Mol. Cells 2019; 42(6): 480-494
Published online June 15, 2019
https://doi.org/10.14348/molcells.2019.0091
© The Korean Society for Molecular and Cellular Biology
Correspondence to : *ylee69@skku.edu
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Aggregates of disease-causing proteins dysregulate cellular functions, thereby causing neuronal cell loss in diverse neurodegenerative diseases. Although many
Keywords α-synuclein, amyloid, fibril, natural compound screen, neurodegenerative disease, peucedanocoumarin III, Tet-Off model
Dysregulation of intracellular protein homeostasis is associated with diverse neurodegenerative diseases (Ross and Poirier, 2004). The aggregation of disease-related proteins (i.e., α-synuclein, polyglutamine (polyQ) repeat expansion-containing huntingtin, TAR DNA-binding protein [TDP43]) can be increased by disease-linked mutations or environmental stresses, thereby affecting the viability of neuronal subtypes in many brain disorders (Brahmachari et al., 2016; Gorbatyuk et al., 2008; Landles and Bates, 2004; Mahul-Mellier et al., 2014; Scherzinger et al., 1997; Scotter et al., 2015; Spillantini and Goedert, 2000). Interestingly, amyloid-like aggregates of disease proteins, including α-synuclein, huntingtin, and TDP43, are localized in the cytoplasm and nucleus (Goers et al., 2003; Kontopoulos et al., 2006; Rousseaux et al., 2016; Scherzinger et al., 1997; Scotter et al., 2014; Wyttenbach et al., 2000), which suggests differential proteinopathic disruption of cellular functions. A recent study showed that cytoplasmic protein aggregates impair nucleo-cytoplasmic protein transport (Woerner et al., 2016), thereby causing robust cellular toxicity. Moreover, pathological nuclear α-synuclein (Goers et al., 2003; Kontopoulos et al., 2006; Rousseaux et al., 2016) and huntingtin (Cui et al., 2006) have been reported to alter gene transcription. Since both cytosolic and nuclear protein aggregates have detrimental effects on cellular function and viability, a therapeutic strategy to control aggregates in both compartments may be important for controlling pathological processes in neurodegenerative diseases, including Parkinson’s disease (PD).
α-Synuclein is a major component of Lewy body inclusions in PD and other α-synucleinopathies (Norris et al., 2004; Spillantini and Goedert, 2000). Recently, pathologies of diseases related to α-synuclein oligomers or fibrils have been shown to propagate via neuron-to-neuron transmission (Luk et al., 2012; Mao et al., 2016). Several natural compounds have been identified and found to inhibit α-synuclein aggregation or convert toxic oligomer or fibril conformations into nontoxic species (Jha et al., 2016; Masuda et al., 2006; Singh et al., 2013). In most cases,
In this study, we established a tetracycline (Tet)-Off cell model expressing nuclear β-sheet amyloid aggregates (nuclear β23, as named in previous studies [Olzscha et al., 2011; Woerner et al., 2016]). β23 was initially developed to aid in the investigation molecular mechanisms of toxicity induced by disease-associated amyloid aggregates (Olzscha et al., 2011). β23 is an artificial protein designed to self-assemble into fibrils with repeated β strands of alternating patterns of polar and nonpolar residues (Olzscha et al., 2011). In the previous study, amyloid aggregate expression of β23 aided in the investigation of sequestration and dysregulation of functionally important endogenous proteins as molecular mechanisms of amyloid-induced cell toxicity (Olzscha et al., 2011). Using Tet-inducible expression and cellular toxicity as readouts, we identified several nuclear β23 inhibitors, including peucedanocoumarin III (PCIII). PCIII enhanced clearance of nuclear, as well as cytosolic, β23 aggregates and prevented the aggregation and toxicity of disease-related proteins (i.e., mutant huntingtin and α-synuclein). Significantly,
The National Development Institute of Korean Medicine (NIKOM) provided the natural compound library, which contained 640 natural compounds of > 80% purity (1 mg/ml). This library was used for nuclear β23 inhibitor high-throughput screening. Natural compounds blocking β23 toxicity (i.e., PCIII, kaempferol-7-O-α-L-rhamnopyranoside, oregonin, and ophiocarpine) were extracted from herbal medications, purified, and validated using high-performance liquid chromatography (HPLC). Thioflavin S, Thioflavin T, 6-OHDA, doxycycline, Alamar blue, trypan blue, MG132, and carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (Z-VAD-FMK) were purchased from Sigma (USA). Doxorubicin was purchased from Selleck Chemicals. The primary antibodies used in this study were mouse antibody to hemagglutinin (HA) (12CA5, 1:1,000; Roche, Switzerland), mouse antibody to FLAG (M2, 1:5,000; Sigma), mouse antibody to α-synuclein (1:3,000; BD Transduction Laboratories, USA), rabbit antibody to green fluorescent protein (GFP) (cat# 2956, 1:5,000; Cell Signaling Technology, USA) mouse antibody to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (GT239, 1:5,000; GeneTex, USA), mouse antibody to poly (ADP-ribose) polymerase 1 (PARP1) (cat# 556494, 1:1,000; BD Bioscience, USA), conformation specific rabbit antibody to α-synuclein filaments (MJFR-14-6-4-2, cat# ab209538, 1:5,000; Abcam, USA) and horseradish peroxidase (HRP)-conjugated mouse antibody to β-actin (AC15; Sigma-Aldrich, USA). The secondary antibodies used were HRP-conjugated sheep antibody to mouse immunoglobulin G (IgG) (cat# RPN4301, 1:5,000; GE Healthcare, USA), HRP-conjugated donkey antibody to rabbit IgG (cat# RPN4101, 1:5,000; GE Healthcare), Alexa Fluor 488-conjugated donkey antibody to mouse IgG (H + L) (cat# A21202, 1:1,000; Invitrogen, USA), Alexa Fluor 568-conjugated donkey antibody to mouse IgG (cat# A10037, 1:1,000; Invitrogen), and Alexa Fluor 647-conjugated donkey antibody to mouse IgG (cat# A31571, 1:1,000; Invitrogen).
The double-strand oligos encoding nuclear β23, β23, and nuclear αS824 sequence were cloned into a pTRE-Dual2 plasmid (Clontech Laboratories, USA). The full sequence of nuclear β23 with tags (NLS-FLAG-β23-HA) is as follows:
ATGCCAAAGAAGAAGCGGAAGGTCGGTTGCGACTACAAGGACGACGACGACAAGGGCATGCAGATCTCCATGGACTACAACATCCAGTTCCACAACAACGGCAACGAGATCCAGTTCGAGATCGACGACTCCGGCGGCGACATCGAGATCGAGATCCGGGGCCCCGGCGGCCGGGTGCACATCCAGCTGAACGACGGCCACGGCCACATCAAGGTGGACTTCAACAACGACGGCGGCGAGCTGCAGATCGACATGCACTACCCATACGACGTCCCAGACTACGCT.
The full DNA and amino acid sequence of β23 with tags (FLAG-β23-HA) is as follows:
ATGTGCGACTACAAGGACGACGACGACAAGGGCATGCAGATCTCCATGGACTACAACATCCAGTTCCACAACAACGGCAACGAGATCCAGTTCGAGATCGACGACTCCGGCGGCGACATCGAGATCGAGATCCGGGGCCCCGGCGGCCGGGTGCACATCCAGCTGAACGACGGCCACGGCCACATCAAGGTGGACTTCAACAACGACGGCGGCGAGCTGCAGATCGACATGCACTACCCATACGACGTCCCAGACTACGCTTAA;
MCDYKDDDDKGMQISMDYNIQFHNNGNEIQFEIDDSGGDIEIEIRGPGGRVHIQLNDGHGHIKVDFNNDGGELQIDMHYPYDVPDYA.
