Mechanism-based rescue of Munc18-1 dysfunction in varied encephalopathies by chemical chaperones

Noah Guy Lewis Guiberson, André Pineda, Debra Abramov, Parinati Kharel, Kathryn E Carnazza, Rachel T Wragg, Jeremy S Dittman, Jacqueline Burré, Noah Guy Lewis Guiberson, André Pineda, Debra Abramov, Parinati Kharel, Kathryn E Carnazza, Rachel T Wragg, Jeremy S Dittman, Jacqueline Burré

Abstract

Heterozygous de novo mutations in the neuronal protein Munc18-1 are linked to epilepsies, intellectual disability, movement disorders, and neurodegeneration. These devastating diseases have a poor prognosis and no known cure, due to lack of understanding of the underlying disease mechanism. To determine how mutations in Munc18-1 cause disease, we use newly generated S. cerevisiae strains, C. elegans models, and conditional Munc18-1 knockout mouse neurons expressing wild-type or mutant Munc18-1, as well as in vitro studies. We find that at least five disease-linked missense mutations of Munc18-1 result in destabilization and aggregation of the mutant protein. Aggregates of mutant Munc18-1 incorporate wild-type Munc18-1, depleting functional Munc18-1 levels beyond hemizygous levels. We demonstrate that the three chemical chaperones 4-phenylbutyrate, sorbitol, and trehalose reverse the deficits caused by mutations in Munc18-1 in vitro and in vivo in multiple models, offering a novel strategy for the treatment of varied encephalopathies.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Neuronal impairments in C. elegans expressing mutant UNC-18. a, b Locomotion of C. elegans. Body bends of indicated worm strains per 30 s were counted. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by Student’s t test; n = 5 independent experiments on ten worms per experiment). c Scheme of the aldicarb assay. d Mutants display reduced acetylcholine release at the worm neuromuscular junction. Paralysis of young adult worms expressing WT or mutant unc-18 was measured 60 min after exposure to aldicarb (see Supplementary Fig. 2d for entire curves). Data are means ± SEM (**p < 0.01, ***p < 0.001 by Student’s t test; n = 6 independent experiments on 20 worms per experiment). e Heat-induced paralysis. Indicated worm strains were exposed to 37 °C over a period of 300 min, and paralysis was scored at indicated time points. Data are means ± SEM (***p < 0.001 by two-way ANOVA, compared to N2; ###p < 0.001 by two-way ANOVA, compared to unc-18-WT in unc-18−/−, n = 9–13 independent experiments on ten worms per experiment). f Worm traces after heat-induced paralysis. Plates were imaged after heat shock analysis in e. gi Locomotion, acetylcholine release, and heat shock paralysis of CRISPR-edited C. elegans. Worms were assayed as in ae. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by Student’s t test in g and h and two-way ANOVA in i; n = 15–20 worms for g, n = 5–6 independent experiments on 20–25 worms per experiment for h, and n = 6 independent experiments on ten worms per experiment for i)
Fig. 2
Fig. 2
Synaptic impairments in neurons expressing mutant Munc18-1. a Primary neurons were plated on a multi-electrode array. bg Munc18-1 knockout neurons (c) or knockout neurons expressing Munc18-1b variants (d) were subjected to analysis of mean firing rate, burst frequency, burst duration, and network burst activity (b, eg). Purple boxes in c and d indicate network activity. Data are means ± SEM (*,#p < 0.05, **,##p < 0.01, ***,###p < 0.001 by Student’s t test; n = 4–6 independent experiments). h, i Uptake of synaptotagmin-1 antibody during high K+ stimulation. Neurons expressing cre recombinase and/or WT or mutant Munc18-1b were subjected to an antibody uptake assay. Endocytosed synaptotagmin-1 antibody was quantified by immunostaining (h), via counting the number of pixels > intensity of 15 (i). Data are means ± SEM (*,#p < 0.05, **,##p < 0.01 by Student’s t test; n = 4 independent experiments). Scale bar in h = 20 µm
Fig. 3
Fig. 3
Increased turnover of Munc18-1 mutants in neurons. a Total protein levels of Munc18-1. WT and mutant Munc18-1b were expressed in primary neurons infected with lentiviral vectors expressing cre recombinase. Total protein levels were quantified by immunoblotting, normalized to the levels of the synaptic protein synaptophysin-1 (SypI). Data are means ± SEM (*p < 0.05, ***p < 0.001 by Student’s t test; n = 3 independent experiments). b Turnover of Munc18-1 by cycloheximide chase. Neurons as in a were subjected to a cycloheximide (CHX) chase experiment for the indicated time to stop protein translation. Remaining protein levels were quantified by immunoblotting, normalized to α-tubulin levels. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by two-way ANOVA; n = 3 independent experiments). cf Turnover of Munc18-1 by Dendra2 photoconversion. HEK293T cells were transfected with WT or mutant Munc18-1b:Dendra2 fusion constructs. Two days after transfection, expressed Dendra2 was photoconverted. The green signal was quantified before and after photoconversion (c, d), and the red signal was quantified at 0, 3, and 24 h after photoconversion (e, f). Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by Student’s t test in d and f, and by two-way ANOVA in e; n = 4–6 independent experiments). Scale bar in c = 50 µm
