a logic-gated nanorobot for targeted transport of …...as a proof of principle, nanorobots loaded...

4
essential in the pairing of CFs and the formation of the n = 5/2 FQH state at low temperature. Our NMR experiments have demonstrated maximal spin polarization for the n = 5/2 FQH state over a wide range of electron densities. These measurements are consistent with the nonabelian Pfaffian state (5) and the anti-Pfaffian state (9, 10), while unambiguously ruling out the unpolarized (331) state (11), which had been the most likely abelian contender (13). However, the exciting prospect of topologically protected quantum op- erations using the n = 5/2 FQH state awaits a direct experimental demonstration of its nonabe- lian nature. References and Notes 1. D. C. Tsui, H. L. Stormer, A. C. Gossard, Phys. Rev. Lett. 48, 1559 (1982). 2. R. B. Laughlin, Phys. Rev. Lett. 50, 1395 (1983). 3. J. K. Jain, Phys. Rev. Lett. 63, 199 (1989). 4. R. L. Willett et al., Phys. Rev. Lett. 59, 1776 (1987). 5. G. Moore, N. Read, Nucl. Phys. B 360, 362 (1991). 6. M. Greiter, X. G. Wen, F. Wilczek, Nucl. Phys. B 374, 567 (1992). 7. C. Nayak, F. Wilczek, Nucl. Phys. B 479, 529 (1996). 8. C. Nayak, S. H. Simon, A. Stern, M. Freedman, S. Das Sarma, Rev. Mod. Phys. 80, 1083 (2008). 9. M. Levin, B. I. Halperin, B. Rosenow, Phys. Rev. Lett. 99, 236806 (2007). 10. S.-S. Lee, S. Ryu, C. Nayak, M. P. A. Fisher, Phys. Rev. Lett. 99, 236807 (2007). 11. B. Halperin, Helv. Phys. Acta 56, 75 (1983). 12. M. Dolev, M. Heiblum, V. Umansky, A. Stern, D. Mahalu, Nature 452, 829 (2008). 13. I. P. Radu et al., Science 320, 899 (2008). 14. R. L. Willett, L. N. Pfeiffer, K. W. West, Proc. Natl. Acad. Sci. U.S.A. 106, 8853 (2009). 15. V. Venkatachalam, A. Yacoby, L. Pfeiffer, K. West, Nature 469, 185 (2011). 16. A. Bid et al., Nature 466, 585 (2010). 17. R. Morf, Phys. Rev. Lett. 80, 1505 (1998). 18. I. Dimov, B. I. Halperin, C. Nayak, Phys. Rev. Lett. 100, 126804 (2008). 19. A. E. Feiguin, E. Rezayi, K. Yang, C. Nayak, S. Das Sarma, Phys. Rev. B 79, 115322 (2009). 20. G. Gamez, K. Muraki, n=5/2 fractional quantum Hall state in low-mobility electron systems: Different roles of disorder; available at http://arxiv.org/abs/1101.5856 (2011). 21. W. Desrat et al., Phys. Rev. Lett. 88, 256807 (2002). 22. N. Kumada, K. Muraki, Y. Hirayama, Phys. Rev. Lett. 99, 076805 (2007). 23. J. H. Smet et al., Nature 415, 281 (2002). 24. Materials and methods are available as supporting material on Science Online. 25. X. G. Wen, Phys. Rev. Lett. 66, 802 (1991). 26. M. Stern et al., Phys. Rev. Lett. 105, 096801 (2010). 27. T. D. Rhone et al., Phys. Rev. Lett. 106, 196805 (2011). 28. I. V. Kukushkin, K. von Klitzing, K. Eberl, Phys. Rev. Lett. 82, 3665 (1999). 29. F. D. M. Haldane, Phys. Rev. Lett. 51, 605 (1983). 30. V. W. Scarola, K. Park, J. K. Jain, Nature 406, 863 (2000). 31. N. Read, D. Green, Phys. Rev. B 61, 10267 (2000). 32. N. Freytag, M. Horvatić , C. Berthier, M. Shayegan, L. P. Lévy, Phys. Rev. Lett. 89, 246804 (2002). Author contributions: L.T. performed all the RD-NMR and transport measurements presented in this paper. G.G. performed transport measurements to optimize the samples. N.K. advised on the RD-NMR measurements. K.M. grew heterostructures and fabricated samples. L.T. and K.M. analyzed the data and wrote the paper. Supporting Online Material www.sciencemag.org/cgi/content/full/science.1216697/DC1 Materials and Methods Supplementary Text Figs. S1 and S2 References (3345) 2 August 2011; accepted 19 January 2012 Published online 26 January 2012; 10.1126/science.1216697 A Logic-Gated Nanorobot for Targeted Transport of Molecular Payloads Shawn M. Douglas,* Ido Bachelet,* George M. ChurchWe describe an autonomous DNA nanorobot capable of transporting molecular payloads to cells, sensing cell surface inputs for conditional, triggered activation, and reconfiguring its structure for payload delivery. The device can be loaded with a variety of materials in a highly organized fashion and is controlled by an aptamer-encoded logic gate, enabling it to respond to a wide array of cues. We implemented several different logical AND gates and demonstrate their efficacy in selective regulation of nanorobot function. As a proof of principle, nanorobots loaded with combinations of antibody fragments were used in two different types of cell-signaling stimulation in tissue culture. Our prototype could inspire new designs with different selectivities and biologically active payloads for cell-targeting tasks. T he DNA origami method (1), in which a multiple-kilobase single-stranded scaffoldis folded into a custom shape by interac- tion with hundreds of oligonucleotide staplestrands, has proved to be extremely versatile for creating custom two- and three-dimensional as- semblies (24) that can template precise arrange- ment of diverse components (57). DNA can also be used to construct devices that perform robotic tasks such as sensing, computation, and actuation (811). Recently, a three-dimensional DNA origami box integrating both structural and computational components was described (12). Inspired by these advances, we sought to design a robotic DNA device capable of selectively in- terfacing with cells to deliver signaling molecules to cell surfaces. Using cadnano, a computer-aided design tool for DNA origami (13), we created a nanorobot in the form of a hexagonal barrel with dimensions of 35 nm × 35 nm × 45 nm. The barrel consists of two domains that are covalently attached in the rear by single-stranded scaffold hinges, and can be noncovalently fastened in the front by staples modified with DNA aptamer based locks (Fig. 1A). Initial self-assembly proceeds in a one-pot reac- tion in which 196 oligonucleotide staple strands direct a 7308-base filamentous phagederived scaf- fold strand into its target shape during a thermal- annealing ramp of rapid heating followed by slow cooling (14). A clasp system based on DNA locks and DNA keys was previously used to control the opening of the lid on a DNA box (12). To operate our device in response to proteins, we designed a DNA aptamerbased lock mechanism that opens in response to binding antigen keys (Fig. 1, B and C). Our lock system was inspired by aptamer beacons (15) and structure-switching aptamers (16), which typically undergo target-induced switching between an aptamer-complement du- plex and an aptamer-target complex. We incor- porated aptamer-complement duplexes on the left and right sides of the front of the barrel, such that the aptamer strands are attached to one do- main and partially complementary strands are attached to the other domain. When both aptamers recognize their targets, the lock duplexes disso- ciate, and the nanorobotacting as an entropic springundergoes a drastic reconfiguration to expose its previously sequestered surfaces. We found that shorter duplexes gave better sensitivity and faster activation rates, but at a cost of increased spontaneous activation (see text S2). We chose a lock duplex length of 23 base pairs (bp) for sub- sequent experiments because it displayed similar sensitivity to a shorter 16-bp duplex (activation at 10 pM) without the unacceptable loss in sensi- tivity observed for longer duplexes (30, 37, and 44 bp required 1 nM). Payloads that were premodified by covalent attachment to the 5end of a 15-base single- stranded DNA oligonucleotide linker were loaded inside the nanorobot. To enable multivalent in- teraction with surface receptors for potent cell stimulation (17), we aimed to load at least two payload molecules per robot. Twelve payload at- tachment sites were arranged in an inward-facing ring in the middle of the barrel (Fig. 1, A and C) to enable different payload orientations and spac- ings. The sites are staple strands with 3extensions Wyss Institute for Biologically Inspired Engineering and De- partment of Genetics, Harvard Medical School, Boston, MA 02115, USA. *These authors contributed equally to this work. To whom correspondence should be addressed. E-mail: http://arep.med.harvard.edu/gmc/email.html www.sciencemag.org SCIENCE VOL 335 17 FEBRUARY 2012 831 REPORTS on February 16, 2012 www.sciencemag.org Downloaded from

