Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Potent Natural Soluble Epoxide Hydrolase Inhibitors from Pentadiplandra brazzeana Baillon: Synthesis, Quantification, and Measurement of Biological Activities In Vitro and In Vivo

  • Seiya Kitamura,

    Affiliation Department of Entomology and Nematology, and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America

  • Christophe Morisseau,

    Affiliation Department of Entomology and Nematology, and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America

  • Bora Inceoglu,

    Affiliation Department of Entomology and Nematology, and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America

  • Shizuo G. Kamita,

    Affiliation Department of Entomology and Nematology, and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America

  • Gina R. De Nicola,

    Affiliation Consiglio per la Ricerca e la sperimentazione in Agricoltura, Centro di Ricerca per le Colture Industriali (CRA-CIN), Bologna, Italy

  • Maximilienne Nyegue,

    Affiliation Départment of Biochemistry and Départment of Microbiology, University of Yaoundé I, Yaoundé, Cameroon

  • Bruce D. Hammock

    bdhammock@ucdavis.edu

    Affiliation Department of Entomology and Nematology, and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America

Abstract

We describe here three urea-based soluble epoxide hydrolase (sEH) inhibitors from the root of the plant Pentadiplandra brazzeana. The concentration of these ureas in the root was quantified by LC-MS/MS, showing that 1, 3-bis (4-methoxybenzyl) urea (MMU) is the most abundant (42.3 μg/g dry root weight). All of the ureas were chemically synthesized, and their inhibitory activity toward recombinant human and recombinant rat sEH was measured. The most potent compound, MMU, showed an IC50 of 92 nM via fluorescent assay and a Ki of 54 nM via radioactivity-based assay on human sEH. MMU effectively reduced inflammatory pain in a rat nociceptive pain assay. These compounds are among the most potent sEH inhibitors derived from natural sources. Moreover, inhibition of sEH by these compounds may mechanistically explain some of the therapeutic effects of P. brazzeana.

Introduction

Soluble epoxide hydrolase (sEH, EC 3.3.2.10) is the major enzyme responsible for the hydrolysis of epoxy fatty acids (EpFAs) to their corresponding vicinal diols in humans and other mammals [1]. These EpFAs include the epoxides of linoleic, arachidonic, eicosapentaenoic, and docosahexaenoic acid that are produced primarily by cytochrome P450s. These natural molecules are pleiotropic endogenous mediators with key functions in inflammation [1], pain [2], and blood pressure regulation [3]. Increasing the levels of endogenous EpFAs by inhibiting sEH has been shown to block and resolve inflammation [4], reduce pain [2,5,6], and lower blood pressure [7] in several in vivo models. Recently, sEH inhibitors were shown to be effective against neuropathic diabetic pain in rodent models [8], and against equine laminitis which is a complex and often fatal disease involving inflammation, hypertension, and severe neuropathic pain [9]. Consequently, sEH has emerged as a potential pharmaceutical target. One sEH inhibitor, AR9281, has undergone a phase II clinical trial for the treatment of hypertension and impaired glucose tolerance [10]. Two other clinical trials are now underway with a different sEH inhibitor that targets chronic obstructive pulmonary disease by Glaxo Smith Kline [11,12]. Several recent studies show that at least some of the beneficial effects associated with dietary supplementation of omega-3 fatty acids (fish oils) are due to the corresponding epoxide metabolites of omega-3 fatty acids [2,13]. Thus sEH inhibitors appear to enhance the positive effects of diet supplementation with fish oils.

A few sEH inhibitors from natural products have been identified. Buscato et al. (2013) screened a library of compounds isolated from entomopathogenic bacteria and reported isopropylstilbene as an sEH inhibitor with an IC50 of 10 μM [14]. Additionally, Lee et al. (2013) reported honokiol and β-amyrin acetate as sEH inhibitors with IC50 values of 3.4 μM and 0.57 μM, respectively [15]. However, the potency of these natural compounds is significantly lower than that of synthetic sEH inhibitors which can possess IC50 values in the low nM to pM range [16,17]. Although purified natural products have been shown to function as sEH inhibitors in vitro, the in vivo efficacy of these natural products as sEH inhibitors is yet to be reported.

In our efforts to search for potent sEH inhibitors from natural products and elucidate their possible therapeutic and nutraceutical applications, we focused on the central pharmacophore of known sEH inhibitors with high potency. The 1,3-disubstituted urea is known as a pharmacophore of potent sEH inhibitors [18]. The urea pharmacophore mimics both the epoxide substrate and the transition state of epoxide hydrolysis, leading to competitive inhibition of sEH. Lipophilic substitutions on the urea are favored for improved potency [19].

Tsopmo et al. reported the isolation and identification of disubstituted urea compounds in the root of the plant Pentadiplandra brazzeana [20]. Pentadiplandra brazzeana (commonly known as Oubli in French) is the sole species in the plant genus Pentadiplandra. This plant grows either as a shrub or as a liana, and is native to Cameroon and other places in West and Central Africa [2123]. The root of this plant is used as a folk remedy against hemorrhoids [24], toothache [25], and as an analgesic for the treatment of chest, abdominal, and intercostal pain, as well as rheumatic disorders [23]. Moreover, the essential oil obtained from the root has anti-inflammatory effects [26]. Tsopmo et al. have isolated and identified three 1,3-dibenzyl ureas in the root of P. brazzeana [20]. However, the biological activity of these ureas was not evaluated.

On the basis of structural analogy, we hypothesized that urea compounds in P. brazzeana are inhibitors of human sEH. To test this hypothesis we measured the inhibitory potency of the crude root extract as well as the individual ureas found in P. brazzeana against recombinant human and recombinant rat sEH. The amount of these inhibitors was quantified using LC-MS/MS, and the analgesic efficacy of the most potent and abundant compound (MMU) was measured in a nociceptive assay using a rat inflammatory pain model.

