You are on page 1of 15

Biochimica et Biophysica Acta 1865 (2017) 589–603

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbapap

Polyphenols in combination with β-cyclodextrin can inhibit and


disaggregate α-synuclein amyloids under cell mimicking conditions: A
promising therapeutic alternative

Saurabh Gautam a, Sandip Karmakar a, Radhika Batra a, Pankaj Sharma b, Prashant Pradhan b, Jasdeep Singh b,
Bishwajit Kundu b,⁎, Pramit K. Chowdhury a,⁎
a
Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
b
Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India

a r t i c l e i n f o a b s t r a c t

Article history: Parkinson's disease is characterized by the presence of insoluble and neurotoxic aggregates (amyloid fibrils) of an
Received 25 July 2016 intrinsically disordered protein α-synuclein. In this study we have examined the effects of four naturally occur-
Received in revised form 17 February 2017 ring polyphenols in combination with β-cyclodextrin (β-CD) on the aggregation of α-synuclein in the presence
Accepted 22 February 2017
of macromolecular crowding agents. Our results reveal that even at sub-stoichiometric concentrations of the in-
Available online 24 February 2017
dividual components, the polyphenol–β-CD combination(s) not only inhibited the aggregation of the proteins
Keywords:
but was also effective in disaggregating preformed fibrils. Curcumin was found to be the most efficient, followed
α-Synuclein by baicalein with (−)-epigallocatechin gallate and resveratrol coming in next, the latter two exhibiting very sim-
β-cyclodextrin ilar effects. Our results suggest that the efficiency of curcumin results from a balanced composition of the phenolic
Polyphenols \\OH groups, benzene rings and flexibility. The latter ensures proper positioning of the functional groups to max-
Amyloid inhibition and disaggregation imize the underlying interactions with both the monomeric form of α-synuclein and its aggregates. The unique-
Parkinson's disease ness of β-CD was reinforced by the observation that none of the other cyclodextrin variants [α-CD and HP-β-CD]
used was as effective, in spite of these possessing better water solubility. Moreover, the fact that the combinations
remained effective under conditions of macromolecular crowding suggests that these have the potential to be de-
veloped into viable drug compositions in the near future. MTT assays on cell viability independently confirmed
this hypothesis wherein these combinations (and the polyphenols alone too) appreciably impeded the toxicity
of the prefibrillar α-synuclein aggregates on the mouse neuroblastoma cell lines (N2a cells).
© 2017 Elsevier B.V. All rights reserved.

1. Introduction characterized by the presence of macroscopic amyloid deposits of the


protein α-synuclein known as Lewy neuritis and Lewy bodies in brain
A number of human pathological diseases such as Alzheimer's dis- tissues [2].
ease, type 2 diabetes, Parkinson's disease, Amyotrophic lateral sclerosis α-Synuclein is copiously present in human brain tissues and also in
(ALS), the prion diseases, and Huntington disease arise from the un- some other tissues such as red blood cells [2]. It is an intrinsically disor-
wanted misfolding, oligomerization and aggregation of proteins [1]. dered protein, composed of 140 amino acid residues that can be struc-
Most of these diseases are characterized by the deposition of amyloid fi- turally divided into three distinct segments (Fig. S1, Supporting
brils in body tissues. Among them Parkinson's disease is one of the most Information) namely: (a) N-terminal amphipathic segment (1–60
common diseases with 7–10 million people worldwide suffering from amino acid residues), (b) a hydrophobic central region (61–95 residues)
it. Parkinson's disease and dementia (due to synucleinopathies) are and (c) C-terminal acidic region (96–140 residues) [3]. The N-terminal
region shares homology with lipoproteins (with amphipathic α-heli-
Abbreviations: β-CD, β-cyclodextrin; CR, congo red; EGCG, (−)-epigallocatechin ces) and contains a characteristic consensus hexameric sequence
gallate; HP-β-CD, (2-Hydroxypropyl)-β-cyclodextrin; IPTG, isopropyl β-D- (KTKEGV) which is repeated about four times. The central hydrophobic
thiogalactopyranoside; SEC, size exclusion chromatography; TEM, transmission electron region is known to be responsible for the aggregation of α-synuclein,
microscopy; ThT, Thioflavin T.
⁎ Corresponding authors.
without which the protein loses its propensity to form amyloids. The
E-mail addresses: bkundu@bioschool.iitd.ac.in (B. Kundu), carboxy terminus region is highly acidic in nature and has been found
pramitc@chemistry.iitd.ac.in (P.K. Chowdhury). to interfere in the formation of aggregates due to its role as an

http://dx.doi.org/10.1016/j.bbapap.2017.02.014
1570-9639/© 2017 Elsevier B.V. All rights reserved.
590 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

intramolecular chaperone [4]. Three tyrosine residues, which are highly synthetic and protein based crowders in an attempt to mimic the phys-
conserved, are also present in this C-terminal region. In vivo, higher ex- iological milieu [40,41]. In the current study, we have tried to extend the
pression level of α-synuclein, increases its tendency to aggregate [5]. use of the combination of polyphenols and β-CD to such cell mimicking
The actual function of this protein is ambiguous with its role in various conditions. Based on our observations, we have also attempted to pro-
cellular activities having been reported, that includes interaction with vide molecular insights into the underlying mechanism and role of
cell membranes/lipids, release of neurotransmitters and maintenance, \\OH groups present in these polyphenols with regards to their ability
storage and transport of vesicles across cells [2,6]. In presence of alco- to modulate protein aggregation. We have used four different naturally
hols, α-synuclein is known to form ordered structures such as α-helix occurring polyphenols namely, resveratrol (from red grapes), baicalein
and β-sheet with low concentrations of ethyl alcohol enhancing its ag- (from the Chinese herbal medicinal plant Scutellaria baicalensis) and
gregation [7]. Soluble oligomers apart from fibrils of α-synuclein have (−)-epigallocatechin gallate (EGCG) (from green tea) along with
also been implicated in neurotoxicity during Parkinson's disease [2]. curcumin which is obtained from turmeric (C. longa) (Fig. 1). The mac-
Numerous small molecule inhibitors (e.g. flavonoids, dopamine, car- romolecular crowding agents used in this study are: Dextran 6, Dextran
bohydrates, antibiotics, synthetic chemical compounds, natural plant 40 and Ficoll 70 with an average molecular weight of 6 kDa, 40 kDa and
products etc.) of amyloid formation by α-synuclein have been reported 70 kDa, respectively. Our results reveal that these polyphenols in an
in literature [8,9]. Polyphenols also constitute an important class of mol- optimized combination with β-CD not only precluded formation of
ecules that have been shown to inhibit and in some cases even disaggre- aggregates but also disaggregated amyloids formed by α-synuclein
gate amyloids formed by α-synuclein [10–20] and other proteins [21, with varying efficiencies in the presence of the macromolecular
22]. Moreover, polyphenols are known to have anti-cancer, anti-oxida- crowding agents. Moreover, the pronounced toxicities displayed by
tive and anti-microbial properties thereby providing several additional the α-synuclein aggregates were significantly diminished when the
medicinal benefits. They are also reported to prevent cardiovascular dis- cells were treated with these polyphenol–β-CD combinations.
eases and control cell death [23]. Recent studies on the mechanism of Taken together our data suggest that these synergistic combinations
inhibition and disaggregation of α-synuclein amyloids by polyphenolic may facilitate the long awaited expedition of such polyphenols to
compounds have shown that the presence of two or more ‘vicinal drug molecules, though views against the same have been put for-
polyhydroxyphenyl groups’ is more important rather than the total ward recently [42].
number of hydroxyl groups [10,13]. Molecular dynamics studies have
also pointed towards the role of these \\OH groups in inhibition and
disaggregation of amyloids [21,24]. However, curcumin, a naturally oc- 2. Materials and methods
curring polyphenol derived from turmeric (Curcuma longa), is a unique
molecule with only monohydroxyphenyl rings but still being a highly 2.1. Materials
potent aggregation inhibitor [25–28]. It has been hypothesized that
curcumin binds to the aliphatic residues present in the middle hydro- Curcumin, resveratrol, baicalein, EGCG, thioflavin T (ThT) and congo
phobic stretch of native monomeric α-synuclein [29]. After this initial red (CR) were purchased from Sigma-Aldrich (St. Louis, MO, USA) and
interaction, the aromatic rings of curcumin bind to nearby hydrophobic used as received. Ethanol was procured from Merck (Mumbai, India).
residues of α-synuclein. The same study proposed that curcumin Ampicillin, LB medium for bacterial growth and isopropyl β-D-thio-
strongly prevents the formation of oligomers and fibrils of α-synuclein galactopyranoside (IPTG) were purchased from Himedia chemicals
by accelerating the intramolecular diffusion of the protein. This leads to (Mumbai, India). All other chemicals used were of highest purity avail-
an increase in the reconfiguration rate of α-synuclein thereby limiting able/analytical grade.
the exposure time of the hydrophobic patches. Another study has
shown that curcumin reduces the toxicity of α-synuclein by binding
to it and minimizing the availability of hydrophobic surface [11]. 2.2. Expression, isolation and purification of human α-synuclein
A recent study from our group has demonstrated that curcumin in
combination with β-cyclodextrin (β-CD) not only inhibited but also dis- Human α-synuclein was expressed, isolated and purified with a sim-
aggregated α-synuclein amyloids in vitro [30]. We hypothesized that ilar process as described earlier [30,43]. Plasmid pT7-7 containing the
curcumin, with its hydrophobic region and \\OH groups, interacts gene for human α-synuclein was first transformed into E. coli BL21
with the hydrophobic middle region and hydrophilic residues of α-sy- (DE3) cells using calcium chloride. Then a single colony was picked
nuclein, respectively. β-CD then further helps by binding to aromatic and inoculated into 100 mL LB medium enriched with 100 mg/mL am-
residues such as phenylalanine present in α-synuclein. β-CD is a cyclic picillin. The culture was incubated at 37 °C and the absorbance at
polysaccharide with seven glucose units. It is a bucket shaped molecule 600 nm was recorded at regular intervals. Induction was carried out
with a hydrophobic internal cavity and hydrophilic exterior [31]. β-CD by adding IPTG (1 mM final concentrations) when absorbance reached
itself is a biologically active molecule and has been used in various ap- 0.7 to 1.0. The cells were harvested and resuspended in 0.75 mL of buffer
plications such as protein folding as a pseudochaperone [32], catalysis (50 mM Tris-HCl, pH 7.5, 10 mM EDTA and 150 mM NaCl) and frozen at
[33], drug delivery [34], lowering of cholesterol [35] and inhibiting pro- −80 °C. Tubes with frozen cells were placed in a boiling water bath for
tein aggregation [36], to name a few. β-CD interacts with polyphenols 7 min and the supernatant was then collected after centrifugation at
and helps in overcoming their limitations such as low bioavailability, in- maximum speed for 5 min. Streptomycin sulfate (136 μL/mL of 10% so-
solubility, instability [37] and also increase their activity, efficiency [30], lution per mL of supernatant) and glacial acetic acid (228 μL/mL of su-
anti-inflammatory and anti-proliferative properties [38]. pernatant) were added and centrifuged for 2 min. Again the
Polyphenolic compounds are not always effective in vivo due to var- supernatant was recovered and precipitated with ammonium sulfate
ious reasons [39]. One of the major factors that add to the intracellular (saturated ammonium sulfate at 4 °C was used 1:1, v/v, with superna-
complexity is the congested cellular interior. The intracellular environ- tant). The protein was collected as a precipitate by centrifugation and
ment is highly crowded with the concentration of macromolecules washed once with 1 mL of ammonium sulfate solution (4 °C, 1:1, v/v,
ranging from 50 g/L to as high as 400 g/L [40]. Such an environment saturated ammonium sulfate and water). The washed pellet was resus-
has been shown to affect protein folding, structure, function, aggrega- pended in 900 μL of 100 mM ammonium acetate (to form a cloudy solu-
tion and other thermodynamic properties primarily via the excluded tion) and precipitated by adding an equal volume of ethanol at room
volume effect [41]. Hence, the experiments carried out in dilute buffer temperature. Precipitation with ethanol was repeated once more. The
solutions may or may not always provide a reliable picture of the in pellet was resuspended in 100 mM ammonium acetate and extensively
vivo environment. To address this issue, researchers have used both dialyzed against 10 mM Tris-HCl buffer, pH 7.4.
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 591

