Review
Twitter Facebook LinkedIn

Advances in the Production of Minor Ginsenosides Using Microorganisms and Their Enzymes

Almando Geraldi1,2,*

1Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, 60115, Indonesia

2Research Center for Bio-Molecule Engineering, Universitas Airlangga, Surabaya, 60115, Indonesia

*Correspondence: Almando Geraldi, Tel: +62-31-5936501; E-mail: almando.geraldi@fst.unair.ac.id

Received: February 29 2020; Revised: March 19 2020; Accepted: April 27 2020; Published Online: May 29 2020


Cite this paper:

Almando Geraldi. Advances in the Production of Minor Ginsenosides Using Microorganisms and Their Enzymes. BIO Integration 2020; 1(1): 15–24.

DOI: 10.15212/bioi-2020-0007. Available at: https://bio-integration.org/

Download citation download

This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See https://bio-integration.org/copyright-and-permissions/

Abstract

Minor ginsenodes are of great interest due to their diverse pharmacological activities such as their anti-cancer, anti-diabetic, neuroprotective, immunomodulator, and anti-inflammatory effects. The miniscule amount of minor ginsenosides in ginseng plants has driven the development of their mass production methods. Among the various production methods for minor ginsenosides, the utilization of microorganisms and their enzymes are considered as highly specific, safe, and environmentally friendly. In this review, various minor ginsenosides production strategies, namely utilizing microorganisms and recombinant microbial enzymes, for biotransforming major ginsenosides into minor ginsenoside, as well as constructing synthetic minor ginsenosides production pathways in yeast cell factories, are described and discussed. Furthermore, the present challenges and future research direction for producing minor ginsenosides using those approaches are discussed.

Keywords

biotransformation, biosynthesis, β-glucosidase, Ginsenosides, minor ginsenosides.

Introduction

Ginseng, a plant belonging to the Araliaceae family and the genus Panax, has been used to treat different kinds of ailments and disease in East Asian countries for millennia [1, 2]. Recently, the popularity of ginseng as a nutraceutical and alternative medicine has been increasing worldwide [3, 4]. The global ginseng market has been reported to be valued at over two billion U.S. dollars and is expected to grow exponentially [5].

The beneficial health effects of ginseng are mainly attributed to ginsenosides, the major bioactive compound of ginseng [6, 7]. Various in vitro and in vivo studies have demonstrated the diverse pharmacological activities of ginsenosides such as anti-microbial, antioxidant, anti-inflammatory, skin-protective, neuroprotective, anti-cancer, and anti-diabetic effects (Table 1). Most of ginsenosides are classified as protopanaxadiol (PPD), and protopanaxatriol (PPT) types [8]. Both types consist of an aglycon (a non-sugar component) of a dammarane skeleton (PPD or PPT) as an aglycon and one to four molecules of sugar moieties at C-3 and C-20 positions in the case of PPD-type ginsenosides or at C-6 and C-20 positions in the case of PPT-type ginsenosides [9, 10].

Table 1 Example of PPD- and PPT-type Ginsenosides with Their Health Benefits

bioi-2020-0007-fx1.jpg

20(S)-Protopanaxadiol-type ginsenoside

Name R1 (C3) R3 (C20) Health Benefits References
Rb1 Major ginsenosides Glc12Glc Glc16Glc Neuroprotective and anti-diabetic effects [11, 12]
Rb2 Glc12Glc Ara(p)16Glc Neuroprotective and anti-inflammatory effects [13, 14]
Rc Glc12Glc Ara(f)16Glc Anti-inflammatory and skin-protective effects [15, 16]
Rd Glc12Glc Glc– Anti-cancer and neuroprotective effects [17, 18]
F2 Minor ginsenosides Glc– Glc– Anti-oxidant, anti-cancer, and skin-protective effects [1921]
Rg3 Glc12Glc H Anti-depressant, anti-virus, and anti-cancer effects [2224]
Gypenoside XVII (GypXVII) Glc– Glc16Glc Cardioprotective and neuroprotective effects [25, 26]
Gypenoside LXXV (GypLXXV) H– Glc16Glc Skin-protective and anti-cancer effects [27, 28]
Rh2 Glc– H– Skin protective, immunomodulator, anti-virus, and anti-cancer effects [2932]
Compound K (CK) H Glc– Anti-epileptic, anti-cancer, and skin-protective effects [3335]
bioi-2020-0007-fx1.jpg

20(S)-Protopanaxatriol-type ginsenoside

Name R2 (C6) R3 (C20) Health Benefit References
Re Major ginsenosides Rha12Glc Glc– Anti-diabetic, kidney-, and heart-protective effects [3638]
Rf Glc12Glc H Neuroprotective and anti-inflammatory effects [39, 40]
Rg1 Glc– Glc– Neuroprotective and hepatoprotective effects [41, 42]
Rg2 Minor ginsenosides Rha12Glc H- Skin-protective, antidepressant, and anti-inflammatory effects [4345]
Rh1 Glc- H Anti-oxidant, anti-inflammatory, immunomodulator, and anti-cancer [46, 47]
F1 H Glc- Anti-aging, anti-oxidant, skin-protective, immunomodulator [48, 49]

Ara(f): α-l-arabinofuranosyl; Ara(p): α-l-arabinopyranosyl; Glc: α-d-glucopyranosyl; Rha: α-l-ahamnopyranosyl.

Meanwhile, based on their abundance in ginseng, ginsenosides can be classified as major ginsenoside and minor ginsenosides. Major ginsenosides [i.e., Rd, Rc, Rb2, and Rb1 (PPD-type); Rg1, Rf, and Re (PPT-type)] constitute more than 90% of the total ginsenosides, while minor ginsenosides [i.e. F2, Rg3, Rh2, and Compound K (PPD-type); Rg2, Rh1, and F1 (PPT-type)] are only present in small quantities in ginseng [50, 51]. Despite showing some therapeutic effects such as, anti-inflammatory, anti-diabetic, and neuroprotective effects, major ginsenosides are not easily absorbed by the human body [52]. On the other hand, while they are only present in very low amounts in ginseng, minor ginsenosides are considered as being more pharmacologically active than major ginsenosides due to their smaller molecule size, better permeability across the cell membrane, and thus their higher bioavailability [53, 54].

Minor ginsenosides are of commercial interest due to their diverse biological activities and high pharmacological activities. However, the miniscule amount of minor ginsenosides extracted from ginseng cannot satisfy the needs of scientific and clinical studies, as well as commercial purposes [55, 56]. Therefore, it is very important to develop useful methods for the mass production of minor ginsenosides.

Minor ginsenosides production methods

The most common approach to producing minor ginsenosides is by hydrolyzing the sugar moieties of major ginsenosides [57]. The hydrolysis can be conducted through physical (heat and microwave transformation), chemical (acid and alkali hydrolysis), and biological (biotransformation) methods. Heat transformation via baking and steaming transforms PPD-type major ginsenosides Rb1, Rb2, and Rc2 into Rd, and finally into minor ginsenosides Rg3, F2, compound K, and Rh2, as well as PPT-type major ginsenosides Re and Rf into minor ginsenosides Rh1 and F1, with by-products of acetyl-ginsenosides [58, 59]. Meanwhile, acid hydrolysis using hydrochloric acid transformed Rb1 into Rg3 [60]. The physical and chemical methods are considered to be fast and simple; however, the use of those methods often results in the formation of undesirable by-products due to the low specificity. Moreover, there are safety and environmental issues, for example, due to the use of high temperature in the heat transformation approach, or strong acid/base in the acid/alkali hydrolysis methods. On the other hand, biological methods which involve enzymes offer higher reaction specificity, can be conducted in mild conditions minimizing safety risks, and are more environmentally friendly [61]. Thus, there is growing interest in using biological methods for the mass production of minor ginsenosides.

