Difference between revisions of "Team:William and Mary/Description"

 
(90 intermediate revisions by 4 users not shown)
Line 4: Line 4:
 
{{Team:William_and_Mary/HEADER2}}
 
{{Team:William_and_Mary/HEADER2}}
  
<!--website by John Mitchell, iGEM 2016
 
Bootstrap template by creative-tim-->
 
  
 
<html lang="en">
 
<html lang="en">
 
<cfheader name="X-XSS-Protection" value="1">
 
<cfheader name="X-XSS-Protection" value="1">
 +
 +
 +
       
 
          
 
          
        <div class="wrapper">
 
 
<div class="section section-numbers-2" id="numbers2" style="padding-top: 0px !important; border-top: 0px !important; margin-top: 0px !important;">
 
  <div class="parallaxWM pattern-image">
 
  <img alt="..." src="https://static.igem.org/mediawiki/2016/c/ce/T--William_and_Mary--rubik-background.jpg"/>
 
  </div>
 
  <div class="container">
 
<div class="col-md-8 col-md-offset-2 text-center">
 
<div class="description">
 
<div class="title">
 
<h2 style='padding-top: 40px; font-family: "Verlag-Book"; font-size: 50px;'>
 
Description
 
</h2>
 
</div>
 
</div>
 
</div>
 
  <div class="clearfix"></div>
 
  </div>
 
</div>
 
 
<p>
 
x
 
</p>
 
  
<div class="section section-we-are-2">
+
<style>
<div class="text-area">
+
      body {
<div class="container">
+
        font-family: 'PT Sans', sans-serif;
<div class="row">
+
        font-size: 13px;
<div class="col-md-4">
+
      }
<div class="title add-animation-stopped">
+
<p class='h2WM' style='padding-left:70px; padding-top: 0px;'>
+
Motivation
+
</p>
+
</div>
+
</div>
+
<div class="col-md-7 col-md-offset-1" style='padding-top: 0px;'>
+
<div class="description add-animation-stopped animation-1">
+
<p class='large' style="color:#A9A9A9;">
+
  Genetic circuits exist in great abundance in nature as complex metabolic pathways which interact in various ways to perform vital cellular processes. Synthetic biologists aim to not only understand naturally occurring circuit networks, but also to modify them or to conceptualize and build entirely new circuits. The inherent versatility of synthetic genetic circuitry has led to a vast array of diverse applications in countless fields. However, the field remains fundamentally limited by the magnitude and specificity of behavioral control over genetic circuits and circuit networks. These limitations can be boiled down to <u>two essential problems</u>:
+
</p>
+
</div>
+
</div>
+
<div class="description">
+
<p class='large'>
+
<b>1.</b> Inherent constraints to behavior based on the nature of a circuit’s constituent genes. </p>
+
<p class='large' style = "padding-left:50px; text-indent:30px;"> The fundamental constraints of integral circuit components limit the ability to design and construct genetic circuits of arbitrary and highly specific behavior. When constructing a circuit with some intended behavior, design is limited by the available input-specific regulators to gene expression and their characteristic regulatory behavior. In order to achieve more precise behavioral control, the ability to tune expression levels of regulatory elements to some desired level is vital. </p>
+
<p class='large' style = "padding-left:50px; text-indent:30px;"> This limitation highlights the need for genetic devices that can modify the behavior of <b>arbitrary genetic circuits</b>; implementing these devices would enable precise behavioral control that is invariant to the constraints of the constituent genes that make up the circuit [1]. </p>
+
</p>
+
<p class='large'>
+
<b>2.</b> The inefficiency of the “design-build-test” cycle which is relied upon for the construction of effective circuit models. </p>
+
<p class='large' style = "padding-left:20px; text-indent:20px;"> The other foundational limitation of genetic circuit construction addresses the inefficiency and unpredictability of the design and construction process itself. The progression from synthesizing parts into a circuit on a plasmid, to transformation and testing in vivo, is a lengthy and expensive process which furthermore is largely variable in terms of actual functionality of the final product [2]. This often leads to a series of trial-and-error testing cycles whose products maintain a persistent level of uncertainty with regard to precise, predictable behavior. Although it is possible to achieve functional genetic circuits in this capacity, greater problems arise regarding the tunability of the product. </p>
+
<p class='large' style = "padding-left:20px; text-indent:20px;"> The success of any genetic circuit relies on the ability to precisely tune a response to a range of input concentrations; it would therefore be desirable to obtain a reliable method for tuning circuit response, ideally <b>without the need to redesign the core of the circuit</b>. Such a method would allow control over output expression to be implemented in a more rapid and predictable manner [3]. </p>
+
</p>
+
</div>
+
<div class="col-md-4">
+
<div class="title add-animation-stopped">
+
<p class='h2WM' style='padding-left:70px;'>
+
The Circuit Control Toolbox
+
</p>
+
</div>
+
</div>
+
<div class="col-md-7 col-md-offset-1" style='padding-top: 0px;'>
+
<div class="description add-animation-stopped animation-1">
+
<p class='large' style="color:#A9A9A9;">
+
Our project aims to provide a modular collection of genetic parts which can specifically and predictably tune the behavior of an arbitrary genetic circuit. This collection, which we have dubbed the “Circuit Control Toolbox,” consists of a suite of parts which can be added to the end of a given genetic circuit; each part provides a specific and independently tunable response which allows direct control over the ultimate output behavior of the circuit.
+
</p>
+
</div>
+
</div>
+
<div class="description">
+
<p class='large' style="padding-left:20px; text-indent:20px;">
+
The overall input/output behavior of any genetic circuit can be represented by a graph known as a transfer function, which relates concentration of input molecule to output protein expression. Likewise, any modifications to the circuit affecting input/output behavior can be visualized by a transformation of the transfer function representing the circuit. The Circuit Control Toolbox consists of three distinct tools which prompt unique behavioral changes to the circuit’s output relative to its input, and therefore generate different transformations of the circuit’s original transfer function.
+
</p>
+
</div>
+
<div class="description">
+
<p class='large' style="padding-left:70px;">
+
The <a href = "https://2016.igem.org/Team:William_and_Mary/RBS"> RBS library </a> provides a collection of ribosome binding sites of varying strength; replacing the RBS within a circuit alters the translational efficiency of the output. This tool effectively allows for scaled changes in the magnitude of a circuit’s output response, thus adjusting the amplitude of the transfer function.
+
</p>
+
<p class='large' style="padding-left:70px;">
+
The <a href = "https://2016.igem.org/Team:William_and_Mary/Binding_Array">  Decoy Binding Array </a> tool implements molecular titration to tune the circuit’s sensitivity to input concentrations. This modification is accompanied by a shift in the threshold of the circuit’s transfer function.
+
</p>
+
  
