You are on page 1of 8

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Biomedical Engineering

Advancing the design and delivery of CRISPR


antimicrobials
Jennie R. Fagen, Daphne Collias, Atul K. Singh and
Chase L. Beisel

Abstract binding and cleavage has given rise to a diverse set of


CRISPR-Cas systems are prokaryotic immune systems whose applications ranging from genome editing and gene
RNA-guided nucleases have been co-opted for applications regulation to in vitro diagnostics, real-time DNA and
ranging from genome editing and gene regulation to in vitro RNA imaging, and gene drives [1,2].
diagnostics and DNA imaging. Here, we review the current
efforts toward repurposing CRISPR nucleases as program- The standard mechanism of CRISPR-based genome
mable antimicrobials. Antimicrobial activity is achieved by editing is the directed repair of cleaved DNA. In
targeted cleavage of multidrug-resistance plasmids or the eukaryotic cells, the cleaved DNA is efficiently repaired
bacterial chromosome, resulting in antibiotic sensitivity or cell through ubiquitous mechanisms such as homology-
death. As part of the review, we discuss the different types of directed repair or non-homologous end joining. How-
nucleases available for CRISPR antimicrobials, the use of ever, in bacteria, multiple studies have indicated that
bacteriophages as delivery vehicles, and opportunities to DNA cleavage by CRISPR nucleases often cannot be
enhance antimicrobial activity, delivery, and specificity. repaired and is therefore lethal. One line of evidence is
Through further advances, these programmable DNA-targeting the dearth of naturally occurring genome-targeting
antimicrobials may help quell the spread of antimicrobial spacers in active CRISPR-Cas systems, even though
resistance and provide a tool for the manipulation of complex incorporation of such sequences regularly occurs during
microbial communities. naive spacer acquisition [3,4]. Separately, the intro-
duction of genome-targeting guide RNAs in bacteria
with an active CRISPR-Cas system arrested the cell
Addresses
Department of Chemical and Biomolecular Engineering, North Car- cycle, leading to eventual death or loss of large genomic
olina State University, Raleigh NC 27695, USA segments containing the target sequence [5,6]. These
insights helped inspire the use of CRISPR nucleases as
Corresponding author: Beisel, Chase L. (cbeisel@ncsu.edu) programmable antibacterial agents that could program-
mably and irreversibly destroy targeted DNA only in
Current Opinion in Biomedical Engineering 2017, 4:57–64
selected bacteria. This mechanism has been developed
primarily to address the rising challenge of antibiotic
This review comes from a themed issue on Synthetic Biology and
Biomedical Engineering
resistance and the diminishing supply of new small-
molecule drugs [7]. Here, we review how CRISPR nu-
Edited by Charlie A. Gersbach
cleases have been harnessed as programmable antimi-
Received 29 June 2017, revised 29 September 2017, accepted 2 crobial agents to combat human disease and the rise of
October 2017
antibiotic resistance, and we discuss opportunities for
the further development of this promising antimicrobial
https://doi.org/10.1016/j.cobme.2017.10.001 strategy.
2468-4511/© 2017 Elsevier Inc. All rights reserved.
CRISPR nuclease selection
Keywords CRISPR-Cas systems commonly act as immune systems
Bacteriophage, Cas9, CRISPR-Cas systems, sgRNA, Tail-fiber by cleaving foreign DNA or RNA, yet a diversity of
proteins. system types have been reported (Figure 1A) [8e10].
These system types are differentiated based on the
Introduction associated proteins responsible for acquisition and
CRISPR-Cas systems are adaptive immune systems in targeting as well as their modes of action. For instance,
bacteria and archaea whose CRISPR nucleases and Type II systems generate blunt-ended cuts in target
guide RNAs have formed the foundation of powerful DNA, Type III systems synergistically cleave target
tools in biotechnology and medicine. The guide RNAs RNA and DNA, and Type VI systems cleave target RNA
direct the nuclease to bind and cleave complementary followed by the non-discriminate cleavage of other
DNA or RNA sequences often flanked by a protospacer- RNAs [10e12]. Given that CRISPR-Cas systems are
adjacent motif (PAM). The ease of designing guide present in roughly half of bacteria, these endogenous
RNAs, the portability of the nuclease-guide RNA pair, systems could be co-opted as antimicrobials through the
and the broad applicability of programmable DNA delivery of a guide RNA targeting the bacterial genome

www.sciencedirect.com Current Opinion in Biomedical Engineering 2017, 4:57–64


58 Synthetic Biology and Biomedical Engineering

Figure 1

Current Opinion in Biomedical Engineering

CRISPR antimicrobial design. (A) Diversity of CRISPR-Cas systems with their respective nucleic-acid targets and mechanisms of attack. Each
displayed PAM is from a representative, well-characterized system. Type IV systems have not been experimentally characterized and are therefore
excluded here. (B) Potential accessory factors for augmenting CRISPR antimicrobials by blocking DNA break repair.

