Está en la página 1de 7

Microbes and Infection 5 (2003) 715–721

www.elsevier.com/locate/micinf

Forum in immunology

Molecular and cellular mechanisms of action of the vacuolating cytotoxin


(VacA) and neutrophil-activating protein (HP-NAP) virulence factors
of Helicobacter pylori
Cesare Montecucco a,b,*, Marina de Bernard a,b
a
Dipartimento di Scienze Biomediche, Università di Padova, Via G. Colombo 3, 35121 Padua, Italy
b
Istituto Veneto di Medicina Molecolare, Via Orus 2, Padua, Italy

Abstract

Helicobacter pylori has elaborated a unique set of virulence factors that allow it to colonise the stomach wall. These factors include urease,
helicoidal shape, flagella and adhesion molecules. Here we discuss the molecular characteristics and mechanisms of action of the vacuolating
cytotoxin, VacA, and the neutrophil-activating protein, HP-NAP. Their activities are discussed in terms of tissue alterations, which promote the
release of nutrients necessary for the growth and survival of the bacterium in its nutrient-poor ecological niche.
© 2003 Éditions scientifiques et médicales Elsevier SAS. All rights reserved.

Keywords: Helicobacter; Inflammation; Cell vacuolation; Epithelia; Neutrophils

1. Introduction lumen with values in the pH range 1–4. Such acid conditions
promote the denaturation of most proteins and their hydroly-
Helicobacter pylori infects the stomach of more than 50% sis by gastric proteases. H. pylori can survive these harsh
of the entire human population [1]. Since its isolation and conditions by buffering its internal and surrounding pH via
identification [2], this microaerophilic Gram-negative bacte- the enzymatic activity of urease with production of ammonia
rium has been associated with chronic superficial gastritis and bicarbonate ions. It is believed that H. pylori rapidly
(inflammation of the gastric mucosa), chronic active gastritis leaves the stomach lumen to enter the mucus layer which
(gastric mucosa inflammation with polymorphonuclear cell covers and protects the mucosa against ulceration due to the
infiltration), peptic ulcer disease (gastric and duodenal ul- gastric fluids.
cers), and gastric cancer [3]. To date, a number of virulence The mucus layer has unique barrier properties and is
factors have been described and well characterised in H. py- relatively impermeable even to small molecules. In addition,
lori. The molecular mechanisms and involvement in the hydrogen ions can freely diffuse from the apical portion of
H. pylori-associated disease processes of two of these viru- the mucosal epithelial cells into the gastric lumen, but not
lence factors will be dealt with in this chapter. vice versa. In contrast, bicarbonate anions (and probably iron
H. pylori occupies a particular ecological niche consisting and nickel ions which are necessary for H. pylori growth) are
of the apical surface of stomach epithelial cells and within poorly permeable. Thus, a pH gradient exists inside the
the protective mucus layer. This environment has peculiar mucus film characterised by slightly acidic values near the
features, which are worth recalling, to understand the activi- apical cell membrane in normal conditions, or by more acid
ties of the virulence factors discussed below. To reach this pH if the thickness of the mucus layer is reduced in patho-
particular site, the bacterium must cross the oral cavity and logical conditions.
the oesophageal tract, which are characterised by the pres- H. pylori can enter the mucus because it releases mucus
ence of hydrolytic enzymes and a neutral pH. Once through hydrolysing enzymes, it has a helicoidal shape and it is
the pylorus, the bacterium faces the acidity of the stomach propelled by flagella. These bacteria adhere strongly to the
apical cell membrane via adhesins and by inducing a reor-
* Corresponding author. Tel.: +39-49-827-6058; fax: +39-49-827-6049. ganisation of the plasma membrane of the host gastric epi-
E-mail address: cesare@civ.bio.unipd.it (C. Montecucco). thelial cells [4]. In this particular region of the body, the
© 2003 Éditions scientifiques et médicales Elsevier SAS. All rights reserved.
DOI: 10.1016/S1286-4579(03)00124-2
716 C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721

supply of nutrients, including ions essential for bacterial


growth, is very limited. Several H. pylori genes are, there-
fore, expected to encode for proteins that affect the physi-
ological state of the stomach cells and tissues in order to
favour H. pylori growth. Here, we will focus on two such
proteins, which are also major virulence factors: the vacu-
olating cytotoxin (VacA) and the neutrophil-activating pro-
tein (HP-NAP).

