Abstract
Cimicifuga racemosa(L.) Nutt. (syn. Actaea racemosa L., black cohosh) is used to relieve menopausal hot flashes, although clinical studies have provided conflicting data, and the active constituent(s) and mechanism(s) of action remain unknown. Since serotonergic receptors and transporters are involved with thermoregulation, black cohosh and its phytoconstituents were evaluated for serotonergic activity using 5-HT7 receptor binding, cAMP induction, and serotonin selective reuptake inhibitor (SSRI) assays. Crude extracts displayed 5-HT7 receptor binding activity and induced cAMP production. Fractionation of the methanol extract lead to isolation of phenolic acids and identification of Nω-methylserotonin by LC/MS-MS. Cimicifuga triterpenoids and phenolic acids bound weakly to the 5-HT7 receptor with no cAMP or SSRI activity. In contrast, Nω-methylserotonin showed 5-HT7 receptor binding (IC50 23 pM), induced cAMP (EC50 22 nM), and blocked serotonin reuptake (IC50 490 nM). These data suggest Nω-methylserotonin may be responsible for the serotonergic activity of black cohosh.
Keywords: black cohosh, N-methylserotonin, Cimicifuga, menopause, serotonin
INTRODUCTION
Women can expect to spend one third of their lives in the post-menstrual phase, with an estimated 55–75% of women experiencing symptoms such as hot flashes, depression, mood swings, sleep deprivation, and loss of sexual drive (1). Among those women suffering these symptoms, approximately 20% will find the symptoms intolerable, and up to 30% of these women will seek out treatment through their healthcare providers (1, 2). Traditionally, the standard treatment recommended to perimenopausal and post-menopausal women to relieve some of these symptoms was hormone replacement therapy (HRT). However, the high HRT usage was prior to the release of the Women’s Health Initiative findings (3, 4), which showed that women on HRT had an increased risk of developing various hormone-dependent cancers (1, 3). Alternatives to traditional HRT are needed to alleviate menopausal symptoms without adverse side effects, leading millions of women to turn to botanical dietary supplements (5). Cimicifuga racemosa (L.) Nutt. (syn. Actaea racemosa L. (Ranunculaceae), black cohosh) is a popular and well documented medicinal plant, native to eastern North America. The roots and rhizomes have many traditional ethnobotanical uses, including relief from rheumatism, as a diuretic, a tonic, an astringent, and for gynecological disorders (1, 6). In addition, black cohosh extracts have become a popular treatment for vasomotor menopausal symptoms, potentially providing an alternative to HRT (1, 6). However, some clinical trials have found no effect of black cohosh extracts on reducing the vasomotor symptoms of menopause (4, 7), although other studies have shown efficacy in alleviating symptoms such as hot flashes, sweating, mood swings, irritability, and sleeplessness (4, 7–9). Conflicting clinical trial results emphasize that the effects of black cohosh on menopausal symptoms and the mechanism(s) of action remain unclear.
Originally, black cohosh was thought to work through estrogenic pathways similar to traditional HRTs (1, 4, 10, 11). An estrogen-like activity was reported initially (4); however, these effects could not be confirmed in later studies which showed no classical estrogenic activity (1, 7, 12, 13). For example, a previous study demonstrated that black cohosh exhibited no estrogenic or antiestrogenic effects in an ovariectomized rat model (14). Interestingly, selective serotonin reuptake inhibitors (SSRIs), which operate through the serotonin transporter (SERT), have been shown to alleviate hot flashes (2). Besides the SERT, the 5-HT7 and 5-HT1A serotonin receptors are also involved in thermoregulation, which suggests that agonists for these receptors might be beneficial for the alleviation of hot flashes (15). Accordingly, the serotonergic system was examined as an alternative pathway through which black cohosh may reduce menopausal hot flashes. Black cohosh extracts had an affinity for various serotonin receptors, particularly the 5-HT1A, 5-HT1D, and 5-HT7 (14). The black cohosh extracts also produced receptor-mediated functional activity as demonstrated by induction of cAMP in the human embryonic kidney (HEK) cells stably over-expressing the 5-HT7 receptor. Data reporting agonistic activity for the 5-HT7 receptor (14) support the need for in vitro evaluation of black cohosh extracts in order to determine the active constituents responsible for serotonergic activity.
The in vivo efficacy of a black cohosh extract for relief of menopausal hot flashes is currently being evaluated in a four-arm randomized double-blind, placebo-controlled Phase II clinical trial (16). The same clinical extract was tested for in vitro serotonergic activity by measuring 5-HT7 receptor competitive binding, the induction of cAMP in 5-HT7-transfected HEK293 cells, and the ability to block the reuptake of serotonin into hSERT-transfected HEK293 cells in the SSRI assay. In addition to the black cohosh clinical extract, several pure compounds isolated from black cohosh, such as triterpenes and polyphenolics (Figure 1) were tested, as well as other fractions obtained through bioassay-guided fractionation (Figure 2).
MATERIALS AND METHODS
Materials and Reagents
All chemicals and reagents were from Fisher (Hanover Park, IL) or Sigma-Aldrich (St. Louis, MO), unless otherwise indicated. All cell culture media were obtained from Invitrogen (Carlsbad, CA). Fetal bovine serum (FBS) was acquired from Biowest (Miami, FL), and dialyzed FBS was acquired from Gemini Bioproducts (West Sacramento, CA). The purity of all reference compounds, including radiochemicals, was determined based on certificates of analysis (CoA) provided by the respective suppliers, and included with the compounds. [3H] Lysergic acid diethylamide (LSD, CoA purity ≥ 99%) was obtained from PerkinElmer Life Sciences (Boston, MA). [3H] 5-Hydroxytryptamine trifluoroacetate ([3H] 5-HT, CoA purity ≥ 99%) was purchased from GE Healthcare UK (Piscataway, NJ). Poly-D-lysine hydrobromide was obtained from BD Biosciences and had a CoA purity > 98% (Bedford, MA). The quantitative 1H NMR (qHNMR) analysis method (17) was used to verify compound purity. 3-(2-Aminoethyl)indol-5-ol (serotonin) as a hydrochloride salt was from Sigma-Aldrich (purity: CoA ≥ 98%; determined by qHNMR > 99% with 0.2% of structurally unrelated impurities). 3-(2-Methylaminoethyl)indol-5-ol (Nω-methylserotonin) was obtained as oxalate salt from Sigma-Aldrich (purity: CoA > 99%; determined by qHNMR > 97.5%, with 2.5% of structurally unrelated impurities) and was used in the stated bioassays.
