post-transcriptional regulation by rnases in streptococcus...

72
Post-transcriptional regulation by RNases in Streptococcus pyogenes Anne-Laure Lécrivain Department of Molecular Biology Umeå Center for Microbial Research (UCMR) Laboratory for Molecular Infection Medicine Sweden (MIMS) Um 2018

Upload: others

Post on 04-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

Post-transcriptional regulation

by RNases

in Streptococcus pyogenes

Anne-Laure Lécrivain

Department of Molecular Biology

Umeå Center for Microbial Research (UCMR)

Laboratory for Molecular Infection Medicine Sweden (MIMS)

Um 2018

Page 2: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

This work is protected by the Swedish Copyright Legislation (Act 1960:729)

Dissertation for PhD

ISBN: 978-91-7601-950-4

ISSN: 0346-6612

New series: 1993

Cover design: Anne-Laure Lécrivain

Electronic version available at: http://umu.diva-portal.org/

Printed by: Print & Media (Umeå University)

Umeå, Sweden 2018

Page 3: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

To everyone who supported me

“C est ce que nous pensons déjà connaître

qui nous empêche souvent d apprendre”

Claude Bernard

(It is what we think we already know that often prevents us from learning)

Page 4: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)
Page 5: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)
Page 6: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

i

Table of Contents

Table of Contents ........................................................................... i

Abstract ....................................................................................... iii

Abbreviations ............................................................................... v

Publications included in this thesis ............................................ vii

I- Introduction ............................................................................... 1

Post-transcriptional regulation of gene expression .................................................. 1

Ribonucleases .............................................................................................................3

RNase Y .................................................................................................................3

RNase III ............................................................................................................... 5

RNase J1 ............................................................................................................... 6

PNPase .................................................................................................................. 7

RNase R ................................................................................................................ 9

YhaM ................................................................................................................... 10

NanoRNases ....................................................................................................... 11

Interplay of RNases .................................................................................................. 11

RNA degradosome and other RNase complexes .............................................. 11

Models of RNA decay ......................................................................................... 13

RNA sequencing........................................................................................................ 14

General principle ................................................................................................ 14

RNA sequencing to identify ribonuclease targets ............................................ 14

Different methods to identify ribonuclease targets.......................................... 15

Streptococcus pyogenes ........................................................................................... 16

A human pathogen ............................................................................................. 16

Post-transcriptional regulation of gene expression in S. pyogenes ................ 18

II- Aims of the thesis .................................................................... 19

III- Results and discussion ......................................................... 20

Paper I ...................................................................................................................... 20

Methodology ...................................................................................................... 20

Characteristics of S. pyogenes RNase III .......................................................... 21

RNase III role in S. pyogenes ............................................................................ 22

Page 7: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

ii

Paper II ..................................................................................................................... 24

Methodology ...................................................................................................... 24

YhaM has a global nibbling activity on transcript 3′ ends ............................ 25

Role and peculiarities of S. pyogenes YhaM .................................................... 26

PNPase role in RNA decay ................................................................................ 28

Limited RNase R activity .................................................................................. 29

Possible redundancy between RNase R and PNPase ...................................... 30

Paper III .................................................................................................................... 31

RNase Y cleaves after a guanosine.................................................................... 31

RNase Y and PNPase act in concert to degrade transcripts .......................... 33

Is RNase Y the major RNase initiating RNA decay?........................................35

The fate of transcript 5′ ends produced by RNase Y ....................................... 36

IV- Main findings of the thesis .................................................... 38

V- Acknowledgements ................................................................ 39

References ................................................................................... 41

Page 8: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

iii

Abstract

Ribonucleases (RNases) are proteins that adjust cellular RNA levels by

processing RNA transcripts, leading to their stabilization or degradation.

RNases are grouped based on their ability to cleave the transcript internally

(endoRNases) or degrade the transcript starting from the ends (exoRNases).

Specificities of RNA degradation vary among bacterial species, attributable to

different sets of endo- and exoRNases. Most of the current knowledge gathered

about the roles of RNases and their targets relies on the study of a few model

bacteria, such as Escherichia coli and Bacillus subtilis. The aim of this thesis

was to understand how Streptococcus pyogenes, a strict human pathogen,

controls and adjusts gene expression by characterizing in vivo RNase activities.

The transcriptome of S. pyogenes was inspected to identify cleavages in vivo

performed by RNases of interest using RNA sequencing. For this purpose, we

developed a method to compare transcript 5′ and 3′ ends in RNase deletion

mutants with those in the parental strain. We first applied our method for the

study of endoRNase III, which cleaves ds RNA, and endoRNase Y, which is

specific for ss RNA. We accurately retrieved RNase III cleavage positions in

structured regions, characterized by 2 nucleotide (nt) 3′ overhangs, and we

showed RNase III nicking activity in vivo. We observed that RNase Y processed

transcripts after a guanosine. The upstream and downstream fragments

generated by a single cleavage event were never both identified, indicating that

RNase Y processing always led to the degradation of one of the two fragments.

To investigate further the degradation of the upstream fragment subsequent to

RNase Y processing, we characterized the 3′-to-5′ exoRNases R, YhaM, and

PNPase. RNase R did not have any detectable activity in standard laboratory

conditions. YhaM is an intriguing enzyme that removed on average 3 nt of the

majority of cellular transcripts. PNPase fully degraded fragments originating

from endoRNase processing and is the main 3′-to-5′ exoRNase involved in RNA

decay in S. pyogenes.

To conclude, in this work, we developed a novel method to analyze RNA

sequencing data. This method was successfully applied to the study of both

Page 9: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

iv

endo- and exoRNases. Most importantly, we identified the targetomes of

RNases III, Y, R, YhaM, and PNPase and we highlighted the distinctive features

of these enzymes.

Page 10: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

v

Abbreviations

5′ OH 5′ hydroxyl

5′ P 5′ monophosphate

5′ PPP 5′ triphosphate

cDNA complementary DNA

CRISPR clustered regularly interspaced short palindromic

repeats

crRNA CRISPR RNA

DNA deoxyribonucleic acid

DNase deoxyribonuclease

ds double-stranded

endoRNase endoribonuclease

exoRNase exoribonuclease

FDR false rate discovery

G guanosine

mRNA messenger ribonucleic acid

NAD 5′- nicotinamide-adenine dinucleotide

nt nucleotide

ORF open reading frame

PAP I poly(A) polymerase I

PFK phosphofructokinase

Pi inorganic phosphate

PNK polynucleotidekinase

poly(A) polyadenine

poly(N) polynucleotide

RBS ribosome binding site

RNA ribonucleic acid

RNase ribonuclease

rRNA ribosomal RNA

SAM S-adenosyl-L-methionine

sRNA small RNA

Page 11: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

vi

ss single-stranded

tmRNA transfer messenger RNA

tracrRNA trans-activating CRISPR RNA

TRAP trp RNA-binding attenuation protein

tRNA transfer RNA

TSS transcriptional start site

UTR untranslated region

WT wild type

∆rnase RNase deletion mutant

Page 12: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

vii

Publications included in this thesis

Paper I

Le Rhun A.*, Lécrivain A.-L.*, Reimegård J., Proux-Wera E., Broglia L., Della

Beffa C., and Charpentier E. (2017). Identification of endoribonuclease specific

cleavage positions reveals novel targets of RNase III in Streptococcus pyogenes.

Nucleic Acids Research. 45:(5), 2329-2340. *Contributed equally.

doi: 10.1093/nar/gkw1316

Paper II

Lécrivain A.-L.*, Le Rhun A*, Renault T. T., Ahmed-Begrich R., Hahnke K., and

Charpentier E. (2018). The in vivo 3′-to-5′ exoribonuclease targetomes of

Streptococcus pyogenes. PNAS. *Contributed equally.

doi: 10.1073/pnas.1809663115

Paper III

Lécrivain A.-L., Broglia L., Renault T. T., Hahnke K., Ahmed-Begrich R., Le

Rhun A. and Charpentier E. Interplay between 3′-to-5′ exoRNases and RNase Y

in Streptococcus pyogenes. Manuscript.

Published but not included in this thesis

Fonfara I.*, Le Rhun A.*, Chylinski K., Makarova K. S., Lécrivain A.-L., Bzdenga

J., Koonin E. V. and Charpentier E. (2014). Phylogeny of Cas9 determines

functional exchangeability of dual-RNA and Cas9 among orthologous type II

CRISPR-Cas systems. Nucleic Acids Research. 42:4, 2577-2590. *Contributed

equally. doi: 10.1093/nar/gkt1074

Broglia L., Materne S., Lécrivain A.-L., Hahnke K., Le Rhun A. and Charpentier

E. (2018). RNase Y-mediated regulation of the streptococcal pyrogenic exotoxin

B. RNA Biology. doi: 10.1080/15476286.2018.1532253.

Page 13: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)
Page 14: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

1

I- Introduction

In order to survive, every organism needs to adapt to multiple changes in

their environment, protect itself from predators and fight various threats. At the

microscopic scale, this translates into adjusting gene expression spatially and

temporally to regulate the levels of RNAs and proteins in the cell. The central

dogma describes the transfer of genetic information, from deoxyribonucleic acid

(DNA) to protein (1): DNA that composes the gene is first transcribed into

messenger ribonucleic acid (mRNA), which is then translated into a protein.

However, RNA – compRising all classes of mRNA, transfer RNA (tRNA),

ribosomal RNA (rRNA) and small RNA (sRNA) − is not merely an intermediate

between DNA and proteins but can alter the information delivered by the DNA

and can have functions on its own (2). Indeed, RNA has the ability to catalyze

enzymatic reactions, to be reverse transcribed into DNA, to be modified and to

control gene expression (2). Therefore, bacteria have developed many ways to

regulate gene expression at the RNA level, also called post-transcriptional

regulation.

Post-transcriptional regulation of gene expression

Post-transcriptional regulation in prokaryotes occurs at the level of

transcriptional elongation, RNA stability and translational initiation (3). Among

the elements that control gene expression without affecting mRNA stability, the

riboswitches are regulatory elements located in the 5′ UTRs of the transcripts

that they regulate. They directly sense environmental changes by binding

ligands, such as uncharged tRNA or S-adenosyl-L-methionine (SAM), or by

sensing temperature changes (4–6). The binding of the ligand triggers the

formation of a terminator or anti terminator hairpin (transcriptional

elongation), or it sequesters or free the ribosome binding site (RBS)

(translation). For more information, refer to the recent review (7).

This section focuses on how a bacterium controls protein expression through

RNA maturation and degradation, affecting mRNA stability. Many factors are

Page 15: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

2

involved but eventually the ribonucleases (RNases) act as the final agents to

clear unwanted RNA from the cells. RNA stability refers to the time a transcript

remains in the cell before it is being degraded (chemical stability) or inactivated

(functional stability) (8, 9). For instance, the functional stability of an mRNA is

defined by the time it undergoes translation and its chemical stability is the time

required for RNases to degrade it (8). Therefore, RNA stability depends on

intrinsic characteristics of the transcript, RNA-binding proteins, and sRNAs; all

further described below.

Each transcript has characteristics that will affect its half-life in the cell. The

5′ end of a primary transcript harbors a triphosphate nucleotide (5′ PPP) or a

5′- nicotinamide-adenine dinucleotide (NAD) cap that blocks endoribonucleases

(exoRNases) and 5′-to-3′ exoRNases (10–14). The sequence of a transcript

controls its targeting by proteins such as decapping enzymes or by sRNAs (15,

16). Structural elements at the transcript 5′ end (e.g. stem-loop or RNA duplex)

and at the 3′ end (e.g. transcriptional terminators) can efficiently prevent the

transcript degradation by exoRNases (17–21). However, these RNA duplexes

can also promote processing by double-stranded (ds) specific endoRNases (22).

RNA-binding molecules can interfere with the stability of transcripts by

preventing or promoting the access of RNases to their binding. RNA helicases

unwind structured transcripts and thereby promote degradation (23).

Polyadenylation performed by the poly(A) polymerase I (PAP I) at the 3′ ends of

transcripts promotes exoribonucleolytic degradation (24–26). Conversely, the

host factor I protein Hfq protects transcripts by binding polyadenine (poly(A))

tails (27). In addition, Hfq promotes the interaction of sRNAs to their mRNA

targets, which can result either in protection or triggering of RNA decay (28).

Ribosomes protect mRNA during translation initiation by binding the RBS (29–

31) and during translation elongation (32, 33). For more detail about the

influence and mechanisms of RNA-binding proteins on RNA stability, refer to

(3, 34).

sRNAs bind mRNAs and can stabilize them by masking an RNase binding-

site or by promoting RNase processing (35–39). Alternatively, sRNAs

destabilize their targets by triggering degradation, either directly by RNase

Page 16: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

3

recruitment or indirectly by blocking translation (40, 41). For more information

about the mechanisms involved, refer to (16, 42).

The final players are the RNases, which catalyze the cleavage of a

phosphodiester bond in a transcript. This leads to the degradation of the

substrate or, conversely, to the maturation of stable RNAs, such as rRNAs and

tRNAs (43, 44).

Ribonucleases

Ribonucleases are classified in endoRNases, which cleave in the body of a

transcript, and in exoRNases, which degrade a transcript starting from the 5′

end (5′-to-3′ exoRNase) or from the 3′ end (3′-to-5′ exoRNase). Some RNases

are phosphorolytic, i.e. they use inorganic phosphate (Pi) to break the

phosphodiester bond and release a nucleoside diphosphate, and others are

hydrolytic enzymes, i.e. they use a water molecule to break the bond and release

a nucleoside monophosphate.

This section focuses on the main RNases that regulate gene expression

and/or perform RNA decay and are present in S. pyogenes. Following is an

overview of the current general knowledge about RNase Y and RNase III

(endoRNases), RNase J1 (5′-to-3′ exoRNase), and YhaM, PNPase, RNase R, and

nanoRNase (3′-to-5′ exoRNases).

RNase Y

The single-stranded (ss) specific RNase Y is a dimeric enzyme that

preferentially targets transcripts with a 5′ monophosphate (5′ P) end (45, 46).

RNase Y is described as the main endoRNase initiating RNA decay in Gram-

positive bacteria such as B. subtilis, Clostridium perfringens and S. pyogenes

(39, 47–50), but has a smaller effect on global RNA stability and abundance in

Staphylococcus aureus (51, 52). This enzyme is involved in sRNA-mediated

RNA decay (39, 53) and in the degradation of 5′ UTR regulatory elements and

sRNAs (45, 47, 48, 51, 54, 55). Notably, RNase Y influences virulence gene

expression and the pathogenicity of S. aureus, S. pyogenes and C. perfringens

(39, 50, 56–60). In S. pyogenes, many virulence genes are controlled by

Page 17: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

4

RNase Y in a glucose-restricted medium (57). Some of these genes are likely

affected by an indirect transcriptional regulation. For example, our laboratory

recently showed that the RNase Y-dependent regulation of the speB transcript

abundance was mainly indirect (60).

RNase Y is located at the bacterial membrane via a transmembrane domain

(45, 61–63); however this does not seem to restrict RNase Y target selection in

vivo (51, 64). RNase Y orthologues present variable RNA structure and sequence

specificities. In S. pyogenes, a guanosine (G) is required for the in vivo

processing of the speB transcript by RNase Y (60). The presence of a G was also

observed directly upstream of 58% of RNase Y processing sites identified in S.

aureus (51) but it was not necessary for the processing of the saePQRS

transcript (65). In this bacterium, RNase Y activity is instead controlled by a

structured element located 6 nucleotides (nt) downstream of the processing site

(65). RNA duplexes and riboswitches were also observed around RNase Y

processing sites in B. subtilis and C. perfringens (39, 40, 45). These structured

regions were not detected in B. subtilis and S. aureus genome-wide studies of

RNase Y processing sites (51, 66). It should be noted that intermolecular

pairings (e.g. sRNA-mRNA duplexes) and complex structured regions are very

difficult to predict on a global scale. Last but not least, RNase Y interacts with

other proteins (see „RNA degradosome and other RNase complexes“ in this

thesis). For instance, the Y-complex – composed of the proteins YlbF, YmcA,

and YaaT – interacts in vivo with RNase Y and is required for the processing of

several polycistronic transcripts in B. subtilis (66, 67). Interestingly, the Y-

complex is not necessary for RNase Y activity but rather is an accessory factor

that potentially directs RNase Y towards specific transcripts (66).

In B. subtilis, RNase Y expression is growth-phase dependent (49). RNase Y

regulates other RNases; in B. subtilis, RNase Y matures RNase P RNA (44) and

potentially regulates its own gene expression and that of other RNases in S.

aureus (51).

Page 18: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

5

RNase III

The dimeric enzyme RNase III cleaves specifically in ds RNA, generating

products with a characteristic 2 nt 3′ end overhang (68). RNase III activity

mostly depends on structural motifs in the target RNA, as presented below.

Some reports have shown that this enzyme is able to cleave only one of the two

strands (i.e. nick) when an internal bulge or loop is present (69–72). The

biological effect of the nicking is illustrated by the study of the bacteriophage T7

transcript (73). The authors inserted a T7 stem-loop nicked by RNase III in a

human gene and showed that the in vivo mRNA stability was increased

compared to the wild type (WT) mRNA (73). Other specific motifs, such as a

bulge–helix–bulge on the dsRNA, allow RNase III to bind but prevent further

processing (70). In vivo, this leads to the stabilization of transcripts (74, 75) or

could activate translation initiation (70, 76, 77). These examples highlight the

various ways by which RNase III regulates gene expression by acting at the post-

transcriptional or translational level.

In addition to maturing the 16S and 23S rRNAs (78), RNase III participates

in the control of gene expression. Studies in E. coli and B. subtilis show that

RNase III affects approximatively 11-12% of the transcriptome (47, 79) and

many processing and binding sites have been identified genome-wide (80–82).

