Next Article in Journal
Recent Advances on the Production of Itaconic Acid via the Fermentation and Metabolic Engineering
Previous Article in Journal
Thermodynamic Equilibrium Study of Anaerobic Digestion through Helmholtz Equation of State
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Method for Improving Microbial Conversion of Diosgenin and Separation and Identification of the Product

1
College of Life Science, Northwest A & F University, Yangling 712100, China
2
College of Chemistry and Pharmacy, Northwest A & F University, Yangling 712100, China
3
Shanxi Xifeng Wine Co., Ltd., Fengxiang County, Baoji 721406, China
4
College of Grassland Agriculture, Northwest A & F University, Yangling 712100, China
*
Authors to whom correspondence should be addressed.
These authors have contributed equally to this work.
Fermentation 2023, 9(1), 70; https://doi.org/10.3390/fermentation9010070
Submission received: 8 November 2022 / Revised: 4 January 2023 / Accepted: 5 January 2023 / Published: 13 January 2023
(This article belongs to the Section Microbial Metabolism, Physiology & Genetics)

Abstract

:
Diosgenin, a hydrolysis product from Dioscorea plants, can be used as a precursor of steroid drugs (e.g., progesterone, testosterone, and glucocorticoid). However, traditional acid hydrolysis production wastes water and causes severe environmental pollution. The extraction of diosgenin through microbial transformation is the most green and environmentally friendly method at present. In order to improve the efficiency of the extraction of diosgenin through microbial transformation, we proposed a new method of strain mutagenesis. After mutagenesis, the response surface methodology was used to optimize the solid-state fermentation medium, thereby improving the diosgenin yield. We found that the optimal formulation was 5.5% sucrose, 0.6% NH4H2PO4, and 26.6% wheat bran. The final extraction rate of diosgenin reached 0.439% (the value of diosgenin per g. of starting plant dry material). Compared with 0.338% before optimization, it had increased 1.29 times. Furthermore, two other compounds were isolated from the fermentation products. These were identified as diosgenone (C27H41O3) and yuccagenone (C27H42O3). Traditional diosgenone is obtained through the oxidation of diosgenin with oxalic acid, but the method in this study is directly obtained from Dioscorea rhizome powder. The price of Dioscorea rhizome powder is much lower than diosgenin, thus greatly reducing the cost of obtaining diosgenone. This method provides a basis for subsequent research on other pharmacological compounds.

1. Introduction

Diosgenin, a C27 spirostan steroid obtained from Dioscorea plants, is a derivative of steroidal sapogenin. It was discovered in Dioscoreaceae plants by the Japanese scientists Matsukawa and Fujii in 1935.
Diosgenin has a variety of biological activities [1]. Sun et al. (2020) found that diosgenin can increase the concentration of Ca2+ through the release and inflow of Ca2+, while it leads to the unrelated death of Ca2+ in PC3 cells, revealing the potential of using diosgenin for human prostate cancer treatment [2]. Sirotkin et al. (2019) tested the direct effects of plant steroidal saponins on basic ovarian cell function in livestock and explored its promotion of ovarian cell function, potentially improving female reproduction [3]. Yang et al. (2021) added diosgenin to yam and fed it to mice. This enhanced the memory of the mice and promoted neurite outgrowth in their neurons [4]. Li et al. (2018) injected lipopolysaccharide into mice to induce motor dysfunction, stimulate inflammation and an oxidation irritable reaction, and activate the TLR/NF-κB pathway in vitro and in vivo [5]. They found that diosgenin could effectively alleviate the LPS-induced response by inhibiting the TLR/NF-κB signal pathway, curing LPS-induced rat Parkinson’s disease (PD) to some extent.
Diosgenin is the synthetic precursor of steroid drugs (e.g., progesterone, testosterone, and glucocorticoid) widely used in clinical practice and it has important industrial application value [6]. However, it mainly exists as glycosides in plants such as turmeric [7], and inversing glycoside is the main strategy used in the extraction of diosgenin [8]. In the traditional industry, direct acid hydrolysis, including hydrochloride acid ethanol extraction and sulfuric acid ethanol extraction, is often used. However, this method requires high volumes of water and results in severe environmental pollution [9]. In order to explore an environmentally friendly extraction method of diosgenin, Shen et al. (2021) proposed the use of phosphorus tailings to prepare solid acid and convert Dioscorea zingiberensis C.H. Wright (DZW) to diosgenin [10]. Zhao et al. (2022) developed and synthesized a new solid acid based on TiO2 as an effective solid acid catalyst by sulfonating the prepared TiO2, which can be used to extract diosgenin from saponin [11]. Although the methods mentioned above solve the problem of the severe environmental harm caused by the large-scale application of sulfuric acid, they have several disadvantages. For example, the concentration of diosgenin extracted from steroidal saponins is low and still involves the production of pollutants. Cheng et al. (2021) developed and optimized a method of homo-enzyme catalysis to produce diosgenin at yields of up to 96.5%. This method’s diosgenin yield is high, with low by-product generation [12]. However, the cost is very high, bringing a new challenge to the industrial production of diosgenin.
Therefore, the purpose of this study is to improve the yield of diosgenin through the mutagenesis of endophytic fungus SYfx213.2 using UV-lithium chloride combined mutagenesis and the optimization of the fermentation culture conditions. In our study, the environmental pollution caused by traditional acid hydrolysis was eliminated, and the steroidal saponins were fully transformed using microorganisms, which reduced the production of by-products and reduced the production cost.

2. Experimental

2.1. Identification of Fungal Strains

In our research, we isolated the SYfx213.2 strain (Preservation number: CGMCC 11517), which can efficiently convert diosgenin from Dioscorea obtained from National Engineering Research Center for Phytochemistry. This strain is an endophytic fungus isolated from Dioscoreaceae. The strain was identified by sequencing the ribosomal ITS sequence. Genomic DNA of SYfx213.2 strain was isolated with DNA extraction kit (Solarbio Science & Technology Co., Ltd., Beijing, China) and used as PCR amplification template. The forward primer ITS1 and reverse primer ITS4 were utilized. The used forward (ITS1) primer was 5′-TCCGTAGGTGAACCTGCGG-3′, and the reverse (ITS4) primer was 5′-TCCTCCGCTTATTGATATGC-3′. Then, 10 μL mix enzymes (Tsingke Biotechnology Co., Ltd., Beijing, China) were mixed with 0.5 μL 10 μmol/L ITS1 primers, 0.5 μL 10 μmol/L ITS4 primer, 5 μL DNA templates, and 4 μL ddH2O. ITS rDNA sequences were amplified using PCR. PCR was performed as follows: (1) the initial denaturation temperature was 94 °C for 5 min; (2) 30 cycles, each cycle 94 °C for 30 s, 55 °C for 30 s, 72 °C for 1 min; (3) the terminal extension step was kept at 72 °C for 5 min [13]. PCR products were sent to China Beijing Aoko Biotechnology Co., Ltd. and purified for sequencing. The ITS sequences obtained were compared in NCBI Blast database to determine the species. The morphology and structure of the colonies were analyzed using scanning electron microscopy (No. 2021252903, Hitachi High-tech Company, Tokyo, Japan) and optical microscopy (NIKON, Tokyo, Japan).

