Download as pdf or txt
Download as pdf or txt
You are on page 1of 83

December 2012 Volume 22, Number 12 pp.

611–692

Special Issue – Synthetic Cell Biology

Editor
Editorial
Rebecca Alvania
Executive Editor, Cell Biology 611 Cell Biology 2.0 Wendell A. Lim, Rebecca Alvania, and
Deborah Sweet Wallace F. Marshall
Journal Manager
Jeanette Bakker
Forum: Science & Society
Journal Administrators 613 Science ⴙ dance = bodystorming Carl Flink and David J. Odde
Patrick Scheffmann
Ria Otten
Opinion
Advisory Editorial Board
Wolfgang Baumeister, Munich, Germany 617 Synthetic multicellularity Michel M. Maharbiz
Dominique Bergmann, Stanford, USA
Pascale Cossart, Paris, France
David Drubin, Berkeley, USA Reviews
William C. Earnshaw, Edinburgh, UK
Scott Emr, Ithaca, USA
624 Designer nucleic acids to probe and Yamuna Krishnan and Mark Bathe
Anne Ephrussi, Heidelberg, Germany program the cell
Susan Gasser, Geneva, Switzerland
David Goldfarb, New York, USA
634 Towards a bottom-up reconstitution Ariadna Martos, Mercedes Jiménez,
Ari Helenius, Zurich, Switzerland of bacterial cell division Germán Rivas, and Petra Schwille
Nobutaka Hirokawa, Tokyo, Japan
644 Engineered, harnessed, and hijacked: Brian S. Goodman, Nathan D. Derr, and
Alan Rick Horwitz, Charlottesville, USA
Tony Hunter, La Jolla, USA synthetic uses for cytoskeletal systems Samara L. Reck-Peterson
Chris Marshall, London, UK
Sascha Martens, Vienna, Austria 653 Principles for designing ordered protein Yen-Ting Lai, Neil P. King, and
Andrew Matus, Basel, Switzerland assemblies Todd O. Yeates
James Nelson, Stanford, USA
Hugh Pelham, Cambridge, UK 662 Designing biological compartmentalization Anna H. Chen and Pamela A. Silver
Hidde Ploegh, Cambridge, USA
671 Directed cytoskeleton self-organization Timothée Vignaud, Laurent Blanchoin,
James R. Woodgett, Toronto, Canada
and Manuel Théry
Editorial Enquiries 683 Directing the assembly of spatially Jennifer S. Liu and Zev J. Gartner
Trends in Cell Biology
Cell Press organized multicomponent tissues from
600 Technology Square the bottom up
Cambridge, MA 02139, USA
Tel: +1 617 386 2105
Fax: +1 617 397 2810
E-mail: tcb@cell.com

Cover: In recent years, the bottom-up approach of synthetic biologists has yielded new insight into fundamental aspects of cell biology.
In this special issue, co-guest edited by Wendell A. Lim and Wallace F. Marshall (editorial on pages 611–612), we highlight some of the
exciting work that has sprung from this intersection between synthetic and cell biology. On the cover, the construction of a single cell is
depicted via an instruction sheet similar to that which might be found in a child’s game. The cover is meant to represent the constructionist
approach to understanding the inner workings of the cell. Cover design by Yvonne Blanco.
Editorial

Special Issue – Synthetic Cell Biology

Cell Biology 2.0


Wendell A. Lim1, Rebecca Alvania3, and Wallace F. Marshall2
1
Department of Cellular and Molecular Pharmacology, University of California, San Francisco, Genentech Hall – N412E,
600 16th Street, San Francisco, CA 94158, USA
2
Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16th Street, San Francisco,
CA 94143-2200, USA
3
Editor, Trends in Cell Biology

‘Verum esse ipsum factum’, the true is in the made – manipulation and perturbation of systems can take place in
Giambattista Vico the milieu of the living cell, with its basic metabolic and
Synthetic Cell Biology sounds intriguing, but the name transcriptional systems and complex spatial environments.
begs the question – why should we try to rebuild or repro- The cell, or even artificially constructed cells, can serve as
gram the cell, especially when we barely understand how the new test tube for logical and systematic analysis of a
cells work? This issue of TiCB explores this emerging area process. As Vico first suggested (and Richard Feynman later
in which scientists are taking apart, rebuilding, repro- reiterated), if we can build it, we can understand it.
gramming, and repurposing parts of the cell. The reviews Predictive, mechanistic understanding of how the parts
cover a wide range of scales, from microscopic molecular of a cell work together to generate cell behaviors is not only
machines to macroscopic, multicellular tissues. These interesting in its own right; it would provide a rational
reviews highlight the fact that, in addition to its role in basis to turn cell biology into an engineering discipline.
harnessing and unleashing the power of cells for new and Synthetic cell biology has the promise of remarkable bio-
future applications, synthetic biology also has an impor- technology applications. Many advances have been made
tant role to play in facilitating the understanding of com- in harnessing cells to create new and useful molecules, by
plex cellular processes. In short, we can learn much about cobbling together new strings of metabolic enzymes. But in
cells through the process of trying to build them (or parts of many ways, this is only the tip of the iceberg of what cells
them). And if we understand cells, we can start to really could do. Current efforts have largely ignored the cell, its
harness their power. structure and regulation, as a tool to harness. Cells are
Cell biology has historically been primarily an observa- some of the most powerful and capable devices we know,
tional science, inextricably linked with tools like the mi- especially in producing complex products. Manipulation of
croscope, which first enabled humans to peer into the cellular organelles, transport systems, and secretion sys-
remarkable world of the cell, with its diversity of forms tems and their regulation could, if properly applied, have
and complex and dynamic inner structure. Yet today, even dramatic effects on cell-based production of fuels, drugs,
as we know the genomic parts list of the cell, we realize that nutrients, and chemicals. Ultimately, learning how to
there is a huge gap in our mechanistic understanding of the reprogram the machinery of the cell extends far beyond
logic of how these systems work. On the one hand, we can biofermentation – it may lead to dramatic advances in
observe these beautiful systems that yield complex cellular designer cells (or cell-inspired devices) that can execute
structures, movements, and regulatory decisions; on the precision therapeutic or regenerative functions. It may
other, we have a list of molecular parts that somehow even be possible to create new cells that have hybrid
underlie these behaviors and decisions. But the middle functions and structures that are completely novel. The
ground linking how these parts fit together and work as a next few decades offer tantalizing promise of how our
system is often missing. We have a serious gap in the understanding of cell biology could be applied, and it is
mesoscopic mechanistic understanding of how microscopic time for cell biologists to start thinking creatively about
molecules work together to yield complex macroscopic how cells as machines could be harnessed.
behavior. The reviews in this issue cover several fundamental
Synthetic biology as applied to cell biology can help to issues. Thery, Schwille, and Odde discuss different ways
address this gap, because in many ways it is a philosophical that reconstitution or synthetic approaches can be used to
descendant of old-school reconstitution biochemistry. It understand the fundamental problem of self-organization
offers a unique and powerful way to construct simple, – how molecules dynamically interact with one another to
stripped down, and manipulable molecular systems, which yield higher-order structure systems such as the cytoskel-
can then be used gain insight into the fundamental rules of eton and the cell division machinery. Odde explores how
how some complex biological process takes place. But now, cellular processes can be reconstituted or visualized using
thanks to the power of genetic engineering, much of this dancers as a medium.
Maharbiz and Gartner explore the intriguing problem
Corresponding authors: Lim, W.A. (lim@cmp.ucsf.edu);
of multicellular assembly, which is intimately linked to
Marshall, W.F. (Wallace.Marshall@ucsf.edu). developmental biology. Can we understand the rules of
0962-8924/$ – see front matter ß 2012 Published by Elsevier Ltd. http://dx.doi.org/10.1016/j.tcb.2012.10.004 Trends in Cell Biology, December 2012, Vol. 22, No. 12 611
Editorial Trends in Cell Biology December 2012, Vol. 22, No. 12

how molecular systems can lead to hierarchical assembly information-processing systems. Yeates explores the way
of multicellular structures like tubes and spheres and in which known protein modules can be used to build novel
eventually into complex tissues? How might we manipu- assemblies. Reck-Petersen discusses how we can harness
late this to gain a better understanding, and is it eventu- the power of cytoskeleton-based transport machines, and
ally possible that we can build novel and arbitrary Silver reviews progress towards harnessing cellular
macroscopic structures from reengineered cells? compartmentalization for metabolic engineering.
Bathe, Yeates, Reck-Petersen, and Silver explore differ- These articles underscore the promise of synthetic cell
ent ways in which we can harness and repurpose the biology, both as an engineering discipline and as an alter-
machinery of the cell. Bathe explores the diversity of native route to understanding the secret of how cells work.
ways in which nucleic acids, with their simple Watson– But this issue is not so much a roadmap, as an invitation to
Crick base pairing can be used as a substrate to build participate. Figuring out how to reimagine the cell biology
complex assemblies, carry out catalysis, and build of the future is left as an exercise for the reader.

612
Forum: Science & Society

Special Issue – Synthetic Cell Biology

Science + dance = bodystorming


Carl Flink1 and David J. Odde2
1
Department of Theatre Arts & Dance, University of Minnesota, Minneapolis, MN 55455, USA
2
Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA

In everyday life, gravity and inertial forces often domi- where kB is Boltzmann’s constant, T is absolute tempera-
nate our movements; in the cell, these forces pale in ture, and m is the mass of the molecule. For water, the most
comparison to thermal forces. The violent, collisional abundant molecule in the cell, the root-mean-square speed
world of the cell, where water moves faster than a jet exceeds 600 m/s (1400 m.p.h.). As water molecules move at
airliner, can be difficult to imagine. To develop our these speeds, they collide with other molecules moving at
intuitive understanding of cellular and molecular pro- similar speeds, creating an extreme collisional environ-
cesses, we are exploring the concept of ‘bodystorming’, ment. It is these collisions that give rise to diffusion, the
where human ‘movers’ act as molecules that diffuse, apparently random movements of molecules. For a dance
undergo reactions, and generate/absorb forces. to convey a cellular process, such as microtubule assembly,
it is important to first appreciate the thermally driven
environment within which this process occurs. Fortunate-
Getting started ly, BLM embraces physicality and seeks safe ways in which
For the past few years, we have worked together to explore collision and contact can be incorporated into dance. To
how science can be informed through dance and vice convey this philosophy, BLM often refers to its dancers as
versa. As a starting point, we considered the concept of movers (Figure 1).
‘catastrophe’, an important feature of microtubule self-as- To simulate diffusion, we started with training exercises
sembly [1], where an abrupt, stochastic switch from an where a coin flip determined the left–right and forward–
assembling state to a disassembling state allows dynamic backward movements of the dancers. Although we have not
microtubules rapidly to explore the intracellular environ- formally checked that the movers are executing truly ran-
ment. Catastrophe is vital for proper formation of the mitotic dom walks, one gets the impression that they are (it is a
spindle, and some of the more widely used anticancer challenge to demonstrate true randomness; we routinely use
drugs (e.g., paclitaxel) specifically target microtubule self- computers to simulate diffusion even though we know that
assembly. computers are deterministic machines). Initially, movers
We came to focus on catastrophe after a chance meeting were protected by body suits, but we found that these were
sponsored by the Institute for Advanced Study (IAS) at the too cumbersome. Instead, we focused on developing trust
University of Minnesota, through which we had separately within the movers. An overarching principle is that movers
developed collaborative projects under the general theme should neither seek nor avoid collisions. In fact, we routinely
of ‘time’. One project focused on ‘Catastrophe’, a research adjust the rate of random stepping to adjust the contact level,
collaborative organized by D.O. and his brother Thomas C. equivalent to increasing/decreasing the temperature.
Odde (film studies), that explored disruption of time from Once we had movers diffusing, we added bond formation.
the multiple perspectives of art-making, biology, and engi- If the movers face each other when bonded, they can form
neering. The other project was entitled ‘Wreck’, an explo- dimers. Because the Debye screening length (the distance
ration of the depths of physical and psychological over which attractive forces are felt between macromole-
endurance in the face of impending destruction, developed cules) is typically of the order of 1 nm or less, the bonds do not
by C.F. as artistic director of the Black Label Movement form until the movers are in direct contact. When the
(BLM) Dance Company. Under the direction of Ann Waltner, bonding is front to back, self-assembled linear filaments
IAS required us to meet with other collaborative leaders, can form. We simulate bond breaking in various ways,
which prompted us to propose The Moving Cell Project, typically by throwing balls at the bonded partners. We then
where the concept of microtubule catastrophe would be enclose all the movers in a circle of movers holding hands to
explored through dance (Box 1). simulate enclosure of the diffusion and reactions within a
membrane bilayer. Working with a professional dance com-
Basic moves pany means that the movers have a high degree of physical
The cytoplasm is a violent place. The root-mean-square intelligence and can build trust quickly. This allows us to
speed of a molecule is: move rapidly with a robust range of physicality available to
rffiffiffiffiffiffiffiffiffiffiffiffi express the scientific concepts.
3kB T
vrms ¼ [1]
m Which model?
Initially, our plan was to convey scientific concepts, such as
Corresponding author: Odde, D.J. (oddex002@umn.edu). microtubule catastrophe, to a lay audience through dance.
613
Forum: Science & Society Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 1. Useful links together we have explored the extent to which dance can
Robert Hammel Vimeo Site
be used as a communication medium in science, most
http://vimeo.com/30346802 – video documenting The Moving Cell recently exemplified in a dance performance at the
Project (20 minutes). 2011 TedX-Brussels conference (http://www.ted.com/talks/
Twin Cities Public Television john_bohannon_dance_vs_powerpoint_a_modest_proposal.
http://www.mnoriginal.org/episode/hit-the-moving-cell-project/ – in- html).
terview with D.O. and C.F. and project clips (10 minutes) In either dance or computer animation, key decisions
http://www.mnoriginal.org/episode/black-label-movement-hit/ must be made about what to include in the performance/
– edited extended excerpt of BLM performing ‘HIT’ (10 minutes) animation and what not to include. Naturally, every sci-
Institute for Advanced Study entist will have his or her own unique scientific views, just
http://ias.umn.edu/2011/09/22/moving-cell-2/ – video podcast of a as choreographers will have their own unique perspectives.
joint class between BMEN 5351 Cell Engineering (D.O, instructor) The question then becomes, which model for a cellular
and DNCE 3010 Modern Dance Technique (C.F., instructor) held in
the Barker Dance Studio at the University of Minnesota (85 minutes)
process should be depicted? A model from a major textbook
or a recent review article? Or perhaps a new model that a
Chicago Tribune review of ‘HIT’
http://articles.chicagotribune.com/2011-03-11/features/ct-live-0312-
scientist is developing? In choreographing a dance, a sci-
same-planet-dance-review20110311_1_dancer-dance-center- entist is making key decisions about which model to depict.
collisions In choosing to depict a cellular process in a way that differs
Twin Cities Star Tribune review of ‘HIT’ from how another scientist would depict it, we have the
ht tp: //w ww. star tri bun e.c om/ ent e rta inm ent /st agea ndar ts/ beginning of a potentially constructive scientific discus-
139153774.html?refer=y sion. It occurred to us that dance could potentially be
BLM website informative not only to the general public, but also to
http://www.blacklabelmovement.com/ scientists at the cutting edges of scientific knowledge as
David Odde’s laboratory they articulate their hypotheses through the dances that
http://oddelab.umn.edu/ they choreograph.

Although microtubules are central to eukaryotic life Bodystorming: rapid prototyping of hypotheses
and the target of many cancer treatments, they remain One might wonder whether one can ‘prove’ anything using
virtually unknown to the general public. Using dance to dance; why not just use mathematics or computer simula-
convey basic ideas is not entirely novel; perhaps the most tions to make testable predictions from hypotheses? In
famous example is a dance performed by Stanford Univer- fact, this is a standard approach in our research and is
sity students in the 1970s entitled ‘A Protein Primer’ fast becoming standard in cell biology research generally,
(http://www.youtube.com/watch?v=u9dhO0iCLww). Since as described in this special issue on synthetic biology.
the 1970s, advances in computer animation have led to Because of the complexity of cellular processes, we gener-
beautifully rendered depictions of cellular processes, such ally need to solve equations numerically or perform Monte
as the ‘Inner Life of the Cell’ video from XVIVO Scientific Carlo simulations, depending on whether the dynamics are
Animation and BioVisions Multimedia at Harvard Univer- fundamentally deterministic or stochastic [2]. In practice,
sity (http://multimedia.mcb.harvard.edu/anim_innerlife_ doing theory via computer simulation requires a signifi-
music.html). Nevertheless, the medium of dance remains cant investment in computers and time spent program-
a vital means of communicating science, as evidenced by ming and debugging code. It is often weeks or months
the success of the ‘Dance Your Ph.D.’ contest, an annual before useful simulation output is obtained; when the
event organized by John Bohannon, a correspondent for simulations are done, we are left with the task of inter-
Science. John has joined our Moving Cell Project and preting the output. Often we need to revise the output
statistics from the simulation and try different ways of
plotting the results or making animations of the output.
Even when the model makes testable predictions that we
test experimentally and find to be correct, we often do not
intuitively understand why the model behaves in the way
that it does. This last phase of modeling, model deconstruc-
tion, is now taking us at least as much time as the model
construction phase. A modeling project can continue for
months before we start to feel confident that the model is
physically sound, makes testable predictions that are cor-
rect, and is sufficiently deconstructed that we understand
it intuitively.
Dance provides us with a useful complement to comput-
er simulation, allowing us to rapidly prototype hypotheses
without having to code and debug each one on a computer.
Using the basic moves described above, we can rapidly
TRENDS in Cell Biology explore the consequences of our hypotheses using movers.
Figure 1. Black Label Movement ‘movers’ Eddie Oroyan and Carl Flink collide in a
Because these dances are most useful at the earlier brain-
simulation of the thermally driven environment of the cell. storming stage, we refer to the process of dancing out our
614
Forum: Science & Society Trends in Cell Biology December 2012, Vol. 22, No. 12

A new way of teaching?


One exciting aspect of the collaboration is how the BLM
movers have responded positively to the science. The BLM
company members are highly trained professional dancers
with scientific backgrounds of varying extent. We have
found that abstract concepts, such as diffusion and self-
assembly, are relatively easy for a dancer to understand
when put into concrete physical terms. When we teach
science, students are normally seated and the teaching is
largely verbal and visual (e.g., lecture with whiteboard
and/or computer slides). When we teach science to dancers,
TRENDS in Cell Biology both teacher and students are on their feet and in motion.
This allows the teacher and student to bring physicality
Figure 2. Scientists and Black Label Movement ‘movers’ simulate filament self-
assembly, an example of bodystorming.
into the learning process, which we suspect is especially
effective for students whose thinking is intimately linked
to physicality (e.g., dancers and athletes). The integration
hypotheses as bodystorming (Figure 2). Often we see in the of bodystorming into our educational system seems rela-
first few moments of a bodystorming dance that the tively unexplored. We envision possible collaborations be-
experimentally observed behavior will not emerge under tween teachers in physical education, arts, and science,
the hypothesized rules. What is especially interesting is with students encouraged to learn actively through partic-
when the reasons for the discrepancy are not evident to ipation in a cellular (or physical–chemical) process. A
the scientists as the dance unfolds. At this point, we future science classroom might look more like a dance
engage in a dialog with the movers: why is the expected studio or gymnasium than a room filled with desks aimed
behavior not occurring? Unlike the computer, the movers at a whiteboard, or teaching can even move out of the
can engage in discussion with the scientists and with classroom and into the outdoors.
each other. In addition, the scientists themselves can
become movers and inhabit the cell that they are imag- Mutations and engineering
ining, which can be a profound and visceral experience. If One early challenge in the collaboration was that the
there are N people engaged in the dance, there are of the dancers would sometimes propose to move in ways that
order of N2 possible discussions that can take place after were incongruent with our current understanding of the
the dance. By contrast, in the traditional approach, the scientific concept we were trying to convey. The motivation
computer is mute and the task is left to the scientist to for these proposals was often to develop dance movements
interpret the numerical output in tabular, graphical, or with aesthetic value. Although it was tempting for the
animated output form. It is interesting to note that there scientist to dismiss these suggestions as scientifically in-
is a general trend toward agent-based simulations in cell valid, we found that it was best not to be too strict in the
biology [2]. In the case of bodystorming, the agents are artistic interpretations. First, the BLM movers are artists,
human beings who can reflect on their experience in the and for a two-way collaboration to work effectively there
simulation. needs to be room for artistic freedom, so that the movers do
To see whether BLM movers could facilitate scientific not simply feel like automatons executing an algorithm.
dialog and debate, we invited biochemists Dyche Mullins Second, the scientists should keep an open mind. Perhaps
(University of California San Francisco) and Enrique De the dancers have injected a new idea into the process.
La Cruz (Yale University) to join us in a rapid prototyp- These proposals could be regarded as ‘mutations’ and allow
ing experiment. Along with John Bohannon, who sug- the sort of ‘what if?’ experimentation that helps us to build
gested the term bodystorming during these sessions, we our physical intuition and reasoning. Can we successfully
explored a wide range of scientific questions. Perhaps predict how this mutated dance will unfold, even though
most stimulating was the question of the effect of mac- we highly doubt that a cell could ever work that way? The
romolecular crowding on association rates in vivo. Most ability to understand how a mutant dance will unfold
of our kinetic and thermodynamic measurements have connects very directly to engineering, where we attempt
been made in vitro, but we do not yet fully understand to steer the system in a direction that has a health or
how the macromolecules of the cytoplasm affect kinetics technological benefit. This could help us to address the
and thermodynamics [3]. Therefore, understanding issue of which molecules to target (e.g., via drugs) to
crowding is vital to linking our understanding of in vitro achieve a desired therapeutic outcome. Making up new
biochemistry to in vivo biochemistry and cell biology. and bizarre rules should not be discouraged.
Although we did not settle any issues definitively, we
developed a better intuitive understanding of crowding, Feeding back into dance
and the issues that were raised prompted us to go back to Another advantage of collaborating with a professional
the laboratory to set up rigorous computer simulations, dance company is that the movements developed through
work that is ongoing. In this case, bodystorming the collaboration may inspire new works of performance
allowed us to engage in rapid prototyping to identify art. In our case, the basic moves described above (e.g.,
key issues for further exploration by more traditional diffusion, reaction), led to a broader exploration of the use
approaches. of impact in creating dance works. In particular, the explo-
615
Forum: Science & Society Trends in Cell Biology December 2012, Vol. 22, No. 12

strain energy, a process that is now integrated into ‘HIT’


(Figure 3) to capture a moment of violent control between
two men. ‘HIT’ is an example of how this collaboration is
leading to serious and recognized art-making (see links to
critical reviews of ‘HIT’ in Box 1).

Future outlook
The Moving Cell Project has led us in many surprising
directions. We recently provided advice to the Citizens
League of Minnesota, a public policy group that is inter-
ested in exploring the application of bodystorming to com-
plex policy issues. In our own work, we are now planning
joint group meetings in a dance studio, which will allow
even greater integration of science and dance into our
work. Last summer we visited the Marine Biological Lab-
oratory (MBL) in Woods Hole, MA and provided a body-
storming capability to the community there through the
physiology course. Students and faculty participated in
bodystorming and rapidly prototyped hypotheses driving
their research projects during the course. In general, the
close integration of dance and science offers new opportu-
nities to learn, teach, and drive new discoveries across
disciplinary boundaries.

Acknowledgments
The Moving Cell Project is funded by the Institute for Advanced Study at
the University of Minnesota, directed by Ann Waltner. The authors thank
BLM, the Odde laboratory group, John Bohannon, Enrique De La Cruz,
and Dyche Mullins for their participation and valuable input into the
collaborative. We are grateful to have worked with the late Robert
Hammel and to Perimeter Productions for their video documentation of
our work. We also thank the MBL, especially Dyche Mullins, Diana
Kenney, Bill Reznikoff, and Gary Borisy, for supporting a BLM residency
TRENDS in Cell Biology
in Woods Hole. Finally, we thank Ann Waltner for her commitment to
interdisciplinary research and for catalyzing our collaboration.
Figure 3. Patrick Jeffrey and Eddie Oroyan create a curling microtubule
protofilament in the new Black Label Movement dance ‘HIT’. References
1 Mitchison, T.J. and Kirschner, M.W. (1984) Dynamic instability of
microtubule growth. Nature 312, 237–242
2 Mogilner, A. and Odde, D. (2011) Modeling cellular processes in 3D.
ration of the technique of impact required to manifest the Trends Cell Biol. 21, 692–700
violent interior of the cell suggested to us that there was a 3 Zhou, H.X. et al. (2008) Macromolecular crowding and confinement:
choreographic application of this vocabulary useful for biochemical, biophysical, and potential physiological consequences.
Annu. Rev. Biophys. 37, 375–397
examining human relationships. In a quartet entitled
‘HIT’, Carl removed the prohibition of violent physicality
in healthy human relationships to examine a dysfunctional
0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved.
community of people. When microtubules disassemble, http://dx.doi.org/10.1016/j.tcb.2012.10.005 Trends in Cell Biology, December 2012,
their protofilaments curl outward releasing mechanical Vol. 22, No. 12

616
Opinion

Special Issue – Synthetic Cell Biology

Synthetic multicellularity
Michel M. Maharbiz
Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA 94720, USA

The ability to synthesize biological constructs on the Multicellularity and emergent behavior
scale of the organisms we observe unaided is probably At its most fundamental level, multicellularity arises when
one of the more outlandish, yet recurring, dreams cells come together and find means to couple their internal
humans have had since they began to modify genes. states in such a way that the connections result in emer-
This review brings together recent developments in gent behavior –generally with improved fitness for a set of
synthetic biology, cell and developmental biology, com- problems – that arises from the collective of cells. This is a
putation, and technological development to provide somewhat weak definition of multicellularity, because it
context and direction for the engineering of rudimenta- allows for numerous collective ensembles (e.g., bacterial
ry, autonomous multicellular ensembles. biofilms or intestinal microflora) that are themselves not
considered organisms, but for the engineer it allows a
What is multicellularity? certain flexibility when approaching the problem. There
Macroscopic organisms generally comprise numerous cells, is now a well-developed literature in applied mathematics,
usually from a common genetic parent, differentiated computer science, and the biology of social insects that
through environmentally sensitive genetic programs. That deals with the analysis and design of similar multi-agent
is, they are multicellular. But what is multicellularity? systems. It has long been known, for instance, that the
How could we approach the multifaceted challenge of complex functions carried out by social insect communities
engineering autonomous multicellular ensembles? What (foraging and detection of environmental signals, construc-
is missing today from the synthetic biological repertoire? tion of complex structures [12], and even specification of
Are there approaches that veer away from the paths taken insect phenotype [13]) emerge from a beautiful interplay
by nature? Lastly, is there a good reason to do this at all? between environmental factors, ‘simple’ behaviors encoded
A discussion of the definition of multicellularity – par- in the individual insects, and the exchange of information
ticularly when appended to the word organism or when between individuals. This cooperation results in a type of
discussed in the context of complex bacterial communities ‘swarm intelligence’ that is robust to insult and environ-
– is beyond the scope of this short paper and excellent mental change and can carry out optimization tasks
reviews already exist [1–4]. Needless to say, any student [14–16]. Numerous engineering efforts, notably in computer
contemplating these issues should take a developmental science and applied mathematics, have taken inspiration
biology course to lose themselves in the myriad routes to from these phenomena to develop software packages for
multicellularity in the natural record; classic textbooks routing and optimization of many real-world problems
include Wolpert [5] and Gilbert [6]. The seminal synthesis [17], to explore the theoretical aspects of emergent systems
by D.W. Thomson, On Growth and Form [7], and the many as computational engines [18,19], and to build computation
highly readable monographs by J.T. Bonner [8–10], al- into physical structures [20,21] (an active endeavor that is
though in many ways dated, issue forth inspiration from fundamentally similar to the topic that concerns us here).
almost any page. Lastly, the elegant little book Vehicles: That many of these ‘social’ behaviors bear striking resem-
Experiments in Synthetic Psychology [11] provides a con- blance to functions and properties of multicellular organ-
ceptual roadmap toward engineered complexity that is isms motivates a closer inspection. The point here is that
well worth the read. before we delve into how multicellular ensembles might be
I will avoid in this paper the notion of coopting existing constructed, we should spend some time learning the rich
mammalian cell lines to form synthetic multicellular computational background that now exists for designing
ensembles; that is primarily the province of tissue engi- and analyzing systems with emergent properties. In this
neering. This is not to say that is not a useful endeavor, of regard, the Dictyostelium discoideum community has per-
course; but these lines already contain within them com- haps some of the best demonstrations of the remarkably
plex – and poorly understood – systems for producing complex, yet computationally tractable, emergent behaviors
specific metazoan forms. They harbor millions of years of arising from cells executing relatively simple processes.
evolutionary baggage, so to speak. To form a working From Bonner [10,22] through Nakagaki’s maze-finding
definition of multicellularity (to which we can later add slime molds [23] to the so-called Tokyo subway experiment
the label synthetic), we would like to start much closer to [24], Dictyostelium has become one of the models for study-
the beginning. ing emergence and multicellularity [25]. The broader point
is that, as with almost all engineering efforts initially
Corresponding author: Maharbiz, M.M. (maharbiz@eecs.berkeley.edu).
inspired by nature, once we computationally understand
Keywords: multicellular; abiotic; interfaces; development; emergent;
cell–cell adhesion. emergence, we are likely to find solutions not available from
0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.09.002 Trends in Cell Biology, December 2012, Vol. 22, No. 12 617
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

the observable natural catalog (e.g., consider the robot Rhex (e.g., cadherins) appear to have arisen before multicellu-
[26] or synthetic nacre [27], both inspired by but very larity, were involved in environmental and prey–predator
different from natural systems). Armed with this conviction, detection, and were coopted during the transition[39].
we can now ask what ‘parts’ are needed to synthesize Numerous studies [1,31,36,40–44] point to the handful
collective behavior in cells. of basic ‘enabling technologies’ we must consider engineer-
ing: the excretion of an extracellular matrix (ECM), the
The transition to multicellularity presence of cytoplasmic bridges, cell–cell adhesion, and
Where might we turn for a first look at what genetic and molecular differentiation (or inherited functional speciali-
epigenetic modules are needed to engineer multicellulari- zation). A fifth observation, likely to be emergent, is that
ty? The biomolecular complexity of the classic model choanoflagellate colonies appear to form not due to aggre-
systems in developmental biology (e.g., Drosophila mela- gation, but due to non-separation after division (with the
nogaster, Caenorhabditis elegans) can be daunting [5]; even concomitant production of a matrix and cell junctions). Let
Hydra [28] and the simple flatworm [29] are already far us consider these enabling technologies one at a time.
down the path into multicellularity. Luckily, the past A recent review [45] provides an overview of ECM
decade has seen an explosion of work on species bordering domains as well as the known phylogeny in the catalog;
the transition from the unicellular to multicellular lifestyle Özbek et al. provides an evolutionary viewpoint [46]. Choa-
[30–34]. It is here that, stripped of all but the barest noflagellates can secrete multiple types of ECM [36], al-
essentials, we may begin to identify the specific compo- though apparently none with the complexity of true
nents we must engineer. Sometime around 600 million metazoan ECM [45]. The type prevalent in colonial phe-
years ago, colonial and truly multicellular ensembles notypes of Salpingoeca rosetta appears ‘amorphous, loose
began to emerge from unicellular flagellates [34]. Today, and space-fitting’, whereas unicellular phenotypes of the
some of the closest relatives to metazoans are the choano- same species are capable of producing denser, more pre-
flagellates (Figure 1) – a collection of small (3–10 mm), cisely shaped ECM as well (the so-called theca ‘goblet’) [36].
flagellated eukaryotes, some of which are unicellular, some Cells of the multicellular alga Volvox embed themselves in
multicellular, and some that can adopt either phenotype a complex ECM and convert part of their cell walls to ECM;
[35]. What is so intriguing about these creatures is not only the massive amount of ECM secreted by these cells allows
that certain species exhibit both unicellular and multicel- for very large colonies (in proportion to their cell number)
lular phenotypes – which makes it possible to observe in [30]. In more complex eukaryotes, the ECM constitutes not
detail the genetic and structural changes that allow multi- only a mechanical support layer, but is a fundamental
cellularity [36,37] – but that spectra of closely related communication channel between cells, allowing chemical
species exist that together capture the heritable transition and mechanical signals to be exchanged by cells attached
to multicellular behavior. Moreover, genomic and proteo- to it [46,47].The fact that ECM is usually laminar with
mic analysis has shown that, remarkably, components of respect to some axis of the ensemble provides a ready
many of the genetic systems once thought specific to reference to establish polarity [6,47]. Bacteria are known
metazoans and bilaterians (cadherins, receptor tyrosine to also secrete some sort of matrix, particularly when
kinases [RTKs], bilaterian miRNAs, Piwi-interacting forming biofilms, but they seem to lack the same richness
RNAs [piRNAs]) and thought to be crucial in the develop- of communication modes as eukaryotes capable of more
ment and maintenance of complex forms are present in complex multicellularity despite the fact that they can
choanoflagellates [30,32,33,38]. Some of these pathways form differentiated spatial structures with different struc-
tural ECM for different functional components [45].
This, of course, brings us to cell–cell adhesion mole-
cules, principally the cadherins. Choanoflagellates pos-
10 um By DHZanee sess cadherins [39] and integrin domains (but no true
N~230 integrins) [45], putatively allowing for cell–cell and cell–
ECM adhesion. This enables the coupling of external
mechanical events and information to internal changes
in cell state. This is one of the functions most sorely lacking
in the bacterial synthetic biology repertoire. Although
ongoing work has demonstrated that the MscL family of
channels in bacteria are sensitive to membrane strain and
affect transmembrane potential [48,49], there are, at
present, no well-characterized modules for coupling strain
or adhesion to genetic expression. Cadherins are crucial
enablers not only of cell polarity, but also of emergent and
coordinated relative motion and reorganization within a
cell ensemble, a topic of recent interest [50–53]. Cyto-
plasmic bridges, which in choanoflagellates seem to arise
from incomplete cytokinesis [36], also seem to play a role,
TRENDS in Cell Biology both in maintaining physical connections among colonial
Figure 1. A spherical colony of Sphaeroeca choanoflagellates. Reproduced, with members and in the constrained exchange of signaling
permission, from http://commons.wikimedia.org/wiki/File:Sphaeroeca-colony.jpg. molecules.
618
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

Lastly, we come to the topic of differentiation (or heri- population. Early seminal efforts demonstrated that
table cell state). In eukaryotic systems, this is a vast field of groups of ‘receiver’ cells sensitive to the presence of AHL
inquiry, well outside the scope of this review. Within could be made to respond in a tunable fashion to the
microbial synthetic biology, switches and feedback loops presence of ‘sender’ cells that produced diffusible AHL
that allow an inducible, switchable state and that can pass [74]. This work was subsequently extended to demonstrate
this state to daughter cells have been demonstrated; I will consensus cooperation in microbial communities [75], syn-
return to this below in the context of circuits and systems thetic predator–prey systems [76], and intercellular com-
already developed by synthetic biologists. It is worth not- munication enabling edge detection across light/dark areas
ing that, in flagellates, there is an inherent trade-off [77], among others. A series of papers from the Hasty
between motility and division, because both processes laboratory have since demonstrated the use of AHL-cou-
make use of microtubule-organizing centers (MTOCs). pled oscillators for entraining and synchronizing popula-
King [34], Buss [54], and Michod [55] have pointed out tions [66,78,79]. More recently, AHL-based signaling was
that differentiation between flagellated cells on the surface used to provide chemical ‘wires’ between cell colonies, each
of a multicell colony and non-flagellated cells in its center carrying out distinct computations to enable more complex
arose as a solution to the MTOC trade-off, because colonies functions than those possible within single cells [80]. Much
containing flagellated and non-flagellated cells would be more remains to be done in this area. True symmetry-
under strong environmental pressure to expose as many breaking systems such as the classic Turing [81] and
motile cells on the surface of the colony as possible. This, of Meinhardt–Gierer [82] diffusible signal pattern generators
course, is what one sees in many species of colonial fla- have not yet been robustly demonstrated, despite recent
gellates. More importantly, it points to the idea that di- experimental [Ting, L. et al. (2008) Pattern formation in a
rected evolution [56] in the presence of some of the basic synthetic multicellular system. 2008 APS March Meeting.
components described above might allow us to recapitulate 53 (http://meetings.aps.org/link/BAPS.2008.MAR.J17.7)]
something as powerful as gastrulation [5,6] and spatial and theoretical results [83]. Despite the early work on
organization [55]. In fact, almost 30 years ago, Edelman the subject, little has been done to create systems in
suggested in his ‘regulator hypothesis’ that selection acting bacteria with multiple, simple, orthogonal cell–cell com-
on just cell–cell adhesion genes and differentiation genes munication motifs operating to produce complex emergent
could account for stable and varied body plans ‘within behavior in communities of cells. As cited above, there is a
relatively short periods of evolutionary time’ [57]. wide literature in computer science and applied mathe-
matics concerning cellular automata and emergent sys-
Moving forward – synthetic engineering beyond the tems, many of which have been shown to be capable of
genetic circuit propulsion, reproduction and size control and to be Turing
Efforts over the past decade have yielded a catalog of basic complete [19,84–86]. As additional orthogonal communi-
cell state circuit motifs [58,59] that include bistable cation channels become available (see below), this type of
switches and memory elements [60–63], oscillators [64– spatial programming should become accessible. Moreover,
66], simple logic gates [67,68], and amplifiers [69,70]. The emergent behavior need not arise exclusively via chemical
complexity of genetic circuitry will no doubt increase in the dynamics. A recent example elegantly demonstrates how
coming years, enabling more sophisticated programming of motility and density can be coupled via a synthetic AHL-
cell state and, thus, at least some degree of synthetic mediated circuit to produce stripe formation in Escherichia
differentiation in microbial models. What else then should coli [87]. Decades of theoretical treatment of the mechani-
we focus on in the quest for multicellularity? cal underpinnings of metazoan morphogenesis have pro-
vided several testable models of how mechanical
Local relationships lead to global organization communication via cell–cell and cell–ECM coupling would
Numerous synthetic intercellular signaling systems have result in complex shape changes and modulation of gene
been demonstrated in the past few years [59,71], with expression [51,88–91]. Emergent pattern formation from
quorum sensing-derived circuits gaining widespread use simple ECM-mediated processes has been documented. An
[72]. Quorum sensing in bacteria relies on the well-known old example [92] demonstrates how fibroblast traction on
acyl homoserine lactone (AHL) family of freely diffusible collagen matrix itself aligns the underlying collagen fibers
and membrane-permeable compounds. The ability to both into tracks toward the cells, driving further aggregation.
induce the production of AHLs via the LuxI gene product This simple mechanical instability was capable of driving
homologs and detect AHL concentration via AHL–LuxR cell density-dependent Turing-like pattern formation. This
complexes acting on inducible promoters allows for the result reflects a still rather unexplored facet of cellular
introduction of diffusible intercellular signaling modules; pattern formation: the interplay between cell processes
an early seminal result was the coupling of an AHL sig- and physicochemical forces in the environment, which
naling circuit to a cell ‘killer’ gene to synthetically regulate together drive complex emergent behavior [51].
bacterial culture growth curves [73]. The number of truly
orthogonal AHL communication channels is somewhat Missing links
limited, but that catalog may grow. One of the outstanding It is believed that several of the genetic and epigenetic
challenges in the field is the demonstration of a robust processes that regulate multicellularity in eukaryotes are
framework for coupling intercellular signaling and circuits missing in bacteria; these include cadherins, tyrosine
encoding cell state to generate programmable patterns in kinases, and complex ECM production (see above). Given
cell state across an initially homogeneous multicellular that bacteria as a whole do exhibit some matrix production,
619
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

Strain
Matrix Cell-cell contact sensor
secreon communicaon Cell polarity

Bacterium
Cell-cell adhesion Le-right symmetry
+ aggregaon
or division

In-out symmetry ‘Fast’ non-transcripon


networks drive molity
TRENDS in Cell Biology

Figure 2. What motifs are necessary for rudimentary synthetic multicellularity? Shown here are distinct ‘enabling technologies’ strung into a putative roadmap to colonial
ensembles. The various ‘missing links’ are discussed in the text.

cell–cell contact-mediated signal transduction [93–95], Here arises the issue of assembly. The classical position
strain sensing [49], rapid membrane potential depolariza- of the genetic engineer is to look for the gene or network of
tion events [96], and even direct electron transfer channels genes, however mythical, that will generate a desired
[97–100], it seems likely that analogs of eukaryotic pro- phenotype when introduced into a cell. This is, in essence,
cesses can be synthetically engineered into or mined from a self-assembly approach: we genetically engineer desired
bacteria. Three specific missing components would be in- behaviors in cells and then allow them to grow and form
valuable to the pursuit of multicellularity and are likely to multicellular systems. A crucial area of work in the context
be obtainable in the near term (Figure 2): a stress/strain of engineering multicellularity via self-assembly is the so-
sensor coupling either membrane stress or stress on an called ‘decomposition problem’ [104–106]. That is, given a
extracellular process (i.e., pili, flagella, or cilia) to gene final form and a palette of possible operations or gene
regulation; a cell–cell contact-mediated channel for either circuits, can I compute the sequence of such operations
cytoplasmic contact or signal exchange; and a system for that form the final form? Can I determine the minimal set
inducing and maintaining cell polarity in bacteria relative of ‘functions’ (that is, cellular behaviors such as adhesion,
to a matrix plane. Coupled with synthetic cell–cell adhe- force production, migration, and signal production) re-
sion, a stress/strain sensor might allow bacteria to ap- quired to arrive at a certain multicellular form?
proach the level of cytomechanical control multicellular Alternatively, we can impose external constraints on
eukaryotes employ for cell sorting and multicell motion. On the cells that direct the formation of multicellularity. This
the issue of cytoplasmic bridging, some reports appear to is an example of constrained assembly, common to classic,
indicate the presence of cytoplasmic exchange – including industrial age manufacturing wherein form or function is
plasmid transfer – across ‘nanotubes’ seen between Bacil- engineered via a sequence of top-down steps (as in the
lus subtilis, Staphylococcus aureus, and E. coli [101]. assembly of a mechanical watch). Biology, in fact, makes
Again, choanoflagellates may be invaluable, because it is extensive use of constrained assembly. The Drosophila egg
thought that metazoan adhesion proteins may be derived chamber, for example, is in part an apparatus for symme-
from proteins used by heterotrophic flagellates to recognize try breaking and driving the constrained assembly of the
and bind bacterial prey [39]. A fourth, and far more diffi- early fly embryo. Developmental biology courses usually
cult, need is for modular RNA- or protein-based systems focus on the beautiful sequence of events that unfold in the
capable of switching states much more rapidly than fertilized embryo starting with the Bicoid gradient (speci-
allowed via transcriptional regulation (i.e., for fast fying, for example, segments and assembling organs) and
responses to the environment) [102,103]. Although compo- ending with an adult fly. What is relevant to this discussion
nents do exist, they are not yet sufficiently well understood, is that the initial anterior–posterior and dorsal–ventral
modular, or orthogonal to enable the flexible engineering axes of polarity are determined by sequestered mRNA laid
of, for example, sensor-to-flagellar control modules in bac- down in unfertilized eggs by maternal nurse cells in the egg
teria (i.e., fast synthetic navigation systems). chamber; thus, symmetry breaking is accomplished in the
embryo by the mother [5,107]. Many examples of these
Putting it all together maternal effects can been documented phenomenological-
Paralleling the increasing complexity of the constructs in ly, but detailed mechanistic understanding appears to be
Valentino Braitenberg’s seminal book Vehicles: Experi- emerging [108].
ments in Synthetic Psychology, it is tempting to entertain When considering synthetic multicellular engineering,
the notion of a series of grand challenges. Who can be it is likely that abiotic ‘maternal effect’ technologies – that
the first to demonstrate a synthetic, motile colony? With is, machines that break symmetry for the developing sys-
programmable sensor-to-motility response? With pro- tem or otherwise impose boundary conditions on the mul-
grammable form? With cell lineage differentiation? With ticellular ensemble to guide development – could play a
reproduction? With memory? role. A vast repertoire of non-biological devices can be
620
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

brought to bear. Whether or not natural colonial and First, a grand challenge around true, synthetic multi-
multicellular systems arise through self-assembly, human cellularity would provide an immediate test bed for the
engineers need not build them that way. Modern advances development of many other platform technologies crucial
have resulted in a wide variety of devices for specifying the to synthetic biology. With properly formulated systems
chemical, mechanical, optical, and electrical milieu both goals, we could outline specifications for the various mod-
intra- and extracellularly. An exhaustive review is outside ules needed and present multicellularity as a competitive
the scope of this piece, but some examples are provided challenge wherein groups arrive at unique solutions. Con-
below. Given the medical utility of many of these methods sider some medium-term applications. Multicellular, mo-
(including drug delivery, tissue engineering, and clinically tile colonies with silicon payloads could be deployed in
relevant sensing [109]), they continue to advance rapidly. rivers and lakes as environmental health monitoring
Simply put, having established an engineering goal (e.g., a devices, capable of interacting with the aquatic biology
1-mm diameter, 1024-cell motile, flagellated colony that at various depths and communicating with man-made
swims up a specified concentration gradient and lyses at a devices. Organized films of swarmer cells could clean
programmed threshold), we should explore technologies surfaces: ingesting small particles, digesting larger ones
beyond those available in natural systems to engineer it. and carrying indigestible particulates (including excess
Hybrid abiotic/biotic systems are likely to be useful even cell mass) to set locations. Cell collectives capable of taxiing
beyond assembly tasks. Consider again our flagellar colo- into and around each other in weaves (and then depositing
ny. Why not assemble such a colony on abiotic parts matrix) could assemble and repair simple textiles on de-
properly functionalized with adhesion proteins? A simple mand in remote places. Sophisticated biofilms could detect
computational engine fabricated in a modern semiconduc- strain non-linearities indicative of the cracking of an un-
tor process can be as small as 100 mm on a side. Chemical derlying stratum and produce binding matrix.
input/output [110–114], electrochemical sensors for DNA Second, this endeavor may drive the development of
hybridization [115], nanoscale and integrated light detec- toolsets for use in non-canonical organisms like the choa-
tors and LEDs [116], polymer strain gauges [117,118], noflagellates (among others), something of great value to
surface energy switching [119], and even nanowire trans- fundamental molecular and comparative biology.
membrane recordings [120] can be integrated into very Lastly, and most importantly, the demonstration of
small systems. From my work in integrating abiotic com- synthetic multicellularity with a complexity eventually
ponents with live insects for flight control [121], it has rivaling that of metazoans or terrestrial plants must surely
become clear that hybrid systems can use the best of both be one of the long-term goals of synthetic biology. This
worlds. Biological systems did not evolve radios or 22-nm arises not from an argument of biomimicry – which, fol-
transistors; man-made communication and computational lowed blindly, leads us down fruitless technological paths –
systems have reached a staggering degree of sophistication but because we will never truly understand the limits of
in incredibly small, low-power packages (e.g., the off-the- developmental regulation, stability, and plasticity until we
shelf ATtiny10 from Atmel is a 12-MHz, 8-bit microcon- have recapitulated developmental processes on our syn-
troller with a 1024-byte flash memory in a package 2 mm thetic platforms. There are likely to be stable areas of the
square; unpackaged, it is about half this size). The power metazoan parameter space that evolution has either se-
requirements for the 100-mm computational engine could lected out or not explored and that possess technological
be <10 mW on average and powered entirely from a tiny value for our society. Just as the 20th century made it
solar cell on-chip. Conversely, man-made energy sources, obvious that humans could arrive at molecules and chem-
motility systems, and material properties still pale in istries not found in nature starting from the same constit-
comparison with the natural arsenal. It is not feasible with uent atoms, it is likely we will find multicellular solutions
modern abiotic technology to make an autonomous 1-mm with technological value not arrived at via natural selec-
swimming robot capable of locomoting indefinitely; we tion. That this has profound ethical and societal conse-
cannot build good synthetic chemotrophs, our actuator quences cannot be overstated.
technologies are poorly suited to that scale, and communi-
cation via radio or optics is not easy in real underwater Concluding remarks
environments. There are other, ancillary advantages of There is additionally – and perhaps controversially – an
hybrid approaches. Abiotic parts would not be subject to ultimately ecological rationale for this vision. For the most
genetic changes or selection pressure; hybrid systems part, the technological base created by the industrial revo-
would not be able to reproduce, alleviating some safety lution communicates poorly with the underlying organic
concerns, and have finite lifetimes. technology of the planet. The rapid explosion of man-made,
acellular, resource-consuming, and waste-producing con-
Why do this? structs is in large part responsible for the ecological and
Lastly is the issue of motivation: is this something worth climactic mess we are in. Mindful of the vast ethical and
doing? Much is still to be worked out to achieve scalable societal questions raised, it is worth considering a future
synthetic control circuits [71], not to mention fundamental wherein our homes, our factories, and our consumer gad-
technology development [122,123]. It is not clear what we gets can ‘understand’ the language of the organic systems
do with the simple ‘colonial’ constructs I have sketched around them and form part of a related or hybrid frame-
above and it is a long way to anything resembling a work of information and material exchange. This notion –
metazoan. The attraction of the problem, however, is that our societal artifacts should be mindful of their natu-
threefold. ral surroundings – has long and deep roots in many
621
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