Upload: others

Post on 14-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A Logic-Gated Nanorobot for Targeted Transport of …...As a proof of principle, nanorobots loaded with combinations of antibody fragments were used in two different types of cell-signaling

essential in the pairing of CFs and the formationof the n = 5/2 FQH state at low temperature.

Our NMR experiments have demonstratedmaximal spin polarization for the n = 5/2 FQHstate over awide range of electron densities. Thesemeasurements are consistent with the nonabelianPfaffian state (5) and the anti-Pfaffian state (9, 10),while unambiguously ruling out the unpolarized(331) state (11), which had been the most likelyabelian contender (13). However, the excitingprospect of topologically protected quantum op-erations using the n = 5/2 FQH state awaits adirect experimental demonstration of its nonabe-lian nature.

References and Notes1. D. C. Tsui, H. L. Stormer, A. C. Gossard, Phys. Rev. Lett.

48, 1559 (1982).2. R. B. Laughlin, Phys. Rev. Lett. 50, 1395 (1983).3. J. K. Jain, Phys. Rev. Lett. 63, 199 (1989).4. R. L. Willett et al., Phys. Rev. Lett. 59, 1776 (1987).5. G. Moore, N. Read, Nucl. Phys. B 360, 362 (1991).6. M. Greiter, X. G. Wen, F. Wilczek, Nucl. Phys. B 374,

567 (1992).7. C. Nayak, F. Wilczek, Nucl. Phys. B 479, 529

(1996).

8. C. Nayak, S. H. Simon, A. Stern, M. Freedman,S. Das Sarma, Rev. Mod. Phys. 80, 1083 (2008).

9. M. Levin, B. I. Halperin, B. Rosenow, Phys. Rev. Lett. 99,236806 (2007).

10. S.-S. Lee, S. Ryu, C. Nayak, M. P. A. Fisher,Phys. Rev. Lett. 99, 236807 (2007).

11. B. Halperin, Helv. Phys. Acta 56, 75 (1983).12. M. Dolev, M. Heiblum, V. Umansky, A. Stern, D. Mahalu,

Nature 452, 829 (2008).13. I. P. Radu et al., Science 320, 899 (2008).14. R. L. Willett, L. N. Pfeiffer, K. W. West, Proc. Natl.