Due to its ability to process RNA duplexes, RNase III is involved in clearing

antisense transcripts generated from pervasive transcription in S. aureus (82,

83), in sRNA processing, and sRNA-mediated gene regulation in many bacteria

(82, 84–87). In B. subtilis, RNase III is essential due to the expression of

prophage-encoded toxins whose transcripts are degraded by the enzyme (40).

Moreover, the enzyme was shown to regulate directly translation initiation in

vivo (82, 88).

In many bacterial species, the intracellular level of RNase III is negatively

autoregulated, via RNase III processing its own transcript (rnc) leading to rnc

decay (80, 82, 89, 90). In addition, the enzymes activity is regulated at the post-

translational level. For instance, in E. coli, RNase III is bound by the protein

YmdB, which correlates with an increase in RNase III activity (91, 92).

RNase III activity is also hijacked by the T7 bacteriophage. The phage produces

Page 19: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

6

a kinase that enhances RNase III activity to increase the maturation of T7

transcripts (93).

RNase J1

RNase J1 is the only bacterial 5′-to-3′ exoRNase described so far and is

mostly present in Gram-positive bacteria (94, 95). This relatively recent

discovery – the 5′-to-3′ ribonucleolytic activity was described in 2007– had a

huge impact in the field, as it opened the possibility of a degradation pathway

starting at the 5′ ends of transcripts, like in eukaryotes (96). An

endoribonucleolytic activity was also described in vitro for RNase J1 (95),

although the in vivo relevance of this activity is now questioned (97). Some

bacteria also encode a paralogue of RNase J1, named RNase J2 (95). In contrast

to RNase J1, RNase J2 mostly has an endoribonucleolytic activity in vitro (94).

RNases J1 and J2 form heteroduplexes (95, 98), which alter the in vitro

cleavage pattern in comparison to the one of each individual enzyme (98). In

vivo, global changes in RNA and protein abundances were observed between

the corresponding single and double deletion strains (99). RNase J1 degrades 5′

P and 5′ hydroxyl (5′ OH) transcripts (94, 95), hence it is likely to rely on the

activity of RppH or endoRNases that initiate the degradation of transcripts (11,

12, 29). In addition, the enzyme requires at least 9 unstructured nucleotides at

the 5′ end to access its substrate and to degrade it (94, 100).

The deletion of RNase J1 affected approximatively 30% of B. subtilis

transcriptome and more than 80% of the Gram-negative Helicobacter pylori

transcriptome (47, 101), highlighting the central role of this enzyme in RNA

degradation. RNase J1 decays transcripts at their 5′ end upon removal of the 5′

PPP by RppH (11) or participates in clearing the decay intermediate fragments

that are generated by endonuclease cleavages (47, 51, 102, 103). Particularly,

RNase J1 is involved in the degradation of decay intermediate fragments

originating from transcript 3′-proximal ends, which contain transcriptional

terminators (80, 102, 103). Interestingly, the 30S ribosomal subunits present on

the 5′ UTRs during translation initiation block RNase J1 5′-to-3′

exoribonucleolytic activity and stabilize the transcripts (38, 94, 100, 104, 105).

Page 20: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

7

The role of RNase J2 is more obscure. In B. subtilis and S. aureus, it does not

seem to have any activity and is rather a structural support for RNase J1 (99,

106, 107). However, in Streptococcus mutans, RNase J2 processes the gbpC

transcript in vivo (37) and the deletion of Enterococcus faecalis RNase J2

influenced RNA abundance independently of RNase J1 (108).

While RNases J1 and J2 are dispensable in B. subtilis, S. aureus and S.

mutans (106, 107, 109), both proteins are essential in S. pyogenes (110).

PNPase

Polynucleotide phosphorylase, or PNPase, is a phosphorolytic enzyme that

has two functions. It uses Pi to degrade RNA in a 3′-to-5′ fashion, a reaction

that releases nucleoside diphosphates, and it also performs the reverse reaction,

i.e. the polymerization of nucleoside diphosphates without requiring a template

or a primer (111, 112). In vivo, however, PNPase behaves mostly as a degradative

enzyme and is thought to use its polymerase activity to add polynucleotide

(poly(N)) tails to transcript 3′ ends in E. coli (113–115). In B. subtilis, the

addition of poly(N) is not a major role of PNPase (116).

PNPase is present in bacteria, chloroplasts, and in mitochondria, but not in

archaea and yeast (117). PNPase is a trimeric enzyme that forms a ring-like

structure, with the catalytic site buried in the channel, and three RNA-binding

domains, which are presented in front of the channel entry (118, 119). The

distance between the RNA-binding domains and the catalytic sites determines

PNPase binding to ss tails of RNA of at least 7 nt (118, 120, 121). PNPase is

generally blocked by secondary structures such as intrinsic transcriptional

terminators (18, 122, 123). To overcome this obstacle, PNPase can use (i)

repeated rounds of tail polymerization at transcript 3′ ends by PAP I, or

allegedly by PNPase itself (124–127), (ii) the help of RNA-unwinding partners,

such as RNA helicases (126, 128–130), or (iii) the help of an endoRNase that

removes the structure (103, 122).

In B. subtilis and S. aureus, RNA decay is the major function of PNPase (34,

114, 131). In E. coli, PNPase is involved in RNA decay together with another 3′-

to-5′ exoRNase, RNase II (113, 132, 133). Despite this important role, the

Page 21: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

8

deletion of PNPase only mildly affects bulk RNA stability (133, 134), which

indicates that there is redundancy between PNPase and the other exoRNases to

degrade transcripts. PNPase principally targets decay intermediate fragments

and degrades them very efficiently, since these fragments are not detectable in

vivo (131, 132, 135, 136). A small fraction of PNPase is bound to the E. coli RNA

degradosome (137, 138), a multiprotein complex organized around the

endoRNase E, where it participates in mRNA and sRNA degradation (see „RNA

degradosome and other RNase complexes“ in this thesis).

In parallel, PNPase acts as a regulatory protein in vivo. In E. coli, the

exoRNase has an important protective role for sRNAs that are dependent on

Hfq (139–141). Conversely, sRNAs independent of Hfq are degraded by PNPase

(139, 140, 142). PNPase matures RliB, a transcript originating from a peculiar

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) array

that protects Listeria monocytogenes against invading plasmids (143). In B.

subtilis, PNPase matures the SR5 antisense RNA that is part of the toxin-

antitoxin system bsrE/SR5 (144) and specifically degrades the trp (tryptophan

operon) leader when it is bound to TRAP (trp RNA-binding attenuation

protein), resulting in the recycling of TRAP (145, 146).

As expected, the deletion of PNPase is associated with various phenotypes

(e.g. reduced survival at cold temperatures, changes in bacterial morphology

and biofilm formation) and affects bacterial competence or DNA damage repair,

among many cellular processes (147).

The expression and the activity of PNPase are tightly controlled. The

mechanisms vary from one bacterium to another and, to keep this section

simple, only a global overview is presented here. At the post-transcriptional

level, RNase III and PNPase act in concert to keep PNPase expression at a basal

level by processing the pnpA (coding for PNPase) 5′ UTR (148, 149). The master

carbon storage regulator (CsrA) prevents the processed pnpA transcript from

being translated (150). pnpA stability and translation is repressed by the

antisense RNA SraG (151). Additionally, PNPase binds its transcript in an

RNase III-independent manner and prevents its translation (152). PNPase

responds to cellular metabolites, such as ATP, magnesium-chelated citrate, or

ppGpp that activates or represses PNPase depending on the bacteria studied

Page 22: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

9

(153–155). During adaptation to cold, PNPase levels in the cell increases (156–

158).

In S. pyogenes, a study reported that PNPase destabilizes a few transcripts

coding for virulence factors (159).

RNase R

RNase R is a hydrolytic protein that belongs to the RNR family, compRising

unspecific 3′-to-5′ exoribonucleolytic enzymes (160). RNase R is a processive

enzyme, meaning that it stays bound to the substrate during the complete

degradative process (121, 161).

A particularity of RNase R is its ATP-independent helicase activity that

allows the efficient degradation of structured fragments (162–165). The

biological importance of the helicase activity is highlighted by the fact that

RNase R rescues the lethal deletion of the helicase CsdA in E. coli independently

of its RNase activity (162).

Like PNPase, RNase R needs a ss tail at the substrate 3′ end to bind and

degrade it (166–168). While 7 nt is a minimum, E. coli RNase R has the best in

vitro degradation rate with 10 nt (163, 168). The composition of the tail is

important, as RNase R prefers poly(A) tails to poly(U) tails downstream of a

structured RNA and does not efficiently bind poly(G) RNA or generally

substrates with a high G content (163, 168).

RNase R is mostly known for participating in quality control and maturation

of rRNAs (169–171) and transfer messenger RNA (tmRNA or SsrA RNA) (172).

tmRNA is involved in the trans-translation system that rescues ribosomes

stalled on mRNA lacking a stop codon and RNase R clears the cells from these

defective transcripts (172, 173). RNase R plays a role in mRNA decay by

degrading structured RNA fragments (136, 167) and transcripts (174, 175).

However, the deletion of RNase R has little impact on RNA abundance in H.

pylori, Pseudomonas putida, and in E. coli (176–178). In addition, the deletion

of RNase R together with other 3′-to-5′ exoRNases does not increase bulk RNA

stability in B. subtilis (179), suggesting a minor role of the protein in RNA decay.

The importance of RNase R becomes obvious during bacterial adaptation to cold

Page 23: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

10

shock, where global mRNA stabilization was observed in absence of RNase R

(180).

In E. coli, RNase R expression and activity are tightly controlled at multiple

levels. Protein abundance and/or activity are increased at elevated temperature

(175), upon cold shock, at the entry in the stationary phase of growth and under

starvation (172, 181). RNase R is normally unstable and the regulation of

expression described above occurs at the post-translational level (182). Indeed,

in absence of acetylation, RNase R interacts less tightly with tmRNA/SmpB

(trans-translation system actors). The weak interaction with tmRNA prevents

RNase R from being targeted by proteases (183–186). In addition, during the

exponential phase of growth, RNase R is mostly bound to the ribosomes, and

free RNase R is degraded to prevent unwanted global RNA degradation (187).

YhaM

YhaM has only been identified so far in Gram-positive bacteria (179). B.

subtilis YhaM efficiently degrades nanoRNA (5-mer) and long ss RNA regions in

vitro (179, 188), but is blocked by secondary structures (179). The S. aureus

orthologue of YhaM, Cbf1, is also able to degrade RNA in vitro, although less

efficiently than B. subtilis YhaM (179). The ribonucleolytic activity of YhaM was

detected in vivo as well (136, 189).

The physiological role of YhaM is unclear. B. subtilis YhaM restored the

viability of E. coli lacking the oligoRNase Orn, which is responsible for

degrading nanoRNAs produced by exoRNases (see “NanoRNases” below) (188).

Bulk RNA stability was not significantly affected when YhaM was removed from

B. subtilis (179) and the study of specific transcripts showed a very limited

participation in RNA decay (136). Interestingly, this enzyme is linked to DNA

replication and repair. Indeed, Cbf1 binds the replication enhancer of the

plasmid pT181 (190) and interacts with DnaC, a helicase involved in DNA

replication (191). B. subtilis YhaM presents a 3'-to-5' exoDNase activity in vitro

(179) and the expression of the protein is induced by UV or mitomycin C (DNA

damaging agent) in a RecA-dependent manner (192).

Page 24: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

11

Little is known about the regulation of YhaM. The enzyme is degraded when

B. subtilis enters the stationary phase of growth (193). In B. subtilis, the YhaM

promoter is bound by the carbon catabolite transcriptional repressor CcpN

(194) and harbors a binding site for the transcriptional repressor LexA (192).

NanoRNases

The nanoRNases NrnA, NrnB in Gram-positive bacteria and Orn in E. coli

are enzymes that degrade nanoRNAs into mononucleotides, an essential process

to replenish the pool of nucleotides available (188, 195–197). These nanoRNAs

(2 - 5 nt long) are usually produced by the exoRNases, such as PNPase and

RNase R, at the end of the RNA degradation process (196).

Interplay of RNases

How do organisms ensure the efficient and coordinated action of different

actors in crowed cellular environments? The compartmentalization of processes

is very well known in eukaryotic cells and is allowed by the presence of

organelles that concentrate the different proteins required for a same process

(e.g. the DNA replication or transcription machineries located in the nucleus).

Some prokaryotic organelles have been described, usually for very specific

processes (198). However, absence of organelles does not mean absence of

compartmentalization. In all organisms, proteins assemble in complex

machineries that gather distinct enzymatic activities in one location and

bacteria are not an exception. For instance in E. coli, the RNA degradosome is a

complex composed of an endoRNase, a 3′-to-5′ exoRNase, an RNA helicase and

a glycolytic enzyme. The characteristics of the RNA degradosome, as well as

other complexes described in Gram-positive bacteria, and its role in mRNA

decay is described below.

RNA degradosome and other RNase complexes

The RNA degradosome in E. coli and other Gram-negative bacteria is very

well studied. It is an assembly of multiple proteins that is involved in bulk

mRNA decay (137, 199). The central protein, RNase E, is generally anchored at

Page 25: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

12

the bacterial membrane through an amphipathic helix (200, 201). This enzyme

is essential (202) and it is presumably due to its importance for several major

processes, such as stable RNA maturation and the initiation of mRNA decay

(203–205). In E. coli, the C-terminal domain of RNase E interacts with the

other proteins from the degradosome, which are PNPase, the RNA helicase

RhlB and the glycolytic enzyme enolase (206). The composition of the

degradosome is dynamic and, under specific growth conditions, it involves other

partners, for instance Hfq or PAP I (207). The degradosome is not necessary for

bulk mRNA decay in E. coli, however this association between an endoRNase, a

3′-to-5′ exoRNase and an RNA helicase facilitates the degradation of structured

or otherwise inaccessible transcripts (206).

In Gram-positive bacteria, the existence of a stable RNA degradosome is

subject to debate (reviewed in (208, 209)). Several RNases have been shown to

interact with other RNases, RNA helicases, and glycolytic enzymes (see below).

Early studies proposed that B. subtilis RNase Y acted as the scaffold protein and

the central endoRNase in this complex (46, 210, 211). Multiple interactions

between RNase Y, PNPase, RNases J1/J2, the RNase RnpA, the RNA helicase

CshA, and the glycolytic enzymes enolase and phosphofructokinase (PFK) were

reported in B. subtilis and S. aureus (210–212). Some were contradicted by

other reports, such as the in vivo interaction between RNase Y and

RNases J1/J2 (94) and other were confirmed by in vitro techniques (213). In

addition, a recent in vivo localization study showed that PNPase, RNases J1/J2,

enolase, PFK, and CshA do not localize to the B. subtilis membrane, where

RNase Y is anchored (62). Many investigations were done on the binary

interactions between these enzymes. For instance, in B. subtilis, the glycolytic

enzyme glyceraldehyde 3-phosphate dehydrogenase (GapA) binds to RNase J1

and to RNase Y (214). In H. pylori and S. aureus, RNase J1 forms a complex

with an RNA helicase (215, 216). In S. pyogenes, RNase Y interacts with enolase

(57). In S. epidermidis, PNPase binds RNase J1 (217). In addition, RNase Y

interacts with the Y-complex in B. subtilis (see “RNase Y” in this thesis). In

conclusion, these studies suggest that the interactions between RNases,

helicases, and glycolytic enzymes are transient and unlikely to happen

altogether in vivo.

Page 26: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

13

Models of RNA decay

A general model of mRNA decay was proposed based on studies using E. coli,

which expresses endoRNases and 3′-to-5′ exoRNases (218). This model has

been updated with the numerous discoveries made over the years and accounts

for the different sets of RNases expressed in Gram-negative and Gram-positive

bacteria (219). The two main degradation pathways in E. coli (representative of

Gram-negative bacteria) and in B. subtilis (representative of Gram-positive

bacteria) are presented below.

In E. coli, the major endoRNase responsible for mRNA decay is RNase E

(206) and two main pathways have been described: the 5′ P-dependent pathway

and the direct entry pathway, each of them initiated by a different enzyme. In

the 5′ P-dependent pathway, the 5′ PPP present on primary transcripts is

removed sequentially by a yet unidentified enzyme and the RNA

pyrophosphohydrolase RppH (10, 220–222). This event triggers RNase E

cleavage in the transcript (10, 220). RNase E processing generates an upstream

decay intermediate fragment, which is not protected by a 3′ end structure and is

therefore degraded by 3′-to-5′ exoRNases; and a downstream decay

intermediate fragment with a 5′ P end. Because RNase E has a strong preference

for 5′ P transcripts, the downstream fragment becomes in turn a substrate for

RNase E that repeats the processing several times on each downstream decay

intermediate fragment generated (223, 224). The 5′ P-independent pathway,

also called “direct entry” pathway, relies on the ability of RNase E to bypass the

5′ P requirement and process a transcript with a 5′ PPP end (225–227). The

final steps of transcript degradation are performed by the two major 3′-to-5′

exoRNases: the hydrolase RNase II and PNPase (113, 132, 133).

In B. subtilis, which expresses the 5’-to-3’ exoRNase J1, two different

pathways are known and they differ at the initiation step. The first pathway

relies on an endoRNase that initiate the degradation of a primary transcript,

generating an upstream and a downstream decay intermediate fragment. The

initiator is mainly RNase Y, and in some cases RNase III (16, 228). The

upstream fragment is degraded by 3’-to-5’ exoRNases, mainly PNPase and

RNase R, and the downstream fragment is degraded by RNases J1/J2. The

Page 27: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

14

second pathway is initiated by RppH. The enzyme converts the transcript 5′ PPP

end in a 5′ P end in two steps, thereby making the transcript available for

RNase J1 exoribonucleolytic activity (11, 15).

For both E. coli and B. subtilis, mRNA degradation is an “all-or-none decay”

process (131, 229). Indeed, the decay intermediate fragments are only visible in

the absence of exoRNases, as shown in the global study of Liu et al. (131).

