Lovastatin improves impaired synaptic plasticity and phasic alertness in patients with neurofibromatosis type 1

Florian Mainberger, Nikolai H Jung, Martin Zenker, Ute Wahlländer, Leonie Freudenberg, Susanne Langer, Steffen Berweck, Tobias Winkler, Andreas Straube, Florian Heinen, Sofia Granström, Victor-Felix Mautner, Karen Lidzba, Volker Mall, Florian Mainberger, Nikolai H Jung, Martin Zenker, Ute Wahlländer, Leonie Freudenberg, Susanne Langer, Steffen Berweck, Tobias Winkler, Andreas Straube, Florian Heinen, Sofia Granström, Victor-Felix Mautner, Karen Lidzba, Volker Mall

Abstract

Background: Neurofibromatosis type 1 (NF1) is one of the most common genetic disorders causing learning disabilities by mutations in the neurofibromin gene, an important inhibitor of the RAS pathway. In a mouse model of NF1, a loss of function mutation of the neurofibromin gene resulted in increased gamma aminobutyric acid (GABA)-mediated inhibition which led to decreased synaptic plasticity and deficits in attentional performance. Most importantly, these defictis were normalized by lovastatin. This placebo-controlled, double blind, randomized study aimed to investigate synaptic plasticity and cognition in humans with NF1 and tried to answer the question whether potential deficits may be rescued by lovastatin.

Methods: In NF1 patients (n = 11; 19-44 years) and healthy controls (HC; n = 11; 19-31 years) paired pulse transcranial magnetic stimulation (TMS) was used to study intracortical inhibition (paired pulse) and synaptic plasticity (paired associative stimulation). On behavioural level the Test of Attentional Performance (TAP) was used. To study the effect of 200 mg lovastatin for 4 days on all these parameters, a placebo-controlled, double blind, randomized trial was performed.

Results: In patients with NF1, lovastatin revealed significant decrease of intracortical inhibition, significant increase of synaptic plasticity as well as significant increase of phasic alertness. Compared to HC, patients with NF1 exposed increased intracortical inhibition, impaired synaptic plasticity and deficits in phasic alertness.

Conclusions: This study demonstrates, for the first time, a link between a pathological RAS pathway activity, intracortical inhibition and impaired synaptic plasticity and its rescue by lovastatin in humans. Our findings revealed mechanisms of attention disorders in humans with NF1 and support the idea of a potential clinical benefit of lovastatin as a therapeutic option.

Figures

Figure 1
Figure 1
Synaptic plasticity in patients with NF1 and controls. A) Depicted is the time course of MEP amplitudes after PAS in healthy controls and patients with NF1. PAS led to a significant MEP increase only in the control group. Abscissa pictures point in time and ordinate mean MEP amplitude. Asterisks indicate significant differences between two groups or points in time (p < 0.05, unpaired t test), error bars represent ± standard error of the mean. B) MEP amplitudes from two representative subjects of controls and NF1before and at POST 1 after PAS. Shown are averages of 20 MEP trials in each case.
Figure 2
Figure 2
SICI in healthy controls and patients with NF1 in on/off medication. Depicted are data of 10 patients of NF1 and healthy controls in on/off medication. Mean inhibition at 60% resting motor threshold (for more details see methods) of the conditioning stimulus was built for ISI of 2, 3, 5 ms and over all ISI. Here, a trend towards increased inhibition in patients with NF1 (treated with placebo but not with lovastatin) compared to healthy controls can be observed. Error bars represent ± standard error of the mean.
Figure 3
Figure 3
Reaction times of TAP in healthy controls and patients with NF1. Data of 10 patients with NF1 and healthy controls are plotted for Alertness, Go/NoGo, Visual Scanning and Incompatibility. Here patients with NF1 (treated with placebo but not with lovastatin) scored in lower ranges than healthy controls. The significance was set at a level of p < 0.05 and error bars represent ± standard error of the mean. Alertness: –WT (without warning tone), +WT (with warning tone); Visual Scanning: CS (critical stimulus), nCS (non critical stimulus); Incompartibility: CC (critical condition), nCC (non critical condition).
Figure 4
Figure 4
Time course of MEP amplitudes of patients with NF1 after PAS with and without lovastatin. A) Shown are data from the NF1 patients (n = 11). Abscissa depicts point in time and ordinate mean MEP amplitude. Administration of lovastatin 4 days before PAS led to a rescue of motor cortex plasticity in patients. Asterisks indicate significant differences between the two drug conditions (p < 0.05, paired t test) and error bars represent ± standard error of the mean. B) MEP amplitudes from one representative subject for placebo and lovastatin condition before and at POST 1 after PAS. Shown are averages of 20 MEP trials in each case.
Figure 5
Figure 5
Reaction times of TAP in patients with NF1 with placebo or lovastatin medication. Data of 10 patients with NF1 are plotted for Alertness, Go/NoGo, Visual Scanning and Incompatibility. For phasic alertness and Incompatibility (KB) patients with NF1 (treated with lovastatin) scored in lower ranges than patients treated with placebo. The significance was set at a level of p < 0.05 and error bars represent ± standard error of the mean. Alertness: –WT (without warning tone), +WT (with warning tone); Visual Scanning: CS (critical stimulus), nCS (non critical stimulus); Incompartibility: CC (critical condition), nCC (non critical condition).
Figure 6
Figure 6
Timeline of PAS experiment. We measured motor evoked potentials (MEP) amplitudes as well as resting motor threshold before and at three points in time after paired associative stimulation (PAS). PAS was performed with an interstimulus interval of 25 ms.

