Enzyme and Microbial Technology 86 (2016) 103–116

Contents lists available at ScienceDirect

Enzyme and Microbial Technology

j ournal homepage: www.elsevier.com/locate/emt

Review

Methylation of flavonoids: Chemical structures, bioactivities, progress

and perspectives for biotechnological production

a b a a

Niranjan Koirala , Nguyen Huy Thuan , Gopal Prasad Ghimire , Duong Van Thang ,

a,∗

Jae Kyung Sohng

a

Department of BT-Convergent Pharmaceutical Engineering, Institute of Biomolecule Reconstruction, Sun Moon University, 100, Kalsan-ri, Tangjeonmyun,

Asansi, Chungnam 336-708, Republic of Korea

b

Center for Molecular Biology, Institute of Research and Development, Duy Tan University, K7/25 Quang Trung Street, Haichau District, Danang City, Viet Nam

a r a

t i b s

c l e i n f o t r a c t

Article history: Among the natural products, flavonoids have been particularly attractive, highly studied and become one

Received 9 November 2015

of the most important promising agent to treat cancer, oxidant stress, pathogenic bacteria, inflamma-

Received in revised form 2 February 2016

tions, cardio-vascular dysfunctions, etc. Despite many promising roles of flavonoids, expectations have

Accepted 9 February 2016

not been fulfilled when studies were extended to the in vivo condition, particularly in humans. Instabil-

Available online 11 February 2016

ity and very low oral bioavailability of dietary flavonoids are the reasons behind this. Researches have

demonstrated that the methylation of these flavonoids could increase their promise as pharmaceuti-

Keywords:

cal agents leading to novel applications. Methylation of the flavonoids via theirs free hydroxyl groups

Flavonoids

Bioavailability or C atom dramatically increases their metabolic stability and enhances the membrane transport, lead-

ing to facilitated absorption and highly increased oral bioavailability. In this paper, we concentrated

Pharmaceutical agents

Methylation on analysis of flavonoid methoxides including O- and C-methoxide derivatives in aspect of structure,

Synthetic biology bioactivities and description of almost all up-to-date O- and C-methyltransferases’ enzymatic character-

Metabolic engineering istics. Furthermore, modern biological approaches for synthesis and production of flavonoid methoxides

using metabolic engineering and synthetic biology have been focused and updated up to 2015. This

review will give a handful information regarding the methylation of flavonoids, methyltransferases and

biotechnological synthesis of the same.

© 2016 Elsevier Inc. All rights reserved.

Contents

1. Introduction ...... 104

2. Structure and bioactivity of methylated flavonoids ...... 104

2.1. O-Methylated flavonoids ...... 104

2.2. C-Methylated flavonoids ...... 104

2.3. Several typical methylated flavonoids and their bioactivities ...... 107

2.3.1. Isoflavonoids and their methylation ...... 107

2.3.2. and theirs methylation...... 107

2.3.3. Flavonols and theirs methylation ...... 107

2.3.4. Flavanones and theirs methylation ...... 107

3. Recent biotechnological progress for methylation of flavonoid ...... 109

3.1. Methyltransferaseas a biological tool for synthesis of methylated flavonoid ...... 109

3.2. Synthesis of methylated flavonoids by in vitro enzymatic reaction ...... 109

Corresponding author. Fax: +82 41 544 2919.

E-mail addresses: [email protected] (N. Koirala),

[email protected] (N.H. Thuan), [email protected]

(G.P. Ghimire), [email protected] (D.V. Thang), [email protected]

(J.K. Sohng).

http://dx.doi.org/10.1016/j.enzmictec.2016.02.003

0141-0229/© 2016 Elsevier Inc. All rights reserved.

104 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

3.3. Approaches for modern biotechnological synthesis of methylated flavonoids ...... 109

4. Conclusions and prospects ...... 112

Competing interest ...... 112

Acknowledgements ...... 112

References ...... 112

1. Introduction and biological activity, and since most of the glycosylated prod-

ucts showed only the increase in solubility and a lack of prominent

Polyphenols such as flavonoids, stilbenes are widely presented biological activity (not in all cases though), methylation of these

in plant kingdom and involved in various defense mechanisms pharmaceutically significant flavonoids may give the compounds a

including auto-defending against herbivores, stress tolerance, competitive advantage.

water-lost resistance, etc. Among them flavonoid have gained Methylation of free hydroxyl groups in flavonoids dramatically

much interests due to their importantly medicinal and cos- increases their metabolic stability and enhances their membrane

metic properties [1]. Till date around 8000 naturally occurring transport, facilitating absorption and greater oral bioavailability



flavonoids have been identified and characterized which are abun- [14,15]. Supporting this, 7-hydroxyflavone, 7,4 -dihydroxyflavone,

dantly deposited in vegetables, stems, fruits, seeds, and other and 5,7-dihydroxyflavone () were undetectable in tissue

organs [2]. Structurally, flavonoids contain fifteen carbon atoms levels after administration to rats, whereas the corresponding

in their basic nucleus: two six membraned rings linked with methylated derivatives reached high tissue levels [16]. Mono

a three carbon unit which may or may not be a part of the and dimethylated flavones showed potent antiproliferative activ-

third ring. For convenience, the rings are labeled A, B, and C ity [17]; they inhibited carcinogenic-activating cytochrome P450

[3]. (CYP) transcription and activities [18], benzo[a]pyrene activating

As most plants contain flavonoids, they are considered as enzymes and DNA binding in human bronchial epithelial BEAS-2B

traditional herbs to treat various types of diseases for long cells [19], and aromatase, an important target in hormone-sensitive

time such as increasing in immunological system via anti- cancers [20]. Similarly, 7-O-methyl genistein and 7-O-methyl

oxidant, anti-inflammatory, anti-allergenic, anti-cancer properties daidzein significantly inhibited TNF-␣-induced invasion of HUVECs

[4–6]. Nowadays, several types of flavonoids such as , at 20 ␮M, a concentration at which no cytotoxicity was observed

rutin, , etc. have been extensively used in single or [21]. Furthermore, there have been several reports on compounds

mixed form to make functional food, cosmetic or drug and like rhamnetin [22–24], sakuranetin [25,26] and

widely commercialized whole the world such as Quercetin [27,28]. These compounds are the methylated metabolites of

B5 Plus Complex (viridian), rutin and vitamin C (Lamberts), quercetin, naringenin and apigenin, respectively, and quercetin

etc. is already in the clinical trial phase. These results and current

Naturally, flavonoids are often in the type of glycosylated or research suggests that methylated forms have higher metabolic sta-

methylated form in plants due to those structures are more sta- bility, oral bioavailabity and biological activity than unmethylated

ble, bioavailability as well as bioactivity. Glycosylation of flavonoids forms. The emphasis is the effect of methylation modification on

have been carried out by a biological tool, glycosyltransferase, in original compounds, including increase in metabolic stability and

which the enzyme catalyzes for the attachment of sugar molecule enhancement of pharmaceutical properties. In this review, we have

into aglycone resulting in glycosides [7,8]. In the similar manner, summarized the structure, bioactivities as well as strategies for

methylation of hydroxyl group in flavonoids occurs in the presence biosynthesis of methylated flavonoids using systematic metabolic

of methyltransferase that attaches methyl moieties to aglycone to engineering.

form methoxides. Methylation can occur via oxygen or carbon atom

to form O-methylated or C-methylated compounds, respectively.

2. Structure and bioactivity of methylated flavonoids

Experimental data revealed that methylation of flavonoids resulted

in dramatic change in pharmacological and biochemical proper-

2.1. O-Methylated flavonoids

ties of methylated compound in compared with its parent [9,10].

Thereby, it is one of the most effective ways to modify of natural

O-methylated derivatives are formed via attachment of methyl

products for drug discovery.

group with oxygen of hydroxyl moiety in flavonoid skeleton and

Depending on the reacted substrates, positions and donor

considered as product of post-modification [29]. Due to numer-

groups, glycosylation and methylation may have different effects.

ous hydroxyl groups in flavonoid core, methylation positions

Many promising applications of glycosylated flavonoids were not

of flavonoids are various. For example, structure of several O-

achieved when studies were extended to in vitro biological activ-

methylated flavonoids are presented in Fig. 1A, extracted from

ity tests. For instance, when nonbenzoquinone geldanamycin was

Cuphea and Diplusodon [30], Friesodielsia discolor [31] or Piper mon-

glycosylated, the glycosylated products demonstrated weaker bio-

tealegreanum [32].

logical activity compared to the original aglycone [11]. Additionally,

in our recent preliminary studies, glycosylated genistein was sub-

jected to biological activity tests. Not much improvement was seen 2.2. C-Methylated flavonoids

in its anti-cancer activity, though it has an added advantage of hav-

ing higher solubility than its parent compounds [12]. There are

Numerous C-methylated flavonoids have been mined from

many unpublished results due to a lack of significant biological

plant extract such as in Pisonia grandis roots [33], in some Myri-

activity related to the glycosylation of flavonoids. This “-enhances

taceae [34] or Cleistocalyx operculatus [35] (Fig. 1B). Bioactivities

solubility” tag of glycosylated analogues is true as exemplified by

of C-methylated flavonoid have been checked as neuraminidase

studies focused on the use of sugar conjugation: glycosylated com-

inhibitors for novel influenza H1N1 [35], antioxidant and radical

pounds can greatly enhance drug solubility (up to >2-fold) and

scavenging effect [36,37].

enhance uptake in vitro[13]. As the motive for various modifica-

It will be important to discuss occurrence and biological

tions of natural products like flavonoids is to increase their stability

activities for other selected flavonoids and their methyl con-

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 105

Table 1

Occurrence and bioactivities of several typical methylated flavonoids.

No. Names of compounds Bioactivities Origin References

1 Genkwanin Anti-bacterial, anti-plasmodial, radical Daphene genkwa Sieb. Et. Zucc; [62,27,63,64]

(7-O-methylapigenin) scavenging, chemo preventive and Rosmarinus officinalis L.; Cistus

anti-inflammatory effect in relation with laurifolius L.

the miR-101/MKP-1/MAPK pathway

2 Sakuranetin (7-O- Inhibition of platelet aggregation, a Oryza sativa L., Boesenbergia [128–130]

methylnaringenin) cytotoxic compound to KB nasopharyngeal pandurata, Eriodictyon californicum,

carcinoma cells Piper aduncum

3 Rhamnetin Anti-melanogenesis,inhibits the formation Rhamnus petiolaris, Coriandrum [22,80,82]

(7-O-methylated of ␤-amyloid,enhancing the radio sativum, Syzygium aromaticum,

quercetin) therapeutic efficacy by inhibiting Prunus ceracus

radiation-induced Notch-1 signaling in

lung cancer cell lines

4 Hesperetin Anti-estrogen, inhibition of breast cancer Natsudaidai, Citrofortunella mitis, [131,132]

cell proliferation, delaying tumorigenesis Citrus reticulate, Citrus limon,

Lamium album, Lamium bifidum

Cirillo.

5 Isorhamnetin Inhibition of over-accumulation of Tagetes lucida, Brassica rapa var. [133–135]

triglyceride in HepG2 cells, antioxidant rapa), Solidago virgaurea, Brassica

and cytotoxicity against HepG2 cells, juncea, Ginkgo biloba

inhibition of neutral endopeptidase,

inhibits xanthine oxidase

6 Isosakuranetin Cytotoxic and fungicide properties Citrus sinensis, Citrus x paradisi, [136,137]

Inhibition on CYP1B1, CYP1A2, CYP1A1 Monarda didyma

cytochromes’ activity

7 Dihydrokaempferide Effective antioxidant and radio protectant, Prunus domestica, Salix caprea, [138–140]

prevention of hypertension and significant Brazilian green propolis

decrease in blood pressure, inhibitors of

fungi

8 Kaempferide Inhibition of TRP1 mRNA expression in Kaempferia galanga [141,142]

B16-4A5 cells and pro-coagulant activity in

human monocyte, cytotoxicity against

A549, HeLa and HT-29 cells

 

9 Syringetin (3 ,5 -O- Antiviral activity against HCV JFH-1 Lysimachia congestiflor [143–145]

dimethylmyricetin) J399EM infected in human cells, a

significant induction of differentiation in

MC3T3-E1 mouse calvaria osteoblasts and

osteoblastic 1.19 cell line.



10 Laricitrin (3 -O- Antioxidant activity Red grape, Vaccinium uliginosum [146,147]

dimethylmyricetin)

11 Anti-aromatase and anti-estrogen Robinia pseudoacacia, Turnera [148,149]

activities, used for the treatment of atrial diffusa

fibrillation (AF)

12 Radical-scavenging activity Orthosiphon stamineus and in [150,151]

orange oil

   

13 2 ,4 -Dihydroxy-3 ,5 - Anti-M. tuberculosis H37Rv activity, Campomanesia adamantium, [152,153]



dimethyl-6 - inhibition of KDR tyrosine kinase, Cleistocalyx operculatus

methoxychalcone mitogen-activated protein kinase (MAPK)

and AKT activation of KDR signal

transduction, inhibition of vascular

endothelial HDMEC cells

14 Matteucinol Antitumor activity against HL60, KB, Melastomataceae, Ericaceae, [154,155]

BGC823 and Bel7402cells, cytotoxic Dryopteridaceae, Rhododendron

activity against theHL-60 and SMMC-7721 hainanense

cell lines.

