Synergy in Herbal Medicines: Part 1
Eric Yarnell, ND, RH(AHG)a
©2015, Eric Yarnell, ND, RH(AHG)
Journal Compilation ©2015, AARM
DOI 10.14200/jrm.2015.4.0104
ABSTRACT
In the following paper, we will review the available literature on synergy and
additive effects involving medicinal herbs and herbal extracts. Several types of
synergistic interactions are discussed, including apparently inactive constituents
enhancing the effects of apparently active constituents within and between herbal
medicines, various herbal compounds altering the absorption of others, reduction
in toxicity of some herbal constituents by others, and direct synergistic therapeutic
effects when active constituents are combined within and between many medicinal
herbs. Species discussed include Artemisia annua (sweet Annie, q ng h o), Ammi
visnaga (khella), Glycyrrhiza glabra (licorice), G. uralensis (Chinese licorice, g
n c o), Panax ginseng (Asian ginseng, rén sh n), Mahonia aquifolium (Oregon
grape), Berberis aetnensis (Mt. Etna barberry), B. trifoliolata (algerita), B. fendleri
(Colorado barberry), and Coptis chinensis (goldthread, huáng lián). Part 2 of this
article will continue this review on other medicinal herb species.
Keywords: Synergy; Medicinal plants; Artemisia annua; Glycyrrhiza; Coptis
a
Corresponding author: Associate Professor, Department of Botanical Medicine, Bastyr University,
14500 Juanita Dr NE, Kenmore, WA 98028, USA. Chief Medical Oicer, Northwest Naturopathic Urology,
3670 Stone Wy N, Seattle, WA 98103, USA. Tel.: +1 425-602-3289;
E-mail:
[email protected]
Journal of Restorative Medicine 2015; 4: page 60
Herbal Synergy
INTRODUCTION
Medicinal plants contains hundreds if not thousands
of unique compounds. Models based on single
molecular entities do not accurately describe or
capture the complexity of interactions among the
constituents in medicine plants and multiconstituent
extracts made from them.
In the mainstream pharmaceutical view, there is
a single active constituent in a plant that explains
its activity and which can be isolated and used as
a conventional drug. This idea is reinforced by the
fact that many widely-used drugs are either single
molecules isolated from plants or semisynthetic
variants of natural molecules.1, 2 However, this
still does not prove that the single compounds are
superior to complex mixtures, particularly when
possible beneficial effects of various compounds
that indirectly support the main desired action have
not been assessed.
In many studies, an assay (such as receptor activation or inhibition, or killing of a microbe or cancer
cell in vitro) can be used to identify a single molecule or several molecules that appear to provide
most or all the effect of a crude plant extract for
that assay. For example, one fractionation assay
ultimately determined that berberine was the single
most active antimalarial constituent found in 14
different Vietnamese medicinal plants.3 This would
seem on the face of it to support the conventional
pharmacological model for medicinal plants.
However, these assays do not evaluate the complex
activities of other compounds in the plant that could
directly affect the outcome being assessed. This can
lead to severe oversights and a failure to recognize
“inactive” constituents as providing important functions. As cited below in depth, flavonoids found
in the same plant as berberine have been shown to
inhibit resistance to berberine in microbes. Thus,
while berberine may be the most important antimicrobial, removing the flavonoids could result in
microbial resistance developing toward berberine
and its failure as an antimicrobial. Activity-guided
modeling misses the benefit of other plant constituents on the overall outcome, as well as other
outcomes not being assessed.
This article will review the evidence on synergy of
multiple compounds in complex herbal extracts,
including those involving multiple herbs. Many
instances are reviewed where various types of
synergy have been studied with both supportive and
some negative results. The term synergy will be
used loosely in this review to include effects greater
than the sum of the parts (true synergy) and simple
additive effects. Overall, however, the current
research on many important medicinal herbs underappreciates the value of complex mixtures compared
with single isolated constituents.4
ARTEMISIA ANNUA (SWEET
ANNIE)
The prolific and widespread weed Artemisia annua
(sweet Annie, q ng h o) illustrates the problem of
activity-guided research overlooking useful effects
beyond the “active” constituents. The sesquiterpene lactone artemisinin (Figure 1) from A. annua
is a potent antimalarial schizonticide.5 Artemisinin
(qinghaosu in Chinese) and various semisynthetic
derivatives of it are among the most widely-used
antiplasmodial drugs in the world. It seems to be
largely assumed in mainstream medicine that this
is the beginning and ending of the sweet Annie
story: artemisinin is the silver bullet, and there is
nothing else worth investigating further in the herb.
However, a clinical trial showing a crude decoction
of A. annua that effectively eliminated symptoms
and dramatically lowered parasite burden in adults in
the Democratic Republic of the Congo with chronic
malaria, with cure rates on average of 74%, despite
providing far lower levels of artemisinin than are
used as an isolated drugs, suggesting that there is
more to A. annua than just artemisinin.6
Polymethoxylated flavonoids in A. annua also
have physiological activity (Table 1), though they
are supposedly not directly antiplasmodial (note
that at least one in vitro study did find artemetin
directly antiplasmodial).24 These flavonoids have
been shown to decrease resistance of Plasmodium
spp. to artemisinin by inhibiting efflux pumps in the
parasite.25 Chrysosplenol D (see Figure 1), chrysosplenitin, circilineol, casticin, and artemetin have
all been shown to significantly lower (by 20%–50%
Journal of Restorative Medicine 2015; 4: page 61
Herbal Synergy
OH
OH
O
O
H 3CO
O
O
H
O
H 3CO
OCH3
Artemisinin
OH
O
O
Chrysosplenol D
Figure 1: Artemisinin and chrysosplenol D.
depending on the compound) the half minimal
inhibitory concentration (IC50) of artemisinin
against Plasmodium spp. in vitro.26, 27 These
flavonoids did not reduce Plasmodium resistance
to chloroquine.26, 27
While artemisinin is readily and stably extracted
by aqueous decoction, flavonoids are best
extracted by alcohol.28 This is interesting in light
of in vitro evidence that only ethanolic and not
aqueous crude extracts of A. annua are antiplasmodial, and an absence of evidence of synergy in
A. annua aqueous extracts in vitro.29, 30 It is also
confusing given the human research on aqueous
infusions of A. annua which shows they are effective.6 It should be noted that traditional methods
of preparation of A. annua in Chinese medicine
by soaking combined with wringing or pounding
followed by juicing lead to extracts with 20 times
Table 1: Select other actions reported for polymethoxylated flavonoids found in Artemisia annua.
