ArticlePDF Available

Biosynthesis of fraxetin from three different substrates using engineered Escherichia coli

Authors:

Abstract

Fraxetin, which is a simple coumarin, is a phytochemical present in medicinal plants, such as Fraxinus rhynchophylla, and Cortex Fraxini. In plants, it serves as a controller of iron homeostasis. The health-enhancing activities of fraxetin, such as anticancer, neuroprotective and antibacterial activities, are known. Scopoletin 8-hydroxylase (S8H) is a key enzyme involved in the synthesis of fraxetin from scopoletin. Scopoletin can be synthesized either from esculetin by O-methylation or from ferulic acid by feruloyl CoA 6′-hydroxylase (F6′H) and 4-coumaric acid CoA ligase (4CL). To enable fraxetin synthesis, the fraxetin biosynthesis pathway was introduced into Escherichia coli. Three distinct routes, from ferulic acid, esculetin, and scopoletin, were designed for the synthesis of fraxetin. In the first approach, E. coli strain harboring S8H was used and found to synthesize 84.8 μM fraxetin from 100 μM scopoletin. Two E. coli strains were used for the other two approaches because these approaches required at least two enzymatic reactions. Through this approach, 41.4 μM fraxetin was synthesized from 100 μM esculetin, while 33.3 μM fraxetin was synthesized from 100 μM ferulic acid.
Anetal. Appl Biol Chem (2020) 63:55
https://doi.org/10.1186/s13765-020-00543-9
NOTE
Biosynthesis offraxetin fromthree dierent
substrates using engineered Escherichia coli
Seung Hoon An, Gyu‑Sik Choi and Joong‑Hoon Ahn*
Abstract
Fraxetin, which is a simple coumarin, is a phytochemical present in medicinal plants, such as Fraxinus rhynchophylla,
and Cortex Fraxini. In plants, it serves as a controller of iron homeostasis. The health‑enhancing activities of fraxetin,
such as anticancer, neuroprotective and antibacterial activities, are known. Scopoletin 8‑hydroxylase (S8H) is a key
enzyme involved in the synthesis of fraxetin from scopoletin. Scopoletin can be synthesized either from esculetin
by O‑methylation or from ferulic acid by feruloyl CoA 6‑hydroxylase (F6H) and 4‑coumaric acid CoA ligase (4CL).
To enable fraxetin synthesis, the fraxetin biosynthesis pathway was introduced into Escherichia coli. Three distinct
routes, from ferulic acid, esculetin, and scopoletin, were designed for the synthesis of fraxetin. In the first approach,
E. coli strain harboring S8H was used and found to synthesize 84.8 μM fraxetin from 100 μM scopoletin. Two E. coli
strains were used for the other two approaches because these approaches required at least two enzymatic reactions.
Through this approach, 41.4 μM fraxetin was synthesized from 100 μM esculetin, while 33.3 μM fraxetin was synthe‑
sized from 100 μM ferulic acid.
Keywords: Coumarin, Fraxetin, Scopoletin 8‑hydroxylase
© The Author(s) 2020. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material
in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material
is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creat iveco
mmons .org/licen ses/by/4.0/.
Introduction
Coumarins (benzo-alpha-pyrones) were first isolated
from the tonka bean (Dipteryx odorata) in 1820. Since
then, their presence has been detected in various parts of
different plants, including the fruit (e.g., in Bael fruit or
Aegle marmelos), seed (e.g., in tonka beans or Calophyl-
lum inophyllum), root (e.g., in Ferulago campestris), and
leaf (e.g., Murraya paniculata) [1]. All coumarins have
a hydroxy or methoxy group at position 7. Scopoletin,
esculetin, umbelliferone, fraxetin, as well as their respec-
tive glycosides, are termed simple coumarins; they are
widespread in higher plants [2]. ese coumarins play
a pivotal role in protecting plants against pathogens [3];
furthermore, a simple coumarin, such as fraxetin, was
found to modulate vital physiological processes such as
iron homeostasis [4]. As naturally occurring phytochemi-
cals, coumarins possess health-enhancing properties,
including anticancer [1], neuroprotective [5], and anti-
bacterial properties [6].
Coumarins were synthesized from hydroxycinnamic
acids, such as p-coumaric acid, caffeic acid, and feru-
lic acid, in plants; p-coumaric acid, caffeic acid, and
ferulic acid resulted in the synthesis of umbelliferone,
esculetin, and scopoletin, respectively. e key enzyme
for coumarin biosynthesis was p-coumaroyl CoA
2-hydroxylase (C2H) or feruloyl CoA 6-hydroxylase
(F6H); the corresponding genes were cloned in Arabi-
dopsis thaliana [7], Ruta graveolens [8], Ipomoea bata-
tas [9], Manihot esculenta [10], Angelica decursiva [11],
and Peucedanum praeruptorum [12]. is enzyme is a
2-oxglutarate-dependent dioxygenase; the hydroxylation
of hydroxycinnamoyl-CoA resulted in the formation of a
pyrone ring.
Fraxetin belongs to the family of simple coumarins and
is synthesized from scopoletin by the hydroxylation of its
carbon at position 8. Fraxetin is involved in iron metab-
olism in plants [4, 13]. Similar to other phytochemicals,
fraxetin was found to exert beneficial effects in humans.
ese included antitumor [10, 14], neuroprotective [15],
Open Access
*Correspondence: jhahn@konkuk.ac.kr
Department of Bioscience and Biotechnology, Bio/Molecular Informatics
Center, Konkuk University, Seoul 05029, Republic of Korea
Page 2 of 6
Anetal. Appl Biol Chem (2020) 63:55
antihyperglycemic [16], and anti-inflammatory [17]
effects.
Since the metabolic pathway responsible for the syn-
thesis of simple coumarins is well established, these
compounds have been synthesized in E. coli. Scopole-
tin, esculetin, umbelliferone, skimming (umbelliferone
7-O-glucoside), and herniarin (7-O-methyl umbellifer-
one) were synthesized in E. coli [18, 19]. For the fraxetin
synthesis process in E. coli, two routes were postulated
(Fig.1). e first route involved the synthesis of escule-
tin from glucose, followed by the conversion of esculetin
into fraxetin by 7-O-methylation and 8-hydroxylation.
e second route started with the synthesis of scopoletin
from ferulic acid, followed by 8-hydroxylation. In the pre-
sent study, one coumarin, namely fraxetin, was synthe-
sized using E. coli via these two routes.
Materials andmethods
Plasmid construction
Reverse transcription polymerase chain reaction (RT-
PCR) was used to clone cDNA of scopoletin 8-hydroxy-
lase (S8H) from Arabidopsis thaliana (AtS8H; GenBank:
DQ446658.1). Two primers 5ʹ-aagaattcaATG GGT ATC
AAT TTC GAG GA-3ʹ and 5ʹ-aagcggccgcTCA CTC GGC
ACG TG-3ʹ were used (restriction sites for EcoRI and
NotI have been underlined). Additionally, the S8H
homologue from Oryza sativa (OsS8H; GenBank:
XM_026024461) was cloned by RT-PCR using two
primers: 5ʹ-aagaattcaATG CCG TCC GGC TAC GAC -3ʹ
and 5ʹ-aagcggccgcCTA ATC TAG ACT AGC GGC GG-3ʹ
(restriction sites for EcoRI and NotI have been under-
lined). AtS8H was digested using the EcoRI and NotI sites
and subcloned into pGEX 5X-3 (pG-AtS8H), pET-duet1
(pE-AtS8H), pRSF-duet1 (pR-AtS8H), and pCDF-duet1
(pC-AtS8H). OsS8H was subcloned into EcoRI/NotI sites
of pGEX 5X-3 (pG-OsS8H).
