Efficacy of fumaric acid esters in the R6/2 and YAC128 models of Huntington's disease

Gisa Ellrichmann, Elisabeth Petrasch-Parwez, De-Hyung Lee, Christiane Reick, Larissa Arning, Carsten Saft, Ralf Gold, Ralf A Linker, Gisa Ellrichmann, Elisabeth Petrasch-Parwez, De-Hyung Lee, Christiane Reick, Larissa Arning, Carsten Saft, Ralf Gold, Ralf A Linker

Abstract

Huntington's disease (HD) is an autosomal dominantly inherited progressive neurodegenerative disease. The exact sequel of events finally resulting in neurodegeneration is only partially understood and there is no established protective treatment so far. Some lines of evidence speak for the contribution of oxidative stress to neuronal tissue damage. The fumaric acid ester dimethylfumarate (DMF) is a new disease modifying therapy currently in phase III studies for relapsing-remitting multiple sclerosis. DMF potentially exerts neuroprotective effects via induction of the transcription factor "nuclear factor E2-related factor 2" (Nrf2) and detoxification pathways. Thus, we investigated here the therapeutic efficacy of DMF in R6/2 and YAC128 HD transgenic mice which mimic many aspects of HD and are characterized by an enhanced generation of free radicals in neurons. Treatment with DMF significantly prevented weight loss in R6/2 mice between postnatal days 80-90. At the same time, DMF treatment led to an attenuated motor impairment as measured by the clasping score. Average survival in the DMF group was 100.5 days vs. 94.0 days in the placebo group. In the histological analysis on day 80, DMF treatment resulted in a significant preservation of morphologically intact neurons in the striatum as well as in the motor cortex. DMF treatment resulted in an increased Nrf2 immunoreactivity in neuronal subpopulations, but not in astrocytes. These beneficial effects were corroborated in YAC128 mice which, after one year of DMF treatment, also displayed reduced dyskinesia as well as a preservation of neurons. In conclusion, DMF may exert beneficial effects in mouse models of HD. Given its excellent side effect profile, further studies with DMF as new therapeutic approach in HD and other neurodegenerative diseases are warranted.

