Sei sulla pagina 1di 9

European Journal of Pharmaceutical Sciences 46 (2012) 17–25

Contents lists available at SciVerse ScienceDirect

European Journal of Pharmaceutical Sciences


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

Neuroprotective effect of undecylenic acid extracted from Ricinus communis L.


through inhibition of l-calpain
Eunyoung Lee a, Ji-Eun Eom a, Hye-Lin Kim a, Da-Hye Kang a, Kyu-Yeon Jun a, Duk Sang Jung b,
Youngjoo Kwon a,⇑
a
College of Pharmacy, Division of Life & Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
b
Department of Chemistry, Cheju National University, 1 Ara 1-dong, Jeju 690-756, Republic of Korea

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

Article history: The key neuropathological features of Alzheimer’s disease are abnormal deposition of Ab plaques and
Received 30 November 2011 insoluble Ab peptides in extracellular brain and intracellular neurofibril tangles induced by abnormal
Accepted 31 January 2012 tau hyperphosphorylation. l-Calpain is one of the factors that bridge these Ab- and hyperphosphorylated
Available online 8 February 2012
tau-mediated pathological pathways. Undecylenic acid (UDA), a naturally occurring unsaturated fatty
acid, was discovered as a l-calpain inhibitor by screening a chemical library using a substrate specific
Keywords: l-calpain assay method. UDA inhibited Ab oligomerization and Ab fibrillation and reversed Ab-induced
l-Calpain inhibitor
neuronal cell death. In addition, UDA scavenged ROS and reversed the levels of proapoptotic proteins
Undecylenic acid
Extraction of Ricinus communis L.
induced by ROS in SH-SY5Y cells. UDA inhibited l-calpain activity with better potency than the known
p25 Generation peptide-like l-calpain inhibitor, MDL28170, in SH-SY5Y and HEK293T cells transfected with the catalytic
Tau hyperphosphorylation subunit of l-calpain. These results suggest that UDA is a novel non-peptide-like l-calpain inhibitor with
good cell permeability and potent neuroprotective effect.
Ó 2012 Elsevier B.V. All rights reserved.

1. Introduction Calpains, cytoplasmic cysteine proteases, are activated by


increased intracellular calcium and are essential for normal physi-
Alzheimer’s disease (AD) is a major neurodegenerative disorder ologic neuronal function. However, alteration in calcium
of the aging brain (Conway et al., 2003). The characteristics of AD homeostasis generated by insoluble Ab leads to persistent hyperac-
are neurofibrillary tangles (NFTs) comprised of hyperphosphoryl- tivation of calpains. The two most widely distributed isoforms are
ated tau and senile plaques made up of aggregated amyloid beta l-calpain (calpain1) and m-calpain (calpain2). l-Calpain requires
protein (Ab) (Selkoe, 1991). Calpain may be one of the factors that micromolar concentrations of calcium for full activity and m-cal-
bridge the two pathways (Camins et al., 2006). The insoluble Ab pain requires millimolar concentrations of calcium in vitro. l-Cal-
peptide is caused by proteolytic cleavage of amyloid precursor pro- pain is the major form in neuronal cells and many blood
tein (APP), a type-1 transmembrane protein (Kang et al., 1987). The elements (Mellgren, 1990; Saido et al., 1992; Salamino et al.,
N-terminal of APP cleaved by b-secretase (also called BACE1/Asp2/ 1993). Hyperactivated l-calpain cleaves a number of neuronal
memapsin2) becomes the soluble APP-b and successive cleavage substrates including p35, which negatively affects neuronal struc-
by c-secretase generates insoluble Ab. Through the secretion ture and function, leading to inhibition of essential neuronal sur-
pathway, sorted insoluble Ab is accumulated in outside the cell vival mechanisms. CDK5, a protein kinase, is involved in terminal
(Buxbaum et al., 1998). Depending on where c-secretase cuts in differentiation of neurons and muscle cells. l-Calpain-mediated
C-terminal APP, Ab1–40 and Ab1–42 are mainly formed (Younkin, p35 cleavage into p25 results in the increase in CDK5 activity be-
1998). In particular, the accumulation of Ab1–42 results in Ab cause p25/CDK5 has a more stable conformation than p35/CDK5.
oligomers and fibrils which result in the forming of extracellular The prolonged activation of CDK5 by complexing with p25 induces
amyloid plaques (Demattos et al., 2001). In AD, chronic toxicity tau to be hyperphosphorylated, leading to the formation of neuro-
of aggregated Ab1–42 could lead to sustained intracellular calcium fibrillary tangles (NFTs) and neurodegeneration in AD (Nikolic
elevations in susceptible central neurons and thus calpain activa- et al., 1996; Patrick et al., 1999; Lee et al., 2000; Hashiguchi
tion (Saito et al., 1993). Eventually, the Ab peptide causes synapse et al., 2002; Cruz et al., 2003; Noble et al., 2003). Conclusively, l-
and neuronal cell loss (Smith et al., 2006). calpain plays an important role in Ab-mediated NFT formation
and the development of a l-calpain inhibitor may be a good strat-
⇑ Corresponding author. Tel.: +82 2 3277 4653; fax: +82 2 3277 3051. egy for drug developers targeting AD. In particular, targeting the
E-mail address: ykwon@ewha.ac.kr (Y. Kwon). p35 cleavage activity of l-calpain among its proteolytic activities

0928-0987/$ - see front matter Ó 2012 Elsevier B.V. All rights reserved.
doi:10.1016/j.ejps.2012.01.015
18 E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25

