You are on page 1of 14

Available online at www.sciencedirect.

com

Metabolic engineering of Clostridium acetobutylicum: recent


advances to improve butanol production
Tina Lütke-Eversloh and Hubert Bahl

The biosynthesis of the solvents 1-butanol and acetone is declined rapidly after the 1950s as a result of the cheaper
restricted to species of the genus Clostridium, a diverse group petrochemical production of butanol [1,2]. As shown in
of Gram-positive, endospore forming anaerobes comprising Figure 1, research activities in academia and industry
toxin-producing strains as well as terrestrial non-pathogenic steeply increased in the early 1980s as a response to the oil
species of biotechnological impact. Among solventogenic crisis in the 1970s with approximately equal efforts in
clostridia, Clostridium acetobutylicum represents the model technical aspects, that is fermentation and downstream
organism and general but yet important genetic tools were processing, and research on physiology and genetics of
established only recently to investigate and understand the solventogenic clostridia. In the context of today’s general
complex life cycle-accompanied physiology and its regulatory interests in biofuels, scientific publications on clostridial
mechanisms. Since clostridial butanol production regained research increased again in the past few years, probably
much interest in the past few years, different metabolic enforced by DuPont’s and British Petrol’s announcement
engineering approaches were conducted — although in 2006 to reconstitute the industrial-scale ABE fermen-
promising and in part successful strategies were employed, the tation in the United Kingdom (URL: http://www.bp.com,
major breakthrough to generate an optimum phenotype with press release date: June 20, 2006).
superior butanol titer, yield and productivity still remains to be
expected. As a consequence, various review articles were published
recently, summarizing general aspects of the ABE fer-
Address mentation [2,3,4,5–7], focussing on production countries
Department of Microbiology, Institute of Biological Sciences, University [8,9], patent review [10], product toxicity and tolerance
of Rostock, Albert Einstein-Str. 3, 18051 Rostock, Germany
[11,12,13], as well as technical process development [14–
Corresponding authors: Lütke-Eversloh, Tina 16], respectively. Reviews on clostridial sporulation
(tina.luetke-eversloh@uni-rostock.de) and Bahl, Hubert [17,18], cellulolytic clostridia [19,20,21,22], and con-
(hubert.bahl@uni-rostock.de) solidated bioprocessing perspectives (e.g., [23,24]) are
also available.
Current Opinion in Biotechnology 2011, 22:634–647
The intention of this review paper is to specifically sum
This review comes from a themed issue on up the development of metabolic engineering tools and
Tissue, cell and pathway engineering
Edited by Uwe T. Bornscheuer and Ali Khademhosseini
strategies for C. acetobutylicum to improve the innate
butanol production. As an update of E. T. Papoutsaki’s
Available online 4th March 2011 review of 2008 [25], engineering approaches conducted
within the past few years are highlighted and important
0958-1669/$ – see front matter
# 2011 Elsevier Ltd. All rights reserved.
physiological aspects of the fermentative metabolism are
discussed.
DOI 10.1016/j.copbio.2011.01.011

Central metabolic pathways and their


regulation
Introduction The fermentation of sugars by clostridia typically causes
The clostridial acetone–butanol–ethanol (ABE) fermen- three different growth phases: first, exponential growth
tation represents one of the oldest industrial fermentation and formation of acids, second, transition to stationary
processes known, ranking second in scale only to ethanol growth phase with reassimilation of acids and concomitant
fermentation by yeast. In the early 1920s, Chaim Weiz- formation of solvents, and third, formation of endospores.
mann, who later became Israel’s first president, discov- C. acetobutylicum can utilize a variety of carbohydrates,
ered the anaerobic bacterium Clostridium acetobutylicum including pentoses, hexoses, oligosaccharides and polysac-
which naturally produces acetone, butanol and ethanol in charides — an important benefit for converting lignocellu-
a ratio of 3:6:1. The initial production plants for the ABE lose hydrolysates into biofuels. Although cellulosome genes
fermentation were developed because of the World War are present and expressed, C. acetobutylicum is not capable of
I-dependent demand of acetone for the cordite manu- using cellulose as a substrate. Recent global transcriptional
facture, but butanol was only an unwanted byproduct. and mutant analyses provided new insights into carbo-
However, butanol became a more important product after hydrate utilization and regulatory constraints such as the
the war. Nevertheless, industrial ABE fermentation well-known carbon catabolite repression [26–29].

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 635

Figure 1 genases and their regulation still remains to be elucidated


[33,34,35].

The biphasic metabolism of C. acetobutylicum is tightly


100
associated with different growth stages, that is exponen-
tially growing cells mainly produce acetic and butyric
80
acid, while acetone and butanol are formed by stationary
Publications

60 cells (Figure 2b). After entering the stationary phase, cells


start to synthesize granulose as intracellular storage com-
40 pound, and these ‘‘clostridial stage’’ cells can be micro-
scopically distinguished from vegetative cells [36].
20 However, the regulatory mechanisms of granulose for-
mation and re-utilization are not known. Subsequently,
0 the sporulation process is initiated and the granulose
1950 1960 1970 1980 1990 2000 2010
granula presumably serve as energy and carbon source
Year
for endospore formation [1,36]. The resistant spores are
Current Opinion in Biotechnology
able to survive for a long period of time and germinate
under suitable environmental conditions.
Scientific publications on solventogenic clostridia since 1950. More than
4500 reference hits obtained from database searches (PubMed, Scopus
and ISI Web of Science; as of December 2010) were screened according The role of the well-known sporulation regulator Spo0A
to their relation to basic and applied research on butanol fermentation of Gram-positive bacteria has been shown to be important
and solventogenic clostridia in general. A total of 1238 publications since for the initiation of endospore and solvent formation in C.
1950 were considered except for conference abstracts, university
reports and references lacking author or source information. The pie
acetobutylicum, but in regard of the quite complex net-
charts on top show the ratio between publications on physiology and works, more regulatory factors are very likely to be
genetics (white) and those covering topics of fermentation and involved [17,18]. However, despite the fact that Spo0A
downstream processing (black) for each decade. is a major regulator of solventogenesis, sporulation itself is
not required for butanol and acetone production — more-
over, sporulation is not desirable. According to the
As summarized in Figure 2, glucose is catabolized to importance of the phosphorylated form of Spo0A for
pyruvate via the Embden–Meyerhof–Parnas pathway the initiation of spore formation and its connection to
and acetyl-CoA is primarily formed by the pyruvate:fer- solventogenesis, the metabolic intermediate butyrylpho-
redoxin oxidoreductase. Under certain growth conditions, sphate was also found to play a putative regulatory role.
such as pH values >5 and iron limitation, lactate can be Experiments including for example, the quantification of
the major fermentation product [30,31]. Acetate is syn- intracellular acetylphosphate and butyrylphosphate
thesized via phosphotransacetylase and acetate kinase levels, exhibited the association of high butyrylphosphate
reactions with the latter reaction providing ATP. For concentrations with the reutilization of carboxylic acids as
the biosynthesis of butyrate, two molecules of acetyl- well as the initiation of solvent biosynthesis, indicating
CoA are condensed to acetoacetyl-CoA, followed by a that butyrylphosphate might be a regulatory molecule as
reduction to butyryl-CoA, which is then converted to well, possibly acting as a phosphodonor for transcriptional
butyrate via phosphotransbutyrylase and butyrate kinase factors [37].
reactions with ATP generation.
Considering modern methodologies, the group of E. T.
As a reaction to the significant decrease of the pH in the Papoutsakis started to provide comprehensive insights
culture, which may destroy the essential proton gradient into the life cycle of C. acetobutylicum by analyzing the
across the membrane, C. acetobutylicum switches its metab- growth-associated stages on a global genomic and tran-
olism from acidogenesis to solventogenesis. Acetate and scriptomic as well as on a morphological level with a
butyrate are reassimilated to their corresponding CoA particular focus on sigma factors putatively involved in
derivatives catalyzed by the acetoacetyl-CoA:acyl-CoA sporulation [18,38]. So far, these explorative approaches
transferase, with acetoacetyl-CoA as the CoA donor. guided the researchers to potential candidates, and inac-
Particularly when reducing equivalents are limiting, acet- tivation of the transcriptional regulators sE and sG exhib-
oacetate is decarboxylated to acetone in order to drive the ited an opportunity to uncouple sporulation and
transferase reaction by acetoacetate removal [32]. Butyr- solventogensis in batch fermentation [39].
aldehyde and butanol dehyrdogenase activities, which
can be provided by different dehydrogenases, convert In conclusion, only little is known on the regulatory
butyryl-CoA to butyraldehyde and finally to butanol, the circuits and molecular mechanisms for the transition
major fermentation product of C. acetobutylicum (Figure between acidogenesis and solventogenesis and the onset
2a). However, the role of the different alcohol dehydro- of the sporulation process. Although many efforts and

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


636 Tissue, cell and pathway engineering

Figure 2

(a) Glucose
2 NAD+

Glycolysis
2 NADH + H+

2 ADP + Pi

NAD+ 2 ATP
NADH
+ H+ Ldh Pdc
Lactate Pyruvate
H2 Fdox CoA
Hyd Pfor NAD+ CO2
2 H+ Fdred CO2 -6.5 +
+ CoA NAD(P)H NAD(P)
CoA NADH + H + +
Pi +H
Acetyl-P Acetyl-CoA Acetaldehyde Ethanol
AAc Pta AdhE AdhE
ADP + Pi Ack -5.4 Acetyl-CoA 3.8 -4.5
ATP 6.4 AAc-CoA Thl
5.3 CoA Ac-CoA/Bu-CoA
CO2
fAB
Ct .2
Ac/Bu
Acetate 0 - Acetone
Acetoacetyl-CoA Acetoacetate
CtfAB Adc
NADH + H + -0.2 -6.5
Hbd
-5.4 NAD+

3-Hydroxybutyryl-CoA
Crt
0.5 H2O

Butyrate AAc-CoA Crotonyl-CoA


AAc Etfred NAD+
Ct Bcd
f NADH + H+
ATP Buk -0 AB -14.6 Etfox NAD(P)+
CoA .
2 NAD+ + CoA NAD(P)H
ADP + Pi 6.4
Pi NADH + H+ + H+
Butyryl-P Butyryl-CoA Butyraldehyde Butanol
Ptb AdhE AdhE
-5.4 3.8 -4.5

(b)
ACIDOGENESIS

Vegetative cells

Spore germination Differentiation

Growth

Spores Clostridial form

Spore maturation Sporulation

Forespores

SOLVENTOGENESIS

Current Opinion in Biotechnology

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 637

Table 1

Stoichiometric reactions for C. acetobutylicum.

