oxidative stress resistance in the francisella tularensis live vaccine strain is associated with

166
University of Iowa University of Iowa Iowa Research Online Iowa Research Online Theses and Dissertations Spring 2016 Oxidative stress resistance in the Francisella tularensis live Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with genetic variability in the ferrous vaccine strain is associated with genetic variability in the ferrous iron uptake gene feoB iron uptake gene feoB Joshua Robert Fletcher University of Iowa Follow this and additional works at: https://ir.uiowa.edu/etd Part of the Genetics Commons Copyright 2016 Joshua Robert Fletcher This dissertation is available at Iowa Research Online: https://ir.uiowa.edu/etd/3083 Recommended Citation Recommended Citation Fletcher, Joshua Robert. "Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with genetic variability in the ferrous iron uptake gene feoB." PhD (Doctor of Philosophy) thesis, University of Iowa, 2016. https://doi.org/10.17077/etd.c5lqs7hk Follow this and additional works at: https://ir.uiowa.edu/etd Part of the Genetics Commons

Upload: others

Post on 11-Sep-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

University of Iowa University of Iowa

Iowa Research Online Iowa Research Online

Theses and Dissertations

Spring 2016

Oxidative stress resistance in the Francisella tularensis live Oxidative stress resistance in the Francisella tularensis live

vaccine strain is associated with genetic variability in the ferrous vaccine strain is associated with genetic variability in the ferrous

iron uptake gene feoB iron uptake gene feoB

Joshua Robert Fletcher University of Iowa

Follow this and additional works at: https://ir.uiowa.edu/etd

Part of the Genetics Commons

Copyright 2016 Joshua Robert Fletcher

This dissertation is available at Iowa Research Online: https://ir.uiowa.edu/etd/3083

Recommended Citation Recommended Citation Fletcher, Joshua Robert. "Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with genetic variability in the ferrous iron uptake gene feoB." PhD (Doctor of Philosophy) thesis, University of Iowa, 2016. https://doi.org/10.17077/etd.c5lqs7hk

Follow this and additional works at: https://ir.uiowa.edu/etd

Part of the Genetics Commons

Page 2: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

Oxidative stress resistance in the Francisella tularensis Live Vaccine Strain is

associated with genetic variability in the ferrous iron uptake gene feoB

by

Joshua Robert Fletcher

A thesis submitted in partial fulfillment

of the requirements for the Doctor of Philosophy

degree in Genetics in the

Graduate College of

The University of Iowa

May 2016

Thesis Supervisor: Professor Bradley D. Jones

Page 3: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

Copyright by

JOSHUA ROBERT FLETCHER

2016

All Rights Reserved

Page 4: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

Graduate College The University of Iowa

Iowa City, Iowa

CERTIFICATE OF APPROVAL

____________________________

PH.D. THESIS

_________________

This is to certify that the Ph.D. thesis of

Joshua Robert Fletcher

has been approved by the Examining Committee for the thesis requirement for the Doctor of Philosophy degree in Genetics at the May 2016 graduation. Thesis Committee: ____________________________________________ Bradley D. Jones, Thesis Supervisor ____________________________________________ Lee-Ann H. Allen ____________________________________________ Craig D. Ellermeier ____________________________________________ Mary E. Wilson ____________________________________________ John R. Kirby

Page 5: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

ii

To my family.

Page 6: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

iii

ACKNOWLEDGEMENTS

No scientist’s endeavors occur in a vacuum, unless you happen to be a

computer scientist in the 1940’s or an astronaut. I am neither. I owe much

gratitude to: my friends and colleagues Matt Faron and Jed Rasmussen, two

fellow graduate students in the lab who were soundboards for my ideas more

often than they probably wanted to be, and who offered many a great

conversation about books, science, life, the universe and Rick Astley’s place in it;

everyone in the Francisella group who helped me learn the field and think critically

about the details of experiments; Bram Slutter from the Harty lab for his help and

expertise with T cell biology; our collaborator Katy Bosio from Rocky Mountain

Labs for being generous with data and strains that served as the foundation for

this document; my mentor Brad Jones for giving me space in the lab and the

freedom to explore it; the Genetics program for admitting me and allowing me to

develop as a scientist. Lastly, I owe an enormous debt of gratitude to my friends

and family for supporting me even though I often put science ahead of them.

Page 7: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

iv

ABSTRACT

Francisella tularensis is a highly virulent bacterial pathogen with an

extremely low infectious dose (~10 CFU) and high rates of mortality if left

untreated (30-60%). F. tularensis has an extensive history as a bioweapon, and

there is no vaccine currently licensed. For these reasons the CDC considers F.

tularensis a Tier 1 Select Agent. The unlicensed F. tularensis subsp. holarctica

Live Vaccine Strain (LVS) provides moderate protection against virulent strains;

however, we have discovered that various “wild type” lab stocks differ in their

virulence and ability to confer immunity. Genome sequencing of high virulence

(RML, LD50 ~200 CFU) and low virulence (ATCC, LD50 ~9,000 CFU) strains has

identified nine differences, of which four are non-synonymous substitutions. One

such mutation occurs in the ferrous iron uptake gene feoB in RML. While iron is

required for cellular function, ferrous iron (Fe2+) can participate in the Fenton

reaction with H2O2, leading to inactivation of essential iron-sulfur cluster

enzymes. Part of the innate immune response involves mitochondria-derived

reactive oxygen species in the cytosol. Fully virulent strains of F. tularensis are

known to be highly resistant to such host defenses, and have low levels of

intracellular iron. Accordingly, the RML strain was highly resistant to exogenous

H2O2 in vitro relative to the ATCC strain. An iron-responsive lacZ reporter had

~2-fold higher induction in the RML strain relative to ATCC during iron limitation.

Overexpression of the functional feoB allele, but not the RML allele, leads to

significantly increased sensitivity to H2O2.

Page 8: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

v

Given the connection of iron and H2O2 toxicity, I revisited a previously

published transposon screen to determine if any of the mutants identified had a

role in iron homeostasis and oxidative stress resistance. One such gene was

annotated as bacterioferritin (bfr), which in other bacteria forms a hollow,

spherical multimer that oxidizes Fe2+ to Fe3+ and stores the oxidized form in the

interior of the sphere. The Δbfr mutant was ~10-fold more sensitive to H2O2 and

was attenuated nearly 8-fold in murine intranasal infection in terms of LD50

relative to the parental RML strain. Importantly, the Δbfr mutant allowed us to

test the hypothesis that H2O2 resistance is critical for the RML LVS to stimulate

productive immunity. At six weeks post-infection, mice previously infected with

either RML or the Δbfr mutant were challenged with an infection of 25 CFU of the

fully virulent F. tularensis Schu S4 strain. All mice immunized with RML survived

this challenge, while all mice immunized with Δbfr succumbed; only displaying a

slight increase in time to death. These results are consistent with the hypothesis

that the H2O2 resistance of RML LVS mediates increased fitness in a host.

Page 9: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

vi

PUBLIC ABSTRACT

Many organisms on Earth require oxygen and iron to support their growth

and metabolism, including many bacteria that can cause disease. One such

bacterium, Francisella tularensis, can cause severe pneumonia that may lead to

death. The current vaccine against infection comes from a live but weakened

version of this bacterium, though it is not very effective. One of the goals of this

thesis was to examine the DNA from different lots of this vaccine strain to see if

there were genetic differences between lots that were more or less effective at

providing immunity. This analysis found that the effective vaccines had a genetic

mutation that made these particular strains of the weakened bacteria less able to

gather iron from their environment. Certain forms of iron can be highly reactive

with oxygen, which can be seen as rust on metal. This form of iron is important

for certain metabolic reactions in most living things, but too much of it can be

toxic to life. Iron can react with hydrogen peroxide, a common antiseptic.

Hydrogen peroxide is also made by the cells of the immune system to kill

invading pathogens. My research found that the more effective vaccine strains of

Francisella tularensis were resistant to the toxic effects of hydrogen peroxide

because they had less iron. I showed that genetically manipulating this strain to

make it have more iron made it sensitive to hydrogen peroxide and made it a less

effective vaccine.

Page 10: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

vii

Table of Contents

LIST OF FIGURES ...............................................................................................ix

TABLE: PRIMER SEQUENCES ........................................................................... 1

CHAPTER I .......................................................................................................... 3

INTRODUCTION .................................................................................................. 3 Francisella tularensis early history .................................................................... 3 Tularemia .......................................................................................................... 6 F. tularensis taxonomy and ecology .................................................................. 8 Francisella tularensis genetics ........................................................................ 10 Bacterial factors that mediate pathogenesis .................................................... 13 Iron uptake in F. tularensis .............................................................................. 17 Intracellular lifecycle ........................................................................................ 20 Immunity to Francisella species ...................................................................... 26

CHAPTER II ....................................................................................................... 30

A VIRULENT BIOVAR OF F. TULARENSIS LVS IS INTRINSICALLY MORE RESISTANT TO HYDROGEN PEROXIDE ........................................................ 30

Introduction ..................................................................................................... 30 Materials and Methods .................................................................................... 32 Results ............................................................................................................ 36

LVS biovars have similar PiglA-lacZ expression and lack low molecular weight O-antigen glycosylated proteins........................................................ 36 RML LVS has less intracellular Fe2+ than the ATCC or Iowa LVS ............... 38 FeoB D471Y does not complement E. coli ΔfeoB fhuF::λplac ..................... 42 Resistance to H2O2 correlates with feoB D471Y allele ................................. 43 Resistance to H2O2 also requires Dyp peroxidase ....................................... 45 Neither feoB nor dyp are essential for intracellular growth in A549 or BMM cells .................................................................................................... 47

Discussion ....................................................................................................... 62

CHAPTER III ...................................................................................................... 67

CHARACTERIZATION OF BACTERIOFERRITIN IN OXIDATIVE STRESS RESISTANCE ..................................................................................................... 67

Introduction ..................................................................................................... 67 Materials and methods .................................................................................... 69 Results ............................................................................................................ 72

A bacterioferritin mutant has modest reduction in PfslA-lacZ activity ............. 72 Bacterioferritin promoter activity is not significantly induced in iron limiting media ............................................................................................... 73 Bacterioferritin protects against H2O2 but is not required for intracellular growth in A549 cells ..................................................................................... 75

Page 11: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

viii

Bacterioferritin contributes to virulence and immunogenicity of RML LVS in vivo ........................................................................................................... 75

CHAPTER IV ...................................................................................................... 92

CONCLUSIONS AND FUTURE DIRECTIONS .................................................. 92

APPENDIX A .................................................................................................... 109

DEVELOPMENT OF RECOMBINANT IGLC-LCMV EPITOPE T CELL REPORTER FUSIONS ..................................................................................... 109

Rationale ....................................................................................................... 109 Materials and methods .................................................................................. 110 Results and discussion .................................................................................. 112

APPENDIX B .................................................................................................... 120

OXYR DOES NOT INFLUENCE OXIDATIVE STRESS RESISTANCE IN F. TULARENSIS LVS ........................................................................................... 121

Rationale ....................................................................................................... 121 Results and discussion .................................................................................. 124

REFERENCES ................................................................................................. 129

Page 12: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

ix

LIST OF FIGURES

Figure II.1. RML LVS has similar FPI reporter gene expression and lacks glycosylated proteins seen in virulent Type A and Type B strains of F. tularensis. ..................................................................................................... 49

Figure II.2. Location of the amino acid residue that is mutated in the RML LVS FeoB. .................................................................................................... 50

Figure II.3. RML LVS exhibits growth restriction under iron limitation on MMH agar. ................................................................................................... 51

Figure II.4. RML LVS has iron-related gene expression consistent with less intracellular Fe2+ in iron limiting media. ....................................................... 52

Figure II.5. Constitutive fur expression inhibits robust growth of RML LVS. ....... 53

Figure II.6. RML feoB allele does not complement an E. coli ΔfeoB iron reporter strain. .............................................................................................. 54

Figure II.7. RML LVS is significantly more resistant to H2O2 lethality. ............... 55

Figure II.8. RML LVS can be sensitized to H2O2 by overexpression of a functional feoB. ............................................................................................ 56

Figure II.9. FeoB-mediated sensitivity to H2O2 is independent of the RML LVS genetic background. ............................................................................. 57

Figure II.10. Dyp peroxidase protects against H2O2. ......................................... 58

Figure II.11. Fold growth of LVS biovars in A549 cells. ...................................... 59

Figure II.12. Contribution of feoB alleles to intracellular growth in A549 cells. ... 60

Figure II.13. Twenty-four-hour growth of LVS biovars and mutant derivatives in murine bone marrow-derived macrophages. ............................................ 61

Figure III.1. Bacterioferritin promoter activity is similar between LVS biovars grown in Chamberlain’s defined medium with high or low iron. .................... 78

Figure III.2. Bacterioferritin promoter activity in is upregulated in ∆feoB and downregulated in Δbfr in MMH broth. ........................................................... 79

Figure III.3. Mutants lacking bfr and feoB differ in their H2O2 sensitivity depending on phase of growth. .................................................................... 80

Figure III.4. The bacterioferritin mutant is proficient for intracellular growth in vitro. ............................................................................................................. 81

Figure III.5. RML Δbfr is modestly attenuated in murine infection via the intranasal route of inoculation. ..................................................................... 82

Page 13: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

x

Figure III.6. RML Δbfr-infected mice lost less weight than mice infected with the wild type RML LVS. ................................................................................ 83

Figure III.7. RML requires bacterioferritin to elicit protection against an intranasal challenge of 25 CFU F. tularensis Schu S4. Mice were infected with either 121 CFU of RML LVS or 215 CFU of the bacterioferritin mutant and allowed to recover for fix weeks before challenge. ..................................................................................................... 84

Figure IV.I Model of iron import in F. tularensis subsp. holarctica Live Vaccine Strain (LVS). ................................................................................. 107

Figure IV.II Model of intracellular outcomes for high and low iron strains. ........ 108

Figure A.I. Low dose immunization survival curves. ......................................... 116

Figure A.II. High dose immunization survival curves. ....................................... 117

Figure A.III. IglC-LCMV epitope fusions are expressed in LVS and do not alter intracellular growth. ............................................................................ 118

Figure A.IV. LVS infected mouse lymphocyte response to LCMV antigens ex vivo. ....................................................................................................... 119

Figure A.V. Survival of LCMV immunized mice challenged with LVS expressing LCMV epitopes. ....................................................................... 120

Figure B.I. The RML LVS ∆oxyR mutant is as resistant to H2O2 as the wild type. ........................................................................................................... 127

Figure B.II. The ΔoxyR mutant has a mild intracellular growth defect in J774A.16 cells. ........................................................................................... 128

Page 14: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

1

TABLE: PRIMER SEQUENCES

5’feoB.up.asc1.bamh1 – ggcgcgccggatccAGCATATCAAACACAAAGAAGAAGTATTG 3’feoB.up.avr2 – cctaggAATCAGTTTTCAGGAGTTTATAGTATAG 5’feoB.down.asc1.avr2 – ggcgcgcccctaggAATTCTAATTTGAATATACAGCTTTA 3’feoB.down.bgl2 – agatctTTAGCATTTCACTAAGATTTGCACC 5’feoB.mut.check – AGATGATTGCTCAGTAATAACAGCAACTTTGC 3’feoB.mut.check – CATTAATAAGAATAGTATTCTCATTTTAAAATACCTC 5’feoB.SNP – AGACTTGCGATATTTTCAGTATTTGC 3’feoB.SNP – TTTACCGGCATATTCAAGTGCTGTGG 5’dyp.up.asc1.bamh1 – ggcgcgccggatccCCATCATCAAAGCTTTGATTTGGG 3’dyp.up.avr2 – cctaggTTAACTTTTCCTTATAAAACGCTC 5’dyp.down.asc1.avr2 – ggcgcgcccctaggAACAAGCTACTTGAAATTCTTTATTTTATA 3’dyp.down.bgl2 - agatctCATAGTACTCAACAAACTTGCCAAC 5’feoBc.kpn1 – ggtaccATGAAATATGCTCTAGTTGGCAATCC 3’feoBc.sal1 – gtcgacATATTTAAAGCTGTATATTCAAATTAG 5’dyp.mut.check – AGTATCCATACGATAATAAAGTTAGTTATAGAAGC 3’dyp.mut.check - ATAGAGAATATTTCTTTCCTTTATCCTCTATTTGTAGC 5’dyp.comp.kpn1 – ggtaccGTGGAAATAATTAAATATCAATTAGGAATAG 3’dyp.comp.sal1 – gtcgacCTAATTTAGTAAAGAAATATCTAGTTTGCC 5’bferr.up.asc1.bamh1 – ggcgcgccggatccTTTTAGTGATACTTTTTGAGACAATTGTCCC 3’bferr.up.asc1.avr2 – ggcgcgcccctaggCATATTGTTACCTCCATTATTTAAAACTCTAATC 5’bferr.down.asc1 – ggcgcgccTAAAGGCTATTATCCTCGATGAGTTTTTCTTC 3’bferr.down.bamh1 – ggatccAAGTATTTATCTGTAGTTACAATGGTGG 5’bfr.mut.check – ATATCATTTTTATTAAAATATCTAGGTTG 3’bfr.mut.check – ATAAATACTTTAAGTCACTAAATATCTCG 5’Pbfr.bamh1 -ggatccATATCATTTTTATTAAAATATCTAGGTTG 3’Pbfr.kpn1 – ggtaccATTGTTACCTCCATTATTTAAAACTCTAATC 5’iglC.kpn1 – ggtaccATGATTATGAGTGAGATGATAACAAG

Page 15: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

2

3’iglC.sal1 - gtcgacCTATGCAGCTGCAATATATCCTATTTTAGC 3’iglC – CTATGCAGCTGCAATATATCCTATTTTAGC 3’iglC.GP33.41 – gtcgacCTATAATATACCCATAGTAGCACCATTATATACAGCTTTTATTGAAGTTGCAGCTGCAATATATCCTATTTTAGCAACTCC gp61.80.1c - GCACAGATAAAGGAGTTGCTAAAATAGGATATATTGCAGCTGCAGGTATGTATGGTTTAAAAGGTCCTGATATATATAAAGG gp61.80.1r – CCTTTATATATATCAGGACCTTTTAAACCATACATACCTGCAGCTGCAATATATCCTATTTTAGCAACTCCTTTATCTGTGC Gp61.80.2c – GGTATGTATGGTTTAAAAGGTCCTGATATATATAAAGGTGTATATCAATTTAAATCAGTAGAATTTGATATGTCACATTAGgtcgac Gp61.80.2r – gtcgacCTAATGTGACATATCAAATTCTACTGATTTAAATTGATATACACCTTTATATATATCAGGACCTTTTAAACCATACATACC 5’oxyR.up – ACTATCCATACAGTCGACTCTAGAGGATCACCAGCTACAGACTTAAGATAAGCATTTGC 3’oxyR.up – AACTAAATCTTATAGTTACTATAAATATTAT 5’oxyR.down – GCTCACATAAATATCATCCAAATACCC 3’oxyR.down – GATGAATTCGAGCTCGGTACCCGGGTTAATTTTAAGGATGGTAAGCG 5’oxyR.up.JC84.ITA – ACTATCCATACAGTCGACTCTAGAGGATCggcgcgccggatccACCAGCTACAGACTTAAGATAAGCATTTGC 3’oxyR.down.JC84.ITA – GATGAATTCGAGCTCGGTACCCGGGGATCggatccGTTAATTTTAAGGATGGTAAGCG 5’oxyR.mut.check – GTTAAGCTACGCTATTATTGTATTACTCTTGC 3’oxyR.mut.check – AGGATTTTTATCATTAACAGAAAATATTATGATG

Page 16: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

3

CHAPTER I

INTRODUCTION

Francisella tularensis early history

The bacterium that was eventually named Francisella tularensis was

isolated in 1911 from squirrels in California by George McCoy and Charles

Chapin, researchers working at the United States Public Health and Marine

Service who were investigating disease epidemiology in squirrels3. Infected

squirrels were described as having plague-like buboes, enlarged spleens, and

lesions in their livers. McCoy and Chapin eventually isolated the causative agent

and characterized it as being quite fastidious, requiring nutrient rich media or

passage through animals for propagation. The disease caused by this bacterium

was differentiated from that caused by Yersinia pestis by infecting plague-

immune guinea pigs with either Y. pestis or the newly discovered pathogen.

They observed that the guinea pigs infected with the latter quickly succumbed,

while the Y. pestis challenged animals survived, suggesting that the newly

discovered bacterium was not immunologically similar to Y. pestis. Not long after

their initial description of the disease and the causative agent, the first case of

human infection with Francisella tularensis was reported4. The infected

individual had recently prepared meat at a restaurant; he presented with

conjunctivitis and developed swollen lymph nodes and the infectious agent

isolated from his conjunctival ulcers matched the description of the organism

described by McCoy and Chapin.

Page 17: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

4

Increased case reports over the following decade made it clear that a

major risk factor for contracting the disease was through the handling or

ingestion of infected meat or by insect bite, typically ticks or deer flies5,6. Edward

Francis, for whom the bacteria is named, characterized outbreaks of tularemia in

rural populations of the Pahvant Valley region of Utah, describing the

ulceroglandular form of the disease as occurring following the bite of a deer fly

that had previously bitten an infected rabbit; Dr. Francis also recorded an early

case of lethal pneumonic tularemia7. In his studies he found that the organism

can persist in lice associated with rodents, rabbits and hares, squirrel-associated

fleas, as well as bedbugs8,9. During the time period in which he was conducting

these studies, Dr. Francis also described the infection of six laboratory workers

who had been working with him to characterize the tularemia outbreaks in Utah.

Two of the individuals were performing field work and may have contracted the

infection naturally, but the remaining four became infected under then standard

laboratory conditions during propagation and characterization of the bacterium7.

Francis later notes that laboratory worker infection during routine passage of the

bacterium through animals was so dangerous that the Lister Institute of

Preventative Medicine in London, England suspended work with the organism7.

The symptoms experienced by both the initial patients and infected laboratory

personnel highlight two of the hallmark features of F. tularensis: 1) the organism

is highly infectious, and 2) the disease it causes is debilitating and can result in

fatality.

Page 18: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

5

The extreme infectiousness of F. tularensis and the incapacitation caused

by tularemia did not escape the notice of the governments of Imperial Japan, the

Soviet Union, and the United States, who in the early and mid-20th century

devoted significant resources towards developing biological weapons

programs10-14. While the United States had forsworn offensive bioweapons

capabilities in 1972, evidence exists that the Soviet Union maintained active

bioweapons stockpiles and research programs through its dissolution in the early

1990s5. Given this information and the ubiquity of F. tularensis in the

environment throughout the Northern hemisphere, fears of the intentional release

of aerosols of the bacteria are not without merit. A model produced by the World

Health Organization estimated that aerosol release of F. tularensis over a large

metropolitan area would likely result in approximately 250,000 casualties and

upwards of 19,000 fatalities15. A more recent model put forth by the Centers for

Disease Control and Prevention estimated that an attack on a population center

of 100,000 persons could result in greater than 6,000 deaths and up to $5.4

billion in associated costs for treatments and prophylaxis16. Indeed, these

scenarios have recently spurred considerable research efforts that are focused

on understanding the pathogenesis of F. tularensis and developing effective

vaccines against the disease. To date there is no licensed vaccine for prevention

of tularemia, however an attenuated derivative of F. tularensis subsp. holarctica

called the Live Vaccine Strain or LVS has been used to vaccinate laboratory

workers in the past11. Vaccination proved useful in reducing incidences of

laboratory acquired infections, but LVS does not protect against moderate doses

Page 19: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

6

of aerosolized F. tularensis subsp. tularensis, and the details of the strain’s

creation are unclear. Furthermore, as the nature of the attenuating mutations in

LVS were not fully understood, there were concerns about potential reversion or

suppression of these mutations that could make LVS virulent again, thus the

strain was discontinued for human use in the United States.

Tularemia

The disease caused by F. tularensis infection can manifest in multiple

ways depending on the route of infection. Typical cases of tularemia are

associated with general flu-like symptoms, such as fever, aches, sore throat, and

occasionally dry cough. The most common form of tularemia (~75-80% of

cases), ulceroglandular, presents with a cutaneous ulcer at the site of infection,

with malaise, chills and fever setting in approximately three or more days after

the infection17,18. This is followed by the swelling of draining lymph nodes, which

can continue to enlarge to the point of suppuration. The ulcerous tissue can

undergo necrosis and forms a structure that is not unlike that of an anthrax-like

eschar. While not associated with high rates of mortality, this form of the disease

can progress to secondary pneumonic tularemia or sepsis by dissemination of

the bacteria to other organs. Oculoglandular tularemia occurs when bacteria are

introduced to the mucus membranes of the eye, and is characterized by

inflammation and ulceration of the conjunctiva; the cervical lymph nodes may

became swollen18. Oropharyngeal tularemia, while rare, occurs after ingestion of

Page 20: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

7

contaminated materials and is associated with oral ulcers, cervical lymph node

swelling, tonsillitis, gastrointestinal discomfort and occasionally diarrhea18,19.

The most serious form of the disease, pneumonic tularemia, results from

the inhalation of F. tularensis and is associated with high mortality in the absence

of antibiotic administration, as high as 60%17,20. This form of the disease is

characterized by symptoms typical of tularemia, as well as a non-productive

cough and chest pains that become worse as the disease progresses, bloody

sputum, pleural effusion, possible hemorrhage, and dissemination of the

organisms to the liver and spleen17,18. If untreated, this can lead to organ failure

and death. While rare, outbreaks of pneumonic tularemia occur in areas where

rodent or lagomorph populations have high rates of infection with F. tularensis,

such as Martha’s Vineyard in Massachusetts, rural areas of Scandinavia or areas

where sanitation and hygiene services have been discontinued due to war or

natural disaster21-25. Outbreaks in these locations are associated with aerosols

of F. tularensis that had been generated by running lawnmowers over infected

rabbit carcasses, and the movement of hay that likely had contact with infected

rodents or their carcasses, as well as contamination of water and food supplies.

Pneumonic tularemia would likely be the most common presentation of the

disease in the event of intentional release of F. tularensis, thus careful

surveillance is needed to distinguish between this and naturally occurring

outbreaks.

Page 21: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

8

F. tularensis taxonomy and ecology

F. tularensis is a member of the γ-proteobacteria, though it is

phylogenetically distant from its most closely-related genera, Wolbachia,

Coxiella, and Legionella26. The Francisella genus has three recognized species:

F. tularensis, F. philomiragia, and F. novicida. The latter two species are rarely

associated with disease in humans; little is known about F. philomiragia, and only

immune compromised individuals are at increased risk of infection with F.

novicida27. The two disease-causing subspecies of F. tularensis are known as

Type A (subsp. tularensis) and Type B (subsp. holarctica), and can be found

across the northern hemisphere28. F. tularensis subsp. tularensis is endemic to

North America and causes the severe forms of tularemia that are associated with

high mortality rates, while subsp. holarctica causes significant but rarely lethal

disease, and can be found in North America, Europe, and Asia. Type A strains

exhibit relatively high genetic diversity within the subspecies, while Type B strains

tend to have lower diversity, consistent with a proposed bottleneck event

sometime in the past29-31. It is interesting to speculate that the reduced virulence

of F. tularensis subsp. holarctica may have contributed to its rapid spread across

the northern hemisphere, as prolonged windows of time for insect mediated

transmission events would likely occur in populations of sick animals where Type

B strains are endemic.

A comprehensive genotyping analysis has identified two major clades

within Type A F. tularensis, called Type A.I (itself further separated into A.Ia and

A.Ib) and A.II32. Type A.I and A.II strains have been shown to be slightly more

Page 22: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

9

virulent than the prototypical virulent laboratory strain, F. tularensis subsp.

tularensis Schu S4 in mouse models of infection33. Epidemiological data suggest

that A.Ib strains cause the most morbidity and mortality among documented

human infections in the United States, while A.Ia and A.II strains are associated

with less mortality than even Type B strains32. A third subspecies of F. tularensis

has been isolated from Central Asia, F. tularensis subsp. mediasiatica that

exhibits some virulence in rabbit, but it is not clear if the organism is connected to

disease in man34.

The two subclades of subsp. tularensis strains exhibit differences in

geographic distributions, with the distribution of common vector species for each

subclade closely overlapping. The A.I strains are typically found in the eastern

portion of North America and are commonly associated with infections stemming

from tick (Dermacentor variabilis or Amblyomma americanum) bites or contact

with infected rabbit flesh30. As these tick species have been reported to harbor F.

tularensis for long periods of time throughout multiple stages of their life cycles, it

is not surprising that a majority (~60%) of recorded tularemia infections in the

central and southern regions of the United States reported contact with ticks near

the time of infection35-37. In contrast, the A.II strains are found in the western

United States; transmission and infection are associated with Dermacentor

andersoni ticks and the deer-fly Chysops discalis30. As with Type A strains, ticks

serve as important reservoirs and vectors for Type B strains in North America

and Europe. Typically, these strains are transmitted by ticks from the Ixodes and

Page 23: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

10

Dermacentor genera, though others may be associated, as well as Laelaps mites

that prey on small rodent species34.

The transmission cycles of both subspecies involve infection of rodents

and lagomorphs through tick and fly vectors, and it is known that tularemia

outbreaks in small mammal populations are associated with outbreaks of human

tularemia38. Interestingly, F. tularensis has an enormous range of hosts that it

can infect, possibly greater than 250 species, including amoebae in soil and

aquatic environments, yet large scale human outbreaks are relatively rare39,40.

Some species are highly susceptible to infection, such as mice, and may not

survive long enough for significant transmission to insect vectors. It is also

possible that the many of the species that can be infected with F. tularensis do

not encounter any of the insect vectors in the wild, or do not contract significant

enough disease upon infection to transmit bacteria to insect vectors.