Fig. 4
Fig. 4
Aggregation of Munc18-1 mutants. a Solubility of Munc18-1. Munc18-1 knockout neurons expressing WT or mutant Munc18-1b were solubilized in 0.1% Triton X-100 (TX). Equal volumes of soluble and insoluble fractions were analyzed by quantitative immunoblotting (α-tubulin = control). Data are means ± SEM (**p < 0.01, ***p < 0.001 by Student’s t test; n = 4 independent experiments). b Limited proteolysis. Neurons as in a were incubated with increasing concentrations of trypsin. Remaining protein levels were analyzed by quantitative immunoblotting (α-tubulin = control). Data are means ± SEM (***p < 0.001 by two-way ANOVA; n = 3 independent experiments). c Aggregation of mutant Munc18-1. Neurons as in a were analyzed for the subcellular localization of Munc18-1b by immunocytochemistry. Arrows depict aggregates. Scale bar = 20 µm. d Locomotion of C. elegans expressing GFP-tagged WT or G544D unc-18. Body bends per 30 s were counted. Data are means ± SEM (***p < 0.001 by Student’s t test; n = 5 independent experiments on ten worms per experiment). e Paralysis of WT worms (N2) or worms expressing GFP-tagged WT or G544D mutant unc-18 after 60 min exposure to aldicarb. Data are means ± SEM (***p < 0.001 by Student’s t test; n = 6 independent experiments on ten worms per experiment). f Lack of axonal localization of mutant UNC-18. C. elegans expressing WT::GFP or G544D::GFP were immobilized, and the ventral nerve cord was imaged (solid arrowheads = pairs of bigger puncta, broken arrowheads = single, smaller puncta. Scale bar = 10 µm). g Worm traces after the heat shock assay (Supplementary Fig. 8e). h, i Aggregation of mutant Munc18-1 in yeast. S. cerevisiae expressing GFP-tagged Munc18-1 variants were imaged 24 h after induction of protein expression (h) to quantify aggregation (i). Data are means ± SEM (n = 3 independent experiments). Scale bar = 5 µm. j Expression of mutant Munc18-1 in yeast. Munc18-1 levels were analyzed by measuring GFP fluorescence in a plate reader 24 h post induction. Data are means ± SEM (**p < 0.01, ***p < 0.001 by Student’s t test; n = 4 independent experiments)
Fig. 5
Fig. 5
Dominant-negative activity of mutant Munc18-1 on wild-type Munc18-1. a Protein levels of Munc18-1 in primary neurons. Levels of GFP-tagged WT or mutant Munc18-1 and endogenous Munc18-1 in heterozygous Munc18-1 neurons were analyzed by quantitative immunoblotting, normalized to α-tubulin levels. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by Student’s t test; n = 3 independent experiments). b Same as in a, except that myc-tagged Munc18-1 variants were co-expressed with HA-tagged WT Munc18-1 in Munc18-1 knockout neurons. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by Student’s t test; n = 5–6 independent experiments). c Co-immunoprecipitation of mutant with wild-type Munc18-1. Lysates of HEK293T cells that were co-transfected with HA-tagged WT and myc-tagged mutant Munc18-1 were subjected to immunoprecipitation with an anti-c-myc antibody (IP) or no antibody (control). Precipitated myc- and HA-tagged Munc18-1 was analyzed with the respective input by immunoblotting. GAPDH served as control. d Solubility of Munc18-1. Primary neurons infected as in b were solubilized in 0.1% Triton X-100 (TX). Equal volumes of TX-soluble and -insoluble fractions were analyzed by quantitative immunoblotting. α-Tubulin served as control. Data are means ± SEM (*p < 0.05, ***p < 0.001 by Student’s t test; n = 5–7 independent experiments). e Locomotion of C. elegans. Body bends per 30 s were counted. Data are means ± SEM (*p < 0.05, **p < 0.01 by Student’s t test; n = 15 independent experiments on ten worms per experiment). f Heat-induced paralysis. Paralysis at 37 °C was scored at indicated time points. Data are means ± SEM (***p < 0.001 by two-way ANOVA, compared to N2; n = 3 independent experiments on ten worms per experiment). g Mutants display reduced acetylcholine release at the worm neuromuscular junction. Paralysis of N2 worms expressing WT or mutant unc-18 was measured after 60 min exposure to aldicarb. Data are means ± SEM (**p < 0.01, ***p < 0.001 by Student’s t test; n = 6 independent experiments on 20 worms per experiment)