Conflict of interest statement

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

Figures

Figure 1. R6/2 mice display higher levels…
Figure 1. R6/2 mice display higher levels of oxidative stress.
(A, B) Representative confocal laser scanning image from the striatum of a 3 months old R6/2 mouse (male, A) after hydroethidine staining in comparison to the striatum of a 4 months old C57BL/6 mouse (B). (C, D) Hydroethidine staining of the motor cortex. The same representative R6/2 mouse (C) and C57BL/6 control mouse (D) as in A,B are shown. The amount of free radicals in neuronal cells is indicated in red. Note the increased immunofluorescence in the R6/2 mouse indicating a higher amount of oxidative stress in both anatomical regions. Bar = 20 µm.
Figure 2. DMF improves survival and prevents…
Figure 2. DMF improves survival and prevents weight loss in R6/2 mice.
(A) Comparison of body weight changes in DMF treated male R6/2 mice (black bars) and controls treated with carrier solution alone (white bars). Treatment with DMF prevented weight loss in R6/2 mice on postnatal days 85 (p = 0.05) and 90 (p = 0.04). Data are shown as change in body weight (g) on days 80, 85 and 90 to the respective baseline weight at the start of treatment. Numbers of mice per day of analysis are indicated in the graph. (B) Kaplan Meier survival analysis of male R6/2 mice treated with DMF (n = 14, black curve) or methocel (n = 19, grey curve). DMF treatment leads to prolonged survival of R6/2 mice. Confidence interval for mean survival: DMF 97.7–104.6 vs. methocel 91.22–99.6 days.
Figure 3. DMF preserves motor functions in…
Figure 3. DMF preserves motor functions in R6/2 mice.
(A) Rotarod analysis. A cohort of R6/2 mice treated with DMF (n = 46) or methocel (n = 42) is shown. Although DMF treatment (black bars) leads to a trend towards longer times on the accelerating rod, there is no significant difference as compared to methocel treated controls (white bars). (B) Representative images of a R6/2 mouse treated with DMF and a mouse treated with methocel at the age of 12 weeks. Note the clasping of the front- and hindlimbs in the control mouse. (C) Clasping Score. A cohort of R6/2 mice treated with DMF (n = 46, black bars) or methocel (n = 42, white bars) is shown. At the age of 12 weeks, there is a significant difference between both groups and DMF treated mice display a reduction of limb dyskinesia.
Figure 4. DMF leads to preservation of…
Figure 4. DMF leads to preservation of morphologically intact neurons.
Representative images of the striatum (A,B and E,F) or motor cortex (C,D and G,H) from DMF (A,C,E,G) or methocel treated mice (B,D,F,H) on day 80 are shown. Bar = 100 µm. Arrows indicate intact, arrowheads indicate degenerating neurons. (A–D) NeuN staining of the striatum (A,B) and the motor cortex (C,D) in R6/2 mice (day 80). Note the higher number of NeuN immunoreactive neurons after DMF treatment (marked by arrows, arrowheads indicate degenerating striatal neurons with loss of NeuN immunoreactivity). (E–H) Cresyl violet staining of the striatum (E,F) and motorcortex (G,H) in R6/2 mice. Note the higher number of intact neurons after DMF treatment (marked by arrows) and the increased number of shrunken, dystrophic neurons after methocel treatment (marked by arrowheads). False color images depicting cresyl violet staining in red are shown.
Figure 5. DMF treatment prevents dark cell…
Figure 5. DMF treatment prevents dark cell degeneration in R6/2 mice.
(A,B) Overview images of semithin sections show the striatum of DMF (A) and methocel treated (B) R6/2 mice at 80 days of age. Representative images are shown. (C,D) At higher enlargement (for localization see rectangles), the light striatal neurons appear intact in the DMF treated mouse (C) whereas dark cells (arrows) are dispersed among intact neurons in the methocel treated mouse (D). (E,F) Electron microscopy confirms neuronal integrity in DMF treated mice (E) and the condensed and shrunken appearance of severely affected striatal neurons in methocel treated mice (F). (G,H) Semithin overviews display regular cortical morphology of the DMF treated mouse (G), but an irregular appearance in the cortex of representative methocel treated mouse (H). (I,K) At higher enlargements (rectangles) of corresponding layers the DMF treated mouse exhibits intact neurons (I) whereas the methocel treated mouse show multiple dark cells of varying size (arrows) dispersed between single intact neurons (K). (L,M) Electron microscopy confirms the dark cell degeneration with shrunken dark cytoplasm (arrows) and a ruffled nuclear envelope of a respective neuron (M) in contrast to the intact cortical neuron of a DMF treated mouse (L). Both the intact and the dark neuron exhibit a prominent intranuclear round inclusion (x). Bars in A,B,G,H = 100 µm; bars in C,D,I,K = 20 µm; bars in E,F,L,M = 1 µm.
Figure 6. Increased Nrf2 immunoreactivity after DMF…
Figure 6. Increased Nrf2 immunoreactivity after DMF treatment.
Representative images of the striatum from DMF (A, C, E) or methocel treated mice (B, D, F) on day 80 are shown. (A,B) In contrast to methocel treated R6/2 mice, there is an increased number of Nrf2 positive cells after DMF treatment (Nrf2 immunopositive cells are marked by arrows). Bar = 100 µm. (C,D) Confocal laser scanning microscopy images after NeuN/Nrf2 double staining. In contrast to vehicle treated mice (D), a higher number of NeuN/Nrf2 double positive cells are observed in the striatum in DMF treated R6/2 mice (C). Arrows mark NeuN/Nrf2 double labelled cells. Bar = 20 µm. (E,F) Confocal laser scanning microscopy images after GFAP/Nrf2 double labelling. In the striatum, significant numbers of Nrf2 immunopositive astrocytes were neither identified after DMF treatment (E), nor in control mice (F). Arrows indicate Nrf2 negative astrocytes. Bar = 20 µm; images represent a Z-stack of 10 µm.
Figure 7. DMF preserves motor functions in…
Figure 7. DMF preserves motor functions in YAC128 mice.
(A) Rotarod analysis. A cohort of YAC128 mice treated with DMF (n = 23, black bars) or methocel (n = 22, white bars) is shown. Although DMF treatment leads to a trend towards longer times on the accelerating rod, there is no significant difference between both groups. (B) Clasping Score. A cohort of YAC128 mice treated with DMF (n = 23) or methocel (n = 22) is shown. In comparison to control mice (white bars), there is a trend towards reduced clasping behaviour in the DMF treated group (black bars) from the age of 47 weeks. (C) Blinded quantification of neuronal densities in the striatum after creysl violet staining on day 365 (n = 3 per group). There is a significant preservation of neuronal numbers after DMF treatment (black bar) as compared to methocel treatment (white bar, p = 0.03). (D) Blinded quantification of neuronal densities in the motor cortex after creysl violet staining on day 365 (n = 3/5 per group). There is a significant preservation of neuronal numbers after DMF treatment (black bar) as compared to methocel treatment (white bar, p = 0.004).

