You are on page 1of 19

Chapter 108

Francisella
Anna-Lena Johansson1, Laila Noppa1, Emelie Näslund Salomonsson1 and Åke Forsberg2
1
FOI Swedish Defense Research Agency, Umeå, Sweden, 2Umeå University, Umeå, Sweden

INTRODUCTION Due to its high infectivity, ease of aerosol dissemina-


tion, and ability to cause severe illness and death,
Francisella tularensis is a non-motile Gram-negative coc- F. tularensis has been categorized as a potential bioter-
cobacillus and a facultative intracellular pathogen that rorism agent. It is currently classified as a Tier 1 select
causes the zoonotic infection known as tularaemia. agent (formerly called category A select agent) which
F. tularensis is associated with a wider range of hosts specifies ‘a select agent with a documented risk of caus-
than most other zoonotic pathogens. The organism has ing a high consequence event’ by the United States
been found in more than 300 species, from large mammals Centers for Disease Control and Prevention (CDC) [9].
and vertebrates to invertebrates, arthropods and amoebas Indeed, subsp. tularensis was developed for use as a bio-
[1]. In the environment F. tularensis has been isolated logical weapon by several countries including Japan, the
from water, soil, and various wild animals, especially rab- USA and the Soviet Union during World War II and the
bits, hares and rodents. Outbreaks of disease in humans Cold War [10,11]. The USA alone reported a 450 kg
often parallel outbreaks of tularaemia in wild animals that stockpile of dry-powder F. tularensis when it scrapped
also often correlates to peaks of rodent populations [2,3]. its bioweapons programme in 1973. The anthrax letters
The most important mammalian species involved in in the aftermath of the 9/11 events in 2001 increased the
human infection differ between the continents. In North concern about biothreat agents in general and resulted in
America voles, mice, squirrels, muskrats and beavers are new initiatives to promote preparedness and research in
the most important, while in Europe and Asia lagomorphs the area of biodefence [12]. This increased funding has
(hares and rabbits) and voles are known hosts. Although had a major impact on tularaemia research over the past
most frequently found to be infected, lagomorphs and decade [12].
rodents are unlikely to be reservoirs for F. tularensis,
since infection often leads to acute disease in these ani-
mals. Infectious transfer to humans and other mammals TAXONOMY, EPIDEMIOLOGY AND
also occurs through aerosolization or direct contact with
infected material or carcasses. Another important route of
ECOLOGY
transfer between mammalian hosts involves blood-feeding The genus Francisella comprises at least four recognized
arthropods (insects) such as ticks, mosquitoes and biting species; F. tularensis, F. hispaniensis, F. philomiragia
flies [4]. The most important vector species also differ and F. noatunensis (Table 108.1). In contrast to F. tular-
between the continents. In North America, ticks and bit- ensis, F. philomiragia is rarely associated with human
ing flies are the most common vectors, in central Europe, disease and has only been isolated from patients who
ticks are the main vector while in Northern Europe and in were nearly drowned [13 15]. F. noatunensis is closely
Russia, the bacterium can also be transmitted by mosqui- related to F. philomiragia and seems to be the only mem-
toes. The bacterium also persists for long periods in ber within the genus able to cause disease in fish
watercourses, possibly in association with protozoa [5,6]. [16 20]. Challenge studies indicate that this pathogen
F. tularensis subspecies tularensis (also denoted transmits the disease (francisellosis) directly between fish
type A) is one of the most infectious bacteria known, [21 23]. Francisellosis in fish has during recent years
since inhalation of as few as 10 15 organisms is suffi- been diagnosed all over the world [18]. In the Atlantic
cient to initiate infection in humans [7,8]. Ocean, both wild and farmed cod have been diagnosed

Molecular Medical Microbiology. DOI: http://dx.doi.org/10.1016/B978-0-12-397169-2.00108-6


© 2015 Elsevier Ltd. All rights reserved. 1991
1992 PART | 17 Animal and Ectoparasitic Source Infections

TABLE 108.1 Recognized Francisella Species, Virulence and Distribution

Species Subspecies Type Virulence Distribution/Region


F. tularensis tularensis A.I A Very high North America Midwest, California,
and Massachusetts

tularensis A.II A Moderate North America California,


Less virulent to humans than mountain states
Type B isolates

holarctica B High Throughout Northern hemisphere

mediasiatica High Central Asia

novicida Low, (immuno-suppressed Broad


individuals)

F. hispaniensis ?

F. philomiragia Low (immuno-suppressed Central Asia


individuals)

F. noatunensis noatunensis High, solely a fish pathogen Broad/Atlantic Ocean

orientalis High, solely a fish pathogen Broad

and the mortality rate appears to be high [20,21]. The risk F. tularensis subsp. tularensis has been further divided
for humans to get infected from the fish and shellfish into two genetically distinct subpopulations (clades), the
pathogenic Francisella species is considered very low highly diverse A.I group and the less diverse A.II group
[18]. In addition, F. hispaniensis was recently accepted as [34]. These subgroups also differ in clinical severity since
a separate species that includes several strains previously A.I isolates are more virulent to humans than A.II isolates
denoted as novicida-like isolates [24 26]. which appear to be even less virulent to humans than
F. tularensis is subdivided into four subspecies; tular- subsp. holarctica (type B) isolates [35,36]. The A.I and
ensis (also denoted type A), holarctica (also denoted type A.II groups show distinct geographic distribution which
B), mediasiatica and novicida which are highly similar at matches well with geographic ranges of known tularaemia
genomic level [27] but differ significantly in virulence hosts and vectors, and abiotic components, suggesting
and geographical distribution. that the two groups occupy separate ecological niches
Subspecies tularensis (endemic in North America) and [34]. F. tularensis subsp. holarctica can also be further
holarctica (predominantly found throughout the Northern divided into clades but these are genetically more homo-
Hemisphere but recently also isolated in Australia [28]) genic. This is interesting since subsp. holarctica is distrib-
are the most prevalent and are responsible for the majority uted in a larger geographic area, throughout North
of human cases [29,30]. F. tularensis is the most virulent America and the whole of Eurasia with some reports also
among the subspecies with a mortality rate of up to 30% from Australia [28]. In Northern Europe and in Russia,
if untreated, while F. holarctica causes a milder form of subsp. holarctica is associated with blood-feeding mos-
the disease that is rarely fatal. F. mediasiatica is found quitoes, in association with watercourses, while in central
within a geographically limited niche, i.e., in central Asia, Europe it is more closely associated with ticks.
and exhibits even lower virulence and only rarely causes Tularaemia ecology is still only partially understood but
human infections [31]. Subsp. novicida has only been iso- the current view describes two ecological niches, one
lated from immune-compromised patients. With the rap- mainly terrestrial, i.e., tularensis, and one more connected
idly increasing number of genome sequences available to aquatic environments, i.e., holarctica.
the knowledge on the diversity within the genus The increased availability of completely sequenced
Francisella has recently expanded substantially. This has Francisella genomes has revealed insights into evolution-
opened up a discussion on the boundaries between taxo- ary processes within the genus and several studies have
nomic units within the genus. One example concerns identified new variants of Francisella from soil and water
F. novicida that previously was considered as a separate [26,37 39], as well as Francisella-like endosymbionts
species but now has been ranked as a subspecies [32,33]. with high similarity to F. tularensis (Fig. 108.1) [40,41].
Chapter | 108 Francisella 1993

F. tularensis subsp. holarctica

F. tularensis subsp. mediasiatica

F. tularensis subsp. tularensis


Clade 1
F. tularensis subsp. novicida

F. hispaniensis

Wolbachia persica

F. noatunensis subsp. orientalis

F. philomiragia Clade 2

F. noatunensis subsp. noatunensis

Fangia hongkongensis

Piscirickettsia salmonis

5000

FIGURE 108.1 An overview of the whole-genome phylogeny of the genus Francisella and two genetic relatives, Fangia hongkongensis and
Piscirickettsia salmonis, based on single nucleotide polymorphism in the core genome. Figure is slightly modified from [40]. The Francisella genus is
divided into two parts, clade 1 (F. tularensis and F. novicida) and clade 2 (F. noatunensis and F. philomiragia) occupying both terrestrial and aquatic/
marine habitats, respectively. The phylogeny data show that pathogenic species in each clade (mammals and fish) have emerged independently by
similar evolutionary paths of host adaptation and that the ancestral Francisella species originated in a marine habitat [40].

Thus, members of the Francisella genus can be found in (usually between 3 5 days) [43]. The clinical symptoms
and appear to be associated with a wide range of different and severity depend on (i) the type of F. tularensis that
ecological niches, from specialized endosymbionts and causes the infection, (ii) the infection dose, (iii) the route
pathogens with different host ranges, to those that appear of infection and (iv) the immune status of the recipient.
to be free-living. The increased knowledge about impor- So far there are no confirmed reports of transfer of the
tant sequence differences between environmental species disease from human to human, even if a theoretical risk
and F. tularensis also allows for the design of highly spe- exists due to the occurrence of F. tularensis in respiratory
cific F. tularensis detection assays with improved ability fluids [44].
to discriminate between strains. This is particularly Clinically relevant species of Francisella that cause
important since current methods frequently falsely detect human infections are F. tularensis subspecies tularensis
F. tularensis in environmental samples [40,42]. The (type A), holarctica (type B), and mediasiatica, and in
genome information can also be used for diagnostic addition immune-compromised individuals could be sus-
purposes and epidemiological studies as well as tracing ceptible to F. novicida and F. philomiragia infections
specific strains when deliberate release is suspected in [13,15,45]. If untreated, type A infections have mortality
bio-forensic analysis and studies. rates of 30 60% [45], while type B infections are much
less severe and mortality following type B infections is
rare [10].
TULARAEMIA
Tularaemia, also called rabbit fever, hare fever, lemming
fever, or deerfly fever, is a zoonotic infection disease
Symptoms
where humans could be infected through contact with Diagnosis of tularaemia is relatively uncomplicated in
infected animals or via flies, ticks or mosquitoes. The dis- areas where the disease is known to occur and cases
ease onsets after an incubation period of 1 21 days can be expected. It is far more demanding when it
1994 PART | 17 Animal and Ectoparasitic Source Infections

TABLE 108.2 Different Types of Tularaemia

Type of Tularaemia Typical Symptoms


Ulceroglandular Skin ulcer(s) and swollen tender glands

Glandular Swollen glands without a skin ulcer

Oculoglandular Painful swollen and red eyes. Swollen cervical lymphnodes

Oropharyngeal and gastrointestinal Sore throat, stomach pain, vomiting, diarrhoea and occasionally bowel ulceration.
Swollen cervical lymphnodes