2.2. Preparation of Spore Solution

The stored SYfx213.2 was removed from the refrigerator at −80 °C and inoculated into PDA medium by spreading plate method, then put into an incubator. The activated SYfx213.2 plate was taken from the incubator, and the spores in the plate were washed off with sterile water in the sterile operating table. The spores were placed in a conical flask with glass beads, fully shaken, and scattered. Then the single spore suspension was obtained using filtration with a syringe stuffed with cotton. The concentration of spores was adjusted to 1 × 106 cfu/mL, and it was added to the fermentation medium under the condition of 5% inoculation amount [14]. The germination state of spores was observed every two hours, and the spore solution was taken for the next experiment.

2.3. UV-LiCl Combined Mutagenesis Method

From the UV mutagenic lethal curves and LiCl mutagenic lethal curves, we obtained the respective lethality condition of 80% (materials), as shown in Table 1. We did crossover experiments with compound mutagenesis to obtain a lethality of 80% for the compound conditions for the next experiment. The spore solution was mutagenized using compound conditions of UV irradiation time under light-protected conditions. After the mutagenesis was completed, the indoor fluorescent light was turned on and the single spores were restored using light for 30 min. Then the fluorescent light was turned off again and UV irradiation was maintained, re-exposing them to an additional 2 min under dark conditions. The mutagenized spore solution was diluted to 1 × 102 cfu/mL with distilled water, and 0.1 mL was taken and coated on the activation medium with the suitable LiCl concentration. Single spores untreated with UV and lithium chloride were used as controls. Cultures were incubated at 33 °C in the dark for 3–5 days.

2.4. Preparation of Fermented Seed Liquid

The seed medium liquid (5 g sucrose, 1 g beef extract-peptone, 7 g Dioscorea root powder, 0.75 g K2HPO4, 0.25 g MgSO4, 0.25 g KCl) was prepared in 1 L distilled water and distributed into ten 250 mL conical flasks. The flasks were closed with eight layers of gauze and then sterilized through autoclaving for 20 min. Sterilized sterile water was drawn into the activated plate (Potato Dextrose Agar Medium), sterilized glass beads were placed on the plate, and the plate was shaken for 1–2 min to elute the spores. The mycelium was filtered out, and the suspension was diluted to a final spore concentration of 107 unit/mL. After the seed media cooled to room temperature, 5 mL of the suspension was inoculated into 100 mL media. The flasks were covered with gauze and incubated at 33 °C and 140 rpm agitation for 24 h.

2.5. Preparation of Solid-State Medium

The raw solid-state fermentation medium (80 g Dioscorea root powder, 3 g yeast extract powder, and 20 mL distilled water) was mixed with the seed liquid culture for 24 h and then inoculated with 50% of the inoculum, stirred evenly, and transferred to a 100-mesh screen. After the transfer, the medium was spread evenly on the screen, incubated in a constant temperature incubator for fermentation at 33 °C, and stirred once a day. The fermentation product was obtained after 10 d of fermentation [15].

2.6. Selection of Significant Factors by the Plackett–Burman Design

The Plackett-Burman design (PBD) was used to screen the variables that significantly affected the transformation of diosgenin by the fungus [16]. A single factor test was performed to determine the low and high levels of each variable (Supplementary Materials), and then PBD (Design Expert v8.0.6) was used to evaluate the seven factors [17]. The contents include (Table 2) sucrose (%), NH4H2PO4 (%), MgSO4 (%), Tween (g/L), Rice Husk (%), KH2PO4 (%), and Wheat Bran (%), which are represented as A, B, C, D, E, F, and G. Diosgenin yield (%) was determined by calculating the average of three replicates of independent measurements. The significant variables obtained by this method are used for further optimization of biological processes.

2.7. Levels of Each Factor Tested in the Box-Behnken

After determining the main influencing factors using PBD, Box-Behnken (BBD) analysis was carried out to obtain the significant influence on the biotransformation of DZW and the interaction between the selected factors, and the optimal value of each variable significantly affecting the yield of diosgenin was determined, so as to obtain the highest yield of diosgenin [18].

2.8. Chromatographic Purification

Chromatographic silica gel was loaded into the column using the wet loading method. The crude diosgenin obtained through extraction was ground and pulverized with liquid nitrogen, and 1.6 g was dissolved in 10–15 mL ethylacetate and added to 5 g of silica gel powder. The sample was stirred and incubated in a 50 °C water bath until the ethyl acetate evaporated. The sample was loaded onto the normal phase column, the eluent (light petroleum) was added, and the effluent liquid was collected. When the first substance was detected by using thin-layer chromatography, it was collected in a test tube. The same compounds were combined, and the liquid was dried with a rotary evaporator.

2.9. Thin Layer Chromatography

The samples collected in Section 2.4 were applied to a TLC plate at 1 cm intervals using a 0.3 mm glass capillary spotter. The TLC plate was placed vertically in the developing solvent tank containing cyclohexane and ethyl acetate at a 4:1 ratio. When the liquid level was 1 cm from the top edge, it was removed and dried. Subsequently, it was immersed in 10% sulfuric acid ethanol developer for 2 s and dried.