Flavonoid
Action
Model
References
Chrysosplenol D
Antioxidant
α-Glucosidase inhibitor
Antiangiogenic
Antiangiogenic
Growth inhibition by mitotic spindle
disruption
Antineoplastic and pro-apoptotic by many
mechanisms
In vitro
In vitro
In vitro
In vitro
Cervical cancer cells
in vitro
Breast, ovarian, lung,
colon, liver cancer cells
in vitro
Metoclopramide-treated
rats
In vitro Toll-like
receptor 2-activated
cells
In vitro
Xie et al., 20147
Ezzat and Salama, 20148
Zhu et al., 20139
Zhu et al., 20139
Kobayakawa et al., 200410
Casticin
Inhibit prolactin secretion by inhibiting
ERα and stimulating ERβ
Co-stimulates IL-1β secretion
Artemetin
Cyclooxygenase-2 inhibition, iNOS
inhibition, NF-κB inhibition
Lipoxygenase inhibition
Lipoxygenase inhibition
Anti-inflammatory
Antibacterial against Gram positive bacteria
Angiotensin converting enzyme inhibition
Hepatoprotective
Antiperoxidative
In vitro
In vitro
Carageenan-induced
edema in rat paws
In vitro
In vitro; normotensive
rats
Carbon tetrachloridetreated rats
In vitro
ER, estrogen receptor; IL, interleukin; iNOS, inducible nitric oxide synthase; NF, nuclear factor.
Journal of Restorative Medicine 2015; 4: page 62
Liu et al., 201411; Jiang
et al., 201312; Ono et al.,
200213; Yang et al., 201114
Ye et al., 201015
Lim et al., 201316
Liou et al., 201417
Choudhary et al., 200918
Choudhary et al., 200918
Sertié et al., 199019
Michielin et al., 200920
de Souza et al., 201121
Sridevi et al., 201222
Dugas et al., 200023
Herbal Synergy
higher levels of artemisinin than just infusion.31
Artemisinin content alone did not explain the
antimalarial activity of these traditional juiced
extracts in vitro; the IC50 concentrations were up
to 18 times lower for the crude juices than pure
artemisinin. One mouse study found that a crude
ethanol extract of A. annua had a median effective
dose (ED50) of 35 mg/kg vs. 122 mg/kg for pure
artemisinin.32 Short-term (3 day) use of this product in an open human trial was as effective, if not
more so, than chloroquine, though only 217 mg of
artemisinin were delivered over the total treatment
period, but had a high recrudescence rate (recurrent disease due to incomplete parasite killing).
Recrudescence was lowered by extending duration
of treatment or combining the A. annua capsule
with primaquine.32
An in vitro analysis found that A. annua infusion
with very low artemisinin concentrations (0.18%
dry weight) was equally active against chloroquineresistant and -sensitive strains of P. falciparum as
pure artemisinin.33 The authors speculated these
results are due either to the presence of other
antiplasmodial compounds or other synergistic
effects.33 Presumably these are compounds other
than the polymethoxylated flavonoids given the
low solubility of such flavonoids in water. Ongoing
work is needed to identify all the synergistic compounds in A. annua.
Other compounds in A. annua have activities that
may enhance its clinical utility for people with
malaria and other conditions beyond direct antimalarial activity. For instance, a mixture of many
sesquiterpene lactones from A. annua besides
artemisinin showed promising analgesic activity in
rodents.34 Qinghao acid, a sesquiterpene precursor
to artemisinin in A. annua, and the coumarin scopoletin were shown to contribute to the bacteriostatic
O
and inflammation-modulating effects of A. annua in
another report.35 Cinnamic acid derivatives (rosmarinic and chlorogenic acids in particular) from
A. annua inhibited the pro-inflammatory cytokines
IL-6 and -8 in vitro.36
Polymethoxylated flavonoids from A. annua
reduce resistance to other natural antimicrobials.
Combining chrysosplenol D or chrysosplenetin
from A. annua with subinhibitory concentrations
of the isoquinoline alkaloid berberine was effective in killing Staphylococcus aureus in vitro.37
Both flavonoids by themselves had minimal direct
antibacterial activity. This suggests another kind
of synergy: that which occurs when multiple herbs
are mixed together. Such formulation is a common
practice in most systems of herbal medicine around
the world and it has been suggested that various
other natural antimalarials should be combined and
tested in humans.38 Other herbal mixtures have been
shown to have synergy against malaria in vitro.39
Other examples of formulation synergy are presented elsewhere in this paper.
AMMI VISNAGA (KHELLA)
There is evidence of two types of synergy in the
native Mediterranean medicinal plant Ammi visnaga (khella). First, there is evidence of improved
absorption of furanocoumarins from crude extracts
than when they are given in isolation. Second,
there is evidence of activity from other constituents in the plant besides furanocoumarins, and
these other compounds enhance the activity of
furanocoumarins.
The most well-known spasmolytic furanocoumarin of Ammi visnaga is called khellin (Figure 2).
OCH3
O
OCH3
O
O
O
Khellin
OCH3
O
Visnagin
Figure 2: Khellin and visnagin.
Journal of Restorative Medicine 2015; 4: page 63
Herbal Synergy
Khellin is absorbed more rapidly and completely
when administered as part of a whole Ammi visnaga
extract than when given in isolation.40 Another
important furanocoumarin in Ammi visnaga, visnagin (see Figure 2), has also been shown to have
its absorption significantly enhanced when administered to 12 male Sprague-Dawley rats as a whole
plant extract compared with a pure compound, with
a ten-fold increase in area under the curve in serum
visnagin levels at the lowest dose tested (containing
0.625 mg visnagin/ml extract) of the extract compared to pure visnagin.41 This difference was highly
statistically significant (P = 0.005). Equal concentrations of visnagin were administered in both forms.
Whole extracts of Ammi visnaga were more
effective at inhibiting the mutagenicity of 2-aminoanthracene, 1-nitropyrene, and daunomycin than
isolated khellin in vitro.42 The identity of other constituents that were synergistic in this model were
not identified. In male Sprague-Dawley rats (n=8
per group) with hyperoxaluria induced by injection
of ethylene glycol and ammonium chloride, a crude
aqueous extract of Ammi visnaga fruits, but not
isolated khellin or visnagin, reduced urine oxalate
levels compared with untreated controls (P<0.01).43
The crude extract and isolated khellin and visnagin
all reduced calcium oxalate crystal deposition compared with untreated controls (P<0.05). However,
the crude extract also increased urinary citrate levels while decreasing urine oxalate excretion, but the
isolated constituents did not have these effects. In
vitro, an aqueous Ammi visnaga extract was superior to khellin or visnagin alone at protecting renal
epithelial cells from lysis due to calcium oxalate.44
These results mostly suggest other compounds in
Ammi visnaga are active with different and beneficial additive or synergistic effects besides khellin or
visnagin.
GLYCYRRHIZA GLABRA (LICORICE)
AND GLYCYRRHIZA URALENSIS
(GĀN C O)
The European/Central Asian Glycyrrhiza glabra
(licorice) and its close cousin from China, G.
uralensis (g n c o, Chinese licorice), are two of the
Journal of Restorative Medicine 2015; 4: page 64
most important and widely used herbs in the world.