with other diverse substrates is of great interest in order to develop The fluorometric l-calpain assay was performed as described
l-calpain specific inhibitors in consideration of several reasons as previously (Kang et al., 1009). l-Calpain (human erythrocyte) was
follows. First, the accumulation of p25 itself was observed in the obtained from Calbiochem (Darmstadt, Germany). Pep1 and pep2
brain of early stage AD patients but not in the terminal stage AD were substrates of l-calpain and synthesized by the Peptron Corp.
patients. Second, p25/CDK5 causes cytoskeletal disruption and (Daejeon, Korea). Pep1 was derived from p35 cleavage site ((2-
apoptosis as well as tau hyperphosphorylation in neurons (Nikolic Abz)-Ser-Thr-Phe-Ala-Gln-Pro-(3-nitrotyrosine)-NH2) and pep2
et al., 1996; Patrick et al., 1999). Third, there are neither introns in originated from the calpain cleavage site in a-spectrin ((2-Abz)-
the open reading frame of p35, a neuronal-specific CDK5 regulator, Glu-Val-Tyr-Gly-Met-Met-(3-nitrotyrosine)-NH2). MDL28170 and
nor internal methionine near the beginning of the p25 sequence, E64d (Sigma, USA) were used as positive controls for l-calpain inhi-
suggesting that p25 is only generated by proteolytic cleavage of bition. The assay was performed in a final volume of 100 lL. Stock
calpain (Lee et al., 2000; Chae et al., 1997). solution of pep1, pep2 and compounds were prepared in dimethyl-
In order to develop l-calpain inhibitors with selectivity against sulfoxide (DMSO) and stored at 20 °C until needed. l-Calpain inhi-
other enzymes belonging to the cysteine protease family and bition was assayed in reaction buffer (50 mM Tris–HCl, 50 mM NaCl,
against other endogenous substrates of l-calpain, we previously 1 mM EDTA, 1 mM EGTA and 5 mM b-mercaptoethanol, pH 7.5)
developed an efficient high throughput screening system using with 100 lM pep1 or pep2, 2.5 mM CaCl2 and 5.25 U/mL l-calpain.
fluorescence probes (Kang et al., 2009). In this study, we were pro- The reaction was initiated by adding in the following order: sub-
vided with a profiled small molecular chemical library containing strate, l-calpain, test compound and CaCl2 solution. Thereafter,
6480 compounds from the Korean Chemical Bank to discover novel the mixture was incubated with shaking at room temperature for
calpain inhibitors as potent drug-like compounds for AD. Several 30 min. Fluorescence intensities were determined by using wave-
potent compounds were selected from the chemical library. lengths of 320 nm for excitation and 420 nm for emission.
Among the 6480 compounds, undecylenic acid (UDA) extracted Cathepsin B and L inhibitory activities were assayed in the reac-
from Ricinus communis L. was chosen for further study due to its tion buffer (50 mM NaOAc–HCl, 2 mM dithiothreitor (DTT), 2 mM
low toxicity and potent calpain inhibitory activity. EDTA, pH 5.5 for cathepsin B and 0.1 M NaOAc–HCl, 1 mM EDTA,
UDA, an 11-carbon monounsaturated fatty acid (C11H2O2), 0.1% b-mercaptoethanol, pH 5.5 for cathepsin L) with 20 lM sub-
found naturally in the body (occurring in sweat), is produced com- strate and 1.5 nM cathepsin B or 4 nM cathepsin L. Cathepsin B
mercially by vacuum distillation of castor oil, via the pyrolysis of and L were obtained from Calbiochem (Darmstadt, Germany).
ricinoleic acid. It is an economical antifungal agent and the active The substrates used were RR-AMC (Sigma, USA) for cathepsin B
ingredient in many topical over-the-counter antifungal prepara- and Z-FR-AMC (Sigma, USA) for cathepsin L. The cathepsins were
tions (Shapiro and Rothaman, 1945). We, in the current study, reductively activated by preincubation in assay buffer at 37 °C for
found UDA to have a neuroprotective effect against neurotoxin- 30 min prior to initiating the reaction with substrate and test com-
induced apoptosis in SH-SY5Y, a human neuroblastoma cell, for pounds. Afterwards, the reaction mixture was incubated at room
the first time. The mechanism underlying the neuroprotective temperature for 30 min. Fluorescence intensities were determined
effect of UDA was studied and it functioned as a l-calpain inhibi- by using wavelengths of 360 nm for excitation and 450 nm for
tor, leading to reduction of p25 formation, CDK5 activation, and tau emission. CA-074, Z-FF-FMK and nimodipine were used as a
phosphorylation. UDA may represent a potential adjuvant strategy cathepsin B inhibitor, cathepsin L inhibitor and calcium channel
for AD treatment. blocker, respectively, and all were purchased from Sigma Chemical
Co. (USA).

2. Materials and methods


2.3. Cell viability test
2.1. Cell cultures
SH-SY5Y cells were seeded in 96-well microplates (105 cells per
SH-SY5Y, a human neuroblastoma cell line (ATCC CRL-2266),
each well in 100 lL medium) for 24 h. Then, cells were rinsed twice
and HEK293T, a human kidney epithelial cell line (ATCC CRL-
with phosphate-buffered saline (PBS). After washing, the cells were
11268), were purchased from the Korean Cell Line Bank (Seoul,
incubated in FBS free media with 10 mM glutamate combined with
Korea). SH-SY5Y and HEK293T cells were grown in Minimum
10 mM glycine, 2 mM H2O2 and 25 lM Ab1–42 (Bachem, Switzer-
Essential Medium (MEM, Welgene, Korea) and Dulbecco’s modified
land), or neurotoxin with compound. Cells were treated with glu-
Eagle’s medium (DMEM, Welgene, Korea), respectively, in a
tamate and H2O2 for 24 h. Ab1–42 was treated for 72 h. Prior to
humidified incubator maintained at 37 °C under 5% CO2. Each med-
Ab1–42 treatment, Ab1–42 needed oligomerization since it has been
ium contained 10% fetal bovine serum (FBS, Hyclone laboratories
reported that the Ab1–42 oligomer was more toxic than Ab1–42 fibril
Inc. USA) and 1% penicillin/streptomycin (Sigma Chemical Co.,
(Dahlgren et al., 2002). The oligomerised Ab1–42 was obtained as
USA).
previously reported with minor modification (Dahlgren et al.,
2002). Lyophilized, HPLC-purified Ab1–42 was reconstituted in
2.2. Fluorometric assay for l-calpain, cathepsin B and cathepsin L hexafluoroisopropanol (HFIP, Sigma, USA) at 20–23 °C using amber
microcentrifuge tubes for 30 min and then evaporated in a speed
A fluorometric assay was performed in 96-well plates and fluo- vacuum to remove residual HFIP. The dried Ab1–42 was dissolved
rescence intensity in each well was measured using a Microplate in DMSO with sonication at room temperature in a water bath
Fluorescence Reader (SpectraMAX GEMINI EM, Molecular De- and then was aliquoted. The Ab1–42 aliquot was kept at 20 °C be-
vices); the IC50 values were obtained using the data graphing soft- fore use. Each aliquot was diluted with cold PBS, vortexed for 30 s
ware, TableCurve 2D (Systat software Inc.). Fluorescence intensity and then incubated for 16 h at 4 °C for oligomerization before
was expressed as relative fluorescence units (RFU). RFU was calcu- treatment of cells. During the experiments, pre-oligomerized Ab
lated by subtracting the RFU of the no-enzyme control from all aliquots were added to a final concentration of 25 lM directly to
other values. To determine percent inhibition, the percent change the cell media. Finally, after treating with neurotoxin only or
in RFU between the activity of the enzyme in the presence and ab- cotreating with neurotoxin and compound, 5 lL of cell counting
sence of compounds was calculated. The RFU value calculated in kit-8 solution (Dojindo, Japan) was added to each well. The micro-
the absence of compound represents 100% enzyme activity. plate was incubated at 37 °C for an additional 1 h and the
E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25 19

absorbance of each well was assessed at 450 nm using an ELISA and Ab1–42 oligomerized with co-incubation of each compound
Microplate Reader (VersaMax, Molecular Devices). was placed on the mica and dried. Then the sample was scanned
by AFM (XE-70, Park Systems, Korea), and analyzed by XEI (Park
2.4. Evaluation of the effect of compound on amyloidal plaque Systems, Korea). The reading scale of AFM micrograph was a
formation 4 lm  4 lm square, composed of 256  256 pixels. Each micro-
graph was measured with 256 maximum heights from each line
SH-SY5Y cells were exposed to 25 lM of pre-oligomerized Ab1– per pixel and average maximum height was obtained. The inhibi-
42 in the absence or presence of each compound which included tion percentage of oligomerization was calculated from average
CA074, Z-FF-FMK, pepstatin A, nimodipine, MDL28170 and UDA maximum heights of compound cotreated versus oligomerized
(all 20 lM) for 72 h at 37 °C in a humidified incubator with 5% Ab1–42 without compound.
CO2. The amyloidal plaque was observed and imaged with a fluo-
rescence inverted microscope with magnification of 20 for objec-
tive lens and 10 for ocular lens (Axiovert 200, Germany). 2.8. Western blotting analysis