2.10. High Performance Liquid Chromatography (HPLC)

The solid fermentation product was dried in an oven at 60 °C. The dried products (4 g) were added to 20 mL petroleum ether in 50 mL centrifuge tubes and extracted with ultrasonic waves for 30 min [19]. The products were shaken for three hours at 180 rpm. The supernatant was collected into a flask and dried using a rotary evaporator, and the above extraction steps were repeated three times. The flask contents were washed with CHCl3 and diluted to 10 mL in a centrifuge tube. Next, 2 mL of liquid were transferred into a 2 mL centrifuge tube, evaporated, then dissolved with 2 mL of chromatographic grade methanol. The liquid passed through an organic filter with a pore diameter of 0.45 microns. Then, 1 mL was aspirated into the HPLC vial using a 1 mL syringe as the completed preparation sample. The concentrations of diosgenin standards were 0.125, 0.250, 0.500, 1.000, and 2.000 g/L. The HPLC column (XTERRA MS C18, 4.6 × 250 mm, 5 um; Waters; MA, USA) was used. The column temperature was 25 °C. The wave length of UV detector was 210 nm. Loading quantity of sample was 10 uL. Acetonitrile: water = 8:2 was the eluent (1.0 mL/min). The yield of diosgenin was calculated with the following Equation:
diosgenin yield (%) = diosgenin production (mg/mL)/DZW (mg/mL) × 100%

2.11. Nuclear Magnetic Resonance Method

Diosgenin standard and sample (30 mg each) were added to nuclear magnetic resonance tubes with a diameter of 5 mm and dissolved in 0.6 mL of chloroform-d using sonication [20]. 1H-NMR, 13C-NMR, and distortionless enhancement by polarization transfer (DEPT) were carried out on a nuclear magnetic resonance spectrometer with tetramethylsilane (TMS) as the standard material [21].

2.12. High Resolution Mass Spectrometry (HR-MS)

The samples were analyzed using mass spectrometry [22]. Next, 2 mg of purified sample was dried in a 5 mL centrifuge tube, and 1 mL of chromatographic grade methanol was added, sonicated until the sample was completely dissolved, and filtered through a 0.22 μm organic filter. The mass spectrometry conditions were as follows: EMS ion source was used with a mass-to-charge ratio (m/z) of 300–500 Da, positive ion mode, spray gas and auxiliary gas of 50 psi, curtain gas of 35 psi, ion source temperature of 550 °C, and spray voltage of 5500 V.

3. Results and Discussion

3.1. The Species Identification of the Target Strain

The optical microscopy and scanning electron microscopy of SYfx213.2 are shown in Figure 1. The mycelium was staggered, the diameter of the conidial pedunculum was about 8 μm, and the diameter of the thin-walled rough conidial head was about 35 um. After microscopy, we performed biological identification. Like Baquiao et al., we used ITS1 and ITS4 as primers for the amplification of specific gene fragments [23]. The obtained gene sequences (Supplementary Materials) were compared in the NCBI blast database, and gene sequence alignment analysis revealed that SYfx213.2 (MZ618696.1) is Aspergillus flavus [24]. It had been preserved in China General Microbiological Culture Collection Center (CGMCC), preservation number: 11517. Its evolutionary tree is detailed in Figure 2.

3.2. Screening of High Yield Diosgenin Strains by Compound Mutagenesis

Feng et al. screened for strains with high protease production using the UV mutagenesis of Bacillus subtilis [25]. Wang et al. combined Co-60 γ irradiation with LiCl mutagenesis to obtain strains with high Nosiheptide production [26], and our study combined UV mutagenesis and LiCl mutagenesis to screen for strains with high diosgenin production. It can be seen in Table 3 that the fatality rate is 80% when the UV irradiation time is 180 s and the dosage of lithium chloride is 2.0%. Therefore, this condition will be selected in subsequent experiments to carry out compound mutagenesis on SYfx213.2. After the mutagenesis, the strains were fermented and cultured, and the saponin yield was detected using HPLC. The test results are shown in Figure 3a.
According to the figure, all the samples in this batch produced saponin, among which the saponin spots of sample No. 11 were the largest and brightest, and it could be preliminarily determined that strain No. 11 had the highest saponin yield [27]. From Figure 3b, using the bar chart to compare the saponin yield after the fermentation of the screened strains, it can be concluded that, among them, the saponin yield of No. 11 was the highest, at 58.15 mg/L, while the saponin yields of No. 15, 8, 6, 23, 38, and 42 decreased successively, which were 54.02 mg/L, 53.34 mg/L, 51.43 mg/L, 50.6 mg/L, 47.47 mg/L, and 43.68 mg/L, respectively. The saponin production of these seven strains was higher than that of the control group (39.56 mg/L). The yield of the other strains was lower than that of the control group. After UV-LiCl compound mutagenesis, we evaluated the No. 11. strain, which we named as 11#SYfx213.2, to have the highest yield of diosgenin. Ten consecutive passages were made and fermentation was performed, and it was found to be genetically stable, with a stable yield of diosgenin. This mutant was used for later experiments.

3.2.1. Plackett-Burman Experiment Results

The mutagenic high-yielding fungi 11#SYfx213.2 were used for subsequent solid fermentation. In this experiment, taking the transformation rate of saponin as the response value (R1), the above seven factors were selected at two levels, i.e., high and low.
The specific Plackett-Burman experimental table and corresponding experimental results are shown in Table 4. (A: Content of sucrose; B: Content of NH4H2PO4; C: Content of MgSO4; D: Content of Tween-80; E: Content of rice husk; F: Content of KH2PO4; G: Content of wheat bran; R1: Extraction rate of diosgenin).
The Pareto charts (Figure 4) showed a Bonferroni limit of 5.74651 and a t-value limit of 2.77645, reflecting significant factors in a group. It was concluded that the influence of Tween-80 is the most significant. However, it had a negative effect and greatly inhibited improving the extraction rate of diosgenin. De Oliveira et al. produced beta-fructofuranosidase (FFase) using solid-state fermentation and found that the sucrose level had a significant effect on FFase production [28]. In Li et al.’s study, wheat bran had a greater positive effect on the bacterial community [29], so we ventured to add ammonium dihydrogen phosphate to the solid-state fermentation medium and found that it had a significant positive effect on the production of diosgenin. In our research, the effects of sucrose, wheat bran, and NH4H2PO4 were also significant; and their effects were positive. Adding MgSO4, rice husk, and KH2PO4 had no significant effect on the extraction rate of diosgenin.
The significance of this experiment was analyzed using the F test, and the results are shown in Table 5.
R1 (%, diosgenin yield) = 0.31 + 0.031 × A + 0.019 × B + 4.333 × 103 × C − 0.054 × D + 8.333 × 103 × E + 9.667 × 103 × F + 0.025
The fitting formula is Formula 1.
The whole model’s p-value ≤ 0.01, which is significant. The effect of Tween-80 was the most significant (p-value = 0.0011). The p-value of sucrose was 0.0084, the p-value of NH4H2PO4 was 0.039, and wheat bran’s p-value was 0.0165. These three factors were significant, and consistent with the results shown in the Pareto chart.

