141
REGULATION OF THE RNase E-MEDIATED TURNOVER OF NON-CODING sRNAs CsrB AND CsrC By YUANYUAN LENG A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2017

ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

REGULATION OF THE RNase E-MEDIATED TURNOVER OF NON-CODING sRNAs CsrB AND CsrC

By

YUANYUAN LENG

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2017

Page 2: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

© 2017 Yuanyuan Leng

Page 3: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

To my family and husband

Page 4: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

4

ACKNOWLEDGMENTS

I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving

me the opportunity to work in his lab and for his guidance, encouragement, care and

support during my PhD study. Under his excellent guidance, I have not only built good

skills and expertise in different areas, but also learned how to be a good scientist, to

think critically and independently, and to keep myself enthusiastic about research. I

truely appreciate his help to develop my presentation skills and his support during my

job search. I would also like to thank my committee members, Dr. Julie Maupin-Furlow,

Dr. Maurice Swanson, Dr. Wayne L. Nicholson and Dr. James Preston for their valuable

suggestions and comments in my research.

I am grateful to my colleagues in Dr. Romeo’s lab, especially Dr. Christopher

Vakulskas for his help and guidance when I joined the lab. I am also thankful to

Anastasia Potts for collaborating with me on the work in chapter 4 and for her help with

the RNA-seq library preparation and RNA-seq data analysis, to Dr. Archana Pannuri for

performing Northern blots to determine the effect of enolase on CsrB/C turnover in

chapter 3, to Dr. Tesfelam Zere for his help with Phos-tag gel, and to John Rice for his

help with lab supplies ordering. They have given me much help in my research studies

and a lot of smiles in my lab life. I want to thank Dr. Sixue Chen, Dr. Jin Koh and

Fanchao Zhu at the ICBR Proteomics and Mass Spectrometry Core for their support

with my research. I would like to thank my parents, who provide me endless support to

study abroad and encourage me when I encounter frustrations in work and life. Most

importantly, I wish to thank my loving and supportive husband, Likui Feng, for providing

unending inspiration and for always being there to help me.

Page 5: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

5

TABLE OF CONTENTS

page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 8

LIST OF FIGURES .......................................................................................................... 9

ABSTRACT ................................................................................................................... 11

CHAPTER

1 GENERAL INTRODUCTION .................................................................................. 13

Bacterial Gene Expression Regulation ................................................................... 13 Bacterial Transcriptional Regulation ................................................................. 13

Bacterial Posttranscriptional Regulation ........................................................... 14 Bacterial small regulatory RNAs ................................................................ 15 RNA binding proteins involved in gene expression regulation ................... 16

Turnover of sRNAs ................................................................................................. 17 Turnover of Base-pairing sRNAs ...................................................................... 18

Turnover of Protein Binding sRNAs .................................................................. 20 The Carbon Storage Regulator (Csr) System ......................................................... 21

CsrA/RsmA ....................................................................................................... 21

Regulation of the Csr System ........................................................................... 22

Transcription of CsrA Inhibitory sRNAs ............................................................ 23 Regulation of the Turnover of CsrA Inhibitory sRNAs ...................................... 24 CsrA Regulates its Own Expression ................................................................. 25

Interaction of Csr System with Other Regulatory Pathways ............................. 25 Project Rationale and Objectives ............................................................................ 27

2 REGULATION OF CsrB/C sRNA DECAY BY EIIAGlc ............................................. 30

Introduction ............................................................................................................. 30 Materials and Methods............................................................................................ 33

Bacterial Strains and Culture Conditions .......................................................... 33 Construction of Plasmids and Mutant Strains ................................................... 33 Expression and Purification of EIIAGlc and CsrD Variants ................................ 35

Northern Blotting .............................................................................................. 35 Western Blotting ............................................................................................... 36

Protein Pull-down Assays ................................................................................. 37 Determination of the EIIAGlc Phosphorylation State .......................................... 38 Gel Filtration Analysis of EIIAGlc in Complex with the EAL Domain or Other

CsrD Variants ................................................................................................ 38 Results .................................................................................................................... 39

EIIAGlc Activates CsrB/C Decay via CsrD ......................................................... 39

Page 6: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

6

In vitro Binding of EIIAGlc to CsrD Requires the EAL Domain ........................... 40

EIIAGlc Regulates CsrB/C Decay in a Phosphorylation-dependent Manner ...... 42

cAMP-Crp Modestly Represses CsrB Turnover ............................................... 43 CsrB/C Decay is Regulated in Response to Carbon Availability via the

Phosphorylation State of EIIAGlc .................................................................... 44 EIIAGlc and MshH Promote Csr sRNA Decay in Vibrio cholerae ....................... 45

Discussion .............................................................................................................. 46

3 EXPLORING THE MOLECULAR MECHANISM BY WHICH CsrD FACILITATES CsrB/C TURNOVER........................................................................ 61

Introduction ............................................................................................................. 61 Materials and Methods............................................................................................ 63

Media and Growth Conditions .......................................................................... 63

Construction of Strains and Plasmids ............................................................... 64 Gel Mobility Shift Assay .................................................................................... 64 In vitro RNase E Cleavage Assays ................................................................... 65

Affinity Purification of CsrD and its Binding RNAs ............................................ 65 Affinity Purification of in vivo Synthesized CsrB and its Associated Proteins ... 66

Mass Spectrometry .......................................................................................... 67 Protein Purification and Western Blots ............................................................. 69

RNA Purification and Northern Blots ................................................................ 69 Results and Discussion........................................................................................... 69

EIIAGlc is not Capable to Stimulate the Binding of CsrD to CsrB in vitro ........... 69 CsrA Influences the CsrD-CsrB Interaction in vitro ........................................... 70

CsrD cannot Facilitate CsrA-mediated Protection of CsrB Cleaved by RNase E ........................................................................................................ 70

CsrD is Unlikely to Bind CsrB in vivo ................................................................ 71

CsrB Associated Proteins and their Influence on CsrB Turnover ..................... 72 Conclusion .............................................................................................................. 75

4 EPISTASIS ANALYSIS USING RNA-SEQ (EPI-SEQ) TO EXPLORE THE REGULATORY ROLE OF CsrD ............................................................................. 84

Introduction ............................................................................................................. 84

Materials and Methods............................................................................................ 86 Media and Growth Conditions .......................................................................... 86 Construction of Strains and Plasmids ............................................................... 87 Glycogen Biosynthesis ..................................................................................... 87

RNA Extraction and Purification ....................................................................... 87 Northern Blotting .............................................................................................. 88 RNA-seq Library Preparation ........................................................................... 88

RNA-seq Data Analysis .................................................................................... 88 qRT-PCR .......................................................................................................... 89

Results and Discussion........................................................................................... 89 Construction and Characterization of Bacterial Strains .................................... 89

Page 7: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

7

CsrA Retains its Global Role in Regulating Transcript Levels in the Absence of CsrD .......................................................................................................... 91

CsrD Effects on Gene Expression Require CsrA ............................................. 93 CsrB/C Have Strong Effects on Gene Expression in the Absence of CsrD ...... 94 CsrD Regulates the Majority of its Target Genes in a CsrB/C Dependent

Manner .......................................................................................................... 94 Conclusion .............................................................................................................. 96

5 GENERAL DISCUSSION AND FUTURE PERSPECTIVES ................................. 104

APPENDIX

A SUPPLEMENTARY FIGURES ............................................................................. 109

B SUPPLEMENTARY TABLES ............................................................................... 110

LIST OF REFERENCES ............................................................................................. 124

BIOGRAPHICAL SKETCH .......................................................................................... 141

Page 8: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

8

LIST OF TABLES

Table page 2-1 Molecular weight of CsrD in solution .................................................................. 54

3-1 Proteins co-purifying with Strepto-CsrB identified from band A by mass-spectrometry ....................................................................................................... 82

3-2 Proteins co-purifying with Strepto-CsrB identified from band B by mass-spectrometry ....................................................................................................... 83

B-1 Bacterial Strains ............................................................................................... 110

B-2 Plasmids used in this study. ............................................................................. 113

B-3 Primers used in this study. ............................................................................... 115

B-4 Transcription regulators regulated by CsrA ...................................................... 118

B-5 sRNAs regulated by CsrA at the transcript level ............................................... 123

Page 9: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

9

LIST OF FIGURES

Figure page 1-1 Outline of the Csr system in E. coli (Vakulskas et al., 2015) .............................. 29

2-1 EIIAGlc affects CsrB/C decay rates and levels in E. coli ...................................... 50

2-2 EIIAGlc (crr) activates CsrB/C decay via CsrD, but does not enhance cellular CsrD levels ......................................................................................................... 51

2-3 EIIAGlc interacts specifically with the EAL domain of CsrD. ................................ 52

2-4 Gel filtration chromatography of CsrD variants ................................................... 53

2-5 CsrD binds only to unphosphorylated EIIAGlc in pull-down assays. .................... 54

2-6 Effects of cAMP-Crp on CsrB/C decay. .............................................................. 55

2-7 Effects of carbon sources on EIIAGlc phosphorylation and CsrB/C decay in minimal media .................................................................................................... 56

2-8 The phosphorylation state of EIIAGlc before and 10 min after shift from LB broth into minimal media. ................................................................................... 57

2-9 CsrB/C decay rates and levels after shift from LB to minimal media .................. 58

2-10 EIIAGlc and MshH (CsrD ortholog) affect CsrB and CsrD decay in V. cholerae.. ............................................................................................................ 59

2-11 Proposed model for the effect of carbon availability on CsrB/C decay. .............. 60

3-1 CsrB decay is regulated in response to carbon availability through its effect on CsrA and CsrD antagonism (Vakulskas et al., 2016).. ................................... 76

3-2 EIIAGlc has no effect on the binding of CsrD to CsrB RNA in vitro.. .................... 77

3-3 CsrA influences the CsrD-CsrB complex in vitro.. .............................................. 78

3-4 Effect of EIIAGlc and CsrD on RNase E-dependent cleavage of CsrB in vitro.. ... 79

3-5 CsrB is not copurified with FLAG-tagged CsrD. .................................................. 80

3-6 CsrD is not recovered by Strepto-CsrB in vivo ................................................... 81

3-7 Enolase has little or no effect on CsrB turnover.................................................. 83

4-1 Two proposed model depicting the epistatic relationship between CsrA and CsrD in regulating mRNA abundance.. ............................................................... 97

Page 10: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

10

4-2 Properties of bacterial strains used in this study.. ............................................... 98

4-3 CsrA retains its global role in regulating mRNA levels in the absence of CsrD. 100

4-4 Venn diagram depicting the overlap of differentially expressed genes induced by csrD deletion/overexpression in csrA WT and csrA mutant backgrounds.. .. 102

4-5 Venn diagram depicting the overlap of differentially expressed genes induced by csrB/C deletion or csrB overexpression in csrD WT and csrD mutant backgrounds. .................................................................................................... 102

4-6 CsrD regulate the majority of its target genes in a CsrB/C dependent manner. 103

A-1 EIIAGlc stimulates CsrB decay in the MG1655 csrDFLAG strain. ......................... 109

A-2 Decay of CsrB/C in a strain expressing EIIAGlc H91D is similar to that of EIIAGlc H91A ..................................................................................................... 109

A-3 CRP has minimal or no effect on CsrD protein levels. ...................................... 109

Page 11: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

11

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

REGULATION OF THE RNase E-MEDIATED TURNOVER OF NON-CODING sRNAs

CsrB AND CsrC

By

Yuanyuan Leng

August 2017

Chair: Tony Romeo Major: Microbiology and Cell Science

Bacteria utilize complex regulatory system to coordinate their gene expression

and make life style decisions in response to changing environment. Carbon Storage

Regulator (Csr) is a conserved global regulatory system in Gammaproteobacteria and it

uses the sequence-specific RNA-binding protein CsrA to activate or repress gene

expression at the posttranscriptional level. In Escherichia coli, CsrA activity is regulated

by two non-coding sRNAs, CsrB and CsrC, which bind to multiple CsrA dimers and

thereby sequester this protein away from its mRNA targets. Both the synthesis and

turnover of CsrB/C are regulated and play important roles in determining the steady

state levels of CsrB/C. Their turnover is initiated with RNase E cleavage, and the decay

intermediates are subsequently degraded by PNPase. This RNase E-mediated turnover

of CsrB/C requires CsrD protein although the exact molecular mechanism remains

unclear. In this study, we revealed the physiological role of CsrD in coupling CsrB/C

sRNA decay to the availability of a preferred carbon source. We demonstrated that

EIIAGlc of the glucose-specific PTS system regulates CsrB/C turnover in a

phosphorylation dependent manner and only the unphosphorylated form of EIIAGlc binds

to CsrD and is capable of activating CsrB/C turnover. On the other hand, the

Page 12: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

12

phosphorylated form of EIIAGlc indirectly and modestly represses CsrB turnover via

cAMP-Crp. In addition, we provided evidence that although CsrD binds to CsrB in vitro,

it does not appear to bind CsrB in vivo and it fails to facilitate the cleavage of CsrA-

protected CsrB by RNase E in vitro. These data suggest that CsrD might act indirectly

and require other unknown factor(s) to facilitate the CsrB/C turnover. Furthermore, the

global regulatory role of CsrD on gene expression was explored on a genome-wide

scale using epistasis analysis and RNA-seq. The transcriptomic data indicated that

CsrD mediates changes in gene expression primarily through its effects on CsrB/C

stability and thereby CsrA activity. Our data also implied that CsrD affects expression of

some genes through an alternative pathway independent of CsrB/C, suggestive of other

regulatory role(s) of CsrD in addition to affecting CsrB/C turnover.

Page 13: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

13

CHAPTER 1 GENERAL INTRODUCTION

Bacterial Gene Expression Regulation

Bacteria are constantly exposed to a variety of environmental stresses, such as

changes in temperature, pH, osmolarity and nutrient availability. To survive in the

changing environments, bacteria have developed complex regulatory systems to

coordinate their gene expression in response to cellular and environmental stimuli and

properly adjust cellular physiology and metabolism.

Bacterial Transcriptional Regulation

Expression of genes can be regulated at the transcriptional level by modulating

transcription initiation with alternative sigma factors (σ factors), transcription factors, or

small ligands (Browning & Busby, 2004, Balleza et al., 2009). Under stress conditions,

alternative σ factors, subunits of RNA polymerase (RNAP) holoenzyme, direct RNAP to

initiate transcription of specific gene sets related to stress responses. The best-studied

alternative σ factor in Escherichia coli is RpoS (Weber et al., 2005). It accumulates in

the cells entering stationary phase or during starvation and controls expression of up to

10% of the E. coli genes (Weber et al., 2005, Patten et al., 2004). This thereby triggers

a global stress response and allows the bacterial cells resistant to stress conditions.

Modulation of transcriptional gene expression during environmental shifts is also

mediated by a large set of transcription factors in bacteria (Browning & Busby, 2004).

By directly binding to promoters of target genes, transcription factors either activate or

repress target gene expression. E. coli possesses seven global transcription factors,

CRP, Fis, ArcA, Lrp, FNR, IHF and NarL, which control expression of numerous genes

Page 14: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

14

involved in complex cellular processes, whereas most other transcription factors govern

only a single promoter.

Small ligands provide another mechanism by which cells ensure proper gene

expression following external stimuli. (p)ppGpp, cyclic AMP (cAMP) and cyclic di-GMP

(c-di-GMP) are three common and versatile small ligands that bacteria utilize to control

various important biological processes, including motility, biofilm formation, carbon

transport and metabolism, virulence and stringent response (Battesti et al., 2011, Jenal

et al., 2017, Römling et al., 2013, Botsford & Harman, 1992). The concentrations of

these ligands fluctuate in response to different environmental stimuli, and they function

by modulating activity of their binding proteins, such as RNAP (ppGpp) (Dalebroux &

Swanson, 2012), CRP (cAMP) (Fic et al., 2009), effector proteins or riboswitches (c-di-

GMP) (Sudarsan et al., 2008, Hengge, 2009).

Bacterial Posttranscriptional Regulation

In addition to be regulated at the transcriptional level, bacterial gene expression

is controlled at the posttranscriptional level. In recent years, a number of new small

regulatory RNAs (sRNAs) and RNA-binding proteins (RBPs) have been identified in

bacteria as major posttranscriptional regulators (Gottesman & Storz, 2011, Storz et al.,

2011, Van Assche et al., 2015). These posttranscriptional regulators typically regulate

gene expression by altering translation initiation, transcription elongation and/or

transcript stability. In a few cases, mRNAs have also been described to have regulatory

functions (Dugar et al., 2016, Sterzenbach et al., 2013). For example, the leader region

of flaA mRNA encoding the major fimbriae in Campylobacter jejuni, acts as an mRNA-

derived RNA antagonist of CsrA, which is the global posttranscriptional regulatory

protein (Dugar et al., 2016).

Page 15: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

15

Bacterial small regulatory RNAs

Bacterial sRNAs range in length from approximately 50 to 500 nucleotides and

they play critical regulatory roles in almost every aspect of bacterial physiology, from

metabolism, virulence, biofilm formation to outer membrane synthesis (Gottesman &

Storz, 2011, Storz et al., 2011). The expression of bacterial sRNAs is often induced

under specific stress conditions, such as glucose starvation, glucose-phosphate stress,

iron deficiency and oxidative stress, and the mechanism of action of these sRNAs has

been extensively studied in recent years.

The majority of sRNAs act by base pairing with their target mRNAs with either

extended or limited complementarity. Pairing between sRNA and mRNA is often

incomplete and requires the presence of the RNA chaperone protein Hfq, which

facilitates base pairing by stabilizing sRNA-mRNA duplexes or affecting the secondary

structure of sRNA or mRNA (Vogel & Luisi, 2011). sRNAs mostly act as a negative

regulator of gene expression by blocking ribosome binding, which in turn inhibits

translation initiation, or by recruiting RNases to destabilize target mRNAs. In some

cases, sRNAs activate expression of target mRNAs by blocking mRNA cleavage or by

disrupting an inhibitory secondary structure that occludes ribosome binding.

In other cases, sRNAs exert their functions by mimicking the nucleic acid

substrates of proteins and thereby sequestering protein activity. So far, only a few

sRNAs of this class have been identified and characterized in bacteria. E. coli 6S RNA

contains a large central “loop” flanked by long double helical arms which mimics a open

DNA promoter complex. Thus, 6S RNA can bind tightly to the housekeeping form of

RNAP (σ70-RNAP) and inhibits its activity, leading to down-regulation of a subset of σ70–

dependent promoters (Wurm et al., 2010, Burenina et al., 2015). Another well known

Page 16: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

16

example is Csr/Rsm sRNAs, which contain many GGA sequences, allowing them to

bind CsrA/RsmA with high affinity, sequestering CsrA/RsmA from lower affinity target

mRNAs and antagonizing their activities (Fig. 1-1) (Romeo et al., 2013).

RNA binding proteins involved in gene expression regulation

Bacterial RNA binding proteins bind to target mRNAs and posttranscriptionally

regulate their expression by altering translation initiation, stability, and/or transcript

elongation. They exert their regulatory effects by modulating 1) the susceptibility of

mRNAs and/or sRNAs to RNases, 2) ribosome binding, 3) interaction of mRNA targets

with sRNA or proteins, and 4) formation of transcription terminator/antiterminator (Van

Assche et al., 2015, Vogel & Luisi, 2011, Romeo et al., 2013).

Two representative posttranscriptional regulatory proteins in bacteria are Hfq and

CsrA. Hfq is a widespread bacterial protein that resembles the eukaryotic Sm family of

proteins involved in splicing and mRNA degradation (Brennan & Link, 2007). Hfq forms

a stable ring-like, multimeric quaternary architecture that supports its interaction with

target RNAs. It affects translation initiation and transcript stability of target mRNAs by

facilitating the formation of stable sRNA-mRNA duplexes, such as Spot42-galK (Møller

et al., 2002), MicA-ompA (Johansen et al., 2006, Udekwu et al., 2005), and SgrS-ptsG

(Kawamoto et al., 2006). Thus, Hfq plays important roles in a variety of physiological

processes, from catabolite repression, envelope stress, biofilm formation, motility, metal

homeostasis to virulence (Holmqvist et al., 2016). CsrA is another major

posttranscriptional regulatory protein that contributes to complex posttranscriptional

networks (Romeo et al., 2013, Vakulskas et al., 2015). Its mechanism of action,

regulation and physiological roles will be discussed in a later section.

Page 17: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

17

In addition to Hfq and CsrA, other RNA binding proteins have been identified to

regulate expression of target mRNAs using distinct mechanisms, including RNA

helicases (Kaberdin & Bläsi, 2013, Vakulskas et al., 2014), ribonucleases (Saramago et

al., 2014), cold shock proteins CspA and CspE (Barria et al., 2013, Michaux et al.,

2017), S1 protein (Hajnsdorf & Boni, 2012), adaptor protein RapZ (Göpel et al., 2013),

and RNA chaperone ProQ (Smirnov et al., 2016, Chaulk et al., 2011). With the constant

development of biochemical techniques for the probing of direct protein-DNA/RNA

interactions, there is no doubt that new posttranscriptional regulatory RBPs will be

identified in the near future.

Turnover of sRNAs

sRNA levels must be tightly controlled to govern expression of target mRNAs or

activity of target proteins. Expression of many sRNA is extensively regulated by

transcription regulators, including global transcription factors Lrp, Crp and Fur (Chaulk

et al., 2011, Papenfort & Vogel, 2011, Thomason et al., 2012, Massé & Gottesman,

2002), σ factors RpoE and RpoS (Opdyke et al., 2004, Guo et al., 2014), two

component signal transduction systems (TCS) (Suzuki et al., 2002, Mandin &

Gottesman, 2010), and stringent response components (p)ppGpp and DksA (Edwards

et al., 2011). This extensive regulation ensures bacterial cells fine-tune sRNA levels and

global gene expression in response to different environmental stimuli. In recent years,

the turnover pathway of sRNAs has also been revealed to play important roles in

determing the steady state levels of sRNA (Saramago et al., 2014, Andrade et al.,

2012). Here, the RNases and RBPs involved in sRNA turnover are reviewed.

Page 18: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

18

Turnover of Base-pairing sRNAs

Pairing of sRNA and target mRNA often induces coupled degradation of the

sRNA-mRNA duplex, which mainly involves cleavage by RNase E and RNase III

(Saramago et al., 2014). RNase E is an essential endoribonuclease in Gram-negative

bacteria, which is responsible for bulk RNA turnover, and it prefers to cleave single-

stranded AU rich regions with adjacent stem-loop structures (Górna et al., 2012,

Mackie, 2013). It provides the scaffolding core of the RNA degradosome protein

complex, which also contains polynucleotide phosphorylase (PNPase), RNA helicase B

(RhlB), and the glycolytic enzyme enolase. RNase E generally recognizes substrates by

two mechansims (Mackie, 2013). The first one requires a 5’ monophosphate group on

the target transcript. By binding to the RNA 5’ end, the affinity of RNase E for the

transcript increases, which allostericall activates the activity of RNase E and facilitates

further cleavage of the transcript to the 3’ end (Prévost et al., 2011, Bandyra et al.,

2012). The other mechanism that RNase E utilizes is termed ‘direct entry’ and it

bypasses the 5’ end requirement. In this mechanism, RNase E binds close to the

internal cleavage site and cleaves the transcript (Baker & Mackie, 2003). RNAs that are

free of protein binding are more susceptible to the ‘direct entry’ cleavage (Göpel et al.,

2013). RNase E often acts together with Hfq to trigger the coupled degradation of

sRNA-mRNA duplexes (Basineni et al., 2009). Mostly, Hfq triggers RNA degradation by

recruiting RNase E and PNPase for cleavage (Sharma et al., 2007, Massé et al., 2003,

Göpel et al., 2013, Vogel & Luisi, 2011, Mohanty et al., 2004, Hankins et al., 2010). In

some cases, Hfq blocks the degradation of sRNA-mRNA duplex by competing with

RNase E for RNA binding (Moll et al., 2003). RNase E also degrades specific duplexes

independent of Hfq (Andrade et al., 2012, Lee & Groisman, 2010, Göpel et al., 2013). In

Page 19: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

19

the absence of Hfq, RNase E can be recruited by other adapter proteins to the target

sRNA (Göpel et al., 2013). For example, E. coli sRNA GlmZ binds and activates glmS

mRNA, which encodes glucosamine-6-phosphate (GlcN6P) synthase. The RNase E-

mediated degradation of GlmZ requires a specialized adaptor protein, RapZ. Under low

levels of GlcN6P, sRNA GlmY accumulates and antagonizes RapZ activity. This results

in stabilization of GlmZ and thereby synthesis of GlcN6P (Göpel et al., 2013).

RNase III is another important endoribonuclease responsible for the degradation

of paired sRNA-mRNA (Afonyushkin et al., 2005, Deltcheva et al., 2011). It specifically

recognizes and cleaves double-stranded (ds) RNA, and is able to cleave many sRNAs

in complex with their target mRNAs. For example, in Salmonella enterica serovar

Typhimurium, RNase III cleaves MicA, the negative regulator of outer membrane porins,

when bound to its target ompA mRNA (Viegas et al., 2011).

In a few cases, binding of sRNA to mRNA only promotes cleavage of mRNA but

not sRNA. One example is sRNA MicC. After degradation of its target ompD mRNA by

RNase E, MicC is released from the duplex and is still capable of pairing and inducing

cleavage of another ompD transcript (Bandyra et al., 2012).

Many sRNAs that are free of binding to the RNA chaperone Hfq and their target

mRNAs exist transiently in the cell. In addition to RNase E (Andrade et al., 2012, Lee &

Groisman, 2010, Göpel et al., 2013), PNPase is another key factor responsible for their

degradation (Andrade et al., 2012). When not associated with Hfq, the U rich region of

the 3’ end of sRNA is exposed and can be recognized by PNPase for rapid degradation

(Sauer & Weichenrieder, 2011, Andrade et al., 2012). PNPase was observed to make a

greater contribution than RNase E in degrading many of the Hfq-free sRNAs, including

Page 20: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

20

MicA, GlmY, RyhB, and SgrS, especially in stationary phase cells (Andrade et al.,

2012).

RNase J1/J2 and RNase Y are major factors in the processing and turnover of

RNAs in Gram-positive bacteria. Similar to RNase E, RNase J1/J2 specifically

recognize an AU-rich single-stranded RNA segments and prefer monophosphorylated 5’

ends (Even et al., 2005, Deikus & Bechhofer, 2009). However, their target sRNas have

not been explored much. A few sRNAs, such as RatA in Bacillus subtilis (Commichau &

Stülke, 2012) and RsaA and Sau63 in Staphylococcus aureus (Abu-Qatouseh et al.,

2010, Geissmann et al., 2009), are identified to be cleaved by RNase Y.

Turnover of Protein Binding sRNAs

Studies on the degradation of CsrA/RsmA inhibitory sRNAs have been limited to

only a few species. E. coli CsrB/C turnover is mediated by RNase E and PNPase and

facilitated by CsrD protein (Suzuki et al., 2006). This process will be discussed in a later

section.

6S RNA functions as a global transcription regulator by interacting with σ70 –

RNAP and inhibiting its activity (Wassarman & Storz, 2000). A recent study has

revealed that E. coli 6S RNA is cleaved by an RNase Z family protein, RNase BN, which

acts in maturation of tRNA precursors (Dutta & Deutscher, 2010, Dutta et al., 2012).

RNase BN level is growth phase dependent, it reaches the highest level in early

exponential phase, and is essentially absent in stationary phase. This leads to low level

of 6S in exponential phase and accumulation of 6S as entering stationary phase (Chen

et al., 2016). As a consequence, the housekeeping RNA polymerase is available for

enhanced transcription during exponential phase of growth, while under stationary

Page 21: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

21

phase, its activity is repressed by accumulated 6S RNA, resulting in decreased

expression of σ70-dependent transcription.

The Carbon Storage Regulator (Csr) System

CsrA/RsmA

To cope with changes in nutrient availability, bacteria utilize both transcriptional

and posttranscriptional regulators to modulate gene expression. RpoS, cAMP-Crp and

ppGpp were discussed earlier as transcriptional regulators governing gene expression

under nutrient-limited conditions. In addition, CsrA and its orthologs RsmA and RsmE

posttranscriptionally modulate gene expression and serve as major switches of bacterial

lifestyle between rapid growth and stress resistant growth. CsrA/RsmA is conserved

throughout Gammaproteobacteria and globally represses stationary phase processes,

including gluconeogenesis, glycogen synthesis, stringent response, biofilm formation

and quorum sensing, while it stimulates expression of genes related to motility and

glycolysis for rapid growth (Romeo, 1998, Babitzke & Romeo, 2007, Romeo et al.,

2013, Vakulskas et al., 2015).

CsrA/RsmA regulates gene expression via a direct binding to mRNA targets.

Structural studies of E. coli CsrA, its homolog RsmA in Yersinia enterocolitica, and the

CsrA/RsmA in complex with RNA revealed that CsrA/RsmA is homodimeric protein with

two RNA binding surfaces formed primarily by the parallel β-1 and β-5 strands of

oppossing polypeptides (Gutiérrez et al., 2005, Heeb et al., 2006, Mercante et al., 2006,

Schubert et al., 2007). The two RNA binding surfaces of CsrA/RsmA allows it to bridege

tow target sites on a single RNA molecule (Mercante et al., 2009). In vitro selection and

in vivo UV crosslinking with RNA deep sequencing (CLIP-seq) revealed that CsrA

Page 22: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

22

prefers to recognize AUGGA sequences present in apical loops of hairpin structures in

many bacterial species (Dubey et al., 2005, Holmqvist et al., 2016).

To achieve its regulatory role, CsrA often interacts with the 5’-UTR or early

coding region and modulates translation efficiency (Baker et al., 2002, Baker et al.,

2007, Dubey et al., 2003), transcript stability (Liu et al., 1995, Wang et al., 2005,

Yakhnin et al., 2013) and/or transcription elongation (Figueroa-Bossi et al., 2014) (Fig.