One of their major uses, particularly in Chinese
medicine, is to act as a corrective assistant, meaning
to decrease the toxicity of other herbs with which
they are combined. This is an intriguing type of
formulation synergy that deserves further research,
though several examples can be cited involving G.
uralensis.
A classic example of this corrective assistant effect
is pairing G. uralensis with Aconitum spp. (aconite,
fù z ) prepared lateral roots to prevent cardiotoxicity. One in vitro study using rat cardiomyocytes
found that G. uralensis prevented the tendency of
Aconitum to increase pulse frequency of the cells,
and also inhibited the increase in lactate dehydrogenase induced by Aconitum.45 Adding Zingiber
officinale (ginger) rhizome extract to these two
herbs further enhances the protective effect of G.
uralensis. This creates a formula known in traditional Chinese medicine as sì nì t ng or “frigid
extremities decoction,” first described in the Sh
ng Hán Lùn (Discussion of Cold Damage) from
circa 220 CE that is actually cardioprotective.46–48
Another observation is that Aconitum by itself
induced cytochrome 3A4 in Sprague-Dawley rats
(n=3), resulting in a significantly decreased absorption of buspirone, with the area under the curve of
the drug reduced by 52.8% (P=0.02).49 This effect
was eliminated when G. uralensis was added to
Aconitum. One possible mechanism for protection
against the toxicity of Aconitum is by increasing the
rate of metabolism of the potentially toxic alkaloids
by G. uralensis.50
There are clear chemical differences between isolated extracts of Aconitum and a combination of G.
uralensis and Aconitum, suggesting that the protective effect of G. uralensis begins during preparation
and is not only due to pharmacologic effects.51, 52
There is also evidence of pharmacokinetic changes
in Aconitum alkaloids such as hypoaconitine
when taken in combination with G. uralensis and
Zingiber that could explain some of the protective
effects of this formulation.53
Another example of the corrective assistant effect
of G. uralensis is that it decreases hepatonephrotoxicity of Dioscorea bulbifera (huáng yào z , air
potato) tuber. When the two are boiled together,
the resulting mixture has lower levels of toxic
Herbal Synergy
HOOC
CH 3
O
CH 3
CH 3
CH 3
HOOC
O
HO
CH 3
HO
O
HOOC
O
O
H 3C
CH 3
Glycyrrhizin
HO
HO
OH
Figure 3: Glycyrrhizin.
compounds.54 Even better documented is both the
reduction of toxicity of the highly toxic immunosuppressive herb Tripterygium wilfordii (thunder
duke vine, léi g ng téng) decorticated root and
potentiation of its antiarthritic effects in a randomized clinical trial by the addition of G. uralensis,
compared with T. wilfordii alone.55 Although this
trial was not blinded, it still provides supporting
evidence for of this type of formulation synergy.
G. glabra and G. uralensis contain triterpenoid
saponins, notably glycyrrhizin (Figure 3). It is
believed that these constituents are the chemical basis for the historical use of these herbs as
solubilizing agents in herbal formulas to increase
absorption of other constituents, an important and
distinctive synergistic feature of herbal medicine.
This property is referred to as a guiding action in
traditional Chinese medicine, and G. uralensis is
the king of guide herbs. It appears in 50% of the
283 formulas in one of the most important materia
medica of Chinese medicine, the Shén Nóng B n C
o J ng (Divine Husbandman’s Classic of the Materia
Medica), compiled between 300 BCE and 200 CE.56
In one in vitro study, adding glycyrrhizin to an
aqueous solution increased the solubility of saikosaponin A from Bupleurum falcatum (thorowax,
chái hú) from 0.1 mg/mL to 5 mg/mL at standard
temperature and pressure.57 Saponins from other
plants have this same solubilizing effect. In the
same study, ginsenoside Ro from Panax ginseng
(Asian ginseng, rén sh n) increased the solubility of saikosaponin A in water from 0.1 mg/mL to
3.4 mg/mL. A combination of ginsenoside Ro and
the dammarane saponins of 20(S)-protopanaxadiol
from P. ginseng were more effective than ginsenoside Ro by itself at solubilizing multiple
saikosaponins in another in vitro study.58 Many
other studies confirm the solubilizing properties
of ginsenoside Ro, which may in part explain its
extremely widespread incorporation into traditional
Chinese herbal formulas; a similar explanation for
the widespread incorporation of G. uralensis in
such formulas is also likely.59–61
Another mechanism whereby G. uralensis (and
likely other saponin-rich herbs) can increase
absorption of various other herbal compounds is
by inhibition of P-glycoprotein (Pgp, also known
as multidrug resistance protein 1, or MDR1) in the
intestines. Pgp is a common efflux pump responsible for removal of a number of drugs and herbal
compounds from intestinal epithelial cells, thus preventing their absorption. Glycyrrhizin significantly
enhanced oral absorption of aconitine (AUC of
aconitine increased 61% with glycyrrhizin, P<0.01
compared with aconitine alone) from Aconitum
through inhibition of Pgp in male Sprague-Dawley
rats (n=5).62 Other licorice compounds such as
licochalcone A (Figure 4) also inhibit Pgp.63 This
is a clinical concern given the potential toxicity
of aconitine, but may be reconciled given all the
other information reviewed above about how G.
uralensis offsets the toxicity of Aconitum. Note that
this property of G. uralensis has been leveraged to
reduce drug resistance and amplify the utility of
various antimicrobial drugs.64 The licorice flavonoid
glabridin (see Figure 4) is itself a Pgp substrate
Journal of Restorative Medicine 2015; 4: page 65
Herbal Synergy
HO
O
Licochalcone A
OH
O
O
O
Glabridin
HO
OH
Figure 4: Licochalcone A and glabridin.
and thus a competitive inhibitor of Pgp, based on
a study looking at absorption of digoxin, another
Pgp substrate.65 Another study found that G. uralensis decoction in vitro had no effect on Pgp, but
after oral ingestion by Wistar rats (n=3) it mildly
increased absorption of rhodamine 123 (P<0.05
compared with controls) and thus confirming a mild
Pgp inhibitory effect.66 This study suggests some
process occurred during digestion that activated
compounds in the decoction.66 Other saponin-rich
herbs such as P. ginseng have also been shown to
be Pgp inhibitors.67
In combination with at least two herbs, crude
Chinese licorice (using the alternative species
Glycyrrhiza inflata) extracts have been shown
to synergistically inhibit Pgp in vitro in the case
of Daphne genkwa (genkwa, yuán hu ) and in
Wistar rats in the case of the endangered species
Euphorbia kansui (kan-sui, g n suì).68, 69 Both herbs
are powerful cathartic laxatives and the authors of
these studies commented that the synergistic inhibition of Pgp by these herbs could actually increase
the toxicity of these herbs by keeping the laxative
compounds in the gut longer allowing them to act
more potently. Both herbs are among the very few
that are traditionally regarded as becoming more
toxic in combination with licorice, a type of toxic
synergy. There is other evidence that G. uralensis
enhances the extraction of potentially toxic diterpenoids and other terpenoids from E. kansui and
Journal of Restorative Medicine 2015; 4: page 66
this might mediate the enhancement of its toxicity.70
Whatever the mechanism, these herbs should not be
combined.