Materials and Methods

General

All reagents and solvents were purchased from commercial suppliers and were used without further purification. Honokiol (purity>98%) was purchased from R&D systems (Minneapolis, MN) and stored at 4°C. All of the synthetic reactions were performed in an inert atmosphere of dry nitrogen or argon. Melting points were determined using an OptiMelt melting point apparatus and are uncorrected. 1H and 13C-NMR spectra were collected using a Varian 600 MHz spectrometer with chemical shifts reported relative to residual deuterated solvent peaks or tetramethylsilane internal standard. Accurate masses were measured using a Micromass LCT ESI-TOF-MS. FT-IR spectra were recorded on a Thermo Scientific NICOLET IR100 FT-IR Spectrometer.

Ethics Statement

The plant samples were harvested under the authority of National Herbarium of Cameroon by Mrs. Ada, a Cameroonian botanist at the National Herbarium of Cameroon. The National Herbarium of Cameroon is the authority in charge of the promotion of research on plants. Cameroonian researchers do not need permission to collect plant samples in Cameroon.

For the nociceptive assays, all of the studies were conducted in line with U.S. federal government regulations and were approved by the Institutional Animal Care and Use Committee at the University of California, Davis.

Plant material and sample preparation

The plant root samples were harvested on February 5th 2010 at Elounden (Yaoundé, Cameroon) by Mrs. Ada, a botanist at the National Herbarium of Cameroon in Yaoundé. A voucher specimen is kept at the National Herbarium of Cameroon in Yaoundé (Identication No. 6538NM/01). Root material was freeze-dried, reduced to a fine powder and kept at -20°C until used for the analysis.

DNA extraction and sequencing of ribosomal DNA and maturase K DNA partial sequences

Total DNA in the root powder was extracted using a Qiagen DNeasy Plant Mini Kit (Qiagen, Valencia, CA, USA) following the manufacturer’s protocol. The partial sequences of 18S ribosomal DNA and maturase K DNA were amplified by PCR. The sequences of the PCR primers are as follows: 18S ribosomal DNA, forward primer-1 (5′-GCCGCGGTAATTCCAGCTCCAATAGCGTATATTT-3′) and reverse primer-1 (5′-GAGTCCTAAAAGCAACATCCGCTGATCCCTG-3′); and forward primer-2 (5′-GCAGTTAAAAAGCTCGTAGTTGGACCTTGGGATG-3′) and reverse primer-2 (5′-TGAGACTAGGACGGTATCTGATCGTCTTCGAG-3′). Maturase K DNA, forward primer-1 (5′-GGAGGAATTTCAAGTATATTTAGAGTTGGATAGAGTTCGGC-3′) and reverse primer-1 (5′-CGCAAGAAATGCAAAGAAGAGGCATCTTTTACCCTG-3′); and forward primer-2 (5′-GCAGTTAAAAAGCTCGTAGTTGGACCTTGGGATG-3′) and reverse primer-2 (5′- TGAGACTAGGACGGTATCTGATCGTCTTCGAG-3′). PCR amplification was performed with these primers using GoTaq Green Master Mix (Promega, Madison, WI, USA) as follows: 95°C, 2 min; 35 cycles of 95°C, 30 sec; 62°C, 30 sec; and 72°C, 45 sec; followed by 72°C, 5 min. This PCR generated a 0.5 kbp-long amplicon that was column-purified using a QIAquick Gel Extraction Kit (Qiagen, Valencia, CA, USA) following the manufacturer’s protocol. The sequences of PCR products were determined by the UC Davis College of Biological Sciences Sequencing Facility.

Extraction

The dried root powder (100 mg) was extracted with dichloromethane (DCM)-methanol (MeOH) (1:1) (3 x 2 ml) at room temperature (24 h per extraction). Concentration of the combined percolates in vacuo yielded a yellowish crude extract (approximately 8 mg).

Synthesis of ureas

1,3-Dibenzylurea (BBU) was previously synthesized [27]. Fig. 1 shows the synthetic scheme for the other 2 ureas described in this study.

thumbnail
Fig 1. Synthetic schemes of urea compounds found in P. brazzeana.

THF = tetrahydrofuran, DCM = dichloromethane, DIEA = diisopropylethylamine.

https://doi.org/10.1371/journal.pone.0117438.g001

1-Benzyl-3-(4-methoxybenzyl) urea (BMU)

To a solution of benzylisocyanate (158 μl, 1.28 mmol) in THF (2 ml) was added 167 μl (1.28 mmol) of (4-methoxyphenyl) methanamine. After stirring for 10 min at room temperature, hexane was added and the resulting white crystals were collected (260 mg, 0.963 mmol, 75%). mp 139.6–140.2°C; 1H-NMR (DMSO-d6, 600 MHz); δ (ppm) 7.31 (2H, dd, J1 = 7.5 Hz, J2 = 5.0 Hz) 7.25–7.20 (m, 3H), 7.17 (2H, d, J = 8.4 Hz), 6.87 (2H, d, J = 8.4 Hz), 6.38 (1H, t, J = 6.0 Hz), 6.33 (1H, t, J = 6.0 Hz), 4.22 (2H, d, J = 6.0 Hz), 4.15 (2H, d, J = 6.0 Hz), 3.73 (3H, s), 13C NMR (151 MHz, DMSO-d6): δ 158.1, 158.0, 140.9, 132.8, 128.3, 128.2, 127.0, 126.5, 113.6, 55.0, 42.9, 42.4; IR (cm-1) 3349, 3317, 3032, 2923, 1625, 1577, 1511, 1242, 1031, ESI-MS [M+Na]+ m/z 293.11 (calcd for C16H18N2NaO2 293.13).

1, 3-Bis (4-methoxybenzyl) urea (MMU)

To an ice-cold solution of triphosgene (100 mg, 0.34 mmol) in 2 ml DCM was added dropwise a solution of 4-methoxybenzylamine (137 mg, 1 mmol) and N,N,-diisopropylethylamine (DIEA) (191 μl, 1.1 mmol) in 2 ml of DCM. This mixture was stirred for 5 min at 0°C. To this mixture was added 4-methoxybenzylamine (137 mg, 1 mmol) and DIEA (191 μl, 1.1 mmol) in 2 ml of DCM, warmed slowly to room temperature, and stirred overnight. To this solution was added DCM, and washed with 1 M aqueous HCl solution three times. The DCM layer was dried over MgSO4 and concentrated. Recrystallization from the DCM resulted in 45 mg (0.15 mmol, 15%) of target compound as a white powder; mp 177.7–178.4°C; 1H-NMR (DMSO-d6, 600 MHz); δ (ppm) 7.16 (4H, d, J = 9.0 Hz), 6.87 (4H, d, J = 8.4), 6.27 (2H, t, J = 6.0), 4.14 (4H, d, J = 6.0), 3.72 (6H, s), 13C NMR (151 MHz, DMSO-d6): δ 158.0, 158.0, 132.8, 128.3, 113.6, 55.0, 42.4; IR (cm-1) 3350, 3318, 2955, 2924, 2837, 1624, 1577, 1509, 1237, 808, ESI-MS [M+Na]+ m/z 323.13 (calcd for C17H20N2NaO3 323.14).

