You are on page 1of 54

Journal Pre-proof

Biotechnological potential and applications of microbial consortia

Xiujuan Qian, Lin Chen, Yuan Sui, Chong Chen, Wenming


Zhang, Jie Zhou, Weiliang Dong, Min Jiang, Fengxue Xin, Katrin
Ochsenreither

PII: S0734-9750(19)30200-9
DOI: https://doi.org/10.1016/j.biotechadv.2019.107500
Reference: JBA 107500

To appear in: Biotechnology Advances

Received date: 9 May 2019


Revised date: 13 November 2019
Accepted date: 17 December 2019

Please cite this article as: X. Qian, L. Chen, Y. Sui, et al., Biotechnological potential
and applications of microbial consortia, Biotechnology Advances (2019), https://doi.org/
10.1016/j.biotechadv.2019.107500

This is a PDF file of an article that has undergone enhancements after acceptance, such
as the addition of a cover page and metadata, and formatting for readability, but it is
not yet the definitive version of record. This version will undergo additional copyediting,
typesetting and review before it is published in its final form, but we are providing this
version to give early visibility of the article. Please note that, during the production
process, errors may be discovered which could affect the content, and all legal disclaimers
that apply to the journal pertain.

© 2019 Published by Elsevier.


Journal Pre-proof

Biotechnological potential and applications of microbial consortia

Xiujuan Qiana, Lin Chena, Yuan Suib, Chong Chenb, Wenming Zhanga,c, Jie Zhoua,c,

Weiliang Donga,c, Min Jianga,c*, Fengxue Xina,c*, Katrin Ochsenreitherd

a
State Key Laboratory of Materials-Oriented Chemical Engineering,

f
College of Biotechnology and Pharmaceutical Engineering,

oo
Nanjing Tech University, Nanjing, China

b pr
2011 College, Nanjing Tech University, Nanjing, China
e-
c
Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM),
Pr

Nanjing Tech University, Nanjing, China

d
Institute of Process Engineering in Life Sciences, Section II: Technical Biology,
al

Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany


u rn
Jo

*Corresponding author: Fengxue Xin, E-mail: xinfengxue@njtech.edu.cn.

Min Jiang, E-mail: bioengine@njtech.edu.cn.

Mailing address: State Key Laboratory of Materials-Oriented Chemical Engineering,

College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University,

Puzhu South Road 30#, Nanjing 211800, P. R. China.

Tel/Fax: 0086-25-58139927
Journal Pre-proof

Abstract

Recent advances in microbial consortia present a valuable approach for

expanding the scope of metabolic engineering. Systems biology enable thorough

understanding of diverse physiological processes of cells and their interactions, which

in turn offers insights into the optimal design of synthetic microbial consortia. Yet, the

study of synthetic microbial consortia is still in early infancy, facing many unknowns

f
and challenges in intercellular communication and construction of stable and

oo
controllable microbial consortia systems. In this review, we comprehensively

pr
discussed the recent application of defined microbial consortia in the fields of human
e-
health monitoring and medicine exploitation, valuable compounds synthesis,
Pr

consolidated bioprocessing of lignocellulosic materials and environmental

bioremediation. Moreover, the outstanding challenges and future directions to


al

advance the development of high-efficient, stable and controllable synthetic microbial


rn

consortia were highlighted.


u
Jo

Key words: Microbial consortia application, Human health supervision, Valuable

compounds synthesis, Consolidated bioprocessing, Environment bio-remediation,

Microbial consortia construction challenges

Introduction

Traditional metabolic engineering using axenic cultures has provided great


Journal Pre-proof

opportunities for the production of bulk chemicals and valuable products (Hall and

Howe, 2012). However, the inherent exclusiveness to foreign genes, the lack of novel

enzymes, the existence of silent pathways and requirements for rigid cultivation

conditions always affect bio-product titer and productivity (Bhatia et al. , 2018, Shong

et al. , 2012). As a result, the number of successful examples of commercialized

microbial processes to produce chemicals and fuels cannot fulfill expectations

f
(Gustavsson and Lee, 2016), leaving the question of whether the traditional metabolic

oo
strategy, i.e. construction of “super cell factories” is sufficient for sustainable

biorefineries. pr
e-
Nevertheless, traditional food fermentation processes, such as cheese and soy
Pr

sauce production are typically carried out by using mixed cultures consisting of

multiple strains or species (Hanemaaijer et al. , 2015). Moreover, more than 99% of
al

microorganisms in the environment cannot be cultured successfully by using


rn

traditional cultivation technologies (Kaeberlein et al. , 2002). The powerful features of


u

natural microbial consortia inspired researchers to extend synthetic biology from


Jo

traditional programming of single cells to mixed culture systems construction. As

expected, more and more studies related to the exploration of microbial consortia

potential, distributing synthetic pathways to different microorganisms within consortia,

cell-cell communication mechanism and development of statistical models for

microbial consortia systems have been reported recently (Fig. 1) (An et al. , 2019,

Roell et al. , 2019, Song et al. , 2014, Wang et al. , 2019b, Zhang and Wang, 2016).

The construction of artificial microbial consortia has opened a new horizon in


Journal Pre-proof

synthetic biology in terms of system complexity and functionality. First, these

complex consortia create a novel microenvironment for strains, potentially resulting

in the activation of silent metabolic pathways, which are not expressed under “normal”

cultivation conditions, leading to discovery of novel chemicals, endowing microbial

consortia a promising prospect especially in novel drugs development (Netzker et al. ,

2018). In addition, microbiome behavior in the body is an important factor influence

f
human health, as humans are co-evolving with trillions of microbes that inhabit in our

oo
bodies, which created complex, body-habitat-specific and adaptive ecosystems to

pr
adapt to the relentlessly changing host physiology (Turnbaugh et al. , 2007). Second,
e-
microbial consortia employ the approach of “division of labor”, allowing a burden
Pr

distribution across the population and permitting improved efficiency and more

complex behavior than monocultures (Hays et al. , 2015, Zhou et al. , 2015). It might
al

be easier to optimize the modularized pathway by changing the ratio of constituent


rn

strains, while a series of molecular engineering with regard to promoters, ribosome


u

binding sites, terminators, and vectors, etc. are needed when using monocultures
Jo

(Roell et al. , 2019). Third, such microbial communities consist of multiple functional

microorganisms, which are more robust towards environmental challenges and

possess expanded metabolic capabilities relative to monocultures (McCarty and

Ledesma-Amaro, 2018).

In this review, recent applications using artificial microbial consortia in the area

of human health supervision, valuable compounds synthesis, consolidated

bioprocessing for lignocellulose biorefinery and environment bioremediation will be


Journal Pre-proof

comprehensively summarized and discussed, with emphasis on the strategies in

synthetic microbial consortia construction, aiming to exploit the promising potential

of microbial consortia in more areas. Further, significant challenges and future

directions to advance the development of artificial microbial consortia were

highlighted.

Promising trends and potential applications of microbial consortia for human

f
health supervision

oo
The serendipitous discovery of penicillin using microbial consortia of Penicillium

pr
and Staphylococcus by Alexander Fleming in 1929 was regarded as one of the most
e-
influential scientific breakthroughs in last century (Bennett and Chung, 2001). Since
Pr

then, more and more novel chemicals have been discovered especially with the

development of biotechnology. Table 1 summarized novel chemicals synthesized


al

using microbial consortia reported in the last decade. As seen, most of them exhibit
rn

antimicrobial properties and can only be found in microbial consortia systems. Due to
u

the increasing number of pathogenic antibiotic-resistant strains, the need for research
Jo

and development of new antibiotic substances is more relevant than ever (Tacconelli

et al. , 2018). The unique characteristic of microbial consortia clearly highlights its

potential in antibiotic chemicals synthesis.

The growing number of available microbial genome sequences showed that many

biosynthetic gene clusters, responsible for synthesis of secondary metabolites (SMs)

are presumably silent under standard laboratory conditions (Marmann et al. , 2014),

e.g. the silent glycopeptide cluster in Amycolatopsis (Spohn et al. , 2014). There is
Journal Pre-proof

evidence that the activation of some silent gene clusters requires the physical presence

of other microorganisms (Bertrand et al. , 2014). In order to discover the unrevealed

interaction mechanisms behind microbial consortia, a series of investigations from the

perspectives of physical contact, chemical communication and gene mutation have

been conducted (Fig. 2).

In terms of chemical communication, microbes can produce some signal

f
compounds, such as N-acyl homoserine lactones (AHLs) and small peptides, to

oo
perform as transcriptional regulators and epigenetic modifiers in a process called

pr
“quorum sensing” (QS) (Joint et al. , 2002). AHLs are major class of autoinducer
e-
signal molecules used by gram-negative bacteria for intraspecies QS (Ng and Bassler,
Pr

2009). Once AHL concentration in the environment reaches a threshold level, they

will activate the transcriptional regulator proteins of LuxR family. The LuxR/AHL
al

complex can activate the expression of multiple target genes, including those required
rn

for AHL synthesis (Joint et al. , 2002) (Fig. 2a). Such QS mechanism has been
u

successfully used to engineer spatio-temporally regulated cell-cell communication in


Jo

Escherichia coli (Basu et al. , 2004), to control the population in microbial consortia

by regulating cell growth and death (Balagaddé et al. , 2008, You et al. , 2004), and to

reduce the substrate competition between different species (Wang et al. , 2019a). In

gram-positive bacteria consortia, small peptides, known as autoinducing peptides

(AIPs) are mostly used as QS molecules (Ng and Bassler, 2009, Thoendel and

Horswill, 2010). Different from AHLs, these peptides vary in sequence and structure,

which are actively exported from the cell by dedicated transporters (Michie et al. ,
Journal Pre-proof

2016). AIPs regulated QS systems typically employ a two-component genetic

regulatory mechanism—a membrane-bound AIP-receptor-histidine kinase (HK) and a

DNA-binding response regulator (Fig. 2b). Once the critical concentration of AIP is

reached in extracellular environment, it will be phosphorylated by the receptor-HK

and imported into the cell. The phosphorylated AIP will bind on the target DNA to

regulate its transcription (Michie et al. , 2016). Beyond the chemical communication

f
in the same genus consortia, QS molecules also seem to influence

oo
prokaryote-eukaryote interactions. For example, orsellinic acid derivatives are usually

pr
found in a fungal/bacterial mutualism, implying the functional role of orsellinic acid
e-
in microbial communication (Brakhage and Schroeckh, 2011, Fischer et al. , 2018,
Pr

Schroeckh et al. , 2009). Owing to these discoveries, it becomes easier to construct a

communication system by linking the production of QS molecules with the


al

corresponding receptors and promoters, ultimately, a controllable microbial


rn

consortium can be designed.


u

In addition to freely diffusible QS molecules mentioned in Fig. 2a and 2b, the


Jo

transport of some signal molecules also needs to be assisted by a special structure. For

example, hydrophobic signals such as long-chain AHLs are transported between cells

by membrane vesicles (MVs) (Toyofuku, 2019) (Fig. 2c). In some cases, an intimate

physical contact of the cell members within a consortium is required to elicit the

specific communication. The classical case was the interaction between Aspergillus

nidulans with actinomycetes, where archetypal polyketides could be only

biosynthesized after physical contact of these two partners in the consortium


Journal Pre-proof

(Schroeckh et al. , 2009) (Fig. 2d).

Further gene level analysis indicated that microbial consortia cause a series of

gene expression changes in the manner of gene loss, histone modification, and

horizontal gene transfer (Fig. 2e). For example, co-culture of Streptomyces

clavuligerus with Staphylococcus aureus N315 led to the loss of a large 1.8 Mbp

plasmid, constituting 21% of the genome content, in S. clavuligerus. Consequently, S.

f
clavuligerus acquired the ability to constitutively produce holomycin within this

oo
microbial consortia system (Charusanti et al. , 2012). Speculatively, the loss of gene

pr
segment in S. clavuligerus may reduce the metabolic burden during replication and
e-
gene expression. In return, the silent holomycin synthetic pathway was specifically
Pr

activated, which is a member of the pyrrhotine class of antibiotics and shows great

cytotoxicity levels against S. aureus (Oliva et al. , 2001). Bacterium could also alter
al

fungal gene expression by inducing histone modification through the main histone
rn

acetyltransferase complex Saga/Ada (Nützmann et al. , 2011). In general, histone


u

acetylation is linked to transcriptional activation so that regulate gene expression.


