+ All Categories
Home > Documents > Metabolons involving plant cytochrome P450s

Metabolons involving plant cytochrome P450s

Date post: 23-Jan-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
14
Abstract Arranging biological processes into ‘‘compartments’’ is a key feature of all eukaryotic cells. Through this mechanism, cells can drasti- cally increase metabolic efficiency and manage complex cellular processes more efficiently, sav- ing space and energy. Compartmentation at the molecular level is mediated by metabolons. A metabolon is an ordered protein complex of sequential metabolic enzymes and associated cellular structural elements. The sub-cellular organization of enzymes involved in the synthesis and storage of plant natural products appears to involve the anchoring of metabolons by cyto- chrome P450 monooxygenases (P450s) to specific domains of the endoplasmic reticulum (ER) membrane. This review focuses on the current evidence supporting the organization of metabo- lons around P450s on the surface of the ER. We outline direct and indirect experimental data that describes P450 enzymes in the phenylpropa- noid, flavonoid, cyanogenic glucoside, and other biosynthetic pathways. We also discuss the limitations and future directions of metabolon research and the potential for application to metabolic engineering endeavors. Keywords Cytochrome P450 Metabolon Enzyme interaction ER localization Cytochrome P450 reductase Abbreviations 4CL 4-Coumarate: CoA ligase AFM Atomic force microscopy C4H Cinnamate 4-hydroxylase CHI Chalcone isomerase CHS Chalcone synthase CPR NADPH-cytochrome P450 reductase DFR Dihydroflavonol reductase ER Endoplasmic reticulum F3H Flavanone 3b-hydroxylase F3¢H Flavonoid 3¢-hydroxylase F3¢5¢H Flavonoid 3¢,5¢-hydroxylase FLIM Fluorescence lifetime imaging microscopy FRET Fluorescence energy resonance transfer I2¢H Isoflavone 2¢-hydroxylase IFS Isoflavone synthase IOMT Isoflavone O-methyltransferase P450 Cytochrome P450 monooxygenase PAL Phenylalanine ammonia-lyase L. Ralston Sigma-Aldrich Biotechnology, Life Science and High Technology Center, 2909 Laclede Avenue, St. Louis, MO 63103, USA O. Yu (&) Donald Danforth Plant Science Center, 975 North Warson Road, St. Louis, Missouri 63132, USA e-mail: [email protected] Phytochem Rev (2006) 5:459–472 DOI 10.1007/s11101-006-9014-4 123 Metabolons involving plant cytochrome P450s Lyle Ralston Oliver Yu Received: 14 January 2006 / Accepted: 7 July 2006 / Published online: 28 October 2006 ȑ Springer Science+Business Media B.V. 2006
Transcript

Abstract Arranging biological processes into

‘‘compartments’’ is a key feature of all eukaryotic

cells. Through this mechanism, cells can drasti-

cally increase metabolic efficiency and manage

complex cellular processes more efficiently, sav-

ing space and energy. Compartmentation at the

molecular level is mediated by metabolons. A

metabolon is an ordered protein complex of

sequential metabolic enzymes and associated

cellular structural elements. The sub-cellular

organization of enzymes involved in the synthesis

and storage of plant natural products appears to

involve the anchoring of metabolons by cyto-

chrome P450 monooxygenases (P450s) to specific

domains of the endoplasmic reticulum (ER)

membrane. This review focuses on the current

evidence supporting the organization of metabo-

lons around P450s on the surface of the ER.

We outline direct and indirect experimental data

that describes P450 enzymes in the phenylpropa-

noid, flavonoid, cyanogenic glucoside, and other

biosynthetic pathways. We also discuss the

limitations and future directions of metabolon

research and the potential for application to

metabolic engineering endeavors.

Keywords Cytochrome P450 Æ Metabolon ÆEnzyme interaction Æ ER localization ÆCytochrome P450 reductase

Abbreviations4CL 4-Coumarate: CoA ligase

AFM Atomic force microscopy

C4H Cinnamate 4-hydroxylase

CHI Chalcone isomerase

CHS Chalcone synthase

CPR NADPH-cytochrome P450 reductase

DFR Dihydroflavonol reductase

ER Endoplasmic reticulum

F3H Flavanone 3b-hydroxylase

F3¢H Flavonoid 3¢-hydroxylase

F3¢5¢H Flavonoid 3¢,5¢-hydroxylase

FLIM Fluorescence lifetime imaging

microscopy

FRET Fluorescence energy

resonance transfer

I2¢H Isoflavone 2¢-hydroxylase

IFS Isoflavone synthase

IOMT Isoflavone O-methyltransferase

P450 Cytochrome P450 monooxygenase

PAL Phenylalanine ammonia-lyase

L. RalstonSigma-Aldrich Biotechnology, Life Science and HighTechnology Center, 2909 Laclede Avenue, St. Louis,MO 63103, USA

O. Yu (&)Donald Danforth Plant Science Center, 975 NorthWarson Road, St. Louis, Missouri 63132, USAe-mail: [email protected]

Phytochem Rev (2006) 5:459–472

DOI 10.1007/s11101-006-9014-4

123

Metabolons involving plant cytochrome P450s

Lyle Ralston Æ Oliver Yu

Received: 14 January 2006 / Accepted: 7 July 2006 / Published online: 28 October 2006� Springer Science+Business Media B.V. 2006

Introduction

The ability to partition biological processes

between discrete compartments is a major

evolutionary advantage of eukaryotes. Metabolic

processes in higher organisms are compartmen-

talized at multiple levels ranging from the macro-

organizational level of a tissue or organ, to

organization within different cell types, to sub-

cellular organization at the level of the organelle,

to molecular organization at the level of the

multienzyme complex or metabolon. Compart-

mentalization at multiple levels allows an organ-

ism to manage various biological processes more

precisely and with greater metabolic efficiency.

The use of metabolons extends the coordination

of metabolic compartmentalization to the

molecular level.

Paul Srere first defined a metabolon as a

‘‘supramolecular complex of sequential metabolic

enzymes and cellular structural elements’’ (Srere

1985). Srere posited a number of characteristics as

being inherent to metabolon structure and func-

tion: that component enzymes would channel

substrates for increased catalytic efficiency; that

flux through the metabolon would be regulated by

the association and dissociation of component

enzymes; that these component enzymes would

form interactions with structural elements of the

cell; and that assembly of the metabolon would be

controlled, at least in part, through the coordinate

regulation of the genes encoding its component

enzymes. Such organization at the molecular level

offers a number of potential benefits. The

advantages of channeling intermediates through a

metabolon include the ability to attain high local

concentrations of pathway intermediates, bypass

kinetic constraints resulting from diffusion of

intermediates into the bulk solvent, decrease

transition time of intermediates between the ac-

tive sites of sequential enzymes, protect labile or

reactive intermediates from the bulk solvent, and

coordinate the metabolic flux through pathways

with shared enzymes and/or intermediates.

Classic examples of metabolons are found in

both prokaryotes and eukaryotes (Srere 1987).

Metabolons play well-established roles in the

synthesis of amino acids, proteins, and nucleic

acids (Srere 1987; Mendes et al. 1995). For

example, the tryptophan synthase complex is a

classical metabolon that forms an intra-molecular

hydrophobic tunnel to channel indole between

the active sites of the two subunits of the enzyme

complex (Anderson et al. 1995; Miles 2001). In

addition, several well-defined examples of me-

tabolons exist in plants. Many of these examples

come from central or primary metabolic pathways

and include enzyme complexes of the Calvin–

Benson cycle (Graciet et al. 2004; Winkel 2004),

peroxisomal enzymes involved in the photore-

spiratory cycle (Reumann 2000), and enzymes

involved in polyamine biosynthesis (Panicot et al.

2002). In many cases, metabolon formation ap-

pears to involve specific interactions between

operationally soluble enzymes, cytoskeletal ele-

ments, or membrane proteins (Mendes et al.