1-1). CsrA binding mostly blocks expression, but it also works as a positive regulator of

gene expression.

Regulation of the Csr System

In Gammaproteobacteria, CsrA/RsmA activity is primarily controlled through

sequestration by non-coding sRNAs, such as CsrB/C in E. coli. These sRNAs contain

multiple stem-loops with the conserved CsrA binding GGA motifs, which allow them to

bind CsrA with high affinity, sequester CsrA away for lower affinity mRNA targets and

therefore antagonize CsrA activity (Fig. 1-1) (Liu et al., 1997). In E. coli, CsrB appears

to be the principal sRNA antagonizing CsrA activity under laboratory growth conditions

(Weilbacher et al., 2003, Liu et al., 1997). Deletion of CsrB affects expression of a

number of CsrA target genes and several physiology processes (Liu et al., 1997). The

effects of CsrC on gene expression and celluar physiology are not observed unless

csrB is deleted (Weilbacher et al., 2003). McaS sRNA is recently identified as another

CsrA inhibitory regulator, but its effects are only substantial when overexpressed

(Jørgensen et al., 2013). The redundant effect of these antagonizing sRNAs might

enhance the robustness of the Csr regulation, and also ensure fine-tuning of CsrA

activity in response to different stress conditions.

Page 23: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

23

Despite the widespread distribution of CsrA, a few bacteria, like Gram-negative

Legionella pneumophila and Gram-positive Bacillus subtilis, appear to lack the CsrA

inhibitory sRNAs. In these species, CsrA activity is regulated by a protein antagonist,

FliW, and these proteins together with a cytoplasmic Hag (flagellin) protein contribute to

the tight control of flagellin homeostasis (Mukherjee et al., 2011, Dugar et al., 2016).

Additionally, two mRNAs (flaA and fimAICDHF) derived RNA antagonists of CsrA inhibit

CsrA activity when highly expressed (Dugar et al., 2016, Sterzenbach et al., 2013).

In bacteria that possess the CsrA inhibitory sRNAs, fluctuations in the levels of

these sRNAs play a central role in regulating the Csr system (Weilbacher et al., 2003,

Liu et al., 1997). Their synthesis and stability have been studied in a few bacteria

species (Vakulskas et al., 2015) .

Transcription of CsrA Inhibitory sRNAs

Multiple factors ensure appropriate expression of Csr sRNAs under different

environmental signals. Transcription of E. coli CsrB/C is primarily regulated by the BarA-

UvrY two component signaling transduction system (TCS), which is highly conserved

across Gammaproteobacteria (Suzuki et al., 2002, Chavez et al., 2010, Martínez et al.,

2014, Zere et al., 2015). BarA protein is a membrane-bound sensor-kinase and its

activity is activated by acetate, formate and other carboxylate compounds (Chavez et

al., 2010, Huang et al., 2008). The response regulator UvrY is subsequently

phosphorylated and stimulates CsrB/C transcription (Zere et al., 2015). In a complex

negative feedback loop, CsrA regulates csrB/C transcription by activating both the

expression of the response regulator UvrY and its phosphorylation by the sensor-kinase

BarA (Suzuki et al., 2002, Suzuki et al., 2006, Camacho et al., 2015). Amino acid

starvation and other stresses activate CsrB/C transcription via the stringent response

Page 24: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

24

components ppGpp and DksA (Edwards et al., 2011). In addition, two DEAD-box RNA

helicases, DeaD and SrmB (Vakulskas et al., 2014), IHF (Zere et al., 2015) and cAMP-

Crp (Pannuri et al., 2016) activate or repress csrB/C transcription by distinct

mechanisms.

Regulation of the Turnover of CsrA Inhibitory sRNAs

The decay of CsrB/C RNAs has only been characterized in a few bacterial

species. In E. coli, CsrB/C turnover is mediated by RNase E and PNPase and facilitated

by protein CsrD (Suzuki et al., 2006). CsrD does not appear to be a nuclease but

renders CsrB/C susceptible to degradation by RNase E, thus affecting the expression of

CsrA-regulated genes in a predictable fashion (Suzuki et al., 2006). CsrD contains

GGDEF and EAL domains, which are often responsible for synthesis and degradation of

the secondary messenger cyclic dimeric (3′→5′) GMP (c-di-GMP) (Suzuki et al., 2006).

However, CsrD lacks the critical catalytic amino acid residues of GGDEF and EAL

domains and displays no c-di-GMP synthetic or hydrolytic activity (Suzuki et al., 2006).

Besides, its activity does not respond to c-di-GMP (Suzuki et al., 2006). So far, the

molecular mechanism of CsrD effects on CsrB/C degradation is unclear. CsrD binds

non-specifically to CsrB/C in vitro. Whether it has evolved from a typical GGDEF-EAL

domain protein to a protein with an alternative function, such as RNA binding, need to

be further studied. In addition, how CsrD activity is regulated is another open question.

CsrA weakly represses csrD expression in E. coli and Salmonella typhimurium (Suzuki

et al., 2006, Jonas et al., 2010) but does not seem to affect CsrB turnover in E. coli

(Suzuki et al., 2006). No other factors besides CsrA are known to affect CsrD activity.

CsrD orthologs are present in a number of bacterial families, including

Enterobacteriaceae, Vibrionaceae and Shewanellaceae (Suzuki et al., 2006). In E. coli,

Page 25: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

25

the presence of CsrD drastically facilitates CsrB/C decay rates from half-lives of more

than 32 min to 1-4min during exponential phase growth. Nevertheless, in Pseudomonas

fluorescens, which lacks CsrD, these sRNAs are much more stable, with half-lives from

∼20 min to >60 min. Moreover, binding of the CsrA homolog RsmA stabilizes these

sRNAs by blocking RNase E cleavage in P. fluorescens (Duss et al., 2014, Reimmann

et al., 2005). However, CsrA displays on substantial effect on CsrB/C turnover in E. coli

(Suzuki et al., 2006). Whether this discrepancy is related to the presence of CsrD in E.

coli remains to be further explored.

CsrA Regulates its Own Expression

The level of CsrA in the cell is subject to a complex autoregulation. CsrA

represses its own translation by binding to the untranslated leader region of its mRNA

and blocking ribosome binding (Yakhnin et al., 2011). In the meanwhile, it activates its

own transcription indirectly through RpoS, which mediates the transcription of the P3

promoter of the csrA gene (Yakhnin et al., 2011). Furthermore, CsrA activity is

controlled by the negative feedback loop that exists within the Csr regulatory circuitry

(Gudapaty et al., 2001, Suzuki et al., 2002, Weilbacher et al., 2003). When CsrA activity

increases to certain level, it stimulates CsrB/C synthesis, therefore antagonizing its own

activity (Romeo et al., 2013). This complex autoregulation of its own expression and

activity fine-tunes the CsrA activity during different growth status and stress conditions.

Interaction of Csr System with Other Regulatory Pathways

CsrA participates in diverse regulatory pathways by directly binding to mRNAs of

the regulatory factors or components of these pathways (Romeo et al., 2013). A few

examples are described here. CsrA represses PGA-dependent biofilm formation at

Page 26: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

26

multiple layers (Romeo et al., 2013). First, it directly binds to the untranslated leader of

pgaABCD mRNA and represses pgaA translation (Wang et al., 2005). In addition, it

inhibits the translation of NhaR, a positive regulator of pgaABCD transcription, by

occluding ribosome binding (Goller et al., 2006). Furthermore, CsrA reduces the

abundance of c-di-GMP, a secondary messenger that activates PGA synthesis and

biofilm formation, by controlling expression of genes responsible for c-di-GMP

metabolism (Jonas et al., 2008, Jonas et al., 2010). Besides, CsrA stimulates the Rho-

dependent transcription termination of pgaABCD mRNA by binding to the upstream

portion of its 5’-UTR and exposing the Rho recognization site within this region

(Figueroa-Bossi et al., 2014).

The same type of multiple layer regulation is also observed in CsrA regulation of

virulence formation in Legionella pneumophila (Sahr et al., 2017). CsrA regulates the

virulence formation by two routes, 1) by regulating the expression of several major

regulatory proteins (FleQ, LqsR, LetE and RpoS) related to virulence formation and 2)

by directly interacting with the transcripts of over 40 Dot/Icm type IV secreted effector

proteins and modulating their synthesis.

In addition, a reciprocal regulatory interaction is observed between the Csr

system and the stringent response (Edwards et al., 2011). During amino acid starvation

and other stresses, synthesis of the secondary messenger, (p)ppGpp, is stimulated and

modulates transcription of a large set of gene for stress resistant response (Cashel &

Gallant, 1969, Potrykus & Cashel, 2008). Synthesis of CsrB/C is strongly stimulated by

(p)ppGpp and the (p)ppGpp-responsive transcription regulator DksA. In the meanwhile,

Page 27: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

27

CsrA binds the transcripts of RelA and SpotT that are involved in ppGpp synthesis and

hydrolysis, and DksA in order to modulate their expression (Edwards et al., 2011).

Project Rationale and Objectives

sRNAs are important factors in posttranscriptional regulation. Their levels

respond to diverse environmental stimuli and thereby properly modulate expression of

particular gene sets, adapting cellular metabolism and physiology to environmental

niches (Gottesman & Storz, 2011, Storz et al., 2011). Unlike most of the sRNAs that act

by base-pairing target mRNAs, E. coli CsrB/C act indirectly by binding to and titrating

the posttranscriptional regulatory protein CsrA (Romeo et al., 2013). Fluctuations of

CsrB/C levels play important roles in governing CsrA activity and many physiological

processes in response to different environmental stimuli. Biosynthesis of CsrB/C is

extensively controlled by multiple regulators that use distinct mechanisms under diverse

stress conditions (Chavez et al., 2010, Huang et al., 2008, Edwards et al., 2011, Suzuki

et al., 2002, Camacho et al., 2015, Vakulskas et al., 2014, Zere et al., 2015, Pannuri et

al., 2016). Interestingly, the decay rates of CsrB/C also play a role in determining their

steady state levels (Suzuki et al., 2006). CsrD protein is identified as a specific protein

that facilitates CsrB/C turnover (Suzuki et al., 2006). The overall goal of this study is to

explore the molecular mechanism of the action of CsrD and the physiological role of

CsrD in the CsrB/C turnover pathway.

The first objective of this study is to understand the physiological role of CsrD in

the CsrB/C decay pathway in E. coli. A recent study discovered the CsrD homolog of

Vibrio cholera as a binding partner of EIIAGlc, a component of the carbohydrate

phosphotransferase system (PTS), which serves in the uptake and phosphorylation of

glucose (Pickering et al., 2012). But no physiological role was assigned to this

Page 28: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

28

interaction. Thus, this study is to illustrate whether EIIAGlc regulates the turnover of

CsrB/C by interacting with CsrD in E. coli and to characterize the molecular mechanism

and physiological consequences of this regulation.

The second objective of this study is to explore the molecular mechanism by

which CsrD facilitates CsrB/C turnover. Degradation of many sRNAs requires additional

factors in addition to RNases, such as Hfq (Basineni et al., 2009) and RapZ (Göpel et

al., 2013), to recruit RNases for rapid degradation. However, until now, the molecular

mechanism of CsrD action in CsrB/C turnover pathway is not yet well-characterized.

CsrD binds to CsrB/C in vitro, albeit non-specifically (Suzuki et al., 2006). It contains

GGDEF and EAL domains, which are typically involved in c-di-GMP synthesis and

hydrolysis, respectively (Hengge, 2009). However, CsrD displays no activity in c-di-

GMP metabolism (Suzuki et al., 2006). Whether CsrD acts directly by binding to CsrB or

indirectly through other factors needs to be investigated.

The third major objective of this study is to explore the regulatory role of CsrD in

a genome-wide scale. CsrD was previously observed to regulate expression of a

number of CsrA target genes due to its effects on CsrB/C levels and CsrA activity

(Suzuki et al., 2006). Thus, the open question is whether CsrD has a broader regulatory

role in addtition to affecting CsrB/C turnover, and whether it regulates gene expression

primarily through CsrB/C and CsrA or other unknown factor(s) or pathway(s). Epistasis

analysis together with RNA-seq will be conducted to resolve these questiones.

Page 29: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

29

Figure 1-1. Outline of the Csr system in E. coli (Vakulskas et al., 2015). CsrA generally binds to conserved GGA motifs in the 5’-untranslated or early coding region of target mRNAs, leading to changes in translation initiation (as shown here), RNA stability, and/or transcription elongation. CsrA activity is controlled primarily by the steady state levels of CsrB and CsrC (CsrB is shown here), which contain many high affinity CsrA binding sites that sequester CsrA from interacting with its lower affinity mRNA regulatory targets. The levels of CsrB/C are regulated at the level of transcription and turnover. Ribosomes are depicted in blue. I have obtained the permission to use this figure from American Society for Microbiology.

Page 30: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

Leng, Y., Vakulskas, C.A., Zere, T.R., Pickering, B.S., Watnick, P.I., Babitzke, P., and Romeo, T. (2016) Regulation of CsrB/C sRNA decay by EIIAGlc of the phosphoenolpyruvate: carbohydrate phosphotransferase system. Mol Microbiol 99: 627-639.

30

CHAPTER 2 REGULATION OF CsrB/C sRNA DECAY BY EIIAGlc

Introduction

The Csr/Rsm system is present in diverse eubacteria, where it globally regulates

metabolism, biofilm formation, motility, virulence, quorum sensing, and stress response

systems (Romeo, 1998, Babitzke & Romeo, 2007, Romeo et al., 2013, Vakulskas et al.,

2015). The RNA binding protein CsrA/RsmA of the Csr system regulates gene

expression by interacting with sequences in mRNA, thus altering translation, mRNA

stability, and/or transcript elongation. CsrA governs genes responsible for bacterial

lifestyle transitions, repressing processes that are triggered upon entry into the

stationary phase of growth and conferring stress resistance, while activating processes

such as glycolysis, which support vigorous growth. In E. coli, CsrA activity is mainly

controlled by the noncoding sRNAs, CsrB and CsrC, which contain multiple CsrA

binding sites, allowing them to antagonize CsrA activity by sequestering it away from

lower affinity mRNA targets (Liu et al., 1997, Weilbacher et al., 2003). Fluctuations in

CsrB/C levels play a central role in regulating Csr system and the bacterial lifestyle.

Multiple factors ensure appropriate expression of csrB/C. Transcription is

activated by the BarA-UvrY TCS in response to carboxylic acids (Suzuki et al., 2002,

Chavez et al., 2010, Martínez et al., 2014). In a complex negative feedback loop, CsrA

regulates csrB/C transcription by activating both the expression of the response

regulator UvrY and its phosphorylation by the sensor-kinase BarA (Suzuki et al., 2006,

Camacho et al., 2015, Suzuki et al., 2002). Amino acid starvation and other stresses

activate csrB/C transcription via the stringent response components ppGpp and DksA

Page 31: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

31

(Edwards et al., 2011) and two DEAD-box RNA helicases, DeaD and SrmB, activate

csrB/C transcription by distinct mechanisms (Vakulskas et al., 2014).

In contrast to synthesis, the decay of CsrB/C RNAs is not well understood. A

specificity factor, CsrD, is necessary for degradation of CsrB/C by the housekeeping

nucleases RNase E and PNPase (Suzuki et al., 2006). CsrD does not appear to be a

nuclease, but renders CsrB/C susceptible to degradation by RNase E, thus affecting the

expression of CsrA-regulated genes in a predictable fashion. CsrD contains GGDEF

and EAL domains, which are often responsible for synthesis and degradation of the

secondary messenger cyclic dimeric (3’→5’) GMP (c-di-GMP). However, biochemical

and genetic studies indicated that CsrD displays no c-di-GMP synthetic or hydrolytic

activity and that CsrD activity is not regulated by c-di-GMP in vivo. At present, the

molecular mechanism of CsrD effects on CsrB/C degradation is unclear. CsrD bound

nonspecifically to CsrB/C in vitro (Suzuki et al., 2006). Accordingly, one hypothesis is

that CsrD evolved from the GGDEF-EAL domain family, becoming an RNA binding

protein. How CsrD activity is regulated is another open question. CsrA weakly represses

csrD expression in E. coli and Salmonella Typhimurium (Suzuki et al., 2006, Jonas et

al., 2010), but does not seem to affect CsrB turnover in E. coli (Suzuki et al., 2006), and

no other factors are known to affect CsrD activity.

Glucose is the preferred carbon and energy source for E. coli and is taken up

primarily by the glucose-specific phosphoenolpyruvate-dependent sugar-

phosphotransferase system, PTS (Deutscher et al., 2006, Deutscher et al., 2014,

Lengeler & Jahreis, 2009). This system consists of two cytoplasmic proteins, enzyme I

(EI) and histidine phosphocarrier protein (HPr) that are used for transporting many

Page 32: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

32

sugars, and two glucose-specific proteins, enzyme IIAGlc (EIIAGlc) and the membrane-

bound enzyme IIBCGlc (EIIBCGlc). Glucose uptake is coupled to its phosphorylation. The

phosphoryl group is donated by PEP and transferred to glucose via a phosphorylation

cascade formed by EI, HPr, EIIAGlc, and EIIBCGlc proteins. Thus, the phosphorylation

state of the PTS proteins depends both on extracellular carbon availability and the

metabolic state of the cell.

The glucose-PTS proteins also mediate regulatory functions (Gabor et al., 2011,

Deutscher et al., 2014). EIIAGlc is a central regulator of carbon metabolism.

Unphosphorylated EIIAGlc mediates inducer exclusion by binding to and inhibiting

transporters of non-PTS sugars (Deutscher et al., 2014, Deutscher et al., 2006). It also

inhibits metabolism of alternative carbon sources, e.g. by binding to glycerol kinase

(Postma et al., 1984). In contrast, phosphorylated EIIAGlc (EIIAGlc-P) binds to and

stimulates the activity of adenylate cyclase, which produces cAMP. This compound acts

as a secondary messenger that binds to the cAMP receptor protein (Crp), forming a

transcription factor (cAMP-Crp) that exerts global effects on the proteome, ensuring

efficient resource utilization (Krin et al., 2002, Park et al., 2006, Bao & Duong, 2013).

With the discovery of novel EIIAGlc binding partners in various species, EIIAGlc has been

found to participate in chemotaxis (Neumann et al., 2012), respiration/fermentation (Koo

et al., 2004), biofilm formation (Pickering et al., 2012) and virulence (Kim et al., 2010,

Mazé et al., 2014). The EIIBCGlc protein also carries out a variety of regulatory functions

(Lux et al., 1995, Nam et al., 2001, Tanaka et al., 2000, Lee et al., 2000).

In a screen for EIIAGlc binding partners in Vibrio cholerae, a CsrD homologue,

MshH was identified (Pickering et al., 2012) . While no function was assigned to this

Page 33: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

33

interaction, it was hypothesized that EIIAGlc might affect the decay of Csr sRNAs. Here,

we present the results of a detailed investigation of the role of EIIAGlc on CsrB/C decay

in E. coli. Unphosphorylated EIIAGlc binds specifically to the EAL domain of CsrD and

stimulates CsrB/C turnover. We propose that this mechanism helps to increase the

concentration of free CsrA when it is needed to support growth, and simultaneously

poises the Csr system for rapid response to changing environmental conditions.

Materials and Methods

Bacterial Strains and Culture Conditions

The bacterial strains used in this study are listed in Table B-1. Bacterial strains

were routinely grown in Luria-Bertani (LB) broth unless otherwise indicated. For

synthetic minimal medium, minimal medium A (Hogema et al., 1998) and M9 minimal

medium supplemented with indicated carbohydrates were used. When necessary, the

following antibiotics were added to the growth media: ampicillin (100 μg mL−1),

tetracycline (15 μg mL−1), kanamycin (50 μg mL−1), and chloramphenicol (25 μg mL−1).

E. coli and V. cholerae strains were grown at 37°C and 27°C, respectively. Stationary

phase cultures were routinely used to inoculate LB broth or minimal media unless

otherwise indicated. For strains carrying cyaA deletion or crp disruption, exponentially

growing cultures were used to inoculate LB broth supplemented with or without 10 mM

cAMP to minimize the growth defect.

Construction of Plasmids and Mutant Strains

The plasmids and related primers and restriction sites used in this study are

listed in Tables B-2 and B-3. The plasmid pCRR, used for complementation of a crr

deletion strain, carries the crr gene under a mutant lacUV5 promoter, in which -8 A of

the -10 hexamer consensus was replaced with -8 T for decreased promoter activity

Page 34: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

34

(Moyle et al., 1991). To generate pCRR, the crr gene was amplified from chromosomal

DNA of E. coli MG1655 and ligated into vector pBR322. Plasmid pCRRH91A expressed

the mutant crr gene, producing the protein EIIAGlcH91A. This plasmid was constructed

similarly to pCRR except that the His91Ala (CAC to GCC) substitution was introduced

by the megaprimer PCR procedure (Ke & Madison, 1997). Plasmid pBYH4 used for

complementation of the csrD deletion strain was described previously (Suzuki et al.,

2006). To construct plasmid pETCRR for expression of C-terminal His-tagged EIIAGlc,

crr was amplified and cloned into plasmid pET24a. Plasmids overexpressing CsrD

variants were generated by amplifying the coding regions corresponding to CsrDΔTM

(residues 156-646), CsrDΔHAMP (residues 192-646), CsrDΔCoil (residues 156-199 and

220-656), CsrDΔGGDEF (residues 156-223 and 393-646), CsrDΔEAL (residues 156-385)

and EAL (residues 393-646) by standard PCR or overlapping PCR (Urban et al., 1997),

and cloning the resulting products into vector pmal-c5x, yielding N-terminally MBP-

tagged CsrD variants.

E. coli gene deletions were created by the standard P1vir transduction procedure

or the lambda Red system as described (Datsenko & Wanner, 2000). Chromosomal C-

terminal FLAG-tagged fusions were generated using the phage lambda Red system as

described (Datsenko & Wanner, 2000, Uzzau et al., 2001). The mutant strain H91A

carrying a chromosomal point mutation of crr, producing EIIAGlc H91A protein, was

constructed by using overlapping PCR mutagenesis and the pKOV gene replacement

protocol as described (Urban et al., 1997, Link et al., 1997).

Page 35: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

35

V. cholerae ∆crr and ∆mshH mutants were created in the C6706str2 wild-type

background (Thelin & Taylor, 1996) by double homologous recombination as previously

described (Haugo & Watnick, 2002, Pickering et al., 2012).

Expression and Purification of EIIAGlc and CsrD Variants

EIIAGlc was overproduced in E. coli strain BL21 (DE3) grown in 1L of M9 minimal

medium supplemented with 0.8% glucose (w/v). Three hours after the induction with

1mM IPTG at OD600 ~ 0.6, cells were harvested and lysed using a French Press. After

centrifugation (20,000 × g, 30 min, 4 ̊C), the soluble fraction of the lysate was applied to

a HisTrap column (1 mL, GE Healthcare) and eluted with a gradient of imidazole (20-

500 mM). Eluted proteins were further purified by gel filtration chromatography

(Superdex 75 10/300, GE Healthcare), dialyzed against dialysis buffer (20 mM Tris-HCl,

pH7.5, 100 mM NaCl, 1 mM DTT, 10% glycerol) and stored for subsequent

experiments.

Overproduction of CsrD variants was from E. coli BL21 (DE3) strains containing

the corresponding plasmids, which were grown in LB medium supplemented with 0.2%

glucose (w/v). Cells were induced with 0.3 mM IPTG at OD600 ~ 0.6 and lysed as

mentioned above. The soluble fraction of the lysate was applied to an MBP Trap column

(1 mL, GE Healthcare) and eluted with a gradient of 0-10 mM maltose. To obtain

homogeneous proteins, eluted proteins were further purified by gel filtration

chromatography (Superdex 200 10/300, GE Healthcare) and dialyzed against dialysis

buffer.

Northern Blotting

Northern blot analysis was performed as previously described, with minor

modifications (Vakulskas et al., 2014). Bacterial culture were immediately stabilized by

Page 36: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

36

the addition of RNA protect TM Bacteria Reagent (Qiagen) or 0.125 volumes of stop

solution (10% phenol/90% ethanol). Total RNA was isolated using the RNeasy mini kit

(Qiagen) according to the manufacturer’s instructions. Total cellular RNA (1-2 μg) was

separated on denaturing 5% acrylamide/7 M urea gels and transferred to a positively

charged nylon membrane (Roche Diagnostics) by electroblotting. The membrane was

cross-linked using UV light and hybridized overnight at 68°C (CsrB/C of E. coli) or 70°C

(CsrB/C/D of V. cholerae) using a DIG-labeled antisense RNA probe (Table B-3), which

was prepared with the DIG Northern Starter kit (Roche Diagnostics). Transcripts were

detected using the DIG Northern Starter kit (Roche Diagnostics) according to the

manufacturer’s instructions. Blots were imaged using the ChemiDoc XRS+ system (Bio-

Rad) and RNAs were quantified using Quantity One image analysis software (Bio-Rad).

Prior to hybridization, the rRNAs (16S and 23S) were stained by methylene blue, which

served as loading controls for signal correction.

Western Blotting

Western blot analysis was performed using standard laboratory protocols as

described (Vakulskas et al., 2014). Briefly, total cellular proteins were separated by

SDS-PAGE and transferred to 0.2 μm polyvinylidene difluoride membranes (Bio-Rad)

by electroblotting. Blots for FLAG epitope-tagged proteins used the anti-FLAG M2

monoclonal antibody (Sigma) at 1:2,000 dilution. Blots for RpoB used anti-RpoB

monoclonal antibodies (Neoclone) at 1:50,000 dilution. Western blots were detected

using horseradish peroxidase-linked secondary antibodies and the Super Signal West

Femto Chemiluminescent Substrate (Pierce) as recommended by the manufacturer.

Proteins were quantified using Quantity One image analysis software (Bio-Rad).

Page 37: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

37

Protein Pull-down Assays

Interaction between CsrD variants and EIIAGlc was assessed using purified His-

tagged EIIAGlc protein to pull down MBP-tagged CsrD variants. In a 60 μL reaction, 8

μM EIIAGlc and 4 μM CsrD variants were incubated with 15 μL Ni-NTA resin (Qiagen) in

binding buffer (50 mM MES pH 6.5, 1 mM DTT, 20 mM Imidazole) at 4°C for 1hour.

Unbound proteins remaining in the supernatant solution were collected after brief

centrifugation of the resin. The resin was washed three times with 1 mL washing buffer

(50 mM MES pH 6.5, 1mM DTT, 60 mM Imidazole), and the bound proteins were eluted

with 45 μL of elution buffer (25 mM Tris-HCl pH 8.0, 500 mM NaCl, and 500 mM

Imidazole).

To determine the phosphorylation dependence of the interaction between EIIAGlc

and CsrD, the MBP-tagged CsrDΔTM was used to pull down His-tagged EIIAGlc. His-

tagged EIIAGlc (8 μM) was incubated in reactions containing EI (1 μM), Hpr (8 μM) and

either 5 mM PEP or 5 mM pyruvate in 60 μL buffer (20mM Tris-HCl pH8.0, 2 mM MgCl2,

1 mM DTT) at room temperature for 10 min. These reactions were designed to produce

the phosphorylated or unphosphorylated form of EIIAGlc, respectively. Reactions were

dialyzed against the binding buffer (50mM MES pH6.5, 1mM DTT) with Slide-A-Lyzer

dialysis cassette (Thermo) for 1 hour and subsequently incubated at 4°C for 1 hr with

CsrDΔTM (95 μg) pre-bound to amylose resin. Unbound proteins were collected after

brief centrifugation of the resin and bound proteins were eluted with 60 μL of elution

buffer (20 mM Tris-HCl pH7.5, 200 mM NaCl, 10 mM maltose, 1 mM DTT) after

extensive rinsing of the resin with binding buffer. Unbound and bound proteins in the

pull-down reactions were detected by SDS-PAGE (10% or 15%, as required) and

Page 38: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

38

Coomassie blue staining. Proteins were quantified using Quantity One image analysis

software (Bio-Rad).

Determination of the EIIAGlc Phosphorylation State

The phosphorylation state of EIIAGlc was determined as previously described

(Hogema et al., 1998), with modifications. Briefly, 0.2 mL of bacterial culture (OD600 ~

0.5) was treated with the addition of 20 μL of 10 M NaOH followed by 1 mL of ethanol

and 180 μL of 3 M sodium acetate, pH 5.2. After chilling at -80°C for 2 hr, precipitates

were collected by centrifugation, rinsed with 70% ethanol, and suspended in 100 μL of

sample buffer (0.16 M Tris HCl, pH 7.5, 4% SDS, 20% glycerol, and 10% 2-

mercaptoethanol). To achieve a good separation of the two forms of EIIAGlc, samples

were fractionated on SDS-PAGE gels containing 50 μM of Phos-tag reagent as

previously described (Vakulskas et al., 2014). Subsequently, gels were washed with

Western transfer buffer (25 mM Tris, 192 mM Glycine, 20% methanol, and 0.1% SDS)

containing 1 mM EDTA for 10 min, followed by a second wash with transfer buffer for 20

min. Western blot analysis was then performed as described above.

Gel Filtration Analysis of EIIAGlc in Complex with the EAL Domain or Other CsrD Variants

As discussed below, a site-directed crr mutant allele, encoding an EIIAGlc protein

that cannot be phosphorylated, H91A, also complemented the crr deletion (Fig. 2-1A-D).

