You are on page 1of 25

Arch. Pharm. Res.

DOI 10.1007/s12272-015-0587-6

RESEARCH AR TIC L E

Anti-inflammatory, antioxidant and anti-acetylcholinesterase


activities of Bouvardia ternifolia: potential implications
in Alzheimer’s disease
Giovanni Garc´ıa-Morales • Maira Huerta-Reyes • Manase´ s Gonza´ lez-Cortazar •

Alejandro Zamilpa • Enrique Jime´ nez-Ferrer • Rau´ l Silva-Garc´ıa •


Rube´n Roma´ n-Ramos • Arturo Aguilar-Rojas

Received: 5 August 2014 / Accepted: 27 February 2015


The Pharmaceutical Society of Korea 2015

Abstract Bouvardia ternifolia has been used medicinally neuroprotective effect against b-amyloid peptide
to treat inflammation. In the present study, we investigate (83.97 ± 5.03 %). Phytochemical analysis, showed the
the anti-Alzheimer’s potential effect of the hydroalcoholic presence of 3-O-quercetin glucopyranoside (415 mg/g),
extract of B. ternifolia through evaluation of anti-inflam- rutin (229.9 mg/g), ursolic and oleanolic acid (54 and
matory and antioxidant activities, quantification of the 20.8 mg/g respectively), 3-O-quercetin rhamnopyranoside
percentage inhibition of acetylcholinesterase activity, pro- (12.8 mg/g), chlorogenic acid (9.5 mg/g), and scopoletin
tection effect against b-amyloid fibrillar-induce neuro- (1.38 mg/g). Our findings support the use of B. ternifolia
toxicity, and the identification of the main constituents. Our since the extract induced significant neuroprotection against
results show that B. ternifolia extract and ethyl acetate b-amyloid peptide, anti-inflammatory, antioxidant and anti-
fraction induced anti-inflammatory effects by reducing in- acetylcholinesterase effects that could be attributed to its
flammation by [70 %, while antioxidant test revealed contents of polyphenols, coumarins, and triterpenes, and
significant IC50 values for flavonoid content fraction encourage further studies for development of this extract as
(30.67 ± 2.09 lg/ml) and ethyl acetate fraction (42.66 ± therapeutic agent in treatment of Alzheimer’s disease.
0.93 lg/ml). The maximum inhibition of acetyl-
cholinesterase was exhibited by scopoletin content fraction Keywords Bouvardia ternifolia Alzheimer’s disease
(38.43 ± 3.94 %), while ethyl acetate fraction exerted Polyphenols Cumarins Triterpens

G. Garc´ıa-Morales M. Huerta-Reyes R. Roma´n-Ramos


M. Gonza´lez-Cortazar A. Zamilpa Departamento de Ciencias de la Salud, DCBS, UAM-Iztapalapa,
E. Jime´nez-Ferrer A. Aguilar-Rojas Mexico, DF, Mexico
Centro de Investigacio´ n Biome´dica del Sur, Instituto
Mexicano del Seguro Social (IMSS), Xochitepec, Morelos, A. Aguilar-Rojas (&)
Mexico Unidad de Investigacio´ n Me´dica en Medicina Reproductiva,
Unidad Me´dica de Alta Especialidad (UMAE) en Ginecolog´ıa
G. Garc´ıa-Morales R. Roma´n-Ramos Obstetricia No. 4 ‘‘Dr. Luis Castelazo Ayala’’, IMSS, Av. R´ıo
Divisio´ n de Ciencias Biolo´ gicas y de la Salud Magdalena No. 289, Col. Tizapa´n San A´ ngel, Delegacio´ n A´
(DCBS), Universidad Auto´ noma Metropolitana- lvaro
Iztapalapa (UAM- Obrego´ n, 01090 Mexico, DF, Mexico
Iztapalapa), Mexico, DF, Mexico e-mail: arturolation@yahoo.com

M. Huerta-Reyes
Unidad de Investigacio´ n Me´dica en Farmacolog´ıa, Hospital
de Especialidades, Centro Me´dico Nacional Siglo XXI
(CMN- SXXI), IMSS, Mexico, DF, Mexico

R. Silva-Garc´ıa
Unidad de Investigacio´ n Me´dica en Inmunolog´ıa, Hospital
de
Pediatr´ıa, CMN-SXXI, IMSS, Mexico, DF, Mexico 123
123
Anti-Alzheimer’s diseases effects of B. ternifolia
G. Garc´ıa-Morales et
al.
Introduction O-Tetradecanoyl

The plant species Bouvardia ternifolia (Cav.) Schldtl.


(Rubiaceae) is distributed in Mesoamerica, Mexico, and
the United States. B. ternifolia is popularly known as
‘‘trompetilla’’ and has been used medicinally to treat
in-
toxication produced by stings from snake bites, scorpion
bites, or bee stings, as well as headache and inflammation
(Argueta-Villamar et al. 1994). The pharmacological ac-
tivities of the B. ternifolia extract as an antitumoral (Jolad
et al. 1977), cytotoxic (Tobey et al. 1978) and as antivenom
in scorpion poisoning (Jimenez-Ferrer et al. 2005) have
been described. On the other hand, preliminary screening
studies also revealed that the hydroalcoholic extract of B.
ternifolia exhibited competitive inhibition of the enzyme
acetylcholinesterase, a key molecule for treatment of Alz-
heimer’s disease (AD) (Herrera-Ruiz et al. 2012).
AD is a progressive disease leading to the degeneration
of cholinergic neurons, loss of cholinergic neurotransmis-
sion, and the formation of abnormal aggregates of b-
amyloid (Ab) protein fragments throughout the brain
(Senol et al. 2011). Although neuronal loss is the main
neuropathologic feature underlying the symptoms of AD,
the accumulation of Ab may initiate the inflammatory
process (Lee et al. 2010), as well as the production of
reactive oxygen species (ROS) and oxidative injury
(Wollen 2010). In conjunction, all of these aspects promote
profound neural damage and the typical symptomatology
of AD (Francis et al. 1999; Weiner et al. 2012).
At present, there is no cure for AD. The few agents ap-
proved by the U.S. Federal Drug Administration (FDA) for
the treatment of AD have only modest effectiveness in terms
of modifying clinical symptoms, and none appear to affect
disease progression or prevention (Pasinetti 2012). In
addition to their low therapeutic effectiveness, these drugs
can cause serious side effects (Dunn et al. 2000; Alfirevic
et al. 2007).
Because AD is a genetically complex, slowly
progressive, and irreversible neurodegenerative disease of
the brain, it has been considered that a multitarget approach
may provide more therapeutic value (Wollen 2010; Hampel
et al. 2011). Because the capacity for inhibiting
acetylcholinesterase, as well as its traditional uses that
suggest the possible ability to modulate some neurological
diseases, we extended these observations to further
investigate the anti-inflammatory and antioxidant activities
of B. ternifolia, as well as to quantify the percentage
inhibition of acetylcholinesterase, evaluate the protective
ef- fects against Ab-induced neurotoxicity, and to identify
the main constituents in the extract. For this purpose, the
an- tioxidant activity of the hydroalcoholic extract of B.
ternifolia, as well as several fractions derived from it, was
determined by the inhibition of thiobarbituric acid reactive
substances (TBARS) formation in rat brain homogenate,
while anti-in- flammatory activity was measured by 12-
Anti-Alzheimer’s diseases effects of B. ternifolia
phorbol-13-acetate (TPA)-induced ear edema assay in
mice. The percentage of inhibition on the
acetylcholinesterase was determined by the Ellman’s assay
while the protective effect against Ab-induced
neurotoxicity was evaluated by neural viability in SH-
SY5Y cell determined by XTT. HPLC and other
chromatographic methods were employed for chemical
characterization of the active extract.

