You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/267037009

Anti-Alzheimer, antioxidant activities and flavonol glycosides of Eryngium


campestre L

Article  in  Current Chemical Biology · January 2013

CITATIONS READS

9 323

4 authors, including:

Usama W Hawas Hanem Awad


National Research Center, Egypt National Research Center, Egypt
80 PUBLICATIONS   704 CITATIONS    101 PUBLICATIONS   1,480 CITATIONS   

SEE PROFILE SEE PROFILE

Hanan Taie
National Research Center, Egypt
43 PUBLICATIONS   540 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Endophytic fungi from Medicinal herbs View project

Redoxcycling and toxicity of catechol containing flavonoids and their glutathione adducts View project

All content following this page was uploaded by Hanan Taie on 20 September 2016.

The user has requested enhancement of the downloaded file.


188 Current Chemical Biology, 2013, 7, 188-195

Anti-Alzheimer, Antioxidant Activities and Flavonol Glycosides of


Eryngium campestre L.
Usama W. Hawas*,1, Lamia T. Abou El-Kassem2, Hanem M. Awad3 and Hanan A.A. Taie4

1
Phytochemistry and Plant Systematic Department; 2Department of Pharmacognosy; 3Department of Tanning Materials
and Leather Technology; 4Department of Plant Biochemistry, National Research Centre, El-Bohooth Street, 12311-
Dokki, Cairo, Egypt

Abstract: Phytochemical investigation of the methanolic extract of Eryngium campestre L. aerial parts led to isolation of
eleven known flavonol glycosides. Structures were elucidated by spectroscopic and chemical methods. The methanol
extract of E. campestre and the isolated flavonols exhibited moderate to strong antioxidant activity in DPPH radical
scavenging and reducing power assays. Eryngium campestre extract showed significant inhibition of the -amyloid A 42
(IC50 = 155.75±7.43 ng/ml) without significant reduction in total A (A 40 + A 42) levels in human H4 cell line, using
sensitive sandwich enzyme linked immunosorbent assay (ELISA). The results showed no inhibition activity of the extract
against COX-1 and COX-2 up to 400 ng/ml concentration.

Keywords: Alzheimer's disease, -amyloid peptide (A), antioxidant, reducing power, flavonol glycosides, Eryngium
campestre.

INTRODUCTION consecutive cleavages of - and -secretase to build up


extracellular A deposits. However, the APP derivative A
Eryngium is the largest and arguably the most taxonomically
42 is thought to be a key factor in the pathogenesis of AD, as
complex genus in the family Apiaceae. The genus is
A 42 is more responsible for fibril formation due to its self-
represented by 317 taxa widespread throughout Central Asia, assembly and turned out to be a major A constituent of
northern Africa, America, Central and Southeast Europe [1].
amyloid plaques. At present, no causal therapy for AD is
Eryngium campestre (Field eryngo) is a rare perennial clinically accessible regardless of intense research
restricted to dry grassy areas near the coast. It is native to endeavours. Anti-oxidant therapy, AChE inhibitors, nicotinic
Spain, France, Germany and Greece and other scattered and muscarinic agonists, nerve growth factor (NGF), low
localities in Europe, and is also found in Africa and Asia [2]. molecular lipophilic compounds, anti-inflammatory drugs,
It has been used in European herbal medicine as an infusion drugs which interfere with A formation and deposition, and
to treat whooping cough, kidney and urinary tract drugs that attenuate A toxicity are the existing
inflammations [3]. Previous phoytochemial studies on pharmacological advances related to AD treatment.
Eryngo revealed the presence of triterpenoid saponins [4], Regardless of the way in which A exerts its deleterious
flavonoids: kaempferol and quercetin derivatives [5, 6], effects on the brain, the amyloid hypothesis predicts that
coumarins: Aegelinol benzoate, agasyllin, grandivittin, and decreased production of the A peptide in the brain,
aegelinol [7]. particularly, A 42 will elicit a therapeutic benefit [9].
Alzheimer's disease (AD) is the leading cause of In this work, we have discussed the isolation and
dementia in the elderly, presenting itself clinically by structure elucidation of eleven flavonoidal compounds from
progressive loss of memory and learning and its prevention the aqueous methanol extract of E. campestre aerial parts.
is a major public health challenge. The key event in the The evaluation of the Eryngo extract and the isolated
progression of AD is the sequential cleavages of -amyloid flavonol glycosides for their DPPH scavenging activity, and
precursor protein (-APP) by two proteolytic enzymes, beta- reducing power was carried out. The inhibition of the A 42
site APP-cleaving enzyme 1 (BACE-1 or memapsin 2) and and A 40 formation were measured in the culture medium
-secretase to produce A 40 and A 42 in the human brain of H4 cells, a human neuroglioma cell line expressing the
[8]. The main constituent of the accumulated amyloidic double Swedish mutation (K595N/M596L) of human APP
plaques in the brain is -amyloid peptide (A). The activity (APPsw). The inhibition of the cyclooxygenase activity was
of two aspartic proteases (-secretase and -secretase) results estimated as well for the plant extract.
in A of length from 38 to 42 amino acids. -Secretase MATERIAL AND METHODS
pathway degrades 90% of the APP rest is degraded by the
NMR spectra were obtained on Varian Unity 300 (1H,
300; 13C, 145 MHz) and Varian Inova 600 (1H, 599.744
*Address correspondence to this author at the Phytochemistry and Plant
MHz; 13C, 150.7 MHz) spectrometers. DMSO-d6 was used
Systematic Department, National Research Centre, El-Bohooth Street,
12311, Dokki, Cairo, Egypt; Tel 00202-37340064; Fax: 202-33370931; as solvent. The  values reported as ppm relative to TMS in
E-mail; hawasusama@yahoo.com DMSO-d6 and J values are given in Hz. UV spectra were
recorded on Shimadzu UV-visible spectrophotometer model-