2.5. Measurement of intracellular reactive oxygen species formation The cells were lysed in lysis buffer solution containing 50 mM
Tris–HCl, 300 mM NaCl, 1% Triton X-100, 10% glycerol, 1.5 mM
Free radical production was measured by incubating the cells MgCl2, 1 mM CaCl2, 1 mM PMSF and 1% protease inhibitor cocktail.
with the fluorescent probe 20 ,70 -dichlorodihydrofluorescein diace- Protein concentrations of soluble extracts in the lysate were deter-
tate (DCFH2-DA, Molecular Probe, USA). DCFH2-DA, which is mined by BCA™ Protein Assay Kit (Pierce, USA). A total of 40 lg of
oxidized to fluorescent 20 ,70 -dichlorofluorescin (DCF) by hydroper- protein was loaded per well. Samples were run on a 12% polyacryl-
oxides, was used to measure the relative levels of cellular perox- amide gel and transferred to a polyvinylidene fluoride membrane
ides (Fu et al., 1998). SH-SY5Y cells were incubated in a 24-well (Millipore Corporation, USA). Membranes were blocked for 1 h at
culture dish (4  105 cells per well in 400 lL of medium) for room temperature in TBST with 5% skim milk. Membranes were
24 h. Subsequently, cells were rinsed with PBS. After washing, cells incubated with each antibody of a-tubulin (1:1000 dilution, load-
were treated with various compounds in FBS free medium and ing control), caspase-3 (1:1000), p35/25 (1:1000), tau and p-tau
then incubated at 37 °C for 24 h. After incubation, cells were trea- (1:1000) in TBST with 5% skim milk overnight at 4 °C. Secondary
ted with 400 lM H2O2 for different durations (5, 10, 15 and antibodies used were anti-mouse IgG horseradish peroxidase for
20 min). Each well was rinsed with standard medium (138 mM tau and p-tau, and anti-rabbit IgG horseradish peroxidase for a-
NaCl, 2.7 mM KCl, 1.2 mM CaCl2, 1.2 mM MgCl2, 10 mM PBS and tubulin and p35/25 antibodies. All primary and secondary antibod-
10 mM glucose, pH 7.4). Next, cells were treated with 5 lM of ies used were purchased from Cell Signaling Technology Inc (USA).
DCFH2-DA for 30 min at 37 °C in the standard medium. Subse- After incubation with each antibody, membranes were washed
quently, cells were washed once with standard medium and the three times with TBST and then were detected with the ECL plus
fluorescence was measured using excitation and emission wave- Western blotting detection reagent (Ab frontier, Korea). Western
lengths of 485 nm and 535 nm, respectively in a microplate fluo- blot images were analyzed using Multi-Gauge Software (Fuji photo
rescence reader (SpectraMAX GEMINI EM, Molecular Devices). film Co., Ltd., Japan).

2.6. Thioflavin T fluorescence assay


2.9. l-Calpain assay in cells
The thioflavin T (ThT) fluorescence assay was performed with
synthetic Ab1–42 (Bachem, Switzerland) as described previously In order to measure l-calpain inhibitory activity of compounds
(Kim et al., 2006). The Ab1–42 solution at a final concentration of in live SH-SY5Y and HEK293T cells, the human CAPN1 gene which
25 lM was incubated at 37 °C for 48 h in PBS (50 mM NaH2PO4, encodes the l-calpain catalytic subunit responsible for proteolytic
100 mM NaCl, 0.02% NaN3) to get aggregation in the presence or activity was synthesized (Bioneer, Korea) (Camins et al., 2006).
absence of test compounds. After incubation, 15 lM ThT was The plasmid of CAPN1-encoding pcDNA3.1/His A was subcloned
added to each well containing the aggregated Ab fibril, followed using the restriction sites EcoR V and Xho I and transfected into
by additional incubation for 15 min at 37 °C. ThT fluorescence SH-SY5Y and HEK293T cells, then incubated for various times using
was measured at 450 nm emission/490 nm excitation wavelengths WelFect QTM Plus (Welgene, Korea). The proper transfection time
in a microplate fluorescence reader (SpectraMAX GEMINI EM, was set up by monitoring the extent of protein expression with
Molecular Devices). Readings were normalized to the DMSO-only Western blotting analysis using l-calpain antibody (Cell Signaling
negative control and expressed as a percentage of relative fluores- Tech., USA). After transfection, cells were incubated with each com-
cence to control. Tannic acid, shown previously to inhibit Ab aggre- pound for 210 min in calpain reaction buffer additionally containing
gation, was used as a positive control (Ono et al., 2004a,b). 0.1% Triton X-100. Cleavage product of pep1 by l-calpain was mea-
sured with a kinetic mode on the microplate fluorescence reader
2.7. Atomic force microscopy (AFM) measurements (SpectraMAX GEMINI EM, Molecular Devices) at 37 °C. Fluorescence
was measured at 320 nm excitation/420 nm emission wavelengths.
The Ab1–42 solution in a final concentration of 12.5 lM was
freshly prepared in PBS and aliquoted. The Ab1–42 solution exists
in monomer when freshly prepared. Oligomerized Ab1–42 was pre- 2.10. Statistical analysis
pared according to the method described in the cell viability assay.
The fibril form of Ab1–42 was arranged by incubation at 37 °C for In all experiments, data are expressed as mean ± standard devi-
48 h. During the oligomerization process, each compound was ation, with at least three replicates in each experimental group.
co-incubated to a final concentration of 20 lM to check the ability Comparison of differences was conducted by using an unpaired,
of compounds to block Ab1–42 oligomerization. Twenty microliters two-tailed Student t-test. The difference was considered statisti-
of each aliquot of monomer, oligomer, fibril Ab1–42 sample solution cally significant when the p value was 60.05.
20 E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25