Acad. Sci. U.S.A. 106, 8853 (2009).15. V. Venkatachalam, A. Yacoby, L. Pfeiffer, K. West,

Nature 469, 185 (2011).16. A. Bid et al., Nature 466, 585 (2010).17. R. Morf, Phys. Rev. Lett. 80, 1505 (1998).18. I. Dimov, B. I. Halperin, C. Nayak, Phys. Rev. Lett. 100,

126804 (2008).19. A. E. Feiguin, E. Rezayi, K. Yang, C. Nayak, S. Das Sarma,

Phys. Rev. B 79, 115322 (2009).20. G. Gamez, K. Muraki, n=5/2 fractional quantum Hall

state in low-mobility electron systems: Different rolesof disorder; available at http://arxiv.org/abs/1101.5856(2011).

21. W. Desrat et al., Phys. Rev. Lett. 88, 256807 (2002).22. N. Kumada, K. Muraki, Y. Hirayama, Phys. Rev. Lett. 99,

076805 (2007).23. J. H. Smet et al., Nature 415, 281 (2002).24. Materials and methods are available as supporting

material on Science Online.

25. X. G. Wen, Phys. Rev. Lett. 66, 802 (1991).26. M. Stern et al., Phys. Rev. Lett. 105, 096801 (2010).27. T. D. Rhone et al., Phys. Rev. Lett. 106, 196805

(2011).28. I. V. Kukushkin, K. von Klitzing, K. Eberl, Phys. Rev. Lett.

82, 3665 (1999).29. F. D. M. Haldane, Phys. Rev. Lett. 51, 605 (1983).30. V. W. Scarola, K. Park, J. K. Jain, Nature 406, 863

(2000).31. N. Read, D. Green, Phys. Rev. B 61, 10267 (2000).32. N. Freytag, M. Horvati!, C. Berthier, M. Shayegan,

L. P. Lévy, Phys. Rev. Lett. 89, 246804 (2002).

Author contributions: L.T. performed all the RD-NMRand transport measurements presented in this paper.G.G. performed transport measurements to optimize thesamples. N.K. advised on the RD-NMR measurements.K.M. grew heterostructures and fabricated samples. L.T. andK.M. analyzed the data and wrote the paper.

Supporting Online Materialwww.sciencemag.org/cgi/content/full/science.1216697/DC1Materials and MethodsSupplementary TextFigs. S1 and S2References (33–45)

2 August 2011; accepted 19 January 2012Published online 26 January 2012;10.1126/science.1216697

A Logic-Gated Nanorobot for TargetedTransport of Molecular PayloadsShawn M. Douglas,* Ido Bachelet,* George M. Church†

We describe an autonomous DNA nanorobot capable of transporting molecular payloads tocells, sensing cell surface inputs for conditional, triggered activation, and reconfiguring itsstructure for payload delivery. The device can be loaded with a variety of materials in a highlyorganized fashion and is controlled by an aptamer-encoded logic gate, enabling it to respond to awide array of cues. We implemented several different logical AND gates and demonstrate theirefficacy in selective regulation of nanorobot function. As a proof of principle, nanorobots loadedwith combinations of antibody fragments were used in two different types of cell-signalingstimulation in tissue culture. Our prototype could inspire new designs with different selectivitiesand biologically active payloads for cell-targeting tasks.

The DNA origami method (1), in which amultiple-kilobase single-stranded “scaffold”is folded into a custom shape by interac-

tion with hundreds of oligonucleotide “staple”strands, has proved to be extremely versatile forcreating custom two- and three-dimensional as-semblies (2–4) that can template precise arrange-ment of diverse components (5–7). DNA canalso be used to construct devices that performrobotic tasks such as sensing, computation, andactuation (8–11). Recently, a three-dimensionalDNA origami box integrating both structural andcomputational components was described (12).

Inspired by these advances, we sought to design arobotic DNA device capable of selectively in-terfacingwith cells to deliver signalingmoleculesto cell surfaces.

Using cadnano, a computer-aided design toolfor DNA origami (13), we created a nanorobot inthe form of a hexagonal barrel with dimensionsof 35 nm ! 35 nm ! 45 nm. The barrel consists oftwo domains that are covalently attached in therear by single-stranded scaffold hinges, and canbe noncovalently fastened in the front by staplesmodifiedwithDNAaptamer–based locks (Fig. 1A).Initial self-assembly proceeds in a one-pot reac-tion in which 196 oligonucleotide staple strandsdirect a 7308-base filamentous phage–derived scaf-fold strand into its target shape during a thermal-annealing ramp of rapid heating followed byslow cooling (14).

A clasp system based on DNA locks andDNA keys was previously used to control the

opening of the lid on a DNA box (12). To operateour device in response to proteins, we designed aDNA aptamer–based lock mechanism that opensin response to binding antigen keys (Fig. 1, Band C). Our lock system was inspired by aptamerbeacons (15) and structure-switching aptamers(16), which typically undergo target-inducedswitching between an aptamer-complement du-plex and an aptamer-target complex. We incor-porated aptamer-complement duplexes on theleft and right sides of the front of the barrel, suchthat the aptamer strands are attached to one do-main and partially complementary strands areattached to the other domain.When both aptamersrecognize their targets, the lock duplexes disso-ciate, and the nanorobot—acting as an entropicspring—undergoes a drastic reconfiguration toexpose its previously sequestered surfaces. Wefound that shorter duplexes gave better sensitivityand faster activation rates, but at a cost of increasedspontaneous activation (see text S2). We chose alock duplex length of 23 base pairs (bp) for sub-sequent experiments because it displayed similarsensitivity to a shorter 16-bp duplex (activation at10 pM) without the unacceptable loss in sensi-tivity observed for longer duplexes (30, 37, and44 bp required 1 nM).