Jo

Moreover, genomic analysis revealed that Rhodococcus 307CO harbors a large DNA

segment derived from Streptomyces within the microbial consortia system, leading to

the production of new isomeric antibiotics of rhodostreptomycin A and B (Kurosawa

et al. , 2008). In fact, horizontal gene transfer between gram-positive and

gram-negative bacteria was commonly found, which could be defined as predation

and lead to the incorporation of prey DNA by predator (Pérez et al. , 2011).

The inter-communication within microbial consortia is a very complex process.


Journal Pre-proof

Currently, the most effective approach to explore the interaction mechanism is

through characterizing small of reduced complexities, to detect the specific metabolic

interactions or introduce reporter strains, then, to build a representative model library

of microbial consortia. Correspondingly, more advanced perceiving and analytical

technologies, such as metagenomics, systems biology imaging mass spectrometry,

microfluidic technique and flow cytometry, cell isolation and printing, and

f
high-throughput cultivation should be developed.

oo
In addition to the antibiotic chemicals discovery, microbial consortia also play

pr
promising roles in clinical research. Microorganisms that colonize the human body,
e-
including mucosal and skin environments are at least as abundant as our somatic cells
Pr

and certainly contain far more genes than our human genome (Gilbert et al. , 2018,

Lloyd-Price et al. , 2016). The body microbiome behavior is an important factor in


al

addition to genetics and environment that influence human health (Turnbaugh et al. ,
rn

2007). For example, the gut microbiome is now emerging as an important player in
u

personalized medicine, as it enables the breakdown of otherwise indigestible


Jo

polysaccharides, and is essential for the development and homeostasis of the immune

system in the gut to resist against pathogenic bacteria (Chen et al. , 2018). An

enhanced understanding of the impact of microbial communities and better

elaboration of the exact mechanism of such communities has also become a priority in

human microbiome study.

Valuable compounds synthesis using microbial consortia through the distribution

of metabolic pathways within different strains


Journal Pre-proof

Synthetic biology and metabolic engineering have made great strides in

constructing and optimizing metabolic pathways in model microbes, such as E. coli

and Saccharomyces cerevisiae for high value product synthesis. Usually, these

products can only be synthesized through long or complex metabolic pathways. For

example, 35 to 51 steps were required for taxol de novo synthesis from glucose.

Modification of metabolic pathway in E. coli gave the highest taxadiene titer of 1.02

f
g/L, which is the precursor of taxol (Ajikumar et al. , 2010). However, the process

oo
was still far from industrial manufacturing. In another study, a biosynthetic pathway

pr
containing 10 genes for the synthesis of dihydrosanguinarine and sanguinarine from
e-
(R, S)-norlaudanosoline was constructed in S. cerevisiae (Fossati et al. , 2014). This
Pr

complex biosynthetic pathway only resulted in 1.5% conversion to

dihydrosanguinarine. Generally, biosynthetic efficiency is significantly reduced when


al

multiple genes are simultaneously introduced into a single microorganism, as this will
rn

cause overwhelming metabolic stresses to chassis. This universal challenge could be


u

potentially overcome through rational design using microbial consortia, which could
Jo

modularize and segregate one biosynthetic pathway into multiple separate cells.

Therefore, each chassis strain can be engineered independently to achieve optimal

function of the combined pathway (Zhang and Wang, 2016).

Table 2 summarized the most recent progress in valuable compound production.

Analyzing these successful attempts according to the types of microorganism

recruited in microbial consortia systems, three groups of bacteria-bacteria,

eukaryote-eukaryote and bacteria-eukaryote have been categorized, providing specific


Journal Pre-proof

experience for more artificial microbial consortium construction.In bacteria-bacteria

consortia systems, E. coli was the most commonly used chassis. Zhang et al. (2015b)

designed a novel E. coli-E. coli co-culture for the production of cis, cis-muconic acid

(MA) and 4-hydroxybenzoic acid (4HB), which both are important platform

intermediates for value-added compounds, such as adipic acid, terephthalic acid,

muconic acid and vanillyl alcohol (Sengupta et al. , 2015, Wang et al. , 2018b). In this

f
study, a first E. coli was used for conversion of glucose into the intermediate

oo
3-dehydroshikimic acid (DHS), which was assimilated and subsequently converted to

pr
MA or 4HB by the second E. coli strain. To eliminate carbon source competition
e-
between these two strains, the phosphotransferase system in the first strain was
Pr

removed, and the xylose isomerase gene xylA, which catalyzes the inter-conversion of

D-xylose and D-xylulose, was deleted in the second strain. The resulting consortium
al

could consume xylose and glucose simultaneously. By using this strategy, the
rn

limitations of high-level intermediate secretion and low-efficiency of sugar mixture


u

utilization were successfully overcome. This principle has also been used for the
Jo

production of cadaverine, the desired raw material of bio-polyamides, from a mixture

of glucose and glycerol (Wang et al. , 2018a), and the production of glycosides from a

mixture of glucose and xylose (Liu et al. , 2018a). Furthermore, through

accommodating the upstream and downstream pathways into two independent E. coli

strains, Zhang et al. constructed a microbial consortia system, in which glycerol was

used as the sole carbon source to support the growth of both strains as well as MA

production. The E. coli-E. coli co-culture can be used for complex biosynthesis
Journal Pre-proof

pathway engineering in the context of sole carbon source, regardless of the growth

competition between these two strains (Zhang et al. , 2015a). By assembling

engineered 4-vinylphenol or 4-vinylcatechol producer modules with

cyanidin-3-O-glucoside producer recombinant cell, Akdemir et al ( 2019) first

reported the synthesis of important red wine pigments pyranoanthocyanins using E.

coli co-cultures. Using this approach, the pyranoanthocyanins production are more

f
stable than the traditional extraction method from plants. Moreover, synthetic E.coli

oo
consortia systems have been designed for more and more valuable compounds

pr
production, including ester compounds like caffeoylmalic acid (Li et al. , 2018),
e-
terpene compounds like α-pinene (Niu et al. , 2018), polyphenol compounds like
Pr

resveratrol (Camacho-Zaragoza et al. , 2016), resveratrol glucosides (Thuan et al. ,

2018b), amino acid derivative like 3-amino-benzoic acid (3AB) (Zhang and
al

Stephanopoulos, 2016), cadaverine (Sgobba et al. , 2018), flavonoids compounds like


rn

apigetrin (Thuan et al. , 2018a), glycoside compounds like salicylate


u

2-O-b-D-glucoside (Ahmadi et al. , 2016), and monolignols (Chen et al. , 2017) etc. In
Jo

addition to E. coli, the engineered co-culture of Gluconobacter oxydans and

Ketogulonicigenium vulgare was also developed for one-step production of

2-keto-L-gulonic acid (2-KGA), which is the precursor of vitamin C (Wang et al. ,

2016).

Compared with bacteria-bacteria consortia systems, the attempts of

eukaryote-eukaryote were seldom reported. A representative case is the biosynthesis

modules of monacolin J and its derivative lovastatin, a very important natural product
Journal Pre-proof

used in anti-hypercholesterolemia pharmaceuticals (Campbell and Vederas, 2010), can

be assembled into two Pichia pastoris strains. The microbial consortia cangenerate

250.8 mg/L of lovastatin and 593.9 mg/L of monacolin J from methanol (Liu et al. ,

2018b). Compared to the monoculture, the biosynthesis capability was improved by

2.2-fold for lovastatin and 13.4% for monacolin J.

Not only limited to the cooperation between prokaryotes, cross-species consortia

f
between bacteria and eukaryotes have also been developed for biosynthesis of

oo
complex valuable products. For example, Rodríguez-Bustamante et al. (2005) isolated

pr
a microbial consortium consisting of the yeast Trichosporon asahii and the bacterium
e-
Paenibacillus amylolyticus, where T. asahii was responsible for cleaving lutein to

β-ionone, and P. amylolyticus reduced β-ionone into 7,8-dihydro-β-ionone and


Pr

7,8-dihydro-β-ionol derivatives, which are the compounds present in tobacco aroma


al

note. In another cross-species co-culture of engineered E. coli and S. cerevisiae, 2


rn

mg/L of oxygenate taxane (potent chemotherapy agent) was produced by using


u

glucose as the sole carbon source, although neither of them can produce the paclitaxel
Jo

precursor (Zhou et al. , 2015). In this process, the engineered E. coli strain was

responsible for upstream de novo biosynthesis of taxadiene, which was then converted

to oxygenated taxanes by S. cerevisiae, in which a cytochrome P450 module was

efficiently expressed. However, ethanol produced by S. cerevisiae significantly

inhibited the growth of E. coli and the production of taxanes. Hence, to better control

the population allocation of these two strains, a mutualistic carbon-source method was

employed, where xylose was consumed and converted to acetate by E. coli, and then
Journal Pre-proof

acetate was further metabolized to oxygenated taxanes by S. cerevisiae. After the

genetic modification, a high level of oxygenated taxanes production of 33 mg/L was

finally obtained. Recently, Zhang et al. (2017) constructed a cross-culture between E.

coli and S. cerevisiae for improving the efficiency of naringenin production, which is

a value-added natural product and widely dispersed in the citrus plant of rutaceae

family. Specifically, the endogenous tyrosine pathway was introduced into E. coli for

f
high level production of tyrosine, which was subsequently converted into naringenin

oo
by a downstream engineered S. cerevisiae. As a result, 21.16 mg/L of naringenin was

pr
finally produced from xylose, showing an 8-fold increased titer compared to that of
e-
yeast monoculture. Such systems have also been designed to exchange tyrosine and
Pr

arginine between engineered yeasts.

Beyond the interaction of two microorganisms, poly-culture consortia containing


al

three or more strains were also successfully exploited for constructing a long tandem
rn

or parallel synthetic pathway . For example, an engineered polyculture containing E.


u

coli, Bacillus subtilis and Shewanella oneidensis has been designed for microbial
Jo

power generation (Liu et al. , 2017). In this process, E. coli firstly digested glucose to

produce lactate, which was used as carbon source and electron donor. Subsequently,

lactate was converted to riboflavin by B. subtilis as an electron shuttle. Finally, S.

oneidensis acted as the exoelectrogen to generate electricity. In return, S. oneidensis

oxidized lactate to produce acetate, which served as carbon source for E. coli and B.

subtilis. Consequently, all three species formed a cross-feeding microbial consortium,

which performed “better together” for energy generation. Under optimized condition,
Journal Pre-proof

11 mM of glucose was converted to stable electricity output of ~550 mV for more

than 15 days. The potential of modular co-culture engineering can address the

challenges associated with complex and non-linear biosynthetic pathways. By

employing mixed carbon substrate utilization to minimize upstream carbon flux

competition, an efficient microbial consortia system of three E.coli strains was

constructed for non-linear rosmarinic acid synthesis, which exhibited 38-fold

f
improvement in rosmarinic acid production compared to that of the mono-culture (Li

oo
et al. , 2019). Another polyculture example was the de novo synthesis of anthocyanin,

pr
an important health-promoting pigment (Jaakola, 2013). In this study, 15 enzymatic
e-
steps involved in the production of phenylpropanoic acids, flavanones, flavan-3-ols,
Pr

and anthocyanins were divided into 4 independent E. coli strains respectively,

realizing the first heterologous production of flavan-3-ols (Jones et al. , 2017). This
al

study represents the most complex synthetic consortia to date, providing a new
rn

paradigm in metabolic engineering for reconstituting the extensive metabolic


u

pathways in non-native hosts.