For analysis of the EIIAGlc-EAL complex, a 0.5 mL reaction containing 33 μM EAL, 78

μM EIIAGlc or both proteins were incubated at 4 °C for 30 min and subjected to gel

filtration analysis using an AKTA-FPLC system (Superdex 200, HiLoadTM 16/60, 120

mL, GE Healthcare), and subsequently eluted at 4°C with a flow rate of 1 mL/min in

buffer containing 20 mM Tris HCl pH 7.5, 100 mM NaCl, 1mM DTT. For CsrD variants,

Page 39: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

39

0.5 mL samples containing purified CsrDΔTM, CsrDΔEAL, CsrDΔHAMP, CsrDΔCoil, or

CsrDΔGGDEF was separated on the same system. Fractions (3 mL) were collected and

analyzed by SDS-PAGE and Coomassie blue staining. For EIIAGlc-EAL analysis, the

column was pre-calibrated using 5 gel filtration molecular weight markers (1, sweet

potato β-amylase, 200 kDa; 2, yeast alcohol dehydrogenase, 150 kDa; 3, bovine serum

albumin, 66 kDa; 4, carbonic anhydrase from bovine erythrocytes, 29 kDa; 5, horse

heart cytochrome C, 12.4 kDa), and blue dextran 2000. For CsrD variants analysis, the

column was calibrated using 5 molecular weight markers (1, equine spleen apoferritin,

443 kDa; 2, sweet potato β-Amylase, 200 kDa; 3, alcohol yeast dehydrogenase, 150

kDa; 4, bovine serum albumin, 66 kDa; 5, carbonic anhydrase from bovine erythrocytes,

29 kDa), and blue dextran 2000. The relative molecular masses of proteins or protein

complexes were calculated by logarithmic interpolation from the standards.

Results

EIIAGlc Activates CsrB/C Decay via CsrD

Because the role of CsrD in CsrB/C decay has been investigated only in E. coli,

we decided to determine if EIIAGlc participates in the degradation of CsrB/C in this

species. To do so, we first determined the stability of CsrB/C in the presence or

absence of EIIAGlc (Δcrr) after the addition of rifampicin to the exponentially growing

cultures. Deletion of crr decreased CsrB and CsrC decay rates by about 5-fold and 3-

fold, respectively (Fig. 2-1A-D). Ectopic expression of crr complemented the crr defect,

confirming that EIIAGlc somehow regulates CsrB/C decay (Fig. 2-1A-D). As discussed

below, a site-directed crr mutant allele, encoding an EIIAGlc protein that cannot be

phosphorylated, H91A, also complemented the crr deletion (Fig. 2-1A-D).

Page 40: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

40

As shown in Fig. 2-1E, while EIIAGlc greatly stimulated CsrB/C decay, it modestly

reduced the levels of CsrB/C in the cell. A similar observation was also made previously

in a csrD mutant strain, and was shown to be the result of the Csr regulatory circuitry

(Suzuki et al., 2006). CsrA indirectly activates transcription of CsrB/C via the BarA-UvrY

TCS (Suzuki et al., 2002). Thus, when CsrB/C decay is inhibited, these sRNAs

accumulate and sequester CsrA, causing a decrease in their transcription and an

attenuated effect on their levels.

We next performed an epistasis experiment to determine whether the effect of

EIIAGlc on CsrB/C decay was dependent on CsrD. A Δcrr ΔcsrD double deletion strain

was severely defective in CsrB/C decay (Fig. 2-2A-B), as reported previously for the

ΔcsrD mutant (Suzuki et al., 2006). While ectopic expression of crr in the Δcrr ΔcsrD

strain failed to restore CsrB/C decay, ectopic overexpression of csrD enhanced CsrB/C

decay rates to wild-type levels (Fig. 2-2A and B). This finding suggested that CsrD

functions downstream of EIIAGlc in CsrB/C turnover.

A possible explanation for the epistasis results is that EIIAGlc affects CsrD levels

in the cell by altering its stability or synthesis. However, deletion of crr had no effect on

the levels of a chromosomally encoded and biologically functional CsrD-FLAG protein

(Fig. 2-2C and Fig. A-1).

In vitro Binding of EIIAGlc to CsrD Requires the EAL Domain

Because EIIAGlc regulates the activities of several proteins via direct binding, it

was reasonable to speculate that EIIAGlc affects CsrB/C decay via direct binding to CsrD

in E. coli. To test this idea, we examined the binding of EIIAGlc and CsrD in an in vitro

binding assay or pull-down assay using His-tagged EIIAGlc and a soluble recombinant

CsrD protein (CsrDΔTM) in which the N-terminal transmembrane domains of CsrD were

Page 41: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

41

replaced with a maltose binding protein (MBP) tag (Fig. 2-3A). Previous studies showed

that the transmembrane domains of CsrD were dispensable for its activity when the

protein was ectopically expressed (Suzuki et al., 2006). In this assay, CsrD was mixed

with Ni-NTA resin with or without His-tagged EIIAGlc. As shown in Fig. 2-3B, the CsrDΔTM

variant was retained by the Ni-NTA resin when EIIAGlc was bound to it, but remained in

the unbound fraction when EIIAGlc was absent, indicating that EIIAGlc bound directly to

CsrD.

To determine which domain of CsrD protein is involved in the interaction with

EIIAGlc, we tested similar MBP fusions of CsrD lacking the EAL (CsrDΔEAL), GGDEF

(CsrDΔGGDEF), or HAMP-like domain (CsrDΔHAMP and CsrDΔCoil), using the pull-down

assays. While the other CsrD variants retained the ability to bind to EIIAGlc, CsrDΔEAL

lost all detectable binding, suggesting that the EAL domain is involved in this interaction

(Fig. 2-3B-C). Moreover, the EAL domain alone bound to EIIAGlc in this assay (Fig. 2-

3D). These results indicated that EIIAGlc binds specifically to the EAL domain of CsrD.

To examine the binding reaction of EIIAGlc with CsrD in more detail, the size and

composition of the EIIAGlc-EAL complex was analyzed by gel-filtration chromatography.

The free EAL and EIIAGlc eluted at positions corresponding to sizes of their monomeric

forms (EAL, 72kDa; EIIAGlc, 20 kDa) (Fig. 2-3E and F). When EIIAGlc and EAL were

mixed to allow binding, and fractionated on Superdex 200 (HiLoadTM 16/60, GE

Healthcare), a new peak was observed at a position corresponding to a size of 98kDa,

approximately that of a heterodimer of EIIAGlc-EAL (92kDa). To determine the ratio of

EIIAGlc and EAL in the complex, column fractions corresponding to the presumptive

EIIAGlc-EAL complex and the free EIIAGlc were analyzed by SDS-PAGE with

Page 42: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

42

Commassie blue staining and quantification of the stained proteins (Fig. 2-3F). This

experiment revealed the molar ratio of EAL bound to EIIAGlc in peak fractions (11 and

12) to be 1:1 suggesting that EIIAGlc binds to the EAL domain of CsrD in a one to one

ratio.

In the pull down assay, the relative amount of CsrDΔTM or CsrDΔGGDEF bound by

EIIAGlc was much greater than that of proteins that lacked the intact HAMP-like domain,

CsrDΔHAMP or CsrDΔCoil (Fig. 2-3C). The HAMP domain typically promotes dimerization

or protein-protein interactions and plays important roles in signal transduction (Hulko et

al., 2006). We used gel filtration assays to determine the in vitro oligomeric states of

CsrD variants containing or lacking the HAMP-like domain. All CsrD variants containing

the HAMP-like domain, CsrDΔTM, CsrDΔGGDEF and CsrDΔEAL, eluted at volumes

consistent with their tetrameric forms (Fig. 2-4 and Table 2-1). In contrast, the CsrD

variants with a disrupted HAMP-like domain, CsrDΔHAMP and CsrDΔCoil, eluted as

apparent monomers (Fig. 2-4 and Table S4). The precise way in which tetramerization

affected EIIAGlc binding by the CsrD variants was not further investigated.

EIIAGlc Regulates CsrB/C Decay in a Phosphorylation-dependent Manner

EIIAGlc typically modulates the activity of its binding partners in a

phosphorylation-dependent manner. Accordingly, we tested the effect of the

phosphorylation state of EIIAGlc on CsrB/C decay. We first investigated the impact of the

unphosphorylated EIIAGlc on CsrB/C turnover using a site-directed mutant protein that

could not be phosphorylated (EIIAGlc H91A). Plasmid complementation of the Δcrr strain

with EIIAGlc H91A restored CsrB/C decay rates to slightly higher than in the wild-type

strain (Fig. 2-1A-D), demonstrating that phosphorylation of EIIAGlc was dispensable for

activation of CsrB/C turnover.

Page 43: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

43

Next, we performed pull-down assays to determine the effect of EIIAGlc

phosphorylation on binding to CsrD. In these experiments, CsrD containing an N-

terminal MBP tag was bound to amylose resin and then mixed with EIIAGlc in reactions

that were designed to produce either the phosphorylated or unphosphorylated form of

this protein. In one reaction, E. coli Hpr, EI and PEP were mixed to provide

phosphorylated EIIAGlc. In the other reaction, pyruvate was added instead of PEP to

maintain EIIAGlc in the unphosphorylated form. Strikingly, while most of the

unphosphorylated EIIAGlc bound to CsrD, no binding was observed between the

phosphorylated EIIAGlc and CsrD (Fig. 2-5). These results were in agreement with the

observation that EIIAGlc did not require phosphorylation for activation of CsrB/C turnover

in vivo (Figs. 2-1A-D), and indicated that the binding of unphosphorylated EIIAGlc to

CsrD activates CsrB/C sRNA decay.

cAMP-Crp Modestly Represses CsrB Turnover

While the unphosphorylated EIIAGlc bound to CsrD in vitro and was able to

activate CsrB/C decay in vivo, we wondered if the phosphorylated form of EIIAGlc might

affect CsrB/C turnover via its important role in cAMP-Crp production (Krin et al., 2002,

Park et al., 2006). Deletion of cyaA or crp modestly increased the CsrB decay rate by 2-

fold (Fig. 2-6A), while exhibiting weak or negligible effects on CsrC decay (Fig. 2-6B).

The increased decay rate of CsrB in the cyaA mutant was restored by exogenous cAMP

(10 mM), confirming that cAMP-Crp somehow inhibits CsrB decay. Deletion of cyaA or

crp in the Δcrr background had twofold effects on CsrB decay rates that were similar to

those in the wild-type background, and deletion of crr had similar fivefold effects on

CsrB decay in both the wild-type strain and its isogenic crp and cyaA mutants (Fig. 2-

6A). These findings confirmed that the major effect of EIIAGlc on CsrB decay is mediated

Page 44: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

44

independently of cAMP-Crp. The modest effect of cAMP-Crp in the Δcrr background

was likely due to basal adenylate cyclase activity in the crr mutant (Lévy et al., 1990,

Feucht & Saier, 1980, Reddy & Kamireddi, 1998).

CsrB/C Decay is Regulated in Response to Carbon Availability via the Phosphorylation State of EIIAGlc

The phosphorylation state of EIIAGlc is determined by carbon sources that are

taken up and metabolized (Hogema et al., 1998, Deutscher et al., 2014). Preferred

carbon sources such as glucose lead to net dephosphorylation of PTS proteins,

including EIIAGlc, whereas unfavorable carbon sources or carbon starvation conditions

cause the accumulation of phosphorylated EIIAGlc. Because the unphosphorylated

EIIAGlc bound to CsrD in vitro and promoted CsrB/C decay in vivo (Figs. 2-1 and 2-5),

we expected that CsrB/C decay rates should be elevated in the presence of glucose.

Consequently, we first examined CsrB/C decay in minimal medium supplemented with

0.2% glucose, glycerol or succinate. Both the phosphorylation state of EIIAGlc and

CsrB/C decay rates responded predictably to these carbon sources; more rapid decay

was observed in glucose compared to glycerol or succinate (Fig. 2-7).

To verify that the phosphorylation state of EIIAGlc determines the decay rates of

CsrB/C in response to different carbon sources, we examined CsrB/C turnover in a

strain that expresses the mutant EIIAGlc protein, H91A, which cannot be phosphorylated.

Because the strain expressing H91A has a significant growth defect in minimal medium

(data not shown) the WT and H91A strains were first grown in LB broth to exponential

phase and then washed and inoculated into minimal medium lacking a carbon

compound or containing 0.2% glucose or succinate. The phosphorylation state of EIIAGlc

was determined from growth in LB (Fig. 2-8A) and 10 min after inoculation into minimal

Page 45: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

45

media (Fig. 2-8B). EIIAGlc phosphorylation in LB was ~40% and increased to ~90% in

media with succinate or lacking a carbon source, while it decreased to 4% at 10 min

after glucose exposure. Decay rates of CsrB/C were determined 10 min after

inoculation (Fig. 2-9). The decay rates of CsrB and CsrC in the wild-type strain (WT)

were ~3.5 and 2.5-fold greater, respectively, in medium with glucose vs. no carbon

source. A more modest, but reproducible difference (~2 fold) was observed for CsrB/C

decay rates in glucose compared to succinate. These data support the observations

described above, showing that CsrB/C decay rates vary in response to different carbon

conditions, although the difference in decay between succinate and carbon-deficient

media does not seem to be explained by EIIAGlc phosphorylation alone (Fig. 2-9), as

both conditions resulted in similar EIIAGlc-P levels (Fig. 2-8B). Most importantly, CsrB/C

decay in the H91A strain was rapid and virtually identical in all three media, confirming

that the phosphorylation state of EIIAGlc determines CsrB/C decay in response to carbon

substrate availability. The levels of CsrB/C RNAs (Fig. 2-9E) were consistent with these

decay rates, but as observed previously (Fig. 2-1), the effects of turnover may be

attenuated via the feedback loop of the Csr circuitry (Fig. 2-1F) (Suzuki et al., 2002,

Suzuki et al., 2006).

EIIAGlc and MshH Promote Csr sRNA Decay in Vibrio cholerae

EIIAGlc, RNase E and CsrD orthologs are widespread in Enterobacteriaceae,

Vibrionaceae, and Shewanellaceae species (Suzuki et al., 2006, Vakulskas et al.,

2015), suggesting that a common mechanism may exist for Csr sRNA decay in

members of these bacterial families. As a proof of principle, we tested the effects of

EIIAGlc and the CsrD homolog, MshH, on decay of the V. cholerae sRNAs, CsrB, CsrC

and CsrD (Lenz et al., 2005). The V. cholerae sRNAs exhibited longer half-lives

Page 46: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

46

compared to the E. coli sRNAs under our growth conditions (Fig. 2-10). Nevertheless,

deletion of mshH, crr or both genes greatly decreased CsrB and CsrD turnover. These

effects were not apparent for CsrC, which was already extremely stable in the wild-type

strain. This experiment demonstrated the potential of EIIAGlc and CsrD to activate the

decay of Csr sRNAs in this important member of the Vibrionaceae family.

Discussion

Here, we identified a new regulatory function for EIIAGlc, in which binding to the

sRNA decay protein CsrD stimulates CsrB/C decay when glucose is present. This

mechanism should enhance the concentration of free CsrA, which activates glycolysis

and represses gluconeogenesis, secondary metabolism, and stress resistance

responses such as biofilm formation (Babitzke & Romeo, 2007, Vakulskas et al., 2015,

Romeo et al., 2013). Because CsrA regulates lifestyle transitions in many bacterial

species and interacts with hundreds of transcripts in E. coli (Babitzke & Romeo, 2007,

Patterson-Fortin et al., 2013, Edwards et al., 2011, Vakulskas et al., 2015), we propose

that this represents a particularly important role for EIIAGlc. A high rate of turnover can

facilitate rapid changes in transcript levels. Therefore, EIIAGlc-CsrD interactions should

not only allow CsrB/C decay rates to be reset in response to changing glucose

availability, but may poise the Csr system for rapid responses to other cues or

conditions when glucose is present. The V. cholerae CsrD ortholog, MshH and EIIAGlc

also activated the decay of CsrB and CsrD sRNAs of (Fig. 2-10). We suspect that the

mechanism described for E. coli CsrB/C turnover operates in many species of

Enterobacteriaceae, Vibrionaceae and Shewanellaceae.

The conclusion that only the unphosphorylated form of EIIAGlc is able to promote

CsrB/C decay through binding interactions with CsrD was based on a combination of

Page 47: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

47

biochemical and genetic evidence. CsrD bound only to the unphosphorylated form of

EIIAGlc in vitro (Fig. 2-5). Furthermore, a non-phosphorylatable protein, EIIAGlc-H91A,

sustained CsrB/C decay rates that were similar to or even greater than the wild-type

protein (Figs.2-1A-D). The presence of glucose also caused net dephosphorylation of

EIIAGlc and supported rapid decay of CsrB/C sRNAs relative to starvation conditions or

alternative carbon sources (Figs. 2-7-9). Importantly, EIIAGlc-H91A supported high

decay rates under all of these conditions, confirming that the effects of carbon

availability on CsrB/C decay are mediated thorough altered phosphorylation of EIIAGlc

(Fig. 2-9). A previous study with MshH of V. cholera concluded that both the

phosphorylated and unphosphorylated forms of EIIAGlc bind to the CsrD homolog. This

conclusion was based on the observation that in a two-hybrid assay, MshH interacted

with mutant proteins designed to mimic the unphosphorylated (EIIAGlc-H91A) or the

phosphorylated (EIIAGlc-H91D) forms of EIIAGlc (Pickering et al., 2012). However, we

caution that EIIAGlc-H91D does not appear to mimic EIIAGlc-P. Another putative EIIAGlc-P

mimic, EIIAGlc-H91E, was unable to activate adenylate cyclase (Reddy & Kamireddi,

1998). Similarly, effects of EIIANtr-P were not mimicked by replacing its

phosphorylatable His residue with either Asp or Glu (Lüttmann et al., 2009). Finally, we

constructed EIIAGlc-H91D in E. coli and found that it behaved similarly to

unphosphorylated EIIAGlc rather than EIIAGlc-P in CsrB/C decay (Fig. A-2).

The phosphorylated form of EIIAGlc activates cAMP synthesis by binding to

adenylate cyclase. Because cAMP and Crp modestly repressed CsrB decay (Fig. 2-6),

we propose that EIIAGlc-P indirectly and modestly represses CsrB turnover, reinforcing

the positive effect of unphosphorylated EIIAGlc on CsrB decay. Because a potential Crp

Page 48: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

48

binding site was predicted in the untranslated leader region of csrD (data not shown),

we tested the possibility that Crp inhibits CsrB decay by controlling csrD expression.

Weakly positive to negligible effects of Crp were observed on CsrD levels (Fig. A-3),

which might contribute to the effect of Crp on CsrB decay.

Given that EIIAGlc acts via CsrD without altering its levels in the cell and that

overexpression of csrD restored CsrB/C decay in a strain deleted for the EIIAGlc gene,

crr (Fig. 2-2), we propose that EIIAGlc functions as an allosteric activator of CsrD,

perhaps similar to the role of EIIAGlc-P in activating adenylate cyclase (Saier, 1989, Park

et al., 2006). Structural studies of EIIAGlc show that it possesses a concave face that

allows it to interact with globular target proteins (Hurley et al., 1993, Chen et al., 2013,

Wang et al., 2000, Cai et al., 2003). Other EIIAs seem unable to duplicate this function

(Deutscher et al., 2006). We deleted the genes for five other EIIAs (fruB, mtlA, chbA,

manX and ptsN) and found that none of them regulated CsrB/C decay (data not shown).

This study also expands our understanding of the functionality of the EAL

domain. This is not the first report of a catalytically inactive EAL domain performing a

regulatory role via protein-protein interactions (Römling et al., 2013). The enzymatically

inactive EAL domain protein YdiV of E. coli binds to the transcription activator FlhD4C2

and prevents it from binding to target DNA (Li et al., 2012). Similarly, the EAL domain of

the E. coli photoreceptor YcgF (BluF) binds to the MerR-like repressor YcgE (BluE) in

the presence of blue light and prevents it from binding to DNA (Tschowri et al., 2009).

Complex regulation exists for the EAL domain of FimX, which binds to c-di-GMP as well

as the PilZ protein, and is required for biogenesis of the Type IV pilus (Guzzo et al.,

2009). In all of these cases, the EAL domain-containing protein acts as a sensor that

Page 49: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

49

uses its EAL domain to transmit information to another protein. In contrast, the EAL

domain of CsrD acts as a receiver, to detect signaling information from a sensory

protein. The modular structure of CsrD suggests that the effect of EIIAGlc might be

transmitted through other CsrD domains, such as the GGDEF domain, which is also

necessary for CsrD activity (Suzuki et al., 2006), although this possibility has not been

explored.

While the circuitry surrounding the Csr system is extensive, its role in carbon and

energy pathways is particularly wide-ranging and important (Edwards et al., 2011,

Patterson-Fortin et al., 2013, Yang et al., 1996, Romeo, 1996, Romeo et al., 2013,

Sabnis et al., 1995, Pernestig et al., 2003, Yang et al., 1996). Previous studies have

shown that carboxylic acid-containing end products of carbon metabolism, such as

acetate and formate, stimulate CsrB transcription via the BarA-UvrY TCS (Chavez et al.,

2010). Thus, the synthesis pathway and the newly discovered turnover pathway for

CsrB/C should mediate reinforcing positive effects on the levels of these sRNAs when

preferred carbon resources have been expended and end products have accumulated.

The resulting decrease in CsrA activity under this condition should promote the

transition from glycolytic metabolism and active growth to gluconeogenesis, glycogen

biosynthesis and the formation of a stress resistant phenotype. We caution that other

regulators influence the expression of CsrB/C RNAs, e.g. ppGpp, DksA, DeaD and

SrmB helicases, and impact the workings of this circuitry. An understanding of the

combinatorial effects of all of these factors, and perhaps unknown ones, will require

additional investigation.

Page 50: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

50

Figure 2-1. EIIAGlc affects CsrB/C decay rates and levels in E. coli. A and C) Decay rates of CsrB/C were determined by Northern blotting of RNA extracted from exponential phase cultures (OD600 ~0.5) at various times following the addition of rifampicin. Culture identities: E. coli MG1655 (WT) or its Δcrr mutant with or without plasmid pBR322, pCRR or pCRRH91A (nonphosphorylatable EIIAGlc). The RNA half-lives were determined from the linear portions of their decay curves, shown in B and D. Standard deviations of values from two independent experiments are shown. E) CsrB/C steady state levels determined by Northern blotting of RNA from exponentially growing cultures (OD600 ~0.5), as above. F) A model for the Csr regulatory circuitry that includes EIIAGlc activation of CsrD-mediated CsrB/C decay. A broken line indicates an undefined mechanism(s).

Page 51: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

51

Figure 2-2. EIIAGlc (crr) activates CsrB/C decay via CsrD, but does not enhance cellular

CsrD levels. A and B) Decay rates of CsrB (A) and CsrC (B) were determined by Northern blotting of RNA from E. coli MG1655 (WT), ΔcsrD, Δcrr ΔcsrD strains with or without plasmid pBR322, pCRR or pBYH4 (expressing CsrD), as described in Figure 2-1. C) Western blots depicting the effect of crr deletion on the level of CsrD protein. RpoB was used as loading control. CsrD protein levels in Δcrr relative to those in the wild-type strain (WT) are shown at the bottom. Standard derivations from triplicate experiments are indicated.

Page 52: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

52

Figure 2-3. EIIAGlc interacts specifically with the EAL domain of CsrD. A) CsrD variants

used to identify the domain that binds to EIIAGlc. B-D) In vitro assays for binding of EIIAGlc to CsrD variants depicted in panel A. Each reaction contained 8 μM of CsrD variants and 16 μM of EIIAGlc. U and B: unbound and bound proteins. Control reactions were performed in the absence of EIIAGlc to confirm that CsrD varients do not bind nonspecifically to Ni-NTA resin. E) Gel filtration assay of EIIAGlc, EAL, and EIIAGlc-EAL mixture. Proteins were fractionated on a Superdex 200 column (HiLoadTM 16/60, 120 mL). The solid line corresponds to EAL domain alone; the dashed line corresponds to the EIIAGlc-EAL mixture. The chromatogram for EIIAGlc was not shown because this protein displays little absorbance at 280 nm. Arrows indicate elution volumes of molecular weight markers used to calibrate the column (Experimental procedures). F) SDS-PAGE and Coomassie blue staining of proteins from gel filtration chromatography fractions of the EIIAGlc-EAL mixture shown in panel E. Fractions (3 mL) were collected starting at 40 mL.

A B

C D

GGDEF EAL HAMP-like

Transmembrane

Coiled coil

CsrD

CsrDΔTM

CsrDΔEAL

CsrDΔGGDEF

CsrDΔHAMP

CsrDΔCoil

EAL

Binding to

EIIAGlc

ND

+

-

+

+

+

+

U B U B U B U B U B U B U B

+ _ + _ CsrDΔTM CsrDΔEAL

+ _ +

CsrDΔGGDEF

EIIAGlc

M (kDa)

CsrD variants:

EIIAGlc:

250 150 100 75

50

37

25 20

15

_

CsrDΔHAMP CsrDΔCoil

EIIAGlc

U B U B U B U B U B U B U B

+ _ + _ CsrDΔTM

+ _ + M (kDa)

CsrD variants:

EIIAGlc:

250 150 100 75

50

37

25 20

15

_

EIIAGlc

EAL

U B U B U B

+ M (kDa)

EAL:

EIIAGlc: _ +

_ +

250 150 100 75

50

37

25

20

15

Page 53: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

53

Figure 2-3. Continued.

Figure 2-4. Gel filtration chromatography of CsrD variants. Each CsrD variant (1 mg)

was passed through a Superdex 200 column (HiLoadTM 16/60, 120 mL). Arrows indicate elution volumes of molecular weight markers used to calibrate the column (Materials and Methods).

E F 10 11 12 13 14 15 16 17 18

EIIAGlc

EAL 75

50

37

25 20

M (kDa)

15

Page 54: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

54

Table 2-1. Molecular weight of CsrD in solution

Protein Molecular Mass (kDa) Quaternary structurec Calculateda Experimentalb

CsrDΔTM 99.3 359 Tetramer CsrDΔHAMP 95.1 107 Monomer CsrDΔCoil 97.0 107 Monomer CsrDΔGGDEF 80.3 344 Tetramer CsrDΔEAL 69.4 296 Tetramer

a. Molecular mass of the protein calculated from the primary sequence. b. Molecular mass determined using size exclusion chromatography. c. Deduced quaternary structure.

Figure 2-5. CsrD binds only to unphosphorylated EIIAGlc in pull-down assays. MBP-

tagged CsrD protein was bound to amylose resin and then mixed with EIIAGlc

in the nonphosphorylated (N) or phosphorylated state (P) to permit binding reactions to occur. The reactions were processed as described in the Experimental procedures. Control reactions were performed without CsrD to test for nonspecific binding of the two forms of EIIAGlc to amylose resin. Note that the two forms of EIIAGlc were resolved from each other on 15% SDS-PAGE gel. U and B refer to proteins that were unbound vs. bound by the amylose resin.

EIIAGlc:

_ + CsrDΔTM:

CsrD

EIIAGlc-P

EIIAGlc

EI

M (kDa) U B U B U B U B

N P N P

_ +

100 75 50

37

25

20

Page 55: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

55

Figure 2-6. Effects of cAMP-Crp on CsrB/C decay. A and B) Decay rates of CsrB/C

were determined by Northern blotting of RNA from E. coli MG1655 (WT) and isogenic deletion mutants: cyaA, crp, cyaA, crr crp, crr cyaA, crr with or without added 10 mM cAMP, as described in Figure 2-1.

Page 56: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

56

Figure 2-7. Effects of carbon sources on EIIAGlc phosphorylation and CsrB/C decay in

minimal media. A) Western blot depicting the phosphorylation state of EIIAGlc of E. coli MG1655 growing in minimal medium A supplemented with 0.2% glucose, glycerol or succinate. Exponential phase extracts were fractionated in gels with Phos-tag™ reagent and analyzed by Western blot. Relative level of total EIIAGlc was analyzed by western blot without using Phos-tag™ reagent. The percentage of phosphorylated (EIIAGlc-P) and unphosphorylated EIIAGlc (EIIAGlc) and relative total EIIAGlc protein levels are given. B and C) Northern blot depicting CsrB/C decay rates in E. coli MG1655 exponentially growing in minimal medium A supplemented with 0.2% glucose, glycerol or succinate. The RNA half-lives were determined as shown in Figure 2-1.

Page 57: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

57

Figure 2-8. The phosphorylation state of EIIAGlc before and 10 min after shift from LB

broth into minimal media. A and B) Western blot depicting the phosphorylation state and relative level of EIIAGlc in E. coli MG1655 (WT), Δcrr and H91A (mutant strain that carrying a chromosomal EIIAGlcH91A point mutation). Extracts were prepared from cultures grown in LB broth (A) and at 10 min after reinoculation into minimal medium without carbon, with 0.2% glucose or succinate (B). The percentage of phosphorylated EIIAGlc relative to total EIIAGlc was determined as described in Figure 2-7.

Page 58: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

58

Figure 2-9. CsrB/C decay rates and levels after shift from LB to minimal media. A-D)

Decay rates of CsrB (A and B) and CsrC (C and D) determined by Northern blotting of RNA from E. coli MG1655 (WT) and mutant strain H91A (unphosphorylatable EIIAGlc). Strains were grown in LB broth to exponential phase, washed and reinoculated into M9 minimal medium without carbon or with 0.2% glucose or succinate. Rifampicin was added 10 min after inoculation into minimal media and RNA half-lives determined as in Figure 2-1. E) CsrB/C steady state levels determined by Northern blotting of RNA from E. coli MG1655 (WT) in LB broth at exponential growth phase and 10 min after inoculation into minimal media.

