You are on page 1of 25

Accelerated Article Preview

Article
doi:10.1038/nature23467

Type III CRISPRCas systems produce cyclic oligoadenylate second

W
messengers
OleNiewoehner, CarmelaGarcia-Doval, Jakob T.Rostl, ChristianBerk, FrankSchwede, LaurentBigler, JonathanHall,

IE
Luciano A.Marraffini & MartinJinek

EV
This is a PDF file of a peer-reviewed paper that has been accepted for publication. Although unedited, the content has

PR
been subjected to preliminary formatting. Nature is providing this early version of the typeset paper as a service to our
customers. The text and figures will undergo copyediting and a proof review before the paper is published in its final form.
Please note that during the production process errors may be discovered which could affect the content, and all legal
disclaimers apply.

LE
C
TI
AR
ED
AT

Cite this article as: Niewoehner, O. et al. Type III CRISPRCas systems produce cyclic oligoadenylate second messengers. Nature
http://dx.doi.org/10.1038/nature23467 (2017).
ER
EL

Competing financial interests statement: F.S. is an employee of BIOLOG Life Science Institute GmbH, which markets nucleotide analogues
used in this study. The other authors declare no competing financial interests
C
AC

received 20 June; accepted 10 July 2017.


Accelerated Article Preview Published online 19 July 2017.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article doi:10.1038/nature23467

Type III CRISPRCas systems produce

W
cyclic oligoadenylate second messengers

IE
OleNiewoehner1*, CarmelaGarcia-Doval1*, JakobT.Rostl2, ChristianBerk3, FrankSchwede4, LaurentBigler5, JonathanHall3,
LucianoA.Marraffini2 and MartinJinek1

EV
In many prokaryotes, type III CRISPRCas systems detect and degrade invasive genetic elements by an RNA-guided, RNA-

PR
targeting multisubunit interference complex. The CRISPR-associated protein Csm6 additionally contributes to interference
by functioning as a standalone ribonuclease that degrades invader RNA transcripts, but the mechanism linking invader
sensing to Csm6 activity is not understood. Here we show that Csm6 proteins are activated through a second messenger
generated by the type III interference complex. Upon target RNA binding by the interference complex, its Cas10 subunit
converts ATP into a cyclic oligoadenylate product, which allosterically activates Csm6 by binding to its CARF domain. CARF
domain mutations that abolish allosteric activation inhibit Csm6 activity in vivo, and mutations in the Cas10 Palm domain

LE
phenocopy loss of Csm6. Together, these results point to an unprecedented mechanism for regulation of CRISPR interference
that bears striking conceptual similarity to oligoadenylate signalling in mammalian innate immunity.

Clustered regularly interspaced short palindromic repeat (CRISPR) loci


and CRISPR-associated (Cas) proteins constitute an adaptive prokar-
C
domains17. Our structural studies further revealed that Csm6 c ontains
a conserved, positively charged cleft at the dimeric interface of the
TI
yotic immune system directed against invasive genetic elements such CARF domains. The CARF domain is a variant of the Rossmann
bacteriophages and plasmids13. Class 1 CRISPR-Cas systems, com- fold and has been proposed to function in nucleotide binding or
prising type I, III and IV systems, mediate nucleic acid interference nucleic acid sensing13,22. Moreover, a recent crystal structure of the
AR

by means of a multiprotein-RNA complex composed of a processed CARF domain-containing protein Csx3 revealed specific binding to
CRISPR RNA (crRNA) and several Cas protein subunits4. In type III a linear tetraribonucleotide23,24. Therefore, we hypothesized that the
systems, the interference complex (known as Csm complex in type ribonuclease activity of Csm6 might be allosterically regulated by oligo-
III-A/D systems and Cmr complex in type III-B/C systems) is assem- nucleotide ligands (Extended Data Fig. 1). To test this, we first analysed
bled from the signature multidomain protein Cas10 (Csm1 in type the effect of linear tetraadenylates on the RNase activity of TtCsm6 in
ED

III-A/D or Cmr2 in type III-B/C systems) and additional Cas proteins assays that employed either a fluorophore-labelled ssRNA substrate
(Csm2-5 in type III-A/D, or Cmr1 and Cmr3-6 in type III-B/C)5. In (Fig. 1a), or a fluorogenic RNA substrate whose cleavage results in a quan-
these systems, interference is transcription-dependent68. The target tifiable increase in fluorescence signal (Fig. 1b, Extended Data Fig. 2).
RNA-bound complex functions as a sequence-specific endoribonu- TtCsm6 was potently activated by tetraadenylate (A4); moreover,
clease (RNase) to cleave the target RNA transcript and in addition has the presence of a 2,3-cyclic phosphate group further potentiated
AT

a target RNA-stimulated non-specific deoxyribonuclease (DNase) activation (Fig. 1a, b). Profiling a panel of oligonucleotides of varying
activity that cleaves single-stranded DNA6,912. lengths revealed that the 2,3-cyclic phosphate-terminated tetradeny-
Members of the Csm6 or the related Csx1 protein families are late (A4>P) had the strongest effect on TtCsm6 (Fig. 1c, Extended
frequently encoded within type III CRISPR-Cas systems 13. These Data Fig. 2a). To test whether oligoA-mediated activation was a general
ER

proteins are characterized by the presence of an N-terminal CRISPR- property of Csm6 enzymes, we additionally examined evolutionarily
associated Rossmann Fold (CARF) domain and a C-terminal Higher divergent Csm6 proteins from Methanothermobacter thermoauto-
Eukaryotes and Prokaryotes Nucleotide-binding (HEPN) RNase trophicus (MtCsm6, type III-D) and Enterococcus italicus (EiCsm6,
domain13,14. In the Staphylococcus epidermidis type III-A system, type III-A) (Extended Data Fig. 2b). Both MtCsm6 and EiCsm6 were
Csm6 is required for efficient antiplasmid and antiviral immunity15,16. efficiently activated by oligoA nucleotides (Extended Data Fig. 2c, d),
EL

Csm6 proteins function as RNases1618 that have been shown to non- with MtCsm6 displaying strongest activation by A4>P (Extended Data
specifically degrade invader-derived RNA transcripts, providing an Fig. 2e) and EiCsm6 by 2,3-cyclic phosphate-terminated hexaadenylate
additional interference mechanism that complements the nuclease (A6>P, Fig. 1d, Extended Data Fig. 2f). Together, these results demon-
activities of the type III interference complex16. As Csm6 ribonucleases strate that Csm6-family ribonucleases are activated by oligoadenylate
C

do not appear to physically associate with the Cas10-containing inter- nucleotides.


ference complex1921, the mechanism linking their nuclease activity to
AC

invader recognition is not known.


The CARF domain mediates allosteric activation of
Allosteric activation of CRISPR-associated Csm6 Csm6
ribonucleases To quantify the activation of Csm6 ribonucleases, we analysed the rib-
We previously showed that Csm6 is a homodimeric RNase that onuclease activity of EiCsm6 as a function of A6>P concentration,
contains a conserved active site at the dimer interface of the HEPN obtaining a half-maximal effective concentration (EC50) of ~60 nM
1
Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland. 2Laboratory of Bacteriology, The Rockefeller University, 1230 York Avenue, New York,
NY 10065-6399, USA. 3Department of Chemistry and Applied Biosciences, Institute for Pharmaceutical Sciences, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland. 4BIOLOG Life Science
Institute GmbH, Flughafendamm 9a, D-28199 Bremen, Germany. 5Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
*These authors contributed equally to this work.

0 0 M onth 2 0 1 7 | V O L 0 0 0 | N A T U R E | 1
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

(Extended Data Fig. 3). Since HEPN domain ribonucleases typically wild-type complex (Fig. 3c). These results indicate that activator gener-
bind their RNA substrates with micromolar affinities25,26, this strongly ation is dependent on target RNA binding but activator is not generated
suggests that the A6>P activator is recognized by an allosteric binding by cleavage of target RNA. Moreover, the observed hyperactivity of the
site in EiCsm6 that is distinct from the ribonuclease active site in the RNase-deficient EiCsm(1-5) complex implies that production of the
HEPN domain. Based on the proposed function of CARF domains Csm6 activator is itself allosterically activated by target RNA binding
in binding nucleotide ligands 13,22, we surmised that the CARF by the type III interference complex.

W
domain functions as the allosteric sensor. To test this, we engineered
a g lutamine-to-alanine substitution in the conserved CARF domain The Palm domain of Cas10 generates a cyclic oligoA
motif Ser112Gln116 in EiCsm6 (Extended Data Fig. 4a), which product
maps to the dimeric CARF domain interface in TtCsm6 and would Having ruled out the crRNA-guided target RNA cleavage activity as

IE
correspond to the ligand-binding face in a canonical Rossmann fold the source of the allosteric activator, we examined other enzymatic
domain (Extended Data Fig. 4b). In contrast to wild-type EiCsm6, the activities within the type III interference complex. The Cas10 (Csm1)
Q116A EiCsm6 mutant (dEiCsm6CARF) no longer responded to A6>P subunit of the complex contains two putative catalytic domains. The

EV
(Fig. 2a,b), indicating that disruption of the CARF domain motif abro- histidine-aspartate (HD) domain of Cas10 possesses sequence non-
gated allosteric activation and confirming that the CARF domain is specific DNase activity and mediates target RNA-dependent DNA
critical for the sensing of oligoA ligands. degradation912. In turn, the Palm domain shares structural similarity
with nucleotidyl cyclases and nucleotide polymerases and contains a

PR
Csm6 is activated by a second messenger generated by putative, conserved Gly-Gly-Asp-Asp (GGDD) catalytic motif29. To test
the type III interference complex whether either of the domains was required for the production of the
Since Csm6 proteins function in the context of type III CRISPR-Cas Csm6 activator, we generated EiCsm(1-5) complexes harbouring inac-
systems, we speculated that allosteric activation of Csm6 is dependent tivating point mutations in the HD domain (dCas10HD) or in the Palm
on the activity multisubunit type III CRISPR interference complex. domain motif (dCas10Palm) of EiCas10. Whereas inactivation of the HD