15 Europinidin Used for treatment of diabetes and Plumbag, Ceratostigma [156]

metabolic syndrome, inhibition for

production of hepatitis C virus

16 Hirsutidin Treatment of a cancerous or precancerous Catharanthus roseus [157]

lesion of the skin

17 Petunidin Inhibition of human RNase H and HIV-2 Aronia sp., Amelanchier alnifolia, [158]

RNase H Vitis vinifera, Vitis rotundifolia

18 Rosinidin Inhibition of natural cyclooxygenase Catharanthus roseus, Primula rosea [159,160]

19 Inhibition of horse BchE and STK33 activity Caucasian vetch [161–163]

20 Antihistaminic activity in rat RBL2H3 cells Rice bran [164–166]

assessed as inhibition of DNP-BSA-induced

beta-hexosaminidase release preincubated

Antioxidant activity assessed as DPPH

radical scavenging activity

Potently inhibits cyclooxygenase enzymes

and interferes with intestinal

carcinogenesis in ApcMin mice

21 Tangeritin Appears to counteract the anticancer drug Citrus peels, citrus juices [167,168]

tamoxifen and to suppress the activity of

natural killer cells

Cholesterol lowering agent

Effects againstParkinson’s disease

106 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

Table 1 (Continued)

No. Names of compounds Bioactivities Origin References

22 Inhibitors of CDK9 that induce apoptosis in Scutellaria baicalensis [169–171]

cancer cells by transcriptional suppression

of Mcl-1

Shown pharmacological effects that

indicate wogonin may have anti-tumor

properties. Possess anticonvulsant effects

23 Zapotin Potential anti-carcinogenic effects against Casimiroa edulis [172,173]

isolated colon cancer cells

Potent anticancer activity of zapotin and

suggests a role for zapotin both as a

chemopreventive and a chemotherapeutic

agent against colon cancer

24 Poriol Dose response confirmation for Pseudotsuga menziesii [174,175]

Mcl-1/Noxa interaction inhibitors

HTS to find inhibitors of pathogenic

pemphigus antibodies

25 Lupeol Antiprotozoal, antimicrobial, Syzygium samarangense, Acacia [176,177]

antiinflammatory, antitumor and visco and Abronia villosa.

chemopreventive properties, antimicrobial

26 Betulin Effective against a variety of tumors. Starts Syzygium samarangense, bark of [178–180]

a process of apoptosis and can slow the birch trees, birch sap

growth of several types of tumor cells.

Decreased the lipid contents in serum and

tissues, and increased insulin sensitivity.

Antiviral activity against HIV1 3B in human

H9 cells assessed as inhibition of viral

replication

27 Demethoxymatteucinol Effective concentration against HIV-1 Desmos, Myrica serrata [154,181,182]

replication in H9 lymphocytic cells,

concentration that inhibits uninfected H9

cell growth, antitumor activity against

human HL60 cells, KB cells, BGC823 cells,

Bel7402 cells

  

28 2 ,6 -Dihydroxy-4 - Antifungal and antibacterial Myrica gale, Myrica serrata [37,181]

 

methoxy-3 ,5 - Inhibition of lipid peroxidationinduced by

dimethyldihydrochalcone tert-butyl hydroperoxide

(myrigalone B) Inhibition of peroxidation,scavenging

activity against the diphenylpicrylhydrazyl

(DPPH) radical, and inhibition of enzymatic

lipid peroxidation in linoleic acid

29 Myrigalone Antifungal and antibacterial Syzygium samarangense, Myrica [181,183]   

H/2 ,4 -dihydroxy-6 - (Cladosporiumcucumerinum, Bacillus serrata



methoxy-3 - subtilis, and Escherichia coli)

methyldihydrochalcone Exhibited significant and selective

inhibition against prolyl endopeptidase

30 Desmosflavanone II Inhibition of HIV-1 replication in H9 Desmos cochinchinensis Lour [182,184]

((2S)-7-hydroxy-5- lymphocytic cells, therapeutic index

methoxy-6-methyl-4- determined by ratio of IC50 to EC50 oxo-2-phenyl-2,3- dihydrochromene-8-

carbaldehyde

31 Desmosdumotin C Cytotoxicity activity against bone (HOS), Desmos dumosus, Mitrella kentii [185,186]

breast (MCF) and ovarian (IA9) cancer cell

lines

Gastroprotective effect and

anti-Helicobacter pylori activity

32 (2S)-5-hydroxy-7- Inhibition of influenza A virus Cleistocalyx operculatus [35,182]

methoxy-6,8- neuraminidase, effective concentration

dimethylflavanone against HIV-1 replication in H9

lymphocytic cells

33 Cariphenone B Antioxidant activity against tumor cell Hypericum carinatum [187,188]

lines

34 Rottlerin Inhibitor of protein kinase Cdelta Mallotu philippensis [189,190]

(PKCdelta)

35 Zidovudine Inhibition of reverse-transcriptase Synthesized [191–193]

inhibitor (NRTI), a component of

HIV-treated drug AZT, antiviral activity

against humanHIV 1-infected C8166 cells

and human HIV 13B-infected H9 cell

36 Pisonivanone Inhibition on growth of Mycobacterium Pisonia aculeate [194]

tuberculosis

jugates which may play a significant role in pharmaceutical ities is given in Table 1. Pharmaceutically significant flavonoids

industries in near future. A comprehensive overview of selected and their methylated derivatives are described to some details

methylated flavonoids focusing on their occurrence and bioactiv- herein.

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 107

2.3. Several typical methylated flavonoids and their bioactivities alkaline phosphatase reporter assay), inhibition of important signal

molecules in production of TNF-␣ and IL-6 [61].

2.3.1. Isoflavonoids and their methylation

Isoflavonoids are a very distinctive subgroup of flavonoids that

2.3.2.3. Apigenin. Apigenin-7-O-methylether also known as

are significantly occurred in soybeans and other leguminous plants.

genkwanin is one of the major non-glycosylated flavonoids found

They are found to play an important roles as precursors for the

in some herbs such as Genkwa Flos (Daphene genkwa Sieb. Et. Zucc),

development of phytoalexins during plant micro interactions [38].

rosemary (Rosmarinus officinalis L.) [62] and Cistus laurifolius L. [63].

The metabolism of isoflavonoids initiates with the fixed carbon

Genkwanin has been proven as a multi-fuctional pharmaceutical

gone through the phenylpropanoid pathway. Following multi-

agents such as anti-bacterial, antiplasmodial, radical scavenging,

ple enzymatic processes, phenolic compounds, isoflavonoids are

chemopreventive, anti-inflammatory effect [63,64], inhibitor for

generated [39]. Szkudelska and Nogowski reviewed the effect of

dehydrogenase type 1 in the 17␤-hydroxysteroid pathway [65].

genistein inducing hormonal and metabolic changes, by virtue of

which they can influence various disease pathways [40] or high

2.3.3. Flavonols and theirs methylation

pharmalogical bioactivities such as anti-cancer, reducing of cardio-

2.3.3.1. Myricetin. Myricetin (3,5,7-trihydroxy-2-(3,4,5-

vascular diseases [41,42] prevention of osteoporosis, attenuation

trihydroxyphenyl)-4-chromenone) is a natural flavonol found

of post-menpopausal problems [43] and loss body mass and fat

in fruits, vegetables, tea, strawberries, red wine, and herbs [66]

tissue [44]. Also it has been studied that ingestion of dietary gen-

that involve in the inhibition of superoxide anions or restrict the

estein showed concentration changes of hormones, such as insulin,

bioactivities of xanthine oxidase [67], or suppressed the cancer for-

thyroid hormones, adrenocorticotropic hormone, cortisol and cor-

mation in rat [68]. Further studies mentioned myricetin as inhibitor

ticosterone, and lipid metabolic changes [45]. Similarly, it has been

of myeloperoxidase [69] or reduction of colon carcinogenesis [66].

shown that daidzein exhibits similar effects with those of genistein

Methylated myricetin derivative such as syringetin and

[46]. Furthermore, daidzein restricted bioactivities of enzymes in

laricetrin have been found throughout the plant kingdom [70] and

the biosynthesis of protein and DNA replication in osteoblasts that

play roles as antioxidants, anti-inflammatory, anti-artherosclerotic

influencing on the bone density [47–49]. It is because of these bio-

agents, etc. [71].

logical properties that isoflavonoids find numerous applications

and makes them priceless in nutraceutical as well as cosmetic

applications and are important constituent of various dietary sup-

2.3.3.2. . Kaempferol (3,5,7-trihydroxy-2-(4-

plements, creams, ointments, moisturizing lotions and gels [50].

hydroxyphenyl)-4H-1-benzopyran-4-one) is a flavonoid widely

7-O-methylation of the bioactive isoflavonoids can enhance

occurred in many edible plants and ingredient of traditional

their biological activities. In our recent study, although the 7-O-

medicine [72]. This compound involves in the apoptosis in human

methylation of genistein did not contribute to a better anti-cancer

lung cancer cell [73] or inhibition of cancer cell line under in vitro

and anti-angiogenic activity compared to genistein, the 7-O-

conditions [73,74].

methylation of daidzein provided more elevated chemosensitivity

7-O-Methyl dihydrokaempferol so-called 7-O-methyl aro-

than that of original compound. These results suggest that 7-O-

madendrin is found in plants that effecting on the development

methyl daidzein may have great potential as a chemotherapeutic

of liver carcinoma cells and adipocytes in vitro[75]. Considering all

agent for human cancers, and the regiospecific methylation strat-

these medicinal applications and the importance of methylation

egy could provide a novel starting point for pre-clinical and clinical

on biological activity of kaempferol, more experimental studies

applications of isoflavonoid compounds in cancer therapy [12,21].

are required for discovering the pharmacological effectiveness of

kaempferide in near future.

2.3.2. Flavones and theirs methylation

2.3.2.1. 7,8-Dihydroxyflavone. These flavonoids are significantly 2.3.3.3. Quercetin. Quercetin is a well-known flavonoid and its bio-

present in some parts of plant such as fruits and vegetables [51]. logical activities have been broadly well documented. Even though

It plays role as neurotrophin in mammalian [52], reducing angio- the use of quercetin in cancer prevention and as chemotherapy

genesis [53], a potent agonist against TrkB [54], antioxidant agent, adjuvant [1] as well as their relevance for the well-being of the

resistance against aging-induced morphological changes. In addi- cardiovascular system [76] for neuroprotection [77] and for the

tion, it can reduce the chances of neurodegenerative diseases improvement of cognitive functions has long been known [78], its

induced by ROS via increase the cellular glutathione level [55]. use as a therapeutic is limited because of its poor bioavailability and

Furthermore, 7,8-DHF was proven to be having a vasorelaxing solubility. The solution for this could be methylation of quercetin

and antihypertensive properties [56] suggesting its use in treat- which will be described in the paragraphs below taking rhamnetin

ment of cardiovascular diseases. 7,8-Dihydroxyflavone (flavone) as an example.

and its methylation possess the cytoprotective effect against Rhamnetin, a naturally occurring 7-O-methylated quercetin

oxidative stress by scavenging intracellular ROS and 7-hydroxy-8- is abundantly occurred in Rhamnus petiolaris [79], Coriandrum

methoxyflavone has potent antioxidant activity without affecting sativum (Apiaceae) [80], and Prunus ceracus (Rosaceae) [81]. It pos-

endothelial cell viability even at long period of treatment [57]. sesses strong anti-inflammatory [82], anti-tumor, anti-cholesterol,

anti-bacterial and anti-melanogenesis [83] activities and inhibits

  the formation of ␤-amyloid [23] or enhancing the radiotherapeutic

2.3.2.2. . Luteolin (3 ,4 ,5,7-tetrahydroxyflavone) belongs

efficiency in lung cancer cell lines [24].

to a group of naturally occurring flavonoids that are found widely

in the plant kingdom. Vegetables and fruits such as celery, parsley,

broccoli, onion leaves, carrot, peppers, cabbages, apple skins, and 2.3.4. Flavanones and theirs methylation

chrysanthemum flowers are rich in luteolin [58–60]. Thereby these 2.3.4.1. Naringenin. 7-O-Methylnaringenin also known as saku-

flavonoid are important ingredients of Chinese herbs for treatment ranetin is a chiral flavanone [84] present in rice (Oryza sativa L.)

of hypertension, inflammatory diseases, etc. [51]. [85], finger root (Boesenbergia pandurata) [86], yerba santa (Eriod-



Double methylations of luteolin at 7- and 3 -OH position pro- ictyon californicum) [84], spiked pepper (Piper aduncum) [87] and

duces velutin. Velutin has been demonstrated as a potent inhibitory Populous davidiana [88]. Sakuranetin has been shown engage in

agent in nuclear factor (NF)-␬B activation (as assessed by secreted various plant defense roles due to its anti-bacterial, anti-fungal,

108 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

Fig. 1. (A) Chemical structures of O-methylated flavonoids and (B) C-methylated flavonoids.

and anti-inflammatory activities [89] and cytotoxicity in carcinoma have been extensively studied such as reducing cardiac arrhyth-

cells [87]. mia and infarct size in rats [90] or anti-fungal [91], anti-bacteria

[92], anti-inflamatory [93], enhancing tyrosine activity of B16F10

2.3.4.2. Pinocembrin. Like naringenin, pinocembrin is a type of fla- melanoma cells [94]. Thereby they have become more important

vanone widely spread in vascular plants, and their bioactivities in science and clinical uses. Considering all these medicinal appli-

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 109

cations and the importance of pinocembrin, more experimental SAM-binding domain and metal-dependent catalytic site have been

studies are required for discovering the pharmacological effective- identified in LiOMT (Leptospira interrogans O-methyltransferase)

ness of methylated pinocembrin in near future. [108]. In addition, structure of several enzymes have been analyzed

as wheat flavone O-methyltransferase [109], caffeic acid/5-

hydroxyferulic acid 3/5-O-methyltransferase [110]. However, no

3. Recent biotechnological progress for methylation of detail information on the characterization of C-methyltransferase

flavonoid is has been presented till now. Most of amino acid sequences

are annotated as putative C-methyltransferases being awaited for

3.1. Methyltransferaseas a biological tool for synthesis of studying as presented in Table 2.

methylated flavonoid

3.2. Synthesis of methylated flavonoids by in vitro enzymatic

S-Adenosyl-l-methionine (AdoMet) dependent O- reaction

methyltransferases (OMT) operates by transferring methyl

moietyto a specific hydroxyl group of an acceptor compound Numerous putative O-methyltransferase have been mined from

resulted in the formation of its methyl ether derivative and bacteria (Bacillus, Streptomyces, Listeria, etc.), fungi (Phanerochaete

l

S-adenosyl- -homocysteine [95]. Generally, OMTs acting on the chrysosporium) and plant for characterization of their substrate

natural products are classified into three classes [96]. In particular, as flavonoids. For example, luteolin, luteolin7-O-glucoside, eri-

class I and class II function in methylation of phenolic hydroxyl odictyol, dihydroquercetin, etc. were used as substrate for an

residues, while class III OMTs works on carboxyl groups to obtain O-methyltransferase purified from soybean suspension cell culture

methyl esters. Furthermore, those classes are subdivided by using [111]. In other ways, those were cloned and heterologous expressed

their biochemical properties such as structure, molecular size and in Escherichia coli, or Bacillus, for example, several regiospe-

