Sei sulla pagina 1di 16

FEMS Microbiology Reviews 28 (2004) 43–58

www.fems-microbiology.org

Shigella flexneri infection: pathogenesis and vaccine development


Amy V. Jennison, Naresh K. Verma *

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


Faculty of Science, School of Biochemistry and Molecular Biology, The Australian National University, Canberra, ACT 0200, Australia
Received 1 May 2003; received in revised form 25 July 2003; accepted 30 July 2003

First published online 21 September 2003

Abstract

Shigella flexneri is a gram-negative bacterium which causes the most communicable of bacterial dysenteries, shigellosis. Shigellosis
causes 1.1 million deaths and over 164 million cases each year, with the majority of cases occurring in the children of developing
nations. The pathogenesis of S. flexneri is based on the bacteriaÕs ability to invade and replicate within the colonic epithelium, which
results in severe inflammation and epithelial destruction. The molecular mechanisms used by S. flexneri to cross the epithelial barrier,
evade the hostÕs immune response and enter epithelial cells have been studied extensively in both in vitro and in vivo models. Con-
sequently, numerous virulence factors essential to bacterial invasion, intercellular spread and the induction of inflammation have been
identified in S. flexneri. The inflammation produced by the host has been implicated in both the destruction of the colonic epithelium
and in controlling and containing the Shigella infection. The hostÕs humoral response to S. flexneri also appears to be important in
protecting the host, whilst the role of the cellular immune response remains unclear. The hostÕs immune response to shigellosis is
serotype-specific and protective against reinfection by the same serotype, making vaccination a possibility. Since the 1940s vaccines for
S. flexneri have been developed with little success, however, the growing understanding of S. flexneri’s pathogenesis and the hostÕs
immune response is assisting in the generation of more refined vaccine strategies. Current research encompasses a variety of vaccine
types, which despite disparity in their efficacy and safety in humans represent promising progress in S. flexneri vaccine development.
Ó 2003 Federation of European Microbiological Societies. Published by Elsevier B.V. All rights reserved.

Keywords: Shigella flexneri; Vaccine development; Pathogenesis

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2. Pathogenesis of S. flexneri . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2.1. Crossing the colonic epithelial layer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
2.2. Macrophage apoptosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
2.3. Adhesion to the basolateral membrane of colonic epithelial cells . . . . . . . . . . . . . . . 46
2.4. Uptake by the epithelial cell . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
2.5. Replication within the epithelial cell and intracellular and intercellular spread . . . . . 47
3. The hostÕs immune response to S. flexneri . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.1. Innate immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.2. Cellular immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.3. Humoral immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
4. S. flexneri vaccine development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
4.1. Subunit vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
4.2. Killed oral vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50

*
Corresponding author. Tel.: +61-2-6125-2666;
fax: +61-2-6125-0313.
E-mail address: naresh.verma@anu.edu.au (N.K. Verma).

0168-6445/$22.00 Ó 2003 Federation of European Microbiological Societies. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.femsre.2003.07.002
44 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

4.3. Non-invasive live vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50


4.4. Invasive live vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
4.5. Hybrid vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
4.6. Multiple-serotype protection strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
5. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53

1. Introduction Consequently, the World Health Organisation has pri-


oritised the development of a safe and effective vaccine

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


Members of the genus Shigella are gram-negative against S. flexneri [4].
facultative anaerobes that belong to the family, Enter- Numerous virulence genes have been identified in S.
obacteriaceae. They share common characteristics with flexneri, with the majority of these genes being located
members of the genus, Escherichia and the genetic re- on a 220 kb plasmid known as the virulence plasmid. At
latedness clearly suggests that they are a subtype of least three pathogenicity islands have also been located
E. coli [1,2]. The genus is divided into four species, on the S. flexneri chromosome, encoding important
Shigella flexneri, Shigella boydii, Shigella sonnei and virulence factors such as the lipopolysaccharide and
Shigella dysenteriae. These species are further divided genes for temperature-dependant regulation of the ex-
into serotypes based on biochemical differences and pression of virulence genes on the plasmid [7–10]. Two
variations in their O-antigen. Based on this classification S. flexneri 2a genome sequences have recently been re-
scheme, Shigella flexneri is divided into 13 serotypes. leased, consisting of a chromosome of approximately
Shigella species invade the colonic and rectal epithe- 4 600 000 and 221 618 bp virulence plasmid [1,2]. These
lium of primates and humans, causing the acute mucosal sequences have confirmed that S. flexneri contains a
inflammation characteristic of shigellosis. Infection is number of bacteriophage-related genes. Some of the
usually confined to the superficial layer of the colonic best-characterised S. flexneri phage genes are the sero-
mucosa, where severe tissue damage leads to abscesses type-conversion genes responsible for the serotype-spe-
and ulceration. Destruction of the epithelial layer leads cific modifications to the basic O-antigen structure [11].
to the clinical symptoms of watery diarrhoea, severe The roles of many S. flexneri virulence genes have been
abdominal pain and cramping, eventuating in the studied in a variety of cell culture experiments such as
bloody mucoid stool characteristic of bacillary dysen- invasion assays and plaque assays or through the use of in
tery. In the absence of effective treatments, shigellosis vivo animal models such as the Sereny test in the guinea
patients may develop secondary complications such pig, the mouse pulmonary model and rabbit ligated–
as septicaemia, pneumonia and haemolytic uremic intestinal–loop model [12–15]. Continued research into
syndrome [3]. S. flexneri virulence and pathogenesis will yield further
Shigellosis occurs in an estimated 164.7 million people understanding into the molecular basis of S. flexneri-
per year, of which 1.1 million cases result in death. 163.2 mediated invasion and destruction of the intestinal mu-
million annual cases occur in developing countries and cosa, as well as the role of the hostÕs subsequent innate,
69% of all patients are children under the age of five [4]. cellular and humoral immune responses. A comprehen-
S. flexneri is endemic in most developing countries sive understanding into S. flexneri’s disease-causing
and causes more mortality than any other Shigella spe- mechanisms will assist in vaccine development.
cies [5]. The predominant serotypes of S. flexneri in
developing countries are serotypes 1b, 2a, 3a, 4a and 6,
whilst in industrialised countries most isolates are 2a [4]. 2. Pathogenesis of S. flexneri
The high incidence of Shigella in developing countries is
generally attributed to the lack of clean water, poor S. flexneri is highly infectious, requiring as little as
sanitation, malnutrition and cost of antibiotic treat- 100 cells to cause disease in adult volunteers [16]. This
ment. Transmission is commonly via the faecal-oral low infective dose is in part attributed to S. flexneri’s
route, which is augmented by poor hygiene and close ability to survive the low acidity of the hostÕs stomach,
personal contact. via an up-regulation in acid resistance genes [17].
Antibiotics can be used to treat shigellosis, reducing Once Shigella reach the colon, they begin to invade
the period of bacterial excretion from the patient. the mucosa, penetrating, replicating within and spread-
However, S. flexneri is increasingly developing antibiotic ing between the mucosal epithelial cells. This behaviour
resistance [6]. This escalation in resistance to the com- and the subsequent inflammatory response of the host
monly used, cheaper antibiotics adds increased strain to destroy the colonic epithelial layer generating the clini-
the limited health services of developing countries. cal symptoms of shigellosis (Fig. 1) [18].
Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58
Fig. 1. Pathogenesis of S. flexneri. 1. Lumenal bacteria invade the colonic epithelial layer by three known mechanisms. S. flexneri can manipulate the tight junction proteins expressed by epithelial
cells, allowing paracellular movement of bacteria into the sub-mucosa. 2. PMN cells recruited by IL-8 and IL-1b produced in response to S. flexneri invasion create gaps between epithelial cells,
through which S. flexneri can transmigrate into the sub-mucosa. 3. Endocytic M cells transcytose bacteria, releasing them into an intraepithelial pocket filled with B and T lymphocytes and
macrophages. 4. Macrophages phagocytose the bacteria. S. flexneri escapes the phagosome and induces the macrophage to undergo apoptosis. The apoptotic macrophage releases IL-1b. 5. Sub-
mucosal S. flexneri contact the basolateral membrane of epithelial cells, activating secretion of proteins through their type-III secretion system. Proteins chaperoned in the cytosol of S. flexneri are
secreted into the epithelial cellÕs cytoplasm through a pore formed by IpaB and IpaC. IpaC polymerises actin, IpgD dissociates the plasma membrane from the actin cytoskeleton, VirA destabilises
microtubules and IpaA forms a complex with vinculin, depolymerising actin. This creates cell surface extensions which form around the bacterium, driving the epithelial cell to take up S. flexneri
into a vacuole. 6. IpaB and IpaC lyse the vacuole, releasing S. flexneri into the epithelial cellÕs cytoplasm. The S. flexneri protein, IcsA is displayed on only one pole of the bacterium, creating a
polymerised actin tail behind the bacterium. This propels S. flexneri through the cytoplasm until it contacts the plasma membrane, the force of the contact creates a protrusion into the
neighbouring epithelial cell. Both membranes are lysed by IpaB and IpaC, releasing S . flexneri into the neighbouring epithelial cell. 7. Intracellular S. flexneri induces the epithelial cell to release
IL-8. IL-8 and the IL-1b released from apopotic macrophages are chemotactic to PMN cells (represented by dotted arrows), attracting and inducing them to migrate through the epithelial layer to
the lumen. This epithelial disruption amplifies S. flexneri invasion of the epithelial layer.

45
46 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

2.1. Crossing the colonic epithelial layer Ironically, PMN-mediated interruption of the barrier
function of the epithelial layer promotes the local spread
Experiments using polarised cell lines have demon- of Shigella, whilst the same PMN cells appear to be
strated that the majority of S. flexneri epithelial cell responsible for restricting the infection to the submu-
invasion occurs through the basolateral pole of colonic cosa and preventing systemic dissemination [25].
epithelial cells [19]. The epithelial layer acts as a barrier Recent research has revealed that the S. flexneri is
to pathogens found in the gut lumen. S. flexneri is able capable of manipulating the tight-junction associated
to penetrate the epithelial lining through the follicular proteins of human intestinal epithelial cells, allowing
associated epithelium (FAE). This is the epithelial layer bacterial paracellular movement through a model in-
found above the mucosa-associated lymph nodes, which testinal barrier. These results suggest that shigellae are
contains highly endocytic M cells (Membranous epi- also capable of penetrating the colonic epithelium via an