Materials and methods

Plant material and preparation of the extract

The aerial parts of B. ternifolia (Cav.) Schltdl. were col-


lected in the state of Morelos, Mexico (Latitude:
19 010 37.3800 N, Longitude: 99 120 38.7600 WE). No
specific
permissions were required for this location and the field
studies no involve endangered or protected species. Abigail
Aguilar, M.Sc., curator of the Mexican Institute of Social
Security Medicinal Herbarium (IMSSM), identified the
botanical material employed. A voucher specimen was
deposited at the IMSSM with number 15,649. The dried
and powdered plant material (1700 g) was extracted by
maceration with 7.5 L of an ethanol solution (60 %) per kg
of plant material for 24 h (93) at room temperature. After
filtration, the solvent was removed under reduced pressure
distillation (Heidolph Instruments). A semisolid extract
(BtHA) (278.2 g) was finally lyophilized and stored at 4 C
in centrifuge tubes until needed.

Evaluation of anti-inflammatory activity

Animal experiments were carried out in strict accordance


with the Mexican Regulations of Experimental Animal
Care (NOM-062-ZOO-1999). The protocol was approved
by the Institutional Research Committee and the Ethics
Committee of the Mexican Institute of the Social Security
(IMSS) (Registry number R-2010-1701-57). Animals were
anesthetized with veterinary pentobarbital sodium, and all
efforts were made to minimize suffering. Animals were
maintained at 23 ± 2 C on 12/12 h light–dark cycles with
free access to food and water.
The assay of 12-O-Tetradecanoylphorbol-13-acetate
(TPA)-induced ear edema in mice was performed as previ-
ously reported (De Young et al. 1989; Recio et al. 1994).
Prior to the assays, the anti-inflammatory reaction progress
curve of BtHA extract was analyzed at increasing concen-
trations. This data shown that 6.5 mg of this were the
highest amount of extract able to inhibit edematous
response. All the curves of anti-inflammatory activity were
performed at this dose. The following conditions were used
for the present experiment. Male ICR-strain mice weighing
28–30 g (Har- lan, S.A. de C.V.) were anesthetized
with veterinary
G. Garc´ıa-Morales et
al.
Anti-Alzheimer’s diseases effects of B. ternifolia
pentobarbital sodium (60 mg/ml) (Pfizer, Inc.). To promote solution, pH 8.0, 333 mM 5,50 -dithiobis(2-nitrobenzoics)
inflammation, 2.5 lg TPA in ethanol (Sigma-Aldrich Che- acid (DTNB) and 0.0125 units/ml of enzyme. One unit (U)
mical Co.) was applied to both sides of the mouse’s right of enzymatic activity was defined as the amount of rat
ear. The left ear received ethanol and was used as brain homogenate (ll) and increasing 0.001 absorbance at
reference. After 412 nm in this condition (Chen and Kubo 2002). The
15 min of TPA treatment, 6.5 mg of the test fractions or reactions were measured on the spectrophotometer Lambda
80 lg of Dexamethasone in acetone (Sigma) as anti-in- 25 (Perkin- Elmer) at 412 nm for 3 min every 20 s.
flammatory (positive control) were applied to both sides of Samples in absences of inhibitor as negative control or in
the right ear. The control group received only ethanol. presences of 0.01 mM Tacrine (9-Amino-1,2,3,4-
Treatments and control were administered at 10 ll of final tetrahydroacridine hydrochlo- ridehydrate) (Sigma) as
volume. 4 h later, the animals were sacrificed by cervical positive control, were employed.
dislocation and a plug was removed from each ear. The Prior to the study of anticholinesterase activity, the
edematous response was measured as the difference in Michaelis constant (Km) and maximum velocity (Vm)
weight between the two plugs after treatment. Percentage values were determined. For this proposes, increasing
inhibition of edema was calculated with the following concentrations of acetylcholine iodide were added and data
equation: % = [(edema A - edema B)/edema A] 9 were analyzed by non-linear regression analysis. The anti-
100. Edema A = edema induced by TPA alone, and acetylcholinesterase activity of the BtHA was evaluated at
edema B = edema induced by TPA plus sample. increasing concentrations of this extract. Data from con-
centration-inhibition experiments of BtHA, were calculated
Evaluation of antioxidant activity by non-linear regression analysis and allowed us to cal-
culate the Half maximal Inhibitory concentrations (IC50).
Antioxidant activity of the B. ternifolia fractions was as- The rest of the inhibition curves were carried out em-
sayed by lipid peroxidation of brain homogenates and was ploying the IC50 data calculated (0.04 mg/ml of extract and
quantified by the contents of TBARS (Ohkawa et al. 1979; fractions) as well as the Km value of acetylcholine iodide
Ng et al. 2000). Quercetin and a-Tocopherol compounds at (1.86 mM) as substrate.
1 lg/ml were utilized as references and an inhibitor-free All the assays were carried out in triplicate and per-
sample was employed as control. The antioxidant activity formed at least four times in independent experiments. The
of the B. ternifolia extract and fractions was evaluated at 10 results were reported as percentage of inhibition following
and 100 lg/ml as previously was shown (Gonza´ lez et the equation: % Inhibition = 100 (1 - Vm1/Vm2), where
al. Vm1 was the maximum velocity values of control group
2009). As oxidative stress promoter, 10 lM of FeSO4 and Vm2, the maximum velocity values of test group.
(Sigma) were added to each sample. Concentration of
TBARS was calculated by interpolation in a standard curve Evaluation of protective effect against Ab-induced
of tetramethoxypropane (TMP) as previously reported neurotoxicity
(Esterbauer and Cheeseman 1990). Final results were ex-
pressed as nmol of TBARS per mg of protein. Increasing Preparation of the peptide
concentrations (10–1000 lg/ml) of BtHA, BtAcOEt,
BtC1N, and Flav3 were employed in displacement curves The Ab1–42 peptide (Sigma) was dissolved in
of the lipid peroxidation assay. Quercetin and a-Toco- hexafluoroiso- propanol (Sigma) to 1 mg/ml and incubated
pherol (0–100 lg/ml) were used as references (Gonza at room tem- perature for 24 h. The hexafluoroisopropanol
´lez et al. 2009). The inhibition ratio (%) was calculated was eliminated by vacuum in a Speed Vac and the resulting
fol- lowing the formula: Inhibition ratio (%) = (C - unaggregated Ab1–42 film, was stored at -20 C until use.
E)/ C 9 100 %, where C was the absorbance of control The Ab1–42 ag- gregations to fibrillar forms were prepared
group and E, the absorbance of test group. according with the method of Dahlgren et al. 2002.