F6H2 from Ipomoea batatas (IbF6H2; GenBank :
AB636154) and 4CL (Os4CL; 4-coumarate: CoA ligase)
from O. sativa had been previously cloned using RT-PCR
[18]. F6H2 was first cloned into pET-duet1 (EcoRI/NotI)
using PCR and, then, Os4CL was subcloned into pET-
duet1 containing F6H2 to generate pE-IbF6H2-Os4CL
(NdeI/XhoI). Subsequently, Os4CL was re-amplified
with a forward primer, adding a NotI site and riboso-
mal-binding site(RBS), and a reverse primer, containing
a XhoI site. ereafter, Os4CL was subcloned into the
NotI/XhoI sites of pET-duet1 containing IbF6H2 to gen-
erate pE-IbF6H-Os4CL controlled by a single promoter
(operon). e IbF6H-Os4CL operon was subcloned into
pGEX 5X-3 (EcoRI/XhoI).
POMT7 (flavone 7-O-methyltransferase) [20] and
POMT9 from Populus deltoids [21] and ROMT9 (flavo-
noid 3-O-methyltransferase) from O. sativa [22] have
also been cloned previously. ese genes were subcloned
into pGEX 5X-3 vector.
Production andanalysis ofmetabolites
For the synthesis of fraxetin from scopoletin, an over-
night culture of an E. coli transformant containing
pG-OsS8H, pG-AtS8H, pC-AtS8H, pE-AtS8H, or pR-
AtS8H was inoculated into fresh LB medium contain-
ing 50 μg/mL ampicillin and grown at 37 °C until the
OD600 reached 0.8; following this, isopropyl β-D-1-
thiogalactopyranoside (IPTG) was added to the medium
at a final concentration of 0.1mM or 1mM and incu-
bated at 18°C for 16h. Cells were harvested and the cell
concentration was adjusted to an OD600 of 3.0. e cells
were resuspended in M9 medium containing 2% glucose,
ampicillin (50μg/mL), and either 0.1mM or 1mM IPTG
POMT7AtS8H
Os4CLIbF6’H AtS8H
Esculetin
HO
HO
OO
Ferulic acid
OH
O
HO
OCH3
Feruloyl-CoA
OH
SCoA
HO
OCH
3
OH
SCoA
HO
OCH
3
OH
HO
H
3
CO
OO
Scopoletin
Fraxetin
HO
H3CO
OO
OH
Fraxetin
HO
H3CO
OO
OH
HO
H
3
CO
OO
Scopoletin
a
b
Fig. 1 Biosynthetic pathways of fraxetin from esculetin (a) and ferulic acid (b). POMT7 is an O‑methyltransferase, which converts esculetin into
scopoletin. AtS8H is a scopoletin 8‑hydroxylase. Os4CL catalyzes the attachment of CoA to ferulic acid. IbF6H encodes feruloyl CoA 6‑hydroxylase
(F6H), which converts feruloyl CoA into scopoletin
Page 3 of 6
Anetal. Appl Biol Chem (2020) 63:55
in a test tube. A total concentration of 100μM of the
substrate (esculetin, isoscopoletin, scopoletin, or scopar-
one) was added, and the resulting culture was incubated
at 30°C for 24h. An E. coli transformant containing the
pG-AtS8H construct was employed to determine the
substrate (scopoletin) concentration. e cell concentra-
tion was adjusted to an OD600 of 3.0. e substrate was
added to the appropriate M9 medium at 0.1, 0.2, 0.3, or
0.5mM. e reaction culture was incubated at 30°C for
24h.
e E. coli transformant harboring ROMT9 was used to
methylate esculetin to scopoletin, isoscopoletin, and sco-
poletin. ree reaction products were purified using thin
layer chromatography (silica gel 60 F254, Millipore). A
mixture of benzene and ethyl acetate (3:1) was used as a
solvent. e E. coli transformant harboring POMT9 was
used to synthesize scopoletin from esculetin. e meth-
ylation reaction using E. coli was carried out as described
by Kim etal. [20].
Analysis of the reaction products was carried out using
ermo Ultimate 3000 HPLC [23]. Mass spectrometry
and proton nuclear magnetic resonance (NMR) were per-
formed as previously described [24, 25]. e 1H NMR of
fraxetin in acetone-d6 (in ppm) is δ 3.87 (3H, s, 6-OCH3),
6.15 (1H, d, J = 9.3Hz, H-3), 6.76 (1H, s, H-5), 7.91 (1H,
d, J = 9.3Hz, H-4) [26].
Results anddiscussion
Biotransformation ofscopoletin intofraxetin using E. coli
harboring scopoletin 8‑hydroxylase
Fraxetin is 8-hydroxy scopoletin. S8H from A. thali-
ana (AtS8H) and its homologue from rice (OsS8H) were
cloned as a glutathione S-transferase fusion protein and
expressed in E. coli. Scopoletin was tested, along with
other structurally-related coumarins, such as esculetin,
isoscopoletin, and scoparone. ese four compounds
have esculetin derivatives. ree methylated esculetins
(isoscopoletin, scopoletin, and scoparone) were synthe-
sized using E. coli harboring ROMT9, purified, and used
as substrates.
E. coli harboring AtS8H or OsS8H was tested for the
conversion of esculetin, isoscopoletin, scopoletin, and
scoparone by the administration of each compound. E.
coli harboring OsS8H did not convert any coumarins
used. However, for E. coli harboring AtS8H, scopole-
tin and isoscopoletin were converted into novel com-
pounds that had retention times different from those of
the corresponding substrates (Fig. 2). Other substrates
0.02.04.06.08.010.0 12.0 14.0 16.0 18.0 20.0
0
2500
2500
2500
250
400
100
140
200
350
340 nm
P1
P2
S1
S2
S3
a
b
c
d
e
f
min
mAU
Fig. 2 HPLC analysis of the reaction in E. coli harboring AtS8H. E. coli harboring AtS8H was administered scopoletin (a), isoscopoletin (b), and
esculetin (c); the reaction product was analyzed. df denote standard scopoletin, isoscopoletin, and esculetin, respectively. P1, reaction product
from scopoletin; P2, reaction product from isoscopoletin; S1, scopletin; S2, isoscopoletin; S3, esculetin
Page 4 of 6
Anetal. Appl Biol Chem (2020) 63:55
(esculetin and scoparone) did not generate any new prod-
uct. e molecular mass of the products from scopoletin
and isoscopoletin was 207.937Da, which is the molecular
mass obtained if hydroxylation occurs. S8H utilized one
methylated esculetins (scopoletin and isoscopoletin) as a
substrate and did not utilize dimethylated (scoparone) or
unmethylated esculetin. Scopoletin was a better substrate
than isoscopoletin; 84.8% of scopoletin was converted, as
opposed to the conversion of only 55% isoscopoletin. To
determine the structure of the biotransformation product
from scopoletin, the reaction product was purified, and
its structure was analyzed using proton NMR. e reac-
tion product was determined to be fraxetin (see Materials
and Methods). E. coli harboring different constructs (pG-
AtS8H, pE-AtS8H, pR-AtS8H, or pC-AtS8H) synthesized
the approximately same amount fraxetin from scopoletin.
