You are on page 1of 9

475

Process design for microbial plastic factories: metabolic


engineering of polyhydroxyalkanoates
Ilana S Aldor and Jay D Keaslingy

Implementing several metabolic engineering strategies, either varied applications as the manufacture of shampoo bot-
individually or in combination, it is possible to construct microbial tles, heat seal resins and heart valves, have made PHA
plastic factories to produce a variety of polyhydroxyalkanoate polymers prime candidates for commodity and speciality
(PHA) biopolymers with desirable structures and material commercial plastic production.
properties. Approaches include external substrate manipulation,
inhibitor addition, recombinant gene expression, host cell Besides the commercial applications of PHAs as bioplas-
genome manipulation and, most recently, protein engineering of tics that are biodegradable and made from renewable
PHA biosynthetic enzymes. In addition, mathematical models resources, from the standpoint of an academic meta-
and molecular methods can be used to elucidate metabolically bolic pathway engineer, PHAs are model compounds
engineered systems and to identify targets for performance for metabolic engineering. Several bio-based polyesters,
improvement. including polylactic acid and 3GT (a polymer of 1,3-
propanediol and terephthalic acid), can have some or all
Addresses of their monomeric constituents produced by microbial
Department of Chemical Engineering, 201 Gilman Hall, University of fermentation. However, unlike these bio-based poly-
California, Berkeley, 94720-1462, USA mers which are chemically synthesized, PHAs and their

e-mail: aldor@gene.com
y
e-mail: keasling@socrates.berkeley.edu
unnatural polythioester analogs (described below) are
unique in that both polymer assembly and accumulation
occur in vivo and can be manipulated by metabolic
Current Opinion in Biotechnology 2003, 14:475–483 engineering. The monomeric composition of the biopo-
This review comes from a themed issue on
lymer depends on the host’s PHA synthase (polymerase)
Biochemical engineering and on the hydroxyacyl-CoA thioester precursors sup-
Edited by Jeremy S Edwards and Kenneth J Kauffman plied to the enzyme, which in turn depend on the meta-
bolic pathways operating in the cell and on the external
0958-1669/$ – see front matter
ß 2003 Elsevier Ltd. All rights reserved. carbon source. The biosynthetic routes to PHA mono-
mers compete with and/or rely on important pathways
DOI 10.1016/j.copbio.2003.09.002 such as the tricarboxylic acid (TCA) cycle, fatty acid
degradation (b-oxidation) and fatty acid biosynthesis
Abbreviations for precursors, and involve central metabolites such as
3HB 3-hydroxybutyrate acetyl-CoA and cofactors such as NADPH (Figure 1).
3HHX 3-hydroxyhexanoate Ultimately, the monomeric composition of the PHA
3HO 3-hydroxyoctanoate biopolymer provides the metabolic engineer with con-
3HV 3-hydroxyvalerate
3MB 3-mercaptobutyrate
siderable insight into the metabolism that was involved
3MP 3-mercaptopropionate in polyester biosynthesis.
PHA polyhydroxyalkanoate
PHB poly(3-hydroxybutyrate) The primary aims of the metabolic engineering of PHAs
P(3HB-co-3HHX) poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)
include controlling different factors that determine poly-
P(3HB-co-3HV) poly(3-hydroxybutyrate-co-3-hydroxyvalerate)
mer material properties, such as monomeric composition,
chain length and copolymer microstructure, as well as
Introduction optimizing yield. Pathway engineering for PHA produc-
Polyhydroxyalkanoates (PHAs) form a class of natural tion offers the opportunity to synthesize novel polymers
polyesters that many organisms in the environment accu- with desirable properties in low-cost, high-productivity
mulate in the form of intracellular granules to store carbon fermentations.
and reducing equivalents. Although the most well-stu-
died PHA is poly(3-hydroxybutyrate) (PHB), a polymer In the history of metabolic engineering, PHAs were
of 3-hydroxybutyrate (3HB), there are over 140 possible among the first target compounds and there is substantial
constituent monomers [1], which have been traditionally literature on this topic including some excellent recent
classified as short chain length (C4 and C5) and medium reviews [2–4,5,6]. Plants may be the most economical
chain length ( C6) hydroxyalkanoates. This wide vari- and environmentally friendly hosts for mass production of
ety of monomers yields PHAs with diverse material biodegradable, commodity plastic [7]. The metabolic
properties that depend on polymer composition. These engineering of PHAs in planta was recently reviewed
material properties, which have proven useful in such [5]. However, microbial hosts provide many more

www.current-opinion.com Current Opinion in Biotechnology 2003, 14:475–483


476 Biochemical engineering

Figure 1

GTP
NADH Succinyl-CoA

Succinate
α-Ketoglutarate FADH

NADH
Citric acid Fumarate
Isocitrate cycle

Malate Carbohydrates

Citrate
NADH
Oxaloacetate

Fatty acids
Acetyl-CoA ATP

Acyl-ACP Acyl-CoA
PhaA
NADPH FADH

Fatty acid
trans-2- 3-Keto- Acetoacetyl-CoA 3-Keto- trans-2-
de novo
Enoyl-ACP acyl-ACP acyl-CoA Enoyl-CoA
synthesis
NADPH

PhaB
NADPH
NADH
(R)-3-Hydroxy- (S)-3-Hydroxy-
acyl-ACP acyl-CoA
PhaJ
PhaG
(R)-3-Hydroxyacyl-CoA

PhaC

Polyhydroxyalkanoate
Current Opinion in Biotechnology

PHA biosynthesis in the context of microbial metabolism. The major enzymes involved in PHA biosynthesis are in red. Abbreviations: PhaA,
3-ketothiolase; PhaB, (R)-3-ketoacyl-CoA reductase (for PHB biosynthesis, this enzyme is acetoacetyl-CoA reductase); PhaC, PHA synthase or
polymerase; PhaG, (R)-3-hydroxyacyl ACP:CoA transacylase; PhaJ, (R)-specific enoyl-CoA hydratase. Dotted lines represent reactions where
intermediate metabolic steps are not included. PhaC is specific for enantiomeric monomers in the (R) configuration.

