ArticlePDF Available

Oestrogenic activity of the hop phyto-oestrogen, 8-Prenylnaringenin

Authors:

Abstract and Figures

The female flowers of the hop plant (hop cones) are used as a preservative and as a flavouring agent in beer. A novel phyto-oestrogen, 8-prenylnaringenin, was recently identified in hops and this study was undertaken to characterize the oestrogenic activity of this compound using a combination of in vitro and in vivo assays. Natural and semi-synthetic 8-prenylnaringenin showed similar bioactivities both in a yeast screen transfected with the human oestrogen receptor and in oestrogen-responsive human Ishikawa Var-I cells. 8-Prenylnaringenin showed comparable binding activity to both oestrogen receptor isoforms (ER alpha and ER beta). 8-Prenylnaringenin extracted from hops contains similar amounts of both (R)- and (S)- enantiomers, indicating that the compound is normally formed non-enzymatically. Both enantiomers showed similar bioactivity in vitro and similar binding characteristics to ER alpha and ER beta. The oestrogenic activity of 8-prenyl-naringenin in vitro was greater than that of established phyto-oestrogens such as coumestrol, genistein and daidzein. The high oestrogenic activity was confirmed in an acute in vivo test using uterine vascular permeability as an end point. When the compound was given to ovariectomized mice in their drinking water, oestrogenic stimulation of the vaginal epithelium required concentrations of 100 mug ml(-1) (about 500-fold greater than can be found in any beer).
Content may be subject to copyright.
Introduction
The female flowers (cones) of hops (
Humulus lupulus
L.)
have been used for centuries as a preservative and as a
flavouring agent in beer. Over the years, a recurring
suggestion has been that hops have a powerful oestrogenic
activity and that beer may also be oestrogenic. Hop baths
have been used for the treatment of gynaecological disorders
and hop extracts have been reported to be effective in
reducing hot flushes in menopausal women. In times when
hops were picked by hand, menstrual disturbances among
female pickers were reportedly common. Although the
oestrogenic activity of hops was attributed to xanthohumol,
a prenylated chalcone, this was without any experimental
support (Verzele, 1986). Initial scientific studies of the
oestrogenic activity of hops proved contradictory: some
early investigations (Koch and Heim, 1953; Zenisek and
Bednar, 1960; Hesse
et al
., 1981) reported that the
oestrogenic activity of hops was very high, but other
investigations (Feneslau and Talalay, 1973) found no
indication of oestrogenicity. These discrepancies over the
potential oestrogenic activity of hops are likely to have been
due to the variable nature of the extracts and the variety of
assays used. However, after bioassay-guided fractionation
of hop extracts, Milligan
et al
. (1999) identified a novel
phyto-oestrogen, 8-prenylnaringenin (Fig. 1). Using both
a yeast-based screen to detect oestrogenic activity and a
receptor binding assay using rat uterine cytosol, 8-
prenylnaringenin appeared to be one of the most potent
phyto-oestrogens, and had an oestrogenic activity con-
siderably greater than that of phyto-oestrogens such as
genistein and daidzein (Fig. 1).
There is considerable interest in whether human
exposure to phyto-oestrogens has any health risks or
benefits (Knight and Eden, 1996; Cassidy and Milligan,
1998). The secretions of the lupulin glands present in the
female hop flowers contain predominantly bitter acids
and hop oil constituents, essential in brewing, along with
8-prenylnaringenin and other prenylflavonoids. 8-Prenyl-
naringenin can be found in many beers at concentrations of
up to 0.24 mg l
–1
(Stevens
et al
., 1999; Rong
et al
., 2000).
There may also be significant exposure of women to
8-prenylnaringenin from a number of commercially
available herbal preparations, including those claiming
breast enlargements (Sauer and Coldham, 2001). In view of
such exposure of humans to 8-prenylnaringenin and its
apparent high oestrogenic activity, the present study was
undertaken to characterize the nature of the oestrogenic
activity of this compound using a combination of
in vitro
and
in vivo
assays, and to examine the binding of 8-
prenylnaringenin to the two isoforms of the oestrogen
Oestrogenic activity of the hop phyto-oestrogen,
8-prenylnaringenin
S. Milligan
1
, J. Kalita
1
, V. Pocock
1
, A. Heyerick
2
, L. De Cooman
2
,
H. Rong
2
and D. De Keukeleire
2
1
Endocrinology and Reproduction Research Group, School of Biomedical Sciences,
King’s College, Guy’s Campus, London SE1 1UL, UK; and
2
Ghent University,
Laboratory of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences,
Harelbekestraat 72, B-9000 Ghent, Belgium
Reproduction
(2002) 123, 235–242
Research
The female flowers of the hop plant (hop cones) are used
as a preservative and as a flavouring agent in beer. A
novel phyto-oestrogen, 8-prenylnaringenin, was recently
identified in hops and this study was undertaken to
characterize the oestrogenic activity of this compound
using a combination of
in vitro
and
in vivo
assays. Natural
and semi-synthetic 8-prenylnaringenin showed similar
bioactivities both in a yeast screen transfected with the
human oestrogen receptor and in oestrogen-responsive
human Ishikawa Var-I cells. 8-Prenylnaringenin showed
comparable binding activity to both oestrogen receptor
isoforms (ERα and ERβ). 8-Prenylnaringenin extracted from
hops contains similar amounts of both (
R
)- and (
S
)-
enantiomers, indicating that the compound is normally
formed non-enzymatically. Both enantiomers showed similar
bioactivity
in vitro
and similar binding characteristics
to ERα and ERβ. The oestrogenic activity of 8-prenyl-
naringenin
in vitro
was greater than that of established
phyto-oestrogens such as coumestrol, genistein and daidzein.
The high oestrogenic activity was confirmed in an acute
in vivo
test using uterine vascular permeability as an
end point. When the compound was given to ovariec-
tomized mice in their drinking water, oestrogenic stimula-
tion of the vaginal epithelium required concentrations of
100 µg ml
–1
(about 500-fold greater than can be found in
any beer).
© 2002 Society for Reproduction and Fertility
1470-1626/2002
Email: Stuart.Milligan@kcl.ac.uk
receptor (ERα and ERβ). In addition, as 8-prenylnaringenin
contains a chiral centre at C(2) and can exist in both an (
R
)-
and (
S
)-form, the oestrogenic activity of both enantiomers
was investigated.
Materials and Methods
Compounds
Oestradiol, oestriol, genistein (4’,5,7-trihydroxyisoflavone)
and daidzein (4’,7-dihydroxyisoflavone) were supplied
by Sigma Chemical Co. Ltd, Poole. Coumestrol (3,9-
dihydroxy-6H-benzofuro-[3,2-c][1]benzopyran-6-one) was
from Acros Organics, NJ and formononetin (7-hydroxy-4’-
methoxyisoflavone) from Extrasynthèse, Genay. The anti-
oestrogen ICI 182,780 was a gift from A. Wakeling (Zeneca
Pharmaceuticals, Macclesfield).
Polyphenolic extracts of hops
Dry hop cones (5 g) were extracted consecutively under
reflux and nitrogen with petroleum ether (200 ml, 30 min;
125 ml, 60 min; 50 ml, 60 min) and
n
-hexane (125 ml,
60 min, three times). All fractions were discarded and the
residue was extracted under reflux and nitrogen with 80 ml
methanol:water (3:1; v/v) for 2 h (three times). This extract
was partitioned with petroleum ether (100 ml, twice) and
n
-
hexane (100 ml, twice), and the aqueous methanolic extract
was adjusted to 100 ml. Individual compounds were isolated
by semi-preparative HPLC (Kontron pump system 32X,
photodiode array detector 440, data system 450-MT2/DAD
series) using a Biosil C18 HL 90-10 (Bio-Rad, Nazareth)
10 250 mm (10 µm) column and a gradient of solvent A
(0.05% (v/v) formic acid in water) and solvent B (5% (v/v)
acetonitrile in methanol). Gradient profile: 0–2 min: isocratic
45% B in A; 2–32 min: 45% B in A to 95% B in A; 32–37
min: 95% B in A; 37–45 min: 95% B in A to 45% B in A;
45–47 min: 45% B in A. Retention times of isoxanthohumol,
8-prenylnaringenin, 6-prenylnaringenin and xanthohumol
were 15.1, 19.4, 23.7 and 26.4 min, respectively.