[5,13]. However, the utility of this approach is limited to antimicrobial. Cui and Bikard directly reported this
bacteria with functionally active CRISPR-Cas systems phenomenon in Escherichia coli, where cleavage events
and is sensitive to the expression and genetic stability of were often repaired by homologous recombination with
the CRISPR-Cas locus. A more robust and self- extra copies of the genome [21]. The repair was
contained antimicrobial design consists of introducing dependent on RecA and allowed the cells to survive
the designed guide RNA and the CRISPR nuclease, the attack when Cas9 targeted a broad range of non-
strategy used for virtually all CRISPR antimicrobials to- essential and even a few essential genes. What remains
date. unclear is why some sites led to potent killing and
whether repair will be a barrier to Cas9-based antimi-
While each type of nuclease has potential as an anti- crobials in other bacteria. Non-specific cytotoxicity from
microbial agent, the primary nuclease of choice has been Cas9 has also been reported [23,24], raising concerns
Cas9, the effector protein from Type II CRISPR-Cas about potential non-specific killing in any cells that
systems. Cas9 is the most thoroughly investigated express exogenous Cas9.
RNA-guided nuclease and has been the trailblazer for
current CRISPR technologies [14e18]. Cas9 has also Despite the intense focus on Cas9, Type I CRISPR-Cas
been the primary nuclease used in CRISPR antimicro- systems have also shown promise as antimicrobial
bials, where it has been successfully used to target agents. The Cas3 nuclease for these systems nick and
antibiotic-resistance genes encoded on plasmids or in processively degrade the non-bound strand of DNA in
the genome [19,20] as well as essential and non- the 30 -to-50 direction, leading to immediate destruction
essential chromosomal genes [5,13,21,22]. of large portions of DNA upstream of the target
sequence [25,26]. Accordingly, multiple groups have co-
One critical feature of Cas9 is that it generates a blunt- opted the I-E CRISPR-Cas system from E. coli for
end double-stranded break that is amenable to DNA potent and selective killing. Gomaa et al. demonstrated
repair, potentially compromising Cas9 as an the utility of the E. coli I-E system as a programmable
Current Opinion in Biomedical Engineering 2017, 4:57–64 www.sciencedirect.com
Advancing CRISPR antimicrobials Fagen et al. 59