The full DNA and amino acid sequence of nuclear αS824 with tags (NLS-FLAG-αS824 -HA) is as follows:
ATGCCAAAGAAGAAGCGGAAGGTCGGTTGCGACTACAAGGACGACGACGACAAGGGCATGTACGGCAAGCTGAACGACCTGCTGGAGGACCTGCAGGAGGTGCTGAAGCACGTGAACCAGCACTGGCAGGGCGGCCAGAAGAACATGAACAAGGTGGACCACCACCTGCAGAACGTGATCGAGGACATCCACGACTTCATGCAGGGCGGCGGCTCCGGCGGCAAGCTGCAGGAGATGATGAAGGAGTTCCAGCAGGTGCTGGACGAGATCAAGCAGCAGCTGCAGGGCGGCGACAACTCCCTGCACAACGTGCACGAGAACATCAAGGAGATCTTCCACCACCTGGAGGAGCTGGTGCACCGGTACCCATACGACGTCCCAGACTACGCTTGA;
MPKKKRKVGCDYKDDDDKGMYGKLNDLLEDLQEVLKHVNQHWQGGQKNMNKVDHHLQNVIEDIHDFMQGGGSGGKLQEMMKEFQQVLDEIKQQLQGGDNSLHNVHENIKEIFHHLEELVHRYPYDVPDYA.
Construct integrity was verified by sequencing. Plasmid cytomegalovirus (pCMV)-tetracycline transactivator (tTA)was purchased from Clontech and the pTreTight-Htt94Q-CFP (Maynard et al., 2009) construct was purchased from Addgene in USA (Plasmid #23966). The HA-α-synuclein construct was generated as previously described (Brahmachari et al., 2016).
In March 2013,
HPLC analysis was performed on an Agilent 1260 system, equipped with an evaporative light-scattering detector (ELSD) and a Kinetex C18 column (4.6 × 150 mm; Phenomenex, USA). The mobile phase consisted of 0.1% trifluoroacetic acid in water (v/v) (A) and acetonitrile (B). The injection volume was 3 μl and flow rate was kept at 0.5 ml/min for a total run time of 30 min. The gradient program was as follows: 2% (start, B), 5% (0–3 min, B), 100% (3–20 min, B), 2% (20–23 min, B), and kept at 2% (B) until the end of the run at 30 min. HPLC analysis found PCIII purity to be 99.9% (Fig. 1).
1H and 13C NMR spectra were recorded on a Jeol ECA-500 MHz NMR instrument, operating at 500 MHz for 1H NMR and 125 MHz for 13C NMR (JEOL, Japan) with tetramethylsilane as the internal standard. The PCIII structure observed was compared with that reported in previous literature (Chen et al., 1996; Takata et al., 1990).
PCIII: colorless needles; ESI-MS
Human neuroblastoma SH-SY5Y cells (ATCC [American Type Culture Collection], USA) or HEK-293T cells were grown in DMEM containing 10% fetal bovine serum (FBS) (vol/vol) and antibiotics in a humidified 5% CO2 atmosphere at 37°C. X-tremeGENE HP transfection reagent (Roche) was used for transient transfections according to the manufacturer’s instructions. For α-synuclein PFF uptake experiments, SH-SY5Y cells were incubated for 4 days in complete media containing PFF (5 μg/ml). PFF was replaced every 2 days. SH-SY5Y cells were briefly washed with phosphate-buffered saline (PBS) and incubated in the presence of natural compounds for an indicated duration followed by western blot analysis of remnant PFF in cell lysates.
SH-SY5Y cells were fractionated into cytosol and nucleus fractions using the Qproteome Mitochondria Isolation Kit (Qiagen, Germany) according to manual instructions. Cytosolic fractions were further concentrated with acetone precipitation. The purity of each fraction was validated with western blots using antibodies to marker proteins for cytosolic (GAPDH, 2% of total cytosolic fraction) and nuclear (PARP1, 5% of total nuclear fraction) fractions.
As outlined in Figure 2C, HEK-293T cells were cotransfected with pCMV-tTA and TetP-nuclear β23 constructs. Cells were kept on doxycycline (200 ng/ml; Sigma)-containing complete media to prevent toxic expression of nuclear β23. After 2 days, transfected HEK-293T cells were plated onto 96-well white flat-bottom plates (Microtiter; Thermo Scientific, USA) at 80% confluency in 100 μl of DMEM containing 10% FBS with penicillin/streptomycin (P/S) and without doxycycline for nuclear β23 induction. After 24 h, each compound (i.e., 640 natural compounds extracted and purified by NIKOM; 0.35 μg/ml each) was added to a well. For homogeneous treatment of each compound, 50 μl of culture media were removed and 50 μl of 2X compound in complete media were then added to each well. After 48 h incubation, cell viability was measured using Alamar blue assay. Briefly, 10 μl of filtered Alamar blue solution (1 mg/ml resazurin in DW filtered through a 0.22 μM pore filter) was added to the 100 μl culture media in 96-well plates. Cell viability was determined by fluorescence reading using a microplate fluorescence spectrometer (excitation: 530 nm, emission: 590 nm; SYNERGYneo microplate reader [BioTek, USA]). Each plate had four wells of the blank with no cell culture, positive controls (dimethyl sulfoxide [DMSO] vehicle treatment with β23 induction) and negative controls (no induction of β23 by maintaining in doxycycline [200 ng/ml]-containing media). To assess the appropriateness of our assay for high-throughput screening, the Z′ factor was quantified as follows:
Z′ factor = 1 − 3(SDp + SDn) / (MEANp − MEANn), where MEANp and MEANn are the means of the positive and negative controls, respectively, and SDp and SDn are the standard deviations of the positive and negative controls, respectively. A high-throughput screening-ready assay should have a Z′ factor between 0.5 and 1.
For Triton X-100-soluble and -insoluble fraction separation, SH-SY5Y cells were harvested and processed into nonionic detergent-soluble and detergent-insoluble fractions in lysis buffer containing PBS, 1% Triton X-100, Phosphatase Inhibitor Cocktail II and III (Sigma-Aldrich), and a complete protease inhibitor mixture. The lysates were centrifuged at 100,000
SH-SY5Y or HEK-293T cells were plated onto poly-D-lysine-coated coverslips at 10,000 cells/cm2. Following experimental procedures, cells were fixed with 4% paraformaldehyde in PBS and blocked in a solution containing 5% normal donkey serum (Jackson ImmunoResearch, USA), 2% BSA (Sigma), and 0.1% Triton X-100 (Sigma) for 1 h at room temperature. Samples were then incubated overnight with corresponding primary antibodies against proteins of interest at 4°C. Briefly, cells grown on coverslips were washed with PBS containing 0.1% Triton X-100 and incubated for 1 h with fluorescence-conjugated secondary antibodies (1:500; Invitrogen) at room temperature. The coverslips were mounted with 4′, 6-diamidino-2-phenylindole (DAPI). Fluorescent images were obtained using a fluorescence microscope (Axiovert, 200 M; Carl Zeiss, Germany) or confocal microscope (TCS SP8 STED; Leica Microsystems, Germany) for four channel fluorescence image acquisition. For thioflavin S staining, thioflavin S (500 mM) was dissolved in 50% ethanol. Fixed SH-SY5Y cells were then stained with thioflavin S for 7 min. As a differentiation step to remove nonspecific binding of the dye, a slide was soaked in 100%, 95%, and 90% ethanol solutions for 10 s each and then transferred to PBS. Subsequent blocking and primary antibody binding steps were followed as described above for immunofluorescence staining.