References

    1. Vonsattel JP, DiFiglia M. Huntington disease. J Neuropathol Exp Neurol. 1998;57:369–384.
    1. Kumar P, Kalonia H, Kuman A. Huntington's disease: pathogenesis to animal models. Pharmacol Rep. 2010;62:1–14.
    1. Mrowietz U, Christophers E, Altmeyer P. Treatment of severe psoriasis with fumaric acid esters: scientific background and guidelines for therapeutic use. The German Fumaric Ester Consensus Conference. Br J Dermatol. 1999;141:424–429.
    1. Schweckendiek W. Treatment of psoriasis vulgaris. Med Monatsschr. 1959;13:103–104.
    1. Linker RA, Lee DH, Stangel M, Gold R. Fumarates for the treatment of multiple sclerosis, potential mechanisms of actions and clinical studies. Expert Rev Neurother. 2008;8:1683–1690.
    1. Linker RA, Lee DH, Ryan S, van Dam AM, Conrad R, et al. Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway. Brain. In press.
    1. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell. 1996;87:493–506.
    1. Stack EC, Kubilius JK, Smith K, Cormier K, Del Signore SJ, et al. Chronology of behavioral symptoms and neuropathological sequela in R6/2 Huntington's disease transgenic mice. J Comp Neurol. 2005;490:354–370.
    1. Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F, et al. A YAC mouse model for Huntington's disease with full-length mutant huntingtin, cytoplasmatic toxicity, and selective striatal neurodegeneration. Neuron. 1999;23:181–192.
    1. Slow EJ, van Raamsdonk J, Rogers D, Coleman SH, Graham RK, et al. Selective striatal neuronal loss in a YAC128 mouse model of Huntington disease. Hum Mol Genet. 2003;12:1555–1567.
    1. van Raamsdonk JM, Pearson J, Slow EJ, Hossain SM, Leavitt BR, et al. Cognitive dysfunction precedes neuropathology and motor abnormalities in the YAC128 mouse model of Huntington's disease. J Neurosci. 2005;25:4169–4180.
    1. Pang TY, Stam NC, Nithianantharajah J, Howard ML, Hannan AJ, et al. Differential effects of voluntary physical exercise on behavioral and brain-derived neurotrophic factor expression deficits in Huntington's disease transgenic mice. Neuroscience. 2006;25:569–584.
    1. van Dellen A, Blakemore C, Deacon R, York D, Hannan AJ. Delaying the onset of Huntington's in mice. Nature. 2000;404:721–722.
    1. Clarke KA, Still J. Gait Analysis in the Mouse. Physiol and Behav. 1999;66:723–729.
    1. Davies SW, Turmaine M, Cozens B, DiFiglia M, Sharp AH, et al. Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell. 1997;90:537–548.
    1. Li H, Li SH, Yu ZX, Shelbourne P, Li XJ. Huntingtin aggregate-associated axonal degeneration is an early pathological event in Huntington's disease mice. J Neurol. 2001;21:8473–8481.
    1. Linker RA, Mäurer M, Gaupp S, Martini R, Holtmann B, et al. CNTF is a major protective factor in demyelinating CNS disease: a neurotrophic cytokine as modulator in neuroinflammation. Nat Med. 2002;8:620–624.
    1. Paxinos G, Franklin KB. The mouse brain in stereotaxic coordinates. Academic Press, Elsevier Publishing Company; 2007.
    1. Petrasch-Parwez E, Nguyen HP, Löbbecke-Schumacher M, Habbes HW, Wieczorek S, et al. Cellular and subcellular localization of Huntingtin [corrected] aggregates in the brain of a rat transgenic for Huntington disease. J Comp Neurol. 2007;501:716–730.
    1. Kleinschnitz C, Grund H, Wingler K, Armitage ME, Jones E, et al. Post-stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and neurodegeneration. PloS Biol. 2010;8(9). Pii:e1000479.