Preparation of 8-prenylnaringenin
Dry hop cones (300 g) were immersed in dichloro-
methane (2 l) for 2 h. The extract was filtered and the
solvent evaporated. After the addition of 300 ml 70% (v/v)
aqueous methanol, the solution was maintained at –20C
overnight. The precipitate was separated; the filtrate was
concentrated to dryness; the residue was redissolved in
50 ml of 5% (v/v) ethanolic potassium hydroxide; and the
solution was refluxed for 30 min under nitrogen. After
cooling to room temperature, the reaction mixture was
poured into 200 ml 5% (v/v) HCl and extracted twice with
ethyl acetate. The combined extracts were dried (Na
2
SO
4
) and
concentrated. The residue was purified by column chroma-
tography (Silica gel, Merck grade 9385, 60 Å) using ethyl
acetate:cyclohexane (9:1) to give 780 mg of isoxanthohumol
(80%). A cooled solution of boron trichloride (650 µl,
0.65 mmol) in dichloromethane was added to a stirred
solution of isoxanthohumol (15 mg, 0.04 mmol) in acetonitrile
(1 ml) at –78C. The reaction mixture was allowed to warm
up to 0C over a period of 3 h, and was then poured on to
ice and extracted with ethyl acetate. The extract was
washed with water, saturated with NaCl, dried over Na
2
SO
4
and concentrated to dryness. After purification by column
chromatography (Silica gel; ethyl acetate:cyclohexane 8:2),
7.6 mg of 8-prenylnaringenin was obtained (53%).
Separation of (R)- and (S)-8-prenylnaringenin
The enantiomers of the prenylated hop flavanones,
isoxanthohumol, 8-prenylnaringenin, and 6-prenylnaringenin
were separated by chiral liquid chromatography (triacetyl
cellulose (TAC), 4.1 250.0 mm, prepared in the Laboratory
of Separation Sciences, Department of Organic Chemistry,
Ghent University) using methanol as eluent at a flow rate of
0.8 ml min
–1
. UV detection was at 280 nm. The fractions
containing the enantiomers of 8-prenylnaringenin were
collected by semi-preparative chiral HPLC and the solvent
was evaporated. (
R
)-8-prenylnaringenin and (
S
)-8-prenyl-
naringenin were dissolved in methanol (3.4 mg (10 ml)
–1
and 3.3 mg (10 ml)
–1
, respectively) and their CD spectra
were recorded on an Olis DSM 10 CD spectrophotomer
(Olis, Bogart, GA; 1 mmol l
–1
in MeOH).
Determination of oestrogenic bioactivity
in vitro
Hop extracts and pure compounds were diluted in
ethanol. Aliquots (20 µl) were added to individual wells in a
96-well plate and the ethanol was evaporated. Oestrogenic
activity was determined in two separate
in vitro
bioassays
.
The first bioassay used a human endometrial cell line,
Ishikawa Var I (a gift from E. Gurpide, Mount Sinai School
of Medicine, NY) that responds to oestrogenic stimulation
by a marked increase in alkaline phosphatase activity
(Markiewicz
et al
., 1993). Cells (2.5 10
4
cells per well in
100 µl) were plated in 96-well plates in oestrogen-free basal
medium (EFBM; phenol red-free Ham’s F12 and Dulbecco’s
modified eagle’s medium (1:1), and 5% (v/v) charcoal-
236
S. Milligan
et al.
Oestradiol 8-Prenylnaringenin 6-Prenylnaringenin
Genistein Daidzein Coumestrol
HO
O
OOH OH
HO
O
O
OH
OH
O
O
O
HO
OH
CH
3
HO
HO
O
OOH
OH
O
OH
OOH
HO
Fig. 1. Structures of oestradiol and established phyto-oestrogens.
stripped FBS). The anti-oestrogen ICI 182,780 was used in
some wells at a final concentration of 0.16 µmol l
–1
to
evaluate the specificity of the response. Alkaline phosphatase
activity was determined after 72 h by monitoring the
hydrolysis of
p
-nitrophenyl phosphate to
p
-nitrophenol at
405 nm.
Oestrogenic bioactivity was also studied using an
oestrogen-inducible yeast screen (
Saccharomyces cerevisiae
)
expressing the human oestrogen receptor and containing
expression plasmids carrying oestrogen-responsive sequences
controlling the reporter gene lac-Z (encoding the enzyme
β-galactosidase; a gift from J. Sumpter, Brunel University).
Oestrogenic activity was determined from the enzymatic
hydrolysis of chlorophenol red β-
D-galactopyranoside by
monitoring the absorbance at 540 nm (Routledge and
Sumpter, 1996).
Receptor binding
Oestrogen receptor binding activity was studied using
recombinant human oestrogen receptor α and β (ERα and
ERβ) obtained from PanVera Corporation (Madison, WI).
Dilutions of the test compounds were incubated in 100 µl
buffer (10 mmol Trizma preset crystals l
–1
(pH 7.5), 10%
glycerol, 2 mmol
DL-dithiothreitol (DTT) l
–1
, 1 mg BSA ml
–1
)
with 15 nmol [2,4,6,7-
3
H]-oestradiol l
–1
(84.0 Ci mmol l
–1
;
Amersham Life Science, Amersham) and ER (1.5 nmol l
–1
).
The mixture was incubated overnight at 4C, and free and
bound hormone were separated using 100 µl 15% (w/v)
hydroxylapatite slurry (in 50 mmol Tris–HCl l
–1
, pH 7.4,
1 mmol EDTA l
–1
). After three washes in buffer (ERα:
40 mmol Tris–HCl l
–1
, pH 7.5, 100 mmol KCl l
–1
, 1 mmol
EDTA l
–1
, 1 mmol EGTA l
–1
; ERβ: 40 mmol Tris–HCl l
–1
,
pH 7.5), the slurry was extracted with two washes of 200 µl
ethanol and the radioactivity in the extracts was
determined.
In vivo
assays
The relative potency of oestrogenic compounds
in vivo
depends on a number of factors, including the route of
administration and the nature of the response monitored.
Estimates of oestrogenic potency are affected markedly
depending on the nature of the
in vivo
bioassay used.
Therefore, two
in vivo
assays were used: the first assay was
based on the rapid response of the uterine vasculature to
oestrogenic stimulation (Arvidson, 1977; Milligan
et al
.,
1998). The second assay was based on the uterotrophic
response and mitotic responses of the uterine and vaginal
epithelium. In both assays, female Swiss albino mice (A.
Tuck & Son Ltd, Battlesbridge, Essex), approximately 2–3
months of age and weighing 20–25 g, were maintained
under constant conditions of lighting (lights on from 06:00 h
to 18:00 h) and temperature (21 1C), and fed on a
pelleted diet (Economy Rodent Maintenance, Essex)
ad
libitum
. Animal care was undertaken according to UK
Home Office guidelines. All experiments were performed
on ovariectomized animals. Ovariectomies were performed
under tribromoethanol anaesthesia at least 2 weeks before
the start of each experiment.