Plant Materials
Due to the large scale of the overall study, which included a phase II clinical trial, authentic C. racemosa (L.) Nutt. (syn. Actaea racemosa L., black cohosh) rhizomes/roots (BC #163) were acquired through our industrial partner, Naturex, formerly PureWorld Botanicals (South Hackensack, NJ) (18) and were botanically verified by the UIC/NIH Center for Botanical Dietary Supplements Research and characterized by PCR, vouchers, and microscopy (19). Further details describing the botanical and phytochemical authentication of C. racemosa have been communicated (18). Voucher specimens of the plant materials are deposited at Naturex (South Hackensack, NJ) as follows: 75% EtOH extract (BC #163); MeOH extract (BC #066). Milled roots/rhizomes of black cohosh were extracted with 75% ethanol by large-scale percolation, vacuum dried at 45 °C and 29–30 inches of vacuum pressure, and milled through 60-mesh screen to yield a powdered extract. The chemical characterization was performed by HPLC (20), and the extract was standardized to 5.6% of four triterpene glycosides (18). The triterpene glycoside reference materials were isolated and characterized as previously described (18, 21, 22).
Bioassay-guided fractionation
For the bioassay-guided fractionation of black cohosh, which is outlined in Figure 2, dried and milled rhizomes/roots (1 kg) of C. racemosa were exhaustively extracted using percolation with 11 L of fresh MeOH (22 × 500 mL) at room temperature, with TLC monitoring. The extract was concentrated in vacuo (< 40 °C) to yield 173 g of syrupy residue. The residue was reconstituted in de-ionized water (250 mL) and partitioned with ethyl acetate (20 × 400 mL; 42 g). The water partition was subjected to column chromatography consisting of Amberlite XAD-2, yielding 2 fractions (H2O and MeOH). A portion of the MeOH-soluble fraction (2.8 g) was further subjected to pH zone refined fast centrifugal partition chromatography (FCPC) using water-butanol-ethylacetate (5:4:1) as the solvent-system. The lower phase was used as mobile phase, adding 2 mg ammonium carbonate per mL phase-volume. 0.2% TFA was added to the stationary phase prior to loading the column. The sample was dissolved in stationary phase and injected, immediately afterwards mobile phase was pumped through the column at a flow-rate of 4 mL/min in a head-to-tail mode. The eluate was combined into seven fractions (1: 0 – 16 mL, 2: 16 – 32 mL, 3: 32 – 40, 4: 40 – 112 mL, 5: 112 –136 mL, 6: 136 – 388 mL, 7: 388 – 508 mL). Fraction 4 contained the cimicifugic acids, which are unique compounds in Cimicifuga species. To obtain pure cimicifugic acids, fraction 4 was subjected to a second step of FCPC in a HemWat +5 system [hexane-ethylacetate-methanol-water (3:7:3:7)] adding 0.2% TFA to both phases (23). The column was equilibrated before injecting the sample, which was dissolved in a mixture of both phases and run in a tail to head mode. The flow-rate was set to 4 mL/min (fraction size 4 mL). Solvents were switched to extrusion mode at K = 3 (24), and yielded the cimicifugic acids A, B, E, F and fukinolic acid at K values of K = 3.4 – 3.7, K = 4.8 – 6.0, K = 1.3 – 1.4, K = 2.2 – 2.5, and K = 9.6 – 23.0, respectively, where K represents the distribution constant of the analyte (25). The extraction progression and collected fractions were evaluated using analytical thin layer chromatography (TLC) on 20 × 20 cm pre-coated silica gel 60 F254 glass plates, (Merck, Darmstadt, Germany). Plates were developed at room temperature in the solvent system EtOAc/HCOOH/H2O (18:1:1) and monitored under UV light (254 and 365 nm) both before and after spraying with Naturstoff reagent (1% 2-aminoethyl-phenylborinate in MeOH). Additional details regarding the extraction procedure have been published separately (26).
Identification of Nω-methylserotonin
Nω-methylserotonin was identified by mass spectrometry by means of accurate mass measurements in combination with a CrossFire Commander (Mimas, The University of Manchester, Manchester, UK) database search and comparison with an authentic sample of the compound. Briefly, the XAD-MeOH fraction 7 (Figure 2), active in the 5-HT7 binding assay, was separated on a Tosoh TSK-Gel Amide-80 column using a gradient from 90–65% acetonitrile/0.1% formic acid over 60 min at a flow rate of 0.2 mL/min. The eluent from the column was introduced into a Micromass (Manchester, UK) Q-TOF-2 hybrid quadrupole/time-of-flight mass spectrometer operated in positive ion electrospray mode. The resolving power was 7000 FWHM. The mass accuracy obtained was better than 10 ppm unless noted otherwise. The ion source parameters were as follows: capillary 3.3 kV, cone voltage 23 V, source block temperature 100 °C, and drying gas temperature 300 °C. Tandem mass spectra were acquired at a quadrupole setting of 13, which corresponds to slightly less than unit resolution of the quadrupole. Tandem mass spectra were acquired at a collision energy of 23 eV using argon as the collision gas at a pressure of 2.0 × 10−5 mBar. The XAD-MeOH fraction 7 and Nω-methylserotonin were also analyzed using an isocratic HPLC method and LC-MS based on a 50 mm × 4.6 mm i.d., 1.8 μm, Zorbax Eclipse C18 (Agilent ) column, as well as the gradient HPLC method with LC-MS-MS based on a YMC ODS column to further support the method of unambiguous compound identification.
Cell Culture Conditions
The human 5-HT7-transfected Chinese hamster ovary (CHO) cell line was generously provided by Dr. David Sibley (National Institutes of Health, Bethesda, MD) and cultured with Ham’s F-12 medium containing 10% FBS, 1% sodium pyruvate, 1% geneticin (50 mg/mL), and 1% penicillin/streptomycin. The 5-HT7-transfected-HEK293 cell line was provided by Dr. Mark Hamblin (University of Washington, Seattle, WA). The 5-HT7-transfected-HEK293 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with puromycin (10 μg/mL), 1% antibiotic-antimycotic, and 10% FBS. Four days before the cAMP assays were performed, the culture medium was replaced with Cellgro Complete serum-free medium from Mediatech (Herndon, VA) supplemented with puromycin (10 μg/mL) and 1% antibiotic-antimycotic. The HEK293 cell line stably expressing human serotonin transporter (hSERT) was a generous gift from Dr. Randy D. Blakely (Vanderbilt University, Nashville, TN). hSERT-transfected-HEK293 cells were maintained in DMEM supplemented with 10% dialyzed FBS, 1% penicillin-streptomycin, 1% glutamax, and 1.2% geneticin (50 mg/mL). Cells were grown in a 37 °C humidified environment with 5% CO2.