1872-3136/13 $58.00+.00 © 2013 Bentham Science Publishers


Anti-Alzheimer, Antioxidant Activities and Flavonol Glycosides Current Chemical Biology, 2013, Vol. 7, No. 2 189

UV 240 (Tokyo, Japan). Polyamide 6S (Riedel de Haen AG, according to Brand-Williams (1995) [10]. 0.05 ml of extract
Hannover, Germany); Sephadex LH-20 (Pharmacia Fine or isolated compound was dissolved in DMSO then added to
Chemicals, Uppsala, Sweden); Solvent mixtures, BAW (n- 0.1 ml of freshly prepared methanolic DPPH solution (20 g
butanol:acetic acid:water, 4:1:5 upper phase), and paper ml-1). The mixture was shaken vigorously and allowed to
chromatography (PC), Whatman No. 1 & 3 MM (46  57 stand at room temperature for 30 min. The absorbance was
cm) (Kent, England). measured at 517 nm by spectrophotometer and compared
with a blank control. Scavenging of DPPH free radical was
Plant Material
measured using the following equation:
The aerial parts of Eryngium campestre L. were collected
Scavenging activity (%) = (control absorbance – sample
in April 2009 from Borg El-Arab city (50 km East of
absorbance)/ (control absorbance)  100.
Alexandria), Egypt. The plant was identified by Prof.
Ibrahium El-Garf, Botany Department, Cairo University, and Reducing Power Assay
voucher specimens have been deposited at the herbarium of
The reducing power of E. campestre extract, and the
the National Research Centre, Dokki, Cairo, Egypt.
isolated compounds was determined according to Oyizu
Extraction and Isolation [11]. The extract and isolated flavonoids were dissolved in
methanol and mixed with phosphate buffer (2.5 ml, 0.2 M,
Ground aerial parts of E. campestre (500 g) were
pH 6.6) and potassium ferrocyanide (2.5 ml, 1%). The
extracted three times with 70% aqueous methanol at room
mixture was incubated at 50°C for 20 min. A portion (2.5
temperature (each 2 L, 12 h). The extract was concentrated ml) of trichloroacetic acid (10%) was added to the mixture,
to give a residue (40 g), which was suspended in water and
which was then centrifuged at 3000 rpm for 10 min. The
then partitioned successively with dichloromethane and ethyl
upper layer of the solution (2.5 ml) was mixed with distilled
acetate. The ethyl acetate soluble fraction (15 g) was applied
water (2.5 ml) and FeCl3 (0.5 ml, 0.1%) and the absorbance
to a polyamide 6S column and eluted with H2O with the
was measured at 700 nm and compared with standards.
proportional increasing of MeOH. All fractions thus obtained
Increased absorbance of the reaction mixture indicated
were recombined according to their paper chromatographic increased reducing power.
analysis to give eight major fractions (I-VIII). Fraction I was
subjected to preparative paper chromatography (3 MM) Statistical Analysis
using 15% acetic acid/water as eluent. The separated bands
The antioxidant data were performed in triplicate and all
were scraped off and eluted with 70% methanol and further
data were expressed with mean standard deviations (SD).
purified by column chromatography (CC) on Sephadex LH-
Data obtained were subjected to standard analysis of
20 using MeOH to get compounds 1 (6 mg), 5 (7 mg) and 9
(11 mg). Compounds 2 (10 mg), 4 (10 mg) and 6 (7 mg), variance procedure where values of LSD were obtained at
0.05% as reported by Snedecore and Cochran [12].
were detected as major components in fractions II, III and
IV, respectively, and purified separately by repeated CC on Anti-Alzheimer activity
Sephadex LH-20 using MeOH and MeOH:H2O (8:2) as
eluents. Fraction V was purified on cellulose column with Chemicals
saturated n-BuOH as eluent to give pure compounds 3 (12 Dimethylsulfoxide (DMSO), nicotinamide adenine
mg) and 7 (8 mg), while compound 8 (23 mg) was dinucleotide (reduced form) NADH, phosphate buffered
spontaneously precipitated in the concentrated fraction VI. saline (PBS), hydrogen peroxide, fetal bovine serum,
Fractions VII and VIII, were combined and subjected to gentamicin sulfate, L-Glutamine,Triton X-100 3-(4,5-
preparative paper chromatography (3 MM) using BAW as dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
eluent followed by successively CC on Sephadex LH-20 (MTT), Dulbecco’s modified Eagle’s media (DMEM) and
with MeOH as eluent to afford pure samples of 10 (6 mg) trypan blue dye were purchased from Sigma Chemical
and 11 (7 mg). Company (St. Louis, MO, USA).
Acid Hydrolysis Culture of H4 Cells Expressing the Double Swedish
Compounds 1-11 (2 mg) in a mixture of 8% HCl (1 ml) Mutation (K595N/M596L) of Human APP (APPsw).
and MeOH (4 ml) were separately refluxed for 2 h. The All cell lines were cultured in DMEM (high glucose)
reaction mixtures were reduced under pressure to dryness, containing 9% heat-inactivated fetal calf serum, 100-Us/mL
dissolved in H2O (3 ml) and neutralized with Na2 CO3. The penicillin, 100- g/mL streptomycin, and 2mM L-glutamine.
neutralized products were subjected to PC (eluent, Stably transfected H4 cells were additionally supplemented
benzene:n-butanol:pyridine:H2O, 1:5:3:3). The with 200- g/mL G418 [13].
chromatograms were sprayed with aniline hydrogen
phthalate followed by heating at 100°C. The sugars were Exposure of H4 Cells to the Extract
identified after comparison with authentic samples. Cells were seeded onto 24-well plates (2x105 cell/well)
Anti-Oxidant Activity and allowed to grow to confluence for 24 h, in 5% CO2/95%
air in humidified atmosphere. Increasing concentrations
DPPH free Radical Scavenging Assay (from 0 - 300 ng/ml) of the extract were added to the cells
The free radical scavenging activity using the 1.1- overnight in a final volume of 0.5 ml. R-flurbiprofen was
diphenyl-2-picrylhyrazil (DPPH) reagent was determined used as positive control (3 - 1000 M). DMSO (0.5%) was
used as negative control. At the end of the incubation, 100 l
190 Current Chemical Biology, 2013, Vol. 7, No. 2 Hawas et al.

