Skip to main content

Advertisement

Log in

Allopregnanolone Treatment Improves Plasma Metabolomic Profile Associated with GABA Metabolism in Fragile X-Associated Tremor/Ataxia Syndrome: a Pilot Study

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Currently, there is no effective treatment for the fragile X-associated tremor/ataxia syndrome (FXTAS), a late-onset neurodegenerative disorder. In this pilot study, we evaluated whether allopregnanolone, a natural neurosteroid that exerts beneficial effects in neurodegenerative diseases, nervous system injury, and peripheral neuropathies, could improve lymphocytic bioenergetics and plasma pharmacometabolomics in six males with FXTAS (68 ± 3 years old; FMR1 CGG repeats 94 ± 4; FXTAS stages ranging from 3 to 5) enrolled in a 12-week open-label intervention study conducted at the University of California Davis from December 2015 through July 2016. Plasma pharmacometabolomics and lymphocytic mitochondria function were assessed at baseline (on the day of the first infusion) and at follow-up (within 48 h from the last infusion). In parallel, quantitative measurements of tremor and ataxia and neuropsychological evaluations of mental state, executive function, learning, memory, and psychological symptoms were assessed at the same time points. Allopregnanolone treatment impacted significantly GABA metabolism, oxidative stress, and some of the mitochondria-related outcomes. Notably, the magnitude of the individual metabolic response, as well as the correlation with some of the behavioral tests, was overwhelmingly carrier-specific. Based on this pilot study, allopregnanolone treatment has the potential for improving cognitive and GABA metabolism in FXTAS aligned with the concept of precision medicine.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Mellon SH, Vaudry H (2001) Biosynthesis of neurosteroids and regulation of their synthesis. Int Rev Neurobiol 46:33–78

    Article  CAS  Google Scholar 

  2. Baulieu EE (1991) Neurosteroids: a new function in the brain. Biol Cell 71:3–10

    Article  CAS  Google Scholar 

  3. Agis-Balboa RC, Pinna G, Zhubi A, Maloku E, Veldic M, Costa E, Guidotti A (2006) Characterization of brain neurons that express enzymes mediating neurosteroid biosynthesis. Proc Natl Acad Sci U S A 103:14602–14607. https://doi.org/10.1073/pnas.0606544103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Pelletier G (2010) Steroidogenic enzymes in the brain: morphological aspects. Prog Brain Res 181:193–207. https://doi.org/10.1016/S0079-6123(08)81011-4

    Article  CAS  PubMed  Google Scholar 

  5. Reddy DS (2010) Neurosteroids: endogenous role in the human brain and therapeutic potentials. Prog Brain Res 186:113–137. https://doi.org/10.1016/B978-0-444-53630-3.00008-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Belelli D, Lambert JJ (2005) Neurosteroids: endogenous regulators of the GABA(A) receptor. Nat Rev Neurosci 6:565–575. https://doi.org/10.1038/nrn1703

    Article  CAS  PubMed  Google Scholar 

  7. Schule C, Nothdurfter C, Rupprecht R (2014) The role of allopregnanolone in depression and anxiety. Prog Neurobiol 113:79–87. https://doi.org/10.1016/j.pneurobio.2013.09.003

    Article  CAS  PubMed  Google Scholar 

  8. Schumacher M, Mattern C, Ghoumari A, Oudinet JP, Liere P, Labombarda F, Sitruk-Ware R, De Nicola AF et al (2014) Revisiting the roles of progesterone and allopregnanolone in the nervous system: resurgence of the progesterone receptors. Prog Neurobiol 113:6–39. https://doi.org/10.1016/j.pneurobio.2013.09.004

    Article  CAS  PubMed  Google Scholar 

  9. Langmade SJ, Gale SE, Frolov A, Mohri I, Suzuki K, Mellon SH, Walkley SU, Covey DF et al (2006) Pregnane X receptor (PXR) activation: a mechanism for neuroprotection in a mouse model of Niemann-Pick C disease. Proc Natl Acad Sci U S A 103:13807–13812. https://doi.org/10.1073/pnas.0606218103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pang Y, Dong J, Thomas P (2013) Characterization, neurosteroid binding and brain distribution of human membrane progesterone receptors delta and {epsilon} (mPRdelta and mPR{epsilon}) and mPRdelta involvement in neurosteroid inhibition of apoptosis. Endocrinology 154:283–295. https://doi.org/10.1210/en.2012-1772