cultures and has modern reflections in, for example, the 33 Nichols, S.A. et al. (2006) Early evolution of animal cell signaling and
adhesion genes. Proc. Natl. Acad. Sci. U.S.A. 103, 12451–12456
natural building movements. It is my contention that the
34 King, N. (2004) The unicellular ancestry of animal development. Dev.
development of synthetic multicellular – and likely hybrid Cell 7, 313–325
– systems is a step down this transformative road. 35 King, N. (2005) Choanoflagellates. Curr. Biol. 15, 113–114
36 Dayel, M.J. et al. (2011) Cell differentiation and morphogenesis in the
References colony-forming choanoflagellate Salpingoeca rosetta. Dev. Biol. 357,
1 Nedelcu, A.M. (2001) Evolution of Multicellularity, John Wiley & Sons 73–82
2 Webb, J.S. et al. (2003) Bacterial biofilms: prokaryotic adventures in 37 Fairclough, S.R. et al. (2010) Multicellular development in a
multicellularity. Curr. Opin. Microbiol. 6, 578–585 choanoflagellate. Curr. Biol. 20, 875–876
3 Monds, R.D. and O’Toole, G.A. (2009) The developmental model of 38 Grimson, A. et al. (2008) Early origins and evolution of microRNAs
microbial biofilms: ten years of a paradigm up for review. Trends and Piwi-interacting RNAs in animals. Nature 455, 1193–1198
Microbiol. 17, 73–87 39 Abedin, M. and King, N. (2008) The premetazoan ancestry of
4 McShea, D.W. (2002) A complexity drain on cells in the evolution of cadherins. Science 319, 946–948
multicellularity. Evolution 56, 441–452 40 Kirk, D.L. (2005) A twelve-step program for evolving multicellularity
5 Wolpert, L. and Tickle, C. (2011) Principles of Development, (4th edn), and a division of labor. Bioessays 27, 299–310
Oxford University Press 41 Kirk, D.L. (1999) Evolution of multicellularity in the volvocine algae.
6 Gilbert, S.F. (2010) Developmental Biology, (9th edn), Sinauer Curr. Opin. Plant Biol. 2, 496–501
Associates 42 Sachs, J.L. (2008) Resolving the first steps to multicellularity. Trends
7 Thompson, D.A.W. (1942) On Growth and Form, The University Press Ecol. Evol. 23, 245–248
8 Bonner, J.T. (2001) First Signals: The Evolution of Multicellular 43 Grosberg, R.K. and Strathmann, R.R. (2007) The evolution of
Development, Princeton University Press multicellularity: a minor major transition? Annu. Rev. Ecol. Evol.
9 Bonner, J.T. (1955) Cells and Societies, Princeton University Press Syst. 38, 621–654
10 Bonner, J.T. (1998) The origins of multicellularity. Integr. Biol. 1, 27– 44 Michod, R.E. (2007) Evolution of individuality during the transition
36 from unicellular to multicellular life. Proc. Natl. Acad. Sci. U.S.A. 104,
11 Braitenberg, V. (1986) Vehicles: Experiments in Synthetic Psychology, 8613–8614
MIT Press 45 Hynes, R.O. (2012) The evolution of metazoan extracellular matrix. J.
12 Hölldobler, B. and Wilson, E.O. (1994) Journey to the Ants: A Story of Cell Biol. 196, 671–679
Scientific Exploration, Belknap Press 46 Özbek, S. et al. (2010) The evolution of extracellular matrix. Mol. Biol.
13 Patalano, S. et al. (2012) Shifting behaviour: epigenetic Cell 21, 4300–4305
reprogramming in eusocial insects. Curr. Opin. Cell Biol. 24, 367– 47 Hynes, R.O. (2009) The extracellular matrix: not just pretty fibrils.
373 Science 326, 1216–1219
14 Dussutour, A. et al. (2005) Amplification of context: the ants. Proc. R. 48 Booth, I.R. and Blount, P. (2012) The MscS and MscL families of
Soc. B: Biol. Sci. 272, 705–714 mechanosensitive channels act as microbial emergency release
15 Reznikova, Z. (2008) Experimental paradigms for studying cognition valves. J. Bacteriol. 194, 4802–4809
and communication in ants (Hymenoptera: Formicidae). Myrmecol. 49 Booth, I.R. et al. (2011) Sensing bilayer tension: bacterial
News 11, 201–214 mechanosensitive channels and their gating mechanisms. Biochem.
16 Panteleeva, S. et al. (2010) Using ideas of Kolmogorov complexity for Soc. Trans. 39, 733–740
studying animal behavioural patterns. In Proceedings of Measuring 50 Borghi, N. and James Nelson, W. (2009) Intercellular adhesion in
Behavior 2010 (Eindhoven, The Netherlands, August 24-27, 2010) morphogenesis: molecular and biophysical considerations. Curr. Top.
(Spink, A.J. and et, al., eds), pp. 174–176, Noldus Information Dev. Biol. 89, 1–32
Technology 51 Newman, S. et al. (2006) Before programs: the physical origination of
17 Michael, L. (2005) Ant-based computing. Artif. Life 15, 337–349 multicellular forms. Int. J. Dev. Biol. 50, 289–299
18 Schiff, J.L. (2011) Cellular Automata: A Discrete View of the World, 52 Brien, L.E.O. et al. (2002) Building epithelial architecture: insights
John Wiley & Sons from three-dimensional culture models. Nat. Rev. Mol. Cell Biol. 3, 1–
19 Wolfram, S. (2002) A New Kind of Science, Wolfram Media 7
20 Griffith, S. et al. (2005) Robotics: self-replication from random parts. 53 Borghi, N. et al. (2010) Regulation of cell motile behavior by crosstalk
Nature 437, 636 between cadherin- and integrin-mediated adhesions. Proc. Natl.
21 Pfeifer, R. et al. (2007) Self-organization, embodiment, and Acad. Sci. U.S.A. 107, 13324–13329
biologically inspired robotics. Science 318, 1088–1093 54 Buss, L.W. (1987) The Evolution of Individuality, Princeton
22 Bonner, J.T. (2010) Brainless behavior: a myxomycete chooses a University Press
balanced diet. Proc. Natl. Acad. Sci. U.S.A. 107, 5267–5268 55 Solari, C.A. et al. (2006) A hydrodynamics approach to the evolution of
23 Nakagaki, T. et al. (2000) Intelligence: maze-solving by an amoeboid multicellularity: flagellar motility and germ-soma differentiation in
organism. Nature 407, 470 volvocalean green algae. Am. Nat. 167, 537–554
24 Tero, A. et al. (2010) Rules for biologically inspired adaptive network 56 Cobb, R.E. et al. (2012) Directed evolution: an evolving and enabling
design. Science 327, 439–442 synthetic biology tool. Curr. Opin. Chem. Biol. 16, 285–291
25 Devreotes, P. (1989) Dictyostelium discoideum: a model system for 57 Edelman, G.M. (1984) Cell adhesion and morphogenesis: the
cell-cell interactions in development. Science 245, 1054–1058 regulator hypothesis. Proc. Natl. Acad. Sci. U.S.A. 81, 1460–1464
26 Saranli, U. et al. (2001) RHex: a simple and highly mobile hexapod 58 Wolf, D.M. and Arkin, A.P. (2003) Motifs, modules and games in
robot. Int. J. Robot Res. 20, 616–631 bacteria. Curr. Opin. Microbiol. 6, 125–134
27 Tang, Z. et al. (2003) Nanostructured artificial nacre. Nat. Mater. 2, 59 Boyle, P.M. and Silver, P.A. (2009) Harnessing nature’s toolbox:
413–418 regulatory elements for synthetic biology. J. R. Soc. Interface 6,
28 Steele, R.E. (2002) Developmental signaling in Hydra: what does it S535–S546
take to build a ‘‘simple’’ animal? Dev. Biol. 248, 199–219 60 Palani, S. and Sarkar, C.A. (2011) Synthetic conversion of a graded
29 Gentile, L. et al. (2010) The planarian flatworm: an in vivo model for receptor signal into a tunable, reversible switch. Mol. Syst. Biol. 7,
stem cell biology and nervous system regeneration. Dis. Model. Mech. 480
4, 12–19 61 Atkinson, M.R. et al. (2003) Development of genetic circuitry
30 Abedin, M. and King, N. (2010) Diverse evolutionary paths to cell exhibiting toggle switch or oscillatory behavior in Escherichia coli.
adhesion. Trends Cell Biol. 20, 734–742 Cell 113, 597–607
31 Ruiz-Trillo, I. et al. (2007) The origins of multicellularity: a multi- 62 Chang, D-E. et al. (2010) Building biological memory by linking
taxon genome initiative. Trends Genet. 23, 113–118 positive feedback loops. Proc. Natl. Acad. Sci. U.S.A. 107, 175–180
32 Young, S.L. et al. (2011) Premetazoan ancestry of the Myc – Max 63 Sallee, N.A. et al. (2007) Engineering modular protein interaction
network. Mol. Biol. Evol. 28, 2961–2971 switches by sequence overlap. J. Am. Chem. Soc. 129, 4606–4611

622
Opinion Trends in Cell Biology December 2012, Vol. 22, No. 12

64 Fung, E. et al. (2005) A synthetic gene-metabolic oscillator. Nature 96 Kralj, J.M. et al. (2011) Electrical spiking in Escherichia coli probed
435, 118–122 with a fluorescent voltage-indicating protein. Science 333, 345–348
65 Stricker, J. et al. (2008) A fast, robust and tunable synthetic gene 97 Jensen, H.M. et al. (2010) Engineering of a synthetic electron conduit
oscillator. Nature 456, 516–519 in living cells. Proc. Natl. Acad. Sci. U.S.A. 107, 19213–19218
66 Mondragon-Palomino, O. et al. (2011) Entrainment of a population of 98 Ross, D.E. et al. (2011) Towards electrosynthesis in Shewanella:
synthetic genetic oscillators. Science 333, 1315–1319 energetics of reversing the Mtr pathway for reductive metabolism.
67 Anderson, J.C. et al. (2007) Environmental signal integration by a PLoS ONE 6, e16649
modular AND gate. Mol. Syst. Biol. 3, 133 99 Shi, L. et al. (2007) Respiration of metal (hydr)oxides by Shewanella
68 Wang, B. et al. (2011) Engineering modular and orthogonal genetic and Geobacter: a key role for multihaem c-type cytochromes. Mol.
logic gates for robust digital-like synthetic biology. Nat. Commun. 2, Microbiol. 65, 12–20
508 100 Baron, D. et al. (2009) Electrochemical measurement of electron
69 Nistala, G. et al. (2010) A modular positive feedback-based gene transfer kinetics by Shewanella oneidensis MR-1. J. Biol. Chem.
amplifier. J. Biol. Eng. 4, 1–8 284, 28865–28873
70 Nandagopal, N. and Elowitz, M.B. (2011) Synthetic biology: 101 Dubey, G.P. and Ben-yehuda, S. (2011) Intercellular nanotubes
integrated gene circuits. Science 333, 1244–1248 mediate bacterial communication. Cell 144, 590–600
71 Purnick, P.E.M. and Weiss, R. (2009) The second wave of synthetic 102 Lucks, J.B. et al. (2011) Versatile RNA-sensing transcriptional
biology: from modules to systems. Nat. Rev. Mol. Cell Biol. 10, 410–422 regulators for engineering genetic networks. Proc. Natl. Acad. Sci.
72 Jayaraman, A. and Wood, T.K. (2008) Bacterial quorum sensing: U.S.A. 108, 8617–8622
signals, circuits, and implications for biofilms and disease. Annu. 103 Qi, L. et al. (2012) Engineering naturally occurring trans-acting non-
Rev. Biomed. Eng. 10, 145–167 coding RNAs to sense molecular signals. Nucleic Acids Res. 40, 5775–
73 You, L. et al. (2004) Programmed population control by cell–cell 5786
communication and regulated killing. Nature 428, 868–871 104 Nagpal, R. (2001) Programmable Self-assembly: Constructing Global
74 Basu, S. et al. (2005) A synthetic multicellular system for programmed Shape Using Biologically-Inspired Local Interactions and Origami
pattern formation. Nature 434, 1130–1134 Mathematics. PhD Thesis, MIT Artificial Intelligence Laboratory
75 Brenner, K. et al. (2007) Engineering bidirectional communication Technical Memo 2001-008, MIT
mediates a consensus in a microbial biofilm consortium. Proc. Natl. 105 Galle, J. et al. (2009) From single cells to tissue architecture—a
Acad. Sci. U.S.A. 104, 17300–17304 bottom-up approach to modelling the spatio-temporal organisation
76 Balagadde, F.K. et al. (2008) A synthetic Escherichia coli predator – of complex multi-cellular systems. J. Math. Biol. 58, 261–283
prey ecosystem. Mol. Syst. Biol. 4, 1–8 106 Kondacs, A. (2003) Biologically-inspired self-assembly of two-
77 Tabor, J.J. et al. (2009) A synthetic genetic edge detection program. dimensional shapes using global-to-local compilation. In
Cell 137, 1272–1281 Proceedings of the 18th International Joint Conference on Artificial
78 Danino, T. et al. (2010) A synchronized quorum of genetic clocks. Intelligence (Gottlob, G. and Walsh, T., eds), pp. 633–638, Morgan
Nature 463, 326–330 Kaufmann
79 Prindle, A. et al. (2012) A sensing array of radically coupled genetic 107 St Johnston, D. and Nüsslein-Volhard, C. (1991) The origin of pattern
‘‘biopixels’’. Nature 481, 39–44 and polarity in the Drosophila embryo. Cell 68, 201–219
80 Tamsir, A. et al. (2011) Robust multicellular computing using 108 Wolf, J.B. and Wade, M.J. (2009) What are maternal effects (and what
genetically encoded NOR gates and chemical ‘wires’. Nature 469, are they not)? Philos. Trans. R. Soc. B: Biol. Sci. 364, 1107–1115
212–215 109 Khademhosseini, A. et al. (2006) Microscale technologies for tissue
81 Turing, A.M. (1952) The chemical basis of morphogenesis. Philos. engineering and biology. Proc. Natl. Acad. Sci. U.S.A. 103, 2480–
Trans. R. Soc. Lond. Ser. B: Biol. Sci. 237, 37–72 2487
82 Gierer, A. and Meinhardt, H. (1972) A theory of biological pattern 110 Bansal, T. et al. (2009) Patterned delivery and expression of gene
formation. Biol. Cybern. 12, 30–39 constructs into zebrafish embryos using microfabricated interfaces.
83 Hsia, J. et al. (2012) A feedback quenched oscillator produces Turing Biomed. Microdevices 11, 633–641
patterning with one diffuser. PLoS Comput. Biol. 8, e1002331 111 Park, J. et al. (2006) A microsystem for sensing and patterning
84 Wolfram, S. (1998) Cellular automata as models of complexity, In oxidative microgradients during cell culture. Lab Chip 6, 611–622
Nonlinear Physics for Beginners: Fractals, Chaos, Solitons, Pattern 112 Frevert, C.W. et al. (2006) Measurement of cell migration in response
Formation, Cellular Automata, Complex Systems, World Scientific to an evolving radial chemokine gradient triggered by a microvalve.
85 Mitchell, M. et al. (2005) Computation in cellular automata: a selected Lab Chip 6, 849–856
review, In In Non-Standard Computation, Wiley-VCH Verlag, pp. 95– 113 Ellis-Davies, G.C.R. (2007) Caged compounds: photorelease
140 technology for control of cellular chemistry and physiology. Nat.
86 Gutowitz, H. (1991) Cellular Automata: Theory and Experiment, MIT Methods 4, 619–628
Press 114 Huschka, R. et al. (2010) Visualizing light-triggered release of
87 Liu, C. et al. (2012) Sequential establishment of stripe patterns in an molecules inside living cells. Nano Lett. 10, 4117–4122
expanding cell population. Science 334, 238–241 115 Gooding, J.J. (2002) Electrochemical DNA hybridization biosensors.
88 Beloussov, L.V. and Grabovsky, V.I. (2006) Morphomechanics: goals, Electroanalysis 14, 1149–1156
basic experiments and models. Int. J. Dev. Biol. 50, 81–92 116 Jiang, X. et al. (2011) Rational growth of branched nanowire
89 Patwari, P. and Lee, R.T. (2008) Mechanical control of tissue heterostructures with synthetically encoded properties and
morphogenesis. Circ. Res. 103, 234–243 function. Proc. Natl. Acad. Sci. U.S.A. 108, 12212–12216
90 Gordon, R. (2006) Mechanics in embryogenesis and embryonics: prime 117 Cohen, D.J. et al. (2012) A highly elastic, capacitive strain gauge
mover or epiphenomenon? Int. J. Dev. Biol. 50, 245–253 based on percolating nanotube networks. Nano Lett. 12, 1821–1825
91 Ingber, D.E. (2006) Mechanical control of tissue morphogenesis 118 Laukhina, E. et al. (2010) Ultrasensitive piezoresistive all-organic
during embryological development. Int. J. Dev. Biol. 50, 255–266 flexible thin films. Adv. Mater. 22, 977–981
92 Harris, A.K. et al. (1984) Generation of spatially periodic patterns by a 119 Kuroki, H. et al. (2012) Responsive surfaces for life science
mechanical instability: a mechanical alternative to the Turing model. applications. Annu. Rev. Mater. Res. 42, 343–372
J. Embryol. Exp. Morphol. 80, 1–20 120 Duan, X. et al. (2012) Intracellular recordings of action potentials by
93 Blango, M.G. and Mulvey, M. (2009) Bacterial landlines: contact- an extracellular nanoscale field-effect transistor. Nat. Nanotechnol. 7,
dependent signaling in bacterial populations. Curr. Opin. 174–179
Microbiol. 12, 177–181 121 Sato, H. et al. (2009) Remote radio control of insect flight. Front.
94 Aoki, S.K. et al. (2009) Contact-dependent growth inhibition causes Integr. Neurosci. 3, 24
reversible metabolic downregulation in Escherichia coli. J. Bacteriol. 122 Ellis, T. et al. (2011) DNA assembly for synthetic biology: from parts to
191, 1777–1786 pathways and beyond. Integr. Biol. 3, 109–118
95 Thanassi, D.G. (2011) The long and the short of bacterial adhesion 123 Ma, S. et al. (2012) DNA synthesis, assembly and applications in
regulation. J. Bacteriol. 193, 327–328 synthetic biology. Curr. Opin. Chem. Biol. 16, 260–267

623
Review

Special Issue – Synthetic Cell Biology

Designer nucleic acids to probe and


program the cell
Yamuna Krishnan1 and Mark Bathe2
1
National Centre for Biological Sciences, TIFR, GKVK, Bellary Road, Bangalore 560 065, India
2
Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

Recent advances in nucleic acid sequencing, structural, As with most emerging technologies, with novelty comes
and computational technologies have resulted in dramat- expectation and anticipation. Notwithstanding, a number
ic progress in our understanding of nucleic acid structure of recent works show real promise for transforming our
and function in the cell. This knowledge, together with the abilities to measure and manipulate cell function in highly
predictable base-pairing of nucleic acids and powerful programmable ways that cannot be achieved with existing
synthesis and expression capabilities now offers the technologies. We review a number of these works and
unique ability to program nucleic acids to form precise provide our perspective on what we expect to be promising
3D architectures with diverse applications in synthetic avenues of research to pursue in this rapidly evolving,
and cell biology. The unique modularity of structural largely uncharted territory.
motifs that include aptamers, DNAzymes, and ribozymes,
together with their well-defined construction rules, Programming nucleic acid nanostructure shape
enables the synthesis of functional higher-order nucleic via self-assembly
acid complexes from these subcomponents. As we illus- A hallmark of DNA nanotechnology is the ability to pro-
trate here, these highly programmable, smart complexes gram structurally well-defined, thermodynamically stable
are increasingly enabling researchers to probe and pro- 3D polyhedra using Watson–Crick base-pairing and con-
gram the cell in a sophisticated manner that moves well struction rules [3,4], where the edges of the polyhedra
beyond the use of nucleic acids for conventional genetic comprise individual DNA duplexes. Now, conventional
manipulation alone. 3D shapes that may be produced with high yield and
sample homogeneity include tetrahedra, icosahedra,
Introduction to functional nucleic acid nanostructures cubes, dodecahedra, and, more recently, larger-scale, con-
Over 50 years ago, it was discovered that RNA had cata- siderably more complex wireframe structures [5] using the
lytic activity in addition to its role as an information- DNA origami approach [6], in which approximately 200
carrying polymer, giving rise to the hypothesis that life short strands are used to ‘staple’ together a longer single-
may have evolved from a precursor ‘RNA World’ without stranded DNA scaffold strand, reviewed in [7–9]
proteins or DNA [1,2]. Today, the diverse structures and (Figure 1a–c). Advanced computational tools enable the
activities adopted by RNAs are revealing a panoply of rational design of these structures accounting for thermo-
cellular functions that were barely imaginable at the time dynamic and mechanical stability key to function [9–12].
of this postulation by Alexander Rich and colleagues [1,2]. The principal advantages of using DNA to design
Unlike polypeptides, polynucleotides are structurally nucleic acid architectures include its long chemical half-
highly programmable polymers due to the specific, pair- life and highly specific Watson–Crick base-pairing that
wise Watson–Crick interactions of nucleic acid bases that offers the ability to design stable nanostructures of precise
result in well-defined secondary structures of predictable 3D form [9,12]. By contrast, non-canonical base-pairing
form. This, together with a simple set of construction rules acquired by RNA primary structures results in consider-
established for DNA in a landmark paper [3], the rapidly ably more diverse, dynamic secondary structures [13,14]
decreasing cost of nucleic acid synthesis, the programma- that are additionally subject to hydrolytic instability. This
bility of RNA expression in living cells, and our increasing
knowledge of higher-order RNA structure and chemical
activity, is resulting in an explosion in the number and Glossary
diversity of nucleic acid structures that are being designed 3WJ: a DNA or RNA motif comprising three duplexes radiating from a single
for diverse applications in science and technology, with a node; for example, the replication fork is a naturally occurring DNA 3WJ.
recently emerging focus on designer nucleic acids to probe Fluorescence in situ hybridization (FISH): a technique used to visualize
endogenous DNA or RNA molecules in cells and tissues using fluorescently
and program the cell. labeled antisense oligonucleotides.
Hybridization chain reaction (HCR): a process by which metastable hairpins
assemble by a cascade of hybridization events to form a long nicked duplex.
Systematic evolution of ligands by exponential enrichment (SELEX): uses in
Corresponding authors: Krishnan, Y. (yamuna@ncbs.res.in); vitro evolution to confer specific chemical properties to DNA and RNA
Bathe, M. (mark.bathe@mit.edu). molecules, typically high-affinity binding to a target small molecule.
Keywords: synthetic biology; RNA nanotechnology; DNA nanotechnology.

624 0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.10.001 Trends in Cell Biology, December 2012, Vol. 22, No. 12
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b) (c)


2.2–3 nm

+ 21 bp
+ +

al
ne
An

Ligate
D

D
B

C
B

E E Ligate
F F
A A

(d) (e) (f)

8 nm
FP
siRNAs
scaffold
Funconalizaon

Dicing

siRNA NPs

A B C D

TRENDS in Cell Biology

Figure 1. (a) Left: schematic of the assembly of four DNA strands to form a DNA tetrahedron via annealing and ligation. Reproduced, with permission, from [18]. Right:
atomic model and cryo-electron microscopy reconstruction of the tetrahedron. Reproduced, with permission, from [102]. (b) Left: overview of the scaffolded DNA origami
concept, showing crossovers formed by staple strands every 21 base pairs to form a 2D rectangle [9]. Right: atomic model of a 52 nm diameter wireframe structure [5,11]
compared with 2D transmission electron microscopy. Reproduced, with permission, from [5]. (c) Top: atomic model of a tetrahedron functionalized with DNA oligomers at
its vertices and cryo-electron microscopy reconstruction. Reproduced, with permission, from [24]. Bottom: schematic of physical encapsulation of fluorescent cargo (yellow
oval) within a DNA icosahedron that binds anionic ligand-binding receptors (ALBRs) to map the pH of endosomes as they mature. Reproduced, with permission, from [22].
(d) Functionalizable RNA nanoring scaffolds (grey) assembled through kissing-loop interactions between RNA modules. Nanoring scaffolds are functionalized with RNAs
(red) that can penetrate the siRNA pathway. Reproduced, with permission, from [28]. (e) RNA cubes constructed from six prefolded RNA building blocks. Reproduced, with
permission, from [27]. (f) Atomic model and 3D cryo-electron microscopy reconstruction of a non-uniform square antiprism constructed from self associating tRNA-based
motifs. Reproduced, with permission, from [31].

relative lack of structural fidelity with respect to DNA, (Figure 3a). The hexagonal barrel remains closed with the
however, is largely offset by its rich conformational and help of hasps comprising an aptamer and its complement
catalytic space that may be used to program associations of that dissociate when specific combinations of ligands bind
high affinity and specificity using aptamers [15], enzymatic the aptameric strand. These act as AND gates for the detec-
activity using ribozymes, and allostery using riboswitches tion of NKL cells from a mixture of NKL cells and leukocytes,
[16,17] (Figure 2). Further, the genetic encodability of RNA with the subsequently opened barrel exposing key Fab frag-
offers the exciting opportunity to program cells to express ments that trigger specific cellular processes such as T cell
precise nanoarchitectures for sensing, scaffolding, and activation or growth arrest. This work demonstrated that
signaling, among other functions, positioning RNA nano- allosteric DNA-based containers may be programmed to
technology as a powerful medium to probe and program expose cargo to cell-surface receptors based on selective,
cell function [18]. specific, logic-based multivalent binding in cell culture.
In cases where the covalent linkage of molecular cargo
Programming DNA architectures for targeted delivery to the DNA scaffold is infeasible either due to lack of
The tetrahedron has arisen as a popular 3D template due reactive groups or because molecular activity would be
to its minimal DNA content and high synthetic yield [18] compromised, alternative approaches are still needed.
(Figure 1a,c). An important early advance was the demon- One means of eliminating chemical conjugation is to use
stration that a single covalently linked protein molecule physical entrapment, whereby the cargo is physically con-
(cytochrome C) could be positioned within the tetrahedral strained within DNA polyhedra. This strategy was
cavity, opening up the possibility to use the interior space employed [21] to retain fluorescent dextrans or gold nano-
as a protected cargo hold for delivery [19]. To display such particles in DNA nanocages, and it was subsequently
protected cargo to external receptors, an innovative barrel- shown that these loaded polyhedra could be targeted spe-
shaped structure was designed [20] that sequesters T cell- cifically to single Caenorhabditis elegans cell types that
stimulating Fab fragments (the cargo) within its interior, expressed receptors for the DNA cage without compromis-
hidden from the external milieu when the barrel is closed ing cargo functionality [22] (Figure 1c). Programmed
625
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Aptamer

Aptazyme

Ribozyme/ Mn+
DNAzyme

Allosteric
+
aptamer

Riboswitches

Transcriponal Translaonal Metabolite-binding


TRENDS in Cell Biology

Figure 2. Schematic showing different functional nucleic acid motifs. Red stars and black circles represent reporter fluorophores. Aptamer: a DNA or RNA molecule that
specifically binds a small molecule or biomolecule (green oval). Aptazyme: a DNA or RNA molecule that is comprised of an aptamer domain fused to a catalytic domain,
where the latter becomes functional upon ligand-binding to the aptameric domain. Ribozyme/DNAzyme: an RNA/DNA molecule with catalytic properties, typically
consisting of a self-cleaving reaction in the presence of metal ions (denoted Mn+). Similar to aptazymes, the cleaving reaction is typically followed by strand dissociation.
Allosteric aptamer: aptameric modules sensitive to distinct ligands, typically an analyte and a reporter molecule, fused to one another so that the reporter molecule (red
star) may bind only when the analyte (green oval) is present. Riboswitches: regulatory RNA elements that act in cis by binding a small molecule, switching gene expression
on or off. Switches control either (left, Transcriptional) the formation of transcription terminator hairpins (red), (middle, Translational) accessibility to the Shine-Dalgarno
sequence (blue), or (right, Metabolite-binding) degradation of the transcript. Adapted from [17] and [79].

release of nanoparticles loaded within DNA scaffolds has motifs displayed in a tetrahedral fashion could effect im-
also been achieved by altering the pore size of the scaffold munostimulation of macrophages by interacting with Toll-
using a complementary DNA strand [23]. This suggests like receptor 9.
that it may be possible to both load and release encapsu- Importantly, such multivalent CpG motifs showed en-
lated molecular cargo from containers in a programmed hanced immunostimulatory effects, presumably due to
manner. Interfacing cell targeting with programmed re- increased ligand density. This increased ligand density
lease of non-tethered cargo, however, remains an open afforded by the immobilization of functional nucleic acid
challenge. motifs on a minimal DNA scaffold display was borne out in
In addition to their ability to encapsulate or sequester vivo [26], when a DNA tetrahedron was designed that
cargo for programmed release, DNA nanostructures have displayed small interfering RNAs (siRNAs) as well as a
the capacity to act as pegboards using small-molecule targeting ligand on each edge of the tetrahedron projecting
conjugation to position functional biomolecules precisely out from the tetrahedral scaffold (Figure 3b). The authors
in space due to their well-defined structure and address- showed impressive control over dosage by creating stoi-
able scaffold. One such demonstration was achieved using chiometrically identical particles in bulk with fixed ratios
DNA tetrahedra, in which short hairpins were grafted onto of the number of siRNA duplexes per tetrahedron. Fur-
the vertices of the scaffold without perturbing the tetrahe- ther, spatial orientation of the targeting ligands affected
dral geometry [24]. Such morphology-rich nucleic acid the efficiency of RNAi, presumably by affecting the uptake
motifs displayed on DNA tetrahedra were shown to retain that is in turn dependent on the surface density of the
functionality [25] when it was demonstrated that CpG ligands. This suggests that aspects of endocytosis, such as
626
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b) (c)


H F H
Cargo
F
Protein H
D0
aptamer F +
RNA aptamer
Scaffold
D0
Epifluorescence

1.8
Unscaffolded A B
1.6 (unorganized)
1.4

1.2
Scaffolded A B
1.0 (organized)

0.8 One- Two-


Discrete
dimensional dimensional

TRENDS in Cell Biology

Figure 3. (a) Functional design of a hexagonal barrel bearing Fab fragments (magenta) internally that opens in response to the binding of a signaling molecule (red circle) to
the aptameric strand (blue) of the hasps keeping the barrel closed. Reproduced, with permission, from [20]. (b) Schematic of tetrahedron assembly and structure obtained
from six DNA strands to which siRNA (orange) is hybridized. The tetrahedron bears one to six strands of siRNA in fixed stoichiometries. siRNA-bearing DNA tetrahedra are
shown to be specifically targeted to a tumor in a mouse model. Reproduced, with permission, from [26]. (c) Schematic of RNA aptamers used to localize protein synthesis
machinery to an RNA scaffold to create a larger-scale synthesis pathway in higher-order 1D and 2D structures. Reproduced, with permission, from [43].

ligand–receptor spacing and distinct uptake mechanisms remain an attractive alternative to de novo assembly from
that facilitate endosomal escape or lysosomal delivery synthesized nucleic acids until complex tertiary structures
may be interrogated using 3D designer DNA scaffolds [22]. can reliably be predicted computationally [38,39] or robust
principles of DNA-based assembly can be adopted in the
Programming RNA architectures for targeted delivery RNA domain. An alternative approach is to use protein
Advanced 3D designer RNA scaffolds have also been real- fragments to guide the assembly of nucleic acid building
ized, including nanocubes [27] and nanorings [28], using blocks into well-defined structures. For example, the L7Ae
the assembly principle ‘RNA tectonics,’ which exploits the protein is known to bind the k-turn motif and force it to adopt
modular nature of RNA molecules to form new architec- a bent conformation [40]. This has been used to form closed
tures [29] (Figure 1d–f). RNA-based particles may be polygonal shapes, with the protein occupying the vertices of
functionalized with siRNA motifs of controllable stoichi- the associating RNA building blocks [41].
ometry and incorporate chemically modified RNA and
DNA analogs due to their synthetic nature, and may Programming biomolecular spatial arrays for synthesis
therefore prove valuable for delivery in nanomedicine and signaling
[18,30]. Larger-scale RNA nanostructures approaching Organizing biomolecules in 3D to control, for example,
the size of DNA origami objects of 5–25 nm have also multi-enzyme or signaling cascades is another attractive
recently been achieved based on tectonics of tRNAs [31], aim for nanostructure design. For example, increasing
yielding fully addressable polyhedra functionalized with local concentrations of reactants by spatially localizing
proteins inside and outside the nanostructure. The bacte- enzymes in close proximity resulted in increased down-
riophage phi29 DNA packaging motor pRNA has been used stream mevalonate production by approximately 70-fold
as an alternative construction module [32], comprising a [42] using synthetic protein scaffolds in E. coli. Limitations
117-nucleotide RNA that folds into a three-way junction of protein-based scaffolds, however, include propensity for
and further associates into dimers, trimers, or hexamers nonspecific aggregation, lack of scaffold addressability, and
[33,34]. This enabled the construction of functional hetero- lack of scaffold structural fidelity due to a limited ability to
dimeric RNA nanoparticles bearing a targeting moiety program precisely higher-order protein nanostructures. As
such as folate, as well as an siRNA cargo that brings about an alternative, extended 1D and 2D RNA-based scaffolds
apoptosis in nasopharyngeal carcinomas [35]. were designed [43] to organize catalytic enzymes in bacte-
One important limitation of RNA nanoparticles assem- ria to achieve enhanced rates of hydrogen synthesis of up to
bled through intermolecular tertiary interactions for cell- approximately 50-fold, demonstrating in vivo structural
based applications, however, is the need for metal salts to engineering of the intracellular environment (Figure 3c).
stabilize intermolecular junctions. Recently, this limitation Similarly, increased metabolic flux in Escherichia coli was
was surmounted by a new thermodynamically stable 3WJ achieved using a plasmid-based DNA scaffold and zinc-
junction based on the pRNA junction that circumvents this finger binding motifs [44]. Synthetic microcompartments
requirement and retains its structure at physiological con- for sequestration of reactants similar to natural organelles,
centrations of Mg2+. Further, this junction was programmed such as carboxysomes, and scaffolding motifs for signaling
using three distinct cargoes: a targeting moiety, an siRNA modules with allosteric functionality [45,46] represent
against an antiapoptotic marker, and an aptamer or ribo- other enticing possibilities. In vitro scaffolded enzyme
zyme, all of which proved to be functional in vivo [36]. cascades [47,48] studied in conjunction with computational
Additionally, the realization of RNase-resistant yet biologi- models will be useful in defining the optimal design criteria
cally active pRNA [37] suggests that assembly of preformed for maximizing chemical synthesis yield and efficiency in
RNA components from native biological constructs will the cell.
627
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (c)

Detecon stage Amplificaon stage


HBI (in GFP)
mRNA target H1 Metastable
Probe binding sites H2 hairpins 13-2 RNA DMHBI + DMHBI +
DMHBI
Spinach Spinach-DFHBI
H1 H2 13-2 RNA control RNA
fluorescent complex
Probe set Tethered
DMHBI
Hybridize probes
and wash Fluorescent H1 H2 fluorescent
in situ HCR amplificaon
and wash polymers
Iniators H2
H1

0 min 15 min 30 min

mRNA target

Minimal Target binding Fluorescent complex


90 min 120 min 150 min
fluorescence reinforces structure formaon

(b) (d)
Tg(flk1:egfp) tpm3 elavl3 ntla sox10

Donor Acceptor D/A


H+

5 min
O1 O2
O3 OH-

17 min
z = 20 µm z = 28 µm

60 min
H+

z = 44 µm z = 52 µm 7

TRENDS in Cell Biology

Figure 4. (a) Hybridization chain reaction (HCR) principle whereby metastable fluorescent RNA hairpins H1 and H2 self-assemble into amplification polymers in the
presence of a specific target RNA initiator. An amplification cascade using fluorescent hairpin pairs results in localized amplification of fluorescence. Reproduced, with
permission, from [55]. (b) HCR applied to the multiplexed detection of five distinct mRNAs in whole-mount zebrafish embryos. Reproduced, with permission, from [55]. (c)
Top: chemical structures of 4-hydroxybenzylidene imidazolinone (HBI) (green) and 3,5-dimethoxy-4-hydroxybenzylidene imidazolinone (DMHBI) (black) are shown in
addition to the RNA aptamer Spinach that binds DFHBI but requires the formation of stem 2. Complexation of DMHBI with its aptamer (13-2) enhances its fluorescence.
Reproduced, with permission, from [58]. Bottom: S-adenosyl methionine (SAM) (magenta hexagon) sensor using RNA aptamer region Spinach (black), a transducer
domain (orange), and a SAM aptamer (blue). SAM binding of the sensor promotes the binding of DFHBI (green circle) to Spinach, leading to fluorescence. Bottom right:
distinct patterns of SAM accumulation after adding methionine to Escherichia coli expressing the sensor. From [60]. Reprinted with permission from AAAS. (d) Top left:
Schematic of an i-motif powered, Förster resonance energy transfer (FRET)-based DNA pH sensor, the I-switch, that undergoes a pH triggered conformational change.
Bottom left: the I-switch is internalized by specific receptors present on coelomocytes (yellow cells) in Caenorhabditis elegans and is able to map spatiotemporal pH
changes associated with maturing endosomes on the anionic ligand-binding receptor (ALBR) pathway. Right: pH heat maps of early endosomes, late endosomes and
lysosomes along the ALBR pathway. Reproduced, with permission, from [63].

Probing the cell with fluorescence-based sensors that forms the basis of localized amplification of the fluo-
Biological sensors aim to recognize molecular inputs with rescent signal. This approach enabled multiplexed RNA–
high specificity and sensitivity to transduce them into FISH in whole-mount embryos of impressive clarity, albeit
readouts with minimal perturbation to the cell. Molecular still in fixed samples (Figure 4b).
beacons are a simple early form of nucleic acid-based Directly visualizing RNA molecules in live cells to probe
biosensor comprising a single hairpin with a complemen- RNA trafficking, localization, and transcriptional bursting
tary probe sequence and quenched Förster resonance en- remains an important challenge [52,57] that currently
ergy transfer (FRET) pair that fluoresces on hybridization limits our understanding of the burgeoning RNA layer of
to the target mRNA/DNA [49,50]. Genomic DNA loci and gene regulation. A promising recent development towards
single RNA transcripts are also now routinely measured in realizing live-cell mRNA imaging exploits the photophysi-
fixed cells using fluorescence in situ hybridization (FISH) cal properties of the green fluorescent protein (GFP) fluor-
[51–54], which tiles numerous fluorescent probes along the ophore using RNA aptamers to yield multispectral RNA
target sequence to enhance reporter signal. Although use- mimics of GFP [58]. 4-Hydroxybenzylidene imidazolinone
ful for fixed cells and tissue sections that require only (HBI), the fluorescent moiety in GFP, fluoresces when
limited imaging depth, FISH still suffers from a limited caged in the beta-barrel core of GFP due to proper fluores-
signal-to-noise ratio due to the finite number of fluoro- cence emission from the excited fluorophore state, entering
phores that can be tiled to the target transcript without the dark state when exposed to bulk solution. SELEX was
amplification and limited probe specificity for short used [58] to obtain an RNA aptamer that binds DMHBI, a
sequences. To overcome these limitations and enable sin- chemical analog of HBI, resulting in fluorescence emission
gle-step, multiplexed RNA imaging, a triggered hybridiza- in the bound state (Figure 4c). Further, the authors syn-
tion chain reaction was used [55,56] to dramatically thesized the small molecule DFHBI that is exclusively in
increase the signal-to-noise ratio (Figure 4a). Here, RNA the phenolate form at neutral pH and also obtained an
probes folded into metastable hairpins harboring fluores- aptamer, termed Spinach, that binds this target molecule.
cent reporters restructure in the presence of a desired DFHBI exhibits fluorescence when bound to Spinach that
mRNA sequence into a nicked double-stranded polymer is approximately half as bright as enhanced GFP (eGFP),
628
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

yet brighter than many other fluorescent proteins, consid- Digital and analog controls of protein expression in
erably more photo-stable than GFP, and does not require living systems are essential tools for probing the role of
the maturation time of GFP. Impressive live-cell imaging protein function in determining cell state and behavior.
of 5S RNA translocation in mammalian cells was achieved Controlling translation using small-molecule inducers is
when incorporated into the 30 UTR, which does not affect of major interest for this purpose, both for the reversible
localization. control intrinsic to such methods per se, and specifically for
To achieve conditional functionality, first described [59] the study, for example, of processes and pathogens not
using the cytotoxic malachite green (MG), a metabolite- amenable to conventional genetic tools, such as the ma-
binding aptamer was fused [60] to Spinach using a commu- larial parasite Plasmodium falciparum. The adenine
nication module that confers proper folding of Spinach and riboswitch is one such ‘translational ON’ switch that
subsequent fluorescence in the presence of DFHBI and the allows translation to occur in the presence of adenine
metabolite in a manner that is dependent on concentration of by restructuring the riboswitch to unmask the Shine-
the latter (Figure 4c). This enabled visualization of S-ade- Dalgarno sequence. This switch was reengineered [68]
nosyl methionine distribution in living bacteria, which, to- to produce two different riboswitches that are responsive
gether with the ability to evolve aptamers to bind nearly any either to ammeline or to 2-aminopurine. In subsequent
small molecule, provides an exciting opportunity to image work [69], the authors went on to independently control in
bioactive small molecules and RNAs in living systems. the same bacterium the coexpression of eGFP and DsRed
In addition to RNA-based switches, DNA-based switches induced by ammeline and 2-aminopurine, respectively, in
may also exhibit large-scale conformational rearrange- a dose-dependent manner (Figure 5a). A major advantage
ments in response to molecular cues including pH that offer of these approaches over analogous protein-based systems
the unique ability to map the latter in live cells. Spatiotem- is the elimination of crosstalk and/or the ability to control
poral maps of pH in endocytic compartments within living protein expression profiles more finely. Further, protein
cells have provided important insights into the stages of stoichiometries may possibly be sculpted in a multiplexed
endosomal maturation, viral entry mechanisms, and their fashion using this approach with a cocktail of inducers at
crosstalk with secretory pathways [61]. Imaging endosomal different concentrations. Such systems thereby facilitate
maturation within a multicellular, live organism was re- quantitative approaches to probing and understanding
cently achieved using a FRET-based, pH-sensitive DNA protein function in live cells. In this respect, a recent
switch called the I-switch [62] that undergoes a conforma- ion-sensitive fluoride riboswitch represents a valuable
tional change under acidic conditions (Figure 4d). This addition for temporal control of gene expression using
conformational change was mediated by i-motif formation orthogonal triggers, because fluoride does not result in
of C-rich sequences on the I-switch and the device was first metabolites that could potentially crosstalk with the trig-
validated in cell culture [63]. Importantly, the functionality ger, is cell-permeable, and may be washed out post-induc-
of this pH-triggered device was quantitatively recapitulated tion [70].
from the in vitro to the in vivo context, providing such The RNA motifs and small molecules described above
bistable DNA devices with a new role inside living systems may be used not only to modulate the expression of any
as probes of the intracellular chemical environments of unrelated gene, but also to turn on a pathway that meta-
specific proteins. bolizes the chemical trigger. It is therefore feasible to
engineer synthetic functions into living systems, such as
Programming the cell to perform desired functions sensing and responding to a non-natural molecule. As an
Synthetic biology seeks to program novel functions into example, a synthetic riboswitch was used to control motili-
living systems, for example, convert or synthesize biomole- ty in E. coli by introducing an atrazine aptamer in the 50
cules including biofuels, sense and destroy biological mate- UTR of the cheZ gene, a key player in the chemotactic E.
rial including pathogens, harvest, convert, and store solar coli motility machinery, in the background of an atrazine-
energy, and test working hypotheses on the function of metabolizing gene [71]. This resulted in ‘seek-and-destroy’
complex living systems [64,65]. Engineered biological sys- bacteria that were programmed to seek out and degrade
tems must therefore have the capacity to receive molecular the toxic herbicide atrazine. This approach to controlling
cues, perform context-dependent computation, and produce chemotaxis is potentially generalizable to myriad chemi-
outputs that lead to desired changes in cell state or function, cals where E. coli may be engineered to clear unwanted
and achieve all of this under the noisy, heterogeneous con- chemicals in the environment or other systems [72].
ditions in which cells operate. Unlike solid-state machines Apart from ON/OFF riboswitches that respond to a
that principally seek to store and compute large amounts of unique molecular cue, there are rare natural examples of
information, typically containing only a single mode of input complex logical control over gene expression through tan-
and output, biological computation exploits an opposing side dem riboswitches on a single transcript [73]. Such control
of the computational spectrum, often comprising only a few was realized synthetically using tandem riboswitches cou-
‘states’ with large numbers of inputs comprising small pled to the hammerhead ribozyme (HHR) on the 30 UTR of a
molecules, metabolites, RNAs, and protein, and outputs given mRNA transcript that mediated logical control over
at the levels of transcription, translation, ligation, cleavage, mRNA cleavage and degradation (Figure 5b). Thus, yeast-
and small-molecule signaling [17,64–67]. Here, we focus on enhanced GFP (yeGFP) was expressed as a function of AND,
selected recent findings of relevance to a breadth of cell OR, NAND, and NOR logic by positionally coupling multiple
biological problems and refer the reader to the preceding in- aptamers via crucial connecting modules on the mRNA
depth reviews for additional background. transcript such that the addition of two small-molecule
629
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b)
AND gate
A Input A Input B
AB Output
B
OFF-state ON-state
3′ [SI 1.2]
Start A B Output
RBS theo tc GFP
0 0 0
3′ 0 1 0
5′ 5′ 1 0 0
AAAAA
1 1 1

Output protein (high when AB)


250

125
5:1 (+)
4:1
NOR gate
Ammeline (μM)

64 Input A Input B
3:1 A
A+B Output
32 2:1 B
[SI 1.3]
1:1 A B Output
16
1:2 theo tc GFP
8 1:3 0 0 1
1:4
0 1 0
4 1:5 (+)
1 0 0
AAAAA
1 1 0
0
0 4 8 16 32 64 125 250
2-aminopurine (μM)

Output protein (high when A+B)

TRENDS in Cell Biology

Figure 5. (a) Top: translational mechanism of gene regulation by the engineered adenine riboswitch responsive to either ammeline or 2-aminopurine (2AP) (blue hexagon).
Binding to the inducer releases the normally sequestered ribosome-binding site (RBS) and start codon, allowing mRNA translation. Bottom: contour plots showing relative
coexpression ratios of enhanced green fluorescent protein (eGFP): DsRed for the full range of ammeline and 2-aminopurine. Reproduced, with permission, from [69]. (b)
Top: an RNA device incorporated into an mRNA that expresses yeast-enhanced GFP (yeGFP) according to AND logic and the associated truth table. In the absence of either
A or B, at least one functional hammerhead ribozyme (HHR) motif cleaves the transcript. In the presence of both A and B, both HHR motifs are inactivated and the yeGFP is
produced. Reproduced, with permission, from [74]. Bottom: an RNA device incorporating two analogous inactivated HHR motifs that become activated on addition of A or B
serves as a NOR gate. Reproduced, with permission, from [74].

inputs resulted in activation or deactivation of mRNA high-throughput approaches are likely to play an impor-
cleavage by HHR [74]. tant future role in identifying suitable candidates for RNA-
Examples of implementation of logical control of cell based function.
function in mammalian systems are limited [75,76,77],
representing a major challenge of interest going forward. Concluding remarks
In a recent success, specific signatures of microRNAs Fifty years after the postulate that RNA stands at the
characteristic of cancer cells were read into a logic-gated crossroads of the biocatalytic and information-bearing
computer and used to induce apoptosis using network logic molecules: proteins and DNA, our rapidly advancing un-
[76]. The logic framework implemented is completely gen- derstanding of nucleic acid form and function, together
eral for use in detecting specific gene expression profiles in with our ability to program these polymers for custom
mammalian cells [78], utilizing a combination of micro- purposes offer immense room for their application to basic
RNA inputs together with different logic gates such as and applied cell biology.
AND and NOT to produce a specific programmed cellular We envision an eventual palette of made-to-order RNA-
output. Various means of using riboswitches, aptamers, based devices to probe cellular machinery including levels
and allosteric aptamers to switch on or off gene expression and localization of metabolites, small molecules, ions, and
are comprehensively discussed in [79]. diverse coding and non-coding RNAs in a multiplexed
The aforementioned studies have largely used manual manner with sensitivities that are inaccessible to existing
approaches to achieve RNA-based programs that modulate technologies that are largely protein-based (Figure 6). Ex-
gene expression. In an important development, a high- ploration of intracellular spaces in an organelle-specific
throughput approach was delineated [80] that screens manner to probe their proteomic composition in individual,
endogenous RNA interactions to identify orthogonal living cells, as well as intercellular spaces such as the
mRNA-regulatory RNA pairs. Using an integrated model- neuronal or immunological synapse [81,82] with such de-
ing–experimental framework based on the free energies of signer devices represent intriguing uncharted territories.
RNA hybridization, the authors realized a library of engi- Utilizing the ability to precisely engineer higher-order
neered RNA regulators for predictable interactors. Such structural scaffolds and nucleic acid architectures from
630
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Neu

lar
rotra

struc molecu
Scaffolds Proteomics

rs
nsm

ture

to
ro

mo
ier

Mac

lar
s

cu
Fo

ole
ule

rce
M
lec

M
et o
ab
oli -m s
te gle sic
s Sin ophy
bi

s
ure
l
-cel

DN
DNA Cell acons

uct
Azy
e r
int

str
me

no
s

Na
Super-resoluon
DNA Signaling
microscopy cascades
Sensors Ribozymes Aptamers Delivery
RNA

Ions he
s
Diagn
oscs

mi
itc

RN
osw

A
Rib

The
A rap
RN e u
cs
Programs

Lig
ns
ei

ht
ot

-h
Pr

ar
Ene versio
n

ve
con
ao

s
rgy
Biosynth
diaon

gn
gul
ore

n
Bioreme
Rib

esis

TRENDS in Cell Biology

Figure 6. Overview showing the distinct layers of tools based on DNA and RNA and their diverse applications to cellular and synthetic biology.

DNA and RNA templates at the scale of macromolecular of force sensation and feedback on binding interactions
assemblies such as the nuclear pore complex, chaperones, mediated by focal adhesions [96] and cell–cell junctions
and the ribosome further offers the unique ability to probe [97], will be key to mechanistically understanding morpho-
the effect of the specific higher-order organization of di- genesis, development, and regeneration.
verse macromolecular structures, including relative posi- As evidenced by the macromolecular architectures and
tioning and orientation, on their function. programs presented in this review, nucleic acids offer the
Active scaffolds integrated with precisely positioned pro- unique capability to design scaffolds with high precision for
teins and protein domains may additionally be designed diverse applications in cell biology. Among the many chal-
with allosteric response to probe signaling cascades [45], lenges we face in bringing these structures to bear broadly
interrogate cell circuits, probe the RNA transcriptome [83], on the study of cell biology are an understanding of how to
modulate enzyme cascades and metabolic pathways [42– control and circumvent crosstalk between thousands of
44], perform single-molecule manipulations, and provide molecules and pathways in the cell to leverage sophisticat-
platforms for structural biology [84,85] as well as super- ed nucleic acid-based in vitro programs [66,98–100], as well
resolution imaging [86,87]. Extrinsic control of nucleic acid as the immense heterogeneity and variability in molecular
duplex stability and conformation with light, as demonstrat- levels that feed back to determine cell state and response,
ed extensively in vitro [88–90], will offer novel means to particularly in complex mammalian systems [17,64,66,75–
spatially and temporally regulate the activity of scaffolds, 78,101]. Although the challenge is great, the ever-increas-
sensors, and inhibitors in living systems from individual ing arsenal of new tools and technologies at our disposal
cells to developing embryos [91]. Finally, construction of ensures that meaningful progress will be achieved in the
extracellular origami using DNA to position cells in 3D near term. Along the way, developing and applying nucleic
[92,93] to probe their interaction with their environment acid-based tools to address problems that are currently
in artificial tissue scaffolds and on 2D substrates [94,95] will intractable using conventional technologies alone will un-
provide insight into their function in organizing higher- doubtedly aid the advance of synthetic biology in its aim to
order tissues. Such studies, together with an understanding engineer biological systems for the benefit of society.
631
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Acknowledgments 27 Afonin, K.A. et al. (2010) In vitro assembly of cubic RNA-based


M.B. acknowledges the Office of Naval Research (ONR N000141210621), scaffolds designed in silico. Nat. Nanotechnol. 5, 676–682
the Army Research Office (MURI W911NF-12-1-0420), and MIT Faculty 28 Grabow, W.W. et al. (2011) Self-assembling RNA nanorings based on
Start-up Funds for financial support. Y.K. acknowledges the Innovative RNAI/II inverse kissing complexes. Nano Lett. 11, 878–887
Young Biotechnologist Award and the Wellcome Trust-DBT India 29 Jaeger, L. and Chworos, A. (2006) The architectonics of programmable
Alliance Senior Research Fellowship. The authors are grateful to RNA and DNA nanostructures. Curr. Opin. Struct. Biol. 16, 531–543
Camille Delebecque, Hendrik Dietz, Shawn Douglas, Luc Jaeger, Samie 30 Afonin, K.A. et al. (2011) Design and self-assembly of siRNA-
Jaffrey, Takayuki Kato, Hyukjin Lee, Chengde Mao, Jason Micklefield, functionalized RNA nanoparticles for use in automated
Niles Pierce, Christina Smolke, and Andrew Turberfield, for providing nanomedicine. Nat. Protoc. 6, 2022–2034
high-resolution versions of published figures that are reproduced here, as 31 Severcan, I. et al. (2010) A polyhedron made of tRNAs. Nat. Chem. 2,
well as Matthew Adendorff for help with rendering figures. The authors 772–779
appreciate discussions and feedback from numerous colleagues in the 32 Guo, P.X. et al. (1998) Inter-RNA interaction of phage phi 29 pRNA to
preparation of this work, and sincerely apologize to those authors whose form a hexameric complex for viral DNA transportation. Mol. Cell 2,
work was not included due to space limitations. 149–155
33 Chen, C.P. et al. (2000) A dimer as a building block in assembling RNA
- a hexamer that gears bacterial virus phi29 DNA-translocating
References
machinery. J. Biol. Chem. 275, 17510–17516
1 Rich, A. (1962) On the problems of evolution and biochemical
34 Shu, D. et al. (2004) Bottom-up assembly of RNA arrays and
information transfer. In Horizons in Biochemistry (Kasha, M. and
superstructures as potential parts in nanotechnology. Nano Lett. 4,
Pullmann, B., eds), pp. 103–126, Plenum Press
1717–1723
2 Gilbert, W. (1986) Origin of life - the RNA world. Nature 319, 618
35 Guo, S. et al. (2006) Construction of folate-conjugated pRNA of
3 Seeman, N.C. (1982) Nucleic acid junctions and lattices. J. Theor. Biol.
bacteriophage phi29 DNA packaging motor for delivery of chimeric
99, 237–247
siRNA to nasopharyngeal carcinoma cells. Gene Ther. 13, 814–820
4 Seeman, N.C. (2010) Nanomaterials based on DNA. Annu. Rev.
36 Shu, D. et al. (2011) Thermodynamically stable RNA three-way
Biochem. 79, 65–87
junction for constructing multifunctional nanoparticles for delivery
5 Dietz, H. et al. (2009) Folding DNA into twisted and curved nanoscale
of therapeutics. Nat. Nanotechnol. 6, 658–667
shapes. Science 325, 725–730
37 Liu, J. et al. (2011) Fabrication of stable and RNase-resistant RNA
6 Rothemund, P.W.K. (2006) Folding DNA to create nanoscale shapes
nanoparticles active in gearing the nanomotors for viral DNA
and patterns. Nature 440, 297–302
packaging. ACS Nano 5, 237–246
7 Nangreave, J. et al. (2010) DNA origami: a history and current
38 Cruz, J.A. et al. (2012) RNA-puzzles: a CASP-like evaluation of RNA
perspective. Curr. Opin. Chem. Biol. 14, 608–615
three-dimensional structure prediction. RNA 18, 610–625
8 Shih, W.M. and Lin, C.X. (2010) Knitting complex weaves with DNA
39 Westhof, E. (2012) RNA modeling, naturally. Proc. Natl. Acad. Sci.
origami. Curr. Opin. Struct. Biol. 20, 276–282
U.S.A. 109, 2691–2692
9 Castro, C.E. et al. (2011) A primer to scaffolded DNA origami. Nat.
40 Turner, B. et al. (2005) Induced fit of RNA on binding the L7Ae protein
Methods 8, 221–229
to the kink-turn motif. RNA 11, 1192–1200
10 Douglas, S.M. et al. (2009) Rapid prototyping of 3D DNA-origami
41 Ohno, H. et al. (2011) Synthetic RNA-protein complex shaped like an
shapes with caDNAno. Nucleic Acids Res. 37, 5001–5006
equilateral triangle. Nat. Nanotechnol. 6, 115–119
11 Kim, D-N. et al. (2012) Quantitative prediction of 3D solution shape
42 Dueber, J.E. et al. (2009) Synthetic protein scaffolds provide modular
and flexibility of nucleic acid nanostructures. Nucleic Acids Res. 40,
control over metabolic flux. Nat. Biotechnol. 27, 753–759
2862–2868
43 Delebecque, C.J. et al. (2011) Organization of intracellular reactions
12 Dirks, R.M. and Pierce, N.A. (2003) A partition function algorithm for
with rationally designed RNA assemblies. Science 333, 470–474
nucleic acid secondary structure including pseudoknots. J. Comput.
44 Conrado, R.J. et al. (2012) DNA-guided assembly of biosynthetic
Chem. 24, 1664–1677
pathways promotes improved catalytic efficiency. Nucleic Acids
13 Isambert, H. (2009) The jerky and knotty dynamics of RNA. Methods
Res. 40, 1879–1889
49, 189–196
45 Good, M.C. et al. (2011) Scaffold proteins: hubs for controlling the flow
14 Shapiro, B.A. et al. (2008) Protocols for the in silico design of RNA
of cellular information. Science 332, 680–686
nanostructures. Methods Mol. Biol. 474, 93–115
46 Bashor, C.J. et al. (2010) Rewiring cells: synthetic biology as a tool to
15 Ellington, A.D. and Szostak, J.W. (1990) In vitro selection of RNA
interrogate the organizational principles of living systems. Annu. Rev.
molecules that bind specific ligands. Nature 346, 818–822
Biophys. 39, 515–537
16 Guo, P.X. (2010) The emerging field of RNA nanotechnology. Nat.
47 Fu, J.L. et al. (2012) Interenzyme substrate diffusion for an enzyme
Nanotechnol. 5, 833–842
cascade organized on spatially addressable DNA nanostructures. J.
17 Isaacs, F.J. et al. (2006) RNA synthetic biology. Nat. Biotechnol. 24,
Am. Chem. Soc. 134, 5516–5519
545–554
48 Wilner, O.I. et al. (2009) Enzyme cascades activated on topologically
18 Goodman, R.P. et al. (2005) Rapid chiral assembly of rigid DNA
programmed DNA scaffolds. Nat. Nanotechnol. 4, 249–254
building blocks for molecular nanofabrication. Science 310, 1661–
49 Tyagi, S. and Kramer, F.R. (1996) Molecular beacons: probes that
1665
fluoresce upon hybridization. Nat. Biotechnol. 14, 303–308
19 Erben, C.M. et al. (2006) Single-molecule protein encapsulation in a
50 Bratu, D.P. et al. (2003) Visualizing the distribution and transport
rigid DNA cage. Angew. Chem. Int. Ed. 45, 7414–7417
of mRNAs in living cells. Proc. Natl. Acad. Sci. U.S.A. 100, 13308–
20 Douglas, S.M. et al. (2012) A logic-gated nanorobot for targeted
13313
transport of molecular payloads. Science 335, 831–834
51 Femino, A. et al. (1998) Visualization of single RNA transcripts in situ.
21 Bhatia, D. et al. (2009) Icosahedral DNA nanocapsules by modular
Science 280, 585–590
assembly. Angew. Chem. Int. Ed. 48, 4134–4137
52 Tyagi, S. (2009) Imaging intracellular RNA distribution and dynamics
22 Bhatia, D. et al. (2011) A synthetic icosahedral DNA-based host-cargo
in living cells. Nat. Methods 6, 331–338
complex for functional in vivo imaging. Nat. Commun. 2, 339
53 Capodieci, P. et al. (2005) Gene expression profiling in single cells
23 Lo, P.K. et al. (2010) Loading and selective release of cargo in DNA
within tissue. Nat. Methods 2, 663–665
nanotubes with longitudinal variation. Nat. Chem. 2, 319–328
54 Raj, A. et al. (2008) Imaging individual mRNA molecules using
24 Zhang, C.A. et al. (2010) Exterior modification of a DNA tetrahedron.
multiple singly labeled probes. Nat. Methods 5, 877–879
Chem. Commun. 46, 6792–6794
55 Choi, H.M.T. et al. (2010) Programmable in situ amplification for
25 Li, J. et al. (2011) Self-assembled multivalent DNA nanostructures for
multiplexed imaging of mRNA expression. Nat. Biotechnol. 28, 1208–
noninvasive intracellular delivery of immunostimulatory CpG
1212
oligonucleotides. ACS Nano 5, 8783–8789
56 Dirks, R.M. and Pierce, N.A. (2004) Triggered amplification by
26 Lee, H. et al. (2012) Molecularly self-assembled nucleic acid
hybridization chain reaction. Proc. Natl. Acad. Sci. U.S.A. 101,
nanoparticles for targeted in vivo siRNA delivery. Nat.
15275–15278
Nanotechnol. 7, 389–393

632
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

57 Armitage, B.A. (2011) Imaging of RNA in live cells. Curr. Opin. Chem. 82 Manz, B.N. and Groves, J.T. (2010) Spatial organization and signal
Biol. 15, 806–812 transduction at intercellular junctions. Nat. Rev. Mol. Cell Biol. 11,
58 Paige, J.S. et al. (2011) RNA mimics of green fluorescent protein. 342–352
Science 333, 642–646 83 Wan, Y. et al. (2011) Understanding the transcriptome through RNA
59 Stojanovic, M.N. and Kolpashchikov, D.M. (2004) Modular aptameric structure. Nat. Rev. Genet. 12, 641–655
sensors. J. Am. Chem. Soc. 126, 9266–9270 84 Douglas, S.M. et al. (2007) DNA-nanotube-induced alignment of
60 Paige, J.S. et al. (2012) Fluorescence imaging of cellular metabolites membrane proteins for NMR structure determination. Proc. Natl.
with RNA. Science 335, 1194 Acad. Sci. U.S.A. 104, 6644–6648
61 Weisz, O.A. (2003) Acidification and protein traffic. Int. Rev. Cytol. 85 Berardi, M.J. et al. (2011) Mitochondrial uncoupling protein 2
226, 259–319 structure determined by NMR molecular fragment searching.
62 Surana, S. et al. (2011) An autonomous DNA nanomachine maps Nature 476, 109–113
spatiotemporal pH changes in a multicellular living organism. Nat. 86 Opazo, F. et al. (2012) Aptamers as potential tools for super-resolution
Commun. 2, 340 microscopy. Nat. Methods 9, 938–939
63 Modi, S. et al. (2009) A DNA nanomachine that maps spatial and 87 Lin, C. et al. (2012) Submicrometre geometrically encoded fluorescent
temporal pH changes inside living cells. Nat. Nanotechnol. 4, 325–330 barcodes self-assembled from DNA. Nat. Chem. 4, 832–839
64 Khalil, A.S. and Collins, J.J. (2010) Synthetic biology: applications 88 Nishioka, H. et al. (2012) A photon-fueled DNA nanodevice that
come of age. Nat. Rev. Genet. 11, 367–379 contains two different photoswitches. Angew. Chem. Int. Ed. 51,
65 Andrianantoandro, E. et al. (2006) Synthetic biology: new engineering 1165–1168
rules for an emerging discipline. Mol. Syst. Biol. 2, 2006.0028 89 Wang, C.M. et al. (2011) Caged molecular beacons: controlling nucleic
66 Benenson, Y. (2012) Biomolecular computing systems: principles, acid hybridization with light. Chem. Commun. 47, 5708–5710
progress and potential. Nat. Rev. Genet. 13, 455–468 90 Asanuma, H. et al. (2007) Synthesis of azobenzene-tethered DNA for
67 Chen, X. and Ellington, A.D. (2010) Shaping up nucleic acid reversible photo-regulation of DNA functions: hybridization and
computation. Curr. Opin. Biotechnol. 21, 392–400 transcription. Nat. Protoc. 2, 203–212
68 Dixon, N. et al. (2010) Reengineering orthogonally selective 91 Toettcher, J.E. et al. (2011) The promise of optogenetics in cell biology:
riboswitches. Proc. Natl. Acad. Sci. U.S.A. 107, 2830–2835 interrogating molecular circuits in space and time. Nat. Methods 8,
69 Dixon, N. et al. (2012) Orthogonal riboswitches for tuneable 35–38
coexpression in bacteria. Angew. Chem. Int. Ed. 51, 3620–3624 92 Gartner, Z.J. and Bertozzi, C.R. (2009) Programmed assembly of 3-
70 Baker, J.L. et al. (2012) Widespread genetic switches and toxicity dimensional microtissues with defined cellular connectivity. Proc.
resistance proteins for fluoride. Science 335, 233–235 Natl. Acad. Sci. U.S.A. 106, 4606–4610
71 Mandelbaum, R.T. et al. (1995) Isolation and characterization of a 93 Selden, N.S. et al. (2012) Chemically programmed cell adhesion with
pseudomonas sp that mineralizes the s-triazine herbicide atrazine. membrane-anchored oligonucleotides. J. Am. Chem. Soc. 134, 765–
Appl. Environ. Microbiol. 61, 1451–1457 768
72 Sinha, J. et al. (2010) Reprogramming bacteria to seek and destroy an 94 Hung, A.M. et al. (2010) Large-area spatially ordered arrays of gold
herbicide. Nat. Chem. Biol. 6, 464–470 nanoparticles directed by lithographically confined DNA origami.
73 Sudarsan, N. et al. (2006) Tandem riboswitch architectures exhibit Nat. Nanotechnol. 5, 121–126
complex gene control functions. Science 314, 300–304 95 Kershner, R.J. et al. (2009) Placement and orientation of individual
74 Win, M.N. and Smolke, C.D. (2008) Higher-order cellular information DNA shapes on lithographically patterned surfaces. Nat.
processing with synthetic RNA devices. Science 322, 456–460 Nanotechnol. 4, 557–561
75 Leisner, M. et al. (2010) Rationally designed logic integration of 96 Geiger, B. et al. (2009) Environmental sensing through focal
regulatory signals in mammalian cells. Nat. Nanotechnol. 5, 666–670 adhesions. Nat. Rev. Mol. Cell Biol. 10, 21–33
76 Xie, Z. et al. (2011) Multi-input RNAi-based logic circuit for 97 Levayer, R. and Lecuit, T. (2012) Biomechanical regulation of
identification of specific cancer cells. Science 333, 1307–1311 contractility: spatial control and dynamics. Trends Cell Biol. 22,
77 Ausländer, S. et al. (2010) A ligand-dependent hammerhead ribozyme 61–81
switch for controlling mammalian gene expression. Molecular 98 Winfree, E. et al. (1998) Design and self-assembly of two-dimensional
BioSystems 6, 807–814 DNA crystals. Nature 394, 539–544
78 Rinaudo, K. et al. (2007) A universal RNAi-based logic evaluator that 99 Seelig, G. et al. (2006) Enzyme-free nucleic acid logic circuits. Science
operates in mammalian cells. Nat. Biotechnol. 25, 795–801 314, 1585–1588
79 Vinkenborg, J.L. et al. (2011) Aptamers for allosteric regulation. Nat. 100 Zhang, D.Y. et al. (2007) Engineering entropy-driven reactions and
Chem. Biol. 7, 519–527 networks catalyzed by DNA. Science 318, 1121–1125
80 Mutalik, V.K. et al. (2012) Rationally designed families of orthogonal 101 Xie, Z. et al. (2010) Logic integration of mRNA signals by an RNAi-
RNA regulators of translation. Nat. Chem. Biol. 8, 447–454 based molecular computer. Nucleic Acids Res. 38, 2692–2701
81 Schratt, G. (2009) MicroRNAs at the synapse. Nat. Rev. Neurosci. 10, 102 Kato, T. et al. (2009) High-resolution structural analysis of a DNA
842–849 nanostructure by cryoEM. Nano Lett. 9, 2747–2750

633
Review

Special Issue – Synthetic Cell Biology

Towards a bottom-up reconstitution of


bacterial cell division
Ariadna Martos1, Mercedes Jiménez2, Germán Rivas2, and Petra Schwille1
1
Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany
2
Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040-Madrid, Spain

The components of the bacterial division machinery Glossary


assemble to form a dynamic ring at mid-cell that drives Amphitropic proteins: proteins that can exist as either soluble cytoplasmic or
cytokinesis. The nature of most division proteins and membrane-attached forms depending on physiological conditions of activa-
their assembly pathway is known. Our knowledge about tion. Their reversible interaction with membranes seems to be mediated by
specific lipids. This concept was proposed by Paul Burn (1988).
the biochemical activities and protein interactions of Atomic force microscopy (AFM): an imaging and measurement tool that
some key division elements, including those responsible provides a 3D map of a sample’s surface. It allows the collection of topography
for correct ring positioning, has progressed considerably images with resolution in the nanometer scale and, at the same time, maps
other interesting properties of the samples. With the capacity to observe and
during the past decade. These developments, together manipulate biological surfaces under physiological conditions, AFM can be
with new imaging and membrane reconstitution tech- used to explore biological structures at the single-molecule level.
nologies, have triggered the ‘bottom-up’ synthetic ap- Division ring: a multiprotein complex whose presence at mid-cell is derived
from the coincident location of several proteins (e.g., FtsZ, FtsA, ZipA, FtsN),
proach aiming at reconstructing bacterial division in the because they are independently visualized as rings in dividing cells [8].
test tube, which is required to support conclusions Divisome: the complete macromolecular machinery able to effect division in
derived from cellular and molecular analysis. Here, we the living cell. All the proteins that form the division ring at mid-cell are part of
the divisome. Other components such as periplasmic and outer membrane
describe recent advances in reconstituting Escherichia proteins and the cell envelope may also be integral parts of the divisome. The
coli minimal systems able to reproduce essential func- term divisome was proposed by Nanne Nanninga (1998).
Fluorescence recovery after photobleaching (FRAP): the observation and
tions, such as the initial steps of division (proto-ring
measurement of FRAP allows scientists to investigate the diffusion and motion
assembly) and one of the main positioning mechanisms of fluorescently labelled biological macromolecules. The fluorescently tagged
(Min oscillating system), and discuss future perspectives biomolecule of interest is visualized at low light intensity followed by
photobleaching of a user-defined region of interest with high-intensity light,
and experimental challenges. causing the fluorophores to bleach in the selected region. The measure of the
rate of recovery of fluorescence allows one to infer how fast macromolecules
The strategy of constructive (bottom-up) synthetic move.
biology of bacterial cell division Fluorescence resonance energy transfer (FRET): a technique for measuring
interactions between two molecules. It relies on the ability to capture
The aim of synthetic biology is to dissect biological entities fluorescent signals from the interactions of labeled molecules in single living
into functional modules (macromolecular assemblies, cells, or fixed cells. If FRET occurs, the donor channel signal will be quenched and
the acceptor channel signal will be sensitized or increased. This allows
tissues, organisms, and their networks), to modify them
investigation of the colocalization of the donor- and acceptor-labeled probes
rationally, or to engineer entirely new ones with the goal of and the molecular associations at close distances.
implementing novel biomimetic functionalities [1,2]. The Minimal divisome: the theoretical minimal set of elements capable of effecting
division. It can be defined genetically, when using mutants defective in some
bottom-up synthetic biology approach has strong physico- of the divisome elements, or biochemically when using artificial cell envelope
chemical foundations; it is interdisciplinary and has a assemblies in the test tube.
rigorous quantitative character. It aims at constructing Membrane-targeting sequence (mts): among other amphitropic proteins, FtsA
and MinD present a highly conserved C-terminal sequence motif that it is
biological parts, devices, and systems with increasing thought to form an amphipathic helix that mediates the direct interaction of
levels of complexity toward a minimal cell-like scaffold these proteins with lipid bilayers. The mts from MinD was incorporated into
[3–5]. The ‘top-down’ approach relies rather on biotechnol- chimeric FtsZ forms to bypass the need for membrane-anchoring proteins.
Proto-ring: the natural initial subset of the division ring able to be anchored to
ogy and systems biology, seeking primarily the program- the cytoplasmic membrane. In E. coli, it is formed by FtsZ, FtsA, and ZipA and it
ming of novel gene circuits within an electronic and is assumed to be the initial step of divisome assembly [10].
computational engineering framework [6,7]. Total internal reflection fluorescence microscopy (TIRFM): a powerful fluores-
cence microscopy technique that allows imaging but also the measurement of
In the context of bottom-up approaches, the in vitro dynamics and single-molecule events taking place in a thin section (100 nm)
reconstitution of minimal bacterial cell division machinery in the contact surface between the specimen and the glass slide. An
is an obvious and attractive goal. Considering the highly evanescent field propagates into the specimen on total reflection of the
incident beam at the liquid/glass interface. In this way, only the fluorophores
dynamic and membrane-associated nature of this essential close to the surface are excited, thus reducing background fluorescence due to
machine, and the difficulty of assaying its functional reac- out-of-focus molecules.
Z-ring: a structure present at mid-cell of dividing cells and visualized by
tions, it is a challenging prospect. However, in the past few
microscopy techniques using FtsZ-specific reagents, such as fusions to
years, new technologies for imaging and functional protein fluorescent proteins or antibodies against FtsZ. It is postulated to be a ring
of variable diameter formed by the accumulation of FtsZ-containing complexes
Corresponding authors: Rivas, G. (grivas@cib.csic.es); Schwille, P. [8]. The Z- ring has been assumed to provide a scaffold on which the remaining
(schwille@biochem.mpg.de). division elements assemble due to their ability to interact directly or indirectly
Keywords: constructive synthetic biology; cytomimetic biochemistry; macromolecular with FtsZ. The term Z-ring was proposed by Joe Lutkenhaus (1991).
interactions; FtsZ; Min system; Escherichia coli.

634 0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.09.003 Trends in Cell Biology, December 2012, Vol. 22, No. 12
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

reconstitution in membranes have put this goal within The bacterial cell division machinery
reach. Today, we know the main elements, their biological Bacterial cell division is mediated by a multiprotein ma-
function, and a large part of their biochemistry; therefore, chine whose elements interact dynamically at mid-cell to
it seems not unreasonable to expect that the application of form the division ring, a structure that drives cytokinesis,
this bottom-up synthetic strategy to construct minimal forming part of the divisome [8,9]. The position of the ring
divisomes (see Glossary) in the test tube is feasible and is highly regulated by two negative control systems that
will yield functional assemblies. This constructive synthet- inhibit its incorrect formation (see below). In the case of E.
ic approach will help us to support conclusions already coli, the division ring is formed by at least ten essential
derived from cellular and molecular analysis and, there- proteins, most of them integrated at the membrane. Their
fore, to complete our understanding of how the bacterial assembly pathway has been mostly derived from the be-
division machinery works. havior of mutants in which divisome elements have been

A
(a) B
Z K Q W I N
L
Zip

Proto-ring Periplasmic Pepdoglycan


connector factory

Cytoplasmic ring Periplasmic ring

Early assembly Late assembly

FtsA-independent assembly

(b) FtsQ
FtsL FtsB FtsL FtsN
FtsK
Out FtsW

Membrane

In
FtsA ZipA

FtsZ
GDP GTP

Nanodiscs Microbeads Bilayers Vesicles


TRENDS in Cell Biology

Figure 1. Assembly of the essential components of the Escherichia coli division ring. (a) First row: FtsZ interacts with FtsA and ZipA at the cytoplasmic membrane forming
the initial assembly of the division machinery, the proto-ring. For the sake of simplicity other division proteins (such as FtsE/X or Zap proteins) are not shown in this scheme.
FtsK is then added sequentially, followed by a group formed by FtsQ, B and L. FtsW and I (PBP3), two proteins involved in septal peptidoglycan synthesis, are then added
together to the ring, which is completed by addition of FtsN. The rest of the rows describe the functionality and localization of the ring components. Republished with
permission from [10]. (b) Proto-ring in cells and reconstruction of proto-rings in membrane systems. Diagram illustrating the position of essential divisome elements in the
living cell and reconstruction of proto-rings in biomimetic membranes: nanodiscs, microbeads, supported lipid bilayers, and vesicles. The proto-ring components are
colored; other elements of the divisome are shown in dark grey. The light grey circles in the upper part of the diagram represent macromolecules that contribute to the
crowding of the cytoplasm. Vesicles show the two alternative orientations of the reconstructed proto-ring relative to the vesicle lumen.

635
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

genetically altered (Figure 1a) [10]. The first macromolec- localization of FtsZ occurs. Gain-of-function mutants of
ular assembly of the divisome is the proto-ring, comprising FtsA, such as FtsA*, can bypass the need for ZipA for
three proteins – FtsZ, FtsA, and ZipA – that associate at recruitment of downstream proteins to form a division ring
the cytoplasmic membrane forming the scaffold structure [13,19]. Addition of FtsA* to FtsZ should be sufficient, in
into which the rest of the essential division proteins are principle, to form an active, membrane-anchored proto-
incorporated in a sequential and concerted manner ring without ZipA. However, there is no evidence suggest-
(Figure 1a). ing that the behaviors of FtsA* and native FtsA are the
Central to assembly of the division machinery is FtsZ same.
(40 kDa), the best-known prokaryotic division protein,
which is widely conserved in bacteria and the ancestor Spatial modulators of divisome assembly – the
of eukaryotic tubulin [11,12]. Although the basic unit of oscillating Min CDE system
FtsZ is a monomer and the tubulin protomer is a hetero- In E. coli, the positioning of the division ring at mid-cell at
dimer, both proteins bind and hydrolyze GTP and form the end of the cell cycle is regulated by two independent
polymers in a GTP-dependent manner, making FtsZ poly- mechanisms, the Min system and nucleoid occlusion, that
mers very plastic and polymorphic. The GTP-dependent cooperate to target the formation of the ring at the right
FtsZ assembly/disassembly cycle it is thought to be essen- place [9,14,20,21]. The control exerted by the bacterial
tial for Z-ring function [12–14]. Current research on FtsZ is nucleoid (bulk chromosomal DNA), largely due to crowd-
aimed at describing the polymerization and force-genera- ing/electrostatic effects and the action of the protein SlmA
tion mechanisms and evaluating the roles of nucleotide [22], is nevertheless not able to block the formation of polar
exchange and hydrolysis [11,15]. FtsZ is cytosolic and Z-rings, suggesting that the task of restricting septation to
attaches to the membrane though interactions with FtsA mid-cell might significantly rely on the Min system.
or ZipA. FtsA (45 kDa) belongs to the actin family and The Min system comprises MinD, E, and C proteins,
contains a short amphipathic helix that it is thought to which oscillate repeatedly from one pole of the cell to the
mediate its association with the membrane [16]. ZipA (35 other (Figures 2 and 3) [23,24]. A description of the bio-
kDa) contains an amino-terminal domain that is integrat- chemical and molecular details that account for the inter-
ed into the membrane and connected to a cytoplasmic FtsZ- change between membrane-bound and cytoplasmic
interacting domain via a flexible linker [17,18]. The locations of the Min proteins are reviewed elsewhere
attachment of FtsZ to the membrane requires either ZipA [25,26]. Briefly, MinC inhibits FtsZ assembly [24,27,28].
or FtsA, but in the simultaneous absence of both, no MinD, a membrane-bound ATPase member of the Walker

(a) (b) MinE-Cy5 MinD-Cy3

70 µm

(c) (d)

+ATP /-ADP 1
Rear Middle Front
intensity (AU)
Normalized

2
3
4b
5 –P
4a

MinD
MinE
Key: MinD-ADP MinC MinC
MinD-ATP MinE

TRENDS in Cell Biology

Figure 2. Min oscillating system. (a) MinE fluorescence (red) is observed as it pushes the green fluorescent signal of MinD (yellow). Scheme from [78]. (b) Confocal
fluorescence micrographs showing spiral waves of MinD and E in the presence of ATP on a supported lipid bilayer in vitro. (c) Simplified scheme of the biochemical
reactions underlying the oscillating behaviour. ATP-bound MinD binds to the membrane as dimers. Both MinC and MinE can bind to membrane-bound MinD, but MinE is
able to displace MinC from MinD. MinE stimulates the ATPase activity of MinD and, on hydrolysis of ATP, MinD and MinE are released from the membrane [26]. (d)
Normalized fluorescence intensity profiles of Min D, E, and C corresponding to waves equivalent to the ones shown in (b). Intensity profiles reveal the relative position of
the Min proteins within the wave. Black arrows indicate the traveling direction of the protein waves.

636
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

MinE-Cy5
(a)

(b) 173 µm

10 min
173 µm

0 µm

20 µm

TRENDS in Cell Biology

Figure 3. MinD/E waves propagating on the outer surface of giant unilamellar vesicles (GUVs). (a) Sequential confocal images of the equatorial plane of a GUV in which
MinD/E oscillations are observed on its outer surface are shown (images starting on the upper left toward the right side of the panel; taken every 5 s). The waves appear as
an alternate increase and decrease in the fluorescence intensity of Cy5-labeled MinE. (b) Kymograph of the GUV shown in (a). The rim of the vesicle is analyzed as a straight
line and its fluorescence intensity represented over time. This reconstruction closely resembles the kymographs of the waves previously observed in supported lipid
bilayers.

A cytoskeletal ATPase (WACA) family, powers the oscilla- by optical microscopy, and the behavior of macromolecules
tion by ATP consumption [25,29,30]. It activates MinC’s (divisome proteins) trapped in the GUV interior can be
inhibitory activity and recruits it to the membrane, while studied by microspectroscopy techniques [36]. Finally,
MinE activates MinD’s ATPase to detach MinD from the coated microbeads, comprising lipid bilayers deposited
membrane and drive the oscillation [31]. Their combined on solid, spherical surfaces, are a useful novel membrane
action results in FtsZ assembly into a ring at mid-cell, model to reconstitute macromolecular assemblies. They
where the local time-averaged concentration of MinC in- offer a robust procedure to generate lipid surfaces with
hibitor is the lowest. Fusions of the Min proteins with uniform curvatures and cell-like sizes, in which both lipid
fluorescent proteins allow following of these pole-to-pole composition and the material of the bead can be modified to
oscillations in vivo. In this way, another prominent feature obtain the desired properties of the system [3,32].
of the Min system can be observed, the so called ‘E-ring’, Nanodiscs, formed by a membrane scaffold protein
comprising a sharply defined band of MinE that travels encircling a phospholipid mixture [37], are a promising
behind MinD and MinC [23]. novel tool to study assembly on a lipidic surface while the
system under study remains soluble. When a target mem-
The use of biomimetic membranes to reconstruct brane protein is included in the mixture, each nanodisc
minimal divisome assemblies self-assembles and incorporates a single target molecule,
One major advantage of in vitro reconstitution experi- preserving its natural structural and functional properties.
ments in biomimetic membranes is that both biochemical Targets may be single proteins (e.g., ZipA) or even a protein
and biophysical parameters can be controlled precisely, complex (e.g., ZipA or FtsA plus FtsZ). Due to their small
providing an opportunity to investigate the complex spa- size, nanodiscs remain soluble, allowing the use of solution
tial and temporal dynamics of membrane-based processes biochemical and biophysical tools [38,39] to characterize
under defined conditions (Figure 1b and Box 1) [32–34]. the embedded protein and to monitor and measure its
Supported membrane structural organization (e.g., the interactions with other proteins.
presence of membrane microdomains) can be studied in
detail by fluorescence microscopy, but are also perfectly Reconstruction of a minimal divisome in the test tube,
suited to be studied by surface probe techniques such as step-by-step
atomic force microscopy (AFM), to yield nanoscopic resolu- For successful reconstitution of bacterial cell division, two
tion. Employing total internal reflection fluorescence essential phenomena observed in intact cells need to be
(TIRF) as an excitation scheme, the dynamics of single achieved by a minimal protein machinery: (i) the assembly
lipid and protein molecules in membranes can be visual- of the divisome components at discrete regions along the
ized and dynamic interactions of soluble proteins with cell length (this occurs usually at mid-cell in normal bac-
membranes can be resolved [35]. Another model mem- terial division, but might vary in mutants such as those
brane system useful in reconstitution experiments is giant defective in the Min system); and (ii) the function of the
unilamellar vesicles (GUVs), which are closed lipid force-generating elements that initiate constriction of the
bilayers with diameters well above the optical resolution membrane. We will first discuss approaches toward recon-
limit, from several to hundreds of micrometers. Due to stituting the Min positioning system and then touch on the
their large size, they are perfectly suited to investigations perspectives of realizing a contractile proto-ring.
637
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 1. The membrane as an element of the divisome dynamic behaviour displayed in vivo, such that MinE
In the bacterial cell, it is likely that membrane component
accumulates following MinD and MinC toward the rear
redistributions during cell division may create lipid domains [79] of the wave, resembling the E-ring in cells. It was con-
and regions highly concentrated in specific components that can firmed that MinC is not required for the cycling, but
trigger membrane deformations (favouring curved structures) assumes the role of a passenger. Interestingly, the wave-
[80,81] that might contribute to the accurate selection of the division length of the in vitro pattern is 10 times larger than that in
site and to the force generation that drives constriction. In addition,
membrane fluidity might also modulate bacterial division, as
an E. coli cell, which has been proposed to be explained by
suggested by recent observations that increasing fluidity favours crowding effects or higher reaction rates [26], although
cytokinesis of L forms [82] and FtsZ polymers modify the neither has been confirmed experimentally yet.
viscoelastic properties of E. coli lipid monolayers to become more According to single-molecule imaging of Min dynamics
fluid and plastic [83,84]. The composition of a bilayer defines its [26], the patterns emerge from an isotropically distributed
mechanical properties [85]. Three major lipids constitute the inner
membrane of E. coli: phosphatidylethanolamine (74%), phosphati-
carpet of MinD on the membrane [40]. A difference in the
dylglycerol (19%), and cardiolipin (3%). This mixture confers a net membrane residence times between MinD and MinE, with
negative charge to the membrane at physiological pH of 7.5. The MinE being attached longer than MinD, originates small
relevance of the lipid composition takes place at two levels, fluctuations in the MinD/MinE relative ratio, such that
modulating the mechanical properties of the membrane and
accumulation of MinE initiates a dominant wave of protein
offering specificity as recruiting factors. First, enriched domains on
cardiolipin were detected at both the cell poles and the septal region detachment, leading to faster detachment of MinD toward
of dividing E. coli cells [86]. This conical phospholipid tends to form the end of the wave. Fluorescence resonance energy trans-
high-curvature rather than bilayer structures, indicating that its fer (FRET) experiments on these supported bilayers con-
accumulation might provoke mechanical destabilization of the firmed that MinE binds to MinD as a dimer [40].
bilayer thus facilitating morphological transformations of the
Regarding a plausible explanation for the formation of
membrane. Second, its anionic character, together with its marked
distribution in key areas of the membrane during cell division, make the E-ring in vivo, evidence from several experiments
cardiolipin a suitable target for amphitropic proteins such as FtsA points in favor of persistent binding of MinE [43–45] rather
[16]. These differences in the distribution of cardiolipin in the than formation of higher-order complexes [46]. The differ-
membrane along the cell suggest an active role of this lipid in cell ence in residence times between MinD and MinE, together
division. In addition, MinD has been shown to bind preferentially to
domains of anionic phospholipids on the inner membrane [87].
with a decrease in the diffusion coefficient of MinE by a
Related to this, it has been reported that membrane potential might factor of two in the back of the wave, may explain the
be relevant for the binding of MinD and FtsA to the membrane [88]. accumulation of MinE in the rear and therefore the E-ring
The high protein content present on the inner membrane of E. in vivo [40]. Fluorescence recovery after photobleaching
coli, where the lipid/protein ratio is 0.4, is notable [89]. This is likely (FRAP) experiments demonstrated that incorporation of
to have a major effect on the physical properties of the membrane,
because imposition of physical constraints on its plasticity would
MinE starts from the front of the wave, further corrobora-
make it less prone to change. In summary, the restoration of the ting accumulation – in contrast to cooperative binding – of
membrane can also be considered a strategy towards the recon- MinE [26]. Experiments using MinE C1, a mutant unable
struction of bacterial division. Accordingly, the manipulation of to bind to the membrane, revealed that transient mem-
membrane composition in membrane models would contribute to
brane binding of MinE could be partially involved in the
revealing its relationship with the constriction process.
reported persistent binding [43,47,48]. Unlike wild-type
MinE, the fluorescence intensity profiles of the much less
Reconstituting Min oscillations in vitro regular waves based on MinE C1 do not show its accumu-
The Min system has been described as a reaction–diffusion lation at the rear, nor displacement of MinC from mem-
system in which, starting from a homogeneous distribu- brane-bound MinD [40].
tion, the oscillatory patterns emerge by self-organization The mechanistic picture that emerges from the data
on dynamic instability powered by the hydrolysis of ATP. obtained by reconstruction of a minimal Min system is
In fact, these dynamics largely depend on collective mem- summarized in Figure 2. Recently, it has been verified that
brane binding and unbinding. Thus, a crucial step toward a the Min patterns can be equally well established on vesicle
minimal system for Min oscillations was achieved by the membranes (Figure 3). The obvious next step is now to
reconstruction of in vivo cycling behaviour on artificial encapsulate the Min protein dynamics into a closed com-
bilayers mimicking the inner membrane of E. coli partment and convert the propagating waves – which do
[26,40]. In the presence of ATP, protein waves emerged not allow directing Z-ring assembly to a specific site – to
on E. coli lipid bilayers on incubation of purified and true oscillations with a clear minimum in Z-ring assembly
fluorescently labelled Min proteins at physiological ratios inhibition.
[41,42] (Figure 2). In the absence of spatial boundaries,
these waves propagated in arbitrary directions across the Minimal bacterial proto-rings in vesicles
bilayer. Using confocal and single molecule TIRF micros- The activity, interactions, and assembly properties of the
copy, all three proteins were simultaneously imaged and proto-ring elements can be quantitatively characterized in
the order of events during wave propagation could be minimal membrane systems structured as nanodiscs, coat-
determined (Figure 2). ed-microbeads, lipid bilayers, and, finally, vesicles
Furthermore, this in vitro system reproduces some (Figure 1b). The first example of this synthetic approach
relevant features of the in vivo oscillations [40], such as [49] demonstrated the assembly of a purified variant of
a similar speed, about 0.5 mm/s, which depends on the FtsZ ring inside unstable vesicles that spontaneously
concentrations of MinD and MinE [26]. In addition, the changed in shape over time, suggesting that FtsZ is able
obtained distribution profiles are compatible with the to at least partly constrict the region of the liposome in
638
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (I) (II)


C-terminal MTS N-terminal MTS FtsZ / ZipA

Unilamellar liposomes

3 9 15 21
Mullamellar tubular
vesicles

(b)

TRENDS in Cell Biology

Figure 4. Reconstruction of the contractile FtsZ element. (a.I) Reconstruction of chimeric FtsZ (C-terminal or N-terminal mutants) in unilamellar liposomes and tubular
vesicles. FtsZ is able to narrow the region of the liposome in which it is located. Composed image of liposome contour (from phase contrast microscopy) and fluorescence
of chimeric FtsZ colored in yellow [49]. In unilamellar liposomes, chimeric FtsZ deforms the bilayer according to the location of the mutant construct, suggesting that FtsZ
presents an intrinsic angle of polymerization. In tubular vesicles, FtsZ is able to condense into several rings, narrowing the region in which it is located. Adapted from [49].
(a.II) Membrane tubulation of giant vesicles containing ZipA driven by native FtsZ polymers as demonstrated by confocal microscopy using Alexa 488-labeled FtsZ (A.
Martos et al., unpublished). Scale bars = 10 mm. (b) (Left panel) Fluorescence image of copolymers of YFP-mts-FtsZ and FtsZ assembled on glass substrates having curved
grooves. The profile of the surface is shown on the right. (Right panel) Atomic force microscopy image of filaments assembled on a similar substrate. Red arrows show
filaments on planar surfaces and white arrows show filaments on the curved grooves. Scale bars = 1 mm [57].

which it is located (Figure 4a.I). The authors used an force to promote lipid vesicle deformation, although the
artificially membrane-targeted FtsZ variant (membrane- biological implications of this force within the (much more
targeting sequence [mts]-FtsZ) to tether it to the liposome challenging) task of full membrane constriction and cell
in the absence of physiological membrane-anchoring pro- division remain unclear [49–51].
teins. The cylindrical geometry of the tubular vesicles The next step towards proto-ring reconstruction has
seems to be important, because no Z-rings were found been achieved by incorporating the native FtsZ form (in-
inside spherical liposomes. The incorporation of mts-FtsZ stead of the mts-FtsZ variant) together with FtsA inside
polymers into the outside of liposomes produced visible giant vesicles obtained from bacterial inner membranes
deformations (liposome tubulation) [50], an observation (Figure 5). Being more complex in composition than artifi-
recently confirmed using a vesicle reconstruction contain- cial lipid vesicles, they maintain a distribution of proteins
ing natural FtsZ and ZipA (Figure 4a.II) (A. Martos et al., and lipids resembling more closely the native membrane,
unpublished). FtsZ polymer bending (associated with the and contain the essential membrane-anchoring factor ZipA
natural curvature of FtsZ) is thought to be a relevant naturally inserted. FtsZ polymerization alters the spatial
feature in generating the force that promotes membrane distribution of the proto-ring complexes inside the vesicle,
deformation, because the geometry of the distortion resulting in dislodging of FtsA from the membrane to
depends on the FtsZ terminal region at which the mem- associate with FtsZ fibers in the vesicle lumen, showing
brane-targeting sequence is attached. These experiments that, under assembly-promoting conditions, the interac-
proved that FtsZ polymers alone are capable of generating tion between FtsZ monomers is stronger than the binding
639
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

bilayers through ZipA form dynamic 2D networks that


retain their capacity to dynamically restructure, to frag-
ment, to anneal, and to condense laterally [56]. In a recent
approach [57], microstructured substrates to support
membranes were employed to investigate the curvature-
dependent membrane attachment, and thus curvature
preference, of FtsZ rings. It could be shown that Z-rings
(b) comprise pre-curved FtsZ filaments, which preferentially
FtsZ GDP (a) FtsA GDP
orient themselves along curvatures resembling the native
inner diameter of E. coli cells. FtsZ filaments seem to avoid
smaller diameters (resembling constriction stages during
cell division), which can be evidenced by the slanted orien-
tation of filaments in membrane-coated grooves of greater
curvature (Figure 4b). This speaks for the requirement of
additional factors to facilitate constriction during cell divi-
sion and against FtsZ filaments or rings being the sole
force-generation system. However, conclusive direct mea-
surements of forces exerted by FtsZ filaments with or
FtsZ GTP (c) FtsA GTP (d)
without additional divisome proteins, similar to motor
protein assays, are still lacking.

Biochemical features of proto-ring proteins in coated


beads and nanodiscs
Lipid- and membrane-coated beads allow measurement of
the binding affinities of soluble proteins to membrane
structures. A recent example is the incorporation of FtsA
in these structures [58], showing that the apparent affinity
of FtsA to the inner membrane is tenfold higher than to the
Contrast (e) Superimposed
(f) E. coli lipids (without integral proteins). The reconstitution
of the three proto-ring elements in coated beads is feasible,
TRENDS in Cell Biology
because these membrane systems appear to retain their
Figure 5. Reconstruction of proto-ring elements in giant unilamellar vesicles capability to interact with FtsZ (Monterroso et al., unpub-
(GUVs). The figure shows the spatial distribution of FtsZ and FtsA inside GUVs lished). Beads encoded with gold/silver sensor particles
made from inner bacterial membrane. Confocal images of Alexa 488-labeled FtsZ
(green; a and c) and Alexa 647-labeled FtsA (red; b and d), formed in the absence (a
before membrane coating are being exploited to design
and b) and in the presence (c and d) of GTP. The superposition of (c) and (d) can be assays to monitor interactions between proto-ring ele-
observed in (f). Differential interference contrast image of the giant unilamellar ments [59].
inner membrane vesicle (e). Scale bars = 10 mm. Republished with permission
from [52]. The nanodisc technology has recently been exploited to
elucidate biochemical features of elements of the bacterial
division machinery and their interactions under well-de-
strength of FtsA to the membrane [52]. These findings fined lipid environments [38]. A single copy of the full-
suggest that in addition to the known role in the associa- length ZipA protein from E. coli has been incorporated in
tion of FtsZ with the membrane during division, FtsA may phospholipid bilayer nanodiscs. ZipA in nanodiscs binds
have other roles related to its amphitropic character, which FtsZ oligomers (GDP forms) and polymers (GTP forms)
may involve signalling of proto-ring assembly completion equally with moderate affinity, which is also similar to the
or the activation of late divisome functions. Interestingly, binding strength of a soluble form of ZipA to FtsZ, suggest-
FtsA interacts with FtsN [53], a late-assembling protein ing that the transmembrane segment of ZipA has no role in
essential in septation, because its absence results in de- complex formation. These findings have provided new
stabilization of the already assembled divisome, even af- insights into the role of ZipA as a passive anchoring device,
fecting the proto-ring elements [54]. These results show supporting the notion that FtsZ tethering to the membrane
that the interaction between elements of the divisome is far has enough plasticity to allow the remodelling of the
more complex than what could be deduced from the visual architecture of the division ring during constriction.
inspection of the assembly of rings in a living cell. These
approaches can help to compare the in vivo and in vitro Outlook: experimental challenges
systems to obtain new knowledge on the process of division. The bottom-up synthetic approaches described here, to test
the properties of the elements of the division machinery
Structural features of minimal proto-rings on supported and their interactions in well-defined and controllable
and free-standing lipid bilayers environments, have helped us to gain new mechanistic
The structural and dynamic organization of FtsZ polymers insights into their functions. However, regarding possible
has been studied by AFM on mica and on lipid bilayers reconstitution of full bacterial cell division in minimal
containing a soluble variant of ZipA, suggesting that they systems, many more experimental challenges await us.
are very plastic [55,56]. FtsZ polymers anchored to lipid First, the true origin of a constriction force will have to be
640
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

elucidated. A model of action of FtsZ (and the potential Box 2. Numbers in E. coli cell division
influence of companion division proteins) needs to be com- Proto-ring elements
pleted to reconcile the FtsZ force-generating properties in It is estimated that each E. coli cell contains 4000–15 000 FtsZ [90,91],
vitro [60] and the FtsZ dynamics in vivo [12], with their 600–3000 FtsA [90], and 1000–1500 ZipA [90,92] molecules. In the
implication on division. Moreover, the mechanical role of case of FtsZ, this would correspond to an average concentration of
the cytoplasmic membrane (Box 1) and cell wall synthesis approximately 3–10 mM (0.1–0.4 mg/ml). Non-essential division
proteins (e.g., Z-ring-stabilizing elements) are estimated to be less
during septum formation are far from being understood. than 5% of FtsZ numbers [93]. Because FtsZ assembly follows a
Second, the correct positioning of the division site by the cooperative mechanism, it would be distributed into soluble and
Min protein system will have to be verified, by introducing polymeric forms whose relative abundance would be modulated in
the Min system as well as the required proto-ring elements the cell by several parameters, such as protein concentration, other
into deformable closed containers, preferably vesicles. companion proteins, physiological ligands such as magnesium,
potassium or the ratio of GTP to GDP [51,94], and nonspecific
These goals will require the successful introduction of electrostatic and crowding effects (see below). Taking all these
functional proteins into the vesicles at physiological salt factors together, the amount of FtsZ that is part of the Z-ring is
conditions without disrupting them (which has proved to estimated not to be over two-thirds of the total cellular FtsZ [12,94],
be rather complicated at large scales so far) and the which is enough to encircle the cell.
identification of physical preconditions to convert traveling
Min proteins
Min waves into steady oscillations. Progress in current The abundance of MinE molecules per cell has been estimated to be
research toward the optimization of procedures to encap- 1100–1400; MinD is present at 2000 [42] and MinC at about 400 [41].
sulate protein complexes with high yield inside vesicles in
a controlled manner using permeable membranes and The crowded bacterial interior
It is estimated that 20–30% of the bacterial cytoplasm is occupied by
microfluidic technologies will be crucial [61–64]. Moreover,
macromolecules, among them 200–300 mg/ml protein, 75–120 mg/
lipid chemistry and microsystems technology will probably ml RNA, and 11–15 mg/ml DNA [95,96]. These macromolecules
be required to establish vesicle size and shape as tunable inside the cell seem to be (transiently) clustered in replicative,
parameters in reconstitution experiments. In a recent structural, and metabolic networks rather than homogeneously
study, [65] it was demonstrated that Min waves and pat- distributed [97]. The abundance of macromolecules in bacteria
causes volume exclusion effects, both in solution and in mem-
terns already respond quite strongly to the structure of branes. These effects substantially increase the effective concentra-
membranes. In a 2D assay with patterned membranes, it tion and reactivity of the proteins and may modify the extent and
was found that the waves could be directed by sensing the rate of protein–protein and protein–membrane interactions [68,98].
dimensions of membrane patches, without any confine- Because crowding leads to phase separation, it may also modify the
ment in 3D being required. This raises the interesting spatial arrangement of macromolecular networks involved in
essential processes [99].
question of whether the inner surface, rather than the
volume of bacterial cells, provides the spatial cue for divi-
sion-related processes. specific ways of interacting with the membrane as one key
The nucleoid, a highly charged structure that occupies a template for all division-related processes. New and im-
large volume of the intracellular space, may significantly proved biophysical methods need to be introduced to bac-
influence protein interaction networks in the cytoplasm terial cell biology, to allow for a better understanding of
and at the membrane; for example, it has been recently membrane binding and dissociation, such as super-resolu-
shown that large DNA molecules act as an exclusion zone tion and single-molecule imaging, dynamic techniques like
for actin fibers within a microsphere [66]. Therefore, future fluorescence correlation spectroscopy, and plasmonic
reconstitution approaches may need to use environments methods invoking the potential of nano-optics [71,72].
that faithfully reproduce natural crowding and its associ-
ated electrostatic/excluded-volume effects when attempt- Concluding remarks
ing to assemble functional divisomes (Box 2). Research To conclude, the strategy discussed in this review together
toward a better understanding of the physicochemical with complementary bottom-up synthetic approaches
and biochemical properties of highly concentrated hetero- using either cell-free expression systems inside giant vesi-
geneous mixtures of proteins and/or other macromolecules cles [73–75] or bacteria with simplified genetic content will
[67,68] and the production of artificial organelles [69] will provide the best chance for making progress in the syn-
be essential. Along the same lines, the reconstitution of thetic approach toward studying bacterial division. It is
fortifying shells around membranes, mimicking the pepti- fair to say that, with optical technology entering the sub-
doglycan layer, appears to be a highly attractive, albeit microscopic regime [76], there are many fundamental
complicated, mosaic stone in a full mechanical understand- discoveries to be made on bacterial systems that take us
ing. However, cell division studies on so-called L forms – a step closer to the phenomenon of life in its minimal
bacterial cells deprived of their cell walls – argue against a representation. However, the most important steps toward
decisive role of this feature in cell division [70]. Their its fundamental understanding will probably still have to
division, which does not require FtsZ, is far from being come through bottom-up reconstitution [77].
understood.
In addition to these fundamental questions, nearly all of Acknowledgments
the subsystems discussed above, such as the Min and Fts We thank Professor Miguel Vicente (CNB-CSIC) for useful comments and
especially for sharing with us unpublished material and contributing the
proteins, deserve a more quantitative study of their bio- cell cycle definitions in the Glossary. We also thank the anonymous
chemical properties to better understand and modulate reviewers for useful remarks. Work in the authors’ laboratories is
their functional reactivity. Of particular interest are their supported by Human Frontiers Science Program through grant

641
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

RGP0050/2010-C102 (to G.R. and P.S.), by the Spanish Government 27 Hu, Z. et al. (1999) The MinC component of the division site selection
through grant BIO2011-C03-03 (to G.R.), by the European Commission system in Escherichia coli interacts with FtsZ to prevent
though contract HEALTH-F3-2009-223432 (to G.R.), and by the DFG polymerization. Proc. Natl. Acad. Sci. U.S.A. 96, 14819–14824
Leibniz Prize (to P.S.). We apologize to the authors whose work we were 28 Dajkovic, A. et al. (2008) MinC spatially controls bacterial cytokinesis
not able to include in this review owing to space limitations. by antagonizing the scaffolding function of FtsZ. Curr. Biol. 18, 235–
244
References 29 Raskin, D.M. and de Boer, P.A. (1997) The MinE ring: an FtsZ-
1 Endy, D. (2005) Foundations for engineering biology. Nature 438, 449– independent cell structure required for selection of the correct
453 division site in E. coli. Cell 91, 685–694
2 de Lorenzo, V. and Danchin, A. (2008) Synthetic biology: discovering 30 Zhou, H. and Lutkenhaus, J. (2003) Membrane binding by MinD
new worlds and new words. EMBO Rep. 9, 822–827 involves insertion of hydrophobic residues within the C-terminal
3 Liu, A.P. and Fletcher, D.A. (2009) Biology under construction: in vitro amphipathic helix into the bilayer. J. Bacteriol. 185, 4326–4335
reconstitution of cellular function. Nat. Rev. Mol. Cell Biol. 10, 644– 31 Hu, Z. and Lutkenhaus, J. (2001) Topological regulation of cell division
650 in E. coli spatiotemporal oscillation of MinD requires stimulation of its
4 Chiarabelli, C. et al. (2012) Approaches to chemical synthetic biology. ATPase by MinE and phospholipid. Mol. Cell 7, 1337–1343
FEBS Lett. 586, 2138–2145 32 Arumugam, S. et al. (2011) Protein-membrane interactions: the virtue
5 Schwille, P. and Diez, S. (2009) Synthetic biology of minimal systems. of minimal systems in systems biology. Wiley Interdiscip. Rev. Syst.
Crit. Rev. Biochem. Mol. Biol. 44, 223–242 Biol. Med. 3, 269–280
6 Purnick, P.E. and Weiss, R. (2009) The second wave of synthetic 33 Loose, M. and Schwille, P. (2009) Biomimetic membrane systems to
biology: from modules to systems. Nat. Rev. Mol. Cell Biol. 10, 410–422 study cellular organization. J. Struct. Biol. 168, 143–151
7 Nandagopal, N. and Elowitz, M.B. (2011) Synthetic biology: integrated 34 Vogel, S.K. and Schwille, P. (2012) Minimal systems to study
gene circuits. Science 333, 1244–1248 membrane-cytoskeleton interactions. Curr. Opin. Biotechnol. 23,
8 Vicente, M. et al. (2006) Septum enlightenment: assembly of bacterial 758–765
division proteins. J. Bacteriol. 188, 19–27 35 Garcia-Saez, A.J. and Schwille, P. (2010) Surface analysis of membrane
9 Wu, L.J. and Errington, J. (2011) Nucleoid occlusion and bacterial cell dynamics. Biochim. Biophys. Acta 1798, 766–776
division. Nat. Rev. Microbiol. 10, 8–12 36 Garcia-Saez, A.J. et al. (2010) Fluorescence correlation spectroscopy for
10 Vicente, M. and Rico, A.I. (2006) The order of the ring: assembly of the study of membrane dynamics and organization in giant
Escherichia coli cell division components. Mol. Microbiol. 61, 5–8 unilamellar vesicles. Methods Mol. Biol. 606, 493–508
11 Mingorance, J. et al. (2010) Strong FtsZ is with the force: mechanisms 37 Ritchie, T.K. et al. (2009) Chapter 11 - Reconstitution of membrane
to constrict bacteria. Trends Microbiol. 18, 348–356 proteins in phospholipid bilayer nanodiscs. Methods Enzymol. 464,
12 Erickson, H.P. et al. (2010) FtsZ in bacterial cytokinesis: cytoskeleton 211–231
and force generator all in one. Microbiol. Mol. Biol. Rev. 74, 504– 38 Hernandez-Rocamora, V.M. et al. (2012) Dynamic interaction of the
528 Escherichia coli cell division ZipA and FtsZ proteins evidenced in
13 Pichoff, S. et al. (2012) FtsA mutants impaired for self-interaction nanodiscs. J. Biol. Chem. 287, 30097–30104
bypass ZipA suggesting a model in which FtsA’s self-interaction 39 Nath, A. et al. (2010) Single-molecule fluorescence spectroscopy
competes with its ability to recruit downstream division proteins. using phospholipid bilayer nanodiscs. Methods Enzymol. 472, 89–
Mol. Microbiol. 83, 151–167 117
14 Adams, D.W. and Errington, J. (2009) Bacterial cell division: assembly, 40 Loose, M. et al. (2008) Spatial regulators for bacterial cell division self-
maintenance and disassembly of the Z ring. Nat. Rev. Microbiol. 7, 642– organize into surface waves in vitro. Science 320, 789–792
653 41 Szeto, T.H. et al. (2001) The dimerization function of MinC resides in a
15 Salvarelli, E. et al. (2011) Independence between GTPase active sites in structurally autonomous C-terminal domain. J. Bacteriol. 183, 6684–
the Escherichia coli cell division protein FtsZ. FEBS Lett. 585, 3880– 6687
3883 42 Shih, Y.L. et al. (2002) Division site placement in E.coli: mutations that
16 Pichoff, S. and Lutkenhaus, J. (2005) Tethering the Z ring to the prevent formation of the MinE ring lead to loss of the normal midcell
membrane through a conserved membrane targeting sequence in arrest of growth of polar MinD membrane domains. EMBO J. 21, 3347–
FtsA. Mol. Microbiol. 55, 1722–1734 3357
17 Hale, C.A. and de Boer, P.A. (1997) Direct binding of FtsZ to ZipA, an 43 Park, K.T. et al. (2011) The Min oscillator uses MinD-dependent
essential component of the septal ring structure that mediates cell conformational changes in MinE to spatially regulate cytokinesis.
division in E. coli. Cell 88, 175–185 Cell 146, 396–407
18 Ohashi, T. et al. (2002) Structural evidence that the P/Q domain of ZipA 44 Derr, J. et al. (2009) Self-organization of the MinE protein ring in
is an unstructured, flexible tether between the membrane and the C- subcellular Min oscillations. Phys. Rev. E: Stat. Nonlin. Soft Matter
terminal FtsZ-binding domain. J. Bacteriol. 184, 4313–4315 Phys. 80, 011922
19 Geissler, B. et al. (2007) The ftsA* gain-of-function allele of Escherichia 45 Meacci, G. and Kruse, K. (2005) Min-oscillations in Escherichia coli
coli and its effects on the stability and dynamics of the Z ring. induced by interactions of membrane-bound proteins. Phys. Biol. 2, 89–
Microbiology 153, 814–825 97
20 Vats, P. et al. (2009) The dynamic nature of the bacterial cytoskeleton. 46 Kang, G.B. et al. (2010) Crystal structure of Helicobacter pylori MinE, a
Cell. Mol. Life Sci. 66, 3353–3362 cell division topological specificity factor. Mol. Microbiol. 76, 1222–
21 Lutkenhaus, J. (2007) Assembly dynamics of the bacterial MinCDE 1231
system and spatial regulation of the Z ring. Annu. Rev. Biochem. 76, 47 Arjunan, S.N. and Tomita, M. (2010) A new multicompartmental
539–562 reaction-diffusion modeling method links transient membrane
22 Bernhardt, T.G. and de Boer, P.A. (2005) SlmA, a nucleoid-associated, attachment of E. coli MinE to E-ring formation. Syst. Synth. Biol. 4,
FtsZ binding protein required for blocking septal ring assembly over 35–53
chromosomes in E. coli. Mol. Cell 18, 555–564 48 Hsieh, C.W. et al. (2010) Direct MinE-membrane interaction
23 Raskin, D.M. and de Boer, P.A. (1999) Rapid pole-to-pole oscillation of a contributes to the proper localization of MinDE in E. coli. Mol.
protein required for directing division to the middle of Escherichia coli. Microbiol. 75, 499–512
Proc. Natl. Acad. Sci. U.S.A. 96, 4971–4976 49 Osawa, M. et al. (2008) Reconstitution of contractile FtsZ rings in
24 Raskin, D.M. and de Boer, P.A. (1999) MinDE-dependent pole-to-pole liposomes. Science 320, 792–794
oscillation of division inhibitor MinC in Escherichia coli. J. Bacteriol. 50 Osawa, M. et al. (2009) Curved FtsZ protofilaments generate bending
181, 6419–6424 forces on liposome membranes. EMBO J. 28, 3476–3484
25 Lutkenhaus, J. (2008) Min oscillation in bacteria. Adv. Exp. Med. Biol. 51 Osawa, M. and Erickson, H.P. (2011) Inside-out Z rings–constriction
641, 49–61 with and without GTP hydrolysis. Mol. Microbiol. 81, 571–579
26 Loose, M. et al. (2011) Protein self-organization: lessons from the Min 52 Jimenez, M. et al. (2011) Reconstitution and organization of
system. Annu. Rev. Biophys. 40, 315–336 Escherichia coli proto-ring elements (FtsZ and FtsA) inside giant

642
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

unilamellar vesicles obtained from bacterial inner membranes. J. Biol. 76 Li, Z. et al. (2007) The structure of FtsZ filaments in vivo suggests a
Chem. 286, 11236–11241 force-generating role in cell division. EMBO J. 26, 4694–4708
53 Busiek, K.K. et al. (2012) The early divisome protein FtsA interacts 77 Alberts, B. (2011) A grand challenge in biology. Science 333, 1200
directly through its 1c subdomain with the cytoplasmic domain of the 78 Meinhardt, H. and de Boer, P.A. (2001) Pattern formation in
late divisome protein FtsN. J. Bacteriol. 194, 1989–2000 Escherichia coli: a model for the pole-to-pole oscillations of Min
54 Rico, A.I. et al. (2010) Role of Escherichia coli FtsN protein in the proteins and the localization of the division site. Proc. Natl. Acad.
assembly and stability of the cell division ring. Mol. Microbiol. 76, 760– Sci. U.S.A. 98, 14202–14207
771 79 Mileykovskaya, E. and Dowhan, W. (2005) Role of membrane lipids in
55 Mateos-Gil, P. et al. (2012) Depolymerization dynamics of individual bacterial division-site selection. Curr. Opin. Microbiol. 8, 135–142
filaments of bacterial cytoskeletal protein FtsZ. Proc. Natl. Acad. Sci. 80 Huang, K.C. et al. (2006) A curvature-mediated mechanism for
U.S.A. 109, 8133–8138 localization of lipids to bacterial poles. PLoS Comput. Biol. 2, e151
56 Mateos-Gil, P. et al. (2012) FtsZ polymers bound to lipid bilayers 81 Norris, V. et al. (2004) A hypothesis to explain division site selection in
through ZipA form dynamic two dimensional networks. Biochim. Escherichia coli by combining nucleoid occlusion and Min. FEBS Lett.
Biophys. Acta 1818, 806–813 561, 3–10
57 Arumugam, S. and Schwille, P. (2012) Surface topology engineering of 82 Mercier, R. et al. (2012) Crucial role for membrane fluidity in
membranes for mechanical investigation of tubulin homologue FtsZ. proliferation of primitive cells. Cell Rep. 1, 417–423
Angew. Chem. Int. Ed. Engl. 51, 1–6 83 Lopez-Montero, I. et al. (2010) Lipid domains and mechanical plasticity
58 Martos, A. et al. (2012) Isolation, characterization and lipid-binding of Escherichia coli lipid monolayers. Chem. Phys. Lipids 163, 56–63
properties of the recalcitrant FtsA division protein from Escherichia 84 Lopez-Montero, I. et al. (2012) Active membrane viscoelasticity by the
coli. PLoS ONE 7, e39829 bacterial FtsZ-division protein. Langmuir 28, 4744–4753
59 Ahijado-Guzman, R. et al. (2012) Surface-enhanced raman scattering- 85 Janmey, P.A. and Kinnunen, P.K. (2006) Biophysical properties of
based detection of the interactions between the essential cell division lipids and dynamic membranes. Trends Cell Biol. 16, 538–546
FtsZ protein and bacterial membrane elements. ACS Nano 6, 7514– 86 Mileykovskaya, E. and Dowhan, W. (2000) Visualization of
7520 phospholipid domains in Escherichia coli by using the cardiolipin-
60 Erickson, H.P. (2009) Modeling the physics of FtsZ assembly and force specific fluorescent dye 10-N-nonyl acridine orange. J. Bacteriol.
generation. Proc. Natl. Acad. Sci. U.S.A. 106, 9238–9243 182, 1172–1175
61 Abkarian, M. et al. (2011) Continuous droplet interface crossing 87 Mazor, S. et al. (2008) Mutual effects of MinD-membrane interaction: I.
encapsulation (cDICE) for high throughput monodisperse vesicle Changes in the membrane properties induced by MinD binding.
design. Soft Matter 7, 4610–4614 Biochim. Biophys. Acta 1778, 2496–2504
62 Richmond, D.L. et al. (2011) Forming giant vesicles with controlled 88 Strahl, H. and Hamoen, L.W. (2010) Membrane potential is important for
membrane composition, asymmetry, and contents. Proc. Natl. Acad. bacterial cell division. Proc. Natl. Acad. Sci. U.S.A. 107, 12281–12286
Sci. U.S.A. 108, 9431–9436 89 Devaux, P.F. and Seigneuret, M. (1985) Specificity of lipid-protein
63 Stachowiak, J.C. et al. (2009) Inkjet formation of unilamellar lipid interactions as determined by spectroscopic techniques. Biochim.
vesicles for cell-like encapsulation. Lab Chip 9, 2003–2009 Biophys. Acta 822, 63–125
64 Takiguchi, K. et al. (2011) Transformation of actoHMM assembly 90 Rueda, S. et al. (2003) Concentration and assembly of the division ring
confined in cell-sized liposome. Langmuir 27, 11528–11535 proteins FtsZ, FtsA, and ZipA during the Escherichia coli cell cycle. J.
65 Schweizer, J. and Schwille, P. (2012) Geometry sensing by self- Bacteriol. 185, 3344–3351
organized protein patterns. Proc. Natl. Acad. Sci. U.S.A. 109, 91 Lu, C. and Erickson, H.P. (1999) The straight and curved conformation
15283–15288 of FtsZ protofilaments-evidence for rapid exchange of GTP into the
66 Negishi, M. et al. (2010) Cooperation between giant DNA molecules curved protofilament. Cell Struct. Funct. 24, 285–290
and actin filaments in a microsphere. Phys. Rev. E Stat. Nonlin. Soft 92 Hale, C.A. and de Boer, P.A. (2002) ZipA is required for recruitment of
Matter Phys. 81, 051921 FtsK, FtsQ, FtsL, and FtsN to the septal ring in Escherichia coli. J.
67 Long, M.S. et al. (2005) Dynamic microcompartmentation in synthetic Bacteriol. 184, 2552–2556
cells. Proc. Natl. Acad. Sci. U.S.A. 102, 5920–5925 93 Gueiros-Filho, F.J. and Losick, R. (2002) A widely conserved bacterial
68 Zhou, H.X. et al. (2008) Macromolecular crowding and confinement: cell division protein that promotes assembly of the tubulin-like protein
biochemical, biophysical, and potential physiological consequences. FtsZ. Genes Dev. 16, 2544–2556
Annu. Rev. Biophys. 37, 375–397 94 Gonzalez, J.M. et al. (2003) Essential cell division protein FtsZ
69 Marguet, M. et al. (2012) Polymersomes in ‘‘gelly’’ polymersomes: assembles into one monomer-thick ribbons under conditions
toward structural cell mimicry. Langmuir 28, 2035–2043 resembling the crowded intracellular environment. J. Biol. Chem.
70 Leaver, M. et al. (2009) Life without a wall or division machine in 278, 37664–37671
Bacillus subtilis. Nature 457, 849–853 95 Zimmerman, S.B. and Trach, S.O. (1991) Estimation of macromolecule
71 Baciu, C.L. et al. (2008) Protein-membrane interaction probed by single concentrations and excluded volume effects for the cytoplasm of
plasmonic nanoparticles. Nano Lett. 8, 1724–1728 Escherichia coli. J. Mol. Biol. 222, 599–620
72 Ament, I. et al. (2012) Single unlabeled protein detection on individual 96 Record, M.T., Jr et al. (1998) Biophysical compensation mechanisms
plasmonic nanoparticles. Nano Lett. 12, 1092–1095 buffering E. coli protein-nucleic acid interactions against changing
73 Pereira de Souza, T. et al. (2009) The minimal size of liposome-based environments. Trends Biochem. Sci. 23, 190–194
model cells brings about a remarkably enhanced entrapment and 97 Vendeville, A. et al. (2011) An inventory of the bacterial
protein synthesis. Chembiochem 10, 1056–1063 macromolecular components and their spatial organization. FEMS
74 Maeda, Y. et al. (2012) Assembly of MreB filaments on liposome Microbiol. Rev. 35, 395–414
membranes: a synthetic biology approach. ACS Synth. Biol. 1, 53–59 98 Rivas, G. et al. (2004) Life in a crowded world. EMBO Rep. 5, 23–27
75 Noireaux, V. et al. (2011) Development of an artificial cell, from self- 99 Walter, H. and Brooks, D.E. (1995) Phase separation, due to
organization to computation and self-reproduction. Proc. Natl. Acad. macromolecular crowding, is the basis for microcompartmentation.
Sci. U.S.A. 108, 3473–3480 FEBS Lett. 361, 135–139

643
Review

Special Issue – Synthetic Cell Biology

Engineered, harnessed, and hijacked:


synthetic uses for cytoskeletal systems
Brian S. Goodman, Nathan D. Derr, and Samara L. Reck-Peterson
Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA

Synthetic biology re-imagines existing biological systems be located in the cell periphery and minus ends near the
by designing and constructing new biological parts, nucleus (Box 1, Figure Ia,b). The dynamic growth and
devices, and systems. In the arena of cytoskeleton-based shrinkage of both actin filaments and microtubules is capa-
transport, synthetic approaches are currently used in two ble of exerting pushing and pulling forces [9]. Large, highly
broad ways. First, molecular motors are harnessed for polarized cells, such as neurons or epithelial cells, are
non-physiological functions in cells. Second, transport especially reliant on the cytoskeleton and cytoskeletal trans-
systems are engineered in vitro to determine the biophys- port for maintaining spatial and temporal localization of
ical rules that govern motility. These rules are then ap- intracellular components [10,11], as evidenced by the grow-
plied to synthetic nanotechnological systems. We review ing list of human diseases resulting from defects in cyto-
recent advances in both of these areas and conclude by skeleton-mediated transport [12].
discussing future directions in engineering the cytoskele- Most naturally occurring cytoskeletal motors move uni-
ton and its motors for transport. directionally along either actin filaments or microtubules.
Dynein and kinesin motors move on microtubules, whereas
Harnessing the cytoskeleton myosin motors move on actin filaments (Box 1, Figure Ic,d).
The eukaryotic cytoskeleton is composed of fibers that For the purposes of synthetic biology, the most useful motors
exert and respond to force and serve as highways for may be those that are capable of moving cargo processively
motors. In cells this cytoskeleton performs an array of over long distances because these are capable of individually
functions that include organizing subcellular compart- producing prolonged movements of cargos and/or filaments,
ments, transporting diverse cargos [1], and producing in contrast to non-processive motors which require larger
unique functional structures such as the mitotic spindle arrays to produce similar results. Of the approximately 100
[2,3] and cilia [4,5]. Although synthetic approaches to genes coding for cytoskeletal motors, at least 20 of their
harness and engineer the cytoskeleton are in the early protein products are capable of moving multiple types of
stages, recent work utilizes the diversity of mechanochem- intracellular cargo over long distances [1]. For microtubules,
ical attributes of both the cytoskeletal motors and their these cargo-transporting motors include the minus-end-di-
tracks. In this review we first introduce the key compo- rected cytoplasmic dyneins-1 and -2 [13,14], and the plus-
nents of the cytoskeleton. We then discuss recent endea- end-directed kinesins-1, -2, and -3 [15]. For actin filaments,
vors to employ these components for novel synthetic the plus-end-directed class V myosins are the best-charac-
purposes. Next, we highlight how engineering molecular terized cargo-transporting motors [16]. Many organisms
motors is providing a modular toolbox for synthetic in vitro have expanded on the types of kinesins and myosins through
transport [6–8]. Finally, we review synthetic transport gene duplication and divergent evolution. Although some of
systems composed of either cytoskeletal motors that occur these motors do not function as long-distance cargo trans-
in nature (‘natural motors’), modified natural motors, or porters endogenously, their range of biophysical properties
synthetic motors. We conclude by describing promising could be useful for synthetic purposes. For example, there
future directions in the synthetic biology of transport. are classes of kinesins and myosins that move in reverse
compared to most of their other family members: kinesin-
The parts list: tracks, motors, and cargos 14s are minus-end-directed kinesins [15,17] and class VI
Long-distance transport within eukaryotic cells occurs on myosins are minus-end-directed myosins [18].
two cytoskeletal track systems: actin filaments and micro- Cargos of motor proteins vary widely in size, shape, and
tubules (Box 1, Figure I). Both tracks are polarized, with fast function, and they include membranous organelles, protein-
growing ends (termed ‘plus’ ends) and slow growing ends aceous signaling molecules, and ribonucleoproteins. For
(termed ‘minus’ ends). Generally, dynamic actin filaments synthetic biology uses it is instructive to understand how
are found near the cell periphery, often with their plus ends motors attach to cargo such that those attachment mecha-
polymerizing near the plasma membrane. Microtubules are nisms can be hijacked or mimicked. Although the molecular
usually nucleated from a perinuclear-organizing center and connections linking natural motors to their physiological
emanate from this point; thus, microtubule plus ends tend to cargos are only beginning to be defined, they include both
Corresponding author: Reck-Peterson, S.L. (reck-peterson@hms.harvard.edu).
protein and lipid receptors [19,20]. The filaments them-
Keywords: dynein; kinesin; myosin; actin; microtubule; motor. selves can also be cargos because motors can slide filaments
644 0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.09.005 Trends in Cell Biology, December 2012, Vol. 22, No. 12
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 1. Cellular tracks and motors


(a) In many mammalian cells, actin filaments form a meshwork microtubule-based motors are kinesin-1, -2, and -3 (plus-end-directed)
throughout the cell and near the cell membrane, whereas micro- and cytoplasmic dynein (minus-end-directed). Most kinesins and
tubules are organized radially and emit outward from a perinuclear dyneins are homodimers of motor-containing subunits. Cytoplasmic
microtubule-organizing center. (b) In mammalian neurons microtu- dynein contains a number of additional subunits that play a role in
bules are similarly polarized in axons, but have mixed polarity in cargo binding. Kinesin-14 is a minus-end-directed kinesin. (d)
dendrites. (c) Microtubules are polymers of a- and b-tubulin Filamentous actin (F-actin) is a polymer of globular actin (G-actin),
heterodimers. They are composed of 13–15 protofilaments (linear and rapid polymerization in cells occurs primarily at the plus end and
arrays of tubulin dimers) and can be many microns in length. In cells, depolymerization at the minus end. Myosin-V moves along actin
they predominately polymerize and depolymerize from their dynamic filaments toward the plus end, whereas myosin-VI moves toward the
plus ends near the cell cortex. The major cargo-transporting minus end.

(a) (b)
Plus Dendrite
Minus end
end

Acn Minus end

Microtubules

Plus end

Axon
(c)
Microtubule-based transport
Kinesin-1 Cytoplasmic dynein

Kinesin-14
Plus
end
Minus
end

Microtubule

(d)
Acn-based transport

Myosin-V Myosin-VI
Minus Plus
end end

Filamentous acn
TRENDS in Cell Biology

Figure I. Cellular tracks and motors.

with respect to one another or relative to a fixed position in Pathogens: model hijackers of cytoskeletal systems
the cell [21]. Thus, motors and the dynamic tracks they Pathogenic organisms have already evolved multiple
propel are versatile building blocks for engineering synthet- mechanisms for harnessing the power of the cytoskeleton.
ic systems. Here we discuss examples wherein viruses or bacteria
645
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

co-opt the microtubule or actin cytoskeleton of infected mimic the dynamic and versatile connections that nature
cells to propagate their pathogenic life cycle. Understand- has provided. Such systems would allow the more complete
ing these mechanisms can provide synthetic biologists with integration of artificial transport with endogenous process-
inspiration for re-engineering the cytoskeleton. es. In addition, synthetic in vivo cargo systems could be
Many viruses hijack the microtubule cytoskeleton at utilized to function in response to internal cues, a feature
two major stages of their life cycle. Invading viruses use that current attachment systems cannot yet provide.
dynein to reach the nucleus, where they replicate. Later in
the infection kinesin is used to reach the cell membrane, Harnessing transport systems to perform non-
where viral budding and exit occurs [22]. Considerable physiological functions
progress has been made in identifying the specific virus– A major goal of synthetic biology as it relates to transport is
motor interactions required for these transport events, to harness existing cellular transport systems for novel
creating a useful resource for synthetic biologists. Inter- purposes, such as delivering recombinant DNA to the
estingly, viruses have evolved a variety of mechanisms for nucleus, targeting intracellular organelles to new loca-
recruiting dynein or kinesin [23]. For example, dynein is tions, and mapping neuronal connectivity.
recruited by adenovirus via interactions between the dy- In a process similar to an invading virus navigating the
nein light intermediate and intermediate chains and a cytoplasm to reach the nucleus, effective synthetic delivery
viral capsid subunit termed hexon [24]. This interaction of recombinant DNA to the cell nucleus is a crucial hurdle
is pH-sensitive, providing an elegant mechanism for en- to overcome for many applications of gene manipulation. A
suring that hexon-coated adenoviruses only associate with goal for synthetic biologists is to design a modular system
dynein after entering the low-pH environment of the lyso- capable of bypassing all barriers from cell entry through to
somal compartment. By contrast, herpes simplex virus transcription of the recombinant DNA. The first experi-
(HSV) interacts with dynein via an interaction between mental attempts to achieve this linked plasmid DNA to the
the dynein light chains RP3/TcTex1 and its capsid protein, microtubule cytoskeleton [33]. Although engineered viral
VP26 [25]. In the case of kinesin, vaccinia virus binds to pathogens are commonly used as vectors, they pose signif-
kinesin-1 via an interaction with the kinesin light chain icant safety risks, making non-viral methods largely pref-
[26], whereas HSV binds directly to kinesin via a region of erable [34]. Taking cues from viruses, recent work has
the kinesin heavy chain [27]. These diverse viral binding sought to deliver DNA to the nucleus by coupling DNA
and recruiting mechanisms highlight methods by which to a dynein-recruiting element [35,36]. For example, re-
motors could be co-opted for other non-physiological func- combinant dynein light chain (LC8) linked to a DNA-
tions. binding domain can interact with plasmid DNA; transfec-
In addition to using the cytoskeletal motors, some tions in the presence of this reagent are more efficient
pathogens hijack the polymerization power of the cytoskel- compared to common transfection reagents or naked DNA
etal filaments [28]. The best-characterized example of this [35]. Here, a likely barrier to achieving higher transfection
is the bacterium Listeria monocytogenes, which recruits the efficiency is the ability of the plasmid DNA–dynein light
actin polymerization process to propel itself rapidly chain complex to escape membrane-bound endosomes [37].
through the cytoplasm at speeds of up to 30 nm/s. When A challenge for the future will be to integrate motor
the pathogen hits the cell cortex, actin polymerization recruitment with other steps required to increase the
generates enough force for the host cell to drive itself into efficiency of nuclear delivery, such as the ability to escape
an adjacent cell in a finger-like projection, which is then endosomes and import across nuclear pores.
phagocytosed by that neighboring cell. This process mimics Another promising synthetic application uses actin- or
non-pathogenic lamellipodia formation in which actin net- microtubule-based motors to target existing intracellular
works produce membrane protrusion and ruffling at the cargo to new locations. This causes spatial sequestering of
leading edge of a motile cell [29]. Other pathogens such as cargo, resulting in either blocked or modified functions. Two
Clostridium difficile use toxins to alter microtubule dy- recent proofs of concept showed that synthetic recruitment
namics, causing large microtubule-filled projections to em- of motors to peroxisomal organelles could drive organelle
anate out from the cell. These projections entangle the redistribution [31,32]. In the first study [32], opposite polar-
extracellular pathogen, and are thus hypothesized to help ity microtubule- (cytoplasmic dynein and kinesin-1 or kine-
it adhere to the cell [28,30]. These distinct mechanisms sin-3) or actin- (myosin-V and myosin-VI) based motors were
demonstrate the malleability of the cytoskeletal compo- recruited to peroxisomes in a drug-dependent manner using
nents to be subverted for many non-physiological purposes the FKBP–FRB system [38] (Figure 1a). The recruitment of
and can provide insight for designing novel synthetic each type of motor yielded a different peroxisomal localiza-
systems. tion pattern in fibroblasts. In the second study, similar
These examples highlight how understanding the mech- results were obtained in hippocampal neurons [31]. Here,
anisms that pathogens use to co-opt the cytoskeleton may axon-specific cargo was redistributed to dendrites by
provide inspiration for synthetic designs. Although purely recruiting dynein via the FKBP–FRB system [31]. This
synthetic attachment methods using standard connection approach is both rapid and reversible; organelle redistribu-
technologies have been employed recently [31,32], endoge- tion occurs on a timescale of minutes and can be reversed by
nous and virus-based attachments have the added benefits drug removal. Finally, the modularity of the FKBP–FRB
of being sensitive to endogenous regulation and cellular system can also be used to drive dynein activation in cells
conditions. If these mechanisms are better understood, through motor dimerization [31], which is necessary for
future synthetic attachment schemes could more closely motor processivity [39] (Figure 1b). In future experiments,
646
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) RapalogA Cargo


localizaon
FKBP* domain
element
RapalogB

FRB domain Cargo


protein
FKBP domain

(b)

RapalogA

(c)

R
RapalogB

(d)

RapalogA RapalogB

TRENDS in Cell Biology

Figure 1. FKBP/FRB motor recruitment system. (a) The FKBP–FRB system is a drug-induced, protein dimerization system used by several different groups to control motor
properties and attachments to cargo [32,39,47]. A mutant form of FKBP, referred to as FKBP*, homodimerizes in the presence of the small rapamycin analogue (AP20187)
referred to as RapalogA [31]. In addition, heterodimers can be produced using a different mutant FKBP and the protein domain FRB in the presence of the small molecule,
RapalogB (AP21967) [96]. (b) Fusing FKBP* to two dynein monomers can produce an inducible dynein dimer. This construction technique can be used to activate dynein-
based transport by enabling processive stepping in the presence of RapalogA. (c) Fusing FKBP to a cargo of interest through a cargo localization element (e.g. a PEX domain
for peroxisome localization), allows recruitment of a recombinant FRB–motor to the cargo in an inducible fashion upon the addition of RapalogB. (d) The orthogonal nature
of this system provides the ability to combine their activities for novel complex uses. For example, a protein of interest (tan circle) fused to several copies of FKBP* and a
single copy of FKBP could be selectively aggregated and sequestered by the addition of RapalogA, and subsequently delivered to a site of interest by recruiting a
recombinant FRB–motor by the addition of RapalogB. These strategies (panels a–d) were successfully used to study trafficking in neurons; aspects of this figure were
adapted from [31].

the FKBP–FRB system could also allow inducible spatial Recruitment of motor proteins can also be used as a tool
control of protein, RNA, or organellar cargo through the to harness intracellular sorting mechanisms, allowing
recruitment of endogenous cytoskeletal motors mapping of neural circuits [41]. Many viruses that infect
(Figure 1c,d). Constitutive methods for co-opting motors neurons, such as rabies virus, spread unidirectionally
have also been developed. By generating a myosin–kinesin across synapses from the peripheral nervous system to
chimera containing the motor domain of a kinesin and the the central nervous system. This type of cell-to-cell infec-
cargo-binding domain of a myosin, fission yeast cells were tion requires navigation of the microtubule network within
rewired to use microtubules in place of actin filaments [40]. neurons. Recently, it was demonstrated that glycoproteins
These studies highlight the versatility of motors to move from different viruses were sufficient to provide directional
non-physiological cargo and provide promise that additional viral particle movement [42]. This was achieved by engi-
innovative uses for synthetic technology will be discovered neering vesicular stomatitis virus (VSV) with glycopro-
in the future. teins from viruses known to move either towards or
647
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

away from the neuron cell body [42]. A current missing link Interchangeable parts for molecular transport machines
is to determine how these viral glycoproteins differentially Although engineering the cytoskeleton in cells for synthet-
recruit dynein and kinesin motors, and which specific iso- ic purposes is still in its infancy, significant work has been
forms they use. These experiments demonstrate the feasi- done in minimal systems in vitro. Such experiments have
bility of using synthetic approaches to recruit intracellular focused on engineering systems to gain insight into
motors, allowing the mapping of complex neural circuits motor mechanism. These approaches have paved the
and perhaps more complex tissue organization in the way for a toolbox of interchangeable motor parts, allowing
future. the design of self-assembling, synthetic motile devices

Box 2. Motility assays


In vitro motility assays provide powerful methods for studying or specifically through adaptors such as antibodies or chemical linkers.
mimicking the behavior of cellular transport. (a) In a cargo transport (b) In gliding assays, filaments are moved by the action of motors
assay crude or purified cellular components are observed to move on attached to a coverslip surface. Motors are typically attached to
immobilized filaments by observing the motion of the cargo. Motors coverslips through antibody linkages. (c) In a single-molecule motility
are either absorbed to beads nonspecifically such that their orienta- assay the filament is immobilized to a coverslip and fluorescently
tion and relative positions on the bead or cargo are not controlled, or labeled motors are observed to move along the filament.

(a) In vitro cargo transport assay

Direcon of
dynein movement

- +

(b) Filament gliding assay

Direcon of
microtubule gliding

+ -

Direcon of
dynein stepping

Y Y Y Y Y Y

(c) Single-molecule molity assay

Direcon of
dynein movement

- +

TRENDS in Cell Biology

Figure I. Motility assays.

648
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(termed ‘molecular shuttles’) destined for performing pro- understand the intramolecular interactions between
grammed transport tasks for biomedical and nanomanu- domains in kinesin and dynein, both of which are normally
facturing applications. homodimers. For kinesin, DNA was used to connect two
Motor proteins have been studied in vitro in both native motor-containing monomer subunits at non-native loca-
and recombinant forms (Box 2, Figure I). Initial single- tions [48]. With dynein, DNA replaced the native dimer-
molecule experiments with kinesin, myosin, and dynein ization domains such that two motor-containing monomer
relied on genetically engineering these motors by truncat- subunits were linked to form a functional heterodimer [46].
ing them to their smallest essential components, and This approach also enabled orthogonal fluorophore label-
adding sites for tags to aid in protein expression, purifica- ing of each motor subunit. Similar work using the FRB–
tion, and labeling (see e.g., [39,43,44]). These tags were FKBP system [38] made use of the concept of interchange-
originally intended for attaching fluorophores for visuali- able parts to study the mechanism of wild type and mutant
zation purposes, but recent advances allow them to serve dynein protomers [39,47]. Although these components
as attachment sites for linking elements that provide have yet to be categorized into truly ‘standard’ parts (such
connectivity [45–49]. This connectivity enables disparate as has been done for nucleic acid elements [51,52]), they are
motor and cargo components to become modular and link- interchangeable, with the potential for standardization
able to myriad other objects. Such linking can be accom- (Figure 2). Thus, a future goal for the field is to create a
plished not only with protein, but also with DNA, arguably toolbox of mechanical components that can be applied to
the most versatile connection medium [50]. controlling cargo transport.
DNA connections provide not only a guaranteed linking Initial work demonstrating the utility, promise, and
specificity through unique sequences, but are inexpensive, challenges associated with employing natural motors for
easily modifiable, allow tunable affinity, and come with engineering purposes has led to several devices, including
many enzymatic tools already provided by nature, such as molecular transporters and sorters, and has been reviewed
restriction enzymes, polymerases, and ligases. The engi- recently [6–8,53]. Most of these efforts to produce molecu-
neering properties of DNA have recently been employed to lar transport devices have relied on the filament-gliding

Interchangable parts Ideal component properes

Membrane-bound
Funconal elements

organelles
Nucleic
acids
· Flexible cargo type/funcon
· Task specific tools
Protein · Mulple funconal units
complexes

· Diverse aachment chemistries


Plaorm

Adaptor/scaffold · Modular construcon


· Orthogonal aachment sites
· Constuve or inducible

· Programmable motor type


Motors

· Biological or synthec
· Tunable motor funcons
· Recombinant or nave

TRENDS in Cell Biology

Figure 2. Synthetic cargo systems. An ideal synthetic motile system would have three major sets of components: task-specific functional elements (top), a scaffolding
platform (middle), and motor machinery (bottom). The scaffolding platform provides connectivity for all active components of the motile system and should be modular,
allowing multiple orthogonal attachments sites to bind motors and functional elements. Depending on in vitro or in vivo applications, it can be purely synthetic in origin, or
composed of biologically produced material. In addition, the scaffolding platform can be activated or induced as needed. The platform could be driven by motors of
synthetic or biological origin. The motors are recruited to the platform through motor-specific linking chemistries, such as DNA or Rapalogs. Motors are chosen from an
array of possibilities depending on need, where the potential task-specific motor attributes include directionally, velocity, track selection, and exogenous control. Finally,
specific functional elements can be bound to the platform. These might include various cargo payloads or specific tools for particular applications.

649
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

assay architecture (Box 2, Figure I) with actin or micro- achieve higher functions? These questions should help drive
tubules serving as the cargo scaffold (see e.g., [54,55]). the design principles when creating synthetic motors de
However, a modular cargo framework based on the cargo novo; however, purely synthetic systems also offer design
transport assay architecture (Box 2, Figure I) with multi- possibilities beyond what is available from the biologically
ple attachment possibilities allows the construction of derived tools. For example, the structural organization of
more complicated assemblies with programmable func- the motor could depart from cytoskeletal models and be
tions. Once a motor or motor component is connected to multimeric, with many discrete track and cargo-binding
a chemical linker, attachments to larger scaffolding struc- domains. Track and motor design offers the potential to
tures are possible. This has recently been performed with create many track–motor combinations or dynamically pro-
myosin [56,57] and kinesin [58], using either quantum dots grammable routes through a network of tracks. In addition,
[56] or DNA [57,58] as scaffolds. This work with DNA- motor tasks could be algorithmically determined depending
based scaffolds made use of a single DNA double helix; on the conditions and instruction sets contained within the
however, DNA origami [59,60] was recently used to create system (see e.g., [75,76]).
a more complicated scaffolding structure containing up to 7 Given the versatility of DNA as a nanotechnological
dynein and kinesin motors [61]. Other structure-building platform [50,77–79], it has become a primary construction
techniques [45] in vitro or in vivo [62] are also possible. material for synthetic molecular motors. These synthetic
Such systems could lead to the creation of more sophisti- motors tend to be significantly smaller than cytoskeletal
cated, varied, and modular molecular shuttles. motors and have focused on design issues of autonomy,
Molecular shuttles and other sophisticated synthetic processivity, velocity, directionality, track selection, and
applications of cytoskeletal motors require the ability to visualization. Some early synthetic DNA motors are termed
control the basic motile properties of the motors that drive ‘clocked’ because they required the sequential addition of
them. Methods for exogenous control over motors and fuel strands to propagate motility (see e.g., [80,81]). Howev-
motor systems will be required to achieve such applica- er, other designs allow for continuous autonomous motion
tions. Although progress has been made in understanding [82–88] and in some cases with coordinated stepping
how the motile properties of biologically derived motors are [84,88,89]. Some of these motors consist solely of DNA
modulated by the cell [14,15,63], there is a need for greater [84,89]; however, others make use of complementary com-
mechanistic understanding of this process. Crucial motor ponents such as DNAzymes with an RNA substrate [82,84]
parameters that require innovative methods of control or ligating [83] and nicking [83,85] enzymes. In these sys-
include regulation and control of cargo attachment, load- tems, directionality can be controlled by loading the motor
ing, and unloading; directionality; processivity; bidirec- onto the track at a specific location [82,85] or by the use of
tional versus unidirectional movement; and reuse or polar tracks [83,84]. In addition, instructions contained
recycling of motors. Recent work to address these issues within the motor’s fuel itself [88–90] can imbue directionali-
using synthetic approaches include protein engineering to ty, providing a directionality mechanism distinct from those
affect directionality [64,65], processivity [66–68], and di- of cytoskeletal motors. In contrast to cytoskeletal motor
merization [66]; chemical control over directionality and systems, in some cases the track is modified or destroyed
processivity [69–71]; motor copy-number effects on direc- as the synthetic motor moves along it [82,84–87].
tionality [72]; and photo-based control over motor function Recently, route-based selection motifs included in the
[73]. Of particular note is a study that engineered a track design have allowed motors to navigate particular
switchable, bidirectional myosin motor [64]. paths by following instructions in solution or intrinsic
parts of the motor [90,91]. Specific construction tasks have
Designing synthetic motors de novo also been demonstrated; an autonomous motor created
As a complement to the biologically evolved cytoskeletal sequential peptide bonds between building blocks analo-
motors, recent work has focused on the creation of synthet- gously to a ribosome [92], and a clocked motor performed a
ic motors capable of motion along a track. Although notable nanomanufacturing task by assembling clusters of gold
progress has been made in this area [74], artificial motors nanoparticles along an assembly line [93]. Furthermore, a
have yet to reach the capabilities of naturally occurring DNA motor visualized in real time was found to take
motors, particularly with regard to velocity and processiv- continuous, discrete steps [85], as was recently observed
ity. Nevertheless, there are many advantages to the de for myosin-V by atomic force microscopy [94]. The visuali-
novo design and fabrication of synthetic motors – attempts zation of single synthetic motor steps has also been
to create a functional motor could allow highly specific achieved using single-molecule motility assays (Box 2,
tasks to be performed and lead to a better understanding of Figure I) [82,95]. Future designs are likely to incorporate
the mechanistic details of naturally occurring motors. hybrid approaches that make use of both de novo and
Many fundamental considerations regarding how a syn- natural motor elements. By building modular components,
thetic motor might operate correlate directly with our new designs and functions could be rapidly created and
attempts to understand better the existing natural cytoskel- tested. One promising possibility is to use biological parts
etal motors. For example, how is directionality programmed as the core machinery, with synthetic adapters that are
into the structures and functions of the motors and their able to sense and interact with their environment.
tracks? What enables the motors to operate processively?
What is the source of fuel and how is it used? Given this fuel, Concluding remarks
what are the energy and entropic considerations that allow Nature has provided excellent nanoscale transporters, and
work to be done? Can multiple motors work together to our understanding of how cells control them is rapidly
650
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 3. Outstanding questions 5 Fisch, C. and Dupuis-Williams, P. (2011) Ultrastructure of cilia and
flagella – back to the future! Biol. Cell 103, 249–270
The cytoskeleton has proved to be an amenable system for 6 Goel, A. and Vogel, V. (2008) Harnessing biological motors to engineer
engineering novel synthetic functions. Some outstanding chal- systems for nanoscale transport and assembly. Nat. Nanotechnol. 3,
lenges in the field include the following questions: 465–475
 Can the cytoskeleton and motor proteins be used to create 7 van den Heuvel, M.G.L. and Dekker, C. (2007) Motor proteins at work
additional novel functionality? This is of particular interest in for nanotechnology. Science 317, 333–336
large, polarized cells where cargo sorting heavily relies on 8 Hess, H. (2011) Engineering applications of biomolecular motors.
transport mechanisms. Annu. Rev. Biomed. Eng. 13, 429–450
 Can the cytoskeleton be reorganized for novel transport require- 9 McIntosh, J.R. et al. (2010) Tubulin depolymerization may be an
ments? For example, can actin or microtubules be nucleated to ancient biological motor. J. Cell Sci. 123, 3425–3434
grow from new sites? 10 Conde, C. and Cáceres, A. (2009) Microtubule assembly, organization
 Can motors be co-opted to enhance the efficiency of synthetic in and dynamics in axons and dendrites. Nat. Rev. Neurosci. 10, 319–332
vivo processes already developed? 11 Müsch, A. (2004) Microtubule organization and function in epithelial
 Can a synthetic toolbox be built to recruit motors to any cargo of cells. Traffic 5, 1–9
interest with standardized parts? 12 Hirokawa, N. et al. (2010) Molecular motors in neurons: transport
 Can motors be exogenously controlled to produce molecular mechanisms and roles in brain function, development, and disease.
shuttles that start, stop, and change direction based on outside Neuron 68, 610–638
input? 13 Scholey, J.M. (2008) Intraflagellar transport motors in cilia: moving
 Can the cytoskeleton be harnessed for efficient directed transport along the cell’s antenna. J. Cell Biol. 180, 23–29
of foreign entities to the nucleus? 14 Kardon, J.R. and Vale, R.D. (2009) Regulators of the cytoplasmic
 Can motors be designed de novo to have the efficiency, velocity, dynein motor. Nat. Rev. Mol. Cell Biol. 10, 854–865
and processivity of biological motors? Can these synthetic motors 15 Verhey, K.J. and Hammond, J.W. (2009) Traffic control: regulation of
be imbued with greater functionality than biological motors? kinesin motors. Nat. Rev. Mol. Cell Biol. 10, 765–777
16 Hammer, J.A. and Sellers, J.R. (2012) Walking to work: roles for class V
myosins as cargo transporters. Nat. Rev. Mol. Cell Biol. 13, 13–26
increasing. The regulatory control issues cells face with 17 Walker, R.A. et al. (1990) The Drosophila claret segregation protein is a
molecular motors are similar to those that synthetic biol- minus-end directed motor molecule. Nature 347, 780–782
18 Wells, A.L. et al. (1999) Myosin VI is an actin-based motor that moves
ogists encounter in harnessing and reengineering these
backwards. Nature 401, 505–508
motors for novel tasks. Similarly, these engineering pro- 19 Akhmanova, A. and Hammer, J.A. (2010) Linking molecular motors to
blems overlap with areas of active research in cell biology. membrane cargo. Curr. Opin. Cell Biol. 22, 479–487
The complementary approaches of working on solutions to 20 Kamal, A. and Goldstein, L.S.B. (2002) Principles of cargo attachment
these design problems, and understanding how the cell has to cytoplasmic motor proteins. Curr. Opin. Cell Biol. 14, 63–68
21 Kulic, I.M. et al. (2008) The role of microtubule movement in
solved these same issues, will enrich the utility and un-
bidirectional organelle transport. Proc. Natl. Acad. Sci. U.S.A. 105,
derstanding of molecular motors. 10011–10016
Although in the early stages of research, the ease of 22 Döhner, K. et al. (2005) Viral stop-and-go along microtubules: taking a
building in vitro transport systems shows that the core ride with dynein and kinesins. Trends Microbiol. 13, 320–327
motor components are amenable to modification and in- 23 Dodding, M.P. and Way, M. (2011) Coupling viruses to dynein and
kinesin-1. EMBO J. 30, 3527–3539
clusion within modular engineered systems. These new 24 Bremner, K.H. et al. (2009) Adenovirus transport via direct interaction
constructs will continue to increase in both standardiza- of cytoplasmic dynein with the viral capsid hexon subunit. Cell Host
tion and complexity, and in the future will be used to probe Microbe 6, 523–535
cellular functions and provide novel synthetic applications. 25 Douglas, M.W. et al. (2004) Herpes simplex virus type 1 capsid
Purely synthetic motors will continue to inform how en- protein VP26 interacts with dynein light chains RP3 and Tctex1 and
plays a role in retrograde cellular transport. J. Biol. Chem. 279,
dogenous motors function. The programmability of artifi- 28522–28530
cial motors will expand their role in synthetic 26 Ward, B.M. and Moss, B. (2004) Vaccinia virus A36R membrane
nanomanufacturing, possibly outpacing the use of natural protein provides a direct link between intracellular enveloped
motors. virions and the microtubule motor kinesin. J. Virol. 78, 2486–2493
27 Diefenbach, R.J. et al. (2002) Herpes simplex virus tegument protein
Despite this exciting progress, there are a number of
US11 interacts with conventional kinesin heavy chain. J. Virol. 76,
outstanding questions in the field and challenges for the 3282–3291
future (Box 3). The constant feedback between synthetic 28 Haglund, C.M. and Welch, M.D. (2011) Pathogens and polymers:
and cellular biologists promises to allow this nascent field microbe-host interactions illuminate the cytoskeleton. J. Cell Biol.
to uncover the mechanisms of cellular motility and engi- 195, 7–17
29 Lambrechts, A. et al. (2008) Listeria comet tails: the actin-based
neer novel transport systems.
motility machinery at work. Trends Cell Biol. 18, 220–227
30 Schwan, C. et al. (2009) Clostridium difficile toxin CDT induces
Acknowledgments formation of microtubule-based protrusions and increases adherence
We thank J. Tytell, X. Su, and S. Wickham for comments on the of bacteria. PLoS Pathog. 5, e1000626
manuscript. 31 Kapitein, L.C. et al. (2010) Mixed microtubules steer dynein-driven
cargo transport into dendrites. Curr. Biol. 20, 290–299
References 32 Kapitein, L.C. et al. (2010) Probing intracellular motor protein activity
1 Vale, R.D. (2003) The molecular motor toolbox for intracellular using an inducible cargo trafficking assay. Biophys. J. 99, 2143–2152
transport. Cell 112, 467–480 33 Lechardeur, D. et al. (2005) Intracellular routing of plasmid DNA
2 Gadde, S. and Heald, R. (2004) Mechanisms and molecules of the during non-viral gene transfer. Adv. Drug Deliv. Rev. 57, 755–767
mitotic spindle. Curr. Biol. 14, R797–R805 34 Edelstein, M.L. et al. (2007) Gene therapy clinical trials worldwide to
3 Wittmann, T. et al. (2001) The spindle: a dynamic assembly of 2007 – an update. J. Gene Med. 9, 833–842
microtubules and motors. Nat. Cell Biol. 3, E28–E34 35 Toledo, M.A.S. et al. (2012) Development of a recombinant fusion
4 Gibbons, I.R. (1996) The role of dynein in microtubule-based motility. protein based on the dynein light chain LC8 for non-viral gene
Cell Struct. Funct. 21, 331–342 delivery. J. Control. Release 159, 222–231

651
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

36 Bergen, J.M. and Pun, S.H. (2007) Evaluation of an LC8-binding 65 Carter, A.P. et al. (2008) Structure and functional role of dynein’s
peptide for the attachment of artificial cargo to dynein. Mol. Pharm. microtubule-binding domain. Science 322, 1691–1695
4, 119–128 66 Shishido, H. and Maruta, S. (2012) Engineering of a novel Ca2+-
37 Miller, A.M. and Dean, D.A. (2009) Tissue-specific and transcription regulated kinesin molecular motor using a calmodulin dimer linker.
factor-mediated nuclear entry of DNA. Adv. Drug Deliv. Rev. 61, 603– Biochem. Biophys. Res. Commun. 423, 386–391
613 67 Clancy, B.E. et al. (2011) A universal pathway for kinesin stepping.
38 Banaszynski, L. et al. (2005) Characterization of the FKBP. Nat. Struct. Mol. Biol. 18, 1020–1027
rapamycin.FRB ternary complex. J. Am. Chem. Soc. 127, 4715–4721 68 Hodges, A.R. et al. (2007) Engineering the processive run length of
39 Reck-Peterson, S.L. et al. (2006) Single-molecule analysis of dynein Myosin V. J. Biol. Chem. 282, 27192–27197
processivity and stepping behavior. Cell 126, 335–348 69 Gerson-Gurwitz, A. et al. (2011) Directionality of individual kinesin-5
40 Lo Presti, L. and Martin, S.G. (2011) Shaping fission yeast cells by Cin8 motors is modulated by loop 8, ionic strength and microtubule
rerouting actin-based transport on microtubules. Curr. Biol. 21, 2064– geometry. EMBO J. 30, 4942–4954
2069 70 Cochran, J.C. et al. (2012) A metal switch for controlling the activity of
41 Enquist, L.W. and Card, J.P. (2003) Recent advances in the use of molecular motor proteins. Nat. Struct. Mol. Biol. 19, 122–127
neurotropic viruses for circuit analysis. Curr. Opin. Neurobiol. 13, 603– 71 Walter, W.J. et al. (2012) Two independent switches regulate
606 cytoplasmic dynein’s processivity and directionality. Proc. Natl.
42 Beier, K.T. et al. (2011) Anterograde or retrograde transsynaptic Acad. Sci. U.S.A. 109, 5289–5293
labeling of CNS neurons with vesicular stomatitis virus vectors. 72 Roostalu, J. et al. (2011) Directional switching of the kinesin Cin8
Proc. Natl. Acad. Sci. U.S.A. 108, 15414–15419 through motor coupling. Science 332, 94–99
43 Warshaw, D.M. et al. (2005) Differential labeling of myosin V heads 73 Yamada, M.D. et al. (2007) Photocontrol of kinesin ATPase activity
with quantum dots allows direct visualization of hand-over-hand using an azobenzene derivative. J. Biochem. 142, 691–698
processivity. Biophys. J. 88, L30–L32 74 von Delius, M. and Leigh, D.A. (2011) Walking molecules. Chem. Soc.
44 Rice, S. et al. (1999) A structural change in the kinesin motor protein Rev. 40, 3656–3676
that drives motility. Nature 402, 778–784 75 Qian, L. et al. (2011) Neural network computation with DNA strand
45 Diehl, M.R. et al. (2006) Engineering cooperativity in biomotor–protein displacement cascades. Nature 475, 368–372
assemblies. Science 311, 1468–1471 76 Soloveichik, D. et al. (2010) DNA as a universal substrate for chemical
46 Qiu, W. et al. (2012) Dynein achieves processive motion using both kinetics. Proc. Natl. Acad. Sci. U.S.A. 107, 5393–5398
stochastic and coordinated stepping. Nat. Struct. Mol. Biol. 19, 193–200 77 Seeman, N.C. (2010) Nanomaterials based on DNA. Annu. Rev.
47 DeWitt, M.A. et al. (2012) Cytoplasmic dynein moves through Biochem. 79, 65–87
uncoordinated stepping of the AAA+ ring domains. Science 335, 78 Bath, J. and Turberfield, A.J. (2007) DNA nanomachines. Nat.
221–225 Nanotechnol. 2, 275–284
48 Miyazono, Y. et al. (2010) Strain through the neck linker ensures 79 Aldaye, F.A. et al. (2008) Assembling materials with DNA as the guide.
processive runs: a DNA–kinesin hybrid nanomachine study. EMBO Science 321, 1795–1799
J. 29, 93–106 80 Sherman, W.B. and Seeman, N.C. (2004) A precisely controlled DNA
49 Guydosh, N.R. and Block, S.M. (2009) Direct observation of the binding biped walking device. Nano Lett. 4, 1203–1207
state of the kinesin head to the microtubule. Nature 461, 125–128 81 Shin, J-S. and Pierce, N.A. (2004) A synthetic DNA walker for
50 Michelotti, N. et al. (2012) Beyond DNA origami: the unfolding molecular transport. J. Am. Chem. Soc. 126, 10834–10835
prospects of nucleic acid nanotechnology. Wiley Interdiscip. Rev. 82 Lund, K. et al. (2010) Molecular robots guided by prescriptive
Nanomed. Nanobiotechnol. 4, 139–152 landscapes. Nature 465, 206–210
51 Shetty, R.P. et al. (2008) Engineering BioBrick vectors from BioBrick 83 Peng, Y. et al. (2004) A unidirectional DNA walker that moves
parts. J. Biol. Eng. 2, 5 autonomously along a track. Angew. Chem. Int. Ed. Engl. 43, 4906–
52 Canton, B. et al. (2008) Refinement and standardization of synthetic 4911
biological parts and devices. Nat. Biotechnol. 26, 787–793 84 Omabegho, T. et al. (2009) A bipedal DNA Brownian motor with
53 Korten, T. et al. (2010) Towards the application of cytoskeletal motor coordinated legs. Science 324, 67–71
proteins in molecular detection and diagnostic devices. Curr. Opin. 85 Wickham, S.F.J. et al. (2011) Direct observation of stepwise movement
Biotechnol. 21, 477–488 of a synthetic molecular transporter. Nat. Nanotechnol. 6, 166–169
54 Schmidt, C. et al. (2012) Tuning the ‘roadblock’ effect in kinesin-based 86 Pei, R. et al. (2006) Behavior of polycatalytic assemblies in a substrate-
transport. Nano Lett. 12, 3466–3471 displaying matrix. J. Am. Chem. Soc. 128, 12693–12699
55 Malcos, J.L. and Hancock, W.O. (2011) Engineering tubulin: 87 Bath, J. et al. (2005) A free-running DNA motor powered by a nicking
microtubule functionalization approaches for nanoscale device enzyme. Angew. Chem. Int. Ed. Engl. 44, 4358–4361
applications. Appl. Microbiol. Biotechnol. 90, 1–10 88 Bath, J. et al. (2009) Mechanism for a directional, processive, and
56 Ali, M.Y. et al. (2011) Myosin Va and myosin VI coordinate their steps reversible DNA motor. Small 5, 1513–1516
while engaged in an in vitro tug of war during cargo transport. Proc. 89 Green, S.J. et al. (2008) Coordinated chemomechanical cycles: a
Natl. Acad. Sci. U.S.A. 108, E535–E541 mechanism for autonomous molecular motion. Phys. Rev. Lett. 101,
57 Lu, H. et al. (2012) Collective dynamics of elastically-coupled myosinV 238101
motors. J. Biol. Chem. 287, 27753–27761 90 Wickham, S.F.J. et al. (2012) A DNA-based molecular motor that can
58 Rogers, A.R. et al. (2009) Negative interference dominates collective navigate a network of tracks. Nat. Nanotechnol. 7, 169–173
transport of kinesin motors in the absence of load. Phys. Chem. Chem. 91 Muscat, R.A. et al. (2011) A programmable molecular robot. Nano Lett.
Phys. 11, 4882–4889 11, 982–987
59 Shih, W.M. and Lin, C. (2010) Knitting complex weaves with DNA 92 He, Y. and Liu, D.R. (2010) Autonomous multistep organic synthesis in
origami. Curr. Opin. Struct. Biol. 20, 276–282 a single isothermal solution mediated by a DNA walker. Nat.
60 Krishnan, Y. and Bathe, M. (2012) Designer nucleic acids to probe and Nanotechnol. 5, 778–782
program the cell. Trends Cell. Biol. 22, 624–633 93 Gu, H. et al. (2010) A proximity-based programmable DNA nanoscale
61 Derr, N.D. et al. (2012) Tug of war in motor protein ensembles revealed assembly line. Nature 465, 202–205
with a programmable DNA origami scaffold. Science 338, 662–665 94 Kodera, N. et al. (2010) Video imaging of walking myosin V by high-
62 Delebecque, C.J. et al. (2011) Organization of intracellular reactions speed atomic force microscopy. Nature 468, 72–76
with rationally designed RNA assemblies. Science 333, 470–474 95 Masoud, R. et al. (2012) Studying the structural dynamics of bipedal
63 Hartman, M.A. et al. (2011) Principles of unconventional myosin DNA motors with single-molecule fluorescence spectroscopy. ACS
function and targeting. Annu. Rev. Cell Dev. Biol. 27, 133–155 Nano 6, 6272–6283
64 Chen, L. et al. (2012) Engineering controllable bidirectional molecular 96 Clackson, T. et al. (1998) Redesigning an FKBP–ligand interface to
motors based on myosin. Nat. Nanotechnol. 7, 252–256 generate chemical dimerizers with novel specificity. Proc. Natl. Acad.
Sci. U.S.A. 95, 10437–10442

652
Review

Special Issue – Synthetic Cell Biology

Principles for designing ordered


protein assemblies
Yen-Ting Lai1, Neil P. King2, and Todd O. Yeates3,4
1
Biomedical Engineering Interdepartmental Degree Program, University of California, Los Angeles, CA, USA
2
Department of Biochemistry, University of Washington, Seattle, WA, USA
3
Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
4
University of California Los Angeles (UCLA)–Department of Energy Institute for Genomics and Proteomics, University of
California, Los Angeles, CA, USA

In nature, many proteins have evolved to have self- a highly flexible molecule self-associates to form a higher-
complementary shapes. This drives them to assemble order assembly, the result is typically an extended and
into supramolecular structures, sometimes of great geometrically irregular material. Such network or mesh-
complexity, and often carrying out sophisticated cellular like materials have interesting bulk properties, which can
functions. Designing novel proteins that can self-assem- sometimes be modulated in useful ways [7]. The second
ble into similarly complex structures is a longstanding group, where the assembly behavior is intended to be
goal in bioengineering. New ideas, combined with con- deterministic, encompasses those strategies aimed at pro-
tinually improving computer algorithms, are making it ducing specific 3D structures [5]. These structures, which
possible to advance on that goal, bringing wide-ranging may be finite in size (e.g., clusters or cages) or indefinite in
applications in synthetic biology within reach. Prospec- extent (e.g., arrays or crystals), can be built with atomic
tive applications range from vaccine design to molecular level features in mind.
delivery to bioactive materials. Recent strategies and Early work on designing geometrically specific protein
examples of successfully designed protein cages, layers, assemblies focused on filamentous structures as design
and crystals are reviewed. targets. That early focus reflects the relatively simple
design requirements for filamentous assemblies: a single
Protein assemblies as a synthetic biology goal self-associating interface can produce end-to-end polymer-
The emerging research area of synthetic biology seeks to ization. Specific well-studied self-associating protein
recreate various complex phenomena exhibited by biologi- motifs have been a rich source of building blocks for
cal systems, especially at the molecular level [1]. The designing filamentous structures. Helical coiled-coils have
phenomena of interest are often characterized by a high been especially useful [8,9]. Cyclic polypeptides composed
degree of order – either in time or space. The emergent of b-strand-preferring amino acids have provided another
behavior or ‘output’ of synthetic systems can be consider- self-associating motif, in this case leading to rigid tubular
ably more complex than the behavior of the individual assemblies [10]. Variations on filamentous designs have
components [2]. Often, this arises from the introduction sought to produce more complex patterns, such as branch-
of non-trivial or self-referring interactions between compo- ing [11], but the end-to-end polymerization strategies cen-
nents. For example, if rather than simply A affecting B, tral to filament design do not extend easily to the problem
instead B also affects C, and C affects A, then an output of creating highly specific 3D architectures.
behavior may arise where molecular concentrations oscil- In this review we focus on strategies for designing
late in time [3]. Likewise, if rather than simply A binding to proteins that self-assemble to give defined structures with
B, instead A binds to itself in multiple ways, surprisingly complex architectures, including cages and extended 2D
large and complex molecular assemblies can arise, leading and 3D crystalline arrays.
to spatial organization of various types, such as compart-
mentalization or long-range propagation of forces by rigid Underlying principles
structures. We concern ourselves here with strategies for In nature, wherever supramolecular structures are built
designing protein molecules that self-associate to produce up by the assembly of multiple copies of the same subunit
large, complex assemblies with potential synthetic biology (or similar subunits), the subunits are nearly always as-
applications. sembled in a symmetrical fashion [12,13]. The reason for
Diverse efforts in the area of designing protein-based this was anticipated as early as 1956 by Crick and Watson
assemblies and materials can be divided into two groups: [14]: symmetric assemblies require fewer distinct kinds of
stochastic and deterministic [4,5]. The stochastic group specific interaction interfaces compared to asymmetric
encompasses several design strategies where the self- assemblies. It is natural then that efforts to design ordered
assembling protein molecule is highly flexible [4,6]. When protein assemblies should rely on principles of symmetry.
We articulate three connected ideas that we believe are
Corresponding author: Yeates, T.O. (yeates@mbi.ucla.edu).
Keywords: protein design; symmetry; cages; biomaterials; nanomaterials; assembly. important to permit full exploitation of symmetry-based
0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.08.004 Trends in Cell Biology, December 2012, Vol. 22, No. 12 653
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

approaches for designing self-assembling molecular sys- assemblies has been a difficult challenge. However, efforts
tems: along multiple lines are beginning to bear fruit. Recalling
(i) A symmetric molecular assembly, whether it is a finite the discussions above on the design requirement of intro-
structure or an extended array, is characterized by its ducing two (or more) modes of self-association into a single
symmetry group. A symmetry group is an exact protein molecule, varied approaches to the problem of
mathematical idea that expresses the complete set of designing self-assembling proteins can be grouped accord-
spatial operations that interrelate a set of individual ing to the strategy used to satisfy this central requirement.
components, in this case a set of structurally identical Different strategies rely to different degrees on natural (or
protein subunits in a 3D assembly. For example, a 24- native-like) protein–protein interfaces versus novel inter-
subunit assembly built with the symmetry of a cube faces created by amino acid sequence design (Figure 1).
(referred to as octahedral symmetry) is described by a
symmetry group with 24 spatial operations. Every pair Fusion of natural oligomers
of subunits is related by some operation in the group. An early idea for introducing two self-associating interfaces
For chiral biological molecules, such as proteins, these into a single protein molecule emerged at a time when the
operations must be rotations, potentially combined prospects for designing novel protein–protein interfaces by
with translations, but excluding reflections. computational methods were limited. In 2001, we proposed
(ii) Any interface formed between two subunits in a that genetically fusing two different, naturally oligomeric
symmetric assembly corresponds to one of the domains into a single protein chain could satisfy the design
operations of the symmetry group (vide supra). requirement of combining two self-associating motifs [16].
(iii) For a hypothetical, symmetric constellation of sub- The problem was how to dictate, or at least predict in
units (whether finite or indefinite) to constitute a advance, what the relative orientation would be between
plausible, physically connected assembly – in other two genetically fused domains; free backbone rotations
words, to not be two or more disjoint sub-complexes – occur at a point of fusion, and this would make the final
the following condition is both necessary and suffi- geometry unpredictable. The solution for how to predict the
cient. Taking any individual subunit as a reference, relative orientation in advance was to use oligomeric
the distinct interface types it uses to contact its domains that began or ended in an a-helix, such that the
neighbors comprise a subset of the operations of the protein backbone (and any a-helix-preferring amino acids
complete symmetry group, and this subset of opera- introduced as a linker) might adopt an unbroken a-helix
tions must be capable of generating the full symmetry running from within one oligomeric domain into the next. In
group. That is, repeated combinations of this typically this way, pairs of oligomeric domains of known structure
small subset of operations must be able to reform the would be combined in hypothetical, predicted arrange-
whole symmetry group. In physical terms, this is ments, and a search could be made for pairs that would
equivalent to stating that it must be possible to trace satisfy specific geometric rules for constructing different
a path from any one molecule to any other molecule in architectures such as cages or crystals. The same paper laid
the assembly through the contacts between molecules out geometric rules for how the symmetry axes of the
in the assembly (Box 1). Otherwise, the collection of component domains would have to be oriented relative to
molecules would be disjoint. each other to obtain various architectures ([16], Box 1). That
initial set of geometric rules only covered combinations of
The first two ideas are relatively well-known to those dimers and trimers because the protein structure database
familiar with crystallography or molecular symmetry, but at that time contained relatively few proteins with both
the third is less obvious. It was first articulated in the higher oligomeric symmetry and subunits ending in helices
context of crystal symmetries [15], and then in the context [16]. A tremendous range of assembly architectures can be
of designed protein assemblies [16]. Particularly surprising, achieved using higher symmetry building blocks; those
and important from a design perspective, was the realiza- possibilities have not been completely enumerated yet.
tion that large, complex symmetries (including some 3D The oligomeric fusion method was first used in the design
crystal symmetries) could be generated using only two of protein filaments – a relatively easy design target – and a
distinct symmetry elements [16]. This translates to the idea 12-subunit molecular cage with a tetrahedral shape, repre-
that if one can design two distinct, geometrically specific, senting the first protein construction of its kind. However,
self-associating interfaces into a single molecule, then a that initially designed protein sequence formed cage-like
wide range of assembly architectures can be realized. Al- assemblies whose sizes were too heterogeneous to charac-
though more than the minimum number of distinct contact terize in atomic detail; crystals could not be obtained [16].
types can be introduced in a design – and large natural This barrier was surmounted in recent work. When the
assemblies such as viral capsids almost always exhibit more original design was revisited, and two amino acid changes
than the minimum number of distinct contact types [17] – were made based on a visual identification of potential steric
the minimum contact number (which is just two for many conflicts, a homogenous 12-subunit cage was obtained and
cases) establishes an important design principle. crystallized [18]. The designed cage is roughly 16 nm in
diameter and contains a central opening about 5 nm in
Design strategies and successes diameter (Figure 2a,b). Interestingly, despite an overall
Owing largely to the complexity of protein molecules, and match to the design, the observed assembly exhibited sig-
our incomplete understanding of the rules by which they nificant deviations (about 8 Å root-mean-square deviation,
fold and recognize each other, designing complex protein RMSD) from perfect symmetry [18].
654
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 1. Principles for designing ordered protein assemblies


A general requirement for engineering complex self-assembling assemblies. In each case, the two types of symmetry elements are
structures is to create a protein molecule bearing multiple distinct noted in bold, together with the angle they must form. The
self-associating interfaces (Figure I, top). This can be achieved symmetries of the resulting assemblies are given (T: tetrahedral;
through the use of natural oligomeric proteins or computational O: octahedral; I: icosahedral). For 2D layers and 3D crystals (not
sequence design, separately or in combination. The multiple shown), rules for constructing the full range of possible symmetries
interfaces must be combined according to specific geometric rules have not been articulated yet [16], though specific instances of 2D
[16] if defined structures are to be created (Figure I, bottom). The layers and 3D crystals have been successfully engineered
geometric design requirements are given for some example [22,35,36].

Generang mulple self-associang interfaces by fusion or sequence design

or or

Natural + natural (fusion) Natural + designed Designed + designed

Examples of designed architectures based on geometrically controlled


combinaons of symmetric interfaces

Cages or shells Layers

4 & 2, 45 4 & 3, 54.7


24 subunits 24 subunits
symmetry O symmetry O

Other possibilies (not shown):


3 & 2, 54.7 12 subunits, symmetry T [18,34] 4 & 2 , parallel
3 & 2, 35.3 24 subunits, symmetry O [34] layer symmetry p422
3 & 2, 20.9 60 subunits, symmetry I
5 & 2, 31.7 60 subunits, symmetry I Other possibilies (not shown):
5 & 3, 37.4 60 subunits, symmetry I - numerous [16,22]

TRENDS in Cell Biology

Figure I. Strategies and geometric rules for combining multiple protein interfaces.

Variations on the fusion approach have been introduced superoxide reductase [20]. These building blocks formed
by others. In one study [19], natural oligomers were used to linear filaments upon mixing. Smaller protein motifs have
supply the two self-associating interfaces required for an also been used in oligomeric fusion approaches. One study
extended 2D array of protein molecules, but in this case the connected a trimeric coiled-coil to a pentameric coiled-coil
components were connected by binding to biotin instead of with the goal of generating 60-subunit icosahedral parti-
by genetic fusion; one of the oligomers was streptavidin, a cles [21]. A disulfide bond was introduced to force a bend
D2 tetramer, and the other was aldolase (RhuA), a C4 between the two helical components; an angle of precisely
tetramer. Arrays of only limited extent were obtained, 37.48 is required to satisfy icosahedral symmetry. The
probably owing to flexibility of the biotin linker and imper- particles formed by this design appeared polymorphic
fect control over the relative orientation of the component but were roughly the intended size of 16 nm.
oligomers. In another study, two types of building blocks A notable extension to the original oligomer fusion
were generated by fusing PDZ domains or PDZ-binding strategy was recently introduced [22] (Figure 2c,d). These
peptides to the termini of a tetrameric (D2 symmetry) authors noted that, if higher-order oligomers are used as
655
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (e)

(b) (c) (d)

TRENDS in Cell Biology

Figure 1. Strategies for introducing a new oligomerization interface. (a) Oligomeric fusion strategy. Two different oligomeric proteins can be fused together to generate two
oligomerization interfaces within a single protein subunit, thereby driving the assembly of complex structures [16]. A single protein chain within the complex is shown in
the black rectangle. In the example shown, the green domain derives from a natural trimer and the magenta domain derives from a natural dimer. Linking residues are
yellow. (b) New interfaces generated through metal-binding. Multiple histidine residues incorporated into the side of an exposed a-helix can constitute a new dimeric
interface [30]. Two different chains are shown in green and yellow, histidine residues are shown in black lines, and metal ions are shown as orange spheres. (c) a-Helix-
based oligomerization. a-Helices can be designed to form different oligomerization states based on well-studied coiled-coil motifs [32]. Different chains are shown in
different colors. (d) b-Sheet-based oligomerization. The open edge of a b-sheet can be used as a site for designing a dimeric interaction [31]. Two different chains are
colored in green and cyan. (e) De novo design of a new interface. A new interface can be introduced into a natural oligomer of relatively low initial symmetry to generate
higher-order assemblies [34]. In the case illustrated, multiple copies of a natural trimer are shown in different colors. One trimer, shown in green, is lifted from its assembled
position. The de novo designed interaction patches are shown in red. The designed interactions between the green subunit and other subunits are indicated by thin lines.

the components, the two oligomers to be genetically fused motif to provide one of the modes of self-association,
can be aligned at an axis of symmetry that they both share. and computer algorithms are then used to introduce an
In such a scenario, there will be two (or more) polypeptide additional interface (or interfaces) (Box 1). This approach
linkers instead of one running between adjacent oligo- minimizes the number of novel interfaces that must be
mers. The requirement for a rigid a-helical linker could successfully designed computationally. Strategies that
then be removed, with the necessary orientational control combine a natural oligomeric interface with the computa-
resulting instead from the multiple chain connections. tional design of an additional interaction were presaged in
This symmetry-matching fusion protein strategy success- experiments [26] in which assemblies such as double-ring
fully generated a linear filament and 2D arrays. A notable structures were generated by introducing relatively sim-
success rate of 40% was achieved when constructing 2D ple interfacial features such as a hydrophobic patch into
arrays, and one of the arrays exhibited exceptionally good simpler, single-ring, natural protein assemblies. The suc-
long-range order. The design of 3D crystals based on this cess rate of that strategy was higher when the starting
strategy led to large solid aggregates having crystal-like structures contained more subunits (for example, C4 com-
morphology, but the long-range order required for crystal- pared to C2), presumably owing to the higher multiplicity
lographic analysis has not been reported yet. of the newly introduced interaction sites. In addition, the
In theory, the shared symmetry axis method can be used observed assemblies typically showed substantial devia-
to create 2D and 3D arrays obeying a variety of different tions from the intended structures, presumably reflecting
symmetries, the full range of which has not been articu- the limited geometric precision provided by the sequence
lated yet. However, an unavoidable constraint of the meth- design strategy.
od is that only extended materials (e.g., 2D arrays and 3D Numerous sequence design strategies for promoting
crystals) can be generated, but not finite structures such as self-association have been demonstrated, with strategies
molecular cages. that deliver the highest geometric specificity placing the
greatest demand on the sequence design process
Interface design (Figure 1b–e). At one end of the spectrum, a simple se-
Computer programs for designing protein–protein inter- quence element such as two histidines at positions i and
actions are becoming increasingly powerful [23–25], mak- i+4 on the exposed surface of an a-helix has proved to be a
ing it possible to design sequence mutations that drive straightforward approach for driving protein self-associa-
specific modes of symmetric self-association. This has tion in the presence of metals [27–29]. However, the spe-
opened up more direct strategies for designing large pro- cific geometries of those associations have been hard to
tein assemblies and extended materials. The most conser- predict [30]. Higher levels of geometric specificity have
vative approaches for designing such large assemblies, been demonstrated in the design of self-associating inter-
including most of the successful experiments reported so actions involving an exposed b-strand in one case [31], and
far, rely on a natural (or native-like) oligomerization a-helical bundles in several other studies [32,33]. At the
656
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b)

2
3

16 nm

(c) 4 2 (d)

c N 2 xn
28 nm
2

2 2
2
2

TRENDS in Cell Biology

Figure 2. Designed assemblies based on two types of oligomer-fusion strategies. (a) Fusion through a semi-rigid helix linker. Two natural oligomeric domains (green and
purple; left) are held in a predetermined orientation by genetic fusion of the two domains and an a-helical linker (yellow; middle) [16]. Multiple copies of the fusion protein
can then self-assemble into a well-defined shape (right). (b) Crystal structure of a successful example by using the helix-linker fusion strategy [18]. Twelve different chains in
the assembly are colored differently; a central sphere of about 5 nm is shown in black. (c) Fusion by common symmetry axis. Short and relatively flexible linkers (green; left)
are used to fuse natural oligomeric domains (pink and yellow) such that the shared symmetry axes tend to align with each other (blue arrow; left) [22]. Multiple subunits can
then self-assemble into extended arrays (right). (d) Molecular model of a layer assembled by the common-symmetry-axis fusion strategy, which was consistent with
electron micrographs of the resulting material. Panels (a) and (b) adapted, with permission, from [18]. Panel (c) adapted, with permission, from [55]. Panel (d) adapted, with
permission, from [22].

extreme end, the sequence design of entirely novel, two symmetric metal-binding motifs each had one open
geometrically specific self-associating interfaces (i.e., not coordination site to promote a further twofold association
modeled on well-characterized natural motifs) has been between dimers. Typically, a tail-to-tail association between
demonstrated in a newly published study [34]. Together, head-to-head dimers tends to produce a filament or strip of
these varied modes of interface design have been exploited molecules. In this study [35] it appears that different exper-
in a recent flurry of experiments demonstrating the crea- imental conditions promoted different arrangements and
tion of complex assemblies and extended materials. associations of such strips, such as side-by-side associations
Metal site design, combined with additional interface to give sheets, or helical wrappings to give tubular struc-
design, was used to create a small protein that self-assem- tures (Figure 3). Assemblies produced by the metal-mediat-
bles into a series of different architectures [35]. A cyto- ed strategy have the potential to be fine-tuned through the
chrome protein that had been engineered by the adjustment of the protein:metal ratio or pH. At the present
introduction of coordinating histidine residues to form a stage, the assembly outcomes in metal-mediated designs
metal-induced dimer was stabilized in that specific geomet- must be established experimentally, and the generation of
rical configuration by further mutations using computer- predictable structures constitutes an important future chal-
aided interface design. In the resulting configuration, the lenge.

(a) (b) (c)

Bend & twist

Zn1 Zn3
i-face i-face

Zn2
i-face
Key: Zn1 Zn2 Zn3

TRENDS in Cell Biology

Figure 3. Layers and nanotubes by metal-mediated assembly. (a) Zinc was used to trigger the assembly of protein subunits with designed di-histidine motifs in two
perpendicular directions into a layer, as shown in (b) [35]. The equivalent part of (a) is shown in the green box. Three differently located zinc ions are colored in cyan,
orange, and red. (c) Bending and twisting around the binding site for zinc #3 caused the layer to curl into a nanotube. Adapted, with permission, from [35].

657
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b)

1.7 nm

4 nm

TRENDS in Cell Biology

Figure 4. A designed 3D protein crystal using coiled-coil building blocks. (a) An assembly obeying P6 crystal symmetry was designed using a parallel 3-helix coiled-coil as
the starting motif [36]. Computational sequence design was used to promote lateral and vertical contacts between trimers. Distinct chains in the trimer are colored in green,
cyan, and magenta. Minor breakage of the intended P6 symmetry is visible. (b) The vertical stacking of layers is illustrated in a perpendicular view from the position of the
black triangle in (a). The ends of the helices in one layer form hydrogen bonds with helices in neighboring layers, forming semi-continuous helices along the longitudinal
crystal dimension. The layer shown in (a) is located between the two solid lines.

The successful design of a self-assembling 3D protein that either expressed insolubly or failed to assemble,
crystal was recently demonstrated [36]. First, a protein emphasizing the need for improved methods for designing
backbone conformation based on a well-studied protein protein–protein interactions if this approach is to become
motif, a three-helix bundle, was placed in a chosen crystal widely used.
lattice. In this case, the template trimer was placed on the
threefold symmetry axis of a P6 crystal unit cell (Figure 4). Strategic variations
Favorable unit cell spacings and trimer orientations were The basic design ideas discussed above admit numerous
then sampled, after which the sequence of the protein was variations, some of which have been demonstrated already.
optimized based on its interactions with symmetry-based For extended protein assemblies, one immediate obstacle
neighboring protein molecules. Following production of the is the production of recombinant proteins in soluble form
designed protein, two different protein crystal forms were during bacterial protein expression. The building blocks for
obtained, one of which agreed very closely with the designed such assemblies tend to associate as soon as they are
crystal model, with deviations in the range of about 1 Å. produced inside the host cell, leading to large aggregates
Two different test cases recently demonstrated the ability and inclusion bodies that seriously complicate the problem
to design a large, novel self-associating interface into a of protein purification. Several groups have overcome this
naturally oligomeric protein subunit to produce large molec- problem by splitting the assemblies into two or more
ular cages and clusters [34] (Figure 5). Novel homodimeric distinct components that can be produced in soluble forms
interfaces were designed into two different naturally trimer- in separate bacterial strains [19,20,22]. Purified, soluble
ic proteins in geometries intended to yield a 24-subunit cage components can then be mixed to form the intended as-
with octahedral symmetry in one case and a 12-subunit semblies in situ. Other methods to trigger protein assem-
assembly with tetrahedral symmetry in the other. This blies rely on the addition of metals or other ligands into the
design strategy relied on a computational docking procedure otherwise soluble protein building blocks [35,39,40]. These
that restrained the rigid body and conformational sampling approaches offer the additional advantage that the metals
to configurations obeying the target symmetry [37,38]. Mul- or ligands can be chelated or scavenged to revert the
tiple crystal structures of the designed proteins revealed protein assemblies to their soluble building blocks.
that the resulting assemblies matched the design models at The surfaces of protein building blocks can also be
atomic-level resolution, exhibiting backbone RMSDs of decorated with various functional groups to generate de-
about 1 Å or less. It should be noted that the initially sirable properties. In one case, a rhodamine chemical
designed sequence for the tetrahedral case yielded an as- group was conjugated to building blocks to promote asso-
sembly that was slightly less accurate (backbone RMSD of ciation between protein layers [35]. In another example,
2.7 Å); introducing three additional mutations yielded the protein sidechains in a repeating pattern along a helical
more accurate assembly. This study demonstrated that peptide were chosen to adhere to the periodic pattern of
designing interfaces based on many weak interactions over carbon atoms in single-walled carbon nanotubes [41]. The
large interface areas – similar to the interfaces found in helical peptides wrapping around the carbon nanotubes
natural protein assemblies – can result in highly ordered further served as a periodic registry for gold nanoparticle
and accurately assembled structures. However, the two attachment, highlighting the potential for generating hy-
successes were accompanied by 39 other designed proteins brid bio-nanomaterials.
658
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) Twofold Threefold (c) (d)


35.2

Computaonal docking
and interface design

13 nm
Oligomeric protein Self-assembling
building blocks protein nanomaterials

(b) 54.7
Computaonal docking
and interface design

11 nm
TRENDS in Cell Biology

Figure 5. Accurate design of large symmetric protein assemblies based on novel interface design. Among the wide variety of symmetric architectures that can be obtained
by interface design, two large cubic assemblies have been demonstrated [34]. Crystal structures of designed (a) 24-subunit octahedral and (b) 12-subunit tetrahedral
assemblies are shown, viewed along their twofold and threefold symmetry axes. Within each assembly, individual trimers are depicted in different colors. The designed
interfaces [regions boxed in (a) and (b)] in the crystal structures (green/magenta) of the (c) octahedral and (d) tetrahedral assemblies closely agree with the designed models
(white). Backbone RMSDs between the observed structures and the designed models were 1.1 Å and 0.6 Å over all subunits for the octahedral and tetrahedral assemblies,
respectively.

Further variations for designing protein assemblies have challenging area of extended 3D materials. For strategies
been conceptualized. In one example, the building blocks that rely on creating new interfaces by computational
comprise smaller fragments of natural proteins, which design, the key challenge is in achieving geometrically
would then be stitched together to produce molecules having precise interfaces. The successful design of a protein crys-
specific self-association properties [42]. Although this ap- tal based on an a-helical polypeptide [36] suggests that
proach has not been realized yet in practice, it highlights the this may be relatively straightforward for protein building
variety of creative ideas being brought to bear on the prob- blocks consisting of simple motifs. By comparison, design-
lem of designing ordered assemblies. ing precise interfaces into larger proteins of arbitrary
Different design strategies offer their own advantages shape appears to present more complex problems. Early
and disadvantages. Recent studies emphasize key distinc- experiments showed that single rings of protein oligomers
tions between the two main strategies discussed here – could be reliably converted to double rings by incorporat-
oligomeric fusion and interface design. The former ap- ing a few nonpolar residues on the surface, but the assem-
proach avoids the introduction of mutations within natu- blies obtained generally deviated from the designs in
rally occurring protein domains, and hence minimizes the detail [26]. King et al. [34] twice succeeded in designing
chance that the natively folded structure will be disrupted. extensive, geometrically-accurate interfaces into globular
On the other hand, de novo interface design should ulti- protein subunits, which consequently gave rise to large,
mately enable the use of arbitrary protein domains as symmetric cubic assemblies in very close agreement with
building blocks, without being limited to those natural the designed models. However, the successful designs
oligomers offered by nature. were only a small fraction of the total designs attempted,
and in one of the two successful designs a second round of
Concluding remarks sequence mutations was necessary to fine-tune the initial
If engineered protein assemblies are to become a reliable design. The successes (and accompanying failures) of var-
technology for real-life applications, the success rate of the ious recent design efforts suggest that further improve-
strategies discussed here will need to be improved. For the ments in interface design algorithms could have an
oligomeric fusion strategies, the major challenge is to important impact.
control better the relative orientation of the fusion part- An ultimate challenge will be to create a large, complex
ners. This is especially true for the helix-fusion strategy assembly without relying on existing oligomerization
given that the recent crystal structure of the designed cage interfaces or well-known self-associating motifs. This will
revealed considerable bending and twisting of the helical require designing at least two distinct interfaces into a
linkers [18]. For the symmetry-matching fusion strategy, naturally monomeric protein. In addition to the high accu-
although the presence of multiple linkers between joined racy required, the need to introduce multiple interfaces
oligomers promotes alignment of the shared symmetric compounds the success-rate problem, and also increases
axes, twisting about this axis may be harder to control the likelihood that the sequence changes will be so many as
[22]. Such rotational flexibility may need to be dealt with if to compromise protein folding and stability. The latter
a high success rate is to be achieved, particularly in the issue may call for more careful consideration of which
659
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

protein folds might be the most suitable as starting tem- 14 Crick, F.H.C. and Watson, J.D. (1956) Structure of small viruses.
Nature 177, 473–475
plates for design.
15 Wukovitz, S.W. and Yeates, T.O. (1995) why protein crystals favor
The success-rate problem might be mitigated if efficient some space-groups over others. Nat. Struct. Biol. 2, 1062–1067
screening methods can be developed. King et al. imple- 16 Padilla, J.E. et al. (2001) Nanohedra: using symmetry to design self
mented a multistep screening method to identify which of assembling protein cages, layers, crystals, and filaments. Proc. Natl.
their many computationally designed protein sequences Acad. Sci. U. S. A. 98, 2217–2221
17 Plevka, P. et al. (2012) Crystal structure of human enterovirus 71.
were soluble and compatible with self-assembly [34]. Al-
Science 336, 1274
though their screening protocol, which involved gel elec- 18 Lai, Y.T. et al. (2012) Structure of a 16-nm cage designed by using
trophoresis or liquid chromatography, was suitable for the protein oligomers. Science 336, 1129
number of designs examined (50), design strategies that 19 Ringler, P. and Schulz, G.E. (2003) Self-assembly of proteins into
exploit much larger sequence pools could benefit from the designed networks. Science 302, 106–109
20 Usui, K. et al. (2009) Nanoscale elongating control of the self-assembled
development of higher-throughput screening strategies protein filament with the cysteine-introduced building blocks. Protein
[43]. Sci. 18, 960–969
The next stage in developing designed protein assemblies 21 Raman, S. et al. (2006) Structure-based design of peptides that self-
will need to focus on applications [44,45]. The possibilities assemble into regular polyhedral nanoparticles. Nanomedicine 2,
are myriad, arising from different combinations of function- 95–102
22 Sinclair, J.C. et al. (2011) Generation of protein lattices by fusing
al elements (such as catalytic sites or recognition motifs) proteins with matching rotational symmetry. Nat. Nanotechnol. 6,
with various types of spatial organization, such as encapsu- 558–562
lation or ordered display [46,47]. Finite structures such as 23 Leaver-Fay, A. et al. (2011) Rosetta3: an object-oriented software suite
cages and shells invite the idea of encapsulation, as well as for the simulation and design of macromolecules. Methods Enzymol.
487, 545–574
targeted delivery based on externally displayed motifs
24 Fleishman, S.J. et al. (2011) Computational design of proteins
[48,49]. Motifs displayed on the surface of large assemblies targeting the conserved stem region of influenza hemagglutinin.
could be useful in their own right, for example in the design Science 332, 816–821
of synthetic vaccines [50]. Extended materials offer addi- 25 Huang, P.S. et al. (2007) A de novo designed protein-protein interface.
tional modes of spatial organization [51]. Numerous areas of Protein Sci. 16, 2770–2774
26 Grueninger, D. et al. (2008) Designed protein–protein association.
synthetic biology could be advanced by materials presenting
Science 319, 206–209
ordered arrangements of enzymes or receptors [52,53]. 27 Radford, R.J. and Tezcan, F.A. (2009) A superprotein triangle driven by
Across all the possible architectures that might be explored, nickel(II) coordination: exploiting non-natural metal ligands in protein
the ability to trigger assembly or disassembly should enable self-assembly. J. Am. Chem. Soc. 131, 9136–9137
a broad range of responsive materials [54]. 28 Salgado, E.N. et al. (2010) Metal templated design of protein interfaces.
Proc. Natl. Acad. Sci. U. S. A. 107, 1827–1832
29 Der, B.S. et al. (2012) Metal-mediated affinity and orientation
Acknowledgments specificity in a computationally designed protein homodimer. J. Am.
The authors thank Jennifer Padilla for helpful discussions and critical Chem. Soc. 134, 375–385
reading of the manuscript. 30 Salgado, E.N. et al. (2010) Metal-directed protein self-assembly. Acc.
Chem. Res. 43, 661–672
References 31 Stranges, P.B. et al. (2011) Computational design of a symmetric
1 Schwille, P. (2011) Bottom-up synthetic biology: engineering in a homodimer using beta-strand assembly. Proc. Natl. Acad. Sci. U. S. A.
tinkerer’s world. Science 333, 1252–1254 108, 20562–20567
2 Tan, C. et al. (2009) Emergent bistability by a growth-modulating 32 Zaccai, N.R. et al. (2011) A de novo peptide hexamer with a mutable
positive feedback circuit. Nat. Chem. Biol. 5, 842–848 channel. Nat. Chem. Biol. 7, 935–941
3 Elowitz, M.B. and Leibler, S. (2000) A synthetic oscillatory network of 33 Korendovych, I.V. et al. (2010) De novo design and molecular assembly
transcriptional regulators. Nature 403, 335–338 of a transmembrane diporphyrin-binding protein complex. J. Am.
4 Zhang, S.G. et al. (2002) Design of nanostructured biological materials Chem. Soc. 132, 15516–15518
through self-assembly of peptides and proteins. Curr. Opin. Chem. Biol. 34 King, N.P. et al. (2012) Computational design of self-assembling
6, 865–871 protein nanomaterials with atomic level accuracy. Science 336,
5 Yeates, T.O. and Padilla, J.E. (2002) Designing supramolecular protein 1171–1174
assemblies. Curr. Opin. Struct. Biol. 12, 464–470 35 Brodin, J.D. et al. (2012) Metal-directed, chemically tunable assembly
6 Zhang, S.G. (2003) Fabrication of novel biomaterials through molecular of one-, two- and three-dimensional crystalline protein arrays. Nat.
self-assembly. Nat. Biotechnol. 21, 1171–1178 Chem. 4, 375–382
7 Petka, W.A. et al. (1998) Reversible hydrogels from self-assembling 36 Lanci, C.J. et al. (2012) Computational design of a protein crystal. Proc.
artificial proteins. Science 281, 389–392 Natl. Acad. Sci. U. S. A. 109, 7304–7309
8 Ogihara, N.L. et al. (2001) Design of three-dimensional domain-swapped 37 Andre, I. et al. (2007) Prediction of the structure of symmetrical protein
dimers and fibrous oligomers. Proc. Natl. Acad. Sci. U. S. A. 98, assemblies. Proc. Natl. Acad. Sci. U. S. A. 104, 17656–17661
1404–1409 38 Wang, C. et al. (2007) Protein-protein docking with backbone flexibility.
9 Woolfson, D.N. (2010) Building fibrous biomaterials from alpha-helical J. Mol. Biol. 373, 503–519
and collagen-like coiled-coil peptides. Biopolymers 94, 118–127 39 Carlson, J.C.T. et al. (2006) Chemically controlled self-assembly of
10 Ghadiri, M.R. et al. (1993) Self-assembling organic nanotubes based on protein nanorings. J. Am. Chem. Soc. 128, 7630–7638
a cyclic peptide architecture. Nature 366, 324–327 40 Fegan, A. et al. (2010) Chemically controlled protein assembly:
11 Ryadnov, M.G. and Woolfson, D.N. (2003) Introducing branches into a techniques and applications. Chem. Rev. 110, 3315–3336
self-assembling peptide fiber. Angew. Chem. Int. Ed. Engl. 42, 3021– 41 Grigoryan, G. et al. (2011) Computational design of virus-like protein
3023 assemblies on carbon nanotube surfaces. Science 332, 1071–1076
12 Levy, E.D. et al. (2008) Assembly reflects evolution of protein 42 Tsai, C.J. et al. (2007) Principles of nanostructure design with protein
complexes. Nature 453, U1262–U1266 building blocks. Proteins 68, 1–12
13 Goodsell, D.S. and Olson, A.J. (2000) Structural symmetry and protein 43 Worsdorfer, B. et al. (2011) Directed evolution of a protein container.
function. Annu. Rev. Biophys. Biomol. Struct. 29, 105–153 Science 331, 589–592

660
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

44 Woolfson, D.N. and Mahmoud, Z.N. (2010) More than just bare scaffolds: 50 Smith, M.L. et al. (2006) Modified tobacco mosaic virus particles as
towards multi-component and decorated fibrous biomaterials. Chem. scaffolds for display of protein antigens for vaccine applications.
Soc. Rev. 39, 3464–3479 Virology 348, 475–488
45 Howorka, S. (2011) Rationally engineering natural protein assemblies 51 Ilk, N. et al. (2011) S-layer fusion proteins – construction principles and
in nanobiotechnology. Curr. Opin. Biotechnol. 22, 485–491 applications. Curr. Opin. Biotechnol. 22, 824–831
46 Tschiggerl, H. et al. (2008) Exploitation of the S-layer self-assembly system 52 Delebecque, C.J. et al. (2011) Organization of intracellular
for site directed immobilization of enzymes demonstrated for an reactions with rationally designed RNA assemblies. Science 333,
extremophilic laminarinase from Pyrococcus furiosus. J. Biotechnol. 470–474
133, 403–411 53 Dueber, J.E. et al. (2009) Synthetic protein scaffolds provide modular
47 Minten, I.J. et al. (2009) Controlled encapsulation of multiple proteins control over metabolic flux. Nat. Biotechnol. 27, 753–759
in virus capsids. J. Am. Chem. Soc. 131, 17771–17773 54 de la Rica, R. and Matsui, H. (2010) Applications of peptide and
48 Tong, G.J. et al. (2009) Viral capsid dna aptamer conjugates as protein-based materials in bionanotechnology. Chem. Soc. Rev. 39,
multivalent cell-targeting vehicles. J. Am. Chem. Soc. 131, 11174–11178 3499–3509
49 Han, M. et al. (2011) Targeted vault nanoparticles engineered with an 55 Yeates, T.O. (2011) Nanobiotechnology: protein arrays made to order.
endosomolytic peptide deliver biomolecules to the cytoplasm. ACS Nat. Nanotechnol. 6, 541–542
Nano 5, 6128–6137

661
Review

Special Issue – Synthetic Cell Biology

Designing biological
compartmentalization
Anna H. Chen1 and Pamela A. Silver1,2
1
Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
2
Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA

Intracellular organization is a key factor in cell metabo- designing compartmentalization, especially in building
lism. Cells have evolved various organizational systems bacterial microcompartments and RNA scaffolds. We also
to solve the challenges of toxic pathway intermediates, analyze the degree to which these mimic natural systems
competing metabolic reactions, and slow turnover rates. and discuss how they aid in our understanding of the
Inspired by nature, synthetic biologists have utilized pro- biological organization of the cell.
teins, nucleic acids, and lipids to construct synthetic
organizational systems that mimic natural systems. Many The need for intracellular organization
of these systems have been applied to metabolic path- Cells face many challenges that benefit from compartmen-
ways and shown to significantly increase the production talization (Figure 1a). First, some enzymes, such as ribu-
of industrially and commercially important chemicals. lose 1,5-bisphosphate carboxylase oxygenase (RuBisCO)
Further engineering and characterization of synthetic [13], suffer from slow turnover, which results in flux imbal-
organizational systems will allow us to better understand ances or bottlenecks in pathways. Reliance on such
native cellular strategies of spatial organization. Here, we enzymes may require establishing local concentration gra-
discuss recent advances and ongoing efforts in designing dients of substrates. This would increase reaction rates to
and characterizing synthetic compartmentalization sys- support adequate pathway flux [14]. Second, diffusion of
tems to mimic natural strategies and increase metabolic volatile intermediates through the cell membrane results
yields of engineered pathways. in their loss from the cell [15]. Third, biosynthetic path-
ways can generate toxic intermediates that inhibit growth,
Compartmentalization benefits natural and engineered such as hydrogen sulfide accumulated during bacterial
systems sulfur metabolism [16]. Finally, metabolites can partici-
Biological complexity requires varying degrees of organi- pate in multiple competing reactions, reducing their avail-
zation. Cells require spatial organization to perform the ability for any single pathway. An example of this is
various enzymatic reactions and processes necessary to malonyl-CoA, an intermediate that is consumed in fatty
sustain life [1]. This is achieved through compartmentali- acid and phospholipid production but is also used in the
zation, the physical separation of biological reactions. biosynthesis of polyketides and flavonoids [17].
Examples of compartmentalization include membrane-
bound organelles, bacterial microcompartments [2,3], mul-
tienzyme complexes, and others [4,5]. Nature’s solutions
Inspired by nature, synthetic biologists have recently To deal with these challenges, nature has evolved compart-
devised strategies to mimic cellular organizational sys- mentalization strategies (Figure 1b), such as large enzyme
tems. These synthetic systems have been predominantly complexes [10,18,19] and organelles [2,20], to spatially
designed toward metabolic engineering of pathways, har- organize metabolism. In eukaryotes, compartmentaliza-
nessing the capability of cells to produce industrially [6,7] tion in the form of membrane-bound organelles is common.
or pharmaceutically useful [8,9] compounds. The peroxisome, for example, encapsulates reactions that
In this review, we describe the various difficulties faced generate or consume hydrogen peroxide, a toxic interme-
by the cell when performing metabolic reactions and natu- diate from the breakdown of organic substrates in oxida-
ral compartmentalization systems that solve these pro- tive reactions [21].
blems. We review recent advances in designing synthetic Until recently, prokaryotes were generally thought to
compartments that provide modular solutions to overcome lack internal organization [22]. However, researchers have
these same challenges. These systems capture the benefits recently discovered different types of bacterial microcom-
of spatial organization and apply them to engineered path- partments that partition the internal space of the bacterial
ways. This has also recently been reviewed in [10–12]. Our cell for specialized functions [2,23]. In cyanobacteria and
review will discuss the latest progress and challenges in other autotrophic prokaryotes, carboxysomes encapsulate
RuBisCO and carbonic anhydrase, enzymes involved in the
rate-limiting step of the Calvin cycle [24,25] (Figure 2a).
Corresponding author: Silver, P.A. (pamela_silver@hms.harvard.edu).
Keywords: compartmentalization; synthetic scaffolds; bacterial microcompartments;
These proteinaceous microcompartments are the primary
biological organization. ‘carbon-concentrating mechanism’ in these bacteria. They
662 0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.07.002 Trends in Cell Biology, December 2012, Vol. 22, No. 12
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Key: = enzymes
(a)
(2) Escape through membrane = metabolites

(1) Flux imbalance

Enzyme 1 Enzyme 2 Enzyme 3


Intermediate A Intermediate B Product

Side Product
Enzyme 4

(3) Toxicity
(4) Compeng reacons

Cell

(b)

(1) Increase local Compartmentalizaon


(2) Sequester concentraon System
intermediates
Enzyme 2
Enzyme 1
Enzyme 3

Intermediate A Intermediate B
(3) Reduce toxicity Product

(4) Reduce
compeon
Enzyme 4
Cell
TRENDS in Cell Biology

Figure 1. Compartmentalization: nature’s solution to various challenges. (a) Nature faces many challenges when conducting the chemical reactions of the cell. (1) Differing
enzyme kinetics may result in flux imbalances. (2) Intermediates may be lost through the cell membrane. (3) Toxic intermediates can result in growth inhibition. (4)
Competing reactions can divert flux through undesired pathways. (b) Compartmentalization systems specifically solve challenges 1–4, respectively, by: (1) creating areas of
local concentrations to favor reaction kinetics; (2) sequestering intermediates; (3) reducing toxicity; and (4) reducing competition. (Adapted from [11].)

are proposed to help overcome the slow turnover rate of example is tryptophan synthase, a multienzyme complex
RuBisCO by providing a high local concentration of carbon that catalyzes the last two reactions in the biosynthesis of
dioxide to the enzyme [26,27]. L-tryptophan [18,31]. The intermediate, indole, is chan-
Two other bacterial proteinaceous microcompartments neled from one active site to the next without being
protect the cell from toxic aldehyde intermediates. The released into the surrounding environment. This is advan-
ethanolamine utilization (Eut) microcompartment tageous for the cell not only because indole is reactive and
sequesters acetaldehyde, a volatile and toxic intermediate easily lost through the cell membrane, but also because, in
of the ethanolamine utilization pathway [28]. Likewise, the absence of indole, tryptophan synthase catalyzes
the 1,2-propanediol utilization (Pdu) microcompartment dehydration of serine to pyruvate at 5% the rate of trypto-
encapsulates propionaldehyde, minimizing its toxicity phan formation [32]. Other multi-enzyme complexes found
[29]. These and numerous other bacterial microcompart- in nature include polyketide synthase [33], carbamoyl
ments have been found in approximately 400 microbial phosphate synthetase [34], and cellulosomes [35], which
genomes [2]. may function similarly by increasing reaction kinetics and
Another method of compartmentalization found in na- reducing the loss of intermediates.
ture is multienzyme complexes, which directly link
enzymes involved in a given pathway. Ideally, this results Synthetic compartmentalization
in substrate channeling, the process by which intermedi- The goal of metabolic engineering is to optimize a given
ates are directly transferred between the active sites of two biosynthetic pathway to increase production of a particular
enzymes that catalyze sequential reactions in the pathway substance [7,36]. Many of these pathways present the same
[30]. Substrate channeling prevents the loss of intermedi- challenges of toxic intermediates, competing reactions,
ates and minimizes competing cross-reactions. A classic and flux imbalances found in nature [10,14]. Therefore,
663
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) Shell RuBisCO

10 nm
CA 2 µm
Pentameric verces
Hexamers with
pores regulang transport

PDZ
(b) (c) HMGR

PDZ
HMGR

}
HMGR
HMGS

PDZ
GBD
AtoB

HMGR SH3 SH3


Enzymes

PDZ
HMGR

AtoB
HMGS
Pepde
}

AtoB
GBD SH3 SH3

HMGS
Ligands PDZ

( )( ) ( ) }

HMGS
HMGS

HM
PDZ
GBD Scaffold

AtoB
SH3 PDZ

HMGS
SH3

HMGS
GBD

AtoB

HMGRGR
x y z SH3

HMGR

HMGR

HMGS
HMGS
GBD SH3 SH3 PDZ

PD
Z
GR PDZ
HM HM
GR SH3

PDZ
SH3 GBD

TRENDS in Cell Biology

Figure 2. Protein scaffolds: utilizing nature’s building material for functional complexes. Nature compartmentalizes reactions via enzyme complexes. Proteins often serve
as scaffolds, bringing together pathway enzymes and increasing flux. (a) Bacterial microcompartments are protein structures that encapsulate reactions [2]. Carboxysomes
contain ribulose 1,5-bisphosphate carboxylase oxygenase (RuBisCO) and carbonic anhydrase. The shell comprises pentamers and hexamers, some of which contain pores
that are thought to regulate substrate transport (left). Carboxysomes spatially align in the cell [51] (right). Heterologous expression of microcompartments and further
engineering of targeting and transport has the potential for making microfactories that may be capable of encapsulating various reactions. (Left: reprinted from [46] with
permission from the authors. Right: image courtesy of David F. Savage, Berkeley, CA, USA). (b) Inspired by natural functional complexes, synthetic protein scaffolds were
created [40]. Eukaryotic protein–protein interaction domains (GBD, SH3, PDZ) were expressed as a single polypeptide, creating a synthetic scaffold. Modular protein
domain–ligand interactions were used to colocalize three enzymes of the mevalonate pathway: AtoB, HMGS, and HMGR. The stoichiometries of the enzymes could be
controlled by changing the numbers (x, y, z) of each of the scaffold domains. Scaffolds balance metabolic flux, sequester a toxic intermediate, HMG-CoA, and create a high
local concentration of intermediates, effectively mimicking nature’s strategies of compartmentalization. (c) Scaffolds are hypothesized to oligomerize into large complexes
that resemble metabolite microdomains, trapping intermediates in the interior and quickly converting them to product before they escape [11].

synthetic biologists have drawn inspiration from nature to on solid substrates in vitro [39], and direct enzyme fusion
design synthetic compartmentalization systems. [10].
Inspired by naturally existing enzyme complexes, a syn-
Compartmentalization by tethering using protein thetic protein scaffold, capable of colocalizing enzymes and
scaffolds increasing product titers, was produced [40] (Figure 2b).
Nature uses proteins and protein–protein interactions to Eukaryotic protein–protein interaction domains were fused
build functional multienzyme complexes. In some cases, to form scaffolds. Each of the domains selectively recruited
non-catalytic scaffold proteins are used to assemble these enzymes fused to cognate peptide ligands, colocalizing them
complexes. One example of such a scaffold protein is Ste5, to the scaffold.
which selectively brings together MAPKK Ste7 and its Protein scaffolds have been tested on various pathways
substrate MAPK Fus3, promoting phosphorylation in this and increases in titers were shown. The mevalonate path-
signaling cascade and disfavoring competing substrates way, for example, produces a toxic intermediate and suf-
[37]. Another example is scaffoldins, which organize pro- fers from flux imbalance due to differing enzyme turnover
tein subunits into the cellulosome and assist in the break- rates. Scaffolding increased product yields by 77-fold [40].
down of cellulose [35]. Bacterial microcompartments, large The glucaric acid pathway was also scaffolded, and 5-fold
assemblies comprising thousands of protein subunits, also improvement was seen over a high baseline titer of 0.6 g/L
function as organizational devices in the cell. Attempts to [41]. This pathway requires high levels of intermediate
mimic natural protein scaffolding mechanisms have in- myoinositol to drive myoinositol oxygenase kinetics. A final
cluded crosslinking enzymes [38], enzyme immobilization example is biological hydrogen production, which suffers
664
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

from competing reactions and requires close contact of the carboxysomes, diffusion of small molecules across the shell
two enzymes involved. This pathway benefited by about is likely, but pores that suggest active regulation of sub-
fivefold from protein scaffolding [42]. In principle, these strate crossing have also been revealed in crystal struc-
synthetic protein scaffolds can replicate natural compart- tures [45]. Heterologously expressed microcompartments,
mentalization strategies found in multienzyme complexes, isolated from their native cellular environment, may help
thereby alleviating the problems sometimes found in engi- us better understand these mechanisms.
neered metabolic reactions. Spatial organization of microcompartments can be reg-
The extent of the increase in yields due to scaffolding ulated within a cell, providing the cell with another mech-
depends on scaffold stoichiometry. Nature uses gene du- anism to optimize metabolic processes. For instance,
plication events and evolution of protein–protein interac- carboxysomes are aligned along the major axis of the cell
tions, events that operate on a long timescale, to change [51] (Figure 2a, right) and the bacterial cytoskeletal com-
the ratio of each of the enzymes in multienzyme complexes. ponent ParA is required for proper alignment to occur. The
Synthetic scaffolds similarly enable us to build and test exact interaction between the cytoskeleton and the bacte-
many different enzyme stoichiometries, but in a short rial microcompartment remains unknown and is an active
period of time, to determine the optimal ratio for produc- area of research. This knowledge would enable the design
tion of a compound of interest. Varying the number of each and construction of a novel synthetic cytoskeletal scaffold-
interaction domain in the scaffold was shown to dramati- ing device. By inducing polymerization and depolymeriza-
cally affect product titers [40]. In this way, designing tion of cytoskeletal proteins in response to substrate
stoichiometries and geometries of synthetic protein scaf- availability or other environmental signals, we may be
folds may result in more effective compartmentalization able to control the spatial organization and number of
mechanisms. compartments tethered to this synthetic cytoskeleton. This
Although the exact mechanism by which protein scaf- may allow dynamic regulation of metabolic reactions in
folds increase yields remains unknown, it has been hypoth- synthetic microcompartments.
esized that they may form higher order complexes due to Another bacterial microcompartment-like encapsula-
enzyme oligomerization [11] (Figure 2c), similar to natu- tion device was recently engineered. A lumazine synthase
rally occurring metabolite microdomains, such as Ca2+ or capsid from Aquifex aeolicus was heterologously expressed
cAMP microdomains [43,44]. If true, intermediates pro- in E. coli [52]. Lumazine synthase, an enzyme catalyzing
duced within the complex would be quickly consumed riboflavin synthesis, forms icosahedral assemblies that do
before they could escape into the cellular milieu, thereby not naturally encapsulate other enzymes. However, elec-
reducing their toxicity. This would allow protein scaffolds trostatic interactions were engineered, enabling it to en-
to more closely approximate bacterial microcompartments. capsulate a toxic enzyme, HIV protease. Directed evolution
was used to select for a capsid with higher loading capacity.
Compartmentalization using protein encapsulation In addition, the assembly state of lumazine synthase was
Bacterial microcompartments are relatively closed to the controlled through engineered point mutations, which
surrounding environment with protein-based pores that changed the quaternary structure of the capsid, thereby
likely regulate transport of substrate into and out of the changing the size and structure of the capsid, without
proteinaceous shell (Figure 2a) [45]. This strategy effec- disrupting the secondary or tertiary structure [53]. The
tively isolates reaction intermediates from the rest of the ability to insulate toxicity from the cytoplasm using a
cellular milieu and could create high local concentrations of designable compartmentalization system is important
substrates, thereby increasing product yields. for metabolic engineering. It allows the expression of het-
Microcompartments have been heterologously erologous pathways that may otherwise be toxic, or even
expressed and could be used to encapsulate foreign path- lethal, to the cell.
ways. As such, these microcompartments have the poten- Overall, protein assemblies have been used effectively
tial to increase pathway flux, making them useful tools for to organize metabolic reactions and increase product
metabolic engineering. For example, carboxysomes from yields. Thus far, however, such increases have been low.
cyanobacteria (Figure 2a) were heterologously expressed Therefore, the present challenge is to further increase
in Escherichia coli and the encapsulated RuBisCO was yields to levels high enough for industrial applications.
functional in in vitro assays [46]. The Eut and Pdu micro- To do so, the mechanism of scaffold action must be further
compartments from Salmonella have also been expressed elucidated. In addition, scaffolds have been applied to only
in E. coli [47,48]. a few select pathways as proof of principle. Generality and
Many challenges remain before heterologously scalability of scaffolds will require a better understanding
expressed microcompartments can be utilized in metabolic of localization to synthetic scaffolds.
engineering. Targeting of foreign enzymes to microcom-
partments is an area of ongoing research and localization Compartmentalization using nucleic acids
sequences have been found for the Pdu and Eut micro- DNA and RNA nanotechnology is a field of research that
compartments [47,49]. However, although associations be- has many promising applications in medicine and industry
tween carboxysome shell proteins and other protein [54–56]. Short strands of DNA or RNA can fold into various
components have been shown [50], targeting novel structures or assemble into dimer or multimer building
enzymes to the carboxysome remains not well understood blocks (Figure 3c). These building blocks can then poly-
[46]. Another challenge is elucidating the mechanism of merize into various 3D structures in vitro, including
substrate transport across the microcompartment shell. In simple structures such as sheets, as well as more complex
665
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b) Hydrogenase Ferredoxin } Enzyme


Protein
} adapter

} RNA aptamer

} Scaffold

{
(c)

Dimerize
Polymerize
{

Dimerizaon
{

domain
Polymerizaon
domain
TRENDS in Cell Biology

Figure 3. Nucleic acid scaffolds: mimicking the diverse geometries found in nature. (a) DNA and RNA nanotechnology can produce many different structures in vitro,
including tubes and sheets [86]. Although nucleic acids are not frequently used in nature for scaffolding, the ease with which synthetic biologists can design and build these
structures make them a versatile tool for building synthetic scaffolds that can mimic various natural compartmentalization strategies of different geometries. (Adapted, with
permission, from [86].) (b) Synthetic RNA scaffolds were designed in silico and expressed in vivo [75]. Binding between RNA aptamers and protein adapters facilitated
recruitment of enzymes to the scaffold. Hydrogenase and ferredoxin, components of a hydrogen production pathway, were scaffolded and pathway output was increased.
(c) Single strands of RNA with aptamers dimerize and these building blocks polymerize into 2D sheets and nanotubes. Polymerization domains are protected with hairpins
before dimerization to prevent self-binding and assure order of assembly. The versatile architectures of RNA nanostructures in vivo allow the mimicking of various natural
compartmentalization systems.

structures such as tubes and capsules [57,58] (Figure 3a). A DNA scaffold, in the form of a plasmid, was con-
In this way, DNA and RNA nanotechnology can be used to structed. It recruited enzymes via zinc finger domains that
build complexes that structurally mimic natural organiza- bound to specific motifs on the scaffold [67]. Several
tional systems, such as enzyme channels and bacterial enzymes were tested using this system, including path-
microcompartments. ways for the production of resveratrol, 1,2-propanediol,
Nucleic acid structures can also functionally mimic nat- and mevalonate. Yields were found to increase as a func-
ural organizational systems. That is, proteins can be tar- tion of scaffold architecture, similar to protein scaffolds.
geted directly to the scaffolds. Several multienzyme systems Scaffolds with more complex geometries were con-
have been successfully organized using nucleic acids in vitro. structed using designed non-coding (nc)RNA (Figure 3b,c).
NADH-flavin mononucleotide (FMN) oxidoreductase and As a building material, RNA has several advantages.
luciferase, enzymes that catalyze two consecutive reaction Because the base-pairing interactions in RNA (and DNA)
steps, were assembled onto DNA scaffolds using streptavi- are well studied [68,69], secondary and tertiary structures of
din–biotin linkages and coupled enzymatic activity was nucleic acids can be predicted in silico [70–72] with nano-
increased relative to unscaffolded enzymes [59]. Glucose meter precision [57] and are relatively easy to design [55].
oxidase (GOX) and horseradish peroxidase (HRP) were RNA can also be expressed in large amounts by engineered
immobilized through covalent conjugation to DNA oligonu- cells and can stably exist outside the nucleus in eukaryotic
cleotides and an increase in reactivity was seen in both cells, making it portable to a wide range of hosts. Addition-
scaffolded systems compared with unscaffolded enzymes ally, RNA can recruit proteins fused to adapters via RNA
alone [60]. Other enzyme cascades have been organized aptamers, which can be evolved to bind with high specificity
using large supramolecular DNA scaffolds [61] and DNA to various ligands [73,74].
origami [62] and reactivity was found to depend on relative A single molecule of RNA (approximately 100 bases) can
enzyme positioning on the scaffold. fold into a linear discrete scaffold that mimics multien-
Although traditional in vitro assembly of DNA or RNA zyme complexes in nature [75] (Figure 3b). The RNA
nanostructures relies on physiologically unsustainable molecule comprises multiple different aptamer motifs,
temperatures and slow cool down to denature and anneal which can recruit proteins, as well as complementation
designed strands correctly [63], isothermal strategies for regions, which form the scaffold (Figure 3b). This scaffold is
both DNA and RNA were recently developed [64–66]. This relatively easy to build and characterize and is suitable for
paved the way for in vivo applications of RNA and DNA expression of specific stoichiometries of enzymes in a chain
scaffolds. [75]. Scale up, which may allow production at industrially
666
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

feasible titers, is also possible; numerous RNA aptamer few select pathways. Further application of nucleic acid
domains can be connected in a single molecule of RNA. In scaffolds will require generation of additional orthogonal
this way, almost 100 proteins were recruited in a chain for RNA aptamers or zinc finger domains. The application of
visualization experiments [76], although this has not yet RNA scaffolds, with its diverse architectures, to various
been adapted for metabolic engineering purposes. pathways will not only allow further improvement of the
An RNA nanotube was built using short single-stranded technology, but will also allow testing of the mechanism of
building blocks that polymerized in vivo [75] (Figure 3c). scaffold action. For example, the effect of scaffolding is
Here, the directionality of enzyme loading was not speci- hypothesized to be greater for non-diffusible intermediates
fied; that is, enzymes were tethered to both the interior and compared with diffusible intermediates if local high con-
the exterior of the nanotube. However, loading enzymes centration of intermediates is an important contributing
selectively to the interior has been shown in vitro [77]. If factor. Overall, RNA scaffolds are capable of becoming
such selective loading could be accomplished in vivo, a versatile, designable scaffolds that can mimic natural
modified version of this 1D scaffold could act like a pipe, systems of differing geometries, compartmentalize various
preventing intermediates from escaping and increasing metabolic pathways, and increase yields, possibly beyond
product yields. Another scaffold architecture, a 2D sheet, nature’s capabilities.
was constructed using a similar strategy and product yield
was highest when using this scaffold (Figure 3c). Although Compartmentalization using lipids
there is no directly analogous system in nature, this Lipids, often in the form of membranes, are widely used to
2D-scaffold is likely to function by increasing the number encapsulate reactions in nature. Lipid vesicles and oil
of neighbors any enzyme has in close proximity, allowing emulsions have been used to perform a wide variety of
exploration of the quantitative effect of colocalizing addi- reactions in vitro, such as gene expression [79], sequencing,
tional proteins in two dimensions; for example, by recruit- and evolution of new enzymes [80]. Even self-replicating
ing them to a membrane. lipid systems have been created: protocells that are capa-
As a demonstration of feasibility, [FeFe]-hydrogenase ble of catalyzing RNA reactions and processing ‘food’
and ferredoxin, enzymes involved in the biological produc- micelles [81]. Although these systems are effective at
tion of hydrogen [78], were recruited to RNA scaffolds. This encapsulation of reactions, targeting specific enzymes to
pathway is a useful test bed because it suffers from com- synthetic lipid vesicles remains difficult because localiza-
peting reactions and requires the enzymes to be in close tion signals or other specific targeting mechanisms to lipid
proximity [42]. Hydrogen output was found to increase as a vesicles are unknown.
function of scaffold architecture. Another compartmentalization strategy utilizes natu-
DNA and RNA scaffolds allow metabolic engineers to rally existing membrane-bound organelles. Unlike lipid
mimic many different strategies implemented by nature vesicles, many membrane-bound organelles have well
for substrate channeling and reducing competition to studied localization mechanisms [82]. One study targeted
increase pathway flux. In vitro work provides a wide array methyl halide transferase to yeast vacuoles to increase
of tested architectures and functionalities that mimic methyl iodide production [83]. The increases seen were
various natural compartmentalization systems in differ- likely to have been due to added access to a cofactor and
ent ways. This ability to design precise scaffold geometry sequestration of halogenated products. Also, targeting of
is a major advantage of nucleic acid scaffolds over protein terpenoid production pathway components to the mito-
and lipid compartmentalization systems, where structure chondria significantly increased yields [84]. The advantage
prediction and design remain difficult. However, the in of this system is that it allows engineering of eukaryotic
vitro to in vivo transition is a major challenge for nucleic cells such as commercially important yeast.
acid scaffolds and is not always possible due to many Lipid compartmentalization systems remain a relative-
constraints, such as isothermal assembly. Specific control ly unexplored area of synthetic biology, because they have
over scaffold architectures and the ability to design cus- many weaknesses compared with nucleic acid or protein
tomized shapes with RNA and DNA isothermally in vivo scaffolds. For example, controlling the architecture of
are crucial next steps in utilizing this technology to in- compartments and stoichiometries of encapsulated
crease yields. If successful, RNA and DNA may even be enzymes is challenging. Other major challenges for in vivo
used to build scaffolds in architectures not found in nature. use of lipid compartmentalization devices include secretion
Although the effects of such structures on the cell’s meta- of final products and, in the case of native organelle utili-
bolic load and the crosstalk with native systems may be zation, potential interactions with the host metabolism. A
problematic, it raises interesting possibilities for metabol- better understanding of these factors and studies in natu-
ic engineers to exceed nature’s capabilities to produce ral lipid compartmentalization systems may yield results
useful compounds. that can inform the development of synthetic lipid com-
RNA scaffolds currently have an added advantage over partments.
DNA and other types of scaffold in that the amount of
scaffold expression can be controlled. However, the effect of Using synthetic scaffolds to understand the limitations
expressing high amounts of nucleic acid on cell growth and of natural systems
viability is not well understood and titration of scaffold Studying engineered scaffolds may give us new insight into
expression has yet to be performed. the function and design principles of natural compartmen-
The generality of nucleic acid scaffolds remains to be talization strategies. For example, scaffold architecture
demonstrated, because they have been used in only a and enzyme stoichiometries were found to be important
667
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

to pathway output [40,75]. Through scaffolding pathways Box 1. Outstanding questions


with known stoichiometries and differing architectures, it
may be possible to probe the limitations of native systems  What is the mechanism by which scaffolds operate to increase
yield? Do they form higher order structures?
and even improve on them.  How can we predictably engineer scaffold architecture and control
Knocking out a native compartment, replacing it with a assembly?
synthetic one, and studying the resulting effects on the cell  For what pathways are different scaffolds most useful? Is there a
may allow us to decouple biological functions. This will pathway size limitation? Does the diffusability of intermediate
help elucidate the functions and limitations of the native substrates influence the effectiveness of scaffold action?
 How do substrates enter and exit synthetic compartments? How
system. Conversely, studying heterologous expression of are enzymes localized?
natural compartments and targeting will allow us to  How can we further improve yields using synthetic scaffold
determine the components that are necessary and suffi- technologies? How do we scale up the existing technologies to
cient for proper function. Mechanisms of localization, industrial scale production?
 Are there other undiscovered natural scaffolding systems that
assembly, and substrate gating may be elucidated from
utilize proteins, nucleic acids, and lipids? How does biological
such studies. organization function natively?
In addition, the well-studied examples of natural
compartmentalization systems have comprised primarily
proteins and lipid membranes. Synthetic systems have References
shown, for example, that nucleic acids can function as 1 Hurtley, S. (2009) Spatial cell biology. Location, location, location.
scaffolds. However, RNA and DNA are not commonly used Introduction. Science 326, 1205
2 Kerfeld, C.A. et al. (2010) Bacterial microcompartments. Annu. Rev.
as materials for scaffolding in nature. Yeast telomerase is
Microbiol. 64, 391–408
one of a few known examples of RNA serving as a flexible 3 Kerfeld, C.A. et al. (2005) Protein structures forming the shell of
scaffold for protein subunits [85]. Given findings of the primitive bacterial organelles. Science 309, 936–938
capabilities of a synthetic RNA scaffold, it may be that an 4 Berlatzky, I.A. et al. (2008) Spatial organization of a replicating
as yet to be discovered function of ncRNA, which consti- bacterial chromosome. Proc. Natl. Acad. Sci. U.S.A. 105, 14136–14140
5 Komeili, A. (2012) Molecular mechanisms of compartmentalization and
tutes 80% of total RNA, is scaffolding. Synthetic scaffolds,
biomineralization in magnetotactic bacteria. FEMS Microbiol. Rev. 36,
in this case, may help us elucidate the organizational 232–255
function of the material from which the scaffold was made. 6 Savage, D.F. et al. (2008) Defossiling fuel: how synthetic biology can
transform biofuel production. ACS Chem. Biol. 3, 13–16
Concluding remarks 7 Lee, S.K. et al. (2008) Metabolic engineering of microorganisms for
biofuels production: from bugs to synthetic biology to fuels. Curr. Opin.
Cells have evolved strategies for regulating and maintain- Biotechnol. 19, 556–563
ing proper metabolic flux. Synthetic biologists have taken 8 Chang, M.C.Y. and Keasling, J.D. (2006) Production of isoprenoid
inspiration from nature and developed synthetic systems pharmaceuticals by engineered microbes. Nat. Chem. Biol. 2, 674–681
that solve some of the same problems. From direct mimicry 9 Khalil, A.S. and Collins, J.J. (2010) Synthetic biology: applications
using proteins to more novel solutions using nucleic acids, come of age. Nat. Rev. Genet. 11, 367–379
10 Conrado, R.J. et al. (2008) Engineering the spatial organization of
synthetic biologists have devised promising strategies for metabolic enzymes: mimicking nature’s synergy. Curr. Opin.
increasing metabolic production of industrially useful che- Biotechnol. 19, 492–499
micals. However, some major challenges remain (Box 1). 11 Lee, H. et al. (2011) Spatial organization of enzymes for metabolic
For example, scaffolds must be applicable to various path- engineering. Metab. Eng. 14, 242–251
12 Agapakis, C.M. et al. (2012) Natural strategies for the spatial
ways outside the test pathways chosen in the proof-of-
optimization of metabolism in synthetic biology. Nat. Chem. Biol. 8,
principle studies conducted so far. Further increases in 527–535
yields must be obtained before scale up to industrially 13 Eichelmann, H. et al. (2009) Rubisco in planta kcat is regulated in
relevant quantities can be achieved. Utilizing multiple balance with photosynthetic electron transport. J. Exp. Bot. 60, 4077–
types of scaffold in the same system, if feasible, may help 4088
14 Jørgensen, K. et al. (2005) Metabolon formation and metabolic
accomplish this, because improvements may be made or-
channeling in the biosynthesis of plant natural products. Curr.
thogonally by each scaffold. In addition, the effect of such Opin. Plant Biol. 8, 280–291
high levels of production on cell growth must be minimized. 15 Manjasetty, B.A. et al. (2003) Crystal structure of a bifunctional
To overcome these challenges, a better understanding of aldolase-dehydrogenase: sequestering a reactive and volatile
scaffold mechanism of action and enzyme localization must intermediate. Proc. Natl. Acad. Sci. U.S.A. 100, 6992–6997
16 Liu, Y. et al. (2012) Methanogens: a window into ancient sulfur
be achieved. If these efforts prove successful, scaffolds can metabolism. Trends Microbiol. 20, 251–258
be widely applied as an orthogonal novel method of in- 17 Xu, P. et al. (2011) Genome-scale metabolic network modeling results
creasing yields in metabolic engineering efforts. In addi- in minimal interventions that cooperatively force carbon flux towards
tion, implementing and studying synthetic systems will malonyl-CoA. Metab. Eng. 13, 578–587
help us understand the native biological organization of 18 Hyde, C.C. et al. (1988) Three-dimensional structure of the tryptophan
synthase alpha 2 beta 2 multienzyme complex from Salmonella
the cell. typhimurium. J. Biol. Chem. 263, 17857–17871
19 Pfeifer, B.A. and Khosla, C. (2001) Biosynthesis of polyketides in
Acknowledgments heterologous hosts. Microbiol. Mol. Biol. Rev. 65, 106–118
The authors thank D. Ducat, C.J. Delebecque, J.E. Dueber, G. Sachdeva, 20 Martin, W. (2010) Evolutionary origins of metabolic
D.F. Savage, J. Torella, and G.C. Wu for discussions and helpful compartmentalization in eukaryotes. Philos. Trans. R. Soc. Lond. B:
suggestions. This work is supported by the National Institutes of Biol. Sci. 365, 847–855
Health (Grant #GM080177) and a Graduate Research Fellowship from 21 Gehrmann, W. and Elsner, M. (2011) A specific fluorescence probe for
the National Science Foundation to A.H.C. and by the Wyss Institute for hydrogen peroxide detection in peroxisomes. Free Radic. Res. 45, 501–
Biologically Inspired Engineering and Department of Defense to P.A.S. 506

668
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

22 Losick, R. and Shapiro, L. (1999) Changing views on the nature of the 49 Fan, C. et al. (2010) Short N-terminal sequences package proteins into
bacterial cell: from biochemistry to cytology. J. Bacteriol. 181, 4143– bacterial microcompartments. Proc. Natl. Acad. Sci. U.S.A. 107, 7509–
4145 7514
23 Yeates, T.O. et al. (2008) Protein-based organelles in bacteria: 50 Kinney, J.N. et al. (2012) Elucidating the essential role of the conserved
carboxysomes and related microcompartments. Nat. Rev. Microbiol. carboxysomal protein CcmN reveals a common feature of bacterial
6, 681–691 microcompartment assembly. J. Biol. Chem. 287, 17729–17736
24 Tanaka, S. et al. (2008) Atomic-level models of the bacterial 51 Savage, D.F. et al. (2010) Spatially ordered dynamics of the bacterial
carboxysome shell. Science 319, 1083–1086 carbon fixation machinery. Science 327, 1258–1261
25 Cannon, G.C. and Shively, J.M. (1983) Characterization of a 52 Wörsdörfer, B. et al. (2011) Directed evolution of a protein container.
homogenous preparation of carboxysomes from Thiobacillus Science 331, 589–592
neapolitanus. Arch. Microbiol. 134, 52–59 53 Chen, H-N. and Woycechowsky, K.J. (2012) Conversion of a
26 Tanaka, S. et al. (2009) Insights from multiple structures of the dodecahedral protein capsid into pentamers via minimal point
shell proteins from the beta-carboxysome. Protein Sci. 18, 108– mutations. Biochemistry 51, 4704–4712
120 54 Pinheiro, A.V. et al. (2011) Challenges and opportunities for structural
27 Badger, M.R. (2003) CO2 concentrating mechanisms in cyanobacteria: DNA nanotechnology. Nat. Nanotechnol. 6, 763–772
molecular components, their diversity and evolution. J. Exp. Bot. 54, 55 Guo, P. (2010) The emerging field of RNA nanotechnology. Nat.
609–622 Nanotechnol. 5, 833–842
28 Penrod, J.T. and Roth, J.R. (2006) Conserving a volatile metabolite: a 56 Famulok, M. and Ackermann, D. (2010) RNA nanotechnology: inspired
role for carboxysome-like organelles in Salmonella enterica. J. by DNA. Nat. Nanotechnol. 5, 634–635
Bacteriol. 188, 2865–2874 57 Aldaye, F.A. et al. (2008) Assembling materials with DNA as the guide.
29 Sampson, E.M. and Bobik, T.A. (2008) Microcompartments for B12- Science 321, 1795–1799
dependent 1,2-propanediol degradation provide protection from DNA 58 Wei, B. et al. (2012) Complex shapes self-assembled from single-
and cellular damage by a reactive metabolic intermediate. J. Bacteriol. stranded DNA tiles. Nature 485, 623–626
190, 2966–2971 59 Niemeyer, C.M. et al. (2002) DNA-directed assembly of bienzymic
30 Zhang, Y-HP. (2011) Substrate channeling and enzyme complexes for complexes from in vivo biotinylated NAD(P)H:FMN oxidoreductase
biotechnological applications. Biotechnol. Adv. 29, 715–725 and luciferase. Chembiochem 3, 242–245
31 Dunn, M.F. (2012) Allosteric regulation of substrate channeling and 60 Müller, J. and Niemeyer, C.M. (2008) DNA-directed assembly of
catalysis in the tryptophan synthase bienzyme complex. Arch. artificial multienzyme complexes. Biochem. Biophys. Res. Commun.
Biochem. Biophys. 519, 154–166 377, 62–67
32 Xu, Y. and Abeles, R.H. (1993) Inhibition of tryptophan synthase by (1- 61 Wilner, O.I. et al. (2009) Enzyme cascades activated on topologically
fluorovinyl)glycine. Biochemistry 32, 806–811 programmed DNA scaffolds. Nat. Nanotechnol. 4, 249–254
33 Tran, L. et al. (2010) Insights into protein-protein and enzyme- 62 Numajiri, K. et al. (2010) Discrete and active enzyme nanoarrays on
substrate interactions in modular polyketide synthases. Chem. Biol. DNA origami scaffolds purified by affinity tag separation. J. Am. Chem.
17, 705–716 Soc. 132, 9937–9939
34 Thoden, J.B. et al. (1997) Structure of carbamoyl phosphate 63 Castro, C.E. et al. (2011) A primer to scaffolded DNA origami. Nat.
synthetase: a journey of 96 A from substrate to product. Methods 8, 221–229
Biochemistry 36, 6305–6316 64 Afonin, K.A. et al. (2010) In vitro assembly of cubic RNA-based scaffolds
35 Fontes, C.M.G.A. and Gilbert, H.J. (2010) Cellulosomes: highly designed in silico. Nat. Nanotechnol. 5, 676–682
efficient nanomachines designed to deconstruct plant cell wall 65 Jungmann, R. et al. (2008) Isothermal assembly of DNA origami
complex carbohydrates. Annu. Rev. Biochem. 79, 655–681 structures using denaturing agents. J. Am. Chem. Soc. 130, 10062–
36 Keasling, J.D. (2010) Manufacturing molecules through metabolic 10063
engineering. Science 330, 1355–1358 66 Yin, P. et al. (2008) Programming biomolecular self-assembly
37 Zeke, A. et al. (2009) Scaffolds: interaction platforms for cellular pathways. Nature 451, 318–322
signalling circuits. Trends Cell Biol. 19, 364–374 67 Conrado, R.J. et al. (2012) DNA-guided assembly of biosynthetic
38 Sheldon, R. (2007) Enzyme immobilization: the quest for optimum pathways promotes improved catalytic efficiency. Nucleic Acids Res.
performance. Adv. Synth. Catal. 349, 1289–1307 40, 1879–1889
39 Mosbach, K. and Mattiasson, B. (1970) Matrix-bound enzymes. II. 68 Jaeger, L. and Chworos, A. (2006) The architectonics of programmable
Studies on a matrix-bound two-enzyme-system. Acta Chem. Scand. RNA and DNA nanostructures. Curr. Opin. Struct. Biol. 16, 531–
24, 2093–2100 543
40 Dueber, J.E. et al. (2009) Synthetic protein scaffolds provide modular 69 Rothemund, P.W.K. (2006) Folding DNA to create nanoscale shapes
control over metabolic flux. Nat. Biotechnol. 27, 753–759 and patterns. Nature 440, 297–302
41 Moon, T.S. et al. (2010) Use of modular, synthetic scaffolds for improved 70 Zadeh, J.N. et al. (2011) NUPACK: analysis and design of nucleic acid
production of glucaric acid in engineered E. coli. Metab. Eng. 12, 298– systems. J. Comput. Chem. 32, 170–173
305 71 Zuker, M. (2003) Mfold web server for nucleic acid folding and
42 Agapakis, C.M. et al. (2010) Insulation of a synthetic hydrogen hybridization prediction. Nucleic Acids Res. 31, 3406–3415
metabolism circuit in bacteria. J. Biol. Eng. 4, 3 72 Andronescu, M. (2003) RNAsoft: a suite of RNA secondary structure
43 Laude, A.J. and Simpson, A.W.M. (2009) Compartmentalized prediction and design software tools. Nucleic Acids Res. 31, 3416–
signalling: Ca2+ compartments, microdomains and the many facets 3422
of Ca2+ signalling. FEBS J. 276, 1800–1816 73 Ellington, A.D. and Szostak, J.W. (1990) In vitro selection of RNA
44 Baillie, G.S. (2009) Compartmentalized signalling: spatial regulation molecules that bind specific ligands. Nature 346, 818–822
of cAMP by the action of compartmentalized phosphodiesterases. 74 Tuerk, C. and Gold, L. (1990) Systematic evolution of ligands by
FEBS J. 276, 1790–1799 exponential enrichment: RNA ligands to bacteriophage T4 DNA
45 Klein, M.G. et al. (2009) Identification and structural analysis of a polymerase. Science 249, 505–510
novel carboxysome shell protein with implications for metabolite 75 Delebecque, C.J. et al. (2011) Organization of intracellular reactions
transport. J. Mol. Biol. 392, 319–333 with rationally designed RNA assemblies. Science 333, 470–474
46 Bonacci, W. et al. (2012) Modularity of a carbon-fixing protein 76 Golding, I. and Cox, E.C. (2004) RNA dynamics in live Escherichia coli
organelle. Proc. Natl. Acad. Sci. U.S.A. 109, 478–483 cells. Proc. Natl. Acad. Sci. U.S.A. 101, 11310–11315
47 Choudhary, S. et al. (2012) Engineered protein nano-compartments for 77 Douglas, S.M. et al. (2007) DNA-nanotube-induced alignment of
targeted enzyme localization. PLoS ONE 7, e33342 membrane proteins for NMR structure determination. Proc. Natl.
48 Parsons, J.B. et al. (2010) Synthesis of empty bacterial Acad. Sci. U.S.A. 104, 6644–6648
microcompartments, directed organelle protein incorporation, and 78 Ducat, D.C. et al. (2011) Rewiring hydrogenase-dependent redox
evidence of filament-associated organelle movement. Mol. Cell 38, circuits in cyanobacteria. Proc. Natl. Acad. Sci. U.S.A. 108, 3941–
305–315 3946

669
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

79 Nomura, S.M. et al. (2003) Gene expression within cell-sized lipid 83 Bayer, T.S. et al. (2009) Synthesis of methyl halides from biomass using
vesicles. Chembiochem 4, 1172–1175 engineered microbes. J. Am. Chem. Soc. 131, 6508–6515
80 Channon, K. et al. (2008) Synthetic biology through biomolecular 84 Farhi, M. et al. (2011) Harnessing yeast subcellular compartments for
design and engineering. Curr. Opin. Struct. Biol. 18, 491–498 the production of plant terpenoids. Metab. Eng. 13, 474–481
81 Zhu, T.F. and Szostak, J.W. (2009) Coupled growth and division of 85 Zappulla, D.C. and Cech, T.R. (2004) Yeast telomerase RNA: a flexible
model protocell membranes. J. Am. Chem. Soc. 131, 5705–5713 scaffold for protein subunits. Proc. Natl. Acad. Sci. U.S.A. 101, 10024–
82 Siddiqui, M.S. et al. (2012) Advancing secondary metabolite 10029
biosynthesis in yeast with synthetic biology tools. FEMS Yeast Res. 86 Douglas, S.M. et al. (2009) Self-assembly of DNA into nanoscale three-
12, 144–170 dimensional shapes. Nature 459, 414–418

670
Review

Special Issue – Synthetic Cell Biology

Directed cytoskeleton self-organization


Timothée Vignaud, Laurent Blanchoin, and Manuel Théry
Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherche en Technologies et Sciences pour le Vivant, CNRS/UJF/
INRA/CEA, 17 Rue des Martyrs, 38054, Grenoble, France

The cytoskeleton architecture supports many cellular Cytoskeleton self-organization is partially regulated by
functions. Cytoskeleton networks form complex intra- the action of proteins modulating the biochemical rules of
cellular structures that vary during the cell cycle and filament growth and interactions. The combination of sim-
between different cell types according to their physio- ple biochemical rules can lead to the formation of complex
logical role. These structures do not emerge spontane- structures [2]. Robust patterns can emerge from oriented
ously. They result from the interplay between intrinsic displacements of cytoskeleton filaments by molecular
self-organization properties and the conditions imposed motors in the absence of any external guidance [3–5].
by spatial boundaries. Along these boundaries, cytoskel- However, these autonomous self-organization processes
eton filaments are anchored, repulsed, aligned, or reor- are extremely sensitive to the presence of spatial boundary
iented. Such local effects can propagate alterations conditions (SBCs). An SBC is an external geometrical cue,
throughout the network and guide cytoskeleton assem- within or at the periphery of the network, that can locally
bly over relatively large distances. The experimental affect the self-organization of the network. For tissues, an
manipulation of spatial boundaries using microfabrica- SBC can be a frontier with an external fluid or a contact
tion methods has revealed the underlying physical pro- with bone, muscle, or other organ. For a cell, an SBC can be
cesses directing cytoskeleton self-organization. Here we a neighboring cell or extracellular matrix (ECM). For
review, step-by-step, from molecules to tissues, how the intracellular cytoskeleton networks, an SBC can be a cell
rules that govern assembly have been identified. We adhesion for the actin network, a centrosome for the mi-
describe how complementary approaches, all based crotubule (MT) network, or a frontier such as the plasma
on controlling geometric conditions, from in vitro recon- membrane or an intracellular organelle.
struction to in vivo observation, shed new light on these How an SBC can direct an autonomous self-organization
fundamental organizing principles. process is the subject of this review. We describe recent
advances in the understanding of the role of SBCs in the
Setting boundaries self-organization of actin networks and MT arrays, how
The reproducible shape and spatial organization of organs these processes are integrated in the internal organization
imply the existence of deterministic rules directing the of a cell, and how this in turn affects tissue architecture. In
assembly of complex biological structures. Organ shape the formation of cytoskeleton networks, an SBC can bias
depends on cell architecture, which is supported by cyto- monomer diffusion and thereby the assembly process ([6]
skeleton networks. The formation of defined and geomet- and references therein). Here, we focus on the role of
rically controlled intracellular structures relies on the self- geometrical constraints on the growth, orientation, anchor-
organization properties of the cytoskeleton. The contribu- age, and production of mechanical forces during cytoskele-
tion of self-organization in cell biology is vast and now well ton assembly.
documented [1]. Cytoskeleton self-organization is a process
in which the consumption (physicists would say dissipa- Actin network self-organization
tion) of energy brings the cytoskeleton away from its Actin is an asymmetric protein that can self-assemble to
thermodynamic equilibrium (i.e., a disordered mixture of form polarized actin filaments [7]. This spontaneous pro-
poorly dynamic filaments) toward defined and reproducible cess can be accelerated and temporally regulated by the
steady states. This differs from the process of self-assem- energy liberated from the release of a phosphate group
bly, in which components assemble spontaneously – with- from the nucleotide triphosphate bound to actin [8]. Actin
out an external energy source – to form a structure filaments can interact to form actin networks. Actin net-
corresponding to the thermodynamic equilibrium. Depend- works can self-organize into several types of structures in
ing on the rules regulating the interaction of cytoskeleton cells: bundles comprising aligned long filaments and mesh-
components, complex structures may self-organize in a works comprising branched and intermingled short fila-
robust manner. The purpose of much of the research ments. Bundles and meshworks form such complex
described in this review has been to identify and formulate intricate networks in cells [9] that it is difficult to identify
these rules to understand how physical principles direct the principles of their self-organization.
biological morphogenesis. Biochemists have developed alternative methods to
analyze self-organization in controlled conditions in vitro
Corresponding author: Théry, M. (manuel.thery@cea.fr).
Keywords: actin; microtubule; architecture; polarity; microfabrication;
by mixing, in defined proportions, the individual compo-
micropatterning. nents (either purified from tissues or from recombinant
0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.08.012 Trends in Cell Biology, December 2012, Vol. 22, No. 12 671
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

bacteria or yeasts). The kinetic parameters of actin poly- Filaments can become aligned by membrane tension. Two
merization measured in vitro and how these parameters filaments pushing orthogonally to a deformable membrane
vary in response to the presence of actin-associated pro- will coalesce and align to reduce the elastic energy of the
teins has provided key information about the regulation of membrane [22]. Preassembled filaments can become
actin assembly dynamics [10]. However, the rules guiding aligned by defining the anchorage positions with regular
the spatial organization of the network can be identified arrays of beads or micropillars and adding filamin to
only by using controlled geometric boundary conditions. crosslink filaments [23,24].
Filaments can become aligned in parallel or antiparallel
Symmetry break configurations by controlling the orientation of their
Mechanical constraints in an actin network can induce a growth. Surface micropatterning can be used to manipu-
symmetry break (i.e., the sudden occurrence of a singular late precisely the geometrical boundary conditions of fila-
axis in isotropic conditions in which all directions were ment growth and orientation [25]. Selective adsorption of
previously equivalent). This propensity for symmetry actin nucleation-promoting factors on micropatterned
breaking in actin networks was elegantly revealed using regions induces localized formation of a branched mesh-
a spherical glass bead coated with actin nucleation factors work. Only non-branched filaments grow out of the micro-
as a simple SBC [11,12]. Actin nucleation is induced from pattern, with their barbed ends reproducibly oriented
the bead, and the presence of capping proteins, which block outward. Steric interactions force growing filaments to
filament elongation from their fast growing end, ensures align parallel to each other, orthogonal to the nucleation
that the actin filaments are short and form a dense region (Figure 1g). Distant from the nucleation region, two
branched meshwork. As the actin filaments grow at the filaments growing toward each other in nearly opposite
bead surface, material accumulates and the stress directions tend to form antiparallel bundles; whereas two
increases in the network up to a critical value inducing filaments growing toward each other but at an oblique
its rupture [13]. The rupture creates an asymmetry in the angle tend to form parallel bundles (Figure 1g). However,
pressure applied on the bead such that the bead is dis- these tendencies can be biased because adjacent filaments
placed (Figure 1a). Repetition of this sequence of events sterically affect each other. The reorientation of filaments
induces saltatory propulsion of the bead [14,15]. during bundle formation guides adjacent filaments also to
In this experimental system, the SBC can easily be align with the bundle (Figure 1g). Bundle formation is thus
manipulated by changing its dimensional parameters. a combination of local probabilistic events, governed by
For example, the larger the bead, the shallower the curva- filament flexibility, and the propagation of the alignment
ture of the bead surface, leading to an increase in the configuration to adjacent filaments by steric interactions
critical value of network thickness before rupture [16,17] [25].
and the periodicity of the saltatory propulsion (Figure 1b). In egg extracts, biochemical conditions are less well
An asymmetric SBC can be created using ellipsoidal beads. defined but closer to intracellular conditions. Encapsula-
The difference in surface curvature of the bead biases the tion of egg extracts in membrane vesicles revealed that
location of network rupture, which occurs preferentially in filaments nucleated at the periphery move inward and
line with or orthogonal to the long axis of the bead [18] align to form a central ring. Interestingly, the ring can
(Figure 1c). Higher aspect ratios, obtained by actin nucle- form only when nucleation is restricted to the vesicle
ation on small glass rods, further increase the spatial bias periphery and not distributed evenly throughout the entire
and branched network growth is restricted to being orthog- volume. A scaling law appears to regulate the ring size in
onal to the long axis of the rod [15]. As the rod length proportion to the vesicle diameter [26] (Figure 1f).
increases, several independent networks can form, reveal-
ing the existence of a critical length for subnetwork inter- Network contraction
connections (Figure 1d). Interestingly, symmetry break Myosins are oriented motors moving toward a defined ex-
and asymmetric force production are not restricted to tremity of actin filaments. Thus, they have specific actions
branched meshworks of actin, but can also be induced depending on actin network architecture. They walk along
by the bundling and alignment of individual filaments parallel filaments, whereas they slide along antiparallel
polymerizing against the bead surface [19]. filaments in opposite directions relative to each other and
thus contract the network [27] (Figure 1i). Myosins can also
Filament alignment induce the contraction of branched meshworks, because
Several self-organization processes can induce actin fila- these networks also contain antiparallel filaments. Howev-
ment alignment in response to an SBC. Filaments can er, the rate of contraction is reduced due to the resistance
become aligned by steric interactions. When two long associated with branches and network anchoring to nucle-
filaments come close to each other, they prevent the inser- ation regions [27]. It has been shown, based on the use of
tion of a short filament between them. Long filaments will actomyosin bundles connecting beads, that the contraction
be further forced to align by the steric interactions of short rate is proportional to bundle length [28]. In more complex
filaments around them. Steric interactions between long structures comprising various types of network, the contrac-
filament bundles will then promote their orientation in line tion rate is determined by the local proportion of parallel and
with the long axis of the volume in which they are confined antiparallel bundles and branched meshwork [27]. Varia-
[20] (Figure 1e). Steric interaction of filaments freely mov- tions of these proportions in a given architecture will induce
ing on a layer of molecular motors can also result in their anisotropic contraction, although myosins are present
alignment along each other [4] and along the SBC [21]. throughout the network (Figure 1i). Therefore, an SBC
672
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) Symmetry break

Time

(b) Nucleaon size (c) Nucleaon shape (e) Container shape

(d) (f)

(g)

(h) (i)

Time Time

Key:
Acn bundles: Acn meshwork:
Acn nucleaon
Intermingled and Pressure
micropaern An-parallel filaments
branched filaments Tension
Confinement Parallel filaments
volume

TRENDS in Cell Biology

Figure 1. Actin network self-organization. (a) Actin meshwork polymerization around beads leads to symmetry breaking, meshwork rupture, and bead propulsion. (b) Bead
size regulates the period and size of meshwork rupture. (c) Bead asymmetry orients meshwork growth. (d) Bar length affects network coherence. (e) Long filaments self-
align to form bundles, which become oriented along the long axis of the container. (f) Inward flow of filaments nucleated at the vesicle periphery leads to the formation of a
ring, the size of which is in proportion with the vesicle diameter. (g) Filament nucleation and growth of micropatterned branched meshworks. The filament interaction angle
modulates the probability of association in either parallel (blue filaments) or antiparallel (red filaments) configurations. (h) Myosins induce the specific contraction and
disassembly of antiparallel bundles and branched meshworks while leaving parallel bundles unaffected. (i) Asymmetric distribution of the ratio between branched and
antiparallel networks leads to asymmetric contraction.

can define the type of network architecture, which in turn required to accelerate the process [29]. Compared with
can define its pattern of contraction. actin filaments, MTs are much more rigid and almost
straight in the dimensions of a single cell. MTs can sustain
MT network self-organization higher compression forces than actin filaments. They can
Similar to the formation of actin filaments from the self- form bundles, but they cannot form branched networks.
assembly of actin monomers, tubulin forms asymmetric MTs are not as numerous as actin filaments in the cell
dimers that can self-assemble into MTs. However, cytoskeleton. MT growth is characterized by long growth
the release of tubulin-bound nucleotide triphosphate is phases alternated with short periods of rapid shortening.
673
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

The ‘plus-end’ of the MT is much more dynamic than the assembly around DNA: the focusing of the minus-ends of
‘minus-end’, which can be attached to a MT-organizing MTs that are associated with large DNA clusters to form
center (MTOC). In most animal cells, the MT network spindle poles, and the antiparallel alignment of the plus-
forms as an aster in which MTs radiate from the MTOC. ends of MTs that are anchored at the two MTOCs such that
As cells divide, the MTOC is duplicated and the network the aster MTs overlap [37].
forms a bipolar spindle. Multimeric minus-end-directed motors, such as
dyneins, induce the formation of spindle poles. DNA pro-
Centering vides guidance cues for initial MT alignment and thus
The most straightforward way to investigate MT aster biases bipolar spindle formation [38,39]. MTs tend to align
positioning in response to an SBC has been to purify parallel to the surface of a DNA-coated bead. The intrinsic
MTOCs from cells and place them in microfabricated molecular machinery supporting spindle pole focusing and
chambers of defined dimensions [30]. Hence, the bound- mitotic spindle spatial organization is robust and initially
aries of the chamber can serve as an external SBC. As MT appeared insensitive to configuration of the DNA complex
plus-ends grow and push against the edges of a square [40]. However, extensive manipulations of the amount of
chamber, MTs are subjected to compression forces that DNA and its spatial distribution using microcontact print-
push the aster toward the geometrical center of the cham- ing revealed the DNA directing role in spindle assembly
ber [30] (Figure 2a). When fluctuations cause the MTOC to [41]. The increase in size of DNA aggregates induces
become off-center in a given direction, MT curvature and spindle lengthening (Figure 2d). Above a critical size, large
pressure increases in that direction and pushes the MTOC DNA aggregates can induce the formation of multiple poles
back toward the center. Thus, an isotropic array of MTs [41] (Figure 2d). Moderately asymmetric distribution of
pushing on peripheral barriers is sufficient to maintain the DNA is sufficient to orient spindle formation [40,41]
aster at the center of the volume in which it is confined. (Figure 2e). Long bars coated with DNA result in the
However, MTs sliding along the periphery could affect the formation of multiple repeats of spindles along the length
stability of this centering mechanism by reorienting MTs. of the bar and thus revealed the existence of an intrinsic
In such conditions, both pushing and pulling forces, by spindle width (Figure 2f). This intrinsic spindle width
minus-end-directed motors attached to the periphery, are seems to be defined by the balance between motors forcing
necessary to ensure efficient stabilization of the MT aster the focusing MT ends and the elastic reaction force due to
at the geometrical center of the SBC [31] (Figure 2a). MT bending. In a certain range of parameters defined by
By contrast, asters with opposite polarities (i.e., with the ratio between the DNA aggregate width and MT
MT plus-ends at the center of the aster) cannot adopt the length, the symmetry is broken and all MTs collapse on
same steady state. As long as MTs contacting the periphery one side of the DNA, resulting in an asymmetric configu-
are short enough to release their elastic energy by straight- ration of spindles with respect to the long axis of the bar
ening, they gently push the aster toward the center. As [41] (Figure 2f). Below this critical range, antiparallel MTs
they get longer, the compression forces in bent MTs in- from opposite poles (with the bar in between) interact to
crease. The clustering of dynamic plus-ends by kinesins at stabilize the formation of symmetric bipolar spindles;
the aster center is not strong enough to resist these forces above this range, the two spindle configurations on oppos-
and so the aster fragments. The MT network then switches ing sides of the DNA bar are independent and both form
to highly robust vortex-like structures [32] (Figure 2b). independent monopolar spindles.

Symmetry break Cellular self-organization


When an aster is trapped in a water droplet encapsulated in In cells, the organizing principles described above appear
oil, MTs cannot attach to the periphery. The spherical applicable but more difficult to reveal and investigate.
water–oil interface has minimal tangential resistance and Both actin and MT networks are regulated by hundreds
is an effective SBC along which MT can slide easily. In these of different types of binding protein. In addition, the as-
conditions, symmetry breaks in the aster configuration can sembly of actin filaments and MTs are affected by each
occur [33] (Figure 2c). In a relatively large spherical volume, other through physical and biochemical interactions. Cy-
few MTs reach the boundaries and the aster is stabilized toskeleton network assembly is regulated at the scale of a
close to the geometrical center. As the size of the spherical cell and is no longer solely dependent on local biochemical
volume is reduced, MTs tend to be longer than the container and geometrical conditions. The implication of biochemical
radius. To minimize their curvature and relax their elastic signals forces the system to break its symmetry and define
energy, MTs slip along the edges and align with the SBC an axis of polarity. Although actin or MT network assembly
[33,34]. This produces an asymmetric redistribution of MTs is more complex in the cellular context than in vitro, some
that pushes the MTOC to the periphery of the droplet [33] self-organizing principles have been identified.
(Figure 2c). Interestingly, when the rigidity of the SBC is In simple conditions as near to cellular conditions as can
reduced, clustered MTs push and deform it to the extent that be achieved in experiments in vitro, similar self-organized
a tubular protrusion can be formed [33,35,36]. structures can be observed. Cytoskeleton networks in cells
from lymphatic lines or in other cells or cell fragments on
Alignment and spindle formation non-adhesive substrates are subjected to no other geomet-
The formation of bipolar mitotic spindles also depends on rical constraints than the flexible plasma membrane. In
geometrical boundary conditions defined by DNA and cell the absence of MT networks, the actin network contracts
shape. Two mechanisms contribute to mitotic spindle and breaks symmetry after a local rupture occurs in the
674
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a)

(b)

(c)

(d) (e)

(f)

Key:
Plus-end directed motor Microtubule DNA Pressure

Minus-end directed motor (free)


Minus end anchor Tension
Minus-end directed motor (anchored)

TRENDS in Cell Biology

Figure 2. Microtubule (MT) network self-organization. (a) Aster off-centering with short MTs in a large container (left). Aster centering by MT sliding and pushing on the
container corners (middle). Highly efficient aster centering by pushing and pulling forces (right). (b) MT length-dependent aster formation and centering. Short MT ‘plus-
end’ coalescence by motors (left). Aster centering by a few MT ‘minus-ends’ reaching and pushing on container edges (middle). Aster fragmentation and vortex formation
by pushing forces exerted by long MTs on container edges (right). (c) MT length-dependent aster off-centering. Aster centering by few MT plus-ends reaching and pushing
on container edges (left). Symmetry break and aster off-centering by a few, sliding MTs pushing on container edges (middle). Cortical alignment of MTs and peripheral
localization of MT-organizing center (MTOC) due to numerous MTs sliding and pushing on container edges (right). (d) DNA cluster size regulates spindle size and pole
formation. (e) DNA cluster asymmetry regulates spindle orientation. (f) DNA cluster width regulates spindle symmetry.

network. With the symmetry breaking, an over-contracted results from the contractile force generated by myosins
region propagates in the network [42–44]. The process of rather than by the pushing force associated with actin
rupture is similar to what happens in branched meshworks polymerization. In the absence of actin networks, MTs
around beads [12], except the occurrence of the rupture pushing on a deformable membrane coalesce, align, and
675
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

break symmetry by forming a long tubular protrusion along the cell’s basal surface are coupled to the assembly
[42,43], reminiscent of their behavior in vesicles of similar structures and force distribution along the cell’s
[33,35,36]. However, in cellular conditions, actin and MT apical surface [62,63]. Aligned myofibrils tend to organize
networks interact and the SBCs are more complex that a their banding patterns in register [64].
freely fluctuating plasma membrane. The ECM and cell Asymmetric SBCs, defined in cell culture by micropat-
neighbors can represent adhesive SBCs. Hence, the precise terned adhesion sites, can lead to the development of
control and manipulation of cell adhesions, which are asymmetric actin networks. Bundles accumulate preferen-
cellular structures that interact with the cell’s structural tially along concave rather than convex cell edges [65]. As
microenvironment, reveal how these SBCs could direct the cell spreads over an adhesive region, conspicuous
intrinsic cytoskeleton self-organizing properties. contractile bundles are formed that connect this region
to other adhesive regions separated by non-adhesive
Directed shape regions [47,66,61], revealing the development of larger
Cells spreading on a defined regular array of adhesion traction forces [67] (Figure 3d). A relatively larger distance
spots revealed that the size and spacing between spots between adhesion sites leads to a reduced edge curvature
was a critical regulator of cell shape. Cells need a minimum and thicker bundles and so probably reflects a larger force
spot size to assemble focal adhesions and cannot extend between these sites [66,68] (Figure 3e).
over a maximal distance between these spots [45–48]. Cell
shape appears to result from the competition between the Directed MT network
force from adhesion-induced spreading and a reaction force The MT network adapts its dynamics to the various con-
from the cell’s elasticity and other internal contraction figurations of the actin network. MTs bend and grow along
forces [49]. However, some cells, such as fibroblasts, have actin contractile bundles, but stop growing when they
an intrinsic mechanism to regulate the length of their long reach a branched actin meshwork [69]. Interestingly, al-
axis regardless of their width, which seems to implicate though an asymmetric actin network will lead to asym-
tight crosstalk between actin and the MT network [50,51]. metric organization of MTs, the MTOC remains at its
Although cell shape elongation, cytoskeletal alignment, central location (Figure 3f). Centrosome positioning
and internal cell polarity orientation are usually correlat- appears to depend on generation of forces by dyneins on
ed, cell shape does not determine actin and MT organiza- MTs [70,71], but also on the forces generated by the less
tion. Modifying the actin network by fluid flow while characterized connections with the actomyosin network
maintaining constant shape reorients the MT network [71,72]. Centrosome central positioning is even more re-
[52]. Similarly, modifying the MT network independently markable given that a large part of the cytoplasm is
of cell shape reorients the actin network [53]. Rather, there occupied by the nucleus, on which MTs can also push
is an intricate coupling between actin and MT networks and pull. The robust mechanism by which the centrosome
that affects their respective spatial organizations and the becomes positioned at the geometrical center of the contour
axis of cell polarity. that describes the cell shape, where the actin network is
asymmetric and the nucleus occupies a large part of the
Directed actin network architecture cytoplasmic volume, remains to be elucidated.
The cellular actin network is organized by a balance be- The positioning of the nucleus in a cell in culture is off-
tween the assembly of a contractile network of aligned center and distal from the cell’s adhesion to the ECM and
filaments and the polymerization of a non-contractile the actin branched meshwork, but is proximal to the
branched meshwork. This balance appears to be finely contractile bundles [69]. Therefore, the internal polarity,
regulated by the degree of cell adhesion [54]. as revealed by the nucleus–centrosome vector, is oriented
The branched meshwork assembles at the cell periph- with respect to ECM and actin network asymmetries
ery. It is preferentially developed along convex rather than [55,69]. Biochemical disruption of the actomyosin network,
concave cell edges [55]; thus, it promotes the formation of the MT network or the nucleus–cytoskeleton connections
larger membrane deformations at a cell apex [56] can perturb polarity orientation with respect to cell–ECM
(Figure 3a), the size of which increases as the angle of SBCs [71,73,75].
the apex is reduced [57]. The centrosome and nucleus are often described as
Contractile bundles of antiparallel filaments are pres- being in the cell spreading plane in culture, but they can
ent throughout the cytoplasm. Peripheral bundles and be positioned on an axis orthogonal to this. Moreover, their
more interior bundles have distinct dynamics and contrac- relative positions can be switched on this axis, depending
tion properties. Components of peripheral bundles move on the degree of confinement imposed by the available
toward the bundle center, whereas components of interior spreading area. Indeed, the centrosome is positioned to-
bundles remain static with respect to the bundle organi- ward the apical curved surface, above the nucleus, in
zation [58] (Figure 3b). This probably reveals key differ- confined cells and below the nucleus in cells that have
ences in the polarity of filaments and thus specific spread extensively [76] (Figure 3g).
contraction properties of these two types of bundle. As cell
spreading or the cell aspect ratio increases, cell contraction Directed migration
increases [59–61]. The cell aspect ratio increase induces The asymmetric cytoskeleton organization in response to
the alignment of contractile bundles, which form struc- an asymmetric SBC can affect the direction of a motile cell.
tures such as stress fibers or myofibrils (Figure 3c). Aligned The relationship between external asymmetry and orient-
stress fibers and the associated anisotropic contraction ed motility is not straightforward, because it seems to
676
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b) (c) (d) (e)

Angle Posioning Aspect rao Adhesion Distance

(f) Centrosome XY centering (g) Centrosome Z off-centering

Side view

(h) Directed moon (i) Polarized moon

Key: Acn filaments:


An-parallel Nucleus

Parallel Centrosome
ECM micropaern Branched Microtubule

TRENDS in Cell Biology

Figure 3. Cellular self-organization. (a) Branched meshwork polymerization in acute-angled regions of the cell periphery. (b) Inward treadmilling (arrows) in peripheral actin
bundles and absence of treadmilling of internal bundles may reveal differences in filament polarities. (c) Alignment of myofibrils in response to cell shape elongation. (d)
Formation of conspicuous actin bundles along non-adhesive edges and thin actin bundles along adhesive edges. (e) Longer peripheral bundles are also thicker. (f)
Microtubules (MTs) adapt their growth to local actin structures. The centrosome maintains its central position in symmetric (left) and asymmetric environments (right). (g)
Centrosome positioned above the nucleus, close to branched actin meshwork, in spatially confined cells (top). Centrosome positioned below the nucleus, close to actin
bundles, in spread cells (bottom). (h) Cells move toward confined spaces above a certain threshold (top) and toward open spaces below that threshold (bottom). (i) Spread
cells move with the centrosome toward the front (top), whereas confined cells move with the centrosome toward the back (bottom).

depend on cell type and the degree of asymmetry. A linear whereas fibroblast speed is greater when the bar width is
track of repeated micropatterns in the shape of isosceles narrower [81]. Interestingly, the orientation of internal cell
triangles can lead cells to move toward the acute angle polarity, revealed by the position of the centrosome with
apices of these triangles [77,78]. By contrast, an elongated respect to the nucleus, also depends on the width of the
isosceles triangle with a very acute angle (several degrees adhesive micropattern. A cell migrates on a wide bar with
only) can lead cells away from the acute angle apex [79] the centrosome nearer the leading edge, whereas on a
(Figure 3h). narrow bar, the centrosome is nearer the trailing edge
Without an external bias such as those created by [82] (Figure 3i). How this centrosome positioning is related
asymmetric SBC, the direction of motility is defined by to the different types of actin organization remains to be
intrinsic cell polarization mechanisms and can be observed investigated.
in motile cells on adhesive micropatterns in the shape of The speed of migrating cells and their persistence in
bars. However, bar width affects actin network organiza- moving in a given direction are both affected in cells whose
tion. Variations in actin network assembly in response to internal polarity orientation process is defective [75]. The
bar width are cell type-specific because keratocytes need systematic connection between the actin network machin-
large transversal spreading to move relatively fast [80], ery powering cell migration and the degree of stability of
677
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

spatial organization of the internal cell polarity was fur- distribution of which directs the migration of cells with
ther supported by the observation in around a hundred respect to their neighbors. The sudden removal of SBCs,
different cell types of a correlation between cell speed and allowing a previously confined group to migrate, revealed
the persistence of the cell in maintaining their direction of that intercellular forces propagate from the migrating front
migration [83]. to the group’s rear [94] (Figure 4b). Intercellular forces did
not appear to pull cells forward but rather to orient the
Directed cell division traction force field they develop on the ECM to migrate.
The adaptation of MT network architecture in relation to An intriguing recent work revealed that global coher-
the actin network architecture and to cell shape is manifest ence can emerge in the spatial organization and collective
during cell division. The tensions in astral MTs, radiating motion of large cell groups [95]. Cells plated as multicellu-
from the spindle pole toward the cell cortex, exert a torque lar groups on micropatterned discs do not display any
on the spindle and direct its orientation. The tension in coherent global motion nor specific cell orientation. How-
these astral MTs is regulated by the presence of cortical ever, on a torus-shaped micropattern, there is a clear
cues associated with the actin network, which orient the asymmetry in cell orientations such that the long cell axes
cell division axis with respect to cell adhesion cues and the tilt at similar angles with respect to the torus center
architecture of the actin network [84–87]. Tension can also (Figure 4c). This appears to be reflected in the direction
be exerted throughout the cytoplasm and therefore be cells adopt when motile on the torus. The angular direction
proportional to astral MT length; differences in astral of cell motility at the peripheral edge of the torus (with
MT length can differ with respect to cell shape elongation positive curvature) tends to be opposite to that at the
and these variations can direct the orientation of the interior edge of the torus (with negative curvature)
division axis accordingly [88,89]. (Figure 4c). Therefore, it appears that the symmetry break
imposed by the torus arises from this directional property
Tissue self-organization of cell motility at the edges of the torus that is propagated
At the level of tissue organization, the complexity of the throughout the entire group of cells. Surprisingly, the
system increases with greater numbers of components. angular bias of endothelial cell orientation is clockwise,
Nevertheless, precise manipulation of the geometries of whereas with myoblasts it is counterclockwise. Thus, var-
SBCs has proven useful in identifying consistent self-or- iations in intracellular parameters presumably can be
ganization rules. manifested as specific asymmetries for different cell types.
However, no explanation has yet been proposed for this
Directed cell positioning geometrically simple organization resulting from a proba-
Self-organization of cells in a given space depends on the bly quite complex mechanism. One area where a mecha-
balance of mechanical forces between the cells and the nism may be identified is in the regulation of cell polarity
surrounding matrix. Two cells in contact constitute a mini- and its relationship to oriented cell motility.
mal multicellular structure where cells can form cell–matrix
adhesions (CMAs) and cell–cell adhesions (CCAs). When Directed cell polarity
confined on a homogeneous micropattern (i.e., when the cell The relationship between the locations of CCAs and CMAs
basal surface is in contact with a continuous layer of ECM), affects nucleus–centrosome axis orientation. The centro-
endothelial cells forming CCAs move regularly around each some, with respect to the nucleus, tends to adopt a more
other in the plane of the culture dish, whereas fibroblasts, distal position from CCAs and a more proximal position to
which cannot form CCAs, do not [90]. Therefore, the forma- CMAs [73,74,96] (Figure 4d). Thus, the asymmetric loca-
tion of CCAs appears to modulate the capacity of the two tions of both CCAs and CMAs are sufficient to bias the
cells to reach a mechanical balance. The two adhesive nucleus–centrosome axis [73,74]. CCAs seem to regulate
systems – CCA and CMA – within a cell can mutually affect centrosome positioning [73,96], whereas CMAs seem to
their respective localizations [91]. Two cells of a given regulate nucleus off-centering [69,73]. Both actin filaments
epithelial cell type confined on micropatterned ECM within [96] and MTs [73] have been shown to be involved in the
a defined area can move or adopt a stationary position in regulation of centrosome positioning away from CCAs.
response to subtle changes in ECM geometry [92] Therefore, the mechanisms by which the cytoskeleton
(Figure 4a). Indeed, the production of tensional forces on affects centrosome and nucleus positioning remain un-
the CCA depends on the spatial organization of the ECM. clear. In addition, the orientation of cell polarity not only
Intercellular force is higher when the CCA is close to the depends on the position of CCAs, but also on the orienta-
ECM. This directs the CCA away from the ECM and stabi- tion of intercellular force fields [97].
lizes the cell position in this configuration, which corre- Given that the self-organization of actin filament and
sponds to global minimization of the overall contractile MT networks is highly sensitive to SBCs and to the distri-
energy [92] (Figure 4a). Conversely, the formation of a bution of mechanical constraints, and that both types of
CCA prevents the formation of proximal CMAs [74,93]. network have intrinsic capacities to break symmetry, per-
The mutual exclusions of the two adhesion systems lead haps biased collective directional motility [95] results from
to their spatial segregation [91] and directs cell positioning. symmetry break in the intracellular actin networks and
the consequent asymmetric orientation of internal cell
Directed collective motion organization [98] (Figure 4e). How these polarized signals
The collective motion of a large multicellular group propagate to adjacent cells and result in collective oriented
depends on the production of intercellular forces, the spatial motility remains to be elucidated. Particularly, the role of
678
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) Directed cell posioning (c) Symmetry break in collecve migraon

(b) Directed collecve migraon (d) Directed cell polarity

Removable Removable
barrier barrier

(e) Directed morphogenesis

Side view

Key:
Intercellular
Acn filament bundle Centrosome
ECM micropaern tensional forces
ECM
Nucleus Branched acn network Microtubule tracon forces

TRENDS in Cell Biology

Figure 4. Tissue self-organization. (a) Two cells move regularly around each other (black arrows) when extracellular matrix (ECM) is present all along the periphery (left),
whereas they stop moving when the extremities of their ECM contact plane reach a region without ECM (right). The presence and absence of ECM regulate intra- and
intercellular forces in opposite ways. (b) Intercellular forces propagate from the front to the center of a migrating cell group. (c) A large multicellular group on a disk of ECM
displays no geometrical bias (left), whereas on a torus, symmetry is broken and cells bias their orientation and move (black arrows) in a coherent fashion (right). (d) Two
adherent cells orient their internal polarity away from their contact plane. (e) Speculation on coherent tissue polarity establishment. Symmetry break first occurs in the actin
network, followed by microtubule (MT) rerouting and internal polarity reorientation. The asymmetric distribution of internal forces associated with these changes is
counterbalanced by asymmetric intercellular forces, which further affect polarity in adjacent cells and propagate asymmetric orientation cues.

internal mechanics and intercellular force transmission cellular architectures and dynamics. Additional efforts
could be the key elements supporting intracellular inte- need to be initiated to connect in vitro and in vivo self-
gration of spatial signals and the establishment of coherent organized cytoskeleton networks and fully to benefit from
cell polarities in dynamic multicellular structures. the former in understanding the latter.
There is currently a gap between the few self-organized
Concluding remarks structures that have been characterized in vitro and the
SBCs play a major role in directing intrinsic cytoskeleton myriad different structures observed in cells. Efforts
self-organization properties, from the architecture of mac- should be made to reconstitute all of these structures
romolecular structures to the distribution of cells in tis- in vitro. This will become possible by: (i) using more
sues. Investigations at each scale – on isolated cytoskeleton complex protein mixtures in vitro to recapitulate their
components, more complex cell extracts, or entire cells – effects on cytoskeleton networks observed in cells; (ii)
provide complementary information. All contribute to the identifying ways to engineer controlled SBCs mimicking
establishment of a working framework, which should ulti- actual biological membrane; and (iii) modulating bio-
mately allow us to formulate the exact rules of cytoskeleton chemical signaling.
self-organization during morphogenesis. However, our un- The regulation of network disassembly is as important
derstanding of the self-organization of minimal molecular as the regulation of assembly in network dynamics. There
systems in vitro is not sufficient to account for genuine is a critical need to further understand how this network
679
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

disassembly is modulated by SBCs. Progress in this direc- 8 De La Cruz, E.M. et al. (2000) Polymerization and structure of
nucleotide-free actin filaments. J. Mol. Biol. 295, 517–526
tion should allow the reconstitution of dynamic steady
9 Xu, K. et al. (2012) Dual-objective STORM reveals three-dimensional
states in which manipulation of SBCs and network assem- filament organization in the actin cytoskeleton. Nat. Methods 9, 185–
bly–disassembly could lead to conditions in which the 188
network persistently self-renews, with its overall structure 10 Pollard, T.D. (2007) Regulation of actin filament assembly by Arp2/3
remaining unaffected. Technological developments are al- complex and formins. Annu. Rev. Biophys. Biomol. Struct. 36, 451–
477
so required to modulate SBCs in real time [47], especially
11 Yarar, D. et al. (1999) The Wiskott-Aldrich syndrome protein directs
for analyzing dynamic systems and cytoskeleton adapta- actin-based motility by stimulating actin nucleation with the Arp2/3
tion to external changes. complex. Curr. Biol. 9, 555–558
However, the considerable efforts made to understand 12 Oudenaarden, A.V. and Theriot, J.A. (1999) Cooperative symmetry-
the regulation of the self-organization properties of actin breaking by actin polymerization in a model for cell motility. Nat. Cell
Biol. 1, 493–499
filament or MT networks will not be sufficient to under- 13 van der Gucht, J. et al. (2005) Stress release drives symmetry breaking
stand their self-organization in a cellular context, because for actin-based movement. Proc. Natl. Acad. Sci. U.S.A. 102, 7847–
the two networks are not independent of each other. In- 7852
stead, the two networks are physically and biochemically 14 Bernheim-Groswasser, A. et al. (2005) Mechanism of actin-based
coupled. It is necessary to design new, controlled in vitro motility: a dynamic state diagram. Biophys. J. 89, 1411–1419
15 Achard, V. et al. (2010) A ‘‘primer’’-based mechanism underlies
biochemical assays in which the two networks can interact branched actin filament network formation and motility. Curr. Biol.
and regulate each other. Such assays should offer the 20, 423–428
possibility to manipulate the geometry of network inter- 16 Noireaux, V. et al. (2000) Growing an actin gel on spherical surfaces.
actions as well as the spatial distribution of crosslinking Biophys. J. 78, 1643–1654
17 Bernheim-Groswasser, A. et al. (2002) The dynamics of actin-
proteins and regulating enzymes such as Rho-GTPases.
based motility depend on surface parameters. Nature 417, 308–
Physical SBCs need to be completed by biochemical SBCs 311
comprising surface-grafted, but also soluble and diffusible, 18 Lacayo, C.I. et al. (2012) Choosing orientation: influence of cargo
cues. geometry and ActA polarization on actin comet tails. Mol. Biol. Cell
Notably, understanding of the basic laws governing 23, 614–629
19 Michelot, A. et al. (2007) Actin-filament stochastic dynamics mediated
cytoskeleton assembly can not only provide insights into
by ADF/cofilin. Curr. Biol. 17, 825–833
cell and tissue morphogenesis, but may also have techno- 20 Soares e Silva, M. et al. (2011) Self-organized patterns of actin
logical applications in the development of microdevices filaments in cell-sized confinement. Soft Matter 7, 10631
requiring complex and dynamic architectures. A structure 21 Månsson, A. et al. (2012) Self-organization of motor-propelled
whose precise architecture is regulated by deterministic cytoskeletal filaments at topographically defined borders. J. Biomed.
Biotechnol. 2012, 647265
assembly rules, that can grow and self-repair because it 22 Liu, A.P. et al. (2008) Membrane-induced bundling of actin filaments.
self-renews, has advantages over a fixed structure that Nat. Phys. 4, 789–793
would have to be repaired or replaced by a prefabricated 23 Roos, W.H. et al. (2003) Freely suspended actin cortex models on arrays
static component. This new sort of manufacturing would be of microfabricated pillars. Chemphyschem 4, 872–877
a useful way to prepare novel biomaterials and should find 24 Uhrig, K. et al. (2009) Optical force sensor array in a microfluidic device
based on holographic optical tweezers. Lab Chip 9, 661–668
promising applications in microelectronics and robotics. 25 Reymann, A-C. et al. (2010) Nucleation geometry governs ordered actin
networks structures. Nat. Mater. 9, 827–832
Acknowledgments 26 Pinot, M. et al. (2012) Confinement induces actin flow in a meiotic
We apologize to authors whose work on cytoskeleton self-organization cytoplasm. Proc. Natl. Acad. Sci. U.S.A. 109, 11705–11710
was instructive and influential but not cited here because the purpose 27 Reymann, A-C. et al. (2012) Actin network architecture can determine
was to focus on the specific role of SBCs. We thank all members of the myosin motor activity. Science 336, 1310–1314
Physics of the Cytoskeleton and Morphogenesis Laboratory for their 28 Thoresen, T. et al. (2011) Reconstitution of contractile actomyosin
experimental work and discussions. This work was supported by grants bundles. Biophys. J. 100, 2698–2705
from the Human Frontier Science Programs (RGP0004/2011 to L.B. and 29 Shelanski, M.L. (1973) Chemistry of the filaments and tubules of brain.
RGY0088/2012 to M.T.) and Institut National du Cancer (PLBIO 2011- J. Histochem. Cytochem. 21, 529–539
141 to M.T.). 30 Holy, T.E. et al. (1997) Assembly and positioning of microtubule asters
in microfabricated chambers. Proc. Natl. Acad. Sci. U.S.A. 94, 6228–
6231
References 31 Laan, L. et al. (2012) Cortical dynein controls microtubule dynamics to
1 Karsenti, E. (2008) Self-organization in cell biology: a brief history. Nat. generate pulling forces that position microtubule asters. Cell 148, 502–
Rev. Mol. Cell Biol. 9, 255–262 514
2 Huber, F. and Käs, J. (2011) Self-regulative organization of the 32 Nedelec, F. et al. (1997) Self-organization of microtubules and motors.
cytoskeleton. Cytoskeleton (Hoboken) 68, 259–265 Nature 389, 305–308
3 Surrey, T. et al. (2001) Physical properties determining self- 33 Pinot, M. et al. (2009) Effects of confinement on the self-organization of
organization of motors and microtubules. Science 292, 1167–1171 microtubules and motors. Curr. Biol. 19, 954–960
4 Schaller, V. et al. (2010) Polar patterns of driven filaments. Nature 467, 34 Cosentino Lagomarsino, M. et al. (2007) Microtubule organization in
73–77 three-dimensional confined geometries: evaluating the role of elasticity
5 Sumino, Y. et al. (2012) Large-scale vortex lattice emerging from through a combined in vitro and modeling approach. Biophys. J. 92,
collectively moving microtubules. Nature 483, 448–452 1046–1057
6 Cortès, S. et al. (2006) Microtubule self-organisation by reaction- 35 Emsellem, V. et al. (1998) Vesicle deformation by microtubules: a phase
diffusion processes in miniature cell-sized containers and diagram. Phys. Rev. E 58, 4807–4810
phospholipid vesicles. Biophys. Chem. 120, 168–177 36 Fygenson, D. et al. (1997) Mechanics of microtubule-based membrane
7 Pollard, T.D. and Cooper, J.A. (1986) Actin and actin-binding proteins. extension. Phys. Rev. Lett. 79, 4497–4500
A critical evaluation of mechanisms and functions. Annu. Rev. 37 Karsenti, E. and Vernos, I. (2001) The mitotic spindle: a self-made
Biochem. 55, 987–1035 machine. Science 294, 543–547

680
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

38 Heald, R. et al. (1996) Self-organization of microtubules into bipolar 66 Théry, M. et al. (2006) Cell distribution of stress fibres in response to
spindles around artificial chromosomes in Xenopus egg extracts. the geometry of the adhesive environment. Cell Motil. Cytoskeleton 63,
Nature 382, 420–425 341–355
39 Halpin, D. et al. (2011) Mitotic spindle assembly around RCC1-coated 67 Tseng, Q. et al. (2011) A new micropatterning method of soft substrates
beads in Xenopus egg extracts. PLoS Biol. 9, e1001225 reveals that different tumorigenic signals can promote or reduce cell
40 Gaetz, J. et al. (2006) Examining how the spatial organization of contraction levels. Lab Chip 11, 2231–2240
chromatin signals influences metaphase spindle assembly. Nat. Cell 68 Bischofs, I.B. et al. (2008) Filamentous network mechanics and active
Biol. 8, 924–932 contractility determine cell and tissue shape. Biophys. J. 95, 3488–
41 Dinarina, A. et al. (2009) Chromatin shapes the mitotic spindle. Cell 3496
138, 502–513 69 Théry, M. et al. (2006) Anisotropy of cell adhesive microenvironment
42 Bornens, M. et al. (1989) The cortical microfilament system of governs cell internal organization and orientation of polarity. Proc.
lymphoblasts displays a periodic oscillatory activity in the absence of Natl. Acad. Sci. U.S.A. 103, 19771–19776
microtubules: implications for cell polarity. J. Cell Biol. 109, 1071–1083 70 Wu, J. et al. (2011) Effects of dynein on microtubule mechanics and
43 Bailly, E. et al. (1991) The cortical actomyosin system of cytochalasin D- centrosome positioning. Mol. Biol. Cell 22, 4834–4841
treated lymphoblasts. Exp. Cell Res. 196, 287–293 71 Hale, C.M. et al. (2011) SMRT analysis of MTOC and nuclear
44 Paluch, E. et al. (2005) Cortical actomyosin breakage triggers shape positioning reveals the role of EB1 and LIC1 in single-cell
oscillations in cells and cell fragments. Biophys. J. 89, 724–733 polarization. J. Cell Sci. 124, 4267–4285
45 Cavalcanti-Adam, E.A. et al. (2007) Cell spreading and focal adhesion 72 Zhu, J. et al. (2010) finding the cell center by a balance of dynein and
dynamics are regulated by spacing of integrin ligands. Biophys. J. 92, myosin pulling and microtubule pushing: a computational study. Mol.
2964–2974 Biol. Cell 21, 4418–4427
46 Schvartzman, M. et al. (2011) Nanolithographic control of the spatial 73 Dupin, I. et al. (2009) Classical cadherins control nucleus and
organization of cellular adhesion receptors at the single-molecule level. centrosome position and cell polarity. J. Cell Biol. 185, 779–786
Nano Lett. 11, 1306–1312 74 Camand, E. et al. (2012) N-cadherin expression level modulates
47 Vignaud, T. et al. (2012) Reprogramming cell shape with laser nano- integrin-mediated polarity and strongly impacts on the speed
patterning. J. Cell Sci. 125, 2134–2140 and directionality of glial cell migration. J. Cell Sci. 125, 844–
48 Lehnert, D. et al. (2004) Cell behaviour on micropatterned substrata: 857
limits of extracellular matrix geometry for spreading and adhesion. J. 75 Lombardi, M.L. et al. (2011) The interaction between nesprins and sun
Cell Sci. 117, 41–52 proteins at the nuclear envelope is critical for force transmission
49 Vianay, B. et al. (2010) Single cells spreading on a protein lattice adopt between the nucleus and cytoskeleton. J. Biol. Chem. 286, 26743–
an energy minimizing shape. Phys. Rev. Lett. 105, 3–6 26753
50 Levina, E.M. et al. (2001) Cytoskeletal control of fibroblast length: 76 Pitaval, A. et al. (2010) Cell shape and contractility regulate
experiments with linear strips of substrate. J. Cell Sci. 114, 4335–4341 ciliogenesis in cell cycle-arrested cells. J. Cell Biol. 191, 303–312
51 Picone, R. et al. (2010) A polarised population of dynamic microtubules 77 Mahmud, G. et al. (2009) Directing cell motions on micropatterned
mediates homeostatic length control in animal cells. PLoS Biol. 8, ratchets. Nat. Phys. 5, 606–612
e1000542 78 Kushiro, K. et al. (2012) Modular design of micropattern geometry
52 Vartanian, K.B. et al. (2008) Endothelial cell cytoskeletal alignment achieves combinatorial enhancements in cell motility. Langmuir 28,
independent of fluid shear stress on micropatterned surfaces. Biochem. 4357–4362
Biophys. Res. Commun. 371, 787–792 79 Yoon, S-H. et al. (2011) A biological breadboard platform for cell
53 Terenna, C.R. et al. (2008) Physical mechanisms redirecting cell adhesion and detachment studies. Lab Chip 11, 3555–3562
polarity and cell shape in fission yeast. Curr. Biol. 18, 1748–1753 80 Csucs, G. et al. (2007) Locomotion of fish epidermal keratocytes
54 Bergert, M. et al. (2012) Cell mechanics control rapid transitions on spatially selective adhesion patterns. Cell Motil. Cytoskeleton 64,
between blebs and lamellipodia during migration. Proc. Natl. Acad. 856–867
Sci. U.S.A. 666, 1–7 81 Doyle, A.D. et al. (2009) One-dimensional topography underlies
55 James, J. et al. (2008) Subcellular curvature at the perimeter of three-dimensional fibrillar cell migration. J. Cell Biol. 184, 481–
micropatterned cells influences lamellipodial distribution and cell 490
polarity. Cell Motil. Cytoskeleton 65, 841–852 82 Pouthas, F. et al. (2008) In migrating cells, the Golgi complex and the
56 Parker, K.K. et al. (2002) Directional control of lamellipodia extension position of the centrosome depend on geometrical constraints of the
by constraining cell shape and orienting cell tractional forces. FASEB substratum. J. Cell Sci. 121, 2406–2414
J. 16, 1195–1204 83 Maiuri, P. et al. (2012) The world first cell race. Curr. Biol. 22, R673–
57 Brock, A. et al. (2003) Geometric determinants of directional cell R675
motility revealed using microcontact printing. Langmuir 19, 1611– 84 Théry, M. et al. (2007) Experimental and theoretical study of mitotic
1617 spindle orientation. Nature 447, 493–496
58 Rossier, O.M. et al. (2010) Force generated by actomyosin contraction 85 Fink, J. et al. (2011) External forces control mitotic spindle positioning.
builds bridges between adhesive contacts. EMBO J. 29, 1055–1068 Nat. Cell Biol. 13, 771–778
59 Tan, J.L. et al. (2003) Cells lying on a bed of microneedles: an approach 86 Samora, C.P. et al. (2011) MAP4 and CLASP1 operate as a safety
to isolate mechanical force. Proc. Natl. Acad. Sci. U.S.A. 100, 1484– mechanism to maintain a stable spindle position in mitosis. Nat. Cell
1489 Biol. 13, 1040–1050
60 Rape, A.D. et al. (2011) The regulation of traction force in relation to cell 87 Kiyomitsu, T. and Cheeseman, I.M. (2012) Chromosome- and spindle-
shape and focal adhesions. Biomaterials 32, 2043–2051 pole-derived signals generate an intrinsic code for spindle position and
61 Kilian, K. et al. (2010) Geometric cues for directing the differentiation orientation. Nat. Cell Biol. 14, 311–317
of mesenchymal stem cells. Proc. Natl. Acad. Sci. U.S.A. 107, 4872– 88 Minc, N. et al. (2011) Influence of cell geometry on division-plane
4877 positioning. Cell 144, 414–426
62 Hu, S. et al. (2004) Mechanical anisotropy of adherent cells probed by a 89 Minc, N. and Piel, M. (2012) Predicting division plane position and
3D magnetic twisting device. Am. J. Physiol. Cell Physiol. 287, C1884– orientation. Trends Cell Biol. 22, 193–200
C1191 90 Huang, S. et al. (2005) Symmetry-breaking in mammalian cell cohort
63 Khatau, S.B. et al. (2009) A perinuclear actin cap regulates nuclear migration during tissue pattern formation: role of random-walk
shape. Proc. Natl. Acad. Sci. U.S.A. 106, 19017–19022 persistence. Cell Motil. Cytoskeleton 61, 201–213
64 Bray, M-A. et al. (2008) Sarcomere alignment is regulated by myocyte 91 Burute, M. and Thery, M. (2012) Spatial segregation of cell–cell and
shape. Cell Motil. Cytoskeleton 651, 641–651 cell–matrix adhesions. Curr. Opin. Cell Biol. 24, 628–636
65 Xu, J. et al. (2011) Effects of micropatterned curvature on the motility 92 Tseng, Q. et al. (2012) Spatial organization of the extracellular matrix
and mechanical properties of airway smooth muscle cells. Biochem. regulates cell-cell junction positioning. Proc. Natl. Acad. Sci. U.S.A.
Biophys. Res. Commun. 415, 591–596 109, 1506–1511

