SynCell 2023

International Conference on Engineering Synthetic Cells and Organelles


May 22-24 2023, Minneapolis, MN


Thank you everyone for a fantastic meeting. SynCell2023 was a great success, thanks to our speakers, chairs, presenters and participants.

We’re looking forward to seeing everyone next year in Toulouse at SynCell2024.

 
 
 
 
 

Aims of the conference

SynCell2023 is an international conference that aims to bring together researchers worldwide to present and discuss the latest results in in bottom-up synthetic biology research. In addition, SynCell2023 also provides room to discuss future scientific and technological perspectives of research on synthetic cells and organelles, and the philosophical and ethical considerations that come with them.

The program will feature plenary as well as parallel sessions with presentations and discussions, and poster sessions. Invited top scientists will give tutorials for young researchers, and technical sessions and talks for all attendees.

A day after SynCell2023 there will be Build-a-Cell Workshop 10, buildacell.org/workshop10

Please considering staying in Minneapolis the one extra day and participating in the workshop.

 
 

Registration

To register for SynCell2023 on site participation:

  1. Fill out this google form forms.gle/EoBoHhC2mKyMHUGJ6

  2. Pay $300 registration fee using link you will receive after filling out the registration form.

Please note that using the above linked form you are registering for SynCell 2023 meeting. If you would like to also attend Build-a-Cell Workshop 10, a day after SynCell 2023, you have to register for the workshop separately. See info on the workshop website.

Deadlines:

To apply for travel funding: register by the end of February 2023.

To propose a talk: register by the end of March 2023.

To show a poster: register by the end of April 2023.

The last day to register for the conference as a participant will be May 18, 2023.

Remote participation

The talks will not be recorded. 

It is not possible to give a talk remotely. The goal of this meeting is to facilitate interactions between all speakers and to allow junior trainees to interact with senior speakers, so everyone giving talks need to be present on site in person. 

By popular demand, we did add remote viewing option for registered participants. This is to allow people who couldn't travel a chance to see the talks and ask questions during discussion.

The remote participation will only be available to registered participants. 

To register, for free, for remote participation, please fill out this form: https://forms.gle/AUwYcaM3Ew8cA8tQA

There is no registration fee for remote participation. 

Only people who register in advance will be able to join the password-protected remote session. 

 
 

Travel funding

We will have travel funding available.

Priority will be given to trainees, but independent researches are welcome to apply as well.

To apply for travel funding, please fill out the registration form for the meeting, forms.gle/EoBoHhC2mKyMHUGJ6.

 
 

Program

The meeting will be divided into scientific sessions, covering most important aspects of synthetic cell engineering.

The sessions will be:

The detailed program is below.

It will be possible to view the talks remotely. To register, for free, for remote participation, please fill out this form: https://forms.gle/AUwYcaM3Ew8cA8tQA

The working groups
During the meeting, we will form ad-hoc working groups to develop short perspective papers on few key issues, roadblocks and opportunities in synthetic cell engineering.
The working group leaders will pitch their working group topics on Monday, at 11:15 - 12:00AM during the Working groups intro plenary session.
We invite you to join one of those working groups, to influence the perspectives paper on the topic that is closest related to your interests.
We hope that a collection of short, impactful papers coming out of this meeting will help to shape the directions of our field. 

 

Detailed program

Monday, May 22

8:00AM - 9:00AM, coffee and registration, Minnesota Room

9:00AM - 9:15AM, Intro and organization, Memorial Hall

9:15AM - 10:00AM, Keynote: Marileen Dogterom, Memorial Hall, abstract

10:00AM - 10:45AM, Keynote: Avi Schroeder, Memorial Hall, abstract

10:45AM - 11:00AM, Break

11:00 - 12:00AM, Working groups intro, working group info, Memorial Hall

12:00PM - 1:00PM, Lunch, Memorial Hall

1:00PM - 2:30PM, sessions: Containers #1, Beyond the Bench #1, Building Blocks #1

2:30PM - 3:00PM, Break

3:00PM - 4:30PM, sessions: Containers #2, Applications #1, Cytoskeleton #1

4:30PM - 4:45PM, Break

4:45PM - 6:00PM, sessions: Building blocks #2, Applications #2

6:00PM - 8:00PM, Dinner, Memorial Hall

8:00PM - 9:00PM, Poster Session and drinks, The commons room, all poster abstracts

Tuesday, May 23

8:00AM - 9:00AM, Breakfast, Minnesota Room

9:00AM - 9:15AM, Intro and organization, Memorial Hall

9:15AM - 10:00AM, Keynote: Dora Tang, Memorial Hall, abstract

10:00AM - 10:45AM, Keynote: Sebastian Maerkl, Memorial Hall, abstract

10:45AM - 11:15AM, Break

11:15AM - 12:00PM, Working group meetings, working group info, location picked by each group

12:00PM - 1:00PM, Lunch, Memorial Hall

1:00PM - 2:30PM, sessions: Containers #3, Applications #3, Building Blocks #3

2:30PM - 3:00PM, Break

3:00PM - 4:30PM, sessions: Containers #4, Applications #4, Building Blocks #4

4:30PM - 4:45PM, Break

4:45PM - 6:00PM, sessions: Containers #5, Cell-Free Systems #1, Building Blocks #5

6:00PM - 9:00PM, Dinner and party at Malcolm Yards (included in your registration price, details below)

Wednesday, May 24

8:00AM - 9:00AM, Breakfast, Minnesota Room

9:00AM - 9:15AM, Intro and organization, Memorial Hall

9:15AM - 10:00AM, Keynote: Paul Freemont, Memorial Hall, abstract

10:00AM - 10:45AM, Keynote: John Glass, Memorial Hall, abstract

10:45AM - 11:30AM, Break

11:30AM - 12:30PM, Poster awards, special surprise guest 🐿️, conference group picture, Memorial Hall

12:00PM - 1:00PM, Lunch, Memorial Hall

1:00PM - 3:00PM, sessions: Cell-Free Systems #2, Building Blocks #6

3:00PM - 4:00PM, Working group meetings, working group info, location picked by each group

4:00PM - 5:00PM, closing remarks, Memorial Hall

Monday, May 22 sessions and keynotes

Monday, May 22, breakout sessions


Containers #1

Monday, May 22, Johnson room, 1:00PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Monday, May 22nd
Johnson room
presenter: Matt Good
all authors: M. Garabedian1, W. Wang3, R. Welles1, M. Guan2, Z. Su2, D. Rich2, B. Tu3, K. Sojitra4, J. Mittal4, D. Hammer2,3, M.C Good*1,3
Synthetic Organelles for Controlling and Augmenting the Functions of Cells
A grand challenge in synthetic biology is bottom-up construction of self-replicating bio-like systems. To achieve this, we must be able to characterize and redesign cellular processes in isolation and test their ability to re-integrate with and control the activities of living systems. Cells subcompartmentalization biochemical processes to enhance the rate and fidelity of these reactions. Numerous regulatory systems of the cell are partitioned to distinct membraneless compartments including nucleoli, nuclear speckles, stress granules and germ granules. These structures or biomolecular condensates are heterotypic assemblies often comprised of intrinsically disordered proteins (IDPs) and RNAs. Designer condensates, constructed from single proteins and which display controllable size, cargo capacity and controlled release functions promise a new strategy for building functionality into synthetic or augmented life. Using a variety of model disordered polypeptides, we have built synthetic organelle platforms that can be genetically encoded in cells and which enable dynamic sequestration-release of native enzyme for modular control of cellular behaviors. We determined the sequence rules for homotypic assembly of the RGG domain of the germ granule protein LAF-1, and re-engineered this protein to assembly and disassemble in response to enzymatic, optical and thermal stimuli. We have biophysically characterized these synthetic coacervates in vitro, in protocells and in living cells and achieved selective concentration of targets to these designer compartments via cognate interaction motifs embedded within the disordered scaffold and specific client. Recently we have begun to decipher the selectivity of IDP coacervation and identified pairs of polypeptides capable of forming discrete condensed phase in vitro and in living cells. Further we have expanded our delivery strategies both to both deploy these minimal organelle systems in a variety of cell types, and for non-genetic uptake of coacervates as signaling hubs. Designer synthetic organelles hold great promise both as linkable modules to build synthetic life and as embedded systems that facilitate programmable control of cellular decision making.

1:00PM - 2:30PM
Monday, May 22nd
Johnson room
presenter: César Rodriguez-Emmenegger
all authors: Cesar Rodriguez-Emmenegger*
Phagocytic Synthetic Cells to protect us against antibiotic-resistant) bacteria
We are devising a therapeutic and prophylactic technology that can be applied to biological interfaces (airways, wounds, implants, contact lenses, etc.) to kill antibiotic-resistant bacteria and thereby prevent or curtail infection severity. Our concept is based on Phagocytic Synthetic Cells (PSCs). The PSCs are a new type of synthetic vesicles decorated with stickers that bind specifically to epitopes on the surface of a bacterium, causing it to be engulfed into a phagosome where it is killed. This dual mode of action is inspired by phagocytosis. Engulfment occurs when the energy gain upon binding to the bacteria overcomes the elastic energy of the PSC’s membrane. This requires PSCs that are extremely flexible yet mechanically stable, properties that cannot be achieved simultaneously by state-of-the-art synthetic vesicles such as liposomes and polymersomes. The stickers must bind to bacteria via specific ultra-weak interactions that are collectively strong. It is important to use only a small number of stickers so that the membrane remains stealth to avoid off-target reactions. To operate with a low density of stickers, they must diffuse rapidly across the PSC’s membrane to the binding site, which requires high lateral mobility. To overcome these challenges, we formed the membranes by the self-assembly of zwitterionic Janus dendrimers and ionically-linked comb-polymers (iCPs) in which the hydrophobic and hydrophilic parts are linked by electrostatic interactions. The zwitterionic Janus dendrimers formed biomimetic membranes with higher flexibility than state-of-the-art lipid and polymer vesicles as needed for engulfment while displaying high chemical, thermal and mechanical stability. Then the periphery of these vesicles is decorated with aptamers that serve as stickers to enable specific capturing of the pathogens. During engulfment, the iCPs segregate to the phagosome where the close apposition with the engulfed bacterium allows their hydrophobic tails to insert, disrupting the bacterial membrane and bursting it. When the PSCs are contacted with Gram positive, negative and biofilm-forming bacteria (clinical isolates) they rapidly engulf them into a phagosome as observed by confocal microscopy. After 30 min, the membrane of the engulfed bacterium fuses with the one of the phagosome resulting in the death of the pathogen. This mechanism could be adjusted by the degree of polymerization and packing parameter of the iCPs. Remarkably, no interactions nor cytotoxicity was observed towards eukaryotic cells. PSCs are a supramolecular quasi-living therapeutic that achieves selectivity through the simultaneous action of multiple stickers and an antimicrobial mechanism based on disrupting the bacterial membrane, an element highly unsusceptible to bacterial evolution. By the same token, the mechanism of action does not provide a selective pressure leading to the emergence of resistance. Thus, we envision that the PSCs will be a safe, facile, drugless antimicrobial technology that can aid in how we protect from and combat antibiotic-resistant infections, one of the biggest global threats for which the world is not yet prepared.

1:00PM - 2:30PM
Monday, May 22nd
Johnson room
presenter: Jessica Lee
all authors: Jessica Lee, Fatma Pir Cakmak, Christine Keating
Combining models of membranes and intracellular condensates
Compartmentalization is crucial for providing localization/isolation of biomolecules, and favorable conditions for bioreactions in cells. Both membrane-bounded and membraneless organelles provide intracellular compartmentalization and are crucial in aspects of cellular life. However, the mechanism of how these organelles interact and influence each other remain largely unknown. Therefore, this research explores the interactions between simple experimental models of membranous and membraneless organelles. We are using coacervate droplets formed by LLPS of charged polymers as models of membraneless organelles, and have observed templated lipid membrane self-assembly around these droplets. The presentation will discuss that how the presence of coacervate droplets impacts membrane morphology, fluidity, and permeability.

1:00PM - 2:30PM
Monday, May 22nd
Johnson room
presenter: Alessandro Fracassi
all authors: Alessandro Fracassi*, Neal K. Devaraj
Nonlamellar lipid compartments for artificial cell development
Spatial organization is a defining feature of life. Living cells regulate many biological functions using lipid membrane compartments. Biomolecules can be concentrated and co localized to favor specific cellular processes while preventing undesired reactions. Although cell membranes are largely comprised of lamellar structures, lipid compartments often adopt various nonlamellar morphologies that are thought to favor important cellular functions such as membrane fission and fusion. Improved methods for controlling the structural organization of artificial model membranes would facilitate investigations into how membrane morphology affects biological functions. We are approaching this challenge by 1) developing synthetic lipids that spontaneously self-assemble into nonlamellar phases and 2) investigating bioconjugation strategies for biomolecule enrichment in the assemblies formed. We studied the atomic underpinnings of lipid nonlamellar assemblies through the synthesis of monoolein (MO) isosteres. Our results show that replacing the MO ester linkage between the hydrophilic headgroup and hydrophobic hydrocarbon chain with a thioester or amide functional group has a dramatic effect on the properties, phase, and morphologies of the assemblies formed. These results improve our understanding of the types of membrane architectures that can be built in artificial cells. We also developed methods to program nonlamellar lipid assemblies to recruit and concentrate biologically relevant molecules. We prepared artificial lipid compartments comprised of a zwitterionic gemini surfactant that spontaneously self-assembles into nonlamellar lipid sponge phase droplets in aqueous media. Such structures can be enriched with simple precursors of bioconjugation reactions, rendering the assemblies capable of confining biological molecules and pathways with high efficiency and in a programmable manner. These results improve our understanding of lipid mesophase assembly and may facilitate the development of nonlamellar compartments for biomedicine and bottom-up synthetic biology.

1:00PM - 2:30PM
Monday, May 22nd
Johnson room
presenter: Elisa Roberti
all authors: Elisa Roberti*, Eugenia De Remigis, Dario Cecchi, Gaia Petrucci, Francesco Bianciardi, Elisa Linda Petrocelli, Jyoti Sharma, Hilda Gomez Bernal, Stefano Palagi
Cell-inspired microrobots for sensing and targeting tumor tissue
Microrobots have size in the micrometer-to-millimeter range, and can move and perform specific tasks - in most cases under control by external fields (e.g. magnetic). The foreseen ability of microrobots to move in confined spaces of the human body makes them particularly interesting for a wide variety of medical applications, such as drug delivery. From this point of view, microrobots could represent a valid alternative or, more likely, a useful complement to nanovectors, whose transport mechanism is mostly based on diffusion and has low efficiency. Indeed, microrobots could be controlled and guided directly to the target, leading to new drug- or nanovector-delivery strategies capable of minimizing side effects on healthy cells and tissues. We aim at realizing cell-inspired microrobots that could move inside body tissues autonomously by sensing chemical or physical gradients. The microrobots consist of giant unilamellar vesicles (GUVs), in which active or responsive particles are loaded. We make the GUV-based microrobots by the inverted emulsion technique, where GUVs are formed by forcing water-in-oil droplets through an oil/water interface. GUVs of different composition are analysed in terms of dimensional distribution, stability, deformability, and particles-loading capabilities. In addition, the responsiveness of the GUVs and of the microrobots to external stimuli to external conditions (e.g. pH) is analysed. Finally, we aim at having them deform and move along chemical and physical gradients, thus obtaining chemotactic movement, by an interplay between the membrane and the encapsulated active or responsive particles.



Beyond the Bench #1

Monday, May 22, Ski-U-Mah room, 1PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Dr. Todd J. Treangen
all authors: Todd J. Treangen
Strengthening, Supporting, and Scaling DNA Screening Practices for Enhanced Biosafety and Biosecurity
While established protocols exist for synthetic sequence screening, predicting the biorisk of novel engineered organisms is a challenging task with no proven best practices nor comprehensive regulatory guidance. In this talk, I will cover SeqScreen, an open-source framework for enhancing biosecurity protocols to fill this need. SeqScreen leverages taxonomic and functional screening for identifying novel sequences of concen, and includes computational modules for mapping sequences to databases of known sequences of concern, such as Biological Select Agents and Toxins (BSATs). I will describe SeqScreen's utility with a few brief vignettes and cover the latest features and updates. To conclude, I will discuss the strengths and limitations of DNA screening approaches, and explore open challenges and opportunities for improved biosafety and biosecurity.

1:00PM - 2:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Rocco Mancinelli
all authors: Rocco Mancinelli
Biosafety commonalities between synthetic biology and planetary protection back contamination
The de novo construction of a functioning synthetic cell or engineering native cells for potential practical applications is a primary goal of synthetic biology and would mark a paradigmatic milestone in science. Although this was the domain of science-fiction just half-century ago, recent developments in synthetic biology may bring the bottom-up generation of a synthetic cell to reality. A key feature of a living synthetic cell is a functional cell cycle, in which DNA replication and segregation as well as cell growth and division are well integrated. Approaches to recreate these processes in a synthetic cell, based on natural systems and/or synthetic alternatives have been developed. A synthetic replicating cell would constitute a life form that would be new to Earth, and it is prudent that safety precautions be taken to ensure that it is contained during its synthesis and while its safety is being assessed. Samples from space and a synthetic cell both constitute a potential hazard to Earth, but both are part of important scientific endeavors. Samples of solar system bodies, whether they be from asteroids, planets, or moons, provide insight into the origins of the solar system and the potential for life to have evolved off Earth. The building of a synthetic cell provides insight into not only how cells function from the most fundamental level and origin of life, but also into how they can be used to produce a variety of applications important in medicine and the production of a broad range of products ranging from biofuels to cosmetics and medicines.

1:00PM - 2:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Nate Hoxie
all authors: Nate Hoxie*, Kate Adamala, Rocco Mancinelli, Becky Mackelprang, Tae Seok Moon, Kathryn Brink, Baris Avsaroglu, John Glass, Anton Jackson-Smith, Elibio Rech, Maya Hey, James Wagstaff
Biosafety in Modern Synthetic Biology: Historical Parallels and Risk Assessment
Research in synthetic biology and, in particular, research aimed towards creating synthetic organisms or resurrecting previously extinct organisms, carry inherent biorisks without established precedent from which to adapt a framework for safe experimentation. Accordingly, researchers, legislators, and funding institutions must draw lessons from a broad historical background, applying principles of risk assessment and minimization from other paradigmatically transformative technologies to inform and supplement existing biosafety and biosecurity guidelines. In this presentation, we examine the history of biosafety in synthetic biology with a particular focus on the rapidly accelerating progress towards engineering synthetic cells, explore a historical parallel in extraterrestrial sampling, and take initial steps towards defining set of guiding principles for responsible synthetic cellular research and engineering.



Building Blocks #1

Monday, May 22, Swain room, 1PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Monday, May 22nd
Swain room
presenter: LYNN J. ROTHSCHILD
all authors: Lynn J. Rothschild, NASA Ames Research Center Garrett Roberts Kingman
How extremophiles can aid Build-a-Cell
All life lives in a physical, chemical, and biological niche space. For a syncell, the chemical composition in which it is created, its temperature and physical movement (shaking), and the other biological entities it interacts with is its ecological niche. Since its origin, life has expanded into every niche on Earth where there is water available; so too, to be useful, syncells will ultimately need be able to move to other environments. Here we suggest a way forward for giving syncells these capabilities based on what is known about the natural evolution of extremophiles and work in our lab creating synthetic extremophiles, which we call “Hell Cells”.

1:00PM - 2:30PM
Monday, May 22nd
Swain room
presenter: Claudia Contini
all authors: Claudia Contini*
Artificial Cells: from Soft Matter to Cell-like Behaviours
My area of research focuses on developing artificial cell-like systems, known as protocells, using synthetic or hybrid molecular building blocks that can be fully controlled through design. These protocells mimic the fundamental structure and mechanisms of natural cells, which can help us better understand biological systems and mechanisms while creating a wide range of functional applications. One of the most exciting challenges in chemical engineering is to develop artificial cell-like systems that can move autonomously and directionally in response to changes in their environment, mimicking biological communication and organization. Creating a life-like system that can be controlled and understood will advance our understanding of the origin of life and drive scientific discovery. Engineering custom-made artificial cells that can move autonomously and directionally will have many applications in clinical and industrial settings. This new research area will lead to the development of artificial-cell devices that are powered by biological compounds and can deliver therapeutics directly to specific targets. Additionally, motile artificial cells could be used for bioremediation or generate artificial tissues and materials through controlled spatiotemporal self-organization.

1:00PM - 2:30PM
Monday, May 22nd
Swain room
presenter: Scott Hansen
all authors: Benjamin Duewell, Naomi Wilson, Sophia Doerr, and Scott Hansen*
A bottom-up approach for deciphering mechanisms of spatial organization in cell signaling
The ability of cells to transiently alter the concentration and spatial distribution of signaling molecules is a hallmark of cellular organization. At the plasma membrane, spatial heterogeneity in cell signaling can emerge from biochemical reactions involving phosphatidylinositol phosphate (PIP) lipids, kinases, phosphatases, and small GTPases. Interconnected positive and negative feedback loops control the communication between these distinct classes of molecules to drive polarization, cortical oscillations, and transient spikes in signaling. Using supported membrane technology, optogenetics, and single molecule imaging, we examine how membrane proximal signaling reactions drive cellular organization. This work provides new insight needed to biochemically reconstitute minimal self-organizing systems that control cell polarity.

1:00PM - 2:30PM
Monday, May 22nd
Swain room
presenter: Yeray Folgar-Cameán
all authors: Yeray Folgar-Cameán,* Richard Booth, Ignacio Insua, Sahnawaz Ahmed, Alicia Rioboo and Javier Montenegro
Supramolecular peptide amphiphiles towards a functional cytoskeleton model
The cytoskeleton is a complex network of protein filaments that plays an essential role in the functionality of eukaryotic cells, providing mechanical stability, adaptability and motility. Bottom-up approaches by synthetic surrogates towards the replication of this complex organelle can help us to understand and possibly transfer the mechanisms underlying this biomolecular scaffold.[1] Here, we report the supramolecular polymerisation of simple amphiphilic peptide monomers that are intended to replicate certain structural and potential functional properties of the cytoskeleton. In one hand, we have focused in controlling the hierarchical one-dimensional assembly of synthetic nanofibers in micron scale. In addition, we have also studied the potential functional consequence of entrapping this fibrillating system into water in oil droplets. The system was accomplished by the in-situ generation of a peptide amphiphile via the oxime condensation of two dormant fragments, which triggered a physically autocatalytic reaction that induced the assembly of the corresponding microfibrillar network.[2] By encapsulating the system into aqueous micro-droplets, it was possible to study the consequence of the supramolecular polymerization in a confined environment, resulting in a series of biomimetic responses by the droplets. Firstly, the spatial reorganisation of the supramolecular network triggered the selective uptake of exogenous chemicals by the droplets. It was also found that this fiber-mediated internalisation of chemicals modulated the coalescence and the contact-based exchange of cargoes between droplet populations. This was tested by incorporating a two-step enzymatic cascade into the droplets, which required exchange of substrate and product between droplet populations to produce a fluorogenic reporter at the end of the biocatalytic pathway. Overall, these simple and hierarchically self-assembling monomers emulated complex functions of the natural cytoskeleton without the need of a sophisticated molecular machinery currently found in living cells. These advances contribute to the bottom-up design and fundamental understanding of supramolecular biomimetic assemblies with life-like properties and stimuli-responsive behaviour. References [1] Mohrdieck C., Dalmas F., et al. Small 2007, 3(6), 1015-1022. [2] Booth R., Insua I., et al. Nat. Commun. 2021, 12(1), 6421.