Reaction Substrate Products ATP yield


(1) 1 Glucose 2 Acetate + 2 CO2 + 4 H2 4
(2) 1 Glucose 1 Butyrate + 2 CO2 + 2 H2 3
(3) 1 Glucose 0.6 Acetate + 0.7 butyrate + 2 CO2 + 2.7 H2 3.3
(4) 1 Glucose 2 Lactate 2
(5) 1 Glucose 1 Acetate + 1 ethanol + 2 CO2 + 2 H2 3
(6) 1 Glucose 2 Ethanol + 2 CO2 2
(7) 1 Glucose 1 Acetone + 3 CO2 + 4 H2 2
(8) 1 Glucose 1 Butanol + 2 CO2 2
(9) 1 Glucose 0.5 Acetone + 0.5 butanol + 2.5 CO2 + 2 H2 2
(10) 1 Glucose 0.3 Acetone + 0.6 butanol + 0.2 ethanol + 2.3 CO2 + 1.2 H2 2
(11) 1 Glucose + 1 acetate 1 Acetone + 1 ethanol + 3 CO2 + 2 H2 2
(12) 1 Glucose + 1 acetate 1 Acetone + 0.5 butanol + 3 CO2 + 2 H2 2
(13) 1 Glucose + 1 butyrate 1 Acetone + 1 butanol + 3 CO2 + 2 H2 2

Both actual and theoretical fermentative reactions were calculated according to balanced reducing equivalents; water and protons were not
considered. Neither other substrates than glucose, acetate and butyrate, nor specific physiological conditions were taken into account. See text for
details.

some progress were made, we do not have a detailed equivalents can be regenerated in the butanol pathway,
picture on the regulation of solvent production in C. provided that reduced ferredoxin transfers its electrons to
acetobutylicum. In addition to Spo0A, other regulators must NAD(P)+ and no molecular hydrogen is formed.
be involved in the solventogenic shift which specifically
control solventogenic operons. But what are the inducing Manipulation of the redox balance has been demon-
signals, how do the regulators interact, and how are the strated to push the metabolism of C. acetobutylicum
regulatory networks connected? The answers to these towards butanol formation. Provision of artificial electron
questions will certainly facilitate straightforward meta- carriers such as methyl viologen or neutral red, increasing
bolic engineering of C. acetobutylicum. the hydrogen partial pressure or gassing with carbon
monoxide led to high butanol and low acetone production
Considerations on redox balance and rates; similar results were obtained by cultivation under
stoichiometry iron-limiting conditions or using whey as a substrate [41–
Regarding the metabolic pathways (Figure 2), ATP is 43,44,31]. All these techniques were employed to reduce
predominantly generated during acidogenesis, whereas hydrogenase activities, and the lack of molecular hydro-
high NAD(P)H levels were proposed to induce solven- gen formation resulted in an electron flow towards buta-
togenesis [40]. Table 1 lists several possible stoichio- nol for the regeneration of the NAD(P)+ pool. The fact
metric reactions of glucose to the different that the ferredoxin/hydrogenase node plays an important
fermentation products considering both carbon and redox role for butanol production was also demonstrated for C.
balances. In practice, however, C. acetobutylicum does saccharoperbutylacetonicum strain N1–4 [45]: employing
usually not follow only one of the simple routes. Instead, antisense RNA against the hupCBA gene cluster, which
multiple products are formed, best approximated by encodes a hydrogen-uptake hydrogenase, the butanol/
reactions (3) for the acidogenic and (10) for the solvento- acetone ratio was decreased from 2.9 to 1.3, because
genic phase, respectively (Table 1). Another important the increased hydrogen formation rate caused a 76%
issue is that solventogenesis can only pursue when glu- decrease in butanol production [45]. Therefore, targeting
cose is concomitantly metabolized, which makes it diffi- the hydrogen production of C. acetobutylicum might con-
cult to stoichiometrically sum up all fermentation stitute a promising metabolic engineering strategy. Inter-
products [1]. But in theory, homo-butanol fermentation estingly, novel energetic properties of some hydrogenase
is possible as shown by reaction (8) (Table 1): two moles representatives were discovered more recently. The tri-
of ATP can be provided by glycolysis and the reducing meric bifurcating hydrogenase of Thermotoga maritima

( Figure 2 Legend ) Acidogenesis and solventogenesis in C. acetobutylicum: Metabolic pathways (a) and their relation to the life cycle stages (b). (a)
The red letters show the enzymes involved in the fermentative pathways: Ldh, lactate dehydrogenase; Pdc, pyruvate decarboxylase; Hyd,
hydrogenase; Pfor, pyruvate:ferredoxin oxidoreductase; Fd, ferredoxin; Pta, phosphotransacetylase; Ack, acetate kinase; AdhE, aldehyde/alcohol
dehyrogenase; CtfAB, acetoacetyl-CoA:acyl-CoA transferase; Adc, acetoacetate decarboxylase; Thl, thiolase; Hbd, 3-hydroxybutyryl-CoA
dehydrogenase; Crt, crotonase, Bcd, butyryl-CoA dehydrogenase; Etf, electron transfer flavoprotein; Ptb, phosphotransbutyrylase, Buk, butyrate
kinase. AAc, acetoacetate; AAc-CoA, acetoacetyl-CoA; Ac/Bu, acetate/butyrate; Ac-CoA/Bu-CoA, acetyl-CoA/butyryl-CoA; ox, oxidized; red,
reduced. The blue numbers represent the standard Gibbs energy changes according to [6]. (b) Acidogenesis and solventogenesis referring to the
respective growth stages. The major (green/blue) and minor (grey) metabolic fluxes are indicated in the miniature pathways, which represent simplified
schemes of (a).

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


638 Tissue, cell and pathway engineering

utilizes reduced ferredoxin and NADH simultaneously, known on the genetic regulation of the energy and redox
but C. beijerinckii is the only solventogenic strain that status in solventogenic clostridia, but more interesting
potentially encodes a homologous protein [46]. Another results can be expected in the near future. Again, soph-
exciting group of enzymes are the energy-converting isticated metabolic engineering approaches will be based
[NiFe] hydrogenases which resemble the well-studied on the identification of the molecular regulatory switches,
complex I and are also designated as ‘‘respiratory’’ hydro- sensing and transferring redox signals to induce or prolong
genases [47]. Although similar genes were found in a few butanol synthesis. Turning such information to account,
clostridial species, none are present in the C. acetobutyli- C. acetobutylicum might be outflanked to increase the
cum genome [48]. carbon flow towards the desired product without redox-
based regulatory constraints.
Yet another scientific breakthrough regarding anaerobic
energy metabolism was achieved recently, solving the Analytical and engineering tools for
hitherto unbalanced stoichiometry of the C. kluyveri etha- C. acetobutylicum
nol/acetate fermentation [49]. Here, the exergonic reac- After publication of the genome sequence of C. acetobu-
tion from crotonyl-CoA to butyryl-CoA is energetically tylicum ATCC 824 [55], several transcriptome analyses
coupled with the endergonic reduction of ferredoxin by related to various physiological aspects such as sporula-
NADH, which is catalyzed by the cytosolic Bcd/Etf tion, solventogenesis or butanol stress were conducted by
(butyryl-CoA dehydrogenase/electron transfer flavopro- the laboratory of E. T. Papoutsakis (e.g., [56–58]). Among
tein) complex [50]. Analogously, the NADH-dependent these, the most comprehensive DNA microarray study on
and ferredoxin-dependent reduction of NADP+ is C. acetobutylicum batch cultures was published in 2008
mediated by the electron bifurcating NfnAB (NADH- [38], providing detailed analyses on all relevant not yet
dependent reduced ferredoxin:NADP+ oxidoreductase) assigned sigma factors putatively involved in the sporula-
complex, an important link for NADPH generation since tion process. The first report from a different laboratory
a transhydrogenase gene is absent [51]. employing DNA microarray methods was published only
in 2009, revealing transcriptional details on detoxification
The energy of the flavin-based electron bifurcation in C. and redox balance mechanisms in C. acetobutylicum related
kluyveri is conserved by pumping protons out of the cell, to oxygen stress [59]. Since recently, global transcrip-
which allows additional ATP generation. Thus, the dogma tional analyses can also be performed with the related
of the past decades that substrate level phosphorylation is strain C. beijerinckii NCIMB 8052 [60], but other solven-
the only energy source in fermenting bacteria was shown to togenic clostridia are thus far not accessible to DNA
be wrong [52]. The major player in the electron transport microarray analyses, albeit other genomes are expected
phosphorylation in C. kluyveri is the membrane-associated to be sequenced or are in progress, respectively [61].
Rnf (Rhodobacter nitrogen fixation) complex providing
ferredoxin:NAD+ oxidoreductase activity, which occurs Proteome analyses can be regarded as a further step
in a variety of microbes and was recently biochemically towards understanding the solventogenic physiology
characterized in Acetobacterium woodii [53,54]. According using ‘omics’ applications. The first system level two-
to the available genome sequences, C. pasteurianum and C. dimensional protein gels from chemostat cells were
acetobutylicum are the only clostridia lacking Rnf-homolo- already published in 2002, and major proteins induced
gous genes. Hence, C. acetobutylicum harbors a cytosolic at the onset of solventogenesis such as the acetoacetate
Bcd/Etf complex to reduce crotonyl-CoA, but obviously, decarboxylase were identified [62]. Four years later,
this reaction is not coupled to a membrane-associated comparative mass spectroscopic analyses of cytosolic
electron transport mechanism. The reason for the absence proteins related to the sporulation master regulator Spo0A
of an energy conserving step is not known. We speculate were conducted and compared accordingly to previous
that because of the severe membrane damaging properties transcriptome data [63,64]. The latter approach was done
of acids and solvents, C. acetobutylicum abandoned this using protein extracts from C. acetobutylicum batch cul-
option for a reduced susceptibility to its own fermentation tures, similar to a recent proteomic study of C. acetobuty-
products. Another reason might be a faster and probably licum strain DSM 1731 as compared to its mutant strain
much more flexible energy metabolism, because all necess- Rh8 obtained earlier by chemical mutagenesis. The phe-
ary enzymes are soluble and usually, those proteins which notype of increased butanol tolerance and yield was
are located in the membrane constitute the limiting step of reflected in the proteome data — although not entirely
a metabolic pathway. However, the fact that a similar or matching previous transcriptome results on butanol stress
different energy conserving mechanism has not been experiments [65,66].
found in C. acetobutylicum does not strictly exclude its
existence. The emphasis on the cultivation conditions for the above
mentioned ‘omics’ publications can be explained as fol-
The above-mentioned findings on energy conservation in lows: simple batch fermentations, even if reproducible,
anaerobic bacteria are quite new and almost nothing is differ in general from continuous chemostat cultures.