Serotonin Receptor Competitive Binding Assay
Cells (human 5-HT7 CHO) were plated into dishes (150 mm × 10 mm) and cultured to confluence in order to collect membranes as previously described (27). Briefly, a hypotonic buffer (15 mM Tris, 1.25 mM MgCl2, and 1 mM EDTA, pH 7.4) was added to the dishes and incubated at 4 °C for 10–15 min. The cells were scraped from the dishes on ice and the lysate was centrifuged. The hypotonic buffer was removed and the membrane pellet was suspended in TEM buffer (75 mM Tris, 12.5 mM MgCl2, 1 mM EDTA, pH 7.4). The cell membranes were homogenized and centrifuged twice at 12,000 g for 20 min. The pellets were dissolved in TEM buffer and stored at −80 °C. Protein concentrations were determined according to the protein assay method using bovine serum albumin (BSA) as the standard from Bio-Rad Laboratories (Hercules, CA).
The 5-HT7 receptor binding assay was performed as described previously (27) with minor modifications using human recombinant CHO cell membrane and [3H] LSD (5.67 nM) in an incubation buffer (50 mM Tris, 10 mM MgCl2, 0.5 mM EDTA, pH 7.4). The plant extracts, tested at 50 μg/mL, were considered active if binding was inhibited by 50% or more. After a 1 h incubation at 37 °C, the mixture was filtered over a 934-AH Whatman filter that had been presoaked in 0.5% polyethylenimine (PEI) and washed three times in ice-cold wash buffer (50 mM Tris buffer, pH 7.4) and simultaneously washed and aspirated five times using a 96-well Tomtec-Harvester (Orange, CT). Each filter was dried, suspended in Wallac microbeta plate scintillation fluid (PerkinElmer Life Sciences, Boston, MA), and counted with a Wallac 1450 Microbeta liquid scintillation counter (PerkinElmer Life Sciences, Boston, MA). Serotonin (1 μM) was used to define nonspecific binding, which accounted for < 10% of total binding. The percent inhibition of [3H] ligand bound to each 5-HT7 receptor was determined using Equation 1.
(1) |
Serotonergic evaluation of Nω-methylserotonin against the other 5-HT receptors was conducted by MDS Pharma (Bothell, WA) and performed in accordance with published assay protocols (28).
Intracellular cAMP Assay
The active samples in the 5-HT7 binding assay were further tested at a concentration of 40 μg/mL of plant extract for modulation of intracellular levels of cyclic adenosine monophosphate (cAMP) to determine if the active components in the extracts acted as agonists or antagonists of the 5-HT7 receptor. Extracts that induced at least 5 pmol/mL of cAMP at the tested concentration were considered to be agonists. The elevation of intracellular cAMP was assayed as previously described (29) with minor modifications. Briefly, human 5-HT7-transfected HEK293 cells were grown for four days in serum-free media and then plated in poly-D-lysine-coated 12-well plates (100 × 104 cells/well) and left for 24 h. The following day, the cells were washed twice with 2 mL of incubation buffer (150 nM NaCl, 5 mM KCl, 1 mM MgSO4, 2 mM CaCl2, 10 mM glucose, 10 mM HEPES, 500 μM isobutylmethylxanthine, 1 μM ascorbic acid, 10 μM pargyline, pH 7.4), and then incubated for 20 min at 37 °C. The compounds and extracts were added and the plates were incubated at 37 °C for 10 min. Forskolin (20 nM) and serotonin (10 nM) were used as positive controls. If stimulation could be blocked by co-incubation of serotonin with the 5-HT7 receptor antagonist SB-269970, the treatment was considered receptor mediated. The reactions were terminated by aspirating the buffer and adding 0.1 M HCl with 0.1% Triton-X detergent to lyse the cells. The cells were collected in microcentrifuge tubes and centrifuged for 5 min at 1000 rpm at room temperature. The cell supernatant was obtained and stored at −80 °C until the treated samples were analyzed. An aliquot of the cell supernatant (100 μL) was transferred to the Trevigen HT Direct cyclic AMP EIA competitive immunoassay kit for the quantitative determination of cyclic AMP in treated cells as per the manufacturer’s protocol (Trevigen Inc., Gaithersburg, MD). The plate was read immediately in a Powerwave 200 microplate spectrophotometer (BioTek Instruments, Winooski, VT) at 405 nm. The concentration of cAMP was determined by measuring the optical density (OD) minus the non-specific binding (NSB). The intensity of the yellow color was inversely proportional to the concentration of cAMP. The percentage of cAMP bound was determined using equation 2, based on the maximum available binding (Bmax). The amount of cAMP was calculated using a standard curve, and the data were expressed as pmol/mL induction and compared to the basal cellular cAMP level observed.
(2) |
Selective Serotonin Reuptake Inhibition (SSRI) Assay
The SSRI assay was performed as previously described (30) with minor modifications. Briefly, 200 μL of poly-D-lysine solution (20 μg/mL) was added to each well of 24-well plates. After shaking the plates for 5 min, the solution was aspirated, and the plates were dried for 1 h. hSERT-HEK293 cells (15 ×104 cells/well) were plated in the pre-coated 24-well culture plates. After 24 h, the plates were washed with KRH buffer (120 mM NaCl, 4.7 mM KCl, 2.2 mM CaCl2, 10 mM Hepes, 1.2 mM KH2PO4, 1.2 mM MgSO4, pH 7.4), incubated with the extracts at 37 °C in KRH buffer containing D-glucose (1.8 g/L), L-ascorbic acid (100 μM), and pargyline (100 μM) for 10 min, and then treated with [3H] 5-HT (20 nM). Fluoxetine (100 μM) was used as a positive control to validate the method. Cells were incubated for an additional 10 min. Uptake of [3H] 5-HT was terminated by washing the plates three times with KRH buffer (30). After adding 200 μL of 1% SDS to each well, the plates were incubated at room temperature for 30 min while gently shaking. The cell solution was transferred to scintillation vials with 4 mL Cytoscint. The tubes were counted using a Beckman LS 5801 (Schaumburg, IL) liquid scintillation counter. The percent inhibition was determined using Equation 1 as previously described. All C. racemosa extracts were subjected to SSRI assays, at 40 μg/mL, and were considered active if inhibition was ≥ 50%.