References

    1. Basu TN, Gutmann DH, Fletcher JA, Glover TW, Collins FS, Downward J. Aberrant regulation of ras proteins in malignant tumour cells from type 1 neurofibromatosis patients. Nature. 1992;13(6371):713–715. doi: 10.1038/356713a0.
    1. DeClue JE, Papageorge AG, Fletcher JA, Diehl SR, Ratner N, Vass WC, Lowy DR. Abnormal regulation of mammalian p21ras contributes to malignant tumor growth in von Recklinghausen (type 1) neurofibromatosis. Cell. 1992;13(2):265–273. doi: 10.1016/0092-8674(92)90407-4.
    1. North K. Neurofibromatosis type 1. Am J Med Genet. 2000;13(2):119–127. doi: 10.1002/1096-8628(200022)97:2<119::AID-AJMG3>;2-3.
    1. Shilyansky C, Lee YS, Silva AJ. Molecular and cellular mechanisms of learning disabilities: a focus on NF1. Annu Rev Neurosci. 2010;13:221–243. doi: 10.1146/annurev-neuro-060909-153215.
    1. Friedman JM. Epidemiology of neurofibromatosis type 1. Am J Med Genet. 1999;13(1):1–6. doi: 10.1002/(SICI)1096-8628(19990326)89:1<1::AID-AJMG3>;2-8.
    1. Hyman SL, Shores A, North KN. The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005;13(7):1037–1044. doi: 10.1212/01.wnl.0000179303.72345.ce.
    1. Mautner VF, Kluwe L, Thakker SD, Leark RA. Treatment of ADHD in neurofibromatosis type 1. Dev Med Child Neurol. 2002;13(3):164–170. doi: 10.1017/S0012162201001876.
    1. Li W, Cui Y, Kushner SA, Brown RA, Jentsch JD, Frankland PW, Cannon TD, Silva AJ. The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Curr Biol. 2005;13(21):1961–1967. doi: 10.1016/j.cub.2005.09.043.
    1. Costa RM, Silva AJ. Molecular and cellular mechanisms underlying the cognitive deficits associated with neurofibromatosis 1. J Child Neurol. 2002;13(8):622–626. doi: 10.1177/088307380201700813.
    1. Silva AJ, Frankland PW, Marowitz Z, Friedman E, Laszlo GS, Cioffi D, Jacks T, Bourtchuladze R. A mouse model for the learning and memory deficits associated with neurofibromatosis type I. Nat Genet. 1997;13(3):281–284. doi: 10.1038/ng0397-281.
    1. Shilyansky C, Karlsgodt KH, Cummings DM, Sidiropoulou K, Hardt M, James AS, Ehninger D, Bearden CE, Poirazi P, Jentsch JD. et al.Neurofibromin regulates corticostriatal inhibitory networks during working memory performance. Proc Natl Acad Sci USA. 2010;13(29):13141–13146. doi: 10.1073/pnas.1004829107.
    1. Cui Y, Costa RM, Murphy GG, Elgersma Y, Zhu Y, Gutmann DH, Parada LF, Mody I, Silva AJ. Neurofibromin regulation of ERK signaling modulates GABA release and learning. Cell. 2008;13(3):549–560. doi: 10.1016/j.cell.2008.09.060.
    1. Krab LC, de Goede-Bolder A, Aarsen FK, Pluijm SM, Bouman MJ, van der Geest JN, Lequin M, Catsman CE, Arts WF, Kushner SA. et al.Effect of simvastatin on cognitive functioning in children with neurofibromatosis type 1: a randomized controlled trial. JAMA. 2008;13(3):287–294. doi: 10.1001/jama.300.3.287.
    1. Acosta MT, Kardel PG, Walsh KS, Rosenbaum KN, Gioia GA, Packer RJ. Lovastatin as treatment for neurocognitive deficits in neurofibromatosis type 1: phase I study. Pediatr Neurol. 2011;13(4):241–245. doi: 10.1016/j.pediatrneurol.2011.06.016.
    1. Rioult-Pedotti MS, Friedman D, Hess G, Donoghue JP. Strengthening of horizontal cortical connections following skill learning. Nat Neurosci. 1998;13(3):230–234. doi: 10.1038/678.