A B

C D

E

Half-life (min)

9.4 ± 1.9

2.3 ± 0.1

5.3 ± 1.7

CsrB-WT

No C

Glc

Suc

0 2 4 6 8 16 32

Time (min)

3.2 ± 0.4

2.6 ± 0.1

2.8 ± 0.2

CsrB-H91A Half-life

(min) 0 2 4 6 8 16 32

No C

Glc

Suc

Time (min)

Half-life (min)

13.2 ± 2.0

4.7 ± 0.3

7.7 ± 2.0

CsrC-WT

No C

Glc

Suc

0 2 4 6 8 16 32

Time (min)

5.4 ± 0.2

5.8 ± 0.4

5.9 ± 0.0

CsrC-H91A Half-life

(min) 0 2 4 6 8 16 32

No C

Glc

Suc

Time (min)

CsrB CsrC

1.0 1.0±0.2 0.4±0.1 0.7±0 1.0 0.9±0.1 0.4±0.1 1.4±0.2

LB No C Glc Suc LB No C Glc Suc

Relative

level

Page 59: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

59

Figure 2-10. EIIAGlc and MshH (CsrD ortholog) affect CsrB and CsrD decay in V.

cholerae. A, C and E) Decay rates of CsrB/C/D sRNAs were determined by Northern blotting, after rifampicin addition (time points indicated) to 27°C exponentially growing (OD600 ~0.5) cultures of V. cholerae 01 EI Tor (WT), Δcrr, ΔmshH and Δcrr ΔmshH strains. The RNA half-lives and standard derivations from duplicate experiments were determined as in Figure 2-1. B, D and F) Quantification of signals obtained from two independent experiments, as presented in panels A, C and E. Standard deviations are indicated.

A B

C D

E F

WT

Δcrr

ΔmshH

Δcrr ΔmshH

Time (min) 0 4 8 16 32

CsrB

14.0 ± 1.2

>32

>32

>32

Half-life (min)

CsrD

0 4 8 16 32

10.8 ± 2.0

>32

>32

>32

WT

Δcrr

ΔmshH

Δcrr ΔmshH

Time (min)Half-life

(min)

CsrC

>32

>32

>32

>32

WT

Δcrr

ΔmshH

Δcrr ΔmshH

Time (min) 0 4 8 16 32 Half-life

(min)

Page 60: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

60

Figure 2-11. Proposed model for the effect of carbon availability on CsrB/C decay.

When glucose is present, EIIAGlc is mostly dephosphorylated, it binds to the EAL domain of CsrD and promotes CsrB/C decay. This results in increased CsrA activity to allow expression of genes and pathways needed for rapid growth, e.g. glycolysis. However, when preferred carbon resources have been expended and end-products have accumulated, the BarA-UvrY TCS activates CsrB/C synthesis and CsrD-dependent turnover is repressed. The resulting accumulation of CsrB/C will antagnize CsrA activity and promote the transition from glycolytic metabolism and active growth to gluconeogenesis, glycogen biosynthesis and the formation of a stress resistant phenotype.

UvrY

GGDEF

EAL

Glucose

Glu-6-P

HPr HPr-P

EI-P EI

PEP Pyruvate

P

P

CsrB/C

Carboxylic acids

mRNA translation, stability and transcription

elongation

CsrD

CsrA

Inner membrane

EIIAGlc EIIAGlc

EIIBGlc

EIICGlc

BarA RNase E

EIIAGlc

Page 61: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

61

CHAPTER 3 EXPLORING THE MOLECULAR MECHANISM BY WHICH CsrD FACILITATES CsrB/C

TURNOVER

Introduction

In the last decade, the importance of sRNAs in gene expression has been

increasingly recognized in prokaryotes and eukaryotes (Gottesman & Storz, 2011, Storz

et al., 2011, Filipowicz et al., 2008, Vaucheret, 2006). While most of the bacterial

sRNAs act by base-pairing with mRNAs and altering gene expression, a small number

of sRNAs directly bind proteins and modulate protein activities by mimicking their

nucleic acid substrates. For example, Csr/Rsm sRNAs control gene expression and

cellular phenotypes indirectly by antagonizing CsrA/RsmA activities (Romeo, 1998,

Babitzke & Romeo, 2007, Romeo et al., 2013, Vakulskas et al., 2015). CsrA/RsmA are

RNA binding proteins that prefer to bind GGA motifs within apical loops of hairpin

structures in sRNAs or in the untranslated leader region or early coding region of target

mRNAs (Dubey et al., 2005, Holmqvist et al., 2016). CsrA/RsmA activate or repress

expression of target mRNAs by affecting the translation efficiency (Baker et al., 2002,

Baker et al., 2007, Dubey et al., 2003), transcription elongation (Figueroa-Bossi et al.,

2014), and/or transcript stability (Liu et al., 1995, Wang et al., 2005, Yakhnin et al.,

2013). In E. coli, CsrA activity is mainly controlled by the steady state levels of two non-

coding sRNAs CsrB and CsrC. These sRNAs contain many high-affinity CsrA binding

sites that sequesting CsrA from binding to its lower affinity target mRNAs (Liu et al.,

1997, Weilbacher et al., 2003).

Levels of CsrB/C are positively and negatively regulated by diverse regulators at

the synthesis level (Chavez et al., 2010, Huang et al., 2008, Edwards et al., 2011,

Suzuki et al., 2002, Camacho et al., 2015, Vakulskas et al., 2014, Zere et al., 2015,

Page 62: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

62

Pannuri et al., 2016) and turnover level (Suzuki et al., 2006, Leng et al., 2016,

Vakulskas et al., 2016) in response to different environmental stimuli. This regulation

permits bacteria cell to fine-tune CsrA activity and gene expression for proper cellular

stress response.

Turnover of CsrB is mediated by RNase E and PNPase. The cleavage of CsrB is

initiated by RNase E at a 9 nt region located immediately upstream of intrinsic

terminator (Vakulskas et al., 2016). Deletion of this region or two adjacent adenine

residues within this region virtually eliminated CsrB turnover. PNPase acts to eliminate

the products from RNase E cleavage (Suzuki et al., 2006). In addition to RNase E and

PNPase, CsrD, EIIAGlc and CsrA are identified as key factors in regulating CsrB/C

turnover, as shown in Figure 4-1 (Suzuki et al., 2006, Leng et al., 2016, Vakulskas et

al., 2016). Deletion of CsrD significantly stabilized CsrB/C from half-lives of 1-2 min to

more than 32 min, indicating that CsrD is required for the rapid degradation of CsrB/C.

CsrD is not a nuclease (Suzuki et al., 2006), but is a membrane-bound signaling protein

containing GGDEF and EAL domains, which typically catalyze the synthesis and

turnover of the secondary messenger c-di-GMP. However, CsrD lacks critical catalytic

residues in these domains and displays no activity in c-di-GMP metabolism (Suzuki et

al., 2006). We recently showed that CsrD is responsible for the activation of CsrB/C

decay by EIIAGlc in the presence of a preferred carbon source (Fig. 3-1) (Leng et al.,

2016). When glucose is being actively transported by the PTS system,

unphosphorylated EIIAGlc accumulates and binds to the EAL domain of CsrD. Binding of

EIIAGlc allosterically activates CsrD activity to facilitate CsrB/C turnover (Leng et al.,

2016). Furthermore, CsrD and CsrA showed antagonistic roles on CsrB turnover (Fig. 3-

Page 63: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

63

1). Specifically, the binding of CsrA to two GGA sites adjacent to the initial RNase E

cleavage region stabilized CsrB by protecting it from RNase E cleavage in a csrD

mutant and in vitro. However, CsrA disruption did not change CsrB decay rate in the

csrD WT strain (Vakulskas et al., 2016). In addition, even though CsrD is required for

the rapid degradation of CsrB in wild-type strain (Suzuki et al., 2006), it is not necessary

in csrA mutant strain, where CsrB is not covered by CsrA and can be rapidly degraded

by RNase E (Vakulskas et al., 2016). These data together suggest that CsrD has

evolved from a c-di-GMP metabolizing enzyme to become a device for decoupling Csr

sRNA turnover from the direct influence of CsrA binding.

So far, exactly how CsrD permits RNase E access to CsrB is not yet clear.

Previous study showed that the GGDEF domain of CsrD was homologous to several

RNA binding proteins, and CsrD bound to CsrB/C sRNAs in vitro, albeit nonspecifically

(Suzuki et al., 2006). Thus, we proposed that CsrD might directly bind CsrB/C, which in

turn releases CsrA and exposes the RNase E cleavage sites. Nevertheless, it is also

possible that CsrD works indirectly through other factors.

Materials and Methods

Media and Growth Conditions

Bacterial strains utilized in this study are listed in Table B-1. E. coli strains were

routinely grown in LB medium (1% tryptone, 1% NaCl, and 0.5% yeast extract) with

appropriate antibiotics when needed: ampicillin (100 μg mL−1), kanamycin (50 μg mL−1),

and gentamicin (10 g μg mL−1). For immunoprecipitation assays, overnight culture

grown in LB broth were inoculated into Kornberg medium (1.1% K2HPO4, 0.85%

KH2PO4, 0.6% yeast extract and 0.5% glucose) to an OD600 of 0.01, and their growth

was monitored at OD600. Strains carrying rne-1 were incubated at 30°C and then shifted

Page 64: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

64

to 43-44°C before culture collection to inactive RNase E. Strains deficient in enolase

(PBAD-eno without arabinose) were grown in M9 medium containing 0.2% succinate and

0.02% glycerol.

Construction of Strains and Plasmids

E. coli gene deletions were created by the standard P1vir transduction procedure

or the lambda Red system as described (Datsenko & Wanner, 2000). The resistance

markers introduced into mutant strains were eliminated using an FLP expression

plasmid pCP20 when necessary (Datsenko & Wanner, 2000). The cat-PBAD-eno was

moved from TM447 (Morita et al., 2004) to MG1655 by P1 vir transduction.

Plasmids and DNA oligonucleotides used in this study are listed in Tables B-2

and B-3. For constructing plasmid p3FLAG-CsrD expressing CsrD containing a 3×FLAG

tag at C-terminal end, plasmid pBYH4 expressing WT-CsrD under the control of its

native promoter (Suzuki et al., 2006), was amplified with primer pair pBR3FLAG-F/R

(containing sequence encoding 3×FLAG tag), and the PCR product was re-ligated using

T4 DNA ligase (NEB). For plasmid p2SLStrep-CsrB expressing the Streptotag-CsrB,

plasmid pCsrB expressing the WT-CsrB under the control of its native promoter

(Vakulskas et al., 2016), was amplified with primer pair pBRStrep-CsrB-F/R (containing

Streptotag sequence), and PCR product was re-ligated.

Gel Mobility Shift Assay

RNAs (CsrB, CsrC, rpsT and GlmZ) were synthesized and end-labeled as

previously described (Suzuki et al., 2006). RNAs were gel purified, suspended in TE

buffer (10 mM Tris-HCl, pH 8.0, 1 mM EDTA) and renatured by heating to 70°C for 5

min and slow cooling to room temperature. Binding reactions (10 μl) contained 10 mM

Tris-HCl, pH7.5, 125 mM KCl, and 2 mM MgCl2, 32.5 ng of yeast RNA, 7.5% glycerol, 1

Page 65: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

65

mM dithiothreitol (DTT), 4 U of RNase inhibitor (Ambion), 0.6 nM CsrB RNA, purified

MBP-tagged CsrDΔTM, His-tagged EIIAGlc and/or His-tagged CsrA and 0.1 mg mL-1

xylene cyanol. For competition studies, assays were carried out with unlabeled RNA

competitors (CsrC, rpsT and GlmZ). Reaction mixtures were incubated for 30 min at

37°C to allow protein–RNA complex formation. Samples were then fractionated on 5%

native Bis-Tris gels. Radioactive bands were visualized and quantified using a

phosphorimager and Image Quant software.

In vitro RNase E Cleavage Assays

RNase E assays were performed by first incubating 0.05 nM RNA at 90°C for 3

min in RNase E reaction buffer (25 mM Tris–HCl, pH 7.9, 5 mM MgCl2, 60 mM KCl, 100

mM NH4Cl, 15 mM DTT and 7.5% glycerol), followed by cooling to 25°C over 10 min.

The reactions were then incubated at 25°C for 10 min in the presence or absence of

CsrA. RNase E was added to 37.5 nM and reactions were incubated at 25°C for an

additional 10 min. Reactions were stopped with two volumes of RNA loading buffer,

incubated at 65°C for 10 min to inactivate CsrA and subsequently kept on ice prior to

electrophoresis. Samples were separated by electrophoresis on 8% acrylamide gels

(19:1) containing 7 M urea for 3 h at 45 Watts. Gels were dried onto chromatography

paper and subjected to autoradiography.

Affinity Purification of CsrD and its Binding RNAs

Strains carrying p3FLAG-CsrD or pBYH4 were grown in 500 mL Kornberg

medium at 37°C, 250 rpm. At OD600 of 1.0, formaldehyde (0.5% final concentration) was

added and the cultures were incubated for 10 min at 30°C, 150 rpm. Glycine (0.125M

final concentration, pH 8.0) was subsequently added to quench the crosslinking reaction

and the samples were mixed with gentle swirling for 5 min at room temperature. The

Page 66: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

66

cells were harvested by centrifugation (7K rpm, 10 min), washed twice with ice-cold 1x

PBS, suspended in 20 mL lysis buffer (20 mM HEPES pH 7.9, 150 mM KCl, 10%

glycerol, 0.5% Triton X-100) with EDTA-free protease inhibitor cocktail (Roche

Diagnostics) and lysed using 4 rounds of 10 sec sonication (power 30%, 0.5 sec pulse

on, 0.5 sec off) on ice. Lysates were cleared by centrifugation (14K rpm, 30 min) and

filtration over Millex-HV 0.45 m PVDF filter-units (Millipore) before mixing with 200 μL of

pre-washed ANTI-FLAG M2 beads (Sigma). Cell lysate with beads were rotated for 4

hours at 4°C to immunoprecipitate FLAG-tagged CsrD. The beads were then washed

three times with 4 mL high salt buffer (lysis buffer with 0.75M KCl), three times with 4mL

lysis buffer and once with elution buffer (20 mM HEPES pH 7.9, 150 mM KCl, 10%

glycerol). At last, CsrD-RNA complexes were eluted from the beads with 3X FLAG

Peptide (sigma), concentrated with Amicon Ultra-0.5 mL Centrifugal Filters (3kDa,

Millipore), and the formaldehyde crosslinking was reversed by heating at 95°C for 20

min. Afterward, RNAs were isolated by phenol chloroform extraction and ethanol

precipitation of the aqueous phase and proteins were isolated by acetone-precipitation

of the organic phase and dissolved in protein loading buffer. CsrB levels in samples

were analyzed by Northern blots and proteins were size-separated by 4–20% Mini-

PROTEAN ® TGX™ Precast gels (Biorad) and visualized by silver staining (kit from

Invitrogen).

Affinity Purification of in vivo Synthesized CsrB and its Associated Proteins

Strains carrying p2SLStrepto-CsrB or pCsrB were grown in 500 mL Kornberg

medium at 30°C, 250 rpm. At exponential phase of growth (OD600 of 0.5-0.6), cultures

were incubated at 50°C for ~15 min (temperature of culture reached 43-44°C) to

inactive RNase E. The cells were then harvested by centrifugation, washed once with

Page 67: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

67

ice-cold 1x PBS, and suspended in 10 mL lysis buffer (10 mM Tris-HCl pH 7.8, 200 mM

KCl, 1 mM MgCl2, 5% glycerol, 0.5% Triton X-100) with EDTA-free protease inhibitor

cocktail (Roche Diagnostics) and SUPERase RNase Inhibitor (Thermo Fisher).

Afterward, Cells were lysed using 4 rounds of 10 sec sonication (power 30%, 0.5 sec

pulse on, 0.5 sec off) on ice. Lysates were then cleared by centrifugation (30 min, 14K

rpm) and filtration over Millex-HV 0.45 m PVDF filter-units (Millipore).

Dihydrostreptomycin (Sigma) coupled to epoxy-activated sepharose 6B (Sigma)

were prepared as previously described (Bachler et al., 1999, Windbichler & Schroeder,

2006) and used to pull down Strepto-CsrB. For preparation of the affinity column, 1 mL

slurry of the resin was applied to Poly-Prep® Chromatography Columns (BioRad). The

prepared column was washed twice with 6 mL lysis buffer. Subsequently, the cleared

bacterial lysate was loaded onto the column and incubated at 4°C for 3 hours on a

rotating wheel, followed by three washes with 6 mL washing buffer (10 mM Tris-HCl pH

7.8, 200 mM KCl, 1 mM MgCl2, 5% glycerol, 0.5% Triton X-100) and one time wash with

elution buffer (10 mM Tris-HCl pH 7.8, 200 mM KCl, 1 mM MgCl2, 5% glycerol). The

beads were eluted twice with 600 μL of freshly prepared 10 μM streptomycin in elution

buffer and elutes were concentrated with Amicon Ultra-0.5 mL Centrifugal Filters (3kDa,

Millipore) to 300 μL. RNA was extracted with phenol-chloroform and ethanol

precipitation of the aqueous phase. For protein isolation, the organic phase was

subjected to acetone precipitation and the pellet was dissolved in protein loading buffer.

Mass Spectrometry

For mass spectrometric analysis of CsrB associated proteins, precipitated

proteins were size-separated on a 4–20% Mini-PROTEAN ® TGX™ Precast gels

(Biorad) and observed with silver staining. Bands of interest were cut-out and the

Page 68: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

68

excised gel pieces were destained with 50 mM ammonium bicarbonate, pH 8.5, and

then reduced by 10 mM Dithiothreitol (DTT, Sigma-Aldrich Inc., St. Louis, MO) at 37 ˚C

for 1h, followed by alkylation by 20 mM iodoacetamide in the dark for 30 min. Trypsin

(Sigma-Aldrich Inc.) was added for digestion (w/w for enzyme: sample = 1 : 100)

overnight at 37 °C. The digested peptides were desalted using micro ZipTip mini-

reverse phase (Millipore Inc., Billerica, MA), and then lyophilized to dryness.

Peptides derived from the protein samples and resuspended in 0.1% formic acid

for mass spectrometric analysis. The bottom-up proteomics data acquisition was

performed on an EASY-nLC 1200 ultra-performance liquid chromatography system

(Thermo Scientific Inc., Odense, Denmark) connected to an Orbitrap Fusion Tribrid

instrument equipped with a nano-electrospray source (Thermo Scientific Inc, San Jose,

CA). The peptide samples were loaded to a C18 trapping column (75 μm i.d. × 2 cm,

Acclaim PepMap® 100 particles with 3 μm size and 100 Å pores) and then eluted using

a C18 analytical column (75 μm i.d. x 15 cm, 3 μm particles with 100 Å pore size). The

flow rate was set at 300 nL/minute with solvent A (0.1% formic acid in water) and

solvent B (0.1% formic acid and 99.9% acetonitrile) as the mobile phases. Separation

was conducted using the following gradient: 2 - 35 % of B over 0 - 20 min; 35 - 98 % of

B over 20 – 21 min, and isocratic at 98% of B over 21-35 min. The full MS1 scan (m/z

350 - 2000) was performed on the Orbitrap with a resolution of 120,000 at m/z 400. Raw

data were analyzed using Mascot (Matrix Science, London, UK; version 2.4.1) against

E. coli K12 MG1655 20161109 database and Scaffold (version Scaffold_4.2.1,

Proteome Software Inc., Portland, OR) was used to validate MS/MS based peptide and

Page 69: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

69

protein identifications. Protein identities were based on a threshold of 99.0 % probability

and < 0.1 % False Discovery Rate (FDR).

Protein Purification and Western Blots

Procedures for purification of His-tagged EIIAGlc and MBP-tagged CsrD and

Western blots were as described in Chapter 2 ‘Materials and Methods’. Purification of

RNase E and CsrA was as described previously (Vakulskas et al., 2016).

RNA Purification and Northern Blots

Total RNA from 20 μL resuspended intact cells, lysate and flow-through were

extracted using hot phenol chloroform (Georgellis et al., 1992) and purified with ethanol-

precipitation. Procedures for Northern blots were as described in Chapter 2 ‘Materials

and Methods’.

Results and Discussion

EIIAGlc is not Capable to Stimulate the Binding of CsrD to CsrB in vitro

A recent study indicated that EIIAGlc directly binds CsrD and allosterically

stimulates its activity (Leng et al., 2016). We therefore hypothesized that EIIAGlc might

promote the binding between CsrD and CsrB if CsrD functions as a CsrB binding

protein. To verify this, we compared the binding affinities and specificities of CsrD to

CsrB in the presence and absence of EIIAGlc using EMSA. Fig. 3-2A showed that CsrD

binds CsrB in vitro, but the binding affinity between CsrD and CsrB is much lower than

that between CsrA and CsrB (Kd value of 0.8-1.6 nM) (Weilbacher et al., 2003). Addition

of EIIAGlc did not alter the binding affinity (Fig. 3-2B). The specificity of the CsrD-CsrB

interaction was investigated by performing competition experiments with specific

(sRNAs CsrB and CsrC) and non-specific (mRNA rpsT and sRNA GlmZ) unlabelled

RNA competitors (Fig. 3-2C). We found that while CsrB, CsrC and rpsT transcripts

Page 70: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

70

competed for CsrD binding, non-specific sRNA GlmZ did not compete (Fig. 3-2C),

suggesting that CsrD binds to CsrB with some specificitiy. Moreover, the specificity of

binding was not altered by addition of EIIAGlc (Fig. 3-2D). This suggests that CsrD binds

to CsrB with some specificitiy and EIIAGlc did not alter the specificity of this binding. All

together, EIIAGlc does not promote the binding affinity or specificity of CsrD to CsrB in

vitro.

CsrA Influences the CsrD-CsrB Interaction in vitro

Another recent study revealed that CsrD is required for CsrB turnover when the

RNase E-mediated CsrB cleavage can be protected by CsrA (Vakulskas et al., 2016).

Then, it is possible that CsrD prefers to bind CsrB in complex with CsrA. Specifically,

binding of CsrA to CsrB may modulate the sencondary or tertiary structure of CsrB and

render it accessible to CsrD binding. To test this, we examined the effect of CsrA on the

binding of CsrD to CsrB using EMSA. As expected, the binding of CsrA to CsrB was

observed in the absence of CsrD (the last lane, Fig. 3-3B). As the concentration of CsrD

increased, an additional shift was observed (Fig. 3-3B). Nevertheless, this shift does not

correspond to the CsrD-CsrB complex shown in Fig. 3-3A, where CsrA is not present.

Addition of EIIAGlc to the binding reaction did not alter the shift pattern (Fig. 3-3C). This

data suggest that addition of CsrA influences the CsrD-CsrB binding complex. We

speculate that the additional shift in Fig. 3-3B might represent a complex formed by

CsrD, CsrB and CsrA, but this needs to be further investigated by western blot analysis.

CsrD cannot Facilitate CsrA-mediated Protection of CsrB Cleaved by RNase E

CsrB contains 22 GGA motifs, most of which are presumed to be CsrA binding

sites (Liu et al., 1997, Weilbacher et al., 2003). Although the binding affinity of CsrD with

CsrB is much lower than that of CsrA with CsrB (Fig. 3-2A) (Weilbacher et al., 2003), we

Page 71: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

71

cannot rule out the possibility that CsrD competes with CsrA for binding to the two GGA

motifs adjacent to the RNase E cleavage site, and expose CsrB for RNase E cleavage.

Thus, we examined whether CsrD facilitates the in vitro cleavage of CsrA-protected

CsrB by RNase E. This assay was performed with a shorter fragment of CsrB (+226 to

+369 relative to the transcription start site), whose turnover remains CsrD-dependent.

Our data indicated that RNase E cleaves the CsrB fragment mainly at nucleotides A327

and A331, which are located within the RNase E initial cleavage region ‘A’ (Vakulskas et

al., 2016). Addition of CsrA completely blocked the cleavage of CsrB by RNase E at two

major cleavage sites. However, inceasing concentration of CsrD with the help of EIIAGlc

did not facilite the cleavage of CsrA-protected CsrB by RNase E (Fig. 3-4). This

suggests that rather than functioning via a direct binding to CsrB, CsrD might require

other factor(s) in addition to EIIAGlc to render the CsrA-protected CsrB accessible to

RNase E cleavage.

CsrD is Unlikely to Bind CsrB in vivo

We further tested the in vivo binding of CsrD to CsrB using formaldehyde

crosslinking and immunoprecipitation, which could potentially capture transient and

unstable binding. If CsrD binds to CsrB in vivo, CsrB is expected to be enriched along

with FLAG-tagged CsrD. A 3×FLAG tag was added to the C terminus of CsrD, which

does not affect the activity of CsrD on CsrB turnover (Fig. 3-5A). FLAG-tagged CsrD

was expressed under the control of its native promoter on a multi-copy plasmid pBR322

in a strain with a csrD genomic deletion. Plasmid pBYH4 expressing untagged CsrD

was used as the negative control. The crosslinked CsrD-RNA complex was captured

and subsequently uncrosslinked by heating at 95 °C for 20 min. Enrichment of CsrD and

CsrB was determined by silver staining and Northern blot, respectively. Fig. 3-5B

Page 72: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

72

showed that FLAG-tagged CsrD was enriched in samples expressing tagged CsrD

comparing to the negative control (lane 1 and 2). Samples without formaldehyde

treatment (lane 3) gave a stronger enrichment of FLAG-tagged CsrD, suggesting that

formaldehyde treatment somehow inhibited the binding of FLAG-tagged CsrD to the

beads. Surprisingly, while CsrD was strongly enriched, no enrichment of CsrB was

detected (Fig. 3-5C). These data suggest that CsrD might not directly bind to CsrB in

vivo. Although it is also possible that binding between CsrD and CsrB is very transient

and could not be captured with the approach we used.

CsrB Associated Proteins and their Influence on CsrB Turnover

To further investigate binding of CsrD with CsrB in vivo, we used a reciprocal

approach, by pulling down proteins from the cell via a tagged CsrB. This approach will

also provide information on the proteins associated with CsrB and may help to identify

the missing factor(s) involved in CsrB/C turnover.

Our previous work showed that the second stem loop of CsrB was not essential

for the CsrD-mediated turnover of CsrB (Vakulskas et al., 2016). This stem loop was

partially substituted with an RNA aptamer Streptotag that specifically binds to

streptomycin with high binding affinity (Bachler et al., 1999, Dangerfield et al., 2006).

Tagged CsrB was expressed under the control of its own promoter on plasmid pBR322

in a strain lacking the endogenous csrB. CsrB decay assays revealed that the RNA

aptamer did not influence the expression of CsrB, and slightly stabilized CsrB from a

half-life of 4.3 min to 8.3 min (Fig. 3-6A). Nevertheless, decay of the tagged CsrB

remains CsrD dependent (Fig. 3-6A). We tried to capture the transient CsrB-protein

complex with formaldehyde crosslinking, but this treatment severely inhibited the

binding of tagged CsrB to the beads (data not shown). Then we performed this

Page 73: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

73

experiment using strains in rne-1 background and the bacterial cultures were shifted

from 30°C to 43-44°C prior to affinity purification to inactive RNase E. Under this

condition, CsrB-protein complex could potentially be stabilized. After affinity purification,

enrichment of CsrB and CsrD was analyzed by Northern blot and Western blot,

respectively. Northern blot results illustrated that while 10% of the total CsrB was

captured using this approach (data not shown), CsrD was not recovered by CsrB,

further supporting our previous finding that CsrD might not directly bind CsrB in vivo

(Fig. 3-6C). CsrB associated proteins were separated on SDS-PAGE gel and observed

with silver staining, as shown in Fig. 3-6B. Of particular interest, in addition to CsrA, the

well-characterized CsrB binding protein, two other bands A and B showed strong

enrichments in the strain expressing tagged CsrB (lane 1 and 2) relative to the negative

control with the wild-type CsrB (lane 3). Using LC-MS/MS, we analyzed all the proteins

from the two bands, as shown in Tables 3-1 and 3-2. Unexpectedly, no protein from

band A was identified to have a size corresponding to band A. In band B, enolase

represents the most abundant protein and has a size matching that of band B. Enolase

is a glycolytic enzyme that catalyzes the interconversion of 2-phospho-D-glycerate and

phosphoenolpyruvate and is a component of the RNA degradosome. The role of

enolase in RNA degradosome is not very clear, but it is required for the destabilization

of ptsG mRNA in response to glucose-6-phosphate (Morita et al., 2005, Morita et al.,

2004). Given that enolase in eukaryotes is capable to bind RNA (Hernández-Pérez et

al., 2011), we hypothesized E. coli enolase directly binds CsrB and facilitates its

turnover. To examine the possible role of enolase in CsrB turnover, we compared CsrB

decay rates in wild-type strain and a mutant strain growing in M9 minimal medium

Page 74: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

74

supplemented with succinate and glycerol, in which the eno gene is under the control of

an arabinose-inducible promoter PBAD (Morita et al., 2004). In the absence of arabinose,

enolase should not be expressed and the decay rate of CsrB was moderately

decreased in enolase deficient strain (half-life of 8.2 min) as compared to in the wild-

type strain (half-life of 5.0 min) (Fig. 3-7A). Since the bacterial culture used for

immunoprecipitation assay was grown in Kornberg medium (Fig. 3-6), we also

examined the effect of enolase on CsrB turnover in Kornberg medium. We first

transferred the exponentially growing culture in M9 minimal medium to Kornberg

medium and then compared CsrB decay rates in enolase deficient strain and wild-type

strain 10 min after the transfer. CsrB decay rates were faster in Kornberg medium than

in the minimal medium with succinate and glycerol likely due to the presence of glucose

(Fig. 3-7A and B). However, decay rates of CsrB were essentially identical in strains

lacking enolase (half-life of 2.3 min) or with enolase (half-life of 2.2 min), suggesting that

enolase has little or no role in CsrB turnover (Fig. 3-7B). Whether enolase directly binds

to CsrB remains to be further determined, but based on our finidng that enolase has no

effect on CsrB turnover and it directly interacts with RNase E (Mackie, 2013), it is

possible that enolase associates with CsrB indirectly via RNase E.

