Autophagy induction enhances TDP43 turnover and survival in neuronal ALS models

Sami J Barmada, Andrea Serio, Arpana Arjun, Bilada Bilican, Aaron Daub, D Michael Ando, Andrey Tsvetkov, Michael Pleiss, Xingli Li, Daniel Peisach, Christopher Shaw, Siddharthan Chandran, Steven Finkbeiner, Sami J Barmada, Andrea Serio, Arpana Arjun, Bilada Bilican, Aaron Daub, D Michael Ando, Andrey Tsvetkov, Michael Pleiss, Xingli Li, Daniel Peisach, Christopher Shaw, Siddharthan Chandran, Steven Finkbeiner

Abstract

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have distinct clinical features but a common pathology--cytoplasmic inclusions rich in transactive response element DNA-binding protein of 43 kDa (TDP43). Rare TDP43 mutations cause ALS or FTD, but abnormal TDP43 levels and localization may cause disease even if TDP43 lacks a mutation. Here we show that individual neurons vary in their ability to clear TDP43 and are exquisitely sensitive to TDP43 levels. To measure TDP43 clearance, we developed and validated a single-cell optical method that overcomes the confounding effects of aggregation and toxicity and discovered that pathogenic mutations shorten TDP43 half-life. New compounds that stimulate autophagy improved TDP43 clearance and localization and enhanced survival in primary murine neurons and in human stem cell-derived neurons and astrocytes harboring mutant TDP43. These findings indicate that the levels and localization of TDP43 critically determine neurotoxicity and show that autophagy induction mitigates neurodegeneration by acting directly on TDP43 clearance.

Conflict of interest statement

STATEMENT OF COMPETING FINANCIAL INTERESTS

We declare that the authors have no competing interests as defined by Nature Publishing Group, or other interests that might be perceived to influence the results and/or discussion reported in this article.