In general, the biological methods for producing minor ginsenosides can be categorized as; the use of microbial cells and enzymes for hydrolyzing sugar moieties of major ginsenosides, and the emerging biosynthesis methods, where the minor ginsenoside biosynthesis pathway is introduced into microorganism hosts. The present paper reviews the advances on using those biological methods for producing minor ginsenosides.

Biotransformation of major ginsenosides into minor ginsenosides using microorganisms

A wide variety of microorganisms have been used in the biotransformation of major ginsenosides into minor ginsenosides (Table 2). The biotransformation activities are mainly attributed to β-glucosidases (β-d-glucopyranoside glucohydrolase) [E.C.3.2.1.21] which hydrolyze the glycosidic bonds of the sugar moieties of the major ginsenosides at the C-3, C-6, and C-20 positions [62]. However, due to specific structures of the aglycon (dammarane skeleton), only specific β-glucosidases, thus specific microorganisms are able to hydrolyze ginsenoside-β-glucoside linkages [63].

Table 2 Production of Minor Ginsenosides in Different Microorganism

Microorganism Source of Microorganism Transformation Pathway Remarks
Flavobacterium sp. GE 32 [65] Root of Panax ginseng (Jilin, China) Rb1 → GypXVII
Rb1 → Rd → Rg3
Burkholderia sp. GE 17–7 [66] P. ginseng (Jilin, China) Rb1 → Rd → Rg3 Rg3 was produced with conversion rate of 98% after 15 h
Arthrinium sp. GE 1718 [67] Root of P. ginseng (Jilin, China) Rb1 → Rd → F2 → CK Endophytic fungi
Cellulosimicrobium sp. TH-20 [68] Soil of ginseng field (Fusong, China) Re → Rg2 Re (30 mg) was transformed into Rg2 (24 mg) with a yield of 96%
C. allociferrii JNO301 [70] Meju (dried fermented soybeans) from South Korea Rb1 → Rd → F2
Rf → Rh1
Yeast
Leuconostoc mesenteroides WIKim19 [71] Kimchi (fermented vegetable) from South Korea Rb1 → Rd → Rg3
Lactobacillus rhamnosus GG [72] Culture collection Rb1 → Rd Probiotic microorganism
Schizophyllum commune [73] Culture collection Rb1 → Rd → F2 → CK Edible and medicinal fungi

Ginseng plantation fields are one of the main sources of microorganisms with major ginsenoside-biotransforming activities. Endophytic microorganisms, spend all or part of their life cycle inside ginseng plants without damaging the plant tissues or inducing defense responses, for example, Burkholderia sp. GE 17-7 and Flavobacterium sp. GE 32, as well as fungi Arthrinium sp. GE 17-18 [64]. Those microorganisms exhibit hydrolysis activities on PPD-type ginsenosides. Flavobacterium sp. GE 32 has been reported to hydrolyze the outer glycosidic linkage of Rb1 at C-3 to produce Gypenoside XVII and the C-20 position to produce Rd. The bacteria also showed hydrolysis activities on the glycosidic linkage at the C-20 position of Rd to generate Rg3 [65]. Similar hydrolysis activities on the terminal and inner glucopyranosyl moieties at the C-20 position of Rb1 to produce Rg3 were also shown by Burkholderia sp. GE 17-7 [66]. Meanwhile Arthrinium sp. GE 17-18 was shown to have hydrolysis activity on terminal and inner glucopyranosyl moieties at the C-3 position and terminal glucopyranosyl moieties at the C-20 position of Rb1 to generate CK [67]. Finally, Cellulosimicrobium sp. TH-20 isolated from rhizosphere soil of ginseng showed biotransformation activities on PPT-type Re to Rg2 by hydrolyzing sugar moieties at the C-20 position [68].

Generally recognized as safe (GRAS) microorganisms (i.e., probiotics, microorganisms from fermented foods, etc.), which are non-pathogenic and considered as safe to be used for nutraceutical and pharmaceutical purposes, were also reported to have major ginsenoside-biotransforming activities [69]. Candida allociferrii JNO301 yeast isolated from Korean fermented soybean showed hydrolysis activity on outer glucopyranosyl moieties at the C-3 and C-20 positions of Rb1 to produce F2. Interestingly the yeast also exhibited biotransformation activity on PPT-type ginsenoside Rf into Rh1, by hydrolyzing outer sugar moieties at the C-6 position [70].

Biotransformation of major ginsenosides into minor ginsenosides using β-glucosidase from microorganisms

Purified recombinant enzymes are considered superior to the enzymes isolated and purified from cultured microorganisms due to their higher selectivity and activity [7476]. Gram-scale quantities of minor ginsenosides were produced using enzymes from Microbacterium sp. Gsoil 167 isolated from ginseng plantation and Lactobacillus ginsenosidimutans EMML 3041 from kimchi (fermented vegetable) [27, 77] (Table 3). Thermostable β-glucosidase were also reported to exhibit ginsenoside-biotransformation activities which resulted in the production of Gram-scale quantities of minor ginsenosides. Those thermostable enzymes are of industrial interest as the enzymes can be used in combination with heat and acid hydrolysis to further accelerate and increase the yield of major ginsenoside biotransformation. While most of ginsenoside-biotransforming microorganisms and enzymes are isolated from the East Asia region, interestingly one bacterial isolate from Indonesia has β-glucosidase that can hydrolyze the outer sugar moieties at C-3 and C-20 of Rb1 to produce F2 [62].

Table 3 Production of Minor Ginsenosides Using Different Recombinant β-Glucosidase