Payloads that were premodified by covalentattachment to the 5! end of a 15-base single-strandedDNAoligonucleotide linker were loadedinside the nanorobot. To enable multivalent in-teraction with surface receptors for potent cellstimulation (17), we aimed to load at least twopayload molecules per robot. Twelve payload at-tachment sites were arranged in an inward-facingring in the middle of the barrel (Fig. 1, A and C)to enable different payload orientations and spac-ings. The sites are staple strands with 3! extensions

Wyss Institute for Biologically Inspired Engineering and De-partment of Genetics, Harvard Medical School, Boston, MA02115, USA.

*These authors contributed equally to this work.†To whom correspondence should be addressed. E-mail:http://arep.med.harvard.edu/gmc/email.html

www.sciencemag.org SCIENCE VOL 335 17 FEBRUARY 2012 831

REPORTS

on

Febr

uary

16,

201

2ww

w.sc

ienc

emag

.org

Down

load

ed fr

om

smd
Comment on Text
Since there's no established definition of nanorobot, we felt we had some flexibility in using the term. We asked robotics expert Rob Wood at Harvard about whether he thought it was appropriate to call this a nanorobot. He said to describe what it does in general terms, and we said that it senses friend versus foe, and when a foe is detected, it changes its shape and attacks. He agreed that this sounded like a function that a robot would perform.
smd
Comment on Text
Origami literally translates to "folding paper" in Japanese, so the term "DNA folding paper" originally seemed a bit awkward to native Japanese speakers.
smd
Comment on Text
As a graduate student in Dr. William Shih's lab, I found that designing each new DNA origami was tedious and error-prone. Fortunately, it was also highly predictable and repetitive, and thus ripe for optimization by software. It took me about two years of tinkering with Python scripts to figure out I needed to start over from scratch and build a graphical user interface. I worked on the new version for about 3 months before Prof. Shih noticed I had been neglecting my experiments and suggested that I shelve the project. I stubbornly insisted that the software tool would be transformative and was thus my top priority, and fortunately, I was given more time. After an additional 5 months of late-night coding, I had the first working version of Cadnano. The impact of this work was immediate and significant: before implementation, we typically needed one month to design a new DNA origami shape. Since implementing the software, the design process can now be completed in one afternoon for a simple shape, and a few days for a more complex origami.
smd
Comment on Text
In the first paragraph we aim to provide concise context for the work. Here we are reporting on technology development, so we cite the foundational work related to DNA origami.
smd
Comment on Text
The second paragraph describes the basic "specs" of the nanorobot. More technical detail, such as the specific DNA sequences, is provided in the supplementary notes.
smd
Comment on Text
The "minimum" number of two payload molecules is somewhat arbitrary — we could have aimed for 3, 4, or more. This is another case where working in new territory affords great flexibility when setting our goals. Many details do not need to be optimized at the outset and can be calibrated in future works.
smd
Comment on Text
Supplemental notes can be a fantastic repository for technical detail that is crucial for the work, but beyond the scope of the centeral "story" of the paper.
smd
Comment on Text
This work may seem like it represented a huge leap forward in technology. However, once we converged on the idea of targeted delivery to cells, implementing the device felt very incremental. In fact, we only had to integrate a handful of molecular components and methods that were already well validated on their own: 3D DNA origami shapes, DNA aptamer switches, non-site-specific DNA-protein conjugation, and cell-stimulating antibody fragments.
smd
Comment on Text
Ned Seeman, the founder of the field of DNA nanotechnology, rates Paul Rothemund's invention of DNA origami as the greatest subsequent contribution to the field. This seminal paper is definitely worth a read.
smd
Comment on Text
The 'proof-of-concept' is meant to hint at future potential of DNA nanotechnology, but we will see that it is still very far from a practical implementation (i.e., a therapeutic that can be used to treat human disease).
Page 2: A Logic-Gated Nanorobot for Targeted Transport of …...As a proof of principle, nanorobots loaded with combinations of antibody fragments were used in two different types of cell-signaling

that are complementary to a linker sequenceattached to the intended cargo (Fig. 1D). Afternanorobot folding and purification, cargo loadingwas carried out by adding linker-modified pay-load in molar excess to attachment sites and in-cubating at room temperature for 12 hours. Twotypes of cargo were loaded: 5-nm gold nano-particles covalently attached to 5!-thiol–modifiedlinkers (18), and various Fab´ antibody fragmentsthat were covalently attached to 5!-amine–modified linkers using a HyNic/4FB couplingkit (Solulink, San Diego, California). We usednegative-stain transmission electron microscopy(TEM) to analyze the device in closed and openstates, with and without cargo (Fig. 1F). We ob-served by manual counting that on average fourattachment sites were populated when loadinggold nanoparticles, and three sites were populatedwhen loading antibody fragments (fig. S11).