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


thelial cells). M cell or PMN independant mechanism (Fig. 1) [27].
M cells sample and transport lumenal antigen across
the epithelial barrier, releasing it into an intraepithelial 2.2. Macrophage apoptosis
pocket formed by the basolateral membrane of the M cell.
This pocket is filled with lymphocytes and macrophages Once released into the intraepithelial pocket of the M
waiting to take up any delivered lumenal antigen and cell, bacteria are engulfed by resident macrophages,
initiate a mucosal immune response [20]. The transcytotic possibly through a bacterial driven macropinocytic
properties of M cells are exploited as a route for invasion event similar to Shigella entry of epithelial cells [28].
of the impermeable epithelial lining by a number of S. flexneri is able to evade the killing mechanisms of the
pathogens, including S. flexneri (Fig. 1) [15]. macrophage by IpaB-mediated lysis of the phagocytic
Shigella appear to enter M cells by the same mem- vacuole (Fig. 1). The membrane lysing properties of the
brane ‘‘ruffling’’ seen in epithelial cell invasion [21]. virulence plasmid IpaB invasin allows the bacteria to
Once internalised in an endocytic vacuole by the M cell, gain free access to the cytoplasm [29]. Once in the
shigellae are moved rapidly through the cell and released macrophage cytosol, secreted IpaB binds and activates
into the intraepithelial pocket. Once the FAE is crossed, caspase-1, a member of the pro-apoptotic cysteine pro-
Shigella can access the basolateral membrane of the teases [30]. Caspase-1 dependant apoptosis is not an
epithelial cells. immunologically silent cell death, as activated caspase-1
In the later stages of a Shigella infection, shigellae cleaves and activates the pro-inflammatory cytokines
exploit the hostÕs inflammatory response in order to IL-1b and IL-18 [31]. Macrophage apoptosis occurs
amplify bacterial penetration of the colonic epithelium. within four hours of in vivo Shigella infection, releasing
Macrophages infected by S. flexneri are induced to bacteria into the sub-mucosa [32].
undergo apoptosis, releasing large amounts of IL-1,
which is important in inducing inflammation and re- 2.3. Adhesion to the basolateral membrane of colonic
cruiting PMN cells to the site of infection. Additionally, epithelial cells
the invasion of epithelial cells by Shigella activates the
transcription and secretion of IL-8. IL-8 is chemotactic It remains unclear exactly why S. flexneri preferen-
for PMN cells and plays a significant role in recruiting tially invades epithelial cells thorough their basolateral
PMN cells to the infected subepithelial area, where they membrane. The apical membrane of the colonic epi-
transmigrate through the epithelial lining to reach lu- thelial cells is covered with glycolipids which form a
menal bacteria (Fig. 1) [22]. mucin layer. This layer may act as a physical barrier
The influx of PMN cells across the epithelial layer in preventing S. flexneri access to the apical membrane,
response to Shigella disrupts the integrity of the epi- interfering with the type-III secretion systems delivery of
thelium allowing lumenal bacteria to cross into the sub- the invasion plasmid antigens (Ipa), required for
mucosa in an M-cell independant mechanism [23]. This Shigella entry into epithelial cells [33].
PMN recruitment has been demonstrated to be crucial Additionally the basolateral membrane of epithelial
for the generation of the inflammation and tissue de- cells may display cellular components utilised by Shigella
struction typical of shigellosis in a number of studies. as cell adhesion receptors. The role of receptor-mediated
Experiments in the rabbit ligated–intestinal–loop model epithelial cell adhesion in Shigella infection is incom-
of Shigella infection where either IL-1 or IL-8 is inhib- pletely understood. However, it is known that the Shigella
ited almost abolished inflammation, tissue destruction Mxi/Spa secreton system requires contact with a host cell
and notably decreased the amount of bacterial invasion to trigger the secretion of the Ipa invasins [34]. A number
[24,25]. Additionally, the blocking of CD18, an adhesion of basolateral receptors capable of binding Shigella
molecule used by PMNs during migration, in the same components have been identified. The Ipa proteins are
animal model also diminished tissue damage and bac- capable of interaction with a5b1 integrin, a basolateral
terial invasion [26]. receptor which binds the extracellular matrix located
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 47

beneath the epithelium [35]. IpaB is also able to bind the tin filaments, organising an entry foci around the bac-
membrane receptor, CD44, which is the major cell surface terium [50,51]. IpgD is injected into the epithelial cell by
receptor for hyaluronic acid and is found on the baso- the S. flexneri type III secretion system, where it acts as
lateral membrane of epithelial cells [36,37]. Both a5b1 a phosphoinositide phosphatase, uncoupling the plasma
integrin and CD44 can act as cytoskeleton linkers, sug- membrane from the actin cytoskeleton, allowing mem-
gesting that upon the binding of Shigella they may con- brane extensions to form [52].
tribute to the cytoskeletal reorganisation seen during VirA has recently been identified as an additional
epithelial invasion [38]. effector molecule of S. flexneri epithelial cell invasion.
More recently, bacterial adherence to epithelial cells in An interaction between VirA and tubulin within the
a polarised model epithelium was shown to be dependant host cytosol destabilises microtubules around the bac-
on the length and presence of the O-antigen [39]. These terial site of entry. It is proposed that this destabilisation

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


results suggest that the S. flexneri LPS may play a role in could stimulate Rac1, a Rho family GTPase, creating
cell-bacteria interactions during epithelial cell invasion. A lamellipodial extensions in the host cell [53].
number of cell receptors capable of binding LPS have The cytoskeletal rearrangements induced by the
been characterised, including CD14 and the Toll family of Shigella effector proteins results in the bacterium being
receptors (TLRs), which are found on the basolateral internalised by the epithelial cell within a macropino-
membrane of epithelial cells [40,41]. cytic vacuole.

2.4. Uptake by the epithelial cell 2.5. Replication within the epithelial cell and intracellular
and intercellular spread
S. flexneri invades epithelial cells through a macr-
opinocytic process, where S. flexneri-induced rear- The macropinocytic vacuole containing the Shigella
rangements of the host cell cytoskeleton engulf the bacterium is rapidly lysed by the IpaB invasin, which
bacterium into a vacuole (Fig. 1). acts as membranolytic toxin in the phagosome mem-
The virulence plasmid of S. flexneri encodes two loci brane, releasing Shigella into the host cell cytoplasm
crucial to this invasive phenotype, the ipa locus and the [29]. The lysis of the phagosome may also involve IpaC,
mxi-spa locus. The ipa operon encodes the ‘‘invasion which is able to disrupt phospholipid membranes upon
plasmid antigens’’, IpaA, IpaB, IpaC and IpaD, which insertion of its hydrophobic regions [54,55].
are the effectors of bacterial entry into the host cell. The S. flexneri can replicate inside the cytoplasm of epi-
mxi-spa operon encodes the components of a type-III thelial cells in vitro with a doubling time of 40 minutes.
secretion system, which is a flagella-like structure used Epithelial cells are observed undergoing necrotic-like
to deliver proteins, such as the Ipa proteins, from the death during shigellosis (Fig. 1) [56]. Although it was
bacterial cytoplasm to the cytoplasmic membrane or initially proposed that Shigella multiplication within the
even cytosol of the host cell [42]. The mxi-spa operon cytosol was the cause of epithelial cell lysis, it seems
and IpaB, IpaC and IpaD are essential for in vitro epi- more likely that the cells are being destroyed by the
thelial cell invasion [43,44]. hostÕs inflammatory response [26]. In fact, Shigella
The detailed mechanisms by which the Ipa proteins would gain little advantage from killing the epithelial
generate Shigella invasion are not completely under- cell as whilst the bacteria are contained within the epi-
stood. The Ipa proteins are synthesised and stored thelial cell they are protected from immune cells and are
within the bacteria, where they are associated with in a favourable environment for replication [56].
chaperone proteins until secretion is activated by con- mxiE, a gene located within the mxi/spa locus has
tact with a host cell [45,46]. A complex formed by IpaB recently been identified as a transcriptional regulator for
and IpaD may play a role in the regulation of this se- a number of putative virulence factors required for vir-
cretion [34]. Once secretion is activated by contact with ulence in the Sereny test. mxiE is only activated when
an epithelial cell, the N-terminus of IpaC binds IpaB the bacterium is within the epithelial cell cytosol sug-
[47]. Both proteins are hydrophobic allowing this com- gesting that its role is to regulate virulence genes used in
plex to insert into the membrane of the host cell to form the post-invasion steps of infection [57].
a pore [48]. It is presumed that the other effector mole- Shigella is able to exploit the host cells actin assembly
cules, delivered by the type III secreton, are able to ac- machinery to move through the host cell cytoplasm and
cess the host cytoplasm through this pore. into adjacent epithelial cells. This intra and intercellular
The C-terminal domain of IpaC activates host cell spread is a crucial step in the virulence of Shigella and is
Rho GTPases, triggering actin polymerisation and fi- driven by the outer membrane protein, IcsA (VirG)
lopodial extensions in the vicinity of the bacteria [49]. [58–60]. IcsA is expressed in a unipolar fashion on the
IpaA is secreted into the cytosol of the host epithelial bacterial surface, with the greatest concentration local-
cell where it binds the cytoskeleton-associated protein ised to the old pole of the bacterium [61]. Newly syn-
vinculin. The IpaA–vinculin complex depolymerises ac- thesised IcsA appears to be directly targeted to the old
48 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

pole by two internal regions, where it is autotransported regulation of a variety of cytokines (IL-1, TNF-a, IL-6,
to the outer membrane [62,63]. The maintenance of IcsAÕs IFN-c, TNF-b, IL-4, IL-10, TGF-b and IL-8) [80]. Al-
unipolar localisation is essential for intracellular move- though some of the clinical symptoms of shigellosis may
ment and appears to be dependant on the structure of the actually be a direct consequence of the cytokines, they
LPS. Mutant S. flexneri strains missing, expressing partial also assist in controlling and containing the infection.
O-antigenÕs or lacking a modal distribution of O-antigen Resident macrophages and infiltrating monocytes are
chain length display non-polar surface localisation of unable to efficiently kill S. flexneri in their phagosomes
IcsA and are unable to spread from cell to cell [64–66]. It is and instead succumb to apoptosis [81,82]. The IL-18
possible that the LPS maintains IcsA polarity by forming released by apoptotic macrophages can target NK cells
interlocking microdomains with its O-antigen side chains and T lymphocytes, inducing production of IFN-c [83].
on the surface of the bacterium, which would prevent IcsA IFN-c deficient mice are five times more susceptible to a