Fig. 1. Chemical structures of the four different polyphenols [curcumin, resveratrol, baicalein and (−)-epigallocatechin gallate (EGCG)], β-cyclodextrin (β-CD) and two types of
macromolecular crowding agents (Dextran and Ficoll) used in this study. The structures were drawn using ChemDraw.

2.3. Preparation of solutions of polyphenols 10 mM Tris-HCl buffer, pH 7.4) resulting in a total volume of 500 μL
with the final concentration of ThT/CR being 20 μM [The aliquots were
Stock solutions of all four polyphenols (curcumin, resveratrol, used directly without ThT/CR for recording circular dichroism spectra,
baicalein and EGCG) were prepared in ethanol. All the compounds size exclusion chromatography (SEC) and transmission electron micros-
were added to protein solutions so that the final concentration of etha- copy (TEM) experiments]. This resulted in a dilution of the aggregating/
nol remained equal to or below 7.5% (v/v) which was required for pro- disaggregating reaction mixture containing the protein and
tein aggregation. polyphenol–β-CD combination. The dilution of the aggregating/disag-
gregating reaction mixture was 8.33 times (60 μL reaction mixture
+ 440 μL ThT in buffer = 500 μL / 60 μL = 8.33) thereby resulting in di-
2.4. Assays to monitor amyloid fibrils (fibril formation assay) lution of polyphenols as well by a factor of 8.33 present in the original
aggregating/disaggregating reaction mixture. ThT fluorescence acquisi-
Amyloid fibrils formed by α-synuclein were monitored by using two tions were then performed by exciting the samples at 430 nm with an
most common techniques namely: ThT based fluorimetric assay [44] emission at 485 nm using an Edinburgh FLS920 spectrofluorimeter (Ed-
and congo red (CR) assay [45]. The protein solution with a concentra- inburgh Instruments Ltd., Livingston, UK). The fluorescence was mea-
tion of 35 μM in 10 mM Tris-HCl buffer, pH 7.4 (containing 0.1 M sured using a quartz cuvette of 10 mm path length (Starna Scientific
NaCl) was stirred with a micro-stirring bar in a glass vial at 37 °C with Ltd., UK) with excitation and emission slit widths kept at 2 and 5 nm, re-
or without different concentrations of macromolecular crowding agents spectively. Relative intensity was obtained by calculating F/Fm, where F
and ethanol, in the presence or absence of different concentrations of is the fluorescence intensity of ThT in the presence of α-synuclein olig-
polyphenols and/or β-CD. At regular intervals, aliquots of 60 μL were omers/aggregates and Fm is the fluorescence intensity of ThT in pres-
taken out from the aggregating/disaggregating reaction mixtures ence of monomeric native α-synuclein. All fluorescence
which contained 35 μM of protein in 10 mM Tris-HCl buffer, pH 7.4 measurements were corrected for buffer contributions. CR absorbance
apart from respective concentrations of ethanol and/or macromolecular (at 540 nm) was recorded using a spectrophotometer (UV-2600,
crowders. The aggregating/disaggregating mixtures also contained Shimadzu, Japan) after incubation for 10 min at 25 °C. The absorbance
polyphenol–β-CD combinations during the inhibition of aggregation of protein only as a control in the absence of CR was also recorded by
and disaggregation experiments. The aliquots of 60 μL from aggregat- taking an aliquot (60 μL) of protein sample with 440 μL of Tris-HCl buff-
ing/disaggregating reaction mixtures were then mixed with 440 μL of er; similarly absorbance of CR only in the absence and presence of native
23 μM stock solution of ThT/CR (stock solution of ThT/CR was in monomeric α-synuclein as a control was also recorded with 20 μM final
592 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

concentration of CR in Tris-HCl buffer. All the experiments were carried with different concentrations of curcumin (7.5 μM), resveratrol (15 μM),
out in triplicate and the error was b 5%. For disaggregation experiments, baicalein (10 μM) and EGCG (15 μM) served as positive controls. Cells in-
preformed aggregates were obtained as described above and then the cubated with α-synuclein prefibrillar oligomers having initial concentra-
compounds were added. Aliquots were then taken out at regular tion of 0.5 mg/mL (or 35 μM) for monomer was served as negative
intervals. control. Test samples of α-synuclein samples having concentration of
Aggregation and disaggregation kinetics of α-synuclein showed sig- 0.5 mg/mL (or 35 μM for monomer) drawn at the native (0 min), oligo-
moidal behaviour reminiscent of nucleation dependent pathway and meric (30 min), prefibrillar (50 min) and filamentous fibrillar (120 min)
were fitted in a sigmoidal curve using OriginPro 8.5: states with or without optimized concentrations of polyphenols
(curcumin, resveratrol, baicalein and EGCG) and/or β-CD were added
A2−A1 and incubated for 12 h. The effective concentrations for curcumin, resver-
Y ¼ A1 þ ð1Þ
1 þ 10ðLOGt 0 −t Þp atrol, baicalein and EGCG in combination with β-CD were 7.5 μM (with 15
μM β-CD), 15 μM (with 30 μM β-CD), 10 μM (with 20 μM β-CD) and 15
where Y is ThT fluorescence intensity or CR absorbance, A1 is Y value at μM (with 30 μM β-CD), respectively. After incubation, MTT (sigma Al-
the bottom of the curve or bottom plateau, A2 is Y value at the top of the drich) was added at a concentration of 1 mg/mL and further incubated
curve or top plateau, LOGt0 is the time (in min) when the response is at 37 °C and 5% CO2 for 2–4 h. DMSO was added to the wells and plates
halfway between Bottom and Top (half maximum) and p is the Hill were read at 570 nm (multiskan Go, Thermoscientific, USA). The resulting
slope or Hill coefficient. absorbance values were converted to percentage viability with respect to
the untreated control cells. The statistical analysis for this data was done
2.5. Size exclusion chromatography (SEC) by paired t-test method.

SEC experiments were carried out using an AKTA purifier FPLC sys-
2.9. All atomistic molecular dynamics simulations
tem (GE Healthcare). The column used was Superdex 200 Increase 10/
300 GL column (I.D.: 1.0 cm × 30 cm). The elution was carried out
Molecular dynamics simulation was done using GROMACS 5.1.1 [47]
using 10 mM sodium phosphate buffer (pH 7.4) with 0.1 M NaCl with
employing GROMOS 54a7 all-atom force field using periodic boundary
a flow rate of 0.50 mL/min. All the samples were injected for chromato-
condition [48]. The starting model (PDB id: 2kkw, Model 1) was solvat-
graphic separation after centrifugation at 10,000 g for 20 min and filtra-
ed in a periodic box with SPC water model. The initial structures of test
tion through a 0.22 μm membrane filter.
compounds were imported from PubChem Compound Database while
the topology and other parameters were assigned using ATB topology
2.6. Circular dichroism spectroscopy
builder [49,50]. In separate setups for α-synuclein, simulations were
carried out in the absence and presence of polyphenols. Briefly, the
Far-UV circular dichroism spectra over the wavelength range of 195
monomers of the protein along with five monomers of individual poly-
to 250 nm were recorded on Jasco J-815 spectropolarimeter (Jasco Cor-
phenols were placed in random orientation in individual cubical boxes.
poration, Japan) equipped with a temperature controller. All the spectra
Counter ions were added to the solvent to neutralize electrical net
were accumulated at 25 °C using a cell with a path length of 0.1 cm and
charge of the system. The system was then minimized for 50,000
corrected for buffer contributions with five scans averaged for each
steps using a steepest decent algorithm followed by an equilibration
spectrum. Circular dichroism data are presented in terms of normalized
run of 100 ps in NVT (constant Number of particles, Volume, and Tem-
ellipticity as a function of wavelength.
perature) ensemble with restraints on the protein atoms. The NPT (con-
stant Number of particles, Pressure, and Temperature) ensemble was
2.7. Transmission electron microscopy (TEM)
used for production simulation. Systems were simulated at 310 K, main-
tained by a Berendsen thermostat [51], with a time constant of 1 ps,
Aliquots (5 μL) were taken out from different α-synuclein samples
with the protein and non-protein molecules coupled separately. Pres-
undergoing aggregation/inhibition of aggregation/disaggregation and
sure coupling was done employing a parrinello-rahman barostat [52]
placed on a carbon-coated Formvar grid (Ted Pella, Inc., Redding, CA,
using a 1 bar reference pressure and a time constant of 2.0 ps with com-
USA) and incubated for 5 min. Thereafter, the grids were washed with
pressibility of 4.5e−5 bar using isotropic scaling scheme. Electrostatic
MilliQ water and negatively stained with 2% (w/v) aqueous phospho-
interactions were calculated using the Particle Mesh Ewald (PME) sum-
tungstic acid (PTA) solution. PTA solution was freshly prepared in MilliQ
mation. The production run was performed for 80 ns each with 2 fs time
water and filtered through 0.22 μm sterile syringe filters before use.
step in every run. The coordinates were recorded at each 10 ps interval.
Electron micrographs were recorded digitally on a FEI Morgagni 268D
The resulting trajectories were analyzed by inbuilt GROMACS tools
instrument operated at 80 kV using magnifications in the range of
while the images were created using PyMol [53] package. Secondary
20,000–40,000X.
structure analysis was done using VMD package [54].
2.8. Cytotoxicity assay
3. Results
The toxicity of various polyphenol-mediated α-synuclein species
were tested on N2a cells. Cells were cultured in DMEM supplemented 3.1. Aggregation
with 10% FBS and 1% penicillin-streptomycin (Cell clone, Genetix bio-
tech Asia Pvt. Ltd., India) and incubated at 37 °C and 5% CO2. The cells Aggregation of α-synuclein was carried out in presence of the mac-
were seeded in 96 well plates at a density of 104 cells and were allowed romolecular crowding agents and was monitored using ThT (Thioflavin
to adhere and grow till the confluency reached to 85–90%. Experiments T) based fluorescence [43] and CR (congo red) binding assays [44]. Both
were set in triplicates on two 96 well plates. The viability of cells was an- these dyes are commonly used for following the progress of aggregation
alyzed using MTT assay that determines the mitochondrial dehydroge- and have been shown to complement other well established tech-
nase activity as described previously [46]. Only the viable cells reduce niques. Moreover due to concerns over ThT fluorescence data interpre-
MTT into formazan crystals by using the mitochondrial dehydrogenase tation in presence of the polyphenols [55], we have also used other
enzyme. To these different cells, components were added in the given techniques to further validate our findings such as circular dichroism,
concentrations as indicated and incubated for 12 h. Cells incubated with transmission electron microscopy, and size exclusion chromatography
buffer alone having final ethanol concentration of b1% (~0.35%, v/v) and apart from congo red absorbance.
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 593