LE
Guided by a processed crRNA, the Cas10-containing complex binds domain had no effect on the production of the Csm6 activator, muta-
to a target RNA, which leads to sequence-specific target RNA cleavage tions in the Palm domain motif abrogated Csm6 activation (Fig. 4a),
and concurrently stimulates sequence non-specific DNase activity indicating that the Cas10 Palm domain generates the Csm6 activator.
of the complex912. Given that Csm6 proteins have not been found Previous structural studies of the Cas10 subunit (Cmr2) from the

C
to be physically associated with the interference complex19,20,27, we type III-B interference complex of Pyrococcus furiosus revealed that the
hypothesized that the complex would additionally generate a d iffusible putative Palm domain active site can accommodate two ATP molecules,
allosteric activator of Csm6 in a target RNA-dependent manner. To while the GGDD motif coordinates divalent metal ions30. Based on this,
TI
this end, we heterologously expressed and purified the interference we tested whether the Palm domain functions as an o ligoA s ynthetase
complex, hereafter referred to as EiCsm(1-5), from the type III-A by directly catalysing 5-3 phosphodiester formation between ATP
AR

system of Enterococcus italicus (Extended Data Fig. 5a). The complex molecules. To this end, we incubated the EiCsm(1-5) complex in the
displayed subunit stoichiometry consistent with the general molecular presence of target RNA, Mg2+ ions and [-32P]-ATP, and visualized
architecture of type III interference complexes (Extended Data Fig. 5b) reaction products by denaturing polyacrylamide gel electrophoresis
and was capable of cleaving a cognate RNA target, yielding a charac- and phosphorimaging. In the presence of cognate target RNA, the
teristic pattern of cleavage products at six-nucleotide intervals19,20,28. EiCsm(1-5) complex converted ATP into a product displaying lower
As expected, target RNA cleavage was dependent on the presence of electrophoretic mobility (Fig. 4b, Extended Data Fig. 6a). In contrast,
ED

a conserved aspartate residue in the Csm3 subunits (Asp32), since product formation did not occur when a non-cognate RNA was
alanine substitution of the Csm3 aspartate (dCsm3D32A) resulted in supplied. Moreover, the activity was dependent on the presence of an
loss of RNase activity (Extended Data Fig. 5c). intact GGDD motif in the Palm domain, whereas inactivating point
To test whether the allosteric activator of Csm6 is produced by the mutations in the Cas10 HD domain or in Csm3 had no effect (Fig. 4b).
AT

type III interference complex, we incubated the EiCsm(1-5) complex Together, these results indicate that within the Csm(1-5) complex, the
together with a target RNA oligonucleotide in the presence of ATP Cas10 Palm domain is allosterically activated by target RNA binding
and magnesium ions at 37oC. We subsequently heat-inactivated the to convert ATP into a specific oligoA product.
complex, removed precipitated proteins by centrifugation and added a Liquid chromatography-mass spectrometry (LC-MS) analysis
ER

fraction of the deproteinized supernatant to an assay reaction mixture revealed that the EiCsm(1-5) complex generates a single dominant
containing EiCsm6 and the fluorogenic RNA substrate. We observed molecular species with a molecular mass of 1974.75 Da, consistent
robust activation of EiCsm6, with 0.01% of the added supernatant with the product being either A6>P or a cyclic hexaadenylate (c-A6),
having slightly stronger effect than 500nM A6>P, suggesting that the both having a theoretical mass of 1974.32 Da (Fig. 4c, Extended Data
EiCsm(1-5) complex produces a lowmolecularweight product capable Fig. 6b). However, the retention time of the purified product (3.93min)
EL

of activating EiCsm6 (Fig. 3a). Activator production was dependent was markedly different from that of a synthetic A6>P standard
on the presence of ATP, which could not be substituted by any of the (3.74min), suggesting that the product is cyclic and not linear
three other nucleotide triphosphates, and required Mg2+ ions, as (Extended Data Fig. 7). Moreover, the product was refractory to treat-
addition of EDTA was inhibitory (Fig. 3b). Moreover, the activator ments with polynucletide kinase (PNK), alkaline phosphatase and the
C

was only p roduced in the presence of a cognate target RNA but not pyrophosphatase RppH, indicating that it lacked 5- or 3-phosphate,
when a non-cognate control RNA was used instead (Fig. 3b). Together, 2,3-cyclic or 5-triphosphate moieties. Digestion of the product with
AC

these results show that the EiCsm(1-5) complex generates a diffusible S1 nuclease yielded a single terminal species, further confirming that
Csm6 activator by an ATP-, magnesium- and target RNA-dependent the product is a cyclic, 3-5 linked hexanucleotide (Fig. 4d, Extended
mechanism. Data Fig. 8a). The purified c-A6 product potently stimulated the RNase
Since crRNA-guided target RNA cleavage by the type III interference activity of EiCsm6 with an EC50 of ~3.5 nM (Fig. 4e, Extended Data
complex yields hexanucleotide products that could potentially act as Fig. 8b). Together, these results imply that the Palm domain of the
Csm6 activators19,20,28, we tested whether the ribonuclease activity of Cas10 subunit is a cyclic oligoadenylate synthetase that uses ATP to
the EiCsm(1-5) complex was required to generate the EiCsm6 activator. generate a cyclic hexaadenylate product. Notably, the molecular mass
Supernatant from a reaction containing EiCsm(1-5) complex in which of the product generated in the presence of all ribonucleotide triphos-
the crRNA-guided RNase activity was abolished by the D32A mutation phates (ATP, UTP, CTP and GTP) remained unchanged (Extended
in the Csm3 subunits activated EiCsm6 even more strongly than the Data Fig. 9), indicating that the EiCsm(1-5) complex only incorporates

2 | N A T U R E | V O L 0 0 0 | 0 0 month 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

adenosines into the cyclic product and does not generate mixed-base are allosterically activated by cyclic oligoadenylate activators through
species. Overall, our results thus suggest that cyclic oligoadenylates are binding to the CARF domain. We also demonstrate that the activator
allosteric activators of Csm6 enzymes in vivo and that the activators are is synthesized by the Palm domain of the Cas10 subunit of the type
synthesized by the Cas10 subunit of the type III interference complex III interference complex in response to target RNA recognition. Our
upon target RNA recognition. In light of our TtCsm6 data, these results findings thus reveal that type III systems use second messengers to
also hint that other type III CRISPR-Cas systems might generate cyclic signal target RNA sensing to CARF domain ribonucleases, as has
been proposed in a previous comparative genomic study22. Although

W
tetranucleotides instead.
bacteria utilize numerous nucleotide metabolites such as c-di-AMP,
In vivo activity of Csm6 is dependent on the c-di-GMP or ppGpp for signalling and stress response pathways31,
oligoadenylate cyclase activity of the Csm complex cyclic oligoA metabolites have not been reported to function as s econd

IE
In the S. epidermidis type III-A CRISPR-Cas system, the RNase messengers in prokaryotes before. However, the mechanism brings up
activities of Csm6 and Csm3 result in the degradation of phage striking parallels with the vertebrate innate immune system, where
transcripts, which is required for efficient anti-phage immunity when detection of viral double-stranded RNAs by oligoadenylate synthetase

EV
the target site is located in a gene transcript expressed late during enzymes triggers production of linear 2-5 linked oligoadenylates that
infection16. To corroborate our biochemical studies, we tested whether allosterically activate ribonuclease L32.
the in vivo activity of Csm6 depends on allosteric activation by cyclic We conclude that the type III interference complex is not only a
oligoadenylates generated by Cas10. We first determined whether crRNA-guided RNase and DNase, but also a cyclic oligoadenylate

PR
EiCsm6 can function together with the S. epidermidis type III system synthetase (Fig. 6). The likely mechanism of cyclic oligoA synthesis by
in the absence of its endogenous csm6 gene. To this end, we engineered Cas10 would involve linear oligomerization, followed by final cycli-
Staphylococcus aureus strains containing an RNase-deficient version zation of the linear intermediate through its 5-triphosphate group
of the S. epidermidis type III-A system that was programmed to target (Extended Data Fig. 10). As the cyclic oligoadenylate synthetase
the gp43 gene of phage NM16, contained an inactivating point activity is stimulated by target RNA binding, this would provide a fail-

LE
mutation in the SeCsm3 protein, and lacked SeCsm6. The strains co- safe interference mechanism in case the intrinsic DNase and RNase
expressed either wild-type EiCsm6 or the unresponsive EiCsm6 Q116A activities of the type III interference complex are insufficient to clear
mutant (dEiCsm6CARF) from a plasmid. Cell cultures were infected with the invader DNA and/or its transcripts, such as when the target gene
NM16 phage at a low multiplicity of infection (MOI ~0.25) and cell is expressed late in the phage lytic cycle or contains mismatches to

C
survival was measured over time (Fig. 5a). Staphylococci e xpressing the crRNA guide16. Although the interference in type III CRISPR-
EiCsm6 were equally, if not more, immune as those expressing cata- Cas systems might in part be due to non-specific RNA degradation
lytically active SeCsm6, demonstrating that the SeCsm(1-5) complex by Csm6 enzymes, in analogy with the proposed mechanism of type
TI
can activate EiCsm6. When the dEiCsm6CARF mutant was expressed VI systems33, we note that the Csm6 activity appears to be confined
instead, CRISPR-mediated immunity was lost and staphylococci to phage-derived transcripts in the S. epidermidis type III system16.
AR

succumbed to phage infection, as did control strains expressing the As CARF domain ribonucleases of the Csm6 and Csx1 families are
HEPN RNase active site mutants of SeCsm6 or EiCsm6 (Fig. 5a). only associated with type III CRISPR-Cas systems containing a Cas10
These results suggest that oligoA sensing by the Csm6 CARF domain subunit with a catalytically competent Palm domain, this n evertheless
is required for Csm6 function in vivo and also affirm that Csm6 activa- hints that the second messenger-mediated activation mechanism
tion relies on a diffusible ligand rather than a direct physical interaction is nearly universally conserved in type III-A and III-B CRISPR-Cas
with the type III interference complex. systems. Moreover, other CRISPR-associated CARF domain proteins,
ED