Francisella tularensis genetics

At approximately 1.89 million base pairs in length, the F. tularensis

genome is relatively small compared to other intracellular pathogens and is AT

rich, with approximately 33% GC content26. Genome comparison between the

two virulent subspecies and a species that is nonpathogenic for humans, F.

novicida, indicates that a high degree of similarity exists in the Francisella genus.

The F. novicida genome has approximately 97-98% identity among conserved

sequences between it and the F. tularensis species, and nearly 86% of the core

genome is common to all three41. F. novicida is an environmental species

Page 24: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

11

usually associated with aquatic environments that exhibits virulence in mice,

though significant differences exist in terms of the disease when compared to

that caused by F. tularensis42. The high degree of similarity between Francisella

species is remarkable given the extreme difference in virulence and disease

phenotypes, especially when one considers the paucity of genes dedicated solely

to virulence, such as those encoding Type III or Type IV secretion systems used

by other intracellular pathogens like Salmonella or Legionella26. One notable

exception is the presence of an approximately 30 kilobase locus, termed the

Francisella Pathogenicity Island (FPI) that encodes genes with weak similarity to

Type VI secretion systems43,44. This locus encodes key functions for the

intracellular lifecycle of Francisella species, and interestingly, two functioning

copies of the FPI are encoded on the chromosomes of subsp. tularensis and

subsp. holarctica, highlighting the importance of these genes for the

pathogenesis of the bacteria. The FPI will be discussed further in a later section.

A key feature of the genomes of the virulent species of Francisella that

distinguishes them from that of F. novicida are the high number of insertion

sequence (IS) elements and genomic rearrangements that have occurred as a

consequence of homologous recombination between these IS elements. Indeed,

Rohmer, et. al. found that the genomes of subsp. holarctica LVS and subsp.

tularensis Schu S4 have 109 and 79 IS elements, respectively, while that of F.

novicida only contains 2641. A consequence of the expansion of these IS

elements is both an increase in the number of genes inactivated by insertion of

these elements and an increase in genomic rearrangements in the virulent

Page 25: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

12

species relative to F. novicida. The organization within genomic blocks and the

presence of conserved sequences flanking the IS elements that have mediated

recombination events suggests that a significant number of these changes

occurred in the ancestor of subsp. tularensis and subsp. holarctica, though

further subspecies-specific changes have occurred since their divergence41. The

proliferation of these IS elements is consistent with what is observed in the

genomes of other pathogenic bacteria. For example, pathogenic species of

Bordetella, Shigella, Yersina, Burkholderia, as well as Mycobacterium leprae all

have genomic signatures of proliferation of IS elements and gene loss or decay,

suggesting that adaptation to the pathogenic lifestyle is associated with loss of

so-called “anti-virulence genes” that are not essential or even deleterious to the

infection process45-49. Many of the genes lost or non-functional in the virulent

species of Francisella encode proteins that are involved in metabolic pathways,

in particular those for amino acid biosynthesis41. In fact, F. tularensis requires

supplementation of thirteen amino acids and numerous other nutrients, such as

thiamine and β-nicotinamide adenine dinucleotide, to support its growth in

standard laboratory culture. This implies that F. tularensis readily obtains these

essential molecules in the preferred niche of the host cell environment.

The F. tularensis subsp. tularensis and subsp. holarctica genomes differ

primarily by the chromosomal location of large blocks of synteny that have

rearranged independently in each subspecies, however the gene order and

nucleotide identity within these blocks are highly conserved41. With the exception

of pseudogenes, the two subspecies differ very little genetically. Over half of the

Page 26: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

13

annotated pseudogenes (160) in each subspecies appear to be inactivated by

the same mutation, which suggests that they occurred before the subspecies

diverged and may have been important for the adaptation to a pathogenic

lifestyle, though a further 94 and 143 pseudogenes have occurred since these

subspecies split41. It is unknown whether all of the pseudogenes unique to each

subspecies contribute to the differences in their virulence, but it is reasonable to

hypothesize that at least some do. The F. tularensis subsp. holarctica LVS

chromosome is highly similar to that of a prototypical subsp. holarctica strain in

terms of synteny blocks and pseudogenes, though LVS has fifteen unique

deletions and pseudogenes that likely account for the strain’s attenuation50. A

deletion that lead to the loss of the pilin gene pilA and the partial deletion of the 3’

end of FTL_0391 and the 5’ end of FTL_0392 that created a gene fusion have

been shown to account for most, if not all of the attenuation of LVS in mouse

models of infection51. Interestingly, low-level genetic variation also exists within

the LVS lineage, some of which is the focus of this thesis.

Bacterial factors that mediate pathogenesis

For a pathogen of such high virulence, F. tularensis lacks many of the

common virulence factors used by other pathogenic bacteria, such as toxins,

hemolysins, and Types III or IV secretion systems26,52,53. While transposon

screens have identified hundreds of genes that affect the ability of the bacteria to

replicate in a host cell or cause disease in vivo, many of the mutations disrupt

genes that are important for the general fitness of the organism, such as those

Page 27: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

14

involved in ribosome biogenesis or protein folding, and relatively few appear to

be dedicated virulence factors54-56. It should be noted, though, that a significant

portion of mutants identified in a F. tularensis Schu S4 transposon screen

encoded genes of unknown function or those without obvious homologs in other

bacterial species, suggesting that they may represent novel virulence factors

unique to Francisella.

The genes within the Francisella Pathogenicity Island (FPI) are among the

few that appear to be specific to the pathogenesis of the bacteria. Studies into F.

tularensis LVS during growth in the cytoplasm of J774 macrophage-like cell line

identified a number of bacterial proteins as being upregulated, including a 23 kilo

Dalton protein that would be later described as belonging to the FPI57.

Interestingly, Golovliov, et. al. noted that this protein was also highly induced

when the bacteria were exposed to hydrogen peroxide, suggesting that oxidative

stress may be a signal within the host cell that the bacteria are able to detect and

respond to with changes in gene expression. Soon after this discovery, F.

novicida transposon mutants were identified as failing to grow in murine

macrophages; one of the mutants mapped to the gene encoding the 23 kilo

Dalton protein and another mapped to a gene upstream within the same operon

(called the intracellular growth locus, or igl; the 23 kilo Dalton protein was named

IglC)58. Subsequent development of tools for molecular genetic analysis and

genome sequencing demonstrated that this locus was duplicated in the

chromosomes of the virulent F. tularensis26,59. These early genetic studies also

found that iglC mutants fail to inhibit the production of proinflammatory cytokines

Page 28: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

15

in J774 cells and human peripheral blood monocytic cells, indicating that a major

component of F. tularensis pathogenesis is to suppress host responses that

typically would result in recognition and clearance of a bacterium60,61.

Further genetic analysis expanded the number of genes linked to the igl

locus, and showed that they belonged to a pathogenicity island of approximately

30 kilo bases and 19 open reading frames43. The GC content of the FPI is

considerably lower than that of the rest of the Francisella chromosome,

suggesting that it is derived from a horizontal transfer event or is the remnant of a

phage. Indeed, bioinformatic analysis indicates that the FPI encodes a secretion

system very distantly related to the Type VI secretion systems of other bacteria,

such as Pseudomonas and Vibrio species, and these systems have similarity to

the tail spike of certain bacteriophages62-64. Numerous studies have found

evidence that FPI proteins can gain access to the cytoplasm of infected cells,

though evidence suggests that some FPI proteins can still reach the cytoplasm in

the absence of a functioning FPI65-69. Recent structural analysis indicates that

the FPI does indeed encode a Type VI secretion system that likely functions

similarly to those of other bacteria70. While the mechanistic details still need

further investigation, it is clear that F. tularensis mutants lacking many of the

genes of the FPI are profoundly attenuated for virulence, often remaining trapped

in the phagosome of host cells71,72.

The FPI genes are regulated largely by the transcription factors FevR

(Francisella effector of virulence regulator) and the Francisella homologs of the

stringent starvation proteins A and B, called MglA and MglB (macrophage growth

Page 29: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

16

locus)73,74. Mutants in these genes show attenuation in vitro and in vivo, and they

show differential expression of FPI genes and approximately 70 genes relative to

the wild type strains, supporting the idea that FPI-independent virulence factors

may exist elsewhere on the chromosome74,75. Interactions with the MglA/B-RNA

polymerase complex and FevR are stabilized by the small molecule guanosine

tetraphosphate, or ppGpp76. Production of ppGpp is essential for FevR

association with the RNA polymerase complex, as mutants that fail to produce

significant amounts of ppGpp have minimal FevR-bound RNA polymerase and

expression of FPI genes is substantially reduced. Our lab has identified mutants

that contribute to ppGpp generation and have shown that each mutant has more

or less severe intracellular growth phenotypes depending on the type of cell

being infected77,78. It is not currently known what stimulates production of ppGpp

in Francisella species, but there may be a connection to metabolic processes.

The FPI genes are also regulated by iron levels in their local environment, as

both lacZ reporter and proteomics analysis of F. tularensis LVS grown in iron

limiting media showed upregulation of FPI genes79,80.

In addition to the FPI, a major F. tularensis virulence factor is the Group 4

capsule and lipopolysaccharide (LPS) structures, which share the same

polysaccharide subunits81-84. The LPS of F. tularensis is tetraacylated rather than

penta- or hexaacylated, and the acyl chains tend to contain 16-18 carbons, rather

than 12-14 like that of other Gram negative bacteria. This atypical structure

renders F. tularensis LPS remarkably inert with respect to activation of TLR4

signaling, which is typically highly activated upon stimulation with LPS from other

Page 30: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

17

species, such as E. coli or Salmonella85-88. Not only is the LPS inert, but it

appears that virulent F. tularensis is able to suppress host signaling pathways

that are activated upon typical LPS engagement of TLR4, suggesting that

avoidance of this pathway is highly important to the pathogenesis of F.

tularensis89. The capsule and LPS prevent complement-mediated lysis and can

bind to complement to mediate uptake in human macrophages through

complement receptor 3, a process associated with inhibition of proinflammatory

signaling. In addition to this, the capsule and LPS are essential for growth in the

host cell cytoplasm, as mutants that lack these structure rapidly induce host cell

death56. These mutants are highly attenuated in vivo, yet cause significant

inflammation and tissue damage, suggesting that capsule and LPS effectively

shield the bacteria from intracellular immune surveillance systems90,91. Our lab

and others have identified multiple mutants that disrupt various aspects of LPS

and capsule biosynthesis but have yet to identify mutants deficient in only one or

the other of these structures.

Iron uptake in F. tularensis

The iron uptake systems of F. tularensis are not dedicated virulence

factors, per se, however they contribute to the ability of the pathogen to cause

disease. Interestingly, Francisella species do not have a homolog of the TonB

system that powers the active import of siderophores in other bacteria, and thus

far no candidate gene to place this function has been found in the Francisella

genome to date92. Francisella species import iron through siderophore-bound

Page 31: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

18

ferric iron via the Fsl and Fup systems, and ferrous iron via FeoB and FupA/B

with the latter fusion protein being unique to LVS93-96. The fsl operon encodes

proteins for the biosynthesis and uptake of the Francisella siderophore, which is

structurally identical to the fungal siderophore rhizoferrin95,97-99. Loss of

siderophore production or uptake does not affect intracellular growth or virulence

in vivo, indicating that other iron uptake mechanisms are able to

compensate94,100. Characterization of a spontaneous mutant of F. tularensis

Schu S4 that was highly attenuated in murine infections found that a deletion had

occurred within ORFs FTT_0918 and FTT_0919, creating a gene fusion101. The

coding sequences of these genes, now called fupA and fupB (Fe utilization

protein), also underwent a similar fusion event in the creation of LVS, with the

fusion protein mostly mediating siderophore uptake. Further analysis of the fupA

showed that it is a paralog of the siderophore receptor gene fslE, and that the

protein is located at the outer membrane, but somewhat surprisingly mediates

ferrous iron uptake in F. tularensis Schu S4; however, these iron molecules

would still need to pass through the inner membrane ferrous iron transporter

FeoB94,96,100,102,103.

In other bacterial species, the feo operon typically consists of feoAB, and

less frequently feoABC; the Francisella chromosome encodes feoA and feoB

separately and lacks feoC94,104. FeoB is a large transmembrane protein that has

an N-terminal G-protein domain and a multi-pass transmembrane C-terminal

domain105-107. In other organisms the Fe2+ import activity of FeoB requires

interaction with the small protein FeoA, though the details of how FeoA

Page 32: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

19

stimulates FeoB activity are not clear108-111. The importance of iron in infection is

reflected by the numerous ways that bacterial pathogens have evolved to obtain

iron in a host, and several works have demonstrated that feoB contributes to or is

required for full pathogenesis of Salmonella, Campylobacter, Helicobacter,

Legionella, and others112-117. Multiple studies have characterized the Fe2+ uptake

function of the F. tularensis FeoB and have linked this to its pathogenesis, though

its contribution to oxidative stress resistance has not been thoroughly

investigated94,96,118.

Nearly all life on earth requires iron for essential metabolic processes,

including bacterial pathogens, yet the utility of iron in metabolism due to its ability

to accept and donate electrons makes it a relatively dangerous molecule inside of

a cell, as it can participate in Fenton Reaction chemistry119,120. F. tularensis

virulence is connected to the ability of the organism to avoid the detrimental

effects of reactive oxygen species (ROS), such as H2O2. The bacterium couples

glutamate metabolism pathways to H2O2 neutralization, and also maintains

optimally low levels of intracellular Fe2+ such that Fenton Reaction-mediated

damage appears to be minimized121,122. When this threshold is breached,

numerous antioxidant enzymes are able to protect the organism by detoxification

of H2O2; mutants lacking enzymes like the Dyp peroxidase, superoxide

dismutases and catalase are more sensitive to H2O2, fail to inhibit host

inflammatory signaling, and can be attenuated in murine infection models123-129.

Page 33: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

20

Intracellular lifecycle

F. tularensis is a facultative intracellular pathogen that has the capacity to

gain access to the cytosol of multiple cell types including macrophages,

polymorphonuclear leukocytes, dendritic cells, lung airway epithelia, endothelia,

and hepatocytes and it is there that the bacteria acquire nutrients and proliferate

to high numbers78,130-136. The process of parasitizing the host cell cytosol begins

with uptake of the bacteria, which can differ depending on the subspecies of

bacteria used for the infection, the host cell type being infected, both in terms of

host lineage (mouse vs. human) and developmental origin (professional

phagocytes vs. epithelial cells, for example)137. Briefly, the process initiates

when the bacteria come into contact with a host cell and interact with one or

more host cell receptors, such as mannose receptor, complement receptor 3,

scavenger receptor-A, Fcγ receptor, etc138. Human monocyte-derived

macrophages take up the bacteria by an actin-dependent process termed

“looping phagocytosis” that requires intact capsule and lipopolysaccharide

carbohydrates on the outer surface of the bacteria, while epithelial cells appear to

utilize macropinocytosis for uptake of F. tularensis139,140. Phagocytosed bacteria

temporarily reside in a phagosome that acquires markers of early and late

endosomes, such as EEA-1 and LAMP-1, before the organism induces

degradation of the phagosome membrane141-144. The bacteria then have access

to the nutrient-rich cytosol where they grow until the death of the cell, whereby

the bacteria can begin the process again in a new cell145,146.

Page 34: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

21

Opsonization mediates increased uptake of the bacteria, and depending

on the opsonin can have profound influence on the fate of the bacteria in terms of

phagosome escape kinetics, growth in the host cell cytosol, and whether an

inflammatory response is stimulated or suppressed138. For example, while

unopsonized F. tularensis typically interacts with the mannose receptor to

mediate phagocytosis, serum opsonization can lead to uptake via complement

receptors138,147. The complement cascade is a complex and highly regulated part

of the innate immune system that can directly kill bacteria via lysis or target them

for receptor-mediated phagocytosis by neutrophils or macrophages, but

Francisella species are resistant to complement-mediated lysis via their LPS and

capsule polysaccharides148,149. Additionally, these structures appear to help the

bacteria to subvert this important host defense by binding and converting

complement component C3b to the inactive C3bi, which does not contribute to

formation of the lytic membrane attack complex, yet still allows for receptor-

mediated phagocytosis149. Natural IgM readily binds to F. tularensis capsule and

LPS and mediates activation of the classical complement pathway, suggesting

that the bacteria have evolved in such a way that interaction with the complement

system may increase its pathogenesis150. Indeed, Dai, et. al. found that

complement receptor 3-mediated phagocytosis of F. tularensis Schu S4 lead to

suppression of TLR2 induced proinflammatory cytokine production in human

monocyte-derived macrophages, arguing that this contributes to the stealthy

nature of F. tularensis infections151. Interestingly, they report that this

immunosuppressive phenotype was not observed when the experiments were

Page 35: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

22

repeated with murine bone marrow-derived macrophages, highlighting the

complexity of outcomes between experiment design, as well as the differences

between human and mouse responses.

Antibody-mediated phagocytosis also leads to considerably more uptake

of the bacteria in murine macrophages; however, the growth of the organisms is

restricted in a NADPH oxidase-dependent manner138. Regardless of the route of

entry into the cell, uptake in vitro is surprisingly low when compared to the very

small infectious dose of approximately 10 CFU via the respiratory route,

suggesting that mechanisms of uptake in the host environment may be

considerably different, or that the bacteria may persist and possibly grow

extracellularly before encountering sufficient numbers of host cells to mediate the

relatively rapid dissemination and high organ burdens observed in mouse models

of tularemia.

Francisella-containing phagosomes initially progress from having

characteristics of early to late endosomes, but the bacteria somehow inhibit

phagosome fusion to lysosomes and avoid damage by the protease cathepsin D

or the acidic nature associated with these vesicles141,143,144,152. Once inside the

phagosome F. tularensis avoids the destructive activity of the respiratory burst of

neutrophils and macrophages. The integral membrane proteins gp91phox and

p22phox come together with the soluble subunits gp47phox, p67phox and p40phox at

the phagosome membrane to form the NADPH oxidase153. This complex is

responsible for the production of high levels of toxic reactive oxygen species,

such as the superoxide anion, hydrogen peroxide, and hypochlorous acid (the

Page 36: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

23

latter being specific to neutrophils)154. These molecules severely damage or kill

ingested microbes, though F. tularensis has evolved as yet undefined

mechanisms for preventing the NADPH oxidase from assembling or functioning

appropriately135,155. Neutrophils infected with F. tularensis do not show

colocalization of internalized bacteria and components of the NADPH oxidase nor

do the infected cells generate a respiratory burst; in fact, infected cells can

prevent this response when treated with compounds that typically stimulate

robust respiratory bursts135,156. Indeed, mice lacking genes coding the gp91phox

subunit of the NADPH oxidase did not exhibit dramatic differences in survival or

organ burdens when infected with F. tularensis Schu S4, suggesting that reactive

oxygen species generated by the NADPH oxidase do not significantly contribute

to controlling infection in wild type mice157. Genetic screening for mutants that

fail to inhibit the burst identified mutants that were defective in various aspects of

uracil metabolism; how these mutants stimulate a burst is not clear, though it may

depend in part on expression levels of genes from the Francisella Pathogenicity

Island158. That the bacteria avoid the respiratory burst strongly argues that a

major mechanism of pathogenesis for F. tularensis is to avoid the toxic effects of

host-induced reactive oxygen species.

Escape from the phagosome occurs shortly after phagocytosis, typically

within hours depending on the host species, Francisella strain, and the presence

or absence of opsonins141-143,159. Acidification of the phagosome via the vacuolar

ATPase has been shown to occur in vitro when unopsonized bacteria infect

murine macrophages; this acidification was important for egress from the

Page 37: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

24

phagosome141. These results are in contrast with those of Clemens, et. al. who

found that opsonized F. tularensis do not induce acidification of the phagosome

in human macrophages, nor do they require this process for escape143,160.

Interestingly, prevention of the acidification of the phagosome has also been

reported to make F. tularensis LVS less able to acquire iron in the phagosome161.

As mentioned previously, the disparities between reports highlights the fact that

interpretation of results of these types of experiments cannot be generalized, as

differing conditions used to examine F. tularensis intracellular growth in vitro lead

to multiple outcomes, and it is currently not known if one or more experimental

paradigms better recapitulate the complexity of the conditions encountered in

vivo.

Degradation of the phagosome occurs by an unknown mechanism;

however, it is clear that many of the genes of the Francisella Pathogenicity Island

are required for this process, as certain mutants remain trapped in the

phagosome75,162. Once in the cytoplasm the bacteria begin to acquire nutrients

and replicate significantly faster than is observed in rich laboratory media, with F.

tularensis Schu S4 doubling in population size in approximately one hour141.

Being an auxotroph for numerous amino acids, F. tularensis must obtain them

from the host cytoplasm. One mechanism by which the bacteria do this appears

to be through the upregulation of host amino acid importers to increase the

cytoplasmic amino acid concentration. Barel et. al. found that infection of an

immortalized macrophage cell line with F. tularensis LVS lead to upregulation of

the host SLC1A5 amino acid importer, and that siRNA knockdown of its transcript

Page 38: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

25

restricted LVS growth in the cytosol163. This group and others have also

identified a number of bacterial factors that mediate uptake of various amino

acids that are essential for growth in the cytoplasm of host cells, consistent with

the idea that evolution towards the intracellular parasitism of F. tularensis

relieved selective pressure for the maintenance of biosynthetic pathways to

produce these molecules, as the bacteria readily obtain them from their host164-

166. Scavenging amino acids and other nutrients from the host also has the

additional survival advantage of providing some of the end products of metabolic

pathways that are inactivated by H2O2 via oxidation of iron-sulfur cluster-

containing enzymes167.

F. tularensis burden in the cytosol of infected host cells can reach high

numbers, as microscopy of in vitro infection shows cells that appear to be entirely

filled with bacteria56,78,168. Presumably, the bacteria exhaust the host cell of its

nutrients, which leads to its death145,146. As with phagosome acidification and

escape, the type of cell death that an infected cell undergoes appears to be

specific to each Francisella species, as well as the species of the host cell being

used for infection. For example, F. novicida and F. tularensis LVS stimulates

pyroptotic cell death in an AIM2 and caspase-1-dependent fashion and have

been fruitful as tools to explore the molecular mechanisms underlying the

function of this host surveillance system, but this mechanism does not appear to

be involved in cell death induced by the virulent species169-172. Rather, apoptotic

cell death pathways induced by subsp. tularensis strains appear to be mediated

by caspase-3 in vivo173,174. Regardless of the pathway responsible, F. tularensis-

Page 39: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

26

infected cells die and facilitate the further infection of adjacent or newly arrived

cells.

Immunity to Francisella species

One of the key features of the murine pneumonic tularemia model is the

rapid time to death (~5-7 days after infection), which precludes the development

of a robust adaptive immune response. This aspect of Francisella virulence has

made vaccine development a daunting challenge, and many labs have used

Francisella tularensis subsp. holarctica LVS as a less virulent substitute to study

aspects of B and T cell biology in sub-lethal infection and immunization, though it

is clear that the host responds to Francisella in a strain-specific fashion175,176. A

vaccine against fully virulent Francisella strains will almost certainly require

stimulation of cellular immunity, as studies of individuals who have recovered

from tularemia indicates a clear Francisella-specific T cell response, even years

after infection177-180.

Protective antibodies were an early target of research into immunity to

Francisella. In early studies utilizing a Type A strain, Foshay, et. al. showed that

transfer of hyperimmune serum from horses or goats provided protection to ~70-

90% of infected rats, although recent studies utilizing the mouse model have

found contradictory results181,182. Kirimanjeswara, et. al. found that murine

alveolar macrophages stimulated with interferon-γ were able to kill Schu S4 that

had been opsonized with serum from LVS-immunized mice, but that the immune

serum did not provide protection against Schu S4 during infection in vivo182.

Page 40: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

27

While the latter is intriguing, Crane, et. al. have shown that Schu S4 may be able

to avoid this fate by interacting with the host serine protease plasmin. Schu S4

can bind to active plasmin, which can degrade Francisella-specific antibodies183.

The authors found that while antibody opsonization of both Schu S4 and LVS

resulted in an increase in TNF-α, the presence of active plasmin on Schu S4, but

not LVS, inhibited the induction of TNF-α in infected cells183. These results

highlight a difference between the two strains that may have important

implications for how virulent Francisella species avoid early detection and

control, and highlight significant pathogenesis mechanisms that will need to be

accounted for when designing a next generation vaccine against F. tularensis.

As with antibodies, the role of B cell activity in providing immunity to

Francisella species is complex; however, it is clear that mice lacking B cells are

less able to control or survive a Francisella infection184. Data is emerging that a

specific subset of B cells have divergent roles depending on the infecting strain

and the model of infection185,186. Immunization with lipopolysaccharide from LVS

generated a B1a cell-dependent protective response against an intraperitoneal

challenge with approximately 103 CFU of LVS185. In contrast, Crane, et. al.

utilized a short-term low dose antibiotic treatment after intranasal infection with

Schu S4 (the convalescent model) and observed that mice largely deficient in

B1a cells (XID mice) survived better than did wild type mice186. The increased

survival in these mice was found to be associated with a reduction in the anti-

inflammatory cytokine IL-10, which is a potent inhibitor of IL-12. The latter

cytokine stimulates interferon-γ production, and is necessary for the survival of

Page 41: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

28

tularemia184. Curiously, IL-10 -/- mice succumb to standard intranasal Schu S4

infection similar to wild type mice, though it should be noted that the time scales

of infection are different between these studies and direct comparisons may not

be appropriate187. The immune responses observed in the convalescent model

would not have time to develop in a standard intranasal infection, although it

would be of interest to see if IL-10 -/- mice phenocopy XID mice in the

convalescent model of Schu S4 infection.

One possible target of the anti-inflammatory activity of IL-10 may be at the

interface of antigen presenting cells and T lymphocytes. Hunt, et. al., identified a

factor released by Francisella infected cells that stimulated IL-10-dependent

degradation of MHC class II molecules in macrophages188. Importantly,

supernatants from Schu S4 infected macrophages also stimulated the

downregulation of MHC class II molecules189. These data suggest that antigen

presentation to CD4+ T cells may be reduced in vivo. The same research group

and others have also previously found that F. novicida and LVS induce

prostaglandin E2 (PGE2) production in infected macrophages189,190. PGE2 has

been shown to inhibit macrophage maturation, and might play a similar role in

downregulating pathways important for intracellular killing of Francisella, as in

Burkholderia pseudomallei infections191,192. Interestingly, they showed that PGE2

induced by Francisella infection inhibited T cell proliferation and interferon-γ

production in vitro. It has long been known that T cells are important for bacterial

control and immunity to Francisella179,184,193. Both CD4+ and CD8+ T cells were

required for survival of a primary Schu S4 infection in the convalescent model of

Page 42: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

29

tularemia, but only partial protection against a secondary challenge was

observed for wild type mice (66% survival)184. It is intriguing to speculate that the

partial protection may be a function of a less than optimal T cell response

mediated by PGE2, both in terms of antigen presentation and T cell proliferation.

Considerable effort has been expended testing potential vaccines and

vaccine strains against infection with F. tularensis, yet as with the disparities

between in vitro infection outcomes, these studies all differ depending on the

details of experimental setup. The differences in protocols include route of

inoculation (intranasal, subcutaneous, or intradermal), immunizing agent (live

versus dead organisms, membrane fractions, purified LPS/capsule, passive

transfer of serum or antibodies), challenge strain (LVS or fully virulent F.

tularensis), and model system (mouse, rat, rabbit, etc.)194. As such, comparisons

between these studies are difficult to interpret. Thus, the major goal of the work

presented in this thesis was to develop a foundation for future understanding of

what makes F. tularensis LVS the “gold standard” live vaccine strain using the

tools of bacterial genetics.

Page 43: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

30

CHAPTER II

A VIRULENT BIOVAR OF F. TULARENSIS LVS IS INTRINSICALLY MORE

RESISTANT TO HYDROGEN PEROXIDE

Introduction

Prior to initiating my thesis work I spent considerable time developing and

testing a low and moderate dose immunization experiment with two attenuated

capsule/O-antigen mutants, waaY and wbtA, that had been identified in

transposon screens from my lab56,91. In this experiment (Appendix A) I included

LVS as a positive control for protection against intranasal challenge against F.

tularensis Schu S4, and found that LVS significantly outperformed waaY and

wbtA, despite the latter strains’ attenuation and stimulation of an obvious host

response (ruffled fur, hunched shoulders, conjunctivitis, etc.). Concurrent with

this experiment, our collaborator at Rocky Mountain Laboratories, Catharine

Bosio, shared data indicating that two LVS stocks in her possession differed in

virulence and ability to provide protection against Schu S4. She graciously

shared these strains and their genome sequences, so I began to explore the

phenotypic differences between these and my lab strain in an effort to

understand the genetic requirements of LVS to cause disease and stimulate an

immune response in a mouse.

Despite significant differences in virulence, members of the Francisella

genus are very similar at the genetic level; many of the observed differences are

genomic rearrangements and single nucleotide polymorphisms (SNPs)41,50. The

similarities across the Francisella strains has made identification of the specific

Page 44: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

31

factors that mediate the high level of virulence displayed by Type A and Type B

strains difficult, although genome comparisons between F. tularensis subsp.

tularensis Schu S4 and less virulent LVS have identified some of the attenuating

mutations. These include fusion of the fupA/B genes and loss of pilA102. Another

area of significant difference between the highly virulent Type A strains and the

intermediately virulent type B strains (and LVS) is the level of intracellular iron,

with virulence being inversely correlated with iron levels122. Iron is an essential

micronutrient, and numerous studies have shown that it is highly sought after by

bacterial pathogens, as host sequestration of iron (a part of nutritional immunity)

can restrict the growth of numerous pathogens113,195. Apparently, F. tularensis

may represent an exception to this paradigm as Lindgren, et al., found that

higher virulence subspecies had low levels of intracellular iron122.