Pneumonic Dry cough and sharp chest pain

Typhoidal (septicaemic) Fevers, chills, muscle pain, and lack of energy. Absence of an ulcer or swollen glands

occurs in areas where tularaemia has rarely been


reported since the symptoms are shared with several
other diseases with flu-like symptoms including sudden
fever, headache and swollen lymph nodes and fatigue.
Respiratory symptoms, like cough and a sore throat,
can occur even if the infection was not acquired by
inhalation. The type of tularaemia depends on the route
of infection, where the most common are ulceroglandu-
lar, glandular and pneumonic, but oropharyngeal, ocu-
loglandular, gastrointestinal and typhoidal forms also
occur (Table 108.2) [43].
Ulceroglandular tularaemia that appears to be the most
common type is usually vector-transmitted, in Central
Europe and the USA predominantly by ticks [4,10,46]
and in Northern Europe mainly by mosquitos [10,47]. A
person with a skin wound could also acquire ulcerogland-
ular tularaemia as a result of handling infected animals
[10,48]. The clinical symptoms may vary but commonly
there is a local cutaneous papule or a granuloma at the
inoculation site, eventually followed by an abscess and an
eschar after a few days. Several days after the inoculation,
typical symptoms include fever and flu-like signs,
together with one or more lymph nodes in the proximity
of the papule becoming swollen. In more severe cases the
lymph node could eventually burst [11]. Symptoms
from glandular tularaemia are the same as during ulcero- FIGURE 108.2 Swollen lymph node in a patient suffering from oculo-
glandular tularaemia but without any skin infection. glandular tularaemia. With permission from: Dr Henrik Eliasson, Örebro
Oculoglandular tularaemia could be transmitted via direct University Hospital, Sweden.
infection/contamination of the eye. The symptoms are
often conjunctivitis followed by a cervical lymphadeno-
pathy (Fig. 108.2). The symptoms of gastrointestinal tularaemia are typically
After ingestion of infected food or water oropharyn- vomiting and diarrhoea, but in the more severe cases also
geal and gastrointestinal tularaemia could occur depend- an ulceration of the bowel [45]. For pneumonic tularaemia
ing on the focus of infection. Oropharyngeal tularaemia is the symptoms are typically flu-like with headache and
the most common presentation in Turkey and in Eastern high fever, and a mild, dry cough that occur at later stages.
European countries [49 53]. The clinical findings may There is a discrepancy between primary pneumonia and
vary but typical symptoms are a painful sore throat, swollen secondary pneumonia, where the former is a result of inha-
neck, cervical lymphadenopathy and tonsillopharyngitis. lation and secondary pneumonia could be found after a
Chapter | 108 Francisella 1995

haematogenic spread from another type of tularaemia. more severe cases aminoglycosides like gentamicin are a
[43,54]. Typhoidal tularaemia is a generic term used to good alternative or complement [65,70]. The levels of
describe systemic illness in the absence of other symptoms protection afforded by antibiotics are highly dependent
indicating another specific type of tularaemia [10]. upon timely administration following infection [71 73].
Furthermore, in rare cases F. novicida, F. philomiragia
and F. noatunensis could cause bacteraemia after near-
drowning accidents in salt water, where the typical
PATHOGENIC LIFESTYLE OF FRANCISELLA
symptoms are pneumonia [13,15,18,55]. The very low infectious dose suggest that Francisella has
evolved to become a highly successful pathogen and cen-
tral to the pathogenic lifestyle is the ability to replicate
Diagnosis
intracellularly and also circumvent or evade the different
Several methods for diagnosis of tularaemia have been stages of immune response by the host. This includes
developed. Different methods are used depending on the strategies to infect via very different routes and means of
time point after onset of the disease, the equipment avail- transmission by different vectors like ticks, mosquitoes
able and the need for exact identification of the and flies or via inhalation, ingestion or small wounds.
Francisella subspecies that has caused the disease. If Irrespective of the route of entry the pathogen can rap-
pneumonic tularaemia is suspected, the pneumonia might idly disseminate from the initial site of infection, prolifer-
be verified using X-ray. Culturing could be performed ate and spread to different organs in the infected host.
from ulcers, sputum, pharyngeal washing, and occasion- Various strategies have evolved to mitigate the initial
ally in blood [10], however culturing could be difficult extracellular innate immune response. Francisella can
and many clinical laboratories do not have the ability to enter various host cells like macrophages, dendritic cells,
handle this pathogen since there is a demand of a bio- polymorphonuclear neutrophils (PMNs), endothelial and
safety 3 level laboratory when handling both type A and alveolar epithelial cells [11,74,75]. Once inside cells the
type B strains [56]. survival and replication absolutely rely on the ability to
Useful tools for diagnosis are agglutination tests, escape from the initial phagosome. This is also a central
ELISA and Western blotting. The main disadvantage of virulence strategy that involves a number of genes
using serological-based assays is the late onset of encoded on the Francisella pathogenicity island (FPI).
detectable antibody levels that usually appear about 2 The subsequent growth in the host cell cytosol also
weeks after infection [57,58]. In order to measure the requires successful tampering with the intracellular innate
prevalence of antibodies, whole killed cells, outer mem- defence mechanisms as well as a metabolic adaption to
brane or LPS could be used as antigens [57,59]. The high- growth inside the host cell.
est level of antibodies could be detected in blood 4 7 In line with the pathogenic lifestyle is that cell-
weeks after the onset of the disease, and detectable levels mediated immunity is very important for development of
have also been found to persist even 31 years after the ini- protective immunity against subsequent infection. Still, sub-
tial infection [60,61]. Therefore identification of antibo- stantial antibody responses are generated in humans for
dies against F. tularensis may represent a previous instance against LPS during human F. tularensis infections
infection of the disease as well as an acute illness. [76 78]. This is not surprising since F. tularensis has also
Immunological analysis could be performed on serum, been shown to exist in an extracellular phase in mice [79].
urine, sputum or on tissue biopsies [10,62] However, due However, due to the mainly intracellular nature of
to the delay of antibody response after onset of the dis- F. tularensis infections, the cell-mediated immune response
ease, the optional method for rapid and early diagnosis of is thought to be the most important for protective immunity
tularaemia is a PCR-based approach [63,64]. against tularaemia [80].
The available genome sequences of several strains of
the different F. tularensis subspecies have shown that the
Therapy overall similarity at genomic level is very high and this
Historically, treatment of tularaemia has been based on also includes the more or less non-pathogenic subsp. novi-
streptomycin or gentamicin treatment, but streptomycin is cida. Even though there are distinct differences even
not commonly used today due to its potential toxicity and between subgroups (clades) within each subsp. it is still
is therefore less available in several countries [10,65 68]. far from clear how the genetic differences translate into
All F. tularensis species are naturally resistant to beta- the differences seen in virulence to humans [81,82]. The
lactam antibiotics [69], but so far development of resis- vast majority of genes involved in immune evasion and/or
tance against any other antibiotics has not been reported. virulence are common for the subspecies.
Today ciprofloxacin and doxycycline represent the first While the ability to infect and replicate inside host
therapeutic choices for treatment of tularaemia, but in cells is the single most important property for successful
1996 PART | 17 Animal and Ectoparasitic Source Infections

infection and virulence in Francisella, the extracellular by bacteria in response to nutritional starvation [89,91].
phase of the infection is clearly also of relevance. It is not MigR regulates gene expression of the FPI and also posi-
entirely clear during what stages of infection Francisella tively regulates transcription of fevR. It is still unclear if
is extracellular, but it could be assumed to occur both the regulation is direct or indirect, due to the absence of a
early after the entry into the host as well as at later stages DNA binding domain in MigR. In addition Mig R, as
when the infection could be more or less systemic. well as FevR, was found to be involved in blockade of
neutrophil NADPH oxidase activity, a feature that is an
important strategy of F. tularensis to evade neutrophil
REGULATION OF GENE EXPRESSION killing [92]. PmrA is another response regulator of a large
group of genes and has also been found to be essential for
DURING HOST INFECTION intracellular survival and virulence in mice [93,94].
Bacteria have evolved the capability to sense environmen- Today, it is unclear how expression of several other
tal changes and adapt to these changes by altering expres- virulence genes outside the FPI is regulated, but it seems
sion of different enzymes, proteins and toxins. In many clear that environmental conditions, like increased tem-
bacterial species, the regulatory systems are well known perature and intracellular growth, are factors that initiate
and a broad range of regulatory genes have been induction of gene expression [95,96].
identified. Many of these regulators are so-called two- In most organisms, small non-coding RNAs (sRNAs)
component systems (TCS), which are typically composed have a regulatory function based on pairing with comple-
of a membrane-localized sensor, that senses the environ- mentary mRNA [97]. This base pairing is often facilitated
ment, and a transcriptional response regulator located in by Hfq, an RNA chaperone. The hfq homologue identified
the cytoplasm [83]. In human virulent F. tularensis spe- in F. tularensis was, as in many other bacteria, found to
cies there is no typical complete TCS, and also only one be important for growth and resistance to different stress
alternative sigma factor, RpoH, is identified [84,85]. conditions [98]. To date a few sRNA are identified in
In general, transcriptional regulators identified in F. tularensis, with variable impact on growth, intracellu-
F. tularensis are relatively few compared to other bacte- lar survival and virulence [99,100].
rial species, but on the other hand it seems that these Taken together the regulatory network in F. tularensis
regulators control transcription of a broad range of genes. is unique compared to many other bacteria and focuses
The best-characterized transcriptional regulators in on assuring timely expression of key genes during the
F. tularensis are; MglA, SspA, FevR, PmrA and MigR. infection cycle where the key events are rapid escape
All these regulators are conserved between F. tularensis from the phagosome and evasion of host innate defence
subspecies, and their regulatory function has been experi- mechanisms.
mentally established in more than one subspecies.
Common to these regulators is that they are involved in
regulation of FPI gene expression, but some of them have EXTRACELLULAR PHASE OF INFECTION
been found to regulate several additional genes.
MlgA was one of the first identified regulatory genes
Spread from Initial Infection Site
in F. tularensis [86]. MglA is a homologue to SspA of With a very low infection dose, even via vector-borne
Escherichia coli where it is a transcriptional factor that transmission, one of the initial challenges for Francisella
responds to nutrient limitation and regulation during star- is to survive the extracellular immune response and to
vation. In F. tularensis MglA is up-regulated during spread from the initial infection site. Francisella encodes
infection in macrophages and an mlgA mutant (novicida) gene clusters for functional type IV pili (Tfp) and there is
is unable to grow in macrophages and amoeba, and also some evidence that these adhesive structures may be
attenuated in mice [86 88]. An additional SspA homo- important for the spread from the initial site of infection.
logue is identified and these two homologues, SspA and Pilin mutants in type B strains were found to require high-
MglA, interact and associate with RNA polymerase to er inoculum to be able to spread and cause systemic infec-
regulate transcription [89]. It is suggested that the MglA/ tion [101,102], while the pilin was dispensable for
SspA complex responds to environmental stress and is intracellular replication. The attenuated vaccine strain
therefore essential for bacterial survival and growth in LVS (type B) also lacks one of the pilin genes [103].
tough environments [90]. FevR (pigR) is another regula- However, it is less clear if Tfp has a major role in the vir-
tory protein that acts in parallel with MglA/SspA and has ulence of the most virulent subsp. tularensis (type A) as
been found to promote expression of genes essential for different mutants in the pilin genes have been found to
survival. This transcriptional up-regulation is a result of be only slightly attenuated or of similar virulence as
the ability to sense the nutritional status of the cell via the wild-type strains when evaluated in mouse infection
ppGpp (guanosine tetraphosphate) a molecule produced models [104,105].
Chapter | 108 Francisella 1997

Subversion of Extracellular Host Defences Another important host defence mechanism is the
cationic antimicrobial peptides defensins and cathelici-
For a successful intracellular pathogen, uptake can be dins [110]. These positively charged antimicrobial pep-
regarded as a major evasion mechanism to avoid extracel- tides are designed to disrupt the negatively charged
lular host defences. Still, it is clear that the ability to resist bacterial membrane and can be found both extracellu-
killing mechanisms and inflammatory responses is impor- larly on mucosal surfaces and within immune cells like
tant also for Francisella (Figure 108.3). Activation of the macrophages and neutrophils. The host defensins and
complement is a major defence mechanism where host cathelicidins appear to have a limited role in defence
recognition of a pathogen via antibodies or lections leads against Francisella. There are two apparently efficient
to activation with host signalling molecules that will pro- mechanisms by which the pathogens avoid these antimi-
mote inflammation and recruit phagocytes and other crobials. One intriguing mechanism is to lower the nega-
immune cells to the site of infection. In addition, comple- tive surface charge by modifying the lipid A moiety of
ment activation also leads to activation of killing mechan- LPS. Lipid A of Francisella lacks two negatively
isms [106]. Francisella avoids activation of complement charged phosphate groups that prevents the binding of
by binding to host factors like plasminogen/plasmin and the positively charged defensins [111]. The second
factor H and thereby prevents activation of both the kill- mechanism whereby Francisella can avoid these antimi-
ing mechanisms and a strong inflammatory response. This crobial peptides is efflux systems like the AcrAB/TolC
strategy also leads to complement-factor-mediated uptake efflux platform that promotes the active efflux of toxic
by phagocytic cells that is a better alternative to activate compounds and thereby preventing their antimicrobial
other powerful host defences [107,108]. The O-antigen action [112].
oligosaccharide, that is part of LPS and present on the
surface, is critical for the complement-inhibiting activity
and O-antigen mutants are also attenuated in animal INTRACELLULAR PHASE OF INFECTION
infection models [109].
Uptake/entry and Survival in the Phagosome
Francisella readily enter different host cells but macro-
phages appear to be the primary cell where it replicates
in vivo. Francisella is also very capable of replicating
inside phagocytes so uptake can also be seen as a strategy
to evade the extracellular innate host defence
(Figure 108.3). Uptake can be promoted via interaction
with different receptors, and binding triggers uptake via
membrane protrusions formed around the bacterium
called pseudopod loops [113]. The uptake appears to
occur via a pseudopod loop mechanism irrespective of
what receptor Francisella binds to and if the bacterium is
opsonized or not. While the overall mechanism appears to
be the same, the efficacy of uptake as well as intracellular
replication differs depending on the host cell receptor
involved. Serum or antibody opsonized bacteria are
mainly taken up via CR3 and FcγR-receptors. Uptake of
FIGURE 108.3 Francisella is a successful intracellular pathogen and opsonized bacteria is about tenfold more efficient than for
in line with this has mechanisms to subvert host defences at different
stages of the infection. For details, see the respective paragraph.
non-opsonized bacteria [114]. This could be an advantage
for an intracellular pathogen, but this route of uptake
1. Subversion of innate extracellular host defences, like complement comes at the price of a slower replication rate and delayed
and antimicrobial peptides
2. Multiple receptors mediate uptake and also affects the level of host
escape from the phagosome. Even though Francisella is
innate defences inside the phagosome fully capable of surviving in the hostile phagosomal envi-
3. Survival in the hostile host phagosome interference with ROS ronment, escape from the phagosome is a direct require-
production and ROS detoxification ment for successful infection of macrophages and in vivo
4. Mechanism to rapidly escape the phagosome mediated by FPI infection of animal hosts. It seems like the time
encoded T6SS-like mechanism
5. Intracellular replication without eliciting a strong inflammatory host
Francisella spends in the phagosome also depends on the
response avoiding TLR and NLR recognition mode of uptake. Opsonized bacteria on average spend a
6. Uptake into autophagosomes after successful intracellular cytosolic longer time within the Francisella-containing phagosome
replication role in human infection somewhat unclear (FCP). There are several lines of host defence that
1998 PART | 17 Animal and Ectoparasitic Source Infections