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


from diffusing away from the old pole [67]. A recent study Shigella infection, as IFN-c activates macrophages and
has revealed the need for S. flexneri to display short length fibroblast cells, which promote bacterial clearance and
O-antigen chains in order to prevent the blocking of Ic- possibly inhibit bacterial replication within epithelial
sAÕs active sites by very long O-antigen chains. This cells [84].
finding suggests that S. flexneri has evolved to express two The most important consequence of the hostÕs innate
O-antigen chain lengths with each contributing to the immune response appears to be the cytokine induced
virulence of the strain; short chains which allow IcsA to migration of PMN cells. The transcription factor NF-j
function and long chains which confer resistance to serum B is activated in Shigella-infected epithelial cells in an
[68]. LPS-dependant mechanism, leading to the production
The Shigella protein DegP also appears to be re- and secretion of IL-8 by the infected cells [39]. IL-8 is a
quired for efficient intracellular spread and polarised potent chemoattractant for PMN cells, as is the IL-1
expression of IcsA. The exact role of DegP is unknown released from apoptotic macrophages.
but it may be important in the delivery of IcsA to the Shigella is unable to escape the phagocytic vacuole of
bacterial surface [69]. PMN cells and are killed inside the phagosome [85].
IcsA at the bacterial pole interacts with the host protein Recent research has implicated the human neutrophil
neural Wiskott–Aldrich syndrome protein (N-WASP) elastase (NE) as a key host defence protein of the neu-
and possibly with vinculin [70–72]. IcsA specifically binds trophil, capable of degrading Shigella virulence proteins
N-WASP and not other members of the WASP family, within 10 min of Shigella infecting the neutrophil [86].
which stimulates actin-related protein (Arp) 2/3 complex- PMN cells ultimately play a crucial role in controlling
mediated actin polymerisation [73,74]. This ligand speci- the Shigella infection, confining extracellular bacteria to
ficity of IcsA–N-WASP may determine which host cells the mucosa, preventing deeper tissue invasion and sys-
allow Shigella to use actin-based motility [74]. temic spread [25,87].
Actin polymerisation at the pole of the bacterium Another host defence mechanism directed against
creates propulsive force, which drives the bacterium Shigella has recently been discovered. The glycoprotein,
through the cytoplasm of the cell until it contacts the host lactoferrin, present in mucosal secretions, breast milk
cell membrane, forming a protrusion into the neigh- and phagocytic cells can impair the ability of S. flexneri
bouring epithelial cell [75]. The protrusion is actively en- to invade HeLa cells, exposing IpaB–IpaC complexes to
docytosed by the neighbouring cell in a myosin light chain protease degradation by disrupting the bacterial surface
kinase dependant mechanism, which also requires cad- [88]. Additionally, a study in transgenic mice expressing
herin expression [76,77]. The bacteria are then sur- a human intestinal defensin has demonstrated an im-
rounded by two cellular membranes, which are lysed by portant role for intestinally-secreted antibiotic peptides
secreted IpaB and IpaC [78]. Another protein, VacJ, has in controlling a Salmonella typhimurium enteric infection
also been shown to be essential for freeing Shigella into [89]. It is highly likely that intestinal defensins would
the cytoplasm of the next cell [79]. Thus, Shigella is able display similar antibiotic properties against enteric
to replicate and spread within the intestinal epithelial S. flexneri.
layer whilst avoiding exposure to the extracellular envi-
ronment and its circulating immune cells. 3.2. Cellular immunity

Very little data is available on the hostÕs cellular im-


3. The hosts immune response to S. flexneri mune response to S. flexneri, especially in comparison to
other intracellular bacteria. Studies have shown in-
3.1. Innate immunity creased T cell activation in shigellosis patients and T cell
clones have been isolated which proliferate in response
The severe inflammation generated by shigellosis can to S. flexneri antigen [90–93]. The cytokines induced by
persist in the gut for over a month, with a general up- Shigella antigens in vaccine studies are suggestive of Th1
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 49

and Th2 lymphocyte responses [94,95]. Additionally, the IgA, especially anti-LPS IgA have been detected in
increased susceptibility of AIDS patients, deficient in humans suffering natural shigellosis in a number of
CD4þ T cells, to shigellosis could suggest that cell- studies and is thought to play an important role in im-
mediated immunity can play a protective role in shig- munity to re-infection [105–109]. Anti-LPS secretory
ellosis [96]. IgA antibodies in the breast milk of mothers exposed to
However, the contribution of T lymphocytes to the shigellosis appear to be responsible for the decreased
hostÕs protective immunity to Shigella was studied in the severity of shigellosis in Shigella-infected infants [110].
mouse pulmonary model where mice deficient in T cells Additionally, the implantation of a serotype-specific
were vaccinated with attenuated S. flexneri. These mice sIgA hybridoma on the back of mice protected them
were suitably protected from challenge with wild type against intranasal challenge with a lethal dose of
bacteria despite their deficiency in T lymphocytes, sug- S. flexneri organisms [111]. This experiment suggests

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


gesting that even if T cell responses develop to Shigella that a mucosal antibody directed against a single LPS
they are not essential for protection [97]. epitope of Shigella could be sufficient for protective
immunity against re-infection by the homologous sero-
3.3. Humoral immunity type.
Despite shigellosis generally being a localised mucosal
Information about the hostÕs humoral response to infection, serum antibodies IgG and IgM are detected in
Shigella infection has been collected from numerous se- natural human infections directed against the LPS and
rological studies of infected humans and experiments virulence plasmid antigens [105–107,112,113]. IgG and
performed in animal models. The data suggests that the possibly IgM directed against the LPS have been shown
humoral immune response is a major component of to play a protective role in immunity to Shigella in mice
protective immunity to shigellosis with both systemic and studies. IgA deficient vaccinated mice are fully protected
mucosal responses activated against the LPS and some against pulmonary Shigella challenge, suggesting that
virulence plasmid encoded proteins, including the Ipa IgG or IgM are able to provide immunity [114]. Im-
proteins. The serotype-specific structure of the LPS is munised mice deficient in all T lymphocytes were pro-
assumed to be the major target of the hostÕs immune re- tected from wild-type Shigella challenge by a
sponse as natural and experimental infections with Shi- predominantly anti-LPS IgM response [97]. It is still
gella confer serotype-specific immunity, where previous unclear what role serum antibodies directed against the
infection or vaccination provides little to no protection LPS of S. flexneri are playing in the generation of se-
against heterologous serotypes [98–100]. However, anti- rotype-specific immunity, although they may be stimu-
bodies directed against epitopes shared between certain lating complement killing of the bacteria or mediating
O-antigen structures do appear to show some cross-re- antibody-dependant cellular cytotoxicity in the mucosal
activity [101]. The protective significance of these cross- area [115–117]. However, it must be stressed that the
reactive antibodies is incompletely understood and is protective role of serum antibodies in controlling
discussed in more detail in Section 4.6. However, the Shigella infection has been predominately characterised
overall importance of an antibody response to Shigella in mice and warrants further investigation as human
infection has been confirmed in a study which showed that vaccination data suggests that the parenteral stimula-
a reduced and delayed humoral immune response in tion of serum Ig does not correlate with protection
comparison to adult patients is the likely cause of the in- [118–121].
creased susceptibility of children to shigellosis [102].
It appears that both the systemic and mucosal arms
of the humoral response are activated as serum IgG, 4. S. flexneri vaccine development
IgM and secretory IgA have all been implicated in the
generation of serotype-specific immunity against The cost of treating shigellosis with antibiotics, es-
S. flexneri. pecially in the developing world, is unrealistic. The se-
Secretory IgA (sIgA) is made up of 2 IgA units and rotype-specific immunity generated by S. flexneri
two polypeptides, the J chain and the secretory com- provides protection against reinfection by the homolo-
ponent (SC). sIgA transcytoses into the lumenal cavity gous serotype, making vaccination a viable option for
of the intestine where the secretory component binds the controlling shigellosis.
mucosal coating of the epithelial cells, forming an an- A suitable vaccine for shigellosis must fulfil certain
tibody shield over the cells [103]. sIgA can also coat the requirements: the mucosal immune system must be
outer membrane of lumenal bacteria, impeding invasion activated and this immunity should be long-lasting,
by preventing their attachment to the mucosal surfaces, the vaccine must be cheap to manufacture, induce
mediate antibody-dependant cell-mediated cytotoxicity minimal side effects and be simple to administer, as
and interfere with bacterial utilisation of growth factors children in developing countries will be the main
[104]. recipients.
50 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

Since the 1940s a number of candidate vaccines for S. content in ribosomal preparations varies, making a
flexneri have been developed but as yet none have been consistent vaccine difficult to manufacture [129].
successful enough for field release. Early attempts to
develop S. flexneri vaccines consisted of inactivated 4.2. Killed oral vaccines
bacteria delivered parenterally, which failed to induce a
protective immune response, despite inducing a high Early challenge experiments in monkeys revealed that
titre of serum anti-LPS antibody [118–121]. The lack orally administered acetone-killed and dried Shigella
of protection was most likely due to the failure of was unable to protect monkeys from infection [130].
the parenteral vaccine in inducing a mucosal immune More recently however, an oral heat-killed S. flexneri
response. vaccine evaluated in a rabbit model was shown to be
Consequently, many recent vaccine strategies have

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


100% protective [131]. Thus, further studies are required
concentrated on developing live vaccine strains which to determine the protective capabilities of killed oral
can be administered orally and will activate the effectors vaccines for S. flexneri in humans.
of mucosal immunity.
4.3. Non-invasive live vaccines
4.1. Subunit vaccines
Mutations in either the S. flexneri chromosome or the
Subunit Shigella vaccines may avoid the safety issues virulence plasmid have been used to generate non-in-
associated with live vaccines. vasive live vaccine strains. Most of these strains were
LPS can be complexed to proteosomes and delivered safe in humans and were able to induce some degree of
intranasally to humans. Clinical trials have revealed that protective immunity in volunteers (Table 1). Probably
a S. flexneri 2a LPS–proteosome vaccine is capable of the most successful of these vaccines is the invasion
generating a serotype-specific immune response in hu- plasmid mutant, S. flexneri 2a Istrati T32 which is 100%
mans [122]. S. flexneri LPS has also been attached to safe in humans and provides up to an 85% protective
proteins and delivered parenterally to volunteers as efficacy. However, it must be administered in large
potential vaccines. These vaccines were safe in humans (1  1011 CFU) multiple doses every six months which is
and induced strong serum antibody responses [123–126]. expensive and difficult to implement in developing
Other subunit vaccines are yet to be evaluated in countries [132].
humans. Mice and guinea pigs were protected from
S. flexneri challenge by mucosal immunisation with a 4.4. Invasive live vaccines
purified complex of IpaB, IpaC, IpaD and LPS [127].
Ribosomal preparations from Shigella delivered paren- Invasive oral Shigella vaccine strain strategies are
terally can generate protective immunity in guinea pigs increasingly being explored as invasive strains deliver
and monkeys [128]. The immune response is directed antigen to the mucosal immune system, provoking a
against O-antigen polysaccharides (L -hapten) purified strong immune response. As the genetic understanding
with the ribosomal preparation. However, the O-antigen of S. flexneri virulence has improved so have the strat-

Table 1
Live non-invasive oral S. flexneri vaccines which have been assessed in monkeys or humans
Vaccine Description Safety Efficacy Comment References
S. flexneri 2a 2457O Spontaneous Reverts to virulence Monkeys are Reactogenic in [130,146,147]
avirulent mutant, virF in humans protected. Caused humans
inactivated by dysentery in 34% of
insertion human volunteers
S. flexneri streptomy- Spontaneous Reversion to Up to 90% protection Unstable [98,148,149]
cin dependant strains mutants incapable of streptomycin in field trials with phenotype and
growing in the independence in multiple doses. US inconsistent
absence of volunteers. Diarrhoea trials found marginal protective efficacy
streptomycin and vomiting in protection
15–35% of volunteers
(5  1010 CFU)
S. flexneri 2a Istrati Spontaneous deletion Safe in humans, mild Around 80% Protection lasts 6 [132,150]
T32 of ipaBCDA, invA adverse effects in very protection in humans months. Reduces
and icsA (virG) from few volunteers at when administered in attack rate of
the virulence plasmid 2  1011 CFU 5 doses heterologous
Shigella serotypes
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 51

egies to construct safe invasive vaccines. Invasive vac- 4.6. Multiple-serotype protection strategies
cine strains are generally attenuated by mutations in
either virulence genes necessary for pathogenesis after Because immunity to S. flexneri is serotype-specific,
cell entry or in metabolic genes which prevent the bac- vaccination against one serotype will only provide pro-
teria from replicating and spreading in the host after tection to infection by the homologous serotype. The
invasion. serotypes of S. flexneri differ in their distribution with up
Mutations in either icsA and/or in a variety of met- to four different serotypes prevalent in an endemic area.
abolic genes have produced attenuated invasive vaccine Thus, the ideal S. flexneri vaccine would provide pro-
strains which are safe and capable of up to 100% pro- tection to all prevalent serotypes of a particular geo-
tection with multiple doses in monkeys (Table 2). A graphical region.
number of auxotrophic vaccine strains, some also car- All S. flexneri serotypes, with the exception of sero-