3.2.2. Response Surface Analysis of the Interaction of Various Factors

The Plackett-Burman experiment indicated that there are four significant factors. However, adding Tween-80 has a clear inhibitory effect on the yield and transformation rate of diosgenin, which is contrary to the purpose of this experiment. Therefore, as shown in the Table 6. Tween-80 was not added in the response surface experiments. Only sucrose, NH4H2PO4, and wheat bran were used in the additive optimization experiment.
Design expert v8.0.6 software was used to conduct quadratic regression analysis on the results of the above experimental design, and the final Equation (Formula 2) was obtained.
R1(%, diosgenin yield) = 0.45 − 6.750 × 103 × A − 9.375 × 103 × B + 8.125 × 103 × C + 5.250 × 103 × A × B − 9.750 × 103 × A × C − 5.000 × 103 × B × C − 0.022 × A2 − 1.500 × 103 × B2 − 0.02 × C2
Each symbol in the formula represents the following factors: R1 is the diosgenin yield (mg/L); A is the concentration of sucrose (g/L); B is the concentration of NH4H2PO4 (g/L); C is the concentration of wheat bran (g/L). The analysis of the model from the BBD shown in Table 7.
As shown in Table 7, the additive effects of sucrose, NH4H2PO4, and wheat bran on the yield of diosgenin are significant. The interactions between sucrose and NH4H2PO4 and between NH4H2PO4 and wheat bran were not significant. However, the interaction between sucrose and wheat bran had a notable effect on the diosgenin yield, with a p-value = 0.0209, indicating that the effects of sucrose and wheat bran on the yield are not simple linear relationships [30]. The strength of various additives’ influence on the diosgenin yield, judged by the F value, are NH4H2PO4 > wheat bran > sucrose. Tanyildizi et al. used response surface methodology (RSM) to optimize the medium composition for α-amylase production by solid substrate fermentation (SSF) and found that (NH4)(2)SO4 had the greatest effect on α-amylase production [31]. Based on the results of this experiment, we conjecture that it is the effect of NH4+ that makes the endophytic Aspergillus flavus strain 11#SYfx213.2 produce more α-amylase and thus decompose the polysaccharide material wrapped around diosgenin and contribute to the conversion of diosgenin. There is not much difference in the lack of fit of the model as prob > F is 0.9459, indicating that the model can simulate the experimental results well [32].
The response surface analysis (Figure 5) confirmed that the interactions between sucrose and NH4H2PO4, NH4H2PO4 and wheat bran, and sucrose and wheat bran affect diosgenin yield greatly [18].

3.3. Model Validation Experiment

The optimal fermentation medium composition was determined to be 5.47% of sucrose, 0.6% of NH4H2PO4, and 26.6% of wheat bran. The extraction rate of diosgenin reached 0.4491%. Considering the operability, the addition amount was adjusted to 5.5% of sucrose, 0.6% of NH4H2PO4, and 26.6% of wheat bran. Under the adjusted conditions, three repeated verification experiments were carried out and an average extraction rate of 0.439% was achieved.

3.4. Separation and Identification of Solid-State Fermentation Products

By separating the crude products using column chromatography, as shown in Figure 6. three substances were obtained, i.e., diosgenin, yuccagenone, and diosgenone.
The results of our analysis of the above chart are shown in the Table 8.
The chemical formula of compound 1 was deduced to be C27H42O3 from the quasi-molecular ion peak m/z 415.3215, [M+H]+, (cald. 415.3207 for C27H43O3) in the HR-MS (Supplementary Materials). In the 1H-NMR spectrum (Table 7), the presence of four methyl peaks in the high field, including two d peaks (δH 0.90, d, 6.8, 3H; 0.70, d, 7.0, 3H) and two s peaks (δH 0.96, s, 3H; 0.72, s, 3H), indicating that one could be a steroid. Olefinic protons (δH 5.28, brs, 1H) in the low field indicated that one had an olefinic fragment. In total, 27 carbon signals appeared in the 13C-NMR spectrum. The DEPT experiment differentiated them into four methyl groups (δC 19.4, 17.2, 16.3, and 14.5), nine methines (including an olefinic methine (δC 121.5)), two oxygenated methines (δC 80.9 and 71.8), and four quaternary carbons (including an olefinic quaternary carbon (δC 141.0), a bi-oxygenated quaternary carbon (δC 109.3) and two aliphatic quaternary carbons (δC 40.3 and 36.6)). It shows that compound 1 is a typical spirostan compound. Compared with the NMR data of diosgenin in the literature Chen reported [33], we can confirm that this compound is diosgenin, the target product of diosgenin chain degradation by microorganisms.
The quasi-molecular ion peak of m/z 415.3215, [M+H]+, (cald. 415.3207 for C27H43O3) appears in the HR-MS spectrum of compound 2. The chemical formula of this compound is C27H42O3, an isomer of diosgenin. However, compared with compound 1, it lacks olefinic protons at position 6 and even tertiary carbon-oxygen protons at position 3 in the 1H-NMR spectrum. The 13C-NMR spectrum lacks C-5, C-6 olefinic carbon signals, and even C-3 tertiary carbon-oxygen signals, but has an extra carbonyl group signal (δC 213.3). In addition, ring A and ring B’s signals in the 13C-NMR spectrum changed greatly, while the other signals were nearly unchanged. Therefore, it is speculated that this compound may be a diosgenin derivative with the 5,6-position double bond reduced and the 3-position hydroxyl group oxidized to a ketone group. The NMR data in previous literature confirmed this compound to be yuccagenone [34].
The quasi-molecular ion peak of m/z 413.3051, [M+H]+, (cald. 413.3050 for C27H41O3), appears in the HR-MS spectrum of compound 3. The chemical formula of this compound is C27H40O3. The NMR data of this compound are similar to those of compound 2, but with an added unsaturation appearing as two olefinic carbon signals (δC 171.4 and 124.1), indicating that this compound has an extra double bond compared with compound 2. Meanwhile, the signal of the carbonyl group significantly shifted to the high field (δC 199.8), and the carbon signals of the double bond are the typical signals of an α, β-unsaturated ketone fragment. Therefore, it is assumed that the carbonyl group of this compound is still at position 3, and the double bond is at positions 4 and 5. Combined with previous literature results, this compound is diosgenone [35].

