ArticlePDF Available

Bifunctional Cytochrome P450 Enzymes Involved in Camptothecin Biosynthesis

Authors:

Abstract and Figures

Camptothecin (CAM) is a well-known complex plant-derived antitumour monoterpenoid indole alkaloid (MIA). Featuring a unique pentacyclic pyrroloquinoline scaffold, CAM is biosynthetically distinct from the other known MIAs, such as antitumour vincristine and vinblastine. Herein, CaCYP72A565 and CaCYP72A610 enzymes involved in the biosynthesis of monoterpenoid moiety of CAM were cloned from CAM-producing Camptotheca acuminata. Heterologous overexpression and functional characterization assays showed that CaCYP72As catalyse two consecutive reactions, the stereoselective hydroxylation at C-7 of 7-deoxyloganic acid and the subsequent carbon-carbon (C-C) bond cleavage between C-7 and C-8 of iridoid glucoside, to generate the intramolecular cyclopentane ring-opening secoiridoid glucoside. Comparative metabolite profiling analyses suggested that C. acuminata synthesizes loganic acid, secologanic acid, and strictosidinic acid as its MIA carboxylic acid intermediates. CaCYP72As are novel bifunctional enzymes that catalyse stereoselective hydroxylation and subsequent C-C bond cleavage reactions to give ring-opening product with two functional groups, an aldehyde and a double bond.
Content may be subject to copyright.
Bifunctional Cytochrome P450 Enzymes Involved in Camptothecin
Biosynthesis
Yun Yang,
,§
Wei Li,
Jing Pang,
,§
Liangzhen Jiang,
Xixing Qu,
Xiang Pu,
Guolin Zhang,
and Yinggang Luo*
,,
Center for Natural Products Research, Chinese Academy of Sciences, Chengdu Institute of Biology, 9 Section 4, Renmin Road
South, Chengdu 610041, China
State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy
of Sciences, 345 Lingling Road, Shanghai 200032, China
§
University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
*
SSupporting Information
ABSTRACT: Camptothecin (CAM) is a well-known, complex, plant-derived
antitumor monoterpenoid indole alkaloid (MIA). Featuring a unique pentacyclic
pyrroloquinoline scaold, CAM is biosynthetically distinct from the other known
MIAs, such as antitumor vincristine and vinblastine. Herein, CaCYP72A565 and
CaCYP72A610 enzymes involved in the biosynthesis of the monoterpenoid moiety of
CAM were cloned from CAM-producing Camptotheca acuminata. Heterologous
overexpression and functional characterization assays showed that CaCYP72As
catalyzes two consecutive reactions, the stereoselective hydroxylation at C-7 of 7-
deoxyloganic acid and the subsequent carboncarbon (CC) bond cleavage between
C-7 and C-8 of iridoid glucoside, to generate the intramolecular cyclopentane ring-
opening secoiridoid glucoside. Comparative metabolite proling analyses suggested
that C. acuminata synthesizes loganic acid, secologanic acid, and strictosidinic acid as
its MIA carboxylic acid intermediates. CaCYP72As are novel bifunctional enzymes
that catalyze stereoselective hydroxylation and subsequent CC bond cleavage
reactions to give a ring-opening product with two functional groups, an aldehyde and a double bond.
Cytochrome P450 monooxygenases (CYPs), haem-con-
taining enzymes, are the largest superfamily of enzymes
that play pivotal roles in metabolism and catabolism in all
kingdoms of life.
15
Generally, CYPs catalyze regio- and
stereoselective monooxygenation/hydroxylation, and they
participate in many biochemical pathways to produce an
immense chemical diversity of species-specic natural products
in secondary metabolism.
1,2
Some CYPs have been shown to
catalyze unique reactions, such as the oxidative rearrangement
of carbon skeletons, methylenedioxy bridge formation, phenol
coupling, and oxidative carboncarbon (CC) bond cleav-
age.
14
For instance, CYP72A1 and its isoform from the
vincristine- and vinblastine-producing Catharanthus roseus
6,7
were functionally proven to catalyze a unique CC bond
cleavage reaction in the cyclopentane ring of loganin to give
the ring-opening product secologanin, which is known to be a
key intermediate for many pharmaceutically important natural
products, such as monoterpenoid indole alkaloids (MIAs),
ipecacuanha alkaloids, and secoiridoid glucosides.
8,9
Featuring a unique pentacyclic pyrroloquinoline scaold,
camptothecin (CAM, Figure 1) is a well-known plant-derived
complex antitumor MIA.
10
As unique DNA topoisomerase I
inhibitors,
11
the CAM-derived topotecan and irinotecan have
been approved by the U.S. Food and Drug Administration as
antitumor drugs for use against many types of tumors, such as
small-cell lung and refractory ovarian cancers.
1214
As a
distinct pyrroloquinoline-containing MIA, CAM was proposed
to biologically synthesize through a modied MIA pathway in
plants (Figure 1).
1517
CYPs have been proposed to catalyze
many biochemical conversion steps to establish the distinctive
pentacyclic pyrroloquinoline MIA.
1517
In the course of the investigations of CAM biosynthesis, we
analyzed the public transcriptome data sets from Camptotheca
acuminata,
18,19
cloned CaCYP72As, and compared them with
the functionally characterized CrSLSs (CYP72A1 and its
isoform),
6,7
the secologanin synthase responsible for the
specicCC bond cleavage conversion of loganin into
secologanin (Figure 1B) in Catharanthus roseus (Supporting
Information (SI), Figures S1S5). The CaCYP72As show high
amino acid residue sequence identity (>60%) with CrSLSs
(Figure S3). Phylogenetic analyses revealed that two
CaCYP72As were clustered into one clade with CrSLSs
(Figure S4), indicating that they may catalyze the oxidative
cyclopentane ring opening of iridoid glucosides to produce
secoiridoid glucosides. The CaCYP72As were individually
Received: December 29, 2018
Accepted: May 21, 2019
Letters
pubs.acs.org/acschemicalbiology
Cite This: ACS Chem. Biol. XXXX, XXX, XXXXXX
© XXXX American Chemical Society ADOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
Downloaded by BUFFALO STATE at 19:50:03:651 on May 23, 2019
from https://pubs.acs.org/doi/10.1021/acschembio.8b01124.
overexpressed (Figure S6) in the Saccharomyces cerevisiae
WAT11 strain, which overexpresses the Arabidopsis thaliana
NADPH P450 reductase 1.
20
The yeast cells/microsomes
overexpressing the pYES2-CT empty vector or the pYES2-CT-
CaCYP72As were incubated with NADPH and loganin. The
HPLC-DAD (Figure S7A) and HPLC-DAD-HRMS (Figure
2A) analyses showed that both Caa_locus_1905 and
Caa_locus_133 catalyze the conversion of loganin into a
new product. The enzymatically converted product showed an
identical HPLC retention time (panels II and IV, Figure 2A;
panels III and V, Figure S7A) and an identical UV spectrum
(panels I and II, Figure S8A) to those of the standard
secologanin (panel V, Figure 2A; panel VI, Figure S7A; panel
III, Figure S8A). HRESIMS of the product and its
fragmentation pattern (panels I and II, Figure S8D) are in
perfect accordance with that of the standard secologanin
(Figure 2A; Figure S8C). Caa_locus_1905 and Caa_lo-
cus_133 displayed intramolecular oxidative CCbond
cleavage activity and were named CYP72A565 and
CYP72A610, respectively, by the CYP nomenclature commit-
tee.
21
As positive experimental controls, CrSLSs were cloned,
overexpressed, and characterized by following the above-
described experimental procedures.