pattern for α-synuclein with a lag phase, log phase (elongation and for-
mation of protofibrils) and a final stationary phase analogous to the gen-
eration of mature amyloid fibrils (Fig. 2 and Fig. S2, Supporting
Information). The rate of aggregation of α-synuclein was slower in case
of Dextran 40 than in the presence of Dextran 6 and Ficoll 70 (Table 1a).
Addition of 150 g/L crowders resulted in an aggregation profile and rate
of fibril formation similar to that of 100 g/L crowders (Table 1a, Fig. 2
and Fig. S2, Supporting Information). It can be deduced that the maximum
fibril formation was observed at a concentration of 100 g/L for all the three
crowders. Beyond this concentration the trend was reversed, showing a
decrease in the rate of aggregation and fibril formation (Table 1a).
To further accelerate the aggregation kinetics of α-synuclein, etha-
nol was used. At low concentrations, ethanol enhances the rate of fibril
formation without changing the morphology of the fibrils [7]. Therefore,
ethanol at three different concentrations of 5% (v/v), 7.5% (v/v) and 10%
(v/v) was added to the reaction system having α-synuclein with 100 g/L
Dextran 40 (Fig. S3, Supporting Information). In our studies we used an
ethanol concentration of 7.5% (v/v) as the optimum concentration
both with regards to the rate of fibril formation and the ease with
which the aggregation process could be followed. Above this concen-
tration (10%, v/v) the solution turned turbid spontaneously, while at
a concentration below this (5%, v/v), the aggregation process was
quite slow (Table S1, Supporting Information).
Aggregation of α-synuclein in the presence of different concentrations
of macromolecular crowding agents both in absence (Fig. 2) and presence
of 7.5% (v/v) ethanol (Fig. S4, Supporting Information) again showed no
change in the pattern of aggregation kinetics. Moreover the highest
amount of fibril formation and fastest aggregation kinetics were observed
at a concentration of 100 g/L for all the three macromolecular crowding
agents irrespective of whether ethanol was present or not (Table 1a and
Table S2, Supporting Information). In addition, slower aggregation kinet-
ics along with increased lag time were observed with an increase in
crowder concentration beyond 150 g/L for both (in presence or absence
of ethanol). Thus the only alteration observed was a decrease in lag
Fig. 2. Aggregation profiles of α-synuclein monitored using ThT fluorescence (λex = time with faster aggregation for the samples having ethanol. These data
430 nm and λem = 485 nm) in the presence of different concentrations of the taken together demonstrate that the only effect of the organic solvent
macromolecular crowding agents namely, Dextran 40, Dextran 6 and Ficoll 70.
was to increase the kinetics of aggregation and reduce the lag time keep-
Aggregation was carried out at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing
0.1 M NaCl with the protein concentration being kept at 35 μM. The data plotted are a
ing the other trends/characteristics intact (Fig. S4, Supporting Informa-
mean of the experiments done in triplicate and the standard deviation so obtained has tion). Finally, for the inhibition of aggregate formation and
been included for the individual data points. disaggregation of preformed synuclein fibrils, our main focus was primar-
ily on the 100 g/L crowder concentration since the latter brought about
The macromolecular crowding agents (Dextran 40, Dextran 6 and the maximum rate enhancement and fibril formation (Table 1a).
Ficoll 70) when added to a solution of α-synuclein (35 μM) decreased
the lag phase and increased the rate of formation of fibrils in a crowder 3.2. Inhibition of aggregation
concentration dependent manner up to 100 g/L for all the three crowders
as shown by the ThT (Fig. 2) and CR assays (Fig. S2, Supporting Informa- First we studied the inhibition of aggregation of α-synuclein by indi-
tion). The aggregation kinetics showed the characteristic sigmoidal vidual polyphenols. Addition of polyphenols decelerated the

Table 1
Kinetic parameters obtained (based on Eq. (1)) for aggregation and disaggregation (of preformed aggregates) of α-synuclein.

(a) Aggregation of α-synuclein in the presence of different concentrations of crowding agents (Fig. 2).

Concentration of the crowding agents (g/L) LOGt0⁎ for Dextran 40 LOGt0⁎ for Dextran 6 LOGt0⁎ for Ficoll 70
0 806.4 806.4 806.4
50 695.7 619.0 575.4
75 590.3 509.8 483.9
100 453.7 364.0 330.5
150 461.3 367.1 342.4
200 582.1 397.7 398.6
250 608.9 462.2 472.1
1.0
(b) Disaggregation of preformed aggregates of α-synuclein in the presence of three different macromolecular crowding agents (Dextran 40, Dextran 6 and Ficoll 70) at a
concentration of 100 g/L by the different optimized polyphenol–β-CD combinations (Fig. 5).

Crowding agent LOGt0⁎ for Curcumin and β-CD LOGt0⁎ for Resveratrol and β-CD LOGt0⁎ for Baicalein and β-CD LOGt0⁎ for EGCG and β-CD
Dextran 40 110.6 117.4 138.7 145.5
Dextran 6 131.0 136.2 159.6 163.9
Ficoll 70 132.8 138.9 162.5 165.9
⁎ LOGt0 = Time in min required for half maximum response.
594 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

aggregation kinetics and increased the lag phase in a polyphenol con- found these to be able to completely abolish the aggregation of α-synu-
centration dependent manner (Fig. 3 and Fig. S5, Supporting Informa- clein under these conditions (Fig. S11, Supporting Information).
tion). The effective concentrations of polyphenols, where no
aggregation was observed, were 25 μM, 60 μM, 30 μM and 60 μM for 3.3. Disaggregation
curcumin, resveratrol, baicalein and EGCG, respectively. These concen-
trations required for complete inhibition of aggregation of α-synuclein After successful inhibition of aggregation of α-synuclein by the
were slightly higher than those necessary in the absence of any of the polyphenol–β-CD combinations, we extended our investigation to the
macromolecular crowding agents (i.e. buffer only) (Table S3, disaggregation of preformed amyloid fibrils of α-synuclein in the ab-
Supporting Information). The trend for effectiveness of polyphenols in sence and presence of macromolecular crowding agents and ethanol.
the presence of Dextran 40 was observed to be curcumin N baicalein N The concentration of polyphenols and β-CD used were the same optimal
resveratrol ≈ EGCG, as defined by the lowest concentrations of the re- ones as obtained from the inhibition studies. Our results show that all
spective polyphenol required for complete inhibition. Addition of β- these combinations successfully disaggregated the preformed fibrils of
CD alone to α-synuclein could inhibit the aggregation but only at a α-synuclein in the presence as well as absence of macromolecular
much higher concentration of 200 μM (Fig. S6, Supporting Information). crowding agents (Fig. 5 and Fig. S12, Supporting Information). Irrespec-
This effect along with the known pseudochaperone action of β-CD [32] tive of whether the aggregates are formed in the presence or absence of
and our previous results with curcumin [22] obligated us to use it in ethanol and macromolecular crowding agents, the disaggregation pro-
combination with the polyphenols. Our earlier study has shown that ad- files by the polyphenol–β-CD combination was similar in all cases
dition of β-CD to curcumin in a ratio of 2:1 was most effective in (Fig. S13, Supporting Information). The kinetics of disaggregation by
inhibiting and disaggregating α-synuclein amyloids [30]. In this work all the combinations demonstrated a sigmoidal curve similar to aggre-
too, the addition of β-CD along with polyphenols in a ratio of 2:1 in gation kinetics after addition of the optimized combinations but with
the presence of Dextran 40 lowered the required concentration of less visible lag phase in all the cases (Fig. 5 and Fig. S12, Supporting In-
each of the polyphenols by a factor of 3 to 4 both in the absence of eth- formation). The rate of disaggregation was significantly faster in buffer
anol (Fig. 4 and Table S3, Supporting Information) as well as in its pres- only than in the presence of macromolecular crowding agents for all
ence (Fig. S7, Supporting Information). The effective concentrations for the combinations, with the shortest time required being for the
curcumin, resveratrol, baicalein and EGCG in combination with β-CD curcumin–β-CD combination (Table 1b). Amongst the macromolecular
were 7.5 μM (with 15 μM β-CD), 15 μM (with 30 μM β-CD), 10 μM crowding agents, the fastest rate of disaggregation was observed for
(with 20 μM β-CD) and 15 μM (with 30 μM β-CD), respectively. These Dextran 40 while Dextran 6 and Ficoll 70 were distinctly slower. All
combinations were also effective with Dextran 6 (Fig. S8, Supporting In- the combinations were able to completely disaggregate preformed am-
formation) and Ficoll 70 (Fig. S9, Supporting Information) both of which yloids of α-synuclein even at the much higher concentration of 200 g/L
induce faster aggregation than Dextran 40 (Fig. S10, Supporting Infor- for crowders both with and without ethanol (Fig. S14, Supporting Infor-
mation), thereby further proving the efficacy of the combinations as in- mation). This, in combination with our previous observations, conclu-
hibitors. We also explored the effectiveness of these optimized sively proves that ethanol's role was only to enhance the rates of
combinations for different crowder concentrations (0–200 g/L) and aggregation. When an enhanced dose/concentration of the combination

Fig. 3. Effect of different polyphenols at various concentrations on the aggregation of α-synuclein using ThT fluorescence (λex = 430 nm and λem = 485 nm) in the presence of Dextran 40
(100 g/L) as the macromolecular crowding agent. The experiments were carried out at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing 0.1 M NaCl and 7.5% (v/v) ethanol with the
protein concentration being kept at 35 μM. The data plotted are a mean of the experiments done in triplicate and the standard deviation so obtained has been included for the individual
data points.
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 595

Fig. 4. Effect of different polyphenols in combination with β-CD on the aggregation of α-synuclein using ThT fluorescence (λex = 430 nm and λem = 485 nm) in the presence of Dextran 40
(100 g/L) as the macromolecular crowding agent: (a) Curcumin with β-CD, (b) resveratrol with β-CD, (c) baicalein with β-CD and (d) EGCG with β-CD. The experiments were carried out
at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing 0.1 M NaCl with the protein concentration being maintained at 35 μM. The data plotted are a mean of the experiments done in
triplicate and the standard deviation so obtained has been included for the individual data points.

was used (keeping the polyphenol to β-CD ratio fixed at 1:2), the time again found to be the most effective one as observed from the rate of
required for disaggregation decreased (Fig. S15 and Table S4, disaggregation followed by baicalein, resveratrol and EGCG (Table S4,
Supporting Information). Curcumin in combination with β-CD was Supporting Information).