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 639

Whereas a batch fermentation resembles the natural life other model organisms such as Escherichia coli or Sacchar-
cycle of C. acetobutylicum, chemostat cultivation is an omyces cerevisiae.
excellent research tool to keep the cells at a steady state,
that is all parameters are perfectly constant and the native Regarding plasmid-based gene overexpression in C. acet-
metabolism can be pushed into a desired direction, such obutylicum, an improvement of the tedious in vivo meth-
as uncoupling solventogenesis from sporulation. Using ylation protocol [79] can now be achieved by using host
steady state C. acetobutylicum cells, distinct proteome strains lacking the very active restriction endonuclease
reference maps for acidogenesis and solventogenesis Cac824I [80,81]. Furthermore, the range of available
were generated and accompanied by detailed transcrip- plasmids was extended by the modular pMTL80000
tome data [67]. Such reference maps provide a suitable series providing different selection markers, a choice of
basis for detailed single parameter analyses with a mini- Gram-positive and Gram-negative replicons, a multiple
mum degree of disturbance, for example, specific exogen- cloning site, tra genes for conjugal transfer and allows
ous factors such as nutrient supply or defined stress simplified cloning in E. coli using blue/white screening.
stimuli can be examined as well as the alteration of Depending on the application, the different shuttle plas-
endogenous factors like an interesting mutant strain. mid modules can be assembled individually and com-
bined with the gene(s) of interest [82].
Continuing the development of more sophisticated
systems biology methods, metabolome analyses can The same research group also developed a reliable
provide important information on metabolic pathways method for directed gene disruption by adapting the
and fluxes. As a simple general rule for system Targetron system to clostridial species, a similar paper
level approaches, the quality of information increases was published by Shao et al. [83,84]. Further modifi-
with proximity to the organism’s actual physiology cations of the ClosTron system allow by now the con-
(i.e., gene < protein < metabolite < pathway) which is struction of multiple knock-out (KO) mutants via
usually associated with increasing experimental complex- flippase-mediated marker recycling [85], providing an
ity and difficulty. Interestingly, the first metabolome data important platform for sustainable metabolic engineering
sets on the central carbon metabolism of C. acetobutylicum of clostridial butanol production. Probably because of
derived from isotope tracer experiments were published commercial licensing issues, other research groups devel-
only recently, exhibiting a complete tricarboxylic acid oped different KO methods for C. acetobutylicum and the
(TCA) cycle with a reductive and oxidative branch laboratory of N. P. Minton also filed a patent for a
towards succinate [68,69]. ClosTron-independent KO system [80,86–89]. Until
now, the most often described method in the literature
A common goal of various ‘omics’ strategies is the to specifically decrease gene expression in C. acetobuty-
development of a computational model resembling liucm was the application of antisense RNA to ‘knock-
the metabolic pathways and fluxes. Stoichiometric cal- down’ clostridial genes [90].
culations for modeling the ABE fermentation were
already described in 1984 [70], and Desai et al. [71] Rational metabolic engineering strategies
conducted metabolic flux analyses of the acid reassi- As shown in Figure 1, the ABE fermentation regained
milation in C. acetobutylicum prior its genome sequence much interest after the oil crisis in the 1980s and many
was available. The first computational model for kinetic studies on optimizing the cultivation conditions and
simulations of the ABE fermentation was published in varying the feeding regime were published. Since clos-
2007 for C. saccharoperbutylacetonicum N1–4 [72,73]. tridia were genetically not accessible at that time, other
Regarding C. acetobutylicum, genome-scale models were approaches were chosen to investigate the physiology and
independently developed by different research groups regulation of butanol biosynthesis. A major improvement
[74–77] and are discussed in a recent review [78]. in butanol production by C. acetobutylicum and other
solventogenic strains was achieved by lowering the redox
The systems biology approaches and in silico models potential to promote butanol formation, for example, by
described above provide a useful tool to predict metabolic using glycerol as co-substrate or addition of methyl violo-
engineering targets, and experimental validation of such gen (for review see e.g., [43,44]). Within the past 15
targets generally requires much more efforts and respect- years, overcoming the burden of genetic inaccessibility
ive literature is still sparse. The major reason for this is the was successful and the number of metabolic engineering
difficult genetic accessibility of clostridia, which ham- approaches for C. acetobutylicum steadily increased; refer-
pered successful engineering of C. acetobutylicum and ences published prior 2008 can be found elsewhere
other solventogenic strains for a long time. However, a [4,25], whereas recent efforts are listed in Table 2.
number of suitable molecular protocols were developed
in recent years, although the clostridial portfolio of The major principle of rationally designing and improv-
genetic methods displays only a minor fraction as com- ing a microbial production strain is to increase the meta-
pared to the metabolic engineering toolbox available for bolic flux towards the desired product, usually

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


640 Tissue, cell and pathway engineering

Table 2

Metabolic engineering of C. acetobutylicum for improved butanol production since 2005.

Parental strain Strategy Phenotype Genotype/plasmid Reference


(compared to control)
C. acetobutylicum Disruption of butyrate, acetone, No information Dbuk, DctfAB, Dldh Dpta/ack [96]
ATCC 824 lactate, and acetate pathways
Dcac1515Dupp
C. acetobutylicum Disruption of butyrate, acetone, No information Dbuk, DctfAB, Dldh DhydA [96]
ATCC 824 lactate, and hydrogen pathways
Dcac1515Dupp
C. acetobutylicum Based on a previously published 178 mM (176) butanol, pCASAAD [93,94]
ATCC 824 idea, promotors for enhanced 61 mM (109) acetone, (pptb-adhE1-pthl-asRNA:ctfB)
adhE1 overexpression and ctfA 305 mM (20) ethanol,
downregulation were exchanged 2 mM (37) butyrate,
85 mM (77) acetate;
earlier butanol production
C. acetobutylicum M5 Based on a previously published 92 mM (84) butanol, p94AAD3 (pptb-adhE1) [86,101]
idea, the native sol promotor was 20 mM (8) ethanol,
exchanged by the ptb promotor 72 mM (99) butyrate,
for adhE1 expression in the 159 mM (101) acetate;
solvent-negative M5 strain no acetone
C. acetobutylicum M5 Expression of the sol operon in 154 mM (69) butanol, pIMP1E1AB (psol-adhE1-ctfAB) [101,102]
the solvent-negative M5 strain 20 mM (9) ethanol,
10 mM (none) acetone,
54 mM (85) butyrate,
227 mM (168) acetate
C. acetobutylicum Disruption of acetone pathway 100 mM (184) butanol, adc::Int(180/181) [95]
EA 2018 36 mM (59) ethanol,
4 mM (49) acetone,
3 mM (6) butyrate,
60 mM (8) acetate;
80% (71) butanol ratio
of total solvents
C. acetobutylicum Co-production of riboflavin as 70 mg/l (none) riboflavin, pJpGN (pptb-ribGBAH) [103]
ATCC 824 a high-value product 193 mM (191) butanol

Tables summarizing previous approaches can be found in [4,32].