    Article  CAS  PubMed  Google Scholar 

  11. Keller EA, Zamparini A, Borodinsky LN, Gravielle MC, Fiszman ML (2004) Role of allopregnanolone on cerebellar granule cells neurogenesis. Brain Res Dev Brain Res 153:13–17. https://doi.org/10.1016/j.devbrainres.2004.07.009

    Article  CAS  PubMed  Google Scholar 

  12. Wang JM, Brinton RD (2008) Allopregnanolone-induced rise in intracellular calcium in embryonic hippocampal neurons parallels their proliferative potential. BMC Neurosci 9(Suppl 2):S11. https://doi.org/10.1186/1471-2202-9-S2-S11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang JM, Johnston PB, Ball BG, Brinton RD (2005) The neurosteroid allopregnanolone promotes proliferation of rodent and human neural progenitor cells and regulates cell-cycle gene and protein expression. J Neurosci 25:4706–4718. https://doi.org/10.1523/JNEUROSCI.4520-04.2005

    Article  CAS  PubMed  Google Scholar 

  14. Karout M, Miesch M, Geoffroy P, Kraft S, Hofmann HD, Mensah-Nyagan AG, Kirsch M (2016) Novel analogs of allopregnanolone show improved efficiency and specificity in neuroprotection and stimulation of proliferation. J Neurochem 139:782–794. https://doi.org/10.1111/jnc.13693

    Article  CAS  PubMed  Google Scholar 

  15. Wang JM, Singh C, Liu L, Irwin RW, Chen S, Chung EJ, Thompson RF, Brinton RD (2010) Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 107:6498–6503. https://doi.org/10.1073/pnas.1001422107

    Article  PubMed  PubMed Central  Google Scholar 

  16. Singh C, Liu L, Wang JM, Irwin RW, Yao J, Chen S, Henry S, Thompson RF et al (2012) Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice. Neurobiol Aging 33:1493–1506. https://doi.org/10.1016/j.neurobiolaging.2011.06.008

    Article  CAS  PubMed  Google Scholar 

  17. Griffin LD, Gong W, Verot L, Mellon SH (2004) Niemann-Pick type C disease involves disrupted neurosteroidogenesis and responds to allopregnanolone. Nat Med 10:704–711. https://doi.org/10.1038/nm1073

    Article  CAS  PubMed  Google Scholar 

  18. He J, Hoffman SW, Stein DG (2004) Allopregnanolone, a progesterone metabolite, enhances behavioral recovery and decreases neuronal loss after traumatic brain injury. Restor Neurol Neurosci 22:19–31

    CAS  PubMed  Google Scholar 

  19. Chen S, Wang JM, Irwin RW, Yao J, Liu L, Brinton RD (2011) Allopregnanolone promotes regeneration and reduces beta-amyloid burden in a preclinical model of Alzheimer's disease. PLoS One 6:e24293. https://doi.org/10.1371/journal.pone.0024293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Djebaili M, Guo Q, Pettus EH, Hoffman SW, Stein DG (2005) The neurosteroids progesterone and allopregnanolone reduce cell death, gliosis, and functional deficits after traumatic brain injury in rats. J Neurotrauma 22:106–118. https://doi.org/10.1089/neu.2005.22.106

    Article  PubMed  Google Scholar 

  21. Sayeed I, Parvez S, Wali B, Siemen D, Stein DG (2009) Direct inhibition of the mitochondrial permeability transition pore: a possible mechanism for better neuroprotective effects of allopregnanolone over progesterone. Brain Res 1263:165–173. https://doi.org/10.1016/j.brainres.2009.01.045