of each supernatant were removed for measuring -amyloid (Biorad, USA). The relative cell viability was expressed as
peptides. the mean percentage of viable cells compared to the
untreated control cells, with the half maximal inhibitory
Detection of A Peptides
concentration (IC50) calculated from the viability dose
Each supernatant was treated with a biotinylated mouse response curve of each cell line.
monoclonal antibody (4G8, Signet Laboratories Inc., Lactate Dehydrogenase (LDH) Cytotoxicity Assay
Dedham, MA, USA), specifically recognizing the 17-24
amino acid region of A and two rabbit polyclonal To determine the effect of the extract on membrane
antibodies (C-term 42 and C-term 40, BioSource permeability in H4 cells, a lactate dehydrogenase (LDH)
International, Camarillo, CA, USA), specifically recognizing release assay was used. The cells were seeded in 96-well
the C-terminus of A 42 and A40, respectively. Antigen– culture plates at a density of 2  104 cells/well in 100 l
antibodies complexes were recognized by TAG-donkey anti- volume and allowed to grow for 18 hours before treatment.
rabbit IgG (Jackson Immuno Research Laboratories, Soham, After treatment, the plates were incubated for 24, 48 and 72
UK). Streptavidin coated magnetic beads captured the hours, respectively. Then, the supernatant (40 l) was
complexes and the signals were read by an transferred to a new 96 well to determine LDH release and
electrochemiluminescence instrument (Origen M8 Analyzer, 6% triton X-100 (40 L) was added to the original plate for
BioVeris Corporation, Gaithersburg, MD, USA). A levels determination of total LDH. An aliquot of 0.1 M potassium
in cell supernatants were evaluated in comparison to an A phosphate buffer (100 L, pH 7.5) containing 4.6 mM
peptide Standard. The total A content was calculated as pyruvic acid was mixed to the supernatant using repeated
sum of A 40 and A 42. pipetting. Then, 0.1 M potassium phosphate buffer (100 L,
pH 7.5) containing 0.4 mg/ml reduced -NADH was added
Inhibition of COX-1 and COX-2 Assay
to the wells. The kinetic changes were read for 1 min using
The inhibition of the cyclooxygenase activity was ELISA microplate reader in absorbance at wavelength 340
estimated measuring by PGE2 production form arachidonic nm. This procedure was repeated with 40 l of the total cell
acid according to a modified method [14]. Recombinant lysate to determine total LDH. The percentage of LDH
human prostaglandin H2 synthase-1 (PGHS-1) and synthase- release was determined by dividing the LDH released into
2 (PGHS-2) were expressed in transfected spodoptera the media by the total LDH following cell lysis in the same
frugiperda (Sf-9) cells (Invitrogen, San Diego, CA, USA). well.
The microsomal fractions were prepared from the transfected
Statistical Analysis
cells and used to assay the enzymatic activities. Briefly, the
enzymes (2 g) reconstituted in a buffer (100 mM Tris-HCl, All experiments of Alzheimer were conducted in
pH 8.0) containing 2 mM phenol, were preincubated with triplicate (n = 3). All the values were represented as mean ±
vehicle (DMSO) or the extract (0 - 300 ng/ml in DMSO) for SD. Significant differences between the means of parameters
20 min at 22ºC. The reaction mixture was completed with as well as IC50s were determined by probit analysis using
1M hematin. The reaction was initiated adding arachidonic SPSS software program (SPSS Inc., Chicago, IL).
acid (4 and 2  for COX-1 and COX-2, respectively) and the
RESULTS
mixture was incubated for 5 min at 22ºC for COX-1 assay,
or for 10 min at 25ºC for COX-2 assay. For control Chemical Characterization of Isolated Compounds
measurements, arachidonic acid was omitted from the
reaction mixture. The reactions were stopped by the Elven flavonol compounds were isolated and identified
sequential addition of 1 M HCl and 1 M Tris-HCl (pH 8.0), as three quercetin glycosides, quercetin 3-O--glucoside (1),
followed by cooling to 4ºC. The amount of PGE2 present in quercetin 3-O--glucuronide-4'-methylether (2), and rutin
the reaction mixture was quantified using an enzyme- (3); five isorhamnetin glycosides, isorhamnetin 3-O--
immunoassay. glucoside (4), isorhamnetin 3-O--galactoside (5),
isorhamnetin 3-O--rhamnoside (6), isorhamnetin 3-O--
MTT Cytotoxicity Assay rutinoside (7), and isorhamnetin 3-O--glucoside-7-O--
rhamnoside (8); and three myricetin glycosides, myricetin 3-
The anti-proliferative activity against H4 human cell line
O--glucoside-3'-methylether (9), myricetin 3-O--
was estimated using the 3-[4,5-dimethyl-2-thiazolyl)-2,5-
glucoside-4'-methylether (10), and myricetin 3-O--
diphenyl-2H-tetrazolium bromide (MTT) assay, which is
galactoside-4'-methylether (11).
based on the cleavage of the tetrazolium salt by
mitochondrial dehydrogenases in viable cells [15]. Cells The structures of the isolated compounds were
were dispensed in a 96 well sterile microplate (5 x 104 established by means of a UV-visible spectrophotometer,
cells/well), and incubated at 37oC with various MS and NMR spectrometer as follows:
concentrations of the extract (0 - 300 ng/ml) or paclitaxel
(taxol, positive control) for 48 h in a serum free medium Quercetin 3-O--glucoside (1)
prior to the MTT assay. After incubation, media were Yellow powder, PC Rf 0.54 (BAW), 0.43 (15% HOAc).
carefully removed, 40 L of MTT (5 mg/ml) were added to UV max (MeOH): 257, 267sh, 324sh 360; (NaOMe): 272,
each well and then incubated for an additional 4 h. The 329, 409; (AlCl3): 272, 430; (AlCl3/HCl): 268, 359sh, 405sh;
purple formazan dye crystals were solubilized by the (NaOAc): 273, 324, 391; (NaOAc/H3BO3): 262, 379. 1 H
addition of 200 L of acidified isopropanol. The absorbance NMR:  = 7.53 (m, H-2'/6'), 6.79 (d, 8.4 Hz, H-5'), 6.34 (d,
was measured at 570 nm using a microplate ELISA reader 2 Hz, H-8), 6.14 (d, 2 Hz, H-6), 5.41 (d, 6.9 Hz, H-1).
Anti-Alzheimer, Antioxidant Activities and Flavonol Glycosides Current Chemical Biology, 2013, Vol. 7, No. 2 191

