 2003;23:1161–1168.
    1. Liu J, Wang XB, Park DS, Lisanti MP. Caveolin-1 expression enhances endothelial capillary tubule formation. The Journal of Biological Chemistry. 2002;277:10661–10668.
    1. Chang SH, Feng D, Nagy JA, Sciuto TE, Dvorak AM, Dvorak HF. Vascular permeability and pathological angiogenesis in caveolin-1-null mice. The American Journal of Pathology. 2009;175:1768–1776.
    1. Sonveaux P, Martinive P, DeWever J, Batova Z, Daneau G, Pelat M, et al. Caveolin-1 expression is critical for vascular endothelial growth factor-induced ischemic hindlimb collateralization and nitric oxide-mediated angiogenesis. Circulation Research. 2004;95:154–161.
    1. 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. The American Journal of Pathology. 2012;180:1702–1714.
    1. Sbaa E, Dewever J, Martinive P, Bouzin C, Frerart F, Balligand JL, et al. Caveolin plays a central role in endothelial progenitor cell mobilization and homing in sdf-1-driven postischemic vasculogenesis. Circulation Research. 2006;98:1219–1227.
    1. Navarro G, Borroto-Escuela DO, Fuxe K, Franco R. Potential of caveolae in the therapy of cardiovascular and neurological diseases. Frontiers in Physiology. 2014;5:370.
    1. Jasmin JF, Malhotra S, Singh Dhallu M, Mercier I, Rosenbaum DM, Lisanti MP. Caveolin-1 deficiency increases cerebral ischemic injury. Circulation Research. 2007;100:721–729.
    1. 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;19:633–650.
    1. Fujimura M, Shimizu H, Inoue T, Mugikura S, Saito A, Tominaga T. Significance of focal cerebral hyperperfusion as a cause of transient neurologic deterioration after extracranial-intracranial bypass for moyamoya disease: Comparative study with non-moyamoya patients using n-isopropyl-p-[(123)i]iodoamphetamine single-photon emission computed tomography. Neurosurgery. 2011;68:957–964. discussion 964-965.
    1. Bang OY, Goyal M, Liebeskind DS. Collateral circulation in ischemic stroke: Assessment tools and therapeutic strategies. Stroke. 2015;46:3302–3309.

Source: PubMed

3
Sottoscrivi