 

cation requirement, etc. For example, cation dependent OMTs sub- cific flavonoid 3 /5 -O-methyltransferases from tomato [112], or

group of class I (so-called caffeoyl coenzyme A OMTs or CCoAOMTs) Chrysosplenium americanum OMT1 and OMT2 were characterized



includes a group of low molecular mass (23–27 kDa) enzymes and as 3 -O-methylation and 3/5-O-methylation of flavonoid, respec-

class II (also known as caffeic acid OMT or COMT) comprises OMTs tively [113]. This method offers some merits like valuable tools to

with a higher subunit molecular mass (38–45 kDa) and no cation study catalytic mechanism, activity as well as kinetic of enzyme.

dependency [97,98]. However, methyltransferases can simply be Furthermore, product is formed regio-selectively and the forma-

catagorized as O-methyltransferase (OMT), N-methyltransferase tion of by-products are extremely less with lower waste emission

(NMT) and C-methyltransferase (CMT) based on their target and energy requirements. However, it requires highly purified sub-

attachments such as oxygen, nitrogen and carbon, respectively strates, hard to scale up the reaction, thus mostly used in the

[99]. Up to now, OMTs are found ubiquitously in nature. Most laboratory.

plant-originated OMTs are characterized to use phenolic com-

pounds like stilbene and flavonoids as their substrate [57,100,101]. 3.3. Approaches for modern biotechnological synthesis of

Five highly conserved regions are proposed as a signature for plant methylated flavonoids

O-methyltransferases, two of which (regions I and IV) are believed

to be involved in S-adenosyl-l-methionine and metal binding, Although much information on biosynthetic pathways of plant

respectively [95]. However, just a few microorganisms-originated methylated flavonoids have been obtained so far it is difficult to

OMTs and their working mechanisms have been biochemically carry out mutagenesis or optimization for production of secondary

characterized. Biochemical and molecular characterization of metabolites using plants as direct hosts due to their huge size

several OMTs have been outlined as in Table 2. genome and complex regulatory network. This resulted in the wide

To investigate the structure-activity relationship, several spreading of direct extraction from plant and chemical synthesis of

flavonoid methyltransferases have been overexpressed, crys- desired compounds before biotechnology still less developed. How-

tallized and analyzed the molecular structure, for instance, ever, directed extraction method is restricted by type of plant, low

2+

Bacillus cereus BcOMT2 – a flavonoid Mg – dependent O- concentration of methylated flavonoids as well as their mixture

methyltransferase [102,103]. In another study, modeling of wheat in natural resource such that this method requires many tedious

(Triticuma estivum L.) O-methyltransferase (TaOMT2), a experiment skills and facilities [114–116]. Chemical synthesis also

O-methyltransferase. Stepwise tricetin methylation was proven often occurs following severe and complex reaction conditions and

via involvement of deprotonation of its hydroxyl groups by a produces many un-wanted by-products leading to impenetrability

His262-Asp263 pair. Futhermore Val309, a conserved amino acid in purification, low yield of product, for example chemical synthesis

in a number of graminaceous flavone OMTs, was determined the of silybin glycoside at C-23 [117].

enzyme TaOMT2 fortricetin as the preferred substrate [104]. In the Biosynthetic methods based on the reconstruction of recom-

similar manner, amino acid of rice-based flavonoid OMT (ROMT9) binant plasmids harboring genes from various sources and uses

was aligned with the Medicago truncatula O-methyltransferase genetically engineered E. coli as host leading to generation of

(COMT) and resulted several specific amino acid residues regard- numerous natural and un-natural products. Such methods provide

ing enzymatic activities such as Asn-275, His-328 [105]. Recently, several advantages such as high yield of product owing to substrate-

Podospora anserina PaMTH1, a putative O-methyltransferase is specific enzyme, easily-occurred reaction, less type of secondary

objective for studying its structure and function. The authors metabolites produced by E. coli limiting formation of by-products.

showed that PaMTH1 is responsible for transferring of the methyl In addition, genetically engineered E. coli is straightforwardly con-

group from SAM to one hydroxyl group of the myricetin in a cation- trolled due to its simple genetical machine. Furthermore, it is easy

dependent manner [106]. to scale up the production of desired compound by using fermenter

Studying on crystal structure of ChOMT (chalcone SAM- [1,8,118].

dependent O-methyltransferase) and IOMT (isoflavone SAM- Most of methylated derivatives of flavonoids can be produced by

dependent O-methyltransferase) provided a structural basis for biotransformation of flavonoids. For example, two types of Strep-

understanding the substrate specificity of the diverse family of tomyces were used for methylation of genistein [119]. Kim et al.

plant OMTs and facilitates the engineering of novel activities in reported the biotransformation of apigenin and quercetin using

this extensive class of natural product biosynthetic enzymes [107]. regioselective 7-O-methyltransferase POMT7 having more than

110 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

Table 2

Origin, accession numbers and biochemical properties of flavonoid methyltransferases.

No. Origins Accession Protein size in Enzymatic activities References

number length (aa)

1 Hordeum vulgare subsp. CAA54616 390 Flavonoid 7-O-methyltransferase, [195,196]

vulgare kinetic parameters of KSAM = 10.9 ␮M,

Kapigenin = 4.6 ␮M, Vmax = 0.45 kat/g

2 Hordeum vulgare subsp. ABQ58825 356 Flavone-specific O-methyltransferase. Prefer [197]

 

vulgare substrate flavone tricetin at 3 ,5 -O position

into tricin

  

3 Triticuma estivum ABB03907 356 Flavonoid O-methyltransferase (prefer 3 , 4 , 5 [198]

position of flavone in to mono, di,tri methyl ether)



4 Glycine max C6TAY1 358 Flavonoid 4 -O-methyltransferase [29]

 

5 Vitis vinifera NP 001290011 235 3 ,5 -O-Methyltransferase showing strong [179,180]

preference foranthocyanins and glycosylated flavonols



6 Catharanthus roseus AAR02419 359 Flavonoid 4 -O-methyltransferase [201]



7 Catharanthus roseus AAM97497 348 Sequential methylations at the 3 - and [202]



5 -positions of the B-ring in myricetin

(flavonol) and

dihydromyricetin(dihydroflavonol)

Preferred substrates flavonol glycosides and

anthocyanins

8 Theobroma cacao EOY28137 380 Flavonoid O-methyltransferase [203]

9 Zea mays NP 001106047 364 Catechol and flavonoid O-methyltransferase [204]

10 Mesembryanthemum AAN61072 237 Flavonoids and caffeoyl-CoA [205] crystallinum



11 Glycyrrhiza echinata Q84KK6 367 Isoflavone 4 -O-methyltransferase [206,207]



12 Lechevalieriaaero Q8KZ94 283 Rebeccamycin sugar 4 -O-methyltransferase [208,209]

colonigenes

13 Synthetic construct AFQ94040 368 Monolignol 4-O-methyltransferase [210]

14 Triticuma estivum Q84N28 360 Caffeic acid O-methyltransferase [211]

15 Oryza sativa Japonica ABB90678 368 O-Methyltransferase [212]

Group

16 Medicago sativa 1FP1 D 372 Chalcone O-methyltransferase. [107]

17 Streptomyces peucetius Q06528 356 Carminomycin 4-O-methyltransferase DnrK [193,194]

18 Streptomyces avermitilis BAB69281 359 7-O-Methyltransferase [215]

19 Streptomyces peucetius AGU42206 223 Flavonoids O-methyltransferase [57]

ATCC 27952

20 Marinithermus AEB11491 395 C-Methyltransferase [216]

hydrothermalis DSM

14884

21 Meiothermus silvanus ADH64788 391 C-Methyltransferase [217]

DSM 9946

22 Haliscomenobacterhydrosis AEE53273 433 C-Methyltransferase [218]

DSM 1100

23 Marinithermus AEB11490 409 C-Methyltransferase [216]

hydrothermalis DSM

14884

24 Thermomonospora ACY98305 405 C-Methyltransferase [219]

curvata DSM 43183

25 Calditerrivibrio ADR19070 408 C-Methyltransferase [219]

nitroreducens DSM

19672

90% conversion [23]. Through the combination of optimized POMT7 to this research, a series of flavonoid biosynthetic gene has been

expression and cofactor production, the production of rhamnetin cloned into recombinant plasmids to convert p-coumaric acid into

(7-O-methyl-quercetin) was increased upto 111 mg/L via using naringenin and dihydrokaempferol. Simultanously, biosynthetic

genetically engineered E. coli as host [120]. The same group genes for enhanced production of malonyl-CoA (a precursor of chal-

had characterized another regiospecific 7-O-methyltyransferase cone synthase) was also reconstructed to increase the heterologous

SaOMT2 showing the conversion of quercetin as 119% compared to production of targeted flavonoids. Later on, 7-O-MT (Streptomyces

naringenin [121]. Biochemically, biosynthesis of those compounds avermitilis MA4680) and flavanone-3-hydroxylase has been used

have been illustrated in Fig. 2. to produce sakuranetin as intermediate and finally 7-O-methyl-

Our group has recently optimized the conditions for pro- aromadendrin. Also, the authors can generate dihydrokaempferol

duction of methylated derivatives of pharmaceutically important using naringenin as substrate before converting this compound

flavonoids using engineered E. coli. Through the combination of into final 7-O-methyl-aromadendrin. The yield of 7-OMA depended

optimized conditions and cofactor production, the maximum yield on fed concentration of p-coumaric acid and achieved 30 mg/L

of 7-O-methyl genistein was 164 ␮M (46.74 mg/L) and 7-O-methyl (99.2 ␮M) after 24 h [122]. Rational design was used for enhanced

daidzein was 382 ␮M (102.75 mg/L) when 200 ␮M of genistein and intracellular tyrosine, a precursor for synthesis of chalcone, was

400 ␮M of daidzein was supplemented in the fed batch culture strategy for production of flavonoid by Kim et al. Consequently,

[21]. To add more data to the current research on methylated com- they used the similar biosynthetic genes of flavonoid such as

pounds, recently, system metabolic engineering strategy has been tyrosine ammonialyase (TAL), 4-coumaroyl CoA ligase (4CL), chal-

applied to produce 7-O-methyl-aromadendrin in E. coli. Regarding cone synthase (CHS) to generate naringenin as an intermediate.

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 111

OH OH

OH

HOOC H OOC

HOOC Cinnamic acid p-coumaric acid Caffeic acid

4CL 4CL 4CL OH OCH O- S CoA OH 3 ACS HO O C O C O CoAS HO O

CH3 CH3 SOMT2

O OH ACC Acid-CoA complex OH Acetate Acetyl-CoA OH O OH O

Kaempferol Kaempferide

O O

CoA

HO S CHS FLS OH 3x Malonyl-CoA OH OH OCH3 HO O HO OH H3CO O HO O SaOMT2 OH OH

OH O

S OH O OH O

O

OH O M H Dihydrokaempferol

T F3 7-O-methyl 2 Naringenin chalcone (Aromadendrin) Ponciretin aromadendrin CHI OH F3'H OH OH HO O HO O H CO O OH 3 NOMT

OH OCH3 SaOM T2 OH O OH O

OH O OH

Naringe nin Dihydroquercetin

7-O-methy l-naringein

(Taxi folin) H3CO O (Sakuran etin) NA

IFS DP T2 OH FNS M H aO OH O CRP FLS +S T9 M Rhamnazin NA RO HO O

OH DP OH OH

+ OH HO O OH OH

OH O HO O H3CO O

POMT7 OH O Genistei n OH OH

Apigenin OH O SaOMT2 OH O

Quercetin 7-O-meth yl-quercetin

SaOMT2 SaOMT2 POMT7 (Rhamneti n) OCH3 OH ROMT9 OH H3CO O H3CO O HO O OH OH O OH

OH O OH O

7- O-methyl-genistein

7-O-methyl-apigenin 3'-O-methyl-quercet in

(genkw anin) (Iso-rhamnetin)

Fig. 2. Metabolic engineering for biological synthesis of various methylated flavonoid. CHS: chalcone synthase, CHI: chalcone isomerase, IFS: isoflavone synthase, 4CL:



4-coumarate-CoA ligase, RS: stilbene synthase (STS), FLS: flavonol synthase, F3’H: flavonoid-3 -hydroxylase, F3H: flavanone-3-hydroxylase, ACS: acetyl-CoA synthase, ACC:

acetyl-CoA carboxylase, CRP: cytochrome P450, FNS: flavone synthase, POMT7: apigenin-7-O-methyltransferase; SaOMT2: Streptomyces avermitilis O-methyltransferase;

NOMT: naringenin O-methyltransferase; SOMT2: soybean O-methyltransferase; ROMT9: rice O-methyltransferase.

However, in order to increase the supply the coenzyme A (CoA), CoA, the yields of naringenin and pinocembrin reached about

one gene (icdA, isocitrate dehydrogenase) was deleted. Finally, 60 mg/L [124]. Later on, introduction of Photorabdus luminescens –

O-methyltransferase (OMT) was applied to produce sakuranetin originated acetyl – CoA carboxylase (ACC) and biotin ligase in com-



(7-O-methyl-naringenin) and ponciretin (4 -O-methyl naringenin) bination with redesigned acetate assimilation pathways in E. coli

with maximal yield of 42.5 mg/L and 40.1 mg/L, respectively [123]. led to much improvement of those flavonoids during 36 h of cul-

Miyahisa et al. constructed recombinants harboring PAL (pheny- ture. And, yields of pinocembrin, naringenin and eriodictyol were

lalanine ammonia-lyase), ScCCL (cinnamate/coumarate:CoA lig- achieved as 429 mg/L, 119 mg/L, and 52 mg/L, respectively [125].

ase), CHS (chalcone synthase) and CHI (chalcone isomerase) genes Recently, Wu et al. reported a biosynthesis pathway of pinocem-

from different sources and heterologously expressed in E. coli. This brin from glucose using system metabolic engineering in E. coli.

resulted in successful production of (2S)-naringenin from tyro- In particular, they designed recombinant vectors harboring genes

sine and (2S)-pinocembrin from phenylalanine. Furthermore by encoding for converting of glucose, synthesis of phenylalanine,

introduction of acetyl-CoA carboxylase (ACC) to enhance malonyl- phenylpropanoid, chalcone, etc. and a recombinant vector support-

112 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

ing for intracellular system balance. It resulted in production of [5] L. Yochum, L.H. Kushi, K. Meyer, A.R. Folsom, Dietary flavonoid intake and

risk of cardiovascular disease in postmenopausal women, Am. J. Epidemiol.

pinocembrin with yield of 40.02 mg/L in minimal media [126]. By

149 (1999) 943–949, http://dx.doi.org/10.1017/S0007114508945694.

using similar strategy but focus on proper modulation of the tar-

[6] S. Zhang, X. Yang, R.A. Coburn, M.E. Morris, Structure activity relationships

get genes and the supply of the co-substrate, Kim’s group could get and quantitative structure activity relationships for the flavonoid-mediated

inhibition of breast cancer resistance protein, Biochem. Pharmacol. 70

much better accumulation of pinocembrin in E. coli (97 mg/L) [127].

