Marys Medicine

 

Mse.lab.scu.edu.cn

NIH Public Access
Author Manuscript
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
NIH-PA Author Manuscript Published in final edited form as: Nat Rev Genet. 2010 May ; 11(5): 367–379. doi:10.1038/nrg2775.
Synthetic Biology: Applications Come of Age
Ahmad S. Khalil1 and James J. Collins1,2,*
1 Howard Hughes Medical Institute, Department of Biomedical Engineering, Center for
BioDynamics, and Center for Advanced Biotechnology, Boston University, Boston, MA 02215, USA
2 Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA Synthetic biology is bringing together engineers and biologists to design and build novelbiomolecular components, networks and pathways, and to use these constructs to rewire andreprogram organisms. These re-engineered organisms will change our lives in the coming years,leading to cheaper drugs, "green" means to fuel our cars, and targeted therapies to attack "superbugs" NIH-PA Author Manuscript and diseases such as cancer. The de novo engineering of genetic circuits, biological modules, andsynthetic pathways is beginning to address these critical problems and is being used in relatedpractical applications.
The circuit-like connectivity of biological parts and their ability to collectively process logicaloperations was first appreciated nearly 50 years ago1. This inspired attempts to describebiological regulation schemes with mathematical models2–5 and to apply circuit analogies fromestablished frameworks in electrical engineering6, 7. Meanwhile, breakthroughs in genomicresearch and genetic engineering (e.g., recombinant DNA technology) were supplying theinventory and methods necessary to physically construct and assemble biomolecular parts. Asa result, synthetic biology was born with the broad goal of engineering or "wiring" biologicalcircuitry—be it genetic, protein, viral, pathway, or genomic—for manifesting logical forms ofcellular control. Synthetic biology, equipped with the engineering-driven approaches ofmodularization, rationalization, and modeling, has progressed rapidly and generated an ever-increasing suite of genetic devices and biological modules.
The successful design and construction of the first synthetic gene networks—the genetic toggle NIH-PA Author Manuscript switch8 and the repressilator9 (Box 1)—demonstrated that engineering-based methodologycould indeed be applied to build sophisticated, computing-like behaviour into biologicalsystems. In these two cases, basic transcriptional regulatory elements were designed andassembled to realize the biological equivalents of electronic memory storage and timekeeping(Box 1). Within the framework provided by these two synthetic systems, biological circuitscan be built from smaller well-defined parts according to model blueprints, they can be studiedand tested in isolation, and their behaviour can be evaluated against model predictions of thesystem dynamics. This methodology has subsequently been applied in the syntheticconstruction of additional genetic switches8, 10–18, memory elements8, 14, 15, 19, andoscillators9, 10, 20–23, as well as of other electronics-inspired genetic devices, including pulsegenerators24, digital logic gates25–30, filters31–33, and communication modules23, 31, 34, 35.
*To whom correspondence should be addressed ([email protected]).
Khalil and Collins NIH-PA Author Manuscript Early synthetic biology designs: switches and oscillators
Switches and oscillators that occur in electronic systems are also seen in biology and havebeen engineered into synthetic biological systems.
In electronics, one of the most basic elements for storing memory is the reset-set (RS) latchbased on logical NOR gates. This device is bistable, in that it possesses two stable statesthat can be toggled with the delivery of specified inputs. Upon removal of the input, thecircuit retains memory of its current state indefinitely. These forms of memory and stateswitching have significant implications in biology, such as in the differentiation of cellsfrom an initially undifferentiated state. One means by which cellular systems can achievebistability is through genetic mutual repression. The natural PR/PRM genetic switch frombacteriophage lambda, which uses this network architecture to govern the lysis/lysogenydecision, consists of two promoters that are each repressed by the gene product of the other(that is, by the Cro and CI repressor proteins). The genetic toggle switch8, constructed byour research group, is a synthetically engineered version of this co-repressed gene regulationscheme. In one version of the genetic toggle, the PL promoter from lambda phage was usedto drive transcription of lacI, whose product represses a second promoter Ptrc-2 (a lac NIH-PA Author Manuscript promoter variant). Ptrc-2, on the other hand, drives expression of a temperature-sensitiveλ CI repressor protein (cI-ts), which inhibits the PL promoter. The activity of the circuit ismonitored through the expression of GFP. The system can be toggled in one direction withthe exogenous addition of the chemical inducer isopropyl-β-D-thiogalactopyranoside(IPTG) or in the other with a transient increase in temperature. Importantly, upon removalof these exogenous signals, the system retains its current state, creating a cellular form ofmemory.
Timing mechanisms, much like memory, are also fundamental to many electronic andbiological systems. Electronic timekeeping can be achieved with basic oscillator circuits,such as the LC circuit (inductor L and capacitor C), which act as resonators for producingperiodic electronic signals. Biological timekeeping, which is widespread among livingorganisms120, is achieved with circadian clocks and similar oscillator circuits, such as theone responsible for synchronizing the critical processes of photosynthesis and nitrogenfixation in cyanobacteria. The circadian clock of cyanobacteria is based on, among otherregulatory mechanisms, intertwined positive and negative feedback loops on the clock genes NIH-PA Author Manuscript kaiA, kaiB, and kaiC. Elowitz and Leibler constructed a synthetic genetic oscillator based,not on clock genes, but on standard transcriptional repressors (the repressilator)9. Here, acyclic negative feedback loop composed of three promoter–gene pairs, in which the ‘first'promoter in the cascade drives expression of the ‘second' promoter's repressor and so on,was used to drive oscillatory output in gene expression.
Now, ten years after the demonstration of synthetic biology's inaugural devices8, 9, engineeredbiomolecular networks are beginning to move into the application stage and yield solutions tomany complex societal problems. While much work remains on elucidating biological designprinciples36, this foray into practical applications signals an exciting coming-of-age time forthe field.
Here, we review the practical application of synthetic biology to biosensing, therapeutics, andthe production of biofuels, pharmaceuticals and novel biomaterials. Many of the examplesherein do not fit exclusively or neatly into only one of these three application categories; Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins however, it is precisely this multivalent applicability that makes synthetic biology platformsso powerful and promising.
NIH-PA Author Manuscript Cells have evolved a myriad of regulatory circuits—from transcriptional to post-translational—for sensing and responding to diverse and transient environmental signals. These circuitsconsist of exquisitely tailored sensitive elements that bind analytes and set signal detectionthresholds, and transducer modules that filter the signals and mobilize a cellular response (Box2). The two basic sensing modules must be delicately balanced: this is achieved byprogramming modularity and specificity into biosensing circuits at the transcriptional,translational and post-translational levels, as described below.
Synthetic biosensor architectures and examples
Biosensors consist of two basic modules (top panel in figure): sensitive elements forrecognizing and binding analytes, and transducer modules for transmitting and reportingsignals.
NIH-PA Author Manuscript Transcriptional biosensors (panel a) are built by linking environment-responsive promotersto engineered gene circuits for programmed transcriptional changes. In the example shown,a transcriptional AND gate was designed to sense and report only the simultaneous presenceof two environmental signals (e.g., salicylate and arabinose)25. At one gate input, theresearchers encoded an environment-responsive promoter (e.g., PBAD) that activatestranscription of a T7 RNA polymerase gene in response to a single environmental signal(e.g., arabinose). The gene, however, carries internally encoded amber stop codons (redasterisks) that function to block translation of its transcript. Activation of the second gateinput is the key to unlocking translation; specifically, translation can be induced when asecond promoter (e.g., Psal) activates transcription of the SupD amber suppressor tRNA inresponse to a second unique signal (e.g., salicylate). In other words, only when the twoenvironmental signals are simultaneously present can the T7 RNA polymerase be faithfullyexpressed and used to activate an output T7 promoter. This is an example of howsophisticated specificity can be programmed into a transducer module by creatively linkingthe sensory information of multiple sensitive elements. Furthermore, the design istranscriptionally modular in that different sets of environment-responsive promoters can beinterfaced to the AND gate.
NIH-PA Author Manuscript Translational biosensors (panel b) are typically built by linking RNA aptamer domains toRNA regulatory domains. The example shown is an OFF "antiswitch". Here, the smallmolecule theophylline is recognized and bound by the aptamer stem of the RNA biosensor.
This causes a conformational change in the molecule that liberates the antisense domainfrom its sequestering stem loop and allows it to inhibit translation of an output reporter11.
Post-translational biosensors (panel c) consist of membrane-bound protein receptors thattrigger signal transduction cascades through signaling proteins, such as response regulatorsof two-component systems. In the example shown, a synthetic protein scaffold wasengineered to physically localize the pathway components of the yeast mating mitogen-activated protein (MAP) kinase pathway, which here is being triggered by the alpha matingfactor79. By recruiting pathway positive and negative modulators (+/−) to the scaffold, the Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins modular system can be tuned to enable desired responses to upstream signals (e.g.,accelerated, delayed, ultrasensitive responses).
NIH-PA Author Manuscript The hybrid example (panel d) shows a synthetic genetic edge detection circuit82. Thesensitive element is a light-dark sensor, Cph8, made as a chimera of the photoreceptordomain of the cyanobacteria phytochrome Cph1 and the kinase domain of E. coli EnvZ.
This synthetic sensor activates an engineered gene circuit that combines cell-cellcommunication (genes and promoters of the Lux operon) with a logical AND gate (Plux-λ)in order to trace the edges of an image. Specifically, the absence of light triggers Cph8kinase activity, which correspondingly activates the ompC promoter. Cells not receivinglight will therefore produce a cell-cell communication molecule 3-oxohexanoyl-homoserine lactone (AHL)—via expression of its biosynthetic enzyme LuxI—and thetranscriptional repressor CI. AHL binds to the constitutively expressed transcription factorLuxR to activate expression from the Plux-λ promoter, which is simultaneously anddominantly repressed by CI. The result is that only cells that receive light and are nearby toAHL-producing dark cells will activate the final gate and produce pigment through β-galactosidase activity.