Because path b (Figure 1) may be present in C. acuminata,
17
we tested whether the CaCYP72As can catalyze the conversion
of loganic acid into secologanic acid. The enzymatic assays
were performed as described above using loganic acid to
replace loganin. The HPLC-DAD (Figure S7B) and HPLC-
DAD-HRMS (Figure 2B) analyses showed that both
CaCYP72A565 and CaCYP72A610 catalyze the conversion
of loganic acid into a new product. The enzymatically
transformed product showed an identical HPLC retention
time (panels II and IV, Figure 2B; panels III and V, Figure
S7B) and an identical UV spectrum (panels I and II, Figure
S8B) to those of the standard secologanic acid (panel V, Figure
2B; panel VI, Figure S7B; panel III, Figure S8B). HRESIMS of
the product and its fragmentation pattern (panels I and II,
Figure S8F) are in perfect accordance with that of the standard
Figure 1. Putative biosynthetic pathway for MIAs. Biosynthetic pathway for the monoterpenoid moieties of CAM (A) and of the MIAs in
Catharanthus roseus (B). (C) The PictetSpengler condensation reaction between tryptamine and the monoterpenoid and the subsequent
biochemical modications to generate MIAs. Enzyme names in bold represent enzymes that have been functionally characterized. Path a, original
pathway; path b, recently modied pathway; DMAPP, dimethylallyl diphosphate; IPP, isopentenyl diphosphate; GPP, geranyl diphosphate; GPPS,
GPP synthase; GES, geraniol synthase; G10H, geraniol 10-hydroxylase; 10HGO, 10-hydroxygeraniol oxidase; IS, iridodial synthase; 7-DLS, 7-
deoxyloganetic acid synthase; 7-DLGT, 7-deoxyloganetic acid glucosyltransferase; DL7H, 7-deoxyloganic acid-7-hydroxylase; LAMT, loganic acid
O-methyltransferase; SLS, secologanin synthase; TDC, tryptophan decarboxylase; STR, strictosidine synthase; STRAS, strictosidinic acid synthase.
ACS Chemical Biology Letters
DOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
B
secologanic acid (Figure 2B; Figure S8E). However, CrSLSs
could not catalyze the above-mentioned conversion reaction.
Phylogenetic analyses (Figure S4) showed that the
CaCYP72As are close to CrDL7H (CYP72A224), the 7-
deoxyloganic acid 7-hydoxylase involved in MIA biosynthesis
in Catharanthus roseus (Figure 1B),
22
which implied that the
CaCYP72As may catalyze the regio- and stereoselective
hydroxylation of 7-deoxyloganic acid to give loganic acid.
The enzymatic assays were performed as described above using
7-deoxyloganic acid as the substrate. The HPLC-DAD (Figure
S9) and HPLC-DAD-HRMS (Figure 3) analyses showed that
both CaCYP72A565 and CaCYP72A610 catalyze the con-
version of 7-deoxyloganic acid into loganic acid. The
enzymatically catalyzed product showed an identical HPLC
retention time (panels II and IV, Figure 3; panels III and V,
Figure S9A) to that of the standard loganic acid (panel V,
Figure 3; panel VI, Figure S9A). UPLC-DAD-HRESIMS of the
product and its fragmentation pattern (panel II, Figure S9B;
panels I and II, Figure S9D) are in perfect accordance with that
of the standard loganic acid (Figure 3;Figure S9C).
From the reaction mixtures of the CaCYP72A-catalyzed
hydroxylation of 7-deoxyloganic acid, a small product peak
showed an identical HPLC retention time (panels II and IV,
Figure 3; panels III and V, Figure S9A) to that of the
secologanic acid standard (panel V, Figure 3; panel VI, Figure
S9A). UPLC-DAD-HRESIMS of the product and its
fragmentation ion (panel I, Figure S9B; panels I and II, Figure
S9F) are in perfect accordance with that of the standard
secologanic acid (Figure 3;Figure S9E). The results indicate
that the CaCYP72As catalyze the regio- and stereoselective
hydroxylation of 7-deoxyloganic acid to loganic acid and the
subsequent oxidative cyclopentane ring-opening of loganic acid
to generate secologanic acid. To validate this conclusion,
authentic loganic acid was added to the CaCYP72A-catalyzed
1 h reaction mixtures using 7-deoxyloganic acid as the
substrate, and the catalytic conversion reaction was continued
for an additional 2 h (panel III, Figure 4; panel IV, Figure
S10). Comparing the inactivated CaCYP72A-catalyzed reac-
tions (panel I, Figure S10) with the enzymatic reactions, a
small amount of secologanic acid could be detected in the 1 h
reaction mixture (panel I, Figure 4; panel II, Figure S10). The
amount of secologanic acid increased clearly in the modied 3
h reaction (panel III, Figure 4; panel IV, Figure S10) compared
with the normal 3 h reaction (panel II, Figure 4; panel III,
Figure S10). Thus, CaCYP72As catalyze consecutive regio-
and stereoselective hydroxylation and further intramolecular
oxidative CC bond cleavage to generate the cyclopentane
ring-opening product with two functional groups, an aldehyde
and a double bond.
Preliminary investigations of the consecutive reactions
implied that the CaCYP72A-catalyzed CC cleavage activity
Figure 2. Catalytic oxidative CC bond cleavage of iridoid glucosides
to generate secoiridoid glucosides by CaCYP72As. (A) Relative
intensenties of the extracted ion chromatogram (EIC) traces from
HPLC-DAD-HRESIMS (positive ionization mode) of the micro-
somal enzymatic reactions using loganin as a substrate. Standard
loganin (blue, [M + Na]+m/z413.1419) and secologanin (red, [M +
Na]+m/z411.1273; panel V). (B) HPLC-DAD-HRMS (positive
ionization mode) EICs of the microsomal enzymatic reactions using
loganic acid as a substrate. Standard loganic acid (blue, [M + Na]+m/
z399.1265) and secologanic acid (red, [M + Na]+m/z397.1111;
panel V). Panel I, boiled CaCYP72A610; panel II, CaCYP72A610;
panel III, boiled CaCYP72A565; panel IV, CaCYP72A565.
Figure 3. Catalytic hydroxylation of 7-deoxyloganic acid to form
loganic acid by CaCYP72As. HPLC-DAD-HRESIMS (negative
ionization mode) EICs of the microsomal enzymatic reactions using
7-deoxyloganic acid as a substrate. Standard 7-deoxyloganic acid
(green, [M H]m/z359.1342), loganic acid (blue, [M H]m/z
375.1299), and secologanic acid (red, [M H]m/z373.1143; panel
V). Panel I, boiled CaCYP72A610; panel II, CaCYP72A610; panel
III, boiled CaCYP72A565; panel IV, CaCYP72A565.
ACS Chemical Biology Letters
DOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
C
is a rate-limiting step (Figures 3 and 4;Figures S9 and S10).
The protein overexpression time, reaction buer pH, temper-
ature, time, NADPH, and substrate concentration were
optimized for the catalytic oxidative CC bond cleavage
catalyzed by CaCYP72As (Figure S11). The steady kinetic
parameters suggested that CaCYP72A565 shows a slightly
higher catalytic eciency than CaCYP72A610 (Table 1,Figure
S12). To investigate the substrate scope of the CaCYP72As,
nine iridoid and secoiridoidglucosides,namely,8-O-
acetylharpagide, harpagide, geniposide, geniposidic acid,
morroniside, sweroside, sanzhiside methyl, sanzhiside, and
verbenalin (Figure S13), were tested for potential catalysis by
the CaCYP72As. HPLC-DAD, HRESIMS, and NMR data
interpretation revealed that geniposidic acid and its methyl
ester geniposide were hydroxylated at C-6βby the
CaCYP72As to give scandoside and its methyl ester,
respectively (Figure S14), indicating that the CaCYP72As
catalyzed stereoselective β-hydroxylation toward a less
hindered cyclopentane moiety of iridoid glucosides. However,
the recombinant CrDL7H can accept only 7-deoxyloganic acid
as a substrate.
