ArticlePDF AvailableLiterature Review

Quorum sensing for population-level control of bacteria and potential therapeutic applications

Authors:

Abstract and Figures

Quorum sensing (QS), a microbial cell-to-cell communication process, dynamically regulates a variety of metabolism and physiological activities. In this review, we provide an update on QS applications based on autoinducer molecules including acyl-homoserine lactones (AHLs), auto-inducing peptides (AIPs), autoinducer 2 (AI-2) and indole in population-level control of bacteria, and highlight the potential in developing novel clinical therapies. We summarize the development in the combination of various genetic circuits such as genetic oscillators, toggle switches and logic gates with AHL-based QS devices in Gram-negative bacteria. An overview is then offered to the state-of-the-art of much less researched applications of AIP-based QS devices with Gram-positive bacteria, followed by a review of the applications of AI-2 and indole based QS for interspecies communication among microbial communities. Building on these general-purpose QS applications, we highlight the disruptions and manipulations of QS devices as potential clinical therapies for diseases caused by biofilm formation, antibiotic resistance and the phage invasion. The last part of reviewed literature is dedicated to mathematical modelling for QS applications. Finally, the key challenges and future perspectives of QS applications in monoclonal synthetic biology and synthetic ecology are discussed.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Cellular and Molecular Life Sciences
https://doi.org/10.1007/s00018-019-03326-8
REVIEW
Quorum sensing forpopulation‑level control ofbacteria andpotential
therapeutic applications
ShengboWu1,2,3· JiahengLiu1,3,4· ChunjiangLiu1,2,3· AidongYang5· JianjunQiao1,3,4
Received: 4 July 2019 / Revised: 13 September 2019 / Accepted: 30 September 2019
© Springer Nature Switzerland AG 2019
Abstract
Quorum sensing (QS), a microbial cell-to-cell communication process, dynamically regulates a variety of metabolism and
physiological activities. In this review, we provide an update on QS applications based on autoinducer molecules includ-
ing acyl-homoserine lactones (AHLs), auto-inducing peptides (AIPs), autoinducer 2 (AI-2) and indole in population-level
control of bacteria, and highlight the potential in developing novel clinical therapies. We summarize the development in
the combination of various genetic circuits such as genetic oscillators, toggle switches and logic gates with AHL-based QS
devices in Gram-negative bacteria. An overview is then offered to the state-of-the-art of much less researched applications
of AIP-based QS devices with Gram-positive bacteria, followed by a review of the applications of AI-2 and indole based
QS for interspecies communication among microbial communities. Building on these general-purpose QS applications, we
highlight the disruptions and manipulations of QS devices as potential clinical therapies for diseases caused by biofilm forma-
tion, antibiotic resistance and the phage invasion. The last part of reviewed literature is dedicated to mathematical modelling
for QS applications. Finally, the key challenges and future perspectives of QS applications in monoclonal synthetic biology
and synthetic ecology are discussed.
Keywords Cell–cell communication· Signaling molecule· Microbial community· Population control· Genetic circuit·
Gut microbiota
List of symbols
[A] Intracellular AHL concentration (mM)
[C] Intracellular CI protein concentration (mM)
[E] Intracellular CcdB protein concentration (mM)
[L] Intracellular LacR concentration (mM)
[LuxR] Intracellular LuxR concentration (mM)
[R] Intracellular AHL/LuxR complex concentration
(mM)
N The cell density (CFUml−1)
Nm The maximum cell density (CFUml−1)
Fpfk The fractional Pfk-1 activity (U/mg)
Kd The cumulative dissociation constant
X Biomass concentration (gL−1)
n1, n2 Transcription factor cooperativity/
multimerization
αC CI protein synthesis rate constant (μMmin−1)
αL1, αL2 LacR protein synthesis rate constants
(μMmin−1)
βC CI repression coefficient (mM)
βL LacR repression coefficient (mM)
d Cell death rate (nM−1 h−1)
dA, dE AHL and CcdB protein decay constant (min−1)
dC CI protein decay constant (min−1)
dL, dR LacR and LuxR–AHL complex decay constants
(min−1)
k Growth rate (h−1)
kE CcdB protein production rate constant (h−1)
vA AHL production rate constant (nMmLh−1)
Cellular andMolecular Life Sciences
* Aidong Yang
aidong.yang@eng.ox.ac.uk
* Jianjun Qiao
jianjunq@tju.edu.cn
1 School ofChemical Engineering andTechnology, Tianjin
University, Tianjin300072, China
2 State Key Laboratory ofChemical Engineering, Tianjin
University, Tianjin300072, China
3 Collaborative Innovation Center ofChemical Science
andEngineering (Tianjin), Tianjin300072, China
4 Key Laboratory ofSystems Bioengineering, Ministry
ofEducation (Tianjin University), Tianjin300072, China
5 Department ofEngineering Science, University ofOxford,
OxfordOX13PJ, UK
S.Wu et al.
1 3
θR LuxR/AHL activation coefficient (mM)
ρR LuxR/AHL dimerization constant (μM−3min−1)
Introduction
Quorum sensing (QS) is a cell–cell communication process,
which is ubiquitous in fungi [1], bacteria and even viruses
[2]. QS regulates a series of physiological and biochemical
functions, such as biofilm formation, conjugation, compe-
tence, bacteriocin production and pathogenesis, achieved
by microbes producing, secreting, sensing and responding
to certain signal molecules which are called autoinducers
(AIs) [3]. Generally, various AIs can be roughly divided
into three types: (i) acylated homoserine lactones (AHLs)
and the diffusible signaling factors (DSFs) utilized by Gram-
negative bacteria; (ii) auto-inducing peptides (AIPs) utilized
by Gram-positive bacteria; and (iii) autoinducer 2 (AI-2) and
indole for interspecies communication of microbial com-
munities [4]. Combining these AIs and their relevant QS
devices with synthetic genetic circuits is of great importance
to the dynamic control of bacterial populations and to the
development of potential clinical therapies (Fig.1).
Dynamic control of bacterial populations usually includes
population size control, dynamic metabolic engineering for
desirable products and the regulation of various physiologi-
cal activities [5] (Fig.1). Metabolic control, a major issue
in dynamic control of metabolic engineering, can be divided
into static metabolic control and dynamic metabolic con-
trol [6]. Usually, static metabolic control involves natural or
slightly modified control systems with knockout, weakening
and overexpression of genes. Dynamic metabolic control
utilizes genetic circuits such as toggle switches and sensor-
regulator to achieve the dynamic adjustment of metabolic
production of microbes [5]. According to the type of genetic
circuits involved, either on–off or continuous dynamic meta-
bolic control can be achieved [7]. A common strategy via
introducing an on–off switch is to close the relevant competi-
tive pathways when the bacteria population reaches a certain
level. This type of genetic circuits has the disadvantages of
requiring proper induction time, increasing production costs
due to the addition of inducer and being incapable of sensing
the changing environment continuously. To overcome these
disadvantages, continuous dynamic metabolic control has
been developed to up-regulate the desirable product using
synthetic feedback loops, such as QS-based devices [7]. The
implementation of dynamic metabolic control can be either
pathway-specific or pathway-independent [8]. The pathway-
specific implementations are achieved by detecting input and
output changes of a relevant intermediate or byproduct [9],
while the pathway-independent implementations are through
nutrients in the medium or by QS [10, 11]. Compared to
the pathway-specific implementations which are restricted
to sense and dynamically control the metabolism of intra-
cellular pathways, pathway-independent implementations
allow microbes to respond to the changing extracellular
environment and adjust accordingly their metabolism and
physiological activities, with QS as an important enabler
[8]. What’s more, significant advances have been made in
synthetic biology which created synthetic pathways and cir-
cuits to control the expression levels of relevant genes, such
as overexpressing the genes for producing glycosides [12]
in engineered bacteria. Transcriptional toggle switches [13]
and transcriptional oscillators [14] are involved in transcrip-
tional regulation of genes, and genetic loops such as bista-
ble positive feedback loops and RNA-based anti-switches
are constructed into biological systems to control post-
transcriptional regulation [15], metabolic flux distribution
[16] and signaling proteins expression [17]. These QS-based
genetic circuits not only make the synthetic systems more
reliable and robust [18], but also provide new avenues to the
dynamic control of bacterial populations [19].
With the increasingly recognized importance of patho-
gens and microbiota for human health, the QS-based mono-
clonal synthetic biology and synthetic ecology have enor-
mous potential in promoting the development of potential
clinical therapies for curing devastating diseases, tackling
antimicrobial resistance [20] (Fig.1). Many bacteria have
been shown to have a tendency to organize in aggregates
generally to adhere to surfaces to form biofilms, and biofilm
formation is a principal virulence factor in many localized
Fig. 1 QS applications for dynamic control of bacteria populations
and its potential clinical therapies for diseases. Dynamic control of
bacterial populations includes three aspects, i.e., the population of
bacteria control, dynamic metabolic engineering control and regula-
tion of physiological activities. They mainly work on the combina-
tions of various genetic circuits such as genetic oscillators, genetic
toggle switches and genetic logic gates. Underpinned by the func-
tioning of autoinducer molecules, i.e., AHLs, DSFs, AIPs, AI-2
and indole, the disruptions and manipulations of QS in the dynamic
control of bacteria populations can be extended to be widely applied
in monoclonal synthetic biology and synthetic ecology to develop
potential clinical therapies
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
chronic infections [21, 22]. As shown by existing stud-
ies [23], QS and quorum quenching [4] can significantly
affect biofilm formation. One of the most important factors
causing the changes of microbiota is the use of antibiotics,
which does not only alter the microbiota but also promote
the emergence of antibiotics resistance [24]. It has recently
been demonstrated that QS inhibition including quorum
quenching and other QS-blocking approaches can decrease
the production of virulence factor [25]. Therefore, coupling
QS devices with microbial consortia in various environ-
ments such as the human gut has much therapeutic potential,
for example, treating chronic infections [26] and relieving
antimicrobial resistance [27]. Besides, as infections of phage
increase at high cell density [28], QS devices have much
potential to regulate the CRISPR-Cas (clustered regularly
interspaced short palindromic repeats; CRISPR-associated)
immune systems to monitor the development of diseases
[29].
Recently, several QS-based reviews have been published
which focus on aspects including QS signals transduction
and architectures, dynamic control for metabolic engineer-
ing, applications of synthetic microbial consortia, socio-
microbiology based on cell–cell communications, the
applications of QS inhibitors in biofilm formation and the
mathematical modelling for QS, as listed in Table1. The
purpose of this current review is to provide an updated sum-
mary of the more recent achievements in applying QS to the
dynamic control of bacterial populations and in developing
potential clinical therapies. We start by presenting the recent
promising achievements, including bacteria population con-
trol, applications of the genetic toggle switches, synthetic
genetic oscillators and genetic logic gates that apply QS
devices which are based on AHLs signal transduction in
Gram-negative bacteria. We then provide an overview of the
AIP-based dynamic regulations of competence and virulence
in Gram-positive bacteria, followed by that of AI-2-based
and indole-based control of metabolism and physiological
activities in microbial communities. Building on the review
of these rather generic QS applications, we further highlight
the important progress in the disruptions and manipulations
of QS devices for therapeutic applications. The last part of
the reviewed literature is on the mathematical modelling
related to QS applications in the dynamic control of bacteria.
Finally, we identify key challenges and suggest directions for
future QS research.
QS applications inGram‑negative bacteria
QS forpopulation control
QS-dependent activities are the result of density-depend-
ent expression of both intra- and extracellular gene
products [47]. It is essential for population-level dynamics
and genetic-level regulation [19]. An etal. [48] and Goo
etal. [49] certified that nutrients are typically limited and
the environment is unfavorable for growth and metabolism
of microbes in a crowded environment. Therefore, it is of
much importance to control the cell density to optimize
metabolic production.
You etal. [50] proposed that cell–cell communication
can be used to programme the dynamics of a population.
Combining the cell survival and death genes to the LuxI/
LuxR QS circuit, they built and characterized a ‘popula-
tion control’ circuit that autonomously regulated the den-
sity of an Escherichia coli population (Fig.2a). Balagadde
etal. [51] applied two E. coli populations to construct a
synthetic ecosystem (predator and prey) (Fig.2b), which
was based on two QS mechanisms (LuxI/LuxR and LasI/
LasR). The predators will die following the expression of
a suicide gene (ccdB) when the density of prey is low. As
the prey density increases, AHL accumulates in the culture
and eventually reaches a sufficiently high concentration,
its combination with LuxR will then work to increase the
expression of an antidote gene (ccdA) to rescue the preda-
tors. However, the predators will produce and accumulate
Las AHL to a sufficient level to bind with LasR, which
will, in turn, activate the expression of ccdB gene to kill
the preys. The series of events leads to the oscillatory
behavior of the two E. coli populations, which is typi-
cal for a two-strain ecosystem. More recently, AHL-based
synthetic E. coli systems were used to test a general rule
deduced for predicting coexistence and productivity of
mutualistic communities [52]. Compared to the previous
work by the same group [51], new features of these experi-
mentally tested systems included the use of Isopropyl β-d-
1-thiogalactopyranoside (IPTG) to induce the expression
of CcdB (representing stress), and the application of anhy-
drotetracycline (aTc) to induce the QS module to impose
the cooperation cost to both strains.
With the development of genetic circuits, the lysis genes
can be coupled with new synthetic circuits such as genetic
oscillators to realize various functions. Din etal. [53] inte-
grated the lysis genes with a microbial drug delivery system
[54] to form a synchronized lysis circuit (SLC) for control-
ling population levels and facilitating drug delivery using
bacteria (Fig.2c). The circuit includes a luxI promoter which
promotes expression of LuxI, a therapeutic gene, a reporter
gene and a lysis gene ϕX174 E. When AHL reaches a target
threshold, the expression of the therapeutic gene and the
reporter gene will be promoted, and bacteria will produce
and release cytotoxic agents continually. At the same time,
the number of bacterium will decrease due to the expression
of the lysis gene. Then, a small number of remaining bacte-
ria will begin to produce AHL again to restart this process
in a cyclical fashion. Compared with existing drug delivery
S.Wu et al.
1 3
strategies, SLC can be used as a novel therapeutic technique
to cure diseases through population control of bacteria.
In a separate study, two orthogonal QS devices and a
population control mechanism were combined together to
control the population densities of competitive microbes of
Salmonella typhimurium strains [55] (Fig.2d). lux and rpa
QS devices can be integrated with two lysis genes to form
SLCs in two bacterial strains to control bacteria population.
When two competitive microbes, the Lux-QS strain and the
Rpa-QS strain, are co-cultured, the latter has a significant
growth advantage over the former. It was observed that
from an initial population ratio of 100:1 between the Lux-
QS strain and the Rpa-QS strain with the lysis gene, the
population ratio became about 1:1 over 10h. Without the
lysis gene, the co-culture would be taken over by the Lux-QS
strain. This demonstrates that the integration of two orthogo-
nal QS devices to form an ‘ortholysis’ system is a potential
strategy to stabilize competitive strains in co-cultures.
Table 1 Recent reviews focusing on QS
Theme Core content References
QS signals transduction and architectures Reviewed various signal–response systems and their applications in
Gram-negative bacteria
[18]
Reviewed types of molecular mechanisms coupled with various QS
devices in Gram-negative and Gram-positive bacteria, and some
network architectures of QS circuits
[30]
Reviewed various signal–response systems in Gram-positive bacteria [31]
Reviewed the mechanisms of intracellular pathway and extracellular
pathway QS system, and the applications of regulating of conjugation,
competence, bacteriocin production, and biofilm formation in Gram-
positive bacteria
[32]
Reviewed the function of indole in bacterial pathogenesis and eukary-
otic immunity
[33]
QS and its applications in marine microbes [34]
Reviewed the diversity, functions, biosynthetic pathways, and turnover
systems for the diffusible signaling factors (DSF) family of QS signals
[35]
Dynamic control for metabolic engineering Reviewed various strategies such as QS system in the applications of
dynamic metabolic engineering
[7]
Reviewed some dynamic control strategies coupling with QS to syn-
chronize cellular activity
[36]
Applications of synthetic microbial consortia Reviewed some engineering cell–cell communication, mainly on QS,
and synthetic microbial consortia for community composition, divi-
sion of labor, and biofilm formation with QS system
[37]
Reviewed some typical synthetic microbial consortia by cell–cell com-
munications, mainly on QS
[38]
Discussed complex interactions and interplays in synthetic microbial
ecology based on QS-based cell–cell communication
[39]
Socio-microbiology based on cell-to-cell communications Discussed the complex signal network and the cooperation with QS
cheating phenotypes in bacterial. And reviewed various and feasible
mechanisms that have been certified to stabilize QS-based cooperation
in microbes
[40]
Reviewed the background and brief history of QS, and the applications
of QS in socio-microbiology
[4]
Applications of QS inhibitors in biofilm formation Reviewed natural and synthetic quorum sensing inhibitors (QSIs) in
various microbes
[25]
Reviewed applications of QS in biotechnology, especially for QSIs and
some other biosensors
[41]
Reviewed the mechanism for pathogenic biofilms formation, and dis-
cussed the current biofilm-targeting therapeutic strategies for disease
which caused by microbial biofilms and drug tolerance
[42]
Reviewed how bacteria deploy QS in realistic, complex and dynami-
cally changing scenarios
[43]
Mathematical modelling for QS Reviewed the modeling approaches on a systemic level [44]
Proposed the core principles of autoinducer systems in bacteria [45]
Reviewed various QS mathematical models [46]
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
The above studies suggest that the combination of lysis
genes, such as ccdB, and various QS devices (LuxI/LuxR,
LasI/LasR, or RpaI/RpaR) can be applied to achieve the
control of bacterial populations in either mon-culture or
co-culture systems. Such control helps meet the essential
requirement on population size needed for realizing vari-
ous high-value metabolic production and microbial clinical
treatments.
QS‑based synthetic genetic oscillators
Using only a small amount of regulators in a large-scale
genetic regulatory network, a relatively large number of
genes and hence the complex cell behavior can be regu-
lated [56, 57]. Many metabolic activities, such as popu-
lation control, respiration, hormone secretion and circa-
dian rhythms, are closely associated with synchronized
oscillators [5860]. Developing synthetic gene oscilla-
tors (SGOs) is one of the main research directions of the
research on synthetic genetic regulatory networks [61].