RNA sequencing

General principle

RNA sequencing relies on the reverse transcription of RNA into

complementary DNAs (cDNAs), which will be then sequenced (230). Many RNA

sequencing methods have been developed to answer specific scientific

questions, recently reviewed in (231) and (232). This section presents the global

workflow used in this thesis, which is based on the RNA sequencing method of

Dotsch et al. (233).

RNA sequencing to identify ribonuclease targets

First, the RNA samples are prepared for the ligation of the adapters. Total

RNA is extracted, DNase-treated and depleted from rRNAs. At this stage,

different treatments allow the selection of different transcripts. For instance, the

treatment with Tobacco Acid Pyrophosphatase (TAP) that converts 5′ PPP to 5′

P ensures the ligation of both primary and processed transcripts. Conversely, in

the absence of TAP treatment, only processed transcripts with a 5′ P are ligated.

The transcripts are mechanically fragmented before the treatment with T4

polynucleotidekinase (PNK), which converts 5′ OH to 5′ P.

The second step consists of preparing the cDNA library. The RNA fragments

are ligated to 5′ adapters (ligated at fragment 3′ ends) and to 3′ adapters

(ligated at fragment 5′ ends). The use of two adapters allows the identification

of the DNA strand of origin (sense or antisense). A primer that binds the 5′

adapter is used for the reverse transcription. Finally, the cDNA libraries are

sequenced.

Page 28: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

15

The last step is the analysis. The sequencing outputs are called “reads”,

which include the transcript sequences. The reads undergo quality control check

and adaptor sequence removal before being mapped to the corresponding

genome. The mapping is visualized as “coverage” of reads over the genome. The

“total coverage” represents the number of reads that cover each nucleotide of

the genome. In addition, depending on the conditions of library preparation and

the RNA sequencing parameters, it is possible to calculate the “read end

coverage”. This corresponds to the number of reads that start (5′ end) or stop

(3′ end) at each nucleotide on the genome, which define the transcript

boundaries.

To compare the gene expression between wild type (WT) and RNase deletion

mutant (∆rnase), the differential expression analysis is done with the

normalized total coverages using edgeR (234) or DEseq2 (235). To determine

the position of a processing, the differential expression analysis is done with the

end coverage between WT and ∆rnase.

Different methods to identify ribonuclease targets

To have a glance at the global impact of RNases in vivo, the simplest method

is to compare global RNA steady-state levels in WT and ∆rnase − i.e. differential

expression analysis. However, the distinction between transcriptional regulation

(i.e. indirect effect) and post-transcriptional regulation (i.e. direct effect) is not

possible with this analysis.

Another method is to arrest the cellular transcription and follow the stability

of transcripts over time and genome-wide. The stability of each mRNA as well as

the global mRNA stability can be calculated. For instance, the median mRNA

half-lives in Bacillus cereus and S. pyogenes were estimated at 2.4-2.6 min and

0.89 min, respectively, and the average mRNA half-life in E. coli was estimated

at 2.5 min (50, 236, 237). The deletion of RNase Y in S. pyogenes increased the

median mRNA half-life to 1.81 min, due to the stabilization of the majority of

the transcripts (50).

During the last years, different techniques have been developed to identify

more accurately the mRNA bound or processed by the RNase of interest. Some

Page 29: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

16

techniques were based on the selection of particular transcripts (structure or

nature of the transcript end) and some examples are described below. A

catalytically inactive variant of RNase III was used to isolate the dsRNA bound

to the enzyme in vivo (82). Alternatively, RNA duplexes were

immunoprecipitated with J2 antibody from WT and RNase III deletion strains,

which revealed a major role of RNase III on degrading dsRNA in E. coli (84).

The ability of metal-ion independent RNases to produce fragments with a 2′, 3′-

cyclic phosphate 3′ end was exploited to determine the transcripts processed by

RNase L (238).

In addition to identify the transcripts targeted by an RNase of interest, the

mapping of transcript ends allows the determination of the exact cleavage

positions (i.e. targetome). This is done by comparing the ends present in WT

and absent in ∆rnase strains. For instance, the mapping of transcript 5′ ends

pinpointed the exact cleavage sites of RNase J1 (5′-to-3′ exoribonucleolytic

activity) (106), RNase Y (51, 66), RNase III (80, 239), RNase E (225, 240) and of

the toxin MazF (241).

The mapping of the transcript 3′ ends has been principally used to study

operon architectures and specificities of transcriptional termination (242–246).

Streptococcus pyogenes

Streptococcus pyogenes (also called GAS, for Group A Streptococcus) is a

non-motile, non-spore forming bacteria that grows in chains. It belongs to the

Firmicutes phylum, the Bacilli class − like B. subtilis and S. aureus, two

bacterial species often mentioned in this thesis − and the Lactobacillales order.

The strains are classified into serotypes according to the sequence of the emm

gene, which encodes a virulence factor called the M protein (247).

A human pathogen

S. pyogenes is a strict human pathogen that is responsible for one of the

widest range of human diseases (248). S. pyogenes is transmitted via direct

Page 30: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

17

contact with a contaminated person or, very rarely, via contaminated food

(249).

Commonly, the bacteria infect the skin and the throat, leading to superficial

conditions (e.g. impetigo or pharyngitis) or to more severe diseases (e.g. scarlet

fever). S. pyogenes can progress deeper in the tissues and cause erysipelas,

cellulitis and necrotizing fasciitis. This last disease gave S. pyogenes the

nickname of “flesh-eating bacteria”. The bacteria are able to spread into the

bloodstream, leading to the streptococcal toxic shock syndrome (STSS), a life-

threatening condition. Due to cross-reactivity of some antibodies developed

against GAS during the infection, several complications can occur after the

bacteria have been efficiently eliminated (250). The most common ones are the

acute post-streptococcal glomerulonephritis (inflammation of the kidneys), the

acute rheumatic fever (mainly inflammation of the joints and heart) and the

rheumatic heart disease (inflammation of the heart).

S. pyogenes is responsible for more than 700 million cases of impetigo and

pharyngeal infections (251) and more than half a million deaths per year

worldwide (252). The treatment of choice is ampicillin, as no resistance to the

antibiotic has ever been developed by S. pyogenes (253). For patients who are

allergic to ampicillin, or in case of deep infections, macrolides are used but the

resistance rates to this molecules are increasing (254). The development of a

vaccine has proven to be challenging, due to the lack of an invariant and safe

antigen, but not impossible, as some vaccines are being evaluated in human

clinical trials (255).

S. pyogenes expresses many virulence factors to successfully establish an

infection (250). For instance, adhesins allow the bacterium to adhere to host

cells (256). SpeB, an unspecific protease that degrades both human and

bacterial proteins, promotes S. pyogenes dissemination (257, 258). To protect

itself against the immune response of the host, the bacterium produces

peptidases that degrade antibodies or a hyaluronic acid capsule that prevents

phagocytosis (259, 260). In addition, S. pyogenes produces toxins, such as

Streptolysin S, which create pores in the host cells (261).

Page 31: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

18

Post-transcriptional regulation of gene expression in S.

pyogenes

The bacterial strain studied in this thesis is S. pyogenes SF370 serotype M1

(ATCC 700294), which was originally isolated from a wound infection (262).

The S. pyogenes chromosome has a size of 1.8 Mb and a low GC content of

38.6% (262, 263).

The study of the post-transcriptional regulation of gene expression in S.

pyogenes focused mainly on the sRNA-mediated regulation and the discovery of

new sRNAs (85, 264, 265). The role of RNases was investigated, in particular of

RNase Y, RNases J1/J2, and PNPase (refer to the corresponding sections in this

thesis). Based on the studies of RNases J1/J2 and PNPase, a model of mRNA

decay dependent on the phase of growth was proposed (110, 159, 266).

According to this model, the transcripts are separated in two classes: the Class I,

which compRises transcripts detectable in exponential phase of growth, and the

Class II, which compRises transcripts mostly detected in stationary phase of

growth (159). Class I transcripts are short-lived, whereas Class II transcripts are

very stable and present a biphasic degradation pattern (159). RNases J1/J2 are

responsible for initiating the decay of transcripts from the two classes, with a

preference for Class I (110). Therefore, in stationary phase of growth,

RNases J1/J2 start to degrade the Class II transcripts after the cells were

depleted in Class I transcripts (110). The transcripts belonging to both classes

are further degraded by PNPase (266).

In conclusion, the post-transcriptional regulation of gene expression is still

poorly characterized in S. pyogenes and further investigations are required,

particularly about the roles of RNases and their specificities.

Page 32: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

19

II- Aims of the thesis

In this thesis, the roles of various RNases on the regulation of gene

expression in the human pathogen S. pyogenes were investigated. The approach

used was to establish the targetomes of these RNases, i.e. the exact processing

sites on a genome-wide scale. The specific objectives were:

1- To develop a novel analysis tool that uses RNA sequencing datasets to

identify the targetome of a given RNase.

2- To characterize the roles and specificities of the endoRNases Y and III,

and the 3′-to-5′ exoRNases R, YhaM, and PNPase.

3- To investigate the interplay between RNase Y and the 3′-to-5′ exoRNases.

Page 33: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

20

III- Results and discussion

Paper I

Identification of endoribonuclease specific cleavage positions

reveals novel targets of RNase III in Streptococcus pyogenes.

RNA sequencing is widely used to identify in vivo genome-wide RNase

cleavage positions (i.e. targetome). However, when our project to establish

RNase III targetome started, very few studies were available and relied only on

the sequencing of transcript 5′ ends (106, 225, 241). The disadvantage of this

method is that the RNase processing events leading to subsequent degradation

of transcript 5′ ends are not identified. We sequenced both transcript 5′ and 3′

ends to increase the detection of processing events. To determine the global

processing positions of an endoRNase of interest, we compared the abundances

of transcript 5′ and 3′ ends in WT and ∆rnase strains. The ds specific RNase III

was a perfect model to validate this method of analysis, as it generates

theoretically two new 5′ ends and two new 3′ ends for each cleavage event.

Methodology

Two different kinds of libraries (p and Pp) were prepared using total RNA of

WT, ∆rnc (lacking RNase III), ∆rny (lacking RNase Y), and double mutant

∆rny_∆rnc strains (Paper I, Supplementary Figure S1). The Pp library

contained the processed and primary transcripts and the p library was enriched

in processed transcripts. The total coverage as well as the coverages of the 5′ end

and the 3′ end (i.e. the ends of the read) were calculated for each position on the

genome. To insure that we compared positions with enough coverage in all

strains, we filtered out the positions with less than 10 reads in the reference

(expressing RNase III, i.e. WT and ∆rny) and mutant strains (not expressing

RNase III, i.e. ∆rnc and ∆rny_∆rnc). Then, we defined several parameters to

isolate the specific cleavage positions from the background noise. These

parameters were less stringent for the p libraries, where the processed

Page 34: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

21

transcript ends were enriched, than for the Pp libraries. The following settings

were used for the p libraries (Paper I, Figure 1): (i) the position of interest

cumulated a minimal read end coverage of 10 in the two strains that were

compared; (ii) the reads that ended at the position of interest represented at

least 16% of all the read ends in a 20 nt-window centered at this position; (iii)

the percentage of reads ending at the position of interest compared to the total

coverage at the position of interest was at least 5%; and (iv) the ratio of the

percentages obtained in (iii) for the reference strain and for the mutant strain

was at least 3.

Characteristics of S. pyogenes RNase III

We detected 92 cleavage positions that were dispatched on 25 transcripts

and on the six pre-rRNA transcripts. For more than half of the 25 transcripts,

we retrieved both RNase III cleavage positions in a stem-loop and in two cases,

in antisense transcripts (Paper I, Figure 3A). The 2 nt 3′ overhang characteristic

of RNase III processing was detected, which validated the accuracy of our

method. We identified a single cleavage site (referred to as stand-alone

positions, or SA) for the rest of the transcripts (Paper I, Figure 3A). Although a

relaxed consensus sequence was described for E. coli RNase III around the

cleavage site (72, 239, 267), we did not observe any conserved sequence or motif

for S. pyogenes RNase III.

When we visually inspected the individual reads mapped at the RNase III

double cleavage positions, we discovered that, for a given target, a portion of the

transcripts were cleaved only at one position (i.e. cleavage in one side of the

RNA stem) (Paper I, Figure 3C). This nicking activity was visible on each strand

for a given target, which means that RNase III did not favor one strand in

particular. It was previously observed that E. coli RNase III had a nicking

activity in vitro and in vivo (70, 71, 267). Our primer extension analyses

confirmed the presence of two alternative 5′ ends (nicking at one or the other

side of the RNA stem) for four transcripts (Paper I, Supplementary Figure S2).

The transcripts where we could observe RNase III nicks harbored at least one

bulge or a loop located near one of the two cleavage positions (Paper I, Figure

Page 35: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

22

3B-C, Supplementary Figure S4). These structural features are known to

destabilize RNase III binding to the RNA after the first cleavage (69, 72). The

accessibility of the upstream fragment, generated by RNase III nicking, to 3′-to-

5′ exoRNases was investigated in Paper II. The study of RNase III nicking

activity on pnpA 5′ UTR showed that PNPase was able to degrade the two

alternative 3′ ends (Paper II, SI appendix, Fig. S7). Thus, for this specific target,

the nicks did not alter the degradation of the upstream fragment. Up to this

date, there is only the T7 transcript that is nicked and stabilized by RNase III,

which illustrates the biological impact of RNase III nicking activity (73). We

showed that RNase III nicking activity is a general phenomenon in S. pyogenes

but the impact on gene regulation remains unknown.

SA processing positions were detected in ss regions of eight transcripts. We

first reasoned that these transcripts base-paired with an unknown transcript

and that the second processing position was in the complementary mRNA or

sRNA. Unfortunately, we could not identify base-pairing partners for these

transcripts. Other explanations were considered: (i) RNase III cleaved in an

unstable ds region (predicted as ss) and the fragment generated by the other

processing was degraded and therefore not detected; (ii) RNase III nicked in an

unstable ds region (predicted as ss); (iii) RNase III cleaved in a RNA stem-loop

that was subsequently trimmed by the 5′-to-3′ exoRNase J1 for the 5′ end

positions, or by the 3′-to-5′ exoRNases for the 3′ end positions, hence the

information about the original cleavage positions of RNase III was not retrieved.

Two SA positions were associated with a change in abundance of neighborhood

transcript (Paper I, Supplementary Table S3), therefore the study of these SA

targets could reveal novel mechanisms of gene regulation by RNase III.

RNase III role in S. pyogenes

As described in other bacteria, we found that S. pyogenes RNase III

participated in rRNA maturation (78). The enzyme processed the pre-rRNA

transcripts in the stem-loops delimiting the 16S and 23S rRNAs, which were

further matured by other RNases (Paper I, Supplementary Figure S3). RNase III

was not essential to this process, as RNase III deletion did not influence the

Page 36: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

23

maturation of the 16S and 23S rRNAs. In this context, we detected additional

processing positions (called substitute processing events) on the pre-rRNAs that

were not present in the WT (Paper I, Supplementary Table S4). This indicated

that the rRNA maturation was performed by other RNases to ensure that

ribosomes were still functional in absence of RNase III.

S. pyogenes RNase III targeted mostly UTRs, as for S. aureus RNase III (82),

but in contrast to what has been described in E. coli and B. subtilis (80, 239).

This suggests that the cleavages would affect gene expression. However, out of

the 25 transcripts cleaved by RNase III (excluding rRNAs), the abundance of

only seven was affected. It is possible that RNase III regulates transcript

stability or translation without affecting the transcript abundance. For instance,

in S. pyogenes, the global transcript stabilization caused by RNase Y deletion

did not reflect on the transcript abundance (50). Therefore, alternative methods

should be used to investigate RNase III regulatory effects.

We identified several mRNAs that were known targets of RNase III in other

bacteria, such as pnpA. In E. coli, RNase III cleaves a stem-loop in the 5′ UTR of

pnpA, which leads to pnpA destabilization (147). We observed a similar cleavage

by S. pyogenes RNase III in pnpA 5′ UTR and pnpA abundance was increased

in absence of RNase III (Paper I, Figure 3B and Supplementary Table S6).

Another target, the secY-adk transcript (coding for Sec translocase and

adenylate kinase), was cleaved by RNase III in S. aureus (82). More specifically,

RNase III cleaves the transcript in the secY ORF and the absence of cleavage

leads to an increased secY abundance and stability (82). We showed that a

different regulatory mechanism occurs in S. pyogenes. RNase III cleaved in the

UTR between the secY and adk, which uncoupled the expression of the two

genes as only adk was more abundant in ∆rnc (Paper I, Figure 3C and

Supplementary Table S6). These examples highlight that, while some regulatory

mechanisms are conserved, bacteria also develop alternative mechanisms to

regulate the expression of similar transcripts using orthologue RNases.

RNase III is involved in sRNA-mediated gene regulation (84, 85, 87, 268). In

this study, few sRNAs were cleaved by RNase III or differentially abundant

(Paper I, Supplementary Table S7). For instance, we retrieved RNase III

processing in the putative 23S methyl regulatory element, in the antisense RNA

Page 37: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

24

SPy_sRNA477741 (85), in CRISPR RNAs (crRNAs), and trans-activating

CRISPR RNA (tracrRNA) (87). sRNAs were lost during our library preparation,

which could explain the limited number of RNase III targets.

In conclusion, we determined that RNase III participates in conserved

regulatory and maturation pathways and has a limited specific role in S.

pyogenes. Further work is required to evaluate the impact of RNase III on

sRNAs, which may be broader than what was described in this study.

Paper II

The in vivo 3′-to-5′ exoribonuclease targetomes of Streptococcus

pyogenes.