β-Glucosidase Source of Microorganism Transformation Pathway Yields
Recombinant β-glucosidase from Microbacterium sp. Gsoil 167 expressed in E. coli BL21(DE3) [27] Soil of ginseng field (Pocheon, South Korea) GypXVII → GypLXXV GypXVII (10 g) was transformed into GypLXXV (5.7 g; 69.6% recovery; and 97.8 chromatography purity)
Recombinant β-glucosidase from Arachidicoccus ginsenosidimutans sp. nov. expressed in E. coli BL21(DE3) [83] Ginseng compost (South Korea) Rb1 → GypXVII → F2 → CK
Rd → F2
Recombinant β-glucosidase from Microbacterium esteraromaticum expressed in E. coli BL21(DE3) [84] Soil of ginseng field (South Korea) Re → Rg2 Re (1 mg) was transformed into Rg2 (0.83 mg; 100% molar conversion yield within 150 min)
Rg1 → Rh1
Rg1 (1 mg) was transformed into Rh1 (0.6 mg; 78% molar conversion yield within 15 min)
Recombinant β-glucosidase from M. esteraromaticum expressed in E. coli BL21(DE3) [85] Soil of ginseng field (South Korea) Rb1 → Rd → CK Rb1 (1 mg/ml) was transformed into CK (0.46 mg/ml; 77% molar conversion yield within 60 min)
Recombinant β-glucosidase from Pseudonocardia sp. Gsoil 1536 expressed in E. coli BL21(DE3) [51] Soil of ginseng field (Pocheon, South Korea) Re → Rg2 Re (150 g) was transformed into Rg2 (150 g; 84.0±1.1% chromatographic purity)
Recombinant β-glucosidase from Lactobacillus brevis expressed in E. coli BL21(DE3) [86] Kimchi (fermented vegetable) from South Korea GypXVII → CK GypXVII was transformed into CK (89% molar conversion yield within 6 h)
Recombinant β-glucosidase from L. ginsenosidimutans EMML 3041 expressed in E. coli BL21(DE3) [77] Kimchi (fermented vegetable) from South Korea Rb1 → Rd → Rg3 Rb1 (50 g) was transformed into Rg3 (30 g; 74.3% chromatographic purity)
Recombinant β-glucosidase from Thermotoga thermarum DSM 5069 expressed in E. coli JM109 (DE3) [87] Culture Collection Re → Rg2 Re (10 g/L) was transformed into Rg2 (8.02 g/L) within 60 min at 85 °C and PH 5.5.
Rg1 → Rh1
Rg1 (2 g/L) was transformed into Rh1 (1.56 g/L) within 60 min at 85 °C and PH 5.5
Recombinant β-glucosidase from Caldicellulosiruptor bescii in E. coli [88] Culture Collection Rb1 → Rd → F2 → CK CK was produced from Rb1 with the productivity of 1000 (μM/h) within 80 min at 80 °C and PH 5.5
Rc → Rd → F2 → CK
CK was produced from Rc with the productivity of 400 (μM/h) within 180 min at 80 °C and and PH 5.5
Recombinant β-glucosidase from Thermotoga neapolitana DSM 4359 in E. coli [89] Culture Collection Re → Rg2 Re (2 mg/ml) was transformed into Rg2 (1.66 mg/ml; 100% molar conversion yield) within 3 h at 85 °C and PH 5.5
Recombinant β-glucosidase from Serratia marcescens LII61 expressed in E. coli BL21(DE3) [62] Slaughterhouse waste (Indonesia) Rb1 → Rd → F2
Rb1 → GypXVII → F2
Recombinant β-glucosidase from M. testaceum ATCC 15829 expressed in C. glutamicum ATCC 13032 [56] Culture Collection Rb1 → GypXVII → GypLXXV → F2 → CK CK (7.59 g/L) was produced from PPD-type ginsenoside mixtures in 24 h
Rd → F2 → CK
Re → F1 F1 (9.42 g/L) was produced from PPT-type ginsenoside mixtures in 24 h
Rg → F1
Recombinant β-glucosidase from Paenibacillus mucilaginosus KCTC 3870 expressed in C. glutamicum ATCC 13032 [81] Culture Collection Rg3 → Rh2
Recombinant β-glucosidase from Flavobacterium johnsoniae expressed in L. lactis subsp. cremoris NZ9000 [90] Culture Collection Rb1 → Rd → F2 → CK
Rb1 → Rd → Rg3
Recombinant β-glucosidase from P. mucilaginosus expressed in L. lactis subsp. cremoris NZ9000 [91] Culture Collection Rb1 → Rd → F2 F2 was produced from PPD-type ginsenoside mixtures with 74% conversion yield

Based on the data in Table 3, Escherichia coli is the most common host for the production of recombinant β-glucosidase. This is due to the simplicity of E. coli genetic modification and its rapid growth in relatively inexpensive media [78, 79]. However, E. coli is not preferable for application in food and pharmaceutical industries due to its non-GRAS status [80]. Efforts to produce ginsenoside transforming-β-glucosidase in GRAS microorganisms, such as Corynebacterium glutamicum and Lactococcus lactis have been attempted. Microbacterium testaceum β-glucosidase was successfully expressed in C. glutamicum and exhibit biotransformation activity for both PPD- and PPT-type ginsenosides, and resulted in the production of Gram-scale quantities of CK and F1 [56]. However, in general, the quantity and activity of ginsenoside biotransforming-β-glucosidases produced in GRAS hosts are not as high as recombinant β-glucosidases produced in E. coli [81, 82].

Biosynthesis of minor ginsenoside in microorganisms

Despite the high efficiency, biotransformation methods for producing minor ginsenosides require ginseng extracts as the raw material which are produced through time-consuming (typically, 5–6 years are required to produce marketable ginseng), labor-intensive, energy-consuming, and high-cost processes and can be affected by many factors such as soil quality, climate, pathogens, and pests [92, 93]. The discovery of genes encoding enzymes involved in the minor ginsenosides synthesis pathway coupled with the advances in synthetic biology tools, has allowed the construction of microbial cell factories, which can provide more sustainable and cost-effective alternative to mass-produced minor ginsenosides from renewable resources [94, 95].

The ginsenoside biosynthesis pathway has mainly been introduced for three species of yeasts (Saccharomyces cerevisiae, Pichia pastoris, and Yarrowia lipolytica) which are on the GRAS list of microorganisms, and are compatible for the expression of plant-derived heterologous enzymes [95]. In general, there are three general strategies to produce minor ginsenosides in yeast cell factories: (1) improving the yield of the yeast native mevalonic acid (MVA) pathway; (2) introducing genes for the synthesis of ginsenosides aglycons; and (3) introducing uridine diphosphate (UDP)-glycosyltransferase genes for the addition of sugar moieties to the aglycons (Figure 1).

Figure 1 Biosynthetic pathways for minor ginsenosides in metabolically engineered S. cerevisiae.

Single arrows represent one-step conversions, while multiple arrows represent multiple steps. Bold, blue arrow represents over-expressed modified yeast endogenous genes (tHMGR: truncated 3-hydroxy-3-methylglutaryl-CoA reductase). Bold, green arrows represent over-expressed yeast endogenous genes (FPS: farnesyl pyrophosphate synthase; SS: squalene synthase; SQE: squalene epoxidase). Bold, red arrows represent exogenous plant genes that were introduced into S. cerevisiae (PgDDS: P. ginseng dammarenediol II synthase; CYP716A47: P. ginseng protopanaxadiol synthase; CYP716A53v2: P. ginseng protopanaxatriol synthase; CPR: A. thaliana/P. ginseng NADPH-cytochrome P450 reductase; PgUGTs: P. ginseng UDP-glycosyltransferases). Dashed arrow represents competing pathway (LS: lanosterol synthase). Intermediates: HMG-CoA: 3-hydroxy-3-methylglutaryl coenzyme A; IPP: isopentenyl pyrophosphate; DMAPP: dimethylallyl pyrophosphate; FPP: farnesyl diphosphate.

bioi-2020-0007-f01.jpg

The MVA pathway produces isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP) which are important precursors for the synthesis of ginsenosides, from acetyl coenzyme A (acetyl-CoA). The native MVA pathway of yeasts is improved by overexpressing 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR) which catalyzes the production of mevalonate from 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA). Furthermore, to eliminate the post-transcriptional feedback inhibition, hmgr gene with a truncated N-terminal (thmgr), which lacks its N-terminal transmembrane sequence coding for membrane-binding activity, is used [96]. Additionally, the acetyl-CoA supply to the pathway can be improved, for example, via the overexpression of the ALD6 gene (encoding NADP-dependent aldehyde dehydrogenase) along with the introduction of a synthetic codon-optimized acetyl-CoA synthase mutant from Salmonella enterica [97].

IPP and DMAPP are then converted to 2,3 oxidosqualene, the precursor of ginsenoside aglycons. The yield of 2,3 oxidosqualene is improved by the overexpression of squalene synthase (SQS)- and squalene epoxidase (SQE)-encoding genes. To minimize the utilization of 2,3 oxidosqualene by competing yeasts in the native ergosterol pathway, downregulation of the Erg7/LS (lanosterol synthase)-encoding gene is conducted. The subsequent conversion of 2,3 oxidosqualene into dammarenediol and protopanaxadiol/protopanaxatriol was facilitated by the introduction of dammarenediol II synthase and cytochrome P450 (CYP) [protopanaxadiol synthase (CYP716A47) and protopanaxatriol synthase (CYP716A53v2)] genes from P. ginseng and NADPH-cytochrome P450 reductase (CPR) from Arabidopsis thaliana or P. ginseng [92, 98, 99].