NIH-PA Author Manuscript As the first dedicated phase of gene expression, transcription serves as one method by whichcells mobilize a cellular response to an environmental perturbation. As such, the transcriptionmachinery, which includes the genes to be expressed, their promoters, RNA polymerase andtranscription factors, all serve as potential engineering components for transcriptionalbiosensors. Most synthetic designs have focused on the promoters and their associatedtranscription factors, given the abundance of known and characterized prokaryotic andeukaryotic environment-responsive promoters, which include the well-known promoters of theEscherichia coli (E. coli) lac, tet, and ara operons.
Both the sensory and transducer behaviours of a biosensor can be placed under synthetic controlby directly engineering environment-responsive promoter sequences. In fact, this was the earlydesign strategy adopted to establish inducible expression systems37–40. By introducing,removing, or modifying activator and repressor sites, a promoter's sensitivity to a moleculecan be tuned. Synthetic mammalian transactivation systems are generic versions of thisstrategy, in which an environmentally-sensitive transcription factor is fused to a mammaliantransactivation domain to cause inducer-dependent changes in gene expression. Syntheticmammalian biosensors based on this scheme have been created for sensing signals such as NIH-PA Author Manuscript antibiotics41–43, quorum-sensing molecules44, 45, gases and metabolites46–49, and temperaturechanges50, 51. Fussenegger and colleagues have even incorporated this transgene design intomammalian circuits, creating synthetic networks that are responsive to electrical signals52.
While the engineering of environment-responsive promoters has been valuable and useful,additional control over modularity and specificity can be achieved by embedding environment-responsive promoters within engineered gene networks. Achieving true modularity withgenetic parts is inherently difficult because of unintended interference among native andsynthetic parts, and therefore requires careful decoupling of functional modules. One suchmodular design strategy was employed by Kobayashi et al.34 to develop whole-cell E. colibiosensors that respond to signals in a programmable fashion. In this design, a sensory module(i.e., an environment-responsive promoter and associated transcription factor) was coupled toan engineered gene circuit that functions like a central processing unit. E. coli cells wereprogrammed to respond to a deleterious endogenous input, specifically, DNA-damagingstimuli such as UV radiation or mitomycin C. The gene circuit, which was chosen to be the Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins toggle switch described above (see Box 1), processes the incoming sensory information andflips from an ‘off' to an ‘on' state when a signal threshold is exceeded. Because the biosensor NIH-PA Author Manuscript has a decoupled, modular nature, it can be wired to any desired output, from the expression ofa standard fluorescent reporter to the activation of natural phenotypes, such as biofilmformation (e.g., via expression of traA gene) or cell suicide (e.g., via expression of ccdB gene).
Sometimes a single signal may be too general to characterize or define an environment. Forsuch situations, Anderson et al.25 devised a transcriptional AND gate that could be used tointegrate multiple environmental signals into a single genetic circuit (Box 2a), thusprogramming the desired level of biosensing specificity. Genetic biosensors of this sort couldbe useful for communicating the state of a specific microenvironment (e.g., in an industrialbioreactor) within a "sea" of environmental conditions such as temperature, metabolite levels,or cell density.
RNA molecules have a diverse and important set of cellular functions53. Noncoding RNAs cansplice and edit RNA, modify ribosomal RNA, catalyze biochemical reactions, and regulategene expression at the level of transcription or translation53–56. This last regulatory subset ofnoncoding RNAs57–59 is well-suited for rational design60 and, in particular, for biosensingapplications. Many regulatory RNA molecules are natural environmental sensors61–69, and NIH-PA Author Manuscript because of their ability to take on complex structures defined by their sequence, these moleculescan mediate diverse modular functions across distinct sequence domains. Riboswitches70, forinstance, bind specific small molecule ligands via aptamer domains and induce conformationalchanges in the 5′ untranslated region (UTR) of an mRNA, thus affecting its expression.
Aptamer domains that are modeled after riboswitches represent versatile and widely usedsensitive elements for RNA-based biosensing. The choice and number of aptamer domains canprovide control over specificity. Building an entire RNA-based biosensor typically requirescoupling the functionalities of an aptamer domain (the sensitive element) and a post-transcriptional regulatory domain (the transducer module) on a modular RNA moleculescaffold.
Antisense RNAs59, 71 are one such class of natural regulatory RNAs that can control geneexpression through post-transcriptional mechanisms. By linking a riboswitch aptamer to anantisense repressor on a single RNA molecule, Bayer and Smolke engineered trans-acting,ligand-responsive riboregulators of gene expression in S. cerevisiae11 (Box 2b). Binding ofthe aptamer to its ligand (e.g., the small molecule theophylline) induces a conformationalchange in the RNA sensor that either sequesters the antisense domain in a stable stem loop(ON switch) or liberates it to inhibit translation of an output gene reporter (OFF switch). As a NIH-PA Author Manuscript result of the cooperative dependence on both ligand and target mRNA, this biosensor exhibitsbinary-like switching at a threshold ligand concentration, similar to the genetic toggle design.
Importantly, this detection threshold can be adjusted by altering the RNA sequence and thusthe thermodynamic properties of the structure. In principle, the "antiswitch" framework ismodular, in other words, aptamers for different ligands and antisense stems targeting differentdownstream genes could be incorporated into the scaffold to devise new sensors. In practice,and as in many current synthetic biology designs, developing new sensors by aptamer andantisense replacement would likely involve re-screening compatible secondary structures tocreate functioning switches. In the future, this platform could be combined with rapid, invitro aptameric selection techniques72–75 for generating a suite of RNA biosensors that reporton the levels of various mRNA species and metabolites within a cell. However, here it shouldalso be noted that aptamers show specificity for a relatively biased ligand space, and as a resultaptamers for a target ligand cannot always be found.
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins Yet another method for transducing the sensory information captured by aptamer domains isto regulate translation through RNA self-cleavage69, 70. RNA cleavage is catalyzed by NIH-PA Author Manuscript ribozymes, some of which naturally possess aptameric domains and are responsive tometabolites69. Yen et al.76 took advantage of this natural framework and encoded ligand-sensitive ribozymes within the mRNA sequences of reporter genes. In the absence of its cognateligand, constitutive autocleavage of the reporter mRNA resulted in little or no signal. The RNAbiosensor is flipped when cognate ligand is present to inhibit the ribozyme's activity. Similarto the "antiswitch" framework (and with the same looming technical challenges), theseengineered RNAs could potentially be used as endogenous sensors for reporting on a varietyof intracellular species and metabolites.
The diversity and complexity of signal transduction pathways are vast. Factors such as thenature of the molecular interactions, the number of interconnected proteins in a cascade, andthe use of spatial mechanisms dictate which signals are transmitted, whether a signal isamplified or attenuated, and the dynamics of the response. Despite the multitude of factors andinteracting components, signal transduction pathways are, in the simplest sense, hierarchicalschemes based on sensitive elements and downstream transducer modules, and can berationalized as such for engineering protein-based biosensors.
NIH-PA Author Manuscript The primary sensitive element for most signal transduction pathways is the protein receptor.
Whereas environment-responsive promoters and RNA aptamers are typically identified fromnature or selected with high-throughput combinatorial methods, protein receptors can bedesigned de novo at the level of molecular interactions. Looger et al.77, for instance, deviseda computational method for redesigning natural protein receptors to bind new target ligands.
Starting with a "basis" of five proteins from the E. coli periplasmic binding protein (PBP)superfamily, the researchers replaced each of the wild-type (WT) ligands with a new, non-native target ligand and then used an algorithm to combinatorially explore all binding pocketresidue mutations and ligand-docking configurations. This procedure was used to predict novelreceptors for a carcinogen and explosive (trinitrotoluene, TNT), a medically-importantmetabolite (L-lactate), and a chemical associated with psychiatric conditions (serotonin). Thepredicted receptor designs were experimentally confirmed to be strong and specific in vitrosensors, as well as in vivo cell-based biosensors.
Protein receptors, such as the ones discussed above, are typically membrane-bound; theytrigger protein signaling cascades that ultimately result in a cellular response. However, severalsynthetic methods can be employed to transmit captured sensory information in a tunable anddesirable manner. Skerker et al.78 rationally rewired the transmission of information through NIH-PA Author Manuscript two-component systems, by identifying rules governing the specificity of a histidine kinase toits cognate response regulator. Alternatively, engineered protein scaffolds can be designed tophysically recruit pathway modulators and synthetically reshape the dynamical responsebehaviour of a system79 (Box 2c). This constitutes a modular method for programming protein-based biosensors to have any desired response, including accelerated, delayed or ultrasensitiveresponses, to upstream signals.
Combining synthetic transcriptional, translational, and post-translational circuits into hybridsolutions, and harnessing desired characteristics from each, could lead to the creation of cell-based biosensors that are as robust as those of natural organisms. Using a synthetic hybridapproach, Voigt and colleagues developed E. coli-based optical sensors80–82. A syntheticsensor kinase was engineered to allow cells to identify and report the presence of red light. Asa result, a bacterial lawn of the engineered cells could faithfully "print" a projected image, in Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins the biological equivalent of photographic film. Specifically, a membrane-bound photoreceptorfrom cyanobacteria was fused to an E. coli intracellular histidine kinase to induce light- NIH-PA Author Manuscript dependent changes in gene expression80 (Box 2d). In a clever example of its use, the bacterialoptical sensor was applied in image edge detection82. In this case, by wiring the optical sensorto transcriptional circuits that perform cell-cell communication (quorum sensing system fromVibrio fischeri) and logical functions (Box 2d), the researchers programmed only the cells thatreceive light and directly neighbor cells that do not receive light to produce a pigment, thustracing the edges of a projected image. Indeed, this work shows that complex behaviour canemerge from properly wiring together smaller genetic programs, with unique advantages toreal-world applications.