(2005) 627–639, http://dx.doi.org/10.1016/j.bcp.2005.05.017.

Methylation and subsequent glycosylation have been proven

[7] R.W. Gantt, P. Peltier-Pain, J.S. Thorson, Enzymatic methods for

as strong strategy to generate novel derivatives and may alter glyco(diversification/randomization) of drugs and small molecules, Nat.

Prod. Rep. 28 (2011) 1811–1853, http://dx.doi.org/10.1039/c1np00045d.

bioactivities of parent compounds. Our group had successfully syn-

[8] N.H. Thuan, R.P. Pandey, T.T.T. Thuy, J.W. Park, J.K. Sohng, Improvement of

thesized several glycosyl flavonoid methoxide derivatives after

␣ l

regio-specific production of myricetin-3-O- - -rhamnoside in engineered

modification of 7,8-dihydroxyflavone using the combination of Escherichia coli, Appl. Biochem. Biotechnol. 171 (2013) 1956–1967, http://

engineered metabolic microbial system and in vitro systems [57]. dx.doi.org/10.1007/s12010-013-0459-9.

[9] H. Cao, X. Jing, D. Wu, Y. Shi, Methylation of genistein and kaempferol

Biological activities of those compounds are under progress of eval-

improves their affinities for proteins, Int. J. Food Sci. Nutr. 64 (2013)

uation. This strategy may prove to be a lethal modification having

437–443, http://dx.doi.org/10.3109/09637486.2012.759186.

a double advantage of enhanced activities and solubility as well. [10] T. Walle, Methylation of dietary flavones greatly improves their hepatic

metabolic stability and intestinal absorption, Mol. Pharm. 4 (2007) 826–832,

http://dx.doi.org/10.1021/mp700071d.

4. Conclusions and prospects [11] C.Z. Wu, J.H. Jang, M. Woo, J.S. Ahn, J.S. Kim, Y.S. Hong, Enzymatic

glycosylation of nonbenzoquinone geldanamycin analogs via Bacillus

UDP-glycosyltransferase, Appl. Environ. Microbiol. 78 (2012) 7680–7686,

Due to special roles in pharmacology, dietary supplement as

http://dx.doi.org/10.1128/AEM.02004-12.

well as cosmetic, flavonoids have been extensively studied and

[12] N. Koirala, R.P. Pandey, D.V. Thang, H.J. Jung, J.K. Sohng, Glycosylation and

become one of the most important promising agent to treat can- subsequent malonylation of isoflavonoids in E. coli: strain development,

production and insights into future metabolic perspectives, J. Ind. Microbiol.

cer, oxidant stress, pathogenic bacteria, etc. There are numerous

Biotechnol. 41 (2014) 1647–1658, http://dx.doi.org/10.1007/s10295-014-

flavonoid-based commercial products that have been extensively 1504-6.

marketed for human life such as Trimology, Flavonoid complex, [13] C. Fernández, O. Nieto, E. Rivas, G. Montenegro, J.A. Fontenla, A.

Fernández-Mayoralas, Synthesis and biological studies of glycosyl dopamine

Lipod Flavonoid Plus trademark names.

derivatives as potential antiparkinsonian agents, Carbohydr. Res. 327 (2000)

In this article, we reviewed several methylated flavonoids,

353–365, http://dx.doi.org/10.1016/S0008-6215(00)00073-2.

derivatives bearing critically biological and pharmaceutical activ- [14] X. Wen, T. Walle, Methylated flavonoids have greatly improved intestinal

absorption and metabolic stability, Drug Metab. Dispos.: Biol. Fate Chem. 34

ities as described. The classification of methylated flavonoid’s

(2006) 1786–1792, http://dx.doi.org/10.1124/dmd.106.011122.

knowledge including chemical structures, activities and biologi-

[15] T. Walle, X. Wen, U.K. Walle, Improving metabolic stability of cancer

cal synthesis methods allows us to get an extensive understanding chemoprotective polyphenols, Exp. Opin. Drug Metab. Toxicol. 3 (2007)

379–388, http://dx.doi.org/10.1517/17425255.3.3.379.

of their nature and outline the novel strategies to exploit theirs

[16] T. Walle, N. Ta, T. Kawamori, X. Wen, P.A. Tsuji, U.K. Walle, Cancer

applications. The advances in the fields of genetic and metabolic

chemopreventive properties of orally bioavailable flavonoids-methylated

engineering has made more convenient to merge various different versus unmethylated flavones, Biochem. Pharmacol. 73 (2007) 1288–1296,

pathways inside a single cells. Though, we do not have a good expla- http://dx.doi.org/10.1016/j.bcp.2006.12.028.

[17] A. Murakami, K. Koshimizu, H. Ohigashi, S. Kuwahara, W. Kuki, Y. Takahashi,

nation for the different results observed, and we still believe that

et al., Characteristic rat tissue accumulation of , a chemopreventive

it is a fertile area for further study such as structure activity rela-

polymethoxyflavonoid, in comparison with luteolin, BioFactors (Oxford

tionship. To conclude; methylated flavonoids could be produced in Engl.) 16 (2002) 73–82, http://dx.doi.org/10.1002/biof.5520160303.

[18] K.L. Morley, P.J. Ferguson, J. Koropatnick, and nobiletin induce G1

industrial scale in future and are very promising agents to be used

cell cycle arrest but not apoptosis in human breast and colon cancer cells,

as a pharmaceutical, cosmeceutical and nutraceutical agents. The

Cancer Lett. 251 (2007) 168–178, http://dx.doi.org/10.1016/j.canlet.2006.11.

future of this class of flavonoid derivative is certainly brighter. 016.

[19] P.A. Tsuji, T. Walle, Benzo[a]pyrene-induced cytochrome P450 1A and DNA

binding in cultured trout hepatocytes—inhibition by plant polyphenols,

Competing interest Chem. Biol. Interact. 169 (2007) 25–31, http://dx.doi.org/10.1016/j.cbi.2007.

05.001.

[20] N. Ta, T. Walle, Aromatase inhibition by bioavailable methylated flavones, J.

The authors declare that they have no competing interests.

Steroid Biochem. Mol. Biol. 107 (2007) 127–129, http://dx.doi.org/10.1016/j.

jsbmb.2007.01.006.

Acknowledgements [21] N. Koirala, N.H. Thuan, G.P. Ghimire, H.J. Jung, T.J. Oh, J.K. Sohng, Metabolic

engineering of E. coli for the production of isoflavonoid-7-O-methoxides and

their biological activities, Biotechnol. Appl. Biochem. (2015), http://dx.doi.

This work was supported by grant from the Next-Generation org/10.1002/bab.1452.

BioGreen 21 Program (SSAC, grant#: PJ0094832), Rural Develop- [22] K. Igarashi, M. Ohmuma, Effects of isorhamnetin, rhamnetin, and quercetin

on the concentrations of cholesterol and lipoperoxide in the serum and liver

ment Administration, Republic of Korea and by Vietnam National

and on the blood and liver antioxidative enzyme activities of rats, Biosci.

Foundation for Science and Technology Development (NAFOSTED)

Biotechnol. Biochem. 59 (1995) 595–601, http://dx.doi.org/10.1271/bbb.59.

under grant number: 106-NN.02-2014.25. 595.

[23] B.G. Kim, H. Kim, H.G. Hur, Y. Lim, J.H. Ahn, Regioselectivity of

7-O-methyltransferase of poplar to flavones, J. Biotechnol. 126 (2006)

References 241–247, http://dx.doi.org/10.1016/j.jbiotec.2006.04.019.

[24] J. Kang, E. Kim, W. Kim, K.M. Seong, H. Youn, J.W. Kim, et al., Rhamnetin and

[1] D. Simkhada, E. Kim, H.C. Lee, J.K. Sohng, Metabolic engineering of cirsiliol induce radiosensitization and inhibition of epithelial-mesenchymal

Escherichia coli for the biological synthesis of 7-O-xylosyl naringenin, Mol. transition (EMT) by miR-34a-mediated suppression of Notch-1 expression

Cells 28 (2009) 397–401, http://dx.doi.org/10.1007/s10059-009-0135-7. in non-small cell lung cancer cell lines, J. Biol. Chem. 288 (2013)

[2] F. Ververidis, E. Trantas, C. Douglas, G. Vollmer, G. Kretzschmar, N. 27343–27357, http://dx.doi.org/10.1074/jbc.M113.490482.

Panopoulos, Biotechnology of flavonoids and other [25] J. Orjala, A.D. Wright, H. Behrends, G. Folkers, O. Sticher, H. Rüegger, et al.,

phenylpropanoid-derived natural products. Part I: chemical diversity, Cytotoxic and antibacterial dihydrochalcones from Piper aduncum, J. Nat.

impacts on plant biology and human health, Biotechnol. J. 2 (2007) Prod. 57 (1994) 18–26, http://dx.doi.org/10.1021/np50103a003.

1214–1234, http://dx.doi.org/10.1002/biot.200700084. [26] A.P. Danelutte, J.H.G. Lago, M.C.M. Young, M.J. Kato, Antifungal flavanones

[3] T.P. Kondratyuk, J.M. Pezzuto, Natural product polyphenols of relevance to and prenylated hydroquinones from Piper crassinervium Kunth,

human health, Pharm. Biol. 42 (2004) 46–63, http://dx.doi.org/10.3109/ Phytochemistry 64 (2003) 555–559, http://dx.doi.org/10.1016/S0031-

13880200490893519. 9422(03)00299-1.

[4] E. Middleton, C. Kandaswami, T.C. Theoharides, The effects of plant [27] F. Cottiglia, G. Loy, D. Garau, C. Floris, M. Casu, R. Pompei, et al.,

flavonoids on mammalian cells: implications for inflammation, heart Antimicrobial evaluation of coumarins and flavonoids from the stems of

disease, and cancer, Pharmacol. Rev. 52 (2000) 673–751, http://dx.doi.org/ Daphne gnidium L, Phytomed.: Int. J. Phytother. Phytopharmacol. 8 (2001)

10.1021/jf8006568. 302–305, http://dx.doi.org/10.1078/0944-7113-00036.

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 113

[28] A.R. Kim, Y.N. Zou, T.H. Park, K.H. Shim, M.S. Kim, N.D. Kim, et al., Active [54] S.W. Jang, X. Liu, M. Yepes, K.R. Shepherd, G.W. Miller, Y. Liu, et al., A

components from Artemisia iwayomogi displaying ONOO(−) scavenging selective TrkB agonist with potent neurotrophic activities by

activity, Phytother. Res.: PTR 18 (2004) 1–7, http://dx.doi.org/10.1002/ptr. 7,8-dihydroxyflavone, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 2687–2692,

1358. http://dx.doi.org/10.1073/pnas.0913572107.

[29] D.H. Kim, B.G. Kim, Y. Lee, J.Y. Ryu, Y. Lim, H.G. Hur, et al., Regiospecific [55] J. Chen, K.W. Chua, C.C. Chua, H. Yu, A. Pei, B.H.L. Chua, et al., Antioxidant

methylation of naringenin to ponciretin by soybean O-methyltransferase activity of 7,8-dihydroxyflavone provides neuroprotection against

expressed in Escherichia coli, J. Biotechnol. 119 (2005) 155–162, http://dx. glutamate-induced toxicity, Neurosci. Lett. 499 (2011) 181–185, http://dx.

doi.org/10.1016/j.jbiotec.2005.04.004. doi.org/10.1016/j.neulet.2011.05.054.

[30] A. Salatino, M.L.F. Salatino, D.Y.A.C. dos Santos, M.C.B. Patrício, Distribution [56] R. Huai, X. Han, B. Wang, C. Li, Y. Niu, R. Li, et al., Vasorelaxing and

and evolution of secondary metabolites in Eriocaulaceae, Lythraceae and antihypertensive effects of 7,8-dihydroxyflavone, Am. J. Hypertens. 27

Velloziaceae from campos rupestres, Genet. Mol. Biol. 23 (2000) 931–940, (2014) 750–760, http://dx.doi.org/10.1093/ajh/hpt220.

http://dx.doi.org/10.1590/S1415-47572000000400038. [57] N. Koirala, R.P. Pandey, P. Parajuli, H.J. Jung, J.K. Sohng, Methylation and

[31] U. Prawat, D. Phupornprasert, A. Butsuri, A.W. Salae, S. Boonsri, P. subsequent glycosylation of 7,8-dihydroxyflavone, J. Biotechnol. 184 (2014)

Tuntiwachwuttikul, Flavonoids from Friesodielsia discolor, Phytochem. Lett. 128–137, http://dx.doi.org/10.1016/j.jbiotec.2014.05.005.