Containers #2

Monday, May 22, Johnson room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Monday, May 22nd
Johnson room
presenter: Siddharth Deshpande
all authors: Niels Appelman, Chang Chen, Ivar Gruppen, Siddharth Deshpande*
Dance of Droplets: Spontaneous Differentiation-Repulsion Behaviour in Two-Component Systems
An important exercise for understanding the emergent behavior in biology is studying minimal synthetic systems under non-equilibrium conditions. With the ongoing efforts on constructing synthetic cells, one of the hallmark properties to recreate are motility and chemotaxis on individual as well as population level. Current biomimetic systems involving directed motion and collective migration require at least three components: two immiscible solvents to form an emulsion, and external agent(s) to give rise to asymmetric interactions. In this talk, I will present the most minimal system thinkable, consisting of only two components, in the form of micron-sized oil droplets dispersed in a finite water reservoir. We report spontaneous emergent dynamics of oil droplets at the air-water interface, in the form of physical differentiation of droplets in a stochastic manner that leads to an immediate repulsive behavior in the surrounding droplets. Through experiments and modelling, we show that this recurring differentiation-repulsion cycle is a result of Marangoni flows and its coupling with evaporative flux. Finally, I will illustrate the potential of this platform by demonstrating control over the event frequency, its use as ‘Marangoni tweezers’, and proof-of-principle reorganization of soft matter assemblies. The presented collective behavior with exceptional chemical simplicity makes it amenable for developing self-assembled biomimetic and bioengineered structures.

3:00PM - 4:30PM
Monday, May 22nd
Johnson room
presenter: Tommaso Fraccia
all authors: Tommaso P. Fraccia*, Tony Z. Jia, D. Cuvelier, Nicolas Martin
Oligonucleotide ligation and compartment-content coupling in potentially evolvable coacervates
Coacervates droplets have been proposed as promising synthetic membrane-less compartments for their ease of formation, ability to segregate biopolymers and sustain biochemical reactions. Anyway, reproducing a coupling between information content and compartments physico-chemical properties represents a still unachieved fundamental step for the realization of evolvable synthetic protocells. Here we report the surprising propensity of DNA oligonucleotide ligation and assembly in liquid crystals (LC) to sustain strong coupling between the structural compartment properties and their nucleic acids content in coacervates. Indeed, we have recently shown that mixtures of oligonucleotides and cationic peptides produce a rich ensemble of phase-separated coacervates, including liquid crystalline coacervate microdroplets [1]. Such phases dramatically depend on oligonucleotide properties, such as sequence, length and secondary structure; and extend the range of different states that coacervates can display as function of environmental parameters, such as ionic strength and temperature [2]. Secondly, these chemically-enriched phases provide a favorable environment for the non-enzymatic ligation of end-reactive oligonucleotides via carbodiimide chemistry, so that long polynucleotides are produced in situ, while the condensation reaction is only poorly effective in the dilute supernatant [3]. Significantly, polymerization yields are strongly correlated with the nature of the phase in which it occurs. Moreover, the phase-dependent growth of the DNA population towards longer polymers during ligation further induces structural changes to the compartments themselves, increasing their stability and causing transitions in the inner coacervate phase. Such positive feedback connection between compartment structure and in situ reactions offers a promising platform for the development of evolvable synthetic protocellular systems. References: [1] Fraccia T. P. & Jia T. Z. (2020) ACS Nano, 14, 15071, DOI: 10.1021/acsnano.0c05083 [2] Fraccia T. P. & Jia T. Z. (2020) Crystals, 10, 964, DOI: 10.3390/cryst10110964 [3] Fraccia T. P. & Martin N. (2022) ChemRxiv, DOI: 10.26434/chemrxiv-2021-zg20r

3:00PM - 4:30PM
Monday, May 22nd
Johnson room
presenter: Hannes Mutschler
all authors: Elia Salibi, Benedikt Peter, Annalena Salditt, Leonie Karr, Kristian Le Vay, Petra Schwille, Dieter Braun, Hannes Mutschler*
Non-isothermal conditions as drivers of protein-free RNA replication
The proliferation of modern cells relies on the complex orchestration of hundreds of evolved proteins. In contrast, it is unclear how primitive precursors of life may have propagated before the emergence of translation. In this talk, we will explore how non-equilibrium conditions such as temperature cycling and temperature gradients may have acted as early drivers of RNA-catalyzed replication of sequence information or facilitated the spread of replicators within protocell populations.



Applications #1

Monday, May 22, Ski-U-Mah room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Allen Liu
all authors: Hossein Moghimianavval, Kyle Loi, Allen Liu
Contact-Dependent Synthetic Cell Communication
Cell-cell communication and signal transduction through direct cell-cell contact are found ubiquitously in biology. Despite the prevalence of direct cell-cell communication at membrane contact sites, it is difficult to engineer functions at membrane-membrane interfaces. Here, we present programmable activation of a split bioluminescent protein, NanoBiT, at the membrane-membrane interface between synthetic cells. We utilize a peptide-protein pair called SpyTag and SpyCatcher as adapter proteins to mediate functional reconstitution of NanoBiT. We illustrate an example of downstream protein activation through a designed light-based communication system by using a pair of photoactivatable proteins, iLID and SspB inside synthetic cells and demonstrate iLID-SspB dimerization triggered by reconstituted NanoBiT bioluminescence.

3:00PM - 4:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Michael Booth
all authors: Michael Booth*
Remote-controlled synthetic cells
Synthetic cells, lipid membrane-bound compartments that mimic living cells, are a promising technology for studying living systems and as drug delivery vehicles. However, real-world application of synthetic cells requires methods of controlling their function for spatiotemporal activation and to reduce off-target effects. To address this, we are developing methods to externally control these soft biomaterials with various biologically- and medically-applicable stimuli, including multiple wavelengths of light, by chemically modifying the nucleic acid component. We are using our synthetic cells to mimic living processes and form controllable interfaces between living and non-living materials.

3:00PM - 4:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Béla Frohn
all authors: Béla Frohn*, Shunshi Kohyama, Leon Babl, Petra Schwille
Bottom-Up Protein Engineering: Screening proteins for complex emergent functions in synthetic cells brings protein design to the next level.
Recently, the utilization of Machine Learning (ML) based methods has led to astonishing progress in protein design, raising high hopes in synthetic biology to build custom protein-based machineries adapted to the individual needs of specific environments. Utilizing such tailored protein machineries instead of machineries found in and pre-optimised for existing organisms has the potential to revolutionize bottom-up synthetic biology. Towards this goal, however, protein design must be pushed to the next level: Up to now, the design of proteins with emergent functions, that is, functions that require higher-order molecular interactions and can only be observed on a macroscale, such as biological pattern formation or membrane deformation, is still in its infancy. A major challenge for this next step of protein engineering is the lack of high-throughput screening pipelines to test for emergent functions of de novo designed proteins in vitro, adapting the typical design-and-screen iterations of protein design projects to bottom-up synthetic biology. Here, we present a comprehensive pipeline enabling such design and screening of proteins with an emergent function, based on bottom-up constructed synthetic cells. We first use ML to design novel distant homologs of a protein whose emergent function is biological pattern formation, and then use a cell-free expression system to produce and encapsulate the designed proteins inside lipid droplets, enabling us to easily screen for pattern formation inside these synthetic cell models. This pipeline allows us to find novel distant homologs that form patterns in vitro, to our knowledge the first design of a protein with such a complex emergent function. Moreover, the protein performing best in the bottom-up screening can fully functionally substitute the wildtype gene in Escherichia coli, leading to a phenotype indistinguishable from the wildtype bacterium. This not only is the first example of complete substitution by a ML-designed artificial homolog in vivo to the best of our knowledge, but also shows the great potential to use bottom-up constructed cell-mimicking systems to screen for complex emergent functions. These results raise great hopes for the next level of protein engineering, where the synthesis of computational design and bottom-up experimental screening will be used to engineer cellular functions.

3:00PM - 4:30PM
Monday, May 22nd
Ski-U-Mah room
presenter: Pavana Khan
all authors: Pavana Khan*, Aaron Engelhart
Aptamer-based Readouts for Isothermal Pathogen Detection
The ongoing SARS-CoV-2 pandemic highlights the importance of early, rapid and widespread pathogen detection tests that are easily accessible and available to all. While real-time-PCR methods are the most well-established tests for SARS-CoV-2, the requirements for expensive reagents and equipment, skilled technicians and more complicated workflows impede simple and fast turnaround of diagnostic results in point-of-care and home settings. Towards meeting this need, we previously developed Apta-NASBA, an engineered diagnostic technique that employs a multienzyme cascade to couple fluorescent aptamer tagging to nucleic acid sequence based amplification (NASBA) (Aufdembrink, Khan, et al, RNA (2020) 10.1261/rna.075192.120). We sought to extend the power of our aptamer tagging approach to a technique that takes advantage of Recombinase Polymerase Amplification, a highly robust isothermal amplification method that occurs in phase-separated, membraneless organelle-like domains. We have extended RPA to develop Apta-RPA: a one-pot, isothermal, nucleic acid amplification-based detection test that is rapid, sensitive, and specific. With a detection limit of 2 copies/reaction and a time to detection of 10 minutes, Apta-RPA couples a fluorescent aptamer to the produced amplicon. T7 RNA polymerase-mediated transcription of RPA amplicon-aptamer-fusions allows real-time fluorescence detection. We demonstrate that the Apta-RPA assay has a low limit of detection and can be used to detect SARS-CoV-2 and other pathogens such as E.coli. Furthermore, we show that it is a useful technique for multiplexed detection of pathogens in a one-pot reaction and for differentiation between multiple SARS-CoV-2 variants without the need for expensive sequencing methods. Apta-RPA is an inexpensive, low-cost tool for rapid, sensitive, and specific detection of SARS-CoV-2 and other pathogens that showcases the value of synthetic biology-based cell-free techniques for diagnostics.



Cytoskeleton #1

Monday, May 22, Swain room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Monday, May 22nd
Swain room
presenter: Lorenzo Di Michele
all authors: Lorenzo Di Michele
Synthetic cells from smart nucleic-acid condensates
Synthetic cells require micro-compartmentalised architectures to regulate transport and establish internal heterogeneity. While membrane-based synthetic-cell scaffolds are common, membrane-less implementations relying on hydrogels or coacervate droplets are becoming increasingly popular due to their robustness and programmability. In this talk, I will discuss the use of nucleic acid-based condensates as scaffolds for building synthetic cells. I will show that condensates self-assembling from amphiphilic DNA nanostructures can host a variety of biomimetic functionalities, including regulated transport, cargo capture/release, triggered assembly and disassembly, and the in-situ synthesis of functional RNA. To recapitulate the internal compartmentalisation seen in biological cells, I will discuss how reaction-diffusion processes and micro-phase separation can be used to create distinct, chemically addressable domains within the SCs, allowing spatial engineering of functionalities and localised morphological responses.

3:00PM - 4:30 PM

Swain room
presenter: Shashank Shekhar
all authors: Shashank Shekhar
Turning actin treadmilling on its head – assembling filaments from their ‘wrong’ end
Actin is an essential protein. For over two decades, intracellular actin filaments have been thought to elongate at their barbed ends and depolymerize from pointed ends. This process, referred to as “treadmilling” has formed the central bedrock of our understanding of actin dynamics. Our recent results have however suggested that the treadmilling dogma might not always hold true. Using a combination of in vitro multicolor single molecule and bottom-up reconstitution experiments, we have discovered two new activities that call for reevaluation of the treadmilling dogma. First, we discovered the first-ever pointed-end polymerase VopF that processively polymerizes filament pointed ends in cells and from purified proteins. Further, VopF accelerates polymerization in presence of profilin and is a mechanosensitive protein - its rate of polymerization increases under pN-range pulling forces. Second, we have recently discovered that twinfilin, a member of the cofilin family of proteins, induces depolymerization of filament barbed ends. Interestingly we find that the depolymerase twinfilin, polymerase formin and blocker CP form a multicomponent complex at the filament barbed end. Taken together, our findings call for taking a fresh look at actin treadmilling and will have important implications on our ability to bottom-up reconstitute actin dynamics for use in synthetic systems.

3:00PM - 4:30PM
Monday, May 22nd
Swain room
presenter: Andreas Fink
all authors: Andreas Fink*, Sunnatullo Fazliev, Charlotte R. Doll, Ana Yagüe Relimpio, Yannik Dreher, Joachim P. Spatz, Kerstin Göpfrich, and Elisabetta Ada Cavalcanti-Adam*
Extracellular cues govern shape and cytoskeletal organization in giant unilamellar lipid vesicles
Actin organization is a key process in cell polarization and directed cell motility, affecting cell shape, but in turn also being affected by external geometrical cues. Here, we present a minimal model to study actin organization and vesicle stiffness in adherent, deformed giant unilamellar vesicles (GUVs) in a bottom-up synthetic approach. We utilize streptavidin micropatterned surfaces to generate external cues to adhere and deform biotin functionalized GUVs. Micropatterning of adhesive ligands is commonly used to study single-cell behaviour and polarization in a well-defined environment. In this work we find that cytoskeletal proteins like actin organize due to geometric constraints in confinment. We further studied the effect of various actin bundlers in such deformed vesicles. Actin filaments crosslinked by fascin orient along the major axis in the lumen of deformed GUVs when the bundle length is similar to the size of the major axis. On the other hand, filaments bundled by crowding agents like dextran localize at the membrane. These show further organization along the adhesion site of the deformed GUVs, likely induced by the strong curvature at the contact area with the pattern. Further, we observe that GUVs deform less in the presence of actin bundled by dextran due to the localization of actin filaments at the GUV membrane. This leads to the conclusion that GUVs can be stabilized mechanically by organizing actin in a cortex-like manner around the membrane. These results underline the importance of actin filament organization on the mechanical properties of synthetic cells. They further show the interplay of organization induced by molecular crowding, as well as deformation. The here presented system can thus serve as a versatile platform to gain further understanding in the delicate interplay of cell adhesion and cytoskeletal organization.

3:00PM - 4:30PM
Monday, May 22nd
Swain room
presenter: Yash K Jawale
all authors: Reza Amini Hounejani, Yash Kiran Jawale*, Eli van der Sluis, Beatriz Orozco Monroy, Koen van Gent, Marileen Dogterom
Designing a minimal DNA segregation system for synthetic cells
Living cells have evolved various mechanisms to faithfully transfer their genomic material to the next generations. Chromosomal DNA segregation is often based on active segregation employing cytoskeletal filaments such as microtubules. We are interested in designing a minimal system for active DNA segregation for application in synthetic cells. Given that the eukaryotic mitotic spindle is highly complicated, we investigate the use of bacterial segregation systems instead. Here we discuss, the possible employment of three bacterial cytoskeletal filaments, two tubulin-like: bacterial microtubules (bMTs) and TubZ filaments, and one actin-like: ParM filaments. The centromeric tubC and parC plasmid DNA attach to the TubZ and ParM filaments with the help of adaptor proteins TubR and ParR respectively. We aim to achieve temporal control over DNA segregation upon completion of DNA replication using optogenetic tools. We have fused filament- and DNA binding- domains of the adaptor proteins with the photo-switchable iLID-SspB protein pair. For each system, we assessed several key requirements: the feasibility of in vitro reconstitution of dynamic filaments with the ability to bind DNA, the potential for encapsulation in liposomes, and the possibility of successful protein expression in the PURE system. We propose possible DNA segregation systems for a synthetic cell based on our findings. The spatiotemporal control using photo-switchable proteins will help to impose external cell-cycle control.

3:00PM - 4:30PM
Monday, May 22nd
Swain room
presenter: Marcos Arribas
all authors: Marcos Arribas Perez* and Gijsje H. Koenderink
Influence of the membrane shape in the organization of actin networks reconstituted in GUVs
Animal cells continuously regulate their shape to adapt to external stimuli and perform vital functions such as cellular nutrition, migration and division. These shape changes are mainly controlled by a complex interplay between the plasma membrane and the actin cytoskeleton. The architecture of the actin cytoskeleton is dictated by a myriad of actin binding proteins that can for instance facilitate actin polymerization, promote or prevent filament elongation and crosslink actin filaments. In addition, myosin motor proteins interact with the actin cytoskeleton and exert force that remodel the filament arrangement. The molecular complexity and dynamic behavior the actin cytoskeleton makes it extremely difficult to understand how the actin cytoskeleton works. In this context, bottom-up reconstitution of minimal systems contributes to gain new insights into fundamental biochemical and biophysical mechanisms behind the shape changes induced by the actin cytoskeleton. Here we use emulsion droplet interface crossing encapsulation (eDICE) to produce giant unilamellar vesicles (GUVs) with an actin cytoskeleton. By modifying the actin binding proteins co-encapsulated in the GUVs together with actin we can tune the architecture of the actin cytoskeleton inside the vesicles. Moreover, we explore how the geometry of the GUVs and the membrane curvature influence the spatial reorganization of the encapsulated actin cytoskeleton using microfluidic traps and curvature-inducing proteins. Such spatial reorganization of the actin networks would allow us to create symmetry breaking events which are extremely important in processes such as cell division and migration.



Building blocks #2

Monday, May 22, Johnson room, 4:45PM - 6:00PM
Zoom link to this session; to get password register as online participant.

4:45PM - 6:00PM
Monday, May 22nd
Johnson room
presenter: Laura Heinen
all authors: Laura Heinen*, Bert Poolman
Robust Cell-Fueling Strategies: A Steady Feed of ATP Across the Membranes of Synthetic Cells
The Synthetic Cell Community needs reliable and practical solutions to supply their artificial life-like vesicle systems with energy. Our engineered synthetic cell envelopes require energy for various reasons, mainly to fulfill active functions e.g., to grow, divide, process information and synthesize the basic building blocks. Ideally, this energy is fed in a continuous and autonomous fashion and the corresponding waste products are constantly removed or recycled. Intact biological cells usually absorb sugars or light, convert this energy, and store the energy in the form of adenosine triphosphate (ATP). However, for example, certain pathogenic intracellular bacteria and organelles also rely on energy parasitism. This means that they take up ATP from the environment to fuel their energy-demanding functions. We make use of this concept and developed synthetic vesicles that can shuttle ATP across their membrane. In my talk I will present ATP cross-feeding synthetic organelles. One population of vesicles produces and exports ATP while a second population of vesicles takes up the ATP and uses this chemical energy to fuel other luminal ATP-dependent processes. The hydrolyzed ATP feeds back into the first vesicle population where it will be recycled, and the interdependent metabolic cycle can start again. The vesicles are a future platform technology to fuel ATP-dependent processes in a sustained fashion and with wide applications in synthetic cells and as biological nanoreactors.

4:45PM - 6:00PM
Monday, May 22nd
Johnson room
presenter: Aaron Engelhart
all authors: Tanner Hoog, Lauren Aufdembrink, Matthew Pawlak, Katarzyna Adamala, Aaron Engelhart
Bridging hard and soft materials with switchable biopolymer electron transfer catalysts
We will show recent results in which we demonstrate that conditionally active electron transfer catalysts can be constructed from DNA. The catalysts are comprised of two strands, one of which can be selectively switched between a G-quadruplex and duplex or single-stranded conformations. This switching ability arises from our recent discovery that perchlorate, a chaotropic Hofmeister ion, selectively destabilizes duplex over G-quadruplex DNA. By varying perchlorate concentration, we show that the device can be operated as a switch or signal amplifier. State switching can be achieved in three ways: thermally, by dilution, or by concentration. In each case, when operated in the presence of the cofactor hemin, the device catalyzes electron transfer in only the G-quadruplex state. Examples of secondary-structure remodeling are observed in life as well. Nucleic acids exhibit considerable polymorphism in biological systems, particularly in sequences that can form G-Quadruplexes. Several proteins can remodel G-quadruplexes, and both energy-dependent (i.e., helicases) and energy-independent systems that can do so have been reported. ATPdependent helicases are known to unwind G-quadruplex structures, and the RNA-binding protein Lin28 has been shown to unfold G-quadruplexes without the requirement for ATP. The structure switching performed by Hofmeister ions in this work thus amounts to a bioinspired means of performing such remodeling, which in turn enables a functional behavior (electron transfer). As the system here functions at >1 order of magnitude higher salt concentration than typical biopolymer-based catalysts, it presents a potential means of bridging "soft" biopolymer-based materials and "hard" inorganic materials, such as nanoelectronics.

4:45PM - 6:00PM
Monday, May 22nd
Johnson room
presenter: Jose L. Alejo
all authors: Jose L. Alejo*, Dylan Girodat, Michael Hammerling, Jessica A. Willi, Michael C. Jewett, Kate P. Adamala
Directed evolution reveals the flexibility of ribosome motion
Controlled domain motion is critical to the function of many biological macromolecules. During ribosome translocation across mRNA, the small subunit head domain swivels, relocating the tRNA and mRNA substrates. The head domain then reverses this swivel, leaving the ribosome in its post translocation state. We have studied the flexibility of the two rRNA hinge regions which this head domain swivels about by carrying out in vitro directed evolution of hinge variant ribosomes. Targeted molecular dynamics simulations of the translocation of variants were carried out to research the malleability of the process. Interestingly, hinge, head domain and tRNA trajectories show considerable variability during forward swivel and reverse swivel. These results suggest the process of translocation is far more conformationally flexible than previously understood.



Applications #2

Monday, May 22, Ski-U-Mah room, 4:45PM - 6:00PM
Zoom link to this session; to get password register as online participant.