LC-MS/MS analysis

Urea compounds were quantified using a Waters Quattro Premier triple quadrupole tandem mass spectrometer (Micromass, Manchester, UK) interfaced to an electrospray ionization (ESI) source. The ESI was performed following HPLC in the positive mode at 2.51 kV capillary voltage. The source and the desolvation temperatures were set at 120 and 300°C, respectively. Cone gas (N2) and desolvation gas (N2) were maintained at flow rates of 10 and 700 l/h, respectively. Optimized conditions for mass spectrometry are shown in Table 1. Dwell time was set to 0.1 s. A regression curve for each compound was obtained from at least eight different concentrations of standard stock solutions (r2 > 0.99). 1, 3-Diphenylurea (DPU) was used as an internal standard and was added just before the analysis. The final concentration of 1, 3-diphenylurea was adjusted to 100 nM. Full scan MS/MS spectra (m/z interval of 10–300) were collected for each of the molecules (precursor ion; MMU m/z: 301, BMU m/z: 271, BBU m/z: 241) using product ion scan mode with cone voltage and collision voltage of 35V and 26V, respectively. The MS was coupled with a Waters Acquity UPLC (Waters, and Milford, MA, USA). The HPLC column Imtakt Cadenza CD-C18 (CD007, 3 μm, 4.6x500 mm) was used to separate these analytes. The HPLC solvent gradient is shown in Table 2. The infusion flow rate to the MS was adjusted to 0.3 ml/min.

thumbnail
Table 1. Optimum mass transition conditions and key fragmentation of the urea compounds.

https://doi.org/10.1371/journal.pone.0117438.t001

thumbnail
Table 2. HPLC solvent gradient for the separation of target ureas.

https://doi.org/10.1371/journal.pone.0117438.t002

Standard addition and recovery experiments

A solution of BBU (20 nmol), BMU (100 nmol), and MMU (200 nmol) in 10 μl of methanol was spiked into 100 mg of dried root powder. The powder was mixed, dried, and kept at -20°C until extraction. Samples were extracted as described above. The recovery percentage was calculated as follows:

Enzyme Purification

Recombinant human and recombinant rat sEH were produced in insect High Five cells using recombinant baculovirus expression vectors, and purified by affinity chromatography as reported previously [28,29]. Each enzyme appeared as a single band (0.3 μg loading) with an estimated purity of more than 95% by Coomassie Brilliant Blue staining following SDS-PAGE separation (S1 Fig.). The final recombinant sEH preparations had no esterase or glutathione S-transferase activity which interferes with the CMNPC assay as described below.

Measurement of sEH inhibition by fluorescent assay (CMNPC assay)

IC50 values were determined as described previously [30] using cyano (2-methoxynaphthalen-6-yl) methyl trans-(3-phenyl-oxyran-2-yl) methyl carbonate (CMNPC) as a fluorescent substrate. Recombinant human or recombinant rat sEH (0.96 nM) was incubated with crude root extract or inhibitors for 5 min in 25 mM bis-Tris/HCl buffer (pH 7.0) containing 0.1 mg/mL of BSA at 30°C prior to substrate introduction ([S] = 5 μM). Activity was measured by determining the appearance of the 6-methoxy-2-naphthaldehyde with an excitation wavelength of 330 nm and an emission wavelength of 465 nm for 10 min.

Measurement of sEH inhibition by radioactivity-based assay (t-DPPO assay)

IC50 values were determined as described previously [31] with slight modifications, using racemic [3H] trans-diphenylpropene oxide (t-DPPO). Purified recombinant human sEH was diluted in 100 mM sodium phosphate buffer (pH 7.4) containing 0.1 mg/ml BSA, and was incubated in triplicate with inhibitors for 5 min at 30°C prior to the introduction of the radiolabeled substrate (t-DPPO: 50 μM; ∼10,000 cpm/assay). The mixture was incubated at 30°C for 10 min and the reaction was quenched by the addition of 60 μl of methanol. The remaining substrate was extracted by vigorous mixing with 200 μl of isooctane. The radioactivity of the aqueous phase was measured using a liquid scintillation counter (Perkin Elmer Tri-Carb 2810TR, Shelton, CT). Epoxide hydrolase activity was determined as a percentage of the radioactivity corresponding to the diol in the aqueous phase relative to the control.

Enzyme kinetics

Dissociation constants were determined following the method described by Dixon [32] for competitive tight binding inhibitors, using t-DPPO as a substrate [31]. Inhibitor MMU at concentrations between 0 and 1000 nM was incubated in triplicate for 5 min in 100 mM sodium phosphate buffer (pH 7.4) at 30°C with 100 μl of the enzyme (2 nM of recombinant human sEH). Substrate (4.0 ≤ [S] final ≤ 30 μM) was then added. Velocity was measured as described previously [31]. For each substrate concentration, the plots of the velocity as a function of the inhibitor concentration allow the determination of an apparent inhibition constant (Kiapp [32]). The plot of the Kiapp values as a function of the substrate concentration allows the determination of Ki when [S] = 0. Kiapp was measured in at least 3 separate assays. Results are presented as average ± SE.

Measurement of solubility of MMU

An excess amount of MMU was added to a vial containing sodium phosphate buffer, 0.1 M pH 7.4 (0.25 mL), and the MMU suspension was equilibrated during 1 h of sonication and 48 h of shaking at 25°C, followed by centrifugation. The amount of MMU in the supernatant was analyzed by LC—MS/MS. The result is presented as mean ± SD of triplicate measurement.

