Cytochrome P450 and matrix metalloproteinase genetic modifiers of disease severity in Cerebral Cavernous Malformation type 1

Hélène Choquet, Eliana Trapani, Luca Goitre, Lorenza Trabalzini, Amy Akers, Marco Fontanella, Blaine L Hart, Leslie A Morrison, Ludmila Pawlikowska, Helen Kim, Saverio Francesco Retta, Hélène Choquet, Eliana Trapani, Luca Goitre, Lorenza Trabalzini, Amy Akers, Marco Fontanella, Blaine L Hart, Leslie A Morrison, Ludmila Pawlikowska, Helen Kim, Saverio Francesco Retta

Abstract

Background: Familial Cerebral Cavernous Malformation type 1 (CCM1) is an autosomal dominant disease caused by mutations in the Krev Interaction Trapped 1 (KRIT1/CCM1) gene, and characterized by multiple brain lesions. CCM lesions manifest across a range of different phenotypes, including wide differences in lesion number, size and susceptibility to intracerebral hemorrhage (ICH). Oxidative stress plays an important role in cerebrovascular disease pathogenesis, raising the possibility that inter-individual variability in genes related to oxidative stress may contribute to the phenotypic differences observed in CCM1 disease. Here, we investigated whether candidate oxidative stress-related cytochrome P450 (CYP) and matrix metalloproteinase (MMP) genetic markers grouped by superfamilies, families or genes, or analyzed individually influence the severity of CCM1 disease.

Methods: Clinical assessment and cerebral susceptibility-weighted magnetic resonance imaging (SWI) were performed to determine total and large (≥5mm in diameter) lesion counts as well as ICH in 188 Hispanic CCM1 patients harboring the founder KRIT1/CCM1 'common Hispanic mutation' (CCM1-CHM). Samples were genotyped on the Affymetrix Axiom Genome-Wide LAT1 Human Array. We analyzed 1,122 genetic markers (both single nucleotide polymorphisms (SNPs) and insertion/deletions) grouped by CYP and MMP superfamily, family or gene for association with total or large lesion count and ICH adjusted for age at enrollment and gender. Genetic markers bearing the associations were then analyzed individually.

Results: The CYP superfamily showed a trend toward association with total lesion count (P=0.057) and large lesion count (P=0.088) in contrast to the MMP superfamily. The CYP4 and CYP8 families were associated with either large lesion count or total lesion count (P=0.014), and two other families (CYP46 and the MMP Stromelysins) were associated with ICH (P=0.011 and 0.007, respectively). CYP4F12 rs11085971, CYP8A1 rs5628, CYP46A1 rs10151332, and MMP3 rs117153070 single SNPs, mainly bearing the above-mentioned associations, were also individually associated with CCM1 disease severity.

Conclusions: Overall, our candidate oxidative stress-related genetic markers set approach outlined CYP and MMP families and identified suggestive SNPs that may impact the severity of CCM1 disease, including the development of numerous and large CCM lesions and ICH. These novel genetic risk factors of prognostic value could serve as early objective predictors of disease outcome and might ultimately provide better options for disease prevention and treatment.

Keywords: Cerebral Cavernous Malformation (CCM); Cerebrovascular disease; Cytochrome P450 (CYP); Disease severity; Familial Cerebral Cavernous Malformation type 1; Free radicals; Genetic markers set association study; Inter-individual variability in disease susceptibility and outcome; Intracerebral hemorrhage; KRIT1/CCM1; Matrix metalloproteinase (MMP); Oxidative stress; Reactive oxygen species (ROS); Vascular brain lesions.

Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract

References

    1. Clatterbuck R.E. Ultrastructural and immunocytochemical evidence that an incompetent blood-brain barrier is related to the pathophysiology of cavernous malformations. J. Neurol. Neurosurg. Psychiatry. 2001;71(2):188–192.
    1. Gault J. Pathobiology of human cerebrovascular malformations: basic mechanisms and clinical relevance. Neurosurgery. 2004;55(1):1–16. (discussion 16-7)
    1. Batra S. Cavernous malformations: natural history, diagnosis and treatment. Nat. Rev. Neurol. 2009;5(12):659–670.
    1. Cavalcanti D.D. Cerebral cavernous malformations: from genes to proteins to disease. J. Neurosurg. 2012;116(1):122–132.
    1. Denier C. Genotype–phenotype correlations in cerebral cavernous malformations patients. Ann. Neurol. 2006;60(5):550–556.
    1. Rigamonti D. Cavernous Malformations of the Nervous System. Cambridge University Press; 2011.
    1. Choquet H. Association of cardiovascular risk factors with disease severity in cerebral cavernous malformation type 1 subjects with the common Hispanic mutation. Cerebrovasc. Dis. 2014;37(1):57–63.
    1. Choquet H. Polymorphisms in inflammatory and immune response genes associated with cerebral cavernous malformation type 1 severity. Cerebrovasc. Dis. 2014;38(6):433–440.
    1. Boulday G. Tissue-specific conditional CCM2 knockout mice establish the essential role of endothelial CCM2 in angiogenesis: implications for human cerebral cavernous malformations. Dis. Models Mech. 2009;2(3–4):168–177.
    1. He Y. Stabilization of VEGFR2 signaling by cerebral cavernous malformation 3 is critical for vascular development. Sci. Signal. 2010;3(116)
    1. Whitehead K.J. The cerebral cavernous malformation signaling pathway promotes vascular integrity via Rho GTPases. Nat. Med. 2009;15(2):177–184.
    1. Whitehead K.J. CCM1 is required for arterial morphogenesis: implications for the etiology of human cavernous malformations. Development. 2004;131(6):1437–1448.
    1. Zheng X.J. CCM3 signaling through sterile 20-like kinases plays an essential role during zebrafish cardiovascular development and cerebral cavernous malformations. J. Clin. Investig. 2010;120(8):2795–2804.
    1. Boulday G. Developmental timing of CCM2 loss influences cerebral cavernous malformations in mice. J. Exp. Med. 2011;208(9):1835–1847.
    1. Fischer A. Cerebral cavernous malformations: from CCM genes to endothelial cell homeostasis. Trends Mol. Med. 2013;19(5):302–308.
    1. Gibson C.C. Strategy for identifying repurposed drugs for the treatment of cerebral cavernous malformation. Circulation. 2015;131(3):289–299.
    1. Chrissobolis S. Oxidative stress and endothelial dysfunction in cerebrovascular disease. Front Biosci. (Landmark Ed.) 2011;16:1733–1745.
    1. Goitre L. Molecular crosstalk between integrins and cadherins: do reactive oxygen species set the talk? J. Signal. Transduct. 2012;2012:807682.
    1. Freeman L.R., Keller J.N. Oxidative stress and cerebral endothelial cells: regulation of the blood–brain-barrier and antioxidant based interventions. Biochim. Biophys. Acta. 2012;1822(5):822–829.
    1. Faraci F.M. Protecting against vascular disease in brain. Am. J. Physiol. Heart Circ. Physiol. 2011;300(5):H1566–H1582.
    1. Fukai T., Ushio-Fukai M. Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid. Redox Signal. 2011;15(6):1583–1606.
    1. Goitre L. KRIT1 loss of function causes a ROS-dependent upregulation of c-Jun. Free Radic Biol Med. 2014;68:134–147.
    1. Ushio-Fukai M. Compartmentalization of redox signaling through NADPH oxidase-derived ROS. Antioxid. Redox Signal. 2009;11(6):1289–1299.
    1. Goitre L. KRIT1 regulates the homeostasis of intracellular reactive oxygen species. PLoS One. 2010;5(7):e11786.