The first SGO, the repressilator, was proposed in 2000
and is illustrated in Fig.3a [14]. Unlike the repressilator
where only repression exists, a relaxation oscillator has
positive feedback loops which can promote the expression
of negative feedback loops [62]. Hasty etal. [63] applied
the common gene regulatory components (ci, lac and PRM)
to construct a SGO model (Fig.3b). Stricker etal. [64]
developed a fast, robust and persistent engineered genetic
oscillator in E. coli with the induction of IPTG and arab-
inose (Fig.3c), and confirmed that its oscillatory period
can be tuned by altering inducer levels, temperature and
the media source.
Due to the complexity of cellular interaction and vari-
ability, it is important to investigate population-level dynam-
ics of microbes, such as synchronization [65, 66] and pro-
grammed population interactions [39, 67, 68]. To avoid the
random phase drift and remove the effects of noise, it is
desirable to introduce measures to coordinate and synchro-
nize each cellular oscillator [69], and QS has been found to
offer an important means for this task [70].
Mcmillen etal. [71] firstly combined a genetic relaxation
oscillator, which is composed of promotor PRE, gene X (cii)
and gene Y (ftsh), with the lux QS mechanism (Fig.3d).
Protein CII can be degraded by protein FtsH, while the com-
plex of AHL and LuxR (LuxR-AHL) can activate the tran-
scription of CII. When the concentration of AHL reaches its
threshold, it will bind with another LuxR in other cells to
regulate their CII level.
Fig. 2 QS for the lysis and population control. a The signaling mol-
ecule 3OC6HSL (Lux AHL) is produced by the LuxI synthase. It will
accumulate in the culture with the increased cell density of E. coli.
When the concentration of the Lux AHL reaches a certain thresh-
old, AHL will diffuse back into E. coli and be recognized by LuxR,
a specific protein receptor to activate the transcriptional expression
of the killer protein LacZa-ccdB to regulate the cell death of E. coli
and consequently control the population density. b Based on the two
QS mechanisms (LuxI/LuxR and LasI/LasR QS devices), two E.
coli strains are engineered to construct a synthetic predator and prey
ecosystem. c When the population reaches the critical threshold, the
AHL will bind with LuxR to become AHL–LuxR complex. It will
facilitate the expression of LuxI, gene ϕX174E for lysis, therapeutic
gene for cytotoxic agents, and sfGFP for reporter. d Genetic circuits
of a two-strain ecosystem including Lux-QS and Rpa-QS S. typhimu-
rium strains
S.Wu et al.
1 3
Danino etal. [17] applied a QS-based approach to syn-
chronized oscillations at the colony level (Fig.3e). In this
circuit, luxI, aiiA and yemGFP genes were controlled by
the luxI promoter. The LuxR–AHL complex activated the
luxI promoter [72]. AHL was degraded by homoserine
lactonase (AiiA).
As the DNA copy number changes with environmental
pressures, it is a challenge to combine gene circuits with-
out predictable dynamic control of gene expression [73].
Treating the DNA copy number as a circuit control element,
Baumgart etal. [74] reported that the expression of some
cut site genes on a plasmid can be repressed by a targeted
Fig. 3 The applications of QS-based synthetic genetic oscillators. a
When the promoter PllacO1 is successfully promoted, the TetR pro-
tein will inhibit the downstream PltetO1 promoter, the CI protein will
not be expressed, the inhibition of the Pr promoter will be released,
LacI protein will be expressed, and the state of the PllacO1 promoter
will be changed from “turn on” to “turn off”. Then the expression of
TetR protein will be inhibited, and the inhibition of the PltetO1 pro-
moter will be released, CI protein will be expressed, Pr promoter
will be inhibited, LacI protein expression will be inhibited, and pro-
moter PllacO1 will resume its “turn on” state. This is the mechanism
of the first repressilator. b A relaxation oscillator with several positive
and negative feedback loops. CI protein promotes the expression of
itself and of LacI protein, while LacI protein inhibits the expression
of itself and of CI protein. c A relaxation oscillator with the induc-
tion of IPTG and arabinose. Arabinose promotes the expression of
AraC, GFP and LacI protein, while LacI protein inhibits the expres-
sion of itself, GFP and CI protein without IPTG. d Network architec-
ture of the proposed gene network. The relaxation oscillator includes
CII protein, FtsH protein LuxI/LuxR type QS system. e The network
architecture of a synchronized oscillation design with LuxI/LuxR
type QS system. f The circuit diagrams of two-plasmid circuit which
includes the activator and repressor plasmid. The activator plasmid
keeps activating luxI promoter to activate the expression of I-SceI in
the repressor plasmid. I-SceI can be used to negatively regulate the
expression of some cut site genes on the activator plasmid, which will
reduces its copy number
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
nuclease (I-SceI) to reduce the copy number. They combined
the negative feedback component with the positive feedback
component of lux QS system to form a synthetic gene oscil-
lator of the plasmid copy number (Fig.3f).
Most SGOs, such as those introduced above, have been
constructed to operate within single, isogenic cellular popu-
lations. Representing a step further, Prindle etal. [75] inte-
grated a genetic relaxation oscillator, lux QS system and
redox signaling (arsenite) to form coupled genetic ‘biopixels’
among different colonies (Fig.4a). This genetic circuit con-
sists of arsenite-responsive promoter, ArsR, lux QS system
and arsenite. When there is no arsenite, ArsR will repress
the expression of luxR, thus no fluorescence or oscillation
is generated. The repression will be removed when there
is a sufficient amount of arsenite, thereby the LuxR–AHL
complex will promote oscillations and the expression of the
fluorescence gene.
Compared to the synchronization of genetic oscillators by
means of coupling with standard transcription factor-based
methods such as QS devices, work on the delay times of
synchronization by competitive protein degradation is much
less [76]. Prindle etal. developed a post-translational cou-
pling platform which worked through shared degradation by
the ClpXP protease [77] to couple various synthetic genetic
modules rapidly and efficiently. This platform was used to
integrate intracellular genetic oscillators (including Plac/ara-1,
lacI and inducers) and the LuxI/LuxR-type QS system to
realize synchronization (Fig.4b).
As an example of synchronization with two QS devices,
Chen etal. [78] constructed a SGO with an “activator” strain
and a “repressor” strain to realize the emergent, population-
level oscillations of two genetically distinct E. coli (Fig.4c).
The activator produces Rhl AHL, which promotes the tran-
scription of target genes for both strains, while the repres-
sor produces Cin AHL, which inhibits the transcription for
both strains mediated by the LacI protein. Besides, there is
another negative feedback loop in which the AiiA protein
can degrade these two AHLs. These feedback loops were
divided into four types of topologies to investigate the pop-
ulation-level dynamics of these two strains.
These studies have demonstrated that incorporating QS
devices can indeed enrich the design and implementation of
SGOs. Such systems hold much potential in regulating the
synchronization of synthetic microbial consortia to benefit
real applications in metabolic engineering, particularly the
development and optimization of production pathways for
high-value metabolites, as well as some medical applica-
tions, such as the drug release process for some probiotic
therapies. On the other hand, these potential applications
may require further optimization of the QS-based SGOs
to achieve more accurate controls, beyond the feasibility
already demonstrated by the proof-of-concept studies.
QS‑based genetic toggle switches
The main objective of metabolic engineering is to increase
the yield and productivity of the desirable production
through genetic engineering [79]. As a synthetic genetic
circuit, a metabolic toggle switch (MTS) is often used to
meet this goal [13, 80]. Demonstrating a modular design
strategy, Kobayashi etal. [81] created four E. coli strains
that contained a genetic toggle switch (Fig.5a). Half of
the strains were interfaced with a transgenic QS signaling
pathway from Vibrio fischeri that detects AHLs. The genetic
circuit combined the QS mechanism and the artificial ON/
OFF genetic toggle switch. The QS circuit was composed
of the luxI, luxR and lacI genes, and would work as follows:
Fig. 4 QS for synchronization of genetic oscillators. a A genetic
relaxation oscillator consists of arsenite-responsive promoter, ArsR,
lux QS system, and arsenite. b Coupling (i) genetic oscillators which
includes promoter Plac/ara-1, lacI gene, arabinose and IPTG inducers
and (ii) LuxI/LuxR type QS system to realize the synchronization by
the ClpXP protease platform. c Genetic circuit diagrams of the activa-
tor with Rhl QS system and repressor strains with Cin QS system
S.Wu et al.
1 3
Initially, the concentration of the AHL is too low to function,
the expression of the target gene, such as gfp, is maintained
at the ‘OFF’ state; when AHL reaches a critical concentra-
tion, the gene will switch to the ‘ON’ state.
Anesiadis etal. [82] proposed an integrated computa-
tional model and showed that a genetic toggle switch can be
effectively employed in dynamic metabolic engineering to
increase bioprocess productivity and yield (Fig.5b). Anesi-
adis etal. [83] also applied the LuxI/LuxR-type QS system
to reconstruct E. coli to improve the productivity of serine
with an ON/OFF genetic toggle switch (Fig.5c). The highest
productivity of the final strain to produce serine was 29.6%
higher than that of the previous mutant strain. To further
the design, they integrated population response, dynamic
metabolic regulation, and the DFBA metabolic modeling
method to construct a mathematical model to analyze the
global sensitivity.
Although the aforementioned on–off two-stage control
strategies have taken into consideration the appropriate
cell density or sufficiently high concentrations of AHLs
into consideration to achieve the intended purposes, the
desirable gene expression often requires a more accurate
cell density to synchronize microbial growth and cellular
activity. To further improve the desirable production, novel
engineering strategies to manage the trade-off between
cell growth and desirable production are needed. Soma
etal. [10] constructed a synthetic lux system to achieve
a dynamic switch of the metabolic flux between the TCA
cycle and the isopropanol synthesis pathway (Fig.5d),
resulting in the yield and the conversion rate improved by
Fig. 5 QS for the dynamic metabolic control in Gram-negative bacte-
ria. a Genetic circuit diagram of a genetic toggle switch and QS sign-
aling pathway. b Genetic circuit diagram of the integration of LuxI/
LuxR type QS system, central carbon metabolism and a genetic tog-
gle switch in E. coli. The genetic toggle switch consists of LacI and
λCI proteins. Their expressions are inhibited mutually. The pta gene,
relevant to ethanol production, is at the downstream of λcI gene. At
low AHL concentration, λcI and pta genes express normally, while
lacI gene is inhibited. When the concentration of the Lux AHLs reach
a certain threshold, the inhibition of lacI gene will be released. Later
on, the expression of λcI and pta genes will be repressed. c Genetic
circuit diagram consists of the genetic controller for serine production
and the QS sensor. d Design of synthetic genetic circuit coupling QS
system and the genetic toggle switch. With the help of IPTG inducer,
this genetic circuit can realize the switch flexibly between two path-
ways: isopropanol synthesis and the TCA cycle. e Schematic of
dynamic control of cell growth and myo-inositol production. f Sche-
matic of two-layer dynamic control of cell growth and d-glucaric acid
production
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
3 and 2.3 times, respectively. The threshold cell density
was controlled by IPTG in this system.
Gupta etal. [8] introduced the QS circuits to control
the expression of pfk-1 gene (which determines the carbon
flux to glycolysis and cell growth) to identify the optimal
point to switch off gene expression in terms of the desired
times and cell densities (Fig.5e). When there is no AHL,
the transcriptional regulator EsaRI70V will bind to the
PesaS promoter. As cell density increases, the accumula-
tion of AHL will reduce the activity of EsaRI70V and turn
off the expression of pfk-1 gene. As a result, most of the
glucose will be switched to the target pathway to increase
the titers of myo-inositol (MI).
Doong etal. [11] combined pathway-independent and
pathway-specific strategies to form a control mechanism
that involved two orthogonal and tunable dynamic regula-
tion strategies, for the purpose of improving the produc-
tion of d-glucaric acid (Fig.5f). The pathway-independent
strategy was based on the QS system to turn glucose utili-
zation from the glycolysis to the production of d-glucaric
acid at the threshold of AHL concentration. The pathway-
specific strategy used myo-inositol as the intermediate
metabolite sensor to achieve dynamic and autonomous
control to improve the production of d-glucaric acid.
More recently, Honjo etal. [84] constructed an engi-
neered microbial community composed of an E. coli strain
producing beta-glucosidase (BGL) and the other E. coli
strain producing isopropanol (IPA) using QS-dependent
cell lysis circuits. Specifically, the BGL-producing strain
will produce BGL to convert cellobiose to glucose as the
carbon source when the desired cell density is reached
by lysing itself. The IPA-producing strain grows with the
help of the BGL release and can detect AHL produced
by the BGL-producing strain to induce the expression of
IPA. With a three-species consortium (Gluconobacter oxy-
dansKetogulonicigenium vulgareBacillus megaterium)
for vitamin C fermentation, Wang etal. [85] applied the
LuxI/LuxR QS system to control the lysis of G. oxydans as
the population level QS-based metabolic toggle switches
for l-sorbose production and for the relieve of l-sorbose
competition with K. vulgare, thus realizing one-step fer-
mentation for vitamin C.
The above studies commonly feature the combination
of synthetic QS systems and metabolic toggle switches,
allowing the synthetic genetic circuit to be activated at the
targeted cell density, and hence resulting in self-induced
metabolic states switching. With the gradual maturation of
the application of metabolic division of labor in metabolic
engineering [86], the combined applications of engineered
microbial communities and QS-induced metabolic toggle
switches appear to offer a potentially promising direction
for metabolic engineering and synthetic biology.
QS‑based logic gates
Positive and negative feedback control loops are prevalent in
physical systems. Analogously, various positive and negative
feedback regulatory structures have been found in biologi-
cal systems [87]. The amazing similarities and sophisticated
connections between the two types of systems have attracted
many researchers to develop biomolecular computing sys-
tems [88] which mainly include the design and simulation
of various genetic circuits such as logic gates [89, 90]. Logic
gates, such as Boolean logic gates, are the basic content and
computing units of digital electronic circuits. Boolean logic
gates for genetic circuits that have been involved in various
applications mainly include AND, OR, NOR, NAND and
XOR [91]. As early as 2002, an AND logic gate based on
exogenously added signals has been established, activating
the expression of GFP as the output based on the addition of
two inputs, IPTG and aTc [92]. Step by step, various logic
gates were constructed in microbes, such as AND logic gates
in Pseudomonas aeruginosa [93] and Shewanella oneidensis
[94]. Due to the ability of coordinating cell behavior at the
population level, the QS devices such as lux, las and rhl have
been combined with other genetic circuits to form various
QS-based logic gates.
QS devices can be used as “wires” to combine genetic
circuits to produce more complex computations in space, as
shown by Tamsir etal. [95]. Firstly, based on previous work
[96], they constructed the simplest NOR gates from NOT
gates with the addition of a repressor. The inputs and the
outputs of the NOR gates were designed to act as promoters
to form multiple gates. Secondly, three NOR gates and a
buffer gate in four separate E. coli cells were wired together
to form an XOR gate via Las AHL and Rhl AHL from P.
aeruginosa PAO1 (Fig.6a).
Different from the logic gates which solely rely on
two exogenously added signals such as IPTG and aTc
[97], Shong etal. [98] developed a synthetic AND gate in
response to the endogenous AHL signal and exogenously
added IPTG or aTc (Fig.6b). The esa QS system from Pan-
toea stewartii was applied to obtain the endogenous Esa
AHL signal to avoid the disadvantages of the lux QS system.
They showed that the downstream gene would not express
without a second exogenous signal, hence demonstrating the
function of this QS-dependent AND gate.
An AND logic gate combined with the lux QS system was
constructed in Shewanella oneidensis to realize the appli-
cation of a logic gate in microbial fuel cells (MFCs) [99].
They firstly integrated the IPTG responding module, a QS
module and an output module (reporter or target gene mtrA)
to form a synthetic AND gate (Fig.6c) to control extracel-
lular electron transfer in S. oneidensis. When the Lux AHL
concentration reaches a critical threshold and binds with
LuxR protein to facilitate the activation of promoter Plux,
S.Wu et al.
1 3
accompanied by the IPTG addition to release the inhibition
of Ptac by LacI protein, this AND logic gate will be switched
on to start extracellular electron transfer.
As stated earlier in “QS-based logic gates”, QS devices
have been widely integrated with the synthetic genetic toggle
switches in metabolic engineering to dynamically regulate
and control the gene expression responsible for the desired
production. As the cell’s physiological state affects meta-
bolic regulation, the stationary phase sensing system and
a QS system were combined by He etal. [100] to obtain
an auto-induced AND gate for monitoring cell growth and
polyhydroxybutyrate (PHB) production (Fig.6d). PrpoS regu-
lates gene expression in the stationary phase [101] and was
chosen here to control the transcription of HrpS, forming
one of the inputs of the AND gate. The other input was from
a QS system that controlled the expression of HrpR. Pro-
moter PhrpL, controlled by the complex of HrpR and HrpS,
was the output.
As a broadly shared paradigm, the LuxI/LuxR-type QS
system has been integrated with synthetic genetic oscillators,
genetic toggle switches, and logic gates, revealing a wide
range of possibilities and potential applications. It should be
emphasized that, although some success has been achieved
in applying QS-based engineering to specific microbes
within the scope of monoclonal synthetic biology, complica-
tions arising from factors such as metabolic load and toxicity
of metabolites could seriously limit what a single microbe
can achieve. Therefore, we anticipate that, following some
of the existing studies reviewed in this section, more explo-
rations will be published on the engineering of microbial
consortia with QS-based synthetic genetic circuits, as a prac-
tice of synthetic ecology, which realize the population-level
synchronization in synthetic communities to overcome the
limit of single species.
QS applications inGram‑positive bacteria
Gram-positive bacteria can also apply their own QS mech-
anism to regulate gene expression at the population level
dynamically. Different from the AHLs adopted in the QS
systems in Gram-negative bacteria, QS of Gram-positive
bacteria is dependent on AIPs, also known as pheromones
[3, 102]. The Gram-positive bacteria can thus sense AIPs to
regulate their own metabolism in a changing environment
[103]. Under certain conditions, AIPs of Gram-positive bac-
teria are produced in the cytoplasm and then secreted by the
oligopeptide transport system to the extracellular medium.
Thereafter, they are either detected at the bacterial surface
by the extracellular pathway or re-internalized by the intra-
cellular pathway [32].
As shown in “QS applications in Gram-negative bac-
teria”, the QS mechanisms in Gram-negative bacteria
(including lux, las QS system and so on) are relatively well
Fig. 6 The applications of QS-based logic gates. a Genetic circuit of
an XOR gate with three NOR gates and a buffer gate in four separate
E. coli colonies. Arabinose (Ara) and anhydrotetracycline (aTc) are
inputs and expression of LasI is the output for the first NOR gate in
cell 1. Based on the first LasI input, Ara and aTc are regarded as the
second inputs for the cell 2 and cell 3. The output of cell 2 and cell 3
is the expression of RhlI. The buffer gate responses to the RhlI input
to express the reporter gene (YFP). b Schematic diagram of QS-
dependent AND logic gate genetic circuits in P. stewartii. c Genetic
circuits schematic diagram of the AND logic gate in S. oneidensis.