Quercetin 3-O--glucuronide-4'-methylether (2) Isorhamnetin 3-O-rutinoside (7)


Yellow powder, PC Rf 0.56 (BAW), 0.44 (15% HOAc). Yellow powder, PC Rf 0.42 (BAW), 0.46 (15% HOAc).
UV max (MeOH): 253, 266sh, 339, 356; (NaOMe): 274, UV max (MeOH): 254, 266sh, 300sh 352; (NaOMe): 272,
321, 388 (dec); (AlCl3): 269, 363, 396; (AlCl3/HCl): 268, 320, 408; (AlCl3): 274, 354, 399; (AlCl3/HCl): 274, 364,
360, 364, 371, 400; (NaOAc): 269, 339, 361; 395; (NaOAc): 275, 371; (NaOAc/H3BO3): 255, 339, 354.
(NaOAc/H3BO3): 255, 358. 1H NMR:  = 8.01 (d, 2 Hz, H-
1
H NMR:  = 7.85 (d, 2.2 Hz, H-2'), 7.45 (dd, 8 and 2.2 Hz,
2'), 7.50 (dd, 8.5 and 2 Hz, H- 6'), 6.9 (d, 8.5 Hz, H-5'), 6.4 H- 6'), 6.90 (d, 8 Hz, H-5'), 6.42 (d, 2 Hz, H-8), 6.20 (d, 2
(d, 2 Hz, H-8), 6.2 (d, 2 Hz, H-6), 5.6 (d, 7 Hz, H-1''), 3.81 Hz, H-6), 5.41 (d, 7.4 Hz, H-1''). 4.42 (s, H-1'''), 3.85 (s, 3'-
(s, 4'-OCH3). 13C NMR:  = 175 (C-4), 170.2 (C-6), 164 (C- OCH3), 0.98 (d, 6 Hz, CH3-Rh). 13C NMR:  = 177 (C-4),
7), 161.5 (C-2), 156.5 (C-5/9), 149 (C-4'), 146.5 (C-3'), 163 (C-7), 161.5 (C-5), 158.5 (C-9), 157 (C-2), 147 (C-3'),
132.5 (C-3), 121.8 (C-2'), 121 (C-6'), 115 (C-1'), 113 (C-5'), 146 (C-4'), 133 (C-3), 122 (C-1'), 121.5 (C-6'), 115.5 (C-5'),
105 (C-10), 101 (C-1), 98.5 (C-6), 93.7 (C-8), 75.9 (C-2), 113.5 (C-2'), 105.5 (C-10), 102.5 (C-1), 102 (C-1'''), 97 (C-
75.7 (C-3), 74.0 (C-5), 71.8 (C-4), 55.4 (4'-OCH3). 6), 95 (C-8), 79 (C-5), 76 (C-3), 73.7 (C-4), 73 (C-2), 71
(C-3), 70.2 (C-2), 69.5 (C-4), 68.1 (C-5), 65.5 (C-6),
56 (CH3O), 17.8 (C-6).
Rutin (3)
Isorhamnetin 3-O--glucoside-7-O--rhamnoside (8)
Yellow powder, PC Rf 0.40 (BAW), 0.47 (15% HOAc).
UV max (MeOH): 257, 267sh, 295sh 360; (NaOMe): 275, Yellow powder, PC Rf 0.43 (BAW), 0.45 (15% HOAc).
320, 415; (AlCl3): 269, 305sh, 335sh, 430; (AlCl3/HCl): 275, UV max (MeOH): 263, 267sh, 354; (NaOMe): 262, 419;
305sh, 345sh, 420; (NaOAc): 270, 320, 385; (AlCl3): 272, 358, 407; (AlCl3/HCl): 271, 402, 408;
(NaOAc/H3BO3): 260, 300sh, 380. 1H NMR:  = 7.50 (dd, (NaOAc): 256, 268, 361; (NaOAc/H3BO3): 255, 368, 361.
2/8 Hz, H-2'/6'), 6.80 (d, 8 Hz, H-5'), 6.35 (d, 2 Hz, H-8),
1
H NMR:  = 7.91 (d, 2.2 Hz, H-2'), 7.63 (d, 8 Hz, H- 6'),
6.15 (d, 2 Hz, H-6), 5.31 (d, 7.5 Hz, H-1''). 4.34 (s, H-1'''), 6.90 (d, 8 Hz, H-5'), 6.81 (d, 2 Hz, H-8), 6.43 (d, 2 Hz, H-6),
0.95 (d, 6 Hz, CH3-Rh). 13C NMR:  = 177.2 (C-4), 164 (C- 5.61 (m, H-1''/ H-1'''), 3.85 (s, 3'-OCH3), 1.12 (d, 6.3 Hz,
7), 161 (C-5), 156.3 (C-9), 156.1(C-2), 148.2 (C-4'), 144.7 CH3-Rh). 13C NMR:  = 177 (C-4), 161.5 (C-5), 161 (C-7),
(C-3'), 133.2 (C-3), 121.4 (C-6'), 121.0 (C-1'), 116.1 (C-5'), 156.5 (C-2), 156 (C-5), 149.5 (C-4'), 146.5 (C-3'), 133.1 (C-
115.1 (C-2'), 103.8 (C-10), 101(C-1), 100.6 (C-1'''), 98.6 3), 122.0 (C-1'), 121.0 (C-6'), 115.0 (C-5'), 113.5 (C-2'),
(C-6), 93.5 (C-8), 76.4 (C-5), 75.8 (C-3), 74.0 (C-2), 71.8 105.5 (C-10), 101(C-1), 99.0 (C-6), 94.3 (C-8), 98.5 (C-