Jo

Although remarkable progress in terms of valuable compounds synthesis using

microbial consortia has been achieved, it remains a great challenge to realize

industrial applications using such multispecies cultures . Except for the modular strain

selection and compromised cultivation conditions optimization, which will be

discussed in the future perspectives, more practical questions should be considered:

① As discussed in the first section, the physical contact of different microorganisms

will cause the production of some secondary chemicals, which brings more
Journal Pre-proof

difficulties in controlling the microorganisms population structure, and whether the

induced secondary chemicals will affect the natural chemicals quality should be taken

into account; ②In contrast to single cell factories, each species within microbial

consortia will interact with each other through the interchange of substrates or

intermediates, and how to improve the efficiency of substrates or intermediates

transfer between cells is the key to construct a highly efficient microbial consortium.

f
In addition, the low concentration of some intermediate metabolites will bring

oo
difficulties for partner microorganism to sense and capture them efficiently. Therefore,

pr
some versatile fermentation equipments should be designed to enhance the substrates
e-
or intermediates transfer within a microbial consortium system; ③ Microbial
Pr

community systems are dynamic and it is difficult to achieve long-term production

stability (Roell et al. , 2019). Thus, it is premature to judge the potential of a microbial
al

consortium from a laboratory perspective. Therefore, more experiments in large scale


rn

and especially long-term cultivation processes should be conducted. As stability,


u

controllability, safety and costs are key elements in assessment of an industrial


Jo

production process, more attention should be paid to these aspects in later experiment

design.

Consolidated bioprocessing for lignocellulose biorefinery using microbial

consortia

Lignocellulose is the most abundant renewable resource in the world, which is

generally considered as a promising feedstock for the production of biofuels and


Journal Pre-proof

bio-based chemicals. However, converting recalcitrant biomass into biochemicals is a

complex process compromising many steps, which convergently accommodates the

production of saccharolytic enzymes, hydrolysis of carbohydrate components to

sugars, and fermentation of hexose and pentose sugars (Klein‐Marcuschamer et al. ,

2012). Nevertheless, separating these tasks will cause inevitably high costs and long

processing periods. Recently, consolidated bioprocessing (CBP), which combines the

f
production of lignocellulose degrading enzymes, lignocellulose hydrolysis and

oo
microbial fermentation in one step has been considered as an economical and efficient

pr
approach to produce desired products from polysaccharides (Olson et al. , 2012, Xin
e-
et al. , 2017). Previous CBP reviews addressed fermentative production of chemicals
Pr

with recombinant lignocellulosic microbes (Fig. 3a) (Jang et al. , 2012, Yang et al. ,

2015, Zhang et al. , 2011), or expression of lignocellulose degrading enzymes in


al

non-lignocellulosic microbes (Fig. 3b) (Edwards et al. , 2011, Favaro et al. , 2015,
rn

Hasunuma and Kondo, 2012). However, each strategy possesses a considerable


u

challenge due to the assembly of differential models into a single synthetic chassis
Jo

(Del Vecchio et al. , 2018). Moreover, it is difficult to find a suitable microorganism

with the capability to produce all desired lignocellulose degrading enzymes (van Zyl

et al. , 2013). Activities of heterologously produced lignocellulose degrading enzymes

in current CBP microorganisms remained at only several hundred filter paper unit

(FPU)/L (den Haan et al. , 2015, Guo et al. , 2018, Singh et al. , 2018), which were

notably lower than that from native cellulolytic fungi, e.g. Trichoderma reesei, a

well-known cellulolytic fungus, in which tens of thousands FPU/L can be achieved.


Journal Pre-proof

A promising alternative is microbial consortia-based bioprocessing, which

consists of multiple microorganisms with complementary metabolic functions, so that

difficult tasks can be divided across a diverse subset of microorganisms. Especially,

co-cultivating of lignocellulose degrading strains with chemical producing microbes

exhibits superior advantages (Fig 3c). When a stable microbial consortium of

wild-type T. reesei and wild-type Lactobacilli sp. were co-cultivated, 19.8 g/L of

f
lactic acid corresponding to 85.2% of the theoretical maximum yield was obtained

oo
from non-detoxified steam-pretreated beech wood (Shahab et al. , 2018). Buzzini et al.

pr
designed a microbial co-culture of wild-type Debaryomyces castellii and wild-type
e-
Rhodotorula glutinis for the production of carotenoids from oligosaccharides and
Pr

dextrins from corn syrup. D. castellii was applied to hydrolyze the raw substrate to

maltose and glucose, which was subsequently converted to carotenoids by R. glutinis.


al

Finally, 8.2 mg/L of carotenoid was produced from corn syrup in the fed-batch
rn

co-culture system (Buzzini, 2001). Through using a symbiotic consortia of the


u

engineered S. cerevisiae and the cellulolytic bacterium Actinotalea fermentans,


Jo

methyl halide production was achieved from unprocessed biomass, such as

switchgrass, corn stover, sugar cane bagasse and poplar (Bayer et al. , 2009).

Moreover, Sgobba et al. (2018) established an E. coli-Corynebacterium glutamicum

mutualism consortium, which realized the production of L-lysine and its derivatives

directly from starch. In detail, α-amylase from S. griseus was heterologously

introduced into starch-negative E. coli, allowing it to hydrolyze starch as carbon

source. The generated glucose from starch by E. coli was then used to feed C.
Journal Pre-proof

glutamicum. In return, C. glutamicum provided lysine for lysine auxotrophic E. coli to

facilitate its growth. The mutualism of each species guaranteed the stable cooperation

of this microbial consortia system.

Particularly, the major contribution of microbial consortia CBP was applied for

bioenergy production, such as bioethanol, biobutanol, microbial lipid and H2 (Table 3).

For bioethanol production, Patle and Lal demonstrated that a microbial community of

f
Zymomonas mobilis and Candida tropicalis could transform enzymatically

oo
hydrolyzed lignocellulosic biomass into ethanol with a high yield of 97.7% (Patle and

pr
Lal, 2007). A polyculture of T. reesei, S. cerevisiae and Scheffersomyces stipitis could
e-
achieve cellulolytic enzyme production, hexose conversion and pentose sugar
Pr

utilization in one pot, realizing ethanol de novo synthesis from slurry dilute acid

pretreated wheat straw without detoxification (Brethauer and Studer, 2014). Similarly,
al

more microbial consortia CBP systems for bioethanol, biobutanol and isobutanol
rn

production among bacteria-bacteria, fungi-bacteria and fungi- fungi co-culture have


u

been designed (Minty et al. , 2013, Patle and Lal, 2007, Suriyachai et al. , 2013,
Jo

Zuroff et al. , 2013). For acetone-butanol-ethanol (ABE) production, Valdez-Vazquez

et al. developed a consortium composed of Clostridium beijerinckii 10132 and

Clostridium cellulovorans 35296, which could produce ABE from biologically

pretreated wheat straw, resulting in 23.3 g/L of total ABE with 3.7 g/L of ethanol, 14.2

g/L of butanol and 5.4 g/L of acetone (Valdez-Vazquez et al. , 2015). In parallel, Wen

et al. (2014) constructed a stable symbiotic consortium through co-culturing a

cellulolytic, anaerobic, butyrate-producing mesophile of C. cellulovorans 743B with a


Journal Pre-proof

non-cellulolytic, solventogenic bacterium of C. beijerinckii NCIMB 8052, realizing

solvents production from alkali extracted deshelled corn cobs (AECC). Under

optimized conditions, the microbial consortium degraded 68.6 g/L of AECC and

produced 11.8 g/L of solvents (2.6 g/L of acetone, 8.3 g/L of butanol, and 0.9 g/L of

ethanol) in less than 80 h, with 17.2% solvents conversion rate. Further genetic

modification of this consortium improved the solvent production to 22.1 g/L from

f
83.2 g/L of lignocellulose hydrolysate, and AECC degradation was increased by

oo
21.3%, while solvents titer was enhanced by 87.2% (Wen et al. , 2017). In terms of

pr
microbial lipid production, Papone et al. (2016) reported a co-culture of microalgae
e-
Chlorella sp. KKU-S2 and yeast Toluraspora globosa YU5/2. A maximum biomass
Pr

concentration of 6.90 g/L with a lipid concentration of 0.33 g/L was obtained using

sugarcane molasses as the substrate. Encouraged by the advantages of co-culture


al

systems over pure monocultures, many attempts have also been made in biogas and
rn

biohydrogen production (Ike et al. , 1999, Laxman Pachapur et al. , 2015, Masset et
u

al. , 2012).
Jo

Generally, substrate degradation efficiency is still the rate limiting step in CBP.

To further improve the performance efficiency of microbial consortia in CBP, the

substrate degradation rate should be enhanced to provide higher monosaccharide

concentrations. As lignocellulose possesses a very complex structure, a single

microbial strain can not efficiently secrete all desirable components of lignocellulose

degradation complex. Microbial co-culture was reported to increase the production of

cellulases and hemicellulases complex (Kumar et al. , 2008). For example, high
Journal Pre-proof

production of cellulase and β-glucosidase was obtained in co-culture of A. ellipticus

and A. fumigatus (Gupte and Madamwar, 2010). Microbial consortia of Pleurotus

ostreatus and Phanerochaete chrysosporium also resulted in the improved production

of ligninolytic enzymes (Verma and Madamwar, 2002). Moreover, higher

α-cellobiohydrolase, β-glucosidase, β-galactosidase and laccase activities were found

for A. oryzae in combination with other fungi, in particular with P. chrysosporium (Hu

f
et al. , 2011). The overall activity of degrading enzyme in the microbial consortia is

oo
not the sole sum of activities of individual microorganism species, which in some

pr
cases exceeded the sum of single incubations.Co-cultivation of these lignocellulose
e-
degrading consortia with biological detoxification species could further improve the =
Pr

efficiency of microbial consortia CBP systems (Kuhar et al. , 2015).

Bioremediation using microbial consortia


al

The growing population and anthropogenic activities are constantly threatening


rn

the ecosystem, such as water quality deterioration, heavy metals pollution and loss of
u

soluble phosphorus, etc (Ahmed et al. , 2019, Campbell-Lendrum and Prüss-Ustün,


Jo

2019, Critchell et al. , 2019, Ibrahim et al. , 2016, Walker et al. , 2019). To achieve

green and sustainable development, biological technologies using specific

microorganisms to degrade various pollutants into non-toxic or less toxic compounds

provides an eco-friendly option to remedy the impaired environment(Azubuike et al. ,

2016). However, the efficiency for biodegradation and bioremediation of

environmental pollutants using single strains is still very low and restricted (Villegas

et al. , 2014). Therefore, more and more attention has been shifted towards microbial
Journal Pre-proof

consortia, since their inherent multiple function, robust and adaptable characteristics.

In terms of wastewater eutrophication, the main task is to remove the excess

elements such as nitrogen (N), phosphorus (P), pollutants and toxins (Mujtaba and

Lee, 2016). Current biological processesinclude anaerobic digestion, nitrification and

denitrification steps, however, several cycles of these three steps are required to

achieve the EU legislation accepted nutrient levels, and each step requires several

f
tanks and internal recycles of activated sludge, resulting in an overall increase of

oo
process cost, complexity and energy input (Gonçalves et al. , 2017). Comparatively,

pr
microalgal consortia (microalgae and bacteria/fungi) provide an efficient approach for
e-
water remediation. For instance, co-culture of microalgae Scenedesmus dimorphus
Pr

and nitrifiers enhanced the removal of N and P from wastewater by 3.4 and 6.5 times,

respectively compared to those using nitrifiers-only (Choi et al. , 2017). Co-culture of


al

microalga Chlorella vulgaris and bacterium P. putida also showed higher removal
rn

efficiency of both nutrients (N and P) and chemical oxygen demand (COD) than each
u

monoculture (Mujtaba et al. , 2017). Using co-culture of Scenedesmus sp. and


Jo

anaerobic sludge, 89% N removal and 80% P removal from starch wastewater were

achieved (Ren et al. , 2015). In the view of symbiotic relationship, microalgae can

release organic compounds and O 2 during photosynthesis, which can be used as

carbon and energy sources by bacteria/fungi. In return, bacteria/fungi will provide

CO2 and growth promoting factors for microalgae, such as vitamins and siderophores

(Fig. 4) (Abinandan et al. , 2018, Bordel et al. , 2009). In addition, microalgae can

also serve as refuge for bacteria, protecting them from hostile environmental
Journal Pre-proof

conditions (Byappanahalli et al. , 2009, Mandal et al. , 2011, Unnithan et al. , 2014).