possibilities for precisely engineering the monomeric (Figure 2). Illustrative examples from the recent litera-
composition and the properties of PHAs; the controlled ture are described.
environment of a bioreactor, as compared with an outdoor
farm field, the ease of microbial genetic manipulation, External substrate manipulation
and the option to use external carbons sources other than The simplest metabolic engineering strategy is to manip-
carbon dioxide are clear advantages. The focus of the ulate the carbon source(s) supplied to the host to control
present article is PHA production in bacteria. and direct carbon flux through the precursor and polymer
biosynthesis enzymes. This strategy has traditionally
Strategies for the metabolic engineering of been exploited to modulate polymer composition by
bacteria for PHA production varying the feed ratio of different substrate precursors.
To benefit from the advantages of metabolically engi-
neered microbes for PHA production, several strategies Recently, there have been novel applications of this
can be implemented either individually or in combination strategy to generate unusual sulfur-containing polymers.

Current Opinion in Biotechnology 2003, 14:475–483 www.current-opinion.com


Metabolic engineering of polyhydroxyalkanoates Aldor and Keasling 477

Figure 2

Evolved protein
Substrate precursor Inhibitor addition
addition

Heterologous gene Host cell genome


expression manipulation
Current Opinion in Biotechnology

Schematic of various strategies for the metabolic engineering of PHAs. These approaches can be implemented individually or in combination.

In one instance, recombinant Ralstonia eutropha was fed For example, acrylate was used to inhibit b-oxidation in
alkylthioalkanoates (thia fatty acids) to produce poly(3- wild-type R. eutropha during growth on octanoate such that
hydroxy-S-propyl-o-thioalkanoate) copolymers contain- the bacteria accumulated a copolymer of 3-hydroxypropio-
ing thioether linkages in their sidechains [8]. In another nate, 3HB, 3-hydroxyhexanoate and 3-hydroxyoctanoate
creative example of substrate feeding, previously (P(3HP-co-3HB-co-3HHX-co-3HO)) containing both short
unknown microbial polythioesters (which have C(O)S and medium chain length monomers [13]. Normally, this
bonds in contrast to the C(O)O bonds of analogous organism accumulates PHAs with only short chain length
PHA polyoxoesters), were incorporated into a copolymer monomers.
by cultivating wild-type R. eutropha in medium containing
gluconate plus, for example, 3-mercaptopropionate Recombinant gene expression
(3MP) [9] or 3-mercaptobutyrate (3MB) [10]. The organ- Key genes and hosts for recombinant PHA biosynthesis
ism accumulated the polythioester-polyoxoester copoly- The main PHA biosynthetic enzymes are shown in
mers P(3MP-co-3HB) and P(3MB-co-3HB), respectively, Figure 1 and the corresponding genes can be expressed
with sulfur in their backbones and a nonrandom micro- in recombinant microbes for the metabolic engineering
structure [9,11]. of PHAs. The most common microbes used as heterolog-
ous hosts and in fermentation development include R.
In another clever example of external substrate manip- eutropha, Pseudomonas putida, Pseudomonas oleovorans and
ulation, block copolymers of PHB and poly(3-hydroxy- Escherichia coli [14].
butyrate-co-3-hydroxyvalerate) (P(3HB-co-3HV)) were
generated in wild-type R. eutropha by intermittent addi- E. coli, which lacks a native PHA synthase and enzymes
tion of valerate (precursor substrate of 3HV) to cultiva- that mediate the formation of various hydroxyacyl-CoA
tion medium containing an excess of fructose [12]. Core precursors with high specificity, deserves special mention
and shell (diblock copolymer) or multilayered granules because of the organism’s numerous advantages as a host
accumulated. (summarized elsewhere [15,16]). This microbe has well-
understood genetics and biochemistry and can be easily
Inhibitor addition manipulated for the biosynthesis of PHAs and for high-
Connected with the idea of a feeding strategy for external productivity fermentation. In E. coli, PHA synthesis may
substrates, is the addition of inhibitors that channel pre- be induced at an optimized point in the process and is not
cursors to PHA synthesis by inhibiting competing and tied to natural regulation, which often involves induction
undesirable pathways or that limit chain elongation. Such by nutrient limitation. E. coli also does not harbor native
compounds have proven particularly useful for incorpor- machinery for polymer degradation, unlike organisms that
ating medium chain length monomers derived from b- can retrieve the stored carbon and reducing equivalents.
oxidation into PHAs. Finally, E. coli cells are relatively weak, which is an