Fig. 6
Fig. 6
Chemical chaperones rescue mutant Munc18-1 deficits in neurons. a Total protein levels of Munc18-1. WT and mutant Munc18-1b were expressed in primary cortical neurons infected with lentiviral vectors expressing cre recombinase and Munc18-1b variants in the presence or absence of chemical chaperones. Total protein levels were quantified 7 days after infection by immunoblotting, normalized to the levels of the post-synaptic protein PSD-93. Data are means ± SEM (*p < 0.05, **p < 0.01 by Student’s t test; n = 5 independent experiments). b Triton X-100 solubility of Munc18-1. WT or mutant Munc18-1b were expressed as above. Seven days after infection, cells were solubilized in 0.1% Triton X-100 (TX). Equal volumes of soluble and insoluble fractions were separated by SDS-PAGE, and TX-soluble Munc18-1 was measured as percent of total Munc18-1 by quantitative immunoblotting. Solubility of PSD-93 served as control (Supplementary Fig. 13b). Data are means ± SEM (*p < 0.05, **p < 0.01 by Student’s t test; n = 5 independent experiments). c, d Uptake of synaptotagmin-1 antibody during high K+ stimulation. Primary cortical neurons infected at 6 DIV with lentivirus expressing cre recombinase and WT or mutant Munc18-1b were subjected to an antibody uptake assay at 13 DIV in the absence or presence of chemical chaperones. Endocytosed synaptotagmin-1 antibody was quantified by immunostaining (c), via counting the number of pixels > intensity of 15 (d). Data are means ± SEM (*,#p < 0.05, **,##p < 0.01, ***,###p < 0.001 by Student’s t test; n = 4 independent experiments). Scale bar in c = 20 µm
Fig. 7
Fig. 7
Chemical chaperones rescue deficits of mutant UNC-18 in C. elegans. a Locomotion of C. elegans. Body bends per 30 s were counted. Data are means ± SEM (**p < 0.01, ***p < 0.001 by Student’s t test; n = 10 independent experiments on ten worms per experiment). b Rescue of reduced acetylcholine release in worms expressing mutant UNC-18 variants. Young adult worms expressing WT or mutant unc-18 were exposed to aldicarb, and paralysis at 60 min was measured. Data are means ± SEM (#p < 0.05, **,#p < 0.01, ***p < 0.001, by Student’s t test; n = 6 independent experiments on 20 worms per experiment). c Heat-induced paralysis. Indicated worm strains were exposed to 37 °C over a period of 180 min, and paralysis was scored at indicated time points. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by two-way ANOVA, compared to control, n = 5–6 independent experiments on ten worms per experiment). d, e Rescue of reduced acetylcholine release and heat-induced paralysis in CRISPR/Cas9-generated P334L and R405H knock-in worms. Worms were analyzed as described in ac. Data are means ± SEM (*p < 0.05, **p < 0.01, ***p < 0.001 by two-way ANOVA, compared to control, n = 4 independent experiments on ten worms per experiment for d, and n = 4 independent experiments on 20 worms per experiments for e). f Rescue of subcellular localization of UNC-18 in worms expressing mutant UNC-18. C. elegans expressing WT::GFP or G544D::GFP were immobilized, and the ventral nerve cord was imaged. Solid arrowheads point to pairs of bigger puncta, broken arrowheads to single, smaller puncta. Scale bar = 10 µm. g Rescue of reduced acetylcholine release in worms expressing GFP-tagged mutant UNC-18 variants. Experiments were performed as described under (b). Data are means ± SEM (*p < 0.05, ##p < 0.01, ###p < 0.001, by Student’s t test; n = 6 independent experiments on 20 worms per experiment)
Fig. 8
Fig. 8
Model of Munc18-1 dysfunction in encephalopathies and rescue of deficits with chemical chaperones. Munc18-1-linked encephalopathies are caused by a dominant-negative disease mechanism. Heterozygous missense mutations in Munc18-1 cause a reduction in functional Munc18-1 levels significantly below 50%, due to accelerated degradation of misfolded mutant Munc18-1, aggregation of misfolded mutant Munc18-1 that is resistant to cellular clearance, and due to co-aggregation of WT Munc18-1. Chemical chaperones not only shift the unfolded–folded protein equilibrium significantly toward a folded state, but also result in an increase in total Munc18-1 levels. This overall increase in Munc18-1 levels and solubility is sufficient to rescue the Munc18-1-linked neuronal deficits in vitro and in vivo

References

    1. Saitsu H, et al. De novo mutations in the gene encoding STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy. Nat. Genet. 2008;40:782–788. doi: 10.1038/ng.150.