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


rying mutations in virulence genes, have been assessed type 6, share a common O-antigen backbone. The ad-
for their safety and ability to induce a serotype-specific dition of glucosyl and/or O-acetyl groups to the sugars
immune response in human volunteers in phase 1 clini- of the backbone generates the type (I, II, IV, V and X)
cal trials [94,133–135]. These strains vary in the levels of and group (3, 4, 6 and 7, 8) antigens that define the
their attenuation and their immunogenicity (Table 2). serotypes. Consequently, some serotypes share type and/
The S. flexneri 2a vaccine strain, SC602 has pro- or group antigens on their LPS [11]. Because the im-
ceeded to phase 2 clinical trials in humans. This strain mune response is primarily directed against the LPS,
carries deletions in icsA as well as the aerobactin iuc some antibodies generated against the group or type
locus, which is involved in iron transport. SC602 is safe antigen of one serotype should be cross-reactive to other
in humans at low doses (1  104 CFU) and capable of serotypes. For example, antibody in human sera raised
providing protection to immunised humans challenged against S. flexneri 2a has been shown to cross-react with
with wild type 2a S. flexneri. However, the vaccine is LPS from heterologous serotypes 1a, 2b, 5a and Y,
only weakly attenuated causing symptoms such as di- which share type or group antigens with the serotype 2a
arrhoea and fever when administered in doses higher O-antigen structure [101]. As cross-reactivity of the hu-
that 1  104 CFU [136]. Thus, despite promising results man sera to all of the different serotypes was not ob-
with S. flexneri invasive vaccine candidates, further served, it appears that the common group 1 antigen,
work is required to achieve a balance between immu- which is shared by all S. flexneri serotypes [143] was not
nogenicity and safety in humans. able to induce any sufficiently cross-reactive antibodies.
This suggests that the group 1 antigen is poorly immu-
4.5. Hybrid vaccines nogenic and may not have a role in inducing protective
antibodies against heterologous serotypes. It also re-
E. coli vaccine candidates have also been used to mains unclear whether the cross-reactive antibodies di-
develop hybrid vaccines expressing Shigella antigens. rected against the other group and type antigens of the
Early attempts using Shigella–E. coli hybrid vaccines LPS, as mentioned above, can provide any protection
developed invasive vaccines which caused symptoms against infection by heterologous serotypes. Therefore,
in human volunteers or which were not protective further research is required to adequately establish the
[137,138]. Strains based on E. coli K12 carrying the role of the cross-reactive O-antigen epitopes in human
group- and type-specific antigen of S. flexneri 2a and the immunity against shigellosis.
virulence plasmid from S. flexneri 5 were unable to in- However, animal studies have shown that mixing a
duce significant protection in immunised volunteers number of S. flexneri vaccines of different serotypes into
[139,140]. a vaccine cocktail can invoke an immune response with
Additionally, S. flexneri candidate vaccine strains are cross-reactive potential. A vaccine cocktail containing
being engineered to express the O-antigenÕs of other serotype 2a and 3a S. flexneri strains was administered
Shigella species. The S. flexneri 2a vaccine strain T32 to guinea pigs in the Sereny test, conferring significant
carrying a plasmid containing the gene cluster coding protection to challenge by serotypes 1b, 2b, 5b and Y
for S. sonnei O-antigen, was capable of providing 100% [144]. Thus, by combining a selection of S. flexneri ser-
protection to mice against challenge with both virulent otypes into a vaccine cocktail, it may be possible to
S. flexneri and S. sonnei [141]. The S. dysenteriae O- cross-protect against most S. flexneri serotypes.
antigen biosynthesis genes were integrated into the Alternatively, single S. flexneri vaccine strains can be
SFL124 (serotype Y) vaccine strain, generating strains engineered to express the O-antigen of more than one
able to induce antibodies specific to both homologous serotype. Such strains should be capable of generating a
and heterologous O-antigen structures in mice [142]. protective immune response in the host directed against
Similar approaches are also being used to generate each of the serotype specific O-antigen structures. This
vaccines protective against multiple S. flexneri serotypes lab has previously reported the serotype-conversion of
and will be discussed below. the serotype Y S. flexneri candidate vaccine strain,
52

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


Table 2
Live invasive oral S. flexneri vaccines which have been assessed in monkeys or humans
Vaccine Description Safety Efficacy Comment References
S. flexneri 5a SC5700 Attenuated by insertions in icsA Mucoid diarrhoea in some About 70% protection in Slightly reactogenic in monkeys [151]
and iuc monkeys with 5  1010 CFU monkeys after 3 doses
S. flexneri 5a SC560 Deletion in icsA Mucoid diarrhoea and mild 100% protective in monkeys Slightly reactogenic in monkeys [60]

A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58


clinical symptoms in some after 3 doses
monkeys with 5  1010 CFU
S. flexneri 5a SC433 Deletion in envZ and ompR Mucoid diarrhoea and mild 100% protective in monkeys Slightly reactogenic in monkeys [60]
clinical symptoms in some after 3 doses
monkeys with 5  1010 CFU
S. flexneri 5a SC445 Deletion in envZ, icsA and 100% safe in monkeys at One out of five monkeys Level of attenuation may be too severe [60]
ompR 5  1010 CFU challenged became sick for consistent protection
S. flexneri 2a vc77 Auxotrophic for purine (Pur ) No symptoms in adults or Dysentery in 2 out of 4 adults Poor protective capabilities [152–154]
and rifampicin resistant children up to 3  109 CFU challenged
S. flexneri Y TSF-21 Thymine requiring (Thy ) and No symptoms in monkeys at 100% protection in monkeys Temperature sensitivity [155,156]
temperature sensitive (Ts ) 1  1011 CFU after 2 doses mutation has a high rate of reversion
S. flexneri Y SFL114 Insertional inactivation of aroD No symptoms in monkeys at 100% protection in monkeys Some reversion of phenotype observed [157]
2–3  1010 CFU after four doses in laboratory
conditions
S. flexneri Y SFL124 Deletion of aroD Very mild symptoms in some 100% protection in monkeys Strain is advantageous as it can be [135,158–160]
human volunteers at 2  109 after three doses of 1  1011 converted to new serotypes
CFU and children at up to CFU
1  109 CFU
S. flexneri 2aSFL1070 Deletion of aroD Mild symptoms in volunteers at About 85% protection in Further attenuation may be necessary [133,161]
1  107 – 1  108 CFU. Increased monkeys after 4 doses 1  1011
clinical symptoms at 1  109 CFU
CFU
S. flexneri 2a Deletions in icsA and aroA No symptoms in humans when Challenge only performed in Strain may require further [134,162,163]
CVD1203 administered in a single dose of guinea pigs, provided 83% attenuation in order to reduce
1  106 CFU protection reactogenicity in humans
S. flexneri Deletions in icsA, sen, set and No symptoms in humans when Challenge only performed in Immunogenicity in humans may not be [94,144]
2aCVD1207 guaAB administered in a single dose of guinea pigs, provided 85% sufficient with just one dose
up to 1  108 CFU protection
S. flexneri 2a SC602 Deletions in icsA gene and the Mild diarrhoea and fever in 3 out of 7 volunteers challenged Provides protection or reduces symp- [136,164]
iuc locus. humans at 1  104 CFU experienced mild diarrhoea toms to infection with S. flexneri 2a.
Further attenuation may be required
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 53

SFL124 to serotype X by the insertion of the bacterio- Chen, R., Ma, D., Qiang, B., Wen, Y., Hou, Y. and Yu, J. (2002)
phage SfX serotype-conversion gene cluster [145]. This Genome sequence of Shigella flexneri 2a: insights into pathoge-
nicity through comparison with genomes of Escherichia coli K12
approach has been utilised to insert the serotype con- and O157. Nucleic Acids Res. 30, 4432–4441.
version gene cluster of bacteriophage SfV and the glu- [3] Bennish, M.L. (1991) Potentially lethal complications of shigel-
cosyl transferase gene of bacteriophage SfII in tandem losis. Rev. Infect. Dis. 13, S319–S324.
into the SFL124 chromosome. The resulting strain dis- [4] Kotloff, K.L., Winickoff, J.P., Ivanoff, B., Clemens, J.D.,
played the 3,4 group antigen and both the II and V type Swedlow, D.L., Sansonetti, P.J., Adak, G.K. and Levine,
M.M.M. (1999) Global burden of Shigella infections: implica-
antigens as detected by monovalent antiserum and tions for vaccine development and implementation of control
simultaneously induced a serotype-specific immune strategies. Bull. World Health Organ. 77, 651.
response to both serotypes 2a and 5a in the mouse [5] Bennish, M.L. and Wojtyniak, B.J. (1991) Mortality due to
pulmonary model (unpublished data, this lab). This shigellosis: community and hospital data. Rev. Infect. Dis. 13,

