You are on page 1of 8

Phytochemistry Letters 10 (2014) 152159

Contents lists available at ScienceDirect

Phytochemistry Letters
journal homepage: www.elsevier.com/locate/phytol

Phytochemical prole of Juniperus oxycedrus ssp. oxycedrus berries:


A new monoterpene glucoside and evaluation of the effects on
cancer cell lines
Simona De Marino a, Carmen Festa a, Franco Zollo a, Fabiola Rusolo b, Francesca Capone b,
Eliana Guerriero b, Susan Costantini b, Vincenzo De Felice c, Maria Iorizzi c,*
a

Dipartimento di Chimica delle Sostanze Naturali, Universita` degli Studi di Napoli Federico II, Via D. Montesano 49, I-80131 Napoli, Italy
Centro Ricerche Oncologiche di Mercogliano, Istituto Nazionale per lo Studio e la Cura dei Tumori Fondazione Giovanni Pascale, IRCCS, Italy
c
Dipartimento di Bioscienze e Territorio, Universita` degli Studi del Molise, Contrada Fonte Lappone, I-86090 Pesche, Isernia, Italy
b

A R T I C L E I N F O

A B S T R A C T

Article history:
Received 14 July 2014
Received in revised form 20 August 2014
Accepted 24 August 2014
Available online 4 September 2014

Phytochemical investigation of polar extract from Juniperus oxycedrus spp. oxycedrus berries leds to the
isolation of one new monoterpene glucoside (3R,6E)-3,7 dimethyl 8-hydroxy-6-octenoic acid 8-O-b-Dglucopyranoside along with seven known components, some of them were initially isolated from
Juniperus communis L. berries. Their structures were established on the basis of extensive 1D and 2D NMR
(1H, 13C, COSY, HMBC, HSQC, ROESY) and ESI-MS studies. The n-butanol fraction and isolated
components, shikimic acid (2), compound 3, 4 and 5 were evaluated, in vitro, for their effect on cell
viability against human malignant melanoma (A375), breast (MCF-7) and lung (H460) cancer cell lines.
Shikimic acid exhibited selective effect on cell viability only against breast MCF-7 cell lines reaching IC50
value at dose of 30 mM and also induced the level decrease of vascular endothelial growth factor (VEGF)
and ve pro-inammatory cytokines suggesting its potential anti-inammatory effect.
2014 Phytochemical Society of Europe. Published by Elsevier B.V. All rights reserved.

Keywords:
Juniperus oxycedrus ssp. oxycedrus L.
Monoterpene glucoside
NMR
Viability evaluation
Cytokinome prole
Anti-inammatory capacity

1. Introduction
Juniperus oxycedrus L. (Cupressaceae) is one of 10 species in the
genus Juniperus throughout the world (Adams, 1998). This shrubs
or small tree grows wild in stony places of the Mediterranean and
Near East countries and is one of the most appreciate plants for its
essential oil richness and for its biologically active components
extensively used in folk medicine. Phytochemical studies carried
out on Juniperus taxa demonstrated the presence of a wide array of
secondary metabolites with a variety of pharmacological effects
(Moreno et al., 1998). Juniper berries have long been used as avors
to aromatize alcoholic beverages such as the famous gin or
aromatic grappa and, in northern Europeae, particularly in
Scandinavian cuisine are used to impart a sharp, clear avour
to meat dishes, especially wild birds and game meats (Montagne,
1999). J. oxycedrus was utilized since the antiquity to produce a
sesquiterpene-rich oil named cade oil obtained by destructive
distillation of branches and wood of the plant. Cade oil, also known

* Corresponding author. Tel.: +39 0874 404100; fax: +39 0874 404123.
E-mail address: iorizzi@unimol.it (M. Iorizzi).

in pharmacy as juniper tar, is an empyreumatic oil widely


employed in human and veterinary dermatology to treat chronic
eczema and other skin diseases (Chalchat et al., 1990). Cade oil has
also keratolytic and antipruritic properties (Leung and Foster,
1996) and rectied juniper tar was used as fragrance component
in soaps, detergents, creams and parfumes (Leung and Foster,
1996). J. oxycedrus has an extensively history of use in global folk
medicine for various disorder, such as hyperglycaemia, bronchitis
and pneumonia (Sanchez de Medina et al., 1994), decoction of the
plant is also used in traditional medicine in Spain as mouth
analgesic and for stomach disorders (Fernandez et al., 1996), in
Turkish folk medicine the boiled fruit extract has widely been used
in the treatment of gastrointestinal disorders, common colds, and
as diuretic (Akkol et al., 2009), while the resin was used for wound
healing (Yesilada et al., 1993).
Aqueous and methanol extracts of J. oxycedrus leaves showed in
vitro antimicrobial activity (Karaman et al., 2003) while methanol
and dichloromethane extracts of leaves and stems have been found
to reduce the blood pressure of normotensive rats (Bello et al.,
1997), to inhibit the response to histamine, serotonin and
acetylcholine (Moreno et al., 1998) and possess analgesic
properties (Moreno et al., 1998). The methanolic extracts of fruits

http://dx.doi.org/10.1016/j.phytol.2014.08.015
1874-3900/ 2014 Phytochemical Society of Europe. Published by Elsevier B.V. All rights reserved.

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

153

coumpound 2, (iv) a slowdown in cell cycle G0/G1 phase respect


in MCF-7 cells after treatment with compound 2 to untreated cells,
and (v) the compound 2 induced the level decrease of VEGF and
other ve pro-inammatory cytokines suggesting its potential
anti-inammatory effect.