Statistics
The results of the serotonin receptor binding, intracellular cAMP, and selective serotonin reuptake inhibition assays were all determined as the average ± standard deviation of at least two independent experiments in triplicate determinations. Data were analyzed using the GraphPad Prism Version 4.01 to determine IC50 and EC50 values. Significant difference for cAMP column statistics compared to DMSO control was determined using the one way ANOVA and one sample t-test. IC50 values provided by MDS Pharma were determined by a non-linear, least squares regression analysis using MathIQ (ID Business Solutions Ltd., UK).
RESULTS AND DISCUSSION
Bioassay-guided fractionation (Figure 2)
Since the serotonin receptor 5-HT7 is involved in thermoregulation, bioassay-guided fractionation was performed using the 5-HT7 competitive binding assay and the MeOH crude extract. The choice of MeOH as an extraction solvent was based on practical phytochemical considerations, including viscosity, evaporation handling, and limitations in the supply of the clinical extract. In addition, while 75% EtOH and 100% MeOH extracts are of similar polarity and can be mutually substituted from a phytochemical perspective, neat MeOH extracts contain less of the polar interfering substances, such as sugars. The decision to prepare a 75% EtOH extract for clinical studies was based on both biological (serotonergic activity, factors affecting in vivo studies) and phytochemical analyses previously detailed (18).
Our initial studies, using the previously mentioned 75% EtOH clinical extract, demonstrated serotonergic activity with an IC50 value of 55 μg/mL. The MeOH crude extract had similar serotonergic activity (IC50 87 μg/mL). After separating the triterpenes and sugars from the crude MeOH extract by solvent partitioning, an enriched MeOH fraction (XAD-MeOH) was obtained by chromatography on the XAD column that exhibited more than ten-fold higher serotonergic binding activity compared to the clinical and crude MeOH black cohosh extracts (IC50 5 μg/mL) (Figure 2). Both the clinical extract and XAD-MeOH fraction were also evaluated for functional serotonergic activity. The 5-HT7 receptor is a G-protein coupled receptor that stimulates adenylate cyclase. Therefore, agonist occupancy of the receptor dissociates the Gα subunit of the G-protein and activates adenylate cyclase to catalyze the formation of cAMP (15). Forskolin, a compound known to stimulate all types of adenylate cyclase, and serotonin were used as positive controls. Co-incubation of serotonin with the 5-HT7 receptor antagonist SB-269970 could block this stimulation; however, the activation caused by forskolin could not be completely blocked. Therefore, cAMP production can be used to characterize serotonergic regulators with potential agonistic or antagonistic properties at functional 5-HT7 receptors. A dose-dependent response was observed for both the clinical extract and the XAD-MeOH fraction in the HEK293 5-HT7 receptor cell line (Figure 3), although the XAD-MeOH fraction was found to induce significantly more cAMP. The clinical extract and enriched XAD-MeOH fraction were also tested for SSRI activity. The SSRI assay was used to evaluate whether the tested plant extracts or compounds could target the SERT and inhibit serotonin reuptake employing hSERT-transfected-HEK293 cells. Fluoxetine was used as a positive control to validate the method. These black cohosh fractions showed no ability to inhibit the reuptake of serotonin (data not shown).
Pure compounds belonging to two major phytoconstituent classes of black cohosh (Figure 1) were also evaluated in the 5-HT7 competitive binding, cAMP induction, and SSRI assays to determine whether or not they are serotonergically active and if the serotonergic activity of the black cohosh extract is affected by their presence. Seventeen individual triterpenes, isolated as previously described (18, 21, 22), were tested in the 5-HT7 competitive binding assay at 50 μM (Table 1). The triterpenes tested displayed no significant activity for binding to the 5-HT7 receptor. For example, 23-epi-26-deoxyactein (also known as 27-deoxyactein) (31), 26-deoxyactein, and actein (R,S), which are among the most abundant triterpenes in C. racemosa (Figure 1) and commonly used markers for chemical standardization of black cohosh dietary supplements, exhibited IC50 values ≥ 100 μM (Figure 2). Cimicifugic acids A, B, E, and F (Figure 1), being characteristic and abundant constituents in Cimicifuga species, all had IC50 values greater than 500 μM, while fukinolic acid had an IC50 value of 100 μM (Figure 2). None of the 17 triterpenes (Table 1) induced cAMP activity (data not shown). The cimicifugic and fukinolic acids also failed to increase the production of cAMP and did not inhibit serotonin reuptake (data not shown). The triterpenes were not tested in the SSRI assay due to limited compound availability. Alkaloids, tannins, and flavonoids are among the other main constituents found in black cohosh (21, 22, 32–35). It was previously shown that antioxidant compounds (methyl caffeate, ferulic acid, caffeic acid, cimicifugic acids A and B, fukinolic acid, and cimiracemate A and B) and cimipronidines were contained in various extracts of black cohosh (32, 36) which may have chemopreventive properties (36); however, these compounds are unlikely to be responsible for the serotonergic activity of black cohosh.
Table 1.
5-HT7 competitive binding
| |
---|---|
Compound (50 μM) | % Inhibition ± SD |
23-epi-26-deoxyactein | NA |
23-O-acetylshengmanol-3-β-D-arabinoside | NA |
25-anhydrocimigenol-3-O- β-xyloside | 31 ± 8 |
26-deoxyactein | 38 ± 4 |
Cimiracemoside P | NA |
Cimiracemoside J | NA |
Cimiracemoside K | NA |
25-O-acetylcimigenol-3-O-β-xyloside | NA |
25-O-acetylcimigenol-3-O-α-arabinoside | NA |
25-anhydrocimigenol-3-O- β-arabinoside | NA |
Actein (R,S) | NA |
23-O-acetylshengmanol-3-β-D-xyloside | 35 ± 10 |
Cimiracemoside N | 18 ± 4 |
Cimiracemoside O | 19 ± 5 |
Cimiracemoside L | NA |
Cimicifugoside H-2 | NA |
Cimifugoside H-1 | NA |
Identification of Nω-methylserotonin
Since none of the previously isolated pure compounds had serotonergic activity, bioassay-guided fractionation was pursued with the XAD-MeOH fraction (Figure 2), mainly composed of phenolic compounds without triterpenes and sugars. All seven fractions resulting from subsequent FCPC separation were evaluated for 5-HT7 receptor binding. Fractions 2 and 3 contained cimipronidine alkaloids and had low receptor binding activity. Fraction 4 contained the polyphenolic acids which exhibited weak 5-HT7 receptor binding activity (Figure 2, Table 1, IC50 ~ 100 μg/mL) and no functional activity (data not shown). Fraction 7 showed enhanced 5-HT7 binding activity (Figure 2, IC50 < 2.5 μg/mL). Positive ion electrospray LC-MS analysis of this active fraction revealed the presence of an abundant protonated molecule of m/z 191 eluting at 22 min (Figures 4A and 4B). Accurate mass measurements determined an elemental composition of C11H14N2O (− 5.4 ppm). A database search (CrossFire Commander) revealed only 13 compounds with this formula, one of which was Nω-methylserotonin. The positive ion product and tandem mass spectrum of this compound and the corresponding compound in fraction 7 (Figures 4C and 4D) exhibited a prominent loss of methylamine (31 mass units) from the precursor ion, indicating the presence of a secondary amine in the structure. Of all the compounds in the database, only Nω-methylserotonin was consistent with these analytical data. Confirmation was obtained by establishing that the retention time and fragmentation pattern of this compound in fraction 7 were identical to those of the authentic Nω-methylserotonin (Figure 4). The XAD-MeOH fraction 7 and Nω-methylserotonin were also analyzed using an isocratic HPLC method and LC-MS based on a Zorbax Eclipse C18 column, as well as the gradient HPLC method with LC-MS-MS based on a YMC ODS column (data not shown) leading to the unambiguous identification of Nω-methylserotonin as a new C. racemosa phytoconstituent.
