Metabolic fingerprints of altered brain growth, osmoregulation and neurotransmission in a Rett syndrome model

Angèle Viola, Véronique Saywell, Laurent Villard, Patrick J Cozzone, Norbert W Lutz, Angèle Viola, Véronique Saywell, Laurent Villard, Patrick J Cozzone, Norbert W Lutz

Abstract

Background: Rett syndrome (RS) is the leading cause of profound mental retardation of genetic origin in girls. Since RS is mostly caused by mutations in the MECP2 gene, transgenic animal models such as the Mecp2-deleted ("Mecp2-null") mouse have been employed to study neurological symptoms and brain function. However, an interdisciplinary approach drawing from chemistry, biology and neuroscience is needed to elucidate the mechanistic links between the genotype and phenotype of this genetic disorder.

Methodology/principal findings: We performed, for the first time, a metabolomic study of brain extracts from Mecp2-null mice by using high-resolution magnetic resonance spectroscopy. A large number of individual water-soluble metabolites and phospholipids were quantified without prior selection for specific metabolic pathways. Results were interpreted in terms of Mecp2 gene deletion, brain cell function and brain morphology. This approach provided a "metabolic window" to brain characteristics in Mecp2-null mice (n = 4), revealing (i) the first metabolic evidence of astrocyte involvement in RS (decreased levels of the astrocyte marker, myo-inositol, vs. wild-type mice; p = 0.034); (ii) reduced choline phospholipid turnover in Mecp2-null vs. wild-type mice, implying a diminished potential of cells to grow, paralleled by globally reduced brain size and perturbed osmoregulation; (iii) alterations of the platelet activating factor (PAF) cycle in Mecp2-null mouse brains, where PAF is a bioactive lipid acting on neuronal growth, glutamate exocytosis and other processes; and (iv) changes in glutamine/glutamate ratios (p = 0.034) in Mecp2-null mouse brains potentially indicating altered neurotransmitter recycling.

Conclusions/significance: This study establishes, for the first time, detailed metabolic fingerprints of perturbed brain growth, osmoregulation and neurotransmission in a mouse model of Rett syndrome. Combined with morphological and neurological findings, these results are crucial elements in providing mechanistic links between genotype and phenotype of Rett syndrome. Ultimately, this information can be used to identify novel molecular targets for pharmacological RS treatment.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1
Figure 1
Typical high-resolution 1H NMR spectrum of the brain extract from a Mecp2-deficient mouse (water-soluble metabolites). The entire spectral region of interest (upper left spectrum) as well as magnified subregions are shown. Creatinine (Crn) was not quantified since the weak singlet was not sufficiently separated from other peaks forming a broad hump between 3.01 and 3.06 ppm. Abbreviations: ac, acetate; ala, alanine; asp, aspartate; Cho, choline; Cr, creatine; GABA, γ-aminobutyrate; gln, glutamine; glu, glutamate; gly, glycine; GPC, glycerophosphocholine; lac, lactate; myo-Ins, myo-inositol; NAA, N-acetylaspartate; NANA, N-acetylneuraminate; PC, phosphocholine; scyllo-Ins, scyllo-inositol; suc, succinate; tau, taurine.
Figure 2
Figure 2
Typical high-resolution 31P NMR spectrum of the brain extract from a Mecp2-deficient mouse (phospholipids). The entire PL region (upper left spectrum) as well as magnified subregions are shown. Processing parameters were varied to highlight important spectral details. Abbreviations: AAPtdC, alkyl-acyl-phosphatidylcholine; AAPtdE, alkyl-acyl-phosphatidylethanolamine; CL, cardiolipin; PLx,y, unassigned phospholipids; PtdI, phosphatidylinositol; PtdA, phosphatidic acid; PtdC, (diacyl)phosphatidylcholine; PtdCplasm, choline plasmalogen; PtdE, (diacyl)phosphatidylethanolamine; PtdEplasm, ethanolamine plasmalogen; PtdG, phosphatidylglycerol; PtdS, phosphatidylserine; SM, sphingomyelin.