Francisella has to deal with once internalized and con- lipids of the bacterial cell membrane have been identi-
tained within FCP. fied [124]. Again, this verifies that Francisella has
Acidification of the phagosome is one mechanism overlapping mechanisms also to subvert the NADPH
used by the host that hampers replication of many bacte- oxidase and ROS, a key host defence against intracellu-
rial pathogens. At the same time there are many patho- lar pathogen.
gens that have specific strategies to handle and survive in
the acidic environment. The role of acidification for
Francisella is not entirely clear but very intriguing. While
Phagosomal Escape
there does not appear to be any direct link or requirement
of acidification for phagosomal escape, uptake of non- As discussed above Francisella is successful in surviv-
opsonized bacteria is followed by transient acidification ing the innate host defence mechanisms prior to uptake
15 30 minutes after uptake and rapid phagosomal escape as well as inside the FCP. At the same time it is clear
around 1 hour after infection [115,116]. Uptake mediated that until this point it is mainly about surviving, there is
via opsonization occurs with little or no acidification but little replication during these initial phases of infection.
in this case phagosomal escape is delayed in comparison Therefore, the last step, phagosomal escape whereby
with uptake without serum or antibody-mediated uptake the bacterium is released from the toxic phagosome
[113,117]. The significance of the transient acidification and reaches the cytosol, is an absolutely essential
after non-opsonized uptake is strengthened by the fact step. Mutants unable to escape the FCP are highly
that agents that block acidification also delay escape attenuated and essentially avirulent (Figure 108.3). The
while the acidification-blocking agents have no effect on timing of escape, as mentioned above, largely depends
escape after uptake of opsonized bacteria. Overall, this on the route of Francisella uptake where non-opsonized
illustrates that Francisella have a broad array of mechan- Francisella can escape as early as within 1 h after entry
isms to deal with host defence at different stages of while opsonized Francisella have been estimated to
infection. escape between 2 4 h after entry into the FCP
Francisella must also handle the reactive oxygen [114,125].
species (ROS) that are produced within the phagosome The mechanism of escape is not known; no
in response to infection (Figure 108.3). ROS molecules membrane-disrupting or pore-forming protein directly
are produced by a membrane-localized enzyme system, mediating the escape has been identified. Still it is very
the NADPH oxidase, which produces highly toxic clear that most of the genes encoded by the Francisella
superoxide anions from oxygen [118]. In response to pathogenicity island (FPI) are absolutely required. The
phagocytosis of a pathogen, the different subunits of the FPI which is duplicated in all subspecies except subspe-
enzyme assemble at the membrane to form the active cies novicida encodes a putative type VI secretion system
NADPH oxidase that then starts to produce ROS. (T6SS) and these genes are essential for the intracellular
Francisella species can interfere with ROS production lifestyle as well as for pathogenesis of Francisella
on different levels. One mechanism is to block the [126 128].
assembly of NADPH oxidase components. Four acid Of the 17 FPI proteins only three, IglA, IglB and
phosphatases (AcpA, AcpB, AcpC and Hap) are DotU, share significant homology to typical T6SS core
required for the ability to inhibit NADPH oxidase components [128] and no homologue of ClpV, the impor-
assembly and/or block ROS production by already- tant AAA 1 ATPase and putative energizer of T6SSs, has
assembled complexes [119]. Francisella species encode been identified. A potential VgrG homologue has been
at least six different acid phosphatases and three of identified in the FPI but the Walker A box commonly
those are truncated and may not be fully functional in present in IcmF homologues is missing from the F. tular-
subspecies tularensis (type A) strains. Their role in viru- ensis PdpB gene. These differences suggest that the
lence and intercellular replication is not entirely clear Francisella FPI system is clearly different compared to
but inactivation of four acid phosphatase genes in a type other T6SS. Even if these observations raise the question
A strain had a significant impact on intracellular repli- whether the FPI actually encodes a T6SS it is still clear
cation as well as on virulence [120]. Even if Francisella that some proteins encoded by FPI are secreted and possi-
is able to suppress activation of the NADPH oxidase, bly also reach the host cell cytosol from the bacteria
low levels of ROS are still produced. Francisella residing within the FCP and that this is related to phago-
encodes proteins that can directly detoxify ROS like cat- somal escape. Somehow FPI-encoded proteins are
alase [121 123]. secreted and can promote or induce disruption of the
In addition, a third class of proteins with similarity phagosomal membrane. More work is needed to elucidate
to Ohr proteins that mediate resistance to organic hydro- the molecular mechanisms of this essential virulence
peroxides created during the interaction of ROS with mechanism in Francisella pathogenesis.
Chapter | 108 Francisella 1999

Evasion of Host Recognition Outside Cells as the primary inducers of the TLR2 response. Even if
and Inside the Phagosome only a few lipoproteins have been directly identified as
TLR2 agonists [136] it is likely that the interplay between
The host defence includes an array of receptor molecules lipoproteins and TLR2 signalling is of major importance
(pathogen recognition receptors (PRRs)) specialized in as Francisella encodes many lipoproteins. There is also
detecting different conserved microbial components known substantial evidence that TLR2 is required for the early
as PAMPs (pathogen-associated molecular patterns) [129]. inflammatory response to Francisella infection, both from
The receptors that recognize these microbial components in vitro infected macrophages, as well as in vivo
can be found in the extracellular space and inside the phago- Francisella infections, where TLR2 has been shown to be
some are denoted Toll-like receptors (TLRs) and these can required for control of infection [135,137].
trigger phagocytosis and inflammatory signalling pathways. Francisella has also evolved ways to deal with the
The TLR signalling activates transcription factors like NF- induced TLR2-dependent signalling. It also seems to be
ĸB and IRF3 that in turn induce expression of inflammatory the case that infection of macrophages with Francisella
cytokines and type I interferons that activate innate and has been shown to be blocked from TLR2 and TLR4 acti-
adaptive immune cells [130]. The different TLRs recognize vation in response to E. coli lipoproteins and LPS and
different bacterial molecules; TLR2 is specific for bacterial infection with F. tularensis has also been shown to reduce
lipoproteins (BLPs) and peptidoglycan (PGN) [131]; TLR4 TLR2 expression [138,139]. Further, the ability of the bac-
for lipopolysaccharide (LPS) structures from Gram-negative teria to resist damage by antimicrobials and minimize the
bacteria; TLR5 for flagellin and TLR9 recognizes bacterial release of BLPs is another indirect but efficient way of
CpG DNA [130]. evading TLR2 signalling. An even more sophisticated
As discussed above most of Francisella’s successful mechanism to avoid TLR2 induction is to selectively use
strategy as a pathogen relies on the ability to avoid recog- the CRISPR/Cas to degrade the mRNA coding for a lipo-
nition and induce a strong host inflammatory response in protein known to induce TLR2 signalling [140]. CRISPR/
the host. A significant part of this strategy is linked to Cas (clustered regularly interspaced palindromic repeats/
successful evasion and/or suppression of inflammatory CRISPR-associated) systems are bacterial defence systems
signalling mediated by TLRs. For E. coli and many other against foreign nucleic acids derive, that use an array of
Gram-negative bacteria TLR4 is a primary sensor of small CRISPR RNAs (crRNAs) with repetitive sequences
infection. In contrast to many other pathogens Francisella flanking unique spacers to recognize their targets and then
LPS does not activate an efficient TLR4 response. One the Cas proteins to mediate degradation of target mRNA.
important difference is that Francisella actively removes This intricate mechanism is nicely illustrated and supported
or modifies LPS structures that induce a strong TLR4 by in vivo experiments showing that strains with a non-
response [132]. The importance of these modifications functional cas gene were highly attenuated as a result of
has been verified by showing that mutation of the gene inability to block lipoprotein expression and that lipopro-
that removes an additional 4phosphate group present in tein mutants rescued virulence [140].
lipid A in other bacteria that induce TLR4 responses also All this shows that TLR2 signalling is a major battle-
resulted in rapid clearance in an in vivo infection model field between host and pathogen that is very decisive for
[111]. the outcome of Francisella infections where the host uses
Similar to TLR4, TLR5, another important host sensor this pathway as a major innate defence mechanism and
of infection, is also not activated during infection, as where the pathogen has multitudes of mechanisms, more
Francisella does not encode flagellin [133]. or less sophisticated to subvert and deal with TLR2
Even though there is evidence supporting that signalling.
Francisella DNA is recognized by the TLR system,
TLR9 seems to be of limited importance for the in vivo
host response to Francisella [134]. It is possible that
Francisella avoids TLR9 activation by its resistance to
Immune Subversion at the Level of
host antimicrobial attacks both before uptake and within Intracellular Replication
the phagosome and thereby prevents release of bacterial After escape from the phagosome Francisella reaches the
DNA. Here it is likely that the rapid escape of Francisella host cell cytosol thus the pathogen has overcome TLRs
from the phagosome is critical for minimizing bacterial and harsh phagosomal defences. Even though this could
damage or killing and subsequent DNA release to prevent be seen as a major achievement, replication in the cytosol
activation of TLR9 (see also above). is also faced with a number of challenges. Until this point
Studies conducted so far indicate that TLR2 is the pri- bacterial replication has normally been limited, but in the
mary TLR involved in the host response to Francisella cytosol replicating to high numbers also increases the risk
infection [135]. So far lipoproteins have been identified of triggering an immune response. Bacterial replication
2000 PART | 17 Animal and Ectoparasitic Source Infections