accomplished by reduced byproduct formation and elimi- [94] picked up this strategy again, but optimized gene
nated enzymatic bottle necks. Depending on the type of expression of the asRNA:ctfB construct using the adc
product, metabolic engineering typically becomes more promotor and the ptb promotor for higher adhE1 expres-
and more difficult with increasing complexity of the sion led to a more distinct phenotype (Table 2). On the
physiology and decreasing knowledge on regulatory basis of metabolic flux analyses, the authors further co-
mechanisms. Thus, engineering C. acetobutylicum to overexpressed the thl gene with adhE1 in order to stimu-
improve butanol production represents in fact a very late the butyrate/butanol (C4) pathway. Although acetate
difficult goal because of the branched fermentative path- production could be reduced, the overall phenotype
way and a severe lack of information regarding regulatory comprising elevated thl expression in C. acetobutylicum
circuits determining the organism’s life cycle. Targeting was not improved [94].
the elimination of the acid forming pathways in C. acet-
obutylicum, buk-negative and pta-negative mutants were The first example of a Targetron-based KO mutant of C.
already described by Green et al. [91] and overexpression acetobutylicum defective in the central fermentative
of adhE1 (also referred to as aad) in different host strains metabolism was published by Jiang et al. [95] who
for increased alcohol production was investigated, too (for disrupted the adc gene by insertion of the group II intron.
review see [4,32]). Regarding the acetone branch, C. The adc KO mutant exhibited small amounts of acetone
acetobutylicum was successfully engineered for reduced because of non-enzymatic decarboxylation of acetoace-
acetone formation employing antisense RNA against ctfB tate, but also significantly lower butanol titers as com-
to improve the butanol:acetone ratio [92]. However, the pared to the parental strain C. acetobutylicum EA2018. The
mutant strain also exhibited reduced butanol production, butanol production was further improved by buffering the
and therefore, the ctfB ‘knock-down’ strain was combined fermentation media with calcium carbonate and addition
with adhE1 overexpression. Interestingly, this strain not of methyl viologen. The results of this study basically
only restored the butanol levels but also showed high confirmed the phenotype of the asRNA strategies pub-
ethanol titers of up to 200 mM [93]. Recently, Sillers et al. lished by the group of E. T. Papoutsakis, that is reduced

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 641

acetone synthesis also led to a reduced butanol pro- alternative acetate forming pathways might exist in C.
duction, although the earlier approaches targeted the ctfB acetobutylicum [102].
gene because ‘knock-down’ of the adc gene was not
successful [92]. Lastly, a simple approach from the economic point of
view is the idea to combine a bulk product with a value-
Recently, a comprehensive trial to effectively decrease added product. Cai and Bennett [103] realized this inven-
byproduct formation in C. acetobutylicum was the gener- tive strategy by engineering C. acetobutylicum for riboflavin
ation of two multiple-KO mutants, one comprising (vitamin B2) production: homologous overexpression of
deleted buk, ctfAB, ldh and pta-ack genes, the other the ribGBAH genes did not exhibit any negative effect on
exhibited KOs of the genes buk, ctfAB, ldh and hydA genes butanol production, but instead a high value compound
[96]. However, except for the butyrate titer and total was co-produced [103].
alcohol and acetone yield values of the buk single-KO
mutant, no phenotypic information on the performance or Combinatorial metabolic engineering
fitness of the engineered strains was provided in this strategies
patent application. Thus far, rational metabolic engineering of solventogenic
clostridia as compiled above revealed only limited suc-
Because the solventogenesis is naturally accompanied by cess. This might be attributed to the small portfolio of
the sporulation process — which eventually ceases buta- genetic tools for this bacterial group and hence, the
nol production — an asporogenous C. acetobutylicum strain eventual success for generating a superior butanol produ-
might constitute an excellent starting point for metabolic cing strain can be expected in the future due to the recent
engineering. Non-sporulating variants of C. acetobutylicum development of suitable techniques. On the other hand,
DSM1731 which were still capable of solvent production the principle of systematic approaches might comprise a
have been selected from continuous cultures after several general limitation because of multiple unknown factors
weeks of operation [97]. More popular asporogenous constituting a specific phenotype. Since native butanol
strains are degenerated variants which lost the megaplas- synthesis is exclusively performed by solventogenic clos-
mid pSOL1, such as C. acetobutylicum M5 and DG1 [98– tridia, the accompanied branched fermentative pathways
100]. The best studied pSOL1-encoded genes are those (Figure 2) have not evolved for the reason to be branched
responsible for solvent formation, that is adhE1 and ctfAB and complex, but to provide a distinct advantage for the
which form the tricistronic sol operon, and the adjacent organism. Therefore, it might be the better alternative to
adc gene, but most of the other relevant functions of look for a strain according to its overall performance, that
pSOL1 genes remain to be elucidated [3]. Complemen- is selecting an improved strain because of its phenotypic
tation of C. acetobutylicum M5 with the adhE1 gene characteristics, ideally combined with a gain of knowl-
restored butanol production without acetone formation, edge on the factors which specifically led to the pheno-
further improvement was achieved by exchanging the type of interest. This issue has been addressed to other
native sol promotor by the strong constitutive ptb promo- biotechnological microbes previously and is often
tor for adhE1 expression [86,101] (Table 2). It is note- referred to as ‘inverse metabolic engineering’ [104,105].
worthy at this point that the adhE1 gene (CAP0162) is not As a prerequisite, combinatorial approaches strictly
the only gene responsible for aldehyde/alcohol dehydro- depend on the availability of suitable screening methods
genase function in C. acetobutylicum, as it was erroneously to select the respective phenotype.
indicated in some publications. Overexpression of adhE2
(CAP0035) in the degenerated strain C. acetobutylicum In the broadest sense, the oldest and easiest screening
DG1 also restored butanol production without acetone procedure is mimicking nature: selection by the cell’s
formation [34]. survival of certain environmental conditions. In fact, this
screening method has been the most successful so far for
However, the major fermentation product of the engin- isolating better butanol producing clostridial strains.
eered pSOL1-free strains described above was acetate, Employing random chemical mutagenesis and butanol
accompanied by high butyrate concentrations, and differ- exposure, butanol tolerant strains of C. acetobutylicum were
ent attempts including KOs of ack and buk as well as co- selected and exhibited enhanced butanol production
overexpression of thl did not alter this phenotypic pattern (e.g., [106,107]). More recently, mutant Rh8 of C. acet-
[86]. A further attempt to address the high acid accumu- obutylicum DSM 1731 was obtained by genome shuffling
lation, Lee et al. [102] co-overexpressed the adhE1 and and selection in the presence of high butanol concen-
ctfAB genes in C. acetobutylicum M5: although acetate still trations [65]. The well-studied mutant C. beijerinckii
remained the major fermentation product, butanol titers BA101 was selected in the presence of 2-deoxyglucose
were increased whereas acetone production constituted after chemical mutagenesis, revealing increased amylo-
only 20% of the wild-type level. The authors speculated lytic activity and enhanced butanol production [108].
that the high acetate concentrations were because of the Details on general issues of butanol toxicity and tolerance
strain’s compensation for ATP generation and that were reviewed recently [11,12].

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


642 Tissue, cell and pathway engineering

Other screening methods of the pre-genome era of C. and validation, since the relation of sporulating cells to
acetobutylicum were described aiming to select solvent- butanol production has been demonstrated previously
negative mutants for classical biochemical and genetic (e.g., [36]). Nevertheless, the application of FACS to
analyses. These approaches included the use of suicide solventogenic clostridia opens up new perspectives
substrates such as allyl alcohol and bromobutyrate and regarding phenotype selection from various populations
solvent-negative mutants were selected because of sur- according to life cycle-associated events.
vival and a colorimetric alcohol assay was employed as
well [98,109,110]. Furthermore, the streptococcal trans- Other microbial butanol producers
poson Tn916 was used for C. acetobutylicum to isolate Because of the portfolio of available physiological and
mutants defective in solvent formation, the first example bioinformatic data, as well as a broad range of genetic
of the employment of defined populations which allow to tools, well-studied microorganisms such as E. coli provide
trace back responsible genes [111–113]. an excellent scientific platform for biofuel production.
Such heterologous approaches do not only allow detailed
The second example of screening defined C. acetobutyli- analytical methods without native regulatory constraints,
cum populations was published more recently by Borden many prospects for further genetic manipulation and
and Papoutsakis [114]. Plasmid-based genomic libraries metabolic engineering are provided. However, such
were generated and homologously expressed in C. acet- laboratory strains often lack important traits, that is good
obutylicum, which were subjected to butanol challenges to growth rates and prototrophy, as well as substrate and/or
enrich those plasmids which conferred enhanced butanol product tolerance, which makes them less appropriate for
tolerance. Among the sixteen enriched genes, overex- industrial applications. Therefore, the demand of strain
pression of the CAC1869-encoded transcriptional regu- robustness for large-scale production often contradicts
lator was verified to enhance butanol tolerance, although with the benefits to specifically engineer the microbes
details on the mechanism and the physiological role on the molecular level. C. acetobutylicum comprises both,
remain to be elucidated [114]. The suitability of this above-mentioned benefits and disadvantages: as demon-
combinatorial overexpression strategy for disclosing inter- strated by industrial ABE fermentation, C. acetobutylicum
esting candidate targets was confirmed by optimizing and and other solventogenic clostridia have been proven to
extending the approach: screening the genomic library for constitute robust production strains. Moreover, they have
butyrate tolerance led to the identification of non-coding obviously evolved mechanisms to maintain viability in an
RNAs mediating improved carboxylic acid tolerance increasingly toxic environment, that is to survive inher-
[115]. ently toxic butanol concentrations of up to 2% until the
cells turn to endurable spores to escape the stressful
Whereas in general the choice of appropriate populations conditions. On the other hand, detailed analytical and
is only restricted to the particular scientific claims, the metabolic engineering methods are just about to be
availability of suitable screening procedures largely established — still being far behind from other model
depends on the phenotype of interest. Regarding butanol, organisms, but highly promising for the near future.
ethanol and other colorless small molecules, screening
and phenotype selection in a high-throughput manner Since solventogenic clostridia represent the focus of this
according to the product quantities can only be performed review, recent attempts for recombinant butanol pro-
indirectly, that is the product of interest must be visual- duction are summarized only briefly. The clostridial
ized physically or (bio-) chemically, such as using chemi- butanol pathway was reconstructed in E. coli [119,120],
cal derivatization or (bio-) indicators like fluorescence or S. cerevisiae [121], Bacillus subtilis and Pseudomonas putida
auxotrophy applications [116]. Such quantitative screen- [122]. Considering butanol tolerance features, lactic acid
ing techniques are not available yet for C. acetobutylicum bacteria might present better production hosts [123,124]
and other solventogenic strains, but considering the and respective genes of C. acetobutylicum were successfully
promising potential of explorative strategies, global introduced into Lactobacillus brevis [125]. Another inter-
approaches for significant phenotype improvements can esting host is the homoacetogen C. ljungdahlii because of
certainly be expected in the future. its ability to utilize CO2/CO and H2 as substrates, and
traces of butanol were detected in recombinant cells
An innovative novel screening technique was described expressing the C. acetobutylicum butanol biosynthetic
by Tracy et al. [117], who developed a flow cytometry pathway [126]. All of these heterologous butanol produ-
assay for analyzing cell types of C. acetobutylicum, cers provide a scientific ‘proof of principle’, but butanol
applicable for high-throughput screenings on a single cell production rates were significantly below 1 g/l and are
level with optional fluorescence-assisted cell sorting thus far not competitive to solventogenic clostridia, which
(FACS) [117,118]. Interestingly, the authors observed naturally produce butanol at concentrations of approxi-
a stronger correlation between solvent production and the mately 13 g/l (without technical product removal during
vegetative cell type than the clostridial form type (Figure fermentation). However, further investigations, process
2b). However, this finding requires further investigation optimization, metabolic engineering and synthetic