1995; Srere 2000).

Growing experimental evidence suggests that

the enzymes of various natural product biosyn-

thetic pathways in plants may be organized as

metabolons on the cytoplasmic surface of the

endoplasmic reticulum (ER). In these metabo-

lons, cytochrome P450 monooxygenases (P450s)

are hypothesized to serve as nucleation points

that anchor the metabolon to a specific region

within the cell (Winkel 2004; Jorgensen et al.

2005). The majority of plant P450s are tethered to

the ER via a membrane-spanning amino-terminal

domain (Chapple 1998) (Fig. 1). This localization

provides a platform for potential interactions

between proteins forming a given metabolon. The

P450-anchored metabolons are suspected to

associate with specific domains on ER mem-

branes, thus forming ‘‘metabolic centers’’ (Galili

et al. 1998). This phenomenon has been hypoth-

esized to be facilitated by small accessory proteins

that mediate interactions between component

enzymes of a metabolon and cytoskeletal ele-

ments (Chuong et al. 2004; Graciet et al. 2004;

Winkel 2004). Since cellular processes must con-

stantly adjust in response to various environ-

mental and physiological stimuli, dynamic

formation and dissociation of metabolons may

provide an additional level of metabolic regula-

tion in biosynthetic pathways.

This review focuses on the organization of

metabolons around P450s in plant natural product

pathways. Beyond a metabolic role, P450s are

460 Phytochem Rev (2006) 5:459–472

123

also thought to play a structural role in these

complexes, providing a platform for their partners

to establish and maintain protein–protein inter-

actions. We will discuss the direct and indirect

evidence supporting the hypothesis of P450-an-

chored metabolons in several plant natural

product pathways.

Interactions between P450s and

NADPH-cytochrome P450 reductases

Given the central role of P450s in anchoring

metabolons of plant natural product biosynthesis,

the molecular architecture of the P450s provides a

framework for organizing larger macromolecular

complexes. All P450s rely on a heme co-factor to

split molecular oxygen, inserting one oxygen

atom to the substrate and reducing the other

oxygen atom to water. To accomplish this reac-

tion, a coupled, stepwise electron supply origi-

nating from NAD(P)H is indispensable (Denisov

et al. 2005). The electron supply is provided by

NADPH-cytochrome P450 reductase (CPR)

(Paine et al. 2005). Depending on the type of

P450, the CPR is either fused to the P450 to form

a multifunctional protein or operates as a sepa-

rate membrane-bound protein.

Structural and functional studies of the multi-

functional or ‘‘self-sufficient’’ P450s, such as the

fatty acid x-2 hydroxylase P450 BM3

(CYP102A1) from bacterium Bacillus megateri-

um, provide insight on the interaction between

the P450 and CPR. The multifunctional P450s

evolved through the fusion of the P450 and CPR

enzymes. This type of P450 consists of two dis-

crete domains that can be functionally separated

genetically and proteolytically. The first 472

amino-terminal residues of P450 BM3 comprise

the monooxygenase domain, while the remainder

of the enzyme makes up the reductase domain

(Munro et al. 2002). In P450 BM3, NADPH re-

duces a FAD flavin to a transient hydroquinone,

and a flavin mononucleotide (FMN)-binding do-

main shuttles electrons between the FAD in the

CPR domain and the heme in the P450 domain

(Sevrioukova et al. 1999). The fusion provides a

rare insight into the mechanism of the coupling of

the two units and how protein–protein interac-

tions facilitate the transfer of electrons (for re-

views see (Munro et al. 2002)). Structural and

biochemical data from P450 BM3 suggests that

the rate of electron transfer between NADPH

and FAD in the bipartate protein is much faster

than the observed catalytic activity of the isolated

monooxygenase domain (Munro et al. 1996). Two

basic residues, Lys572 and Lys580, stabilize the

hydroquinone-form of FAD and are essential

for efficient coupling of the electron transfer

chain (Sevrioukova et al. 1999; Murataliev and

Fig. 1 A plant cytochrome P450 localized to the corticalER membrane. (A) Soybean isoflavone synthase (IFS,CYP93C1) was fused in-frame with a yellow fluorescentprotein (EYFP) and transiently expressed in tobacco leafepidermal cells. IFS was localized to the ER network(yellow color, Ralston and Yu, unpublished data). (B) As a

control, soybean chalcone isomerase (CHI) was fused in-frame with a cyan fluorescent protein (ECFP) andtransiently expressed in tobacco leaf epidermal cells.Soluble CHI was localized to the cytoplasm and nucleus(green color). The size bar is 10 lm

Phytochem Rev (2006) 5:459–472 461

123

Feyereisen 2000). Overall, for any P450 to be

functional, a tightly coupled CPR is essential.

In contrast to the multifunctional P450s, the

typical eukaryotic P450 interacts with a separate

membrane-bound CPR (Paine et al. 2005). The

amino-terminal region of CPR (approximately 60

amino acids in length) is a hydrophobic mem-

brane-spanning domain that anchors CPR to

the cytoplasmic surface of the ER. Similar to the

carboxyl-terminal region of the P450 BM3, the

function of the membrane-bound CPR is to ferry

electrons from NAD(P)H through FAD and

FMN to the P450 (Murataliev et al. 2004). Com-

pared to the vast number of P450 genes that exist

in plant genomes, the number of CPR genes is

much lower, and sequence homology is more

highly conserved than in P450s (Koopmann and

Hahlbrock 1997; Urban et al. 1997). This suggests

that a limited number of CPRs would couple with

a diverse array of P450 partners and, by exten-

sion, interact with a variety of metabolons.

Interactions between CPR and P450s have

been investigated mainly in mammalian systems.

A rat CPR without the amino-terminal mem-

brane domain has been crystallized (Wang et al.

1997). The enzyme is composed of three domains:

an FAD-binding domain similar to ferredoxin-

NADP+ reductase, an FMN-binding domain

similar to flavodoxins, and a linear linker that

connects the other two domains. The linker do-

main is responsible for the positioning of the co-

factors FMN and FAD in close proximity to the

heme of the P450 to facilitate efficient electron

transfer. Mutations in the linker region can cause

significant differences in the relative positions of

the two flavin domains and significantly reduce a

P450’s catalytic activities (Hubbard et al. 2001).

Similarly, removing the membrane anchor of a

mammalian CPR will produce a soluble and

enzymatically active CPR. However, the soluble

CPR will be unable to support the activity of a

P450 in vitro, due to the uncoupling of the elec-

tron transfer chain (Venkateswarlu et al. 1998).

The direct interaction of CPR with P450s was

visualized by atomic force microscopy (AFM) on

a reconstituted phospholipid bilayer disk con-

taining CPR and the P450 CYP2B4 (Bayburt

et al. 1998; Bayburt and Sligar 2002). CYP2B4

was found to protrude above the lipid surface,

3.5 nm. This is thought to orient the CPR such

that both the FMN- and FAD-binding domains

lie close to the membrane surface, allowing a

close interaction between FMN and the P450

heme (Wang et al. 1997). The lipid bilayer of the

ER membrane may also contribute to interac-

tions between CPR and P450 by altering the

composition of the bilayers to provide a relatively

stable support for the enzyme complexes when

needed (Ingelman-Sundberg et al. 1981, 1983).

More importantly, a group of basic residues,

Arg45, Lys46, Lys47, and Lys48, which lies at the

beginning of FMN-binding domain, may interact

with the phospholipid head groups of the mem-

brane and restrict the movement of CPR on the

membrane surface (Wang et al. 1997; Zhao et al.

1999). Similarly, a second group of basic residues

(Lys72, Lys74, Lys75, Arg78, Arg97, Lys100,

His103, and Arg108) at the surface of the FMN-

binding domain may provide an additional

membrane-binding site.