Total RNA was extracted with QIAzol Lysis Reagent (cat# 79306; Qiagen) and then treated with DNase I to eliminate trace DNA contamination. cDNA was synthesized from total RNA (1.5 μg) using a first-strand cDNA synthesis kit (iScript cDNA synthesis kit; Bio-Rad, USA). The relative quantities of β23 mRNA expression were analyzed using real-time PCR (QuantStudio 6 flex Real-Time PCR System; Applied Biosystems, USA). SYBR Green PCR master mix (cat# 4309155; Applied Biosystems) was used according to the manufacturer’s instructions. The relative mRNA expression levels of β23 were calculated by the ∆∆Ct method (Livak and Schmittgen, 2001) using GAPDH as an internal loading control. The primer sequences for real-time gene amplification are as follows:
SH-SY5Y cells were plated in 6-well plates at a density of 0.5 × 106 cells per well. Following transient transfection with indicated constructs, cells were grown in DMEM containing low serum (2.5% FBS) for indicated days of α-synuclein, Tet-regulated β23, or mutant huntingtin (Htt94Q-CFP) expression. For α-synuclein toxicity, SH-SY5Y cells were treated with 6-OHDA or PBS vehicle control. SH-SY5Y cells were trypsinized to yield single-cell suspensions that were washed twice with PBS before resuspension in serum-free DMEM. Resuspended cells were mixed with an equal volume of 0.4% trypan blue (wt/vol) and incubated for 2 min at room temperature. Live and dead cells were analyzed automatically using the Countess II Automated Cell Counter (Life Technologies, USA). Cell Counting Kit-8 (CCK8) (Dojindo Molecular Technologies, USA) was also used to assess viability of HEK-293T cells by following the manufacturer’s instructions.
Plasmid pRK172-human α-synuclein was transformed to BL21-competent
To investigate the disaggregating effects of PCIII against α-synuclein PFFs, α-synuclein fibrils (50 μM) prepared from the 7 days incubation of monomeric α-synuclein were subjected to
Recombinant human α-synuclein from
Quantitative data are presented as the mean ± standard error of the mean (SEM). Statistical significance was assessed with either an unpaired two-tailed Student’s
To establish a cellular model of nuclear amyloid accumulation, we generated a Tet-Off construct capable of expressing artificial strands of β-sheet structure with an N-terminal nuclear localization sequence (NLS), FLAG tag and C-terminal influenza HA tag (Fig. 1A). In this manuscript, this construct is subsequently called nuclear β23, following previous studies in which β23’s amyloid-like structure has been characterized (Olzscha et al., 2011; Woerner et al., 2016). Since β23 and reporter protein mCherry expression is under the control of a tetracycline-responsive promoter (TetP-β23), coexpression of a tTA is required for the activation of the Tet promoter and subsequent transcription of mCherry-internal ribosome entry site (IRES)-β23 (Fig. 1B). As designed, cotransfection with pCMV-tTA and TetP-nuclear β23 resulted in the expression of nuclear β23 and fluorescent reporter mCherry in HEK-293T cells (Fig. 1C,
Following this, we assessed whether nuclear expression of amyloid β23 is toxic to HEK-293T cells. HEK-293T cells were transfected with pCMV-tTA and TetP-β23 constructs and incubated in complete media containing either doxycycline or vehicle as a control. In nuclear β23-expressing cells cultured for 2 days in the absence of doxycycline, cell viability measured by Alamar blue assay decreased to about 50% (Fig. 2A). Since the cell viability assay provided consistent and robust toxicity results, we used the Tet-Off nuclear β23 expression system to screen natural compounds that could potentially inhibit nuclear aggregate toxicity. For timed expression of β23, HEK-293T cells were transfected with pCMV-tTA and TetP-nuclear β23 and plated onto 96-well plates in the presence of doxycycline to suppress nuclear β23 expression (Figs. 2B and 2C). After plating, cells were treated with 640 natural compounds extracted and purified from clinically prescribed herbal medications (0.35 μg/ml). On the following day, nuclear β23 expression was induced by removing doxycycline. After 2 days of β23 induction, high-throughput cell viability was assessed by reading Alamar blue fluorescence (Fig. 2D). Z′ factors calculated for each plate using Alamar blue fluorescence intensities for negative (doxycycline treatment) and positive (no doxycycline + DMSO vehicle) controls were between 0.5 and 1.0 (
To determine whether cell death inhibition by these novel compounds is rather selective against toxic protein aggregation, an additional toxic insult (doxorubicin) was introduced into HEK-293T cells; after which, the cytoprotective effect of β23 inhibitors was examined (
To assess the potential central nervous system applications for inhibitors of nuclear protein aggregation toxicity, we used the SH-SY5Y neuroblastoma cell line to examine the cytoprotective functions of select compounds. Transient transfection with pCMV-tTA and TetP-nuclear β23 in SH-SY5Y cells led to the expression of nuclear amyloid β23 and reporter protein mCherry (Fig. 3A). Nuclear β23 expression for 24 h in SH-SY5Y cells resulted in approximately 80% cell toxicity (Fig. 3B). All compounds (PCIII, kaempferol, oregonin, and ophiocarpine) increased SH-SY5Y cell viability against nuclear β23 induction, with PCIII providing maximal protection (up to 60% cell viability; Fig. 3B). Following this, we investigated whether these compounds improved cell viability against nuclear β23 toxicity through the alteration of protein aggregate clearance. To monitor β23 clearance over time, SH-SY5Y cells were transfected with pCMV-tTA and TetP-nuclear β23 (Fig. 3B; top panel). Twenty-four hours after transfection, doxycycline was added to stop further β23 and mCherry transcription in SH-SY5Y cells, and β23 stability was assessed by immunofluorescence using anti-FLAG antibodies. Immunofluorescence showed that PCIII treatment markedly increased the removal of FLAG-tagged β23 from the nucleus, whereas β23 still remained in the vehicle treatment control for 37 h following the addition of doxycycline (Figs. 3C and 3D). To a lesser extent, oregonin facilitated the removal of nuclear FLAG-tagged β23 (Figs. 3C and 3D). In contrast, despite their cytoprotective ability, kaempferol and ophiocarpine had no effect on β23 degradation. These compounds had no effect on reporter protein expression and stability as shown by mCherry fluorescence intensity (Fig. 3C). Moreover, transcription of β23 mRNA was not altered by β23 inhibitors (Fig. 3E).
Regulation of the stability of nuclear β23 by PCIII was further determined by quantitative western blot. Immunoblot results showed no difference in β23 levels over time, which suggests that nuclear amyloid β23 was highly stable and resistant to degradation (Figs. 3F and 3G). Consistent with the immunofluorescence results, PCIII treatment led to approximately 70% degradation of β23 (Figs. 3F and 3G). Taken together, these results show that PCIII’s inhibition of nuclear β23-induced toxicity correlates with its ability to accelerate the degradation of nuclear amyloid β23, which is otherwise highly stable and cannot be cleared via an intrinsic degradation machinery.
