Vous êtes sur la page 1sur 15

IJASR International Journal of Academic Scientific Research

ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35


www.ijasrjournal.org

www.ijasrjournal.org 21 | Page




Microbiological, Biochemical and Functional Characterization of
Helicobacter pylori infection.
Review article
M.Abdelrazik*, H.Baghdadi** and Khalil Alali*

*Medical laboratory department, Fculty of applied medical sciences,
**Biochemistery & molecular medicine department, Faculty of medicine
Taiba Univerisity, Almadina Almanwara, KSA.

INTRODUCTION

In 1979 Robin Warren, a pathologist in Perth, Western Australia, began to notice that curved bacteria
were often present in gastric biopsy specimens submitted for histological examination.These organisms were not
present within the gastric mucosa but were present in the mucus layer overlying the tissue [2]. Warren found that
because they could never be isolated, they were ignored and ultimately forgotten by generations of physicians and
scientists. A young trainee in internal medicine, Barry Marshall, became interested in Warrens observations, and
together the two sought to isolate the organisms from biopsy specimens.Since the organisms had the appearance of
curved, gram-negative rods, the investigators used methods for the isolation of Campylobacter species, including
inoculating the biopsy specimens on to selective media and incubating the cultures under microaerobic conditions.
Since most Campylobacters grow within 48 h under such conditions, plates without visible growth were discarded
within 3 days. The initial cultures from approximately 30 patients were negative, but by chance one culture was
incubated for 5 days over an Easter holiday and colonies were seen [60] subsequently, organisms were isolated from
11 patients. the organism was characterized and called Campylobacter pyloridis (now known as Helicobacter
pylori). Following publication of this seminal report, investigators all over the world rapidly confirmed the presence
of these organisms in the gastric mucus [62]. It had become clear that H. pylori infection was strongly associated
with the presence of inflammation in the gastric mucosa (chronic superficial gastritis), and especially with
polymorphonuclear cell infiltration (chronic active gastritis), [31].

Marshall and Warren noted that H. pylori infection was associated with duodenal ulceration [60], and
this observation too was rapidly confirmed and extended to include gastric ulceration [30]. By 1994, the National
Institutes of Health conference concluded that H. pylori was a major cause of peptic ulcer disease and recommended
that infected individuals with ulcers should be treated to eradicate the organism [74]. Several evidences had
indicated that chronic gastritisis linked to the development of adeno carcinoma of the stomach, the most important
gastric malignancy in the world [57], but the causation of the gastritis was then unknown. In 1991,four reports first
showed associations between H. pylori infection and the presence or the development of gastric cancer [86,123]. In
1994, the International Agency for Cancer Research, of the World Health Organization, reviewed the available
evidence and declared that H. pylori was a carcinogen of humans [26].

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 22 | Page


Microbiological characteristics of H. pylori :
Morphology : H. pylori is a gram-negative bacterium with bluntly rounded ends in gastric biopsy
specimens, measuring 2 to 4 m in length and 0.5 to 1 m in width. Although usually spiral-shaped, the bacterium
can appear as a rod [49]. However, when cultured on solid medium,the bacteria assume a rod-like shape, spiral
shapes are infrequent or absent [48]. After prolonged culture on solid or in liquid medium, coccoid forms typically
predominate [38]. By electron microscopy, coccoid forms appear as U shaped bacilli with the ends of the two arms
joined by a membranous structure. Coccoid forms are metabolically active; however, they cannot be cultured in
vitro [38]. In gastric biopsy specimens, H. pylori organisms are 2.5 to 5.0 mm longand 0.5 to 1.0 mm wide; there are
four to six unipolar sheathed flagella [48], which are essential for bacterial motility. Flagella exhibit a characteristic
terminal bulb, which is an extension of the flagellar sheath [128]. The flagellar sheath exhibits the typical bilayer
structure of a membrane [128]. Ultrastructurally, when tannic acid is used as a mordant, it can be seen that the outer
membrane of H. pylori is coated with a glycocalyx-like structure; in gastric biopsy specimens, the surface of
individual bacteria may be linked to gastric epithelial microvilli by thread-like extensions of the glycocalyx [49].
The surface of viable H. pylori cells grown on agar plates is coated with 12- to 15-nm ring-shaped aggregates of
urease and HspB,a homolog of the GroEL heat shock protein [28]. Urease and HspB are also associated with the
surface of viable H. pylori invivo [43].
Respiration and Metabolism :
In tests involving routine microbiological methods, H. pylori does not appear to utilize carbohydrates
either fermentatively or oxidatively [48]. However, studies begun to elucidate metabolic pathways in H. pylori.The
studied isolates exhibit glucose kinase activity [65],which is associated with the bacterial cell membrane. In
addition,enzyme activity characteristic of the pentose phosphate pathwayhas been identified [65]. Thus, H. pylori
appears to be capable of catabolizing D-glucose. H. pylori possess specific D-glucose transporters; some
characteristics of the glucose transport system appear to be unique [64].