Nociceptive assay using rat inflammatory Pain Model

Male Sprague-Dawley rats weighing 235–280 g were obtained from Charles River Laboratories and maintained in the UC Davis animal housing facility with ad libitum water and food on a 12 h/12 h light-dark cycle. Behavioral nociceptive testing was conducted by assessing mechanical withdrawal threshold using an electronic von Frey anesthesiometer apparatus (IITC, Woodland Hills, CA) [33]. The controller was set to “maximum holding” mode so that the highest applied force (in grams) upon withdrawal of the paw was displayed. At least three measurements were taken at 1–2 min inter-stimulus intervals. Data were normalized to percentage values using the formula (mechanical withdrawal threshold (g)) ×100/ (mechanical withdrawal threshold (g) of naïve animals). The analgesic effect of MMU was tested using the intraplantar carrageenan elicited local inflammatory pain model [16,33]. Following baseline measurements, carrageenan (50 μl, 1% solution of carrageenan) was administered into the plantar area of one hind paw in order to induce inflammatory pain. At 4 h post administration of carrageenan, postcarrageenan responses were measured, and immediately afterwards, MMU (10 μg in 10 μl of PEG400), morphine sulfate (10 μg in 10 μl saline), the potent synthetic sEH inhibitor 1-(1-(Cyclopropanecarbonyl) piperidin-4-yl)-3-(4-(trifluoromethoxy) phenyl) urea (TPCU) (10 ng in 10 μl of PEG400), or the vehicle (10 μl of PEG400) was administered into the inflamed paw by intraplantar injection. Subsequently, the ability of these treatments to reduce the carrageenan-induced inflammatory pain was monitored over the course of 2.5 h.

Statistical Analysis

Data were analyzed using SigmaPlot 11.0 for windows (Systat Software Inc., San Jose, CA). Kruskal-Wallis One Way ANOVA on Ranks followed by Tukey Test was performed with p values<0.05 considered signicant.

Results and Discussion

Inhibition of human soluble epoxide hydrolase by crude root extract

Determination of the plant species of the root tissues was supported by comparing the partial sequences of 18S ribosomal DNA (NCBI accession number: AF070972) and maturase K DNA (AY483239) as described in a previous study [34]. The sequence alignments are shown in S1 Text. The crude root extract of P. brazzeana was prepared by percolation in DCM-methanol (1:1), and the inhibitory activity of the extract toward human sEH was measured by both fluorescent assay (CMNPC assay) and radioactivity based assay (t-DPPO assay) [30,31]. The root extract showed an IC50 value of 1.9 ± 0.4 μg crude extract/ml in the CMNPC assay and 35 ± 3 μg crude extract/ml in the t-DPPO assay.

Optimization of LC-MS/MS and analysis of plant extracts

Two of 3 urea derivatives found in P. brazzeana as reported by Tsopmo et al. [20] were synthetically prepared as shown in Fig. 1. Fig. 2A shows a representative chromatogram of the separation of synthetic standards of the 3 ureas reported by Tsopmo et al. with our internal standard, 1,3-diphenylurea (DPU). The LC-MS/MS analysis clearly showed that the plant root extract contains these ureas, based on the retention time of the analytes and the mass to charge ratio of the molecular and fragment ions (Figs. 2B and S2S4).

thumbnail
Fig 2. Root of P. brazzeana contains urea-based sEH inhibitors.

The chemical structure of target analytes and their mass transition are shown as inserts in the figure. The LC-MS/MS conditions are described in Materials and Methods. (A) MRM chromatograms of synthetic standard ureas. (B) MRM chromatograms of crude root extract from P. brazzeana (250 μg root/ml, 10μl). cps: counts per second.

https://doi.org/10.1371/journal.pone.0117438.g002

To determine the concentration of these ureas in plant samples an LC-MS/MS method was optimized in multiple reaction monitoring (MRM) mode. The concentrations of these ureas are shown in Table 3. 1, 3-bis (4-Methoxybenzyl) urea (MMU) was the most abundant, followed by an asymmetric urea (BMU) and then non-substituted benzyl urea (BBU). In order to determine the extraction efficiency from the plant sample, standard addition and recovery experiments were performed. As shown in Table 3, the recovery percentages of MMU, BMU, and BBU were between 89–99%, showing that our method efficiently extracted the target analytes.

thumbnail
Table 3. Concentration of ureas in the root of P. brazzeana and extraction recovery percentage.

https://doi.org/10.1371/journal.pone.0117438.t003

sEH inhibitory activity of ureas from P. brazzeana

The ability of MMU, BMU, and BBU to inhibit the activity of human sEH was measured using two different substrates. The IC50 values of these urea compounds are shown in Table 4. As reported previously, the t-DPPO assay gives higher IC50 values (lower potency) for the same compound than the CMNPC assay [30]. The symmetric bis (methoxybenzyl) urea (MMU), which was found at the highest concentration in the root extract, showed the highest potency toward human sEH when determined with the CMNPC or t-DPPO assays. The inhibitory activity of the asymmetric urea (BMU) was of about 4-fold lower than that of MMU with both assays. The non-substituted benzyl urea, BBU, showed the lowest potency. These results are consistent with the previous structure-activity relationship of sEH inhibitors in that para methoxy substitution on the phenyl ring leads to higher potency in comparison to non-substituted inhibitors [16].

thumbnail
Table 4. sEH inhibition potency of ureas found in the root of P. brazzeana.

https://doi.org/10.1371/journal.pone.0117438.t004

In order to compare our compounds with previously reported natural product sEH inhibitors, the potency of honokiol was measured using our radioactivity-based assay. Honokiol showed an IC50 of 20.3 μM with the t-DPPO assay. Based on this result, MMU is about 8-fold more potent than honokiol.

The inhibitory potency of these compounds against affinity purified, recombinant rat sEH was measured using the CMNPC assay. The ureas from P. brazzeana showed a similar pattern of inhibitory potency with the rat recombinant sEH as was found with the recombinant human sEH (Table 4).