antimicrobial and demonstrated activity when express- any accessory factors that will enhance antimicrobial
ing the system endogenously or heterologously [13]. activity will be an important part of the overall CRISPR
Furthermore, Gomaa et al. used the I-E system antimicrobial design.
expressed heterologously to kill individual or multiple
strains based on a unique or shared genomic sequence. CRISPR antimicrobial delivery
Yosef et al. took a similar approach to immunize E. coli Arguably the most pervasive challenge to the develop-
against the uptake of antibiotic resistance plasmids [27]. ment of CRISPR antimicrobials is efficient delivery to
Finally, Caliando and Voigt engineered E. coli with a diverse bacteria. To-date, CRISPR antimicrobials have
chromosomally integrated, inducible I-E CRISPR-Cas been principally encoded as DNA and delivered using
system to conditionally degrade regions of the genome bacteriophage particles [20e22,27] (Figure 2A). Bacte-
that are proprietary [26]. Critically, Gomaa et al. riophage (or phage for short) are adept at injecting their
observed low escape rates regardless of the genomic genetic material across the bacterial cell wall and into
target site, suggesting that Type I systems could offer the cytoplasm of its host. CRISPR antimicrobials have
more flexible antimicrobials than those reliant on Cas9 been encoded in two forms to ensure packaging by the
[13]. The drawback to Type I systems is that they phage particles: phagemids and the phage genome
require the coordinated expression of four to seven (Figure 2B). Phagemids are plasmids that contain
proteins, potentially complicating their delivery and packaging signals along with a selection marker and
expression in diverse bacteria. Instead, given that Type I origin-of-replication and therefore do not encode any
systems are the most prevalent type of CRISPR-Cas additional phage components that could inadvertently
system in nature [10], active endogenous systems affect cell physiology. As plasmids, phagemids can be
could be co-opted through the delivery of a genome- equipped with CRISPR antimicrobials through standard
targeting guide RNA [13]. in vitro assembly techniques. Accordingly, multiple
groups have used phagemids for phage-mediated de-
Recently, new types of CRISPR nucleases have been livery of CRISPR antimicrobials to E. coli and Staphylo-
discovered that could also serve as antimicrobial coccus aureus [19,20,22]. The second option is
agents. The Type V-A CRISPR effector protein Cpf1 introducing the CRISPR antimicrobial DNA into the
(Cas12a) functions similarly to Cas9 but tends to be phage genome. DNA manipulation is more laborious for
smaller and leaves a staggered DNA break [28,29]. It phage genomes than for phagemids, and phage genomes
remains to be seen whether this staggered cut is can introduce unwanted phage components. However,
readily repaired similar to the blunt cut from Cas9. A phage genomes regularly outcompete phagemids for
separate, recently discovered Type VI-A nuclease with packaging and encode factors that can block bacterial
antimicrobial potential is C2c2 (Cas13a). This protein immune systems such as restriction-modification sys-
specifically binds and cleaves complementary RNA but tems that degrade foreign DNA. Yosef et al. successfully
then undergoes a conformational change that leads to introduced a Type I CRISPR-Cas system into the
promiscuous RNA cleavage [30,31]. Abudayyeh genome of l phage that integrates into the bacterial
showed that this activity could arrest cell growth [30], genome, thereby enabling heritable targeting of
although the effect may be bacteriostatic rather than multidrug-resistance plasmids and stymying the spread
bactericidal. Given that other CRISPR nucleases likely of multidrug resistance through a bacterial population
await discovery, the best nuclease for antimicrobial [27]. CRISPR antimicrobials have been encoded in the
applications remains to be determined and could be genomes of temperate phage, where CRISPR is the sole
dependent on the mode of delivery and the physiology agent of cell killing. However, lytic phage could also be
of the target bacterium. promising delivery vehicles, as CRISPR could augment
the natural killing abilities of the phage. In the long
While DNA cleaved by Cas9 may undergo repair, addi- term, it will be interesting to see whether lytic or lyso-
tional effectors may be added to increase its lethality genic phage equipped with killing mechanisms such as
(Figure 1B). For instance, Cui and Bikard showed that CRISPR prove to serve as superior antimicrobials
the expression of Cas9 and the Mu phage protein GamS, [22,28].
an inhibitor of the RecBCD DNA repair machinery,
blocked DNA repair and resulted in no bacterial survi- For phage to deliver their genetic cargo, they must
vors [21]. Other viral proteins that inhibit DNA repair efficiently bind to the surface of the target microbeda
such as the Ref protein from P1 phage could similarly process known as adsorption. Phage traditionally adsorb
improve the efficacy of Cas9 and other CRISPR nucle- to a narrow range of bacteria, with most phage limited to
ases by hindering DNA break repair [32,33]. Finally, the a subset of bacterial strains within a single species. This
co-dominant negative RecA56 mutant has been shown narrow adsorption range presents an immediate chal-
to inhibit certain functions of the bacterial DNA repair lenge for utilizing phage as generalized DNA delivery
pathway [34], and could help to further prevent recov- vehicles. A potential solution is the use of a phage
ery from CRISPR-induced DNA cleavage. Going for- cocktail or broad-host phage such as P1 [35,36]. Another
ward, the selection of a potent CRISPR nuclease and solution being explored is to alter or exchange the tail
www.sciencedirect.com Current Opinion in Biomedical Engineering 2017, 4:57–64
60 Synthetic Biology and Biomedical Engineering

Figure 2

A Phage anatomy B Forms of DNA encoding CRISPR antimicrobials


Capsid
Origin of replication

DNA packaging sites

Tail sheath Phagemid


CRISPR Phage genome
nuclease(s)
Tail fibers Genetic material
Genome
packaged within capsid
targeting spacer(s)