Surface residues on the P450 are also likely to

contribute to the CPR–P450 interaction. Specific

lysine and arginine residues on rat CYP1A1 are

shown to be involved in the formation of an

electron transfer complex with CPR (Shimizu

et al. 1991). Similarly, mammalian CYP2B1 and

CYP2B4 may also rely on complementary elec-

trostatic charges to bind with CPR (Shen and

Strobel 1993; Bridges et al. 1998). Site-directed

mutagenesis of CYP2B4 identified a series of ly-

sine and arginine residues on the proximal surface

of the heme region that interact with CPR’s FMN

region (Bridges et al. 1998). Moreover, chemical

cross-linking studies indicate that several acidic

residues on CPR are involved in the interaction

with CYP1A1 (Nadler and Strobel 1991). Other

evidence also suggests that the FMN-binding do-

main is the docking surface of the two enzymes,

as with P450 BM3. Electrostatic potential mea-

surement of the FMN-binding domain of a human

CPR shows clusters of acidic residues that could

form ion-pair interactions with a P450 (Shen and

Strobel 1995). In fact, a decreased association (Kd

10–90 nM) between rabbit CPR and CYP2B4 was

observed when the ionic strength of the buffer

was increased 10-fold (Davydov et al. 2000).

Mutations of the clusters located at the opposite

side of the FMN domain significantly reduces

462 Phytochem Rev (2006) 5:459–472

123

P450 activity, suggesting that the P450 may par-

tially ‘‘engulf’’ CPR at the FMN domain to bring

the heme and FMN in closer contact (Zhao et al.

1999).

In spite of the above evidence, the proposed

metabolon model has to reconcile with the fact

that, at least in mammalian cells, P450s are

present in a 10–25-fold molar excess over CPR

(Estabrook et al. 1971; Peterson et al. 1976). This

ratio dictates that any association of CPR with a

given P450 has to be transient and dynamic to

allow multiple P450s from different pathways to

have access to this essential partner. Such inter-

actions in plant cells are likely to be more com-

plicated due to the higher number of P450 genes

found in plant genomes.

Entry into the phenylpropanoid pathway:the CYP73A-related metabolon

Arguably the strongest physical evidence for the

existence of metabolic channels in a plant natural

product pathway involves the interaction between

the first two enzymes of the phenylpropanoid

pathway (Fig. 2A). The phenylpropanoid path-

way exists in all higher plants and is a major

consumer of energy and carbon fixed by photo-

synthesis (Werck-Reichhart and Feyereisen

2000). This pathway produces a myriad of

structurally diverse phenolic compounds, includ-

ing lignins, stilbenes, aurones, flavonoids, and

isoflavonoids. These compounds serve as signal

molecules, phytoalexins, anti-feedants, pigments,

UV protectants, and cell wall components in

plants. Since this pathway consumes large

amounts of biochemical resources, phenylpropa-

noid metabolism is tightly regulated both trans-

criptionally and post-transcriptionally (Weisshaar

and Jenkins 1998). Metabolic channeling between

enzymes at the entry point of pathway is sug-

gested to provide an additional level of regula-

tion.

The entry point enzyme of the phenylpropa-

noid pathway is phenylalanine ammonia-lyase

(PAL). PAL commits phenylalanine to the phe-

nylpropanoid pathway, deaminating phenylala-

nine and converting it into trans-cinnamate and

ammonia. In all species examined so far, PAL is

encoded by a multi-gene family (Dixon et al.

2002). Based on differential gene expression

patterns, it has long been suspected that the

Fig. 2 Schematic diagram of the phenylpropanoid (A)and dhurrin pathways (B). The P450 enzymes are in reditalics. Enzyme abbreviations are listed in the text, except

for CHR, chalcone reductase; STS, stilbene synthase; HID,2-hydroxyisoflavanone dehydratase; and IFR, isoflavonereductase

Phytochem Rev (2006) 5:459–472 463

123

various PAL isoforms are activated by different

environmental signals to divert phenylalanine

into different branches of the phenylpropanoid

pathway (Kao et al. 2002).

The second enzyme of the phenylpropanoid

pathway is the P450 cinnamate 4-hydroxylase

(CYP73A, C4H). C4H adds a hydroxyl group at

the C4 (para) position of trans-cinnamate to form

p-coumarate (Russell and Conn 1967; Schoch

et al. 2003). C4H is encoded by a single copy gene

in Arabidopsis and occurs in only one or two

copies in other species studied to date (Plant

Cytochrome P450 Database; http://www.drnel-

son.utmem.edu/P450dbplant.html). C4H was the

first plant P450 to be functionally characterized,

and it is among the most extensively studied of

the plant P450s (Chapple 1998).

PAL exists as an operationally soluble homo-

tetramer with no obvious membrane localization

domains (Wanner et al. 1995); however, a num-

ber of reports describe the association of PAL

with endomembranes in plant cells. In 1975,

based on sub-cellular fractionation and micro-

some enzyme activity assays, Czichi and Kindl

reported that PAL and C4H may assemble on

microsomal membranes as an enzyme complex

(Czichi and Kindl 1975). While C4H activity is

located exclusively in the microsomal fraction of

plant cells and PAL activity resides in the soluble

fraction of plant cell extracts, they noted that both

PAL and C4H activities co-localized to the

microsomal fraction. Substrate feeding experi-

ments suggested that cinnamate produced from

[3H]-phenylalanine by PAL is preferred over

exogenously added [14C]-cinnamate by C4H.

Hence, the majority of the C4H product,

p-coumarate, was derived from radiolabeled

phenylalanine (Czichi and Kindl 1977). Further

feeding assays carried out by others demonstrated

that disrupting the microsomal structure, which

may disrupt the interaction of the two enzymes,

led to the increased conversion of exogenously

added cinnamate conversion to p-coumarate

(Hrazdina and Wagner 1985). Taken together,

these earlier experiments provide strong indirect

evidence that PAL and C4H are co-localized on

the cytoplasmic surface of the ER and that cin-

namate is channeled directly to the active site of

C4H from PAL.

Rick Dixon’s lab has produced a good deal of

evidence for the existence of a tobacco PAL-C4H

metabolon. Rasmussen et al. demonstrated that

cinnamate produced from [3H]-phenylalanine

does not equilibrate with exogenously added

[14C]-cinnamate, indicating channeling between

PAL and C4H (Rasmussen and Dixon 1999).

Further, sub-cellular fractionation and protein gel

blot analysis demonstrated that tobacco PAL1 is

localized to both the soluble and microsomal

protein fractions, whereas the PAL2 isozyme is

strictly localized to the soluble fraction. More

recently, Achinine et al. provided more evidence

of direct enzyme interactions between PAL and

C4H. They used a combination of biochemical

and microscopic techniques to study the locali-

zation of PAL1, PAL2, and C4H (Achnine et al.

2004). Using these techniques, they verified the

sub-cellular localization of PAL1 and PAL2.

Moreover, they showed that over-expression of

C4H resulted in the ER localization of both

PAL1 and PAL2. This suggests that C4H itself

may be responsible for the membrane association

of PAL. Surprisingly, co-expression of unlabeled

PAL1 with PAL2-GFP resulted in a shift of flu-

orescence from ER membranes to the cytoplasm

in C4H over-expressing plants, whereas co-

expression of unlabeled PAL2 with PAL1-GFP

did not affect PAL1-GFP localization. This indi-

cates that PAL1 has a higher affinity for its

membrane localization site than PAL2. Fluores-

cence energy resonance transfer (FRET) studies

based on dual-labeling immuno-fluorescence

showed the PAL and C4H are located within

100 A of each other on the membrane. In addi-

tion, negative photo-bleaching results may sug-

gest that the connection between PAL and C4H is

fluid (Achnine et al. 2004).