Acute uterine vascular response
In the assay based on the rapid response of the uterine
vasculature to oestrogenic stimulation, a quantitative index
of the vascular permeability was obtained from the leakage
of radiolabelled albumin from the circulation (Arvidson,
1977; Milligan
et al
., 1998). Three and a half hours after s.c.
injection of the test substance (in 0.1 ml saline), 0.5 µCi
125
I-labelled human serum albumin (50 µl) was injected
into the jugular vein of the mice anaesthetized with
tribromoethanol. After 30 min, a blood sample was taken
from the suborbital canthal sinus with a heparinized
capillary pipette and the animals were killed by cervical
dislocation. The blood sample was centrifuged for 5 min at
1000
g
to provide a 100 µl plasma sample. The uteri and a
sample of thigh muscle were removed, washed briefly in
saline and then weighed. The radioactivity in the uterus,
plasma sample and muscle was determined. Previous
studies on the uterus (Arvidson, 1977; Milligan
et al
., 1998)
have indicated that the blood volume of the uterus is very
much smaller than the albumin space (after a circulation
time of 30 min) and that the extravascular albumin space is
the major determinant of the total tissue albumin space. The
tissue-specific extravascular albumin volume (EAV) was
expressed as a ratio of [
125
I] c.p.m. mg
–1
of tissue:[
125
I]
c.p.m. µl
–1
plasma and used as an index of tissue vascular
permeability. Each day s.c. injections of ICI 182,780
(100 µg (100 µl)
–1
arachis oil) were given to some animals
for 4 days before administration of 8-prenylnaringenin or
oestradiol to investigate whether the vascular responses
could be blocked by an anti-oestrogen.
In vivo
assays of vaginal and uterine mitosis and
uterotrophic response
The oestrogenic potency of 8-prenylnaringenin was
tested after continuous administration in the drinking water
by monitoring the uterotrophic response and cell mitoses in
the vaginal and uterine epithelia. Mice were allocated
randomly (
n
= 6 per treatment) to one of the following
treatment groups: (i) 100 ng oestrogen ml
–1
, (ii) 100, 20, 4
and 0.8 µg 8-prenylnaringenin ml
–1
or (iii) a drinking water
control of 1% (v/v) Tween 80 (polyoxyethylenesorbitan
monooleate) in water; the treatment bottles were weighed
and replaced each day. Uterine and vaginal cell proliferation
were assessed 72 h after the start of treatment. Mice were
injected i.p. with 0.1 ml colchicine (BDH, Poole) in saline
(1 mg ml
–1
) 2 h before they were killed. The uterine horns
were removed, blotted on tissue and weighed. The vaginae
and uterine horns were fixed in Bouin’s solution, embedded
in paraffin wax, cut into serial sections (5 µm), and stained
with haematoxylin and eosin. The number of cells under-
going mitosis in the entire uterine luminal epithelium and
random areas of the basal layer of the vaginal epithelium
were counted from four or more sections (at least 1000 cells
Oestrogenic activity of 8-prenylnaringenin
237
counted for each tissue). The mitotic index (% mitosis) was
calculated by the total number of mitoses divided by the
total number of cells counted 100.
Statistical analysis
Results were expressed as mean SEM and analysed
initially using one-way ANOVA and the Tukey’s multiple
comparison test. Data that did not pass either the normality
test or the equal variance test were analysed using the
Kruskal–Wallis one-way analysis on Ranks followed by
Dunn’s all pairwise multiple comparison test.
Results
Comparison of the oestrogenic activity of natural and
semi-synthetic 8-prenylnaringenin
Natural (purified from hops) and semi-synthetic 8-
prenylnaringenin showed equivalent oestrogenic activity in
both the Ishikawa Var-I assay and the yeast screen (Fig. 2).
The EC
50
values for oestradiol, semi-synthetic 8-prenyl-
naringenin and natural 8-prenylnaringenin were 0.82 0.01,
4.24 0.01 and 4.41 0.02 nmol l
–1
, respectively, in
the Ishikawa assay and 0.33 0.01, 43.7 1.4 and
40.0 1.3 nmol l
–1
, respectively, in the yeast screen. The
responses of Ishikawa Var-I cells were blocked completely
by 10 µmol ICI 182,780 l
–1
(data not shown). The
in vitro
oestrogenic potency of 8-prenylnaringenin was greater than
that of other established phyto-oestrogens in both assays.
The EC
50
values for oestradiol, 8-prenylnaringenin, 6-
prenylnaringenin, coumestrol, genistein and daidzein were
0.8, 4, 500, 30, 200 and 1500 nmol l
–1
, respectively, in the
Ishikawa cell assay, and 0.3, 40, > 4000, 70, 1200 and
2200 nmol l
–1
, respectively, in the yeast screen.
The oestrogenic activity of (R)- and (S)-8-prenylnaringenin
Resolution of 8-prenylnaringenin by chiral HPLC
revealed approximately equal amounts of the enantiomers
in both natural and semi-synthetic 8-prenylnaringenin (Fig.
3a). The circular dichroism spectrum of peak ‘A’ was almost
identical to that reported for (
S
)-naringenin (Gaffield, 1970),
whereas the spectrum of peak ‘B’ was the reflection of ‘A’.
The two enantiomers showed similar oestrogenic activity in
both the recombinant yeast cell and Ishikawa Var-I bioassays
(Fig. 4c). The enantiomers of 8-prenylnaringenin competed
strongly with oestradiol for binding to both ERα and ERβ
with a relative binding affinity of about 0.01 (oestradiol = 1)
(Fig. 4a,b).
The oestrogenic activity of 8-prenylnaringenin
in vivo
The uterine vascular responses to oestradiol, oestriol and
the phyto-oestrogens are shown (Fig. 4d). Oestriol was only
slightly less effective than oestradiol in inducing a rapid
increase in uterine vascular permeability within 4 h of
administration, but both 8-prenylnaringenin and coumestrol
were considerably less potent (< 1% relative to oestradiol).
The dose–response relationship for 8-prenylnaringenin was
similar to that of coumestrol, and a large stimulatory effect
was produced by 100 nmoles 8-prenylnaringenin. The
amount of genistein required to produce the same effect
was at least ten-fold greater. The administration of daidzein
produced no detectable uterine vascular response at the doses
used. The responses to 8-prenylnaringenin and oestradiol
were blocked completely by prior treatment of the animals
with the anti-oestrogen ICI 182,780 (Table 1). There were
no changes in the vascular permeability of the muscle tissue
control in any treatment group.
In vivo
assays of vaginal and uterine mitosis and
uterotrophic response
The addition of 8-prenylnaringenin or oestradiol to the
drinking water of mice is an effective way of providing
continuous, non-invasive oral administration of these
238
S. Milligan
et al.
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
10
–2
10
–1
10
0
10
1
10
2
10
3
10
4
Concentration (nmol l
–1
)
Absorbance at 405 nm
(a)
2.0
1.8
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
10
–2
10
–1
10
0
10
1
10
2
10
3
10
4
Concentration (nmol l
–1
)
Absorbance at 540 nm
(b)
Fig. 2. Relative oestrogenic activity of oestradiol (), 8-prenylnaringenin (semi-synthetic; ), 8-prenylnaringenin (natural; ) and other
phyto-oestrogens (coumestrol (), genistein (), daidzein ()) in (a) Ishikawa Var I cells and (b) yeast screen bearing the human oestrogen
receptor. Results are means
SEM;
n
= 6 wells per point. Where no error bars are visible, the errors were smaller than the symbols.
Oestrogenic activity of 8-prenylnaringenin
239
50
40
30
20
10
0
0.0 2.5 5.0 7.5 10.0 12.5 15.0
Time (min)
mAU
(a)
A
B
17.5
8-Prenylnaringenin
HO
O
OOH
OH
HO
OH O
O
OH
(
R
)-8-Prenylnaringenin(
S
)-8-Prenylnaringenin
(b)
Fig. 3. (a) Enantioseparation of (
S
)-8-prenylnaringenin (peak A) and (
R
)-8-prenylnaringenin (peak B). (b) Absolute structures of the
enantiomers.