    1. Carter RJ, Lione LA, Humby T, Mangiarini L, Mahal A, et al. Characterization of Progressive Motor Deficits in Mice Transgenic for the Human Huntington's Disease Mutation. J Neurosci. 1999;19:3248–3257.
    1. Browne SE, Bowling AC, MacGarvey U, Baik MJ, Berger SC, et al. Oxidative damage and metabolic dysfunction in Huntington's disease: selective vulnerability of the basal ganglia. Ann Neurol. 1997;41:646–653.
    1. Bogdanov MB, Andreassen OA, Dedeoglu A, Ferrante RJ, Beal MF. Increased oxidative damage to DNA in a transgenic mouse model of Huntington's disease. J Neurochem. 2001;79:1246–1249.
    1. Begleiter A, Sivananthan K, Curphey TJ, Bird RP. Induction of NAD(P)H quinone: oxidoreductase1 inhibits carcinogen-induced aberrant crypt foci in colons of Sprague-Dawley rats. Cancer Epidemol Biomarkers Prev. 2003;12:566–572.
    1. Schmidt TJ, Ak M, Mrowietz U. Reactivity of dimethyl fumarate and methylhydrogen fumarate towards glutathione and N-acetyl-L-cysteine-preparation of S-substituted thiosuccinic acid esters. Bioorg Med Chem. 2007;15:333–342.
    1. Wierinckx A, Breve J, Mercier D, Schultzberg M, Drukarch B, et al. Detoxication enzyme inducers modify cytokine production in rat mixed glial cells. J Neuroimmunol. 2005;166:132–143.
    1. Fernández-Checa JC, Fernández A, Morales A, Marí M, García-Ruiz C, et al. Oxidative stress and altered mitochondrial function in neurodegenerative diseases: lessons from mouse models. CNS Neurol Disord Drug Targets. 2010;9(4):439–454.
    1. Ang ET, Tai YK, Lo SQ, Seet R, Soong TW. Neurodegenerative diseases: exercising toward neurogenesis and neurodegeneration. Front Aging Neurosci. 2010;2:25.
    1. Jung KA, Kwalk MK. The Nrf2 system as a potantial target for the development of indirect antioxidants. Molecules. 2010;15(10):7266–7291.
    1. Duffy S, So A, Murphy TH. Activation of endogenous antioxidant defenses in neuronal cells prevents free radical-mediated damage. J Neurochem. 1998;71:69–77.
    1. Su JY, Duffy S, Murphy TH. Reduction of H2O2 evoked, intracellular caldium increases in the rat N18-RE-105 neuronal cell line by pretreatment with an electrophilic antioxidant inducer. Neurosci Lett. 1999;273:109–112.
    1. Moharregh-Khiabani D, Blank A, Skripuletz T, Miller E, Kotsiari A, et al. Effects of fumaric acids on cuprizone induced central nervous system de- and remyelination in the mouse. PLoS One. 2010;5(7):e11769.
    1. Thiessen A, Schmidt MM, Dringen R. Fumaric acid dialkyl esters deprive cultured rat oligodendroglial cells of glutathione and upregulate the expression of heme oxygenase 1. Neurosci Lett. 2010;475:56–60.
    1. Calkins MJ, Jakel RJ, Johnson DA, Chan K, Kan YW, et al. Protection from mitochondrial complex II inhibition in vitro and in vivo by Nrf2-mediated transcription. Proc Natl Acad Sci. 2005;102(1):244–9.
    1. Lee JM, Shih AY, Murphy TH, Johnson JA. NF-E2-related factor-2 mediates neuroprotection against mitochondrial complex I inhibitors and increased concentrations of intracellular calcium in primary cortical neurons. J Biol Chem. 2003;278:37948–37956.
    1. Satoh T, Okamoto SI, Cui J, Watanabe Y, Furunta K, et al. Activation of the Keap1/Nrf2 pathway for neuroprotection by electrophilic [correction of electrophilic] phase II inducers. Proc Natl Acad Sci U S A. 2006;103:768–773.
    1. Shih AY, Imbeault S, Barakauskas V, Erb H, Jiang L, et al. Induction of the Nrf2-driven antioxidant response confers neuroprotection during mitochondrial stress in vivo. J Biol Chem. 2005;280:22925–22936.