Whole root aqueous extracts of G. glabra have
been shown to result in lower and more delayed
absorption of glycyrrhizin (described below)
and its aglycone glycyrrhetinic acid, and thus to
lower toxicity of the crude extract compared with
glycyrrhizin given in isolation.71, 72 Unidentified
lipophilic constituents appear to be responsible for
the effect of the whole root extract on glycyrrhizin
pharmacokinetics.
There are many other aspects of synergy in licorice
and G. uralensis. Table 2 lists a selection of many
other intriguing reports in this vein, including at
least one negative report.
MAHONIA AQUIFOLIUM (OREGON
GRAPE) AND BERBERIS SPP.
(BARBERRY)
Berberine (Figure 5), an isoquinoline alkaloid,
is traditionally regarded as the active constituent
from the roots of Mahonia aquifolium (Oregon
grape) and the many species in the genus Berberis.
However, multiple alkaloids are found in these species and multiple studies have found crude extracts
Herbal Synergy
Table 2: Other reports of synergy in Glycyrrhiza spp.
Comparators
Model
Result
References
Liquiritin apioside, liquiritin,
liquiritigenin, or all three combined
Capsaicin-induced
cough in guinea pigs
Combination inhibited cough significantly
more than any compound in isolation
Kamei
et al., 200573
Glycyrrhizin, glycyrrhetinic acids,
licorice extract
Multiple in vitro
mutagenicity assays
All were antimutagenic but licorice
extract had broadest effects
Zani et al.,
199374
Various flavonoids and chalcones,
G. uralensis extract
Granulomatous
inflammation in rodents
Isoliquiritin many times more potent
inhibitor than whole root extract
Kobayashi
et al., 199575
Glycyrrhizin, 18β-glycyrrhetinic acid
(18BGA), aqueous licorice extract
Respiratory syncytial
virus inhibition in vitro
Aqueous extract and 18BGA very active
while isolated glycyrrhizin inactive
Feng Yeh
et al., 201376
of the roots from various species more effective
than isolated alkaloids. Furthermore, non-alkaloid
compounds, including those found in the leaves,
have been shown to reduce bacterial resistance to
the alkaloids, suggesting a potential type of synergy
between different parts of the same medicinal plant.
found in the leaves of M. aquifolium (Figure 7)
with no intrinsic antistaphylococcal activity have
been shown to enhance this activity in berberine
from roots of the plant through inhibition of the
NorA efflux pump in Staphylococcus aureus.77
Another compound found in M. aquifolium leaves,
pheophorbide A (Figure 8), a natural degradation product of chlorophyll, was also a strong
resistance pump inhibitor in S. aureus, greatly
magnifying the antibacterial activity of berberine in vitro.78 The flavonolignan silymarin from
Silybum marianum (milk thistle) seed was also
very active as a resistance pump inhibitor enhancing the efficacy of berberine in this study.
Methyoxylated flavonolignans such as
5′-methoxyhydnocarpin (Figure 6) primarily
O
N+
O
B. trifoliolata (algerita) leaf also contains
5′-methoxyhydnocarpin and B. fendleri (Colorado
barberry) leaf also contains pheophorbide A with
drug efflux pump-inhibitor effects in S. aureus.79
B. aetnensis (Mt. Etna barberry) has also been
shown to contain similar pump inhibitors in its
OCH3
Berberine
OCH3
Figure 5: Berberine.
O
O
OH
HO
O
O
OH
5′-Methoxyhydnocarpin
OH
O
O
Figure 6: 5′-Methoxyhydnocarpin.
Journal of Restorative Medicine 2015; 4: page 67
Herbal Synergy
COPTIS CHINENSIS (GOLDTHREAD)
Figure 7: Mahonia aquifolium leaves.
leaves as that of M. aquifolium and to also inhibit
the efflux of ciprofloxacin from drug-resistant S.
aureus, thereby enhancing the efficacy of the drug.80
This research suggests that combining some fruit or
leaf of M. aquifolium with the root would optimize
efficacy, at least when treating patients with staphylococcal infections.
A crude ethanol extract of B. vulgaris (barberry)
was more effective than isolated alkaloids or alkaloid fractions in rodent models of acute and chronic
inflammation.81 A crude methanol extract of the root
of B. aetnensis was more active against Candida
albicans and other pathogenic species of Candida
in vitro than an alkaloid fraction or pure berberine.82
These findings suggest a need for human research
on the synergistic effects of not only whole root
extracts, but also root/leaf extracts of Berberis and
Mahonia spp.
Figure 8: Pheophorbide A.
Journal of Restorative Medicine 2015; 4: page 68
Another berberine-containing medicinal species
studied for synergistic properties that is frequently
used in Chinese medicine, is Coptis chinensis
(goldthread, huáng lián) rhizome. Total extracts of
the rhizome are more active hypoglycemics than
an alkaloid-only fraction of C. chinensis.83 A combination of four C. chinensis alkaloids (berberine,
coptisine, palmatine, and epiberberine) were more
hypoglycemic and less toxic to liver cells than
berberine by itself.84, 85 An alkaloid-enriched extract
of C. chinensis killed 100% of male and female
Kunming mice (n=20) at a dose of 0.28 g/kg; no
mice were killed by a total ethanol extract of C.
chinensis at this dose.86 The LD50 of the total extract
was 2950 mg/kg in rats, but it was 160–210 mg/kg
for the alkaloid-enriched extract used in this study.
Statistical significance was not reported. While
berberine and total aqueous extract of C. chinensis
rhizome both limited hepatic damage from carbon
tetrachloride in male Sprague-Dawley rats, the
total extract was more effective (statistical significance not stated).87 In a similar study, the aqueous
rhizome extract and berberine had similar efficacy
at preventing carbon tetrachloride liver damage in
rats.88 The reason for the differences between these
two studies is unknown, as the extracts and experimental designs were very similar.