2.0
1.8
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
10
–2
Concentration (nmol l
–1
)
Absorbance at 540 nm
(c)
10
–1
10
0
10
1
10
2
10
3
30
25
20
15
10
5
0
10
–12
Moles injected
Uterine vascular permeability (µl mg
–1
)
(d)
10
–11
10
–10
10
–9
10
–8
10
–6
10
–7
10
–5
120
100
80
60
40
20
0
10
0
Concentration (nmol l
–1
)
[
3
H]Oestradiol bound (%)
(a)
10
1
10
2
10
3
10
4
10
5
120
100
80
60
40
20
0
Concentration (nmol l
–1
)
[
3
H]Oestradiol bound (%)
(b)
10
0
10
1
10
2
10
3
10
4
10
5
ERα ERβ
Fig. 4. Competitive displacement of [2,4,6,7-
3
H]oestradiol from isolated (a) oestrogen receptor α (ERα) and (b) ERβ by oestradiol () and
(
R
)-8-prenylnaringenin () and (
S
)-8-prenylnaringenin (). (c) The relative oestrogenic activity of oestradiol, (
R
)-8-prenylnaringenin and
(
S
)-8-prenylnaringenin in the yeast screen bearing the transfected human oestrogen receptor (
n
= 4; mean SEM) and (d) the oestrogenic
activity of oestradiol (), oestriol (), 8-prenylnaringenin (), coumestrol (), genistein () and daidzein (), and the control () on
uterine vascular permeability in ovariectomized mice 4 h after s.c. administration (
n
= 6–10 per treatment). Vascular permeability was
expressed as (100) [
125
I] c.p.m. mg
–1
of tissue/[
125
I] c.p.m. µl
–1
plasma (mean SEM).
compounds. There was little variation in the amount of
liquid consumed between treatment groups (3.72 0.26 ml
per mouse per day) and on average, 15.9 mg 8-prenyl-
naringenin per kg per day was ingested by the treatment
group that consumed the highest concentration of 8-
prenylnaringenin. Both 8-prenylnaringenin (100 µgml
–1
)
and oestradiol (100 ng ml
–1
) produced significant increases
in vaginal mitosis after 72 h compared with the negative
control (
P
< 0.05 and
P
< 0.005, respectively; Fig. 5).
However, although oestradiol also produced significant
increases in uterine mass and in epithelial mitosis, there
were no significant differences in the mice exposed to any
of the 8-prenylnaringenin treatments (data not shown).
Discussion
Results from the present study confirm that 8-prenylnaringenin
is one of the most potent phyto-oestrogens known to date
(Milligan
et al
., 1999). The results from the
in vitro
and
in
vivo
bioassays and oestrogen receptor binding assays in the
present study and from that of Kitaoka
et al
. (1998) are
consistent with its oestrogenic activity being considerably
greater than that of genistein or daidzein. In the
in vitro
assays, the oestrogenic activity of 8-prenylnaringenin also
slightly exceeded that of coumestrol. The apparent differences
in potency of 8-prenylnaringenin between the yeast screen
and the Ishikawa cell assays are consistent with similar
observations comparing other compounds in these assays
(Le Gueval and Pakdel, 2001).
In addition to its
in vitro
activity, 8-prenylnaringenin
shows oestrogenic effects
in vivo
. 8-Prenylnaringenin
induces the rapid uterine vascular response typical of
oestrogens and this response is blocked by prior treatment
with an anti-oestrogen. The compound has been reported to
induce uterine growth and to suppress ovariectomy-induced
bone loss in ovariectomized rats when administered each
day s.c. at a dose of 30 mg per kg per day (Miyamoto
et al
.,
1998). In the present study, it was noted that 100 µg 8-
prenylnaringenin ml
–1
in the drinking water (equivalent to
an intake of about 15 mg per kg per day) was able to induce
the characteristic oestrogenic mitotic response in the vaginal
epithelium of ovariectomized mice. The apparent lack of
effect of 8-prenylnaringenin on uterine epithelial mitoses
may reflect either the limited amounts given or the temporal
differences in the mitotic responses induced by oestrogens
in the uterine luminal, uterine glandular and vaginal luminal
epithelia (Finn and Martin, 1973; Finn and Publicover, 1981).
Although it is difficult to compare the continuous exposure
of mice to 8-prenylnaringenin (via oral intake) experimentally
with human exposure to the compound via beer consump-
tion, it has to be noted that the oral concentrations required
to produce an oestrogenic effect in mice are about 500-fold
greater than can be found in any beer (Stevens
et al
., 1999;
Rong
et al
., 2000). In addition, many beers are now made
using extracts from hops or hop products rather than whole
hops: depending on the nature of the hop product used, these
beers have even lower concentrations of 8-prenylnaringenin
or contain no 8-prenylnaringenin at all (Stevens
et al
., 1999;
Rong
et al
., 2000).
A number of phyto-oestrogens (for example, coumestrol,
genistein) have a stronger binding affinity for ERβ than for
240
S. Milligan
et al.
Table 1. The ability of the anti-oestrogen ICI 182,780 to inhibit the increases in uterine vascular permeability (expressed as (100) [
125
I]
c.p.m. mg
–1
of tissue per [
125
I] c.p.m. µl
–1
plasma) induced by oestradiol and 8-prenylnaringenin
Treatment Uterus Muscle
Control (arachis oil) 4.70 0.84 0.53 0.02
ICI 182,780 pre-treatment 4.20 0.27 0.47 0.04
Oestradiol (10
–10
mol l
–1
) 12.53 1.17 0.53 0.07
ICI 182,780 pre-treatment + oestradiol (10
–10
mol l
–1
) 5.83 1.73*** 0.61 0.03
8-Prenylnaringenin (10
–7
mol l
–1
) 22.19 1.9 0.54 0.02
ICI 182,780 pre-treatment + 8-prenylnaringenin (10
–7
mol
–1
) 5.96 0.58*** 0.64 0.06
Values are mean SEM.
***Significantly different compared with treatment without ICI 182, 780 (
P
< 0.001;
n
= 6 mice per treatment).
10
8
6
4
2
0
Negative
control
Treatments in drinking water
Mitosis in vaginal epithelium (%)
0.8 µg ml
–1
8PN
4 µg ml
–1
8PN
20 µg ml
–1
8PN
100 µg ml
–1
8PN
100 ng ml
–1
E
2
**
*
Fig. 5. Mitotic response of the vaginal luminal epithelium of
ovariectomized mice to 8-prenylnaringenin (8PN) or oestradiol (E
2
)
administered for 72 h in drinking water. Data are mean
SEM
(
n
= 6 per treatment). Significant differences are denoted by
*
P
< 0.05, **
P
< 0.005 (Kruskal–Wallis test on ranks and Dunn’s
multiple comparison test).
ERα (Kuiper
et al.
, 1997; Casanova
et al.
, 1999), and ERβ
may be an important mediator for the action of phyto-
oestrogens. This study shows that 8-prenylnaringenin can
compete with oestradiol for binding with the ERα and ERβ,
although there was no marked difference in binding affinity
for the two receptors. The relative ligand binding affinities,
tissue distribution and developmental pattern of expression
of ERα and ERβ may help to explain the action of ER
agonists, such as phyto-oestrogens (Brandenburger
et al
.,
1997; Tetsuka
et al
., 1998). (
R
)- and (
S
)-prenylnaringenin
showed comparable activities both in the
in vitro
bioassays
and in the competitive binding assay to ERα and ERβ. The
enantiomers have also been shown to behave similarly in a
competitive binding assay with uterine oestrogen receptors
(Kitaoka
et al
., 1998).
Many natural flavanones prevail in the (
S
)-configuration
and are formed from the corresponding chalcones as a
consequence of the action of chalcone isomerase. However,
the lupulin glands of hops lack chalcone isomerase and the
presence of equal amounts of (
R
)- and (
S
)-8-prenylnaringenin
indicates that the compound may be derived from
xanthohumol or desmethylxanthohumol via a non-enzymatic
route (for example, thermal Michael-type cycloaddition).