Baseplate

C Modulation of phage adsorption range


Tail fiber exchange Tail fiber mutation

T7 WT host T3 WT host
Phagemid encoding
A B tail fiber genes (black)

Random mutagenesis
of tail fiber genes

Transfer T3 tail fiber


genes on T7 phagemid Produce phage library
with mutant tail fiber genes

Produce T7/T3
Screen for DNA delivery
hybrid
to bacteria of interest

Screen for DNA delivery A B C Bacteria


to bacteria of interest

T7 delivery range
A B Bacteria

T7 delivery range Shifted delivery range

T3 delivery range Expanded delivery range

T3 delivery range Shifted delivery range

Current Opinion in Biomedical Engineering

Engineering phage as delivery vehicles for CRISPR antimicrobials. (A) Anatomy of a representative tailed phage. (B) DNA-encoded CRISPR
antimicrobials can be incorporated into a phagemid or the phage genome for viral packaging and intracellular delivery. (C) Strategies for modifying tail
fiber proteins to alter the phage host range. The model T7 lytic phage has been the focus of recent phage engineering efforts through tail fiber gene
exchange or mutation.

Current Opinion in Biomedical Engineering 2017, 4:57–64 www.sciencedirect.com


Advancing CRISPR antimicrobials Fagen et al. 61

fiber proteins responsible for binding the bacterial cell Future challenges and opportunities
surface (Figure 2C). Recent studies using E. coli phage Even if DNA encoding a CRISPR antimicrobial is effi-
T7 showed that this phage can accommodate the tail ciently delivered to the target cell, the DNA must avoid
fiber proteins of related phage, thereby allowing delivery the host’s immune systems to allow sufficient antimi-
to varying enteric bacteria [37,38]. Yosef et al. further crobial expression. Different types of immune systems
applied directed evolution to the tail fiber proteins of are known, such as restriction-modification, CRISPR-
T7, resulting in increased infectivity in desired bacterial Cas, and abortive infection among others, and the spe-
strains [38]. A similar strategy could be employed to cific barriers can vary widely even between related
quickly tailor the host range to target all strains for a bacteria. Some of these barriers can be overcome
given pathogen or target a broad set of pathogens. through common techniques that have been developed
through efforts to improve DNA transformation [40].
Phage aside, other delivery methods are being explored For example, mimicry of the target’s DNA methylation
and could hold promise for broad intracellular DNA and codon optimization of any delivered DNA may help
delivery. Kang et al. developed polymer-based nano- to overcome the target bacterium’s restriction-
particles that delivered Cas9-sgRNA ribonucleoprotein modification systems and improve translation effi-
complexes to bacterial cells [39]. While the delivery ciency, respectively [41,42]. However, more work is
efficiency was low, this DNA-independent approach has needed to understand which major barriers are posed in
the advantage of bypassing innate bacterial defenses each strain and how to systematically overcome each
against mobile genetic elements as well as functioning barrier.
independently of the host’s transcription/translation
machinery. CRISPR antimicrobials have also been Aside from barriers to foreign DNA, there are multiple
introduced through conjugation [20]. While conjugation scenarios in which a fully functional CRISPR antimi-
is currently less efficient than phage-based delivery, crobial may fail to kill the target bacterium (Figure 3).
conjugation may be more broadly applicable as it can Multiple modes of heritable resistance have been sug-
occur between unrelated bacteria and requires minimal gested or demonstrated, such as loss of the target site, or
modification of the conjugative donor. Furthermore, mutation of the surface receptors for phage adsorption
conjugation could allow a CRISPR antimicrobial to [19]. Other potential mechanisms include the expres-
propagate through a mixed bacterial population, sion of compatible anti-CRISPR proteins in a lysoge-
potentially offering a means of community-wide sur- nized phage that could inhibit the activity of delivered
veillance fordand eradication ofdundesirable strains or CRISPR antimicrobials [43], or the presence of a
genetic traits. lysogenized phage that blocks infections by similar

Figure 3

Targeted killing with


A CRISPR antimicrobial
B Heritable resistance to killing C Off-target killing
Immune Unable to Superinfection Anti-CRISPR Mutated Guide Activation of Cas9 toxicity
Phage binding systems bind to host exclusion genomic target competition endogenous
to host guides
surface
receptor