In whole, there is extensive compelling evi-

dence suggesting that C4H anchors an ER-local-

ized metabolon at the entry point of the

phenylpropanoid pathway. However, at least one

example can be found in the literature disputing

the existence of a metabolon between PAL and

C4H. Ro and Douglas argue that enzyme kinetics

and biochemical coupling of PAL and C4H are

enough to drive carbon flux into the phenyl-

propanoid pathway (Ro and Douglas 2004). They

reconstituted the entry point of the pathway by

464 Phytochem Rev (2006) 5:459–472

123

expressing PAL, C4H, and CPR in yeast. They

then fed the engineered yeast [3H]-phenylalanine

and [14C]-cinnamate simultaneously. In this sys-

tem, inhibition of C4H by P450 inhibitors indeed

reduced PAL activity, demonstrating biochemical

coupling. However, unlike experiments using

plant microsome fractions, the two radiolabeled

substrates showed equal accessibility to C4H,

indicating that no channeling occurs between

PAL and C4H. Earlier analysis using transgenic

plants supported this biochemical coupling the-

ory: when the C4H gene was silenced in tobacco,

PAL activity was significantly reduced while PAL

silencing did not disturb C4H activity. This data

provides further evidence of a feedback loop at

the entry point of the phenylpropanoid pathway

(Blount et al. 2000). Clearly, biochemical cou-

pling is part of the regulation mechanism between

PAL and C4H. Yet the physical interactions be-

tween these enzymes inside plant cells should not

be excluded based on experiments in yeast.

Cytoskeletal elements and accessory proteins

critical for interactions between PAL and C4H

may well be lacking in a heterologous host. It is

possible that at the entry point of the phenyl-

propanoid pathway, both regulation mechanisms

co-exist inside plant cells.

Flavonoid biosynthesis: CYP75B-related

metabolon

While the majority of the carbon flux into the

phenylpropanoid pathway is directed towards

lignin production (discussed below), the second

greatest carbon sink from this pathway is flavo-

noid biosynthesis. Flavonoid compounds are

produced by the coordinate action of a series of

operationally soluble and membrane-bound

cytoplasmic enzymes (Fig. 2A) (Winkel-Shirley

2001a). Soluble enzymes of the flavonoid branch

pathway include 4-coumarate: CoA ligase (4CL),

chalcone synthase (CHS), chalcone isomerase

(CHI), flavanone 3b-hydroxylase (F3H), and di-

hydroflavonol reductase (DFR). A number of

P450s also participate in this branch of the path-

way. Flavonoid 3¢-hydroxylase (CYP75B, F3¢H)

modifies the B-ring of the flavanone backbone at

the C3¢ position. Another P450, flavonoid 3¢,5¢-

hydroxylase (CYP75A, F3¢5¢H), is found in a

limited number of species and modifies the B-ring

at the C5¢ position (Winkel-Shirley 2001b).

There is strong evidence suggesting that the

operationally soluble enzymes of the flavonoid

branch pathway participate in metabolons. In a

yeast two-hybrid assay, Burbulis and Winkel-

Shirley demonstrated that CHS, CHI, and DFR

interact with one another in an orientation-

dependent manner (Burbulis and Winkel-Shirley

1999). Affinity chromatography and immuno-

precipitation assays also demonstrated interac-

tions between CHS, CHI, and F3H in lysates from

Arabidopsis seedlings (Burbulis and Winkel-

Shirley 1999). Although three-dimensional struc-

tures are available for CHS and CHI (Ferrer et al.

1999; Jez et al. 2000), no structural model of this

proposed macromolecular complex has been

published. However, mechanistic studies of CHS

and CHI suggest the need for metabolic chan-

neling in this pathway. The non-enzymatic cycli-

zation of chalcones into flavanones readily occurs

in solution but yields an enantiomeric mix of

biologically inactive and active isomers. Chan-

neling between CHS and CHI would prevent the

formation of mixed isomers (Jez et al. 2002).

Interestingly, the catalytic efficiency of CHI (kcat/

Km = 109 M–1 min–1) approaches the diffusion

limit, so one might ask why CHI would need

metabolites to be directed towards it. One possi-

ble answer is that the moderately lipophilic nat-

ure of chalcones and flavanones may require

channeling to limit the potential for sequestration

in cellular membranes. Moreover, in legumes,

channeling through the metabolon may ‘‘slow’’

the activity of CHI and protect the chalcone pool

from complete conversion into naringenin (Jez

et al. 2002).

There is no direct evidence that the complex

formed by CHS, CHI, F3H, and DFR interacts

with P450s. However, if these enzyme do form

such an interaction, the most likely candidate is

F3¢H (or perhaps F3¢5¢H, if it is expressed). The

complex could also involve the upstream P450

C4H, thus forming a metabolon that encompasses

the majority of the enzymes of the general phe-

nylpropanoid and flavonoid branch pathways

(Winkel 2004; Jorgensen et al. 2005). There is

interesting indirect evidence suggesting that CHS/

Phytochem Rev (2006) 5:459–472 465

123

CHI-related metabolons are indeed localized to

the ER. First, CHI contains a motif that indicates

the enzyme might be modified post-translation-

ally by a fatty acid moiety with a thiol-sensitive

linkage. This would provide a mechanism for the

membrane localization of this soluble enzyme

(Burbulis and Winkel-Shirley 1999). Earlier re-

ports indicate that in the hypocotyl epidermis of

buckwheat (Fagopyrum esculentum), CHS activ-

ity and protein were detected at the cytoplasmic

face of the rough ER, based on linear sucrose

density gradient fractionation, immuno-blots, and

immuno-gold analysis (Hrazdina et al. 1987). Fi-

nally, membrane localization of CHS, and co-

localization of CHS and CHI were observed at

the ER and tonoplast in Arabidopsis root cells

(Saslowsky and Winkel-Shirley 2001). The anti-

bodies detected an electron-dense region with

membrane structures. In the Arabidopsis tt7 mu-

tant in which the P450 F3¢H is deleted, the elec-

tron-dense regions containing these two enzymes

were not detected. However, CHS and CHI were

still found to co-localize to the ER and tonoplast

in the tt7 mutant, suggesting that other proteins

may function in recruiting the soluble flavonoid

pathway enzymes to membranes (Saslowsky and

Winkel-Shirley 2001). Interestingly, recent data

suggests that flavonoids and CHS and CHI pro-

teins are also localized to nucleus of Arabidopsis

(Saslowsky et al. 2005). However, the implica-

tions of this discovery are not yet clear. Collec-

tively, the evidence amassed over the past

30 years suggests the existence of metabolons

involving enzymes of the general phenylpropa-

noid and flavonoid branch pathways that are an-

chored to the cytoplasmic surface of the ER by

P450s.

Isoflavonoid biosynthesis: CYP93C-relatedmetabolons

In legumes, the majority of the naringenin pro-

duced by CHS is converted to isoflavonoids by

the P450 isoflavone synthase (CYP93C, IFS)

(Yu et al. 2003). Isoflavonoids are major defense

compounds for legumes. While isoflavonoids are

constitutively produced at low levels, their bio-

synthesis is drastically induced upon pathogen

infection. During the defense response, isoflavo-

noids are further metabolized into more complex

and more potent phytoalexins by a set of legume-

specific enzymes (Dixon et al. 2002). In the

Medicago family, the first such modification is the

methylation of the 4¢-hydroxy on the B-ring of

the isoflavones. This step is mediated by an

operationally soluble isoflavone-O-methyltrans-

ferase (IOMT).