<p class='large' style="padding-left:70px;">
+
</style>
The <a href = "https://2016.igem.org/Team:William_and_Mary/Synthetic_Enhancer">  Synthetic Enhancer Suite </a> exploits a synthetically modified enhancer/promoter system engineered to allow genetic circuits to generate multi-state responses. In other words, circuits are prompted to produce distinct levels of output based on the concentration of input molecule.This creates a staircase-like curve in the transfer function for the circuit.
+
</p>
+
<p class='large' style="padding-left:70px;">
+
Each of these tools functions orthogonally to the activity of the other tools; furthermore, each tool is independently tunable to a specific degree. By implementing and adjusting multiple tools to the desired degree, a diverse range of circuit output behaviors can be achieved, generating a plethora of unique transfer function responses.
+
</p>
+
<div class="col-md-4">
+
<div class="title add-animation-stopped">
+
<p class='h2WM' style='padding-left:70px;'>
+
Using the Toolbox
+
</p>
+
</div>
+
</div>
+
  
<p class='large' style="padding-left:70px; padding-top: 50px;">
+
</head>
The implementation of this Toolbox relies on its generalizability and consistency over any arbitrary genetic circuit. A circuit’s relative output behavior may be influenced by the coding sequence for the output which it controls. In order to ensure that behavior remains consistent across any range of coding sequences, we offer an additional <a href= "https://2016.igem.org/Team:William_and_Mary/RiboJ">Ribozyme Insulator</a> tool. This ribozyme part, known as RiboJ, insulates a circuit’s promoter activity from the genetic context of the coding sequence, allowing for consistency in the levels of relative expression across multiple coding sequence insertions. The addition of RiboJ as an insulator justifies the application of Toolbox components to the end of any genetic circuit, irrespective of its choice of final output protein [4].</p>
+
 