The promoter Ptac is inhibited by LacI protein, and it can be relieved
by IPTG addition in IPTG responding module. The AND logic gate
functions by the combination of the IPTG responding module and QS
regulation of LuxR–AHL complex (QS module). d Schematic dia-
gram of the AND logic gate with the control of HrpR and HrpS in
E. coli
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
understood and have been shown to be applicable to the
design and construction of genetic toggle switches, oscil-
lators and logic gates. In contrast, QS in Gram-positive
bacteria still has a number of unknown mechanisms [104],
and there is much less research on the application of QS in
Gram-positive bacteria than that of Gram-negative bacte-
ria [41]. Recently, Marchand etal. [105] reported the first
construction of a synthetic QS system to realize cell–cell
communication among Gram-positive bacteria. They incor-
porated the agr QS system of Staphylococcus aureus into
Bacillus megaterium to synthesize a genetic circuit for
monitoring cell growth. In this circuit, the original P2 pro-
moter for the expression of agrB, agrD, agrC, and agrA
genes was replaced with the PxylA promoter. The AgrD pro-
tein was transported by ArgB and SipM protein to the cul-
ture medium to become mature AIPs, which would then be
recognized by a two-component system (TCS). The TCS
consisted of the AgrC receptor and the AgrA transcriptional
activator which were used to regulate the P3 promoter and
the expression of the target gene such as gfp reporter gene.
However, different from the aforementioned work [105],
most of the other reported AIPs systems of Gram-positive
bacteria are based on the up-regulation or down-regulation
of their own native QS devices as opposed to “borrowing”
one from a different species, as summarized in Table2.
These QS systems are disrupted and manipulated in their
own specific bacteria. Compared with the AHLs, which can
diffuse through the cell membrane freely, the secretion of
AIPs requires the aid of the oligopeptides transport system
[103]. Besides, it should be taken into consideration that the
rates of diffusion of the larger oligopeptides are slower than
the smaller AHLs, especially in a solid culture [31]. These
differences between AHLs and AIPs are arguably among the
reasons for fewer studies on the QS application with Gram-
positive bacteria.
QS forpopulation‑level control based
oninterspecies communication
QS signals can either be used by bacteria to form coopera-
tion or exploited by individuals which do not secrete them;
the latter is termed as cheating phenotypes [129]. QS cheat-
ing in microbes is one of the most important parts of QS
research. As listed in Table1, achievements on cheating
phenotypes and AHL-based social interactions have been
Table 2 Recent QS applications in Gram-positive bacteria with native devices
Pathways AIPs Bacteria species Function controlled References
Extracellular pathway Agr type peptides Clostridium botulinum Neurotoxin production and sporulation [106]
Clostridium acetobutylicum Granulose formation, sporulation [107]
Listeria monocytogenes Population dynamics in soil [108]
Clostridium perfringens Virulence and toxin production [109]
Staphylococcus epidermidis Biofilms and infection [110]
Clostridium perfringens Toxin production and virulence [111]
peptides that contain Gly–Gly motifs Streptococcus thermophilus Production of Blp Bacteriocins [112]
Streptococcus pneumoniae Competence development [113]
Streptococcus pneumoniae Competence control [114]
Streptococcus mutans Competence control [115]
Streptococcus pneumoniae Competence control [116]
Intracellular pathway Rap/NprR/PlcR/PrgX (RNPP family) Bacillus Competence control (Rap) [117]
Bacillus cereus group protease production in sporulation
(NprR)
[118]
Bacillus cereus Necrotrophism (NprR) [119]
Bacillus cereus Virulence regulation (PlcR) [120]
Bacillus cereus and Bacil-
lus thuringiensis Virulence and necrotrophic properties
(NprR)
[121]
Enterococcus faecalis Regulation of conjugation (PrgX) [122]
Rgg-like family Streptococci mutans Competence control [123]
Streptococci genus cross-talk between these different
SHP/Rgg systems
[124]
Streptococcus genus Competence control [125]
Streptococcus thermophilus Competence control [126]
Streptococci genus Competence control [127]
Streptococcus mutans Genetic competence [128]
S.Wu et al.
1 3
extensively reviewed by Whiteley etal. [4] and Asfahl etal.
[40]. Different from the intra-species signal responses which
are mainly dependent on AHLs and AIPs (as reviewed in
QS applications in Gram-negative bacteria” and “QS appli-
cations in Gram-positive bacteria”), the signals for inter-
species communication are mainly autoinducer 2 (AI-2)
[130132] and indole [133, 134] that regulate cooperation
and competition in microbial communities, which is the
focus of this section.
AI‑2‑based communication
AI-2 is a product of the LuxS enzyme, which widely exists
in Gram-positive and Gram-negative bacteria, and even in
fungi [3]. LuxS enzymes synthesize 4,5-dihydroxy-2,3-pen-
tanedione (DPD), which can be regarded as the precursor
of AI-2 [135]. DPD is a by-product of the S-adenosyl-
methionine (SAM) metabolism, which is included in the
activated methyl cycle (AMC). SAM can transfer methyl
groups to methyl-transferases and substrates to produce
S-adenosylhomocysteine (SAH). Catalyzed by a series
of relevant protease, SAH can be converted to homocyst-
eine and DPD [136]. DPD is a highly active molecule that
can spontaneously cyclize into different DPD derivatives,
which can be identified as a signal molecule, AI-2, by dif-
ferent bacteria [137]. Chen etal. [131] certified that AI-2
produced by Vibrio harveyi contains boron. In contrast,
the AI-2 signals of S. typhimurium and E. coli [138] are
non-borated cyclized. There are existing reviews [136, 137]
which explain the mechanism of the AI-2 signaling systems
at length.
Xavier etal. [139] testified that AI-2 can mediate two-
way communication between E. coli and Vibrio harveyi in
co-culture. The AI-2 produced by E. coli can be sensed by
V. harveyi to induce bioluminescence, and reciprocally, the
AI-2 produced by V. harveyi can be detected by E. coli to
regulate its Lsr system. Armbruster etal. [140] found that
Haemophilus influenzae and Moraxella catarrhalis, which
are responsible for one of the common childhood infections
named otitis media, have reciprocal effects on biofilm for-
mation via the AI-2 QS signal. They pointed out that the
former promotes the biofilm formation of the latter. This
and the other studies show the potential of the exploration on
AI-2-based control to further the understanding of interac-
tions between microbes and hosts and to develop strategies
to influence the physiological and biochemical functions of
various pathogenic bacteria for curing the relevant diseases.
Indole‑based communication
With l-tryptophan as the reactant, indole is synthetized
by tryptophanase (TnaA) in many bacteria [141]. More
recently, indole is regarded as a signaling molecule which
is relevant to various bacterial physiology, such as bio-
film formation [142], plasmid stability [143], popula-
tion-based resistance [144], virulence [145], persister
formation [146], spore formation [147], and cell division
[148] in indole-producing bacteria. Yee etal. [149] had
studied biofilm formation with indole-producing bacteria
(E. coli) and non-indole-producing bacteria (P. fluores-
cens), where indole was converted to isoindigo by toluene
o-monooxygenase (TOM), with the TOM gene introduced
from the soil bacterium Burkholderia cepacia G4. Their
results indicated that E. coli was present in a higher den-
sity when co-cultured with P. fluorescens which expressed
TOM. Han etal. [150] proposed that indole oxidation by
TOM will increase the electricity generation in an E. coli-
catalyzed microbial fuel cell. Lee etal. [142] reported that
indole increases the biofilm formation of P. aeruginosa
that does not synthesize indole. Also, indole derivatives
(Indole-3-acetaldehyde) from pathogen Rhodococcus sp.
BFI 332 have been verified for inhibition to biofilm forma-
tion of E. coli O157:H7 [151]. Indole and its derivative
7-benzyloxyindole (7BOI) were investigated for their inhi-
bition to the virulence of S. aureus [152]. Lee etal. [153]
found that indole influences the growth, biofilm formation,
antibiotic tolerance, and motility of Agrobacterium tume-
faciens. Chu etal. [154] investigated the interaction and
competitiveness between E. coli and P. aeruginosa in a
mixed culture. They concluded that the major indole-based
protection for the growth of E. coli in the mixed culture
was due to direct inhibition of QS-based virulence factors,
such as pyocyanin and elastase, in P. aeruginosa. Focus-
ing on the host-microbe interactions of various bacteria
and Caenorhabditis elegans, Lee etal. [155] concluded
that indole and its derivatives will influence the egg-lay-
ing behavior, chemotaxis, and the survival of C. elegans.
Further details and examples of indole-based QS can be
found in several reviews, such as the one by Lee etal.
[133] which covered at length indole-producing bacteria
and the mechanisms and applications of the signaling sys-
tems based on indole and its derivatives. Recently, the per-
spectives on indole signaling systems have been expanded
from interspecies to inter-kingdom. For example, Lee etal.
[33] and Tomberlin etal. [156] provided overviews on
how indole and its derivatives affect various physiological
activities in fungi, insects, plants, and animals.
The functioning of AI-2 and Indole, as two main sign-
aling molecules facilitating the inter-species communica-
tion, greatly expand the presence of QS in the microbial
world. As the understanding of their mechanisms deepens,
one would expect that engineering and medical innova-
tions leveraging such knowledge will start to emerge,
despite their current relative insignificance.
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
QS applications inpotential clinical
therapies
Biolm formation andinhibition
Many bacteria have a tendency to be organized in aggre-
gates, commonly adhering to surfaces to form biofilms
[157]. Compared to the free-living counterparts, there are
some unique properties in bacteria forming biofilms, such
as antimicrobial tolerance [158]. In fact, biofilms forma-
tion has been widely regarded as one of the most impor-
tant virulence factors for microbial toxicity and infections
[159]. When there is a biofilm for bacteria within the
human host, the infections will be hard to treat.
Many studies have reported that the QS of AI-2/luxS and
DSF-based QS systems play an important role in biofilm
development and disassembly [4, 42, 160, 161]. From the
studies using flow-cell systems [160], QS and biofilms have
been shown to be inextricably linked. Lebeer etal. [162]
conducted the first investigation on the relationship between
the LuxS and biofilm formation in Lactobacillus rhamnosus
GG, which is one of the probiotics for human. They found
that LuxS enzyme is crucial for the gastric stress resistance
and the metabolism of L. rhamnosus GG. Sun etal. [163]
found that the over-expression of luxS or AI-2 supplementa-
tion enhanced biofilm formation of Bifidobacterium longum
NCC2705 by about 50%. Furthermore, exogenously addition
of signaling molecule AI-2 was found to promote the biofilm
formation of P. aeruginosa PAO1 [164], Helicobacter pylori
[165], and Staphylococcus epidermidis [166]. Laganenka
etal. [167] showed that self-produced AI-2 can mediate
autoaggregation of E. coli to enhance bacterial stress resist-
ance and promote biofilm formation. Papenfort etal. [168]
discovered 3, 5-dimethylpyrazin-2-ol (DPO) in Vibrio chol-
erae which can be regarded as a new QS signal that regulates
biofilm formation and virulence. Liu etal. [169] identified
that d-Ribose can be applied to decrease the activity of AI-2
to inhibit biofilm formation of Lactobacillus paraplantarum
L-ZS9. Besides, based on the DSF QS systems, Ryan etal.
[170] proposed that DSF underpins the interspecies signal-
ing between Stenotrophomonas maltophilia and P. aerugi-
nosa, with the latter influencing the former’s biofilm forma-
tion. Dean etal. [171] demonstrated that the Burkholderia
diffusible signal factor (BDSF) plays an important role in the
inhibition and dispersion of biofilm formed by Francisella
novicida. It has also been shown that DSF signaling regu-
lates many functions that contribute to biofilm formation
in Stenotrophomonas maltophilia [172] and Helicobacter
pylori [173]. What’s more, DSF-based QS systems can be
used to regulate antibiotic tolerance (e.g., [174]) and the
production of virulence factors (e.g., [175]); this area has
been reviewed at length [176].
Given the close connections between QS and biofilm
formation, the development of novel antimicrobial thera-
pies by QS inhibitors has attracted extensive attention from
researchers [177]. QS inhibitors are functioned by the deg-
radation of QS signals (quorum quenching) [178] or some
other QS-blocking approaches. Shen etal. [179] synthesized
a series of structural analogues of the substrate S-ribosyl-
homocysteine (SRH) and a 2-ketone intermediate to inhibit
LuxS enzyme. Zhang etal. [180] proved that two small pep-
tides, 5411 and 5906, could inhibit the AI-2 activity and
influence biofilm formation and virulence of Edwardsiella
tarda. Ni etal. [181] reviewed seven approaches to inhibit-
ing QS pathways. Brackman etal. [182] certified that cinna-
maldehyde and cinnamaldehyde derivatives can interact with
the AI-2 QS signal by reducing the DNA-binding ability
of LuxR. Brackman etal. [183] investigated the relation-
ship between the susceptibility of biofilms to antibiotics and
the antibiofilm effect of quorum sensing inhibitors (QSI)
invitro and invivo model systems. Christensen etal. [184]
discovered, by an high-throughput cell-free screen, three
AHLs inhibitors which can be used as potential therapeutic
agents for virulence and microbial infections. O’Loughlin
et al. [185] reported that the meta-bromo-thiolactone
(mBTL) can not only inhibit the production of pyocyanin
and biofilm formation but also reduce the activity of two
QS receptors, LasR and RhlR, in P. aeruginosa. Starkey
etal. [186] identified several compounds as QSI (all with
the structure of benzamide-benzimidazole) which can inhibit
the expression of the mvfR QS system which is one of the
key reasons for multidrug-resistant and antibiotic-tolerant
infections. Ouyang etal. [187] also reported that quercetin
can be applied to inhibit biofilm formation and virulence
factors in P. aeruginosa.
Consortia‑based therapies forP. aeruginosa
infection
Pseudomonas aeruginosa, a multidrug resistant pathogen,
can cause disease in plants and animals, including humans
[188]. Biofilms of P. aeruginosa can cause chronic oppor-
tunistic infections, especially for immunocompromised
patients and the elderly. These biofilms also appear to pro-
tect the bacteria from traditional antibiotic therapies [189].
Therefore, research on the discovery of new treatments, such
as consortia-based therapies against P. aeruginosa is much
needed. In particular, engineering microbial consortia with
genetic circuits based on QS devices to inhibit biofilm for-
mation offers a potentially attractive therapeutic technique
to deal with the infectious pathogens.
Taking the potential applications in bioremediation
(among other applications) into consideration, Hong
etal. [23] combined the LasI/LasR-type QS system from
P. aeruginosa with biofilm dispersal genes to control
S.Wu et al.
1 3
biofilm displacement in a microfluidic device. The engi-
neered microbial consortia consisted of disperser cells
and initial colonizer cells. In a disperser cell, the LasI
protein which is the precursor of the signaling molecule
Las AHL is produced continuously, and the biofilm-dis-
persing Hha13D6 protein is encoded when the relevant
gene is induced by IPTG. In an initial colonizer cell, the
LasR protein is encoded, which couples with Las AHL
to form a complex to promote the expression of another
biofilm-dispersing protein, BdcAE50Q (Fig.7a).
Saeidi etal. [190] integrated the LasI/LasR-type QS
system, a lysing device for engineered E. coli, and pyocin
for killing P. aeruginosa. The Las AHL is produced by P.
aeruginosa and detected by the engineered E. coli. When
the Las AHL concentration reaches a threshold, the Las
LasR–AHL complex promotes the transcription of PluxR
and then facilitates the expression of pyocin S5 and lysis
E7 genes. E7 lysis protein will accumulate and then lyse
E. coli cell to release S5 pyocin to inhibit biofilm forma-
tion and kill P. aeruginosa (Fig.7b).
In a further study, Gupta etal. [191] integrated a secre-
tion module to the genetic circuit “sense-kill” system for
P. aeruginosa. They applied a novel pathogen-specific
bacteriocin CoPy, the combination of Colicin E3 and
Pyocin S3 to kill P. aeruginosa. What’s more, they uti-
lized a secretion tag, FlgM, to transport the bacteriocin
CoPy into the culture to kill P. aeruginosa (Fig.7c).
Hwang etal. [192] combined the LasI/LasR-type QS
system, motility control and a killing device to form a
novel genetic circuit engineered into E. coli to kill P.
aeruginosa. The Las LasR–AHL complex promotes the
expression of gene lasI, gene cheZ (controlling the motil-
ity of E. coli toward P. aeruginosa), gene Dnasel (control-
ling biofilm degradation of P. aeruginosa), and gene mcsS
(killing P. aeruginosa) (Fig.7d).
Based on a previous work [190], Hwang etal. [193]
introduced an auxotrophic marker in E. coli to avoid the
horizontal gene transfer of the antibiotic resistance to
other bacteria. The alr and dadX genes which help the
interconversion of d-alanine and l-alanine were knocked
out in the novel engineered E. coli. To construct a modi-
fied “sense-kill” system for P. aeruginosa, they first com-
plemented the auxotrophic E. coli with an alr + plasmid
(pEaak) to ensure the growth and other physiological
activities and then added the dspB gene to the previous
genetic circuits (Fig.6b) to inhibit the biofilm forma-
tion more efficiently. The dspB gene encodes dispersin B
(DspB), an anti-biofilm enzyme for degrading mature bio-
films. It was proven that combining the dspB and pyoS5
genes together to disassemble biofilm formation is an
efficient strategy to kill P. aeruginosa (Fig.7e).