Afterwards, the mechanisms of PCIII-mediated nuclear β23 clearance in SH-SY5Y cells were elucidated. Tet-Off expression construct of β23 with no NLS sequence (TetP-β23) was generated to assess whether PCIII’s cytoprotective effect on amyloid toxicity is specific to nuclear aggregates. SH-SY5Y cells were co-transfected with pCMV-tTA and TetP-β23 constructs. β23 was expressed for the initial 24 h and further expression was terminated by doxycycline treatment. Cell viability was assessed 24 h after doxycycline treatment. Expression of β23 amyloid in SH-SY5Y cells resulted in robust cell death which was largely prevented by PCIII treatment (
Amyloid selectivity of PCIII-mediated protein degradation was examined by generating an artificial protein containing an alpha helix structure (αS824), following previous reports (Olzscha et al., 2011; Woerner et al., 2016). Tet-Off construct of αS824 was cloned with NLS and FLAG sequence in the N terminus and HA tag in the C terminus (TetP-nuclear αS824). In contrast to aggregate-forming β23, αS824 was distributed to the soluble fraction (
Next, amyloid clearance facilitated by PCIII was investigated. The proteasome inhibitor, MG132, was treated for 24 h during clearance of nuclear β23 or αS824 in the presence or absence of PCIII (
Of the compounds that were cytoprotective against nuclear and cytosolic aggregates, PCIII demonstrated the greatest protective and disaggregative effects. PCIII extracted and purified from
To determine PCIII’s therapeutic effects on disease-associated proteins, we selected a cellular model of mutant huntingtin aggregation, which is linked to hyperkinetic movement disorders in humans, such as Huntington’s disease (Scherzinger et al., 1997). To examine whether PCIII can regulate nuclear huntingtin aggregation in cells, we transfected SH-SY5Y cells with pCMV-tTA and 94 polyglutamine repeat-containing huntingtin protein (TetP-Htt94Q-CFP) to induce aggregate formation (Fig. 4A). A proteasome inhibitor (MG132) was added to facilitate nuclear aggregate formation as previously described (Maynard et al., 2009; Wyttenbach et al., 2000). Separation of protein lysates into detergent (1% Triton X-100)-soluble and -insoluble fractions demonstrated that PCIII treatment markedly prevented insolubility of mutant huntingtin aggregates and redistributed huntingtin into soluble fractions (Figs. 4B and 4C). Consistent with western blot results, immunofluorescence demonstrated that PCIII treatment of SH-SY5Y cells was associated with substantial removal of cytosolic and nuclear aggregate puncta formed because of Htt94Q expression (Figs. 4D–F). Moreover, the growth of mutant huntingtin aggregate size in DMSO-treated controls was inhibited or diminished by PCIII treatment in the nucleus and cytoplasm, respectively (Figs. 4E and 4F). Mutant huntingtin aggregate expression led to 80% cell death in SH-SY5Y cells; however, PCIII treatment improved cell viability by approximately 20% (Fig. 4G).
After observing the promising effects of PCIII on amyloid β23 and huntingtin aggregate toxicity, we tested PCIII’s protective effects on pathological α-synuclein aggregates associated with PD. α-Synuclein was selected since its aggregates have been observed in the cytosol and nucleus (Goers et al., 2003; Gorbatyuk et al., 2008; Kontopoulos et al., 2006; Rousseaux et al., 2016). Moreover, the presence of α-synuclein aggregates in Lewy body inclusions characterizes a wide range of human brain disorders (Shults, 2006; Spillantini and Goedert, 2000). Since PCIII was able to enhance clearance of established nuclear amyloid structures (i.e., β23), we hypothesized that α-synuclein PFFs could also be modulated by PCIII. To test this possibility, HA-α-synuclein expressing SH-SY5Y cells were incubated with PFFs (5 μg/ml;
In addition to the above mentioned cellular model of α-synuclein aggregation, toxin induced cellular PD model was also generated by treating HA-α-synuclein expressing SH-SY5Y cells with 6-OHDA to induce α-synuclein aggregation and toxicity (
To investigate the mechanisms by which PCIII decreased α-synuclein aggregate formation, recombinant α-synuclein was incubated in solution in the presence or absence of PCIII. Thioflavin T fluorescence, which is sensitive to amyloid fibril formation, demonstrated that the amount of α-synuclein oligomers or fibrils increased over time and reached maximum levels at approximately 4 to 5 days of incubation (Figs. 6A and 6B). Under a treatment of 10 μM PCIII, α-synuclein aggregation was slightly inhibited. Following 50 μM PCIII treatment, α-synuclein aggregation was substantially diminished. In 50 μM PCIII-added reaction, α-synuclein oligomer or fibril aggregation was only 30% of the amount accumulated in vehicle controls (Figs. 6A and 6B). These results indicate that PCIII directly affects α-synuclein aggregation processes
Next, we sought to determine whether PCIII is capable of disintegrating already established α-synuclein PFFs of β-strand conformation. In vitro incubation of PFF with 100 μM PCIII for 7 days substantially decreased the thioflavin T fluorescence signal compared with that at the start of incubation (i.e., a 40% decrease; Fig. 6C). In contrast,
Here, we report the discovery of nuclear amyloid inhibitors that could serve as potential therapeutic agents for PD and other proteinopathies. The identification of several natural compounds was made possible by using a novel Tet-inducible nuclear β23 expression system. Previous studies have induced artificial β23 expression within the cytoplasm or nucleus to understand the molecular mechanisms of protein aggregate toxicity (Woerner et al., 2016). However, constitutive expression has limited applications for high-throughput screening. Synchronizing toxic protein expression and compound treatments in 96-well plates is difficult with constitutive expression. Moreover, timing concerns can compromise stable plating practices. In contrast, with the Tet-Off system, the timing of transgene expression is controlled by the addition of doxycycline. Indeed, this type of conditional gene expression system is commonly used to model toxic protein expression in cells and
There have been many studies on compounds that inhibit protein aggregation, including α-synuclein and amyloid β (Herva et al., 2014; Jha et al., 2016; Kim et al., 2010; 2015; Singh et al., 2013). Most of these studies performed
The nuclear β23 toxicity-inhibiting natural compounds identified in this study include PCIII, oregonin, kaempferol, and ophiocarpine. Interestingly, the chemical structure of oregonin resembles that of curcumin and its analog, which bind to α-synuclein fibrils and exert protective functions against fibril-induced toxicity (Jha et al., 2016; Singh et al., 2013). Of these compounds, PCIII was thoroughly examined due to its strong protective effects. Isothermal titration calorimetry analysis of PCIII revealed weak interaction of this compound with α-synuclein fibril (data not shown). Unfortunately, the interaction between PCIII and α-synuclein fibril seems too weak or slow to obtain thermodynamic values (over mM range of Km value) although we observed slowly increasing series of spikes of heat flow with PCIII injections. Ongoing disaggregation of fibrils into monomers in competition with compound-fibril binding might interfere with ITC measurements. More sensitive binding assays, such as surface plasmon resonance, could be applied to elucidate thermodynamic binding parameters of PCIII with diverse disease-associated amyloid aggregates. Nevertheless, relatively weak binding affinity of PCIII for α-synuclein fibril seems sufficient for disassembly of protein aggregates
We have shown that PCIII prevents β23-, mutant huntingtin-, and α-synuclein-induced proteotoxicity by facilitating clearance of protein aggregates
Mol. Cells 2019; 42(6): 480-494
Published online June 30, 2019 https://doi.org/10.14348/molcells.2019.0091
Copyright © The Korean Society for Molecular and Cellular Biology.