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 643

biology approaches can sustainably improve recombinant Although 25 years old, this review nicely outlines the details on the
physiology and metabolic stoichiometry of Clostridium acetobutylicum.
butanol production in the future [127–129]. By far the most cited review on ABE fermentation until today.
2. Dürre P: Biobutanol: an attractive biofuel. Biotechnol J 2007,
Finally, as an innovatively different approach, the 2- 2:1525-1534.
ketoacid pathway for alternative biofuel production in 3. Dürre P: Fermentative butanol production: bulk chemical and
E. coli was developed by the laboratory of J. C. Liao to biofuel. Ann NY Acad Sci 2008, 1125:353-362.
synthesize various alcohols via a non-natural pathway 4. Lee SY, Park JH, Jang SH, Nielsen LK, Kim J, Jung KS:
[130,131]. Several interesting reviews on general issues  Fermentative butanol production by clostridia. Biotechnol
Bioeng 2008, 101:209-228.
of recombinant biofuel production were published Detailed review on various aspects of the ABE fermentation in general,
recently (e.g., [23,24,132–134]). with an emphasis on the urgent need of systems biology approaches.
5. Gheshlaghi R, Scharer JM, Moo-Young M, Chou CP: Metabolic
Conclusions pathways of clostridia for producing butanol. Biotechnol Adv
2009, 27:764-781.
The conversion of the classic ABE fermentation into a
6. Zheng YN, Li LZ, Xian M, Ma YJ, Yang JM, Xu X, He DZ: Problems
single product, that is butanol, process is a prerequisite with the microbial production of butanol. J Ind Microbiol
for a successful industrial revival. C. acetobutylicum as well Biotechnol 2009, 36:1127-1138.
as all other related bacteria harbors complex metabolic 7. Huang H, Liu H, Gan YR: Genetic modification of critical
pathways with several branching points which makes it enzymes and involved genes in butanol biosynthesis from
biomass. Biotechnol Adv 2010, 28:651-657.
difficult to direct the carbon flow exclusively to butanol.
The achievements in the past few years with respect to 8. Zverlov VV, Berezina O, Velikodvorskaya GA, Schwarz WH:
Bacterial acetone and butanol production by industrial
the development of genetic tools are very promising for fermentation in the Soviet Union: use of hydrolyzed
sustainable metabolic engineering strategies and the data agricultural waste for biorefinery. Appl Microbiol Biotechnol
2006, 71:587-597.
gathered by ‘omics’ technologies allowed deeper insights
into the physiology of C. acetobutylcum. However, a major 9. Ni Y, Sun Z: Recent progress on industrial fermentative
production of acetone–butanol–ethanol by Clostridium
drawback is the lack of knowledge on how the metabolic acetobutylicum in China. Appl Microbiol Biotechnol 2009,
shift from acid to solvent production is regulated on the 83:415-423.
molecular level, for example, what are the inducing 10. Kharkwal S, Karimi IA, Chang MW, Lee DY: Strain improvement
signals, which regulators are involved, how do they  and process development for biobutanol production. Rec Pat
Biotechnol 2009, 3:202-210.
interact, and how are the regulatory networks connected. Interesting review with an exclusive focus on recent patent applications.
Therefore, which products in which quantities are pro- 11. Liu S, Qureshi N: How microbes tolerate ethanol and butanol.
duced by C. acetobutylicum is a question of not only which Nat Biotechnol 2009, 26:117-121.
genes are present, but also how the carbon and electron 12. Ezeji T, Milne C, Price ND, Blaschek HP: Achievements and
flow is regulated to maintain the redox balance. The  perspectives to overcome the poor solvent resistance in
acetone and butanol-producing microorganisms. Appl
regulation is governed by three major issues: first, a Microbiol Biotechnol 2010, 85:1697-1712.
balanced redox status is of crucial importance for the A recent review focussed on butanol toxicity issues. Besides an update
anaerobe C. acetobutylicum; second, the energy yield must on the authors’ expertise on fermentation process engineering, metabolic
engineering efforts on innate and heterologous butanol producers to
be as efficient as possible; and third, the survival of the improve solvent resistance are compiled.
self-poisoning fermentation products must be ensured. 13. Nicolaou SA, Gaida SM, Papoutsakis ET: A comparative
Numerous experimental examples have shown that C. view of metabolite and substrate stress and tolerance in
acetobutylicum can be obliged to alter its product spectrum microbial bioprocessing: From biofuels and chemicals, to
biocatalysis and bioremediation. Metab Eng 2010,
to favor butanol production, and implementing the 12:307-331.
respective molecular mechanisms will most likely reveal 14. Ezeji TC, Qureshi N, Blaschek HP: Butanol fermentation
new perspectives for metabolic engineering approaches. research: upstream and downstream manipulations. Chem
Rec 2004, 4:305-314.
We are optimistic that a butanol-only clostridial strain
can be generated. Whether inactivation of byproduct 15. Ezeji TC, Qureshi N, Blaschek HP: Bioproduction of butanol from
biomass: from genes to bioreactors. Curr Opin Biotechnol 2007,
formation, improved strain robustness, abandonment 18:220-227.
of sporulation, tuning global regulators, etc. or combi-
16. Qureshi N, Ezeji TC: Butanol, ‘a superior biofuel’ production
nations thereof will lead to superior butanol producing from agricultural residues (renewable biomass): Recent
phenotypes offer challenging questions for metabolic progress in technology. Biofuels Bioprod Biorefin 2008,
2:319-330.
engineers.
17. Dürre P, Hollergschwandner C: Initiation of endospore formation
in Clostridium acetobutylicum. Anaerobe 2004, 10:69-74.
References and recommended reading
Papers of particular interest, published within the period of review, 18. Paredes CJ, Alsaker KV, Papoutsakis ET: A comparative
have been highlighted as:  genomic view of clostridial sporulation and physiology. Nat
Rev Microbiol 2005, 3:969-978.
 of special interest Thus far a unique review on clostridial sporulation and related genomics.
 of outstanding interest
19. Lynd LR, Weimer PJ, van Zyl WH, Pretorius IS: Microbial
 cellulose utilization: fundamentals and biotechnology.
1. Jones DT, Woods DR: Acetone-butanol fermentation revisited. Microbiol Mol Biol Rev 2002, 66:506-577.
 Microbiol Rev 1986, 50:484-524. A very detailed review on the mechanisms of cellulose degradation.