Liu et al. observed that IOMT from alfalfa

(Medicago sativa) was translocated to the ER upon

elicitor treatment, co-localizing with IFS (Liu and

Dixon 2001). They also noted that feeding studies

in alfalfa cell suspension cultures suggested chan-

neling between IFS and the IOMT. Radiolabel

from [3H]-liquiritigenin was preferentially incor-

porated into formononetin, the product of IOMT,

as well as other downstream isoflavonoids. How-

ever, radiolabel from daidzein, the in vitro sub-

strate of IOMT, was not incorporated into

downstream isoflavonoids. This data suggests that

the methyltransferase is closely associated with

IFS, facilitating rapid methylation and stabilization

of the product of IFS. This association was also

used to explain the observed differences in IOMT

regio-specificity between in vivo and in vitro assays

(Liu and Dixon 2001). However, different types of

IOMTs have been cloned, indicating that these

differences in regio-specificity may instead be

caused by the activity of different IOMT enzymes

(Akashi et al. 2000, 2003). Liu et al. provided a

model suggesting that intermediates of the isofl-

avonoid pathway could rapidly flow from one en-

zyme center anchored by IFS to next enzyme

center in a metabolon anchored by another P450,

isoflavone 2¢-hydroxylase (CYP81E, I2¢H), and

eventually enter into the vacuole (Liu and Dixon

2001). This process is more complicated in other

legume species. In these species, the product of

I2¢H is prenylated by plastid-associated pren-

yltransferases to form pterocarpans and fur-

anocoumarins before entering the vacuole (Dhillon

and Brown 1976). This must require the shuttling of

compounds between different organelles for the

metabolon model to hold true.

The association of IFS with upstream enzymes

such as CHI and CHS can only be implied. Sev-

eral groups have shown that efforts to engineer

isoflavonoid synthesis in non-legume plants by

466 Phytochem Rev (2006) 5:459–472

123

introducing IFS resulted in a disproportionate

decrease in flavonoid production, which cannot

be explained by a shift in metabolic flux alone

(Yu et al. 2000; Liu et al. 2002). In transgenic

Arabidopsis expressing IFS, the production of

genistein can be increased 3-fold by irradiation

with UV light, but the ratio of genistein in total

flavonoid levels actually decreased. This suggests

that the pool of naringenin substrate is not

equally accessible to the flavonoid and isoflavo-

noid branches of the pathway (Yu et al. 2000).

Similar experiments using transgenic tobacco and

maize cell cultures demonstrated similar ‘‘com-

partmentalization’’ of the naringenin substrate

(Yu et al. 2000). Additionally, the treatment of

tobacco leaves with UV light to increase na-

ringenin levels resulted in elevated flavonol levels

but failed to raise anthocyanin or isoflavone lev-

els. This data suggests that flux through the phe-

nylpropanoid pathway is tightly channeled to

flavonol production in tobacco leaves, and IFS

has difficulty accessing this substrate.

To delineate enzyme interactions between IFS

and endogenous flavonoid pathway genes, a CHI

from alfalfa, shown to be an legume-specific

isomerase by its ability to convert isoliquiritigenin

into liquiritigenin, was transformed into Arabid-

opsis and then genetically crossed into a trans-

genic Arabidopsis line carrying the IFS gene (Liu

et al. 2002). The combination of genes enhanced

genistein accumulation moderately. However,

flavonol accumulation was significantly reduced

compared to transgenic plants carrying only the

legume-specific CHI. The disproportionate

reduction of flavonol biosynthesis caused by the

presence of IFS further confirmed that the flux of

substrate is preferentially channeled towards

endogenous flavonoid biosynthesis (Liu et al.

2002). One possible explanation offered for this

outcome is that the membrane-localized IFS

might interfere with the assembly of metabolic

complexes dedicated to flavonoid biosynthesis,

which may be anchored to the ER by the P450

F3¢H in Arabidopsis (Yu et al. 2000). When the

IFS and CHI were introduced into a tt6/tt3 double

mutant background, genistein accumulation was

enhanced by up to 30-fold as compared to plants

expressing IFS alone. This mutant has structural

defects in both F3¢H and DFR genes and is thus

blocked in flavonol and anthocyanin production.

Again, this suggests that the bottleneck for isof-

lavone production in Arabidopsis is competition

for flavanones between IFS and endogenous fla-

vonol biosynthetic enzymes (Liu et al. 2002).

This brings us to an interesting topic regarding

the regulation of the competing branch pathways.

In legumes, where the flavonoid and isoflavonoid

pathways co-exist, the flux of flavanone interme-

diates should be differentially divided into the

two pathways in response to abiotic and biotic

stresses. Plants can activate the expression of a

subset of genes that divert the precursor phenyl-

alanine into specific products of the pathway

using a number of relatively well-defined tran-

scription factors (Yu et al. 2003). However, me-

tabolons could play a major role in controlling

flux through the pathway. In fact, two functionally

different CHI exist in legumes (Shimada et al.

2003; Ralston et al. 2005): the type I CHI exists in

all higher plants and converts tretrahydroxy-

chalcones into the flavanone naringenin, while the

legume specific type II CHI converts both tetra-

hydroxy-chalcones and trihydroxy-chalcones into

naringenin and liquiritigenin, respectively. The

two types of CHI are differentially expressed, and

only the type II CHI is induced by defense signals

(Ralston et al. 2005). It’s possible that the type II

CHI, together with other isoflavone biosynthetic

enzymes may form their own metabolic channels,

independent of the flavonoid channels described

in Arabidopsis (Fig. 3) (Winkel 2004; Yu and

McGonigle 2005).

Cyanogenic glucoside biosynthesis: CYP79A/

CYP71E-related metabolons and others

The biosynthesis of cyanogenic glucosides in sor-

ghum adds another dimension to the involvement

of P450s in metabolons. Dhurrin is a type of cya-

nogenic glucoside that is produced from the amino

acid tyrosine by two P450s, CYP79A1 and

CYP71E1, and the UDP-glucosyltransferase

UGT85B1 (Fig. 2B) (Celenza 2001; Morant et al.

2003; Jorgensen et al. 2005). Feeding assays using

the radioisotopically labeled substrate tyrosine and

the intermediate Z-p-hydroxyphenylacetal dox-

ime demonstrated clearly that the two P450s are

Phytochem Rev (2006) 5:459–472 467

123

highly channeled, with the majority of the radio-

label in dhurrin coming from tyrosine (Moller and

Conn 1980). Epitope tagging and confocal laser

scanning microscopy revealed that this metabolon

is located in a distinct domain of the ER (Nielsen

and Moller 2005). From an evolutionary point of

view, the coupling of the three enzymes likely arose

out of the need to prevent the highly labile and

toxic p-hydroxymandelonitrile intermediate from

releasing cyanide into the cytoplasm.

Genetic engineering has provided further sup-

port for the existence of P450 anchored metabo-

lons. When the entire cyanogenic glucoside

pathway was introduced into Arabidopsis by

expressing all three enzymes, the production of

dhurrin had virtually no effect on the metabolic

profiles of the host plants (Kristensen et al. 2005).

This toxic product was effectively contained and

sequestered in vacuoles. However, when only the

two P450s were expressed in Arabidopsis, without

the metabolon partner UGT85B1, plants showed

a variety of toxicity-related phenotypes. Inter-

estingly, when introduced separately into Ara-

bidopsis, the first enzyme CYP79A1 was able to

establish highly efficient interactions with down-

stream glucosinolate-producing enzymes, and al-

tered the overall glucosinolate profile of

Arabidopsis (Bak et al. 1999).

With the rapid advances in the understanding

of cellular and biochemical processes, metabolons

have been invoked in other metabolic pathways.

For example, the lignin biosynthetic enzymes

have previously been proposed to form a meta-

bolic grid, in which the flux could proceed in a

relatively random manner (Humphreys and

Chapple 2002; Boudet et al. 2003), however, re-

cent molecular analysis, particularly the new en-

zyme kinetic and substrate specificity

observations suggested that many of the routes

through the initially proposed grid are unlikely to

function in vivo, thus suggesting a more or less

linear metabolic pathway. Dixon et al. proposed

an interesting model suggesting that the linearity

of enzyme reactions is the result of metabolic

channeling among the key lignin enzymes (Dixon

et al. 2001). The metabolons are thought to pre-

vent the intermediates from coming into contact

with competing enzymes that might lead to the

development of a less efficient metabolic grid.