Phenotypic variability exists between different LVS biovars in terms of

virulence and vaccine efficacy51,196. These differences provide a naturally

occurring genetic experiment to identify bacterial factors that correlate with

vaccine efficacy. To explore this, the genomes of two LVS biovars were

sequenced (Bosio lab), previously labeled RML (higher virulence; provides better

protection against Schu S4 challenge) and ATCC (lower virulence; poor

protection against Schu S4 challenge) and I sought to characterize the genetic

differences here. Interestingly, two mutations occurred in genes known to be

involved either in iron uptake (FTL_0133, feoB) or oxidative stress resistance

(FTL_1773, dyp peroxidase). FeoB is an inner membrane protein that imports

ferrous iron into the bacterial cytoplasm, and has previously been linked to

Page 45: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

32

virulence as one of two major iron uptake pathways in LVS94,96,197. Dyp is a

member of a broad class of dye-decolorizing peroxidases with wide substrate

specificity and important industrial uses198. In agreement with the data reported

in this thesis, Binesse, et. al. characterized the LVS dyp allele with a 93 base pair

deletion as contributing to sensitivity to H2O2, and found that complementation

with a full length dyp (found in Schu S4 and some LVS lots) restored H2O2

resistance51,127. These data suggest that resistance to host-induced oxidative

stress is a significant component of the virulence of F. tularensis, and may

contribute to the vaccine efficacy of LVS given that the ATCC biovar of LVS has

the 93 base pair deletion and was relatively attenuated in murine infections and

stimulated weak immunity against a challenge of F. tularensis Schu S4196.

Materials and Methods

Bacterial strains and growth conditions: Three different biovars of F.

tularensis subsp. holarctica LVS were used in this work - the LVS strain used in

the Jones lab for several years, here called Iowa LVS (University of Iowa) and

the RML LVS and ATCC LVS, (Rocky Mountain Laboratories). Bacteria were

routinely cultured on modified Mueller-Hinton (supplemented with 1% glucose,

0.025% ferric pyrophosphate, and 2% IsoVitaleX) agar or in broth, with 50 μg/mL

of kanamycin or spectinomycin, as needed. Mueller-Hinton agar was also

prepared without ferric pyrophosphate supplementation to assay for growth in

lower iron conditions. Bacteria were also cultured in Chamberlain’s defined

medium with either 35 μM FeSO4 or 350 nM FeSO4, supplemented with

Page 46: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

33

antibiotics as needed. Agar plates were incubated at 37°C with humidity and 5%

CO2 while broth cultures were grown at 37°C, shaken at 200 rpm.

Mutant construction and complementation: Deletion of feoB (FTL_0133) and

dyp (FTL_1773) gene was achieved by homologous recombination using

derivatives of the non-replicating plasmid pJC84. Primer sequences are shown

in Table 1. Briefly, upstream and downstream flanking DNA was amplified via

PCR, and amplicons were cloned into the multiple cloning site of pCR2.1 (Life

Technologies). A spectinomycin resistance cassette was cloned into the AvrII

site in the 3’ end of each upstream PCR fragment. The upstream-spectinomycin

resistance fragment was removed by digestion with AscI, and cloned into the

AscI site in the 5’ region of the downstream PCR plasmid. The entire upstream-

spectinomycin resistance-downstream fragment was cloned into the BamHI site

of the suicide plasmid pJC84. Finally, the spectinomycin resistance cassette was

removed by digestion with AvrII and the plasmid was re-ligated. Plasmids were

electroporated into LVS (2.5 kV, 25 μF, and 600 Ω), and the bacteria were plated

onto MMH agar with 50 μg/mL kanamycin after 2-3 hours of outgrowth.

Organisms were then grown overnight in broth lacking kanamycin, and were

plated onto MMH agar with 8% sucrose for sacB-mediated counter-selection.

Kanamycin sensitive colonies were screened by colony PCR to detect deletion of

the feoB or dyp gene. Primers were designed such that they flanked the coding

sequence of each gene such that an amplicon would be produced regardless of

genotype. For complementation of each mutant, the coding sequence of each

Page 47: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

34

allele of feoB or dyp was PCR amplified using the Phusion high fidelity

polymerase. Primers were designed such that 5’ KpnI and 3’ SalI sites were

included for cloning purposes. The PCR amplicons were purified and A-tailed

with Taq polymerase (New England Biolabs) per the manufacturer’s instructions

and cloned into pCR2.1. Inserts were removed by digestion with KpnI and SalI,

and cloned into the respective sites downstream of the groE promoter in

pTrc99a; the groE promoter has a 5’ BamHI site. The entire PgroE-coding

sequence was moved to the BamHI and SalI sites of pBB103 to create the final

plasmids. The waaY::Trgtn mutants were constructed as described previously56.

Iron-regulated β-galactosidase reporter activity: To assess iron uptake-

regulated gene expression, Miller assays were performed with Iowa, RML, and

ATCC LVS carrying the fslA promoter-lacZ fusion on the pBB103 plasmid77.

Bacteria were grown in Chamberlain’s defined medium (supplemented with 50

μg/mL spectinomycin) to late-log phase, and β-galactosidase activity was

assayed using the standard Miller assay199.

FeoB function in a heterologous reporter system: The coding sequence of

feoB was PCR-amplified from both Iowa and RML LVS using the high fidelity

Phusion polymerase (New England Biolabs). Sanger sequencing was performed

to confirm that the SNP observed in the RML LVS background was present. The

feoA coding sequence was cloned into the KpnI/SalI sites of a derivative of

pTrc99a, immediately downstream of the groE promoter. The entire PgroE-feoA

Page 48: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

35

fragment was removed by digestion with BamHI and SalI, and ligated into the

same sites in pBB103. Both this plasmid and the pWKS30 containing feoB were

transformed into the E. coli H1771 strain (MC4100 aroB feoB7 fhuF::plac Mu).

Control strains were also generated that carried only one of the plasmids. Miller

assays were performed to measure FeoB Fe2+ import activity via the readout of

Fur repression of fhuF::placZ. All strains were grown in LB supplemented with 50

μg/mL of spectinomycin and 100 μg/mL of ampicillin as necessary.

Hydrogen peroxide sensitivity: To measure resistance to H2O2-mediated

killing, mid- to late-log phase organisms were pelleted at 13,200 RPM for 5 min,

washed in PBS, and ~106 CFU were resuspended into 200 μL PBS with or

without 100 μM fresh hydrogen peroxide (H2O2) in a 96-well dish. The samples

were incubated at 37°C with humidity and 5% CO2 for thirty minutes or one hour.

The culture from each well was then serially diluted, plated onto modified

Mueller-Hinton agar (with antibiotic where necessary) and the number of

surviving organisms for each strain was enumerated after 2-3 days.

In vitro infections: Murine bone marrow-derived macrophages (BMM) were

generated by harvesting bone marrow from C57BL/6 mice, 6-10 weeks old. The

bone marrow was differentiated in DMEM supplemented with 10% heat-

inactivated fetal bovine serum (FBS), 10% L929 cell-conditioned DMEM and Pen

Strep (Gibco) for 5-6 days. Subsequently, non-adherent cells were removed by

PBS wash, and adherent cells were lifted by a brief incubation with Versene

Page 49: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

36

(Gibco) for ~5-6 minutes at 37°C. Cells were enumerated and seeded into 24

well dishes at a density of 3x105 per mL in DMEM with 10% L929-conditioned

media, 10% heat-inactivated FBS. Multiplicity of infection (MOI) was estimated

by measuring the OD600 value of mid to late-log phase grown bacteria, and

confirmed via serial dilution onto modified Mueller-Hinton agar and enumeration

after 2-3 days growth at 37°C with humidity, 5% CO2. Bacteria were allowed to

associate with the BMMS for 3 hours, after which time the cells were washed 3x

with PBS, and incubated in 10% L929-conditioned DMEM supplemented with

100 μg/mL of gentamicin for one hour, cells were then washed 3x with PBS and

either lysed or incubated in antibiotic-free media for a further 20 hours. At the 4

and 24-hour time points 0.1% saponin was added to each well. Wells were

scraped and vigorously pipetted to disrupt the cells and liberate intracellular

bacteria for serial dilution and enumeration. Infection of A549 cells was

performed identically as BMM infections, however the cells were maintained in

DMEM with 10% heat-inactivated FBS with Pen Strep as needed, and cells were

lysed with saponin at the four-hour time point rather than at three hours.

Results

LVS biovars have similar PiglA-lacZ expression and lack low molecular weight O-

antigen glycosylated proteins

Prior to having access to the genome sequences of RML LVS and

ATCC LVS I investigated two phenotypes putatively connected to virulence that

our lab had identified as differing between virulent strains and LVS: FPI gene

Page 50: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

37

reporter expression and the presence of protein species glycosylated with O-

antigen sugars. The Francisella Pathogenicity Island (FPI) encodes numerous

genes essential for the pathogenesis of Francisella, and as such the genes at

this locus are known to be highly expressed. Previous data from our laboratory

has shown that a PiglA-lacZ reporter has different activity levels between

subspecies, with expression in the Type A strain F. tularensis subsp. tularensis

Schu S4 being about two-fold higher than the Type B F. tularensis subsp.

holartica. The LVS exhibited the lowest level expression, approximately one-

third that of Schu S4 expression. While we do not have evidence that these iglA

expression level differences mediate the relative virulence of each strain, it

remains an intriguing observation that virulent strains have higher expression of

fundamental virulence factor genes. To test the hypothesis that the more virulent

RML LVS biovar had higher expression of a FPI gene relative to two less virulent

biovars, I introduced the PiglA-lacZ reporter plasmid to each strain and performed

Miller assays after overnight growth. As seen in Figure II.1A, no significant

difference was observed between the strains under these conditions, suggesting

that the difference in virulence between them is not likely due to differential

regulation of the igl operon.

Additionally, it was previously shown by our lab that the virulent

subspecies of F. tularensis produce low molecular weight O-antigen positive,

proteinase K sensitive protein species while the Iowa LVS does not81. To test the

hypothesis that the more virulent RML LVS biovar produce such species, I

constructed a waaY::TrgTn mutant in both RML LVS and ATCC LVS; an identical

Page 51: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

38

mutant in the Iowa LVS background was constructed previously. Each sample

was prepared with or without overnight treatment with proteinase K, separated by

SDS-PAGE, and blotted with an antibody that recognizes F. tularensis O-antigen

sugars (FB11). As shown in Figure II.1B, the positive control F. tularensis Schu

S4 waaY::trgTn sample has several low molecular weight bands that are not

present when the sample is treated overnight with proteinase K. Like the Iowa

and ATCC LVS (latter strain not shown), the RML LVS sample did not have

these bands irrespective of treatment, suggesting that the higher virulence of this

strain is not likely due to the presence of O-antigen glycosylated proteins.

RML LVS has less intracellular Fe2+ than the ATCC or Iowa LVS

Genome sequences of the RML and ATCC LVS indicated a

nonsynonymous substitution in the RML LVS feoB allele that changed an

aspartate at residue 471 to a tyrosine (Fig. II.2). The feoB D471Y allele

represented an interesting target to identify the virulence difference between the

RML and the ATCC strain due to the well characterized nature of FeoB function

in Fe2+ uptake and the connections between iron content and virulence. The

D471Y mutation maps to a cytoplasmic loop, adjacent to a highly conserved

glycine residue that is predicted to be functionally important by ConSurf200,201.

The substitution of a large, hydrophobic tyrosine for an aspartate at residue 471

led us to hypothesize that the FeoB encoded by the RML allele imports Fe2+

poorly, or not at all. Although the genome of the lab stock of LVS from the Jones

lab (hereafter referred to as “Iowa”) has not been completely sequenced, the

Page 52: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

39

strain was also included in the experiments described here. Sanger sequencing

confirmed that the Iowa feoB encodes an aspartate at residue 471 and a full

length dyp peroxidase, suggesting that it is potentially an intermediate between

the RML and ATCC strains.

We tested the hypothesis that the RML LVS has less intracellular iron than

either the Iowa LVS or the ATCC LVS by growing the strains overnight in

standard modified Mueller-Hinton (MMH) broth, serially diluting in PBS, and

spotting ten-fold dilutions onto MMH agar of varying concentrations of iron (Fig.

II.3A). Growth was identical among each strain when spotted onto the control

agar containing the typical concentration of iron (0.0025% ferric pyrophosphate)

routinely used for propagation of F. tularensis. When the iron concentration was

reduced by 50%, both the Iowa and ATCC strains had growth patterns similar to

that observed on the control plate, however, the RML strain exhibited growth

restriction at this concentration of iron. Fewer isolated colonies were observed

and lawn growth was less luxurious when compared to the Iowa and ATCC LVS.

On MMH agar lacking added iron, all three strains exhibited growth restriction,

however the reduced growth phenotype of RML was exacerbated, with extremely

poor growth even at the lowest dilution plated. Iowa and ATCC strains both grew

as lawns at these dilutions, indicating that they were able to scavenge sufficient

iron from the agar plate environment, while the RML strain could not. The

decrease in the RML strain growth was approximately two orders of magnitude

greater than that observed for either the Iowa or ATCC LVS, consistent with the

hypothesis that the RML strain has less intracellular iron under conditions of iron

Page 53: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

40

limitation. In support of this, disc-diffusion assays on MMH agar lacking added

iron showed that the RML LVS grew smaller halos around discs soaked with

ferric pyrophosphate solutions of varying concentrations (Fig. II.3B).

As the RML strain grew less well under iron limiting conditions and the

feoB gene encodes a ferrous (Fe2+) iron importer, we hypothesized that the RML

strain would have less intracellular Fe2+. Levels of intracellular Fe2+ are tightly

regulated, as free iron can participate in the toxic Fenton reaction with H2O2,

causing cellular damage in the forms of lipid peroxidation, DNA damage, and

most importantly, poisoning of the iron-sulfur cluster enzymes of essential

metabolic pathways. To avoid this toxicity, bacteria have evolved transcriptional

regulatory mechanisms to maintain iron homeostasis. One such example is the

Fur system202. The Fur transcriptional repressor is an allosteric regulator that

uses Fe2+ as a co-repressor; when iron levels are sufficient, the Fur-Fe2+

complex binds to Fur Box sequence motifs in the promoters of iron uptake genes

to mediate their repression. When iron is limiting, less Fe2+ is bound to Fur,

decreasing its ability to bind DNA. This leads to de-repression of iron uptake

genes; the Fur regulatory system has been described to function similarly in

Francisella95,97,98. To assess F. tularensis LVS intracellular Fe2+ levels under iron

limiting conditions, a Fur regulated PfslA-lacZ construct was introduced into the

RML, Iowa, and ATCC strains. Iron limitation is known to relieve Fur repression

at the fslA promoter, so increasing gene expression correlates with decreasing

concentrations of intracellular Fe2+77. Each bacterial strain was grown in

Chamberlain’s Defined Medium (CDM) with 35 μM (high iron) or 350 nM (low

Page 54: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

41

iron) FeSO4, and β-galactosidase activity was measured after overnight growth

(Fig. II.4). As expected, iron starvation induced high expression of PfslA-lacZ in all

three strains; however, induction in the RML LVS was approximately 2-fold

higher than that observed for Iowa LVS or ATCC LVS. The higher activity of the

PfslA-lacZ construct in the RML strain is consistent with the hypothesis that this

strain has lower levels of intracellular Fe2+ when iron is limiting in the growth

medium.

To assess iron levels in the RML LVS by an independent method, we

genetically manipulated the ability of each strain to acquire iron by

overexpression of Fur. The F. tularensis fur gene was placed under the control

of the groE promoter, and the construct introduced into each strain. Bacteria

were grown in MMH broth with normal iron concentrations overnight, serially

diluted in PBS and plated onto standard MMH agar (Fig. II.5). The Iowa and

ATCC strains, when overexpressing Fur, exhibited colony growth similar to that

of their vector controls, with no obvious decrease in the size of isolated colonies

or robustness of lawn growth at lower dilutions. The RML LVS, however, had

much smaller colony size and poor lawn formation, indicating a general growth

delay that was not observed in the RML vector control strain. We interpret these

results to indicate that Fur overexpression in RML LVS had a greater impact on

growth because the strain has significantly smaller pools of intracellular iron and

therefore inhibition of iron uptake systems has a greater impact on RML LVS

growth and survival than for the Iowa LVS or ATCC LVS, even though all strains

were grown in iron-rich media. This is consistent with the hypothesis that

Page 55: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

42

intracellular Fe2+ pool of RML LVS are smaller than in Iowa LVS or ATCC LVS

and become limiting for growth in conditions that are not limiting for the other two

strains.

FeoB D471Y does not complement E. coli ΔfeoB fhuF::λplac

The low iron growth phenotypes and Fe2+-regulated gene expression of

RML LVS strongly suggest that the FeoB D471Y protein encoded by the strain

has significantly reduced Fe2+ import activity. To assess the ability of the FeoB

D471Y protein to transport iron, we made use of a well-characterized E. coli iron

reporter strain that lacks feoB (Fig. II.6)111,203. This strain, E. coli H1771, has a

chromosomal lacZ insertion in the Fur-regulated gene fhuF, and has high β-

galactosidase activity as a result of low Fe2+ levels. The feoB alleles from Iowa

and RML (D471Y) were introduced into H1771 on the low copy number plasmid

pWKS30, and Miller assays were performed. Initial experiments supplying only

feoB (either allele) showed no repression of fhuF::lacZ, indicating that FeoB

alone is not sufficient for Fe2+ import in this reporter system (data not shown). In

most bacterial species encoding a feo system, the small gene feoA is encoded

either in an operon with feoB or elsewhere on the chromosome and the encoded

protein is required to stimulate FeoB Fe2+ import activity104,109,111. When the

Francisella feoA was supplied on pBB103 in concert with feoB (Iowa/ATCC

allele) on pWKS30, significant repression of the fhuF-lacZ reporter was observed

(~ 80% reduction in expression relative to the empty vector control), indicating

that Fe2+ import was significantly increased. In contrast, complementation of

Page 56: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

43

H1771 with feoA and feoB D471Y (RML allele) failed to repress fhuF::lacZ. This

result indicates that the FeoB D471Y protein failed to significantly import Fe2+

and further supports the hypothesis that RML has less intracellular Fe2+ under

conditions of iron limitation in vitro. I included the Schu S4 feoB allele in this

analysis as it differs from the annotated LVS allele by four amino acids, however

significant reduction in fhuF::lacZ reporter activity was observed, indicating that it

is competent to import ferrous iron.

Resistance to H2O2 correlates with feoB D471Y allele

Since the RML LVS had less intracellular Fe2+ and Lindgren et. al.

demonstrated that strains with lower intracellular iron concentrations were more

resistant to H2O2, we hypothesized that RML LVS would be more resistant to

killing by H2O2 than Iowa LVS or ATCC LVS. Bacteria were exposed to PBS

alone or PBS with 100 μM H2O2 in a 96-well dish for one hour at 37°C with 5%

CO2 and humidity. Following incubation, the bacteria were serially diluted in PBS

and plated onto MMH agar (Fig. II.7). The RML strain was approximately ten-fold

more resistant to H2O2 than the Iowa strain, and approximately one-hundred-fold

more resistant than ATCC. The genome of ATCC encodes a Dyp peroxidase

with a 93 base pair deletion; the contribution of Dyp function to H2O2 resistance is

described in a later section.

Given the increased sensitivity of Iowa LVS to H2O2, and the functional

complementation of an E. coli feoB mutant by the Iowa feoB allele, we next

tested if the RML LVS could be sensitized to H2O2 by increasing the intracellular

Page 57: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

44

Fe2+ pool via overexpression of a functional feoB (Fig. II.8). This was achieved

by transforming the strain with a plasmid carrying the overexpression constructs

Pgro-feoB (Iowa/ATCC allele - high Fe2+ transport) and Pgro-feoB (D471Y; RML

allele - low Fe2+ transport). H2O2 sensitivity assays were performed as described

previously, however, a 30-minute time point was also included in case

overexpression lead to increased susceptibility such that no viable organisms

would be recovered at one hour. Overexpression of either feoB allele led to

increased sensitivity to H2O2; however, the effect mediated by the Iowa/ATCC

feoB was orders of magnitude greater than that of the poorly functional RML feoB

D471Y allele. Nearly one log of killing was observed at 30 minutes for the RML +

Pgro-feoB (Iowa/ATCC), while RML + Pgro-feoB (D471Y) only had a two-fold

reduction in viability. The effect was more pronounced at the one-hour time

point, with no viable bacteria recovered at the level of detection (102 CFU) from

RML + Pgro-feoB (Iowa/ATCC). In contrast, over 104 CFU were recovered at one

hour from the RML + Pgro-feoB (D471Y). These data demonstrate that the SNP

in feoB encoded in the RML LVS genome mediates increased resistance to

H2O2.

Although there were few differences between the genomes of RML LVS

and ATCC LVS, it was possible that the H2O2 sensitivity of RML LVS

overexpressing the Iowa/ATCC feoB allele may have been due to genetic

interactions between feoB and SNPs unique to the RML LVS and not feoB alone.

To confirm that the H2O2 resistance phenotype conferred by feoB D471Y is not

unique to the RML genetic background, I constructed a ΔfeoB mutant in the Iowa

Page 58: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

45

LVS background and complemented it with the Pgro-feoB constructs (Fig. II.9).

While the genome of this strain is not sequenced, the Iowa LVS was chosen over

the ATCC LVS, as the latter strain was approximately 100 fold more sensitive to

H2O2 (see below). H2O2 sensitivity assays were performed as before and,

consistent with results in the RML strain background, overexpression of feoB

D471Y mediated significant resistance to H2O2 relative to that when wild type

feoB was overexpressed. This result strongly supports the hypothesis that the

feoB allele in the RML LVS genome encodes a FeoB protein with minimal ferrous

iron import and that this mediates higher resistance to the lethal effects of H2O2.

Resistance to H2O2 also requires Dyp peroxidase

The ATCC LVS was observed to be approximately two-to-three orders of

magnitude more sensitive to the killing effects of H2O2 than the RML strain. The

ATCC LVS genome, in addition to encoding a feoB that is competent for robust

Fe2+ import, encodes a dyp-type heme peroxidase that is missing nucleotides

737 to 830 relative to the first nucleotide of the coding sequence. This result in a

protein that lacks 31 amino acids near the C-terminus, and bioinformatic analysis

indicates that this deleted region encodes a conserved phenylalanine residue

that is part of the heme-binding pocket, a critical functional component for Dyp

enzymatic activity in other organisms198. Dyp peroxidases are known for having

a wide range of substrates, so ascertaining the natural function of the enzyme in

vivo has been difficult, though as a peroxidase Dyp uses H2O2 as a cofactor in its

reactions.

Page 59: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

46

To assess the contribution of Dyp to H2O2 resistance, I constructed a ∆dyp

deletion mutant in the Iowa LVS and performed H2O2 sensitivity assays as

described in the materials and methods (Fig. II.10). The ∆dyp mutant was

significantly more sensitive relative to the parental Iowa strain, indicating that the

Dyp enzyme provides substantial protection against the killing activity of H2O2.

This indicates that the 93 base pair deletion observed in the ATCC LVS dyp

allele may disrupt its function in this capacity, rendering it highly sensitive to

H2O2. To confirm that the loss of these 93 base pairs is what abrogates Dyp

function in the ATCC LVS, I generated two plasmid-borne complementation

constructs with each dyp allele being expressed from the groE promoter. H2O2

sensitivity assays were performed as before and the ∆dyp mutant that had been

complemented with the full length allele exhibited both a reduction in overall

viability and an increase in sensitivity to H2O2, while the strain complemented

with the 93 base pair deletion allele exhibited modest partial complementation

and no obvious defect in overall viability. While surprising at first, this may be

consistent with the identity of Dyp as a heme-binding protein. Overexpression

may result in a concentration of Dyp protein that far exceeds its normal levels; in

fact, a Pdyp-lacZ reporter exhibits negligible β-galactosidase activity (data not

shown), suggesting that the gene is expressed at low levels. In this scenario the

full length protein may be binding and sequestering a larger percentage of the

cell’s available heme such that other enzymes requiring heme, such as the

catalase KatG, lack this essential cofactor and are thus catalytically inert. The

truncated protein encoded by the allele with the 93 base pair deletion is predicted

Page 60: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

47

to lose a conserved residue in the heme-binding pocket and may bind negligible

quantities of heme when expressed naturally in the ATCC LVS, leading to an

inactive Dyp protein and H2O2 sensitivity in this strain. Alternatively,

overexpression of wild type Dyp may lead to levels of the enzyme that exceed its

natural substrate(s), and it may erroneously oxidize/reduce molecules important

for oxidative stress resistance.

Neither feoB nor dyp are essential for intracellular growth in A549 or BMM cells

After establishing the importance of both FeoB D471Y and Dyp in

resistance to the lethal effects of H2O2 I examined their contributions toward

intracellular growth in two cell types, the human adenocarcinoma A549 cell line

and primary murine bone-marrow-derived macrophages. The former cell line

was chosen for in vitro infection because a feoB mutant in the ATCC LVS lineage

was reported to exhibit poor intracellular growth over twenty-four hours, and the

latter cell type was chosen as growth within macrophages is a preferred niche for

F. tularensis96. No significant difference in growth in either cell type was

observed for the three LVS biovars, approximately 100-fold for A549 cells and

approximately 10-fold for BMMs (Fig. II.11 and Fig. II.13). These data suggest

that the genetic differences between these strains do not detectably alter

intracellular growth kinetics.

As it had already been established that the ATCC LVS feoB mutant

exhibited a growth defect in A549 cells, I generated clean deletion mutants in all

three parental strains and performed in vitro infections to see if this was a

Page 61: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

48

general ∆feoB phenotype, or if the phenotype was unique to the ATCC LVS

genetic background (Fig. II.12). Surprisingly, only the ATCC LVS ∆feoB mutant

exhibited an intracellular growth defect, while the mutants in the Iowa LVS and

RML LVS genetic backgrounds grew similar to their respective parental strains.

During the course of these infections it was observed that approximately 1.5- to

2-fold higher CFUs were obtained at the four-hour time point for the ATCC LVS

∆feoB. Previous work in our lab has identified increased uptake as a phenotype

associated with genetic defects in capsule and LPS biosynthesis, so an

immunoblot for each of these structures was performed on the ATCC LVS ∆feoB

(not shown). This revealed that the the ATCC LVS ∆feoB indeed lacked capsule

and LPS, possibly due to an undefined second site mutation; it was not pursued

further.

Page 62: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

49

0

20

40

60

80

100

120

140

160

Iowa RML ATCC fevR

Mill

er u

nits

(nor

m. t

o no

pro

mot

er

cont

rol)

PiglA-lacZ β-galactosidase activity

Figure II.1. RML LVS has similar FPI reporter gene expression and lacks glycosylated proteins seen in virulent Type A and Type B strains of F. tularensis. A) Promoter activity of the Francisella Pathogenicity Island gene iglA in three LVS biovars. Miller assays were performed as described in the materials and methods. No significant differences were observed between Iowa, RML, or ATCC LVS. Shown is combined data from three replicates. B) Immunoblot analysis of low molecular weight O-antigen positive, proteinase K sensitive bands observed in virulent F. tularensis subsp. tularensis and subsp. holarctica, but not in Iowa LVS. O-antigen ligase (FTT1238/FTL0706c) mutants were generated via the TargetTron method. Samples were treated with proteinase K (PK) overnight and separated by SDS-PAGE. Membranes were probed with the anti-O-antigen monoclonal antibody FB11. Shown is a representative blot from two replicates.

A B

Page 63: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

50

Figure II.2. Location of the amino acid residue that is mutated in the RML LVS FeoB. The protein sequence was submitted to HMMTOP to predict transmembrane domains, then visualized using TMRPres2D1,2. The mutated residue is predicted to occur in a cytoplasmic loop near the membrane and its location is indicated by an arrow in the figure above.

Page 64: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

51

Figure II.3. RML LVS exhibits growth restriction under iron limitation on MMH agar. A) Bacteria were grown in standard MMH broth overnight, then serially diluted in PBS and spotted onto MMH agar containing 0.025%, 0.0125 %, or no added ferric pyrophosphate. B) Bacteria were grown in standard MMH broth overnight, and then ~106 CFU were spread onto MMH agar lacking added ferric pyrophosphate. Sterile discs were then placed onto the plates and 10 μl of ten-fold dilutions of ferric pyrophosphate were spotted onto the discs: 0.025% (upper left), 0.0025% (upper right), 0.00025% (bottom left), or sterile water (bottom right). Shown are representative data from two similarly designed replicates.

A

B

Page 65: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

52

0.00

500.00

1000.00

1500.00

2000.00

Iowa RML ATCC

Mill

er u

nits

PfslA-lacZ activity 1x Fe

0.01x Fe

Figure II.4. RML LVS has iron-related gene expression consistent with less intracellular Fe2+ in iron limiting media. Each strain carried the PfslA-lacZ plasmid which is responsive to changes in iron concentrations. Bacteria were grown overnight in Chamberlain’s defined medium with either 35 μM FeSO4 or 350 nM FeSO4 and Miller assays were performed as described in the materials and methods. Shown is a representative Miller assay from three replicates.