3. Results DMSO. The decreased cell viability mediated by oligomerized


Ab1–42 was attenuated by co-treatment with CA074, Z-FF-FMK,
3.1. Discovery of a novel l-calpain inhibitor by screening a small pepstatin A, MDL28170 and UDA (Fig. 1A). Among the compounds,
molecular chemical library Z-FF-FMK, which is known as a specific inhibitor of cathepsin L,
inhibited both cathepsin B and L to a similar extent (Table 1) and
A small molecular chemical library provided by Korean Chemi- recovered the cell viability up to 98.7 ± 0.4%. UDA co-treatment
cal Bank was screened to discover novel calpain inhibitors using also enhanced viability of Ab1–42-treated cells to 91.3 ± 0.7% with
the fluorometric l-calpain assay method established in our labora- a statistical significance, even though its effect was much less than
tory. The library was composed of 6480 compounds which have di- that of Z-FF-FMK (Fig. 1A). The cell viability was decreased to
verse chemical cores. Several potent compounds were selected 45.6 ± 9.5% in glutamate-treated cells and was recovered by co-
from the chemical library (Kang et al., 2009). UDA showed a mild treatment with compounds to a very small extent. The viability
l-calpain inhibitory activity compared to MDL28170 but was more of glutamate-treated cells was recovered up to 63.6 ± 9.7% by
potent than E64d in cell-free assay (Table 1). MDL28170 has been UDA (Fig. 1B). The treatment of hydrogen peroxide decreased cell
known as one of l-calpain inhibitors categorized into peptidyl viability to 56.4 ± 13.5% compared to control. Only UDA co-treat-
aldehyde derivatives while E64d belongs to the peptidyl epoxide ment, compared to other compounds, recovered the viability of
derivative family (Shirasaki et al., 2006; Wang and Yuen, 1997). H2O2-treated cells significantly and up to 80.0 ± 5.4% (Fig. 1C). In
Most of the known l-calpain inhibitors including peptidyl alde- summary, UDA showed neuroprotective effect against glutamate,
hyde, peptidyl epoxide, diketopiperazine and a-ketoamide deriva- H2O2 and Ab1–42 induced cell death. Among these, the neuroprotec-
tives possess peptide characteristics and highly reactive aldehyde tive effect of UDA against H2O2 was most significant.
or epoxide moieties which cause inadequate selectivity for
l-calpain to other cysteine proteases as well as poor metabolic
stability or/and cell permeability. MDL28170 inhibited cathepsin 3.3. Effect of UDA against Ab-induced cell aggregation, Ab
B and cathepsin L to almost the same extent as l-calpain inhibition oligomerization and fibrillation in vitro
and showed more cathepsin L inhibitory activity over l-calpain
(Table 1). UDA has a totally different structure compared to known Exposure to Ab1–42 led to significant Ab aggregates in SH-SY5Y
l-calpain inhibitors and originates from R. communis L. Among cells called amyloidal plaques. The amyloidal plaques are one of
several potent compounds selected from the chemical library the major pathological events in brains of AD patients (Conway
screening, UDA was of interest and selected for further study. et al., 2003; Selkoe, 1991). Changes in Ab-induced cell aggregation
in SH-SY5Y cells treated with 25 lM Ab1–42 in the absence and
presence of test compounds (20 lM) were monitored and imaged
3.2. Neuroprotective effect of UDA against neurotoxin-induced SH- with a fluorescence inverted microscope (Fig. 2A). Cells treated
SY5Y cell death with only aggregated Ab1–42 showed significant morphological
changes. But there were little or no aggregated forms in cells co-
Glutamate, H2O2, and Ab1–42 are all well known neurotoxins treated with Z-FF-FMK, MDL28170 and UDA, while cells co-treated
which have been previously shown to induce apoptosis in neuronal with CA074, pepstatin A and nimodipine showed severe aggrega-
cell types including SH-SY5Y (Smith et al., 2006; Choi and Roth- tion. We further tested the effect of UDA on aggregation of
man, 1990; Hoyt et al., 1997). Cell viability of SH-SY5Y was mark- Ab1–42 treatment. This test was performed with formation of amy-
edly decreased after exposure to these neurotoxins, including loid fibrils, which can be readily assayed by binding to ThT (Reinke
glutamate and H2O2 (Fig. 1A). Oligomerized Ab1–42 caused apopto- et al., 2009). Synthetic Ab1–42 (25 lM) was incubated in the
sis in SH-SY5Y cells to a lesser extent compared to treatment of absence or presence of either MDL28170, UDA and tannic acid at
glutamate and H2O2, respectively (Fig. 1B and C). Therefore, the final concentrations of 20 or 50 lM at 37 °C for 48 h. UDA inhibited
morphological changes of SH-SY5Y cells were monitored after Ab fibril formation by 46.1 ± 5.3%, although UDA was less potent
treatment with oligomerized Ab1–42 in the absence or presence of than tannic acid which was used as positive control (Ono et al.,
compounds (Fig. 2A). Some synthetic compounds showed good 2004a,b) (Fig. 2B). Ab1–42 has been reported to aggregate to form
l-calpain inhibitory activity from the library, however unlike oligomers and fibrils en route to deposition of amyloid plagues
UDA, they were not studied further due to their high toxicity, (data associated with AD. Since oligomers are more toxic to mouse neu-
not shown). The 72 h treatment of oligomerized Ab1–42 decreased rons than fibrils, we evaluated whether UDA could inhibit Ab olig-
cell viability to 84.5 ± 2.8% versus controls treated with 0.1% omerization using AFM micrographs which capture the differences

Table 1
The inhibitory activity of compounds against l-calpain, cathepsin B, and cathepsin L.

Compound Inhibitory activity as IC50 (lM)a of


l-Calpain Cathepsin B Cathepsin L
b c
On pep1 On pep2
MDL28170 0.0990 ± 0.0002 0.0552 ± 0.0001 0.0958 ± 0.0045 0.0031 ± 0.0003
CA074 >100 >100 0.0037 ± 0.0003 >100
Z-FF-FMK 0.3971 ± 0.0004 1.1100 ± 0.010 0.0656 ± 0.0029 0.0837 ± 0.0008
E64d 62.26 ± 7.73 77.60 ± 1.43 14.39 ± 0.18 2.41 ± 0.09
UDA 5.37 ± 0.71 >100 >100 2.85 ± 0.26
a
Each data point represents mean ± SD from three different experiments performed in triplicate.
b
Calpain inhibitory activity of chemicals on pep1 substrate was evaluated in reaction buffer (50 mM Tris–HCl, 50 mM NaCl, 1 mM EDTA, 5 mM b-mercaptoethanol, pH 7.5)
with 100 lM pep1, 2.5 mM CaCl2, and 5.25 U/ml l-calpain. Thereafter, reaction mixture was incubated with shaking at RT for 30 min. Fluorescence intensities were measured
using 360 nm excitation and 420 nm emission wavelengths. The substrate of pep1 possess the l-calpain-cleavage site in p35 which is [2-Abz]-Ser-Thr-Phe-Ala-Gln-Pro-[3-
nitrotyrosine]-NH2.
c
Calpain inhibitory activity of chemicals on pep2 substrate was assayed as following the method used for that on pep1. The pep2 substrate ([2-Abz]-Glu-Val-Tyr-Gly-Met-
Met-[3-nitrotyrosine]-NH2) was originated from a-spectrin.
E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25 21

Fig. 1. UDA prevented neurotoxin-induced viability loss and apoptosis of SH-SY5Y cells. SH-SY5Y cells were incubated with each neurotoxin including 25 lM pre-
oligomerized Ab1–42 for 72 h (A), 10 mM glutamate with 10 mM glycine for 24 h (B) and 2 mM H2O2 for 24 h (C) in the absence and presence of 20 lM of each compound at
37 °C. Bars represent the mean ± SD from three different experiments performed in triplicate. ⁄p < 0.05, ⁄⁄p < 0.01, significantly different from each neurotoxin-only-treated
group.