and leaves exhibited antinociceptive and anti-inammatory


activities (Akkol et al., 2009), antioxidant (Lizzo et al., 2007) and
antifungal activities have been also reported (Amri et al., 2013).
Recently, polyphenolic fraction from J. oxycedrus badia exhibited acethylcholinesterase inhibitory activity and protected
neurons from injury in the neurodegeneration cell model (Tavares
et al., 2012).
There are many reports on the chemical composition of the oils
from leaves (Chalchat et al., 1990; Adams et al. 1999), berries (Boti
et al., 2006) and less frequently wood (Barrero et al., 1993) of
J. oxycedrus, however intraspecic variation in the chemical components of these essential oil was observed according to harvesting time,
geographical origin and starting material. Concerning leaf oil of
J. oxycedrus, chemical studies have reported in varying details the
composition from different geographic areas as from Corsica (Boti
et al., 2006), Croatia (Milos and Radonic, 2000), Spain (Adams, 1998),
Italy (Valentini et al., 2003), Tunisia (Medini et al., 2010), Greece (Stassi
et al., 1995), Lebanon (Lizzo et al., 2007), Turkey (Sezik et al., 2005).
Most of the study reported in literature up to now on J.
oxycedrus are focused on the chemical composition of the berrys
essential oil and limited data are reported on non-volatile
components. The methanol extract from ripe berries of J. oxycedrus
from Turkey was recently tested on cancer cell viability by using a
human hepatocellular liver carcinoma (HepG2) cell line (Taviano
et al., 2013). In previous works we have analyzed the seasonal
variations in the metabolic ngerprint of the J. communis berries
(Falasca et al., 2014) and the ability of imbricatolic acid to prevent
cell cycle progression in human lung tumor cells (CaLu-6) (De
Marino et al., 2011).
As a part of our ongoing investigations toward the discovery of
bioactive secondary metabolites from plants origin, one new
monoterpene glucoside (1) and seven known components were
isolated.
Herein, we also report the evaluation of effects of crude
n-butanol extract and its pure compounds 2, 3, 4 and 5 on three
human cancer cell lines, A375 (malignant melanoma), MCF-7,
(breast), H460 (lung). In overall the experiments performed on
cells showed that: (i) the n-butanol extract decreased the cell
viability only in MCF-7 cells, (ii) the compound 2 had effect on
MCF-7 viability at the concentration of 30 mM, (iii) no signicant
increase of apoptosis in MCF-7 cells after treatment with

2. Results and discussion


Natural products obtained mainly from plants have been used
as prominent source of phytochemicals that have been shown to
exert a wide range of biological activity.
The methanolic crude extracts of J. oxycedrus ssp. oxycedrus
berries were submitted to extractive and chromatographic
analyses. Four extracts were obtained using n-hexane, CHCl3,
n-BuOH and water following the modied Kupchans partitioning
procedure (Kupchan et al., 1973).
Purication of the n-buOH fractions let to the isolation of one
new monoterpene glucoside (1), with four known ones 25
identied as shikimic acid (2) (Ali et al., 2009), the monoterpene
glucoside (3) previously isolated from J. communis var. depressa
(Nakanishi et al., 2005), the n-butyl b-D-fructofuranoside (4) (Hu
and Chen, 1997) and the phenyl propanoid rhamnoside (5) isolated
from Pinus massoniana Lamb. (Lundgren et al., 1985) and from
Chamaecyparies obtusa Sieb. et Zucc. (Matsubara et al., 1990)
(Fig. 1). Their structures were elucidated using chemical and
spectroscopic methods including 1D and 2D NMR experiments and
spectrometric techniques.
2.1. Structural elucidation
Compound 1 was obtained as an amorphous powder and
exhibited a negative optical rotation ([a]D25 5.7, c. 0.12, MeOH).
In the positive and negative ESI-MS quasimolecular ion peaks were
observed at m/z 371 [M+Na]+ and m/z 347 [M] respectively. Its
molecular formula was determined as C18H28O8 based on the
positive-mode HRESI-MS analysis (m/z 371.1679 [M+Na]+; calc. for
C18H28O8Na 371.1676) and 13C NMR data. The 1H NMR spectrum
(Table 1) exhibited signals due to two methyl groups (dH 0.95 and
1.70), one olenic proton at dH 5.50, one methine, three methylene
and one primary alcoholic function resonating at dH 4.05 and 4.20.
Furthermore, one anomeric signal at dH 4.26 (d, J = 7.8 Hz) was

Table 1
1
H and 13C NMR data (CD3OD, 500 and 125 MHz) of compound 1 and selected signals in the (R)- and (S)-PGME amide of 1.
Position
1
2
3
4
5
6
7
8
9
10
Glucose
10
20
30
40
50
60
a

Signals overlapped.

dH (J in Hz)

dC

HMBC

1.96 m
2.19 m
1.95 m
1.24 m
1.41 m
2.09 m
2.07 m
5.50 br t (6.6)

4.05 d (11.7)
4.20 d (11.7)
0.95 d (6.1)
1.70 s

181.7
46.9

C-1, C-3, C-9

4.26 d (7.8)
3.19 dd (7.8, 8.3)
3.34a
3.26a
3.24a
3.66 dd (11.9, 5.4)
3.86 dd (11.9, 2.3)

102.1
74.8
77.8
71.5
77.5
62.6

(R)-PGME amide

(S)-PGME amide

5.475 (br t)

5.487 (br t)

C-2, C-3, C-4


C-6, C-7, C-8

0.934 (6.2)
1.731 s

0.893 (6.2)
1.746 s

C-8

4.272 (7.7)

4.271 (7.7)

31.6
37.9
26.0
130.5
132.4
75.5
19.9
13.8

C-4, C-6, C-7


C-8
0

C-6, C-7, C-1 , C-10

154

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

recognized with a large vicinal coupling constant (3JH1,H2)


indicating the trans-diaxial orientation (b-conguration). The
13
C NMR spectrum of 1 showed signals due to b-glucose unit and
one signal for a CO group (dC 181.7). The 1H1H-COSY implied
connectivity in the alkylic chain, from H2-2 to H-6, an isolated spin
system for H2-8 and CH3-10, together with signals of a Dglucopyranose. The HMBC experiment showed correlations
reported in Table 1 and indicated the glycosidic linkage at C-8
position, claried with a cross peak between H-10 (dH 4.26) and C-8
(dC 75.5). The double bond between C-6 and C-7 was E-congured
based on a NOESY cross-peak observed between H-6 (dH 5.50) and
H2-8 at dH 4.20 and dH 4.05. NOESY was also observed for CH3-10
(dH 1.70) with H-5 at dH 2.07 (Fig. 1).
Acidic hydrolysis of 1 afforded D-glucose which was conrmed
by GCMS analysis (Hara et al., 1987). The absolute conguration
at C-3 in 1 was determined by NMR methodology (PGME method);
1 was converted into its (R)- and (S)-phenylglicine methyl ester
(PGME) amides which enabled the determination of the absolute
conguration of b,b disubstituted carboxylic acids (Yabuuchi and
Kusumi, 2000).
The Dd [d (R-PGME amide)-d (S-PGME amide)] values obtained
from the 1H NMR spectra of the PGME amides (Table 1) suggested
that the absolute conguration at C-3 in 1 was R and was
elucidated as shown in Fig. 2. These data were in accordance with
the presence of (3R,6E)-3,7-dimethyl-8-hydroxy-6 octenoic acid 8O-b-D-glucopyranoside.
In the present study we also characterized four known labdane
diterpenoids from n-hexane and chloroform extracts. It is
especially noteworthy that the isolated metabolites of the J.
oxycedrus, are similar to those identied in the corresponding
extracts from berries of J. communis, a plant which grows in the
same geographical area in the central Italy. The n-hexane soluble
fraction was puried by chromatographic separation yielding the
known labdane diterpenoids cis-communic acid, myrcecommunic
acid, sandarocopimaric acid (De Marino et al., 2011). The
purication of CHCl3 soluble fraction gave the natural imbricatolic
acid (De Marino et al., 2011). Labdane diterpenes, isolated from
several plants families, show a variety of biological activities