    1. Otsuka M, et al. STXBP1 mutations cause not only Ohtahara syndrome but also West syndrome--result of Japanese cohort study. Epilepsia. 2010;51:2449–2452. doi: 10.1111/j.1528-1167.2010.02767.x.
    1. Carvill GL, et al. GABRA1 and STXBP1: novel genetic causes of Dravet syndrome. Neurology. 2014;82:1245–1253. doi: 10.1212/WNL.0000000000000291.
    1. Epi KC, et al. De novo mutations in epileptic encephalopathies. Nature. 2013;501:217–221. doi: 10.1038/nature12439.
    1. Vatta M, et al. A novel STXBP1 mutation causes focal seizures with neonatal onset. J. Child Neurol. 2012;27:811–814. doi: 10.1177/0883073811435246.
    1. Olson HE, et al. Mutations in epilepsy and intellectual disability genes in patients with features of Rett syndrome. Am. J. Med. Genet. A. 2015;167A:2017–2025. doi: 10.1002/ajmg.a.37132.
    1. Hamdan FF, et al. Intellectual disability without epilepsy associated with STXBP1 disruption. Eur. J. Hum. Genet. 2011;19:607–609. doi: 10.1038/ejhg.2010.183.
    1. Deprez L, et al. Clinical spectrum of early-onset epileptic encephalopathies associated with STXBP1 mutations. Neurology. 2010;75:1159–1165. doi: 10.1212/WNL.0b013e3181f4d7bf.
    1. Milh M, et al. Epileptic and nonepileptic features in patients with early onset epileptic encephalopathy and STXBP1 mutations. Epilepsia. 2011;52:1828–1834. doi: 10.1111/j.1528-1167.2011.03181.x.
    1. Stamberger H, et al. STXBP1 encephalopathy: a neurodevelopmental disorder including epilepsy. Neurology. 2016;86:954–962. doi: 10.1212/WNL.0000000000002457.
    1. Keogh MJ, et al. A novel de novo STXBP1 mutation is associated with mitochondrial complex I deficiency and late-onset juvenile-onset parkinsonism. Neurogenetics. 2015;16:65–67. doi: 10.1007/s10048-014-0431-z.
    1. Donovan LE, et al. Analysis of a membrane-enriched proteome from postmortem human brain tissue in Alzheimer’s disease. Proteom. Clin. Appl. 2012;6:201–211. doi: 10.1002/prca.201100068.
    1. Jacobs EH, Williams RJ, Francis PT. Cyclin-dependent kinase 5, Munc18a and Munc18-interacting protein 1/X11alpha protein up-regulation in Alzheimer’s disease. Neuroscience. 2006;138:511–522. doi: 10.1016/j.neuroscience.2005.11.017.
    1. McTague A, Cross JH. Treatment of epileptic encephalopathies. CNS Drugs. 2013;27:175–184. doi: 10.1007/s40263-013-0041-6.
    1. Aalto MK, Ruohonen L, Hosono K, Keranen S. Cloning and sequencing of the yeast Saccharomyces cerevisiae SEC1 gene localized on chromosome IV. Yeast. 1991;7:643–650. doi: 10.1002/yea.320070613.