Finally, the addition of sugar moieties to PPD and PPT aglycons was facilitated by the introduction of glycosyltransferases-encoding genes. UDP-glycosyltransferases from P. ginseng, PgUGT45 and PgUGT74AE2, catalyze the transfer of a glucose moiety from UDP-glucose (UDP-Glc) to the C3 hydroxyl groups of PPD to form Rh2, whereas PgUGT29 and PgUGT94Q2 catalyze the transfer of a glucose moiety from UDP-Glc to Rh2 to form Rg3 [100, 101]. Meanwhile, PgUGT100 and PgUGT1 transfer a glucose moiety from UDP-glucose (UDP-Glc) to the C-6 and C-20 hydroxyl groups of PPT to form Rh1 and F1, respectively [102]. The application of the general strategies along with other strategy such as protein engineering resulted in the production of minor ginsenoside in Gram-scale quantities (Table 4).

Table 4 Production of Minor Ginsenosides Using Different Yeast Cell Factories

Host Microorganism Strategies Products Yield
S. cerevisiae strain BY4742 (MATα, his3Δ1, leu2Δ0, lys2Δ0, ura3Δ0) Enhancing MVA pathway of S. cerevisiae strain BY4742 Rh2, Rg3 Rg3 (3.49 ± 0.14 μmol/g dry cell weight) and Rh2 (1.45 ± 0.27 μmol/g dry cell weight) were produced after 6 days [103]
Introducing dammarenediol synthase gene and protopanaxadiol synthase genes and UDP-glycosyltransferase genes from P. ginseng (PgUGT45 and PgUGT29) into S. cerevisiae strain BY4742
S. cerevisiae strain ZW-PPD-B (PPD-producing yeast strain) [103] Introducing cytochrome P450 CYP716A53v2, cytochrome P450 reductase PgCPR1, and UDP-glycosyltransferase PgUGT100 genes into S. cerevisiae strain ZW-PPD-B to construct an Rh1-producing yeast strain Rh1, F1 Rh1 (92.8 ± 12.5 mg/L) and F1 (42.1 ± 3.2 mg/L) were produced after 6 days [102]
Introducing CYP716A53v2, PgCPR1, and PgUGT1 genes into S. cerevisiae strain ZW-PPD-B to construct an F1-producing yeast strain
S. cerevisiae strain ZD-PPD-016 (PPD-producing yeast strain) [96] Conducting site-directed mutagenesis and iterative saturation mutagenesis to S. cerevisiae glycosyltransferase-encoding gene (UGT51) Rh2 Rh2 (300 mg/L) was produced via a 5 L fed-batch fermentation [104]
Introducing mutant UGT51 gene into S. cerevisiae strain ZD-PPD-016 to construct an Rh2-producing yeast strain
S. cerevisiae strain BY4742 (MATα, his3Δ1, leu2Δ0, lys2Δ0, ura3Δ0) Modular engineering of the MVA pathway Rh2 Rh2 (2.25 g/L) was produced via a 10 L fed-batch fermentation [105]
Optimizing the expression level of cytochrome P450
Increasing the copy number of UDP-glycosyltransferase-encoding gene (UGTPg45 from P. ginseng) and engineering its promoter to increase the expression level of the enzyme
Conducting in vivo directed evolution to the in vivo activity of UGTPg45 in yeast
Y. lipolytica ATCC 201249 Overexpressing key genes in the MVA pathway CK CK (161.8 mg/L) was produced via 5 L fed-batch fermentation [106]
Constructing fusion of cytochrome NADPH-P450 reductase, and P450 monooxygenase (PPDS), and introducing the gene into Y. lipolytica ATCC 201249
Introducing UDP-glycosyltransferase genes UGT1 from P. ginseng into Y. lipolytica ATCC 201249

MVA: Mevalonic acid.

Conclusion and future perspectives

Due to their medical importance and high economical value, investigations on the mass-production methods of minor ginsenosides, especially those that involve microorganisms and their enzymes, have garnered great interest. Various microorganisms have been reported to be able to convert major ginsenosides into minor ginsenosides, including GRAS microorganisms which would be suitable for the food and drugs industries. To date the highest yield of minor ginsenosides has been obtained via the biotransformation of major ginsenosides using recombinant β-glucosidase expressed in E. coli systems. However, E. coli is considered as unsafe, and is an inedible bacteria, which would limit its application in the nutraceutical and pharmaceutical industries. Thus, the improvements of recombinant β-glucosidase production in GRAS strains such as C. glutamicum and L. lactis, for example, through the optimization of growth condition, and media, as well as genetic engineering, are needed.

Finally, the costly, time-consuming, and labor-intensive process of producing ginseng extract as substrates for biotransformation, has driven the development of more sustainable ways to produce minor ginsenosides, especially via biosynthesis in yeast cell factories. The introduction of ginsenosides biosynthesis genes from P. ginseng coupled with the optimization of yeast native pathways has led to the successful production of minor ginsenosides. Further optimization using protein engineering, synthetic biology, and metabolic engineering approaches, as well as the development of efficient fermentation strategies are critical to unleash the full potential of yeast cell factories to mass produce minor ginsenosides.

Competing interests

The authors declare that they have no competing interests.