Determination of acetylcholinesterase percent Cell culture


inhibition
Human neuronal cell line (SH-SY5Y) was purchased from
The anti-acetylcholinesterase activity of B. ternifolia American Type Culture Collection (ATCC, Manassas, VA,
extract was determined spectrophotometrically following USA). Cell line was routinely grown at 37 C in a hu-
the method described by Ellman (Ellman et al. 1961), midified incubator with 5 % CO2 in Dulbecco’s modified
using acetyl- cholinesterase from rat brain homogenate Eagle’s medium (DMEM) supplemented with antibiotics,
and acetylcholine iodide as substrate. All assays were 10 % fetal bovine serum and 2 mM glutamine (Invitrogen).
performed in a final vol- ume of 3.0 ml in the presence
of 0.1 M phosphate-buffer
G. Garc´ıa-Morales et
al.
Anti-Alzheimer’s diseases effects of B. ternifolia
Cell viability assay C18) (Macherey–Nagel). Gradient mixture of water/ace-
tonitrile was used as mobile phase to obtain four fractions:
The protective effect of B. ternifolia in AD was evaluated BtC1M(100:0); BtC1N(80:20); BtC1Q(70:30), and
by the quantification of the cell viability of the neurob- BtFlav3(50:50) (Fig. 1). The fraction BtC1M was mainly
lastoma cells, SH-SY5Y. Neurons were treated with in- constituted of the polyphenolic compound chlorogenic acid
creasing concentrations of fibrillar preparations of Ab1–42 (3) which experimental spectroscopic and spectrometric
(Dahlgren et al. 2002; Patel and Good 2007) in presence of data of the compound isolated where according with pre-
the extract or fractions of B. ternifolia. Previously at the vious reports (Sari 2010). Regarding the BtFlav3 fraction,
assays, the effective dose (ED50) of the extract and frac- its principally content was the flavonol 3-O-quercetin
tions were determined. Briefly, SH-SY5Y cells were plated rhamnopyranoside (4), following by 3-O-quercetin glu-
3
(5 9 10 cell/well) in 96-well plates in 100 ll of media. copyranoside (5), and rutin (6). In the case of compounds 4
24 h later, the cells were rinsed with PBS. Vehicle or and 5, the identification was achieved by comparison be-
peptide (0.001–100 lM) were prepared in serum-free tween the experimental data obtained by spectroscopic and
medium and added to the cell in presence of extract or spectrometric data of the isolated compounds and the lit-
fractions of B. ternifolia (6.25 lg/well). The treated cells erature (Kombal and Glasl 1995; Yen et al. 2009). The
were incubated for 24 h in routinely growth conditions. identification of compound 6 was determined by HPLC
Cell viability was determinate by the Cell Proliferation Kit analysis when experimental peak overlapped with the
II (XTT) (Roche Applied Science) following the commercial standard, although the detection wavelength
manufacturer’s protocol. Viability was expressed as per- was scanned at 190–400 nm. In the remaining fraction
centage of control [(absorbance of treated neurons/ab- BtC1N, we identified the coumarin scopoletin (7) by
sorbance of untreated neurons) 9 100)] (Tarozzi et al. overlapping between the experimental peak and the com-
2010). Assays were performed in triplicate incubations mercial standard (190–400 nm scanned wavelength).
from three independent experiments. Data were analyzed In order to isolate and quantified these triterpens, we
by one-way ANOVA followed by Dunnet´ts test for com- followed the method described by Nagafuji (Nagafuji et al.
parisons against control. Values of p B 0.05 (*) and 2004). Briefly, 20 g of BtHA were suspended in 60 %
p B 0.001 (***) were considered statistically significant. methanol and centrifuged (at 3000 rpm, 50 min, 25 C) to
obtain a supernatant (BtS/NHA) and a precipitate (pp)
Phytochemical analysis of BtHA (Fig. 1). Then, 2 g of precipitate were recovered, sus-
pended in 60 % methanol solution, and passed through a
Purification of the main compounds of BtHA was per- Diaion HP-20 column (Merck & Co., Inc) (BtFR1). The
formed by successive chromatography methods (Fig. 1) methanol solution was passed through the same column
whereas the identification of the isolated compounds was (BtFR2), where a spontaneous precipitate (BtPPF2) and a
1 13
carried on by spectroscopic ( H NMR, C NMR, HMQC), supernatant (BtS/NF2) were obtained. Finally, the ethyl
spectrometric (EI) and HPLC techniques. The quantifica- acetate solution was treated in the same way (BtFR3)
tion of the identified compounds was achieved by HPLC (Fig. 1). An abundant mixture of ursolic and oleanolic acid
using calibration curves. Experimental conditions for iso- was detected in BtPPF2 and BtS/NF2 which experimental
lation, identification and quantification of compounds de- spectroscopic data and HPLC analysis were found to be in
tected in BtHA extract are described: 26 g of BtHA were good agreement with the values reported by
fractioned by a bipartition process with ethyl acetate/water, Razborsˇek et al. (2008).
obtaining BtAcOEt fraction and BtAq fraction. The BtAq The HPLC analysis of BtHA was conducted on a Waters
fraction was dissolved in methanol to obtain a supernatant 2695 liquid chromatographer equipped with a Waters 2996
(BtMeOH) and a precipitate (BtPP) (Fig. 1). 4 g of BtA- photodiode array detector in order to quantify the com-
cOEt were subjected to an open chromatography column pounds detected. Separation was carried out using a RP
previously packed with silica gel (mesh 70-230) (Merck). C-18 Superspher (Merck) HLPC column (120 9 4 mm;
The elution solvent consisted of an n-hexane/ethyl acetate/ 5 lm) with the following solvent ratios for the mobile
methanol gradient mixture, in which we obtained six phase where solvent A is water and solvent B is acetoni-
fractions: Fr-A(100:0:0); Fr-B(75:25:0); Fr-C(5:5:0); Fr- trile: A:B = 100:0 (0 9 1 min); 90:10 (2–4 min); 80:20
D(5:5:1); Fr-E(5:5:2.5), and Fr-F(0:0:100) (Fig. 1). From (5–9 min); 70:30 (10–15 min); 60:40 (16–18 min); 40:60
the subsequent fractionation of Fr-A, Fr-B, and Fr-C by (19–20 min); 0:100 (21–30 min), and 100:0 (31–32 min).
normal open chromatography column, we detected ursolic Sample injection volume was 10 ll with a 1 ml/min flow
(1) and oleanolic acid triterpens (2) (Fig. 1). rate. The detection wavelength was scanned at
On the other hand, fractions Fr-D, Fr-E, and Fr-F were 190–400 nm. Quantification of the main compounds was
grouped and purified by reversed phase (Polygoprep 60-50 achieved using calibration curves (linear regression where
G. Garc´ıa-Morales et
al.