We then proceeded to test whether the function of the endogenous notably Csa313,34,35, appear to be transcription factors rather than
Csm6 protein is dependent on the enzymatic activity of the Palm nucleases. This raises the intriguing possibility that CRISPR-Cas
domain of Cas10. To this end, we infected staphylococci containing systems also use cyclic oligoA signalling for transcriptional regula-
the S. epidermidis system programmed to target the late-expressed gene tion across different CRISPR-Cas loci within the same organism, or to
AT

gp43 with phage NM16. Whereas the activity of either Csm3 or induce other genome defence or stress response pathways upon invader
Csm6 is sufficient to support immunity at low multiplicity of infection detection.
(MOI ~1)16, at high MOI (~30) mutation of the Csm6 HEPN RNase
domain (dSeCsm6HEPN) resulted in a significant reduction of cell Online Content Methods, along with any additional Extended Data display items and
Source Data, are available in the online version of the paper; references unique to
ER

survival, even in the presence of active Csm3 (Fig. 5b). Mutation of the these sections appear only in the online paper.
GGDD motif in the Cas10 subunit (dSeCas10Palm) resulted in similar
reduction in cell survival as that observed for the dSeCsm6HEPN mutant received 20 June; accepted 10 July 2017.
(Fig. 5b). It was previously reported that this mutation in S. e pidermidis Published online 19 July 2017.
Cas10 prevents DNA cleavage in vitro6, and therefore the defect in
EL

immunity could be attributed to a functional role of the Palm domain 1. Mohanraju, P. et al. Diverse evolutionary roots and mechanistic variations of
the CRISPR-Cas systems. Science 353, aad5147 (2016).
in Csm6 activation, DNA cleavage or both. To distinguish between 2. Marraffini, L. A. CRISPR-Cas immunity in prokaryotes. Nature 526, 5561
these possibilities we additionally tested a dSeCsm6HEPN/dSeCas10Palm (2015).
double mutant strain, reasoning that the combined effect of the muta- 3. Sorek, R., Lawrence, C. M. & Wiedenheft, B. CRISPR-mediated adaptive
C

immune systems in bacteria and archaea. Annu Rev Biochem 82, 237266
tions would not be additive if immunity is a consequence of Cas10- (2013).
dependent Csm6 activation. Indeed, the double mutant displayed a cell 4. Koonin, E. V., Makarova, K. S. & Zhang, F. Diversity, classification and evolution
AC

survival profile similar to each of the individual mutants, indicating that of CRISPR-Cas systems. Curr Opin Microbiol 37, 6778 (2017).
5. Makarova, K. S. et al. An updated evolutionary classification of CRISPRCas
Cas10 Palm domain and Csm6 RNase activity are genetically linked. systems. Nat Rev Microbiol (2015). doi:10.1038/nrmicro3569
Altogether, these results demonstrate that the in vivo activity of Csm6 6. Samai, P. et al. Co-transcriptional DNA and RNA Cleavage during Type III
is dependent on the cyclic oligoadenylate synthetase activity of the type CRISPR-Cas Immunity. Cell 161, 11641174 (2015).
III interference complex. 7. Goldberg, G. W., Jiang, W., Bikard, D. & Marraffini, L. A. Conditional tolerance of
temperate phages via transcription-dependent CRISPR-Cas targeting. Nature
514, 633637 (2014).
Discussion 8. Deng, L., Garrett, R. A., Shah, S. A., Peng, X. & She, Q. A novel interference
Previous studies demonstrated that Csm6 family ribonucleases provide mechanism by a type IIIB CRISPR-Cmr module in Sulfolobus. Mol. Microbiol.
87, 10881099 (2013).
an additional interference mechanism in type III CRISPR-Cas systems 9. Elmore, J. R. et al. Bipartite recognition of target RNAs activates DNA cleavage
by targeting invader transcripts16. Here we show that Csm6 proteins by the Type III-B CRISPR-Cas system. Genes Dev 30, 447459 (2016).

0 0 M onth 2 0 1 7 | V O L 0 0 0 | N A T U R E | 3
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

10. Estrella, M. A., Kuo, F.-T. & Bailey, S. RNA-activated DNA cleavage by the Type 34. He, F., Vestergaard, G., Peng, W., She, Q. & Peng, X. CRISPR-Cas type I-A
III-B CRISPR-Cas effector complex. Genes Dev 30, 460470 (2016). Cascade complex couples viral infection surveillance to host transcriptional
11. Kazlauskiene, M., Tamulaitis, G., Kostiuk, G., Venclovas, C. & Siksnys, V. regulation in the dependence of Csa3b. Nucleic Acids Res 45, 19021913
Spatiotemporal Control of Type III-A CRISPR-Cas Immunity: Coupling DNA (2017).
Degradation with the Target RNA Recognition. Mol Cell 62, 295306 (2016). 35. Lintner, N. G. et al. The structure of the CRISPR-associated protein Csa3
12. Tamulaitis, G., Venclovas, C. & Siksnys, V. Type III CRISPR-Cas Immunity: Major provides insight into the regulation of the CRISPR/Cas system. J Mol Biol 405,
Differences Brushed Aside. Trends Microbiol. 25, 4961 (2017). 939955 (2011).
13. Makarova, K. S., Anantharaman, V., Grishin, N. V., Koonin, E. V. & Aravind, L. 36. Geertsma, E. R. FX cloning: a simple and robust high-throughput cloning
CARF and WYL domains: ligand-binding regulators of prokaryotic defense method for protein expression. Methods Mol. Biol. 1116, 153164 (2014).

W
systems. Front Genet 5, 102 (2014). 37. Kreiswirth, B. N. et al. The toxic shock syndrome exotoxin structural gene is not
14. Anantharaman, V., Makarova, K. S., Burroughs, A. M., Koonin, E. V. & Aravind, L. detectably transmitted by a prophage. Nature 305, 709712 (1983).
Comprehensive analysis of the HEPN superfamily: identification of novel roles 38. Krissinel, E. & Henrick, K. Secondary-structure matching (SSM), a new tool for
in intra-genomic conflicts, defense, pathogenesis and RNA processing. Biol fast protein structure alignment in three dimensions. Acta Crystallogr D Biol

IE
Direct 8, 15 (2013). Crystallogr 60, 22562268 (2004).
15. Hatoum-Aslan, A., Maniv, I., Samai, P. & Marraffini, L. A. Genetic characterization 39. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple
of antiplasmid immunity through a type III-A CRISPR-Cas system. J. Bacteriol. sequence alignments using Clustal Omega. Molecular Systems Biology 7,
196, 310317 (2014). 539539 (2011).

EV
16. Jiang, W., Samai, P. & Marraffini, L. A. Degradation of Phage Transcripts by 40. Gouet, P., Courcelle, E., Stuart, D. I. & Mtoz, F. ESPript: analysis of multiple
CRISPR-Associated RNases Enables Type III CRISPR-Cas Immunity. Cell 164, sequence alignments in PostScript. Bioinformatics 15, 305308
710721 (2016). (1999).
17. Niewoehner, O. & Jinek, M. Structural basis for the endoribonuclease activity of 41. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. E. The
the type III-A CRISPR-associated protein Csm6. RNA 22, 318329 (2016). Phyre2 web portal for protein modeling, prediction and analysis. Nat Protoc

PR
18. Sheppard, N. F., Glover, C. V. C., Terns, R. M. & Terns, M. P. The CRISPR- 10, 845858 (2015).
associated Csx1 protein of Pyrococcus furiosus is an adenosine-specific
endoribonuclease. RNA 22, 216224 (2016). Supplementary Information is available in the online version of the paper.
19. Staals, R. H. J. et al. RNA targeting by the type III-A CRISPR-Cas Csm complex
of Thermus thermophilus. Mol Cell 56, 518530 (2014). Acknowledgements We thank members of the Jinek and Marraffini laboratories
20. Tamulaitis, G. et al. Programmable RNA shredding by the type III-A CRISPR-Cas for helpful discussions and critical comments on the manuscript. We thank
system of Streptococcus thermophilus. Mol Cell 56, 506517 (2014). Daan Swarts for technical assistance and sharing reagents. We thank Undine

LE
21. Hatoum-Aslan, A., Samai, P., Maniv, I., Jiang, W. & Marraffini, L. A. A Ruler Protein Manzau for technical assistance during A4>P synthesis and purification. This
in a Complex for Antiviral Defense Determines the Length of Small Interfering study was supported by a Swiss National Science Foundation project grant
CRISPR RNAs. Journal of Biological Chemistry 288, 2788827897 (2013). to M.J. (SNSF 31003A_149393) and by funding from the Swiss National
22. Burroughs, A. M., Zhang, D., Schffer, D. E., Iyer, L. M. & Aravind, L. Comparative Competence Center for Research (NCCR) RNA & Disease (to M.J. and J.H.).
genomic analyses reveal a vast, novel network of nucleotide-centric systems M.J. is International Research Scholar of the Howard Hughes Medical Institute
in biological conflicts, immunity and signaling. Nucleic Acids Res 43, and Vallee Scholar of the Bert L & N Kuggie Vallee Foundation. C.G.-D. was
1063310654 (2015).
23. Yan, X., Guo, W. & Yuan, Y. A. Crystal structures of CRISPR-associated Csx3
reveal a manganese-dependent deadenylation exoribonuclease. RNA biology
C
supported by a Long-Term Fellowship from the European Molecular Biology
Organization (EMBO). J.T.R. was supported by a Boehringer Ingelheim Fonds
PhD fellowship. L.A.M. is supported by the Rita Allen Scholars Program, a
TI
12, 749760 (2015). Burroughs Wellcome Fund PATH award, an NIH Directors New Innovator Award
24. Topuzlu, E. & Lawrence, C. M. Recognition of a pseudo-symmetric RNA (1DP2AI104556-01) and a HHMI-Simons Faculty Scholar Award.
tetranucleotide by Csx3, a new member of the CRISPR associated Rossmann
AR