+
 
 +
<body>
 +
<br></br>
 +
<div style='padding-top: 30px;'></div>
 +
 
 +
<center><img src="https://static.igem.org/mediawiki/2017/3/36/T--William_and_Mary--descript.jpeg" width=380px;/></center>
 +
 
 +
 
 +
<div style='padding-top: 30px;'></div>
 +
 
 +
<div style = 'padding-left: 190px; padding-bottom: 10px;font-size: 25px' ><b>Motivation</b></div>
 +
 
 +
<div style='background: #808080; margin: 0px 190px 20px 190px; height:1px;></div>
 +
 
 +
<div style='padding-top: 0px;'></div>
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >
 +
One of the main goals of synthetic biology is to create a modular genetic basis for the independent control of circuit behavior properties. Much progress has been made in achieving this aim for properties like gene expression strength (where well-characterized ribosome binding sites (RBSs) can be swapped within a genetic part), circuit architecture (where promoters can be swapped out to introduce connections and feedback architectures), and even gene expression noise (through a combination of the above two modulations). However, in order to move into the next phase of synthetic biology, we need to be able to control the dynamical properties of circuits— we want to move beyond circuits that focus on endpoint, steady-state values and explore the rich variety of dynamical systems. Fundamentally, gaining controlling of dynamical systems implies gaining control of temporal dynamics. 
 +
</div>
 +
 
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >
 +
Currently, there is no good way to control the temporal dynamics of gene expression. Current control strategies require either a rewiring of the circuit architecture to achieve different time-dependent dynamics [1, 2] or a complete circumvention of transcriptional circuitry altogether, relying on post-translational dynamics like phosphorylation [3] or protein-protein interactions [4] to transmit information through a circuit. These approaches are often inaccessible to iGEM teams because they require too drastic an overhaul of existing circuit implementations. To alleviate this issue, and to enable future iGEM teams to create robust dynamical circuits, we created a protein degradation based ‘plug-and-play’ style system that allows modular and predictable control of the gene expression speed of a given circuit without requiring a fundamental redesign of existing circuit architecture.
 +
</div>
 +
 
 +
 
 +
<div style='padding-top: 60px;'></div>
 +
 
 +
 
 +
<div style = 'padding-left: 190px; padding-bottom: 10px;font-size: 25px' ><b>Degradation Based Control of Gene Expression Speed</b></div>
 +
 
 +
 
 +
<div style='background: #808080; margin: 0px 190px 20px 190px; height:1px;></div>
 +
 
 +
<div style='padding-top: 0px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >The relationship between the degradation rate of a gene's protein product and the response speed of its expression is a well-established concept in the theoretical biology community. Consider the following simple mathematical model presented by Uri Alon [5]. X is an inducible gene which, once activated, produces its protein product x at rate α. If x is degraded at rate γ, then we can use the following differential equation to track the concentration of protein x over time:</div>
 +
 
 +
<figure style='padding-left: 200px;'>
 +
<img src='https://static.igem.org/mediawiki/2017/0/02/T--William_and_Mary--truDescriptionMath1.png' height = "70%" width = "70%"/>
 +
<figcaption><div style='padding-left: 5px;padding-top: 15px; color: #808080; font-size: 14px;'>Figure 1: A simple kinetic model tracking the changing concentration of protein x over time. </div><div style='padding-left: 5px;color: #808080; font-size: 14px;'>Gene circuit component images adapted from Newcastle iGEM 2010; protein images adapted from Cameron and Collins 2014 [7]. </div></figcaption>
 +
</figure>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; line-height: 25px;' >We solve the above differential equation to obtain the following function, which gives the protein concentration at a given time t after activation of X: </div>
 +
 
 +
<figure style='padding-left: 200px;'>
 +
<img src='https://static.igem.org/mediawiki/2017/7/71/T--William_and_Mary--truDescriptionMath2.png' height = "30%" width = "30%"/>
 +
<figcaption><div style='padding-left: 5px;padding-top: 15px; color: #808080; font-size: 14px;'>Figure 2: Concentration of protein x as a function of time; the solution to the differential equation in Figure 1. </div></figcaption>
 +
</figure>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; line-height: 25px;' >We are able to graph this function and determine that the steady state concentration for x is equal to production rate over degradation rate. Finally, defining т<sub>1/2</sub> as the time it takes for x to reach half its steady-state concentration, we find the following: </div>
 +
 