4. Conclusions

We isolated Aspergillus flavus strains from an endophyte fungus. After mutagenesis, response surface methodology was used to optimize the solid-state fermentation medium, thereby improving the diosgenin yield [36]. The extraction of diosgenin using microbial conversion provides a new diosgenin production method to solve the problems of environmental pollution, resource waste, and high production cost caused by the traditional production process. In this study, the efficiency of the extraction of diosgenin using microbial transformation was greatly improved by means of strain mutagenesis and medium optimization. The most productive diosgenin strain was selected through strain mutation, and the solid-state fermentation was carried out. The optimization of the solid-state fermentation medium using the Plackett-Burman test indicated four substances that have a significant effect on diosgenin production, i.e., sucrose, NH4H2PO4, Tween-80, and wheat bran. The BBD test was carried out to test the three significant positively influencing factors [25]. The optimal addition amounts were 5.5% sucrose, 0.6% NH4H2PO4, and 26.6% wheat bran. The final extraction rate of diosgenin reached 0.439%. Compared with 0.338% before optimization, it had increased 1.29 times. Obviously, this method can effectively improve the extraction rate of diosgenin transformed by microorganisms.
This study not only produced diosgenin from the fermentation products, but also obtained and identified diosgenone after separation. This means that diosgenone was directly obtained using microbial transformation, avoiding the step of producing it from diosgenin through oxidation. This method greatly reduces the cost of producing diosgenone, and provides a basis for subsequent research on other pharmacological compounds.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/fermentation9010070/s1, Figure S1. Fatality rate curve of ultraviolet mutagenesis. Figure S2. Fatality rate curve of lithium chloride mutagenesis. Figure S3. Activation of strains and solid state fermentation. Table S1. The single factor experimental design. Figure S4. The effects of various additives on the extraction rate of diosgenin. Figure S5. HR-MS three isolated compounds. Figure S6. High performance liquid chromatography.

Author Contributions

F.M. and J.T.; Methodology, S.H.; Software, Y.L. and R.C.; Validation, F.M. and J.T.; Formal analysis, R.S.; Investigation, Y.Z.; Resources, S.F. and Y.S.; Data curation, F.M.; Writing—original draft preparation, J.T.; Writing—review and editing, Y.L.; Visualization, B.Q.; Supervision and administration. F.M. and J.T. are co-first authors of the article. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by the Shaanxi Province Key R&D Program (2022ZDLSF05-02), Shaanxi Province Strategic Emerging Industry Major Product (Group) Project (No. 2015KTCQ03-05) and the Shaanxi Province Key R&D Program (2021SF-387).

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Teaching and Research Core Facility at College of Life Science (Ningjuan Fan, Xiyan Chen, and Hui Duan) and Life Science Research Core Services, Northwest A&F University for the technical support.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds (diosgenin, yuccagenone, and diosgenone) and plant endophytic fungi SYfx213.2 are available from the authors.