fold superfamily. RNA biology 13, 254257 (2016). Author Contributions O.N, C.G-D. and M.J. conceived the study. O.N., C.G.-D.,
25. Carroll, S. S. et al. Activation of RNase L by 2,5-oligoadenylates. Kinetic J.T.R., L.M. and M.J. designed experiments. O.N. expressed and purified
characterization. J Biol Chem 272, 1919319198 (1997). recombinant Csm6 proteins, carried out oligoA activation and ATPase assays
26. Prischi, F., Nowak, P. R., Carrara, M. & Ali, M. M. U. Phosphoregulation of Ire1 and performed enzymatic probing of the cyclic oligoA product. C.G.-D.
RNase splicing activity. Nat Commun 5, 3554 (2014). expressed and purified recombinant EiCsm(1-5) complexes, performed
27. Staals, R. H. J. et al. Structure and Activity of the RNA-Targeting Type III-B oligoA activation assays and assisted with LC-MS analysis. J.T.R. performed
CRISPR-Cas Complex of Thermus thermophilus. Mol Cell 52, 135145 (2013). phage infection assays under supervision of L.M. C.B. synthesized 2,3-cyclic
28. Hale, C. R., Cocozaki, A., Li, H., Terns, R. M. & Terns, M. P. Target RNA capture phosphate-terminated nucleotides and carried out LC-MS analysis under
ED

and cleavage by the Cmr type III-B CRISPR-Cas effector complex. Genes Dev supervision of J.H. F.S. synthesized 2,3-cyclic phosphate-terminated A4
28, 24322443 (2014). nucleotide and advised on nucleotide chemistry. L.B. performed additional
29. Makarova, K. S. et al. Evolution and classification of the CRISPR-Cas systems. LC-MS analyses of Csm6 activators. O.N., C.G.-D. and M.J. wrote the manuscript,
Nat Rev Microbiol 9, 467477 (2011). with input from the remaining authors.
30. Osawa, T., Inanaga, H. & Numata, T. Crystal Structure of the Cmr2-Cmr3
AT

Subcomplex in the CRISPR-Cas RNA Silencing Effector Complex. J Mol Biol Author Information Reprints and permissions information is available at
425, 38113823 (2013). www.nature.com/reprints. The authors declare competing financial interests:
31. Kalia, D. et al. Nucleotide, c-di-GMP, c-di-AMP, cGMP, cAMP, (p)ppGpp signaling details are available in the online version of the paper. Readers are welcome to
in bacteria and implications in pathogenesis. Chem Soc Rev 42, 305341 comment on the online version of the paper. Publishers note: Springer Nature
(2013). remains neutral with regard to jurisdictional claims in published maps and
32. Hornung, V., Hartmann, R., Ablasser, A. & Hopfner, K.-P. OAS proteins and institutional affiliations. Correspondence and requests for materials should be
ER

cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. addressed to M.J. (jinek@bioc.uzh.ch).
Nat. Rev. Immunol. 14, 521528 (2014).
33. East-Seletsky, A. et al. Two distinct RNase activities of CRISPR-C2c2 enable Reviewer Information Naturethanks S. Bailey, P. Kranzusch and R. Stalls for
guide-RNA processing and RNA detection. Nature 538, 270273 (2016). their contribution to the peer review of this work.
EL
C
AC

4 | N A T U R E | V O L 0 0 0 | 0 0 month 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

D E 4XHQFKHU
51$
OLQNHU
)OXRURSKRUH

7W&VP 7W&VP 7W&VP &VP


Q0 Q0 Q0
4XHQFKHU )OXRURSKRUH

$ 2+
$2+ $!3

!3
Q0 Q0


$
 7W&VP
7LPH PLQ                Q0

W
VV51$ 

)OXRUHVFHQFH DX
VXEVWUDWH
 Q0
$SR


IE
$2+
SURGXFWV  $3

 $!3

EV

   

F 7W&VP G
7LPH VHFRQGV
(L&VP
Q0 S0

PR




)OXRUHVFHQFH DX

)OXRUHVFHQFH DX






LE
 



       

C
7LPH VHFRQGV 7LPH VHFRQGV

$2+ $2+ $!3 $2+ $2+ $!3


TI
Q0 $2+ $!3 $!3 Q0 $2+ $!3 $!3

$2+ $!3 $SR $2+ $!3 $SR

Figure 1 | Csm6 is allosterically activated by oligoA nucleotides. of tetraadenylates containing 3-OH, 3-phosphate or 2,3-cyclic phosphate
AR

a, TtCsm6 ribonuclease activity assay using a Cy5-labelled ssRNA groups. c, d, TtCsm6 and EiCsm6 RNase activities in the presence of
substrate, in the presence of linear tetraadenylates containing 3-OH or oligoadenylates of varying lengths containing 3-OH or 2,3-cyclic
2,3-cyclic phosphate. b, Top: schematic representation of fluorogenic phosphate. All data points represent the mean of three replicates s.e.m.
ribonuclease activity assay; bottom: TtCsm6 RNase activity in the presence
ED
AT
ER
EL
C
AC

0 0 M onth 2 0 1 7 | V O L 0 0 0 | N A T U R E | 5
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

D E 
(L&VP
S0
(L&VP
:7 :7 G&VP&$5) G&VP&$5) Q0

)OXRUHVFHQFH DX

$!3 $!3 Q0 

7LPH PLQ             

W
VV51$ 
VXEVWUDWH

IE

   
7LPH VHFRQGV

SURGXFWV &VP:7$!3
 &VPDSR

EV
Q0 G&VP+(31$!3 G&VP&$5)DSR
G&VP&$5)$!3 G&VP+(31DSR

Figure 2 | The CARF domain mediates allosteric activation of Csm6. of A6>P. b, Ribonuclease activity assay using WT EiCsm6, dEiCsm6HEPN
a, Ribonuclease activity assay using a Cy5-labelled ssRNA and either wild- (R372A/N373A) and dEiCsm6CARF proteins in the presence of A6>P.

PR
type (WT) EiCsm6 or dEiCsm6CARF (Q116A) in the presence or absence All data points represent the mean of three replicates s.e.m.

LE
C
TI
AR
ED
AT
ER
EL
C
AC

6 | N A T U R E | V O L 0 0 0 | 0 0 month 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

D 
(L&VP
(L&VP  FU51$ S0
WDUJHW51$
$73 
0J

)OXRUHVFHQFH DX

$SR

VXSHUQDWDQW $!3>Q0@

W
RQDW& DGGHGWR (L&VP 
USP 
PLQDW& (L&VP VXSHUQDWDQW>@
51DVHUHDFWLRQ 

IE


EV
   
7LPH VHFRQGV
E  (L&VP F  (L&VP
S0 S0



PR
)OXRUHVFHQFH DX
)OXRUHVFHQFH DX








LE


 
   
  
7LPH VHFRQGV

C
7LPH VHFRQGV
(L&VP  >@ $SR

(L&VP  $73>@ (L&VP  :7>@


TI
(L&VP  WDUJHW51$>@ (L&VP  G&VP'$>@
(L&VP  $73WDUJHW51$>@

AR

(L&VP  $73QRQWDUJHW51$>@



(L&VP  $73WDUJHW51$('7$>@
(L&VP  WDUJHW51$873&73*73>@

Figure 3 | The Cas10 complex activates Csm6 via a diffusible second identify components necessary for activator production by the
messenger. a, Left: Schematic representation of EiCsm(1-5) activation EiCsm(1-5). c, EiCsm6 RNase activity in the presence of the product
assay; right: RNase activity of EiCsm6 in the presence of synthetic generated by WT EiCsm(1-5) and EiCsm(1-5)-dCsm3D32A. All data points
ED

A6>P or the product generated by EiCsm(1-5). b, Control assay to represent the mean of three replicates s.e.m.
AT
ER
EL
C
AC

0 0 M onth 2 0 1 7 | V O L 0 0 0 | N A T U R E | 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

D (L&VP E

 ' OP
G&  + P
$
S0

DV  3DO
G DV '

$
VP 3D
G&  '


& 
G& V


VP
D
(L&VP 

7
7
7
7
G&
:
:
:
:
)OXRUHVFHQFH DX
(L&VP  :7>@ WDUJHW51$      

(L&VP  G&VP'$>@ QRQFRJQDWH51$ 
 (L&VP  G&DV3DOP>@  0J        
(L&VP  G&DV3DOPG&VP'$
 >@ 

W

(L&VP  G&DV+'>@
 1RSURGXFWDGGHG

IE

   

F
7LPH VHFRQGV

PLQ
0: 1XFOHDVH6

EV
G

5 $3 3
VW $7

$ P$8

)D 

 LQ
 LQ
LQ
+
BB

 Q
 Q
 LQ
P
P
P
.
.

V
V
L
L
SS

P
P
P
31
31






(L&VP  G&VP'$

PR

   
7LPH PLQ

PLQ
0:

LE

$ P$8

H
  (& Q0
$!3VWDQGDUG &, Q0WRQ0

C
 5 

,QLWDOYHORFLW\ DXV

   
7LPH PLQ
TI


PLQ
0: $73GLPHU 
AR


$ P$8

 (L&VP  



G&DV3DOPG&VP'$    
>F$@ 0

   
ED

7LPH PLQ

Figure 4 | The Palm domain of Cas10 generates cyclic hexaadenylate d, Treatment of the product generated by EiCsm(1-5)-dCsm3D32A by
in vitro. a, EiCsm6 RNase activity in the presence of products generated by T4 PNK, FastAP, pyrophosphatase RppH and S1 nuclease. e, Log(dose)-
WT and mutant EiCsm(1-5). b, ATP oligomerization using [-32P] ATP. versus-response curve and EC50 derived from assays in Extended Data
c, LC-MS analysis of the products generated by EiCsm(1-5)-dCsm3D32A, Fig. 8a. Errors are indicated as 95% confidence interval (CI).
AT

EiCsm(1-5)-dCas10Palm/dCsm3D32A, and a synthetic A6>P standard.