To optimize the initial concentration of scopoletin and
the final yield of fraxetin, E. coli harboring AtS8H was
prepared at an OD600 of 3.0 after induction of AtS8H.
Subsequently, four different concentrations of scopole-
tin (100, 200, 300, and 500μM) were added. e highest
rate of conversion of scopoletin into fraxetin was seen
at 100μM scopoletin; 84.8μM fraxetin was synthesized
(84.8% conversion rate). However, fraxetin produc-
tion was highest at 200 μM scopoletin; approximately
139.5 μM fraxetin was synthesized (69.8% conversion
rate). Above 200μM scopoletin, the production level of
fraxetin registered a decline. e optimum initial cell
concentrations were also determined. Five initial cell
concentrations (OD600 = 1.0, 2.0, 3.0, 4.0, and 5.0) were
tested and 200μM scopoletin was administered. As the
initial cell concentration increased, the conversion of
scopoletin also registered a concomitant increase. At an
OD600 of 5.0, approximately 152.0μM of scopoletin was
converted into fraxetin.
Synthesis offraxetin fromesculetin andferulic acid
Fraxetin may also be synthesized from esculetin. Two
enzymatic reactions are required; the first is the conver-
sion of esculetin into scopoletin by an O-methyltrans-
ferase (OMT), and the second is the synthesis of fraxetin
from scopoletin by S8H. For the synthesis of scopoletin
from esculetin, three OMT genes (POMT7, POMT9,
and ROMT9) were evaluated. E. coli harboring POMT7
synthesized 56.8μM scopoletin from 100μM esculetin
(Fig.3a). However, E. coli harboring ROMT9 produced
three methylated esculetins (isoscopoletin, scopoletin,
and scoparone), with isoscopoletin as a major product.
e ratio of isoscopoletin, scopoletin, and scoparone was
83: 13: 3. POMT9 generated almost the same amounts of
isoscopoletin (38.1 μM) and scopoletin (37.4μM) from
100μM esculetin. erefore, E. coli harboring POMT7
was utilized to synthesize scopoletin from esculetin.
A two-step reaction was conducted using two E. coli
transformants to augment the final yield of fraxetin. e
first reaction was carried out using E. coli harboring
POMT7. Approximately 56.9μM scopoletin was synthe-
sized from 100 μM esculetin (Fig. 3a). Further incuba-
tion did not result in the conversion of more esculetin.
ereafter, the culture filtrate from the first reaction
was combined with E. coli harboring AtS8H. Approxi-
mately 41.4μM fraxetin was synthesized from 56.9μM
scopoletin (Fig. 3b), indicating that there was approxi-
mately 72.7% conversion from the synthesized fraxetin.
0.02.04.06.08.010.0 12.0 14.016.0 18.0 20.0
0
100
180
0
100
200
300
S1
P1
S1
P1
P2
a
b
min
340 nm
mAU
Fig. 3 Synthesis of fraxetin from esculetin using two E. coli transformants. a Conversion of esculetin into scopoletin using E. coli harboring POMT7.
E. coli harboring POMT7 was administered esculetin (S1), following which the reaction product was analyzed. P1 denotes the reaction product from
scopoletin. b Synthesis of fraxetin from scopoletin using E. coli harboring AtS8H. The culture filtrate from E. coli harboring POMT7 was administered
to the E. coli harboring AtS8H, and the reaction product was analyzed
Page 5 of 6
Anetal. Appl Biol Chem (2020) 63:55
Fraxetin was successfully synthesized from esculetin by
a two-step reaction. e final yield of fraxetin synthe-
sized from esculetin was lower than that from scopoletin;
moreover, the conversion rate of scopoletin into fraxetin
in the two-step reaction was lower than that seen for the
direct conversion. is could possibly be attributed to the
metabolite(s) in the first step inhibiting the second reac-
tion. It was attempted herein to synthesize fraxetin from
esculetin using an E. coli transformant harboring both
POMT7 and AtS8H. Only 3.4 μM fraxetin and 17.2μM
scopoletin were synthesized from 100μM esculetin.
Next, fraxetin was synthesized from ferulic acid. ree
enzymatic reactions are required for this. Previously,
scopoletin was successfully synthesized from ferulic
acid using E. coli harboring IbF6H2 and Os4CL. It was
reasoned that introducing AtS8H into E. coli harbor-
ing IbF6H2 and Os4CL could result in the synthesis of
fraxetin from ferulic acid. ree genes (IbF6H, Os4CL,
and AtS8H) were introduced into E. coli, and the result-
ing transformant was administered ferulic acid. e E.
coli transformant synthesized fraxetin from ferulic acid.
To optimize fraxetin synthesis, several initial ferulic acid
concentrations (100, 200, 300, and 500μM) were tested.
e synthesis of fraxetin was optimal at 100μM of initial
ferulic acid, and approximately 33.3μM fraxetin was syn-
thesized (Fig. 4). Unreacted ferulic acid and scopoletin
were accumulated at the higher concentrations of ferulic
acid.
Fraxetin was synthesized from three different sub-
strates (scopoletin, esculetin, and ferulic acid). As shown
in Fig. 1, more enzymes are required when fraxetin is
synthesized from ferulic acid or esculetin than when it is
synthesized from scopoletin. Consequently, the final yield
of fraxetin was higher (84.8μM) when it was synthesized
from scopoletin (100μM). Its yield was decreased when
synthesis was carried out from esculetin (41.4μM) or fer-
ulic acid (33.3μM).
An attempt was made to synthesize fraxetin from
esculetin or ferulic acid. One E. coli transformant
harboring both POMT7 and AtS8H synthesized a lower
amount of fraxetin from esculetin than the other two E.
coli transformants, each of which conducted one reac-
tion. However, fraxetin was successfully synthesized
from ferulic acid using one E. coli transformant harbor-
ing three genes (Os4CL, IbF6H, and AtS8H). Esculetin
may compete with scopoletin for AtS8H. In the E. coli
transformant harboring both POMT7 and AtS8H, escu-
letin served as a substrate for POMT7 and an inhibitor
of AtS8H. erefore, following the synthesis of sco-
poletin by POMT7, AtS8H could not utilize scopoletin
because it was inhibited by esculetin. Conversely, when
two independent E. coli transformants were used, more
scopoletin synthesized by the first E. coli transformant
harboring POMT7 was present in the medium and was
converted into fraxetin by the second E. coli transfor-
mant harboring AtS8H. When fraxetin was synthesized
from ferulic acid, only scopoletin was synthesized;
therefore, it was possible to synthesize fraxetin using E.
coli harboring Os4CL, IbF6H, and AtS8H.
Acknowledgements
The present study was supported by grants from the Next‑Generation Bio‑
Green 21 Program (PJ01326001), Rural Development Administration, Republic
of Korea.
Authors’ contributions
SHA and JHA designed the experiments. SHA, GSC, and JHA performed the
experiments and analyzed the data. SHA, GSC, and JHA wrote the manuscript.
All authors read and approved the final manuscript.
Funding
Funding was received from the Next‑Generation BioGreen 21 Program, Rural
Development Administration (PJ01326001).
Availability of data and materials
All data generated or analyzed during the present study are included in this
published article.
Competing interests
The authors declare that they have no competing interests.
