Next Article in Journal
Development of New 1,3-Diazaphenoxazine Derivatives (ThioG-Grasp) to Covalently Capture 8-Thioguanosine
Previous Article in Journal
TiO2 and Fe2O3 Films for Photoelectrochemical Water Splitting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

An Improved HPLC-DAD Method for Quantitative Comparisons of Triterpenes in Ganoderma lucidum and Its Five Related Species Originating from Vietnam

1
National Institute of Medicinal Materials (NIMM), 3B Quangtrung, Hoankiem, Hanoi 100000, Vietnam
2
Faculty of Chemistry, University of Science, Vietnam National University-HoChiMinh City, 227 Nguyen Van Cu, District 5, Ho Chi Minh City 227-01, Vietnam
3
College of Pharmacy, Catholic University of Daegu, Gyeungbuk 712–702, Korea
4
Department of Biology, University of Taynguyen, 567 Le Duan, Dac Lac City 630000, Vietnam
*
Authors to whom correspondence should be addressed.
Molecules 2015, 20(1), 1059-1077; https://doi.org/10.3390/molecules20011059
Submission received: 25 November 2014 / Accepted: 5 January 2015 / Published: 9 January 2015
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
An HPLC-DAD method for the quality control of wild and cultivated Ganoderma lucidum (Linhzhi) and related species samples was developed and validated. The quantitative determination of G. lucidum and its related species using 14 triterpene constituents, including nine ganoderma acids (compounds 412), four alcohols (compounds 1316), and one sterol (ergosterol, 17) were reported. The standard curves were linear over the concentration range of 7.5–180 µg/mL. The LOD and LOQ values for the analyses varied from 0.34 to 1.41 µg/mL and from 1.01 to 4.23 µg/mL, respectively. The percentage recovery of each reference compound was found to be from 97.09% to 100.79%, and the RSD (%) was less than 2.35%. The precision and accuracy ranged from 0.81%–3.20% and 95.38%–102.19% for intra-day, and from 0.43%–3.67% and 96.63%–103.09% for inter-day, respectively. The study disclosed in detail significant differences between the quantities of analyzed compounds in different samples. The total triterpenes in wild Linhzhi samples were significantly higher than in cultivated ones. The total constituent contents of the five related Linhzhi samples were considerably lower than that in the G. lucidum specimens, except for G. australe as its constituent content outweighed wild Linhzhi’s content by 4:1.

1. Introduction

Ganoderma lucidum (Leyss. Ex Fr.) Karst (Linhzhi) is an ancient traditional medicine that is highly valued in Asian countries as well as in the West for the treatment and prevention of many diseases such as neurasthenia, insomnia, anorexia, dizziness, chronic hepatitis, hypercholesterolemia, mushroom poisoning, coronary heart disease, hypertension, and carcinoma [1,2]. Linhzhi has been considered a tincture of life for thousands of years. Currently, a number of Linhzhi commercial products such as teas, capsules, tablets, raw herb, powders, and extracts are available and are used medicinally [1,2,3]. A number of bioactive components from G. lucidum have been identified, including polysaccharides, triterpenoids, lectins, steroids, and proteins, which have been used in the treatment and the prevention numerous of diseases [2]. Among those components, triterpenoids, including highly oxygenated lanostane derivatives and common fungal steroids derived from ergosterol, have received considerable attention owing to their well-known pharmacological activities such as anti-HIV-1 (ganoderiol F, ganodermanontriol, ganoderic acids A, B, H, and C1) [4], anticholesterol (ganoderic acids A, B, and C) [5], antinociceptive (ganoderic acids A, B, G, and C6) [6], hepatoprotective (ganodermanontriol, ergosterol) [7], antihistamine (ganoderic acids C2 and D) [8], and antitumor (ganoderic acids T, Me) [9] activities, as well as H/R-induced oxidative stress and inflammatory responses (ergosterol) [10]. Hence, triterpenoids could be considered the “marker compounds” for the chemical evaluation and the standardization of the bioactive components from G. lucidum. The chemical structures of the triterpenoids in G. lucidum are quite similar and difficult to isolate in sufficient amounts for the simultaneous analysis of multiple components. Therefore, the quality control and standardization of the isolation of the components of G. lucidum is a challenging task. However, high-performance liquid chromatography (HPLC) has been used successfully to overcome these difficulties [11,12,13,14].
Despite the valuable dietary and therapeutic benefits of Linhzhi, chemical investigations of the active components have been conducted mainly in China, Korea, Japan, and United States, but few experiments with the local Ganoderma genus (the family: Ganodermataceae) have been carried out [12,15,16]. In Vietnam, several studies have been reported regarding the taxonomy and distribution of Vietnamese Ganodermataceae [17,18]. Vietnam possesses 27 species of the Ganoderma genus [18]; however, the classification and distribution of the Ganoderma genus in Vietnam continues to be studied. In a 2013 report by Nguyen et al., 43 species of the Ganoderma genus were identified in the Highlands of Vietnam [17]. Five of these have been successfully cultivated in Vietnam including G. lucidum, G. applanatum, G. australe, G. colossum, and G. subresinosum. However, the chemical compositions of the G. lucidum species collected in Vietnam have not been reported, and no data exist that compare the components of wild-harvested, cultivated, and other related Linhzhi species from Vietnam. In this study, a reverse-phase HPLC method was developed for the fingerprint analysis and simultaneous determination of 17 compounds including a new lanostane triterpene (butyl lucidenate E2 (11) [19], uracil (1), 5-dihydrobenzoic acid (3, gentisic acid) [20], 12 lanostane triterpene derivatives (compounds 410 and 1216), adenosine (2), and ergosterol (17). The developed method was successfully applied to the quantification of 14 triterpenoids in six wild and four cultivated G. lucidum samples, and five related Ganoderma species.

2. Results and Discussion

2.1. Optimization of Sample Preparation Condition

Several methods for the extraction of the fruiting bodies of Ganoderma species were surveyed including ultrasonication, refluxing, and maceration using methanol, but the ultrasonication method was the most effective, therefore we used this method to evaluate the effect of different solvents (100% methanol and 100% ethanol) on the amount of sample extracted. When 100% methanol was used, the content of the sample extracted was higher. To test the necessary time to accomplish the extraction, samples were prepared for 30, 60, 90, and 120 min. Since, the amount of the sample extracted after 90 min was same as the 120 min sample and higher than the 30 min sample, 90 min was selected as the optimal extraction time.