4:45PM - 6:00PM
Monday, May 22nd
Ski-U-Mah room
presenter: Oskar Staufer
all authors: Oskar Staufer
Bottom-up assembly of synthetic cell-based tissues and their application for immunotherapy
Synthetic biology has revolutionized the way we approach tissue engineering and regenerative medicine, offering innovative tools for the creation of artificial tissues from the bottom-up. One of the most exciting and promising approaches involves the use of synthetic cells that can self-assemble into higher order multicellular systems. I present a simple and scalable process to construct synthetic cells, which form dense macroscale structures interconnected by DNA-aptamer networks for programmable and hierarchical self-assembly. The synthetic cell can be instructed to assemble into artificial tissues with precise spatial organization and distinct mechanical features, allowing for the creation of sophisticated multicellular systems. To demonstrate the application potential of the synthetic cell tissues in the field of immunotherapy, I showcase how they can be engineered to mimic the structure and function of natural lymph nodes, for instance, by interacting with natural human immune cells to induce adaptive immune responses. The programmability of these systems allows for the precise tuning of immune cell activation and differentiation, enabling the creation of custom-designed synthetic tissues for specific applications. The potential for these synthetic tissues to interact with natural human cells and be used in immunotherapy represents a major step forward in our ability to harness the power of synthetic biology for real-world applications.

4:45PM - 6:00PM
Monday, May 22nd
Ski-U-Mah room
presenter: Bo Zheng
all authors: Jiqin, Yao, Bo Zheng*
Communicating Artificial Cells
In this talk I will introduce the hydrogel based artificial cells, which are constructed by covalently immobilizing the proteinaceous factors of cell-free system onto the polymer backbone of functionalized hydrogel particles. The experiment demonstrated the ability of the artificial cells to communicate with each other via mRNA and to communicate with bacteria. The work illustrates the potential antibacterial and antibiofilm applications of the hydrogel based artificial cells.

4:45PM - 6:00PM
Monday, May 22nd
Ski-U-Mah room
presenter: Jochen Estebano Hernandez Bücher
all authors: Jochen Estebeno Hernandez Bücher (1), Oskar Staufer (1), Lukas Ostertag (1), Ulrike Mersdorf (2), Ilia Platzman (1) and Joachim P. Spatz (1) (1) Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany (2) Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, D 69120, Heidelberg, Germany
Bottom-up assembly of target-specific cytotoxic synthetic cells
Immunotherapy is a rapidly evolving field that has become the fourth pillar of cancer treatment by harnessing the immune system's specificity and cytotoxicity. However, the current top-down genetic engineering approaches, such as CAR-T cell technology, have significant limitations in terms of their turn-over time, cost-effectiveness, versatility, and off-target side effects. To overcome these challenges, we propose a bottom-up approach using synthetic biology to assemble fully synthetic immune-inspired cells from predefined molecular components. In our study, we used giant unilamellar vesicles (GUVs) as the building blocks for the synthetic immune cells. By combining the Fas-ligand-based apoptosis-inducing immune cell module with an antibody-mediated cellular cytotoxicity-inspired system, we created target-specific cytotoxic immune cells that could identify leukemia cells by specific surface antigens. To minimize unspecific interactions with non-target cells, we coated the membrane surface with polyethylene glycol (PEG). We characterized the morphology of the target-specific cytotoxic synthetic cells using fluorescence confocal microscopy and their cytotoxicity towards cancer cell lines using microfluidics, live cell, and flow cytometry. We also examined their surface properties using transmission electron microscopy (TEM) and dynamic light scattering. TEM analysis showed that the GUV surface composition and decoration could be precisely adjusted, demonstrating a high degree of control over synthetic immune cells. Most notably, we found that the synthetic immune cells could bind to cancer cells and form a stable interaction, eventually inducing cell death in their targets. Moreover, we showed that these processes could be tuned by adjusting the PEG and antibody decoration. Overall, this study demonstrates the potential of bioinspired construction of effector immune cells from defined molecular building blocks, which may provide a cost-efficient and controllable alternative to natural-cell based therapies. Our approach also enables learning-by-building approaches in synthetic immunology, allowing for a deeper understanding of cellular phenomena as molecularly defined systems.



Monday, May 22, keynote lectures
Zoom link to this session; to get password register as online participant.


Marileen Dogterom

Monday, May 22, Memorial Hall, 9:15AM - 10:00AM

A minimal cytoskeleton for synthetic cells

abstract



Avi Schroeder

Monday, May 22, Memorial Hall, 10:00AM - 10:45AM

Synthetic cells as platforms for drug delivery

Medicine is taking its first steps toward patient-specific cancer care. Nanoparticles have many potential benefits for treating cancer, including the ability to transport complex molecular cargoes, including siRNA and protein, as well as targeting specific cell populations.
The talk will explain the fundamentals of nanotechnology, from ‘barcoded nanoparticles’ that target sites of cancer where they perform a programmed therapeutic task. Specifically, liposomes diagnose the tumor and metastasis for their sensitivity to different medications, providing patient-specific drug activity information that can be used to improve the medication choice.
The talk will also describe how liposomes can be used for degrading the pancreatic stroma to allow subsequent drug penetration into pancreatic adenocarcinoma and how nanoparticle’ biodistribution and anti-cancer efficacy are impacted by the patient’s gender and, more specifically, the menstrual cycle.
The evolution of drug delivery systems into synthetic cells, programmed nanoparticles that have an autonomous capacity to synthesize diagnostic and therapeutic proteins inside the body, and their promise for treating cancer and immunotherapy, will be discussed.



Monday, May 22 poster session

Posters should be no bigger than 36" x 48".

Posters will be grouped by session

 

Session Containers: liposome, microfluidics, coacervates etc. posters


Abbey Robinson

Abbey Robinson*, Jessica Lee, Anders Cameron, Christine D Keating, Katarzyna P. Adamala

poster number 10

Cell-free expressed membraneless organelles sequester RNA in synthetic cells

Compartments within living cells create specialized microenvironments, allowing for multiple reactions to be carried out simultaneously and efficiently. While some organelles are bound by a lipid bilayer, others are formed by liquid-liquid phase separation, such as P-granules and nucleoli1–3. Many of these membraneless condensates are formed by intrinsically disordered proteins (IDPs) that lack a discrete structure. LAF-1, an RNA helicase found in Caenorhabditis elegans P-granules, contains an intrinsically disordered region termed the RGG domain which plays a strong role in promoting phase separation4. Engineering compartments within synthetic cells facilitates the creation of more complex and multifaceted synthetic life-like systems. We describe a synthetic cell expressing phase-separating protein to form artificial membraneless organelles that are capable of sequestering RNA and reducing protein expression. We create complex microenvironments within synthetic cell cytoplasm, and introduce a tool to modulate protein expression in synthetic cells.


Anastassiya Schramm

Anastassiya Schramm*, Dr. Ilia Platzman, Prof. Dr. Joachim Spatz

poster number 13

Design of Stimuli-Responsive Multicompartment Vesicles for Immunological Applications

Programmed Death-Ligand 1 (PD-L1) together with its receptor, Programmed Cell-Death Protein 1 (PD-1) are crucial members of a PD-L1 - PD-1 checkpoint, which plays a major role in downregulating the immune system. A wide range of tumors has evolved to express PD-L1 on their surface, as well as on their exosomes in order to evade the immune system response. After prolonged interaction with this protein, T-cells become exhausted due to the continuous blockade of their PD-1 receptors and lose their “killing” ability. As a result, the body cannot effectively resist the infection or tumors, which can lead to the shutdown of the immune system. A current method to overcome this problem is by using checkpoint inhibitors, such as anti-PD-L1 or anti-PD-1 antibodies. Despite certain improvements in T cell responses, the major drawbacks of antibody therapy are its non-specificity, which leads to serious side-effects and autoimmune response, as well as its high cost. Therefore, finding a safer and more affordable solution would greatly benefit the field of immunomodulation and checkpoint inhibition. In this project, we propose a modular multicompartment trigger-responsive system that can potentially inhibit PD-L1 on the tumor and its exosomes. The system consists of multicompartment lipid vesicles, functionalized with NIR-responsive gold nanorods and magneto-responsive iron nanoparticles, which enable the rupture of the vesicle and its manipulation in space. Furthermore, the presence of tumor-recognition ligand on the vesicle is expected to provide the specificity for the selected tumor, while components such as CD47 and PEG would prolong the circulation time in the system and prevent unspecific interactions. Finally, selected immunomodulation agents could potentially facilitate the blockade of PD-L1 on the tumor cells and their exosomes. This modular vesicle, when injected in proximity to the tumor, could be moved to a location of interest through the magnetic field and then ruptured with the NIR light. The irradiation would cause the release of the immunomodulation components, which could restore the activity of the T cells and thereby recover the immune system.


Ashley Tafoya

Ashley N. Tafoya*, Aidira D.Y. Macias Gonzalez, Jacqueline De Lora, Nick J. Carroll, Gabriel P. López

poster number 14

Comparative study of methods for making cell-like compartments containing membrane-less organelles

Compartmentalization is an essential hallmark of living cells, allowing for control over distinct inter-cellular and intra-cellular biological functions. A major obstacle to the fabrication of synthetic cells is the ability to engineer membrane-bound and membrane-less synthetic organelles in a cell-mimicking environment. Similar to the proteins that comprise membrane-less organelles, elastin-like polypeptides (ELPs) contain regions of disorder, are stimulus-responsive proteins, and undergo liquid-liquid phase separation (LLPS), allowing them to function as components of synthetic membrane-less organelles. Finding a method for forming giant unilamellar vesicles (GUVs) that are mechanically and chemically stable enough to incorporate ELPs would allow for a simplified cell model to be studied. It would be a step closer to engineering a life-like structure from nonliving matter for the ultimate goal of better understanding natural cells. Our goal is specifically to load an ELP into a synthetic cell-like architecture to make synthetic membrane-less organelles. This work investigates several encapsulation techniques for this purpose. The three techniques studied here include: (1) one-pot production of droplet stabilized GUVs relying on a charge-mediated fusion of SUVs inside of a surfactant-stabilized droplet which can be released to an aqueous environment as a GUV, (2) glass capillary microfluidic production of monodisperse GUVs, and (3) formation of supported lipid bilayers on porous silica microparticles. The one-pot platform is a simple and straightforward method with capacity to be scaled up and to create complex GUV-based synthetic cells in physiologically relevant environments. The microfluidics platform utilizes a water-oil-water (W/O/W) based double emulsion, which leads to dewetting of the oil phase creating monodisperse GUVs. The porous silica particles provide support for the lipid bilayer while allowing for the encapsulation of biological components within the pores of the beads. This study investigates the flexibility of each method for incorporating proteins and nucleic acids, the ability to tune sizes for different compartments, complexity, and overall yield.


Aukse Gaizauskaite

Aukse Gaizauskaite*, Henrike Niederholtmeyer

poster number 15

Engineering Biomolecular Condensates in Cell-Free Systems

Several proteins, that have intrinsically disordered regions (IDRs) and high multivalency, possess the ability to self-associate into molecularly crowded liquid-like droplets, so called biomolecular condensates. In nature, living cells employ condensates as membranelles organelles for spatial organization, which allows effective separation of chemical reactions, concentration of molecules, and regulation of reaction kinetics. Cell-free transcription and translation (TX-TL) systems have emerged as alternatives to living cells in bioengineering, but they are still lacking ways to compartmentalize molecules. In our approach, microfluidic techniques and specifically designed microfluidic devices are applied to characterize and engineer protein-condensates in cell-free systems. Micrometer size environments in microfluidic chips with diffusive supply of TX-TL reagents ensure long-term continuous protein synthesis with the constant supply of fresh reaction components. Synthetic protein condensates, as programmable and dynamic molecular assemblies, can be useful for the construction of life-like systems from the bottom up, as well as for the creation of biochemically active compartments by programming the sequestration of different molecules or signalling networks.


Bert Van Herck

Bert Van Herck*, Gijsje Koenderink, Cees Dekker

poster number 16

Development of an integrated lab-on-a-chip system to establish an artificial life cycle of synthetic cells

Recent advances in synthetic biology considerably accelerated the progress toward developing synthetic cells. One crucial step in the process of generating an autonomous synthetic cell is the establishment of a cell cycle that mimics the sequence of events that takes place as the cell grows and divides. Many of these elements have already been individually recapitulated in vitro. Now the time has come to combine these modules into an integrated system of growing and dividing liposomes, mimicking a continuous life cycle of a living cell. For this purpose, we are developing an integrated lab-on-a-chip system that can be used as a fundamental tool to develop a living synthetic cell that eventually can undergo successive evolutionary cycles. More specifically, we will expand Octanol-assisted Liposome Assembly (OLA), for the production of cell-sized liposomes, and trap the liposomes in microfluidic structures for further manipulation according to our need to establish on-chip growth, division, and selection. We will present our first steps in developing the microfluidic device, and provide an outlook on how we plan to integrate these modules into one single microfluidic device.


Cathrine Abild Meyer

Abild Meyer, C.*, De Dios Andres, P., Brodszkij, E., Westensee, I. N., Lyons, J., Vaz, S. H., Städler, B

poster number 17

Crosslinked hybrid vesicles and their interactions with astrocytes cultured in paper-based chips

Astrocytes have been attracting increasing attention through the last couple of years due to their newly discovered role in neurodevelopment and function in diseases and under physiological conditions. However, the understanding of how astrocytes interact and respond remains scarce. Here, we explore how polymer lipid hybrid vesicles associate with the astrocytic cell line C8-D1A or primary cells cultured in polystyrene tissue culture or floating paper-based chips. The paper-based chips provided a 3D scaffold for the cells to mimic the physiological tissue. Consequently, they allowed the astrocytes to form a 3D network with preserved viability, calcium signaling ability, and glial fibrillary acidic protein expression. Hybrid vesicles consisting of the amphiphilic block copolymer poly(cholesteryl methacrylate)-block-poly(2-carboxyethyl acrylate) and 1,2-dioleoyl-sn-glycerol-3-phosphoethanolamine lipids, which can be crosslinked with EDC/NHS chemistry, and offer a potential useful scaffold for artificial organelles. The hybrid vesicles show no short-term toxicity to the C8-D1A cell line or the primary astrocytes. Finally, the internalization of the hybrid vesicles by the astrocytes was confirmed when cultured in the paper-based chips. These findings are a step toward creating a more stable scaffold for artificial organelles and contribute to our fundamental understanding of the interaction between astrocytes and soft-material-based nanoparticles. Abild Meyer, C., De Dios Andres, P., Brodszkij, E., Westensee, I. N., Lyons, J., Vaz, S. H., Städler, B., Astrocytes in Paper Chips and Their Interaction with Hybrid Vesicles. Adv. Biology 2023, 2200209. https://doi.org/10.1002/adbi.202200209


Mengfei Gao

Mengfei Gao*, Dishi Wang, Weihua Leng, Michaela Wilsch-Bräuninger, Dietmar Appelhans, T-Y. Dora Tang

poster number 32

Cell free expression in native protein-PNIPAAm based proteinosome

Artificial cell systems have rapidly advanced through the use of cell free expression, allowing the constructions of predicable and programmable behaviours in vitro. But the current approaches are often limited by long term stability and resources that can access the interior of a membrane bound compartment. To address these challenges, we introduce a new method to produce protein-polymer protocells (proteinosomes) directly with native proteins that are stable, porous and can support cell free transcription and translation. Furthermore, we demonstrate the feasibility of regulating and compartmentalizing cell free expression reactions through tuning the protein-polymer crosslinking. Our simplified methodology provides a versatile route to establishing robust synthetic cells that provide architectural features close to a nuclear pore as well as offering a promising platform for developing sophisticated gene circuits by combining multiple proteinosome systems in cell free expression-based reactions


Paula de Dios Andres

Paula De Dios Andres*, Joseph Lyons, Brigitte Städler

poster number 36

Towards an Artificial Cell with Transmembrane Asymmetry

Bottom-up synthetic biology is an approach that uses designed building blocks to assemble life-like units. The membrane of living cells and their organelles have commonly transmembrane asymmetry, but their synthetic counterpart rarely have this property. The bottom-up assembly of polymer-containing vesicles typically results in random distribution of the membrane constituents. Polymer-lipid hybrid vesicles are an alternative to liposomes and polymersomes that combine the self-assembly ability of the lipids and benefits of modern polymer chemistry. Only limited reports show hybrid vesicles with transmembrane heterogeneity, obtained due to the assembly process. However, ideally, transmembrane asymmetry can be regulated post-assembly. Here, we explore the use of lipid flippases, which are reconstituted in polymer-lipid hybrid vesicles, to re-arrange the anionic lipids to obtain transmembrane asymmetry. We employed hybrid vesicles composed of 1,2-dioleoyl-sn-glycero-3-phosphocholine, 1,2-dioleoyl-sn-glycero-3-phospho-L-serine, 1,2-dioctanoyl-sn-glycero-3-phospho-(1'-myo-inositol-4'-phosphate) and the amphiphilic block copolymer poly(cholesteryl methacrylate)-block-poly(2-carboxyethyl acrylate) to reconstitute P4-ATPase Drs2p-Cdc50p flippase. The ATP depletion confirmed the presence of the flippase in the hybrid vesicle membrane. An Annexin-binding assay in small and giant hybrid vesicles was used to illustrate the flipping of phospholipids. Taken together, the transmembrane transport of phospholipids is the first step towards the control over the location of the membrane constituents in artificial cells with the potential for specifically tuning the lipid orientation mimicking the behaviour of mammalian cells.


Samuel Chen

Yen-Yu Hsu*, Samuel J. Chen*, Allen P. Liu

poster number 37

Building mechanically activated synthetic cells with calcium-induced exocytosis

Exocytosis is essential for cell-cell communication in multicellular organisms and is important for numerous biological functions. In cells, exocytosis requires membrane fusion mediated by SNARE proteins, whose activities are calcium dependent. Several non-native membrane fusion mechanisms have been demonstrated; however, few are capable of sensing and responding to environmental stimuli. Here, we aim to develop synthetic cells that couple mechanosensing to calcium-triggered exocytosis in a vesicle-in-vesicle system. Artificial exocytosis is mediated by first anchoring DNA oligonucleotides on opposing membranes. Membrane fusion is controlled by shielding DNA oligos on both surfaces with membrane-bound, calpain-cleavable PEG polymers. As calpain is calcium-activated, an influx of calcium into a giant unilamellar vesicle (GUV) causes cleavage of PEG polymers, enabling membrane fusion between the encapsulated small unilamellar vesicles (SUVs) with the GUV, thus releasing the SUV contents. Mechanosensitive capabilities are introduced by cell-free reconstitution of mechanosensitive channel of large conductance (MscL) on GUV membranes . Using membrane fusion and content mixing experiments, control over DNA oligo-mediated membrane fusion on SUV-GUV, SUV-SUV, and SUV-lipid-coated bead systems were demonstrated. Furthermore, functional reconstitution of MscL on GUVs was confirmed by observing activation of MscL via osmotic pressure and the subsequent calcium influx into GUV. This new system is the first to recapitulate the coupling of calcium influx to trigger secretion in a synthetic cell model, which mimics the biochemical activation of natural SNARE-mediated membrane fusion machineries. This platform could see applications in smart drug delivery, as well as uses in exploring communications between synthetic and natural cells.


Yen-Yu Hsu

Yen-Yu Hsu*, Samuel Chen*, Allen Liu

poster number 44

Building mechanically activated synthetic cells with calcium-induced exocytosis

Exocytosis is essential for cell-cell communication in multicellular organisms and is important for numerous biological functions. In cells, exocytosis requires membrane fusion mediated by SNARE proteins, whose activities are calcium dependent. Several non-native membrane fusion mechanisms have been demonstrated; however, few are capable of sensing and responding to environmental stimuli. Here, we aim to develop synthetic cells that couple mechanosensing to calcium-triggered exocytosis in a vesicle-in-vesicle system. Artificial exocytosis is mediated by first anchoring DNA oligonucleotides on opposing membranes. Membrane fusion is controlled by shielding DNA oligos on both surfaces with membrane-bound, calpain-cleavable PEG polymers. As calpain is calcium-activated, an influx of calcium into a giant unilamellar vesicle (GUV) causes cleavage of PEG polymers, enabling membrane fusion between the encapsulated small unilamellar vesicles (SUVs) with the GUV, thus releasing the SUV contents. Mechanosensitive capabilities are introduced by cell-free reconstitution of mechanosensitive channel of large conductance (MscL) on GUV membranes. Using membrane fusion and content mixing experiments, control over DNA oligo-mediated membrane fusion on SUV-GUV, SUV-SUV, and SUV-lipid-coated bead systems were demonstrated. Furthermore, functional reconstitution of MscL on GUVs was confirmed by observing activation of MscL via osmotic pressure and the subsequent calcium influx into GUV. This new system is the first to recapitulate the coupling of calcium influx to trigger secretion in a synthetic cell model, which mimics the biochemical activation of natural SNARE-mediated membrane fusion machineries. This platform could see applications in smart drug delivery, as well as uses in exploring communications between synthetic and natural cells.


Emma Gehlbach

Emma Gehlbach*, Abbey Robinson, Katarzyna Adamala

poster number 51

Using sequential encapsulation to improve encapsulation efficiency of liposomes formed by gentle hydration

Synthetic cells are simple, man-made objects that mimic the structure and function of living cells and can be programmed to produce proteins through cell-free protein synthesis (CFPS). Liposomes are spherical lipid bilayers made of amphiphilic lipids that can be used to form synthetic cells by mimicking the structure and function of the plasma membrane. Liposomes can be created through several methods, including gentle hydration of lipids with aqueous buffer. Gentle hydration has an advantage over other some other methods of liposome formation because it makes more pure liposomes without organic contaminants in the lipid bilayer. However, compared to other methods of liposome formation, gentle hydration is less efficient at encapsulating aqueous solution within the liposome, which is a necessary step for making synthetic cells that can perform CFPS. Improving the encapsulation efficiency of the gentle hydration process would make it a more useful technique for synthetic cell research. We hypothesized that adding lipids to the buffer solution in increments (sequential encapsulation) instead of all at once (non-sequential encapsulation) would increase the encapsulation efficiency of gentle hydration. We tested this method in HEPES buffer (used in CFPS) and CHAN buffer (used for liposome storage) using cholesterol and POPC lipids, as they mimic mammalian membrane lipid composition. We used lipid concentrations ranging from 1-9 mM. We found that sequential encapsulation increases percent encapsulation of liposomes in HEPES buffer by up to 3.5-fold, but sequential encapsulation does not increase percent encapsulation of liposomes in CHAN buffer.