Page 66: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

53

Figure II.5. Constitutive fur expression inhibits robust growth of RML LVS. Each strain was transformed with the vector control plasmid pBB103 or pBB103 with Pgro-fur and grown overnight in standard MMH broth. Bacteria were serially diluted in PBS and plated onto standard MMH agar. Shown is a representative assay.

Page 67: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

54

0.00

100.00

200.00

300.00

400.00

500.00

600.00

Empty vector V. choleraefeoABC

Ft feoAB Ft feoAB D471Y Schu S4 feoAB

Mill

er u

nits

E. coli ΔfeoB fhuF::λplac Mu activity

Figure II.6. RML feoB allele does not complement an E. coli ΔfeoB iron reporter strain. E.coli H1771 was transformed with the control plasmids pWKS30 (empty vector), the positive control plasmid with the Feo system from V. cholerae in pWKS30, or pWKS30 expressing either LVS feoB allele, or the Schu S4 feoB allele. The latter three strains also carried derivatives of pBB103 with Pgro-feoA as FeoB is known to require FeoA in other bacterial species. Miller assays were performed as described in the materials and methods. Shown is a representative assay from three replicates.

Page 68: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

55

1.E-01

1.E+00

1.E+01

1.E+02

Iowa RML ATCC

Perc

ent s

urvi

val (

norm

aliz

ed to

RM

L)

Survival 100 μM H2O2

Figure II.7. RML LVS is significantly more resistant to H2O2 lethality. Bacteria were grown in standard MMH broth overnight, centrifuged at 13,200 rpm for 5 minutes, and re-suspended in sterile PBS with or without 100 μM H2O2 in 200 μl total volume in a 96 well dish. These were incubated without shaking for one hour at 37° C, 5% CO2 before serial dilution in PBS and spotting onto standard MMH agar. Shown is a representative assay from >3 replicates.

Page 69: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

56

1.E+00

1.E+01

1.E+02

RML+feoB (D471Y) RML+feoB (WT)

Perc

ent s

urvi

val 1

00 μ

M H

2O2

Survival 100 μM H2O2

30 min

1 hr

Figure II.8. RML LVS can be sensitized to H2O2 by overexpression of a functional feoB. RML LVS was transformed with pBB103 containing either feoB allele being expressed from the groE promoter. H2O2 sensitivity assays were performed as described in the materials and methods. No colonies were observed from RML + feoB (WT) after one hour H2O2 treatment at the level of detection of 102 CFU. Shown is a representative assay from two replicates.

Page 70: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

57

1.E+00

1.E+01

1.E+02

1.E+03

Iowa Iowa ΔfeoB Iowa ΔfeoB+feoB (WT)

Iowa ΔfeoB+feoB (D471Y)

Perc

ent s

urvi

val (

norm

aliz

ed to

Iow

a)

Survival 100 μM H2O2

Figure II.9. FeoB-mediated sensitivity to H2O2 is independent of the RML LVS genetic background. A clean feoB deletion mutant was constructed in the Iowa LVS genetic background and the mutant was complemented with each allele expressed from the gro promoter. H2O2 sensitivity assays were performed as described in the materials and methods. Shown is a representative assay from two replicates.

Page 71: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

58

0

20

40

60

80

100

120

Iowa IowaΔdyp IowaΔdyp + Pgro-dyp

IowaΔdyp + Pgro-dyp93

Perc

ent s

urvi

val (

norm

aliz

ed to

Iow

a)

Survival 100 μM H2O2

Figure II.10. Dyp peroxidase protects against H2O2. A clean deletion mutant of dyp was constructed and subsequently complemented with plasmid-borne Pgro-dyp and Pgro-dyp93 constructs. H2O2 sensitivity assays were performed as described in the materials and methods. Shown is a representative assay from two replicates.

Page 72: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

59

1.E+00

1.E+01

1.E+02

1.E+03

Iowa RML ATCC

Fold

gro

wth

24 hour growth in A549 cells

Figure II.11. Fold growth of LVS biovars in A549 cells. Cells were infected at a multiplicity of infection of 100:1 and gentamicin protection assays were performed as described in the materials and methods. One replicate is shown.

Page 73: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

60

1.E+00

1.E+01

1.E+02

1.E+03

Fold

gro

wth

24 hour growth A549 cells

1.E+00

1.E+01

1.E+02

RML ΔfeoB

Fold

gro

wth

24 hour growth A549 cells

Figure II.12. Contribution of feoB alleles to intracellular growth in A549 cells. Bacteria were centrifuged onto cells at 600 rpm for four minutes at a multiplicity of infection of 500:1. A) Gentamicin protection assays were performed as described in the materials and methods. Only ATCCΔfeoB and ATCCΔfeoB + Pgro-feoB (WT) were significantly different from all strains. Shown is the combined fold growth for three replicates. B) The RML LVS feoB mutant trends towards less intracellular growth than the parental strain, but the two are not statistically significant (P>0.5). Shown are combined fold growths from two replicates.

A

B

Page 74: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

61

1.E+00

1.E+01

1.E+02

Iowa RML ATCC IowaΔfeoB IowaΔdyp

Fold

gro

wth

24 hour growth in BMM

Figure II.13. Twenty-four-hour growth of LVS biovars and mutant derivatives in murine bone marrow-derived macrophages. Bacteria were passively infected (no centrifugation) for three hours at a multiplicity of infection of 20:1. Gentamicin protection assays were performed as described in the materials and methods. Shown is the combined fold growth for three replicates. No significant differences were observed between strains.

Page 75: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

62

Discussion

Francisella tularensis is a highly pathogenic bacterium for which no

effective licensed vaccine exists. Additionally, the extreme infectiousness of the

organism has raised the possibility of its intentional release, thus precipitating the

need for a vaccine. The Live Vaccine Strain of F. tularensis subsp. holarctica

has been used to vaccinate laboratory workers in the past, but the immunity

stimulated by it was not sufficient to protect against moderate levels of

aerosolized virulent subspecies11. Furthermore, the genetic mutations that

attenuated LVS were unknown, and it was not clear if reversion to virulence was

a possibility. While LVS is not likely to be re-licensed for human vaccination, it

has served as a standard in animal models by which to measure candidate

vaccines, and as a tool to discover correlates of immunity to F. tularensis.

Indeed, in a previous report it was found that the RML LVS biovar stimulated

protective immunity in a mouse model of vaccine against virulent F. tularensis

Schu S4 by inducing higher numbers of effector T cells196. Building on these

observations, I sought to characterize some of the genetic differences between

biovars of LVS and explore how they may affect the outcomes of mouse models

of virulence and immunity. Here I report that a difference between the RML LVS

and the Iowa or ATCC LVS biovars is in the homeostasis of ferrous iron.

Iron is one of the most abundant metals on the planet but is often found in

the environment in an insoluble oxidized state, thus bacteria have evolved

sophisticated molecular machinery to scavenge iron from their local

environment120. Bacterial pathogens must acquire iron within the host

Page 76: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

63

environment, however they must also contend with the host immune response,

which can sequester iron and limit a pathogen’s growth204-207. F. tularensis also

requires iron for productive infection, as double mutants in LVS and F. tularensis

Schu S4 lacking both ferrous and ferric iron uptake pathways are incapable of

intracellular growth in vitro and are attenuated in murine infections94,100.

Additionally, F. tularensis induced upregulation of a number of host iron related

genes, including transferrin receptor, to increase the labile iron pool in the cytosol

of infected host cells208. Although F. tularensis requires iron for growth in a host,

it appears to be unique amongst pathogens in that the bacterium’s virulence may

depend in part on maintaining optimally low levels of iron within its cytoplasm, as

the highly virulent subsp. tularensis strains had approximately 4 to 5-fold less iron

than the moderately virulent subsp. holarctica strains122.

Consistent with these observations, I have shown that the more virulent

RML LVS biovar encodes a non-synonymous mutation in the feoB gene that

affects its ferrous iron levels. The strain grows similar to the Iowa and ATCC

LVS biovars in standard propagation media, yet is restricted for growth when the

iron concentrations are significantly reduced. This suggests that the intracellular

iron concentrations may be optimally low in all strains under normal growth, but

when extracellular iron levels in the media are significantly lowered, the RML

LVS is not as proficient at scavenging iron as the Iowa and ATCC LVS.

Numerous labs have reported that the fsl iron uptake operon genes are tightly

regulated by iron concentrations in the media, so we used a lacZ reporter the

measure the promoter activity of fslA, the first gene of the fsl operon in each

Page 77: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

64

strain under rich and limiting iron conditions95,98,102. As with growth phenotypes,

there was no detectable difference in PfslA-lacZ activity under normal growth

conditions; however, the RML LVS had increased reporter activity relative to the

Iowa and ATCC LVS after growth in iron limiting media. This is consistent with

the strain having less intracellular ferrous iron under these conditions, as Fur

regulates this promoter; higher reporter activity is interpreted to mean that less

Fe2+ is available to bind Fur protein and mediate repression of the fslA

promoter95.

The PfslA-lacZ expression is likely specific to the feoB encoded by RML

LVS, as disruptive mutations in this gene are predicted to directly impact ferrous

iron levels and no genetic differences were detected in other iron related loci. It

was important, however, to show specificity to this allele. To achieve this, I

tested the ability of each feoB to functionally complement an E. coli ΔfeoB iron

reporter strain. The results showed that the RML LVS feoB allele did not

complement the reporter mutant, while the allele from Iowa/ATCC LVS did.

Thus, the FeoB D471Y protein did not significantly import ferrous iron in a

heterologous system. While little or no function was observed in E. coli, we

believe that the FeoB D471Y does retain minimal function in LVS, as the PfslA-

lacZ reporter activity is higher in a RML LVS ΔfeoB mutant than in the parental

RML LVS (data not shown). To our knowledge this is the first direct genetic

evidence that explains differences in iron content between F. tularensis strains,

though it is almost certainly unique to LVS and the effect that we observed is

Page 78: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

65

likely subtler than that when comparing between subsp. tularensis and subsp.

holarctica strains.

The low ferrous iron phenotypes exhibited by subsp. tularensis have been

reported to correlate with resistance to the lethal effects of H2O2, so I sought to

characterize the relative sensitivity of each LVS biovar in vitro. Indeed, the RML

LVS exhibited nearly ten-fold more resistance than the Iowa LVS, which encodes

a functional FeoB, and one hundred-fold or more resistance than the ATCC LVS.

The latter strain is significantly more sensitive to H2O2 as it is competent for Fe2+

uptake through its functional FeoB, and because it encodes a Dyp peroxidase

lacking 31 amino acids near the C-terminal portion of the protein. The mutant

Dyp has been shown previously to increase H2O2 sensitivity, and a clean deletion

of dyp in the Iowa LVS recapitulates this phenotype127. I found that the RML LVS

could be sensitized to the H2O2 lethality by supplying the functional feoB allele

under constitutive expression on a plasmid; this effect was much less

pronounced when the allele supplied was its own. I further showed that the

increased H2O2 lethality associated with overexpression of a functional feoB was

not unique to the RML LVS genetic background, as an Iowa LVS ΔfeoB mutant

complemented with the mutant allele was more resistant to H2O2 than that

complemented with the functional allele. I think these sets of experiments are

significant, as the strains only differed by a single nucleotide in feoB, yet

exhibited nearly ten-fold differences in their sensitivities to H2O2, irrespective of

genetic background. By virtue of its lower function FeoB, the RML LVS may be

better at avoiding Fe2+ associated toxicity in the context of the host environment.

Page 79: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

66

Somewhat surprisingly, I found that the RML and Iowa LVS ΔfeoB strains

did not exhibit the same intracellular growth defect in A549 cells as I did with the

ATCC ΔfeoB; this defect had originally been reported by Thomas-Charles, et.

al.96. Unfortunately, further analysis of this mutant showed that it had lost

capsule and LPS structures, likely through a second, unspecified mutation.

Phase variation with these structures has been reported for LVS before, so the

feoB mutation may somehow increase the probability of phase variation

occurring209,210. The mutagenesis strategy that I employed makes use of a two-

step homologous recombination process, and the mutagenesis vector is

integrated onto the chromosome during the first step. Immunoblotting of the

parental ATCC strain with the integrated vector revealed no capsule or LPS

defects, so I may have been unlucky when picking an ATCC ΔfeoB colony for

long-term storage. It is not known if the intracellular growth defect of the ATCC

LVS feoB mutant reported by Thomas-Charles, et. al. is due to the same

phenomenon as I observed, or if the differences between mutagenesis schemes

would result in different phenotypes. The mutant that I constructed lacked the

entire coding sequence of feoB, while that of Thomas-charles, et. al. only

removed 410 amino acids from the middle of the protein, which is 747 amino

acids in length. Curiously, I did observe modest recovery of intracellular growth

when the ATCC ΔfeoB was complemented on a plasmid with the low function

RML LVS feoB allele, though there was significant variability between assays.

Page 80: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

67

CHAPTER III

CHARACTERIZATION OF BACTERIOFERRITIN IN OXIDATIVE STRESS

RESISTANCE

Introduction

A growing body of literature has shown that F. tularensis utilizes numerous

strategies to avoid activation of ROS-dependent host signaling pathways and

killing by host-generated reactive oxygen and nitrogen species125-129,156,158,211,212.

Furthermore, at least one phenotypic difference between the virulent F. tularensis

Schu S4 and the non-pathogenic F. novicida is that the latter is considerably

more sensitive to H2O2; this sensitivity is associated with activation of the AIM2

inflammasome via ROS likely derived from the mitochondria213. Given these

reports I mined a previously published transposon screen to look for genes

associated with iron homeostasis and oxidative stress resistance to assess the

role these processes play in pathogenesis and immunogenicity. This screen

identified the bacterioferritin (bfr) gene as important for F. tularensis Schu S4

growth in human monocyte-derived macrophages infected with pools of

transposon mutants56.

Bacterioferritin is a widely conserved member of the ferritin family of

proteins found in bacteria and archea, but its essentiality in iron storage and

H2O2 detoxification appears to vary cross taxa214,215. The bacterioferritin protein

exists as a spherical 24-mer, typically in the cytosol where it can sequester iron

within its hollow cavity216. While structurally and functionally similar to bacterial

ferritins, most bacterioferritins have been reported to bind heme, which is not

Page 81: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

68

observed with the former215,216. This associated heme may be important for the

controlled release of iron via electron transfer and the activity of accessory

proteins like the bacterioferritin-associated ferredoxin to meet cellular iron needs,

although it is unknown if this is a widely conserved function of bacterioferritin

among bacteria217.

Evidence exists for some species that bacterioferritin may be upregulated

by iron starvation and can protect against H2O2 toxicity214,218-221. For example, a

Pseudomonas aeruginosa mutant lacking the bacterioferritin gene bfrA exhibited

increased sensitivity to H2O2, yet showed no significant difference in total iron

content relative to the wild type222. Interestingly, while the iron levels were

similar, it was found that the BfrA protein may serve as a specific source of iron

for the catalase enzyme, as its activity was reduced by approximately half in the

bfrA mutant. In contrast, mutation of bfr alone did not enhance sensitivity of E.

coli to H2O2, but when mutated in combination with the ferric uptake regulator fur,

did provide a synergistic effect over that of the fur mutant alone. This study also

determined that ferritin served as the main reservoir for iron storage in E. coli,

with bacterioferritin contributing minimally in this capacity. Importantly,

Mycobacterium tuberculosis mutants lacking one or more bfr genes were more

susceptible to H2O2 than wild type, grew poorly in THP-1 cells, and were

attenuated in mouse and guinea pig models of infection223,224.

Bacterioferritin has appeared in the Francisella literature as being one of

the targets of the cellular and antibody immune responses in LVS immunized

mice, however, passive protection is not conferred by transfer of anti-Bfr

Page 82: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

69

antibodies225,226. Interestingly, a LVS sodB mutant upregulated bacterioferritin

almost 2.5-fold, suggesting that it may play a role in the oxidative stress

response of F. tularensis by sequestering free Fe2+ or by detoxifying H2O2225. It

is unknown if the F. tularensis bacterioferritin functions similarly to homologs in

other bacteria, and to my knowledge a mutant has not been reported beyond that

from Lindemann, et. al56. I report here that bacterioferritin provides protection

against H2O2 and modestly attenuates and alters the immunogenicity of a more

virulent biovar of LVS in a model of challenge with the fully virulent F. tularensis

Schu S4.

Materials and methods

Bacterial strains and growth conditions: Iowa LVS (University of Iowa) and

the RML LVS were routinely cultured on modified Mueller-Hinton (supplemented

with 1% glucose, 0.025% ferric pyrophosphate, and 2% IsoVitaleX) agar or in

broth, with 50 μg/mL of kanamycin or spectinomycin, as needed. Agar plates

were incubated at 37°C with humidity and 5% CO2 while broth cultures were

grown at 37°C, shaken at 200 rpm.

Mutant construction: Deletion of bfr (FTL_0617) was achieved by homologous

recombination using derivatives of the non-replicating plasmid pJC84. Briefly,

upstream and downstream flanking DNA was amplified via PCR, and amplicons

were cloned into the multiple cloning site of pCR2.1 (Life Technologies). A

spectinomycin resistance cassette was cloned into the AvrII site in the 3’ end of

Page 83: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

70

each upstream PCR fragment. The upstream-spectinomycin resistance fragment

was removed by digestion with AscI, and cloned into the AscI site in the 5’ region

of the downstream PCR plasmid. The entire upstream-spectinomycin resistance-

downstream fragment was cloned into the BamHI site of the suicide plasmid

pJC84. Finally, the spectinomycin resistance cassette was removed by digestion

with AvrII and the plasmid was re-ligated. Plasmids were electroporated into

LVS (2.5 kV, 25 μF, and 600 Ω), and the bacteria were plated onto MMH agar

with 50 μg/mL kanamycin after 2-3 hours of outgrowth. Organisms were then

grown overnight in broth lacking kanamycin, and were plated onto MMH agar

with 8% sucrose for sacB-mediated counter-selection. Kanamycin sensitive

colonies were screened by colony PCR to detect deletion of the bfr gene.

Primers were designed such that they flanked the coding sequence of each gene

such that an amplicon would be produced regardless of genotype.

β-galactosidase reporter activity: To assess iron uptake-regulated gene

expression, Miller assays were performed with Iowa, RML, and ATCC LVS, as

well as the RML ∆feoB and RML ∆bfr strains carrying the bfr promoter-lacZ

fusion on the pBB103 plasmid77. RML and its feoB and bfr mutant derivatives

were also assayed for PfslA-lacZ activity to estimate relative intracellular Fe2+

levels. Bacteria were grown in Chamberlain’s defined medium (supplemented

with 50 μg/mL spectinomycin) or standard MMH broth overnight, and β-

galactosidase activity was assayed using the standard Miller assay199.

Page 84: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

71

Hydrogen peroxide sensitivity: To measure resistance to H2O2-mediated

killing, mid- to late-log phase organisms were pelleted at 13,200 RPM for 5 min,

washed in PBS, and ~106 CFU were resuspended into 200 μL PBS with or

without 100 μM fresh hydrogen peroxide (H2O2) in a 96-well dish. The samples

were incubated at 37°C with humidity and 5% CO2 for one hour. The culture

from each well was then serially diluted, plated onto modified Mueller-Hinton agar

(with antibiotic where necessary) and the number of surviving organisms for each

strain was enumerated after 2-3 days.

In vitro infections: A549 cells were cultivated in DMEM supplemented with

10% heat-inactivated fetal bovine serum (HI-FBS). Cells were enumerated and

seeded into 24 well dishes at a density of 3x105 per mL in DMEM with 10%

L929-conditioned media, 10% heat-inactivated FBS. Multiplicity of infection

(MOI) was estimated by measuring the OD600 value of mid to late-log phase

grown bacteria, and confirmed via serial dilution onto modified Mueller-Hinton

agar and enumeration after 2-3 days’ growth at 37°C with humidity, 5% CO2.

Bacteria were allowed to associate with the cells for 3 hours, after which time the

cells were washed 3x with PBS, and incubated in DMEM supplemented with HI-

FBS and 100 μg/mL of gentamicin for one hour, cells were then washed 3x with

PBS and either lysed or incubated in antibiotic-free media for a further 20 hours.

At the 4 and 24-hour time points 0.1% saponin was added to each well. Wells

were scraped and vigorously pipetted to disrupt the cells and liberate intracellular

bacteria for serial dilution and enumeration.

Page 85: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

72

Murine infections: 8-10 week old female C57BL/6 mice were maintained on

corncob bedding for one week prior to infection. Mice were intranasally infected

with 25 μL of various doses of F. tularensis LVS strains and mutant derivatives

that had been resuspended in PBS. Inocula were calculated by measuring the

OD600 value of mid- to late-log phase grown organisms, and were confirmed by

serial dilution onto modified Mueller-Hinton agar and enumeration after 2-3 days’

growth at 37°C with humidity, 5% CO2. Moribund animals (defined as having lost

25% of the initial body weight) were sacrificed in accordance with the protocol

approved by the University of Iowa Institutional Animal Care and Use Committee.

Results

A bacterioferritin mutant has modest reduction in PfslA-lacZ activity

Data from the second chapter of this thesis highlighted the connection

between ferrous iron homeostasis and resistance to toxicity associated with

exposure to H2O2. To further extend these findings, I mined a published

transposon screen from our lab to look for mutants that may have defects in iron

metabolism and oxidative stress resistance. One gene fit these criteria, a

putative bacterioferritin encoded at the FTT_1441 locus56. A significant

proportion of the genes identified as important for growth in human macrophages

were those with unknown function or no obvious homologs, so it is not clear if

any of these has an association with iron metabolism.

Page 86: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

73

Bacterioferritin forms large spherical multimers that store iron via a

reaction that oxidizes Fe2+ to Fe3+, making a mutant in F. tularensis an attractive

target for this study215. I constructed a clean Δbfr mutant in the RML LVS

background and supplied the PfslA-lacZ reporter construct on a plasmid to test for

differences in iron regulated promoter activity between the mutant and parental

strains (Fig. III.2). After overnight growth in standard MMH broth, reporter activity

was approximately 15-20% lower in the RML LVS Δbfr mutant than in the

parental strain, consistent with the hypothesis that a mutant lacking the

bacterioferritin protein complex may have an increased intracellular pool of Fe2+

in standard laboratory culture conditions. Construction of a complemented

mutant is underway at the time of writing, however, the bacterioferritin gene is not

in an operon and the ~600 base pairs of sequence immediately downstream is

not annotated to encode a gene or other functional element, so it seems unlikely

that the mutation has polar effects.

Bacterioferritin promoter activity is not significantly induced in iron limiting media

Although the bfr promoter has been reported as having high basal activity,

it was of interest to see if there was differential regulation amongst the LVS

biovars227. Sequence analysis shows that there is a Fur box motif (ATAATGAT)

36 base pairs upstream of the annotated start codon, suggesting that bfr

expression may be responsive to iron levels. Differential expression of an iron

storage gene could represent another phenotypic difference between the lower

iron RML LVS and the Iowa and ATCC LVS biovars. A plasmid construct

Page 87: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

74

containing 150 base pairs upstream of the bfr coding sequence fused to lacZ was

introduced into each LVS biovar and promoter activity was assayed in both

standard MMH and CDM with high or low iron. Unlike the PfslA-lacZ reporter, no

strong induction in low iron CDM was observed in any of the strains, suggesting

that the bacterioferritin promoter may not be regulated by intracellular iron levels

directly (Fig. III.1). Reporter activity in the RML and ATCC LVS tended to be

slightly higher than in the Iowa LVS in low iron CDM, but significance was only

reached for the RML LVS, though the minimal fold induction in low iron that was

observed in each experiment was such that it is not clear that these are

physiologically significant differences between each strain.

Interestingly, the bacterioferritin promoter showed a modest change in

activity in the RML LVS ΔfeoB and Δbfr mutant backgrounds after overnight

growth in standard MMH broth. Relative to RML LVS the ΔfeoB mutant

consistently exhibited ~20-30% increased expression from the bacterioferritin

promoter, while the Δbfr mutant had ~20% less expression. These gene

expression patterns match that seen from the PfslA-lacZ reporter in each mutant

background, indicating that there may be subtle regulation at the bacterioferritin

promoter in wild type strains that is not detectable using Miller assays under the

conditions used here.

Page 88: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

75

Bacterioferritin protects against H2O2 but is not required for intracellular growth in

A549 cells

To assay the role of bacterioferritin in F. tularensis oxidative stress

resistance, I performed H2O2 sensitivity assays as described in the materials and

methods. As before, the RML LVS exhibited relatively high level resistance to

H2O2, while the Δbfr mutant had ~10-fold fewer viable organisms, levels similar to

the Iowa LVS (Fig. III.3). This data suggests that bacterioferritin protects against

oxidative stress and is consistent with what is known for Neisseria gonorrhea and

Mycobacterium tuberculosis. I next performed in vitro infections with RML LVS

and the Δbfr mutant in the human adenocarcinoma A549 cell line to model

intracellular growth (Fig. III.4). These experiments revealed that the Δbfr mutant

is competent to escape the phagosome and grow in the cytosol of these cells,

thus bacterioferritin is dispensable for this aspect of F. tularensis pathogenesis.

Bacterioferritin contributes to virulence and immunogenicity of RML LVS in vivo

Strains with increased sensitivity to H2O2 are known to have altered

virulence properties, so we sought to determine if this phenotype resulted in

attenuation in a mouse model of infection125,127,212. The RML LVS was previously

reported to have a relatively low LD50 (174 CFU) in C57BL6 mice; this was

confirmed in an independent experiment with two groups of mice (n = 5 per

group) infected with either 133 CFU or 1.33x103 CFU of RML LVS (Fig. III.5)196.

All mice succumbed by day 7 to infection with 1,330 CFU RML LVS. Sixty

percent survival was observed in the group infected with 133 CFU; the average

Page 89: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

76

weight loss was approximately 23 percent of the pre-infection weights. To test if

bacterioferritin was important for virulence in vivo three groups of mice (n = 5 per

group) were intranasally infected with 215 CFU, 2.15x103, or 2.15x104 CFU of

the RML LVS Δbfr mutant, and their weight and health status was measured. All

mice that received 2x102 CFU survived (average weight loss of ~12 percent)

while forty percent survived infection with 2x103 CFU (average weight loss of ~22

percent); none survived 2x104 CFU (Fig. III.5 and III.6). Importantly, it took

>2,000 CFU Δbfr to lead to survival curves and weight loss that approximated

that seen for mice infected with only 133 CFU of RML LVS, a greater than 15-fold

difference in dose. This is consistent with the hypothesis that bacterioferritin

contributes to the fitness of F. tularensis LVS in the host environment.

It was previously shown that an H2O2 sensitive biovar, ATCC LVS,

stimulates relatively weak protection against challenge with F. tularensis Schu

S4, while the H2O2 resistant RML LVS provided considerably better protection.

To test the hypothesis that the efficacy of RML LVS immunization depended on

H2O2 resistance, we challenged surviving Δbfr-infected mice (n = 5 for 215 CFU

Δbfr, n = 2 for 2,150 CFU Δbfr) and a control group of mice that had been

intranasally infected with 121 CFU of RML LVS (n = 3) coincident with the Δbfr

infections (Fig. III.7). Approximately six weeks after the initial infection the

groups were challenged with 25 CFU F. tularensis Schu S4. All mice previously

infected with RML survived the challenge, while all mice previously infected with

either 215 CFU or 2,150 CFU Δbfr succumbed with only a slight increase in time-

Page 90: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

77

to-death relative to naïve controls, indicating that bacterioferritin is necessary for

RML LVS to stimulate an optimal immune response.

Page 91: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

78

0

20

40

60

80

100

120

140

160

Iowa RML ATCC

Mill

er u

nits

nor

mal

ized

to Io

wa

LVS

Pbfr-lacZ activity

1x iron0.01x iron

Figure III.1. Bacterioferritin promoter activity is similar between LVS biovars grown in Chamberlain’s defined medium with high or low iron. The Pbfr-lacZ construct was introduced to each strain and reporter activity was determined via Miller assay, as described in the materials and methods. All values were normalized to those from Iowa LVS. Shown are combined data from four replicates.

Page 92: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

79

020406080

100120140160180200

RML RMLΔfeoB RMLΔbfr

Mill

er u

nits

nor

mal

ized

to R

ML

Promoter activity of fslA and bfr

fslA-lacZ

bfr-lacZ

Figure III.2. Bacterioferritin promoter activity in is upregulated in ∆feoB and downregulated in Δbfr in MMH broth. The PfslA-lacZ and Pbfr-lacZ constructs were introduced to each strain and reporter activity was determined via Miller assay, as described in the materials and methods. Data were normalized to the RML LVS. Shown is combined data from five experiments with RML and Δbfr, and four experiments with ∆feoB for PfslA-lacZ activity, and four experiments for Pbfr-lacZ.

Page 93: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

80

1.E+00

1.E+01

1.E+02

1.E+03

RML RMLΔfeoB RMLΔbfr

Perc

ent s

urvi

val n

orm

aliz

ed to

RM

L

Survival 100 μM H2O2 Log

Overnight

Figure III.3. Mutants lacking bfr and feoB differ in their H2O2 sensitivity depending on phase of growth. Strains were subjected to H2O2 sensitivity assays at ~mid-log phase and after overnight growth. Data were normalized to RML. Shown is combined data from four experiments.

Page 94: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

81

1.E+00

1.E+01

1.E+02

1.E+03

RML RMLΔbfr

Fold

gro

wth

24 hour growth A549 cells

Figure III.4. The bacterioferritin mutant is proficient for intracellular growth in vitro. Gentamicin protection assays were performed as described in the materials and methods. Shown is combined data from four experiments.