5 (2012) 809–813, http://dx.doi.org/10.1016/j.phytol.2012.09.007. [58] M.L. Neuhouser, Dietary flavonoids and cancer risk: evidence from human

[32] H. Da, M. De, M.C. De, Three new compounds form piper montealegreanum population studies, Nutr. Cancer 50 (2004) 1–7, http://dx.doi.org/10.1207/

yuncker (Piperaceae), J. Braz. Chem. Soc. 22 (2011) 1610–1615, http://dx. s15327914nc5001 1.

doi.org/10.1590/S0103-50532011000800026. [59] K.H. Miean, S. Mohamed, Flavonoid (myricetin, quercetin, kaempferol,

[33] S. Sutthivaiyakit, C. Seeka, N. Wetprasit, P. Sutthivaiyakit, C-methylated luteolin, and apigenin) content of edible tropical plants, J. Agric. Food Chem.

flavonoids from Pisonia grandis roots, Phytochem. Lett. 6 (2013) 407–411, 49 (2001) 3106–3112, http://dx.doi.org/10.1021/jf000892m.

http://dx.doi.org/10.1016/j.phytol.2013.05.001. [60] M.A. Gates, S.S. Tworoger, J.L. Hecht, I. De Vivo, B. Rosner, S.E. Hankinson, A

[34] E. Wollenweber, R. Wehde, M. Dörr, G. Lang, J.F. Stevens, prospective study of dietary flavonoid intake and incidence of epithelial

C-methyl-flavonoids from the leaf waxes of some Myrtaceae, ovarian cancer, Int. J. Cancer 121 (2007) 2225–2232, http://dx.doi.org/10.

Phytochemistry 55 (2000) 965–970, http://dx.doi.org/10.1016/S0031- 1002/ijc.22790.

9422(00)00348-4. [61] C. Xie, J. Kang, Z. Li, A.G. Schauss, T.M. Badger, S. Nagarajan, et al., The ac¸ aí

[35] T.T. Dao, B.T. Tung, P.H. Nguyen, P.T. Thuong, S.S. Yoo, E.H. Kim, et al., flavonoid velutin is a potent anti-inflammatory agent: blockade of

C-methylated flavonoids from Cleistocalyx operculatus and their inhibitory LPS-mediated TNF-␣ and IL-6 production through inhibiting NF-␬B

effects on novel influenza A (H1N1) neuraminidase, J. Nat. Prod. 73 (2010) activation and MAPK pathway, J. Nutr. Biochem. 23 (2012) 1184–1191,

1636–1642, http://dx.doi.org/10.1021/np1002753. http://dx.doi.org/10.1016/j.jnutbio.2011.06.013.

[36] L. Mathiesen, K.E. Malterud, R.B. Sund, Antioxidant activity of fruit exudate [62] G. Altinier, S. Sosa, R.P. Aquino, T. Mencherini, R. Della Loggia, A. Tubaro,

and C-methylated dihydrochalcones from Myrica gale, Planta Med. 61 Characterization of topical anti-inflammatory compounds in Rosmarinus

(1995) 515–518, http://dx.doi.org/10.1055/s-2006-959360. officinalis L, J. Agric. Food Chem. 55 (2007) 1718–1723, http://dx.doi.org/10.

[37] K.E. Malterud, O.H. Diep, R.B. Sund, C-methylated dihydrochalcones from 1021/jf062610+.

Myrica gale L.: effects as antioxidants and as scavengers of [63] S.K. Sadhu, E. Okuyama, H. Fujimoto, M. Ishibashi, E. Yesilada, Prostaglandin

1,1-diphenyl-2-picrylhydrazyl, Pharmacol. Toxicol. 78 (1996) 111–116, inhibitory and antioxidant components of Cistus laurifolius, a Turkish

http://dx.doi.org/10.1111/j.1600-0773.1996.tb00190.x. medicinal plant, J. Ethnopharmacol. 108 (2006) 371–378, http://dx.doi.org/

[38] T. Aoki, T. Akashi, S. Ayabe, Flavonoids of leguminous plants: structure, 10.1016/j.jep.2006.05.024.

biological activity, and biosynthesis, J. Plant Res. 113 (2000) 475–488, [64] Y. Gao, F. Liu, L. Fang, R. Cai, C. Zong, Y. Qi, Genkwanin inhibits

http://dx.doi.org/10.1007/PL00013958. proinflammatory mediators mainly through the regulation of

[39] B. Weisshaar, G.I. Jenkins, Phenylpropanoid biosynthesis and its regulation, miR-101/MKP-1/MAPK pathway in LPS-activated macrophages, PLoS One 9

Curr. Opin. Plant Biol. 1 (1998) 251–257, http://dx.doi.org/10.1016/S1369- (2014) e96741, http://dx.doi.org/10.1371/journal.pone.0096741.

5266(98)80113-1. [65] P. Brozic, P. Kocbek, M. Sova, J. Kristl, S. Martens, J. Adamski, et al.,

[40] K. Szkudelska, L. Nogowski, Genistein-a dietary compound inducing Flavonoids and cinnamic acid derivatives as inhibitors of 17

hormonal and metabolic changes, J. Steroid Biochem. Mol. Biol. 105 (2007) -hydroxysteroid dehydrogenase type 1, Mol. Cell. Endocrinol. 301 (2009)

37–45, http://dx.doi.org/10.1016/j.jsbmb.2007.01.005. 229–234, http://dx.doi.org/10.1016/j.mce.2008.09.004.

[41] R.A. Dixon, D. Ferreira, Genistein, Phytochemistry 60 (2002) 205–211, [66] J.M. Harnly, R.F. Doherty, G.R. Beecher, J.M. Holden, D.B. Haytowitz, S.

http://dx.doi.org/10.1016/S0031-9422(02)00116-4. Bhagwat, et al., Flavonoid content of U.S. fruits, vegetables, and nuts, J. Agric.

[42] H. Si, D. Liu, Phytochemical genistein in the regulation of vascular function: Food Chem. 54 (2006) 9966–9977, http://dx.doi.org/10.1021/jf061478a.

new insights, Curr. Med. Chem. 14 (2007) 2581–2589, http://dx.doi.org/10. [67] J. Robak, R.J. Gryglewski, Flavonoids are scavengers of superoxide anions,

2174/092986707782023325. Biochem. Pharmacol. 37 (1988) 837–841, http://dx.doi.org/10.1016/0006-

[43] H. Marini, L. Minutoli, F. Polito, A. Bitto, D. Altavilla, M. Atteritano, et al., 2952(88)90169-4.

Effects of the phytoestrogen genistein on bone metabolism in osteopenic [68] P. Nirmala, M. Ramanathan, Effect of myricetin on 1,2 dimethylhydrazine

postmenopausal women: a randomized trial, Ann. Intern. Med. 146 (2007) induced rat colon carcinogenesis, J. Exp. Ther. Oncol. 9 (2011) 101–108,

839–847, http://dx.doi.org/10.1210/jc.2006-2295. http://dx.doi.org/10.1016/j.ejphar.2010.11.034.

[44] A. Ørgaard, L. Jensen, The effects of soy isoflavones on obesity, Exp. Biol. [69] F.C. Meotti, R. Senthilmohan, D.T. Harwood, F.C. Missau, M.G. Pizzolatti, A.J.

Med. (Maywood, N.J.) 233 (2008) 1066–1080, http://dx.doi.org/10.3181/ Kettle, Myricitrin as a substrate and inhibitor of myeloperoxidase:

0712-MR-347. implications for the pharmacological effects of flavonoids, Free Radic. Biol.

[45] F.H. Lo, N.K. Mak, K.N. Leung, Studies on the anti-tumor activities of the soy Med. 44 (2008) 109–120, http://dx.doi.org/10.1016/j.freeradbiomed.2007.

isoflavone daidzein on murine neuroblastoma cells, Biomed. Pharmacother. 09.017.

61 (2007) 591–595, http://dx.doi.org/10.1016/j.biopha.2007.08.021. [70] H. Hibasami, A. Mitani, H. Katsuzaki, K. Imai, K. Yoshioka, T. Komiya,

[46] M. Yamaguchi, E. Sugimoto, Stimulatory effect of genistein and daidzein on Isolation of five types of flavonol from seabuckthorn (Hippophae

protein synthesis in osteoblastic MC3T3-E1 cells: activation of rhamnoides) and induction of apoptosis by some of the flavonols in human

aminoacyl-tRNA synthetase, Mol. Cell. Biochem. 214 (2000) 97–102, http:// promyelotic leukemia HL-60 cells, Int. J. Mol. Med. 15 (2005) 805–809,

dx.doi.org/10.1023/A.1007199120295. http://dx.doi.org/10.3892/ijmm.15.5.805.

[47] E. Sugimoto, M. Yamaguchi, Stimulatory effect of daidzein in osteoblastic [71] K.C. Ong, H.E. Khoo, Biological effects of myricetin, Gen. Pharmacol. 29

MC3T3-E1 cells, Biochem. Pharmacol. 59 (2000) 471–475, http://dx.doi.org/ (1997) 121–126, http://dx.doi.org/10.1016/S0306-3623(96)00421-1.

10.1016/S0006-2952(99)00351-2. [72] P. Rajendran, T. Rengarajan, N. Nandakumar, R. Palaniswami, Y. Nishigaki, I.

[48] M. Messina, S. Barnes, The role of soy products in reducing risk of cancer, J. Nishigaki, Kaempferol, a potential cytostatic and cure for inflammatory

Natl. Cancer Inst. 83 (1991) 541–546, http://dx.doi.org/10.1093/jnci/83.8. disorders, Eur. J. Med. Chem. 86 (2014) 103–112, http://dx.doi.org/10.1016/

541. j.ejmech.2014.08.011.

[49] R. Civitelli, In vitro and in vivo effects of ipriflavone on bone formation and [73] H.W.C. Leung, C.J. Lin, M.J. Hour, W.H. Yang, M.Y. Wang, H.Z. Lee, Kaempferol

bone biomechanics, Calcif. Tissue Int. 61 (Suppl. 1) (1997) S12–S14. induces apoptosis in human lung non-small carcinoma cells accompanied

[50] T. Cornwell, W. Cohick, I. Raskin, Dietary phytoestrogens and health, by an induction of antioxidant enzymes, Food Chem. Toxicol. 45 (2007)

Phytochemistry 65 (2004) 995–1016, http://dx.doi.org/10.1016/j. 2005–2013, http://dx.doi.org/10.1016/j.fct.2007.04.023.

phytochem.2004.03.005. [74] C.S. Bestwick, L. Milne, S.J. Duthie, Kaempferol induced inhibition of HL-60

[51] J.B. Harborne, C.A. Williams, Advances in flavonoid research since 1992, cell growth results from a heterogeneous response, dominated by cell cycle

Phytochemistry 55 (2000) 481–504, http://dx.doi.org/10.1016/S0031- alterations, Chem. Biol. Interact. 170 (2007) 76–85, http://dx.doi.org/10.

9422(00)00235-1. 1016/j.cbi.2007.07.002.

[52] C.B. Mantilla, L.G. Ermilov, The novel TrkB receptor agonist [75] R. Fang, P.J. Houghton, P.J. Hylands, Cytotoxic effects of compounds from Iris

7,8-dihydroxyflavone enhances neuromuscular transmission, Muscle Nerve tectorum on human cancer cell lines, J. Ethnopharmacol. 118 (2008)

45 (2012) 274–276, http://dx.doi.org/10.1002/mus.22295. 257–263, http://dx.doi.org/10.1016/j.jep.2008.04.006.

[53] J. Williams, 7,8-Dihydroxyflavone, A Selective Tyrosine Kinase Receptor B [76] A.J. Larson, J.D. Symons, T. Jalili, Therapeutic potential of quercetin to

Agonist and BDNF Mimic, Promotes Angiogenesis, East Tennessee State decrease blood pressure: review of efficacy and mechanisms, Adv. Nutr.

University, 2011, http://dc.etsu.edu/cgi/viewcontent. (Bethesda, Md.) 3 (2012) 39–46, http://dx.doi.org/10.3945/an.111.001271.

cgi?article=1027&context=honors (accessed 20.06.15).

114 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

[77] J. Pan, C. Yuan, C. Lin, Z. Jia, R. Zheng, Pharmacological activities and [102] J.H. Cho, Y. Lim, J.H. Ahn, S. Rhee, Crystallization and preliminary X-ray

mechanisms of natural phenylpropanoid glycosides, DiePharmazie 58 diffraction analysis of BcOMT2 from Bacillus cereus: a family of

(2003) 767–775, http://dx.doi.org/10.1111/j.1365-313X.2006.02872.x. O-methyltransferase, J. Microbiol. Biotechnol. 17 (2007) 369–372, http://dx.

[78] E.P. Cherniack, A berry thought-provoking idea: the potential role of plant doi.org/10.1016/j.jmb.2008.07.080.

polyphenols in the treatment of age-related cognitive disorders, Br. J. Nutr. [103] J.H. Cho, Y. Park, J.H. Ahn, Y. Lim, S. Rhee, Structural and functional insights

108 (2012) 794–800, http://dx.doi.org/10.1017/S0007114512000669. into O-methyltransferase from Bacillus cereus, J. Mol. Biol. 382 (2008)

[79] M. Ozipek, I. Calis¸ , M. Ertan, P. Rüedi, Rhamnetin 3-p-coumaroyl 987–997, http://dx.doi.org/10.1016/j.jmb.2008.07.080.

rhamninoside from Rhamnus petiolaris, Phytochemistry 37 (1994) 249–253, [104] W. Zhou, B. Feng, H. Huang, Y. Qin, Y. Wang, L. Kang, et al., Enzymatic

http://dx.doi.org/10.1016/0031-9422(94)85035-6. synthesis of alpha-glucosyl-timosaponin BII catalyzed by the extremely

[80] N.M.A. Chaudhry, P. Tariq, Bactericidal activity of black pepper, bay leaf, thermophilic enzyme: Toruzyme 3.0 L, Carbohydr. Res. 345 (2010)

aniseed and coriander against oral isolates, Pak. J. Pharm. Sci. 19 (2006) 1752–1759, http://dx.doi.org/10.1016/j.carres.2010.05.027.

214–218, http://dx.doi.org/10.13040/IJPSR.0975-8232. [105] B.S. Hong, B.G. Kim, N.R. Jung, Y. Lee, Y. Lim, Y. Chong, et al., Structural

[81] M.E. Szabo, E. Gallyas, I. Bak, A. Rakotovao, F. Boucher, J. de Leiris, et al., modeling and biochemical characterization of flavonoid

Heme oxygenase-1-related carbon monoxide and flavonoids in O-methyltransferase from Rice, Bull. Korean Chem. Soc. 30 (2009)

ischemic/reperfused rat retina, Invest. Ophthalmol. Vis. Sci. 45 (2004) 2803–2805, http://dx.doi.org/10.5012/bkcs.2009.30.11.2803.