(C-4), 70.4 (C-3), 70.2 (C-2), 69.9 (C-4), 68.1 (C-5), 1), 77.1 (C-5), 76.6 (C-3), 74.4 (C-2), 76.9 (C-3), 71.8
66.9 (C-6), 17.5 (C-6). (C-4), 70.6 (C-3), 70.3 (C-2), 70.2 (C-4), 61.0 (C-6),
55.0 (3'-OCH3), 17.8 (CH3-Rh).
Isorhamnetin 3-O--glucoside (4) Myricetin 3-O--glucoside-3'-methylether (9)
Yellow powder, PC Rf 0.56 (BAW), 0.39 (15% HOAc). Yellow powder, PC Rf 0.41 (BAW), 0.27 (15% HOAc).
UV max (MeOH): 257, 267sh, 324sh 357; (NaOMe): 272, UV max (MeOH): 253, 267sh, 356; (NaOMe): 275, 324,
329, 408; (AlCl3): 265, 385; (AlCl3/HCl): 265, 359sh, 405sh; 381; (AlCl3): 274, 365, 408; (AlCl3/HCl): 273, 357, 399,
(NaOAc): 273, 324, 391; (NaOAc/H3BO3): 262, 379. 1 H 409; (NaOAc): 274, 324, 391; (NaOAc/H3 BO3): 254, 267,
NMR:  = 7.51 (s, H-2'), 7.34 (d, 8.4 Hz, H- 6'), 6.85 (d, 8.4 379. 1H NMR:  = 7.59 (s, H-2'), 7.14 (s, H-6'), 6.42 (d, 2
Hz, H-5'), 6.22 (d, 2 Hz, H-8), 6.03 (d, 2 Hz, H-6), 5.51 (d, 7 Hz, H-8), 6.20 (d, 2 Hz, H-6), 5.58 (d, 6.8 Hz, H-1''). 3.72 (s,
Hz, H-1''), 3.78 (s, 3'-OCH3). 3'-OCH3). 13C NMR:  = 177 (C-4), 164.5 (C-2), 161.0 (C-
7), 156 (C-5/9), 148 (C-4'), 145.7 (C-5'), 138 (C-3'), 133 (C-
Isorhamntin 3-O--galactoside (5) 3/2'), 129 (C-6'), 121.0 (C-1'), 121.8 (C-6'), 103.5 (C-10),
101 (C-1), 99.2 (C-6), 93.5 (C-8), 77.4 (C-3), 76.3 (C-5),
Yellow powder, PC Rf 0.46 (BAW), 0.35 (15% HOAc). 74.2 (C-2), 69.8 (C-4), 62.7 (C-6), 55 (3'-OCH3).
UV max (MeOH): 257, 267sh, 324sh 359; (NaOMe): 272,
329, 408; (AlCl3): 265, 385; (AlCl3/HCl): 265, 359sh, 405sh; Myricetin 3-O--glucoside-4'-methylether (10)
(NaOAc): 273, 324, 391; (NaOAc/H3BO3): 262, 379. 1 H Yellow powder, PC Rf 0.56 (BAW), 0.32 (15% HOAc).
NMR:  = 7.99 (d, 2 Hz, H-2'), 7.45 (dd, 8.4 and 2 Hz, H- UV max (MeOH): 253, 267sh, 357; (NaOMe): 275, 324,
6'), 6.88 (d, 8.4 Hz, H-5'), 6.39 (d, 2 Hz, H-8), 6.15 (d, 2 Hz, 381(dec); (AlCl3): 277, 365, 412; (AlCl3/HCl): 276, 357,
H-6), 5.52 (d, 6.9 Hz, H-1''), 3.79 (s, 3'-OCH3). 399, 412; (NaOAc): 274, 324, 391; (NaOAc/H3BO3): 254,
267, 379. 1H NMR:  = 7.53 (s, H-2'/6'), 6.45 (d, 2 Hz, H-8),
Isorhamnetin 3-O--rhamnoside (6) 6.20 (d, 2 Hz, H-6), 5.62 (d, 6.8 Hz, H-1''). 3.85 (s, 4'-
OCH3).
Yellow powder, PC Rf 0.55 (BAW), 0.33 (15% HOAc);
UV max (MeOH): 257, 268, 358; (NaOMe): 272, 324, Myricetin 3-O--galactoside-4'-methylether (11)
420; (AlCl3): 268, 301, 360, 398; (AlCl3/HCl): ) 262, 302,
Yellow powder, PC Rf 0.55 (BAW), 0.32 (15% HOAc).
402; (NaOAc): 270, 322, 408; (NaOAc/H3 BO3): 256, 266,
UV max (MeOH): 254, 266sh, 339, 358; (NaOMe): 273,
362. 1H NMR:  = 7.91 (d, 2 Hz, H-2'), 7.53 (d, 8.0 Hz, H-
324, 422 (dec); (AlCl3): 273, 306, 396, 403; (AlCl3/HCl):
6'), 6.86 (d, 8.0 Hz, H-5'), 6.41 (d, 2.3 Hz, H-8), 6.18 (d, 2.3 273, 306, 358, 400; (NaOAc): 274, 324, 366, 422;
Hz, H-6), 5.53 (br s, H-1''), 3.81 (s, 3'-OCH3), 1.07 (d, 6.1 (NaOAc/H3BO3): 255, 339, 361. 1H NMR:  = 7.52 (s, H-
Hz, CH3-Rh).
192 Current Chemical Biology, 2013, Vol. 7, No. 2 Hawas et al.