between monomers, oligomers and fibrils of Ab1–42 (Ahmed et al., dose-dependent manner in cells (data not shown). Pre-incubation
2010). AFM micrographs of the fibril form of Ab1–42 were distinctly with 20 lM MDL28170 or UDA for 24 h prior to treatment with
different from those of monomers and oligomers as shown in the 400 lM H2O2 prevented the increase in fluorescence generated
upper panel of Fig. 2C. When either UDA or MDL28170 was co- by H2O2 (Fig. 3). UDA reduced the levels of free radicals generated
incubated during the Ab1–42 oligomerization process, as shown in by H2O2 in cells with the maximum effect within 10 min after
bottom panel of Fig. 2C, both compounds inhibited the formation treatment and with a better potency than MDL28170.
of Ab1–42 oligomers. MDL28170 and UDA each inhibited Ab1–42 oli-
gomer formation by 75.31 ± 4.28% and 64.65 ± 3.07%, respectively,
3.5. Effect of UDA on tau hyperphosphorylation and apoptosis induced
which were calculated by using average maximum height obtained
by GOx in SH-SY5Y cells
from XEI program (Fig. 2D). These results suggested that UDA
might be able to inhibit Ab oligomerization as well as fibrillation.
We observed UDA attenuated H2O2-induced apoptosis through
scavenging of free radicals in SH-SY5Y cells. These results necessi-
3.4. Scavenging effect of UDA on oxidative stress-induced free radical tated the evaluation of UDA effects on expression of proteins re-
formation lated to l-calpain activity. When l-calpain is over-activated in
cells, it results in p25 generation and tau hyperphosphorylation,
UDA attenuated H2O2-induced apoptosis effectively in SH-SY5Y ultimately leading to neuronal cell death. SH-SY5Y cells were trea-
cells (Fig. 1C). Therefore, the ability of UDA to directly scavenge ted with glucose oxidase (GOx) which catalyses the oxidation of
H2O2-induced reactive oxygen species (ROS) formation was evalu- glucose to D-glucono-d-lactone with the production of H2O2. GOx
ated to determine a potential mechanism underlying the neuro- induced apoptosis by continual release of H2O2 (Markesbery,
protective effect of UDA. ROS formation was investigated within 1997). GOx at 30 mU/mL elevated l-calpain hyperactivation and
SH-SY5Y cells by monitoring a conversion of DCFH2-DA to DCF. apoptosis related proteins such as p25, hyperphosphorylated tau,
We first observed H2O2-producing free radicals in a rapid and activated caspase and cleaved poly(ADP-ribose)polymerase
22 E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25

(A) (B)

(C) (D)

Fig. 2. UDA reduced the formation of amyloidal plaques in SH-SY5Y cells induced by 25 lM pre-oligomerized Ab1–42 (A) through preventing Ab fibrillation (B) and
oligomerization in vitro (C and D). (A) SH-SY5Y cells were incubated for 72 h with 25 lM pre-oligomerized Ab1–42 in the absence or presence of 20 lM of test compounds. The
amyloidal plaque formed was observed and imaged with a fluorescence inverted microscope at 200 magnification (Axiovert 200, Germany). (B) Ab1–42 aggregation was
induced in the absence or presence of compounds through incubation at 37 °C for 48 h. Then, ThT was added at a final concentration of 10 lM followed by incubation for
15 min at 37 °C. The fluorescence measured at 450 nm (ex) and 490 nm (em) was directly proportional to the amount of Ab1–42 fibril formed. Bars represent means ± SD from
six independent experiments. ⁄p < 0.05, ⁄⁄p < 0.01 vs control group treated with only Ab1–42. (C) AFM micrographs showed the difference between Ab1–42 monomers,
oligomers and fibrils. Ab1–42 was incubated at 20 °C (monomers), at 4 °C for 16 h (oligomers), and at 37 °C for 48 h (fibrils) in PBS. UDA or MDL28170 was co-incubated
during the oligomerization process. Twenty microliters of each aliquot of monomer, oligomer, fibril Ab1–42 sample solution and Ab1–42 oligomerized with co-incubation of
each compound was placed on the mica and dried. Then each sample was scanned by AFM. (D) The inhibition % of oligomerization was calculated as indicated in Methods.
Bars represent means ± SD from three independent experiments. ⁄⁄⁄p < 0.001 vs Ab1–42 oligomer without compounds treated.

(PARP). Caspase-3 is one of the main executioner caspases in the hyperphosphorylation of tau, caspase activation and PARP activa-
apoptotic process. Caspase-3 exists within the cell as an inactive tion with better or equivalent potency in SH-SY5Y cells than
proform called procaspase-3 which undergoes proteolytic cleavage MDL28170. These results led us to develop an assay method to
to be an active protein. The caspase-3 activation can be also veri- evaluate l-calpain inhibitory activity of test compound in cells
fied by the proteolytic cleavage of PARP, an endogenous caspase- which might predict directly l-calpain-mediated biological activ-
3 substrate (Tenn and Wang, 2007). The treatment of 30 mU/mL ity. We synthesized the CAPN1 gene encoding the large subunit
GOx resulted in an increase in levels of p25, phospho-tau, cleaved of l-calpain, which is responsible for catalytic proteolysis. Then,
PARP, and cleaved caspase-3 compared to control values (second it was transfected into HEK293T and SH-SY5Y cells. A transfection
lane in Fig. 4), but UDA treatment (20 lM) remarkably decreased time of 8 h showed the maximum degree of l-calpain protein
these levels as shown in Fig. 4. On the other hand, the treatment expression (Fig. 5A) in both cell types. Cells were treated with
of 20 lM MDL28170 did not decrease these levels as much as UDA. the substrate, 10 lM pep1, together with 1 lM ionomycin in the
absence or presence of MDL28170 and UDA after the 8 h transfec-
3.6. l-Calpain inhibitory activity of UDA in cells tion. Ionomycin, a calcium ionophore, enhances calcium influx into
the cell (Morgan and Jacob, 1994), which then activates l-calpain.
UDA inhibited l-calpain activity with much less potency than The cells treated with pep1 and co-treated with 1 lM ionomycin
MDL28170 in a cell free in vitro experiment (Table 1). However, showed higher fluorescence intensity compared to pep1 treated
UDA reduced l-calpain-mediated p35 cleavage to p25, cells. This was due to increases in pep1 cleavage induced by
E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25 23