Sangwoo Ham1, Hyojung Kim1, Seojin Hwang2, Hyunook Kang3, Seung Pil Yun4,5,6, Sangjune Kim4,5,6 , Donghoon Kim4,5,6, Hyun Sook Kwon7
, Yun-Song Lee1
, MyoungLae Cho7, Heung-Mook Shin7, Heejung Choi3
, Ka Young Chung8, Han Seok Ko4,5,6, Gum Hwa Lee2, and Yunjong Lee1,9,*
1Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea, 2College of Pharmacy, Chosun University, Gwangju 61452, Korea, 3School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea, 4Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA, 5Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA, 6Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA, 7National Development Institute of Korean Medicine, Gyeongsan 38540, Korea, 8School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea, 9Samsung Medical Center, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Seoul 06351, Korea
Correspondence to:*ylee69@skku.edu
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Aggregates of disease-causing proteins dysregulate cellular functions, thereby causing neuronal cell loss in diverse neurodegenerative diseases. Although many
Keywords: α-synuclein, amyloid, fibril, natural compound screen, neurodegenerative disease, peucedanocoumarin III, Tet-Off model
Dysregulation of intracellular protein homeostasis is associated with diverse neurodegenerative diseases (Ross and Poirier, 2004). The aggregation of disease-related proteins (i.e., α-synuclein, polyglutamine (polyQ) repeat expansion-containing huntingtin, TAR DNA-binding protein [TDP43]) can be increased by disease-linked mutations or environmental stresses, thereby affecting the viability of neuronal subtypes in many brain disorders (Brahmachari et al., 2016; Gorbatyuk et al., 2008; Landles and Bates, 2004; Mahul-Mellier et al., 2014; Scherzinger et al., 1997; Scotter et al., 2015; Spillantini and Goedert, 2000). Interestingly, amyloid-like aggregates of disease proteins, including α-synuclein, huntingtin, and TDP43, are localized in the cytoplasm and nucleus (Goers et al., 2003; Kontopoulos et al., 2006; Rousseaux et al., 2016; Scherzinger et al., 1997; Scotter et al., 2014; Wyttenbach et al., 2000), which suggests differential proteinopathic disruption of cellular functions. A recent study showed that cytoplasmic protein aggregates impair nucleo-cytoplasmic protein transport (Woerner et al., 2016), thereby causing robust cellular toxicity. Moreover, pathological nuclear α-synuclein (Goers et al., 2003; Kontopoulos et al., 2006; Rousseaux et al., 2016) and huntingtin (Cui et al., 2006) have been reported to alter gene transcription. Since both cytosolic and nuclear protein aggregates have detrimental effects on cellular function and viability, a therapeutic strategy to control aggregates in both compartments may be important for controlling pathological processes in neurodegenerative diseases, including Parkinson’s disease (PD).
α-Synuclein is a major component of Lewy body inclusions in PD and other α-synucleinopathies (Norris et al., 2004; Spillantini and Goedert, 2000). Recently, pathologies of diseases related to α-synuclein oligomers or fibrils have been shown to propagate via neuron-to-neuron transmission (Luk et al., 2012; Mao et al., 2016). Several natural compounds have been identified and found to inhibit α-synuclein aggregation or convert toxic oligomer or fibril conformations into nontoxic species (Jha et al., 2016; Masuda et al., 2006; Singh et al., 2013). In most cases,
In this study, we established a tetracycline (Tet)-Off cell model expressing nuclear β-sheet amyloid aggregates (nuclear β23, as named in previous studies [Olzscha et al., 2011; Woerner et al., 2016]). β23 was initially developed to aid in the investigation molecular mechanisms of toxicity induced by disease-associated amyloid aggregates (Olzscha et al., 2011). β23 is an artificial protein designed to self-assemble into fibrils with repeated β strands of alternating patterns of polar and nonpolar residues (Olzscha et al., 2011). In the previous study, amyloid aggregate expression of β23 aided in the investigation of sequestration and dysregulation of functionally important endogenous proteins as molecular mechanisms of amyloid-induced cell toxicity (Olzscha et al., 2011). Using Tet-inducible expression and cellular toxicity as readouts, we identified several nuclear β23 inhibitors, including peucedanocoumarin III (PCIII). PCIII enhanced clearance of nuclear, as well as cytosolic, β23 aggregates and prevented the aggregation and toxicity of disease-related proteins (i.e., mutant huntingtin and α-synuclein). Significantly,
The National Development Institute of Korean Medicine (NIKOM) provided the natural compound library, which contained 640 natural compounds of > 80% purity (1 mg/ml). This library was used for nuclear β23 inhibitor high-throughput screening. Natural compounds blocking β23 toxicity (i.e., PCIII, kaempferol-7-O-α-L-rhamnopyranoside, oregonin, and ophiocarpine) were extracted from herbal medications, purified, and validated using high-performance liquid chromatography (HPLC). Thioflavin S, Thioflavin T, 6-OHDA, doxycycline, Alamar blue, trypan blue, MG132, and carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (Z-VAD-FMK) were purchased from Sigma (USA). Doxorubicin was purchased from Selleck Chemicals. The primary antibodies used in this study were mouse antibody to hemagglutinin (HA) (12CA5, 1:1,000; Roche, Switzerland), mouse antibody to FLAG (M2, 1:5,000; Sigma), mouse antibody to α-synuclein (1:3,000; BD Transduction Laboratories, USA), rabbit antibody to green fluorescent protein (GFP) (cat# 2956, 1:5,000; Cell Signaling Technology, USA) mouse antibody to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (GT239, 1:5,000; GeneTex, USA), mouse antibody to poly (ADP-ribose) polymerase 1 (PARP1) (cat# 556494, 1:1,000; BD Bioscience, USA), conformation specific rabbit antibody to α-synuclein filaments (MJFR-14-6-4-2, cat# ab209538, 1:5,000; Abcam, USA) and horseradish peroxidase (HRP)-conjugated mouse antibody to β-actin (AC15; Sigma-Aldrich, USA). The secondary antibodies used were HRP-conjugated sheep antibody to mouse immunoglobulin G (IgG) (cat# RPN4301, 1:5,000; GE Healthcare, USA), HRP-conjugated donkey antibody to rabbit IgG (cat# RPN4101, 1:5,000; GE Healthcare), Alexa Fluor 488-conjugated donkey antibody to mouse IgG (H + L) (cat# A21202, 1:1,000; Invitrogen, USA), Alexa Fluor 568-conjugated donkey antibody to mouse IgG (cat# A10037, 1:1,000; Invitrogen), and Alexa Fluor 647-conjugated donkey antibody to mouse IgG (cat# A31571, 1:1,000; Invitrogen).
The double-strand oligos encoding nuclear β23, β23, and nuclear αS824 sequence were cloned into a pTRE-Dual2 plasmid (Clontech Laboratories, USA). The full sequence of nuclear β23 with tags (NLS-FLAG-β23-HA) is as follows:
ATGCCAAAGAAGAAGCGGAAGGTCGGTTGCGACTACAAGGACGACGACGACAAGGGCATGCAGATCTCCATGGACTACAACATCCAGTTCCACAACAACGGCAACGAGATCCAGTTCGAGATCGACGACTCCGGCGGCGACATCGAGATCGAGATCCGGGGCCCCGGCGGCCGGGTGCACATCCAGCTGAACGACGGCCACGGCCACATCAAGGTGGACTTCAACAACGACGGCGGCGAGCTGCAGATCGACATGCACTACCCATACGACGTCCCAGACTACGCT.