Fig. 5. Disaggregation of preformed aggregates of α-synuclein in the presence and absence of different macromolecular crowding agents: (a) curcumin (7.5 μM) with β-CD (15 μM), (b)
resveratrol (15 μM) with β-CD (30 μM), (c) baicalein (10 μM) with β-CD (20 μM) and (d) EGCG (15 μM) with β-CD (30 μM). Disaggregation profiles were measured using ThT fluorescence
(λex = 430 nm and λem = 485 nm) of preformed aggregates of α-synuclein (obtained after 800 min of aggregation) after addition of the respective polyphenol–β-CD combinations. The
experiments were carried out at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing 0.1 M NaCl with a protein concentration of 35 μM. The concentration of the macromolecular
crowding agents was 100 g/L. The data plotted are a mean of the experiments done in triplicate and the standard deviation so obtained has been included for the individual data points.
596 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

3.4. Secondary structure of α-synuclein decreased considerably with simultaneous appearance of higher molec-
ular weight peaks, the latter signifying the presence of α-synuclein olig-
Far UV circular dichroism spectroscopy was carried out to further omers and protofibrils. At 60 min, the monomeric protein peak
understand the effect of these polyphenols on the secondary structure disappeared completely and the peak due to intermediate oligomers
of α-synuclein. Native monomeric α-synuclein is an intrinsically disor- also started to disappear. This was accompanied by a decrease in the in-
dered protein and therefore showed a negative peak at around 200 nm tensity of the peak obtained near void volume corresponding to
corresponding to its random structure (Fig. S16, Supporting Informa- protofibrils due to the formation of higher molecular weight and insol-
tion). Aggregation of α-synuclein by incubating in the presence of dif- uble amyloid fibrils that cannot be analyzed using SEC. After 120 min,
ferent macromolecular crowding agents resulted in the formation of there was only one peak corresponding to protofibrils with significantly
β-sheet rich structure which is a characteristic of amyloids (Fig. S16, lower intensity. This decrease in the intensity of protofibrils was due to
Supporting Information). Introduction of optimized concentrations of the formation of higher molecular weight insoluble amyloid fibrils of α-
different polyphenols along with β-CD maintained the native random synuclein which were removed during centrifugation and filtration be-
structure of α-synuclein thereby preventing the formation of any inter- fore SEC analysis.
mediate molten globular or β-sheet conformation essential for amyloid We then investigated the effect of optimized combinations of poly-
formation (Fig. S17a, Supporting Information). This supported the phenols and β-CD on the elution profile of α-synuclein. No change
above results obtained using ThT and CR. Furthermore, we added poly- was observed in elution profile of α-synuclein (same as native α-synu-
phenols in combination with β-CD to the preformed amyloid fibrils of clein) under aggregating conditions even after 120 min in the presence
α-synuclein. The resultant α-synuclein species upon incubation with of optimized combinations of polyphenols and β-CD namely: curcumin
polyphenols and β-CD reverted to the random native structure rather (7.5 μM) with β-CD (15 μM), resveratrol (15 μM) with β-CD (30 μM),
than the initial β-sheet rich structure present in preformed amyloids baicalein (10 μM) with β-CD (20 μM) and EGCG (15 μM) with β-CD
(Fig. S17b, Supporting Information). (30 μM) (Fig. S18, Supporting Information). These data clearly demon-
strated that presence of these polyphenol–β-CD combinations did not
3.5. Size exclusion chromatography (SEC) analyses allow the protein to undergo aggregation or oligomerization and there-
by supported our ThT and CR data discussed earlier.
SEC analysis was carried out for the supernatants containing soluble Disaggregation of the preformed aggregates of α-synuclein was also
protein fractions after centrifugation. Elution profile of native mono- analyzed using SEC (Fig. 6b). Addition of curcumin (7.5 μM) and β-CD
meric α-synuclein during SEC analysis showed a peak resembling a (15 μM) to the aggregated α-synuclein showed a trend reverse of that
globular protein of 55 kDa (Fig. 6a). This is a well known property of observed during the aggregation process (Fig. 6a). After 30 min, elution
α-synuclein due to its intrinsically disordered nature and has been profile started to show peaks corresponding to oligomers and mono-
shown by other groups too [56]. We then studied the aggregation of mers. The intensity of the monomeric α-synuclein increased with
α-synuclein using SEC as a function of time (Fig. 6a). After 20 min, the time and only monomeric α-synuclein was present after 240 min show-
intensity of peak corresponding to native monomeric α-synuclein ing the completion of disaggregation process. Similar results were

Fig. 6. SEC analyses showing the elution profile of α-synuclein. (a) Aggregation of α-synuclein as a function of time and (b) Disaggregation profile after addition of curcumin (7.5 μM) with
β-CD (15 μM) to the preformed aggregates of α-synuclein. Aggregation was carried out at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing 0.1 M NaCl and ethanol (7.5%, v/v) with a
protein concentration of 35 μM in the presence of Dextran 40 (100 g/L). Disaggregation of preformed aggregates (obtained after 120 min of aggregation) was carried out after addition of
curcumin with β-CD under the same conditions. The arrows indicate the elution volumes of proteins used as molecular weight standards: (1) Void volume, (2) IgG (158 kDa), (3)
conalbumin (75 kDa), (4) bovine serum albumin (66.5 kDa), (5) ovalbumin (44 kDa), (6) super oxide dismutase 1 (32 kDa) and (7) lysozyme (14.4 kDa).
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 597

obtained with other optimized combinations of resveratrol (15 μM) polyphenols at sub-stoichiometric (with respect to that of α-synuclein)
with β-CD (30 μM), baicalein (10 μM) with β-CD (20 μM) and EGCG concentrations.
(15 μM) with β-CD (30 μM) and disaggregation was completed within
300 min in all these cases (Fig. S19, Supporting Information) as was
also shown by the ThT and CR assays. The SEC data thus also pointed to- 3.7. Role of β-cyclodextrin (β-CD)
wards the fact that these optimized combinations can not only inhibit
but also reverse the aggregation process. We also attempted to assess the role of β-CD used in combination
with the polyphenols in this study. As shown above, polyphenols
alone without β-CD were required at much higher concentration (Fig.
3.6. Transmission electron microscopy (TEM) 3 and Table S3, Supporting Information) than in combination to
completely inhibit the aggregation of α-synuclein. Substitution of β-
Aggregation of α-synuclein with different macromolecular CD in the optimized combination by other commonly used cyclodextrin
crowding agents (Dextran 40, Dextran 6 and Ficoll 70) resulted in the variants had a negative influence on the inhibition of aggregation (Fig.
formation of amyloid fibrils with similar morphology (Fig. S20a&b, 8a&b). Incorporation of either α-CD (Fig. 8a) or HP-β-CD [(2-Hydroxy-
Supporting Information). Incorporation of optimized combination of propyl)-β-cyclodextrin] (Fig. 8b) along with the polyphenols (with the
different polyphenols with β-CD to the solution of α-synuclein in the same optimized concentrations as used earlier with β-CD) could not in-
presence of macromolecular crowding agents completely inhibited the hibit the aggregate formation by α-synuclein. This implied that neither
formation of amyloid fibrils (Fig. S20d–g, Supporting Information). We α-CD nor HP-β-CD were as efficient as β-CD. The effect of CDs only, that
further analyzed the effect of these combinations of polyphenols and is, without the presence of the polyphenolic compounds, on inhibition
β-CD on the preformed amyloid fibrils of α-synuclein (Fig. 7). All the of aggregation was also investigated (Fig. 8c). β-CD was effective in
combinations disaggregated the preformed fibrils (Fig. 7 and Fig. S21, inhibiting the aggregation only when used at a high concentration of
Supporting Information) within the time duration (300 min) as ob- 200 μM with α-CD being the least effective while HP-β-CD showed mar-
served earlier using ThT fluorescence and CR absorbance. Therefore, ginal insignificant inhibition (Fig. 8c). Thus further experiments with α-
TEM images further confirmed the effectiveness of the unique, comple- CD or HP-β-CD on their abilities (in combination with the polyphenols)
mentary/synergistic and potent combination of β-CD with various to disaggregate α-synuclein aggregates were not performed. These data

Fig. 7. TEM images showing the disaggregation of preformed aggregates of α-synuclein in the presence of curcumin (7.5 μM) with β-CD (15 μM) at different time intervals of (a) 0 min, (b)
60 min, (c) 120 min, (d) 180 min (e) 210 min and (f) 240 min. Disaggregation of preformed aggregates of α-synuclein (obtained after 120 min of aggregation) was carried out after
addition of respective polyphenols with β-CD. Aggregation and disaggregation experiments were carried out at 37 °C with stirring in 10 mM Tris-HCl, pH 7.4 containing 0.1 M NaCl
and ethanol (7.5%, v/v) with a protein concentration of 35 μM in the presence of Dextran 40 (100 g/L). Scale bar is 200 nm.
598 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

therefore established the important role played by β-CD and its essence
in the combination with polyphenols.

3.8. Stoichiometry between protein and polyphenols/β-CD combination is


also important

The optimized combinations of polyphenol and β-CD were able to


inhibit the aggregation of α-synuclein present at a concentration of 35
μM (Fig. 4). But when the concentration of the protein was increased
even by 5 μM (i.e. a total protein concentration of 40 μM), complete in-
hibition of aggregation was not achieved by the optimized combination
of curcumin (7.5 μM) and β-CD (15 μM) (Fig. S22a, Supporting Informa-
tion). Further increase in the concentration of α-synuclein (studied up
to 100 μM) enhanced the level of aggregation and was proportional to
an increase in protein concentration (Fig. S22a, Supporting Information)
as expected. It is worth noting that the ratio of α-synuclein (with 35 μM
concentration) to curcumin in the optimized combination (7.5 μM
curcumin along with 15 μM β-CD) is around 4.66. With increased con-
centration of α-synuclein (100 μM), similar protein to polyphenol
ratio of 5.0 was able to completely inhibit the aggregation (20 μM
curcumin along with 40 μM β-CD) (Fig. S22b, Supporting Information)
thereby implying the significance of the stoichiometry in the present
case.