Enzyme inhibition kinetics

In order to define the mode of enzyme inhibition and to determine the dissociation constant (Ki), we performed enzyme kinetic analysis of MMU using recombinant human sEH. As shown in the insert in Fig. 3, the Kiapp values increase with an increase in the concentration of the substrate. This strongly suggests that MMU inhibits sEH as a competitive inhibitor, which is consistent with other urea-based sEH inhibitors [18]. The dissociation constant (Ki) of MMU was determined to be 54±13 nM. In terms of potency this is approximately 40-fold lower than the highly potent synthetic inhibitor t-AUCB (Ki = 1.5 nM), but approximately 2-fold higher than the synthetic sEH inhibitor APAU (Ki = 125 nM) which has undergone a phase II clinical trial [35,36].

thumbnail
Fig 3. Enzyme inhibition kinetics of MMU on human soluble epoxide hydrolase.

Enzyme kinetic analysis was performed on recombinant human soluble epoxide hydrolase (2 nM) using [3H] t-DPPO as a substrate. For each substrate concentration (4.0–30.0 μM), the velocity was plotted as a function of inhibitor concentration (0–1000 nM), allowing the determination of an apparent inhibition constant (Kiapp). Kiapp values are plotted as a function of the substrate concentration (insert). For [S] = 0, a Ki value of 54 nM was found.

https://doi.org/10.1371/journal.pone.0117438.g003

Nociceptive assay in rat inflammatory pain model

Several studies have shown that sEH inhibitors effectively reduce inflammatory pain [4,5,16]. In order to evaluate the in vivo efficacy of MMU, a rat inflammatory pain model was used to measure its analgesic effects. MMU was selected based on its inhibitory potency on sEH and its concentration in P. brazzeana. As shown in Fig. 4, local administration of either MMU (10 μg/intraplantar injection), morphine (10 μg/intraplantar injection), or potent synthetic sEH inhibitor TPCU (10 ng/intraplantar injection) significantly reduced carrageenan induced inflammatory pain (Kruskal-Wallis One Way ANOVA on Ranks, p≤0.001). In this assay, MMU, TPCU, and morphine appeared to have similar efficacy in terms of reducing enhanced pain perception but MMU showed a longer duration of efficacy compared to morphine (MMU vs morphine, Tukey’s post hoc test, p<0.05). TPCU showed an IC50 of 0.4 nM with Ki value of 0.67 nM on human sEH [16,36], which is approximately 230-fold more potent than MMU based on the IC50 values. Thus, it is reasonable that MMU showed similar or slightly better efficacy in comparison to TPCU when MMU was administered at a 1000-fold higher dose than TPCU.

thumbnail
Fig 4. MMU effectively reduces carrageenan-induced inflammatory pain in rat.

Administration of the inflammatory agent carrageenan (CAR) induces a stable hyperalgesic response for the duration of the experiment. Treatment with MMU (▲, 10 μg/paw, intraplantar), morphine (●, 10 μg/paw, intraplantar) or the potent synthetic sEH inhibitor (TPCU) (△, 10 ng/paw, intraplantar) significantly reduced pain levels (Kruskal-Wallis One Way ANOVA on Ranks, p≤0.001, Tukey’s post hoc test (MMU vs vehicle, morphine vs vehicle, TPCU vs vehicle, and MMU vs morphine) p<0.05). Mean ± SE (n = 6) of mechanical withdrawal threshold (% of naïve baseline) are shown. Vehicle (○), morphine (●), and TPCU (△) treatment data are from Rose et al., (2010).

https://doi.org/10.1371/journal.pone.0117438.g004

The root of P. brazzeana is used as a traditional analgesic against various types of pain [23,25]. For example, dried root bark powder is applied by scarification to treat intercostal and abdominal pain, while the crushed root or root bark is applied as a topical ointment or as an infusion drink to soothe chest pain, lumbago, rheumatism, and haemorrhoids [23]. In order to relate our results to the ethnomedicinal use of P. brazzeana, it is necessary to determine how much of the ureas are absorbed into the body, and whether they reach a dose that can effectively inhibit sEH. In the case of application by scarification, which is similar to intraplantar injection, these urea compounds could be absorbed and reach the site of action. In the case of oral administration, MMU may have limited bioavailability given that the synthetic MMU has low solubility (10.6±2.1 μM (mean±SD)) in sodium phosphate buffer (pH7.4). It should be noted, however, that the root of P. brazzeana may have compounds such as lipids and polysaccharides, which could solubilize MMU, BMU or BBU and facilitate their absorption after ingestion. Determination of the bioavailability of these inhibitors after oral administration of the root, or topical ointment treatment is the subject of further research. A recent study showed that sEH inhibitors are highly effective against neuropathic diabetic pain [8]. Determining the efficacy of the compound/plant extract on neuropathic pain is also the subject of further study. Determining the localization of MMU within the plant root, especially whether it is localized within the root bark, will help us to optimize a method for efficient isolation of MMU or other target ureas from P. brazzeana.

The theoretical IC50 of the crude extract of P. brazzeana against recombinant human sEH is approximately 29 μg crude root extract/ml using the CMNPC assay. This calculated IC50 of the crude root extract is based on the IC50 of MMU and its concentration in the plant (assuming for simplicity that MMU is the main component that inhibits sEH). Experimentally, however, the crude root extract gave an IC50 of 1.9 μg crude root extract/ml. This experimentally-derived potency is approximately 15-fold higher than the calculated potency, suggesting that the crude root extract contains unknown sEH inhibitor(s), or another factor that improves its potency. It should be noted, however, that in different preparations of the crude root extract, significant variation in the inhibitory potency (IC50 = 4.1–176 μg/ml in t-DPPO assay) was found while the concentrations of MMU, BMU, and BBU were very similar. This may indicate that the root extract contains unknown inhibitors that are sensitive to the extraction process that is utilized. Optimization of the extraction/processing method is still needed in order to precisely measure the level of sEH inhibition by the crude plant extract. In our preliminary HPLC separation of the crude root extract and measurement of sEH inhibition by each fraction, we observed strong sEH inhibition in fractions that were different from the fractions containing MMU (S5 and S7 Figs., S1 and S2 Tables). This indicates that there are other unidentified sEH inhibitors in this plant. Addition of dithiothreitol (100 μM) to either the crude extract or to each fraction did not change the inhibitory potency, indicating that the unknown inhibitors are not Michael acceptors. The purification and identification of these inhibitors are the subjects of further study.