could result in release of PAMPs that in turn could be rec- autophagosomes [150] (Figure 108.3). It is not clear
ognized by cytosolic PRRs (Figure 108.3). The host cell whether this is a host-induced response to control infec-
cytosol also has numerous PRRs capable of recognizing a tion or a mechanism induced by the pathogen to promote
large number of bacterial products and elicits a host infection. F. tularensis modulates the expression of
response to eliminate the invaders. The Nod-like receptor autophagy-related genes that could indicate that
(NLR) family responds to a diverse set of PAMPs includ- Francisella subverts this host defence system [151]. An
ing PGN (Nod1 and Nod2), flagellin (NLRC4, NAIP5 important thing to bear in mind, with regard to the
and NAIP6) [141] and damage induced by pore-forming significance of autophagy, is that it has only been
toxins (NLRP3) [142]. The role of Nods during observed in murine macrophages and has not yet been
Francisella infection is less well established than TLRs, observed in human macrophages [152,153].
but it is possible that they recognize and respond to PGN. The extensive and rapid replication in the cytosol of
The presence of less pathogenic subspecies novicida in host cells that occurs without induction of an inflamma-
the cytosol is sensed in a type I interferon (IFN)-depen- tory response is a major hallmark of the successful and
dent manner that activates the absent melanoma (AIM2)- essential immune subversion employed by F. tularensis.
containing inflammasome [143]. The AIM2 complex acti- Many of the molecular determinants and mechanisms that
vation leads to induction of secretion of proinflammatory promote the evasion of cytosolic defences remain to be
cytokines IL1ß and pro-IL18 and programmed inflamma- identified.
tory cell death by pyroptosis. This will recruit and activate
other immune cells that can promote bacterial clearance
[144]. Thereby the bacteria released into the extracellular
Nutritional Requirements and Defences
environment can be taken up by other cells like neutrophils An intracellular pathogen must be well adapted for intra-
that under these inducing conditions are not permissive for cellular replication and enough nutrients must be
replication [145]. The activation of the AIM2 inflamma- available and possible for Francisella to acquire, to
some is potentiated by TLR2 signalling and very important sustain high growth. The host on the other hand has
for eliminating Francisella from its intracellular prolifera- developed mechanisms that limit the availability of spe-
tion niche inside macrophages [146,147]. AIM2 has been cific nutrients, thereby establishing defence against patho-
shown to recognize double-stranded DNA and at least for gens at the nutritional level and the battle for nutrients is
subspecies novicida the AIM2 activation has been found to another critical aspect of host pathogen interactions.
be linked to release of bacterial DNA in the cytosol [147]. Several specific metabolic pathways critical to
It is important to bear in mind that these studies have Francisella replication in the host, such as the biosynthe-
been performed in murine systems and the less virulent sis of purines and pyrimidines, gluconeogenesis and gly-
subspecies novicida. In addition, macrophages infected colysis have been identified [154]. The host also actively
with the highly virulent subspecies tularensis secrete only limits some nutrients during infection and for these the
low levels of the inflammasome dependent cytokine pathogen needs to develop strategies to directly access
interleukin (IL)-18 and dendritic cells infected with these nutrients.
F. tularensis fails to efficiently activate the inflamma- Iron is necessary for a variety of enzymatic functions
some [148]. Still, the findings strongly support that TLR2 in bacteria and is a vital component of various redox reac-
and IFN signalling are the two major host defence path- tions that take place during growth. Within the host, most
ways in controlling Francisella infections and both con- of the total iron is found in the intracellular space [155].
tribute to inflammasome activation. It is likely that the The iron outside host cells is associated with iron storage
higher virulence of type A strains, at least in part, is the molecules that bind iron with very high affinity like
result of having additional ways to limit or prevent haem, lactoferrin and transferrin. In principle Francisella
inflammasome activation. in part overcomes the barrier to iron acquisition when it
It is not clear how Francisella after extensive replica- enters the host cell, where the iron content is significantly
tion exits from infected host cells and disseminates higher. Also within host cells, iron-containing redox
throughout the host without evoking inflammation. These enzymes and iron-storage proteins sequester iron and
infected macrophages could serve as Trojan horses, traf- therefore iron acquisition for the pathogen is still very dif-
ficking the bacteria to promote systemic spread. ficult [156,157]. In addition, the host also sequesters iron
Xenophagy is another host strategy to capture intracel- away in response to infection [158]. In response to the
lular microbes within a double-membrane vacuole, the challenges from the host, Francisella utilizes a
autophagosome, for delivery and degradation in the lyso- rhizoferrin-like siderophore [159]. Siderophores are small
somal compartment [149]. After long periods of cytosolic molecules that bind iron with very high affinity and can
replication (.18 hours), some of the intracellular compete for iron from the host iron-sequestering com-
Francisella become enclosed and reside inside pounds [160]. The Francisella genes that encode the iron-
Chapter | 108 Francisella 2001

sequestering system are fslABCDE (also termed via non-activating receptors, escape of phagosomal killing
figABCDE). These genes encode the proteins that provide and modulation of cytosolic defence pathways too.
production as well as export of the Francisella sidero- One important observation is that if macrophages are
phore [159,161]. FslE is not involved in siderophore pro- activated by the pro-inflammatory cytokine IFN-gamma,
duction but is instead required for siderophore binding they become highly capable of preventing its replication
and/or import [161,162]. Francisella appears to lack the in the cytosol [174,175]. IFN-gamma that is primarily
highly conserved siderophore receptor and importer genes produced by NK cells and dendritic cells during infec-
tonB, exbB and exbD genes found in many bacteria [163]. tion regulates more than 200 genes. Many of these are
Instead there are two fslE orthologues present in the involved in enhancing nitric oxide production, inducing
genome that may promote siderophore import that are autophagy, and promoting enhanced major histocompati-
denoted fupAB and in the type B vaccine strain LVS a bility complex (MHC) class I and II antigen presentation
deletion event has resulted in these two genes being fused [176,177]. All these pathways could enhance clearance
to a single protein. This deletion event is also linked to of Francisella and promote host survival. In line with
significant virulence attenuation indicating the importance this lack of IFN-gamma in vivo leads to increased suscepti-
of this gene for siderophore import [162,164,165]. It is bility of mice to Francisella infection [175,178]. F. tular-
clear that siderophore production and iron acquisition pro- ensis also induces production of immunomodulatory lipid
teins are vital for Francisella to maintain iron homeosta- prostaglandin E2 (PGE2) in macrophages [179]. PGE2
sis and to survive and sustain replication in the host. The lowers IFN-gamma production from T cells and it also
importance of fslABC, fupAB and feoB, a ferrous iron induces the expression of a host factor that directs
importer [159,166] have each been verified by in vivo vir- ubiquitin-dependent lysosomal degradation of MHC class
ulence screening or by direct mutagenesis identified in II molecules, thereby significantly lowering the number of
in vivo virulence screens in macrophages or in animal MHC class II on the surface of the macrophages [180].
models [167,168]. Thereby Francisella also affects the induction of adaptive
The host also limits the intracellular concentration of immune responses by limiting antigen presentation through
the amino acid tryptophan in response to infection [169]. MHC class II.
Francisella overcomes this nutritional host defence by its Dendritic cells produce cytokines that control T-cell
ability to synthesize tryptophan de novo [170] and strains activation and the immune responses that they then pro-
with mutations in genes involved in tryptophan biosynthe- mote in induction of cell-mediated immunity. F. tularen-
sis are also significantly attenuated in vivo [171,172] con- sis can suppresses proinflammatory cytokine production
firming the importance of tryptophan prototrophy for from dendritic cells [181] and one of these, IL-12, is
Francisella pathogenesis. Francisella has several muta- important for the development of IFN-gamma-producing
tions in genes encoding enzymes required for several bio- T cells [182].
synthetic pathways [154,173] and as a consequence it is F. tularensis also subverts dendritic cell function to
auxotrophic for 13 amino acids, i.e. Francisella can not make them produce anti-inflammatory cytokines like
de novo synthesize these amino acids. The only amino transforming growth factor beta (TGF-β) and IL-10
acids that Francisella can synthesize de novo are alanine, [179,181], which also contributes to the lack of an early
glutamate, glutamine, asparagine, glycine, phenylalanine inflammatory response as IL-10 inhibits macrophage pro-
and tryptophan [154,163,173]. Therefore, the ability to liferation and proinflammatory cytokine production [183].
acquire these essential amino acids within the host is also This can also lead to reduced MHC class II expression
essential for pathogenesis. and further suppression of the adaptive immune response
[184] and they can also promote development of regula-
tory T cells that in turn can suppress the inflammatory
EVASION OF DEVELOPMENT OF activity of other T cells [185].
The molecular mechanisms of the F. tularensis-medi-
ADAPTIVE IMMUNITY ated redirection of APCs towards an anti-inflammatory
As discussed above F. tularensis efficiently evades early response are not known but inducing immune tolerance
immune responses and can also modulate the activity of through the induction of IL-10 or TGF-β is a strategy also
important innate immune cells. Macrophages and den- used by important pathogens such as Yersinia pestis,
dritic cells have a very important role as antigen- Coxiella burnetii and Chlamydia pneumoniae [186].
presenting cells (APCs) and the ability of these cells to Cell-mediated immunity is regarded as essential for
present microbial antigens and activate T cells is a key controlling intracellular bacterial infection. It is appar-
link between the innate and adaptive immune systems. As ent that Francisella encodes multiple strategies to early
discussed above Francisella uses several different modulate APCs that can interfere with activation of the
mechanisms to subvert macrophage defences like entry adaptive immune system. This provides an environment
2002 PART | 17 Animal and Ectoparasitic Source Infections

that promotes replication in subsequent phases of TULARAEMIA VACCINE DEVELOPMENT


infection by interfering with the activation of CD8 T
cells, which could kill infected host cells harbouring One of the long-term goals with tularaemia research has
Francisella. been to develop a vaccine against the disease. Currently,
there is no licensed vaccine for general use against tular-
aemia, but vaccine development has become a renewed
priority since the anthrax letters in the aftermath of the
INFECTION MODELS 9/11 events in 2001. Vaccine efforts against tularaemia
Unlike some intracellular pathogens, F. tularensis natu- started in the 1930s and since then two different types of
rally infects a wide variety of mammals, including tularaemia vaccine have been studied in humans. The
humans and rodents, via multiple routes including the Foshay vaccine, in honour of the inventor, Dr. Lee
respiratory and gastrointestinal tracts, the skin and the Foshay, consisted of chemically killed (phenol-killed)
conjunctiva [1]. An advantage with this is that there are F. tularensis and was effective at reducing the incidence
many different natural infections and infection routes that of laboratory-acquired tularaemia cases from 100% to
can be used in research to study naturally occurring infec- 30% in the 1950s (Box 108.1) [198]. However, this vac-
tion. Importantly, there are many significant differences cine provided only minimal protection against aerosol
when hosts and routes for infections are used. challenge with type A Francisella. At the same time a
The small animal models used; mouse, guinea pig, live vaccine strain (LVS) derived from type B
rabbit and hamster, also differ in sensitivity. The viru- Francisella (F. tularensis subsp. holarctica) was used to
lence of the attenuated live vaccine strain, LVS (subsp. immunize millions of individuals in the former Soviet
holarctica) in the most commonly used animal model, Union. Samples of the Russian vaccine strain which was
mice, has been shown to be dependent on the route of attenuated by several in vitro passages were given to the
delivery, while the subsp. tularensis strain SCHU S4 is United States as part of a formal scientific exchange pro-
fully virulent regardless of the route of infection. To date, gramme. However, tularaemia in the former Soviet
none of the small animal models described in the litera- Union was exclusively caused by type B subsp. holarcti-
ture behaves as the human host with respect to their sus- ca strains and therefore the efficacy of the Russian vac-
ceptibility to different subspecies and strains like cine against type A, subsp. tularensis was unknown.
SCHU S4 (type A) or LVS (attenuated type B). The fact During the 1950s and 1960s, the LVS vaccine was fur-
that the vaccine strain LVS is still virulent in mice makes ther characterized and after several passages in labora-
this model limited for evaluation of virulence properties tory media and through mice, the strain was finally
for the human infection [187 189]. Further, F. novicida, designated LVS [199].
which is essentially avirulent in humans, is more virulent This vaccine provided better protection against aerosol
in the mouse infection model compared to LVS (type B) challenge with type A Francisella than the Foshay-
[190,191]. Because of these differences and limitations vaccine and has ever since been used to vaccinate at-risk
there is a need to develop and use other animal models. staff (laboratory and military personnel) in some coun-
Non-human primates are important and highly relevant tries. The vaccine confers a long-lasting humoral and cell-
for vaccine and protection studies. The LVS vaccine mediated immunity that is effective for up to 25 years
strain provides protection in monkeys in a similar way as [200]. This is in accordance with the protective immunity
in humans [192,193]. Still, the model is not ideal as mon- obtained after a natural infection [201 203].
keys are more sensitive to infections with type B strains
compared to humans [194]. A rat model has also been
used but is less well established. Rats are less susceptible
Box 108.1 Laboratory-Acquired Tularaemia
to LVS but instead they appear to be more resistant to
SCHU S4 (type A) that may limit their use [195]. Tularaemia is caused by F. tularensis and due to the very
Arthropod vectors are important transmission routes low infection dose and risk for aerosolization during han-
dling of bacterial cultures and plates, there is a relatively
for tularaemia, and the fruitfly Drosophila melanogaster
high frequency of accidental infection of laboratory person-
has recently been included in the list of possible in vivo nel exposed to F. tularensis. One illustration of this is when
models of Francisella infections. F. tularensis can spread 12 microbiology laboratory personnel were exposed to
throughout the tissues of the fly but it is not yet known F. tularensis, and received prophylactic antibiotics [197].
how Francisella survives the efficient arthropod immune To avoid this type of accident it is very important that labo-
response. This model might be valuable as it can enable ratory personnel are well trained and informed about the
studies of bacterial survival and virulence in an arthropod high risk for aerosol exposure and that appropriate protec-
host and also provide insight into vector-borne transmis- tive measures are implemented routinely.
sion [196].
Chapter | 108 Francisella 2003

