Vous êtes sur la page 1sur 7

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Systems Biology

Systems biology of lactic acid bacteria: For food and


thought
Bas Teusink1,2 and Douwe Molenaar1,2

Abstract enormous diversity in species and strains with different


Lactic acid bacteria (LAB) ferment plants, fish, meats and milk functionalities, such as flavour production profiles or
and turn them into tasty food products with increased shelf life; texturing properties. Furthermore, one may change pH
other LAB help digesting food and create a healthy environ- and temperature a bit, or change some ingredients
ment in the intestine. The economic and societal importance of perhaps.
these relatively simple and small bacteria is immense. In this
review we hope to show that their adaptations to nutrient-rich It is also not so easy to decide what to change, or how to
environments provides fascinating and often puzzling behav- change it: foods are chemically complex and undefined,
iours that give rise to many fundamental evolutionary biological and often the fermentation is not carried out by a single
questions in need of a systems biology approach. We will strain, but a complex mixture of LABs. Finally, analyses
provide examples of such questions, compare the (metabolic) in sticky, solid and inhomogeneous food matrices can
behaviour of LAB to that of other model organisms, and pro- be tedious, and they often are. Therefore, in contrast
vide the latest insights, if available. to the industrial biotechnology field that produces
biobased chemicals, largely on the basis of monocultures
Addresses in relatively well-defined growth media, research in
1
Amsterdam Institute for Molecules, Medicines and Systems, Vrije
LAB has much less adopted engineering and systems
Universiteit Amsterdam, O|2 Building, Section Systems Bioinformatics,
Location Code 2E51, De Boelelaan 1085, NL-1081HV Amsterdam, approaches e although metabolic engineering activities
The Netherlands in LAB are ongoing [2e4].
2
Top Institute Food and Nutrition, 6700 AN Wageningen, The
Netherlands We believe this is a pity, from both sides: Systems
Corresponding author: Teusink, Bas (b.teusink@vu.nl)
biology has a lot to offer to the LAB field, and the LABs
have a lot to offer to the systems biology field. LABs
provide questions, challenges, and biological examples
Current Opinion in Systems Biology 2017, 6:7–13 of adaptations to environments rich in nutrients and full
This review comes from a themed issue on Systems biology of model of stress [5]. They can offer interesting and relevant
organisms (2017) cases to test the generality of findings in other, better
Edited by Jens Nielsen and Kiran Raosaheb Patil studied, model organisms, Escherichia coli in particular.
For a complete overview see the Issue and the Editorial
Systems biology on the other hand, can provide struc-
ture to, and understanding of, the complex systems
Available online 18 July 2017
comprising LAB. In this review, we will describe some
http://dx.doi.org/10.1016/j.coisb.2017.07.005 features of LAB physiology that we believe are inter-
2452-3100/© 2017 The Authors. Published by Elsevier Ltd. This is an esting from a systems biology perspective, and we will
open access ar ticle under the CC BY-NC-ND license (http:// describe our current understanding and open questions.
creativecommons.org/licenses/by-nc-nd/4.0/).
We hope this will attract more systems biologists to
these fascinating microorganisms.
Keywords
Lactic acid bacteria, Lactococcus, Lactobacillus, Food fermentation,
Systems biology, Biotechnology. Metabolic adaptation to rich nutrient
environments: auxotrophies and division of
labour
Introduction Environments in which LAB thrive are rich in sugars and
With over 100 billion Euros annually [1], the economic protein; fats, vitamins and nucleotides are also often
value of foods fermented by such small bacteria e LAB available. Consequently, most LAB are auxotrophic for a
have genome sizes of only 2e3 Mbp-is impressive. large number of amino acids and vitamins. The loss of
Yogurt, cheese and sauerkraut, but also breads, hams, function in the presence of some specific nutrients may
and olives, or pickles, soy- and fish sauce require the be caused by genetic drift or provides a selective
metabolic activities of LAB. Many of these foods have a advantage. Recent works make strong cases for the
long history, and the associated industry is rather tradi- latter. It was shown experimentally in E. coli that
tional and wary of revolutionary changes in production introduction of auxotrophies and subsequent exchange
processes, let alone genetic modifications to improve of amino acids provided the auxotrophic mutant an
traits. Rather, the industry takes advantage of the increased growth rate over the wildtype [6]. These traits