2.2. Selection of HPLC Conditions and Validation of the Developed Method

Although several HPLC methods have been reported for the determination of the constituents of Linhzhi samples [11,12,14,21,22], few constituents have been analyzed within the same study due to a lack of standard reference compounds. Our previous chemical investigation of G. lucidum from Vietnam resulted in the extraction and isolation of 17 compounds 117 (Figure 1). The chemical structures of the isolated compounds were identified using UV-Vis, IR, 1H- and 13C-NMR, and mass spectrometry as well as by the comparison of these spectroscopic data with those reported in literature as a new lanostane triterpene (butyl lucidenate E2 (11) [19], adenosine (2), and two compounds isolated from the first time in G. lucidum, namely uracil (1) and gentisic acid (3) [20]. In addition, 11 lanostane steroids were identified, including lucidenic acid N (4), lucidenic acid E2 (5), ganoderic acid A (6), lucidenic acid A (7), ganoderic acid E (8), methyl lucidenate E2 (9), methyl lucidenate A (10), and butyl ganoderate A (12) [23], lucidadiol (13), ganodermanontriol (14), ganoderiol F (15), ganodermadiol (16), and ergosterol (17) [19,20,24]. The purities of the compounds were greater than 95%, as estimated using an HPLC-DAD method. Most Linhzhi triterpenoids contain a conjugated skeleton, and their UV absorption peaks are concentrated at 210, 237, 243, 253, and 255 nm [11,16]. The analytes were divided into three groups including lanostane triterpenoid-type alcohols and acids, sterol, and others including gentisic acid and adenosine. Based on the maximum absorption of the compounds, the detection wavelengths were set at 256 nm for the acids and their derivatives and 243 nm for the others. The retention times of the compounds in the analyzed samples were distinguished by comparing with those of each reference compound, which are shown in Table 1. The sample preparation conditions for the extraction of the compounds in the Ganoderma species were optimized, which are described in Section 2.1. This study describes the results of the fingerprint analysis of the compounds 117 and the quality analysis of 14 of them (compounds 417). The chromatographic fingerprints of the Ganoderma species are shown in Figure 2, which are divided into three groups including (A) the wild Linhzhi group, (B) the cultivated Linhzhi group, and (C) the related species group.
Figure 1. Chemical compounds 117 isolated from G. lucidum.
Figure 1. Chemical compounds 117 isolated from G. lucidum.
Molecules 20 01059 g001
This HPLC-DAD method was validated for linearity, the limits of detection (LOD) and limits of quantitation (LOQ), recovery, and reproducibility. Each coefficient of correlation (r2) was >0.999, as determined by least square analysis, suggesting good linearity between the peak area ratio versus the compound concentration (Table 1). The LOD and LOQ were examined based on the lowest detectable peak in the chromatogram with a signal-to-noise (S/N) ratio of 3 and 10, respectively. Under our experimental conditions, we determined the LOD and LOQ for the 14 reference compounds in Table 2. The values obtained for both the LOD and LOQ in these analyses were low enough to detect traces of the compounds in the crude extract.
Figure 2. Chromatographic fingerprints of Ganoderma species. (A) the wild collected Linhzhi group (Group A); (B) the cultivated Linhzhi group (Group B); (C) the related Linhzhi species group (Group C). Compounds 117 were isolated from Ganoderma lucidum from the previous studies.
Figure 2. Chromatographic fingerprints of Ganoderma species. (A) the wild collected Linhzhi group (Group A); (B) the cultivated Linhzhi group (Group B); (C) the related Linhzhi species group (Group C). Compounds 117 were isolated from Ganoderma lucidum from the previous studies.
Molecules 20 01059 g002
Table 1. Retention time reproducibilities, regression equations, correlation coefficients (r2), F-test, p (F), linearity ranges, LOD, and LOQ of the compounds 417 (n = 8).
Table 1. Retention time reproducibilities, regression equations, correlation coefficients (r2), F-test, p (F), linearity ranges, LOD, and LOQ of the compounds 417 (n = 8).
CompoundsRetention Time Reproducibility (min, n = 8)Regression Equationr2*F*p (F)Linearity Range (μg/mL)LOD (μg/mL)LOQ (μg/mL)
Acid
Lucidenic acid N (4)48.176 ± 0.110y = 20041.5x − 11913.80.999912.670.003147.50–180.000.882.64
Lucidenic acid E2 (5)56.463 ± 0.161y = 10679.7x − 4195.80.999912.740.003087.50–180.000.822.45
Ganoderic acid A (6) & Lucidenic acid A (7)74.075 ± 0.130y = 19900.9x − 11310.70.999912.770.003057.50–180.000.341.01
Ganoderic acid E (8)83.898 ± 0.240y = 5790.0x − 4699.30.999912.600.003207.50–180.001.173.52
Methyl lucidenate E2 (9)93.330 ± 0.083y = 6087.3x − 5267.70.999912.590.003217.50–180.000.912.74
Methyl lucidenate A (10)97.480 ± 0.080y = 31602.7x − 12081.80.999912.740.003087.50–180.000.682.04
Butyl lucidenate E2 (11) & Butyl ganoderate A (12)101.827 ± 0.116y = 9737.0x − 12899.60.999912.650.003167.50–180.001.103.31
Alcohol
Lucidadiol (13)104.729 ± 0.115y = 9573.6x − 3631.90.999912.740.003087.50–180.000.571.72
Ganodemanontriol (14)105.589 ± 0.213y = 34791.4x + 110902.30.999913.890.002267.50–180.001.394.18
Ganoderiol F (15)111.230 ± 0.245y = 7417.9x − 14493.50.999912.250.003537.50–180.001.414.23
Ganodermadiol (16)121.772 ± 0.258y = 11250.1x − 5594.50.999912.700.003117.50–180.001.374.12
Other
Ergosterol (17)128.915 ± 0.267y = 18821.0x − 5702.90.999912.770.003067.50–180.000.942.82
* F-test and p (F) values were analysed by one-way analysis of variance (ANOVA using the Origin V7.5 software (Originlab, Northampton, MA, USA). Statistical significance was assumed at below the 0.05 probability level.
Table 2. Recovery rates of the compounds 417 from extracts of G. lucidum.
Table 2. Recovery rates of the compounds 417 from extracts of G. lucidum.
CompoundsAdded (µg/mL)Detected (µg/mL)Recovery (%)M ± SD (%)RSD (%)
Acid
Lucidenic acid N (4)9.509.4799.6899.51 ± 1.481.49
19.0019.17100.89
38.0037.2297.95
Lucidenic acid E2 (5)10.2010.23100.2998.44 ± 1.611.64
20.4019.9297.65
40.8039.7397.38
Ganoderic acid A (6) & Lucidenic acid A (7)10.4010.2198.1798.71 ± 1.231.25
20.8020.3597.84
41.6041.65100.12
Ganoderic acid E (8)10.8010.5497.5999.20 ± 1.451.46
21.6021.69100.42
43.2043.0299.58
Methyl lucidenate E2 (9)10.0010.28102.80100.79 ± 1.741.73
20.0019.9599.75
40.0039.9399.83
Methyl lucidenate A (10)10.109.8297.2398.39 ± 1.761.79
20.2019.7097.52
40.4040.57100.42
Butyl lucidenate E2 (11) and Butyl ganoderate A (12)10.109.9398.3297.52 ± 1.952.00
20.2019.2595.30
40.4039.9798.94
Alcohol
Lucidadiol (13)10.5010.1296.3897.17 ± 2.282.35
21.0020.0395.38
42.0041.8999.74
Ganodemanontriol (14)10.2010.1299.2299.32 ± 1.271.27
20.4020.53100.64
40.8040.0398.11
Ganoderiol F (15)10.4010.0596.6397.09 ± 1.121.15
20.8020.4698.37
41.6040.0596.27
Ganodermadiol (16)10.009.9499.4099.45 ± 1.631.63
20.0020.22101.10
40.0039.1497.85
Other
Ergosterol (17)10.4010.0796.8397.70 ± 2.212.26
20.8019.9896.06
41.6041.69100.22
For the recovery, each reference compound was spiked into 1 g of each Ganoderma species at three levels, as described in the Experimental Section. The spiked samples were assayed, and the recoveries of each reference compounds were found to be 97.09% to 100.79%, and the relative standard deviation (RSD) (%) was less than 2.31% (Table 2). The average recovery was represented by the formula: R (%) = [(amount from the sample spiked standard − amount from the sample)/amount from the spiked standard] × 100. Table 3 shows the intra-day and inter-day precision (%RSD) of this HPLC method. The precision and accuracy ranged from 0.81%–3.20% and 95.38%–102.19% for intra-day and from 0.40%–3.67% and 95.63%–103.09% for inter-day, respectively. The data demonstrate that the method was acceptable in terms of linearity, accuracy, and reproducibility.