Fig. 1 Schematic representation of the fractionation of the hydroalcoholic extract of Bouvardia ternifolia. Black boxes are the fractions tested.
Dashes boxes are the chemical compounds identified
Anti-Alzheimer’s diseases effects of B. ternifolia

2
r [ 0.9807), which were separately constructed with 0.12 mg), and BtF3 (1.71 ± 0.45 mg) (Fig. 2c) show a
pure standards. The HPLC-solvents and HPLC-references significant anti-inflammatory effect by reducing inflam-
were purchased from Sigma or Merck. mation by [70 % in the TPA-induced ear edema assay. In
contrast, no anti-inflammatory effects were detected in
Statistical analysis fractions BtMeOH, BtPP (Fig. 2a), BtC1Q, BtFlav3
(Fig. 2b), BtS/NHA, BtFR1, and BtS/NF2 (Fig. 2c) when
All data were analyzed using the GraphPad prism software compared with the Dexamethasone-treated control (0.97 ±
(GraphPad Software) and represented as mean ± Standard 0.35 mg).
error of the mean (SEM). Data were evaluated by one-way
ANOVA followed by Dunnett’s test for comparisons Evaluation of antioxidant activity
against control. Values of p B 0.05 or higher were con-
sidered statistically significant. IC50 were estimated by BtHA showed lipid peroxidation protection at 100 lg/ml
means of a linear regression equation. (8.79 ± 1.7 nM/mg protein), as well as the BtAcOEt
fraction (2.68 ± 0.46 nM/mg protein) (Fig. 3a) and
BtC1M (2.65 ± 0.19 nM/mg protein) (Fig. 3b), in com-
Results parison with the control. However, 10-lg/ml concentra-
tions of these same fractions did not show significant
Evaluation of anti-inflammatory activity effects. In Fig. 3b, the antioxidant effect of fractions
BtC1N and BtFlav3 was dose-dependent. BtC1N showed
BtHA extract (3.09 ± 0.74 mg) and fractions BtAcOEt 6.67 ± 0.49 nM/mg proteins of TBARS at 10 lg/ml and
(1.48 ± 0.19 mg) (Fig. 2a), BtC1M (1.95 ± 0.26 mg), 0.64 ± 0.04 nM/mg protein at 100 lg/ml, while in the case
BtC1N (1.25 ± 0.37 mg) (Fig. 2b), BtPPF2 (0.76 ± of BtFlav3, the latter showed 6.02 ± 0.44 nM/mg proteins

123
Fig. 2 Evaluation of Bouvardia ternifolia as anti-inflammatory by [70 % when compared with the Dexamethasone-treated control. In
(TPA)-induced ear edema in mice. BtHA extract and fractions contrast, no anti-inflammatory effects were detected in the remaining
BtAcOEt (a), BtC1M, BtC1N (b), BtPPF2, and BtF3 (c) show a fractions. Results represent mean ± SEM. ***p B 0.001 compared to
significant anti-inflammatory effect by reducing inflammation by TPA. Each value represents the mean of six observations

Fig. 3 Antioxidant activities of Bouvardia ternifolia evaluated by BtPPF2, and BtF3 (c) did not show protection against lipid
TBARS assays. BtHA and BtAcOEt fraction showed lipid peroxida- peroxidation. Data are shown as nmoles of TBARS per mg of
tion protection at 100 lg/ml (a) as well as BtC1M fraction (b), in protein. Each value represents the mean of four observations ± SEM.
comparison with the control. b The antioxidant effect of fractions The value p B 0.05 (*) and p B 0.01 (**) were considered as
BtC1N and BtFlav3 was dose-dependent. The remaining fractions, significant difference with respect to the standard
BtMeOH, BtPP (a), BtC1Q (b), and BtS/NHA, BtFR1, BtS/NF2,