681
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

93 McCain, M.L. et al. (2012) Cooperative coupling of cell-matrix and cell- 96 Desai, R.A. et al. (2009) Cell polarity triggered by cell-cell adhesion via
cell adhesions in cardiac muscle. Proc. Natl. Acad. Sci. U.S.A. 109, E-cadherin. J. Cell Sci. 122, 905–911
9881–9886 97 Reffay, M. et al. (2011) Orientation and polarity in collectively
94 Serra-Picamal, X. et al. (2012) Mechanical waves during tissue migrating cell structures: statics and dynamics. Biophys. J. 100,
expansion. Nat. Phys. 8, 628–634 2566–2575
95 Wan, L.Q. et al. (2011) Micropatterned mammalian cells exhibit 98 Mullins, R.D. (2010) Cytoskeletal mechanisms for breaking cellular
phenotype-specific left-right asymmetry. Proc. Natl. Acad. Sci. symmetry. Cold Spring Harb. Perspect. Biol. 2, a003392
U.S.A. 108, 12295–12300

682
Review

Special Issue – Synthetic Cell Biology

Directing the assembly of spatially


organized multicomponent tissues
from the bottom up
Jennifer S. Liu and Zev J. Gartner
Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 95108, USA

The complexity of the human body derives from numer- tissue engineering approaches are ‘top down’ and require
ous modular building blocks assembled hierarchically the use of patterned substrates, molds, or ECM scaffolds to
across multiple length scales. These building blocks, assist cells in finding their appropriate positions and dif-
spanning sizes ranging from single cells to organs, in- ferentiation states within a tissue. In principle, a 3D
teract to regulate development and normal organismal scaffold of ECM of the precise composition and organiza-
function but become disorganized during disease. Here, tion can provide all the necessary structural and microen-
we review methods for the bottom-up and directed vironmental cues to direct the organization of individual
assembly of modular, multicellular, and tissue-like con- cells into a functional tissue or organ, as evidenced by
structs in vitro. These engineered tissues will help refine recent experiments using decellularized organs [2]. How-
our understanding of the relationship between form and ever, de novo construction of scaffolds with the requisite
function in the human body, provide new models for the level of detail at all length scales is not currently possible.
breakdown in tissue architecture that accompanies dis- As a consequence, tissue reconstruction starting from cells
ease, and serve as building blocks for the field of regen- or cell aggregates remains challenging, because mixtures
erative medicine. of dissociated cells do not typically reconstitute complex
tissue structures or functions without pre-organization
Investigating the relationship between tissue form and into the correct 3D geometry. Therefore, additional means
function with in vitro engineered tissues of controlling the spatial organization of cells or groups of
Humans contain trillions of cells spanning over 200 spe- cells will facilitate tissue engineering.
cialized subtypes. This complex cellular community grows Bottom-up or synthetic approaches are emerging as
within a web of extracellular matrix (ECM) to form the valuable and alternative means to more prevalent top-
tissues and organs that perform the numerous functions of down approaches for pre-organizing groups of cells into
our bodies. Cells, tissues, and organs constitute a hierar- tissue-like structures. Bottom-up approaches are distinct
chy of structures spanning tens of microns to meters, in from top-down approaches in that they link together sim-
which the arrangement of building blocks at one scale plified building blocks to generate objects that are struc-
forms the building block for the next. A major goal of cell turally organized at larger length scales [3]. Directing the
and developmental biology is to delineate how the form of assembly of building blocks from the bottom up may pro-
the body – or the composition and physical organization of vide enhanced control over the relative spatial arrange-
its building blocks – affects function at the level of tissues, ment of cells in engineered tissues when used together
organs, or the whole organism [1]. However, this remains a with currently available top-down approaches. In addition
challenging goal because direct and general methods for to the advantages of the top-down tissue engineering
controlling the relative spatial position of cells in tissues strategies outlined above, bottom-up methods have several
and organs do not exist. other desirable features. First, they are inherently modu-
One powerful approach for elucidating fundamental lar, allowing for the simple replacement of specific cells or
principles relating form to function is an engineering nodes in a network of interacting cells, tissues, or organs
strategy that builds tissue-like structures ex vivo. Like (Box 1). This feature makes bottom-up engineering attrac-
studies in model organisms, tissue-engineering strategies tive as a versatile method for incorporating multiple cell
can incorporate genetically modified cellular building types into tissues as well as for building different tissue
blocks. Unlike studies using model organisms, however, types or states (for example, functional or pathological) by
tissue-engineering strategies are also compatible with the interchanging building blocks. Furthermore, these meth-
use of primary or immortalized human cells, advanced ods are inherently scalable; many nearly identical tissue
imaging techniques, and techniques that control the constructs can be prepared without the need for complex or
non-cellular components of the microenvironment. Most specialized scaffolds. Finally, bottom-up approaches are
ideally suited for studying the direct interactions between
Corresponding author: Gartner, Z.J. (zev.gartner@ucsf.edu).
Keywords: bottom up; programmed assembly; tissue engineering;
individual building blocks. Recent research has highlight-
cell–cell interactions; paracrine signaling. ed the importance of interactions between heterogeneous
0962-8924/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2012.09.004 Trends in Cell Biology, December 2012, Vol. 22, No. 12 683
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

Box 1. The hierarchical organization of a modular organ: the breast


The human breast contains an organized hierarchy of structures built For this modular and hierarchically organized tissue to function,
up from modular units, from nanometer-sized proteins of the mechanical, chemical, and electrical signals must be detected by
basement membrane to micron-sized cells to millimeter-sized tissues individual cells and transmitted across each hierarchy of the organ to
[80]. The bilayered epithelium of the mammary gland, for example, synchronize cellular behaviors. Single epithelial cells integrate
has two principle building blocks: luminal epithelial and myoepithe- chemical and mechanical cues from the basement membrane, a
lial cells. Considerable heterogeneity exists even within each of these specialized ECM, to maintain cell polarity and secrete milk [83].
cell types. For example, subpopulations of luminal cells express Epithelial cells are also mechanically coupled with their neighbors
estrogen and progesterone receptors. When stimulated, these cells and distant tissues via the cytoskeleton and the ECM, respectively
release growth factors triggering the growth of their neighbors. In [84,85]. The cytosols of epithelial cells are chemically and electrically
addition, luminal and myoepithelial cells play distinct functional linked through gap junctions [86], and an array of secreted and
roles, serving to secrete and pump milk, respectively (Figure Ia). membrane-localized signaling proteins coordinate tissue homeosta-
These cellular building blocks are organized into ducts and acini that sis and function across the epithelium. Additionally, secreted
are further supported by fibroblasts that synthesize and reside in a endocrine factors link the mammary gland with stromal adipocytes,
collagenous ECM. Endothelial cells provide additional support for the nervous system, and other reproductive organs [87]. Processes
these structures through a meshwork of capillaries delivering such as breastfeeding require not only coordination between multiple
nutrients, facilitating circulation of lymphocytes, and relaying hor- cell types and modules within the breast but also between sensory
mones from distant organs (Figure Ib). Ducts and acini are further cells in the nervous system, which further synchronize actions in
organized into terminal ductal lobular units (TDLUs) that are distant organs to those in the breast. Although it is appreciated that
surrounded by a secondary and specialized ECM containing a denser overlapping signals across this hierarchy of building blocks are
collagenous matrix and beds of adipocytes that add additional form required for the higher order function of the breast and all other
to the organ (Figure Ic) [80]. Finally, TDLUs are organized into tissues, the structural organization of building blocks that serves as
multiple lobes that drain into large ducts, together delivering milk to the foundation for the proper exchange of intercellular signaling
the nipple [82] (Figure Id). Although the overall architecture of the events remains challenging to study. By spatially pre-organizing cells,
gland is drastically remodeled over the course of a woman’s lifetime, tissues and organs, bottom-up and directed tissue engineering
the relative position of the different cell types with respect to each strategies aim to control and understand the exchange of signals
other and the modular organization of the organ remain constant in between building blocks at all levels of structural hierarchy in the
healthy tissue. human body.

Endocrine signals Stroma


(a) (b) (c) (d) Nervous
system

Growth TDLU
factors Mammary
ER+ gland
ER- Epithelium
ER-
Nipple
Fibroblast

10–5 10–4 10–3 10–1m


TRENDS in Cell Biology

Figure I. The modular and hierarchical organization of the human mammary gland. (a) Individual glandular epithelial cells exchange signals with each other and the
basement membrane. (b) Epithelial cells of the ducts and acini also exchange signals with the surrounding lobular stroma. (c) Ducts and acini are organized into
terminal ductal lobular units (TDLUs) that are embedded in a second type of collagenous extracellular matrix (ECM) that also contains many adipocytes. (d) The entire
organ is integrated with the rest of the body to mediate its function in delivering milk during breastfeeding. Adapted from [1].

cell types on tissue behaviors, whether the interactions or progenitor sources, or synthetically engineered genetic
occur within an epithelium [4–7], between the epithelium circuits. The reader is directed to several recent reviews for
and surrounding stroma [8], or even between cells in discussion of these topics [10–15].
different organ systems [9]. Although spatially organizing
multiple heterogeneous cellular interactions can be chal- Directing the bottom-up assembly of tissues using
lenging using top-down tissue engineering approaches, a single cells as building blocks
multiplicity of interacting partners can be systematically Control over the relative position of single cells provides
incorporated using a modular, bottom-up approach. the fine resolution necessary for probing interactions be-
This review focuses on bottom-up and directed-assem- tween neighboring cells in a tissue. This level of spatial
bly approaches that utilize predefined building blocks to resolution is required for recreating stem cell niches [16] or
construct spatially defined multicellular structures con- when rebuilding cell–cell connections found in fully differ-
taining more than one cell type. The goal of these methods entiated tissues [17]. In some cases, a group of cells has the
is to mimic the cellular heterogeneity and physical ar- ability to self-assemble into specific structures at these
rangement of the modular repeating units found in mam- length scales. Townes and Holtfreter famously found that
malian tissues (Figure 1) by directing their assembly from dissociated cells from amphibian embryos would aggregate
simpler building blocks. This review will not focus on other and self-sort into germ layers without outside intervention
techniques that control the spatial organization of tissues [18]. This strategy occasionally allows a multiplicity of cell
through organ printing, microscale technologies, genetic or types to self-organize in wells or in hanging drops [19–21].
optogenetic techniques, directed differentiation of stem cell However, isolated mixtures of cells of multiple types do not
684
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) (b) (c) (d)

TRENDS in Cell Biology

Figure 1. Modular functional units in mammalian organs. (a) Human skeletal muscle cross section. (b) Human mammary terminal ductal lobular unit (TDLU) cross-section.
Arrow indicates extralobular stroma; arrowhead indicates lobular stroma. (c) Pig liver cross-section showing repeating lobules. (d) Mouse embryonic kidney. Reproduced
with permission from [80], Pathpedia, Werning, S. (2007) (http://calphotos.berkeley.edu/cgi/img_query?seq_num=223971&one=T), and [81].

always spontaneously organize into structures that mimic microns. Neither phenotype was observed in aggregates
their tissue of origin without the aid of external ECM homogeneous for Ras-activated cells nor in aggregates as-
scaffolds. In the absence of these external positioning cues, sembled from single WT MCF10A cells with Ras-activated
bottom-up and directed-assembly techniques may be used neighbors (Figure 2b,c). Because this method controls the
to spatially position cells in relation to each other at the initial architecture of aggregates, the emergent phenotypes
microscale. could be directly attributed to the underlying cell-to-cell
DNA-programmed assembly is a recently developed ap- variability in Ras activity.
proach to direct the organization of multicellular structures DNA-programmed assembly of cellular building blocks
in vitro with single-cell resolution [22]. Key to this approach enables the study of cell–cell interactions in the context of
is the covalent or non-covalent remodeling of the adhesive multicellular structures with precise spatial arrange-
properties of the cell surface with single-stranded DNA ments. Because essentially any cell type can be modified
(ssDNA). ssDNA is linked to the cell surface by several with ssDNA using the various DNA-labeling methods out-
means. In one approach, cells are first cultured in the lined above and a nearly unlimited set of orthogonal DNA
presence of an azide-modified monosaccharide that is incor- sequences is available, DNA-programmed assembly can be
porated into cell surface glycans. The accessible azides react used to reconstitute various complex heterotypic cell–cell
with chemically modified oligonucleotides by Staudinger interactions for study. Importantly, unlike in genetic engi-
ligation [23] or [1,3]-dipolar cycloaddition [22] to covalently neering, the DNA used to program cellular interactions is
attach the DNA to the glycocalyx. In an alternative ap- temporary and degrades rapidly at 37 8C to leave unmodi-
proach, N-hydroxysuccinimide-modified DNA is added to fied interacting cells [24,25]. This technique closely
cell suspensions, where it reacts covalently with free lysines apposes interacting cell surfaces and is ideally suited for
on the cell surface [24]. Lastly, lipid–DNA conjugates are the study of multicellular circuits that operate over short
added to culture medium where they passively partition into distances. Interactions amenable to study with this ap-
the cell membrane for non-covalent cell surface modification proach include those mediated by electrical and chemical
[25–27]. Labeling or binding other interacting biomolecules signals through gap junctions [33], short-range mechanical
to cell surfaces will also direct the programmed assembly of signals coupled to the cytoskeleton [34], juxtacrine signal-
cells, though these are often limited to only single pairs of ing such as through the Notch pathway [35], and short-
interacting molecules [28–31]. range paracrine signaling such as through the Wnt and
Mixing different cell populations labeled with comple- Hedgehog pathways [36,37]. However, because products of
mentary ssDNA strands or molecules directs the formation microscale cell–cell assembly have been limited to around
of heterogeneous microtissues, whereas changing the ratios 100 microns in diameter, cell–cell interactions that occur
of labeled populations can be used to achieve discrete mul- between neighboring or distant tissues or tissue compart-
ticellular arrangements (Figure 2a). This strategy has been ments have evaded study using this strategy alone. Access
used to recapitulate synthetic paracrine signaling networks to these larger structures might be achieved by combining
[22] and to mimic immune cell homing to sites of inflamma- programmed assembly with top-down techniques or by
tion [29]. More recently, a DNA-mediated programmed using the products of programmed assembly themselves
assembly approach was used to investigate the conse- as building blocks for further elaboration.
quences of cell-to-cell variability among mammary epitheli-
al cells during the dynamic process of morphogenesis [32]. Directing the bottom-up assembly of tissues using cell
Wild-type (WT) MCF10A mammary epithelial cells and sheets and aggregates as building blocks
derivatives with elevated Ras activation were labeled with Many tissues and organs contain repetitive subunits com-
ssDNA and combinatorially assembled to form homoge- prising groups of cells with dimensions of hundreds of
neous and mosaic microtissues of defined compositions. microns to a millimeter. Such structures include pancre-
Mosaic epithelial aggregates assembled from single Ras- atic islets, lymph nodes, and the lobules of the breast and
activated cells and WT MCF10A neighbors displayed emer- liver. Most cells within these repeating units are fully
gent behaviors: Ras-activated cells were basally extruded or differentiated and structurally integrated with their neigh-
led motile multicellular protrusion that directed the motility bors and the ECM. Therefore, approximating these units
of the surrounding WT cells across tens to hundreds of with cell aggregates that contain fully formed cell–cell
685
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

(a) Assemble Purify Culture


Label cell lines aggregates aggregates microssues

DNA ‘a’
‘a’
Gene

Gen DNA ‘b’


e ‘b’

0.5 h 6h 12 h 18 h 24 h
(b)

Normal H2B-GFP

Extrusion

Protrusion

(c)
Normal Extrusion Protrusion

WT WT ns ns
Ras Ras
***
***

WT Ras
0
0

10

20

30

40
20

40

60

80

10

20

30

40
10

% Frequency
TRENDS in Cell Biology

Figure 2. DNA-programmed assembly. (a) Fluorescent epithelial cells labeled with complementary single-stranded DNA (ssDNA) (or other interacting molecules) are
brought together through molecular recognition. Mixing cell populations 1:50 results in discrete multicellular aggregates that can be purified using fluorescence activated
cell sorting. Assembled aggregates are then cultured in laminin-rich extracellular matrix (ECM) to form polarized microtissues. Genetically distinct input cells can be
incorporated to build mosaic aggregates. (b) Mosaic microtissues assembled from single histone H2B-green fluorescent protein (GFP)-expressing MCF10AT cells, which
express low levels of H-RasV12, and wild type MCF10A neighbors display emergent behaviors that are not observed in homogeneous assemblies (scale bar, 10 mm). (c)
Quantification of the emergent behaviors in homogeneous and mosaic aggregates. Mean values of greater than 500 observations are displayed, with error bars
representing the standard deviation of the mean. WT, wild type; Ras, Ras-activated MCF10neoT. Reproduced with permission from [32].

junctions may provide a means of constructing tissues at various shapes and sizes [38–41]. Such an approach has
larger length scales than can be achieved from assembly of been used to combine aggregates of human fibroblasts and
single cells alone. rat hepatoma cells. When precultured together in spher-
Spherical cell aggregates of the appropriate size have oids, these two cell types self-sort into an inner core of
been engineered using top-down molding techniques and fibroblasts surrounded by hepatoma cells. When directed
then assembled within microwells into larger structures of to assemble in rectangular molds, these precultured
686
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

CD31 Merged
(a) (d) No. 2 EC - Fb - Fb (e) No. 2-1-3Dy No. 2-2-3Dy

(b) 20 µm 20 µm

No. 3-1-3Dy No. 3-2-3Dy


No. 3 Fb - EC - Fb

20 µm 20 µm

No. 4-1-3Dy No. 4-2-3Dy


No. 4
Fb - Fb - EC

(c)
20 µm 20 µm

No. 5 Co - Co - Co No. 5-1-3Dy No. 5-2-3Dy

20 µm 20 µm

TRENDS in Cell Biology

Figure 3. Directed assembly of cell aggregates. (a) Preformed, spheroidal cell aggregates (100 mm in diameter) assembled in trough-shaped wells fused into rod-shaped
structures over 24 hours. (b) Precultured heterogeneous spheroids with a core of human fibroblasts (red) and surrounding rat hepatoma (green) cells cultured in wells for
24 hours. (c) Confocal image shows that the assembled spheroids retained the inner fibroblast core during spheroid fusion (scale bars, 200 mm). (d) Different combinations
of three cell sheet layers are stacked to make heterogeneous tissues of endothelial (green) and fibroblast (pink) cells. (e) After 3 days, endothelial sheets formed vessels
(outlined with indirect CD31 staining in green) when stacked within or under fibroblast sheets (actin staining in red in merged images; scale bars, 20 mm). Reproduced with
permission from [40] and [51].

spheroidal building blocks fuse into a rod but maintain the prepared by manually stacking different cell sheets. For
fibroblast cores for at least 24 hours [40] (Figure 3a–c). instance, myoblast cell sheets intercalated with either
Directed assembly with globular cell aggregates (Table 1) layers, lines, or dissociated human umbilical vein endothe-
has been demonstrated only with modules of a single type, lial cells (HUVECs) [50–52] (Figure 3d,e) formed vascular
though the same basic strategy should be applicable to networks in vitro that could integrate with host vasculature
modules made of different cell types. However, the overall in vivo when grafted subcutaneously into rats [52].
architecture in large assemblies of cell aggregates may By starting with multicellular building blocks with an
require further structural support from the microenviron- established geometry, directed assembly of cell aggregates
ment to be stable over the long term [39]. of different cell types can be used to design and spatially
Cell sheets have also been used as building blocks and can position tissue-sized networks of interacting cells. These
be stacked manually to build macroscopic tissue structures. higher-order structures are amenable to further elabora-
Sheets are cultured in suspension or released using ther- tion with layers of individual cells. For example, pancreatic
mal- or ion-sensitive coatings [42–44]. Single sheets have islets isolated from rodents were labeled with lipid-modi-
been made from various cell types, including fibroblasts, fied DNA and surrounded with mammalian cells bearing
endothelial cells, and epithelial cells [45,46]. Cell–cell adhe- complementary DNA to provide a barrier function to the
sions and secreted ECM components are maintained in sensitive cell aggregates [53]. In principle, such a strategy
lifted sheets [47,48], which can also promote tissue-like could also introduce components of the surrounding tissue
functions that are not observed with dissociated cells [49]. stroma to provide additional trophic support between
3D tissue constructs comprising various cell types can be modular units. The modular organization of assembled

Table 1. Features, advantages, and opportunities for directed assembly approaches


Building block Major advantage Size of products (relevant Current applications Challenges and opportunities
biological structures)
Cell Single cell spatial resolution 10–100 mm (groups of Study of cell–cell interactions Assembling more complex tissues;
cells, niches) across short length scales; Incorporation of products into
Study of heterotypic cell–cell multiscale tissues;
interactions; Control of product architecture
Drug screening over time
Cell aggregate Mature cell–cell junctions 100 mm to 1 mm Study of cell aggregation; Control of product architecture
or sheet (functional tissue units) Study of interactions between over time;
cell populations; Nutrient delivery and/or
Production of engineered and vascularization
functional tissues
Cell-laden Incorporation of specialized 100 mm to 1 mm Study of paracrine signaling; Matching matrix properties to
hydrogel ECM or ECM-like material; (functional tissue units Generation of tissue engineering in vivo tissue;
Easily interfaced with or multicomponent constructs Nutrient delivery and/or
microscale engineering tissues) vascularization;
approaches Responsive or smart hydrogels

687
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

building blocks may also be stratified with exogenous containing rat myocardiocytes onto a planar nylon filter
ECM-like components to mimic tissue architectures of then added temporary alginate glue. The resulting modular
increasing complexity. tissue formed a sheet-like structure that contracted on
electrical stimulation [57]. Collagen hydrogels containing
Directing the bottom-up assembly of tissues using cell- different cell types have also been assembled into a linear
laden hydrogels as building blocks array using microfluidics to constrain hydrogel orientation
The minimal functional unit of many tissues comprises [59].
small groups of structurally organized cells embedded in In contrast to the random but constrained assembly of
specialized ECM. Collections of these functional units are subunits, directed-assembly schemes can guide the forma-
further organized in 3D space within additional ECM that is tion of a multiplicity of hydrogel building blocks into more
frequently of a different composition. The terminal ductal defined structures. Assembling submillimeter-sized poly-
lobular units (TDLUs) and the intervening adipocyte-rich ethylene glycol (PEG)-based hydrogels in hydrophobic me-
regions of the mammary gland exemplify this organization dia, for example, promotes the self-association of the
(Box 1). These structures should be modeled with explicit hydrophilic objects. Hydrogel building blocks containing
inclusion of spatially segregated ECM-like materials in two different cell types can be prepared by sequential
building blocks, particularly when matrix components are photolithography steps that meld two concentric rings of
critical to cell and tissue function or when the cells cannot cell-laden PEG hydrogel together into a single unit
generate the volume and composition of the ECM found in (Figure 4a). Additional assembly of these two-component
biological tissues on their own. Fortunately, many natural building blocks into linear arrays generated tubes contain-
and synthetic polymers are available for designing ECM- ing an inner layer of endothelial cells surrounded by
like hydrogels with specific structural and mechanical prop- smooth muscle cells that mimicked the architecture of
erties to mimic different tissues within the human body vasculature [60] (Figure 4b). Another way to generate
[54,55]. Although ECM-like materials can be incorporated spatially defined heterogeneous tissues using this direct-
into tissues by mixing microspheres of hydrogel with indi- ed-assembly method is to mix two different populations of
vidual cells [56], cell-laden hydrogel modules with dimen- hydrogels containing different cell types; changing the
sions of hundreds of microns to millimeters are closer in size ratio of these two hydrogel monomers can bias the compo-
to the ECM-encapsulated repeating units found in many sition of the end products [61]. Nanofibers added to hydro-
tissues in vivo. Therefore, these may be a more tractable gel monomers can provide additional mechanical coupling
building block for constructing interactions between repeat- between modules, facilitating cell differentiation and tis-
ing units within tissues or organs. sue function [62]. An additional level of control over the
The straightforward packing of modular cell-laden interface between two populations of hydrogel-embedded
hydrogel units into a defined space can direct their assembly cells can be added by using gels with lock-and-key designs
into larger structures. Such an approach was used with (Figure 4c,d) to access precise architectures with controlled
building blocks of cell-laden and collagen-based cylinders stoichiometries of building blocks [61] (Figure 4e,f).
covered with an endothelial cell layer [57,58]. The Similar to cellular building blocks, hydrogels can also be
authors assembled multiple endothelium-coated cylinders labeled with ssDNA or other biomolecules to program their

(a) (b) (c) (d)

(e) (f) (g)

100 µm

TRENDS in Cell Biology

Figure 4. Directed assembly of cell-laden hydrogels. (a) Donut-shaped hydrogels with an inner hydrogel ring loaded with endothelial cells (green) surrounded by an outer
ring loaded with smooth muscle cells (red) were made using sequential photolithography steps. (b) Side view of tubular structure formed after sequential assembly of
hydrogel units from a (scale bars, 100 mm). (c,d) Lock-and-key (cross- and rod-shaped) hydrogels stained with fluorescent dextran were made using photolithography. (e,f)
Lock-and-key hydrogels loaded with fluorescent murine fibroblast cells assembled through self-association in a hydrophobic medium (scale bars, 200 mm). (g) 3D volume
reconstruction of DNA-directed hydrogel assemblies. Spherical hydrogels bearing green fluorescent tracking beads and labeled with single-stranded DNA (ssDNA) were
bound to a microarray template. A second layer of hydrogels loaded with red beads and labeled with complementary DNA was assembled onto the first population (scale
bar, 100 mm). Reproduced with permission from [60,61,63].

688
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

assembly. Such an approach avoids the necessity to fabri- components and soluble factors in tissues can be extreme-
cate hydrogel units of complex shape but requires addi- ly dynamic [75,76] and may differ substantially from
tional chemical labeling steps either before or after formulations typically used in culture. Because the chem-
fabrication. A two-step approach was used to label uniform, ical, mechanical, and structural organization of ECM can
spherical PEG-based hydrogel units with streptavidin, affect tissue and cell behaviors [77,78], use of smart
followed by modification with biotinylated ssDNA [63]. materials with adjustable and controllable properties,
Additional control of building block positioning was dem- such as encapsulating hydrogels, could allow for dynamic
onstrated by annealing the ssDNA-coated hydrogels onto control of the ECM and aid the proper morphogenesis of
microarray templates or by directed assembly of two popu- the engineered construct [79]. Additionally, secreted
lations of hydrogels bearing complementary ssDNA on molecules can be integrated into the design of tissue
their surfaces (Figure 4g). Recent reports suggest that modules for gradual release [22,68,69] or the necessary
additional control over interfacial interactions between secreted factors could be delivered and controlled exter-
hydrogel building blocks using non-biological molecules nally such as through microfluidics or photo-uncaging.
could add further complexity to tissues engineered at this
length scale [54,64]. Concluding remarks
Due to advances in top-down fabrication techniques, The applications of bottom-up strategies for directing the
hydrogel building blocks of various sizes, shapes, and com- assembly of specific tissue architectures are still in their
positions are readily designed to mimic architectures and infancy, but they have potential to address important
interfaces observed in tissues in vivo. The density of cells questions relating cellular organization to the coordination
within the hydrogel units can also be controlled, though of multicellular behaviors. At the spatial resolution of
achieving tissue-like cell densities may require extended single cells, directed assembly strategies will aid the study
periods of culture [59]. Because cells are physically con- of stem cell niches from multiple tissue types, with respect
strained within the building blocks, cell-laden hydrogels to both their structure and composition. These methods
may be especially appropriate for studying the exchange will also benefit the study of cell-to-cell variability in gene
of soluble factors between different cell populations [56]. expression or pathway activation in normal and diseased
Moreover, hydrogel compositions can be adjusted using tissues. Finally, these methods will provide a means of
biologically derived or synthetic polymers [65,66] designed studying the coordination of cellular behaviors during
to match the physical and chemical properties of ECM of processes such as morphogenesis and tissue repair. At
specific tissues, or to probe the consequence of changing the larger spatial resolution of cell aggregates and whole
ECM properties on cellular behaviors. Such control would be tissues, directed-assembly strategies will impact the field
especially useful for modeling tissues that contain multiple of regenerative medicine as well as the study of mechanical
compartments with different matrices and cell types, such and chemical coupling of cell groups, particularly between
as between neighboring TDLUs in the breast. Finally, cell- epithelial cells and the components of the stroma.
laden hydrogels of various levels of complexity can be Numerous opportunities exist for improving the direct-
implanted in vivo. In one example, gel-encapsulated cells ed assembly of tissues (Table 1). One avenue of interest is
coated with an endothelial layer show enhanced survival in the combination of the various approaches described
and differentiation, as well as a favorable host response to above to direct the hierarchical assembly of tissues with
growth factors secreted by the encapsulated cells [67–69]. spatial precision across multiple length scales, spanning
that of single cells to full organs. Some progress along these
Considerations lines has been made [53] or would be a logical extension of
The techniques outlined here are designed to direct the existing work [63]. Another opportunity will be in directing
assembly of cells into a specific position relative to other the assembly of anisotropic or asymmetric tissue struc-
cells in the context of a multicellular tissue. To form a tures. Finally, introducing genetic circuits into modules to
functional tissue, however, the cells incorporated into control interactions between neighboring cells or tissues
building blocks must also retain the capacity to interact will provide additional mechanisms for engineering the
properly with their neighbors and to deposit and remodel processes of development and morphogenesis. We antici-
their own ECM. Unfortunately, established cell lines prop- pate that progress towards these challenges, better inte-
agated in 2D culture are frequently used as building blocks gration with top-down engineering techniques, and new
in tissue engineering applications. In these cell lines, applications will be forthcoming in this exciting area.
tissue-specific gene expression patterns and cell surface
proteins necessary for directing heterotypic cell–cell inter- Acknowledgments
actions are often downregulated or lost [70,71]. In fact, The authors would like to acknowledge support for a portion of the work
covered in this review by DOD grant W81XWH-10-1-1023 to Z.J.G., by
even primary cells can begin to lose markers of differenti-
grant P50 GM081879 to the UCSF Center for Systems and Synthetic
ation after brief growth on tissue-culture plastic [72,73]. Biology, and by NSF grant DGE-0648991 to J.S.L.
Therefore, care must be taken to validate that the cell
types used in an engineered tissue retain expression of
appropriate markers of differentiation and the ability to References
1 Nelson, C.M. and Bissell, M.J. (2005) Modeling dynamic reciprocity:
interact with neighboring cell populations. engineering three-dimensional culture models of breast architecture,
Optimization of other components in the microenviron- function, and neoplastic transformation. Sem. Cancer Biol. 15, 342–352
ment may also be required for assembled cells to transition 2 Ott, H.C. et al. (2008) Perfusion-decellularized matrix: using nature’s
to tissue-like organization [74]. For instance, ECM platform to engineer a bioartificial heart. Nat. Med. 14, 213–221

689
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

3 Elbert, D.L. (2011) Bottom-up tissue engineering. Curr. Opin. 31 Hamon, M. et al. (2011) Avidin-biotin-based approach to forming
Biotechnol. 22, 674–680 heterotypic cell clusters and cell sheets on a gas-permeable
4 Hogan, C. et al. (2009) Characterization of the interface between membrane. Biofabrication 3, 034111
normal and transformed epithelial cells. Nat. Cell Biol. 11, 460–467 32 Liu, J.S. et al. (2012) Programmed assembly of mosaic cell aggregates
5 Leung, C.T. and Brugge, J.S. (2012) Outgrowth of single oncogene- reveals the consequences of cell-to-cell variability in Ras activity on the
expressing cells from suppressive epithelial environments. Nature 482, collective behavior of mammary epithelial cells. Cell Rep. http://
410–413 dx.doi.org/10.1016/j.celrep.2012.08.037
6 Eisenhoffer, G.T. et al. (2012) Crowding induces live cell extrusion to 33 Locke, D. (1998) Gap junctions in normal and neoplastic mammary
maintain homeostatic cell numbers in epithelia. Nature 484, 546–549 gland. J. Pathol. 186, 343–349
7 Johnston, L.A. (2009) Competitive interactions between cells: death, 34 Eyckmans, J. et al. (2011) A hitchhiker’s guide to mechanobiology. Dev.
growth, and geography. Science 324, 1679–1682 Cell 21, 35–47
8 Engelhardt, J.J. et al. (2012) Marginating dendritic cells of the tumor 35 Sprinzak, D. et al. (2010) Cis-interactions between Notch and Delta
microenvironment cross-present tumor antigens and stably engage generate mutually exclusive signalling states. Nature 465, 86–90
tumor-specific T cells. Cancer Cell 21, 402–417 36 Taipale, J. and Beachy, P.A. (2001) The Hedgehog and Wnt signalling
9 DeNardo, D.G. et al. (2009) CD4(+) T cells regulate pulmonary pathways in cancer. Nature 411, 349–354
metastasis of mammary carcinomas by enhancing protumor 37 Takebe, N. et al. (2011) Targeting cancer stem cells by inhibiting Wnt,
properties of macrophages. Cancer Cell 16, 91–102 Notch, and Hedgehog pathways. Nat. Rev. Clin. Oncol. 8, 97–106
10 Mironov, V. et al. (2009) Organ printing: tissue spheroids as building 38 Tejavibulya, N. et al. (2011) Directed self-assembly of large scaffold-
blocks. Biomaterials 30, 2164–2174 free multi-cellular honeycomb structures. Biofabrication 3, 034110
11 Kaji, H. et al. (2011) Engineering systems for the generation of 39 Livoti, C.M. and Morgan, J.R. (2010) Self-assembly and tissue fusion of
patterned co-cultures for controlling cell-cell interactions. Biochim. toroid-shaped minimal building units. Tissue Eng. Part A 16, 2051–
Biophys. Acta 1810, 239–250 2061
12 Toettcher, J.E. et al. (2011) The promise of optogenetics in cell biology: 40 Rago, A.P. et al. (2009) Controlling cell position in complex heterotypic
interrogating molecular circuits in space and time. Nat. Methods 8, 35– 3D microtissues by tissue fusion. Biotechnol. Bioeng. 102, 1231–1241
38 41 Rago, A.P. et al. (2009) Encapsulated arrays of self-assembled
13 Miesenbock, G. (2011) Optogenetic control of cells and circuits. Annu. microtissues: an alternative to spherical microcapsules. Tissue Eng.
Rev. Cell Dev. Biol. 27, 731–758 Part A 15, 387–395
14 Wu, S.M. and Hochedlinger, K. (2011) Harnessing the potential of 42 Jean, J. et al. (2011) Bioengineered skin: the self-assembly approach. J.
induced pluripotent stem cells for regenerative medicine. Nat. Cell Tissue Sci. Eng. S5, 001
Biol. 13, 497–505 43 Haraguchi, Y. et al. (2012) Fabrication of functional three-dimensional
15 Lenas, P. et al. (2009) Developmental engineering: a new paradigm for tissues by stacking cell sheets in vitro. Nat. Protoc. 7, 850–858
the design and manufacturing of cell-based products. Part I: from 44 Zahn, R. et al. (2012) Ion-induced cell sheet detachment from standard
three-dimensional cell growth to biomimetics of in vivo cell culture surfaces coated with polyelectrolytes. Biomaterials 33,
development. Tissue Eng. Part B: Rev. 15, 381–394 3421–3427
16 Losick, V.P. et al. (2011) Drosophila stem cell niches: a decade of 45 Yang, J. et al. (2007) Reconstruction of functional tissues with cell sheet
discovery suggests a unified view of stem cell regulation. Dev. Cell engineering. Biomaterials 28, 5033–5043
21, 159–171 46 Labbe, B. et al. (2011) Cell sheet technology for tissue engineering: the
17 Desai, R.A. et al. (2009) Cell polarity triggered by cell-cell adhesion via self-assembly approach using adipose-derived stromal cells. Methods
E-cadherin. J. Cell Sci. 122, 905–911 Mol. Biol. 702, 429–441
18 Townes, P.L. and Holtfreter, J. (1955) Directed movement and selective 47 Nishida, K. et al. (2004) Functional bioengineered corneal epithelial
adhesion of embryonic amphibian cells. J. Exp. Zool. 128, 53–120 sheet grafts from corneal stem cells expanded ex vivo on a temperature-
19 Kunz-Schughart, L.A. et al. (2006) Potential of fibroblasts to regulate the responsive cell culture surface. Transplantation 77, 379–385
formation of three-dimensional vessel-like structures from endothelial 48 Ohashi, K. et al. (2007) Engineering functional two- and three-
cells in vitro. Am. J. Physiol. Cell Physiol. 290, C1385–C1398 dimensional liver systems in vivo using hepatic tissue sheets. Nat.
20 Wenger, A. et al. (2005) Development and characterization of a Med. 13, 880–885
spheroidal coculture model of endothelial cells and fibroblasts for 49 Sekine, H. et al. (2011) Cardiac cell sheet transplantation improves
improving angiogenesis in tissue engineering. Cells Tissues Organs damaged heart function via superior cell survival in comparison with
181, 80–88 dissociated cell injection. Tissue Eng. Part A 17, 2973–2980
21 Foty, R. (2011) A simple hanging drop cell culture protocol for 50 Tsuda, Y. et al. (2007) Cellular control of tissue architectures using a
generation of 3D spheroids. J. Vis. Exp. 51, 2720 three-dimensional tissue fabrication technique. Biomaterials 28, 4939–
22 Gartner, Z.J. and Bertozzi, C.R. (2009) Programmed assembly of 3- 4946
dimensional microtissues with defined cellular connectivity. Proc. 51 Asakawa, N. et al. (2010) Pre-vascularization of in vitro three-
Natl. Acad. Sci. U.S.A. 106, 4606–4610 dimensional tissues created by cell sheet engineering. Biomaterials
23 Chandra, R.A. et al. (2006) Programmable cell adhesion encoded by 31, 3903–3909
DNA hybridization. Angew. Chem. Int. Ed. Engl. 45, 896–901 52 Sasagawa, T. et al. (2010) Design of prevascularized three-dimensional
24 Hsiao, S.C. et al. (2009) Direct cell surface modification with DNA for cell-dense tissues using a cell sheet stacking manipulation technology.
the capture of primary cells and the investigation of myotube formation Biomaterials 31, 1646–1654
on defined patterns. Langmuir 25, 6985–6991 53 Teramura, Y. et al. (2010) Microencapsulation of islets with living cells
25 Selden, N.S. et al. (2012) Chemically programmed cell adhesion with using polyDNA-PEG-lipid conjugate. Bioconjug. Chem. 21, 792–796
membrane-anchored oligonucleotides. J. Am. Chem. Soc. 134, 765–768 54 Seliktar, D. (2012) Designing cell-compatible hydrogels for biomedical
26 Teramura, Y. et al. (2010) Control of cell attachment through polyDNA applications. Science 336, 1124–1128
hybridization. Biomaterials 31, 2229–2235 55 Correia, A.L. and Bissell, M.J. (2012) The tumor microenvironment is a
27 Liu, H. et al. (2011) Membrane anchored immunostimulatory dominant force in multidrug resistance. Drug Resist. Updat. 15, 39–49
oligonucleotides for in vivo cell modification and localized 56 Scott, E.A. et al. (2010) Modular scaffolds assembled around living cells
immunotherapy. Angew. Chem. Int. Ed. Engl. 50, 7052–7055 using poly(ethylene glycol) microspheres with macroporation via a non-
28 Liu, X. et al. (2011) Targeted cell-cell interactions by DNA nanoscaffold- cytotoxic porogen. Acta Biomater. 6, 29–38
templated multivalent bispecific aptamers. Small 7, 1673–1682 57 Leung, B.M. and Sefton, M.V. (2010) A modular approach to cardiac
29 Zhao, W. et al. (2011) Mimicking the inflammatory cell adhesion tissue engineering. Tissue Eng. Part A 16, 3207–3218
cascade by nucleic acid aptamer programmed cell-cell interactions. 58 McGuigan, A.P. and Sefton, M.V. (2006) Vascularized organoid
FASEB J. 25, 3045–3056 engineered by modular assembly enables blood perfusion. Proc.
30 Dutta, D. et al. (2011) Synthetic chemoselective rewiring of cell Natl. Acad. Sci. U.S.A. 103, 11461–11466
surfaces: generation of three-dimensional tissue structures. J. Am. 59 Bruzewicz, D.A. et al. (2008) Fabrication of a modular tissue construct
Chem. Soc. 133, 8704–8713 in a microfluidic chip. Lab Chip 8, 663–671

690
Review Trends in Cell Biology December 2012, Vol. 22, No. 12

60 Du, Y. et al. (2011) Sequential assembly of cell-laden hydrogel endothelial cells outside the lymphoid tissue microenvironment.
constructs to engineer vascular-like microchannels. Biotechnol. Blood 103, 4164–4172
Bioeng. 108, 1693–1703 74 Rivron, N.C. et al. (2009) Tissue assembly and organization:
61 Du, Y. et al. (2008) Directed assembly of cell-laden microgels for developmental mechanisms in microfabricated tissues. Biomaterials
fabrication of 3D tissue constructs. Proc. Natl. Acad. Sci. U.S.A. 30, 4851–4858
105, 9522–9527 75 Maller, O. et al. (2010) Extracellular matrix composition reveals
62 Dvir, T. et al. (2011) Nanowired three-dimensional cardiac patches. complex and dynamic stromal-epithelial interactions in the
Nat. Nanotechnol. 6, 720–725 mammary gland. J. Mammary Gland Biol. Neoplasia 15, 301–318
63 Li, C.Y. et al. (2011) DNA-templated assembly of droplet-derived PEG 76 Muller, P. and Schier, A.F. (2011) Extracellular movement of signaling
microtissues. Lab Chip 11, 2967–2975 molecules. Dev. Cell 21, 145–158
64 Zheng, Y. et al. (2012) Switching of macroscopic molecular recognition 77 Gudjonsson, T. et al. (2002) Normal and tumor-derived myoepithelial
selectivity using a mixed solvent system. Nat. Commun. 3, 831 cells differ in their ability to interact with luminal breast epithelial
65 Van Vlierberghe, S. et al. (2011) Biopolymer-based hydrogels as scaffolds cells for polarity and basement membrane deposition. J. Cell Sci. 115,
for tissue engineering applications: a review. Biomacromolecules 12, 39–50
1387–1408 78 Lu, P. et al. (2012) The extracellular matrix: a dynamic niche in cancer
66 Jabbari, E. (2011) Bioconjugation of hydrogels for tissue engineering. progression. J. Cell Biol. 196, 395–406
Curr. Opin. Biotechnol. 22, 655–660 79 Samchenko, Y. et al. (2011) Multipurpose smart hydrogel systems. Adv.
67 Gupta, R. and Sefton, M.V. (2011) Application of an endothelialized Colloid Interface Sci. 168, 247–262
modular construct for islet transplantation in syngeneic and 80 Damassa, D.A. et al. (1996) Purification and characterization of the sex
allogeneic immunosuppressed rat models. Tissue Eng. Part A 17, hormone-binding globulin in serum from Djungarian hamsters. Comp.
2005–2015 Biochem. Physiol. B: Biochem. Mol. Biol. 113, 593–599
68 Butler, M.J. and Sefton, M.V. (2012) Cotransplantation of adipose- 81 Davies, J.A. (2006) A method for cold storage and transport of viable
derived mesenchymal stromal cells and endothelial cells in a modular embryonic kidney rudiments. Kidney Int. 70, 2031–2034
construct drives vascularization in SCID/bg mice. Tissue Eng. Part A 82 Nelson, C.M. and Bissell, M.J. (2006) Of extracellular matrix, scaffolds,
18, 1628–1641 and signaling: tissue architecture regulates development, homeostasis,
69 Vallbacka, J.J. and Sefton, M.V. (2007) Vascularization and improved and cancer. Annu. Rev. Cell Dev. Biol. 22, 287–309
in vivo survival of VEGF-secreting cells microencapsulated in HEMA- 83 Streuli, C.H. et al. (1991) Control of mammary epithelial differentiation:
MMA. Tissue Eng. 13, 2259–2269 basement membrane induces tissue-specific gene expression in the
70 Birgersdotter, A. et al. (2005) Gene expression perturbation in vitro–a absence of cell-cell interaction and morphological polarity. J. Cell
growing case for three-dimensional (3D) culture systems. Semin. Biol. 115, 1383–1395
Cancer Biol. 15, 405–412 84 Paszek, M.J. et al. (2005) Tensional homeostasis and the malignant
71 Petersen, O.W. et al. (1992) Interaction with basement membrane phenotype. Cancer Cell 8, 241–254
serves to rapidly distinguish growth and differentiation pattern of 85 Guo, C.L. et al. (2012) Long-range mechanical force enables self-
normal and malignant human breast epithelial cells. Proc. Natl. assembly of epithelial tubular patterns. Proc. Natl. Acad. Sci. U.S.A.
Acad. Sci. U.S.A. 89, 9064–9068 109, 5576–5582
72 Chaffer, C.L. et al. (2011) Normal and neoplastic nonstem cells can 86 McLachlan, E. et al. (2007) Connexins and gap junctions in mammary
spontaneously convert to a stem-like state. Proc. Natl. Acad. Sci. gland development and breast cancer progression. J. Membr. Biol. 218,
U.S.A. 108, 7950–7955 107–121
73 Lacorre, D.A. et al. (2004) Plasticity of endothelial cells: rapid 87 Brisken, C. and O’Malley, B. (2010) Hormone action in the mammary
dedifferentiation of freshly isolated high endothelial venule gland. Cold Spring Harb. Perspect. Biol. 2, a003178

691

You might also like