To our knowledge, this is the first report that Nω-methylserotonin has been identified in C. racemosa roots/rhizomes. Extracts from another species, Ranunculus sceleratus L., (cursed buttercup, marsh-crowfoot), belonging to the same family as black cohosh (Ranunculaceae), also contains serotonin and other tryptamine analogues (49, 50). Due to the very low levels of Nω-methylserotonin in the black cohosh extract, isolation of the target compound was not attempted. A fully characterized sample of Nω-methylserotonin, analyzed by MS, 1D and 2D NMR, and qHNMR for purity evaluation, was used in the bioassays. In the black cohosh extract, Nω-methylserotonin is present at very low levels (~30 ppm) that do not allow detection by NMR due to the relatively low sensitivity and dynamic range of the method. Mass spectrometry was used in this instance as a well-established method for the detection and identification of known compounds present in complex mixtures at low levels. In this study we employed an approach that is widely accepted not only in pharmaceutical industry, but also in the chemical industry and forensics and regulatory area. This approach includes determination of molecular composition by accurate mass measurement combined with the comparison of retention times and fragmentation patterns of the test compound with the authentic standard. For the purpose of comparison of retention characteristics, we employed two different chromatographic modes (normal vs. reversed phase and two different reversed phase columns and elution systems), which further strengthens the identification of Nω-methylserotonin as the active principle.
Serotonergic and SSRI activity of Nω-methylserotonin
Nω-methylserotonin was evaluated in the 5-HT7 receptor binding assay, and it revealed a potent binding affinity, IC50 23 pM (Figure 5). To test selectivity, Nω-methylserotonin was also screened against all the known serotonin receptors (MDS Pharma). Nω-methylserotonin showed a higher selectivity for the 5-HT1, 5-HT6, and 5-HT7 subfamilies (Table 2), particularly the 5-HT1A and 5-HT7 receptors, and the least selectivity for the 5-HT3 and 5-HT4 receptors which do not appear to be associated with thermoregulation. In general, the addition of the methyl group to the omega nitrogen appears to increase the selectivity for most of the serotonin receptors when compared to serotonin itself. In the cAMP induction assay, Nω-methylserotonin demonstrated a strong ability to increase intracellular cAMP levels (Figure 6). cAMP formation could be reversed by co-incubation with the 5-HT7 antagonist, SB 269970 (Figure 6A), which indicated the process was receptor mediated. The EC50 was determined to be 22 nM, and the maximum cAMP induction (43 pmol/mL) was observed at 100 nM, the highest tested concentration. Additionally, Nω-methylserotonin showed strong SSRI activity, IC50 490 nM, in this assay (Figure 7) compared to the SSRI antidepressant, fluoxetine hydrochloride (Prozac, IC50 150 nM). While the fluoxetine and Nω-methylserotonin SSRI activities appear to be of a comparable level, the black cohosh extracts failed to exhibit any SSRI activity.
Table 2.
IC50 [nM]
| |||
---|---|---|---|
Receptor | Nω-methylserotonina | Serotoninc | Positive controlsc |
5-HT1A | 2 | 1.9 | Metergoline: 4 |
5-HT1B | 15 | 19 | Serotonin: 19 |
5-HT2A | -- | 410 | Ketanserin: 0.65 |
5-HT2B | 97 | 55 | Ketanserin: 290 |
5-HT2C | 101 | 67 | Mesulergine: 0.7 |
5-HT3 | 1670 | 700 | MDL-72222: 11 |
5-HT4 | 6700 | 300 | R,S-235970-190: 90 |
5-HT5A | 289 | 100 | Methiothepin: 2.9 |
5-HT6 | 55 | 340 | Methiothepin: 2.8 |
5-HT7 | 0.02b | 0.86 | Methiothepin: 0.4 |
The results represent primary screening data (three points with duplicate determinations) tested at MDS Pharma, Bothell, WA, except for the binding data with the 5-HT7 receptor.
These represent results performed in triplicate in three independent experiments.
The results represent MDS Pharma results in accordance with published assay protocols.
Quantitative analysis of the clinical extract also indicated the presence of Nω-methylserotonin and the amount was determined to be 0.0031% by weight of the excipient-free extract. Thus Nω-methylserotonin was independently identified in both a 75% EtOH and a 100% MeOH extract suggesting that methylation of serotonin was not an artifact of the extraction process. Further analysis found 100 μg/mL of the clinical extract would contain approximately 16 nM of Nω-methylserotonin. At this concentration, cAMP induction is expected to be 17 pmol/mL; however, less than half of that amount was found to be induced in the clinical extract (Figure 3) probably due to the fact that other phytoconstituents in the extract may modulate the activity. In contrast, the XAD-MeOH fraction induced twice the amount of cAMP expected at 100 μg/mL (Figure 3).