However, it is also possible that the precursors are
converted to racemic 8-prenylnaringenin during drying,
storage and extraction of hops (Stevens
et al
., 1999; Rong
et al.
, 2000). The same may also be true for some other hop-
derived prenylated flavanones, as isoxanthohumol and
6-prenylnaringenin also occurred in both (
R
)- and (
S
)-forms
(data not shown). Further conversions and decompositions
may occur during the brewing process and such reactions
may account for the variable amounts of 8-prenylnaringenin
and other prenylflavonoids found in beer (Stevens
et al
.,
1999; Rong
et al
., 2000).
Although 8-prenylnaringenin has also been isolated from
a crude Thai drug derived from the heartwood of
Anaxagorea luzonensis
A. Gray (Annonaceae) (Kitaoka,
1998), hops probably represent the most significant source
of exposure of humans to the compound. In the past, hop
workers would have been exposed to this oestrogenic
compound via inhalation of hop dust or through
transcutaneous absorption rather than via dietary intake and
such exposure may provide an explanation for the
menstrual disturbances in female hop workers (Verzele,
1986). The reported ability of hop extracts to reduce hot
flushes in post-menopausal women (Goetz, 1990) is also
consistent with the presence of oestrogenic activity.
Although hops are used to impart flavour, bitterness and
other properties to beer, the amount of hops used in
brewing is relatively small and, therefore, 8-
prenylnaringenin is not a significant dietary component.
However, it is important to note that hops are now
incorporated into a number of herbal preparations for
women, including some claiming ‘breast enlargement’.
Claims for the effectiveness of at least some such ‘breast
enlargement’ preparations are partly based on their phyto-
oestrogen content. The concentration of 8-prenylnaringenin
in one type of these ‘breast-enhancing’ preparations is
10.9 µggm
–1
, which would result in a daily intake of
approximately 130 µg per day (Sauer and Coldham, 2001).
This value should be compared with the fact that some
beers have been reported to contain concentrations of
240 µg 8-prenylnaringenin l
–1
(although most are below
30 µgl
–1
; Stevens
et al
., 1999). Therefore, although moderate
beer intake could provide a daily intake of 8-prenylnaringenin
in the same range as that provided by some ‘breast
enhancement’ products, it should be noted that there are
no reports of clinical trials demonstrating either the
effectiveness or oestrogenic activity of such herbal
preparations in humans. Indeed, Sauer and Coldham (2001)
found that the supplement tested was inactive in a mouse
uterotrophic assay after administration either in the feed or
by s.c. injection. In addition, the present study showed that
the concentrations of 8-prenylnaringenin required to
produce oestrogenic responses in mice are 100 µgml
–1
,
that is, at least 400-fold greater than found in any beer.
Finally, although there may be interactions between alcohol
intake and the risk of breast cancer (Hulka and Moorman,
2001), there is no evidence linking beer intake with
breast enhancement. Other biological activities of 8-
prenylnaringenin have been described, including anti-
proliferative effects on breast and colon cancer cell lines,
inhibition of cytochrome P450-mediated activation of
procarcinogens, inhibition of bone resorption and inhibition
of diacylglycerol acyltransferase activity (Miranda
et al
.,
1999; Henderson
et al
., 2000). The biological effects, if any,
of consumption of 8-prenylnaringenin via beer or herbal
preparations remain to be clarified.
The authors are grateful to The Leverhulme Trust and to The
Royal Society for supporting this work.
References
Arvidson NG (1977) Early estrogen-induced changes in uterine albumin
exchange in mice
Acta Physiologica Scandinavica
100 325–331
Brandenberger AW, Tee MK, Lee JY, Chao V and Jaffe RB (1997) Tissue
distribution of estrogen receptors alpha (ER-alpha) and beta (ER-beta)
mRNA in the mid-gestational human fetus
Journal of Clinical
Endocrinology and Metabolism
82 3509–3512
Casanova M, You L, Gaido KW, Archibeque-Engle S, Janszen DB and Heck
HA (1999) Developmental effects of dietary phytoestrogens in
Sprague–Dawley rats and interactions of genistein and daidzein with rat
estrogen receptors alpha and beta
in vitro. Toxicological Sciences
51
236–244
Cassidy A and Milligan SR (1998) How significant are environmental
estrogens to women?
Climacteric
1 1–12
Fenselau C and Talalay P (1973) Is estrogenic activity present in hops?
Food
and Cosmetics Toxicology
11 597–603
Finn CA and Martin L (1973) Endocrine control of gland proliferation in the
mouse uterus
Biology of Reproduction
8 585–588
Finn CA and Publicover M (1981) Hormonal control of cell death in the
luminal epithelium of the mouse uterus
Journal of Endocrinology
91
335–340
Gaffield W (1970) Circular dichroism, optical rotatory dispersion and
absolute configuration of flavanones, 3-hydroxyflavanones and their
glycosides
Tetrahedron
26 4093–4108
Goetz P (1990) Traitement des bouffées de chaleur par insuffisance
Oestrogenic activity of 8-prenylnaringenin
241
ovarienne par l’extrait de houblon (
Humulus lupulus
).
Revue de
Phytothérapie Pratique
4 13–15
Henderson MC, Miranda CL, Stevens JF, Deinzer ML and Buhler DR (2000)
In vitro
inhibition of human P450 enzymes by prenylated flavonoids
from hops,
Humulus lupulus
.
Xenobiotica
30 235–251
Hesse R, Hoffman B, Karg H and Vogt K (1981) Untersuchungen über den
Nachweis von Phytoöstrogenen in Futterpflanzen und Hopfen mit Hilfe
eines Rezeptortests
Zentralblatt fur Veterinärmedizin
28 422–454
Hulka BS and Moorman PG (2001) Breast cancer: hormones and other risk
factors
Maturitas
38 103–113
Kitaoka M, Kadokawa H, Sugano M
et al.