    Article  CAS  PubMed  Google Scholar 

  22. Conde V, Palomar FJ, Lama MJ, Martinez R, Carrillo F, Pintado E, Mir P (2013) Abnormal GABA-mediated and cerebellar inhibition in women with the fragile X premutation. J Neurophysiol 109:1315–1322. https://doi.org/10.1152/jn.00730.2012

    Article  CAS  PubMed  Google Scholar 

  23. Giulivi C, Napoli E, Tassone F, Halmai J, Hagerman R (2016) Plasma biomarkers for monitoring brain pathophysiology in FMR1 premutation carriers. Front Mol Neurosci 9:71. https://doi.org/10.3389/fnmol.2016.00071

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Giulivi C, Napoli E, Tassone F, Halmai J, Hagerman R (2016) Plasma metabolic profile delineates roles for neurodegeneration, pro-inflammatory damage and mitochondrial dysfunction in the FMR1 premutation. Biochem J 473:3871–3888. https://doi.org/10.1042/BCJ20160585

    Article  CAS  PubMed  Google Scholar 

  25. D'Hulst C, Heulens I, Brouwer JR, Willemsen R, De Geest N, Reeve SP, De Deyn PP, Hassan BA et al (2009) Expression of the GABAergic system in animal models for fragile X syndrome and fragile X associated tremor/ataxia syndrome (FXTAS). Brain Res 1253:176–183. https://doi.org/10.1016/j.brainres.2008.11.075

    Article  CAS  PubMed  Google Scholar 

  26. Hagerman RJ, Leehey M, Heinrichs W, Tassone F, Wilson R, Hills J, Grigsby J, Gage B et al (2001) Intention tremor, parkinsonism, and generalized brain atrophy in male carriers of fragile X. Neurology 57:127–130

    Article  CAS  Google Scholar 

  27. Berry-Kravis E, Abrams L, Coffey SM, Hall DA, Greco C, Gane LW, Grigsby J, Bourgeois JA et al (2007) Fragile X-associated tremor/ataxia syndrome: clinical features, genetics, and testing guidelines. Mov Disord 22:2018–2030, quiz 2140. https://doi.org/10.1002/mds.21493

    Article  PubMed  Google Scholar 

  28. Cao Z, Hulsizer S, Tassone F, Tang HT, Hagerman RJ, Rogawski MA, Hagerman PJ, Pessah IN (2012) Clustered burst firing in FMR1 premutation hippocampal neurons: amelioration with allopregnanolone. Hum Mol Genet 21:2923–2935. https://doi.org/10.1093/hmg/dds118

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hall D, Todorova-Koteva K, Pandya S, Bernard B, Ouyang B, Walsh M, Pounardjian T, Deburghraeve C et al (2016) Neurological and endocrine phenotypes of fragile X carrier women. Clin Genet 89:60–67. https://doi.org/10.1111/cge.12646

    Article  CAS  PubMed  Google Scholar 

  30. Kaddurah-Daouk R, Weinshilboum RM, Pharmacometabolomics Research N (2014) Pharmacometabolomics: implications for clinical pharmacology and systems pharmacology. Clin Pharmacol Ther 95:154–167. https://doi.org/10.1038/clpt.2013.217

    Article  CAS  PubMed  Google Scholar 

  31. Zhu H, Bogdanov MB, Boyle SH, Matson W, Sharma S, Matson S, Churchill E, Fiehn O et al (2013) Pharmacometabolomics of response to sertraline and to placebo in major depressive disorder—possible role for methoxyindole pathway. PLoS One 8:–e68283. https://doi.org/10.1371/journal.pone.0068283

    Article  CAS  Google Scholar 

  32. Wang J-Y, Trivedi AM, Yang J, Schneider A, Giulivi C, Adams P, Tassone F, Kim K et al (2017) Open label allopregnanolone treatment of men with FXTAS. Neurotherapeutics 14:1073–1083. https://doi.org/10.1007/s13311-017-0555-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Seritan AL, Nguyen DV, Mu Y, Tassone F, Bourgeois JA, Schneider A, Cogswell JB, Cook KR et al (2014) Memantine for fragile X-associated tremor/ataxia syndrome: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry 75:264–271. https://doi.org/10.4088/JCP.13m08546