H. pylori exhibit urea cycle activity, which may serve as an effective mechanism to extrude excess
nitrogen from bacterial cells [66]. The Entner-Doudoroff pathway has been demonstrated in H. pylori [67].
Fumarate reductase is an essential component of the metabolism of H. pylori and as such constitutes a possible
target for therapeutic intervention [68].H. pylori can metabolize amino acids by fermentative pathways similar to
those in anaerobic bacteria [221]. Cytochromes involved in termination of the respiratory chain of H. pylori have
been characterized [59, 73]. The elevated level of CO2 required for growth of H. pylori in vitro may be due in part
to activity of the enzyme acetyl coenzyme A carboxylase [34].

H. pylori cells contain polyphosphate granules, which may function as a reserve energy source in
bacteria associated with a degenerated epithelium, where an exogenous energy source may be absent [32, 36]. Such
studies of the metabolism of H. pylori are rapidly expanding our basic understanding of this important gastric
pathogen; a practical outcome may be the identification of unique metabolic pathways which may serve as
therapeutic targets with minimal effect upon the host [33].
Metabolism : H. pylori can be grown only in chemically defined medium with the additional amino acids
arginine, histidine, isoleucine, leucine, methionine, phenylalanine and valine, and some strains also require alanine
and/or serine [21]. H. pylori are urease, catalase, and oxidase positive, characteristics which are often used in
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 23 | Page


identification of H. pylori. H. pylori can catabolize glucose, and both genomic and biochemical information
indicates that other sugars cannot be catabolized by H. pylori [5, 12].
(i) Oxidative stress defense: H. pylori is a micro aerophilic bacterium that does not tolerate high oxygen
conditions, but it requires at least 2% O
2
.This is because H. pylori uses oxygen as a terminal electron acceptor. H.
pylori cannot utilize alternative electron acceptors, such as nitrate or formate [15].
(ii) Nitrogen metabolism: The gastric environment contains amino acids and urea asa two major sources
of nitrogen. Ammonia is a key component in nitrogen metabolism as well as acid resistance [22]. Consequently, H.
pylori can utilize several alternative sources of ammonia [16, 24]. Also, H. pylori produces large amounts of urease,
which has been estimated as up to 10% of the total protein content of H. pylori. Urease is a nickel-containing
enzyme that consists of 12 UreA and 12 UreB subunits [10]. The UreA and UreB subunits have molecular masses of
27 kDa and 62 kDa [6].
(iii) Metal metabolism: Metals which are involved in a wide variety of chemical reactions. Metals are
cofactors of enzymes, catalyzing basic functions such as electron transport, redox reactions, and energy metabolism,
and are essential for maintaining the osmotic pressure of the cell.Consequently, they play an important role in the
metabolism of all organisms,. Since both metal limitation and metal overload delay growth and can cause cell death,
metal homeostasis is of critical importance to all living organisms.
(a) Nickel: H. pylori requires efficient acquisition of nickel, as it is the metal cofactor of the essential
colonization factors urease and hydrogenase. Nickel availability in human serum is very low (2 to 11 nM), and the
nickel concentration in ingested food varies significantly depending on the diet and on food sources [3].
(b) Iron: In human or animal host tissues, the concentration of free iron is too low to support bacterial
growth, as most iron is complexed into hemoglobin or chelated by transferrin in serum or by lactoferrin at mucosal
surfaces. Iron sources available in the gastric mucosa are lactoferrin, heme compounds released from damaged
tissues, and iron derived from pepsin-degraded food. It is thought that metals such as iron display increased
solubility under the acidic conditions of the gastric mucosa and that eukaryotic iron-complexing proteins display
lowered binding affinity under these conditions. The H. pylori genome encodes 11 proteins predicted to be involved
in iron transport and 2 proteins thought to function as iron storage proteins [23].