2. Vacuolating cytotoxin (VacA)

2.1. Structure and binding to cells

The production of a cytotoxin by H. pylori was deduced


by the seminal finding that the supernatants of H. pylori
isolates induced striking vacuolar degeneration in cultured
eukaryotic cells [5]. This effect was subsequently shown to
be caused by a secreted protein toxin, thereafter termed VacA
[6].
The vacA gene encodes a pro-toxin approximately
140 kDa in mass; both an amino-terminal signal sequence
and a carboxy-terminal fragment are proteolitically cleaved
during the process of VacA secretion and an ~88-kDa mature
toxin is exported [6,7]. Mature toxin molecules are released
into the extracellular space, or may be retained on the surface
of the bacterium [8]. The single polypeptide forming the
mature VacA tends to undergo cleavage at the site of an
exposed, protease-sensitive loop into N-terminal 34-kDa
(p37) and C-terminal 54-kDa (p58) fragments [9]. These two
fragments remain non-covalently associated, and may corre-
spond to distinct structural and functional subunits. VacA is Fig. 1. Structure and activity of VacA. (a) Low-resolution structure of
purified from the culture supernatant as an oligomer with a oligomeric VacA: alternate monomers are shaded differently to represent the
mass >900 kDa [6]. Imaging by deep-etch electron micros- interaction of the P37 portion of one monomer with the P58 portion of the
adjacent monomer. The dotted line represents the loop connecting P58 and
copy shows individual VacA oligomers as flower-shaped P37. (b) Current model of the cellular activities of VacA. The toxin binds to
complexes about 30 nm in diameter (Fig. 1 panel a). These the apical portion of epithelial cells and inserts into the plasma membrane
structures are thought to be composed of one or two rings, via hydrophobic protein–lipid interactions. This insertion leads to the for-
each made up of six to seven VacA monomers [9,10]. Acidic mation of anion-selective channels of low conductance, which are capable of
[11,12] or basic [13] pH solutions disassemble the oligomers releasing bicarbonate, chloride and urea from the cell cytosol. Bicarbonate
and urea can thus enter the carbon and nitrogen cycles of H. pylori.
into membrane-inserting monomers [14] and are associated Eventually, the VacA toxin channel is internalised and changes the anion
with a marked enhancement of VacA cytotoxicity [11,13]. permeability of late endosomal compartments, with enhancement of the
Transfection of HeLa cells with plasmids expressing the vacuolar ATPase proton pumping activity. In the presence of weak bases, and
amino-terminal 422 amino acids of VacA is sufficient to in particular of the ammonia generated by the H. pylori urease, osmotically
induce vacuolisation of HeLa cells [15,16]. Small trunca- active acidotropic ions (NH4+) will accumulate in the endosomes. This leads
to water influx and vesicle swelling, an essential step in vacuole formation.
tions, internal deletions, and several point mutations in the Somehow, the VacA toxin is also capable of altering cell–cell junctions and
amino-terminal portion of VacA abrogate toxin activity when of modifying the trans-epithelial electrical resistance, with concomitant
assessed in transiently transfected cells [17]. Similarly, puri- increased fluxes of iron and nickel ions and small metabolites from the
fied toxin from a mutant Helicobacter strain, carrying a mucosa to the bacterium, thus providing it with essential nutrients. Adapted
deletion of amino acid residues 6–27, fails to induce cyto- from [42].
plasmic vacuolation, is defective in the capacity to form
membrane channels (see below) and inhibits the activity of been suggested to be especially important for cell line-
the wild-type toxin [18]. Taken together, these data suggest specific recognition and binding [19].
that the amino-terminal region of VacA plays a very impor- Although saturable binding of VacA to sensitive cell lines
tant role in toxin activity and that the active membrane- suggests the presence of specific receptor sites, at least three
inserted toxin is an oligomer. separate proteins have been implicated in toxin recognition
The first step of VacA entry into target cells consists of the [12,20–22]. In addition, cell sensitivity to VacA has been
binding to the plasma membrane. Residues 480 and 700 have associated with the presence of one or several GPI-anchored
C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721 717