 +
<figure style='padding-left: 200px;'>
 +
<img src='https://static.igem.org/mediawiki/2017/6/65/T--William_and_Mary--DescriptionMath3.png' height = "70%" width = "70%"/>
 +
<figcaption><div style='padding-left: 5px;padding-top: 15px; color: #808080; font-size: 14px;'>Figure 3: We find that т<sub>1/2</sub> is a function of γ, the degradation rate for protein x. </div></figcaption>
 +
</figure>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; line-height: 25px;' >The above expression reveals that in this model, it is not only sufficient but also necessary to tune the degradation rate γ in order to control the gene expression speed (represented here as т<sub>1/2</sub>). This property remains true for more complex circuit architectures like feedforward loops, provided that the steady-state concentration is held constant [5]. </div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >Thus in order to create a plug-and-play style system to control gene expression speed, we developed and characterized a suite of BioBrick parts which allow for simple, modular and predictable changes to the gene expression speed of arbitrary proteins via protein degradation.</div>
 +
 
 +
 
 +
<div style='padding-top: 60px;'></div>
 +
 
 +
 
 +
<div style = 'padding-left: 190px; padding-bottom: 10px;font-size: 25px' ><b>Orthogonal Degradation Tags</b></div>
 +
 
 +
 
 +
<div style='background: #808080; margin: 0px 190px 20px 190px; height:1px;></div>
 +
 
 +
<div style='padding-top: 0px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >When we created our gene expression control system, we wanted to make sure that it was both usable across a variety of biological systems and circuits, and easily accessible to other iGEM teams. To this end, we chose to use the <i>Mesoplasma florum</i> Lon (mf-Lon) protease system which was characterized by Gur and Sauer in 2008 [6] and developed into a modular suite of genetic parts by Cameron and Collins in 2014 [7]. This system consists of of a AAA+ protease and its associated protein degradation tags (pdt), which operate in a mechanistically similar manner to the E. Coli endogenous protease ClpXP and its associated ssrA tags. However, unlike ClpXP and ssrA tags, mf-Lon and pdts are completely orthogonal to the endogenous protein degradation systems in E. coli. Using this orthogonal degradation system helps eliminate cross-talk between our system and endogenous E. coli proteins. Further, since there are pdts with a wide range of different affinities, we are able to tune degradation rate, and thus gene expression speed to a wide variety of values. This represents not only a practical advancement in the tuning of circuits, but also an important contribution in our theoretical understanding of biological processes, as to our knowledge there has so far been no direct experimental measurement in the literature of the predicted 1/γ scaling of a gene's speed with its degradation rate. See our <a href='https://2017.igem.org/Team:William_and_Mary/Results' style='text-decoration: underline;'>results</a> page for more details.</div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style='padding-top: 60px;'></div>
 +
 
 +
 
 +
<div style = 'padding-left: 190px; padding-bottom: 10px;font-size: 25px' ><b>Modeling</b></div>
 +
 
 +
 
 +
<div style='background: #808080; margin: 0px 190px 20px 190px; height:1px;></div>
 +
 
 +
<div style='padding-top: 0px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 50px;line-height: 25px;' >Finally, by constructing a mechanistic model of our degradation system, we were able to rigorously analyze our timeseries datasets using Bayesian Parameter Inference and obtain parameter distributions (Figure 4). We then used this analysis to feed back into our predictive saturation model, which we used to develop the beginnings of a theoretical framework for understanding the relationship between protease-driven speed control and saturation effects on the protease. By combining our characterization measurements with our conceptual insights, future iGEM teams are now enabled to use the pdt system to predictably control the temporal dynamics of their genetic circuits. See our <a href='https://2017.igem.org/Team:William_and_Mary/Model' style='text-decoration: underline;'>modeling</a> page for details
 +
</div>
 +
 
 +
<center>
 +
<figure style='padding-left: px;'>
 +
<img src='https://static.igem.org/mediawiki/2017/5/5a/T--William_and_Mary--MCMC-F2.png' width = "45%"/>
 +
<figcaption><div style='padding-left: 20%;padding-right:20%; padding-top: 15px; color: #808080; font-size: 14px;'>Figure 4: MCMC parameter estimation for a simple protein production model correctly estimates the simulated values for both beta and gamma (15 and .03 respectively). In addition, the MCMC identifies a strong positive correlation between beta and gamma. This makes sense as steady-state levels of protein are set by the ratio Beta/Gamma. Therefore as beta goes up we should see a corresponding increase in gamma to best fit the model.
 +
</div></figcaption>
 +
</figure>
 +
</center>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style='padding-top: 60px;'></div>
 +
 