www.sciencedirect.com Current Opinion in Systems Biology 2017, 6:7–13


8 Systems biology of model organisms (2017)

are evolvable in laboratory evolution experiments and almost all LAB require either glutamine or glutamate,
make the mutants depend on cross-feeding [7]. because they cannot synthesize its precursor a-keto-
glutarate de novo, by lack of a complete TCA cycle
The differences between LAB species and even strains [11]. Alternatively, the presence of some amino acids
from the same species, however, is large and although it can provide feedback inhibition on the synthesis of
was explained in general by niche adaptation for another amino acid. This happens e.g. for aromatic and
Lactococcus lactis strains [8], the diversity in amino acid branched chain amino acids [14]. Therefore, auxotro-
metabolism and corresponding auxotrophies remains phies predicted from genome-scale metabolic models
puzzling as the environments in which LAB are isolated require careful experimental validation, as recently
often appear equally rich in amino acids, peptides and/ done extensively for Enterococcus faecalis [15] and
or proteins. This matters for LAB applications as their Streptococcus pyogenes [16].
catabolic products are flavour compounds [9], or bitter
or health-promoting [10] peptides. The physiological The similarities between auxotrophies of LAB species
role of such catabolism is not always clear: its wide and humans perhaps suggest an underlying cause or
spread occurrence may be a consequence of our selec- constraint. Methionine or cysteine are always required,
tion for flavour production, but it may also contribute to as is histidine (except for Lactobacillus plantarum); Aro-
stress and energy metabolism [11], chemical warfare, matic and branched chain amino acids, especially valine,
hostemicrobe interactions [12] or possibly geographic are often needed for growth, or at least for fast growth
spreading through attracted insects. [14]. Why these amino acids? Are these the most
expensive to make, or require complex co-factors or vi-
In Table 1 the experimentally-determined auxotro- tamins, and are the genes therefore most easily lost? A
phies for amino acids are shown for a number of rep- comparative study in lactobacilli showed a great di-
resentatives of LAB species. However, defining such versity for different amino acids in true loss of biosyn-
auxotrophies is nontrivial by dependencies on the thetic genes and loss of activity by mutations [17].
presence of other nutrients: also in humans the di- However, the growth of all tested lactobacilli could not
chotomy between essential and non-essential amino be restored without glutamate, even after mutagenesis
acids was questioned and condition-dependent essen- and selective plating. The collective loss by LAB of the
tial amino acids introduced [13]. For example, if ability to synthesize such a key amino acid as glutamate
glutamine is present, glutamate is not required, but is mysterious.

Table 1

Amino acid requirement for selected LAB and man (in black box). Green indicates that growth is possible (but often at a lower
rate) in the absence of the amino acid, red means no or extremely poor growth is observed. Based on studies of specific strains of
S.t. [18], L. l. [19], L.p. [14], S.p. [16], E.f. [16], H.s. [13] and L.a. [17]. Note that auxotrophies between different strains of the same
species can differ.

Current Opinion in Systems Biology 2017, 6:7–13 www.sciencedirect.com


Systems biology of lactic acid bacteria Teusink and Molenaar 9

From diversity in auxotrophies to microbial Yet, subsequent laboratory evolution experiments in