protein(s) [23,24]. Depletion of plasma membrane choles- in the human stomach. Indeed, evidence for a reduced im-
terol decreases the amount of VacA internalised into HeLa mune response at the level of the stomach mucosa was found
cells by more than 70%, and cell-bound VacA was found to [35].
be associated with rafts, suggesting that VacA may directly
associate with cholesterol-rich microdomains [23–25]. 2.3. Effect on polarised epithelial monolayers
and intracellular survival of H. pylori
2.2. Cell vacuolation and endocytic pathway alteration
Addition of VacA to various polarised epithelial monolay-
VacA induces the formation of membrane-delimited ers in vitro results in a rapid increase in ion conductivity,
‘vacuoles’ in intoxicated cells. Membranes of these vacuoles which can be measured as a drop in trans-epithelial resis-
contain both late endosomal and lysosomal markers [26]. tance (TER). This appears to be due to modulation of the
Vacuolation depends not only on VacA but also on the pres- resistance of cell–cell junctions (the paracellular pathway)
ence in the extracellular medium of permeant weak base, [36]. This effect of VacA is not accompanied by either vacu-
such as ammonia [6]. Several cellular enzymes required for olation or inhibition of epidermal growth factor degradation;
vacuole development and maintenance have been identified TER decrease is not inhibited by bafilomycin A1 and is not
[27–29]. Vacuoles observed in biopsies of stomach mucosa influenced by the presence of weak bases, in contrast to the
from H. pylori-infected patients and in HeLa cells in culture effect of these compounds on vacuolation. Not only ions but
contain electron dense material, but are largely devoid of the also small neutral molecules with a molecular weight
multivesicular bodies which are characteristic of late endo- <450 kDa (for example, mannitol and sucrose) exhibit in-
somes (LE) and lysosomes (LY) [30]. It appears that VacA creased trans-epithelial diffusion upon VacA treatment.
induces considerable rearrangement of the organisation of These results indicate that VacA increases the supply of
LE and LY, with extensive membrane fusion and swelling. essential nutrients that are necessary for bacterial growth on
These events do not require SNARE proteins, and it has been the mucosa. This action has been proposed to enhance H.
suggested that the vacuolar limiting membrane results from pylori growth in the stomach by allowing increased diffusion
successive processes of fusion between the internal mem- of nutrients from gastric mucosa into the mucus layer [36].
branes among themselves and with the limiting membrane of Recently, VacA was shown to improve the intracellular
late endosomal compartments [31]. survival of H. pylori within AGS cells (human gastric cell
A role for dynamin, a GTPase functioning in intracellular line derived from an antral adenocarcinoma) [37]. If con-
vesicle formation, in VacA-induced vacuolisation was sug- firmed, this would represent another major role of VacA, as it
gested by the finding that a dominant negative mutant of emerges that intracellular vacuoles may constitute a reservoir
dynamin inhibits cell vacuolisation in transfected cells with- of live H. pylori, one that is difficult to be reached by
out affecting VacA binding or internalisation [32]. antibiotics and by inflammatory cells [38]. Moreover, H. py-
VacA alters the functions of the late endocytic pathway lori strains expressing VacA interrupt phagosome maturation
and LY even in the absence of detectable vacuolation. With in macrophage cell line RAW 264.7 and in human
no added weak bases, treatment of HeLa cells with VacA macrophage-like cell line THP-1: failure of macrophages to
results in missorting of lysosomal hydrolases to the extracel- kill bacteria could explain, at least in part, the persistence of
lular medium, and impairs the degradation of extracellular H. pylori [39].
ligands, such as epidermal growth factor. Both effects may be
ascribed to the capacity of VacA to partially neutralise the pH 2.4. Biophysical and biochemical activities
of acidic cellular compartments [33]. VacA-induced alter-
ations in the function of the endocytic pathway also probably Studies with planar lipid bilayers showed that VacA forms
account for the capacity of the toxin to interfere with antigen anion-selective and voltage-dependent channels only at low
processing by B lymphocytes [34]. pH or after low-pH pre-activation [40]. The VacA channel in
The possible impairment of the intracellular endocytic the membrane is hexameric and assembles with the same
pathway may have cytotoxic implications. In fact, protein functional properties on the plasma membrane of HeLa cells
degradation is a cellular function essential for the removal of [41]. After binding and pore formation, VacA is slowly inter-
non-functional cell membrane proteins and extracellular nalised by endocytosis, and the toxin anion channel, due to
ligands and for the consequent re-utilisation of their amino its acid resistance, is expected to permeabilise to anions the
acids. Moreover, the processing of protein antigens by im- endocytic membrane as well. However, the activity of such
mune antigen-presenting cells takes place mainly inside the intracellular anion-specific channels is only expected to be of
antigen-processing compartment, a specialised form of importance to organelles endowed with the vacuolar ATPase
LE/LY compartment, which is capable of fusing with the (v-ATPase) proton pump. This pump is electrogenic, i.e. as it
plasma membrane. The inhibition of antigen processing and acidifies the lumen, it generates a proton gradient that de-
presentation by VacA could be part of a strategy of survival presses its further activity. The presence of an anion channel
for H. pylori, because the depression of antigen processing should strongly promote the v-ATPase activity, leading to an
within antigen-presenting cells of the mucosa could signifi- enhanced accumulation of protons, which in turn would drive
cantly contribute to the long-lasting infection with H. pylori the uptake of weak bases that can permeate through the
718 C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721