Black cohosh has become a very popular alternative therapy for alleviating menopausal symptoms, particularly hot flashes (1, 4, 9). However, many questions about its efficacy as well as mechanism(s) of action or active constituent(s) remain unclear. In vitro and in vivo experiments designed to determine the mechanism(s) of action found that black cohosh does not seem to work through classical estrogenic pathways (7, 12, 13). For example, an in vivo animal study using ovariectomized rats found that black cohosh failed to maintain consistent body weight, did not increase uterine weight, and did not lead to cell differentiation of the vaginal tract, all of which are markers of estrogenic activity (14). Since black cohosh exhibited no estrogenic or antiestrogenic activity in vivo, an alternative serotonergic mechanism linked to thermoregulation (14) was examined. In support of this hypothesis, several Phase II and III clinical trials have found that SSRIs were able to reduce hot flashes in breast cancer survivors treated with tamoxifen, raloxifene, aromatase inhibitors, soy products, and/or vitamin E (2, 37). Additional studies have also shown that SSRIs have an affinity to the 5-HT1A receptor, resulting in its downregulation (38, 39). As a result, serotonergic pathways could play a role in black cohosh mediated thermoregulation, although the precise mechanism of action remains unclear. In addition to serotonergic pathways, it is possible that dopaminergic (41–43) and opiate systems (44, 45) play a role in mitigation of hot flashes. Similarities exist between hot flashes experienced as a result of going through menopause or opiate withdrawal (46), which is why opiate-dependent animal models have been used to evaluate hot flashes in vivo (47). It has been reported that black cohosh extracts behave as competitive ligands and partial agonists for opiate receptors (44, 48) similar to serotonin receptors (14). A competitive binding assay using a CHO cell line stably transfected with human μ opioid receptors and the same clinical extract used in this study gave an IC50 of 170 μg/mL (44) compared to 55 μg/mL for the 5-HT7 receptor. Although direct comparisons between the opiate experiments and the serotonergic studies are difficult, these data suggest at least two relevant activities are found in black cohosh and both may play a role in thermoregulation of hot flashes.
In summary, the in vitro serotonergic activity of black cohosh extracts appears to be mediated by Nω-methylserotonin. Nω-methylserotonin was active in both the 5-HT7 receptor and the SERT pathways, both of which have been shown to affect thermoregulation and can increase serotonin activity. Nω-methylserotonin displayed 5-HT7 receptor binding activity at pM levels, cAMP induction activity at nM levels, and SSRI activity in the μM range. Therapeutically, a 120 mg daily dose of the black cohosh clinical extract would contain approximately 3.7 μg of Nω-methylserotonin. The current in vitro studies suggest that Nω-methylserotonin could be responsible for in vivo serotonergic activity, although further studies are in progress to evaluate oral bioavailability. The data have also shown that while triterpenes and Cimicifuga polyphenols are suitable markers for chemical standardization, they do not correlate with the serotonergic activity. It is possible that effective black cohosh extracts, beneficial for relieving menopausal symptoms, should be biologically standardized to compounds that show serotonergic activity, such as Nω-methylserotonin. However, further studies are necessary and ongoing in our laboratory, and will be reported in due course.
Acknowledgments
This work was supported by grant P50 AT00155 provided jointly by the Office of Dietary Supplements (ODS), the National Center for Complementary and Alternative Medicine (NCCAM), the Office for Research on Women’s Health (ORWH), and the National Institute of General Medicine NIGMS) of the National Institutes of Health (NIH). S.L.P. is grateful for a Ruth L. Kirschstein NCCAM predoctoral fellowship F31 AT 003995. The contents of this paper are solely the responsibility of the authors and do not necessarily represent the official views of NIH.
ABBREVIATIONS USED
- Bmax
maximum available binding
- BSA
bovine serum albumin
- cAMP
cyclic adenosine monophosphate
- CHO
Chinese hamster ovary
- CoA
certificate of analysis
- DMEM
Dulbecco’s modified Eagle’s medium
- FBS
fetal bovine serum
- FCPC
fast centrifugal partition chromatography
- HEK
human embryonic kidney
- HRT
hormone replacement therapy
- hSERT
human serotonin transporter
- KRH
Krebs/Ringer/Hepes
- LSD
lysergic acid diethylamide
- MeOH
methanol
- N-methylserotonin
3-(2-Methylaminoethyl)indol-5-ol
- NSB
non-specific binding
- OD
optical density
- PEI
polyethylenimine
- qHNMR
quantitative 1H NMR
- SB-269970
(R)-3-[2-[2-(4-Methylpiperidin-1-yl)ethyl]pyrrolidine-1-sulfonyl]phenol hydrochloride
- SERT
serotonin transporter
- SSRI
selective serotonin reuptake inhibitor
- TIC
total ion chromatogram
- 5-HT
5-hydroxytryptamine, serotonin, 3-(2-Aminoethyl)indol-5-ol
LITERATURE CITED
- 1.Mahady GB. Is black cohosh estrogenic? Nutr Rev. 2003;61:183–186. doi: 10.1301/nr.2003.may.183-186. [DOI] [PubMed] [Google Scholar]
- 2.Carpenter JS. State of the science: hot flashes and cancer. Part 2: management and future directions. Oncol Nurs Forum. 2005;32:969–978. doi: 10.1188/05.ONF.969-978. [DOI] [PubMed] [Google Scholar]
- 3.Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321–333. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
- 4.Borrelli F, Ernst E. Black cohosh (Cimicifuga racemosa) for menopausal symptoms: a systematic review of its efficacy. Pharmacol Res. 2008;58:8–14. doi: 10.1016/j.phrs.2008.05.008. [DOI] [PubMed] [Google Scholar]