    1. Fidalgo M. Adaptor protein cerebral cavernous malformation 3 (CCM3) mediates phosphorylation of the cytoskeletal proteins ezrin/radixin/moesin by mammalian Ste20-4 to protect cells from oxidative stress. J. Biol. Chem. 2012;287(14):11556–11565.
    1. Moglia A. Evaluation of the bioactive properties of avenanthramide analogs produced in recombinant yeast. Biofactors. 2015;41(1):15–27.
    1. Jung K.H. Proteomic analysis reveals KRIT1 as a modulator for the antioxidant effects of valproic acid in human bone-marrow mesenchymal stromal cells. Drug Chem. Toxicol. 2015;38(3):286–292.
    1. Marchi S. Defective autophagy is a key feature of cerebral cavernous malformations. EMBO Mol. Med. 2015;7(11):1403–1417.
    1. Trapani E., Retta S.F. Cerebral cavernous malformation (CCM) disease: from monogenic forms to genetic susceptibility factors. J. Neurosurg. Sci. 2015;59(3):201–209.
    1. Guzik T.J. Functional effect of the C242T polymorphism in the NAD(P)H oxidase p22phox gene on vascular superoxide production in atherosclerosis. Circulation. 2000;102(15):1744–1747.
    1. Heslop C.L. Combined polymorphisms in oxidative stress genes predict coronary artery disease and oxidative stress in coronary angiography patients. Ann. Hum. Genet. 2012;76:435–447.
    1. Klaunig J.E. Oxidative stress and oxidative damage in chemical carcinogenesis. Toxicol. Appl. Pharmacol. 2011;254(2):86–99.
    1. L. Lyrenas, E. Wincent, L. Forsberg, U. De Faire, R. Morgenstern, The potential impact of polymorphism on oxidative stress status, in: Redox-Genome Interactions in Health and Disease, CRC Press, 2003.
    1. Cheng S.L., Yu C.J., Yang P.C. Genetic polymorphisms of cytochrome p450 and matrix metalloproteinase in chronic obstructive pulmonary disease. Biochem. Genet. 2009;47(7–8):591–601.
    1. van Eeden S.F., Sin D.D. Oxidative stress in chronic obstructive pulmonary disease: a lung and systemic process. Can. Respir. J. 2013;20(1):27–29.
    1. Nebert D.W., Wikvall K., Miller W.L. Human cytochromes P450 in health and disease. Philos. Trans. R. Soc. Lond. B: Biol. Sci. 2013;368(1612):20120431.
    1. Nebert D.W., Dalton T.P. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat. Rev. Cancer. 2006;6(12):947–960.
    1. Zangar R.C., Davydov D.R., Verma S. Mechanisms that regulate production of reactive oxygen species by cytochrome P450. Toxicol. Appl. Pharmacol. 2004;199(3):316–331.
    1. Shahabi P. Human cytochrome P450 epoxygenases: variability in expression and role in inflammation-related disorders. Pharmacol. Ther. 2014;144(2):134–161.
    1. Hedlund E., Gustafsson J.A., Warner M. Cytochrome P450 in the brain; a review. Curr. Drug Metab. 2001;2(3):245–263.
    1. Haining R.L., Nichols-Haining M. Cytochrome P450-catalyzed pathways in human brain: metabolism meets pharmacology or old drugs with new mechanism of action? Pharmacol. Ther. 2007;113(3):537–545.
    1. Ghosh C. Pattern of P450 expression at the human blood–brain barrier: roles of epileptic condition and laminar flow. Epilepsia. 2010;51(8):1408–1417.
    1. Imig J.D. Cytochrome P450 eicosanoids and cerebral vascular function. Expert Rev. Mol. Med. 2011;13:e7.
    1. Donnelly M.K. Genetic markers in the EET metabolic pathway are associated with outcomes in patients with aneurysmal subarachnoid hemorrhage. J. Cereb. Blood Flow Metab. 2015;35(2):267–276.
    1. Gebremedhin D., Gopalakrishnan S., Harder D.R. Endogenous events modulating myogenic regulation of cerebrovascular function. Curr. Vasc. Pharmacol. 2014;12(6):810–817.
    1. Gu S. Association between genetic polymorphisms of cytochrome P450 2C19 and the risk of cerebral ischemic stroke in Chinese. BMC Med. Genet. 2014;15:83.
    1. Lee C.R. CYP2J2 and CYP2C8 polymorphisms and coronary heart disease risk: the Atherosclerosis Risk in Communities (ARIC) study. Pharmacogenet. Genom. 2007;17(5):349–358.
    1. Zanger U.M., Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 2013;138(1):103–141.
