25 research outputs found

    Trade-offs between retroactivity and noise in connected transcriptional components

    Get PDF
    At the interconnection of two gene transcriptional components in a biomolecular network, the noise in the downstream component can be reduced by increasing its gene copy number. However, this method of reducing noise increases the load applied to the upstream system, called retroactivity, thereby causing a perturbation in the upstream system. In this work, we quantify the error in the system trajectories caused by perturbations due to retroactivity and noise, and analyze the trade-off between these two perturbations. We model the system as a set of nonlinear chemical Langevin equations and quantify the trade-off by employing contraction theory for stochastic systems.National Science Foundation (U.S.). Division of Computing and Communication Foundations (Award 1058127)United States. Air Force Office of Scientific Research (Award FA9550-12-1-0129

    Modular Composition of Gene Transcription Networks

    Get PDF
    Predicting the dynamic behavior of a large network from that of the composing modules is a central problem in systems and synthetic biology. Yet, this predictive ability is still largely missing because modules display context-dependent behavior. One cause of context-dependence is retroactivity, a phenomenon similar to loading that influences in non-trivial ways the dynamic performance of a module upon connection to other modules. Here, we establish an analysis framework for gene transcription networks that explicitly accounts for retroactivity. Specifically, a module's key properties are encoded by three retroactivity matrices: internal, scaling, and mixing retroactivity. All of them have a physical interpretation and can be computed from macroscopic parameters (dissociation constants and promoter concentrations) and from the modules' topology. The internal retroactivity quantifies the effect of intramodular connections on an isolated module's dynamics. The scaling and mixing retroactivity establish how intermodular connections change the dynamics of connected modules. Based on these matrices and on the dynamics of modules in isolation, we can accurately predict how loading will affect the behavior of an arbitrary interconnection of modules. We illustrate implications of internal, scaling, and mixing retroactivity on the performance of recurrent network motifs, including negative autoregulation, combinatorial regulation, two-gene clocks, the toggle switch, and the single-input motif. We further provide a quantitative metric that determines how robust the dynamic behavior of a module is to interconnection with other modules. This metric can be employed both to evaluate the extent of modularity of natural networks and to establish concrete design guidelines to minimize retroactivity between modules in synthetic systems.United States. Air Force Office of Scientific Research (FA9550-12-1-0129

    Principles of genetic circuit design

    Get PDF
    Cells navigate environments, communicate and build complex patterns by initiating gene expression in response to specific signals. Engineers seek to harness this capability to program cells to perform tasks or create chemicals and materials that match the complexity seen in nature. This Review describes new tools that aid the construction of genetic circuits. Circuit dynamics can be influenced by the choice of regulators and changed with expression 'tuning knobs'. We collate the failure modes encountered when assembling circuits, quantify their impact on performance and review mitigation efforts. Finally, we discuss the constraints that arise from circuits having to operate within a living cell. Collectively, better tools, well-characterized parts and a comprehensive understanding of how to compose circuits are leading to a breakthrough in the ability to program living cells for advanced applications, from living therapeutics to the atomic manufacturing of functional materials.National Institute of General Medical Sciences (U.S.) (Grant P50 GM098792)National Institute of General Medical Sciences (U.S.) (Grant R01 GM095765)National Science Foundation (U.S.). Synthetic Biology Engineering Research Center (EEC0540879)Life Technologies, Inc. (A114510)National Science Foundation (U.S.). Graduate Research FellowshipUnited States. Office of Naval Research. Multidisciplinary University Research Initiative (Grant 4500000552