100
90

Scavenging Activity (%)


80
70
60
50
40
30
20
10
0
Extract 1 2 3 5 6 7 8 9 10 11 BHT
Compounds

*LSD 0.05%=0.914
Values are means ±S.D. (n=3).

Fig. (1). DPPH radical-scavenging activities (%) of E. campestre extract and isolated compounds (0.5 mg/ml).

2.5

2
Absorbance(700nm)

1.5

0.5

0
Extract 1 2 3 5 6 7 8 9 10 11 BHT

Compounds

*LSD 0.05%=0.105
Values are means ±S.D. (n=3)

Fig. (2). Reducing Power of E. campestre extract and isolated compounds (0.5 mg/ml).

2'/6'), 6.42 (d, 2 Hz, H-8), 6.17 (d, 2 Hz, H-6), 5.58 (d, 6.5 power (1.8) compared to the isolated flavonol glycosides 1-3
Hz, H-1''), 3.83 (s, 4'-OCH3). and 5-11. Quercetin 3-O--glucuronide-4'-methylether (2),
isorhamnetin 3-O--glucoside-7-O--rhamnoside (8) and
DPPH Free Radical Scavenging Activity
rutin (3) showed relatively similar reducing power activity
The stable radical DPPH has been used widely for the (1.47, 1.43 and1.34 absorbance at 700 nm), respectively.
determination of primary antioxidant activity. The assay is
Anti-Alzheimer Activity
based on the reduction of DPPH radicals in methanol which
causes an absorbance drop at 517nm. As shown in Fig. (1), In this study we examined whether the extract E.
the antioxidant activity of E. campestre extract exhibited campestre alters APP processing and generation of A,
relatively high DPPH free radical scavenging activity particularly the A 42 species. H4 human neuroglioma cells
(66.3%). Among the isolated compounds, rutin (3) showed expressing the double Swedish mutation (K595N/M596L) of
the highest DPPH free radical scavenging activity (56.2 %) human APP (APPsw) were used and treated with increasing
followed by isorhamnetin 3-O--glucoside 7-O-- concentrations of the E. campestre extract, and analysed for
rhamnoside (8) and isorhamnetin 3-O--galactoside (5) with A 40 and A 42 levels in culture medium by sensitive
55.1% and 51.1% free radical scavenging activities, sandwich enzyme link immunosorbent assay (ELISA).
respectively, whereas myricetin 3-O--glucoside-3'-
In H4 cells, a 73% reduction in the A 42 supernatant
methylether (9) exhibited the lowest DPPH free radical
scavenging activity (20.53%). concentration was achieved at concentration of 300 ng/ml
without significant reduction in total A (A 40 + A 42)
Reducing Power level. A dose-dependent inhibition of A 42 secretion was
observed (Fig. 4) with IC50 of (155.75 ± 7.43 ng/ml). No
Fe (III) reduction is often used as an indicator of toxicity was detected by both standard MTT assay and LDH
electrondonating activity, which is an important mechanism assay in H 4 cells treated with extract concentration up to
of phenolic antioxidant action [16]. From Fig. (2), it was 500 ng/ml. In addition, the inhibition activity of different
found that E. campestre extract revealed significant reducing concentrations of the extract against COX-1 and COX-2 was
Anti-Alzheimer, Antioxidant Activities and Flavonol Glycosides Current Chemical Biology, 2013, Vol. 7, No. 2 193