    1. Hosono R, et al. The unc-18 gene encodes a novel protein affecting the kinetics of acetylcholine metabolism in the nematode Caenorhabditis elegans. J. Neurochem. 1992;58:1517–1525. doi: 10.1111/j.1471-4159.1992.tb11373.x.
    1. Grone BP, et al. Epilepsy, behavioral abnormalities, and physiological comorbidities in syntaxin-binding protein 1 (STXBP1) mutant zebrafish. PLoS ONE. 2016;11:e0151148. doi: 10.1371/journal.pone.0151148.
    1. Harrison SD, Broadie K, van de Goor J, Rubin GM. Mutations in the Drosophila Rop gene suggest a function in general secretion and synaptic transmission. Neuron. 1994;13:555–566. doi: 10.1016/0896-6273(94)90025-6.
    1. Verhage M, et al. Synaptic assembly of the brain in the absence of neurotransmitter secretion. Science. 2000;287:864–869. doi: 10.1126/science.287.5454.864.
    1. Novick P, Schekman R. Secretion and cell-surface growth are blocked in a temperature-sensitive mutant of Saccharomyces cerevisiae. Proc. Natl Acad. Sci. USA. 1979;76:1858–1862. doi: 10.1073/pnas.76.4.1858.
    1. Hosono R, Kamiya Y. Additional genes which result in an elevation of acetylcholine levels by mutations in Caenorhabditis elegans. Neurosci. Lett. 1991;128:243–244. doi: 10.1016/0304-3940(91)90270-4.
    1. Weimer RM, et al. Defects in synaptic vesicle docking in unc-18 mutants. Nat. Neurosci. 2003;6:1023–1030. doi: 10.1038/nn1118.
    1. Yook KJ, Proulx SR, Jorgensen EM. Rules of nonallelic noncomplementation at the synapse in Caenorhabditis elegans. Genetics. 2001;158:209–220.
    1. Wu MN, Littleton JT, Bhat MA, Prokop A, Bellen HJ. ROP, the Drosophila Sec1 homolog, interacts with syntaxin and regulates neurotransmitter release in a dosage-dependent manner. EMBO J. 1998;17:127–139. doi: 10.1093/emboj/17.1.127.
    1. Toonen RF, et al. Munc18-1 expression levels control synapse recovery by regulating readily releasable pool size. Proc. Natl Acad. Sci. USA. 2006;103:18332–18337. doi: 10.1073/pnas.0608507103.
    1. Chai YJ, et al. Munc18-1 is a molecular chaperone for alpha-synuclein, controlling its self-replicating aggregation. J. Cell. Biol. 2016;214:705–718. doi: 10.1083/jcb.201512016.
    1. Patzke C, et al. Analysis of conditional heterozygous STXBP1 mutations in human neurons. J. Clin. Invest. 2015;125:3560–3571. doi: 10.1172/JCI78612.
    1. Saitsu H, et al. STXBP1 mutations in early infantile epileptic encephalopathy with suppression-burst pattern. Epilepsia. 2010;51:2397–2405. doi: 10.1111/j.1528-1167.2010.02728.x.
    1. Martin S, et al. Increased polyubiquitination and proteasomal degradation of a Munc18-1 disease-linked mutant causes temperature-sensitive defect in exocytosis. Cell Rep. 2014;9:206–218. doi: 10.1016/j.celrep.2014.08.059.
    1. Cote M, et al. Munc18-2 deficiency causes familial hemophagocytic lymphohistiocytosis type 5 and impairs cytotoxic granule exocytosis in patient NK cells. J. Clin. Invest. 2009;119:3765–3773. doi: 10.1172/JCI40732.
    1. Han GA, et al. A pivotal role for pro-335 in balancing the dual functions of Munc18-1 domain-3a in regulated exocytosis. J. Biol. Chem. 2014;289:33617–33628. doi: 10.1074/jbc.M114.584805.
    1. Hu SH, et al. Possible roles for Munc18-1 domain 3a and Syntaxin1 N-peptide and C-terminal anchor in SNARE complex formation. Proc. Natl Acad. Sci. USA. 2011;108:1040–1045. doi: 10.1073/pnas.0914906108.
    1. Xu Y, Su L, Rizo J. Binding of Munc18-1 to synaptobrevin and to the SNARE four-helix bundle. Biochemistry. 2010;49:1568–1576. doi: 10.1021/bi9021878.