Figures

Figure 1. The toxicity of TDP43 depends…
Figure 1. The toxicity of TDP43 depends strongly on dose
(a, b) Histograms of TDP43(WT)-EGFP and TDP43(A315T)-EGFP levels in individual neurons. Increasing amounts of DNA significantly shifted the distribution of log-transformed expression levels towards higher values (p 2 = 0.9353) or TDP43(A315T)-EGFP (R2 = 0.9574) level. The total TDP43 level was determined by quantitative immunocytochemistry using TDP43-specific antibodies (Supplementary Fig. 3) and normalized to the amount of TDP43 in non-transfected neurons. Dotted lines, reference (HR = 1).
Figure 2. TDP43 turnover in primary neurons
Figure 2. TDP43 turnover in primary neurons
(a) Primary neurons were labeled with 35S-labeled methionine, and endogenous TDP43 was immunoprecipitated using anti-TDP43 antibodies (IP-TDP43) or non-specific IgG antibodies (IP-IgG). (b) Log-normal plot of normalized TDP43 levels. TDP43 half-life was determined by fitting a first-order exponential curve to the data (R2 = 0.8984). Values pooled from five independent experiments. (c) Potential confounders in half-life analyses. Ideally (top), total protein decreases over time in a predictable manner (top right panel). Toxicity (middle) reduces the amount of measured protein, potentially shortening the calculated half-life (middle right panel). Protein aggregation (bottom) might also shorten half-life estimations if the protein cannot be immunoprecipitated (dark arrows, bottom right panel), or lengthen it if the protein is more stable and successfully immunoprecipitated (grey arrows, bottom right panel). (d) Optical pulse labeling. Primary cortical neurons were transfected with EGFP and TDP43(WT)-Dendra2, photoconverted then imaged by AFM. Dead cells (arrow) were excluded at the time of death. (e) The half-life of TDP43(WT)-Dendra2 was determined by fitting a first-order exponential curve to the data (R2 0.9558). (f) Including cells with aggregates prolonged TDP43(WT)-Dendra2 half-life (purple line, R2 = 0.9003; * p < 0.0001, F 15.34, extra sum-of-squares F-test). Excluding cells that died over the 36 h experiment also prolonged half-life (blue line, R2 = 0.7171; ** p < 0.0001, extra sum-of-squares F-test), suggesting that toxicity reduces measured half-life. Error bars represent ± SEM. Values pooled from eight wells per condition, performed in quadruplicate.
Figure 3. Optical pulse labeling of TDP43
Figure 3. Optical pulse labeling of TDP43
(a) Scatter plot depicting half-life measurements for individual cells expressing TDP43(WT)-Dendra2 (cyan, n=206) and TDP43(A315T)-Dendra2 (red, n=251). * p=0.0029, U = 11797, Mann-Whitney test. (b) Probability density plot for single-cell half-life measurements, demonstrating a reduced half-life of TDP43(A315T)-Dendra2 (red, n=251), compared to TDP43(WT)-Dendra2 (cyan, n=206; ** p=0.0007, two-sided Kolmogorov-Smirnov test). Values were pooled from eight wells per condition, performed in quadruplicate. Probability density plots of single-cell half-life measurements for TDP43(WT)-Dendra2 (c) and TDP43(A315T)-Dendra2 (d). At 0.5 μM, all three compounds significantly reduced TDP43(WT)-Dendra2 half-life, compared to vehicle control (DMSO, n=110; FPZ, n=96, *f p=1×10−4; MTM, n=129, *m p=2×10−7; NCP, n=111, *n p=2×10−5, two-sided Kolmogorov-Smirnov test). The compounds also reduced TDP43(A315T)-Dendra2 half-life, in comparison to vehicle control (DMSO, n=120; FPZ, n=155, *f p=2×10−9; MTM, n=152, *m p=0.004; NCP, n=111, *n p=0.005, two-sided Kolmogorov-Smirnov test). Colored hash marks represent values from individual neurons in (b), (c) and (d). Values were obtained from eight wells per condition, performed in triplicate.
Figure 4. Induction of autophagy by a…
Figure 4. Induction of autophagy by a family of small molecules