Fig. 7 The applications of QS devices relevant to biofilm formation. a
Diagram of genetic circuits of disperser cell and initial colonizer cell.
b Schematic of genetic circuit coupling QS, killing, and lysing sys-
tems to kill P. aeruginosa. c Genetic circuit architectural of “sense-
kill” system of P. aeruginosa with transport system of the bacteriocin
CoPy. d Schematic of engineering E. coli to sense, migrate and kill P.
aeruginosa. e Diagram of novel “sense-kill” genetic circuit coupling
QS, dspB and pyoS5 gene (for killing), and lysing systems to kill P.
aeruginosa
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
Probiotic therapies fordiseases relevant togut
microbiota
Gut microbiota has been shown to clearly relate to a range
of diseases and conditions of human, such as type 2 diabe-
tes [194], cardiovascular disease [195], clostridium difficile
infection (CDI) [196], epithelial tumors [197] and obesity
[198]. With the large quantities of antibiotics widely used
in the past decades, antibiotic resistance is currently ubiqui-
tous and hard to deal with, especially in the human gut [20,
199, 200]. Much research has thus been focusing on finding
alternative antimicrobial therapies. Taking S. typhimurium,
enterohaemorrhagic E. coli (EHEC) and Clostridium difficile
as representative microbes, Bäumler etal. [60] reviewed the
interactions among the microbiota, the host and these three
pathogenic bacteria when treated by antibiotics. Antibiot-
ics treatment appears to increase free sialic acid (from the
host) and succinate (from the microbiota) level. The elevated
sialic acid and succinate in turn promote the expansion of
the S. typhimurium and C. difficile, which will do harm to
the intestinal epithelium cells (IECs). Besides, EHEC was
found to use a fucose-sensing signaling-transduction QS
system [201] to avoid the nutrient competition with com-
mensal E. coli. To avoid the defect of antibiotics treatment
leading to antibiotic resistance, multiple attempts have been
made to develop probiotic therapies which utilize probiotic
bacteria such as lactic acid bacteria [54] to serve as vectors
for delivery of drug and signaling molecules [202]. As one
of the most important ways for cell–cell communication,
QS devices hold enormous potential in sensing the patho-
gens (stated in “Consortia-based therapies for P. aeruginosa
infection”) for probiotic therapies.
Diarrhoeal diseases, caused by the invasion of Vibrio
cholera, are nightmares for both children and adults [203].
Co-culturing the Ruminococcus obeum and Vibrio cholera
in AKI medium, Hsiao etal. [204] found that the patho-
genicity of V. cholerae was reduced by AI-2 of R. obeum.
The results of experiments in the gnotobiotic mice model
illustrated that the virulence of V. cholerae was regulated via
a novel regulatory pathway in R. obeum. It relates to a new
mechanism based on the VqmA virulence regulator rather
than the known pathway based on HapR.
Considering that the gut microbiota mainly includes
Bacteroidetes and Firmicutes, Thompson etal. [27] chose
these two microbes to investigate the influence of AI-2 on
the ratio of them. Firstly, they used streptomycin to induce
gut dysbiosis, which had previously been applied to inves-
tigate the relationship between streptomycin treatment and
colonization resistance in intestinal E. coli [205]. They sub-
sequently engineered E. coli to manipulate the AI-2 level in
the mouse intestine and investigated the influence on strep-
tomycin-induced dysbiosis. By increasing the level of AI-2,
the ratio of Firmicutes and Bacteroidetes was increased so
as to relieve the strong effect of the antibiotic and restore the
dysbiosis (Fig.8a). Analogously, Xavier etal. also proposed
that AI-2 can make some difference on the composition of
gut microbiota in mouse [206].
Lactococcus lactis and Enterococcus faecalis are ubiq-
uitous in human gastrointestinal tract [207]. Borrero etal.
[208] proposed using L. lactis, generally recognized as safe
(GRAS) for human, to kill E. faecalis which is responsible
for hospital-acquired infections such as enterococcal infec-
tions [209]. In this bi-directional system, L. lactis produces
three antimicrobial peptides (AMPs), enterocin A, hiracin
JM79 and enterocin P, to inhibit the growth of E. faeca-
lis, including vancomycin-resistant enterococcus (VRE)
strains. E. faecalis produces and secrets the sex pheromone
cCF10 (as an AIP) to be detected by the engineered L.
lactis (Fig.8b). When the concentration of the sex phero-
mone cCF10 expressed by E. faecalis reaches the thresh-
old, it will be imported by PrgZ protein and oligopeptide
permease (Opp) system of the engineered L. lactis into its
cytoplasm. cCF10 will then integrate with PrgX protein to
form a PrgX–cCF10 complex which increases RNA poly-
merase access to PQ [104] and enhances the expression of
the downstream antimicrobial peptides to kill E. faecalis.
As concluded by Coyte etal. [210], understanding the
interactions between microbes, especially for the competi-
tion and cooperation among pathogens and probiotics, is
key to revealing the mechanisms of gut microbiota-related
diseases. To understand these complex processes, the studies
reviewed above show that it is important to recognize the
Fig. 8 The applications of QS devices in the gut microbiota. a Bal-
ance of the gut microbiota. Once treated by antibiotics such as strep-
tomycin, Firmicutes and AI-2 producing will reduce, while metabo-
lite such as sialic acid, succinate and fucose will increase in the gut
microbiota. Artificially increasing the levels of AI-2 produced by
engineered E. coli reliefs the dysbiosis, increases the ratio of Firm-
icutes, and reverses state partially. b Diagram of genetic circuits of
applying Lactococcus lactis to sense and kill Enterococcus
S.Wu et al.
1 3
QS signaling molecules as the language or bonds that link
together the members of the microbiome, and also as the
bridge for between the bacteria and the host [211].
Therapies based onCRISPR‑Cas immune systems
Bacteria often suffer from invasion by foreign mobile genetic
elements such as bacteriophage infestation [212] and plas-
mids conjugation [213] which forms a basis for developing
potential therapies to treat human diseases caused by patho-
genic bacteria. Bacteria possess natural defense systems,
named CRISPR-Cas immune systems [76], to reject foreign
phages or plasmids (Fig.9). The regulation of CRISPR-Cas
immune systems can occur during different stages of an inte-
grated network which involves pre-emptive warning, first
contact, breaking the silence, detecting infection, and dedi-
cated regulators [214]. Due to infections of phage increase at
high cell density [28], QS devices have much potential to be
applied to monitor the infections [29]. It is costly to continu-
ously defend the whole CRISPR-Cas immune systems with
an integrated network of various sensors [14]. Therefore,
combining with QS devices, microbes can be “instructed”
to regulate CRISPR-Cas immune systems only at high cell
density, hence minimizing the cost. This is in line with the
work of many researchers.
In response to phage and plasmids invasion, this is in line
with the work of Rossmann etal. [56] which proposed that
AI-2 regulates enterococcal pathogenicity and induces the
horizontal gene transfer (HGT) of virulence genes by con-
jugation or transformation from phages to the commensal
enterococci.
Høyland-Kroghsbo etal. [59] demonstrated that the
CRISPR-Cas activity can be regulated by QS devices in the
pathogen P. aeruginosa. The type I-F CRISPR-Cas system,
LasI/LasR- and RhlI/RhlR-type QS devices were utilized
to analyze the consequence of QS regulations on CRISPR-
Cas activity. When lasI and rhlI genes were knocked out,
the mutant exhibited obvious decreases in the expression of
CRISPR-Cas relative to the wild type. Also, CRISPR-Cas
activity restored to wild type levels when certain auto-induc-
ers were artificially replenished. Therefore, it is possible to
suppress the CRISPR-Cas immune system by QSIs as a cost-
effective way to promote the killing of the pathogens by the
phage.phage therapies.
Patterson etal. [215] combined the SmaI/smaR-type QS
system and homologs of the LuxI/LuxR-type QS system
with type I-E, I-F, and III-A CRISPR-Cas systems, respec-
tively, to investigate the regulation effect of QS on HGT in
Serratia sp. ATCC39006. The signaling molecule Sma AHL
is produced by smaI gene and bonded with SmaR protein
to form a SmaR–AHL complex. When the concentration
of Sma AHL, Lux AHL homologs, is low at low cell den-
sity, the SmaR transcriptional regulator will act as a DNA-
binding repressor for the CRISPR-Cas operon. As the cell
density increases, Sma AHL accumulates in the culture and
eventually reaches sufficiently high concentrations, and the
SmaR–AHL complex will then inhibit the DNA binding
activity of SmaR, which causes the expression of CRISPR-
Cas to increase (Fig.9).
The population-level resistance of bacteria upon invasion
by foreign phages or pathogenic bacteria is commonly con-
sidered as important for maintaining the healthy state of the
microbiome [214]. The development QS-based CRISPR-Cas
technologies such as those reviewed above thus have the
potential to bring useful additions to the toolbox for realizing
population-level resistance.
Mathematical modelling forQS applications
Complementing experimental explorations and supported by
the richness of biological information, a variety of synthetic
genetic circuits and established databases [216], mathemati-
cal modelling has been widely applied in systems biology
and synthetic biology to achieve systematic understandings
of cellular behavior [217] and to optimize TYR for desir-
able products in engineering applications [218]. In particu-
lar, various modeling methods, such as flux balance analysis
(FBA) [219], dynamic flux balance analysis (DFBA) [220],
and sensitivity analysis have been combined with ordinary
differential equations (ODE) [221223] to construct math-
ematical models for QS. As listed in Table1, there exist
comprehensive reviews of deterministic and stochastic mod-
els and modelling approaches for QS devices, particularly
of their molecular mechanisms. Not repeating these exist-
ing reviews, this section is intended to focus on the typi-
cal approaches and recent achievements of mathematical
Fig. 9 The schematic of QS regulation on CRISPR-Cas systems in
Serratia. Mechanisms have been described in the text. The more spe-
cific introduction for the mechanisms of CRISPR-Cas immune sys-
tems can be found in the review paper (Patterson etal. [214])
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
modelling of the applications of QS for bacterial population
control as reviewed earlier in this work.
QS applications modelling inmonoclonal synthetic
biology
As a commonly shared paradigm for QS modelling, the
LuxI/LuxR-type QS has been modelled by a number of
researchers. To predict the function of the circuit illustrated
earlier in Fig.2a (and explained in “QS-based synthetic
genetic oscillators”), You etal. built a mathematical model
which includes cell growth, cell death, production and deg-
radation of CcdB protein, and change of AHL concentration
in this system [50]. The equations of the model are shown in
Eqs.1a1c, which are derived following five assumptions:
(a) Without IPTG induction, cell density changes will fol-
low the logistic model;
(b) When induced by IPTG, the rate of cell death will be
proportional to the concentration of CcdB protein;
(c) The generation rate of CcdB protein is proportional
to the concentration of AHL, and the intracellular and
extracellular AHL concentrations are equal;
(d) The generation rate of AHL is proportional to cell den-
sity;
(e) Degradation of CcdB protein and AHL follows a first-
order kinetics.
A number of QS models similar to the above have sub-
sequently been developed, making predictions based on the
capture of the interactions between cell density, concentra-
tion of AIs, complex of AIs and their correspond receptors
[44].
In principle, QS is not only dictated by reactions of vari-
ous molecules but also affected by diffusion. Basu etal.
[224] designed a synthetic multicellular system with QS
devices, which consists of “sender” and “receiver” cells. Due
to the diffusion of AHL, the concentration of AHL gener-
ated by the “sender” decreases gradually from the cell to
the periphery. Consequently, the “receiver” cells in different
regions respond to different concentrations of AHLs and
express different colors of fluorescent protein, thus forming
different colors and different ring-like patterns.
(1a)
dN
dt
=kN
(
1
N
Nm)
d[E]N
,
(1b)
d[A]
dt
=vANdA[A]
,
(1c)
d[E]
dt
=kE[A]dE[E]
.
Building on the two models mentioned above, Anesiadis
etal. [82] constructed a mechanistic model (Eqs.2a2e)
for investigating the dynamics of the genetic circuit includ-
ing a genetic toggle switch (introduced earlier in Fig.5a).
The same research group [83] further integrated the QS
model (Eqs.2a2e) and a DFBA model to maximize serine
production.
As shown earlier in Fig.5e and explained in “QS-based
genetic toggle switches”, a synthetic genetic toggle switch
can be designed and applied for MI production (Eqs.3a3c).
Gupta etal. [8] modified the population control equation
Eq.1a by removing the lysis term and adding a variable
which denotes the fractional Pfk-1 activity to predict the
circuit’s function.
QS applications modelling insynthetic ecology
Based on the aforementioned relatively simple models
for monoclonal synthetic biology, several more complex
models for combinatorial quorum sensing [70] in synthetic
ecology have been reported. Building on the model of You
etal. [50], Balagadde etal. [51] modelled the dynamics of
a synthetic E. coli predator–prey system, which includes
two QS devices (lux QS and las QS) (introduced earlier in
Fig.2b). Further, Song etal. [225], from the group of You,
expanded their model to investigate the spatiotemporal
(2a)
dX
dt
=kX
,
(2b)
d[A]
dt
=vAXdA[A]
,
(2c)
d[R]
dt
=𝜌R[LuxR]2[A]2dR[R]
,
(2d)
d
[L]
dt=𝛼L1
1+
(
[C]
𝛽C
)
n1+𝛼L2 R
n2
(𝜃R)n2+Rn2dL[L]
,
(2e)
dt=𝛼C
1+
[L]
𝛽L
n1dC[C]
(3a)
d
N
dt
=FPfk kN
(
1N
FPfk Nm
),
(3b)
d[A]
dt
=vANdA[A]
,
(3c)
d
FPfk
dt=
(
Kd
(Kd +[A])
2
)
×(vANdA[A])
.
S.Wu et al.
1 3
dynamics of the predator–prey system by adding the fac-
tors of chemical diffusion, cellular motility, and nutrient
consumption. To study the dynamics of emergent genetic
oscillations in a synthetic microbial consortium (intro-
duced earlier in Fig.4c), Chen etal. [78] developed a
model depicting the system with three compartments,
namely the intracellular space of the activator strain, the
intracellular space of the repressor strain, and the extra-
cellular space, to reduce the difficulty of modelling. Scott
etal. [55] combined agent-based modeling and determin-
istic modeling to describe the population-level dynamics
of synchronized oscillations (introduced earlier in Fig.2d).
Based on the models derived from two-strain consortia,
Kong etal. [226] developed models to investigate the
dynamics of three- and four-strain ecosystems induced by
nisin, a QS molecule of Gram-positive bacteria.
QS stochastic modelling
Stochastic models for QS are needed to account for the
natural stochasticity in gene expression. In particular,
when the rates of gene expression are relatively low
and the amount of the reactants is relatively small, the
effects of stochasticity on the system’s behavior cannot
be ignored [227]. It thus becomes necessary to adopt a
stochastic model to more faithfully capture the nature of
genetic circuits such as synthetic genetic oscillators [56,
58]. Tian etal. [228] recoganized the importance of noise
in the switching of bistable systems, such as the QS-based
genetic toggle switch (Fig.5a). They developed quantita-
tive stochastic models for large-scale genetic regulatory
networks by introducing Poisson random variables into
deterministic models to analyze the influence of noise. The
model developed by Baumgart etal. [74] considered the
dynamics of the concentrations of LuxI, I-SceI and GFP,
and was used for conducting robustness analysis of a sto-
chastic process that involved the positive feedback of QS,
negative feedback of plasmid copy number, and intracel-
lular delay in feedback, which were integrated to describe
the oscillator in DNA copy number control (mechanism
shown in Fig.3f).
The modelling studies reviewed in this section show that
lumped-parameter (i.e., well-mixed), deterministic models,
which are relatively simple, can already predict the effect
of applications of QS-based genetic circuits in metabolic
engineering and to offer useful insights on the dynamics of
such systems. On the other hand, more advanced modelling
schemes, such as those taking into account spatial heteroge-
neity and stochasticity, could offer more faithful and more
detailed representation, which would become particularly
important when such complexities play a decisive role in
shaping the behavour and function of a QS-based system.
Summary andfuture perspectives
QS devices have been shown to be able to play a key role
in engineered dynamic control of metabolism and various
microbial physiological activities, such as the TYR (titer,
yield and rate) increase of desirable metabolites, microbiota
synchronization, and regulation of sporulation, virulence,
competence and toxin production. These applications of QS
have been realized by five main types of signaling molecules,
namely AHLs, DSFs, AIPs, AI-2 and indole. Genetic oscil-
lators, toggle switches and logic gates have been constructed
by coupling with AHL-based QS devices in Gram-negative
bacteria. With Gram-positive bacteria, both extracellular
and intracellular pathway AIP-based QS devices have been
developed for controlling physiological activities. AI-2 and
indole, on the other hand, can be regarded as languages
of communication in microbial communities. Engineered
applications of QS in microbes can be either “constructive”
(through introducing new/foreign or enhancing existing QS
mechanisms) or “destructive” (through inhibiting existing
QS mechanisms). In constructive cases, the QS devices
could be either natural or synthetic. Furthermore, a synthetic
QS device can be either constructed partly from naturally
occurring QS modules or synthesized from scratch. Among
other areas, both constructive and destructive QS applica-
tions have found increasing potential in developing clinical
therapies for a range of diseases caused by biofilm forma-
tion, antibiotic resistance and phage invasion.
For QS to be more effective and more widely applicable,
further work is needed to address several general challenges.
First, the target QS modulators must recognize precisely the
corresponding signals in an extremely miscellaneous pool.
Second, compared to the long-range effective electrical sig-
nals [229, 230], quenching problems are common in chemi-
cal signals in QS devices; answering questions such as how
to realize the spatiotemporal and tempo control of signal-
ing molecules is essential, too. Third, there are couplings or
crosstalk between different QS signals and receptors (e.g.,
AHL-based QS and indole-based QS affect each other);
understanding how to decouple these pathways is necessary
and important to achieve their applications with larger scale
genetic oscillators in demanding tasks such as investigat-
ing the population-level dynamics of microbes. Fourth, the
cheating behaviors aggravate the complexity of microbial
communities; the questions of how to constrain cheaters
when desirable and how to exploit cheating behaviors to
inhibit QS are not only theoretically interesting for microbial
ecology but also practically important for the development
of QSIs. Finally, nonlinearities and stochasticity need to be
considered and resolved appropriately in QS modelling.
With synthetic biology making great leaps forward,
the future perspectives of QS applications are expected
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
to keep pace with it. Currently, most of the engineered
dynamic regulation mechanisms build on AHL-based QS
devices of Gram-negative bacteria. In contrast, existing
QS-based interventions in Gram-positive bacteria are
mostly in the form of up-regulating or down-regulating
naturally occurring processes, and there is very limited
research on synthetic QS in this type of bacteria. On the
other hand, Gram-positive bacteria, such as Lactococ-
cus lactis have been viewed as important candidates for
building engineered microbial cell factories and vaccine
delivery systems, and they play an irreplaceable role in
metabolic engineering and medical applications, such as
producing dairy fermentations [231, 232], industrial prod-
ucts and various vaccines [233]. Given their significance,
more research is needed to discover, study and apply the
QS mechanisms in Gram-positive bacteria. In particular,
one may expect that there is great potential to couple cer-
tain synthetic TCS-based or AIP-based QS devices with
the original pathways in Gram-positive bacteria to improve
the TYR of desirable products in Gram-positive cell facto-
ries and to develop future therapeutic systems.