Received: 5 August 2020 Accepted: 9 September 2020
0.02.04.06.08.010.0 12.0 14.0 16.0 18.0 20.0
0
100
200
S1
P1
P2
min
340 nm
mAU
Fig. 4 Synthesis of fraxetin from ferulic acid using E. coli harboring Os4CL, IbF6H, and AtS8H. S1, ferulic acid; P1, scopoletin; P2, fraxetin
Page 6 of 6
Anetal. Appl Biol Chem (2020) 63:55
References
1. Thakur A, Singla R, Jaitak V (2015) Coumarins as anticancer agents: a
review on synthetic strategies, mechanism of action and SAR studies. Eur
J Med Chem 101:476–495
2. Bourgaud F, Hehn A, Larbat R, Doerper S, Gontier E, Kellner S, Matern U
(2006) Biosynthesis of coumarins in plants: a major pathway still to be
unraveled for cytochrome P450 enzymes. Phytochem Rev 5:293–308
3. Stringlis IA, de Jonge R, Pieterse CMJ (2019) The age of coumarins in
plant–microbe interactions. Plant Cell Physiol 60:1405–1419
4. Tsai HH, Rodríguez‑Celma J, Lan P, Wu YC, Vélez‑Bermúdez IC, Schmidt W
(2018) Scopoletin 8‑hydroxylase‑mediated fraxetin production is crucial
for iron mobilization. Plant Physiol 177:194–207
5. Kang SY, Kim YC (2007) Neuroprotective coumarins from the root
of Angelica gigas: structure‑activity relationships. Arch Pharm Res
30:1368–1373
6. Kayser O, Kolodziej H (1999) Antibacterial activity of simple coumarins:
structural requirements for biological activity. Z Naturforsch, C: J Biosci
54:169–174
7. Kai K, Mizutani M, Kawamura N, Yamamoto R, Tamai M, Yamaguchi H,
Sakata K, Schimizu B‑I (2008) Scopoletin is biosynthesized via ortho‑
hydroxylation of feruloyl CoA by a 2‑oxoglutarate‑dependent dioxyge‑
nase in Arabidopsis thaliana. Plant J 55:989–999
8. Vialart G, Hehn A, Olry A, Ito K, Krieger C, Larbat R, Paris C, Bun‑Ichi S,
Sugimoto Y, Mizutani M, Bourgaud F (2012) A 2‑oxoglutarate depend‑
ent dioxygenase from Ruta graveolens L. exhibits p‑coumaroyl CoA 2
hydroxylase activity (C2H): a missing step in the synthesis of umbellifer‑
one. in plants. Plant J 70:460–470
9. Matsumoto S, Mizutani M, Sakata K, Shimizu B (2012) Molecular cloning
and functional analysis of the ortho‑hydroxylases of p‑coumaroyl coen‑
zyme A/feruloyl coenzyme A involved in formation of umbelliferone and
scopoletin in sweet potato, Ipomoea batatas (L.) Lam. Phytochemistry
74:49–57
10. Liu S, Zainuddin IM, Vanderschuren H, Doughty J, Beeching JR (2017)
RNAi inhibition of feruloyl CoA 6‑hydroxylase reduces scopoletin biosyn‑
thesis and post‑harvest physiological deterioration in cassava (Manihot
esculenta Crantz) storage roots. Plant Mol Biol 94:185–195
11. Zhao Y, Jian X, Wu J, Huang W, Huang C, Luo J, Kong L (2019) Elucidation
of the biosynthesis pathway and heterologous construction of a sustain‑
able route for producing umbelliferone. J Biol Eng 13:44
12. Yao R, Zhao Y, Liu T, Huang C, Xu S, Sui Z, Luo J, Kong L (2017) Identifica‑
tion and functional characterization of a p‑coumaroyl CoA 2‑hydroxylase
involved in the biosynthesis of coumarin skeleton from Peucedanum
praeruptorum Dunn. Plant Mol Biol 95:199–213
13. Siwinska J, Siatkowska K, Olr y A, Grosjean J, Hehn A, Bourgaud F, Meharg
AA, Carey M, Lojkowska E, Ihnatowicz A (2018) Scopoletin 8‑hydroxylase:
a novel enzyme involved in coumarin biosynthesis and iron‑deficiency
responses in Arabidopsis. J Exp Bot 69:1735–1748
14. Kimura Y, Sumiyoshi M (2015) Antitumor and antimetastatic actions of
dihydroxycoumarins (esculetin or fraxetin) through the inhibition of M2
macrophage differentiation in tumor‑associated macrophages and/or G1
arrest in tumor cells. Eur J Pharm 746:115–125
15. Molina‑Jiménez MF, Sánchez‑Reus MI, Andres D, Cascales M, Benedi J
(2004) Neuroprotective effect of fraxetin and myricetin against rotenone‑
induced apoptosis in neuroblastoma cells. Brain Res 1009:9–16
16. Murali R, Srinivasan S, Ashokkumar N (2013) Antihyperglycemic effect
of fraxetin on hepatic key enzymes of carbohydrate metabolism in
streptozotocin‑induced diabetic rats. Biochimie 95:1848–1854
17. Kundu J, Chae IG, Chun K‑S (2016) Fraxetin induces heme oxygenase‑1
expression by activation of Akt/Nrf2 or AMP‑activated protein kinase α/
Nrf2 pathway in HaCaT cells. J Cancer Prev 21:135–143
18. Yang S‑M, Shim GY, Kim B‑G, Ahn J‑H (2015) Biological synthesis of cou‑
marins in Escherichia coli. Microb Cell Fact 14:65
19. Chu LL, Pandey RP, Lim HN, Jung HJ, Thuan NH, Kim T‑S, Sohng JK (2017)
Synthesis of umbelliferone derivatives in Escherichia coli and their biologi‑
cal activities. J Biol Eng 11:15
20. Kim BG, Lee YJ, Lee S, Lim Y, Cheong Y, Ahn J‑H (2008) Altered regioselec‑
tivity of a poplar O‑methyltransferase, POMT‑7. J Biotech 138:107–111
21. Kim BG, Lee Y, Hur HG, Lim Y, Ahn J‑H (2006) Production of Three O‑Meth‑
hylated Esculetins with E. coli Expressing O‑Methyltransferase from Poplar.
Biosci Biotech Biochem 70:1269–1272
22. Kim BG, Lee Y, Hur H‑G, Lim Y, Ahn J‑H (2006) Flavonoid 3O‑methyltrans‑
ferase from rice: cDNA cloning, characterization and functional expres‑
sion. Phytochemistry 67:387–394
23. Lee SJ, Sim GY, Kang H, Yeo WS, Kim B‑G, Ahn J‑H (2018) Synthesis of
avenanthramides using engineered Escherichia coli. Microb Cell Fact
17:46
24. Song MK, Cho AR, Sim GY, Ahn J‑H (2019) Synthesis of diverse hydroxy‑
cinnamoyl phenylethanoid esters using Escherichia coli. J Agric Food
Chem 67:2028–2035
25. Yoon J‑A, Kim B‑G, Lee WJ, Lim Y, Chong Y, Ahn J‑H (2012) Production of a
novel quercetin glycoside through metabolic engineering of Escherichia
coli. Appl Env Microbiol. 78:4256–4262
26. Yu M, Sun A, Zhnag Y, Liu R (2014) Purification of coumarin compounds
from Cortex fraxinus by adsorption chromatography. J Chromatogr Sci
52:1033–1037
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
... Esculetin can be converted to scopoletin by an O-methyltransferase, and then scopoletin can be synthesized to fraxetin by scopoletin 8-hydroxylase. Ferulic acid can also be synthesized to scopoletin by E. coli, and then further synthesized to fraxetin [36]. Coumarin was found only in NTJ, not in NTF or NTR, which indicated that coumarin was newly generated through fermentation process. ...