2.3. Quantitative Comparison of Different Ganoderma Species

The amount of the chemical compounds within the samples was influenced by various factors such as the place of origin, type of study sample (cultivated or wild samples; different species of the same genus), and harvesting season. The variation of the lanostane triterpenoid alcohol or acid derivatives and ergosterol in the different Ganoderma species originating from the wild and cultivated collections from Vietnam was evaluated. The fingerprint analysis of the wild Linhzhi group (Figure 2A) showed a similarity across the chromatograms, and the 17 analytes were present in all the samples. Figure 2B,C show the differences among the wild Linhzhi group (A group), the cultivated Linhzhi group (B group), and the related Linhzhi species group (C group). In particular, the chromatograms of the five related Linhzhi species including G. sp, G. applanatum, G. australe, G. colossum, and G. subresinosum showed obvious differences. Compounds 13 were not distinctly separated using the developed method, so they were not quantified. This study focused on the simultaneous determination of the remaining 14 compounds 417 by using the developed HPLC-DAD method for the all samples (Groups A, B and C), which are summarized in Table 4, Table 5 and Table 6, respectively. Each sample was analyzed in triplicate to ensure the reproducibility of the quantitative results. The comparison of the 14 compounds in the Linhzhi samples (both wild and cultivated samples) showed that the number of acids and their derivatives in all the samples is significantly higher than the number of alcohols. However, there are differences between the wild and cultivated samples. While the total amount of the acids and alcohols in the wild samples vary from 2089.40 to 44,703.07 μg/g and 917.41 to 2498.68 μg/g, respectively, those from the cultivated samples fluctuate between 1003.83 and 1720.69 μg/g and 153.31 to 549.32 μg/g, respectively. Similarly, the total amount of the acids in G. australe outweighs that of the wild and cultivated Linhzhi samples and other related Linhzhi species (Table 4, Table 5 and Table 6,) with a total amount of 19,999.28 μg/g. In addition, the amount of the 2 compounds 11 and 12 in the analyzed samples was below LOQ except for TGau, which had 131.29 ± 1.31 µg/g.
Table 3. Intraday and interday repeatability of 417.
Table 3. Intraday and interday repeatability of 417.
CompoundsNominal Conc. (µg/mL)Inter-DayIntra-Day
Observed Conc. (M ± SD), (μg/mL)Accuracy (%)Precision (%)Observed Conc. (M ± SD), (μg/mL)Accuracy (%)Precision (%)
Lucidenic acid N (4)30.2530.02 ± 0.1399.240.4330.17 ± 0.4299.741.39
60.5060.13 ± 2.1099.393.4961.02 ± 1.95100.863.20
121.00120.04 ± 2.2999.211.91120.11 ± 2.2599.261.87
Lucidenic acid E2 (5)29.7530.33 ± 0.12101.950.4030.12 ± 0.50101.241.66
59.5060.17 ± 0.97101.131.6158.27 ± 0.7997.931.36
119.00118.56 ± 1.9699.631.65118.39 ± 0.9699.490.81
Ganoderic acid A (6) & Lucidenic acid A (7)30.7530.28 ± 1.1198.473.6730.10 ± 0.5997.891.96
61.5060.00 ± 1.0597.561.7559.88 ± 0.8897.371.47
123.00122.12 ± 1.2799.281.04121.16 ± 2.0598.501.69
Ganoderic acid E (8)29.7529.59 ± 0.5999.461.9929.03 ± 0.7697.582.62
59.5059.27 ± 1.1299.611.8958.36 ± 0.9998.081.70
119.00118.33 ± 0.9599.440.80118.13±1.1399.270.96
Methyl lucidenate E2 (9)28.7527.94 ± 0.8497.183.0129.13 ± 0.89101.323.06
57.5058.24 ± 1.10101.291.8956.02 ± 0.9397.431.66
115.00116.05 ± 1.27100.911.09114.12 ± 2.1199.231.85
Methyl lucidenate A (10)31.7529.62 ± 0.6693.292.2330.59 ± 0.7396.352.39
63.5062.21 ± 0.9897.971.5862.58 ± 1.0698.551.69
127.00125.76 ± 0.7499.020.59126.29 ± 1.9499.441.54
Butyl lucidenate E2 (11) and Butyl ganoderate A (12)33.7529.45 ± 0.5887.261.9732.19 ± 0.8295.382.55
67.5066.54 ± 1.2098.581.8066.83 ± 1.0299.011.53
135.00133.50 ± 1.3198.890.98134.20 ± 2.0799.411.54
Lucidadiol (13)32.5030.08 ± 0.6992.552.2932.19 ± 0.8399.052.58
65.0063.89 ± 1.0198.291.5863.97 ± 1.3298.422.06
130.00128.95 ± 1.1799.190.91128.04 ± 1.9598.491.52
Ganodemanontriol (14)30.7529.85 ± 0.5897.071.9429.83 ± 0.5997.011.98
61.5061.59 ± 0.94100.151.5360.82 ± 0.8398.891.36
123.00122.11 ± 1.2799.281.04122.39 ± 1.9699.501.60
Ganoderiol F (15)29.2529.78 ± 0.55101.811.8529.89 ± 0.55102.191.84
58.5058.02 ± 1.0199.181.7458.01 ± 1.0399.161.78
117.00115.38 ± 1.7898.621.54116.12 ± 2.1199.251.82
Ganodermadiol (16)30.2530.05 ± 0.7399.342.4330.11 ± 0.4999.541.63
60.5060.02 ± 0.7999.211.3260.12 ± 1.3799.372.28
121.00120.11 ± 1.0299.260.85120.37 ± 2.0599.481.70
Ergosterol (17)29.7530.67 ± 0.63103.092.0530.17 ± 0.73101.412.42
59.5058.03 ± 0.9297.531.5959.20 ± 1.5299.502.57
119.00119.97 ± 1.12100.820.93118.79 ± 1.9999.821.68
Table 4. Contents of 417 in the wild collected Linhzhi samples (Group A).
Table 4. Contents of 417 in the wild collected Linhzhi samples (Group A).
CompoundsContent (µg/g) a
VN1VN12VN13VN16VN18VN21
Acid
Lucidenic acid N (4)388.16 ±5.10444.58 ± 3.84845.46 ± 9.89420.71 ± 2.40257.80 ± 3.88884.05 ± 7.45
Lucidenic acid E2 (5)776.90 ± 9.831766.75 ± 24.01319.47 ± 6.931151.08 ± 7.64430.79 ± 7.041695.01 ± 16.32
Ganoderic acid A (6) & Lucidenic acid A (7)228.88 ± 3.12401.93 ± 2.37349.93 ± 5.05140.97 ± 1.71891.05 ± 7.68509.76 ± 7.05
Ganoderic acid E (8)878.27 ± 7.751528.73 ± 12.88574.54 ± 4.71797.83 ± 18.762100.20 ± 13.11 b1614.25 ± 9.27
Methyl lucidenate E2 (9)
Methyl lucidenate A (10)46.62 ± 1.47
Butyl lucidenate E2 (11)
& Butyl ganoderate A (12)
Total acid2318.82 ± 6.244141.98 ± 22.152089.40 ± 24.292510.58 ± 29.773679.85 ± 21.664703.07 ± 38.72
Alcohol