Human health is afflicted by new and old foes, including emergent drug-resistant microbes,cancer, and obesity. All the while, progress in medicine is faced with challenges at each stageof the therapeutic spectrum, ranging from the drying up of pharmaceutical pipelines to limitedglobal access to viable medicines. In a relatively short amount of time, synthetic biology hasmade promising strides in reshaping and streamlining this spectrum (Box 3). Indeed, therational and model-guided construction of biological parts is enabling new therapeuticplatforms, from the identification of disease mechanisms and druggable targets to theproduction and delivery of small molecules.
NIH-PA Author Manuscript Synthetic biology's impact on the therapeutic spectrum
Drug discovery. A synthetic mammalian gene circuit that enabled drug discovery
for anti-tuberculosis compounds90. The antibiotic ethionamide is rendered
cytotoxic to M. tuberculosis by the enzyme EthA within infected cells. Because
EthA is natively repressed by EthR, resistance to ethionamide treatment is
common. In the gene circuit, a fusion of EthR and the mammalian transactivator
VP16 binds a minimal promoter with a synthetic EthR operator site, and activates
expression of a reporter gene SEAP (human placental secreted alkaline
phosphatase). This platform allows for the rapid screening of EthR inhibitors
within mammalian cells.
Treatment & delivery. A synthetic mammalian genetic switch for tight, tunable,
and reversible control of a desired gene for therapeutic or gene delivery
applications. In the OFF configuration, expression of the gene of interest (green)
is repressed both at the levels of transcription and translation. Constitutively
NIH-PA Author Manuscript expressed LacI repressor (red) binds to the lac operator sites in the transgenemodule of the gene of interest, thus repressing its transcription. Any transcriptionalleakage is repressed at the level of translation by an interfering RNA (blue), whichtargets the gene's 3′ UTR. The system is switched ON by addition of IPTG, whichbinds LacI repressor proteins and consequently relieves both forms of repression.
Drug production & access. The discovery of drugs does not always translate to
the people who need them the most, because drug production processes can be
difficult and costly. Antibiotics are industrially produced from microbes and fungi,
and are therefore widespread and cheap. Conversely, many other drugs are isolated
from hosts that are not as amenable to large-scale production and are therefore
costly and in short supply. Such drugs include the antimalaria drug artemisinin and
the anticancer drug taxol. Fortunately, global access to drugs is being enabled by
hybrid synthetic biology and metabolic engineering strategies for the microbial
production of rare natural products. In the case of artemisinin, there exist two
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins biosynthetic pathways for the synthesis of the universal precursors to allisoprenoids, the large and diverse family of natural products of which artemisinin NIH-PA Author Manuscript is a member. The native pathway found in E. coli (deoxyxylulose 5-phosphate,DXP, pathway) has been difficult to optimize, so the researchers syntheticallyconstructed and tested in piece-wise fashion (e.g., "Top" and "Bottom" operons)the entire S. cerevisiae mevalonate-dependent (MEV) pathway in E. coli. Theresearchers initially used a simple, orthogonal host platform to construct, debug,and optimize a large metabolic pathway105. They then linked the optimizedheterologous pathway to a codon-optimized form of the plant terpene synthase,ADS, in order to funnel metabolic production to the specific terpene precursor toartemisinin. This work allowed them to build a full, optimized solution that couldbe ultimately and seamlessly deployed back into S. cerevisiae for cost-effectivesynthesis and purification of industrial quantities of the immediate drug precursorof artemisinin106. Pathway intermediate abbreviations: FPP, farnesylpyrophosphate.
An electrical engineer is likely to prototype portions of a circuit on a breadboard before printingit as an entire integrated circuit. This allows for the rigorous testing of submodules in an NIH-PA Author Manuscript isolated, well-characterized environment. Similarly, synthetic biology provides a frameworkfor synthetically reconstructing natural biological systems to explore how pathologicalbehaviours may emerge. This strategy was employed to lend mechanistic insight into a primaryimmunodeficiency, known as agammaglobulinemia, in which patients cannot generate matureB cells and as a result are unable to properly fight infections83. The researchers developed asynthetic testbed by systematically reconstructing the various components of the human B cellantigen receptor (BCR) signaling pathway in an orthogonal environment. This allowed themto identify network topology features that trigger BCR signaling and assembly. A rare mutationin the Igβ-encoding gene, identified in one patient, was then introduced into the syntheticsystem and shown to abolish assembly of the BCR on the cell surface, thereby linking thisfaulty pathway component with disease onset. Pathogenic viral genomes can similarly bereconstructed to study the molecular underpinnings of infectious disease pandemics. Forinstance, synthetic reconstruction of the Severe Acquired Respiratory Syndrome (SARS)coronavirus84 and the 1918 Spanish influenza virus85 helped to identify genetic mutations thatmay have conferred human tropism and increased virulence.
Drug Target Identification
NIH-PA Author Manuscript Building up synthetic pathways and systems from individual parts is one way of identifyingdisease mechanisms and therapeutic targets. Another is to deploy synthetic biology devices tosystematically probe the function of individual components of a natural pathway. Our group,for instance, has engineered modular riboregulators that can be used to tune the expression ofa toxic protein or any gene within a biological network86. To achieve post-transcriptionalcontrol over a target gene, the mRNA sequence of its 5′-UTR was designed to form a hairpinstructure that sequesters the ribosomal binding site (RBS) and prevents ribosome access to it.
Translational repression of this cis-repressed mRNA could then be alleviated by anindependently regulated trans-activating RNA that targets the stem-loop for unfolding.
Engineered riboregulators were used in a subsequent study to tightly regulate the expressionof CcdB, a toxic bacterial protein that inhibits DNA gyrase, so as to gain a better understandingof the sequence of events leading to induced bacterial cell death87. These synthetic biologystudies, in conjunction with systems biology studies of quinolones (antibiotics that inhibitgyrase)87, led to the discovery that all major classes of bactericidal antibiotics induce a commonoxidative damage cellular death pathway88, 89. This work provided new insights into how Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins bacteria respond to lethal stimuli, and paved the way for the development of more effectiveantibacterial therapies.
NIH-PA Author Manuscript Once a faulty pathway component or target is identified, whole-cell screening assays can bedesigned using synthetic biology strategies for drug discovery. As a demonstration of thisapproach, Fussenegger and colleagues90 developed a synthetic platform for screening smallmolecules that could potentiate a Mycobacterium tuberculosis antibiotic (Box 3a).
Ethionamide, currently the last line of defense in the treatment of multidrug-resistanttuberculosis, depends on activation by the M. tuberculosis enzyme EthA for efficacy. Due totranscriptional repression of ethA by the protein EthR, however, ethionamide-based therapy isoften rendered ineffective. To address this problem, the researchers designed a syntheticmammalian gene circuit, featuring an EthR-based transactivator of a reporter gene, and usedit to screen for and identify EthR inhibitors that could abrogate resistance to ethionamide.
Importantly, because the system is a cell-based assay, it intrinsically enriches for inhibitorsthat are nontoxic and membrane-permeable to mammalian cells, which are key drug criteriaas M. tuberculosis is an intracellular pathogen. This framework, in which drug discovery isapplied to whole cells that have been engineered with circuits that highlight a pathogenicmechanism, could be extended to other diseases and phenotypes.
NIH-PA Author Manuscript Synthetic biology devices have additionally been developed to serve as therapies themselves.
Entire engineered viruses and organisms can be programmed to target specific pathogenicagents and pathological mechanisms. In two separate studies91, 92, for instance, engineeredbacteriophages were deployed to combat antibiotic-resistant bacteria, by endowing them withgenetic mechanisms that target and thwart bacteria's antibiotic evasion techniques. The firststudy was prompted by the observation that biofilms, in which bacteria are encapsulated in anextracellular matrix, have inherent resistance to antimicrobial therapies and are implicatedsources of persistent infections. To more effectively penetrate this protective environment, T7phage was engineered to express the biofilm matrix-degrading enzyme dispersin B (DspB)upon infection91. The two-pronged attack of phage-induced lysis fueling the creation andspread of matrix-degrading enzyme resulted in 99.997% removal of biofilm bacterial cells.
In the second study92, it was hypothesized that inhibition of certain bacterial genetic programscould help current antibiotic therapies achieve more effective activity. In this case,bacteriophages were deliberately designed to be non-lethal so as not to elicit resistancemechanisms; instead, non-lytic M13 phage was used to suppress the bacterial SOS DNA NIH-PA Author Manuscript damage response by overexpression of its repressor, lexA3. The engineered bacteriophagesignificantly enhanced killing by three major classes of antibiotics in traditional cell cultureand in E. coli-infected mice, potentiated killing of antibiotic-resistant bacteria, and importantlyreduced the incidence of antibiotic-induced resistant cells.
Synthetically engineered viruses and organisms that are able to sense and link their therapeuticactivity to pathological cues may be useful in the treatment of cancer, where current therapiesoften indiscriminately attack tumors and normal tissues. Adenoviruses, for instance, wereprogrammed to couple their replication to the state of the p53 pathway in human cells93. Normalp53 production would result in inhibition of a critical viral replication component, whereas adefunct p53 pathway, which is characteristic of tumor cells, would allow viral replication andcell killing. In another demonstration of translational synthetic biology applied to cancertherapy, Voigt and colleagues94 developed cancer-targeting bacteria and linked their ability toinvade the cancer cells to specific environmental signals. Constitutive expression of theheterologous inv gene (from Yersinia pseudotuberculosis) can induce E. coli cells to invade Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins both normal and cancer human cell lines. So, to preferentially invade cancer cells, theresearchers placed inv under the control of transcriptional operons that are activated by NIH-PA Author Manuscript environmental signals specific to the tumor microenvironment. These engineered bacteriacould be made to carry or synthesize cancer therapies for the treatment of tumors.