www.current-opinion.com Current Opinion in Biotechnology 2003, 14:475–483


478 Biochemical engineering

advantage for recovering recombinant granules that tend phaBCA from Acinetobacter was co-expressed (Figure 3a).
to be large in this microbe. This proof-of-concept study showed how it is possible to
use a novel pathway to generate the copolymer from a
Gene dosage simple carbon source without the need for external pro-
One approach to metabolically engineer PHA synthesis pionate, a costly chemical.
involves increasing the copy number of relevant genes,
often genes that are identical or homologous to those In another recent example of producing a PHA copolymer
already expressed on the host’s chromosome, to augment from an unrelated carbon source, P(3HB-co-3HHX) was
PHA content or to modify polymer composition. Histori- made from fructose in recombinant R. eutropha harboring
cally, this strategy has had mixed success [3]. the crotonyl-CoA reductase gene (crr) from Streptomyces
cinnamonensis and phaCJ from A. punctata [22]. This work
Fukui et al. [17] found that Aeromonas punctata (formerly also took advantage of several native host enzyme activ-
known as Aeromonas caviae) harboring additional copies of ities. The pathway functioned sufficiently to incorporate
its own gene cluster phaPCJ (encoding an uncharacter- 1.5 mol% 3HHX in the copolymer, resulting in noticeable
ized, granule-associated ‘phasin’, PHA synthase and improvement of its material properties.
enoyl-CoA hydratase, respectively), accumulated P(3HB-
co-3HHX) at higher levels and with an increased 3HHX In another example of an engineered multistep pathway,
fraction than the wild-type strain. Another group found butyrate kinase (Buk) and phosphotransbutyrylase (Ptb)
that R. eutropha harboring additional phaC copies accu- from Clostridium acetobutylicum and PHA synthase from
mulated significantly more 3HV in P(3HB-co-3HV) than Thiocapsa pfennigii were recently applied to the innovative
the parent strain, but total copolymer levels were un- production of homopolythioesters in E. coli (Figure 3b)
changed [18]. [23]. 3-Mercaptoalkanoates were supplied in a polymer
production phase that followed the glucose-fed growth
A similar strategy was attempted by expressing R. eutro- phase of the fermentation. P(3MP), P(3MB) or P(3MV)
pha PHA synthesis genes (phaCAB) in a Synechocystis strain were formed depending on the 3-mercaptoalkanoate
that could naturally accumulate PHAs when grown on feed, and these polymers have unique thermoplastic
carbon dioxide and various carbon sources [19]. Only a properties compared with PHAs and petrochemical-
marginal change in PHA accumulation was observed, derived polymers. Such homopolythioesters cannot be
although synthase activity increased twofold. Clearly, produced in a natural PHA accumulator like R. eutropha,
the effectiveness of a strategy involving increased gene because in this organism the PHB precursor also accu-
dosage depends on the limiting factor for polymer synth- mulates and polythioester-polyoxoester copolymers are
esis in the individual case. formed (as described above).

Introduction of heterologous precursor production Choosing the source of genes


pathways With ongoing genome sequencing projects, newly iden-
Metabolic engineers use heterologous precursor produc- tified genes continuously become available. It is common
tion pathways to facilitate the synthesis of polymers that in PHA work to utilize homologous genes encoding the
would not naturally accumulate in the host of interest, or same enzyme, but with different activities or specificities,
possibly anywhere in nature, and which may have desir- for various metabolically engineered systems. Together
able structures and material properties. Another major with the use of diverse, heterologous pathways for the
goal is to develop novel pathways for efficient precursor synthesis of precursors, this facilitates a combinatorial
production using unrelated, simple and inexpensive car- biosynthetic approach to PHA metabolic engineering.
bon sources, which may also facilitate fast growth to a high
cell density. As PHA synthase specificity is a crucial parameter for
determining polymer composition, in the example given
Historically, the random copolymer P(3HB-co-3HV) has above for producing polythioesters, the heterodimeric
been the most extensively studied and was sold com- ‘type III’ PhaEC from T. pfennigii was used because of
mercially under the tradename BiopolTM. Production its broad substrate range [24]. It is also becoming clear
traditionally involved feeding propionate to produce that certain enoyl-CoA hydratases are more useful than
intracellular propionyl-CoA, the precursor of 3HV [20]. others for transferring medium chain length precursors
We metabolically engineered a pathway for the formation from b-oxidation to PHA synthase, owing to the enantio-
of propionyl-CoA from succinyl-CoA, derived from the selectivity and chain length specificity of various enoyl-
TCA cycle, in recombinant Salmonella enterica serovar CoA hydratases [25].
Typhimurium using the recently discovered E. coli
enzymes, sleeping beauty mutase (Sbm) and (R)-methyl- Bioprospecting for novel genes and pathways
malonyl-CoA decarboxylase (YgfG) [21]. This facilitated Metabolic engineers can take advantage of natural bio-
the production of P(3HB-co-3HV) from glycerol when diversity and bioprospect among organisms, including

Current Opinion in Biotechnology 2003, 14:475–483 www.current-opinion.com


Metabolic engineering of polyhydroxyalkanoates Aldor and Keasling 479

Figure 3

(a)
O O
(b)

O O
x y O
P(3HB-co-3HV)
Glycerol
HS OH
5
3MV
3-Hydroxy- 3-Hydroxy- PEP ATP
valeryl-CoA butyryl-CoA Butyrate kinase
(Buk) ADP
4 Pyruvate
3MV-Pi
3-Keto- Aceto- Phospho- CoA
valeryl-CoA acetyl-CoA transbutyrylase
3 (Ptb) Pi
Acetyl-CoA 3MV-CoA
6
2-Methylcitrate
PHA synthase
Citrate Oxaloacetate (PhaEC)
Propionyl-CoA CoA
Malate 2-Methyl-
Isocitrate Citric
citric 2-Methyl-cis-
acid O
CO2 acid aconitate
cycle Fumarate
2-Ketoglutarate cycle
Succinate S
2 2-Methyl-
Succinyl-CoA isocitrate x
(R)-Methyl- P(3MV)
Pyruvate
malonyl-CoA 1

Current Opinion in Biotechnology

Examples of recently engineered pathways using heterologous precursor production pathways for the biosynthesis of PHA and related biopolymers.
(a) Production of P(3HB-co-3HV) from an unrelated carbon source. The key precursor of 3HV, propionyl-CoA, is synthesized from succinyl-CoA
derived from the TCA cycle. Enzymes are numbered: (1) sleeping beauty mutase (Sbm); (2) (R)-methylmalonyl-CoA decarboxylase (YgfG); (3) 3-
ketothiolase (PhaA); (4) acetoacetyl-CoA reductase (PhaB); (5) PHA synthase (PhaC); (6) 2-methylcitrate synthase (PrpC). The gene encoding PrpC was
deleted from the chromosome of the S. enterica host to channel propionyl-CoA to 3HV by preventing propionyl-CoA catabolism via the 2-methylcitric
acid cycle. Dotted lines represent reactions where intermediate metabolic steps are not included. (b) Production of an unnatural polythioester P(3MV),
a sulfur analog to the polyhydroxyalkanoate (polyoxoester) P(3HV), in recombinant E. coli.