Mutated
CRISPR or lost
antimicrobial host surface
DNA delivery Restriction receptor
enzyme Endogenous
CRISPR
RNA-guided
guides
endonuclease
complex
Unknown
mechanism

Chromosome Processing of
targeting and endogenous
cleavage guides

Cell death Cell survival Cell death

Current Opinion in Biomedical Engineering

Potential modes of heritable bacterial resistance or off-target killing. (A) Steps of effective delivery and targeted killing. (B) Potential modes of
heritable resistance from CRISPR antimicrobials through DNA degradation by the host’s immune systems, lost or mutated surface receptor for the
phage delivery vehicle, superinfection exclusion due to previous phage infection, expression of an antagonistic anti-CRISPR protein, lack of a complete
target site flanked by the appropriate PAM, or competitive binding of the CRISPR nuclease by host encoded guide RNAs. (C) Potential modes of
unintended bacterial killing through self-targeting using a native CRISPR array or cytotoxicity associated with nuclease overexpression.

www.sciencedirect.com Current Opinion in Biomedical Engineering 2017, 4:57–64


62 Synthetic Biology and Biomedical Engineering

phage through superinfection exclusion [44]. Despite process with little precedent for phage-based thera-
these potential mechanisms of resistance, current peutic strategies. Despite these envisioned challenges,
studies have found that deleterious mutations in the these efforts should continue given the increasing need
CRISPR antimicrobial constructs are the primary driver to combat multidrug-resistant infections and an
of escapes rather than innate bacterial resistance. For emerging emphasis on preserving a healthy human
instance, Bikard et al. found that all S. aureus cells that microbiome. There is also the potential to use CRISPR
survived phage-delivered Cas9 antimicrobials remained antimicrobials to selectively perturb complex microbial
susceptible to reinfection by the phage [19], while communities or clear niches for introduced bacteria.
Gomaa et al. found that E. coli cells expressing the I-E Overall, there remains incredible potential for CRISPR
CRISPR-Cas system and transformed with genome- as an antimicrobial agent that could greatly expand the
targeting CRISPR plasmid survived through recombi- umbrella of CRISPR technologies and address existing
nation within the guide RNAs prior to plasmid isolation. challenges in the prevention and treatment of human
Therefore, resistance to CRISPR antimicrobials may be infectious diseases.
less prominent, although future research efforts should
aim to elucidate and counteract the most common Acknowledgements
mechanisms of bacterial escape. This work was supported by the Bill and Melinda Gates Foundation [grant
number OPP1140021], the North Carolina Biotechnology Center [Tech-
nology Enhancement Grant], and the Bay Area Lyme Foundation
Finally, CRISPR antimicrobials could also adversely [Emerging Leader Award]. C.L.B. is a co-founder of Locus Biosciences and
impact non-targeted bacteria (Figure 3C). Aside from is an inventor on patent applications related to CRISPR technologies.