References

  1. Guo D, Cheng L, Zhang Y, Zheng H, Ma H, et al. An improved method for the preparation of Ginsenoside Rg5 from ginseng fibrous root powder. Heliyon 2019;5:e02694. [PMID: 31687518 DOI: 10.1016/j.heliyon.2019.e02694]
  2. Li X, Sun L, Zhao D. Current status and problem-solving strategies for ginseng industry. Chin J Integr Med 2019;25:883-6. [PMID: 31630359 DOI: 10.1007/s11655-019-3046-2]
  3. Baeg I-H, So S-H. The world ginseng market and the ginseng (Korea). J Ginseng Res 2013;37:1-7. [PMID: 23717152 DOI: 10.5142/jgr.2013.37.1]
  4. Choi J, Kim T-H, Choi T-Y, Lee MS. Ginseng for health care: a systematic review of randomized controlled trials in Korean literature. PLoS One 2013;8:e59978. [PMID: 23560064 DOI: 10.1371/journal.pone.0059978]
  5. DesRochers N, Walsh PJ, Renaud BJ, Seifert AK, Yeung K-CK, et al. Metabolomic profiling of fungal pathogens responsible for root rot in American Ginseng. Metabolites 2020;10:35. [PMID: 31947697 DOI: 10.3390/metabo10010035]
  6. Xu J, Chu Y, Liao B, Xiao S, Yin Q, et al. Panax ginseng genome examination for ginsenoside biosynthesis. Gigascience 2017;6:1-15. [PMID: 29048480 DOI: 10.1093/gigascience/gix093]
  7. Yu JS, Roh H-S, Baek K-H, Lee S, Kim S, et al. Bioactivity-guided isolation of ginsenosides from Korean red Ginseng with cytotoxic activity against human lung adenocarcinoma cells. J Ginseng Res 2018;42:562-70. [PMID: 30337817 DOI: 10.1016/j.jgr.2018.02.004]
  8. Wu T, Kwaku OR, Li H-Z, Yang C-R, Ge L-J, et al. Sense ginsenosides from ginsengs: structure-activity relationship in autophagy. Nat Prod Commun 2019;14:1934578X19858223. [DOI: 10.1177/1934578X19858223]
  9. Yang L, Zou H, Gao Y, Luo J, Xie X, et al. Insights into gastrointestinal microbiota-generated ginsenoside metabolites and their bioactivities. Drug Metab Rev 2020;52:125-38. [PMID: 31984805 DOI: 10.1080/03602532.2020.1714645]
  10. Park C-S, Yoo M-H, Noh K-H, Oh D-K. Biotransformation of ginsenosides by hydrolyzing the sugar moieties of ginsenosides using microbial glycosidases. Appl Microbiol Biotechnol 2010;87:9-19. [PMID: 20376631 DOI: 10.1007/s00253-010-2567-6]
  11. Zhou P, Xie W, He S, Sun Y, Meng X, et al. Ginsenoside Rb1 as an anti-diabetic agent and its underlying mechanism analysis. Cells 2019;8:204. [PMID: 30823412 DOI: 10.3390/cells8030204]
  12. Ahmed T, Raza SH, Maryam A, Setzer WN, Braidy N, et al. Ginsenoside Rb1 as a neuroprotective agent: a review. Brain Res Bull 2016;125:30-43. [PMID: 27060612 DOI: 10.1016/j.brainresbull.2016.04.002]
  13. Huang Q, Wang T, Wang H. Ginsenoside Rb2 enhances the anti-inflammatory effect of ω-3 fatty acid in LPS-stimulated RAW264.7 macrophages by upregulating GPR120 expression. Acta Pharmacol Sin 2017;38:192-200. [PMID: 28017961 DOI: 10.1038/aps.2016.135]
  14. Kim DH, Kim DW, Jung BH, Lee JH, Lee H, et al. Ginsenoside Rb2 suppresses the glutamate-mediated oxidative stress and neuronal cell death in HT22 cells. J Ginseng Res 2019;43:326-34. [PMID: 30976171 DOI: 10.1016/j.jgr.2018.12.002]
  15. Oh Y, Lim H-W, Park KH, Huang Y-H, Yoon J-Y, et al. Ginsenoside Rc protects against UVB induced photooxidative damage in epidermal keratinocytes. Mol Med Rep 2017;16:2907-14. [PMID: 28713942 DOI: 10.3892/mmr.2017.6943]
  16. Yu T, Yang Y, Kwak Y-S, Song GG, Kim M-Y, et al. Ginsenoside Rc from Panax ginseng exerts anti-inflammatory activity by targeting TANK-binding kinase 1/interferon regulatory factor-3 and p38/ATF-2. J Ginseng Res 2017;41:127-33. [PMID: 28413316 DOI: 10.1016/j.jgr.2016.02.001]
  17. Cong L, Chen W. Neuroprotective effect of ginsenoside Rd in spinal cord injury rats. Basic Clin Pharmacol Toxicol 2016;119:193-201. [PMID: 26833867 DOI: 10.1111/bcpt.12562]
  18. Zhang E, Shi H, Yang L, Wu X, Wang Z. Ginsenoside Rd regulates the Akt/mTOR/p70S6K signaling cascade and suppresses angiogenesis and breast tumor growth. Oncol Rep 2017;38:359-67. [PMID: 28534996 DOI: 10.3892/or.2017.5652]
  19. Jeong WI, Kim MH, Jeong JM, Kim SY, Kim SC, inventors; INTELLIGENT SYNTHETIC BIOLOGY CENTER, assignee. Composition for preventing or treating liver cancer containing ginsenoside F2. United States patent US 9,943,534. 2018 Apr 17.
  20. Liu D, Pan F, Liu J, Wang Y, Zhang T, et al. Individual and combined antioxidant effects of ginsenoside F2 and cyanidin-3-O-glucoside in human embryonic kidney 293 cells. RSC Adv 2016;6:81092-100. [DOI: 10.1039/C6RA14831J]
  21. Park S-H, Seo W, Eun HS, Kim SY, Jo E, et al. Protective effects of ginsenoside F2 on 12-O-tetradecanoylphorbol-13-acetate-induced skin inflammation in mice. Biochem Biophys Res Commun 2016;478:1713-9. [PMID: 27596969 DOI: 10.1016/j.bbrc.2016.09.009]
  22. Kim S-J, Jang JY, Kim E-J, Cho EK, Ahn D-G, et al. Ginsenoside Rg3 restores hepatitis C virus-induced aberrant mitochondrial dynamics and inhibits virus propagation. Hepatology 2017;66:758-71. [PMID: 28329914 DOI: 10.1002/hep.29177]
  23. Sun M, Ye Y, Xiao L, Duan X, Zhang Y, et al. Anticancer effects of ginsenoside Rg3 (Review). Int J Mol Med 2017;39:507-18. [PMID: 28098857 DOI: 10.3892/ijmm.2017.2857]
  24. You Z, Yao Q, Shen J, Gu Z, Xu H, et al. Antidepressant-like effects of ginsenoside Rg3 in mice via activation of the hippocampal BDNF signaling cascade. J Nat Med 2017;71:367-79. [PMID: 28013484 DOI: 10.1007/s11418-016-1066-1]
  25. Meng X, Luo Y, Liang T, Wang M, Zhao J, et al. Gypenoside XVII enhances lysosome biogenesis and autophagy flux and accelerates autophagic clearance of amyloid-β through TFEB activation. J Alzheimer’s Dis 2016;52:1135-50. [PMID: 27060963 DOI: 10.3233/JAD-160096]
  26. Yang K, Zhang H, Luo Y, Zhang J, Wang M, Liao P, et al. Gypenoside XVII prevents atherosclerosis by attenuating endothelial apoptosis and oxidative stress: Insight into the ERα-mediated PI3K/Akt pathway. Int J Mol Sci 2017;18:77. [PMID: 28208754 DOI: 10.3390/ijms18020077]
  27. Cui C-H, Kim DJ, Jung S-C, Kim S-C, Im W-T. Enhanced production of gypenoside LXXV using a Novel Ginsenoside-transforming β-glucosidase from Ginseng-Cultivating Soil Bacteria and its anti-cancer property. Molecules 2017;22:844. [PMID: 28534845 DOI: 10.3390/molecules22050844]