(Demetzos and Dimas, 2001) including the inhibition of cell


proliferation in several cell lines (Dimas et al., 2006).
2.2. Colorimetric assay with sulforhodamine B, analysis of apoptosis
and cell cycle
Cancer chemotherapy regimen makes use of cytotoxic compounds that target proliferating cell populations and most of
cytotoxic drugs are from plants origin.
The cell viability of several cancer lines of melanoma (A375),
breast (MCF-7) and lung (H460) was determined after 24 h
stimulation with crude n-butanol extract by colorimetric assay
(sulforhodamine B) to identify the IC50 concentration, corresponding to the chemical extract amount that causes 50% inhibition of
cell growth (Fig. 3). However, it is important to underline that
rstly we performed the tests after 24, 36, 48 and 60 h and the
results were perfectly conserved. Hence we reported in this paper
the results obtained after 24 h in according to other our recent
papers (Rusolo et al., 2013; Costantini et al., 2014). Moreover, since
the solvent can be not healthy for the cells, we tested the effect of
only n-butanol on three cancer cell lines and veried that it had not
biological effect on cells and the related cell viability remained
equal to 100%. Therefore the cellular viability of untreated cells
was used as control.
The decrease in cell viability became signicant only for breast
cancer cell line, MCF-7, reaching IC50 at the concentrations of
200 mg. Then, we tested on MCF-7 cells the pure compounds 2, 3, 4
and 5. Only compound 2, shikimic acid, as evidenced in Fig. 4,
showed a signicant decrease in cell viability of reaching IC50 value
at dose of 30 mM. Compound 1 has not been tested because its
amount was not enough for the test.
In the next step we evaluated the ability of compound 2 to
induce apoptosis in MCF-7 and has been calculated the rate of
apoptosis by identication of four populations:
1) live cells which are not undergoing to apoptosis: annexin V ()
and cells dead marker ();
2) cells in early apoptosis: annexin V (+) and cells dead marker ();

Fig. 1. Natural compound isolated from J. oxycedrus ssp. oxycedrus L.

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

155

Table 2
Rate of apoptosis (A) and cell percentages in G0/G1, S and G2/M phases (B)
evaluated in MCF-7 cells, non treated and treated for 24 h, with 30 mM of compound
2A.
Cells

Fig. 2. Dd values [Dd (in ppm) dR  dS] obtained for the (R)- and (S)-PGME amides
of 1.

Fig. 3. Cell viability determined after 24 h stimulation with J. oxycedrus n-butanol


extract on A375, MCF-7 and H460 cells and estimated as % compared to untreated
cells.

Fig. 4. Cell viability determined after 24 h stimulation with different concentrations


of the compounds 2, 3, 4 and 5 on MCF-7 cells and estimated as % compared to
untreated cells.

3) cells in late apoptosis: annexin V (+) and cells dead marker (+);
4) dead cells which do not cross the apoptotic process: annexin V
() and cells dead marker (+).
In details, the cells dead marker used is 7-Aminoactinomycin D
(7-AAD).
The treatment with compound 2 did not show signicant
increase of apoptosis in MCF-7 (Table 2A). This was shown by a
density increase of cells in the plot of live cells and the presence of
few cells in early apoptosis/dead and, even further, in late
apoptosis.
We also evaluated the cell cycle in MCF-7 cells treated with
compound 2. The treatment showed a slowdown in cell cycle
G0/G1 phase respect to untreated cells (Table 2B) with a slight
lengthening of G0/G1 phase peak and a shortening of S phase
peaks. In fact, the cell percentages in G0/G1, S and G2/M phases
were 48.5%, 32.5% and 17.4% in untreated cells and 56.5%, 21.3%
and 19.2% in treated cells.
2.3. Cytokine level evaluation on MCF-7 supernatants
We also evaluated the cytokine orescence intensities in MCF-7
supernatants after treatment with compounds 2, 3, 4 and 5 by

A
MCF-7 non treated
MCF-7 treated

Live
cells

Cells in
early
apoptosis

Cells in late
apoptosis

Dead
cells

95.40%
92.31%

0.48%
3.40%

1.89%
2.78%

2.23%
1.51%

Cells

G0/G1

G2/M

B
MCF-7 non treated
MCF-7 treated

48.5%
56.5%

32.5%
21.3%

17.4%
19.2%

Human Cytokine 27-Plex Panel. The results obtained were


compared with untreated cells used as control. These experiments
showed that the cytokine levels in the supernatants after
treatment with compounds 3, 4 and 5 were similar to those in
control cells whereas the levels of VEGF and ve pro-inammatory
cytokines, like IL-8, IL-12, IP-10 (CXCL10), RANTES (CCL5) and
MCP-1 (CCL2) decreased in dose dependent way at increasing
levels of compound 2 (Table 3 and Fig. 5).
The cytokines IL-8 and VEGF promote tumor angiogenesis,
growth, and metastasis and are coexpressed in a variety of cancers.
The promoters of their genes contain different recognition sites for
transcription factors nuclear factor (NF)-kappaB (Bancroft et al.,
2001).
MCP-1 and RANTES are potent chemoattractants belonging to
the CC subfamily of chemokines that are involved in immunoregulatory and inammatory processes with ability to recruit,
activate, co-stimulate T cells and monocytes and attract above
mentioned cells into the place of destruction/inammation (Conti
and Di Gioacchino, 2001). In literature it is reported that MCP-1
and RANTES expression is regulated from NF-kB and the blocking
of MCP-1 activity in immunodecient mice bearing human breast
carcinomas resulted in signicant prolongation of the survival of
tumor-bearing mice (Salcedo et al., 2000).
Therefore, the reduction of IL-8, VEGF, MCP-1 and RANTES could
indicate that they inhibit the activation of NF-kB by blocking
tumor growth.
On the other hand IL-12 is an interleukin, naturally produced by
dendritic cells, macrophages and human B-lymphoblastoid cells in
response to antigenic stimulation. It stimulates, from T and natural
killer (NK) cells, the production of interferon-gamma (IFN-g),
which in turn increases the production of a chemokine called
inducible protein-10 (CXCL10) that mediates this anti-angiogenic
effect (Coma et al., 2006). This can explain because both IL-12 and
IP-10 decreased after treatment with compound 2 in MCF-7 cells.
Taking into consideration these data, it is suggest a possible
anti-inammatory effect of compound 2 on MCF-7 cells and its
capacity of reducing NF-kB activation.
Recent data have evidenced that inammation is a critical
component of tumor development. A growing body of epidemiologic evidence has emerged showing that long-standing inammation promotes tumor development, growth, and progression
(Multhoff et al., 2012). For this reason, many research groups are
working to nd new possible anti-inammatory molecules.
Therefore, concerning breast cancer is a complex and heterogeneous disease, we have evaluated the cytokine orescence
intensities after treatment with compounds 2 by Human Cytokine
27-Plex Panel also in supernatants of another breast cancer cell
line, MDA-MB231, and in this case the cytokine levels resulted
similar to those in control cells. It could depend from the intrinsic
difference between MDA-MB231 and MCF7 human breast cancer