As a broad perspective for future research, the explora-
tion of QS for population-level control of bacteria and its
potential in therapeutic applications may proceed in hori-
zontal and vertical dimensions, respectively. Horizontally,
QS applications can be centered on multi-circuit systems
(the combination of various QS-based genetic circuits),
multi-production (two or more products from one single
cell), and microbial communities (engineering microbiota
to produce one or more products). Vertically, QS applica-
tions in medical therapeutics have the potential to advance
by addressing the interactions between microbes (virus,
bacterium and fungi) and the host [212, 234236]. The
understanding of these interactions will accelerate the
development of more reliable strategies for manipulating
the microbiota against infectious diseases and antibiotic
resistance, which is of great significance for human health.
In the future horizontal and vertical developments of QS
applications, mathematic models are expected to continue
their supporting role to construct QS networks, by not only
embracing nonlinearities and stochasticity, but also inte-
grating across component, circuit, cellular and community
levels and addressing the interactions between microbes
and their hosts and other environments, to offer the power
of making holistic predictions.
Acknowledgements This study was supported by the National Key
Research and Development Project of China (2017YFD0201400), the
National Natural Science Foundation of China (31570089, 31170076),
and the Funds for Creative Research Groups of China (21621004), Dr.
Jianjun Qiao was supported by The New Century Outstanding Talent
Support Program, Education Ministry of China.
Compliance with ethical standards
Conflict of interest The authors declare no competing financial inter-
ests.
References
1. Homer CM, Summers DK, Goranov AI, Clarke SC, Wiesner DL,
Diedrich JK, Moresco JJ, Toffaletti D, Upadhya R, Caradonna
I, Petnic S, Pessino V, Cuomo CA, Lodge JK, Perfect J, Yates
JR 3rd, Nielsen K, Craik CS, Madhani HD (2016) Intracellular
action of a secreted peptide required for fungal virulence. Cell
Host Microbe 19:849–864
2. Erez Z, Steinberger-Levy I, Shamir M, Doron S, Stokar-Avihail
A, Peleg Y, Melamed S, Leavitt A, Savidor A, Albeck S (2017)
Communication between viruses guides lysis-lysogeny decisions.
Nature 541:488–493
3. Melissa B, Miller BL (2001) Quorum sensing in bacteria. Annu
Rev Microbiol 55(1):165–199
4. Whiteley M, Diggle SP, Greenberg EP (2017) Progress in
and promise of bacterial quorum sensing research. Nature
551:313–320
5. Chubukov V, Gerosa L, Kochanowski K, Sauer U (2014) Coordi-
nation of microbial metabolism. Nat Rev Microbiol 12:327–340
6. Holtz WJ, Keasling JD (2010) Engineering static and dynamic
control of synthetic pathways. Cell 140:19–23
7. Venayak N, Anesiadis N, Cluett WR, Mahadevan R (2015)
Engineering metabolism through dynamic control. Curr Opin
Biotechnol 34:142–152
8. Gupta A, Reizman IM, Reisch CR, Prather KL (2017) Dynamic
regulation of metabolic flux in engineered bacteria using a
pathway-independent quorum-sensing circuit. Nat Biotechnol
35:273–279
9. Liu D, Evans T, Zhang F (2015) Applications and advances of
metabolite biosensors for metabolic engineering. Metab Eng
31:35–43
10. Soma Y, Hanai T (2015) Self-induced metabolic state switch-
ing by a tunable cell density sensor for microbial isopropanol
production. Metab Eng 30:7–15
11. Doong SJ, Gupta A, Prather KLJ (2018) Layered dynamic regula-
tion for improving metabolic pathway productivity in escherichia
coli. Proc Natl Acad Sci USA 115(12):2964–2969
12. Liu X, Li XB, Jiang J, Liu ZN, Qiao B, Li FF, Cheng JS, Sun X,
Yuan YJ, Qiao J (2018) Convergent engineering of syntrophic
Escherichia coli coculture for efficient production of glycosides.
Metab Eng 47:243–253
13. Gardner TS, Cantor CR, Collins JJ (2000) Construc-
tion of a genetic toggle switch in Escherichia coli. Nature
403(6767):339–342
14. Elowitz MB, Leibler S (2000) A synthetic oscillatory network of
transcriptional regulators. Nature 403:335–338
15. Win MN, Smolke CD (2007) A modular and extensible rna-based
gene-regulatory platform for engineering cellular function. Proc
Natl Acad Sci USA 104:14283–14288
16. Dueber JE, Wu GC, Malmirchegini GR, Moon TS, Petzold CJ,
Ullal AV, Prather KL, Keasling JD (2009) Synthetic protein scaf-
folds provide modular control over metabolic flux. Nat Biotech-
nol 27(8):753–759
17. Danino T, Mondragon-Palomino O, Tsimring L, Hasty J (2010)
A synchronized quorum of genetic clocks. Nature 463:326–330
18. Papenfort K, Bassler BL (2016) Quorum sensing signal–
response systems in gram-negative bacteria. Nat Rev Microbiol
14:576–588
S.Wu et al.
1 3
19. Diggle SP, Griffin AS, Campbell GS, West SA (2007) Coop-
eration and conflict in quorum-sensing bacterial populations.
Nature 450:411–414
20. Modi SR, Collins JJ, Relman DA (2014) Antibiotics and the
gut microbiota. J Clin Investig 124:4212–4218
21. Flemming HC, Wingender J, Szewzyk U, Steinberg P, Rice SA,
Kjelleberg S (2016) Biofilms: an emergent form of bacterial
life. Nat Rev Microbiol 14:563–575
22. Yan J, Bassler BL (2019) Surviving as a community: antibi-
otic tolerance and persistence in bacterial biofilms. Cell Host
Microbe 26:15–21
23. Hong SH, Hegde M, Kim J, Wang X, Jayaraman A, Wood TK
(2012) Synthetic quorum-sensing circuit to control consortial
biofilm formation and dispersal in a microfluidic device. Nat
Commun 3:613–620
24. Cho I, Yamanishi S, Cox L, Methé BA, Zavadil J, Li K, Gao
Z, Mahana D, Raju K, Teitler I (2012) Antibiotics in early
life alter the murine colonic microbiome and adiposity. Nature
488:621–626
25. Kalia VC (2013) Quorum sensing inhibitors: an overview. Bio-
technol Adv 31:224–245
26. Ma S, Zhou Z (2017) Recent advances in the discovery of pqsd
inhibitors as antimicrobial agents. ChemMedChem 12:420–425
27. Thompson JA, Oliveira RA, Djukovic A, Ubeda C, Xavier KB
(2015) Manipulation of the quorum sensing signal ai-2 affects
the antibiotic-treated gut microbiota. Cell Rep 10:1861–1871.
https ://doi.org/10.1016/j.celre p.2015.02.049
28. Abedon ST (2012) Spatial vulnerability: bacterial arrange-
ments, microcolonies, and biofilms as responses to low rather
than high phage densities. Viruses 4:663–687
29. Semenova E, Severinov K (2016) Come together: Crispr-cas
immunity senses the quorum. Mol Cell 64:1013–1015
30. Hawver LA, Jung SA, Ng WL (2016) Specificity and complex-
ity in bacterial quorum-sensing systems. FEMS Microbiol Rev
40:738–752
31. Monnet V, Gardan R (2015) Quorum-sensing regulators in
gram-positive bacteria: ‘Cherchez le peptide’. Mol Microbiol
97:181–184
32. Monnet V, Juillard V, Gardan R (2014) Peptide conversations
in gram-positive bacteria. Crit Rev Microbiol 42:339–351
33. Lee JH, Wood TK, Lee J (2015) Roles of indole as an interspe-
cies and interkingdom signaling molecule. Trends Microbiol
23:707–718
34. Hmelo LR (2017) Quorum sensing in marine microbial envi-
ronments. Ann Rev Mar Sci 9:257–281
35. Zhou L, Zhang LH, Camara M, He YW (2017) The dsf family
of quorum sensing signals: diversity, biosynthesis, and turno-
ver. Trends Microbiol 25:293–303
36. Xu P (2017) Production of chemicals using dynamic control of
metabolic fluxes. Curr Opin Biotechnol 53:12–19
37. Shong J, Diaz MRJ, Collins CH (2012) Towards synthetic
microbial consortia for bioprocessing. Curr Opin Biotechnol
23:798–802
38. Song H, Ding MZ, Jia XQ, Ma Q, Yuan YJ (2014) Synthetic
microbial consortia: from systematic analysis to construction
and applications. Chem Soc Rev 43:6954–6981
39. Dolinsek J, Goldschmidt F, Johnson DR (2016) Synthetic
microbial ecology and the dynamic interplay between micro-
bial genotypes. FEMS Microbiol Rev 40:961–979
40. Asfahl KL, Schuster M, Gibbs K (2017) Social interactions in
bacterial cell–cell signaling. FEMS Microbiol Rev 41:92–107
41. Choudhary S, Schmidt-Dannert C (2010) Applications of
quorum sensing in biotechnology. Appl Microbiol Biotechnol
86:1267–1279
42. Koo H, Allan RN, Howlin RP, Stoodley P, Hall-Stoodley L
(2017) Targeting microbial biofilms: current and prospective
therapeutic strategies. Nat Rev Microbiol 15:740–755
43. Mukherjee S, Bassler BL (2019) Bacterial quorum sensing
in complex and dynamically changing environments. Nat Rev
Microbiol 17:371–382
44. Goryachev AB (2011) Understanding bacterial cell–cell
communication with computational modeling. Chem Rev
111:238–250
45. Hense BA, Schuster M (2015) Core principles of bacterial auto-
inducer systems. Microbiol Mol Biol Rev 79:153–169
46. Pérez-Velázquez J, Gölgeli M, García-Contreras R (2016) Math-
ematical modelling of bacterial quorum sensing: a review. Bull
Math Biol 78:1–55
47. Boyer M, Wisniewski-Dye F (2009) Cell–cell signalling in bac-
teria: not simply a matter of quorum. FEMS Microbiol Ecol
70:1–19
48. An JH, Goo E, Kim H, Seo YS, Hwang I (2014) Bacterial quo-
rum sensing and metabolic slowing in a cooperative population.
Proc Natl Acad Sci USA 111:14912–14917
49. Goo E, Majerczyk CD, An JH, Chandler JR, Seo YS, Ham H,
Lim JY, Kim H, Lee B, Jang MS, Greenberg EP, Hwang I (2012)
Bacterial quorum sensing, cooperativity, and anticipation of sta-
tionary-phase stress. Proc Natl Acad Sci USA 109:19775–19780
50. You L, Cox RS 3rd, Weiss R, Arnold FH (2004) Programmed
population control by cell–cell communication and regulated
killing. Nature 428:868–871
51. Balagadde FK, Song H, Ozaki J, Collins CH, Barnet M, Arnold
FH, Quake SR, You L (2008) A synthetic escherichia coli preda-
tor–prey ecosystem. Mol Syst Biol 4:187–194
52. Wu F, Lopatkin AJ, Needs DA, Lee CT, Mukherjee S, You L
(2019) A unifying framework for interpreting and predicting
mutualistic systems. Nat Commun 10:242–251
53. Din MO, Danino T, Prindle A, Skalak M, Selimkhanov J, Allen
K, Julio E, Atolia E, Tsimring LS, Bhatia SN (2016) Syn-
chronized cycles of bacterial lysis for invivo delivery. Nature
536:81–85
54. Mays ZJ, Nair NU (2018) Synthetic biology in probiotic lactic
acid bacteria: at the frontier of living therapeutics. Curr Opin
Biotechnol 53:224–231
55. Scott SR, Din MO, Bittihn P, Xiong L, Tsimring LS, Hasty J
(2017) A stabilized microbial ecosystem of self-limiting bac-
teria using synthetic quorum-regulated lysis. Nat Microbiol
2:17083–17091
56. Woods ML, Leon M, Perezcarrasco R, Barnes CP (2016) A sta-
tistical approach reveals designs for the most robust stochastic
gene oscillators. ACS Synth Biol 5:459–470
57. Jörg DJ, Morelli LG, Jülicher F (2018) Chemical event chain
model of coupled genetic oscillators. Phys Rev E 97(3–1):1–11
(032409)
58. Potvin-Trottier L, Lord ND, Vinnicombe G, Paulsson J (2016)
Synchronous long-term oscillations in a synthetic gene circuit.
Nature 538:514–517
59. Tu BP, Mcknight SL (2006) Metabolic cycles as an underlying
basis of biological oscillations. Nat Rev Mol Cell Biol 7:696–701
60. Mondragon-Palomino O, Danino T, Selimkhanov J, Tsimring L,
Hasty J (2011) Entrainment of a population of synthetic genetic
oscillators. Science 333:1315–1319
61. Purcell O, Savery NJ, Grierson CS, Bernardo MD (2010) A
comparative analysis of synthetic genetic oscillators. J R Soc
Interface 7(52):1503–1524
62. Barkai N, Leibler S (2000) Circadian clocks limited by noise.
Nature 403(6767):267–268
63. Hasty J, Dolnik M, Rottschäfer V, Collins JJ (2002) Synthetic
gene network for entraining and amplifying cellular oscillations.
Phys Rev Lett 88(14):1–4 (148101)
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
64. Stricker J, Cookson S, Bennett MR, Mather WH, Tsimring LS,
Hasty J (2008) A fast, robust and tunable synthetic gene oscilla-
tor. Nature 456:516–519
65. Kellogg R, Tay S (2015) Noise facilitates transcriptional control
under dynamic inputs. Cell 160:381–392
66. Tokuda IT, Okamoto A, Matsumura R, Takumi T, Akashi M
(2017) Potential contribution of tandem circadian enhancers to
nonlinear oscillations in clock gene expression. Mol Biol Cell
28(17):2333–2342
67. Wintermute EH, Silver PA (2010) Dynamics in the mixed micro-
bial concourse. Genes Dev 24:2603–2614
68. Xavier JB (2011) Social interaction in synthetic and natural
microbial communities. Mol Syst Biol 7:483–493
69. Chuang JS (2012) Engineering multicellular traits in synthetic
microbial populations. Curr Opin Chem Biol 16:370–378
70. Scott SR, Hasty J (2016) Quorum sensing communication mod-
ules for microbial consortia. ACS Synth Biol 5:969–977
71. Mcmillen D, Kopell N, Hasty J, Collins JJ (2002) Synchronizing
genetic relaxation oscillators by intercell signaling. Proc Natl
Acad Sci USA 99:679–684
72. Waters CM, Bassler BL (2005) Quorum sensing: cell-to-cell
communication in bacteria. Annu Rev Cell Dev Biol 21:319–346
73. Slager J, Kjos M, Attaiech L, Veening JW (2014) Antibiotic-
induced replication stress triggers bacterial competence by
increasing gene dosage near the origin. Cell 157:395–406
74. Baumgart L, Mather W, Hasty J (2017) Synchronized DNA
cycling across a bacterial population. Nat Genet 49(8):1282–1285
75. Prindle A, Samayoa P, Razinkov I, Danino T, Tsimring LS, Hasty
J (2012) A sensing array of radically coupled genetic/’biopixels/’.
Nature 481:39–44
76. Prindle A, Selimkhanov J, Li H, Razinkov I, Tsimring LS, Hasty
J (2014) Rapid and tunable post-translational coupling of genetic
circuits. Nature 508:387–391
77. Levchenko I, Seidel M, Sauer RT, Baker TA (2000) A specific-
ity-enhancing factor for the clpxp degradation machine. Science
289:2354–2356
78. Chen Y, Kim JK, Hirning AJ, Josić K, Bennett MR (2015) Emer-
gent genetic oscillations in a synthetic microbial consortium. Sci-
ence 349(6251):986–989
79. Feist AM, Palsson BO (2010) The biomass objective function.
Curr Opin Microbiol 13:344–349
80. Soma Y, Tsuruno K, Wada M, Yokota A, Hanai T (2014) Meta-
bolic flux redirection from a central metabolic pathway toward a
synthetic pathway using a metabolic toggle switch. Metab Eng
23:175–184
81. Kobayashi H, Kaern M, Araki M, Chung K, Gardner TS, Can-
tor CR, Collins JJ (2004) Programmable cells: interfacing nat-
ural and engineered gene networks. Proc Natl Acad Sci USA
101:8414–8419
82. Anesiadis N, Cluett WR, Mahadevan R (2008) Dynamic meta-
bolic engineering for increasing bioprocess productivity. Metab
Eng 10:255–266
83. Anesiadis N, Kobayashi H, Cluett WR, Mahadevan R (2013)
Analysis and design of a genetic circuit for dynamic metabolic
engineering. ACS Synth Biol 2:442–452
84. Honjo H, Iwasaki K, Soma Y, Tsuruno K, Hamada H, Hanai T
(2019) Synthetic microbial consortium with specific roles des-
ignated by genetic circuits for cooperative chemical production.
Metab Eng 55:268–275
85. Wang EX, Liu Y, Ma Q, Dong XT, Ding MZ, Yuan YJ (2019)
Synthetic cell–cell communication in a three-species consortium
for one-step vitamin c fermentation. Biotechnol Lett 41:951–961
86. Tsoi R, Dai Z, You L (2019) Emerging strategies for engineering
microbial communities. Biotechnol Adv 37(6):1–9 (107372)
87. Nandagopal N, Elowitz MB (2011) Synthetic biology: integrated
gene circuits. Science 333:1244–1248
88. Benenson Y (2012) Biomolecular computing systems: princi-
ples, progress and potential. Nat Rev Genet 13(7):455–468
89. Moon TS, Lou C, Tamsir A, Stanton BC, Voigt CA (2012)
Genetic programs constructed from layered logic gates in sin-
gle cells. Nature 491:249–253
90. Baig H, Madsen J (2017) Simulation approach for timing anal-
ysis of genetic logic circuits. ACS Synth Biol 6(7):1169–1179
91. Nielsen AA, Der BS, Shin J, Vaidyanathan P, Paralanov
V, Strychalski EA, Ross D, Densmore D, Voigt CA (2016)
Genetic circuit design automation. Science 352(aac7341):1–11
92. Hasty J, McMillen D, Collins JJ (2002) Engineered gene cir-
cuits. Nature 420:224–230
93. Li Z, Rosenbaum MA, Venkataraman A, Tam TK, Katz E,
Angenent LT (2011) Bacteria-based and logic gate: a deci-
sion-making and self-powered biosensor. Chem Commun
47:3060–3062
94. Arugula MA, Shroff N, Katz E, He Z (2012) Molecular and
logic gate based on bacterial anaerobic respiration. Chem
Commun 48:10174–10176
95. Tamsir A, Tabor JJ, Voigt CA (2010) Robust multicellular
computing using genetically encoded nor gates and chemical
/`wires/’. Nature 469:212–215
96. Yokobayashi Y, Weiss R, Arnold F (2002) Directed evolution
of a genetic circuit. Proc Natl Acad Sci USA 99:16587–16591
97. Miyamoto T, Razavi S, Derose R, Inoue T (2013) Synthesiz-
ing biomolecule-based boolean logic gates. ACS Synth Biol
2:72–82
98. Shong J, Collins CH (2014) Quorum sensing-modulated and-
gate promoters control gene expression in response to a combi-
nation of endogenous and exogenous signals. ACS Synth Biol
3:238–246
99. Hu Y, Yang Y, Katz E, Song H (2015) Programming the quorum
sensing-based and gate in shewanella oneidensis for logic gated-
microbial fuel cells. Chem Commun 51:4184–4187
100. He X, Chen Y, Liang Q, Qi Q (2017) An autoinduced and-gate
controlling metabolic pathway dynamically in response to micro-
bial communities and cell physiological state. ACS Synth Biol
6:463–470
101. Takayanagi Y, Tanaka K, Takahashi H (1994) Structure of the 5
upstream region and the regulation of the rpos gene of Escheri-
chia coli. Mol Genet Genomics 243:525–531
102. Keller L, Surette MG (2006) Communication in bacteria: an
ecological and evolutionary perspective. Nat Rev Microbiol
4:249–258
103. Papadimitriou K, Alegria A, Bron PA, de Angelis M, Gobbetti
M, Kleerebezem M, Lemos JA, Linares DM, Ross P, Stanton
C, Turroni F, van Sinderen D, Varmanen P, Ventura M, Zuniga
M, Tsakalidou E, Kok J (2016) Stress physiology of lactic acid
bacteria. Microbiol Mol Biol Rev 80:837–890
104. Cook LC, Federle MJ (2014) Peptide pheromone signal-
ing in streptococcus and enterococcus. FEMS Microbiol Rev
38:473–492
105. Marchand N, Collins CH (2016) Synthetic quorum sensing and
cell–cell communication in gram-positive bacillus megaterium.