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


strategy could be easily applied to any newly developed S245–S251.
[6] Ashkenazi, S., Levy, I., Kazaronovski, V. and Samra, Z. (2003)
serotype Y vaccine candidates, ultimately generating a Growing antimicrobial resistance of Shigella isolates. J. Anti-
variety of S. flexneri polyvalent vaccine strains which microb. Chemother. 51, 427–429.
could be combined into vaccine cocktails designed for [7] Moss, J.E., Cardozo, T.J., Zychlinsky, A. and Groisman, E.A.
specific geographical areas. (1999) The SelC-associated SHI-2 pathogenicity island of
Shigella flexneri. Mol. Microbiol. 33, 74–83.
[8] Rajakumar, K., Sasakawa, C. and Adler, B. (1997) Use of a
novel approach, termed island probing, identifies the Shigella
5. Conclusions flexneri she pathogenicity island which encodes a homolog of the
immunoglobulin A protease-like family of proteins. Infect.
Through in vitro and in vivo studies we are beginning Immun. 65, 4606–4614.
[9] Turner, S.A., Luck, S.N., Sakellaris, H., Rajakumar, K. and
to develop a detailed picture of how S. flexneri invades
Adler, B. (2001) Nested deletions of the SRL pathogenicity
the intestinal mucosa and causes disease. Further re- island of Shigella flexneri 2a. J. Bacteriol. 183, 5535–5543.
search into the hostÕs immune response will ultimately [10] Walker, J.C. and Verma, N.K. (2002) Identification of a putative
reveal all inflammatory mediators involved in shigellosis pathogenicity island in Shigella flexneri using subtractive hy-
and clarify whether cellular immunity plays an impor- bridisation of the S. flexneri and Escherichia coli genomes.
FEMS Microbiol. Lett. 10580, 1–8.
tant role in the control of Shigella and in protection
[11] Allison, G.E. and Verma, N.K. (2000) Serotype-converting
against reinfection. Virulence factors are crucial to bacteriophages and O-antigen modification in Shigella flexneri.
S. flexneri for the development and maintenance of Trends Microbiol. 8, 17–23.
disease. Undoubtedly, many more remain to be identi- [12] Mallet, C.P., VanDeVerg, L.L., Collins, H.H. and Hale, T.L.
fied and characterised. Discovery of new virulence fac- (1993) Evaluation of Shigella vaccine safety and efficacy in an
intranasally challenged mouse model. Vaccine 11, 190–196.
tors and an understanding of gene regulation and
[13] Oaks, E.V., Wingfield, M.E. and Formal, S.B. (1985) Plaque
conditional gene expression will be greatly assisted by formation by virulent Shigella flexneri. Infect. Immun. 48, 124–
the release of the S. flexneri 2a genome sequence. A 129.
number of the Shigella vaccine strategies mentioned [14] Sereny, B. (1957) Experimental keratoconjunctivitis Shigellosa.
above have shown promise in animal studies and initial Acta Microbiol. Acad. Sci. Hung., 4.
[15] Wassef, J.S., Keren, D.F. and Mailloux, J.L. (1989) Role of M
human clinical trials, significantly advancing the status
cells in initial antigen uptake and in ulcer formation in the rabbit
of Shigella vaccine development. However, to com- intestinal loop model of shigellosis. Infect. Immun. 57, 858–863.
pletely protect against natural Shigella infections, the [16] DuPont, H.L., Levine, M.M., Hornick, R.B. and Formal, S.B.
development of vaccine cocktails and polyvalent vac- (1989) Inoculum size in shigellosis and implications for expected
cines must be addressed. Future vaccine research should mode of transmission. J. Infect. Dis. 159, 1126–1127.
[17] Small, P., Blankenhorn, D., Welty, D., Zinser, E. and Slonczewski,
encompass trials of mixed vaccines designed to confer
J.L. (1994) Acid and base resistance in Escherichia coli and Shigella
protection against multiple serotypes. flexneri: role of rpoS and growth pH. J. Bacteriol. 176, 1729–1737.
[18] Philpott, D.J., Edgeworth, J.D. and Sansonetti, P.J. (2000) The
pathogenesis of Shigella flexneri infection: lessons from in vitro
References and in vivo studies. Philos. Trans. R. Soc. Lond. B. Biol. Sci.
355, 575–586.
[1] Wei, J., Goldberg, M.B., Burland, V., Venkatesan, M.M., Deng, [19] Mounier, J., Vasselon, T., Hellio, R., Lesourd, M. and Sanso-
W., Fournier, G., Mayhew, G., Plunkett III, G., Rose, D., netti, P.J. (1992) Shigella flexneri enters the human colonic Caco-
Darling, A., Mau, B., Perna, N.T., Payne, S.M., Runyen- 2 epithelial cells through the basolateral pole. Infect. Immun. 60,
Janecky, L., Zhou, S., Schwartz, D.C. and Blattner, F.R. (2003) 237–248.
Complete genomic sequence and comparative genomics of [20] Neutra, M.R., Pringault, E. and Kraehenbuhl, J.P. (1996)
Shigella flexneri serotype 2a strain 2457T. Infect. Immun. 71, Antigen sampling across epithelial barriers and induction of
2775–2786. mucosal immune responses. Annu. Rev. Immunol. 14.
[2] Jin, Q., Yuan, Z., Xu, J., Wang, Y., Shen, Y., Lu, W., Wang, J., [21] Sansonetti, P.J. and Phalipon, A. (1999) M cells as ports of entry
Liu, H., Yang, J., Yang, F., Zhang, X., Zhang, J., Yang, G., Wu, for enteroinvasive pathogens: mechanisms of interaction,
H., Qu, D., Dong, J., Sun, L., Xue, Y., Zhao, A., Gao, Y., Zhu, consequences for the disease process. Semin. Immunol. 11,
J., Kan, B., Ding, K., Chen, S., Cheng, H., Yao, Z., He, B., 193–203.
54 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

[22] Beatty, W.L. and Sansonetti, P.J. (1997) Role of lipopolysac- CD11/CD18 and CD14 share a common lipid A signalling
charide in signaling to subepithelial polymorphonuclear leuko- pathway. J. Immunol. 161, 5413–5420.
cytes. Infect. Immun. 65, 4395–4404. [42] Hueck, C.J. (1998) Type III protein secretion systems in bacterial
[23] Perdomo, J.J., Gounon, P. and Sansonetti, P.J. (1994) Polymor- pathogens of animals and plants. Microbiol. Mol. Biol. Rev. 62,
phonuclear leukocyte transmigration promotes invasion of colonic 379–433.
epithelial monolayer by Shigella flexneri. J. Clin. Invest. 93, 633– [43] Menard, R., Sansonetti, P.J. and Parsot, C. (1993) Non-polar
643. mutagenesis of the ipa genes defines IpaB, IpaC and IpaD as
[24] Sansonetti, P.J., Arondel, J., Cavaillon, J.M. and Huerre, M. effectors of Shigella entry into epithelial cells. J. Bacteriol. 175,
(1995) Role of interleukin-1 in the pathogenesis of experimental 5899–5906.
shigellosis. J. Clin. Invest. 96, 884–892. [44] Sasakawa, C., Kamata, K., Sakai, T., Makino, S., Yamada, M.,
[25] Sansonetti, P.J., Arondel, J., Huerre, M., Harada, A. and Okada, N. and Yoshikawa, M. (1988) Virulence-associated
Matsushima, K. (1999) Interleukin-8 controls bacterial transepi- genetic regions comprising 31 kilobases of the 230-kilobase
thelial translocation at the cost of epithelial destruction in plasmid in Shigella flexneri 2a. J. Bacteriol. 170, 2480–2484.

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


experimental shigellosis. Infect. Immun. 67, 1471–1480. [45] Menard, R., Sansonetti, P.J., Parsot, C. and Vasselon, T. (1994)
[26] Perdomo, O.J., Cavaillon, J.M., Huerre, M., Ohayon, H., Extracellular association and cytoplasmic partitioning of the
Gounon, P. and Sansonetti, P.J. (1994) Acute inflammation IpaB and IpaC invasins of Shigella flexneri. Cell 79, 515–525.
causes epithelial invasion and mucosal destruction in experimen- [46] Page, A.L., Sansonetti, P.J. and Parsot, C. (2002) Spa15 of
tal shigellosis. J. Exp. Med. 180, 1307–1319. Shigella flexneri, a third type of chaperone in the type III
[27] Sakaguchi, T., Kohler, H., Gu, X., McCormick, B.A. and secretion pathway. Mol. Microbiol. 43, 1533–1542.
Reinecker, H. (2002) Shigella flexneri regulates the tight [47] Harrington, A.T., Hearn, P.D., Picking, W.L., Barker, J.R.,
junction-associated proteins in human intestinal epithelial cells. Wessel, A. and Picking, W.D. (2003) Structural characterization
Cell. Microbiol. 4, 367–381. of the N terminus of IpaC from Shigella flexneri. Infect. Immun.
[28] Kuwae, A., Yoshida, S., Tamano, K., Mimuro, H., Suzuki, T. 71, 1255–1264.
and Sasakawa, C. (2001) Shigella invasion of macrophage [48] Blocker, A., Gounon, P., Larquet, E., Niebuhr, K., Cabiaux, V.,
requires the insertion of IpaC into the host plasma membrane. Parsot, C. and Sansonetti, P.J. (1999) The tripartite type III
J. Biol. Chem. 276, 32230–32239. secretion of Shigella flexneri inserts IpaB and IpaC into hosts
[29] High, N., Mounier, J., Prevost, M.C. and Sansonetti, P.J. (1992) membranes. J. Cell Biol. 147, 683–693.
IpaB of Shigella flexneri causes entry into epithelial cells and [49] Tran Van Nhieu, G., Caron, E., Hall, A. and Sansonetti, P.J.
escape from the phagocytic vacuole. EMBO J. 11, 1991–1999. (1999) IpaC induces actin polymerization and filopodia forma-
[30] Chen, R., Smith, M.R., Thirumalai, K. and Zychlinsky, A. tion during Shigella entry into epithelial cells. EMBO J. 18,
(1996) A bacterial invasin induces macrophage apoptosis by 3249–3262.
binding directly to ICE. EMBO J. 15, 3853–3860. [50] Bourdet-Sicard, R., Rudiger, M., Jockusch, B.M., Gounon, P.,
[31] Dinarello, C.A. (1998) Interleukin-1b, Interleukin-18 and the Sansonetti, P.J. and Tran Van Nhieu, G. (1999) Binding of the
Interleukin-1b converting enzyme. Ann. N.Y. Acad. Sci. 856, 1– Shigella protein IpaA to vinculin induces F-actin depolymeriza-
11. tion. EMBO J. 18, 5853–5862.
[32] Zychlinsky, A., Thirumalai, K., Arondel, J., Cantey, J.R., [51] Tran Van Nhieu, G., Ben-ZeÕev, A. and Sansonetti, P.J. (1997)
Aliprantis, A.O. and Sansonetti, P.J. (1996) In vivo apoptosis Modulation of bacterial entry into epithelial cells by association
in Shigella flexneri infections. Infect. Immun. 64, 5357–5365. between vinculin and the Shigella IpaA invasin. EMBO J. 16,
[33] Nutten, A., Sansonetti, P.J., Huet, G., Bourdon-Bisiaux, C., 2717–2729.
Meresse, B., Colombel, J. and Desreumaux, P. (2002) Epithelial [52] Niebuhr, K., Giuriato, S., Pedron, T., Philpott, D.J., Gaits, F.,
inflammation response induced by Shigella flexneri depends on Sable, J., Sheetz, M.P., Parsot, C., Sansonetti, P.J. and Payrastre,
mucin gene expression. Microbes Infect. 4, 1121–1124. B. (2002) Conversion of PtdIns(4,5)P2 into PtdIns(5)P by the S.
[34] Menard, R., Sansonetti, P.J. and Parsot, C. (1994) The secretion flexneri effector IpgD reorganizes host cell morphology. EMBO
of the Shigella flexneri Ipa invasions is activated by epithelial J. 21, 5069–5078.
cells and controlled by IpaB and IpaD. EMBO J. 13, 5293–5302. [53] Yoshida, S., Katayama, E., Kuwae, A., Mimuro, H., Suzuki, T. and
[35] Watarai, M., Funato, S. and Sasakawa, C. (1996) Interaction of Sasakawa, C. (2002) Shigella deliver an effector protein to trigger host
Ipa proteins of Shigella flexneri with alpha5beta1 integrin microtubule destabilization, which promotes Rac1 activity and
promotes entry of the bacteria into mammalian cells. J. Exp. efficient bacterial internalization. EMBO J. 21, 2923–2935.
Med. 183, 991–999. [54] De Geyter, C., Vogt, B., Benjelloun-Touimi, Z., Sansonetti, P.J.,
[36] Alho, A.M. and Underhill, C.B. (1989) The hyaluronate receptor Ruysschaert, J.M. and Cabiaux, V. (1997) Purification of IpaC, a
is preferentially expressed on proliferating epithelial cells. J. Cell protein involved in entry of Shigella flexneri into epithelial cells
Biol. 108, 1557–1565. and characterization of its interaction with lipid membranes.
[37] Skoudy, A., Mounier, J., Aruffo, A., Ohayon, H., Gounon, P., FEBS Lett. 400, 149–154.
Sansonetti, P.J. and Tran Van Nhieu, G. (2000) CD44 binds to [55] Kueltzo, L.A., Osiecki, J., Barker, J., Picking, W.L., Ersoy, B.,
the Shigella IpaB protein and participates in bacterial invasion of Picking, W.D. and Middaugh, C.R. (2003) Structure–function
epithelial cells. Cell. Microbiol. 2, 19–33. analysis of Invasion Plasmid Antigen C (IpaC) from Shigella
[38] Tran Van Nhieu, G. and Sansonetti, P.J. (1999) Mechanism of flexneri. J. Biol. Chem. 278, 2792–2798.
Shigella entry into epithelial cells. Curr. Opin. Microbiol. 2, 51–55. [56] Mantis, N., Prevost, M.C. and Sansonetti, P.J. (1996) Analysis of
[39] Kohler, H., Rodrigues, S.P. and McCormick, B.A. (2002) S. epithelial cell stress response during infection by Shigella flexneri.
flexneri interactions with the basolateral membrane domain of Infect. Immun. 64, 2474–2482.
polarised model intestinal epithelium: role of LPS in cell invasion [57] Kane, C.D., Schuch, R., Day, W.A. and Maurelli, A.T. (2002)
and in activation of the mitogen activated protein kinase ERK. MxiE regulates intracellular expression of factors secreted by the
Infect. Immun. 70, 1150–1158. Shigella flexneri 2a type III secretion system. Infect. Immun. 184,
[40] Aderem, A. and Ulevitch, R.J. (2000) Toll-like receptors in the 4409–4419.
induction of the Innate Immune Response. Science 406, 782–787. [58] Bernardini, M.L., Mounier, J., dÕHauteville, H., Coquis-Ron-
[41] Ingalls, R.R., Monks, B.G., Savedra, R., Christ, W.J., Delude, don, M. and Sansonetti, P.J. (1989) Identification of icsA, a
R.L., Medvedev, A.E., Espevik, T. and Golenbock, D.T. (1998) plasmid locus of Shigella flexneri that governs bacterial intra-
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 55