(c) Copper: Copper is a cofactor for several proteins involved in electron transport, oxidases, and
hydroxylases, but may also contribute to the formation of reactive oxygen species. H. pylori expresses several
proteins which either are involved in copper transport or may act as copper chaperones [20].
(d) Cobalt : Cobalt is a trace metal cofactor of the arginase enzyme, which plays an important role in
nitrogen metabolism of H. pylori [13] but also in modulating the immune response to H. pylori [8,9].It has been
suggested that H. pylori is exquisitely sensitive to cobalt in vitro [4], and it has been mentioned that cobalt may be
used in non antibiotic therapy of H. pylori infections [1].
Biochemical and functional characterization:
Host inflammatory responses and effector molecules produced by H. pylori infected cells cause changes
in the gastric mucosa during infection, requiring H. pylori to be able to adapt to an ever-changing environment.
Several factors with vital roles in the colonization of H. pylori and the development of disease have been described.
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 24 | Page


These factors include the urease enzyme, flagella, the vacuolating cytotoxin VacA and the cag-pathogenicity island
(cagPAI), which encodes the CagA protein and a type IV secretion (T4S) machinery that translocates CagA into
host cells [151; 139-142]. Both phosphorylated and non phosphorylated CagA have been shown to interfere with
host signalling pathways and cellular functions [110 108; 155].Adherence is important for persistent infection of a
host and is mediated by the binding of bacteria to glycoproteins and/or glycolipids present on the host cell surface.
Analysis of gastric biopsy material has shown that H. pylori populations are found deep in the mucous layer, and a
subset is attached to the surface of epithelial cells [109]. Recent reports have also suggested H. pylori to be localized
intracellularly [152; 143 144; 90]. Several receptor structures for adherence of H. pylori to human gastric epithelial
cells have been described, suggesting that H.pylori exhibits a multitude of adherence modes, which may change as
infection progresses [106]. Two functional receptors for H. pylori, fucosylated ABO/Lewis bantigens (Leb) [100,
90] and sialyl-Lewis x/a antigens (sLex/a) human gastric epithelium, only minute levels of sialylated glycans are
detected, but upon infection by H. pylori, increased expression of the inflammation/selectin as asociated sLex occurs
[133, 121]. Bacterial adherence to host cells is mediated by adhesins on the bacterial cell surface. Correspondingly,
the H. pylori blood group antigen binding adhesin (BabA) mediates binding to the ABO/Leb receptor structures
[114], and binding to sLex/a is mediated by the sialicacid binding adhesin (SabA) [133, 90].

Gram-negative bacteria continuously shed vesicles from the cell surface during their growth. These
vesiclesare 20300 nm in size and are surrounded by an outer membrane (OM) layer, which is described to have a
composition derived from the bacterias OM. Upon shedding from the bacterial cell surface, some of the under lying
periplasmic and cytoplasmic proteins, as well as DNA and RNA, are contained within the vesicles [98,124]. The
biological role of these vesicles has not been fully elucidated, but is described to be involved in toxin delivery,
protein and DNA transfer, and signalling between bacteria. Biochemical characterization of vesicles isolated from
pathogenic species has identified virulence associated factors such as toxins, invasins and hosteffector molecules
[112]. Electron micrographs of plasma from a person infected with Neisseria meningitides sero group B that died of
septic shock showed that numerous vesicles were present [136]. H. pylori vesicles were found in gastric mucosal
biopsy material and shown to contain the Vac Acyto toxin [104]. The protein content of H. pylori vesicles differs
significantlyfrom whole bacterial cells as well as of the OM.Using immunoblots,the presence of aseries of virulence
factors in the vesicles such as adherencecomponents and the oncoproteinCagAwas confirmed [98].

The biochemical characterization and functional analysis of adherence components present on the
surface of H. pylori vesicles was studied. Receptor conjugatesfor electron microscopy studies and an in vitroassay to
characterize adherence to human gastric tissue sections was used. The results of these studies provide valuable
insight into the mechanisms involved in bacterialhost interactions as they relate to H. pylori vesicles and their
intimate interaction with human gastric mucosa.

Biochemical characterization of H. pylori vesicles:
(i) Characterization of major phospholipid vesicle components:
Phospholipids present in H. pylori vesicles were identified using two-dimensional (2D) 31P,1H NMR
correlation spectra. The type and abundance of phospholipid components present in isolated vesicles, outer and inner
membrane (IM) fractions, and whole H. pylori cells was determined. By definition, IMs have fewer proteins than
OMs, thereby allowing IMs to be separated from OMs using density gradient centrifugation. Lipids were extracted
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 25 | Page


from whole bacteria, OM, IM and purified vesicles for fractions localization. For analysis of the 2D spectra, 1D
projections were integrated .Phosphatidyl ethanolamine (PE) and cardiolipin (CL) were identified asthe major
phospholipids present in all fractions. IM fractions contained considerably lower amounts of phosphatidyl glycerol
(PG) and lyso-phophatidyl etanolamine (LPE) compared with the OM or vesicle fractions. Phophatidyl choline (PC)
was below the detection limit in IM, but was readily detected in all other fractions. These data represent the
differences in phospholipid composition between the analysed fractions, and suggests that the phospholipid
composition of vesicles most closely resembles that of the OM. Helicobacter pylori membranes have been reported
to contain cholesterol [115] .The molar fraction of cholesterol estimated to be10% relative to that of total
phospholipids present in both whole bacterial cells as well as in vesicles