In Paper I, we identified and validated RNase processing positions by

sequencing both the 5′ and the 3′ ends of transcripts. This opened the

possibility to study the targetomes of 3′-to-5′ exoRNases. PNPase and RNase R

have been intensively studied in various organisms, yet the global identification

of their direct targets was missing. We applied our method to the study of

YhaM, PNPase, and RNase R, the three 3′-to-5′ exoRNases present in S.

pyogenes.

Methodology

We improved the previous method presented in Paper I by adding a step of

normalization between the samples and including statistical power using false

discovery rate (FDR). Total RNA was extracted in triplicates from WT, ∆yhaM

(lacking YhaM), ∆pnpA (lacking PNPase), and ∆rnr (lacking RNase R). EdgeR

was used to analyse the differential abundance of transcript 5′ and 3′ ends

between the reference strain and the mutant strain (234). The positions with an

absolute fold change of at least 2 and an FDR below 0.05 were used for further

analysis. We applied the parameters from Paper I on these positions: the

percentage of reads ending at the nucleotide of interest compared to all the

reads that mapped at the nucleotide of interest was at least 2%; and the ratio of

Page 38: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

25

the percentage obtained for the reference and the mutant strains was at least 3.

We used the Pp libraries for all the strains.

The positions with a negative fold change (i.e. more abundant in the WT)

corresponded to exoRNase trimming stop positions and those with a positive

fold change (i.e. more abundant in the ∆exornase) corresponded to exoRNase

trimming start positions. The distance between them represented the

processivity of the enzyme. Approximatively 90% of all the positions more

abundant in the WT were transcript 3′ ends, which confirmed the in vivo 3′-to-

5′ exoribonucleolytic activity of these enzymes.

YhaM has a global nibbling activity on transcript 3′ ends

We identified 602 trimming stop positions for YhaM. The conservation of a

secondary structure and a stretch of uridines signaled the presence of intrinsic

transcriptional terminators upstream half of these positions (Paper II, Fig. 2

and SI appendix, Fig. S2A-B). The second half of the positions presented no

particular conservation of sequence or structure (Paper II, SI appendix, Fig.

S2C-D). These positions were mostly found in ORFs, which suggested that these

transcript 3′ ends originated from endoRNase processing. We estimated that

the enzyme trimmed approximatively half of the terminated transcripts ends, in

addition of the transcript 3′ ends produced by endoRNases. Therefore, YhaM

has a broad targetome in S. pyogenes. It would be of interest to understand why

some transcripts are not targeted by YhaM. Perhaps factors such as low

transcript abundance, too short or absent ss tail, or the localization of the

transcripts in the cell can restrict YhaM activity.

The 602 trimming stop positions were associated with the closest 171

trimming start positions (maximum distance of 10 nt) to estimate YhaM

processivity. We obtained 112 pairs located within 10 nt from transcriptional

terminators and the 42 pairs located at putative processing sites. The two

groups of pairs revealed that YhaM trimmed 3 nt on average (Paper II, Dataset

S2), which implied that the terminator structure did not limit YhaM

processivity. YhaM nibbling activity was additionally validated by Northern

blotting analyses (Paper II, SI appendix, Fig. S4). Almost all the trimming start

Page 39: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

26

positions were paired with the stop positions, supporting that the short

processivity was not a bias of the distance of pairing. Oppositely, the paired

trimming stop positions represented only a small fraction of the total number of

trimming stop positions. This can be explained by the fact that many YhaM

trimming start positions were not identified (for more details about the

limitation of our method, refer to Paper II, SI appendix, Fig. S3A-B). Indeed, by

visual screening, we could observed these trimming start positions 1-3 nt

downstream of the trimming stop positions (examples presented in Paper II, SI

appendix, Fig. S3C). We cannot exclude that, among the unpaired trimming

start positions, some may be indicative of a processivity for YhaM higher than 3

nt. However, this would be a minor activity compared to YhaM nibbling activity.

Role and peculiarities of S. pyogenes YhaM

It has been suggested that YhaM acted more as a DNase than as an RNase in

vivo (269). Our study shows that S. pyogenes YhaM is an unspecific RNase that

broadly targets transcript 3′ ends originating from termination or processing by

other RNases. The removal of 3 nt on average at transcript ends raised several

questions.

First, what prevents YhaM from trimming further the transcript ends? (i) It

could be due to the intrinsic biochemical properties of the enzyme. However, B.

subtilis YhaM was able to degrade a 110 nt substrate in vitro (179). (ii) It could

be transcript secondary structures; yet, we failed to detect any structure that

could have blocked YhaM at the transcript 3′ ends generated by endoRNases.

(iii) It could be a balance between the cellular concentration of YhaM and of the

transcripts, yet we did not observe a correlation between transcript abundance

and trimming. In E. coli, a similar observation was recently made for the 3′-to-

5′ exoRNase II, which nibbles 1-3 nt at transcript intrinsic terminators (243).

The authors did not comment on whether RNase II nibbled only at the

transcript intrinsic terminators or at unstructured transcript 3′ ends as well. As

RNase II is blocked approximately 9 nt downstream of secondary structures

(270), this physical constraint could be an explanation. The mechanisms behind

the short processivity of YhaM and RNase II are likely to be different.

Page 40: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

27

Interestingly, the same study described a narrow YhaM nibbling activity at

intrinsic transcriptional terminators in B. subtilis (15% of the intrinsic

terminators) (243). It is possible that YhaM has a broader targetome in S.

pyogenes than in other bacteria. The comparison of the targetomes of YhaM

orthologues belonging to the Bacillales order (e.g. B. subtilis and S. aureus) and

to the Lactobacillales order (e.g. S. pyogenes) will help answering this question.

Secondly, what is the biological purpose of this nibbling? RNase II is

suggested to have a protective role on transcripts by shortening the tail required

for PNPase and RNase R binding and degradation (133, 271, 272). We observed

that the ss tail downstream of terminators was reduced from 9 to 6 nt on

average in the WT compared to ∆yhaM (Paper II, SI appendix, Fig. S1). For E.

coli PNPase, these tails would be too short and the transcripts would be

protected (120). To gain some insight on the YhaM possible protective role, we

looked at the differential gene expression in ∆yhaM compared to the WT. Forty

transcripts were differentially expressed and nineteen were downregulated in

∆yhaM (Paper II, Dataset 3). This small number of transcripts did not correlate

with the global nibbling performed by YhaM. Additionally, the transcript 3′

ends generated by endoRNases did not have a secondary structure that would

protect them from PNPase or RNase R (Paper II, SI appendix, Fig. S2D). Thus,

removing 3 nt from the ss tail of these transcripts seems unnecessary.

We observed that S. pyogenes growth at cold temperature was greatly

impacted by the deletion of YhaM (Paper II, SI appendix, Fig. S9B).

Interestingly, the deletion of PNPase or RNase R, which are known to be

essential or important for optimal growth at cold temperatures in various

bacteria (147, 172), did not have such an effect. It was previously shown in B.

subtilis that the deletion of YhaM in the double mutant ∆pnpA_∆rnr slowed

bacterial growth at cold temperatures (179). We observed a synergic effect when

YhaM was deleted in ∆rnr but not in ∆pnpA (Paper II, SI appendix, Fig. S9B). It

is tempting to speculate that YhaM activity becomes important when S.

pyogenes encounters specific growth conditions. In these conditions, it is not

known whether the slower growth phenotype is due to the absence of global

nibbling on the transcript 3′ ends, to an indirect effect of the enzyme deletion on

specific targets, or to its putative DNA-binding activity.

Page 41: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

28

PNPase role in RNA decay

The deletion of PNPase revealed 1255 trimming start positions and 183

trimming stop positions. The small amount of trimming stop positions implied

that PNPase fully degraded its targets (i.e. until the 5′ end). Therefore, we

assumed that transcript 5′ ends should be more abundant in absence of PNPase.

It should be noted that the fragments whose 5′ end corresponds to

transcriptional start sites (TSS) were not identified with our method (for a

detailed explanation, refer to Paper II, SI Appendix, Fig. S3). Indeed, we noticed

336 transcript 5′ ends that accumulated in ∆pnpA (24 and 8 times more than in

∆yhaM and ∆rnr, respectively; Paper II, Fig. 1A). By pairing the transcript 5′

ends with the closest trimming start positions, we showed that 185 fragments

with a median size of 115 nt were present in ∆pnpA. As 85% of these fragments

were located in ORFs, we assumed that they originated principally from the

mRNA decay pathway initiated by endoribonucleolytic activities. These

fragments were not detected in the WT, therefore they were immediately

degraded by PNPase. In B. subtilis, PNPase degrades principally decay

intermediate fragments originating from the 5′ proximal part of transcripts

(131). We could not observe a similar pattern, due to the impossibility to detect

fragments accumulating at the TSS in ∆pnpA (for a detailed explanation, refer

to Paper II, SI Appendix, Fig. S3).

Less than 20% of PNPase total trimming start positions were located in

UTRs. Some of them were downstream of regulatory 5′ UTRs, for instance T-

boxes that controlled aminoacyl-tRNA synthetase expression by binding tRNAs.

Northern blotting analyses of 10 regulatory 5′ UTRs revealed that decay

intermediate fragments accumulated in ∆pnpA for 9 of them (Paper II, Figs. 3B

and 4, and SI Appendix, Fig. S8). For example, when the pyrR leader was

probed, the decay intermediate fragments were longer than the full length

leader, indicating that the initial processing events were in the pyrR coding

sequence instead of the 5′ UTR. Indeed, several PNPase trimming start

positions were detected downstream of the leader (Paper II, SI Appendix, Fig.

S8). With the notable exception of FMN, the stability of the full-length leaders

was unaffected by the deletion of PNPase, hence PNPase is not initiating the

Page 42: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

29

decay. Additionally, we observed that the dpr leader was matured by PNPase

(Paper II, Fig. 3C and SI Appendix, Fig. S8). Interestingly, this transcript codes

for a peroxide resistance protein (273), thus the function of this stable sRNA in

oxidative stress should be investigated. In view of these results, our study shows

that PNPase is also responsible for degrading 5′ UTRs in S. pyogenes. Many

regulatory 5′ UTRs adopt different structural conformations based on external

stimuli such as temperature or binding of a ligand (274). For instance, B.

subtilis PNPase preferentially degrades the regulatory 5′ UTR of trp when it is

bound by TRAP (145, 146). It would be of interest to study whether this

conformational preference is observed for S. pyogenes PNPase.

As PNPase relies on endoRNases to degrade the decay intermediate

fragments, we compared PNPase trimming stop positions with RNase III

cleavage positions (Paper I) (275). Indeed, PNPase trimmed fragments

produced by RNase III, for example the decay intermediate fragments generated

during rRNA maturation and the processing of pnpA 5′ UTR (Paper II, SI

appendix, Figs. S6 and S7). PNPase also trimmed approximatively 25 nt of rplQ

3′ UTR upstream of RNase III cleavage (Paper II, SI appendix, Fig. S7).

In conclusion, PNPase is the major 3′-to-5′ exoRNase involved in S.

pyogenes RNA decay and the processing of the dpr leader hints toward a

possible regulatory role for PNPase in S. pyogenes, as it is the case in many

bacteria.

Limited RNase R activity

A total of 82 trimming positions were retrieved for RNase R. Only two

transcripts harboring a trimming start position and a trimming stop position

and six decay intermediate fragments were identified. One trimming start

position was located in the glyQ leader, a T-box riboswitch, which was also

degraded by PNPase (Paper II, SI Appendix, Fig. S8). Northern blotting

analyses confirmed that glyQ leader was degraded by PNPase and RNase R

(Paper II, Fig. 4). However, the decay intermediate fragment was more stable in

absence of RNase R than of PNPase. We hypothesized that (i) RNase R could be

more efficient in degrading this decay intermediate fragment than PNPase

Page 43: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

30

(redundant activity), or (ii) there could be two populations of intermediate

fragments (tRNA-bound and free) and the population accumulating in ∆pnpA is

different from the one accumulating in ∆rnr, as the trimming start positions of

RNase R and PNPase were few nt apart (specific activity) (Paper II, SI

Appendix, Fig. S8). In the second case, each 3′-to-5′ exoRNase would then be

responsible for degrading one population of glyQ leader. In absence of RNase R,

PNPase would degrade both glyQ leader populations and would need more time

to degrade the RNase R-specific population. Reciprocally, in absence of PNPase,

RNase R would be able to degrade both glyQ leader populations, and would

need more time to degrade the PNPase-specific population.

Possible redundancy between RNase R and PNPase

Several reasons could explain the very limited targetome observed for

RNase R. First, it could indicate a low expression or a reduced activity of the

enzyme under standard laboratory conditions of growth (mid-logarithmic phase

of growth, rich medium, optimal temperature). Indeed, RNase R in Gram-

negative bacteria is more stable and more expressed under stress conditions

(276). Secondly, RNase R could degrade transcripts from transcriptional

terminator to TSS, which were not detected by our method (Paper II, SI

appendix, Fig. S3E). The third explanation is that PNPase would compensate

when RNase R is absent, thereby preventing the identification of RNase R

targets. In favor of this hypothesis, RNase R activity is mostly observed in

absence of PNPase in E. coli and B. subtilis (136, 167). In addition, the double

mutant ∆pnpA_∆rnr could not be obtained in the conditions tested in S.

pyogenes. In E. coli, ∆pnpA_∆rnr is also lethal, probably due to the

accumulation of abnormal rRNA precursors (169, 277). Interestingly, in B.

subtilis, all the 3′-to-5′ exoRNases (PNPase, RNase R, YhaM, and RNase PH)

can be deleted together (136), which suggests a different pathway of rRNA

quality control than in E. coli. If the lethality of ∆pnpA_∆rnr in S. pyogenes is

confirmed, it would be of interest to investigate the biological reason behind.

The decay intermediate fragments originating from the regulatory 5′ UTRs

that accumulated in ∆pnpA were eventually degraded by another exoRNase

Page 44: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

31

(Paper II, SI Appendix Fig. S8). Therefore another RNase, presumably RNase R

or RNase J1, can compensate the loss of PNPase in RNA decay pathway. In this

case, the redundancy is only partial, as we observed many specific PNPase

trimming positions in our screening (Paper II, Dataset S1). B. subtilis PNPase is

also the major 3′-to-5′ exoRNase (131, 136) and in its absence, RNase R degrade

some decay intermediate fragments (136). However, the combined deletion of

PNPase, RNase R, RNase PH, and YhaM is not lethal in B. subtilis (136),

suggesting either that the 3′-to-5′ degradation pathway is not necessary or that

an additional 3′-to-5′ exoRNase remains to be identified.

Both B. subtilis and S. pyogenes express RNases J1 and J2, however these

enzymes are only essential in S. pyogenes (107, 110). The essentiality of the 3′-

to-5′ degradation pathway and of the 5′-to-3′ degradation pathway can imply

that they have distinct functions in S. pyogenes.

Paper III

Interplay between 3′-to-5′ exoRNases and RNase Y in

Streptococcus pyogenes.

As a continuation of Paper II, we decided to investigate the implication of

RNase Y in the generation of transcript 3′ ends that are targeted and degraded

by the 3′-to-5′ exoRNases YhaM, PNPase, and RNase R. First, we applied the

analysis method used in Paper II (see Paper II, ”Methodology” in this thesis) to

determine the targetome of RNase Y. Total RNA was extracted from WT, ∆rny

(lacking RNase Y), and ∆rny::rny (complemented strain) that was used as a

second reference strain. Secondly, we compared the targetome of RNase Y with

the targetomes of the 3′-to-5′ exoRNases.

RNase Y cleaves after a guanosine

We identified 320 processing positions, 60% were detected as 5′ ends (190)

and 40% were detected as 3′ ends (130). The majority of transcript 5′ ends

(87.4%) generated by RNase Y were located immediately after a guanosine (G)

(Paper III, Figure 1D). A recent study from our laboratory showed that RNase Y

Page 45: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

32

requires a G to process the speB transcript in vivo (60). The conserved presence

of a G in our screening suggests that this requirement is a general feature of S.

pyogenes RNase Y.

It is difficult to envision how the G alone can restrict S. pyogenes RNase Y

activity to the limited number of processing positions we identified. A G residue

was previously identified upstream of 58% of the 99 transcript 5′ ends produced

by RNase Y in S. aureus (51), which is similar to what we observed. In

comparison, Salmonella enterica RNase E prefers a U located 2 nt downstream

of the processing position and 22000 processing sites were reported in vivo

(240). Thus, the specificity towards 1 nt is unlikely to restrict RNase Y activity

enough to explain the small targetomes detected in S. pyogenes and S. aureus.

The specificity of S. aureus RNase Y was further investigated on the saePQRS

transcript, which was processed just downstream of a U (65). Instead of a

sequence motif, S. aureus RNase Y requires a downstream structured element

to cleave the saePQRS transcript (65). Global studies done in B. subtilis, in S.

aureus as well as our study in S. pyogenes did not reveal structured regions near

RNase Y processing positions (51, 66). The identification of complex structured

regions among the targets is difficult; therefore, it is possible that undetected

structures are important to direct RNase Y processing of some transcripts.

Another possibility is that S. pyogenes RNase Y specificity would be altered

when RNase Y associates with other proteins, such as the Y-complex described

in B. subtilis (66). The localization to the membrane could restrict RNase Y

access to the transcripts. However, this was not the case in S. aureus and S.

pyogenes (51, 64). It is possible that RNase Y has a broader targetome than

what we detected, which is undetectable because the exoRNases degraded the

transcripts produced by RNase Y. It is not clear whether RNase Y orthologues

have different specificities or whether RNase Y responds to multiple signals that

have not been fully characterized. The co-crystal structure of this protein with

its target is crucially missing to improve our understanding on RNase Y

specificity.