MMU and BMU have not been reported in other plants, while BBU has been detected in plants such as moringa (Moringa oleifera) and garden cress (Lepidium sativum) [37,38]. A regioisomer of MMU, bis (m-methoxybenzyl) urea (salvadourea), was reported in the miswak, Salvadora persica, plant [39]. Interestingly, P. brazzeana, moringa, garden cress, and miswak all belong to the plant order Brassicales. In addition these plants produce glucosinolates or their corresponding isothiocyanate products [4043], suggesting that they may share a metabolic pathway. A genetic comparison between these species based on ribosomal DNA sequences, however, failed to find any similarity between these species. Chemotaxonomical analysis is needed to find shared biosynthetic pathways in these plants.

Conclusions

We demonstrate that urea compounds found in the root of P. brazzeana have potent sEH inhibitory activities. These inhibitors were quantified by LC-MS/MS, showing that the most potent inhibitor, MMU, is found at the highest concentration in the plant root in comparison to BMU and BBU. MMU showed local analgesic effects in an in vivo inflammatory pain model. These findings may explain, at least in part, the pharmacological mechanisms of traditional medicinal uses of the root of P. brazzeana. Our findings illustrate that plants could provide leads for novel and therapeutically valuable sEH inhibitors and that plant natural products that inhibit sEH could be valuable supplements to an omega-3 fatty acid-rich diet.

Supporting Information

S1 Fig. SDS-PAGE analysis of human and rat recombinant sEH for purity assessment.

Each enzyme appeared as a single band (0.3 μg loading) of ca. 62 kDa by Coomassie Brilliant Blue staining following SDS-PAGE separation. The migration of molecular weight markers (in kDa) are indicated to the left.

https://doi.org/10.1371/journal.pone.0117438.s001

(TIF)

S2 Fig. Full scan MS/MS spectra of MMU synthetic standard (upper panel) and plant extract (lower panel).

Full scan MS/MS spectra (m/z interval of 10–300) were collected for MMU setting precursor ion m/z as 301 with cone voltage and collision voltage of 35V and 26V, respectively. cps: counts per second.

https://doi.org/10.1371/journal.pone.0117438.s002

(TIF)

S3 Fig. Full scan MS/MS spectra of BMU synthetic standard (upper panel) and plant extract (lower panel).

Full scan MS/MS spectra (m/z interval of 10–300) were collected for BMU setting precursor ion m/z as 271 with cone voltage and collision voltage of 35V and 26V, respectively. cps: counts per second.

https://doi.org/10.1371/journal.pone.0117438.s003

(TIF)

S4 Fig. Full scan MS/MS spectra of BBU synthetic standard (upper panel) and plant extract (lower panel).

Full scan MS/MS spectra (m/z interval of 10–300) were collected for BBU setting precursor ion m/z as 241 with cone voltage and collision voltage of 35V and 26V, respectively. cps: counts per second.

https://doi.org/10.1371/journal.pone.0117438.s004

(TIF)

S5 Fig. Normal phase HPLC fraction collection and sEH inhibition by the fractions.

Crude root extract (approximately 2 mg) was injected into a normal phase HPLC column (YMC-Pack SIL-06) and eluted with 20% isopropanol in hexane with a flow rate of 4 ml/min for 15 min, followed by 100% isopropanol with a flow rate of 2 ml/min for 25 min. The relative intensity of the UV absorption at the wavelength of 210 and 276 nm are shown (top and middle). Fractions were collected for every 4 ml of eluent. After the solvent was evaporated the residue was reconstituted in 50 μl DMSO. The inhibition percentage by each fractions (100 times dilution of reconstituted solution) was measured using the CMNPC assay with recombinant human sEH (bottom). The black circles (●) represent the inhibition percentage by each of the fractions. The crude extract mixture (100 times dilution of 2 mg extract/ml DMSO) showed complete inhibition of sEH activity (black circle shown at time 0 min). The retention times of the 3 synthetic ureas (BBU, BMU, and MMU) are indicated by arrows in the figure.

https://doi.org/10.1371/journal.pone.0117438.s005

(TIF)

S6 Fig. Procedure for the reverse phase HPLC fraction collection.

https://doi.org/10.1371/journal.pone.0117438.s006

(TIF)

S7 Fig. Reverse phase HPLC fraction collection and sEH inhibition by the fractions.

Crude root extract (approximately 5 mg) was injected into a reverse phase HPLC column (Waters SunFire Prep C18, 5 μm, 10x100 mm) and eluted with 10% acetonitrile in water with a flow rate of 2 ml/min for 10 min, followed by a linear gradient elution of acetonitrile 10% to 100% at a flow rate of 2 ml/min for 25 min, and eluted with 100% acetonitrile for 15 min at a flow rate of 2 ml/min. The relative intensity of the UV absorption at the wavelength of 210 and 280 nm are shown (top and middle). The retention times of the 3 synthetic ureas (BBU, BMU, and MMU) are indicated by an arrow in the figure. Fractions were collected for every 4 ml of eluent. After the solvent was evaporated the residue was reconstituted in 50 μl DMSO. The inhibition percentage by each fractions (100 times dilution of reconstituted solution) was measured using the CMNPC assay with recombinant human sEH (bottom). The black circles (●) represent the inhibition percentage by each of the fractions. The crude extract C (100 times dilution of 5 mg extract/ml DMSO) showed complete inhibition of sEH activity (black circle shown at time 0 min).

https://doi.org/10.1371/journal.pone.0117438.s007

(TIF)

S1 Table. Relative potency of normal phase HPLC fractions.

https://doi.org/10.1371/journal.pone.0117438.s008

(DOCX)

S2 Table. Relative potency of reverse phase HPLC fractions.

https://doi.org/10.1371/journal.pone.0117438.s009

(DOCX)

S3 Table. Effect of extraction solvent on human sEH inhibitory potency.

https://doi.org/10.1371/journal.pone.0117438.s010

(DOCX)

S1 Text. Sequence alignments of PCR products from root sample of P. brazzeana vs sequences in NCBI database.

https://doi.org/10.1371/journal.pone.0117438.s011

(DOCX)

S2 Text. Methods for HPLC fraction collection and sEH inhibition by the fractions.

https://doi.org/10.1371/journal.pone.0117438.s012

(DOCX)

Author Contributions

Conceived and designed the experiments: SK. Performed the experiments: SK CM BI SGK GRDN. Analyzed the data: SK CM. Contributed reagents/materials/analysis tools: GRDN MN. Wrote the paper: SK CM BI SGK GRDN MN BDH. Wrote the first draft and finalized the manuscript: SK.