Page 95: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

82

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post-infection

RML intranasal infection

RML 133 CFU (n = 5)

RML 1,330 CFU (n = 5)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post-infection

RML Δbfr intranasal infection

RMLΔbfr 215 CFU (n = 5)

RMLΔbfr 2,150 CFU (n = 5)

RMLΔbfr 21,500 CFU (n = 4)

Figure III.5. RML Δbfr is modestly attenuated in murine infection via the intranasal route of inoculation. Mice were infected ten-fold dilutions of RML LVS or the Δbfr mutant that had been suspended in 25 μl sterile PBS. Health was monitored for the duration of infection and mice were humanely sacrificed if ≥25% of initial weight was lost. Data shown are from one replicate.

Page 96: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

83

0

5

10

15

20

25

30

35

RML 133 CFU RML 1,330CFU

Δbfr 215 CFU Δbfr 2,150 CFU Δbfr 21,500 CFU

Aver

age

perc

ent w

eigh

t los

t Average lowest weight

Figure III.6. RML Δbfr-infected mice lost less weight than mice infected with the wild type RML LVS. Shown is the average lowest weight for each group of infected mice from Figure III.5. Animals were humanely sacrificed if ≥25% of initial weight was lost. Data shown is from one replicate, n = 5 mice per experimental condition.

Page 97: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

84

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post-challenge

Schu S4 challenge - 25 CFU

121 CFU RML (n = 3)

215 CFU RMLΔbfr (n = 5)

2,150 CFU RMLΔbfr (n = 2)

Naïve (n = 10)

Figure III.7. RML requires bacterioferritin to elicit protection against an intranasal challenge of 25 CFU F. tularensis Schu S4. Mice were infected with either 121 CFU of RML LVS or 215 CFU of the bacterioferritin mutant and allowed to recover for fix weeks before challenge. The challenge inoculum was suspended in 25 μl sterile PBS and administered intranasally. Health was monitored for the duration of infection and mice were humanely sacrificed if ≥25% of initial weight was lost. Data shown is from one replicate.

Page 98: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

85

Discussion

That Francisella tularensis has numerous enzymes for the avoidance and

detoxification of H2O2 is not surprising, given that all aerobic organisms must

avoid the damaging and potentially lethal consequences of the Fenton

reaction228. Some of the ways that bacteria have evolved to limit this toxicity is to

produce antioxidant enzymes like catalase and superoxide dismutase, and these

enzymes function similarly in F. tularensis125,211. Bacteria also avoid Fenton

chemistry by limiting the availability of redox active metals like ferrous iron by

oxidizing and storing them in complexes formed by ferritin or bacterioferritin,

usually both, though one typically predominates. Here I provide the first

description of a bacterioferritin mutant in F. tularensis and provide preliminary

data suggesting a role in virulence.

Bacterioferritin is known to oxidize ferrous iron and store the ferric oxide

mineral in the hollow cavity formed by the 24-mer sphere215. I reasoned that a F.

tularensis RML LVS mutant lacking bacterioferritin may be unable to sequester

Fe2+ as effectively as the wild type strain, and that this may be reflected in iron

related gene expression. The PfslA-lacZ reporter indeed showed a modest

decrease in expression in the Δbfr mutant, indicating that the strain may have

more Fe2+ available to interact with Fur and repress expression of the reporter.

This result is consistent with the hypothesis that F. tularensis LVS uses

bacterioferritin to alter the pools of intracellular Fe2+ levels by oxidation and

storage. The reporter expression decreased by only ~15-20%, implying that the

Page 99: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

86

bacteria may also store iron via other mechanisms, possibly the ferritin protein; a

similar mutant analysis may prove useful.

The activity of the bacterioferritin promoter was not dramatically altered by

growth in low iron media, suggesting that the Fur box motif may not be important

under this level of iron limitation. Early proteomic studies compared the

proteomes of LVS grown in high and low iron conditions found either ~2-fold

induction of bacterioferritin in iron limiting media, or that the protein spot was only

observed in iron-starved bacteria79. Interestingly, a promoter trap library from

Zaide, et. al. found that the bacterioferritin promoter was approximately 10-fold

more active than that from the groEL promoter, itself known to be highly active in

Francisella species227. These two observations suggest that there may be more

complex regulation of bacterioferritin, possibly post-transcriptional. Francisella

encodes small RNAs in its genome, but their contribution to the physiology of

Francisella has been little studied229. Still, the RML LVS had a slight but

significant increase in bacterioferritin promoter activity relative to the Iowa LVS in

iron limiting media, and the RML LVS ∆feoB mutant had approximately 30%

higher expression than that observed in the parental strain. These data suggest

a link between FeoB activity, iron levels and the bacterioferritin promoter, though

more work is needed to flesh this relationship out. The ATCC LVS also had

somewhat increased activity, but tended to exhibit variability between replicates

and was not significantly different from either the RML or Iowa LVS biovars. This

may be similar to what is known for the oxidant-sensitive LVS superoxide

dismutase mutant, which upregulated bacterioferritin nearly 2.5-fold225. I interpret

Page 100: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

87

this to mean that LVS may be able to upregulate bacterioferritin in response to

oxidative stress and that the ATCC LVS, being one hundred to one thousand-fold

more sensitive to H2O2 than RML LVS, may increase bacterioferritin expression

to sequester free Fe2+. Alternatively, the bacterioferritin protein complex may be

required to supply the requisite iron for antioxidant enzymes, such catalase,

which may be upregulated to respond to the increased H2O2 stress experienced

by the ATCC LVS217,230.

As the iron regulated reporter indicated that the Δbfr mutant had increased

levels of intracellular ferrous iron, I hypothesized that the strain would be more

sensitive to H2O2. I found that the Δbfr mutant had approximately 75-90%

reduction in viability relative to the parental strain, indicating that bacterioferritin

has a role in resistance to oxidative stress in F. tularensis LVS. Interestingly, in

the course of these experiments I also observed that the RML LVS ∆feoB mutant

exhibited almost complete resistance to the levels of H2O2 used in these assays.

Coupled with the observation that this mutant also had significantly increased

levels of bacterioferritin promoter activity, these data suggest that the high level

resistance of the feoB mutant may be due both to having less intracellular Fe2+

and an increase in the H2O2 protective activity of bacterioferritin. This is

consistent with the observation that the RML LVS (with low FeoB activity) had a

small but significant increase in bacterioferritin promoter activity under iron

limitation. Attempts to construct a double ∆feoB Δbfr mutant are underway at the

time of writing.

Page 101: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

88

Surprisingly, I did not detect an intracellular growth defect from the RML

LVS Δbfr mutant, at least in A549 cells. Certainly, A549 cells are not likely to

replicate the cytosolic environment of a primary human macrophage, but the

transposon screen from which bacterioferritin was mined was based on the

decreased intracellular growth of a F. tularensis Schu S4 bacterioferritin

mutant56. Multiple possibilities exist to explain this discrepancy, and they are not

mutually exclusive. First, A549 cells are immortalized adenocarcinoma cells, not

professional phagocytes, and may not provide a sufficiently challenging growth

environment, thus the mild H2O2 sensitivity of the mutant may have negligible

effect on the mutant’s fitness. Secondly, the requirement of bacterioferritin for

efficient growth in the cytosol may be more important for the fitness of Schu S4

than it is for LVS. In this scenario LVS may encode factors that sufficiently

compensate for loss of bacterioferritin that may not be present or may not

function similarly in Schu S4. Indeed, Lee et. al. report that proteomics analysis

of LVS and a virulent F. tularensis subsp. tularensis clinical isolate exhibited

differences in bacterioferritin isoforms, suggesting subtle differences in protein

processing226. Lastly, the transposon screen in which bacterioferritin was

identified relied on infecting human macrophages with pools of transposon

mutants. It is possible that the Schu S4 bacterioferritin transposon mutant may

have been exposed to a more stressful intracellular environment if macrophages

were co-infected with one or more mutants that induced a host response that

increased bactericidal activities. Two replicates of murine BMM infection suggest

that an Iowa LVS Δbfr exhibits ~30% less fold growth than Iowa LVS (not

Page 102: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

89

shown), though time and resources have precluded me from rigorously testing

the RML LVS Δbfr in BMMs.

In the previous chapter I showed that a major phenotypic difference

between the RML and ATCC LVS biovars is the ability to resist H2O2 mediated

killing, due in part to differences in intracellular Fe2+ pools. I reasoned that the

increased H2O2 resistance phenotype of the RML LVS was critical to the

enhanced virulence it displays relative to ATCC LVS. The Δbfr mutant allowed

me to test the virulence of a RML LVS strain with increased H2O2 sensitivity

mediated by higher intracellular Fe2+ levels. Intranasal infections of mice

confirmed that bacterioferritin was required for full virulence of the strain, with the

mutant’s LD50 estimated to be ~1,500 CFU, nearly an eight-fold increase relative

to the parental strain (<200 CFU). Additionally, the mice exhibited differences in

the severity of disease, as mice infected with 133 CFU lost considerable weight

during the course of the infection, approximately twenty-three percent on

average. In contrast, the mice infected with 215 CFU of the Δbfr mutant lost

significantly less body weight during infection (approximately twelve percent,

P=0.0004), even though the dose was sixty percent higher. Only at a ten-fold

higher dose of the Δbfr mutant did the mice lose similar amounts of weight as

those infected with the lowest dose of the parent strain. I interpret this to mean

that the increased intracellular iron pool of this mutant makes it more sensitive to

host defense mechanisms that depend, at least in part, on oxidative stress.

Additionally, the RML LVS is known to be more efficacious than the ATCC

LVS at stimulating productive immunity, so survivors from the LD50 experiment

Page 103: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

90

were challenged with Schu S4 to test the hypothesis that RML LVS stimulates

protective immunity by a mechanism that requires resistance to H2O2. As in

Griffin, et. al., I found that mice given a sublethal infection of RML LVS all

survived challenge with 25 CFU Schu S4, while naïve controls succumbed to

infection. Consistent with my hypothesis, the RML LVS Δbfr did not stimulate

robust immunity in mice infected with this strain, as all succumbed from Schu S4

infection by day eight. These data suggest that H2O2 resistance somehow

contributes to the efficacy of the RML LVS as a vaccine against tularemia in

mice, however it is possible that other interpretations of the data are valid. For

example, the bacterioferritin protein is known to stimulate antibodies and T cell

proliferation in LVS immunized mice, however it isn’t clear that antibody

responses are effective at controlling infection with virulent F. tularensis Schu

S4225. Schu S4 is able to bind the active form of the host protease plasmin in

vitro, which can cleave Francisella specific antibodies to avoid the ensuing

bactericidal and inflammatory consequences of phagocytosis via antibody

mediated opsonization183. Furthermore, transfer of monoclonal antibodies to

Francisella bacterioferritin did not provide protection against lethal LVS

challenge, even though antibodies can to be highly effective at protecting against

this strain185,231. Taken together, this suggests that the lack of a bacterioferritin-

specific antibody response is not likely to explain the poor efficacy of the RML

LVS Δbfr at protecting against Schu S4.

It is possible that the absence of bacterioferritin and the lack of

subsequent antigen-specific T cell responses to the protein explain the poor

Page 104: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

91

protection of mice immunized with Δbfr, rather than the increased H2O2 sensitivity

of the strain. One way to test this hypothesis would be to complement a Δbfr

mutant with a mutant bfr allele with site-directed mutations at conserved residues

important for the H2O2 protective activity. Assuming protein structure and

stability were not affected, this could help clarify interpretation of the data in Fig.

II.7. For example, if the H2O2 protective activity of bacterioferritin is required for

RML LVS induced productive immunity, then a future rationally designed vaccine

strain might be designed such that its method of attenuation preserves H2O2

resistance. Alternatively, if the antigen provided by the bacterioferritin protein is

required but its activity is dispensable, then the data from Fig. III.7 would be

strong evidence that bacterioferritin is a major T cell antigen in vivo that is

required for an effective immune response to eliminate virulent F. tularensis.

This is important, as the difference in survival between mice immunized with

RML LVS or the Δbfr mutant was significant, and to my knowledge most studies

of F. tularensis T cell antigens typically utilize purified proteins for in vitro cell

stimulation, or vaccination of mice. The latter approach is specific, but does not

fully recapitulate the effectiveness of vaccination with LVS, as immunization of

mice with multiple Francisella specific T cell antigens failed to provide significant

protection against F. tularensis Schu S4232. If the alternative hypothesis is

correct, then a future vaccine strain may be designed to have constitutively high

expression of bacterioferritin.

Page 105: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

92

CHAPTER IV

CONCLUSIONS AND FUTURE DIRECTIONS

Francisella tularensis is a fascinating bacterium of extreme infectiousness

and virulence, yet the mechanisms governing these processes are still not well

defined. That inhalation of as few as 10 CFU is sufficient to cause a significant

and potentially life-threatening disease is quite remarkable, even among other

pathogens of similar stature. The bacterium apparently has few dedicated

virulence factors, and does not encode complex regulatory or signal transduction

systems. Even less well understood are the ways in which the bacteria prevent

the generation of robust immune responses, though it seems to be and active

process that contributes to the stealthy nature of the bacteria. This is likely due

to the combination of physical shielding of antigenic structures on the bacterium

via its capsule and the atypical LPS, as well as mechanisms that may depend on

the activity of the FPI system. Evidence is accumulating, however, that the

bacteria can actively influence host signaling systems to inhibit the production of

certain inflammatory cytokines, or to prevent antigen presentation to

lymphocytes233,234.

Although the Live Vaccine Strain of F. tularensis subsp. holarctica has

been used to vaccinate humans in the past, the strain retains some of the

mechanisms of virulence and immunosuppression. The bacterial factors

underlying these mechanisms may contribute to the inability of the LVS to induce

sterilizing immunity against fully virulent F. tularensis in a host, especially at

moderate to high dose challenge. It should be noted, though, that LVS is still

Page 106: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

93

among the most effective vaccines in mouse models for the study of immunity to

tularemia, and it represents a “gold standard” that future potential vaccines must

surpass, both in terms of efficacy but also safety.

This thesis has its roots in a desire to identify mutant strains of F.

tularensis that are attenuated and stimulate protective immunity in a mouse

model of immunization and disease. Our lab had previously identified and

described a number of mutants that lacked capsule and had truncated LPS

structures, and found that they stimulate a response that triggers host cell death

and significant inflammation in vivo, thus we reasoned that they may elicit a

productive immune response. These strain induced significant host tissue

damage at high inoculum, ~106 CFU, indicating that lower immunizing doses

would be needed. This is consistent with other immunization paradigms in the

Francisella field, and models of T cell development in environments with high or

low levels of inflammation235,236. Accordingly, I infected mice with two doses of

either waaY::trgtn or wbtA::Tn5 mutants or one dose of Iowa LVS and

subsequently challenged the mice with two doses of virulent F. tularensis Schu

S4. Surprisingly, the LVS immunized mice survived both doses of Schu S4

challenge significantly better than mice immunized with low doses of either

waaY::trgtn or wbtA::Tn5. Mice immunized with moderate doses of the mutants

had survival rates approaching that of LVS immunized mice, though they did not

surpass them. It should be reiterated that the waaY::trgtn or wbtA::Tn5 mutants

stimulate significant inflammation and tissue damage, so future evaluation of the

Page 107: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

94

ability of these mutants will need to account for that possibility when deciding on

the dose and route of inoculation.

As LVS was more effective at protecting mice against virulent F. tularensis

challenge than attenuated F. tularensis mutants, I began to focus my attention on

what attributes of LVS might be important for immunity. Through collaboration

with the Bosio lab at Rocky Mountain Laboratories I obtained two phenotypically

different biovars of LVS (RML and ATCC) as well as their genomes. The RML

LVS was reported to have a 50-fold lower LD50 than the ATCC LVS, yet it

elicited significantly better protection against subsequent challenge with F.

tularensis Schu S4. These biovars differ very little at the genetic level, though

two important differences exist. The first and most obvious was a 93 base pair

deletion in FTL_1773 of the ATCC LVS genome, which encodes a Dyp-type

heme peroxidase. This deletion had been reported before, and while I pursued a

genetic approach to characterize this locus another group published phenotypic

data on a dyp mutant using a similar approach127. The second genetic difference

between the LVS biovars was a non-synonymous substitution in the feoB

(FTL_0133) gene in the RML LVS genome; this changed the aspartate at residue

471 to a tyrosine. As it is known that virulent subspecies of F. tularensis have

lower iron levels and higher resistance to H2O2 than less virulent subspecies, I

hypothesized that the RML LVS was more virulent and a better vaccine strain

than the ATCC LVS as a consequence of it having lower levels of intracellular

iron and a fully functional Dyp-type peroxidase.

Page 108: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

95

I tested this hypothesis in a number of ways, including growth under

conditions where iron was limiting or in excess. I found that decreasing the iron

levels in MMH agar lead to growth restriction in all three of the LVS biovars

tested, including the LVS that had been used in our lab. The RML LVS exhibited

a much greater inability to grow under severe iron limitation than did either the

Iowa or ATCC LVS strains. This growth defect was also recapitulated in disc

diffusion assays, where the RML LVS grew in considerably smaller haloes

surrounding sterile discs spotted with iron solution. Interestingly, the RML LVS

does not exhibit iron limitation growth phenotypes in broth, suggesting that the

diffusion of iron and siderophores is sufficient for the strain to grow in this

condition. To date, iron-related growth defects reported in the literature have

been comparisons of mutants to wild type or between F. tularensis subspecies,

making the genetic data included here the first report of variable iron-related

growth phenotypes within nominally wild type bacteria from the same

subspecies96,103,122.

I extended this by showing that a lacZ reporter fusion to the promoter of

fslA was significantly more active in the RML LVS than in the other biovars when

the bacteria were grown in iron limiting media. The fslA gene is the first in the

siderophore biosynthesis and uptake operon, and is regulated by the Fur

transcriptional repressor97. The higher expression level in RML LVS is consistent

with the hypothesis that this biovar has less intracellular iron under iron limiting

conditions. Furthermore, the PfslA-lacZ reporter data indicate that the RML LVS

specifically has less ferrous iron, the reduced form of the metal that is imported in

Page 109: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

96

a FeoB-dependent manner in other bacteria. Consistent with this, artificially

increasing the intracellular levels of Fur via overexpression rendered RML LVS

growth on MMH agar sickly compared to the vector control and the Iowa and

ATCC biovars. These data strongly implicate the RML LVS FeoB D471Y as the

causative factor for the biovar’s iron and Fur-related growth restriction and

increased fslA promoter activity.

The functionality of FeoB from each strain was compared by

complementation of an E. coli feoB mutant with a chromosomal lacZ insertion at

the iron-regulated fhuF locus. The basal β-galactosidase activity of this strain is

high due to the lack of FeoB-mediated ferrous iron uptake, though it can be

repressed by complementation with a functional feo system111.

Complementation with the Iowa/ATCC LVS feoB allele mediated significant

repression of the β-galactosidase activity while complementation with the RML

LVS feoB did not. This indicates that the low iron growth restriction and high fslA

promoter activity of RML LVS is likely due to poor ferrous iron import mediated by

FeoB D471Y. The assay does not, however, give any indication on the stability

or localization of FeoB D471Y within the heterologous host, so it is possible that

the protein may be degraded by proteases, or insert in the inner membrane

improperly. Raising an antibody to FeoB or creation of an epitope-tagged

version of FeoB D471Y could be useful for addressing these possibilities.

Though not novel for the field of bacterial iron homeostasis, these experiments

also confirmed the requirement of FeoA for Francisella FeoB function, as no

repression of the iron-regulated reporter was observed in the absence of FeoA.

Page 110: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

97

As it is established that the Type A strains of F. tularensis have

approximately 4 to 5-fold less intracellular iron than the Type B strains, I included

the Schu S4 feoB allele in the E. coli iron reporter experiments. The encoded

protein differs from that of the annotated LVS protein by four amino acids,

suggesting that there may be functional differences that could account for the

strain’s low iron levels. Surprisingly, the Schu S4 feoB allele complemented the

E. coli reporter strain nearly as well as the Iowa/ATCC feoB allele, indicating that

the extremely low iron levels in this prototypical Type A strain are not due to a

decreased capacity of the Schu S4 FeoB to import iron, though it should be

noted that the FeoA supplied in these experiments was from F. tularensis LVS

and it differs by one amino acid. It is possible that the amino acid difference in

Schu S4 FeoA may decrease its ability to stimulate FeoB import activity.

I have also compared the promoter sequences of two known Fur-

regulated genes between Schu S4 and LVS to see if mutations may have

occurred that might decrease expression in the Type A strains. The promoters

for fslA and feoB are highly similar, though some nucleotide differences exist in

the latter. I created PfeoB-lacZ reporters for each strain’s promoter, but neither

exhibited much β-galactosidase activity so I did not pursue this further (data not

shown). Negligible color change was observed in Miller assay samples over the

course of an hour. There are multiple possible reasons for why minimal activity

was observed. First, the promoter sequences may not have contained all of the

relevant regulatory elements for transcription to occur, or may have included

elements that became inhibitory outside of their native chromosomal context due

Page 111: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

98

to the lack of associated upstream elements. Another possibility is that the basal

promoter activity is inherently low, which would be consistent with the potentially

dangerous effects of excess FeoB-mediated ferrous iron import. Differential

expression of feoB has been reported in the form of quantitative RT-PCR, so it is

possible that Miller Assays are not sensitive enough, or there may be post-

transcriptional of feoB mRNA via sRNA or RNAse-mediated degradation.

Whatever the case, the low iron status of Type A strains is likely due to complex

factors that will require the analysis of gene expression and protein activity of

both iron uptake pathways.

The low iron Type A strains were also shown to be more resistant to H2O2

in vitro, thus I hypothesized that the RML LVS is also more resistant as a result

of its low iron levels. Ferrous iron can participate in the Fenton reaction and lead

to lipid peroxidation, DNA damage, and inhibition of essential metabolic

enzymes. Indeed, I observed that the RML LVS was as much as 10-fold more

resistant than the Iowa LVS, which encodes a functional feoB in its genome. The

RML LVS was 100 to 1,000-fold than the ATCC LVS. As mentioned above, the

ATCC LVS encodes both a functional FeoB and a truncated Dyp-type peroxidase

that lacks a conserved heme-binding residue that likely renders the truncated

enzyme less functional. I confirmed that the Dyp enzyme protects against H2O2

by constructing a dyp deletion mutant in the Iowa LVS, which encodes a full

length dyp, and testing its sensitivity. Curiously, I was unable to construct a dyp

mutant in the RML LVS, though repeated attempts were made. The biological

significance of this is not clear, as Dyp peroxidases are known to have an

Page 112: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

99

extraordinarily broad substrate range and a potential genetic interaction with a

low function FeoB has no obvious explanation198.

The connection between the low functioning FeoB D471Y and H2O2

resistance was shown by overexpression of each feoB allele in the wild type RML

LVS and repeating the H2O2 sensitivity assays. As predicted, overexpression of

the Iowa/ATCC LVS feoB allele rendered the RML LVS significantly more

sensitive, while overexpression of the D471Y allele only modestly increased

sensitivity. This means that a single nucleotide change in feoB is responsible for

the enhanced resistance to H2O2 displayed by the RML LVS. This was not

unique to RML LVS, as an Iowa LVS ∆feoB mutant complemented with the

overexpression construct displayed similar phenotypes. These experiments also

showed that the feoB from RML LVS is not likely a null mutant, as its

overexpression did slightly increase sensitivity. Likewise, data from chapter III

shows that a ∆feoB mutant in RML LVS exhibits approximately 30-40% higher

PfslA-lacZ activity than the parent, suggesting that FeoB D471Y retains some

minimal level of function in F. tularensis LVS.

None of the mutants or overexpression strains exhibited intracellular

growth defects in vitro, save the ATCC ∆feoB mutant. As discussed in chapter II,

this strain had an undefined second site mutation that led to loss of capsule and

O-antigen polysaccharides. While it may be interesting that overexpression with

the feoB D471Y allele led to modest partial complementation, the undefined

nature of the causative mutation(s) that affected the capsule and LPS makes

interpretation of this result difficult. That the feoB mutants in RML LVS and Iowa

Page 113: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

100

LVS did not display growth defects in in vitro infections is largely consistent with

reports in the literature, save one96. As discussed in chapter II, Thomas-Charles,

et. al. report that a F. tularensis ATCC LVS feoB mutant fails to grow in the

human adenocarcinoma A549 cell line, though I see no such defects from the

Iowa or RML LVS. If this phenotype is due to a similar capsule/LPS defect to

what I observe with an ATCC LVS ∆feoB mutant, then it would be expected that

their mutant has growth defects in more than one cell type. This is not the case,

as their mutant replicated in human monocyte-derived macrophages, which have

been shown to undergo early cell death upon infection with capsule/LPS

mutants. Given that our mutant construction protocols are different, it is possible

that their mutant (internal deletion of ~400 amino acids) and my mutant (clean

deletion of coding sequence) simply have different phenotypes.

Given that the original report of multiple biovars of LVS demonstrated

differences in virulence and vaccine efficacy phenotypes, I wanted to perform

mouse studies with strains that had different combinations of feoB and dyp

alleles complemented on the chromosomes. Unfortunately, I have been

unsuccessful in my attempts to create these strains as of the time of writing,

though not for lack of effort. Complementation on the chromosome would allow

for regulation by the native promoters and remove the problem of gene dosage

effects introduced by constitutive expression on a multi-copy plasmid.

While working on these strains, I also created a bacterioferritin mutant in

the RML LVS. Lindemann, et. al identified a F. tularensis Schu S4 bacterioferritin

transposon mutant as being less fit in the environment of human monocyte-

Page 114: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

101

derived macrophages in vitro, suggesting a possible connection to iron, H2O2

resistance, and virulence. I confirmed that the RML LVS ∆bfr mutant had lower

PfslA-lacZ levels, suggesting that this strain had an increase in the intracellular

ferrous iron pool. The mutant was also more sensitive to H2O2, indicating that

the protein can protect F. tularensis LVS from H2O2 possibly by using H2O2 as a

cofactor to oxidize ferrous iron during the process of storing it as has been

reported for bacterioferritin-mediated iron storage mechanisms in other

organisms. Biochemical approaches to dissect Francisella bacterioferritin are

needed to describe its mechanism of action.

As discussed in chapter III, the RML LVS ∆bfr mutant did not exhibit

intracellular growth defects, suggesting that the transposon mutant identified in F.

tularensis Schu S4 may be more severely affected by loss of bacterioferritin

function, or that the way the screen through cells was performed may have

affected its viability. Interestingly, an early proteomics study of F. tularensis

reported that bacterioferritin protein expression was specific to subsp. holarctica,

but a later study by the same group found that Schu S4 expressed the protein,

but had reduced quantities of it in the membrane relative to Type B strains237,238.

The significance of this is not clear, but it suggests that bacterioferritin expression

patterns may differ in Schu S4 compared to Type B strains, or that its localization

is different. If this is the case, it is interesting to speculate that the increased

expression/localization of bacterioferritin in Type B strains may be a detriment to

the bacteria, as it is known that bacterioferritin is a potent Francisella antigen226.

This would imply that Schu S4 and other Type A strains may express

Page 115: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

102

bacterioferritin only in environments where it is absolutely necessary for the

survival of the bacterium, such as a host cell. Further work is needed with a

defined deletion mutant of bacterioferritin in the Schu S4 background examined

outside of the context of a mutant pool in vitro infection.

Despite the lack of an intracellular growth defect, the RML LVS ∆bfr

mutant allowed me to test the hypothesis that a key component of the virulence

of RML LVS and its ability to stimulate immunity in a mouse is its resistance to

H2O2 due to increased intracellular ferrous iron. Murine infections were

consistent with this hypothesis, as the mutant was mildly attenuated relative to

the parent strain. More importantly, subsequent challenge of ∆bfr immunized

mice with F. tularensis Schu S4 showed that bacterioferritin is a necessary factor

for RML LVS to elicit protective immunity, as all challenged mice succumbed

within eight days. In comparison, one-hundred percent survival was observed in

control mice that had been immunized with wild type RML LVS.

As discussed in chapter III, it is possible that the absence of the protein

and not necessarily its H2O2 protective enzymatic activity that leads to a less than

optimal immune response. Future work with site-directed mutants at conserved

residues critical for enzyme function should be performed to examine this

possibility. However, I interpret these results to mean that the H2O2 sensitivity of

the mutant stimulates a qualitatively different host response. It is known that the

H2O2 sensitive ATCC LVS elicits less poor protection relative to the resistant

RML LVS, even though its bacterioferritin gene is intact and the protein is likely

expressed appropriately196. I suspect that the mild to moderate H2O2 sensitivity

Page 116: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

103

of these strains leads to occasional loss of bacterial membrane integrity inside a

host cell that could lead to host surveillance systems recognizing bacterial factors

from these low level “leaky” events, such as bacterial DNA, and responding by

induction of proinflammatory cytokines. F. novicida is highly sensitive to H2O2

relative to F. tularensis and it activates the AIM2 inflammasome in murine

macrophages by a process that depends in part on H2O2 from the host’s

mitochondria213. My working model is that inflammation derived from a similar

process coupled to the as yet undefined immunosuppressive mechanisms that

inhibit production of certain proinflammatory cytokines and antigen presentation

lead to a smaller population of Francisella-specific effector memory T cells in

mice immunized with H2O2 sensitive strains of LVS.

This model is necessarily oversimplified, as the full scope of intracellular

events that are actively modulated by F. tularensis is still unknown. Additionally,

further experiments need to be performed, as the model is built on assumptions

that need testing. First, I am assuming that strains and mutants with excess

intracellular ferrous iron exhibit occasional instability in their membranes that

results in detectable loss of integrity due to Fenton reactions between the ferrous

iron and H2O2 generated from endogenous metabolism. In the cytoplasm of a

host cell there would be a contribution of H2O2 from host metabolism as well.