Meanwhile, we also observed other dramatically enriched proteins in amounts

less abundant than enolase and with sizes that do not correspond to bands A and B

(Tables 3-1 and 3-2). They are represented by RNase E, poly(A) polymerase (PAP I),

RhlB and multiple ribosomal proteins. It is known that RNase E directly binds CsrB and

cleaves it. RhlB, the component of of RNA degradosome comprising of RhlB, PNPase,

enolase and RNase E, could have been recruited by RNase E. PAP I promotes

Page 75: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

75

degradation of sRNA and mRNA by adding a single stranded poly(A) tail at the 3’ end of

the RNAs (Xu et al., 1993, Hajnsdorf et al., 1995), but it does not influence CsrB

turnover (Suzuki et al., 2006). It is also reported to associate with PNPase and might

have been recuirted via the RNA degradosome (Mohanty et al., 2004). Ribosomal

proteins were capable to interact directly with RNase E (Tsai et al., 2012), so they might

directly bind the multiple AGGA motifs within CsrB, or indirectly associate with CsrB via

its interaction with RNase E. Overall, copurification of these proteins suggest that CsrB

might provide a platform for binding of a specific set of proteins, but whether these

proteins are functionally relevant to CsrB decay or have other functions remains to be

further explored.

Conclusion

Previous study demonstrated that CsrD facilitates CsrB turnover by overcoming

the CsrA mediated protection and promoting the initinal cleavage of CsrB by RNase E.

This study suggested that CsrD might not work as a RNA binding protein to render

CsrA-protected CsrB accessible to RNase E cleavage. In vitro assays suggest that

other factor(s) in addition to EIIAGlc and CsrA might be required to transmit the

regulatory role of CsrD to CsrB turnover. In addition, proteins associated with CsrB were

detected in this study, but whether these interactions have any physiological functions,

such as affecting CsrB turnover, needs to be further determined.

Page 76: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

76

Figure 3-1. CsrB decay is regulated in response to carbon availability through its effect

on CsrA and CsrD antagonism (Vakulskas et al., 2016). The phosphorylation state of the PTS protein EIIAGlc serves as an indicator of carbon availability. P-EIIAGlc predominates when a preferred carbon source such as glucose is unavailable, and this form is unable to bind to CsrD. During glucose transport, EIIAGlc becomes dephosphorylated and able to bind to CsrD and potentiate CsrB decay. CsrA binding to CsrB protects it from RNase E cleavage in the absence of CsrD-EIIAGlc. A broken line indicates that the molecular mechanism of CsrD remains to be determined. RNase E, PNPase and other nucleases degrade CsrB to nucleotides (NTDs).

Page 77: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

77

Figure 3-2. EIIAGlc has no effect on the binding of CsrD to CsrB RNA in vitro. A and B)

EMSA experiments were carried out with 0.6 nM labeled CsrB and increasing concentrations of CsrD (0-320 nM) in the absence or presence of EIIAGlc. C and D) Competition assays were performed using 0.6 nM labeled CsrB with indicated concentration of CsrD and EIIAGlc in the absence or presence of 100-fold and 1,000-fold molar excess of unlabeled competitor sRNAs, CsrB, CsrC, and GlmZ and mRNA, rpsT.

Page 78: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

78

Figure 3-3. CsrA influences the CsrD-CsrB complex in vitro. EMSA experiments were

carried out with 0.6 nM labeled CsrB and increasing concentrations of CsrD (0-320 nM) in the absence or presence of CsrA and EIIAGlc.

0 10 20 40 80 120 160 250 320 CsrD (nM) 0 10 20 40 80 120 160 250 320 0 CsrD (nM)

CsrA (nM) 0 500

A B

C

0 10 20 40 80 120 160 250 320 0 CsrD (nM)

CsrA (nM) 0 500

EIIAGlc(nM) 0 1280

Page 79: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

79

Figure 3-4. Effect of EIIAGlc and CsrD on RNase E-dependent cleavage of CsrB in vitro.

This experiment was performed with the +226 CsrB RNA (CsrB alleles starting from nucleotide +226 of CsrB to the end of csrB gene), 0.5 mM CsrA, 37.5 nM RNase E, EIIAGlc (0.5, 2 or 6 μM), and CsrD (0.125, 0.5 or 1.5 μM) as indicated. Premixed EIIAGlc-CsrD complex or each protein individually was added to reactions and incubated at 25 °C for 10 min. CsrA and RNase E were then sequentially added to reactions and cleavage was allowed to proceed for 10 min at 25 °C. Control experiments were performed with RNA only. Partial alkaline hydrolysis (OH) and RNase T1 digestion (T1) ladders are also indicated. Numbering is with respect to the full length CsrB sequence.

Page 80: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

80

Figure 3-5. CsrB is not copurified with FLAG-tagged CsrD. A) Northern blot depicting

the CsrB decay rates in MG1655 ΔcsrD strains carrying pBYH4 or p3FLAG-CsrD, which express WT CsrD and FLAG-tagged CsrD, respectively. B) Proteins co-purifying with FLAG-tagged CsrD were observed by silver staining. 1, MG1655 ΔcsrD carrying pBYH4 with formaldehyde crosslinking; 2, MG1655 ΔcsrD carrying p3FLAG-CsrD with formaldehyde crosslinking; 3, MG1655 ΔcsrD carrying p3FLAG-CsrD without formaldehyde crosslinking. C) Northern blot depicting CsrB levels before, during and after the affinity purification. RNA samples were prepared from intact cells before lysate preparation (Tot), from the cleared lysate (Lys), from the flow-through fraction (FT) and from the eluates (Elu1 and Elu2). The amount of RNA loaded in Tot, Lys, FT each corresponds to 30 μL of bacterial culture, in Elu1 and Elu2 correspond to 4.5 mL and 60 mL of bacterial culture, respectively.

1 2 3 1 2 3 1 2 3 1 2 3 1 2 3

Tot Lys FT Elu1 Elu2

A

B

C

CsrDFLAG

1 2 3

0 2 4 8 16 0 2 4 8 16 Time (min)

CsrDWT CsrDFLAG

CsrB

Page 81: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

81

Figure 3-6. CsrD is not recovered by Strepto-CsrB in vivo. A) Streptotag does not affect

expression of CsrB and the CsrD-mediated turnover of CsrB. Rifampcin was added to exponentially growing cultures of MG1655 ΔcsrB and MG1655 ΔcsrB ΔcsrD and decay rates of CsrB were determined by Northern blots as described in Chapter 2 ‘Materials and Methods’. B) Proteins co-purifying with Strepto-CsrB were observed by silver staining. 1, MG1655 rne-1 ΔcsrB harboring p2SLStrep-CsrB; 2, MG1655 rne-1 ΔcsrB ΔcsrD harboring p2SLStrep-CsrB; 3, MG1655 rne-1 ΔcsrB harboring pCsrB. C) Western blot depicting CsrD levels before, during and after the affinity purification. Proteins samples were prepared from intact cells before lysate preparation (Tot), from the cleared lysate (Lys), from the flow-through fraction (FT) and from the eluates (Elu1 and Elu2). The amount of proteins loaded in Tot, Lys, FT each corresponds to 250 μL of bacterial culture, in Elu1 and Elu2 correspond to 16.5 mL and 250 mL of bacterial culture, respectively.

CsrB

1 2 3

A?

B?

CsrA

kDa

75

50

37

25 20 15

100

A

C

B

0 2 4 8 16 0 2 4 8 16

0 4 8 16 32 0 4 8 16 32

WT-CsrB Strepto-CsrB

WT-CsrB Strepto-CsrB

csrB

csrB csrD

Half-lives

Half-lives

4.3 min 8.3min

>32min >32min

ΔcsrD WT-CsrB Strepto-CsrB

Tot Tot Lys FT Elu1 Elu2 Tot Lys FT Elu1 Elu2

Page 82: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

82

Table 3-1. Proteins co-purifying with Strepto-CsrB identified from band A by mass-spectrometry

Protein Gene MW (kDa)

Ratio of Total Unique Spec

Counts

Coverage (%)

Enolase eno 46 3 6.5 RNase E rne 118 3 13

RNA helicase RhlB

rhlB 47 3 7.4

50S ribosomal protein L2

rplB 30 3 17

50S ribosomal protein L3

rplC 22 5 23

50S ribosomal protein L15

rplO 15 6 40

50S ribosomal protein L15

rplI 16 4 35

50S ribosomal protein L21

rplU 12 4 37

50S ribosomal protein L1

rplA 25 3 17

30S ribosomal protein S3

rpsC 26 3 18

30S ribosomal protein S5

rpsE 18 4 34

Protein identities were based on a thresholdof 99.0 % probability and < 0.1 % False Discovery Rate (FDR).

Page 83: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

83

Table 3-2. Proteins co-purifying with Strepto-CsrB identified from band B by mass-spectrometry

Protein Gene MW (kDa)

Ratio of Total Unique Spec Counts

Coverage (%)

R1 R2 R1 R2

Enolase eno 46 3 9 41 64 Poly(A) polymerase

I pcnB 54 5 16 12 27

RNase E rne 118 8 7 8 8 50S ribosomal

subunit protein L9 rplI 16 3 10 40 61

50S ribosomal subunit protein L3

rplC 22 4 3 17 30

50S ribosomal subunit protein L21

rplU 12 3 5 25 32

Experiment was done in replicate. R1, repeat 1; R2, repeat 2. Protein identities were based on a thresholdof 99.0 % probability and < 0.1 % False Discovery Rate (FDR)

Figure 3-7. Enolase has little or no effect on CsrB turnover. Decay rates of CsrB were

determined by Northen blotting of RNA from E. coli MG1655 (WT) and PBAD- eno strains. A) Rifampicin was added to bacterial cultures growing in M9 minimal medium containing 0.2% succinate and 0.02% glycerol entering exponential phase. B) Strains were grown in M9 minimal medium containing 0.2% succinate and 0.02% glycerol to exponential phase, washed and reinoculated into Kornberg medium. Rifampicin was added 10 min after the reinoculation. RNA half-lives were determined as in Figure 2-1.

WT

0 2 4 6 8 16 32 Time (min) Half-lives (min)

5.0

8.2

WT

0 2 4 6 8 16 32 Time (min) Half-lives (min)

2.2

2.3

A

B

PBAD-eno

PBAD-eno

Page 84: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

84

CHAPTER 4 EPISTASIS ANALYSIS USING RNA-SEQ (EPI-SEQ) TO EXPLORE THE REGULATORY ROLE OF CsrD

Introduction

The Csr (Rsm) system is a global regulatory system that is conserved among

Gammaproteobacteria (Vakulskas et al., 2015, Zere et al., 2015). It controls complex

phenotypes, including glycogen metabolism (Baker et al., 2002, Romeo et al., 1993),

biofilm formation (Jonas et al., 2008, Sterzenbach et al., 2013, Wang et al., 2005),

motility (Wei et al., 2001, Yakhnin et al., 2013), and virulence (Bhatt et al., 2009,

Martínez et al., 2011, Vakulskas et al., 2015). The Csr system in E. coli consists of the

RNA binding protein CsrA, the inhibitory small RNAs (sRNAs) CsrB and CsrC, and a

specificity factor for the turnover of the sRNAs CsrD (Fig. 4-1A) (Vakulskas et al., 2015).

Generally, CsrA binds to conserved GGA motifs in the 5’-untranslated or early coding

region of mRNAs leading to changes in RNA structure (Patterson-Fortin et al., 2013),

RNA stability (Liu et al., 1995, Wang et al., 2005, Yakhnin et al., 2013), translation

initiation (Baker et al., 2002, Baker et al., 2007, Dubey et al., 2003), and/or transcription

elongation (Figueroa-Bossi et al., 2014). CsrA activity is controlled primarily by the

steady state levels of CsrB/C, which contain many high affinity CsrA binding sites that

sequester CsrA from interacting with its lower affinity mRNA regulatory targets (Liu et

al., 1997, Weilbacher et al., 2003, Vakulskas et al., 2015). Transcription of these sRNAs

is controlled by the BarA-UvrY TCS in response to the accumulation of end products of

metabolism, including acetate and formate (Chavez et al., 2010, Zere et al., 2015).

RNase E-dependent turnover of CsrB/C is regulated by CsrD although the exact

molecular mechanism remains unclear (Fig. 4-1A) (Suzuki et al., 2006, Vakulskas et al.,

2016, Leng et al., 2016). CsrD is a membrane-bound protein that has degenerate

Page 85: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

85

GGDEF and EAL domains. Although these domains are often involved in the synthesis

and degradation of c-di-GMP, CsrD neither produces nor degrades c-di-GMP (Suzuki et

al., 2006). However, CsrD is essential for the turnover of CsrB/C (Suzuki et al., 2006,

Vakulskas et al., 2016). Loss of CsrD strongly stabilizes these RNAs but only leads to a

modest increase in their steady states levels, likely due to regulatory feedback loops

that also control their transcription (Fig. 4-1A) (Suzuki et al., 2006).

When CsrD was initially described as a regulator of CsrB/C turnover, it was also

found to regulate CsrA-dependent gene expression and phenotypes such as biofilm

formation and glycogen synthesis in a CsrA and CsrB/C dependent manner (Suzuki et

al., 2006). This led to the development of a model, in which CsrD affects gene

expression through changes in CsrB/C levels, which then affect CsrA activity (Model 1,

Fig. 4-1A). According to this model CsrA acts as the most downstream regulator of gene

expression in the Csr system. It directly regulates gene expression posttranscriptionally

by affecting transcription elongation and transcript stability, but it can also mediate

indirect transcriptional and posttranscriptional effects through regulators it controls

directly. However, a recent transcriptomics study found that in addition to global effects

of CsrA on RNA stability and steady state RNA levels, CsrD had many effects on RNA

levels but not stability (Esquerré et al., 2016). These results led the authors to propose

an alternative model (Model 2, Fig. 4-1B). This model proposes that CsrA acts as a

posttranscriptional regulator for its direct targets, but it mediates most of its indirect

effects on RNA abundance indirectly though transcriptional effects controlled by CsrD

and perhaps some additional effects through other unknown factors.

Page 86: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

86

In this work we used RNA-seq to untangle the epistatic relationships among the

members of the Csr system (Epi-seq). To determine on a genome-wide scale whether

CsrD affects gene expression primarily through its effects on CsrB/C levels and

therefore CsrA activity, we compared the impact of CsrD on transcriptomic profile in the

presence and absence of CsrA or CsrB/C. If most of the effects of CsrD on gene

expression were lost in the absence of CsrA or CsrB/C, this would support Model 1 in

Fig. 4-1A, where CsrD primarily affects gene expression through CsrB/C and CsrA.

Likewise, CsrA effects on RNA abundance were assessed in strains with and without

CsrD. If most of CsrA effects on transcript levels were eliminated in the absence of

CsrD, this would support Model 2 in Fig. 4-1B, where CsrA primarily acts through CsrD

to mediate changes in transcription. We found that CsrA and CsrB mediate vast

transcriptional changes independently of CsrD, and the majority of CsrD-dependent

effects on gene expression require CsrA and CsrB/C. These results support Model 1,

where CsrD works primarily upstream of and through CsrB/C to regulate CsrA activity,

which is the major regulator of gene expression in the Csr system.

Materials and Methods

Media and Growth Conditions

Bacterial strains utilized in this study are listed in Table B-1. E. coli strains were

routinely grown in LB medium (1% tryptone, 1% NaCl, and 0.5% yeast extract) with

appropriate antibiotics when needed: ampicillin (100 μg mL−1), kanamycin (50 μg mL−1),

chloramphenicol (25 μg mL−1), and gentamicin (10 μg mL−1). For growth curve and

RNA-seq analysis, overnight culture grown in LB broth were inoculated into Kornberg

medium (1.1% K2HPO4, 0.85% KH2PO4, 0.6% yeast extract and 0.5% glucose) to an

Page 87: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

87

OD600 of 0.01, cultures were then grown at 37°C with shaking at 250 rpm and their

growth was monitored at OD600.

Construction of Strains and Plasmids

MG1655 pgaC880::cam, in which pagC gene was disrupted by transposon mini-

Tn10cam, was used as a parent wild-type strain (Wang et al., 2004). E. coli gene

deletions and disruptions were created by vir transduction procedure using E. coli donor

strains from previous studies (Suzuki et al., 2006, Wang et al., 2004, Vakulskas et al.,

2016) and the Keio library (Baba et al., 2006), as shown in Table B-1. The FRT-flanked

antibiotic resistance cassettes introduced into mutant strains were eliminated using an

FLP expression plasmid pCP20 when necessary (Datsenko & Wanner, 2000).

Plasmids and DNA oligonucleotides used in this study are listed in Tables B-2

and B-3. Plasmids p2VR112 (referred as pCsrA in this study) and pBRY4 (referred as

pCsrD in this study), express genes csrA and csrD, respectively, under the control of

their own promoters on plasmid pBR322 (Suzuki et al., 2006, Vakulskas et al., 2016).

To construct plasmid pCsrB for expression of CsrB, the csrB gene with 494 base pairs

(bp) upstream and 36 bp downstream was amplified from the genomic DNA and cloned

into plasmid pBR322. Control strains for pCsrA, pCsrD or pCsrB containing strains were

transformed with pBR322.

Glycogen Biosynthesis

Glycogen production was examined by staining colonies with iodine vapor (Liu et

al., 1997)

RNA Extraction and Purification

During transition to stationary phase of growth (OD600 of 2.0), 1 mL of cell culture

was collected and immediately mixed with 0.125 mL of stop solution (10% phenol / 90%

Page 88: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

88

ethanol) to stabilize RNA. Total RNA was extracted using hot phenol chloroform

followed by ethanol precipitation (Georgellis et al., 1992). Genomic DNA was removed

by treating 20 μg of nucleic acid with 4U of Turbo DNase (Ambion), and RNA was

purified from these reactions with the RNeasy kit (Qiagen). The integrity of the RNA was

verified using denaturing gel electrophoresis and RNA Bioanalyzer (Agilent) analysis.

Northern Blotting

Procedures for Northern blots were as described as in Chapter 2 ‘Materials and

Methods’.

RNA-seq Library Preparation

For each strain, independently grown biological triplicates were collected.

Ribosomal RNA was depleted from 5 μg of total RNA using the Ribo-Zero rRNA

Removal Kit for Gram-Negative Bacteria (Illumina). The concentrations of the rRNA-

depleted samples were determined with the Qubit RNA HS Assay Kit (ThermoFisher).

RNA-seq libraries were then generated with the Stranded RNA-Seq Library Preparation

Kit for Illumina (KAPA) and NEBNext Multiplex Oligos for Illumina adaptors (NEB)

according to the manufacturer’s instructions for 100 ng of starting material and a mean

insert size of 200-300 bases. Final libraries were purified with Pure Beads (KAPA).

Sequencing library size and integrity were verified with DNA Bioanalyzer analysis

(Agilent). Libraries were pooled and sequenced on 2 lanes of 50SE HiSeq 2500

(Illumina) by the Genomic Services Laboratory at the HudsonAlpha Institute for

Biotechnology.

RNA-seq Data Analysis

Raw reads were demultiplexed and analyzed with FastQC to ensure there were

no quality control issues. Sequencing reads were mapped to the E. coli rRNA

Page 89: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

89

sequences with Bowtie 2 (Langmead & Salzberg, 2012), and unmapped reads were

retained. rRNA depleted reads were then mapped to the E. coli genomic DNA sequence

(NC_000913.3) with Bowtie 2 (Langmead & Salzberg, 2012). Read counts per gene

were calculated with htseq-count (Anders et al., 2015). Read counts were filtered to

remove genes with an average of 10 reads per sample across all samples. Differential

expression was analyzed with limma voom (Liu et al., 2015), and genes with fold

changes greater than 2 and a p-value less 0.05 were considered significant.

qRT-PCR

Quantitative reverse transcriptase PCR (qRT-PCR) was conducted using iTaq

Universal SYBR Green One-Step Kit (Bio-Rad) and an iQ5 iCycler real time PCR

system (Bio-Rad) according to the manufacturer’s instructions. Reactions of 10 μL

contained 200 ng of RNA or DNA standard, 300 nM of each primer, iScript reverse

transcriptase, and 1x iTaq universal SYBR Green reaction mix. Reactions were

incubated for 10 min of RT at 50°C, 1 min of denaturation and RT inactivation at 95°C,

and then 45 cycles of 10 sec of denaturation at 95°C and 20 sec of annealing,

extension, and imaging at 60°C. Melt curve analysis was used to verify the specificity of

amplifications with the parameters: 95°C for 1 min, 55°C for 1 min, and increasing the

temperature 0.5°C/10 sec until reaching 95°C. RNA abundances were determined

relative to a standard curve of PCR products and normalized to 16s rRNA levels.

Results and Discussion

Construction and Characterization of Bacterial Strains

The csrA::gm gene disruption mutant (hereafter csrA mutant) used in this study is

not a deletion, but expresses a functionally impaired CsrA protein (Vakulskas et al.,

2016). Unlike a csrA deletion strain which exhibits a severe growth defects and rapidly

Page 90: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

90

accumulates suppressor mutations (Lawhon et al., 2003, Timmermans & Van Melderen,

2009, Yakhnin et al., 2013), this csrA mutant grows similarly to the wild-type in rich

media. For example, during the transition to stationary phase of growth (OD600 of 2.0) in

Kornberg (KB) media where we collected RNA for this study, the csrA mutant grows

only slightly slower than the csrA wild-type strain (Fig. 4-2A). As a csrAD double mutant

showed dramatically enhanced cell aggregation and biofilm formation (data not shown),

all of the strains were constructed in a pgaC disruption mutant background, which

cannot synthesize PGA or form PGA-dependent biofilm (Wang et al., 2004).

Northern blots and quantitative reverse transcriptase PCR (qRT-PCR) (Figs. 4-

2B-D) showed that while CsrD is essential for normal turnover of CsrB/C (Suzuki et al.,

2006), deletion of csrD only moderately increased CsrB levels and slightly decreased

CsrC levels and this phenotype can be complemented by ectopic expression of csrD

(Figs. 4-2B-D). Likely this is due to a feedback loop within Csr regulatory circuitry, in

which CsrB antagonizes CsrA activity to repress CsrB and CsrC transcription through

the BarA-UvrY TCS (Fig. 4-1A). CsrB is the principal sRNA antagonist of CsrA. Thus,

we expect that even with a minor decrease in CsrC level, the increased CsrB level

caused by the csrD deletion is sufficient to reduce CsrA activity. As expected (Camacho

et al., 2015), disruption of csrA in either the csrD mutant or wild-type strains significantly

reduced CsrB/C levels via BarA-UvrY TCS and this reduction can be restored by

ectopic expression of csrA (Figs. 4-2 B-D). Overall, these observations are consistent

with the known regulatory circuitry outlined in Model 1, where multiple feedback loops

existing to regulate the levels of CsrB/C.

Page 91: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

91

We also used iodine staining and qRT-PCR to determine glycogen abundance

and glgC RNA transcript levels in all the strains. As expected, the csrA mutant

accumulated much more glycogen and significantly increased glgC expression 8.7-fold

relative to the wild-type strain (Figs. 4-2E and F), whereas deletion of csrD resulted in a

slight increase in glycogen levels and increased glgC expression 1.4-fold relative to the

wild-type strain (Fig. 4-2E and F). Ectopic expression of csrA in the csrAD mutant and

csrBC in csrBCD significantly reduced and stimulated glycogen synthesis, respectively.

The analysis of glgC mRNA levels as determined by qRT-PCR were also consistent

with these observations of glycogen levels (Fig. 4-2F). All of these results confirmed that

the strains we constructed for RNA-seq analysis behave as expected.

CsrA Retains its Global Role in Regulating Transcript Levels in the Absence of CsrD

Differential expression analysis revealed 1,054 genes with greater than 2-fold

change in RNA levels and p-values less than 0.05 between the wild-type and csrA

mutant strains (Fig. 4-3A). CsrA repressed the expression of 828 genes and activated

226. In addition, 807 out of the 1054 genes were differentially expressed between the

csrAD double mutant strain and the csrD mutant strain (csrAD-csrD, Fig. 4-3B),

suggesting that CsrA retains its global influence on 80% of its target genes in the

absence of CsrD. To examine whether CsrD influences CsrA-mediated changes in gene

expression, we compared the log2 transformed fold change in RNA abundance caused

by the csrA mutation in the wild-type (csrA - WT) and csrD mutant strain (csrAD - csrD)

backgrounds. The high value of Spearman’s correlation coeficient between these data

sets demonstrated that the absence of csrD had little impact on the overall effect of

CsrA on gene expression (ρ=0.78, Fig. 4-3C). In addition, few genes varied in

Page 92: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

92

expression in the presence or absence of CsrD (Fig. 4-3C). Similarly, ectopic

expression of csrA in the csrAD double mutant resulted in vast changes in gene

expression (csrAD pCsrA - csrAD, Fig. 4-3B). The log2 transformed fold changes from

this comparison showed a strong negative correlation with those resulting from mutation

of csrA in either a wild-type (csrA - WT) or csrD mutant strain (csrAD - csrD)

background (ρ=0.82 and 0.87, respectively, Figs. 4-3D and E). Together these data

indicate that CsrA does not require CsrD to exert global effects on transcript levels. The

data do not support Model 2 in which CsrA effects on the transcriptome are primarily

mediated through CsrD (Fig. 4-1B).

To validate our RNA-seq results, we used qRT-PCR to analyze the effects of

CsrA on fabB and ftnA expression, which seem to be regulated by CsrA at the level of

their transcription (Esquerré et al., 2016) (Potts et al., unpublished). As Fig. 4-3F shown,

csrA mutation significantly increased expression of fabB and ftnA, and csrA

overexpression dramatically decreased their expression, both in the csrD wild-type and

csrD mutant backgrounds. These results also support a CsrD-independent role of CsrA

on gene expression.

The finding that CsrA regulates gene expression largely independently of CsrD

does not support Model 2 in which CsrA mediates transcriptional changes primarily

through CsrD (Fig. 4-1B). This raises a question as to how CsrA affects the

transcriptional landscape. We suspect that it does this directly through its

posttranscriptional regulation of RNA stability and transcription elongation, and also

indirectly by controlling other transcriptional and posttranscriptional regulators. Indeed,

our data showed that CsrA affected the expression of 79 transcription regulators,

Page 93: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

93

including two component systems (TCS), σ factors, transcription factors, and enzymes

involved in the metabolism of the secondary messenger c-di-GMP (Table B-4). This is

consistent with recent studies in our group, which demonstrated that CsrA interacts with

many mRNAs encoding transcriptional regulators in vivo as determined using UV-

crosslinking immunoprecipitation and sequencing (CLIP-seq) (Potts et al., unpublished).

In addition, CsrA affected the abundance of 8 sRNAs in addition to CsrB and CsrC

(Table B-5). sRNAs have recently been discovered to participate in transcriptional

regulation via various transcriptional regulators (Göpel & Görke, 2012, Mika & Hengge,

2014, Lee & Gottesman, 2016, Mandin & Guillier, 2013). It seems that integration of

posttranscriptional regulation into transcriptional regulatory networks is a common

theme used by bacterial cells. As result, posttranscriptional regulators (such as CsrA

and sRNAs) can have vast effects on gene expression and control a broad range of

genes and cellular functions. Overall, our data suggest that rather than acting through

CsrD to alter transcript levels, CsrA mediates changes in the transcription of many

genes indirectly through other regulators.

CsrD Effects on Gene Expression Require CsrA

The next question we addressed was whether CsrD affects gene expression in a

CsrA-dependent manner. We identified in total of 74 genes that were differentially

expressed between the wild-type and csrD mutant strains. These data suggest that

CsrD has a limited effect on transcriptome compared to CsrA under our experimental

condition (Figs. 4-4 and 4-3A). Importantly, deletion of csrD in the csrA mutant

background did not change expression of any genes with the exception of csrD and

csrB (csrAD – csrA, Fig. 4-4). This indicated that almost all of CsrD effects on gene

expression require CsrA. Furthermore, while ectopic expression of csrA caused vast

Page 94: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

94

transcriptional changes in the csrAD mutant background (csrAD pCsrA – csrAD, Fig. 4-

3B), ectopic expression of csrD in this strain did not result in any changes in gene

expression except of csrD and csrB (csrAD pCsrD – csrAD, Fig. 4-4). These results

confirm that CsrA functions downstream of CsrD to regulate gene expression.

CsrB/C Have Strong Effects on Gene Expression in the Absence of CsrD

CsrB/C affect gene expression indirectly through sequestration of CsrA (Liu et al., 1997,

Weilbacher et al., 2003, Vakulskas et al., 2015). According to Model 1 (Fig. 4-1A),

CsrB/C can impact the transcriptional landscape independently from CsrD. As shown in

Fig. 4-5, deletion of csrBC resulted in changes in expression of 40 genes in the wild-

type background (csrBC - WT) and 218 genes in the csrD mutant strain background

(csrBCD - csrD). This suggested that CsrB/C do not require CsrD in order to affect gene

expression. The more effective effects of the csrBC deletion in the csrD mutant than in

the wild-type background was likely due to higher level of CsrB in the csrD mutant,

which reduces CsrA activity more than in the wild type strain and results in greater

changes to the CsrA regulon upon CsrB/C deletion. Furthermore, overexpression of

CsrB in the csrBCD mutant strain resulted in changes in expression of 912 genes (Fig.

4-5), suggesting that CsrB/C retain their global roles in regulating gene expression in

the absence of CsrD. These data together with our observation that CsrA globally

affects gene expression independently of CsrD (Fig. 4-3) demonstrated that neither

CsrB/C nor CsrA acts primarily through CsrD to mediate changes in gene expression, in

stark contrast to the prediction of Model 2 (Fig. 4-1B).

CsrD Regulates the Majority of its Target Genes in a CsrB/C Dependent Manner

Model 1 predicts that CsrD indirectly modulates CsrA activity through its effect on

CsrB/C levels (Suzuki et al., 2006). To test if CsrD depends on CsrB/C to mediate

Page 95: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

95

changes in gene expression, we compared the impact of CsrD on the transcriptional

landscape in the presence and absence of CsrB/C. Differential expression analysis

revealed 60 genes that respond to csrD deletion in the wild-type background (csrD –

WT, Fig. 4-6A) but not in csrBC mutant background (csrBCD – csrBC, Fig. 4-6A),

suggesting that most of CsrD effects on gene expression require the presence of

CsrB/C. We also validated the dependence of CsrD-mediated ftnB expression on csrA

and CsrB/C using qRT-PCR and confirmed that CsrD lost its regulation of ftnB

expression in csrA or csrBC mutant background (Fig. 4-6B).