    Beyond the two-state model of switching in biology and computation

    Get PDF
    The thesis presents various perspectives on physical and biological computation. Our fundamental object of study in both these contexts is the notion of switching/erasing a bit. In a physical context, a bit is represented by a particle in a double well, whose dynamics is governed by the Langevin equation. We define the notions of reliability and erasing time-scales in addition to the work required to erase a bit for a given family of control protocols. We call bits “optimal” if they meet the required reliability and erasing time requirements with minimal work cost. We find that optimal bits always saturate the erasing time requirement, but may not saturate the reliability time requirement. This allows us to eliminate several regions of parameter space as sub-optimal. In a biological context, our bits are represented by substrates that are acted upon by catalytic enzymes. We define retroactivity as the back-signal propagated by the downstream system when connected to the upstream system. We analyse certain upstream systems that can help mitigate retroactivity. However, these systems require a substantial pool of resources and are therefore not optimal. As a consequence, we turn our attention to insulating networks called push-pull motifs. We find that high rates of energy consumption are not essential to alleviate retroactivity in push-pull motifs; all we need is to couple weakly to the upstream system. However, this approach is not resilient to cross-talk caused by leak reactions in the circuit. Next, we consider a single enzyme-substrate reaction and analyse its mechanism. Our system has two intermediate states (enzyme-substrate complexes). Our main question is “How should we choose binding energies of the intermediates to minimize sequestra- tion of substrates (retroactivity), whilst maintaining a minimum flux at steady-state?”. Choosing very low binding energies increases retroactivity since the system spends a considerable proportion of time in the intermediate states. Choosing binding energies that are very high reduces retroactivity, but hinders the progress of the reaction. As a result, we find that the the optimal binding energies are both moderate, and indeed tuned with each other. In particular, their difference is related to the free energy difference between the products and reactants.Open Acces

    From Microbial Communities to Distributed Computing Systems

    Get PDF
    A distributed biological system can be defined as a system whose components are located in different subpopulations, which communicate and coordinate their actions through interpopulation messages and interactions. We see that distributed systems are pervasive in nature, performing computation across all scales, from microbial communities to a flock of birds. We often observe that information processing within communities exhibits a complexity far greater than any single organism. Synthetic biology is an area of research which aims to design and build synthetic biological machines from biological parts to perform a defined function, in a manner similar to the engineering disciplines. However, the field has reached a bottleneck in the complexity of the genetic networks that we can implement using monocultures, facing constraints from metabolic burden and genetic interference. This makes building distributed biological systems an attractive prospect for synthetic biology that would alleviate these constraints and allow us to expand the applications of our systems into areas including complex biosensing and diagnostic tools, bioprocess control and the monitoring of industrial processes. In this review we will discuss the fundamental limitations we face when engineering functionality with a monoculture, and the key areas where distributed systems can provide an advantage. We cite evidence from natural systems that support arguments in favor of distributed systems to overcome the limitations of monocultures. Following this we conduct a comprehensive overview of the synthetic communities that have been built to date, and the components that have been used. The potential computational capabilities of communities are discussed, along with some of the applications that these will be useful for. We discuss some of the challenges with building co-cultures, including the problem of competitive exclusion and maintenance of desired community composition. Finally, we assess computational frameworks currently available to aide in the design of microbial communities and identify areas where we lack the necessary tool

    Control Theory for Synthetic Biology: Recent Advances in System Characterization, Control Design, and Controller Implementation for Synthetic Biology

    Get PDF
    Living organisms are differentiated by their genetic material-millions to billions of DNA bases encoding thousands of genes. These genes are translated into a vast array of proteins, many of which have functions that are still unknown. Previously, it was believed that simply knowing the genetic sequence of an organism would be the key to unlocking all understanding. However, as DNA sequencing technology has become affordable, it has become clear that living cells are governed by complex, multilayered networks of gene regulation that cannot be deduced from sequence alone. Synthetic biology as a field might best be characterized as a learn-by-building approach, in which scientists attempt to engineer molecular pathways that do not exist in nature. In doing so, they test the limits of both natural and engineered organisms