Page 46: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

33

RNase Y and PNPase act in concert to degrade transcripts

The RNase Y-dependent 130 transcript 3′ ends did not present the conserved

G. We assumed that these transcripts were initially processed by RNase Y and

were then further trimmed by 3′-to-5′ exoRNases. The transcript 3′ ends

detected in our screening would therefore correspond to the trimming stop

positions of the exoRNases. To prove this hypothesis, we compared the

targetome of RNase Y with the trimming start and stop positions of YhaM,

PNPase, and RNase R previously published in Paper II (Paper III, Figure 2).

The positions of six and forty-six RNase Y-dependent transcript 3′ ends

corresponded to PNPase trimming start and stop positions, respectively (Paper

III, Figure 2A, panels 1-2 and Tables S2-S3). The six PNPase trimming start

positions, which would correspond to the nucleotide upstream of RNase Y

original processing positions, were located at a G (Paper III, Table S3). These

results showed that most of the transcript 3′ ends produced by RNase Y and

trimmed by PNPase were immediately trimmed (i.e. mostly trimming stop

positions detected). It was shown that RNase Y and PNPase interact in B.

subtilis, which could explain the immediate degradation of transcripts produced

by RNase Y (278). However, the disruption of the complex did not affect the

decay of the transcripts studied (278). In addition, we retrieved 27 transcript 3′

ends nibbled by YhaM and one transcript 3′ end trimmed by RNase R, in

accordance with YhaM broad nibbling activity and RNase R limited detectable

activity described in Paper II. The rest of the RNase Y-dependent transcript 3′

ends did not present a conservation of G. They could originate from PNPase and

RNase R redundant trimming (which we cannot detect using our method), from

another unidentified 3′-to-5′ exoRNase expressed in S. pyogenes, or from

another RNase whose activity is dependent on RNase Y.

We did not retrieve both the transcript 3′ end (i.e. upstream fragment) and

5′ end (i.e. downstream fragment) produced by a single RNase Y processing

event. Because our results showed that 3′-to-5′ exoRNases trimmed transcripts

produced by RNase Y, we searched for the 3′-to-5′ exoRNase trimming start

positions located near the transcript 5′ ends produced by RNase Y (Paper III,

Figure 2A, panel 3). Both transcript ends were observed for 23 RNase Y

Page 47: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

34

processing events and PNPase trimmed all the transcript 3′ ends identified

(Paper III, Table S4). Many RNase Y-dependent transcript 5′ ends (167) were

not paired with the corresponding upstream fragments. This implies that other

3′ ends are missing, probably because they were fully degraded or because they

were trimmed by RNase R in absence of PNPase.

PNPase generally cannot degrade Rho independent terminators and depends

on the activity of endoRNases to remove terminators in E. coli and B. subtilis

(103, 122, 123, 203). S. pyogenes does not have a Rho orthologue (279),

therefore all the transcriptional terminators are in theory intrinsic terminators.

Some RNase Y processing positions were located upstream of transcriptional

terminators and attenuators. For instance, RNase Y removed the transcriptional

attenuator from the glyQ leader (Paper III, Table S2), which was further

processed by PNPase and RNase R (Paper II, SI Appendix, Fig. S8). An RNA

decay mechanism such as the one initiated by RNase Y on the glyQ leader must

be general. We do not know whether RNase Y is the global initiator of decay by

removing intrinsic terminator structures, because few positions close to the

transcriptional terminators were observed. In some cases, the small size of the

downstream fragments containing the terminators prevented the detection of

RNase Y processing positions at the terminators. Their identification could be

prevented as well by RNase J1, shown to degrade the fragments containing the

transcriptional terminators in B. subtilis (80, 102, 103).

Because PNPase degraded the transcript 3′ ends generated by RNase Y, we

had a closer look at the transcript ends that accumulated in ∆pnpA (Paper II, SI

appendix, Fig. S2E). A conserved G was visible directly upstream of transcript 5′

ends, which pinpointed towards RNase Y activity. We repeated the sequence

analysis on the decay intermediate fragments (Paper III, Fig. S5) and showed

that RNase Y was responsible for the generation of some decay intermediate

fragment 5′ ends but not for the 3′ ends (Paper III, Figure 2C). Therefore,

PNPase degrades the decay intermediate fragment 3′ end generated by an

unidentified endoRNase until it reaches the fragment 5′ end produced by

RNase Y. This highlights that the interplay between PNPase and RNase Y is not

exclusive and that PNPase processes transcript 3′ ends that are generated by

other endoRNases.

Page 48: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

35

Is RNase Y the major RNase initiating RNA decay?

S. pyogenes RNase Y is involved in RNA decay, as illustrated by the interplay

with PNPase to degrade RNA fragments. Several facts support that RNase Y has

a minor role in this process and are developed below: the small targetome of

RNase Y, the absence of strong phenotype for ∆rny, and the presence of

RNase Y-independent but PNPase-dependent degradative pathway.

First, with the addition of putative processing positions of RNase Y found in

∆pnpA (336 transcript 5′ ends with a small conservation of the G, Paper II), the

total number of RNase Y processing positions that we detected in these

conditions would not be above 500. The S. pyogenes strain SF370 used in our

studies has a genome of 1.85 Mb and 1801 genes (262, 263). Thus, the RNase Y

targetome represents a small fraction of processed transcripts, as described for

B. subtilis and S. aureus (51, 66). This is opposite to the 22000 processing

positions published for RNase E, the major initiator of mRNA decay, in S.

enterica (240). Secondly, the deletion of RNase Y mildly affected the bacterial

growth (Paper III, Figure S3). We observed an absence of hemolysis on agar

blood plates and of protease activity on silk-milk plates (it has been shown to be

due to SpeB (60)), but no susceptibility to several antibiotics, no susceptibility

to osmotic stress, no differences in metabolic reactions with Api 20 Strep

(Biomérieux) were detected. This is opposite to the severe phenotypes observed

in B. subtilis (107) and to the essentiality of RNase E in Gram-negative bacteria

(202). Thirdly, the size of the fragments degraded by PNPase (120 nt on

average) indicates that transcripts are processed multiple times. The

conservation of sequence around the fragments degraded by PNPase (Paper III,

Figure S5) suggested that other RNases were involved. Thus, PNPase degrades

transcripts in an RNase Y-independent manner. This was supported by the

growth of the double deletion mutant ∆rny_∆pnpA, which is slower than the

single deletion mutants ∆rny or ∆pnpA (Paper II, SI appendix, Fig. S9; to

compare with Paper III, Figure S3).

Taken together, our data supports the existence of a degradation pathway

initiated by RNase Y and involving PNPase for the final degradation steps.

However, this pathway does not seem central in mRNA decay when bacteria are

Page 49: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

36

grown under standard laboratory conditions. The study of a different S.

pyogenes strain (NZ131) grown in C-medium, which is a peptide-rich and

carbon-poor medium that mimics host environment during deep tissue invasion

(280, 281), revealed that the bulk RNA half-life was increased by 2 fold in

absence of RNase Y (50). It is possible that RNase Y has a more prominent role

under other conditions.

The fate of transcript 5′ ends produced by RNase Y

The main finding of our study is the different fate of the transcript 3′ ends

and 5′ ends produced by RNase Y. On one side, we showed that most of the

transcript 3′ ends are further trimmed by 3′-to-5′ exoRNases and on the other

side, we observed that 87.4% of the transcript 5′ ends corresponded to the

original RNase Y processing positions. Therefore, these transcript 5′ ends are

not subsequently partially trimmed by RNase J1, which would give observable

RNase J1 trimming stop positions. However, since 107 transcript 3′ ends were

not paired with their corresponding downstream transcript 5′ ends, a number of

transcript 5′ ends were not detected in our study. A part of these transcript 5′

ends were degraded by PNPase, which started at the downstream 3′ end until it

reached the 5′ end produced by RNase Y (Paper III, Figure 2D). We assume that

RNase J1 is also involved in degrading these transcript 5′ ends. The interplay

between RNase Y and RNase J1 was indeed observed in vivo in B. subtilis and S.

aureus (40, 51). An interesting observation was that either the transcript 5′ ends

were original RNase Y processing positions or they were not detected, i.e.

degraded to completion. The signals deciding the different fates of these

transcript 5′ ends should be further investigated.

The interplay between RNase Y and RNase J1 does not seem as broad as the

interplay between RNase Y and PNPase. S. pyogenes RNases J1 and J2 are

essential (110) and the fact that the deletion of RNase Y is not lethal suggests

that RNase J1 and/or RNase J2 perform RNA decay mainly in an RNase Y-

independent manner. To this date, we have been unable to obtain the single

deletions of RNase J1, RNase J2 and the double deletion of PNPase and RNase

R in S. pyogenes, suggesting that these deletions are lethal. This highlights that

Page 50: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

37

both 3′-to-5′ degradation and 5′-to-3′ degradation pathways are vital for S.

pyogenes. Whether it is due to a global role in one particular cellular process

(such as RNA decay) or to the maturation or degradation of a specific target is

currently unknown.

Page 51: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

38

IV- Main findings of the thesis

The aim of this thesis was to investigate the poorly characterized post-

transcriptional regulation mediated by RNases in S. pyogenes. A method was

developed to identify the specific in vivo targetomes of endoRNases and

exoRNases. It was applied to study the roles of RNase Y, RNase III, YhaM,

PNPase, and RNase R in S. pyogenes, as well as the interplay between these

RNases. The three papers included in this thesis led to the conclusions

presented below.

We reported a general in vivo nicking activity of RNase III and a broad

nibbling activity of YhaM, whose impact on the regulation of gene expression in

S. pyogenes is currently unknown.

PNPase is the major 3′-to-5′ exoRNase responsible for RNA decay in S.

pyogenes. Our results additionally suggest a redundant role between PNPase

and RNase R in this process. However, while PNPase compensated the loss of

RNase R, the reverse was only partial.

S. pyogenes RNase Y processes transcripts downstream of a G and initiates

the subsequent degradation of the upstream fragment by 3′-to-5′ exoRNases.

These fragments can be fully or partially degraded. The downstream fragments

are either stable or fully degraded, as we did not observe partial degradation.

RNase Y is not a major player in S. pyogenes mRNA decay in standard

laboratory conditions. Instead, S. pyogenes, like other Gram-positive bacteria,

seems to rely on several endoRNases to initiate mRNA decay, opposite to E. coli

that relies principally on RNase E.

In conclusion, the method presented in this thesis is particularly suitable for

the study of sequential processing events performed by multiple RNases. Given

the importance of S. pyogenes as a human pathogen, it will be relevant to

extend the use of this method to study the role of these RNases in conditions

mimicking human infection.

Page 52: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

39

V- Acknowledgements

Many persons made this journey possible and unforgettable. It started in

Umeå seven years ago and I am glad to come back there for the final step (even

if it is in December).

I am grateful to my supervisor Emmanuelle Charpentier for the opportunity

she gave me to work in her lab, to develop my projects independently, to attend

international conferences, and for making me discover Berlin.

I am grateful to my co-supervisors and my PhD committee members Bernt

Eric Uhlin and Tracy Nissan, to my PhD committee member Jan Larsson, to

Sven Bergström for being my examiner and to the members of the BEU journal

club and of the RNA group for the fruitful conversations. In particular, Tracy,

thank you for including me in your group during my “lonely” years in Umeå, for

the lab meetings and the journal clubs “à l’américaine”, for the time you

invested in me, for your critics and your support and for your knowledge (I am

sure that your brain is more complete that my Endnote libraries will ever be). I

learnt so much from you during this time, you are a truly inspiring scientist!

Thanks to my third co-supervisor, AnaÏs, for your support, dedication, constant

diverging opinions and for improving my writing .

For their precious help to organize the defense from Berlin and for their fast

answers to any question, thank you Eva-Maria Diehl and Ulrich von Pawel-

Rammingen! Thank you Jörgen Johansson for your help when you were

responsible for the PhD students, and for the numerous “C’est la vie” at the

coffee machine. I am also very grateful to the persons working at the

administration of Mol Biol, MPIIB and MPUSP.

From my time in Umeå: the original lab members : Sophie, for constantly

mocking my French accent, which improved it (a bit); Geet, for the best

cheesecakes ever; Khan, for being the kindest person in the lab; and of course,

where is Shi Pey? Already gone to Berlin… The beer corner times will stay

forever in my memory, as much for the people, the discussions and the music as

for the affordable beers… Susanne, well, you know… thank you for all of that. I

remember the night when half of the EC lab left, I was really down. Then, truly

Page 53: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

40

amazing persons appeared in my life: Rosh, you are the best kind of crazy

person; Akbar: keep moving those hips! And keep the discussions about

everything and nothing; Ala: ma petite, I will always fight to keep the last bit,

even though I know I don’t stand a chance; Nabil: Exaaaaactlyyyyyy…; I love

you guys, you made Umeå the best place in the world. The lovely members of

the Spanish group and Vassili group, Ági, Mridula, Radha, Chinmay, Hélène,

Sveta, Sandrine, thank you for all the parties, outings, the curling, ultimate

Frisbee, etc… Thanks to the TAs during the fun/stressful teaching times.

From my time in Berlin: Laura, my PE buddy, my double loading gel partner,

it was so fun and motivating to work with you in the lab! Andres, there is

something I still want to say: “Yes, I did it with the radioactive substrate and it

did not work!!”; Karin, you are the best technician and such a sweet person;

Thibaud, for your constant flow of ideas, discussions and ideas (and

discussions) (and ideas..); Stefan, you are one of a kind, I am glad that I met

you!; Rina, thanks for introducing me to bouldering; Hagen, James, Majda,

Lina, Eric, Thomas, Frank, Katja, Sandra, Dior, Marlene, Vanessa, thank you for

the fun, the outings, Metallica concert, Croatian dinner, etc... I want to thank as

well the students who helped me: Franziska, Juan, Ann, and Quentin. And of

course all the member of the EC/RIIB/MPUSP lab! Lya and Lisa, thank you for

your kindness; Teresa, Paul, Sara, thank you for the strand bar, French-German

conversations and diners! Ines (there you are ), thank you for being my friend,

for being such a lovable person, thank you for being there when I needed it and

thank you for just being yourself… Andrey and your incessant flow of fun facts,

historical facts, and fabulous facts, thank you for being part of my life and a

great friend. Laurianne, thank you for all your caring, support and friendship!

Du fond de mon coeur, merci à ma famille pour son soutien: Maman, tu es l’une

des femmes les plus solides que je connaisse; Papa, tu me manques…; mes

frères Emmanuel et Christophe, pour être là, tout simplement; Odile, Jean-

Marc, Véro et le reste de la famille. Loïc, tu étais un grand soleil dans ma vie…

Mes amies de toujours: Anso, Laura, Titia, Aude, Titi, merci d’être là, même de

loin :)

Finally, thanks to all the musicians who put their souls in their music and

release it out there…

Page 54: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

41

References

1. Crick F (1970) Central dogma of molecular biology. Nature. doi:10.1038/227561a0.

2. Shapiro JA (2009) Revisiting the central dogma in the 21st century. Annals of the New York Academy of Sciences doi:10.1111/j.1749-6632.2009.04990.x.

3. Van Assche E, Van Puyvelde S, Vanderleyden J, Steenackers HP (2015) RNA-binding proteins involved in post-transcriptional regulation in bacteria. Front Microbiol 6:141.

4. Grundy FJ, Henkin TM (1993) tRNA as a positive regulator of transcription antitermination in B. subtilis. Cell. doi:10.1016/0092-8674(93)80049-K.

5. Epshtein V, Mironov AS, Nudler E (2003) The riboswitch-mediated control of sulfur metabolism in bacteria. Proc Natl Acad Sci. doi:10.1073/pnas.0531307100.

6. Johansson J, et al. (2002) An RNA thermosensor controls expression of virulence genes in Listeria monocytogenes. Cell. doi:10.1016/S0092-8674(02)00905-4.

7. Sherwood A V., Henkin TM (2016) Riboswitch-mediated gene regulation: Novel RNA architectures dictate gene expression responses. Annu Rev Microbiol. doi:10.1146/annurev-micro-091014-104306.

8. Kennell D (2002) Processing endoribonucleases and mRNA degradation in bacteria. J Bacteriol 184(17):4645–4657.

9. Newbury SF, Smith NH, Robinson EC, Hiles ID, Higgins CF (1987) Stabilization of translationally active mRNA by prokaryotic REP sequences. Cell 48(2):297–310.

10. Celesnik H, Deana A, Belasco JG (2007) Initiation of RNA decay in Escherichia coli by 5’ pyrophosphate removal. Mol Cell 27(1):79–90.

11. Richards J, et al. (2011) An RNA pyrophosphohydrolase triggers 5’-exonucleolytic degradation of mRNA in Bacillus subtilis. Mol Cell 43(6):940–949.

12. Frindert J, et al. (2018) Identification, biosynthesis, and decapping of NAD-capped RNAs in B. subtilis. Cell Rep 24(7):1890–1901.

13. Cahová H, Winz ML, Höfer K, Nübel G, Jäschke A (2015) NAD captureSeq indicates NAD as a bacterial cap for a subset of regulatory RNAs. Nature. doi:10.1038/nature14020.

14. Bird JG, et al. (2016) The mechanism of RNA 5′ capping with NAD+, NADH and desphospho-CoA. Nature. doi:10.1038/nature18622.

15. Piton J, et al. (2013) Bacillus subtilis RNA deprotection enzyme RppH recognizes guanosine in the second position of its substrates. Proc Natl Acad Sci U S A 110(22):8858–8863.

16. Durand S, Tomasini A, Braun F, Condon C, Romby P (2015) sRNA and mRNA turnover in Gram-positive bacteria. FEMS Microbiol Rev 39(3):316–330.

Page 55: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

42

17. Belasco JG, Beatty JT, Adams CW, von Gabain A, Cohen SN (1985) Differential expression of photosynthesis genes in R. capsulata results from segmental differences in stability within the polycistronic rxcA transcript. Cell 40(1):171–181.