This loss of integrity could be tested via commonly used live/dead stain assays.

Secondly, I assume that occasional loss of integrity occurs in the cytosolic

environment of infected cells and that this correlates with activation of an

intracellular immune surveillance system and production of proinflammatory

Page 117: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

104

cytokines. This could be addressed with confocal microscopy of macrophages

infected with strains and mutants generated in this thesis. Similar experiments

include infection of host cells with bacteria harboring a plasmid with luciferase

driven by a eukaryotic promoter; it is thought that loss of membrane integrity

leads to leakage of the plasmid from the bacteria to the host cell cytoplasm, and

that the host then transcribes and translates the luciferase239. Thus, I could

infect cells with my strains and mutants and measure relative luciferase activity.

The model predicts that high sensitivity to H2O2 would correlate with high

luciferase activity.

In support of this model, studies of how F. tularensis physiology and

pathogenicity change after growth in modified Mueller-Hinton broth (MMH) or

Brain-Heart Infusion (BHI) have linked growth in MMH to the AIM2-dependant

stimulation of proinflammatory cytokines in infected murine macrophages240,241.

Importantly, BHI-grown mutants that are deficient in oxidative stress resistance,

katG and mglA, failed to inhibit cytokine production. I mention this because I

have compared PfslA-lacZ iron reporter activity and H2O2 sensitivity in strains

grown in MMH or BHI (data not shown) and found that bacteria grown in BHI

have five to seven-fold higher iron reporter activity and negligible sensitivity to

H2O2 at levels that cause significant lethality from bacteria grown in MMH. It is

important to note that the MMH used for F. tularensis propagation incudes

0.025% ferric pyrophosphate, making it an iron-rich medium. That the iron

reporter activity after growth in BHI is so high is striking; it is comparable to that

of bacteria grown in Chamberlain’s defined medium with 350 nM FeSO4, a

Page 118: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

105

concentration 100-fold less than typically used in this medium for standard

Francisella propagation. Taken together, this shows that wild type bacteria

grown in MMH have higher intracellular iron levels, increased instances of

membrane instability as shown by live/dead stain of broth grown organisms, and

they stimulate significantly high levels of proinflammatory cytokines from infected

murine macrophages via AIM2. Interestingly, the authors report that BHI is a low

iron media, but they do not pursue the role of high iron concentrations in MMH in

mediating membrane instability or induction of cytokines. My model predicts that

strains with excess intracellular ferrous iron may stimulate an inflammatory

response that likely leads to clearance of the bacteria and polarization of naïve T

cells to a short-lived effector state, rather than longer-lived memory state.

The ultimate goal of this thesis was to both identify the bacterial factors

responsible for the variability between F. tularensis biovars with respect to their

virulence and to characterize the host responses both in vitro and in vivo due to

these factors. Unfortunately, I was not able to accomplish this; however, the low

and moderate dose immunization experiments and the IglC-LCMV epitope fusion

tools that I developed in appendix A can clearly be used to supplement the

findings in this thesis. For example, the IglC-LCMV epitope fusion could be used

to characterize antigen-specific T cell populations in mice immunized with strains

generated in this thesis. These tools could test the hypothesis that H2O2

sensitive strains weakly stimulate proliferation of antigen-specific T cells, and that

resistant strains are more effective at this process. These tools could also be

used in conjunction with mouse lines that lack components of intracellular

Page 119: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

106

surveillance mechanisms, as well as mice that lack various inflammatory

cytokines to characterize the contribution of each in mediating the increased or

decreased vaccine efficacy of each strain. In the broader context, I had hoped

that this work and the tools built herein could help clarify the some of the

requirements of a live vaccine strain to elicit immunity against infection with F.

tularensis so that a future rationally designed vaccine strain could incorporate

and build upon the strengths and avoid the weaknesses of F. tularensis LVS.

Page 120: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

107

Figure IV.I Model of iron import in F. tularensis subsp. holarctica Live Vaccine Strain (LVS). The RML LVS feoB allele has a nucleotide substitution that changes the aspartate at position 471 to a tyrosine; this change decreases the ferrous iron import activity of FeoB in the RML LVS, which correlates with enhanced resistance to H2O2. Once in the cytosol, ferrous iron can likely be oxidized by and stored within the central cavity of the bacterioferritin complex.

Page 121: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

108

Figure IV.II Model of intracellular outcomes for high and low iron strains. RML LVS (left half of cell model) is more resistant to the bactericidal activity of IFNγ stimulated murine macrophages due in part to having less intracellular ferrous iron content. Strains that have excess ferrous iron, such as ATCC, are more sensitive to such bactericidal activities.

Page 122: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

109

APPENDIX A

DEVELOPMENT OF RECOMBINANT IGLC-LCMV EPITOPE T CELL

REPORTER FUSIONS

Rationale

While developing a mouse model of low and moderate dose immunization

with F. tularensis Schu S4 mutants with capsule and LPS defects, I sought to

create tools to facilitate the study of T cell responses of mice immunized within

this model. To do this I made plasmids that carried the Francisella iglC gene with

the coding sequence of either of two well-known epitopes (GP33-41 and GP61-80)

from the lymphocytic choriomeningitis virus, or LCMV242. IglC was chosen for C-

terminal fusion of the epitopes because it is among the most highly induced

genes within the macrophage environment and the protein is among those that

are T cell antigens in LVS immunized mice243,244.

The rationale for this approach was to use these tools with mouse lines

that are T cell receptor restricted to these specific epitopes to characterize T cell

proliferation and maturation after immunization with a F. tularensis strain, as well

as use the strength of mouse genetics to identify host factors that are necessary

for generation of a T cell response to F. tularensis. Additionally, as the immune

responses to these epitopes are well described, their use in F. tularensis during

vaccination may also help identify mechanisms of immune suppression that

interfere with the generation of a robust and long-lived cellular immune response,

and/or be useful in identifying correlates of immunity.

Page 123: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

110

Materials and methods

Bacterial strains and growth conditions: Iowa LVS (University of Iowa) and F.

tularensis Schu S4 and were routinely cultured on modified Mueller-Hinton

(supplemented with 1% glucose, 0.025% ferric pyrophosphate, and 2%

IsoVitaleX) agar or in broth, with 50 μg/mL of spectinomycin, as needed. Agar

plates were incubated at 37°C with humidity and 5% CO2 while broth cultures

were grown at 37°C, shaken at 200 rpm. Plasmids were electroporated into LVS

(2.5 kV, 25 μF, and 600 Ω), and the bacteria were plated onto MMH agar with 50

μg/mL spectinomycin after 2-3 hours of outgrowth.

Low and moderate dose mouse immunization and Schu S4 challenge: 6-8

week old female BALB/c mice were inoculated via the intranasal with either the

~500 CFU Iowa LVS, ~85 or 850 CFU F. tularensis waaY::Trgtn, or ~22 or 107

CFU wbtA::Tn5 that had been suspended in sterile PBS. Inoculum dose from

each strain was serially diluted and plated for enumeration. Mice were monitored

for signs of distress daily. Six weeks after immunization the mice were

challenged with either ~66 or 660 CFU of F. tularensis Schu S4 and monitored

for survival.

Recombinant IglC-LCMV epitope construction: The coding sequence of iglC

was PCR amplified and subsequently used as template in splice-overlap-

extension PCR (SOE-PCR) with large primers that included the 3’ end of iglC

and GP33-41 or GP61-80 that had been codon-optimized for the Francisella genome

Page 124: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

111

(http://www.jcat.de). All PCRs used the high-fidelity Phusion polyermase per the

manufacturer’s recommendations. The 5’ primers included a KpnI site and the

final 3’ primers included a SalI site. The SOE-PCR product was A-tailed using

Taq polyermase and cloned into pCR2.1. Inserted sequences were excised by

digestion with KpnI and SalI, then ligated into the respective sites of pTrc99a,

immediately downstream of the iglA promoter that had been cloned previously.

Similar plasmids were created with the groE promoter. The promoter-iglC

fragments were removed by digestion with BamHI and SalI and cloned into the

respective sites of the final vector, pBB103.

In vitro infections: HepG2 cells were cultivated in DMEM supplemented with

10% heat-inactivated fetal bovine serum (HI-FBS). Cells were enumerated and

seeded into 24 well dishes at a density of 3x105 per mL in DMEM with 10%

L929-conditioned media, 10% heat-inactivated FBS. Multiplicity of infection

(MOI) was estimated by measuring the OD600 value of mid to late-log phase

grown bacteria, and confirmed via serial dilution onto modified Mueller-Hinton

agar and enumeration after 2-3 days’ growth at 37°C with humidity, 5% CO2.

Bacteria were allowed to associate with the cells for 3 hours, after which time the

cells were washed 3x with PBS, and incubated in DMEM supplemented with HI-

FBS and 100 μg/mL of gentamicin for one hour, cells were then washed 3x with

PBS and either lysed or incubated in antibiotic-free media for a further 20 hours.

At the 4 and 24-hour time points 0.1% saponin was added to each well. Wells

Page 125: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

112

were scraped and vigorously pipetted to disrupt the cells and liberate intracellular

bacteria for serial dilution and enumeration.

T cell response to antigen ex vivo: Briefly, mice were infected intraperitoneally

with ~500 CFU of the Iowa LVS harboring either of the plasmids encoding the

IglC-LCMV epitope fusion and splenic lymphocytes were incubated with antigen

peptides, and the proportion of cells responding, as measured by interferon γ

production, were quantified via FACS protocol similar to Badovinac et. al245.

LCMV immunization and epitope-expressing LVS challenge: 6-8 week old

female BALB/c mice were inoculated intraperitoneally with 2.5x105 PFU LCMV.

Six weeks after inoculation, these mice and unimmunized controls were infected

via the intranasal route with either Iowa LVS carrying the control plasmid

expressing iglC alone, iglC-GP33-41, or iglC-GP61-80. Weight loss was monitored

daily and mice were humanely sacrificed if ≥25% of initial weight was lost.

Results and discussion

The low and moderate dose immunization experiment included Iowa LVS

as a control and a benchmark against which to compare the protection afforded

mice by intranasal inoculation with two F. tularensis Schu S4 mutants with

defects in capsule and LPS that our lab had previously characterized as

attenuated. Each group of mice was challenged with two doses of F. tularensis

Page 126: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

113

Schu S4 and the survival of each group was compared to that of the LVS

immunized group (Fig. A.1 and A.2). From the group immunized with low dose

waaY::trgtn or wbtA::Tn5, no mice survived challenge with F. tularensis Schu S4,

but 80% of the LVS immunized group survived 66 CFU, though only an increase

in time to death was observed when challenged with 660 CFU. Mice immunized

with higher doses of the mutants survived the 66 CFU challenge better, with 20%

and 60% survival in mice from the waaY::trgtn and wbtA::Tn5 groups,

respectively, though still not as well as the LVS immunized mice. Survival of the

moderate immunization dose group after challenge with 660 CFU was roughly

comparable to that of the control LVS group; in fact one wbtA::Tn5 immunized

mouse survived the challenge for the duration of the experiment, though it should

be noted that this individual was considerably larger than its littermates.

To begin to characterize the differences in T cell responses between LVS

and the F. tularensis Schu S4 capsule/O-antigen mutants, I built IglC-LCMV

epitope fusion constructs. Fusion of the LCMV epitopes to IglC resulted in

detectable upward shifts of the IglC signal in Western blots from E. coli, LVS, and

the LVS fevR::trgtn mutant, indicating that the fusion constructs were being

expressed specifically from the groE promoter; no signal would be predicted from

E. coli lacking the plasmids and very low signal is expected from the fevR

mutant. The signal strength appears to be weaker from proteins with the

epitopes, indicating that they may be inherently less stable or are made targets of

proteases with the additional amino acids. Importantly, the presence of the

protein fusions did not appear to alter the intracellular growth kinetics of LVS in

Page 127: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

114

HepG2 cells, suggesting that if the fusion proteins do negatively impact the FPI

function within the bacteria, it is not a dominant phenotype.

Mice infected with LVS expressing the fusion proteins generate a low level

response to IglC-GP33-41, as treatment of their lymphocytes ex vivo with purified

GP33-41 peptide stimulated only a four-fold increase in the number of interferon γ

positive cells relative to untreated cells. No GP61-80 response was detected in this

assay, suggesting that the decreased stability of the fusion protein may reduce

the protein levels to below a threshold where enough antigen is presented to be

detectable with this approach. It is also possible that the process of antigen

presentation is decreased or inhibited in vivo by F. tularensis as it is in vitro190.

To test the hypothesis that antigen-specific T cells can contribute to an

immune response to LVS expressing these epitopes, mice were given a sub-

lethal dose of LCMV that is known to stimulate cellular immunity. These mice

were challenged with the LVS strains approximately six weeks later and

monitored for weight loss and survival. Mice infected with the control LVS

expressing native IglC exhibited the most mortality, with only one mouse

surviving the intranasal challenge with ~6,000 CFU. In both naïve control mice

and LCMV-immunized mice there was a decrease in mortality when challenged

with both GP33-41 and GP61-80 expressing strains, indicating that the epitopes alter

the virulence of the bacteria, possibly as a result of the strength of the response

to the antigenic epitopes. It will be of interest to describe the events that occur in

naïve mice in response to these strains, as they may give insight into some of the

Page 128: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

115

host response pathways that may be engaged for the control of intracellular

Francisella, and thus may aid in identifying correlates of immunity.

Ultimately, these tools were built in an effort to dissect the host response

to attenuated strains of F. tularensis that could contribute towards a vaccine

against tularemia. These tools were intended to be integrated into the high and

moderate dose immunization experimental paradigm to compare the T cell

responses to F. tularensis in environments with differing levels of inflammation,

which is can affect the developmental fate of naïve T cells246.

Page 129: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

116

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post challenge

Low CFU immunization - 66 CFU Schu Challenge

Naïve - 19 CFU Schu (n=5)

LVS 500 CFU - 66 CFU Schu (n=5)

85 CFU waaY::Trgtn - 66 CFU Schu(n=5)22 CFU wbtA::Tn5 - 66 CFU Schu (n=5)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post challenge

Low CFU immunization - 660 CFU Schu Challenge

Naïve - 19 CFU Schu (n=5)

LVS 500 CFU - 660 CFU Schu (n=5)

85 CFU waaY::Trgtn - 660 CFU Schu (n=5)

22 CFU wbtA::Tn5 - 660 CFU Schu (n=5)

Figure A.I. Low dose immunization survival curves. Groups of mice were inoculated via the intranasal route with LVS or two different F. tularensis Schu S4 capsule/O-antigen mutants six weeks prior to challenge with two doses of wild type Schu S4, as per the materials and methods. All data are from one replicate.

Page 130: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

117

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14Perc

ent s

urvi

val

Days post challenge

High CFU immunization - 66 CFU Schu Challenge

Naïve - 19 CFU Schu (n=5)

LVS 500 CFU - 66 CFUSchu850 CFU waaY::Trgtn - 66CFU Schu (n=5)107 CFU wbtA::Tn5 - 66CFU Schu (n=5)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14

Perc

ent s

urvi

val

Days post challenge

High CFU immunization - High Schu Challenge

Naïve - 19 CFU Schu (n=5)

LVS 500 CFU - 660 CFU Schu

850 CFU waaY::Trgtn - 660 CFUSchu (n=4)107 CFU wbtA::Tn5 - 660 CFU Schu(n=4)

Figure A.II. High dose immunization survival curves. Groups of mice were inoculated via the intranasal route with LVS or two different F. tularensis Schu S4 capsule/O-antigen mutants six weeks prior to challenge with two doses of wild type Schu S4, as per the materials and methods. All data are from one replicate.

Page 131: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

118

1

100

LVS LVS C LVS C33 LVS C61

Fold

gro

wth

24 hour growth HepG2 cells

LVS

E. c

oli +

em

pty

vect

or

E. c

oli +

Pgr

o-ig

lC

E. c

oli +

Pgr

o-ig

lC-G

P33

E. c

oli +

Pgr

o-ig

lC-G

P61

LVS

LVS

fevR

::Trg

Tn

LVS

fevR

::Trg

Tn +

Pgr

o-ig

lC

LVS

fevR

::Trg

Tn +

Pgr

o-ig

lC-G

P33

LVS

fevR

::Trg

Tn +

Pgr

o-ig

lC-G

P61

LVS

+ P

gro-

iglC

LVS

+ P

gro-

iglC

-GP

33

LVS

+ P

gro-

iglC

-GP

61

Figure A.III. IglC-LCMV epitope fusions are expressed in LVS and do not alter intracellular growth. A) Plasmids with the coding sequence for expression of the IglC fusion proteins were transformed into E. coli, LVS, and the LVS fevR mutant. Lysates of each strain were subjected to Western blot and probed with the anti-IglC mAb. B) HepG2 cells were infected in vitro with either LVS or LVS carrying one of the recombinant IglC plasmids. LVS C = LVS expressing IglC with no epitope; LVS C33 = LVS expressing IglC-GP33-41; and LVS C61 = LVS expressing IglC-GP61-80. All data shown are from one replicate.

A

B

Page 132: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

119

CD4+ CD4+

IFNγ

IFNγ

CD8+ CD8+

Figure A.IV. LVS infected mouse lymphocyte response to LCMV antigens ex vivo. Naïve mice were infected via the intraperitoneal route with ~500 CFU of LVS expressing either IglC-GP33-41 or IglC-GP61-81. Splenic lymphocyte responses to purified antigens were measured seven days later, as described in the materials and methods. Daa are from one replicate.

Page 133: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

120

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12

Perc

ent s

urvi

val

Days post-infection

Naïve mice

LVS iglC (n=5)

LVS iglC-GP33 (n=4)

LVS iglC-GP61 (n=5)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 10 11 12

Perc

ent s

urvi

val

Days post infection

LCMV immunized

LCMV + LVS iglC (n=4)

LCMV + LVS iglC-GP33 (n=5)

LCMV + LVS iglC-GP61 (n=5)

Figure A.V. Survival of LCMV immunized mice challenged with LVS expressing LCMV epitopes. Mice were given 2.5x105 PFU of LCMV intraperitoneally and allowed to convalesce for six weeks, after which they were challenged by the intranasal route with ~6,000 CFU of LVS with or without epitopes fused to IglC, as described in the materials and methods. Data are from one replicate.

Page 134: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

121

APPENDIX B

OXYR DOES NOT INFLUENCE OXIDATIVE STRESS RESISTANCE IN F.

TULARENSIS LVS

Rationale

Analysis of the genomes of Francisella indicates that a well conserved

homolog of the LysR transcription factor OxyR can be found. OxyR is known to

be an allosteric regulator of genes in the oxidative stress resistance pathways of

many bacteria, changing its DNA binding ability upon oxidation of cysteine

residues by H2O2 and activating transcription of protective genes like catalase

and the alkylhydroperoxide reductase247-250. Additionally, a transposon screen of

F. novicida identified oxyR as a mutant with increased sensitivity to H2O2 and

decreased fitness in a Drosophila model of infection. For these reasons I

hypothesized that the F. tularensis homolog of OxyR may be important for

upregulation of H2O2 protective factors, thus I created a mutant in F. tularensis

LVS and tested its role in resistance to H2O2.

Materials and methods

Bacterial strains and growth conditions: RML LVS was routinely cultured on

modified Mueller-Hinton (supplemented with 1% glucose, 0.025% ferric

pyrophosphate, and 2% IsoVitaleX) agar or in broth, with 25 or 50 μg/mL of

kanamycin or spectinomycin as needed, respectively. Agar plates were

Page 135: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

122

incubated at 37°C with humidity and 5% CO2 while broth cultures were grown at

37°C, shaken at 200 rpm.

Mutant construction: Deletion of oxyR (FTL_1014) was achieved by

homologous recombination using derivatives of the non-replicating plasmid

pJC84. Briefly, upstream and downstream flanking DNA was amplified via PCR,

and amplicons were cloned into the multiple cloning site of pCR2.1 (Life

Technologies). A spectinomycin resistance cassette was cloned into the AvrII

site in the 3’ end of each upstream PCR fragment. The upstream-spectinomycin

resistance fragment was removed by digestion with AscI, and cloned into the

AscI site in the 5’ region of the downstream PCR plasmid. The entire upstream-

spectinomycin resistance-downstream fragment was cloned into the BamHI site

of the suicide plasmid pJC84. In contrast to the mutants made in chapters II and

II, the spectinomycin resistance cassette was not removed. Plasmids were

electroporated into LVS (2.5 kV, 25 μF, and 600 Ω), and the bacteria were plated

onto MMH agar with 50 μg/mL kanamycin after 2-3 hours of outgrowth.

Organisms were then grown overnight in broth lacking kanamycin, and were

plated onto MMH agar with 8% sucrose for sacB-mediated counter-selection.

Kanamycin sensitive colonies were screened by colony PCR to detect deletion of

the oxyR gene.

Hydrogen peroxide sensitivity: To measure resistance to H2O2-mediated

killing, mid- to late-log phase organisms were pelleted at 13,200 RPM for 5 min,

Page 136: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

123

washed in PBS, and ~106 CFU were resuspended into 200 μL PBS with or

without 100 μM fresh hydrogen peroxide (H2O2) in a 96-well dish. The samples

were incubated at 37°C with humidity and 5% CO2 for one hour. The culture

from each well was then serially diluted, plated onto modified Mueller-Hinton agar

(with antibiotic where necessary) and the number of surviving organisms for each

strain was enumerated after 2-3 days. This assay was also modified in two

ways to test for the production and/or secretion of an H2O2 protective factor into

the culture media. First, a standard quantity of bacteria were added to fresh

media and allowed to outgrow for one hour, followed by addition of 500 µM H2O2

directly to the culture, from which samples were removed and serially diluted

onto MMH agar every hour for five hours; and second, the ∆oxyR mutant was

suspended in spent media from wild type RML LVS or itself with or without 1 mM

H2O2 for 30 minutes before serial dilution onto MMH agar.

In vitro infections: J774.A16 cells were cultivated in DMEM supplemented with

10% heat-inactivated fetal bovine serum (HI-FBS). Cells were enumerated and

seeded into 24 well dishes at a density of 3x105 per mL in DMEM with 10%

L929-conditioned media, 10% heat-inactivated FBS. Multiplicity of infection

(MOI) was estimated by measuring the OD600 value of mid to late-log phase

grown bacteria, and confirmed via serial dilution onto modified Mueller-Hinton

agar and enumeration after 2-3 days’ growth at 37°C with humidity, 5% CO2.

Bacteria were allowed to associate with the cells for 3 hours, after which time the

cells were washed 3x with PBS, and incubated in DMEM supplemented with HI-

Page 137: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

124

FBS and 100 μg/mL of gentamicin for one hour, cells were then washed 3x with

PBS and either lysed or incubated in antibiotic-free media for a further 20 hours.

At the 4 and 24-hour time points 0.1% saponin was added to each well. Wells

were scraped and vigorously pipetted to disrupt the cells and liberate intracellular

bacteria for serial dilution and enumeration.

Results and discussion

The F. tularensis LVS OxyR protein shares 36% identity to that of E. coli

K12 and 35% identity to its homolog in Salmonella (via BLASt analysis),

significantly high values for such a phylogenetically distant organism. Such

conservation and the clear link of OxyR to oxidative stress resistance in many

bacteria, including other pathogens, made the F. tularensis LVS OxyR an

attractive target for further study. Furthermore, the oxyR gene is in a divergent

genomic orientation with the alkylhydroperoxide reductase gene ahpC, consistent

with the orientation of other LysR family genes, suggesting that this locus may

represent a functional unit in Francisella251. The mutant exhibits no obvious

growth defects in broth culture, though the colony size on an agar plate is smaller

than that of RML LVS; the significance of this is not clear. Quite surprisingly, the

∆oxyR mutant did not display increased H2O2 sensitivity in any of the three

assays that were used, though it did exhibit a very modest intracellular growth

defect in J774A.16 cells; its twenty-four hour fold growth was approximately 30%

that of the parental RML LVS.

Page 138: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

125

Multiple possibilities exist to explain why the ∆oxyR mutant was not more

sensitive to H2O2, though only further experimentation can confirm or rule them

out. The first possibility is that OxyR may be a molecular fossil and no longer

regulates genes involved in oxidative stress resistance. This could occur by

mutations that affect the ability of OxyR to be oxidized by H2O2, mutations that

affect its ability to bind DNA, or by mutations within the promoters of oxidative

stress resistance genes. This would mean that the genes previously regulated

by OxyR may be constitutively expressed, possibly to the bacteria’s benefit.

The second possibility is that the F. tularensis LVS OxyR responds to a

different stress than H2O2, such as superoxide or reactive nitrogen species.

Indeed, Binesse, et. al. report that while a F. tularensis Schu S4 ahpC mutant

was not more sensitive to H2O2, it did have increased sensitivity to paraquat and

the compound SIN-1, which ultimately generates peroxynitrite127. If the oxyR-

ahpC divergent gene locus is a functional LysR-like regulatory unit, then it is

possible that the lack of H2O2 sensitivity displayed by the F. tularensis LVS

∆oxyR mutant would be consistent with the data from Binesse, et. al. Preliminary

experiments with this mutant and SIN-1 are inconclusive and need optimization

as of the time of writing.

A third possibility is redundancy in transcription factors that regulate H2O2

protective genes. Many bacteria have multiple pathways that can overlap in

terms of the genes they activate or repress, including those for oxidative stress

resistance252. For example, the FTL_1634 ORF encodes a LysR-like

transcription factor that shares 42% identity with OxyR at the amino acid level. If

Page 139: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

126

this ORF is transcribed and encodes a functional homolog of OxyR, then it could

compensate and mask H2O2 associated phenotypes in the ∆oxyR mutant. There

are other, less conserved LysR-like genes encoded elsewhere on the

chromosome that could compensate for loss of OxyR, as well.

Page 140: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

127

020406080

100120

10 uM 50 uM 100 uM

Perc

ent s

urvi

val

H2O2 in PBS

RML

ΔoxyR

0.001

0.01

0.1

1

1 2 3 4 5

Perc

ent s

urvi

val

Hours post H2O2

500 uM H2O2 in broth

RML

ΔoxyR

0.1

1

10

MMH RCM OCM

Perc

ent s

urvi

val

1 mM H2O2 in conditioned media

A

B

C

Figure B.I. The RML LVS ∆oxyR mutant is as resistant to H2O2 as the wild type. A) The bacteria were exposed to three doses of H2O2 in PBS for one hour. B) Bacteria were sub-cultured into fresh media for one hour prior to addition of 500 µM H2O2. Samples were taken at one hour intervals. C) ∆oxyR was exposed to 1 mM H2O2 for thirty minutes in either fresh MMH broth or filtered spent media from the wild type RML LVS or the ∆oxyR mutant. MMH = modified Mueller-Hinton; RCM = RML LVS conditioned media; OCM = ∆oxyR conditioned media.

Page 141: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

128

0

20

40

60

80

100

120

RML ΔoxyR

Fold

gro

wth

nor

mal

ized

to

RM

L 24 hour growth in J774.A16 cells

Figure B.II. The ΔoxyR mutant has a mild intracellular growth defect in J774A.16 cells. Infections were performed as described in the materials and methods. Combined fold growths are shown from four independent experiments.

Page 142: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

129

REFERENCES 1 Tusnady, G. E. & Simon, I. The HMMTOP transmembrane topology

prediction server. Bioinformatics 17, 849-850 (2001). 2 Spyropoulos, I. C., Liakopoulos, T. D., Bagos, P. G. & Hamodrakas, S. J.

TMRPres2D: high quality visual representation of transmembrane protein models. Bioinformatics 20, 3258-3260, doi:10.1093/bioinformatics/bth358 (2004).

3 McCoy, G. W. & Chapin, C. W. Studies of plague, a plague-like disease,

and tuberculosis among rodents in California. I. A study of the virulence of cultures of Bacillus pestis of various sources and ages. II. The virulence of Bacillus pestis of ground squirrel origin. III. Immunity of wild rats (Mus norvegicus) to plague infection. IV. Susceptibility of a ground squirrel (Ammospermophilus leucurus, Merriam) to plague. V. Bacterium tularense the cause of a plague-like disease of rodents. VI. A note on the susceptibility of ground squirrels (Citellus beechyi, richardson) to tuberculosis. (Govt. print. off., 1912).

4 Wherry, W. B. & Lamb, B. H. Infection of man with Bacterium tularense.

1914. J Infect Dis 189, 1321-1329 (2004). 5 Sjostedt, A. Tularemia: history, epidemiology, pathogen physiology, and

clinical manifestations. Ann N Y Acad Sci 1105, 1-29, doi:10.1196/annals.1409.009 (2007).

6 Chapin, C. W. Review of Our Knowledge of Bacterium Tularense.

American journal of public health 11, 529-532 (1921). 7 Francis, E. Landmark article April 25, 1925: Tularemia. By Edward

Francis. Jama 250, 3216-3224 (1983). 8 Francis, E. & Lake, G. C. Transmission of Tularaemia by the Mouse Louse

Polyplax Serratus (Burm.). Public health reports 37, 96-101 (1922). 9 Francis, E. & Lake, G. C. Transmission of Tularaemia by the Bedbug,

Cimex Lectularius. Public health reports 37, 83-95, doi:Doi 10.2307/4576251 (1922).

10 McLendon, M. K., Apicella, M. A. & Allen, L. A. Francisella tularensis:

taxonomy, genetics, and Immunopathogenesis of a potential agent of biowarfare. Annu Rev Microbiol 60, 167-185, doi:10.1146/annurev.micro.60.080805.142126 (2006).