uncultivable species, in a variety of environments to may also reveal useful genes for polymer production. For
find PHA genes and pathways suitable for different example, upstream of phaC (encoding PHA synthase) on
applications. the Aeromonas genome, orf1 (phaP) was discovered, a gene
which has proved useful for the production of P(3HB-co-
For example, a couple of studies used genes from Aero- 3HHX) with a high 3HHX fraction [27] and facilitated the
monas species isolated from raw sewage for recombinant incorporation of 3HV in the terpolymer, P(3HB-co-3HV-
PHA production in E. coli [26,27]. Furthermore, techniques co-3HHX) [26].
like degenerate PCR and screening procedures such as
restriction fragment length polymorphism (RFLP) permit An interesting recent example of the elucidation of a novel
the isolation of novel, PHA-related genes from community pathway from an environmental organism involved the use
DNA extracted from various environments (S Yilmaz, IS of 13C labeling and in vivo nuclear magnetic resonance
Aldor, KD McMahon, JD Keasling and D Jenkins, unpub- (NMR) to describe propionate metabolism by uncultiva-
lished data), facilitating the discovery of PHA synthesis ble, phosphorus-accumulating organisms from activated
genes that are homologous to known sequences. sludge that performed enhanced biological phosphorus
removal [28]. The microbes accumulate a copolymer of
Exploration of the sequence adjacent to well-character- 3HB, 3HV, 3-hydroxy-2-methylbutyrate and 3-hydroxy-2-
ized open reading frames involved in PHA biosynthesis methylvalerate (P(3HB-co-3HV-co-3H2MB-co-3H2MV)),

www.current-opinion.com Current Opinion in Biotechnology 2003, 14:475–483


480 Biochemical engineering

which is not found anywhere else in nature and whose In the case of A. punctata PHA synthase, applying single
synthesis would be an interesting pathway to reconstitute gene shuffling to a targeted region and screening for
in a heterologous host. higher PHB-accumulators yielded two mutants that
demonstrated increased activity towards 3HB-CoA and
Host cell genome manipulation led to enhanced accumulation of PHB and P(3HB-co-
Host cell genome manipulation can eliminate competing 3HHX) [40]. In addition, both mutants incorporated a
pathways or modify native regulation for improved func- markedly increased 3HHX fraction compared with wild
tion of desirable pathways. This strategy was involved in type. In a separate study, A. punctata PHA synthase was
the study described above for the production of P(3HB- elegantly engineered in vivo using the mutator strain
co-3HV) using Sbm and YgfG. The host was a prpC strain E. coli XL1-Red, and mutants were again screened for
of S. enterica, in which 2-methylcitrate synthase was promoting enhanced PHB accumulation in recombinant
deleted to prevent catabolism of propionyl-CoA via the E. coli [41]. Overproducing mutants with higher in vitro
2-methylcitric acid cycle and to increase the pool of this specific activity (up to fivefold) and yielding higher PHA
precursor available to 3-ketothiolase [21] (Figure 3a). content (up to 126% of wild type) were isolated, although
in vitro and in vivo activity did not directly correlate. In
In addition, in a separate study, we utilized a mutant S. this work, mutants synthesized PHAs with increased
enterica prpE, ackA and acs host, which was unable to weight average molecular mass, but in contrast to the
activate propionate to propionyl-CoA [29]. We recon- previous study the 3HHX fraction was only slightly
stituted prpE (encoding propionyl-CoA synthetase) different from wild-type composition.
expression under the control of an isopropyl-b-D-thioga-
lactopyranoside (IPTG)-inducible promoter, indepen- In the latest example of PHA synthase engineering,
dent from the promoter expressing the Acinetobacter Pseudomonas sp. 61-3 PHA synthase was engineered to
PHA synthesis operon. Thus, by eliminating native pro- enhance PHB accumulation in recombinant E. coli [42].
pionate activation activity in the host, we could modulate This polymerase, which synthesizes copolymers consist-
the composition of the P(3HB-co-3HV) copolymer at a ing of C4 to C12 units, was targeted for improved PHB
fixed, external propionate concentration by varying IPTG accumulation because it possesses low in vitro substrate
addition, creating a ‘dial-a-composition’ system that specificity towards 3HB-CoA compared to precursors
facilitated adjustment of material properties according with longer sidechains. Error-prone PCR generated sin-
to inducer level. gle mutants that indicated two positions where mutations
would lead to increased PHB accumulation. The amino
Protein engineering of PHA biosynthetic enzymes acids at these respective locations were optimized using
Protein engineering via mutagenesis and molecular evo- saturation mutagenesis, in which all possible substitu-
lution allows metabolic engineers to optimize enzyme tions at the selected sites were tested. A dramatic
performance to obtain desirable polymer properties and increase in PHB content of up to 400-fold was achieved
yield. The recent target of this strategy has been PHA by generating double mutants with optimal residues at
synthase, the key enzyme that catalyzes the polymeriza- the two positions.
tion of hydroxyacyl-CoA thioesters, provided by precursor
pathways, into water-insoluble, PHA granules. A full Protein engineering for metabolic engineering of PHAs
structural and mechanistic characterization of this protein has a promising future. All the examples above utilized
from different microbes is ongoing [30–37]. single-gene shuffling techniques. However, given the
large number of cloned PHA synthase genes [15], it will
In general, the use of molecular evolution for enhanced be interesting to apply family gene shuffling to synthase
PHA production has lagged behind the utilization of this engineering as well [43]. Furthermore, we expect other
technique for the production of other metabolic engineer- PHA enzymes to be targets in the near future. Two
ing targets, such as carotenoids, because of the lack of a candidates include the recently characterized (R)-3-
good screening procedure for desirable mutations. The hydroxyacyl ACP:CoA transacylase (PhaG) from P. putida
screen used in the examples described below involves [44] and (R)-specific enoyl-CoA hydratase (PhaJ) from A.
Nile red, a lipophilic dye that imparts a pinkish stain to punctata [45] (see Update).
PHA-containing cells cultured on plates upon exposure to
ultraviolet light. This method selects for high PHA con- Analysis of metabolically engineered, PHA-
tent and traditionally has not been able to discriminate producing systems using mathematical
between polymers with different compositions [38]. A models and molecular techniques
recent report, however, suggests that, at least, it is pos- Once the above strategies have been implemented to
sible to distinguish between PHAs composed of short create microbial plastic factories, mathematical models,
chain length versus medium chain length monomers genetics, microarray analysis, proteomics and metabolo-
using spectrofluorometry with Nile red-stained colonies mics can be used to study the manipulated biological
suspended in water [39]. systems and suggest new metabolic engineering targets