The full DNA and amino acid sequence of β23 with tags (FLAG-β23-HA) is as follows:
ATGTGCGACTACAAGGACGACGACGACAAGGGCATGCAGATCTCCATGGACTACAACATCCAGTTCCACAACAACGGCAACGAGATCCAGTTCGAGATCGACGACTCCGGCGGCGACATCGAGATCGAGATCCGGGGCCCCGGCGGCCGGGTGCACATCCAGCTGAACGACGGCCACGGCCACATCAAGGTGGACTTCAACAACGACGGCGGCGAGCTGCAGATCGACATGCACTACCCATACGACGTCCCAGACTACGCTTAA;
MCDYKDDDDKGMQISMDYNIQFHNNGNEIQFEIDDSGGDIEIEIRGPGGRVHIQLNDGHGHIKVDFNNDGGELQIDMHYPYDVPDYA.
The full DNA and amino acid sequence of nuclear αS824 with tags (NLS-FLAG-αS824 -HA) is as follows:
ATGCCAAAGAAGAAGCGGAAGGTCGGTTGCGACTACAAGGACGACGACGACAAGGGCATGTACGGCAAGCTGAACGACCTGCTGGAGGACCTGCAGGAGGTGCTGAAGCACGTGAACCAGCACTGGCAGGGCGGCCAGAAGAACATGAACAAGGTGGACCACCACCTGCAGAACGTGATCGAGGACATCCACGACTTCATGCAGGGCGGCGGCTCCGGCGGCAAGCTGCAGGAGATGATGAAGGAGTTCCAGCAGGTGCTGGACGAGATCAAGCAGCAGCTGCAGGGCGGCGACAACTCCCTGCACAACGTGCACGAGAACATCAAGGAGATCTTCCACCACCTGGAGGAGCTGGTGCACCGGTACCCATACGACGTCCCAGACTACGCTTGA;
MPKKKRKVGCDYKDDDDKGMYGKLNDLLEDLQEVLKHVNQHWQGGQKNMNKVDHHLQNVIEDIHDFMQGGGSGGKLQEMMKEFQQVLDEIKQQLQGGDNSLHNVHENIKEIFHHLEELVHRYPYDVPDYA.
Construct integrity was verified by sequencing. Plasmid cytomegalovirus (pCMV)-tetracycline transactivator (tTA)was purchased from Clontech and the pTreTight-Htt94Q-CFP (Maynard et al., 2009) construct was purchased from Addgene in USA (Plasmid #23966). The HA-α-synuclein construct was generated as previously described (Brahmachari et al., 2016).
In March 2013,
HPLC analysis was performed on an Agilent 1260 system, equipped with an evaporative light-scattering detector (ELSD) and a Kinetex C18 column (4.6 × 150 mm; Phenomenex, USA). The mobile phase consisted of 0.1% trifluoroacetic acid in water (v/v) (A) and acetonitrile (B). The injection volume was 3 μl and flow rate was kept at 0.5 ml/min for a total run time of 30 min. The gradient program was as follows: 2% (start, B), 5% (0–3 min, B), 100% (3–20 min, B), 2% (20–23 min, B), and kept at 2% (B) until the end of the run at 30 min. HPLC analysis found PCIII purity to be 99.9% (Fig. 1).
1H and 13C NMR spectra were recorded on a Jeol ECA-500 MHz NMR instrument, operating at 500 MHz for 1H NMR and 125 MHz for 13C NMR (JEOL, Japan) with tetramethylsilane as the internal standard. The PCIII structure observed was compared with that reported in previous literature (Chen et al., 1996; Takata et al., 1990).
PCIII: colorless needles; ESI-MS
Human neuroblastoma SH-SY5Y cells (ATCC [American Type Culture Collection], USA) or HEK-293T cells were grown in DMEM containing 10% fetal bovine serum (FBS) (vol/vol) and antibiotics in a humidified 5% CO2 atmosphere at 37°C. X-tremeGENE HP transfection reagent (Roche) was used for transient transfections according to the manufacturer’s instructions. For α-synuclein PFF uptake experiments, SH-SY5Y cells were incubated for 4 days in complete media containing PFF (5 μg/ml). PFF was replaced every 2 days. SH-SY5Y cells were briefly washed with phosphate-buffered saline (PBS) and incubated in the presence of natural compounds for an indicated duration followed by western blot analysis of remnant PFF in cell lysates.
SH-SY5Y cells were fractionated into cytosol and nucleus fractions using the Qproteome Mitochondria Isolation Kit (Qiagen, Germany) according to manual instructions. Cytosolic fractions were further concentrated with acetone precipitation. The purity of each fraction was validated with western blots using antibodies to marker proteins for cytosolic (GAPDH, 2% of total cytosolic fraction) and nuclear (PARP1, 5% of total nuclear fraction) fractions.
As outlined in Figure 2C, HEK-293T cells were cotransfected with pCMV-tTA and TetP-nuclear β23 constructs. Cells were kept on doxycycline (200 ng/ml; Sigma)-containing complete media to prevent toxic expression of nuclear β23. After 2 days, transfected HEK-293T cells were plated onto 96-well white flat-bottom plates (Microtiter; Thermo Scientific, USA) at 80% confluency in 100 μl of DMEM containing 10% FBS with penicillin/streptomycin (P/S) and without doxycycline for nuclear β23 induction. After 24 h, each compound (i.e., 640 natural compounds extracted and purified by NIKOM; 0.35 μg/ml each) was added to a well. For homogeneous treatment of each compound, 50 μl of culture media were removed and 50 μl of 2X compound in complete media were then added to each well. After 48 h incubation, cell viability was measured using Alamar blue assay. Briefly, 10 μl of filtered Alamar blue solution (1 mg/ml resazurin in DW filtered through a 0.22 μM pore filter) was added to the 100 μl culture media in 96-well plates. Cell viability was determined by fluorescence reading using a microplate fluorescence spectrometer (excitation: 530 nm, emission: 590 nm; SYNERGYneo microplate reader [BioTek, USA]). Each plate had four wells of the blank with no cell culture, positive controls (dimethyl sulfoxide [DMSO] vehicle treatment with β23 induction) and negative controls (no induction of β23 by maintaining in doxycycline [200 ng/ml]-containing media). To assess the appropriateness of our assay for high-throughput screening, the Z′ factor was quantified as follows:
Z′ factor = 1 − 3(SDp + SDn) / (MEANp − MEANn), where MEANp and MEANn are the means of the positive and negative controls, respectively, and SDp and SDn are the standard deviations of the positive and negative controls, respectively. A high-throughput screening-ready assay should have a Z′ factor between 0.5 and 1.
For Triton X-100-soluble and -insoluble fraction separation, SH-SY5Y cells were harvested and processed into nonionic detergent-soluble and detergent-insoluble fractions in lysis buffer containing PBS, 1% Triton X-100, Phosphatase Inhibitor Cocktail II and III (Sigma-Aldrich), and a complete protease inhibitor mixture. The lysates were centrifuged at 100,000
SH-SY5Y or HEK-293T cells were plated onto poly-D-lysine-coated coverslips at 10,000 cells/cm2. Following experimental procedures, cells were fixed with 4% paraformaldehyde in PBS and blocked in a solution containing 5% normal donkey serum (Jackson ImmunoResearch, USA), 2% BSA (Sigma), and 0.1% Triton X-100 (Sigma) for 1 h at room temperature. Samples were then incubated overnight with corresponding primary antibodies against proteins of interest at 4°C. Briefly, cells grown on coverslips were washed with PBS containing 0.1% Triton X-100 and incubated for 1 h with fluorescence-conjugated secondary antibodies (1:500; Invitrogen) at room temperature. The coverslips were mounted with 4′, 6-diamidino-2-phenylindole (DAPI). Fluorescent images were obtained using a fluorescence microscope (Axiovert, 200 M; Carl Zeiss, Germany) or confocal microscope (TCS SP8 STED; Leica Microsystems, Germany) for four channel fluorescence image acquisition. For thioflavin S staining, thioflavin S (500 mM) was dissolved in 50% ethanol. Fixed SH-SY5Y cells were then stained with thioflavin S for 7 min. As a differentiation step to remove nonspecific binding of the dye, a slide was soaked in 100%, 95%, and 90% ethanol solutions for 10 s each and then transferred to PBS. Subsequent blocking and primary antibody binding steps were followed as described above for immunofluorescence staining.