(1998) Prenylflavonoids: a
new class of non-steroidal phytoestrogens (Part 1). Isolation of 8-
isopentenylnaringenin and an initial study on its structure–activity
relationship
Planta Medica
64 511–515
Knight DC and Eden JA (1996) A review of the clinical effects of
phytoestrogens
Obstetrics and Gynecology
87 897–904
Koch W and Heim G (1953) Hormone in hopfen und bier
Brauwissenschaft
8 132–133
Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S
and Gustafsson JA (1997) Comparison of the ligand binding specificity
and transcript tissue distribution of estrogen receptors alpha and beta
Endocrinology
138 863–870
Le Gueval R and Pakdel F (2001) Assessment of oestrogenic potency of
chemicals used as a growth promoter by
in vitro
methods
Human
Reproduction
16 1030–1036
Markiewicz L, Garey J, Adlercreutz H and Gurpide E (1993)
In vitro
bioassays of non-steroidal phytoestrogens
Journal of Steroid
Biochemistry and Molecular Biology
45 399–405
Milligan SR, Balasubramanian AV and Kalita JC (1998) Relative potency of
xenobiotic oestrogens in an acute
in vivo
mammalian assay
Environmental Health Perspectives
106 23–26
Milligan SR, Kalita JC, Heyerick A, Rong H, De Cooman L and
De Keukeleire D (1999) Identification of a potent phytoestrogen in hops
(
Humulus lupulus
L.) and beer
Journal of Clinical Endocrinology and
Metabolism
83 2249–2252
Miranda CL, Stevens JF, Helmrich A, Henderson MC, Rodriguez RJ,
Yang YH, Deinzer ML, Barnes DW and Buhler DR (1999)
Antiproliferative and cytotoxic effects of prenylated flavonoids from
hops (
Humulus lupulus
) in human cancer cell lines
Food and Chemical
Toxicology
37 271–285
Miyamoto M, Matsushita Y, Kiyokawa A, Fukuda C, Iijima Y, Sugano M and
Akiyama T (1998) Prenylflavonoids: a new class of non-steroidal
phytoestrogens (Part 2). Estrogenic effects of 8-isopentenylnaringenin on
bone metabolism
Planta Medica
64 516–519
Rong H, Zhao Y, Lazou K, De Keukeleire D, Milligan SR and Sandra P
(2000) Quantitation of 8-prenylnaringenin, a novel phytoestrogen in
hops (
Humulus lupulus
L.), hop products and beers, by benchtop HPLC
using electrospray ionization
Chromatographia
51 545–552
Routledge EJ and Sumpter JP (1996) Estrogenic activity of surfactants and
some of their degradation products assessed using a recombinant yeast
screen
Environmental Toxicology and Chemistry
15 241–258
Sauer MJ and Coldham NG (2001) Phyto-oestrogen content and
oestrogenic activity of a hop-containing food supplement
Reproduction
Abstracts Series
27 15
Stevens JF, Taylor AW and Deinzer ML (1999) Quantitative analysis of
xanthohumol and related prenylflavonoids in hops and beer by liquid
chromatography-tandem mass spectrometry
Journal of Chromatography
A
832 97–107
Tetsuka M, Milne M and Hillier SG (1998) Expression of oestrogen receptor
isoforms in relation to enzymes of oestrogen synthesis in rat ovary
Molecular and Cellular Endocrinology
141 29–35
Verzele M (1986) 100 years of hop chemistry and its relevance to brewing
Journal of the Institute of Brewing
92 32–48
Zenisek A and Bednar IJ (1960) Contribution to the identification of
estrogen activity in hops
American Perfumer Aromatics
75 61–62
Received 13 September 2001.
First decision 16 October 2001.
Accepted 2 November 2001.
242
S. Milligan
et al.
... The estrogenicity of hops was attributed to xanthohumol (XN) without any reported scientific data [2]. Preliminary studies on estrogenic potency of hop showed contradictory results: some studies revealed high estrogenic activity [4,5], while others found no or low estrogenic activity of hop [6]. These discrepancies might have been due to the varieties of extracts and the nature of assays used [5]. ...
... Preliminary studies on estrogenic potency of hop showed contradictory results: some studies revealed high estrogenic activity [4,5], while others found no or low estrogenic activity of hop [6]. These discrepancies might have been due to the varieties of extracts and the nature of assays used [5]. In 1999, Milligan et al. reported of a novel compound, , after a successful bioassay-guided fractionation of hop extracts, and provided the first evidence of its prominent estrogenic activity [7]. ...
... 8-PN is one of the most potent phytoestrogens currently known. This has been confirmed by a variety of in vitro assays for estrogenic activity (Table 2), including yeastbased screens expressing the human estrogen receptor (ER) and a reporter gene (usually luciferase, β-galactosidase or chloramphenicol acetyltransferase) under the control of estrogen-responsive sequences (ERE) [5,46,58,59], human cell lines responsive to estrogenic stimulation [5,7,60,61], and ER binding [5,7,61,62]. Compared with established phytoestrogens, such as coumestrol, genistein, and daidzein, the estrogenic activity of 8-PN has generally proven to be 0. , and 50-1500-fold stronger, respectively [5,7,59,62] (but see ERβ-specific data below), while concomitantly 5-250 times weaker than that of 17β-estradiol (E2) [5,7,46,[59][60][61][62][63][64]. ...
Article
8-prenylnaringenin (8-PN) is a prenylated flavonoid, occurring, in particular, in hop, but also in other plants. It has proven to be one of the most potent phytoestrogens in vitro known to date, and in the past 20 years, research has unveiled new effects triggered by it in biological systems. These findings have aroused the hopes, expectations, and enthusiasm of a “wonder-drug” for a host of human diseases. However, the majority of 8-PN effects require such high concentrations that they cannot be reached by normal dietary exposure, only pharmacologically; thus, adverse impacts may also emerge. Here, we provide a comprehensive and up-to-date review on this fascinating compound, with special reference to the range of beneficial and untoward health consequences that may ensue from exposure to it.
... As a novel phytoestrogen, 8-PN is unique in that its receptor specificity and potency is higher than any other phytoestrogens investigated thus far [9,10]. In vitro, 8-PN was shown to enhance differentiation and maturation of osteoblast and inhibit differentiation of osteoclast with intensities of response stronger than that observed with soy isoflavones [11]. ...
... The main mechanism of action of HE is the estrogenic activity of 8-PN, which has been demonstrated in vitro and in vivo and which has been recently extensively reviewed [9][10][11]54]. Along with its metabolites, 8-PN was only detected in the plasma and urine of women supplemented with HE, confirming adherence to treatment and an exposition to these active compounds in this group only. No correlation was found between the levels of total 8-PN and change in total body BMD after 48 weeks. ...
Article
Full-text available
Estrogen deficiency increases the risk of osteoporosis and fracture. The aim of this study was to investigate whether a hop extract standardized in 8-prenylnaringenin (8-PN), a potent phytoestrogen, could improve bone status of osteopenic women and to explore the gut microbiome roles in this effect. In this double-blind, placebo-controlled, randomized trial, 100 postmenopausal, osteopenic women were supplemented with calcium and vitamin D3 (CaD) tablets and either a hop extract (HE) standardized in 8-PN (n = 50) or a placebo (n = 50) for 48 weeks. Bone mineral density (BMD) and bone metabolism were assessed by DXA measurements and plasma bone biomarkers, respectively. Participant’s quality of life (SF-36), gut microbiome composition, and short-chain fatty acid (SCFA) levels were also investigated. In addition to the CaD supplements, 48 weeks of HE supplementation increased total body BMD (1.8 ± 0.4% vs. baseline, p < 0.0001; 1.0 ± 0.6% vs. placebo, p = 0.08), with a higher proportion of women experiencing an increase ≥1% compared to placebo (odds ratio: 2.41 ± 1.07, p < 0.05). An increase in the SF-36 physical functioning score was observed with HE versus placebo (p = 0.05). Gut microbiome α-diversity and SCFA levels did not differ between groups. However, a higher abundance of genera Turicibacter and Shigella was observed in the HE group; both genera have been previously identified as associated with total body BMD. These results suggest that an 8-PN standardized hop extract could beneficially impact bone health of postmenopausal women with osteopenia.
... The previous pharmacological studies reported that, the bioactive compounds Harmine (Patel et al., 2012), Bergapten (Santoro et al., 2016), 8-Prenylnaringenin (Milligan et al., 2002), Isoxanthohumol (Possemiers et al., 2006;Torrens-Mas and Roca, 2020), Bergamottin (He et al., 1998) and Colubrinic acid (Arai et al., 2008) having anticancer properties. Lactucopicrin a sesquiterpene lactone reported as a potent anti-malarial (against Plasmodium falciparum) compound which is also obtain in the roots of Cichorium intybus L. (Bischoff et al., 2004). ...