In addition to engineered therapeutic organisms, synthetic circuits and pathways can be usedfor the controlled delivery of drugs as well as for gene and metabolic therapy. In some cases,sophisticated, kinetic control over drug release in the body may yield therapeutic advantagesand reduce undesired side effects. Most hormones in the body are released in time-dependentpulses. Glucocorticoid secretion, for instance, has a circadian and ultradian pattern of release,with important transcriptional consequences for glucocorticoid-responsive cells95. Faithfullymimicking these patterns in the administration of synthetic hormones to patients withglucocorticoid-responsive diseases, such as rheumatoid arthritis, may decrease known sideeffects and improve therapeutic response95. Periodic synthesis and release of biologic drugscan be autonomously achieved with synthetic oscillator circuits9, 10, 20–22 or programmedtime-delay circuits96. In other cases, one may wish to place a limit on the amount of drugreleased by programming the synthetic system to self-destruct after a defined number of cellcycles or drug release pulses. Our group has recently developed two variants of a syntheticgene counter14 that could be adapted for such an application.
NIH-PA Author Manuscript Gene therapy is beginning to make some promising advances in clinical areas where traditionaldrug therapy is ineffective, such as in the treatment of many hereditary and metabolic diseases.
Synthetic circuits offer a more controlled approach to gene therapy, such as the ability todynamically silence, activate, and tune the expression of desired genes. In one suchexample12, a genetic switch was developed in mammalian cells that couples transcriptionalrepressor proteins and an RNA interference (RNAi) module for tight, tunable, and reversiblecontrol over the expression of desired genes (Box 3b). This system would be particularly usefulin gene silencing applications, as it was shown to yield > 99% repression of a target gene.
Additionally, the construction of non-native pathways offers a unique and versatile approachto gene therapy, such as for the treatment of metabolic disorders. Operating at the interface ofsynthetic biology and metabolic engineering, for instance, Liao and colleagues97 recentlyintroduced the glyoxylate shunt pathway into mammalian liver cells and mice to explore itseffects on fatty acid metabolism and, more broadly, whole-body metabolism. Remarkably, theresearchers found that when transplanted into mammals the shunt actually increased fatty acidoxidation, evidently by creating an alternative cycle. Furthermore, mice expressing the shuntshowed resistance to diet-induced obesity when placed on a high-fat diet, with corresponding NIH-PA Author Manuscript decreases in total fat mass, plasma triglycerides, and cholesterol levels. This work offers a newsynthetic biology model for studying metabolic networks and disorders, and for developingtreatments for the increasing problem of obesity.
The discovery of drugs and effective treatments may not quickly or ever translate to the peoplewho need them the most, because drug production processes can be difficult and costly. Asreviewed below, here synthetic biology is bringing more cost-effective manufacturingprocesses to the production of such rare and costly drugs (Box 3c).
BIOFUELS, PHARMACEUTICALS AND BIOMATERIALS
Recent excitement surrounding the production of biofuels, pharmaceuticals and biomaterialsfrom engineered microorganisms is matched by the challenges that loom in bringing thesetechnologies to production scale and quality. The most widely used biofuel is ethanol produced Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins from corn or sugar cane98; however, the heavy agricultural burden combined with thesuboptimal fuel properties of ethanol make this approach to biofuels problematic and limited.
NIH-PA Author Manuscript Microorganisms engineered with optimized biosynthetic pathways to efficiently convertbiomass into biofuels are an alternative and promising source of renewable energy. Thesestrategies will succeed only if their production costs can be made to compete with, or even out-compete, current fuel production costs. Similarly, there are many drugs whose productionprocesses preclude their capacity for a wider and more cost-effective therapeutic reach. Newsynthetic biology tools would also greatly advance the microbial production of biomaterialsand the development of altogether novel materials.
Constructing biosynthetic pathways
One of the first design decisions when engineering for biofuels, drugs, or biomaterials is whichbiosynthetic pathway(s) to construct around and, further to that, which host organism to use.
Typically, this decision originates with searching for organisms innately capable of achievingsome desired biosynthetic activity or phenotype99. For biofuel production, for instance,microorganisms have evolved to be proficient in converting lignocellulosic material to ethanol,biobutanol, and other biofuels. These native isolates possess unique catabolic activity,heightened tolerances for toxic materials, and a host of enzymes designed to break down thelignocellulosic components. Unfortunately, these highly desired properties exist in pathways NIH-PA Author Manuscript that are tightly regulated according to the host's evolved needs and therefore may not be suitablein their native state for production scale. A longstanding challenge in metabolic and geneticengineering is determining whether to improve the isolate host's production capacity orwhether to transplant the desired genes/pathways into an industrial model host, such as E.
coli
or S. cerevisiae; these important considerations and tradeoffs are reviewed elsewhere99.
The example of the microbial production of biobutanol, a higher energy density alternative toethanol, provides a useful glimpse into these design tradeoffs. Butanol is converted naturallyfrom acetyl-coenzyme A (acetyl-coA) by Clostridium acetobutylicum100. However, it isproduced in low yields and as a mixture with acetone and ethanol, thus requiring significantcellular engineering of a strain for which most standard molecular biology techniques do notapply101, 102. On the other hand, importing the biosynthetic genes into an industrial microbialhost can lead to metabolic imbalances103. In an altogether different approach, Liao andcolleagues104 bypassed standard fermentation pathways and recognized that a broad set of the2-keto acid intermediates of E. coli amino acid biosynthesis could be synthetically shunted toachieve high-yield production of butanol and other higher alcohols in two enzymatic steps.
Complementary to efforts in traditional metabolic and genetic engineering is the use of NIH-PA Author Manuscript engineering principles for constructing functional, predictable, and non-native biologicalpathways de novo to control and improve microbial production. In an exemplary illustrationof this, Keasling and colleagues engineered the microbial production of precursors to theantimalarial drug artemisinin to industrial levels105, 106 (Box 3c). There are now many suchexamples of the successful application of synthetic approaches to biosynthetic pathwayconstruction, for instance in the microbial production of fatty-acid-derived fuels and chemicals(e.g., fatty esters, fatty alcohols, and waxes)107, methyl halide-derived fuels and chemicals108, polyketide synthases that make cholesterol-lowering drugs109, and polyketides made frommegaenzymes that are encoded by very large synthetic gene clusters110.
Optimizing pathway flux
Once biosynthetic pathways are constructed, the expression levels of all the components needto be orchestrated to optimize metabolic flux and achieve high product titers. A standardapproach is to drive the expression of pathway components with strong and exogenously Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins tunable promoters, such as the PLtet, PLlac, and PBAD promoters from the tet, lac, and araoperons of E. coli, respectively. To this end, there are ongoing synthetic biology efforts to NIH-PA Author Manuscript create and characterize more reusable, biological control elements based on promoters forpredictably tuning expression levels111, 112. Further to this, synthetic biologists have deviseda number of alternative methods for biological pathway balance, ranging from reconfiguringnetwork connectivity to fine-tuning individual components. A richer discussion of these topics,including the fine-tuning of parts, model-guided approaches, and the development of next-generation interoperable parts, is presented elsewhere113. In Box 4, we detail a few suchsynthetic biology strategies that specifically pertain to the optimization of metabolic pathwayflux. These strategies span a spectrum from those driven by evolutionary techniques to thosedriven by rational design and in silico models, to those that combine both approaches.
Synthetic biology approaches to controlling metabolic flux
Evolutionary strategies. In the production of artemisinin precursors, the native
E. coli isoprenoid pathway (DXP) was eschewed in favor of a heterologous
pathway so as to circumvent the complex regulatory control imposed by the host
(see Box 3c). In an alternative method of relieving regulatory control over the large
number of DXP pathway components, Wang et al.121 rapidly diversified and
NIH-PA Author Manuscript evolved, and thus optimized, the native DXP biosynthetic pathway in E. coli. Theresearchers developed a rapid, automated method for the in vivodirected evolutionof pathways, which they termed multiplex automated genome engineering(MAGE); they then applied it to evolve the translational efficiencies of DXPpathway components to achieve maximal lycopene production. Specifically, cellswere subjected to cycles of genetic modifications (via oligo-mediated allelicreplacement) in an automated fashion to explore sufficient genomic diversity foroptimizing biosynthetic pathways in laboratory time scales. Pathway intermediateabbreviations: G3P, glyceraldehydes 3-phosphate; DXP, 1-deoxy-D-xylulose 5-phosphate; IPP, isopentenyl diphosphate; DMAPP, dimethylallyl diphosphate;FPP, farnesyl diphosphate.
Rational design. At the other end of the spectrum are strategies that rely on
quantitative models and blueprints for the rational design of optimized networks
and pathways. Typically, a component of interest (e.g., an engineered promoter,
P*, or RBS sequence, RBS*) will be built into a simple test network. The network
and its input/output data will then be fed into a model, which attempts to determine
a parameter set that optimally describes the component's dynamics within the
NIH-PA Author Manuscript framework of the model. Finally, the optimized parameter set will be used toforward-engineer altogether new networks and components. For example,stochastic biochemical models have been developed to capture the expressiondynamics of synthetically engineered promoters; these models were subsequentlyemployed to predict the correct, in vivo behaviour of different and more complexgene networks built from the modeled components122, 123. Similarly, at the levelof translation, thermodynamic models that predict the relative translationalinitiation rates of proteins can be used to rationally forward-engineer syntheticRBS sequences to give desired expression levels124. Such techniques harnessmodeled genetic parameters (transcriptional or translational) to predict the levelof expression of proteins and enzymes within a network.
Hybrid approaches. In a hybrid rational-combinatorial approach, Dueber et al.