Current Opinion in Biotechnology 2003, 14:475–483 www.current-opinion.com


Metabolic engineering of polyhydroxyalkanoates Aldor and Keasling 481

and approaches. PHA researchers are taking advantage of substrate specificity of (R)-enoyl-CoA hydratase from A.
the mathematical and molecular tools available for such punctata. Increased fractions of 3HO and 3-hydroxyde-
purposes. canoate (3HD) could be incorporated into the PHA
copolymer composed of short and medium chain length
Stoichiometric flux analysis of recombinant E. coli monomers, which was formed from dodecanoate in
recently predicted the importance of the Entner–Dou- recombinant E. coli expressing the mutant hydratase
doroff pathway in PHB production [46]. This was con- and PHA synthase from Pseudomonas sp. strain 61-3.
firmed experimentally [46] and was consistent with
results obtained from a proteomic analysis using two- In a second recent paper, a different strategy utilizing
dimensional polyacrylamide gel electrophoresis [47]. gene dosage and host cell genome manipulation ap-
Mathematical models have also been used to describe proaches was taken to modulate the composition of PHA
the dynamics of PHA copolymer structure and properties copolymer composed of medium chain length monomers
during its in vivo accumulation. A population balance for material property improvement [53]. Amplification of
model identified an optimal carbon source switching E. coli fadD and fadE, encoding acyl-CoA synthetase and
strategy for the production of block copolymers in the acyl-CoA dehydrogenase, respectively, in a fadA and/or
R. eutropha system described above [48,49]. fadB mutant of recombinant E. coli that harbored a PHA
synthase from Pseudomonas sp. strain 61-3 (different from
Also, a recent study utilizing genetics and enzyme activ- the synthase from this strain mentioned above), led to
ity assays provides a good example of how a metabolically enrichment of 3-hydroxyalkanoate monomers of the
engineered system can yield insight into native host same carbon number as the fatty acid carbon source.
metabolism and enzyme function [50]. The finding that
YfcX (a subunit of a putative fatty acid oxidation com- References and recommended reading
plex) is crucial for metabolic engineering of PHAs com- Papers of particular interest, published within the annual period of
review, have been highlighted as:
posed of medium chain length monomers from fatty
 of special interest
acids in E. coli fadB strains preceded work that described  of outstanding interest
the enzyme’s participation in a previously unknown,
1. Steinbuchel A, Valentin HE: Diversity of bacterial
anaerobic b-oxidation pathway and provided evidence polyhydroxyalkanoic acids. FEMS Microbiol Lett 1995,
for various aspects of enzyme function in the novel 128:219-228.
metabolism [51]. 2. Witholt B, Kessler B: Perspectives of medium chain length
poly(hydroxyalkanoates), a versatile set of bacterial
bioplastics. Curr Opin Biotechnol 1999, 10:279-285.
Conclusions
This review covers the recent, published work on PHA 3. Madison LL, Huisman GW: Metabolic engineering of
poly(3-hydroxyalkanoates): from DNA to plastic. Microbiol Mol
metabolic engineering involving both rational manipula- Biol Rev 1999, 63:21-53.
tion of pathways and random protein engineering tech- 4. Steinbuchel A: Perspectives for biotechnological production
niques; however, it is also noteworthy that industry and utilization of biopolymers: metabolic engineering of
continues to invest in PHAs. In the past, BiopolTM was polyhydroxyalkanoate biosynthesis pathways as a successful
example. Macromol Biosci 2001, 1:1-24.
produced by ICI, Zeneca and Monsanto. Metabolix
5. Snell KD, Peoples OP: Polyhydroxyalkanoate polymers and
recently acquired the BiopolTM patents from Monsanto  their production in transgenic plants. Metab Eng 2002,
and is producing several PHA polymers in bacteria and 4:29-40.
plants. Also, Proctor and Gamble developed PHA copo- A comprehensive review of the current status of the production of PHAs in
transgenic plants, a fascinating area not covered in this review. This
lymers of short and medium chain length monomers publication is from scientists in industry currently working on the meta-
under the tradename NodaxTM, and licensed the tech- bolic engineering of PHAs and contains interesting practical insights.
nology to the Kaneka Corporation (a Japanese manufac- 6. Steinbuchel A, Lutke-Eversloh T: Metabolic engineering and
turer of plastics and resins that is focusing on the  pathway construction for biotechnological production of
relevant polyhydroxyalkanoates in microorganisms.
production of P(3HB-co-3HHX)). Current, industrial pro- Biochem Eng J 2003, 3734:1-16.
duction systems extensively utilize metabolically engi- The latest review by academic leaders in the PHA field with interesting
perspectives on the metabolic engineering of specific PHAs. The focus is
neered bacteria to produce specialized polymers with on P(3HB-co-3HV), PHAs composed of medium chain length monomers,
specific, desirable properties. This attests to the value P(4HB) and P(4HV) homopolyesters, copolymers containing 4HB and
copolymers composed of short and medium chain length monomers.
of PHAs as commercially viable products and to the utility
of metabolic engineering strategies for their production. 7. Kurdikar D, Fournet L, Slater S, Paster M, Gruys KJ, Gerngross TU,
Coulon R: Greenhouse gas profile of a plastic material derived
We can expect that many more microbial plastic factories from a genetically modified plant. J Ind Ecol 2000, 4:107-122.
will be constructed in the near future. 8. Ewering C, Lutke-Eversloh T, Luftmann H, Steinbuchel A:
Identification of novel sulfur-containing bacterial polyesters:
Update biosynthesis of poly(3-hydroxy-S-propyl-x-thioalkanoates)
containing thioether linkages in the side chains. Microbiol 2002,
In recent work, Tsuge et al. [52] used information 148:1397-1406.
obtained from a crystallographic structure analysis with 9. Lutke-Eversloh T, Bergander K, Luftmann H, Steinbuchel A:
site-directed mutagenesis to engineer the chain length Identification of a new class of biopolymer: bacterial synthesis