22
It should be noted that we did not test 7-
deoxyloganin, the methyl ester of 7-deoxyloganic acid, as a
substrate since it could not be obtained commercially or be
derived from 7-deoxyloganic acid via methylation due to the
limited amount of 7-deoxyloganic acid. None of these substrate
analogues could be converted into the corresponding ring-
opening products by the CaCYP72As. Thus, for catalytic
oxidative CC bond cleavage, CaCYP72As have strict
substrate recognition similar to that of CrSLSs.
6,7
CrSLSs
cannot catalyze the hydroxylation of 7-deoxyloganic acid but
do catalyze a further oxidation of secologanin to generate
secoxyloganin.
7
Secoxyloganin, an acidic derivative of
secologanin, cannot be condensed with tryptamine by
strictosidine synthase, which is ascribed to the lack of the
aldehyde functional group.
7
The microsomal SLS from cell
suspension cultures of Lonicera japonica was shown to have
strict substrate specicity, accepting only loganin as a
substrate.
23
A few bifunctional CYPs have been shown to
catalyze both oxidation and unique reactions (SI). However,
CaCYP72As are novel bifunctional enzymes that catalyze
stereoselective hydroxylation and subsequent intramolecular
cyclopentane ring opening through CC cleavage to give one
product with two functional groups, an aldehyde and a double
bond.
The quantitative real-time PCR analyses showed that both
CaCYP72A565 and CaCYP72A610 genes were widely ex-
pressed in dierent tissues of C. acuminata (Figure S15). The
accumulation of secologanic acid is positively correlated with
the transcription levels of CaCYP72As within dierent plant
tissues (Figure S15). The CAM content in the leaves is
positively associated with the transcription levels of the
CaCYP72As. In the stems, CAM was found at slightly lower
levels than in the leaves, while the stems showed the lowest
transcriptional levels of CaCYP72As. The seemingly contra-
dictory results suggest that the leaves are the biosynthetic
compartment for CAM and a portion of freshly synthesized
CAM is transported to the stems and stored (Figure S15),
since the biosynthesis of MIA is thought to be compartmen-
talized in dierent plant tissues.
13
Five dierent plant tissues, including roots (SR), stems (SS),
and leaves (SL) from seedlings and young leaves (YL) and
ower buds (FB) from wild mature C. acuminata, were
collected and extracted by two solvent systems to obtain the
putative intermediates of the CAM biosynthesis pathway in C.
acuminata.
17,25
The freshly prepared extracts were immediately
subjected to HPLC-DAD and UPLC-HRESIMS analyses
(Figure S16). Ten putative biosynthetic intermediates, CAM,
and 10-hydroxycamptothecin were detected and identied by
comparing the HPLC retention times, UV spectra, and
accurate molecular ions observed in the UPLC-HRESIMS
mass spectra with those of the standards and/or the proposed
biosynthetic intermediates (Table S2). Tryptamine, secolo-
ganic acid, and strictosidinic acid were detected in all plant
Figure 4. Consecutive conversion reactions from 7-deoxyloganic acid
to secologanic acid via loganic acid catalyzed by CaCYP72A610 (A)
and CaCYP72A565 (B). HPLC-DAD-HRESIMS (negative ionization
mode) EIC analyses of the microsomal enzymatic reactions using 7-
deoxyloganic acid as a substrate. Standard 7-deoxyloganic acid (green,
[M H]m/z359.1342), loganic acid (blue, [M H]m/z
375.1299), and secologanic acid (red, [M H]m/z373.1143;
panels IV). Panel I, 1 h reaction; panel II, 3 h reaction; panel III, an
additional 1 mM of loganic acid was added to the reaction mixtures of
the whole reaction at 1 h and then the reaction was continued for 2
more hours.
Table 1. Kinetic Parameters of CaCYP72As with Dierent Substrates
enzyme substrate Km
a
Vmax
b
Kcat
c
Kcat/Km
CaCYP72A610 7-deoxyloganic acid 0.33 ±0.07 0.29 ±0.02 0.09 0.28
loganic acid 1.78 ±0.63 0.14 ±0.04 0.04 0.02
loganin 1.10 ±0.10 0.09 ±0.01 0.03 0.026
CaCYP72A565 7-deoxyloganic acid 0.33 ±0.07 0.67 ±0.11 0.21 0.63
loganic acid 1.50 ±0.48 0.25 ±0.04 0.08 0.05
loganin 2.29 ±0.17 0.18 ±0.01 0.06 0.025
a
Km, mM.
b
Vmax,μM/min.
c
Kcat,μM/min/mM enzyme. All experimental values represent the means of three replicates ±standard deviation.
ACS Chemical Biology Letters
DOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
D
tissues. Loganic acid was detected in the SL, YL, and FB
tissues. As expected, C. acuminata accumulated loganic acid,
secologanic acid, and strictosidinic acid but lacked detectable
levels of the corresponding methyl ester derivatives loganin,
secologanin, and strictosidine. The monoterpenoid intermedi-
ates geraniol, 10-hydroxygeraniol, and 7-deoxyloganic acid
could not be detected in any of the ve plant tissues, which
may be ascribed to their instability and volatility. The
biosynthetic intermediates of the downstream pathway of
CAM,
17,2427
strictosamide and its ketolactam and epoxide,
pumiloside, and deoxypumiloside, were identied in all ve
plant tissues. CAM and 10-hydroxycamptothecin were
accumulated in all plant tissues. The comparative metab-
olomics analyses (Figure S16, Table S2) suggested that C.
acuminata might follow an alternative secoiridoid glucoside
pathway with carboxylic acid intermediates to strictosidinic
acid (path b, Figure 1), which is highly consistent with the
results of the latest report.
17
However, in the best studied
MIA-producing plant Catharanthus roseus, the methyl-esteried
intermediates, not the acidic intermediates, have been proven
to be involved in MIA biosynthesis.
24,28
The results presented
here and the previous report
17
implied that CAM biosynthesis
may be distinct from that of other MIAs. However, the methyl-
esteried intermediate pathway may not be ruled out, since the
enzymatic activity assays indicated that the CaCYP72As can
catalyze the conversion of loganin into secologanin (Figure 2A;
Figures S7, S8, S10, and S11), and we could not clarify
whether 7-deoxyloganin can be hydroxylated by CaCYP72As
at the moment due to the limited amounts of reagents. More
studies, such as molecular cloning and functional character-
ization of strictosidinic acid synthase (Figure 1), are in
progress to uncover the biosynthetic mysteries of CAM, which
will be fundamental to developing novel biological platforms to
produce CAM eciently.
In summary, we mined the transcriptome data of CAM-
producing C. acuminata and cloned candidate CYP72As for the
biosynthesis of the monoterpenoid moiety of CAM.
CaCYP72A565 and CaCYP72A610 were shown to identically
catalyze consecutive reactions, the stereoselective hydroxyla-
tion at C-7 of 7-deoxyloganic acid, and the subsequent CC
cleavage between C-7 and C-8 of iridoid glucoside, to generate
the intramolecular cyclopentane ring-opening secoiridoid
glucoside. Comparative metabolite proling analyses revealed
that C. acuminata synthesizes loganic acid, secologanic acid,
and strictosidinic acid as its carboxylic acid intermediates.
CaCYP72As are bifunctional CYP enzymes that catalyze
stereoselective hydroxylation and subsequent CC cleavage
reactions to give a ring-opening product with two functional
groups, an aldehyde and a double bond.
METHODS
General Experimental Procedures. A detailed description of
general experimental procedures is given in the Supporting
Information.
ASSOCIATED CONTENT
*
SSupporting Information
The Supporting Information is available free of charge on the
ACS Publications website at DOI: 10.1021/acschem-
bio.8b01124.
Experimental procedures, supplementary results, includ-
ing transcriptome data mining, molecular cloning, and
previously reported bifunctional CYPs, supplementary
gures, references, and tables (PDF)
AUTHOR INFORMATION
Corresponding Author
*E-mail: yinggluo@cib.ac.cn.