References

  1. 1. Morisseau C, Hammock BD (2013) Impact of Soluble Epoxide Hydrolase and Epoxyeicosanoids on Human Health. Annu Rev Pharmacol Toxicol 53: 37–58. pmid:23020295
  2. 2. Morisseau C, Inceoglu B, Schmelzer K, Tsai HJ, Jinks SL, et al. (2010) Naturally occurring monoepoxides of eicosapentaenoic acid and docosahexaenoic acid are bioactive antihyperalgesic lipids. J Lipid Res 51: 3481–3490. pmid:20664072
  3. 3. Lin W-K, Falck JR, Wong PYK (1990) Effect of 14,15-epoxyeicosatrienoic acid infusion on blood pressure in normal and hypertensive rats. Biochemical and Biophysical Research Communications 167: 977–981. pmid:2322287
  4. 4. Inceoglu B, Jinks SL, Schmelzer KR, Waite T, Kim IH, et al. (2006) Inhibition of soluble epoxide hydrolase reduces LPS-induced thermal hyperalgesia and mechanical allodynia in a rat model of inflammatory pain. Life Sci 79: 2311–2319. pmid:16962614
  5. 5. Inceoglu B, Schmelzer KR, Morisseau C, Jinks SL, Hammock BD (2007) Soluble epoxide hydrolase inhibition reveals novel biological functions of epoxyeicosatrienoic acids (EETs). Prostaglandins Other Lipid Mediat 82: 42–49. pmid:17164131
  6. 6. Wagner K, Inceoglu B, Gill SS, Hammock BD (2011) Epoxygenated fatty acids and soluble epoxide hydrolase inhibition: novel mediators of pain reduction. J Agric Food Chem 59: 2816–2824. pmid:20958046
  7. 7. Yu Z, Xu F, Huse LM, Morisseau C, Draper AJ, et al. (2000) Soluble Epoxide Hydrolase Regulates Hydrolysis of Vasoactive Epoxyeicosatrienoic Acids. Circ Res 87: 992–998. pmid:11090543
  8. 8. Inceoglu B, Wagner KM, Yang J, Bettaieb A, Schebb NH, et al. (2012) Acute augmentation of epoxygenated fatty acid levels rapidly reduces pain-related behavior in a rat model of type I diabetes. Proc Natl Acad Sci U S A 109: 11390–11395. pmid:22733772
  9. 9. Guedes AGP, Morisseau C, Sole A, Soares JHN, Ulu A, et al. (2013) Use of a soluble epoxide hydrolase inhibitor as an adjunctive analgesic in a horse with laminitis. Veterinary Anaesthesia and Analgesia 40: 440–448. pmid:23463912
  10. 10. A randomized, double-blind, placebo-controlled, dose-ranging, exploratory, 28-day study to examine the effects of AR9281 on blood pressure and glucose tolerance in patients with mild to moderate hypertension and impaired glucose tolerance. ClinicalTrials.gov, ID NCT00847899. Available: http://clinicaltrials.gov/ct2/show/NCT00847899?term=NCT00847899.&rank=1, Accessed 2014 Apr 25.
  11. 11. A Study to Assess the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of Single Doses of GSK2256294 in Healthy Volunteers, and Single and Repeat Doses of GSK2256294 in Adult Male Moderately Obese Smokers. ClinicalTrials.gov, ID NCT01762774, Available: http://www.clinicaltrials.gov/ct2/show/NCT01762774?term=soluble+epoxide+hydrolase&rank=2, Accessed 2014 Apr 25.
  12. 12. A Study to Investigate the Safety and Pharmacokinetics of a Single Dose of GSK2256294 in Healthy Young Males and Elderly Subjects. ClinicalTrials.gov, ID NCT02006537, Available: https://clinicaltrials.gov/ct2/show/NCT02006537?term=GSK2256294&rank=2, Accessed 2014 Dec 12.
  13. 13. Zhang G, Panigrahy D, Mahakian LM, Yang J, Liu J-Y, et al. (2013) Epoxy metabolites of docosahexaenoic acid (DHA) inhibit angiogenesis, tumor growth, and metastasis. Proc Natl Acad Sci U S A 110: 6530–6535. pmid:23553837
  14. 14. El Buscató, Büttner D, Brüggerhoff A, Klingler F-M, Weber J, et al. (2013) From a Multipotent Stilbene to Soluble Epoxide Hydrolase Inhibitors with Antiproliferative Properties. ChemMedChem 8: 919–923. pmid:23596124
  15. 15. Lee GH, Oh SJ, Lee SY, Lee J-Y, Ma JY, et al. (2014) Discovery of soluble epoxide hydrolase inhibitors from natural products. Food and Chemical Toxicology 64: 225–230. pmid:24309146
  16. 16. Rose TE, Morisseau C, Liu JY, Inceoglu B, Jones PD, et al. (2010) 1-Aryl-3-(1-acylpiperidin-4-yl)urea inhibitors of human and murine soluble epoxide hydrolase: structure-activity relationships, pharmacokinetics, and reduction of inflammatory pain. J Med Chem 53: 7067–7075. pmid:20812725
  17. 17. Thalji RK, McAtee JJ, Belyanskaya S, Brandt M, Brown GD, et al. (2013) Discovery of 1-(1,3,5-triazin-2-yl)piperidine-4-carboxamides as inhibitors of soluble epoxide hydrolase. Bioorg Med Chem Lett 23: 3584–3588. pmid:23664879
  18. 18. Morisseau C, Goodrow MH, Dowdy D, Zheng J, Greene JF, et al. (1999) Potent urea and carbamate inhibitors of soluble epoxide hydrolases. Proc Natl Acad Sci U S A 96: 8849–8854. pmid:10430859
  19. 19. Shen HC, Hammock BD (2012) Discovery of inhibitors of soluble epoxide hydrolase: a target with multiple potential therapeutic indications. J Med Chem 55: 1789–1808. pmid:22168898
  20. 20. Tsopmo A, Ngnokam D, Ngamga D, Ayafor JF, Sterner O (1999) Urea Derivatives from Pentadiplandra brazzeana. Journal of Natural Products 62: 1435–1436. pmid:10543911