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


644 Tissue, cell and pathway engineering

20. Lynd LR, van Zyl WH, McBride JE, Laser M: Consolidated 37. Zhao Y, Tomas CA, Rudolph FB, Papoutsakis ET, Bennett GN:
bioprocessing of cellulosic biomass: an update. Curr Opin Intracellular butyryl phosphate and acetyl phosphate
Biotechnol 2005, 16:577-583. concentrations in Clostridium acetobutylicum and their
implications for solvent formation. Appl Environ Microbiol 2005,
21. Demain AL, Newcomb M, Wu JHD: Cellulase, clostridia, and 71:530-537.
ethanol. Microbiol Mol Biol Rev 2005, 69:124-154.
38. Jones SW, Paredes CJ, Tracy B, Cheng N, Sillers R, Senger RS,
22. Fontes CM, Gilbert HJ: Cellulosomes: highly efficient  Papoutsakis ET: The transcriptional program underlying the
 nanomachines designed to deconstruct plant cell wall physiology of clostridial sporulation. Genome Biol 2008,
complex carbohydrates. Annu Rev Biochem 2010, 79:655-681. 9:R114.
An appropriate update of [19]. The most detailed transcriptome analyses on C. acetobutylicum batch
cultures thus far, focussing on life cycle-related mechanisms with parti-
23. Alper H, Stephanopoulos G: Engineering for biofuels: exploiting cular emphasis on the molecular sporulation events.
 innate microbial capacity or importing biosynthetic potential?
Nat Rev Microbiol 2009, 7:715-723. 39. Tracy BP, Jones SW, Papoutsakis ET: Inactivation of sE and sG
An expert’s view on fundamental consolidated bioprocessing issues. in Clostridium acetobutylicum illuminates their roles in
clostridial-cell form biogenesis, granulose synthesis,
24. Weber C, Farwick A, Benisch F, Brat D, Dietz H, Subtil T, Boles E: solventogenesis, and spore morphogenesis. J Bacteriol 2011
Trends and challenges in the microbial production of doi: 10.1128/JB.01380-10.
lignocellulosic bioalcohol fuels. Appl Microbiol Biotechnol 2010,
87:1303-1315. 40. Grupe H, Gottschalk G: Physiological events in Clostridium
acetobutylicum during the shift from acidogenesis to
25. Papoutsakis ET: Engineering solventogenic clostridia. Curr solventogenesis in continous culture and presentation
 Opin Biotechnol 2008, 19:420-429. of a model for shift induction. Appl Environ Microbiol 1992,
Excellent state-of-the-art review: it concisely summarizes major strate- 58:3896-3902.
gies for sustainable metabolic engineering of clostridial butanol produc-
tion. 41. Rao G, Ward PJ, Mutharasan R: Manipulation of end-product
distribution in strict anaerobes. Ann NY Acad Sci 1987,
26. Mitchell WJ, Albasheri KA, Yazdanian M: Factors affecting 506:76-83.
utilization of carbohydrates by clostridia. FEMS Microbiol Rev
1995, 17:317-329. 42. Girbal L, Soucaille P: Regulation of Clostridium acetobutylicum
metabolism as revealed by mixed-substrate steady-state
27. Servinsky MD, Kiel JT, Dupuy NF, Sund CJ: Transcriptional continuous cultures: role of NADH/NAD ratio and ATP pool.
analysis of differential carbohydrate utilization by Clostridium J Bacteriol 1994, 176:6433-6438.
acetobutylicum. Microbiology 2010, 156:3478-3491.
43. Girbal L, Croux C, Vasconcelos I, Soucaille P: Regulation of
28. Grimmler C, Held C, Liebl W, Ehrenreich A: Transcriptional metabolic shifts in Clostridium acetobutylicum ATCC 824.
analysis of catabolite repression in Clostridium FEMS Microbiol Rev 1995, 17:287-297.
acetobutylicum growing on mixtures of D-glucose and D-
44. Girbal L, Soucaille P: Regulation of solvent production in
xylose. J Biotechnol 2010, 150:315-323.
 Clostridium acetobutylicum. Trends Biotechnol 1998, 16:1-16.
29. Ren C, Gu Y, Hu S, Wu Y, Wang P, Yang Y, Yang C, Yang S, This group did some pioneering work on the redox balance of C.
Jiang W: Identification and inactivation of pleiotropic regulator acetobutylicum’s metabolism in the past which is summarized in this
CcpA to eliminate glucose repression of xylose utilization in brief review. Unfortunately, it is the authors’ most recent journal review.
Clostridium acetobutylicum. Metab Eng 2010, 12:446-454. 45. Nakayama S, Kosaka T, Hirakawa H, Matsuura K, Yoshino S,
30. Bahl H, Gottschalk G: Parameters affecting solvent production Furukawa K: Metabolic engineering for solvent productivity by
by Clostridium acetobutylicum in continuous culture. downregulation of the hydrogenase gene cluster hupCBA in
Biotechnol Bioeng Symp 1984, 14:215-223. Clostridium saccharoperbutylacetonicum strain N1–4. Appl
Microbiol Biotechnol 2008, 78:483-493.
31. Bahl H, Gottwald M, Kuhn A, Rale, Andersch W, Gottschalk G: 46. Schut GJ, Adams MW: The iron-hydrogenase of Thermotoga
Nutritional factors affecting the ratio of solvents produced by
maritima utilizes ferredoxin and NADH synergistically: a new
Clostridium acetobutylicum. Appl Environ Microbiol 1986, perspective on anaerobic hydrogen production. J Bacteriol
52:169-172.
2009, 191:4451-4457.
32. Dürre P: Formation of solvents in clostridia. In Handbook on 47. Hedderich R, Forzi L: Energy-converting [NiFe]-hydrogenases:
Clostridia. Edited by Dürre P. Boca Raton, FL: CRC Press; 2005: more than just H2 activation. J Mol Microbiol Biotechnol 2005,
671-693. 10:92-104.
33. Chen JS: Alcohol dehydrogenase: multiplicity and relatedness 48. Calusinska M, Happe T, Joris B, Wilmotte A: The surprising
in the solvent-producing clostridia. FEMS Microbiol Rev 1995,  diversity of clostridial hydrogenases: a comparative genomic
17:263-273. perspective. Microbiology 2010, 156:1575-1588.
A nice review which comprehensively summarizes current knowledge on
34. Fontaine L, Meynial-Salles I, Girbal L, Yang X, Croux C, clostridial hydrogenases based on genomic data and recent literature.
 Soucaille P: Molecular characterization and transcriptional
analysis of adhE2, the gene encoding the NADH-dependent 49. Seedorf H, Fricke WF, Veith B, Brüggemann H, Liesegang H,
aldehyde/alcohol dehydrogenase responsible for butanol Strittmatter A, Miethke M, Buckel W, Hinderberger J, Li F et al.: The
production in alcohologenic cultures of Clostridium genome of Clostridium kluyveri, a strict anaerobe with unique
acetobutylicum ATCC 824. J Bacteriol 2002, metabolic features. Proc Nat Acad Sci USA 2008, 105:2128-
184:821-830. 2133.
This study first provided experimental evidence on the function of the
bifunctional aldehyde/alcohol dehydrogenase 2 (AdhE2, CAP0035) in 50. Li F, Hinderberger J, Seedorf H, Zhang J, Buckel W, Thauer RK:
cultures of C. acetobutyliucm with clearly reduced redox potential, the  Coupled ferredoxin and crotonyl coenzyme A (CoA) reduction
so-called alcohologenic cultures. with NADH catalyzed by the butyryl-CoA dehydrogenase/Etf
complex from Clostridium kluyveri. J Bacteriol 2008, 190:843-
35. Grimmler C, Janssen H, Krauße D, Fischer RJ, Bahl H, Dürre P, 850.
Liebl W, Ehrenreich A: Genome-wide gene expression analysis This and the following Ref. [51] were important contributions for the
of the switch between acidogenesis and solventogenesis in understanding of the energetic coupling and electron bifurcation mechan-
continuous cultures of Clostridium acetobutylicum. J Mol isms in C. kluyveri.
Microbiol Biotechnol 2011, 20:1-15.
51. Wang S, Huang H, Moll J, Thauer RK: NADP+ reduction with
36. Jones DT, van der Weshuizen A, Long S, Allcock ER, Reid SJ,  reduced ferredoxin and NADP+ reduction with NADH are
Woods DR: Solvent production and morphological changes in coupled via an electron bifurcating enzyme complex in
Clostridium acetobutylicum. Appl Environ Microbiol 1982, Clostridium kluyveri. J Bacteriol 2010, 192:5115-5123.
43:1434-1439. See comment on [50].