membrane. The relevance of the VacA channel activity to the sphingomyelin [53]. Purified recombinant HP-NAP has been
proton pumping of v-ATPase is supported by the finding that produced in Bacillus subtilis to avoid contamination by
chloride channel defective cells have fewer acidic intracellu- Escherichia coli LPS. This purified material is chemotactic
lar organelles. An increased lumenal concentration of anions for human neutrophils and monocytes [51] and induces sur-
and cations has been suggested to cause an osmotically face expression of b2-integrins which are necessary for en-
driven swelling with generation of vacuoles [42] (Fig. 1 dothelial trans-migration [50], suggesting that HP-NAP
panel b). Accordingly, anion-channel blockers inhibit vacu- plays a role in the accumulation of these cells at the site of
olation of HeLa cells exposed to VacA [41,43]. Moreover, infection with H. pylori. HP-NAP is a powerful stimulant of
mutations within the amino-terminal hydrophobic region of the production of reactive oxygen radicals. HP-NAP acts via
VacA, which impair membrane channel formation inhibit a cascade of intracellular activation events (Fig. 2), including
vacuolation [44]. Recent evidence that cell vacuolation re- increase of cytosolic calcium ion concentration and phospho-
quires extracellular chloride and is indeed associated with rylation of proteins, leading to the assembly of functional
intracellular accumulation of this anion, of NH4+ and of NADPH oxidase on the neutrophil plasma membrane
water further support this model [45]. The importance of the through a PTX-sensitive pathway involving ERK and p38-
anion-channel activity of VacA is also confirmed by the fact MAPK [54].
that the anion channel blocker NPPB prevents and reverses HP-NAP has also been shown to increase the synthesis of
the VacA-induced decrease of TER of polarised epithelia tissue factor and the secretion of the inhibitor-2 of the plas-
[41]. Also the VacA-induced apical anion secretion found in minogen activator in mononuclear cells [55]. By inducing the
rat intestine [46] can be ascribed to its anion-channel activity. coordinate expression of cell pro-coagulant and antifibrin-
Very recently, Debellis et al. [47] were able to monitor pH olytic activities, HP-NAP might favour fibrin deposition and
changes in epithelial cells of the stomach mucosa of frogs contribute to the inflammatory reaction of gastric mucosa
and have found that VacA forms an NPPB-sensitive perme- elicited by H. pylori [55]. HP-NAP is also capable of cross-
ation pathway that allows the exit of bicarbonate anions from ing epithelial monolayers and of inducing the activation of
the cell cytosol. Such anions could contribute to the buffering the underlying mast cells [56]. These data further support the
of the apical domain of the tissue and could enter the carbon idea that HP-NAP has an important role in the in vivo trig-
cycle of H. pylori. gering and maintaining of the inflammatory events observed
Given the importance of urea in H. pylori metabolism and during H. pylori infection. Once released from the bacte-
survival, the finding that urea permeates out of the cell rium, HP-NAP would cross the stomach epithelial layer,
through the VacA channel may be a very relevant one [48], as reaching the underlying tissue where mast cells reside. The
VacA could contribute to bacterial growth also by inducing subsequent activation of mast cells by HP-NAP with release
the release of urea on the apical domain. The urea permease of the content of granules and of the pro-inflammatory cytok-
inhibitor phloretin inhibits urea permeation through the VacA ine IL-6 is known to recruit monocytes and neutrophils.
channel and also cell vacuolation and TER decrease in epi- Thus, HP-NAP can act at different stages of the inflammatory
thelia [48], reinforcing the central role of the VacA channel in response by activating mast cells with release of pro-
its multiple biological effects. inflammatory molecules able to activate neutrophils and
monocytes, and additionally by acting directly on neutro-
phils and monocytes by promoting their recruitment and
3. H. pylori neutrophil-activating protein activation.
The napA gene is highly conserved within 12 different
isolates of H. pylori, at variance from vacA, which may
The infiltration of neutrophils and mononuclear inflam- indicate a lack of immune selection for diversification of
matory cells within the H. pylori-infected stomach mucosa is HP-NAP. The atomic structure of HP-NAP shows a ball-
a common finding, and the degree of mucosal damage corre- shaped docecamer formed by four-helix bundled subunits
lates with neutrophil infiltration. This bacterium induces the with a hollow central part, similar to the E. coli DNA-binding
release of several pro-inflammatory cytokines from the tissue protein Dps [57]. Dps proteins are a diverse family of bacte-
[49]. In addition, one or more protein components present in rial stress proteins that are induced during periods of nutrient
H. pylori extracts directly attract and activate neutrophils and limitation. However, unlike Dps, which binds DNA, HP-
other inflammatory cells. Among these, a 150-kDa oligo- NAP can bind up to 500 atoms of iron per dodecamer. Indeed,
meric protein isolated from H. pylori was found to promote HP-NAP was originally thought to be a bacterial ferritin,
neutrophil adhesion to endothelial cells [50]. This protein based on the nucleotide sequence homologies [58]. The nap
was designated HP-NAP (H. pylori neutrophil-activating gene has several A+T-rich regions of dyad symmetry imme-
protein) because of its ability to induce neutrophils to pro- diately upstream of its start codon, which could function as
duce reactive oxygen radicals [51]. HP-NAP is released in Fur-regulated promoters for iron regulation. Similar A+T-
the medium, most likely after cell lysis, and binds to the rich regions exist upstream of the H. pylori ferritin pfr gene.
bacterial surface, where it can act as an adhesin, mediating All these data indicate that HP-NAP is important for iron
binding to mucin [52] or to polymorphonuclear leukocyte uptake by H. pylori. Iron is an essential nutrient for the
C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721 719