also investigated. The results showed no inhibition activity showed only the signals of A- and B-ring, while in 13C NMR
of the extract against COX-1 and COX-2 up to 400 ng/ml spectra the characteristic carbon resonance of flavonol C-3
concentration. (C 133-135 ppm) was observed. 2D-NMR experiments were
also used to confirm the glycosidic linkage in proposed
DISCUSSION
structures of flavonol diglycosides (3, 7 and 8) as well as the
Dry aerial parts of E. campestre were extracted with 70% di-substituted flavonols (2, 9 and 10).
MeOH then subjected to successive fractional partition by
Phenols are very important plant constituents because of
chloroform and ethyl acetate. The ethyl acetate was
their scavenging ability due to their hydroxyl groups [18].
chromatographed on a polyamide column followed by
The aqueous methanol extract of E. campestre showed the
repeated CC on Sephadex LH-20 or cellulose and
highest scavenging activity against DPPH. This activity
preparative paper chromatography, to obtain the pure eleven
attributed to the presence of flavonoid contents of this
flavonoids 1-11. species. In this study, chemical structure-activity has shown
All compounds appeared as dark purple spots on PC that quercetin diglycoside (3) are a potent peroxyl radical
under UV light, changing to yellow when exposed to scavenger followed by O-methylated derivatives (5). This
ammonia vapor, except compounds 2 and 10 where no finding is in accordance with the results reported by Bors
change in color was observed which indicates flavonoids 2 and co-works (1990) [19] and confirms that the O-dihydroxy
and 10 with a free 5- and substituted 4´-hydroxyl groups structure in the B-ring is important for enhanced radical
[17]. Complete acid hydrolysis for isolated flavonol O- scavenging activity. It has been demonstrated that the spin
glycosides was carried out, and followed by paper co- distribution during oxidation of quercetin remain entirely on
chromatography with authentic samples to identify the the B-ring favouring the donation of two electrons leading to
hydrolytic products whether flavonol aglycons and sugar the formation of an ortho-quinone. This has also been related
moieties. to the total conjugation of quercetin molecule over the B and
C rings. Flavonols containing a 4'-monohydroxyl group are
The UV spectra of the isolated compounds 1-11 in less potent antioxidant, the mechanism of action probably via
MeOH are characteristic of flavonols showing an intense formation of a phenoxyl radical.
band I between 352-361 nm. The bathochromic shift of 42-
55 nm observed for band Ia after addition of the reagent Different studies have indicated that the electron
AlCl3/ HCl indicates 5-hydroxy-3-substituted flavonols. The donation capacity reflects the reducing power of bioactive
absence of a band II bathochromic shift in the presence of compounds is associated with antioxidant activity. In this
NaOAc is in agreement with a 7-O-substitution as proposed study, the reducing power assay using Fe (III) reduction as
for flavonol diglycoside (8). A band I hypsochromic shift an indicator of electron donating showed the presence of
with low intensity after addition of NaOMe was observed for antioxidants in the sample reduced the Fe (III) to Fe (II) by
flavonols 2 and 10. The 1H NMR spectra of flavonols 1-11 donating an electron. Amount of Fe (II) complex can be then

OH
CH3
O
OR1
OH

HO O
R1O O

OR2 OR2

OH O OH O

1: R1 = H, R2 = Glucose 4: R1 = H, R2 = Glucose
2: R1 = CH3, R2 = Glucuronic acid 5: R1 = H, R2 = Galactose
3: R1= H, R2 = Rutinoside 6: R1 = H, R2 = Rahmnose
7: R1 = H, R2 = Rutinoside
8: R1 = Rhamnose, R2 = Glucose
OH

O OH
CH3
O
CH3
HO O
OH
HO O
OH
OH

O
O
OH OR
OH O
OH OH
OH O

9 10: R = Glucose
11: R = Galactose
Fig. (3).
194 Current Chemical Biology, 2013, Vol. 7, No. 2 Hawas et al.