    1. Haorah J. Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction. J. Neurochem. 2007;101(2):566–576.
    1. Spinale F.G. Matrix metalloproteinases: regulation and dysregulation in the failing heart. Circ. Res. 2002;90(5):520–530.
    1. Kameda K. Correlation of oxidative stress with activity of matrix metalloproteinase in patients with coronary artery disease. Possible role for left ventricular remodelling. Eur. Heart J. 2003;24(24):2180–2185.
    1. Chen Y. Soluble endoglin modulates aberrant cerebral vascular remodeling. Ann. Neurol. 2009;66(1):19–27.
    1. Tang J. Mmp-9 deficiency enhances collagenase-induced intracerebral hemorrhage and brain injury in mutant mice. J. Cereb. Blood Flow Metab. 2004;24(10):1133–1145.
    1. Kelly P.J. Oxidative stress and matrix metalloproteinase-9 in acute ischemic stroke: the Biomarker Evaluation for Antioxidant Therapies in Stroke (BEAT-Stroke) study. Stroke. 2008;39(1):100–104.
    1. Garcia-Alloza M. Matrix metalloproteinase inhibition reduces oxidative stress associated with cerebral amyloid angiopathy in vivo in transgenic mice. J. Neurochem. 2009;109(6):1636–1647.
    1. Thomas D.D. Signaling and stress: the redox landscape in NOS2 biology. Free Radic. Biol. Med. 2015;87:204–225.
    1. Yoon S. Genetic analysis of MMP3, MMP9, and PAI-1 in Finnish patients with abdominal aortic or intracranial aneurysms. Biochem. Biophys. Res. Commun. 1999;265(2):563–568.
    1. Huai C. Allelic variation of the MMP3 promoter affects transcription activity through the transcription factor C-MYB in human brain arteriovenous malformations. PLoS One. 2013;8(3):e57958.
    1. Sun B. The rs9509 polymorphism of MMP-9 is associated with risk of hemorrhage in brain arteriovenous malformations. J. Clin. Neurosci. 2012;19(9):1287–1290.
    1. Zhao Y. The rs522616 polymorphism in the matrix metalloproteinase-3 (MMP-3) gene is associated with sporadic brain arteriovenous malformation in a Chinese population. J. Clin. Neurosci. 2010;17(12):1568–1572.
    1. Lamblin N. Polymorphisms in the promoter regions of MMP-2, MMP-3, MMP-9 and MMP-12 genes as determinants of aneurysmal coronary artery disease. J. Am. Coll. Cardiol. 2002;40(1):43–48.
    1. Li H. 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.
    1. Al-Shahi Salman R. Hemorrhage from cavernous malformations of the brain: definition and reporting standards. Angioma Alliance Scientific Advisory Board. Stroke. 2008;39(12):3222–3230.
    1. Hoffmann T.J. Next generation genome-wide association tool: design and coverage of a high-throughput European-optimized SNP array. Genomics. 2011;98(2):79–89.
    1. Florczak-Rzepka M. Matrix metalloproteinases in human spontaneous intracerebral hemorrhage: an update. Cerebrovasc. Dis. 2012;34(4):249–262.
    1. Yadav L. Matrix metalloproteinases and cancer-roles in threat and therapy. Asian Pac. J. Cancer Prev. 2014;15(3):1085–1091.
    1. Lin C.C. Functional polymorphisms in matrix metalloproteinases-1,-3,-9 are associated with arteriovenous fistula patency in hemodialysis patients. Clin. J. Am. Soc. Nephrol. 2010;5(10):1805–1814.
    1. Corbin Z.A. White matter hyperintensity volume correlates with matrix metalloproteinase-2 in acute ischemic stroke. J. Stroke Cerebrovasc. Dis. 2014;23(6):1300–1306.
    1. Niu W.Q., Qi Y. Matrix metalloproteinase family gene polymorphisms and risk for coronary artery disease: systematic review and meta-analysis. Heart. 2012;98(20):1483–1491.
    1. Huang D.W. The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007;8(9)
    1. Ferguson C.S., Tyndale R.F. Cytochrome P450 enzymes in the brain: emerging evidence of biological significance. Trends Pharmacol. Sci. 2011;32(12):708–714.
    1. Kalsotra A., Strobel H.W. Cytochrome P450 4F subfamily: at the crossroads of eicosanoid and drug metabolism. Pharmacol. Ther. 2006;112(3):589–611.