(ii) Characterization of major proteins present in vesicles:
The major protein components of H. pylori vesicles were analysed using mass spectrometry (MS).
Since many of the H. pylori OM proteins are of similar molecular masses, exhibit high pI values and do not resolve
on 2D gels, 1DSDS-PAGE was combined with analysis by the sensitive nanoflow LC FT-ICR MS/MS method.
Using H. pylorivesicles purified according to density, samples with adensity of 1.151.20 g ml-1 from fractions 39
were pooled. Initial separation of the samples by SDS-PAGE identified a series of 34 bands that were excised and
subjected to peptide tandem mass determination. All mass data obtained were searched by MASCOT against all
species in the NCBI database. An H.pylori proteome consists of approximately 1500 putative proteins [89, 143, 97],
and in total 306 different H. pylori proteins were identified in the 34 excised bands. Identified vesicle proteins were
classified according to clusters of orthologous groups (COG) for the total H. pylori proteome. Several H. pylori OM
proteinshave been associated with gastric disease [162], and therefore H. pylori proteins are listed as a separate
group as classified according to [89]. A majority of all OM proteins (77%) were identified in the vesicles.. Several
of the identified vesicle proteins are associated with important roles in H. pylori colonization and virulence, such as
urease subunits, the cytotoxin VacA, CagA, and the BabA and SabAadhesins. Also, g-glutamyl transpeptidase has
been shown to exhibit immunosuppressive effects similar to the VacA protein [150] and the protease HtrA [130].
Among the series of cytoplasmic proteins GroEL, catalase, metabolic and ribosomal proteins were found.A second
MS analysis (MS II) was performed on an additional vesicle sample of a narrower density interval (1.171.18 g ml-
1). Separation by SDS-PAGE resolved a series of 32 bands that were isolated and subjected to nanoflow LC FT-ICR
MS/MS analysis. From this sample, 126 different H. pylori proteins were identified.Among these, 93% were
identical to the previous analysis and 7% were specific for MS II .

Mechanisms of Infection pathogensis:
H. pylori is able to colonize and persist in a unique biological niche within the gastric lumen. The
putative pathogenic determinants of H. pylori can be divided into two major groups virulence factors, which
contribute to the pathogenetic effects of the bacterium, and maintenance factors, which allow the bacterium to
colonize and remain within the host. H. pylori has three major pathogenic effects, gastric inflammation, disruption
of the gastric mucosal barrier, and alteration of gastric physiology.

Induction of gastric inflammation:
(i) Interleukin-8: Interleukin-8 (IL-8) is a small peptide (chemokine) secreted by a variety of cell types,
which serves asa potent inflammatory mediator recruiting and activating neutrophils. Several studies have
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 26 | Page


demonstrated that H. pylori strains are capable of inducing IL-8 secretion from gastric carcinoma cells in vitro.
Wild-type strains that were VacA1 CagA1 produced significantly more IL-8 than did wild-type VacA2 CagA2
strains [38, 50, 78].

(ii) Neutrophil adherence: The150-kDa proteins which increases the expression of neutrophil
CD11b/CD18 and increases neutrophil adherence to endothelial cells have been characterized. The protein
designated HP-NAP, is a polymer of 10 identical subunits. The gene (napA) shows homology tothe gene encoding
the bacterio ferritin family of proteins [114].

(iii) Platelet-activating factor: Platelet-activating factor (PAF) is a phospholipid mediator which is
recognized as a potent ulcerogenic agent. Lyso-PAF is produced by gastric mucosal cells under basal conditions and
in response to gastrin in healthy persons [298]. PAF stimulates gastric acid secretion via specific parietal cell
receptors H. pylori can metabolize the non ulcerogenic precursor lyso-PAF into PAF Thus, through synthesis of
PAF, H. pylori may induce mucosal injury directly or indirectly via increased acid secretion.[81].

(iv) Lipopolysaccharide: H. pylori LPS disrupts the gastric mucus coat by interfering with the interaction
between mucin and its mucosal receptor; the anti-ulcer agent ebrotidine counteracts this effect [279]. However, the
outstanding feature of the H. pylori LPS is its low proinflammatory activity [71].This phenomenon is mediated by
its unique lipid structure and affects binding to CD-14 [54].