ACS Synth Biol 5:597–606
106. Cooksley CM, Davis IJ, Winzer K, Chan WC, Peck MW, Minton
PN (2010) Regulation of neurotoxin production and sporulation
by a putative agrbd signaling system in proteolytic clostridium
botulinum. Appl Environ Microbiol 76:4448–4460
107. Steiner E, Scott J, Minton NP, Winzer K (2012) An agr quorum
sensing system that regulates granulose formation and sporu-
lation in clostridium acetobutylicum. Appl Environ Microbiol
78:1113–1122
108. Vivant AL, Garmyn D, Gal L, Piveteau P (2014) The agr commu-
nication system provides a benefit to the populations of listeria
monocytogenes in soil. Front Cell Infect Microbiol 4(160):1–7
S.Wu et al.
1 3
109. Ma M, Li J, Mcclane BA (2015) Structure-function analysis of
peptide signaling in the clostridium perfringens agr-like quorum
sensing system. J Bacteriol 197:1807–1818
110. Yang T, Talgan Y, Paharik AE, Horswill AR, Blackwell HE
(2016) Structure-function analyses of a staphylococcus epider-
midis autoinducing peptide reveals motifs critical for agrc-type
receptor modulation. ACS Chem Biol 11:1982–1991
111. Yu Q, Lepp D, Mehdizadeh GI, Wu T, Zhou H, Yin X, Yu H,
Prescott JF, Nie SP, Xie MY (2017) The agr-like quorum sensing
system is required for necrotic enteritis pathogenesis in poultry
caused by clostridium perfringens. Infect Immun 85:e00975-16
112. Fontaine L, Boutry C, Guédon E, Guillot A, Ibrahim M, Gros-
siord B, Hols P (2007) Quorum-sensing regulation of the pro-
duction of blp bacteriocins in Streptococcus thermophilus. J
Bacteriol 189:7195–7205
113. Håvarstein LS (2010) Increasing competence in the genus strep-
tococcus. Mol Microbiol 78:541–544
114. Mirouze N, Bergé MA, Soulet AL, Mortierbarrière I, Quentin Y,
Fichant G, Granadel C, Noirotgros MF, Noirot P, Polard P (2013)
Direct involvement of dpra, the transformation-dedicated reca
loader, in the shut-off of pneumococcal competence. Proc Natl
Acad Sci USA 110:352–361
115. Reck M, Tomasch J, Wagner-Dobler I (2015) The alternative
sigma factor sigx controls bacteriocin synthesis and competence,
the two quorum sensing regulated traits in streptococcus mutans.
PLoS Genet 11:e1005353
116. Moreno-GãmS Sorg RA, Domenech A, Kjos M, Weissing FJ,
van Doorn GS, Veening JW (2017) Quorum sensing integrates
environmental cues, cell density and cell history to control bacte-
rial competence. Nat Commun 8:854–865
117. Gallego del Sol F, Marina A (2013) Structural basis of rap phos-
phatase inhibition by phr peptides. PLoS Biol 11:e1001511
118. Perchat S, Dubois T, Zouhir S, Gominet M, Poncet S, Lemy C,
Aumont-Nicaise M, Deutscher J, Gohar M, Nessler S (2011) A
cell–cell communication system regulates protease production
during sporulation in bacteria of the bacillus cereus group. Mol
Microbiol 82:619–633
119. Zouhir S, Perchat S, Nicaise M, Perez J, Guimaraes B, Lereclus
D, Nessler S (2013) Peptide-binding dependent conformational
changes regulate the transcriptional activity of the quorum-sen-
sor nprr. Nucleic Acids Res 41:7920–7933
120. Grenha R, Slamti L, Nicaise M, Refes Y, Lereclus D, Nessler S
(2013) Structural basis for the activation mechanism of the plcr
virulence regulator by the quorum-sensing signal peptide papr.
Proc Natl Acad Sci USA 110:1047–1052
121. Slamti L, Perchat S, Huillet E, Lereclus D (2014) Quorum sens-
ing in bacillus thuringiensis is required for completion of a full
infectious cycle in the insect. Toxins 6:2239–2255
122. Chen Y, Bandyopadhyay A, Kozlowicz BK, Haemig HAH,
Tai A, Hu WS, Dunny GM (2017) Mechanisms of peptide sex
pheromone regulation of conjugation in enterococcus faecalis.
Microbiologyopen 6(e492):1–13
123. Mashburn-Warren L, Morrison DA, Federle MJ (2010) A
novel double-tryptophan peptide pheromone controls compe-
tence in streptococcus spp. via an rgg regulator. Mol Microbiol
78:589–606
124. Fleuchot B, Guillot A, Mézange C, Besset C, Chambellon E,
Monnet V, Gardan R (2013) Rgg-associated shp signaling pep-
tides mediate cross-talk in streptococci. PLoS One 8:e66042
125. Parashar V, Aggarwal C, Federle MJ, Neiditch MB (2015) Rgg
protein structure-function and inhibition by cyclic peptide com-
pounds. Proc Natl Acad Sci USA 112:5177–5182
126. Haustenne L, Bastin G, Hols P, Fontaine L (2015) Modeling of
the comrs signaling pathway reveals the limiting factors control-
ling competence in Streptococcus thermophilus. Front Microbiol
6(1413):1–20
127. Talagas A, Fontaine L, Ledesma-García L, Mignolet J, Li de la
Sierra-Gallay I, Lazar N, Aumont-Nicaise M, Federle MJ, Prehna
G, Hols P, Nessler S (2016) Structural insights into streptococcal
competence regulation by the cell-to-cell communication system
comrs. PLoS Pathog 12:e1005980
128. Underhill SAM, Shields RC, Kaspar JR, Haider M, Burne RA,
Hagen SJ (2018) Intracellular signaling through the comrs
system in streptococcus mutans genetic competence. mSphere
3(5):e00444-18
129. Garcíacontreras R, Nuñezlópez L, Jassochávez R, Kwan BW,
Belmont JA, Rangelvega A, Maeda T, Wood TK (2014) Quo-
rum sensing enhancement of the stress response promotes resist-
ance to quorum quenching and prevents social cheating. ISME J
9(1):115–125
130. Bassler BL, Wright M, Showalter RE, Silverman MR (1993)
Intercellular signalling in vibrio harveyi: sequence and function
of genes regulating expression of luminescence. Mol Microbiol
9:773–786
131. Chen X (2002) Structural identification of a bacterial quorum-
sensing signal containing boron. Nature 415:545–549
132. Sedlmayer F, Hell D, Muller M, Auslander D, Fussenegger M
(2018) Designer cells programming quorum-sensing interference
with microbes. Nat Commun 9(1822):1–13
133. Lee JH, Lee J (2010) Indole as an intercellular signal in microbial
communities. FEMS Microbiol Rev 34:426–444
134. Wang D, Ding X, Rather PN (2001) Indole can act as an extracel-
lular signal in Escherichia coli. J Bacteriol 183:4210–4216
135. Bassler BL, Losick R (2006) Bacterially speaking. Cell
125:237–246
136. De Keersmaecker SC, Sonck K, Vanderleyden J (2006) Let luxs
speak up in ai-2 signaling. Trends Microbiol 14:114–119
137. Pereira CS, Thompson JA, Xavier KB (2013) Ai-2-mediated
signalling in bacteria. FEMS Microbiol Rev 37:156–181
138. Xavier KB, Bassler BL (2005) Regulation of uptake and process-
ing of the quorum-sensing autoinducer ai-2 in Escherichia coli.
J Bacteriol 187:238–248
139. Xavier KB, Bassler BL (2005) Interference with ai-2-mediated
bacterial cell–cell communication. Nature 437:750–753
140. Armbruster CE, Hong W, Pang B, Weimer KED, Juneau RA,
Turner J, Swords WE (2010) Indirect pathogenicity of haemophi-
lus influenzae and moraxella catarrhalis in polymicrobial otitis
media occurs via interspecies quorum signaling. Mbio 1:119–121
141. Newton WA, Snell EE (1965) Formation and interrelationships
of tryptophanase and tryptophan synthetases in Escherichia coli.
J Bacteriol 89:355–364
142. Lee J, Jayaraman A, Wood TK (2007) Indole is an inter-species
biofilm signal mediated by sdia. BMC Microbiol 7:1–15
143. Chant EL, Summers DK (2007) Indole signalling contributes to
the stable maintenance of Escherichia coli multicopy plasmids.
Mol Microbiol 63:35–43
144. Lee HH, Molla MN, Cantor CR, Collins JJ (2010) Bacterial char-
ity work leads to population-wide resistance. Nature 467:82–85
145. Jintae L, Can A, Cirillo SLG, Cirillo JD, Wood TK (2010) Indole
and 7-hydroxyindole diminish Pseudomonas aeruginosa viru-
lence. Microb Biotechnol 2:75–90
146. Vega NM, Allison KR, Khalil AS, Collins JJ (2011) Signaling-
mediated bacterial persister formation. Nat Chem Biol 8:431
147. Kim YG, Lee JH, Cho MH, Lee J (2011) Indole and 3-indoly-
lacetonitrile inhibit spore maturation in paenibacillus alvei. BMC
Microbiol 11:119
148. Catalin C, Field CM, Silvia PF, Keyser UF, Summers DK
(2012) Indole prevents Escherichia coli cell division by modu-
lating membrane potential. Biochim Biophys Acta Biomembr
1818:1590–1594
149. Yee DC, Maynard JA, Wood TK (1998) Rhizoremedia-
tion of trichloroethylene by a recombinant, root-colonizing
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
pseudomonas fluorescens strain expressing toluene ortho-
monooxygenase constitutively. Appl Environ Microbiol
64:112–118
150. Han TH, Cho MH, Lee J (2014) Indole oxidation enhances elec-
tricity production in an E. coli-catalyzed microbial fuel cell.
Biotechnol Bioprocess E 19:126–131
151. Lee JH, Kim YG, Kim CJ, Lee JC, Cho MH, Lee J (2012) Indole-
3-acetaldehyde from Rhodococcus sp. Bfi 332 inhibits Escheri-
chia coli o157:h7 biofilm formation. Appl Microbiol Biotechnol
96:1071–1078
152. Lee JH, Cho HS, Kim Y, Kim JA, Banskota S, Cho MH, Lee
J (2013) Indole and 7-benzyloxyindole attenuate the viru-
lence of Staphylococcus aureus. Appl Microbiol Biotechnol
97:4543–4552
153. Lee JH, Kim YG, Baek KH, Cho MH, Lee J (2015) The mul-
tifaceted roles of the interspecies signalling molecule indole in
Agrobacterium tumefaciens. Environ Microbiol 17:1234–1244
154. Chu W, Zere TR, Weber MM, Wood TK, Whiteley M, Hidalgo-
Romano B Jr, Valenzuela E, Mclean RJ (2012) Indole produc-
tion promotes Escherichia coli mixed-culture growth with
Pseudomonas aeruginosa by inhibiting quorum signaling. Appl
Environ Microbiol 78:411–419
155. Lee JH, Kim YG, Kim M, Kim E, Choi H, Kim Y, Lee J (2017)
Indole-associated predator–prey interactions between the nema-
tode Caenorhabditis elegans and bacteria. Environ Microbiol
19:1776–1790
156. Tomberlin JK, Crippen TL, Wu G, Griffin AS, Wood TK, Kil-
ner RM (2017) Indole: an evolutionarily conserved influencer of
behavior across kingdoms. BioEssays 39(1600203):1–12
157. Costerton JW, Cheng KJ, Geesey GG, Ladd TI, Nickel JC, Das-
gupta M, Marrie TJ (1987) Bacterial biofilms in nature and dis-
ease. Annu Rev Microbiol 41:435–464
158. Van AH, Van DP, Coenye T (2014) Molecular mechanisms of
antimicrobial tolerance and resistance in bacterial and fungal
biofilms. Trends Microbiol 22:326–333
159. Lebeaux D, Ghigo JM, Beloin C (2014) Biofilm-related infec-
tions: bridging the gap between clinical management and fun-
damental aspects of recalcitrance toward antibiotics. Microbiol
Mol Biol Rev 78:510–543
160. Parsek MR, Greenberg EP (2005) Sociomicrobiology: the con-
nections between quorum sensing and biofilms. Trends Microbiol
13:27–33
161. Deng YY, Wu JE, Tao F, Zhang LH (2011) Listening to a new
language: Dsf-based quorum sensing in gram-negative bacteria.
Chem Rev 111:160–173
162. Lebeer S, Claes IJ, Verhoeven TL, Shen C, Lambrichts I, Ceup-
pens JL, Vanderleyden J, De Keersmaecker SC (2008) Impact
of luxs and suppressor mutations on the gastrointestinal tran-
sit of Lactobacillus rhamnosus gg. Appl Environ Microbiol
74:4711–4718
163. Sun Z, He X, Brancaccio VF, Yuan J, Riedel CU (2014) Bifi-
dobacteria exhibit luxs-dependent autoinducer 2 activity and
biofilm formation. PLoS One 9:e88260
164. Li H, Li X, Wang Z, Fu Y, Ai Q, Dong Y, Yu J (2015) Autoin-
ducer-2 regulates pseudomonas aeruginosa pao1 biofilm forma-
tion and virulence production in a dose-dependent manner. BMC
Microbiol 15:1–8
165. Anderson JK, Huang JY, Wreden C, Sweeney EG, Goers J, Rem-
ington SJ, Guillemin K (2015) Chemorepulsion from the quorum
signal autoinducer-2 promotes Helicobacter pylori biofilm dis-
persal. mBio 6:e00379
166. Xue T, Ni J, Shang F, Chen X, Zhang M (2015) Autoinducer-2
increases biofilm formation via an ica- and bhp-dependent
manner in staphylococcus epidermidis rp62a. Microbes Infect
17:345–352
167. Laganenka L, Colin R, Sourjik V (2016) Corrigendum: chemot-
axis towards autoinducer 2 mediates autoaggregation in Escheri-
chia coli. Nat Commun 7:13979
168. Papenfort K, Silpe JE, Schramma KR, Cong JP, Seyedsayamdost
MR, Bassler BL (2017) A vibrio cholerae autoinducer-receptor
pair that controls biofilm formation. Nat Chem Biol 13:551–557
169. Liu L, Wu R, Zhang J, Shang N, Li P (2017) D-ribose interferes
with quorum sensing to inhibit biofilm formation of Lactobacil-
lus paraplantarum l-zs9. Front Microbiol 8:1860
170. Ryan RP, Fouhy Y, Garcia BF, Watt SA, Niehaus K, Yang L,
Tolker-Nielsen T, Dow JM (2008) Interspecies signalling via the
Stenotrophomonas maltophilia diffusible signal factor influences
biofilm formation and polymyxin tolerance in Pseudomonas aer-
uginosa. Mol Microbiol 68:75–86
171. Dean SN, Chung MC, van Hoek ML (2015) Burkholderia dif-
fusible signal factor signals to Francisella novicida to disperse
biofilm and increase siderophore production. Appl Environ
Microbiol 81:7057–7066
172. An SQ, Tang JL (2018) Diffusible signal factor signaling regu-
lates multiple functions in the opportunistic pathogen Steno-
trophomonas maltophilia. BMC Res Notes 11:569–575
173. Krzyzek P, Gosciniak G (2018) A proposed role for diffusible
signal factors in the biofilm formation and morphological trans-
formation of Helicobacter pylori. Turk J Gastroenterol 29:7–13
174. Deng YY, Lim A, Lee J, Chen SH, An SW, Dong YH, Zhang LH
(2014) Diffusible signal factor (dsf) quorum sensing signal and
structurally related molecules enhance the antimicrobial efficacy
of antibiotics against some bacterial pathogens. BMC Microbiol
14:51–59
175. Barel V, Chalupowicz L, Barash I, Sharabani G, Reuven M, Dror
O, Burdman S, Manulis-Sasson S (2015) Virulence and in planta
movement of Xanthomonas hortorum pv. Pelargonii are affected
by the diffusible signal factor (dsf)-dependent quorum sensing
system. Mol Plant Pathol 16:710–723
176. Dow JM (2017) Diffusible signal factor-dependent quorum sens-
ing in pathogenic bacteria and its exploitation for disease control.