Session Applications: anything useful and moneymaking, like medicine, bioengineering etc. posters


Aidira D. Y. Macias Gonzalez

Aidira D. Y. Macias Gonzalez*, Jacqueline De Lora, Ashley N. Tafoya, Matthew L. Lakin, Joachim P. Spatz, and Gabriel P. Lopez

poster number 11

Developing Biosensing Coatings by Embedding SynCells in Porous Silica Films

Porous silica coatings hold promise for biomedical and sensor applications due to their biocompatibility and flexible structural and chemical attributes. We describe the fabrication of hybrid materials using biofriendly sol-gel chemistry for biomedical applications. Our long-term goal is to develop porous silica coatings that incorporate synthetic cells (SynCells) that can sense and respond to exogenous biochemical cues. Giant unilamellar vesicle (GUV)-based SynCells entrapped in porous silica films might offer this possibility. To produce porous materials, we use tetramethyl orthosilicate as a precursor, with a low molecular weight polyethylene glycol to conserve the integrity of rudimentary models for SynCells entrapped within the silica film. The sol-gel chemistry employed differs from conventional methods as it does not use methanol or other alcohols as a cosolvent and potentially harmful catalysts and byproducts. Vaporization is used to eliminate the methanol formed during silica condensation in the sol-gel process. We then entrap SynCells, comprising GUVs, to preserve their stability for long periods of time and show that this further depends on storage temperature of either 20 or 4°C. SynCells entrapped in silica matrices exhibit long-term stability especially when stored at lower temperature. We present easy to fabricate hybrid, porous silica matrices that extend the structural integrity of embedded GUV-based SynCells and probe chemical communication using an aptamer-steroid sensor platform to determine the ability to interact with small molecules that can freely diffuse through the porous silica matrix.


Ana Maria Restrepo

Ana María Restrepo*, Zhanar Abil, Christophe Danelon.

poster number 12

CADGE: Clonal amplification-enhanced gene expression for cell-free directed evolution

In cell-free gene expression, low input DNA concentration severely limits the phenotypic output, which may impair in vitro protein evolution efforts. We address this challenge by developing CADGE, a strategy that is based on clonal isothermal amplification of a linear gene-encoding dsDNA template by the minimal Φ29 replication machinery and in situ transcription-translation. We demonstrate the utility of CADGE in bulk and in clonal liposome microcompartments to boost up the phenotypic output of soluble and membrane-associated proteins, as well as to facilitate the recovery of encapsulated DNA. Moreover, we report that CADGE enables the enrichment of a DNA variant from a mock gene library either via a positive feedback loop-based selection or high-throughput screening. This new biological tool can be implemented for cell-free protein engineering and the construction of a synthetic cell.


Hannah Sleath

Hannah Sleath*, Janna Lowensohn, Bortolo Mognetti, Yuval Elani, Lorenzo Di Michele

poster number 21

Engineering Artifical Cell Chemotaxis using DNA Nanotechnology

One key goal of the field of nanomedicine and drug delivery is to achieve long-range targeting of diseased tissues, to simultaneously increase the effectiveness and decrease the toxicity of therapeutic treatments. Diseased tissues often release substances as signals to the immune system, such as in the inflammatory response. Therefore, one potential solution for targeted drug delivery is to design artificial cells that can move up a concentration gradient of the signalling substance to its source, a type of motion known as chemotaxis. The aim of this work is to engineer chemotactic artificial cells and use them to study the multivalent interactions between molecular “receptors” embedded in the cell membranes and “ligands” in the surrounding environment. We have established an experimental system in which DNA-functionalised giant unilamellar vesicles (GUVs) perform chemotactic motion by migrating along a surface density gradient of ligands adsorbed to a surface. A variety of system parameters and their effects on the motion have been explored, including varying the binding and unbinding rates of the ligand-receptor pairs, varying the surface density of DNA constructs, and varying the size of the GUVs. Experimental outcomes are compared with stochastic computer simulations, to investigate the theory governing the motion of the GUVs over ligand-coated surfaces and the effect that the ligand gradients have on this motion. The current focus of the work is to optimise an updated version of the experimental system, in which a catalytic mechanism involving toehold-mediated strand displacement is employed to boost the speed of GUV motion.


Judee Sharon

Judee A. Sharon*, Chelsea Dasrath, Aiden Fujiwara, Alessandro Synder, Mace Blank, Sam O’Brien, Lauren M. Aufdembrink, Aaron E. Engelhart, Katarzyna P. Adamala

poster number 23

Trumpet: a new operating system for biocomputing

Biological computation is becoming a viable and fast-growing alternative to traditional electronic computing. Here we present a new biocomputing technology called Trumpet: Transcriptional RNA Universal Multi-Purpose GatE PlaTform. Trumpet combines the simplicity and robustness of the simplest in vitro biocomputing methods, adding signal amplification and programmability, while avoiding common shortcomings of live cell-based biocomputing solutions. We have demonstrated the use of Trumpet to build all universal Boolean logic gates. We have also built a web-based platform for designing Trumpet gates and created a primitive processor by networking several gates as a proof-of-principle for future development. The Trumpet offers a new paradigm in biocomputing, providing an efficient and easily programmable biological logic gate operating system.


Kaitlin Eversole

Kaitlin Eversole*, Peter Davenport, Matthew R. Lakin

poster number 25

Programmable genetic networks for synthetic cell engineering

Synthetic cells are a promising approach for advanced therapeutics, bioproduction, and diagnostics. However, the genetic devices that control cellular phenotype and behavior are often hardwired and cannot be modified inside the host cell, thereby constraining design space and current applications. To address this limitation, we engineered genetic constructs capable of switching between multiple configurations in situ, thus enabling programmable control of cellular behavior. This poster will present preliminary results from the development and optimization of recombinase-programmable genetic circuits, capable of being primed for subsequent molecular computations through site-specific enzymatic recombination. To simplify the rewiring process and expand recombinase-programmable circuit design space, CRISPR transcription factors were utilized, thus facilitating straightforward scalability for future multilayer designs. Similar bottom-up engineering approaches could be used in future work to build genetic circuits capable of sensing and autonomously reprogramming the behavior of rationally engineered synthetic cells as determined by previous experiences.


Marco Oliveira

Marco Oliveira*, Rayane Lima, Lilian Hasegawa, Mariana Severo, Leila Barros, Cristiano Lacorte, Elibio Rech

poster number 27

Int-Plex@ (INTegrase PLant EXpression): a serine-integrase-based genetic switch system for inversion and excision of target genes inserted in the genome of Nicotiana benthamiana

Serine-integrases are a promising biotechnological tool to modulate plant metabolic pathways and regulate gene expression under controlled conditions. Initially present in nature as a mechanism for phage DNA integration into bacterial host genomes, these recombinases can rearrange long sequences of DNA in different ways, as excision or sequential inverting events turning on and off a target gene expression in a reversible and specific manner, according to the manipulation of their recombination sites pairs’ sequences, positions and distribution on a synthetic construct. In this work, we showed the assembly and operation of Int-Plex@, a genetic switch system based on applying four distinct prophage-derived serine-integrases as an input trigger mechanism for the inversion or excision of a target gene stably integrated into the genome of Nicotiana benthamiana. The genetic switch was formed by a mGFP gene sequence flanked by a combination of attachment sites for serine-integrases Bxb1, PhiC31, Int 13 and Int9. It can be divided into two modules: an inversion and an excision modules. The transient expression of Int9 or Int13 activates the inversion module. In this module, the activation results in the mgfp sequence being flipped to its forward orientation with the resulting expression of the mGFP reporter as a switch output. The excision module is activated by transient expression of Bxb1 or phiC31. In this case, the DNA sequence flanked by their attachment sites is irreversibly excised from the genome. The inputs can be activated sequentially, turning the target expression on and off or deleting the cassette thoroughly after modulations. Different delivery strategies for serine-integrase heterologous terms were successfully tested, although yielding varying levels of activation efficiency: leaf tissue agroinfiltration of agrobacterium transformed with viral vector Potato virus X (PVX) or with binary vector pCAMBIA2300 and biolistic system. This memory switch system can be used in future genetic circuits to engineer and modulate plant metabolic pathways of economic and environmental importance while also addressing transgene biocontainment issues, given the possibility of cassette excision.


Meline Macher

Meline Macher*, Rahul Rimal, Assa Yeroslaviz, Rinho Kim, Saradaprasan Muduli, Eva Bertosin, Smriti Singh, Ilia Platzman, Joachim P. Spatz

poster number 31

Bottom-up Assembly of Fully Synthetic Extracellular Vesicles against Inflammatory Skin Disease

Extracellular Vesicles (EVs) are naturally produced compartments for intercellular communication. Depending on their biochemical composition, they have intrinsic therapeutic potential against various physical impairments such as cardiovascular, immune and skin diseases. Especially the pro-regenerative and immunomodulatory effects of human mesenchymal stem cell (hMSC)-derived EVs are in the focus of biomedical research and technology. Despite the large biomedical potential of EVs, their conventional enrichment from cell culture medium hampers translation into clinics, as natural EV biogenesis is inherently difficult to control. This leads to high EV heterogeneity and challenges in production and mechanistic understanding. To overcome these limitations, we developed and apply bottom-up synthetic biology for a quantitative and well-controlled production of liposome-based fully synthetic EVs (synEVs). The goal of our research is to specifically mimic adipose-derived hMSC EVs and to achieve similar functionalities using synEVs in a human organotypic tissue model of inflammatory skin disease in vitro. To this end, we have isolated hMSC EVs and characterized their biophysical and biochemical properties. To cover the size range of natural EVs, we have produced two types of synEVs, based on either small (diameter below 200 nm) or large (200-1000 nm) unilamellar vesicles, both with a lipid composition and a slightly negative surface charge mimicking natural EVs. We have identified abundant surface proteins and miRNAs in the hASC EVs and have been testing the corresponding synthetic miRNAs and recombinant protein domains as active cargo of the synEVs. We have found that after topical application onto skin models, synEVs penetrate through the epidermis and even into the dermal layers. We show that a set of 5 surface proteins on synEVs is sufficient to improve histology and restore epidermal thickness and differentiation in the inflamed skin models. This is particularly interesting as it demonstrates direct effects of synEVs on skin cells without immune cell contributions. To identify the molecular mechanisms of synEVs, we are now analysing our transcriptomics data from the healthy, diseased and synEV-treated tissue models. Further optimization of synEV composition will be required to dissect the function of individual EV components. The developed synEVs have the potential to overcome challenges in the clinical translation of EVs and present a potential novel therapeutic for inflammatory skin diseases as well as a tool to better understand the mechanisms of action of natural EVs.


Sung-Won Hwang

Sung-Won Hwang, Hossein Moghimianavval, Cong Truc Huynh, Chung-Man Lim, Eben Alsberg, Nicholas A. Kotov, Allen P. Liu

poster number 40

Mechano-responsive self-degradable hydrogel using giant unilamellar vesicles

Scaffold-based tissue engineering requires the scaffold to degrade as cells proliferate and a new extracellular matrix develops. However, it is difficult to predict tissue regeneration profile and to engineer a scaffold that degrades by itself. Given that tissue growth generates pressure, we have developed a hydrogel that can degrade in response to compressive stress. To introduce mechanoresponsive behavior to a calcium-crosslinked alginate hydrogel, we employed giant unilamellar vesicles (GUVs) loaded with a gel-degrading compound, ethylene glycol tetraacetic acid (EGTA). EGTA-loaded GUVs were generated using a continuous droplet interface crossing encapsulation technique, mixed with alginate, and stiffened with calcium chloride solution. The embedded GUVs in the gel were found to be stable over 2 weeks. When the gel was subjected to compressive stress, the GUVs in the gel matrix ruptured and released EGTA, causing the hydrogel to degrade. We characterized a gel-to-solution transition by analyzing the Brownian motion of fluorescent beads embedded within the gel. Stiffness and the residual gel amount after applying pressure were also analyzed and have shown significant reduction. The results demonstrate that the GUV-embedded hydrogel can be a promising material for various biomedical applications, including tissue engineering and drug delivery, that require rapid and on-demand degradation of drug release in response to compression.


Sparky Mitra

Sparky Mitra*, Yanzhe Liu*, Andrew Nguyen*, Aniket Dhar*, Amit Klein*, Wesam Kanim*

poster number 46

Carbon Fixation in a Minimal Cell

Atmospheric carbon dioxide release has been increasing, and resulted in changes in global temperature. Uncontrolled release of greenhouse gasses brings an urgent and existential threat for organisms on Earth. Current carbon capture technologies aim to use industrial processes to convert carbon dioxide into other forms, or mitigate their release into the atmosphere through renewable energy forms. Our team proposes the use of synthetic biology to create an artificial organism able to capture carbon more efficiently, leveraging natural and synthetic carbon fixation pathways. One such pathway, the POAP cycle, has fewer reactions than any natural or synthetic pathway to date, totalling only four reactions. This pathway will then be inserted into the J. Craig Venter Institute’s Syn 3.0 minimal cell that has been evolved using TALE from 37°C to 43.4°C (via growing cells at previous bottleneck temperatures) to create a minimized organism that can perform thermodynamically-feasible carbon sequestration in isolation. The genes are inserted into Escherichia coli for assembly with two being unequivocally successful. So far, evolution and pathway assembly have been successful with other steps moving forward as well boding well for ultimately successful transformation of the minimal cell. The transformed E. coli is then purified, after which evolved minimal cells are transformed with assembled constructs. Then oxalate production is then measured by LC/GC-MS to assess initial efficiency of the synthetic pathway in vivo. For optimal efficiency, the genomes of the engineered cells will eventually be transplanted with genes assisting ferredoxin protection that will directly aid the POAP cycle. In the future, the efficiency of the organism can be measured through carbon-14 labeling within the cells to compare against other known rates. The transformed cells can also be tested in conjunction with bioreactors to assess if efficiency can be increased via environmental stimuli for further applicability to large-scale carbon sequestration.



Session Building blocks: metabolism, gene replication, membrane synthesis, ribosome assembly etc. posters


Gaku Sato

Gaku Sato, Nobuhide Doi, Kei Fujiwara

poster number 20

In vitro reconstitution of glycolysis as the core metabolism of synthetic cell

Glycolysis is a central metabolic system found in almost all living cells, producing energy and precursors for important biomolecules. However, the complexity of glycolysis has hindered its application in synthetic cell research. In this study, we developed a kinetic model to overcome the complex behavior of glycolysis and applied it to the energy regeneration system of the PURE system, an in vitro protein synthesis system using purified elements. We then attempted to synthesize important biomolecules from glucose with high yields and found that 100% conversion of glucose can be achieved by adding ATPase. Theoretically, any ATP-consuming system, including protein synthesis and genome replication, could replace ATPase. Our findings contribute to the construction of a synthetic cell with glycolysis as its core metabolic system.


Joseph Heili

Joseph Heili

poster number 22

Activation of caged functional RNAs via oxidative desulfurization of 2-thiouridine

The origins of life field, or abiogenesis, seeks to understand the processes by which life arose from non-living matter. My favorite area of research in this field is the RNA world hypothesis, purporting that RNA preceded both DNA and proteins evolutionarily. This hypothesis relies on initial abiotic generation of nucleosides and simple RNA polymers and researchers have demonstrated templated nonenzymatic polymerization of RNA rich in GC sequences using activated nucleotide derivatives. In contrast, AU-rich sequences exhibit diminished fidelity and speed. This is due in part to G-U wobble pairing, which is ca. isoenergetic with A-U base pairing. Recently, it was shown that substitution of 2-thiouridine (2sU) for U resulted in diminished wobble pairing and stronger base pairing with A, enabling faster, higher-fidelity base pairing. While this solves the problem of wobble pairing in nonenzymatic RNA copying, it brings about a second issue. In functional RNA, which are critical to the RNA world hypothesis, G-U wobble pairs are common secondary structure elements and metal ion binding motifs. Taking advantage of a recent finding that 2sU can be oxidatively desulfurized to U, we have explored the potential for rescue of functionally caged RNA polymers initially transcribed with 2sU. In experiments with a fluorescent aptamer and a ribozyme nuclease, each reliant on critical interactions with U for proper folding, we have shown that the 2sU RNA exhibiting diminished function can be at least partially rescued via the oxidative desulfurization reaction. Because the oxidative desulfurization reaction is prebiotically plausible these findings support the hypothesis that RNA polymers could be replicated non-enzymatically while also evolving function. Looking forward, I am characterizing the kinetics of the oxidative desulfurization reaction and identifying further functional RNA targets that rely critically on U for proper folding.


Kai Kean

N/A

poster number 24

N/A

N/A


Marijn van den Brink

Marijn van den Brink

poster number 29

Evolutionary optimization of synthetic genomes by image-assisted selection of liposomes

Synthetic cells provide a minimal model system to study fundamental principles of life. One approach to the construction of synthetic cells requires the stepwise integration of primary cellular functions, such as DNA replication, cell growth and division. We aim at accelerating the optimization and integration of these biological functions in gene-expressing liposomes using laboratory evolution. One essential step is the selection of liposomes displaying the desired phenotype from a large population of genetic variants. We are developing a microscopy-based selection method enabling high-resolution image acquisition for phenotypic interrogation and real-time labelling of synthetic cells via photoactivatable fluorophores, whereafter the selected vesicles are sorted by fluorescence-activated cell sorting. This technique has the potential for high-throughput selection of liposomes based on multidimensional features, including vesicle shape, protein localization and dynamic behaviours, which is indispensable for directed evolution of complex synthetic genomes. We show that liposomes can be selectively labelled using a confocal microscope and subsequently isolated from the liposome pool. By integrating artificial intelligence-assisted detection of the ‘fittest’ liposomes, we aim to develop an automated platform for rapid identification and sorting of synthetic cells.


Mireia Casanovas Montasell

Mireia Casanovas Montasell*, Alexander N. Zelikin

poster number 33

Chemical zymogens: synthetic proenzymes based on the protein cysteinome

In nature enzymes are activated and deactivated on demand, forming the basis of signal transduction while maintaining enzymatic homeostasis. These interconversions between an active enzyme to its inactive form are performed through different mechanisms, and are highly specific in space and time. However, synthesis of zymogens using tools of organic chemistry and chemical biology remains a big challenge. In this work, we address this task and engineer chemical zymogens based on the protein cysteinome. We use the protein cysteine groups to create several types of synthetic zymogens, all based on reversible conjugation of the cysteine thiol to a blocking group. Through this, we obtain inactive enzymes, which then we can reactivate on demand. In one design, we engineer enzyme (de)activation via artificial protein-protein interactions. In another, we take steps towards establishing specificity of enzyme recovery. In yet another example, we engineer a system whereby enzyme activity is controlled by steroids, in a manner that mimics nature. The specific application for the developed platform is the design of artificial signalling cascades in fully synthetic cells, and we believe it represents a big step forward in the field of synthetic, chemical zymogens.


Miyer Patiño-Ruiz

Miyer Patiño-Ruiz*, Bauke F. Gaastra, Zaid Ramdhan Anshari, Dirk J. Slotboom, Bert Poolman

poster number 34

Providing energy for the uptake of building blocks in a synthetic cell

The cell membrane constitutes a protective barrier that keeps a defined and life-friendly interior separated from an external and sometimes hostile environment. Rather than being a static physical barrier the membrane is a dynamic structure constituted mostly by lipids and proteins. Indeed, the functional activity of the cell membrane is intimately associated with the proteins embedded in this hydrophobic fluidic mosaic. Integral membrane transporters are essential to keep the cell homeostasis by mediating the uptake of nutrients and at the same time discarding waste products whose accumulation might be deleterious for life. Although a fraction of these transport processes can occur via diffusion of small molecules (passive or facilitated) it is well known that the cell invests a considerable portion of the energy budget in the uptake of nutrients and export of waste metabolic products [1], which are in most cases charged or large hydrophilic molecules. We have recognized this active transport as an essential and desirable feature for any potential synthetic cell designed to mimic the of out-of-equilibrium conditions of actual living cells [2]. We have previously demonstrated that the arginine degradation pathway provides sufficient energy in the form of ATP to activate the uptake of an osmolyte in response to an osmotic shock [3] and even to eventually energize the synthesis of lipids [4]. In this work, we present a simple strategy to provide a synthetic cell with free energy in the form of a proton electrochemical gradient that can be used to drive the translocation of building blocks across the lipid membrane. For that, we use a precursor-product system existing in fermentative bacteria as secondary source of proton motive force based on the decarboxylation of L-malate [5]. We reconstituted the L-malate decarboxylation pathway from Lactococcus lactis in lipid vesicles and demonstrated the sustained generation of a chemical proton gradient and an electrical gradient over a period of several hours. By co-reconstitution of the Escherichia coli H+ symporters GltP and LacY we showed that the free energy from the L-malate decarboxylation stored as proton motive force can drive the sustained accumulation of glutamate and lactose at concentrations compatible with those required for competitive metabolism in living cells. [1] R. Milo, R., Phillips, Cell Biology by the Numbers (Garland, Science 2016). [2] Sikkema, H. R.; Gaastra, B. F.; Pols, T.; Poolman, B. Cell Fuelling and Metabolic Energy Conservation in Synthetic Cells. ChemBioChem. 2019, pp 2581–2592. [3] Pols, T.; Sikkema, H. R.; Gaastra, B. F.; Frallicciardi, J.; Śmigiel, W. M.; Singh, S.; Poolman, B. A Synthetic Metabolic Network for Physicochemical Homeostasis. Nat Commun 2019, 10 (1) [4] Bailoni, E.; Poolman, B. ATP Recycling Fuels Sustainable Glycerol 3-Phosphate Formation in Synthetic Cells Fed by Dynamic Dialysis. ACS Synth Biol 2022, acssynbio.2c00075. [5] Poolman, B., Molenaar, D., Smid, E., Ubbink, T., Abee, T., Renault, P. P., Konings, W. N., Malolactic fermentation: electrogenic malate uptake and malate/lactate antiport generate metabolic energy. J. Bacteriol. 1991. 19, 6030-6037.


Shanny Ackerman

Shanny Ackerman*, Patricia Mora-Riamundo, Avi schroeder

poster number 38

Energy pathways in synthetic cells

The success of synthetic cells (SCs) in many translational applications requires their prolonged and dynamic activity, which heavily relies on energy availability. Without an external energy supply, the maximum protein production capacity inside SCs was measured in a few hours. Different techniques have shown successful production of ATP inside SCs. However, most methods were based on structural changes and the addition of complex enzymatic reactions to the SCs. Nevertheless, finding a simple solution to the energy supply challenge is essential for advancing applications with SCs, especially medical ones. This study aims to introduce a human-made and uniquely structured energy-supplying organelle into the SCs and prolong their protein production for several days.


Tanner Hoog

Tanner Hoog*, Matt Pawlak, Nathan Gaut, Katarzyna Adamala, and Aaron Engelhart

poster number 42

Utility of the Selective Denaturant Perchlorate in Origins of Life and Synthetic Cell Research

The Hofmeister ions are categorized by their ability to increase biomolecular secondary structure stability (kosmotropes) or to decrease biomolecular secondary structure stability (chaotropes). Here we show that the G quadruplex, a noncanonical nucleic acid secondary structure, retains stability in solutions of anionic chaotropes relative to several other nucleic acid secondary structures, including a duplex and i-Motif. This relative insensitivity of G quadruplexes compared to duplexes enables a switchable nucleic acid electron transfer device, which can be switched using heat or dilution. Additionally, we show that ribozymes retain activity in perchlorate brines. This is of interest from an origins of life perspective because transient pools of liquid water can be stabilized on Mars due to perchlorate’s innate hygroscopicity.


Viktoriia Belousova

Viktoriia Belousova, Kareem Al Nahas, Petra Schwille

poster number 43

Mutation of alpha-hemolysin creates a cation-controlled transmembrane pore for synthetic cells

Transport of molecules across the membrane is crucial for cell communication, organization, and regulation. In nature, there are many sorts of membrane transporters, ion channels and pore proteins that are regulated and coordinated by complex signaling cascades. In synthetic cells based on biomimetic membrane compartments, implementing and regulating transport across the membrane remains a challenge. Ion channels and pores were reconstituted in synthetic cell mimics like GUVs before, but are often suboptimal modules to be regulated under the conditions of minimal systems. Generally, pore proteins that can be regulated by ions or small molecules constitute a more promising tool for this goal. In this work, we use a variant of Staphylococcus aureus pore protein alpha-hemolysin with a 5 amino acid mutation that makes it susceptible to Zn2+ ions. We demonstrate that this alpha-hemolysin mutant can be expressed in GUVs using a cell-free protein expression system and that it allows small molecules to diffuse across the GUV membrane. We also show that this passive transport can be controlled by addition of Zn2+ ions that bind to the pore protein and close it, blocking transmembrane diffusion. This variant of alpha-hemolysin thus appears to be a promising tool for future applications in synthetic cells that require the control of signaling and communication.


Ryota Miyachi

*Ryota Miyachi, Yoshihiro Shimizu, Norikazu Ichihashi

poster number 48

Transfer RNA synthesis-coupled translation and DNA replication in a reconstituted transcription/translation system

Cell-free or in vitro synthetic biology aims to reconstitute the biological functions from defined molecules, such as DNA, RNA, and proteins, to reveal the design principles of living systems and also provide new biotechnology (Wang et al., 2021). Despite various cellular functions have been reconstructed, however, the construction of an artificial self-reproductive system, which requires regeneration of the gene expression system from the gene expression system itself, remains a significant challenge. Especially, In vitro synthesis of Transfer RNAs (tRNAs), central factors in translation, and their coupled translation are important challenges in the construction of a self-regenerative molecular system (Hibi et al., 2020). Here, we first examined luciferase translation coupled with tRNA expression in a one-pot reaction. We tested different strategies for expression schemes with 5'-G and non-5'-G tRNAs and demonstrated expression of the 21 tRNAs that are minimally required for translation. Moreover, we showed DNA replication through expression of a tRNA encoded by DNA, mimicking information processing within the cell. Our findings highlight the feasibility of an in vitro self-reproductive system in which tRNAs can be synthesized from replicating DNA.


Evan Kalb

Mace Blank, Joshua Davisson, Evan Kalb, Angelin Prasad, Aaron Engelhart, Kate Adamala

poster number 52

Eutectic phase flexizyme reactions improve acylation and cell-free translation yield

Flexizymes are ubiquitous for acylating an enormous chemical diversity of compounds onto tRNA, enabling the translation of radically different peptide backbones, bonds, and side chains. However, flexizyme reactions suffer from several drawbacks such as single turnover kinetics, high magnesium carryover in in vitro translation reactions, and rapid hydrolysis. Utilizing a water ice-eutectic phase, we have developed novel flexizyme reaction conditions which demonstrate increased aminoacylation yields, low magnesium concentrations, and slow hydrolysis of the aminoacyl bond. Eutectic flexizyme reactions attain similar or higher yields than literature and maintain this high yield for 48+ hours, much longer than a standard flexizyme reaction. Slow hydrolysis means flexible flexizyme reaction timing and increased utility and flexibility of aminoacylation reactions. Furthermore, flexizyme reactions require an excess of MgCl2 (up to 600 mM). This concentration of Mg2+ has been shown to carryover into cell free protein synthesis reactions, inhibiting protein synthesis. Eutectic flexizyme reactions use 30 times lower MgCl2, reducing carryover and increasing peptide/protein production for in vitro translation reactions. Flexizyme reactions have been shown to be practically single turnover. Surprisingly, eutectic flexizyme reactions have increased turnover, further increasing aminoacylation yield. Overall, we present a new methodology for performing in vitro aminoacylation reactions, with increased yields and more flexible timing.


Scott Hansen

Grace Waddell, Benjamin Duewell, Naomi Wilson, Emma Drew, Sophia Doerr, Sarah Peabody, Andres Olavarrieta, Katie Faris, Reed Talus, & Scott D. Hansen*

poster number 53

Mechanisms controlling spatial patterning of PIP lipids in cell signaling

Cells interpret chemical cues in their environment and make decisions that control their fate. For example, human neutrophils respond to a variety of pathogenic signals by sensing, polarizing, and targeting the foreign pathogens. Critical for these cellular processes is the relay of signals between cell surface receptors, small GTPases, and phosphatidylinositol phosphate (PIP) lipids. Collectively, these molecules orchestrate numerous biochemical reactions at the plasma membrane that give rise to emergent properties, such as symmetry breaking, polarization, and spatial patterning during cell signaling. My lab uses a combination of biochemistry and quantitative cell biology to define the mechanisms that control these properties. Using supported membrane technology and single molecule imaging techniques, we have characterized several classes of lipid kinases and phosphatases that regulate the rapid amplification and down-regulation of PIP lipids in various spatial patterning programs. Long-term, we want to understand the design principles that allow PIP lipids to organize signaling reactions on cellular membranes. Given the critical role that PIP lipids play in cellular organization and signal transduction, our research will also help decipher mechanisms of misregulation that are associated with numerous human diseases.



Session Beyond the bench: education, outreach, ethics, biosafety posters


Orion Venero

Orion M Venero, Katarzyna P Adamala

poster number 35

Synthetic Systematics: The Question of Naming Synthetic Life

To fully apply the findings of synthetic cell research to a broader range of fields and research interests, it is paramount that there is a common language with which to discuss synthetic biotic constructs. In this respect, such systematization must be compatible with existing classification methodologies for terrestrial life: acting as an aesthetic or conceptual overlay atop existing systems of nomenclature. The development of a universal system of nomenclature for biology is deeply linked with the formulation of a universal theory of life because cogent systematization is contingent on semantically understanding the same logical and aesthetic problems faced when trying to proffer a universal theory of life. Evincing a universal theory of life is outside the scope of this effort, but the development of an aesthetic model which relates the classifiers ‘biotic’ and ‘living’ will allow for the formulation of universal systematics and perhaps aid future efforts to advance a universal theory of life. Accordingly, the biotic interval model, that we present here, demonstrates the precedent for, and feasibility of, the phylogenetic classification of synthetic life alongside classical terrestrial life.


William L Gannon (Bill)

William L. Gannon*, Department of Biology, Philosophy, and Graduate Studies, University of New Mexico, Albuquerque, NM 87131 (wgannon@unm.edu ) and, Gabriel P. Lopez, Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131 (gplopez@unm.edu)

poster number 49

Increasing Accessibility of Emerging Technologies to URM Students: Lessons from a Graduate Student Seminar in Synthetic Cell Biology

Abstract -- The purpose of this research is to describe characteristics and motivations of students who have entered the university as undergraduate and graduate students to pursue research in synthetic cell biology, a field focused on engineering an artificial cell, synthetic cell or minimal cell as an engineered particle that mimics one or many functions of a biological cell. We provide details of a seminar course that was specifically created for students interested in synthetic cell biology and engineering with emphasis on both technological content and ethical considerations as this field emerges. Student participation included discussion, research, and presentation of student-identified projects. Given the special nature of the University of New Mexico as a minority-majority, Hispanic Serving Institution, the project is aimed to characterize the stories of students in the field about how they decided to enter a graduate program, what sort of barriers or hardships that they have encountered, where they hope their careers will develop, and what they feel about the future of synthetic cell research and applications. We describe how our synthetic cell biology course was conducted and provide recommendations for better teaching practices for future course offerings at other institutions derived from student feedback. We consider courses such as these to be effective ways to recruit and support current and potential students from underrepresented groups into the field.


Joshua Davisson

Joshua Davisson, Evan Kalb, Mace Blank, Kate Adamala

poster number 56

Complete genetic code reprogramming subverts current DNA purchasing biosafety measures

Highly conserved across all known life forms, the genetic code carries information transported by mRNA and translated into protein. Genetic code reprogramming alters this biological Rosetta Stone, liberating codons for the translation of noncanonical amino acids or to reassign a canonical amino acid into a new codon, weakening the link between nucleic acid and protein sequence. Recently, aminoacyl tRNA synthetase (aaRS)-free translation of proteins was demonstrated by supplementing aminoacylated tRNA into a protein synthesis using recombinant elements (PURE) reaction (Chen et al., 2021). This was accomplished using flexizymes, small synthetic ribozymes that charge tRNAs in vitro. However, flexizyme aminoacylation is not specific for any tRNA and can attach almost any amino acid to any tRNA. By aminoacylating all 20 amino acids to their noncognate tRNAs, complete genetic code reprogramming is possible, translating proteins using a novel genetic code. This genetically reprogrammed PURE reaction would enable the subversion of DNA purchasing biosecurity measures which heavily rely on sequence homology (Elworth et al., 2020). This is particularly relevant after a paper synthesized infectious poliovirus particles in vitro from assembled DNA oligos using a cell free protein synthesis reaction (Cello et al., 2002). The increasingly weak link between nucleotide and protein sequence highlights the need for further genetic biosafety considerations. This project demonstrates that genetic biosecurity and biosafety must adapt with novel genetic engineering tools like genetic code reprogramming.



Session Cell-free translation systems: the cytoplasm of synthetic cells posters


Matas Deveikis

Matas Deveikis

poster number 30

Exploring limiting factors in the PURE cell-free system

Cell-free protein synthesis (CFPS) systems have emerged as a valuable tool for protein production outside a living chassis. Protein synthesis using recombinant elements (PURE) cell-free system has been especially promising for use in engineering synthetic cells due to the ability to finely model the system and adjust the levels of its' components. However, comparatively low yields have made the PURE system an unattractive candidate in practice. In this study, we aim to explore the limiting factors stopping PURE cell-free reactions and propose methods to increase the efficacy of the system.


Shota Nishikawa

Shota Nishikawa*, Po-Hsiang Wang, Shawn McGlynn, and Kosuke Fujishima

poster number 39

De novo synthesis of iron-sulfur proteins using recombinant SUF system in a cell-free system

Iron-sulfur clusters are essential cofactors mediating single-electron transfer in respiratory and metabolic networks. However, heterologous iron-sulfur protein expression is challenging due to (i) the requirements for iron-sulfur cluster assembly, (ii) the O2 lability of iron-sulfur clusters. Here, we established a facile and efficient protocol to express mature [4Fe-4S] proteins in the reconstituted cell-free translation system (i.e., the PURE system) under aerobic conditions. The incorporation of recombinant SUF helper proteins to the PURE system enabled streamlined mRNA translation and [4Fe-4S] cluster assembly under anaerobic conditions. To reconcile the O2 lability of [4Fe-4S] clusters, we further added an O2-scavenging three-enzyme cascade to the PURE system. In this cascade, formate dehydrogenase, flavin reductase and catalase are used to reduce O2 to water. This cascade also supplies FADH2 to the SUF helper proteins as well as removing O2 from the reactions, allowing a one-pot synthesis of mature [4Fe-4S] proteins under aerobic conditions with a high maturation rate (>90%). Our new approach will expand the repertoire of cell-free biocatalysis, paving the way for future reconstruction of redox-active synthetic cells.


Surendra

Surendra Yadav

poster number 41

Building Synthetic Metabolism to Efficiently Drive the PURE Cell-free system

Cell-free protein synthesis (CFPS) platforms have recently emerged as powerful tools to produce recombinant proteins rapidly and inexpensively, by harnessing biological gene expression inside in vitro biochemical reactions. The Protein Synthesis Using Recombinant Elements (PURE) system combines purified molecular components required for transcription and translation with energetic substrates and cofactors to carry out CFPS. This completely defined system can be rapidly modulated, offers straightforward product purification, and is well-suited to the construction of minimal cells. However, the system is notably limited by low reaction yields and lifetimes (~2h) compared to conventional lysate-based systems. One way to improve reaction performance is to reengineer the biochemical energy regeneration system. The PURE system uses a basic ATP regeneration system based on creatine phosphate, which leads to accumulation of inorganic phosphate, a by-product inhibitory to CFPS at high concentrations. The aim of this project is to explore different synthetic systems to regenerate ATP, starting with a three-enzyme system based on pyruvate oxidase, catalase, and acetate kinase, which was successfully used to control phosphate accumulation in lysates. Our preliminary results show that this system is thermodynamically as well as kinetically feasible in the PURE reaction conditions. The three enzymes have been purified and are being tested in the PURE system for their energy regeneration role. The successful implementation of these new pathways in the PURE system will help improve performance in terms of reaction lifetime and yield.


Kirill Alexandrov

Shayli Varasteh Moradi, Sergey Mureev, Wayne A Johnston, Yue Wu, Patricia Walden, Zhenling Cui and Kirill Alexandrov

poster number 45

Development and applications of a eukaryotic cell-free expression system based on Leishmania tarentolae

Proteins are workhorses of the biotech industry and Life sciences. Yet our ability to create protein-based systems with desired features is woefully underdeveloped. In an effort to create effective tools for rapid protein production and engineering we have exploited the unique biology of Kinetoplastida to develop Leishmania tarentolae-based in vitro expression systems . We present the motivations, opportunities and hurdles that we faced creating and disseminating different versions of the system. The system has been tested in a range of cell-biological, biotechnological and protein engineering projects. We will discuss the lessons learned and show case several examples including the use of Leishmania cell free system to map protein:protein interaction network of SARS-CoV-2 virus. We will offer my perspective on the place of the developed technology in the Life Sciences toolbox.


Vadim Bogatyr

Vadim Bogatyr¹*, Andreas Biebricher¹, Maike Hansen², Roel Maas², Wilhelm Huck², Gijs Wuite¹ 1 – Vrije Universiteit Amsterdam, Amsterdam, the Netherlands 2 – Radboud University, Nijmegen, the Netherlands

poster number 47

Counting mRNAs: towards single-molecule in vitro transcription (smIVT) assay

Transcription is a vital cellular process in which an RNA polymerase produces messenger RNAs (mRNA) from a DNA template. Therefore, on its trajectory toward a viable artificial cell, synthetic biology aims to reproduce transcription reliably and understand how it works in a confined environment. To this end, a number of commercially-available or lab-made transcription systems have been established for in vitro applications. Moreover, using fluorescent probes, researchers could follow live the changes in RNA concentration over time inside giant unilamellar vesicles (GUVs). However, most of these measurements have concerned DNA concentrations far exceeding those in natural cells, where there is usually just a single DNA copy. Here we show our progress towards a single-molecule transcription assay that goes beyond these limitations and allows us to track single DNA and RNA in solution and inside GUVs. We have achieved this with two approaches, both reliant on fluorescent probes that become more fluorescently active after an interaction with the transcript mRNA. In the first one, we use molecular beacons (MB), a 26-base long DNA hairpin modified with ATTO 647N dye and an Iowa Black RQ quencher. Closed and dim in the solution, they open and light up upon binding to the complementary sequence on the mRNA. The template DNA here had 32 MB-complementary repeats to ensure a signal-to-noise ratio sufficient for detecting single mRNAs. In the second approach, we have utilized Spinach aptamer, a specific mRNA sequence that, upon folding, can stabilize a DFHBI dye molecule and enhance its signal. Similarly, to improve detection, 32 repeats of the Spinach sequence were encoded on the DNA construct. We demonstrate these two single-molecule approaches for tracking transcription in real-time at picomolar DNA concentrations, discuss their performance and compare different transcription systems.


Elisabeth Edgerton

Elisabeth Edgerton*, Maddie Westenberg, Wakana Sato, Madilyn Stahl, Ahmed Shahkhan, Akshay Peddi, Dadir Awad, Aaron Engelhart, Katarzyna Adamala

poster number 54

Cellulator: Developing a Validated Computational Model of Cell-free Protein Synthesis

Cell-free protein synthesis is an efficient method for expressing and producing proteins, but predicting yields can be difficult. Computational models can help to predict the impact of varying parameters, but few models exist that have been experimentally validated and include all the necessary biological components. We developed a computational model of cell-free protein synthesis that includes transcription, translation, energy regeneration, and aminoacylation. Experimental kinetic data collected from the literature was used to set parameters for the model, and we ran a series of simulations to predict protein synthesis for the protein HiBiT. To validate the model, we ran experimental trials in which the concentrations of DNA, tRNA, amino acids, and rNTPs were individually varied, and the resulting protein yields were measured. Experimental results demonstrated that the model accurately predicted the changes in protein output when the concentrations of DNA and tRNA were varied. However, when the concentrations of amino acids and rNTPs were varied, the experimental results differed from the model predictions, possibly due to a lack of consideration of magnesium ions in the model. Additionally, the numerical protein output generated by the model was not consistent with the actual protein yields measured in the experiments, which indicates that the model could be improved upon by accounting for additional parameters or further refining the kinetic data. Overall, our computational model provides a valuable tool for predicting the impact of varying system parameters on protein synthesis and could potentially be used to optimize the PURE cell-free system for specific protein production. By comparing the simulation results with experimental data, we can continue to build upon the model and improve its accuracy for further testing.


Christopher Deich

Christopher Deich, Nathaniel Gaut, Brock Cash, Tanner Hoog, Aaron Engelhart, Katarzyna Adamala

poster number 55

Synthetic minimal cell with 90kb genome undergoing natural selection

Assembling a live cell from non-living components is one of the biggest challenges of experimental science. Here we demonstrate a complete cell cycle of a synthetic cell undergoing natural selection: DNA replication, gene expression, genetically encoded growth, and resource acquisition via feeding. To enable growth and feeding, we engineered a genetically encoded mechanism for liposome fusion. The competition between two genomes results in natural selection, where one population feeds and grows faster, producing more offspring. The synthetic cells presented here have fully defined chemical composition, demonstrating minimal chemical recipe for life-like system. Overall, we present a cell cycle coupled with natural selection, that creates a chassis for the evolution of new lineages of life.



Session Minimal cell on live chassis: Mycoplasma and others posters


Daniela Bittencourt

Daniela Matias de C. Bittencourt*, Marco Antônio de Oliveira, Raquel Sampaio de Oliveira, Mariana Mathias C. Araujo, Elibio Rech, John I. Glass

poster number 18

Development of chromosomal DNA-free Mycoplasmas as recipients of functional genetic constructs

The study and development of minimal genomes serve as a basis for better understanding the structural and functional organization of the genome and, consequently, how to manipulate it to perform specific functions in response to external stimuli. Therefore, the development of synthetic cells and effective tools to build them will contribute to more precise genetic regulation and the generation of increasingly innovative processes and products. In this study we report the development of chromosome-free cells based on the minimal cell JCVI-Syn3.0B, as a prototype for the design of synthetic cells capable of acting as biosensors and performing specific functions in response to external stimuli. To this end, we engineered a genetic circuit using serine integrases (INT9) to activate the expression of the endonuclease I-Ceul, which together with endogenous endonucleases, completely degrades the host cell genome. The endonuclease I-Ceul recognizes a specific 26 base pairs sequence (5'-TAACTATAACGGTCCTAAGGTAGCGA-3'), present in most bacterial genomes encoded within the 23S rRNA rrl gene. To analyze the functionality of the new cell (SimCell_JCVI-syn3B), a genetic construct with the complete metabolic pathway of arginine deaminase was also used. Due to the lack of a chromosome, SimCell_JCVI-syn3B is unable to replicate, which lessens the biosafety concerns associated with genetically modified organisms. This approach will not only enable the development of innovative and safe biological inputs for application in the different productive sectors of agriculture, industry and medicine, but it also comes as an alternative method to prepare recipients for synthetic genomes.


Feilun Wu

Feilun Wu, Nacyra Assad-Garcia, Eugenia Romantseva, Elizabeth Strychalski, John Glass

poster number 19

Expanding toolkit for whole genome transplantation

Whole genome transplantation (WGT) is a process of installing an isolated bacterial genome into a suitable recipient cell, resulting in a transplant with the phenotype and genotype of the donor genome. Building cells programmed with synthetic or other radically engineered genomes will be a slow and expensive process until a method like genome transplantation can be used for a wide range of bacteria species. However, currently, whole genome transplantation is only successful in mycoplasma. In order to expand whole genome transplantation beyond mycoplasma, fundamental understanding of the mechanism of whole genome transplantation needs to be acquired, and new techniques and technologies such as automation and microfluidics devices need to be developed. Here we outline our plan for the expansion of whole genome transplantation toolkit.


Ronald Rodriguez

Ronald Rodriguez, John I Glass

poster number 50

Genetic and Physiological Characterization of a Genome Minimized, Synthetic Bacterial Cell

The construction of living cells with minimized genomes required development of methodologies that have been extremely valuable to synthetic biologists performing grand scale genome engineering. JCVI-Syn 3.0 (Syn 3.0) is a synthetic, minimal bacterial cell derived from Mycoplasma mycoides containing only 473 genes and is currently being utilized as a model system to study the basic principles of life. Syn 3.0 can replicate its genome and undergo cell division, despite the absence of genes thought to be necessary for normal cell division. Syn 3.0 cell division generates irregularly shaped cells and filamentous structures through an unknown mechanism. Normal cell division can be restored by the addition of seven non-essential genes, including cell division genes ftsZ, sepF, and five genes with unknown functions. To develop a better understanding of cell division in Syn 3.0, we will examine the cellular localization of these non-essential gene products with high-resolution light microscopy. Bacterial histone-like protein (HupA) is a high-copy essential protein in natural bacteria. HupA copy number per cell decreased from approximately 6000 to 28 in our minimal cell, presumably due to deletion of a non-essential gene located directly upstream of hupA. We hypothesize this is why chromosome conformation capture studies of our minimal cell displays a lack of long-range chromosome interactions. Perhaps by dramatically reducing the copy number of HupA per cell, we have recapitulated a physiological state of primordial cells before the evolution of chromatin. We will restore the non-essential gene upstream of hupA and determine if our long-range chromosomal interactions date is similar to what occurs in other bacteria. Our planned minimal cell experiments should expand our understanding of minimal cell physiology and potentially provide insight into what primordial cells may have looked like before the emergence of modern cell division mechanisms and chromatin in bacteria.



Session Cytoskeleton: protein, DNA, other structures posters


LAURA CASAS FERRER

Casas Ferrer Laura, Xiangting Lei, Honts Jerry, Bhamla Saad

poster number 26

Mechanical behavior of optically controlled motor-free cytoskeletal networks under biomimetic confinement

In the present work we study the behavior of a motor-free cytoskeletal network under physical confinement. Our system is made of the tetrahymena calcium-binding protein 2 (tcb2), which has the ability to assemble into network-like structures and to contract upon increase of free calcium in solution. The network is enclosed inside a cell-sized liposome, which mimics the structure and composition of a cellular membrane. The contraction of these networks is possible thanks to a photolabile compound that chelates calcium and releases it upon irradiation with UV light, which is also enclosed inside the lipid bilayer. We study the response of the protein network upon controlled calcium release within the membrane boundaries, the membrane deformations, and the motion that arises from it. The study of motor-free cytoskeletons is important in the context of bioengineering, since it opens new venues for the design of more energetically efficient systems and devices. The ability to optically control calcium signaling pathways is also a rising field in biomedicine, which has already offered solutions for diagnosis and treatment of various diseases, including cancer, and several muscular and neurological disorders. The convergence of these two disciplines in the presented work will contribute to building a solid ground for the development of the next generation of optically-controlled bioactuators and biosensing devices.


María Reverte López

María Reverte-López*, Marion Jasnin, Petra Schwille

poster number 28

Positioning of actomyosin rings and vesicle blebbing mediated by the MinDE system

One of the challenges of bottom-up synthetic biology is the engineering of a minimal set of modules for self-division of synthetic cells. To produce the contractile force required for the excision of cell-like compartments such as lipid vesicles, reconstituted cytokinetic rings made of actin are considered the most promising constituent of a potential synthetic division machinery. However, control over the assembly and localization of actomyosin rings inside giant unilamellar vesicles (GUVs) is still required in order to reach effective mid-vesicle constriction. Here, we show the combined in vitro reconstitution within GUVs of actomyosin rings and a synthetic positioning module: the bacterial MinDE protein system, a versatile tool that allows the active transport of any diffusible cargo molecule on membranes. Incorporating Min oscillations to actin-containing GUVs induces the stacking and localization of actin rings at the equatorial plane of lipid vesicles. Remarkably, the synergistic effect of Min oscillations with the contractile nature of actomyosin bundles induces mid-vesicle membrane deformations and blebbing, which lead to the breaking of spherical GUV symmetry and dramatic outward membrane expansion. Our system thus demonstrates that it is possible to integrate synthetic modules dissimilar in nature, and showcases how emerging effects arising from their joint reconstitution could contribute to the development of a synthetic division toolset.



Tuesday, May 23 sessions and keynotes

Tuesday, May 23, breakout sessions


Containers #3

Tuesday, May 23, Johnson room, 1:00PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Tuesday, May 23rd
Johnson room
presenter: James Hindley
all authors: Ignacio Gispert, Yuval Elani, Oscar Ces
Modular construction of synthetic signaling pathways
Synthetic cells are nano and microsystems that aim to replicate the structures, functions and behaviours of living cells. One such behaviour, communication, is essential to the functioning of living systems. Signaling enables them to sense their surroundings, and in response adapt their state. It is therefore critical for synthetic cells to be able to sense and respond to specific stimuli in their environment if they are to successfully mimic their biological counterparts. In this talk I will discuss how to construct signaling pathways in synthetic cells, employing biochemical parts and responsive lipid membranes in a modular approach to implement (i) signal transduction, (ii) user-controlled, stimuli-responsive cell-cell communication and (iii) feedback loop design. Integrating each of these systems within synthetic cells has the potential to drive their use as biological models, as well as generate new technologies suitable for commercial or clinical translation.

1:00PM - 2:30PM
Tuesday, May 23rd
Johnson room
presenter: Rizal F. Hariadi
all authors: Ranjan Sasmal, Nirbhik Acharya, Gde Bimananda Mahardika Wisna, Youssef Hassan, Carter Swanson, Hao Yan, Rizal F. Hariadi*
Building GPCR-inspired molecular devices with amphiphilic DNA for signal transduction across membranes.
Plasma membranes are fundamental to all living organisms, separating the intracellular and extracellular environments. Membrane-spanning proteins enable molecules and information to be transported across otherwise impermeable membranes. Design principles for these proteins are often elucidated using scientific approaches across scales, from single-molecule biophysics to cell and developmental biology. In this talk, I will describe my group's work using synthetic approaches to understand the fundamental limits of membrane-spanning proteins. Rather than studying the intricate workings of these proteins in vitro and in vivo, we apply their design principles to engineer functional membrane-spanning devices. These membrane-spanning protein analogs can transduce signals from the interior of synthetic and biological vesicles to produce specific responses on the outside. Driven by hydrophobic interactions, these amphiphilic molecules target and insert themselves into lipid membranes while translocating some parts of the molecules across the membrane. Furthermore, the sensing domain of the membrane-spanning nanodevices is designed to bind complementary oligonucleotides on the inner side of a lipid bilayer and generate a fluorescence signal. Confocal microscopy and flow cytometry assays reveal that the sensor binding does not compromise the membrane's structural integrity. We evaluated the potential of our sensor by assessing its dynamic range, specificity, and kinetics, as well as its integrity and orientation across mammalian plasma membranes with minimal cytotoxicity. Moreover, we observed a novel phenomenon of sequence-specific DNA-gated passage of small molecules inside synthetic vesicles, termed DNA-gated nanopores. Finally, I will conclude with a discussion on the transformative potential and future directions of non-destructive sensing of cytosolic nucleic acid biomarkers, including live-cell isolation based on their transcriptomic profile for CRISPR screening and cell therapy.

1:00PM - 2:30PM
Tuesday, May 23rd
Johnson room
presenter: Jorik Waeterschoot
all authors: Jorik Waeterschoot*, Jeroen Lammertyn, Erin Koos, Xavier Casadevall i Solvas
Formation of giant unilamellar vesicles assisted by fluorinated silica nanoparticles
One of the main impediments to producing artificial cells is the recreation of the cellular membrane. Among the existing model systems that mimic these membranes, giant unilamellar vesicles (GUVs) are especially interesting due to their resemblance in size and shape to eukaryotic cells. Many techniques have been developed to produce GUVS, such as electroformation, thin film hydration and microfluidics, all having inherent advantages and disadvantages. Here, we show that fluorinated silica nanoparticles (FNPs) used to form Pickering emulsions in a fluorinated oil can destabilise lipid nanosystems to template the formation of GUVs inside microdroplets. This technique enables GUV release across a broader spectrum of buffer condition than previously described, also preventing the leakage of the droplet encapsulated components into the oil phase. A further added benefit is the simple use of centrifugation for the release of GUVs into an aqueous phase, thus bypassing the need to use emulsion-destabilising chemicals. By using fluorescently-labelled FNPs and TEM we were able to confirm that our FNPs are removed together with the fluorinated oil, producing GUVs that are free of any contaminants. Further experiments to assess the lateral diffusion of the lipids and the unilamellarity of our GUVs demonstrate that they are comparable to GUVs produced via electroformation. Hence this new methodology constitutes a valuable addition to the toolkit of GUV production in the growing field of synthetic biology and open a new route toward the production of more biologically relevant artificial cells containing active biomolecules.

1:00PM - 2:30PM
Tuesday, May 23rd
Johnson room
presenter: Matthew Allen
all authors: Matthew E. Allen*, James W. Hindley, Nina O’Toole, Elina Kamilova, Hannah S. Cooke, Oscar Ces, Yuval Elani
Hydrogels as functional components in artificial cell systems
Artificial cells are biomimetic structures formed from molecular building blocks that replicate biological processes, behaviours, and architectures. Of these building blocks hydrogels have emerged as ideal, yet underutilised candidates to provide a gel-like chassis in which to incorporate both biological and non-biological componentry which enables the replication of cellular functionality. As a consequence, we have developed a microfluidic strategy to assemble hydrogel-based artificial cells that can interface with a variety of living and non-living components. These newly developed hybrid microstructures have enabled mimicry of existing biological structures and the gaining of new capabilities that can be applied in fields as diverse as therapeutic delivery, biosensing, cell therapy and bioremediation.

1:00PM - 2:30PM
Tuesday, May 23rd
Johnson room
presenter: Orion Venero
all authors: Orion M Venero, Katarzyna P Adamala
Modeling Membrane Growth with Engineered Organelles
Recapitulating binary division of a cell membrane is an eminent goal in synthetic cell research. However, this classical cytokinesis, which results in two equal daughter-cells, is a non-trivial process; itself predicated on both membrane growth, and the orchestration of membrane division machineries. While many efforts have proffered tractable methods for recapitulating either membrane growth or membrane division, few have unified both in the same synthetic cell system. Moreover, those methods which have been investigated, are constrained by the non-regenerative consumption of starting reagents, in bottom-up synthetic cell systems. Here, we describe a hybrid system that is comprised of a top-down artificial endosymbiont encapsulated within a TX;TL competent bottom-up synthetic cell. Molecular programming of the artificial endosymbiont up-regulates fatty acid (FA) production, driving FA secretion into the synthetic cytoplasm, and eliciting FA-mediated growth of the synthetic cell membrane. The endosymbiont is an S. elongatus cyanobacteria; hence, modulation of photoperiod can be used to tailor endosymbiont metabolism and FA production in confluence with TX;TL affected division machineries. This represents a preliminary step towards developing a robust photo-inducible membrane growth system that could serve as a test platform for various synthetic cytokinesis strategies.



Applications #3

Tuesday, May 23, Ski-U-Mah room, 1:00PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Manish Kushwaha
all authors: Amir Pandi, Mathilde Koch, Paul Soudier, Angelo Cardoso Batista, Antoine Levrier, Peter L. Voyvodic, Ana Zuniga, Thomas Duigou, Kenza Bazi-Kabbaj, Julie A. Vendrell, Tatjana Achmedov, Tristan Reif-Trauttmansdorff, Angelique DeVisch, Martin Cohen-Gonsaud, Jerome Solassol, Chase L. Beisel, Jerome Bonnet, Jean-Loup Faulon, Manish Kushwaha*
Information processing circuits in cell-free systems: beyond gene expression regulation
Cell-free expression systems have emerged as promising tools for implementing information-processing circuits. Most of these circuits employ biosensors directly linked to gene expression regulation, using digital logic encoded on plasmid DNA. Here, we present several new approaches for building biological circuits that process information via metabolic transducers: combining analogue and digital logic in the metabolic perceptron, routing metabolic sensing through a common hub molecule, and screening transducer enzymes using linear DNA templates. The examples introduced here lay the groundwork for more advanced metabolic circuits for rapid and scalable multiplex sensing.

1:00PM - 2:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Brian Belardi
all authors: Brian Belardi*, Unyime Effiong, Ahmed Sihorwala
Controlling Molecular Assembly State En Route to Synthetic Tissue
Multicellular tissue – the hallmark of all animals – exhibits an exquisite array of morphological structures and material properties. The three-dimensional structures and physical properties of living tissue are, however, far from static as they can adapt and evolve during developmental programs and when exposed to biological insults. Recent advances in synthetic cell technology now point to a future where construction of synthetic tissue is within reach, a boon to pressing material needs in biomedical implants, drug delivery systems, adhesives, filters, and storage devices. Yet, controlling properties of synthetic tissue in a dynamic manner akin to its natural counterpart demands new molecular technologies. Here, I present on my lab’s recent efforts toward controlling synthetic tissue properties in real-time by manipulating molecular assembly state of cell junctions with user-defined inputs. I’ll first discuss our work engineering gap junction connexon assembly to enable material transfer and communication among synthetic cells. I’ll also cover our work designing protein switches that change conformation in response to an external stimulus, ultimately allowing control over cytoskeletal engagement.

1:00PM - 2:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Tae Seok Moon
all authors: Austin G. Rottinghaus, Steven Vo, and Tae Seok Moon*
Developing multilevel CRISPR-based kill-switches for biocontainment of genetically engineered microbes and microbiota
Microbial biocontainment is a critical goal for engineering safe living therapeutics, pollutant-degrading microbes, and nitrogen-fixing bacteria [1,2]. However, the genetic stability of biocontainment circuits, including kill switches, is a huge challenge that must be addressed. Kill switches are among the most difficult circuits to maintain due to the strong selection pressure they impart, leading to high potential for evolution of escape mutant populations. We developed two CRISPR-based kill switches in the probiotic Escherichia coli Nissle 1917: a single-input chemical-responsive switch and a 2-input chemical- and temperature-responsive switch [3]. We employed parallel strategies to address kill switch stability, including functional redundancy within the circuit, modulation of the SOS response, antibiotic-independent plasmid maintenance, and provision of intra-niche competition by a closely related strain. We demonstrate that strains harboring either kill switch can be selectively and efficiently killed inside the murine gut, while strains harboring the 2-input switch are additionally killed upon excretion into the environment. Leveraging redundant or layered strategies, we demonstrate robust biocontainment of our kill switch strains and provide a generalizable platform for future kill switch development, facilitating safe applications of genetically engineered microbes and microbiota in agricultural, environmental, medical, and manufacturing sectors [4]. [1] Amrofell, MB., Rottinghaus, AG., Moon, TS., Engineering microbial diagnostics and therapeutics with smart control. Current opinion in biotechnology 2020, 66, 11-17. [2] Rottinghaus, AG., Amrofell, MB., Moon, TS., Biosensing in Smart Engineered Probiotics. Biotechnology journal 2020, 15, 1900319. [3] Rottinghaus, AG., Ferreiro, A., Fishbein, SRS., Dantas, G., Moon, TS., Genetically stable CRISPR-based kill switches for engineered microbes. Nature communications. 2022, 13, 672 [4] Rottinghaus, AG., Vo, S., Moon TS, Computational design of CRISPR guide RNAs to enable strain-specific control of microbial consortia. PNAS. 2022, 120, e2213154120.

1:00PM - 2:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Ana Yagüe Relimpio
all authors: Ana Yagüe Relimpio*, Andreas Fink, Sebastian Fabritz, Alessia Ruggieri, Ilia Platzman, Joachim P. Spatz
Bottom-up assembled synthetic SARS-CoV-2 viruses reveal lipid membrane affinity of Omicron variant spike glycoprotein
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent for the still ongoing COVID-19 pandemic. The spike glycoprotein (S) decorates the viral envelope providing it with a crown-like appearance and is responsible for the first interaction of the virus with the cell. S binds to the angiotensin-converting enzyme 2 (ACE2) on the cell membrane, which initiates the infection process. From all the variants of concern, Omicron presents the highest amount of mutations in S. These changes in amino acid residues hint at new interacting partners, such as the cell membrane or proteoglycans. In this study, bottom-up assembled synthetic SARS-CoV-2 viruses (MiniVs) are implemented to study the binding properties of variants Omicron and Alpha. To that end, a functional screening assay is developed to investigate the affinity of Omicron and Alpha S to ACE2-functionalized and non-functionalized supported lipid bilayers. Furthermore, both variants are tested for competitive interaction using giant unilamellar vesicles as a cell membrane model. The findings evidence a significantly higher affinity of Omicron S to both functionalized and non-functionalized lipid membranes. The research presented here highlights the advantages of implementing synthetic SARS-CoV-2 MiniVs to study S binding affinity in a well-controlled manner and identify new interacting partners for future infectivity studies.



Building Blocks #3

Tuesday, May 23, Swain room, 1:00PM - 2:30PM
Zoom link to this session; to get password register as online participant.

1:00PM - 2:30PM
Tuesday, May 23rd
Swain room
presenter: Larry Wackett
all authors: Larry Wackett
Building a Cell That Loves PFAS
Fluorine is more abundant than carbon in the Earth’s crust and yet biological cells almost universally avoid fluorine. By contrast, the chemical industries have embraced fluorine, generating more than 19 million non-natural fluorinated chemicals, many of which are known by the common term PFAS. Part of the allure derives from the resistance of organofluorine (C-F) compounds to metabolism. Recent findings regarding the toxicity of C-F compounds and fluoride anion call for research to create new metabolism, to build cells that love PFAS. We have taken a systematic approach. First, we developed a convenient microtiter-well assay for C-F bond cleavage to detect fluoride anion at nmol concentrations. Using that, we identified existing enzymes that adventitiously react with certain C-F compounds. Following that, a cell sensor regulating hydrocarbon metabolism was co-opted and shown to induce enzymes reacting with C-F compounds. To use evolutionary selection, we constructed a Pseudomonas putida bacterium that was dependent on C-F compound metabolism to supply carbon and energy for cell proliferation. Dependence on C-F compounds released copious levels of fluoride anion. Fluoride is known for binding to biologically essential metals like calcium and magnesium, causing toxicity, and fluoride stress management is necessary. The current focus is on how bacteria can mitigate intracellular fluoride stress during C-F compound metabolism.

1:00PM - 2:30PM
Tuesday, May 23rd
Swain room
presenter: Prof. Pablo Ivan Nikel
all authors: P. I. Nikel
Designing and implementing synthetic metabolism in environmental bacteria
Fluorine is a key element for the industrial synthesis of molecules broadly used in medicine, agriculture and materials. Adding fluorine atoms onto organic structures is a unique strategy for tuning their molecular properties—yet organofluorines are only rarely found in Nature, and approaches to integrate fluorometabolites into the chemistry of living cells are scarce. Synthetic metabolism offers a unique opportunity towards expanding the chemical landscape of bacterial cells, thus providing alternative biosynthetic strategies for fluorinated building-blocks. This general approach will be illustrated by showing how novel fluorinating enzymes mined from extreme environments and synthetic gene circuits can be combined in the platform bacterium Pseudomonas putida for biosynthesis of organofluorine compounds. To this end, fluoride-responsive riboswitches and orthogonal RNA polymerases were exploited to drive the expression of the key elements needed to activate a synthetic biofluorination metabolism. Biosynthesis of fluoronucleotides, fluorosugars and fluorinated organic acids in engineered P. putida is demonstrated with mineral fluoride both as the only source of fluorine (i.e. as a substrate of the pathway) and as inducer of the synthetic circuits. Building on these results, prospects for bioproduction of fluorinated building blocks and materials in fluorination-addicted systems will be likewise discussed.

1:00PM - 2:30PM
Tuesday, May 23rd
Swain room
presenter: Donghoon Lee (Hoon)
all authors: Donghoon M. Lee*, Chanhee Park, Daehwan Kim
Life virtualization platform with L++: a complete programming language to build large-scale virtual biosystems
The genome is the blueprint for life, consisting of four nucleic acid bases that cells read and process. However, it is challenging for humans to directly interpret or modify the genome without annotation. We have developed L++, a comprehensive programming language specifically designed for biologists to unambiguously describe, design, execute, analyze, and debug large-scale biological systems, including organisms. We decoupled biology from computation and introduced intuitive syntax and grammar that can be learned in less than a day, making it accessible to individuals with minimal traditional computer programming experience. L++ features modular programming and includes essential traditional programming functions to enable efficient biology programming. Using L++, we are writing 100 biochemical pathways of E. coli, totaling around 500 pages of reusable code. This code integrates research from an array of heterogeneous databases and resources and can be used and repurposed through simulations and visualizations to efficiently create biochemical pathways, reprogram cells, and design and eventually create organisms. Our ultimate goal is to reverse engineer our knowledge of biology to reconstruct and execute human code, enabling full-scale systematic analysis and debugging of human diseases. With L++, we aim to facilitate the discovery of novel treatments and therapies, transforming the way we approach human health.

1:00PM - 2:30PM
Tuesday, May 23rd
Swain room
presenter: Rakesh Mukherjee
all authors: Rakesh Mukherjee*, Aditya Sengar, Javier Cabello-Garcia, Krizan Jurinovic, Luke Fehily, Thomas Ouldridge
A novel strand displacement motif to achieve catalytic control in DNA-templated reactions
Far-from-equilibrium templating is one of the fundamental features of many biological processes like DNA replication, RNA transcription, and protein translation. In such processes, an information-carrying template recognises the specific building blocks from a pool of possible monomers to form the desired product. These processes are also marked by the detachment of the template from the formed product so that a single copy of the template molecule can facilitate multiple reaction cycles and act as a catalyst. A synthetic mechanism with such features will be useful to design more complex nano systems. To demonstrate such functionalities in a DNA strand displacement network, we have developed a reaction motif called Handhold Mediated Strand Displacement (HMSD). In this presentation, we demonstrate the utilisation of HMSD to catalyse a sequence-specific dimerization reaction. Albeit this process working efficiently, the specificity is not extremely high. Such discrimination challenges in biological systems are mitigated by a process called “kinetic proofreading” where a fuel molecule is used to allow the small free-energy differences between the right and wrong building blocks to be used multiple times to increase the accuracy of the system. Here we demonstrate a DNA-based synthetic kinetic proofreading system to enhance the accuracy of HMSD. We also adjusted the system to detect single nucleotide polymorphism in ssDNA. Finally, we have extended the HMSD motif to design a self-replicator where both the “product” and the “template” are dimeric units of DNA. When a very low amount of pre-formed template is added to a pool of all those four DNA units, it catalyses the formation of the dimeric “product”. The “product” also catalyses the formation of the “template” via a similar HMSD process. In this complete network, the components show a sigmoidal growth characteristic of an autocatalytic system.



Containers #4

Tuesday, May 23, Johnson room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Tuesday, May 23rd
Johnson room
presenter: Chris Kempes
all authors: As part of the talk I will be reviewing published work by many co-authors, shall I provide that entire list even if none of them will be directly involved in this talk?
Cellular Scaling "Rules" for Synthetic Cell Design
In order to design synthetic cells it is important to find fundamental and condensed representations of cell physiology that capture the most important cellular "rules". In this talk I will review several categories of cellular rules that span the entire range of bacterial diversity. Primarily I will focus on scaling laws that determine cellular requirements for energy, macromolecules, and stoichiometry. I will discuss some recent results illustrating the connection between these scaling relationships and how they can be applied to synthetic cell design.

3:00PM - 4:30PM
Tuesday, May 23rd
Johnson room
presenter: Dieter Braun
all authors: Noël Yeh Martín, Alexander Floroni, Laura Weise, Sheref S. Mansy, Hannes Mutschler, Christof B. Mast, Dieter Braun
Thermal non-equilibria to assemble a synthetic cell
The bottom-up assembly of cells requires the autonomous accumulation of molecular building blocks into a spatially resolved, low entropy state. We found that temperature differences can lead to the assembly of such a structure, thereby generating membrane-free synthetic cells. A dilute and thus inactive transcription-translation machinery was accumulated by molecular thermophoresis and fluid convection, both triggered by the same temperature difference. Gene expression was activated by a coordinated increase of DNA, RNA, protein, and ion concentrations. The temperature difference confined the biomolecules over long periods of time, even when disturbed by a constant flow of water. The findings show how temperature differences can generate cell-like spatial organization without an active cell membrane, enabling a minimal way to engineer microfluidic cellular systems.

3:00PM - 4:30PM
Tuesday, May 23rd
Johnson room
presenter: Rebecca Schulman
all authors: Rebecca Schulman
Toward distributed computation in synthetic cell systems
A central goal of synthetic cell engineering is to create synthetic tissues in which different synthetic cells perform different small tasks in coordination so that together, they complete an otherwise hopelessly complex task. I will discuss how synthetic cells might solve such an example problem: could a community of synthetic cells collectively "see" a pattern, like the shape of a digit, by each detecting the local state and then together processing this information to make a coordinated decision? I will describe our ongoing efforts to address this question: building architectures of synthetic hydrogel cells using automated digital photolithograhy, developing a biochemical language involving transcriptional circuits for distributed computation, and learning to write programs for this discrimination that can be executed through simple local computation and information sharing by synthetic cells.



Applications #4

Tuesday, May 23, Ski-U-Mah room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Sam Schaffter
all authors: Samuel W Schaffter*, Molly E Wintenberg, Terence Murphy, Elizabeth Strychalski
Genetically encodable RNA circuits for programmable molecular computation in synthetic cells
The programmable and predictable control of nucleic acid strand displacement reactions make them a powerful tool for engineering diverse circuits that process chemical information. However, state-of-the-art strand displacement circuits composed of DNA are single-use and not easily genetically encoded, limiting their synthetic biology applications. Here, we present cotranscriptionally encoded RNA strand displacement (ctRSD) circuits, in which RNA strand displacement components can be transcribed together without substantial cross reaction, enabling genetic encoding of state-of-the-art strand displacement circuits that can be continuously produced in situ. We have programmed ctRSD circuits to execute a range of computational tasks with predictable and tunable dynamics in vitro. To facilitate construction of large circuits, we identified a toolkit of functional sequence domains and design approaches for reliably integrating these domains into circuit components and multi-layer cascades. Further, we developed open-source software for simulating the dynamics of diverse circuits. We are also characterizing ctRSD circuits with select RNases to enable coupled production and degradation so circuits can respond multiple times to changing inputs. These circuits could be genetically encoded in living or synthetic cells for designer molecular computation on a variety of chemical inputs. Towards these goals we are leveraging ongoing automation, cell free characterization, and chemical sensor engineering efforts in the Cellular Engineering Group at NIST.

3:00PM - 4:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: John Chaput
all authors: Chim N, Setterholm N, Lozoya-Colinas A, Majumdar B, Sarma D, Negrya S, Yik E, Lee E, Medina E, Hajjar M, Maola V, Yu Y, and Chaput JC
Redesigning the Genetic Polymers of Life
The birth of biotechnology marked a defining moment in American science that launched a multi-billion-dollar industry built on the concept that enzymes found in nature could be used to improve human health. This relatively simple idea revolutionized medicine by providing the tools need to synthesize, sequence, and manipulate DNA with exquisite precision. The panoply of applications that have emerged because of recombinant DNA technology—the core scientific discovery that fuelled the revolution—represent the vanguard of drug discovery and personalized medicine. Now, some forty-years later, we have begun asking the question: What discoveries could we make or what new technologies could we develop if we are no longer constrained to the natural genetic polymers of life? This talk will focus on the rise of artificial genetic systems, commonly referred to as XNAs, where X denotes a sugar other than ribose or deoxyribose. I will illustrate cases where natural enzymes have been redesigned by directed evolution to synthesize, propagate, and evolve artificial genetic polymers with backbone structures that are distinct from those found in nature. I will then demonstrate how XNAs are beginning to drive new applications in synthetic biology, biotechnology, and medicine.

3:00PM - 4:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Cheemeng Tan
all authors: Cheemeng Tan
Towards artificial cells for biomedical applications
It is challenging to create smart nano- and micro-devices that rival the dynamic chemical-biological response of natural cells. The nano- and micro-devices are biohybrid systems that mimic specific dynamical properties of living systems. To overcome the challenge, my lab investigates the operating principles of protein and gene networks. We then apply the principles to engineer various nano- and micro-devices. In this talk, I will discuss our recent work on using cell-free synthetic biology for the rapid prototyping of biological pathways, nano-scale drug-delivery vesicles, and membrane proteins. In addition, I will present the engineering of a new kind of micro-devices, termed Cyborg Cells, for broad biomedical applications. The Cyborg Cells are created by integrating synthetic materials with single natural cells. Cyborg Cells do not divide but preserve essential cellular functions, including cellular metabolism, motility, protein synthesis, and compatibility with genetic circuits. Cyborg Cells also acquire new abilities to resist stressors that otherwise kill natural cells. Finally, we demonstrate the therapeutic potential of Cyborg Cells by showing their invasion of various mammalian cells. Our work uses a unique holistic approach that integrates synthetic and systems biology. Furthermore, our work establishes a new paradigm in cellular bioengineering by creating non-living synthetic devices that mimic and potentially out-perform their living counterparts.

3:00PM - 4:30PM
Tuesday, May 23rd
Ski-U-Mah room
presenter: Jorin Riexinger
all authors: Jorin Riexinger, Thomas Caganek, Kevin R. Foster, Ravinash Krishna Kumar and Hagan Bayley
Patterned communication between synthetic tissues and living cells
Here, we describe the interaction between 3D-printed synthetic tissues and living cells by the patterned release of a chemical inducer. The release rate of the inducer can be tuned by a range of parameters, such as membrane pore or inducer concentration. By precisely matching inducer uptake rates of surrounding cells, patterned gene expression can be achieved. Additionally, two synthetic tissues may act as a switch, allowing for controlled inducer release once a specific connection is established between them.



Building Blocks #4

Tuesday, May 23, Swain room, 3:00PM - 4:30PM
Zoom link to this session; to get password register as online participant.

3:00PM - 4:30PM
Tuesday, May 23rd
Swain room
presenter: Kirill Alexandrov
all authors: Zhong Guo, Oleh Smutok, Cagla Ergun Ayva, Evgeny Katz and Kirill Alexandrov*
Towards real time information processing systems based on artificial allosteric proteins
Living organisms are multilayer energy and information processing systems where the external layer must operate rapidly in response to the environmental fluctuations. This layer is composed of protein-based networks to perform complex real time computation. The idiosyncratic relationship of protein structure and function, so far, prevented design and deployment of real time artificial protein-based signaling networks. We combine protein design and directed evolution to create artificial allosteric protein switches with tuneable input and output parameters. We discuss the thermodynamic parameters that determine the behaviour of natural and artificial switches and propose solutions for overcoming some of the inherent trade-offs. We demonstrate construction of protein switches controlled by ions, small molecules and proteins and discuss the ways they can be assembled into artificial signalling cascades.

3:00PM - 4:30PM
Tuesday, May 23rd
Swain room
presenter: Amelie Benk
all authors: Amelie S. Benk1*, Lucia T. Benk1, Jonas V. Schaefer2, Birgit Dreier2, Seraphine V. Wegner3, Yoav Peleg4, Shira Albeck4, Tamar Unger4, Peter Comba5, Benjamin Geiger6, Andreas Plückthun2, Joachim P. Spatz1 1Max Planck Institute for Medical Research, Dept. of Cellular Biophysics; 2Department of Biochemistry, University of Zurich; 3Max Planck Institute for Polymer Research and Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster; 4Structural Proteomics Center, The Weizmann Institute of Science; 5Inorganic-Chemical Institute and Interdisciplinary Center for Scientific Computing, Heidelberg University; 6Department of Molecular Cell Biology, The Weizmann Institute of Science
Controlled linkage of different proteins in synthetic cells
Shedding light into fundamental biological processes is often hindered by the extensive complexity discovered in living cells. Synthetic cell models with reduced and controlled complexity can therefore be a valuable tool for the detailed characterization of various vital mechanisms regarding individual proteins, protein systems and the cell. Recently, our lab could demonstrate the sequential bottom-up assembly of well-controlled, giant unilamellar vesicle (GUV)-based protocells equipped with purified cytoskeletal and adhesion associated proteins using high-throughput droplet-based microfluidic technologies (1). Controlled linkage of different proteins is a fundamental key in order to regulate self-assembly and functionality of cellular systems. Two different approaches are developed in our laboratory: (i) Implementing Designed Ankyrin Repeat Proteins (DARPins) as synthetic adaptor modules between natural proteins reduces the biological complexity by the restriction of the protein function to its binding capability. (ii) Beside these protein-mediated biochemical linkages, we additionally developed a simple, chemistry-based method for the coupling of proteins to surfaces and labels via metal complexes (2,3). Our compound proves site-specificity and kinetically inert immobilization of a protein to its target under physiological conditions providing a universal tool for various protein conjugation approaches. (1) Weiss M, Frohnmayer JP, Benk LT, Haller B, Janiesch JW, Heitkamp T, Börsch M, Lira RB, Dimova R, Lipowsky R, Bodenschatz E, Baret JC, Vidakovic-Koch T, Sundmacher K, Platzman I, Spatz JP. Sequential bottom-up assembly of mechanically stabilized synthetic cells by microfluidics. Nat Mater. 2018 Jan;17(1):89-96. doi: 10.1038/nmat5005. (2) Wegner SV, Spatz JP. Cobalt(III) as a stable and inert mediator ion between NTA and His6-tagged proteins. Angew Chem Int Ed Engl. 2013 Jul 15;52(29):7593-6. doi: 10.1002/anie.201210317. (3) Benk, AS, Schenk F, Wegner SV, Comba P, Spatz JP. Site-specific, kinetically inert conjugation of labels and/or carriers to target molecules such as his-tagged proteins via metal complex reagents. 2020. International Patent Number WO2020254539A1

3:00PM - 4:30PM
Tuesday, May 23rd
Swain room
presenter: Michael Levy
all authors: Michael Levy*, Reuven Falkovich, Shirley S. Daube, Roy. H. Bar-Ziv
Autonomous biogenesis of a ribosomal subunit on a chip
To qualify for being alive, protein-based minimal artificial systems should autonomously reproduce all their parts, including ribosomes. Establishing a scenario for concomitant synthesis and assembly of over sixty ribosomal proteins (r-proteins) and RNAs (r-RNA) into intact ribosomes has long been a bottleneck toward realization of a self-replicating biosystem. Here we reconstitute the biogenesis of the E. coli’s small ribosomal subunit (SSU), accomplished by expressing one r-RNA, twenty r-proteins and six assembly factors from surface-immobilized genes in a minimal cell-free reaction. The confinement to the surface created necessary conditions for concomitant synthesis and interactions of parts and for the visualization of nascent subunits, which accumulated on the surface spatially segregated from original ribosomes. Our real-time fluorescence measurements verified correct assembly by showing hierarchal binding, cooperative interactions, unstable intermediates, and specific binding to large ribosomal subunits (LSU). Using only synthetic genes, our methodology is a crucial step towards creation of a self-replicating artificial cell and a general strategy for the mechanistic investigation of diverse multi-component macromolecular machines.

3:00PM - 4:30PM
Tuesday, May 23rd
Swain room
presenter: Ane B. Søgaard
all authors: Ane B. Søgaard*, Andreas B. Pedersen, Alexander N. Zelikin
Transmembrane signaling by synthetic receptors in artificial cells
Transmembrane signaling is an essential process in cellular life. Natural cells sense, respond and adapt to their external environment by signal transduction pathways. The high complexity of this process makes it one of the major challenges in cell mimicry, toward the design of a fully synthetic cell. Herein, we present the design of synthetic enzyme activating receptor molecules made using tools of organic chemistry. The receptor is synthesized as a modular scaffold containing a lipid anchor to enable membrane association and a self-immolative linker as the mechanism of signal transduction. The modularity applies to both the exofacial trigger moiety, which can be exchanged to adjust the activating stimuli and the internally released messenger molecule, to fit the desired downstream function. We proved successful transmembrane signaling of multiple designs of synthetic receptors in cell-mimicking liposomes by measuring the fluorescence development of an encapsulated fluorogenic enzyme substrate. We investigated the signal transduction mechanism by comparing receptor-mediated signaling to simple diffusive signaling of the free messenger molecule and demonstrated that only receptor-mediated signaling lead to significant internal enzyme activation. The enzyme activation was also maintained in the presence of extracellular trap molecules, which confirms the position and internal release of the messenger molecule. Our results demonstrate the development of fully synthetic receptors that fulfills a hallmark of natural receptors, by enabling signal transduction based on an external stimulus that mediates internal activation of a downstream enzymatic signaling cascade. It represents an advancement within the field of artificial signal transduction and encourages the future of engineered responsive cells and cell mimicry.



Containers #5

Tuesday, May 23, Johnson room, 4:45PM - 6:00PM
Zoom link to this session; to get password register as online participant.

4:45PM - 6:00PM
Tuesday, May 23rd
Johnson room
presenter: Andreas Walther
all authors: Andreas Walther & Avik Samanta
Metabolic DNA Protocells
Living self-organizing systems operate far-from-equilibrium and display energy-dependent functionalities that are orchestrated through feedback loops and metabolic reaction networks to allow response, adaptation and communication in complex sensory landscapes. A cell is a formidable example that exhibits the Sense-Process-Act paradigm to sense and translate external signals, process and memorize them using Boolean and non-Boolean Algebra using its internal signaling network and act based on this outcome in order to internally adapt or communicate to the surrounding. This allows phenotype changes and communication for emergence in tissue formation and for morphogenesis. In this talk, I will discuss the formation of DNA-based protocell architectures using a recently discovered liquid-liquid phase separation process of ssDNA. Furthermore, I will show how secondary structures can be accessed inside as an artificial cytoskeleton, and that uptake of reactive species does not follow Fickian diffusion. Towards incorporation of a metabolism, I will reveal how abiotic catalysts and DNAzymes can be embedded within these artificial cells to drive downstream morphological adaptations.

4:45PM - 6:00PM
Tuesday, May 23rd
Johnson room
presenter: Nick Brooks
all authors: Nick Brooks*, Rob Law, Oscar Ces, Mohit Devgan, Giulia Mignini Urdaneta
Life in the extremes: Exploring membrane assembly in extreme environments
Oceans cover over 71% of the Earth’s surface with depths reaching almost 11km and pressures of over 1000 bar, and it is remarkable that we are still discovering hundreds of new species of marine life with every exploration. This reflects how the largest environment on earth remains the least explored and certainly least understood. The lipid membrane is the most pressure sensitive cellular component and pressure tolerant organisms are able to modify their molecular make-up in real time to ensure that their membranes remain functional, however, the mechanisms behind this adaptation and the parameters that are regulated remain elusive. We have developed a unique suite of instruments to study model and cellular membranes under high pressure, aiming to both understand the regulatory mechanisms employed by biology and use this evolutionary inspiration to develop active synthetic membranes that change and adapt their structure in response to changes in their environmental conditions. Further afield, there is currently huge interest in the deep sub-surface liquid water environments on Jupiter’s icy moons (particularly Europa and Ganymede). The ESA Juice probe which launched in April 2023, and the NASA Europa Clipper which will launch in October 2024 will give an unprecedented view of these environments with the explicit aim of understanding if the oceans on these moons could support basic life. Understanding how life on Earth survives in extreme environments is pivotal to understand how proto-life may blossom on other planetary systems and our instruments will form a laboratory testbed for exploring molecular assembly under the conditions found by these missions.

4:45PM - 6:00PM
Tuesday, May 23rd
Johnson room
presenter: Nina Conzelmann
all authors: Nina F. Conzelmanna*, Rongcong Luo, Sadaf Pashapour, Ilia Platzman and Joachim P. Spatz
Development of multifunctional, porous polymer-based microcapsules for antibacterial applications
Infections caused by pathogens such as bacteria are of rising concern in health care related sectors. Particularly, periprosthetic infections became a heavy burden due to the increased application of endoprostheses in society. Therefore, developing and improving strategies to avoid and battle implant-associated infections are of particular importance. Current antibacterial approaches are mainly based on the use of antibiotics and/or coatings that directly lyse the bacteria. However, the drawbacks of these strategies are the development of antibiotic resistances, and in the case of coatings, lysing bacteria in an ‘open space’ leads to acute inflammations due exposure of surrounding tissue to bacterial endotoxins and fragments. To circumvent these drawbacks, we developed multifunctional, porous, polymer-based microcapsules capable of capture, isolate and kill bacteria in a confined space. The pore geometry of the microcapsules was precisely tuned by a proper combination of functional biocompatible polymers. The optimized geometry of the pores allowed efficient penetration and isolation of bacteria within the hollow cavity of mechanically stable microcapsules. Moreover, we implemented biochemical modules within the microcapsule to enhance the killing efficiency of the captured bacteria. Furthermore, magnetic particles and gold nanorods (AuNRs) were integrated into the polymeric shell to further improve the bacteria capturing efficiency and their local killing by near-infrared radiation, respectively. The developed modular microcapsules can be potentially implemented either as efficient antibacterial coatings on various medical devices or as flushing solution.



Cell-free #1

Tuesday, May 23, Ski-U-Mah room, 4:45PM - 6:00PM
Zoom link to this session; to get password register as online participant.

4:45PM - 6:00PM

Ski-U-Mah room
presenter: Daisuke Kiga
all authors: Daisuke Kiga
Simplified genetic code for directed evolution of proteins without specific amino acid species
"During the early stages of life, it is probable that the genetic code consisted of fewer than the canonical set of 20 amino acids. What would the consequence have been for protein evolution during this period? To facilitate directed evolution with such restricted codes, we have developed artificial genetic codes utilizing a reduced alphabet. To facilitate directed evolution with such restricted codes, we have developed artificial genetic codes utilizing a reduced alphabet. Additionally, this fundamental modification of the biological system serves as an innovative engineering tool because some amino acids can be a weak point: Additionally, this fundamental modification of the biological system serves as an innovative engineering tool because some amino acids can be a weak point: For instance, the oxidation of Cysteine or Methionine can significantly reduce the activity of proteins.

4:45PM - 6:00PM

Ski-U-Mah room
presenter: Danielle Pedrolli
all authors: Brenner, G.; Franco, R.A.L.; Zocca, V.F.B.; de Paiva, G.B.; Lima, R.N.; Rech, E.L.; Lins, M.R.C.R.; Pedrolli, D.B.*
Signal-amplification for cell-free biosensors, an analog-to-digital converter
Toehold switches are biosensors useful for the detection of endogenous and environmental RNAs. They have been engineered to detect virus RNAs in cell-free gene expression reactions. Their inherent sequence programmability makes engineering a fast and predictable process. Despite improvements in the design, toehold switches suffer from leaky translation in the OFF state, which compromises the fold change and sensitivity of the biosensor. To address this, we constructed and tested signal amplification circuits for three toehold switches triggered by Dengue and Sars-CoV-2 RNAs and an artificial RNA. The serine integrase circuit efficiently contained leakage, boosted the expression fold-change from OFF to ON, and decreased the detection limit of the switches by three to four orders of magnitude. Ultimately, the integrase circuit converted the analog switches’ signals into digital-like output. The circuit is broadly useful for biosensors and eliminates the hard work of designing and testing multiple switches to find the best possible performer.

4:45PM - 6:00PM

Ski-U-Mah room
presenter: Evan Kalb
all authors: Evan Kalb*, Jose L. Alejo, Josh Davisson, Kate Adamala
Post-transcriptionally modified synthetic tRNAs for enhanced cell-free translation
Transfer RNAs (tRNAs) act as the mediators of translation by delivering amino acids to the ribosome. Native tRNAs are often extensively adorned with many post-transcriptional modifications which can serve as identity elements for aminoacylation, enable faithful decoding, or enhance interactions with the translational machinery. For convenience, synthetic tRNAs are usually generated using in vitro transcription which renders them devoid of these important modifications. To enable the use of synthetic tRNAs which bear post-transcriptional modifications, we have developed a method which couples the heterologous expression of tRNA to a hybridizing-probe purification process. Using this method, we have purified the orthogonal tRNApCNF from M. jannaschii at a >10 nmole scale with high purity. We show that the in vivo tRNApCNF is indeed modified by the host E. coli and outperforms its in vitro synthesized counterpart in stop-codon suppression assays. Our results underscore the importance of post-transcriptional modification within tRNAs and offer a simple and convenient way to generate these substrates.



Building Blocks #5

Tuesday, May 23, Swain room, 4:45PM - 6:00PM
Zoom link to this session; to get password register as online participant.

4:45PM - 6:00PM
Tuesday, May 23rd
Swain room
presenter: Alexander Zelikin
all authors: Alexander Zelikin
Adaptive, responsive behavior in synthetic cells
Design and engineering of artificial, synthetic cells is fundamentally intriguing and important for a suit of emerging applications in biosensing and synthetic biology. In our work, we focus on the development of adaptive, responsive synthetic cells. Learning from nature, we aim to achieve this using receptors as the communications chassis, and encapsulated catalytic cascades and down-stream signaling events. More specifically, we aim to achieve this using tools of chemistry. Thus, the overarching theme of our research is the development of artificial chemical receptors and engineering of tools for reversible inactivation of enzymes. One deliverable of our research is the development of chemical zymogens for diverse enzymes: proteases, kinases, polymerases. In each work, chemical zymogens are engineered around the protein cysteinome such that the conversion of a protein thiol into a corresponding mixed disulfide reversibly encaptivates the enzyme. Another highlight is the development of transmembrane signaling whereby an enzymatic reaction in the extracellular space ensues the signal transduction across a lipid bilayer and culminates in the activation of enzymes in the intracellular space of a synthetic cell. Artificial receptors designed in our work are engineered to release thiol-containing secondary messenger molecules to directly couple to the engineered chemical zymogens. Using these tools, in synthetic cells and using chemical receptors and chemical zymogens, we achieved transmembrane activation of proteolysis, production of ATP, and/or initiation of transcription. Synthetic cells engineered in our work were responsive to biochemical and even inorganic stimuli, mimicking natural adaptive behavior of natural cells and possible responses in protocells at the birth of life.