Total RNA was extracted with QIAzol Lysis Reagent (cat# 79306; Qiagen) and then treated with DNase I to eliminate trace DNA contamination. cDNA was synthesized from total RNA (1.5 μg) using a first-strand cDNA synthesis kit (iScript cDNA synthesis kit; Bio-Rad, USA). The relative quantities of β23 mRNA expression were analyzed using real-time PCR (QuantStudio 6 flex Real-Time PCR System; Applied Biosystems, USA). SYBR Green PCR master mix (cat# 4309155; Applied Biosystems) was used according to the manufacturer’s instructions. The relative mRNA expression levels of β23 were calculated by the ∆∆Ct method (Livak and Schmittgen, 2001) using GAPDH as an internal loading control. The primer sequences for real-time gene amplification are as follows:
SH-SY5Y cells were plated in 6-well plates at a density of 0.5 × 106 cells per well. Following transient transfection with indicated constructs, cells were grown in DMEM containing low serum (2.5% FBS) for indicated days of α-synuclein, Tet-regulated β23, or mutant huntingtin (Htt94Q-CFP) expression. For α-synuclein toxicity, SH-SY5Y cells were treated with 6-OHDA or PBS vehicle control. SH-SY5Y cells were trypsinized to yield single-cell suspensions that were washed twice with PBS before resuspension in serum-free DMEM. Resuspended cells were mixed with an equal volume of 0.4% trypan blue (wt/vol) and incubated for 2 min at room temperature. Live and dead cells were analyzed automatically using the Countess II Automated Cell Counter (Life Technologies, USA). Cell Counting Kit-8 (CCK8) (Dojindo Molecular Technologies, USA) was also used to assess viability of HEK-293T cells by following the manufacturer’s instructions.
Plasmid pRK172-human α-synuclein was transformed to BL21-competent
To investigate the disaggregating effects of PCIII against α-synuclein PFFs, α-synuclein fibrils (50 μM) prepared from the 7 days incubation of monomeric α-synuclein were subjected to
Recombinant human α-synuclein from
Quantitative data are presented as the mean ± standard error of the mean (SEM). Statistical significance was assessed with either an unpaired two-tailed Student’s
To establish a cellular model of nuclear amyloid accumulation, we generated a Tet-Off construct capable of expressing artificial strands of β-sheet structure with an N-terminal nuclear localization sequence (NLS), FLAG tag and C-terminal influenza HA tag (Fig. 1A). In this manuscript, this construct is subsequently called nuclear β23, following previous studies in which β23’s amyloid-like structure has been characterized (Olzscha et al., 2011; Woerner et al., 2016). Since β23 and reporter protein mCherry expression is under the control of a tetracycline-responsive promoter (TetP-β23), coexpression of a tTA is required for the activation of the Tet promoter and subsequent transcription of mCherry-internal ribosome entry site (IRES)-β23 (Fig. 1B). As designed, cotransfection with pCMV-tTA and TetP-nuclear β23 resulted in the expression of nuclear β23 and fluorescent reporter mCherry in HEK-293T cells (Fig. 1C,
Following this, we assessed whether nuclear expression of amyloid β23 is toxic to HEK-293T cells. HEK-293T cells were transfected with pCMV-tTA and TetP-β23 constructs and incubated in complete media containing either doxycycline or vehicle as a control. In nuclear β23-expressing cells cultured for 2 days in the absence of doxycycline, cell viability measured by Alamar blue assay decreased to about 50% (Fig. 2A). Since the cell viability assay provided consistent and robust toxicity results, we used the Tet-Off nuclear β23 expression system to screen natural compounds that could potentially inhibit nuclear aggregate toxicity. For timed expression of β23, HEK-293T cells were transfected with pCMV-tTA and TetP-nuclear β23 and plated onto 96-well plates in the presence of doxycycline to suppress nuclear β23 expression (Figs. 2B and 2C). After plating, cells were treated with 640 natural compounds extracted and purified from clinically prescribed herbal medications (0.35 μg/ml). On the following day, nuclear β23 expression was induced by removing doxycycline. After 2 days of β23 induction, high-throughput cell viability was assessed by reading Alamar blue fluorescence (Fig. 2D). Z′ factors calculated for each plate using Alamar blue fluorescence intensities for negative (doxycycline treatment) and positive (no doxycycline + DMSO vehicle) controls were between 0.5 and 1.0 (
To determine whether cell death inhibition by these novel compounds is rather selective against toxic protein aggregation, an additional toxic insult (doxorubicin) was introduced into HEK-293T cells; after which, the cytoprotective effect of β23 inhibitors was examined (
To assess the potential central nervous system applications for inhibitors of nuclear protein aggregation toxicity, we used the SH-SY5Y neuroblastoma cell line to examine the cytoprotective functions of select compounds. Transient transfection with pCMV-tTA and TetP-nuclear β23 in SH-SY5Y cells led to the expression of nuclear amyloid β23 and reporter protein mCherry (Fig. 3A). Nuclear β23 expression for 24 h in SH-SY5Y cells resulted in approximately 80% cell toxicity (Fig. 3B). All compounds (PCIII, kaempferol, oregonin, and ophiocarpine) increased SH-SY5Y cell viability against nuclear β23 induction, with PCIII providing maximal protection (up to 60% cell viability; Fig. 3B). Following this, we investigated whether these compounds improved cell viability against nuclear β23 toxicity through the alteration of protein aggregate clearance. To monitor β23 clearance over time, SH-SY5Y cells were transfected with pCMV-tTA and TetP-nuclear β23 (Fig. 3B; top panel). Twenty-four hours after transfection, doxycycline was added to stop further β23 and mCherry transcription in SH-SY5Y cells, and β23 stability was assessed by immunofluorescence using anti-FLAG antibodies. Immunofluorescence showed that PCIII treatment markedly increased the removal of FLAG-tagged β23 from the nucleus, whereas β23 still remained in the vehicle treatment control for 37 h following the addition of doxycycline (Figs. 3C and 3D). To a lesser extent, oregonin facilitated the removal of nuclear FLAG-tagged β23 (Figs. 3C and 3D). In contrast, despite their cytoprotective ability, kaempferol and ophiocarpine had no effect on β23 degradation. These compounds had no effect on reporter protein expression and stability as shown by mCherry fluorescence intensity (Fig. 3C). Moreover, transcription of β23 mRNA was not altered by β23 inhibitors (Fig. 3E).
Regulation of the stability of nuclear β23 by PCIII was further determined by quantitative western blot. Immunoblot results showed no difference in β23 levels over time, which suggests that nuclear amyloid β23 was highly stable and resistant to degradation (Figs. 3F and 3G). Consistent with the immunofluorescence results, PCIII treatment led to approximately 70% degradation of β23 (Figs. 3F and 3G). Taken together, these results show that PCIII’s inhibition of nuclear β23-induced toxicity correlates with its ability to accelerate the degradation of nuclear amyloid β23, which is otherwise highly stable and cannot be cleared via an intrinsic degradation machinery.