Fig. 2. Scheme of the intracellular events involved in HP-NAP activation of human leukocytes. HP-NAP binds to a specific receptor, which is coupled to a
trimeric G protein sensitive to pertussis toxin. Binding triggers the entry of calcium via plasma membrane calcium channels and via the activation of channels
located on the endoplasmic reticulum, which are opened by IP3, produced by activated phospholipase C. The activated HP-NAP receptor also activates a PI3
kinase, which is inhibited by wortmannin. The rise in the cytosolic calcium concentration and the activity of PI3 kinase leads to phosphorylation of the cytosolic
subunits of the NADPH oxidase and to their migration to the plasma membrane, causing enzyme activation with production of superoxide anions. HP-NAP
rapidly activates ERK and p38, and this activation is essential for the HP-NAP-induced superoxide anion generation, adhesion and chemotaxis of human
neutrophils. Adapted from [42].

growth of most bacteria, including H. pylori. The unique References


niche inhabited by H. pylori is probably very poor in iron,
and H. pylori possesses several mechanisms for iron uptake. [1] M.J. Blaser, Helicobacter pylori: microbiology of a “slow” bacterial
These observations suggest that HP-NAP may have a role in infection, Trends Microbiol. 1 (1993) 255–259.
iron binding, but recent data from our laboratory reveal that [2] J.R. Warren, B.J. Marshall, Unidentified curved bacilli on gastric
HP-NAP is constitutively expressed under iron-depleted epithelium in active chronic gastritis, Lancet 1 (1983) 1273–1275.
conditions, its expression is not regulated by the presence or [3] N.I.H. Consensus Development Panel on Helicobacter pylori in peptic
ulcer disease, Helicobacter pylori in peptic ulcer disease, J. Am. Med.
absence of iron and that it does not play a part in the metal Assoc. 272 (1994) 65–69.
resistance of H. pylori [59]. An alternative possibility is that [4] A. Covacci, R. Rappuoli, Tyrosine-phosphorylated bacterial proteins:
HP-NAP has evolved unique properties of a pro- Trojan horses for the host cell, J. Exp. Med. 191 (2000) 587–592.
inflammatory molecule to induce a very moderate state of [5] R.D. Leunk, P.T. Johnson, B.C. David, W.G. Kraft, D.R. Morgan,
inflammation, which would promote H. pylori growth by the Cytotoxin activity in broth-culture filtrates of Campylobacter pylori,
release of nutrient factors from the inflammed tissue. The J. Med. Microbiol. 26 (1988) 93–99.
comparison of the structure of HP-NAP with those of similar [6] T.L. Cover, M.J. Blaser, Purification and characterization of the vacu-
molecules produced by other bacteria shows the unique pres- olating toxin from Helicobacter pylori, J. Biol. Chem. 267 (1992)
10570–10575.
ence of a cluster of positive charges, which could be involved
[7] W. Schmitt, R. Haas, Genetic analysis of the Helicobacter pylori
in the activation of inflammatory cells [57].
vacuolating cytotoxin: structural similarities with the IgA protease
type of exported protein, Mol. Microbiol. 12 (1994) 307–319.
[8] J.L. Telford, P. Ghiara, M. Dell’Orco, M. Comanducci, D. Burroni,
M. Bugnoli, M.F. Tecce, S. Censini, A. Covacci, Z. Xiang, E. Papini,
C. Montecucco, L. Parente, R. Rappuoli, Gene structure of the Heli-
Acknowledgements cobacter pylori cytotoxin and evidence of its key role in gastric
disease, J. Exp. Med. 179 (1994) 1653–1658.
[9] P. Lupetti, J.E. Heuser, R. Manetti, S. Lanzavecchia, P.L. Bellon,
Work carried out in the authors’ laboratories is supported R. Dallai, R. Rappuoli, J.L. Telford, Oligomeric and subunit structure
by the CNR-MURST 5% Project and by MURST 40% of the Helicobacter pylori vacuolating cytotoxin, J. Cell Biol. 133
Project on Inflammation. (1996) 801–807.
720 C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721