156

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

as internal standard. The Heteronuclear Single-Quantum Coherence (HSQC) spectra were optimized for an average 1JCH of
140 Hz; the gradient-enhanced Heteronuclear Multiple Bond
Correlation (HMBC) experiment were optimized for a 3JCH of
8 Hz. Nuclear Overhauser Effect (NOE) measurements were
performed by 2D ROESY experiment. Through-space 1H connectivities were evidenced using a ROESY experiment with mixing
times of 300 ms. HPLC was performed using a Waters 510 pump
equipped with a Waters Rheodine injector and a Waters 401
differential refractometer as detector, using a 30 cm  3.9 mm;
i.d., C18 m-Bondapak (Waters, Milford, MA, USA) columns; ow
rate was 1 mL min1, and HPLC on a JASCO PU-2089 Plus
equipped with a UV/vis detector (UV-2075 Plus JASCO) at
270 nm using a Luna C18 column (150 mm  4.60 mm i.d., 3 mm,
Phenomenex).
The GC/MS analysis was carried out with an Agilent Technologies 6890N Network gas chromatograph coupled to an Agilent
Technologies 5973 Network quadrupole mass selective spectrometer and provided with a split/splitless injection port. Helium was
used as carrier gas at a linear velocity of 40 cm/s. Droplet countercurrent chromatography (DCCC) was performed on a DCC-A
apparatus (Tokyo Rikakikai Co., Tokyo, Japan) equipped with 250
glass-columns. The purity of compounds was determined to be
greater than 95% by HPLC, MS and NMR.
3.2. Plant material
Selected ripe berries of wild growing plants of J. oxycedrus L.
(Cupressaceae) were collected in the mountain areas of Isernia
(central Italy) in October 2010 and identied by Dr. Paola Fortini. A
voucher specimen is deposited (JO-301-10) at the Herbarium of
Dipartimento di Bioscienze e Territorio, University of Molise
(Pesche, Isernia). Berries were kept frozen at 20 8C until analyzed.
3.3. Extraction and isolation

Fig. 5. Cytokine levels evaluated in MCF-7 cells after treatment with compound 2.
We indicate with * the comparisons between the levels in the untreated and treated
cells resulted statistically signicant with p < 0.05 by T-test. The labels show the
concentrations corresponding to the different colors.

cell lines. Indeed, MCF7 cells are estrogen receptor alpha


(ERalpha)-positive, weakly invasive and luminal epithelial-like,
whereas MDA-MB231 cells are ERalpha-negative, highly invasive
and broblast-like. Hence, further studies will be performed to
evaluate the cytokine levels on other breast cancer cell lines
because many different cell lines are known for this cancer
(Holliday and Speirs, 2011).
3. Experimental
3.1. General experiment procedure
High-resolution ESI mass spectrometry (HR-ESI-MS) was
recorded on a Micromass QTOF spectrometer and electrospray
ionization mass spectrometry (ESI-MS) experiments were performed on an Applied Biosystem API 2000 triple-quadrupole mass
spectrometer. Optical rotations were determined on a Jasco P-2000
polarimeter. NMR spectra were obtained on Varian Inova 500 NMR
spectrometer (1H at 500 MHz and 13C at 125 MHz) equipped with a
Sun hardware, d (ppm), J in Hz, spectra referred to CD3OH (dH = 3.31)

Fresh ripe berries (100 g) were crushed and extracted with


MeOH (3 700 mL) at room temperature. The combined extracts
(15 g) were concentrated and subjected to a modied Kupchans
(Kupchan et al., 1973) partitioning procedure, as reported in De
Marino et al. (2011), obtaining three fractions: n-hexane extract
(3.2 g), CHCl3 extract (2.5 g), n-butanol extract (2.0 g) and an
aqueous residue. The n-hexane extract was separated by column
chromatography on silica gel and then puried by HPLC in the
same conditions reported in De Marino et al. (2011), to give mainly
cis-communic acid and myrcecommunic acid. The CHCl3 extract
(1.5 g) was fractionated by DCCC using CHCl3/MeOH/H2O (7:13:8)
in the ascending mode (the lower phase was the stationary phase)
to give three main fractions. Fractions were combined and
monitored by TLC on silica gel with CHCl3/MeOH/H2O (80:18:2)
as eluent and then separated by HPLC (C18 m-Bondapak column;
30 cm  3.9 mm i.d.). Fraction 1 puried by HPLC with MeOH/H2O
(7:3) contained mainly imbricatolic acid; fraction 2 processed with
MeOH/H2O 8:2 yielded myrcecommunic acid and sandarocopimaric acid; fraction 3 contained several aldehydes and were
puried by HPLC (MeOH/H2O 75:25) as eluent.
The n-BuOH extract (2 g) was submitted to DCCC with n-BuOH/
Me2CO/H2O (3:1:5) in the descending mode (the upper phase was
the stationary phase), to give three main fractions A-C. The
obtained fractions were monitored by TLC on Silica gel plates with
n-BuOH/OHAc/H2O (12:3:5) and CHCl3/MeOH/H2O (80:18:2).
Fraction A (180 mg) mainly contained free sugars and compound
2 (6.3 mg) puried by HPLC on a C18 m-Bondapak column (MeOH/
H2O 15:85) as eluent. Fraction B (550 mg) contained the new
monoterpene glucoside 1 (1.2 mg), the glucoside 3 (1.8 mg) and
the fructofuranoside derivative 4 (mg 1.5) and they were puried

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

157

Table 3
Cytokine levels evaluated in MCF-7 cells after treatment with compound 2 by Human Cytokine 27-Plex Panel.