of TBARS production at 10 lg/ml and 0.34 ± 0.02 nM/mg 0.20 nM/mg protein), and a-Tocopherol (1.46 ± 0.56 nM/
protein at 100 lg/ml. Both fractions exhibited a higher mg protein) (Fig. 3b). The remaining fractions, BtMeOH,
antioxidant effect than the references: Quercetin (0.98 ± BtPP (Fig. 3a), BtC1Q (Fig. 3b), and BtS/NHA, BtFR1,
BtS/NF2, BtPPF2, and BtF3 (Fig. 3c) did not show pro- Protective effect of Bouvardia ternifolia in Alzheimer’s
tection against lipid peroxidation. disease
On the other hand, the IC50 values of fractions BtFlav3
(30.67 ± 2.09 lg/ml), BtAcOEt (42.66 ± 0.93 lg/ml), The protective effect of B. ternifolia against Ab1–42 fib-
and BtC1N (59.94 ± 10 lg/ml) exhibited an antioxidant rillar-induce neurotoxicity in terms of neuronal viability in
effect in contrast to the BtHA crude extract (356.21 ± SH-SY5Y cell was evaluated. The toxic effects of in-
22.89 lg/ml) (Fig. 4). However, these IC50 values were creasing concentrations (0.001–100 lM) of fibrillar form
greater than those exhibited by references Quercetin of Ab1–42 were determined by using the XTT assays. When
(0.50 ± 0.01 lg/ml) and a-Tocopherol (2.92 ± 0.93 lg/ b-amyloid peptide was tested individually, a maximal
ml) (Fig. 4). neurotoxicity was observed at 1.0, 10 and 100 lM of
peptide (84.63 ± 2.80, 69.98 ± 5.47 and 38.39 ± 4.06 %
Determination of percentages of inhibition of neuronal viability, respectively) (Fig. 6). On the other
on acetylcholinesterase hand, 6.5 lg/well of BtAcOEt fraction was added to b-
amyloid treated SH-SY5Y cells. This fraction was able to
BtHA extract (12.26 ± 2.53 %) and fraction BtAcOEt revert significantly the fibrillar peptide toxicity at 10 and
(13.82 ± 2.04 %) showed significant inhibition on acetyl- 100 lM of peptide (83.97 ± 5.03 and 69.33 ± 4.56 % of
cholinesterase enzyme when compared to the control free neuronal viability, respectively) (Fig. 6). In the case of
on inhibitor (Fig. 5a), while BtMeOH and BtPP did not BtHA extract, it was observed an improving in neuronal
show significant effect. In Fig. 5b, the inhibitory effect of protection at 10 and 100 lM of peptide (88.90 ± 5.26 and
fractions BtC1M (38.43 ± 3.94 %) and BtFlav3 (34.08 ± 49.77 ± 3.98 % of neuronal viability, respectively); how-
3.52 %) on the enzyme was significant, and in the case of ever, it did not shown significantly decreased of Ab-in-
fraction BtC1M the percentages of inhibition even com- duced cell death with respect to the control (p B 0.05)
parable with Tacrine (40.61 ± 2.28 %). The remaining (Fig. 6). Fractions BtMeOH, BtPP, BtCIM, BtC1N,
fractions, BtC1N, BtC1Q (Fig. 5b), and BtS/NHA, BtFR1, BtC1Q, BTFlav3, BtS/NHA, BtFR1, BtS/NF2, BtPPF2 and
BtS/NF2, BtPPF2, and BtF3 (Fig. 5c) did not show inhi- BtFR3 did not shown significant statically differences with
bition on the acetylcholinesterase enzyme. respect to the control.

Phytochemical analysis of BtHA

The chemical compounds detected and isolated from the


active BtHA extract were identified as polyphenols
(chlorogenic acid, 3-O-quercetin rhamnopyranoside, 3-O-
quercetin glucopyranoside, rutin), triterpens (ursolic and
oleanolic acid) and a cumarin (scopoletin). The quantita-
tive analysis showed that 3-O-quercetin glucopyranoside
was the main compound found in BtHA, since the quan-
tification analysis showed as the most abundant (415 mg/g
of extract) (Table 1). The quantification values obtained for
the rest of the compounds are showed in Table 1.

Discussion

At present, cholinesterase inhibitors are one of the most


prescribed drug classes for AD treatment (Senol et al.
2011); however, these drugs cause many side effects (Dunn
et al. 2000; Alfirevic et al. 2007). In consequence, finding
new sources for the safest and most effective drugs against
this disease becomes relevant. Recent evidence suggests
that inflammatory responses and antioxidant properties
Fig. 4 Displacements assays. Data shown percentage of inhibition of
lipid peroxidation (%) by increasing concentrations of the BtHA, may contribute significantly to the progression and
BtAcOEt, BtC1N and BtFlav3. Quercetin and a-Tocopherol were chronicity of AD (Heneka and O’Banion 2007). Therefore,
employed as references. Data represents the mean of 4 assays ± SEM in the current study was shown the properties of the B.
123
Fig. 5 Anti-acetylcholinesterase activity of Bouvardia ternifolia. NHA, BtFR1, BtS/NF2, BtPPF2, and BtF3 (c) did not show inhibition
BtHA extract and fraction BtAcOEt showed significant inhibition on the acetylcholinesterase enzyme. Each value represents the mean
on acetylcholinesterase enzyme when compared to the control free on of six observations ± SEM. The value p B 0.05 (*) and p B 0.01
inhibitor (a), in contrast with BtMeOH and BtPP fractions. b The (**) were considered as significant difference with respect to the
inhibitory effect of fractions BtC1M and BtFlav3 on the enzyme was standard
significant. The remaining fractions, BtC1N, BtC1Q (b), and BtS/