(a). Each compound significantly increased the LC3-II/GAPDH (b) and LC3-II/LC3-I (c) ratios at 5 μM, but not 0.5 μM [ns, p > 0.05; * 0.05 2 = 0.7333; Beclin, R2 = 0.6968, FPZ, R2 = 0.8752; MTM, R2 = 0.8447; NCP, R2 = 0.8357; * p < 0.0001 for global trend, F = 12.6, extra sum-of-squares F-test). Error bars represent ± SEM. (f) The median single-cell half-life of LC3-Dendra2 was significantly reduced by beclin expression or treatment with 0.5 μM NCP, MTM, and FPZ (* p < 0.01, ** p < 0.001, Kruskal Wallis statistic = 52.56, Kruskal-Wallis with Dunn’s test). (g) Probability density plot of LC3-Dendra2 half-life measurements from individual neurons treated with vehicle control (DMSO, n=140), NCP (n=128), MTM (n=91), or FPZ (n=77), or those expressing beclin (n=101). *n, p=6×10−4; *m, p=1×10−10; *f, p=5×10−9; *b, p=2×10−3; two-sided Kolmogorov-Smirnov test). Colored hash marks represent values from individual neurons. Values pooled from eight wells per condition, performed in triplicate.
Figure 5. Autophagy induction reduces TDP43 levels,…
Figure 5. Autophagy induction reduces TDP43 levels, aggregation, and cytoplasmic mislocalization
(a) FPZ (n=490) and MTM (n=580), but not NCP (n=523), lowered total TDP43(WT)-EGFP levels in comparison to vehicle (DMSO, n=1007). (b) Histogram of normalized, log-transformed TDP43(WT)-EGFP levels. (c) Each compound also reduced TDP43(A315T)-EGFP levels (DMSO, n=983; FPZ, n=550; MTM, n=542; NCP, n=493). (d) Histogram of single-cell TDP43(A315T)-EGFP levels. Asterisks in (b) and (d) denote median values for cells exposed to vehicle (cyan), FPZ (green), MTM (blue) or NPZ (purple). For FPZ, MTM, NCP vs. DMSO in (a) and (d), p < 1×10−3 by two-sided Kolmogorov-Smirnov test. (e) No compound reduced TDP43(WT)-EGFP inclusions 48 h after transfection (DMSO, n=360; FPZ, n=194; MTM, n=234; NCP, n=203). (f) FPZ and MTM, but not NCP, reduced TDP43(A315T)-EGFP inclusions at 48 h (DMSO, n=327; FPZ, n=226; MTM, n=262; NCP, n=253). (g) No compound affected TDP43(WT)-EGFP localization 24 h after transfection (DMSO, n=256; FPZ, n=212; MTM, n=250; NCP, n=241). Each compound (h) prevented TDP43(A315T)-EGFP cytoplasmic mislocalization (DMSO, n=233; FPZ, n=268; MTM, n=263; NCP, n=270) and (i) reduced cytoplasmic TDP43(A315T)-EGFP levels (DMSO, n=125; FPZ, n=102; MTM, n=121; NCP, n=144. * p < 0.05; ns, p > 0.05). For (a), (c), and (e–i), * p < 0.05; ns, p > 0.05, one-way ANOVA with Dunnett’s test. Error bars represent ± SEM. Values in (a–d) pooled from 8–12 wells per condition, from six experiments; values in (e–h) pooled from eight wells per condition, in triplicate; values in (i) pooled from five wells per condition, in duplicate.
Figure 6. Autophagic stimulation improves survival in…
Figure 6. Autophagic stimulation improves survival in neuronal and astrocyte models of ALS
Primary neurons transfected with TDP43(WT)-EGFP (a) or TDP43(A315T)-EGFP (b) were treated with autophagy inducers and survival determined by AFM. Data obtained from eight wells per condition, performed in triplicate. See Supplementary Table 2 for the number of neurons per condition, hazard ratios, 95% CI, and p values as determined by Cox hazards analysis. (c) Autophagic induction improved survival in TDP43 M337V HB9-positive human iPSC-derived MNs. (d) FPZ and MTM also reduced the risk of death in MAP2-positive human iPSC-derived MNs Values in (c) and (d) pooled from 18 wells per condition, performed in triplicate. (e) Astrocytes differentiated from human iPSCs exhibit mutant TDP43-related toxicity that is mitigated by FPZ and MTM. Values pooled from 18 wells per condition, performed in triplicate. See Supplementary Table 3 for the number of human iPSC-derived neurons and astrocytes per condition, hazard ratios, 95% CI, and p values as determined by Cox hazards analysis.