(v) Urease: H. pylori urease is a potent stimulus of mono nuclear phagocyte activation and inflammatory
cytokine production [146]. In vitro, urease activity also is toxic to human gastric epithelial cells [80]. Thus, urease
appears to function as both a colonization [maintenance] factor and a virulence factor. There is evidence that urease
is associated with the outer membrane of H. pylori.

Disruption of the gastric mucosal barrier:
H. pylori can inhibit the secretory response of mucus cells in vitro, indicating a potential deleterious
effect on this primary defense mechanism of the gastric mucosa [69].

(i) Phospholipase: H. pylori disrupts the protective phospholipid-rich layer at the apical membrane of
mucus cells [61]. Furthermore, changes can be induced in phospholipid layers invitro by phospholipases A2 and C
expressed by H. pylori [75]; their effects can be inhibited by bismuth salts [75].

(ii) Mucinase: H. pylori possesses a gene that is almost identical to a mucinase gene of Vibrio cholera
[79]. Such mucinase activity, if expressed in vivo, would probably contribute to disruption of the gastric mucosal
barrier.

(iii) Vacuolating cytotoxin: Almost one-half of H. pylori strains produce a vacuolating cytotoxin in vitro
[55]. This cytotoxin induces acidic vacuoles in the cytoplasm of eukaryotic cells; these vacuoles accumulate neutral
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 27 | Page


red dye, facilitating their analysis [37]. The purified vacuolating cytotoxin, which is active on a wide range of
eukaryotic cells, migrates as an approximately 87-kDa protein under denaturing, reducing conditions [63].

(iv) Reactive oxygen species : H. pylori induces the synthesis of reactive oxygen species (ROS) in gastric
mucosa in vivo.There is a positive association between the amount of ROS present, the infective load of H. pylori,
and the extent of gastricmucosal injury [39]. Smoking, drugs, and alcohol consumption had no independent effect
upon ROS production in vivo [40].There is no evidence for ROS participation in gastric mucosal injury in cases not
related to H. pylori infection [40]. H. pylori stimulates mucosal ROS production both in vitro [29] and in vivo; this
phenomenon appears to be of pathogenic significance [39].

(v) Inducible nitric oxide synthase: High-output nitric oxide production by inducible nitric oxide synthase
(iNOS) is associated with immune activation and tissue injury. H. pylori induces iNOS in macrophages in vitro
[335]. Eradication of H. pylori reduces iNOS in human gastric epithelial cells, suggesting that H. pylori also,
induces the activity of this enzyme invivo [58].

(vi) Apoptosis: H. pylori appears to induce programmed cell death (apoptosis) of gastric epithelial cells
[58] and to stimulate oxidative DNA damage in infected human gastric mucosa [21]. In addition, H. pylori appears
to inhibit gastric epithelial cell migration and proliferation [77]. Thus, H. pylori infection can induce gastric mucosal
injury both directly and indirectly.

Altered gastric homeostasis :
H. pylori infection reversibly induces expression of the acid-stimulating peptide gastrin and suppresses
expression of the acid-inhibitory hormone somatostatin [35]. These effects may not be due to H. pylori but may be
related to the degree of gastric inflammation present. H. pylori increases the duodenal acid load under some
circumstances but appears to decrease acid secretion under other conditions [85]. When acid secretion is suppressed
with omeprazole, the relative susceptibility of H. pylori in the antrum and corpus differs, yielding the appearance of
migration of bacteria from the antrum to the corpus [56].

(i) Motility: Motility is an essential colonization factor based on infection inability of the aflagellate,
nonmotile variants of H. pylori [45]. Normally, H. pylori possesses two to six polar, sheathed flagella, whose
filaments consist of two types, encoded by the flaAand flab genes [53]. These genes have been cloned; the use of
induced mutations has shown that both are essential for full motility [53] and for colonization [46]. The gene (flbA)
that codes for the H. pylori homolog of a family of conserved proteins (LcrDInvA-FlbF family), which is involved
in the regulation of motility, has been identified and cloned [83]. Mutants with mutationsin the flbAgene are
nonmotile and fail to express either flagellin or the hook protein [83].

(ii) Urease: All H. pylori isolates, as well as each of the gastric Helicobacter species, produce large
quantities of the enzyme urease. There is evidence that urease is associated with the outer membrane of H. pylori. It
is generally assumed that urease activity is required for production of a neutral microenvironment for the organism
within the gastric lumen. [42].
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 28 | Page



(iii) Catalase and superoxide dismutase: The genes encoding the superoxide dismutase and catalase
enzymes of H. pylori demonstrate significant homology to those of intracellular pathogenic microorganisms [82],
suggesting a role in resistance to killing by polymorphonuclear leukocytes. Of interest, a significant fraction of
catalase [76] and superoxide dismutase [82] is associated with the surface of viable H. pylori; whether surface
association of these enzymes is essential for protection against oxygen-dependent killing of H. pylori by neutrophils
is not known.