  28. Park S, Ko E, Lee HJ, Song Y, Cui C-H, et al. Gypenoside LXXV promotes cutaneous wound healing in vivo by enhancing connective tissue growth factor levels via the glucocorticoid receptor pathway. Molecules 2019;24:1595. [PMID: 31018484 DOI: 10.3390/molecules24081595]
  29. Kang S, Im K, Kim G, Min H. Antiviral activity of 20(R)-ginsenoside Rh2 against murine gammaherpesvirus. J Ginseng Res 2017;41:496-502. [PMID: 29021696 DOI: 10.1016/j.jgr.2016.08.010]
  30. Chen F, Sun Y, Zheng S-L, Qin Y, Julian McClements D, et al. Antitumor and immunomodulatory effects of ginsenoside Rh2 and its octyl ester derivative in H22 tumor-bearing mice. J Funct Foods 2017;32:382-90. [DOI: 10.1016/j.jff.2017.03.013]
  31. Yang Z, Zhao T, Liu H, Zhang L. Ginsenoside Rh2 inhibits hepatocellular carcinoma through β-catenin and autophagy. Sci Rep 2016;6:19383. [PMID: 26783250 DOI: 10.1038/srep19383]
  32. Ko E, Park S, Lee HJ, Cui C-H, Hou J, et al. Ginsenoside Rh2 ameliorates atopic dermatitis in NC/Nga mice by suppressing NF-kappaB-mediated thymic stromal lymphopoietin expression and T helper type 2 differentiation. Int J Mol Sci 2019:20:6111. [PMID: 31817146 DOI: 10.3390/ijms20246111]
  33. Zeng X, Hu K, Chen L, Zhou L, Luo W, et al. The effects of ginsenoside compound K against epilepsy by enhancing the γ-aminobutyric acid signaling pathway. Front Pharmacol 2018:9;1020. [PMID: 30254585 DOI: 10.3389/fphar.2018.01020]
  34. Kim EH, Kim W. An insight into ginsenoside metabolite compound K as a potential tool for skin disorder. In: Shin S-H, editor. Evid Based Complement Alternat Med 2018;2018:8075870. [PMID: 30046346 DOI: 10.1155/2018/8075870]
  35. Chen L, Meng Y, Sun Q, Zhang Z, Guo X, et al. Ginsenoside compound K sensitizes human colon cancer cells to TRAIL-induced apoptosis via autophagy-dependent and -independent DR5 upregulation. Cell Death Dis 2016;7:e2334. [PMID: 27512955 DOI: 10.1038/cddis.2016.234]
  36. Shi Y, Wan X, Shao N, Ye R, Zhang N, Zhang Y. Protective and antiangiopathy effects of ginsenoside Re against diabetes mellitus via the activation of p38 MAPK, ERK1/2 and JNK signaling. Mol Med Rep 2016;14:4849-56. [PMID: 27748921 DOI: 10.3892/mmr.2016.5821]
  37. Chen R-C, Wang J, Yang L, Sun G-B, Sun X-B. Protective effects of ginsenoside Re on lipopolysaccharide-induced cardiac dysfunction in mice. Food Funct 2016;7:2278-87. [PMID: 27074714 DOI: 10.1039/c5fo01357g]
  38. Wang Z, Li Y, Han X, Sun Y, Zhang L, et al. Kidney protection effect of ginsenoside Re and its underlying mechanisms on cisplatin-induced kidney injury. Cell Physiol Biochem 2018;48:2219-29. [PMID: 30110677 DOI: 10.1159/000492562]
  39. Du Y, Fu M, Wang YT, Dong Z. Neuroprotective effects of ginsenoside Rf on amyloid-beta-induced neurotoxicity in vitro and in vivo. J Alzheimers Dis 2018;64:309-22. [PMID: 29865080 DOI: 10.3233/JAD-180251]
  40. Ahn S, Siddiqi MH, Aceituno VC, Simu SY, Yang DC. Suppression of MAPKs/NF-κB activation induces intestinal anti-inflammatory action of ginsenoside Rf in HT-29 and RAW264.7 cells. Immunol Invest 2016;45:439-49. [PMID: 27224660 DOI: 10.3109/08820139.2016.1168830]
  41. Zhou T, Zu G, Zhang X, Wang X, Li S, et al. Neuroprotective effects of ginsenoside Rg1 through the Wnt/β-catenin signaling pathway in both in vivo and in vitro models of Parkinson’s disease. Neuropharmacology 2016;101:480-9. [PMID: 26525190 DOI: 10.1016/j.neuropharm.2015.10.024]
  42. Gao Y, Chu S, Zhang Z, Chen N. Hepataprotective effects of ginsenoside Rg1 – a review. J Ethnopharmacol 2017;206:178-83. [PMID: 28427912 DOI: 10.1016/j.jep.2017.04.012]
  43. Ren Y, Wang J-L, Zhang X, Wang H, Ye Y, et al. Antidepressant-like effects of ginsenoside Rg2 in a chronic mild stress model of depression. Brain Res Bull 2017;134:211-9. [PMID: 28842305 DOI: 10.1016/j.brainresbull.2017.08.009]
  44. Jin Y, Baek N, Back S, Myung C-S, Heo K-S. Inhibitory effect of ginsenosides Rh1 and Rg2 on oxidative stress in LPS-Stimulated RAW 264.7 cells. J Bacteriol Virol 2018;48:156-65. [DOI: 10.4167/jbv.2018.48.4.156]
  45. Chung YH, Jeong SA, Choi HS, Ro S, Lee JS, et al. Protective effects of ginsenoside Rg2 and astaxanthin mixture against UVB-induced DNA damage. Anim Cells Syst (Seoul) 2018;22:400-6. [PMID: 30533262 DOI: 10.1080/19768354.2018.1523806]
  46. Tam DNH, Truong DH, Nguyen TTH, Quynh LN, Tran L, et al. Ginsenoside Rh1: a systematic review of its pharmacological properties. Planta Med 2018;84:139-52. [PMID: 29329463 DOI: 10.1055/s-0043-124087]
  47. Lee W, Cho S-H, Kim J-E, Lee C, Lee J-H, et al. Suppressive effects of ginsenoside Rh1 on HMGB1-mediated septic responses. Am J Chin Med 2019;47:119-33. [PMID: 30630344 DOI: 10.1142/S0192415X1950006X]
  48. Moon SS, Lee JH, Mathiyalagan R, Kim JY, Yang UD, et al. Synthesis of a novel α-glucosyl ginsenoside F1 by cyclodextrin glcanotransferase and its in vitro cosmetic applications. Biomolecules 2018;8:142. [PMID: 30423825 DOI: 10.3390/biom8040142]
  49. Kim SC, Kim HS, inventors; INTELLIGENT SYNTHETIC BIOLOGY CENTER, assignee. Composition for enhancing immunity including ginsenoside f1 as an active ingredient. United States patent application US 15/461,722. 2017 Dec 28.
  50. Lee DG, Lee JS, Kim K-T, Kim HY, Lee S. Analysis of major ginsenosides in various ginseng samples. J Appl Biol Chem 2019;62:87-91. [DOI: 10.3839/jabc.2019.013]
  51. Du J, Cui C-H, Park SC, Kim J-K, Yu H-S, et al. Identification and characterization of a ginsenoside-transforming β-glucosidase from Pseudonocardia sp. Gsoil 1536 and its application for enhanced production of minor ginsenoside Rg2(S). PLoS One 2014;9:e96914. [PMID: 24911166 DOI: 10.1371/journal.pone.0096914]
  52. Leung KW, Wong AS-T. Pharmacology of ginsenosides: a literature review. Chin Med 2010;5:20. [PMID: 20537195 DOI: 10.1186/1749-8546-5-20]
  53. Cui L, Wu S, Zhao C, Yin C. Microbial conversion of major ginsenosides in ginseng total saponins by Platycodon grandiflorum endophytes. J Ginseng Res 2016;40:366-74. [PMID: 27746689 DOI: 10.1016/j.jgr.2015.11.004]
  54. Wang Y, Choi K-D, Yu H, Jin F, Im W-T. Production of ginsenoside F1 using commercial enzyme cellulase KN. J Ginseng Res 2016;40:121-6. [PMID: 27158232 DOI: 10.1016/j.jgr.2015.06.003]