References

  1. Fan, R.; He, W.; Fan, Y.; Xu, W.; Xu, W.; Yan, G.; Xu, S. Recent advances in chemical synthesis, biocatalysis, and biological evaluation of diosgenin derivatives—A review. Steroids 2022, 180, 108991. [Google Scholar] [CrossRef] [PubMed]
  2. Sun, G.C.; Jan, C.R.; Liang, W.Z. Exploring the impact of a naturally occurring sapogenin diosgenin on underlying mechanisms of Ca2+ movement and cytotoxicity in human prostate cancer cells. Environ. Toxicol. 2020, 35, 395–403. [Google Scholar] [CrossRef] [PubMed]
  3. Sirotkin, A.V.; Alexa, R.; Alwasel, S.; Harrath, A.H. The phytoestrogen, diosgenin, directly stimulates ovarian cell functions in two farm animal species. Domest. Anim. Endocrinol. 2019, 69, 35–41. [Google Scholar] [CrossRef] [PubMed]
  4. Yang, X.; Nomoto, K.; Tohda, C. Diosgenin content is a novel criterion to assess memory enhancement effect of yam extracts. J. Nat. Med. 2021, 75, 207–216. [Google Scholar] [CrossRef] [PubMed]
  5. Li, B.; Xu, P.; Wu, S.; Jiang, Z.; Huang, Z.; Li, Q.; Chen, D. Diosgenin Attenuates Lipopolysaccharide-Induced Parkinson’s Disease by Inhibiting the TLR/NF-kappa B Pathway. J. Alzheimers Dis. 2018, 64, 943–955. [Google Scholar] [CrossRef]
  6. Cai, D.; Qi, J.; Yang, Y.; Zhang, W.; Zhou, F.; Jia, X.; Guo, W.; Huang, X.; Gao, F.; Chen, H.; et al. Design, Synthesis and Biological Evaluation of Diosgenin-Amino Acid Derivatives with Dual Functions of Neuroprotection and Angiogenesis. Molecules 2019, 24, 4025. [Google Scholar] [CrossRef] [Green Version]
  7. Zeng, X.; Liu, D.; Huang, L. Metabolome Profiling of Eight Chinese Yam (Dioscorea polystachya Turcz.) Varieties Reveals Metabolite Diversity and Variety Specific Uses. Life 2021, 11, 687. [Google Scholar] [CrossRef]
  8. Zhang, R.; Li, P.; Xu, L.; Chen, Y.; Sui, P.; Zhou, L.; Li, J. Enhancement of Diosgenin Production in Dioscorea zingiberensis Cell Culture by Oligosaccharide Elicitor from its Endophytic Fungus Fusarium oxysporum Dzf17. Nat. Prod. Commun. 2009, 4, 1459–1462. [Google Scholar]
  9. Li, P.; Mou, Y.; Shan, T.; Xu, J.; Li, Y.; Lu, S.; Zhou, L. Effects of Polysaccharide Elicitors from Endophytic Fusarium oxysporium Dzf17 on Growth and Diosgenin Production in Cell Suspension Culture of Dioscorea zingiberensis. Molecules 2011, 16, 9003–9016. [Google Scholar] [CrossRef] [Green Version]
  10. Shen, B.W.; Yu, X.J.; Jiang, W.X.; Yuan, H.; Zhao, M.Q.; Zhou, H.; Pan, Z.Q. Green Conversion of Saponins to Diosgenin in an Alcoholysis System Catalyzed by Solid Acid Derived from Phosphorus Tailings. ACS Omega 2021, 6, 5423–5435. [Google Scholar] [CrossRef]
  11. Zhao, M.Q.; Pan, Z.Q.; Chen, Q.R.; Zhou, H. Catalytic Alcoholysis to Prepare Diosgenin with a Solid Acid Based on Nano TiO2. Catal. Lett. 2022, 152, 3453–3464. [Google Scholar] [CrossRef]
  12. Cheng, L.Y.; Zhang, H.; Cui, H.Y.; Davari, M.D.; Wei, B.; Wang, W.Y.; Yuan, Q.P. Efficient enzyme-catalyzed production of diosgenin: Inspired by the biotransformation mechanisms of steroid saponins in Talaromyces stollii CLY-6. Green Chem. 2021, 23, 5896–5910. [Google Scholar] [CrossRef]
  13. Gerocs, A.; Nagy, T.; Nemes-Barnas, K.; Majer, J.; Szoke, B.A.; Kovago, R.; Magalhaes, F.; Gibson, B.; Szekeres, A.; Juhasz, A.; et al. Characterization of Saccharomyces Strains Isolated from “Keknyelu” Grape Must and Their Potential for Wine Production. Fermentation 2022, 8, 416. [Google Scholar] [CrossRef]
  14. Nunez Perez, J.; Chavez Arias, B.S.; de la Vega Quintero, J.C.; Zarate Baca, S.; Manuel Pais-Chanfrau, J. Multi-Objective Statistical Optimization of Pectinolytic Enzymes Production by an Aspergillus sp. on Dehydrated Coffee Residues in Solid-State Fermentation. Fermentation 2022, 8, 170. [Google Scholar] [CrossRef]
  15. Banat, I.M.; Carboue, Q.; Saucedo-Castaneda, G.; de Jesus Cazares-Marinero, J. Biosurfactants: The green generation of speciality chemicals and potential production using Solid-State fermentation (SSF) technology. Bioresour. Technol. 2021, 320, 124222. [Google Scholar] [CrossRef]
  16. Krongrawa, W.; Limmatvapirat, S.; Saibua, S.; Limmatvapirat, C. Optimization of Ultrasound-Assisted Extraction of Yields and Total Methoxyflavone Contents from Kaempferia parviflora Rhizomes. Molecules 2022, 27, 4162. [Google Scholar] [CrossRef]
  17. Hallol, M.; Helmy, O.; Shawky, A.E.; El-Batal, A.; Ramadan, M. Optimization of Alpha-Amylase Production by a Local Bacillus paramycoides Isolate and Immobilization on Chitosan-Loaded Barium Ferrite Nanoparticles. Fermentation 2022, 8, 241. [Google Scholar] [CrossRef]
  18. Ghazanfar, M.; Nadeem, M.; Shakir, H.A.; Khan, M.; Ahmad, I.; Franco, M.; Chen, L.; Irfan, M. Valorization of Bombax ceiba Waste into Bioethanol Production through Separate Hydrolysis and Fermentation and Simultaneous Saccharification and Fermentation. Fermentation 2022, 8, 386. [Google Scholar] [CrossRef]
  19. He, S.; Wang, X.; Chen, J.; Li, X.; Gu, W.; Zhang, F.; Cao, G.; Yu, J. Optimization of the Ultrasonic-Assisted Extraction Technology of Steroidal Saponins from Polygonatum kingianum Collett & Hemsl and Evaluating Its Quality Planted in Different Areas. Molecules 2022, 27, 1463. [Google Scholar] [CrossRef]
  20. Yao, H.Y.Y.; Wang, J.Q.; Yin, J.Y.; Nie, S.P.; Xie, M.Y. A review of NMR analysis in polysaccharide structure and conformation: Progress, challenge and perspective. Food Res. Int. 2021, 143, 110290. [Google Scholar] [CrossRef]
  21. Chen, G.; Ma, T.; Ma, Y.; Han, C.; Zhang, J.; Sun, Y. Chemical Composition, Anti-Breast Cancer Activity and Extraction Techniques of Ent-Abietane Diterpenoids from Euphorbia fischeriana Steud. Molecules 2022, 27, 4282. [Google Scholar] [CrossRef] [PubMed]
  22. Ica, R.; Mlinac-Jerkovic, K.; Ilic, K.; Sajko, T.; Munteanu, C.V.A.; Zamfir, A.D.; Kalanj-Bognar, S. Gangliosidome of a Human Hippocampus in Temporal Lobe Epilepsy Resolved by High-Resolution Tandem Mass Spectrometry. Molecules 2022, 27, 4056. [Google Scholar] [CrossRef] [PubMed]