3.9. Cytotoxicity assays

To investigate whether the aggregation of α-synuclein and the com-


bination strategy employed by us for its inhibition or disaggregation can
be promoted as effective therapeutics, we tested their effect on the via-
bility of cultured N2a cells (mouse neuroblastoma cell lines). Since
prefibrillar aggregates are considered to be the most toxic species,
therefore cell viability assay was carried out with α-synuclein samples
taken at time points corresponding to prefibrillar aggregates (obtained
after 50 min of aggregation) [57]. As expected, α-synuclein at
Fig. 8. Effect of different cyclodextrins and polyphenols on the aggregation of α-synuclein prefibrillar stages induced drastic reduction (b 30%) in cell viability as
using ThT fluorescence (λex = 430 nm and λem = 485 nm). (a) α-CD in combination with compared to the controls that had no α-synuclein (Fig. 9). However,
different polyphenols, (b) HP-β-CD in combination with different polyphenols and (c)
α-synuclein species incubated with polyphenols alone showed signifi-
cyclodextrins only. The experiments were carried out at 37 °C with stirring in 10 mM
Tris-HCl, pH 7.4 containing 0.1 M NaCl and ethanol (7.5%, v/v) with a protein cant (N80%) increase in cell viability as compared to the prefibrillar spe-
concentration of 35 μM in the presence of Dextran 40 (100 g/L). [Abbreviations used: cies added without them (Fig. 9). Notably, when β-CD was also included
cur = curcumin; res = resveratrol; baic = baicalein; EGCG = (−)-epigallocatechin with these polyphenols a further increase in cell viability was observed,
gallate; ACD = α-CD; HPBCD = 2-Hydroxypropyl-β-cyclodextrin and BCD = β-CD]. with the most pronounced effect visible in the case of EGCG. Since the

Fig. 9. MTT reduction assay after 12 h incubation of N2a cells showing percentage viability in the presence or absence of α-synuclein prefibrillar aggregates (α-synuclein (P)). These are
species formed at 50 min time point of the aggregation kinetics. Cells incubated without or with polyphenols (curcumin, resveratrol, baicalein & ECGC) and/or β-CD for 12 h served as
positive controls, as represented by colored bars where there was almost no loss of viability. Incubation of cells with α-synuclein P alone reduced its viability drastically (to ~20%) and
hence served as negative control. Note that α-synuclein + β-CD combination also reduced cell viability. Compared to these, significant increase in cell viability (N80%) was observed
when cells were incubated with α-synuclein species formed in the presence of polyphenols and polyphenol + β-CD. P *p 0.05, non significant p values are not shown here.
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 599

* *

Fig. 10. Effect of decreasing concentrations of curcumin in presence of fixed concentration of β-CD in molar ratios of 5:1, 5:0.75, 5:0.5, 5:0.25 (as shown by dark green color bars) as plotted
along with their respective controls (as shown by light green color bars) that shows significant increase in the cell protective activity of polyphenol (curcumin) in presence of β-CD. P *p
0.05, non significant p values are not shown here.

effect of β-CD was not that evident as the polyphenols themselves effect showed a decrease (Fig. S23). This could be due to a narrow range
showed substantial effects, we performed another series of experiments of operational stoichiometry where the disaggregation effect of poly-
where the concentration of curcumin, was lowered from its optimum phenol is enhanced by the presence of β-CD.
value of 7.5 μM (Fig. 10). The data reveal that there is a significant in-
crease in cell survivability when β-CD is used in combination with poly- 4. Molecular dynamics simulations
phenol, confirming our hypothesis on the synergism exhibited by the
polyphenol-βCD combiation. The most pronounced increase was ob- To gain atomistic insights into the mechanism of action of these
served at the lowest concentration of curcumin (1.875 μM) in presence polyphenols, all-atom MD simulations were carried for α-synuclein
of β-CD (37.5 μM) as compared to that of curcumin (1.875 μM) alone. alone and in the presence of phenolic compounds. To correlate with
This effect however was visible only upto a certain stoichiometric ex-vivo outcomes, solution structure of human micelle-bound α-synu-
ratio of the polyphenol-βCD combination, beyond which the synergistic clein was used. The amino acid sequence of α-synuclein can be divided

Fig. 11. Gibbs free energy landscapes projected as function RMSD (nm) and Rg (nm) for simulations of alpha-synuclein alone (A) and in the presence of curcumin, baicalein, resveratrol and
EGCG (B–E). Conformations corresponding to lowest free energy basin attained during simulations are shown in above panel. The corresponding lower panel indicates changes in
secondary structure contents over the entire simulation of 80 ns. Transitions of residues to beta-conformation in simulations of alpha-synuclein alone are shown as marked by the red
rectangle.
600 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

into three regions, N-terminal region comprising of residues 1–60, cen- exhibit [40]. Remarkably, at higher concentrations (N150 g/L) of the
tral hydrophobic region consisting of residues 61–95 that are hydropho- macromolecular crowding agents, the rate of aggregation slowed
bic and C-terminal region comprising of residues 96–140, rich in acidic down, that is, showed a reverse trend for all the crowders (Table 1a).
residues (Fig. S1). The central region has proved to be crucial for aggre- Since the aggregation process includes the diffusional encounter of
gation, as its truncation results in reduced aggregation of protein. The monomers/oligomers with each other, it is natural to expect that the in-
Gibbs free energy landscape was projected as a function of backbone crease of viscosity that accompanies such high crowder concentrations,
RMSD and gyration radius (Rg). Backbone RMSD (root mean square de- would slow down the self-assembly process thereby giving rise to the
viation) is a measure of protein main-chain atomic deviations over the observed reduction in rates [62]. However, it has also been shown that
entire trajectory of simulation as compared to its initial starting confor- viscosity is not the major or sole reason for the observed decrease in ag-
mation (called the reference frame). The radius of gyration (Rg) is the gregation kinetics [63,64]. Schreiber and coworkers [59] have discussed
measure of protein compactness by calculating gyration about the x, y four distinct regimes of behavior depending on the polymer concentra-
and z-axes, as a function of time with atoms considered explicitly tion, namely, dilute, crossover, semi-dilute and concentrated regimes,
mass weighted. In simulation of α-synuclein alone, two distinct lowest with context to protein-protein association rates. In the semi-dilute re-
energy basins were observed (Fig. 11A). The corresponding coordinates gime, polymer concentration is high enough such that macromolecular
showed presence of distinct out of register/anti-parallel β-sheets crowders form a cage-like environment around the associating protein
(shown in snapshots and secondary structure profiles of residues). molecules thereby increasing the effective encounter rate leading to en-
However, in the presence of the polyphenols, no such intra-molecular hanced aggregation kinetics. However, in the concentrated regime, the
rearrangements were observed and the low energy basin drifted in polymers have the tendency to be situated in between the aggregating
the presence of compounds (Fig. 11B–E) towards a conformational sub- species, posing as obstacles and thereby slowing down the assembly
space having higher Rg values, suggestive of higher disordered struc- process by minimizing complex formation. Our observations are a
tures. Specifically in the presence of curcumin and EGCG, the lowest clear reflection of this concept with 100–150 g/L corresponding to the
energy basin drifted the most. Resveratrol however, instead of showing semi-dilute regime while beyond 200 g/L the concentrated polymer re-
such a drift, gave rise to multiple low energy basins accompanied by an gime comes into effect. On the other hand, the difference in the afore-
increase in the scatter of the Rg, signifying more conformational disor- mentioned rates of fibril formation in the presence of different
der. When analyzed, the corresponding lowest energy conformations macromolecular crowding agents (Table 1a) are attributed to the intrin-
showed binding of all the polyphenols to the core central region resi- sic differences in the molecular nature of the polymeric crowder mole-
dues. In the absence of compounds, these regions showed lower fluctu- cules exerting different extents of excluded volume. While dextran is a
ations, possibly due to stable intra-molecular interaction networks. polymer of glucose, Ficoll 70 is formed from the copolymerization of su-
Conversely, in the presence of the polyphenols, the core-amyloid re- crose with epichlorohydrin (Fig. 1). It is evident that both these crowder
gions (47–56 and 68–78) of the proteins showed higher RMSF (root molecules (Fig. 1) have enough \\OH groups to exhibit polar interac-
mean square fluctuations) possibly due to frequent contacts with tions and\\CH2\\moieties for hydrophobic contacts with the test pro-
these inhibitory compounds (Fig. S24). Moreover, these interactions tein. Such soft interactions between protein and crowder molecules
are dominated by stable H-bond networks (Fig. S25). A recent report have been shown to play an important role in protein conformational
on the role of DNA intercalators in modulating amyloidogenesis process distribution [65]. Dextran 6 with lowest average molecular weight
show dominant role of hetero-aromatic associations [58,59]. Detailed among the crowders used here has the maximum number of molecules
structural analysis revealed that besides other aromatic interactions, present at any given concentration giving rise to a higher packing den-
the Phe-95 of the central hydrophobic region of α synuclein is involved sity. This results in an increased excluded volume effect and hence the
in aromatic associations with the planar groups of the compounds. Res- faster aggregation rates for Dextran 6 as compared to Dextran 40 was
veratrol was an exception showing more intricate aromatic interactions expected. Had one followed the same line of argument, Ficoll 70 should
involving Tyr 125, 133 and 136 of α synuclein (Fig. S26). The simulation have shown the least effect on the aggregation. However, our observa-
outcomes suggests that direct interactions of compounds through H- tions reveal that both Dextran 6 and Ficoll 70 have comparable rates
bonding and aromatic associations subject monomeric α synuclein to suggesting that excluded volume is not the only dominant factor. We
unstructured aggregates thus controlling fibrillogenesis and associated propose that the enhancement of aggregation rates seen in presence
cytotoxicity. of Ficoll 70, can be attributed to the presence of significant soft interac-
tions between protein and crowder molecules in combination with the
5. Discussion excluded volume so exerted at the concentrations of the crowding agent
used. Thus, the slower aggregation kinetics seen in presence of Dextran
The physiological interior of cells present a very complex environ- 40 arises from the fact that this being of intermediate molecular weight,
ment, one which is not only congested, but at the same time also ex- it neither exerts as high an excluded volume as Dextran 6 nor exhibits
hibits a multitude of interactions (electrostatic, hydrophobic, the extent of soft interactions as that of Ficoll 70.
dispersion forces, excluded volume etc.) of the surrounding macromol- Based on the complexity of a congested environment, it is expected
ecules with the test protein [60]. Thus it is not surprising that dilute that the concentration of polyphenols and β-CD required for complete
buffer solutions might not be able to always provide an apt representa- inhibition of aggregation will be different than those in absence of the
tion of the cellular interior. Moreover given the fact that protein aggre- macromolecular crowding agents. Indeed this is apparent by the fact
gation is itself a very heterogeneous and intricate process, this in that the polyphenols alone or in combination with β-CD were required
combination with the added vagaries of the crowded milieu make the in a slightly higher concentration to bring about complete inhibition of
study of such self-assembly processes even more challenging. Further, aggregation in presence of the macromolecular crowders (Table S3,
any related attempts at trying to inhibit aggregate formation and/or try- Supporting Information). Curcumin in combination with β-CD was
ing to break up preformed protein aggregates need to be performed in most effective with lowest concentration required (among all four poly-
cellular-like environments such that any obvious shortcomings or prob- phenols used) for inhibiting and disaggregating amyloids formed by α-
lems can be addressed before taking such efforts to the next level (that synuclein. It was followed by baicalein while resveratrol and EGCG were
is, in vivo). In the present study, the aggregation of α-synuclein was ob- equally effective in combination with β-CD.
served to be much accelerated in presence of the macromolecular The next obvious question is ‘why β-CD’? Our comparative studies
crowders as compared to that in buffer only (Table 1a), very similar to with other commonly used cyclodextrin variants revealed that β-CD is
what has been observed before [61]. Such an increase has been attribut- the most effective one. Among the naturally occurring cyclodextrins,
ed to the excluded volume effect that these crowders are known to β-CD contains seven glucose units as compared to six in case of α-CD
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 601

and eight for γ-CD which directly correlates with the size of internal hy- vicinal \\OH groups as being one of the primary characteristics of the
drophobic cavity being 4.7–5.3 Å, 6.0–6.5 Å and 7.5–8.3 Å for α-CD, β- polyphenols [10,13] that are able to inhibit aggregation of α-synuclein.
CD and γ-CD, respectively [30]. β-CD, not being on the extreme ends However, our results reveal that other factors such as molecular flexibil-
in terms of size and properties, is the most commonly used cyclodextrin ity, hydrophobic interactions and size, as discussed above, also play a
in pharmaceutical formulations and drug delivery [34]. In the current major role in inhibition or disaggregation of protein aggregates. In this
study, β-CD showed excellent synergism along with the polyphenols regard, curcumin, which does not have vicinal polyhydroxy groups
in not only helping in inhibition of the aggregation but also disaggregat- but still is the most effective among the compounds studied here, is an
ing fibrils at sub-stoichiometric concentrations. Moreover, β-CD helps apt example of the possible structural aspects that an effective com-
in overcoming the limitations of polyphenolic compounds such as low pound needs to have when so designed.
bioavailability, in vivo instability [37], low efficiency [30] and inability The polyphenol–β-CD combinations were also effective in complete-
to cross blood-brain barrier at higher concentrations (i.e. 20 μM) etc. ly disaggregating the preformed aggregates of α-synuclein. Surprising-
[66]. Independent studies from our group have shown that β-CD signif- ly, the rate of disaggregation in the presence of Dextran 40 was faster as
icantly enhanced the in vitro stability of curcumin as compared to α-CD compared to Dextran 6 or Ficoll 70, this being just opposite to the trend
or HP-β-CD. HP-β-CD, a hydroxypropyl derivative of β-CD with similar observed for the aggregation kinetics (Table 1b and Fig. 5). This observa-
cavity dimensions but enhanced water solubility, was however not very tion fits quite well into our following hypothesis: since Dextran 40 nei-
effective and helpful for inhibiting amyloid formation. Thus, it can be ther exhibits high excluded volume nor extensive soft interactions, the
deduced that the optimum hydrophobicity (such as that present in β- access of the combination to the aggregates is easier thereby leading
CD) along with the cavity size is an important pre-requisite for to faster disassembly. Curcumin–β-CD showed the fastest disaggrega-
exhibiting the effects as seen in this study. tion kinetics than the other combinations in the presence of all three
The inhibition of α-synuclein aggregation involves the synergistic macromolecular crowding agents (Table 1b). Dose dependent increase
effect of both the polyphenols and β-CD. Curcumin has been shown to in the rate of disaggregation (Table S4 and Fig. S15, Supporting Informa-
act by concealing the hydrophobic patches on the protein molecules tion) further highlighted that the combination is working in a synergis-
and later on interacting via hydrogen bonding with the side chains of tic way with curcumin being most effective. The efficacy of the
amino acid residues present in protein molecules [24,29]. This interferes combinations used for disaggregation was same as that observed for
with two most important pre-requisites for amyloid formation: initial the inhibition of aggregate formation, thereby suggesting that similar
hydrophobic interactions required for nucleation and its growth and types of interactions between the polyphenol–β-CD units and the pro-
second, the hydrogen bonds stabilizing the β-sheets of amyloids [67]. teins were present. Berhanu & Masunov et al. [29,32] have shown
The two aromatic rings impart curcumin the ability to exhibit hydro- using MD simulations that polyphenols such as curcumin, exifone and
phobic interactions while the phenolic\\OH groups along with the car- myricetin disaggregate the fibrils by forming hydrogen bonds not just
bonyl groups help in interacting with protein molecules via hydrogen with side chain \\NH groups of protein molecules in fibrils but with
bonds. The linker between both the phenolic rings along with the ability the \\NH groups present in peptide backbone as well. This results in
for keto-enol tautomerism provides curcumin with a greater flexibility the loss of fibrillar hydrogen bonding leading to the dissociation of β-
in terms of adapting conformations that can help facilitate interactions sheets. Besides the intermolecular hydrogen bonding that the polyphe-
leading to the impediment of protein aggregation. Moreover, Reinke nols used in this study can exhibit with the aggregates, the presence of
and Gestwicki [68] have proposed three important features in mole- β-CD ensures, through hydrophobic interactions, that on dissociation,
cules required for inhibiting fibrillation: (a) existence of two terminal the component units do not recombine, thereby preventing auxiliary
aromatic groups required for interacting with protein molecules, (b) aggregation [72]. Besides, EGCG has been shown to exhibit a range of in-
substitution on the aromatic groups i.e. presence of hydroxyl groups teractions with monomeric and oligomeric proteins thereby hinting at
and (c) the length of the linker connecting the two aromatic groups the universality of these polyphenols being used more aggressively in
which should be 6–19 Å. Curcumin fulfills all these criteria and it is developing effective therapeutic approaches for neurodegenerative dis-
therefore not surprising that this polyphenol is the most effective eases arising from protein misfolding and aggregation [14,70].
among the ones studied here. A recent study has also provided insights There exist some key differences between the mechanisms of inhibi-
into the mechanism of action of curcumin and similarly found that it in- tion of aggregation and disaggregation. First, while for inhibition the
teracts via both hydrophobic effects and hydrogen bonding [69]. Pres- concentration of polyphenols (7.5 μM for curcumin, 10 μM for baicalein
ence of β-CD further helps in reducing the availability of hydrophobic and 15 μM for both resveratrol and EGCG) used is lesser than that of α-
patches by interacting with these, especially with the phenylalanine synuclein (35 μM), however during disaggregation the scenario is re-
residues present in α-synuclein [36]. The three other polyphenols (res- versed. In the latter, presence of fibrils or higher molecular weight ag-
veratrol, baicalein and EGCG) seem to interact mainly through the –OH gregates ensures that their concentration is much reduced as
groups by forming hydrogen bonds with residues of α-synuclein due to compared to the prevailing concentration of the polyphenols, since
the presence of vicinal polyhydroxyphenyl groups [10,13,14,70]. There- each aggregate is composed of many synuclein monomers. Second,
after, hydrophobic interactions take place and minimize further inter- the nature of the surface of α-synuclein exposed for interactions with
molecular interactions among α-synuclein molecules [71]. EGCG con- the polyphenols is different for the processes of inhibition and disaggre-
tains more\\OH groups than other polyphenols used in this study but gation. During inhibition, the exposed hydrophobic stretch (NAC re-
is least effective and is required at the highest concentration for gion) of the disordered protein, which is the main nucleation point for
inhibiting or disaggregating amyloids. It must be emphasized that the association, is blocked by the combination(s), presumably initiated via
size of EGCG arising from the bulkiness of three aromatic rings is higher interactions that are predominantly hydrophobic. On the other hand,
than others. This results in sterically limiting the movement and flexibil- in the aggregates, along with the presence of extensively hydrogen
ity of side groups present in EGCG as compared to curcumin which con- bonded β-sheets well-defined hydrophobic clefts also exist amongst
sists of a long aliphatic linker between two aromatic rings. On the the interdigitized monomers/oligomers. Such voids/cavities provide
contrary, though resveratrol contains a small aliphatic linker, it lacks more extensive hydrophobic interactions with the polyphenols used,
the vicinal\\OH groups which along with the restricted rotation around thereby further abetting subsequent cleavage into monomers with β-
its aliphatic double bond makes it far less effective than curcumin and CD helping to sequester the dissociated components through its
baicalein. Baicalein meanwhile possesses three vicinal polyhydroxy pseudochaperone-like action. Molecular dynamics simulations provid-
groups which coupled with the smaller size of the molecule, help in ed important insights on association of the compounds with monomeric
interacting even with interior i.e. relatively inaccessible segments of α-synuclein through favourable H-bond and aromatic interactions.
the aggregates. Previous studies have emphasized the presence of These interactions altered the conformational free energy subspace of
602 S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603