  55. Biswas T, Mathur AK, Mathur A. A literature update elucidating production of Panax ginsenosides with a special focus on strategies enriching the anti-neoplastic minor ginsenosides in ginseng preparations. Appl Microbiol Biotechnol 2017;101:4009-32. [PMID: 28411325 DOI: 10.1007/s00253-017-8279-4]
  56. Cui C, Jeon B-M, Fu Y, Im W-T, Kim S-C. High-density immobilization of a ginsenoside-transforming β-glucosidase for enhanced food-grade production of minor ginsenosides. Appl Microbiol Biotechnol 2019;103:7003-15. [PMID: 31289903 DOI: 10.1007/s00253-019-09951-4]
  57. Shin K-C, Oh D-K. Classification of glycosidases that hydrolyze the specific positions and types of sugar moieties in ginsenosides. Crit Rev Biotechnol 2016;36:1036-49. [PMID: 26383974 DOI: 10.3109/07388551.2015.1083942]
  58. Li X, Yao F, Fan H, Li K, Sun L, et al. Intraconversion of polar ginsenosides, their transformation into less-polar ginsenosides, and ginsenoside acetylation in ginseng flowers upon baking and steaming. Molecules 2018:23:759. [PMID: 29587462 DOI: 10.3390/molecules23040759]
  59. Shin J-H, Park YJ, Kim W, Kim D-O, Kim B-Y, et al. Change of ginsenoside profiles in processed ginseng by drying, steaming, and puffing. J Microbiol Biotechnol 2019;29:222-9. [PMID: 30609886 DOI: 10.4014/jmb.1809.09056]
  60. Lu C, Yin Y. Optimum conversion of major ginsenoside Rb1 to minor ginsenoside Rg3 (S) by pulsed electric field-assisted acid hydrolysis treatment. Open Chem 2018;16:283-90. [DOI: 10.1515/chem-2018-0031]
  61. Zheng M, Xu F, Li Y, Xi X, Cui X, et al. Study on transformation of ginsenosides in different methods. In: Mahady GB, editor. Biomed Res Int 2017;2017:8601027. [PMID: 29387726 DOI: 10.1155/2017/8601027]
  62. Geraldi A, Fatimah N, Cui C-H, Nguyen TT, et al. Enzymatic biotransformation of ginsenoside Rb1 by recombinant β-glucosidase of bacterial isolates from Indonesia. Biocatal Agric Biotechnol 2020;23:101449. [DOI: 10.1016/j.bcab.2019.101449]
  63. Yang X-D, Yang Y-Y, Ouyang D-S, Yang G-P. A review of biotransformation and pharmacology of ginsenoside compound K. Fitoterapiat 2015;100:208-20. [PMID: 25449425 DOI: 10.1016/j.fitote.2014.11.019]
  64. Khan Chowdhury MDE, Jeon J, Ok Rim S, Park Y-H, Kyu Lee S, et al. Composition, diversity and bioactivity of culturable bacterial endophytes in mountain-cultivated ginseng in Korea. Sci Rep 2017;7:10098. [PMID: 28855721 DOI: 10.1038/s41598-017-10280-7]
  65. Fu Y. Biotransformation of ginsenoside Rb1 to Gyp-XVII and minor ginsenoside Rg3 by endophytic bacterium Flavobacterium sp. GE 32 isolated from Panax ginseng. Lett Appl Microbiol 2019;68:134-41. [PMID: 30362617 DOI: 10.1111/lam.13090]
  66. Fu Y, Yin Z-H, Yin C-Y. Biotransformation of ginsenoside Rb1 to ginsenoside Rg3 by endophytic bacterium Burkholderia sp. GE 17-7 isolated from Panax ginseng. J Appl Microbiol 2017;122:1579-85. [PMID: 28256039 DOI: 10.1111/jam.13435]
  67. Fu Y, Yin Z-H, Wu L-P, Yin C-R. Biotransformation of ginsenoside Rb1 to ginsenoside C-K by endophytic fungus Arthrinium sp. GE 17-18 isolated from Panax ginseng. Lett Appl Microbiol 2016;63:196-201. [PMID: 27316666 DOI: 10.1111/lam.12606]
  68. Yu S, Zhou X, Li F, Xu C, Zheng F, et al. Microbial transformation of ginsenoside Rb1, Re and Rg1 and its contribution to the improved anti-inflammatory activity of ginseng. Sci Rep 2017;7:138. [PMID: 28273939 DOI: 10.1038/s41598-017-00262-0]
  69. Ku S. Finding and producing probiotic glycosylases for the biocatalysis of ginsenosides: a mini review. Molecules 2016:21:645. [PMID: 27196878 DOI: 10.3390/molecules21050645]
  70. Lee S, Lee Y-H, Park J-M, Bai D-H, Jang JK, et al. Bioconversion of ginsenosides from red ginseng extract using Candida allociferrii JNO301 isolated from Meju. Mycobiology 2014;42:368-75. [PMID: 25606009 DOI: 10.5941/MYCO.2014.42.4.368]
  71. Park B, Hwang H, Lee J, Sohn S-O, Lee SH, et al. Evaluation of ginsenoside bioconversion of lactic acid bacteria isolated from kimchi. J Ginseng Res 2017;41:524-30. [PMID: 29021699 DOI: 10.1016/j.jgr.2016.10.003]
  72. Ku S, You HJ, Park MS, Ji GE. Whole-cell biocatalysis for producing ginsenoside Rd from Rb1 using Lactobacillus rhamnosus GG. J Microbiol Biotechnol 2016;26:1206-15. [PMID: 27012233 DOI: 10.4014/jmb.1601.01002]
  73. Liu Z, Li J-X, Wang C-Z, Zhang D-L, Wen X, et al. Microbial conversion of protopanaxadiol-type ginsenosides by the edible and medicinal mushroom schizophyllum commune: a green biotransformation strategy. ACS Omega 2019;4:13114-23. [PMID: 31460439 DOI: 10.1021/acsomega.9b01001]
  74. Kim B-N, Yeom S-J, Kim Y-S, Oh D-K. Characterization of a β-glucosidase from Sulfolobus solfataricus for isoflavone glycosides. Biotechnol Lett 2012;34:125-9. [PMID: 21898127 DOI: 10.1007/s10529-011-0739-9]
  75. Hong H, Cui C-H, Kim J-K, Jin F-X, Kim S-C, et al. Enzymatic biotransformation of ginsenoside Rb1 and gypenoside XVII into ginsenosides Rd and F2 by recombinant β-glucosidase from Flavobacterium johnsoniae. J Ginseng Res 2012;36:418-24. [PMID: 23717145 DOI: 10.5142/jgr.2012.36.4.418]
  76. Eom SJ, Kim K-T, Paik H-D. Microbial bioconversion of ginsenosides in Panax ginseng and their improved bioactivities. Food Rev Int 2018;34:698-712. [DOI: 10.1080/87559129.2018.1424183]
  77. Siddiqi ZM, Srinivasan S, Park YH, Im W-T. Exploration and characterization of novel glycoside hydrolases from the whole genome of Lactobacillus ginsenosidimutans and enriched production of minor ginsenoside Rg3(S) by a recombinant enzymatic process. Biomolecules 2020;10:288. [PMID: 32059542 DOI: 10.3390/biom10020288]
  78. Rosano GL, Morales ES, Ceccarelli EA. New tools for recombinant protein production in Escherichia coli: a 5-year update. Protein Sci 2019;28:1412-22. [PMID: 31219641 DOI: 10.1002/pro.3668]
  79. Hayat SMG, Farahani N, Golichenari B, Sahebkar A. Recombinant protein expression in Escherichia coli (E. coli): What we need to know. Curr Pharm Des 2018;24:718-25. [PMID: 29384059 DOI: 10.2174/1381612824666180131121940]
  80. Taguchi S, Ooi T, Mizuno K, Matsusaki H. Advances and needs for endotoxin-free production strains. Appl Microbiol Biotechnol 2015;99:9349-60. [PMID: 26362682 DOI: 10.1007/s00253-015-6947-9]