ecology milk showed a loss of the protease and a decrease in
Quite a number of lactobacilli that express proteases and fitness [30]. The trait is evolutionary unstable because
thus can rely on proteolysis, lost the ability to synthesize of cheaters that profit from the peptides released by the
or interconvert many more amino acids. As a result, efforts of others. The loss of protease activity can be
excess amino acids are excreted that depend on the prevented by cultivation at low densities. Extracellular
protein composition, protease specificity and the amino sucrose hydrolysis by invertase expression in yeast
acid interconversion metabolism of the protease positive showed a similar instability [31]. These examples
strain. This in turn may provide specific amino acid demonstrate the need to consider the strategies of
niches for other bacteria. Some LAB-residents of the competitors, and to seriously consider “the tragedy of
human gut even specialized into growth on peptides the commons”, i.e. the idea that common goods drive
derived from gastric proteolysis [20]. And so, perhaps individuals to behaviours that are good for themselves,
the specific auxotrophies we observe today are the result but not for the population as a whole [32].
of co-evolution dynamics and testimonies of diversities
arising by chance and necessity [21] in complex eco- This brings us to the metabolic activity that gave LAB
systems. Systems biology may help explain the necessity their name: the fermentation of sugars to lactic acid. For
[22]. many LAB, lactate is the only major catabolic byproduct,
but for many others, alternative fermentation routes
Auxotrophies introduce diversity and dependencies that exist. Of these “mixed acid fermentation” whereby a
result in ecological interactions such as cross feeding, mixture of acetate, formate and ethanol is produced is
which was readily observed in spatiotemporal models of the most prevalent case (Figure 1A). Lactate fermen-
bacterial communities that mimic the gut [23]. The gut tation extracts only 2 ATP per glucose, whereas mixed
may be too complex and experimentally inaccessible, acid fermentation does slightly better with 3 ATP per
however, for deeper quantitative systems approaches. glucose. L. lactis shows the high-ATP yield mixed acid
Many consortia of LAB (sometimes with yeasts) that fermentation strategy at low growth rates, and switches
turn milk into yogurt or cheese, or wheat into sour- to lactate formation as the growth rate increases [33].
bread, are of a more manageable complexity, between This is reminiscent of the Crabtree effect in yeast [35],
two species for yogurt and in the order of a hundred for or of overflow metabolism E coli [36]. Thus, production
cheese and kefir (Kiran Patil, personal communication) of lactate, associated with fast and wasteful growth,
cultures. Moreover, many of these consortia are stable as seems the result of a tragedy of the commons. Indeed,
they are formed by long histories of back-swapping, and when we privatized the common sugar by propagation in
their fitness may have been optimized [24,25], making water-in-oil emulsions, we could select L. lactis mutants
them interesting model systems for systems-level that switched to mixed acid fermentation [37]. The
studies of ecosystem functioning. mechanism by which mutants achieved this switch,
contains an interesting general lesson: we found muta-
One such study pointed to other causes for community tions in the glucose transport system that we hypothe-
diversity beyond metabolic interactions. For cheese size create an internal state that mimics a low-glucose
starter cultures, the dynamics of strain frequencies environment. Such internal states can subsequently
during serial cultivation in milk was followed [26], and drive metabolic regulation, either through allosteric in-
genetic heterogeneity in the culture was stabilized by teractions or gene expression (see Refs. [38,39] for
frequency-dependent phage predation, coined a “kill- recent examples in E. coli, or [40] in L. lactis).
the-winner” mechanism e possibly a specific case of
frequency-dependent predation known in ecology. This In E. coli, it was recently shown that differences in
mechanism prevents one lineage to take over the pop- “proteome-efficiency” can explain overflow metabolism
ulation. Resistance to phages -or bacteriocins, naturally [36]. Indeed, the perspective of (optimal [25,41])
produced antibiotics-introduce another layer of in- allocation of limited resources has appeared as a
teractions that is extremely relevant for food production powerful determinant of regulatory strategies [42,43].
and evoke fundamental scientific questions, from mo- The central idea is that protein is costly [44] and ATP-
lecular mechanistic [27] to evolutionary [28]. The efficient pathways cost more resources to implement,
function of Crispr as a phage immune response was which at higher growth rates becomes too large a burden
discovered in a LAB: Streptococcus thermophilus [29]. and drives cells to switch to cheaper and less ATP-
efficient pathways [36,42]. We tested this idea by a
careful characterisation of the metabolic switch in
Metabolic adaptation to rich nutrient
L. lactis, and found that the proteome hardly changed
environments: cheating and overflow
with growth rate [33]. Glycolytic enzyme levels
Proteases appear useful in protein-rich environments:
remained largely constant despite a more than threefold
indeed, providing a L. lactis plant isolate with casein
increase in flux. The apparent overcapacity of glycolytic
(milk protein) protease increased fitness substantially.
enzymes at the lower growth rates, also observed for
www.sciencedirect.com Current Opinion in Systems Biology 2017, 6:7–13
10 Systems biology of model organisms (2017)