3727–3732, http://dx.doi.org/10.1167/iovs.03-1324. [106] D. Chatterjee, D. Kudlinzki, V. Linhard, K. Saxena, U. Schieborr, S.L. Gande,

[82] H.N. Jnawali, E. Lee, K.W. Jeong, A. Shin, Y.S. Heo, Y. Kim, Anti-inflammatory et al., Structure and Biophysical Characterization of the

activity of rhamnetin and a model of its binding to c-Jun NH2-terminal S-adenosylmethionine-dependent O-methyltransferase PaMTH1, a putative

kinase 1 and p38 MAPK, J. Nat. Prod. 77 (2014) 258–263, http://dx.doi.org/ enzyme accumulating during senescence of Podospora anserina, J. Biol.

10.1021/np400803n. Chem. 290 (2015) 16415–16430, http://dx.doi.org/10.1074/jbc.M115.

[83] Y.J. Kim, Rhamnetin attenuates melanogenesis by suppressing oxidative 660829.

stress and pro-inflammatory mediators, Biol. Pharm. Bull. 36 (2013) [107] C. Zubieta, X.Z. He, R.A. Dixon, J.P. Noel, Structures of two natural product

1341–1347, http://dx.doi.org/10.1016/j.fitote.2014.06.012. methyltransferases reveal the basis for substrate specificity in plant

[84] Y.L. Liu, D.K. Ho, J.M. Cassady, V.M. Cook, W.M. Baird, Isolation of potential O-methyltransferases, Nat. Struct. Biol. 8 (2001) 271–279, http://dx.doi.org/

cancer chemopreventive agents from Eriodictyon californicum, J. Nat. Prod. 10.1038/85029.

55 (1992) 357–363, http://dx.doi.org/10.1021/np50081a012. [108] X. Hou, Y. Wang, Z. Zhou, S. Bao, Y. Lin, W. Gong, Crystal structure of

[85] R. Rakwal, G.K. Agrawal, M. Yonekura, O. Kodama, Naringenin SAM-dependent O-methyltransferase from pathogenic bacterium Leptospira

7-O-methyltransferase involved in the biosynthesis of the flavanone interrogans, J. Struct. Biol. 159 (2007) 523–528, http://dx.doi.org/10.1016/j.

phytoalexin sakuranetin from rice (Oryza sativa L.), Plant Sci. 155 (2000) jsb.2007.04.007.

213–221, http://dx.doi.org/10.1016/S0168-9452(00)00223-5. [109] J.A. Kornblatt, J.M. Zhou, R.K. Ibrahim, Structure-activity relationships of

[86] P. Tuchinda, V. Reutrakul, P. Claeson, U. Pongprayoon, T. Sematong, T. wheat flavone O-methyltransferase—a homodimer of convenience, FEBS J.

Santisuk, et al., Anti-inflammatory cyclohexenyl chalcone derivatives in 275 (2008) 2255–2266, http://dx.doi.org/10.1111/j.1742-4658.2008.06377.

Boesenbergia pandurata, Phytochemistry 59 (2002) 169–173, http://dx.doi. x.

org/10.1016/S0031-9422(01)00451-4. [110] C. Zubieta, P. Kota, J. Ferrer, R.A. a Dixon, J.P. Noel, Structural basis for the

[87] J. Orjala, A.D. Wright, H. Behrends, G. Folkers, O. Sticher, H. Rüegger, et al., modulation of lignin monomer methylation by caffeic acid/5-hydroxyferulic

Cytotoxic and antibacterial dihydrochalcones from Piper aduncum, J. Nat. acid 3/5-O-methyltransferase, Plant Cell 14 (2002) 1265–1277, http://dx.

Prod. 57 (1994) 18–26, http://dx.doi.org/10.1021/np50103a003. doi.org/10.1105/tpc.001412.nylpropanoids.

[88] X. Zhang, T.M. Hung, P.T. Phuong, T.M. Ngoc, B.S. Min, K.S. Song, et al., [111] J.E. Poulton, K. Hahlbrock, H. Grisebach, O-Methylation of flavonoid

Anti-inflammatory activity of flavonoids from Populus davidiana, Arch. substrates by a partially purified enzyme from soybean cell suspension

Pharm. Res. 29 (2006) 1102–1108, http://dx.doi.org/10.1007/BF02969299. cultures, Arch. Biochem. Biophys. 180 (1977) 543–549, http://dx.doi.org/10.

[89] S. Tamogami, O. Kodama, Coronatine elicits phytoalexin production in rice 1016/0003-9861(77)90071-6.

leaves (Oryza sativa L.) in the same manner as jasmonic acid, Phytochemistry [112] M.-H. Cho, H.L. Park, J.-H. Park, S.-W. Lee, S.H. Bhoo, T.-R. Hahn,

 

54 (2000) 689–694, http://dx.doi.org/10.1016/S0031-9422(00)00190-4. Characterization of regiospecific flavonoid 3 /5 -O-methyltransferase from

[90] A. Lungkaphin, A. Pongchaidecha, S. Palee, P. Arjinajarn, W. Pompimon, N. tomato and its application in flavonoid biotransformation, J. Korean Soc.

Chattipakorn, Pinocembrin reduces cardiac arrhythmia and infarct size in Appl. Biol. Chem. 55 (2012) 749–755, http://dx.doi.org/10.1007/s13765-

rats subjected to acute myocardial ischemia/reperfusion, Appl. Physiol. Nutr. 012-2193-3.

Metab. 40 (2015) 1031–1037, http://dx.doi.org/10.1139/apnm-2015-0108. [113] A. Gauthier, P.J. Gulick, R.K. Ibrahim, Characterization of two cDNA clones

[91] L. Peng, S. Yang, Y.J. Cheng, F. Chen, S. Pan, G. Fan, Antifungal activity and which encode O-methyltransferases for the methylation of both flavonoid

action mode of pinocembrin from propolis against Penicillium italicum, Food and phenylpropanoid compounds, Arch. Biochem. Biophys. 351 (1998)

Sci. Biotechnol. 21 (2012) 1533–1539, http://dx.doi.org/10.1007/s10068- 243–249, http://dx.doi.org/10.1006/abbi.1997.0554.

012-0204-0. [114] L.W. Wulf, C.W. Nagel, Identification and changes of flavonoids in merlot

[92] R.J. Weston, K.R. Mitchell, K.L. Allen, Antibacterial phenolic components of and carbernet sauvignon wines, J. Food Sci. 45 (1980) 479–484, http://dx.

New Zealand manuka honey, Food Chem. 64 (1999) 295–301, http://dx.doi. doi.org/10.1111/j.1365-2621.1980.tb04080.x.

org/10.1016/S0308-8146(98)00100-9. [115] S. Sabatier, M.J. Amiot, M. Tacchini, S. Aubert, Identification of flavonoids in

[93] M.A. Saad, R.M. Abdel Salam, S.A. Kenawy, A.S. Attia, Pinocembrin attenuates sunflower Honey, J. Food Sci. 57 (1992) 773–774, http://dx.doi.org/10.1111/

hippocampal inflammation, oxidative perturbations and apoptosis in a rat j.1365-2621.1992.tb08094.x.

model of global cerebral ischemia reperfusion, Pharmacol. Rep.: PR 67 [116] M. Bimakr, R.A. Rahman, F.S. Taip, A. Ganjloo, L.M. Salleh, J. Selamat, et al.,

(2015) 115–122, http://dx.doi.org/10.1016/j.pharep.2014.08.014. Comparison of different extraction methods for the extraction of major

[94] N. Nasr Bouzaiene, F. Chaabane, A. Sassi, L. Chekir-Ghedira, K. Ghedira, Effect bioactive flavonoid compounds from spearmint (Mentha spicata L.) leaves,

of apigenin-7-glucoside, genkwanin and naringenin on tyrosinase activity Food Bioprod. Process. 89 (2011) 67–72, http://dx.doi.org/10.1016/j.fbp.

and melanin synthesis in B16F10 melanoma cells, Life Sci. 144 (2016) 2010.03.002.

80–85, http://dx.doi.org/10.1016/j.lfs.2015.11.030. [117] V. Kren,ˇ J. Kubisch, P. Sedmera, P. Halada, V. Prikrylová,ˇ A. Jegorov, et al.,

[95] R.K. Ibrahim, A. Bruneau, B. Bantignies, Plant O-methyltransferases: Glycosylation of silybin, J. Chem. Soc. Perkin Trans. 1 (1997) 2467–2474,

molecular analysis, common signature and classification, Plant Mol. Biol. 36 http://dx.doi.org/10.1039/a703283h.

(1998) 1–10, http://dx.doi.org/10.1023/A.1005939803300. [118] N.H. Thuan, J.W. Park, J.K. Sohng, Toward the production of

[96] J.P. Noel, R.A. Dixon, E. Pichersky, C. Zubieta, J.L. Ferrer, Integrative flavone-7-O-␤-d-glucopyranosides using Arabidopsis glycosyltransferase in

phytochemistry: from ethnobotany to molecular ecology, Pergamon, U.S.A., Escherichia coli, Process Biochem. 48 (2013) 1744–1748, http://dx.doi.org/

2003. 10.1016/j.procbio.2013.07.005.

[97] J.G. Kopycki, D. Rauh, A.A. Chumanevich, P. Neumann, T. Vogt, M.T. Stubbs, [119] M. Hosny, J.P.N. Rosazza, Microbial hydroxylation and methylation of

Biochemical and structural analysis of substrate promiscuity in plant genistein by Streptomycetes, J. Nat. Prod. 62 (1999) 1609–1612, http://dx.doi.

2+

Mg -dependent O-methyltransferases, J. Mol. Biol. 378 (2008) 154–164, org/10.1021/np9901783.

http://dx.doi.org/10.1016/j.jmb.2008.02.019. [120] S. Lee, S.Y. Shin, Y. Lee, Y. Park, B.G. Kim, J.H. Ahn, et al., Rhamnetin

[98] J.C. Qin, Y.M. Zhang, C.Y. Lang, Y.H. Yao, H.Y. Pan, X. Li, Cloning and production based on the rational design of the poplar O-methyltransferase

functional characterization of a caffeic acid O-methyltransferase from enzyme and its biological activities, Bioorg. Med. Chem. Lett. 21 (2011)

Trigonella foenumgraecum L, Mol. Biol. Rep. 39 (2012) 1601–1608, http://dx. 3866–3870, http://dx.doi.org/10.1016/j.bmcl.2011.05.043.

doi.org/10.1007/s11033-011-0899-7. [121] B.G. Kim, B.R. Jung, Y. Lee, H.G. Hur, Y. Lim, J.H. Ahn, Regiospecific flavonoid

[99] H.L. Schubert, R.M. Blumenthal, X. Cheng, Many paths to methyltransfer: a 7-O-methylation with Streptomyces avermitilis O-methyltransferase

chronicle of convergence, Trends Biochem. Sci. 28 (2003) 329–335, http:// expressed in Escherichia coli, J. Agric. Food Chem. 54 (2006) 823–828, http://

dx.doi.org/10.1016/S0968-0004(03)00090-2. dx.doi.org/10.1021/jf0522715.

[100] A. Schmidt, C. Li, A.D. Jones, E. Pichersky, Characterization of a flavonol [122] S. Malla, M.A.G. Koffas, R.J. Kazlauskas, B.G. Kim, Production of 7-O-methyl

3-O-methyltransferase in the trichomes of the wild tomato species Solanum aromadendrin, a medicinally valuable flavonoid, in Escherichia coli, Appl.

habrochaites, Planta 236 (2012) 839–849, http://dx.doi.org/10.1007/s00425- Environ. Microbiol. 78 (2012) 684–694, http://dx.doi.org/10.1128/AEM.

012-1676-0. 06274-11.

[101] E.J. Joe, B.G. Kim, B.C. An, Y. Chong, J.H. Ahn, Engineering of flavonoid [123] M.J. Kim, B.G. Kim, J.H. Ahn, Biosynthesis of bioactive O-methylated

O-methyltransferase for a novel regioselectivity, Mol. Cells 30 (2010) flavonoids in Escherichia coli, Appl. Microbiol. Biotechnol. 97 (2013)

137–141, http://dx.doi.org/10.1007/s10059-010-0098-8. 7195–7204, http://dx.doi.org/10.1007/s00253-013-5020-9.

N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116 115

[124] I. Miyahisa, M. Kaneko, N. Funa, H. Kawasaki, H. Kojima, Y. Ohnishi, et al., [148] P. Zhao, L. Bai, J. Ma, Y. Zeng, L. Li, Y. Zhang, et al., Amide N-glycosylation by

Efficient production of (2S)-flavanones by Escherichia coli containing an Asm25, an N-glycosyltransferase of ansamitocins, Chem. Biol. 15 (2008)

artificial biosynthetic gene cluster, Appl. Microbiol. Biotechnol. 68 (2005) 863–874, http://dx.doi.org/10.1016/j.chembiol.2008.06.007.

498–504, http://dx.doi.org/10.1007/s00253-005-1916-3. [149] G.R. Li, H.B. Wang, G.W. Qin, M.W. Jin, Q. Tang, H.Y. Sun, et al., Acacetin, a

[125] E. Leonard, Y. Yan, Z.L. Fowler, Z. Li, C.G. Lim, K.H. Lim, et al., Strain natural flavone, selectively inhibits human atrial repolarization potassium

improvement of recombinant Escherichia coli for efficient production of currents and prevents atrial fibrillation in dogs, Circulation 117 (2008)

plant flavonoids, Mol. Pharm. 5 (2016) 257–265, http://dx.doi.org/10.1021/ 2449–2457, http://dx.doi.org/10.1161/CIRCULATIONAHA.108.769554.

mp7001472. [150] G.A. Akowuah, Z. Ismail, I. Norhayati, A. Sadikun, The effects of different

[126] J. Wu, G. Du, J. Zhou, J. Chen, Metabolic engineering of Escherichia coli for extraction solvents of varying polarities on polyphenols of Orthosiphon

(2S)-pinocembrin production from glucose by a modular metabolic strategy, stamineus and evaluation of the free radical-scavenging activity, Food Chem.

Metab. Eng. 16 (2013) 48–55, http://dx.doi.org/10.1016/j.ymben.2012.11. 93 (2005) 311–317, http://dx.doi.org/10.1016/j.foodchem.2004.09.028.