One obstacle to overcome in constructing a“spring-loaded” device was to ensure assemblyto high yield in its closed state. Like hands thatset a mousetrap, two “guide” staples were in-

corporated adjacent to the lock sites that span thetop and bottom domains of the device (Fig. 1E).The guide staples include 8-base toehold over-hangs and could be removed after folding andpurification steps by adding a 10:1 excess of fullycomplementary strands to the mixture (19). Weobserved that foldingwith the aid of guide staplesincreased the yield of closed robots from 48%to 97.5%, as assessed by manual counting ofnanorobots images by TEM (fig. S17).

To examine nanorobot function, we selected apayload such that robot activation would be cou-pled to labeling of an activating cell (Fig. 2A).Robots loaded with fluorescently labeled anti-body fragments against human leukocyte antigen(HLA)–A/B/Cwere mixed with different cell typesexpressing humanHLA-A/B/C and various “key”combinations (described below) and were ana-lyzed by flow cytometry. In the absence of thecorrect combination of keys, the robot remainedinactive. In the inactive state, the sequesteredantibody fragments were not able to bind the cellsurface, resulting in a baseline fluorescence sig-

nal. However, when the robot encountered theproper combination of antigen keys, it was freedto open and bind to the cell surface via its anti-body payload, causing an increase in fluorescence.We used key-neutralizing antibodies in compe-titive inhibition control experiments to verify thatnanorobots were not activated by a non–ligand-based mechanism (fig. S25).

The robot could be programmed to activate inresponse to a single type of key by using thesame aptamer sequence in both lock sites. Al-ternatively, different aptamer sequences could beencoded in the locks to recognize two inputs.Both locks needed to be opened simultaneouslyto activate the robot. The robot remained inactivewhen only one of the two locks was opened. Thelock mechanism is thus equivalent to a logicalAND gate, with possible inputs of cell surfaceantigens not binding or binding (0 or 1, respec-tively) to aptamer locks, and possible outputs ofremaining closed or a conformational rearrange-ment to expose the payload (0 or 1, respectively)(Fig. 2B).

FA C+ +

lock

edun

lock

edtop

front

top

side

side

++

D E

guideremoval

guide stapleswith toeholds

++

B

23bp 18–24

TTTT

TTTT

Fig. 1. Design and TEM analysis of aptamer-gated DNA nanorobot. (A) Sche-matic front orthographic view of closed nanorobot loaded with a protein pay-load. Two DNA-aptamer locks fasten the front of the device on the left (boxed)and right. (B) Aptamer lock mechanism, consisting of a DNA aptamer (blue)and a partially complementary strand (orange). The lock can be stabilized in adissociated state by its antigen key (red). Unless otherwise noted, the lockduplex length is 24 bp, with an 18- to 24-base thymine spacer in the nonaptamerstrand. (C) Perspective view of nanorobot opened by protein displacement ofaptamer locks. The two domains (blue and orange) are constrained in the rear by

scaffold hinges. (D) Payloads such as gold nanoparticles (gold) and antibody Fab´fragments (magenta) can be loaded inside the nanorobot. (E) Front and sideviews show guide staples (red) bearing 8-base toeholds aid assembly of nano-robot to 97.5% yield in closed state as assessed by manual counting. Afterfolding, guide staples are removed by addition of fully complementary oligos(black). Nanorobots can be subsequently activated by interaction with antigenkeys (red). (F) TEM images of robots in closed and open conformations. Leftcolumn, unloaded; center column, robots loaded with 5-nm gold nanoparticles;right column, robots loaded with Fab´ fragments. Scale bars, 20 nm.

17 FEBRUARY 2012 VOL 335 SCIENCE www.sciencemag.org832

REPORTS

on

Febr

uary

16,

201

2ww

w.sc

ienc

emag

.org

Down

load

ed fr

om

smd
Comment on Text
We knew from experience that gold nanoparticles would provide good contrast in electron microscopy. While they are not strictly necessary for the subsequent experiments, including them in Fig 1F helps reinforce that we do in fact have good spatial control over where the payloads are located.
smd
Comment on Text
Here is another case where we borrowed a well-validated method in the DNA nanotechnology field: toehold switches. The initial low yield of "closed" nanorobots would have been a deal-breaker for the project, but luckily we saw that our yield more than doubled! This simple modification essentially worked on the first try, which is a testament to the power and versatility of DNA nanotechnology.
smd
Comment on Text
The DNA oligos are attached to surface lysines on the antibody fragments in a non-site-specific manner. Thus, we believe that some fraction of the Fab's are oriented such that their binding site is occluded by the origami. Achieving a site-specific attachment would have been more technically challenging, but fortunately it not necessary to see some positive signal in this work and could be left for a future follow-up project.
smd
Comment on Text
In retrospect, it was overly bombastic to describe our locks as logic gates, especially since the locks are not reusable and couldn't be used to implement meaningful computation. In practice, the locks are more like a single-use "rip cord" or a spring-loaded latch.
smd
Comment on Text
When reporting on a technological or methodological advance, we use Figure 1 to illustrate the overall concept in the paper. Typically, we include a schematic illustration and provide some routine characterization of the system or device.
smd
Comment on Text
In this paper, the entire story is built around a single structure, so we found it helpful to include a fancy "hero" rendering of the 3D model.
smd
Comment on Text
We should not have shown equal-length dual-arrows between the two panels in Figure B, as we did not have any evidence that the activation was fully reversible. We might have collected more data to test what happens, and then adjusted the relative arrow lengths depending on the reversibility between the locked and unlocked states.
smd
Comment on Text
While it's fun to draw cartoon schematics, I love to use as much experimental data as possible to tell the story. One particularly attractive feature of DNA origami is that it can be visualized at the individual particle level using electron microscopy, and visualized at the population level using gel electrophoresis.
smd
Comment on Text
Scale bars should always be included with every micrograph.
Page 3: A Logic-Gated Nanorobot for Targeted Transport of …...As a proof of principle, nanorobots loaded with combinations of antibody fragments were used in two different types of cell-signaling