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Filipovic-Sadic S, Sah S, Chen L, Krosting J, Sekinger E, Zhang W, Hagerman PJ, Stenzel TT et al (2010) A novel FMR1 PCR method for the routine detection of low abundance expanded alleles and full mutations in fragile X syndrome. Clin Chem 56:399–408. https://doi.org/10.1373/clinchem.2009.136101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Pretto D, Yrigollen CM, Tang HT, Williamson J, Espinal G, Iwahashi CK, Durbin-Johnson B, Hagerman RJ et al (2014) Clinical and molecular implications of mosaicism in FMR1 full mutations. Front Genet 5:318. https://doi.org/10.3389/fgene.2014.00318

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Song G, Napoli E, Wong S, Hagerman R, Liu S, Tassone F, Giulivi C (2016) Altered redox mitochondrial biology in the neurodegenerative disorder fragile X-tremor/ataxia syndrome: use of antioxidants in precision medicine. Mol Med 22:1. https://doi.org/10.2119/molmed.2016.00122

    Article  CAS  Google Scholar 

  37. Giulivi C, Zhang YF, Omanska-Klusek A, Ross-Inta C, Wong S, Hertz-Picciotto I, Tassone F, Pessah IN (2010) Mitochondrial dysfunction in autism. Jama 304:2389–2396. https://doi.org/10.1001/jama.2010.1706

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xia J, Sinelnikov IV, Han B, Wishart DS (2015) MetaboAnalyst 3.0—making metabolomics more meaningful. Nucleic Acids Res 43:W251–W257. https://doi.org/10.1093/nar/gkv380

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Szumilas M (2010) Explaining odds ratios. J Can Acad Child Adolesc Psychiatry 19:227–229

    Article  Google Scholar 

  40. Gibson KM, Hoffmann GF, Hodson AK, Bottiglieri T, Jakobs C (1998) 4-Hydroxybutyric acid and the clinical phenotype of succinic semialdehyde dehydrogenase deficiency, an inborn error of GABA metabolism. Neuropediatrics 29:14–22. https://doi.org/10.1055/s-2007-973527

    Article  CAS  PubMed  Google Scholar 

  41. Gibson KM, Aramaki S, Sweetman L, Nyhan WL, DeVivo DC, Hodson AK, Jakobs C (1990) Stable isotope dilution analysis of 4-hydroxybutyric acid: an accurate method for quantification in physiological fluids and the prenatal diagnosis of 4-hydroxybutyric aciduria. Biomed Environ Mass Spectrom 19:89–93. https://doi.org/10.1002/bms.1200190207

    Article  CAS  PubMed  Google Scholar 

  42. Brown GK, Cromby CH, Manning NJ, Pollitt RJ (1987) Urinary organic acids in succinic semialdehyde dehydrogenase deficiency: evidence of alpha-oxidation of 4-hydroxybutyric acid, interaction of succinic semialdehyde with pyruvate dehydrogenase and possible secondary inhibition of mitochondrial beta-oxidation. J Inherit Metab Dis 10:367–375. https://doi.org/10.1007/BF01799979

    Article  CAS  PubMed  Google Scholar 

  43. Bernasconi R, Mathivet P, Bischoff S, Marescaux C (1999) Gamma-hydroxybutyric acid: an endogenous neuromodulator with abuse potential? Trends Pharmacol Sci 20:135–141

    Article  CAS  Google Scholar 

  44. Maitre M, Andriamampandry C, Kemmel V, Schmidt C, Hode Y, Hechler V, Gobaille S (2000) Gamma-hydroxybutyric acid as a signaling molecule in brain. Alcohol 20:277–283

    Article  CAS  Google Scholar 

  45. Karu N, McKercher C, Nichols DS, Davies N, Shellie RA, Hilder EF, Jose MD (2016) Tryptophan metabolism, its relation to inflammation and stress markers and association with psychological and cognitive functioning: Tasmanian Chronic Kidney Disease pilot study. BMC Nephrol 17:171. https://doi.org/10.1186/s12882-016-0387-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. de Bruin NM, Kruse CG (2015) 5-HT6 receptor antagonists: potential efficacy for the treatment of cognitive impairment in schizophrenia. Curr Pharm Des 21:3739–3759