ORCID
Yinggang Luo: 0000-0002-8647-0948
Author Contributions
Y.L. conceived and designed the study. Y.Y., X.Q., X.P., and
L.J. mined the transcriptome data and cloned the candidate
CYPs. Y.Y., W.L., J.P., X.Q., and X.P. performed the
recombinant plasmids construction, heterologous expression,
enzymatic activity assay, product characterization, and kinetics
assay. Y.Y. and X.P. carried out the metabolomics analysis. Y.L.,
Y.Y., and G.Z. analyzed the data, and Y.L., Y.Y., and G.Z. wrote
the manuscript.
Funding
This work was supported in part by the 21172216 Project from
the National Natural Science Foundation of China, the ZSTH-
003 Project from the Chinese Academy of Sciences, and the
Applied and Basic Research Program of Sichuan Province
(2015JY0058).
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
We thank D. Nelson at University of Tennessee, USA, for
naming the CYPs according to the standardized CYP
nomenclature system. We thank T. Xia at Anhui Agricultural
University, China, for kindly supplying the S. cerevisiae WAT11
strain. We thank L. Zhou at Northwest A and F University,
China, for kindly supplying 7-deoxyloganic acid. We thank S.
Song at Shenyang Pharmaceutical University, China, for kindly
supplying secologanic acid and secoxyloganin.
REFERENCES
(1) Hamberger, B., and Bak, S. (2013) Plant P450s as versatile
drivers for evolution of species-specific chemical diversity. Philos.
Trans. R. Soc., B 368, 20120426.
(2) Ghosh, S. (2017) Triterpene structural diversification by plant
cytochrome P450 enzymes. Front. Plant Sci. 8, 1886.
(3) Mizutani, M., and Sato, F. (2011) Unusual P450 reactions in
plant secondary metabolism. Arch. Biochem. Biophys. 507, 194203.
(4) Guengerich, F. P., and Munro, A. W. (2013) Unusual
cytochrome P450 enzymes and reactions. J. Biol. Chem. 288,
1706517073.
(5) Guengerich, F. P., and Yoshimoto, F. K. (2018) Formation and
cleavage of C-C bonds by enzymatic oxidation-reduction reactions.
Chem. Rev. 118, 65736655.
(6) Irmler, S., Schröder, G., St-Pierre, B., Crouch, N. P., Hotze, M.,
Schmidt, J., Strack, D., Matern, U., and Schröder, J. (2000) Indole
alkaloid biosynthesis in Catharanthusroseus: new enzyme activities and
identification of cytochrome P450 CYP72A1 as secologanin synthase.
Plant J. 24, 797804.
(7) Dugéde Bernonville, T., Foureau, E., Parage, C., Lanoue, A.,
Clastre, M., Londono, M. A., Oudin, A., Houillé
, B., Papon, N.,
Besseau, S., Glé
varec, G., Atehortùa, L., Giglioli-Guivarch, N., St-
Pierre, B., De Luca, V., OConnor, S. E., and Courdavault, V. (2015)
Characterization of a second secologanin synthase isoform producing
both secologaninand secoxyloganin allows enhanced de novo assembly
of a Catharanthusroseus transcriptome. BMC Genomics 16, 619.
(8) Battersby, A. R., and Parry, R. J. (1971) Biosynthesis of the
Ipecac alkaloids and of ipecoside. J. Chem. Soc. D 1971, 901902.
ACS Chemical Biology Letters
DOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
E
(9) Inouye, H., Ueda, S., Inoue, K., and Takeda, Y. (1974) Studies
on monoterpene glucosides and related natural products. XXIII.
Biosynthesis of the secoiridoid glucosides, gentiopicroside, morroni-
side, oleuropein, and jasminin. Chem. Pharm. Bull. 22, 676686.
(10) Wall, M. E., Wani, M. C., Cook, C. E., Palmer, K. H., McPhail,
A. T., and Sim, G. A. (1966) Plant antitumor agents. I. The isolation
and structure of camptothecin, a novel alkaloidal leukemia and tumor
inhibitor from Camptothecaacuminata.J. Am. Chem. Soc. 88, 3888
3890.
(11) Hsiang, Y. H., Hertzberg, R., Hecht, S., and Liu, L. (1985)
Camptothecin induces protein-linked DNA breaks via mammalian
DNA topoisomerase I. J. Biol. Chem. 260, 1487314878.
(12) Demain, A. L., and Vaishnav, P. (2011) Natural products for
cancer chemotherapy. Microb. Biotechnol. 4, 687699.
(13) Sirikantaramas, S., Asano, T., Sudo, H., Yamazaki, M., and
Saito, K. (2007) Camptothecin: therapeutic potential and biotechnol-
ogy. Curr. Pharm. Biotechnol. 8, 196202.
(14) Thomas, C. J., Rahier, N. J., and Hecht, S. M. (2004)
Camptothecin: current perspectives. Bioorg. Med. Chem. 12, 1585
1604.
(15) Kai, G., Wu, C., Gen, L., Zhang, L., Cui, L., and Ni, X. (2015)
Biosynthesis and biotechnological production of anti-cancer drug
Camptothecin. Phytochem. Rev. 14, 525539.
(16) Qu, X., Pu, X., Chen, F., Yang, Y., Yang, L., Zhang, G., and Luo,
Y. (2015) Molecular cloning, heterologous expression, and functional
characterization of an NADPH-cytochrome P450 reductase gene from
Camptothecaacuminata, a camptothecin-producing plant. PLoS One
10, No. e0135397.
(17) Sadre, R., Magallanes-Lundback, M., Pradhan, S., Salim, V.,
Mesberg, A., Jones, A. D., and DellaPenna, D. (2016) Metabolite
diversity in alkaloid biosynthesis: a multilane (diastereomer) highway
for camptothecin synthesis in Camptothecaacuminata.Plant Cell 28,
19261944.
(18) Sun, Y., Luo, H., Li, Y., Sun, C., Song, J., Niu, Y., Zhu, Y., Dong,
L., Lv, A., Tramontano, E., and Chen, S. (2011) Pyrosequencing of
the Camptothecaacuminata transcriptome reveals putative genes
involved in camptothecin biosynthesis and transport. BMC Genomics
12, 533.
(19) Góngora-Castillo, E., Childs, K. L., Fedewa, G., Hamilton, J. P.,
Liscombe, D. K., Magallanes-Lundback, M., Mandadi, K. K., Nims, E.,
Runguphan, W., Vaillancourt, B., Varbanova-Herde, M., DellaPenna,
D., McKnight, T. D., O'Connor, S., and Buell, C. B. (2012)
Development of transcriptomic resources for interrogating the
biosynthesis of monoterpene indole alkaloids in medicinal plant
species. PLoS One 7, No. e52506.
(20) Pompon, D., Louerat, B., Bronine, A., and Urban, P. (1996)
Yeast expression of animal and plant P450 in optimized redox
environments. Methods Enzymol. 272,5164.
(21) Nelson, D. R. (2006) Cytochrome P450 nomenclature, 2004.
Methods Mol. Biol. 320,110.
(22) Salim, V., Yu, F., Altarejos, J., and De Luca, V. (2013) Virus-
induced gene silencing identifies Catharanthusroseus 7-deoxyloganic
acid-7-hydroxylase, a step in iridoid and monoterpene indole alkaloid
biosynthesis. Plant J. 76, 754765.
(23) Yamamoto, H., Katano, N., Ooi, A., and Inoue, K. (2000)
Secologanin synthase which catalyzes the oxidative cleavage of loganin
into secologanin is a cytochrome P450. Phytochemistry 53,712.
(24) Dugéde Bernonville, T., Clastre, M., Besseau, S., Oudin, A.,
Burlat, V., Glé
varec, G., Lanoue, A., Papon, N., Giglioli-Guivarch, N.,
St-Pierre, B., and Courdavault, V. (2015) Phytochemical genomics of
the Madagascar periwinkle: Unravelling the last twists of the alkaloid
engine. Phytochemistry 113,923.