 +
<!---References----->
 +
 
 +
 
 +
 
 +
<div style = 'padding-left: 190px; padding-bottom: 10px;font-size: 25px' ><b>References</b></div>
 +
 
 +
<div style='background: #808080; margin: 0px 190px 20px 190px; height:1px;></div>
 +
 
 +
<div style='padding-top: 0px;'></div>
 +
 
 +
<div style='padding-top: 30px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >[1]  Nitzan Rosenfeld, Michael B Elowitz, and Uri Alon. Negative autoregulation speeds the response times of transcription networks. Journal of molecular biology, 323(5):785–793, 2002.
 +
</div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
 
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >[2] Shmoolik Mangan and Uri Alon. Structure and function of the feed-forward loop network motif. Proceedings of the National Academy of Sciences, 100(21):11980–11985, 2003.
 +
</div>
 +
<div style='padding-top: 15px;'></div>
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >
 +
[3] Russell M Gordley, Reid E Williams, Caleb J Bashor, Jared E Toettcher, Shude Yan, and Wendell A Lim. Engineering dynamical control of cell fate switching using synthetic phospho- regulons. Proceedings of the National Academy of Sciences, 113(47):13528–13533, 2016.</div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >
 +
[4] Anselm Levskaya, Orion D Weiner, Wendell A Lim, and Christopher A Voigt. Spatiotemporal control of cell signalling using a light-switchable protein interaction. Nature, 461(7266):997– 1001, 2009.
 +
</div>
 +
<div style='padding-top: 15px;'></div>
 +
 
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >
 +
[5]  Uri Alon. An introduction to systems biology: design principles of biological circuits. CRC press, 2006
 +
</div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >
 +
[6] Eyal Gur and Robert T Sauer. Evolution of the ssra degradation tag in mycoplasma: specificity switch to a different protease. Proceedings of the National Academy of Sciences, 105(42):16113– 16118, 2008.
 +
</div>
 +
 
 +
<div style='padding-top: 15px;'></div>
 +
<div style = 'padding-right: 190px; padding-left: 190px; text-indent: 0px;line-height: 25px;' >
 +
[7] D Ewen Cameron and James J Collins. Tunable protein degradation in bacteria. Nature biotechnology, 32(12):1276–1281, 20
 +
</div>
 +
 
 +
 
 +
 
 +
 
 +
 
 +
</body>
  
  
<p class='large' style="padding-left:70px; padding-bottom:30px; padding-top:50px;">
 
<b>Our Circuit Control Toolbox can easily be implemented in any project concerned with the behavior of genetic circuitry by working through the following sequence of events</b>:
 
</p>
 
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>1.</b> Visualize the original behavior of the circuit in question by constructing a characteristic transfer function.
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>2.</b> Determine the appropriate Toolbox parts-to-use using our mathematical model. This model has been parameterized such that the model parameters correspond to actual physical variables (e.g. number of tetO arrays, plasmid backbone).
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>3.</b> Swap out the final protein coding sequence in the original circuit with a Ribo-J insulated repressor sequence that is compatible with the Toolbox.
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 50px;">
 
<b>4.</b> Apply the appropriate Toolbox parts to the end of your circuit, and express your original final output protein at the end of the series of Toolbox components.
 
</p>
 
 
<p class='large' style="padding-left:70px; padding-bottom: 100px;">
 
In this manner, future iGEM teams and synthetic biologists will be able to easily obtain higher levels of precision and control over the behavior of their genetic networks.
 