maintained significantly activity in the entire biological test


considered at the in vitro AD model. In opposite fashion,
although BtHA and BtC1M shown anti-inflammatory, an-
tioxidant, and inhibitory properties on the percentage of
inhibition on acetylcholinesterase enzyme, they were un-
able to prevent neural damage. The fraction BtFlav3 was
able to show antioxidant effect and acetylcholinesterase
inhibition, but this fraction not exhibited effect in the anti-
inflammatory test, as well as the fraction BtC1N, which
exerted anti-inflammatory and antioxidant effects, but not
inhibitory effect on the acetylcholinesterase. Thus,
although BtAcOEt is a fraction derivate from BtHA, it is
also the direct result of removing the most polar fraction
from the whole crude extract. In this way, as many reports
indicate (Lin et al. 2007; Deharo and Ginsburg 2011;
Rosoanaivo et al., 2011), it is well known fact in phyto-
Fig. 6 Neuronal viability of SH-SY5Y cells treated with increasing
chemistry that crude plant extracts, that include multiple
concentrations of fibrillar preparations of Ab1–42 in absence or and diverse compounds (Kurapati et al. 2013), are usually
presence of Bouvardia ternifolia extract or fractions (6.25 lg/well). more powerful medicines than pure isolated compounds.
Cell viability was expressed as percentage of control ± Standard This effect may be due to the multiple actions of complex
error of the mean (SEM) from three independent experiments.
mixtures, but could also arise from synergistic interactions
Statistically significant differences compared to controls are shown
[p \ 0.05 (*), p B 0.001 (***)] among their components (Fernandez et al. 2005).
The chemical analysis of BtHA performed in the present
ternifolia extract in an in vitro AD model which comprised contribution revealed polyphenolic compounds as the ma-
the anti-inflammatory, the antioxidant, the percentage of joritary content. Among them, 3-O-quercetin glucopyra-
inhibition on acetylcholinesterase enzyme, and additional- noside is the majority constituent and could be considered
ly, the neuroprotective effects of this extract, supporting its as the main element involved in the anti-inflammatory and
multi-target action. antioxidant effects observed. This is consistent with find-
In the present study, among the BtHA extract and the ings in which quercetin and its glucopyranoside derivatives
fractions derived, BtAcOEt is the only fraction that are one of the most abundant polyphenols found in
Table 1 Chemical compounds isolated and identified in the active inflammatory and antioxidant activities ([90 %) which
hydroalcoholic extract of Bouvardia ternifolia (BtHA)
are in agreement with previous reports of the biological
Compound Quantity present properties of this coumarin (Kim et al. 2004; Chang et al.
in BtHA (mg/g of extract) 2012). Scopoletin is a coumarin that exerts inhibitory ef-
Ursolic acid 54 fects on the production of PGE2, TNF-a, IL-1b, and IL-6 in
Oleanolic acid 20.8 macrophages (Kim et al. 2004). Likewise, the triterpenes
Chlorogenic acid 9.5 and ursolic and oleanolic acids (mainly concentrated in
3-O-quercetin rhamnopyranoside 12.8 fraction BtPPF2) showed potent anti-inflammatory activity
3-O-quercetin glucopyranoside 415 in the present study that is consistent with the literature
Rutin 229.9 (Liu 1995). Some reports indicate that ursolic acid also
Scopoletin 1.38 reduces Ab-induced oxidative damage, such as free radical
formation and lipid peroxidation, in in vitro assay systems,
in turn also reducing the production of proinflammatory
vegetables and fruits with potent anti-inflammatory and cytokines, leading to a neuroprotective effect against Ab
antioxidant properties (Materska 2008). On the other hand, (Yoo and Park 2012; Yoon et al. 2014). Therefore, taking
protection against lipid peroxidation and high bioavail- all of this information together, there is the possibility that
ability in the digestive tract has been shown for both the neuroprotector properties exerted by BtHA may be due
quercetin and glucopyranoside derivatives (Wagner et al. to the anti-inflammatory, the antioxidant and the inhibitory
2006). on acetylcholinesterase effects that could be attributed to
The remainder of the polyphenols detected may also be its contents of polyphenols, coumarins, and triterpenes. The
involved in the biological effects observed: rutin, in par- establishment of the precise role and combination of these
ticular, has been described as a multifunctional agent that active components requires further investigation.
can inhibit the specific pathogenic processes involved in
AD, such as Ab aggregation, prevent mitochondrial dam-
age, reduce ROS production, reduce the generation of Conclusion
Nitric oxide (NO), diminish the activity of inducible Nitric
oxide synthase (iNOS), and decrease the production of The hydroalcoholic extract of B. ternifolia, which contains
proinflammatory cytokines (Pu et al. 2007; Wang et al. polyphenol compounds such as, 3-O-quercetin glucopyra-
2012). In the same line, chlorogenic acid is able to regulate noside, 3-O-quercetin rhamnopyranoside, rutin, and
inflammation by the increase of proinflammatory factors chlorogenic acid, as well as coumarin, scopoletin, and the
such as Interleukin-6 (IL-6) and Tumor necrosis factor- triterpenes ursolic and oleanolic acid, is able to induce
alpha (TNF-a) (Du et al. 2013) and by inhibition of significant anti-inflammatory and antioxidant effects, as
Prostaglandin E2 (PGE2) production (Shan et al. 2009). well as inhibition on acetylcholinesterase enzyme, and
The antioxidant activities of chlorogenic acid are also ob- conferring neuroprotection against b-amyloid peptide for
served in its inhibiting the formation of ROS or its scav- AD. These data support its use in traditional medicine and
enging of the latter (Morishita and Ohnishi 2001). encourages future studies for its development as a
Interestingly, in this work, we also found that fraction therapeutic agent in the treatment of AD.
BtC1M, which mainly content is chlorogenic acid, was