Lucidadiol (13)144.44 ± 2.32212.81 ± 1.35111.31 ± 1.00128.84 ± 0.54243.95 ± 6.43219.25 ± 2.00
Ganodemanontriol (14)168.35 ± 2.53364.27 ± 3.33232.15 ± 1.82129.31 ± 0.95394.10 ± 5.78291.04 ± 2.44
Ganoderiol F (15)697.42 ± 13.75643.68 ± 11.49795.69 ± 5.07563.94 ± 2.861635.06 ± 13.63689.01 ± 2.02
Ganodermadiol (16)218.99 ± 4.14159.87 ± 0.9591.38 ± 1.0395.33 ± 0.60225.56 ± 0.49348.52 ± 4.90
Total alcol1229.20 ± 21.161380.63 ± 14.301230.54 ± 6.38917.41 ± 3.652498.68 ± 7.411547.83 ± 9.55
Other
Ergosterol (17)205.82 ± 1.72700.74 ± 4.76222.27 ± 4.93325.85 ± 1.87862.09 ± 12.71160.74 ± 4.56
(−): Lower than test limit and could not be quantified; a Data were expressed as mean ± SD of three experiments; b Out of linear range.
Table 5. Contents of 417 in the cultivated Linhzhi samples (Group B).
Table 5. Contents of 417 in the cultivated Linhzhi samples (Group B).
CompoundsContent (µg/g) a
GL1GL2GL3VN10
Acid
Lucidenic acid N (4)139.08 ± 1.8279.58 ± 1.4752.53 ± 1.06
Lucidenic acid E2 (5)481.31 ± 4.07420.91 ± 2.78357.89 ± 2.90258.06 ± 1.31
Ganoderic acid A (6) & Lucidenic acid A (7)83.35 ± 2.40232.55 ± 5.6670.17 ± 0.6676.19 ± 0.18
Ganoderic acid E (8)730.00 ± 4.2785.54 ± 0.89207.77 ± 1.79773.92 ± 4.75
Methyl lucidenate E2 (9)286.94 ± 2.33446.45 ± 8.63288.41 ± 1.73
Methyl lucidenate A (10)
Butyl lucidenate E2 (11) & Butyl ganoderate A (12)
Total acid1720.69 ± 9.231185.46 ± 3.011003.83 ± 5.021160.36 ± 7.87
Alcohol
Lucidadiol (13)69.15 ± 1.4049.43 ± 0.69109.00 ± 1.0050.47 ± 1.59
Ganodemanontriol (14)107.04 ± 3.32208.34 ± 2.6250.85 ± 0.80
Ganoderiol F (15)86.16 ± 1.2465.03 ± 0.71153.75 ± 0.92226.71 ± 1.29
Ganodermadiol (16)59.59 ± 0.6078.23 ± 0.6437.07 ± 0.41
Total alcol155.31 ± 1.38281.10 ± 4.06549.32 ± 3.44365.10 ± 0.93
Other
Ergosterol (17)135.14 ± 1.52194.68 ± 3.74795.96 ± 5.65647.79 ± 10.62
(−): Lower than test limit and could not be quantified; a Data were expressed as mean ± SD of three experiments.
Table 6. Contents of 4–17 in the related Linzhi samples (Group C).
Table 6. Contents of 4–17 in the related Linzhi samples (Group C).
CompoundsContent (µg/g) a
TGLsTGapTGauTGcTGs
Acid
Lucidenic acid N (4)144.18 ± 1.1563.13 ± 1.45207.73 ± 2.0557.50 ± 0.65
Lucidenic acid E2 (5)413.76 ± 2.04121.65 ± 4.50201.92 ± 2.45
Ganoderic acid A (6) & Lucidenic acid A (7)89.18 ± 0.9778.09 ± 2.97118.24 ± 3.09
Ganoderic acid E (8)475.23 ± 3.28114.89 ± 1.891213.14 ± 15.26337.62 ± 4.9151.10 ± 1.30
Methyl lucidenate E2 (9)2499.52 ± 17.6518,247.10 ± 48.40 b1023.84 ± 28.71
Methyl lucidenate A (10)144.89 ± 3.34
Butyl lucidenate E2 (11) & Butyl ganoderate A (12)65.65 ± 0.65
Total acid1119.26 ± 4.482613.80 ± 17.0819,999.28 ± 41.661889.35 ± 20.91108.60 ± 1.07
Alcohol
Lucidadiol (13)54.04 ± 1.3270.36 ± 0.0698.44 ± 1.1946.08 ± 0.78
Ganodemanontriol (14)72.99 ± 1.88
Ganoderiol F (15)84.61 ± 0.85126.42 ± 1.32272.73 ± 3.4956.58 ± 0.70
Ganodermadiol (16)47.55 ± 0.71
Total alcol138.65 ± 1.4870.36 ± 0.06224.86 ± 1.23439.35 ± 5.2256.58 ± 0.70
Other
Ergosterol (17)116.32 ± 1.59112.71 ± 0.52148.30 ± 2.63221.45 ± 2.49158.35 ± 1.92
(−): Lower than test limit and could not be quantified; a Data were expressed as mean ± SD of three experiments; b Out of linear range.
As shown in Table 4, Table 5, Table 6 and Figure 3, the total amount of all the compounds in the wild samples was appreciably higher than in the cultivated ones. For example, the amount of compound 4 (lucidenic acid N) in the wild G. lucidum sample varied from 257.80–845.46 μg/g; however, this compound was not observed in GL2, and in the other cultivated samples (Group C) it fluctuated between 52.53–139.08 μg/g. Another good example is lucidenic acid E2 (5), which was found in the wild G. lucidum samples in a range from 319.47 to 1,766.75 μg/g in comparison with the cultivated G. lucidum samples in a range from 258.06 to 481.31 μg/g. In addition, the wild samples contained significantly more ganoderiol F (15) than the cultivated samples, which varied between 563.94 μg/g and 1,635.06 μg/g and between 65.03 μg/g and 226.71 μg/g, respectively. Interestingly, a different trend was observed for methyl lucidenate E2 (9), which was under the LOQ in the wild samples but was found in GL1, GL2, and GL3 in the range between 286.94 and 446.95 μg/g. On the whole, the samples from Bac Giang (VN16 and VN18) seemed to have a higher amount of the constituents than the specimens from Quang Nam (VN1, VN12, and VN13). The total amount of constituents in the related Linhzhi samples (Group C) including in TGau, TGLs, TGap, TGc, and TGs was considerably lower than the amount in the Linhzhi samples of Groups A and B. The amount of constituents in TGau outweighed that of the others, as it was about 4 times as high as that of the wild Linhzhi samples (VN12).
Figure 3. Content of 417 in various Linhzhi samples belong to groups A, B and C.
Figure 3. Content of 417 in various Linhzhi samples belong to groups A, B and C.
Molecules 20 01059 g003
The proportion of constituents in the other species is different from G. lucidum. More specifically, the proportion of lucidenic acid E2 (5), which is one of the major compounds in G. lucidum, is low in TGap and TGS and is not found in TGau. Similarly, while almost G. lucidum samples contain a considerable amount of ganodemanontriol, it was only seen in trace amounts in the TGLs, TGap, TGau and TGs samples. In contrast, the amount of methyl lucidenate E2 (9), which is not observed in G. lucidum, is the major compound in the TGap, TGau, and TGc samples. It is noteworthy that the amount of the constituents in TGs was substantially smaller than others, and 50 percent was ergosterol.