(25) Montoro, P., Maldini, M., Piacente, S., Macchia, M., and Pizza,
C. (2010) Metabolite fingerprinting of Camptothecaacuminata and the
HPLC-ESI-MS/MSanalysis of camptothecin and related alkaloids. J.
Pharm. Biomed. Anal. 51, 405415.
(26) Carte, B. K., DeBrosse, C., Eggleston, D., Hemling, M.,
Mentzer, M., Poehland, B., Troupe, N., Westley, J. W., and Hecht, S.
M. (1990) Isolation and characterization of a presumed biosynthetic
precursor of camptothecin from extracts of Camptothecaacuminata.
Tetrahedron 46, 27472760.
(27) Hutchinson, C. R., Heckendorf, A. H., Straughn, J. L., Daddona,
P. E., and Cane, D. E. (1979) Biosynthesis of camptothecin. 3.
Definition of strictosamide as the penultimate biosynthetic precursor
assisted by carbon-13 and deuterium NMR spectroscopy. J. Am.
Chem. Soc. 101, 33583369.
(28) OConnor, S. E., and Maresh, J. J. (2006) Chemistry and
biology of monoterpene indole alkaloid biosynthesis. Nat. Prod. Rep.
23, 532547.
ACS Chemical Biology Letters
DOI: 10.1021/acschembio.8b01124
ACS Chem. Biol. XXXX, XXX, XXXXXX
F
... The recombinant plasmids and pYES2/CT empty plasmid were transformed (1.5 kV) into the WAT11 competent strain using a MicroPulser 165-2100 (Bio-Rad, Hercules, CA, USA). The obtained WAT11 strains were recovered and screened on SC-Ura nutrient-deficient solid medium as described previously (Yang et al., 2019). Single colonies were verified by yeast colony PCR (95 • C for 10 min; 30 cycles of 95 • C for 30 s, 57 • C for 30 s, and 72 • C for 1 min; 12 • C for 10 min). ...
... The verification primers are listed in Table S1. The positive WAT11 strain was activated in SC-Ura liquid medium and induced in YPGal medium, as described previously (Yang et al., 2019). ...
... The structures and biosynthetic pathway for the pentacyclic triterpenes isolated from C. acuminata. X. Pu et al. according the method previously described (Yang et al., 2019). The CYP716 protein concentrations were determined according to their extinction coefficient (ε 280nm ) calculated from ExPasy ProtParam (Yang et al., 2019;Gill and Hippel, 1989). ...
Article
Dozens of triterpenes have been isolated from Camptotheca acuminata, however, triterpene metabolism in this plant remains poorly understood. The common C28 carboxy located in the oleanane-type and ursane-type triterpenes indicates the existence of a functionally active triterpene, C28 oxidase, in this plant. Thorough mining and screening of the CYP716 genes were initiated using the multi-omics database for C. acuminata. Two CYP716A (CYP716A394 and CYP716A395) and three CYP716C (CYP716C80-CYP716C82) were identified based on conserved domain analyses and hierarchical cluster analyses. CYP716 microsomal proteins were prepared and their enzymatic activities were evaluated in vitro. The CYP716 classified into the CYP716C subfamily displays β-amyrin oxidation activity, and CYP716A displays α-amyrin and lupeol oxidation activity, based on gas chromatography-mass spectrometry analyses. The oxidation products were determined based on their mass and nuclear magnetic resonance spectrums. The optimum reaction conditions and kinetic parameters for CYP716C were determined, and functions were verified in Nicotiana benthaminana. Relative quantitative analyses revealed that these CYP716C genes were enriched in the leaves of C. acuminata plantlets after 60 d. These results indicate that CYP716C plays a dominant role in oleanane-type triterpene metabolism in the leaves of C. acuminata via a substrate-specific manner, and CYP716A is responsible for ursane- and lupane-type triterpene metabolism in fruit. This study provides valuable insights into the unique CYP716C-mediated oxidation step of pentacyclic triterpene biosynthesis in C. acuminata.
... The obtained WAT11 strains were recovered and screened on an SC-Ura nutrient deficiency solid medium as previously described. 12 The single colony was further verified by PCR. The PCR parameters were set as follows: 95°C for 10 min; 30 cycles of 95°C for 30 s, 57°C for 30 s, 72°C for 1 min; and 12°C for 10 min. ...
... The positive WAT11 strain was activated in the SC-Ura liquid medium and induced in the YPGal medium as previously described. 12 Recombinant WAT11 pellets were prepared by centrifugation (3500g, 4°C, 10 min). The pellets were crushed with ceramic beads (0.5 mm) using a freezing grinder (Jingxin, Shanghai, China). ...
... The supernatant was carefully transferred, and the microsomal proteins were prepared according to the method previously described. 12 The CYP71 protein concentrations were determined according to their extinction coefficient (ε 280 nm ) calculated from ExPasy ProtParam. 12,60 Western blotting was performed to verify CYP71 expression in Saccharomyces cerevisiae. ...
Article
The detailed metabolic map for camptothecin (CPT) biosynthesis in Camptotheca acuminata has been proposed according to our combined omics results. However, the CYP450-mediated epoxidation step in CPT biosynthesis remains unexplored. A proteomics-guided approach was used to identify and annotate the proteins enriched during the vigorous CPT metabolism period in mature C. acuminata and seedlings. Comparative analyses revealed that the CPT and flavonoid biosyntheses were vigorous in stems and all of the samples except the leaves, respectively. The CYP71BE genes were screened based on their enrichment patterns at the transcriptomic-proteomic level and biochemically characterized in Saccharomyces cerevisiae WAT11. Four CYP71BE proteins exhibited in vitro isoliquiritigenin epoxidase activity. Additionally, CYP71BE206 showed epoxidase activity toward strictosamide, the critical precursor for CPT biosynthesis, both in vitro and in Nicotiana benthamiana. In planta functional verification suggested that CYP71BE206 is involved in CPT biosynthesis. Their catalytic conditions were optimized, and the enzymatic parameters were determined. This study provides valuable insight into the CYP71BE-mediated epoxidation step for CPT biosynthesis and offers evidence to verify that the newly characterized epoxidase (CYP71BE206) is simultaneously responsible for the biosynthesis of CPT and the flavonoid in this plant. An evolution event probably happened on ancestral CYP71BE, resulting in the neofunctionalization of CYP71BE206.
... However, some E. coli endogenous enzymes that show substrate promiscuity will consume substrates unexpectedly, resulting in the reduction or failure of the target products. In the course of functional characterization of the bifunctional Camptotheca acuminata cytochrome P450 monooxygenase genes, CYP72A565 and CYP72A610, in Saccharomyces cerevisiae WAT11 (Yang et al. 2019), we initiated to express the CYP72As as fusion proteins with their redox partner, NADPH-dependent cytochrome P450 reductase (CPR), in E. coli BL21(DE3) (Qu 2014) based on the previous reports (Barnes et al. 1991;Hotze et al. 1995;Irmler et al. 2000). The catalytic activity assays revealed that a small amount of secologanin was detected while a large build-up of a compound was observed with the same UV profile as that of loganin/secologanin (Qu 2014). ...
... The primers Ca565-pETDuet-MS1(NcoI)-F and -MS1(NotI)-R (Table S1 in Supplementary Data) were designed and synthesized to amplify the coding sequence of CaCYP72A565 (GenBank: MH763569; Yang et al. 2019) by using a HiFiTaq DNA polymerase (TransGen Biotech (Beijing) Co., Ltd, China) with the following cycling parameters: 1 cycle of 94°C for 5 min; 30 cycles of 94°C for 30 s, 56°C for 30 s, 72°C for 80 s; followed by a final extension at 72°C for 10 min. The PCR amplification products were gel-purified, digested with Nco I and Not I endonucleases, and subcloned into a pETDuet-1 vector (Novagen, Madison, USA) harboring tCaCPR (Qu et al. 2015) that was digested with Nco I and Not I endonucleases, to afford the expression construct pETDuet-Ca565-tCaCPR. ...