References

    1. Rett A. Über ein eigenartiges hirnatrophisches Syndrom bei Hyperammonämie im Kindesalter. Wien Med Wochenschr. 1966;116:723–726.
    1. Hagberg B, Aicardi J, Dias K, Ramos O. A progressive syndrome of autism, dementia, ataxia, and loss of purposeful hand use in girls: Rett's syndrome: report of 35 cases. Ann Neurol. 1983;14:471–479.
    1. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23:185–188.
    1. Shahbazian MD, Zoghbi HY. Rett syndrome and MeCP2: linking epigenetics and neuronal function. Am J Hum Genet. 2002;71:1259–1272.
    1. Zoghbi HY. MeCP2 dysfunction in humans and mice. J Child Neurol. 2005;20:736–740.
    1. Hagberg B, Witt-Engerstrom I. Rett syndrome: a suggested staging system for describing impairment profile with increasing age towards adolescence. Am J Med Genet Suppl. 1986;1:47–59.
    1. Armstrong DD. Rett syndrome neuropathology review 2000. Brain Dev. 2001;23(Suppl 1):S72–76.
    1. Reiss AL, Faruque F, Naidu S, Abrams M, Beaty T, et al. Neuroanatomy of Rett syndrome: a volumetric imaging study. Ann Neurol. 1993;34:227–234.
    1. Subramaniam B, Naidu S, Reiss AL. Neuroanatomy in Rett syndrome: cerebral cortex and posterior fossa. Neurology. 1997;48:399–407.
    1. Hagberg B, Hanefeld F, Percy A, Skjeldal O. An update on clinically applicable diagnostic criteria in Rett syndrome. Comments to Rett Syndrome Clinical Criteria Consensus Panel Satellite to European Paediatric Neurology Society Meeting, Baden Baden, Germany, 11 September 2001. Eur J Paediatr Neurol. 2002;6:293–297.
    1. Hagberg G, Stenbom Y, Witt Engerstrom I. Head growth in Rett syndrome. Acta Paediatr. 2000;89:198–202.
    1. Gotoh H, Suzuki I, Maruki K, Mitomo M, Hirasawa K, et al. Magnetic resonance imaging and clinical findings examined in adulthood-studies on three adults with Rett syndrome. Brain Dev. 2001;23(Suppl 1):S118–121.
    1. Dani VS, Chang Q, Maffei A, Turrigiano GG, Jaenisch R, et al. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc Natl Acad Sci U S A. 2005;102:12560–12565.
    1. Guy J, Hendrich B, Holmes M, Martin JE, Bird A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat Genet. 2001;27:322–326.
    1. Horike S, Cai S, Miyano M, Cheng JF, Kohwi-Shigematsu T. Loss of silent-chromatin looping and impaired imprinting of DLX5 in Rett syndrome. Nat Genet. 2005;37:31–40.
    1. Nuber UA, Kriaucionis S, Roloff TC, Guy J, Selfridge J, et al. Up-regulation of glucocorticoid-regulated genes in a mouse model of Rett syndrome. Hum Mol Genet. 2005;14:2247–2256.
    1. Nelson ED, Kavalali ET, Monteggia LM. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr Biol. 2006;16:710–716.
    1. Viemari JC, Roux JC, Tryba AK, Saywell V, Burnet H, et al. Mecp2 deficiency disrupts norepinephrine and respiratory systems in mice. J Neurosci. 2005;25:11521–11530.
    1. Nagai K, Miyake K, Kubota T. A transcriptional repressor MeCP2 causing Rett syndrome is expressed in embryonic non-neuronal cells and controls their growth. Brain Res Dev Brain Res. 2005;157:103–106.
    1. Saywell V, Viola A, Confort-Gouny S, Le Fur Y, Villard L, et al. Brain magnetic resonance study of Mecp2 deletion effects on anatomy and metabolism. Biochem Biophys Res Commun. 2006;340:776–783.
    1. Gokcay A, Kitis O, Ekmekci O, Karasoy H, Sener RN. Proton MR spectroscopy in Rett syndrome. Comput Med Imaging Graph. 2002;26:271–275.
    1. Naidu S, Kaufmann WE, Abrams MT, Pearlson GD, Lanham DC, et al. Neuroimaging studies in Rett syndrome. Brain Dev. 2001;23(Suppl 1):S62–71.
    1. Pan JW, Lane JB, Hetherington H, Percy AK. Rett syndrome: 1H spectroscopic imaging at 4.1 Tesla. J Child Neurol. 1999;14:524–528.
    1. Ham AL, Kumar A, Deeter R, Schanen NC. Does genotype predict phenotype in Rett syndrome? J Child Neurol. 2005;20:768–778.
    1. Lutz NW. From metabolic to metabolomic NMR spectroscopy of apoptotic cells. Metabolomics. 2005;1:251–268.
    1. Purves D, Augustine GJ, Fitzpatrick D, Hall WC, Lamantia AS, et al., editors. Neuroscience. ed: Sinauer Associates; 2001. 2nd ed.
    1. Viola A, Chabrol B, Nicoli F, Confort-Gouny S, Viout P, et al. Magnetic resonance spectroscopy study of glycine pathways in nonketotic hyperglycinemia. Pediatr Res. 2002;52:292–300.
    1. Baslow MH. Evidence supporting a role for N-acetyl-L-aspartate as a molecular water pump in myelinated neurons in the central nervous system. An analytical review. Neurochem Int. 2002;40:295–300.
    1. Macala LJ, Yohe HC. In situ accessibility of murine macrophage gangliosides. Glycobiology. 1995;5:67–75.