LVS is administered by scarification, and induces a few as ten organisms can be sufficient to establish an
high level of protection against type B infections, while infection in humans that if untreated could be fatal.
protection against type A is more limited [199]. LVS vac- The success of F. tularensis as a pathogen lies in its
cination by aerosol, i.e. administration directly into the unique ability to adapt a lifestyle as an intracellular patho-
lungs, improved the efficacy against respiratory challenge gen and to very timely subvert host defences both at
but also elicited serious side effects [204]. However, extracellular and intracellular levels. Key events in the
although the LVS vaccine has been used as prophylaxis in infection process are the ability to rapidly escape the pha-
a large number of people over the last few decades, it has gosome after uptake by phagocytic cells and the ability to
not yet been licensed for general use. Major obstacles for replicate to high levels inside the host cells without evok-
approval are concerns regarding its undefined attenuation, ing a host inflammatory response.
mechanism of protection and reversion frequency [203].
Recent studies using the mouse infection model have
revealed that the virulence attenuation of LVS is the REFERENCES
result of two gene-deletion events, one affecting a pilin
[1] Keim P, Johansson A, Wagner DM. Molecular epidemiology,
gene and the other a protein mediating iron uptake [103].
evolution, and ecology of Francisella. Ann N Y Acad Sci
The total number of clinical type A cases (type A
2007;1105:30 66.
restricted to North America) is low but due to renewed [2] Morner T. The ecology of tularaemia. Rev Sci Tech
concerns regarding the threat of bioterrorism, there has 1992;11:1123 30.
been an increased interest in licensing a tularaemia vac- [3] Tarnvik A, Sandstrom G, Sjostedt A. Epidemiological analysis of
cine for general use. Therefore, the goal has been to tularemia in Sweden 1931 1993. FEMS Immunol Med Microbiol
develop a Francisella vaccine that can also provide pro- 1996;13:201 4.
tection from an aerosol challenge, i.e., the pneumonic [4] Petersen JM, Mead PS, Schriefer ME. Francisella tularensis: an
form of the disease, in the event of a deliberate release in arthropod-borne pathogen. Vet Res 2009;40:7.
a biological attack. [5] Berdal BP, Mehl R, Meidell NK, Lorentzen-Styr AM, Scheel O.
New experimental vaccines, including novel formula- Field investigations of tularemia in Norway. FEMS Immunol Med
Microbiol 1996;13:191 5.
tions of LVS, live (and defined) attenuated type A strains,
[6] Abd H, Johansson T, Golovliov I, Sandstrom G, Forsman M.
killed whole cell, and subunit vaccine candidates are
Survival and growth of Francisella tularensis in Acanthamoeba
currently developed and under evaluation [12]. Subunit- castellanii. Appl Environ Microbiol 2003;69:600 6.
based vaccines would be preferable from a safety [7] Saslaw S, Eigelsbach HT, Wilson HE, Prior JA, Carhart S.
perspective but have proven not to confer high levels of Tularemia vaccine study. I. Intracutaneous challenge. Arch Intern
protection. This is also the experience from other intracel- Med 1961;107:689 701.
lular bacterial pathogens. The most common strategy for [8] Saslaw S, Eigelsbach HT, Prior JA, Wilson HE, Carhart S.
the on-going vaccine development is to find ways to Tularemia vaccine study. II. Respiratory challenge. Arch Intern
attenuate type A strains to be both safe and provide high Med 1961;107:702 14.
levels of protection [205]. [9] Darling RG, Catlett CL, Huebner KD, Jarrett DG. Threats in bio-
A vaccine must elicit both innate immunity and immu- terrorism. I: CDC category A agents. Emerg Med Clin North Am
2002;20:273 309.
nological memory, i.e. persist long enough to develop a
[10] Dennis DT, Inglesby TV, Henderson DA, Bartlett JG, Ascher MS,
strong adaptive immunity. Live attenuated vaccines would
Eitzen E, Working Group on Civilian, B., et al. Tularemia as a
be able to elicit both and confer protection. biological weapon: medical and public health management.
JAMA 2001;285:2763 73.
[11] Oyston PC, Sjostedt A, Titball RW. Tularaemia: bioterrorism
CONCLUSIONS defence renews interest in Francisella tularensis. Nat Rev
Microbiol 2004;2:967 78.
Francisella tularensis, the causative agent of tularaemia, [12] Conlan JW. Tularemia vaccines: recent developments and remain-
is a zoonotic intracellular pathogen that can be found in a ing hurdles. Future Microbiol 2011;6:391 405.
very large number of species ranging from large mam- [13] Hollis DG, Weaver RE, Steigerwalt AG, Wenger JD, Moss CW,
mals and vertebrates to invertebrates, arthropods and Brenner DJ. Francisella philomiragia comb. nov. (formerly
amoebas. Disease in humans often occurs in parallel with Yersinia philomiragia) and Francisella tularensis biogroup novici-
da (formerly Francisella novicida) associated with human disease.
tularaemia in wild animals.
J Clin Microbiol 1989;27:1601 8.
Human infection can occur through aerosolization,
[14] Mailman TL, Schmidt MH. Francisella philomiragia adenitis and
direct contact with infected animals and via arthropod pulmonary nodules in a child with chronic granulomatous disease.
vectors like ticks, mosquitos and biting flies. Can J Infect Dis Med Microbiol 2005;16:245 8.
F. tularensis subspecies tularensis (type A) is one of [15] Wenger JD, Hollis DG, Weaver RE, Baker CN, Brown GR,
the most infectious bacteria known and inhalation of as Brenner DJ, et al. Infection caused by Francisella philomiragia
2004 PART | 17 Animal and Ectoparasitic Source Infections

(formerly Yersinia philomiragia). A newly recognized human [31] Sandstrom G, Sjostedt A, Forsman M, Pavlovich NV, Mishankin
pathogen. Ann Intern Med 1989;110:888 92. BN. Characterization and classification of strains of Francisella
[16] Brevik OJ, Ottem KF, Nylund A. Multiple-locus, variable number tularensis isolated in the central Asian focus of the Soviet Union
of tandem repeat analysis (MLVA) of the fish-pathogen and in Japan. J Clin Microbiol 1992;30:172 5.
Francisella noatunensis. BMC Vet Res 2011;7:5. [32] Johansson A, Celli J, Conlan W, Elkins KL, Forsman M, Keim
[17] Ottem KF, Nylund A, Karlsbakk E, Friis-Moller A, Kamaishi T. PS, et al. Objections to the transfer of Francisella novicida to the
Elevation of Francisella philomiragia subsp. noatunensis subspecies rank of Francisella tularensis. Int J Syst Evol
Mikalsen et al. (2007) to Francisella noatunensis comb. nov. [syn. Microbiol 2010;60:1717 8; author reply 1718 20.
Francisella piscicida Ottem et al. (2008) syn. nov.] and character- [33] Busse HJ, Huber B, Anda P, Escudero R, Scholz HC, Seibold E,
ization of Francisella noatunensis subsp. orientalis subsp. nov., et al. Objections to the transfer of Francisella novicida to the sub-
two important fish pathogens. J Appl Microbiol species rank of Francisella tularensis - response to Johansson
2009;106:1231 43. et al. Int J Syst Evol Microbiol 2010;60:1718 20.
[18] Birkbeck TH, Feist SW, Verner-Jeffreys DW. Francisella infec- [34] Farlow J, Wagner DM, Dukerich M, Stanley M, Chu M, Kubota
tions in fish and shellfish. J Fish Dis 2011;34:173 87. K, et al. Francisella tularensis in the United States. Emerg Infect
[19] Colquhoun DJ, Duodu S. Francisella infections in farmed and Dis. 2005;11:1835 41.
wild aquatic organisms. Vet Res 2011;42:47. [35] Staples JE, Kubota KA, Chalcraft LG, Mead PS, Petersen JM.
[20] Olsen AB, Mikalsen J, Rode M, Alfjorden A, Hoel E, Straum-Lie Epidemiologic and molecular analysis of human tularemia, United
K, et al. A novel systemic granulomatous inflammatory disease in States, 1964 2004. Emerg Infect Dis 2006;12:1113 8.
farmed Atlantic cod, Gadus morhua L., associated with a bacte- [36] Kugeler KJ, Mead PS, Janusz AM, Staples JE, Kubota KA,
rium belonging to the genus Francisella. J Fish Dis Chalcraft LG, et al. Molecular epidemiology of Francisella tular-
2006;29:307 11. ensis in the United States. Clin Infect Dis 2009;48:863 70.
[21] Nylund A, Ottem KF, Watanabe K, Karlsbakk E, Krossoy B. [37] Barns SM, Grow CC, Okinaka RT, Keim P, Kuske CR. Detection
Francisella sp. (Family Francisellaceae) causing mortality in of diverse new Francisella-like bacteria in environmental samples.
Norwegian cod (Gadus morhua) farming. Arch Microbiol Appl Environ Microbiol 2005;71:5494 500.
2006;185:383 92. [38] Petersen JM, Carlson J, Yockey B, Pillai S, Kuske C, Garbalena
[22] Ostland VE, Stannard JA, Creek JJ, Hedrick RP, Ferguson HW, G, et al. Direct isolation of Francisella spp. from environmental
Carlberg JM, et al. Aquatic Francisella-like bacterium associ- samples. Lett Appl Microbiol 2009;48:663 7.
ated with mortality of intensively cultured hybrid striped [39] Schrallhammer M, Schweikert M, Vallesi A, Verni F, Petroni G.
bass Morone chrysops x M. saxatilis. Dis Aquat Organ Detection of a novel subspecies of Francisella noatunensis as
2006;72:135 45. endosymbiont of the ciliate Euplotes raikovi. Microb Ecol
[23] Kamaishi T, Miwa S, Goto E, Matsuyama T, Oseko N. Mass mor- 2011;61:455 64.
tality of giant abalone Haliotis gigantea caused by a Francisella [40] Sjodin A, Svensson K, Ohrman C, Ahlinder J, Lindgren P, Duodu
sp. bacterium. Dis Aquat Organ 2010;89:145 54. S, et al. Genome characterisation of the genus Francisella reveals
[24] Escudero R, Elia M, Saez-Nieto JA, Menendez V, Toledo A, insight into similar evolutionary paths in pathogens of mammals
Royo G, et al. A possible novel Francisella genomic species iso- and fish. BMC Genomics 2012;13:268.
lated from blood and urine of a patient with severe illness. Clin [41] Scoles GA. Phylogenetic analysis of the Francisella-like endo-
Microbiol Infect 2010;16:1026 30. symbionts of Dermacentor ticks. J Med Entomol
[25] Whipp MJ, Davis JM, Lum G, de Boer J, Zhou Y, Bearden SW, 2004;41:277 86.
et al. Characterization of a novicida-like subspecies of Francisella [42] Kugeler KJ, Gurfield N, Creek JG, Mahoney KS, Versage JL,
tularensis isolated in Australia. J Med Microbiol Petersen JM. Discrimination between Francisella tularensis and
2003;52:839 42. Francisella-like endosymbionts when screening ticks by PCR.
[26] Kugeler KJ, Mead PS, McGowan KL, Burnham JM, Hogarty MD, Appl Environ Microbiol 2005;71:7594 7.
Ruchelli E, et al. Isolation and characterization of a novel [43] Tarnvik A, Berglund L. Tularemia. Eur Respir J 2003;21:361 73.
Francisella sp. from human cerebrospinal fluid and blood. J Clin [44] Jones RM, Nicas M, Hubbard A, Sylvester MD, Reingold A. The
Microbiol 2008;46:2428 31. infectious dose of Francisella tularensis (Tularemia). Appl
[27] Johansson A, Farlow J, Larsson P, Dukerich M, Chambers E, Biosafety 2005;10:227 39.
Byström M, et al. Worldwide genetic relationships among [45] Ellis J, Oyston PCF, Green M, Titball RW. Tularemia. Clin
Francisella tularensis isolates determined by multiple-locus vari- Microbiol Rev 2002;15:631 46.
able-number tandem repeat analysis. J Bacteriol 2004;186: [46] Goethert HK, Telford 3rd SR. Differential mortality of dog tick
5808 18. vectors due to infection by diverse Francisella tularensis tularen-
[28] Jackson J, McGregor A, Cooley L, Ng J, Brown M, Ong CW, sis genotypes. Vector Borne Zoonotic Dis 2011;11:1263 8.
et al. Francisella tularensis Subspecies holarctica, Tasmania, [47] Li J, Ryder C, Mandal M, Ahmed F, Azadi P, Snyder DS, et al.
Australia, 2011. Emerg Infect Dis 2012;18:1484 6. Attenuation and protective efficacy of an O-antigen-deficient mutant
[29] Olsufjev NG, Meshcheryakova IS. Infraspecific taxonomy of tula- of Francisella tularensis LVS. Microbiology 2007;153:3141 53.
remia agent Francisella tularensis McCoy et Chapin. J Hyg [48] Francis E. Tularemia. JAMA 1925;84:7.
Epidemiol Microbiol Immunol 1982;26:291 9. [49] Helvaci S, Gedikoglu S, Akalin H, Oral HB. Tularemia in Bursa,
[30] Ellis J, Oyston PC, Green M, Titball RW. Tularemia. Clin Turkey: 205 cases in ten years. Eur J Epidemiol 2000;16:
Microbiol Rev 2002;15:631 46. 271 6.
Chapter | 108 Francisella 2005