    1. Sloviter RS, Nilaver G. Immunocytochemical localization of GABA-, cholecystokinin-, vasoactive intestinal polypeptide-, and somatostatin-like immunoreactivity in the area dentata and hippocampus of the rat. J. Comp. Neurol. 1987;256:42–60. doi: 10.1002/cne.902560105.
    1. Wittner L, et al. Preservation of perisomatic inhibitory input of granule cells in the epileptic human dentate gyrus. Neuroscience. 2001;108:587–600. doi: 10.1016/S0306-4522(01)00446-8.
    1. Rizo J, Sudhof TC. The membrane fusion enigma: SNAREs, Sec1/Munc18 proteins, and their accomplices--guilty as charged? Annu. Rev. Cell Dev. Biol. 2012;28:279–308. doi: 10.1146/annurev-cellbio-101011-155818.
    1. Brenner S. The genetics of Caenorhabditis elegans. Genetics. 1974;77:71–94.
    1. Martin JA, Hu Z, Fenz KM, Fernandez J, Dittman JS. Complexin has opposite effects on two modes of synaptic vesicle fusion. Curr. Biol. 2011;21:97–105. doi: 10.1016/j.cub.2010.12.014.
    1. Heeroma JH, et al. Trophic support delays but does not prevent cell-intrinsic degeneration of neurons deficient for munc18-1. Eur. J. Neurosci. 2004;20:623–634. doi: 10.1111/j.1460-9568.2004.03503.x.
    1. Kaeser PS, et al. RIM proteins tether Ca2+channels to presynaptic active zones via a direct PDZ-domain interaction. Cell. 2011;144:282–295. doi: 10.1016/j.cell.2010.12.029.
    1. Kraszewski K, et al. Synaptic vesicle dynamics in living cultured hippocampal neurons visualized with CY3-conjugated antibodies directed against the lumenal domain of synaptotagmin. J. Neurosci. 1995;15:4328–4342. doi: 10.1523/JNEUROSCI.15-06-04328.1995.
    1. Sheehan P, Zhu M, Beskow A, Vollmer C, Waites CL. Activity-dependent degradation of synaptic vesicle proteins requires rab35 and the ESCRT pathway. J. Neurosci. 2016;36:8668–8686. doi: 10.1523/JNEUROSCI.0725-16.2016.
    1. Loffing J, Moyer BD, Reynolds D, Stanton BA. PBA increases CFTR expression but at high doses inhibits Cl(-) secretion in Calu-3 airway epithelial cells. Am. J. Physiol. 1999;277:L700–L708.
    1. Collins AF, et al. Oral sodium phenylbutyrate therapy in homozygous beta thalassemia: a clinical trial. Blood. 1995;85:43–49.
    1. Du Y, et al. Histone deacetylase 6 regulates cytotoxic alpha-synuclein accumulation through induction of the heat shock response. Neurobiol. Aging. 2014;35:2316–2328. doi: 10.1016/j.neurobiolaging.2014.04.029.
    1. Mimori S, et al. Protective effects of 4-phenylbutyrate derivatives on the neuronal cell death and endoplasmic reticulum stress. Biol. Pharm. Bull. 2012;35:84–90. doi: 10.1248/bpb.35.84.
    1. Tanaka M, et al. Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington disease. Nat. Med. 2004;10:148–154. doi: 10.1038/nm985.
    1. Yu WB, et al. Trehalose inhibits fibrillation of A53T mutant alpha-synuclein and disaggregates existing fibrils. Arch. Biochem. Biophys. 2012;523:144–150. doi: 10.1016/j.abb.2012.04.021.
    1. Fisher RJ, Pevsner J, Burgoyne RD. Control of fusion pore dynamics during exocytosis by Munc18. Science. 2001;291:875–878. doi: 10.1126/science.291.5505.875.
    1. Graham ME, Barclay JW, Burgoyne RD. Syntaxin/Munc18 interactions in the late events during vesicle fusion and release in exocytosis. J. Biol. Chem. 2004;279:32751–32760. doi: 10.1074/jbc.M400827200.
    1. Graham ME, Sudlow AW, Burgoyne RD. Evidence against an acute inhibitory role of nSec-1 (munc-18) in late steps of regulated exocytosis in chromaffin and PC12 cells. J. Neurochem. 1997;69:2369–2377. doi: 10.1046/j.1471-4159.1997.69062369.x.