However, direct evidence of enzyme interactions

in this pathway is scarce. While the existence of

metabolons has been postulated in other plant

natural product pathways, including the alkaloid

and terpenoid pathways (Chappell 1995; Meme-

link 2004), investigators have yet to produce

compelling evidence of this phemonenon.

Conclusion and perspectives

A considerable amount of evidence argues for the

formation of metabolic P450-anchored enzyme

complexes in plant natural product pathways.

However, unlike the more conventional concept

of the stable multienzyme complexes that are

focused on completing one particular task more

efficiently, the P450-related metabolons probably

take on broader roles in regulating complicated

pathways and even metabolic grids. Such func-

tions require these metabolons to be able to

accommodate the constantly changing environ-

ment both inside and outside of the cell. There-

fore, most of these metabolons are likely to be

very dynamic and transient. The assembly and

Fig. 3 Proposed models of the flavonoid (A) and isofl-avonoid (B) metabolons. The enzyme abbreviations arethe same as in Fig. 2, except for UFGT, UDP-glucose:anthocyanidin-3-O-glucosyl trasnsferase; LDOX, leuco-anthocyanidin deoxygenase

468 Phytochem Rev (2006) 5:459–472

123

disassociation of enzymes within these complexes

is expected to occur rapidly in response to inter-

nal and external stimuli. As a consequence, it has

been very difficult to isolate and study these

complexes. Traditional biochemical assays,

including isotope dilution, substrate feeding, and

sub-cellular fractionation, while valuable, are

limited in their ability to provide direct evidence

of enzyme interactions. Co-immunoprecipitation,

protein cross-linking and affinity purifications are

best suited for stable interactions (Blancaflor and

Gilroy 2000); and yeast two-hybrid assays are not

applicable for membrane bound P450s. There-

fore, newer technologies, such as multiphoton

confocal co-localization (Blancaflor and Gilroy

2000), FRET and fluorescence lifetime imaging

microscopy (FLIM) analysis (Wallrabe and Pe-

riasamy 2005), split ubiquitin assays (Thaminy

et al. 2004), protein fragment complementation

assays (Subramaniam et al. 2001), and surface

plasmon resonance assays (Magee et al. 2002)

may provide direct and in vivo enzyme interac-

tion analysis.

If one or more plant P450 structures can be

resolved by X-ray crystallography, it will certainly

provide tremendous help in understanding of

nature of these protein–protein interactions, since

many of the soluble partners of P450s have al-

ready been resolved structurally. This will allow

directed mutagenesis to be carried out to evaluate

the interactions at the interface of these proteins.

However, the crystallization of membrane-bound

proteins is notoriously difficult and may require

intensive labor and extensive modifications of the

P450. In addition, direct visualization based on

AFM (Janovjak et al. 2005) or cryo-EM (Chiu

et al. 2005) is also possible if the enzyme com-

plexes can be functionally reconstituted or iso-

lated.

Understanding the structure and function of

the P450-related metabolons clearly is a consid-

erable challenge. However, these complexes very

likely provide another layer of cellular regulation

mechanisms and play important roles in plant

growth and development. The information ob-

tained on metabolons will also guide new meta-

bolic engineering efforts and allow the plant

community to harvest the desired products with

more certainty and higher efficiency.

Acknowledgements We would like to thank Dr. Joe Jezof the Danforth Center for the critical review of thismanuscript and for many thoughtful discussions on thetopic. The research in Dr. Yu’s lab is supported by theNational Science Foundation (MCB0519634), UnitedStates Department of Agriculture (CSREES: 2005-05190),and Missouri Soybean Merchandising Council (02-242).

References

Achnine L, Blancaflor EB, Rasmussen S, Dixon RA(2004) Colocalization of L-phenylalanine ammonia-lyase and cinnamate 4-hydroxylase for metabolicchanneling in phenylpropanoid biosynthesis. PlantCell 16:3098–3109

Akashi T, Sawada Y, Aoki T, Ayabe S (2000) Newscheme of the biosynthesis of formononetin involving2,7,4¢-trihydroxyisoflavanone but not daidzein as themethyl acceptor. Biosci Biotechnol Biochem 64:2276–2279

Akashi T, Sawada Y, Shimada N, Sakurai N, Aoki T,Ayabe S (2003) cDNA cloning and biochemicalcharacterization of S-adenosyl-L-methionine: 2,7,4¢-trihydroxyisoflavanone 4¢-O-methyltransferase, acritical enzyme of the legume isoflavonoid phyto-alexin pathway. Plant Cell Physiol 44:103–112

Anderson KS, Kim AY, Quillen JM, Sayers E, Yang XJ,Miles EW (1995) Kinetic characterization of channelimpaired mutants of tryptophan synthase. J BiolChem 270:29,936–29,944

Bak S, Olsen CE, Petersen BL, Moller BL, Halkier BA(1999) Metabolic engineering of p-hydroxybenzylg-lucosinolate in Arabidopsis by expression of the cya-nogenic CYP79A1 from Sorghum bicolor. PlantJ 20:663–671

Bayburt TH, Carlson JW, Sligar SG (1998) Reconstitutionand imaging of a membrane protein in a nanometer-size phospholipid bilayer. J Struct Biol 123:37–44

Bayburt TH, Sligar SG (2002) Single-molecule heightmeasurements on microsomal cytochrome P450 innanometer-scale phospholipid bilayer disks. Proc NatlAcad Sci USA 99:6725–6730

Blancaflor EB, Gilroy S (2000) Plant cell biology in thenew millennium: new tools and new insights. AmJ Bot 87:1547–1560

Blount JW, Korth KL, Masoud SA, Rasmussen S, Lamb C,Dixon RA (2000) Altering expression of cinnamicacid 4-hydroxylase in transgenic plants provides evi-dence for a feedback loop at the entry point into thephenylpropanoid pathway. Plant Physiol 122:107–116

Boudet AM, Kajita S, Grima-Pettenati J, Goffner D(2003) Lignins and lignocellulosics: a better control ofsynthesis for new and improved uses. Trends Plant Sci8:576–581

Bridges A, Gruenke L, Chang YT, Vakser IA, Loew G,Waskell L (1998) Identification of the binding site oncytochrome P450 2B4 for cytochrome b5 and cyto-chrome P450 reductase. J Biol Chem 273:17,036–17,049

Phytochem Rev (2006) 5:459–472 469

123

Burbulis IE, Winkel-Shirley B (1999) Interactions amongenzymes of the Arabidopsis flavonoid biosyntheticpathway. Proc Natl Acad Sci USA 96:12,929–12,934

Celenza JL (2001) Metabolism of tyrosine and tryptophan– new genes for old pathways. Curr Opin Plant Biol4:234–240

Chappell J (1995) Biochemistry and molecular biology ofthe isoprenoid biosynthetic pathway in plants. AnnRev Plant Physiol Mol Biol 46:521–547

Chapple C (1998) Molecular genetics analysis of plantcytochrome P450-dependent monooxygenases. AnnRev Plant Physiol Mol Biol 49:311–343

Chiu W, Baker ML, Jiang W, Dougherty M, Schmid MF(2005) Electron cryomicroscopy of biological machinesat subnanometer resolution. Structure 13:363–372

Chuong SD, Good AG, Taylor GJ, Freeman MC, Moor-head GB, Muench DG (2004) Large-scale identifica-tion of tubulin-binding proteins provides insight onsubcellular trafficking, metabolic channeling, and sig-naling in plant cells. Mol Cell Proteomics 3:970–983