009. [151] K. Steinke, E. Jose, D. Sicker, H.U. Siehl, K.P. Zeller, S. Berger, Sinensetin,

[127] B.G. Kim, H. Lee, J.H. Ahn, Biosynthesis of pinocembrin from glucose using Chem. Unserer Zeit 47 (2013) 158–163, http://dx.doi.org/10.1002/ciuz.

engineered Escherichia coli, J. Microbiol. Biotechnol. 24 (2014) 1536–1541, 201300627.

http://dx.doi.org/10.4014/jmb.1406.06011. [152] X.F. Zhu, B.F. Xie, J.M. Zhou, G.K. Feng, Z.C. Liu, X.Y. Wei, et al., Blockade of

[128] P. Tuchinda, V. Reutrakul, P. Claeson, U. Pongprayoon, T. Sematong, T. vascular endothelial growth factor receptor signal pathway and antitumor

    

Santisuk, et al., Anti-inflammatory cyclohexenyl chalcone derivatives in activity of ON-III (2 ,4 -dihydroxy-6 -methoxy-3 ,5 -dimethylchalcone), a

Boesenbergia pandurata, Phytochemistry 59 (2002) 169–173, http://dx.doi. component from Chinese herbal medicine, Mol. Pharmacol. 67 (2005)

org/10.1016/S0031-9422(01)00451-4. 1444–1450, http://dx.doi.org/10.1124/mol.104.009894.

[129] X. Zhang, T.M. Hung, P.T. Phuong, T.M. Ngoc, B.S. Min, K.S. Song, et al., [153] F.R. Pavan, C.Q.F. Leite, R.G. Coelho, I.D. Coutinho, N.K. Honda, C.A.L. Cardoso,

Anti-inflammatory activity of flavonoids from Populus davidiana, Arch. et al., Evaluation of anti-Mycobacterium tuberculosis activity of

Pharm. Res. 29 (2006) 1102–1108, http://dx.doi.org/10.1007/BF02969299. Campomanesia adamantium (Myrtaceae), Química Nova 32 (2009)

[130] Y. Ogawa, H. Oku, E. Iwaoka, M. Iinuma, K. Ishiguro, Allergy-preventive 1222–1226, http://dx.doi.org/10.1590/S0100-40422009000500026.

flavonoids from Xanthorrhoea hastilis, Chem. Pharm. Bull. 55 (2007) [154] L. Shi, X.E. Feng, J.R. Cui, L.H. Fang, G.H. Du, Q.S. Li, Synthesis and biological

675–678, http://dx.doi.org/10.1248/cpb.55.675. activity of flavanone derivatives, Bioorg. Med. Chem. Lett. 20 (2010)

[131] F.V. So, N. Guthrie, A.F. Chambers, M. Moussa, K.K. Carroll, Inhibition of 5466–5468, http://dx.doi.org/10.1016/j.bmcl.2010.07.090.

human breast cancer cell proliferation and delay of mammary [155] J. Zhao, H.X. Ding, D.G. Zhao, C.M. Wang, K. Gao, Isolation, modification and

tumorigenesis by flavonoids and citrus juices, Nutr. Cancer 26 (1996) cytotoxic evaluation of flavonoids from Rhododendron hainanense, J. Pharm.

167–181, http://dx.doi.org/10.1080/01635589609514473. Pharmacol. 64 (2012) 1785–1792, http://dx.doi.org/10.1111/j.2042-7158.

[132] B. Zarebczan, S.N. Pinchot, M. Kunnimalaiyaan, H. Chen, Hesperetin, a 2012.01560.x.

potential therapy for carcinoid cancer, Am. J. Surg. 201 (2011) 329–332, [156] J.B. Harborne, Comparative biochemistry of the flavonoids-IV,

http://dx.doi.org/10.1016/j.amjsurg.2010.08.018 (discussion 333). Phytochemistry 6 (1967) 1415–1428, http://dx.doi.org/10.1016/S0031-

[133] D. Zhang, L. Xie, G. Jia, S. Cai, B. Ji, Y. Liu, et al., Comparative study on 9422(00)82884-8.

antioxidant capacity of flavonoids and their inhibitory effects on oleic [157] A. Piovan, R. Filippini, D. Favretto, Characterization of the anthocyanins of

acid-induced hepatic steatosis in vitro, Eur. J. Med. Chem. 46 (2011) Catharanthus roseus (L.) G. Donin vivo andin vitro by electrospray ionization

4548–4588, http://dx.doi.org/10.1016/j.ejmech.2011.07.031. ion trap mass spectrometry, Rapid Commun. Mass Spectrom. 12 (1998)

[134] B. Bauvois, M.L. Puiffe, J.B. Bongui, S. Paillat, C. Monneret, D. Dauzonne, 361–367, http://dx.doi.org/10.1002/(SICI)1097-0231(19980415)12:73.0.

Synthesis and biological evaluation of novel flavone-8-acetic acid CO;2-U.

derivatives as reversible inhibitors of aminopeptidase N/CD13, J. Med. [158] S.M. Cerritelli, R.J. Crouch, Cloning, expression, and mapping of

Chem. 46 (2003) 3900–3913, http://dx.doi.org/10.1021/jm021109f. ribonucleases H of human and mouse related to bacterial RNase HI,

[135] A. Nagao, M. Seki, H. Kobayashi, Inhibition of xanthine oxidase by Genomics 53 (1998) 300–307, http://dx.doi.org/10.1006/geno.1998.5497.

flavonoids, Biosci. Biotechnol. Biochem. 63 (1999) 1787–1790, http://dx.doi. [159] K. Toki, N. Saito, Y. Irie, F. Tatsuzawa, A. Shigihara, T. Honda, 7-O-Methylated

org/10.1080/00021369.1985.10867027. anthocyanidin glycosides from Catharanthus roseus, Phytochemistry 69



[136] S. Sacco, M. Maffei, The effect of isosakuranetin (5,7-dihydroxy 4 -methoxy (2008) 1215–1219, http://dx.doi.org/10.1016/j.phytochem.2007.11.005.

flavanone) on potassium uptake in wheat root segments, Phytochemistry 46 [160] T. Iwashina, The structure and distribution of the flavonoids in plants, J.

(1997) 245–248, http://dx.doi.org/10.1016/S0031-9422(97)00289-6. Plant Res. 113 (2000) 287–299, http://dx.doi.org/10.1007/PL00013940.

[137] H. Takemura, T. Itoh, K. Yamamoto, H. Sakakibara, K. Shimoi, Selective [161] O.A. Andreeva, M.N. Ivashev, I.I. Ozimina, G.V. Maslikova, Diosmetin

inhibition of methoxyflavonoids on human CYP1B1 activity, Bioorg. Med. glycosides from caucasian vetch: isolation and study of biological activity,

Chem. 18 (2010) 6310–6315, http://dx.doi.org/10.1016/j.bmc.2010.07.020. Pharm. Chem. J. 32 (1998) 595–597, http://dx.doi.org/10.1007/BF02465832.

[138] K.E. Malterud, T.E. Bremnes, A. Faegri, T. Moe, E.K.S. Dugstad, T. Anthonsen, [162] G. Brunhofer, A. Fallarero, D. Karlsson, A. Batista-Gonzalez, P. Shinde, C. Gopi

et al., Flavonoids from the wood of Salix caprea as inhibitors of Mohan, et al., Exploration of natural compounds as sources of new

wood-destroying fungi, J. Nat. Prod. 48 (1985) 559–563, http://dx.doi.org/ bifunctional scaffolds targeting cholinesterases and beta amyloid

10.1021/np50040a007. aggregation: the case of chelerythrine, Bioorg. Med. Chem. 20 (2012)

[139] H. Maruyama, Y. Sumitou, T. Sakamoto, Y. Araki, H. Hara, Antihypertensive 6669–6679, http://dx.doi.org/10.1016/j.bmc.2012.09.040.

effects of flavonoids isolated from brazilian green propolis in spontaneously [163] C. Scholl, S. Fröhling, I.F. Dunn, A.C. Schinzel, D.A. Barbie, S.Y. Kim, et al.,

hypertensive rats, Biol. Pharm. Bull. 32 (2009) 1244–1250, http://dx.doi.org/ Synthetic lethal interaction between oncogenic KRAS dependency and

10.1248/bpb.32.1244. STK33 suppression in human cancer cells, Cell 137 (2009) 821–834, http://

[140] N. Pandurangan, C. Bose, A. Banerji, Synthesis and antioxygenic activities of dx.doi.org/10.1016/j.cell.2009.03.017.

seabuckthorn flavone-3-ols and analogs, Bioorg. Med. Chem. Lett. 21 (2011) [164] H. Cai, M. Al-Fayez, R.G. Tunstall, S. Platton, P. Greaves, W.P. Steward, et al.,

5328–5330, http://dx.doi.org/10.1016/j.bmcl.2011.07.008. The rice bran constituent tricin potently inhibits cyclooxygenase enzymes

[141] H. Matsuda, S. Nakashima, Y. Oda, S. Nakamura, M. Yoshikawa, and interferes with intestinal carcinogenesis in ApcMin mice, Mol. Cancer

Melanogenesis inhibitors from the rhizomes of Alpinia officinarum in B16 Ther. 4 (2005) 1287–1292, http://dx.doi.org/10.1158/1535-7163.MCT-05-

melanoma cells, Bioorg. Med. Chem. 17 (2009) 6048–6053, http://dx.doi. 0165.

org/10.1016/j.bmc.2009.06.057. [165] M. Maver, E.F. Queiroz, J.L. Wolfender, K. Hostettmann, Flavonoids from the

[142] A. Lale, J.M. Herbert, J.M. Augereau, M. Billon, M. Leconte, J. Gleye, Ability of stem of Eriophorum scheuchzeri, J. Nat. Prod. 68 (2005) 1094–1098, http://dx.

different flavonoids to inhibit the procoagulant activity of adherent human doi.org/10.1021/np0580107.

monocytes, J. Nat. Prod. 59 (1996) 273–276, http://dx.doi.org/10.1021/ [166] H. Kuwabara, K. Mouri, H. Otsuka, R. Kasai, K. Yamasaki, Tricin from a

np960057s. malagasy connaraceous plant with potent antihistaminic activity, J. Nat.

[143] M.M. Liu, L. Zhou, P.L. He, Y.N. Zhang, J.Y. Zhou, Q. Shen, et al., Discovery of Prod. 66 (2003) 1273–1275, http://dx.doi.org/10.1021/np030020p.

flavonoid derivatives as anti-HCV agents via pharmacophore search [167] E.M. Kurowska, J.A. Manthey, Hypolipidemic effects and absorption of citrus

combining molecular docking strategy, Eur. J. Med. Chem. 52 (2012) 33–43, polymethoxylated flavones in hamsters with diet-induced

http://dx.doi.org/10.1016/j.ejmech.2012.03.002. hypercholesterolemia, J. Agric. Food Chem. 52 (2004) 2879–2886, http://dx.

[144] Y.L. Hsu, H.L. Liang, C.H. Hung, P.L. Kuo, Syringetin, a flavonoid derivative in doi.org/10.1021/jf035354z.

grape and wine, induces human osteoblast differentiation through bone [168] K.P. Datla, M. Christidou, W.W. Widmer, H.K. Rooprai, D.T. Dexter, Tissue

morphogenetic protein-2/extracellular signal-regulated kinase 1/2 distribution and neuroprotective effects of citrus flavonoid tangeretin in a

pathway, Mol. Nutr. Food Res. 53 (2009) 1452–1461, http://dx.doi.org/10. rat model of Parkinson’s disease, NeuroReport 12 (2001) 3871–3875, http://

1002/mnfr.200800483. dx.doi.org/10.1016/j.yexcr.2010.01.001.

[145] J. Guo, D.L. Yu, L. Xu, M. Zhu, S.L. Yang, Flavonol glycosides from Lysimachia [169] J. Gao, W.A. Morgan, A. Sanchez-Medina, O. Corcoran, The ethanol extract of

congestiflora, Phytochemistry 48 (1998) 1445–1447, http://dx.doi.org/10. Scutellaria baicalensis and the active compounds induce cell cycle arrest and

1016/S0031-9422(97)01025-X. apoptosis including upregulation of p53 and Bax in human lung cancer cells,

[146] F. Mattivi, R. Guzzon, U. Vrhovsek, M. Stefanini, R. Velasco, Metabolite Toxicol. Appl. Pharmacol. 254 (2011) 221–228, http://dx.doi.org/10.1016/j.

profiling of grape: flavonols and anthocyanins, J. Agric. Food Chem. 54 taap.2011.03.016.

(2006) 7692–7702, http://dx.doi.org/10.1021/jf061538c. [170] H.G. Park, S.Y. Yoon, J.Y. Choi, G.S. Lee, J.H. Choi, C.Y. Shin, et al.,

[147] A.K. Lätti, L. Jaakola, K.R. Riihinen, P.S. Kainulainen, Anthocyanin and Anticonvulsant effect of wogonin isolated from Scutellaria baicalensis, Eur. J.

flavonol variation in bog bilberries (Vaccinium uliginosum L.) in Finland, J. Pharmacol. 574 (2007) 112–119, http://dx.doi.org/10.1016/j.ejphar.2007.07.

Agric. Food Chem. 58 (2010) 427–433, http://dx.doi.org/10.1021/jf903033m. 011.

116 N. Koirala et al. / Enzyme and Microbial Technology 86 (2016) 103–116

[171] G. Polier, J. Ding, B.V. Konkimalla, D. Eick, N. Ribeiro, R. Köhler, et al., [194] M.C. Wu, C.F. Peng, I.S. Chen, I.L. Tsai, Antitubercular chromones and

Wogonin and related natural flavones are inhibitors of CDK9 that induce flavonoids from Pisonia aculeata, J. Nat. Prod. 74 (2011) 976–982, http://dx.

apoptosis in cancer cells by transcriptional suppression of Mcl-1, Cell Death doi.org/10.1021/np1008575.

Dis. 2 (2011) e182, http://dx.doi.org/10.1038/cddis.2011.66. [195] L. Gregersen, A.B. Christensen, J. Sommer-Knudsen, D.B. Collinge, A putative

[172] A. Maiti, M. Cuendet, T. Kondratyuk, V.L. Croy, J.M. Pezzuto, M. Cushman, O-methyltransferase from barley is induced by fungal pathogens and UV

Synthesis and cancer chemopreventive activity of zapotin, a natural product light, Plant Mol. Biol. 26 (1994) 1797–1806, http://dx.doi.org/10.1186/1471-

from Casimiroa edulis, J. Med. Chem. 50 (2007) 350–355, http://dx.doi.org/ 2229-8-88.