ER
EL
C
AC

8 | N A T U R E | V O L 0 0 0 | 0 0 month 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

D E 
 


2' QP

2' QP






,QIHFWLRQ

W

,QIHFWLRQ

 
 

IE
 
             
7LPH PLQ 7LPH PLQ
 6SDFHU G6H&VP+(31

EV
6H&VP G6H&VP +(31
(L&VP :7
G(L&VP+(31 G(L&VP&$5) G6H&DV3DOP G6H&VP+(31G&DV3DOP

Figure 5 | In vivo activity of Csm6 is dependent on cyclic oligoA b, Growth curves of S. aureus harbouring the S. epidermidis
and Cas10. a, Growth of S. aureus cells containing S. epidermidis type type III-A system with a gp43 spacer and indicated mutations. Infection
III-A (dCsm3D32A/Csm6) system programmed with a gp43 spacer with NM16 is initiated at 60minutes at a MOI of 30. Each data points

PR
supplemented with wild-type or mutated S. epidermidis or E. italicus represents the mean of three replicates s.e.m.
Csm6. Cells were infected at 60minutes with NM16 at a MOI of 0.25.

LE
C
TI
AR
ED
AT
ER
EL
C
AC

0 0 M onth 2 0 1 7 | V O L 0 0 0 | N A T U R E | 9
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

&VP &VP
+'
FU51$
3DOP
&VP &DV
&VP

'1$WHPSODWHRIWDUJHW51$
51DVH &VP

W
WDUJHW51$
'1DVH +'
&DV

IE
F\FOLF
ROLJR$ &DV

EV
V\QWKHWDVH 3DOP $73

FROLJR$

PR
VV51$
&VP

Figure 6 | Proposed model for the molecular mechanism of type activity harboured by the HD domain of Cas10 and (iii) target RNA-
III CRISPR-Cas systems. The type III interference complex has three stimulated cyclic oligoadenylate synthetase activity harboured by the Palm
enzymatic activities: (i) a crRNA-guided RNase activity harboured by the domain of Cas10 to allosterically activate Csm6 RNase.

LE
Csm3 subunits to cleave target RNA, (ii) target RNA-stimulated DNase

C
TI
AR
ED
AT
ER
EL
C
AC

1 0 | N A T U R E | V O L 0 0 0 | 0 0 month 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

Methods c entrifugal filters (Merck Millipore), and samples were analysed by SDS-PAGE
Plasmid DNA constructs. DNA sequences encoding for TtCsm6 (Thermus using a 4-15% gradient polyacrylamide gel (Bio-Rad). All mutant complexes were
thermophilus DSM-579), EiCsm6 (Enterococcus italicus DSM-15952) and MtCsm6 purified using the same protocol as for the wild-type complex. The concentration
(Methanothermobacter thermoautotrophicus DSM-1053) were amplified from their of EiCsm(1-5) complex was calculated according to a formula that takes nucleic
respective genomic DNAs by a standard PCR protocol and were subsequently acid contamination (i.e. crRNA) into consideration.
cloned into a 2HR-T vector (Addgene #29718) using ligation-independent cloning,
(1.55 OD280) (0.75 OD260)
resulting in constructs carrying an N-terminal hexa-histidine tag followed by a

W
StrepII tag, a Tobacco Etch Virus (TEV) protease cleavage site, and the Csm6 Synthetic RNA oligonucleotides. A synthetic target oligonucleotide for the
polypeptide sequence. To obtain a plasmid vector for heterologous expression a EiCsm(1-5) was obtained from IDT with the sequence 5-GGGUAAGGAUAGU
functional EiCsm(1-5) interference complex from the E. italicus DSM-15952 type GAAAUAGAUGCAGUGGAAGCAUACCAAGAGCA-3. A non-cognate control
III-A CRISPR locus, a DNA fragment spanning from the cas10 gene to the end RNA contained the sequence 5-GGUCUAUUGCCCUCUAUAUCGGGCUGUU

IE
of the second spacer (GenBank GL622241.1; complement 1999065-1991480) was CUCCAAACCGGUCUGGGUACCUUCGUACGGACGACCUUC-3.
amplified from E. italicus genomic DNA using primers oCGD208 and oCGD209 Linear 3-hydroxylated oligoA nucleotides were obtained from Dharmacon.
and cloned into a p7XNH3 vector using FX cloning36, resulting in the pCGD253 Linear tetraadenylate carrying a 3-phosphate group (ApApApAp) was obtained

EV
plasmid. In this plasmid, Cas10 (Csm1) was N-terminally fused downstream from Biospring AG, and subsequently converted to A4>P as follows. 2.5ml
of a deca-histidine tag and a human rhinovirus 3C protease cleavage site. Point 2-(N-morpholino)ethanesulfonic acid buffer (MES, 20mM, pH 6) and 1-Ethyl-
mutations in all constructs were inserted by inverse PCR using the primers and 3-(3-dimethylaminopropyl)carbodiimide (EDC, 5.9mg, 38mol, 3.1 eq.) were
templates listed in Supplementary Table 1, and resulting plasmids were verified by added to ApApApAp (12.3mol in 486l water) and the resulting solution was
DNA sequencing. Synthetic DNA and RNA oligonucleotides were obtained from stirred at room temperature. Further portions of 1ml MES-buffer and 1.91mg

PR
Sigma-Aldrich or IDT and are listed in Supplementary Table 2. (1 eq.) EDC were added to the reaction mixture after 24hours and 48hours.
The plasmids containing the wild-type S. epidermidis RP62a CRISPR system HPLC analysis after 55hours indicated ~90% product formation. The reaction
with an anti-gp43 spacer and wild-type SeCsm6 (pWJ191) or inactive dCsm6HEPN mixture was diluted with 10ml water and was extracted with chloroform
(R364A, H369A) mutant (pWJ241), as well as the no-spacer control plasmid (4 x 40ml). The combined organic phases were back-extracted with 10ml water
(pGG-BsaI-R) were previously described 7,16. To construct the Cas10 Palm GGAA and the combined product-containing aqueous phase was filtered (regenerated
(D586A, D587A) mutation (dCas10Palm), the wild-type plasmid was amplified cellulose membrane, 5m) to remove particulate components. The product

LE
by PCR with primer pairs W852/W1169 and W614/W1170, and the resulting solution was diluted with water to ~90ml and applied to a Q Sepharose Fast
fragments were ligated by Gibson assembly. The plasmid carrying the dCsm6HEPN Flow anion exchange column (40165m; 100 x 10mm), previously regenerated
(R364A, H369A) and dCas10Palm double mutation (pJTR147) was generated by with 2M sodium chloride and washed with water. The column was washed with
amplifying PCR products from pWJ241 with primers W614/PS93 and from water, followed by a gradient of 0-2 M NaCl. The title compound eluted with

C
pWJ299 with primers W852/PS94, and them by Gibson assembly. pWJ235 was ~0.20.3 M NaCl. Product-containing fractions were carefully concentrated to
generated by amplifying PCR products from the plasmid pWJ192 with primer a final volume of approximately 10ml. Desalting of A4>P was accomplished by
pairs PS465/W1022 and W494/PS466, and joining them via Gibson assembly. semi-preparative reverse-phase HPLC. The product solution was applied to a
TI
pWJ156, containing SeCsm6, was constructed by a one-piece Gibson reaction YMC*GEL ODS-A 12nm column (10m; 250 x 16mm) pre-equilibrated with
using the PCR product from the amplification of pAS9 with primers W795 and 100% water. The column was washed with water to remove excess NaCl. 50%
W796. The plasmid carrying dCsm6HEPN was generated by amplifying pWJ156 methanol in water was used to elute residual title compound. Product-containing
AR

with primers NP38/PS247 and NP39/PS248, and joining the products by Gibson fractions of appropriate purity were pooled, concentrated with a rotary evaporator
ligation. The plasmids containing wild-type EiCsm6 (pJTR179), dEiCsm6HEPN and subsequently lyophilised in high vacuum for 16 hours to yield 5.8mol A4>P,
(pJTR180), and dEiCsm6CARF (pJTR181) were generated by amplifying DNAs from sodium salt (theoretical yield: 47.2%; purity: 99.96% by HPLC).
plasmids pOP214, pOP215, or pOP218 respectively with primers JTR504/JTR505. All other linear oligoA nucleotides containing 2,3-cyclic phosphate groups
Each resulting fragment was combined by Gibson ligation with the PCR product (A5>P, A6>P, A7>P, A8>P) were synthesized as follows. Phosphoramidites
amplified from pWJ156 with primers JTR502/JTR503. were obtained from Thermo Fisher Scientific (Waltham, MA, USA). 2,3-cyclic
ED

Protein expression and purification. For expression of TtCsm6, MtCsm6, phosphate oligonucleotides were synthesized on a MM12 synthesizer from Bio
EiCsm6, and EiCsm(1-5) complex, the corresponding plasmid was transformed Automation (BioAutomation Corp., Irving, TX, USA) using 5mg 5'-DMT-2',
into E. coli BL21 Rosetta2 (DE3) cells. Cells were grown in LB supplemented 3'-Cyclic Phosphate Adenosine (N-Bz) 1000 CPG (ChemGenes, Wilmington,
with appropriate antibiotics until they reached OD600nm ~0.6 and expression was MA, USA) and 2-TBDMS Adenosine (N-Bz) CED phosphoramidite with a
induced by addition of 0.2mM IPTG (isopropyl--D-thiogalactopyranoside). coupling time of 2 x 90s. Phosphoramidite was prepared as a 0.08M solution
AT

Proteins were expressed at 18C for 16h. For TtCsm6, MtCsm6 and EiCsm6 the in dry acetonitrile (ACN), the activator 5-(Benzylthio)-1H-tetrazole (Biosolve
cells were harvested and resuspended in lysis buffer containing 20mM HEPES BV, Valkenswaard, Netherlands) was prepared as a 0.24M solution in dry ACN.
pH 8.0, 500mM KCl, and 5mM imidazole pH 8.0. The cell suspension was lysed Oxidizer was prepared as a 0.02M I2 solution in THF/pyridine/H2O (70:20:10,
by ultrasonication and lysate was cleared by centrifugation at 20,000 g for 40min. w/v/v). Capping reagent A was THF/lutidine/acetic anhydride (8:1:1) and capping
ER