To test the generality and robustness of theaptamer-encoded logic gating, we designed sixdifferent robots using pairwise combinations ofaptamer locks drawn from a set of three well-characterized aptamer sequences: 41t, againstplatelet-derived growth factor (PDGF) (20), shownin red; TE17 (21), shown in yellow; and sgc8c (22),shown in blue (Fig. 2C). The six robot versions—plus a permanently locked negative control and ano-lock positive control—were loaded with fluo-rescently labeled antibody to human HLA-A/B/CFab´ and used to probe six different cell lines, eachexpressing different profiles of the “key” anti-gens recognized by the three chosen aptamers.

A Burkitt’s lymphoma cell line (Ramos) activatednone of the robots. An acute myeloid leukemiacell line (Kasumi-1) activated all robots. A thirdcell line, isolated from a patient with large granularlymphocytic leukemia, aggressive NK type (NKL)(23), activated robots with two 41t locks, twoTE17 locks, andwith one 41t and one TE17 lock.The baseline negative signal for NKL cells washigher than other cell lines because of NKL back-ground fluorescence. A fourth cell line, isolatedfrom acute T cell leukemia (Jurkat), activated ro-bots bearing two TE17 locks, two sgc8c locks, aswell as one TE17 lock and one sgc8c lock. Thefifth line was isolated from acute lymphoblastic

leukemia (CCRF-CEM) and had a similar pro-file to Jurkat cells. Finally, a neuroblastoma cellline (SH-SY5Y) activated robots with two sgc8clocks. The lower intensity of the positive signalsin the neuroblastoma cell line may be caused bythe lower level of surface HLA-A/B/C expressedon these cells, and fluorescence intensities maybe affected in general by expression levels of bothaptamer keys and antibody payload targets.

We further examined the performance of thelogic-gating mechanism in three ways. First, wetested the ability of the robot to selectively bind toa single cell type (NKLPTK7+/+) in a mixed pop-ulation of two cell types (NKLPTK7+/+ andRamos).

wholeblood++ NKL

cells

forward scatter

side

sca

tter

Ramos

CD20CD20

D E F G

HLA

-A/B

/C

HLA

-A/B

/C

HLA

-A/B

/C

CD20

NKLPTK7+/+ RamosNKLPTK7+/+ RamosNKLPTK7+/+

C

HLA–A/B/C HLA–A/B/C HLA–A/B/C HLA–A/B/C HLA–A/B/C HLA–A/B/C

Acutelymphoblasticleukemia Neuroblastoma

AggressiveNK leukemia

AcutemyeloblasticleukemiaBurkitt's lymphoma T-cell leukemia

B

+

001

+

111

+

00 0

+

010

A

FITC

++

FITC

++

FITC

coun

tco

unt

coun

t

++

FITC:anti-HLAcargo

HLA+

HLA+

HLA+key+

cells labeled

cells labeled

cells unlabeled

unlocked

locked

locked

+

Fig. 2. Analysis of logic-gated nanorobot activation by different cell types. (A)Experimental scheme. Nanorobots were loaded with fluorescently labeled anti-body Fab´ fragments to human HLA-A/B/C. In their unlocked state, nanorobotswill bind to any cell expressing the HLA-A/B/C antigen (top). They remain inactivein the presence of key– cells (middle) but activate upon engaging key+ cells(bottom). (B) Truth table for nanorobot activation. The aptamer-encoded locksfunction as an ANDgate responding tomolecular inputs (keys) expressed by cells.Color match indicates lock-and-key match. Aptamer-antigen activation stateserves as input; output is manifested as nanorobot conformation. (C) Eightdifferent nanorobot versions (10 to 100 fmol, loaded with anti–HLA-A/B/C anti-body fragments at a molar excess of 20) were tested with six different cell typesexpressing various combinations of antigen keys by incubating for 5 hours. Each

histogram displays the count of cells versus fluorescence due to anti–HLA-A/B/Clabeling. (D) NKL cells (50,000 per sample, with transiently induced expression ofPTK7 (NKLPTK7+/+), Ramos cells (100,000 per sample), fluorescein isothiocyanate(FITC)–labeled antibody to human CD20 (0.1 mg/ml), and permanently lockedrobots loaded with allophycocyanin–labeled antibody to human HLA-A/B/C Fab´were incubated at room temperature for 5 hours. No labeling is observed, aslocked robots remain inactive. (E) Unlocked robots react with both cell pop-ulations. (F) sgc8c-gated robots react only with the cell population expressing thePTK7 key. (G) Forward- versus side-scatter dot plot of 4:1 mixture of healthyhumanwhole-blood leukocytes andNKL cells. Nanorobots loaded with anti-CD33Fab´ payload and gated with 41t locks selectively label NKL cells. Off-targetbinding occurred in 0.6% of sampled cells.

