A suppressor screen in Mecp2 mutant mice implicates cholesterol metabolism in Rett syndrome

Christie M Buchovecky, Stephen D Turley, Hannah M Brown, Stephanie M Kyle, Jeffrey G McDonald, Benny Liu, Andrew A Pieper, Wenhui Huang, David M Katz, David W Russell, Jay Shendure, Monica J Justice, Christie M Buchovecky, Stephen D Turley, Hannah M Brown, Stephanie M Kyle, Jeffrey G McDonald, Benny Liu, Andrew A Pieper, Wenhui Huang, David M Katz, David W Russell, Jay Shendure, Monica J Justice

Abstract

Mutations in MECP2, encoding methyl CpG-binding protein 2, cause Rett syndrome, the most severe autism spectrum disorder. Re-expressing Mecp2 in symptomatic Mecp2-null mice markedly improves function and longevity, providing hope that therapeutic intervention is possible in humans. To identify pathways in disease pathology for therapeutic intervention, we carried out a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis suppressor screen in Mecp2-null mice and isolated five suppressors that ameliorate the symptoms of Mecp2 loss. We show that a stop codon mutation in Sqle, encoding squalene epoxidase, a rate-limiting enzyme in cholesterol biosynthesis, underlies suppression in one line. Subsequently, we also show that lipid metabolism is perturbed in the brains and livers of Mecp2-null male mice. Consistently, statin drugs improve systemic perturbations of lipid metabolism, alleviate motor symptoms and confer increased longevity in Mecp2 mutant mice. Our genetic screen therefore points to cholesterol homeostasis as a potential target for the treatment of patients with Rett syndrome.