[50] Kantardjiev T, Ivanov I, Velinov T, Padeshki P, Popov B, Nenova [69] Baker CN, Hollis DG, Thornsberry C. Antimicrobial susceptibility
R, et al. Tularemia outbreak, Bulgaria, 1997 2005. Emerg Infect testing of Francisella tularensis with a modified Mueller-Hinton
Dis 2006;12:678 80. broth. J Clin Microbiol 1985;22:212 5.
[51] Meric M, Sayan M, Dundar D, Willke A. Tularaemia outbreaks in [70] Johansson A, Berglund L, Gothefors L, Sjostedt A, Tarnvik A.
Sakarya, Turkey: case-control and environmental studies. Ciprofloxacin for treatment of tularemia in children. Pediatr Infect
Singapore Med J 2010;51:655 9. Dis J 2000;19:449 53.
[52] Tarnvik A, Priebe HS, Grunow R. Tularaemia in Europe: an epi- [71] Piercy T, Steward J, Lever MS, Brooks TJ. In vivo efficacy of
demiological overview. Scand J Infect Dis 2004;36:350 5. fluoroquinolones against systemic tularaemia infection in mice. J
[53] Willke A, Meric M, Grunow R, Sayan M, Finke EJ, Splettstosser Antimicrob Chemother 2005;56:1069 73.
W, et al. An outbreak of oropharyngeal tularaemia linked to natu- [72] Mills E, Gold R, Thipphawong J, Barreto L, Guasparini R,
ral spring water. J Med Microbiol 2009;58:112 6. Meekison W, et al. Safety and immunogenicity of a combined five-
[54] Matyas BT, Nieder HS, Telford 3rd SR. Pneumonic tularemia on component pertussis-diphtheria-tetanus-inactivated poliomyelitis-
Martha’s Vineyard: clinical, epidemiologic, and ecological charac- Haemophilus B conjugate vaccine administered to infants at two,
teristics. Ann N Y Acad Sci 2007;1105:351 77. four and six months of age. Vaccine 1998;16:576 85.
[55] Brett M, Doppalapudi A, Respicio-Kingry LB, Myers D, Husband [73] Steward J, Piercy T, Lever MS, Simpson AJ, Brooks TJ.
B, Pollard K, et al. Francisella novicida bacteremia after a near- Treatment of murine pneumonic Francisella tularensis infection
drowning accident. J Clin Microbiol 2012;50:2826 9. with gatifloxacin, moxifloxacin or ciprofloxacin. Int J Antimicrob
[56] Titball RW, Sjostedt A, Pavelka Jr. MS, Nano FE. Biosafety and Agents 2006;27:439 43.
selectable markers. Ann N Y Acad Sci 2007;1105:405 17. [74] McCaffrey RL, Allen LA. Francisella tularensis LVS evades kill-
[57] Bevanger L, Maeland JA, Naess AI. Agglutinins and antibodies to ing by human neutrophils via inhibition of the respiratory burst
Francisella tularensis outer membrane antigens in the early diag- and phagosome escape. J Leukoc Biol 2006;80:1224 30.
nosis of disease during an outbreak of tularemia. J Clin Microbiol [75] Hall JD, Craven RR, Fuller JR, Pickles RJ, Kawula TH. Francisella
1988;26:433 7. tularensis replicates within alveolar type II epithelial cells in vitro
[58] Brown SL, McKinney FT, Klein GC, Jones WL. Evaluation of a and in vivo following inhalation. Infect Immun 2007;75:1034 9.
safranin-O-stained antigen microagglutination test for Francisella [76] Sundaresh S, Randall A, Unal B, Petersen JM, Belisle JT, Hartley
tularensis antibodies. J Clin Microbiol 1980;11:146 8. MG, et al. From protein microarrays to diagnostic antigen discov-
[59] Grunow R, Splettstoesser W, McDonald S, Otterbein C, O’Brien ery: a study of the pathogen Francisella tularensis. Bioinformatics
T, Morgan C, et al. Detection of Francisella tularensis in biologi- 2007;23:i508 18.
cal specimens using a capture enzyme-linked immunosorbent [77] Cole LE, Elkins KL, Michalek SM, Qureshi N, Eaton LJ,
assay, an immunochromatographic handheld assay, and a PCR. Rallabhandi P, et al. Immunologic consequences of Francisella
Clin Diagn Lab Immunol 2000;7:86 90. tularensis live vaccine strain infection: role of the innate immune
[60] Koskela P, Salminen A. Humoral immunity against Francisella response in infection and immunity. J Immunol 2006;176:6888 99.
tularensis after natural infection. J Clin Microbiol 1985;22:973 9. [78] Koskela P, Herva E. Cell-mediated and humoral immunity
[61] Sato T, Fujita H, Ohara Y, Homma M. Microagglutination test for induced by a live Francisella tularensis vaccine. Infect Immun
early and specific serodiagnosis of tularemia. J Clin Microbiol 1982;36:983 9.
1990;28:2372 4. [79] Forestal CA, Malik M, Catlett SV, Savitt AG, Benach JL, Sellati
[62] Tarnvik A, Lofgren S, Ohlund L, Sandstrom G. Detection of anti- TJ, et al. Francisella tularensis has a significant extracellular
gen in urine of a patient with tularemia. Eur J Clin Microbiol phase in infected mice. J Infect Dis 2007;196:134 7.
1987;6:318 9. [80] Sjostedt A, Sandstrom G, Tarnvik A, Jaurin B. Molecular cloning
[63] Sjöstedt A, Eriksson U, Berglund L, Tarnvik A. Detection of and expression of a T-cell stimulating membrane protein of
Francisella tularensis in ulcers of patients with tularemia by PCR. Francisella tularensis. Microb Pathog 1989;6:403 14.
J Clin Microbiol 1997;35:1045 8. [81] Molins CR, Carlson JK, Coombs J, Petersen JM. Identification of
[64] Johansson A, Ibrahim A, Göransson I, Eriksson U, Gurycova D, Francisella tularensis subsp. tularensis A1 and A2 infections by
Clarridge 3rd JE, et al. Evaluation of PCR-based methods real-time polymerase chain reaction. Diagn Microbiol Infect Dis
for discrimination of Francisella species and subspecies and 2009;64:6 12.
development of a specific PCR that distinguishes the two major [82] Champion MD, Zeng Q, Nix EB, Nano FE, Keim P, Kodira CD,
subspecies of Francisella tularensis. J Clin Microbiol 2000;38: et al. Comparative genomic characterization of Francisella tular-
4180 5. ensis strains belonging to low and high virulence subspecies.
[65] Eliasson H, Broman T, Forsman M, Back E. Tularemia: current PLoS Pathog 2009;5:e1000459.
epidemiology and disease management. Infect Dis Clin North Am [83] Beier D, Gross R. Regulation of bacterial virulence by two-
2006;20:289 311, ix. component systems. Curr Opin Microbiol 2006;9:143 52.
[66] Francis E. Streptomycin in treatment of tularemia. Trans Assoc [84] Bell BL, Mohapatra NP, Gunn JS. Regulation of virulence gene
Am Physicians 1947;60:181 6. transcripts by the Francisella novicida orphan response regulator
[67] Maurin M, Mersali NF, Raoult D. Bactericidal activities of anti- PmrA: role of phosphorylation and evidence of MglA/SspA inter-
biotics against intracellular Francisella tularensis. Antimicrob action. Infect Immun 2010;78:2189 98.
Agents Chemother 2000;44:3428 31. [85] Hrstka R, Stulik J, Vojtesek B. The role of MAPK signal path-
[68] Sanford JP. Landmark perspective: Tularemia. JAMA 1983; ways during Francisella tularensis LVS infection-induced apopto-
250:3225 6. sis in murine macrophages. Microbes Infect 2005;4:4.
2006 PART | 17 Animal and Ectoparasitic Source Infections