www.sciencemag.org SCIENCE VOL 335 17 FEBRUARY 2012 833

REPORTS

on

Febr

uary

16,

201

2ww

w.sc

ienc

emag

.org

Down

load

ed fr

om

smd
Comment on Text
In general, the order of figures in a manuscript does not correspond to the temporal order in which the experiments were performed in the lab. The best projects often play out in a non-linear fashion, where new experiments are quickly improvised when the data is unexpected, and then later fit into a cohesive narrative. In our case, Figures 2D, 2E, 2F represent the very first experimental validation we performed for the entire system, and obtaining that first result made for the most exciting day of the entire project. Only after we convinced ourselves that the nanotobots would activate in a cell-specific manner in a mixed background did we go back and systematically test the various lock, key, and cell line combinations shown in Figure 2C.
smd
Comment on Text
It can take a long time to translate an exciting preliminary result into a published manuscript. The first experimental data that convinced us the project would be viable was obtained in July 2010. Fortunately, in recent years, sharing of preprints (on servers such as bioRxiv) greatly accelerating the time between data acquisition and dissemination of results to the community.
smd
Comment on Text
Figure 2B is also dense. It is a condensed version of Figure 2A that introduces the idea that when the two locks on the nanorobot recognize different keys, then both keys must be present to fully unlock the robot.
smd
Comment on Text
If I recall correctly, this was George Church's blood, and we spiked in NKL cells.
smd
Comment on Text
Figure 2A is visually dense, but you can think of it like three little comic strips. The top row, starting with an unlocked robot, we add the cargo (FITC fluorescently labelled antibody fragments that bind to HLA) and then mix it with cells that express HLA. We then image the cells using flow cytometry, resulting in a histogram with a right-shifted FITC peak, indicating that the cells are bound by the cargo. The second row shows the case where the robot is locked and the cell lacks the key, so there is no shift in the FITC peak. The third row uses cells that do express the key, and we now see the FITC peak shift similar to the positive control in the first row.
smd
Comment on Text
In Figure 2 of a technology report, we typically want to present a thorough and convincing characterization of our new system or device. For us, the big question a skeptical reader would be trying to answer is something like "Does this nanorobot actually target cells, and if so, how well?" Here, we first show a cartoon schematic that is meant to briefly explain the experimental approach we are using to answer that question, i.e. fluorescently labeled nanorobots in various activation states, measured by flow cytometry. In Fig 2C we demonstrate the modular versatility of the nanorobot by measuring activation with several lock combos and cell lines. In Fig 2D-G we highlight the robots' specificity by showing they do not activate when encountering non-target bystander cells.
Page 4: A Logic-Gated Nanorobot for Targeted Transport of …...As a proof of principle, nanorobots loaded with combinations of antibody fragments were used in two different types of cell-signaling

Three versions of the robot were tested: a per-manently closed version in which the guide sta-ples were not removed, an open version in whichno guide staples or aptamer locks were included,and a gated version using two sgc8c locks, whichwe expected to target only the PTK7-expressingNKL cells (Fig. 2, D to F, and figs. S22 and S23).The gated version discriminated between the twocell types, only binding to NKL cells. Second,to further simulate physiological conditions, wemixed NKL cells with healthy whole-blood leu-kocytes and found that robots gated with 41tlocks discriminated those NKL cells with highprecision (Fig. 2G and fig. S19). Third, we ex-amined the sensitivity of the nanorobots to cellconcentration using a background of 1e6 Ramoscells, tested for the ability of an sgc8c-gated robotto bind to JurkatPTK7+/+ cells. Nanorobots wereactivated for every cell count, from 1e6 cells downto the single-cell level (fig. S24).

Finally, we investigated the ability of an ac-tivated robot to interface with cells and stimulatetheir signaling in separate inhibition and activa-tion tasks. We first chose to target NKL cellsusing a pair of 41t locks, and loaded the robotswith a combination of antibody to human CD33and antibody to human CDw328 Fab´ fragments,which have been shown to induce growth arrest inleukemic cells (24). The robots induced growtharrest in NKL cells in a dose-dependent fash-ion (Fig. 3, A and B). The robots suppressed JunN-terminal kinase (JNK) (Fig. 3C) and Akt (pro-tein kinase B) signaling in this process (fig. S27).We then used T cells to test various methods of

signaling pathway activation (Fig. 3, D to F).Robots loaded with combinations of antibody tohuman CD3e Fab´ and antibody to flagellin Fab´were mixed with T cells and found to induceactivation (Fig. 3, D and F). Previous work hasshown that flagellin can augment T cell activa-tion (25). We found that our nanorobots wereable to collect flagellin from a 100 pg/ml solutionand induce augmented Tcell activation (Fig. 3, Eand F). These findings demonstrate that therobots can induce a variety of tunable changesin cell behavior. Furthermore, biologically activepayloads may be bound indirectly via interac-tions with antibody fragments, enabling applica-tions in which the robot carries out a scavengingtask before targeted payload delivery.

References and Notes1. P. W. K. Rothemund, Nature 440, 297 (2006).2. Y. Ke et al., Nano Lett. 9, 2445 (2009).3. S. M. Douglas et al., Nature 459, 414 (2009).4. H. Dietz, S. M. Douglas, W. M. Shih, Science 325,

725 (2009).5. A. Kuzuya et al., ChemBioChem 10, 1811 (2009).6. N. V. Voigt et al., Nat. Nanotechnol. 5, 200

(2010).7. N. Stephanopoulos et al., Nano Lett. 10, 2714

(2010).8. M. N. Stojanovic, D. Stefanovic, Nat. Biotechnol. 21,

1069 (2003).9. G. Seelig, D. Soloveichik, D. Y. Zhang, E. Winfree,

Science 314, 1585 (2006).10. Y. Benenson, B. Gil, U. Ben-Dor, R. Adar, E. Shapiro,

Nature 429, 423 (2004).11. B. Ding, N. C. Seeman, Science 314, 1583

(2006).12. E. S. Andersen et al., Nature 459, 73 (2009).