the potential for off-target cleavage, undesired killing
could result from nuclease cytotoxicity or promiscuous References
Papers of particular interest, published within the period of review,
cleavage activity at elevated expression levels have been highlighted as:
[23,24,45]. The introduction of Cas9 has also been
* of special interest
shown to interact with orphaned Type II CRISPR arrays, * * of outstanding interest
the natural form of guide RNAs, in Enterococcus faecalis
that could result in incidental self-targeting [46]. These 1. Sander JD, Joung JK: CRISPR-Cas systems for editing, regu-
challenges could be overcome in part by using a rare lating and targeting genomes. Nat Biotechnol 2014, 32:
347–355.
CRISPR nuclease that is unlikely to be compatible with
2. Wright AV, Nuñez JK, Doudna JA: Biology and applications of
an endogenous array. Rare nucleases would also have the CRISPR systems: harnessing nature’s toolbox for genome
added benefit of being unlikely to encounter inhibitory engineering. Cell 2016, 164:29–44.
anti-CRISPR proteins. Overall, CRISPR antimicrobials 3. Stern A, Keren L, Wurtzel O, Amitai G, Sorek R: Self-targeting by
face multiple potential hurdles to ensure efficient de- CRISPR: gene regulation or autoimmunity? Trends Genet
2010, 26:335–340.
livery and on-target killing, although the versatility and
tractability of CRISPR-Cas systems and phage would 4. Staals RHJ, Jackson SA, Biswas A, Brouns SJJ, Brown CM,
Fineran PC: Interference-driven spacer acquisition is domi-
suggest that each barrier can be overcome through nant over naive and primed adaptation in a native CRISPR–
further engineering efforts. Cas system. Nat Commun 2016, 7:12853.
5. Selle K, Klaenhammer TR, Barrangou R: CRISPR-based
screening of genomic island excision events in bacteria. Proc
Conclusions and future perspectives Natl Acad Sci U S A 2015, 112:8076–8081.
CRISPR antimicrobials represent one of many uses of 6. Vercoe RB, Chang JT, Dy RL, Taylor C, Gristwood T, Clulow JS,
CRISPR-derived technologies. The CRISPR antimi- Richter C, Przybilski R, Pitman AR, Fineran PC: Cytotoxic
chromosomal targeting by CRISPR/Cas systems can reshape
crobial field is still in its infancy and faces numerous bacterial genomes and expel or remodel pathogenicity
challenges for the design and delivery of these antimi- islands. PLoS Genet 2013, 9:e1003454.
crobials. We also envision future hurdles as CRISPR 7. Koch G, Yepes A, Förstner KU, Wermser C, Stengel ST,
antimicrobials move from the lab to animal models and Modamio J, Ohlsen K, Foster KR, Lopez D: Evolution of resis-
tance to a last-resort antibiotic in Staphylococcus aureus via
finally to human trials. Specifically, further investigation bacterial competition. Cell 2014, 158:1060–1071.
and engineering is needed to ensure that the phage 8. Koonin EV, Makarova KS, Zhang F: Diversity, classification and
delivery vehicles reach the site of infection and effi- evolution of CRISPR-Cas systems. Curr Opin Microbiol 2017,
ciently deliver the CRISPR payload to pathogens in the 37:67–78.