125 hypothesized that metabolic flux could be controlled by spatially recruiting the
enzymes of a desired biosynthetic pathway using synthetic protein scaffolds. To
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins construct the enzyme scaffolding, the researchers tethered protein-proteininteraction domains from metazoan signaling proteins (e.g., GBD, SH3, PDZ NIH-PA Author Manuscript domains) that recognize and bind cognate peptides fused to the enzymes to berecruited (atoB, HMGS, HMGR). By varying the number of repeats of aninteraction domain, they could additionally control the stoichiometry of theenzymes recruited to the complex. Once again, using the heterologous MEVpathway in E. coli as a model, they combinatorially (albeit, at a significantlysmaller scale) optimized the stoichiometry of the three enzymes responsible forproducing mevalonate from acetyl-coA. Finally, they showed that the optimizedsynthetic scaffold could significantly increase product titer while also reducing themetabolic load on the host; in other words, their high product titers did not requirethe overexpression of biosynthetic enzymes in the cell. Gene symbolabbreviations: atoB, acetoacetyl-CoA thiolase from E. coli; HMGS, HMG-CoAsynthase from S. cerevisiae; HMGR, HMG-CoA reductase from S. cerevisiae.
Programming novel functionality and materials
Beyond facilitating metabolic tasks, synthetic systems can infuse novel functionality intoengineered organisms for production purposes or for building new materials. Early work in thefield laid the groundwork for constructing basic circuits that could sense and process signals, NIH-PA Author Manuscript perform logic operations, and actuate biological responses114. Wiring these modules togetherto bring about reliable, higher-order functionality is one of synthetic biology's next majorgoals113, and an important application of this objective is the layering of "smart" controlmechanisms over metabolic engineering. For instance, circuits designed to sense the bioreactorenvironment and shift metabolic phases accordingly would further improve biofuel production.
Alternatively, autonomous timing circuits could be used to shut down metabolic processesafter a prescribed duration of time. Biological timers of this sort have been developed usinggenetic toggle switches that had been deliberately rendered imbalanced through model-guidedpromoter engineering112. These genetic timers were used to program the time-dependentflocculation of yeast cells to facilitate the separation of cells from, for instance, the alcoholproduced in industrial fermentation processes.
Synthetic control systems can also be employed to extract and purify the synthesized product.
This is particularly important in the production of recombinant proteins, bioplastics, and otherlarge biomaterials, whose high titer can lead to intracellular accumulation, the formation ofinclusion bodies, and cell toxicity. For instance, to export recombinant spider silk monomers,Widmaier et al.115 searched for a secretion system that would enable efficient andindiscriminate secretion of proteins through both bacterial membranes. The Salmonella type NIH-PA Author Manuscript III secretion system (T3SS) not only fulfills these criteria but also possesses a natural regulatoryscheme that ties expression of the protein to be secreted to the secretion capacity of the cell;as a result, the desired protein is only expressed when sufficient secretion complexes have beenbuilt. The researchers needed only to engineer a control circuit that hitches their heterologousgenes to the innate genetic machinery for environmental sensing and secretion commitment toobtain superior secretion rates of recombinant silk protein.
Finally, there is an emerging branch of synthetic biology that seeks to program coordinatedbehaviour in populations of cells, which could lead to the fabrication of novel biomaterials fora variety of applications. The engineering of synthetic multicellular systems is typicallyachieved with cell-cell communication and associated intracellular signal processing modules,as was elegantly employed by Hasty and colleagues to bring about synchronized oscillationsin a population of bacterial cells23. Weiss and colleagues have similarly done pioneering workin building biomolecular signal processing architectures that are able to filter communication Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins signals originating from ‘sender' cells24, 31. These systems, which can be programmed to formintricate multicellular patterns from a solid-phase cellular lawn, would aid the development of NIH-PA Author Manuscript fabrication-free scaffolds for tissue engineering.
FUTURE CHALLENGES AND CONCLUSIONS
The future of translational synthetic biology is dependent on and limited by the developmentof reliable means for connecting smaller functional circuits to realize higher-order networkswith predictable behaviours. In a previous perspective113, we outlined four research effortsaimed at improving and accelerating the overall design cycle and allowing more seamlessintegration of biological circuitry (Box 5).
Recommendations for improving the synthetic biology design cycle
Scaling up to larger and more complex biological systems while simultaneouslyminimizing interference among parts will require an expanded synthetic biologytoolkit and, in particular, libraries of interoperable parts. Eukaryotic systems arefertile grounds for discovering such parts, as many synthetic biology devices arebased on a small repertoire of prokaryotic regulatory elements.
NIH-PA Author Manuscript Modeling and fine-tuning of synthetic networks should be emphasized,particularly as the network size and complexity increases. This will facilitateproper matching of input/output behaviours (i.e., transfer functions) when distinctmodules are connected.
There is a need to develop new probes and high-throughput methods for the invivo measurement of circuit dynamics in order to rapidly characterize parts anddebug networks.
The development of cellular testing platforms are needed to quicken the pace ofidentifying problematic network nodes and ease failure-prone jumps associatedwith either building a more complex network or deploying a network in a morecomplex organism. These testing platforms may be cells engineered to haveminimal genomes126–129 or lower model organisms that have been equipped withspecific machinery from higher organisms.
Beyond the challenge of improving the design cycle, applied synthetic biology would benefitfrom once again summoning the original inspiration of biocomputing. The ability to program NIH-PA Author Manuscript higher-level decision-making into synthetic networks would yield more robust and dynamicorganisms, including ones that can accomplish many tasks simultaneously. Furthermore, asadaptive and predictive behaviours are naturally present in all organisms (including microbes)116, 117, synthetic learning networks built from genetic and biological parts118, 119 would infuseengineered organisms with more sophisticated automation for biosensing and relatedapplications.
Finally, the majority of synthetic biology is currently practiced in microbes. Yet, many of themost pressing problems, and in particular those of human health, are inherently problems withmammalian systems. Therefore, a more concerted effort towards advancing mammaliansynthetic biology is critical to next-generation therapeutic solutions, including engineeringsynthetic gene networks for stem cell generation and differentiation.
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins By addressing such challenges, we will be limited, not by the technicalities of construction orthe robustness of synthetic gene networks, but only by the imagination of researchers and the NIH-PA Author Manuscript number of societal problems and applications for which synthetic biology can resolve.
We thank members of the Collins Lab for helpful discussions and K.M. Flynn for help with artwork. We also thankthe Howard Hughes Medical Institute and the NIH Director's Pioneer Award Program for their financial support.
A device that is used to store information about the current state of asystem A circuit or device used to generate pulses. A biological version hasbeen implemented in a multicellular bacterial system, whereby receivercells respond to a chemical signal with a transient burst of geneexpression, whose amplitude and duration depends on the distance fromthe sender cells Digital logic gates A digital logic gate implements Boolean logic (such as AND, OR, NOT)on one or more logic inputs to produce a single logic output. Electronic NIH-PA Author Manuscript logic gates are implemented using diodes and transistors and operateon input voltages or currents, whereas biological logic gates operate oncellular molecules (chemical or biological) A digital logic gate that implements logical NOR, or the negation of theOR operator. It produces a HIGH output (1) only if both inputs to thegate are LOW (0) An algorithm or device for removing or enhancing parts or frequencycomponents from a signal Refers to the capacity of a system or component to functionindependently of context Promoters that directly transduce environmental signals (e.g., heavy metal ions, hormones, chemicals, temperature) that are captured by their associated, sensory transcription factors A cell-to-cell communication mechanism in many species of bacteria,whereby cells measure their local density (via the accumulation of a NIH-PA Author Manuscript signaling molecule) and subsequently coordinate gene expression Oligonucleic acids that bind to a specific target molecule, such as asmall molecule, protein, or nucleic acid. Nucleic acid aptamers aretypically developed through in vitro selection schemes but are alsofound naturally (e.g., RNA aptamers in riboswitches) Antisense RNAs bind segments of mRNA in trans to inhibit translation Among the simplest types of signal transduction pathways. In bacteria, they consist of two domains, a membrane-bound histidine kinase(sensitive element) that senses a specific environmental stimulus andits cognate response regulator (transducer domain) that triggers acellular response A cellular environment or host into which genetic material is transplanted to avoid undesired, native host interference or regulation.
Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins Orthogonal hosts are often organisms with sufficient evolutionary NIH-PA Author Manuscript distance from the native host Small regulatory RNAs found in prokaryotes that can activate or repressgene expression by binding segments of mRNA in trans. They aretypically expressed in response to an environmental signaling event A type II DNA topoisomerase that catalyzes the ATP-dependentsupercoiling of closed-circular dsDNA by strand breakage and rejoiningreactions. Control of chromosomal topological transitions is essentialfor DNA replication and transcription in bacteria, making gyrase aneffective target for antimicrobial agents (e.g., the quinolone class ofantibiotics) The surface-associated communities of bacterial cells encapsulated inan extracellular polymeric substances (EPS) matrix. Biofilms are anantibiotic-resistant mode of microbial life found in natural andindustrial settings Periods or cycles that are repeated throughout a 24-hour circadian day A two-enzyme metabolic pathway unique to bacteria and plants that is NIH-PA Author Manuscript activated when sugars are not readily available. This pathway divertsthe tricarboxylic acid (TCA) cycle so that fatty acids are not completelyoxidized but, instead, converted into carbon energy sources Rate of flow of metabolites through a metabolic pathway, which isregulated by the enzymes in the pathway A specific form of cell aggregation in yeast triggered by certainenvironmental conditions, such as the absence of sugars, e.g., once thesugar in a beer brew has been fermented into ethanol 1. Monod J, Jacob F. General conclusions: telenomic mechanisms in cellular metabolism, growth, and differentiation. Cold Spring Harb Symp Quant Biol 1961;26:389–401. [PubMed: 14475415] 2. Glass L, Kauffman SA. The logical analysis of continuous, non-linear biochemical control networks.