10.1021/jm060915+. [196] A.B. Christensen, P.L. Gregersen, C.E. Olsen, D.B. Collinge, A flavonoid

[173] G. Murillo, W.H. Hirschelman, A. Ito, R.M. Moriarty, A.D. Kinghorn, J.M. 7-O-methyltransferase is expressed in barley leaves in response to pathogen

Pezzuto, et al., Zapotin, a phytochemical present in a Mexican fruit, prevents attack, Plant Mol. Biol. 36 (1998) 219–227, http://dx.doi.org/10.1023/A.

colon carcinogenesis, Nutr. Cancer 57 (2007) 28–37, http://dx.doi.org/10. 1005985609313.

1080/01635580701268097. [197] J.M. Zhou, Y. Fukushi, E. Wollenweber, R.K. Ibrahim, Characterization of two

[174] Z. Nikolovska-Coleska, R. Wang, X. Fang, H. Pan, Y. Tomita, P. Li, et al., O-methyltransferase-like genes in barley and maize, Pharm. Biol. 46 (2008)

Development and optimization of a binding assay for the XIAP BIR3 domain 26–34, http://dx.doi.org/10.1080/13880200701729745.

using fluorescence polarization, Anal. Biochem. 332 (2004) 261–273, http:// [198] J.M. Zhou, N.D. Gold, V.J.J. Martin, E. Wollenweber, R.K. Ibrahim, Sequential

dx.doi.org/10.1016/j.ab.2004.05.055. O-methylation of tricetin by a single gene product in wheat, Biochim.

[175] G.M. Barton, New C-methylflavanones from Douglas-fir, Phytochemistry 11 Biophys. Acta 1760 (2006) 1115–1124, http://dx.doi.org/10.1016/j.bbagen.

(1972) 426–429, http://dx.doi.org/10.1016/S0031-9422(00)90036-0. 2006.02.008.

[176] C.M. Starks, R.B. Williams, V.L. Norman, J.A. Lawrence, M.G. Goering, M. [201] G. Schröder, E. Wehinger, R. Lukacin, F. Wellmann, W. Seefelder, W. Schwab,



O’Neil-Johnson, et al., Abronione, a rotenoid from the desert annual Abronia et al., Flavonoid methylation: a novel 4 -O-methyltransferase from

villosa, Phytochem. Lett. 4 (2011) 72–74, http://dx.doi.org/10.1016/j.phytol. Catharanthus roseus, and evidence that partially methylated flavanones are

2010.08.004. substrates of four different flavonoid dioxygenases, Phytochemistry 65

[177] A. Wal, A. Rai, P. Wal, G. Sharma, Biological activities of lupeol, Syst. Rev. (2004) 1085–1094, http://dx.doi.org/10.1016/j.phytochem.2004.02.010.

Pharm. 2 (2011) 96, http://dx.doi.org/10.4103/0975-8453.86298. [202] S. Cacace, G. Schröder, E. Wehinger, D. Strack, J. Schmidt, J. Schröder, A

[178] S. Alakurtti, T. Mäkelä, S. Koskimies, J. Yli-Kauhaluoma, Pharmacological flavonol O-methyltransferase from Catharanthus roseus performing two

properties of the ubiquitous natural product betulin, Eur. J. Pharm. Sci. 29 sequential methylations, Phytochemistry 62 (2003) 127–137, http://dx.doi.

(2006) 1–13, http://dx.doi.org/10.1016/j.ejps.2006.04.006. org/10.1016/j.phytochem.2011.01.036.

[179] J.J. Tang, J.G. Li, W. Qi, W.W. Qiu, P.S. Li, B.L. Li, et al., Inhibition of SREBP by a [203] J.C. Motamayor, K. Mockaitis, J. Schmutz, N. Haiminen, D. Livingstone, O.

small molecule, betulin, improves hyperlipidemia and insulin resistance and Cornejo, et al., The genome sequence of the most widely cultivated cacao

reduces atherosclerotic plaques, Cell Metab. 13 (2011) 44–56, http://dx.doi. type and its use to identify candidate genes regulating pod color, Genome

org/10.1016/j.cmet.2010.12.004. Biol. 14 (2013) r53, http://dx.doi.org/10.1186/gb-2013-14-6-r53.

[180] T. Fujioka, Y. Kashiwada, R.E. Kilkuskie, L.M. Cosentino, L.M. Ballas, J.B. Jiang, [204] P.S. Schnable, D. Ware, R.S. Fulton, J.C. Stein, F. Wei, S. Pasternak, et al., The

et al., Anti-AIDS agent. 11. Betulinic acid and platanic acid as anti-HIV B73 maize genome: complexity, diversity, and dynamics, Science (New York,

principles from Syzigium claviflorum, and the anti-HIV activity of N.Y.) 326 (2009) 1112–1115, http://dx.doi.org/10.1126/science.1178534.

2+

structurally related triterpenoids, J. Nat. Prod. 57 (1994) 243–247, http://dx. [205] M. Ibdah, X.H. Zhang, J. Schmidt, T. Vogt, A novel Mg -dependent

doi.org/10.3109/10409238.2014.953628. O-methyltransferase in the phenylpropanoid metabolism of

[181] S. Gafner, J.L. Wolfender, S. Mavi, K. Hostettmann, Antifungal and Mesembryanthemum crystallinum, J. Biol. Chem. 278 (2003) 43961–43972,

antibacterial chalcones from Myrica serrata, Planta Med. 62 (1996) 67–69, http://dx.doi.org/10.1074/jbc.M304932200.

http://dx.doi.org/10.1055/s-2006-957804. [206] T. Akashi, H.D. VanEtten, Y. Sawada, C.C. Wasmann, H. Uchiyama, S. Ayabe,

[182] J.H. Wu, X.H. Wang, Y.H. Yi, K.H. Lee, Anti-AIDS agents 54. A potent anti-HIV Catalytic specificity of pea O-methyltransferases suggests gene duplication

chalcone and flavonoids from genus Desmos, Bioorg. Med. Chem. Lett. 13 for (+)-pisatin biosynthesis, Phytochemistry 67 (2006) 2525–2530, http://

(2003) 1813–1815, http://dx.doi.org/10.1016/S0960-894X(03)00197-5. dx.doi.org/10.1016/j.phytochem.2006.09.010.

[183] E.C. Amor, I.M. Villasenor,˜ A. Yasin, M.I. Choudhary, Prolyl endopeptidase [207] T. Akashi, Y. Sawada, N. Shimada, N. Sakurai, T. Aoki, S. Ayabe, cDNA cloning

inhibitors from Syzygium samarangense (Blume) Merr. & L.M. Perry, J. Biosci. and biochemical characterization of S-adenosyl-l-methionine:

 

(Zeitschrift Fnr˜ Naturforschung. C) 59 (2004) 86–92, http://dx.doi.org/10. 2,7,4 -trihydroxyisoflavanone 4 -O-methyltransferase, a critical enzyme of

1016/j.foodchem.2007.08.086. the legume isoflavonoid phytoalexin pathway, Plant Cell Physiol. 44 (2003)

[184] H. Wu, X. Liao, L. Mao, Q. Liang, L. Wang, W. Su, Desmoflavone II: a new 103–112, http://dx.doi.org/10.1093/pcp/pcg034.

flavanone from Desmo cochinchinensis Lour, J. Chin. Pharm. Sci. 6 (1997) 3–5, [208] C. Zhang, C. Albermann, X. Fu, N.R. Peters, J.D. Chisholm, G. Zhang, et al.,

http://dx.doi.org/10.1016/j.ejvs.2009.08.009. RebG- and RebM-catalyzed indolocarbazole diversification, ChemBioChem:

[185] J.H. Wu, A.T. McPhail, K.F. Bastow, H. Shiraki, J. Ito, K.H. Lee, Desmosdumotin Eur. J. Chem. Biol. 7 (2006) 795–804, http://dx.doi.org/10.1002/cbic.

C, a novel cytotoxic principle from Desmos dumosus, Tetrahedron Lett. 43 200500504.

(2002) 1391–1393, http://dx.doi.org/10.1016/S0040-4039(02)00026-6. [209] S. Singh, J.G. McCoy, C. Zhang, C.A. Bingman, G.N. Phillips, J.S. Thorson,

[186] H.M.A. Sidahmed, A.H.S. Azizan, S. Mohan, M.A. Abdulla, S.I. Abdelwahab, Structure and mechanism of the rebeccamycin sugar



M.M.E. Taha, et al., Gastroprotective effect of desmosdumotin C isolated 4 -O-methyltransferase RebM, J. Biol. Chem. 283 (2008) 22628–22636,

from Mitrella kentii against ethanol-induced gastric mucosal hemorrhage in http://dx.doi.org/10.1074/jbc.M800503200.

rats: possible involvement of glutathione, heat-shock protein-70, sulfhydryl [210] K. Zhang, M.W. Bhuiya, J.R. Pazo, Y. Miao, H. Kim, J. Ralph, et al., An

compounds, nitric oxide, and anti-Helicobacter pylori, BMC Complementa. engineered monolignol 4-O-methyltransferase depresses lignin

Altern. Med. 13 (2013) 183, http://dx.doi.org/10.1186/1472-6882-13-183. biosynthesis and confers novel metabolic capability in Arabidopsis, Plant Cell

[187] A.P.M. Bernardi, A.B.F. Ferraz, D.V. Albring, S.A.L. Bordignon, J. Schripsema, R. 24 (2012) 3135–3152, http://dx.doi.org/10.1105/tpc.112.101287.

Bridi, et al., Benzophenones from Hypericum carinatum, J. Nat. Prod. 68 [211] J.M. Zhou, Y.W. Seo, R.K. Ibrahim, Biochemical characterization of a putative

(2005) 784–786, http://dx.doi.org/10.1021/np040149e. wheat caffeic acid O-methyltransferase, Plant Physiol. Biochem. 47 (2009)

[188] A.V. Pinhatti, F.M.C. de Barros, C.B. de Farias, G. Schwartsmann, G.L. von 322–326, http://dx.doi.org/10.1016/j.plaphy.2008.11.011.



Poser, A.L. Abujamra, Antiproliferative activity of the dimeric phloroglucinol [212] B.G. Kim, Y. Lee, H.G. Hur, Y. Lim, J.H. Ahn, Flavonoid 3 -O-methyltransferase

and benzophenone derivatives of Hypericum spp. native to southern Brazil, from rice: cDNA cloning, characterization and functional expression,

Anti Cancer Drugs 24 (2013) 699–703, http://dx.doi.org/10.1097/CAD. Phytochemistry 67 (2006) 387–394, http://dx.doi.org/10.1016/j.phytochem.

0b013e3283626626. 2005.11.022.

[189] S.P. Soltoff, Rottlerin: an inappropriate and ineffective inhibitor of PKCdelta, [215] S. Omura, H. Ikeda, J. Ishikawa, A. Hanamoto, C. Takahashi, M. Shinose, et al.,

Trends Pharmacol. Sci. 28 (2007) 453–458, http://dx.doi.org/10.1016/j.tips. Genome sequence of an industrial microorganism Streptomyces avermitilis:

2007.07.003. deducing the ability of producing secondary metabolites, Proc. Natl. Acad.

[190] M. Gschwendt, H.J. Müller, K. Kielbassa, R. Zang, W. Kittstein, G. Rincke, Sci. U. S. A. 98 (2001) 12215–12220, http://dx.doi.org/10.1073/pnas.

et al., Rottlerin, a novel protein kinase inhibitor, Biochem. Biophys. Res. 211433198.

Commun. 199 (1994) 93–98, http://dx.doi.org/10.1006/bbrc.1994.1199. [216] A. Copeland, W. Gu, M. Yasawong, A. Lapidus, S. Lucas, S. Deshpande, et al.,

[191] R.G. Ptak, B.G. Gentry, T.L. Hartman, K.M. Watson, M.C. Osterling, R.W. Complete genome sequence of the aerobic, heterotroph Marinithermus

Buckheit, et al., Inhibition of human immunodeficiency virus type 1 by hydrothermalis type strain (T1(T)) from a deep-sea hydrothermal vent

triciribine involves the accessory protein nef, Antimicrob. Agents chimney, Stand. Genom. Sci. 6 (2012) 21–30, http://dx.doi.org/10.4056/sigs.

Chemother. 54 (2010) 1512–1519, http://dx.doi.org/10.1128/AAC.01443-09. 2435521.

[192] L.L. Pan, P.L. Fang, X.J. Zhang, W. Ni, L. Li, L.M. Yang, et al., Tigliane-type [217] J. Sikorski, B.J. Tindall, S. Lowry, S. Lucas, M. Nolan, A. Copeland, et al.,

diterpenoid glycosides from Euphorbia fischeriana, J. Nat. Prod. 74 (2011) Complete genome sequence of Meiothermus silvanus type strain (VI-R2),

1508–1512, http://dx.doi.org/10.1021/np200058c. Stand. Genom. Sci. 3 (2010) 37–46, http://dx.doi.org/10.4056/sigs.1042812.

[193] R.T. D’Aquila, V.A. Johnson, S.L. Welles, A.J. Japour, D.R. Kuritzkes, V. [218] H. Daligault, A. Lapidus, A. Zeytun, M. Nolan, S. Lucas, T.G. Del Rio, et al.,

DeGruttola, et al., Zidovudine resistance and HIV-1 disease progression Complete genome sequence of Haliscomenobacter hydrossis type strain (O),

during antiretroviral therapy. AIDS Clinical Trials Group Protocol 116B/117 Stand. Genom. Sci. 4 (2011) 352–360, http://dx.doi.org/10.4056/sigs.

Team and the Virology Committee Resistance Working Group, Ann. Intern. 1964579.

Med. 122 (1995) 401–408, http://dx.doi.org/10.7326/0003-4819-122-6- [219] O. Chertkov, J. Sikorski, M. Nolan, A. Lapidus, S. Lucas, T.G. Del Rio, et al.,

199503150-00001. Complete genome sequence of Thermomonospora curvata type strain (B9T),

Stand. Genom. Sci. 4 (2011) 13–22, http://dx.doi.org/10.4056/sigs.1453580.