www.current-opinion.com Current Opinion in Biotechnology 2003, 14:475–483


482 Biochemical engineering

of a sulfur-containing polymer with thioester linkages. recombinant R. eutropha. This is the first example of the synthesis of
Microbiol 2001, 147:11-19. 3HHX from an unrelated carbon source and further development of this
design will hopefully generate higher 3HHX fractions.
10. Lutke-Eversloh T, Bergander K, Luftmann H, Steinbuchel A:
Biosynthesis of poly(3-hydroxybutyrate-co-3- 23. Lutke-Eversloh T, Fischer A, Remminghorst U, Kawada J,
mercaptobutyrate) as a sulfur analogue to poly(3-  Marchessault RH, Bogershausen A, Kalwei M, Eckert H, Reichelt R,
hydroxybutyrate) (PHB). Biomacromolecules 2001, Liu S-J et al.: Biosynthesis of novel thermoplastic polythioesters
2:1061-1065. by engineered Escherichia coli. Nat Mater 2002, 1:236-240.
E. coli harboring genes for phosphotransbutyrylase (Ptb) and butyrate
11. Lutke-Eversloh T, Kawada J, Marchessault RH, Steinbuchel A: kinase (Buk) from C. acetobutylicum and type III PHA synthase (PhaEC)
Characterization of microbial polythioesters: physical from T. pfennigii converts 3-mercaptoalkanoates into polythioesters,
properties of novel copolymers synthesized by Ralstonia which are relatives of PHAs that have sulfur in their backbones. The
eutropha. Biomacromolecules 2002, 3:159-166. homopolythioesters formed in this work are the first to be synthesized in
12. Kelley AS, Mantzaris NV, Daoutidis P, Srienc F: Controlled bacteria, and the authors demonstrate that these polymers have unique
synthesis of polyhydroxyalkanoic (PHA) nanostructures in properties compared with PHAs and petrochemical-derived polymers.
R. eutropha. Nano Lett 2001, 1:481-485. PHA synthases can clearly have a surprising and fascinating lack of
specificity.
13. Green PR, Kemper J, Schechtman L, Guo L, Satkowski M,
 Fiedler S, Steinbuchel A, Rehm BHA: Formation of short chain 24. Liu SJ, Steinbuchel A: A novel genetically engineered pathway
length/medium chain length polyhydroxyalkanoate for synthesis of poly(hydroxyalkanoic acids) in Escherichia coli.
copolymers by fatty acid b-oxidation inhibited Ralstonia Appl Environ Microbiol 2000, 66:739-743.
eutropha. Biomacromolecules 2002, 3:208-213.
25. Tsuge T, Taguchi K, Taguchi S, Doi Y: Molecular characterization
A thought-provoking piece of work that challenges the idea that R.
and properties of (R)-specific enoyl-CoA hydratases from
eutropha PHA synthase is only capable of incorporating short chain
Pseudomonas aeruginosa: metabolic tools for synthesis of
length monomers into PHAs, a long-held belief that originated with in
polyhydroxyalkanoates via fatty acid b-oxidation. Int J Biol
vitro enzyme assays. Upon treatment with the fatty acid b-oxidation
Macromol 2003, 31:195-205.
inhibitor acrylate, during growth on octanoate, wild-type R. eutropha
accumulated P(3HP-co-3HB-co-3HHX-co-3HO). 26. Park SJ, Ahn WS, Green PR, Lee SY: Biosynthesis of
14. Lee SY, Choi J: Production of microbial polyester by poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-
fermentation of recombinant microorganisms. Biopolyesters. hydroxyhexanoate) by metabolically engineered Escherichia
Adv Biochem Eng Biotechnol, vol 71. Edited by Steinbuchel A, coli strains. Biotechnol Bioeng 2001, 74:81-86.
Babel W. Berlin: Springer-Verlag; 2001:185-207. 27. Park SJ, Ahn WS, Green PR, Lee SY: Production of
15. Steinbuchel A, Hein S: Biochemical and molecular basis of poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) by
microbial synthesis of polyhydroxyalkanoates in metabolically engineered Escherichia coli strains.
microorganisms. Biopolyesters. Adv Biochem Eng Biotechnol, vol Biomacromolecules 2001, 2:248-254.
71. Edited by Steinbuchel A, Babel W. Berlin: Springer-Verlag; 28. Lemos PC, Serafim LS, Santos MM, Reis MAM, Santos H:
2001:82-123.  Metabolic pathway for propionate utilization by
16. Lee SY: E. coli moves into the plastic age. Nat Biotechnol 1997, phosphorus-accumulating organisms in activated sludge:
13
15:17-18. C labeling and in vivo nuclear magnetic resonance.
Appl Environ Microbiol 2003, 69:241-251.
17. Fukui T, Kichise T, Iwata T, Doi Y: Characterization of 13 kDa A ground-breaking example of the use of 13C in vivo NMR to elucidate the
granule-associated protein in Aeromonas caviae and complex carbon metabolism in activated sludge, composed primarily of
biosynthesis of polyhydroxyalkanoate with altered molar uncultivable organisms, performing the wastewater treatment process
composition by recombinant bacteria. Biomacromolecules known as enhanced biological phosphorus removal (EBPR). An accurate
2001, 2:148-153. model for EBPR metabolism has been the subject of debate for many
years. This paper is a significant contribution towards understanding this
18. Choi JC, Shin HD, Lee YH: Modulation of 3-hydroxyvalerate complicated metabolism, including the associated PHA accumulation
molar fraction in poly(3-hydroxybutyrate-3-hydroxyvalerate) that involves a novel PHA copolymer and that has an important role in the
using Ralstonia eutropha transformant co-amplifying phbC treatment process.
and NADPH generation-related zwf genes. Enzyme Microb
Technol 2003, 32:178-185. 29. Aldor I, Keasling JD: Metabolic engineering of poly(3-
hydroxybutyrate-co-3-hydroxyvalerate) in recombinant
19. Sudesh K, Taguchi K, Doi Y: Effect of increased PHA synthase Salmonella enterica serovar Typhimurium. Biotechnol Bioeng
activity on polyhydroxyalkanoates biosynthesis in 2001, 76:108-114.
Synechocystis sp. PCC6803. Int J Biol Macromol 2002,
30:97-104. 30. Zhang S, Kolvek S, Lenz RW, Goodwin S: Mechanism of the
polymerization reaction initiated and catalyzed by the
20. Byrom D: Polymer synthesis by microorganisms: technology polyhydroxybutyrate synthase of Ralstonia eutropha.
and economics. Trends Biotechnol 1987, 5:246-250. Biomacromolecules 2003, 4:504-509.
21. Aldor IS, Kim SW, Prather KLJ, Keasling JD: Metabolic 31. Hezayen FF, Steinbuchel A, Rehm BHA: Biochemical and
 engineering of a novel propionate-independent pathway for the enzymological properties of the polyhydroxybutyrate synthase
production of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) from the extremely halophilic archaeon strain 56. Arch Biochem
in recombinant Salmonella enterica serovar Typhimurium. Biophys 2002, 403:284-291.
Appl Environ Microbiol 2002, 68:3848-3854.
This paper demonstrates an effective route to the 3HV monomer in 32. Jia Y, Yuan W, Wodzinska J, Park C, Sinskey AJ, Stubbe J:
P(3HB-co-3HV) from an unrelated carbon source using the recently Mechanistic studies on class I polyhydroxybutyrate (PHB)
discovered E. coli enzymes Sbm and YgfG, which form part of a novel synthase from Ralstonia eutropha: class I and III synthases
pathway to convert succinyl-CoA to propionyl-CoA. Further work with the share a similar catalytic mechanism. Biochemistry 2001,
recombinant S. enterica host, which can synthesize vitamin B12 de novo 40:1011-1019.
under anaerobic conditions, might eliminate the requirement for exogen-
ous cyanocobalamin addition for the B12-dependent mutase. 33. Yuan W, Jia Y, Tian JM, Snell KD, Muh U, Sinskey AJ, Lambalot RH,
Walsh CT, Stubbe J: Class I and III polyhydroxyalkanoate
22. Fukui T, Abe H, Doi Y: Engineering of Ralstonia eutropha for synthases from Ralstonia eutropha and Allochromatium
 production of poly(3-hydroxybutyrate-co-3- vinosum: characterization and substrate specificity studies.
hydroxyhexanoate) from fructose and solid-state properties of Arch Biochem Biophys 2001, 394:87-98.
the copolymer. Biomacromolecules 2002, 3:618-624.
A sophisticated pathway design utilizing the heterologous expression of 34. Rehm BHA, Antonio RV, Spiekermann P, Amara AA, Steinbuchel A:
PHA synthase and enoyl-CoA hydratase (PhaCJ) from A. punctata, Molecular characterization of the poly(3-hydroxybutyrate)
crotonyl-CoA reductase (Ccr) from Streptomyces cinnamonensis and (PHB) synthase from Ralstonia eutropha: in vitro evolution,
native 3-ketothiolase, acetoacetyl-CoA reductase and (S)-3HB-CoA site-specific mutagenesis and development of a PHB synthase
dehydrogenase activities to make P(3HB-co-3HHX) from fructose in protein model. Biochim Biophys Acta 2002, 1594:178-190.