α-synuclein restricting its coil to cross-β transitions. Absence of such to delineate the details about the underlying forces at play at the molec-
transitions by polyphenols helped in remodelling the protein into an al- ular level.
ternative state which is less prone to aggregation. Such transitions could
be the triggering point for full amyloidogenesis of the protein. Transparency document
Interestingly the prefibrillar aggregates of α-synuclein in presence
of β-CD along with polyphenolic compounds were much less toxic The Transparency document associated with this article can be
than prefibrils alone as shown by cytotoxicity studies with N2a cells. found, in the online version.
However, the polyphenols alone were also effective in significantly re-
ducing the toxicity of α-synuclein prefibrils. Polyphenols alone have Acknowledgements
been shown to reduce the toxicity of α-synuclein oligomers/fibrils ear-
lier as well [11,14]. Recent studies based on the interaction of EGCG with PKC thanks the Department of Science and Technology (DST), New
amyloids have proposed that this naturally occurring polyphenol brings Delhi, India, for financial support under the Fast Track Scheme for Young
about restructuring of amyloids thereby rendering these aggregates Scientists (SR/FT/CS-007/2010) and IIT Delhi for startup funding. SG, PS
non-toxic and hence increasing cell-viability [14,71]. Otzen and co- and PP thank CSIR and SK thanks IIT Delhi for providing their fellow-
workers [73] have reported that EGCG helps in reducing cell toxicity in- ships. The authors thank IIT Delhi HPC facility for computational
duced by α-synuclein oligomers by preventing membrane disruption resources.
rather than bringing about any modulation of the structures of the pro-
tein aggregates. Reducing the concentration of curcumin by keeping References
that of β-CD fixed (Fig. 10) clearly brought out the effect of this pseu-
[1] D. Eisenberg, M. Jucker, The amyloid state of proteins in human diseases, Cell 148
do-chaperone and hence the role it plays in making this combination (2012) 1188–1203.
a potent one. These and many other studies, including the present [2] O. Marques, T.F. Outeiro, Alpha-synuclein: from secretion to dysfunction and death,
one, thus suggest that while polyphenols provide a promising approach Cell Death Dis. 3 (2012), e350.
[3] M. Bisaglia, S. Mammi, L. Bubacco, Structural insights on physiological functions and
for targeting neurodegenerative diseases, however, the exact mecha- pathological effects of alpha-synuclein, FASEB J. 23 (2009) 329–340.
nisms of their actions, in combination with improved bioavailability of [4] F. Cheng, G. Vivacqua, S. Yu, The role of alpha-synuclein in neurotransmission and
the same [74] need to be explored in depth at the molecular level before synaptic plasticity, J. Chem. Neuroanat. 42 (2011) 242–248.
[5] V.N. Uversky, Neuropathology, biochemistry, and biophysics of alpha-synuclein ag-
taking the next step towards drug design/formulation.
gregation, J. Neurochem. 103 (2007) 17–37.
[6] J.T. Bendor, T.P. Logan, R.H. Edwards, The function of alpha-synuclein, Neuron 79
(2013) 1044–1066.
[7] L.A. Munishkina, C. Phelan, V.N. Uversky, A.L. Fink, Conformational behavior and ag-
6. Conclusion
gregation of alpha-synuclein in organic solvents: modeling the effects of mem-
branes, Biochemistry 42 (2003) 2720–2730.
The combination(s) of the drug encapsulator β-CD and four poly- [8] G. Toth, S.J. Gardai, W. Zago, C.W. Bertoncini, N. Cremades, S.L. Roy, M.A. Tambe, J.C.
phenols are effective not only in inhibiting α-synuclein aggregation Rochet, C. Galvagnion, G. Skibinski, S. Finkbeiner, M. Bova, K. Regnstrom, S.S. Chiou,
J. Johnston, K. Callaway, J.P. Anderson, M.F. Jobling, A.K. Buell, T.A. Yednock, T.P.
but also in breaking up preformed aggregates of the disordered protein, Knowles, M. Vendruscolo, J. Christodoulou, C.M. Dobson, D. Schenk, L.
at much lower doses than had the components been used in isolation. McConlogue, Targeting the intrinsically disordered structural ensemble of alpha-sy-
The different chemical structures of these polyphenols have allowed nuclein by small molecules as a potential therapeutic strategy for Parkinson's dis-
ease, PLoS One 9 (2014) e87133.
us to throw insights into the mechanistic aspects underlining the [9] L. Breydo, J.W. Wu, V.N. Uversky, Alpha-synuclein misfolding and Parkinson's dis-
mode of action of the polyphenol–β-CD combinations. While the effica- ease, Biochim. Biophys. Acta 1822 (2012) 261–285.
cy of a polyphenol depends on a host of factors like structural flexibility, [10] M. Caruana, T. Högen, J. Levin, A. Hillmer, A. Giese, N. Vassallo, Inhibition and disag-
gregation of α-synuclein oligomers by natural polyphenolic compounds, FEBS Lett.
presence of phenolic \\OH groups and a flavour of hydrophobicity as 585 (2011) 1113–1120.
brought in by the benzene moiety, the uniqueness of β-CD arises from [11] P.K. Singh, V. Kotia, D. Ghosh, G.M. Mohite, A. Kumar, S.K. Maji, Curcumin modulates
its apt cavity size and associated hydrophobic interior thereby allowing α-synuclein aggregation and toxicity, ACS Chem. Neurosci. 4 (2013) 393–407.
[12] M.E. Herva, S. Zibaee, G. Fraser, R.A. Barker, M. Goedert, M.G. Spillantini, Anti-amy-
the latter to interact with hydrophobic amino acid side chains and
loid compounds inhibit alpha-synuclein aggregation induced by protein misfolding
hence block subsequent self-association of α-synuclein. Moreover, the cyclic amplification (PMCA), J. Biol. Chem. 289 (2014) 11897–11905.
ability of β-CD to help solubilize and stabilize the polyphenols through [13] X. Meng, L.A. Munishkina, A.L. Fink, V.N. Uversky, Molecular mechanisms underlying
the flavonoid-induced inhibition of alpha-synuclein fibrillation, Biochemistry 48
known hydrophobic encapsulation thus making these more bioavail-
(2009) 8206–8224.
able is an added advantage point of the combination employed in this [14] J. Bieschke, J. Russ, R.P. Friedrich, D.E. Ehrnhoefer, H. Wobst, K. Neugebauer, E.E.
study. Macromolecular crowding influenced the aggregation kinetics Wanker, EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and re-
as expected but in a subtle manner, such that the effects observed duces cellular toxicity, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 7710–7715.
[15] K.J. Korshavn, M. Jang, Y.J. Kwak, A. Kochi, S. Vertuani, A. Bhunia, S. Manfredini, A.
could not be ascribed to the excluded volume phenomenon alone. Addi- Ramamoorthy, M.H. Lim, Reactivity of metal-free and metal-associated amyloid-β
tionally, the fact that the sub-stoichiometric concentrations of these with glycosylated polyphenols and their esterified derivatives, Sci. Rep. 5 (2015)
compounds were equally effective under cell-mimicking conditions fur- 17842–17856.
[16] A.S. Detoma, S. Salamekh, A. Ramamoorthy, M.H. Lim, Misfolded proteins in
ther lend credence to and provide a scope for developing these with an Alzheimer's disease and type II diabetes, Chem. Soc. Rev. 41 (2012) 608–621.
eye on possible therapeutics. The ex vivo experiments with N2a cells [17] S.S. Hindo, A.M. Mancino, J.J. Braymer, Y. Liu, S. Vivekanandan, A. Ramamoorthy,
(mouse neuroblastoma cells) also supported the in vitro results demon- M.H. Lim, Small molecule modulators of copper-induced Aβ aggregation, J. Am.
Chem. Soc. 131 (2009) 16663–16665.
strating the effectiveness of the four polyphenols and their concoction [18] S. Lee, X. Zheng, J. Krishnamoorthy, M.G. Savelieff, H.M. Park, J.R. Brender, J.H. Kim,
with β-CD. This successful correlation exemplified the significance of J.S. Derrick, A. Kochi, H.J. Lee, C. Kim, A. Ramamoorthy, M.T. Bowers, M.H. Lim, Ratio-
using cell mimicking conditions (i.e. macromolecular crowding agents) nal design of a structural framework with potential use to develop chemical re-
agents that target and modulate multiple facets of Alzheimer's disease, J. Am.
while carrying out experiments in vitro rather than dilute buffer solu-
Chem. Soc. 136 (2013) 299–310.
tions. Since curcumin and EGCG have already received attention in [19] S.J. Hyunga, A.S. DeTomaa, J.R. Brender, S. Lee, S. Vivekanandan, A. Kochia, J.S. Choic,
modulating the aggregation of several neurodegenerative disease caus- A. Ramamoorthya, B.T. Ruotoloa, M.H. Lim, Insights into antiamyloidogenic proper-
ties of the green tea extract (−)-epigallocatechin-3-gallate toward metal-associated
ing proteins, it is worthwhile to try the aforementioned polyphenol–β-
amyloid-β species, Proc. Natl. Acad. Sci. 110 (2013) 3743–3748.
CD cocktail(s) for such cases too in order to probe the universality of the [20] J.S. Choi, J.J. Braymer, R.P.R. Nanga, A. Ramamoorthy, M.H. Lim, Design of small mol-
prescribed approach. Indeed with extensive focus worldwide on com- ecules that target metal-Aβ species and regulate metal-induced Aβ aggregation and
bating neurodegeneration and related ailments, such examples as neurotoxicity, Proc. Natl. Acad. Sci. 107 (2010) 21990–21995.
[21] W.M. Berhanu, A.E. Masunov, Natural polyphenols as inhibitors of amyloid aggrega-
used in this study provide alternate promising routes that one can em- tion. Molecular dynamics study of GNNQQNY heptapeptide decamer, Biophys.
bark upon, involving more focused attention and subsequent research, Chem. 149 (2010) 12–21.
S. Gautam et al. / Biochimica et Biophysica Acta 1865 (2017) 589–603 603