The mixture of six parts C. chinensis and one part
Tetradium rutaecarpa (evodia, wú zh yú) fruit
is known as Zu J n Wán, “Left Metal Pill,” in
Chinese medicine. It originated in 1481 CE in
D n X X n F (Essential Teachings of Dan-Xi) and
is used to treat a wide range of digestive problems
ranging from ulcers and esophageal reflux to nausea
and vomiting. Administration of this formula to rats
resulted in a complex pharmacokinetic interaction,
notably an increase in absorption of major important compounds from T. rutaecarpa and a decrease
in bioavailability of alkaloids from C. chinensis,
notably coptisine and berberine.89, 90 Pretreatment
of rats with T. rutaecarpa fruit aqueous extract for
2 weeks also resulted in significant reduction in
berberine bioavailability from C. chinensis.91 The
mechanism appeared to be through the constituents
of T. rutaecarpa inducing hepatic UGT1A1, increasing first pass metabolism of berberine. C. chinensis
is commonly combined with T. rutaecarpa as a
Herbal Synergy
corrective assistant because C. chinensis is “cold”
while T. rutaecarpa is “hot.”92 This pharmacokinetic
outcome could be the desired result of the combination though it is difficult to be certain at this time.
One other line of evidence suggesting a balancing
effect of these two herbs in combination comes
from an in vitro study looking at the effects of C.
chinensis and T. rutaecarpa separately and together
on bovine adrenal medullary cells.93 The two herbs
had diametrically opposed effects, with the alkaloids of C. chinensis inhibiting secretion from the
cells and the alkaloids of T. rutaecarpa stimulating
it. While this might suggest they simply cancel each
other out and it would make little sense to combine
them, it is more likely given the endurance of this
herb pair in Chinese herbal medicine that the effect
is balancing and beneficial in some way.
There are other hints in the literature that combining these two herbs as Zu J n Wán may be
superior to either alone. Evodiamine is an alkaloid
found in T. rutaecarpa with multiple antineoplastic actions against gastric cancer cells in vitro, but
it also increases IL-8 secretion by the cells, which
is associated with increased risk of metastasis.
Berberine blocked this effect.94 Berberine and
evodiamine in combination were significantly better at inducing apoptosis in human hepatocellular
carcinoma cells in vitro than either compound
alone.95 All these results suggest a possible benefit
of combining these two herbs in cancer patients.
medicinal herbs. Some of these are straightforward, and with multiple compounds with similar
actions showing greater efficacy together than
any one of them in isolation, such as the hypoglycemic activities of alkaloids in C. chinensis.16, 17
Others are more subtle and complex; for instance,
showing that compounds can indirectly enhance
the efficacy of other compounds with direct
effects, such as the methyloxylated flavonolignans in M. aquifolium leaf reducing resistance in
Staphylococcus aureus to berberine in M. aquifolium root.9 Effects on pharmacokinetics between
herbal compounds can be beneficially inhibitory (as in the case of the interaction between T.
rutaecarpa and C. chinensis)21–23 or enhancing (as
is the case with G. uralensis and many compounds).78 Another type of synergy is reduction of
toxicity of one herb by another, as demonstrated
with G. uralensis and several very different types
of herbs.62, 71, 72
Further research is warranted to determine the
extent and importance of these effects, particularly in humans, as much of the existing research
is preclinical. Currently herbal medicine is still
practiced by many practitioners based on the
idea that these synergistic effects are relevant,
as herbal formulas are still in widespread use.
Further verifying that this approach is clinically
useful and determining optimal formulations
would be of great benefit to many patients around
the globe.
CONCLUSION
DISCLOSURE OF INTEREST
Numerous lines of inquiry demonstrate synergistic and additive effects among and between
Dr. Yarnell reports a financial interest in Heron
Botanicals, outside the submitted work.
REFERENCES
1.
Samuelsson G. Drugs of Natural Origin: A
Textbook of Pharmacognosy, 4th edn. Stockholm:
Apotekarsocieteten; 1999.
2.
Evans WC. Trease and Evans’ Pharmacognosy, 13th
edn. London: Baillière Tindall; 1989.
3.
Tran QL, Tezuka Y, Ueda JY, et al. In vitro antiplasmodial activity of antimalarial medicinal plants used in
Vietnamese traditional medicine. J Ethnopharmacol.
2003;86(2–3):249–52.
4.
Wagner H. Synergy research: approaching a new generation
of phytopharmatceuticals. Fitoterapia. 2011;82(1):34–7.
5.
Yarnell E. Artemisia annua (sweet Annie), other
Artemisia species, artemisinin, artemisinin derivatives,
and malaria. J Restorative Med. 2014;3(1):69–84.
Journal of Restorative Medicine 2015; 4: page 69
Herbal Synergy
6.
Mueller MS, Runyambo N, Wagner I, et al.
Randomized controlled trial of a traditional preparation
of Artemisia annua L (annual wormwood) in the
treatment of malaria. Trans R Soc Trop Med Hyg.
2004;98(5):318–21.
7.
Xie XY, Chen FF, Shi YP. Simultaneous determination of
eight flavonoids in the flowers of Matricaria chamomilla
by high performance liquid chromatography. J AOAC
Int. 2014;97(3):778–83.
8.
Ezzat SM, Salama MM. A new α-glucosidase inhibitor
from Achillea fragrantissima (Forssk) Sch Bip growing
in Egypt. Nat Prod Res. 2014;28(11):812–8.
9.
Zhu XX, Yang L, Li YJ, et al. Effects of sesquiterpene,
flavonoid and coumarin types of compounds from
Artemisia annua L on production of mediators of angiogenesis. Pharmacol Rep. 2013;65(2):410–20.
10.
Kobayakawa J, Sato-Nishimori F, Moriyasu M,
Matsukawa Y. G2-M arrest and antimitotic activity
mediated by casticin, a flavonoid isolated from Viticis
Fructus (Vitex rotundifolia Linne fil). Cancer Lett.
2004;208(1):59–64.
20.
Michielin EM, Salvador AA, Riehl CA, et al. Chemical
composition and antibacterial activity of Cordia
verbenacea extracts obtained by different methods.
Bioresour Technol. 2009;100(24):6615–23.
21.
de Souza P, Gasparotto A Jr, Crestani S, et al.
Hypotensive mechanism of the extracts and artemetin
isolated from Achillea millefolium L (Asteraceae) in rats.
Phytomedicine. 2011;18(10):819–25.
22.
Sridevi VK, Chouhan HS, Singh NK, Singh
SK. Antioxidant and hepatoprotective effects of
ethanol extract of Vitex glabrata on carbon tetrachloride-induced liver damage in rats. Nat Prod Res.
2012;26(12):1135–40.
23.
Dugas AJ Jr, Castañeda-Acosta J, Bonin GC, et al.
Evaluation of the total peroxyl radical-scavenging capacity of flavonoids: structure-activity relationships. J Nat
Prod. 2000;63(3):327–31.
24.
Ortet R, Prado S, Regalado EL, et al. Furfuran lignans
and a flavone from Artemisia gorgonum Webb and
their in vitro activity against Plasmodium falciparum. J
Ethnopharmacol. 2011;138(2):637–40.