Czichi U, Kindl H (1975) Formation of p-coumaric acid ando-coumaric acid from L-phenylalanine by microsomalmembrane fractions from potato: evidence of mem-brane-bound enzyme complexes. Planta 125:115–125

Czichi U, Kindl H (1977) Phenylalanine ammonia lyaseand cinamic acid hydroxylases as assembled consec-utive enzymes on microsomal membranes of cucum-ber cotyledons: cooperation and subcellulardistribution. Planta 134:133–143

Davydov DR, Kariakin AA, Petushkova NA, Peterson JA(2000) Association of cytochromes P450 with theirreductases: opposite sign of the electrostatic interac-tions in P450BM-3 as compared with the microsomal2B4 system. Biochemistry 39:6489–6497

Denisov IG, Makris TM, Sligar SG, Schlichting I (2005)Structure and chemistry of cytochrome P450. ChemRev 105:2253–2277

Dhillon DS, Brown SA (1976) Localization, purification,and characterization of dimethylallylpyrophosphate:umbelliferone dimethylallythransferase from Rutagraveolens. Arch Biochem Biophys 177:74–83

Dixon RA, Achnine L, Kota P, Liu C-J, Reddy MSS,Wang LJ (2002) The phenylpropanoid pathway andplant defense – a genomics perspective. Mol PlantPathol 3:371–390

Dixon RA, Chen F, Guo D, Parvathi K (2001) The bio-synthesis of monolignols: a ‘‘metabolic grid’’, orindependent pathways to guaiacyl and syringyl units?Phytochemistry 57:1069–1084

Estabrook RW, Hildebrandt AG, Baron J, Netter KJ,Leibman K (1971) A new spectral intermediate asso-ciated with cytochrome P-450 function in liver micro-somes. Biochem Biophys Res Commun 42:132–139

Ferrer JL, Jez JM, Bowman ME, Dixon RA, Noel JP(1999) Structure of chalcone synthase and themolecular basis of plant polyketide biosynthesis. NatStruct Biol 6:775–784

Galili G, Sengupta-Gopalan C, Ceriotti A (1998) Theendoplasmic reticulum of plant cells and its role inprotein maturation and biogenesis of oil bodies. PlantMol Biol 38:1–29

Graciet E, Lebreton S, Gontero B (2004) Emergence ofnew regulatory mechanisms in the Benson–Calvinpathway via protein–protein interactions: a glyceral-dehyde-3-phosphate dehydrogenase/CP12/phospho-ribulokinase complex. J Exp Bot 55:1245–1254

Hrazdina G, Wagner GJ (1985) Metabolic pathways asenzyme complexes: evidence for the synthesis ofphenylpropanoids and flavonoids on membrane asso-ciated enzyme complexes. Arch Biochem Biophys237:88–100

Hrazdina G, Zobel AM, Hoch HC (1987) Biochemical,immunological, and immunocytochemical evidencefor the association of chalcone synthase with endo-plasmic reticulum membranes. Proc Natl Acad SciUSA 84:8966–8970

Hubbard PA, Shen AL, Paschke R, Kasper CB, Kim JJ(2001) NADPH-cytochrome P450 oxidoreductase:structural basis for hydride and electron transfer.J Biol Chem 276:29,163–29,170

Humphreys JM, Chapple C (2002) Rewriting the ligninroadmap. Curr Opin Plant Biol 5:224–229

Ingelman-Sundberg M, Blanck J, Smettan G, Ruckpaul K(1983) Reduction of cytochrome P-450 LM2 byNADPH in reconstituted phospholipid vesicles isdependent on membrane charge. Eur J Biochem134:157–162

Ingelman-Sundberg M, Haaparanta T, Rydstrom J (1981)Membrane charge as effector of cytochromeP-450LM2 catalyzed reactions in reconstituted lipo-somes. Biochemistry 20:4100–4106

Janovjak H, Kedrov A, Cisneros DA, Sapra KT, Struck-meier J, Muller DJ (2005) Imaging and detectingmolecular interactions of single transmembrane pro-teins. Neurobiol Aging 88:1423–1428

Jez JM, Bowman ME, Dixon RA, Noel JP (2000) Struc-ture and mechanism of the evolutionarily unique plantenzyme chalcone isomerase. Nat Struct Biol 7:786–791

Jez JM, Bowman ME, Noel JP (2002) Role of hydrogenbonds in the reaction mechanism of chalcone isom-erase. Biochemistry 41:5168–5176

Jorgensen K, Rasmussen AV, Morant M, Nielsen AH,Bjarnholt N, Zagrobelny M, Bak S, Moller BL (2005)Metabolon formation and metabolic channeling in thebiosynthesis of plant natural products. Curr OpinPlant Biol 8:280–291

Kao YY, Harding SA, Tsai CJ (2002) Differentialexpression of two distinct phenylalanine ammonia-lyase genes in condensed tannin-accumulating andlignifying cells of quaking aspen. Plant Physiol130:756–760

Koopmann E, Hahlbrock K (1997) Differentiallyregulated NADPH: cytochrome p450 oxidoreducta-ses in parsely. Proc Natl Acad Sci USA 94:14,954–14,959

Kristensen C, Morant M, Olsen CE, Ekstrom CT, Gal-braith DW, Moller BL, Bak S (2005) Metabolicengineering of dhurrin in transgenic Arabidopsisplants with marginal inadvertent effects on the me-tabolome and transcriptome. Proc Natl Acad Sci USA102:1779–1784

470 Phytochem Rev (2006) 5:459–472

123

Liu CJ, Blount JW, Steele CL, Dixon RA (2002) Bottle-necks for metabolic engineering of isoflavone glyco-conjugates in Arabidopsis. Proc Natl Acad Sci USA99:14,578–14,583

Liu CJ, Dixon RA (2001) Elicitor-induced association ofisoflavone O-methyltransferase with endomembranesprevents the formation and 7-O-methylation ofdaidzein during isoflavonoid phytoalexin biosynthesis.Plant Cell 13:2643–2658

Magee T, Pirinen N, Adler J, Pagakis SN, Parmryd I(2002) Lipid rafts: cell surface platforms for T cellsignaling. Biol Res 35:127–131

Memelink J (2004) Putting the opium in poppy to sleep.Nat Biotechnol 22:1526–1527

Mendes P, Douglas KB, Welch GR (1995). Metabolicchannelling in organized enzyme systems: experimentsand models. In: Brindle KM (ed) Enzymology in vivo,vol 11. JAI Press Inc., Greenwich, CN, pp 1–19

Miles EW (2001) Tryptophan synthase: a multienzymecomplex with an intramolecular tunnel. Chem Rec1:140–151

Moller BL, Conn EE (1980) The biosynthesis of cyano-genic glucosides in higher plants. Channeling ofintermediates in dhurrin biosynthesis by a microsomalsystem from Sorghum bicolor (linn) Moench. J BiolChem 255:3049–3056

Morant M, Bak S, Moller BL, Werck-Reichhart D (2003)Plant cytochromes P450: tools for pharmacology,plant protection and phytoremediation. Curr OpinBiotechnol 14:151–162

Munro AW, Daff S, Coggins JR, Lindsay JG, ChapmanSK (1996) Probing electron transfer in flavocyto-chrome P-450 BM3 and its component domains. Eur JBiochem 239:403–409

Munro AW, Leys DG, McLean KJ, Marshall KR, Ost TW,Daff S, Miles CS, Chapman SK, Lysek DA, MoserCC, Page CC, Dutton PL (2002) P450 BM3: the verymodel of a modern flavocytochrome. Trends BiochemSci 27:250–257

Murataliev MB, Feyereisen R (2000) Functional interac-tions in cytochrome P450BM3. Evidence thatNADP(H) binding controls redox potentials of theflavin cofactors. Biochemistry 39:12,699–12,707