Page 143: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

130

11 Saslaw, S., Eigelsbach, H. T., Prior, J. A., Wilson, H. E. & Carhart, S. Tularemia vaccine study. II. Respiratory challenge. Archives of internal medicine 107, 702-714 (1961).

12 Saslaw, S., Eigelsbach, H. T., Wilson, H. E., Prior, J. A. & Carhart, S.

Tularemia vaccine study. I. Intracutaneous challenge. Archives of internal medicine 107, 689-701 (1961).

13 Harris, S. Japanese biological warfare research on humans: a case study

of microbiology and ethics. Ann N Y Acad Sci 666, 21-52 (1992). 14 Christopher, G. W., Cieslak, T. J., Pavlin, J. A. & Eitzen, E. M., Jr.

Biological warfare. A historical perspective. Jama 278, 412-417 (1997). 15 World Health Organization. Health aspects of chemical and biological

weapons. Report of a WHO Group of Consultants. (1970). 16 Kaufmann, A. F., Meltzer, M. I. & Schmid, G. P. The economic impact of a

bioterrorist attack: are prevention and postattack intervention programs justifiable? Emerg Infect Dis 3, 83-94, doi:10.3201/eid0302.970201 (1997).

17 Dennis, D. T. et al. Tularemia as a biological weapon: medical and public

health management. Jama 285, 2763-2773 (2001). 18 Foley, J. E. & Nieto, N. C. Tularemia. Veterinary microbiology 140, 332-

338, doi:10.1016/j.vetmic.2009.07.017 (2010). 19 Aktas, D. et al. Oropharyngeal Tularemia Outbreak Associated with

Drinking Contaminated Tap Water, Turkey, July-September 2013. Emerg Infect Dis 21, 2194-2196, doi:10.3201/eid2112.142032 (2015).

20 McCrumb, F. R. Aerosol Infection of Man with Pasteurella Tularensis.

Bacteriol Rev 25, 262-267 (1961). 21 Grunow, R., Kalaveshi, A., Kuhn, A., Mulliqi-Osmani, G. & Ramadani, N.

Surveillance of tularaemia in Kosovo, 2001 to 2010. Euro Surveill 17 (2012).

22 Syrjala, H., Kujala, P., Myllyla, V. & Salminen, A. Airborne transmission of

tularemia in farmers. Scand J Infect Dis 17, 371-375 (1985). 23 Teutsch, S. M. et al. Pneumonic tularemia on Martha's Vineyard. N Engl J

Med 301, 826-828, doi:10.1056/NEJM197910113011507 (1979).

Page 144: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

131

24 Feldman, K. A. et al. An outbreak of primary pneumonic tularemia on Martha's Vineyard. N Engl J Med 345, 1601-1606, doi:10.1056/NEJMoa011374 (2001).

25 Johansson, A. et al. An outbreak of respiratory tularemia caused by

diverse clones of Francisella tularensis. Clin Infect Dis 59, 1546-1553, doi:10.1093/cid/ciu621 (2014).

26 Larsson, P. et al. The complete genome sequence of Francisella

tularensis, the causative agent of tularemia. Nat Genet 37, 153-159, doi:10.1038/ng1499 (2005).

27 Brett, M. E. et al. Outbreak of Francisella novicida bacteremia among

inmates at a louisiana correctional facility. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 59, 826-833, doi:10.1093/cid/ciu430 (2014).

28 Olsufjev, N. G. & Emelyanova, O. S. Immunological relationships between

Old and New World varieties of tularaemic bacteria. J Hyg Epidemiol Microbiol Immunol 7, 178-187 (1963).

29 Vogler, A. J. et al. Phylogeography of Francisella tularensis: global

expansion of a highly fit clone. Journal of bacteriology 191, 2474-2484, doi:10.1128/JB.01786-08 (2009).

30 Farlow, J. et al. Francisella tularensis in the United States. Emerging

infectious diseases 11, 1835-1841, doi:10.3201/eid1112.050728 (2005). 31 Johansson, A. et al. Worldwide genetic relationships among Francisella

tularensis isolates determined by multiple-locus variable-number tandem repeat analysis. Journal of bacteriology 186, 5808-5818, doi:10.1128/JB.186.17.5808-5818.2004 (2004).

32 Kugeler, K. J. et al. Molecular Epidemiology of Francisella tularensis in the

United States. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 48, 863-870, doi:10.1086/597261 (2009).

33 Molins, C. R. et al. Virulence difference between the prototypic Schu S4

strain (A1a) and Francisella tularensis A1a, A1b, A2 and type B strains in a murine model of infection. BMC infectious diseases 14, 67, doi:10.1186/1471-2334-14-67 (2014).

34 Keim, P., Johansson, A. & Wagner, D. M. Molecular epidemiology,

evolution, and ecology of Francisella. Ann N Y Acad Sci 1105, 30-66, doi:10.1196/annals.1409.011 (2007).

Page 145: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

132

35 Taylor, J. P. et al. Epidemiologic characteristics of human tularemia in the

southwest-central states, 1981-1987. Am J Epidemiol 133, 1032-1038 (1991).

36 Hopla, C. E. The ecology of tularemia. Advances in veterinary science and

comparative medicine 18, 25-53 (1974). 37 Hopla, C. E. Experimental studies on tick transmission of tularemia

organisms. Am J Hyg 58, 101-118 (1953). 38 Morner, T. The ecology of tularaemia. Rev Sci Tech 11, 1123-1130

(1992). 39 Abd, H., Johansson, T., Golovliov, I., Sandstrom, G. & Forsman, M.

Survival and growth of Francisella tularensis in Acanthamoeba castellanii. Applied and environmental microbiology 69, 600-606 (2003).

40 Ellis, J., Oyston, P. C., Green, M. & Titball, R. W. Tularemia. Clin Microbiol

Rev 15, 631-646 (2002). 41 Rohmer, L. et al. Comparison of Francisella tularensis genomes reveals

evolutionary events associated with the emergence of human pathogenic strains. Genome biology 8, R102, doi:10.1186/gb-2007-8-6-r102 (2007).

42 Kingry, L. C. & Petersen, J. M. Comparative review of Francisella

tularensis and Francisella novicida. Front Cell Infect Microbiol 4, 35, doi:10.3389/fcimb.2014.00035 (2014).

43 Nano, F. E. et al. A Francisella tularensis pathogenicity island required for

intramacrophage growth. J Bacteriol 186, 6430-6436, doi:10.1128/JB.186.19.6430-6436.2004 (2004).

44 Ludu, J. S. et al. The Francisella pathogenicity island protein PdpD is

required for full virulence and associates with homologues of the type VI secretion system. J Bacteriol 190, 4584-4595, doi:10.1128/JB.00198-08 (2008).

45 Brotcke, A. et al. Identification of MglA-regulated genes reveals novel

virulence factors in Francisella tularensis. Infection and immunity 74, 6642-6655, doi:10.1128/IAI.01250-06 (2006).

46 Maurelli, A. T., Fernandez, R. E., Bloch, C. A., Rode, C. K. & Fasano, A.

"Black holes" and bacterial pathogenicity: a large genomic deletion that enhances the virulence of Shigella spp. and enteroinvasive Escherichia coli. Proc Natl Acad Sci U S A 95, 3943-3948 (1998).

Page 146: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

133

47 Parkhill, J. et al. Comparative analysis of the genome sequences of

Bordetella pertussis, Bordetella parapertussis and Bordetella bronchiseptica. Nat Genet 35, 32-40, doi:10.1038/ng1227 (2003).

48 Bliven, K. A. & Maurelli, A. T. Antivirulence genes: insights into pathogen

evolution through gene loss. Infect Immun 80, 4061-4070, doi:10.1128/IAI.00740-12 (2012).

49 Moore, R. A. et al. Contribution of gene loss to the pathogenic evolution of

Burkholderia pseudomallei and Burkholderia mallei. Infect Immun 72, 4172-4187, doi:10.1128/IAI.72.7.4172-4187.2004 (2004).

50 Rohmer, L. et al. Potential source of Francisella tularensis live vaccine

strain attenuation determined by genome comparison. Infect Immun 74, 6895-6906, doi:10.1128/IAI.01006-06 (2006).

51 Salomonsson, E. et al. Reintroduction of two deleted virulence loci

restores full virulence to the live vaccine strain of Francisella tularensis. Infect Immun 77, 3424-3431, doi:10.1128/IAI.00196-09 (2009).

52 Costa, T. R. et al. Secretion systems in Gram-negative bacteria: structural

and mechanistic insights. Nat Rev Microbiol 13, 343-359, doi:10.1038/nrmicro3456 (2015).

53 Michalska, M. & Wolf, P. Pseudomonas Exotoxin A: optimized by

evolution for effective killing. Front Microbiol 6, 963, doi:10.3389/fmicb.2015.00963 (2015).

54 Qin, A. & Mann, B. J. Identification of transposon insertion mutants of

Francisella tularensis tularensis strain Schu S4 deficient in intracellular replication in the hepatic cell line HepG2. BMC Microbiol 6, 69, doi:10.1186/1471-2180-6-69 (2006).

55 Weiss, D. S. et al. In vivo negative selection screen identifies genes

required for Francisella virulence. Proc Natl Acad Sci U S A 104, 6037-6042, doi:10.1073/pnas.0609675104 (2007).

56 Lindemann, S. R. et al. Francisella tularensis Schu S4 O-antigen and

capsule biosynthesis gene mutants induce early cell death in human macrophages. Infect Immun 79, 581-594, doi:10.1128/IAI.00863-10 (2011).

57 Golovliov, I., Ericsson, M., Sandstrom, G., Tarnvik, A. & Sjostedt, A.

Identification of proteins of Francisella tularensis induced during growth in

Page 147: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

134

macrophages and cloning of the gene encoding a prominently induced 23-kilodalton protein. Infect Immun 65, 2183-2189 (1997).

58 Gray, C. G., Cowley, S. C., Cheung, K. K. & Nano, F. E. The identification

of five genetic loci of Francisella novicida associated with intracellular growth. FEMS Microbiol Lett 215, 53-56 (2002).

59 Golovliov, I., Sjostedt, A., Mokrievich, A. & Pavlov, V. A method for allelic

replacement in Francisella tularensis. FEMS Microbiol Lett 222, 273-280 (2003).

60 Telepnev, M., Golovliov, I., Grundstrom, T., Tarnvik, A. & Sjostedt, A.

Francisella tularensis inhibits Toll-like receptor-mediated activation of intracellular signalling and secretion of TNF-alpha and IL-1 from murine macrophages. Cell Microbiol 5, 41-51 (2003).

61 Telepnev, M., Golovliov, I. & Sjostedt, A. Francisella tularensis LVS

initially activates but subsequently down-regulates intracellular signaling and cytokine secretion in mouse monocytic and human peripheral blood mononuclear cells. Microbial pathogenesis 38, 239-247, doi:10.1016/j.micpath.2005.02.003 (2005).

62 Pukatzki, S. et al. Identification of a conserved bacterial protein secretion

system in Vibrio cholerae using the Dictyostelium host model system. Proc Natl Acad Sci U S A 103, 1528-1533, doi:10.1073/pnas.0510322103 (2006).

63 Hachani, A. et al. Type VI secretion system in Pseudomonas aeruginosa:

secretion and multimerization of VgrG proteins. J Biol Chem 286, 12317-12327, doi:10.1074/jbc.M110.193045 (2011).

64 Cianfanelli, F. R., Monlezun, L. & Coulthurst, S. J. Aim, Load, Fire: The

Type VI Secretion System, a Bacterial Nanoweapon. Trends Microbiol 24, 51-62, doi:10.1016/j.tim.2015.10.005 (2016).

65 de Bruin, O. M., Ludu, J. S. & Nano, F. E. The Francisella pathogenicity

island protein IglA localizes to the bacterial cytoplasm and is needed for intracellular growth. BMC Microbiol 7, 1, doi:10.1186/1471-2180-7-1 (2007).

66 Barker, J. R. et al. The Francisella tularensis pathogenicity island encodes

a secretion system that is required for phagosome escape and virulence. Mol Microbiol 74, 1459-1470 (2009).

67 Broms, J. E., Lavander, M., Meyer, L. & Sjostedt, A. IglG and IglI of the

Francisella pathogenicity island are important virulence determinants of

Page 148: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

135

Francisella tularensis LVS. Infect Immun 79, 3683-3696, doi:10.1128/IAI.01344-10 (2011).

68 Broms, J. E., Meyer, L., Sun, K., Lavander, M. & Sjostedt, A. Unique

substrates secreted by the type VI secretion system of Francisella tularensis during intramacrophage infection. PLoS One 7, e50473, doi:10.1371/journal.pone.0050473 (2012).

69 Hare, R. F. & Hueffer, K. Francisella novicida pathogenicity island

encoded proteins were secreted during infection of macrophage-like cells. PLoS One 9, e105773, doi:10.1371/journal.pone.0105773 (2014).

70 Clemens, D. L., Ge, P., Lee, B. Y., Horwitz, M. A. & Zhou, Z. H. Atomic

structure of T6SS reveals interlaced array essential to function. Cell 160, 940-951, doi:10.1016/j.cell.2015.02.005 (2015).

71 Nano, F. E. & Schmerk, C. The Francisella pathogenicity island. Ann N Y

Acad Sci 1105, 122-137, doi:10.1196/annals.1409.000 (2007). 72 Long, M. E., Lindemann, S. R., Rasmussen, J. A., Jones, B. D. & Allen, L.

A. Disruption of Francisella tularensis Schu S4 iglI, iglJ, and pdpC genes results in attenuation for growth in human macrophages and in vivo virulence in mice and reveals a unique phenotype for pdpC. Infect Immun 81, 850-861, doi:10.1128/IAI.00822-12 (2013).

73 Baron, G. S. & Nano, F. E. MglA and MglB are required for the

intramacrophage growth of Francisella novicida. Mol Microbiol 29, 247-259 (1998).

74 Brotcke, A. & Monack, D. M. Identification of fevR, a novel regulator of

virulence gene expression in Francisella novicida. Infect Immun 76, 3473-3480, doi:10.1128/IAI.00430-08 (2008).

75 Bonquist, L., Lindgren, H., Golovliov, I., Guina, T. & Sjostedt, A. MglA and

Igl proteins contribute to the modulation of Francisella tularensis live vaccine strain-containing phagosomes in murine macrophages. Infect Immun 76, 3502-3510, doi:10.1128/IAI.00226-08 (2008).

76 Charity, J. C., Blalock, L. T., Costante-Hamm, M. M., Kasper, D. L. &

Dove, S. L. Small molecule control of virulence gene expression in Francisella tularensis. PLoS Pathog 5, e1000641, doi:10.1371/journal.ppat.1000641 (2009).

77 Buchan, B. W., McCaffrey, R. L., Lindemann, S. R., Allen, L. A. & Jones,

B. D. Identification of migR, a regulatory element of the Francisella tularensis live vaccine strain iglABCD virulence operon required for normal

Page 149: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

136

replication and trafficking in macrophages. Infect Immun 77, 2517-2529, doi:10.1128/IAI.00229-09 (2009).

78 Faron, M. et al. The Francisella tularensis migR, trmE, and cphA genes

contribute to F. tularensis pathogenicity island gene regulation and intracellular growth by modulation of the stress alarmone ppGpp. Infect Immun 81, 2800-2811, doi:10.1128/IAI.00073-13 (2013).

79 Lenco, J. et al. Proteomics analysis of the Francisella tularensis LVS

response to iron restriction: induction of the F. tularensis pathogenicity island proteins IglABC. FEMS Microbiol Lett 269, 11-21, doi:10.1111/j.1574-6968.2006.00595.x (2007).

80 Buchan, B. W., McLendon, M. K. & Jones, B. D. Identification of

differentially regulated francisella tularensis genes by use of a newly developed Tn5-based transposon delivery system. Appl Environ Microbiol 74, 2637-2645, doi:10.1128/AEM.02882-07 (2008).

81 Jones, B. D., Faron, M., Rasmussen, J. A. & Fletcher, J. R. Uncovering

the components of the Francisella tularensis virulence stealth strategy. Frontiers in cellular and infection microbiology 4, 32, doi:10.3389/fcimb.2014.00032 (2014).

82 Vinogradov, E., Perry, M. B. & Conlan, J. W. Structural analysis of

Francisella tularensis lipopolysaccharide. Eur J Biochem 269, 6112-6118 (2002).

83 Thomas, R. M. et al. The immunologically distinct O antigens from

Francisella tularensis subspecies tularensis and Francisella novicida are both virulence determinants and protective antigens. Infect Immun 75, 371-378, doi:10.1128/IAI.01241-06 (2007).

84 Apicella, M. A. et al. Identification, characterization and immunogenicity of

an O-antigen capsular polysaccharide of Francisella tularensis. PLoS One 5, e11060, doi:10.1371/journal.pone.0011060 (2010).

85 Beutler, B. & Poltorak, A. Positional cloning of Lps, and the general role of

toll-like receptors in the innate immune response. Eur Cytokine Netw 11, 143-152 (2000).

86 Miller, S. I., Ernst, R. K. & Bader, M. W. LPS, TLR4 and infectious disease

diversity. Nat Rev Microbiol 3, 36-46, doi:10.1038/nrmicro1068 (2005). 87 Barker, J. H., Weiss, J., Apicella, M. A. & Nauseef, W. M. Basis for the

failure of Francisella tularensis lipopolysaccharide to prime human

Page 150: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

137

polymorphonuclear leukocytes. Infect Immun 74, 3277-3284, doi:10.1128/IAI.02011-05 (2006).

88 Hajjar, A. M. et al. Lack of in vitro and in vivo recognition of Francisella

tularensis subspecies lipopolysaccharide by Toll-like receptors. Infection and immunity 74, 6730-6738, doi:10.1128/IAI.00934-06 (2006).

89 Walters, K. A. et al. Prior infection with Type A Francisella tularensis

antagonizes the pulmonary transcriptional response to an aerosolized Toll-like receptor 4 agonist. BMC Genomics 16, 874, doi:10.1186/s12864-015-2022-2 (2015).

90 Rasmussen, J. A. et al. Francisella tularensis Schu S4 lipopolysaccharide

core sugar and O-antigen mutants are attenuated in a mouse model of tularemia. Infect Immun 82, 1523-1539, doi:10.1128/IAI.01640-13 (2014).

91 Rasmussen, J. A., Fletcher, J. R., Long, M. E., Allen, L. A. & Jones, B. D.

Characterization of Francisella tularensis Schu S4 mutants identified from a transposon library screened for O-antigen and capsule deficiencies. Front Microbiol 6, 338, doi:10.3389/fmicb.2015.00338 (2015).

92 Crosa, L. M., Crosa, J. H. & Heffron, F. Iron transport in Francisella in the

absence of a recognizable TonB protein still requires energy generated by the proton motive force. Biometals 22, 337-344, doi:10.1007/s10534-008-9170-7 (2009).

93 Ramakrishnan, G. & Sen, B. The FupA/B protein uniquely facilitates

transport of ferrous iron and siderophore-associated ferric iron across the outer membrane of Francisella tularensis live vaccine strain. Microbiology 160, 446-457, doi:10.1099/mic.0.072835-0 (2014).

94 Perez, N. M. & Ramakrishnan, G. The reduced genome of the Francisella

tularensis live vaccine strain (LVS) encodes two iron acquisition systems essential for optimal growth and virulence. PLoS One 9, e93558, doi:10.1371/journal.pone.0093558 (2014).

95 Ramakrishnan, G., Meeker, A. & Dragulev, B. fslE is necessary for

siderophore-mediated iron acquisition in Francisella tularensis Schu S4. J Bacteriol 190, 5353-5361, doi:10.1128/JB.00181-08 (2008).

96 Thomas-Charles, C. A. et al. FeoB-mediated uptake of iron by Francisella

tularensis. Infection and immunity 81, 2828-2837, doi:10.1128/IAI.00170-13 (2013).

97 Sullivan, J. T., Jeffery, E. F., Shannon, J. D. & Ramakrishnan, G.

Characterization of the siderophore of Francisella tularensis and role of

Page 151: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

138

fslA in siderophore production. J Bacteriol 188, 3785-3795, doi:10.1128/JB.00027-06 (2006).

98 Deng, K., Blick, R. J., Liu, W. & Hansen, E. J. Identification of Francisella

tularensis genes affected by iron limitation. Infection and immunity 74, 4224-4236, doi:10.1128/IAI.01975-05 (2006).

99 Kiss, K., Liu, W., Huntley, J. F., Norgard, M. V. & Hansen, E. J.

Characterization of fig operon mutants of Francisella novicida U112. FEMS Microbiol Lett 285, 270-277, doi:10.1111/j.1574-6968.2008.01237.x (2008).

100 Perez, N., Johnson, R., Sen, B. & Ramakrishnan, G. Two parallel

pathways for ferric and ferrous iron acquisition support growth and virulence of the intracellular pathogen Francisella tularensis Schu S4. Microbiologyopen, doi:10.1002/mbo3.342 (2016).

101 Twine, S. et al. A mutant of Francisella tularensis strain SCHU S4 lacking

the ability to express a 58-kilodalton protein is attenuated for virulence and is an effective live vaccine. Infection and immunity 73, 8345-8352, doi:10.1128/IAI.73.12.8345-8352.2005 (2005).

102 Lindgren, H. et al. The 58-kilodalton major virulence factor of Francisella

tularensis is required for efficient utilization of iron. Infection and immunity 77, 4429-4436, doi:10.1128/IAI.00702-09 (2009).

103 Ramakrishnan, G., Sen, B. & Johnson, R. Paralogous outer membrane

proteins mediate uptake of different forms of iron and synergistically govern virulence in Francisella tularensis tularensis. J Biol Chem 287, 25191-25202, doi:10.1074/jbc.M112.371856 (2012).

104 Cartron, M. L., Maddocks, S., Gillingham, P., Craven, C. J. & Andrews, S.

C. Feo--transport of ferrous iron into bacteria. Biometals : an international journal on the role of metal ions in biology, biochemistry, and medicine 19, 143-157, doi:10.1007/s10534-006-0003-2 (2006).

105 Andrews, S. C., Robinson, A. K. & Rodriguez-Quinones, F. Bacterial iron

homeostasis. FEMS microbiology reviews 27, 215-237 (2003). 106 Hantke, K. Is the bacterial ferrous iron transporter FeoB a living fossil?

Trends in microbiology 11, 192-195 (2003). 107 Marlovits, T. C., Haase, W., Herrmann, C., Aller, S. G. & Unger, V. M. The

membrane protein FeoB contains an intramolecular G protein essential for Fe(II) uptake in bacteria. Proceedings of the National Academy of

Page 152: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

139

Sciences of the United States of America 99, 16243-16248, doi:10.1073/pnas.242338299 (2002).

108 Kim, H., Lee, H. & Shin, D. The FeoA protein is necessary for the FeoB

transporter to import ferrous iron. Biochemical and biophysical research communications 423, 733-738, doi:10.1016/j.bbrc.2012.06.027 (2012).

109 Lau, C. K., Ishida, H., Liu, Z. & Vogel, H. J. Solution structure of

Escherichia coli FeoA and its potential role in bacterial ferrous iron transport. Journal of bacteriology 195, 46-55, doi:10.1128/JB.01121-12 (2013).

110 Su, Y. C. et al. Structure of Stenotrophomonas maltophilia FeoA

complexed with zinc: a unique prokaryotic SH3-domain protein that possibly acts as a bacterial ferrous iron-transport activating factor. Acta crystallographica. Section F, Structural biology and crystallization communications 66, 636-642, doi:10.1107/S1744309110013941 (2010).

111 Weaver, E. A., Wyckoff, E. E., Mey, A. R., Morrison, R. & Payne, S. M.

FeoA and FeoC are essential components of the Vibrio cholerae ferrous iron uptake system, and FeoC interacts with FeoB. Journal of bacteriology 195, 4826-4835, doi:10.1128/JB.00738-13 (2013).

112 Nagy, T. A., Moreland, S. M. & Detweiler, C. S. Salmonella acquires

ferrous iron from haemophagocytic macrophages. Molecular microbiology 93, 1314-1326, doi:10.1111/mmi.12739 (2014).

113 Cassat, J. E. & Skaar, E. P. Iron in infection and immunity. Cell host &

microbe 13, 509-519, doi:10.1016/j.chom.2013.04.010 (2013). 114 Aranda, J. et al. Contribution of the FeoB transporter to Streptococcus

suis virulence. International microbiology : the official journal of the Spanish Society for Microbiology 12, 137-143 (2009).

115 Velayudhan, J. et al. Iron acquisition and virulence in Helicobacter pylori: a

major role for FeoB, a high-affinity ferrous iron transporter. Molecular microbiology 37, 274-286 (2000).

116 Robey, M. & Cianciotto, N. P. Legionella pneumophila feoAB promotes

ferrous iron uptake and intracellular infection. Infection and immunity 70, 5659-5669 (2002).

117 Naikare, H., Palyada, K., Panciera, R., Marlow, D. & Stintzi, A. Major role

for FeoB in Campylobacter jejuni ferrous iron acquisition, gut colonization, and intracellular survival. Infection and immunity 74, 5433-5444, doi:10.1128/IAI.00052-06 (2006).

Page 153: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

140

118 Lindgren, H., Lindgren, L., Golovliov, I. & Sjostedt, A. Mechanisms of

heme utilization by Francisella tularensis. PloS one 10, e0119143, doi:10.1371/journal.pone.0119143 (2015).

119 Imlay, J. A. The molecular mechanisms and physiological consequences

of oxidative stress: lessons from a model bacterium. Nat Rev Microbiol 11, 443-454, doi:10.1038/nrmicro3032 (2013).

120 Crichton, R. R. & Pierre, J. L. Old iron, young copper: from Mars to Venus.

Biometals : an international journal on the role of metal ions in biology, biochemistry, and medicine 14, 99-112 (2001).

121 Ramond, E. et al. Glutamate utilization couples oxidative stress defense

and the tricarboxylic acid cycle in Francisella phagosomal escape. PLoS pathogens 10, e1003893, doi:10.1371/journal.ppat.1003893 (2014).

122 Lindgren, H. et al. Iron content differs between Francisella tularensis

subspecies tularensis and subspecies holarctica strains and correlates to their susceptibility to H(2)O(2)-induced killing. Infection and immunity 79, 1218-1224, doi:10.1128/IAI.01116-10 (2011).

123 Bakshi, C. S. et al. Superoxide dismutase B gene (sodB)-deficient mutants

of Francisella tularensis demonstrate hypersensitivity to oxidative stress and attenuated virulence. Journal of bacteriology 188, 6443-6448, doi:10.1128/JB.00266-06 (2006).

124 Llewellyn, A. C., Jones, C. L., Napier, B. A., Bina, J. E. & Weiss, D. S.

Macrophage replication screen identifies a novel Francisella hydroperoxide resistance protein involved in virulence. PloS one 6, e24201, doi:10.1371/journal.pone.0024201 (2011).

125 Melillo, A. A. et al. Identification of Francisella tularensis live vaccine strain

CuZn superoxide dismutase as critical for resistance to extracellularly generated reactive oxygen species. Journal of bacteriology 191, 6447-6456, doi:10.1128/JB.00534-09 (2009).

126 Melillo, A. A., Bakshi, C. S. & Melendez, J. A. Francisella tularensis

antioxidants harness reactive oxygen species to restrict macrophage signaling and cytokine production. The Journal of biological chemistry 285, 27553-27560, doi:10.1074/jbc.M110.144394 (2010).

127 Binesse, J., Lindgren, H., Lindgren, L., Conlan, W. & Sjostedt, A. Roles of

reactive oxygen species-degrading enzymes of Francisella tularensis SCHU S4. Infect Immun 83, 2255-2263, doi:10.1128/IAI.02488-14 (2015).

Page 154: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

141

128 Shakerley, N. L., Chandrasekaran, A., Trebak, M., Miller, B. A. & Melendez, J. A. Francisella tularensis catalase restricts immune function by impairing TRPM2 channel activity. The Journal of biological chemistry, doi:10.1074/jbc.M115.706879 (2015).

129 Rabadi, S. M. et al. Antioxidant defenses of Francisella tularensis

modulate macrophage function and production of proinflammatory cytokines. The Journal of biological chemistry, doi:10.1074/jbc.M115.681478 (2015).

130 Anthony, L. D., Burke, R. D. & Nano, F. E. Growth of Francisella spp. in

rodent macrophages. Infection and immunity 59, 3291-3296 (1991). 131 Ben Nasr, A. et al. Critical role for serum opsonins and complement

receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in phagocytosis of Francisella tularensis by human dendritic cells (DC): uptake of Francisella leads to activation of immature DC and intracellular survival of the bacteria. Journal of leukocyte biology 80, 774-786, doi:10.1189/jlb.1205755 (2006).

132 Buddingh, G. J. & Womack, F. C. Observations on the Infection of Chick

Embryos with Bacterium Tularense, Brucella, and Pasteurella Pestis. The Journal of experimental medicine 74, 213-222 (1941).

133 Lindemann, S. R., McLendon, M. K., Apicella, M. A. & Jones, B. D. An in

vitro model system used to study adherence and invasion of Francisella tularensis live vaccine strain in nonphagocytic cells. Infection and immunity 75, 3178-3182, doi:10.1128/IAI.01811-06 (2007).

134 Faron, M., Fletcher, J. R., Rasmussen, J. A., Apicella, M. A. & Jones, B.

D. Interactions of Francisella tularensis with Alveolar Type II Epithelial Cells and the Murine Respiratory Epithelium. PLoS One 10, e0127458, doi:10.1371/journal.pone.0127458 (2015).