    Article  Google Scholar 

  47. Kasten CR, Boehm SL 2nd (2015) Identifying the role of pre-and postsynaptic GABA(B) receptors in behavior. Neurosci Biobehav Rev 57:70–87. https://doi.org/10.1016/j.neubiorev.2015.08.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gall WE, Beebe K, Lawton KA, Adam KP, Mitchell MW, Nakhle PJ, Ryals JA, Milburn MV et al (2010) Alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One 5:e10883. https://doi.org/10.1371/journal.pone.0010883

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lord RS, Bralley JA (2008) Clinical applications of urinary organic acids. Part I: detoxification markers. Altern Med Rev 13:205–215

    PubMed  Google Scholar 

  50. Capuron L, Schroecksnadel S, Feart C, Aubert A, Higueret D, Barberger-Gateau P, Laye S, Fuchs D (2011) Chronic low-grade inflammation in elderly persons is associated with altered tryptophan and tyrosine metabolism: role in neuropsychiatric symptoms. Biol Psychiatry 70:175–182. https://doi.org/10.1016/j.biopsych.2010.12.006

    Article  CAS  PubMed  Google Scholar 

  51. Dantzer R, O'Connor JC, Lawson MA, Kelley KW (2011) Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology 36:426–436. https://doi.org/10.1016/j.psyneuen.2010.09.012

    Article  CAS  PubMed  Google Scholar 

  52. Napoli E, Song G, Wong S, Hagerman R, Giulivi C (2016) Altered bioenergetics in primary dermal fibroblasts from adult carriers of the FMR1 premutation before the onset of the neurodegenerative disease fragile X-associated tremor/ataxia syndrome. Cerebellum 15:552–564. https://doi.org/10.1007/s12311-016-0779-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Myint AM, Kim YK (2014) Network beyond IDO in psychiatric disorders: revisiting neurodegeneration hypothesis. Prog Neuro-Psychopharmacol Biol Psychiatry 48:304–313. https://doi.org/10.1016/j.pnpbp.2013.08.008

    Article  CAS  Google Scholar 

  54. Armstrong MD (1979) N-delta-acetylornithine and S-methylcysteine in blood plasma. Biochim Biophys Acta 587:638–642

    Article  CAS  Google Scholar 

  55. McClay JL, Vunck SA, Batman AM, Crowley JJ, Vann RE, Beardsley PM, van den Oord EJ (2015) Neurochemical metabolomics reveals disruption to sphingolipid metabolism following chronic haloperidol administration. J NeuroImmune Pharmacol 10:425–434. https://doi.org/10.1007/s11481-015-9605-1

    Article  PubMed  PubMed Central  Google Scholar 

  56. Leucht S, Cipriani A, Spineli L, Mavridis D, Orey D, Richter F, Samara M, Barbui C et al (2013) Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet 382:951–962. https://doi.org/10.1016/S0140-6736(13)60733-3

    Article  CAS  PubMed  Google Scholar 

  57. Marsden CD, Jenner P (1980) The pathophysiology of extrapyramidal side-effects of neuroleptic drugs. Psychol Med 10:55–72

    Article  CAS  Google Scholar 

  58. Yang J, Chen T, Sun L, Zhao Z, Qi X, Zhou K, Cao Y, Wang X et al (2013) Potential metabolite markers of schizophrenia. Mol Psychiatry 18:67–78. https://doi.org/10.1038/mp.2011.131

    Article  CAS  PubMed  Google Scholar 

  59. Song G, Napoli E, Wong S, Hagerman R, Liu S, Tassone F, Giulivi C (2016) Altered redox mitochondrial biology in the neurodegenerative disorder fragile X-tremor/ataxia syndrome: use of antioxidants in precision medicine. Mol Med 22:1. https://doi.org/10.2119/molmed.2016.00122