    1. Thomas GM, Huganir RL. MAPK cascade signalling and synaptic plasticity. Nat Rev Neurosci. 2004;13(3):173–183.
    1. Guilding C, McNair K, Stone TW, Morris BJ. Restored plasticity in a mouse model of neurofibromatosis type 1 via inhibition of hyperactive ERK and CREB. Eur J Neurosci. 2007;13(1):99–105. doi: 10.1111/j.1460-9568.2006.05238.x.
    1. Cooke SF, Bliss TV. Plasticity in the human central nervous system. Brain. 2006;13(Pt 7):1659–1673.
    1. Stefan K, Kunesch E, Cohen LG, Benecke R, Classen J. Induction of plasticity in the human motor cortex by paired associative stimulation. Brain. 2000;13(Pt 3):572–584.
    1. Stefan K, Kunesch E, Benecke R, Cohen LG, Classen J. Mechanisms of enhancement of human motor cortex excitability induced by interventional paired associative stimulation. J Physiol. 2002;13(Pt 2):699–708.
    1. Delvendahl I, Jung NH, Mainberger F, Kuhnke NG, Cronjaeger M, Mall V. Occlusion of bidirectional plasticity by preceding low-frequency stimulation in the human motor cortex. Clin Neurophysiol. 2010;13(4):594–602. doi: 10.1016/j.clinph.2009.09.034.
    1. Pascual-Leone A, Valls-Sole J, Wassermann EM, Hallett M. Responses to rapid-rate transcranial magnetic stimulation of the human motor cortex. Brain. 1994;13(Pt 4):847–858.
    1. Kujirai T, Caramia MD, Rothwell JC, Day BL, Thompson PD, Ferbert A, Wroe S, Asselman P, Marsden CD. Corticocortical inhibition in human motor cortex. J Physiol. 1993;13:501–519.
    1. Battaglia F, Wang HY, Ghilardi MF, Gashi E, Quartarone A, Friedman E, Nixon RA. Cortical plasticity in Alzheimer’s disease in humans and rodents. Biol Psychiatry. 2007;13(12):1405–1412. doi: 10.1016/j.biopsych.2007.02.027.
    1. Frantseva MV, Fitzgerald PB, Chen R, Moller B, Daigle M, Daskalakis ZJ. Evidence for impaired long-term potentiation in schizophrenia and its relationship to motor skill learning. Cereb Cortex. 2008;13(5):990–996. doi: 10.1093/cercor/bhm151.
    1. Battaglia F, Quartarone A, Rizzo V, Ghilardi MF, Di Rocco A, Tortorella G, Girlanda P. Early impairment of synaptic plasticity in patients with Down’s syndrome. Neurobiol Aging. 2008;13(8):1272–1275. doi: 10.1016/j.neurobiolaging.2007.02.025.
    1. Bagnato S, Agostino R, Modugno N, Quartarone A, Berardelli A. Plasticity of the motor cortex in Parkinson’s disease patients on and off therapy. Mov Disord. 2006;13(5):639–645. doi: 10.1002/mds.20778.
    1. Costa RM, Federov NB, Kogan JH, Murphy GG, Stern J, Ohno M, Kucherlapati R, Jacks T, Silva AJ. Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature. 2002;13(6871):526–530. doi: 10.1038/nature711.
    1. Ziemann U, Muellbacher W, Hallett M, Cohen LG. Modulation of practice-dependent plasticity in human motor cortex. Brain. 2001;13(Pt 6):1171–1181.
    1. Ziemann U, Siebner HR. Modifying motor learning through gating and homeostatic metaplasticity. Brain Stimul. 2008;13(1):60–66. doi: 10.1016/j.brs.2007.08.003.
    1. Russmann H, Lamy JC, Shamim EA, Meunier S, Hallett M. Associative plasticity in intracortical inhibitory circuits in human motor cortex. Clin Neurophysiol. 2009;13(6):1204–1212. doi: 10.1016/j.clinph.2009.04.005.
    1. Tobert JA, Hitzenberger G, Kukovetz WR, Holmes IB, Jones KH. Rapid and substantial lowering of human serum cholesterol by mevinolin (MK-803), an inhibitor of hydroxymethylglutaryl-coenzyme a reductase. Atherosclerosis. 1982;13(1):61–65. doi: 10.1016/0021-9150(82)90070-3.