and intercellular spread through interaction with F-actin. Proc. [76] Rathman, M., de Lanerolle, P., Ohayon, H., Gounon, P. and
Natl. Acad. Sci. USA. 86, 3867–3871. Sansonetti, P.J. (2000) Myosin light chain kinase plays an
[59] Lett, M., Sasakawa, C., Okada, N., Sakai, T., Makino, S., essential role in S. flexneri dissemination. J. Cell Sci. 113, 3375–
Yamada, M., Komatsu, K. and Yoshikawa, M. (1989) irG, a 3386.
plasmid-coded virulence gene of Shigella flexneri: identification [77] Sansonetti, P.J., Mounier, J., Prevost, M.C. and Mege, R.M.
of the virG protein and determination of the complete coding (1994) Cadherin expression is required for the spread of Shigella
sequence. J. Bacteriol. 171, 353–359. flexneri between epithelial cells. Cell 76, 829–839.
[60] Sansonetti, P.J., Arondel, J., Fontaine, A., dÕHauteville, H. and [78] Page, A.L., Ohayon, H., Sansonetti, P.J. and Parsot, C. (1999)
Bernardini, M.L. (1991) OmpB (osmo-regulation) and IcsA (cell– The secreted IpaB and IpaC invasins and their cytoplasmic
cell spread) mutants of Shigella flexneri; vaccine candidates and chaperone IpgC are required for intercellular dissemination of
probes to study the pathogenesis of shigellosis. Vaccine 9, 416– Shigella flexneri. Cell. Microbiol. 1, 183–193.
422. [79] Suzuki, T., Murai, T., Fukuda, T., Tobe, T., Yoshikawa, M. and
[61] Goldberg, M.B., Barzu, O., Parsot, C. and Sansonetti, P.J. Sasakawa, C. (1994) Identification and characterization of a

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


(1993) Unipolar localization and ATPase activity of IcsA, a chromosomal virulence gene, vacJ, required for intercellular
Shigella flexneri protein involved in intracellular movement. J. spreading of Shigella flexneri. Mol. Microbiol. 11, 31–41.
Bacteriol. 175, 2189–2196. [80] Raqib, R., Lindberg, A.A., Wretlind, B., Bardhan, P.K.,
[62] Steinhauer, J., Agha, R., Pham, T., Varga, A.W. and Goldberg, Andersson, U. and Andersson, J. (1995) Persistence of local
M.B. (1999) The unipolar Shigella surface protein IcsA is cytokine production in shigellosis in acute and convalescent
targeted directly to the bacterial old pole: IcsP cleavage of IcsA stages. Infect. Immun. 63, 289–296.
occurs over the entire bacterial surface. Mol. Microbiol. 32, 367– [81] Hathaway, L.J., Griffin, G.E., Sansonetti, P.J. and Edgeworth,
377. J.D. (2002) Human monocytes kill Shigella flexneri but then die
[63] Charles, M., Perez, M., Kobil, J.H. and Goldberg, M.B. (2001) by apoptosis associated with suppression of proinflammatory
Polar targeting of Shigella virulence factor IcsA in Enterobac- cytokine production. Infect. Immun. 70, 3833–3842.
teriacae and Vibrio. Proc. Natl. Acad. Sci. USA 98, 9871–9876. [82] Zychlinsky, A., Prevost, M.C. and Sansonetti, P.J. (1992)
[64] Sandlin, R.C., Goldberg, M.B. and Maurelli, A.T. (1996) Effect Shigella flexneri induces apoptosis in infected macrophages.
of O side-chain length and composition on the virulence of Nature 358, 167–169.
Shigella flexneri 2a. Mol. Microbiol. 22, 63–73. [83] Biet, F., Locht, C. and Kremer, L. (2002) Immunoregulatory
[65] Sandlin, R.C., Lampel, K.A., Keasler, S.P., Goldberg, M.B., functions of interleukin 18 and its role in defense against
Stolzer, A.L. and Maurelli, A.T. (1995) Avirulence of rough bacterial pathogens. J. Mol. Med. 80, 147–162.
mutants of Shigella flexneri: requirement of O antigen for correct [84] Way, S.S., Borczuk, A.C., Dominitz, R. and Goldberg, M.B.
unipolar localisation of IcsA in the bacterial outer membrane. (1998) An essential role for gamma interferon in innate
Infect. Immun. 63, 229–237. resistance to Shigella flexneri infection. Infect. Immun. 66,
[66] Van Den Bosch, L., Manning, P.A. and Morona, R. (1997) 1342–1348.
Regulation of O-antigen chain length is required for Shigella [85] Mandic-Mulec, I., Weiss, J. and Zychlinsky, A. (1997) Shigella
flexneri virulence. Mol. Microbiol. 23, 765–775. flexneri is trapped in polymorphonuclear leukocyte vacuoles and
[67] Robbins, J.R., Monack, D.M., McCallum, S.J., Vegas, A., efficiently killed. Infect. Immun. 65, 110–115.
Pham, E., Goldberg, M.B. and Theriot, J.A. (2001) The making [86] Weinrauch, Y., Drujan, D., Shapiro, S.D., Weiss, J. and
of a gradient: IcsA (VirG) polarity in Shigella flexneri. Mol. Zychlinsky, A. (2002) Neutrophil elastase targets virulence
Microbiol. 41, 861–872. factors of enterobacteria. Nature 417, 91–94.
[68] Morona, R., Daniels, C. and Van Den Bosch, L. (2003) Genetic [87] Zhang, J., Jin, L., Champion, G., Seydel, K.B. and Stanley, S.L.
modulation of Shigella flexneri 2a lipopolysaccharide O antigen (2001) Shigella infection in a SCID mouse–human intestinal
modal chain length reveals that it has been optimized for xenograft model: role for neutrophils in containing bacterial
virulence. Microbiology 149, 925–939. dissemination in human intestine. Infect. Immun. 69, 3240–3247.
[69] Purdy, G.E., Hong, M. and Panyne, S. (2002) Shigella flexneri [88] Gomez, H.F., Ochoa, T.J., Carlin, L.G. and Cleary, T.G. (2003)
DegP facilitates IcsA surface expression and is required for Human lactoferrin impairs virulence of Shigella flexneri. J.
efficient intercellular spread. Infect. Immun. 70, 6355–6364. Infect. Dis. 187, 87–95.
[70] Suzuki, T., Miki, H., Takenawa, T. and Sasakawa, C. (1998) [89] Salzman, N.H., Ghosh, D., Huttner, K.M., Paterson, Y. and
Neural Wiskott–Aldrich syndrome protein is implicated in the Bevins, C.L. (2003) Protection against enteric salmonellosis in
actin based motility of Shigella flexneri. EMBO J. 17, 2767–2776. transgenic mice expressing a human intestinal defensin. Nature
[71] Goldberg, M.B. (1997) Shigella actin-based motility in the 422, 522–526.
absence of vinculin. Cell Motil. Cytoskeleton. 37, 44–53. [90] Islam, D. and Christensson, B. (2000) Disease dependant changes
[72] Suzuki, T., Saga, S. and Sasakawa, C. (1996) Functional analysis in T-cell populations in patients with shigellosis. APMIS 108, 251–
of Shigella VirG domains essential for interaction with vinculin 260.
and actin-based motility. J. Biol. Chem. 271. [91] Islam, D., Bardhan, P.K., Lindberg, A.A. and Christensson, B.
[73] Egile, C., Loisel, T.P., Laurent, V., Li, R., Pantaloni, D., (1995) Shigella infection Induces cellular activation of T and B
Sansonetti, P.J. and Carlier, M.F. (1999) Activation of the cells and distinct species-related changes in peripheral blood
CDC42 effector N-WASP by the Shigella flexneri IcsA protein lymphocyte subsets during the course of the disease. Infect.
promotes actin nucleation by Arp2/3 complex and bacterial Immun. 63, 2941–2949.
actin-based motility. J. Cell Biol. 146, 1319–1332. [92] Islam, D., Wretlind, B., Lindberg, A.A. and Christensson, B.
[74] Suzuki, T., Mimuro, H., Suetsugu, S., Miki, H., Takenawa, T. (1996) Changes in the peripheral blood T-cell receptor V b
and Sasakawa, C. (2002) Neural Wiskott–Aldrich syndrome repertoire in vivo and in vitro during shigellosis. Infect. Immun.
protein (N-WASP) is the specific ligand for Shigella VirG among 64, 1391–1399.
the WASP family and determines the host cell type allowing [93] Zwillich, S.H., Duby, A.D. and Lipsky, P.E. (1989) T-lymphocyte
actin-based spreading. Cell. Microbiol. 4, 223–233. clones responsive to Shigella flexneri. J. Clin. Microbiol. 27, 417–421.
[75] Monack, D.M. and Theriot, J.A. (2001) Actin-based motility is [94] Kotloff, K.L., Noriega, F.R., Samandari, T., Sztein, M.B.,
sufficient for bacterial membrane protrusion formation and host Losonsky, G.A., Nataro, J.P., Picking, W.D., Barry, E.M. and
cell uptake. Cell. Microbiol. 2, 633–647. Levine, M.M. (2000) Shigella flexneri 2a strain CVD 1207, with
56 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