2.4. Discussion

To date, several previous studies have reported using HPLC analytical methods for the analysis of Ganoderma lucidum and its related products. For example, Zhao et al. used HPLC for the determination of 9 triterpenes and sterols for the quality evaluation of G. lucidum [25]. In a study from Wang et al., an RP-HPLC method was developed for the determination of six ganoderic acids [22]. In 2004, Gao and coworkers reported the quantitative determination of 19 triterpene constituents, including six ganoderma alcohols and 13 ganoderic acids [11]. These studies and others focused only on the ganoderic acids and their derivatives [12,26]. Nucleosides, nucleobases, and polysaccharides were used for the qualitative and quantitative analyses of Ganoderma spp [27]. However, these studies are insufficient for a comprehensive evaluation of G. lucidum, and there is little data that compare G. lucidum from different origins or compare G. lucidum and its related species.
In this paper, we developed and optimized an HPLC-DAD method that allows for the specific identification of many terpenes. Fourteen triterpenes, including nine ganodermic acids 412, four alcohols 1316, and one sterol (ergosterol, 17), were used for the quantitative determination of G. lucidum and its related species. Eight of the nine ganoderma acids (all but ganoderic acid A) had never been analyzed before. Two new lanostane triterpenes 11 and 12, which recently were discovered by our group and Lee Iksoo et al. [24], were used in the fingerprint analysis and quantitative determination for the first time [19,23]. Alcohols 13 and 16, which had never been examined quantitatively in previous studies, were used to evaluate G. lucidum and its related species chemically using HPLC-DAD. Two Ganoderma species investigated in this study were studied quantitatively using HPLC for the first time, except for G. applanatum [28]. However, in a study by Liu et al., G. applanatum was evaluated by using five ganoderic acids. Moreover, two compounds, uracil and gentisic acid, which were found in G. lucidum for the first time, were confirmed using the HPLC fingerprint technique.
In comparison with the results of previous studies, this study showed both similarities and differences. In a study by Gao et al. [11], the amount of ganondermanontriol (14) in Japanese Linhzhi ranged widely from 19.2 to 235.3 μg/g. In our study, there was a wide variation of compound 14 in the wild, cultivated, and related Linhzhi samples ranging from 129.31 to 394.10 μg/g, 50.85 to 208.34 μg/g, and 72.99 ± 1.88 μg/g (related Linhzhi species, TGc), respectively. Therefore, the amount of compound 14 in the Japanese samples was similar to the Vietnamese cultivated samples but was lower than that found in the Vietnamese wild Linhzhi. These results indicate that the amount of compound 14 may not depend on geographic factors but instead is affected by the cultivation conditions. Gao’s study showed that the contents of ganoderiol F (15) ranged from 18.9 to 156.5 μg/g [11]. However, the Vietnamese wild samples contained 563.94–1635.06 μg/g of 15, and the Vietnamese cultivated samples contained 65.03–226.71 μg/g of 15. Both the wild and cultivated Linhzhi samples from Vietnam contain more compound 15 than the Linhzhi from Japan. Similar to the results found with compound 14, these results indicate that amount of 15 is not only affected by geographic factors but also by cultivation conditions. In a study from the Yuan group, the content of ergosterol (17) from sporoderm-broken germinating spores of Linhzhi varied from 32 μg/g to 1202 μg/g in the cultivated Linhzhi from China [21], corresponding with the results from this study, as the content of 17 in the cultivated Linhzhi ranged between 135.14 μg/g to 795.96 μg/g. With regard to methyl lucidenate E2 (9), there was a huge difference in the amounts found among the wild and cultivated Linhzhi and its related species. While a quantitative determination of 9 in the wild species could not be made, its content was above 288 μg/g in the cultivated species. Interestingly, compound 9 was the major compound in related species of Linhzhi as its content was at least 1023.84 μg/g across the species and was as high as 2499.52 μg/g in G. australe.