[86] Baron GS, Nano FE. MglA and MglB are required for the intra- of a type IV pilin gene results in major virulence attenuation of
macrophage growth of Francisella novicida. Mol Microbiol Francisella tularensis. Mol Microbiol 2006;59:1818 30.
1998;29:247 59. [102] Salomonsson EN, Forslund AL, Forsberg A. Type IV Pili in
[87] Bonquist L, Lindgren H, Golovliov I, Guina T, Sjostedt A. MglA Francisella - A Virulence Trait in an Intracellular Pathogen.
and Igl proteins contribute to the modulation of Francisella Front Microbiol 2011;2:29.
tularensis live vaccine strain-containing phagosomes in murine [103] Salomonsson E, Kuoppa K, Forslund AL, Zingmark C,
macrophages. Infect Immun 2008;76:3502 10. Golovliov I, Sjostedt A, et al. Reintroduction of two deleted viru-
[88] Lauriano CM, Barker JR, Yoon SS, Nano FE, Arulanandam BP, lence loci restores full virulence to the live vaccine strain of
Hassett DJ, et al. MglA regulates transcription of virulence factors Francisella tularensis. Infect Immun 2009;77:3424 31.
necessary for Francisella tularensis intraamoebae and intrama- [104] Forslund AL, Salomonsson EN, Golovliov I, Kuoppa K, Michell
crophage survival. Proc Natl Acad Sci USA 2004;101:4246 9. S, Titball R, et al. The type IV pilin, PilA, is required for full vir-
[89] Charity JC, Costante-Hamm MM, Balon EL, Boyd DH, Rubin EJ, ulence of Francisella tularensis subspecies tularensis. BMC
Dove SL. Twin RNA polymerase-associated proteins control viru- Microbiol 2010;10:227.
lence gene expression in Francisella tularensis. PLoS Pathog [105] Ark NM, Mann BJ. Impact of Francisella tularensis pilin homo-
2007;3:e84. logs on pilus formation and virulence. Microb Pathog
[90] Guina T, Radulovic D, Bahrami AJ, Bolton DL, Rohmer L, 2011;51:110 20.
Jones-Isaac KA, et al. MglA regulates Francisella tularensis [106] Rus H, Cudrici C, Niculescu F. The role of the complement sys-
subsp. novicida (Francisella novicida) response to starvation and tem in innate immunity. Immunol Res 2005;33:103 12.
oxidative stress. J Bacteriol 2007;189:6580 6. [107] Ben Nasr A, Klimpel GR. Subversion of complement activation at
[91] Brotcke A, Monack DM. Identification of fevR, a novel regulator the bacterial surface promotes serum resistance and opsonophago-
of virulence gene expression in Francisella novicida. Infect cytosis of Francisella tularensis. J Leukoc Biol 2008;84:77 85.
Immun 2008;76:3473 80. [108] Clinton SR, Bina JE, Hatch TP, Whitt MA, Miller MA. Binding
[92] Buchan BW, McCaffrey RL, Lindemann SR, Allen LA, Jones and activation of host plasminogen on the surface of Francisella
BD. Identification of migR, a regulatory element of the tularensis. BMC Microbiol 2010;10:76.
Francisella tularensis live vaccine strain iglABCD virulence [109] Clay CD, Soni S, Gunn JS, Schlesinger LS. Evasion of
operon required for normal replication and trafficking in macro- complement-mediated lysis and complement C3 deposition are
phages. Infect Immun 2009;77:2517 29. regulated by Francisella tularensis lipopolysaccharide O antigen.
[93] Mohapatra NP, Soni S, Bell BL, Warren R, Ernst RK, Muszynski J Immunol 2008;181:5568 78.
A, et al. Identification of an orphan response regulator required [110] Cederlund A, Gudmundsson GH, Agerberth B. Antimicrobial
for the virulence of Francisella spp. and transcription of pathoge- peptides important in innate immunity. FEBS J 2011;278:
nicity island genes. Infect Immun 2007;75:3305 14. 3942 51.
[94] Sammons-Jackson WL, McClelland K, Manch-Citron JN, [111] Wang X, Ribeiro AA, Guan Z, Abraham SN, Raetz CR.
Metzger DW, Bakshi CS, Garcia E, et al. Generation and charac- Attenuated virulence of a Francisella mutant lacking the lipid A
terization of an attenuated mutant in a response regulator gene of 4’-phosphatase. Proc Natl Acad Sci U S A 2007;104:4136 41.
Francisella tularensis live vaccine strain (LVS). DNA Cell Biol [112] Bina XR, Lavine CL, Miller MA, Bina JE. The AcrAB RND
2008;27:387 403. efflux system from the live vaccine strain of Francisella tularen-
[95] Horzempa J, Carlson Jr. PE, O’Dee DM, Shanks RM, Nau GJ. sis is a multiple drug efflux system that is required for virulence
Global transcriptional response to mammalian temperature pro- in mice. FEMS Microbiol Lett 2008;279:226 33.
vides new insight into Francisella tularensis pathogenesis. BMC [113] Clemens DL, Lee BY, Horwitz MA. Francisella tularensis enters
Microbiol 2008;8:172. macrophages via a novel process involving pseudopod loops.
[96] Wehrly TD, Chong A, Virtaneva K, Sturdevant DE, Child R, Infect Immun 2005;73:5892 902.
Edwards JA, et al. Intracellular biology and virulence determi- [114] Geier H, Celli J. Phagocytic receptors dictate phagosomal escape
nants of Francisella tularensis revealed by transcriptional profil- and intracellular proliferation of Francisella tularensis. Infect
ing inside macrophages. Cell Microbiol 2009;11:1128 50. Immun 2011;79:2204 14.
[97] Waters LS, Storz G. Regulatory RNAs in bacteria. Cell [115] Santic M, Asare R, Skrobonja I, Jones S, Abu Kwaik Y.
2009;136:615 28. Acquisition of the vacuolar ATPase proton pump and phagosome
[98] Janovska S, Pavkova I, Hubalek M, Lenco J, Macela A, Stulik J. acidification are essential for escape of Francisella tularensis
Identification of immunoreactive antigens in membrane proteins into the macrophage cytosol. Infect Immun 2008;76:
enriched fraction from Francisella tularensis LVS. Immunol Lett 2671 7.
2007;108:151 9. [116] Chong A, Wehrly TD, Nair V, Fischer ER, Barker JR, Klose KE,
[99] Postic G, Dubail I, Frapy E, Dupuis M, Dieppedale J, Charbit A, et al. The early phagosomal stage of Francisella tularensis deter-
et al. Identification of a novel small RNA modulating mines optimal phagosomal escape and Francisella pathogenicity
Francisella tularensis pathogenicity. PLoS One 2012;7:e41999. island protein expression. Infect Immun 2008;76:5488 99.
[100] Postic G, Frapy E, Dupuis M, Dubail I, Livny J, Charbit A, et al. [117] Clemens DL, Lee BY, Horwitz MA. Francisella tularensis pha-
Identification of small RNAs in Francisella tularensis. BMC gosomal escape does not require acidification of the phagosome.
Genomics 2010;11:625. Infect Immun 2009;77:1757 73.
[101] Forslund AL, Kuoppa K, Svensson K, Salomonsson E, [118] Nauseef WM. Assembly of the phagocyte NADPH oxidase.
Johansson A, Bystrom M, et al. Direct repeat-mediated deletion Histochem Cell Biol 2004;122:277 91.
Chapter | 108 Francisella 2007

[119] Mohapatra NP, Soni S, Rajaram MV, Dang PM, Reilly TJ, [135] Abplanalp AL, Morris IR, Parida BK, Teale JM, Berton MT.
El-Benna J, et al. Francisella acid phosphatases inactivate TLR-dependent control of Francisella tularensis infection and
the NADPH oxidase in human phagocytes. J Immunol host inflammatory responses. PLoS One 2009;4:e7920.
2010;184:5141 50. [136] Thakran S, Li H, Lavine CL, Miller MA, Bina JE, Bina XR, et
[120] Mohapatra NP, Soni S, Rajaram MV, Strandberg KL, Gunn JS. al. Identification of Francisella tularensis lipoproteins that stim-
Type A Francisella tularensis acid phosphatases contribute to ulate the toll-like receptor (TLR) 2/TLR1 heterodimer. J Biol
pathogenesis. PLoS One 2013;8:e56834. Chem 2008;283:3751 60.
[121] Lindgren H, Shen H, Zingmark C, Golovliov I, Conlan W, [137] Cole LE, Shirey KA, Barry E, Santiago A, Rallabhandi P, Elkins
Sjostedt A. Resistance of Francisella tularensis strains against KL, et al. Toll-like receptor 2-mediated signaling requirements
reactive nitrogen and oxygen species with special reference to for Francisella tularensis live vaccine strain infection of murine
the role of KatG. Infect Immun 2007;75:1303 9. macrophages. Infect Immun 2007;75:4127 37.
[122] Bakshi CS, Malik M, Regan K, Melendez JA, Metzger DW, [138] Telepnev M, Golovliov I, Grundstrom T, Tarnvik A, Sjostedt
Pavlov VM, et al. Superoxide dismutase B gene (sodB)-deficient A. Francisella tularensis inhibits Toll-like receptor-mediated
mutants of Francisella tularensis demonstrate hypersensitivity activation of intracellular signalling and secretion of TNF-
to oxidative stress and attenuated virulence. J Bacteriol alpha and IL-1 from murine macrophages. Cell Microbiol
2006;188:6443 8. 2003;5:41 51.
[123] Melillo AA, Mahawar M, Sellati TJ, Malik M, Metzger DW, [139] Butchar JP, Cremer TJ, Clay CD, Gavrilin MA, Wewers MD,
Melendez JA, et al. Identification of Francisella tularensis live Marsh CB, et al. Microarray analysis of human monocytes
vaccine strain CuZn superoxide dismutase as critical for resis- infected with Francisella tularensis identifies new targets of host
tance to extracellularly generated reactive oxygen species. response subversion. PLoS One 2008;3:e2924.
J Bacteriol 2009;191:6447 56. [140] Sampson TR, Saroj SD, Llewellyn AC, Tzeng YL, Weiss DS. A
[124] Llewellyn AC, Jones CL, Napier BA, Bina JE, Weiss DS. CRISPR/Cas system mediates bacterial innate immune evasion
Macrophage replication screen identifies a novel Francisella and virulence. Nature 2013;497:254 7.
hydroperoxide resistance protein involved in virulence. PLoS [141] Kofoed EM, Vance RE. Innate immune recognition of bacterial
One 2011;6:e24201. ligands by NAIPs determines inflammasome specificity. Nature
[125] Golovliov I, Baranov V, Krocova Z, Kovarova H, Sjostedt A. An 2011;477:592 5.
attenuated strain of the facultative intracellular bacterium [142] Miao EA, Mao DP, Yudkovsky N, Bonneau R, Lorang CG,
Francisella tularensis can escape the phagosome of monocytic Warren SE, et al. Innate immune detection of the type III secre-
cells. Infect Immun 2003;71:5940 50. tion apparatus through the NLRC4 inflammasome. Proc Natl
[126] Barker JR, Chong A, Wehrly TD, Yu JJ, Rodriguez SA, Liu J, et al. Acad Sci U S A 2010;107:3076 80.
The Francisella tularensis pathogenicity island encodes a secretion [143] Jones JW, Kayagaki N, Broz P, Henry T, Newton K, O’Rourke
system that is required for phagosome escape and virulence. Mol K, et al. Absent in melanoma 2 is required for innate immune
Microbiol 2009;74:1459 70. recognition of Francisella tularensis. Proc Natl Acad Sci USA
[127] Nano FE, Schmerk C. The Francisella pathogenicity island. Ann 2010;107:9771 6.
N Y Acad Sci 2007;1105:122 37. [144] Fink SL, Cookson BT. Apoptosis, pyroptosis, and necrosis:
[128] Broms JE, Sjostedt A, Lavander M. The Role of the Francisella mechanistic description of dead and dying eukaryotic cells.
Tularensis pathogenicity island in type VI secretion, intracellular Infect Immun 2005;73:1907 16.
survival, and modulation of host cell signaling. Front Microbiol [145] Sjostedt A, Conlan JW, North RJ. Neutrophils are critical for
2010;1:136. host defense against primary infection with the facultative intra-
[129] Janeway Jr. CA, Medzhitov R. Innate immune recognition. Annu cellular bacterium Francisella tularensis in mice and participate
Rev Immunol 2002;20:197 216. in defense against reinfection. Infect Immun 1994;62:2779 83.
[130] Kawai T, Akira S. The role of pattern-recognition receptors in [146] Fernandes-Alnemri T, Yu JW, Juliana C, Solorzano L, Kang S,
innate immunity: update on Toll-like receptors. Nat Immunol Wu J, et al. The AIM2 inflammasome is critical for innate
2010;11:373 84. immunity to Francisella tularensis. Nat Immunol 2010;11:
[131] Asong J, Wolfert MA, Maiti KK, Miller D, Boons GJ. Binding 385 93.
and cellular activation studies reveal that toll-like receptor 2 can [147] Jones CL, Weiss DS. TLR2 signaling contributes to rapid inflam-
differentially recognize peptidoglycan from Gram-positive and masome activation during F. novicida infection. PLoS One
Gram-negative bacteria. J Biol Chem 2009;284:8643 53. 2011;6:e20609.
[132] Raetz CR, Guan Z, Ingram BO, Six DA, Song F, Wang X, et al. [148] Wickstrum JR, Bokhari SM, Fischer JL, Pinson DM, Yeh HW,
Discovery of new biosynthetic pathways: the lipid A story. J Horvat RT, et al. Francisella tularensis induces extensive
Lipid Res 2009;50(Suppl):S103 8. caspase-3 activation and apoptotic cell death in the tissues of
[133] Li H, Nookala S, Bina XR, Bina JE, Re F. Innate immune infected mice. Infect Immun 2009;77:4827 36.
response to Francisella tularensis is mediated by TLR2 and [149] Levine B, Mizushima N, Virgin HW. Autophagy in immunity
caspase-1 activation. J Leukoc Biol 2006;80:766 73. and inflammation. Nature 2011;469:323 35.
[134] Elkins KL, Rhinehart-Jones TR, Stibitz S, Conover JS, Klinman [150] Checroun C, Wehrly TD, Fischer ER, Hayes SF, Celli J.
DM. Bacterial DNA containing CpG motifs stimulates Autophagy-mediated reentry of Francisella tularensis into the
lymphocyte-dependent protection of mice against lethal infection endocytic compartment after cytoplasmic replication. Proc Natl
with intracellular bacteria. J Immunol 1999;162:2291 8. Acad Sci U S A 2006;103:14578 83.
2008 PART | 17 Animal and Ectoparasitic Source Infections