    1. Rossi S, Hallett M, Rossini PM, Pascual-Leone A. Safety, ethical considerations, and application guidelines for the use of transcranial magnetic stimulation in clinical practice and research. Clin Neurophysiol. 2009;13(12):2008–2039. doi: 10.1016/j.clinph.2009.08.016.
    1. Brasil-Neto JP, Cohen LG, Panizza M, Nilsson J, Roth BJ, Hallett M. Optimal focal transcranial magnetic activation of the human motor cortex: effects of coil orientation, shape of the induced current pulse, and stimulus intensity. J Clin Neurophysiol. 1992;13(1):132–136. doi: 10.1097/00004691-199201000-00014.
    1. Kaneko K, Kawai S, Fuchigami Y, Morita H, Ofuji A. The effect of current direction induced by transcranial magnetic stimulation on the corticospinal excitability in human brain. Electroencephalogr Clin Neurophysiol. 1996;13(6):478–482.
    1. Jung NH, Delvendahl I, Kuhnke NG, Hauschke D, Stolle S, Mall V. Navigated transcranial magnetic stimulation does not decrease the variability of motor-evoked potentials. Brain Stimul. 2010;13(2):87–94. doi: 10.1016/j.brs.2009.10.003.
    1. Awiszus F. TMS and threshold hunting. Suppl Clin Neurophysiol. 2003;13:13–23.
    1. Mishory A, Molnar C, Koola J, Li X, Kozel FA, Myrick H, Stroud Z, Nahas Z, George MS. The maximum-likelihood strategy for determining transcranial magnetic stimulation motor threshold, using parameter estimation by sequential testing is faster than conventional methods with similar precision. JECT. 2004;13(3):160–165.
    1. Rossini PM, Barker AT, Berardelli A, Caramia MD, Caruso G, Cracco RQ, Dimitrijevic MR, Hallett M, Katayama Y, Lucking CH. et al.Non-invasive electrical and magnetic stimulation of the brain, spinal cord and roots: basic principles and procedures for routine clinical application. Report of an IFCN committee. Electroencephalogr Clin Neurophysiol. 1994;13(2):79–92. doi: 10.1016/0013-4694(94)90029-9.
    1. Muellbacher W, Ziemann U, Wissel J, Dang N, Kofler M, Facchini S, Boroojerdi B, Poewe W, Hallett M. Early consolidation in human primary motor cortex. Nature. 2002;13(6872):640–644. doi: 10.1038/nature712.
    1. Stefan K, Wycislo M, Classen J. Modulation of associative human motor cortical plasticity by attention. J Neurophysiol. 2004;13(1):66–72. doi: 10.1152/jn.00383.2003.
    1. Peurala SH, Muller-Dahlhaus JF, Arai N, Ziemann U. Interference of short-interval intracortical inhibition (SICI) and short-interval intracortical facilitation (SICF) Clin Neurophysiol. 2008;13(10):2291–2297. doi: 10.1016/j.clinph.2008.05.031.
    1. Orth M, Snijders AH, Rothwell JC. The variability of intracortical inhibition and facilitation. Clin Neurophysiol. 2003;13(12):2362–2369. doi: 10.1016/S1388-2457(03)00243-8.
    1. Stinear CM, Byblow WD. Elevated threshold for intracortical inhibition in focal hand dystonia. Mov Disord. 2004;13(11):1312–1317. doi: 10.1002/mds.20160.
    1. Zimmermann P, Fimm B. Testbatterie zur Aufmerksamkeitsprüfung [Testbattery of Attentional Performance (TAP)] version 1.7. Herzogenrath: Psytest; 2002.
    1. Igel M, Sudhop T, von BK. Pharmacology of 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins), including rosuvastatin and pitavastatin. J Clin Pharmacol. 2002;13(8):835–845. doi: 10.1177/009127002401102731.

Source: PubMed

3
Tilaa