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 645

52. Herrmann G, Jayamani E, Mai G, Buckel W: Energy conservation 67. Janssen H, Döring C, Ehrenreich A, Voigt B, Hecker M, Bahl H,
 via electron-transferring flavoprotein in anaerobic bacteria. J  Fischer RJ: A proteomic and transcriptional view of acidogenic
Bacteriol 2008, 190:784-791. and solventogenic steady-state cells of Clostridium
Brilliant review connecting recent findings with biochemical knowledge acetobutylicum in a chemostat culture. Appl Microbiol
on anaerobic energy metabolism. Biotechnol 2010, 87:2209-2226.
Comprehensive proteome study of acetogenesis and solventogenesis
53. Biegel E, Schmidt S, Müller V: Genetic, immunological and providing respective reference maps for C. acetobutylicum using
biochemical evidence for a Rnf complex in the acetogen steady-state cells. The data were accompanied by detailed transcriptome
Acetobacterium woodii. Environ Microbiol 2009, 11:1438-1443. analyses and provide an excellent basis for future systems biology
approaches due to the high degree of reproducibility of chemostat cultures.
54. Biegel E, Müller V: Bacterial Na+-translocating ferredoxin:NAD+
 oxidoreductase. Proc Natl Acad Sci USA 2010, 107:18138- 68. Amador-Noguez D, Feng XJ, Fan J, Roquet N, Rabitz H,
18142.  Rabinowitz JD: Systems-level metabolic flux profiling
This recent study provided detailed biochemical evidence for the ener- elucidates a complete, bifurcated TCA cycle in Clostridium
getic coupling of ferredoxin oxidation and sodium ion translocation in acetobutylicum. J Bacteriol 2010, 192:4452-4461.
Acetobacterium woodii. Simultaneously to [69], metabolomic studies provided important insights
into the central carbon metabolism of C. acetobutylicum.
55. Nölling J, Breton G, Omelchenko MV, Makarova KS, Zeng Q,
 Gibson R, Lee HM, Dubois J, Qiu D, Hitti J et al.: Genome 69. Crown SB, Indurthi DC, Ahn WS, Choi J, Papoutsakis ET,
sequence and comparative analysis of the solvent-producing  Antoniewicz MR: Resolving the TCA cycle and pentose-
bacterium Clostridium acetobutylicum. J Bacteriol 2001, phosphate pathway of Clostridium acetobutylicum ATCC 824:
183:4823-4838. isotopomer analysis, in vitro activities and expression
The early publication of the C. acetobutylicum genome sequence paved analysis. Biotechnol J 2010 doi: 10.1002/biot.201000282.
the way for the current knowledge of this unique microorganism. See comment on [68].
56. Tomas CA, Alsaker KV, Bonarius HP, Hendriksen WT, Yang H, 70. Papoutsakis ET: Equations and calculations for fermentations
Beamish JA, Paredes CJ, Papoutsakis ET: DNA array-based of butyric acid bacteria. Biotechnol Bioeng 1984, 26:174-187.
transcriptional analysis of asporogenous, nonsolventogenic
Clostridium acetobutylicum strains SKO1 and M5. J Bacteriol 71. Desai RP, Nielsen LK, Papoutsakis ET: Stoichiometric modeling
2003, 185:4539-4547. of Clostridium acetobutylicum fermentations with non-linear
constraints. J Biotechnol 1999, 71:191-205.
57. Alsaker KV, Paredes CJ, Papoutsakis ET: Design, optimization
and validation of genomic DNA microarrays for examining the 72. Shinto H, Tashiro Y, Yamashita M, Kobayashi G, Sekiguchi T,
Clostridium acetobutylicum transcriptome. Biotechnol Hanai T, Kuriya Y, Okamoto M, Sonomoto K: Kinetic modeling
Bioprocess Eng 2005, 10:432-443. and sensitivity analysis of acetone–butanol–ethanol
production. J Biotechnol 2007, 131:45-56.
58. Paredes CJ, Senger RS, Spath IS, Borden JR, Sillers R,
Papoutsakis ET: A general framework for designing and 73. Shinto H, Tashiro Y, Kobayashi G, Sekiguchi T, Hanai T, Kuriya Y,
validating oligomer-based DNA microarrays and its Okamoto M, Sonomoto K: Kinetic study of substrate
application to Clostridium acetobutylicum. Appl Environ dependency for higher butanol production in acetone–
Microbiol 2007, 73:4631-4638. butanol–ethanol fermentation. Proc Biochem 2008, 43:1452-
1461.
59. Hillmann F, Döring C, Riebe O, Ehrenreich A, Fischer RJ, Bahl H:
 The role of PerR in O2-affected gene expression of Clostridium 74. Lee J, Yun H, Feist AM, Palsson BØ, Lee SY: Genome-scale
acetobutylicum. J Bacteriol 2009, 191:6082-6093. reconstruction and in silico analysis of the Clostridium
First global transcriptional view on the oxidative stress response of C. acetobutylicum ATCC 824 metabolic network. Appl Microbiol
acetobutylicum and its aerotolerant PerR mutant. Biotechnol 2008, 80:849-862.

60. Shi Z, Blaschek HP: Transcriptional analysis of Clostridium 75. Senger RS, Papoutsakis ET: Genome-scale model for
beijerinckii NCIMB 8052 and the hyper-butanol-producing Clostridium acetobutylicum. Part I. Metabolic network
mutant BA101 during the shift from acidogenesis to resolution and analysis. Biotechnol Bioeng 2008,
solventogenesis. Appl Environ Microbiol 2008, 101:1036-1052.
74:7709-7714. 76. Senger RS, Papoutsakis ET: Genome-scale model for
Clostridium acetobutylicum. Part II. Development of specific
61. Hemme CL, Mouttaki H, Lee YJ, Zhang G, Goodwin L, Lucas S,
proton flux states and numerically determined sub-systems.
Copeland A, Lapidus A, Glavina del Rio T, Tice H et al.:
Biotechnol Bioeng 2008, 101:1053-1071.
Sequencing of multiple clostridial genomes related to
biomass conversion and biofuels production. J Bacteriol 2010, 77. Haus S, Jabbari S, Millat T, Janssen H, Fischer RJ, Bahl H, King JR,
192:6494-6496. Wolkenhauer O: A systems biology approach to investigate the
effect of pH-induced gene regulation on solvent production by
62. Schaffer S, Isci N, Zickner B, Dürre P: Changes in protein Clostridium acetobutylicum in continuous culture. BMC Syst
 synthesis and identification of proteins specifically induced Biol 2011, 5:10.
during solventogenesis in Clostridium acetobutylicum.
Electrophoresis 2002, 23:110-121. 78. Senger RS: Biofuel production improvement with genome-
First proteome analysis on the solventogenic physiology of C. acetobu-  scale models: the role of cell composition. Biotechnol J 2010,
tylicum. 5:671-685.
This review concisely discusses the current computational models for C.
63. Sullivan L, Bennett GN: Proteome analysis and comparison of acetobutylicum, understandable also for non-bioinformatic researchers.
Clostridium acetobutylicum ATCC 824 and Spo0A strain
variants. J Ind Microbiol Biotechnol 2006, 33:298-308. 79. Mermelstein LD, Papoutsakis ET: In vivo methylation in
Escherichia coli by the Bacillus subtilis phage phi3TI
64. Alsaker KV, Spitzer TR, Papoutsakis ET: Transcriptional analysis methyltransferase to protect plasmids from restriction upon
of spo0A overexpression in Clostridium acetobutylicum and transformation of Clostridium acetobutylicum ATCC 824. Appl
its effect on the cell’s response to butanol stress. J Bacteriol Environ Microbiol 1993, 59:1077-1081.
2004, 186:1959-1971.
80. Soucaille P, Figge G, Croux C: Process for chromosomal
65. Mao S, Luo Y, Zhang T, Li J, Bao G, Zhu Y, Chen Z, Zhang Y, Li Y, integration and DNA sequence replacement in clostridia.
Ma Y: Proteome reference map and comparative proteomic International patent WO 2008/040387.
analysis between a wild type Clostridium acetobutylicum DSM
1731 and its mutant with enhanced butanol tolerance and 81. Dong H, Zhang Y, Dai Z, Li Y: Engineering Clostridium strain to
butanol yield. J Proteome Res 2010, 9:3046-3061. accept unmethylated DNA. PLoS One 2010, 5:e9038.
66. Tomas CA, Beamish J, Papoutsakis ET: Transcriptional analysis 82. Heap JT, Pennington OJ, Cartman ST, Minton NP: A modular
of butanol stress and tolerance in Clostridium acetobutylicum.  system for Clostridium shuttle plasmids. J Microbiol Methods
J Bacteriol 2004, 186:2006-2018. 2009, 78:79-85.

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647


646 Tissue, cell and pathway engineering

The authors established a very useful shuttle plasmid collection for 99. Stim-Herndon KP, Nair RV, Papoutsakis ET, Bennett GN: Analysis
simplified gene cloning and expression in C. acetobutylicum and other of degenerate variants of Clostridium acetobutylicum ATCC
clostridia with a wide range of application-dependent opportunities. 824. Anaerobe 1996, 2:11-18.
83. Heap JT, Pennington OJ, Cartman ST, Carter GP, Minton NP: The 100. Cornillot E, Nair RV, Papoutsakis ET, Soucaille P: The genes for
 ClosTron: a universal gene knock-out system for the genus butanol and acetone formation in Clostridium acetobutylicum
Clostridium. J Microbiol Methods 2007, 70:452-464. ATCC 824 reside on a large plasmid whose loss leads to
First publication on a reliable and reproducible method for targeted and degeneration of the strain. J Bacteriol 1997, 179:5442-5447.
stable gene inactivation in C. acetobutylicum and other clostridia.
101. Nair RV, Papoutsakis ET: Expression of plasmid-encoded aad in
84. Shao L, Hu S, Yang Y, Gu Y, Chen J, Yang Y, Jiang W, Yang S: Clostridium acetobutylicum M5 restores vigorous butanol
Targeted gene disruption by use of a group II intron (targetron) production. J Bacteriol 1994, 176:5843-5846.
vector in Clostridium acetobutylicum. Cell Res 2007,
17:963-965. 102. Lee JY, Jang YS, Lee J, Papoutsakis ET, Lee SY: Metabolic
engineering of Clostridium acetobutylicum M5 for highly
85. Heap JT, Kuehne SA, Ehsaan M, Cartman ST, Cooksley CM, selective butanol production. Biotechnol J 2009,
Scott JC, Minton NP: The ClosTron: mutagenesis in Clostridium 4:1432-1440.
refined and streamlined. J Microbiol Methods 2010,
80:49-55. 103. Cai X, Bennett GN: Improving the Clostridium acetobutylicum
butanol fermentation by engineering the strain for co-
86. Sillers R, Chow A, Tracy B, Papoutsakis ET: Metabolic production of riboflavin. J Ind Microbiol Biotechnol 2010 doi:
engineering of the non-sporulating, non-solventogenic 10.1007/s10295-010-0875-6.
Clostridium acetobutylicum strain M5 to produce butanol
without acetone demonstrate the robustness of the acid- 104. Bailey JE, Sburlati A, Hatzimanikatis V, Lee K, Renner WA, Tsai PS:
formation pathways and the importance of the electron Inverse metabolic engineering: a strategy for directed genetic
balance. Metab Eng 2008, 10:321-332. engineering of useful phenotypes. Biotechnol Bioeng 2002,
79:568-579.
87. Tracy BP, Papoutsakis ET: Methods and compositions for
genetically engineering clostridia species. International patent 105. Santos CN, Stephanopoulos G: Combinatorial engineering of
WO 2009/137778. microbes for optimizing cellular phenotype. Curr Opin Chem
Biol 2008, 12:168-176.
88. Heap JT, Minton NP: Methods. International patent WO 2009/
106. Hermann M, Fayolle F, Marchal R, Podvin L, Sebald M,
101400.
Vandecasteele JP: Isolation and characterization of butanol-
89. Cartman ST, Minton NP: Method of double crossover resistant mutants of Clostridium acetobutylicum. Appl Environ
homologous recombination in clostridia. International patent Microbiol 1985, 50:1238-1243.
WO 2010/084349. 107. Matta-el-Ammouri G, Janati-Idrissi R, Rambourg JM,
90. Desai RP, Papoutsakis ET: Antisense RNA strategies for Petitdemange H, Gay R: Acetone butanol fermentation by a
metabolic engineering of Clostridium acetobutylicum. Appl Clostridium acetobutylicum mutant with high solvent
Environ Microbiol 1999, 65:936-945. productivity. Biomass 1986, 10:109-119.