specific deletions in virG, sen, set and guaBA, is highly attenuated [111] Phalipon, A., Kaufman, M., Michetti, P., Cavaillon, J.M.,
in humans. Infect. Immun. 68, 1034–1039. Huerre, M., Sansonetti, P.J. and Kraehenbuhl, J.P. (1995)
[95] van de Verg, L.L., Mallet, C.P., Collins, H.H., Larsen, T., Monoclonal immunoglobulin A antibody directed against sero-
Hammack, C. and Hale, T.L. (1995) Antibody and cytokine type-specific epitope of Shigella flexneri lipopolysaccharide
responses in a mouse pulmonary model of Shigella flexneri protects against murine experimental shigellosis. J. Exp. Med.
serotype 2a infection. Infect. Immun. 63, 1947–1954. 182, 769–778.
[96] Nelson, M.R., Shanson, D.C., Hawkins, D.A. and Gazzard, [112] Oaks, E.V., Hale, T.L. and Formal, S.B. (1986) Serum immune
B.G. (1992) Salmonella, Campylobacter and Shigella in HIV- response to Shigella protein antigens in Rhesus monkeys and
seropositive patients. AIDS 6, 1495–1498. humans infected with Shigella species. Infect. Immun. 53, 57–
[97] Way, S.S., Borczuk, A.C. and Goldberg, M.B. (1999) Thymic 63.
independance of adaptive immunity to the intracellular pathogen [113] van de Verg, L.L., Herrington, D.A., Boslego, J., Lindberg, A.A.
Shigella flexneri serotype 2a. Infect. Immun. 67, 3970–3979. and Levine, M.M. (1992) Age-specific prevalence of serum
[98] DuPont, H.L., Hornick, R.B., Synder, M.J., Libonati, J.P., antibodies to the invasion plasmid and lipopolysaccharide

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


Formal, S.B. and Gangarosa, E.J. (1972) Immunity in shigellosis. antigens of Shigella species in Chilean and North American
II. Protection induced by oral live vaccine or primary infection. populations. J. Infect. Dis. 166, 158–161.
J. Infect. Dis. 125, 12–16. [114] Way, S.S., Borczuk, A.C. and Goldberg, M.B. (1999) Adaptive
[99] Formal, S.B., Oaks, E.V., Olsen, R.E., Wingfield, M.E., Snoy, immune response to Shigella flexneri 2a cydC in immunocom-
P.J. and Cogan, J.P. (1991) Effect of prior infection with virulent petent mice and mice lacking immunoglobulin A. Infect. Immun.
Shigella flexneri 2a on the resistance of monkeys to subsequent 67, 2001–2004.
infection with Shigella sonnei. J. Infect. Dis. 164, 533–537. [115] Cohen, D., Green, M.S., Block, C., Slepon, R. and Ofek, I.
[100] Mel, D.M., Bogoljub, L.A., Nikolic, B.D. and Radovanic, M.L. (1991) Prospective study of the association between serum
(1968) Studies on vaccination against bacillary dysentery. 4. Oral antibodies to lipopolysaccharide O-antigen and the attack rate
immunization with live monotypic and combined vaccines. Bull. of shigellosis. J. Clin. Microbiol. 29, 386–389.
World Health Organ. 39, 375–380. [116] Lowell, G.H., MacDermott, R.P., Summers, P.L., Reeder, A.A.,
[101] Van De Verg, L.L., Bendiuk, N.O., Kotloff, K., Marsh, M.M., Bertovich, M.J. and Formal, S.B. (1980) Antibody-dependant
Ruckert, J.L., Puryear, J.L., Taylor, D.N. and Hartman, A.B. cell-mediated antibacterial activity: K lymphocytes, monocytes
(1996) Cross-reactivity of Shigella flexneri serotype 2a O antigen and granulocytes are effective against Shigella. J. Immunol. 125.
antibodies following immunization or infection. Vaccine 14, [117] Reed, W.P. and Albright, E.L. (1974) Serum factors responsible
1062–1068. for killing of Shigella. Immunology 26, 205–215.
[102] Raqib, R., Qadri, F., Sarker, P., Mia, S.M., Sansonetti, P.J., [118] Barnes, L.A., Cooper, M.L., Jerome, E.A., Durant, R.C. and
Albert, M.J. and Andersson, J. (2002) Delayed and reduced Smith, A.B. (1951) Field trial of Shigella flexneri III vaccine. VI.
adaptive humoral immune responses in children with shigellosis Mouse protective studies. J. Immunol. 66, 515–525.
compared with in adults. Scand. J. Immunol. 55, 414–423. [119] Bennett, I.L., Gordon, R.S. and Barnes, L.A. (1949) A field trial
[103] Phalipon, A., Cardona, A., Kraehenbuhl, J.P., Edelman, L., of Shigella flexneri III vaccine. II. Serum agglutination studies.
Sansonetti, P.J. and Corthesy, B. (2002) Secretory component: a J. Infect. Dis. 85, 180–194.
new role in secretory IgA-mediated immune exclusion in vivo. [120] Cooper, M.L., Tepper, J. and Keller, H.M. (1948) Studies in
Immunity 17, 107–115. dysentery vaccination. IV. Primary vaccination of children with
[104] Iijima, H., Takahashi, I. and Kiyono, H. (2001) Mucosal monovalent vaccines of Shigella. J. Immunol. 60, 189–203.
immune network in the gut for the control of infectious diseases. [121] Higgins, A., Floyd, T. and Kader, M. (1955) Studies in
Rev. Med. Virol. 11, 117–133. shigellosis. III. A controlled evaluation of monovalent Shigella
[105] Cohen, D., Block, C., Green, M.S., Lowell, G.H. and Ofek, I. vaccine in a highly endemic environment. Am. J. Trop. Med.
(1989) Immunoglobulin M, A and G antibody response to Hyg. 4, 281–288.
lipopolysaccharide O antigen in symptomatic and asymptomatic [122] Fries, L.F., Montemarano, A.D., Mallet, C.P., Taylor, D.N.,
Shigella infections. J. Clin. Microbiol. 27, 162–167. Hale, T.L. and Lowell, G.H. (2001) Safety and immunogenicity
[106] Islam, D., Veress, B., Bardhan, P.K., Lindberg, A.A. and of a proteosome-Shigella flexneri 2a lipopolysaccharide vaccine
Christensson, B. (1997) Quantitative assessment of IgG and IgA administered intranasally to healthy adults. Infect. Immun. 69,
subclass producing cells in rectal mucosa during shigellosis. J. 4545–4553.
Clin. Pathol. 50, 513–520. [123] Ashkenazi, S., Passwell, J.H., Harlev, E., Miron, D., Dagan, R.,
[107] Oberhelman, R.A., Kopecko, D.J., Salazar-Lindo, E., Gotuzzo, Farzan, N., Ramon, R., Majadly, F., Bryla, D.A., Karpas, A.B.,
E., Buysse, J.M., Venkatesan, M.M., Fernandez-Prada, C., Robbins, J.R. and Schneerson, R. (1999) Safety and immuno-
Guzman, M. and Leon-Barua, R. (1991) Prospective study of genicity of Shigella sonnei and Shigella flexneri 2a O-specific
systemic and mucosal immune responses in dysenteric patients to polysaccharide conjugates in children. J. Infect. Dis. 179, 1565–
specific Shigella invasion plasmid antigens and lipopolysaccha- 1568.
rides. Infect. Immun. 59, 2341–2350. [124] Cohen, D., Ashkenazi, S., Green, M.S., Lerman, Y., Slepon, R.,
[108] Rasolofo-Razanamparany, V., Cassel-Beraud, A.M., Roux, J., Robin, G., Orr, N., Taylor, D.N., Sadoff, J.C., Chu, C.,
Sansonetti, P.J. and Phalipon, A. (2001) Predominance of Shiloach, J., Schneerson, R. and Robbins, J.R. (1996) Safety
serotype-specific mucosal antibody response in Shigella flexneri and immunogenicity of investigational Shigella conjugate vac-
-infected humans living in an area of endemicity. Infect. Immun. cines in Israeli volunteers. Infect. Immun. 64, 4074–4077.
69, 5230–5234. [125] Passwell, J.H., Harlev, E., Ashkenazi, S., Chu, C., Miron, D.,
[109] Schultsz, C., Qadri, F., Hossain, S.A., Ahmed, F. and Ciznar, I. Ramon, R., Farzan, N., Shiloach, J., Bryla, D.A., Majadly, F.,
(1992) Shigella-specific IgA in saliva of children with bacillary Roberson, R., Robbins, J.R. and Schneerson, R. (2001) Safety
dysentery. FEMS Microbiol. Immunol. 4, 65–72. and immunogenicity of improved Shigella O-specific polysac-
[110] Clemens, J.D., Stanton, B., Stoll, B., Shahid, N.S., Banu, H. and charide-protein conjugate vaccines in adults in Israel. Infect.
Chowdhury, A.K. (1986) Breastfeeding as a determinant of Immun. 69, 1351–1357.
severity in shigellosis: evidence for protection throughout the [126] Taylor, D.N., Trofa, A.C., Sadoff, J.C., Chu, C., Bryla, D.A.,
first three years of life in Bangladesh children. Am. J. Epidemiol. Shiloach, J., Cohen, D., Ashkenazi, S., Lerman, Y. and Egan, W.
123, 710–720. (1993) Synthesis, characterization and clinical evaluation of
A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58 57

conjugate vaccines composed of the O-specific polysaccharides of candidate strain against Shigella flexneri 2a and Shigella sonnei.
Shigella dysenteriae type 1, Shigella flexneri type 2a and Shigella Chin. J. Biotechnol. 12, 89–97.
sonnei (Plesiomonas shigelloides) bound to bacterial toxoids. [142] Klee, S.R., Tzscgaschel, B.D., Singh, M., Falt, I., Lindberg,
Infect. Immun. 61, 3678–3687. A.A., Timmis, K.N. and Guzman, C.A. (1997) Construction and
[127] Turbyfill, K.R., Hartman, A.B. and Oaks, E.V. (2000) Isolation characterization of genetically-marked bivalent anti-Shigella
and characterization of a Shigella flexneri invasin complex dysenteriae and anti-Shigella flexneri Y live vaccine candidates.
subunit vaccine. Infect. Immun. 68, 6624–6632. Microb. Pathogenesis 22, 363–376.
[128] Levenson, V.I., Chernokhvostova, E.V., Lyubinskaya, M.M., [143] Carlin, N.I.A. and Lindberg, A.A. (1987) Monoclonal antibodies
Salamatova, S.A., Dzhikidze, E. and Stasilevitch, Z.K. (1988) specific for Shigella flexneri lipopolysaccharides: clones binding
Parental immunization with Shigella ribosomal vaccine elicits to type IV, V and VI antigens, group 3,4 antigen and an epitope
local IgA response and primes for mucosal memory. Int. Arch. common to all Shigella flexneri and Shigella dysenteriae type 1
Allergy Immunol. 87, 25–31. strains. Infect. Immun. 55, 1412–1420.
[129] Levenson, V.I. and Egorova, T.P. (1990) Polysaccharide nature [144] Noriega, F.R., Liao, F.M., Maneval, D.R., Ren, S., Formal, S.B.