    1. Stack C, Ho D, Wille E, Calingasan NY, Williams C, et al. Triterpenoids CDDO-ethyl amide and CDDO-trifluoroethyl amide improve the behavioral phenotype and brain pathology in a transgenic mouse model of Huntington's disease. Free Radic Biol Med. 2010;49:147–158.
    1. Chen PC, Vargas MR, Pani AK, Smeyne RJ, Johnson DA, et al. Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson's disease: Critical role for the astrocyte. Proc Natl Acad Sci U S A. 2009;24:2933–2938.
    1. Kraft AD, Johnson DA, Johnson JA. Nuclear factor E2-related factor 2-dependent antioxidant response element activation by tert-buthylhydroquinone and sulforaphane occuring preferentially in astrocytes conditions neurons against oxidative insult. J Neurosci. 2004;24:1101–1112.
    1. Shih AY, Johnson DA, Wong G, Kraft AD, Jiang L, et al. Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress. J Neurosci. 2003;23:3394–3406.
    1. Vargas MR, Johnson DA, Sirkis DW, Messing A, Johnson JA. Nrf2 activation in astrocytes protects against neurodegeneration in mouse models of familial amyotrophic lateral sclerosis. J Neurosci. 2008;28:13574–13581.
    1. De Jong R, Bezemer AC, Zomerdijk TP, Pouw-Kraan T, Ottenhoff TH, et al. Selective stimulation of T helper 2 cytokine responses by the anti-psoriasis agent monomethylfumarate. Eur J Immunol. 1996;26:2067–2074.
    1. Loewe R, Holnthoner W, Groger M. Dimethylfumarate inhibits TNF-induced nuclear entry of NF-kappa B/p65 in human endothelial cells. J Immunol. 2002;168:203–208.
    1. Ockenfels HM, Schultewolter T, Ockenfels G, Funk R, Goos M. The antipsoriatic agent dimethylfumarate immunomodulates T-cell cytokine secretion and inhibits cytokines of the psoriatic cytokine network. Br J Dermatol. 1998;139:390–395.
    1. Sebok B, Bonnekoh B, Vetter R, Schneider I, Gollnick H, et al. The antipsoriatic dimethyl-fumarate suppresses interferon-gamma-induced ICAM-d1 and HLA-DR expression on hyperproliferative keratinocytes. Quantification by a culture plate-directed APAAP-ELISA technique. Eur J Dermatol. 1998;8:29–32.
    1. Vandermeeren M, Janssens S, Borgers M, Geysen J. Dimethylfumarate is an inhibitor of cytokine-induced E-selectin, VCAM-1, and ICAM-1 expression in human endothelial cells. Biochem Biophys Res Commun. 1997;234:19–23.
    1. Clifford JJ, Drago J, Natoli AL, Wong JYF, Kinsella A, et al. Essential fatty acids given from conception prevent topographies of motor deficits in a transgenic model of Huntington's disease. Neuroscience. 2002;109:81–88.
    1. Smith KM, Matson S, Matson WR, Cormier K, Del Signore SJ, et al. Dose ranging and efficacy study of high-dose coenzyme Q10 formulations in Huntington's disease mice. Biochem Biophys Acta. 2006;1762:616–626.
    1. Keene CD, Rodrigues CM, Eich T, Chhabra MS, Steer CJ, et al. Tauroursodeoxycholic acid, a bile acid, is neuroprotective in a transgenic animal model of Huntington's disease. Proc Natl Acad Sci U S A. 2002;99:10671–10676.
    1. Gil JM, Rego C. The R6 lines of transgenic mice: A model for screening new therapies for Huntington's disease. Brain Res Rev. 2009;59:410–431.
    1. Menalled LB, Sison JD, Wu Y, Olivieri M, Li XJ, et al. Early motor dysfunction and striosomal distribution of huntingtin microaggregates in Huntington's disease knock-in mice. J Neurosci. 2002;22:8266–8276.