91. Green EM, Boynton ZL, Harris LM, Rudolph FB, Papoutsakis ET, 108. Qureshi N, Blaschek HP: Recent advances in ABE fermentation:
Bennett GN: Genetic manipulation of the acid formation hyper-butanol producing Clostridium beijerinckii BA101. J Ind
pathways by gene inactivation in Clostridium acetobutylicum Microbiol Biotechnol 2001, 27:287-291.
ATCC 824. Microbiology 1996, 142:2079-2086. 109. Dürre P, Kuhn A, Gottschalk G: Treatment with allyl alcohol
selects specifically for mutants of Clostridium acetobutylicum
92. Tummala SB, Welker NE, Papoutsakis ET: Design of antisense
defective in butanol synthesis. FEMS Microbiol Lett 1986,
RNA constructs for downregulation of the acetone formation
36:77-81.
pathway of Clostridium acetobutylicum. J Bacteriol 2003,
185:1923-1934. 110. Rogers P, Palosaari N: Clostridium acetobutylicum mutants
that produce butyraldehyde and altered quantities of solvents.
93. Tummala SB, Junne SG, Papoutsakis ET: Antisense RNA Appl Environ Microbiol 1987, 53:2761-2766.
downregulation of coenzyme A transferase combined with
alcohol-aldehyde dehydrogenase overexpression leads to 111. Bertram J, Dürre P: Conjugal transfer and expression of
predominantly alcohologenic Clostridium acetobutylicum streptococcal transposons in Clostridium acetobutylicum.
fermentations. J Bacteriol 2003, 185:3644-3653. Arch Microbiol 1989, 151:551-557.
94. Sillers R, Al-Hinai MA, Papoutsakis ET: Aldehyde-alcohol 112. Bertram J, Kuhn A, Dürre P: Tn916-induced mutants of
dehydrogenase and/or thiolase overexpression coupled with Clostridium acetobutylicum defective in regulation of solvent
CoA transferase downregulation lead to higher alcohol titers formation. Arch Microbiol 1990, 153:373-377.
and selectivity in Clostridium acetobutylicum fermentations.
Biotechnol Bioeng 2008, 102:38-49. 113. Babb BL, Collett HJ, Reid SJ, Woods DR: Transposon
mutagenesis of Clostridium acetobutylicum P262:
95. Jiang Y, Xu C, Dong F, Yang Y, Jiang W, Yang S: Disruption of the isolation and characterization of solvent deficient and
 acetoacetate decarboxylase gene in solvent-producing metronidazole resistant mutants. FEMS Microbiol Lett 1993,
Clostridium acetobutylicum increases the butanol ratio. Metab 114:343-348.
Eng 2009, 11:284-291.
The first example of engineering the central fermentative metabolism of C. 114 . Borden JR, Papoutsakis ET: Dynamics of genomic-library
acetobutylicum using the Targetron technology.  enrichment and identification of solvent-tolerant genes in
Clostridium acetobutylicum. Appl Environ Microbiol 2007,
96. Soucaille P: Process for the biological production of n-butanol 73:3061-3068.
at high yield. International patent WO 2008/052973. This unconventional approach was only the second combinatorial exam-
ple for screening defined C. acetobutylicum populations to allow retracing
97. Meinecke B, Bahl H, Gottschalk G: Selection of an the phenotype-related genotype.
asporogenous strain of Clostridium acetobutylicum in
continous culture under phosphate limitation. Appl Environ 115. Borden JR, Jones SW, Indurthi D, Chen Y, Papoutsakis ET: A
Microbiol 1984, 48:1064-1065. genomic-library based discovery of a novel, possibly
synthetic, acid-tolerance mechanism in Clostridium
98. Clark SW, Bennett GN, Rudolph FB: Isolation and acetobutylicum involving non-coding RNAs and ribosomal
characterization of mutants of Clostridium acetobutylicum RNA processing. Metab Eng 2010, 12:268-281.
ATCC 824 deficient in acetoacetyl-coenzyme A:acetate/
butyrate:coenzyme A-transferase (EC 2.8.3.9) and in other 116 . Dietrich JA, McKee AE, Keasling JD: High-throughput metabolic
solvent pathway Enzymes. Appl Environ Microbiol 1989,  engineering: advances in small-molecule screening and
55:970-976. selection. Annu Rev Biochem 2010, 79:563-590.

Current Opinion in Biotechnology 2011, 22:634–647 www.sciencedirect.com


Metabolic engineering of Clostridium acetobutylicum Lütke-Eversloh and Bahl 647

A nicely written review on the necessity of developing suitable high- 125. Berezina OV, Zakharova NV, Brandt A, Yarotsky SV, Schwarz WH,
throughput screening techniques for combinatorial metabolic engineer- Zverlov VV: Reconstructing the clostridial n-butanol metabolic
ing strategies: examples, limits and perspectives are provided. pathway in Lactobacillus brevis. Appl Microbiol Biotechnol
2010, 87:635-646.
117. Tracy BP, Gaida SM, Papoutsakis ET: Development and
 application of flow-cytometric techniques for analyzing and 126. Köpke M, Held C, Hujer S, Liesegang H, Wiezer A, Wollherr A,
sorting endospore-forming clostridia. Appl Environ Microbiol Ehrenreich A, Liebl W, Gottschalk G, Dürre P: Clostridium
2008, 74:7497-7506. ljungdahlii represents a microbial production platform based
The most inventive recent publication on the development of a sophis- on syngas. Proc Natl Acad Sci USA 2010, 107:13087-13092.
ticated high-throughput screening method for clostridia based on the life
cycle-associated morphological changes. 127. Fischer CR, Klein-Marcushamer D, Stephanopoulos G: Selection
and optimization of microbial hosts for biofuels production.
118 . Tracy BP, Gaida SM, Papoutsakis ET: Flow cytometry for Metab Eng 2008, 10:295-304.
 bacteria: enabling metabolic engineering, synthetic biology
and the elucidation of complex phenotypes. Curr Opin 128. Dellomonaco C, Rivera C, Campbell P, Gonzalez R: Engineered
Biotechnol 2010, 21:85-99. respiro-fermentative metabolism for the production of
A concise survey on flow cytometry applications from a technical point of biofuels and biochemicals from fatty acid-rich feedstocks.
view, opportunities and challenges for explorative metabolic engineering Appl Environ Microbiol 2010, 76:5067-5078.
approaches are discussed.
129. Ranganathan S, Maranas CD: Microbial 1-butanol production:
119. Inui M, Suda M, Kimura S, Yasuda K, Suzuki H, Toda H, identification of non-native production routes and in silico
Yamamoto S, Okino S, Suzuki N, Yukawa H: Expression of engineering interventions. Biotechnol J 2010, 5:716-725.
Clostridium acetobutylicum butanol synthetic genes in
Escherichia coli. Appl Microbiol Biotechnol 2008, 77:1305-1316. 130 . Atsumi S, Hanai T, Liao JC: Non-fermentative pathways for
 synthesis of branched-chain higher alcohols as biofuels.
120. Atsumi S, Cann AF, Connor MR, Shen CR, Smith KM, Nature 2008, 451:86-89.
Brynildsen MP, Chou KJ, Hanai T, Liao JC: Metabolic Application and genetic engineering of a novel non-natural biosynthetic
engineering of Escherichia coli for 1-butanol production. pathway for second-generation biofuel production in E. coli.
Metab Eng 2008, 10:305-311.
131. Atsumi S, Liao JC: Metabolic engineering for advanced biofuel
121. Steen EJ, Chan R, Prasad N, Myers S, Petzold CJ, Redding A,
production from Escherichia coli. Curr Opin Biotechnol 2008,
Ouellet M, Keasling JD: Metabolic engineering of
19:414-419.
Saccharomyces cerevisiae for the production of n-butanol.
Microb Cell Fact 2008, 7:36. 132. Ghim CM, Kim T, Mitchell RJ, Lee SK: Synthetic biology for
122. Nielsen DR, Leonard E, Yoon SH, Tseng HC, Yuan C, Prather KL: biofuels: Building designer microbes from the scratch.
Engineering alternative butanol production platforms in Biotechnol Bioproc Eng 2010, 15:11-21.
heterologous bacteria. Metab Eng 2009, 11:262-273.
133. Dellomonaco C, Fava F, Gonzalez R: The path for next
123. Knoshaug EP, Zhang M: Butanol tolerance in a selection of generation biofuels: successes and challenges in the era of
microorganisms. Appl Biochem Biotechnol 2009, 153:13-20. synthetic biology. Microb Cell Fact 2010, 9:3.

124. Li J, Zhao JB, Zhao M, Yang YL, Jiang WH, Yang S: Screening and 134. Clomburg JM, Gonzalez R: Biofuel production in Escherichia
characterization of butanol-tolerant microorganisms. Lett coli: the role of metabolic engineering and synthetic biology.
Appl Microbiol 2010, 50:373-379. Appl Microbiol Biotechnol 2010, 86:419-434.

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:634–647

You might also like