able to inhibit acetylcholinesterase in values comparable to Acknowledgments This study was supported by grant R-2010-
control (Tacrine). This information is in agreement with 1701-57 (to A. A.-R.) from the FIS-Instituto Mexicano del Seguro
Social, Me´xico. G. Garc´ıa-Morales is grateful for a doctoral
Kwon and collaborators, who reported the inhibitory ac- fellow- ship from CONACyT (Exp. No. 212,779). The authors wish
tivity on acetylcholinesterase by chlorogenic acid in a dose to thank Jonathan Ordun˜ o, Arturo Pe´rez (CIBIS-IMSS), and
dependent-manner (Kwon et al. 2010). Thus, the potential Antonio Nieto- Camacho (Instituto de Qu´ımica-UNAM) for
neuroprotective effects of the chemical compounds present technical assistance.
in BtHA permit us to suggest this plant as an alternative Conflict of interest The authors declare that there is no conflict of
therapeutic agent against AD. interests regarding the publication of this article.
The fact that polyphenols are the main compounds de-
tected in BtHA does not preclude their possible active in-
volvement in the anti-inflammatory and antioxidant References
activities of the remaining compounds identified in BtHA,
such as coumarin and the triterpenes. This is the case of Argueta-Villamar, A., L.M. Cano-Asseleih, and M.E. Rodarte. 1994.
Atlas de las Plantas de la Medicina Tradicional Mexicana.
fraction BtC1N, where scopoletin was identified as the Mexico D.F.: Instituto Nacional Indigenista.
main compound. This fraction exerted high anti- Alfirevic, A., T. Mills, D. Carr, B.J. Barratt, A. Jawaid, J. Sherwood,
J.C. Smith, J. Tugwood, R. Hartkoorn, A. Owen, K.B. Park, and
123
M. Pirmohamed. 2007. Tacrine-induced liver damage: An Bouvardin and deoxybouvardin, antitumor cyclic hexapeptides
analysis of 19 candidate genes. Pharmacogenetics and Genomics from Bouvardia ternifolia (Rubiaceae). Journal of the American
17: 1091–1100. Chemical Society 99: 8040–8044.
Chang, T.N., J.S. Deng, Y.C. Chang, C.Y. Lee, and L. Jung-Chun. Kim, H.J., S.I. Jang, Y.J. Kim, H.T. Chung, Y.G. Yun, T.H. Kang,
2012. Ameliorative effects of scopoletin from Crossostephium O.S. Jeong, and Y.C. Kim. 2004. Scopoletin suppresses pro-
chinensis against inflammation pain and its mechanisms in mice. inflammatory cytokines and PGE2 from LPS-stimulated cell
Evidence-Based Complementary and Alternative Medicine 2012: line, RAW 264.7 cells. Fitoterapia 75: 261–266.
1–10. Kombal, R., and H. Glasl. 1995. Flavan-3-ols and flavonoids from
Chen, Q.X., and I. Kubo. 2002. Kinetics of mushroom tyrosinase Potentilla anserina. Planta Medica 61: 484–485.
inhibition by quercetin. Journal of Agriculture and Food Kurapati, K.R.V., V.S.R. Atluri, T. Samikkannu, and M.P.N. Nair.
Chemistry 50: 4108–4112. 2013. Ashwagandha (Withania somnifera) reverses b-amyloid1-
Dahlgren, K.N., A.M. Manelli, W.B. Stine Jr, L.K. Baker, G.A. 42 induced toxicity in human neuronal cells: Implications in
Krafft, and M.J. LaDu. 2002. Oligomeric and fibrillar species of HIV-associated neurocognitive disorders (HAND). PLoS One 8:
amyloid-beta peptides differentially affect neuronal viability. e77624.
Journal of Biological Chemistry 277: 32046–32053. Kwon, S.H., H.K. Lee, J.A. Kim, S.I. Hong, H.C. Kim, T.H. Jo, Y.I.
De Young, L.M., J.B. Kheifets, S.J. Ballaron, and J.M. Young. 1989. Park, C.K. Lee, Y.B. Kim, S.Y. Lee, and C.G. Jang. 2010.
Edema and cell infiltration in the phorbol ester-treated mouse ear Neuroprotective effects of chlorogenic acid on scopolamine-
are temporally separate and can be differentially modulated by induced amnesia via anti-acetylcholinesterase and anti-oxidative
pharmacologic agents. Agents and Actions 26: 335–341. activities in mice. European Journal of Pharmacology 649:
Deharo, E., and H. Ginsburg. 2011. Analysis of additivity and 210–217.
synergism in the anti-plasmodial effect of purified compounds Lee, Y.J., S.B. Han, S.Y. Nam, K.W. Oh, and J.T. Hong. 2010.
from plant extracts. Malaria Journal 10(Suppl 1): S5. Inflammation and Alzheimer’s disease. Archives of Pharmacal
Du, W.Y., C. Chang, Y. Zhang, Y.Y. Liu, and K. Sun. 2013. High- Research 33: 1539–1556.
dose chlorogenic acid induces inflammation reactions and Lin, F.M., L.R. Chen, E.H. Lin, F.C. Ke, H.Y. Chen, M.J. Tsai, and
oxidative stress injury in rats without implication of mast cell P.W. Hsiao. 2007. Compounds from Wedelia chinensis syner-
degranulation. Journal of Ethnopharmacology 147: 74–83. gistically suppress androgen activity and growth in prostate
Dunn, N.R., G.L. Pearce, and S.A. Shakir. 2000. Adverse effects cancer cells. Carcinogenesis 28: 2521–2529.
associated with the use of donepezil in general practice in Liu, J. 1995. Pharmacology of oleanolic acid and ursolic acid. Journal
England. Journal of Psychopharmacology 14: 406–408. of Ethnopharmacology 49: 57–68.
Ellman, G.L., K.D. Courtney, B. Andres, and R.M. Featherstone Jr. Materska, M. 2008. Quercetin and its derivatives: Chemical structure
1961. New and rapid colorimetric determination of acetyl- and bioactivity—a review. Polish Journal of Food and Nutrition
cholinesterase activity. Biochemical Pharmacology 7: 88–95. Sciences 58: 407–413.
Esterbauer, H., and K.H. Cheeseman. 1990. Determination of Morishita, H., and M. Ohnishi. 2001. Absorption, metabolism and
aldehydic lipid peroxidation products: Malonaldehyde and biological activities of chlorogenic acids and related compounds.
4-hydroxynonenal. Methods in Enzymology 186: 407–421. Studies in Natural Products Chemistry 25: 919–953.
Fernandez, S.P., C. Wasowski, A.C. Paladini, and M. Marder. 2005. Nagafuji, S., H. Okabe, H. Akahane, and F. Abe. 2004. Trypanocidal
Synergistic interaction between hesperidin, a natural flavonoid, constituents in plants 4. Withanolides from the aerial parts of
and diazepam. European Journal of Pharmacology 512: Physalis angulata. Biological and Pharmaceutical Bulletin 27:
189–198. 193–197.
Francis, P.T., A.M. Palmer, M. Snape, and G.K. Wilcock. 1999. The Ng, T.B., F. Liu, and Z.T. Wang. 2000. Antioxidative activity of