Article
Nitraria tangutorum Bobrov is a berry shrub with white flowers and red fruits, which grows in the deserts in the Tibetan Plateau, Mongolia, and western China. Its fruit N. tangutorum Fruit (NTF) contains various bioactive compounds, with anti-fatigue, anti-inflammatory, and neuroprotective functions. However, the high saccharide content of NTF makes it unsuitable for diabetic patients. In this study, we fermented NTF to obtain N. tangutorum Fermented Juice (NTJ), and N. tangutorum Fermented Residue (NTR), which are suitable for diabetics to consume. We characterized the bioactive compounds in NTF, NTR, and NTJ, and found that fermentation increased the diversity of bioactive compounds, and greatly reduced sucrose, glucose, and fructose content while generating trehalose, which has the potential to manage blood glucose levels. Further, NTJ displays anti-diabetic potential due to various compounds anti-diabetic properties. This study provides a basis for further clinical research on NTJ’s anti-diabetic function in humans.
... Esculetin can be converted to scopoletin by an O-methyltransferase, and then scopoletin can be synthesized to fraxetin by scopoletin 8-hydroxylase. Ferulic acid can also be synthesized to scopoletin by E.coli., and then further synthesized to fraxetin [36]. Coumarin was found only in NTJ, not in NTF or NTR, which indicated that coumarin was newly generated through fermentation process. ...
Preprint
Full-text available
Nitraria tangutorum Bobrov is a berry shrub with white flowers and red fruits, which grows in the deserts in the Tibetan Plateau, Mongolia, and western China. Its fruit N. tangutorum fruit (NTF) contains various bioactive compounds, with anti-fatigue, anti-inflammatory, and neuroprotective functions. However, the high saccharide content of NTF makes it unsuitable for diabetic patients. In this study, we fermented NTF to obtain N. tangutorum fermented juice (NTJ), and N. tangutorum fermented residue (NTR), which are suitable for diabetics to consume. We characterized the bioactive compounds in NTF, NTR, and NTJ, and found that fermentation increased the diversity of bioactive compounds, and greatly reduced sucrose, glucose, and fructose content while generating trehalose, which has the potential to manage blood glucose levels. Further, NTJ displays anti-diabetic potential due to various compounds anti-diabetic properties. This study provides a basis for further clinical research on NTJ's anti-diabetic function in humans.
... Yang et al. [71] developed the synthetic pathway based on the cascade enzymes of TAL-4CL-F6'H to synthesize 82.9 mg/l umbelliferone from p-coumaric acid, 79.5 mg/l esculetin from caffeic acid, and 52.3 mg/l scopoletin from ferulic acid in E. coli. An et al. [72] harnessed the E. coli harboring 4CL, F6'H, O-methyltransferase (OMT), and scopoletin 8-hydroxylase to synthesize 175 μg/l fraxetin from ferulic acid. Zhao et al. [73] constructed a biocatalytic pathway containing HpaBC, OMT, and a fusion of 4CL-F6'H in the engineered S. cerevisiae, forming 4.79 mg/l scopoletin from lignin hydrolysate. ...
Article
Full-text available
Lignin is one of the most widespread organic compounds found on earth, boasting a wealth of aromatic molecules. The use of lignin feedstock for biochemical productions is of great importance for achieving “carbon neutrality.” In recent years, a strategy for lignin valorization known as the “bio-funnel” has been proposed as a means to generate a variety of commercially valuable chemicals from lignin-derived compounds. The implementation of biocatalysis and metabolic engineering techniques has substantially advanced the biotransformation of depolymerized lignin into chemicals and materials within the supply chain. In this review, we present an overview of the latest advancements in microbial upcycling of depolymerized lignin into value-added chemicals. Besides, the review provides insights into the problems facing current biological lignin valorization while proposing further research directions to improve these technologies for the extensive accomplishment of the lignin upcycling.
... The biosynthesis of coumarins was first studied in Arabidopsis thaliana from cinnamic acid [39]. Apart from plants, coumarins have been successfully synthesized in Escherichia coli [40,41]. Various reports have documented umbelliferone as the primary source of osthole biosynthesis. ...
Article
Full-text available
Neurodegenerative diseases constitute a major threat to human health and are usually accompanied by progressive structural and functional loss of neurons. Abnormalities in synaptic plasticity are involved in neurodegenerative disorders. Aberrant cell signaling cascades play a predominant role in the initiation, progress as well as in the severity of these ailments. Notch signaling is a pivotal role in the maintenance of neural stem cells and also participates in neurogenesis. PI3k/Akt cascade regulates different biological processes including cell proliferation, apoptosis, and metabolism. It regulates neurotoxicity and mediates the survival of neurons. Moreover, the activated BDNF/TrkB cascade is involved in promoting the transcription of genes responsible for cell survival and neurogenesis. Despite significant progress made in delineating the underlying pathological mechanisms involved and derangements in cellular metabolic promenades implicated in these diseases, satisfactory strategies for the clinical management of these ailments are yet to be achieved. Therefore, the molecules targeting these cell signaling cascades may emerge as useful leads in developing newer management strategies. Osthole is an important ingredient of traditional Chinese medicinal plants, often found in various plants of the Apiaceae family and has been observed to target these aforementioned mediators. Until now, no review has been aimed to discuss the possible molecular signaling cascades involved in osthole-mediated neuroprotection at one platform. The current review aimed to explore the interplay of various mediators and the modulation of the different molecular signaling cascades in osthole-mediated neuroprotection. This review could open new insights into research involving diseases of neuronal origin, especially the effect on neurodegeneration, neurogenesis, and synaptic plasticity. The articles gathered to compose the current review were extracted by using the PubMed, Scopus, Science Direct, and Web of Science databases. A methodical approach was used to integrate and discuss all published original reports describing the modulation of different mediators by osthole to confer neuroprotection at one platform to provide possible molecular pathways. Based on the inclusion and exclusion criteria, 32 articles were included in the systematic review. Moreover, literature evidence was also used to construct the biosynthetic pathway of osthole. The current review reveals that osthole promotes neurogenesis and neuronal functioning via stimulation of Notch, BDNF/Trk, and P13k/Akt signaling pathways. It upregulates the expression of various proteins, such as BDNF, TrkB, CREB, Nrf-2, P13k, and Akt. Activation of Wnt by osthole, in turn, regulates downstream GSK-1β to inhibit tau phosphorylation and β-catenin degradation to prevent neuronal apoptosis. The activation of Wnt and inhibition of oxidative stress, Aβ, and GSK-3β mediated β-catenin degradation by osthole might also be involved in mediating the protection against neurodegenerative diseases. Furthermore, it also inhibits neuroinflammation by suppressing MAPK/NF-κB-mediated transcription of genes involved in the generation of inflammatory cytokines and NLRP-3 inflammasomes. This review delineates the various underlying signaling pathways involved in mediating the neuroprotective effect of osthole. Modulation of Notch, BDNF/Trk, MAPK/NF-κB, and P13k/Akt signaling pathways by osthole confers protection against neurodegenerative diseases. The preclinical effects of osthole suggest that it could be a valuable molecule in inspiring the development of new drugs for the management of neurodegenerative diseases and demands clinical studies to explore its potential. An effort has been made to unify the varied mechanisms and target sites involved in the neuroprotective effect of osthole. The comprehensive description of the molecular pathways in the present work reflects its originality and thoroughness. The reviewed literature findings may be extrapolated to suggest the role of othole as a “biological response modifier” which contributes to neuroprotection through kinase modulatory, immunomodulatory, and anti-oxidative activity, which is documented even at lower doses. The current review attempts to emphasize the gaps in the existing literature which can be explored in the future.