Cleared lysate was applied to a 5ml Ni-NTA cartridge (Qiagen), the column was reagent B was 16% N-methylimidazole/THF. 3% dichloroacetic acid in dichlo-
washed with 10 column volumes of lysis buffer and bound proteins were eluted romethane was used as deblock solution. 2,3-cyclic phosphate oligonucleotides
with 5 column volumes of the same buffer supplemented with 250mM imidazole were synthesized DMT-off. After completion of the automated synthesis, oligo-
pH 8.0. Eluted proteins were dialysed overnight against 20mM HEPES pH 7.5 nucleotides were cleaved from the solid support and the protecting groups on
and 500mM KCl in the presence of TEV (tobacco etch virus) protease. Dialysed the exocyclic amino groups (N6-benzoyl) and the backbone (2-cyanoethyl) were
EL

proteins were passed through a 5ml Ni-NTA cartridge. The flow-through fraction removed using a 1:1 mixture of 40% aqueous methylamine and 25% aqueous
was concentrated and further purified by size-exclusion chromatography using ammonia for 1 h at 65C. 2-O-tert-butyldimethylsilyl groups (TBDMS) were
an S200 (16/600) column (GE Healthcare) in 20mM HEPES pH 7.5 and 500mM removed and simultaneously the 2,3-cyclic phosphate terminus was formed using
KCl, yielding pure, monodisperse proteins. Purified proteins were concentrated a fresh mixture of N-methyl-2-pyrrolidone (60l), triethylamine (30l) and trieth-
to 5-75mg ml-1 using 30,000 MWCO centrifugal filters (Merck Millipore) and ylamine trihydrofluoride (40l) at 40C for 6h. Desilylation was quenched with
C

flash-frozen in liquid nitrogen. trimethylethoxysilane (200l), and diisopropyl ether (200l) was subsequently
For EiCsm(1-5) complex, cells were harvested and resuspended in lysis buffer added to precipitate the oligonucleotide. The precipitate was dissolved in H2O
AC

containing 20mM HEPES pH 8.0, 400mM KCl, and 5mM imidazole supple- and purified on an Agilent 1200 series preparative HPLC fitted with a Waters
mented with EDTA-free protease inhibitor (Roche) and lysed using an HPL6 cell XBridge Oligonucleotide BEH C18 column (1050mm, 2.5m) at 65C using
homogenizer. Lysate was clarified by centrifugation at 20,000 g for 40min. The a gradient of 5-20% buffer B over 8min and flow rate of 5ml min-1. Buffer A was
lysate was applied to a 5ml Ni-NTA cartridge (Qiagen), the column was washed 0.1M triethylammonium acetate pH 8.0; buffer B was methanol. Fractions were
in two steps with lysis buffer supplemented 10 and 50mM imidazole. The complex pooled, dried under vacuum and dissolved in H2O. The c-A6 product generated
was eluted with elution buffer containing 20mM HEPES pH 7.5, 400mM KCl, and by EiCsm(1-5) was purified using the same protocol.
250mM Imidazole. The complex was concentrated and further directly purified Gel-based Csm6 ribonuclease activity assay. Gel-based cleavage assays were
by size-exclusion chromatography using two Superose-6 (10/300) columns performed as described previously17. Briefly, a 24-nucleotide synthetic RNA
connected in tandem, or a Sephacryl S-300 (26/600) column (GE Healthcare), (250nM), 5-end labelled with Cy5, was incubated with Csm6 in the presence of
in buffer containing 20mM HEPES pH 7.5, 300mM KCl, and 5mM MgCl2. indicated concentrations of oligonucleotide activators at 37oC. At indicated time
Appropriate fractions were pooled and concentrated using 100,000 MWCO points, the reaction was quenched by addition of an equal volume of quencher

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

s olution (95% formamide, 5% glycerol and 0.005% bromophenol blue) and 1l 10x buffer and 6.5l H2O. Reactions were incubated at 37C for 90min and
products were resolved on a 16% denaturing (7M urea) polyacrylamide gel and products were resolved and imaged as above.
visualised using a fluorescence gel scanner (Typhoon FLA 9500, GE Healthcare). LC-MS analysis. To analyse the product generated by EiCsm(1-5) complex, the
Protein concentrations are indicated in the respective figure panels. EiCsm(1-5)-dCsm3D32A complex was used to prevent cleavage of the target RNA
Fluorescence-based Csm6 ribonuclease activation assay. To obtain kinetics by the Csm3 subunits and ensure persistent activation of the complex. A reaction
of Csm6 ribonuclease activity, 2l of 2M RNase Alert substrate (IDT) and 1l containing 0.15mg ml-1 of EiCsm(1-5) complex (500nM), 10M target RNA,
of synthetic oligoA or supernatant from a EiCsm(1-5) cyclic oligoadenylate 1mM MgCl2 and 0.5mM ATP in buffer containing 20mM HEPES pH 7.5 and

W
synthetase reaction were added to a well in a 96-well plate. The reaction was 50mM KCl was incubated for 30 minutes at 37C. To test the incorporation of
started by addition of 97l Csm6 protein in 20mM HEPES pH 7.5 and 50mM KCl. other ribonucleotides, the reaction was additionally supplemented with 0.5mM
Fluorescence signal (excitation at 490nm, emission at 520nm) was measured over of GTP, CTP and UTP. Reactions were stopped by heating to 95C for 10min, the
time in TECAN Infinite M1000 or PHERAstar FSX (BMG Labtech) multimodal denatured protein was removed by centrifugation and 1l of the deproteinized

IE
plate readers. Final protein and ligand concentrations used in the assay reactions supernatant was diluted in 30l of H2O and subjected to LC-MS. Analysis of
are indicated in corresponding figure panels. purified oligonucleotides and enzymatic assays was conducted on an Agilent
Calculation of EC50. Cleavage kinetics for reactions containing EiCsm6 sup- 1200/6130 LC-MS system fitted with a Waters Acquity UPLC OST C18 column

EV
plemented with indicated concentrations of A6>P or c-A6 were measured and (2.150 mm, 1.7 m) at 65C, with a gradient of 5-20% buffer B in 10min with
initial velocities were calculated over the first 10s of the reaction. Initial velocities a flowrate of 0.3mlmin1. Buffer A was aqueous hexafluoroisopropanol (0.4M)
were then plotted against ligand concentrations in GraphPad and fitted by using supplemented with triethylamine (15mM). Buffer B was methanol. The super-
a nonlinear regression analysis using the log(dose)-versus-response relationship, natants obtained from the EiCsm(1-5)-dCsm3D32A complex or the EiCsm(1-5)-
assuming a variable Hill coefficient. dCsm3D32A/dCas10Palm complex were analysed using the same protocol. To analyse

PR
In vitro cyclic oligoadenylate synthetase assays using EiCsm(1-5). To utilise the the S1 nuclease digest of the EiCsm(1-5) product, 15l of the supernatant were
EiCsm(1-5) complex and mutants for in vitro synthesis of cyclic o ligoadenylates, treated with 0.5U l-1 of nuclease S1 enzyme for 30min at 37C. The sample was
0.15mg ml-1 EiCsm(1-5) complex was mixed with 10M target RNA, 1mM MgCl2 mixed with 20l chloroform, vortexed and centrifuged for 5min at 5000 rpm.
and 0.5mM ATP in a buffer containing 20mM HEPES pH 7.5 and 50mM KCl The upper (aqueous) phase was transferred to a fresh tube without disturbing the
in a total volume of 30l. The reaction was incubated at 37C for ~16h and the interphase and analysed on the same system as above with a gradient of 1-10%
proteins were subsequently denatured at 95C for 10min. Denatured protein was buffer B in 10min.

LE
pelleted by centrifugation and the deproteinized supernatant was transferred to RNA cleavage assay by EiCsm(1-5) complexes. The reaction was initiated by
a fresh tube. The supernatant was diluted as indicated and added to the Csm6 adding 250nM target RNA substrate (3-labelled with Alexa 352) to 250nM
ribonuclease activity assay as described above. EiCsm(1-5) complex (wild-type or inactive mutants) in a buffer containing 20mM
To directly visualize the synthesis of cyclic oligodenylates, the same reaction HEPES pH 7.5 and 50mM KCl. After 1 minute at 37C, the reaction was stopped

C
was supplemented with 1Ci of [-32P]-ATP (Hartmann Analytic GmbH) and by addition of equal volume of quencher solution (90% formamide, 5% glycerol,
the reaction time was shortened to 30min. The reaction was stopped by heat- 25mM EDTA) and heated at 95C for 5min. 10% of the final reaction was resolved
inactivation at 95C for 10min. The products were mixed with equal volumes of on a 16% denaturing (7M urea) polyacrylamide gel. The products were visualized
TI
quencher solution (95% formamide, 5% glycerol and 0.005% bromophenol blue) using a Typhoon FLA 9500 fluorescence gel scanner.
and subsequently resolved on a 20% denaturing (7M urea) polyacrylamide gel run Phage infection assays. S. aureus RN422037 was grown in tryptic soy broth
at 60 W for ~5h. The gel was dried and products were detected by phosphorim- (TSB) medium at 37C, and supplied with 10g ml-1 chloramphenicol or
AR

aging using a Typhoon FLA 9500 imager (GE). 10g ml-1 erythromycin for plasmid maintenance. During bacteriophage
For time-course experiments, 0.05mg ml-1 EiCsm(1-5) complex was mixed with infection, 5mM CaCl2 was added to the media. For both EiCsm6 complementa-
10M target RNA, 1mM MgCl2 and 0.5mM ATP in a buffer containing 20mM tion and Csm6-Cas10 phenocopy experiments, overnight cultures initiated from
HEPES pH 7.5 and 50mM KCl in a total volume of 30l. 3 l aliquots were taken at single colonies were diluted 1:20 in TSB broth. After growth for 1 h, cell d
ensities
indicated time points and reactions were quenched by addition into 7l quencher were normalised and cultures were added to a 96-microwell plate (Cellstar,
solution. Samples were resolved and imaged as above. 655180) in the presence of appropriate antibiotics and 5mM CaCl2. The plate
ED