In the absence of CsrB/C, 26 genes still respond to CsrD (csrBCD – csrBC, Fig.

4-6A), suggesting that an alternative pathway may allow CsrD to mediates changes

through some unknown factor(s) rather than CsrB/C. The sRNA McaS has been

observed to serve as antagonists of CsrA and inhibits CsrA activity when overexpressed

(Jørgensen et al., 2013, Sterzenbach et al., 2013). Also, other sRNA were recently

discovered to directly bind CsrA in vivo in Samonella and E. coli (Holmqvist et al.,

2016)(Potts et al., unpublished). We predict that CsrD might mediate some changes in

gene expression through effects on other transcripts in a CsrA-dependent manner, it

might also function independently of CsrA. In addition, we observed 12 genes that were

regulated by CsrD only in csrBC mutant background (csrBCD – csrBC, Fig. 4-6A) but

not in wild-type background (csrD – WT, Fig. 4-6A). This implied that the CsrB/C

independent pathway might have opposite roles as the CsrB/C dependent pathway in

controlling expression of these 12 genes. Surprisingly, the expression level changes of

these 12 genes caused by csrD deletion in csrBC mutant background were not

complemented by ectopic expression of csrD (csrBCD pCsrD – csrBCD, Fig. 4-6A). The

Page 96: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

96

reason for this requires further investigation. Taken together, these results strongly

support Model 1 (Fig. 4-1A) in which most of CsrD effects on CsrA activity are mediated

through its effect on CsrB/C turnover. However, CsrD might also regulate other RNAs

that affect CsrA activity. We cannot rule out the possibility that CsrD regulates a limited

number of genes independently of CsrA, but this possibility needs to be further

explored.

Conclusion

Overall, we provide evidence on a genome-wild scale that CsrA protein and

CsrB/C sRNAs mediate vast transcriptional changes independently of CsrD. Moreover,

CsrD primarily affects gene expression through its effects on CsrB/C and CsrA. In

addition, some of our data suggest that CsrD may affect expression of genes through

additional factors, something that warrants further investigation. Altogether, the use of

Epi-seq to clarify the epistatic relationships among the components of the Csr system

supports the original model on CsrD function, in which CsrA acts as the most

downstream regulator of gene expression in the Csr system (Model 1, Fig. 4-1A) and

our data do not support Model 2, wherein CsrD acts as the primary regulator of the

indirect effects of CsrA (Fig. 4-1B).

Page 97: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

97

Figure 4-1. Two proposed model depicting the epistatic relationship between CsrA and CsrD in regulating mRNA abundance. A) CsrD affects gene expression through changes in CsrB/C levels, which then affect the activity of CsrA to directly or indirectly control mRNA expression (Suzuki et al., 2006). B) CsrA acts as a posttranscriptional regulator affecting RNA stability, but it mediates most of its indirect effects on mRNA abundance though transcriptional effects controlled by CsrD (Esquerré et al., 2016).

Page 98: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

98

Figure 4-2. Properties of bacterial strains used in this study. A) Growth curve of the wild-

type and mutant strains carrying plasmids pBR322, pCsrA, pCsrD or pCsrB growing in KB medium at 37 °C. Control strains for pCsrA, pCsrD or pCsrB containing strains were transformed with plasmid pBR322. B) Northern blots of CsrB and CsrC RNAs in 37°C cultures of wild-type and mutant strains carrying plasmids pBR322, pCsrA, pCsrD, or pCsrB in KB medium at the transition from exponential to stationary phase of growth. RNA levels in mutant strains relative to those in the wild-type (WT) strain were shown at the bottom. ND, not detected. C, D and F) qRT-PCR analysis of the transcript levels of csrB, csrC and glgC genes in mutant strains with plasmids pBR322, pCsrA, pCsrD or pCsrB relative to those in the WT strain at the transition from exponential to stationary phase of growth. Error bars represent the mean results ± S.D. from three biological replicates. Asterisks indicate level of significance (*p≤0.05, **p≤0.01, ***p≤0.001) by Student’s t-test. E) Glycogen production in WT and mutant strains with plasmids pBR322, pCsrA, pCsrD or pCsrB were determined by iodine staining.

CsrC

WT

csrA

csrA

pCsr

A

csrD

csrD

pCsr

D

csrA

D

csrA

D p

Csr

A

csrA

D p

Csr

D

csrBC

csrB

CD

csrB

CD p

Csr

B

csrB

CD p

Csr

D

0

2

4

6

Re

lati

ve

exp

res

sio

n le

velCsrB

0

2

4

6

8

10

Re

lati

ve

ex

pre

ss

ion

le

ve

l

A B

C D

CsrB

1.0 0.1 1.3 ND 0.3 1.8 0.1 ND 1.3 ND

CsrC

1.0 0.1 0.8 ND 0.1 2.3 0.1 ND ND ND

csrB

C

cs

rAD

csrA

D

pCsr

A

csrA

D

pCsr

D

csrB

CD

csrB

CD

pC

srB

csrB

CD

pc

srD

Ratio to WT

WT csrA csrD

Ratio to WT

0 2 4 6 8 10 12 14 16 18 20 22 240.01

0.1

1

10

Time (Hour)

OD

600

WT

csrA

csrD

csrBC

csrAD

csrAD pCsrA

csrAD pCsrD

csrBCD

csrBCD pCsrB

csrBCD pCsrD

WT

csrA

csrA

pCsr

A

csrD

csrD

pCsr

D

csrA

D

csrA

D p

Csr

A

csrA

D p

Csr

D

csrB

C

csrB

CD

csrB

CD p

Csr

B

csrB

CD p

Csr

D

* *

* *

Page 99: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

99

Figure 4-2. Continued.

Page 100: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

100

Figure 4-3. CsrA retains its global role in regulating mRNA levels in the absence of

CsrD. A) Volcano Plot of the log2 fold change of RNA levels between the csrA

mutant and its isogenic wild-type strain. P-value ≤ 0.05 and a log2 fold

change higher than 1 or lower than − 1 were used as cutoff. The numbers of downregulated and upregulated genes (black dots) were shown on the top. B) Venn diagram depicting overlap of differentially expressed genes in csrA versus WT, csrAD versus csrD, and csrAD pCsrA versus csrAD. C, D and E) The log2 transformed fold change in RNA abundance caused by loss or overexpression of CsrA in the WT and/or csrD mutant backgrounds. Blue and red dots represent the genes that are only differentially expressed in one strain background. Black and grey dots represent the genes that are differentially expressed in both of the backgrounds, or neither of the background, respectively. The Spearman's correlation coefficients (ρ) were shown. F) qRT-PCR analysis of the transcript levels of acnA and ftnB genes in mutant strains csrA, csrD and csrAD carrying plasmids pBR322, pCsrA or pCsrD relative to those in the WT strain at the transition from exponential to stationary phase of growth. Error bars represent the mean results ± S.D. from three biological replicates. Asterisks indicate level of significance (*p≤0.05, **p≤0.01, ***p≤0.001) and n.s. indicates not significant by Student’s t-test.

Page 101: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

101

Figure 4-3. Continued.

Page 102: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

102

Figure 4-4. Venn diagram depicting the overlap of differentially expressed genes

induced by csrD deletion/overexpression in csrA WT and csrA mutant backgrounds. Specifically, differentially expressed genes in csrD versus WT, csrAD versus csrA, csrAD pCsrD versus csrAD were compared.

Figure 4-5. Venn diagram depicting the overlap of differentially expressed genes

induced by csrB/C deletion or csrB overexpression in csrD WT and csrD mutant backgrounds. Specifically, differentially expressed genes in csrBC versus WT, csrBCD versus csrD, csrBCD pCsrB versus csrBCD were compared.

Page 103: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

103

Figure 4-6. CsrD regulate the majority of its target genes in a CsrB/C dependent

manner. A) Venn diagram depicting the overlap of differentially expressed genes in csrD versus WT, csrBCD versus csrBC, and csrBCD pCsrD versus csrBCD. B) qRT-PCR analysis of the transcript levels of ftnB gene in mutant strains carrying plasmids pBR322, pCsrA, pCsrD or pCsrB relative to those in the wild-type strain at the transition from exponential to stationary phase of growth. Error bars represent the mean results ± S.D. from three biological replicates. Asterisks indicate level of significance (*p≤0.05, **p≤0.01, ***p≤0.001) and n.s. indicates not significant by Student’s t-test.

Page 104: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

104

CHAPTER 5 GENERAL DISCUSSION AND FUTURE PERSPECTIVES

Csr/Rsm sRNAs (for example E. coli CsrB/C) are non-coding sRNAs that

indirectly control gene expression by binding to and sequestering the activity of the

posttranscriptional regulatory proteins, CsrA/RsmA (Romeo et al., 2013, Vakulskas et

al., 2015). Synthesis and turnover of CsrB/C in E. coli are both controlled by

environmental signals and play important roles in governing CsrA activity and bacterial

lifestyle (Chavez et al., 2010, Huang et al., 2008, Edwards et al., 2011, Camacho et al.,

2015, Pannuri et al., 2016, Leng et al., 2016). The turnover of CsrB/C is initiated by

RNase E cleavage and subsequently faciliated by PNPase (Suzuki et al., 2006,

Vakulskas et al., 2016). This RNase E-dependent turnover of CsrB/C also requires

CsrD protein. The absence of CsrD significantly stabilized CsrB/C (Suzuki et al., 2006).

In this study, the physiological role of CsrD, the exact molecular mechanism how CsrD

facilitates CsrB/C turnover and whether CsrD has broader regulatory roles in addition to

regulating CsrB/C turnover were explored.

First, we revealed a physiological role of CsrD in coupling CsrB/C decay to

availability of preferred carbon sources. The CsrD effect is achieved by a direct

interaction of EIIAGlc of the glucose-specific PTS system to the EAL domain of CsrD. We

demonstrated that EIIAGlc regulates CsrB/C turnover in a phosphorylation dependent

manner and only the unphosphorylated form of EIIAGlc bound to CsrD and was capable

of activating CsrB/C turnover. On the other hand, the phosphorylated form of EIIAGlc

indirectly and modestly represses CsrB turnover via cAMP-Crp, reinforcing the positive

effect of unphosphorylated EIIAGlc on CsrB decay. This regulatory pathway couples

CsrB/C sRNA decay to the availability of a preferred carbon source, glucose. While

Page 105: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

105

CsrB/C in strains lacking CsrD protein are extremely stable, the presence of CsrD in E.

coli ensures the high decay rates of CsrB/C in the presence of preferred carbon source

and poises the Csr system for rapid response to environmental stimuli.

This study also uncovered an important new way in which EIIAGlc shapes global

regulatory circuitry in response to nutritional status. Previous studies have identified

regulatory roles of EIIAGlc in carbon metabolism (Deutscher et al., 2014, Deutscher et

al., 2006), chemotaxis (Neumann et al., 2012), respiration/fermentation (Koo et al.,

2004), biofilm formation (Pickering et al., 2012) and virulence (Kim et al., 2010, Mazé et

al., 2014). Here, we showed that EIIAGlc facilitates the turnover of sRNAs CsrB/C in the

presence of glucose, resulting in decreased CsrB/C levels and therefore increased CsrA

activity to stimulate expression of genes for rapid growth.

In addition, this study uncovered a novel function of the EAL domain. A few

catalytically inactive EAL domains have been discovered to perform regulatory roles via

protein-protein interactions (Römling et al., 2013, Li et al., 2012, Tschowri et al., 2009,

Guzzo et al., 2009), but they all act as sensors to transmit information to another

protein. Here, the EAL domain of CsrD was revealed to act as a receiver and detect

signaling information from a sensory protein EIIAGlc. Also, it would be very intriguing to

figure out the molecular mechanism of the interaction between EAL domain and EIIAGlc

by crystallography.

More importantly, these findings revealed a new physiological influence on the

workings of the Csr system. Here we discovered that the presence of glucose

stimulates CsrB/C turnover and reduces CsrB/C levels. Previous studies have shown

that carboxylic acid-containing end products of carbon metabolism, such as acetate and

Page 106: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

106

formate, stimulate CsrB transcription via the BarA-UvrY TCS (Chavez et al., 2010).

Thus, carbon availability is capable to mediate reinforcing effects on the levels of these

sRNAs through their synthesis pathway and the newly discovered turnover pathway.

This allows bacterial cells to rapidly alter the concentration of CsrA and properly

mediate gene expression in order to switch between active growth and stress resistant

growth upon changes in carbon availability. Of particular interest, even though CsrB/C

turnover rates are severely inhibited by the lack of EIIAGlc or CsrD, their levels are only

modestly changed due to feedback loops. We wonder that whether the turnover

pathway has a bigger impact in earlier stage upon changes in carbon availability and

the impact lessens over time when the transcriptional response takes over. It would be

of great interest to characterize the signaling dynamics of each pathway under different

carbon conditions, specifically, the real-time response patterns of CsrB/C turnover and

the transcriptional response could be investigated.

Besides, another question worthy of further investigation is whether other

environmental signals in addition to carbon availability affect CsrB/C turnover. The Csr

system captures environmental stimuli mainly through fluctuations of the steady state

levels of CsrB/C and then converges this into global regulation through CsrA. Multiple

regulatory factors that mediate stress responses, including stringent response

components (p)ppGpp/DksA (Edwards et al., 2011), the global stress σ factors RpoS

(Yakhnin et al., 2011), RNA chaperon Hfq (Suzuki et al., 2006), RNA helicases

Dead/SrmB (Vakulskas et al., 2014), and Crp (Pannuri et al., 2016) have been

discovered to control the transcription of CsrB/C. It would be interesting to know

Page 107: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

107

whether these factors also regulate the Csr system through affecting the turnover of

CsrB/C.

Our recent work demonstrated that RNase E-mediated turnover of CsrB is

antagonistically controlled by CsrA and CsrD in E. coli (Leng et al., 2016). The binding

of CsrA to CsrB blocks the RNase E-mediated cleavage of CsrB both in vitro and in

vivo, while CsrD facilitates CsrB turnover by overcoming the CsrA-mediated protection

in vivo. In this study, we showed that although CsrD binds to CsrB in vitro, it did not

seem to bind CsrB in vivo and it failed to facilitate the cleavage of CsrA-protected CsrB

by RNase E in vitro. These data suggest that rather than functioning as an RNA binding

protein, CsrD might act indirectly and require other unknown factor(s) for its regulatory

role on CsrB/C turnover.

Proteins that are associated with CsrB in vivo were analyzed in this study by

immunoprecipitation and mass-spectrometry. In addition to CsrA, components of the

RNA degradosome, ribosomal proteins and PAP I were identified. While enolase was

strongly enriched along with purified CsrB, it does not influence CsrB/C turnover.

Whether other proteins, such as ribosomal proteins, are functionally relevant to CsrB

decay or have other functions remains to be further explored. Identifying the missing

factors involved in CsrB turnover will not only help to elucidate how CsrD works but also

provides new insights into the complexity of sRNAs degradation pathway. Genetic

screening can be conduced in future work to search for these factors.

In this study, we clarified the epistatic relationships of CsrA, CsrB/C and CsrD in

Csr system using Epi-seq. According to our previous study (Suzuki et al., 2006), CsrD

was proposed to affect gene expression through its effects on CsrB/C turnover and then

Page 108: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

108

CsrA activity (Model 1, Fig. 4-1A). However, a recent transcriptomics study has

proposed an alternative model that CsrD works downstream of CsrA for regulating gene

expression and it is responsible for most of the indirect effects of CsrA on RNA

abundance (Model 2, Fig. 4-1B) (Esquerré et al., 2016). Our data illustrated on a

genome-wide scale that CsrD mediates changes in gene expression primarily through

its effects on CsrB/C and CsrA. Moreover, CsrA does not require CsrD to exert its

global effects on transcript levels, and its effects on indirect targets might be mediated

via other transcriptional and posttranscriptional regulators. In addition, our data

suggested that CsrD affects expression of some genes through an alternative pathway

independent of CsrB/C, suggestive of other regulatory role(s) of CsrD in addition to

facilitating CsrB/C turnover. Further investigation will be required to identify these

additional factors and pathways.

Page 109: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

109

APPENDIX A SUPPLEMENTARY FIGURES

Figure A-1. EIIAGlc stimulates CsrB decay in the MG1655 csrDFLAG strain. Northern blots

depicting the effect of EIIAGlc on CsrB decay in strains MG1655 csrDFLAG and MG1655 Δcrr csrDFLAG. Half-lives were determined as in Figure 2-1.

Figure A-2. Decay of CsrB/C in a strain expressing EIIAGlc H91D is similar to that of EIIAGlc H91A. Decay rates of CsrB/C were determined in the Δcrr strain containing plasmid pCRRH91D, as described for the pCRRH91A-containing strain in Figure 2-1A and C.

Figure A-3. CRP has minimal or no effect on CsrD protein levels. Western blots depicting effects of crp deletion on CsrD protein levels. RpoB was used as loading control. CsrD protein levels in Δcrp relative to those in the wild-type strain (WT) were given. Standard derivations from triplicate experiments are indicated.

0.1 0.5 1.0 2.0

WT Δcrp WT Δcrp WT Δcrp WT Δcrp

OD600

1.5 ± 0.3 1.3 ± 0.1 1.0 ± 0.2 1.0 ± 0.1 Δcrp/WT

CsrDFLAG

RpoB

Page 110: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

110

APPENDIX B SUPPLEMENTARY TABLES

Table B-1. Bacterial Strains

Strain Description Reference

MG1655 Prototrophic E. coli K12 Michael Cashel MG1655 Δcrr MG1655 with unmarked crr deletion This study (Chapter 2) MG1655 ΔcsrD MG1655 with unmarked csrD

deletion This study (Chapter 2)

MG1655 Δcrp MG1655 with marked crp disruption-Camr

This study (Chapter 2)

MG1655 ΔcyaA MG1655 with marked cyaA deletion-Kanr

This study (Chapter 2)

MG1655 Δcrr ΔcsrD

MG1655 with unmarked crr deletion and marked csrD deletion-Kanr

This study (Chapter 2)

MG1655 Δcrr Δcrp

MG1655 with unmarked crr deletion and marked crp disruption-Camr

This study (Chapter 2)

MG1655 Δcrr ΔcyaA

MG1655 with unmarked crr deletion and marked cyaA deletion-Kanr

This study (Chapter 2)

MG1655 csrDFLAG

MG1655 with in-frame, CTD 3X-FLAG tag at native csrD locus

This study (Chapter 2)

MG1655Δcrr csrDFLAG

csrDFLAG allele introduced by transduction using P1vir - Kanr

This study (Chapter 2)

MG1655 Δcrp csrDFLAG

csrDFLAG allele introduced by transduction using P1vir - Kanr

This study (Chapter 2)

MG1655 crrFLAG MG1655 with in-frame, CTD 3X-FLAG tag at native crr locus

This study (Chapter 2)

MG1655 H91A MG1655 with a His91Ala exchange (CAC to GCC) at native crr locus

This study (Chapter 2)

MG1655 H91A crrFLAG

crrFLAG allele introduced by transduction using P1vir

This study (Chapter 2)

BL21(DE3) Host for expression (Studier & Moffatt, 1986) DE5α Host for plasmid amplification (Woodcock et al., 1989) PW1096 Vibrio cholerae C6706str2 (Thelin & Taylor, 1996) PW1197 PW1096 with in frame mshH deletion This study (Chapter 2) PW1198 PW1096 with in frame crr deletion This study (Chapter 2) PW1207 PW1096 with in fram mshH and crr

deletions This study (Chapter 2)

MG1655 rne-1 ΔcsrB pCsrB

MG1655 rne-1 with marked csrB deletion-Gmr, harboring plasmid pCsrB

This study (Chapter 3)

MG1655 rne-1 ΔcsrB p2SLStrepto-CsrB

MG1655 rne-1 with marked csrB deletion-Gmr, harboring plasmid p2SLStrepto-CsrB

This study (Chapter 3)

Page 111: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

111

Table B-1. Continued.

Strain Description Reference

MG1655 rne-1 ΔcsrB ΔcsrD pCsrB

MG1655 rne-1 ΔcsrB pCsrB with marked csrD deletion-Kanr

This study (Chapter 3)

MG1655 rne-1 ΔcsrB ΔcsrD p2SLStrepto-CsrB

MG1655 rne-1 ΔcsrB p2SLStrepto-CsrB with marked csrD deletion-Kanr

This study (Chapter 3)

MG1655 rne-1 ΔcsrB csrDFLAG pCsrB

MG1655 rne-1 ΔcsrB pCsrB with in-frame CTD 3X-FLAG tag at native csrD locus

This study (Chapter 3)

MG1655 rne-1 ΔcsrB csrDFLAG p2SLStrepto-CsrB

MG1655 rne-1 ΔcsrB p2SLStrepto-CsrB with in-frame CTD 3X-FLAG tag at native csrD locus

This study (Chapter 3)

MG1655 ΔcsrD pBYH4 MG1655 with marked csrD deletion-Kanr, harboring plasmid pBYH4

This study (Chapter 3)

MG1655 ΔcsrD p3FLAG-CsrD

MG1655 with marked csrD deletion-Kanr, harboring plasmid p3FLAG-CsrD

This study (Chapter 3)

XWC880 MG1655 with marked pgaC disruption-Camr

(Wang et al., 2004)

MG1655 csrA::gm MG1655 with csrA disrupted after amino acid position 50 - Gmr

(Vakulskas et al., 2016)

KDMG MG1655 with marked csrD deletion-Kanr

(Suzuki et al., 2006)

BW25113 csrB::kan

BW25113 with marked csrB deletion-Kanr

Baba, T 2006 (Baba et al., 2006)

BW25113 csrC::kan BW25113 with marked csrC deletion-Kanr

Baba, T 2006 (Baba et al., 2006)

Wild-type (WT)

MG1655 with marked pgaC disruption-Camr, carrying plasmid pBR322

This study (Chapter 4)

csrA

MG1655 with marked pgaC disruption-Camr and CsrA disruption-Camr, carrying plasmid pBR322

This study (Chapter 4)

csrD

MG1655 with marked pgaC disruption-Camr and csrD deletion-Kanr, carrying plasmid pBR322

This study (Chapter 4)

csrBC

MG1655 with marked pgaC disruption-Camr and unmarked csrB and csrC deletions, carrying plasmid pBR322

This study (Chapter 4)

Page 112: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

112

Table B-1. Continued.

Strain Description Reference

csrAD

MG1655 with marked pgaC disruption-Camr, marked csrA disrutpion-Gmr and marked csrD deletion-Kanr, carrying plasmid pBR322

This study (Chapter 4)

csrA pCsrA

MG1655 with marked pgaC disruption-Camr and marked csrA disruption-Gmr, carrying plasmid pCsrA

This study (Chapter 4)

csrD pCsrD

MG1655 with marked pgaC disruption-Camr and marked csrD deletion-Kanr, carrying plasmid pCsrD

This study (Chapter 4)

csrAD pCsrD

MG1655 with marked pgaC disruption-Camr, marked csrA disrutpion-Gmr and marked csrD deletion-Kanr, carrying plasmid pCsrD

This study (Chapter 4)

csrAD pCsrA

MG1655 with marked pgaC disruption-Camr, marked csrA disrutpion-Gmr, and marked csrD deletion-Kanr, carrying plasmid pCsrA

This study (Chapter 4)

csrBCD

MG1655 with marked pgaC disruption-Camr, marked csrD deletion-Kanr and unmarked csrB and csrC deletions, carrying plasmid pBR322

This study (Chapter 4)

csrBCD pCsrB

MG1655 with marked pgaC disruption-Camr, marked csrD deletion-Kanr and unmarked csrB and csrC deletions, carrying plasmid pCsrB

This study (Chapter 4)

csrBCD pCsrD

MG1655 with marked pgaC disruption-Camr, marked csrD deletion-Kanr and unmarked csrB and csrC deletions, carrying plasmid pCsrD

This study (Chapter 4)

Page 113: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

113

Table B-2. Plasmids used in this study.

Name Description Relevant Primers

Reference

pBR322 Cloning vector - Ampr Tetr N/A (Bolivar et al., 1977)

pCRR

pBR322 derivative carrying crr gene between EcoRI and HindIII sites- Ampr

P1/P2 This study (Chapter 2)

pCRRH91A pCRR derivative carrying crr with a His91Ala exchange (CAC to GCC)

P1/P17/P2

This study (Chapter 2)

pCRRH91D pCRR derivative carrying crr with a His91Asp exchange (CAC to GAC)

P1/P19/P2

This study (Chapter 2)

pBYH4

pBR322 derivative carrying csrD gene in EcoRI site- Ampr

N/A

(Suzuki et al., 2006)

pET24CRR pET24a derivative carrying crr gene between NdeI and XhoI sites

P3 /P4 This study (Chapter 2)

pDTM

pMAL-c5x derivative carrying DNA encoding 156-646 aa of CsrD between NcoI and EcoRI sites

P5/P10

This study (Chapter 2)

pDEAL

pMAL-c5x derivative carrying DNA encoding 156-385 aa of CsrD between NcoI and EcoRI sites

P6/P7

This study (Chapter 2)

pDGGDEF pMAL-c5x derivative carrying DNA encoding 156-223 and 393-646 aa of CsrD between NcoI and EcoRI sites

P6/P8 and P9/P10

This study (Chapter 2)

pDHAMP pMAL-c5x derivative carrying DNA encoding 192-646 aa of CsrD between NcoI and EcoRI sites

P11/P10

This study (Chapter 2)

pDcoil pMAL-c5x derivative carrying DNA encoding 156-199 and 220-646 aa of CsrD between NcoI and EcoRI sites

P6/P12 and P13/P10

This study (Chapter 2)

pEAL pMAL-c5x derivative carrying DNA encoding 393-646 aa of CsrD between NcoI and EcoRI sites

P14/P10

This study (Chapter 2)

pKOV Vector for homologous recombination

N/A (Link et al., 1997)

pKOVH91A pKOV derivative carrying crr with a His91Ala exchange (CAC to GCC) between NotI and BamHI sites

P15/P17and P18/ P16

This study (Chapter 2)

pCsrB pBR322 derivative carrying csrB gene with 494bp upstream and 36bp downstream between BamHI and HindIII sites- Ampr

csrB-494upBamHI/csrB-36downHindIII

This study (Chapters 3 and 4)

Page 114: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

114

Table B-2. Continued.

Name Description Relevant Primers

Reference

p2SLStrepto-CsrB

pBR322 derivative expressing Strepto-CsrB- Ampr

p2SLStrepto-CsrB-F/R

This study (Chapter 3)

p3FLAG-CsrD pBR322 derivative expressing 3FLAG tagged CsrD- Ampr

p3FLAG-CsrD-F/R

This study (Chapter 3)

p2VR112 csrA gene cloned into the EcoRI-BamHI sites of pBR322 - Ampr

N/A (Vakulskas et al., 2016)

Page 115: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

115

Table B-3. Primers used in this study.

Primer Sequence (5’-3’) Function

P1 CAGTACCAGGAATTCTTTACACTTTATGCTTCCGGCTCGTATATTGTGTGGAAGAAATAATTTTGTTTAACTTTAAG

pCRR and pH91D construction

P2 CAGGACCATAAGCTTTTACTTCTTGATGCGGATAACCGGGGT

pCRR construction

P3 ACATGATCTCATATGGGTTTGTTCGATAAACTGAAATCTC

pET24CRR construction

P4 ACATGATCTCTCGAGCTTCTTGATGCGGATAACCGGGGTT

pET24CRR construction

P5 CCGATTCCATGGGCCGCTGGTTACAACGGCAACTTGCCG

pDTM construction

P6 CATGCCATGGGCCGCTGGTTACAACGGCAACTTGC

pDEAL, pDGGDEF and pDcoil construction

P7 ACCGGAATTCTTAGTAAATAGCCCAGCTATTGCCGC

pDEAL construction

P8 AACATTACCGCGTCCATAAGAGCGGATCAG pDGGDEF construction

P9 CTGATCCGCTCTTATGGACGCGGTAATGTT pDGGDEF construction

P10 ACCGGAATTCTTAAACCGAGTATCTTTGTGAATAT

pDTM, pDGGDEF, pDHAMP, pDcoil and pEAL construction

P11 CATGCCATGGGCCCGCCCAGAACCAGCAGTGC pDHAMP construction

P12 GGCGGCATAAGAGCGGATCAGCGCACTGCTGGTTCT

pDcoil construction

P13 AGAACCAGCAGTGCGCTGATCCGCTCTTATGCCGCC

pDcoil construction

P14 CATGCCATGGGCGGACGCGGTAATGTTCGCTGGCGTA

pEAL construction

P15 ATAAGAATGCGGCCGCAAAGATCTGCCAGCTATTACGCTGG

pKOVH91A construction

P16 CGCGGATCCTGATAGCCGATTTGACTGCCAGAAT pKOVH91A construction

P17 GGTGTCGATACCGAAGGCGACGAACAGTTCAAC pCRRH91A and pKOVH91A construction

P18 GTTGAACTGTTCGTCGCCTTCGGTATCGACACC pKOVH91A construction

P19 GGTGTCGATACCGAAGTCGACGAACAGTTCAAC pCRRH91D construction

Page 116: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

116

Table B-3. Continued.