Article
Full-text available
Escherichia coli, a well-known prokaryotic organism, has been widely employed as a versatile host for heterologous overexpression of proteins/biocatalysts and the production of pharmaceutically important intermediates/small molecules. However, some E. coli endogenous enzymes showing substrate promiscuity may disturb the heterologous metabolic flux, which will result in the reduction of substrates, intermediates, and target products. Here we reported an unexpected E. coli-catalyzed regioselective O-acetylation of various glucosides. The regioselectively O-acetylated products, 6′-O-acetyl-loganin and 6′-O-acetyl-loganic acid, were obtained and characterized from the enzymatic reaction in which the supernatants of E. coli expressing either CaCYP72A565 and CaCPR, the key enzymes involved in camptothecin biosynthesis, or empty vector were used as catalyst and loganin and loganic acid as independent substrate. An alkaloidal glucoside strictosamide was converted into the regioselectively O-acetylated product 6′-O-acetyl-strictosamide, implying substrate promiscuity of the E. coli-catalyzed O-acetylation reaction. Furthermore, 8 glucosides, including 5 iridoid glucosides and 3 flavonoid glucosides, were successfully converted into the regioselectively O-acetylated products by E. coli, indicating the wide substrate range for the unexpected E. coli-catalyzed O-acetylation. E. coli maltose O-acetyltransferase was demonstrated to be responsible for the mentioned regioselective O-acetylation at the 6-OH of the glucopyranosyl group of multiple classes of natural product glucosides through candidate acetyltransferase-encoding gene analysis, gene knock-out, gene complementation, and the relevant enzymatic reaction activity assays. The present study not only provides an efficient biocatalyst for regioselective O-acetylation but also notifies cautions for metabolic engineering and synthetic biology applications in E. coli. Graphical abstract Key points • 6-OH of glucosyl of multiple glucosides was regioselectively O-acetylated by E. coli. • Endogenous EcMAT is responsible for the regioselective O-acetylation reaction.
... Two of its semi-synthetic derivatives, irinotecan and topotecan, were clinically used as one of the best-selling plant-sourced antineoplastic drugs [3]. Owing to their pharmaceutical and commercial importance, substantial research has been carried out to optimize extraction procedures [4], develop chemical synthetic routes [5], mine alternative plant [6,7] or microbial resources [8], and elucidate the biosynthetic pathway for this star molecule [9][10][11][12][13][14][15][16]. In addition to its versatile alkaloid-producing ability, C. acuminata serves as an excellent producer of flavonoids as well. ...
... As shown in ▶ Table 2 and CPT (13) [40] and its representative natural analogues 9HCPT (6) [41] and 9MCPT (16) [10] are known compounds with substantial anticancer activity. Strictosidine (STR, 20) and strictosidine acid (STRA, 19) are key intermediates in the CPT biosynthetic pathway [42][43][44]. 9MMPE-Glc (1) is a novel E-ring-opened analogue of CPT, which may have good anticancer activity after being modified [34]. The content of these six compounds in different tissues of N. tomentosa, N. nimmoniana, C. acuminata, and O. japonica was then quantitatively analyzed by HPLC-Q-ESI-TOF-MS. ...
Article
Camptothecin (CPT) and its derivatives have attracted worldwide attention due to their notable anticancer activity. However, the growing demand for CPT in the global pharmaceutical industry has caused a severe shortage of CPT-producing plant resources. In this study, phytochemical analysis of Nothapodytes tomentosa resulted in the isolation and identification of CPT (13) and 16 analogues (1-12, 14-17), including a new (1) and five known (9, 10, 12, 15 and 17) CPT analogues with an open E-ring. In view of the potential anticancer activity of CPT analogues with an open E-ring, the fragmentation pathways and mass spectra profiles of these six CPT analogues (1, 9, 10, 12, 15 and 17) were investigated, providing a reference for the rapid detection of these compounds in other plants. Furthermore, based on the fragmentation patterns of CPT (13) and known analogues (2-8, 11, 14, 16, 18-26), the distribution and content of these compounds in different tissues of N. tomentosa, N. nimmoniana, Camptotheca acuminata, and Ophiorrhiza japonica were further studied. Our findings not only provide an alternative plant resource for further expanding the development and utilization of CPT and its analogues, but also lay a foundation for improving the utilization of known CPT-producing plant resources.
... There are SLS orthologs in multiple MIA-producing species including Rauvolfia serpentina, Ophiorrhiza pumila, Amsonia hubrichtii, Vinca minor, Tabernaemontana elegans, Camptotheca acuminata, and Nothapodytes nimmonia (Rather et al. 2020). It was recently shown that two SLS isoforms from C. acuminata, CYP72A565 and CYP72A610, will oxidize both 7-deoxyloganic acid and loganin, demonstrating the dual DL7H and SLS activity of this enzyme (Table 1; Figure S1 and S2) (Yang et al. 2019). These SLS isoforms from C. acuminata may have a more flexible binding site than the related P450s, allowing space for the additional alcohol and acetyl groups of loganin, without loss of oxidation activity. ...
Article
Full-text available
The large family of cytochrome P450 enzymes are heme containing proteins generally associated with the cytoplasmic face of the endoplasmic reticulum in Eukaryotic cells. They play essential roles in detoxification mechanisms of cellular processes and are key components in biosynthesis and evolution of specialized metabolites having various biological activities in the plant kingdom. The assembly of complex monoterpenoid indole alkaloids and benzylisoquinoline alkaloids involves numerous cytochrome P450 enzymes that participate in their chemical diversification and that act as central scaffolds for recruitment of the biosynthetic enzymes required for their production. The present review discusses the roles played by different CYP families (-71, -72, -75, -76, 80-, 82-, -86 and -719) in the diversification of MIA and BIA pathways that have been discovered and characterized. Recent studies using homology model guided site-directed mutagenesis coupled with determination of biochemical function are described that provide insights about how small modifications in protein structures allowed the evolution of new substrate specificity and the appearance of new monoterpenoid indole alkaloids and benzylisoquinoline alkaloids in Nature.
Article
Terpene indole alkaloids (TIAs) are plant-derived natural products synthesized in low levels in medicinal plants such as Catharanthus roseus and Camptotheca acuminata. TIA pathways species utilize several CYP72A subfamily members to form loganic acid from 7-deoxyloganic acid (a simple hydroxylation) as well as secologanin and secologanic acid from loganin and loganic acid (a C-C bond scission). Divergences in the specificities of these P450s have allowed Camptotheca secologanic acid synthases (SLASs) to become bifunctional enzymes capable of performing both reactions. In contrast, Catharanthus 7-deoxyloganic acid hydroxylase (7DLH) and secologanin synthase (SLS) have remained monofunctional enzymes capable either of monooxygenation or C-C bond scission. Our in vitro reconstitutions have now demonstrated that Camptotheca also contains a monofunctional 7DLH capable only of hydroxylating 7-deoxyloganic acid. Mutageneses aimed at evaluating residues important for the tight specificity of Camptotheca 7DLH (CYP72A729) and the broad specificity of SLAS (CYP72A564) have identified several residues where reciprocal switches substantially affect their activities: Lys128His in 7DLH increases hydroxylation of 7-deoxyloganic acid, and His132Lys in SLAS decreases this hydroxylation and C-C bond scissions of loganic acid and loganin; Gly321Ser in 7DLH does not affect hydroxylation of 7-deoxyloganic acid, whereas Ser324Gly in SLAS significantly increases C-C bond scission of loganic acid; Asp332Glu in the acid-alcohol pair of 7DLH increases hydroxylation of 7-deoxyloganic acid, whereas Glu335Asp in SLAS completely eliminates both of its activities. These mutations that enhance or eliminate these respective activities have significant potential to aid engineering efforts aimed at increasing TIA production in cell cultures, microbial systems, and/or other plants.