... Scopoletin 8-hydroxylase (S8H) has been reported as a strong Fe-responsive gene encoding a 2-oxoglutarate-dependent dioxygenase [27,28], and it can also catalyze scopoletin to form fraxetin in plants via o-hydroxylation at the C8 position. Overexpression of S8H improved fraxetin biosynthesis in Escherichia coli [36]. In the presence of CYP82C4, fraxetin can be converted to the oxidized and reduced forms of sideretin [33]. ...
Article
Full-text available
Scopoletin is a typical example of coumarins, which can be produced in plants. Scopoletin acts as a precursor for pharmaceutical and health care products, and also possesses promising biological properties, including antibacterial, anti-tubercular, anti-hypertensive, anti-inflammatory, anti-diabetic, and anti-hyperuricemic activity. Despite the potential benefits, the production of scopoletin using traditional extraction processes from plants is unsatisfactory. In recent years, synthetic biology has developed rapidly and enabled the effective construction of microbial cell factories for production of high value-added chemicals. Herein, this review summarizes the progress of scopoletin biosynthesis in artificial microbial cell factories. The two main pathways of scopoletin biosynthesis are summarized firstly. Then, synthetic microbial cell factories are reviewed as an attractive improvement strategy for biosynthesis. Emerging techniques in synthetic biology and metabolic engineering are introduced as innovative tools for the efficient synthesis of scopoletin. This review showcases the potential of biosynthesis of scopoletin in artificial microbial cell factories.
... The production of industrially essential compounds from sustainable biomass is attracting attention as a critical technology for solving serious global problems, including climate change [1][2][3][4]. Escherichia coli strains have been widely used as a host for developing the cell factory to produce chemicals from sustainable biomass [5][6][7][8][9][10]. One reason is the rich and well-defined information on genetics, physiology, and tools, for metabolic rewiring by engineering [11][12][13]. ...
Article
Full-text available
Escherichia coli has been used as a host to construct the cell factory for biobased production of chemicals from renewable feedstocks. Because galactose is found in marine biomass as a major component, the strategy for galactose utilization in E. coli has been gained more attention. Although galactose and glucose co-fermentation has been reported using the engineered E. coli strain, few reports have covered fermentation supplemented with galactose as a sole carbon source in the mutant lacking the repressor-specific carbon catabolite repression (CCR). Here, we report the effects of the deregulation of the repressor-specific CCR (galR− and galS−) in fermentation supplemented with galactose as a sole carbon source, using the engineered E. coli strains. In the fermentation using the galR− and galS− double mutant (GR2 strain), an increase of rates in sugar consumption and cell growth was observed compared to the parent strain. In the glucose fermentation, wild-type W3110 and its mutant GR2 and GR2PZ (galR−, galS−, pfkA−, and zwf−) consumed sugar at a higher rate than those values obtained from galactose fermentation. However, the GR2P strain (galR−, galS−, and pfkA−) showed no difference between fermentations using glucose and galactose as a sole carbon source. This study provides essential information for galactose fermentation using the CCR-deregulated E. coli strains.
Article
Full-text available
Fraxetin is a bioactive molecule present in various natural plants, especially Cortex Fraxini. Evidenced outcomes in phytochemical and biological analyses for this agent are now available in the literature, but an insightful review is yet unknown. The goal of the current research is to offer a panoramic illustration of natural observation, biosynthesis, synthesis, pharmacology, and pharmacokinetics for fraxetin. Esculetin and ferulic acid acted as precursors in the enzymatic biosynthetic route, whereas fraxetin could be easily synthesized from simple phenols. A great deal of interest was obtained in using this molecule for pharmacological targets. Herein, its pharmacological value included anticancer, antioxidative, anti-inflammatory, antidiabetic, antiobesity, and antimicrobial activities, as well as the protection of the liver, neurons, heart, bone, lung, kidney, and others. Anticancer activity may involve the inhibition of proliferation, invasion, and migration, together with apoptotic induction. Health benefits from this molecule were deduced from its ability to suppress cytokines and protect the immune syndrome. Various signaling pathways, such as Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3), phosphoinositide 3 kinase (PI3K)/protein kinase B (Akt), nuclear factor kappa B (NF-κB)/NLRP3, Akt/AMPK, have been proposed for in vitro and in vivo mechanisms of action. Fraxetin is highly distributed to rat plasma and several organs. However, more pharmacokinetic studies to improve its bioavailability are needed since its solubility in water is still limited.
Article
Full-text available
Purpose: Chronic unpredictable stress (CUS) induces long-term neuronal and synaptic plasticity with a neurohormonal disbalance leading to the development of co-existing anxiety, depression, and cognitive decline. The side effects and delayed onset of current clinically used antidepressants has prompted a quest for antidepressants with minimum drawbacks. Fraxetin is a natural coumarin derivative with documented antioxidant and neuroprotective activity though its effects on stress are unknown. This study therefore aimed to investigate any possible acute effect of fraxetin in behavioral tests including a CUS paradigm in correlation with brain regional neurochemical changes. Methods: Mice were subjected to a series of mild stressors for 14 days to induce CUS. Furthermore, behavioral performance in the open field test, forced swim test (FST), Y-maze and elevated plus-maze were evaluated. Postmortem frontal cortical, hippocampal and striatal tissues were analyzed via high-performance liquid chromatography (HPLC) for neurochemical changes. Result: Acute administration of fraxetin (20–60 mg/kg, orally) decreased depression-like behavior in the FST and behavioral anxiety in both the open field test and elevated plus-maze. Memory deficits induced during the CUS paradigm were markedly improved as reflected by enhanced Y maze performance. Concurrent biochemical and neurochemical analyses revealed that only the two higher fraxetin doses decreased elevated serum corticosterone levels while diminished serotonin levels in the frontal cortex, striatum and hippocampus were reversed, though noradrenaline was only raised in the striatum. Concomitantly, dopamine levels were restored by fraxetin at the highest dose exclusively in the frontal cortex. Conclusion: Acute treatment with fraxetin attenuated CUS-induced behavioral deficits, ameliorated the increased corticosterone level and restored altered regional neurotransmitter levels and this may indicate a potential application of fraxetin in the management of anxiety and depression modeled by CUS. However, further studies are warranted regarding the chronic effects of fraxetin behaviorally and neurochemically.