</p>
 
</div>
 
<div align="center">
 
<h3 style='padding-top: 50px; padding-bottom: 50px;'>References</h3>
 
</div>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>1.</b> Nielsen, A. A., Segall-Shapiro, T. H., & Voigt, C. A. (2013). Advances in genetic circuit design:
 
Novel biochemistries, deep part mining, and precision gene expression. Current Opinion in Chemical Biology,
 
17(6), 878-892. doi:<a>http://dx.doi.org/10.1016/j.cbpa.2013.10.003</a>
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>2.</b> Sun, Z. Z., Yeung, E., Hayes, C. A., Noireaux, V., & Murray, R. M.Linear DNA for rapid prototyping of
 
synthetic biological circuits in an escherichia coli based TX-TL cell-free system. ACS Synthetic Biology, (6), 387.
 
doi:10.1021/sb400131a
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>3.</b> Lucks, J. B., Qi, L., Whitaker, W. R., & Arkin, A. P. (2008). Toward scalable parts families for predictable
 
design of biological circuits. Current Opinion in Microbiology, 11(6), 567-573. doi:<a>http://dx.doi.org/10.1016/j.mib.2008.10.002</a>
 
</p>
 
<p class='large' style="padding-left:70px; padding-bottom: 30px;">
 
<b>4.</b> Lou, C., Stanton, B., Chen, Y., Munsky, B., & Voigt, C. A. (2012). Ribozyme-based insulator parts buffer
 
synthetic circuits from genetic context. Nature Biotechnology, (30), 1137-1142.
 
</p>
 
</div>
 
</div>
 
</div>
 
<!--
 
<div id="carousel">
 
<div id="section-we-are-1" class="carousel slide add-animation-stopped-1" data-interval="20000">
 
<div class="carousel-inner pattern-image animate">
 
<div class="item  active">
 
<img alt="..." class="full-image" src="assets/img/builder/projects/project.jpg">
 
</div>
 
<div class="item">
 
<img alt="..." class="full-image" src="assets/img/builder/projects/project.jpg">
 
</div>
 
</div>
 
<a class="left carousel-control" href="#section-we-are-1" data-slide="prev"><span class="fa fa-angle-left hidden-md hidden-lg"></span></a>     
 
<a class="right carousel-control" href="#section-we-are-1" data-slide="next"><span class="fa fa-angle-right hidden-ms hidden-lg"></span></a>     
 