Murataliev MB, Feyereisen R, Walker FA (2004) Electrontransfer by diflavin reductases. Biochim Biophys Acta1698:1–26

Nadler SG, Strobel HW (1991) Identification and charac-terization of an NADPH-cytochrome P450 reductasederived peptide involved in binding to cytochromeP450. Arch Biochem Biophys 290:277–284

Nielsen KA, Moller BL (2005) Cytochrome P450 in plants.In: Ortiz de Montellano PR (ed) Cytochrome P450:structure, mechanism, and biochemistry. KluwerAcademic/Plenum Publishers, New York, pp 553–583

Paine MJ, Scrutton NS, Munro AW, Gutierrez A, RobertsGC, Wolf CR (2005) Electron transfer partners ofcytochrome P450. In: Ortiz de Montellano PR (ed)Cytochrome P450: structure, mechanism, and bio-chemistry. Kluwer Academic/Plenum Publishers, NewYork, pp 115–148

Panicot M, Minguet EG, Ferrando A, Alcazar R, BlazquezMA, Carbonell J, Altabella T, Koncz C, Tiburcio AF(2002) A polyamine metabolon involving aminopro-pyl transferase complexes in Arabidopsis. Plant Cell14:2539–2551

Peterson JA, Ebel RE, O’Keeffe DH, Matsubara T, Es-tabrook RW (1976) Temperature dependence ofcytochrome P-450 reduction. A model for NADPH-cytochrome P-450 reductase: cytochrome P-450interaction. J Biol Chem 251:4010–4016

Ralston L, Subramanian S, Matsuno M, Yu O (2005)Partial reconstruction of flavonoid and isoflavonoidbiosynthesis in yeast using soybean Type I andType II chalcone isomerases. Plant Physiol 137:1375–1388

Rasmussen S, Dixon RA (1999) Transgene-mediated andelicitor-induced perturbation of metabolic channelingat the entry point into the phenylpropanoid pathway.Plant Cell 11:1537–1552

Reumann S (2000) The structural properties of plantperoxisomes and their metabolic significance. BiolChem 381:639–648

Ro DK, Douglas CJ (2004) Reconstitution of the entrypoint of plant phenylpropanoid metabolism in yeast(Saccharomyces cerevisiae): implications for controlof metabolic flux into the phenylpropanoid pathway.J Biol Chem 279:2600–2607

Russell DW, Conn EE (1967) The cinnamic acid 4-hydroxylase of pea seedlings. Arch Biochem Biophys122:256–258

Saslowsky D, Winkel-Shirley B (2001) Localization offlavonoid enzymes in Arabidopsis roots. Plant J 27:37–48

Saslowsky DE, Warek U, Winkel BS (2005) Nuclearlocalization of flavonoid enzymes in Arabidopsis.J Biol Chem 280:23,735–23,740

Schoch GA, Attias R, Le Ret M, Werck-Reichhart D(2003) Key substrate recognition residues in the activesite of a plant cytochrome P450, CYP73A1. Homol-ogy guided site-directed mutagenesis. Eur J Biochem270:3684–3695

Sevrioukova IF, Li H, Zhang H, Peterson JA, Poulos TL(1999) Structure of a cytochrome P450-redox partnerelectron-transfer complex. Proc Natl Acad Sci USA96:1863–1868

Shen S, Strobel HW (1993) Role of lysine and arginineresidues of cytochrome P450 in the interactionbetween cytochrome P4502B1 and NADPH-cyto-chrome P450 reductase. Arch Biochem Biophys304:257–265

Shen S, Strobel HW (1995) Functional assessment ofspecific amino acid residues of cytochrome P4501A1using anti-peptide antibodies. Arch Biochem Biophys320:162–169

Shimada N, Aoki T, Sato S, Nakamura Y, Tabata S, AyabeS (2003) A cluster of genes encodes the two types ofchalcone isomerase involved in the biosynthesis ofgeneral flavonoids and legume-specific 5-deoxy(iso)flavonoids in Lotus japonicus. Plant Physiol131:941–951

Phytochem Rev (2006) 5:459–472 471

123

Shimizu T, Tateishi T, Hatano M, Fujii-Kuriyama Y (1991)Probing the role of lysines and arginines in the cata-lytic function of cytochrome P450d by site-directedmutagenesis. Interaction with NADPH-cytochromeP450 reductase. J Biol Chem 266:3372–3375

Srere PA (1985) The metabolon. Trends Biochem Sci10:109–110

Srere PA (1987) Complexes of sequential metabolic en-zymes. Annu Rev Biochem 56:89–124

Srere PA (2000) Macromolecular interactions: tracing theroots. Trends Biochem Sci 25:150–153

Subramaniam R, Desveaux D, Spickler C, Michnick SW,Brisson N (2001) Direct visualization of proteininteractions in plant cells. Nat Biotechnol 19:769–772

Thaminy S, Miller J, Stagljar I (2004) The split-ubiquitinmembrane-based yeast two-hybrid system. MethodsMol Biol 261:297–312

Urban P, Mignotte C, Kazmaier M, Delorme F, Pompon D(1997) Cloning, yeast expression, and characterizationof the coupling of two distantly related Arabidopsisthaliana NADPH-cytochrome P450 reductases withP450 CYP73A5. J Biol Chem 272:19,176–19,186

Venkateswarlu K, Lamb DC, Kelly DE, Manning NJ,Kelly SL (1998) The N-terminal membrane domain ofyeast NADPH-cytochrome P450 (CYP) oxidoreduc-tase is not required for catalytic activity in sterolbiosynthesis or in reconstitution of CYP activity.J Biol Chem 273:4492–4496

Wallrabe H, Periasamy A (2005) Imaging protein mole-cules using FRET and FLIM microscopy. Curr OpinBiotechnol 16:19–27

Wang M, Roberts DL, Paschke R, Shea TM, Masters BS,Kim JJ (1997) Three-dimensional structure ofNADPH-cytochrome P450 reductase: prototype forFMN- and FAD-containing enzymes. Proc Natl AcadSci USA 94:8411–8416

Wanner LA, Li G, Ware D, Somssich IE, Davis KR (1995)The phenylalanine ammonia-lyase gene family inArabidopsis thaliana. Plant Mol Biol 27:327–338

Weisshaar B, Jenkins GI (1998) Phenylpropanoid biosyn-thesis and its regulation. Curr Opin Plant Biol 1:251–257

Werck-Reichhart D, Feyereisen R (2000) CytochromesP450: a success story. Genome Biol 1:3003.1–3003.9

Winkel-Shirley B (2001a) Flavonoid biosynthesis. A col-orful model for genetics, biochemistry, cell biology,and biotechnology. Plant Physiol 126:485–493

Winkel-Shirley B (2001b) It takes a garden. How work ondiverse plant species has contributed to an under-standing of flavonoid metabolism. Plant Physiol127:1399–1404

Winkel BSJ (2004) Metabolic channeling in plants. AnnRev Plant Physiol Mol Biol 55:85–107

Yu O, Jung W, Shi J, Croes RA, Fader GM, McGonigle B,Odell JT (2000) Production of the isoflavones geni-stein and daidzein in non-legume dicot and monocottissues. Plant Physiol 124:781–794

Yu O, McGonigle B (2005) Metabolic engineering ofisoflavone biosynthesis. Adv Agron 86:147–190

Yu O, Shi J, Hession AO, Maxwell CA, McGonigle B,Odell JT (2003) Metabolic engineering to increaseisoflavone biosynthesis in soybean seed. Phytochem-istry 63:753–763

Zhao Q, Modi S, Smith G, Paine M, McDonagh PD, WolfCR, Tew D, Lian LY, Roberts GC, Driessen HP(1999) Crystal structure of the FMN-binding domainof human cytochrome P450 reductase at 1.93 A res-olution. Protein Sci 8:298–306

472 Phytochem Rev (2006) 5:459–472

123


Recommended