11.
Liu LP, Cao XC, Liu F, et al. Casticin induces breast
cancer cell apoptosis by inhibiting the expression of forkhead box protein M1. Oncol Lett.
2014;7(5):1711–7.
25.
Elford BC, Roberts MF, Phillipson JD, Wilson RJ.
Potentiation of the antimalarial activity of qinghaosu
by methoxylated flavones. Trans R Soc Trop Med Hyg.
1987;81(3):434–6.
12.
Jiang L, Cao XC, Cao JG, et al. Casticin induces ovarian
cancer cell apoptosis by repressing FoxM1 through the
activation of FOXO3a. Oncol Lett. 2013;5(5):1605–10.
26.
Liu KCS, Yang SY, Roberts MF, et al. The contribution
of flavonoids to the antimalarial activity of Artemisia
annua. Planta Med. 1989;55(7):654–5.
13.
Ono M, Yanaka T, Yamamoto M, et al. New diterpenes
and norditerpenes from the fruits of Vitex rotundifolia.
J Nat Prod. 2002;65(4):537–41.
27.
Liu KC, Yang SL, Roberts MF, et al. Antimalarial activity of Artemisia annua flavonoids from whole plants and
cell cultures. Plant Cell Rep. 1992;11(12):637–40.
14.
Yang J, Yang Y, Tian L, et al. Casticin-induced
apoptosis involves death receptor 5 upregulation in
hepatocellular carcinoma cells. World J Gastroenterol.
2011;17(38):4298–307.
28.
Weathers PJ, Towler MJ. The flavonoids casticin
and artemetin are poorly extracted and are unstable
in an Artemisia annua tea infusion. Planta Med.
2012;78(10):1024–6.
15.
Ye Q, Zhang QY, Zheng CJ, et al. Casticin, a flavonoid isolated from Vitex rotundifolia, inhibits prolactin
release in vivo and in vitro. Acta Pharmacol Sin.
2010;31(12):1564–8.
29.
Ferreira JF, Peaden P, Keiser J. In vitro trematocidal
effects of crude alcoholic extracts of Artemisia annua,
A. absinthium, Asimina triloba, and Fumaria officinalis:
trematocidal plant alcoholic extracts. Parasitol Res.
2011;109(6):1585–92.
16.
Lim EK, Mitchell PJ, Brown N, et al. Regiospecific
methylation of a dietary flavonoid scaffold selectively
enhances IL-1β production following Toll-like receptor 2 stimulation in THP-1 monocytes. J Biol Chem.
2013;288(29):21126–35.
30.
Mouton J, Jansen O, Frédérich M, van der Kooy F. Is
artemisinin the only antiplasmodial compound in the
Artemisia annua tea infusion? An in vitro study. Planta
Med. 2013;79(6):468–70.
31.
Wright CW, Linley PA, Brun R, et al. Ancient
Chinese methods are remarkably effective for the
preparation of artemisinin-rich extracts of Qing
Hao with potent antimalarial activity. Molecules.
2010;15(2):804–12.
32.
Wan YD, Zang QZ, Wang JS. Studies on the antimalarial
action of gelatin capsule of Artemisia annua. Zhongguo
Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi.
1992;10(4):290–4 [in Chinese].
33.
De Donno A, Grassi T, Idolo A, et al. First-time comparison of the in vitro antimalarial activity of Artemisia
17.
Liou CJ, Len WB, Wu SJ, et al. Casticin inhibits COX-2
and iNOS expression via suppression of NF-κB and
MAPK signaling in lipopolysaccharide-stimulated mouse
macrophages. J Ethnopharmacol. 2014;158PA:310–6.
18.
Choudhary MI, Azizuddin, Jalil S, et al.
Antiinflammatory and lipoxygenase inhibitory
compounds from Vitex agnus-castus. Phytother Res.
2009;23(9):1336–9.
19.
Sertié JA, Basile AC, Panizza S, et al. Anti-inflammatory
activity and sub-acute toxicity of artemetin. Planta Med.
1990;56(1):36–40.
Journal of Restorative Medicine 2015; 4: page 70
Herbal Synergy
annua herbal tea and artemisinin. Trans R Soc Trop Med
Hyg. 2012;106(11):696–700.
46.
Li Y, Fu CM, Ren B, et al. Study on attenuate and synergistic mechanism between aconiti lateralis praeparata
radix and glycyrrhizae radix for toxicity reduction based
on metabonomic of MI-RI mouse cardiomyocytes.
Zhongguo Zhong Yao Za Zhi. 2014;39(16):3166–71 [in
Chinese].
47.
Zhao MQ, Wu WK, Zhao DY, et al. Protective effects of
sini decoction on adriamycin-induced heart failure and
its mechanism. Zhong Yao Cai. 2009;32(12):1860–3 [in
Chinese].
48.
Liu J, Peter K, Shi D, et al. Traditional formula,
modern application: Chinese medicine formula sini
tang improves early ventricular remodeling and cardiac
function after myocardial infarction in rats. Evid Based
Complement Alternat Med. 2014;2014:141938.
49.
Zhang G, Zhu L, Zhou J, et al. Effect of aconiti laterlis
radix compatibility of glycyrrhizae radix on CYP3A4 in
vivo. Zhongguo Zhong Yao Za Zhi. 2012;37(15):2206–9
[in Chinese].
50.
Shen H, Wu J, Di LQ, et al. Enhancement by
Glycyrrhizae Radix of hepatic metabolism of hypaconitine, a major bioactive and toxic component of Aconiti
Laterlis Radix, evaluated by HPLC-TQ-MS/MS analysis.
Biomed Chromatogr. 2013;27(5):556–62.
51.
Zhang JM, Li L, Gao F, et al. Chemical ingredient
analysis of sediments from both single Radix Aconiti
Lateralis decoction and Radix Aconiti Lateralis – Radix
Glycyrrhizae decoction by HPLC-MS. Yao Xue Xue Bao.
2012;47(11):1527–33 [in Chinese].
52.
Yang Y, Yin XJ, Guo HM, et al. Identification and
comparative analysis of the major chemical constituents in the extracts of single fuzi herb and fuzi-gancao
herb-pair by UFLC-IT-TOF/MS. Chin J Nat Med.
2014;12(7):542–53.
34.
Favero F de F, Grando R, Nonato FR, et al. Artemisia
annua L: evidence of sesquiterpene lactones’ fraction
antinociceptive activity. BMC Complement Altern Med.
2014;14:266.
35.
Huang L, Liu JF, Liu LX, et al. Antipyretic and antiinflammatory effects of Artemisia annua L. Zhongguo
Zhong Yao Za Zhi 1993;18:44–8, 63–4 [in Chinese].
36.