Fig. (4). Analysis of A from H4 human neuroglioma cultured cells treated with the E. campestre extract by ELISA. A 42 and total A
levels (A 40 + A 42 values) were normalized to values obtained from vehicle-treated cells. Treatment with extract preferentially reduced
A levels in the medium of H4 human neuroglioma cells in a dose-dependent manner. Data are shown as the mean ± s.d. of all experiments.

be monitored by measuring the formation of Perl’s Prussian ACKNOWLEDGEMENTS


blue (Fe4[-Fe(CN)6]3) at 700 nm. The methanol extract of
Declared none.
E. campestre and the isolated flavonol glycosides 1-3 and 5-
11 exhibited moderate reducing power comparable with that
of standard antioxidant butylated hydroxy toluene (BHT). REFERENCES
[1] Wörz A. On the distribution and relationships of the South-West
In this study, we describe the activity of E. campestre Asian species of Eryngium L. (Apiaceae-Saniculoideae). Turk J
extract that reduced A production by functionally inhibiting Bot 2004; 28: 85-92.
-secretase. Therefore, we examined whether the extract [2] Clapham AR, Tutin TG. Flora of the British Isles. 1st ed.
alters APP processing and generation of A, particularly the Cambridge: Cambridge University Press 1952.
A 42 species using H4 human neuroglioma cells expressing [3] Gruenwald J, Brendler T, Jaenicke C. PDR for Herbal Medicines.
2nd ed. New Jersey: Medical Economics Company 2000; 729-33.
the double Swedish mutation (K595N/M596L) of human [4] Kartal M, Mitaine-Offer AC, Paululat T, Abu-Asaker M, Wagner
APP (APPsw). The results indicated that this extract has H, Mirjolet JF, et al. Triterpene saponins from Eryngium
reduced the A 42 level at very low concentration without campestre. J Nat Prod 2006; 69(7): 1105-8.
significant reduction in total A (A 40 + A 42) levels. [5] Kartnig T, Wolf J. Flavonoids from the Aboveground Parts of
Eryngium campestre. Planta Med 1993; 59(3): 285.
The cytotoxicity of E. campestre extract was assessed, in [6] Hohmann J, Páll Z, Günther G, Máthé I. Flavonol acyl glycosides
of the aerial parts of Eryngium campestre. Planta Med 1997; 63(1):
order to insure that A 42 reduction is not related to 96.
cytotoxicity. Furthermore, the results showed no inhibition [7] Erdelmeier CAJ, Sticher O. Coumarin derivatives from Eryngium
activity of the extract against COX-1, COX-2, and campestre. Planta Med 1985; 51: 407-9.
concomitant reduction in prostaglandin synthesis, indicating [8] Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease.
Lancet 2006; 368: 387-403.
that the reduction in A 42 levels may be independent of [9] Schmidt B, Braun HA, Narlawar R. Drug development and PET-
COX activity. Therefore, the E. campestre extract may diagnostics for Alzheimer's disease. Curr Med Chem 2005; 12:
affects only the secretion of the A peptide without 1677-95.
significantly affecting the secretion of either  and  forms [10] Brand-Williams W, Cuvelier ME, Berset C. Use of a free radical
method to evaluate antioxidant activity. LWT Food Sci Technol
of APP as the total A level did not significantly changed. 1995; 28: 25-30.
This finding suggests that the pharmacological action of E. [11] Oyaizu M. Studies on products of browning reactions: antioxidant
campestre extract is via the functional inhibition of - activities of products of browning reaction prepared from glucose
secretase. The results obtained indicate that E. campestre amine. Jap J Nutr 1986; 44: 307-15.
[12] Snedecor GW, Cochran WG. Statistical Methods. 7th Edn. Ames.
extract may be used as a potent candidate drug for the Iowa: Lowa State University Press 1980.
treatment of Alzheimer's disease (AD) or at least ameliorate [13] Haugabook SJ, Yager DM, Eckman EA. High throughput screens
or delay the bad effect of this disease. However, further in for the identification of compounds that alter the accumulation of
vivo and pre-clinical studies will be necessary to assess the Alzheimer's amyloid beta peptide (Abeta). J Neurosci Methods
2001; 108:171-79.
whether this extract is capable of altering -amyloidosis and [14] Glaser K, Sung Ml, O'Neill K, Belfast M, Hartman D, Carlson R.
neuropathology upon prolonged treatment. Etodolac selectively inhibits human prostaglandin G/H synthase 2
(PGHS-2) versus human PGHS-1. Eur J Pharmacol 1995; 281:
107-11.
CONFLICT OF INTEREST STATEMENT [15] Hansen MB, Nielsen SE, Berg K. Re-examination and further
development of a precise and rapid dye method for measuring cell
The author(s) confirm that this article content has no growth /cell kill. J Immunol Methods 1989; 119: 203-10.
conflict of interest. [16] Nabavi SM, Ebrahimzadeh MA, Nabavi SF, Fazelian M, Eslami B.
In vitro antioxidant and free radical scavenging activity of
Anti-Alzheimer, Antioxidant Activities and Flavonol Glycosides Current Chemical Biology, 2013, Vol. 7, No. 2 195

Diospyros lotus and Pyrus boissieriana growing in Iran. tannins and related polyphenols on superoxide anion radical and on
Pharmacognosy Magazine 2009; 4(18): 123-27. DPPH radical. Chem Pharm Bull 1989; 37: 2016-21
[17] Harborne JB, Mabry TJ, Mabry H. The Flavonoids. London: [19] Bors W, Heller W, Michel C, Saran M. Flavonoids as antioxidants:
Chapman and Hall 1975. determination of radical scavenging efficiencies. Methods enzymol
[18] Hatano T, Edamatsu R, Mori A, Fujita Y, Yasuhara E. Effect of 1990; 186: 343-55.
interaction of tannins with co-existing substances. VI. Effect of

Received: June 07, 2012 Revised: July 31, 2012 Accepted: August 01, 2012

View publication stats

You might also like