PDGF-bb
IL-1b
IL-1ra
IL-2
IL-4
IL-5
IL-6
IL-7
IL-8
IL-9
IL-10
IL-12
IL-13
IL-15
IL-17
Eotaxin
FGF basic
G-CSF
GM-CSF
IFN-g
IP-10
MCP-1
MIP-1a
MIP-1b
RANTES
TNF-a
VEGF

CTRL

5 mM

10 mM

20 mM

30 mM

40 mM

50 mM

52.8
33.8
35.2
116.5
28.3
16.0
97.7
46.2
612.2
140.7
359.0
1082.2
81.3
216.5
99.3
29.7
93.7
42.3
1732.2
39.2
26.0
3028.7
33.5
65.3
159.8
25.7
4923.7

52.2
32.6
35.7
115.5
26.0
14.7
93.8
39.8
590.3
142.7
344.7
915.2
80.0
214.8
109.0
29.5
95.7
37.7
1738.2
31.8
16.3
2508.8
32.0
69.8
65.0
24.3
4395.0

50.0
31.9
34.7
114.0
27.2
14.2
97.8
37.5
169.0
137.3
347.7
846.0
77.8
219.0
104.5
28.5
93.8
34.7
1725.0
32.3
15.7
920.5
31.8
69.0
66.7
23.0
3985.5

51.9
32.4
34.2
115.0
24.7
13.0
95.5
41.5
42.8
137.8
356.9
732.7
79.7
219.8
108.3
28.0
92.2
35.6
1729.7
29.3
15.3
235.8
28.8
66.0
60.3
22.0
3539.3

49.6
32.8
34.8
112.5
23.8
12.7
94.5
41.5
24.7
139.0
358.8
372.3
77.2
207.5
104.5
28.2
92.3
37.3
1713.5
34.3
14.3
163.2
26.7
65.2
59.8
20.0
2136.0

49.1
32.9
34.5
113.6
23.6
12.5
93.6
39.9
23.6
136.9
349.6
372.1
76.5
211.5
103.5
28.1
91.7
37.5
1723.9
34.8
13.9
163.2
29.6
67.6
59.3
21.3
2136.0

49.3
32.3
34.4
113.1
23.3
12.6
93.2
40.1
22.9
138.7
348.9
371.3
77.9
213.9
103.9
28.5
90.3
36.7
1698.7
34.6
13.7
163.2
28.5
66.4
58.9
20.9
2136.0

by HPLC with MeOH/H2O 15:85, 25:75 and 20:80 respectively, as


eluent; fraction C was separated by HPLC (MeOH/H2O 45:55) to
give compound 5 (2.2 mg) and a mixture of polyphenolic
components.
3.3.1. Acid hydrolysis of 1
Compound 1 (0.3 mg) was hydrolyzed with 2 N CF3CO2H (2 mL)
at 110 8C in a sealed tube for 8 h. After cooling, the solution was
diluted with H2O (5 mL) and extracted with AcOEt (3 2 mL). The
aqueous layer was evaporated to dryness under reduced pressure
and the residue was reacted with 0.1 M L-cysteine methyl ester
hydrochloride in anhydrous pyridine (200 mL) for 1 h at 60 8C
(Hara, Okabe, & Mihashi, 1987). 1-(Trimethylsilyl)imidazole in
pyridine was added and the thiazolidine derivatives analyzed by
GCMS. D-Glucose were conrmed in 1 by comparison of the
retention times of their derivatives with those of D-glucose
(Rt = 17.88 min) and L-glucose (Rt = 19.20 min). The hydrolysate
was subjected to GCMS for identication of the sugar moiety on a
HP-5MS capillary column (30 m  0.25 mm, 0.25 mm lm thickness, Agilent USA). GC oven temperature was kept constant at
180 8C. The injector temperature was 230 8C. The temperature of
the ion source and the transfer line was 250 and 280 8C,
respectively. Mass spectra were taken at 70 eV and the mass
range was from 40 to 350 a.m.u.

(R)-PGME amide of compound 1: 1H NMR (500 MHz in CD3OD) d


7.38 (5H, m, phenyl), 5.57 (1H, d, CHNH), 3.75 (3H, s, OCH3). Other
signals are in Table 1. ESI-MS m/z 496 [M+H]+.
(S)-PGME amide of compound 1: 1H NMR (500 MHz in CD3OD) d
7.40 (5H, m, phenyl), 5.58 (1H, d, CHNH), 3.77 (3H, s, OCH3). Other
signals are in Table 1. ESI-MS m/z 496 [M+H]+. See supplementary
material for 1H NMR, COSY, HSQC, HMBC spectra.
3.3.3. Shikimic acid (2)
White amorphous powder, ([a]D25 148.7, c. 0.20, MeOH);
positive ESI-MS m/z 175 [M+H]+.
3.3.4. Monoterpene glucoside (3)
White amorphous powder, ([a]D25 35.0, c. 0.18, MeOH);
positive ESI-MS m/z 369 [M+Na]+.
3.3.5. n-Butyl b-D-fructofuranoside (4)
Colorless oil, ([a]D25 28.8, c. 0.15, MeOH); positive ESI-MS m/z
237 [M+H]+.
3.3.6. Phenyl propanoid rhamnoside (5)
White amorphous powder, ([a]D25 +33.9, c. 0.22, MeOH);
positive ESI-MS m/z 515 [M+Na]+.
3.4. Cell culture

3.3.2. (R)- and (S)-PGME amides of compound 1


To an ice cooled DMF solution (500 mL) of 1 (0.4 mg) and (R)PGME (0.5 mg) were added PyBOP (1.0 mg), HOBt (0.5 mg) and Nmethylmorpholine (25 mL) and stirring was continued at room
temperature for 5 h. After addition of 5% HCl (1 mL), the mixture
was extracted with EtOAc (2.5 mL). The extract was washed with
saturated NaHCO3 aq (1 mL) and brine (1 mL), and then
concentrated in vacuo to afford the (R)-PGME amide of 1
(0.5 mg). The (S)-PGME amide of 1 was prepared according to
the same procedure as described above. Each diasteroisomer was
subjected to HPLC (Luna C18 3 mm) eluting with MeOH/H2O (from
20% to 100% MeOH) gradient 20% to 100% MeOH in 15 min, then
hold 100% MeOH for 10 min, by UV detector at 270 nm.