ture of UDA is much different from known calpain inhibitors and


the function of UDA regarding neuroprotective effect has not yet
been reported so far (Kang et al., 2009; Shirasaki et al., 2006; Wang
and Yuen, 1997; Cuerrier et al., 2007). Pep1 substrate is derived
from p35, a CDK5 activity regulator and pep2 originates from spec-
trin. The IC50 of UDA on l-calpain against pep1 was 5.37 ± 0.71 lM,
while that against pep2 was more than 100 lM. Further, other
known l-calpain inhibitors, MDL28170 and E64d, showed no
substrate specificity (Table 1). Although UDA has much less
activity than MDL28170 (0.0990 ± 0.0002 lM for pep1 and
0.0552 ± 0.0001 lM for pep2), we suggest UDA could be a novel
potent l-calpain inhibitor with influential activity in vivo due to
following reasons. Although E64d in vitro activity against calpains
and cathepsins is poorer than other known inhibitors including
MDL28170, E64 and E64c (Lampi et al., 1992; Azuma et al., 1992;
Fig. 3. UDA prevented oxidative stress-induced free radical formation in SH-SY5Y Tsubokawa et al., 2006), E64d has been extensively utilized in
cells. SH-SY5Y cells were loaded with 5 lM DCFH2-DA for 30 min at 37 °C and then in vivo experiments because of its enhanced cell permeability,
exposed to 400 lM of H2O2 for 30 min. Before H2O2 treatment, cells were pre- leading to good in vivo efficacy. In our in vitro experiments, UDA
incubated for 24 h with 20 lM MDL28170 and UDA. Values are the means ± SD of
three independent experiments.
showed much better l-calpain inhibitory activity than E64d
(62.26 ± 7.73 lM for pep1 and 77.60 ± 1.43 lM for pep2). Addi-
tionally, UDA is much less toxic; the LD50 of UDA after oral treat-
ment for mice is 8150 mg/kg (Newell et al., 1949). UDA also
showed competent neuroprotective effects against glutamate-,
H2O2- and Ab1–42-induced cell death in SH-SY5Y cells (Fig. 1).
Therefore, UDA was further evaluated in this study for its potential
use as an AD therapeutic. The key neuropathological features of AD
are the abnormal deposition of Ab1–42-mediated plaques and re-
lated insoluble peptides in extracellular spaces in brain. The neuro-
toxicity exerted by aggregated Ab may be mediated by several
mechanisms, such as the generation of ROS, escalation of intracel-
lular Ca2+ concentrations and induction of apoptosis (Selkoe, 1991;
Camins et al., 2006; Esteban, 2004). Another distinct biomarker of
AD is intracellular neurofibril tangles which are induced by abnor-
mal tau hyperphosphorylation. Alteration in calcium homeostasis
generated by insoluble Ab leads to persistent hyperactivation of
calpains. Hyperactivated l-calpain cleaves a number of neuronal
substrates including p35. The cleavage of p35 to p25 produces a
more stable CDK5-p25 complex. This leads to prolonged CDK5 acti-
vation and mislocation of CDK5, which results in tau hyperphosph-
orylation, neurofibril tangles and ultimately neuronal cell death
(Saito et al., 1993; Patrick et al., 1999). UDA attenuated Ab-medi-
ated cell death in SH-SY5Y cells with equipotency compared to
MDL28170 (Fig. 1A and Fig. 2A). Barry et al. demonstrated that
Z-FF-FMK, known as a cathepsin L inhibitor, could block
Fig. 4. UDA has a neuroprotective effect against a continuous oxidative stress. UDA Ab-mediated induction of the apoptotic cascade (Bloland and
down regulated the conversion of p35 to p25 leading to a decrease in tau Campbell, 2004). The inhibitory activity of UDA against cathepsin
phosphorylation. The elevated levels of both PARP and caspase-3 by GOx treatment
L (IC50 value of 2.85 ± 0.26 lM) was much weaker than that of
in SH-SY5Y cells showed decreases in the active forms when UDA was treated. SH-
SY5Y cells were treated with GOx (30 mU/mL) in the absence or presence of 20 lM Z-FF-FMK (0.0837 ± 0.0008 lM) in vitro, while UDA protected
MDL28170 and UDA for 4 h at 37 °C in a humidified atmosphere with 5% CO2. The Ab-mediated cell death with almost equipotency to Z-FF-FMK. In
cells were lysed and the soluble protein was analyzed by Western blot. addition, UDA could inhibit Ab fibril formation by 46.1 ± 5.3%
which was determined in the ThT assay (Fig. 2B). Since the ThT as-
l-calpain activation when calcium influx occurred. The saturation say cannot differentiate Ab oligomers from Ab fibrils, we estimated
on fluorescence intensity was observed after about 80 min when the effect of UDA on the formation of oligomers using AFM analy-
signals were measured in kinetic mode (Fig. 5B). The effect of sis. AFM micrographs of UDA treatment during Ab oligomerization
MDL28170 and UDA on l-calpain activity in HEK293T (Fig. 5C) (oligomer vs UDA micrographs in Fig. 2C) showed no fibril shape
and SH-SY5Y cells (Fig. 5D) is shown using the value at 80 min. and the size of oligomers was lessened. It can be suggested that
UDA inhibited l-calpain with better potency than MDL28170 in UDA blocked the oligomerization process. It was recently reported
HEK293T cells and with equivalent potency in SH-SY5Y cells. that the Ab oligomer was more toxic than Ab fibrils and Ab fibril
formation was reported as a self-detoxification mechanism (Sakon-
4. Discussion o and Zako, 2010; Walsh et al., 2000; Wang et al., 2008). In this
sense, it is worthy to note that UDA is able to inhibit Ab oligomer-
We have screened a chemical library made up of 6480 com- ization. It has been known that insoluble Ab1–42 generates ROS
pounds which consisted of both synthetic and natural products and induces cell membrane damage, which leads to calcium
possessing diverse pharmacophores. Using the screening method influx (Selkoe, 1991; Camins et al., 2006; Walsh et al., 2000).
developed previously in our laboratory (Kang et al., 2009), UDA Therefore, the ability of UDA to alleviate Ab oligomerization and
was found to be a substrate-specific l-calpain inhibitor. The struc- fibrillation might affect intracellular Ca2+ levels. Among the tested
24 E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25

(A) (B)

(C) (D)

Fig. 5. UDA reduced l-calpain activity in cells with better potency than MDL28170. (A) HEK293T and SH-SY5Y cells were transfected with 3 lg of CAPN1 encoded with
pcDNA3.1/His A for diverse time periods and then the extent of l-calpain expression was determined using Western blot. (B) Cells transfected with pcDNA 3.1/His A_CAPN1
for 8 h were harvested and resuspended in calpain reaction buffer containing 0.1% triton X-100. The cells were incubated with pep1 and ionomycin in the absence or presence
of either MDL28170 or UDA. The relative change in fluorescence induced by l-calpain-mediated pep1 cleavage was measured in kinetic mode using a microplate fluorescence
reader (SpectraMAX GEMINI EM, Molecular Devices) at 37 °C. Fluorescence was measured at 320 nm (ex) and 420 nm (em). (C),(D) Fluorescence obtained at an endpoint of
80 min from (B) were depicted in bar graphs in HEK293T and SH-SY5Y cells, respectively. Values are the means ± SD of three independent experiments performed in
triplicate. ns, not significant; ⁄⁄p < 0.01, ⁄⁄⁄p < 0.001 vs ionomycin-treated group only.