J Appl Microbiol 122:2–11
177. Allen RC, Popat R, Diggle SP, Brown SP (2014) Targeting viru-
lence: can we make evolution-proof drugs? Nat Rev Microbiol
12:300–308
178. Dong YH, Wang LH, Xu JL, Zhang HB, Zhang XF, Zhang LH
(2001) Quenching quorum-sensing-dependent bacterial infection
by an n-acyl homoserine lactonase. Nature 411:813–817
179. Shen G, Rajan R, Zhu J, Bell CE, Pei D (2006) Design and
synthesis of substrate and intermediate analogue inhibitors of
s-ribosylhomocysteinase. J Med Chem 49:3003–3011
180. Zhang M, Jiao X, Hu Y, Sun L (2009) Attenuation of edwards-
iella tarda virulence by small peptides that interfere with luxs/
autoinducer type 2 quorum sensing. Appl Environ Microbiol
75:3882–3890
181. Ni N, Li M, Wang J, Wang B (2009) Inhibitors and antagonists
of bacterial quorum sensing. Med Res Rev 29:65–124
182. Brackman G, Defoirdt T, Miyamoto C, Bossier P, Calenbergh
SV, Nelis H, Coenye T (2008) Cinnamaldehyde and cinnamalde-
hyde derivatives reduce virulence in vibriospp. By decreasing the
DNA-binding activity of the quorum sensing response regulator
luxr. BMC Microbiol 8:149
183. Brackman G, Cos P, Maes L, Nelis HJ, Coenye T (2011) Quorum
sensing inhibitors increase the susceptibility of bacterial biofilms
to antibiotics invitro and invivo. Antimicrob Agents Chemother
55:2655–2661
184. Christensen QH, Grove TL, Booker SJ, Greenberg EP (2013) A
high-throughput screen for quorum-sensing inhibitors that tar-
get acyl-homoserine lactone synthases. Proc Natl Acad Sci USA
110:13815–13820
S.Wu et al.
1 3
185. O’Loughlin CT, Miller LC, Siryaporn A, Drescher K, Semmel-
hack MF, Bassler BL (2013) A quorum-sensing inhibitor blocks
pseudomonas aeruginosa virulence and biofilm formation. Proc
Natl Acad Sci USA 110:17981–17986
186. Starkey M, Lepine F, Maura D, Bandyopadhaya A, Lesic B, He
J, Kitao T, Righi V, Milot S, Tzika A (2014) Identification of
anti-virulence compounds that disrupt quorum-sensing regulated
acute and persistent pathogenicity. PLoS Pathog 10:e1004321
187. Ouyang J, Sun F, Feng W, Sun Y, Qiu X, Xiong L, Liu Y, Chen
Y (2016) Quercetin is an effective inhibitor of quorum sensing,
biofilm formation and virulence factors in Pseudomonas aerugi-
nosa. J Appl Microbiol 120:966–974
188. Zerfaß C, Chen J, Soyer OS (2018) Engineering microbial com-
munities using thermodynamic principles and electrical inter-
faces. Curr Opin Biotechnol 50:121–127
189. McCardell RD, Huang S, Green LN, Murray RM (2017) Con-
trol of bacterial population density with population feedback and
molecular sequestration. https ://doi.org/10.1101/22504 5
190. Saeidi N, Wong CK, Lo TM, Nguyen HX, Ling H, Leong SS,
Poh CL, Chang MW (2011) Engineering microbes to sense and
eradicate Pseudomonas aeruginosa, a human pathogen. Mol Syst
Biol 7(521):1–11
191. Gupta S, Bram EE, Weiss R (2013) Genetically programmable
pathogen sense and destroy. ACS Synth Biol 2:715–723
192. Hwang IY, Tan MH, Koh E, Ho CL, Poh CL, Chang MW (2014)
Reprogramming microbes to be pathogen-seeking killers. ACS
Synth Biol 3:228–237
193. Hwang IY, Koh E, Wong A, March JC, Bentley WE, Lee YS,
Chang MW (2017) Engineered probiotic Escherichia coli can
eliminate and prevent Pseudomonas aeruginosa gut infection in
animal models. Nat Commun 8:15028
194. Zhao L, Zhang F, Ding X, Wu G, Lam YY, Wang X, Fu H,
Xue X, Lu C, Ma J (2018) Gut bacteria selectively promoted by
dietary fibers alleviate type 2 diabetes. Science 359:1151–1156
195. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bar-
tolomaeus H, Haase S, Mähler A, Balogh A, Markó L (2017)
Salt-responsive gut commensal modulates th17 axis and disease.
Nature 551:585–589
196. Crotty MP, Jackson PJ (2017) Terminal room disinfection: how
much betr can it get? Lancet 389:765–766
197. Routy B, Le CE, Derosa L, Cpm D, Alou MT, DaillãRe R,
Fluckiger A, Messaoudene M, Rauber C, Roberti MP (2018) Gut
microbiome influences efficacy of pd-1-based immunotherapy
against epithelial tumors. Science 359:91–97
198. Zou J, Chassaing B, Singh V, Pellizzon M, Ricci M, Fythe MD,
Kumar MV, Gewirtz AT (2018) Fiber-mediated nourishment of
gut microbiota protects against diet-induced obesity by restoring
il-22-mediated colonic health. Cell Host Microbe 23:41–53
199. Sun Z, Grimm V, Riedel CU (2015) Ai-2 to the rescue against
antibiotic-induced intestinal dysbiosis? Trends Microbiol
23:327–328
200. Kamada N, Kim YG, Sham HP, Vallance BA, Puente JL, Mar-
tens EC, Núñez G (2012) Regulated virulence controls the abil-
ity of a pathogen to compete with the gut microbiota. Science
336:1325–1329
201. Dandekar AA, Greenberg EP (2012) Bacterial quorum sensing
and metabolic incentives to cooperate. Science 338:264–266
202. Bongaerts GP, Severijnen RS (2016) A reassessment of the
propatria study and its implications for probiotic therapy. Nat
Biotechnol 34:55–63
203. Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M,
Alam MA, Benezra A, Destefano J, Meier MF, Muegge BD
(2014) Persistent gut microbiota immaturity in malnourished
bangladeshi children. Nature 510:417–421
204. Hsiao A, Ahmed AM, Subramanian S, Griffin NW, Drewry LL
Jr, Petri WA, Haque R, Ahmed T, Gordon JI (2014) Members
of the human gut microbiota involved in recovery from vibrio
cholerae infection. Nature 515:423–426
205. Daniel R, Rubens JR, Sarpeshkar R, Lu TK (2013) Synthetic
analog computation in living cells. Nature 497:619–623
206. Bivar Xavier K (2018) Bacterial interspecies quorum sensing
in the mammalian gut microbiota. C R Biol 341:297–299
207. Gilmore MS, Lebreton F, Van SW (2013) Genomic transi-
tion of enterococci from gut commensals to leading causes of
multidrug-resistant hospital infection in the antibiotic era. Curr
Opin Microbiol 16:10–16
208. Borrero J, Chen Y, Dunny GM, Kaznessis YN (2015) Modified
lactic acid bacteria detect and inhibit multiresistant entero-
cocci. ACS Synth Biol 4:299–306
209. Arias CA, Murray BE (2012) The rise of the enterococcus:
beyond vancomycin resistance. Nat Rev Microbiol 10:266–278
210. Coyte KZ, Rakoff-Nahoum S (2019) Understanding competi-
tion and cooperation within the mammalian gut microbiome.
Curr Biol 29:538–544
211. Li Q, Ren Y, Fu X (2019) Inter-kingdom signaling between gut
microbiota and their host. Cell Mol Life Sci 76:2383–2389
212. Mirzaei MK, Maurice CF (2017) Menage a trois in the human
gut: interactions between host, bacteria and phages. Nat Rev
Microbiol 15:397–408
213. Bondydenomy J, Pawluk A, Maxwell KL, Davidson AR (2013)
Bacteriophage genes that inactivate the crispr/cas bacterial
immune system. Nature 493:429–432
214. Patterson AG, Yevstigneyeva MS, Fineran PC (2017) Regula-
tion of crispr-cas adaptive immune systems. Curr Opin Micro-
biol 37:1–7
215. Patterson A, Jackson S, Taylor C, Evans G, Salmond GC, Przy-
bilski R, Staals RJ, Fineran P (2016) Quorum sensing controls
adaptive immunity through the regulation of multiple crispr-
cas systems. Mol Cell 64:1102–1108
216. Brophy JA, Voigt CA (2014) Principles of genetic circuit
design. Nat Methods 11:508–520
217. Buescher JM (2012) Global network reorganization during
dynamic adaptations of bacillus subtilis metabolism. Science
335:1099–1103
218. Woolston BM, Edgar S, Stephanopoulos G (2013) Metabolic
engineering: past and future. Annu Rev Chem Biomol Eng
4:259–288
219. Orth JD, Thiele I, Palsson BØ (2010) What is flux balance
analysis? Nat Biotechnol 28:245–248
220. Meadows AL, Karnik R, Lam H, Forestell S, Snedecor
B (2010) Application of dynamic flux balance analysis
to an industrial escherichia coli fermentation. Metab Eng
12:150–160
221. Hill A (1913) The combinations of haemoglobin with oxygen
and with carbon monoxide. I. Biochem J 7:577–586
222. Schmidt SK, Simkins S, Alexander M (1985) Models for the
kinetics of biodegradation of organic compounds not support-
ing growth. Appl Environ Microbiol 50:323–331
223. English BP, Min W, van Oijen AM, Lee KT, Luo G, Sun H,
Cherayil BJ, Kou SC, Xie XS (2006) Ever-fluctuating single
enzyme molecules: Michaelis-menten equation revisited. Nat
Chem Biol 2:87–94
224. Basu Subhayu, Gerchman Yoram, Collins Cynthia H, Arnold
FH, Weiss R (2005) A synthetic multicellular system for pro-
grammed pattern formation. Nature 434:1130–1134
225. Song H, Stephen P, Meagan G, You L (2009) Spatiotemporal
modulation of biodiversity in a synthetic chemical-mediated
ecosystem. Nat Chem Biol 5:929–935
226. Kong W, Meldgin DR, Collins JJ, Lu T (2018) Designing
microbial consortia with defined social interactions. Nat Chem
Biol 14:821–829
Quorum sensing forpopulation-level control ofbacteria andpotential therapeutic…
1 3
227. Mads K, Elston TC, Blake WJ, Collins JJ (2005) Stochasticity
in gene expression: from theories to phenotypes. Nat Rev Genet
6:451–464
228. Tian T, Burrage K (2006) Stochastic models for regulatory net-
works of the genetic toggle switch. Proc Natl Acad Sci USA
103:8372–8377
229. Prindle A, Liu J, Asally M, Ly S, Garciaojalvo J, Süel GM (2015)
Ion channels enable electrical communication within bacterial
communities. Nature 527:59–63
230. Popkin G (2017) Bacteria use brainlike bursts of electricity to
communicate. Quanta Magazine, New York
231. Liu J, Zhou J, Wang L, Ma Z, Zhao G, Ge Z, Zhu H, Qiao J
(2017) Improving nitrogen source utilization from defatted soy-
bean meal for nisin production by enhancing proteolytic function
of Lactococcus lactis f44. Sci Rep 7:6189
232. Wu H, Song S, Tian K, Zhou D, Wang B, Liu J, Zhu H, Qiao J
(2018) A novel small rna s042 increases acid tolerance in Lacto-
coccus lactis f44. Biochem Biophys Res Commun 500:544–549
233. Song AA, In LLA, Lim SHE, Rahim RA (2017) A review on
Lactococcus lactis: from food to factory. Microb Cell Fact 16:55
234. Guo CJ, Chang FY, Wyche TP, Backus KM, Acker TM, Funa-
bashi M, Mao T, Donia MS, Nayfach S, Pollard KS (2017) Dis-
covery of reactive microbiota-derived metabolites that inhibit
host proteases. Cell 168:517–526
235. Foster KR, Schluter J, Coyte KZ, Rakoff-Nahoum S (2017) The
evolution of the host microbiome as an ecosystem on a leash.
Nature 548:43–51
236. Fernandez L, Rodriguez A, Garcia P (2018) Phage or foe: an
insight into the impact of viral predation on microbial communi-
ties. ISME J 12:1171–1179
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... This has been noted in the interaction of Escherichia coli and Vibrio harveyi. The AI-2 produced by E. coli can induce the QS of V. harveyi to produce bioluminescence and the AI-2 of the V. harveyi can be sensed by E. coli to induce its Lsr system [3,20]. ...
... Quorum sensing is a molecular approach of signaling that mediates cellto-cell communication [2].Bacteria havea well-organized social life.QS network provides bacteria with a method tocollect information from its surrounding and make appropriate decisions using the stimuli from the environment [2]. Using bacterial quorum sensing, both intra-species and inter-species gene regulation is possible and the bacteria can control a variety of traits and several physiological and biochemical functions [3]. The QS helps in regulation of a range of social behavior such as those which promote the fitness of other bacteria or have a positive impact in presence of other competing bacteria by secreting substances that can alter the environment or by secreting toxins that can negatively affect other bacterial strains or species, and horizontal gene transfer [1]. ...
... Quorum signaling helps bacteria in detecting the population size of bacteria by sensing concentration of signal molecules in their environment and thereby controlling investment of energy in processes regulating costly phenotypic traits which is possible only in high cell-density [10]. The synthesis and the release of QS signals leads to the regulation of the production of diverse kinds of extracellular factors from cells which controls behaviors like swarming and motility,virulence, conjugation, competence,scavenging for nutrients, bacteriocin production, pathogenesis, synthesis of antibiotics, suppression of host immune system, formation of biofilms [3,10].These behaviors can only be achieved by cooperation and coordination of the all members of bacterial community [10]. At lower cell-density, QS signals are produced by cell at basal rate. ...
Chapter
Biotechnology is one of the emerging fields that can add new and better application in a wide range of sectors like health care, service sector, agriculture, and processing industry to name some. This book will provide an excellent opportunity to focus on recent developments in the frontier areas of Biotechnology and establish new collaborations in these areas. The book will highlight multidisciplinary perspectives to interested biotechnologists, microbiologists, pharmaceutical experts, bioprocess engineers, agronomists, medical professionals, sustainability researchers and academicians. This technical publication will provide a platform for potential knowledge exhibition on recent trends, theories and practices in the field of Biotechnology.
... The quorum-sensing system controls key mechanisms such as virulence, gene expression, pathogenicity, and antibiotic resistance [22]. This system plays a crucial role in bacterial pathogenicity [23] by regulating the activity of efflux pumps, thereby promoting resistance and tolerance to various antibiotics and disinfectants [24]. In pathogenic bacteria, the coordinated "behaviour and actions" are essential for a successful infection, which is achieved by the QS communication between them [25]. ...
... In pathogenic bacteria, the coordinated "behaviour and actions" are essential for a successful infection, which is achieved by the QS communication between them [25]. However, impaired quorumsensing communication leads to decreased bacterial pathogenicity and the blockade of infection progression [5,6,23,26,27]. Anti-quorum-sensing agents aim not to kill bacteria, but rather to reduce their bacterial virulence [28]. ...
Article
Full-text available
The formation of microbial biofilm is a self-organizing process among bacterial cells, regulated by quorum-sensing (QS) mechanisms, contributing to development of infections. These processes, either separately or in combination, significantly contribute to bacterial resistance to antibiotics and disinfectants. A novel approach to addressing the challenge of treating infections due to antibacterial resistance involves the use of plant metabolites. In recent years, there has been increasing recognition of different phytochemicals as potential modulators. In our study, we evaluated the synergistic effect of chloroform and methanol extracts from Inula species against key virulence factors, including biofilm formation, violacein production, and swarming motility. Each of the 11 examined plant extracts demonstrated the ability to reduce biofilms and pigment synthesis in C. violaceum. Two of the extracts from I. britannica exhibited significant anti-biofilm and anti-quorum-sensing effects with over 80% inhibition. Their inhibitory effect on violacein synthesis indicates their potential as anti-QS agents, likely attributed to their high concentration of terpenoids (triterpenoids, sesquiterpene lactones, and diterpenoids). Scanning electron microscopy revealed a notable reduction in biofilm biomass, along with changes in biofilm architecture and cell morphology. Additionally, fluorescence microscopy revealed the presence of metabolically inactive cells, indicating the potent activity of the extracts during treatment. These new findings underscore the effectiveness of the plant extracts from the genus Inula as potential anti-virulent agents against C. violaceum. They also propose a promising strategy for preventing or treating its biofilm formation.
... There are many significant associations between human gut microbiota and gastrointestinal diseases [1][2][3]. The dynamic homeostasis of the human body is dependent on the complex interactions among different gut microbes [4][5][6][7]. While most human microbiome researches focus on associating health outcomes to microbial taxonomic indicators, there is a growing realization that it is not the microbes themselves are responsible for specific health effects, but rather their metabolites [8][9][10][11]. ...
Article
Full-text available
How to resolve the metabolic dark matter of microorganisms has long been a challenging problem in discovering active molecules. Diverse omics tools have been developed to guide the discovery and characterization of various microbial metabolites, which make it gradually possible to predict the overall metabolites for individual strains. The combinations of multi-omic analysis tools effectively compensates for the shortcomings of current studies that focus only on single omics or a broad class of metabolites. In this review, we systematically update, categorize and sort out different analysis tools for microbial metabolites prediction in the last five years to appeal for the multi-omic combination on the understanding of the metabolic nature of microbes. First, we provide the general survey on different updated prediction databases, webservers, or software that based on genomics, transcriptomics, proteomics, and metabolomics, respectively. Then, we discuss the essentiality on the integration of multi-omics data to predict metabolites of different microbial strains and communities, as well as stressing the combination of other techniques, such as systems biology methods and data-driven algorithms. Finally, we identify key challenges and trends in developing multi-omic analysis tools for more comprehensive prediction on diverse microbial metabolites that contribute to human health and disease treatment.
... Microorganisms 2024, 12, 985 ...
Article
Full-text available
Pseudomonas aeruginosa is an opportunistic pathogen that usually causes chronic infections and even death in patients. The treatment of P. aeruginosa infection has become more challenging due to the prevalence of antibiotic resistance and the slow pace of new antibiotic development. Therefore, it is essential to explore non-antibiotic methods. A new strategy involves screening for drugs that target the quorum-sensing (QS) system. The QS system regulates the infection and drug resistance in P. aeruginosa. In this study, veratryl alcohol (VA) was found as an effective QS inhibitor (QSI). It effectively suppressed the expression of QS-related genes and the subsequent production of virulence factors under the control of QS including elastase, protease, pyocyanin and rhamnolipid at sub-inhibitory concentrations. In addition, motility activity and biofilm formation, which were correlated with the infection of P. aeruginosa, were also suppressed by VA. In vivo experiments demonstrated that VA could weaken the pathogenicity of P. aeruginosa in Chinese cabbage, Drosophila melanogaster, and Caenorhabditis elegans infection models. Molecular docking, combined with QS quintuple mutant infection analysis, identified that the mechanism of VA could target the LasR protein of the las system mainly. Moreover, VA increased the susceptibility of P. aeruginosa to conventional antibiotics of tobramycin, kanamycin and gentamicin. The results firstly demonstrate that VA is a promising QSI to treat infections caused by P. aeruginosa.