Figures

Figure 1. A dominant suppressor screen shows…
Figure 1. A dominant suppressor screen shows inheritance of longevity in five lines
a) ENU-treated C57BL/6J males were mated to 129.Mecp2tm1.1Bird/+ females. G1Mecp2tm1.1Bird/Y males were aged and assessed for fore- and hindlimb clasping, tremors, body size, cage activity, and longevity. b) N2 animals from five lines (352 (Sum1m1Jus; blue), 856 (Sum2m1Jus; green), 895 (Sum3m1Jus; red), 1395 (Sum4m1Jus; brown) and 1527 (Sum5m1Jus; purple)) produced N3 offspring that exhibited increased longevity. A closed circle represents offspring of male N2 parents, and an open circle represents offspring of female N2 parents. The longevity of the G1 founder of each line is indicated by the colored square.
Figure 2
Figure 2
Survival curves for each line with a confirmed modifier map location are shown assessed at the N3 generation. Survival of Mecp2tm1.1Bird/Y mice is significantly increased by the presence of each suppressing mutation: a) Line 352 (Sum1m1Jus); p=.001. b) Line 856 (Sum2m1Jus); p=.005. c) Line 895 (Sum3m1Jus); p=.002. d) Line 1395 (Sum4m1Jus); p=.016.
Figure 3. A stop codon mutation in…
Figure 3. A stop codon mutation in Sqle confers rescue at Sum3m1Jus
a) The ENU-induced Sqle R399× mutation occurs in exon 7 in the squalene epoxidase domain. b)Sqle is not expressed in homozygous E8.0 mutant embryos. SqleΔ7 is the short predicted transcript lacking exon 7. c) Western blot on stage-matched E8.0 embryos shows that the expected 64kDa protein and 36kDa degradation product are absent in homozygous mutant embryos. d) Expression of Sqle but not Hmgcr is decreased in SqleSum3/+ brain; e) gene expression is unchanged by SqleSum3/+ in liver. i) Brain concentration of cholesterol precursors lanosterol and desmosterol is also decreased by SqleSum3/+. f) Serum cholesterol concentration is unchanged by SqleSum3/+. Tissue (d–g) analysis performed at P70; N=6 per group. Mecp2tm1.1Bird/Y SqleSum3/+ animals at backcross generation N7 to 129S6/SvEvTac show g) significantly improved rotarod performance at P56 (p=.0001), h) improved open field activity at P70. All error bars represent s.e.m.
Figure 4. Cholesterol metabolism is disrupted in…
Figure 4. Cholesterol metabolism is disrupted in Mecp2 null male mice
a) A simplified schematic of the enzymes and products in cholesterol biosynthesis via desmosterol is shown. b) Expression of Hmgcr, Sqle and Cyp46a1 in Mecp2tm1.1Bird/Y and Mecp2tm1.1Jae/Y are similar in brain. c) Lanosterol (Lan), desmosterol (Des) and total cholesterol (TC) concentrations are displayed per gram of brain tissue at P56 (N=8 per group) and P70 (N=4 per group). d) Cholesterol synthesis is decreased in Mecp2tm1.1Jae/Y brain at P56 (wild type N=4; null N=5). e) Expression of Hmgcr and Sqle in Mecp2tm1.1Bird/Y and Mecp2tm1.1Jae/Y differ in liver. f) Triacylglyceride (TAG) and TC concentrations are displayed per gram of liver tissue at P56 (N=6 per group). g) Cholesterol synthesis is slightly increased in Mecp2tm1.1Jae/Y liver per gram of tissue at P56 (wild type N=4; null N=5). Serum h) total cholesterol, i) LDL-cholesterol and j) triglyceride levels are significantly higher in Mecp2tm1.1Bird/Y mice by P56 (N=8–11 per group), but unchanged in Mecp2tm1.1Jae/Y mice (N=6 per group). For gene expression data (b,e) Bird: N=6 per genotype at P28, and 12 per genotype at P56; Jae: N=4 per genotype at P28, and 6 per genotype at P56. Tissue data (b–g) represent percentage change from wild type levels. *p≤0.05; All error bars represent s.e.m.
Figure 5. Statin treatment improves health in…
Figure 5. Statin treatment improves health in 129.Mecp2tm1.1Bird/Y males
Total animals assessed were 37 Mecp2tm1.1Bird/Y fluvastatin-treated, 12 Mecp2tm1.1Bird/Y lovastatin-treated, 31 Mecp2tm1.1Bird/Y vehicle-treated, 29 wild type +/Y fluvastatin-treated, 8 +/Y lovastatin-treated, and 29 wild type +/Y vehicle-treated mice for the following tests. a) Fluvastatin treatment of 129.Mecp2tm1.1Bird/Y confers increased longevity: median 122 days compared to 87 days with 57% survival beyond 120 days (p<.0001). Three animals were sacrificed due to dermatitis (boxes) while active and otherwise healthy. b) Rotarod performance improves in P56 treated null males (fluvastatin p=.015; lovastatin p=.009), c) Open field activity is increased in P70 treated null males as assessed by beam breaks (fluvastatin: p=.011, lovastatin: p=.049). d) Statin treatment lowers plasma cholesterol by P70 (fluvastatin: p=.001, lovastatin: p=.001). e) Statin treatment ameliorates elevated lipid concentration in 129.Mecp2tm1.1Bird/Y livers at P70 (fluvastatin p=.020; lovastatin: p=.386). The concentration of f) lanosterol slightly increases and g) desmosterol significantly increases in the brains of fluvastatin-treated 129.Mecp2tm1.1Bird/Y mice at P70 (N=4 per group; p=.042). All error bars represent s.e.m.
Figure 6. Fluvastatin treatment improves health in…
Figure 6. Fluvastatin treatment improves health in 129.Mecp2tm1.1Bird/+ females
a) No fluvastatin-treated 129.Mecp2tm1.1Bird/+ females died prior to eight months, but three vehicle-treated females died. b) Rotarod performance improves in five-month-old fluvastatin-treated 129.Mecp2tm1.1Bird/+ females (p=.001). c) Open field activity assessed at four months shows no significant differences in fluvastatin- or vehicle-treated groups. d) Fluvastatin treatment does not significantly change serum cholesterol levels at eight months. e) Fluvastatin treatment ameliorates elevated lipid concentration in 129.Mecp2tm1.1Bird/+ livers assessed at eight months (p=.045). All error bars represent s.e.m.