  23. Baquiao, A.C.; de Oliveira, M.M.M.; Reis, T.A.; Zorzete, P.; Atayde, D.D.; Correa, B. Polyphasic approach to the identification of Aspergillus section Flavi isolated from Brazil nuts. Food Chem. 2013, 139, 1127–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Li, Y.; Huang, Y.; Long, H.; Yu, Z.; Huang, M.; Liu, X. Biodiversity and Oenological Property Analysis of Non-Saccharomyces Yeasts Isolated from Korla Fragrant Pears (Pyrus sinkiangensis Yu). Fermentation 2022, 8, 388. [Google Scholar] [CrossRef]
  25. Lu, F.; Chao, J.; Zhao, X.; Betchem, G.; Ding, Y.; Yang, X.; Li, Y.; Ma, H. Enhancing protease activity of Bacillus subtilis using UV-laser random mutagenesis and high-throughput screening. Process Biochem. 2022, 119, 119–127. [Google Scholar] [CrossRef]
  26. Wang, Q.L.; Zhang, D.; Li, Y.D.; Zhang, F.M.; Wang, C.; Liang, X.L. Genome Shuffling and Ribosome Engineering of Streptomyces actuosus for High-Yield Nosiheptide Production. Appl. Biochem. Biotechnol. 2014, 173, 1553–1563. [Google Scholar] [CrossRef]
  27. Widyastuti, W.; Setiawan, F.; Al Afandy, C.; Irawan, A.; Laila, A.; Juliasih, N.; Setiawan, W.A.; Arai, M.; Hendri, J.; Setiawan, A.; et al. Antifungal Agent Chitooligosaccharides Derived from Solid-State Fermentation of Shrimp Shell Waste by Pseudonocardia antitumoralis 18D36-A1. Fermentation 2022, 8, 353. [Google Scholar] [CrossRef]
  28. De Oliveira, R.L.; da Silva, M.F.; Converti, A.; Porto, T.S. Production of β-fructofuranosidase with transfructosylating activity by Aspergillus tamarii URM4634 Solid-State Fermentation on agroindustrial by-products. Int. J. Biol. Macromol. 2020, 144, 343–350. [Google Scholar] [CrossRef]
  29. Li, X.Q.; Liu, D.Q. Effects of wheat bran co-fermentation on the quality and bacterial community succession during radish fermentation. Food Res. Int. 2022, 157, 111229. [Google Scholar] [CrossRef]
  30. Foudah, A.I.; Alshehri, S.; Shakeel, F.; Alqarni, M.H.; Aljarba, T.M.; Alam, P. Simultaneous Estimation of Escitalopram and Clonazepam in Tablet Dosage Forms Using HPLC-DAD Method and Optimization of Chromatographic Conditions by Box-Behnken Design. Molecules 2022, 27, 4209. [Google Scholar] [CrossRef]
  31. Tanyildizi, M.S.; Selen, V.; Ozer, D. Optimization of alpha-Amylase Production in solid substrate fermentation. Can. J. Chem. Eng. 2009, 87, 493–498. [Google Scholar] [CrossRef]
  32. Kumar, M.; Dahuja, A.; Tiwari, S.; Punia, S.; Tak, Y.; Amarowicz, R.; Bhoite, A.G.; Singh, S.; Joshi, S.; Panesar, P.S.; et al. Recent trends in extraction of plant bioactives using green technologies: A review. Food Chem. 2021, 353, 129431. [Google Scholar] [CrossRef]
  33. Chen, Y.; Dong, Y.; Chi, Y.L.; He, Q.; Wu, H.; Ren, Y. Eco-friendly microbial production of diosgenin from saponins in Dioscorea zingiberensis tubers in the presence of Aspergillus awamori. Steroids 2018, 136, 40–46. [Google Scholar] [CrossRef]
  34. Blunden, G.; Carabot, A.; Cripps, A.L.; Jewers, K. Ruizgenin, a new steroidal sapogenin diol from Agave lecheguilla. Steroids 1980, 35, 503–510. [Google Scholar] [CrossRef]
  35. Hernandez Linares, M.-G.; Guerrero-Luna, G.; Bernes, S.; Flores-Alamo, M.; Fernandez-Herrera, M.A. Diosgenone: A second P21 polymorph. Acta Crystallogr. Sect. E 2012, 68, o2358. [Google Scholar] [CrossRef] [Green Version]
  36. Choi, K.R.; Jang, W.D.; Yang, D.; Cho, J.S.; Park, D.; Lee, S.Y. Systems Metabolic Engineering Strategies: Integrating Systems and Synthetic Biology with Metabolic Engineering. Trends Biotechnol. 2019, 37, 817–837. [Google Scholar] [CrossRef]
Figure 1. (a) The sporangia of SYfx213.2 fungus under the optical microscope at 800-times magnification; (b) spores of SYfx213.2 fungus under the optical microscope at 800-times magnification; (c) 1000-times magnification of mycelia under the scanning electron microscope; (d) 2500-times magnification of Conidial head under the scanning electron microscope.
Figure 1. (a) The sporangia of SYfx213.2 fungus under the optical microscope at 800-times magnification; (b) spores of SYfx213.2 fungus under the optical microscope at 800-times magnification; (c) 1000-times magnification of mycelia under the scanning electron microscope; (d) 2500-times magnification of Conidial head under the scanning electron microscope.
Fermentation 09 00070 g001
Figure 2. Fungal strain SYfx213.2 and its internal transcribed spacer ITS DNA phylogenetic tree. Numbers in brackets indicate GenBank accession number.
Figure 2. Fungal strain SYfx213.2 and its internal transcribed spacer ITS DNA phylogenetic tree. Numbers in brackets indicate GenBank accession number.
Fermentation 09 00070 g002
Figure 3. (a) Thin layer chromatogram of diosgenin from screened mutant strains. Strain 0 was the unmutagenized strain SYfx213.2 as the control. Strain 6, 8, 11, 15, 23, 38, 42 were strains with higher yield of diosgenin than the control strain. (b) Comparison diagram of diosgenin yield of mutant strains. Unmutagenized strain SYfx213.2 used as the control group.
Figure 3. (a) Thin layer chromatogram of diosgenin from screened mutant strains. Strain 0 was the unmutagenized strain SYfx213.2 as the control. Strain 6, 8, 11, 15, 23, 38, 42 were strains with higher yield of diosgenin than the control strain. (b) Comparison diagram of diosgenin yield of mutant strains. Unmutagenized strain SYfx213.2 used as the control group.
Fermentation 09 00070 g003
Figure 4. (a) Pareto chart of the Plackett-Burman experiment; (b) distribution chart of the Plackett-Burman experiment results. (A: Content of sucrose; B: Content of NH4H2PO4; C: Content of MgSO4; D: Content of Tween-80; E: Content of rice husk; F: Content of KH2PO4; G: Content of wheat bran; R1: Extraction rate of diosgenin).
Figure 4. (a) Pareto chart of the Plackett-Burman experiment; (b) distribution chart of the Plackett-Burman experiment results. (A: Content of sucrose; B: Content of NH4H2PO4; C: Content of MgSO4; D: Content of Tween-80; E: Content of rice husk; F: Content of KH2PO4; G: Content of wheat bran; R1: Extraction rate of diosgenin).