3. Experimental Section

3.1. Ganoderma Species

The wildly collected samples (Group A) (Ganoderma lucidum) were VN1, VN12; VN13 originating from Tienphuoc, Quangnam Province; VN16 and VN18 originating from Bacgiang Province; and VN21 originating from Trami, Quangnam Province. The cultivated samples (Group B) (Ganoderma lucidum) were VN10 from Vietnam Academy of Agricultural Sciences, GL1-GL3 from Linh chi Vina company (Ho Chi Minh, Vietnam). The other species of Ganoderma genus (Group C) including G. applanatum (TGap), G. colossum (TGc), G. subresinosum (TGs), Ganoderma sp (TGls), and G. australe (TGau) were gifts from Linh chi Vina Company. The samples were botanically identified by Msc. Co Duc Trong, Linh chi Vina Company, where the voucher specimens were deposited. The other specimens of the test samples have been verified by KiHwan Bae from College of Pharmacy, Chungnam National University, Korea and deposited in the Department of Phytochemistry, NIMM, Vietnam. All dried samples were ground and transferred to the laboratory for preparation of the plant extracts.

3.2. Standard Compounds

The compounds 117 were isolated from the chloroform- and water-soluble fractions of the methanol extracts from fruiting bodies of Ganoderma lucidum, which were published previously by our group including a new lanostane triterpene butyl lucidenate E2 (11) and adenosine (2) and two compounds isolated from the first time in G. lucidum, namely uracil (2) and gentisic acid (3), 12 lanostane steroids including lucidenic acid N (4), lucidenic acid E2 (5), ganoderic acid A (6), lucidenic acid A (7), ganoderic acid E (8), methyl lucidenate E2 (9), methyl lucidenate A (10), butyl ganoderate A (12), lucidadiol (13), ganodermanontriol (14), ganoderiol F (15), ganodermadiol (16), and ergosterol (17) [19,20,24] (Figure 1). All chemical standards were isolated from G. lucidum, theirs purities were checked by the HPLC-DAD method and the chemical structures were elucidated by 1H-NMR, IR, UV/VIS spectroscopic analysis and MS spectral data.

3.3. Chemicals and Reagents

Acetonitrile and methanol (MeOH) of analytical HPLC grade was purchased from Merck (Darmstadt, Germany). Phosphoric acid of analytical reagent grade was obtained from Sigma-Aldrich (St Louis, MO, USA). The other organic solvents and other chemical reagents were of analytical reagent grade.

3.4. Reference Compound Preparation

To determine the content of fourteen markers (compounds 417) of Linhzhi and related Linhzhi samples, the dried powders were used for extraction. The same amounts (about 1 g) of pulverized fruiting bodies were weighed and sieved through 50 mesh and then placed into a volumetric flask, methanol (10 mL) was added, the weight was accurately measured and the samples were ultrasonically extracted for 90 min at 50 °C. The solution was cooled, weighed again, and made up the loss in weight with methanol. The solution was filtered through 0.45 µm membrane filter prior to HPLC analysis.

3.5. HPLC

Analytical HPLC was carried out on a LC 20A system (Shimadzu, city, Japan) consisting of a LC-20AD quaternary gradient pump, an autosampler, and a SPD-M20A diode array detector, connected to a LC solution singer ver. 1.25 software. A Zorbax XDB C18 (4.6 × 250 mm, 5 µm, Agilent Technologies, Inc., Santa Clara, California, CA, USA) was used. A binary gradient solution system consisted of 0.1% phosphoric acid in water (A) and acetonitrile (B) and separation was achieved using the following gradient program: 0 min, 4% B; 10 min, 11% B; 15 min, 30% B; 60 min, 45% B; 90 min, 85% B; 110 min, 100 B%; 130–140 min, 100% B; and finally, reconditioning the column with 4% B isocratic for 10 min. The flow rate was 0.5 mL/min, the system operated at 40 °C and the detection wavelengths were set at 243 and 256 nm for ganoderma alcohols and acids, respectively.

3.6. Method Validation

Every standard compound was accurately weighed and dissolved in 100% MeOH to prepare a stock solution of 1.0 mg/mL concentration. Working standard solutions of ganoderma alcohols and acids were prepared by repeated dilution to give eight respective concentrations with methanol (7.5–180 µg/mL). Eight concentrations of 14 analyses were injected in triplicate, and then the calibration curves were constructed by plotting the peak areas versus the concentrations of each analysis. The linearity was demonstrated by a correlation coefficient (r2) greater than 0.999. The limit of detection (LOD) and the limit of quantification (LOQ) were determined based on signal-to noise ratios (S/N) of 3:1 and 10:1, respectively. Intra- and inter-day variations were chosen to determine the precisions of the developed method. The relative standard deviation (RSD) was taken as a measure of precision. Intra- and inter-day repeatability was determined on five times within one day and five separate days, respectively. The recovery tests were prepared by mixing a powdered sample (1 g) with three concentration levels (25%, 50%, and 100%) of each compound. The mixture was then extracted by following the section of preparation of sample solution for HPLC analysis. The extract solutions were filtered through a 0.45 µM membrane. The HPLC-DAD analysis experiments were performed in triplicate for each control level. Precision were determined by multiple analysis (n = 5) of quality control samples. All samples were then subjected to HPLC analysis to calculate the recovery rates.