  81. Siddiqi MZ, Cui C-H, Park S-K, Han NS, Kim S-C, et al. Comparative analysis of the expression level of recombinant ginsenoside-transforming β-glucosidase in GRAS hosts and mass production of the ginsenoside Rh2-Mix. PLoS One 2017;12:e0176098. [PMID: 28423055 DOI: 10.1371/journal.pone.0176098]
  82. Liu X, Yang Y, Zhang W, Sun Y, Peng F, et al. Expression of recombinant protein using Corynebacterium glutamicum: progress, challenges and applications. Crit Rev Biotechnol 2016;36:652-64. [PMID: 25714007 DOI: 10.3109/07388551.2015.1004519]
  83. Siddiqi MZ, Shafi SM, Im W-T. Complete genome sequencing of Arachidicoccus ginsenosidimutans sp. nov., and its application for production of minor ginsenosides by finding a novel ginsenoside-transforming β-glucosidase. RSC Adv 2017;7:46745-59. [DOI: 10.1039/C7RA02612A]
  84. Quan L-H, Min J-W, Sathiyamoorthy S, Yang D-U, Kim Y-J, et al. Biotransformation of ginsenosides Re and Rg1 into ginsenosides Rg2 and Rh1 by recombinant β-glucosidase. Biotechnol Lett 2012;34:913-7. [PMID: 22261865 DOI: 10.1007/s10529-012-0849-z]
  85. Quan L-H, Min J-W, Jin Y, Wang C, Kim Y-J, et al. Enzymatic biotransformation of ginsenoside Rb1 to compound K by recombinant β-glucosidase from Microbacterium esteraromaticum. J Agric Food Chem 2012;60:3776-81. [PMID: 22428991 DOI: 10.1021/jf300186a]
  86. Zhong F-L, Dong W-W, Wu S, Jiang J, Yang D-C, et al. Biotransformation of gypenoside XVII to compound K by a recombinant β-glucosidase. Biotechnol Lett 2016;38:1187-93. [PMID: 27060008 DOI: 10.1007/s10529-016-2094-3]
  87. Pei J, Wu T, Yao T, Zhao L, Ding G, et al. Biotransformation of ginsenosides Re and Rg1 into Rg2 and Rh1 by thermostable β-glucosidase from Thermotoga thermarum. Chem Nat Compd 2017;53:472-7. [DOI: 10.1007/s10600-017-2025-0]
  88. Shin K-C, Kim T-H, Choi J-H, Oh D-K. Complete biotransformation of protopanaxadiol-type ginsenosides to 20-O-β-glucopyranosyl-20(S)-protopanaxadiol using a novel and thermostable β-glucosidase. J Agric Food Chem 2018;66:2822-9. [PMID: 29468877 DOI: 10.1021/acs.jafc.7b06108]
  89. Bi Y-F, Wang X-Z, Jiang S, Liu J-S, Zheng M-Z, et al. Enzymatic transformation of ginsenosides Re, Rg1, and Rf to ginsenosides Rg2 and aglycon PPT by using β-glucosidase from Thermotoga neapolitana. Biotechnol Lett 2019;41:613-23. [PMID: 29468877 DOI: 10.1021/acs.jafc.7b06108]
  90. Li L, Lee SJ, Yuan QP, Im WT, Kim SC, et al. Production of bioactive ginsenoside Rg3(S) and compound K using recombinant Lactococcus lactis. J Ginseng Res 2018;42:412-8. [PMID: 30337801 DOI: 10.1016/j.jgr.2017.04.007]
  91. Li L, Shin S-Y, Lee SJ, Moon JS, Im WT, et al. Production of ginsenoside F2 by using Lactococcus lactis with enhanced expression of β-Glucosidase gene from Paenibacillus mucilaginosus. J Agric Food Chem 2016;64:2506-12. [PMID: 26494255 DOI: 10.1021/acs.jafc.5b04098]
  92. Dai Z, Wang B, Liu Y, Shi M, Wang D, et al. Producing aglycons of ginsenosides in bakers’ yeast. Sci Rep 2014;4:3698. [PMID: 24424342 DOI: 10.1038/srep03698]
  93. Kim Y-J, Zhang D, Yang D-C. Biosynthesis and biotechnological production of ginsenosides. Biotechnol Adv 2015;33:717-35. [PMID: 25747290 DOI: 10.1016/j.biotechadv.2015.03.001]
  94. Dai Z, Liu Y, Guo J, Huang L, Zhang X. Yeast synthetic biology for high-value metabolites. FEMS Yeast Res 2015;15:1-11. [PMID: 25047863 DOI: 10.1111/1567-1364.12187]
  95. Chu LL, Montecillo JAV, Bae H. Recent Advances in the metabolic engineering of yeasts for ginsenoside biosynthesis. Front Bioeng Biotechnol 2020;8:139. [PMID: 32158753 DOI: 10.3389/fbioe.2020.00139]
  96. Dai Z, Liu Y, Zhang X, Shi M, Wang B, et al. Metabolic engineering of Saccharomyces cerevisiae for production of ginsenosides. Metab Eng 2013;20:146-56. [PMID: 24126082 DOI: 10.1016/j.ymben.2013.10.004]
  97. Zhao F, Bai P, Nan W, Li D, Zhang C, et al. A modular engineering strategy for high-level production of protopanaxadiol from ethanol by Saccharomyces cerevisiae. AIChE J 2019;65:866-74. [DOI: 10.1002/aic.16502]
  98. Han J-Y, Hwang H-S, Choi S-W, Kim H-J, Choi Y-E. Cytochrome P450 CYP716A53v2 catalyzes the formation of protopanaxatriol from protopanaxadiol during ginsenoside biosynthesis in Panax ginseng. Plant Cell Physiol 2012;53:1535-45. [PMID: 22875608 DOI: 10.1093/pcp/pcs106]
  99. Park S-B, Chun J-H, Ban Y-W, Han JY, Choi YE. Alteration of Panax ginseng saponin composition by overexpression and RNA interference of the protopanaxadiol 6-hydroxylase gene (CYP716A53v2). J Ginseng Res 2016;40:47-54. [PMID: 26843821 DOI: 10.1016/j.jgr.2015.04.010]
  100. Jung S-C, Kim W, Park SC, Jeong J, Park MK, et al. Two Ginseng UDP-glycosyltransferases synthesize ginsenoside Rg3 and Rd. Plant Cell Physiol 2014;55:2177-88. [PMID: 25320211 DOI: 10.1093/pcp/pcu147]
  101. Yang J-L, Hu Z-F, Zhang T-T, Gu A-D, Gong T, et al. Progress on the studies of the key enzymes of ginsenoside biosynthesis. Molecules 2018;23:589. [PMID: 29509695 DOI: 10.3390/molecules23030589]
  102. Wei W, Wang P, Wei Y, Liu Q, Yang C, et al. Characterization of Panax ginseng UDP-glycosyltransferases catalyzing protopanaxatriol and biosyntheses of bioactive ginsenosides F1 and Rh1 in metabolically engineered yeasts. Mol Plant 2015;8:1412-24. [PMID: 26032089 DOI: 10.1016/j.molp.2015.05.010]
  103. Wang P, Wei Y, Fan Y, Liu Q, Wei W, et al. Production of bioactive ginsenosides Rh2 and Rg3 by metabolically engineered yeasts. Metab Eng 2015;29:97-105. [PMID: 25769286 DOI: 10.1016/j.ymben.2015.03.003]
  104. Zhuang Y, Yang G-Y, Chen X, Liu Q, Zhang X, et al. Biosynthesis of plant-derived ginsenoside Rh2 in yeast via repurposing a key promiscuous microbial enzyme. Metab Eng 2017;42:25-32. [PMID: 28479190 DOI: 10.1016/j.ymben.2017.04.009]
  105. Wang P, Wei W, Ye W, Li X, Zhao W, et al. Synthesizing ginsenoside Rh2 in Saccharomyces cerevisiae cell factory at high-efficiency. Cell Discov 2019;5:5. [PMID: 30652026 DOI: 10.1038/s41421-018-0075-5]
  106. Li D, Wu Y, Zhang C, Sun J, Zhou Z, et al. Production of triterpene ginsenoside compound K in the non-conventional yeast Yarrowia lipolytica. J Agric Food Chem 2019;67:2581-8. [PMID: 30757901 DOI: 10.1021/acs.jafc.9b00009]