compounds, UDA also showed the most potent neuroprotective ef- 5. Conclusion
fect against H2O2-mediated toxicity in SH-SY5Y cells (Fig. 1C).
These results can be explained by a ROS scavenging effect of A multitude of neurodegenerative cellular events induced by l-
UDA. UDA inhibited H2O2-generated ROS by measuring the calpain hyperactivation leads to caspase-3 protease activity that
DCFH2-DA conversion to DCF with a better potency than controls programmed cell death (Tenn and Wang, 2007; Eliasson
MDL28170 (Fig. 3). From the Western blots of SH-SY5Y cells, it et al., 1997). Thus, UDA is likely able to slow the progression of
was confirmed that l-calpain inhibitory activity of UDA reduced AD by inhibiting l-calpain and the many pathways such as insol-
the cleavage of p35 to p25, decreased tau hyperphosphorylation, uble Ab oligomerization and fibrillation, Ab-mediated ROS forma-
and ultimately inhibited the activation of PARP and caspase-3 in tion, neurofibril tangles formation and cell death. The treatment
GOx-treated SH-SY5Y cell (Fig. 4). of combination of Docosahexaenoic acid (a type of omega-3 fatty
Most of the peptide-like l-calpain inhibitors showed no speci- acid), uridine, and choline to aged gerbils showed significantly bet-
ficity to l-calpain over other cysteine proteases. Other problems ter performance on learning and memory tests than untreated ger-
of these l-calpain inhibitors are poor membrane permeability bil group (Holguin et al., 2008). Although further in vivo studies are
and poor metabolic stability (Kang et al., 2009; Shirasaki et al., necessary to demonstrate the potential of UDA as an adjuvant ther-
2006; Wang and Yuen, 1997; Cuerrier et al., 2007). l-Calpain apeutic for AD, the current study may pave the way to develop
(Calpain 1) exists as a heterodimer consisting of a large 80 kDa cat- non-peptide-like novel l-calpain inhibitors which can reverse
alytic subunit encoded by CAPN1 and a common 29 kDa regulatory the levels of insoluble Ab- and hyperphosphorylated tau-induced
subunit encoded by CAPN4 (Aoki et al., 1986; Ohno et al., 1986). apoptotic proteins.
Upon exposure to Ca2+, l-calpain is activated by subunit autolysis
by separation from the regulatory subunit (Goll et al., 1992). UDA
reduced l-calpain activity with better potency than MDL28170 in Acknowledgements
SH-SY5Y and HEK293T cells when the catalytic subunit of l-cal-
pain was overexpressed (Fig. 5). UDA could be a distinctive potent This research was supported by Basic Science Research Program
l-calpain inhibitor possessing good cell permeability, non-toxicity through the National Research Foundation of Korea (NRF) funded by
and structural stability. the Ministry of Education, Science and Technology (2011-0011007)
E. Lee et al. / European Journal of Pharmaceutical Sciences 46 (2012) 17–25 25