References

    1. Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci. 2013;14:248–264.
    1. Neumann M. Molecular Neuropathology of TDP-43 Proteinopathies. Int J Mol Sci. 2009;10:232–246.
    1. van Blitterswijk M, Landers JE. RNA processing pathways in amyotrophic lateral sclerosis. Neurogenetics. 2010 doi: 10.1007/s10048-010-0239-4.
    1. Ayala YM, et al. Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci. 2008;121:3778–3785.
    1. Moisse K, et al. Cytosolic TDP-43 expression following axotomy is associated with caspase 3 activation in NFL−/− mice: support for a role for TDP-43 in the physiological response to neuronal injury. Brain Res. 2009;1296:176–186.
    1. Dewey CM, et al. TDP-43 is directed to stress granules by sorbitol, a novel physiological osmotic and oxidative stressor. Mol Cell Biol. 2010 doi: 10.1128/MCB.01279-10.
    1. Davidson YS, et al. TDP-43 pathological changes in early onset familial and sporadic Alzheimer’s disease, late onset Alzheimer’s disease and Down’s Syndrome: association with age, hippocampal sclerosis and clinical phenotype. Acta Neuropathol. 2011;122:703–713.
    1. Barmada SJ, Finkbeiner S. Pathogenic TARDBP mutations in amyotrophic lateral sclerosis and frontotemporal dementia: disease-associated pathways. Rev Neurosci. 2010;21:251–272.
    1. Barmada SJ, et al. Cytoplasmic mislocalization of TDP-43 is toxic to neurons and enhanced by a mutation associated with familial amyotrophic lateral sclerosis. J Neurosci. 2010;30:639–649.
    1. Tsao W, et al. Rodent models of TDP-43: Recent advances. Brain Res. 2012:1–14.
    1. Roberson ED. Mouse models of frontotemporal dementia. Ann Neurol. 2012;72:837–849.
    1. Ling SC, et al. ALS-associated mutations in TDP-43 increase its stability and promote TDP-43 complexes with FUS/TLS. Proc Natl Acad Sci USA. 2010;107:13318–13323.
    1. Watanabe S, Kaneko K, Yamanaka K. Accelerated disease onset with stabilized familial Amyotrophic Lateral Sclerosis (ALS)-linked TDP-43 mutations. J Biol Chem. 2013;288:3641–3654.
    1. Roscic A, Baldo B, Crochemore C, Marcellin D, Paganetti P. Induction of autophagy with catalytic mTOR inhibitors reduces huntingtin aggregates in a neuronal cell model. J Neurochem. 2011;119:398–407.
    1. Prakash A, Levy DE. Regulation of IRF7 through cell type-specific protein stability. Biochem Biophys Res Comm. 2006;342:50–56.
    1. Wang X, et al. Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system. Neurosci Lett. 2010;469:112–116.
    1. Urushitani M, Sato T, Bamba H, Hisa Y, Tooyama I. Synergistic effect between proteasome and autophagosome in the clearance of polyubiquitinated TDP-43. J Neurosci Res. 2010;88:784–797.
    1. Zhang X, et al. Rapamycin treatment augments motor neuron degeneration in SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Autophagy. 2011;7:412–425.
    1. Hetz C, et al. XBP-1 deficiency in the nervous system protects against amyotrophic lateral sclerosis by increasing autophagy. Genes Dev. 2009;23:2294–2306.
    1. Wang IF, et al. Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies of the TAR DNA-binding protein 43. Proc Natl Acad Sci USA. 2012;109:15024–15029.
    1. Fox JH, et al. The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease. Mol Neurodegen. 2010;5:26.
    1. Tsvetkov AS, et al. A small-molecule scaffold induces autophagy in primary neurons and protects against toxicity in a Huntington disease model. Proc Natl Acad Sci USA. 2010;107:16982–16987.
    1. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274–293.
    1. Gitcho MA, et al. TDP-43 A315T mutation in familial motor neuron disease. Ann Neurol. 2008;63:535–538.
    1. Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004;431:805–810.
    1. Xu YF, et al. The Pathological Phenotypes of Human TDP-43 Transgenic Mouse Models Are Independent of Downregulation of Mouse Tdp-43. PLoS ONE. 2013;8:e69864.
    1. Chudakov DM, Lukyanov S, Lukyanov KA. Tracking intracellular protein movements using photoswitchable fluorescent proteins PS-CFP2 and Dendra2. Nat Protoc. 2007;2:2024–2032.
    1. Wong E, Cuervo AM. Integration of clearance mechanisms: the proteasome and autophagy. Cold Spring Harb Perspect Biol. 2010;2:a006734.
    1. Klionsky DJ, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4:151–175.
    1. Wang J. Beclin 1 bridges autophagy, apoptosis and differentiation. Autophagy. 2008;4:947–948.
    1. Kim J, Dalton VM, Eggerton KP, Scott SV, Klionsky DJ. Apg7p/Cvt2p is required for the cytoplasm-to-vacuole targeting, macroautophagy, and peroxisome degradation pathways. Mol Biol Cell. 1999;10:1337–1351.
    1. Bilican B, et al. Mutant induced pluripotent stem cell lines recapitulate aspects of TDP-43 proteinopathies and reveal cell-specific vulnerability. Proc Natl Acad Sci USA. 2012;109:5803–5808.
    1. Singh Roy N, et al. Enhancer-specified GFP-based FACS purification of human spinal motor neurons from embryonic stem cells. Exp Neurol. 2005;196:224–234.
    1. Geser F, et al. Clinical and pathological continuum of multisystem TDP-43 proteinopathies. Arch Neurol. 2009;66:180–189.
    1. Kosik KS, Finch EA. MAP2 and tau segregate into dendritic and axonal domains after the elaboration of morphologically distinct neurites: an immunocytochemical study of cultured rat cerebrum. J Neurosci. 2001;7:3142–3153.
    1. Boillée S, Vande Velde C, Cleveland DW. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron. 2006;52:39–59.
    1. Serio A, et al. Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc Natl Acad Sci USA. 2013 doi: 10.1073/pnas.1300398110.
    1. Egawa N, et al. Drug Screening for ALS Using Patient-Specific Induced Pluripotent Stem Cells. Sci Transl Med. 2012;4:145ra104–145ra104.
    1. Stavrovskaya IG. Clinically Approved Heterocyclics Act on a Mitochondrial Target and Reduce Stroke-induced Pathology. J Exp Med. 2004;200:211–222.
    1. Szczudlik A, Tomik B, Słowik A, Kasprzyk K. Assessment of the efficacy of treatment with pimozide in patients with amyotrophic lateral sclerosis. Introductory notes. Neurol Neurochir Pol. 1998;32:821–829.
    1. Krauss S, et al. Translation of HTT mRNA with expanded CAG repeats is regulated by the MID1–PP2A protein complex. Nat Comm. 1AD;4:1511–9.
    1. Miller J, et al. Quantitative Relationships between Huntingtin Levels, Polyglutamine Length, Inclusion Body Formation, and Neuronal Death Provide Novel Insight into Huntington’s Disease Molecular Pathogenesis. J Neurosci. 2010;30:10541–10550.
    1. Finkbeiner S. Bridging the Valley of Death of therapeutics for neurodegeneration. Nat Med. 2010;16:1227–1232.
    1. Dolmetsch R, Geschwind DH. The Human Brain in a Dish: The Promise of iPSC-Derived Neurons. Cell. 2011;145:831–834.
    1. Jacquier A, Bellouze S, Blanchard S, Bohl D, Haase G. Astrocytic protection of spinal motor neurons but not cortical neurons against loss of Als2/alsin function. Hum Mol Genet. 2009;18:2127–2139.
    1. Haidet-Phillips AM, et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat Biotechnol. 2011;29:824–828.
    1. Ko LW, Ko HHC, Lin WL, Kulathingal JG, Yen SHC. Aggregates assembled from overexpression of wild-type alpha-synuclein are not toxic to human neuronal cells. J Neuropathol Exp Neurol. 2008;67:1084–1096.
    1. Mucke L, et al. High-level neuronal expression of abeta 1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci. 2000;20:4050–4058.
    1. Tsvetkov AS, et al. Proteostasis of polyglutamine varies among neurons and predicts neurodegeneration. Nat Chem Biol. 2013;9:586–592.
    1. Saudou F, Finkbeiner S, Devys D, Greenberg ME. Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell. 1998;95:55–66.

Source: PubMed

3
S'abonner