3.7. Statistical Analysis

The data were analyzed using the unpaired Student’s t-test between the control and compounds. Data compiled from three independent experiments and values are expressed as mean ±SD.

4. Conclusions

This is the first time a HPLC-DAD method for quantitative analysis of constituents in Ganoderma lucidum and its related species batches originating from Vietnam was established. In the present work, we have reported for the first time the presence of lanostane triterpenes, ergosterol, uracil, adenosine, and gentisic acids in the Vietnamese G. lucidum and its related species. Especially, two new lanostanes, butyl lucidenate E2 and butyl ganoderate A, were reported for the first time in G. lucidum originating from Vietnam and its four related species using a HPLC-DAD method. In addition, the highest content of methyl lucidenate E2 was found in G. australe, G. applatatum, and G. colossum, respectively. In the present study the profile of the 17 compounds differed significantly in the all analyzed samples. It can be also concluded that the geographical distributions, growth conditions, and substrates might be the key to differences in producing chemical compositions. This present work suggested an accurate and sufficient method for quantitative evaluation, which is suitable for quality evaluation of Ganoderma products.

Supplementary Materials

Supplementary materials can be accessed at: https://www.mdpi.com/1420-3049/20/01/1059/s1.

Acknowledgments

This work was supported by a grant from Vietnam National Foundation for Science and Technology Development (NAFOSTED: 106.99-2011.54). We are grateful to Co Duc Trong for supporting of the cultivated Linhzhi and related Linhzi samples.