[10] J.M. Reyrat, S. Lanzavecchia, P. Lupetti, M. de Bernard, C. Pagliac- [26] M. Molinari, C. Galli, N. Norais, J.L. Telford, R. Rappuoli, J.P. Luzio,
cia, V. Pelicic, M. Charrel, C. Ulivieri, N. Norais, X. Ji, V. Cabiaux, C. Montecucco, Vacuoles induced by Helicobacter pylori toxin con-
E. Papini, R. Rappuoli, J.L. Telford, 3D imaging of the 58 kDa cell tain both late endosomal and lysosomal markers, J. Biol. Chem. 272
binding subunit of the Helicobacter pylori cytotoxin, J. Mol. Biol. 290 (1997) 25339–25344.
(1999) 459–470. [27] E. Papini, E. Gottardi, B. Satin, M. de Bernard, J.L. Telford, P. Mas-
[11] M. de Bernard, E. Papini, V. de Filippis, E. Gottardi, J.L. Telford, sari, R. Rappuoli, S.B. Sato, C. Montecucco, The vacuolar ATPase
R. Manetti, A. Fontana, R. Rappuoli, C. Montecucco, Low pH acti- proton pump is present on intracellular vacuoles induced by Helico-
vates the vacuolating toxin of Helicobacter pylori, which becomes bacter pylori, J. Med. Microbiol. 44 (1996) 1–6.
acid and pepsin resistant, J. Biol. Chem. 270 (1995) 23937–23940. [28] E. Papini, B. Satin, C. Bucci, M. de Bernard, J.L. Telford, R. Manetti,
[12] T.L. Cover, P.I. Hanson, J.E. Heuser, Acid-induced dissociation of R. Rappuoli, M. Zerial, C. Montecucco, The small GTP binding
VacA, the Helicobacter pylori vacuolating toxin, reveals its pattern of protein rab7 is essential for cellular vacuolation induced by Helico-
assembly, J. Cell Biol. 138 (1997) 759–769. bacter pylori cytotoxin, EMBO J. 16 (1997) 15–24.
[13] K. Yahiro, T. Niidome, M. Kimura, T. Hatakeyama, H. Aoyagi, [29] N.A. Hotchin, T.L. Cover, N. Akhtar, Cell vacuolation induced by the
H. Kurazono, K. Imagawa, A. Wada, J. Moss, T. Hirayama, Activation VacA cytotoxin of Helicobacter pylori is regulated by the Rac1
of Helicobacter pylori VacA toxin by alkaline or acid conditions GTPase, J. Biol. Chem. 275 (2000) 14009–14012.
increases its binding to a 250-kDa receptor protein–tyrosine phos-
[30] T.L. Cover, A.H. Susan, M.J. Blaser, Characterization of HeLa cell
phatase beta, J. Biol. Chem. 274 (1999) 36693–36699.
vacuoles induced by Helicobacter pylori broth culture supernatant,
[14] M. Molinari, C. Galli, M. de Bernard, N. Norais, J.M. Ruysschaert,
Hum. Pathol. 23 (1992) 1004–1010.
R. Rappuoli, C. Montecucco, The acid activation of Helicobacter
pylori toxin VacA: structural and membrane binding studies, Bio- [31] M. de Bernard, M. Moschioni, A. Habermann, G. Griffiths, C. Mon-
chem. Biophys. Res. Commun. 24 (1998) 334–340. tecucco, Cell vacuolization induced by Helicobacter pylori VacA
[15] M. de Bernard, D. Burroni, E. Papini, R. Rappuoli, J.L. Telford, cytotoxin does not depend on late endosomal SNAREs, Cell. Micro-
C. Montecucco, Identification of the Helicobacter pylori VacA toxin biol. 4 (2002) 11–18.
domain active in the cell cytosol, Infect. Immun. 66 (1998) [32] J. Suzuki, H. Ohnsihi, H. Shibatam, A. Wada, T. Hirayama, T. Iiri,
6014–6016. N. Ueda, C. Kanamaru, T. Tsuchida, H. Mashima, H. Yasuda,
[16] D. Ye, D.C. Willhite, S.R. Blanke, Identification of the minimal T. Fujita, Dynamin is involved in human epithelial cell vacuolation
intracellular vacuolating domain of the Helicobacter pylori vacuolat- caused by the Helicobacter pylori-produced cytotoxin VacA, J. Clin.
ing toxin, J. Biol. Chem. 274 (1999) 9277–9282. Invest. 107 (2001) 363–370.
[17] D. Ye, S.R. Blanke, Mutational analysis of the Helicobacter pylori [33] B. Satin, N. Norais, J.L. Telford, R. Rappuoli, M. Murgia, C. Mon-
vacuolating toxin amino terminus: identification of the amino acids tecucco, E. Papini, Vacuolating toxin of Helicobacter pylori inhibits
essential for cellular vacuolation, Infect. Immun. 68 (2000) maturation of procathepsin D and degradation of epidermal growth
4354–4357. factor in HeLa cells through a partial neutralization of acidic intracel-
[18] A.D. Vinion-Dubiel, M.S. McClain,M. Czajkowsky, H. Iwamoto, lular compartments, J. Biol. Chem. 272 (1997) 25022–25028.