J Theor Biol 1973;39:103–29. [PubMed: 4741704] 3. Savageau MA. Comparison of classical and autogenous systems of regulation in inducible operons.
Nature 1974;252:546–9. [PubMed: 4431516] NIH-PA Author Manuscript 4. Kauffman S. The large scale structure and dynamics of gene control circuits: an ensemble approach.
J Theor Biol 1974;44:167–90. [PubMed: 4595774] 5. Glass L. Classification of biological networks by their qualitative dynamics. J Theor Biol 1975;54:85– 107. [PubMed: 1202295] 6. McAdams HH, Arkin A. Towards a circuit engineering discipline. Curr Biol 2000;10:R318–20.
[PubMed: 10801411] 7. McAdams HH, Shapiro L. Circuit simulation of genetic networks. Science 1995;269:650–6. [PubMed: 8. Gardner TS, Cantor CR, Collins JJ. Construction of a genetic toggle switch in Escherichia coli. Nature 2000;403:339–42. [PubMed: 10659857] 9*. Elowitz MB, Leibler S. A synthetic oscillatory network of transcriptional regulators. Nature 2000;403:335–8. Two seminal papers demonstrating synthetic biology's first devices--the genetictoggle switch and the repressilator--and establishing the engineeing-based methodology forconstructing sophisticated, dynamical behaviours in biological systems from simple regulatoryelements. [PubMed: 10659856] Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 10. Atkinson MR, Savageau MA, Myers JT, Ninfa AJ. Development of genetic circuitry exhibiting toggle switch or oscillatory behavior in Escherichia coli. Cell 2003;113:597–607. [PubMed: 12787501] NIH-PA Author Manuscript 11*. Bayer TS, Smolke CD. Programmable ligand-controlled riboregulators of eukaryotic gene expression. Nat Biotechnol 2005;23:337–43. Establishes an RNA scaffold for the ligand-dependenton/off switching of gene expression. [PubMed: 15723047] 12. Deans TL, Cantor CR, Collins JJ. A tunable genetic switch based on RNAi and repressor proteins for regulating gene expression in mammalian cells. Cell 2007;130:363–72. [PubMed: 17662949] 13*. Dueber JE, Yeh BJ, Chak K, Lim WA. Reprogramming control of an allosteric signaling switch through modular recombination. Science 2003;301:1904–8. Provides a modular framework forprogramming the input/output behaviour of eukaryotic signaling protein circuits, and demonstratessynthetic switch proteins with a rich set of gating behaviours. [PubMed: 14512628] 14. Friedland AE, et al. Synthetic gene networks that count. Science 2009;324:1199–202. [PubMed: 15. Ham TS, Lee SK, Keasling JD, Arkin AP. A tightly regulated inducible expression system utilizing the fim inversion recombination switch. Biotechnol Bioeng 2006;94:1–4. [PubMed: 16534780] 16. Ham TS, Lee SK, Keasling JD, Arkin AP. Design and construction of a double inversion recombination switch for heritable sequential genetic memory. PLoS ONE 2008;3:e2815. [PubMed:18665232] 17. Kramer BP, Fussenegger M. Hysteresis in a synthetic mammalian gene network. Proc Natl Acad Sci U S A 2005;102:9517–22. [PubMed: 15972812] 18. Kramer BP, et al. An engineered epigenetic transgene switch in mammalian cells. Nat Biotechnol NIH-PA Author Manuscript 2004;22:867–70. [PubMed: 15184906] 19. Ajo-Franklin CM, et al. Rational design of memory in eukaryotic cells. Genes Dev 2007;21:2271–6.
[PubMed: 17875664] 20. Fung E, et al. A synthetic gene-metabolic oscillator. Nature 2005;435:118–22. [PubMed: 15875027] 21. Stricker J, et al. A fast, robust and tunable synthetic gene oscillator. Nature 2008;456:516–9. [PubMed: 22. Tigges M, Marquez-Lago TT, Stelling J, Fussenegger M. A tunable synthetic mammalian oscillator.
Nature 2009;457:309–12. [PubMed: 19148099] 23. Danino T, Mondragon-Palomino O, Tsimring L, Hasty J. A synchronized quorum of genetic clocks.
Nature 2010;463:326–30. [PubMed: 20090747] 24. Basu S, Mehreja R, Thiberge S, Chen MT, Weiss R. Spatiotemporal control of gene expression with pulse-generating networks. Proc Natl Acad Sci U S A 2004;101:6355–60. [PubMed: 15096621] 25. Anderson JC, Voigt CA, Arkin AP. Environmental signal integration by a modular AND gate. Mol Syst Biol 2007;3:133. [PubMed: 17700541] 26. Guet CC, Elowitz MB, Hsing W, Leibler S. Combinatorial synthesis of genetic networks. Science 2002;296:1466–70. [PubMed: 12029133] 27. Rackham O, Chin JW. Cellular logic with orthogonal ribosomes. J Am Chem Soc 2005;127:17584– NIH-PA Author Manuscript 5. [PubMed: 16351070] 28. Rinaudo K, et al. A universal RNAi-based logic evaluator that operates in mammalian cells. Nat Biotechnol 2007;25:795–801. [PubMed: 17515909] 29. Stojanovic MN, Stefanovic D. A deoxyribozyme-based molecular automaton. Nat Biotechnol 2003;21:1069–74. [PubMed: 12923549] 30. Win MN, Smolke CD. Higher-order cellular information processing with synthetic RNA devices.
Science 2008;322:456–60. [PubMed: 18927397] 31*. Basu S, Gerchman Y, Collins CH, Arnold FH, Weiss R. A synthetic multicellular system for programmed pattern formation. Nature 2005;434:1130–4. An excellent demonstration of syntheticcontrol over a population of cells. By splitting the Vibrio fischeri quorum sensing circuit between‘sender' and ‘receiver' cells, bacteria were programmed to communicate in order to generateintricate, two-dimensional patterns. [PubMed: 15858574] 32. Hooshangi S, Thiberge S, Weiss R. Ultrasensitivity and noise propagation in a synthetic transcriptional cascade. Proc Natl Acad Sci U S A 2005;102:3581–6. [PubMed: 15738412] Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 33. Sohka T, et al. An externally tunable bacterial band-pass filter. Proc Natl Acad Sci U S A 2009;106:10135–40. [PubMed: 19502423] NIH-PA Author Manuscript 34. Kobayashi H, et al. Programmable cells: interfacing natural and engineered gene networks. Proc Natl Acad Sci U S A 2004;101:8414–9. [PubMed: 15159530] 35. You L, Cox RS 3rd, Weiss R, Arnold FH. Programmed population control by cell-cell communication and regulated killing. Nature 2004;428:868–71. [PubMed: 15064770] 36. Mukherji S, van Oudenaarden A. Synthetic biology: understanding biological design from synthetic circuits. Nat Rev Genet 2009;10:859–71. [PubMed: 19898500] 37. Brown M, et al. lac repressor can regulate expression from a hybrid SV40 early promoter containing a lac operator in animal cells. Cell 1987;49:603–12. [PubMed: 3034429] 38. Deuschle U, et al. Regulated expression of foreign genes in mammalian cells under the control of coliphage T3 RNA polymerase and lac repressor. Proc Natl Acad Sci U S A 1989;86:5400–4.
[PubMed: 2664783] 39. Hu MC, Davidson N. The inducible lac operator-repressor system is functional in mammalian cells.
Cell 1987;48:555–66. [PubMed: 3028641] 40. Lutz R, Bujard H. Independent and tight regulation of transcriptional units in Escherichia coli via the LacR/O, the TetR/O and AraC/I1-I2 regulatory elements. Nucleic Acids Res 1997;25:1203–10.
[PubMed: 9092630] 41. Fussenegger M, et al. Streptogramin-based gene regulation systems for mammalian cells. Nat Biotechnol 2000;18:1203–8. [PubMed: 11062442] NIH-PA Author Manuscript 42. Gossen M, Bujard H. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc Natl Acad Sci U S A 1992;89:5547–51. [PubMed: 1319065] 43. Weber W, et al. Macrolide-based transgene control in mammalian cells and mice. Nat Biotechnol 2002;20:901–7. [PubMed: 12205509] 44. Neddermann P, et al. A novel, inducible, eukaryotic gene expression system based on the quorum- sensing transcription factor TraR. EMBO Rep 2003;4:159–65. [PubMed: 12612605] 45. Weber W, et al. Streptomyces-derived quorum-sensing systems engineered for adjustable transgene expression in mammalian cells and mice. Nucleic Acids Res 2003;31:e71. [PubMed: 12853648] 46. Malphettes L, et al. A novel mammalian expression system derived from components coordinating nicotine degradation in arthrobacter nicotinovorans pAO1. Nucleic Acids Res 2005;33:e107.
[PubMed: 16002786] 47. Mullick A, et al. The cumate gene-switch: a system for regulated expression in mammalian cells.
BMC Biotechnol 2006;6:43. [PubMed: 17083727] 48. Weber W, Link N, Fussenegger M. A genetic redox sensor for mammalian cells. Metab Eng 2006;8:273–80. [PubMed: 16473537] 49. Weber W, et al. Gas-inducible transgene expression in mammalian cells and mice. Nat Biotechnol 2004;22:1440–4. [PubMed: 15502819] 50. Boorsma M, et al. A temperature-regulated replicon-based DNA expression system. Nat Biotechnol NIH-PA Author Manuscript 2000;18:429–32. [PubMed: 10748525] 51. Weber W, et al. Conditional human VEGF-mediated vascularization in chicken embryos using a novel temperature-inducible gene regulation (TIGR) system. Nucleic Acids Res 2003;31:e69.