Article
Magnolia hodgsonii (Hook. f. & Thomson) H. Keng has been a traditionally important plant among the tribes of the Northeastern region of India. The present study aimed at the identification of this indigenous tree M. hodgsonii using DNA barcoding and analysis of their phytoconstituents using GC-MS and LC-MS/MS. The GC-MS analysis of petroleum ether extract (HSTP01_PET), detected 31 compounds including sesquiterpenes, monoterpenes, fatty acid methyl ester and lignan derivatives. Among them methyl 6,8-octadecadiynoate (33.58%), alpha-bisabolol (20.25%), 1h-cycloprop[e] azulene, decahydro-1,1,7-trimethyl (15.65%), cyclohexene, 3-(1,5-dimethyl-4-hex-enyl)-6-methylene (13.44%) and furan 2,5-bis(3,4-dimethoxyphenyl) tetrahydro-3,4-di (1.12%). In LC-MS/MS analysis, both methanolic (HSTM-01) and aqueous (HSTA-01) extracts showed the presence of phenols, flavo-noids, alkaloids, terpenes, coumarin and amino derivative compounds. Five bioactive compounds namely, 4-hy-droxy 5 (3′, 4′ dihydroxyphenyl) valeric acid, Harmine, (+)-Catechin-3-O glucose, Lysine-Betaxanthin and Rheumlhasol A were detected in both the extracts. The rbcL and matK sequence was employed as a molecular marker in DNA barcoding. The phylogenetic tree was generated in MEGA X using the maximum likelihood method, and the robustness of the tree was evaluated using bootstrapping with 1000 replicates. This study revealed that M. hodgsonii is closely related to M. pterocarpa and M. liliifera. This is the first study of the chemical components of essential oil isolated from M. hodgsonii leaves. The use of molecular barcoding in identifying M. hodgsonii as well as the chemical composition may form an important marker in the compilation of future herbal pharmacopeia for its proper identification and medicinal use.
... Two further aroma compounds found in this study were associated with the green spicy herbal aroma of hop cones are humulene and caryophyllene (Van Opstaele et al., 2013). Another spicy but also moderately sharp, fragrant, mint-or acetone-like aroma compound was identified as 2butanone, which was not described in earlier studies or reviews on hop aroma to our knowledge (Almaguer et al., 2014;Forster & Gahr, 2014;Milligan et al., 2002;Schönberger & Kostelecky, 2011;Su & Yin, 2021;Vázquez-Araújo et al., 2013). 2-butanone might be specific to this newly bred hop cultivar and could contribute to the passion fruit aroma in beers brewed with 'Callista' (Hanke et al., 2016). ...
Article
Hops (Humulus lupulus L.) are essential ingredients in brewing, contributing to beer’s flavor, aroma, and stability. This study pioneers an in-depth analysis of the 'Callista' cultivar, aiming to unravel how harvest timing, annual variations, and cultivation location synergistically influence its molecular profile, sensory perception, and biochemistry. Leveraging high-performance liquid chromatography and gas chromatography-mass spectrometry-olfactometry, we identified significant year-to-year and location-based fluctuations in bitter acids—the quintessential aroma constituents in hops. Our comprehensive aroma profiling discerned 55 volatile compounds, marking the first-ever sensory detection of 2-butanone in hops, with its presence showing remarkable interannual variability. This study showed significant differences among the three years tested, whereas hops were perceived “fruitier” and more “citrusy” in 2021, even though the bitter acid and aroma analysis showed that 2022 sticks out due to extremely high lupulone values up to 10% dry cone weight and 78% β-myrcene in the oil fraction compared to 60% and 45% in 2020 and 2021, respectively. Molecular analysis of key enzymes involved in hop aroma biosynthesis revealed no significant associations with location, but a strong diurnal pattern for all genes. The results indicated that especially the hot temperatures of 2022 may have induced significant changes of cone quality, while 2021 was more interesting from the sensory evaluations, which may justify the usage of viticultural terms such as “vintage” for hop marketing. These findings contribute to a better understanding of the factors influencing hop aroma and quality.
... Because 8-PN has been identified as a phytoestrogen that has potent ER-agonistic activity [31], the question of whether 8-PN displays its pharmacological effects in an ERdependent manner or not is an important issue for understanding its molecular mechanism. In contrast to most other phytoestrogens that show higher binding affinity for ERβ than for ERα, 8-PN has been shown to have a higher affinity for ERα in in vitro competitive binding assays and transfected yeast cells [32,33]. ...
Article
Full-text available
It has been shown that citrus flavanone naringenin and its prenyl derivative 8-prenylnaringenin (8-PN) possess various pharmacological activities in in vitro and in vivo models. Interestingly, it has been proposed that prenylation can enhance biological potentials, including the estrogen-like activities of flavonoids. The objective of this study was to investigate the anti-diabetic potential and molecular mechanism of 8-PN in streptozotocin (STZ)-induced insulin-deficient diabetic mice in comparison with naringenin reported to exhibit hypoglycemic effects. The oral administration of naringenin and 8-PN ameliorated impaired glucose homeostasis and islet dysfunction induced by STZ treatment. These protective effects were associated with the suppression of pancreatic β-cell apoptosis and inflammatory responses in mice. Moreover, both naringenin and 8-PN normalized STZ-induced insulin-signaling defects in skeletal muscles and apoptotic protein expression in the liver. Importantly, 8-PN increased the protein expression levels of estrogen receptor-α (ERα) in the pancreas and liver and of fibroblast growth factor 21 in the liver, suggesting that 8-PN could act as an ERα agonist in the regulation of glucose homeostasis. This study provides novel insights into the mechanisms underlying preventive effects of naringenin and 8-PN on the impairment of glucose homeostasis in insulin-deficient diabetic mice.
... The compound 4 has been previously extracted from hops, a bitter component present in beer, and exhibits various physiological activities, such as estrogenlike effect and muscle-aging inhibitory effect. 12,13 In addition, 3-hydroxy-5 ′ -methoxy-licoflavanone (6) exhibited the secondhighest antibacterial activity among the tested compounds. In these samples, the compounds with a few prenyl groups tended to exhibit relatively high antibacterial activities. ...
Article
Full-text available
Escherichia coli is an important bacterium for preventing food poisoning and biofilm infections. The emergence of antibiotic-resistant microorganisms necessitates the development of new antibiotics. The formation of bacterial biofilm is a drug-resistance mechanism utilized by diverse microorganisms. Therefore, it is important to identify compounds that can inhibit biofilm formation and cell survival, without triggering drug resistance. Herein, the antibacterial and antibiofilm activities of 2 types of Thai propolis (collected from Chiang Mai and Chanthaburi) against E. coli were investigated. The antibacterial activity was evaluated using the paper-disc method, while the minimum inhibitory concentration assay was performed using 2-fold serial dilution. Both types of Thai propolis and their isolated compounds showed antibacterial activity against E. coli (minimum inhibitory concentration: 32 µg/mL). The biofilm growth and development were assessed using a crystal violet solution. In particular, the extracts of the Chiang Mai propolis exhibited a significant antibiofilm formation activity against E. coli. Four prenylflavonoids, present in high proportions in the Chiang Mai propolis extracts, inhibited biofilm formation at low concentrations, contributing to the overall antibiofilm activity. These findings indicate that Thai propolis, a natural product, exhibits antibacterial and antibiofilm activities against E. coli.
Article
Estrogen receptors (ERs) are ligand-regulated transcription factors that modulate essential transcriptional programs by either promoting or repressing targeted gene expression. Given the impact of ER signaling on development, metabolism and physiology, it is no surprise to find impaired ER function as the basis of many disorders; thus, ERs have long been recognized as important biological and pharmaceutical targets. While 17b-estradiol (E2) is the main ER cognate ligand, ERs can be activated by diverse estrogen-mimicking compounds e.g. phytoestrogens, capable of binding receptors in a variable manner and influencing estrogen-dependent pathways, with both, beneficial and harmful health consequences. In this review we assessed current knowledge in the field of phytoestrogens as ER alternative ligands.
Article
The Expert Panel for Cosmetic Ingredient Safety (Panel) assessed the safety of Humulus Lupulus (Hops) Extract (reported functions include antimicrobial agent and hair conditioning agent) and Humulus Lupulus (Hops) Oil (reported function is fragrance). The Panel reviewed the relevant data related to these ingredients. Because final product formulations may contain multiple botanicals, each containing the same constituents of concern, formulators are advised to be aware of these constituents and to avoid reaching levels that may be hazardous to consumers. For these ingredients, the Panel was concerned about the presence of 8-prenylnaringenin, β-myrcene, and quercetin in cosmetics, which could result in estrogenic effects, dermal irritation, and genotoxicity, respectively. Industry should use current good manufacturing practices to limit impurities and constituents of concern. The Panel concluded that Humulus Lupulus (Hops) Extract and Humulus Lupulus (Hops) Oil are safe in cosmetics in the present practices of use and concentration when formulated to be non-sensitizing.