D. Ye, P. Cao, W. Schraw, G. Szabo, S.R. Blanke, Z. Shao, T.L. Cover, [34] M. Molinari, M. Salio, C. Galli, N. Norais, R. Rappuoli, A. Lanzavec-
A dominant negative mutant of Helicobacter pylori vacuolating toxin chia, C. Montecucco, Selective inhibition of Li-dependent antigen
(VacA) inhibits VacA-induced cell vacuolation, J. Biol. Chem. 274 presentation by Helicobacter pylori toxin VacA, J. Exp. Med. 187
(1999) 37736–37742. (1998) 135–140.
[19] H.J. Wang, W.C. Wang, Expression and binding analysis of GST- [35] M. Shirai, T. Arichi, T. Nakazawa, J.A. Berzofsky, Persistent infection
VacA fusions reveals that the C-terminal approximately 100-residue by Helicobacter pylori down-modulates virus-specific CD8+ cyto-
segment of exotoxin is crucial for binding in HeLa cells, Biochem. toxic T cell response and prolongs viral infection, J. Infect. Dis. 177
Biophys. Res. Commun. 278 (2000) 449–454. (1998) 72–80.
[20] P.I. Padilla, A. Wada, K. Yahiro, M. Kimura, T. Niidome, H. Aoyagi, [36] E. Papini, B. Satin, N. Norais, M. de Bernard, J.L. Telford, R. Rap-
A. Kumatori, M. Anami, T. Hayashi, J. Fujisawa, H. Saito, J. Moss, puoli, C. Montecucco, Selective increase of the permeability of polar-
T. Hirayama, Morphologic differentiation of HL-60 cells is associated ized epithelial cell monolayers by Helicobacter pylori vacuolating
with appearance of RPTPbeta and induction of Helicobacter pylori toxin, J. Clin. Invest. 102 (1998) 813–820.
VacA sensitivity, J. Biol. Chem. 275 (2000) 15200–15206.
[37] A.M. Petersen, K. Sorensen, J. Blom, K.A. Krogfelt, Reduced intrac-
[21] K. Seto, Y. Hayashi-Kuwabara, T. Yoneta, H. Suda, H. Tamaki, Vacu-
ellular survival of Helicobacter pylori vacA mutants in comparison
olation induced by cytotoxin from Helicobacter pylori is mediated by
with their wild-types indicates the role of VacA in pathogenesis,
the EGF receptor in HeLa cells, FEBS Lett. 431 (1998) 347–350.
FEMS Immunol. Med. Microbiol. 30 (2001) 103–108.
[22] K. Yahiro, T. Niidome, T. Hatakeyama, H. Aoyagi, H. Kurazono,
P.I. Padilla, A. Wada, T. Hirayama, Helicobacter pylori vacuolating [38] M.R. Amieva, N.R. Salama, L.S. Tompkins, S. Falkow, Helicobacter
cytotoxin binds to the 140-kDa protein in human gastric cancer cell pylori enter and survive within multivesicular vacuoles of epithelial
lines, AZ-521 and AGS, Biochem. Biophys. Res. Commun. 238 cells, Cell. Microbiol. 4 (2002) 677–690.
(1997) 629–632. [39] P.Y. Zheng, N.L. Jones, Helicobacter pylori strains expressing the
[23] V. Ricci, A. Galmiche, A. Doye, V. Necchi, E. Solcia, P. Boquet, High vacuolating cytotoxin interrupt phagosome maturation in macroph-
cell sensitivity to Helicobacter pylori VacA toxin depends on a ages by recruiting and retaining TACO (coronin 1) protein, Cell.
GPI-anchored protein and is not blocked by inhibition of the clathrin- Microbiol. 5 (2003) 25–40.
mediated pathway of endocytosis, Mol. Biol. Cell 11 (2000) [40] F. Tombola, C. Carlesso, I. Szabò, M. de Bernard, J.M. Reyrat,
3897–3909. J.L. Telford, R. Rappuoli, C. Montecucco, E. Papini, M. Zoratti,
[24] W. Schraw, Y. Li, M.S. McClain, F.G. van der Goot, T.L. Cover, Helicobacter pylori vacuolating toxin forms anion-selective channels
Association of Helicobacter pylori vacuolating toxin (VacA) with in planar lipid bilayers: possible implications for the mechanism of
lipid rafts, J. Biol. Chem. 277 (2002) 34642–34650. cellular vacuolation, Biophys. J. 96 (1999) 1401–1409.
[25] H.K. Patel, R.M. Willhite, R.M. Patel, D. Ye, C.L. Williams, [41] I. Szabò, S. Brutsche, F. Tombola, M. Moschioni, B. Satin, J.L. Tel-
E.M. Torres, K.B. Marty, R.A. MacDonald, S.R. Blanke, Plasma ford, R. Rappuoli, C. Montecucco, E. Papini, M. Zoratti, Formation of
membrane cholesterol modulates cellular vacuolation induced by the anion-selective channels in the cell plasma membrane by the toxin
Helicobacter pylori vacuolating cytotoxin, Infect. Immun. 70 (2002) VacA of Helicobacter pylori is required for its biological activity,
4112–4123. EMBO J. 18 (1999) 5517–5527.
C. Montecucco, M. de Bernard / Microbes and Infection 5 (2003) 715–721 721