Afterwards, the mechanisms of PCIII-mediated nuclear β23 clearance in SH-SY5Y cells were elucidated. Tet-Off expression construct of β23 with no NLS sequence (TetP-β23) was generated to assess whether PCIII’s cytoprotective effect on amyloid toxicity is specific to nuclear aggregates. SH-SY5Y cells were co-transfected with pCMV-tTA and TetP-β23 constructs. β23 was expressed for the initial 24 h and further expression was terminated by doxycycline treatment. Cell viability was assessed 24 h after doxycycline treatment. Expression of β23 amyloid in SH-SY5Y cells resulted in robust cell death which was largely prevented by PCIII treatment (
Amyloid selectivity of PCIII-mediated protein degradation was examined by generating an artificial protein containing an alpha helix structure (αS824), following previous reports (Olzscha et al., 2011; Woerner et al., 2016). Tet-Off construct of αS824 was cloned with NLS and FLAG sequence in the N terminus and HA tag in the C terminus (TetP-nuclear αS824). In contrast to aggregate-forming β23, αS824 was distributed to the soluble fraction (
Next, amyloid clearance facilitated by PCIII was investigated. The proteasome inhibitor, MG132, was treated for 24 h during clearance of nuclear β23 or αS824 in the presence or absence of PCIII (
Of the compounds that were cytoprotective against nuclear and cytosolic aggregates, PCIII demonstrated the greatest protective and disaggregative effects. PCIII extracted and purified from
To determine PCIII’s therapeutic effects on disease-associated proteins, we selected a cellular model of mutant huntingtin aggregation, which is linked to hyperkinetic movement disorders in humans, such as Huntington’s disease (Scherzinger et al., 1997). To examine whether PCIII can regulate nuclear huntingtin aggregation in cells, we transfected SH-SY5Y cells with pCMV-tTA and 94 polyglutamine repeat-containing huntingtin protein (TetP-Htt94Q-CFP) to induce aggregate formation (Fig. 4A). A proteasome inhibitor (MG132) was added to facilitate nuclear aggregate formation as previously described (Maynard et al., 2009; Wyttenbach et al., 2000). Separation of protein lysates into detergent (1% Triton X-100)-soluble and -insoluble fractions demonstrated that PCIII treatment markedly prevented insolubility of mutant huntingtin aggregates and redistributed huntingtin into soluble fractions (Figs. 4B and 4C). Consistent with western blot results, immunofluorescence demonstrated that PCIII treatment of SH-SY5Y cells was associated with substantial removal of cytosolic and nuclear aggregate puncta formed because of Htt94Q expression (Figs. 4D–F). Moreover, the growth of mutant huntingtin aggregate size in DMSO-treated controls was inhibited or diminished by PCIII treatment in the nucleus and cytoplasm, respectively (Figs. 4E and 4F). Mutant huntingtin aggregate expression led to 80% cell death in SH-SY5Y cells; however, PCIII treatment improved cell viability by approximately 20% (Fig. 4G).
After observing the promising effects of PCIII on amyloid β23 and huntingtin aggregate toxicity, we tested PCIII’s protective effects on pathological α-synuclein aggregates associated with PD. α-Synuclein was selected since its aggregates have been observed in the cytosol and nucleus (Goers et al., 2003; Gorbatyuk et al., 2008; Kontopoulos et al., 2006; Rousseaux et al., 2016). Moreover, the presence of α-synuclein aggregates in Lewy body inclusions characterizes a wide range of human brain disorders (Shults, 2006; Spillantini and Goedert, 2000). Since PCIII was able to enhance clearance of established nuclear amyloid structures (i.e., β23), we hypothesized that α-synuclein PFFs could also be modulated by PCIII. To test this possibility, HA-α-synuclein expressing SH-SY5Y cells were incubated with PFFs (5 μg/ml;
In addition to the above mentioned cellular model of α-synuclein aggregation, toxin induced cellular PD model was also generated by treating HA-α-synuclein expressing SH-SY5Y cells with 6-OHDA to induce α-synuclein aggregation and toxicity (
To investigate the mechanisms by which PCIII decreased α-synuclein aggregate formation, recombinant α-synuclein was incubated in solution in the presence or absence of PCIII. Thioflavin T fluorescence, which is sensitive to amyloid fibril formation, demonstrated that the amount of α-synuclein oligomers or fibrils increased over time and reached maximum levels at approximately 4 to 5 days of incubation (Figs. 6A and 6B). Under a treatment of 10 μM PCIII, α-synuclein aggregation was slightly inhibited. Following 50 μM PCIII treatment, α-synuclein aggregation was substantially diminished. In 50 μM PCIII-added reaction, α-synuclein oligomer or fibril aggregation was only 30% of the amount accumulated in vehicle controls (Figs. 6A and 6B). These results indicate that PCIII directly affects α-synuclein aggregation processes
Next, we sought to determine whether PCIII is capable of disintegrating already established α-synuclein PFFs of β-strand conformation. In vitro incubation of PFF with 100 μM PCIII for 7 days substantially decreased the thioflavin T fluorescence signal compared with that at the start of incubation (i.e., a 40% decrease; Fig. 6C). In contrast,
Here, we report the discovery of nuclear amyloid inhibitors that could serve as potential therapeutic agents for PD and other proteinopathies. The identification of several natural compounds was made possible by using a novel Tet-inducible nuclear β23 expression system. Previous studies have induced artificial β23 expression within the cytoplasm or nucleus to understand the molecular mechanisms of protein aggregate toxicity (Woerner et al., 2016). However, constitutive expression has limited applications for high-throughput screening. Synchronizing toxic protein expression and compound treatments in 96-well plates is difficult with constitutive expression. Moreover, timing concerns can compromise stable plating practices. In contrast, with the Tet-Off system, the timing of transgene expression is controlled by the addition of doxycycline. Indeed, this type of conditional gene expression system is commonly used to model toxic protein expression in cells and
There have been many studies on compounds that inhibit protein aggregation, including α-synuclein and amyloid β (Herva et al., 2014; Jha et al., 2016; Kim et al., 2010; 2015; Singh et al., 2013). Most of these studies performed
The nuclear β23 toxicity-inhibiting natural compounds identified in this study include PCIII, oregonin, kaempferol, and ophiocarpine. Interestingly, the chemical structure of oregonin resembles that of curcumin and its analog, which bind to α-synuclein fibrils and exert protective functions against fibril-induced toxicity (Jha et al., 2016; Singh et al., 2013). Of these compounds, PCIII was thoroughly examined due to its strong protective effects. Isothermal titration calorimetry analysis of PCIII revealed weak interaction of this compound with α-synuclein fibril (data not shown). Unfortunately, the interaction between PCIII and α-synuclein fibril seems too weak or slow to obtain thermodynamic values (over mM range of Km value) although we observed slowly increasing series of spikes of heat flow with PCIII injections. Ongoing disaggregation of fibrils into monomers in competition with compound-fibril binding might interfere with ITC measurements. More sensitive binding assays, such as surface plasmon resonance, could be applied to elucidate thermodynamic binding parameters of PCIII with diverse disease-associated amyloid aggregates. Nevertheless, relatively weak binding affinity of PCIII for α-synuclein fibril seems sufficient for disassembly of protein aggregates
We have shown that PCIII prevents β23-, mutant huntingtin-, and α-synuclein-induced proteotoxicity by facilitating clearance of protein aggregates
Soyeon Lee, and Kyung-Tai Min
Mol. Cells 2018; 41(12): 1000-1007 https://doi.org/10.14348/molcells.2018.0438Jinyoung Kim, Yu-Mi Lim, and Myung-Shik Lee
Mol. Cells 2018; 41(1): 11-17 https://doi.org/10.14348/molcells.2018.2228