    Physics of epigenetic landscapes and statistical inference by cells

    Full text link
    Biology is currently in the midst of a revolution. Great technological advances have led to unprecedented quantitative data at the whole genome level. However, new techniques are needed to deal with this deluge of high-dimensional data. Therefore, statistical physics has the potential to help develop systems biology level models that can incorporate complex data. Additionally, physicists have made great strides in understanding non-equilibrium thermodynamics. However, the consequences of these advances have yet to be fully incorporated into biology. There are three specific problems that I address in my dissertation. First, a common metaphor for describing development is a rugged "epigenetic landscape" where cell fates are represented as attracting valleys resulting from a complex regulatory network. I introduce a framework for explicitly constructing epigenetic landscapes that combines genomic data with techniques from spin-glass physics. The model reproduces known reprogramming protocols and identifies candidate transcription factors for reprogramming to novel cell fates, suggesting epigenetic landscapes are a powerful paradigm for understanding cellular identity. Second, I examine the dynamics of cellular reprogramming. By reanalyzing all available time-series data, I show that gene expression dynamics during reprogramming follow a simple one-dimensional reaction coordinate that is independent of both the time and details of experimental protocol used. I show that such a reaction coordinate emerges naturally from epigenetic landscape models of cell identity where cellular reprogramming is viewed as a "barrier-crossing" between the starting and ending cell fates. Overall, the analysis and model suggest that gene expression dynamics during reprogramming follow a canonical trajectory consistent with the idea of an "optimal path"' in gene expression space for reprogramming. Third, an important task of cells is to perform complex computations in response to external signals. Intricate networks are required to sense and process signals, and since cells are inherently non-equilibrium systems, these networks naturally consume energy. Since there is a deep connection between thermodynamics, computation, and information, a natural question is what constraints does thermodynamics place on statistical estimation and learning. I modeled a single chemical receptor and established the first fundamental relationship between the energy consumption and statistical accuracy of a receptor in a cell

    Advances and Computational Tools towards Predictable Design in Biological Engineering

    Get PDF
    The design process of complex systems in all the fields of engineering requires a set of quantitatively characterized components and a method to predict the output of systems composed by such elements. This strategy relies on the modularity of the used components or the prediction of their context-dependent behaviour, when parts functioning depends on the specific context. Mathematical models usually support the whole process by guiding the selection of parts and by predicting the output of interconnected systems. Such bottom-up design process cannot be trivially adopted for biological systems engineering, since parts function is hard to predict when components are reused in different contexts. This issue and the intrinsic complexity of living systems limit the capability of synthetic biologists to predict the quantitative behaviour of biological systems. The high potential of synthetic biology strongly depends on the capability of mastering this issue. This review discusses the predictability issues of basic biological parts (promoters, ribosome binding sites, coding sequences, transcriptional terminators, and plasmids) when used to engineer simple and complex gene expression systems in Escherichia coli. A comparison between bottom-up and trial-and-error approaches is performed for all the discussed elements and mathematical models supporting the prediction of parts behaviour are illustrated

    Control Theory for Synthetic Biology: Recent Advances in System Characterization, Control Design, and Controller Implementation for Synthetic Biology

    Get PDF
    Living organisms are differentiated by their genetic material-millions to billions of DNA bases encoding thousands of genes. These genes are translated into a vast array of proteins, many of which have functions that are still unknown. Previously, it was believed that simply knowing the genetic sequence of an organism would be the key to unlocking all understanding. However, as DNA sequencing technology has become affordable, it has become clear that living cells are governed by complex, multilayered networks of gene regulation that cannot be deduced from sequence alone. Synthetic biology as a field might best be characterized as a learn-by-building approach, in which scientists attempt to engineer molecular pathways that do not exist in nature. In doing so, they test the limits of both natural and engineered organisms

    Cell-Free Synthetic Biology Platform for Engineering Synthetic Biological Circuits and Systems

    Get PDF
    Synthetic biology brings engineering disciplines to create novel biological systems for biomedical and technological applications. The substantial growth of the synthetic biology field in the past decade is poised to transform biotechnology and medicine. To streamline design processes and facilitate debugging of complex synthetic circuits, cell-free synthetic biology approaches has reached broad research communities both in academia and industry. By recapitulating gene expression systems in vitro, cell-free expression systems offer flexibility to explore beyond the confines of living cells and allow networking of synthetic and natural systems. Here, we review the capabilities of the current cell-free platforms, focusing on nucleic acid-based molecular programs and circuit construction. We survey the recent developments including cell-free transcription– translation platforms, DNA nanostructures and circuits, and novel classes of riboregulators. The links to mathematical models and the prospects of cell-free synthetic biology platforms will also be discussed.11Yscopu
    corecore