Melillo de Magalhães P, Dupont I, Hendrickx A,
et al. Anti-inflammatory effect and modulation of
cytochrome P450 activities by Artemisia annua tea
infusions in human intestinal Caco-2 cells. Food Chem.
2012;134(2):864–71.
37.
Stermitz FR, Scriven LN, Tegos G, Lewis K. Two
flavonols from Artemisa annua which potentiate the
activity of berberine and norfloxacin against a resistant strain of Staphylococcus aureus. Planta Med.
2002;68(12):1140–1.
38.
Rasoanaivo P, Wright CW, Willcox ML, Gilbert B.
Whole plant extracts versus single compounds for the
treatment of malaria: synergy and positive interactions.
Malar J. 2011;10(Suppl 1):S4.
39.
Gathirwa JW, Rukunga GM, Njagi EN, et al.
The in vitro anti-plasmodial and in vivo anti-malarial
efficacy of combinations of some medicinal plants
used traditionally for treatment of malaria by the
Meru community in Kenya. J Ethnopharmacol.
2008;115(2):223–31.
40.
Eder M, Mehnert W. Plant excipients–valuable pharmaceutical aids or superfluous ballast? Pharm Unserer Zeit.
2000;29(6):377–84 [in German].
41.
Haug KG, Weber B, Hochhaus G, Butterweck V.
Pharmacokinetic evaluation of visnagin and Ammi
visnaga aqueous extract after oral administration in rats.
Planta Med. 2012;78(17):1831–6.
53.
Schimmer O, Rauch P. Inhibition of metabolic
activation of the promutagens, benzo[a]pyrene,
2-aminofluorene and 2-aminoanthracene by furanochromones in Salmonella typhimurium. Mutagenesis.
1998;13(4):385–9.
Zhang W, Zhang H, Sun S, et al. Comparative pharmacokinetics of hypaconitine after oral administration of pure
hypaconitine, Aconitum carmichaelii extract and sini
decoction to rats. Molecules. 2015;20(1):1560–70.
54.
Vanachayangkul P, Chow N, Khan SR, Butterweck V.
Prevention of renal crystal deposition by an extract of
Ammi visnaga L and its constituents khellin and visnagin
in hyperoxaluric rats. Urol Res. 2011;39(3):189–95.
Ma L. Studies on the Changes of the Main Compounds in
Co-boiling Extracts, Pooled Individual Boiling Extracts
in Dioscorea bulbifera L and Glycyrrhiza uralensis Fisch
Decoction (Thesis, Fujian University of Traditional
Chinese Medicine), 2011.
55.
Li YS, Tong PJ, Ma HZ. Toxicity attenuation and
efficacy potentiation effect of liquorice on treatment of rheumatoid arthritis with Tripterygium
wilfordii. Zhongguo Zhong Xi Yi Jie He Za Zhi.
2006;26(12):1117–9 [in Chinese].
56.
Wang X, Zhang H, Chen L, et al. Liquorice, a unique
‘guide drug’ of traditional Chinese medicine: a review
of its role in drug interactions. J Ethnopharmacol.
2013;150(3):781–90.
57.
Nie RL, Tanaka T, Miyakoshi M, et al. A triterpenoid
saponin from Thladiantha hookeri var pentadactyla.
Phytochemistry. 1989;28(6):1711–5.
42.
43.
44.
45.
Vanachayangkul P, Byer K, Khan S, Butterweck V. An
aqueous extract of Ammi visnaga fruits and its constituents khellin and visnagin prevent cell damage caused
by oxalate in renal epithelial cells. Phytomedicine.
2010;17(8–9):653–8.
Xie S, Zhang G, Sun G, et al. Detoxication experimental
study on different compatibility proportion of Aconiti
Lateralis Radix Praeparata and glycyrrhizae radix et rhizoma. Zhongguo Zhong Yao Za Zhi. 2012;37(15):2210–4
[in Chinese].
Journal of Restorative Medicine 2015; 4: page 71
Herbal Synergy
58.
Wantanabe K, Fujino H, Morita T, et al. Solubilization
of saponins of bupleuri radix with ginseng saponins:
cooperative effect of dammarane saponins. Planta Med.
1988;54(5):405–9.
59.
Kimata H, Sumida N, Matsufuji N, et al. Interaction
of saponin of bupleuri radix with ginseng
saponin: solubilization of saikosaponin-a with
chikusetsusaponin-V (=ginsenoside Ro). Chem Pharm
Bull. 1985;33(7):2849–53.
60.
Zhou X, Kasai R, Yoshikawa M, et al. Solubilization
of saponins of Bupleuri radix with Ginseng saponins:
effect of malonyl-ginsenosides on water solubility of
saikosaponin-b. Chem Pharm Bull. 1991;39(5):1250–2.
61.
Guo J, Shang E, Zhao J, et al. Data mining and
frequency analysis for licorice as a ‘two-face’ herb in
Chinese formulae based on Chinese Formulae Database.
Phytomedicine. 2014;21(11):1281–6.
62.
Chen L, Yang J, Davey AK, et al. Effects of diammonium glycyrrhizinate on the pharmacokinetics
of aconitine in rats and the potential mechanism.
Xenobiotica. 2009;39(12):955–63.
63.
Nabekura T, Hiro T, Kawasaki T, Uwai Y. Effects of
natural nuclear factor-kappa B inhibitors on anticancer
drug efflux transporter human P-glycoprotein. Biomed
Pharmacother. 2015;70:140–5.
64.
Bhattacharjee A, Majumder S, Majumdar SB, et al.
Co-administration of glycyrrhizic acid with the antileishmanial drug sodium antimony gluconate (SAG) cures
SAG-resistant visceral leishmaniasis. Int J Antimicrob
Agents. 2015;45(3):268–77.
71.
Cantelli-Forti G, Maffei F, Hrelia P, et al. Interaction
of licorice on glycyrrhizin pharmacokinetics. Environ
Health Perspect. 1994;102(suppl 9):65–8.
72.
Wang Z, Nishioka M, Kurosaki Y, et al.
Gastrointestinal absorption characteristics of glycyrrhizin from Glycyrrhiza extract. Biol Pharm Bull.
1995;18(9):1238–41.
73.
Kamei J, Saitoh A, Asano T, et al. Pharmacokinetic and
pharmacodynamic profiles of the antitussive principles
of Glycyrrhizae radix (licorice), a main component of
the Kampo preparation Bakumondo-to (Mai-men-dongtang). Eur J Pharmacol. 2005;507(1–3):163–8.
74.
Zani F, Cuzzoni MT, Daglia M, et al. Inhibition
of mutagenicity in Salmonella typhimurium by
Glycyrrhiza glabra extract, glycyrrhizinic acid, 18α- and
18β-glycyrrhetinic acids. Planta Med. 1993;59(6):502–7.
75.