4:45PM - 6:00PM
Tuesday, May 23rd
Swain room
presenter: Shankar Mukherji
all authors: Shankar Mukherji
Building the cell from unreliable parts: coordinating stochastic organelle biogenesis with cellular growth
Perhaps the defining feature of the eukaryotic cell is its organization into membrane-bound compartments known as organelles. While the processes underlying the biogenesis of individual organelles are often well-known, the precision with which individual cells exert quantitative control over individual organelle properties, such as number and size, and coordinate these properties at systems-scale across the cell’s many different types of organelles remain frontier problems in cell biology and biophysics. Using a combination of theory and quantitative fluorescence imaging, I will describe our recent efforts to show that cells exhibit substantial limits to the precision with which they can control organelle numbers and sizes, but despite this appear to collectively organize systems-level organelle biogenesis into specific “modes” that balance the need for cell growth to be both robust yet responsive to the environment.

4:45PM - 6:00PM
Tuesday, May 23rd
Swan room
presenter: Céline Cleij
all authors: Céline Cleij*, Laura Sierra Heras, Pascale Daran-Lapujade, Christophe Danelon
Building the genome: yeast assembly of synthetic chromosomes for expression in the PURE system
De novo design and assembly of a DNA genome represents a major challenge for the construction of a minimal synthetic cell. We have explored homologous recombination in the yeast Saccharomyces cerevisiae as a method for the assembly of large coding DNA (>50 kb) from multiple fragments (>10). The compatibility of this approach with the genetic constraints imposed by usage of an in vitro transcription-translation system, called PURE system, was tested. Two challenges were identified: (1) assembly of sequences with repeats arising from the limited choice of regulatory elements in PURE and (2) isolation of the assembled synthetic chromosome from yeast, which results in DNA with low concentration and purity for expression in PURE system. In this study, we show that in-yeast assembly of 10 DNA fragments with repeats and 10 marker fragments into a 68-kb chromosome is possible, albeit with low efficiency. We propose a screening pipeline to strategically identify correct assemblies by using auxotrophic, fluorescent, antibiotic resistance and color markers. Assemblies are verified by long-read Nanopore sequencing. Preliminary results for isolation of a small-sized prototype chromosome from yeast containing a reporter gene, show successful expression in PURE system. The isolation protocol needs further improvement to enable PURE expression of multiple genes from larger constructs. Future efforts will focus on engineering in-yeast assembled synthetic genomes with origins of replication and creating genetic diversity, for information continuity and evolution in liposomes.



Tuesday, May 23, keynote lectures
Zoom link to this session; to get password register as online participant.


Dora Tang

Tuesday, May 23, Memorial Hall, 9:15AM - 10:00AM

Using bottom up approaches to unravel the chemical-physico principles of life

One of the goals of bottom-up synthetic biology is to build living cells from scratch. Biology is well equipped in exploiting a large number of out-of-equilibrium processes to support life. A complete understanding of these mechanisms is still in its infancy due to the complexity and number of the individual components involved in the reactions. However, a bottom-up approach allows us to replicate key biological processes using a small number of basic building blocks. Moreover, this methodology has the added advantage that properties and characteristics of the artificial cell can be readily tuned and adapted.
In this talk, I will provide an overview of the strategies we adopt in our lab to build living systems from scratch that rely on compartmentalisation as the defining feature to support out-of-equilibrium behaviour. Specifically, I will talk about the design and synthesis of artificial cells based on liquid-liquid phase separation (coacervation) and hydrophobic effects such as lipid vesicles and proteinosomes and describe how these compartments may be used as platforms for implementing dynamic biological behaviours including: RNA catalysis and communication. I propose that our bottom-up approaches are effective in establishing living systems from scratch and in doing so provide unique model systems that can help to unravel the physico-chemical principles of living systems.



Sebastian Maerkl

Tuesday, May 23, Memorial Hall, 10:00AM - 10:45AM

On Biochemical Constructors and Synthetic Cells

Cell-free synthetic biology emerged as a viable in vitro alternative for biological network engineering. Cell-free synthetic biology implements biological systems in a coupled transcription – translation reaction and therefore is a well-defined environment that is easier to control and interrogate than complex cellular systems. I will discuss several technological and methodological advances including the development of microfluidic chemostat devices, a high-throughput microfluidic device, and a method to easily produce a recombinant cell-free system. With these tools we were able to rapidly prototype genetic networks and transplant them into living hosts and engineered gene regulatory networks from the bottom-up with synthetic Zinc-finger transcriptional regulators. More recently, we were able to demonstrate that it is possible to create a partially self-regenerating cell-free system that continuously produces up to seven essential proteins and does so for an extended period of time. This work lays the foundation for the development of a universal biochemical constructor and may ultimately enable the creation of a synthetic cell.



Tuesday, May 23 conference dinner

The conference dinner will be at Malcolm Yards. All registered participants are invited to attend; dinner and one drink is included in conference registration price.

Malcolm Yards is a food hall located 20 minute walk from the conference venue.

Address: The Market At Malcolm Yards, 501 30th Ave SE, Minneapolis, MN 55414.

Wednesday, May 24 sessions and keynotes

Wednesday, May 24, breakout sessions


Cell-Free Systems #2

Wednesday, May 24, Ski-U-Mah room, 1:00PM - 3:00PM
Zoom link to this session; to get password register as online participant.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Kosuke Fujishima
all authors: Shota Nishikawa, Po-Hsiang Wang, Shawn McGlynn, and *Kosuke Fujishima
De novo synthesis of iron-sulfur proteins using recombinant SUF with O2-scavenging cascade in a cell-free system
Iron-sulfur clusters are essential cofactors mediating single-electron transfer in respiratory and metabolic networks. However, heterologous iron-sulfur protein expression is challenging due to (i) the requirements for iron-sulfur cluster assembly, (ii) the O2 lability of iron-sulfur clusters. Here, we established a facile and efficient protocol to express mature [4Fe-4S] proteins in the reconstituted cell-free translation system (i.e., the PURE system) under aerobic conditions. The incorporation of recombinant SUF helper proteins to the PURE system enabled streamlined mRNA translation and [4Fe-4S] cluster assembly under anaerobic conditions. To reconcile the O2 lability of [4Fe-4S] clusters, we further added an O2-scavenging three-enzyme cascade to the PURE system. In this cascade, formate dehydrogenase, flavin reductase and catalase are used to reduce O2 to water. This cascade also supplies FADH2 to the SUF helper proteins as well as removing O2 from the reactions, allowing a one-pot synthesis of mature [4Fe-4S] proteins under aerobic conditions with a high maturation rate (>90%). Our new approach will expand the repertoire of cell-free biocatalysis, paving the way for future reconstruction of redox-active synthetic cells.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: James Pelletier
all authors: James F. Pelletier*, Lijie Sun, Kim S. Wise, Nacyra Assad-Garcia, Bogumil J. Karas, Thomas J. Deerinck, Mark H. Ellisman, Andreas Mershin, Neil Gershenfeld, Ray-Yuan Chuang, Saúl Ares, John I. Glass, and Elizabeth A. Strychalski
Relating genotype and phenotype during cell division in the genomically minimal cell JCVI-syn3.0
Engineered to have as few genes as possible, genomically minimal cells offer a simplified system to study relationships between genotype and phenotype. Even in the minimal genomic context of JCVI-syn3.0, which has the smallest genome of any free-living cell, fundamental physiological processes often depend on many genes. For example, cell division depends on multiple genes of known and unknown function. To guide future experiments, we consider a physical description of cellular mechanics as an interlayer between genotype and phenotype. We review current knowledge of genes contributing to two physical parameters relevant to cell division, namely, the surface-area-to-volume ratio and membrane curvature. This physical view of JCVI-syn3.0 may inform the attribution of gene functions and conserved processes in bacterial physiology, as well as whole-cell models and the engineering of synthetic cells.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Ion Ioannou
all authors: Ion Ioannou*, Carolina Monck, Nick Brooks, Marina Kuimova, Francesca Ceroni and Yuval Elani
Genetic driven communication in compartmentalised synthetic cells
Compartmentalisation of biologically relevant processes in cell-sized liposomes provides the groundwork towards engineering bottom-up artificial cells of increased complexity. Encapsulation of cell-free protein synthesis systems (CFPSs) has also been employed in the field, not only to control transcription and translation of selected genes but also to integrate the power and versatility of genetic systems in synthetic cell engineering. To date, CFPSs have been used successfully in separate liposomal/microdroplet compartments. In this project we developed a method for the production of compartmentalised giant unilamellar vesicles (GUVs) and for the first time achieved genetic driven trans-compartmental communication. Encapsulation of CFPSs in the core compartment enabled the formation of pores and substrate transfer from the external compartment, in order for enzymatic reactions to occur in the cell core. Our results demonstrate how a novel and versatile liposomal architecture can be combined with cell-free expression to effectively produce controlled enzymatic responses, having the potential to be further developed and support more complex out of equilibrium behaviours.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Andrei Sakai
all authors: Andrei Sakai*, Aafke J. Jonker, Frank H. T. Nelissen, Evan M. Kalb, Christopher R. Deich, Bob van Sluijs, Hans A. Heus, Katarzyna P. Adamala, John I. Glass, Wilhelm T. S. Huck
Cell-free expression system derived from JCVI-syn3A cells
Cell-free expression (CFE) systems are a necessary platform for the construction of a synthetic cell. Here, we report the successful accomplishment of a CFE system derived from JCVI-syn3A. In previous studies, high ribonuclease activity found in Syn3A lysates produced by traditional cell disruption methods (e.g. French press or sonication) impeded the establishment of functional CFE systems. Now, we describe how an unusual cell disruption method (nitrogen decompression) yielded Syn3A lysates that supported in vitro expression. To improve the protein yields in the Syn3A CFE system, we optimized the Syn3A CFE reaction mixture using an active machine learning tool. In conclusion, we hope the JCVI-syn3A CFE system can be useful for the synthetic cell community.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Anton Jackson-Smith
all authors: Anton Jackson-Smith, Will Roberts, Youngju Kim, Wah Chiu, Drew Endy
Integration and Measurement for Building Synthetic Cells
Integration–combining the many cell-essential molecular functions such that they work together–is the key challenge is building a functioning synthetic cell. In order to integrate, we must be able to measure multiple genetic circuits and molecular components working together, in the same cell, at the same time. We have prototyped a platform for integration, using cryo-electron microscopy to visualize individual synthetic cells operating integrated molecular modules, quantifying and characterizing their performance.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Akshay Maheshwari
all authors: Akshay Maheshwari, Jonathan Calles, Sean Waterton, Drew Endy
Engineering tRNA abundances for synthetic cellular systems
Making routine the engineering of synthetic cells will require specifying beforehand how many of each molecule are needed. First-principles tools for specifying molecular abundances enabling whole-cell synthetic biology are missing. We use a colloidal dynamics simulator to make predictions for how tRNA abundances impact protein synthesis rates. We use rational design and direct RNA synthesis to make 21 synthetic tRNA surrogates from scratch. We use evolutionary algorithms within a computer aided design framework to design translation systems predicted to work faster or slower depending on tRNA abundance differences. We build and test the so-specified synthetic systems and find that expected and observed system performance match. First-principles modeling combined with bottom-up experiments can help molecular-to-cellular scale synthetic biology realize “design, build, work” frameworks that transcend tinker-and-test.

1:00PM - 3:00PM
Wednesday, May 24th
Ski-U-Mah room
presenter: Kaito Seo
all authors: Kaito Seo*, Norikazu Ichihashi
Investigating compatibility of non-protein factors in DNA replication, transcription, and translation
Essential functions of cells have been reconstituted in vitro to understand the system of life. The next challenge toward the construction of a fully artificial living system is the integration of each reconstituted function. In this study, we focused on reactions consisting of central dogma: DNA replication, transcription, and translation, and the effect of non-protein factors in the in vitro system for each reaction. While these reactions were integrated into the one-pot reaction in the past study, the optimal conditions for each reaction were different. Our results showed that few factors have inhibiting effects while promoting the other. For example, dNTP is required for DNA replication as a substrate but inhibits translation reaction. rNTP and tRNA are necessary for translation, but both inhibit DNA replication by various DNA polymerases. Also, some of the inhibiting effects were resolved by adjustment of magnesium concentration. As a result of optimization, we achieved transcription/translation coupled DNA replication with higher transcription and translation rate, with a constant amount of DNA replication. Our result would be useful for constructing artificial cells implemented with central dogma and pose a question of how these incompatibilities are solved within living cells.



Building Blocks #6

Wednesday, May 24, Swain room, 1:00PM - 3:00PM
Zoom link to this session; to get password register as online participant.

1:00PM - 3:00PM
Wednesday, May 24th
Swain room
presenter: Matthew Lakin
all authors: Tracy L. Mallette, Matthew R. Lakin
Heterochiral DNA for information processing in synthetic cells
DNA chirality is an underused design variable in DNA nanotechnology. Integrating mirror-chirality L-DNA into DNA nanostructres and nanodevices enables the creation of bioorthogonal molecular devices that resist degradation by naturally occurring nucleases. Combining naturally occurring D-DNA with L-DNA leads to heterochiral DNA devices, which can be used to translate input signals of one chirality into output signals of the opposing chirality via heterochiral DNA strand displacement. Integrating protective L-DNA caps can also be used to enhance the resistance of D-DNA domains in heterochiral devices to degradation by nucleases in biological environments. Such devices could be used to construct or augment information processing systems in engineered synthetic cells to operate independently of any biologically derived components, such as crude cell extracts.

1:00PM - 3:00PM
Wednesday, May 24th
Swain room
presenter: Lado Otrin
all authors: Lado Otrin*, Nika Otrin, Claudia Bednarz, Kai Sundmacher
Bottom-up Assembly of a Prokaryotic Synthetic Cell
We set out to build from the bottom-up a new form of synthetic life – a motile omnitroph capable of sensing and processing the surrounding environment. The organism can detect a variety of stimuli and can react to them by approaching or evading them. It is comprised of several functional modules powered by a hybrid energy and cofactor regenerating engine, designed for simultaneous harnessing of light and chemical energy. The assembly of such organism requires finely controlled integration of natural building blocks from all domains of life with the synthetic materials. In this context, different assembly tools and technologies, such as recursive SNARE-mediated fusion, are discussed.

1:00PM - 3:00PM
Wednesday, May 24th
Swain room
presenter: Isabella Nymann Westensee
all authors: Isabella Nymann Westensee*, Lars Paffen, Brigitte Städler
Mimicry of Communication between Artificial and Natural Cells
Selected structural and functional elements of natural cells can be reflected in artificial cells (ACs). Communication between ACs and mammalian cells is an important feature to mimic when aiming to fabricate hybrid systems that can coordinate their behaviour in response to external challenges. We explore the communication between alginate-based ACs and HepG2 cells in two different approaches. First, a one-way signal transfer between two different types of ACs and HepG2 cells is demonstrated. Metalloporphyrins (MPs) are used as artificial enzymes that mimic cytochrome P450 enzyme (CYP450) activity, and provide catalytic function to the ACs. The MPs catalyse a dealkylation and hydroxylation reaction, which produce fluorescent products as model signalling molecules that can diffuse into neighbouring HepG2 cells. In a subsequent effort, the MP-functionalized ACs were bioprinted together with HepG2 cell aggregates to obtain bionic structures with a specific liver-like activity. Second, we illustrate the potential of ACs to eavesdrop on the CYP450 activity in HepG2 cells. Alginate-based ACs are functionalized with D-cysteine, which was release when triggered. The available D-cysteine reacted with a conversion product from CYP450 in HepG2 cells to produce D-luciferin. This latter compound was detected as a luminescence signal upon enzymatic conversion by luciferase encapsulated in the ACs. Taken together, these efforts are a step towards reciprocal communication between mammalian cells and ACs towards the assembly of adaptive semi-synthetic tissue.

1:00PM - 3:00PM
Wednesday, May 24th
Swain room
presenter: Ned Budisa
all authors: N/A
Top-down approach to synthetic life through directed evolution of synthetic bacterial cells with alternative genetic codes
Life on Earth is a single entity thanks to the universal genetic code, i.e. the genetic code is basically the same for all organisms - all living things use the same "genetic language". This biological framework enables the translation of the genetic message written in DNA into life-sustaining proteins consisting of 20 canonical amino acids. Why "only" 20? Recently, we proposed the Alanine World Model, which explains the selection of amino acid monomers in the genetic code repertoire from the perspective of peptide and protein chemistry. Our model, together with the RNA world hypothesis (and the Hartman-Smith model) and co-evolution theory, provides a plausible scenario for the chemical etiology of the amino acid repertoire in the genetic code. The best way to test these models and theories is to experimentally attempt to create parallel biological worlds based on fundamentally different chemistry, metabolism, and energetics of life, with dramatically different genetic codes.

1:00PM - 3:00PM
Wednesday, May 24th
Swain room
presenter: Eleonora Bailoni
all authors: Eleonora Bailoni, Miyer F. Patiño-Ruiz, Andreea R. Stan, Ronald van der Meulen, Gea K. Schuurman-Wolters, Marten Exterkate, Arnold J.M. Driessen and Bert Poolman
ATP recycling coupled to phospholipid biosynthesis in feeder-utilizer synthetic organelles
ATP is an universal energy currency essential for life. Using purified proteins reconstituted in vesicles, ATP can be generated from the breakdown of L-arginine. We exploit L-arginine-dependent ATP generation for the synthesis and export of glycerol 3-phosphate, and we use the precursor for phospholipid biosynthesis on the outer surface of utilizer vesicles. Glycerol 3-phosphate export is coupled to the import of inorganic phosphate, ensuring a constant pool of phosphate for the synthesis of ATP. We thus develop an out-of-equilibrium metabolic network for the generation of ATP and the coupling of metabolic energy to the synthesis of lipids in a feeder-utilizer, organelle-like system inspired by compartmentalization in eukaryotic cells.



Wednesday, May 24, keynote lectures
Zoom link to this session; to get password register as online participant.


Paul Freemont

Wednesday, May 24, Memorial Hall, 9:15AM - 10:00AM

Constructing synthetic cells – Biology’s 21st century grand challenge

The construction of synthetic cells constitutes one of the greatest scientific challenges of the 21st century and involves researchers from many disciplines including chemistry, physics, biology, engineering and maths to name a few. This intensely interdisciplinary activity confronts the fundamental nature of living systems by attempting to construct a synthetic cell with the properties of life. Through such a grand challenge we will not only learn about the detailed molecular functions of living systems but also extend the materiality of living systems to elements not found in life. I will discuss different approaches to address this grand challenge and also introduce the role of biofoundries to enable systematic measurement and reproducible construction providing access for many more researchers to join the field. I will also cover our recent work on cell-free engineering of extracellular vesicles and introduce the concept of hybrid synthetic cells.



John Glass

Wednesday, May 24, Memorial Hall, 10:00AM - 10:45AM

Design, Construction, and Analysis of a Synthetic Minimal Bacterial Cell

The minimal cell is the hydrogen atom of cellular biology. Such a cell, because of its simplicity and absence of redundancy, would be a platform for investigating just what biological components are required for life, and how those parts work together to make a living cell. Since the late 1990s, our team at the Venter Institute has been developing a suite of synthetic biology tools that enabled us to build what previously has only been imagined, a minimal cell. Specifically, a bacterial cell with a genome that expresses only the minimum set of genes needed for the cell to divide every two hours that can be grown in pure culture. That minimal cell has about half of the genes that are in the bacterium on which it was based, Mycoplasma mycoides JCVI syn1.0, the so-called synthetic bacteria we reported on in 2010. We used transposon bombardment to identify non-essential genes, and genes needed to maintain rapid growth in M. mycoides. Those findings required re-design and re-synthesis of some reduced genome segments. Three cycles of design, synthesis, and testing, with retention of quasi‐essential genes, produced synthetic bacterium JCVI‐Syn3.0 (531 kb, 474 genes), which has a genome smaller than that of any autonomously replicating cell found in nature. Synthetic bacterium JCVI-Syn3.0 retains almost all genes involved in synthesis and processing of macromolecules. Surprisingly, it also contained 149 genes with unknown biological functions, suggesting the presence of undiscovered functions essential for life. This minimal cell is a versatile platform for investigating the core functions of life, and for exploring whole‐genome design. Since it was initially reported in 2016, we have identified functions for about 50 of the original 149 genes of unknown function. These findings have been used to create flux balance analysis and kinetic whole cell computational models of our minimal cell that replicate laboratory observations about our minimal cell. Additionally, we have used JCVI-syn3.0, which has an abnormal cell division and cell morphology phenotype, and a JCVI-syn3.0 mutant containing an additional seven non-essential genes that has divides normally and looks like wild type Mycoplasma mycoides to investigate how modern cell division and cell size control might have evolved.



 
 

Local information

Meeting will be on the Minneapolis campus of the University of Minnesota, at the McNamara Alumni Center.

200 SE Oak St, Minneapolis, MN 55455

All participants are responsible for their own accommodations.

Travel

Minneapolis International Airport, MSP, is well connected with the University campus. You can take the train from the airport directly to campus, or you can take taxi or ride share (Uber or Lyft).

Hotels

Please book your accommodations early! May is the time when prospective students and parents visit the campus, and UMn has about 6,000 freshmen each year. That corresponds to a lot of on campus visits, and hotels in this area fill up fast.

We do not have conference rate at any of the local hotels.

Below is a list of hotels walking distance to the McNamara Center, the conference site.

The Graduate Hotel, graduatehotels.com/minneapolis/ - this hotel is connected by skyway with the McNamara Center.

Hilton Garden Inn Minneapolis University Area, hilton.com/en/hotels/msputgi-hilton-garden-inn-minneapolis-university-area/

Days Hotel by Wyndham University Ave SE, wyndhamhotels.com/days-inn/minneapolis-minnesota/days-inn-hotel-university-ave-se/overview

Also close, about 15 minute walk from the conference site:

Hampton Inn & Suites Minneapolis University Area, hilton.com/en/hotels/mspuahx-hampton-suites-minneapolis-university-area/

Visa

If you need visa invitation letter, please email Kate Adamala.

 
 

SynCell conference series

Previous meetings in the SynCell series: SynCell 2022, SynCell 2021, SynCell 2020.

Spiritual predecessor of SynCell series: BaSyC2018.

 
 

Organizers

The organizing committee

  • Jacqueline De Lora – Max Planck Institute for Medical Research, Stuttgart

  • Marileen Dogterom – Delft University of Technology

  • Gabriel López – University of New Mexico

  • Joachim Spatz – Max Planck Institute for Medical Research, Stuttgart

  • Christophe Danelon - Delft University of Technology

  • Stefania Usai - Delft University of Technology

  • Kate Adamala – University of Minnesota

  • Carlise Sorenson - University of Minnesota

 
 

Sponsors

We are grateful for sponsors who made this meeting possible.