Bacillus subtilis [45] and yeast [46], is difficult to recon- but why? Structural kinetic modeling of L. lactis central
cile with optimal proteome management. We performed metabolism hinted at a negative control of the acetate
prolonged chemostat cultivation studies at different branch on the glycolytic flux, through its impact on
growth rates, and found that expression of membrane- NADH levels [48]. How this subsequently affects
associated processes was specifically affected, indi- growth rate and fitness is unclear, however, but glyco-
cating that under these conditions uptake of nutrient, lytic flux as biological objective does fit with an old
rather than its further intracellular processing, limits observation that the glycolytic flux in L. lactis appears
growth [Claire Price, Filipe Branco dos Santos, unpub- always on, independent from ATP demand [49]. Alter-
lished]. Genome-scale metabolism and expression natively, we should also keep in mind that resource
modeling in E. coli also suggested a distinction between allocation and the associated bacterial growth laws are
nutrient (or uptake) limitation at low growth rates, and steady state relationships. At constant conditions,
proteome limitation at high growth rates [47]. metabolic regulation is not needed and costly, which
points to a role under dynamic conditions. Recent
What remains puzzling, however, is that in the absence studies have shown how important it is to properly
of an apparent proteome limitation, L. lactis still switches respond to extracellular dynamics [50], and how cellular
to lactic acid formation as growth rate increases [33]. heterogeneity in such responses can create sub-
This strongly suggests a dominant metabolic regulation, populations of responders and non-responders, in

Figure 1

Fermentation pathways in L. lactis to lactate or mixed acid fermentation. (A) Overview of the biochemical reactions, with external metabolites in white
boxes, lumped metabolite pools in grey boxes, and positive (green) and negative (red) regulation by F16bP. (B) Is the metabolic regulation of acetate
versus lactate observed in the chemostat (lower figure, based on [33]) a series of still pictures from a normally fast transition to glucose depletion (upper
figure, based on [34])? During a brief period of glucose depletion, growth rate decreases rapidly, and a shift of lactate towards acetate production takes
place.