Primer Sequence (5’-3’) Function

S1 CGTAACCGTGGGTGAAACCCCGGTTATCCGCATCAAGAAGGACTACAAAGACCATGACGG

MG1655 crrFLAG

construction S2 AAATGGCGCCGATGGGCGCCATTTTTCACTGCG

GCAAGAACATATGAATATCCTCCTTAG MG1655 crrFLAG

construction S3 TGATACTAACGTGAAAAAATATTCACAAAGATAC

TCGGTTGACTACAAAGACCATGACGG MG1655 csrDFLAG construction

S4 AGCGCGCATTATTCTACGTGAAAACGGATTAAACGGCAGGCATATGAATATCCTCCTTAG

MG1655 CsrDFLAG construction

R1 GTAATACGACTCACTATAGTCGACAGGGAGTCAGACAAC

RNA CsrB synthesis

R2 AAAAAAAGGGAGCACTGTATTCACAGCGCTCCCGGTTCGTTTCGCAG

RNA CsrB synthesis

R3 GTAATACGACTCACTATAGGATAGAGCGAGGACGCTAACAGGAAC

RNA CsrC synthesis

R4 AAGAAAAAAGGCGACAGATTACTCTGTCGCCTTTTTTCCTGACTC

RNA CsrC synthesis

R5 GTAATACGACTCACTATAGGCCTTTGAATTGTCCATATAGAACAC

RNA rpsT synthesis

R6 AAAAAAACCCGCTTGCGCGGGCTTTTTCACAAAGCTTCAGC

RNA rpsT synthesis

R7 TAATACGACTCACTATAGGGTAGATGCTCATTCCATCTC

RNA GlmZ synthesis

R8 AAAAAAACGCCTGCTCTTATTACGGAGCAGGCGTTAAAAC

RNA GlmZ synthesis

RP1 TAATACGACTCACTATAGGGTAAAAGGTGCTCCCTGCATCTAATC

V. cholerae CsrB probe synthesis

RP2 TGGTGATCTTCAGGAAGAAGAATCG V. cholerae CsrB probe synthesis

RP3 TAATACGACTCACTATAGGGATCCTTTCAGCGAACTCCGAGCATC

V. cholerae CsrC probe synthesis

RP4 CAGGATGAGAAGTGGTGAGGATGAC V. cholerae CsrC probe synthesis

RP5 TAATACGACTCACTATAGGGCAATCCCGCTACTAATAGGTGCTCC

V. cholerae CsrD probe synthesis

RP6 CAAGGATTGGTCATCTTCAGGACGA V. cholerae CsrD probe synthesis

RP7 GTAATACGACTCACTATAGGTTCGTTTCGCAGCATTCCAG

E. coli CsrB probe synthesis

RP8 GCGTTAAAGGACACCTCCAGG E. coli CsrB probe synthesis

RP9 GTAATACGACTCACTATAGGTCTTACAATCCTTGCAGGC

E. coli CsrC probe synthesis

RP10 GAGGACGCTAACAGGAACAATG E. coli CsrC probe synthesis

Page 117: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

117

Table B-3. Continued.

Primer Sequence (5’-3’) Function

3FLAG-CsrD-F

TGATATCGACTACAAAGATGACGACGATAAATAGTAACCTGCCGTTTAATCCGTTTTCA

p3FLAG-CsrD construction

3FLAG-CsrD-R

TGATCTTTATAATCACCGTCATGGTCTTTGTAGTCAACCGAGTATCTTTGTGAATATTTT

p3FLAG-CsrD construction

2SLStrepto-CsrB-F

GCAAGGGCACCACGGTCGGATCCCACTTCTGCAGGACACACCAGGAT

p2SLStrepto-CsrB construction

2SLStrepto-CsrB-R

GGGCAGAAGTCCAAATGCGATCCCACTTCGTTGTCTGACTCCCTGTCG

p2SLStrepto-CsrB construction

csrB-494upBamHI

CGCGGATCCATATGCACGCGCAGTTTGGCGATAT pCsrB construction

csrB-36downHindIII

CCCAAGCTTACCTCAATAAGAAAAACTGCCGCGA pCsrB construction

70upCsrA

TGGCGTTATATGATGGATAATGCCG csrA::gm confirmation

100downCsrA

GAGACTTAAGTTGAATGAACGGGAG csrA::gm confirmation

Page 118: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

118

Table B-4. Transcription regulators regulated by CsrA

Gene Locus Gene

Regulator Type Directon Description

b2127 mlrA Transcription factor Repressed Transcriptional regulator of csgD

b3555 yiaG Transcription factor Repressed

Predicted transcriptional regulator, function unknown

b1306 pspC Transcription factor Repressed

Positive regulatory gene, cooperatively with PspB; facilitates binding of PspA to PspB; membrane protein; dimer

b0313 betI Transcription factor Repressed

Transcriptional repressor for the betIBA-betT divergent operon, choline-inducible

b2664 csiR Transcription factor Repressed Repressor of csiD promoter

b1399 paaX Transcription factor Repressed

Repressor for the paa operon, phenylacetyl-CoA induced

b4396 rob Transcription factor Repressed

Right oriC-binding transcriptional activator, AraC family

b1299 puuR Transcription factor Repressed

Repressor for the divergent puu operons, putrescine inducible; putrescine utilization pathway

b4135 yjdC Transcription factor Repressed

Putative transcriptional repressor, function unknown

b4313 fimE Transcription factor Repressed

Site-specific recombinase, fimA promoter inversion; biased towards the "ON to OFF" fimbriae phase switching direction

b2706 srlM Transcription factor Repressed

srl operon transcriptional activator, sorbitol-responsive

b2537 hcaR Transcription factor Repressed

Transcriptional activator for the hca operon; inducd by 3-phenylpropionate and cinnamic acid; autoregulatory

b3995 rsd Transcription factor Repressed

Regulates RNA polymerase holoenzyme formation; interacts with free σ 70 and core RNA polymerase; stationary phase protein; anti-σ

b1217 chaB Transcription factor Repressed

Accessory and regulatory protein for chaA

b3023 ygiV Transcription factor Repressed

Represses mcbR, involved in biofilm regulation

b0487 cueR Transcription factor Repressed

Activator of copper-responsive regulon genes cueO and copA; MerR homolog

b2398 yfeC Transcription factor Repressed

Predicted DNA-binding protein, DUF1323 family

Page 119: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

119

Table B-4. Continued.

Gene Locus Gene

Regulator Type Directon Description

b0435 bolA Transcription factor Repressed

Stationary-phase morphogene, transcriptional repressor; predicted reductase; regulates mreB, dacA, dacC, and ampC transcription;

b4340 yjiR Transcription factor Repressed

Predicted HTH transcriptional regulator with aminotransferase domain, function unknown; MocR family

b2980 glcC Transcription factor Repressed

Transcriptional reperssor for glc operon, glycolate-binding

b0330 prpR Transcription factor Repressed

Transcriptional regulator of prp operon; propionate catabolism via 2-methylcitrate cycle, characterized primarily in Salmonella

b1422 ydcI Transcription factor Repressed Putative transcriptional regulator

b1014 putA Transcription factor Repressed

Proline dehydrogenase and repressor for the putAP divergon

b1384 feaR Transcription factor Repressed

Transcriptional activator for tynA and feaB, AraC family

b2427 murR Transcription factor Repressed

Repressor for murPQ, MurNAc 6-P inducible

b3604 lldR Transcription factor Repressed

Dual role activator/repressor for lldPRD operon

b1622 malY Transcription factor Repressed

Antagonist of MalT transcriptional activator of maltose regulon, binds MalT in absence of maltotriose; cysteine desulfhydrase

b0603 ybdO Transcription factor Repressed

Required for swarming phenotype, function unknown; probable LysR-family transcriptional regulator

b0020 nhaR Transcription factor Repressed

Positive regulator of nhaA, Na(+)-dependent

b0145 dksA Transcription factor Repressed

RNAP-binding protein modulating ppGpp and iNTP regulation; reduces open complex half-life on rRNA promoters; removes transcriptional roadblocks to replication

b3680 yidL Transcription factor Repressed

Predicted transcriptional regulator, AraC family, function unknown

b0064 araC Transcription factor Repressed

Transcriptional activator for the ara regulon

Page 120: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

120

Table B-4. Continued.

Gene Locus Gene

Regulator Type Directon Description

b2399 yfeD Transcription factor Repressed

Predicted DNA-binding protein, DUF1323 family

b4365 yjjQ Transcription factor Repressed

Putative transcriptional regulator, function unknown; H-NS-repressed, dimeric

b3021 mqsA Transcription factor Repressed

Antitoxin for MqsR toxin; transcriptional repressor

b2846 yqeH Transcription factor Repressed

Predicted LuxR family transcriptional regulator; part of T3SS PAI ETT2 remnant

b1450 mcbR Transcription factor Repressed

MqsR-controlled colanic acid and biofilm regulator; represses mcbA

b0346 mhpR Transcription factor Repressed

Transcriptional activator, mhp operon; utilizes MHP

b1499 ydeO Transcription factor Repressed

UV-inducible global regulator, EvgA-, GadE-dependent; transcriptional activator for mdtEF; AraC family

b0076 leuO Transcription factor Repressed

Pleiotropic transcriptional regulator; regulates dsrA; relieves bgl silencing, multi-copy represses cadC

b2491 hyfR Transcription factor Repressed

Formate-sensing regulator for hyf operon

b3022 mqsR Transcription factor Repressed

GCU-specific mRNA interferase, toxin-antitoxin pair MqsRA; motility, quorum-sensing biofilm regulator;

b4366 bglJ Transcription factor Repressed

Transcriptional activator for the silent bgl operon; requires the bglJ4 allele to function; LuxR family

b2709 norR Transcription factor Repressed

Transcription regulator for norVW, NO-responsive; σ 54-dependent activator with a GAF domain

b0294 ecpR Transcription factor Repressed

Putative transcriptional regulator for the ecp operon

b2531 iscR Transcription factor Repressed

isc operon transcriptional repressor; suf operon transcriptional activator; icsR regulon regulator;

b2561 yfhH Transcription factor Repressed

putative DNA-binding transcriptional regulator

b0564 appY Transcription factor Activated

Global transcription regulator, AraC family, DLP12 prophage

b2714 ascG Transcription factor Activated

Repressor of asc operon; inducer unknown; prpBC operon repressor

Page 121: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

121

Table B-4. Continued.

Gene Locus Gene

Regulator Type Directon Description

b3556 cspA Transcription factor Activated

Cold-inducible RNA chaperone and antiterminator; aids gene expression at low temperature;

b3261 fis Transcription factor Activated

Transcriptional activator for rRNA operons, bends DNA; interacts with RNAP; nucleoid-associated protein;

b1891 flhC Transcription factor Activated

Transcriptional activator of flagellar class II operons; forms heterotetramer with FlhD;

b1892 flhD Transcription factor Activated

Transcriptional activator of flagellar class II operons; forms heterotetramer with FlhC

b1658 purR Transcription factor Activated Purine regulon repressor

b3481 nikR Transcription factor Activated

Nickel-responsive regulator of the nik operon; homodimer

b4116 adiY Transcription factor Activated

Transcriptional activator for adiA, AraC family

b0450 glnK Transcription factor Activated

Potent activator of NRII (GlnL/NtrB) phosphatase; trimeric.

b4128 ghoS Transcription factor Activated

Antitoxin of GhoTS toxin-antitoxin pair; endonuclease for ghoT mRNA

b3071 yqjI Transcription factor Activated

Transcriptional repressor for yqjH, nickel- or iron-inducible; autorepressor

b0483 ybaQ Transcription factor Activated

Predicted transcriptional regulator, function unknown

b1983 yeeN Transcription factor Activated

UPF0082 family protein, function unknown

b1988 nac Transcription factor Activated

Repressor of gdhA transcription; RpoN, GlnG (NtrC) regulons; pleiotropic effects

b3869 glnL Sensor kinase Activated

Bifunctional kinase/phosphatase, nitrogen regulator II, NRII; homodimeric

b1222 narX Sensor kinase Repressed

Two-component nitrate/nitrite sensor-transmitter protein; NarL is cognate regulator;

b1609 rstB Sensor kinase Repressed

Sensory histidine kinase of RstAB two-component system, low Mg-responsive via PhoQP

b2078 baeS Sensor kinase Repressed

Sensor kinase for mdtABCD, acrD and spy

Page 122: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

122

Table B-4. Continued.

Gene Locus Gene

Regulator Type Directon Description

b2503 yfgF Phosphodiesterase Activated

Cyclic-di-GMP phosphodiesterase, anaerobic; dual domain protein; defective cyclase domain; predicted membrane sensor protein

b1489 dosP Phosphodiesterase Repressed

Heme-regulated oxygen sensor, c-di-GMP phosphodiesterase; biofilm regulator;

b1815 yoaD Phosphodiesterase Repressed

Predicted membrane-anchored cyclic-di-GMP phosphodiesterase; regulation of cellulose production

b1285 gmr Phosphodiesterase Repressed

Cyclic-di-GMP phosphodiesterase; csgD regulator;

b1956 yedQ Diguanylate cyclase Repressed

Predicted membrane-anchored diguanylate cyclase

b2067 yegE Diguanylate cyclase Repressed

Predicted diguanylate cyclase, dual domain protein; defective phosphodiesterase domain;

b1341 ydaM Diguanylate cyclase Repressed

Diguanylate cyclase, csgD regulator; also regulates GGDEF protein YaiC (AdrA)

b1535 dgcZ Diguanylate cyclase Repressed Diguanylate cyclase, zinc-sensing

b1025 ycdT Diguanylate cyclase Repressed

Diguanylate cyclase, membrane-anchored

b1490 dosC Diguanylate cyclase Repressed

Diguanylate cyclase, binds oxygen, positive biofilm regulator; cold- and stationary phase-induced

b2741 rpoS σ factor Repressed RNA polymerase subunit, stress and stationary phase σ S; σ 38

b4293 fecI σ factor Repressed RNA polymerase σ-19 factor; fecA promoter RNAP σ factor

Page 123: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

123

Table B-5. sRNAs regulated by CsrA at the transcript level

Gene Locus Gene Direction Description

b4458 oxyS Repressed OxyS sRNA activates genes that detoxify oxidative damage

b4577 sgrS Repressed sRNA that destabilzes ptsG mRNA

b4441 glmY Repressed sRNA activator of glmS mRNA, glmZ processing antagonist

b4444 omrA Repressed sRNA downregulating OM proteins and curli; positively regulated by OmpR/EnvZ

b4445 omrB Repressed sRNA downregulating OM proteins and curli; positively regulated by OmpR/EnvZ

b4459 ryjA Repressed Novel sRNA, function unknown

b4698 mgrR Repressed sRNA affecting sensitivity to antimicrobial peptides; regulated by PhoPQ and Mg2+

b4699 fnrS Activated FNR-activated anaerobic sRNA; mediates negative FNR regulation; Hfq-dependent

Page 124: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

124

LIST OF REFERENCES Abu-Qatouseh, L.F., Chinni, S.V., Seggewiss, J., Proctor, R.A., Brosius, J.,

Rozhdestvensky, T.S., Peters, G., von Eiff, C., and Becker, K. (2010) Identification of differentially expressed small non-protein-coding RNAs in Staphylococcus aureus displaying both the normal and the small-colony variant phenotype. J Mol Med (Berl) 88: 565-575.

Afonyushkin, T., Vecerek, B., Moll, I., Bläsi, U., and Kaberdin, V.R. (2005) Both RNase E and RNase III control the stability of sodB mRNA upon translational inhibition by the small regulatory RNA RyhB. Nucleic Acids Res 33: 1678-1689.

Anders, S., Pyl, P.T., and Huber, W. (2015) HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 31: 166-169.

Andrade, J.M., Pobre, V., Matos, A.M., and Arraiano, C.M. (2012) The crucial role of PNPase in the degradation of small RNAs that are not associated with Hfq. RNA 18: 844-855.

Baba, T., Ara, T., Hasegawa, M., Takai, Y., Okumura, Y., Baba, M., Datsenko, K.A., Tomita, M., Wanner, B.L., and Mori, H. (2006) Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol Syst Biol 2: 2006.0008.

Babitzke, P., and Romeo, T. (2007) CsrB sRNA family: sequestration of RNA-binding regulatory proteins. Curr Opin Microbiol 10: 156-163.

Bachler, M., Schroeder, R., and von Ahsen, U. (1999) StreptoTag: a novel method for the isolation of RNA-binding proteins. RNA 5: 1509-1516.

Baker, C.S., Eöry, L.A., Yakhnin, H., Mercante, J., Romeo, T., and Babitzke, P. (2007) CsrA inhibits translation initiation of Escherichia coli hfq by binding to a single site overlapping the Shine-Dalgarno sequence. J Bacteriol 189: 5472-5481.

Baker, C.S., Morozov, I., Suzuki, K., Romeo, T., and Babitzke, P. (2002) CsrA regulates glycogen biosynthesis by preventing translation of glgC in Escherichia coli. Mol Microbiol 44: 1599-1610.

Baker, K.E., and Mackie, G.A. (2003) Ectopic RNase E sites promote bypass of 5'-end-dependent mRNA decay in Escherichia coli. Mol Microbiol 47: 75-88.

Balleza, E., López-Bojorquez, L.N., Martínez-Antonio, A., Resendis-Antonio, O., Lozada-Chávez, I., Balderas-Martínez, Y.I., Encarnación, S., and Collado-Vides, J. (2009) Regulation by transcription factors in bacteria: beyond description. FEMS Microbiol Rev 33: 133-151.

Page 125: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

125

Bandyra, K.J., Said, N., Pfeiffer, V., Górna, M.W., Vogel, J., and Luisi, B.F. (2012) The seed region of a small RNA drives the controlled destruction of the target mRNA by the endoribonuclease RNase E. Mol Cell 47: 943-953.

Bao, H., and Duong, F. (2013) Phosphatidylglycerol directs binding and inhibitory action of EIIAGlc protein on the maltose transporter. J Biol Chem 288: 23666-23674.

Barria, C., Malecki, M., and Arraiano, C.M. (2013) Bacterial adaptation to cold. Microbiology 159: 2437-2443.

Basineni, S.R., Madhugiri, R., Kolmsee, T., Hengge, R., and Klug, G. (2009) The influence of Hfq and ribonucleases on the stability of the small non-coding RNA OxyS and its target rpoS in E. coli is growth phase dependent. RNA Biol 6: 584-594.

Battesti, A., Majdalani, N., and Gottesman, S. (2011) The RpoS-mediated general stress response in Escherichia coli. Annu Rev Microbiol 65: 189-213.

Bhatt, S., Edwards, A.N., Nguyen, H.T., Merlin, D., Romeo, T., and Kalman, D. (2009) The RNA binding protein CsrA is a pleiotropic regulator of the locus of enterocyte effacement pathogenicity island of enteropathogenic Escherichia coli. Infect Immun 77: 3552-3568.

Bolivar, F., Rodriguez, R.L., Greene, P.J., Betlach, M.C., Heyneker, H.L., Boyer, H.W., Crosa, J.H., and Falkow, S. (1977) Construction and characterization of new cloning vehicles. II. A multipurpose cloning system. Gene 2: 95-113.

Botsford, J.L., and Harman, J.G. (1992) Cyclic AMP in prokaryotes. Microbiol Rev 56: 100-122.

Brennan, R.G., and Link, T.M. (2007) Hfq structure, function and ligand binding. Curr Opin Microbiol 10: 125-133.

Browning, D.F., and Busby, S.J. (2004) The regulation of bacterial transcription initiation. Nat Rev Microbiol 2: 57-65.

Burenina, O.Y., Elkina, D.A., Hartmann, R.K., Oretskaya, T.S., and Kubareva, E.A. (2015) Small Noncoding 6S RNAs of Bacteria. Biochemistry (Mosc) 80: 1429-1446.

Cai, M., Williams, D.C., Wang, G., Lee, B.R., Peterkofsky, A., and Clore, G.M. (2003) Solution structure of the phosphoryl transfer complex between the signal-transducing protein IIAGlucose and the cytoplasmic domain of the glucose transporter IICBGlucose of the Escherichia coli glucose phosphotransferase system. J Biol Chem 278: 25191-25206.

Page 126: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

126

Camacho, M.I., Alvarez, A.F., Chavez, R.G., Romeo, T., Merino, E., and Georgellis, D. (2015) Effects of the global regulator CsrA on the BarA/UvrY two-component signaling system. J Bacteriol 197: 983-991.

Cashel, M., and Gallant, J. (1969) Two compounds implicated in the function of the RC gene of Escherichia coli. Nature 221: 838-841.

Chaulk, S.G., Smith Frieday, M.N., Arthur, D.C., Culham, D.E., Edwards, R.A., Soo, P., Frost, L.S., Keates, R.A., Glover, J.N., and Wood, J.M. (2011) ProQ is an RNA chaperone that controls ProP levels in Escherichia coli. Biochemistry 50: 3095-3106.

Chavez, R.G., Alvarez, A.F., Romeo, T., and Georgellis, D. (2010) The physiological stimulus for the BarA sensor kinase. J Bacteriol 192: 2009-2012.

Chen, H., Dutta, T., and Deutscher, M.P. (2016) Growth Phase-dependent Variation of RNase BN/Z Affects Small RNAs: REGULATION OF 6S RNA. J Biol Chem 291: 26435-26442.

Chen, S., Oldham, M.L., Davidson, A.L., and Chen, J. (2013) Carbon catabolite repression of the maltose transporter revealed by X-ray crystallography. Nature 499: 364-368.

Commichau, F.M., and Stülke, J. (2012) A mystery unraveled: essentiality of RNase III in Bacillus subtilis is caused by resident prophages. PLoS Genet 8: e1003199.

Dalebroux, Z.D., and Swanson, M.S. (2012) ppGpp: magic beyond RNA polymerase. Nat Rev Microbiol 10: 203-212.

Dangerfield, J.A., Windbichler, N., Salmons, B., Günzburg, W.H., and Schröder, R. (2006) Enhancement of the StreptoTag method for isolation of endogenously expressed proteins with complex RNA binding targets. Electrophoresis 27: 1874-1877.

Datsenko, K.A., and Wanner, B.L. (2000) One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645.

Deikus, G., and Bechhofer, D.H. (2009) Bacillus subtilis trp Leader RNA: RNase J1 endonuclease cleavage specificity and PNPase processing. J Biol Chem 284: 26394-26401.

Deltcheva, E., Chylinski, K., Sharma, C.M., Gonzales, K., Chao, Y., Pirzada, Z.A., Eckert, M.R., Vogel, J., and Charpentier, E. (2011) CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471: 602-607.

Page 127: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

127

Deutscher, J., Aké, F.M., Derkaoui, M., Zébré, A.C., Cao, T.N., Bouraoui, H., Kentache, T., Mokhtari, A., Milohanic, E., and Joyet, P. (2014) The bacterial phosphoenolpyruvate:carbohydrate phosphotransferase system: regulation by protein phosphorylation and phosphorylation-dependent protein-protein interactions. Microbiol Mol Biol Rev 78: 231-256.

Deutscher, J., Francke, C., and Postma, P.W. (2006) How phosphotransferase system-related protein phosphorylation regulates carbohydrate metabolism in bacteria. Microbiol Mol Biol Rev 70: 939-1031.

Dubey, A.K., Baker, C.S., Romeo, T., and Babitzke, P. (2005) RNA sequence and secondary structure participate in high-affinity CsrA-RNA interaction. RNA 11: 1579-1587.

Dubey, A.K., Baker, C.S., Suzuki, K., Jones, A.D., Pandit, P., Romeo, T., and Babitzke, P. (2003) CsrA regulates translation of the Escherichia coli carbon starvation gene, cstA, by blocking ribosome access to the cstA transcript. J Bacteriol 185: 4450-4460.

Dugar, G., Svensson, S.L., Bischler, T., Wäldchen, S., Reinhardt, R., Sauer, M., and Sharma, C.M. (2016) The CsrA-FliW network controls polar localization of the dual-function flagellin mRNA in Campylobacter jejuni. Nat Commun 7: 11667.

Duss, O., Michel, E., Yulikov, M., Schubert, M., Jeschke, G., and Allain, F.H. (2014) Structural basis of the non-coding RNA RsmZ acting as a protein sponge. Nature 509: 588-592.

Dutta, T., and Deutscher, M.P. (2010) Mode of action of RNase BN/RNase Z on tRNA precursors: RNase BN does not remove the CCA sequence from tRNA. J Biol Chem 285: 22874-22881.

Dutta, T., Malhotra, A., and Deutscher, M.P. (2012) Exoribonuclease and endoribonuclease activities of RNase BN/RNase Z both function in vivo. J Biol Chem 287: 35747-35755.

Edwards, A.N., Patterson-Fortin, L.M., Vakulskas, C.A., Mercante, J.W., Potrykus, K., Vinella, D., Camacho, M.I., Fields, J.A., Thompson, S.A., Georgellis, D., Cashel, M., Babitzke, P., and Romeo, T. (2011) Circuitry linking the Csr and stringent response global regulatory systems. Mol Microbiol 80: 1561-1580.

Esquerré, T., Bouvier, M., Turlan, C., Carpousis, A.J., Girbal, L., and Cocaign-Bousquet, M. (2016) The Csr system regulates genome-wide mRNA stability and transcription and thus gene expression in Escherichia coli. Sci Rep 6: 25057.

Page 128: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

128

Even, S., Pellegrini, O., Zig, L., Labas, V., Vinh, J., Bréchemmier-Baey, D., and Putzer, H. (2005) Ribonucleases J1 and J2: two novel endoribonucleases in B.subtilis with functional homology to E.coli RNase E. Nucleic Acids Res 33: 2141-2152.

Feucht, B.U., and Saier, M.H. (1980) Fine control of adenylate cyclase by the phosphoenolpyruvate:sugar phosphotransferase systems in Escherichia coli and Salmonella typhimurium. J Bacteriol 141: 603-610.

Fic, E., Bonarek, P., Gorecki, A., Kedracka-Krok, S., Mikolajczak, J., Polit, A., Tworzydlo, M., Dziedzicka-Wasylewska, M., and Wasylewski, Z. (2009) cAMP receptor protein from Escherichia coli as a model of signal transduction in proteins--a review. J Mol Microbiol Biotechnol 17: 1-11.

Figueroa-Bossi, N., Schwartz, A., Guillemardet, B., D'Heygère, F., Bossi, L., and Boudvillain, M. (2014) RNA remodeling by bacterial global regulator CsrA promotes Rho-dependent transcription termination. Genes Dev 28: 1239-1251.

Filipowicz, W., Bhattacharyya, S.N., and Sonenberg, N. (2008) Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 9: 102-114.

Gabor, E., Göhler, A.K., Kosfeld, A., Staab, A., Kremling, A., and Jahreis, K. (2011) The phosphoenolpyruvate-dependent glucose-phosphotransferase system from Escherichia coli K-12 as the center of a network regulating carbohydrate flux in the cell. Eur J Cell Biol 90: 711-720.

Geissmann, T., Chevalier, C., Cros, M.J., Boisset, S., Fechter, P., Noirot, C., Schrenzel, J., François, P., Vandenesch, F., Gaspin, C., and Romby, P. (2009) A search for small noncoding RNAs in Staphylococcus aureus reveals a conserved sequence motif for regulation. Nucleic Acids Res 37: 7239-7257.

Georgellis, D., Arvidson, S., and von Gabain, A. (1992) Decay of ompA mRNA and processing of 9S RNA are immediately affected by shifts in growth rate, but in opposite manners. J Bacteriol 174: 5382-5390.

Goller, C., Wang, X., Itoh, Y., and Romeo, T. (2006) The cation-responsive protein NhaR of Escherichia coli activates pgaABCD transcription, required for production of the biofilm adhesin poly-beta-1,6-N-acetyl-D-glucosamine. J Bacteriol 188: 8022-8032.

Gottesman, S., and Storz, G. (2011) Bacterial small RNA regulators: versatile roles and rapidly evolving variations. Cold Spring Harb Perspect Biol 3.

Gudapaty, S., Suzuki, K., Wang, X., Babitzke, P., and Romeo, T. (2001) Regulatory interactions of Csr components: the RNA binding protein CsrA activates csrB transcription in Escherichia coli. J Bacteriol 183: 6017-6027.

Page 129: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

129

Guo, M.S., Updegrove, T.B., Gogol, E.B., Shabalina, S.A., Gross, C.A., and Storz, G. (2014) MicL, a new σE-dependent sRNA, combats envelope stress by repressing synthesis of Lpp, the major outer membrane lipoprotein. Genes Dev 28: 1620-1634.

Gutiérrez, P., Li, Y., Osborne, M.J., Pomerantseva, E., Liu, Q., and Gehring, K. (2005) Solution structure of the carbon storage regulator protein CsrA from Escherichia coli. J Bacteriol 187: 3496-3501.

Guzzo, C.R., Salinas, R.K., Andrade, M.O., and Farah, C.S. (2009) PILZ protein structure and interactions with PILB and the FIMX EAL domain: implications for control of type IV pilus biogenesis. J Mol Biol 393: 848-866.

Górna, M.W., Carpousis, A.J., and Luisi, B.F. (2012) From conformational chaos to robust regulation: the structure and function of the multi-enzyme RNA degradosome. Q Rev Biophys 45: 105-145.

Göpel, Y., and Görke, B. (2012) Rewiring two-component signal transduction with small RNAs. Curr Opin Microbiol 15: 132-139.

Göpel, Y., Papenfort, K., Reichenbach, B., Vogel, J., and Görke, B. (2013) Targeted decay of a regulatory small RNA by an adaptor protein for RNase E and counteraction by an anti-adaptor RNA. Genes Dev 27: 552-564.

Hajnsdorf, E., and Boni, I.V. (2012) Multiple activities of RNA-binding proteins S1 and Hfq. Biochimie 94: 1544-1553.

Hajnsdorf, E., Braun, F., Haugel-Nielsen, J., and Régnier, P. (1995) Polyadenylylation destabilizes the rpsO mRNA of Escherichia coli. Proc Natl Acad Sci U S A 92: 3973-3977.

Hankins, J.S., Denroche, H., and Mackie, G.A. (2010) Interactions of the RNA-binding protein Hfq with cspA mRNA, encoding the major cold shock protein. J Bacteriol 192: 2482-2490.

Haugo, A.J., and Watnick, P.I. (2002) Vibrio cholerae CytR is a repressor of biofilm development. Mol Microbiol 45: 471-483.

Heeb, S., Kuehne, S.A., Bycroft, M., Crivii, S., Allen, M.D., Haas, D., Cámara, M., and Williams, P. (2006) Functional analysis of the post-transcriptional regulator RsmA reveals a novel RNA-binding site. J Mol Biol 355: 1026-1036.