Article
Camptotheca acuminata Decne., the main source of camptothecin (CPT), has received increasing attention for its remarkable antitumor activity. Many CPT derivatives are clinically used as effective anticancer agents worldwide. However, their biosynthesis mechanism remains unclear, and uncovering this pathway would greatly facilitate development of alternative CPT production methods to replace current inefficient plant-derived ones. The expression of >30,000 genes was accurately quantified using unique molecular identifier RNA sequencing in ten C. acuminata tissues, and 7,854 proteins from five tissues were quantified with label-free quantitative proteomics. Fifteen full-length transcriptomes were sequenced with long-read Oxford Nanopore Technologies, and 5,692 alternative splicing events were discovered among 4,746 genes. Integrated transcriptome and proteome analysis provided novel insights into CPT biosynthesis and its hierarchical regulation. Five cytochrome P450s and three O-methyltransferases were considered as candidates involved in the biosynthesis of CPT and its derivatives, while fifteen transcription factors potentially regulating CPT biosynthesis were screened. These findings provide important clues for elucidating the biosynthetic mechanisms of CPT and its derivatives and substantially contribute to the future production of these anticancer agents with synthetic biology. The generated large-scale multiomics data also provide valuable resources for investigating the functional genomics of the most important CPT-producing plant species-C. acuminata. This article is protected by copyright. All rights reserved.
Article
Cholesterol serves as a key precursor for many high-value chemicals such as plant-derived steroidal saponins and steroidal alkaloids, but a plant chassis for effective biosynthesis of high levels of cholesterol has not been established. Plant chassis have significant advantages over microbial chassis in terms of membrane protein expression, precursor supply, product tolerance, and regionalization synthesis. Here, using Agrobacterium tumefaciens-mediated transient expression technology, Nicotiana benthamiana, and a step-by-step screening approach, we identified nine enzymes (SSR1-3, SMO1-3, CPI-5, CYP51G, SMO2-2, C14-R-2, 8,7SI-4, C5-SD1, and 7-DR1-1) from the medicinal plant Paris polyphylla and established detailed biosynthetic routes from cycloartenol to cholesterol. Specfically, we optimized HMGR, a key gene of the mevalonate pathway, and co-expressed it with the PpOSC1 gene to achieve a high level of cycloartenol (28.79 mg/g dry weight, which is a sufficient amount of precursor for cholesterol biosynthesis) synthesis in the leaves of N. benthamiana. Subsequently, using a one-by-one elimination method we found that six of these enzymes (SSR1-3, SMO1-3, CPI-5, CYP51G, SMO2-2, and C5-SD1) were crucial for cholesterol production in N. benthamiana, and we establihed a high-efficiency cholesterol synthesis system with a yield of 5.63 mg/g dry weight. Using this strategy, we also discovered the biosynthetic metabolic network responsible for the synthesis of a common aglycon of steroidal saponin, diosgenin, using cholesterol as a substrate, obtaining a yield of 2.12 mg/g dry weight in N. benthamiana. Our study provides an effective strategy to characterize the metabolic pathways of medicinal plants that lack a system for in vivo functional verification, and also lays a foundation for the synthesis of active steroid saponins in plant chassis.
Article
Full-text available
Cytochrome P450 monooxygenases (P450s) represent the largest enzyme family of the plant metabolism. Plants typically devote about 1% of the protein-coding genes for the P450s to execute primary metabolism and also to perform species-specific specialized functions including metabolism of the triterpenes, isoprene-derived 30-carbon compounds. Triterpenes constitute a large and structurally diverse class of natural products with various industrial and pharmaceutical applications. P450-catalyzed structural modification is crucial for the diversification and functionalization of the triterpene scaffolds. In recent times, a remarkable progress has been made in understanding the function of the P450s in plant triterpene metabolism. So far, ∼80 P450s are assigned biochemical functions related to the plant triterpene metabolism. The members of the subfamilies CYP51G, CYP85A, CYP90B-D, CYP710A, CYP724B, and CYP734A are generally conserved across the plant kingdom to take part in plant primary metabolism related to the biosynthesis of essential sterols and steroid hormones. However, the members of the subfamilies CYP51H, CYP71A,D, CYP72A, CYP81Q, CYP87D, CYP88D,L, CYP93E, CYP705A, CYP708A, and CYP716A,C,E,S,U,Y are required for the metabolism of the specialized triterpenes that might perform species-specific functions including chemical defense toward specialized pathogens. Moreover, a recent advancement in high-throughput sequencing of the transcriptomes and genomes has resulted in identification of a large number of candidate P450s from diverse plant species. Assigning biochemical functions to these P450s will be of interest to extend our knowledge on triterpene metabolism in diverse plant species and also for the sustainable production of valuable phytochemicals.
Article
Full-text available
Camptothecin (CAM), a complex pentacyclic pyrroloqinoline alkaloid, is the starting material for CAM-type drugs that are well-known antitumor plant drugs. Although many chemical and biological research efforts have been performed to produce CAM, a few attempts have been made to uncover the enzymatic mechanism involved in the biosynthesis of CAM. Enzyme-catalyzed oxidoreduction reactions are ubiquitously presented in living organisms, especially in the biosynthetic pathway of most secondary metabolites such as CAM. Due to a lack of its reduction partner, most catalytic oxidation steps involved in the biosynthesis of CAM have not been established. In the present study, an NADPH-cytochrome P450 reductase (CPR) encoding gene CamCPR was cloned from Camptotheca acuminata, a CAM-producing plant. The full length of CamCPR cDNA contained an open reading frame of 2127-bp nucleotides, corresponding to 708-amino acid residues. CamCPR showed 70 ~ 85% identities to other characterized plant CPRs and it was categorized to the group II of CPRs on the basis of the results of multiple sequence alignment of the N-terminal hydrophobic regions. The intact and truncate CamCPRs with N- or C-terminal His6-tag were heterologously overexpressed in Escherichia coli. The recombinant enzymes showed NADPH-dependent reductase activity toward a chemical substrate ferricyanide and a protein substrate cytochrome c. The N-terminal His6-tagged CamCPR showed 18- ~ 30-fold reduction activity higher than the C-terminal His6-tagged CamCPR, which supported a reported conclusion, i.e., the last C-terminal tryptophan of CPRs plays an important role in the discrimination between NADPH and NADH. Co-expression of CamCPR and a P450 monooxygenase, CYP73A25, a cinnamate 4-hydroxylase from cotton, and the following catalytic formation of p-coumaric acid suggested that CamCPR transforms electrons from NADPH to the heme center of P450 to support its oxidation reaction. Quantitative real-time PCR analysis showed that CamCPR was expressed in the roots, stems, and leaves of C. acuminata seedlings. The relative transcript level of CamCPR in leaves was 2.2-fold higher than that of roots and the stems showed 1.5-fold transcript level higher than the roots. The functional characterization of CamCPR will be helpful to disclose the mysterious mechanisms of the biosynthesis of CAM. The present study established a platform to characterize the P450 enzymes involved in the growth, development, and metabolism of eukaryotic organisms.