(iv) Heat shock protein homologs: The sequence of the gene encoding the HspB protein of H. pylori is
highly conserved compared with those of heat shock proteins of other bacteria and humans [57]. This conservation
suggests that the sequence cannot be modified without affecting the function of the protein. Based on structural
similarity, HspB may functionas a molecular chaperone for urease [44] The gene (hspB) is part of a bicistronic
operon (hspA-hspB), which has been cloned and sequenced .The hspAgene, which islocated upstream of hspB, codes
for the H. pylori homolog of the GroES heat shock protein homolog. The H. pylori hspAgene is unique in that it
contains a nickel-binding site at its Cterminus Expression of the hspAand hspBheat shock proteins together with the
H. pylori urease increases the activity of urease in functional complementation experiments .Thus, hspAmay play a
role in the integration of nickel into the functional urease molecule [84].

(v) P-type ATPase : ATPases are of particular interest in H. pylori research, because an ATPase is
presumed to be the target of the bactericidal action of proton pump inhibitors, such as lansoprazole or omeprazole,
on H. pylori [72]. AP-type ATPase has been cloned and sequenced [63]. Isogenic mutants lacking this gene were
viable; inactivation ofthe ATPase gene had no effect on the MIC of omeprazole, suggesting that this ATPase is not
the target of the bactericidal action of omeprazole [127].

(vi) Siderophores : Iron is an essential element for bacterial growth and metabolism. H. pylori does not
produce siderophores, (iron scavenging proteins) under iron-limiting conditions. However, H. pylori could grow in
the presence of human lactoferrin and has surface receptors for this iron-binding protein [51]. In contrast, the
production of siderophores was detected but did not affect the binding of lactoferrin to H. pylori proteins [52]. Thus,
it is not yet clear how H. pylori acquires iron for its growth.
Diagnosis and treatement :
Diagnosis of infection: A variety of tests are now available to diagnose H. pylori infection. Histological
examination of gastric tissue,bacterial culture, rapid urease testing, use of DNA probes, and PCR analysis, when
used to test gastric tissue, all require endoscopy; therefore they incur expense and a risk of complication due to the
procedure. In contrast, breath tests, serology, gastric juice PCR, and urinary excretion of ammonia are non invasive
tests that do not require endoscopy.The choice of test used for diagnosis of H. pylori infection will depend, in most
cases, on the clinical information sought and the local availability and cost of individual tests [25].
Treatment: Although H. pylori is sensitive to a wide range of antibiotics in vitro, they all fail as
monotherapy in vivo. In infected patients, the most effective single drug is clarithromycin, which leads to an
approximate eradication rate of 40% when given twice daily for 10 to 14 days [14]. The lack of efficacy of
monotherapy is related to the niche of H. pylori, residing at lower pH in a viscous mucus layer. Dual therapies,
combining twice-daily-dosed PPI with, in particular, amoxicillin, are still in use in some countries, but dual
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 29 | Page


therapies have mostly been replaced by triple therapies. These combine two antibiotics with either a bismuth
compound or a PPI. A further alternative is provided by quadruple therapies, which combine the bismuth compound
and PPI with two antibiotics. The exact mode of action of bismuth compounds is unknown, but H. pylori is
susceptible to these compounds both in vivo and in vitro [11]. Tetracycline, amoxicillin, imidazoles [predominantly
metronidazole and tinidazole], and a few selected macrolides [in particular clarithromycin, sometimes azithromycin]
are probably the drugs most widely used for H. pylori eradication therapy recently, the use of rifabutin [19] and
furazolidone [7] has been promoted. However, as their effectiveness is limited and many patients do not tolerate
furazolidone, the primary use of these two antibiotics is a second-line rescue therapy of patients harboring
metronidazole-resistant isolates [18].
REFERENCES

[1]. Bytzer, P., and C. O .Morain. 2005. Treatment of Helicobacter pylori. Helicobacter 10 Suppl.[ 1] :40-46.

[2]. Marshall, B. J., and J. R. Warren. 1984. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet
i:13111315.

[3]. Christensen, J. M., J. Kristiansen, N. H. Nielsen, T. Menne, and K. Byrialsen. 1999. Nickel concentrations in serum and urine of patients with
nickel eczema. Toxicol.Lett. 108:185-189.

[4]. Contreras, M., J. M. Thiberge, M. A. Mandrand-Berthelot, and A. Labigne. 2003. Characterization of the roles of NikR, a nickel-responsive
pleiotropic autoregulator of Helicobacter pylori. Mol. Microbiol. 49:947-963.