- 5.Tesch BJ. Herbs commonly used by women: An evidence-based review. Am J Obstet Gynecol. 2003;188:S44–S55. doi: 10.1067/mob.2003.402. [DOI] [PubMed] [Google Scholar]
- 6.Blumenthal M. Complementary & Alternative Medicine: The use of black cohosh to treat symptoms of menopause. Sexuality, Reproduction, & Menopause. 2004;2:27–34. [Google Scholar]
- 7.Borrelli F, Izzo AA, Ernst E. Pharmacological effects of Cimicifuga racemosa. Life Sci. 2003;73:1215–1229. doi: 10.1016/s0024-3205(03)00378-3. [DOI] [PubMed] [Google Scholar]
- 8.McKenna DJ, Jones K, Humphrey S, Hughes K. Black cohosh: efficacy, safety, and use in clinical and preclinical applications. Altern Ther Health Med. 2001;7:93–100. [PubMed] [Google Scholar]
- 9.Osmers R, Friede M, Liske E, Schnitker J, Freudenstein J, Henneicke-von Zepelin HH. Efficacy and safety of isopropanolic black cohosh extract for climacteric symptoms. Obstet Gynecol. 2005;105:1074–1083. doi: 10.1097/01.AOG.0000158865.98070.89. [DOI] [PubMed] [Google Scholar]
- 10.Bolle P, Mastrangelo S, Perrone F, Evandri MG. Estrogen-like effect of a Cimicifuga racemosa extract sub-fraction as assessed by in vivo, ex vivo and in vitro assays. J Steroid Biochem Mol Biol. 2007;107:262–269. doi: 10.1016/j.jsbmb.2007.03.044. [DOI] [PubMed] [Google Scholar]
- 11.Kretzschmar G, Nisslein T, Zierau O, Vollmer G. No estrogen-like effects of an isopropanolic extract of rhizoma Cimicifugae racemosae on uterus and vena cava of rats after 17 day treatment. J Steroid Biochem Mol Biol. 2005;97:271–277. doi: 10.1016/j.jsbmb.2005.05.008. [DOI] [PubMed] [Google Scholar]
- 12.Liu J, Burdette JE, Xu H, Gu C, van Breemen RB, Bhat KP, Booth N, Constantinou AI, Pezzuto JM, Fong HH, Farnsworth NR, Bolton JL. Evaluation of estrogenic activity of plant extracts for the potential treatment of menopausal symptoms. J Agric Food Chem. 2001;49:2472–2479. doi: 10.1021/jf0014157. [DOI] [PubMed] [Google Scholar]
- 13.Stromeier S, Petereit F, Nahrstedt A. Phenolic esters from the rhizomes of Cimicifuga racemosa do not cause proliferation effects in MCF-7 cells. Planta Med. 2005;71:495–500. doi: 10.1055/s-2005-864148. [DOI] [PubMed] [Google Scholar]
- 14.Burdette JE, Liu J, Chen SN, Fabricant DS, Piersen CE, Barker EL, Pezzuto JM, Mesecar A, van Breemen RB, Farnsworth NR, Bolton JL. Black cohosh acts as a mixed competitive ligand and partial agonist of the serotonin receptor. J Agric Food Chem. 2003;51:5661–5670. doi: 10.1021/jf034264r. [DOI] [PubMed] [Google Scholar]
- 15.Hedlund PB, Sutcliffe JG. Functional, molecular and pharmacological advances in 5-HT7 receptor research. Trends Pharmacol Sci. 2004;25:481–486. doi: 10.1016/j.tips.2004.07.002. [DOI] [PubMed] [Google Scholar]
- 16.Farnsworth NR, Krause EC, Bolton JL, Pauli GF, van Breemen RB, Graham JG. The University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements Research for Women’s Health: from plant to clinical use. Am J Clin Nutr. 2008;87:504S–508S. doi: 10.1093/ajcn/87.2.504S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pauli GF, Jaki BU, Lankin DC. Quantitative 1H NMR: development and potential of a method for natural products analysis. J Nat Prod. 2005;68:133–149. doi: 10.1021/np0497301. [DOI] [PubMed] [Google Scholar]
- 18.Fabricant DS. PhD dissertation. University of Illinois; Chicago: 2006. Pharmacognostic investigation of black cohosh (Cimicifuga racemosa (L.) Nutt.) [Google Scholar]
- 19.Xu H, Fabricant DS, Piersen CE, Bolton JL, Pezzuto JM, Fong H, Totura S, Farnsworth NR, Constantinou AI. A preliminary RAPD-PCR analysis of Cimicifuga species and other botanicals used for women’s health. Phytomedicine. 2002;9:757–762. doi: 10.1078/094471102321621403. [DOI] [PubMed] [Google Scholar]
- 20.Li W, Chen S, Fabricant D, Angerhofer CK, Fong HHS, Farnsworth NR, Fitzloff JF. High-performance liquid chromatographic analysis of black cohosh (Cimicifuga racemosa) constituents with in-line evaporative light scattering and photodiode array detection. Anal Chim Acta. 2002;471:61–75. [Google Scholar]
- 21.Chen SN, Fabricant DS, Lu ZZ, Fong HH, Farnsworth NR. Cimiracemosides I-P, new 9,19-cyclolanostane triterpene glycosides from Cimicifuga racemosa. J Nat Prod. 2002;65:1391–1397. doi: 10.1021/np0200818. [DOI] [PubMed] [Google Scholar]
- 22.Chen SN, Fabricant DS, Lu ZZ, Zhang H, Fong HH, Farnsworth NR. Cimiracemates A-D, phenylpropanoid esters from the rhizomes of Cimicifuga racemosa. Phytochemistry. 2002;61:409–413. doi: 10.1016/s0031-9422(02)00209-1. [DOI] [PubMed] [Google Scholar]
- 23.Friesen JB, Pauli GF. Rational development of solvent system families in counter-current chromatography. J Chromatogr A. 2007;1151:51–59. doi: 10.1016/j.chroma.2007.01.126. [DOI] [PubMed] [Google Scholar]
- 24.Berthod A, Friesen JB, Inui T, Pauli GF. Elution-extrusion countercurrent chromatography: theory and concepts in metabolic analysis. Anal Chem. 2007;79:3371–3382. doi: 10.1021/ac062397g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Friesen JB, Pauli GF. Reciprocal symmetry plots as a representation of countercurrent chromatograms. Anal Chem. 2007;79:2320–2324. doi: 10.1021/ac062007q. [DOI] [PubMed] [Google Scholar]
- 26.Gödecke T, ND, Lankin DC, Chen S, Powell SL, Dietz B, Bolton JL, Van Breemen RB, Farnsworth NR, Pauli GF. Phytochemistry of cimicifugic acids and associated bases in Cimicifuga racemosa root extracts. Phytochemical Analysis. 2008 doi: 10.1002/pca.1106. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Roth BL, Craigo SC, Choudhary MS, Uluer A, Monsma FJ, Jr, Shen Y, Meltzer HY, Sibley DR. Binding of typical atypical antipsychotic agents to 5-hydroxytryptamine-6, 5-hydroxytryptamine-7, receptors. J Pharmacol Exp Ther. 1994;268:1403–1410. [PubMed] [Google Scholar]
- 28.MDSPharmaServices. 2008 Oct 24; http://discovery.mdsps.com.