and by the Ewha Global Top5 Grant 2011 of Ewha Womans Kim, W., Kim, Y., Min, J., Kim, D.J., Chang, Y.T., Hecht, M.H., 2006. A high-throughput
screen for compounds that inhibit aggregation of the Alzheimer’s peptide.
University. Jun K.Y. was partially supported by Ewha Womans
Chem. Biol. 1, 461–469.
University. Lampi, K.J., Kadoya, K., Azuma, M., David, L.L., Shearer, T.R., 1992. Comparison of
cell-permeable calpain inhibitors and E64 in reduction of cataract in cultured
rat lenses. Toxicol. Appl. Pharmacol. 117, 53–57.
Lee, M.S., Kwon, Y.T., Li, M., Peng, J., Friedlander, R.M., Tsai, L.H., 2000. Neurotoxicity
References induces cleavage of p35 to p25 by calpain. Nature 405, 360–364.
Markesbery, W.R., 1997. Oxidative stress hypothesis in Alzheimer’s disease. Free
Rad. Biol. Med. 23, 134–147.
Ahmed, M., Davis, J., Aucoin, D., Sato, T., Ahuja, S., Aimoto, S., Elliott, J.I., Van Mellgren, R.L., 1990. Interaction of human erythrocyte multicatalytic proteinase
Nostrand, W.E., Smith, S.O., 2010. Structural conversion of neurotoxic amyloid- with polycations. Biochim. Biophys. Acta 1040, 28–34.
beta(1–42) oligomers to fibrils. Nat. Struct. Mol. Biol. 17, 561–567. Morgan, A.J., Jacob, R., 1994. Ionomycin enhances Ca2+ influx by stimulating store-
Aoki, K., Imajoh, S., Ohno, S., Emori, Y., Koike, M., Kosaki, G., Suzuki, K., 1986. regulated cation entry and not by a direct action at the plasma membrane.
Complete amino acid sequence of the large subunit of the low-Ca2+-requiring Biochem. J. 300, 665–672.
form of human Ca2+-activated neutral protease (lCANP) deduced from its cDNA Newell, G.W., Petretti, A.K., Reiner, L., 1949. Studies of the acute and chronic toxicity
sequence. FEBS Lett. 205, 312–317. of undecylenicacid. J. Invest. Dermatol. 13, 145–149.
Azuma, M., David, L.L., Shearer, T.R., 1992. Superior prevention of calcium ionophore Nikolic, M., Dudek, H., Kwon, Y.T., Ramos, Y.F., Tsai, L.H., 1996. The CDK5/p35 kinase
cataract by E64d. Biochim. Biophys. Acta 1180, 215–220. is essential for neurite outgrowth during neuronal differentiation. Gene Dev. 10,
Bloland, B., Campbell, V., 2004. Abeta-mediated activation of the apoptotic cascade 816–825.
in cultured cortical neurons: a role for cathepsin-L. Neurobiol. Aging 25, 83–91. Noble, W., Olm, V., Takata, K., Casey, E., Mary, O., Meyerson, J., Gaynor, K.,
Buxbaum, J.D., Liu, K.N., Luo, Y., Slack, J.L., Stocking, K.L., Peschon, J.J., Johnson, R.S., LaFrancois, J., Wang, L., Kondo, T., Davies, P., Burns, M., 2003. Veeranna, Nixon R,
Castner, B.J., Cerretti, D.P., Black, R.A., 1998. Evidence that tumor necrosis factor Dickson D, Matsuoka Y, Ahlijanian M, Lau LF, Duff K. CDK5 is a key factor in tau
alpha converting enzyme is involved in regulated alpha-secretase cleavage of aggregation and tangle formation in vivo. Neuron 38, 555–565.
the Alzheimer amyloid protein precursor. J. Biol. Chem. 273, 27765–27767. Ohno, S., Emori, Y., Suzuki, K., 1986. Nucleotide sequence of a cDNA coding for the
Camins, A., Verdaguer, E., Folch, J., Pallàs, M., 2006. Involvement of calpain small subunit of human calcium-dependent protease. Nucleic Acids Res. 14,
activation in neurodegenerative processes. CNS Drug Rev. 12, 135–148. 5559.
Chae, T., Kwon, Y.T., Bronson, R., Dikkes, P., Li, E., Tsai, L.H., 1997. Mice lacking p35, a Ono, K., Hasegawa, K., Naiki, H., Yamada, M., 2004a. Anti-amyloidogenic activity of
neuronal specific activator of Cdk5, display cortical lamination defects, seizures, tannic acid and its activity to destabilize Alzheimer’s b-amyloid fibril in vitro.
and adult lethality. Neuron 18, 29–42. Biochim. Biophys. Acta 1690, 193–202.
Choi, D.W., Rothman, S.M., 1990. The role of glutamate neurotoxicity in hypoxic- Ono, K., Hasegawa, K., Naiki, H., Yamada, M., 2004b. Antiamyloidogenic activity of
ischemic neuronal death. Annu. Rev. Neurosci. 13, 171–182. tannic acid and its activity to destabilize Alzheimer’s b-amyloid fibrils in vitro.
Conway, K.A., Baxter, E.W., Felsenstein, K.M., Reits, A.B., 2003. Emerging beta- Biochim. Biophys. Acta 1690, 193–202.
amyloid therapies for the treatment of Alzheimer’s disease. Curr. Pharm. Des. 9, Patrick, G.N., Zukerberg, L., Nikolic, M., de la Monte, S., Dikkes, P., Tsai, L.H., 1999.
427–447. Conversion of p35 to p25 deregulates CDK5 activity and promotes
Cruz, J.C., Tseng, H.C., Goldman, J.A., Shih, H., Tsai, L.H., 2003. Aberrant CDK5 neurodegeneration. Nature 402, 615–622.
activation by p25 triggers pathological events leading to neurodegeneration Reinke, A.A., She, H.Y., Gestwicki, J.E., 2009. A chemical screening approach reveals
and neurofibrillary tangles. Neuron 30, 471–483. that indole fluorescence is quenched by pre-fibrillar but not fibrillar amyloid-
Cuerrier, D., Moldoveanu, T., Campbell, R.L., Yoruk, B., Verhelst, S.H., Greenbaum, D., beta. Bioorg. Med. Chem. Lett. 19, 4952–4957.
Bogyo, M., Davies, P.L., 2007. Development of calpain-specific inactivator by Saido, T.C., Shibata, M., Takenawa, T., Murofushi, H., Suzuki, K., 1992. Positive
screening of positional-screening epoxide libraries. J. Biol. Chem. 282, 9600– regulation of mu-calpain action by polyphosphoinositides. J. Biol. Chem. 267,
9611. 24585–24590.
Dahlgren, K.N., Manelli, A.M., Stine Jr., W.B., Baker, L.K., Krafft, G.A., LaDu, M.J., 2002. Saito, K., Elce, J.S., Hamos, J.E., Nixon, R.A., 1993. Widespread activation of calcium-
Oligomeric and fibrillar species of amyloid-b peptides differentially affect activated neutral proteinase (calpain) in the brain in Alzheimer disease: a
neuronal viability. J. Biol. Chem. 277, 32046–32053. potential molecular basis for neuronal degeneration. Proc. Natl. Acad. Sci. USA
Demattos, R.B., Bales, K.R., Cummins, D.J., Dodart, J.C., Paul, S.M., Holtzman, D.M., 90, 2628–2632.
2001. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance Sakono, M., Zako, T., 2010. Amyloid oligomers: formation and toxicity of Ab
and decreases brain A beta burden in a mouse model of Alzheimer’s disease. oligomers. FEBS J. 277, 1348–1358.
Proc. Natl. Acad. Sci. USA 98, 8850–8855. Salamino, F., De Tullio, R., Mengotti, P., Viotti, P.L., Melloni, E., Pontremoli, S., 1993.
Eliasson, M.J., Sampei, K., Mandir, A.S., Hurn, P.D., Traystman, R.J., Bao, J., Pieper, A., Site-directed activation of calpain is promoted by a membrane-associated
Wang, Z.Q., Dawson, T.M., Snyder, S.H., Dawson, V.L., 1997. Poly(ADP-ribose) natural activator protein. Biochem. J. 290, 191–197.
polymerase gene disruption renders mice resistant to cerebral ischemia. Nat. Selkoe, D.J., 1991. The molecular pathology of Alzheimer’s disease. Neuron 6, 487–
Med. 3, 1089–1095. 498.
Esteban, J.A., 2004. Living with the enemy: a physiological role for the bata-amyloid Shapiro, A.L., Rothaman, S., 1945. Undecylenic acid in the treatment of
peptide. Trends Neurosci. 27, 1–3. dermatomycoses. Arch. Dermatol. Syphilol. 52, 166–171.
Fu, W., Luo, H., Parthasarathy, S., 1998. Catecholamines potentiate amyloid b- Shirasaki, Y., Nakamura, M., Yamaguchi, M., Miyashita, H., Sakai, O., Inoue, J., 2006.
peptide neurotoxicity: involvement of oxidative stress, mitochondrial Exploration of orally available calpain inhibitors 2: peptidylhemiacetal
dysfunction, and perturbed calcium homeostasis. Neurobiol. Dis. 5, 229–243. derivatives. J. Med. Chem. 49, 3926–3932.
Goll, D.E., Thompson, V.F., Taylor, R.G., Zalewska, T., 1992. Is calpain activity Smith, W.W., Gorospe, M., Kusiak, J.W., 2006. Signaling mechanisms underlying
regulated by membrane and autolysis or by calcium and calpastatin? BioEssays Abeta toxicity: potential therapeutic targets for Alzheimer’s disease. CNS
14, 549–556. Neurol. Disord. Drug Targets 5, 355–361.
Hashiguchi, M., Saito, T., Hisanaga, S., Hashiguchi, T., 2002. Truncation of CDK5 Tenn, C.C., Wang, Y., 2007. VX-induced cell death involves activation of caspase-3 in
activator p35 induces intensive phosphorylation of Ser202/Thr205 of human cultured rat cortical neurons. Neurosci. Lett. 417, 155–159.
tau. J. Biol. Chem. 277, 44525–44530. Tsubokawa, T., Solaroglu, I., Yatsushige, H., Cahill, J., Yata, K., Zhang, J.H., 2006.
Holguin, S., Martinez, J., Chow, C., Wurtman, R., 2008. Dietary uridine enhances the Cathepsin and calpain inhibitor E64d attenuates matrix metalloproteinase-9
improvement in learning and memory produced by administering DHA to activity after focal cerebral ischemia in rats. Stroke 37, 1888–1894.
gerbils. FASEB J. 22, 3938–3946. Walsh, D.M., Klyubin, I., Fadeeva, J.V., Cullen, W.K., Anwyl, R., Wolfe, M.S., Rowan,
Hoyt, K.R., Gallagher, A.J., Hastings, T.G., Reynolds, I.J., 1997. Characterization of M.J., Selkoe, D.J., 2000. Naturally secreted oligomers of amyloid beta protein
hydrogen peroxide toxicity in cultured rat forebrain neurons. Neurochem. Res. potently inhibit hippocampal long-term potentiation in vivo. Nature 416, 535–
22, 333–340. 539.
Kang, D.H., Jun, K.Y., Lee, J.P., Pak, C.S., Na, Y., Kwon, Y., 2009. Identification of 3- Wang, K.K., Yuen, P.W., 1997. Development and therapeutic potential of calpain
acetyl-2-aminoquinolin-4-one as a novel, nonpeptidic scaffold for specific inhibitors. Adv. Pharmacol. 37, 117–1152.
calpain inhibitory activity. J. Med. Chem. 52, 3093–3097. Wang, L., Maji, S.K., Sawaya, M.R., Eisenberg, D., Riek, R., 2008. Bacterial inclusion
Kang, J., Lemaire, H.G., Unterbeck, A., Salbaum, J.M., Masters, C.L., Grzeschik, K.H., bodies contain amyloid-like structure. PLoS Biol. 6, e195.
Multhaup, G., Beyreuther, K., Müller-Hill, B., 1987. The precursor of Alzheimer’s Younkin, S.G., 1998. The role of A beta 42 in Alzheimer’s disease. J. Physiol. Paris 92,
disease amyloid A4 protein resembles a cell-surface receptor. Nature 325, 733– 289–292.
736.

Potrebbero piacerti anche