[5]. Doig, P., B. L. de Jonge, R. A. Alm, E. D. Brown, M. Uria-Nickelsen, B. Noonan, S. D. Mills, P. Tummino, G. Carmel, B. C. Guild, D. T.
Moir, G. F. Vovis, and T. J. Trust. 1999. Helicobacter pylori physiology predicted from genomic comparison of two strains. Microbiol. Mol.
Biol. Rev. 63:675-707

[6]. Dunn, B. E., G. P. Campbell, G. I. Perez-Perez, and M. J. Blaser. 1990. Purification and characterization of urease from Helicobacter pylori.
J. Biol. Chem. 265:9464-9469.

[7]. Fakheri, H., S. Merat, V. Hosseini, and R. Malekzadeh. 2004. Low-dose furazolidone in triple and quadruple regimens for Helicobacter
pylori eradication. Aliment.Pharmacol.Ther. 19:89-93

[8]. Gobert, A. P., Y. Cheng, J. Y. Wang, J. L. Boucher, R. K. Iyer, S. D. Cederbaum, R. A. Casero, Jr., J. C. Newton, and K. T. Wilson. 2002.
Helicobacter pylori induces macrophage apoptosis by activation of arginaseII. J. Immunol. 168:4692-4700.

[9]. Gobert, A. P., D. J. McGee, M. Akhtar, G. L. Mendz, J. C. Newton, Y.Cheng, H. L. Mobley, and K. T. Wilson. 2001. Helicobacter pylori
arginase inhibits nitric oxide production by eukaryotic cells: a strategy for bacterial survival. Proc. Natl. Acad. Sci. USA 98:13844-13849.

[10]. Ha, N. C., S. T. Oh, J. Y. Sung, K. A. Cha, M. H. Lee, and B. H. Oh. 2001. Supramolecular assembly and acid resistance of Helicobacter
pylori urease. Nat. Struct. Biol. 8:505-509.

[11]. Lambert, J. R., and P. Midolo. 1997. The actions of bismuth in the treatment of Helicobacter pylori infection. Aliment.Pharmacol.Ther.
11:27-33.

[12]. Marais, A., G. L. Mendz, S. L. Hazell, and F. Megraud. 1999. Metabolism and genetics of Helicobacter pylori: the genome era. Microbiol.
Mol. Biol. Rev. 63:642-674

[13]. McGee, D. J., F. J. Radcliff, G. L. Mendz, R. L. Ferrero, and H. L. Mobley.1999. Helicobacter pylori rocF is required for arginase activity
and acid protection in vitro but is not essential for colonization of mice or for urease activity. J. Bacteriol. 181:7314-7322.

[14]. Megraud, F. 1995. Rationale for the choice of antibiotics for the eradication of Helicobacter pylori. Eur. J. Gastroenterol. Hepatol. 7[Suppl.
1]:S49-S54.

[15]. Mendz, G. L., A. J. Shepley, S. L. Hazell, and M. A. Smith. 1997. Purine metabolism and the microaerophily of Helicobacter pylori. Arch.
Microbiol. 168:448-456

[16]. Merrell, D. S., M. L. Goodrich, G. Otto, L. S. Tompkins, and S. Falkow. 2003. pH-regulated gene expression of the gastric pathogen
Helicobacter pylori. Infect. Immun. 71:3529-3539.

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 30 | Page


[17]. Mohammadi, M., S. Czinn, R. Redline, and J. Nedrud. 1996. Helicobacter-specific cell-mediated immune responses display a predominant
Th1 phenotype and promote a delayed-type hypersensitivity response in the stomachs of mice. J. Immunol. 156:4729-4738.

[18]. Parente, F., C. Cucino, and G. Bianchi Porro. 2003. Treatment options for patients with Helicobacter pylori infection resistant to one or
more eradication attempts. Dig. Liver Dis. 35:523-528.

[19]. Perri, F., V. Festa, R. Clemente, M. Quitadamo, and A. Andriulli. 2000. Rifabutin-based rescue therapy for Helicobacter pylori infected
patients after failure of standard regimens. Aliment.Pharmacol.Ther. 14:311-316.

[20]. Rensing, C., and G. Grass. 2003. Escherichia coli mechanisms of copper homeostasis in a changing environment. FEMS Microbiol.Rev.
27:197-213.

[21]. Reynolds, D. J., and C. W. Penn. 1994. Characteristics of Helicobacter pylori growth in a defined medium and determination of its amino
acid requirements. Microbiology 140:2649-2656.

[22]. Stingl, K., K. Altendorf, and E. P. Bakker. 2002. Acid survival of Helicobacter pylori: how does urease activity trigger cytoplasmic pH
homeostasis? Trends Microbiol. 10:70-74.