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


of O-antigen in protective ribosomal preparations from Shigella: and Levine, M.M. (1999) Strategy for cross-protection among
experimental evidence and implications from the ribosomal Shigella flexneri serotypes. Infect. Immun. 67, 782–788.
vaccine concept. Res. Microbiol. 141, 707–720. [145] Guan, S. and Verma, N.K. (1998) Serotype conversion of a
[130] Formal, S.B., LaBrec, E., Palmer, A. and Falkow, S. (1965) Shigella flexneri candidate vaccine strain via a novel site specific
Protection of monkeys against experimental shigellosis with chromosome-integration system. FEMS Microbiol. Lett. 166, 79–
attenuated vaccines. J. Bacteriol. 90, 63–68. 87.
[131] Chakrabarti, M.K., Bhattacharya, J., Bhattacharya, M.K., Nair, [146] DuPont, H.L., Hornick, R.B., Snyder, M., Libonati, J.P.,
G.B., Bhattacharya, S.K. and Mahalanabis, D. (1999) Killed Formal, S.B. and Gangarosa, E.J. (1972) Immunity in shigellosis.
oral Shigella vaccine made from Shigella flexneri 2a protects I. Response of man to attenuated strains of Shigella. J. Infect.
against challenge in the rabbit model of shigellosis. Acta Dis. 125, 5–11.
Paediatr. 88, 161–165. [147] Mills, J.A., Venkatesan, M.M., Baron, L.S. and Buysse, J.M.
[132] Meitert, T., Pencu, E., Ciudin, L. and Tonciu, M. (1984) Vaccine (1992) Spontaneous insertion of an IS1-like element into the virF
strain Sh. flexneri T32 -ISTRATI. Studies in animals and volun- gene is responsible for avirulence in opaque colonial variants of
teers. Antidysentery immunoprophylaxis and immunotherapy by Shigella flexneri 2a. Infect. Immun. 60, 175–182.
live vaccine VADIZEN (Sh. flexneri T32 -ISTRATI). Arch. [148] Levine, M.M., Gangarosa, E.J., Werner, M. and Morris, G.K.
Roum. Pathol. Exp. Microbiol. 43, 251–278. (1974) Shigellosis in custodial institutions. III. Prospective
[133] Karnell, A., Li, A., Zhao, C.R., Karlsson, K., Nguyen, B.M. and clinical and bacteriologic surveillance of children vaccinated
Lindberg, A.A. (1995) Safety and immunogenicity study of the with oral attenuated Shigella vaccines. J. Pediatr. 84, 803–806.
auxotrophic Shigella flexneri 2a vaccine SFL1070 with a deleted [149] Mel, D.M., Terzin, A.L. and Vuksic, L. (1965) Studies on
aroD gene in adult Swedish volunteers. Vaccine 13, 88–99. vaccination against bacillary dysentery. 3. Effective oral immu-
[134] Kotloff, K.L., Noriega, F.R., Losonsky, G.A., Sztein, M.B., nization against Shigella flexneri 2a in a field trial. Bull. World
Wasserman, S.S., Nataro, J.P. and Levine, M.M. (1996) Safety, Health Organ. 32, 647–655.
immunogenicity, and transmissibility in humans of CVD1203, a [150] Venkatesan, M.M., Fernandez-Prada, C., Buysse, J.M., Formal,
live oral Shigella flexneri 2a vaccine candidate attenuated by S.B. and Hale, T.L. (1991) Virulence phenotype and genetic
deletions in aroA and virG. Infect. Immun. 64, 4542–4548. characteristics of the T32 -ISTRATI Shigella flexneri 2a vaccine
[135] Li, A., Pal, T., Forsum, U. and Lindberg, A.A. (1992) Safety and strain. Vaccine 9, 358–363.
immunogenicity of the live oral auxotrophic Shigella flexneri [151] Sansonetti, P.J. and Arondel, J. (1989) Construction and
SFL124 in volunteers. Vaccine 10, 395–404. evaluation of a double mutant of S. flexneri as a candidate for
[136] Coster, T.S., Hoge, C.W., van de Verg, L.L., Hartman, A.B., oral vaccination against shigellosis. Vaccine 7, 443–450.
Oaks, E.V., Venkatesan, M.M., Cohen, D., Robin, G., Fontaine- [152] Dentchev, V., Marinova, S., Vassilev, T., Bratoyeva, M. and
Thompson, A., Sansonetti, P.J. and Hale, T.L. (1999) Vaccina- Linde, K. (1990) Live Shigella flexneri 2a and Shigella sonnei I
tion against shigellosis with attenuated Shigella flexneri 2a strain vaccine candidate strains with two attenuating markers. II.
SC602. Infect. Immun. 67, 3437–3443. Preliminary results of vaccination of adult volunteers and
[137] Falkow, S., Schneider, H., Baron, L.S. and Formal, S.B. (1963) children aged 2–17 years. Vaccine 8, 30–34.
Virulence of Escherichia–Shigella genetic hybrids for the guinea [153] Linde, K., Dentchev, V., Bondarenko, V., Marinova, S.,
pig. J. Bacteriol. 86, 1251–1258. Randhagen, B., Bratoyeva, M., Tsvetanov, Y. and Romanova,
[138] Levine, M.M., Woodward, D., Formal, S.B., Gemski, P., DuPont, Y. (1990) Live Shigella flexneri and Shigella sonnei I vaccine
H.L., Hornick, R.B. and Snyder, M. (1977) Studies with a new candidate strains with two attenuating markers. I. Construction
generation of oral attenuated Shigella vaccine: Escherichia coli bearing of vaccine candidate strains with retained invasiveness but
surface antigens of Shigella flexneri. J. Infect. Dis. 136, 577–582. reduced intracellular multiplication. Vaccine 8, 25–29.
[139] Kotloff, K.L., Herrington, D.A., Hale, T.L., Newland, J.W., van [154] Linde, K., Randhagen, B., Beer, J., Dentchev, V., Marinova, S.,
de Verg, L.L., Cogan, J.P., Snoy, P.J., Sadoff, J.C., Formal, S.B. Vassilev, T. and Bratoyeva, M. (1993) Shigella flexneri 2a and
and Levine, M.M. (1992) Safety, immunogenicity and efficacy in sonnei I vaccine with two attenuating markers: construction,
monkeys and humans of invasive Escherichia coli K-12 vaccine tolerability and immunogenicity in 143 children aged 3–17 years.
candidates expressing Shigella flexneri 2a somatic antigen. Infect. Vaccine 11, 197–199.
Immun. 60, 2218–2224. [155] Ahmed, Z.U., Sarker, M.R. and Sack, D.A. (1990) Protection of
[140] Kotloff, K.L., Losonsky, G.A., Nataro, J.P., Wasserman, S.S., adult rabbits and monkeys from lethal shigellosis by oral
Hale, T.L., Taylor, D.N., Newland, J.W., Sadoff, J.C., Formal, immunization with a thymine-requiring and temperature-sensi-
S.B. and Levine, M.M. (1995) Evaluation of the safety, immu- tive mutant of Shigella flexneri Y. Vaccine 8, 153–158.
nogenicity and efficacy in healthy adults of four doses of live oral [156] Ashraf, M.M., Giri, D.K., Batra, H.V., Khandekar, P., Ahmed,
hybrid Escherichia coli–Shigella flexneri 2a vaccine strain EcSf2a- Z.U. and Talwar, G.P. (1991) Potentials of Shigella flexneri Y
2. Vaccine 13, 495–502. strain TSF21 as a candidate vaccine against shigellosis: safety,
[141] Rui, X., Xu, Y., Wan, H., Su, G. and Huang, C. (1996) immunogenicity and protective efficacy in Bonnet monkeys.
Construction of a stable and non-resistant bivalent vaccine FEMS Microbiol. Immunol. 3, 165–170.
58 A.V. Jennison, N.K. Verma / FEMS Mircobiology Reviews 28 (2004) 43–58

[157] Karnell, A., Sweiha, H. and Lindberg, A.A. (1992) Auxotrophic [161] Karnell, A., Cam, P.D., Verma, N.K. and Lindberg, A.A. (1993)
live oral Shigella flexneri vaccine protects against challenge with AroD deletion attenuates Shigella flexneri strain 2457T and
S. flexneri of different serotypes. Vaccine 10, 167–174. makes it a safe and efficacious oral vaccine in monkeys. Vaccine
[158] Karnell, A., Stocker, B.A., Katakura, S., Reinholt, F.P. and 11, 830–836.
Lindberg, A.A. (1992) Live oral auxotrophic Shigella flexneri [162] Noregia, F., Wang, J., Losonsky, G., Maneval, D., Hone, D. and
SFL124 vaccine with a deleted aroD gene: characterisation and Levine, M. (1994) Construction and characterization of attenu-
monkey protection studies. Vaccine 10, 389–394. ated delta aroA delta virG Shigella flexneri 2a strain CVD 1203, a
[159] Li, A., Cam, P.D., Islam, D., Minh, N.B., Huan, P.T., Rong, prototype live oral vaccine. Infect. Immun. 62, 5168–5172.
Z.C., Karlsson, K., Lindberg, A.A. and Lindberg, G. (1994) [163] Noriega, F.R., Losonsky, G.A., Wang, J.Y., Formal, S.B. and
Immune response in vietnamese children after a single dose of the Levine, M.M. (1996) Further characterisation of aroA virG
auxotrophic, live Shigella flexneri Y vaccine strain SFL124. J. Shigella flexneri 2a strain CVD 1203 as a mucosal Shigella
Infect. 28, 11–23. vaccine and as a live-vector vaccine for delivering antigens of
[160] Li, A., Karnell, A., Huan, P.T., Cam, P.D., Minh, N.B., Tram, enterotoxigenic Escherichia coli. Infect. Immun. 64, 23–27.

Downloaded from https://academic.oup.com/femsre/article-abstract/28/1/43/635550 by guest on 10 December 2019


L.N., Quy, N.P., Trach, D.D., Karlsson, K. and Lindberg, G. [164] Barzu, S., Fontaine, A., Sansonetti, P.J. and Phalipon, A. (1996)
(1993) Safety and immunogenicity of the live oral auxotrophic Induction of a local anti-IpaC antibody response in mice by use
Shigella flexneri SFL124 in adult vietnamese volunteers. Vaccine of a Shigella flexneri 2a vaccine candidate: implications for use of
11, 180–189. IpaC as a protein carrier. Infect. Immun. 64, 1190–1196.

Potrebbero piacerti anche