References

    1. Amir RE, et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nature genetics. 1999;23:185–188.
    1. Bienvenu T, Chelly J. Molecular genetics of Rett syndrome: when DNA methylation goes unrecognized. Nature reviews. 2006;7:415–426.
    1. Guy J, Hendrich B, Holmes M, Martin JE, Bird A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nature genetics. 2001;27:322–326.
    1. Shepherd GM, Katz DM. Synaptic microcircuit dysfunction in genetic models of neurodevelopmental disorders: focus on Mecp2 and Met. Current opinion in neurobiology. 2011;21:827–833.
    1. Kavalali ET, Nelson ED, Monteggia LM. Role of MeCP2, DNA methylation, and HDACs in regulating synapse function. J Neurodev Disord. 2011;3:250–256.
    1. Guy J, Gan J, Selfridge J, Cobb S, Bird A. Reversal of neurological defects in a mouse model of Rett syndrome. Science (New York, N.Y. 2007;315:1143–1147.
    1. Collins AL, et al. Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Human molecular genetics. 2004;13:2679–2689.
    1. Chahrour M, et al. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science (New York, N.Y. 2008;320:1224–1229.
    1. Stancheva I, Collins AL, Van den Veyver IB, Zoghbi H, Meehan RR. A mutant form of MeCP2 protein associated with human Rett syndrome cannot be displaced from methylated DNA by notch in Xenopus embryos. Molecular cell. 2003;12:425–435.
    1. St Johnston D. The art and design of genetic screens: Drosophila melanogaster. Nature reviews. 2002;3:176–188.
    1. Carpinelli MR, et al. Suppressor screen in Mpl−/− mice: c-Myb mutation causes supraphysiological production of platelets in the absence of thrombopoietin signaling. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:6553–6558.
    1. Matera I, et al. A sensitized mutagenesis screen identifies Gli3 as a modifier of Sox10 neurocristopathy. Human molecular genetics. 2008;17:2118–2131.
    1. Justice MJ, Siracusa LD, Stewart AF. Technical approaches for Mouse Models of Human Disease. Disease Models and Mechanisms. 2011;4:305–310.
    1. Derecki NC, et al. Wild-type microglia arrest pathology in a mouse model of Rett syndrome. Nature. 2012;484:105–109.
    1. Neuhaus IM, Beier DR. Efficient localization of mutations by interval haplotype analysis. Mamm Genome. 1998;9:150–154.
    1. Moran JL, et al. Utilization of a whole genome SNP panel for efficient genetic mapping in the mouse. Genome research. 2006;16:436–440.
    1. Fairfield H, et al. Mutation discovery in mice by whole exome sequencing. Genome biology. 2011;12:R86.
    1. Jurevics HA, Kidwai FZ, Morell P. Sources of cholesterol during development of the rat fetus and fetal organs. J Lipid Res. 1997;38:723–733.
    1. Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57:178–201.
    1. Gill S, Stevenson J, Kristiana I, Brown AJ. Cholesterol-dependent degradation of squalene monooxygenase, a control point in cholesterol synthesis beyond HMG-CoA reductase. Cell metabolism. 2011;13:260–273.
    1. Cory EJ, Russey WE, Ortiz de Montellano PR. 2,3-oxidosqualene, an intermediate in the biological synthesis of sterols from squalene. J Am Chem Soc. 1966;88:4750–4751.
    1. Yamamoto S, Bloch K. Studies on squalene epoxidase of rat liver. J Biol Chem. 1970;245:1670–1674.
    1. Shibata N, et al. Supernatant protein factor, which stimulates the conversion of squalene to lanosterol, is a cytosolic squalene transfer protein and enhances cholesterol biosynthesis. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:2244–2249.
    1. Astruc M, Tabacik C, Descomps B, de Paulet AC. Squalene epoxidase and oxidosqualene lanosterol-cyclase activities in cholesterogenic and non-cholesterogenic tissues. Biochim Biophys Acta. 1977;487:204–211.
    1. Ingham PW, Nakano Y, Seger C. Mechanisms and functions of Hedgehog signalling across the metazoa. Nature reviews. 2011;12:393–406.
    1. Pose D, Botella MA. Analysis of the arabidopsis dry2/sqe1-5 mutant suggests a role for sterols in signaling. Plant signaling & behavior. 2009;4:873–874.
    1. Pose D, et al. Identification of the Arabidopsis dry2/sqe1–5 mutant reveals a central role for sterols in drought tolerance and regulation of reactive oxygen species. The Plant journal : for cell and molecular biology. 2009;59:63–76.
    1. Nieweg K, Schaller H, Pfrieger FW. Marked differences in cholesterol synthesis between neurons and glial cells from postnatal rats. J Neurochem. 2009;109:125–134.
    1. Dietschy JM, Turley SD, Spady DK. Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans. J Lipid Res. 1993;34:1637–1659.
    1. Dietschy JM. Central nervous system: cholesterol turnover, brain development and neurodegeneration. Biological chemistry. 2009;390:287–293.