In addition to wastewater purification, biomass harvested from effluent can also be

used as sustainable substrates (or feedstocks) for biofuels or biochemicals production

(Bhatia et al. , 2017, Wrede et al. , 2014), and animal feed preparation (Stiles et al. ,

2018), which further improving process economy. Despite the obvious advantages of

microalgae-bacteria/fungi symbiosis systems, the availability of land, sufficient light

f
intensities and appropriate temperature still seriously limits its scale-up progress in

oo
wastewater treatment (Osundeko et al. , 2019). In addition, the recovery and

pr
processing of microalgae from the cultivation medium are still a challenge, since
e-
needs a high energy input is needed (Osundeko et al. , 2019). More functional facility
Pr

and more effective harvesting technology need to be developed to accommodate

microalgae treatment and decrease cell mass recovery cost.


al

Heavy metals, such as zinc and nickel are considered as the most hazardous
rn

pollutants, because they can affect the structure of nucleic acids and proteins.
u

Currently, it is impossible to adopt a single method to treat heavy-metal-polluted


Jo

wastewater (Bhatia et al. , 2018). Though various microbes with different

metal-reduction potential have been identified (Sathiyanarayanan et al. , 2016,

Shafique et al. , 2017), the metal removal capability using monoculture is still

restricted, as the compositions in wastewater are too diverse and complicated.

Microbial consortia contain various robust metal-degrading microorganisms provides

an economical and eco-friendly option for heavy-metal degradation through division

of labor. For instance, co-culture of algae Scenedesmus and Pseudokirchneriella was


Journal Pre-proof

able to absorb a mixture of Zn(II) and Ni(II) ions (Kipigroch et al. , 2012). Ilamathi et

al. used an alginate-bead immobilized mixed-culture of yeast and different bacteria, P.

aeruginosa, B. subtilis and E. coli in a liquid-solid fluidized bed for heavy metal

biosorption, which resulted in 84.62%, 67.17%, 49.25% and 61.02% recovery of

copper, cadmium, chromium and nickel, respectively (Ilamathi et al. , 2014).

Azo dyes, the largest chemical class of dyes with the greatest variety of colors are

f
commonly released from textile, dyestuff and dyeing industry, which cause serious

oo
environmental pollution because of their color, biorecalcitrance and potential toxicity

pr
to animal and human (Senan and Abraham, 2004). Nevertheless, it is very difficult to
e-
decolorize these dyes by common physical and chemical methods (Khan et al. , 2013).
Pr

Microbial strains are able to decolorize azo dyes, but the degradation products are

frequently toxic aromatic amines or metabolites that are even more difficult to
al

degrade than the parent dye (Solís et al. , 2012). Microbial consortia treatment
rn

systems could achieve a higher degree of biodegradation and mineralization due to the
u

synergistic metabolic activities of the microbial community (Shanmugam et al. ,


Jo

2017). For instance, orange II can be completely decolorized by microbial consortia

of Enterobacter cloacae and Enterococcus casseliflavus, while only 10%

decolorization rate was obtained by E. cloacae and 23% by E. casseliflavus alone

(Chan et al. , 2011). The complete decolorization of scarlet R was accomplished

within only 3 h by microbial consortia of Proteus vulgaris and Micrococcus

glutamicus, superior to 14 h by P. vulgaris and 20 h by M. glutamicus alone (Saratale

et al. , 2009). The decolorization of sulfonated reactive dye Green HE4BD using
Journal Pre-proof

microbial consortia of P. vulgaris and Micrococcus glutamicus is significantly higher

than that using the individual microorganisms (Saratale et al. , 2010). In these

microbial consortia, individual strains may attack the dye molecule at different

positions or even utilize resulting degradation products, such as the toxic aromatic

amines produced by partner strains for further decomposition (Saratale et al. , 2011).

Microbial consortia also demonstrate unique capability in degradation of other

f
pollutants, like pesticides, antibiotics and other toxins. For example, co-culture of

oo
Pseudomonas and Staphylococcus was shown to be more efficient in removing phenol

pr
than their monocultures (Senthilvelan et al. , 2014). Co-culture of Serratia and
e-
Trichosporon sp. could completely mineralize chlorpyrifos, one of the most widely
Pr

used organophosphate insecticide (Xu et al. , 2007). Similarly, microbial consortia of

Klebsiella pneumoniae and Ralstonia sp. showed better tolerance to thiocyanate with
al

a degradation rate of 500 mg/L/d. Poly-culture of Arthrobacter sp. NB1, Serratia sp.
rn

NB2 and Stenotrophomonas sp. NB3 showed enhanced degradation efficiency of


u

nitrobenzene compared with monoculture (Jin et al. , 2012). Moreover, other studies
Jo

showed that microbial consortia have higher removal efficiencies in polycyclic

aromatic hydrocarbons (PAHs) than monocultures (González et al. , 2011, Simarro et

al. , 2011).

Future perspectives

The application of synthetic microbial consortia in the area of health, chemistry,

energy and environment, etc. has prompted the rapid development of biotechnology.
Journal Pre-proof

Taking industrial biotechnology as an example, microbial consortia could synthesize

more complex chemicals, which only exist in plants and can not be synthesized by

single microorganism (Song et al. , 2014 ). Synthetic microbial consortia could

accomplish this complex task through the division of labor. However, these synthetic

microbial consortia are still relatively simple, in which usually two or three wild type

or genetically engineered microorganisms were recruited. In addition, the construction

f
of synthetic microbial consortia is still maintained at trial stage and lack of theory

oo
guide, as microorganisms assembly seems random. To construct a more robust, stable

pr
and controllable microbial consortium, several challenges must be addressed.
e-
Stable coexistence of different species within one microbial consortium is
Pr

prerequisite for microbial consortia construction. Microorganisms within a microbial

consortium often possess different growth features, such as temperature, pH, or


al

dissolved oxygen et al. To harmonize the mismatching of growth conditions,


rn

sequential cultivation mode through multistage conditions regulation is the most


u

commonly used one, in which growth conditions were adjusted based on different
Jo

strains (Shahab, 2019). However, this would lead to longer fermentation duration and

affect final product productivity. Alternatively, more specific bio-materials and

facilities could be designed to obtain stable microbial consortia. For example,

microcapsule and microfluidic laminar flow techniques can help create a relatively

optimal microenvironment for individual cells, and the mechanical separation of each

microbial species would not affect microbial growth in a complex microbial consortia

system (Wang et al. , 2019, Lindemann et al. , 2016). And also, bioreactor equipped
Journal Pre-proof

with some inlets, on which parceled with gas permeable dense membranes or other

nutrient selective permeable membrane can form a gradient cultivation condition,

which could meet the requirement for different nutrient of microbial consortia

members (Shahab et al. , 2020).

A rational population structure and substrates allocation modes are another two

key factors to achieve an efficient synthetic microbial consortium. Regulation of

f
inoculum size and time of different species within microbial consortia is the most

oo
direct and efficient way to adjust the population structure. However, the substrate

pr
competition may exist if each species was fed by the same carbon source, and this
e-
would lead to the uncontrollable growth of each species within microbial consortia.
Pr

Alternatively, designing a parallel pathway, where the microbial consortia members

can specifically utilize different carbon sources, and this would efficiently reduce the
al

growth competition. For instance, pentose and hexose utilization pathways can be
rn

metabolically constructed in different species (Zhang et al. , 2015b). This would not
u

only eliminate the substrate competition, but also achieve simultaneous utilization of
Jo

mixed sugars in lignocellulosic hydrolysate. Another approach is to construct a

sequential utilization mode of substrate and intermediate. For example,

monosaccharides can be first metabolized into some intermediates, such as acetates by

first strain. Then partner strain, which lost the utilization capability of

monosaccharides can further convert intermediates to final product (Zhou et al. ,

2015). The ratio of these two species could be balanced by the substrate and

intermediate ratio. Recently, with the deeper understanding of signal communication


Journal Pre-proof

mechanism between different species of microbial consortia, QS system could also be

constructed within microbial consortia. Through the inter-communication of each

member, growth related genes could be regulated at different levels to further adjust

growth ratio of different species (You et al. , 2004).

Efficient mass transfer, including the intermediates, energy and cofactors can

improve the product efficiency by using microbial consortia. In contrast to single cell

f
factories, intermediates have to be delivered to other partner strains. The complex

oo
metabolic pathways within all species of microbial consortia made the metabolic

pr
regulation more difficult. Herein, a spatial organization concept, or named
e-
3-dimensional (3D) began to show up. In a 3D consortium, microorganisms are
Pr

well-organized, which will ensure efficient population structure of members with

lower fitness, enhance the efficient metabolite and signal molecule transfer among
al

different cells, and/or improve the overall resilience to environmental stresses


rn

(Agapakis et al. , 2012, Johns et al. , 2016). In fact, such 3D microbial consortia
u

widely exist in natural environment. A typical example is the anaerobic sludge granule,
Jo

which is a spatial organization of many species of microorganisms. Acidogens would

wrap outside of granules to break down complex organic molecules into acids, which

are subsequently transformed to H2 by acetogens in middle layer. H2 and CO 2

produced by these outer layers will be consumed by methanogens in central core

(MacLeod et al. , 1990). This spatial organization creates optimal growth environment

for different species of microorganisms in these three layers, and realizes efficient

mass transfer with orderly arranged microbes.


Journal Pre-proof

To construct synthetic 3D microbial consortia, two strategies including

self-organization and artificial-organization can be adopted. Self-organization can

immobilize core microorganisms in bacterial formed matrix without the use of

synthetic binding agents. One inspirational example is the microbial consortia

consisting of bacterium Acetobacter aceti and photosynthetic microalga

Chlamydomonas reinhardtii (Das et al. , 2016). Within this system, A. aceti produced

f
cellulose and acetate mats at the air-water interface when growing in liquid cultures.

oo
Consequently, C. reinhardtii thrived in an acetate-rich medium and produced oxygen

pr
for A. aceti. By co-cultivating these two microorganisms, researchers created a
e-
composite material using cellulose produced in situ by Acetobacter, which offered a
Pr

matrix to favor the cell growth and entrap microalgae cells (Das et al. , 2016). Another

example is the widespread existence of biofilms, which is formed by microorganism


al

aggregation for surviving harsh environmental conditions (Azeredo et al. , 2017). In


rn

biofilms, bacteria live in a self-produced matrix of hydrated extracellular polymeric


u

substances (EPS) and organize themselves into a coordinated functional community,


Jo

in which cells can share nutrients and are sheltered from harmful factors in the

environment, such as detergents and antibiotics (Flemming and Wingender, 2010). As

for artificial-organization strategy, smart devices or materials can be designed to help

form spatial-arranged microbial consortia. Various physical segregation techniques

have been developed recently (Wondraczek et al. , 2019). For example, microfluidic

and microwell devices have been used to help building microbial communities, where

individual species are grown in separated chambers, allowing intermediates to


Journal Pre-proof

exchange freely (Kim et al. , 2008). Strategies using inkjet printing cells and

multiphoton 3D printing of gelatin techniques can allow specific members to be

arranged in defined geometric patterns, which has been used for microbial

communities construction with more complex structures (Connell et al. , 2013, Hays

et al. , 2015). Moreover, fiber structure like calcium alginate fibers with one species in

the core and another in the exterior can also realize strains ordered arrangement with a

f
complex consortium system (Kim et al. , 2010). With the increasing understanding of

oo
systems biology and metabolic dynamics in various microbial consortia, more

pr
biocompatible and functionalized supporting materials for organizing microbial
e-
consortia will be developed in the future.
Pr

Not only limited to cultivation strategies and biomaterials or facilities design,


al

more advanced computational analysis tools to predict community behavior should be


rn

developed (Jones et al. , 2016, Klitgord and Segre, 2010, Lindemann et al. , 2016,
u

Manikandan and Viruthagiri, 2010, Panda et al. , 2010, Park et al., 2011, Sun et al. ,
Jo

2010, Suriyachai et al. , 2013). For this purpose, Minty et al. (2013) have designed an

equation model consisting of 50 parameters to describe and predict the behavior of a

symbiotic consortium of E. coli and T. reesei. The sophisticated model allowed the

identification of key parameters during isobutanol production from lignocellulosic

biomass, as well as in-depth evaluation of the co-culture stability, which marked a

significant progress in developing modular co-culture engineering. Moreover, the

development of computational tools can also help elaborate the interaction mechanism
Journal Pre-proof

of many natural microbial consortia models, such as methanogenic communities

(Stams and Plugge, 2009), the inner life of sludge (Liu et al. , 2002), soil and

rhizosphere ecosystems (Kent and Triplett, 2002), and marine environment (Ka and

Keerthi, 2017), which will further provide theoretical guidance for construction of

synthetic microbial consortia.

Conclusion

f
Synthetic microbial consortia possess several promising characteristics and

oo
potential over monocultures, which harness the resources and metabolic power of

pr
multiple microbial strains to meet the requirements of discovery of novel chemicals
e-
and functional reconstitution of complex pathways. More importantly, microbial
Pr

consortia offer a viable option to share the undesired metabolic burden among

different microbial strains and go beyond the limit of the metabolic capacity within a
al

single microbial strain. And it also presents a new approach for efficient utilization of
rn

complex substrates and remediation of the environment. However, many uncertain


u

factors, such as the unclear mechanism of intercellular communications, change of


Jo

microbial population structure and compromise of cultivation conditions, etc. still

exist when synthetic microbial communities were constructed. To address these

problems, a comprehensive study focusing on interaction mechanism of natural

microbial consortia, introduction of functional supporting materials, and development

of analytical and predictable computational models should be carried out in the future.

Ultimately, high-efficient, stable and controllable synthetic microbial communities

will be rationally designed for many intriguing applications in the foreseeable future.
Journal Pre-proof

Acknowledgements

This work was supported by National Key Research and Development Program

of China (2018YFA0902200), National Natural Science Foundation of China (No.

21978130, No. 31961133017, No.21706125), Jiangsu Province Natural Science

Foundation for Youths (BK20170993, BK20170997), the Jiangsu Synergetic

Innovation Center for Advanced Bio-Manufacture, Jiangsu Key Lab of

f
Biomass-based Green Fuels and Chemicals Foundation (JSBEM201908), and Project

oo
of State Key Laboratory of Materials-Oriented Chemical Engineering (ZK201601).

Reference pr
Abinandan S., et al., 2018. Nutrient removal and biomass production: advances in
e-
microalgal biotechnology for wastewater treatment. Crit. Rev. Biotechnol. 38,
1244-1260.
Pr

Agapakis CM., Boyle PM., Silver PA., 2012. Natural strategies for the spatial
optimization of metabolism in synthetic biology. Nat. Chem. Biol. 8, 527.
Ahmadi MK., et al., 2016. E. coli metabolic engineering for gram scale production of
al

a plant-based anti-inflammatory agent. Metab. Eng. 38, 382-388.


Ahmed N., Thompson S., Glaser M., 2019. Global aquaculture productivity,
rn

environmental sustainability, and climate change adaptability. Environ. Manage. 63,


159-172.
u

Ajikumar PK., et al., 2010. Isoprenoid pathway optimization for Taxol precursor
overproduction in Escherichia coli. Science. 330, 70-74.
Jo

Akdemir H., et al., 2019. Production of pyranoanthocyanins using Escherichia coli


co-cultures. Metab. Eng.
An X., et al., 2019. Engineering microbial consortia for high-performance cellulosic
hydrolyzates-fed microbial fuel cells. Front. Microbiol. 10, 409.
Azeredo J., et al., 2017. Critical review on biofilm methods. Crit. Rev. Microbiol. 43,
313-351.
Azubuike CC., Chikere CB., Okpokwasili GC., 2016. Bioremediation
techniques–classification based on site of application: principles, advantages,
limitations and prospects. World. J. Microb. Biot. 32, 180.
Balagaddé FK., et al., 2008. A synthetic Escherichia coli predator–prey ecosystem.
Mol Syst Biol. 4.
Basu S., et al., 2004. Spatiotemporal control of gene expression with pulse-generating
networks. P. Natl. Acad. Sci. USA. 101, 6355-6360.
Bayer TS., et al., 2009. Synthesis of methyl halides from biomass using engineered
Journal Pre-proof

microbes. J. Am. Chem. Soc. 131, 6508-6515.


Bennett JW., Chung K-T., 2001. Alexander Fleming and the discovery of penicillin.
Bertrand S., et al., 2014. Metabolite induction via microorganism co-culture: a
potential way to enhance chemical diversity for drug discovery. Biotechnol. Adv. 32,
1180-1204.
Bhatia SK., et al., 2018. Biotechnological potential of microbial consortia and future
perspectives. Crit. Rev. Biotechnol. 38, 1209-1229.
Bhatia SK., Bhatia RK., Yang Y-H., 2017. An overview of microdiesel—a sustainable
future source of renewable energy. Renew. Sust. Energ. Rev. 79, 1078-1090.
Bordel S., Guieysse B., Munoz R., 2009. Mechanistic Model for the reclamation of
industrial wastewaters using algal− bacterial photobioreactors. Environ. Sci. Technol.
43, 3200-3207.
Brakhage AA., Schroeckh V., 2011. Fungal secondary metabolites–strategies to

f
activate silent gene clusters. Fungal. Genet. Biol. 48, 15-22.

oo
Brethauer S., Studer MH., 2014. Consolidated bioprocessing of lignocellulose by a
microbial consortium. Energ. Environ. Sci. 7, 1446-1453.
pr
Buzzini P., 2001. Batch and fed‐ batch carotenoid production by Rhodotorula
glutinis–Debaryomyces castellii co‐ cultures in corn syrup. J. Appl. Microbiol. 90,
843-847.
e-
Byappanahalli MN., et al., 2009. Seasonal stability of Cladophora-associated
Salmonella in Lake Michigan watersheds. Water. Res. 43, 806-814.
Pr

Camacho- Zaragoza JM., et al., 2016. Engineering of a microbial coculture of


Escherichia coli strains for the biosynthesis of resveratrol. Microb. Cell. Fact. 15,
163.
al

Campbell-Lendrum D., Prüss-Ustün A., 2019. Climate change, air pollution and
noncommunicable diseases. B. World. Health. Organ. 97, 160.
rn

Campbell CD., Vederas JC., 2010. Biosynthesis of lovastatin and related metabolites
formed by fungal iterative PKS enzymes. Biopolymers. 93, 755-763.
u

Chagas FO., Dias LG., Pupo MT., 2013. A mixed culture of endophytic fungi
increases production of antifungal polyketides. J. Chem. Ecol. 39, 1335-1342.
Jo

Chan GF., et al., 2011. Identification and optimization of novel NAR-1 bacterial
consortium for the biodegradation of Orange II. Insight. Biotechnol. 1, 7-16.
Charusanti P., et al., 2012. Exploiting adaptive laboratory evolution of Streptomyces
clavuligerus for antibiotic discovery and overproduction. Plos. One. 7, e33727.
Chen L., et al., 2018. A system biology perspective on environment–host–microbe
interactions. Hum. Mol. Genet. 27, R187-R194.
Chen Z., et al., 2017. Metabolic engineering of Escherichia coli for microbial
synthesis of monolignols. Metab. Eng. 39, 102-109.
Choi K-J., et al., 2017. Co-culture consortium of Scenedesmus dimorphus and
nitrifiers enhances the removal of nitrogen and phosphorus from artificial wastewater.
KSCE. J. Civ. Eng. 1-7.
Connell JL., et al., 2013. 3D printing of microscopic bacterial communities. P. Natl.
Acad. Sci. USA. 110, 18380-18385.
Critchell K., et al., 2019. Plastic pollution in the coastal environment: current
Journal Pre-proof

challenges and future solutions. Estuar. Coast. 595-609.


Das AA., et al., 2016. Fabrication of living soft matter by symbiotic growth of
unicellular microorganisms. J. Mater. Chem. B. 4, 3685-3694.
Del Vecchio D., et al., 2018. Future systems and control research in synthetic biology.
Annu. Rev. Control. 45, 5-17.
Den Haan R., et al., 2015. Progress and challenges in the engineering of
non-cellulolytic microorganisms for consolidated bioprocessing. Curr. Opin. Biotech.
33, 32-38.
Edwards MC., et al., 2011. Addition of genes for cellobiase and pectinolytic activity
in Escherichia coli for fuel ethanol production from pectin- rich lignocellulosic
biomass. Appl. Environ. Microbiol. 77, 5184-5191.
Fang Z., et al., 2018. Engineering Escherichia coli Co‐ Cultures for Production of
Curcuminoids From Glucose. Biotech. J. 13, 1700576.

f
Favaro L., et al., 2015. Consolidated bioprocessing of starchy substrates into ethanol

oo
by industrial Saccharomyces cerevisiae strains secreting fungal amylases. Biotechnol.
Bioeng. 112, 1751-1760.
pr
Fischer J., et al., 2018. Fungal chromatin mapping identifies BasR, as the re gulatory
node of bacteria-induced fungal secondary metabolism. BioRxiv. 211979.
Flemming H-C., Wingender J., 2010. The biofilm matrix. Nat. Rev. Microbiol. 8, 623.
e-
Fossati E., et al., 2014. Reconstitution of a 10- gene pathway for synthesis of the plant
alkaloid dihydrosanguinarine in Saccharomyces cerevisiae. Nat. Commun. 5, 3283.
Pr

Ganesan V., et al., 2017. Heterologous biosynthesis of natural product naringenin by


co-culture engineering. Synth Syst Biotechnol. 2, 236-242.
Gilbert JA., et al., 2018. Current understanding of the human microbiome. Nat. Med.
al

4, 392.
Gonçalves AL., Pires JC., Simões M., 2017. A review on the use of microalgal
rn

consortia for wastewater treatment. Algal. Res. 24, 403-415.


González N., et al., 2011. Effect of surfactants on PAH biodegradation by a bacterial
u

consortium and on the dynamics of the bacterial community during the process.
Bioresource. Technol. 102, 9438-9446.
Jo

Guo Z-p., et al., 2018. Developing cellulolytic Yarrowia lipolytica as a platform for
the production of valuable products in consolidated bioprocessing of cellulose.
Biotechnol. Biofuels. 11, 141.
Gupte A., Madamwar D., 2010. Solid state fermentation of lignocellulosic waste for
cellulase and β‐ glucosidase production by cocultivation of Aspergillus ellipticus and
Aspergillus fumigatus. Biotechnol. Progr. 13, 166-169.
Gustavsson M., Lee SY., 2016. Prospects of microbial cell factories developed
through systems metabolic engineering. Microb. Biotechnol. 9, 610-617.
Hall GM., Howe J., 2012. The impact of synthetic biology in chemical
engineering—Educational issues. Educ. Chem. Eng. 7, e51-e55.
Hanemaaijer M., et al., 2015. Systems modeling approaches for microbial community
studies: from metagenomics to inference of the community structure. Front. Microbiol.
6, 213.
Hasunuma T., Kondo A., 2012. Development of yeast cell factories for consolidated
Journal Pre-proof

bioprocessing of lignocellulose to bioethanol through cell surface engineering.


Biotechnol. Adv. 30, 1207-1218.
Hays SG., et al., 2015. Better together: engineering and application of microbial
symbioses. Curr. Opin. Biotech. 36, 40-49.
Hu HL., et al., 2011. Improved enzyme production by co-cultivation of Aspergillus
niger and Aspergillus oryzae and with other fungi. Int. Biodeter. Biodegr. 65,
248-252.
Ibrahim RK., et al., 2016. Environmental application of nanotechnology: air, soil, and
water. Environ. Sci. Pollut. R. 23,13754-13788.
Ike A., et al., 1999. Photoproduction of hydrogen from raw starch using a ha lophilic
bacterial community. J. Biosci. Bioeng. 88, 72-77.
Ilamathi R., Nirmala G., Muruganandam L., 2014. Heavy metals biosorption in liquid
solid fluidized bed by immobilized consortia in alginate beads. J. Bioprocess. Biotech.

f
4, 1-6.

oo
Jaakola L., 2013. New insights into the regulation of anthocyanin biosynthesis in
fruits. Trends. Plant. Sci. 18, 477-483.
pr
Jang Y-S., et al., 2012. Enhanced butanol production obtained by reinforcing the
direct butanol-forming route in Clostridium acetobutylicum. MBio. 3, e00314-12.
Jin D., et al., 2012. Optimization of a bacterial consortium for nitrobenzene
e-
degradation. Water. Sci. Technol. 65, 795-801.
Johns NI., et al., 2016. Principles for designing synthetic microbial communities. Curr.
Pr

Opin. Microbiol. 31, 146-153.


Joint I., et al., 2002. Cell-to-cell communication across the prokaryote-eukaryote
boundary. Science. 298, 1207.
al

Jones JA., et al., 2017. Complete biosynthesis of anthocyanins using E. coli


polycultures. MBio. 8, e00621-17.
rn

Jones JA., et al., 2016. Experimental and computational optimization of an


Escherichia coli co-culture for the efficient production of flavonoids. Metab. Eng. 35,
u

55-63.
König CC., et al., 2013. Bacterium induces cryptic meroterpenoid pathway in the
Jo

pathogenic fungus Aspergillus fumigatus. Chembiochem. 14, 938-942.


Ka AK., Keerthi T., 2017. Co-culture as the novel approach for drug discovery from
marine environment. Nov. Appro. Drug. Des. Dev. 2, 1-4.
Kaeberlein T., Lewis K., Epstein SS., 2002. Isolating" uncultivable" microorganisms
in pure culture in a simulated natural environment. Science. 296, 1127-1129.
Kent AD., Triplett EW., 2002. Microbial communities and their interactions in soil
and rhizosphere ecosystems. Annu. Rev. Microbiol. 56, 211-236.
Khan R., Bhawana P., Fulekar M., 2013. Microbial decolorization and degradation of
synthetic dyes: a review. Rev. Environ. Sci. Bio. 12, 75-97.
Kim HJ., et al., 2008. Defined spatial structure stabilizes a synthetic multispecies
bacterial community. P. Natl. Acad. Sci. USA. 105, 18188-18193.
Kim HJ., Du W., Ismagilov RF., 2010 Complex function by design using spatially
pre-structured synthetic microbial communities: degradation of pentachlorophenol in
the presence of Hg (II). Integr Biol. 3,126-133.
Journal Pre-proof

Kipigroch K., Janosz-Rajczyk M., Wykrota L., 2012. Biosorption of heavy metals
with the use of mixed algal population. Arch. Environ. Prot. 38, 3-10.
Klein‐ Marcuschamer D., et al., 2012. The challenge of enzyme cost in the
production of lignocellulosic biofuels. Biotechnol. Bioeng. 109, 1083-1087.
Klitgord N., Segre D., 2010. Environments that induce synthetic microbial
ecosystems. Plos. Comput. Biol. 6, e1001002.
Kuhar F., Castiglia V., Levin L., 2015. Enhancement of laccase production and
malachite green decolorization by co-culturing Ganoderma lucidum and Trametes
versicolor in solid-state fermentation. Int. Biodeter. Biodegr. 104, 238-243.
Kumar R., Singh S., Singh OV., 2008. Bioconversion of lignocellulosic biomass:
biochemical and molecular perspectives. J. Ind. Microbiol. Biot. 35, 377-391.
Kurosawa K., et al., 2008. Rhodostreptomycins, antibiotics biosynthesized following
horizontal gene transfer from Streptomyces padanus to Rhodococcus fascians. J. Am.

f
Chem. Soc. 130, 1126-1137.

oo
Laxman Pachapur V., et al., 2015. Co‐ culture strategies for increased biohydrogen
production. Int. J. Energ. Res. 39, 1479-1504.
pr
Li T., et al., 2018. Production of caffeoylmalic acid from glucose in engineered
Escherichia coli. Biotechnol. Lett. 40, 1057-1065.
Li Z., Wang X., Zhang H., 2019. Balancing the non-linear rosmarinic acid
e-
biosynthetic pathway by modular co-culture engineering. Metab. Eng. 54, 1-11.
Lindemann SR., et al., 2016. Engineering microbial consortia for controllable outputs.
Pr

ISME. J. 10, 2077.


Liu W-T., Chan O-C., Fang HH., 2002. Characterization of microbial community in
granular sludge treating brewery wastewater. Water. Res. 36, 1767-1775.
al

Liu X., et al., 2018. Convergent engineering of syntrophic Escherichia coli coculture
for efficient production of glycosides. Metab. Eng. 47, 243-253.
rn

Liu Y., et al., 2017. A three-species microbial consortium for power generation. Energ.
Environ. Sci. 10, 1600-1609.
u

Liu Y., et al., 2018. Engineered monoculture and co-culture of methylotrophic yeast
for de novo production of monacolin J and lovastatin from methanol. Metab. Eng. 45,
Jo

189-199.
Lloyd-Price J., Abu-Ali G., Huttenhower C., 2016. The healthy human microbiome.
Genome. Med. 8, 51.
MacLeod F., Guiot S., Costerton J., 1990. Layered structure of bacterial aggregates
produced in an upflow anaerobic sludge bed and filter reactor. Appl Environ
Microbiol. 56,1598-1607.
Mandal SK., Singh RP., Patel V., 2011. Isolation and characterization of
exopolysaccharide secreted by a toxic dinoflagellate, Amphidinium carterae Hulburt
1957 and its probable role in harmful algal blooms (HABs). Microb. Ecol. 62,
518-527.
Manikandan K., Viruthagiri T., 2010. Optimization of C/N ratio of the medium and
fermentation conditions of ethanol production from tapioca starch using co–culture of
Aspergillus niger and Saccharomyces cerevisiae. Int. J. Chem. Tech. Res. 2, 947-955.
Marmann A., et al., 2014. Co-cultivation—a powerful emerging tool for enhancing
Journal Pre-proof

the chemical diversity of microorganisms. Mar. Drugs. 12, 1043-1065.


Martínez I., et al., 2016. Engineering synthetic bacterial consortia for enhanced
desulfurization and revalorization of oil sulfur compounds. Metab. Eng. 35, 46-54.
Masset J., et al., 2012. Fermentative hydrogen production from glucose and starch
using pure strains and artificial co-cultures of Clostridium spp. Biotechnol. Biofuels. 5,
35.
McCarty NS., Ledesma-Amaro R., 2018. Synthetic biology tools to engineer
microbial communities for biotechnology. Trends. Biotechnol. 37,181-197
Michie KL., Cornforth DM., Whiteley M., 2016. Bacterial tweets and podcasts#
signaling# eavesdropping# microbialfightclub. Mol. Biochem. Parasit. 208, 41-48.
Minty JJ., et al., 2013. Design and characterization of synthetic fungal-bacterial
consortia for direct production of isobutanol from cellulosic biomass. P. Natl. Acad.
Sci. USA. 110, 14592-14597.

f
Mujtaba G., Lee K., 2016. Advanced treatment of wastewater using symbiotic

oo
co-culture of microalgae and bacteria. Appl. Chem. Eng. 27, 1-9.
Mujtaba G., Rizwan M., Lee K., 2017. Removal of nutrients and COD from
pr
wastewater using symbiotic co-culture of bacterium Pseudomonas putida and
immobilized microalga Chlorella vulgaris. J. Ind. Eng. Chem. 49, 145-151.
Nützmann H-W., et al., 2011. Bacteria- induced natural product formation in the
e-
fungus Aspergillus nidulans requires Saga/Ada- mediated histone acetylation. P. Natl.
Acad. Sci. USA. 108, 14282-14287.
Pr

Netzker T., et al., 2018. Microbial interactions trigger the production of antibiotics.
Curr. Opin. Microbiol. 45, 117-123.
Ng W-L., Bassler BL., 2009. Bacterial quorum-sensing network architectures. Annu.
al

Rev. Genet. 43, 197-222.


Niu F-X., et al., 2018. Enhancing production of pinene in Escherichia coli by using a
rn

combination of tolerance, evolution, and modular co-culture engineering. Front.


Microbiol. 9.
u

Nonaka K., et al., 2011. Enhancement of metabolites productivity of Penicillium


pinophilum FKI-5653, by co-culture with Trichoderma harzianum FKI-5655. J.
Jo

Antibiot. 64, 769.


Oh D-C., et al., 2007. Induced production of emericellamides A and B from the
marine-derived fungus Emericella sp. in competing co-culture. J. Nat. Prod. 70,
515-520.
Oliva B., et al., 2001. Antimicrobial properties and mode of action of the pyrrothine
holomycin. Antimicrob Agents Chemother. 45,532-9.
Olson DG., et al., 2012. Recent progress in consolidated bioprocessing. Curr. Opin.
Biotech. 23, 396-405.
Osundeko O., et al., 2019. Promises and challenges of growing microalgae in
wastewater. Water Conservation, Recycling and Reuse: Issues and Challenges:
Springer; 29-53.
Pérez J., et al., 2011. Myxococcus xanthus induces actinorhodin overproduction and
aerial mycelium formation by Streptomyces coelicolor. Microb. Biotechnol. 4,
175-183.
Journal Pre-proof

Panda BP., Javed S., Ali M., 2010. Optimization of fermentation parameters for higher
lovastatin production in red mold rice through co-culture of Monascus purpureus and
Monascus ruber. Food. Bioprocess. Tech. 3, 373-378.
Papone T., et al., 2016. Producing of microbial oil by mixed culture of microalgae and
oleaginous yeast using sugarcane molasses as carbon substrate. J. Clean. Energy.
Technol. 4, 253-256.
Park J., et al., 2011. Microdroplet-enabled highly parallel co-cultivation of microbial
communities. Plos. One. 6, e17019.
Patle S., Lal B., 2007. Ethanol production from hydrolysed agricultural wastes using
mixed culture of Zymomonas mobilis and Candida tropicalis. Biotechnol. Lett. 29,
1839-1843.
Peterson R., Nevalainen H., 2012. Trichoderma reesei RUT-C30–thirty years of strain
improvement. Microbiology. 158, 58-68.

f
Rateb ME., et al., 2013. Induction of diverse secondary metabolites in Aspergillus

oo
fumigatus by microbial co-culture. RSC. Adv. 3, 14444-14450.
Ren H-Y., et al., 2015. Hydrogen and lipid production from starch wastewater by
pr
co-culture of anaerobic sludge and oleaginous microalgae with simultaneous COD,
nitrogen and phosphorus removal. Water. Res. 85, 404-412.
Rodríguez-Bustamante E., et al., 2005. Bioconversion of lutein using a microbial
e-
mixture—maximizing the production of tobacco aroma compounds by manip ulation
of culture medium. Appl. Microbiol. Biot. 68, 174-182.
Pr

Roell GW., et al., 2019. Engineering microbial consortia by division of labor. Microb.
Cell. Fact. 18:35.
Saratale R., et al., 2010. Decolorization and biodegradation of reactive dyes and dye
al

wastewater by a developed bacterial consortium. Biodegradation. 21, 999-1015.


Saratale R., et al., 2009. Enhanced decolorization and biodegradation of textile azo
rn

dye Scarlet R by using developed microbial consortium- GR. Bioresource. Technol.


100. 2493-2500.
u

Saratale RG., et al., 2011. Bacterial decolorization and degradation of azo dyes: a
review. J. Taiwan. Inst. Chem. E. 42, 138-157.
Jo

Sathiyanarayanan G., et al., 2011. Metal removal and reduction potential of an


exopolysaccharide produced by Arctic psychrotrophic bacterium Pseudomonas sp.
PAMC 28620. RSC. Adv. 6, 96870-96881.
Schroeckh V., et al., 2009. Intimate bacterial–fungal interaction triggers biosynthesis
of archetypal polyketides in Aspergillus nidulans. P. Natl. Acad. Sci. USA. 106,
14558-14563.
Senan RC., Abraham TE., Bioremediation of textile azo dyes by aerobic bacterial
consortium aerobic degradation of selected azo dyes by bacterial consortium.
Biodegradation. 15, 275-280.
Sengupta S., et al., 2015. Metabolic engineering of a novel muconic acid biosynthesis
pathway via 4-hydroxybenzoic acid in Escherichia coli. Appl. Environ. Microbiol. 81,
8037-8043.
Senthilvelan T., et al., 2014. Biodegradation of phenol by mixed microbial culture: an
eco-friendly approach for the pollution reduction. Clean. Technol. Envir. 16, 113-126.
Journal Pre-proof

Sgobba E., et al., 2018. Synthetic Escherichia coli-Corynebacterium glutamicum


consortia for l- lysine production from starch and sucrose. Bioresource. Technol. 260,
302-310.
Shafique M., Jawaid A., Rehman Y., 2017. As (V) reduction, As (III) oxidation, and
Cr (VI) reduction by multi- metal-resistant Bacillus subtilis, Bacillus safensis, and
Bacillus cereus species isolated from wastewater treatment plant. Geomicrobiol. J. 34,
687-694.
Shahab RL. 2019. Labor division in engineered cross-kingdom consortia:
Consolidated bioprocessing of lignocellulosic biomass to carboxylic acids. PhD thesis
EPFL, Switzerland.
Shahab RL., et al., 2018. Consolidated bioprocessing of lignocellulosic biomass to
lactic acid by a synthetic fungal‐ bacterial consortium. Biotechnol. Bioeng. 115,
1207-1215.

f
Shahab RL., et al., 2020. Engineering of ecological niches to create stable artificial

oo
consortia for complex biotransformations. Curr Opin Biotechnol. 62,129-136.
Shanmugam BK., et al., 2017. Metabolic pathway and role of individual species in the

Chemosphere. 188, 81-89.


pr
bacterial consortium for biodegradation of azo dye: a biocalorimetric investigation.

Shong J., Diaz MRJ., Collins CH., 2012. Towards synthetic microbial consortia for
e-
bioprocessing. Curr. Opin. Biotech. 23, 798-802.
Simarro R., et al., 2011. Optimisation of key abiotic factors of PAH (naphthalene,
Pr

phenanthrene and anthracene) biodegradation process by a bacterial consortium.


Water. Air. Soil. Poll. 217, 365-374.
Singh N., et al., 2018. Enhanced cellulosic ethanol production via consolidated
al

bioprocessing by Clostridium thermocellum ATCC 31924☆. Bioresource. Technol.


250, 860-867.
rn

Solís M., et al., 2012. Microbial decolouration of azo dyes: a review. Process.
Biochem. 47, 1723-1748.
u

Song H., et al., 2014. Synthetic microbial consortia: from systematic analysis to
construction and applications. Chem. Soc. Rev. 43, 6954-6981.
Jo

Spohn M., et al., 2014. Overproduction of ristomycin A by activation of a silent gene


cluster in Amycolatopsis japonicum MG417-CF17. Antimicrob. Agents. Ch. 58,
6185-6196.
Stams AJ., Plugge CM., 2009. Electron transfer in syntrophic communities of
anaerobic bacteria and archaea. Nat. Rev. Microbiol. 7, 568.
Stiemsma LT., Michels KB., 2018. The role of the microbiome in the developmental
origins of health and disease. Pediatrics. 141, e20172437.
Stiles WA., et al., 2018. Using microalgae in the circular economy to valorise
anaerobic digestate: challenges and opportunities. Bioresource. Technol. 267,
732-742.
Sun Q., et al., 2010. Statistical optimization of biohydrogen production from sucrose
by a co-culture of Clostridium acidisoli and Rhodobacter sphaeroides. Int. J.
Hydrogen. Energ. 35, 4076-4084.
Suriyachai N., et al., 2013. Optimized simultaneous saccharification and
Journal Pre-proof

co-fermentation of rice straw for ethanol production by Saccharomyces cerevisiae and


Scheffersomyces stipitis co-culture using design of experiments. Bioresource. Technol.
142, 171-178.
Tacconelli E., et al., 2018. Discovery, research, and development of new antibiotics:
the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet. Infect.
Dis. 18, 318-327.
Thoendel M., Horswill AR., 2010. Biosynthesis of peptide signals in gram-positive
bacteria. Adv. Appl. Microbiol. 91-112.
Thuan NH., et al., 2018. Engineering co-culture system for production of apigetrin in
Escherichia coli. J. Ind. Microbiol. Biot. 45, 175-185.
Thuan NH., et al., 2018. Escherichia coli modular coculture system for resveratrol
glucosides production. World. J. Microb. Biot. 34, 75.
Toyofuku M., 2019. Bacterial communication through membrane vesicles. Biosci.

f
Biotech. Bioch. 1-7.

oo
Turnbaugh PJ., et al., 2007. The human microbiome project. Nature. 449, 804.
Unnithan VV., Unc A., Smith GB., 2014. Mini- review: a priori considerations for

Algal. Res. 4, 35-40.


pr
bacteria–algae interactions in algal biofuel systems receiving municipal wastewaters.

Valdez- Vazquez I., et al., 2015. Hydrogen and butanol production from native wheat
e-
straw by synthetic microbial consortia integrated by species of Enterococcus and
Clostridium. Fuel. 159, 214-222.
Pr

Van Zyl WH., den Haan R., la Grange DC., 2013. Developing cellulolytic organisms
for consolidated bioprocessing of lignocellulosics. K. Gupta, M.G. Tuohy (Eds.),
Biofuel technologies recent developments, Springer, Berlin. 189-220.
al

Verma P., Madamwar D., 2002. Production of ligninolytic enzymes for dye
decolorization by cocultivation of white-rot fungi Pleurotus ostreatus and
rn

Phanerochaete chrysosporium under solid-state fermentation. Appl. Biochem.


Biotech. 102, 109-118.
u

Villegas LB., et al., 2014. Microbial consortia, a viable alternative for cleanup of
contaminated soils. In: Bioremediation in Latin America. Springer. 135-148.
Jo

Wang C., et al., 2019. A novel microbe consortium, nano-visible light photocatalyst
and microcapsule system to degrade PAHs. Chem Eng J. 359,1065-1074.
Walker D., et al., 2019. Surface water pollution. Environ. Sci. Pollut. R. 261-292.
Wang E-X., et al., 2016. Reorganization of a synthetic microbial consortium for
one-step vitamin C fermentation. Microb. Cell. Fact. 15, 21.
Wang E-X., et al., 2019. Synthetic cell–cell communication in a three-species
consortium for one-step vitamin C fermentation. Biotechnol. Lett. 1-11.
Wang J-p., et al., 2013. Induced production of depsipeptides by co-culturing Fusarium
tricinctum and Fusarium begoniae. Tetrahedron. Lett. 54, 2492-2496.
Wang J., et al., 2018. A novel process for cadaverine bio-production using a
consortium of two engineered Escherichia coli. Front. Microbiol. 9.
Wang S., et al., 2018. 4-Hydroxybenzoic acid—a versatile platform intermediate for
value-added compounds. Appl. Microbiol. Biot. 102, 3561-3571.
Wang X., et al., 2019. Engineering a microbial consortium based whole-cell system
Journal Pre-proof

for efficient production of glutarate from L-lysine. Front. Microbiol. 10, 341.
Wen Z., et al., 2017. Enhanced solvent production by metabolic engineering of a
twin-clostridial consortium. Metab. Eng. 39, 38-48.
Wen Z., et al., 2014. Artificial symbiosis for acetone-butanol-ethanol (ABE)
fermentation from alkali extracted deshelled corn cobs by co-culture of Clostridium
beijerinckii and Clostridium cellulovorans. Microb. Cell. Fact. 13, 92.
Willrodt C., et al., 2015. Coupling limonene formation and oxyfunctionalization by
mixed‐ culture resting cell fermentation. Biotechnol. Bioeng. 112, 1738-1750.
Wondraczek L., et al., 2019. Artificial microbial arenas: Materials for observing and
manipulating microbial consortia. Adv. Mater. 1900284.
Wrede D., et al., 2014. Co-cultivation of fungal and microalgal cells as an efficient
system for harvesting microalgal cells, lipid production and wastewater treatment.
Plos. One. 9, e113497.

f
Xin F., et al., 2017. Strategies for improved isopropanol–butanol production by a

oo
Clostridium strain from glucose and hemicellulose through consolidated
bioprocessing. Biotechnol. Biofuels. 10, 118.
pr
Xu G., et al., 2007. Mineralization of chlorpyrifos by co-culture of Serratia and
Trichosporon spp. Biotechnol. Lett. 29, 1469-1473.
Yamada R., Hasunuma T., Kondo A., 2013. Endowing non-cellulolytic
e-
microorganisms with cellulolytic activity aiming for consolidated bioprocessing.
Biotechnol. Adv. 31, 754-763.
Pr

Yang X., Xu M., Yang S-T., 2015. Metabolic and process engineering of Clostridium
cellulovorans for biofuel production from cellulose. Metab. Eng. 32, 39-48.
You L., et al., 2004. Programmed population control by cell–cell communication and
al

regulated killing. Nature. 428, 868.


Zhang H., et al., 2015. Engineering E. coli–E. coli cocultures for production of
rn

muconic acid from glycerol. Microb. Cell. Fact. 14, 134.


Zhang H., et al., 2015. Engineering Escherichia coli coculture systems for the
u

production of biochemical products. P. Natl. Acad. Sci. USA. 112, 8266-8271.


Zhang H., Stephanopoulos G., et al., 2016. Co‐ culture engineering for microbial
Jo

biosynthesis of 3‐ amino‐ benzoic acid in Escherichia coli. Biotech. J. 11, 981-987.


Zhang H., Wang X., 2016. Modular co-culture engineering, a new approach for
metabolic engineering. Metab. Eng. 37, 114-121.
Zhang W., et al., 2017. Production of naringenin from D‐ xylose with co‐ culture of E.
coli and S. cerevisiae. Eng. Life. Sci. 17, 1021-1029.
Zhang X-Z., et al., 2011. One-step production of lactate from cellulose as the sole
carbon source without any other organic nutrient by recombinant cellulolytic Bacillus
subtilis. Metab. Eng. 13, 364-372.
Zhou K., et al., 2015. Distributing a metabolic pathway among a microbial
consortium enhances production of natural products. Nat. Biotechnol. 33, 377.
Zhou Y., et al., 2019. Establishing microbial co‐ cultures for 3‐ hydroxybenzoic acid
biosynthesis on glycerol. Eng. Life. Sci. 19, 389-395.
Zomorrodi AR., Segre D., 2016. Synthetic ecology of microbes: mathematical models
and applications. J. Mol. Biol. 428, 837-861.
Journal Pre-proof

Zuck KM., Shipley S., Newman DJ., 2011. Induced production of N-formyl alkaloids
from Aspergillus fumigatus by co-culture with Streptomyces peucetius. J. Nat. Prod.
74, 1653-1657.
Zuroff TR., Xiques SB., Curtis WR., 2013. Consortia- mediated bioprocessing of
cellulose to ethanol with a symbiotic Clostridium phytofermentans/yeast co-culture.
Biotechnol. Biofuels. 6, 59.

f
oo
pr
e-
Pr
al
u rn
Jo
Journal Pre-proof

Table 1. Recent studies on mixed cultures identifying novel secondary metabolites.


Production in
Microorganisms Novel product Activity Reference
monoculture
Alternaria tenuissima & Stemphyperylenol, alterperylenol Antifungal activity Yes (Chagas et al. , 2013)
Nigrospora sphaerica
Aspergillus fumigates & Fumicyclines A, B Antibiotic activity
o f No (König et al. , 2013)
Streptomyces rapamycinicus
r o
A. fumigatus & Ergosterol, diketopiperazine

- p
Antibacterial and No (Rateb et al. , 2013)

e
S. bullii alkaloids antiprotozoal
Fusarium tricinctum &
Fusarium begoniae
Subenniatins A and B

P rAntibiotic activity No (Wang et al. , 2013)

al
Trichophyton rubrum & 4″-hydroxysulfoxy-2,2″ N/A No (Bertrand et al. ,

rn
Bionectria ochroleuca -dimethylthielavin P 2014)
Aspergillus nidulans & Orsellinic acid, lecanoric acid N/A No (Schroeckh et al. ,
Collection of 58 actinomycetes

o u
F-9775A, F-9775B (1) (1) Cathepsin K inhibitors 2009)
Streptomyces clavuligerus &
Staphylococcus aureus
Salinispora arenicola &
J
Holomycin (2)

Emericellamides A and B
(2) Antimicrobial activity

Antibacterial activities
No

Yes
(Charusanti et al. ,
2012)
(Oh et al. , 2007)
Emericella sp.
Penicillium pinophilum & Secopenicillide C N/A No (Nonaka et al. , 2011)
Trichoderma harzianum Penicillide, MC-141 and Yes
Stromemycin
Journal Pre-proof

Rhodococcus fascians & Rhodostreptomycin A and B Antibiotic activities No (Kurosawa et al. ,


Streptomyces padanus 2008)
A. fumigates & Fumiformamide (1) (1) Inactive No (Zuck et al. , 2011)
Streptomyces peucetius N,N’-((1Z,3Z)-1,4-bis(4-methoxyp (2) Cytotoxicit
henyl)buta-1,3-diene-2,3-diyl)
diformamide (2)

o f
r o
- p
r e
l P
n a
u r
J o
Journal Pre-proof

Table 2. Summary of recent progress in valuable compound production applying modular co-culture engineering.

Improvement compared to
Co-culture Product Reference
monoculture/other method*

Bacteria-Bacteria E. coli-E. coli Muconic acid (MA)


o
19-fold titer improvement
f (Zhang, Li, 2015a)
E. coli-E. coli Pyranoanthocyanins
r o
more stable than plant extraction (Akdemir et al. , 2019)

E. coli-E. coli
4-hydroxybenzoic acid
(4HB)
- p
8.6-fold titer improvement (Zhang et al. , 2015b)

E. coli-E. coli
3-hydroxybenzoic acid
r e
5.3-fold titer improvement (Zhou et al. , 2019)

P
(3HB)

E. coli-E. coli Cadaverine


al 3-fold and 2.7-fold higher in cadaverine (Wang et al. , 2018a)

rn
production and productivity
E. coli-E. coli Apigetrin 2.1-fold titer improvement (Thuan et al. , 2018a)
E. coli-E. coli
o u
Caffeoylmalic acid 5-fold titer improvement (Li et al. , 2018)
E. coli-E. coli
E. coli-E. coli
J Salidroside
Resveratrol glucosides
Over 20-fold titer improvement
2.9-fold titer improvement
(Liu et al. , 2018a)
(Thuan et al. , 2018b)
E. coli-E. coli Naringenin 1.5-fold titer improvemen (Ganesan et al. , 2017)
Bisdemethoxycurcumin 6.28 mg/L in titer, BDMC first de novo
E. coli-E. coli (Fang et al. , 2018)
(BDMC) production from glucose*
E. coli-E. coli Caffeyl alcohol 12-fold improvement (Chen et al. , 2017)
Journal Pre-proof

E. coli-E. coli Muconic acid (MA) 14-fold titer improvement (Zhang et al. , 2015a)
3-amino-benzoic acid (Zhang and
E. coli-E. coli 15-fold titer improvement
(3AB) Stephanopoulos, 2016)
22.6 mg/L titer, first de novo production (Camacho-Zaragoza et
E. coli-E. coli Resveratrol
from glycerol al. , 2016)
E. coli-E. coli Perillyl acetate (POHAc)
o f
3.3-fold titer and ~34-fold productivity
(Willrodt et al. , 2015)

ro
improvement
E. coli-E. coli Flavonoids 970-fold titer improvement (Jones et al. , 2016)
E. coli-E. coli Pinene
-p
1.9-fold titer improvement

e
(Niuet al. , 2018)

E. coli-E. coli
Salicylate
2-O-β-d-glucoside

P r2.5 g/L titer, higher than the


monoculture
(Ahmadi et al. , 2016)

al
89.7% of the theoretical yield,
G. oxydans-K. 2-keto-L-gulonic acid
comparable to the conventional (Wang et al. , 2016)
vulgare (2-KGA)

n
two-step fermentation
P. putida-P. putida
r
2-hydroxybiphenyl

u
(2HBP)
Up to 50% 2HBP formation (Martínez et al. , 2016)

oneidensis
J
E. coli-B. subtilis-S.
o Electricity
0.28 g of glucose was converted to
electricity (~550 mV) for more than 15
days
(Liu et al. , 2017)

E. coli-E. coli-E. coli Rosmarinic acid 38-fold titer improvement (Li et al. , 2019)
E. coli-E. coli-E. 9.5 mg/L of titer, the first report in
Callistephin (Jones et al. , 2017)
coli- E. coli non-plant host*
Eukaryote- P. pastoris- Monacolin J, 55% improvement of monacolin J and
(Liu et al. , 2018b)
Eukaryote P. pastoris Lovastatin 71%improvement of lovastatin from
Journal Pre-proof

methanol
Bacteria-Eukaryote 33 mg/L vs non-detected by
E. coli-S.cerevisiae Oxygenated taxanes (Zhou et al., 2015)
monoculture
21.16 mg/L of titer, 8-fold titer
E. coli-S.cerevisiae Naringenin (Zhang et al. , 2017)

f
improvement
*No report using mono-culture de novo synthesis.

o o
p r
e -
P r
a l
r n
o u
J
Table 3. An overview of microbial consortia mediated consolidated bioprocessing (CBP) systems for bulk chemical production.
Journal Pre-proof

Product Co-culture strains Substrate Achievement Reference


Lactic acid T. reesei & Non-detoxified steam‐ 19.8 g/L, 85.2% of the
(Shahab et al. , 2018)
Lactobacilli sp. pretreated beech wood theoretical maximum
L-lysine, 12.3  mM of L-lysine, 6.8mM

f
E. coli &
cadaverine and Starch of cadaverine and 3.4 mM of (Sgobba et al. , 2018)

o
C. glutamicum
L-pipecolic acid L-pipecolic acid

ro
Ethanol T. reesei &
(Brethauer and Studer,

p
S. cerevisiae & wheat straw ~ 9 g/L, 67% yield
2014)
S. stipitis
C. phytofermentans & S.
e -
cerevisiae
α-cellulose

P r 22 g/L
15.2 g/L, 99% of the
(Zuroff et al. , 2013)
(Suriyachai et al. ,
S. cerevisiae & S. stipitis
Z. mobilis &
a l
Pretreated rice straw
Enzymatically hydrolysed
theoretical maximum 2013)

n 52 g/L, 97.7% yield (Patle and Lal, 2007)

ur
C. tropicalis lignocellulosic biomass
Isobutanol T. reesei & E. coli Corn Stover 1.88 g/L, 62% of the
(Minty, Singer, 2013)
ABE
C. cellulovorans &
C. beijerinckii J o Corn cobs (alkali extracted)
theoretical maximum
22.1 g/L solvents (4.25 g/L
acetone, 11.5 g/L butanol and (Wen et al. , 2017)
6.37 g/L ethanol)
23.3 g/L solvents (3.7 g/L
C. beijerinckii & Biologically treated wheat (Valdez-Vazquez,
ethanol, 14.2 g/L butanol, and
C. cellulovorans straw Pérez-Rangel, 2015)
5.4 g/L acetone)
C. beijerinckii & Alkali extracted deshelled 11.8 g/L solvents (2.64 g/L
(Wen et al. , 2014)
C. cellulovorans corn cobs acetone, 8.30 g/L butanol and
Journal Pre-proof

0.87 g/L ethanol)


Microbial lipid Chlorella sp. KKU-S2 & (Papone, Kookhunthod,
Sugarcane molasses 0.33g/L
T.globosa YU5/2 2016)

o f
r o
- p
r e
l P
n a
u r
J o
Journal Pre-proof

Figure legends

Figure 1. A brief timeline of some of the key milestones in microbial consortia

development.

Figure 2. The interaction mechanism models of microbial consortia. (a) Quorum

sensing in Gram-negative bacteria consortium. In typical Gram-negative LuxIR

f
circuits, the LuxI-type protein catalyzes the synthesis of an N-acylhomoserine

oo
lactones (AHLs) autoinducer (pentagons). Once AHLs reach a threshold level in the

pr
environment, they will activate the transcriptional regulator proteins of LuxR family.
e-
The LuxR/AHL complex can activate the expression of multiple target genes,
Pr

including those required for AHL synthesis. (b) Quorum sensing in Gram-positive

bacteria consortium. Autoinducing peptides (AIPs) are actively exported from the cell
al

and bind to the sensor domain of an extracellular receptor-HK. Once a critical


rn

concentration of AIP is reached in the environment, it will be phosphorylated by the


u

receptor-HK and imported into the cell, the phosphorylated AIP will bind on the target
Jo

DNA to regulate its transcription. (c) A model of signal molecule transport using

membrane vesicles (MVs). Gram-negative bacteria pinches off of a producing cell

from cell surface, which forms a spherical container. The MVs fuse with a receptor

cell, delivering hydrophobic signal molecules, subsequently activate the transcription

of target genes. (d) The model of physical contact. The typical case is the intimate

contact between A. nidulans and actinomycetes, which led to an increased acetylation

of histone H3 catalyzed by the Saga/Ada complex. (e) Gene level changes in microbial
Journal Pre-proof

consortia. The mixed culture can cause a serial changes in gene expression in the

manner of gene loss, histone modification, and horizontal gene transfer.

Figure 3. Schematic illustrations of different consolidated bioprocessing engineering

strategies. (a) The “native” Lignocellulolytic Strategy means recombinant solvents

synthesis pathway in the plant biomass degrading microorganism, while (b)

f
Recombinant Lignocellulolytic Strategy means expression these lignocelluloses

oo
degrading enzymes in solvents producing microorganisms. (c) Consolidated

pr
bioprocessing Strategy for lignocellulose biorefinery using microbial consortia, where
e-
biomass is first degraded into fermentable carbon source catalyzed by degrading
Pr

enzyme released from cellulosic microorganism, subsequently, the produced

fermentation carbon source will be utilized by non-cellulosic microorganism to


al

produce various valuable chemicals. Main enzymes involved in in cellulose and


rn

hemicelluloses degradation are illustrated with in different symbols.


u
Jo

Figure 4. Symbiotic relationship between microalgae and bacteria/fungi during

wastewater treatment. Microalgae can sustain and drive an engineered consortium

with other microorganisms through metabolite exchange, while microalgae can

release organic compounds and O 2 during photosynthesis for bacteria/fungi, In return,

bacteria/fungi will provide microalgae CO 2 and growth promoting factors. The

harvested biomass of microalgae and co-cultured microorganism can further be used

as feedstock for biofuel or biochemical production.


Journal Pre-proof

f
oo
pr
e-
Pr
al
rn
u
Jo
Journal Pre-proof

Highlights:

 This review comprehensively summarized the distinguished advantages of

microbial consortia against monocultures and classified the functional

applications of microbial consortia in multiple fields.

 Comprehensively discussed the unrevealed interaction mechanisms behind the

microbial consortia from the perspectives of physical contact, chemical

f
communication and gene mutation.

oo
 Providing guidance and experience in the construction of artificial microbial

consortia systems. pr
e-
 The outstanding challenges and future directions to advance artificial microbial
Pr

consortia development were highlighted, especially on spatial organization

schemes design and supporting materials introduction.


al
u rn
Jo

You might also like