18. McLaren RS, Newbury SF, Dance GS, Causton HC, Higgins CF (1991) mRNA degradation by processive 3’-5’ exoribonucleases in vitro and the implications for prokaryotic mRNA decay in vivo. J Mol Biol 221(1):81–95.

19. Emory SA, Bouvet P, Belasco JG (1992) A 5’-terminal stem-loop structure can stabilize mRNA in Escherichia coli. Genes Dev. doi:10.1101/gad.6.1.135.

20. Hambraeus G, Persson M, Rutberg B (2000) The aprE leader is a determinant of extreme mRNA stability in Bacillus subtilis. Microbiology. doi:10.1099/00221287-146-12-3051.

21. Sharp JS, Bechhofer DH (2005) Effect of 5’-proximal elements on decay of a model mRNA in Bacillus subtilis. Mol Microbiol 57(2):484–495.

22. Mohanty BK, Kushner SR (2016) Regulation of mRNA Decay in Bacteria. Annu Rev Microbiol 70:25–44.

23. Kaberdin VR, Blasi U (2013) Bacterial helicases in post-transcriptional control. Biochim Biophys Acta 1829(8):878–883.

24. O’Hara EB, et al. (1995) Polyadenylylation helps regulate mRNA decay in Escherichia coli. Proc Natl Acad Sci USA. doi:10.1073/pnas.92.6.1807.

25. Mohanty BK, Kushner SR (1999) Analysis of the function of Escherichia coli poly(A) polymerase I in RNA metabolism. Mol Microbiol. doi:10.1046/j.1365-2958.1999.01673.x.

26. Joanny G, et al. (2007) Polyadenylation of a functional mRNA controls gene expression in Escherichia coli. Nucleic Acids Res. doi:10.1093/nar/gkm120.

27. Folichon M, et al. (2003) The poly(A) binding protein Hfq protects RNA from RNase E and exoribonucleolytic degradation. Nucleic Acids Res. doi:10.1093/nar/gkg915.

28. Nogueira T, Springer M (2000) Post-transcriptional control by global regulators of gene expression in bacteria. Curr Opin Microbiol. doi:10.1016/S1369-5274(00)00068-0.

29. Braun F, Durand S, Condon C (2017) Initiating ribosomes and a 5’/3’-UTR interaction control ribonuclease action to tightly couple B. subtilis hbs mRNA stability with translation. Nucleic Acids Res 45(19):11386–11400.

30. Agaisse H, Lereclus D (1996) STAB-SD: A Shine-Dalgarno sequence in the 5′ untranslated region is a determinant of mRNA stability. Mol Microbiol. doi:10.1046/j.1365-2958.1996.5401046.x.

31. Hambraeus G, Karhumaa K, Rutberg B (2002) A 5′ stem-loop and ribosome binding but not translation are important for the stability of Bacillus subtilis aprE leader mRNA. Microbiology. doi:10.1099/00221287-148-6-1795.

32. Braun F, Le Derout J, Régnier P (1998) Ribosomes inhibit an RNase E cleavage which induces the decay of the rpsO mRNA of Escherichia coli. EMBO J. doi:10.1093/emboj/17.16.4790.

33. Deana A, Belasco JG (2005) Lost in translation: the influence of ribosomes on bacterial mRNA decay. Genes Dev 19(21):2526–2533.

Page 56: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

43

34. Anderson KL, Dunman PM (2009) Messenger RNA turnover processes in Escherichia coli, Bacillus subtilis, and emerging studies in Staphylococcus aureus. Int J Microbiol. doi: 10.1155/2009/525491.

35. Opdyke JA, Fozo EM, Hemm MR, Storz G (2011) RNase III participates in GadY-dependent cleavage of the gadX-gadW mRNA. J Mol Biol 406(1):29–43.

36. Brantl S (2012) Acting antisense: Plasmid- and chromosome-encoded sRNAs from Gram-positive bacteria. Future Microbiol. doi:10.2217/fmb.12.59.

37. Liu N, et al. (2015) The Streptococcus mutans irvA gene encodes a trans-acting riboregulatory mRNA. Mol Cell 57(1):179–190.

38. Durand S, Braun F, Helfer AC, Romby P, Condon C (2017) sRNA-mediated activation of gene expression by inhibition of 5’-3’ exonucleolytic mRNA degradation. Elife 6. doi:10.7554/eLife.23602.

39. Obana N, Nakamura K, Nomura N (2017) Role of RNase Y in Clostridium perfringens mRNA decay and processing. J Bacteriol 199(2). doi:10.1128/JB.00703-16.

40. Durand S, Gilet L, Condon C (2012) The essential function of B. subtilis RNase III is to silence foreign toxin genes. PLoS Genet. doi:10.1371/journal.pgen.1003181.

41. Morita T, Mochizuki Y, Aiba H (2006) Translational repression is sufficient for gene silencing by bacterial small noncoding RNAs in the absence of mRNA destruction. Proc Natl Acad Sci. doi:10.1073/pnas.0509638103.

42. Brantl S, Brückner R (2014) Small regulatory RNAs from low-GC Gram-positive bacteria. RNA Biol. doi:10.4161/rna.28036.

43. Deutscher MP (2006) Degradation of RNA in bacteria: comparison of mRNA and stable RNA. Nucleic Acids Res 34(2):659–666.

44. Gilet L, DiChiara JM, Figaro S, Bechhofer DH, Condon C (2015) Small stable RNA maturation and turnover in Bacillus subtilis. Mol Microbiol 95(2):270–282.

45. Shahbabian K, Jamalli A, Zig L, Putzer H (2009) RNase Y, a novel endoribonuclease, initiates riboswitch turnover in Bacillus subtilis. EMBO J 28(22):3523–3533.

46. Lehnik-Habrink M, et al. (2011) RNase Y in Bacillus subtilis: a natively disordered protein that is the functional equivalent of RNase E from Escherichia coli. J Bacteriol 193(19):5431–5441.

47. Durand S, Gilet L, Bessieres P, Nicolas P, Condon C (2012) Three essential ribonucleases-RNase Y, J1, and III-control the abundance of a majority of Bacillus subtilis mRNAs. PLoS Genet. doi: 10.1371/journal.pgen.1002520

48. Laalami S, et al. (2013) Bacillus subtilis RNase Y activity in vivo analysed by tiling microarrays. PLoS One. doi: 10.1371/journal.pone.0054062

49. Lehnik-Habrink M, et al. (2011) RNA processing in Bacillus subtilis: identification of targets of the essential RNase Y. Mol Microbiol 81(6):1459–1473.

50. Chen Z, Itzek A, Malke H, Ferretti JJ, Kreth J (2013) Multiple roles of RNase Y in Streptococcus pyogenes mRNA processing and degradation. J Bacteriol 195(11):2585–2594.

Page 57: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

44

51. Khemici V, Prados J, Linder P, Redder P (2015) Decay-initiating endoribonucleolytic cleavage by RNase Y is kept under tight control via sequence preference and sub-cellular localisation. PLoS Genet. doi:/10.1371/journal.pgen.1005577.

52. Marincola G, et al. (2012) RNase Y of Staphylococcus aureus and its role in the activation of virulence genes. Mol Microbiol 85(5):817–832.

53. Durand S, et al. (2015) A nitric oxide regulated small RNA controls expression of genes involved in redox homeostasis in Bacillus subtilis. PLoS Genet. doi: 10.1371/journal.pgen.1004957.

54. Deikus G, Bechhofer DH (2011) 5’ End-independent RNase J1 endonuclease cleavage of Bacillus subtilis model RNA. J Biol Chem 286(40):34932–34940.

55. Jahn N, Brantl S (2016) Heat shock induced refolding entails rapid degradation of bsrG toxin mRNA by RNases Y and J1. Microbiology. doi:10.1099/mic.0.000247.

56. Le Scornet A, Redder P (2018) Post-transcriptional control of virulence gene expression in Staphylococcus aureus. Biochim Biophys Acta. doi:10.1016/j.bbagrm.2018.04.004.

57. Kang SO, Caparon MG, Cho KH (2010) Virulence gene regulation by CvfA, a putative RNase: the CvfA-enolase complex in Streptococcus pyogenes links nutritional stress, growth-phase control, and virulence gene expression. Infect Immun 78(6):2754–2767.

58. Kang SO, et al. (2012) Thermoregulation of capsule production by Streptococcus pyogenes. PLoS One. doi: 10.1371/journal.pone.0037367.

59. Chen Z, Itzek A, Malke H, Ferretti JJ, Kreth J (2012) Dynamics of speB mRNA transcripts in Streptococcus pyogenes. J Bacteriol 194(6):1417–1426.

60. Broglia L, et al. (2018) RNase Y-mediated regulation of the streptococcal pyrogenic exotoxin B. RNA Biol. doi: 10.1080/15476286.2018.1532253.

61. Hunt A, Rawlins JP, Thomaides HB, Errington J (2006) Functional analysis of 11 putative essential genes in Bacillus subtilis. Microbiology 152:2895–2907.

62. Cascante-Estepa N, Gunka K, Stulke J (2016) Localization of components of the RNA-degrading machine in Bacillus subtilis. Front Microbiol 7:1492.

63. Koch G, et al. (2017) Attenuating Staphylococcus aureus Virulence by Targeting Flotillin Protein Scaffold Activity. Cell Chem Biol 24(7):845–857.

64. Chen Z, Mashburn-Warren L, Merritt J, Federle MJ, Kreth J (2017) Interference of a speB 5’ UTR partial deletion with mRNA degradation in Streptococcus pyogenes. Mol Oral Microbiol. doi:10.1111/omi.12181.

65. Marincola G, Wolz C (2017) Downstream element determines RNase Y cleavage of the saePQRS operon in Staphylococcus aureus. Nucleic Acids Res. doi:10.1093/nar/gkx296.

66. DeLoughery A, Lalanne JB, Losick R, Li GW (2018) Maturation of polycistronic mRNAs by the endoribonuclease RNase Y and its associated Y-complex in Bacillus subtilis. Proc Natl Acad Sci U S A. doi:10.1073/pnas.1803283115.

67. DeLoughery A, Dengler V, Chai Y, Losick R (2016) Biofilm formation by Bacillus subtilis requires an endoribonuclease-containing multisubunit complex that controls mRNA levels for the matrix gene repressor SinR. Mol Microbiol

Page 58: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

45

99(2):425–437.

68. Court DL, et al. (2013) RNase III: Genetics and function; structure and mechanism. Annu Rev Genet 47:405–431.

69. Robertson HD (1982) Escherichia coli ribonuclease III cleavage sites. Cell 30(3):669–672.

70. Calin-Jageman I, Nicholson AW (2003) RNA structure-dependent uncoupling of substrate recognition and cleavage by Escherichia coli ribonuclease III. Nucleic Acids Res 31(9):2381–2392.

71. Kim K, Sim SH, Jeon CO, Lee Y, Lee K (2011) Base substitutions at scissile bond sites are sufficient to alter RNA-binding and cleavage activity of RNase III. FEMS Microbiol Lett 315(1):30–37.

72. Nicholson AW (2014) Ribonuclease III mechanisms of double-stranded RNA cleavage. Wiley Interdiscip Rev RNA 5(1):31–48.

73. Panayotatos N, Truong K (1985) Cleavage within an RNase-III site can control messenger RNA stability and protein synthesis in vivo. Nucleic Acids Res 13(7):2227–2240.

74. Guarneros G (1988) Retroregulation of bacteriophage lambda int gene expression. Curr Top Microbiol Immunol 136:1–19.

75. Dasgupta S, et al. (1998) Genetic uncoupling of the dsRNA-binding and RNA cleavage activities of the Escherichia coli endoribonuclease RNase III - the effect of dsRNA binding on gene expression. Mol Microbiol 30(3):679.

76. Altuvia S, Locker-Giladi H, Koby S, Ben-Nun O, Oppenheim AB (1987) RNase III stimulates the translation of the cIII gene of bacteriophage lambda. Proc Natl Acad Sci U S A 84(18):6511–6515.

77. Altuvia S, Kornitzer D, Teff D, Oppenheim AB (1989) Alternative mRNA structures of the cIII gene of bacteriophage lambda determine the rate of its translation initiation. J Mol Biol 210(2):265–280.

78. Deutscher MP (2009) Maturation and degradation of ribosomal RNA in bacteria. Prog Mol Biol Transl Sci 85:369–391.

79. Stead MB, et al. (2011) Analysis of Escherichia coli RNase E and RNase III activity in vivo using tiling microarrays. Nucleic Acids Res 39(8):3188–3203.

80. DiChiara JM, Liu B, Figaro S, Condon C, Bechhofer DH (2016) Mapping of internal monophosphate 5’ ends of Bacillus subtilis messenger RNAs and ribosomal RNAs in wild-type and ribonuclease-mutant strains. Nucleic Acids Res. doi:10.1093/nar/gkw073.

81. Gordon GC, Cameron JC, Pfleger BF (2017) RNA sequencing identifies new RNase III cleavage sites in Escherichia coli and reveals increased regulation of mRNA. MBio 8(2). doi:10.1128/mBio.00128-17.

82. Lioliou E, et al. (2012) Global regulatory functions of the Staphylococcus aureus endoribonuclease III in gene expression. PLoS Genet. doi:10.1371/journal.pgen.1002782.

83. Lasa I, et al. (2011) Genome-wide antisense transcription drives mRNA processing in bacteria. Proc Natl Acad Sci U S A 108(50):20172–20177.

84. Lybecker M, Zimmermann B, Bilusic I, Tukhtubaeva N, Schroeder R (2014) The

Page 59: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

46

double-stranded transcriptome of Escherichia coli. Proc Natl Acad Sci U S A 111(8):3134–3139.

85. Le Rhun A, Beer YY, Reimegård J, Chylinski K, Charpentier E (2016) RNA sequencing uncovers antisense RNAs and novel small RNAs in Streptococcus pyogenes. RNA Biol 13(2):177–195.

86. Romilly C, et al. (2012) Loop-loop interactions involved in antisense regulation are processed by the endoribonuclease III in Staphylococcus aureus. RNA Biol 9(12):1461-72.

87. Deltcheva E, et al. (2011) CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. doi:10.1038/nature09886.

88. Wilson HR, Yu D, Peters 3rd HK, Zhou JG, Court DL (2002) The global regulator RNase III modulates translation repression by the transcription elongation factor N. EMBO J 21(15):4154–4161.

89. Bardwell JC, et al. (1989) Autoregulation of RNase III operon by mRNA processing. EMBO J 8(11):3401–3407.

90. Xu W, Huang J, Cohen SN (2008) Autoregulation of absB (RNase III) expression in Streptomyces coelicolor by endoribonucleolytic cleavage of absB operon transcripts. J Bacteriol. doi:10.1128/JB.00558-08.

91. Kim KS, Manasherob R, Cohen SN (2008) YmdB: A stress-responsive ribonuclease-binding regulator of E. coli RNase III activity. Genes Dev. doi:10.1101/gad.1729508.

92. Paudyal S, et al. (2015) Combined computational and experimental analysis of a complex of ribonuclease III and the regulatory macrodomain protein, YmdB. Proteins 83(3):459–472.

93. Mayer JE, Schweiger M (1983) RNase III is positively regulated by T7 protein kinase. J Biol Chem. 258(9):5340-3.

94. Mathy N, et al. (2007) 5’-to-3’ exoribonuclease activity in bacteria: role of RNase J1 in rRNA maturation and 5’ stability of mRNA. Cell 129(4):681–692.

95. Even S, et al. (2005) Ribonucleases J1 and J2: two novel endoribonucleases in B. subtilis with functional homology to E. coli RNase E. Nucleic Acids Res 33(7):2141–2152.

96. Condon C (2010) What is the role of RNase J in mRNA turnover? RNA Biol 7(3):316–321.

97. Durand S, Condon C (2018) RNases and helicases in Gram-positive bacteria. Microbiol Spectr 6(2). doi:10.1128/microbiolspec.RWR-0003-2017.

98. Mathy N, et al. (2010) Bacillus subtilis ribonucleases J1 and J2 form a complex with altered enzyme behaviour. Mol Microbiol 75(2):489–498.

99. Mader U, Zig L, Kretschmer J, Homuth G, Putzer H (2008) mRNA processing by RNases J1 and J2 affects Bacillus subtilis gene expression on a global scale. Mol Microbiol 70(1):183–196.

100. Dorleans A, et al. (2011) Molecular basis for the recognition and cleavage of RNA by the bifunctional 5’-3’ exo/endoribonuclease RNase J. Structure 19(9):1252–1261.

101. Redko Y, et al. (2016) RNase J depletion leads to massive changes in mRNA

Page 60: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

47

abundance in Helicobacter pylori. RNA Biol. 13(2): 243–253.

102. Deikus G, Condon C, Bechhofer DH (2008) Role of Bacillus subtilis RNase J1 endonuclease and 5’-exonuclease activities in trp leader RNA turnover. J Biol Chem 283(25):17158–17167.

103. Yao S, Bechhofer DH (2010) Initiation of decay of Bacillus subtilis rpsO mRNA by endoribonuclease RNase Y. J Bacteriol 192(13):3279–3286.

104. Daou-Chabo R, Mathy N, Benard L, Condon C (2009) Ribosomes initiating translation of the hbs mRNA protect it from 5’-to-3’ exoribonucleolytic degradation by RNase J1. Mol Microbiol 71(6):1538–1550.

105. Hasenöhrl D, Konrat R, Bläsi U (2011) Identification of an RNase J ortholog in Sulfolobus solfataricus: Implications for 5′-to-3′ directional decay and 5′-end protection of mRNA in Crenarchaeota. RNA. doi:10.1261/rna.2418211.

106. Linder P, Lemeille S, Redder P (2014) Transcriptome-wide analyses of 5’-ends in RNase J mutants of a Gram-positive pathogen reveal a role in RNA maturation, regulation and degradation. PLoS Genet. doi: 10.1371/journal.pgen.1004207.