[22] S.A. Hudson, H. Ecroyd, F.C. Dehle, I.F. Musgrave, J.A. Carver, (−)-Epigallocatechin-3- [50] A.K. Malde, L. Zuo, M. Breeze, M. Stroet, D. Poger, P.C. Nair, C. Oostenbrink, A.E. Mark,
gallate (EGCG) maintains kappa-casein in its pre-fibrillar state without redirecting An automated force field topology builder (ATB) and repository: version 1.0, J.
its aggregation pathway, J. Mol. Biol. 392 (2009) 689–700. Chem. Theory Comput. 7 (2011) 4026–4037.
[23] D. Vauzour, A. Rodriguez-Mateos, G. Corona, M.J. Oruna-Concha, J.P. Spencer, Poly- [51] M. Parinello, A. Rahman, Polymorphic transitions in single crystals: a new molecular
phenols and human health: prevention of disease and mechanisms of action, Nutri- dynamics method, J. Appl. Phys. 52 (1981) 7182–7190.
ents 2 (2010) 1106–1131. [52] T. Darden, D. York, L. Pedersen, Particle mesh Ewald: an N-log(N) method for Ewald
[24] W.M. Berhanu, A.E. Masunov, Atomistic mechanism of polyphenol amyloid aggrega- sums in large systems, J. Chem. Phys. 98 (1993) 10089–10092.
tion inhibitors: molecular dynamics study of curcumin, exifone, and myricetin inter- [53] LLC. Schrodinger, The PyMOL Molecular Graphics System, Version 1.8, 2015.
action with the segment of tau peptide oligomer, J. Biomol. Struct. Dyn. 1-13 (2014). [54] W. Humphrey, A. Dalke, K. Schulten, VMD—visual molecular dynamics, J. Mol.
[25] B.B. Aggarwal, C. Sundaram, N. Malani, H. Ichikawa, Curcumin: the Indian solid gold, Graph. 14 (1996) 33–38.
Adv. Exp. Med. Biol. 595 (2007) 1–75. [55] Y. Suzuki, J.R. Brender, K. Hartman, A. Ramamoorthy, E.N.G. Marsh, Alternative path-
[26] N. Pandey, J. Strider, W.C. Nolan, S.X. Yan, J.E. Galvin, Curcumin inhibits aggregation ways of human islet amyloid polypeptide aggregation distinguished by 19F nuclear
of alpha-synuclein, Acta Neuropathol. 115 (2008) 479–489. magnetic resonance detected kinetics of monomer consumption, Biochemistry 51
[27] A.S. Pithadia, A. Bhunia, R. Sribalan, V. Padmini, C.A. Fierke, A. Ramamoorthy, Influ- (2012) 8154–8162.
ence of a curcumin derivative on hIAPP aggregation in the absence and presence [56] W. Ariesandi, C.F. Chang, T.E. Chen, Y.R. Chen, Temperature-dependent structural
of lipid membranes, Chem. Commun. 52 (2016) 942–945. changes of Parkinson's alpha-synuclein reveal the role of pre-existing oligomers in
[28] P. Arya, A. Srivastava, S.V. Vasaikar, G. Mukherjee, P. Mishra, B. Kundu, Selective in- alpha-synuclein fibrillization, PLoS One 8 (2013), e53487.
terception of gelsolin amyloidogenic stretch results in conformationally distinct ag- [57] D.P. Karpinar, M.B. Balija, S. Kugler, F. Opazo, N. Rezaei-Ghaleh, N. Wender, H.Y. Kim,
gregates with reduced toxicity, ACS Chem. Neurosci. 5 (2014) 982–992. G. Taschenberger, B.H. Falkenburger, H. Heise, A. Kumar, D. Riedel, L. Fichtner, A.
[29] B. Ahmad, L.J. Lapidus, Curcumin prevents aggregation in α-synuclein by increasing Voigt, G.H. Braus, K. Giller, S. Becker, A. Herzig, M. Baldus, H. Jackle, S. Eimer, J.B.
reconfiguration rate, J. Biol. Chem. 287 (2012) 9193–9199. Schulz, C. Griesinger, M. Zweckstetter, Pre-fibrillar alpha-synuclein variants with
[30] S. Gautam, S. Karmakar, A. Bose, P.K. Chowdhury, beta-Cyclodextrin and curcumin, a impaired beta-structure increase neurotoxicity in Parkinson's disease models,
potent cocktail for disaggregating and/or inhibiting amyloids: a case study with EMBO J. 28 (2009) 3256–3268.
alpha-synuclein, Biochemistry 53 (2014) 4081–4083. [58] J. Singh, A. Srivastava, P. Sharma, P. Pradhan, B. Kundu, DNA intercalators as amyloid
[31] E.M.M. Del Valle, Cyclodextrins and their uses: a review, Process Biochem. 39 (2004) assembly modulators: mechanistic insights, RSC Adv. 7 (2017) 493–506.
1033–1046. [59] A. Srivastava, S. Sharma, S. Sadanandan, S. Gupta, J. Singh, S. Gupta, V. Harids, B.
[32] S. Machida, S. Ogawa, S. Xiaohua, T. Takaha, K. Fujii, K. Hayashi, Cycloamylose as an Kundu, Modulation of prion polymerization and toxicity by rationally designed
efficient artificial chaperone for protein refolding, FEBS Lett. 486 (2000) 131–135. peptidomimetics, Biochem. J. 474 (2017) 123–147.
[33] F. Hapiot, H. Bricout, S. Tilloy, E. Monflier, Functionalized cyclodextrins as first and [60] R.J. Ellis, Macromolecular crowding: an important but neglected aspect of the intra-
second coordination sphere ligands for aqueous organometallic catalysis, Eur. J. cellular environment, Curr. Opin. Struct. Biol. 11 (2001) 114–119.
Inorg. Chem. (2012) 1571–1578. [61] V.N. Uversky, M.E. Cooper, K.S. Bower, J. Li, A.L. Fink, Accelerated alpha-synuclein fi-
[34] J. Zhang, P.X. Ma, Cyclodextrin-based supramolecular systems for drug delivery: re- brillation in crowded milieu, FEBS Lett. 515 (2002) 99–103.
cent progress and future perspective, Adv. Drug Deliv. Rev. 65 (2013) 1215–1233. [62] L.A. Munishkina, E.M. Cooper, V.N. Uversky, A.L. Fink, The effect of macromolecular
[35] Z.T. Wehrmann, T.W. Hulett, K.L. Huegel, K.T. Vaughan, O. Wiest, P. Helquist, H. crowding on protein aggregation and amyloid fibril formation, J. Mol. Recognit. 17
Goodson, Quantitative comparison of the efficacy of various compounds in lowering (2004) 456–464.
intracellular cholesterol levels in Niemann-Pick type C fibroblasts, PLoS One 7 [63] Y.Y. Kuttner, N. Kozer, E. Segal, G. Schreiber, G. Haran, Separating the contribution of
(2012), e48561. translational and rotational diffusion to protein association, J. Am. Chem. Soc. 127
[36] X.R. Qin, H. Abe, H. Nakanishi, NMR and CD studies on the interaction of Alzheimer (2005) 15138–15144.
beta-amyloid peptide (12-28) with beta-cyclodextrin, Biochem. Biophys. Res. [64] N. Kozer, Y.Y. Kuttner, G. Haran, G. Schreiber, Protein-protein association in polymer
Commun. 297 (2002) 1011–1015. solutions: from dilute to semidilute to concentrated, Biophys. J. 92 (2007)
[37] Z.X. Fang, B. Bhandari, Encapsulation of polyphenols—a review, Trends Food Sci. 2139–2149.
Technol. 21 (2010) 510–523. [65] L. Sapir, D. Harries, Is the depletion force entropic? Molecular crowding beyond ste-
[38] V.R. Yadav, S. Prasad, R. Kannappan, J. Ravindran, M.M. Chaturvedi, L. Vaahtera, J. ric interactions, Curr. Opin. Colloid Interface Sci. 20 (2015) 3–10.
Parkkinen, B.B. Aggarwal, Cyclodextrin-complexed curcumin exhibits anti-inflam- [66] L. Baum, A. Ng, Curcumin interaction with copper and iron suggests one possible
matory and antiproliferative activities superior to those of curcumin through higher mechanism of action in Alzheimer's disease animal models, J. Alzheimers Dis. 6
cellular uptake, Biochem. Pharmacol. 80 (2010) 1021–1032. (2004) 367–377 (discussion 443-369).
[39] M. D'Archivio, C. Filesi, R. Vari, B. Scazzocchio, R. Masella, Bioavailability of the poly- [67] K.E. Marshall, K.L. Morris, D. Charlton, N. O'Reilly, L. Lewis, H. Walden, L.C. Serpell,
phenols: status and controversies, Int. J. Mol. Sci. 11 (2010) 1321–1342. Hydrophobic, aromatic, and electrostatic interactions play a central role in amyloid
[40] H.X. Zhou, G. Rivas, A.P. Minton, Macromolecular crowding and confinement: bio- fibril formation and stability, Biochemistry 50 (2011) 2061–2071.
chemical, biophysical, and potential physiological consequences, Annu. Rev. [68] A.A. Reinke, J.E. Gestwicki, Structure-activity relationships of amyloid beta-aggrega-
Biophys. 37 (2008) 375–397. tion inhibitors based on curcumin: influence of linker length and flexibility, Chem.
[41] D. Gnutt, M. Gao, O. Brylski, M. Heyden, S. Ebbinghaus, Excluded-volume effects in Biol. Drug Des. 70 (2007) 206–215.
living cells, Angew. Chem. Int. Ed. Engl. 54 (2015) 2548–2551. [69] N.K. Bhatia, A. Srivastava, N. Katyal, N. Jain, M.A. Khan, B. Kundu, S. Deep, Curcumin
[42] J. Baell, M.A. Walters, Chemistry: Chemical con artists foil drug discovery, Nature binds to the pre-fibrillar aggregates of Cu/Zn superoxide dismutase (SOD1) and al-
513 (2014) 481–483. ters its amyloidogenic pathway resulting in reduced cytotoxicity, Biochim. Biophys.
[43] M.J. Volles, P.T. Lansbury, Relationships between the sequence of alpha-synuclein Acta 1854 (2015) 426–436.
and its membrane affinity, fibrillization propensity, and yeast toxicity, J. Mol. Biol. [70] P. Cao, D.P. Raleigh, Analysis of the inhibition and remodeling of islet amyloid poly-
366 (2007) 1510–1522. peptide amyloid fibers by flavanols, Biochemistry 51 (2012) 2670–2683.
[44] H. LeVine, Quantification of beta-sheet amyloid fibril structures with thioflavin T, [71] F.L. Palhano, J. Lee, N.P. Grimster, J.W. Kelly, Toward the molecular mechanism(s) by
Methods Enzymol. 309 (1999) 274–284. which EGCG treatment remodels mature amyloid fibrils, J. Am. Chem. Soc. 135
[45] W.E. Klunk, R.F. Jacob, R.P. Mason, Quantifying amyloid by congo red spectral shift (2013) 7503–7510.
assay, Methods Enzymol. 309 (1999) 285–305. [72] V.V. Shvadchak, M.M. Claessens, V. Subramaniam, Fibril breaking accelerates alpha-
[46] J. van Meerloo, G.J. Kaspers, J. Cloos, Cell sensitivity assays: the MTT assay, Methods synuclein fibrillization, J. Phys. Chem. B 119 (2015) 1912–1918.
Mol. Biol. 731 (2011) 237–245. [73] N. Lorenzen, S.B. Nielsen, Y. Yoshimura, B.S. Vad, C.B. Andersen, C. Betzer, J.D.
[47] GROMACS: high performance molecular simulations through multi-level parallel- Kaspersen, G. Christiansen, J.S. Pedersen, P.H. Jensen, F.A. Mulder, D.E. Otzen, How
ism from laptops to supercomputers. epigallocatechin gallate can inhibit alpha-synuclein oligomer toxicity in vitro, J.
[48] Z. Lin, W.F. van Gunsteren, Refinement of the application of the GROMOS 54A7 force Biol. Chem. 289 (2014) 21299–21310.
field to beta-peptides, J. Comput. Chem. 34 (2013) 2796–2805. [74] M. Singh, M. Arseneault, T. Sanderson, V. Murthy, C. Ramassamy, Challenges for re-
[49] K.B. Koziara, M. Stroet, A.K. Malde, A.E. Mark, Testing and validation of the Automat- search on polyphenols from foods in Alzheimer's disease: bioavailability, metabo-
ed Topology Builder (ATB) version 2.0: prediction of hydration free enthalpies, J. lism, and cellular and molecular mechanisms, J. Agric. Food Chem. 56 (2008)
Comput. Aided Mol. Des. 28 (2014) 221–233. 4855–4873.

You might also like