[42] C. Montecucco, R. Rappuoli, Living dangerously: how Helicobacter [52] F. Namavar, M. Sparrius, E.C. Veerman, B.J. Appelmelk, C.M. Van-
pylori survives in the human stomach, Nat. Rev. Mol. Cell. Biol. 2 denbroucke-Grauls, Neutrophil-activating protein mediates adhesion
(2001) 457–466. of Helicobacter pylori to sulfated carbohydrates on high-molecular-
[43] F. Tombola, F. Oregna, S. Brutsche, I. Szabò, G. Del Giudice, R. Rap- weight salivary mucin, Infect. Immun. 66 (1998) 444–447.
puoli, C. Montecucco, E. Papini, M. Zoratti, Inhibition of the vacu-
olating and anion channel activities of the VacA toxin of Helicobacter [53] S. Teneberg, H. Miller-Podraza, H.C. Lampert, D.J. Evans Jr.,
pylori, FEBS Lett. 460 (1999) 221–225. D.G. Evans, D. Danielsson, K.A. Karlsson, Carbohydrate binding
[44] M.S. McClain, H. Iwamoto, P. Cao, A.D. Vinion-Dubiel, Y. Ly, specificity of the neutrophil-activating protein of Helicobacter pylori,
G. Szabo, Z. Shao, T.L. Cover, Essential role of a GXXXG motif for J. Biol. Chem. 272 (1997) 19067–19071.
membrane channel formation by Helicobacter pylori vacuolating
[54] H. Nishioka, I. Baesso, G. Semenzato, L. Trentin, R. Rappuoli, G. Del
toxin, J. Biol. Chem. 278 (2003) 12101–12108.
Giudice, C. Montecucco, The neutrophil activating protein of Helico-
[45] L. Morbiato, F. Tombola, S. Campello, G. Del Giudice, R. Rappuoli,
bacter pylori (HP-NAP) activates the MAPK pathway in human
M. Zoratti, E. Papini, Vacuolation induced by VacA toxin of Helico-
neutrophils, Eur. J. Immunol. 33 (2003) 840–849.
bacter pylori requires the intracellular accumulation of membrane
permeant bases, Cl(–) and water, FEBS Lett. 508 (2001) 479–483. [55] P. Montemurro, G. Barbuti, W.G. Dundon, G. Del Giudice, R. Rap-
[46] A. Guarino, M. Bisceglia, R.B. Canani, M.C. Boccia, G. Mallardo, puoli, M. Colucci, P. De Rinaldis, C. Montecucco, N. Semeraro,
E. Bruzzese, P. Massari, R. Rappuoli, J.L. Telford, Enterotoxic effect E. Papini, Helicobacter pylori neutrophil-activating protein stimu-
of the vacuolating toxin produced by Helicobacter pylori in Caco-2 lates tissue factor and plasminogen activator inhibitor-2 production by
cells, J. Infect. Dis. 178 (1998) 1373–1378. human blood mononuclear cells, J. Infect. Dis. 183 (2001)
[47] L. Debellis, E. Papini, R. Caroppo, C. Montecucco, S. Curci, Helico- 1055–1062.
bacter pylori cytotoxin VacA increases alkaline secretion in gastric
epithelial cells, Am. J. Physiol. Gastrointest. Liver Physiol. 281 [56] P. Montemurro, H. Nishioka, W.G. Dundon, M. de Bernard, G. Del
(2001) G1440–G1448. Giudice, R. Rappuoli, C. Montecucco, The neutrophil-activating pro-
[48] F. Tombola, L. Morbiato, G. Del Giudice, R. Rappuoli, M. Zoratti, tein of Helicobacter pylori is a potent stimulant of mast cells, Eur. J.
E. Papini, The Helicobacter pylori VacA toxin is a urea permease that Immunol. 32 (2002) 671–676.
promotes urea diffusion across epithelia, J. Clin. Invest. 108 (2001)
[57] G. Zanotti, E. Papinutto, W.G. Dundon, R. Battistuta, M. Seveso,
929–937.
G. Del Giudice, R. Rappuoli, C. Montecucco, Structure of the
[49] J.E. Crabtree, Cytokine response in Helicobacter pylori infection, in:
Neutrophil-activating protein from Helicobacter pylori, J. Mol. Biol.
M. Achtman, S. Suerbaum (Eds.), Helicobacter pylori: Molecular and
323 (2002) 125–130.
Cellular Biology, Norfolk Orizon Scientific Press, 2001, pp. 63–83.
[50] D.J. Evans Jr., D.J. Evans, T. Takemura, H. Nakano, H.C. Lampert, [58] D.J. Evans Jr., D.J. Evans, H.C. Lampert, H. Nakano, Identification of
D.Y. Graham, D.N. Dranger, P.R. Kvietys, Characterization of a four new prokaryotic bacterioferritins, from Helicobacter pylori,
Helicobacter pylori neutrophil-activating protein, Infect. Immun. 63 Anabaena variabilis, Bacillus subtilis and Treponema pallidum, by
(1995) 2213–2220. analysis of gene sequences, Gene 153 (1995) 123–127.
[51] B. Satin, G. Del Giudice, V. Della Bianca, S. Dusi, C. Laudanna,
F. Tonello, D. Kelleher, R. Rappuoli, C. Montecucco, F. Rossi, The [59] W.G. Dundon, H. Nishioka, A. Polenghi, E. Papinutto, G. Zanotti,
neutrophil-activating protein (HP-NAP) of Helicobacter pylori is a P. Montemurro, G. Del Giudice, R. Rappuoli, C. Montecucco, The
protective antigen and major virulence factor, J. Exp. Med. 191 (2000) neutrophil-activating protein of Helicobacter pylori, Int. J. Med.
1467–1476. Microbiol. 291 (2002) 545–550.

También podría gustarte