107. Figaro S, et al. (2013) Knockouts of the genes encoding ribonucleases RNase Y and J1 are viable in B. subtilis, with major defects in cell morphology, sporulation and competence. J Bacteriol. doi:10.1128/JB.00164-13.

108. Gao P, et al. (2017) Functional studies of E. faecalis RNase J2 and its role in virulence and fitness. PLoS One. doi: 10.1371/journal.pone.0175212.

109. Chen X, Liu N, Khajotia S, Qi F, Merritt J (2015) RNases J1 and J2 are critical pleiotropic regulators in Streptococcus mutans. Microbiology. doi:10.1099/mic.0.000039.

110. Bugrysheva J V, Scott JR (2010) The ribonucleases J1 and J2 are essential for growth and have independent roles in mRNA decay in Streptococcus pyogenes. Mol Microbiol 75(3):731–743.

111. Grunberg-Manago M, Ochoa S (1955) Enzymatic synthesis and breakdown of polynucleotides; polynucleotide phosphorylase. J Am Chem Soc. doi:10.1021/ja01616a093.

112. Grunberg-Manago M, Ortiz PJ, Ochoa S (1955) Enzymatic synthesis of nucleic acidlike polynucleotides. Science. doi:10.1126/science.122.3176.907.

113. Kinscherf TG, Apirion D (1975) Polynucleotide phosphorylase can participate in decay of mRNA in Escherichia coli in the absence of ribonuclease II. Mol Gen Genet 139(4):357–362.

114. Deutscher MP, Reuven NB (1991) Enzymatic basis for hydrolytic versus phosphorolytic mRNA degradation in Escherichia coli and Bacillus subtilis. Proc Natl Acad Sci U S A 88(8):3277–3280.

115. Mohanty BK, Kushner SR (2000) Polynucleotide phosphorylase functions both as a 3’-to-5’ exonuclease and a poly(A) polymerase in Escherichia coli. Proc Natl Acad Sci U S A 97(22):11966–11971.

116. Campos-Guillen J, Bralley P, Jones GH, Bechhofer DH, Olmedo-Alvarez G (2005) Addition of poly(A) and heteropolymeric 3’ ends in Bacillus subtilis wild-type and polynucleotide phosphorylase-deficient strains. J Bacteriol 187(14):4698–4706.

117. Lin-Chao S, Chiou NT, Schuster G (2007) The PNPase, exosome and RNA

Page 61: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

48

helicases as the building components of evolutionarily-conserved RNA degradation machines. J Biomed Sci 14(4):523–532.

118. Symmons MF, Jones GH, Luisi BF (2000) A duplicated fold is the structural basis for polynucleotide phosphorylase catalytic activity, processivity, and regulation. Structure 8(11):1215–1226.

119. Hardwick SW, Gubbey T, Hug I, Jenal U, Luisi BF (2012) Crystal structure of Caulobacter crescentus polynucleotide phosphorylase reveals a mechanism of RNA substrate channelling and RNA degradosome assembly. Open Biol. doi:10.1098/rsob.120028.

120. Spickler C, Mackie GA (2000) Action of RNase II and polynucleotide phosphorylase against RNAs containing stem-loops of defined structure. J Bacteriol 182(9):2422–2427.

121. Fazal FM, Koslover DJ, Luisi BF, Block SM (2015) Direct observation of processive exoribonuclease motion using optical tweezers. Proc Natl Acad Sci U S A 112(49):15101–15106.

122. Deikus G, Bechhofer DH (2007) Initiation of decay of Bacillus subtilis trp leader RNA. J Biol Chem 282(28):20238–20244.

123. Liu B, Kearns DB, Bechhofer DH (2016) Expression of multiple Bacillus subtilis genes is controlled by decay of slrA mRNA from Rho-dependent 3’ ends. Nucleic Acids Res 44(7):3364–3372.

124. Coburn GA, Miao X, Briant DJ, Mackie GA (1999) Reconstitution of a minimal RNA degradosome demonstrates functional coordination between a 3’ exonuclease and a DEAD-box RNA helicase. Genes Dev 13(19):2594–2603.

125. Blum E, Carpousis AJ, Higgins CF (1999) Polyadenylation promotes degradation of 3’-structured RNA by the Escherichia coli mRNA degradosome in vitro. J Biol Chem 274(7):4009–4016.

126. Khemici V, Carpousis AJ (2004) The RNA degradosome and poly(A) polymerase of Escherichia coli are required in vivo for the degradation of small mRNA decay intermediates containing REP-stabilizers. Mol Microbiol 51(3):777–790.

127. Mohanty BK, Kushner SR (2011) Bacterial/archaeal/organellar polyadenylation. Wiley Interdiscip Rev RNA 2(2):256–276.

128. Chen X, et al. (2013) An RNA degradation machine sculpted by Ro autoantigen and noncoding RNA. Cell 153(1):166–177.

129. Huen J, et al. (2017) Structural insights into a unique dimeric DEAD-Box helicase CshA that promotes RNA decay. Structure 25(3):469–481.

130. Liou GG, Chang HY, Lin CS, Lin-Chao S (2002) DEAD box RhlB RNA helicase physically associates with exoribonuclease PNPase to degrade double-stranded RNA independent of the degradosome-assembling region of RNase E. J Biol Chem 277(43):41157–41162.

131. Liu B, et al. (2014) Global analysis of mRNA decay intermediates in Bacillus subtilis wild-type and polynucleotide phosphorylase-deletion strains. Mol Microbiol. doi:10.1111/mmi.12748.

132. Donovan WP, Kushner SR (1986) Polynucleotide phosphorylase and ribonuclease II are required for cell viability and mRNA turnover in Escherichia coli K-12. Proc Natl Acad Sci U S A 83(1):120–124.

Page 62: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

49

133. Mohanty BK, Kushner SR (2003) Genomic analysis in Escherichia coli demonstrates differential roles for polynucleotide phosphorylase and RNase II in mRNA abundance and decay. Mol Microbiol 50(2):645–658.

134. Wang W, Bechhofer DH (1996) Properties of a Bacillus subtilis polynucleotide phosphorylase deletion strain. J Bacteriol 178(8):2375–2382.

135. Braun F, Hajnsdorf E, Regnier P (1996) Polynucleotide phosphorylase is required for the rapid degradation of the RNase E-processed rpsO mRNA of Escherichia coli devoid of its 3’ hairpin. Mol Microbiol 19(5):997–1005.

136. Oussenko IA, Abe T, Ujiie H, Muto A, Bechhofer DH (2005) Participation of 3’-to-5’ exoribonucleases in the turnover of Bacillus subtilis mRNA. J Bacteriol 187(8):2758–2767.

137. Bernstein JA, Lin P-H, Cohen SN, Lin-Chao S (2004) Global analysis of Escherichia coli RNA degradosome function using DNA microarrays. Proc Natl Acad Sci. doi:10.1073/pnas.0308747101.

138. Carpousis AJ, Van Houwe G, Ehretsmann C, Krisch HM (1994) Copurification of E. coli RNase E and PNPase: Evidence for a specific association between two enzymes important in RNA processing and degradation. Cell. doi:10.1016/0092-8674(94)90363-8.

139. Cameron TA, De Lay NR (2016) The phosphorolytic exoribonucleases polynucleotide phosphorylase and RNase PH stabilize sRNAs and facilitate regulation of their mRNA targets. J Bacteriol 198(24):3309–3317.

140. Bandyra KJ, Sinha D, Syrjanen J, Luisi BF, De Lay NR (2016) The ribonuclease polynucleotide phosphorylase can interact with small regulatory RNAs in both protective and degradative modes. RNA 22(3):360–372.

141. De Lay N, Gottesman S (2011) Role of polynucleotide phosphorylase in sRNA function in Escherichia coli. RNA 17(6):1172–1189.

142. Andrade JM, Pobre V, Matos AM, Arraiano CM (2012) The crucial role of PNPase in the degradation of small RNAs that are not associated with Hfq. RNA 18(4):844–855.

143. Sesto N, et al. (2014) A PNPase dependent CRISPR System in Listeria. PLoS Genet. doi: 10.1371/journal.pgen.1004065.

144. Müller P, et al. (2016) A multistress responsive type I toxin-antitoxin system: bsrE/SR5 from the B. subtilis chromosome. RNA Biol. doi:10.1080/15476286.2016.1156288.

145. Deikus G, Babitzke P, Bechhofer DH (2004) Recycling of a regulatory protein by degradation of the RNA to which it binds. Proc Natl Acad Sci U S A 101(9):2747–2751.

146. Deikus G, Bechhofer DH (2009) Bacillus subtilis trp Leader RNA: RNase J1 endonuclease cleavage specificity and PNPase processing. J Biol Chem 284(39):26394–26401.

147. Briani F, Carzaniga T, Deho G (2016) Regulation and functions of bacterial PNPase. Wiley Interdiscip Rev RNA 7(2):241–258.

148. Robertlemeur M, Portier C (1992) Escherichia coli polynucleotide phosphorylase expression is autoregulated through an RNase-III-dependent mechanism. Embo J 11(7):2633–2641.

Page 63: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

50

149. Jarrige AC, Mathy N, Portier C (2001) PNPase autocontrols its expression by degrading a double-stranded structure in the pnp mRNA leader. EMBO J 20(23):6845–6855.

150. Park H, Yakhnin H, Connolly M, Romeo T, Babitzke P (2015) CsrA participates in a PNPase autoregulatory mechanism by selectively repressing translation of pnp transcripts that have been previously processed by RNase III and PNPase. J Bacteriol 197(24):3751–3759.

151. Fontaine F, et al. (2016) The small RNA SraG participates in PNPase homeostasis. RNA 22(10):1560–1573.

152. Carzaniga T, Deho G, Briani F (2015) RNase III-independent autogenous regulation of Escherichia coli polynucleotide phosphorylase via translational repression. J Bacteriol 197(11):1931–1938.

153. Del Favero M, et al. (2008) Regulation of Escherichia coli polynucleotide phosphorylase by ATP. J Biol Chem. doi:10.1074/jbc.C800113200.

154. Nurmohamed S, et al. (2011) Polynucleotide phosphorylase activity may be modulated by metabolites in Escherichia coli. J Biol Chem. doi:10.1074/jbc.M110.200741.

155. Siculella L, et al. (2010) Guanosine 5’-diphosphate 3’-diphosphate (ppGpp) as a negative modulator of polynucleotide phosphorylase activity in a “rare” actinomycete. Mol Microbiol 77(3):716–729.

156. Zangrossi S, et al. (2000) Transcriptional and post-transcriptional control of polynucleotide phosphorylase during cold acclimation in Escherichia coli. Mol Microbiol 36(6):1470–1480.

157. Mathy N, Jarrige AC, Robert-Le Meur M, Portier C (2001) Increased expression of Escherichia coli polynucleotide phosphorylase at low temperatures is linked to a decrease in the efficiency of autocontrol. J Bacteriol 183(13):3848–3854.

158. Jones PG, VanBogelen RA, Neidhardt FC (1987) Induction of proteins in response to low temperature in Escherichia coli. J Bacteriol 169(5):2092–2095.

159. Barnett TC, Bugrysheva J V, Scott JR (2007) Role of mRNA stability in growth phase regulation of gene expression in the Group A Streptococcus. J Bacteriol 189(5):1866–1873.

160. Zuo Y, Deutscher MP (2001) Exoribonuclease superfamilies: structural analysis and phylogenetic distribution. Nucleic Acids Res 29(5):1017–1026.

161. Carpousis AJ, Luisi BF, McDowall KJ (2009) Endonucleolytic initiation of mRNA decay in Escherichia coli. Prog Mol Biol Transl Sci 85:91–135.

162. Awano N, et al. (2010) Escherichia coli RNase R has dual activities, helicase and RNase. J Bacteriol 192(5):1344–1352.

163. Cheng ZF, Deutscher MP (2002) Purification and characterization of the Escherichia coli exoribonuclease RNase R. Comparison with RNase II. J Biol Chem 277(24):21624–21629.

164. Lee G, Bratkowski MA, Ding F, Ke A, Ha T (2012) Elastic coupling between RNA degradation and unwinding by an exoribonuclease. Science. 336(6089):1726–1729.

165. Chu LY, et al. (2017) Structural insights into RNA unwinding and degradation by RNase R. Nucleic Acids Res 45(20):12015–12024.

Page 64: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

51

166. Oussenko IA, Bechhofer DH (2000) The yvaJ gene of Bacillus subtilis encodes a 3’-to-5’ exoribonuclease and is not essential in a strain lacking polynucleotide phosphorylase. J Bacteriol 182(9):2639–2642.

167. Cheng ZF, Deutscher MP (2005) An important role for RNase R in mRNA decay. Mol Cell 17(2):313–318.

168. Vincent HA, Deutscher MP (2006) Substrate recognition and catalysis by the exoribonuclease RNase R. J Biol Chem 281(40):29769–29775.

169. Cheng ZF, Deutscher MP (2003) Quality control of ribosomal RNA mediated by polynucleotide phosphorylase and RNase R. Proc Natl Acad Sci USA 100(11):6388–6393.

170. Sulthana S, Deutscher MP (2013) Multiple exoribonucleases catalyze maturation of the 3’ terminus of 16S ribosomal RNA (rRNA). J Biol Chem 288(18):12574–12579.

171. Baumgardt K, Gilet L, Figaro S, Condon C (2018) The essential nature of YqfG, a YbeY homologue required for 3’ maturation of Bacillus subtilis 16S ribosomal RNA is suppressed by deletion of RNase R. Nucleic Acids Res. doi:10.1093/nar/gky488.

172. Cairrao F, Cruz A, Mori H, Arraiano CM (2003) Cold shock induction of RNase R and its role in the maturation of the quality control mediator SsrA/tmRNA. Mol Microbiol 50(4):1349–1360.

173. Richards J, Mehta P, Karzai AW (2006) RNase R degrades non-stop mRNAs selectively in an SmpB-tmRNA-dependent manner. Mol Microbiol 62(6):1700–1712.

174. Andrade JM, Cairrao F, Arraiano CM (2006) RNase R affects gene expression in stationary phase: regulation of ompA. Mol Microbiol 60(1):219–228.

175. Andrade JM, Hajnsdorf E, Regnier P, Arraiano CM (2009) The poly(A)-dependent degradation pathway of rpsO mRNA is primarily mediated by RNase R. RNA 15(2):316–326.

176. Pobre V, Arraiano CM (2015) Next generation sequencing analysis reveals that the ribonucleases RNase II, RNase R and PNPase affect bacterial motility and biofilm formation in E. coli. BMC Genomics 16. doi:10.1186/s12864-015-1237-6.

177. Tsao MY, Lin TL, Hsieh PF, Wang JT (2009) The 3’-to-5’ exoribonuclease (encoded by HP1248) of Helicobacter pylori regulates motility and apoptosis-inducing genes. J Bacteriol 191(8):2691–2702.

178. Fonseca P, Moreno R, Rojo F (2008) Genomic analysis of the role of RNase R in the turnover of Pseudomonas putida mRNAs. J Bacteriol 190(18):6258–6263.

179. Oussenko IA, Sanchez R, Bechhofer DH (2002) Bacillus subtilis YhaM, a member of a new family of 3’-to-5’ exonucleases in Gram-positive bacteria. J Bacteriol 184(22):6250–6259.

180. Zhang Y, et al. (2018) A stress response that monitors and regulates mRNA structure is central to cold shock adaptation. Mol Cell. doi:10.1016/j.molcel.2018.02.035.

181. Chen C, Deutscher MP (2005) Elevation of RNase R in response to multiple stress conditions. J Biol Chem 280(41):34393–34396.

182. Chen C, Deutscher MP (2010) RNase R is a highly unstable protein regulated by

Page 65: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

52

growth phase and stress. RNA 16(4):667–672.

183. Liang W, Deutscher MP (2012) Transfer-messenger RNA-SmpB protein regulates ribonuclease R turnover by promoting binding of HslUV and Lon proteases. J Biol Chem 287(40):33472–33479.

184. Liang W, Deutscher MP (2012) Post-translational modification of RNase R is regulated by stress-dependent reduction in the acetylating enzyme Pka (YfiQ). RNA 18(1):37–41.

185. Liang W, Malhotra A, Deutscher MP (2011) Acetylation regulates the stability of a bacterial protein: growth stage-dependent modification of RNase R. Mol Cell 44(1):160–166.

186. Liang W, Deutscher MP (2010) A novel mechanism for ribonuclease regulation: transfer-messenger RNA (tmRNA) and its associated protein SmpB regulate the stability of RNase R. J Biol Chem 285(38):29054–29058.

187. Liang W, Deutscher MP (2013) Ribosomes regulate the stability and action of the exoribonuclease RNase R. J Biol Chem 288(48):34791–34798.

188. Fang M, et al. (2009) Degradation of nanoRNA is performed by multiple redundant RNases in Bacillus subtilis. Nucleic Acids Res 37(15):5114–5125.

189. Redko Y, Condon C (2010) Maturation of 23S rRNA in Bacillus subtilis in the absence of Mini-III. J Bacteriol 192(1):356–359.

190. Zhang Q, Soares de Oliveira S, Colangeli R, Gennaro ML (1997) Binding of a novel host factor to the pT181 plasmid replication enhancer. J Bacteriol 179(3):684–688.

191. Noirot-Gros MF, et al. (2002) An expanded view of bacterial DNA replication. Proc Natl Acad Sci USA 99(12):8342–8347.

192. Au N, et al. (2005) Genetic composition of the Bacillus subtilis SOS system. J Bacteriol 187(22):7655–7666.

193. Gerth U, et al. (2008) Clp-dependent proteolysis down-regulates central metabolic pathways in glucose-starved Bacillus subtilis. J Bacteriol 190(1):321–331.

194. Eckart RA, Brantl S, Licht A (2009) Search for additional targets of the transcriptional regulator CcpN from Bacillus subtilis. FEMS Microbiol Lett 299(2):223–231.