Article
Full-text available
Objective: Safety concerns or contraindications in the use of hormones have resulted in a rise in the use of nutritional medicinal products for the management of menopausal symptoms. The aim of the present study was to demonstrate the efficacy and safety of Exelvit Menopause®. Patients and methods: A prospective, open, observational, and multicentre study was performed, including 156 menopausal women. The patients received the nutritional product containing evening primrose oil 50 mg; hop extract 0.127-0.212 mg; saffron Stigmas Extract 0.6 mg; tryptophan 71.25 mg, vitamins B6, D3, K2, B12, and B9 once per day for 12 weeks. The validated menopausal rating score (MRS) was used for recording symptoms. Results: A decrease in the MRS of all menopausal symptoms was observed after 12 weeks compared to baseline (p < 0. 0001). Overall, hot flashes were reduced by 48.15%, heart discomfort by 33.3%, sleep disturbance by 46.2%, joint and muscular discomfort by 27.8%, depressive mood by 45.0%, irritability by 47.6%, anxiety by 44.4%, physical problems by 36.4%, sexual problems by 30.0%, bladder problems 31.3%, and vaginal dryness by 33.3%. Conclusions: The nutritional product Exelvit Menopause® significantly reduced menopausal symptoms.
Article
Full-text available
We demonstrated the anti-inflammatory and anti-oxidative effects of Humulus lupulus (HL) extract on solar simulator-irradiated primary human keratinocytes (PHKs) by analyzing ERK and p38 MAPK phosphorylation and production of IL-6 and IL-8. The anti-inflammatory effect of topically applied HL was further tested in vivo on human skin. To this end, we developed an oil-in-water (O/W) and a water-in-oil (W/O) cream with a lipid content of 40%. The anti-inflammatory effect of 1% HL extract incorporated in these two vehicles was assessed in a randomized, prospective, placebo controlled, double-blind UVB erythema study with 40 healthy volunteers. Hydrocortisone acetate (HCA) in the corresponding vehicle served as positive control. Surprisingly, both HL and HCA were only effective in the O/W system but not in the W/O formulation. Release studies using vertical diffusion cells (Franz cells) revealed that HCA was released in much higher amounts from the O/W cream compared to the W/O formulation. In summary, we have shown that 1% HL extract exerts anti-inflammatory effects comparable to 1% HCA, but only when incorporated in our O/W cream. Our findings confirm the critical role of the vehicle in topical anti-inflammatory systems.
Article
Full-text available
An estrogen-inducible screen was developed in yeast (Saccharomyces cerevisiae) in order to assess whether surfactants and their major degradation products are estrogenic. The DNA sequence of the human estrogen receptor (hER) was integrated into the yeast genome, which also contained expression plasmids carrying estrogen-responsive sequences (ERE) controlling the expression of the reporter gene lac-Z (encoding the enzyme β-galactosidase). Thus, in the presence of estrogens, β-galactosidase is synthesized and secreted into the medium, where it causes a color change from yellow to red. This recombinant strain was used to determine whether representatives of major surfactant classes and some of their principal degradation products possess estrogenic activity. The results were compared to the effects of the main natural estrogen 17β-estradiol. None of the parent surfactants tested possessed estrogenic activity. However, one class of surfactants, the alkylphenol polyethoxylates, degrade to persistent metabolites that were weakly estrogenic. Another group of degradation products, the sulfophenyl carboxylates, which are derived from the biodegradation of linear alkylbenzene sulfonates, do not appear to possess estrogenic activity.
Article
Chemical research has shown that the α-acids are the main quality factors of hops and that the bitter taste of hopped beer derives from the iso-α-acids. This has provided the basis for improvements in hop breeding, conditioning, extraction, use, analysis etc. Structural chemical development of hop constituents was completed about ten years ago. At the present moment the accent of chemical research is on analytical aspects, mainly using High Performance Liquid Chromatography. From the standpoint of the brewery the situation of chemical research is not completely satisfactory, since a clear understanding of the more subtle chemical quality factors of hops and their impact on beer quality is still lacking. Hops are much more important for beer than is generally estimated, even in brewery circles.
Article
Flavanones of 2S configuration and 3-hydroxyflavanones of 2R,3R configuration, having equatorial 2-aryl substituents in the former or diequatorial 2,3-substituents in the latter, exhibit a positive Cotton effect due to the n → π* transition (∼ 330 nm) and a negative Cotton effect in the n α π* region (∼ 280–290 nm). Flavanone glycosides possessing chiral aglycones show Cotton effects quite similar to their optically active aglycones while flavanone glycosides having racemic aglycones show only weak Cotton effects at 250–350 nm. The π → π* Cotton effect is more suitable for determining aglycone chirality in flavanone glycosides. In addition to the two high wavelength bands, 2S-flavanones generally showed a positive Cotton effect at 245–270 nm and a negative Cotton effect at 225–240 nm. trans 2R,3R-3-Hydroxyflavanones gave two Cotton effects below 270 nm both of which were positive.
Article
The effect of the oestrogens, oestradiol and oestriol, on plasma albumin dynamics in the uterus was studied in mice. A double isotope technique with 125I- and 131I-labelled human serum albumin was used. The optimum conditions to measure the extravascular accumulation of albumin were to use one labelled albumin as "permeability marker" (30 min circulation time) the other as plasma volume indicator (3 min circulation time). The oestrogens increased the extravascular accumulation of labelled albumin in the uterus but not in diaphragm or heart muscle. The accumulation was maximal after 4 h (75% higher than in controls) and was significant as early as 2 h after oestrogen administration. It cannot be decided whether the increase in labelled albumin accumulation was due to an increase in permeability and/or to an increase in the total surface area of perfused capillaries.
Article
Glandular epithelial cells of the mouse uterus divide in response to estrogens, but the pattern of response is different from that in the luminal epithelium. During continuous treatment the luminal epithelium shows maximum mitosis by 24 hr whereas the glands show little response until 72 hr and require three daily injections for maximum response. The incidence of mitosis then drops for the duration of treatment but there is a second wave of mitosis in the glands 72 hr after the cessation of estrogen administration. No such secondary wave occurs in the luminal epithelium. Progesterone inhibits both waves of glandular mitosis and the luminal mitosis.
Article
During the last 20 yr, several claims have been made for the presence of large quantities of oestrogens in hops and it has been alleged that this hormonal activity appears in beer. The oestrogenic activities reported were very high in comparison with those in other plants. A number of purified hop components, including the essential-oil fraction and the α-and β-bitter acids obtained from hop resins, have now been examined and various organicsolvent extracts of hops grown in the USA and in Europe have been prepared and tested for oestrogenic activity, both directly and following saponification in base. Commercial hop extracts were also tested for oestrogenic activity. Wide ranges of dilutions of all preparations were examined by the uterine-weight assay in the immature female mouse. No oestrogenic activity was detectable in any of these preparations.
Article
An attempt has been made to assess quantitatively the extent of cell death in the uterine epithelium after oestrogen treatment. [3H]Thymidine was injected into ovariectomized mice at an interval after oestrogen treatment when many of the luminal epithelial cells were in the S phase of mitosis. Uptake of [3H]thymidine was confirmed by autoradiography of sections of uterus and scintillation counting of trichloracetic acid-insoluble fraction of whole uterine horns. Radioactivity declined after the cessation of oestrogen treatment but remained high if treatment was continued. The decline appears to be correlated with the cell death previously demonstrated in histological sections of uteri under similar conditions.