</div>
 
</div>
 
-->
 
</div>
 
 
</html>
 
</html>
 +
 +
 +
  
 
{{Team:William_and_Mary/JS_SOURCE2}}
 
{{Team:William_and_Mary/JS_SOURCE2}}
 
{{Team:William_and_Mary/FOOTER_SMALL}}
 
{{Team:William_and_Mary/FOOTER_SMALL}}

Latest revision as of 03:07, 2 November 2017




Motivation
One of the main goals of synthetic biology is to create a modular genetic basis for the independent control of circuit behavior properties. Much progress has been made in achieving this aim for properties like gene expression strength (where well-characterized ribosome binding sites (RBSs) can be swapped within a genetic part), circuit architecture (where promoters can be swapped out to introduce connections and feedback architectures), and even gene expression noise (through a combination of the above two modulations). However, in order to move into the next phase of synthetic biology, we need to be able to control the dynamical properties of circuits— we want to move beyond circuits that focus on endpoint, steady-state values and explore the rich variety of dynamical systems. Fundamentally, gaining controlling of dynamical systems implies gaining control of temporal dynamics.
Currently, there is no good way to control the temporal dynamics of gene expression. Current control strategies require either a rewiring of the circuit architecture to achieve different time-dependent dynamics [1, 2] or a complete circumvention of transcriptional circuitry altogether, relying on post-translational dynamics like phosphorylation [3] or protein-protein interactions [4] to transmit information through a circuit. These approaches are often inaccessible to iGEM teams because they require too drastic an overhaul of existing circuit implementations. To alleviate this issue, and to enable future iGEM teams to create robust dynamical circuits, we created a protein degradation based ‘plug-and-play’ style system that allows modular and predictable control of the gene expression speed of a given circuit without requiring a fundamental redesign of existing circuit architecture.
Degradation Based Control of Gene Expression Speed
The relationship between the degradation rate of a gene's protein product and the response speed of its expression is a well-established concept in the theoretical biology community. Consider the following simple mathematical model presented by Uri Alon [5]. X is an inducible gene which, once activated, produces its protein product x at rate α. If x is degraded at rate γ, then we can use the following differential equation to track the concentration of protein x over time:
Figure 1: A simple kinetic model tracking the changing concentration of protein x over time.
Gene circuit component images adapted from Newcastle iGEM 2010; protein images adapted from Cameron and Collins 2014 [7].
We solve the above differential equation to obtain the following function, which gives the protein concentration at a given time t after activation of X:
Figure 2: Concentration of protein x as a function of time; the solution to the differential equation in Figure 1.
We are able to graph this function and determine that the steady state concentration for x is equal to production rate over degradation rate. Finally, defining т1/2 as the time it takes for x to reach half its steady-state concentration, we find the following:
Figure 3: We find that т1/2 is a function of γ, the degradation rate for protein x.
The above expression reveals that in this model, it is not only sufficient but also necessary to tune the degradation rate γ in order to control the gene expression speed (represented here as т1/2). This property remains true for more complex circuit architectures like feedforward loops, provided that the steady-state concentration is held constant [5].
Thus in order to create a plug-and-play style system to control gene expression speed, we developed and characterized a suite of BioBrick parts which allow for simple, modular and predictable changes to the gene expression speed of arbitrary proteins via protein degradation.
Orthogonal Degradation Tags
When we created our gene expression control system, we wanted to make sure that it was both usable across a variety of biological systems and circuits, and easily accessible to other iGEM teams. To this end, we chose to use the Mesoplasma florum Lon (mf-Lon) protease system which was characterized by Gur and Sauer in 2008 [6] and developed into a modular suite of genetic parts by Cameron and Collins in 2014 [7]. This system consists of of a AAA+ protease and its associated protein degradation tags (pdt), which operate in a mechanistically similar manner to the E. Coli endogenous protease ClpXP and its associated ssrA tags. However, unlike ClpXP and ssrA tags, mf-Lon and pdts are completely orthogonal to the endogenous protein degradation systems in E. coli. Using this orthogonal degradation system helps eliminate cross-talk between our system and endogenous E. coli proteins. Further, since there are pdts with a wide range of different affinities, we are able to tune degradation rate, and thus gene expression speed to a wide variety of values. This represents not only a practical advancement in the tuning of circuits, but also an important contribution in our theoretical understanding of biological processes, as to our knowledge there has so far been no direct experimental measurement in the literature of the predicted 1/γ scaling of a gene's speed with its degradation rate. See our results page for more details.
Modeling
Finally, by constructing a mechanistic model of our degradation system, we were able to rigorously analyze our timeseries datasets using Bayesian Parameter Inference and obtain parameter distributions (Figure 4). We then used this analysis to feed back into our predictive saturation model, which we used to develop the beginnings of a theoretical framework for understanding the relationship between protease-driven speed control and saturation effects on the protease. By combining our characterization measurements with our conceptual insights, future iGEM teams are now enabled to use the pdt system to predictably control the temporal dynamics of their genetic circuits. See our modeling page for details
Figure 4: MCMC parameter estimation for a simple protein production model correctly estimates the simulated values for both beta and gamma (15 and .03 respectively). In addition, the MCMC identifies a strong positive correlation between beta and gamma. This makes sense as steady-state levels of protein are set by the ratio Beta/Gamma. Therefore as beta goes up we should see a corresponding increase in gamma to best fit the model.
References
[1]  Nitzan Rosenfeld, Michael B Elowitz, and Uri Alon. Negative autoregulation speeds the response times of transcription networks. Journal of molecular biology, 323(5):785–793, 2002.
[2] Shmoolik Mangan and Uri Alon. Structure and function of the feed-forward loop network motif. Proceedings of the National Academy of Sciences, 100(21):11980–11985, 2003.
[3] Russell M Gordley, Reid E Williams, Caleb J Bashor, Jared E Toettcher, Shude Yan, and Wendell A Lim. Engineering dynamical control of cell fate switching using synthetic phospho- regulons. Proceedings of the National Academy of Sciences, 113(47):13528–13533, 2016.
[4] Anselm Levskaya, Orion D Weiner, Wendell A Lim, and Christopher A Voigt. Spatiotemporal control of cell signalling using a light-switchable protein interaction. Nature, 461(7266):997– 1001, 2009.
[5]  Uri Alon. An introduction to systems biology: design principles of biological circuits. CRC press, 2006
[6] Eyal Gur and Robert T Sauer. Evolution of the ssra degradation tag in mycoplasma: specificity switch to a different protease. Proceedings of the National Academy of Sciences, 105(42):16113– 16118, 2008.
[7] D Ewen Cameron and James J Collins. Tunable protein degradation in bacteria. Nature biotechnology, 32(12):1276–1281, 20