    Article  CAS  Google Scholar 

  60. Brinton RD (2016) Neuroendocrinology: oestrogen therapy affects brain structure but not function. Nat Rev Neurol 12:561–562. https://doi.org/10.1038/nrneurol.2016.147

    Article  CAS  PubMed  Google Scholar 

  61. Kleppner SR, Tobin AJ (2001) GABA signalling: therapeutic targets for epilepsy, Parkinson's disease and Huntington's disease. Expert Opin Ther Targets 5:219–239. https://doi.org/10.1517/14728222.5.2.219

    Article  CAS  PubMed  Google Scholar 

  62. Adams PE, Adams JS, Nguyen DV, Hessl D, Brunberg JA, Tassone F, Zhang W, Koldewyn K et al (2010) Psychological symptoms correlate with reduced hippocampal volume in fragile X premutation carriers. Am J Med Genet B Neuropsychiatr Genet 153B:775–785. https://doi.org/10.1002/ajmg.b.31046

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kim JM, Kim DH, Lee Y, Park SJ, Ryu JH (2014) Distinct roles of the hippocampus and perirhinal cortex in GABAA receptor blockade-induced enhancement of object recognition memory. Brain Res 1552:17–25. https://doi.org/10.1016/j.brainres.2014.01.024

    Article  CAS  PubMed  Google Scholar 

  64. Prut L, Prenosil G, Willadt S, Vogt K, Fritschy JM, Crestani F (2010) A reduction in hippocampal GABAA receptor alpha5 subunits disrupts the memory for location of objects in mice. Genes Brain Behav 9:478–488. https://doi.org/10.1111/j.1601-183X.2010.00575.x

    Article  CAS  PubMed  Google Scholar 

  65. Lewis DA, Volk DW, Hashimoto T (2004) Selective alterations in prefrontal cortical GABA neurotransmission in schizophrenia: a novel target for the treatment of working memory dysfunction. Psychopharmacology 174:143–150. https://doi.org/10.1007/s00213-003-1673-x

    Article  CAS  PubMed  Google Scholar 

  66. Reis J, Cohen LG, Pearl PL, Fritsch B, Jung NH, Dustin I, Theodore WH (2012) GABAB-ergic motor cortex dysfunction in SSADH deficiency. Neurology 79:47–54. https://doi.org/10.1212/WNL.0b013e31825dcf71

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rodriguez-Revenga L, Madrigal I, Pagonabarraga J, Xuncla M, Badenas C, Kulisevsky J, Gomez B, Mila M (2009) Penetrance of FMR1 premutation associated pathologies in fragile X syndrome families. Eur J Hum Genet 17:1359–1362. https://doi.org/10.1038/ejhg.2009.51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bishnoi M, Chopra K, Kulkarni SK (2008) Progesterone attenuates neuroleptic-induced orofacial dyskinesia via the activity of its metabolite, allopregnanolone, a positive GABA(A) modulating neurosteroid. Prog Neuro-Psychopharmacol Biol Psychiatry 32:451–461. https://doi.org/10.1016/j.pnpbp.2007.09.017

    Article  CAS  Google Scholar 

  69. Singh A, Kumar A (2008) Possible GABAergic modulation in the protective effect of allopregnanolone on sleep deprivation-induced anxiety-like behavior and oxidative damage in mice. Methods Find Exp Clin Pharmacol 30:681–689. https://doi.org/10.1358/mf.2008.30.9.1186076

    Article  CAS  PubMed  Google Scholar 

  70. Tamazian G, Ho Chang J, Knyazev S, Stepanov E, Kim KJ, Porozov Y (2015) Modeling conformational redox-switch modulation of human succinic semialdehyde dehydrogenase. Proteins 83:2217–2229. https://doi.org/10.1002/prot.24937

    Article  CAS  PubMed  Google Scholar 

  71. Murphy TC, Amarnath V, Gibson KM, Picklo MJ Sr (2003) Oxidation of 4-hydroxy-2-nonenal by succinic semialdehyde dehydrogenase (ALDH5A). J Neurochem 86:298–305