Current Opinion in Biotechnology 2003, 14:475–483 www.current-opinion.com


Metabolic engineering of polyhydroxyalkanoates Aldor and Keasling 483

35. Liebergesell M, Rehalkar S, Steinbuchel A: Analysis of the 3-hydroxyacyl ACP:CoA transacylase, which diverts
Thiocapsa pfennigii polyhydroxyalkanoate synthase: intermediates of fatty acid de novo biosynthesis. J Biol Chem
subcloning, molecular characterization and generation of 2002, 277:42926-42936.
hybrid synthases with the corresponding Chromatium vinosum
enzyme. Appl Microbiol Biotechnol 2000, 54:186-194. 45. Hisano T, Tsuge T, Fukui T, Iwata T, Miki K, Doi Y: Crystal structure
of the (R)-specific enoyl-CoA hydratase from Aeromonas
36. Zhang S, Kamachi M, Takagi Y, Lenz RW, Goodwin S: caviae involved in polyhydroxyalkanoate biosynthesis. J Biol
Comparative study of the relationship between monomer Chem 2003, 278:617-624.
structure and reactivity for two polyhydroxyalkanoate
synthases. Appl Microbiol Biotechnol 2001, 56:131-136. 46. Hong SH, Park CJ, Moon SY, Park JP, Lee SY: In silico prediction
and validation of the importance of the Entner-Doudoroff
37. Qi Q, Steinbuchel A, Rehm BHA: In vitro synthesis of pathway in poly(3-hydroxybutyrate) production by
poly(3-hydroxydecanoate): purification and enzymatic metabolically engineered Escherichia coli. Biotechnol Bioeng
characterization of type II polyhydroxyalkanoate synthases 2003, 83:854-863.
PhaC1 and PhaC2 from Pseudomonas aeruginosa.
Appl Microbiol Biotechnol 2000, 54:37-43. 47. Han M, Yoon SS, Lee SY: Proteome analysis of metabolically
engineered Escherichia coli producing poly(3-
38. Spiekermann P, Rehm BHA, Kalscheuer R, Baumeister D, hydroxybutyrate). J Bacteriol 2001, 183:301-308.
Steinbuchel A: A sensitive, viable-colony staining method using
Nile red for direct screening of bacteria that accumulate 48. Mantzaris NV, Kelley AS, Srienc F, Daoutidis P: Optimal carbon
polyhydroxyalkanoic acids and other lipid storage compounds. source switching strategy for the production of PHA
Arch Microbiol 1999, 171:73-80. copolymers. AIChE J 2001, 47:727-743.