    1. Christmas P. Role of cytochrome P450s in inflammation. Adv. Pharmacol. 2015;74:163–192.
    1. Sehgal N. Cytochrome P4504f, a potential therapeutic target limiting neuroinflammation. Biochem. Pharmacol. 2011;82(1):53–64.
    1. Fang Y.C. Induction of prostacyclin/PGI2 synthase expression after cerebral ischemia–reperfusion. J. Cereb. Blood Flow Metab. 2006;26(4):491–501.
    1. Nakayama T. Prostacyclin synthase gene: genetic polymorphisms and prevention of some cardiovascular diseases. Curr. Med. Chem. Cardiovasc. Hematol. Agents. 2005;3(2):157–164.
    1. Ospina J.A., Krause D.N., Duckles S.P. 17Beta-estradiol increases rat cerebrovascular prostacyclin synthesis by elevating cyclooxygenase-1 and prostacyclin synthase. Stroke. 2002;33(2):600–605.
    1. Meliton A. KRIT1 mediates prostacyclin-induced protection against lung vascular permeability induced by excessive mechanical forces and TRAP6. Am. J. Respir. Cell Mol. Biol. 2015
    1. Duckles S.P., Krause D.N. Cerebrovascular effects of oestrogen: multiplicity of action. Clin. Exp. Pharmacol. Physiol. 2007;34(8):801–808.
    1. Krause D.N., Duckles S.P., Pelligrino D.A. Influence of sex steroid hormones on cerebrovascular function. J. Appl. Physiol. (1985) 2006;101(4):1252–1261.
    1. Stirone C. Estrogen increases mitochondrial efficiency and reduces oxidative stress in cerebral blood vessels. Mol. Pharmacol. 2005;68(4):959–965.
    1. Lorbek G., Lewinska M., Rozman D. Cytochrome P450s in the synthesis of cholesterol and bile acids-from mouse models to human diseases. FEBS J. 2012;279(9):1516–1533.
    1. Poli G., Biasi F., Leonarduzzi G. Oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 2013;1:125–130.
    1. Lund E.G. Knockout of the cholesterol 24-hydroxylase gene in mice reveals a brain-specific mechanism of cholesterol turnover. J. Biol. Chem. 2003;278(25):22980–22988.
    1. Russell D.W. Cholesterol 24-hydroxylase: an enzyme of cholesterol turnover in the brain. Annu. Rev. Biochem. 2009;78:1017–1040.
    1. Hughes T.M. Markers of cholesterol metabolism in the brain show stronger associations with cerebrovascular disease than Alzheimer’s disease. J. Alzheimers Dis. 2012;30(1):53–61.
    1. Sakaki T. Metabolism of vitamin D3 by cytochromes P450. Front. Biosci. 2005;10:119–134.
    1. Schuster I. Cytochromes P450 are essential players in the vitamin D signaling system. Biochim. Biophys. Acta. 2011;1814(1):186–199.
    1. H.J. Ting, Y.F. Lee, Vitamin D and oxidative stress, in: A.F. Gombart (Ed.), Vitamin D: Oxidative Stress, Immunity, and Aging, CRC Press, 2012, pp. 131–150
    1. Norman P.E., Powell J.T. Vitamin D and cardiovascular disease. Circ. Res. 2014;114(2):379–393.
    1. Van Hove I. Matrix metalloproteinase-3 in the central nervous system: a look on the bright side. J. Neurochem. 2012;123(2):203–216.
    1. Bruno G. Vascular extracellular matrix remodeling in cerebral aneurysms. J. Neurosurg. 1998;89(3):431–440.
    1. Yang Y., Rosenberg G.A. Blood–brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke. 2011;42(11):3323–3328.
    1. Fujimura M. Expression of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase (TIMP) in cerebral cavernous malformations: immunohistochemical analysis of MMP-2, -9 and TIMP-2. Acta Neurochir. (Wien) 2007;149(2):179–183. (discussion 183)
    1. Geer L.Y. The NCBI bioSystems database. Nucl. Acids Res. 2010;38:D492–D496.
    1. Singh R. Chronic oxidative stress sensitizes hepatocytes to death from 4-hydroxynonenal by JNK/c-Jun overactivation. Am. J. Physiol. Gastrointest. Liver Physiol. 2009;297(5):G907–G917.
    1. Miksys S.L., Tyndale R.F. Drug-metabolizing cytochrome P450s in the brain. J. Psychiatry Neurosci. 2002;27(6):406–415.
    1. Wang Y. Gene regulation of CYP4F11 in human keratinocyte HaCaT cells. Drug Metab. Dispos. 2010;38(1):100–107.