Current Opinion in Systems Biology 2017, 6:7–13 www.sciencedirect.com


Systems biology of lactic acid bacteria Teusink and Molenaar 11

L. lactis [51], but also E. coli [52] and Saccharomyces production. Trends Biotechnol 2017, http://dx.doi.org/10.1016/
j.tibtech.2017.05.002.
cerevisiae [53]. If LAB have adapted to conditions of feast
and famine, with relatively fast and frequent transitions 3. Petersen KV, Liu J, Chen J, Martinussen J, Jensen PR, Solem C:
Metabolic characterization and transformation of the non-
between them, metabolic regulation to rapidly switch dairy Lactococcus lactis strain KF147, for production of
fluxes may make sense. One idea (Frank Bruggeman, ethanol from xylose. Biotechnol J 2017, http://dx.doi.org/
10.1002/biot.201700171.
personal communication), is that the titration of growth
rate through the dilution rate in the chemostat may 4. Song AA-L, In LLA, Lim SHE, Rahim RA: A review on Lacto-
coccus lactis: from food to factory. Microb Cell Factories 2017,
provide artificial steady-state still pictures of what 16:55.
normally is a brief transition for the organism 5. Papadimitriou K, Alegría Á, Bron PA, de Angelis M, Gobbetti M,
(Figure 1B). Kleerebezem M, Lemos JA, Linares DM, Ross P, Stanton C,
et al.: Stress physiology of lactic acid bacteria. Microbiol Mol
Biol Rev 2016, 80:837–890.
The molecular mechanism by which metabolic regula-
tion achieves the switch between mixed acid and lactate 6. Pande S, Merker H, Bohl K, Reichelt M, Schuster S, de
Figueiredo LF, Kaleta C, Kost C: Fitness and stability of obli-
fermentation, is also not yet resolved, despite much gate cross-feeding interactions that emerge upon gene loss
research in the (distant) past. Complexity arises from in bacteria. ISME J 2014, 8:953–962.
the involvement of moiety conservations and couplings 7. D’Souza G, Kost C: Experimental evolution of metabolic de-
* pendency in bacteria. PLoS Genet 2016, 12:e1006364.
between enzyme activities through the cofactors Simple but effective set of laboratory evolution experiments in the
NADH/NAD and ATP/ADP [54]. But similar perhaps to presence and absence of exogenous amino acids show that auxotro-
what has been suggested for yeast [55] and E. coli [56], phies and cross feeding are selected for.
regulation by fructose 1,6-bisphosphate seems impor- 8. Kelleher P, Bottacini F, Mahony J, Kilcawley KN, van Sinderen D:
tant for the switch. F16bP activates lactate dehydroge- Comparative and functional genomics of the Lactococcus
lactis taxon; insights into evolution and niche adaptation.
nase of L. lactis, E. faecalis and S. pyogenes, but not that of BMC Genom 2017, 18:267.
L. plantarum [57]; suggestively, only the latter organism 9. Liu M, Nauta A, Francke C, Siezen RJ: Comparative genomics
does not switch to acetate production at low dilution of enzymes in flavor-forming pathways from amino acids in
rate [58]. This activation by F16bP is different from that lactic acid bacteria. Appl Environ Microbiol 2008, 74:
4590–4600.
on pyruvate kinase, observed from LAB to man: in
10. Rai AK, Sanjukta S, Jeyaram K: Production of angiotensin I
L. lactis the current suggestion from computational converting enzyme inhibitory (ACE-I) peptides during milk
studies is that feedforward activation ensures robustness fermentation and their role in reducing hypertension. Crit Rev
to feast-famine dynamics [48], possibly through main- Food Sci Nutr 2017, 57:2789–2800.
taining adequate phosphoenol pyruvate levels [50], the 11. Fernández M, Zúñiga M: Amino acid catabolic pathways of
lactic acid bacteria. Crit Rev Microbiol 2006, 32:155–183.
substrate required for sugar uptake and phosphorylation.
12. Goffin P, van de Bunt B, Giovane M, Leveau JHJ, Höppener-
Ogawa S, Teusink B, Hugenholtz J: Understanding the physi-
Conclusion ology of Lactobacillus plantarum at zero growth. Mol Syst Biol
2010, 6:413.
We have given a brief overview of a number of what we
suggest are adaptive behaviours of lactic acid bacteria to 13. Reeds PJ: Dispensable and indispensable amino acids for
humans. J Nutr 2000, 130:1835S–1840S.
their rich nutrient environments: a fascinating mix of
chemical warfare, cheating, robustness and rat races. We 14. Teusink B, van Enckevort FHJ, Francke C, Wiersma A,
Wegkamp A, Smid EJ, Siezen RJ: In silico reconstruction of
focused on current open scientific questions and our lat- the metabolic pathways of Lactobacillus plantarum:
ests insights, and on the commonalities and differences comparing predictions of nutrient requirements with those
from growth experiments. Appl Environ Microbiol 2005, 71:
with other model organisms that are more often studied in 7253–7262.
the systems biology field. The models and advanced mo- 15. Veith N, Solheim M, van Grinsven KWA, Olivier BG, Levering J,
lecular and functional genomics tools are available [59]. Grosseholz R, Hugenholtz J, Holo H, Nes I, Teusink B, et al.:
Using a genome-scale metabolic model of Enterococcus
faecalis V583 to assess amino acid uptake and its impact on
Acknowledgements central metabolism. Appl Environ Microbiol 2015, 81:
We thank prof J Ellers for useful comments. LAB-related work in our group 1622–1633.
is supported by STW grant 13858, TI Food and Nutrition project 16MF01,
EraSysApp project SysMilk: NWO 832.14.001 and the ITN MicroWine: 16. Levering J, Fiedler T, Sieg A, van Grinsven KWA, Hering S,
Horizon2020 grant 643031. Veith N, Olivier BG, Klett L, Hugenholtz J, Teusink B, et al.:
Genome-scale reconstruction of the Streptococcus
pyogenes M49 metabolic network reveals growth re-
References quirements and indicates potential drug targets. J Biotechnol
Papers of particular interest, published within the period of review, 2016, 232:25–37.
have been highlighted as:
17. Morishita T, Deguchi Y, Yajima M, Sakurai T, Yura T: Multiple
nutritional requirements of lactobacilli: genetic lesions affecting
* of special interest
* * of outstanding interest amino acid biosynthetic pathways. J Bacteriol 1981, 148:64–71.
18. Pastink MI, Teusink B, Hols P, Visser S, de Vos WM,
1. de Vos WM: Systems solutions by lactic acid bacteria: from Hugenholtz J: Genome-scale model of Streptococcus ther-
paradigms to practice. Microb Cell Factories 2011, 10:S2. mophilus LMG18311 for metabolic comparison of lactic acid
bacteria. Appl Environ Microbiol 2009, 75:3627–3633.
2. Sauer M, Russmayer H, Grabherr R, Peterbauer CK, Marx H: The
efficient clade: lactic acid bacteria for industrial chemical 19. Flahaut NAL, Wiersma A, van de Bunt B, Martens DE, Schaap PJ,
Sijtsma L, Dos Santos VAM, de Vos WM: Genome-scale