    1. Wu G, Lu ZH, Wang J, Wang Y, Xie X, et al. Enhanced susceptibility to kainate-induced seizures, neuronal apoptosis, and death in mice lacking gangliotetraose gangliosides: protection with LIGA 20, a membrane-permeant analog of GM1. J Neurosci. 2005;25:11014–11022.
    1. Lekman AY, Hagberg BA, Svennerholm LT. Membrane cerebral lipids in Rett syndrome. Pediatr Neurol. 1991;7:186–190.
    1. Lekman AY, Hagberg BA, Svennerholm LT. Cerebrospinal fluid gangliosides in patients with Rett syndrome and infantile neuronal ceroid lipofuscinosis. Eur J Paediatr Neurol. 1999;3:119–123.
    1. Papadimitriou JM, Hockey A, Tan N, Masters CL. Rett syndrome: abnormal membrane-bound lamellated inclusions in neurons and oligodendroglia. Am J Med Genetics. 1988;29:365–368.
    1. Baburina I, Jackowski S. Cellular responses to excess phospholipid. J Biol Chem. 1999;274:9400–9408.
    1. Zhang XH, Zhao C, Seleznev K, Song K, Manfredi JJ, et al. Disruption of G1-phase phospholipid turnover by inhibition of Ca2+-independent phospholipase A2 induces a p53-dependent cell-cycle arrest in G1 phase. J Cell Sci. 2006;119:1005–1015.
    1. Johnston MV, Hohmann C, Blue ME. Neurobiology of Rett syndrome. Neuropediatrics. 1995;26:119–122.
    1. Westergaard N, Fosmark H, Schousboe A. Metabolism and release of glutamate in cerebellar granule cells cocultured with astrocytes from cerebellum or cerebral cortex. J Neurochem. 1991;56:59–66.
    1. Kimelberg HK. The problem of astrocyte identity. Neurochem Int. 2004;45:191–202.
    1. Bazan NG. Synaptic lipid signaling: significance of polyunsaturated fatty acids and platelet-activating factor. J Lipid Res. 2003;44:2221–2233.
    1. Johnston MV, Jeon OH, Pevsner J, Blue ME, Naidu S. Neurobiology of Rett syndrome: a genetic disorder of synapse development. Brain Dev 23 Suppl. 2001;1:S206–213.
    1. Segovia G, Del Arco A, Mora F. Role of glutamate receptors and glutamate transporters in the regulation of the glutamate-glutamine cycle in the awake rat. Neurochem Res. 1999;24:779–783.
    1. Stanimirovic DB, Ball R, Durkin JP. Stimulation of glutamate uptake and Na,K-ATPase activity in rat astrocytes exposed to ischemia-like insults. Glia. 1997;19:123–134.
    1. Newsholme P, Procopio J, Lima MM, Pithon-Curi TC, Curi R. Glutamine and glutamate–their central role in cell metabolism and function. Cell Biochem Funct. 2003;21:1–9.
    1. Shokati T, Zwingmann C, Leibfritz D. Contribution of extracellular glutamine as an anaplerotic substrate to neuronal metabolism: a re-evaluation by multinuclear NMR spectroscopy in primary cultured neurons. Neurochem Res. 2005;30:1269–1281.
    1. Zwingmann C, Shokati T, Butterworth RF, Leibfritz D. Use of NMR spectroscopy for the study of ammonia metabolism in astrocytes and neurons: role of glutamine synthesis in astrocytes. Neurochem Res. in press.
    1. Patel AB, de Graaf RA, Mason GF, Rothman DL, Shulman RG, et al. The contribution of GABA to glutamate/glutamine cycling and energy metabolism in the rat cortex in vivo. Proc Natl Acad Sci U S A. 2005;102:5588–5593.
    1. Brand A, Richter-Landsberg C, Leibfritz D. Multinuclear NMR studies on the energy metabolism of glial and neuronal cells. Dev Neurosci. 1993;15:289–298.
    1. Kreis R, Ernst T, Ross BD. Development of the human brain: in vivo quantification of metabolite and water content with proton magnetic resonance spectroscopy. Magn Reson Med. 1993;30:424–437.
    1. Picker JD, Yang R, Ricceri L, Berger-Sweeney J. An altered neonatal behavioral phenotype in Mecp2 mutant mice. Neuroreport. 2006;17:541–544.
    1. Lutz NW, Tome ME, Aiken NR, Briehl MM. Changes in phosphate metabolism in thymoma cells suggest mechanisms for resistance to dexamethasone-induced apoptosis. A 31P NMR spectroscopic study of cell extracts. NMR Biomed. 2002;15:356–366.
    1. Lilliefors HW. The Kolmogorov-Smirnov test for normality with mean and variance unknown. J Amer Stat Assn. 1967;62:399–402.
    1. Sokal RR, Rohlf FJ. Biometry. New York: Freeman; 1995.
    1. Tome ME, Lutz NW, Briehl MM. Overexpression of catalase or Bcl-2 delays or prevents alterations in phospholipid metabolism during glucocorticoid-induced apoptosis in WEHI7.2 cells. Biochim Biophys Acta. 2003;1642:149–162.
    1. Lykidis A, Baburina I, Jackowski S. Distribution of CTP:phosphocholine cytidylyltransferase (CCT) isoforms. Identification of a new CCTbeta splice variant. J Biol Chem. 1999;274:26992–27001.
    1. Rice SQ, Southan C, Boyd HF, Terrett JA, MacPhee CH, et al. Expression, purification and characterization of a human serine-dependent phospholipase A2 with high specificity for oxidized phospholipids and platelet activating factor. Biochem J. 1998;330(Pt 3):1309–1315.

Source: PubMed

3
Suscribir