Nuclease S1 digest of Ei(Csm1-5) product. The radiolabelled product from was shaken in a microplate reader (TECAN Infinite 200 PRO) at 37C, with
the EiCsm(1-5) assay was digested with Nuclease S1 (Thermo Fisher Scientific). optical measurements taken at 600nm every 10 minutes. Bacteriophage N M16 7
Cleavage reactions contained 4l 5x Nuclease S1 buffer, 15l of [-32P] ATP- was added at an MOI of approximately 0.25 or 30 after 60min of shaking. For the
labelled oligoadenylates and 1l of Nuclease S1 diluted 1:50 in its commercial complementation experiment, cells containing plasmid pWJ235 (gp43 spacer,
protein storage buffer (final concentration 0.1U l-1) in a total volume of 20l. dCsm3, Csm6) in combination with one of the following plasmids; pWJ156,
AT

The reaction was incubated at 37C and 2l aliquots were taken at indicated time pJTR67, pJTR179, pJTR180, or pJTR181, were inoculated from single colonies
points. Reactions were quenched by addition into 10 volumes quencher solution and grown in TSB supplemented with 10g ml-1 chloramphenicol and 10g ml-1
and subsequently resolved and imaged as described above. erythromycin. For the phenocopy experiment, overnight cultures of S. aureus
Enzymatic probing of Ei(Csm1-5) product. Aliquots of the radiolabelled prod- RN4220 containing pWJ191, pWJ241, pWJ299, pJTR147, or pGG-BsaI-R were
uct from the EiCsm(1-5) assay were treated with PNK (NEB) in the absence and inoculated from single colonies and grown in in TSB supplemented with 10g ml-1
ER

presence of 1mM ATP, with Fast Alkaline Phosphatase (Thermo Fisher Scientific) chloramphenicol.
and E. coli RNA 5' Pyrophosphohydrolase (RppH, purified in-house). All r eactions Data availability. Raw images of uncropped gels shown in in Fig. 1a, Fig. 2a,
were performed in reaction buffers optimised for the respective enzymes as Fig. 4b, Fig. 4d and Extended Data Figs. 1, 2 and 5 are provided in Supplementary
recommended by the supplier. All reactions were performed in total volumes Figure 1. Other data are available from the corresponding author upon reasonable
of 10l containing 1l enzyme, 1.5l [-32P] ATP-labelled oligoadenylates,
EL

request.
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

AfCsx3 TtCsm6 Superimposition


(PDB: 3WZI) (PDB: 5FSH)

W
IE
EV
Extended Data Figure 1 | AfCsx3 resembles the CARF domain of TtCsm6. Left: Structure of AfCsx3 (PDB: 3WZI) bound to a tetraribonucleotide.

PR
Center: Structure of the CARF domain dimer of TtCsm6 (PDB: 5FSH). Right: Superimposition of AfCsx3 (grey) and TtCsm6 (pink and blue). The
structures were aligned using PDBeFold38.

LE
C
TI
AR
ED
AT
ER
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

a b

F
AR
iC 6 H EP

T
dE m6 T
dE sm T

W
C
s W
iC W

M m6

6
TtCsm6

Ei m6

sm
s
s
50 nM kDa

tC
C
C
Tt
150
5 M 250
apo 130

W
A3-OH 100
100 A4-OH
Fluorescence (a.u.)

70
A5-OH

IE
A6-OH 55
A7-OH
50 A8-OH
A9-OH 35

EV
A10-OH
25
0
0 250 500 750 1000
Time (seconds)

PR
15

c d

LE
MtCsm6 EiCsm6
150 4 nM 150 20 nM
5 M 5 M
apo apo

C
A3-OH A3-OH
Fluorescence (a.u.)

Fluorescence (a.u.)

100 A4-OH 100 A4-OH


A5-OH A5-OH
TI
A6-OH A6-OH
A7-OH A7-OH
50 A8-OH 50 A8-OH
AR

A9-OH A9-OH
A10-OH A10-OH

0 0
0 250 500 750 1000 0 250 500 750 1000
Time (seconds) Time (seconds)
ED

e f
MtCsm6
300 (1.5 nM) 125

250
AT

100
Fluorescence (a.u.)
Fluorescence (a.u.)

200
75

150
ER

50
100

25
50
EL

0
0
0 250 500 750
0 250 500 750 Time (seconds)
Time (seconds)
A4-OH A7-OH A6>P
20 nM TtCsm6 / 500 nM A4>P 200 pM EiCsm6 / 500 nM A4>P
C

250 nM A5-OH A4>P A7>P 20 nM TtCsm6 / 500 nM A6>P 200 pM EiCsm6 / 500 nM A6>P

A6-OH A5>P Apo 20 nM TtCsm6 200 pM EiCsm6


AC

Extended Data Figure 2 | Activation of Csm6 ribonucleases by EiCsm6 ribonuclease activity assays in the presence of 3-hydroxylated
oligoadenylates. a, TtCsm6 ribonuclease activity assay in the presence oligoadenylates ranging from A3 to A10. e, MtCsm6 ribonuclease activity
of 3-hydroxylated oligoadenylates ranging from A3 to A10. b, SDS- assay in the presence of oligoadenylates carrying 3-hydroxyl or 2,3-cyclic
PAGE analysis of purified WT and mutant Csm6 proteins from Thermus phosphate groups. f, TtCsm6 and EiCsm6 ribonuclease activity assay in
thermophilus (TtCsm6), Methanothermobacter thermoautotrophicus the presence of A4>P and A6>P. All data points represent the mean of
(MtCsm) and Enterococcus italicus (EiCsm6). c,d, MtCsm6, and three replicates s.e.m.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

a EiCsm6 b
(400 pM)
5

375 EC50 = 63 nM
4 95% CI = 53 nM to 75 nM
R2 = 0.9818
300

Inital velocity (a.u./s)


Fluorescence (a.u.)

W
3
225

IE
150

1
75

EV
0
0 10-9 10-8 10-7 10-6 10-5
0 250 500 750
Time (seconds) [A6>P] (M)
1,000 nMA6>P 50 nM A6>P 2.5 nM A6>P

PR
500 nM A6>P 25 nM A6>P Apo

250 nM A6>P 10 nM A6>P

100 nM A6>P 5 nM A6>P

Extended Data Figure 3 | Activation of EiCsm6 by A6>P. a, EiCsm6 RNase assay in the presence of varying concentrations of A6>P. b, Log(dose)-
versus-response curve and EC50 derived from assays in a. Errors are indicated as 95% confidence interval (CI). All data points represent the mean of

LE
three replicates s.e.m.

C
TI
AR
ED
AT
ER
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

a
*
E. italicus Csm6

W
IE
EV
PR
b

LE
Q116
T114
M117

T111 S112
E118

S112 E118 T111


C
TI
M117
T114
AR

Q116

Extended Data Figure 4 | Conservation and structure of the CARF using Clustal Omega39 and visualized using ESPript 340. Green asterisk
domain motif. a, Multiple sequence alignment of the CARF domain denotes the conserved glutamine residue mutated to abrogate allosteric
motifs in Csm6. The polypeptide sequence of EiCsm6 (denoted with an activation of EiCsm6. b, Zoom-in view of a structural model of the
arrow) was used for a BLAST search against the NCBI non-redundant dimeric interface of the CARF domains in the EiCsm6 dimer. The model
protein sequences database and 19 best scoring hits with >95% query was generated using Phyre241 based on the TtCsm6 crystal structure
ED

coverage were selected. The multiple sequence alignment was performed (PDB: 5FSH).
AT
ER
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

a E. italicus DSM 15952

cas1 cas2 cas10 csm2 csm3 csm4 csm5 csm6 cas6

W
csm4 csm5
csm3 csm6

IE
csm2
cas6
R-S1-R-S2

EV
cas10

6xHis
T7 prom KanR

PR
b c
lm
dC 3 D32A

lm
2A
dC 0 HD

Pa

D
Pa
3 D3

H
LE
10

10

10
l

l
1

tro

tro
sm

sm
as

as

as

as
W

on

on
T

T
dC

dC

dC

dC
kDa
W
M

W
C

C
250 target ssRNA

130

C
TI
100 Csm1
Csm1 proteolytic
70
degradation product
55
cleavage
AR

Csm5 products

35 Csm4
Csm3
25

15 Csm2
ED

10

Extended Data Figure 5 | Purification and crRNA-guided RNA cleavage repeat-spacer units in the CRISPR array was cloned into the expression
by the EiCsm(1-5) complex. a, Enterococcus italicus DSM15952 contains plasmid. b, SDS-PAGE analysis of purified wild-type and mutant
EiCsm(1-5) complexes. c, Denaturing gel of the products of a target
AT

a CRISPR-Cas locus with 9 cas genes followed by a repeat-spacer array.


For heterologous expression of the EiCsm(1-5) complex, the genomic RNase cleavage assay performed using a fluorophore-labelled target
DNA fragment spanning genes csm1-csm6 and cas6 and the first two ssRNA substrate in the presence of WT or mutant EiCsm(1-5) complexes.
ER
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

a
EiCsm(1-5)dCsm3D32A + target RNA

in

in
in

in

in

m
s

m
15

30

10

30
0

5
c-A6

W
[-32P] ATP

IE
EV
b
EiCsm(1-5)-dCsm3D32A peak at 4.084 min

100

986.2

PR
80

60
657.2

40

1974.0

LE
20

C
1000 2000 m/z
TI
A6>P standard peak at 3.771 min

100
986.2

AR

80

60
657.2

40
ED

1973.8

20

0
AT

1000 2000 m/z

EiCsm(1-5)-
dCas10Palm/dCsm3D32A peak at 0.798 min
ER

100
505.9

80
EL

60
766.0

40
528.0

1013.0
C

20

0
AC

1000 2000 m/z


Extended Data Figure 6 | The EiCsm(1-5) complex generates cyclic generated by the EiCsm(1-5)-dCsm3D32A and EiCsm(1-5)-dCas10Palm/
hexaadenylate. a, Denaturing 20% polyacrylamide gel indicating time- dCsm3D32A complexes shown in Fig. 4c. The mass spectrum of synthetic
resolved [-32P]-ATP oligomerization by the EiCsm(1-5) complex. A6>P standard is shown for comparison.
b, Mass spectra of LC-MS peaks from the analysis of the products