www.sciencedirect.com Current Opinion in Systems Biology 2017, 6:7–13


12 Systems biology of model organisms (2017)

metabolic model for Lactococcus lactis MG1363 and its more protein per ATP than overflow metabolism, explaining the latter
application to the analysis of flavor formation. Appl Microbiol phenomenon as a resource allocation strategy.
Biotechnol 2013, 97:8729–8739.
37. Bachmann H, Fischlechner M, Rabbers I, Barfa N, Branco Dos
20. Arakawa K, Matsunaga K, Takihiro S, Moritoki A, Ryuto S, Santos F, Molenaar D, Teusink B: Availability of public goods
Kawai Y, Masuda T, Miyamoto T: Lactobacillus gasseri re- shapes the evolution of competing metabolic strategies. Proc
quires peptides, not proteins or free amino acids, for growth Natl Acad Sci U S A 2013, http://dx.doi.org/10.1073/
in milk. J Dairy Sci 2015, 98:1593–1603. pnas.1308523110.
21. Pál C, Papp B, Lercher MJ, Csermely P, Oliver SG, Hurst LD: 38. You C, Okano H, Hui S, Zhang Z, Kim M, Gunderson CW,
Chance and necessity in the evolution of minimal metabolic Wang Y-P, Lenz P, Yan D, Hwa T: Coordination of bacterial
networks. Nature 2006, 440:667–670. proteome with metabolism by cyclic AMP signalling. Nature
2013, 500:301–306.
22. Zelezniak A, Andrejev S, Ponomarova O, Mende DR, Bork P,
Patil KR: Metabolic dependencies drive species co- 39. Kochanowski K, Gerosa L, Brunner SF, Christodoulou D,
occurrence in diverse microbial communities. Proc Natl Acad * * Nikolaev YV, Sauer U: Few regulatory metabolites coordinate
Sci U S A 2015, 112:6449–6454. expression of central metabolic genes in Escherichia coli.
Mol Syst Biol 2017, 13:903.
23. MJA van Hoek, Merks RMH: Emergence of microbial diversity This paper combines promoter activity and metabolomics to conclude
due to cross-feeding interactions in a spatial model of gut that the coordination of gene expression seems relievingly simple. For
microbial metabolism. BMC Syst Biol 2017, 11:56. a theoretical underpinning, see bioRxiv 115428; doi: https://doi.org/10.
1101/115428.
24. Gottstein W, Olivier BG, Bruggeman FJ, Teusink B: Constraint-
based stoichiometric modelling from single organisms to 40. Hove-Jensen B, Andersen KR, Kilstrup M, Martinussen J,
microbial communities. J R Soc Interface 2016:13. Switzer RL, Willemoe€s M: Phosphoribosyl diphosphate
(PRPP): biosynthesis, enzymology, utilization, and metabolic
25. Towbin BD, Korem Y, Bren A, Doron S, Sorek R, Alon U: Opti-
significance. Microbiol Mol Biol Rev MMBR 2017:81.
mality and sub-optimality in a bacterial growth law. Nat
Commun 2017, 8:14123. 41. Bosdriesz E, Molenaar D, Teusink B, Bruggeman FJ: How fast-
growing bacteria robustly tune their ribosome concentration
26. Erkus O, de Jager VCL, Spus M, van Alen-Boerrigter IJ, van
* * Rijswijck IMH, Hazelwood L, Janssen PWM, van Hijum SAFT, to approximate growth-rate maximization. FEBS J 2015, http://
dx.doi.org/10.1111/febs.13258.
Kleerebezem M, Smid EJ: Multifactorial diversity sustains mi-
crobial community stability. ISME J 2013, 7:2126–2136. 42. Molenaar D, van Berlo R, de Ridder D, Teusink B: Shifts in
Population dynamics study of a cheese starter culture (a mixture of LAB growth strategies reflect tradeoffs in cellular economics. Mol
strains added to milk to start the cheese making process) that identifies Syst Biol 2009, 5:323.
phage sensitivity as a major driver in maintaining genetic diversity.
43. Scott M, Gunderson CW, Mateescu EM, Zhang Z, Hwa T: Inter-
27. Mahony J, McDonnell B, Casey E, van Sinderen D: Phage-host dependence of cell growth and gene expression: origins and
interactions of cheese-making lactic acid bacteria. Annu Rev consequences. Science 2010, 330:1099–1102.
Food Sci Technol 2016, 7:267–285.
44. Kafri M, Metzl-Raz E, Jona G, Barkai N: The cost of protein
28. Ferenci T: Trade-off mechanisms shaping the diversity of * production. Cell Rep 2016, 14:22–31.