Hengge, R. (2009) Principles of c-di-GMP signalling in bacteria. Nat Rev Microbiol 7: 263-273.

Hernández-Pérez, L., Depardón, F., Fernández-Ramírez, F., Sánchez-Trujillo, A., Bermúdez-Crúz, R.M., Dangott, L., and Montañez, C. (2011) α-Enolase binds to RNA. Biochimie 93: 1520-1528.

Page 130: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

130

Hogema, B.M., Arents, J.C., Bader, R., Eijkemans, K., Yoshida, H., Takahashi, H., Aiba, H., and Postma, P.W. (1998) Inducer exclusion in Escherichia coli by non-PTS substrates: the role of the PEP to pyruvate ratio in determining the phosphorylation state of enzyme IIAGlc. Mol Microbiol 30: 487-498.

Holmqvist, E., Wright, P.R., Li, L., Bischler, T., Barquist, L., Reinhardt, R., Backofen, R., and Vogel, J. (2016) Global RNA recognition patterns of post-transcriptional regulators Hfq and CsrA revealed by UV crosslinking in vivo. EMBO J 35: 991-1011.

Huang, Y., Suyemoto, M., Garner, C.D., Cicconi, K.M., and Altier, C. (2008) Formate acts as a diffusible signal to induce Salmonella invasion. J Bacteriol 190: 4233-4241.

Hulko, M., Berndt, F., Gruber, M., Linder, J.U., Truffault, V., Schultz, A., Martin, J., Schultz, J.E., Lupas, A.N., and Coles, M. (2006) The HAMP domain structure implies helix rotation in transmembrane signaling. Cell 126: 929-940.

Hurley, J.H., Faber, H.R., Worthylake, D., Meadow, N.D., Roseman, S., Pettigrew, D.W., and Remington, S.J. (1993) Structure of the regulatory complex of Escherichia coli IIIGlc with glycerol kinase. Science 259: 673-677.

Jenal, U., Reinders, A., and Lori, C. (2017) Cyclic di-GMP: second messenger extraordinaire. Nat Rev Microbiol 15: 271-284.

Johansen, J., Rasmussen, A.A., Overgaard, M., and Valentin-Hansen, P. (2006) Conserved small non-coding RNAs that belong to the sigmaE regulon: role in down-regulation of outer membrane proteins. J Mol Biol 364: 1-8.

Jonas, K., Edwards, A.N., Ahmad, I., Romeo, T., Römling, U., and Melefors, O. (2010) Complex regulatory network encompassing the Csr, c-di-GMP and motility systems of Salmonella Typhimurium. Environ Microbiol 12: 524-540.

Jonas, K., Edwards, A.N., Simm, R., Romeo, T., Römling, U., and Melefors, O. (2008) The RNA binding protein CsrA controls cyclic di-GMP metabolism by directly regulating the expression of GGDEF proteins. Mol Microbiol 70: 236-257.

Jørgensen, M.G., Thomason, M.K., Havelund, J., Valentin-Hansen, P., and Storz, G. (2013) Dual function of the McaS small RNA in controlling biofilm formation. Genes Dev 27: 1132-1145.

Kaberdin, V.R., and Bläsi, U. (2013) Bacterial helicases in post-transcriptional control. Biochim Biophys Acta 1829: 878-883.

Kawamoto, H., Koide, Y., Morita, T., and Aiba, H. (2006) Base-pairing requirement for RNA silencing by a bacterial small RNA and acceleration of duplex formation by Hfq. Mol Microbiol 61: 1013-1022.

Page 131: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

131

Ke, S.H., and Madison, E.L. (1997) Rapid and efficient site-directed mutagenesis by single-tube 'megaprimer' PCR method. Nucleic Acids Res 25: 3371-3372.

Kim, Y.J., Ryu, Y., Koo, B.M., Lee, N.Y., Chun, S.J., Park, S.J., Lee, K.H., and Seok, Y.J. (2010) A mammalian insulysin homolog is regulated by enzyme IIAGlc of the glucose transport system in Vibrio vulnificus. FEBS Lett 584: 4537-4544.

Koo, B.M., Yoon, M.J., Lee, C.R., Nam, T.W., Choe, Y.J., Jaffe, H., Peterkofsky, A., and Seok, Y.J. (2004) A novel fermentation/respiration switch protein regulated by enzyme IIAGlc in Escherichia coli. J Biol Chem 279: 31613-31621.

Krin, E., Sismeiro, O., Danchin, A., and Bertin, P.N. (2002) The regulation of Enzyme IIAGlc expression controls adenylate cyclase activity in Escherichia coli. Microbiology 148: 1553-1559.

Langmead, B., and Salzberg, S.L. (2012) Fast gapped-read alignment with Bowtie 2. Nat Methods 9: 357-359.

Lawhon, S.D., Frye, J.G., Suyemoto, M., Porwollik, S., McClelland, M., and Altier, C. (2003) Global regulation by CsrA in Salmonella typhimurium. Mol Microbiol 48: 1633-1645.

Lee, E.J., and Groisman, E.A. (2010) An antisense RNA that governs the expression kinetics of a multifunctional virulence gene. Mol Microbiol 76: 1020-1033.

Lee, H.J., and Gottesman, S. (2016) sRNA roles in regulating transcriptional regulators: Lrp and SoxS regulation by sRNAs. Nucleic Acids Res 44: 6907-6923.

Lee, S.J., Boos, W., Bouché, J.P., and Plumbridge, J. (2000) Signal transduction between a membrane-bound transporter, PtsG, and a soluble transcription factor, Mlc, of Escherichia coli. EMBO J 19: 5353-5361.

Leng, Y., Vakulskas, C.A., Zere, T.R., Pickering, B.S., Watnick, P.I., Babitzke, P., and Romeo, T. (2016) Regulation of CsrB/C sRNA decay by EIIA(Glc) of the phosphoenolpyruvate: carbohydrate phosphotransferase system. Mol Microbiol 99: 627-639.

Lengeler, J.W., and Jahreis, K. (2009) Bacterial PEP-dependent carbohydrate: phosphotransferase systems couple sensing and global control mechanisms. Contrib Microbiol 16: 65-87.

Lenz, D.H., Miller, M.B., Zhu, J., Kulkarni, R.V., and Bassler, B.L. (2005) CsrA and three redundant small RNAs regulate quorum sensing in Vibrio cholerae. Mol Microbiol 58: 1186-1202.

Page 132: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

132

Li, B., Li, N., Wang, F., Guo, L., Huang, Y., Liu, X., Wei, T., Zhu, D., Liu, C., Pan, H., Xu, S., Wang, H.W., and Gu, L. (2012) Structural insight of a concentration-dependent mechanism by which YdiV inhibits Escherichia coli flagellum biogenesis and motility. Nucleic Acids Res 40: 11073-11085.

Link, A.J., Phillips, D., and Church, G.M. (1997) Methods for generating precise deletions and insertions in the genome of wild-type Escherichia coli: application to open reading frame characterization. J Bacteriol 179: 6228-6237.

Liu, M.Y., Gui, G., Wei, B., Preston, J.F., Oakford, L., Yüksel, U., Giedroc, D.P., and Romeo, T. (1997) The RNA molecule CsrB binds to the global regulatory protein CsrA and antagonizes its activity in Escherichia coli. J Biol Chem 272: 17502-17510.

Liu, M.Y., Yang, H., and Romeo, T. (1995) The product of the pleiotropic Escherichia coli gene csrA modulates glycogen biosynthesis via effects on mRNA stability. J Bacteriol 177: 2663-2672.

Liu, R., Holik, A.Z., Su, S., Jansz, N., Chen, K., Leong, H.S., Blewitt, M.E., Asselin-Labat, M.L., Smyth, G.K., and Ritchie, M.E. (2015) Why weight? Modelling sample and observational level variability improves power in RNA-seq analyses. Nucleic Acids Res 43: e97.

Lux, R., Jahreis, K., Bettenbrock, K., Parkinson, J.S., and Lengeler, J.W. (1995) Coupling the phosphotransferase system and the methyl-accepting chemotaxis protein-dependent chemotaxis signaling pathways of Escherichia coli. Proc Natl Acad Sci U S A 92: 11583-11587.

Lévy, S., Zeng, G.Q., and Danchin, A. (1990) Cyclic AMP synthesis in Escherichia coli strains bearing known deletions in the pts phosphotransferase operon. Gene 86: 27-33.

Lüttmann, D., Heermann, R., Zimmer, B., Hillmann, A., Rampp, I.S., Jung, K., and Görke, B. (2009) Stimulation of the potassium sensor KdpD kinase activity by interaction with the phosphotransferase protein IIANtr in Escherichia coli. Mol Microbiol 72: 978-994.

Mackie, G.A. (2013) RNase E: at the interface of bacterial RNA processing and decay. Nat Rev Microbiol 11: 45-57.

Mandin, P., and Gottesman, S. (2010) Integrating anaerobic/aerobic sensing and the general stress response through the ArcZ small RNA. EMBO J 29: 3094-3107.

Mandin, P., and Guillier, M. (2013) Expanding control in bacteria: interplay between small RNAs and transcriptional regulators to control gene expression. Curr Opin Microbiol 16: 125-132.

Page 133: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

133

Martínez, L.C., Martínez-Flores, I., Salgado, H., Fernández-Mora, M., Medina-Rivera, A., Puente, J.L., Collado-Vides, J., and Bustamante, V.H. (2014) In silico identification and experimental characterization of regulatory elements controlling the expression of the Salmonella csrB and csrC genes. J Bacteriol 196: 325-336.

Martínez, L.C., Yakhnin, H., Camacho, M.I., Georgellis, D., Babitzke, P., Puente, J.L., and Bustamante, V.H. (2011) Integration of a complex regulatory cascade involving the SirA/BarA and Csr global regulatory systems that controls expression of the Salmonella SPI-1 and SPI-2 virulence regulons through HilD. Mol Microbiol 80: 1637-1656.

Massé, E., Escorcia, F.E., and Gottesman, S. (2003) Coupled degradation of a small regulatory RNA and its mRNA targets in Escherichia coli. Genes Dev 17: 2374-2383.

Massé, E., and Gottesman, S. (2002) A small RNA regulates the expression of genes involved in iron metabolism in Escherichia coli. Proc Natl Acad Sci U S A 99: 4620-4625.

Mazé, A., Glatter, T., and Bumann, D. (2014) The central metabolism regulator EIIAGlc switches Salmonella from growth arrest to acute virulence through activation of virulence factor secretion. Cell Rep 7: 1426-1433.

Mercante, J., Edwards, A.N., Dubey, A.K., Babitzke, P., and Romeo, T. (2009) Molecular geometry of CsrA (RsmA) binding to RNA and its implications for regulated expression. J Mol Biol 392: 511-528.

Mercante, J., Suzuki, K., Cheng, X., Babitzke, P., and Romeo, T. (2006) Comprehensive alanine-scanning mutagenesis of Escherichia coli CsrA defines two subdomains of critical functional importance. J Biol Chem 281: 31832-31842.

Michaux, C., Holmqvist, E., Vasicek, E., Sharan, M., Barquist, L., Westermann, A.J., Gunn, J.S., and Vogel, J. (2017) RNA target profiles direct the discovery of virulence functions for the cold-shock proteins CspC and CspE. Proc Natl Acad Sci U S A 114: 6824-6829.

Mika, F., and Hengge, R. (2014) Small RNAs in the control of RpoS, CsgD, and biofilm architecture of Escherichia coli. RNA Biol 11: 494-507.

Mohanty, B.K., Maples, V.F., and Kushner, S.R. (2004) The Sm-like protein Hfq regulates polyadenylation dependent mRNA decay in Escherichia coli. Mol Microbiol 54: 905-920.

Moll, I., Afonyushkin, T., Vytvytska, O., Kaberdin, V.R., and Bläsi, U. (2003) Coincident Hfq binding and RNase E cleavage sites on mRNA and small regulatory RNAs. RNA 9: 1308-1314.

Page 134: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

134

Morita, T., Kawamoto, H., Mizota, T., Inada, T., and Aiba, H. (2004) Enolase in the RNA degradosome plays a crucial role in the rapid decay of glucose transporter mRNA in the response to phosphosugar stress in Escherichia coli. Mol Microbiol 54: 1063-1075.

Morita, T., Maki, K., and Aiba, H. (2005) RNase E-based ribonucleoprotein complexes: mechanical basis of mRNA destabilization mediated by bacterial noncoding RNAs. Genes Dev 19: 2176-2186.

Moyle, H., Waldburger, C., and Susskind, M.M. (1991) Hierarchies of base pair preferences in the P22 ant promoter. J Bacteriol 173: 1944-1950.

Mukherjee, S., Yakhnin, H., Kysela, D., Sokoloski, J., Babitzke, P., and Kearns, D.B. (2011) CsrA-FliW interaction governs flagellin homeostasis and a checkpoint on flagellar morphogenesis in Bacillus subtilis. Mol Microbiol 82: 447-461.

Møller, T., Franch, T., Udesen, C., Gerdes, K., and Valentin-Hansen, P. (2002) Spot 42 RNA mediates discoordinate expression of the E. coli galactose operon. Genes Dev 16: 1696-1706.

Nam, T.W., Cho, S.H., Shin, D., Kim, J.H., Jeong, J.Y., Lee, J.H., Roe, J.H., Peterkofsky, A., Kang, S.O., Ryu, S., and Seok, Y.J. (2001) The Escherichia coli glucose transporter enzyme IICBGlc recruits the global repressor Mlc. EMBO J 20: 491-498.

Neumann, S., Grosse, K., and Sourjik, V. (2012) Chemotactic signaling via carbohydrate phosphotransferase systems in Escherichia coli. Proc Natl Acad Sci U S A 109: 12159-12164.

Opdyke, J.A., Kang, J.G., and Storz, G. (2004) GadY, a small-RNA regulator of acid response genes in Escherichia coli. J Bacteriol 186: 6698-6705.

Pannuri, A., Vakulskas, C.A., Zere, T., McGibbon, L.C., Edwards, A.N., Georgellis, D., Babitzke, P., and Romeo, T. (2016) Circuitry Linking the Catabolite Repression and Csr Global Regulatory Systems of Escherichia coli. J Bacteriol 198: 3000-3015.

Papenfort, K., and Vogel, J. (2011) Sweet business: Spot42 RNA networks with CRP to modulate catabolite repression. Mol Cell 41: 245-246.

Park, Y.H., Lee, B.R., Seok, Y.J., and Peterkofsky, A. (2006) In vitro reconstitution of catabolite repression in Escherichia coli. J Biol Chem 281: 6448-6454.

Patten, C.L., Kirchhof, M.G., Schertzberg, M.R., Morton, R.A., and Schellhorn, H.E. (2004) Microarray analysis of RpoS-mediated gene expression in Escherichia coli K-12. Mol Genet Genomics 272: 580-591.

Page 135: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

135

Patterson-Fortin, L.M., Vakulskas, C.A., Yakhnin, H., Babitzke, P., and Romeo, T. (2013) Dual posttranscriptional regulation via a cofactor-responsive mRNA leader. J Mol Biol 425: 3662-3677.

Pernestig, A.K., Georgellis, D., Romeo, T., Suzuki, K., Tomenius, H., Normark, S., and Melefors, O. (2003) The Escherichia coli BarA-UvrY two-component system is needed for efficient switching between glycolytic and gluconeogenic carbon sources. J Bacteriol 185: 843-853.

Pickering, B.S., Smith, D.R., and Watnick, P.I. (2012) Glucose-specific enzyme IIA has unique binding partners in the vibrio cholerae biofilm. MBio 3: e00228-00212.

Postma, P.W., Epstein, W., Schuitema, A.R., and Nelson, S.O. (1984) Interaction between IIIGlc of the phosphoenolpyruvate:sugar phosphotransferase system and glycerol kinase of Salmonella typhimurium. J Bacteriol 158: 351-353.

Potrykus, K., and Cashel, M. (2008) (p)ppGpp: still magical? Annu Rev Microbiol 62: 35-51.

Prévost, K., Desnoyers, G., Jacques, J.F., Lavoie, F., and Massé, E. (2011) Small RNA-induced mRNA degradation achieved through both translation block and activated cleavage. Genes Dev 25: 385-396.

Reddy, P., and Kamireddi, M. (1998) Modulation of Escherichia coli adenylyl cyclase activity by catalytic-site mutants of protein IIAGlc of the phosphoenolpyruvate:sugar phosphotransferase system. J Bacteriol 180: 732-736.

Reimmann, C., Valverde, C., Kay, E., and Haas, D. (2005) Posttranscriptional repression of GacS/GacA-controlled genes by the RNA-binding protein RsmE acting together with RsmA in the biocontrol strain Pseudomonas fluorescens CHA0. J Bacteriol 187: 276-285.

Romeo, T. (1996) Post-transcriptional regulation of bacterial carbohydrate metabolism: evidence that the gene product CsrA is a global mRNA decay factor. Res Microbiol 147: 505-512.

Romeo, T. (1998) Global regulation by the small RNA-binding protein CsrA and the non-coding RNA molecule CsrB. Mol Microbiol 29: 1321-1330.

Romeo, T., Gong, M., Liu, M.Y., and Brun-Zinkernagel, A.M. (1993) Identification and molecular characterization of csrA, a pleiotropic gene from Escherichia coli that affects glycogen biosynthesis, gluconeogenesis, cell size, and surface properties. J Bacteriol 175: 4744-4755.

Page 136: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

136

Romeo, T., Vakulskas, C.A., and Babitzke, P. (2013) Post-transcriptional regulation on a global scale: form and function of Csr/Rsm systems. Environ Microbiol 15: 313-324.

Römling, U., Galperin, M.Y., and Gomelsky, M. (2013) Cyclic di-GMP: the first 25 years of a universal bacterial second messenger. Microbiol Mol Biol Rev 77: 1-52.

Sabnis, N.A., Yang, H., and Romeo, T. (1995) Pleiotropic regulation of central carbohydrate metabolism in Escherichia coli via the gene csrA. J Biol Chem 270: 29096-29104.

Sahr, T., Rusniok, C., Impens, F., Oliva, G., Sismeiro, O., Coppée, J.Y., and Buchrieser, C. (2017) The Legionella pneumophila genome evolved to accommodate multiple regulatory mechanisms controlled by the CsrA-system. PLoS Genet 13: e1006629.

Saier, M.H. (1989) Protein phosphorylation and allosteric control of inducer exclusion and catabolite repression by the bacterial phosphoenolpyruvate: sugar phosphotransferase system. Microbiol Rev 53: 109-120.

Saramago, M., Bárria, C., Dos Santos, R.F., Silva, I.J., Pobre, V., Domingues, S., Andrade, J.M., Viegas, S.C., and Arraiano, C.M. (2014) The role of RNases in the regulation of small RNAs. Curr Opin Microbiol 18: 105-115.

Sauer, E., and Weichenrieder, O. (2011) Structural basis for RNA 3'-end recognition by Hfq. Proc Natl Acad Sci U S A 108: 13065-13070.

Schubert, M., Lapouge, K., Duss, O., Oberstrass, F.C., Jelesarov, I., Haas, D., and Allain, F.H. (2007) Molecular basis of messenger RNA recognition by the specific bacterial repressing clamp RsmA/CsrA. Nat Struct Mol Biol 14: 807-813.

Sharma, C.M., Darfeuille, F., Plantinga, T.H., and Vogel, J. (2007) A small RNA regulates multiple ABC transporter mRNAs by targeting C/A-rich elements inside and upstream of ribosome-binding sites. Genes Dev 21: 2804-2817.

Smirnov, A., Förstner, K.U., Holmqvist, E., Otto, A., Günster, R., Becher, D., Reinhardt, R., and Vogel, J. (2016) Grad-seq guides the discovery of ProQ as a major small RNA-binding protein. Proc Natl Acad Sci U S A 113: 11591-11596.

Sterzenbach, T., Nguyen, K.T., Nuccio, S.P., Winter, M.G., Vakulskas, C.A., Clegg, S., Romeo, T., and Bäumler, A.J. (2013) A novel CsrA titration mechanism regulates fimbrial gene expression in Salmonella typhimurium. EMBO J 32: 2872-2883.

Storz, G., Vogel, J., and Wassarman, K.M. (2011) Regulation by small RNAs in bacteria: expanding frontiers. Mol Cell 43: 880-891.

Page 137: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

137

Studier, F.W., and Moffatt, B.A. (1986) Use of bacteriophage T7 RNA polymerase to direct selective high-level expression of cloned genes. J Mol Biol 189: 113-130.

Sudarsan, N., Lee, E.R., Weinberg, Z., Moy, R.H., Kim, J.N., Link, K.H., and Breaker, R.R. (2008) Riboswitches in eubacteria sense the second messenger cyclic di-GMP. Science 321: 411-413.

Suzuki, K., Babitzke, P., Kushner, S.R., and Romeo, T. (2006) Identification of a novel regulatory protein (CsrD) that targets the global regulatory RNAs CsrB and CsrC for degradation by RNase E. Genes Dev 20: 2605-2617.

Suzuki, K., Wang, X., Weilbacher, T., Pernestig, A.K., Melefors, O., Georgellis, D., Babitzke, P., and Romeo, T. (2002) Regulatory circuitry of the CsrA/CsrB and BarA/UvrY systems of Escherichia coli. J Bacteriol 184: 5130-5140.

Tanaka, Y., Kimata, K., and Aiba, H. (2000) A novel regulatory role of glucose transporter of Escherichia coli: membrane sequestration of a global repressor Mlc. EMBO J 19: 5344-5352.

Thelin, K.H., and Taylor, R.K. (1996) Toxin-coregulated pilus, but not mannose-sensitive hemagglutinin, is required for colonization by Vibrio cholerae O1 El Tor biotype and O139 strains. Infect Immun 64: 2853-2856.

Thomason, M.K., Fontaine, F., De Lay, N., and Storz, G. (2012) A small RNA that regulates motility and biofilm formation in response to changes in nutrient availability in Escherichia coli. Mol Microbiol 84: 17-35.

Timmermans, J., and Van Melderen, L. (2009) Conditional essentiality of the csrA gene in Escherichia coli. J Bacteriol 191: 1722-1724.

Tsai, Y.C., Du, D., Domínguez-Malfavón, L., Dimastrogiovanni, D., Cross, J., Callaghan, A.J., García-Mena, J., and Luisi, B.F. (2012) Recognition of the 70S ribosome and polysome by the RNA degradosome in Escherichia coli. Nucleic Acids Res 40: 10417-10431.

Tschowri, N., Busse, S., and Hengge, R. (2009) The BLUF-EAL protein YcgF acts as a direct anti-repressor in a blue-light response of Escherichia coli. Genes Dev 23: 522-534.

Udekwu, K.I., Darfeuille, F., Vogel, J., Reimegård, J., Holmqvist, E., and Wagner, E.G. (2005) Hfq-dependent regulation of OmpA synthesis is mediated by an antisense RNA. Genes Dev 19: 2355-2366.

Urban, A., Neukirchen, S., and Jaeger, K.E. (1997) A rapid and efficient method for site-directed mutagenesis using one-step overlap extension PCR. Nucleic Acids Res 25: 2227-2228.

Page 138: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

138

Uzzau, S., Figueroa-Bossi, N., Rubino, S., and Bossi, L. (2001) Epitope tagging of chromosomal genes in Salmonella. Proc Natl Acad Sci U S A 98: 15264-15269.

Vakulskas, C.A., Leng, Y., Abe, H., Amaki, T., Okayama, A., Babitzke, P., Suzuki, K., and Romeo, T. (2016) Antagonistic control of the turnover pathway for the global regulatory sRNA CsrB by the CsrA and CsrD proteins. Nucleic Acids Res 44: 7896-7910.

Vakulskas, C.A., Pannuri, A., Cortés-Selva, D., Zere, T.R., Ahmer, B.M., Babitzke, P., and Romeo, T. (2014) Global effects of the DEAD-box RNA helicase DeaD (CsdA) on gene expression over a broad range of temperatures. Mol Microbiol 92: 945-958.

Vakulskas, C.A., Potts, A.H., Babitzke, P., Ahmer, B.M., and Romeo, T. (2015) Regulation of Bacterial Virulence by Csr (Rsm) Systems. Microbiol Mol Biol Rev 79: 193-224.

Van Assche, E., Van Puyvelde, S., Vanderleyden, J., and Steenackers, H.P. (2015) RNA-binding proteins involved in post-transcriptional regulation in bacteria. Front Microbiol 6: 141.

Vaucheret, H. (2006) Post-transcriptional small RNA pathways in plants: mechanisms and regulations. Genes Dev 20: 759-771.

Viegas, S.C., Silva, I.J., Saramago, M., Domingues, S., and Arraiano, C.M. (2011) Regulation of the small regulatory RNA MicA by ribonuclease III: a target-dependent pathway. Nucleic Acids Res 39: 2918-2930.

Vogel, J., and Luisi, B.F. (2011) Hfq and its constellation of RNA. Nat Rev Microbiol 9: 578-589.

Wang, G., Louis, J.M., Sondej, M., Seok, Y.J., Peterkofsky, A., and Clore, G.M. (2000) Solution structure of the phosphoryl transfer complex between the signal transducing proteins HPr and IIAGlucose of the Escherichia coli phosphoenolpyruvate:sugar phosphotransferase system. EMBO J 19: 5635-5649.

Wang, X., Dubey, A.K., Suzuki, K., Baker, C.S., Babitzke, P., and Romeo, T. (2005) CsrA post-transcriptionally represses pgaABCD, responsible for synthesis of a biofilm polysaccharide adhesin of Escherichia coli. Mol Microbiol 56: 1648-1663.

Wang, X., Preston, J.F., and Romeo, T. (2004) The pgaABCD locus of Escherichia coli promotes the synthesis of a polysaccharide adhesin required for biofilm formation. J Bacteriol 186: 2724-2734.

Wassarman, K.M., and Storz, G. (2000) 6S RNA regulates E. coli RNA polymerase activity. Cell 101: 613-623.

Page 139: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

139

Weber, H., Polen, T., Heuveling, J., Wendisch, V.F., and Hengge, R. (2005) Genome-wide analysis of the general stress response network in Escherichia coli: sigmaS-dependent genes, promoters, and sigma factor selectivity. J Bacteriol 187: 1591-1603.

Wei, B.L., Brun-Zinkernagel, A.M., Simecka, J.W., Prüss, B.M., Babitzke, P., and Romeo, T. (2001) Positive regulation of motility and flhDC expression by the RNA-binding protein CsrA of Escherichia coli. Mol Microbiol 40: 245-256.

Weilbacher, T., Suzuki, K., Dubey, A.K., Wang, X., Gudapaty, S., Morozov, I., Baker, C.S., Georgellis, D., Babitzke, P., and Romeo, T. (2003) A novel sRNA component of the carbon storage regulatory system of Escherichia coli. Mol Microbiol 48: 657-670.

Windbichler, N., and Schroeder, R. (2006) Isolation of specific RNA-binding proteins using the streptomycin-binding RNA aptamer. Nat Protoc 1: 637-640.

Woodcock, D.M., Crowther, P.J., Doherty, J., Jefferson, S., DeCruz, E., Noyer-Weidner, M., Smith, S.S., Michael, M.Z., and Graham, M.W. (1989) Quantitative evaluation of Escherichia coli host strains for tolerance to cytosine methylation in plasmid and phage recombinants. Nucleic Acids Res 17: 3469-3478.

Wurm, R., Neusser, T., and Wagner, R. (2010) 6S RNA-dependent inhibition of RNA polymerase is released by RNA-dependent synthesis of small de novo products. Biol Chem 391: 187-196.

Xu, F., Lin-Chao, S., and Cohen, S.N. (1993) The Escherichia coli pcnB gene promotes adenylylation of antisense RNAI of ColE1-type plasmids in vivo and degradation of RNAI decay intermediates. Proc Natl Acad Sci U S A 90: 6756-6760.

Yakhnin, A.V., Baker, C.S., Vakulskas, C.A., Yakhnin, H., Berezin, I., Romeo, T., and Babitzke, P. (2013) CsrA activates flhDC expression by protecting flhDC mRNA from RNase E-mediated cleavage. Mol Microbiol 87: 851-866.

Yakhnin, H., Yakhnin, A.V., Baker, C.S., Sineva, E., Berezin, I., Romeo, T., and Babitzke, P. (2011) Complex regulation of the global regulatory gene csrA: CsrA-mediated translational repression, transcription from five promoters by Eσ70 and EσS, and indirect transcriptional activation by CsrA. Mol Microbiol 81: 689-704.

Yang, H., Liu, M.Y., and Romeo, T. (1996) Coordinate genetic regulation of glycogen catabolism and biosynthesis in Escherichia coli via the CsrA gene product. J Bacteriol 178: 1012-1017.

Page 140: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

140

Zere, T.R., Vakulskas, C.A., Leng, Y., Pannuri, A., Potts, A.H., Dias, R., Tang, D., Kolaczkowski, B., Georgellis, D., Ahmer, B.M., and Romeo, T. (2015) Genomic Targets and Features of BarA-UvrY (-SirA) Signal Transduction Systems. PLoS One 10: e0145035.

Page 141: ufdcimages.uflib.ufl.edu · 4 ACKNOWLEDGMENTS I want to express my deepest gratitude to my advisor, Dr. Tony Romeo for giving me the opportunity to work in his lab and for his guidance,

141

BIOGRAPHICAL SKETCH

Yuanyuan Leng was born in Weifang, Shandong, China. She received her

bachelor’s degree in biotechonology from Shandong University, China in June of

2009. She then joined the program of State Key Laboratory of Microbial Technology at

Shandong University and received her master’s degree in microbiology in June of

2012. After that, in August of 2012, she joined the graduate program at the

Department of Microbiology and Cell Science at the University of Florida. After three

rotations, she joined Dr. Tony Romeo’s lab and worked with him to study the

regulation of the RNase E-mediated turnover of sRNAs CsrB/C. She received her

Ph.D. from the University of Florida in the summer of 2017. Yuanyuan Leng plans to

continue research training as a postdoctoral associate at National Cancer Institute.