[PubMed: 12799458] 52. Weber W, et al. A synthetic mammalian electro-genetic transcription circuit. Nucleic Acids Res 2009;37:e33. [PubMed: 19190091] 53. Eddy SR. Non-coding RNA genes and the modern RNA world. Nat Rev Genet 2001;2:919–29.
[PubMed: 11733745] 54. Doudna JA, Cech TR. The chemical repertoire of natural ribozymes. Nature 2002;418:222–8.
[PubMed: 12110898] 55. Guerrier-Takada C, Gardiner K, Marsh T, Pace N, Altman S. The RNA moiety of ribonuclease P is the catalytic subunit of the enzyme. Cell 1983;35:849–57. [PubMed: 6197186] 56. Kruger K, et al. Self-splicing RNA: autoexcision and autocyclization of the ribosomal RNA intervening sequence of Tetrahymena. Cell 1982;31:147–57. [PubMed: 6297745] Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 57. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993;75:843–54. [PubMed: 8252621] NIH-PA Author Manuscript 58. Stougaard P, Molin S, Nordstrom K. RNAs involved in copy-number control and incompatibility of plasmid R1. Proc Natl Acad Sci U S A 1981;78:6008–12. [PubMed: 6171808] 59. Wagner EG, Simons RW. Antisense RNA control in bacteria, phages, and plasmids. Annu Rev Microbiol 1994;48:713–42. [PubMed: 7826024] 60. Isaacs FJ, Dwyer DJ, Collins JJ. RNA synthetic biology. Nat Biotechnol 2006;24:545–54. [PubMed: 61. Gelfand MS, Mironov AA, Jomantas J, Kozlov YI, Perumov DA. A conserved RNA structure element involved in the regulation of bacterial riboflavin synthesis genes. Trends Genet 1999;15:439–42.
[PubMed: 10529804] 62. Johansson J, et al. An RNA thermosensor controls expression of virulence genes in Listeria monocytogenes. Cell 2002;110:551–61. [PubMed: 12230973] 63. Lease RA, Belfort M. A trans-acting RNA as a control switch in Escherichia coli: DsrA modulates function by forming alternative structures. Proc Natl Acad Sci U S A 2000;97:9919–24. [PubMed:10954740] 64. Majdalani N, Hernandez D, Gottesman S. Regulation and mode of action of the second small RNA activator of RpoS translation, RprA. Mol Microbiol 2002;46:813–26. [PubMed: 12410838] 65. Mandal M, Boese B, Barrick JE, Winkler WC, Breaker RR. Riboswitches control fundamental biochemical pathways in Bacillus subtilis and other bacteria. Cell 2003;113:577–86. [PubMed:12787499] NIH-PA Author Manuscript 66. Mironov AS, et al. Sensing small molecules by nascent RNA: a mechanism to control transcription in bacteria. Cell 2002;111:747–56. [PubMed: 12464185] 67. Morita MT, et al. Translational induction of heat shock transcription factor sigma32: evidence for a built-in RNA thermosensor. Genes Dev 1999;13:655–65. [PubMed: 10090722] 68. Winkler W, Nahvi A, Breaker RR. Thiamine derivatives bind messenger RNAs directly to regulate bacterial gene expression. Nature 2002;419:952–6. [PubMed: 12410317] 69. Winkler WC, Nahvi A, Roth A, Collins JA, Breaker RR. Control of gene expression by a natural metabolite-responsive ribozyme. Nature 2004;428:281–6. [PubMed: 15029187] 70. Winkler WC, Breaker RR. Regulation of bacterial gene expression by riboswitches. Annu Rev Microbiol 2005;59:487–517. [PubMed: 16153177] 71. Good L. Translation repression by antisense sequences. Cell Mol Life Sci 2003;60:854–61. [PubMed: 72. Cox JC, et al. Automated selection of aptamers against protein targets translated in vitro: from gene to aptamer. Nucleic Acids Res 2002;30:e108. [PubMed: 12384610] 73. Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature 1990;346:818–22. [PubMed: 1697402] 74. Hermann T, Patel DJ. Adaptive recognition by nucleic acid aptamers. Science 2000;287:820–5.
NIH-PA Author Manuscript [PubMed: 10657289] 75. Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990;249:505–10. [PubMed: 2200121] 76. Yen L, et al. Exogenous control of mammalian gene expression through modulation of RNA self- cleavage. Nature 2004;431:471–6. [PubMed: 15386015] 77. Looger LL, Dwyer MA, Smith JJ, Hellinga HW. Computational design of receptor and sensor proteins with novel functions. Nature 2003;423:185–90. [PubMed: 12736688] 78. Skerker JM, et al. Rewiring the specificity of two-component signal transduction systems. Cell 2008;133:1043–54. [PubMed: 18555780] 79. Bashor CJ, Helman NC, Yan S, Lim WA. Using engineered scaffold interactions to reshape MAP kinase pathway signaling dynamics. Science 2008;319:1539–43. [PubMed: 18339942] 80. Levskaya A, et al. Synthetic biology: engineering Escherichia coli to see light. Nature 2005;438:441– 2. [PubMed: 16306980] 81. Levskaya A, Weiner OD, Lim WA, Voigt CA. Spatiotemporal control of cell signalling using a light- switchable protein interaction. Nature. 2009 Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 82*. Tabor JJ, et al. A synthetic genetic edge detection program. Cell 2009;137:1272–81. An oustanding example of how smaller circuits can be combined to realize larger genetic programs withsophisticated and predictable behaviour. Here, logic gates and cell-cell communication modules NIH-PA Author Manuscript were coupled to program a population of bacteria to sense and trace the edges of a projected image.
[PubMed: 19563759] 83. Ferrari S, et al. Mutations of the Igbeta gene cause agammaglobulinemia in man. J Exp Med 2007;204:2047–51. [PubMed: 17709424] 84. Becker MM, et al. Synthetic recombinant bat SARS-like coronavirus is infectious in cultured cells and in mice. Proc Natl Acad Sci U S A 2008;105:19944–9. [PubMed: 19036930] 85. Tumpey TM, et al. Characterization of the reconstructed 1918 Spanish influenza pandemic virus.
Science 2005;310:77–80. [PubMed: 16210530] 86. Isaacs FJ, et al. Engineered riboregulators enable post-transcriptional control of gene expression. Nat Biotechnol 2004;22:841–7. [PubMed: 15208640] 87. Dwyer DJ, Kohanski MA, Hayete B, Collins JJ. Gyrase inhibitors induce an oxidative damage cellular death pathway in Escherichia coli. Mol Syst Biol 2007;3:91. [PubMed: 17353933] 88. Kohanski MA, Dwyer DJ, Hayete B, Lawrence CA, Collins JJ. A common mechanism of cellular death induced by bactericidal antibiotics. Cell 2007;130:797–810. [PubMed: 17803904] 89. Kohanski MA, Dwyer DJ, Wierzbowski J, Cottarel G, Collins JJ. Mistranslation of membrane proteins and two-component system activation trigger antibiotic-mediated cell death. Cell 2008;135:679–90.
[PubMed: 19013277] 90. Weber W, et al. A synthetic mammalian gene circuit reveals antituberculosis compounds. Proc Natl NIH-PA Author Manuscript Acad Sci U S A 2008;105:9994–8. [PubMed: 18621677] 91. Lu TK, Collins JJ. Dispersing biofilms with engineered enzymatic bacteriophage. Proc Natl Acad Sci U S A 2007;104:11197–202. [PubMed: 17592147] 92*. Lu TK, Collins JJ. Engineered bacteriophage targeting gene networks as adjuvants for antibiotic therapy. Proc Natl Acad Sci U S A 2009;106:4629–34. A pair of studies in which engineeredbacteriophage were used to deliver synthetic enzymes and perturb gene networks for combattingantibiotic-resistant strains of bacteria. [PubMed: 19255432] 93. Ramachandra M, et al. Re-engineering adenovirus regulatory pathways to enhance oncolytic specificity and efficacy. Nat Biotechnol 2001;19:1035–41. [PubMed: 11689848] 94*. Anderson JC, Clarke EJ, Arkin AP, Voigt CA. Environmentally controlled invasion of cancer cells by engineered bacteria. J Mol Biol 2006;355:619–27. Provides a potential synthetic approach tocancer therapy, whereby bacteria were programmed to sense environmental cues of the tumormicroenvironment and respond to them by invading malignant cells. [PubMed: 16330045] 95. Stavreva DA, et al. Ultradian hormone stimulation induces glucocorticoid receptor-mediated pulses of gene transcription. Nat Cell Biol 2009;11:1093–102. [PubMed: 19684579] 96. Weber W, et al. A synthetic time-delay circuit in mammalian cells and mice. Proc Natl Acad Sci U S A 2007;104:2643–8. [PubMed: 17296937] 97. Dean JT, et al. Resistance to diet-induced obesity in mice with synthetic glyoxylate shunt. Cell Metab NIH-PA Author Manuscript 2009;9:525–36. [PubMed: 19490907] 98. Fortman JL, et al. Biofuel alternatives to ethanol: pumping the microbial well. Trends Biotechnol 2008;26:375–81. [PubMed: 18471913] 99. Alper H, Stephanopoulos G. Engineering for biofuels: exploiting innate microbial capacity or importing biosynthetic potential? Nat Rev Microbiol 2009;7:715–23. [PubMed: 19756010] 100. Jones DT, Woods DR. Acetone-butanol fermentation revisited. Microbiol Rev 1986;50:484–524.
[PubMed: 3540574] 101. Tummala SB, Welker NE, Papoutsakis ET. Design of antisense RNA constructs for downregulation of the acetone formation pathway of Clostridium acetobutylicum. J Bacteriol 2003;185:1923–34.