    Article  CAS  Google Scholar 

  72. Picklo MJ, Montine TJ, Amarnath V, Neely MD (2002) Carbonyl toxicology and Alzheimer's disease. Toxicol Appl Pharmacol 184:187–197

    Article  CAS  Google Scholar 

  73. Napoli E, Ross-Inta C, Wong S, Omanska-Klusek A, Barrow C, Iwahashi C, Garcia-Arocena D, Sakaguchi D et al (2011) Altered zinc transport disrupts mitochondrial protein processing/import in fragile X-associated tremor/ataxia syndrome. Hum Mol Genet 20:3079–3092. https://doi.org/10.1093/hmg/ddr211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kennedy AD, Pappan KL, Donti TR, Evans AM, Wulff JE, Miller LAD, Reid Sutton V, Sun Q et al (2017) Elucidation of the complex metabolic profile of cerebrospinal fluid using an untargeted biochemical profiling assay. Mol Genet Metab 121:83–90. https://doi.org/10.1016/j.ymgme.2017.04.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Trushina E, Dutta T, Persson XM, Mielke MM, Petersen RC (2013) Identification of altered metabolic pathways in plasma and CSF in mild cognitive impairment and Alzheimer's disease using metabolomics. PLoS One 8:e63644. https://doi.org/10.1371/journal.pone.0063644

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Griffin JL, Salek RM (2007) Metabolomic applications to neuroscience: more challenges than chances? Expert Rev Proteomics 4:435–437. https://doi.org/10.1586/14789450.4.4.435

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank the carriers that participated in this study. We thank Drs. Roberta Brinton and Robert Irwin for their protocol guidance and expertise regarding allopregnanolone treatment and for providing the sulfobutylether-β-cyclodextrin for this study. We would like to thank Dr. Daniel Tancredi (Department of Pediatrics, UC Davis School of Medicine) for his excellent contribution to the statistical analysis of this study. We also thank Ms. Catherine Ross-Inta, Gyu Song, and Ilaria Marsilio for their valuable technical assistance and Dr. Gerhart Bauer for his expertise in preparing the allopregnanolone solutions. This study was made possible by private donations to FXTAS research, NICHD grant HD036071, and the MIND Institute IDDRC U54HD079125.

Author information

Authors and Affiliations

Authors

Contributions

EN assessed most of the mitochondrial outcomes and helped in drafting, edited, and approved the final version of the manuscript; AS carried out cognitive and psychological testing on the patients, revised the manuscript, and approved the final manuscript; JW acquired neuroimaging data, performed statistical analysis, interpreted the results, and reviewed and approved the final version of the manuscript; AT and NRC analyzed neuropsychiatric tests and reviewed and approved the final version of the manuscript; FT provided lymphocytes, performed the genotyping, and revised and approved the manuscript; MR provided expertise in intravenous injection of allopregnanolone, reviewed the manuscript, and approved its final version; RJH carried out clinical assessment of these subjects, wrote clinical findings, and revised and approved the final version of the manuscript; and CG conceptualized the study, designed the experiments, analyzed the metabolomics data, and wrote the manuscript.

Corresponding author

Correspondence to Cecilia Giulivi.

Ethics declarations

Conflict of Interest

RJH has received funding from Novartis, Roche/Genentech, Alcobra, and Neuren for treatment trials in fragile X syndrome, autism, and Down syndrome. She has also consulted with Novartis, Fulcrum, Zynerba, and Roche/Genentech regarding treatment for fragile X syndrome. The other authors have no conflicts of financial interest relevant to this article to disclose.

Electronic supplementary material

ESM 1

(XLSX 32 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Napoli, E., Schneider, A., Wang, J.Y. et al. Allopregnanolone Treatment Improves Plasma Metabolomic Profile Associated with GABA Metabolism in Fragile X-Associated Tremor/Ataxia Syndrome: a Pilot Study. Mol Neurobiol 56, 3702–3713 (2019). https://doi.org/10.1007/s12035-018-1330-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1330-3

Keywords

Navigation