... This circuit is based on a production-lysis oscillation in which the lysis gene E is activated using acyl homoserine lactone (AHL) quorum-sensing signaling, resulting in controlled lysis of some bacteria. [43][44][45] The SLC-Ag system was designed to colonize tumors and undergo intratumoral quorum lysis, leading to the simultaneous local delivery of Ag, DOX, hsulf-1, and immunostimulatory lysed bacterial adjuvants to stimulate antitumor immunity and promote tumor regression ( Figure 1A). ...
Article
Full-text available
Background New treatment modalities for hepatocellular carcinoma (HCC) are desperately critically needed, given the lack of specificity, severe side effects, and drug resistance with single chemotherapy. Engineered bacteria can target and accumulate in tumor tissues, induce an immune response, and act as drug delivery vehicles. However, conventional bacterial therapy has limitations, such as drug loading capacity and difficult cargo release, resulting in inadequate therapeutic outcomes. Synthetic biotechnology can enhance the precision and efficacy of bacteria-based delivery systems. This enables the selective release of therapeutic payloads in vivo. Methods In this study, we constructed a non-pathogenic Escherichia coli (E. coli) with a synchronized lysis circuit as both a drug/gene delivery vehicle and an in-situ (hepatitis B surface antigen) Ag (ASEc) producer. Polyethylene glycol (CHO-PEG2000-CHO)-poly(ethyleneimine) (PEI25k)-citraconic anhydride (CA)-doxorubicin (DOX) nanoparticles loaded with plasmid encoded human sulfatase 1 (hsulf-1) enzyme (PNPs) were anchored on the surface of ASEc (ASEc@PNPs). The composites were synthesized and characterized. The in vitro and in vivo anti-tumor effect of ASEc@PNPs was tested in HepG2 cell lines and a mouse subcutaneous tumor model. Results The results demonstrated that upon intravenous injection into tumor-bearing mice, ASEc can actively target and colonise tumor sites. The lytic genes to achieve blast and concentrated release of Ag significantly increased cytokine secretion and the intratumoral infiltration of CD4/CD8⁺T cells, initiated a specific immune response. Simultaneously, the PNPs system releases hsulf-1 and DOX into the tumor cell resulting in rapid tumor regression and metastasis prevention. Conclusion The novel drug delivery system significantly suppressed HCC in vivo with reduced side effects, indicating a potential strategy for clinical HCC therapy.
... As previously described, Gram-positive bacteria QS is mediated by autoinducer peptides (AIPs), which are typically released extracellularly and are, therefore, present in the surrounding medium. Gram-positive bacteria can detect and respond to AIPs to regulate their metabolic activities [15]. This feedback loop involving AIPs may enhance QS activity, leading to gene regulation that potentially triggers higher QQ activity, resulting in the detection of AHL molecules and the increase in degradation by B. velezensis D-18. ...
Article
Full-text available
Amid growing concerns about antibiotic resistance, innovative strategies are imperative in addressing bacterial infections in aquaculture. Quorum quenching (QQ), the enzymatic inhibition of quorum sensing (QS), has emerged as a promising solution. This study delves into the QQ capabilities of the probiotic strain Bacillus velezensis D-18 and its products, particularly in Vibrio anguillarum 507 communication and biofilm formation. Chromobacterium violaceum MK was used as a biomarker in this study, and the results confirmed that B. velezensis D-18 effectively inhibits QS. Further exploration into the QQ mechanism revealed the presence of lactonase activity by B. velezensis D-18 that degraded both long-and short-chain acyl homoserine lactones (AHLs). PCR analysis demonstrated the presence of a homologous lactonase-producing gene, ytnP, in the genome of B. velezensis D-18. The study evaluated the impact of B. velezensis D-18 on V. anguillarum 507 growth and biofilm formation. The probiotic not only controls the biofilm formation of V. anguillarum but also significantly restrains pathogen growth. Therefore, B. velezensis D-18 demonstrates substantial potential for preventing V. anguillarum diseases in aquaculture through its QQ capacity. The ability to disrupt bacterial communication and control biofilm formation positions B. velezensis D-18 as a promising eco-friendly alternative to conventional antibiotics in managing bacterial diseases in aquaculture.
... Pathogenic bacteria, including E. coli, use QS as a regulator for various biological processes such as biofilm formation, the production of secondary metabolites, and interactions with hosts and other microorganisms. In particular, QS plays a crucial role in the generation of virulence factors and the development of antibiotic resistance [13]. The AI-2, one of the QS signaling molecules, is produced by both Gram-positive and Gram-negative bacteria and is involved in the regulation of multiple bacterial processes [14]. ...
Article
Full-text available
Porcine extraintestinal pathogenic Escherichia coli (ExPEC) is a pathogenic bacterium that causes huge economic losses to the pig farming industry and considerably threatens human health. The quorum sensing (QS) system plays a crucial role in the survival and pathogenesis of pathogenic bacteria. Hence, it is a viable approach to prevent ExPEC infection by compromising the QS system, particularly the LuxS/AI-2 system. In this study, we investigated the effects of baicalin on the LuxS/AI-2 system of ExPEC. Baicalin at concentrations of 25, 50, and 100 μg/mL significantly diminished the survival ability of ExPEC in hostile environments and could inhibit the biofilm formation and autoagglutination ability in ExPEC. Moreover, baicalin dose-dependently decreased the production of AI-2 and down-regulated the expression level of luxS in PCN033. These results suggest that baicalin can weaken the virulence of PCN033 by inhibiting the LuxS/AI-2 system. After the gene luxS was deleted, AI-2 production in PCN033 was almost completely eliminated, similar to the effect of baicalin on the production of AI-2 in PCN033. This indicates that baicalin reduced the production of AI-2 by inhibiting the expression level of luxS in ExPEC. In addition, the animal experiment further showed the potential of baicalin as a LuxS/AI-2 system inhibitor to prevent ExPEC infection. This study highlights the potential of baicalin as a natural quorum-sensing inhibitor for therapeutic applications in preventing ExPEC infection by targeting the LuxS/AI-2 system.
Article
Full-text available
Iron is a transition metal used as a cofactor in many biochemical reactions. In bacteria, iron homeostasis involves Fur-mediated de-repression of iron uptake systems, such as the iron-chelating compounds siderophores. In this work, we identified and characterized novel regulatory systems that control siderophores in the environmental opportunistic pathogen Chromobacterium violaceum. Screening of a 10,000-transposon mutant library for siderophore halos identified seven possible regulatory systems involved in siderophore-mediated iron homeostasis in C. violaceum. Further characterization revealed a regulatory cascade that controls siderophores involving the transcription factor VitR acting upstream of the quorum-sensing (QS) system CviIR. Mutation of the regulator VitR led to an increase in siderophore halos, and a decrease in biofilm, violacein, and protease production. We determined that these effects occurred due to VitR-dependent de-repression of vioS. Increased VioS leads to direct inhibition of the CviR regulator by protein-protein interaction. Indeed, insertion mutations in cviR and null mutations of cviI and cviR led to an increase of siderophore halos. RNA-seq of the cviI and cviR mutants revealed that CviR regulates CviI-dependent and CviI-independent regulons. Classical QS-dependent processes (violacein, proteases, and antibiotics) were activated at high cell density by both CviI and CviR. However, genes related to iron homeostasis and many other processes were regulated by CviR but not CviI, suggesting that CviR acts without its canonical CviI autoinducer. Our data revealed a complex regulatory cascade involving QS that controls siderophore-mediated iron homeostasis in C. violaceum. IMPORTANCE The iron-chelating compounds siderophores play a major role in bacterial iron acquisition. Here, we employed a genetic screen to identify novel siderophore regulatory systems in Chromobacterium violaceum, an opportunistic human pathogen. Many mutants with increased siderophore halos had transposon insertions in genes encoding transcription factors, including a novel regulator called VitR, and CviR, the regulator of the quorum-sensing (QS) system CviIR. We found that VitR is upstream in the pathway and acts as a dedicated repressor of vioS, which encodes a direct CviR-inhibitory protein. Indeed, all QS-related phenotypes of a vitR mutant were rescued in a vitRvioS mutant. At high cell density, CviIR activated classical QS-dependent processes (violacein, proteases, and antibiotics production). However, genes related to iron homeostasis and type-III and type-VI secretion systems were regulated by CviR in a CviI- or cell density-independent manner. Our data unveil a complex regulatory cascade integrating QS and siderophores in C. violaceum.
Article
Full-text available
Microbiomes are foundational components of the environment that provide essential services relating to food security, carbon sequestration, human health, and the overall well-being of ecosystems. Microbiota exert their effects primarily through complex interactions at interfaces with their plant, animal, and human hosts, as well as within the soil environment. This review aims to explore the ecological, evolutionary, and molecular processes governing the establishment and function of microbiome-host relationships, specifically at interfaces critical to One Health – a transdisciplinary framework that recognises that the health outcomes of people, animals, plants and the environment are tightly interconnected. Within the context of One Health, the core principles underpinning microbiome assembly will be discussed in detail, including biofilm formation, microbial recruitment strategies, mechanisms of microbial attachment, community succession and the effect these processes have on host function and health. Finally, this review will catalogue recent advances in microbiology and microbial ecology methods that can be used to profile microbial interfaces, with particular attention to multi-omic, advanced imaging, and modelling approaches. These technologies are essential for delineating the general and specific principles governing microbiome assembly and functions, mapping microbial interconnectivity across varying spatial and temporal scales, and for the establishment of predictive frameworks that will guide the development of targeted microbiome-interventions to deliver One Health outcomes.
Preprint
Full-text available
Genetic engineering technology has become sophisticated enough to allow precise manipulation of bacterial genetic material. Engineering efforts with these technologies have created modified bacteria for various medical, industrial, and environmental purposes, but organisms designed for specific functions require improvements in stability, longevity, or efficiency of function. Most bacteria live in multispecies communities, whose composition may be closely linked to the effect the community has on the environment. Bacterial engineering efforts will benefit from building communities with regulated compositions, which will enable more stable and powerful community functions. We present a design of a synthetic two member bacterial community capable of maintaining its composition at a defined ratio of [ cell type 1]: [ cell type 2]. We have constructed the genetic motif that will act in each cell in the two member community, containing an AHL-based negative feedback loop that activates ccdB toxin, which caps population density with increasing feedback strength. It also contains one of two ccdB sequestration modules, either the ccdA protein antitoxin, or an RNA device which prevents transcription and translation of ccdB mRNA, that rescues capped population density with induction. We compare absorbance and colony counting methods of estimating bacterial population density, finding that absorbance-based methods overestimate viable population density when ccdB toxin is used to control population density. Prior modeling results show that two cell types containing this genetic circuit motif that reciprocally activate the other’s ccdB sequestration device will establish a steady state ratio of cell types. Experimental testing and tuning the full two member community will help us improve our modeling of multi-member bacterial communities, learn more about the strengths and weaknesses of our design for community composition control, and identify general principles of design of compositionally-regulated microbial communities.
Article
Full-text available
Objectives A three-species consortium for one-step fermentation of 2-keto-l-gulonic acid (2-KGA) was constructed to better strengthen the cell–cell communication. And the programmed cell death module based on the LuxI/LuxR quorum-sensing (QS) system was established in Gluconobacter oxydans to reduce the competition that between G. oxydans and Ketogulonicigenium vulgare. Results By constructing and optimizing the core region of the promoter, which directly regulated the expression of lethal ccdB genes in QS system, IR3C achieved the best lethal effect. The consortium of IR3C- K. vulgare–Bacillus megaterium (abbreviated as 3C) achieved the highest 2-KGA titer (68.80 ± 4.18 g/l), and the molar conversion rate was 80.7% within 36 h in 5 l fermenter. Metabolomic analysis on intracellular small molecules of consortia 3C and 1C showed that most amino acids (such as glycine, leucine, methionine and proline) and TCA cycle intermediates (such as succinic acid, fumaric acid and malic acid) were significantly affected. These results further validated that the programmed cell death module based on the LuxI/LuxR QS system in G. oxydans could also faciliate better growth and higher production of consortium 3C for one-step fermentation. Conclusions We successfully constructed a novel three-species consortia for one-step vitamin C fermentation by strengthening the cell–cell communication. This will be very useful for probing the rational design principles of more complex multi-microbial consortia.
Article
Full-text available
The crosstalk between prokaryotic bacteria and eukaryotic gut epithelial cells has opened a new field for research. Quorum sensing system (QS) molecules employed by gut microbiota may play an essential role in host–microbial symbioses of the gut. Recent studies on the gut microbiome will unveil evolved mechanisms of the host to affect bacterial QS and shape bacterial composition. Bacterial autoinducers (AIs) could talk to the host’s gut by eliciting proinflammatory effects and modulating the activities of T lymphocyte, macrophage, dendritic cells, and neutrophils. In addition, the gut mucosa could interfere with bacterial AIs by degrading them or secreting AI mimics. Moreover, bacterial AIs and gut hormones epinephrine and noradrenaline may be interchangeable in the crosstalk between the microbiota and human gut. Therefore, inter-kingdom signaling between gut microbiota and host may provide a novel target in the management of gut microbiota-related conditions or diseases in the future.
Article
Full-text available
Coarse-grained rules are widely used in chemistry, physics and engineering. In biology, however, such rules are less common and under-appreciated. This gap can be attributed to the difficulty in establishing general rules to encompass the immense diversity and complexity of biological systems. Furthermore, even when a rule is established, it is often challenging to map it to mechanistic details and to quantify these details. Here we report a framework that addresses these challenges for mutualistic systems. We first deduce a general rule that predicts the various outcomes of mutualistic systems, including coexistence and productivity. We further develop a standardized machine-learning-based calibration procedure to use the rule without the need to fully elucidate or characterize their mechanistic underpinnings. Our approach consistently provides explanatory and predictive power with various simulated and experimental mutualistic systems. Our strategy can pave the way for establishing and implementing other simple rules for biological systems.
Article
Full-text available
The ComRS system can function as a quorum sensing trigger for genetic competence in S. mutans . The signal peptide XIP, which is derived from the precursor ComS, enters the cell and interacts with the Rgg-type cytosolic receptor ComR to activate comX , which encodes the alternative sigma factor for the late competence genes. Previous studies have demonstrated intercellular signaling via ComRS, although release of the ComS or XIP peptide to the extracellular medium appears to require lysis of the producing cells. Here we tested the complementary hypothesis that ComRS can drive comX through a purely intracellular mechanism that does not depend on extracellular accumulation or import of ComS or XIP. By combining single-cell, coculture, and microfluidic approaches, we demonstrated that endogenously produced ComS can enable ComRS to activate comX without requiring processing, export, or import. These data provide insight into intracellular mechanisms that generate noise and heterogeneity in S. mutans competence.
Article
Synthetic microbial consortia consisting of microorganisms with different synthetic genetic circuits or divided synthetic metabolic pathway components can exert functions that are beyond the capacities of single microorganisms. However, few consortia of microorganisms with different synthetic genetic circuits have been developed. We designed and constructed a synthetic microbial consortium composed of an enzyme-producing strain and a target chemical-producing strain using Escherichia coli for chemical production with efficient saccharification. The enzyme-producing strain harbored a synthetic genetic circuit to produce beta-glucosidase, which converts cellobiose to glucose, destroys itself via the lytic genes, and release the enzyme when the desired cell density is reached. The target chemical-producing strain was programmed by a synthetic genetic circuit to express enzymes in the synthetic metabolic pathway for isopropanol production when the enzyme-producing strain grows until release of the enzyme. Our results demonstrate the benefits of synthetic microbial consortia with distributed tasks for effective chemical production from biomass.
Article
Biofilms are surface-associated bacterial communities that play both beneficial and harmful roles in nature, medicine, and industry. Tolerant and persister cells are thought to underlie biofilm-related bacterial recurrence in medical and industrial contexts. Here, we review recent progress aimed at understanding the mechanical features that drive biofilm resilience and the biofilm formation process at single-cell resolution. We discuss findings regarding mechanisms underlying bacterial tolerance and persistence in biofilms and how these phenotypes are linked to antibiotic resistance. New strategies for combatting tolerance and persistence in biofilms and possible methods for biofilm eradication are highlighted to inspire future development.
Article
The mammalian gut harbors a vast community of microorganisms - termed the microbiota - whose composition and dynamics are considered to be critical drivers of host health. These factors depend, in part, upon the manner in which microbes interact with one another. Microbes are known to engage in a myriad of different ways, ranging from unprovoked aggression to actively feeding each other. However, the relative extent to which these different interactions occur between microbes within the gut is unclear. In this minireview we assess our current knowledge of microbe-microbe interactions within the mammalian gut microbiota, and the array of methods used to uncover them. In particular, we highlight the discrepancies between different methodologies: some studies have revealed rich networks of cross-feeding interactions between microbes, whereas others suggest that microbes are more typically locked in conflict and actively cooperate only rarely. We argue that to reconcile these contradictions we must recognize that interactions between members of the microbiota can vary across condition, space, and time - and that only through embracing this dynamism will we be able to comprehensively understand the ecology of our gut communities.
Article
Quorum sensing is a process of bacterial cell-to-cell chemical communication that relies on the production, detection and response to extracellular signalling molecules called autoinducers. Quorum sensing allows groups of bacteria to synchronously alter behaviour in response to changes in the population density and species composition of the vicinal community. Quorum-sensing-mediated communication is now understood to be the norm in the bacterial world. Elegant research has defined quorum-sensing components and their interactions, for the most part, under ideal and highly controlled conditions. Indeed, these seminal studies laid the foundations for the field. In this Review, we highlight new findings concerning how bacteria deploy quorum sensing in realistic scenarios that mimic nature. We focus on how quorums are detected and how quorum sensing controls group behaviours in complex and dynamically changing environments such as multi-species bacterial communities, in the presence of flow, in 3D non-uniform biofilms and in hosts during infection.
Article
From biosynthesis to bioremediation, microbes have been engineered to address a variety of biotechnological applications. A promising direction in these endeavors is harnessing the power of designer microbial consortia that consist of multiple populations with well-defined interactions. Consortia can accomplish tasks that are difficult or potentially impossible to achieve using monocultures. Despite their potential, the rules underlying microbial community maintenance and function (i.e. the task the consortium is engineered to carry out) are not well defined, though rapid progress is being made. This limited understanding is in part due to the greater challenges associated with increased complexity when dealing with multi-population interactions. Here, we review key features and design strategies that emerge from the analysis of both natural and engineered microbial communities. These strategies can provide new insights into natural consortia and expand the toolbox available to engineers working to develop novel synthetic consortia.