bacteria. Trends Microbiol 2016, 24:209–223. Elegant study in yeast with reporter constructs to address whether
costs of making a protein are in transcription or translation; this, it
29. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P,
turned out, depends on the conditions.
Moineau S, Romero DA, Horvath P: CRISPR provides acquired
resistance against viruses in prokaryotes. Science 2007, 315: 45. Chubukov V, Uhr M, Le Chat L, Kleijn RJ, Jules M, Link H,
1709–1712. Aymerich S, Stelling J, Sauer U: Transcriptional regulation is
insufficient to explain substrate-induced flux changes in
30. Bachmann H, Molenaar D, Kleerebezem M, van Hylckama
Bacillus subtilis. Mol Syst Biol 2013, 9:709.
Vlieg JET: High local substrate availability stabilizes a coop-
erative trait. ISME J 2011, 5:929–932. 46. Solis-Escalante D, Kuijpers NGA, Barrajon-Simancas N, van den
Broek M, Pronk JT, Daran J-M, Daran-Lapujade P: A minimal set
31. Gore J, Youk H, van Oudenaarden A: Snowdrift game dynamics
of glycolytic genes reveals strong redundancies in Saccha-
and facultative cheating in yeast. Nature 2009, 459:253–256.
romyces cerevisiae central metabolism. Eukaryot Cell 2015,
32. Hardin G: The tragedy of the commons. The population 14:804–816.
problem has no technical solution; it requires a fundamental
47. O’Brien EJ, Lerman JA, Chang RL, Hyduke DR, Palsson BØ:
extension in morality. Science 1968, 162:1243–1248. * * Genome-scale models of metabolism and gene expression
33. Goel A, Eckhardt TH, Puri P, de Jong A, Branco Dos Santos F, extend and refine growth phenotype prediction. Mol Syst Biol
* Giera M, Fusetti F, de Vos WM, Kok J, Poolman B, et al.: Protein 2013, 9:693.
costs do not explain evolution of metabolic strategies and The capabilities of a ME model of E. coli were demonstrated, showing
regulation of ribosomal content: does protein investment for the first time how augmenting metabolic networks with protein costs
explain an anaerobic bacterial Crabtree effect? Mol Microbiol can enhance their biological interpretation and capabilities, including
2015, 97:77–92. overflow metabolism.
Careful quantitative study of the shift from mixed acid to lactate
48. Murabito E, Verma M, Bekker M, Bellomo D, Westerhoff HV,
fermentation in the chemostat, showing at the level of mRNA, protein
Teusink B, Steuer R: Monte-Carlo modeling of the central
and enzyme activity that flux increases over threefold without appre-
carbon metabolism of Lactococcus lactis: insights into
ciable changes in corresponding protein level.
metabolic regulation. PLoS One 2014, 9:e106453.
34. Neves AR, Ventura R, Mansour N, Shearman C, Gasson MJ,
49. Koebmann BJ, Solem C, Pedersen MB, Nilsson D, Jensen PR:
Maycock C, Ramos A, Santos H: Is the glycolytic flux in
Expression of genes encoding F(1)-ATPase results in
Lactococcus lactis primarily controlled by the redox charge?
uncoupling of glycolysis from biomass production in
Kinetics of NAD(+) and NADH pools determined in vivo by
Lactococcus lactis. Appl Environ Microbiol 2002, 68:
13C NMR. J Biol Chem 2002, 277:28088–28098.
4274–4282.
35. van Dijken JP, Weusthuis RA, Pronk JT: Kinetics of growth and
50. Dolatshahi S, Fonseca LL, Voit EO: New insights into the
sugar consumption in yeasts. Ant Van Leeuwenhoek 1993, 63:
complex regulation of the glycolytic pathway in Lactococcus
343–352.
lactis. II. Inference of the precisely timed control system
36. Basan M, Hui S, Okano H, Zhang Z, Shen Y, Williamson JR, regulating glycolysis. Mol Biosyst 2015, 12:37–47.
* * Hwa T: Overflow metabolism in Escherichia coli results from
51. Solopova A, van Gestel J, Weissing FJ, Bachmann H, Teusink B,
efficient proteome allocation. Nature 2015, 528:99–104.
Kok J, Kuipers OP: Bet-hedging during bacterial diauxic shift.
A set of elegant quantitative perturbation studies are interpreted with
Proc Natl Acad Sci U S A 2014, 111:7427–7432.
phenomenological bacterial growth laws to show that respiration costs