Article
Full-text available
Background: Coumarins play roles in many biological processes. Angelica decursiva is one of the major sources of coumarins in China. Due to increasing demand for coumarins in the marketplace, traditional extraction from plants is now considered economically insufficient and unsustainable. Microbial synthesis is a promising strategy for scalable production of coumarins. However, the biosynthetic pathway of coumarin remains poorly understood, and even more, the genes associated with this process have not been characterized in A. decursiva. Results: RNA-seq was employed to elucidate the umbelliferone biosynthetic pathway. The results indicated that three enzymes, phenylalanine ammonia-lyase (PAL), 4-Coumarate: Coenzyme A Ligase (4CL), and p-coumaroyl CoA 2'-hydroxylase (C2'H) were involved in umbelliferone biosynthesis. Using the cloned genes, we generated a synthetic biology based microbial cell factory that produces coumarins from tyrosine utilizing Rhodotorula glutinis tyrosine ammonia lyase (RgTAL) to bypass cinnamic acid 4-hydroxylase (C4H). With metabolic engineering strategies, we deleted prephenate dehydratase (pheA), anthranilate synthase (trpE) and transcriptional regulatory protein (tyrR) and overexpressed six related genes involved in tyrosine biosynthesis, to drive the carbon flux from tyrosine. To overcome the limitation of 4CL, a virtual screening and site-specific mutagenesis-based protein engineering approach was applied. In addition, induction/culture conditions and different ions were employed to further improve the yield of umbelliferone. Finally, a yield of 356.59 mg/L umbelliferone was obtained. Conclusions: The current study elucidated the umbelliferone biosynthesis pathway in A. decursiva. The results also demonstrated the feasibility of integrating gene mining with synthetic biology techniques to produce natural compounds.
Article
Full-text available
Coumarins are a family of plant-derived secondary metabolites that are produced via the phenylpropanoid pathway. In the past decade, coumarins have emerged as iron-mobilizing compounds that are secreted by plant roots and aid in iron uptake from iron-deprived soils. Members of the coumarin family are found in many plant species. Besides their role in iron uptake, coumarins have been extensively studied for their potential to fight infections in both plants and animals. Coumarin activities range from antimicrobial and antiviral to anticoagulant and anticancer. In recent years, studies in the model plant species tobacco and Arabidopsis have significantly increased our understanding of coumarin biosynthesis, accumulation, secretion, chemical modification and their modes of action against plant pathogens. Here, we review current knowledge on coumarins in different plant species. We focus on simple coumarins and provide an overview on their biosynthesis and role in environmental stress responses, with special attention for the recently discovered semiochemical role of coumarins in aboveground and belowground plant–microbe interactions and the assembly of the root microbiome.
Article
Full-text available
Background: Hydroxycinnamoyl anthranilates, also known as avenanthramides (avns), are a group of phenolic alkaloids with anti-inflammatory, antioxidant, anti-itch, anti-irritant, and antiatherogenic activities. Some avenanthramides (avn A-H and avn K) are conjugates of hydroxycinnamic acids (HC), including p-coumaric acid, caffeic acid, and ferulic acid, and anthranilate derivatives, including anthranilate, 4-hydroxyanthranilate, and 5-hydroxyanthranilate. Avns are primarily found in oat grain, in which they were originally designated as phytoalexins. Knowledge of the avns biosynthesis pathway has now made it possible to synthesize avns through a genetic engineering strategy, which would help to further elucidate their properties and exploit their beneficial biological activities. The aim of the present study was to synthesize natural avns in Escherichia coli to serve as a valuable resource. Results: We synthesized nine avns in E. coli. We first synthesized avn D from glucose in E. coli harboring tyrosine ammonia lyase (TAL), 4-coumarate:coenzyme A ligase (4CL), anthranilate N-hydroxycinnamoyl/benzoyltransferase (HCBT), and anthranilate synthase (trpEG). A trpD deletion mutant was used to increase the amount of anthranilate in E. coli. After optimizing the incubation temperature and cell density, approximately 317.2 mg/L of avn D was synthesized. Avn E and avn F were then synthesized from avn D, using either E. coli harboring HpaBC and SOMT9 or E. coli harboring HapBC alone, respectively. Avn A and avn G were synthesized by feeding 5-hydroxyanthranilate or 4-hydroxyanthranilate to E. coli harboring TAL, 4CL, and HCBT. Avn B, avn C, avn H, and avn K were synthesized from avn A or avn G, using the same approach employed for the synthesis of avn E and avn F from avn D. Conclusions: Using different HCs, nine avns were synthesized, three of which (avn D, avn E, and avn F) were synthesized from glucose in E. coli. These diverse avns provide a strategy to synthesize both natural and unnatural avns, setting a foundation for exploring the biological activities of diverse avns.
Article
Full-text available
Iron (Fe) is an essential mineral nutrient and an important factor for the composition of natural plant communities. Low Fe availability in aerated soils with neutral or alkaline pH has led to the evolution of elaborate mechanisms that extract Fe from the soil solution. In Arabidopsis thaliana, Fe is acquired by an orchestrated strategy that comprises mobilization, chelation, and reduction of Fe3+ prior to its uptake. We show here that At3g12900, previously annotated as scopoletin 8-hydroxylase (S8H), participates in Fe acquisition by mediating the biosynthesis of fraxetin (7,8-dihydroxy-6-methoxycoumarin), a coumarin derived from the scopoletin pathway. S8H is highly induced in roots of Fe-deficient plants both at the transcript and protein levels. Mutants defective in the expression of S8H showed increased sensitivity to growth on pH 7.0 media supplemented with an immobile source of Fe and reduced secretion of fraxetin. Transgenic lines overexpressing S8H exhibited an opposite phenotype. Homozygous s8h mutants grown on media with immobilized Fe accumulated significantly more scopolin, the storage form of scopoletin, supporting the designated function of S8H in scopoletin hydroxylation. Fraxetin exhibited Fe-reducing properties in vitro with higher rates being observed at neutral relative to acidic pH. Supplementing the media containing immobile Fe with fraxetin partially rescued the s8h mutants. In natural Arabidopsis accessions differing in their performance on media containing immobilized Fe, the amount of secreted fraxetin was highly correlated with growth and Fe and chlorophyll content, indicating that fraxetin secretion is a decisive factor for calcicole-calcifuge behavior (i.e. the ability/inability to thrive on alkaline soils) of plants.
Article
Full-text available
Iron (Fe) deficiency is a serious agricultural problem, particularly for alkaline soils. Secretion of coumarins by Arabidopsis thaliana roots is induced under Fe-deficiency. An essential enzyme for the biosynthesis of major Arabidopsis coumarins, scopoletin and its derivatives, is Feruloyl-CoA 6'-Hydroxylase1 (F6'H1), that belongs to a large enzyme family of the 2-oxoglutarate and Fe(II)-dependent dioxygenases. Another member of this family, which is a close homologue of F6'H1, and is encoded by a strongly Fe-responsive gene, At3g12900, is functionally characterized in the presented work. We purified the At3g12900 protein heterologously expressed in Escherichia coli and demonstrated that it is involved in the conversion of scopoletin into fraxetin, via hydroxylation at the C8-position, scopoletin 8-hydroxylase (S8H). Its function in plant cells was confirmed by the transient expression of S8H protein in Nicotiana benthamiana leaves, followed by the metabolite profiling and the biochemical and ionomic characterization of Arabidopsis s8h knockout lines grown under various Fe regimes. Our results indicate that S8H is involved in coumarin biosynthesis, as part of mechanisms used by plants to assimilate Fe.