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

a b Purified oligoadenylate
2 00 peak at 3.930min

Purified EiCsm(1-5)-dCsm3D32A product 100

986.2
1 50
80
3.930 min

W
MW: 1973.9 60
1 00

657.2
40

1973.9
50

IE
20

0
0

EV
1000 2000
1 2 3 4 5
-50
c d Purified A6>P peak at 3.738 min

PR
3.738 min
2 00 MW: 1974.1 100

986.2
A6>P standard
80
1 50
60

LE 657.2
40
1 00

1974.1
20

50

C
0
1000 2000
TI
0
1 2 3 4 5
AR

-50 peak at 3.775 min


100
986.2
e Purified EiCsm(1-5)-dCsm3D32A product f
80
2 00 spiked with A6>P standard
60
3.775 min
657.2
ED

MW: 1973.8
1 50 40

20 1974.1
3.922 min
1 00 MW: 1973.8
0
AT

1000 2000
50

0
ER

1 2 3 4 5 peak at 3.922 min


100
986.2

-50
80
EL

60
657.2

40
1973.8
C

20

0
AC

1000 2000

Extended Data Figure 7 | LC-MS analysis of purified EiCsm(1-5) purified product spiked with the A6>P standard. b, d, f, Mass spectra of
product. a, c, e, LC-MS analysis of the HPLC-purified activator product the peaks indicated in panels a, c and e.
generated by the EiCsm(1-5) complex as well as an A6>P standard and

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

a
S1 digestion product peak at 0.80 min
8 00 100

693.1
0.80 min
MW: 697.11 (AMP dimer) MW A (267) or pA>P (409)
80
A260 (mAU)

not present
6 00

715.1
60

W
4 00
S1 digestion product

1062.1
40

584.1
730.9
2 00

IE
20
0
0
2 4 6 8

EV
Time (min) 1000 2000

b c
EiCsm6

PR
(150 pM)
EC50 = 3.5 nM
50
150 95% CI = 3.32 nM to 3.74 nM
R2 = 0.9957
125 40

Inital velocity (a.u./s)


100
Fluorescence (a.u.)

LE
30
A6>P
75
c-A6
20
50

C
EC50 = 63 nM
10
25 95% CI = 53 nM to 76 nM
R2 = 0.9818
TI
0 0
0 250 500 750 10-10 10-9 10-8 10-7 10-6 10-5
[A6>P] (M)
25 nM cA6 1 nM cA6 Apo
AR

10 nM cA6 0.5 nM cA6

5 nM cA6 0.25 nM cA6

2.5 nM cA6 0.1 nM cA6

Extended Data Figure 8 | The EiCsm(1-5) complex generates cyclic of c-A6. c, Overlay of EC50 values obtained for A6>P and c-A6. Errors are
ED

hexaadenylate. a, Left: LC-MS analysis of the nuclease S1 digested indicated as 95% confidence interval (CI) and data points represent mean
activator. Right: Mass spectrum profile of the peak obtained after S1 of three replicates s.e.m.
digest. b, EiCsm6 RNase assay in the presence of varying concentrations
AT
ER
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Article RESEARCH

a
EiCsm6 EiCsm6
(200 pM) (200 pM)
175 3.0

150

2.5
125
Fluorescence (a.u.)

Fluorescence (a.u.)

W
100
2.0
75

50
1.5

IE
25

0 1.0
0 250 500 750 0 250 500 750

EV
Time (seconds)
A A/C/U/G G/C G G/C

A/G G G/U C G/U

A/C C C/U/G U C/U/G

A/U U

PR
b
2 00
100

986.2
EiCsm(1-5)-dCsm3D32A
1 50 + ATP/UTP/GTP/CTP 80 peak at 3.905 min
A260 (mAU)

LE
657.2
3.905 min 60
1 00
MW: 1,974.74

1973.9
50 40

612.2

C
20
0
2 4 6 8 0
TI
Time (min) 1000 2000
AR

EiCsm(1-5)-dCsm3D32A 100
1 00
ATP
986.7

3.933 min
MW: 1,974.55 80
peak at 3.933 min
ED
A260 (mAU)

60
50
40
657.2

1974.3

20
AT

0 0
2 4 6 8
1000 2000
Time (min)
Extended Data Figure 9 | The EiCsm(1-5) complex selectively samples from panel a. The elution profile (left) and mass spectrum (right)
ER

incorporates adenosines into the activator. a, EiCsm6 RNase activity of the product remains unaltered upon addition of all four ribonucleotide
in the presence of products generated by EiCsm(1-5) under addition of triphosphates, indicating ATP selectivity of the EiCsm(1-5) complex.
various ribonucleotide combinations. b, LC-MS analysis of indicated
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Article

O O O NH2

- - N
O P O P O P O N O O O NH2

- N - - N
-
O O O N O P O P O P O N
O
- - N
O O O N
O

OH OH

W
O O O NH2 O OH NH2

N N

ATP
- - -
O P O P O P O N O P O N

IE
- - N
PPi N
O O O N O N
O O

EV
OH OH OH OH

ATP
PPi

PR
ATP
PPi

LE
O O O NH2

H2N

- - N
O P O P O P O N N
NH2
N
N

C
- - N N
O O O N
N

PPi
OH N
O N

O N
O
TI O O
P OH

O OH NH2 -
O O
O
O

- N HO P O -
O P O N O P
AR

O -
N O O
O
N
N N
N
O N H2N
O O
O
NH2
N N
N

n
O -
N
O O
- O
P O
O P OH

O OH NH2
PP i O
O O
-
O
ED

- N
HO
O P O N P

O O
N O O
N
O N
N
O N HO N
N

N
N
H2N
N
AT

OH OH NH2

Extended Data Figure 10 | Proposed mechanism of cyclic addition of successive AMP moieties to the 3 end to generate a linear
oligoadenylate formation. The Cas10 Palm domain initially catalyses 5-3 oligonucleotide. Final cyclisation involves the 3-hydroxyl group attacking
phosphodiester bond formation by a nucleophilic attack of the 3-hydroxyl the -phosphate group of the 5-triphosphate moiety in the linear
group of one ATP substrate molecule onto the -phosphate group of oligoadenylate intermediate. Notably, all reaction steps utilize the same
ER

a second ATP molecule, yielding a 5-triphosphorylated dinucleotide catalytic mechanism and thus could be carried out by a single active site in
and inorganic pyrophosphate (PPi). The dinucleotide is extended by the Palm domain of Cas10.
EL
C
AC

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
nature research | life sciences reporting summary
Corresponding author(s): Martin Jinek
Initial submission Revised version Final submission

Life Sciences Reporting Summary


Nature Research wishes to improve the reproducibility of the work that we publish. This form is intended for publication with all accepted life

W
science papers and provides structure for consistency and transparency in reporting. Every life science submission will use this form; some list
items might not apply to an individual manuscript, but all fields must be completed for clarity.
For further information on the points included in this form, see Reporting Life Sciences Research. For further information on Nature Research

IE
policies, including our data availability policy, see Authors & Referees and the Editorial Policy Checklist.

EV
` Experimental design
1. Sample size
Describe how sample size was determined. not applicable

PR
2. Data exclusions
Describe any data exclusions. no applicable
3. Replication

LE
Describe whether the experimental findings were Experimental findings in all figures were reliably reproduced.
reliably reproduced.
4. Randomization
Describe how samples/organisms/participants were
allocated into experimental groups.
not applicable

C
TI
5. Blinding
AR

Describe whether the investigators were blinded to not applicable


group allocation during data collection and/or analysis.
Note: all studies involving animals and/or human research participants must disclose whether blinding and randomization were used.

6. Statistical parameters
D

For all figures and tables that use statistical methods, confirm that the following items are present in relevant figure legends (or in the
Methods section if additional space is needed).
E

n/a Confirmed
AT

The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement (animals, litters, cultures, etc.)
A description of how samples were collected, noting whether measurements were taken from distinct samples or whether the same
sample was measured repeatedly
ER

A statement indicating how many times each experiment was replicated


The statistical test(s) used and whether they are one- or two-sided (note: only common tests should be described solely by name; more
complex techniques should be described in the Methods section)
EL

A description of any assumptions or corrections, such as an adjustment for multiple comparisons


The test results (e.g. P values) given as exact values whenever possible and with confidence intervals noted
A clear description of statistics including central tendency (e.g. median, mean) and variation (e.g. standard deviation, interquartile range)
C

Clearly defined error bars


AC

June 2017

See the web collection on statistics for biologists for further resources and guidance.

1
` Software

nature research | life sciences reporting summary


Policy information about availability of computer code
7. Software
Describe the software used to analyze the data in this GraphPad and Microsoft Excel
study.
For manuscripts utilizing custom algorithms or software that are central to the paper but not yet described in the published literature, software must be made
available to editors and reviewers upon request. We strongly encourage code deposition in a community repository (e.g. GitHub). Nature Methods guidance for

W
providing algorithms and software for publication provides further information on this topic.

` Materials and reagents

IE
Policy information about availability of materials
8. Materials availability

EV
Indicate whether there are restrictions on availability of The A4>P was custom-made by an oligonucleotide synthesis company (BIOLOG
unique materials or if these materials are only available GmbH) per one-time order. As we only have limited quantities, we are unable to
for distribution by a for-profit company. distribute large quantities of the material - this would require another custom
synthesis, either by BIOLOG or other oligonucleotide CMOs.

PR
9. Antibodies
Describe the antibodies used and how they were validated not applicable
for use in the system under study (i.e. assay and species).
10. Eukaryotic cell lines

LE
a. State the source of each eukaryotic cell line used. not applicable

b. Describe the method of cell line authentication used. no applicable

c. Report whether the cell lines were tested for


mycoplasma contamination.
not applicable

C
TI
d. If any of the cell lines used are listed in the database not applicable
of commonly misidentified cell lines maintained by
AR

ICLAC, provide a scientific rationale for their use.

` Animals and human research participants


Policy information about studies involving animals; when reporting animal research, follow the ARRIVE guidelines
D

11. Description of research animals


Provide details on animals and/or animal-derived not applicable
E

materials used in the study.


AT

Policy information about studies involving human research participants


12. Description of human research participants
Describe the covariate-relevant population not applicable
ER

characteristics of the human research participants.


EL
C
AC

June 2017

You might also like