affected tissue. Depending on the pathogen type and 9. Shmakov S, Smargon A, Scott D, Cox D, Pyzocha N, Yan W,
Abudayyeh OO, Gootenberg JS, Makarova KS, Wolf YI, et al.:
infection site, CRISPR antimicrobials may be more or Diversity and evolution of class 2 CRISPR-Cas systems. Nat
less amenable as therapeutic agents; for instance, topical Rev Microbiol 2017, 15:169–182.
infections offering potentially simpler targets than gut 10. Makarova KS, Wolf YI, Alkhnbashi OS, Costa F, Shah SA,
infections while planktonic pathogens may be simpler Saunders SJ, Barrangou R, Brouns SJJ, Charpentier E, Haft DH,
et al.: An updated evolutionary classification of CRISPR-Cas
targets than intracellular pathogens or those that form systems. Nat Rev Microbiol 2015, 13:722–736.
biofilms. Aside from the challenge of targeting patho-
11. Chylinski K, Makarova KS, Charpentier E, Koonin EV: Classifi-
gens in vivo, additional barriers are anticipated as this cation and evolution of type II CRISPR-Cas systems. Nucleic
technology begins to navigate a regulatory approval Acids Res 2014, 42:6091–6105.

Current Opinion in Biomedical Engineering 2017, 4:57–64 www.sciencedirect.com


Advancing CRISPR antimicrobials Fagen et al. 63

12. Samai P, Pyenson N, Jiang W, Goldberg GW, Hatoum-Aslan A, This work generated a phage-encoded CRISPR antimicrobial that
Marraffini LA: Co-transcriptional DNA and RNA cleavage cleared plasmids encoding antibiotic resistance genes and protected
during type III CRISPR-Cas immunity. Cell 2015, 161: against subsequent challenge by an engineered lytic phage in E. coli.
1164–1174. This two-stage approach sought to replace the antibiotic bacteria with
sensitive strains rather than the eradication of pathogens.
13. Gomaa AA, Klumpe HE, Luo ML, Selle K, Barrangou R, Beisel CL:
* * Programmable removal of bacterial strains by use of genome- 28. Krom RJ, Bhargava P, Lobritz MA, Collins JJ: Engineered
targeting CRISPR-Cas systems. MBio 2014, 5. e00928–13. phagemids for nonlytic, targeted antibacterial therapies.
This work demonstrated the utility of heterologous and endogenous Nano Lett 2015, 15:4808–4813.
CRISPR-Cas systems as programmable antimicrobials. Importantly,
the authors quantitatively manipulated a microbial community using 29. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM,
this antimicrobial. Makarova KS, Essletzbichler P, Volz SE, Joung J, van der Oost J,
Regev A, et al.: Cpf1 is a single RNA-guided endonuclease of
14. Gasiunas G, Barrangou R, Horvath P, Siksnys V: Cas9-crRNA a class 2 CRISPR-Cas system. Cell 2015, 163:759–771.
ribonucleoprotein complex mediates specific DNA cleavage
for adaptive immunity in bacteria. Proc Natl Acad Sci U S A 30. Abudayyeh OO, Gootenberg JS, Konermann S, Joung J,
2012, 109:E2579–E2586. Slaymaker IM, Cox DBT, Shmakov S, Makarova KS,
Semenova E, Minakhin L, et al.: C2c2 is a single-component
15. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, programmable RNA-guided RNA-targeting CRISPR effector.
Charpentier E: A programmable dual-RNA-guided DNA endo- Science 2016, 353:aaf5573.
nuclease in adaptive bacterial immunity. Science 2012, 337:
816–821. 31. Liu L, Li X, Wang J, Wang M, Chen P, Yin M, Li J, Sheng G,
Wang Y: Two distant catalytic sites are responsible for C2c2
16. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, RNase activities. Cell 2017, 168:121–134. e12.
Arkin AP, Lim WA: Repurposing CRISPR as an RNA-guided
platform for sequence-specific control of gene expression. 32. Ronayne EA, Wan YCS, Boudreau BA, Landick R, Cox MM: P1
Cell 2013, 152:1173–1183. Ref endonuclease: a molecular mechanism for phage-
enhanced antibiotic lethality. PLoS Genet 2016, 12:e1005797.
17. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X,
Jiang W, Marraffini LA, et al.: Multiplex genome engineering 33. Wilkinson M, Troman L, Wan Nur Ismah WA, Chaban Y,
using CRISPR/Cas systems. Science 2013, 339:819–823. Avison MB, Dillingham MS, Wigley DB: Structural basis for the
inhibition of RecBCD by Gam and its synergistic antibacterial
18. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, effect with quinolones [Internet]. Elife 2016, 5.
Norville JE, Church GM: RNA-guided human genome engi-
neering via Cas9. Science 2013, 339:823–826. 34. Lauder SD, Kowalczykowski SC: Negative co-dominant inhibi-
tion of RecA protein function. Biochemical properties of the
19. Bikard D, Euler CW, Jiang W, Nussenzweig PM, Goldberg GW, RecA1, RecA13 and RecA56 proteins and the effect of
* * Duportet X, Fischetti VA, Marraffini LA: Exploiting CRISPR-Cas RecA56 protein on the activities of the wild-type RecA protein
nucleases to produce sequence-specific antimicrobials. Nat function in vitro. J Mol Biol 1993, 234:72–86.
Biotechnol 2014, 32:1146–1150.
This work equipped a S. aureus phage with a Cas9-based CRISPR 35. Yen M, Cairns LS, Camilli A: A cocktail of three virulent bac-
antimicrobial, which selectively eliminated plasmids conferring anti- teriophages prevents Vibrio cholerae infection in animal
biotic resistance. models. Nat Commun 2017, 8:14187.