Article
Full-text available
Transcriptome sequencing offers a great resource for the study of non-model plants such as Catharanthus roseus, which produces valuable monoterpenoid indole alkaloids (MIAs) via a complex biosynthetic pathway whose characterization is still undergoing. Transcriptome databases dedicated to this plant were recently developed by several consortia to uncover new biosynthetic genes. However, the identification of missing steps in MIA biosynthesis based on these large datasets may be limited by the erroneous assembly of close transcripts and isoforms, even with the multiple available transcriptomes. Secologanin synthases (SLS) are P450 enzymes that catalyze an unusual ring-opening reaction of loganin in the biosynthesis of the MIA precursor secologanin. We report here the identification and characterization in C. roseus of a new isoform of SLS, SLS2, sharing 97 % nucleotide sequence identity with the previously characterized SLS1. We also discovered that both isoforms further oxidize secologanin into secoxyloganin. SLS2 had however a different expression profile, being the major isoform in aerial organs that constitute the main site of MIA accumulation. Unfortunately, we were unable to find a current C. roseus transcriptome database containing simultaneously well reconstructed sequences of SLS isoforms and accurate expression levels. After a pair of close mRNA encoding tabersonine 16-hydroxylase (T16H1 and T16H2), this is the second example of improperly assembled transcripts from the MIA pathway in the public transcriptome databases. To construct a more complete transcriptome resource for C. roseus, we re-processed previously published transcriptome data by combining new single assemblies. Care was particularly taken during clustering and filtering steps to remove redundant contigs but not transcripts encoding potential isoforms by monitoring quality reconstruction of MIA genes and specific SLS and T16H isoforms. The new consensus transcriptome allowed a precise estimation of abundance of SLS and T16H isoforms, similar to qPCR measurements. The C. roseus consensus transcriptome can now be used for characterization of new genes of the MIA pathway. Furthermore, additional isoforms of genes encoding distinct MIA biosynthetic enzymes isoforms could be predicted suggesting the existence of a higher level of complexity in the synthesis of MIA, raising the question of the evolutionary events behind what seems like redundancy.
Article
Full-text available
Camptothecin (CPT) is a kind of modified monoterpene indole alkaloid firstly identified from woody plant Camptotheca acuminata, and its semisynthetic CPT analogs irinothecan and topothecan are clinically used for the treatment of various cancers throughout the world. However, the extraction of CPT from limited natural CPT-producing plant resources couldn’t meet the rapidly increasing market need. The development of plant metabolic engineering provides one alternative way to increase CPT yield by genetic manipulation, which relies on in-depth understanding of the CPT biosynthesis pathway. Several attempts have been also made to obtain CPT by biotechnological approaches such as cell suspensions, endophytic fungi, hairy roots, elicitation as well as metabolic engineering in the past decade. Here, recent advances in knowledge of biosynthesis of CPT, gene isolation, molecular regulation, production improvement and biotechnological methods are summarized and future perspectives are also discussed in this review.
Article
Many oxidation–reduction (redox) enzymes, particularly oxygenases, have roles in reactions that make and break C–C bonds. The list includes cytochrome P450 and other heme-based monooxygenases, heme-based dioxygenases, nonheme iron mono- and dioxygenases, flavoproteins, radical S-adenosylmethionine enzymes, copper enzymes, and peroxidases. Reactions involve steroids, intermediary metabolism, secondary natural products, drugs, and industrial and agricultural chemicals. Many C–C bonds are formed via either (i) coupling of diradicals or (ii) generation of unstable products that rearrange. C–C cleavage reactions involve several themes: (i) rearrangement of unstable oxidized products produced by the enzymes, (ii) oxidation and collapse of radicals or cations via rearrangement, (iii) oxygenation to yield products that are readily hydrolyzed by other enzymes, and (iv) activation of O2 in systems in which the binding of a substrate facilitates O2 activation. Many of the enzymes involve metals, but of these, iron is clearly predominant.
Article
Camptothecin is a monoterpene indole alkaloid (MIA) used to produce semi-synthetic anti-tumor drugs. We investigated camptothecin synthesis in Camptotheca acuminata by combining transcriptome and expression data with reverse genetics, biochemistry, and metabolite profiling. RNAi silencing of enzymes required for the indole and seco-iridoid (monoterpene) components identified transcriptional cross-talk coordinating their synthesis in roots. Metabolite profiling and labeling studies of wild type and RNAi lines identified plausible intermediates for missing pathway steps and demonstrated nearly all camptothecin pathway intermediates are present as multiple isomers. Unlike previously characterized MIA-producing plants, C. acuminata does not synthesize 3-alpha(S)-strictosidine as its central MIA intermediate and instead uses an alternative seco-iridoid pathway that produces multiple isomers of strictosidinic acid. NMR analysis demonstrated that the two major strictosidinic acid isomers are (R) and (S) diastereomers at their glucosylated C21 positions. The presence of multiple diastereomers throughout the pathway is consistent with their use in synthesis before finally being resolved to a single camptothecin isomer after deglucosylation, much as a multi-lane highway allows parallel tracks to converge at a common destination. A model "diastereomer" pathway for camptothecin biosynthesis in C. acuminata is proposed that fundamentally differs from previously studied MIA pathways.
Article
Tracer experiments revealed that (10-3H)-loganin (2) was incorporated into gentiopicroside (3) in Gentiana triflora var. japonica. (7-3H)-2 and (carbo-14C-methoxy)-secolo-ganin (10) were also found to be incorporated into morroniside (4) in Gentiana thunbergii and Cornus officinalis, respectively. The biosynthetic pathway of oleuropein-type glucosides in Olea europaea and Jasminum primulinum was further examined by employing (10-3H)-7-deoxyloganic acid (1), (10-3H)-loganin (2), (carbo-14C-methoxy)-secologanin (10), (8-3H)-and (carbo-3H-methoxy)-kingiside (11), together with (8-3H)-and (carbo-14C-methoxy)-8-epikingiside (18).
Article
The Madagascar periwinkle produces a large palette of Monoterpenoid Indole Alkaloids (MIAs), a class of complex alkaloids including some of the most valuable plant natural products with precious therapeutical values. Evolutionary pressure on one of the hotspots of biodiversity has obviously turned this endemic Malagasy plant into an innovative alkaloid engine. Catharanthus is a unique taxon producing vinblastine and vincristine, heterodimeric MIAs with complex stereochemistry, and also manufactures more than 100 different MIAs, some shared with the Apocynaceae, Loganiaceae and Rubiaceae members. For over 60years, the quest for these powerful anticancer drugs has inspired biologists, chemists, and pharmacists to unravel the chemistry, biochemistry, therapeutic activity, cell and molecular biology of Catharanthus roseus. Recently, the "omics" technologies have fuelled rapid progress in deciphering the last secret of strictosidine biosynthesis, the central precursor opening biosynthetic routes to several thousand MIA compounds. Dedicated C. roseus transcriptome, proteome and metabolome databases, comprising organ-, tissue- and cell-specific libraries, and other phytogenomic resources, were developed for instance by PhytoMetaSyn, Medicinal Plant Genomic Resources and SmartCell consortium. Tissue specific library screening, orthology comparison in species with or without MIA-biochemical engines, clustering of gene expression profiles together with various functional validation strategies, largely contributed to enrich the toolbox for plant synthetic biology and metabolic engineering of MIA biosynthesis.
Article
Iridoids make up a major group of biologically active molecules present in thousands of plant species and one versatile iridoid, secologanin, is a precursor for the assembly of thousands of monoterpenoid indole alkaloids (MIAs) as well as a number of quinoline alkaloids. This study uses bioinformatics to screen large databases of annotated transcripts from various MIA producing plant species to select candidate genes that may be involved in iridoid biosynthesis. Virus induced gene silencing of the selected genes combined with metabolite analyses of silenced plants is then used to identify the 7-deoxyloganic acid-7-hydroxylase (CrDL7H) that is involved in the 3(rd) to last step in secologanin biosynthesis. The silencing of CrDL7H reduced secologanin levels by at least 70% and increased the levels of 7-deoxyloganic acid to over 4 mg/leaf gram fresh weight compared to control plants where this iridoid is not detected. Functional expression of this CrDL7H in yeast confirmed its biochemical activity and substrate specificity studies showed its preference for 7-deoxyloganic acid over other closely related substrates. Together these results suggest that hydroxylation precedes carboxy-O-methylation in the secologanin pathway in Catharanthus roseus. This article is protected by copyright. All rights reserved.