    1. Iannicola C, Moreno S, Oliverio S, Nardacci R, Ciofi-Luzzatto A, et al. Early alterations in gene expression and cell morphology in a mouse model of Huntington's disease. J Neurochem. 2000;75:830–839.
    1. Turmaine M, Raza A, Mahal A, Mangiarini L, Bates GP, et al. Nonapoptotic neurodegeneration in a transgenic mouse model of Huntington's disease. Proc Natl Acad Sci U S A. 2000;97:8093–8097.
    1. Yu ZX, Li SH, Evans J, Pillarisetti A, Li H, et al. Mutant huntingtin causes context-dependent neurodegeneration in mice with Huntington's disease. J Neurosci. 2003;23:2193–2202.
    1. Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, et al. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J Neurosci. 2003;23:9418–9427.
    1. Hockly E, Richon VM, Woodman B, Smith DL, Zhou X, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model af Huntington's disease. Proc Natl Acad Sci U S A. 2003;100:2041–2046.
    1. Heiser V, Scherzinger E, Boeddrich A, Nordhoff E, Lurz R, et al. Inhibition of huntingtin fibrillogenesis by specific antibodies and small molecules: implication for Huntington's disease therapy. Proc Natl Acad Sci U S A. 2000;97:6739–6744.
    1. Klunk WE, Pettegrew JW, Abraham DJ. Quantitative evaluation of Congo red binding to amyloid-like proteins with a beta-pleated sheet conformation. J Histochem Cytochem. 1989;37:1273–1281.
    1. Tanaka M, Machida Y, Niu S, Ikeda T, Jana NR, et al. Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington's disease. Nat Med. 2004;10:148–154.
    1. Masuda N, Peng Q, Li Q, Jiang M, Liang Y, et al. Tiagabine is neuroprotective in the N171-82Q and R6/2 mouse models of Huntington's disease. Neurobiol Dis. 2004;30:293–302.
    1. Dedeoglu A, Kubilus JK, Jeitner TM, Matson SA, Bogdanov M, et al. Therapeutic effects of cystamine in a murine model of Huntington's disease. J Neurosci. 2002;22:8942–8950.
    1. Karpuj MV, Becher MW, Springer JE, Chabas D, Youssef S, et al. Prolonged survival and decreased abnormal movements in transgenic model of Huntington disease, with administration of the transglutaminase inhibitor cystamine. Nat Med. 2002;8:143–149.
    1. Wellington CL, Ellerby LM, Hackam AS, Margoli RL, Trifiro MA, et al. Caspase cleavage of gene products associated with triplet expansion disorders generates truncated fragments containig the polyglutamine tract. J Biol Chem. 1998;273:9158–9167.
    1. Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, et al. Minocycline inhibitis caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington's disease. Nat Med. 2000;6:797–801.
    1. Smith DL, Woodman B, Mahal A, Sathasivam K, Ghazi-Noori S, et al. Minocycline and doxycycline are not beneficial in a model of Huntington's disease. Ann Neurol. 2003;54:186–196.
    1. Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG. Minocycline inhibits caspase-independent and –dependent mitochondrial cell death pathways in models of Huntington's disease. Proc Natl Acad Sci U S A. 2003;100:10483–10487.
    1. Menalled LB, Patry M, Ragland N, Lowden PAS, Goodmann J, et al. Comprehensive behavioral testing in the R6/2 mouse model of Huntington's disease shows no benefit from CoQ10 or Minocycline. Plos One. 2010;5:e9793.
    1. Menalled LB, El-Khodor BF, Patry M, Suarez-Farinas M, Orenstein SJ, et al. Systematic behavioral evaluation of Huntington's disease transgenic and knock-in mouse models. Neurobiol Dis. 2009;35:319–336.

Source: PubMed

3
Abonnere