  21. 21. Jiofack T, Ayissi I, Fokunang C, Guedje N, Kemeuze V (2009) Ethnobotany and phytomedicine of the upper Nyong valley forest in Cameroon. Afr J Pharm Pharmacol 3: 144–150.
  22. 22. Titanji VP, Zofou D, Ngemenya MN (2008) The antimalarial potential of medicinal plants used for the treatment of malaria in Cameroonian folk medicine. Afr J Trad Complement Altern Med 5: 302–321. pmid:20161952
  23. 23. Dounias E (2008) Pentadiplandra brazzeana Baill. In: Schmelzer GH, Gurib-Fakim A (Editors). Prota 11(1): Medicinal plants/Plantes médicinales 1. [CD-Rom]. PROTA, Wageningen, Netherlands.
  24. 24. Makumbelo E, Lukoki L, Paulus J, Luyindula N (2008) Stratégie de valorisation des espèces ressources des produits non ligneux de la savane des environs de Kinshasa:II. Enquête ethnobotanique (aspects médicinaux). Tropicultura 26: 129–134.
  25. 25. Betti JL (2004) An ethnobotanical study of medicinal plants among the Baka pygmies in the Dja biosphere reserve, Cameroon. Afr Study Monogr 25: 1–27.
  26. 26. Nyegue M, Ndoyé F, Amvam Zollo P, Etoa F, Agnaniet H, et al. (2009) Chemical and biological evaluation of essential oil of Pentadiplandra brazzeana (Bail.) roots from Cameroon. Adv Phytotherapy Res: 91–107.
  27. 27. Nakagawa Y, Wheelock CE, Morisseau C, Goodrow MH, Hammock BG, et al. (2000) 3-D QSAR analysis of inhibition of murine soluble epoxide hydrolase (MsEH) by benzoylureas, arylureas, and their analogues. Bioorg Med Chem 8: 2663–2673. pmid:11092551
  28. 28. Beetham JK, Tian TG, Hammock BD (1993) cDNA Cloning and Expression of a Soluble Epoxide Hydrolase from Human Liver. Arch Biochem Biophys 305: 197–201. pmid:8342951
  29. 29. Wixtrom RN, Silva MH, Hammock BD (1988) Affinity purification of cytosolic epoxide hydrolase using derivatized epoxy-activated Sepharose gels. Anal Biochem 169: 71–80. pmid:3369689
  30. 30. Jones PD, Wolf NM, Morisseau C, Whetstone P, Hock B, et al. (2005) Fluorescent substrates for soluble epoxide hydrolase and application to inhibition studies. Anal Biochem 343: 66–75. pmid:15963942
  31. 31. Borhan B, Mebrahtu T, Nazarian S, Kurth MJ, Hammock BD (1995) Improved Radiolabeled Substrates for Soluble Epoxide Hydrolase. Anal Biochem 231: 188–200. pmid:8678300
  32. 32. Dixon M (1972) The graphical determination of K m and K i. Biochem J 129: 197–202. pmid:4630451
  33. 33. Inceoglu B, Jinks SL, Ulu A, Hegedus CM, Georgi K, et al. (2008) Soluble epoxide hydrolase and epoxyeicosatrienoic acids modulate two distinct analgesic pathways. Proc Natl Acad Sci U S A 105: 18901–18906. pmid:19028872
  34. 34. Rodman J, Soltis P, Soltis D, Sytsma K, Karol K (1998) Parallel evolution of glucosinolate biosynthesis inferred from congruent nuclear and plastid gene phylogenies. American Journal of Botany 85: 997. pmid:21684983
  35. 35. Liu JY, Park SH, Morisseau C, Hwang SH, Hammock BD, et al. (2009) Sorafenib has soluble epoxide hydrolase inhibitory activity, which contributes to its effect profile in vivo. Mol Cancer Ther 8: 2193–2203. pmid:19671760
  36. 36. Lee KS, Morisseau C, Yang J, Wang P, Hwang SH, et al. (2013) Forster resonance energy transfer competitive displacement assay for human soluble epoxide hydrolase. Anal Biochem 434: 259–268. pmid:23219719
  37. 37. Sashidhara KV, Rosaiah JN, Tyagi E, Shukla R, Raghubir R, et al. (2009) Rare dipeptide and urea derivatives from roots of Moringa oleifera as potential anti-inflammatory and antinociceptive agents. Eur J Med Chem 44: 432–436. pmid:18243423
  38. 38. Bahroun A, Damak M (1985) Contribution to the study of Lepidium sativum (Cruciferae). Structure of a new compound isolated from the seed: lepidine. Journal de la Societe Chimique de Tunisie 2: 15–24.
  39. 39. Ray A, Chand L, Dutta S (1975) Salvadourea. New urea derivative from Salvadora persica. Chem Ind 12: 517–518.
  40. 40. Bennett RN, Mellon FA, Foidl N, Pratt JH, Dupont MS, et al. (2003) Profiling Glucosinolates and Phenolics in Vegetative and Reproductive Tissues of the Multi-Purpose Trees Moringa oleifera L. (Horseradish Tree) and Moringa stenopetala L. J Agric Food Chem 51: 3546–3553. pmid:12769522
  41. 41. Gil V, MacLeod AJ (1980) Studies on glucosinolate degradation in Lepidium sativum seed extracts. Phytochemistry 19: 1369–1374.
  42. 42. De Nicola GR, Nyegue M, Montaut S, Iori R, Menut C, et al. (2012) Profile and quantification of glucosinolates in Pentadiplandra brazzeana Baillon. Phytochemistry 73: 51–56. pmid:21993210
  43. 43. Sofrata A, Santangelo EM, Azeem M, Borg-Karlson A- K, Gustafsson A, et al. (2011) Benzyl Isothiocyanate, a Major Component from the Roots of Salvadora Persica Is Highly Active against Gram-Negative Bacteria. PLOS One 6: e23045. pmid:21829688