39. Wu H, Sheu D, Lee C: Rapid differentiation between short-chain- 49. Mantzaris NV, Kelley AS, Daoutidis P, Srienc F: A population
length and medium-chain-length polyhydroxyalkanoate-  balance model describing the dynamics of molecular weight
accumulating bacteria with spectrofluorometry. J Microbiol distributions and the structure of PHA copolymer chains.
Methods 2003, 53:131-135. Chem Eng Sci 2002, 57:4643-4663.
For the mathematically inclined, a rigorous population balance model is
40. Kichise T, Taguchi S, Doi Y: Enhanced accumulation and presented that describes the dynamics of the synthesis of P(3HB-co-
 changed monomer composition in polyhydroxyalkanoate 3HV) and PHB-P(3HB-co-3HV) block copolymers in nongrowing R.
(PHA) copolyester by in vitro evolution of Aeromonas caviae eutropha cells. Predicted optimal conditions for di-block and tri-block
PHA synthase. Appl Environ Microbiol 2002, 68:2411-2419. copolymer synthesis fell in the range of feasible bioprocessing manipula-
An in vitro approach (error-prone PCR) was used for the molecular tions.
evolution of A. punctata PHA synthase for enhanced accumulation of
P(3HB-co-3HHX) in recombinant E. coli harboring phaABJP. Interest- 50. Snell KD, Feng F, Zhong L, Martin D, Madison LL: YfcX enables
ingly, in contrast to other work [41], the 3HHX fraction was affected by  medium-chain-length poly(3-hydroxyalkanoate) formation
evolution of the synthase. The mutant generation and screening meth- from fatty acids in recombinant Escherichia coli fadB strains.
odology is nicely illustrated in a figure in this paper. J Bacteriol 2002, 184:5696-5705.
An excellent example of reverse engineering to understand why fadB
41. Amara AA, Steinbuchel A, Rehm BHA: In vivo evolution of the mutants of E. coli, with a disrupted fatty acid b-oxidation complex, can
 Aeromonas punctata polyhydroxyalkanoate (PHA) synthase: still incorporate monomer units with a chain length shorter than that of the
isolation and characterization of modified PHA synthases with fatty acid carbon source. YfcX (subunit of fatty acid oxidation complex)
enhanced activity. Appl Microbiol Biotechnol 2002, 59:477-482. was shown to be crucial for the synthesis of PHAs composed of medium
A pioneering protein engineering effort to use the XL1-Red mutator strain chain length monomers in these strains and this resolves a long-standing
to evolve A. punctata PHA synthase in vivo. All other attempts to engineer ambiguity regarding recombinant PHA biosynthesis in E. coli.
the enzyme to date have relied on in vitro mutagenesis. Mutant synthases
showed higher in vivo specific activity, resulted in higher P(3HB-co-3HHX) 51. Campbell JW, Morgan-Kiss RM, Cronan JE: A new Escherichia
accumulation in recombinant E. coli harboring phaAB and treated with coli metabolic competency: growth on fatty acids by a novel
acrylate, and also generated polymers with increased weight average anaerobic b-oxidation pathway. Mol Microbiol 2003,
molecular mass. However, insignificant changes were seen in composi- 47:793-805.
tion compared with wild type.
52. Tsuge T, Hisano T, Taguchi S, Doi Y: Alteration of chain length
42. Takase K, Taguchi S, Doi Y: Enhanced synthesis of poly(3-  substrate specificity of Aeromonas caviae R-enantiomer-
hydroxybutyrate) in recombinant Escherichia coli by means of specific enoyl-coenzyme A hydratase through site-directed
error-prone PCR mutagenesis, saturation mutagenesis, and mutagenesis. Appl Environ Microbiol 2003, 69:4830-4836.
in vitro recombination of the type II polyhydroxyalkanoate An interesting recent paper that illustrates the potential for rational design
synthase gene. J Biochem (Tokyo) 2003, 133:139-145. in protein engineering of PHA biosynthetic enzymes. This approach is
feasible for engineering (R)-enoyl-CoA hydratase, because its crystal
43. Crameri A, Raillard SA, Bermudez E, Stemmer WPC: DNA shuffling structure has been determined.
of a family of genes from diverse species accelerates directed
evolution. Nature 1998, 391:288-291. 53. Park SJ, Park JP, Lee SY, Doi Y: Enrichment of specific monomer
in medium-chain-length poly(3-hydroxyalkanoates) by
44. Hoffmann N, Amara AA, Beermann BB, Qi Q, Hinz HJ, Rehm BHA: amplification of fadD and fadE genes in recombinant
Biochemical characterization of the Pseudomonas putida Escherichia coli. Enzyme Microb Technol 2003, 33:62-70.

www.current-opinion.com Current Opinion in Biotechnology 2003, 14:475–483

You might also like