    1. Norlin M. Androgen receptor-mediated regulation of the anti-atherogenic enzyme CYP27A1 involves the JNK/c-jun pathway. Arch. Biochem. Biophys. 2011;506(2):236–241.
    1. Miodovnik M. The inflammatory response of keratinocytes and its modulation by vitamin D: the role of MAPK signaling pathways. J. Cell. Physiol. 2012;227(5):2175–2183.
    1. De Gobbi M. A regulatory SNP causes a human genetic disease by creating a new transcriptional promoter. Science. 2006;312(5777):1215–1217.
    1. von Ahsen N., Oellerich M. The intronic prothrombin 19911 A>G polymorphism influences splicing efficiency and modulates effects of the 20210G>A polymorphism on mRNA amount and expression in a stable reporter gene assay system. Blood. 2004;103(2):586–593.
    1. Li S.C., Tang P., Lin W.C. Intronic microRNA: discovery and biological implications. DNA Cell Biol. 2007;26(4):195–207.
    1. Cunningham F. Ensembl 2015. Nucl. Acids Res. 2015;43(D1):D662–D669.
    1. Dammann P. Two-hit mechanism in cerebral cavernous malformation? A case of monozygotic twins with a CCM1/KRIT1 germline mutation. Neurosurg. Rev. 2013;36(3):483–486.
    1. Nebert D.W., Russell D.W. Clinical importance of the cytochromes P450. Lancet. 2002;360(9340):1155–1162.
    1. Fleming I. Endothelium-derived hyperpolarizing factor synthase (Cytochrome P450 2C9) is a functionally significant source of reactive oxygen species in coronary arteries. Circ. Res. 2001;88(1):44–51.
    1. Ingelman-Sundberg M. Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol. Ther. 2007;116(3):496–526.
    1. Kim D.H. A Promoter polymorphism (rs17222919, -1316T/G) of ALOX5AP is associated with intracerebral hemorrhage in Korean population. Prostaglandins Leukot. Essent. Fatty Acids. 2011;85(3–4):115–120.
    1. Harrod C.G., Batjer H.H., Bendok B.R. Deficiencies in estrogen-mediated regulation of cerebrovascular homeostasis may contribute to an increased risk of cerebral aneurysm pathogenesis and rupture in menopausal and postmenopausal women. Med. Hypotheses. 2006;66(4):736–756.
    1. Jamous M.A. Role of estrogen deficiency in the formation and progression of cerebral aneurysms. Part I: experimental study of the effect of oophorectomy in rats. J. Neurosurg. 2005;103(6):1046–1051.
    1. Galluzzi S. Association of blood pressure and genetic background with white matter lesions in patients with mild cognitive impairment. J. Gerontol. A Biol. Sci. Med. Sci. 2008;63(5):510–517.
    1. Garcia A.N. Cyp46 polymorphisms in Alzheimer’s disease: a review. J. Mol. Neurosci. 2009;39(3):342–345.
    1. Norlin M. Androgen receptor-mediated regulation of the anti-atherogenic enzyme CYP27A1 involves the JNK/c-jun pathway. Arch. Biochem. Biophys. 2011;506(2):236–241.
    1. Nunes M.J. Okadaic acid inhibits the trichostatin A-mediated increase of human CYP46A1 neuronal expression in a ERK1/2-Sp3-dependent pathway. J. Lipid Res. 2012;53(9):1910–1919.
    1. Leoni V., Caccia C. Oxysterols as biomarkers in neurodegenerative diseases. Chem. Phys. Lipids. 2011;164(6):515–524.
    1. Jones G., Prosser D.E., Kaufmann M. Cytochrome P450-mediated metabolism of vitamin D. J. Lipid Res. 2014;55(1):13–31.
    1. Fornage M. Family-based association study of matrix metalloproteinase-3 and -9 haplotypes with susceptibility to ischemic white matter injury. Hum. Genet. 2007;120(5):671–680.
    1. Golden M.J. Increased number of white matter lesions in patients with familial cerebral cavernous malformations. AJNR Am. J. Neuroradiol. 2015;36(5):899–903.
    1. Efroni S., Schaefer C.F., Buetow K.H. Identification of key processes underlying cancer phenotypes using biologic pathway analysis. PLoS One. 2007;2(5):e425.
    1. Wu C.C., Strong L.C., Shete S. Effects of measured susceptibility genes on cancer risk in family studies. Hum. Genet. 2010;127(3):349–357.

Source: PubMed

3
订阅