13. S. M. Douglas et al., Nucleic Acids Res. 37, 5001(2009).

14. See supporting material on Science Online.15. N. Hamaguchi, A. Ellington, M. Stanton, Anal.

Biochem. 294, 126 (2001).16. R. Nutiu, Y. Li, J. Am. Chem. Soc. 125, 4771 (2003).17. J. J. Boniface et al., Immunity 9, 459 (1998).18. A. P. Alivisatos et al., Nature 382, 609 (1996).19. B. Yurke, A. J. Turberfield, A. P. Mills Jr., F. C. Simmel,

J. L. Neumann, Nature 406, 605 (2000).20. L. S. Green et al., Biochemistry 35, 14413 (1996).21. Z. Tang et al., Anal. Chem. 79, 4900 (2007).22. Y. F. Huang et al., ChemBioChem 10, 862 (2009).23. M. J. Robertson et al., Exp. Hematol. 24, 406

(1996).24. C. Vitale et al., Proc. Natl. Acad. Sci. U.S.A. 98,

5764 (2001).25. Z. Ye, C. M. L. Lee, G. W. Sun, Y. H. Gan, Infect. Immun.

76, 2541 (2008).

Acknowledgments: We thank J. Ritz for providing NKL cells;C. Reynolds for technical assistance with NKL cells; C. Strongand G. McGill for assistance with 3D visualization usingMolecular Maya; A. Marblestone for helpful discussions; andJ. Markson for comments on the manuscript. S.M.D. holds aCareer Award at the Scientific Interface from the BurroughsWellcome Fund and a Wyss Technology DevelopmentFellowship, and I.B. is supported by a fellowship from theLife Sciences Research Foundation. S.M.D. and I.B. designedand performed the experiments and analyzed the data; S.M.D.,I.B., and G.M.C. wrote the manuscript.

Supporting Online Materialwww.sciencemag.org/cgi/content/full/335/6070/831/DC1Materials and MethodsSOM TextFigs. S1 to S27Table S1

16 September 2011; accepted 12 January 201210.1126/science.1214081

CD69102 103 1040

coun

t +anti-CD3!+anti-Flg

unloaded

+anti-CD3!+anti-Flg+Flagellin

F

T cell

E

+anti-CD3!+anti-Flg

+anti-CD3!+anti-Flg:Flagellin

+ +

Flagellin

D

T cell+anti-CD3!+anti-Flg

+

B C control

gated

unlockedcontrol

gated

unlocked

% G

0/Apo

ptot

ic

(p)-

JNK

med

ian

robot conc. (nM) robot conc. (nM)

0

10

20

30

40

50

60

0 0.01 0.1 1 10 1000

1000

2000

3000

4000

5000

1001010.10.010

A

+

PDGF aptamer locks+anti-CD33

+anti-CDw328

Aggressive NKleukemia cells

modifiedsignaling

activatednanorobotsbind to cells

Fig. 3. Nanorobots manipulate target cell signaling. (A) Experimentalscheme. A single dose of nanorobots loaded with an equal mixture of anti-body to human CD33 and antibody to human CDw328/Siglec-7 Fab´ frag-ments (cyan and magenta, respectively, each at a molar excess of 10 overnanorobots) and gated by 41t locks recognizing platelet-derived growthfactor (PDGF) were used to treat NKL cells at various concentrations (0 to100 nM). Phosphorylation of JNK was measured after 72 hours by intra-cellular flow cytometry. (B) NKL cells (10,000 per sample) treated with nano-robots were analyzed after 72 hours for cell cycle distribution by propidium

iodide (5 mg/ml). (C) Phosphorylation level of JNK as a function of robotconcentration (lowest, 0; highest, 100 nM) after 72 hours. Error bars for (B)and (C) represent SEM of two biological replicates. (D and E) Incrementalactivation of T cells by nanorobots loaded with antibody to human CD3e(blue) and antibody to flagellin Fab´ (black). Nanorobots (50 nM) with (E) andwithout (D) pre-incubation with flagellin (red; 100 pg/ml) were reacted withJurkat T cells (pink) for 1 hour at 37°C. (F) Histograms showing T cell acti-vation after nanorobot treatment, as measured by labeling with FITC-labeledantibody to CD69.

17 FEBRUARY 2012 VOL 335 SCIENCE www.sciencemag.org834

REPORTS

on

Febr

uary

16,

201

2ww

w.sc

ienc

emag

.org

Down

load

ed fr

om

smd
Comment on Text
In Figure 3 of this type of paper, we typically want to showcase the most dramatic and convincing "functional" demonstration of the new technology. In Figure 2, we demonstrated binding to cells, but for any real future application of this technology, we will want the device to actually do something, and the most obvious thing to do with a cell is to stimulate it to change state. Again, none of the stimulation assays we performed were novel by themselves; we used well-characterized activators and readouts. The novel aspect is only that our nanorobot device was mediating the activation.
smd
Comment on Text
Here, we use (p)-JNK as an indicator that the expected apoptotic signaling pathways were engaged, and the measured cell death in Fig 3B is not merely a side effect of, for example, nonspecific nanorobot-induced toxicity.