Cell cultures were kept in growth conditions identical or as


similar as possible. A375 and MCF-7, malignant melanoma and
breast cancer cell lines, were kept in culture and expanded at 37 8C
in a humidied atmosphere of 5% CO2 in culture medium DMEM
(Dulbeccos Modied Eagles Medium, Lonza, Verviers, Belgium),
supplemented with 10% FBS (Invitrogen, Camarillo, CA, USA) at 1%
penicillin/streptomycin 100 (Euroclone, Devon, UK), 1% Glutamax 100 (Invitrogen). The other human lung cancer cell line,
H460, was kept in culture and expanded in humidied atmosphere
at 37 8C and 5% CO2 incubator in culture medium RPMI1640 w/o LGlutamine (Lonza, Verviers, Belgium), supplemented with 10% FBS
(Invitrogen, Camarillo, CA, USA) at 1%, penicillin/streptomycin

158

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

100 (Euroclone, Devon, UK), 1% Glutamax 100 (Invitrogen).


Phosphate buffer 10 (PBS phosphate buffered saline Ca2+ and
Mg2+ free) and trypsin (Ca2+ and Mg2+ free) were supplied by
Euroclone.
3.5. Colorimetric assay with sulforhodamine B
First of all, cell proliferation was assessed in presence of
n-butanolic extract, by colorimetric assay with sulforhodamine
B (SRB, Sigma Aldrich) on the three cell lines and compared to
untreated cells. The cells (4.5  1045  104) were seeded in
96-multiwell plates in 200 mL of culture medium, and left to grow
for 24 h at 37 8C for allowing adhesion. Then, the cells were treated
with J. oxycedrus extract using different concentrations: 50 mg,
80 mg, 200 mg, 300 mg and 400 mg and then incubated for 24 h.
In particular, the extract was dissolved in dimethyl sulfoxide
(DMSO, SigmaAldrich) at a concentration of 100 mg/mL. In cell
cultures the DMSO concentration remained always below 0.1%, a
dose that did not exert toxic effects. Then, the cells were xed by
the addiction of 10% trichloroacetic acid (TCA, SigmaAldrich) per
well for at least 1 h at 4 8C. Subsequently, the cells were washed
with distilled water and were dried to air.
100 mL of SRB was added to each well and the plate was
incubated for 30 min at RT, protected from light. To remove the dye
excess the cells were washed using 1% acetic acid. The number of
viable cells was directly proportional to the protein bound-dye
formation which was then solubilized with 100 mL of 10 mM Tris
base solution pH 10.5 per well, shaking the plates for at least
15 min on a gyratory shaker to homogenize the dye solution.
Measure of OD was performed by using an automated 96-well
plate reader (Bio-Rad, Hercules, CA, USA; Microplate Reader) at a
wavelength of 540 nm. All experiments were performed in
triplicate and were repeated for three times. The cellular viability
was estimated as % compared to untreated cells.
Based on the reduction in cell viability observed in MCF-7 cell
line after 24 h of treatment, we also treated the MCF-7 with the
compounds 2, 3, 4 and 5 at different concentrations 5 mM, 10 mM,
20 mM, 30 mM, 40 mM and 50 mM.
3.6. Apoptosis assay
The cells (1  106) have been harvested and washed two times
with cold PBS. Subsequently, the cells have been labeled by
Annexin V & Dead Cell Assay kit in according to instructions of
producer (Merck Millipore, Darmstadt, Germany). This assay is
based on the detection of phosphatidylserine (PS) on the surface of
apoptotic cells using Annexin V conjugated FITC in combination
with cell death marker, 7-actinomycin D (7-AAD). This staining
allows quantitative analysis of live cells, cells in early and later
apoptosis and dead cells. The samples have been tested on MuseTM
Analyzer (Merck Millipore) and were analyzed by a software
supplied by the same company.
3.7. Cell Cycle Assay
The MuseTM Cell Cycle Assay uses a premixed reagent which
includes the nuclear DNA intercalating stain propidium iodide (PI)
and RNAse A in a proprietary formulation. PI discriminates cells at
different stages of the cell cycle, based on differential DNA content
in the presence of RNAse to increase the specicity of DNA staining.
It is important to have a single cell suspension prior to ethanol
xation and the minimum recommended number of cells for
xation in a tube is 1  106 cells. The samples were centrifuged at
300  g for 5 min and after removing and discarding the
supernatant, an appropriate volume of PBS was added to each
tube (1 mL of PBS per 1  106 cells). After centrifugation and

removing of the supernatant, 1 mL of ice cold 70% ethanol was


added to the resuspending cell pellet in the residual PBS. The
tubes were capped and freezed at 20 8C for at least 3 h prior to
staining. Ethanol-xed cells were centrifuged at 300  g for
5 min at room temperature and pellet was resuspended in PBS.
The cells were centrifuged again at 300  g for 5 min at room
temperature, the supernatant was removed and discarded and
cell pellet was resuspended in 200 mL of MuseTM Cell Cycle
Reagent and incubated for 30 min at room temperature,
protected from light. Cell suspension samples were transferred
to a 1.5-mL microcentrifuge tube prior to analysis on MuseTM
Cell Analyzer. The MuseTM Cell Cycle Assay uses a premixed
reagent which includes the nuclear DNA intercalating stain
propidium iodide (PI) and RNAse A in a proprietary formulation.
PI discriminates cells at different stages of the cell cycle, based
on differential DNA content in the presence of RNAse to increase
the specicity of DNA staining. Resting cells (G0/G1) contain two
copies of each chromosome. As cells begin cycling, they
synthesize chromosomal DNA (S phase). Fluorescence intensity
from PI increases until all chromosomal DNA has doubled (G2/M
phase). At this stage, the G2/M cells uoresce with twice the
intensity of the G0/G1 population. The G2/M cells eventually
divide into two cells.
The assay utilizes PI-based staining of DNA content to
discriminate and measure the percentage of cells in each cell
cycle phase (G0/G1, S, and G2/M). The Muse Cell Cycle Software
Module performs calculations automatically.
Data is displayed in two plots:
 DNA content index and cell size index dot plot.
 DNA content index histogram with markers available to analyze
the cell populations in each phase of the cycle.