    1. Schutz D, Zilly F, Lang T, Jahn R, Bruns D. A dual function for Munc-18 in exocytosis of PC12 cells. Eur. J. Neurosci. 2005;21:2419–2432. doi: 10.1111/j.1460-9568.2005.04095.x.
    1. Tsuboi T, Rutter GA. Multiple forms of “kiss-and-run” exocytosis revealed by evanescent wave microscopy. Curr. Biol. 2003;13:563–567. doi: 10.1016/S0960-9822(03)00176-3.
    1. Parisotto D, et al. An extended helical conformation in domain 3a of Munc18-1 provides a template for SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex assembly. J. Biol. Chem. 2014;289:9639–9650. doi: 10.1074/jbc.M113.514273.
    1. Kaushik JK, Bhat R. A mechanistic analysis of the increase in the thermal stability of proteins in aqueous carboxylic acid salt solutions. Protein Sci. 1999;8:222–233. doi: 10.1110/ps.8.1.222.
    1. Colaco CALS, et al. Chemistry of protein stabilization by trehalose. ACS Sym. Ser. 1994;567:222–240. doi: 10.1021/bk-1994-0567.ch014.
    1. Kumar V, Sharma VK, Kalonia DS. Effect of polyols on polyethylene glycol (PEG)-induced precipitation of proteins: Impact on solubility, stability and conformation. Int. J. Pharm. 2009;366:38–43. doi: 10.1016/j.ijpharm.2008.08.037.
    1. Wu P, Bolen DW. Osmolyte-induced protein folding free energy changes. Proteins. 2006;63:290–296. doi: 10.1002/prot.20868.
    1. Singer MA, Lindquist S. Multiple effects of trehalose on protein folding in vitro and in vivo. Mol. Cell. 1998;1:639–648. doi: 10.1016/S1097-2765(00)80064-7.
    1. Diamant S, Eliahu N, Rosenthal D, Goloubinoff P. Chemical chaperones regulate molecular chaperones in vitro and in cells under combined salt and heat stresses. J. Biol. Chem. 2001;276:39586–39591. doi: 10.1074/jbc.M103081200.
    1. Cortez Leonardo, Sim Valerie. The therapeutic potential of chemical chaperones in protein folding diseases. Prion. 2014;8(2):197–202. doi: 10.4161/pri.28938.
    1. Ono K, et al. A chemical chaperone, sodium 4-phenylbutyric acid, attenuates the pathogenic potency in human alpha-synuclein A30P+1A53T transgenic mice. Park. Relat. Disord. 2009;15:649–654. doi: 10.1016/j.parkreldis.2009.03.002.
    1. Sarkar S, et al. Neuroprotective effect of the chemical chaperone, trehalose in a chronic MPTP-induced Parkinson’s disease mouse model. Neurotoxicology. 2014;44:250–262. doi: 10.1016/j.neuro.2014.07.006.
    1. Schaeffer V, Goedert M. Stimulation of autophagy is neuroprotective in a mouse model of human tauopathy. Autophagy. 2012;8:1686–1687. doi: 10.4161/auto.21488.
    1. Wiley JC, Pettan-Brewer C, Ladiges WC. Phenylbutyric acid reduces amyloid plaques and rescues cognitive behavior in AD transgenic mice. Aging Cell. 2011;10:418–428. doi: 10.1111/j.1474-9726.2011.00680.x.
    1. Maestri NE, Brusilow SW, Clissold DB, Bassett SS. Long-term treatment of girls with ornithine transcarbamylase deficiency. N. Engl. J. Med. 1996;335:855–859. doi: 10.1056/NEJM199609193351204.
    1. Rubenstein RC, Zeitlin PL. A pilot clinical trial of oral sodium 4-phenylbutyrate (Buphenyl) in deltaF508-homozygous cystic fibrosis patients: partial restoration of nasal epithelial CFTR function. Am. J. Respir. Crit. Care Med. 1998;157:484–490. doi: 10.1164/ajrccm.157.2.9706088.
    1. Arribere JA, et al. Efficient marker-free recovery of custom genetic modifications with CRISPR/Case9 in Caenorhabdites elegans. Genetics. 2014;198:87–846. doi: 10.1534/genetics.114.169730.
    1. Bakkum DJ, et al. Parameters for burst detection. Front. Comput. Neurosci. 2014;7:193. doi: 10.3389/fncom.2013.00193.

Source: PubMed

3
Předplatit