cholinergic hypothesis of Alzheimer’s disease: A review of natural products from plants. Life Sciences 66: 709–723.
progress. Journal of Neurology, Neurosurgery and Psychiatry Ohkawa, H., N. Ohishi, and K. Yagi. 1979. Assay for lipid peroxides
66: 137–147. in animal tissues by thiobarbituric acid reaction. Analytical
Gonza´lez, A., E. Go´ mez, A. Corte´s-Lozada, S. Herna´ndez, T. Biochemistry 95: 351–358.
Ramirez- Apan, and A. Nieto-Camacho. 2009. Pasinetti, G.M. 2012. Novel role of red wine-derived polyphenols in
Hepatocoordinate Tin(IV) the prevention of Alzheimer’s disease dementia and brain
compounds derived from pyridine Schiff bases: Synthesis, char- pathology: Experimental approaches and clinical implications.
acterization, in vitro cytotoxicity, anti-inflammatory and an- Planta Medica 78: 1614–1619.
tioxidant activity. Chemical & Pharmaceutical Bulletin 57: 5–15. Patel, D., and T. Good. 2007. A rapid method to measure beta-
Hampel, H., D. Prvulovic, S. Teipel, F. Jessen, and C. Luckhaus. amyloid induced neurotoxicity in vitro. Journal of Neuroscience
2011. The future of Alzheimer’s disease: The next 10 years. Methods 161: 1–10.
Progress in Neurobiology 95: 718–728. Pu, F., K. Mishima, K. Irie, K. Motohashi, Y. Tanaka, K. Orito, T.
Heneka, M.T., and M.K. O’Banion. 2007. Inflammatory processes in Egawa, Y. Kitamura, N. Egashira, K. Iwasaki, and M. Fujiwara.
Alzheimer’s disease. Journal of Neuroimmunology 184: 69–91. 2007. Neuroprotective effects of quercetin and rutin on spatial
Herrera-Ruiz, M., G. Garc´ıa-Morales, A. Zamilpa, M. Gonza memory impairment in an 8-arm radial maze task and neuronal
´lez- Cortazar, J. Tortoriello, E. Ventura-Zapata, and E. Jime death induced by repeated cerebral ischemia in rats. Journal Of
´nez- Ferrer. 2012. Inhibition of acetylcholinesterase activity Pharmacological Sciences 104: 329–334.
by hidroalcoholic extract and their fractions of Bouvardia ternifolia Razborsˇek, M.I., D.B. Voncˆina, V. Dolecˆek, and E. Voncˆina.
(Cav.) Schltdl (Rubiaceae). Boletin Latinoamericano y del 2008.
Caribe de Plantas Medicinales y Aromaticas 11: 526–541. Jimenez- Determination of oleanolic, betulinic and ursolic acid in
Ferrer, E., I. Reynosa-Zapata, Y. Perez-Torres, and J. Lamiaceae and mass spectral fragmentation of their trimethylsi-
Tortoriello. 2005. The secretagogue effect of the poison from lylated derivatives. Chromatographia 67: 433–440.
Centruroides limpidus limpidus on the pancreas of mice and the Recio, M.C., R.M. Giner, S. Manez, and J.L. Rios. 1994. Structural
antagonistic action of the Bouvardia ternifolia extract. Phy- considerations on the iridoids as anti-inflammatory agents.
tomedicine 12: 65–71. Planta Medica 60: 232–234.
Jolad, S.D., J.J. Hoffmann, S.J. Torrance, R.M. Wiedhopf, J.R. Cole, Rosoanaivo, P., C.W. Wright, M.I. Willcox, and B. Gilbert. 2011.
S.K. Arora, R.B. Bates, R.L. Gargiulo, and G.R. Kriek. 1977. Whole plant extracts versus single compounds for the treatment
of malaria: Synergy and positive interactions. Malaria Journal Rutin inhibits beta-amyloid aggregation and cytotoxicity, at-
10(Suppl 1): S4. tenuates oxidative stress, and decreases the production of nitric
Sari, A. 2010. Two new 3-benzylphthalides from Scorzonera oxide and proinflammatory cytokines. Neurotoxicology 33:
veratrifolia Fenzl. Natural Product Research 24: 56–62. 482–490.
Senol, F.S., K. Skalicka Wozniak, M.T.H. Khan, I. Erdogan Orhan, B. Weiner, M.W., D.P. Veitch, P.S. Aisen, L.A. Beckett, N.J. Cairns,
Sener, and K. Głowniak. 2011. An in vitro and in silico approach R.C. Green, D. Harvey, C.R. Jack, W. Jagust, E. Liu, J.C.
to cholinesterase inhibitory and antioxidant effects of the Morris, R.C. Petersen, A.J. Saykin, M.E. Schmidt, L. Shaw, J.A.
methanol extract, furanocoumarin fraction, and major coumarins Siuciak, H. Soares, A.W. Toga, and J.Q. Trojanowski. 2012. The
of Angelica officinalis L. fruits. Phytochemistry Letters 4: Alzheimer’s disease neuroimaging initiative: A review of papers
462–467. published since its inception. Alzheimer’s & Dementia 8: S1–
Shan, J., J. Fu, Z. Zhao, X. Kong, and H. Huang. 2009. Chlorogenic S68.
acid inhibits lipopolysaccharide-induced cyclooxygenase-2 ex- Wollen, K.A. 2010. Alzheimer’s disease: The pros and cons of
pression in RAW264.7 cells through suppressing NF-kappaB pharmaceutical, nutritional, botanical, and stimulatory therapies,
and JNK/AP-1 activation. International Immunopharmacology with a discussion of treatment strategies from the perspective of
9: 1042–1048. patients and practitioners. Alternative Medicine Review 15:
Tarozzi, A., F. Morroni, A. Merlicco, C. Bolondi, G. Teti, M. Falconi, 223–244.
G. Cantelli-Forti, and P. Hrelia. 2010. Neuroprotective effects of Yen, C.T., P.W. Hsieh, T.L. Hwang, Y.H. Lan, F.R. Chang, and Y.C.
cyanidin 3-O-glucopyranoside on amyloid beta (25-35) oligo- Wu. 2009. Flavonol glycosides from Muehlenbeckia platyclada
mer-induced toxicity. Neuroscience Letters 473: 72–76. and their anti-inflammatory activity. Chemical & Pharmaceuti-
Tobey, R.A., D.J. Orlicky, L.L. Deaven, L.B. Rall, and R.J. Kissane. cal Bulletin 57: 280–282.
1978. Effects of Bouvardin (NSC 259968), a cyclic hexapeptide Yoo, K.Y., and S.Y. Park. 2012. Terpenoids as potential anti-
from Bouvardia ternifolia, on the progression capacity of Alzheimer’s disease therapeutics. Molecules 17: 3524–3538.
cultured Chinese hamster. Cancer Research 38: 4415–4421. Yoon, J.H., K. Youn, C.T. Ho, M.V. Karwe, W.S. Jeong, and M. Jun.
Wagner, C., R. Fachinetto, C.L. Dalla Corte, V.B. Brito, D. Severo, 2014. p-Coumaric acid and ursolic acid from Corni fructus
G. de Oliveira Costa Dias, A.F. Morel, C.W. Nogueira, and J.B. attenuated b-amyloid (25-35)-induced toxicity through regula-
Rocha. 2006. Quercitrin, a glycoside form of quercetin, prevents tion of the NF-jB signaling pathway in PC12 cells. Journal of
lipid peroxidation in vitro. Brain Research 1107: 192–198. Agriculture and Food Chemistry 62: 4911–4916.
Wang, S.W., Y.J. Wang, Y.J. Su, W.W. Zhou, S.G. Yang, R. Zhang,
M. Zhao, Y.N. Li, Z.P. Zhang, D.W. Zhan, and R.T. Liu. 2012.

You might also like