Fermentation 09 00070 g004
Figure 5. (a,b): sucrose × NH4H2PO4 response surface plot and contour map; (c,d): sucrose × wheat bran response surface plot and contour map; (e,f): NH4H2PO4 × wheat bran response surface plot and contour map.
Figure 5. (a,b): sucrose × NH4H2PO4 response surface plot and contour map; (c,d): sucrose × wheat bran response surface plot and contour map; (e,f): NH4H2PO4 × wheat bran response surface plot and contour map.
Fermentation 09 00070 g005aFermentation 09 00070 g005b
Figure 6. NMR spectra of the three compounds (a) diosgenin C-NMR, (b) diosgenin 1H-NMR, (c) yuccagenone C-NMR, (d) yuccagenone 1H-NMR, (e) diosgenone C-NMR, (f) diosgenone 1H-NMR.
Figure 6. NMR spectra of the three compounds (a) diosgenin C-NMR, (b) diosgenin 1H-NMR, (c) yuccagenone C-NMR, (d) yuccagenone 1H-NMR, (e) diosgenone C-NMR, (f) diosgenone 1H-NMR.
Fermentation 09 00070 g006aFermentation 09 00070 g006bFermentation 09 00070 g006c
Table 1. Cross experiment table of compound mutagenesis.
Table 1. Cross experiment table of compound mutagenesis.
Impact FactorA: Uv Irradiation Time (s)B: LiCl Concentration (%)
Level 1180 s1.5%
Level 2210 s2.0%
Level 3240 s2.5%
Table 2. The Plackett-Burman experimental design.
Table 2. The Plackett-Burman experimental design.
CodeFactor−11
AContent of sucrose6%4%
BContent of NH4H2PO40.6%0.8%
CContent of MgSO40.4%0.8%
DContent of Tween-801 g/L3 g/L
EContent of rice husk20%30%
FContent of KH2PO40.5%1.5%
GContent of wheat bran20%30%
Table 3. The results of crossover experiments with compound mutagenesis conditions.
Table 3. The results of crossover experiments with compound mutagenesis conditions.
Number of TrialsA: Uv Irradiation Time (s)B: LiCl Concentration (%)Fatality Rate (%)
1180 s1.5%70%
2180 s2.0%80%
3180 s2.5%90%
4210 s1.5%70%
5210 s2.0%90%
6210 s2.5%100%
7240 s1.5%90%
8240 s2.0%90%
9240 s2.5%100%
Table 4. Solid-state fermentation to produce diosgenin Plackett-Burman experimental table and results.
Table 4. Solid-state fermentation to produce diosgenin Plackett-Burman experimental table and results.
Number of TestsABCDEFGR1/%
1−111−11110.419
2111−1−1−110.427
3−1111−1−1−10.203
4−1−11−111−10.291
5−1−1−1−1−1−1−10.275
61−1−1−11−110.392
711−1−1−11−10.376
8−11−111−110.244
91−1111−1−10.258
10−1−1−11−1110.241
1111−1111−10.303
121−111−1110.285
Table 5. Regression Equation F test table of this experiment.
Table 5. Regression Equation F test table of this experiment.
Quadratic SumDegree of FreedomVarianceFProb > F
Model0.06078.607 × 10317.870.0072
A-sucrose0.01110.01123.430.0084
B-NH4H2PO44.408 × 10314.408 × 1039.150.0390
C-MgSO42.253 × 10−412.253 × 10−40.470.5316
D-Tween-800.03510.03572.200.0011
E-rice husk8.333 × 10−418.333 × 10−41.730.2587
F-KH2PO41.121 × 10311.121 × 1032.330.2018
G-wheat bran7.600 × 10317.600 × 10315.780.0165
Residual1.927 × 10344.817 × 10−4
Cor Total0.06211
Table 6. Solid-state fermentation to produce diosgenin response surface experimental table and results.
Table 6. Solid-state fermentation to produce diosgenin response surface experimental table and results.
Number of TestsA: Content of SucroseB: Content of NH4H2PO4C: Content of Wheat BranR1/%
150.6200.426
250.7250.452
350.7250.437
450.8200.419
540.8250.418
660.7300.401
740.7300.436
860.7200.402
940.7200.398
1050.7250.459
1140.6250.449
1250.7250.455
1350.7250.452
1460.8250.417
1550.6300.45
1660.6250.427
1750.8300.423
Table 7. Statistical analysis of the model from the BBD.
Table 7. Statistical analysis of the model from the BBD.
Sum of SquaresdfMean SquareF-Valuep-Value
Model0.006190.000715.770.0007
A-sucrose0.000410.00048.440.0228
B-NH4H2PO40.000710.000716.280.0050
C-wheat bran0.000510.000512.230.0100
AB0.000110.00012.550.1541
AC0.000410.00048.810.0209
BC0.000110.00012.320.1719
A^20.002010.002046.130.0003
B^29.474 × 10−619.474 × 10−60.21940.6537
C^20.001710.001739.010.0004
The rest of item0.000370.0000
Lack of fit0.000038.083 × 10−60.11630.9459
Pure error0.000340.0001
Total0.006416
Table 8. 1H NMR-(400MHz) 13C NMR-(400MHZ).
Table 8. 1H NMR-(400MHz) 13C NMR-(400MHZ).
1 2 3
The Number of CarbonsδH (J in Hz)δCδH (J in Hz)δCδH (J in Hz)δC
1 37.3 37.2 35.7
2 31.4 37.1 34.0
33.46 (br s)71.8 213.3 199.8
4 42.3 42.45.66 (s)124.1
5 141.0 44.3 171.4
65.28 (br s)121.5 26.1 32.9
7 32.1 26.6 32.2
8 31.6 35.2 35.3
9 50.1 40.8 53.8
10 36.6 35.0 38.7
11 20.9 21.1 20.8
12 39.8 40.1 39.7
13 40.3 40.7 40.4
14 56.5 56.3 55.7
15 32.1 31.8 31.7
164.34 (q, 7.3)80.94.34 (q, 7.2)80.84.34 (q, 7.2)80.7
17 62.1 62.3 62.0
180.72 (s, 3H)16.30.72 (s, 3H)16.60.75 (s, 3H)16.4
190.96 (s, 3H)19.40.97 (s, 3H)22.71.13 (s, 3H)17.5
20 41.6 41.6 41.7
210.90 (d, 6.8, 3H)14.50.91 (d, 6.8, 3H)14.60.90 (d, 6.8, 3H)14.6
22 109.3 109.3 109.4
23 31.4 31.4 31.4
24 28.8 28.8 28.8
25 30.3 30.3 30.3
263.30 (t, 11.0)
3.41 (dd, 11.0, 3.3)
66.93.31 (t, 11.0)
3.41 (dd, 11.0, 3.3)
66.93.30 (t, 11.0)
3.41 (dd, 11.0, 3.3)
66.9
270.74 (d, 7.0, 3H)17.20.72 (overlap, 3H)17.20.73 (d, 6.5, 3H)17.2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mou, F.; Tian, J.; Li, Y.; Han, S.; Shang, R.; Song, Y.; Feng, S.; Zhang, Y.; Cao, R.; Qin, B. A Method for Improving Microbial Conversion of Diosgenin and Separation and Identification of the Product. Fermentation 2023, 9, 70. https://doi.org/10.3390/fermentation9010070

AMA Style

Mou F, Tian J, Li Y, Han S, Shang R, Song Y, Feng S, Zhang Y, Cao R, Qin B. A Method for Improving Microbial Conversion of Diosgenin and Separation and Identification of the Product. Fermentation. 2023; 9(1):70. https://doi.org/10.3390/fermentation9010070

Chicago/Turabian Style

Mou, Fangyuan, Junmian Tian, Yulu Li, Shiyao Han, Ruifen Shang, Yuxin Song, Shirong Feng, Yongli Zhang, Rang Cao, and Baofu Qin. 2023. "A Method for Improving Microbial Conversion of Diosgenin and Separation and Identification of the Product" Fermentation 9, no. 1: 70. https://doi.org/10.3390/fermentation9010070

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop