ArticlePDF AvailableLiterature Review

Synthetic Biology Goes Cell-Free

Springer Nature
BMC Biology
Authors:
  • Liberum Biotech

Abstract and Figures

Cell-free systems (CFS) have recently evolved into key platforms for synthetic biology applications. Many synthetic biology tools have traditionally relied on cell-based systems, and while their adoption has shown great progress, the constraints inherent to the use of cellular hosts have limited their reach and scope. Cell-free systems, which can be thought of as programmable liquids, have removed many of these complexities and have brought about exciting opportunities for rational design and manipulation of biological systems. Here we review how these simple and accessible enzymatic systems are poised to accelerate the rate of advancement in synthetic biology and, more broadly, biotechnology.
This content is subject to copyright. Terms and conditions apply.
R E V I E W Open Access
Synthetic Biology Goes Cell-Free
Aidan Tinafar, Katariina Jaenes and Keith Pardee
*
Abstract
Cell-free systems (CFS) have recently evolved into key
platforms for synthetic biology applications. Many
synthetic biology tools have traditionally relied on
cell-based systems, and while their adoption has
shown great progress, the constraints inherent to the
use of cellular hosts have limited their reach and
scope. Cell-free systems, which can be thought of as
programmable liquids, have removed many of these
complexities and have brought about exciting
opportunities for rational design and manipulation of
biological systems. Here we review how these simple
and accessible enzymatic systems are poised to
accelerate the rate of advancement in synthetic
biology and, more broadly, biotechnology.
Moving towards a new bioengineering platform
Since its emergence, the field of synthetic biology has
given rise to the development of many technologies that
are implemented using the whole cell [1]. These have in-
cluded biosensors capable of detecting broad ranges of
analytes [25], systems that can count [6] or perform
complex logic [710], engines for the bioproduction of
valuable commodities [1114], gene-circuit-driven chas-
sis for regenerative medicine [15,16], and engineered
CAR-T cells [17]. Such technologies are on track to
transform many aspects of modern life, yet their require-
ment for a cellular host has limited their reach and
scope. For example, concerns over biosafety have re-
stricted the use of engineered cells, and the systems they
host, largely to laboratory settings. The self-replicability
of cell-based systems carries the risk of escapeor con-
tamination that could impact human health, food secur-
ity, and the environment. While the development of
safeguards to prevent these types of events is an active
area of research [18,19], failure-free implementation of
such systems is not a trivial task.
Another substantial limitation of cell-based synthetic
biology is the requirement for laborious genetic encod-
ing of its design features into a living cell, which can
limit its functionality and significantly slow down de-
signbuildtest cycles. In cell-based systems, genetic in-
structions often need to be assembled into a vector,
imported into the cell, and maintained by using a select-
able marker or by genomic integration. Only then can
the instructions be evaluated. Furthermore, designs must
be iteratively tested to minimize cross-talk with en-
dogenous molecular programs while balancing between
the metabolic burden on the cellular host and the de-
sired outcome.
Cell-free systems offer a means to circumvent many of
theselimitations.Theywereoriginallyconceivedastoolsto
facilitate in vitro protein synthesis and consist of molecular
machinery extracted from cells. They typically contain en-
zymes necessary for transcription and translation, and ac-
cordingly are able to perform the fundamental processes of
the central dogma (DNARNAprotein) independent of
a cell. These systems can be derived from eukaryotes (e.g.,
vertebrates, plants, insects, fungi) [2027] or prokaryotes
(e.g., Escherichia coli,Vibrio natriegens,Bacillus subtilis)
[2843]andmaybepreparedaseitherpurifiedcompo-
nents [36,44] or semi-processed cellular extracts [38]. CFS
can be made sterile via simple filtration, which provides for
a biosafe format for use outside of the lab.
The open nature of CFS means that there is no phys-
ical barrier (e.g., a cell wall) to programming and modifi-
cation. CFS can be augmented with proteins or small
molecules that improve the performance of synthetic
gene networks [45,46] or the productivity of reactions
[39,47]. More importantly, genetically encoded instruc-
tions can be added directly to CFS at desired concentra-
tions and stoichiometries using linear or circular
formats. This means that conceptual designs can go
from computational instructions to chemical synthesis
and amplification (e.g., through PCR) to CFS without
the need for selective markers or cell-based cloning
steps. Such simplicity allows for rapid prototyping of
molecular tools.
© The Author(s). 2019 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
* Correspondence: keith.pardee@utoronto.ca
Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St.,
Toronto, ON M5S 3M2, Canada
Tinafar et al. BMC Biology (2019) 17:64
https://doi.org/10.1186/s12915-019-0685-x
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Importantly, CFS can be freeze-dried, enabling room
temperature storage and distribution [46,48]. Freeze-
dried cell-free (FD-CF) systems can then be activated at
the time of need simply by adding water [46]. This fea-
ture has been used to deploy biosafe, genetically encoded
tools outside of the laboratory as diagnostics and as plat-
forms for biomanufacturing [49,50], as well as their de-
ployment in altogether new contexts, such as global
health and education.
Below we will discuss how CFS are enabling new tech-
nologies and accelerating the coming revolution in bio-
engineering, highlighting some of the most active areas
of research in the cell-free community (Fig. 1).
Development of sensors
Molecular recognition underlies almost every biological
process, including the nucleic acid base pairing that im-
parts specific syntax to the central dogma. Scientists and
engineers have long worked to usher these processes
into cell-free in vitro environments to understand and
exploit their underlying molecular mechanisms for pur-
poses such as diagnostics and detection of molecules.
One of the fruits from such efforts is the polymerase
chain reaction (PCR), which is now an indispensable tool
utilized in most molecular biology laboratories, includ-
ing those for clinical diagnostics. There is currently a
growing need for de-centralized, portable diagnostics
that can be rapidly deployed in the field, for instance
during infectious disease outbreaks or for agricultural
purposes. However, sensing technologies such as PCR
and others have largely remained confined to laborator-
ies in large urban centers due to their requirement for
specialized equipment and personnel.
The biosafe and stable nature of FD-CF systems offers
an alternative molecular venue to address the unmet
need for distributed and low-cost sensing. Here, the
transcription and translation properties of CFS can be
used to host gene circuit-based sensors that can detect
small molecules and nucleic acids with exquisite sensi-
tivity and specificity. Many of the biosensors and circuits
that have been developed for cell-based applications can
be operated in the cell-free environment. These include,
among others, many classic switches (e.g., TetO- and
LacI-based systems), logic gates, negative feedback loops,
transcriptional cascades [37,41,5356] and ring oscilla-
tors [57]. This cross-compatibility between CFS and cell-
based systems has also been exploited for rapid proto-
typing of regulatory elements that can be brought back
to the cell-based environment.
FD-CF systems do not require a temperature-
controlled environment and cold-chain logistics intrinsic
to many other diagnostic approaches, as they remain
Fig. 1 Cell-free protein expression systems and their applications. Capitalizing on their open nature, CFS can be rationally assembled to include
cell lysates, purified proteins, energy sources (e.g., ATP), amino acids, other substrates (such as modified tRNAs and membrane mimics) and RNA
or DNA (circular or linear). CFS can be applied in portable diagnostic devices [46,50] and also hold great potential for biomolecular
manufacturing [49,51]. Additionally, CFS can enable discovery of novel enzymes (e.g., through directed evolution) [52]
Tinafar et al. BMC Biology (2019) 17:64 Page 2 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
active for at least a year without refrigeration, enabling
room temperature storage and distribution [46]. This,
however, does not circumvent the challenges arising
from handling these molecular tools in liquid phasefor
instance upon their resuspension outside of the labora-
tory environment. Inspired by systems like pH paper
and lateral-flow diagnostics, we embedded FD-CF reac-
tions into porous materials (e.g., paper), demonstrating
that low-volume reactions (12μL) could readily be
achieved within this medium. Such paper-based cell-free
systems enabled the deployment of poised synthetic gene
networks outside of the laboratory in a contained and
biosafe format for the first time [46].
With this new ruggedized paper-based format, simple
sensing such as anhydrotetracycline (ATc)-inducible ex-
pression of GFP and mCherry was established [46].
However, to demonstrate the real-world potential for
this system, a sensing platform that could be rationally
designed to detect a wide range of practical analytes was
needed. This was realized with the introduction of toe-
hold switches [58], a new class of riboregulators, into
FD-CF reactions. The use of toehold switches, which
can be designed to recognize virtually any sequence of
interest, was first demonstrated in paper-based FD-CF
reactions for the detection of genes responsible for anti-
biotic resistance and strain-specific detection of the
Ebola virus [46]. While the demonstration of this sensing
capacity in a portable format was exciting, the system
lacked the sensitivity necessary to detect RNA levels
generally present in patient samples.
This sensitivity challenge was addressed by placing
an isothermal amplification step (e.g., NASBA) in the
workflow upstream of the cell-free reaction. This im-
proved the threshold of detection by orders of magni-
tude (10
6
). Since isothermal amplification is a primer-
directed process, combination with toehold-based
sensing results in two sequence-specific checkpoints.
An opportunity to test out the improved system pre-
sented itself in early 2016 when the outbreak of the
mosquito-borne Zika virus was reported in Brazil.
With the improved embodiment, FD-CF toehold sen-
sors could detect all global strains of the Zika virus
at clinically relevant concentrations (down to 2.8 fem-
tomolar) from viremic plasma [50]. Moreover, pow-
ered by the first CRISPR-based system in an in vitro
diagnostic system, viral genotypes could be distin-
guished with single base pair resolution (e.g., Ameri-
can vs African Zika strains). Most recently the Collins
group extended these concepts in a tour de force ef-
fort that demonstrated quantitative detection of ten
gut bacterial species from patient samples [59]. This
work demonstrated detection at clinically relevant
concentrations with sensing performance that mapped
well with parallel measurements done with RT-qPCR.
It also showcased the ability to detect a toxin-related
sequence for the diagnosis of Clostridium difficile
infections.
Following the initial work outlining the potential for
the FD-CF format, a body of work ensued demonstrating
many biosensing applications and improvements on FD-
CF preparations. In one of the earliest examples, Duyen
et al. developed a sensor for the detection of antibiotic
contamination based on protein synthesis inhibition
caused by some antibiotics [60]. The Freemont group
applied their expertise in CFS to develop sensors for the
detection of Pseudomonas aeruginosa in cystic fibrosis
patient samples [61], demonstrating that the quorum-
sensing molecule from P. aeruginosa (3-oxo-C12-HSL)
could be detected down to low nanomolar concentra-
tions. Another novel approach used CFS to express engi-
neered protein fusions containing nuclear receptor
ligand binding domains for the detection of endocrine-
disrupting compounds [62,63]. This work showcased
sensitivity in the nanomolar range, and, interestingly,
demonstrated that CFS could operate in the presence of
contaminants in environmental and clinical samples. In
another example, detection of mercury contamination
using the mercury(II)-responsive transcriptional repres-
sor MerR was accomplished [45] (Fig. 2).
Manufacturing of therapeutics
Another active area in CFS research is the biomanufac-
turing of therapeutics and other protein-based reagents.
Natural biological systems have evolved a remarkable
capacity to synthesize a variety of molecules ranging
from metabolites to biopolymers. Cell-free protein ex-
pression systems allow the incorporation of such reac-
tions into a highly controlled process that allows
production of molecules as needed and in the field. Our
primary focus here will be on a subset of biopolymers,
namely therapeutic proteins. The ongoing work in this
field rests on decades of research that have led to the
productive and practical systems currently available [28,
29,3638,40]. Recent advances in high-throughput
preparation techniques [40,45] and in the development
of systems that can use more economical energy sources
[64,65] have made CFS highly accessible. Meanwhile,
significant strides are being made towards resolving vari-
ous protein folding issues and shortcomings in post-
translational modifications [66] associated with trad-
itional CFS. Recent advances have showcased the poten-
tial for scaling up cell-free reactions, with some having
demonstrated reaction volumes reaching 100 liters [67,
68] to 1000 liters [69]. Cell-free expression has been
used as a platform for the production of a wide range of
potential therapeutics, some of which have been summa-
rized in Table 1. A number of these products have been
validated in animal models [49,76].
Tinafar et al. BMC Biology (2019) 17:64 Page 3 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Two primary modes of CFS have been pursued. The
first, used by commercial efforts such as Sutro [94], fo-
cuses on large, centralized production. This approach le-
verages the advantages of synthesis outside of the cell
for biomanufacturing. For these applications, CFS not
only allow for rapid production, but also significantly
speed up the drug development process [95]. Remark-
ably, Sutro has reportedly increased their cell-free pro-
duction to an incredible 1000 liters [69], showcasing the
scalability of centralized cell-free production. The sec-
ond mode uses FD-CF systems to de-centralize bioma-
nufacturing capacity for small-batch production of
therapeutics, with applications in global health and
emergency response [49,73,96,97]. Using this mode of
production, we have recently demonstrated the proof-of-
concept capacity to manufacture over 50 therapeutics
and lab reagents, including proteins (e.g., vaccines, anti-
bodies, and antimicrobial peptides) and small molecules
[49], with applications outside of the laboratory setting.
Cell-free biomanufacturing is particularly well-suited
for vaccine production due to its potential for rapid
scale-up in response to public health emergencies. Suc-
cessful cell-free expression of a number of recombinant
vaccines (e.g., botulinum, diphtheria, anthrax) has been
demonstrated [49,8690,98], with some having been
validated in animal models, such as mice [49,90]. Con-
sidering the low dose requirements (microgram range)
for many of these therapeutics, commercialization of
CFS-derived vaccines will likely see rapid growth in the
coming years. Production of antibodies has also been an
area of focus for the cell-free community [20,49,51,
7480,99,100]. Due to their compact size and relatively
high expression levels in CFS, single-domain antibodies
have garnered particular attention and seem strategically
well-placed to serve the emerging needs in personalized
medicine, i.e., for therapeutics and diagnostics.
Antibiotic resistance has been recognized as a major
threat to global health, resulting in approximately two
million illnesses and 23,000 deaths in the US alone every
year [101]. Accordingly, cell-free production of anti-
microbial compounds, including antimicrobial peptides
and small molecule drugs, has become the focus of some
groups [49,93]. A number of labs have also demon-
strated the power of CFS to express phages [56,102
104]. The upward trend in the reported antibiotic resist-
ance cases has led to a resurgence in viewing phage
Fig. 2 Overview of the use of biosensors in CFS. The general workflow usually involves in silico design of gene circuits encoding biosensors and
reporter proteins, followed by chemical synthesis of such circuits. Meanwhile, patient or environmental samples are collected, target analytes are
extracted, and, in some cases, amplified. The gene circuits and target analytes are then added to CFS. Examples of biosensors in CFS have
included a) mercury (II) detection using the MerR repressor[45], b) viral and bacterial nucleic acid sensing using toehold switch-based sensors [46,
50,59], c) identification of P. aeruginosa infection by its quorum sensing molecule, 3-oxo-C12-HSL, using the LasRV sensor [61] and d) recognition
of an endocrine-disrupting compound by utilizing an allosterically activated fusion protein containing the ligand binding domain of a human
estrogen receptor [62,63]. Reporters (e.g., colorimetric or fluorescent) can then produced, contingent upon analyte detection, enabling clinical
diagnosis (e.g., using standard spectrophotometers)
Tinafar et al. BMC Biology (2019) 17:64 Page 4 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
therapy as a potentially viable alternative to current anti-
biotic regimens [101,105]. The use of phages has also
been evaluated as an effective treatment strategy for a
number of plant diseases, with some phages now being
commercially available for mass consumption [106].
CFS-based production of these non-traditional antimi-
crobials could play a significant role in battling the anti-
biotic resistance crisis and could also help improve food
security around the globe.
Below, we will highlight some of the areas in which
CFS have shown great potential for enhancing current
methods of therapeutics development and manufactur-
ing. These advances are rapidly transforming CFS into
an integral part of the manufacturing ecosystem.
Membrane proteins
While approximately 70% of all drugs act on membrane
proteins[107], working with these proteins is notoriously
difficult due to their enrichment in hydrophobic surfaces.
Cell-based expression of membrane proteins is often
fraught with challenges, such as toxicity caused by their
membrane incorporation or their incompatibility with the
hosts physiology [108]. Recently, cell-free approaches have
been used to tackle this challenging category of proteins,
the coding sequences of which comprise 2030% of all
known genes [107]. When compared to current cell-based
methods, CFS can be a powerful tool in the production of
solubleactivemembraneproteins[109]. The ability to
integrate steps that can tackle the challenging aspects of
membrane protein synthesis is particularly valuable. For in-
stance, previous efforts in cell-based systems have demon-
strated that membrane mimics can be successfully used to
synthesize and stabilize a wide range of membrane proteins
such as G-protein-coupled receptors [110,111], the epider-
mal growth factor receptor [71], hepatitis C virus mem-
brane proteins [112], and an ATP synthase [109,113].
These mimics include surfactants, liposomes, and nano-
discs [114116] and can be added directly to CFS co-
translationally or post-translationally. There is also evidence
suggesting that functioning single-span membrane proteins
can be synthesized simply in the presence of an oilwater
interface (e.g., through the use of emulsions) [117].
Macromolecular production
Molecular research has highlighted the importance of
proteinprotein interactions and the resulting complexes
that these interactions can generate. Whether it is for
the biophysical study of these complexes or as vehicles
for new therapeutic delivery (e.g., virus-like scaffolds for
vaccines), there is a growing need for developing robust
tools aimed at synthesis of such complexes. As in the
case of membrane proteins, CFS have also demonstrated
higher yields, compared to in vivo strategies, in the pro-
duction of macromolecular assemblies such as virus-like
particles (VLPs) [109]. Groundbreaking work by the
Swartz group, demonstrating the cell-free expression of
hepatitis B core antigen VLP (2 subunits) [91]inanE.
coli-based cell-free system, opened the door to other re-
searchers expressing a variety of macromolecular assem-
blies including the E. coli RNA polymerase (5 subunits)
[118] and an ATP synthase (25 subunits) [113]. Earlier
work with reticulocyte lysate had also demonstrated cell-
free expression of the human T-cell receptor (7 sub-
units) [119]. Remarkably, a number of bacteriophages
have now also been successfully expressed in CFS, in-
cluding the T4 phage, which structurally contains 1500
proteins from 50 genes [56,102104] (Fig. 3).
Non-identical subunits of a protein complex are often
referred to as hetero subunits. In some instances, such
hetero subunits require co-translation to yield active
complexes [120]. Thus, the ability of CFS to concur-
rently translate multiple mRNAs facilitates the produc-
tion of active complexes composed of a number of
different subunits [121]. Some CFS such as E. coli-based
preparations are generally not capable of producing pro-
teins that contain disulfide bonds, which are critical to
numerous pharmaceutically relevant proteins (e.g., anti-
bodies and many cytokines) [121]. However, recent ef-
forts have augmented these systems to enable the
production of complex proteins requiring multiple disul-
fide bonds [85,99,122], expanding the range of therap-
ies that can be made in CFS.
Table 1 Examples of potential therapeutics expressed in CFS to
date
Therapeutic proteins Granulocyte macrophage colony-stimulating
factor (GM-CSF) [68,70]
Erythropoietin [7072]
Cytotoxic protein onconase [73]
Antibodies [51,74,75] and antibody
fragments [49,7679]
Bispecific antibodies [80]
Antibody-drug conjugates [49,81]
Tissue-type plasminogen activator [8285]
Vaccine antigens Picornaviral capsid intermediate structures [86]
Trimeric influenza hemagglutinin head [87]
and stem [88] proteins
Trivalent vaccine based on Hc fragments of
botulinum toxins A, B, and E [89]
Anthrax protective antigen and diphtheria
toxoid [49]
Virus-like particles A B-cell lymphoma vaccine [90]
Anti-hepatitis B VLPs [91]
A virus-like nanoparticle scaffold for vaccines
and drug delivery [92]
Antimicrobials Antimicrobial peptides [49,93]
Small molecules such as violacein [49,56]
Tinafar et al. BMC Biology (2019) 17:64 Page 5 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Modification of proteins and codon tables
Effectiveness of many protein-based therapeutics hinges
upon precise control over natural or non-natural modifica-
tion of their peptide sequences. One of the most compel-
ling uses of such modifications is in the development of
antibodydrug conjugates (ADCs), which are quickly
gaining favor as a new class of therapeutics against cancer.
Classic conjugation techniques result in a heterogeneous
mixture of labeled antibodies due to their reliance on arbi-
trary conjugation to multiple amino acid side chains.
Recent studies, however, suggest that pharmacologic prop-
erties of ADCs could be improved through site-specific
conjugation. Non-natural amino acids provide an efficient
avenue for such site-specific conjugation [123]. To date, co-
translational incorporation of over 100 different non-
natural amino acids has been demonstrated in vivo [124],
allowing for a wide range of modifications [125129]. Many
of these modifications have been demonstrated in the cell-
free context for a variety of applications, including
orientation-controlled immobilization [92,98]andsite-
specific functionalization (e.g., phosphorylation [130],
PEGylation [131], or drug conjugation [81]) [132134].
CFS platforms circumvent some of the cell-based tox-
icity and permeability limitations and offer greater control
and versatility in making protein modifications [109,135].
Incorporation of non-natural amino acids in cell-based
approaches has typically relied on repurposing stop co-
dons to minimize the negative impacts of recoding on
cell-viability [109]. In a cell-free system, however, the en-
tire codon table can in theory be reprogrammed, allowing
not only for the incorporation of non-natural amino acids,
but also for the creation of entirely novel codon tables.
Taken to its extreme, the latter could help with the
protection of intellectual property. DNA sequences
could be obfuscated such that they are rendered non-
functional outside of their specialized cell-free context.
This obfuscated code would make proprietary designs
difficult to copy. Codon obfuscation could also pose ser-
ious challenges for the detection of DNA sequences that
may be employed by malevolent entities. For example,
DNA synthesis companies would have a much more dif-
ficult time screening against DNA sequences that could
be used for nefarious activities (e.g., bioterrorism). Re-
cent work has shown that the size of the codon table
can also be expanded by augmenting the four-letter gen-
etic alphabet with unnatural base pairs [136,137]. Thus,
proteins made in CFS couldat least in theoryhold an
unlimited number of non-natural amino acids.
CFS can also be employed for making naturally occur-
ring modifications to proteins. An example of these is
the grafting of sugars (i.e., glycans) referred to as glyco-
sylation. Successful production of many therapeutics is
often contingent upon highly efficient glycosylation, as
lack of proper glycosylation can reduce the efficacy and
circulation half-life of many therapeutic proteins [138].
Some CFS (e.g., insect, Chinese hamster ovary, and hu-
man K562 extract-based systems) are inherently capable
of glycosylation. However, their repertoire of glycan
structures tends to be limited to those naturally synthe-
sized by their lysatessource cell type. Additionally, gly-
cosylation in these systems often requires recapitulation
of the source cells protein trafficking mechanisms [109].
Thus, creation of synthetic glycosylation pathways in
CFS has become an area of focus in recent years [135,
139]. Success in this domain will likely serve as a key
catalyst in bringing cell-free-produced vaccines and
other therapeutics to the masses. Figure 4outlines some
of the possible protein modifications in CFS.
Directed evolution
Directed evolution is a powerful tool for aptamer and pro-
tein engineering that uses iterative rounds of mutagenesis
Fig. 3 Multi-subunit protein complex synthesis in CFS. Various groups have demonstrated the production of increasingly intricate protein
complexes. These have included the hepatitis B core antigen (HBc) VLP (2 subunits) [91], the E. coli RNA polymerase (5 subunits) [118], the human
T-cell receptor (7 subunits) [119], an ATP synthase (25 subunits) [113], and the T4 phage (1500 subunits) [102104]
Tinafar et al. BMC Biology (2019) 17:64 Page 6 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and selection to modify or tune specific bimolecular prop-
erties (e.g., an enzymes substrate activity). Utility of apta-
mers or proteins, in a given context, with respect to their
corresponding nucleotide sequences is often described as
a fitness landscape. Directed evolution provides a mas-
sively parallel method for searching through a fitness land-
scape to find optimal variants and their corresponding
genotypes [144]. This generally requires one-to-one map-
ping of phenotype to genotype. Although cells have a
built-in capacity for such mapping due to their compart-
mentalized nature, using cells to conduct directed evolu-
tion can impose limits on the size of candidate libraries
screened, and restricts the type of solvents, buffers, and
temperatures that can be sampled [145]. As a result, cell-
free directed evolution platforms have gained favor [145],
starting with the first truly cell-free systems published in
the late 90s [146,147]. More recently, connecting pheno-
type to genotype has been accomplished through artificial
compartmentalization (e.g., using emulsion, microbeads,
and liposomes) [145,148151]. Applications have in-
cluded the design and optimization of Fab antibody frag-
ments [77,152], membrane proteins [151], and, as we will
discuss below, enzyme discovery [52].
Platform for discovery
Engineered transcription and translation systems can
also greatly catalyze research in the laboratory. As previ-
ously mentioned, the absence of a cell wall means that
candidate genes can be readily screened for function. It
also means that substrates, including those difficult to
use in the cellular context, can easily be brought into
contact with enzyme libraries to screen for novel reac-
tions. Below we look at some of the recent work using
CFS as a platform for discovery.
Biosynthetic pathways
From the early days of synthetic biology, it was clear that
there was great potential for synergy with the field of
chemical synthesis. Metabolic pathways responsible for
the synthesis of valuable compounds (e.g., drugs, scents,
and flavors) were thus moved out of organisms that did
not easily lend themselves to production and into heterol-
ogous hosts, such as yeast. This microorganism-based ap-
proach has been incredibly successful and has led to the
assembly of genes from disparate sources to create engi-
neered pathways. Enzyme-based catalysis has the advan-
tage of allowing for stereo-selectivity in aqueous, low-
energy reactions (e.g., green chemistry) [153]. By lever-
aging naturally occurring pathways, it has been possible to
generate tremendous chemical diversity, as seen in isopre-
noids, from simple precursors [154]. An exemplar of this
approach is the synthesis of amorpha-4,11-diene and arte-
misinic acid, which are precursors to the anti-malarial
compound artemisinin [154157]. This process has been
repeated for other pharmaceutical pathways, enabling the
production of opioids [158,159] and taxol [160], as well
as for the generation of molecules for the energy industry
and the agriculture sector [13,161].
While microorganisms are currently a mainstay for
biomanufacturing of commodities, their use for these
purposes is nontrivial. For example, assembly, fine-
tuning, and host strain integration of the industrial-
ized pathway for the bioproduction of artemisinic acid
is estimated to have taken over 150 person-years
[162]. Another challenge to microbial bioproduction
is that a significant portion of inputs are lost to gen-
eral cellular metabolism and growth, reducing effi-
ciency of the overall process [67,134]. Cell-free
synthetic biology alleviates some of these challenges.
For instance, enzyme discoverythe identification of
enzymes that can be used for biosynthetic purposes
via CFS has proven to be effective. Enzymes and their
homologs can be rapidly screened for performance
without the cumbersome steps required for cell-based
screening (e.g., plasmid assembly and transformation).
This approach can be extended to simple prototyping
of pathways or the automated multiplexed shuffling of
complex pathway components. Unlike with cell-based
prototyping, the cell-free environment allows for the
use of enzymes encoded as linear constructs (DNA or
RNA). Substrate preference can also be evaluated
without the need for enzyme purification.
Fig. 4 Protein modifications in CFS. Possible protein modifications
include but are not limited to glycosylation, disulfide-bond
formation, acetylation [140], phosphorylation [141], and PEGylation
[131] (which may be accomplished through the use of non-natural
amino acids). Non-natural amino acids can also be used for the
conjugation of a wide range of compounds such as drugs (e.g.,
through click chemistry) [81] or fluorescent molecules [142]. Figure
adapted from Pagel et al.[143]
Tinafar et al. BMC Biology (2019) 17:64 Page 7 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
In many cases, enzymes and pathways discovered in
CFS will be brought back into cells for scale-up [163].
However, there is a growing case for using CFS dir-
ectly as the production medium. Commercial ventures
(e.g., Sutro, Greenlight) have already demonstrated
that CFS can provide economic advantages for the
production of protein and RNA products [69]. Thus,
it would be reasonable to think that a similar ap-
proach could provide a viable source of high-value
small molecules. Such systems have the advantage of
enabling bioproduction without metabolic inefficien-
cies, toxicity limitations, complex extraction steps, or
the need for integration into a host strain [67,134,
164]. Combined with the capacity for efficient proto-
typing, these systems are generating significant enthu-
siasm. The field is now beginning to focus on more
complex pathways (more than eight enzymes) and lar-
ger reaction volumes (> 100 L) [67].
Single enzyme reactions are highly simplified cell-free
systems that have been used for decades at scale for
washing (e.g., dish and laundry detergents) and for pro-
cessing food, wood pulp, and fuel [165]. Once fully oper-
ationalized, more complex cell-free enzymatic pathways
could revolutionize the chemical industry and enable
greater accessibility to bioproduction. Earlier attempts at
engineering such pathways outside of a cell were gener-
ally made by using purified components. These pathways
have included those designed for the production of
amorpha-4,11-diene [166], isoprene [167], fatty acids
[168], and nucleotides [169]. Recent work has showcased
the use of 27 purified enzymes that can work together to
convert glucose into terpenes such as limonene, pinene,
and sabinene [170]. Here, production can operate con-
tinuously for 5 days with a single addition of glucose,
with glucose conversion of greater than 95%, to generate
high product concentrations (> 15 g/L) that are well
above levels toxic to microbes. While exciting, expres-
sion and purification of each individual component for
such an approach is quite laborious.
Transitioning these metabolic pathways into CFS, where
expression of enzyme-encoding sequences could lead to the
self-assemblyofpathways,wouldbeincrediblyenabling.To
date, a number of reports have validated this approach.
Three- and six-enzyme pathways have recently been gener-
ated de novo from DNA inputs in CFS to produce N-
acetylglucosamine and a peptidoglycan precursor, respect-
ively [171,172]. A five-enzyme pathway that transforms tryp-
tophan into a bioactive pigment called violacein has also
been demonstrated [49,56]. Additionally, a combinatorial
strategy has recently been used to build a 17-step enzyme
pathway for n-butanol [173]. It is intriguing to envision how
this approach could influence the synthesis of high-value
commodities (e.g., small-molecule drugs, cosmetic ingredi-
ents, food additives, and scented compounds), and move
production towards more sustainable enzyme-catalyzed
processes.
The cell-free assembly of engineered metabolic path-
ways has led to parallel approaches in the areas of en-
ergy production, biomaterials, and even the development
of artificial cells. Below we introduce some of the related
efforts in these fields.
Energy storage and generation
Cell-free enzymatic pathways have recently been used to
create biobatteries with small environmental footprints
and energy-storage densities superior to that of current
lithium-ion devices [174]. Moreover, previous studies
have demonstrated ATP generation on electrode surfaces
[175,176]. Since both the assembly of ATP synthase
[113] and the synthesis of membrane proteins into teth-
ered lipid bilayers [177] have been shown in CFS, one
potential application of CFS could be rapid prototyping
and construction of novel energy-generating biodevices
that would be capable of producing electricity from low-
value commodities (i.e., biomass or waste) [109]. One
could readily imagine CFS simply powered by light [178]
or electricity, which could help lower the cost of manu-
facturing industrially relevant biomolecules as discussed
above.
Biomaterials
As noted earlier, CFS have not only been used to screen
the natural diversity of enzymes, but also to sculpt en-
zymatic activity. In an example of this, Bawazer et al.
used CFS to synthesize solid-state materials [52]. A cell-
free system was used to exert evolutionary selection on
biomineralizing enzymes called silicateins that are cap-
able of synthesizing silicon dioxide or titanium dioxide.
DNA fragments coding for two isoforms of silicatein
were digested and reassembled by DNA shuffling to cre-
ate a library of chimeric enzymes. Through a clever
scheme of selection, variants were then chosen for their
ability to deposit silica or titanium dioxide onto
microbeads in an oil-water emulsion. The success of this
methodology through the use of CFS raises the exciting
prospect of using green chemistry for the deposition of
semi-conductor materials. This type of green deposition
could also be modified such that it is guided by a CFS-
compatible photolithography technique similar to that
demonstrated by the Bar-Ziv group [55,179,180].
Artificial cells
Artificial cells have traditionally been defined as encap-
sulated bioactive materials (e.g., RNA, DNA, and en-
zymes) within a membrane compiled to perform a
designated function [134]. Incorporation of CFS into li-
posomes pre-dates much of the cell-free synthetic biol-
ogy discussed above [181,182] and provides a powerful
Tinafar et al. BMC Biology (2019) 17:64 Page 8 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
platform for engineering artificial cells [37,151,183
185]. Artificial cells have many important applications;
they can be used to link phenotype to genotype in vitro
for directed evolution applications, and to spatially sep-
arate synthesis of different proteins [185]. There is also
evidence indicating that confinement, a feature common
to many types of artificial cells, can be used to boost
protein expression yields of CFS [186]. Furthermore,
artificial cells may allow for prolonged expression with-
out relying on traditional dialysis methods that are often
used to provide a continuous supply of reaction precur-
sors. For example, early work by the Noireaux group
showed that membrane-based artificial cells can be aug-
mented with α-hemolysin pore proteins from Staphylo-
coccus aureus in order to achieve selective permeability
for nutrients [182,187].
Artificial cells may also be constructed in the form of
solid-state two-dimensional compartments. Silicon has
been used to fabricate two-dimensional artificial cells
capable of carrying out many of the features possible in
cell-based systems. These features include simple metab-
olism, operation of gene circuits (e.g., oscillators), and
even communication between compartments. Control
over fabrication geometry allows for precise evaluation
of the effects of diffusion gradients and can help tune
protein turnover [55,179].
Looking forward, perhaps one of the most exciting
and promising applications of artificial cells is the ability
to express membrane proteins efficiently. This could
allow for cell-free engineering of signaling pathways
[188], such as those involving G-protein-coupled recep-
tors (GPCRs) [189,190]. Approximately 34% of all FDA-
approved drugs act on GPCR targets [191]. As such,
artificial cells could become an invaluable tool in the
drug discovery process. Artificial cells also have the po-
tential to be used for in vivo therapeutics. For example,
they could be designed to perform sensing, logic, or
therapeutic functions. Artificial cells may be designed to
accumulate at a tumor site through the enhanced per-
meability and retention (EPR) effect [192] or by using
targeting molecules on their surface. They can also be
constructed to protect therapeutic enzymes while being
permeable to specific substrates and products, thus in-
creasing active circulation time and expanding their
therapeutic potential [193,194].
Education
Given their potential for biosafety and portability,
cell-free systems offer a great platform for teaching
key concepts in synthetic biology. The Cold Spring
Harbor Laboratory course in synthetic biology, for ex-
ample, includes modules that utilize cell-free systems
[195]. In recent work led by Jim Collins and Michael
Jewett, the ability of CFS to support on-demand and
on-site sensing and manufacturing was further ex-
tended to bring synthetic biology capabilities to the
classroom [196,197]. Here FD-CF components were
used to create kits that enable students to experience
rational design of reactions, such as creating their
own unique colors by mixing DNA coding for differ-
ent fluorescent proteins. Other applications included
the on-demand creation of fluorescent hydrogels,
scents, and even sensors that could distinguish be-
tween DNA from banana, kiwi, and strawberry.
Reflecting an important trend in the field of synthetic
biology, this work included the testing of tools under
field conditions with the help of high school students.
This work sets the important groundwork for inspir-
ing curiosity and passion in students who will drive
the next generation of synthetic biology.
The future of biotechnology with cell-free systems
The merger of cell-free systems with the vast array of
genetically programmable tools is transforming the syn-
thetic biology landscape, creating powerful in vitro plat-
forms. These platforms have already begun to bring
about de-centralization of health care through portable
diagnostics and drug manufacturing. They also have
great potential for the efficient, centralized production
of high-value commodities. Cell-free synthetic biology
approaches will take biology and biotechnology to new
horizons and will surely produce many creative and un-
expected outcomes. We expect the field to continue to
expand and to merge with other engineered systems.
One could envision programmed interactions with mate-
rials on the nano-scale and interplay with a variety of
engineered enzymes. We are excited to see how CFS will
bring synthetic biology closer to electronics, computa-
tion, and machine learning.
Acknowledgments
Not applicable.
Authorscontributions
AT and KP co-authored and edited the manuscript. KJ edited the manuscript
and created the figures as well as their legends. All authors read and ap-
proved the final manuscript.
Funding
This work was supported by the CIHR Foundation Grant Program
(201610FDN-375469), CIHR/IDRC team grant (149783), and the Canada
Research Chair Program (CIHR, 950-231075) to K.P.
Availability of data and materials
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Tinafar et al. BMC Biology (2019) 17:64 Page 9 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
References
1. Clancy K, Voigt CA. Programming cells: towards an automated Genetic
Compiler. Curr Opin Biotechnol. 2010;21(4):57281. https://doi.org/10.1016/j.
copbio.2010.07.005.
2. van der Meer JR, Belkin S. Where microbiology meets microengineering:
design and applications of reporter bacteria. Nat Rev Microbiol. 2010;8(7):
51122. https://doi.org/10.1038/nrmicro2392.
3. Mao N, Cubillos-Ruiz A, Cameron DE, Collins JJ. Probiotic strains detect and
suppress cholera in mice. Sci Transl Med. 2018;10(445):eaao2586. https://doi.
org/10.1126/scitranslmed.aao2586.
4. Siciliano V, DiAndreth B, Monel B, Beal J, Huh J, Clayton KL, et al.
Engineering modular intracellular protein sensor-actuator devices. Nat
Commun. 2018;9(1):1881. https://doi.org/10.1038/s41467-018-03984-5.
5. Kotula JW, Kerns SJ, Shaket LA, Siraj L, Collins JJ, Way JC, et al.
Programmable bacteria detect and record an environmental signal in the
mammalian gut. Proc Natl Acad Sci U S A. 2014;111(13):483843. https://doi.
org/10.1073/pnas.1321321111.
6. Friedland AE, Lu TK, Wang X, Shi D, Church G, Collins JJ. Synthetic gene
networks that count. Science. 2009;324(5931):1199202. https://doi.org/1
0.1126/science.1172005.
7. Green AA, Kim J, Ma D, Silver PA, Collins JJ, Yin P. Complex cellular logic
computation using ribocomputing devices. Nature. 2017;548(7665):11721.
https://doi.org/10.1038/nature23271.
8. Kitada T, DiAndreth B, Teague B, Weiss R. Programming gene and
engineered-cell therapies with synthetic biology. Science. 2018;359(6376):
eaad1067. https://doi.org/10.1126/science.aad1067.
9. Simpson ML, Sayler GS, Fleming JT, Applegate B. Whole-cell biocomputing.
Trends Biotechnol. 2001;19(8):31723.
10. Yehl K, Lu T. Scaling computation and memory in living cells. Curr Opin
Biomed Eng. 2017;4:14351. https://doi.org/10.1016/j.cobme.2017.10.003.
11. Anderson LA, Islam MA, Prather KLJ. Synthetic biology strategies for
improving microbial synthesis of greenbiopolymers. J Biol Chem. 2018;
293(14):505361. https://doi.org/10.1074/jbc.TM117.000368.
12. Fossati E, Ekins A, Narcross L, Zhu Y, Falgueyret J-P, Beaudoin GAW, et al.
Reconstitution of a 10-gene pathway for synthesis of the plant alkaloid
dihydrosanguinarine in Saccharomyces cerevisiae. Nat Commun. 2014;5:
3283. https://doi.org/10.1038/ncomms4283.
13. Smanski MJ, Zhou H, Claesen J, Shen B, Fischbach MA, Voigt CA. Synthetic
biology to access and expand natures chemical diversity. Nat Rev Microbiol.
2016;14(3):13549. https://doi.org/10.1038/nrmicro.2015.24.
14. Nielsen J, Keasling JD. Engineering cellular metabolism. Cell. 2016;164(6):
118597. https://doi.org/10.1016/j.cell.2016.02.004.
15. Wagner TE, Becraft JR, Bodner K, Teague B, Zhang X, Woo A, et al. Small-
molecule-based regulation of RNA-delivered circuits in mammalian cells.
Nat Chem Biol. 2018;14(11):104350. https://doi.org/10.1038/s41589-018-
0146-9.
16. Scheller L, Strittmatter T, Fuchs D, Bojar D, Fussenegger M. Generalized
extracellular molecule sensor platform for programming cellular behavior.
Nat Chem Biol. 2018;14(7):7239. https://doi.org/10.1038/s41589-018-0046-z.
17. Cho JH, Collins JJ, Wong WW. Universal chimeric antigen receptors for
multiplexed and logical control of T cell responses. Cell. 2018;173(6):1426
1438.e11. https://doi.org/10.1016/j.cell.2018.03.038.
18. Lee JW, Chan CTY, Slomovic S, Collins JJ. Next-generation biocontainment
systems for engineered organisms. Nat Chem Biol. 2018;14(6):5307. https://
doi.org/10.1038/s41589-018-0056-x.
19. Jia B, Qi H, Li B-Z, Pan S, Liu D, Liu H, et al. Orthogonal ribosome biofirewall.
ACS Synth Biol. 2017;6(11):210817. https://doi.org/10.1021/acssynbio.
7b00148.
20. Martin RW, Majewska NI, Chen CX, Albanetti TE, Jimenez RBC, Schmelzer AE,
et al. Development of a CHO-based cell-free platform for synthesis of active
monoclonal antibodies. ACS Synth Biol. 2017;6(7):13709. https://doi.org/1
0.1021/acssynbio.7b00001.
21. Mikami S, Masutani M, Sonenberg N, Yokoyama S, Imataka H. An efficient
mammalian cell-free translation system supplemented with translation
factors. Protein Expr Purif. 2006;46(2):34857. https://doi.org/10.1016/j.pep.2
005.09.021.
22. Tran K, Gurramkonda C, Cooper MA, Pilli M, Taris JE, Selock N, et al. Cell-free
production of a therapeutic protein: Expression, purification, and
characterization of recombinant streptokinase using a CHO lysate.
Biotechnol Bioeng. 2018;115(1):92102. https://doi.org/10.1002/bit.26439.
23. Burgenson D, Gurramkonda C, Pilli M, Ge X, Andar A, Kostov Y, et al. Rapid
recombinant protein expression in cell-free extracts from human blood. Sci
Rep. 2018;8(1):9569. https://doi.org/10.1038/s41598-018-27846-8.
24. Ezure T, Suzuki T, Higashide S, Shintani E, Endo K, Kobayashi S-i, et al. Cell-
free protein synthesis system prepared from insect cells by freeze-thawing.
Biotechnol Prog. 2006;22(6):15707. https://doi.org/10.1021/bp060110v.
25. Buntru M, Vogel S, Stoff K, Spiegel H, Schillberg S. A versatile coupled cell-
free transcription-translation system based on tobacco BY-2 cell lysates.
Biotechnol Bioeng. 2015;112(5):86778. https://doi.org/10.1002/bit.25502.
26. Harbers M. Wheat germ systems for cell-free protein expression. FEBS Lett.
2014;588(17):276273. https://doi.org/10.1016/j.febslet.2014.05.061.
27. Hodgman CE, Jewett MC. Optimized extract preparation methods and
reaction conditions for improved yeast cell-free protein synthesis.
Biotechnol Bioeng. 2013;110(10):264354. https://doi.org/10.1002/bit.24942.
28. Yang WC, Patel KG, Wong HE, Swartz JR. Simplifying and streamlining
Escherichia coli-based cell-free protein synthesis. Biotechnol Prog. 2012;
28(2):41320. https://doi.org/10.1002/btpr.1509.
29. Kigawa T, Yabuki T, Matsuda N, Matsuda T, Nakajima R, Tanaka A, et al.
Preparation of Escherichia coli cell extract for highly productive cell-free
protein expression. J Struct Funct Genomics. 2004;5(12):638. https://doi.
org/10.1023/B:JSFG.0000029204.57846.7d.
30. Moore SJ, Lai H-E, Needham H, Polizzi KM, Freemont PS. Streptomyces
venezuelaeTX-TL - a next generation cell-free synthetic biology tool.
Biotechnol J. 2017;12(4):1600678. https://doi.org/10.1002/biot.201600678.
31. Moore SJ, MacDonald JT, Wienecke S, Ishwarbhai A, Tsipa A, Aw R, et al.
Rapid acquisition and model-based analysis of cell-free transcription-
translation reactions from nonmodel bacteria. Proc Natl Acad Sci U S A.
2018. https://doi.org/10.1073/pnas.1715806115.
32. Kelwick R, Webb AJ, MacDonald JT, Freemont PS. Development of a Bacillus
subtilis cell-free transcription-translation system for prototyping regulatory
elements. Metab Eng. 2016;38:37081. https://doi.org/10.1016/J.YMBEN.2016.
09.008.
33. Li J, Wang H, Jewett MC. Expanding the palette of Streptomyces -based
cell-free protein synthesis systems with enhanced yields. Biochem Eng J.
2018;130:2933. https://doi.org/10.1016/j.bej.2017.11.013.
34. Li J, Wang H, Kwon Y-C, Jewett MC. Establishing a high yielding
streptomyces-based cell-free protein synthesis system. Biotechnol Bioeng.
2017;114(6):134353. https://doi.org/10.1002/bit.26253.
35. Failmezger J, Scholz S, Blombach B, Siemann-Herzberg M. Cell-free protein
synthesis from fast-growing Vibrio natriegens. Front Microbiol. 2018;9:1146.
https://doi.org/10.3389/fmicb.2018.01146.
36. Shimizu Y, Inoue A, Tomari Y, Suzuki T, Yokogawa T, Nishikawa K, et al. Cell-
free translation reconstituted with purified components. Nat Biotechnol.
2001;19(8):7515. https://doi.org/10.1038/90802.
37. Shin J, Noireaux V. An E.coli cell-free expression toolbox: Application to
synthetic gene circuits and artificial cells. ACS Synth Biol. 2012;1(1):2941.
https://doi.org/10.1021/sb200016s.
38. Jewett MC, Calhoun KA, Voloshin A, Wuu JJ, Swartz JR. An integrated cell-
free metabolic platform for protein production and synthetic biology. Mol
Syst Biol. 2008;4:220. https://doi.org/10.1038/msb.2008.57.
39. Li J, Gu L, Aach J, Church GM. Improved cell-free RNA and protein synthesis
system. PLoS One. 2014;9(9):e106232. https://doi.org/10.1371/journal.pone.01
06232.
40. Kwon Y-C, Jewett MC. High-throughput preparation methods of crude
extract for robust cell-free protein synthesis. Sci Rep. 2015;5:8663. https://
doi.org/10.1038/srep08663.
41. Sun ZZ, Hayes CA, Shin J, Caschera F, Murray RM, Noireaux V. Protocols for
implementing an Escherichia coli based TX-TL cell-free expression system
for synthetic biology. J Vis Exp. 2013;79:e50762. https://doi.org/10.3791/5
0762.
42. Caschera F, Noireaux V. Synthesis of 2.3 mg/ml of protein with an all
Escherichia coli cell-free transcriptiontranslation system. Biochimie. 2014;99:
1628. https://doi.org/10.1016/j.biochi.2013.11.025.
43. Wiegand DJ, Lee HH, Ostrov N, Church GM. Establishing a cell-free Vibrio
natriegens expression system. bioRxiv. 2018:331645. https://doi.org/10.11
01/331645.
44. Tuckey C, Asahara H, Zhou Y, Chong S. Protein synthesis using a
reconstituted cell-free system. Curr Protoc Mol Biol. 2014;108:16.31.122.
https://doi.org/10.1002/0471142727.mb1631s108.
Tinafar et al. BMC Biology (2019) 17:64 Page 10 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
45. Didovyk A, Tonooka T, Tsimring L, Hasty J. Rapid and scalable preparation of
bacterial lysates for cell-free gene expression. ACS Synth Biol. 2017;6(12):
2198208. https://doi.org/10.1021/acssynbio.7b00253.
46. Pardee K, Green AA, Ferrante T, Cameron DE, DaleyKeyser A, Yin P, et al.
Paper-based synthetic gene networks. Cell. 2014;159(4):94054. https://doi.
org/10.1016/j.cell.2014.10.004.
47. Chan P, Thomas CJ, Sprang SR, Tall GG. Molecular chaperoning function of
Ric-8 is to fold nascent heterotrimeric G protein αsubunits. Proc Natl Acad
Sci U S A. 2013;110(10):37949. https://doi.org/10.1073/pnas.1220943110.
48. Smith MT, Berkheimer SD, Werner CJ, Bundy BC. Lyophilized Escherichia
coli-based cell-free systems for robust, high-density, long-term storage.
Biotechniques. 2014;56(4):18693. https://doi.org/10.2144/000114158.
49. Pardee K, Slomovic S, Nguyen PQ, Lee JW, Donghia N, Burrill D, et al.
Portable, on-demand biomolecular manufacturing. Cell. 2016;167(1):248
254.e12. https://doi.org/10.1016/j.cell.2016.09.013.
50. Pardee K, Green AA, Takahashi MK, Connor DHO, Gehrke L, Collins JJ, et al. Rapid,
low-cost detection of Zika virus using programmable biomolecular components.
Cell. 2016;165(5):125566. https://doi.org/10.1016/j.cell.2016.04.059.
51. Yin G, Garces ED, Yang J, Zhang J, Tran C, Steiner AR, et al. Aglycosylated
antibodies and antibody fragments produced in a scalable in vitro
transcription-translation system. MAbs. 2012;4(2):21725. https://doi.org/1
0.4161/mabs.4.2.19202.
52. Bawazer LA, Izumi M, Kolodin D, Neilson JR, Schwenzer B, Morse DE.
Evolutionary selection of enzymatically synthesized semiconductors from
biomimetic mineralization vesicles. Proc Natl Acad Sci U S A. 2012;109(26):
E170514. https://doi.org/10.1073/pnas.1116958109.
53. Sun ZZ, Yeung E, Hayes CA, Noireaux V, Murray RM. Linear DNA for rapid
prototyping of synthetic biological circuits in an Escherichia coli based TX-TL cell-
free system. ACS Synth Biol. 2014;3(6):38797. https://doi.org/10.1021/sb400131a.
54. Takahashi MK, Chappell J, Hayes CA, Sun ZZ, Kim J, Singhal V, et al. Rapidly
characterizing the fast dynamics of RNA genetic circuitry with cell-free
transcription-translation (TX-TL) systems. ACS Synth Biol. 2015;4(5):50315.
https://doi.org/10.1021/sb400206c.
55. Karzbrun E, Tayar AM, Noireaux V, Bar-Ziv RH. Programmable on-chip DNA
compartments as artificial cells. Science. 2014;345(6198):82932. https://doi.
org/10.1126/science.1255550.
56. Garamella J, Marshall R, Rustad M, Noireaux V. The all E.coli TX-TL Toolbox 2.
0: A Platform for Cell-Free Synthetic Biology. ACS Synth Biol. 2016;5(4):344
55. https://doi.org/10.1021/acssynbio.5b00296.
57. Niederholtmeyer H, Sun ZZ, Hori Y, Yeung E, Verpoorte A, Murray RM, et al.
Rapid cell-free forward engineering of novel genetic ring oscillators. Elife.
2015;4:e09771. https://doi.org/10.7554/eLife.09771.
58. Green AA, Silver PA, Collins JJ, Yin P. Toehold switches: de-novo-designed
regulators of gene expression. Cell. 2014;159(4):92539. https://doi.org/10.1
016/j.cell.2014.10.002.
59. Takahashi MK, Tan X, Dy AJ, Braff D, Akana RT, Furuta Y, et al. A low-cost
paper-based synthetic biology platform for analyzing gut microbiota and
host biomarkers. Nat Commun. 2018;9(1):3347. https://doi.org/10.1038/s4146
7-018-05864-4.
60. Duyen TTM, Matsuura H, Ujiie K, Muraoka M, Harada K, Hirata K. Paper-based
colorimetric biosensor for antibiotics inhibiting bacterial protein synthesis. J
Biosci Bioeng. 2016;123(1):96100. https://doi.org/10.1016/j.jbiosc.2016.07.015.
61. Wen KY, Cameron L, Chappell J, Jensen K, Bell DJ, Kelwick R, et al. A cell-free
biosensor for detecting quorum sensing molecules in P. aeruginosa-infected
respiratory samples. ACS Synth Biol. 2017;6(12):2293301. https://doi.org/1
0.1021/acssynbio.7b00219.
62. Salehi ASM, Shakalli Tang MJ, Smith MT, Hunt JM, Law RA, Wood DW, et al.
Cell-free protein synthesis approach to biosensing hTRβ-specific endocrine
disruptors. Anal Chem. 2017;89(6):3395401. https://doi.org/10.1021/acs.
analchem.6b04034.
63. Salehi ASM, Yang SO, Earl CC, Shakalli Tang MJ, Porter Hunt J, Smith MT,
et al. Biosensing estrogenic endocrine disruptors in human blood and urine:
A RAPID cell-free protein synthesis approach. Toxicol Appl Pharmacol. 2018;
345:1925. https://doi.org/10.1016/j.taap.2018.02.016.
64. Calhoun KA, Swartz JR. Energizing cell-free protein synthesis with glucose
metabolism. Biotechnol Bioeng. 2005;90(5):60613. https://doi.org/10.1002/
bit.20449.
65. Kim T-W, Kim H-C, Oh I-S, Kim D-M. A highly efficient and economical cell-
free protein synthesis system using the S12 extract of Escherichia coli.
Biotechnol Bioprocess Eng. 2008;13(4):4649. https://doi.org/10.1007/s12257-
008-0139-8.
66. Carlson ED, Gan R, Hodgman CE, Jewett MC. Cell-free protein synthesis:
applications come of age. Biotechnol Adv. 2012;30(5):118594. https://doi.
org/10.1016/j.biotechadv.2011.09.016.
67. Dudley QM, Karim AS, Jewett MC. Cell-free metabolic engineering:
biomanufacturing beyond the cell. Biotechnol J. 2015;10(1):6982. https://
doi.org/10.1002/biot.201400330.
68. Zawada JF, Yin G, Steiner AR, Yang J, Naresh A, Roy SM, et al. Microscale to
manufacturing scale-up of cell-free cytokine production--a new approach
for shortening protein production development timelines. Biotechnol
Bioeng. 2011;108(7):15708. https://doi.org/10.1002/bit.23103.
69. Breaking free from cells; Synthetic biology. Econ. 2017 (May 6).
70. Sullivan CJ, Pendleton ED, Sasmor HH, Hicks WL, Farnum JB, Muto M, et al.
A cell-free expression and purification process for rapid production of
protein biologics. Biotechnol J. 2016;11(2):23848. https://doi.org/10.1002/
biot.201500214.
71. Stech M, Brödel AK, Quast RB, Sachse R, Kubick S. Cell-free systems:
Functional modules for synthetic and chemical biology. Adv Biochem Eng
Biotechnol. 2013;137:67102. https://doi.org/10.1007/10_2013_185.
72. Brödel AK, Wüstenhagen DA, Kubick S. Cell-free protein synthesis systems
derived from cultured mammalian cells. Methods Mol Biol. 2015;1261:129
40. https://doi.org/10.1007/978-1-4939-2230-7_7.
73. Salehi ASM, Smith MT, Bennett AM, Williams JB, Pitt WG, Bundy BC. Cell-free
protein synthesis of a cytotoxic cancer therapeutic: Onconase production
and a just-add-water cell-free system. Biotechnol J. 2016;11(2):27481.
https://doi.org/10.1002/biot.201500237.
74. Groff D, Armstrong S, Rivers PJ, Zhang J, Yang J, Green E, et al. Engineering
toward a bacterial endoplasmic reticulumfor the rapid expression of
immunoglobulin proteins. MAbs. 2014;6(3):6718. https://doi.org/10.4161/
mabs.28172.
75. Cai Q, Hanson JA, Steiner AR, Tran C, Masikat MR, Chen R, et al. A simplified
and robust protocol for immunoglobulin expression in Escherichia coli cell-
free protein synthesis systems. Biotechnol Prog. 2015;31(3):82331. https://
doi.org/10.1002/btpr.2082.
76. Kanter G, Yang J, Voloshin A, Levy S, Swartz JR, Levy R. Cell-free production
of scFv fusion proteins: an efficient approach for personalized lymphoma
vaccines. Blood. 2007;109(8):33939. https://doi.org/10.1182/blood-2006-07-
030593.
77. Stafford RL, Matsumoto ML, Yin G, Cai Q, Fung JJ, Stephenson H, et al. In
vitro Fab display: a cell-free system for IgG discovery. Protein Eng Des Sel.
2014;27(4):97109. https://doi.org/10.1093/protein/gzu002.
78. Kawasaki T, Gouda MD, Sawasaki T, Takai K, Endo Y. Efficient synthesis of a
disulfide-containing protein through a batch cell-free system from wheat
germ. Eur J Biochem. 2003;270(23):47806.
79. Stech M, Merk H, Schenk JA, Stöcklein WFM, Wüstenhagen DA, Micheel B,
et al. Production of functional antibody fragments in a vesicle-based
eukaryotic cell-free translation system. J Biotechnol. 2013;164(2):22031.
https://doi.org/10.1016/j.jbiotec.2012.08.020.
80. Xu Y, Lee J, Tran C, Heibeck TH, Wang WD, Yang J, et al. Production of
bispecific antibodies in knobs-into-holesusing a cell-free expression
system. MAbs. 2015;7(1):231. https://doi.org/10.4161/19420862.2015.989013.
81. Zimmerman ES, Heibeck TH, Gill A, Li X, Murray CJ, Madlansacay MR, et al.
Production of site-specific antibodydrug conjugates using optimized non-
natural amino acids in a cell-free expression system. Bioconjug Chem. 2014;
25(2):35161. https://doi.org/10.1021/bc400490z.
82. Stech M, Quast RB, Sachse R, Schulze C, Wüstenhagen DA, Kubick S. A
continuous-exchange cell-free protein synthesis system based on extracts
from cultured insect cells. PLoS One. 2014;9(5):e96635. https://doi.org/10.13
71/journal.pone.0096635.
83. Bulleid NJ, Bassel-Duby RS, Freedman RB, Sambrook JF, Gething MJ. Cell-free
synthesis of enzymically active tissue-type plasminogen activator. Protein
folding determines the extent of N-linked glycosylation. Biochem J. 1992;
286(Pt 1):27580. https://doi.org/10.1042/BJ2860275.
84. Oh I-S, Kim D-M, Kim T-W, Park C-G, Choi C-Y. Providing an oxidizing
environment for the cell-free expression of disulfide-containing proteins by
exhausting the reducing activity of Escherichia coli S30 extract. Biotechnol
Prog. 2006;22(4):12258. https://doi.org/10.1021/bp060051l.
85. Yin G, Swartz JR. Enhancing multiple disulfide bonded protein folding in a
cell-free system. Biotechnol Bioeng. 2004;86(2):18895. https://doi.org/10.1
002/bit.10827.
86. Palmenberg AC. In vitro synthesis and assembly of picornaviral capsid
intermediate structures. J Virol. 1982;44(3):9006.
Tinafar et al. BMC Biology (2019) 17:64 Page 11 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
87. Welsh JP, Lu Y, He X-S, Greenberg HB, Swartz JR. Cell-free production of
trimeric influenza hemagglutinin head domain proteins as vaccine antigens.
Biotechnol Bioeng. 2012;109(12):29629. https://doi.org/10.1002/bit.24581.
88. Lu Y, Welsh JP, Swartz JR. Production and stabilization of the trimeric
influenza hemagglutinin stem domain for potentially broadly protective
influenza vaccines. Proc Natl Acad Sci U S A. 2014;111(1):125130. doi:
https://doi.org/10.1073/pnas.1308701110
89. Zichel R, Mimran A, Keren A, Barnea A, Steinberger-Levy I, Marcus D, et al.
Efficacy of a potential trivalent vaccine based on Hc fragments of
botulinum toxins A, B, and E produced in a cell-free expression system. Clin
Vaccine Immunol. 2010;17(5):78492. https://doi.org/10.1128/CVI.00496-09.
90. Ng PP, Jia M, Patel KG, Brody JD, Swartz JR, Levy S, et al. A vaccine directed
to B cells and produced by cell-free protein synthesis generates potent
antilymphoma immunity. Proc Natl Acad Sci U S A. 2012;109(36):1452631.
https://doi.org/10.1073/pnas.1211018109.
91. Bundy BC, Franciszkowicz MJ, Swartz JR. Escherichia coli-based cell-free
synthesis of virus-like particles. Biotechnol Bioeng. 2008;100(1):2837.
https://doi.org/10.1002/bit.21716.
92. Lu Y, Chan W, Ko BY, VanLang CC, Swartz JR. Assessing sequence plasticity
of a virus-like nanoparticle by evolution toward a versatile scaffold for
vaccines and drug delivery. Proc Natl Acad Sci U S A. 2015;112(40):123605.
https://doi.org/10.1073/pnas.1510533112.
93. Martemyanov KA, Shirokov VA, Kurnasov OV, Gudkov AT, Spirin AS. Cell-free
production of biologically active polypeptides: application to the synthesis
of antibacterial peptide cecropin. Protein Expr Purif. 2001;21(3):45661.
https://doi.org/10.1006/prep.2001.1400.
94. Sutro Biopharma, Inc. https://www.sutrobio.com/.
95. The Economist. Cell-free biotech will make for better products:
Biotechnology. Econ. 2017.
96. Adiga R, Al-adhami M, Andar A, Borhani S, Brown S, Burgenson D, et al.
Point-of-care production of therapeutic proteins of good-manufacturing-
practice quality. Nat Biomed Eng. 2018;2(9):67586. https://doi.org/10.1038/
s41551-018-0259-1.
97. Murphy TW, Sheng J, Naler LB, Feng X, Lu C. On-chip manufacturing of
synthetic proteins for point-of-care therapeutics. Microsyst Nanoeng. 2019;
25(5):13. https://doi.org/10.1038/s41378-019-0051-8.
98. Lu Y, Welsh JP, Chan W, Swartz JR. Escherichia coli-based cell free
production of flagellin and ordered flagellin display on virus-like particles.
Biotechnol Bioeng. 2013;110(8):207385. https://doi.org/10.1002/bit.24903.
99. Goerke AR, Swartz JR. Development of cell-free protein synthesis platforms
for disulfide bonded proteins. Biotechnol Bioeng. 2008;99(2):35167. https://
doi.org/10.1002/bit.21567.
100. Jiang X, Ookubo Y, Fujii I, Nakano H, Yamane T. Expression of Fab fragment
of catalytic antibody 6D9 in an Escherichia coli in vitro coupled
transcription/translation system. FEBS Lett. 2002;514(2):2904. https://doi.
org/10.1016/S0014-5793(02)02383-9.
101. Laxminarayan R, Duse A, Wattal C, AKMKM Z, HFLFL W, Sumpradit N, et al.
Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;
13(12):105798. https://doi.org/10.1016/S1473-3099(13)70318-9.
102. Shin J, Jardine P, Noireaux V. Genome replication, synthesis, and assembly
of the bacteriophage T7 in a single cell-free reaction. ACS Synth Biol. 2012;
1(9):40813. https://doi.org/10.1021/sb300049p.
103. Rustad M, Eastlund A, Jardine P, Noireaux V. Cell-free TXTL synthesis of
infectious bacteriophage T4 in a single test tube reaction. Synth Biol. 2018;
3(1):17. https://doi.org/10.1093/synbio/ysy002.
104. Rustad M, Eastlund A, Marshall R, Jardine P, Noireaux V. Synthesis of
infectious bacteriophages in an E. coli-based cell-free expression system. J
Vis Exp. 2017;126. https://doi.org/10.3791/56144.
105. Potera C. Phage renaissance: new hope against antibiotic resistance. Environ
Health Perspect. 2013;121(2):a4853. https://doi.org/10.1289/ehp.121-a48.
106. Balogh B, Jones JB, Iriarte FB, Momol MT. Phage therapy for plant disease
control. Curr Pharm Biotechnol. 2010;11(1):4857.
107. Schlegel S, Hjelm A, Baumgarten T, Vikström D, de Gier J-W. Bacterial-based
membrane protein production. Biochim Biophys Acta Mol Cell Res. 2014;
1843(8):173949. https://doi.org/10.1016/J.BBAMCR.2013.10.023.
108. Schneider B, Junge F, Shirokov VA, Durst F, Schwarz D, Dötsch V, et al.
Membrane protein expression in cell-free systems. Methods Mol Biol. 2010;
601:16586. https://doi.org/10.1007/978-1-60761-344-2_11.
109. Perez JG, Stark JC, Jewett MC. Cell-free synthetic biology: Engineering
beyond the cell. Cold Spring Harb Perspect Biol. 2016;8(12):a023853. https://
doi.org/10.1101/cshperspect.a023853.
110. Kaiser L, Graveland-Bikker J, Steuerwald D, Vanberghem M, Herlihy K, Zhang
S. Efficient cell-free production of olfactory receptors: Detergent
optimization, structure, and ligand binding analyses. Proc Natl Acad Sci U S
A. 2008;105(41):1572631. https://doi.org/10.1073/pnas.0804766105.
111. Wang X, Corin K, Baaske P, Wienken CJ, Jerabek-Willemsen M, Duhr S, et al.
Peptide surfactants for cell-free production of functional G protein-coupled
receptors. Proc Natl Acad Sci U S A. 2011;108(22):904954. https://doi.org/1
0.1073/pnas.1018185108.
112. Fogeron M-L, Badillo A, Jirasko V, Gouttenoire J, Paul D, Lancien L, et al.
Wheat germ cell-free expression: Two detergents with a low critical micelle
concentration allow for production of soluble HCV membrane proteins.
Protein Expr Purif. 2015;105:3946. https://doi.org/10.1016/J.PEP.2014.10.003.
113. Matthies D, Haberstock S, Joos F, Dötsch V, Vonck J, Bernhard F, et al. Cell-
free expression and assembly of ATP synthase. J Mol Biol. 2011;413(3):593
603. https://doi.org/10.1016/j.jmb.2011.08.055.
114. Junge F, Haberstock S, Roos C, Stefer S, Proverbio D, Dötsch V, et al.
Advances in cell-free protein synthesis for the functional and structural
analysis of membrane proteins. N Biotechnol. 2011;28(3):26271. https://doi.
org/10.1016/J.NBT.2010.07.002.
115. Sachse R, Dondapati SK, Fenz SF, Schmidt T, Kubick S. Membrane
protein synthesis in cell-free systems: From bio-mimetic systems to bio-
membranes. FEBS Lett. 2014;588(17):277481. https://doi.org/10.1016/J.
FEBSLET.2014.06.007.
116. Panganiban B, Qiao B, Jiang T, DelRe C, Obadia MM, Nguyen TD, et al.
Random heteropolymers preserve protein function in foreign environments.
Science. 2018;359(6381):123943. https://doi.org/10.1126/science.aao0335.
117. Yunker PJ, Asahara H, Hung K-C, Landry C, Arriaga LR, Akartuna I, et al. One-
pot system for synthesis, assembly, and display of functional single-span
membrane proteins on oil-water interfaces. Proc Natl Acad Sci U S A. 2016;
113(3):60813. https://doi.org/10.1073/pnas.1504992113.
118. Asahara H, Chong S. In vitro genetic reconstruction of bacterial transcription
initiation by coupled synthesis and detection of RNA polymerase
holoenzyme. Nucleic Acids Res. 2010;38(13):e141. https://doi.org/10.1093/
nar/gkq377.
119. Huppa JB, Ploegh HL. In vitro translation and assembly of a complete T cell
receptor-CD3 complex. J Exp Med. 1997;186(3):393403. https://doi.org/10.1
084/jem.186.3.393.
120. Matsumoto K, Tomikawa C, Toyooka T, Ochi A, Takano Y, Takayanagi N,
et al. Production of yeast tRNA (m7G46) methyltransferase (Trm8Trm82
complex) in a wheat germ cell-free translation system. J Biotechnol. 2008;
133(4):45360. https://doi.org/10.1016/J.JBIOTEC.2007.11.009.
121. Casteleijn MG, Urtti A, Sarkhel S. Expression without boundaries: Cell-free
protein synthesis in pharmaceutical research. Int J Pharm. 2013;440(1):3947.
https://doi.org/10.1016/j.ijpharm.2012.04.005.
122. Kim D-M, Swartz JR. Efficient production of a bioactive, multiple disulfide-
bonded protein using modified extracts of Escherichia coli. Biotechnol
Bioeng. 2004;85(2):1229. https://doi.org/10.1002/bit.10865.
123. Hallam TJ, Wold E, Wahl A, Smider VV. Antibody conjugates with unnatural
amino acids. Mol Pharm. 2015;12(6):184862. https://doi.org/10.1021/acs.
molpharmaceut.5b00082.
124. ODonoghue P, Ling J, Wang Y-S, Söll D. Upgrading protein synthesis for
synthetic biology. Nat Chem Biol. 2013;9(10):5948. https://doi.org/10.1038/
nchembio.1339.
125. Nguyen DP, Garcia Alai MM, Kapadnis PB, Neumann H, Chin JW. Genetically
encoding N-methyl- l -lysine in recombinant histones. J Am Chem Soc.
2009;131(40):141945. https://doi.org/10.1021/ja906603s.
126. Neumann H, Hancock SM, Buning R, Routh A, Chapman L, Somers J, et al. A
method for genetically installing site-specific acetylation in recombinant
histones defines the effects of H3 K56 acetylation. Mol Cell. 2009;36(1):153
63. https://doi.org/10.1016/j.molcel.2009.07.027.
127. Virdee S, Kapadnis PB, Elliott T, Lang K, Madrzak J, Nguyen DP, et al.
Traceless and site-specific ubiquitination of recombinant proteins. J Am
Chem Soc. 2011;133(28):1070811. https://doi.org/10.1021/ja202799r.
128. Alfonta L, Zhang Z, Uryu S, Loo JA, Schultz PG. Site-specific incorporation of
a redox-active amino acid into proteins. J Am Chem Soc. 2003;125(48):
146623. https://doi.org/10.1021/ja038242x.
129. Cornish VW, Benson DR, Altenbach CA, Hideg K, Hubbell WL, Schultz PG.
Site-specific incorporation of biophysical probes into proteins. Proc Natl
Acad Sci U S A. 1994;91(8):29104.
130. Oza JP, Aerni HR, Pirman NL, Barber KW, ter Haar CM, Rogulina S, et al.
Robust production of recombinant phosphoproteins using cell-free
Tinafar et al. BMC Biology (2019) 17:64 Page 12 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
protein synthesis. Nat Commun. 2015;6(1):8168. https://doi.org/10.1038/
ncomms9168.
131. Shozen N, Iijima I, Hohsaka T. Site-specific incorporation of PEGylated amino
acids into proteins using nonnatural amino acid mutagenesis. Bioorg Med
Chem Lett. 2009;19(17):490911. https://doi.org/10.1016/j.bmcl.2009.07.105.
132. Patel KG, Swartz JR. Surface functionalization of virus-like particles by direct
conjugation using azide-alkyne click chemistry. Bioconjug Chem. 2011;22(3):
37687. https://doi.org/10.1021/bc100367u.
133. Martin RW, Majewska NI, Chen CX, Albanetti TE, Jimenez RBC, Schmelzer AE,
et al. Cell-free protein synthesis from genomically recoded bacteria enables
multisite incorporation of noncanonical amino acids. Nat Commun. 2018;
9(1):1203. https://doi.org/10.1038/s41467-018-03469-5.
134. Lu Y. Cell-free synthetic biology: Engineering in an open world. Synth Syst
Biotechnol. 2017;2(1):237. https://doi.org/10.1016/j.synbio.2017.02.003.
135. Jaroentomeechai T, Stark JC, Natarajan A, Glasscock CJ, Yates LE, Hsu KJ,
et al. Single-pot glycoprotein biosynthesis using a cell-free transcription-
translation system enriched with glycosylation machinery. Nat Commun.
2018;9(1):2686. https://doi.org/10.1038/s41467-018-05110-x.
136. Zhang Y, Ptacin JL, Fischer EC, Aerni HR, Caffaro CE, San Jose K, et al. A
semi-synthetic organism that stores and retrieves increased genetic
information. Nature. 2017;551(7682):6447. https://doi.org/10.1038/
nature24659.
137. Hoshika S, Leal NA, Kim M-J, Kim M-S, Karalkar NB, Kim H-J, et al. Hachimoji
DNA and RNA: A genetic system with eight building blocks. Science. 2019;
363(6429):8847. https://doi.org/10.1126/science.aat0971.
138. Li H, DAnjou M. Pharmacological significance of glycosylation in
therapeutic proteins. Curr Opin Biotechnol. 2009;20(6):67884. https://doi.
org/10.1016/j.copbio.2009.10.009.
139. Guarino C, DeLisa MP. A prokaryote-based cell-free translation system that
efficiently synthesizes glycoproteins. Glycobiology. 2012;22(5):596601.
https://doi.org/10.1093/glycob/cwr151.
140. Gibbs PEM, Zouzias DC, Freedberg IM. Differential post-translational
modification of human type I keratins synthesized in a rabbit reticulocyte
cell-free system. Biochim Biophys Acta Gene Struct Expr. 1985;824(3):24755.
https://doi.org/10.1016/0167-4781(85)90055-7.
141. Dan S, Kang B, Duan X, Wang Y-J. A cell-free system toward deciphering
the post-translational modification barcodes of Oct4 in different cellular
contexts. Biochem Biophys Res Commun. 2015;456(3):71420. https://doi.
org/10.1016/J.BBRC.2014.12.043.
142. Kang S-H, Jun S-Y, Kim D-M. Fluorescent labeling of cell-free synthesized
proteins by incorporation of fluorophore-conjugated nonnatural amino
acids. Anal Biochem. 2007;360(1):16. https://doi.org/10.1016/J.AB.2006.10.02
9.
143. Pagel O, Loroch S, Sickmann A, Zahedi RP. Current strategies and findings in
clinically relevant post-translational modification-specific proteomics. Expert
Rev Proteomics. 2015;12(3):23553. https://doi.org/10.1586/14789450.2015.1
042867.
144. Voigt CA, Mayo SL, Arnold FH, Wang ZG. Computational method to reduce
the search space for directed protein evolution. Proc Natl Acad Sci U S A.
2001;98(7):377883. https://doi.org/10.1073/pnas.051614498.
145. Dodevski I, Markou GC, Sarkar CA. Conceptual and methodological
advances in cell-free directed evolution. Curr Opin Struct Biol. 2015;33:17.
https://doi.org/10.1016/j.sbi.2015.04.008.
146. Roberts RW, Szostak JW. RNA-peptide fusions for the in vitro selection of
peptides and proteins. Proc Natl Acad Sci U S A. 1997;94(23):12297302.
147. Hanes J, Plückthun A. In vitro selection and evolution of functional proteins
by using ribosome display. Proc Natl Acad Sci U S A. 1997;94(10):493742.
148. Sepp A, Tawfik DS, Griffiths AD. Microbead display by in vitro
compartmentalisation: selection for binding using flow cytometry. FEBS Lett.
2002;532(3):4558.
149. Paul S, Stang A, Lennartz K, Tenbusch M, Überla K. Selection of a T7
promoter mutant with enhanced in vitro activity by a novel multi-copy
bead display approach for in vitro evolution. Nucleic Acids Res. 2013;41(1):
e29e29. doi: https://doi.org/10.1093/nar/gks940
150. Diamante L, Gatti-Lafranconi P, Schaerli Y, Hollfelder F. In vitro affinity
screening of protein and peptide binders by megavalent bead surface
display. Protein Eng Des Sel. 2013;26(10):71324. https://doi.org/10.1093/
protein/gzt039.
151. Fujii S, Matsuura T, Sunami T, Kazuta Y, Yomo T. In vitro evolution of α-
hemolysin using a liposome display. Proc Natl Acad Sci U S A. 2013;110(42):
16796801. https://doi.org/10.1073/pnas.1314585110.
152. Sumida T, Yanagawa H, Doi N. In vitro selection of Fab fragments by mRNA
display and gene-linking emulsion PCR. J Nucleic Acids. 2012;2012:19.
https://doi.org/10.1155/2012/371379.
153. Adrio JL, Demain AL. Microbial enzymes: tools for biotechnological processes.
Biomolecules. 2014;4(1):11739. https://doi.org/10.3390/biom4010117.
154. Martin VJJ, Pitera DJ, Withers ST, Newman JD, Keasling JD. Engineering a
mevalonate pathway in Escherichia coli for production of terpenoids. Nat
Biotechnol. 2003;21(7):796802. https://doi.org/10.1038/nbt833.
155. Chang MCY, Eachus RA, Trieu W, Ro D-K, Keasling JD. Engineering
Escherichia coli for production of functionalized terpenoids using plant
P450s. Nat Chem Biol. 2007;3(5):2747. https://doi.org/10.1038/
nchembio875.
156. Newman JD, Marshall J, Chang M, Nowroozi F, Paradise E, Pitera D, et al.
High-level production of amorpha-4,11-diene in a two-phase partitioning
bioreactor of metabolically engineered Escherichia coli. Biotechnol Bioeng.
2006;95(4):68491. https://doi.org/10.1002/bit.21017.
157. Westfall PJ, Pitera DJ, Lenihan JR, Eng D, Woolard FX, Regentin R, et al.
Production of amorphadiene in yeast, and its conversion to
dihydroartemisinic acid, precursor to the antimalarial agent artemisinin. Proc
Natl Acad Sci U S A. 2012;109(3):E1118. https://doi.org/10.1073/pnas.111
0740109.
158. Galanie S, Thodey K, Trenchard IJ, Filsinger Interrante M, Smolke CD.
Complete biosynthesis of opioids in yeast. Science. 2015;349(6252):1095
100. https://doi.org/10.1126/science.aac9373.
159. Nakagawa A, Matsumura E, Koyanagi T, Katayama T, Kawano N, Yoshimatsu
K, et al. Total biosynthesis of opiates by stepwise fermentation using
engineered Escherichia coli. Nat Commun. 2016;7:10390. https://doi.org/10.1
038/ncomms10390.
160. Ajikumar PK, Xiao W-H, Tyo KEJ, Wang Y, Simeon F, Leonard E, et al.
Isoprenoid pathway optimization for Taxol precursor overproduction in
Escherichia coli. Science. 2010;330(6000):704. https://doi.org/10.1126/
science.1191652.
161. Jagadevan S, Banerjee A, Banerjee C, Guria C, Tiwari R, Baweja M, et al.
Recent developments in synthetic biology and metabolic engineering in
microalgae towards biofuel production. Biotechnol Biofuels. 2018;11:185.
https://doi.org/10.1186/s13068-018-1181-1.
162. Keasling JD. Synthetic biology and the development of tools for metabolic
engineering. Metab Eng. 2012;14(3):18995. https://doi.org/10.1016/j.
ymben.2012.01.004.
163. Demain AL. Small bugs, big business: the economic power of the microbe.
Biotechnol Adv. 2000;18(6):499514.
164. Jiang L, Zhao J, Lian J, Xu Z. Cell-free protein synthesis enabled rapid
prototyping for metabolic engineering and synthetic biology. Synth Syst
Biotechnol. 2018;3(2):906. https://doi.org/10.1016/j.synbio.2018.02.003.
165. Chapman J, Ismail A, Dinu C. Industrial applications of enzymes: Recent
advances, techniques, and outlooks. Catalysts. 2018;8(6):238. https://doi.
org/10.3390/catal8060238.
166. Chen X, Zhang C, Zou R, Zhou K, Stephanopoulos G, Too HP. Statistical
experimental design guided optimization of a one-pot biphasic
multienzyme total synthesis of amorpha-4,11-diene. PLoS One. 2013;8(11):
e79650. https://doi.org/10.1371/journal.pone.0079650.
167. Korman TP, Sahachartsiri B, Li D, Vinokur JM, Eisenberg D, Bowie JU. A
synthetic biochemistry system for the in vitro production of isoprene from
glycolysis intermediates. Protein Sci. 2014;23(5):57685. https://doi.org/10.1
002/pro.2436.
168. Liu T, Vora H, Khosla C. Quantitative analysis and engineering of fatty acid
biosynthesis in E. coli. Metab Eng. 2010;12(4):37886. https://doi.org/10.1
016/j.ymben.2010.02.003.
169. Schultheisz HL, Szymczyna BR, Scott LG, Williamson JR. Enzymatic de novo
pyrimidine nucleotide synthesis. J Am Chem Soc. 2011;133(2):297304.
https://doi.org/10.1021/ja1059685.
170. Korman TP, Opgenorth PH, Bowie JU. A synthetic biochemistry platform for
cell free production of monoterpenes from glucose. Nat Commun. 2017;8:
15526. https://doi.org/10.1038/ncomms15526.
171. Sheng J, Huang L, Zhu X, Cai J, Xu Z. Reconstitution of the peptidoglycan
cytoplasmic precursor biosynthetic pathway in cell-free system and rapid
screening of antisense oligonucleotides for Mur enzymes. Appl Microbiol
Biotechnol. 2014;98(4):178594. https://doi.org/10.1007/s00253-013-5467-8.
172. Zhou J, Huang L, Lian J, Sheng J, Cai J, Xu Z. Reconstruction of the UDP-N-
acetylglucosamine biosynthetic pathway in cell-free system. Biotechnol Lett.
2010;32(10):14816. https://doi.org/10.1007/s10529-010-0315-8.
Tinafar et al. BMC Biology (2019) 17:64 Page 13 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
173. Karim AS, Jewett MC. A cell-free framework for rapid biosynthetic pathway
prototyping and enzyme discovery. Metab Eng. 2016;36:11626. https://doi.
org/10.1016/j.ymben.2016.03.002.
174. Zhu Z, Kin Tam T, Sun F, You C, Percival Zhang Y-H. A high-energy-density
sugar biobattery based on a synthetic enzymatic pathway. Nat Commun.
2014;5(1):3026. https://doi.org/10.1038/ncomms4026.
175. Dobson PJ, Hill HAO, Leigh PA, Mazumdar S, Safranov AY. Adenosine
triphosphate synthesis using an electrochemically-driven proton pump. J
Chem Soc Chem Commun. 1994;0(7):807. doi: https://doi.org/10.1039/c3994
0000807
176. Gutiérrez-Sanz Ó, Natale P, Márquez I, Marques MC, Zacarias S, Pita M, et al.
H
2
-fueled ATP synthesis on an electrode: Mimicking cellular respiration.
Angew Chemie Int Ed. 2016;55(21):621620. https://doi.org/10.1002/anie.2
01600752.
177. Zieleniecki JL, Nagarajan Y, Waters S, Rongala J, Thompson V, Hrmova M,
et al. Cell-free synthesis of a functional membrane transporter into a
tethered bilayer lipid membrane. Langmuir. 2016;32(10):24459. https://doi.
org/10.1021/acs.langmuir.5b04059.
178. Berhanu S, Ueda T, Kuruma Y. Artificial photosynthetic cell producing
energy for protein synthesis. Nat Commun. 2019;10(1):1325. https://doi.org/1
0.1038/s41467-019-09147-4.
179. Buxboim A, Bar-Dagan M, Frydman V, Zbaida D, Morpurgo M, Bar-Ziv R. A
single-step photolithographic interface for cell-free gene expression and
active biochips. Small. 2007;3(3):50010. https://doi.org/10.1002/smll.2006004
89.
180. Bar M, Bar-Ziv RH. Spatially resolved DNA brushes on a chip: gene activation
by enzymatic cascade. Nano Lett. 2009;9(12):44626. https://doi.org/10.1021/
nl902748g.
181. Ishikawa K, Sato K, Shima Y, Urabe I, Yomo T. Expression of a cascading
genetic network within liposomes. FEBS Lett. 2004;576(3):38790. https://doi.
org/10.1016/j.febslet.2004.09.046.
182. Noireaux V, Libchaber A. A vesicle bioreactor as a step toward an artificial
cell assembly. Proc Natl Acad Sci U S A. 2004;101(51):1766974. https://doi.
org/10.1073/pnas.0408236101.
183. Kuruma Y, Stano P, Ueda T, Luisi PL. A synthetic biology approach to the
construction of membrane proteins in semi-synthetic minimal cells. Biochim
Biophys Acta Biomembr. 2009;1788(2):56774. https://doi.org/10.1016/J.
BBAMEM.2008.10.017.
184. Wu F, Tan C. The engineering of artificial cellular nanosystems using
synthetic biology approaches. Wiley Interdiscip Rev Nanomed
Nanobiotechnol. 2014;6(4):36983. https://doi.org/10.1002/wnan.1265.
185. Elani Y, Law RV, Ces O. Protein synthesis in artificial cells: using
compartmentalisation for spatial organisation in vesicle bioreactors. Phys
Chem Chem Phys. 2015;17(24):155347. https://doi.org/10.1039/C4CP05933F.
186. Sakamoto R, Noireaux V, Maeda YT. Anomalous scaling of gene expression
in confined cell-free reactions. Sci Rep. 2018;8(1):7364. https://doi.org/10.103
8/s41598-018-25532-3.
187. Noireaux V, Bar-Ziv R, Godefroy J, Salman H, Libchaber A. Toward an
artificial cell based on gene expression in vesicles. Phys Biol. 2005;2(3):P1
P8. doi: https://doi.org/10.1088/1478-3975/2/3/P01
188. Ho KKY, Murray VL, Liu AP. Engineering artificial cells by combining HeLa-
based cell-free expression and ultrathin double emulsion template. Methods
Cell Biol. 2015;128:30318. https://doi.org/10.1016/BS.MCB.2015.01.014.
189. Segers K, Masure S. Cell-free expression of G protein-coupled receptors. Curr
Protoc Protein Sci. 2015;81:29.14.129.14.29. https://doi.org/10.1002/047114
0864.ps2914s81.
190. Sonnabend A, Spahn V, Stech M, Zemella A, Stein C, Kubick S. Production of
G protein-coupled receptors in an insect-based cell-free system. Biotechnol
Bioeng. 2017;114(10):232838. https://doi.org/10.1002/bit.26346.
191. Hauser AS, Attwood MM, Rask-Andersen M, Schiöth HB, Gloriam DE. Trends
in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug
Discov. 2017;16(12):82942. https://doi.org/10.1038/nrd.2017.178.
192. Usmani A, Mishra A, Ahmad M. Nanomedicines: a theranostic approach for
hepatocellular carcinoma. Artif Cells Nanomed Biotechnol. 2018;46(4):680
90. https://doi.org/10.1080/21691401.2017.1374282.
193. Chang TMM. Semipermeable microcapsules. Science. 1964;146(3643):5245.
https://doi.org/10.1126/SCIENCE.146.3643.524.
194. Xu C, Hu S, Chen X. Artificial cells: from basic science to applications. Mater Today
(Kidlington). 2016;19(9):51632. https://doi.org/10.1016/j.mattod.2016.02.020.
195. Cold Spring Harbor Laboratory Course in Synthetic Biology [Internet]
(https://meetings.cshl.edu/courses.aspx?course=C-SYNBIO&year=19).
196. Huang A, Nguyen PQ, Stark JC, Takahashi MK, Donghia N, Ferrante T, et al.
BioBits
TM
Explorer: A modular synthetic biology education kit. Sci Adv. 2018;
4(8):eaat5105. https://doi.org/10.1126/sciadv.aat5105.
197. Stark JC, Huang A, Nguyen PQ, Dubner RS, Hsu KJ, Ferrante TC, et al.
BioBits
TM
Bright: A fluorescent synthetic biology education kit. Sci Adv. 2018;
4(8):eaat5107. https://doi.org/10.1126/sciadv.aat5107.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Tinafar et al. BMC Biology (2019) 17:64 Page 14 of 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Cell-free protein synthesis (CFPS) systems provide a promising alternative for the rapid expression of target proteins (Gregorio et al., 2019;Perez et al., 2016;Zemella et al., 2015). Utilizing an appropriate DNA template facilitates the synthesis of target proteins within a few hours, thereby enabling the study of challenging proteins, including those with disulfide bonds that are sometimes difficult to express in conventional cellbased systems (Chiba et al., 2021;Stech et al., 2017;Tinafar et al., 2019). CFPS has been successfully used to produce various antibody fragments and full-length IgGs (Buntru et al., 2015;Martin et al., 2017;Murakami et al., 2019;Xu et al., 2015). ...
Article
Full-text available
Antibodies are critical tools in medicine and research, and their affinity for their target antigens is a key determinant of their efficacy. Traditional antibody affinity maturation and interaction analyses are often hampered by time‐consuming steps such as cloning, expression, purification, and interaction assays. To address this, we have developed FASTIA (Fast Affinity Screening Technology for Interaction Analysis), a novel platform that integrates rapid gene fragment preparation, cell‐free protein synthesis, and bio‐layer interferometry with non‐regenerative analysis. Using this approach, we can analyze the intermolecular interactions of over 20 variants over 2 days, requiring only the parent protein expression plasmid and basic equipment. We have demonstrated the ability of FASTIA to discriminate between single‐domain antibody variants with different binding affinities using the anti‐HEL VHH antibody D2‐L29, and mapped the results to the crystal structure to identify key interaction sites. FASTIA provides results comparable to those obtained using traditional methods. Our system bypasses the need for genetic engineering facilities and can be easily adopted by laboratories, accelerating the protein engineering and optimization processes. In addition, FASTIA is applicable to other protein–protein interactions, making it a versatile tool for studying molecular recognition. FASTIA facilitates efficient affinity maturation, protein engineering, and analysis of protein–protein interactions. This provides a rapid and accessible route for improving antibodies and a broader understanding of protein interactions.
Article
Full-text available
Synthetic cells encapsulating cell-free protein synthesis machinery are currently limited to laboratory use due to preservation challenges. They are typically produced and used immediately. We present drying methods for long-term...
Article
Heme is a chemical compound crucial for various biological processes and industrial applications. However, the microbial production of heme is often limited by its intracellular accumulation and associated toxicity. To address this, we employed a two-step approach involving in vivo cell cultivation for the production of a heme precursor (coproporphyrin III or coproheme) followed by its in vitro conversion(s) to heme. For the first step, we engineered Escherichia coli strains by implementing the coproporphyrin-dependent (CPD) pathway for bacterial cell cultivation, extracellularly producing up to 251 mg/L coproporphyrin III and 85 mg/L coproheme, respectively. For the second step, we cloned the hemH and hemQ genes for expression in E. coli, and the expressed gene products, i.e., coproheme decarboxylase (ChdC/HemH) and heme synthase (HemQ), were purified. Using the purified enzymes with modulated reaction conditions, we achieved up to a 77.2% yield to convert coproporphyrin III to coproheme and a 45.8% yield to convert coproheme to heme. This in vitro approach not only bypassed the intracellular toxicity constraint associated with in vivo cell cultivation but also enabled precise reaction control, leading to a higher efficiency and yield for heme (and coproheme) production. By applying novel strategies in strain engineering and bioprocessing to overcome inherent bioprocess challenges, this study paves the way for industrial biotechnology for the sustainable, efficient, and even large-scale bio-based production of heme.
Article
Here we report on the development of a CRISPR-based assay for the sensitive and specific detection of antibodies and antigens directly in complex sample matrices. The assay, called Molecular Assay based on antibody-Induced Guide-RNA Enzymatic Transcription (MAIGRET), is based on the use of a responsive synthetic DNA template that triggers the cell-free in vitro transcription of a guide RNA strand upon recognition of a specific target antibody. Such transcribed guide RNA activates the DNA collateral activity of the Cas12a enzyme, leading to the downstream cleavage of a fluorophore/quencher-labeled reporter and thus resulting in an increase in the measured fluorescence signal. We have used MAIGRET for the detection of six different antibodies with high sensitivity (detection limit in the picomolar range) and specificity (no signal in the presence of non-target antibodies). MAIGRET can also be adapted to a competitive approach for the detection of specific antigens. With MAIGRET, we significantly expand the scope and applicability of CRISPR-based sensing approaches to potentially enable the measurement of any molecular target for which an antibody is available.
Article
Full-text available
The Cell-Free Protein Synthesis (CFPS) is an innovative technique used to produce various proteins. It has several advantages, including short expression times, no strain engineering is required, and toxic proteins such as membrane proteins can be produced. However, the most important advantage is that it eliminates the need for a living cell as a production system. Membrane proteins (MPs) are difficult to express in heterologous strains such as Escherichia coli. Modified strains must be used, and sometimes the strain produces them as inclusion bodies, which makes purification difficult. CFPS can avoid the problem of toxicity and, with the use of additives, allows the production of folded and functional membrane proteins. In this review, we focus on describing what cell-free systems are. We address the advantages and disadvantages of the different organisms that can be used to obtain cell extracts, including PURE systems, where the components are obtained recombinantly, and the methodologies that allow the synthesis of membrane proteins in cell-free systems, which, given their hydrophobic nature, require additives for their correct folding. Graphical Abstract
Article
Cell-free systems are powerful tools in synthetic biology with versatile and wide-ranging applications. However, a significant bottleneck for these systems, particularly the PURE cell-free system, is their limited reaction lifespan and yield. Dialysis offers a promising approach to prolong reaction lifetimes and increase yields, yet most custom dialysis systems require access to sophisticated equipment like 3D printers or microfabrication tools. In this study, we utilized an easy-to-assemble, medium-scale dialysis system for cell-free reactions using commercially available components. By employing dialysis with periodic exchange of the feeding solution, we achieved a protein yield of 1.16 mg/mL GFP in the PURE system and extended protein synthesis for at least 12.5 consecutive days, demonstrating the system’s excellent stability.
Article
Full-text available
Microfluidics: Providing therapeutic proteins at the point of care A microfluidic reactor enables researchers to synthesize therapeutic proteins at the point of care. Therapeutic proteins are usually produced in a central location, requiring costly refrigeration and transport to the location where they’re administered. This hinders care for patients in remote or resource-scarce areas. Engineers from the United States’ Virginia Tech, led by Chang Lu, have now developed a microfluidic reactor that uses cell components and a DNA template, completed by a protein purification device. The team validated their device by producing an easily quantifiable protein GFP, before producing therapeutically viable concentrations of cecropin B, a protein used in the treatment of microbial infections. The functionality of the synthesized cecropin B was confirmed by its successful inhibition of Escherichia coli. The devices are low-cost, and future research could extend their utility to other clinically beneficial proteins.
Article
Full-text available
Attempts to construct an artificial cell have widened our understanding of living organisms. Many intracellular systems have been reconstructed by assembling molecules, however the mechanism to synthesize its own constituents by self-sufficient energy has to the best of our knowledge not been developed. Here, we combine a cell-free protein synthesis system and small proteoliposomes, which consist of purified ATP synthase and bacteriorhodopsin, inside a giant unilamellar vesicle to synthesize protein by the production of ATP by light. The photo-synthesized ATP is consumed as a substrate for transcription and as an energy for translation, eventually driving the synthesis of bacteriorhodopsin or constituent proteins of ATP synthase, the original essential components of the proteoliposome. The de novo photosynthesized bacteriorhodopsin and the parts of ATP synthase integrate into the artificial photosynthetic organelle and enhance its ATP photosynthetic activity through the positive feedback of the products. Our artificial photosynthetic cell system paves the way to construct an energetically independent artificial cell.
Article
Full-text available
Expanding the genetic code DNA and RNA are naturally composed of four nucleotide bases that form hydrogen bonds in order to pair. Hoshika et al. added an additional four synthetic nucleotides to produce an eight-letter genetic code and generate so-called hachimoji DNA. Coupled with an engineered T7 RNA polymerase, this expanded DNA alphabet could be transcribed into RNA. Thus, new forms of DNA that add information density to genetic biopolymers can be generated that may be useful for future synthetic biological applications. Science , this issue p. 884
Article
Full-text available
Synthetic mRNA is an attractive vehicle for gene therapies because of its transient nature and improved safety profile over DNA. However, unlike DNA, broadly applicable methods to control expression from mRNA are lacking. Here we describe a platform for small-molecule-based regulation of expression from modified RNA (modRNA) and self-replicating RNA (replicon) delivered to mammalian cells. Specifically, we engineer small-molecule-responsive RNA binding proteins to control expression of proteins from RNA-encoded genetic circuits. Coupled with specific modRNA dosages or engineered elements from a replicon, including a subgenomic promoter library, we demonstrate the capability to externally regulate the timing and level of protein expression. These control mechanisms facilitate the construction of ON, OFF, and two-output switches, with potential therapeutic applications such as inducible cancer immunotherapies. These circuits, along with other synthetic networks that can be developed using these tools, will expand the utility of synthetic mRNA as a therapeutic modality.
Article
Full-text available
There is a need for large-scale, longitudinal studies to determine the mechanisms by which the gut microbiome and its interactions with the host affect human health and disease. Current methods for profiling the microbiome typically utilize next-generation sequencing applications that are expensive, slow, and complex. Here, we present a synthetic biology platform for affordable, on-demand, and simple analysis of microbiome samples using RNA toehold switch sensors in paper-based, cell-free reactions. We demonstrate species-specific detection of mRNAs from 10 different bacteria that affect human health and four clinically relevant host biomarkers. We develop a method to quantify mRNA using our toehold sensors and validate our platform on clinical stool samples by comparison to RT-qPCR. We further highlight the potential clinical utility of the platform by showing that it can be used to rapidly and inexpensively detect toxin mRNA in the diagnosis of Clostridium difficile infections.
Article
Full-text available
Synthetic biology offers opportunities for experiential educational activities at the intersection of the life sciences, engineering, and design. However, implementation of hands-on biology activities in classrooms is challenging because of the need for specialized equipment and expertise to grow living cells. We present BioBits™ Bright, a shelf-stable, just-add-water synthetic biology education kit with easy visual outputs enabled by expression of fluorescent proteins in freeze-dried, cell-free reactions. We introduce activities and supporting curricula for teaching the central dogma, tunable protein expression, and design-build-test cycles and report data generated by K-12 teachers and students. We also develop inexpensive incubators and imagers, resulting in a comprehensive kit costing <US$100 per 30-person classroom. The user-friendly resources of this kit promise to enhance biology education both inside and outside the classroom.
Article
Full-text available
Hands-on demonstrations greatly enhance the teaching of science, technology, engineering, and mathematics (STEM) concepts and foster engagement and exploration in the sciences. While numerous chemistry and physics classroom demonstrations exist, few biology demonstrations are practical and accessible due to the challenges and concerns of growing living cells in classrooms. We introduce BioBits™ Explorer, a synthetic biology educational kit based on shelf-stable, freeze-dried, cell-free (FD-CF) reactions, which are activated by simply adding water. The FD-CF reactions engage the senses of sight, smell, and touch with outputs that produce fluorescence, fragrances, and hydrogels, respectively. We introduce components that can teach tunable protein expression, enzymatic reactions, biomaterial formation, and biosensors using RNA switches, some of which represent original FD-CF outputs that expand the toolbox of cell-free synthetic biology. The BioBits™ Explorer kit enables hands-on demonstrations of cutting-edge science that are inexpensive and easy to use, circumventing many current barriers for implementing exploratory biology experiments in classrooms.
Article
Full-text available
Manufacturing technologies for biologics rely on large, centralized, good-manufacturing-practice (GMP) production facilities and on a cumbersome product-distribution network. Here, we report the development of an automated and portable medicines-on-demand device that enables consistent, small-scale GMP manufacturing of therapeutic-grade biologics on a timescale of hours. The device couples the in vitro translation of target proteins from ribosomal DNA, using extracts from reconstituted lyophilized Chinese hamster ovary cells, with the continuous purification of the proteins. We used the device to reproducibly manufacture His-tagged granulocyte-colony stimulating factor, erythropoietin, glucose-binding protein and diphtheria toxoid DT5. Medicines-on-demand technology may enable the rapid manufacturing of biologics at the point of care.
Article
Full-text available
The emerging discipline of bacterial glycoengineering has made it possible to produce designer glycans and glycoconjugates for use as vaccines and therapeutics. Unfortunately, cell-based production of homogeneous glycoproteins remains a significant challenge due to cell viability constraints and the inability to control glycosylation components at precise ratios in vivo. To address these challenges, we describe a novel cell-free glycoprotein synthesis (CFGpS) technology that seamlessly integrates protein biosynthesis with asparagine-linked protein glycosylation. This technology leverages a glyco-optimized Escherichia coli strain to source cell extracts that are selectively enriched with glycosylation components, including oligosaccharyltransferases (OSTs) and lipid-linked oligosaccharides (LLOs). The resulting extracts enable a one-pot reaction scheme for efficient and site-specific glycosylation of target proteins. The CFGpS platform is highly modular, allowing the use of multiple distinct OSTs and structurally diverse LLOs. As such, we anticipate CFGpS will facilitate fundamental understanding in glycoscience and make possible applications in on demand biomanufacturing of glycoproteins.
Article
The fast growing bacterium Vibrio natriegens is an emerging microbial host for biotechnology. Harnessing its productive cellular components may offer a compelling platform for rapid protein production and prototyping of metabolic pathways or genetic circuits. Here, we report the development of a V. natriegens cell-free expression system. We devised a simplified crude extract preparation protocol and achieved >260 μg/mL of super-folder GFP in a small-scale batch reaction after three hours. Culturing conditions, including growth media and cell density, significantly affect translation kinetics and protein yield of extracts. We observed maximal protein yield at incubation temperatures of 26°C or 30°C, and show improved yield by tuning ions crucial for ribosomal stability. This work establishes an initial V. natriegens cell-free expression system, enables probing of V. natriegens biology, and will serve as a platform to accelerate metabolic engineering and synthetic biology applications.