[PubMed: 12618456] 102. Shao L, et al. Targeted gene disruption by use of a group II intron (targetron) vector in Clostridium acetobutylicum. Cell Res 2007;17:963–5. [PubMed: 17971808] 103. Inui M, et al. Expression of Clostridium acetobutylicum butanol synthetic genes in Escherichia coli.
Appl Microbiol Biotechnol 2008;77:1305–16. [PubMed: 18060402] Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 104. Atsumi S, Hanai T, Liao JC. Non-fermentative pathways for synthesis of branched-chain higher alcohols as biofuels. Nature 2008;451:86–9. [PubMed: 18172501] NIH-PA Author Manuscript 105. Martin VJ, Pitera DJ, Withers ST, Newman JD, Keasling JD. Engineering a mevalonate pathway in Escherichia coli for production of terpenoids. Nat Biotechnol 2003;21:796–802. [PubMed:12778056] 106*. Ro DK, et al. Production of the antimalarial drug precursor artemisinic acid in engineered yeast.
Nature 2006;440:940–3. A paradigm for the application of synthetic biology to the constructionand optimization of biosynthetic pathways for cost-effective and high-yield microbial production.
In these two papers, the authors demonstrate industrial production of the direct precursor to theantimalarial drug artemisinin, in a broader effort to address worldwide shortages of rare drugs.
[PubMed: 16612385] 107. Steen EJ, et al. Microbial production of fatty-acid-derived fuels and chemicals from plant biomass.
Nature 2010;463:559–62. [PubMed: 20111002] 108. Bayer TS, et al. Synthesis of methyl halides from biomass using engineered microbes. J Am Chem Soc 2009;131:6508–15. [PubMed: 19378995] 109. Ma SM, et al. Complete reconstitution of a highly reducing iterative polyketide synthase. Science 2009;326:589–92. [PubMed: 19900898] 110. Kodumal SJ, et al. Total synthesis of long DNA sequences: synthesis of a contiguous 32-kb polyketide synthase gene cluster. Proc Natl Acad Sci U S A 2004;101:15573–8. [PubMed:15496466] 111. Alper H, Fischer C, Nevoigt E, Stephanopoulos G. Tuning genetic control through promoter NIH-PA Author Manuscript engineering. Proc Natl Acad Sci U S A 2005;102:12678–83. [PubMed: 16123130] 112. Ellis T, Wang X, Collins JJ. Diversity-based, model-guided construction of synthetic gene networks with predicted functions. Nat Biotechnol 2009;27:465–71. [PubMed: 19377462] 113. Lu TK, Khalil AS, Collins JJ. Next-generation synthetic gene networks. Nat Biotechnol 2009;27:1139–50. [PubMed: 20010597] 114. Voigt CA. Genetic parts to program bacteria. Curr Opin Biotechnol 2006;17:548–57. [PubMed: 115. Widmaier DM, et al. Engineering the Salmonella type III secretion system to export spider silk monomers. Mol Syst Biol 2009;5:309. [PubMed: 19756048] 116. Mitchell A, et al. Adaptive prediction of environmental changes by microorganisms. Nature 2009;460:220–4. [PubMed: 19536156] 117. Tagkopoulos I, Liu YC, Tavazoie S. Predictive behavior within microbial genetic networks. Science 2008;320:1313–7. [PubMed: 18467556] 118. Fernando CT, et al. Molecular circuits for associative learning in single-celled organisms. J R Soc Interface 2009;6:463–9. [PubMed: 18835803] 119. Fritz G, Buchler NE, Hwa T, Gerland U. Designing sequential transcription logic: a simple genetic circuit for conditional memory. Syst Synth Biol 2007;1:89–98. [PubMed: 19003438] 120. Dunlap JC. Molecular bases for circadian clocks. Cell 1999;96:271–90. [PubMed: 9988221] NIH-PA Author Manuscript 121*. Wang HH, et al. Programming cells by multiplex genome engineering and accelerated evolution.
Nature 2009;460:894–8. Provides a combinatorial/evolutionary approach to optimizingbiosynthetic pathway components through rapid, in vivo genome engineering by cycles of targetedgenome modification and phenotype selection. [PubMed: 19633652] 122. Blake WJ, MKA, Cantor CR, Collins JJ. Noise in eukaryotic gene expression. Nature 2003;422:633– 7. [PubMed: 12687005] 123. Guido NJ, et al. A bottom-up approach to gene regulation. Nature 2006;439:856–60. [PubMed: 124. Salis HM, Mirsky EA, Voigt CA. Automated design of synthetic ribosome binding sites to control protein expression. Nat Biotechnol 2009;27:946–50. [PubMed: 19801975] 125. Dueber JE, et al. Synthetic protein scaffolds provide modular control over metabolic flux. Nat Biotechnol 2009;27:753–9. [PubMed: 19648908] 126. Gibson DG, et al. Complete chemical synthesis, assembly, and cloning of a Mycoplasma genitalium genome. Science 2008;319:1215–20. [PubMed: 18218864] Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins 127. Glass JI, et al. Essential genes of a minimal bacterium. Proc Natl Acad Sci U S A 2006;103:425– 30. [PubMed: 16407165] NIH-PA Author Manuscript 128. Lartigue C, et al. Genome transplantation in bacteria: changing one species to another. Science 2007;317:632–8. [PubMed: 17600181] 129. Lartigue C, et al. Creating bacterial strains from genomes that have been cloned and engineered in yeast. Science. 2009 Early synthetic biology designs, namely the genetic toggle switch and repessilator,demonstrated that regulatory components can be characterized and assembled to bringabout complex, electronics-inspired behaviors in living systems (e.g., memory storageand timekeeping).
Through the characterization and assembly of genetic parts and biological buildingblocks, many more devices have been constructed, including switches, memoryelements, oscillators, pulse generators, digital logic gates, filters, and communicationmodules Advances in the field are now allowing the expansion beyond small gene networks,to the realm of larger biological programs that hold promise for a wide range ofapplications, including biosensing, therapeutics, and the production of biofuels, NIH-PA Author Manuscript pharmaceuticals, and biomaterials.
Synthetic biosensing circuits consist of sensitive elements that bind analytes andtransducer modules that mobilize cellular responses. Balancing these two modulesinvolves engineering modularity and specificity into the various circuits.
Biosensor sensitive elements include environment-responsive promoters(transcriptional), RNA aptamers (translational), and protein receptors (post-translational).
Biosensor transducer modules include engineered gene networks (transcriptional),non-coding regulatory RNAs (translational), and protein signal transduction circuits(post-translational).
Synthetic biology's contributions to therapeutics have included engineered networksand organisms for disease mechanism elucidation, drug target identification, drugdiscovery platforms, therapeutic treatment, therapeutic delivery, and drug productionand access.
In the microbial production of biofuels and pharmaceuticals, synthetic biology has NIH-PA Author Manuscript supplemented traditional genetic and metabolic engineering efforts by aiding in theconstruction of optimized biosynthetic pathways.
Optimizing metabolic flux through biosynthetic pathways is traditionallyaccomplished by driving the expression of pathway enzymes with strong, induciblepromoters. New synthetic approaches include the rapid diversification of the variouspathway components, the rational and model-guided assembly of pathwaycomponents, as well as hybrid solutions.
James J. Collins is an Investigator of the Howard Hughes Medical Institute, and a William F.
Warren Distinguished Profesor, University Professor, Professor of Biomedical Engineering,and Co-Director of the Center for BioDynamics at Boston University. He is also a core foundingfaculty member of the Wyss Institute for Biologically Inspired Engineering at Harvard Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins University. His research group works in synthetic biology and systems biology, with aparticular focus on network biology approaches to antibiotic action and bacterial defense NIH-PA Author Manuscript Ahmad S. Khalil is a Howard Hughes Medical Institute Postdoctoral Fellow training in the labof Jim Collins at Boston University. His research interests are in the advancement of systemsand synthetic biology through novel technologies, and in particular through integratedmicrofluidic devices and automation. He received his doctorate in mechanical engineeringfrom the Massachusetts Institute of Technology, where he worked in the labs of Angela Belcherand Matt Lang on single-molecule studies of biological systems. While at MIT, he was awardeda Charles Stark Draper Laboratory Fellowship in Biomedical Engineering.
NIH-PA Author Manuscript NIH-PA Author Manuscript Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins NIH-PA Author Manuscript FIGURE 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins NIH-PA Author Manuscript NIH-PA Author Manuscript FIGURE 2.
NIH-PA Author Manuscript Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins NIH-PA Author Manuscript FIGURE 3.
NIH-PA Author Manuscript NIH-PA Author Manuscript Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.
Khalil and Collins NIH-PA Author Manuscript FIGURE 4.
NIH-PA Author Manuscript NIH-PA Author Manuscript Nat Rev Genet. Author manuscript; available in PMC 2010 November 1.

Source: http://mse.lab.scu.edu.cn/www/407/discuz/attachments/month_1111/111126234395cc53b4f4ab4d52.attach

Microsoft word - part

Part 1 - General Polices, Procedures and Definitions CIVIL AVIATION RULES AND STANDARDS FEDRAL DEMOCRATIC REPUBLIC OF ETHIOPIA PART 1 — GENERAL POLICIES, PROCEDURES, AND DEFINITIONS General Polices, Procedures and Definitions [THIS PAGE INTENTIONALLY LEFT BLANK] Part 1 - General Polices, Procedures and Definitions Part 1 - General Polices, Procedures and Definitions

Clostridium difficile diagnostik

How to treat multiresistant bacterial infections? Reno Frei, M.D. Klinische Mikrobiologie Universitätsspital Basel 4031 Basel [email protected] www.labormedizin-uhbs.ch Restricted Use in Children -1  Fluoroquinolones (Cipro-, Levo-, Moxifloxacin) • Not approved for children age <18 years