3.8. Bio-Plex assay


The levels of a panel of numerous cytokines, chemokines, and
growth factors were evaluated at the same time by BioPlex assay.
The simultaneous quantitative determination of a large panel of
cytokines, able to report the correct ratios and dynamics between
highly and poorly represented molecules, has emerged as an
accurate, simple, specic, noninvasive, reproducible and less
expensive method (Rusolo et al., 2013; Costantini et al., 2014).
The multiplex biometric ELISA-based immunoassay, containing
dyed microspheres conjugated with a monoclonal antibody
specic for a target protein was used, according to the manufacturers instructions (Bio-Plex Bio-Rad), to evaluate the levels of
different cytokines by Human Cytokine 27-Plex Panel after
treatment with compounds 2, 3, 4 and 5 in MCF-7 supernatants.
In particular, the following cytokines were evaluated: fourteen
interleukins (IL), IL-1b, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9,
IL-10, IL-12 (p70), IL-13, IL-15, IL-17, and eotaxin (CCL11), basic
broblast growth factor (FGF), granulocyte colony-stimulating
factor (G-CSF), granulocyte macrophage colony-stimulating factor
(GM-CSF), interferon-gamma (IFN-g), chemokine (C-X-C motif)
ligand 10 (CXCL10), monocyte chemo-attractant protein-1 (MCP-1),
two macrophage inammatory protein1s, MIP-1a and MIP-1b,
platelet-derived growth factor-b (PDGF-b), RANTES (regulated on
activation, normal T cell expressed and secreted), tumor necrosis
factor-a (TNF-a) and VEGF. Each experiment was performed in
duplicate as previously described (Rusolo et al., 2013). Protein levels
were determined using a Bio-Plex array reader (Luminex, Austin, TX,
USA) that quantitates multiplex immunoassays in a 96-well format
with very small uid volumes. The analyte level was calculated using
a standard curve, with software provided by the manufacturer (BioPlex Manager Software).

S. De Marino et al. / Phytochemistry Letters 10 (2014) 152159

The cytokines levels evaluated in MCF-7 supernatants were


compared by T-test. Values of p < 0.05 were considered to be
statistically signicant.
Acknowledgements
MS and NMR spectra were provided by Laboratorio di Analisi
Strumentale (LAS), Dipartimento di Farmacia, Universita` di Napoli
Federico II, Napoli, Italy. We are also grateful to Dr. Francesca
Fantasma (Universita` del Molise) for the technical support.
Appendix A. Supplementary data
Supplementary data associated with this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.phytol.2014.08.015.
References
Adams, R.P., 1998. The leaf essential oils and chemotaxonomy of Juniperus. Biochem. Syst. Ecol. 26, 637645.
Adams, R.P., Altajeros, J., Fernandez, C., Camacho, A., 1999. The leaf essential oils and
taxonomy of Juniperus oxycedrus L. subsp. oxycedrus, subsp. badia (H Gay) Debeaux,
and subsp. macrocarpa (Sibth e sm) Ball. J. Essent. Oil Res. 11, 167172.
Akkol, E.K., Guvenc, A., Yesilada, E., 2009. A comparative study on the antinociceptive and anti-inammatory activities of ve Juniper taxa. J. Ethnopharmacol.
125, 330336.
Ali, K., Maltese, F., Zyprian, E., Rex, M., Choi, Y.H., Veerporte, R., 2009. NMR metabolic
ngerprinting based identication of grapewine metabolites associated with
downy mildew resistance. J. Agric. Food Chem. 57, 95999606.
Amri, I., Hamrouni, L., Gargouri, S., Banana, M., Jamoussi, B., 2013. Chemical
composition and antifungal activity of essential oils isolated from Juniperus
oxycedrus L. Int. J. Appl. Biol. Pharm. Technol. 4, 227233.
Bancroft, C.C., Chen, Z., Dong, G., Sunwoo, J.B., Yeh, N., Park, C., Van Waes, C., 2001.
Coexpression of proangiogenic factors IL-8 and VEGF by human head and neck
squamous cell carcinoma involves coactivation by MEK-MAPK and IKK-NFkappaB signal pathways. Clin. Cancer Res. 7, 435442.
Barrero, A.F., Oltra, J.E., Altarejos, J., Barragan, A., Lara, A., Laurent, R., 1993. Minor
components in the essential oil of Juniperus oxycedrus L. wood. Flavour Fragr. J.
8, 185189.
Bello, R., Moreno, L., Beltran, B., Primo-Yufera, E., Esplugues, J., 1997. Effects on
arterial blood pressure of methanol and dichloromethanol extracts from J.
oxycedrus L. Phytother. Res. 11, 161162.
Boti, J.B., Bighelli, A., Cavaleiro, C., Salgueiro, L., Casanova, J., 2006. Chemical variability of Juniperus oxycedrus ssp. oxycedrus berry and leaf oils from Corsica, analysed
by combination of GC, GCMS and 13C NMR. Flavour Fragr. J. 21, 268273.
Chalchat, J.C., Garry, R.P., Michet, A., Peyron, L., 1990. Chemical components of
natural and empyreumatic oil and extracts from Juniperus oxycedrus and
Juniperus phoenicea wood. J. Essent. Oil Res. 2, 231236.
a, R., Blanco, J., Blanco, J., Rosell, A., Borras, F.E., Este, J.A., Clotet, B., Ruiz,
Coma, G., Pen
L., Parkhouse, R.M., Boll, M., 2006. Treatment of monocytes with interleukin
(IL)-12 plus IL-18 stimulates survival, differentiation and the production of CXC
chemokine ligands CXCL8, CXCL9 and CXCL10. Clin. Exp. Immunol. 145, 535544.
Conti, P., Di Gioacchino, M., 2001. MCP-1 and RANTES are mediators of acute and
chronic inammation. Allergy Asthma Proc. 22, 133137.
Costantini, S., Rusolo, F., De Vito, V., Moccia, S., Picariello, G., Capone, F., Guerriero, E.,
Castello, G., Volpe, M.G., 2014. Potential anti-inammatory effects of the
hydrophilic fraction of pomegranate (Punica granatum L.) seed oil on breast
cancer cell lines. Molecules 19, 86448660.
De Marino, S., Cattaneo, F., Festa, C., Zollo, F., Iaccio, A., Ammendola, R., Incollingo, F.,
Iorizzi, M., 2011. Imbricatolic acid from Juniperus communis L. prevents cell
cycle progression in CaLu-6 cells. Planta Med. 77 (16) 18221828.
Demetzos, C., Dimas, K., 2001. Labdane-type diterpenes: chemistry and biological
activity. In: Atta-Ur-Rahman (Eds.), Studies in Natural Products Chemistry of
Bioactive Natural Products, vol. 25. Elsevier Science, Oxford, pp. 235292.
Dimas, K., Papadaki, M., Tsimplouli, C., Hatziantoniou, S., Alevizopoulos, K., Pantazis,
P., Demetzos, C., 2006. Labd-14-ene-8,13-diol (sclareol) induces cell cycle arrest
and apoptosis in human breast cancer cells and enhances the activity of
anticancer drugs. Biomed. Pharmacother. 60, 127133.
Falasca, A., Melck, D., Paris, D., Saviano, G., Motta, A., Iorizzi, M., 2014. Seasonal
changes in the metabolic ngerprint of Juniperus communis L. berry extracts by
1
H NMR-based metabolomics. Metabolomics 10, 165174.