Author Contributions

D.T.H. and B.S.M. conceived and designed the research; L.T.L., T.M.H., L.V.N.H., N.M.K., N.P.D.N., and L.V.D. performed the research and analyzed the data; D.T.H. and B.S.M. wrote the manuscript. All the authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Long, S.C.; Birmingham, J.M. Medicinal benefits of the mushroom Ganoderma. Adv. Appl. Microbiol. 1992, 37, 101–134. [Google Scholar] [PubMed]
  2. Boh, B.; Berovic, M.; Zhang, J.; Zhi-Bin, L. Ganoderma lucidum and its pharmaceutically active compounds. Biotech. Ann. Rev. 2007, 13, 265–301. [Google Scholar]
  3. Kim, H.W.; Kim, B.K. Biomedicinal triterpenoids of Ganoderma lucidum (Curt.: Fr.) P. Karst (aphyllophoromycetidae). Intern. J. Med. Mushrooms 1999, 1, 121–138. [Google Scholar] [CrossRef]
  4. El-Mekkawy, S.; Meselhy, M.R.; Nakamura, N.; Tezuka, Y.; Hattori, M.; Kakiuchi, N.; Shimotohno, K.; Kawahata, T.; Otake, T. Anti-HIV-1 and anti-HIV-1-protease substances from Ganoderma lucidum. Phytochemistry 1998, 49, 1651–1657. [Google Scholar] [CrossRef] [PubMed]
  5. Kodora, Y.; Shimizu, M.; Sonoda, Y.; Sato, Y. Ganoderic acid and its derivatives as cholesterol synthesis inhibitors. Chem. Pharm. Bull. 1990, 38, 1359–1364. [Google Scholar] [CrossRef] [PubMed]
  6. Koyama, K.; Akiba, M.; Imaizumi, T.; Kinoshita, K.; Takahashi, K.; Suzuki, A.; Yano, S.; Horie, S.; Watanabe, K. Antinociceptive constituents of Auricularia polytricha. Planta Med. 2002, 68, 284–285. [Google Scholar] [CrossRef] [PubMed]
  7. Ha, D.T.; Oh, J.; Khoi, N.M.; Dao, T.T.; Dung, L.V.; Do, T.N.Q.; Lee, S.M.; Jang, T.S.; Jeong, G.S.; Na, M. In vitro and in vivo hepatoprotective effect of ganodermanontriol against t-BHP-induced oxidative stress. J. Ethnopharmacol. 2013, 150, 875–885. [Google Scholar] [CrossRef] [PubMed]
  8. Andoh, T.; Zhang, Q.; Yamamoto, T.; Tayama, M.; Hattori, M.; Tanaka, K.; Kuraishi, Y. Inhibitory effects of the methanol extract of Ganoderma lucidum on mosquito allergy-induced itch-associated responses in mice. J. Pharmacol. Sci. 2010, 114, 292–297. [Google Scholar] [CrossRef] [PubMed]
  9. Lindequist, U.; Niedermeyer, T.H.; Jülich, W.D. The pharmacological potential of mushrooms. eCAM 2005, 2, 285–299. [Google Scholar] [PubMed]
  10. Ma, J.-Q.; Liu, C.-M.; Qin, Z.-H.; Jiang, J.-H.; Sun, Y.-Z. Ganoderma applanatum terpenes protect mouse liver against benzo(α)pyren-induced oxidative stress and inflammation. Environ. Toxicol. Pharmacol. 2011, 31, 460–468. [Google Scholar] [CrossRef] [PubMed]
  11. Gao, J.-J.; Nakamura, N.; Min, B.-S.; Hirakawa, A.; Zuo, F.; Hattori, M. Quantitative determination of bitter principles in specimens of Ganoderma lucidum using high-performance liquid chromatography and its application to the evaluation of ganoderma products. Chem. Pharm. Bull. 2004, 52, 688–695. [Google Scholar] [CrossRef] [PubMed]
  12. Keypour, S.; Rafati, H.; Riahi, H.; Mirzajani, F.; Fata Moradali, M. Qualitative analysis of ganoderic acids in Ganoderma lucidum from iran and china by rp-hplc and electrospray ionisation-mass spectrometry (ESI-MS). Food Chem. 2010, 119, 1704–1708. [Google Scholar] [CrossRef]
  13. Ruan, W.; Popovich, D.G. Ganoderma lucidum triterpenoid extract induces apoptosis in human colon carcinoma cells (Caco-2). Biomed. Prev. Nutr. 2012, 2, 203–209. [Google Scholar] [CrossRef]
  14. Chen, Y.; Yan, Y.; Xie, M.-Y.; Nie, S.-P.; Liu, W.; Gong, X.-F.; Wang, Y.-X. Development of a chromatographic fingerprint for the chloroform extracts of Ganoderma lucidum by hplc and LC–MS. J. Pharm. Biomed. Anal. 2008, 47, 469–477. [Google Scholar] [CrossRef] [PubMed]
  15. Joseph, S.; Sabulal, B.; George, V.; Smina, T.P.; Janardhanan, K.K. Antioxidative and anti-inflammatory activities of the chloroform extract of Ganoderma lucidum found in south india. Sci. Pharm. 2009, 77, 111–121. [Google Scholar] [CrossRef]
  16. Salteralli, R.; Ceccaroli, P.; Lotti, M.; Zambonelli, A.; Buffalini, M.; Casadei, L. Biochemical characterization and antioxidant activity of mycelium of Ganoderma lucidum from central italy. Food Chem. 2009, 116, 143–151. [Google Scholar] [CrossRef]
  17. Nguyen, P.D.N.; Do, H.T.; Le, B.D. Characteristics of ecological factors and their distribution of Ganodermataceae Donk. in highlands of vietnam. J. Biol. 2013, 35, 198–205. [Google Scholar]
  18. Dam, N.; Nguyen, G.C.; Nguyen, B.; Trinh, T.K. Ganoderma species in vietnam. Hanoi-J. Med. Mat. 1997, 2, 10–13. [Google Scholar]
  19. Tung, N.T.; Cuong, T.D.; Hung, T.M.; Lee, J.H.; Woo, M.H.; Choi, J.S.; Kim, J.; Ryu, S.H.; Min, B.S. Inhibitory effect on no production of triterpenes from the fruiting bodies of Ganoderma lucidum. Bioorg. Med. Chem. Lett. 2013, 23, 1428–1432. [Google Scholar] [CrossRef] [PubMed]
  20. Ha, D.T.; Thu, N.T.; Khoi, N.M. Derivatives of nucleic and benzoic acids isolated from Ganoderma lucidum collected in quangnam-danang, vietnam. Hanoi-J. Med. Mat. 2014, 19, 216–219. [Google Scholar]
  21. Yuan, J.-P.; Wang, J.-H.; Liu, X.; Kuang, H.-C.; Huang, X.-N. Determination of ergosterol in ganoderma spore lipid from the germinating spores of Ganoderma lucidum by high-performance liquid chromatography. J. Agric. Food Chem. 2006, 54, 6172–6176. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, X.M.; Yang, M.; Guan, S.H.; Liu, R.X.; Xia, J.M.; Bi, K.S.; Guo, D.A. Quantitative determination of six major triterpenoids in Ganoderma lucidum and related species by high performance liquid chromatography. J. Pharm. Biomed. Anal. 2006, 41, 838–844. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, I.; Seo, J.; Kim, J.; Kim, H.; Youn, U.; Lee, J.; Jung, H.; Na, M.; Hattori, M.; Min, B.; et al. Lanostane triterpenes from the fruiting bodies of Ganoderma lucidum and their inhibitory effects on adipocyte differentiation in 3T3-L1 cells. J. Nat. Prod. 2010, 73, 172–176. [Google Scholar] [CrossRef] [PubMed]
  24. Ha, D.T.; Thuy, P.T.; Khoi, N.M. Ergostane and lanostane steroids from fruiting body of Ganoderma lucidum collected in quangnam, danang. Hanoi-J. Med. Mat. 2013, 18, 389–394. [Google Scholar]
  25. Zhao, J.; Zhang, X.Q.; Li, S.P.; Yang, F.Q.; Wang, Y.T.; Ye, W.C. Quality evaluation of Ganoderma through simultaneous determination of nine triterpenes and sterols using pressurized liquid extraction and high performance liquid chromatography. J. Sep. Sci. 2006, 29, 2609–2615. [Google Scholar] [CrossRef] [PubMed]
  26. Lu, J.; Qin, J.-Z.; Chen, P.; Chen, X.; Zhang, Y.-Z.; Zhao, S.-J. Quality difference study of six varieties of Ganoderma lucidum with different origins. Front. Pharmacol. 2012, 3, 57. [Google Scholar] [PubMed]
  27. Gao, J.L.; Leung, K.S.Y.; Wang, Y.T.; Lai, C.M.; Li, S.P.; Hu, L.F.; Lu, G.H.; Jiang, Z.H.; Yu, Z.L. Qualitative and quantitative analyses of nucleosides and nucleobases in Ganoderma spp. By HPLC-DAD-MS. J. Pharm. Biomed. Anal. 2007, 44, 807–811. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, Y.; Liu, Q.; Qiu, F.; Di, X. Sensitive and selective liquid chromatography-tandem mass spectrometry method for the determination of five ganoderic acids in Ganoderma lucidum and its related species. J. Pharm. Biomed. Anal. 2011, 54, 717–721. [Google Scholar] [CrossRef] [PubMed]
  • Sample Availability: Samples of the compounds are available from the authors.

Share and Cite

MDPI and ACS Style

Ha, D.T.; Loan, L.T.; Hung, T.M.; Han, L.V.N.; Khoi, N.M.; Dung, L.V.; Min, B.S.; Nguyen, N.P.D. An Improved HPLC-DAD Method for Quantitative Comparisons of Triterpenes in Ganoderma lucidum and Its Five Related Species Originating from Vietnam. Molecules 2015, 20, 1059-1077. https://doi.org/10.3390/molecules20011059

AMA Style

Ha DT, Loan LT, Hung TM, Han LVN, Khoi NM, Dung LV, Min BS, Nguyen NPD. An Improved HPLC-DAD Method for Quantitative Comparisons of Triterpenes in Ganoderma lucidum and Its Five Related Species Originating from Vietnam. Molecules. 2015; 20(1):1059-1077. https://doi.org/10.3390/molecules20011059

Chicago/Turabian Style

Ha, Do Thi, Le Thi Loan, Tran Manh Hung, Le Vu Ngoc Han, Nguyen Minh Khoi, Le Viet Dung, Byung Sun Min, and Nguyen Phuong Dai Nguyen. 2015. "An Improved HPLC-DAD Method for Quantitative Comparisons of Triterpenes in Ganoderma lucidum and Its Five Related Species Originating from Vietnam" Molecules 20, no. 1: 1059-1077. https://doi.org/10.3390/molecules20011059

Article Metrics

Back to TopTop