195. Niyogi SK, Datta AK (1975) A novel oligoribonuclease of Escherichia coli. I. Isolation and properties. J Biol Chem 250(18):7307–7312.

196. Liao H, Liu M, Guo X (2018) The special existences: nanoRNA and nanoRNase. Microbiol Res 207:134–139.

197. Mechold U, Fang G, Ngo S, Ogryzko V, Danchin A (2007) YtqI from Bacillus subtilis has both oligoribonuclease and pAp-phosphatase activity. Nucleic Acids Res 35(13):4552–4561.

198. Murat D, Byrne M, Komeili A (2010) Cell biology of prokaryotic organelles. Cold Spring Harb Perspect Biol. doi:10.1101/cshperspect.a000422.

199. Lopez PJ, Marchand I, Joyce SA, Dreyfus M (1999) The C-terminal half of RNase E, which organizes the Escherichia coli degradosome, participates in mRNA degradation but not rRNA processing in vivo. Mol Microbiol.

Page 66: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

53

doi:10.1046/j.1365-2958.1999.01465.x.

200. Khemici V, Poljak L, Luisi BF, Carpousis AJ (2008) The RNase E of Escherichia coli is a membrane-binding protein. Mol Microbiol. doi:10.1111/j.1365-2958.2008.06454.x.

201. Strahl H, et al. (2015) Membrane recognition and dynamics of the RNA degradosome. PLoS Genet. doi: 10.1371/journal.pgen.1004961.

202. Goldblum K, Apririon D (1981) Inactivation of the ribonucleic acid-processing enzyme ribonuclease E blocks cell division. J Bacteriol 146(1):128–132.

203. Li Z, Deutscher MP (2002) RNase E plays an essential role in the maturation of Escherichia coli tRNA precursors. RNA 8(1):97–109.

204. Hammarlof DL, Bergman JM, Garmendia E, Hughes D (2015) Turnover of mRNAs is one of the essential functions of RNase E. Mol Microbiol. doi:10.1111/mmi.13100.

205. Himabindu P, Anupama K (2017) Decreased expression of stable RNA can alleviate the lethality associated with RNase E deficiency in Escherichia coli. J Bacteriol. doi:10.1128/jb.00724-16.

206. Mackie GA (2013) RNase E: at the interface of bacterial RNA processing and decay. Nat Rev Microbiol 11(1):45–57.

207. Ait-Bara S, Carpousis AJ (2015) RNA degradosomes in Bacteria and Chloroplasts: classification, distribution and evolution of RNase E homologs. Mol Microbiol. doi:10.1111/mmi.13095.

208. Redder P (2018) Molecular and genetic interactions of the RNA degradation machineries in Firmicute bacteria. Wiley Interdiscip Rev RNA. doi:10.1002/wrna.1460.

209. Cho KH (2017) The structure and function of the Gram-positive bacterial RNA degradosome. Front Microbiol 8:154.

210. Commichau FM, et al. (2009) Novel activities of glycolytic enzymes in Bacillus subtilis: interactions with essential proteins involved in mRNA processing. Mol Cell Proteomics 8(6):1350–1360.

211. Lehnik-Habrink M, et al. (2010) The RNA degradosome in Bacillus subtilis: identification of CshA as the major RNA helicase in the multiprotein complex. Mol Microbiol. doi:10.1111/j.1365-2958.2010.07264.x.

212. Roux CM, DeMuth JP, Dunman PM (2011) Characterization of components of the Staphylococcus aureus mRNA degradosome holoenzyme-like complex. J Bacteriol 193(19):5520–5526.

213. Newman JA, et al. (2012) Dissection of the network of interactions that links RNA processing with glycolysis in the Bacillus subtilis degradosome. J Mol Biol 416(1):121–136.

214. Gimpel M, Brantl S (2016) Dual-function sRNA encoded peptide SR1P modulates moonlighting activity of B. subtilis GapA. RNA Biol 13(9):916–926.

215. Redko Y, et al. (2013) A minimal bacterial RNase J-based degradosome is associated with translating ribosomes. Nucleic Acids Res 41(1):288–301.

216. Giraud C, et al. (2015) The C-terminal region of the RNA helicase CshA is required for the interaction with the degradosome and turnover of bulk RNA in

Page 67: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

54

the opportunistic pathogen Staphylococcus aureus. RNA Biol 12(6):658–674.

217. Raj R, Mitra S, Gopal B (2018) Characterization of Staphylococcus epidermidis Polynucleotide phosphorylase and its interactions with ribonucleases RNase J1 and RNase J2. Biochem Biophys Res Commun 495(2):2078–2084.

218. Apirion D (1973) Degradation of RNA in Escherichia coli. A hypothesis. Mol Gen Genet 122(4):313–322.

219. Hui MP, Foley PL, Belasco JG (2014) Messenger RNA degradation in bacterial cells. Annu Rev Genet 48:537–559.

220. Deana A, Celesnik H, Belasco JG (2008) The bacterial enzyme RppH triggers messenger RNA degradation by 5’ pyrophosphate removal. Nature 451(7176):355–358.

221. Luciano DJ, Vasilyev N, Richards J, Serganov A, Belasco JG (2017) A novel RNA phosphorylation state enables 5’ end-dependent degradation in Escherichia coli. Mol Cell 67(1):44-54.

222. Luciano DJ, Vasilyev N, Richards J, Serganov A, Belasco JG (2018) Importance of a diphosphorylated intermediate for RppH-dependent RNA degradation. RNA Biol. doi: 10.1080/15476286.2018.1460995.

223. Mackie GA (1998) Ribonuclease E is a 5’-end-dependent endonuclease. Nature. doi:10.1038/27246.

224. Jiang X, Belasco JG (2004) Catalytic activation of multimeric RNase E and RNase G by 5’-monophosphorylated RNA. Proc Natl Acad Sci U S A. doi:10.1073/pnas.0401382101.

225. Clarke JE, Kime L, Romero AD, McDowall KJ (2014) Direct entry by RNase E is a major pathway for the degradation and processing of RNA in Escherichia coli. Nucleic Acids Res. doi:10.1093/nar/gku808.

226. Kime L, Jourdan SS, Stead JA, Hidalgo-Sastre A, McDowall KJ (2010) Rapid cleavage of RNA by RNase E in the absence of 5’ monophosphate stimulation. Mol Microbiol 76(3):590–604.

227. Kime L, Clarke JE, Romero A. D, Grasby JA, McDowall KJ (2014) Adjacent single-stranded regions mediate processing of tRNA precursors by RNase E direct entry. Nucleic Acids Res. doi:10.1093/nar/gkt1403.

228. Laalami S, Zig L, Putzer H (2014) Initiation of mRNA decay in bacteria. Cell Mol Life Sci 71(10):1799–1828.

229. Kaberdin VR, Singh D, Sue LC (2011) Composition and conservation of the mRNA-degrading machinery in bacteria. J Biomed Sci 18. doi:10.1186/1423-0127-18-23.

230. Croucher NJ, Thomson NR (2010) Studying bacterial transcriptomes using RNA-seq. Curr Opin Microbiol. doi:10.1016/j.mib.2010.09.009.

231. Sharma CM, Vogel J (2014) Differential RNA-seq: The approach behind and the biological insight gained. Curr Opin Microbiol. doi:10.1016/j.mib.2014.06.010.

232. Hör J, Gorski SA, Vogel J (2018) Bacterial RNA biology on a genome scale. Mol Cell. doi:10.1016/j.molcel.2017.12.023.

233. Dötsch A, et al. (2012) The Pseudomonas aeruginosa transcriptome in planktonic cultures and static biofilms using RNA sequencing. PLoS One.

Page 68: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

55

doi:10.1371/journal.pone.0031092.

234. Robinson MD, McCarthy DJ, Smyth GK (2009) edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. doi:10.1093/bioinformatics/btp616.

235. Love MI, Huber W, Anders S (2014) Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. doi:10.1186/s13059-014-0550-8.

236. Kristoffersen SM, et al. (2012) Global mRNA decay analysis at single nucleotide resolution reveals segmental and positional degradation patterns in a Gram-positive bacterium. Genome Biol. doi:10.1186/gb-2012-13-4-r30.

237. Chen H, Shiroguchi K, Ge H, Xie XS (2015) Genome-wide study of mRNA degradation and transcript elongation in Escherichia coli. Mol Syst Biol. doi:10.15252/msb.20145794.

238. Cooper DA, Jha BK, Silverman RH, Hesselberth JR, Barton DJ (2014) Ribonuclease L and metal-ion-independent endoribonuclease cleavage sites in host and viral RNAs. Nucleic Acids Res. doi:10.1093/nar/gku118.

239. Altuvia Y, et al. (2018) In vivo cleavage rules and target repertoire of RNase III in Escherichia coli. Nucleic Acids Res. doi:10.1093/nar/gky684.

240. Chao Y, et al. (2017) In vivo cleavage map illuminates the central role of RNase E in coding and non-coding RNA pathways. Mol Cell 65(1):39–51.

241. Schifano JM, et al. (2014) An RNA-seq method for defining endoribonuclease cleavage specificity identifies dual rRNA substrates for toxin MazF-mt3. Nat Commun. doi:10.1038/ncomms4538.

242. Dar D, Sorek R (2018) High-resolution RNA 3’-ends mapping of bacterial Rho-dependent transcripts. Nucleic Acids Res. doi:10.1093/nar/gky274.

243. Lalanne JB, et al. (2018) Evolutionary Convergence of Pathway-Specific Enzyme Expression Stoichiometry. Cell 173(3):749–761 e38.

244. Dar D, et al. (2016) Term-seq reveals abundant ribo-regulation of antibiotics resistance in bacteria. Science. doi:10.1126/science.aad9822.

245. Dar D, Prasse D, Schmitz RA, Sorek R (2016) Widespread formation of alternative 3’ UTR isoforms via transcription termination in archaea. Nat Microbiol. doi:10.1038/nmicrobiol.2016.143.

246. Dar D, Sorek R (2018) Extensive reshaping of bacterial operons by programmed mRNA decay. PLoS Genet. doi:10.1371/journal.pgen.1007354.

247. Bessen DE (2016) Molecular basis of serotyping and the underlying genetic organization of Streptococcus pyogenes doi:NBK333428 [bookaccession].

248. Ralph AP, Carapetis JR (2013) Group A Streptococcal diseases and their global burden. Curr Top Microbiol Immunol. doi:10.1007/82-2012-280.

249. Katzenell U, Shemer J, Bar-Dayan Y (2001) Streptococcal contamination of food: an unusual cause of epidemic pharyngitis. Epidemiol Infect. 127(2):179-84.

250. Walker MJ, et al. (2014) Disease manifestations and pathogenic mechanisms of Group A Streptococcus. Clin Microbiol Rev. doi:10.1128/CMR.00101-13.

251. Vos T, et al. (2016) Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic

Page 69: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

56

analysis for the Global Burden of Disease Study 2015. Lancet. doi:10.1016/S0140-6736(16)31678-6.

252. Carapetis JR, Steer AC, Mulholland EK, Weber M (2005) The global burden of Group A Streptococcal diseases. Lancet Infect Dis. doi:10.1016/S1473-3099(05)70267-X.

253. Cattoir V (2016) Mechanisms of Antibiotic Resistance. Streptococcus Pyogenes : Basic Biology to Clinical Manifestations, eds Ferretti JJ, Stevens DL, Fischetti VA. doi: NBK333414 [bookaccession].

254. Spellerberg B, Brandt C (2016) Laboratory Diagnosis of Streptococcus pyogenes (Group A Streptococci). Streptococcus Pyogenes : Basic Biology to Clinical Manifestations, eds Ferretti JJ, Stevens DL, Fischetti VA. doi: NBK333414 [bookaccession].

255. Dale JB, Batzloff MR, Cleary PP, Courtney HS, Good MF, Grandi G, Halperin S, Margarit IY, McNeil S, Pandey M, Smeesters PR, Steer AC . (2016) Current Approaches to Group A Streptococcal Vaccine Development. Streptococcus Pyogenes: Basic Biology to Clinical Manifestations doi:NBK333413 [bookaccession].

256. Nobbs AH, Lamont RJ, Jenkinson HF (2009) Streptococcus adherence and colonization. Microbiol Mol Biol Rev. doi:10.1128/MMBR.00014-09.

257. Chiang-Ni C, Wu JJ (2008) Effects of streptococcal pyrogenic exotoxin B on pathogenesis of Streptococcus pyogenes. J Formos Med Assoc. doi:10.1016/S0929-6646(08)60112-6.

258. Nelson DC, Garbe J, Collin M (2011) Cysteine proteinase SpeB from Streptococcus pyogenes-A potent modifier of immunologically important host and bacterial proteins. Biol Chem. doi:10.1515/BC.2011.208.

259. Von Pawel-Rammingen U, Johansson BP, Björck L (2002) IdeS, a novel streptococcal cysteine proteinase with unique specificity for immunoglobulin G. EMBO J. doi:10.1093/emboj/21.7.1607.

260. Wessels MR (2016) Cell Wall and Surface Molecules: Capsule. Streptococcus Pyogenes : Basic Biology to Clinical Manifestations, eds Ferretti JJ, Stevens DL, Fischetti VA. doi: NBK333414 [bookaccession].

261. Molloy EM, Cotter PD, Hill C, Mitchell DA, Ross RP (2011) Streptolysin S-like virulence factors: The continuing sagA. Nat Rev Microbiol. doi:10.1038/nrmicro2624.

262. Ferretti JJ, et al. (2001) Complete genome sequence of an M1 strain of Streptococcus pyogenes. Proc Natl Acad Sci. doi:10.1073/pnas.071559398.

263. Nasser W, et al. (2014) Evolutionary pathway to increased virulence and epidemic Group A Streptococcus disease derived from 3,615 genome sequences. Proc Natl Acad Sci. doi:10.1073/pnas.1403138111.

264. Le Rhun A, Charpentier E (2012) Small RNAs in streptococci. RNA Biol. doi:10.4161/rna.20104.

265. Miller EW, Cao TN, Pflughoeft KJ, Sumby P (2014) RNA-mediated regulation in Gram-positive pathogens: an overview punctuated with examples from the Group A Streptococcus. Mol Microbiol 94(1):9–20.

266. Bugrysheva J V., Scott JR (2010) Regulation of virulence gene expression in Streptococcus pyogenes: Determinants of differential mRNA decay. RNA Biol.

Page 70: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

57

doi:10.4161/rna.7.5.13097.

267. Pertzev A V, Nicholson AW (2006) Characterization of RNA sequence determinants and antideterminants of processing reactivity for a minimal substrate of Escherichia coli ribonuclease III. Nucleic Acids Res 34(13):3708–3721.

268. Lalaouna D, Simoneau-Roy M, Lafontaine D, Masse E (2013) Regulatory RNAs and target mRNA decay in prokaryotes. Biochim Biophys Acta 1829(6–7):742–747.

269. Condon C, et al. (2008) Assay of Bacillus subtilis ribonucleases in vitro. Methods Enzym 447:277–308.

270. Coburn GA, Mackie GA (1996) Overexpression, purification, and properties of Escherichia coli ribonuclease II. J Biol Chem 271(2):1048–1053.

271. Hajnsdorf E, Steier O, Coscoy L, Teysset L, Regnier P (1994) Roles of RNase E, RNase II and PNPase in the degradation of the rpsO transcripts of Escherichia coli: stabilizing function of RNase II and evidence for efficient degradation in an ams pnp rnb mutant. EMBO J 13(14):3368–3377.

272. Coburn GA, Mackie GA (1998) Reconstitution of the degradation of the mRNA for ribosomal protein S20 with purified enzymes. J Mol Biol 279(5):1061–1074.

273. Tsou CC, et al. (2008) An iron-binding protein, Dpr, decreases hydrogen peroxide stress and protects Streptococcus pyogenes against multiple stresses. Infect Immun. doi:10.1128/IAI.00477-08.

274. Winkler WC, Breaker RR (2005) Regulation of bacterial gene expression by riboswitches. Annu Rev Microbiol. doi:10.1146/annurev.micro.59.030804.121336.

275. Le Rhun A, et al. (2017) Identification of endoribonuclease specific cleavage positions reveals novel targets of RNase III in Streptococcus pyogenes. Nucleic Acids Res 45(5):2329–2340.

276. Deutscher MP (2015) How bacterial cells keep ribonucleases under control. FEMS Microbiol Rev 39(3):350–361.

277. Cheng ZF, Zuo Y, Li Z, Rudd KE, Deutscher MP (1998) The vacB gene required for virulence in Shigella flexneri and Escherichia coli encodes the exoribonuclease RNase R. J Biol Chem 273(23):14077–14080.

278. Salvo E, Alabi S, Liu B, Schlessinger A, Bechhofer DH (2016) Interaction of Bacillus subtilis polynucleotide phosphorylase and RNase Y: structural mapping and effect on mRNA turnover. J Biol Chem. doi:10.1074/jbc.M115.711044.

279. Washburn RS, Marra A, Bryant AP, Rosenberg M, Gentry DR (2001) rho is not essential for viability or virulence in Staphylococcus aureus. Antimicrob Agents Chemother 45(4):1099–1103.

280. Lyon WR, Gibson CM, Caparon MG (1998) A role for Trigger Factor and an Rgg-like regulator in the transcription, secretion and processing of the cysteine proteinase of Streptococcus pyogenes. EMBO J. doi:10.1093/emboj/17.21.6263.

281. Valdes KM, et al. (2018) Glucose levels alter the Mga virulence regulon in the Group A Streptococcus. Sci Rep. doi:10.1038/s41598-018-23366-7.

Page 71: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)

58

Page 72: Post-transcriptional regulation by RNases in Streptococcus …umu.diva-portal.org/smash/get/diva2:1261772/FULLTEXT01.pdf · 2018-11-08 · Printed by: Print & Media (Umeå University)