Current Opinion in Systems Biology 2017, 6:7–13 www.sciencedirect.com


Systems biology of lactic acid bacteria Teusink and Molenaar 13

52. Kotte O, Volkmer B, Radzikowski JL, Heinemann M: Phenotypic 57. Feldman-Salit A, Hering S, Messiha HL, Veith N, Cojocaru V,
bistability in Escherichia coli’s central carbon metabolism. Sieg A, Westerhoff HV, Kreikemeyer B, Wade RC, Fiedler T:
Mol Syst Biol 2014, 10:736. Regulation of the activity of lactate dehydrogenases from
four lactic acid bacteria. J Biol Chem 2013, 288:21295–21306.
53. van Heerden JH, Wortel MT, Bruggeman FJ, Heijnen JJ,
Bollen YJM, Planqué R, Hulshof J, O’Toole TG, Wahl SA, 58. Teusink B, Wiersma A, Molenaar D, Francke C, de Vos WM,
Teusink B: Lost in transition: start-up of glycolysis yields Siezen RJ, Smid EJ: Analysis of growth of Lactobacillus
subpopulations of nongrowing cells. Science 2014, 343: plantarum WCFS1 on a complex medium using a genome-
1245114. scale metabolic model. J Biol Chem 2006, 281:40041–40048.
54. Christensen CD, Hofmeyr J-HS, Rohwer JM: Tracing regulatory 59. Grosseholz R, Ching-Chiek K, Veith N, Fiedler T, Strauss M,
routes in metabolism using generalised supply-demand * Olivier BG, Collins BC, Schubert OT, Bergman FT,
analysis. BMC Syst Biol 2015, 9:89. Kreikemeyer B, et al.: Integrating highly quantitative prote-
omics and genome-scale metabolic modeling to study pH
55. van Heerden JH, Bruggeman FJ, Teusink B: Multi-tasking of adaptation in the human pathogen Enterococcus faecalis.
biosynthetic and energetic functions of glycolysis explained NPJ Syst Biol Appl 2016, http://dx.doi.org/10.1038/
by supply and demand logic. BioEssays News Rev Mol Cell npjsba.2016.17.
Dev Biol 2015, 37:34–45. Comprehensive time-resolved proteomics data after a pH challenge
that was integrated in a genome-scale metabolic model.
56. Kochanowski K, Volkmer B, Gerosa L, Haverkorn van
Rijsewijk BR, Schmidt A, Heinemann M: Functioning of a
metabolic flux sensor in Escherichia coli. Proc Natl Acad Sci U
S A 2013, 110:1130–1135.

www.sciencedirect.com Current Opinion in Systems Biology 2017, 6:7–13

Vous aimerez peut-être aussi