159

Fernandez, A., Ortuno, I., Martos, A., Fernandez, C., 1996. Saber Y utilizacion de
plantas en la provincia de Jae`n. Campagna de 1993. Boletin del Istituto de
Estudios Giennenses 161, 199318.
Hara, S., Okabe, H., Mihashi, K., 1987. Gasliquid chromatographic separation of
aldose enantiomers as trimethylsilyl ethers of methyl 2-(polyhydroxyalkyl)thiazolidine-4-(R) carboxylates. Chem. Pharm. Bull. 35, 501506.
Holliday, D.L., Speirs, V., 2011. Choosing the right cell line for breast cancer research.
Breast Cancer Res. 13, 215221.
Hu, L.H., Chen, Z.L., 1997. Structure elucidation of a new n-pentyl fructofuranoside
in Dendranthema morifolium (Ramat) Tzvel. Acta Bot. Sin. 39, 181184.
Karaman, I., Sahin, F., Gulluce, M., Ogutcu, H., Sengul, M., Adiguzel, A., 2003.
Antimicrobial activity of aqueous and methanol extracts of Juniperus oxycedrus
L. J. Ethnopharmacol. 85, 231235.
Kupchan, S.M., Britton, R.W., Ziegler, M.F., Siegel, C.W., 1973. Bruceantin, a new
potent antileukemic simaroubolide from Brucea antidysenterica. J. Org. Chem.
38, 178179.
Leung, A.Y., Foster, S., 1996. Encyclopedia of Common Natural Ingredients. Wiley,
New York, pp. 109.
Lizzo, M.R., Tundis, R., Conforti, F., Saab, A.M., Statti, G.A., Menichini, F., 2007.
Comparative chemical composition, antioxidant and hypoglycaemic activities
of Juniperus oxycedrus ssp. oxycedrus L. berry and wood oils from Lebanon. Food
Chem. 105, 572578.
Lundgren, L.N., Shen, Z., Theander, O., 1985. The constituents of conifer needles. Dilignol
glycosides from Pinus massoniana Lamb. Acta Chem. Scand. B39, 241248.
Matsubara, Y., Nakano, T., Sawabe, A., Iizuka, Y., Okamoto, K., 1990. Structure and
physiological activity of phenyl propanoid glycosides in the leaf of hinoki
(Chamaecyparies obtusa Sieb. et Zucc.). Agric. Biol. Chem. 54 (2) 557559.
Medini, H., Eliassi, A., Khouja, M.L., Chraief, I., Farhat, F., Hammami, M., Chemli, R.,
Harzallah-Skhir, F., 2010. Leaf essential oil of Juniperus oxycedrus L. (Cupressaceae) harvested in northern Tunisia: composition and intra-specic variability.
Chem. Biodivers. 7, 12541266.
Milos, M., Radonic, A., 2000. Gas chromatography mass spectral analysis of free and
glycosidically bound volatile compounds from Juniperus oxycedrus L. growing
wild in Croazia. Food Chem. 68, 333338.
Montagne, P., 1999. The Concise Larousse Gastronomique. Hamlyn, London, UK, pp.
691.
Moreno, L., Bello, R., Beltran, B., Calatayud, S., Promo-Yufera, E., Esplugues, J., 1998.
Pharmacological screening of different Juniperus oxycedrus L. extracts. Pharmacol. Toxicol. 82, 108112.
Multhoff, G., Molls, M., Radons, J., 2012. Chronic inammation in cancer development. Front. Immunol. 2, 98108.
Nakanishi, T., Iida, N., Inatomi, Y., Murata, H., Inada, A., Murata, J., Lang, F.A., Inuma,
M., Tanaka, T., Sakagami, Y., 2005. A monoterpene glucoside and three megastigmane glycosides from Juniperus communis var. depressa. Chem. Pharm. Bull.
53 (7) 783787.
Rusolo, F., Pucci, B., Colonna, G., Capone, F., Guerriero, E., Milone, M.R., Nazzaro, M.,
Volpe, M.G., Di Bernardo, G., Castello, G., Costantini, S., 2013. Evaluation of
selenite effects on selenoproteins and cytokinome in human hepatoma cell
lines. Molecules 18, 25492562.
Salcedo, R., Ponce, M.L., Young, H.A., Wasserman, K., Ward, J.M., Kleinman, H.K.,
Oppenheim, J.J., Murphy, W.J., 2000. Human endothelial cells express CCR2 and
respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression.
Blood 196, 3440.
Sanchez de Medina, F., Gamez, M.I., Jmenez, J., Osuna, J.I., Zarzuelo, A., 1994.
Hypoglycaemic activity of juniper berries. Planta Med. 60, 197200.
Sezik, E., KocaKulaK, E., Baser, K.H.B., Ozek, T., 2005. Composition of essential oils of
Juniperus oxycedrus L. ssp. macrocarpa from Turkey. Chem. Nat. Compd. 41, 352354.
Stassi, V., Verykokidou, E., Loukis, A., Harvala, A., Philianos, S., 1995. Essential oil
from Juniperus oxycedrus L. subsp. macrocarpa (Sm) Ball. J. Essent. Oil Res. 7,
675676.
Tavares, L., McDougall, G.J., Fortalezas, S., Stewart, D., Ferreira, R.B., Santos, C.N.,
2012. The neuroprotective potential of phenolic-enriched fractions from four
Juniperus species found in Portugal. Food Chem. 135, 562570.
Taviano, M.F., Marino, A., Trovato, A., Bellinghieri, V., Melchini, A., Dugo, P., Cacciola,
F., Donato, P., Mondello, L., Guvenc, A., De Pasquale, R., Miceli, N., 2013. Juniperus
oxycedrus L. subsp. oxycedrus and Juniperus oxycedrus L. subsp. macrocarpa
(Sibth & Sm.) Ball. berries from Turkey: comparative evaluation of phenolic
prole, antioxidant, cytotoxic and antimicrobial activities. Food Chem. Toxicol.
58, 2229.
Valentini, G., Bellomaria, B., Maggi, F., Manzi, A., 2003. The leaf and female cone oils
of Juniperus oxycedrus L. ssp. oxycedrus and J. oxycedrus L. ssp. macrocarpa (Sibth
e Sm.) Ball. from Abruzzo. J. Essent. Oil Res. 15, 418421.
Yabuuchi, T., Kusumi, T., 2000. Phenylglycine methyl ester, a useful tool for absolute
conguration determination of various chiral carboxylic acids. J. Org. Chem. 65,
397404.
Yesilada, E., Honda, G., Sezik, E., Tabata, M., Goto, K., Ikeshiro, Y., 1993. Traditional
medicine in Turkey IV. Folk medicine in the Mediterranean subdivision. J.
Ethnopharmacol. 39, 3138.

You might also like