Kobayashi S, Miyamoto T, Kimura I, Kimura M.
Inhibitory effect of isoliquiritin, a compound in licorice
root, on angiogenesis in vivo and tube formation in vitro.
Biol Pharm Bull. 1995;18(10):1382–6.
76.
Feng YC, Wang KC, Chiang LC, et al. Water extract of
licorice had anti-viral activity against human respiratory
syncytial virus in human respiratory tract cell lines. J
Ethnopharmacol. 2013;148(2):466–73.
77.
Stermitz FR, Lorenz P, Tawara JN, et al. Synergy in a
medicinal plant: antimicrobial action of berberine potentiated by 5’-methoxyhydnocarpin, a multidrug pump
inhibitor. Proc Natl Acad Sci. 2000;97(4):1433–7.
78.
Stermitz FR, Tawara-Matsuda J, Lorenz P, et al.
5’-Methoxyhydnocarpin-D and pheophorbide a: Berberis
species components that potentiate berberine growth
inhibition of resistant Staphylococcus aureus. J Nat
Prod. 2000;63(8):1146–9.
79.
Stermitz FR, Beeson TD, Mueller PJ, et al.
Staphylococcus aureus MDR efflux pump inhibitors
from a Berberis and a Mahonia (sensu strictu) species.
Biochem Syst Ecol. 2001;29(8):793–8.
65.
Cao J, Chen X, Liang J, et al. Role of P-glycoprotein in
the intestinal absorption of glabridin, an active flavonoid
from the root of Glycyrrhiza glabra. Drug Metab Dispos.
2007;35(4):539–53.
66.
Yao HW, Fu XY, Xie QD, et al. Effect of liquorice
decoction on rat intestinal P-glycoprotein. Nan Fang Yi
Ke Da Xue Xue Bao. 2009;29(8):1571–3 [in Chinese].
67.
Zhang J, Zhou F, Wu X, et al. 20(S)-Ginsenoside Rh2
noncompetitively inhibits P-glycoprotein in vitro and
in vivo: a case for herb-drug interactions. Drug Metab
Dispos. 2010;38(12):2179–87.
80.
Musumeci R, Speciale A, Costanzo R, et al. Berberis
aetnensis C Presl extracts: antimicrobial properties and
interaction with ciprofloxacin. Int J Antimicrob Agents.
2003;22(1):48–53.
68.
Huang BB, Li GF, Ren F, et al. Effect of Glycyrrhiza
inflata and Daphne genkwa on permeabilities of rhodamine 123, a P-glycoprotein substrate across rat jejunum
membranes in vitro. Zhongguo Zhong Yao Za Zhi.
2008;33(21):2521–6 [in Chinese].
81.
Ivanovska N, Philipov S. Study on the anti-inflammatory action of Berberis vulgaris root extract, alkaloid
fractions and pure alkaloids. Int J Immunopharmacol.
1996;18(10):553–61.
82.
69.
Sun YB, Li GF, Tang ZK, Wu BY. Modulation on
the P-glycoprotein in the jejunum by combined use
of Glycyrrhiza inflata and kansui. Yao Xue Xue Bao.
2010;45(4):510–6 [in Chinese].
Iauk L, Costanzo R, Caccamo F, et al. Activity of
Berberis aetnensis root extracts on Candida strains.
Fitoterapia. 2007;78(2):159–61.
83.
Chen HY, Ye XL, Cui XL, et al. Cytotoxicity and
antihyperglycemic effect of minor constituents
from Rhizoma Coptis in HepG2 cells. Fitoterapia.
2012;83(1):67–73.
84.
Zhu J, Xin FQ, Chen X, et al. Synergism of alkaloids
from Coptis Rhizoma. Shi Zhen Med Mater Med Res.
2010;21:2282–4 [in Chinese].
70.
Shen J, Mo X, Tang Y, et al. Analysis of herb-herb
interaction when decocting together by using ultrahigh-performance liquid chromatography-tandem
mass spectrometry and fuzzy chemical identification
strategy with poly-proportion design. J Chromatogr A.
2013;1297:168–78.
Journal of Restorative Medicine 2015; 4: page 72
Herbal Synergy
85.
Yi J, Ye X, Wang D, et al. Safety evaluation of main
alkaloids from Rhizoma Coptidis. J Ethnopharmacol.
2013;145(1):303–10.
86.
Ma BL, Ma YM, Shi R, et al. Identification of the toxic
constituents in Rhizoma Coptidis. J Ethnopharmacol.
2010;128(2):357–64.
87.
Feng Y, Wang N, Ye X, et al. Hepatoprotective effect
and its possible mechanism of Coptidis rhizoma
aqueous extract on carbon tetrachloride-induced
chronic liver hepatotoxicity in rats. J Ethnopharmacol.
2011;138(3):683–90.
88.
Ye X, Feng Y, Tong Y, et al. Hepatoprotective effects
of Coptidis rhizoma aqueous extract on carbon
tetrachloride-induced acute liver hepatotoxicity in rats.
J Ethnopharmacol. 2009;124(1):130–6.
89.
Yan R, Wang Y, Shen W, et al. Comparative pharmacokinetics of dehydroevodiamine and coptisine in rat plasma
after oral administration of single herbs and Zuojinwan
prescription. Fitoterapia. 2011;82(8):1152–9.
90.
Jia X, Jiang J, Chen B, et al. Progress in research
of processing of fructus Evodiae-Rhizoma
Coptidis and research thoughts and methods of its
processing mechanism. Zhongguo Zhong Yao Za Zhi.
2009;34(10):1314–7 [in Chinese].
91.
Ma BL, Yao MK, Han XH, et al. Influences of
Fructus evodiae pretreatment on the pharmacokinetics of Rhizoma coptidis alkaloids. J Ethnopharmacol.
2011;137(3):1395–401.
92.
Dharmananda S. Evodia: Traditional and Modern Uses.
Institute for Traditional Medicine, Portland, OR. http://
www.itmonline.org/articles/evodia/evodia.htm [accessed
19 Feb 2015], 2010.
93.
Zhao FR, Mao HP, Zhang H, et al. Antagonistic
effects of two herbs in Zuojin Wan, a traditional
Chinese medicine formula, on catecholamine secretion in bovine adrenal medullary cells. Phytomedicine.
2010;17(8–9):659–68.
94.
Shi HL, Wu XJ, Liu Y, Xie JQ. Berberine counteracts
enhanced IL-8 expression of AGS cells induced by
evodiamine. Life Sci. 2013;93(22):830–9.
95.
Wang XN, Han X, Xu LN, et al. Enhancement of
apoptosis of human hepatocellular carcinoma SMMC7721 cells through synergy of berberine and evodiamine.
Phytomedicine. 2008;15(12):1062–8.
Journal of Restorative Medicine 2015; 4: page 73