135 McCaffrey, R. L. & Allen, L. A. Francisella tularensis LVS evades killing by

human neutrophils via inhibition of the respiratory burst and phagosome escape. J Leukoc Biol 80, 1224-1230, doi:10.1189/jlb.0406287 (2006).

136 Hall, J. D. et al. Infected-host-cell repertoire and cellular response in the

lung following inhalation of Francisella tularensis Schu S4, LVS, or U112. Infect Immun 76, 5843-5852, doi:10.1128/IAI.01176-08 (2008).

137 Celli, J. & Zahrt, T. C. Mechanisms of Francisella tularensis intracellular

pathogenesis. Cold Spring Harb Perspect Med 3, a010314, doi:10.1101/cshperspect.a010314 (2013).

Page 155: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

142

138 Geier, H. & Celli, J. Phagocytic receptors dictate phagosomal escape and intracellular proliferation of Francisella tularensis. Infect Immun 79, 2204-2214, doi:10.1128/IAI.01382-10 (2011).

139 Clemens, D. L., Lee, B. Y. & Horwitz, M. A. Francisella tularensis enters

macrophages via a novel process involving pseudopod loops. Infect Immun 73, 5892-5902, doi:10.1128/IAI.73.9.5892-5902.2005 (2005).

140 Bradburne, C. E. et al. Temporal transcriptional response during infection

of type II alveolar epithelial cells with Francisella tularensis live vaccine strain (LVS) supports a general host suppression and bacterial uptake by macropinocytosis. J Biol Chem 288, 10780-10791, doi:10.1074/jbc.M112.362178 (2013).

141 Chong, A. et al. The early phagosomal stage of Francisella tularensis

determines optimal phagosomal escape and Francisella pathogenicity island protein expression. Infect Immun 76, 5488-5499, doi:10.1128/IAI.00682-08 (2008).

142 Golovliov, I., Baranov, V., Krocova, Z., Kovarova, H. & Sjostedt, A. An

attenuated strain of the facultative intracellular bacterium Francisella tularensis can escape the phagosome of monocytic cells. Infection and immunity 71, 5940-5950 (2003).

143 Clemens, D. L., Lee, B. Y. & Horwitz, M. A. Virulent and avirulent strains

of Francisella tularensis prevent acidification and maturation of their phagosomes and escape into the cytoplasm in human macrophages. Infect Immun 72, 3204-3217, doi:10.1128/IAI.72.6.3204-3217.2004 (2004).

144 Checroun, C., Wehrly, T. D., Fischer, E. R., Hayes, S. F. & Celli, J.

Autophagy-mediated reentry of Francisella tularensis into the endocytic compartment after cytoplasmic replication. Proc Natl Acad Sci U S A 103, 14578-14583, doi:10.1073/pnas.0601838103 (2006).

145 Lai, X. H., Golovliov, I. & Sjostedt, A. Francisella tularensis induces

cytopathogenicity and apoptosis in murine macrophages via a mechanism that requires intracellular bacterial multiplication. Infect Immun 69, 4691-4694, doi:10.1128/IAI.69.7.4691-4694.2001 (2001).

146 Lai, X. H. & Sjostedt, A. Delineation of the molecular mechanisms of

Francisella tularensis-induced apoptosis in murine macrophages. Infect Immun 71, 4642-4646 (2003).

Page 156: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

143

147 Schulert, G. S. & Allen, L. A. Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor. J Leukoc Biol 80, 563-571, doi:10.1189/jlb.0306219 (2006).

148 Ben Nasr, A. & Klimpel, G. R. Subversion of complement activation at the

bacterial surface promotes serum resistance and opsonophagocytosis of Francisella tularensis. J Leukoc Biol 84, 77-85, doi:10.1189/jlb.0807526 (2008).

149 Clay, C. D., Soni, S., Gunn, J. S. & Schlesinger, L. S. Evasion of

complement-mediated lysis and complement C3 deposition are regulated by Francisella tularensis lipopolysaccharide O antigen. J Immunol 181, 5568-5578 (2008).

150 Schwartz, J. T. et al. Natural IgM mediates complement-dependent uptake

of Francisella tularensis by human neutrophils via complement receptors 1 and 3 in nonimmune serum. J Immunol 189, 3064-3077, doi:10.4049/jimmunol.1200816 (2012).

151 Dai, S., Rajaram, M. V., Curry, H. M., Leander, R. & Schlesinger, L. S.

Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis. PLoS Pathog 9, e1003114, doi:10.1371/journal.ppat.1003114 (2013).

152 Santic, M., Molmeret, M., Klose, K. E., Jones, S. & Kwaik, Y. A. The

Francisella tularensis pathogenicity island protein IglC and its regulator MglA are essential for modulating phagosome biogenesis and subsequent bacterial escape into the cytoplasm. Cell Microbiol 7, 969-979, doi:10.1111/j.1462-5822.2005.00526.x (2005).

153 Nauseef, W. M. Assembly of the phagocyte NADPH oxidase. Histochem

Cell Biol 122, 277-291, doi:10.1007/s00418-004-0679-8 (2004). 154 Allen, L. A. Mechanisms of pathogenesis: evasion of killing by

polymorphonuclear leukocytes. Microbes Infect 5, 1329-1335 (2003). 155 Rada, B. K., Geiszt, M., Kaldi, K., Timar, C. & Ligeti, E. Dual role of

phagocytic NADPH oxidase in bacterial killing. Blood 104, 2947-2953, doi:10.1182/blood-2004-03-1005 (2004).

156 McCaffrey, R. L. et al. Multiple mechanisms of NADPH oxidase inhibition

by type A and type B Francisella tularensis. J Leukoc Biol 88, 791-805, doi:10.1189/jlb.1209811 (2010).

Page 157: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

144

157 KuoLee, R., Harris, G., Conlan, J. W. & Chen, W. Role of neutrophils and NADPH phagocyte oxidase in host defense against respiratory infection with virulent Francisella tularensis in mice. Microbes Infect 13, 447-456, doi:10.1016/j.micinf.2011.01.010 (2011).

158 Schulert, G. S. et al. Francisella tularensis genes required for inhibition of

the neutrophil respiratory burst and intramacrophage growth identified by random transposon mutagenesis of strain LVS. Infect Immun 77, 1324-1336, doi:10.1128/IAI.01318-08 (2009).

159 Santic, M., Molmeret, M. & Abu Kwaik, Y. Modulation of biogenesis of the

Francisella tularensis subsp. novicida-containing phagosome in quiescent human macrophages and its maturation into a phagolysosome upon activation by IFN-gamma. Cell Microbiol 7, 957-967, doi:10.1111/j.1462-5822.2005.00529.x (2005).

160 Clemens, D. L., Lee, B. Y. & Horwitz, M. A. Francisella tularensis

phagosomal escape does not require acidification of the phagosome. Infect Immun 77, 1757-1773, doi:10.1128/IAI.01485-08 (2009).

161 Fortier, A. H. et al. Growth of Francisella tularensis LVS in macrophages:

the acidic intracellular compartment provides essential iron required for growth. Infect Immun 63, 1478-1483 (1995).

162 Lindgren, H. et al. Factors affecting the escape of Francisella tularensis

from the phagolysosome. J Med Microbiol 53, 953-958, doi:10.1099/jmm.0.45685-0 (2004).

163 Barel, M., Meibom, K., Dubail, I., Botella, J. & Charbit, A. Francisella

tularensis regulates the expression of the amino acid transporter SLC1A5 in infected THP-1 human monocytes. Cell Microbiol 14, 1769-1783, doi:10.1111/j.1462-5822.2012.01837.x (2012).

164 Marohn, M. E. et al. Members of the Francisella tularensis phagosomal

transporter subfamily of major facilitator superfamily transporters are critical for pathogenesis. Infect Immun 80, 2390-2401, doi:10.1128/IAI.00144-12 (2012).

165 Gesbert, G. et al. Asparagine assimilation is critical for intracellular

replication and dissemination of Francisella. Cell Microbiol 16, 434-449, doi:10.1111/cmi.12227 (2014).

166 Gesbert, G. et al. Importance of branched-chain amino acid utilization in

Francisella intracellular adaptation. Infect Immun 83, 173-183, doi:10.1128/IAI.02579-14 (2015).

Page 158: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

145

167 Jang, S. & Imlay, J. A. Micromolar intracellular hydrogen peroxide disrupts metabolism by damaging iron-sulfur enzymes. The Journal of biological chemistry 282, 929-937, doi:10.1074/jbc.M607646200 (2007).

168 Chong, A. & Celli, J. The francisella intracellular life cycle: toward

molecular mechanisms of intracellular survival and proliferation. Frontiers in microbiology 1, 138, doi:10.3389/fmicb.2010.00138 (2010).

169 Mariathasan, S., Weiss, D. S., Dixit, V. M. & Monack, D. M. Innate

immunity against Francisella tularensis is dependent on the ASC/caspase-1 axis. J Exp Med 202, 1043-1049, doi:10.1084/jem.20050977 (2005).

170 Henry, T., Brotcke, A., Weiss, D. S., Thompson, L. J. & Monack, D. M.

Type I interferon signaling is required for activation of the inflammasome during Francisella infection. J Exp Med 204, 987-994, doi:10.1084/jem.20062665 (2007).

171 Jones, J. W. et al. Absent in melanoma 2 is required for innate immune

recognition of Francisella tularensis. Proc Natl Acad Sci U S A 107, 9771-9776, doi:10.1073/pnas.1003738107 (2010).

172 Dotson, R. J. et al. Repression of inflammasome by Francisella tularensis

during early stages of infection. J Biol Chem 288, 23844-23857, doi:10.1074/jbc.M113.490086 (2013).

173 Wickstrum, J. R. et al. Francisella tularensis induces extensive caspase-3

activation and apoptotic cell death in the tissues of infected mice. Infect Immun 77, 4827-4836, doi:10.1128/IAI.00246-09 (2009).

174 Parmely, M. J., Fischer, J. L. & Pinson, D. M. Programmed cell death and

the pathogenesis of tissue injury induced by type A Francisella tularensis. FEMS Microbiol Lett 301, 1-11, doi:10.1111/j.1574-6968.2009.01791.x (2009).

175 Kurtz, S. L., Foreman, O., Bosio, C. M., Anver, M. R. & Elkins, K. L.

Interleukin-6 is essential for primary resistance to Francisella tularensis live vaccine strain infection. Infection and immunity 81, 585-597, doi:10.1128/IAI.01249-12 (2013).

176 Laws, T. R., Clark, G. & D'Elia, R. V. Differential role for interleukin-6

during Francisella tularensis infection with virulent and vaccine strains. Infection and immunity 81, 3055-3056, doi:10.1128/IAI.00234-13 (2013).

177 Ray, H. J. et al. Oral live vaccine strain-induced protective immunity

against pulmonary Francisella tularensis challenge is mediated by CD4+ T

Page 159: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

146

cells and antibodies, including immunoglobulin A. Clinical and vaccine immunology : CVI 16, 444-452, doi:10.1128/CVI.00405-08 (2009).

178 Conlan, J. W. Tularemia vaccines: recent developments and remaining

hurdles. Future microbiology 6, 391-405, doi:10.2217/fmb.11.22 (2011). 179 Eneslatt, K. et al. Signatures of T cells as correlates of immunity to

Francisella tularensis. PloS one 7, e32367, doi:10.1371/journal.pone.0032367 (2012).

180 Eneslatt, K. et al. Persistence of cell-mediated immunity three decades

after vaccination with the live vaccine strain of Francisella tularensis. Eur J Immunol 41, 974-980, doi:10.1002/eji.201040923 (2011).

181 Foshay, L. A comparative study of the treatment of tularemia with immune

serum, hyperimmune serum and streptomycin. The American journal of medicine 1, 180-188 (1946).

182 Kirimanjeswara, G. S., Olmos, S., Bakshi, C. S. & Metzger, D. W. Humoral

and cell-mediated immunity to the intracellular pathogen Francisella tularensis. Immunological reviews 225, 244-255, doi:10.1111/j.1600-065X.2008.00689.x (2008).

183 Crane, D. D., Warner, S. L. & Bosio, C. M. A novel role for plasmin-

mediated degradation of opsonizing antibody in the evasion of host immunity by virulent, but not attenuated, Francisella tularensis. Journal of immunology 183, 4593-4600, doi:10.4049/jimmunol.0901655 (2009).

184 Crane, D. D., Scott, D. P. & Bosio, C. M. Generation of a convalescent

model of virulent Francisella tularensis infection for assessment of host requirements for survival of tularemia. PloS one 7, e33349, doi:10.1371/journal.pone.0033349 (2012).

185 Cole, L. E. et al. Antigen-specific B-1a antibodies induced by Francisella

tularensis LPS provide long-term protection against F. tularensis LVS challenge. Proceedings of the National Academy of Sciences of the United States of America 106, 4343-4348, doi:10.1073/pnas.0813411106 (2009).

186 Crane, D. D., Griffin, A. J., Wehrly, T. D. & Bosio, C. M. B1a cells enhance

susceptibility to infection with virulent Francisella tularensis via modulation of NK/NKT cell responses. Journal of immunology 190, 2756-2766, doi:10.4049/jimmunol.1202697 (2013).

187 Metzger, D. W., Salmon, S. L. & Kirimanjeswara, G. Differing effects of

interleukin-10 on cutaneous and pulmonary Francisella tularensis live vaccine strain infection. Infection and immunity 81, 2022-2027, doi:10.1128/IAI.00024-13 (2013).

Page 160: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

147

188 Hunt, D. et al. Francisella tularensis elicits IL-10 via a PGE(2)-inducible

factor, to drive macrophage MARCH1 expression and class II down-regulation. PloS one 7, e37330, doi:10.1371/journal.pone.0037330 (2012).

189 Wilson, J. E., Katkere, B. & Drake, J. R. Francisella tularensis induces

ubiquitin-dependent major histocompatibility complex class II degradation in activated macrophages. Infection and immunity 77, 4953-4965, doi:10.1128/IAI.00844-09 (2009).

190 Woolard, M. D. et al. Francisella tularensis-infected macrophages release

prostaglandin E2 that blocks T cell proliferation and promotes a Th2-like response. J Immunol 178, 2065-2074 (2007).

191 Zaslona, Z., Serezani, C. H., Okunishi, K., Aronoff, D. M. & Peters-Golden,

M. Prostaglandin E2 restrains macrophage maturation via E prostanoid receptor 2/protein kinase A signaling. Blood 119, 2358-2367, doi:10.1182/blood-2011-08-374207 (2012).

192 Asakrah, S. et al. Post-exposure therapeutic efficacy of COX-2 inhibition

against Burkholderia pseudomallei. PLoS neglected tropical diseases 7, e2212, doi:10.1371/journal.pntd.0002212 (2013).

193 Cowley, S. C. & Elkins, K. L. Immunity to francisella. Frontiers in

microbiology 2, 26, doi:10.3389/fmicb.2011.00026 (2011). 194 Skyberg, J. A. Immunotherapy for tularemia. Virulence 4, 859-870,

doi:10.4161/viru.25454 (2013). 195 Parrow, N. L., Fleming, R. E. & Minnick, M. F. Sequestration and

scavenging of iron in infection. Infection and immunity 81, 3503-3514, doi:10.1128/IAI.00602-13 (2013).

196 Griffin, A. J., Crane, D. D., Wehrly, T. D. & Bosio, C. M. Successful

protection against tularemia in C57BL/6 mice is correlated with expansion of Francisella tularensis-specific effector T cells. Clin Vaccine Immunol 22, 119-128, doi:10.1128/CVI.00648-14 (2015).

197 Su, J. et al. Genome-wide identification of Francisella tularensis virulence

determinants. Infection and immunity 75, 3089-3101, doi:10.1128/IAI.01865-06 (2007).

198 Singh, R. & Eltis, L. D. The multihued palette of dye-decolorizing

peroxidases. Archives of biochemistry and biophysics 574, 56-65, doi:10.1016/j.abb.2015.01.014 (2015).

Page 161: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

148

199 Zubay, G., Morse, D. E., Schrenk, W. J. & Miller, J. H. Detection and isolation of the repressor protein for the tryptophan operon of Escherichia coli. Proceedings of the National Academy of Sciences of the United States of America 69, 1100-1103 (1972).

200 Ashkenazy, H., Erez, E., Martz, E., Pupko, T. & Ben-Tal, N. ConSurf 2010:

calculating evolutionary conservation in sequence and structure of proteins and nucleic acids. Nucleic acids research 38, W529-533, doi:10.1093/nar/gkq399 (2010).

201 Landau, M. et al. ConSurf 2005: the projection of evolutionary

conservation scores of residues on protein structures. Nucleic acids research 33, W299-302, doi:10.1093/nar/gki370 (2005).

202 Troxell, B. & Hassan, H. M. Transcriptional regulation by Ferric Uptake

Regulator (Fur) in pathogenic bacteria. Frontiers in cellular and infection microbiology 3, 59, doi:10.3389/fcimb.2013.00059 (2013).

203 Kammler, M., Schon, C. & Hantke, K. Characterization of the ferrous iron

uptake system of Escherichia coli. Journal of bacteriology 175, 6212-6219 (1993).

204 Soares, M. P. & Weiss, G. The Iron age of host-microbe interactions.

EMBO Rep 16, 1482-1500, doi:10.15252/embr.201540558 (2015). 205 Radtke, A. L. & O'Riordan, M. X. Intracellular innate resistance to bacterial

pathogens. Cell Microbiol 8, 1720-1729, doi:10.1111/j.1462-5822.2006.00795.x (2006).

206 Paradkar, P. N. et al. Iron depletion limits intracellular bacterial growth in

macrophages. Blood 112, 866-874, doi:10.1182/blood-2007-12-126854 (2008).

207 Chlosta, S. et al. The iron efflux protein ferroportin regulates the

intracellular growth of Salmonella enterica. Infect Immun 74, 3065-3067, doi:10.1128/IAI.74.5.3065-3067.2006 (2006).

208 Pan, X., Tamilselvam, B., Hansen, E. J. & Daefler, S. Modulation of iron

homeostasis in macrophages by bacterial intracellular pathogens. BMC Microbiol 10, 64, doi:10.1186/1471-2180-10-64 (2010).

209 Hartley, G. et al. Grey variants of the live vaccine strain of Francisella

tularensis lack lipopolysaccharide O-antigen, show reduced ability to survive in macrophages and do not induce protective immunity in mice. Vaccine 24, 989-996, doi:10.1016/j.vaccine.2005.08.075 (2006).

Page 162: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

149

210 Soni, S. et al. Francisella tularensis blue-gray phase variation involves structural modifications of lipopolysaccharide o-antigen, core and lipid a and affects intramacrophage survival and vaccine efficacy. Frontiers in microbiology 1, 129, doi:10.3389/fmicb.2010.00129 (2010).

211 Lindgren, H. et al. Resistance of Francisella tularensis strains against

reactive nitrogen and oxygen species with special reference to the role of KatG. Infection and immunity 75, 1303-1309, doi:10.1128/IAI.01717-06 (2007).

212 Ma, Z. et al. EmrA1 membrane fusion protein of Francisella tularensis LVS

is required for resistance to oxidative stress, intramacrophage survival and virulence in mice. Molecular microbiology 91, 976-995, doi:10.1111/mmi.12509 (2014).

213 Crane, D. D., Bauler, T. J., Wehrly, T. D. & Bosio, C. M. Mitochondrial

ROS potentiates indirect activation of the AIM2 inflammasome. Front Microbiol 5, 438, doi:10.3389/fmicb.2014.00438 (2014).

214 Bou-Abdallah, F., Lewin, A. C., Le Brun, N. E., Moore, G. R. & Chasteen,

N. D. Iron detoxification properties of Escherichia coli bacterioferritin. Attenuation of oxyradical chemistry. The Journal of biological chemistry 277, 37064-37069, doi:10.1074/jbc.M205712200 (2002).

215 Honarmand Ebrahimi, K., Hagedoorn, P. L. & Hagen, W. R. Unity in the

biochemistry of the iron-storage proteins ferritin and bacterioferritin. Chem Rev 115, 295-326, doi:10.1021/cr5004908 (2015).

216 Andrews, S. C. The Ferritin-like superfamily: Evolution of the biological

iron storeman from a rubrerythrin-like ancestor. Biochim Biophys Acta 1800, 691-705, doi:10.1016/j.bbagen.2010.05.010 (2010).

217 Weeratunga, S. K. et al. Binding of Pseudomonas aeruginosa

apobacterioferritin-associated ferredoxin to bacterioferritin B promotes heme mediation of electron delivery and mobilization of core mineral iron. Biochemistry 48, 7420-7431, doi:10.1021/bi900561a (2009).

218 Chen, C. Y. & Morse, S. A. Neisseria gonorrhoeae bacterioferritin:

structural heterogeneity, involvement in iron storage and protection against oxidative stress. Microbiology 145 ( Pt 10), 2967-2975, doi:10.1099/00221287-145-10-2967 (1999).

219 Mey, A. R., Wyckoff, E. E., Kanukurthy, V., Fisher, C. R. & Payne, S. M.

Iron and fur regulation in Vibrio cholerae and the role of fur in virulence. Infection and immunity 73, 8167-8178, doi:10.1128/IAI.73.12.8167-8178.2005 (2005).

Page 163: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

150

220 Velayudhan, J., Castor, M., Richardson, A., Main-Hester, K. L. & Fang, F.

C. The role of ferritins in the physiology of Salmonella enterica sv. Typhimurium: a unique role for ferritin B in iron-sulphur cluster repair and virulence. Molecular microbiology 63, 1495-1507, doi:10.1111/j.1365-2958.2007.05600.x (2007).

221 Smith, J. L. The physiological role of ferritin-like compounds in bacteria.

Crit Rev Microbiol 30, 173-185, doi:10.1080/10408410490435151 (2004). 222 Ma, J. F. et al. Bacterioferritin A modulates catalase A (KatA) activity and

resistance to hydrogen peroxide in Pseudomonas aeruginosa. J Bacteriol 181, 3730-3742 (1999).

223 Reddy, P. V., Puri, R. V., Khera, A. & Tyagi, A. K. Iron storage proteins

are essential for the survival and pathogenesis of Mycobacterium tuberculosis in THP-1 macrophages and the guinea pig model of infection. Journal of bacteriology 194, 567-575, doi:10.1128/JB.05553-11 (2012).

224 Pandey, R. & Rodriguez, G. M. A ferritin mutant of Mycobacterium

tuberculosis is highly susceptible to killing by antibiotics and is unable to establish a chronic infection in mice. Infection and immunity 80, 3650-3659, doi:10.1128/IAI.00229-12 (2012).

225 Bakshi, C. S. et al. An improved vaccine for prevention of respiratory

tularemia caused by Francisella tularensis SchuS4 strain. Vaccine 26, 5276-5288, doi:10.1016/j.vaccine.2008.07.051 (2008).

226 Lee, B. Y., Horwitz, M. A. & Clemens, D. L. Identification, recombinant

expression, immunolocalization in macrophages, and T-cell responsiveness of the major extracellular proteins of Francisella tularensis. Infection and immunity 74, 4002-4013, doi:10.1128/IAI.00257-06 (2006).

227 Zaide, G. et al. Identification and characterization of novel and potent

transcription promoters of Francisella tularensis. Appl Environ Microbiol 77, 1608-1618, doi:10.1128/AEM.01862-10 (2011).

228 Mishra, S. & Imlay, J. Why do bacteria use so many enzymes to scavenge

hydrogen peroxide? Archives of biochemistry and biophysics 525, 145-160, doi:10.1016/j.abb.2012.04.014 (2012).

229 Postic, G. et al. Identification of small RNAs in Francisella tularensis. BMC

genomics 11, 625, doi:10.1186/1471-2164-11-625 (2010).

Page 164: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

151

230 Bradley, J. M., Le Brun, N. E. & Moore, G. R. Ferritins: furnishing proteins with iron. J Biol Inorg Chem 21, 13-28, doi:10.1007/s00775-016-1336-0 (2016).

231 Savitt, A. G., Mena-Taboada, P., Monsalve, G. & Benach, J. L. Francisella

tularensis infection-derived monoclonal antibodies provide detection, protection, and therapy. Clinical and vaccine immunology : CVI 16, 414-422, doi:10.1128/CVI.00362-08 (2009).

232 Rotem, S. et al. Protective immunity against lethal F. tularensis holarctica

LVS provided by vaccination with selected novel CD8+ T cell epitopes. PloS one 9, e85215, doi:10.1371/journal.pone.0085215 (2014).

233 Bauler, T. J., Chase, J. C., Wehrly, T. D. & Bosio, C. M. Virulent

Francisella tularensis destabilize host mRNA to rapidly suppress inflammation. J Innate Immun 6, 793-805, doi:10.1159/000363243 (2014).

234 Navratil, A. R., Brummett, A. M., Bryan, J. D. & Woolard, M. D. Francisella

tularensis LVS induction of prostaglandin biosynthesis by infected macrophages requires specific host phospholipases and lipid phosphatases. Infect Immun 82, 3299-3311, doi:10.1128/IAI.02060-14 (2014).

235 Rockx-Brouwer, D. et al. Low dose vaccination with attenuated Francisella

tularensis strain SchuS4 mutants protects against tularemia independent of the route of vaccination. PloS one 7, e37752, doi:10.1371/journal.pone.0037752 (2012).

236 Richer, M. J., Nolz, J. C. & Harty, J. T. Pathogen-specific inflammatory

milieux tune the antigen sensitivity of CD8(+) T cells by enhancing T cell receptor signaling. Immunity 38, 140-152, doi:10.1016/j.immuni.2012.09.017 (2013).

237 Hubalek, M. et al. Comparative proteome analysis of cellular proteins

extracted from highly virulent Francisella tularensis ssp. tularensis and less virulent F. tularensis ssp. holarctica and F. tularensis ssp. mediaasiatica. Proteomics 4, 3048-3060, doi:10.1002/pmic.200400939 (2004).

238 Pavkova, I., Hubalek, M., Zechovska, J., Lenco, J. & Stulik, J. Francisella

tularensis live vaccine strain: proteomic analysis of membrane proteins enriched fraction. Proteomics 5, 2460-2467, doi:10.1002/pmic.200401213 (2005).

239 Peng, K., Broz, P., Jones, J., Joubert, L. M. & Monack, D. Elevated AIM2-

mediated pyroptosis triggered by hypercytotoxic Francisella mutant strains

Page 165: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

152

is attributed to increased intracellular bacteriolysis. Cell Microbiol 13, 1586-1600, doi:10.1111/j.1462-5822.2011.01643.x (2011).

240 Hazlett, K. R. et al. Adaptation of Francisella tularensis to the mammalian

environment is governed by cues which can be mimicked in vitro. Infect Immun 76, 4479-4488, doi:10.1128/IAI.00610-08 (2008).

241 Singh, A. et al. Discordant results obtained with Francisella tularensis

during in vitro and in vivo immunological studies are attributable to compromised bacterial structural integrity. PLoS One 8, e58513, doi:10.1371/journal.pone.0058513 (2013).

242 Bocharov, G., Argilaguet, J. & Meyerhans, A. Understanding Experimental

LCMV Infection of Mice: The Role of Mathematical Models. J Immunol Res 2015, 739706, doi:10.1155/2015/739706 (2015).

243 Wehrly, T. D. et al. Intracellular biology and virulence determinants of

Francisella tularensis revealed by transcriptional profiling inside macrophages. Cell Microbiol 11, 1128-1150, doi:10.1111/j.1462-5822.2009.01316.x (2009).

244 Valentino, M. D. et al. A broadly applicable approach to T cell epitope

identification: application to improving tumor associated epitopes and identifying epitopes in complex pathogens. J Immunol Methods 373, 111-126, doi:10.1016/j.jim.2011.08.007 (2011).

245 Badovinac, V. P., Porter, B. B. & Harty, J. T. Programmed contraction of

CD8(+) T cells after infection. Nat Immunol 3, 619-626, doi:10.1038/ni804 (2002).

246 Pearce, E. L. & Shen, H. Making sense of inflammation, epigenetics, and

memory CD8+ T-cell differentiation in the context of infection. Immunol Rev 211, 197-202, doi:10.1111/j.0105-2896.2006.00399.x (2006).

247 Liu, X. et al. OxyR Is a Key Regulator in Response to Oxidative Stress in

Streptomyces avermitilis. Microbiology, doi:10.1099/mic.0.000251 (2016). 248 Ishiga, Y. & Ichinose, Y. Pseudomonas syringae pv. tomato OxyR Is

Required for Virulence in Tomato and Arabidopsis. Mol Plant Microbe Interact 29, 119-131, doi:10.1094/MPMI-09-15-0204-R (2016).

249 Chung, C. H., Fen, S. Y., Yu, S. C. & Wong, H. C. Influence of oxyR on

Growth, Biofilm Formation, and Mobility of Vibrio parahaemolyticus. Appl Environ Microbiol 82, 788-796, doi:10.1128/AEM.02818-15 (2015).

Page 166: Oxidative stress resistance in the Francisella tularensis live vaccine strain is associated with

153

250 Christman, M. F., Storz, G. & Ames, B. N. OxyR, a positive regulator of hydrogen peroxide-inducible genes in Escherichia coli and Salmonella typhimurium, is homologous to a family of bacterial regulatory proteins. Proc Natl Acad Sci U S A 86, 3484-3488 (1989).

251 Schell, M. A. Molecular biology of the LysR family of transcriptional

regulators. Annu Rev Microbiol 47, 597-626, doi:10.1146/annurev.mi.47.100193.003121 (1993).

252 Imlay, J. A. Transcription Factors That Defend Bacteria Against Reactive

Oxygen Species. Annu Rev Microbiol 69, 93-108, doi:10.1146/annurev-micro-091014-104322 (2015).