- 29.Albert PR, Zhou QY, Van Tol HH, Bunzow JR, Civelli O. Cloning, functional expression, and mRNA tissue distribution of the rat 5-hydroxytryptamine1A receptor gene. J Biol Chem. 1990;265:5825–5832. [PubMed] [Google Scholar]
- 30.Rodriguez GJ, Roman DL, White KJ, Nichols DE, Barker EL. Distinct recognition of substrates by the human and Drosophila serotonin transporters. J Pharmacol Exp Ther. 2003;306:338–346. doi: 10.1124/jpet.103.048751. [DOI] [PubMed] [Google Scholar]
- 31.Chen SN, Li W, Fabricant DS, Santarsiero BD, Mesecar A, Fitzloff JF, Fong HH, Farnsworth NR. Isolation, structure elucidation, and absolute configuration of 26-deoxyactein from Cimicifuga racemosa and clarification of nomenclature associated with 27-deoxyactein. J Nat Prod. 2002;65:601–605. doi: 10.1021/np010494t. [DOI] [PubMed] [Google Scholar]
- 32.Fabricant DS, Nikolic D, Lankin DC, Chen SN, Jaki BU, Krunic A, van Breemen RB, Fong HH, Farnsworth NR, Pauli GF. Cimipronidine, a cyclic guanidine alkaloid from Cimicifuga racemosa. J Nat Prod. 2005;68:1266–1270. doi: 10.1021/np050066d. [DOI] [PubMed] [Google Scholar]
- 33.He K, Pauli GF, Zheng B, Wang H, Bai N, Peng T, Roller M, Zheng Q. Cimicifuga species identification by high performance liquid chromatography-photodiode array/mass spectrometric/evaporative light scattering detection for quality control of black cohosh products. J Chromatogr A. 2006;1112:241–254. doi: 10.1016/j.chroma.2006.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jiang B, Kronenberg F, Nuntanakorn P, Qiu MH, Kennelly EJ. Evaluation of the botanical authenticity and phytochemical profile of black cohosh products by high-performance liquid chromatography with selected ion monitoring liquid chromatography-mass spectrometry. J Agric Food Chem. 2006;54:3242–3253. doi: 10.1021/jf0606149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nuntanakorn P, Jiang B, Einbond LS, Yang H, Kronenberg F, Weinstein IB, Kennelly EJ. Polyphenolic constituents of Actaea racemosa. J Nat Prod. 2006;69:314–318. doi: 10.1021/np0501031. [DOI] [PubMed] [Google Scholar]
- 36.Burdette JE, Chen SN, Lu ZZ, Xu H, White BE, Fabricant DS, Liu J, Fong HH, Farnsworth NR, Constantinou AI, van Breemen RB, Pezzuto JM, Bolton JL. Black cohosh (Cimicifuga racemosa L.) protects against menadione-induced DNA damage through scavenging of reactive oxygen species: bioassay-directed isolation and characterization of active principles. J Agric Food Chem. 2002;50:7022–7028. doi: 10.1021/jf020725h. [DOI] [PubMed] [Google Scholar]
- 37.Loprinzi CL, Flynn PJ, Carpenter LA, Atherton P, Barton DL, Shanafelt TD, Rummans TA, Sloan JA, Adjei AA, Mincey BA, Fitch TR, Collins M. Pilot evaluation of citalopram for the treatment of hot flashes in women with inadequate benefit from venlafaxine. J Palliat Med. 2005;8:924–930. doi: 10.1089/jpm.2005.8.924. [DOI] [PubMed] [Google Scholar]
- 38.Czachura JF, Rasmussen K. Effects of acute and chronic administration of fluoxetine on the activity of serotonergic neurons in the dorsal raphe nucleus of the rat. Naunyn-Schmiedeberg’s archives of pharmacology. 2000;362:266–275. doi: 10.1007/s002100000290. [DOI] [PubMed] [Google Scholar]
- 39.Rossi DV, Burke TF, McCasland M, Hensler JG. Serotonin-1A receptor function in the dorsal raphe nucleus following chronic administration of the selective serotonin reuptake inhibitor sertraline. J Neurochem. 2008;105:1091–1099. doi: 10.1111/j.1471-4159.2007.05201.x. [DOI] [PubMed] [Google Scholar]
- 40.Sugimoto Y, Yamada J, Horisaka K Department of Pharmacology, K. W. s. C. o. P. J. Activation of peripheral serotonin2 receptors induces hypothermia in mice. Life Sci. 1991;48(5):419–423. doi: 10.1016/0024-3205(91)90497-y. [DOI] [PubMed] [Google Scholar]
- 41.Yamada J, Sugimoto Y, Wakita H, Horisaka K. The involvement of serotonergic and dopaminergic systems in hypothermia induced in mice by intracerebroventricular injection of serotonin. Jpn J Pharmacol. 1988;48:145–148. doi: 10.1254/jjp.48.145. [DOI] [PubMed] [Google Scholar]
- 42.Cox B, Ary M, Lomax P. Dopaminergic mechanisms in withdrawal hypothermia in morphine dependent rats. Life Sci. 1975;17:41–42. doi: 10.1016/0024-3205(75)90231-3. [DOI] [PubMed] [Google Scholar]
- 43.Cox B, Tha SJ. The role of dopamine and noradrenaline in temperature control of normal and reserpine-pretreated mice. J Pharm Pharmacol. 1975;27:242–247. doi: 10.1111/j.2042-7158.1975.tb10693.x. [DOI] [PubMed] [Google Scholar]
- 44.Rhyu MR, Lu J, Webster DE, Fabricant DS, Farnsworth NR, Wang ZJ. Black cohosh (Actaea racemosa, Cimicifuga racemosa) behaves as a mixed competitive ligand and partial agonist at the human mu opiate receptor. J Agric Food Chem. 2006;54:9852–9857. doi: 10.1021/jf062808u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Stomati M, Rubino S, Spinetti A, Parrini D, Luisi S, Casarosa E, Petraglia F, Genazzani AR. Endocrine, neuroendocrine and behavioral effects of oral dehydroepiandrosterone sulfate supplementation in postmenopausal women. Gynecol Endocrinol. 1999;13:15–25. doi: 10.1080/09513599909167527. [DOI] [PubMed] [Google Scholar]
- 46.Simpkins JW, Katovich MJ, Song IC. Similarities between morphine withdrawal in the rat and the menopausal hot flush. Life Sci. 1983;32:1957–1966. doi: 10.1016/0024-3205(83)90047-4. [DOI] [PubMed] [Google Scholar]
- 47.Katovich MJ, Simpkins JW, O’Meara J. Effects of acute central LH-RH administration of the skin temperature response in morphine dependent rats. Brain Res. 1989;494:85–94. doi: 10.1016/0006-8993(89)90146-7. [DOI] [PubMed] [Google Scholar]
- 48.Reame NE, Lukacs JL, Padmanabhan V, Eyvazzadeh AD, Smith YR, Zubieta JK. Black cohosh has central opioid activity in postmenopausal women: evidence from naloxone blockade and positron emission tomography neuroimaging. Menopause. 2008;15:832–840. doi: 10.1097/gme.0b013e318169332a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bhargava KP, Kishor K, Pant MC, Saxena PR. Identification of tryptamine derivatives in Ranunculus sceleratus L. Br J Pharmacol Chemother. 1965;25:743–750. doi: 10.1111/j.1476-5381.1965.tb01796.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Smith TA. Tryptamine and related compounds in plants. Phytochemistry. 1977;16:171–175. [Google Scholar]