    1. Russell DW, Halford RW, Ramirez DM, Shah R, Kotti T. Cholesterol 24-hydroxylase: an enzyme of cholesterol turnover in the brain. Annual review of biochemistry. 2009;78:1017–1040.
    1. Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nature genetics. 2001;27:327–331.
    1. Pfrieger FW, Ungerer N. Cholesterol metabolism in neurons and astrocytes. Progress in lipid research. 2011;50:357–371.
    1. Xie C, Lund EG, Turley SD, Russell DW, Dietschy JM. Quantitation of two pathways for cholesterol excretion from the brain in normal mice and mice with neurodegeneration. J Lipid Res. 2003;44:1780–1789.
    1. Ko M, et al. Cholesterol-mediated neurite outgrowth is differently regulated between cortical and hippocampal neurons. The Journal of biological chemistry. 2005;280:42759–42765.
    1. Jolley CD, Dietschy JM, Turley SD. Genetic differences in cholesterol absorption in 129/Sv and C57BL/6 mice: effect on cholesterol responsiveness. The American journal of physiology. 1999;276:G1117–G1124.
    1. Bellosta S, Paoletti R, Corsini A. Safety of statins: focus on clinical pharmacokinetics and drug interactions. Circulation. 2004;109:III50–III57.
    1. Garcia-Sabina A, Gulin-Davila J, Sempere-Serrano P, Gonzalez-Juanatey C, Martinez-Pacheco R. Specific considerations on the prescription and therapeutic interchange of statins. Farmacia hospitalaria: organo oficial de expresion cientifica de la Sociedad Espanola de Farmacia Hospitalaria. 2012;36:97–108.
    1. Osterweil EK, et al. Lovastatin corrects excess protein synthesis and prevents epileptogenesis in a mouse model of fragile×syndrome. Neuron. 2013;77:243–250.
    1. Ardern-Holmes SL, North KN. Therapeutics for childhood neurofibromatosis type 1 and type 2. Current treatment options in neurology. 2011;13:529–543.
    1. Keber R, et al. Mouse knockout of the cholesterogenic cytochrome P450 lanosterol 14alpha-demethylase (Cyp51) resembles Antley-Bixler syndrome. The Journal of biological chemistry. 2011;286:29086–29097.
    1. Waterham HR. Defects of cholesterol biosynthesis. FEBS letters. 2006;580:5442–5449.
    1. Bjorkhem I, Hansson M. Cerebrotendinous xanthomatosis: an inborn error in bile acid synthesis with defined mutations but still a challenge. Biochem Biophys Res Commun. 2010;396:46–49.
    1. Lund EG, et al. Knockout of the cholesterol 24-hydroxylase gene in mice reveals a brain-specific mechanism of cholesterol turnover. The Journal of biological chemistry. 2003;278:22980–22988.
    1. Lioy DT, et al. A role for glia in the progression of Rett's syndrome. Nature. 2011;475:497–500.
    1. Vance JE. Dysregulation of cholesterol balance in the brain: contribution to neurodegenerative diseases. Disease models & mechanisms. 2012;5:746–755.
    1. Vance JE, Karten B, Hayashi H. Lipid dynamics in neurons. Biochem Soc Trans. 2006;34:399–403.
    1. Cibickova L. Statins and their influence on brain cholesterol. Journal of clinical lipidology. 2011;5:373–379.
    1. Stranahan AM, Cutler RG, Button C, Telljohann R, Mattson MP. Diet-induced elevations in serum cholesterol are associated with alterations in hippocampal lipid metabolism and increased oxidative stress. J Neurochem. 2011;118:611–615.
    1. Day CP, James OF. Steatohepatitis: a tale of two "hits"? Gastroenterology. 1998;114:842–845.
    1. Fyffe SL, et al. Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response to stress. Neuron. 2008;59:947–958.
    1. Percy AK. Rett syndrome: exploring the autism link. Archives of neurology. 2011;68:985–989.
    1. Lyst MJ, et al. Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT transcriptional co-repressor. Nature Neuroscience. 2013
    1. Ebert DH, et al. Activity-dependent phosphorylation of MECP2 threonine 308 regulates interaction with NcoR. Nature. 2013
    1. Knutson SK, et al. Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks. The EMBO journal. 2008;27:1017–1028.
    1. Sun Z, et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nature medicine. 2012;18:934–942.
    1. Feng D, et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science. 2011;331:1315–1319.
    1. Kile BT, et al. Functional genetic analysis of mouse chromosome 11. Nature. 2003;425:81–86.
    1. McDonald JG, Smith DD, Stiles AR, Russell DW. A comprehensive method for extraction and quantitative analysis of sterols and secosteroids from human plasma. J Lipid Res. 2012;53:1399–1409.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–408.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26:589–595.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079.
    1. Broman KW, Wu H, Sen S, Churchill GA. R/qtl: QTL mapping in experimental crosses. Bioinformatics. 2003;19:889–890.

Source: PubMed

3
Tilaa