Article
Full-text available
Key message: A p-coumaroyl CoA 2'-hydroxylase responsible for the formation of coumarin lactone ring was identified from Peucedanum praeruptorum Dunn and functionally characterized in vitro. Coumarins are important plant secondary metabolites with a variety of biological activities. Ortho-hydroxylation of cinnamates leads to the formation of coumarin lactone ring and is generally thought to be a key step in coumarin biosynthesis. However, ortho-hydroxylases, especially p-coumaroyl CoA 2'-hydroxylase (C2'H) responsible for the biosynthesis of the most common coumarin skeleton, have received insufficient attention. Here, a putative ortho-hydroxylase PpC2'H was isolated from P. praeruptorum Dunn, a traditional Chinese medicinal herb rich in coumarins. Expression profile indicated that PpC2'H exhibited the highest transcript level in roots and could be up-regulated by MeJA elicitation. Subcellular localization of PpC2'H was demonstrated to be cytosol in planta. In order to functionally characterize PpC2'H, the purified recombinant protein was incubated with various potential substrates. HPLC-ESI-MS analysis indicated that PpC2'H catalyzed the conversion of p-coumaroyl CoA into hydroxylated intermediate, which then underwent spontaneous lactonization to generate umbelliferone. Our data also showed that light would promote the spontaneous process. In addition, based on homology modeling and site-directed mutagenesis, amino acid residues Phe-130, Lys-141, Asn-207, His-224, Asp-226, His-282 and Phe-298 were verified essential for enzymatic activity. These findings provide insight into structure-function relationship of this pivotal ortho-hydroxylase and also contribute to elucidating the biosynthetic mechanism of coumarin skeleton.
Article
Full-text available
Background Umbelliferone, also known as 7-hydroxycoumarin, is a phenolic metabolite found in many familiar plants. Its derivatives have been shown to have various pharmacological and chemo-preventive effects on human health. A uridine diphosphate glycosyltransferase YjiC from Bacillus licheniformis DSM 13, a cytochrome P450BM3 (CYP450 BM3) variant namely mutant 13 (M13) from Bacillus megaterium, and an O-methyltransferase from Streptomyces avermitilis (SaOMT2) were used for modifications of umbelliferone. Results Three umbelliferone derivatives (esculetin, skimmin, and herniarin) were generated through enzymatic and whole cell catalysis. To improve the efficiencies of biotransformation, different media, incubation time and concentration of substrate were optimized and the production was scaled up using a 3-L fermentor. The maximum yields of esculetin, skimmin, and herniarin were 337.10 μM (67.62%), 995.43 μM (99.54%), and 37.13 μM (37.13%), respectively. The water solubility of esculetin and skimmin were 1.28-folds and 3.98-folds as high as umbelliferone, respectively, whereas herniarin was 1.89-folds less soluble than umbelliferone. Moreover, the antibacterial and anticancer activities of herniarin showed higher than umbelliferone, esculetin and skimmin. Conclusions This study proves that both native and engineered enzymes could be employed for the production of precious compounds via whole cell biocatalysis. We successfully produced three molecules herniarin, esculetin and skimmin in practical amounts and their antibacterial and anticancer properties were accessed. One of the newly synthesized molecules the present research suggests that the combinatorial biosynthesis of different biosynthetic enzymes could rapidly promote to a novel secondary metabolite.
Article
Full-text available
Cassava (Manihot esculenta Crantz) is a major world crop, whose storage roots provide food for over 800 million throughout the humid tropics. Despite many advantages as a crop, the development of cassava is seriously constrained by the rapid post-harvest physiological deterioration (PPD) of its roots that occurs within 24–72 h of harvest, rendering the roots unpalatable and unmarketable. PPD limits cassava’s marketing possibilities in countries that are undergoing increased development and urbanisation due to growing distances between farms and consumers. The inevitable wounding of the roots caused by harvesting triggers an oxidative burst that spreads throughout the cassava root, together with the accumulation of secondary metabolites including phenolic compounds, of which the coumarin scopoletin (7-hydroxy-6-methoxy-2H-1-benzopyran-2-one) is the most abundant. Scopoletin oxidation yields a blue-black colour, which suggests its involvement in the discoloration observed during PPD. Feruloyl CoA 6′-hydroxylase is a controlling enzyme in the biosynthesis of scopoletin. The cassava genome contains a seven membered family of feruloyl CoA 6′-hydroxylase genes, four of which are expressed in the storage root and, of these, three were capable of functionally complementing Arabidopsis T-DNA insertion mutants in this gene. A RNA interference construct, designed to a highly conserved region of these genes, was used to transform cassava, where it significantly reduced feruloyl CoA 6′-hydroxylase gene expression, scopoletin accumulation and PPD symptom development. Collectively, our results provide evidence that scopoletin plays a major functional role in the development of PPD symptoms, rather than merely paralleling symptom development in the cassava storage root. Electronic supplementary material The online version of this article (doi:10.1007/s11103-017-0602-z) contains supplementary material, which is available to authorized users.
Article
Caffeic acid phenethyl ester (CAPE) is an ester of a hydroxycinnamic acid (phenylpropanoid) and a phenylethanoid (2-phenylethanol; 2-PE) that has long been used in traditional medicine. Here, we synthesized 54 hydroxycinnamic acid-phenylethanoid esters by feeding 64 combinations of hydroxycinnamic acids and phenylethanols to Escherichia coli harboring the rice genes OsPMT and Os4CL. The same approach was applied for ester synthesis with caffeic acid and eight different phenyl alcohols. Two hydroxycinnamoyl phenethyl esters, p-coumaroyl tyrosol and CAPE, were also synthesized from glucose using engineered E. coli by introducing genes for the synthesis of substrates. Consequently, we synthesized approximately 393.4 mg/L p-coumaroyl tyrosol and 23.8 mg/L CAPE with this approach. Overall, these findings demonstrate that the rice PMT and 4CL proteins can be used for the synthesis of diverse hydroxycinnamoyl phenylethanoid esters owing to their promiscuity and that further exploration of the biological activities of these compounds is warranted.
Article
Coumarins are important compounds that contribute to the adaptation of plants to biotic or abiotic stresses. Among coumarins, umbelliferone occupies a pivotal position in the plant phenylpropanoid network. Previous studies indicated that umbelliferone is derived from the ortho-hydroxylation of p-coumaric acid by an unknown biochemical step to yield 2,4-dihydroxycinnamic acid, which then undergoes spontaneous lactonization. Based on a recent report of a gene encoding a 2-oxoglutarate-dependent dioxygenase from Arabidopsis thaliana that exhibited feruloyl CoA 6'-hydroxylase activity (Bourgaud et al., 2006), we combined a bioinformatic approach and a cDNA library screen to identify an orthologous ORF (Genbank accession number JF799117) from Ruta graveolens L. This ORF shares 59% amino acid identity with feruloyl CoA 6'-hydroxylase, was functionally expressed in Escherichia coli, and converted feruloyl CoA into scopoletin and p-coumaroyl CoA into umbelliferone with equal activity. Its bi-functionality was further confirmed in planta: transient expression of JF799117 in Nicotiana benthamiana yielded plants with leaves containing high levels of umbelliferone and scopoletin when compared to control plants, which contained barely detectable traces of these compounds. The expression of JF799117 was also tightly correlated to the amount of umbelliferone that was found in UV-elicited R. graveolens leaves. Therefore, JF799117 encodes a p-coumaroyl CoA 2'-hydroxylase in R. graveolens, which represents a previously uncharacterized step in the synthesis of umbelliferone in plants. Psoralen, which is an important furanocoumarin in R. graveolens, was found to be a competitive inhibitor of the enzyme, and it may exert this effect through negative feedback on the enzyme at an upstream position in the pathway.