[151] Cremer TJ, Amer A, Tridandapani S, Butchar JP. Francisella [169] Taylor MW, Feng GS. Relationship between interferon-gamma,
tularensis regulates autophagy-related host cell signaling path- indoleamine 2,3-dioxygenase, and tryptophan catabolism.
ways. Autophagy 2009;5:125 8. FASEB J 1991;5:2516 22.
[152] Edwards JA, Rockx-Brouwer D, Nair V, Celli J. Restricted [170] Zegarra-Moran O, Folli C, Manzari B, Ravazzolo R, Varesio L,
cytosolic growth of Francisella tularensis subsp. tularensis Galietta LJ. Double mechanism for apical tryptophan depletion
by IFN-gamma activation of macrophages. Microbiology in polarized human bronchial epithelium. J Immunol
2010;156:327 39. 2004;173:542 9.
[153] Akimana C, Al-Khodor S, Abu Kwaik Y. Host factors required for [171] Chu P, Rodriguez AR, Arulanandam BP, Klose KE. Tryptophan
modulation of phagosome biogenesis and proliferation of prototrophy contributes to Francisella tularensis evasion of gamma
Francisella tularensis within the cytosol. PLoS One 2010;5:e11025. interferon-mediated host defense. Infect Immun 2011;79:
[154] Meibom KL, Charbit A. Francisella tularensis metabolism and 2356 61.
its relation to virulence. Front Microbiol 2010;1:140. [172] Peng K, Monack DM. Indoleamine 2,3-dioxygenase 1 is a lung-
[155] Bridges K, Seligman P. Disorders of iron metabolism. specific innate immune defense mechanism that inhibits growth
In: Handlin RI, Lux SE, Stossel TP, London JB, editors. Blood of Francisella tularensis tryptophan auxotrophs. Infect Immun
principles and practice of hematology. Lippincott Company; 2010;78:2723 33.
1995. p. 1433 72. [173] Rohmer L, Fong C, Abmayr S, Wasnick M, Larson Freeman TJ,
[156] Braun V. Iron uptake mechanisms and their regulation in patho- Radey M, et al. Comparison of Francisella tularensis genomes
genic bacteria. Int J Med Microbiol 2001;291:67 79. reveals evolutionary events associated with the emergence of
[157] Wandersman C, Stojiljkovic I. Bacterial heme sources: the role human pathogenic strains. Genome Biol 2007;8:R102.
of heme, hemoprotein receptors and hemophores. Curr Opin [174] De Pascalis R, Taylor BC, Elkins KL. Diverse myeloid and lym-
Microbiol 2000;3:215 20. phoid cell subpopulations produce gamma interferon during early
[158] Mulero V, Brock JH. Regulation of iron metabolism in murine innate immune responses to Francisella tularensis live vaccine
J774 macrophages: role of nitric oxide-dependent and -indepen- strain. Infect Immun 2008;76:4311 21.
dent pathways following activation with gamma interferon and [175] Leiby DA, Fortier AH, Crawford RM, Schreiber RD, Nacy CA.
lipopolysaccharide. Blood 1999;94:2383 9. In vivo modulation of the murine immune response to
[159] Sullivan JT, Jeffery EF, Shannon JD, Ramakrishnan G. Francisella tularensis LVS by administration of anticytokine
Characterization of the siderophore of Francisella tularensis and antibodies. Infect Immun 1992;60:84 9.
role of fslA in siderophore production. J Bacteriol 2006;188: [176] Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to
3785 95. interferon-gamma. Annu Rev Immunol 1997;15:749 95.
[160] Miethke M, Marahiel MA. Siderophore-based iron acquisition [177] Virgin HW, Levine B. Autophagy genes in immunity. Nat
and pathogen control. Microbiol Mol Biol Rev 2007;71:413 51. Immunol 2009;10:461 70.
[161] Kiss K, Liu W, Huntley JF, Norgard MV, Hansen EJ. [178] Elkins KL, Rhinehart-Jones TR, Culkin SJ, Yee D, Winegar RK.
Characterization of operon mutants of Francisella novicida Minimal requirements for murine resistance to infection with
U112. FEMS Microbiol Lett 2008;285:270 7. Francisella tularensis LVS. Infect Immun 1996;64:3288 93.
[162] Ramakrishnan G, Meeker A, Dragulev B. fslE is necessary for [179] Woolard MD, Wilson JE, Hensley LL, Jania LA, Kawula TH,
siderophore-mediated iron acquisition in Francisella tularensis Drake JR, et al. Francisella tularensis-infected macrophages
Schu S4. J Bacteriol 2008;190:5353 61. release prostaglandin E2 that blocks T cell proliferation and pro-
[163] Larsson P, Oyston PC, Chain P, Chu MC, Duffield M, Fuxelius motes a Th2-like response. J Immunol 2007;178:2065 74.
HH, et al. The complete genome sequence of Francisella tular- [180] Wilson JE, Katkere B, Drake JR. Francisella tularensis induces
ensis, the causative agent of tularemia. Nat Genet ubiquitin-dependent major histocompatibility complex class II deg-
2005;37:153 9. Epub 2005 Jan 09. radation in activated macrophages. Infect Immun 2009;77:4953 65.
[164] Lindgren H, Honn M, Golovlev I, Kadzhaev K, Conlan W, [181] Bosio CM, Dow SW. Francisella tularensis induces aberrant
Sjostedt A. The 58-kilodalton major virulence factor of activation of pulmonary dendritic cells. J Immunol 2005;175:
Francisella tularensis is required for efficient utilization of iron. 6792 801.
Infect Immun 2009;77:4429 36. [182] Elkins KL, Cooper A, Colombini SM, Cowley SC, Kieffer TL.
[165] Sen B, Meeker A, Ramakrishnan G. The fslE homolog, FTL_0439 In vivo clearance of an intracellular bacterium, Francisella tular-
(fupA/B), mediates siderophore-dependent iron uptake in ensis LVS, is dependent on the p40 subunit of interleukin-12
Francisella tularensis LVS. Infect Immun 2010;78:4276 85. (IL-12) but not on IL-12 p70. Infect Immun 2002;70:1936 48.
[166] Cartron ML, Maddocks S, Gillingham P, Craven CJ, Andrews SC. [183] O’Farrell AM, Liu Y, Moore KW, Mui AL. IL-10 inhibits mac-
Feo transport of ferrous iron into bacteria. Biometals rophage activation and proliferation by distinct signaling
2006;19:143 57. mechanisms: evidence for Stat3-dependent and -independent
[167] Su J, Yang J, Zhao D, Kawula TH, Banas JA, Zhang JR. pathways. EMBO J 1998;17:1006 18.
Genome-wide identification of Francisella tularensis virulence [184] Mahnke K, Schmitt E, Bonifaz L, Enk AH, Jonuleit H.
determinants. Infect Immun 2007;75:3089 101. Immature, but not inactive: the tolerogenic function of immature
[168] Weiss DS, Brotcke A, Henry T, Margolis JJ, Chan K, Monack dendritic cells. Immunol Cell Biol 2002;80:477 83.
DM. In vivo negative selection screen identifies genes required [185] Fontenot JD, Rudensky AY. A well adapted regulatory contriv-
for Francisella virulence. Proc Natl Acad Sci USA ance: regulatory T cell development and the forkhead family
2007;104:6037 42. transcription factor Foxp3. Nat Immunol 2005;6:331 7.
Chapter | 108 Francisella 2009

[186] Cyktor JC, Turner J. Interleukin-10 and immunity against pro- [194] Schricker RL, Eigelsbach HT, Mitten JQ, Hall WC. Pathogenesis
karyotic and eukaryotic intracellular pathogens. Infect Immun of tularemia in monkeys aerogenically exposed to Francisella
2011;79:2964 73. tularensis 425. Infect Immun 1972;5:734 44.
[187] Fortier AH, Slayter MV, Ziemba R, Meltzer MS, Nacy CA. Live [195] Lyons CR, Wu TH. Animal models of Francisella tularensis
vaccine strain of Francisella tularensis: infection and immunity infection. Ann N Y Acad Sci 2007;1105:238 65.
in mice. Infect Immun 1991;59:2922 8. [196] Vonkavaara M, Telepnev MV, Rydén P, Sjöstedt A, Stöven S.
[188] Conlan JW, Vinogradov E, Monteiro MA, Perry MB. Mice Drosophila melanogaster as a model for elucidating the
intradermally-inoculated with the intact lipopolysaccharide, but pathogenicity of Francisella tularensis. Cell Microbiol
not the lipid A or O-chain, from Francisella tularensis LVS rap- 2008;10:1327 38.
idly acquire varying degrees of enhanced resistance against sys- [197] Shapiro DS, Schwartz DR. Exposure of laboratory workers to
temic or aerogenic challenge with virulent strains of the Francisella tularensis despite a bioterrorism procedure. J Clin
pathogen. Microb Pathog 2003;34:39 45. Microbiol 2002;40:2278 81.
[189] Wu TH, Hutt JA, Garrison KA, Berliba LS, Zhou Y, Lyons CR. [198] Burke DS. Immunization against tularemia: analysis of the effec-
Intranasal vaccination induces protective immunity against intra- tiveness of live Francisella tularensis vaccine in prevention of
nasal infection with virulent Francisella tularensis biovar A. laboratory-acquired tularemia. J Infect Dis 1977;135:55 60.
Infect Immun 2005;73:2644 54. [199] Eigelsbach HT, Downs CM. Prophylactic effectiveness of live
[190] Shen H, Chen W, Conlan JW. Susceptibility of various mouse and killed tularemia vaccines. I. Production of vaccine and eval-
strains to systemically- or aerosol-initiated tularemia by virulent uation in the white mouse and guinea pig. J Immunol
type A Francisella tularensis before and after immunization with 1961;87:415 25.
the attenuated live vaccine strain of the pathogen. Vaccine [200] Eneslatt K, Rietz C, Ryden P, Stoven S, House RV, Wolfraim
2004;22:2116 21. LA, et al. Persistence of cell-mediated immunity three decades
[191] Kieffer TL, Cowley S, Nano FE, Elkins KL. Francisella novicida after vaccination with the live vaccine strain of Francisella tular-
LPS has greater immunobiological activity in mice than F. tular- ensis. Eur J Immunol 2011;41:974 80.
ensis LPS, and contributes to F. novicida murine pathogenesis. [201] Cowley SC, Elkins KL. Immunity to Francisella. Front
Microbes Infect 2003;5:397 403. Microbiol 2011;2:26.
[192] Eigelsbach HT, Tulis JJ, McGavran MH, White JD. Live tulare- [202] Ericsson M, Sandström G, Sjöstedt A, Tärnvik A. Persistence of
mia vaccine I. : Host-Parasite Relationship in Monkeys cell-mediated immunity and decline of humoral immunity to the
Vaccinated Intracutaneously or Aerogenically. J Bacteriol intracellular bacterium Francisella tularensis 25 years after natu-
1962;84:1020 7. ral infection. J Infect Dis 1994;170:110 4.
[193] White JD, McGavran MH, Prickett PA, Tulis JJ, Eigelsbach [203] Wayne Conlan J, Oyston PC. Vaccines against Francisella tular-
HT. Morphologic and immunohistochemical studies of the path- ensis. Ann N Y Acad Sci 2007;1105:325 50.
ogenesis of infection and antibody formation subsequent to vac- [204] Hornick RB, Eigelsbach HT. Aerogenic immunization of man
cination of Macaca irus With an Attenuated Strain of with live Tularemia vaccine. Bacteriol Rev 1966;30:532 8.
Pasteurella tularensis: II. Aerogenic Vaccination. Am J Pathol [205] Marohn ME, Barry EM. Live attenuated tularemia vaccines:
1962;41:405 13. Recent developments and future goals. Vaccine 2013;31:3485 91.

You might also like