20. Citorik RJ, Mimee M, Lu TK: Sequence-specific antimicrobials 36. Murooka Y, Harada T: Expansion of the host range of coli-
* * using efficiently delivered RNA-guided nucleases. Nat Bio- phage P1 and gene transfer from enteric bacteria to other
technol 2014, 32:1141–1145. gram-negative bacteria. Appl Environ Microbiol 1979, 38:
In this work, the authors showed the efficacy of phage-delivered 754–757.
CRISPR antimicrobials against enterohemorrhagic E. coli in cell cul-
37. Ando H, Lemire S, Pires DP, Lu TK: Engineering modular viral
ture and in a wax-moth infection model. * scaffolds for targeted bacterial population editing. Cell Syst
21. Cui L, Bikard D: Consequences of Cas9 cleavage in the 2015, 1:187–196.
chromosome of Escherichia coli. Nucleic Acids Res 2016, 44: This work developed a tailed bacteriophage delivery vehicle where the
4243–4251. DNA delivery range was altered through the exchange of tailfiber
genes.
22. Park JY, Moon BY, Park JW, Thornton JA, Park YH, Seo KS:
* Genetic engineering of a temperate phage-based delivery 38. Yosef I, Goren MG, Globus R, Molshanski-Mor S, Qimron U:
system for CRISPR/Cas9 antimicrobials against Staphylo- * Extending the host range of bacteriophage particles for DNA
coccus aureus. Sci Rep 2017, 7:44929. transduction. Mol Cell 2017, 66:721–728. e3.
This work engineered a S. aureus phage for Cas9-based antimicrobial This work further expanded the DNA delivery range of a tailed phage
delivery in part by altering the phage host range through manipulating through directed evolution.
the phage’s tail fibers.
39. Kang YK, Kwon K, Ryu JS, Lee HN, Park C, Chung HJ: Nonviral
23. Jiang W, Brueggeman AJ, Horken KM, Plucinak TM, Weeks DP: genome editing based on a polymer-derivatized CRISPR
Successful transient expression of Cas9 and single guide nanocomplex for targeting bacterial pathogens and antibiotic
RNA genes in Chlamydomonas reinhardtii. Eukaryot Cell resistance. Bioconjugate Chem 2017, 28:957–967.
2014, 13:1465–1469.
40. Waller MC, Bober JR, Nair NU, Beisel CL: Toward a genetic tool
24. Jiang Y, Qian F, Yang J, Liu Y, Dong F, Xu C, Sun B, Chen B, development pipeline for host-associated bacteria. Curr Opin
Xu X, Li Y, et al.: CRISPR-Cpf1 assisted genome editing of Microbiol 2017, 38:156–164.
Corynebacterium glutamicum. Nat Commun 2017, 8:15179.
41. Zhang G, Wang W, Deng A, Sun Z, Zhang Y, Liang Y, Che Y,
25. Sinkunas T, Gasiunas G, Fremaux C, Barrangou R, Horvath P, Wen T: A mimicking-of-DNA-methylation-patterns pipeline for
Siksnys V: Cas3 is a single-stranded DNA nuclease and ATP- overcoming the restriction barrier of bacteria. PLoS Genet
dependent helicase in the CRISPR/Cas immune system. 2012, 8:e1002987.
EMBO J 2011, 30:1335–1342.
42. Grosjean H, Fiers W: Preferential codon usage in prokaryotic
26. Caliando BJ, Voigt CA: Targeted DNA degradation using a genes: the optimal codon-anticodon interaction energy and
CRISPR device stably carried in the host genome. Nat the selective codon usage in efficiently expressed genes.
Commun 2015, 6:6989. Gene 1982, 18:199–209.

27. Yosef I, Manor M, Kiro R, Qimron U: Temperate and lytic bac- 43. Bondy-Denomy J, Garcia B, Strum S, Du M, Rollins MF, Hidalgo-
* * teriophages programmed to sensitize and kill antibiotic- Reyes Y, Wiedenheft B, Maxwell KL, Davidson AR: Multiple
resistant bacteria. Proc Natl Acad Sci U S A 2015, 112: mechanisms for CRISPR-Cas inhibition by anti-CRISPR pro-
7267–7272. teins. Nature 2015, 526:136–139.

www.sciencedirect.com Current Opinion in Biomedical Engineering 2017, 4:57–64


64 Synthetic Biology and Biomedical Engineering

44. Susskind MM, Botstein D, Wright A: Superinfection exclusion cyanobacterium Synechococcus elongatus UTEX 2973.
by P22 prophage in lysogens of Salmonella typhimurium. III. Microb Cell Fact 2016, 15:115.
Failure of superinfecting phage DNA to enter sieA+ lysogens.
Virology 1974, 62:350–366. 46. Price VJ, Huo W, Sharifi A, Palmer KL: CRISPR-Cas and
restriction-modification act additively against conjugative
45. Wendt KE, Ungerer J, Cobb RE, Zhao H, Pakrasi HB: CRISPR/ antibiotic resistance plasmid transfer in Enterococcus
Cas9 mediated targeted mutagenesis of the fast growing faecalis. mSphere 2016, 1. e00064–16.

Current Opinion in Biomedical Engineering 2017, 4:57–64 www.sciencedirect.com

You might also like