[23]. van Vliet, A. H. M., S. Bereswill, and J. G. Kusters. 2001. Ion metabolism and transport, p. 193-206. In H. L. T. Mobley, G. L. Mendz, and
S. L. Hazell [ed.], Helicobacter pylori: physiology and genetics. ASM Press, Washington, D.C.
[24. van Vliet, A. H. M., J. Stoof, S. W. Poppelaars, S. Bereswill, G. Homuth, M. Kist, E. J. Kuipers, and J. G. Kusters. 2003. Differential
regulation of amidase- and formamidase-mediated ammonia production by the Helicobacter pylori Fur repressor. J. Biol. Chem. 278:9052-9057

[25]. Zagari, R. M., F. Bazzoli, P. Pozzato, S. Fossi, L. De Luca, G. Nicolini, D. Berretti, and E. Roda.1999. Review article: non-invasive
methods for the diagnosis of Helicobacter pylori infection. Ital. J. Gastroenterol. Hepatol

[26].Anonymous.1994. Schistosomes, liver flukes and Helicobacter pylori.IARCMonogr.Eval.Carcinog. Risks Hum.61:1241.

[28.Austin, J. W., P. Doig, M. Stewart, and T. J. Trust.1992. Structural comparison of urease and a GroEL analog from Helicobacter pylori. J.
Bacteriol.174:74707473.

[29].Bagchi, D., G. Bhattacharya, and S. J. Stohs.1996. Production of reactive oxygen species by gastric cells in association with Helicobacter
pylori. FreeRadical Res. 24:439450.

[30.Blaser, M. J. 1987. Gastric campylobacter-like organisms, gastritis and peptic ulcer disease. Gastroenterology 93:371383.

[31].Blaser, M. J. 1990. Helicobacter pylori and the pathogenesis of gastroduodenalinflammation. J. Infect. Dis. 161:626633.

[32].Bode, G., F. Mauch, H. Ditschuneit, and P. Malfertheiner.1993. Identificationof structures containing polyphosphate in Helicobacter pylori.
J. Gen.Microbiol. 139:30293033.

[33].Bottomley, J. R., C. L. Clayton, P. A. Chalk, and C. Kleanthous.1996. Cloning, sequencing, expression, purification and preliminary
characterization of a type II dehydroquinase from Helicobacter pylori.Biochem. J.319:559565.

[34].Burns, B. P., S. L. Hazell, and G. L. Mendz.1995. Acetyl-CoA carboxylase activity in Helicobacter pylori and the requirement of increased
CO2 forgrowth. Microbiology 141:31133118.

[35].Calam, J.1995. Helicobacter pylori, acid and gastrin. Eur. J. Gastroenterol. Hepatol.7:310317.

[36].Caselli, M., A. Aleotti, P. Boldrini, M. Ruina, and V. Alvisi.1993. Ultrastructuralpatterns of Helicobacter pylori. Gut 34:15071509.
63. Cover, T. L., and M. J. Blaser. 1992. Purification and characterization of the vacuolating toxin from Helicobacter pylori. J. Biol. Chem.
267:1057010575.

[37].Cover, T. L., L. Y. Reddy, and M. J. Blaser.1993. Effects of ATPase inhibitors on the response of HeLa cells to Helicobacter pylori
vacuolatingtoxin. Infect. Immun. 61:14271431.

[38].Crabtree, J. E., A. Covacci, S. M. Farmery, Z. Xiang, D. S. Tompkins, S.Perry, I. J. Lindley, and R. Rappuoli.1995. Helicobacter pylori
inducedinterleukin-8 expression in gastric epithelial cells is associated with cagApositive phenotype. J. Clin. Pathol.48:4145.

[39].Davies, G. R., N. Banatvala, C. E. Collins, M. T. Sheaff, Y. Abdi, L.Clements, and D. S. Rampton.1994. Relationship between infective load
ofHelicobacter pylori and reactive oxygen metabolite production in antralmucosa. Scand. J. Gastroenterol. 29:419424.

[40].Davies, G. R., N. J. Simmonds, T. R. Stevens, M. T. Sheaff, N. Banatvala,I. F. Laurenson, D. R. Blake, and D. S. Rampton. 1994.
Helicobacter pyloristimulatesantral mucosal reactive oxygen metabolite production in vivo.Gut35:179185.

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 31 | Page


[41].Desai, M., D. Linton, R. J. Owen, and J. Stanley.1994. Molecular typing of Helicobacter pylori isolates from asymptomatic, ulcer and
gastritis patientsby urease gene polymorphism. Epidemiol. Infect. 112:151160.

[42].Dunn, B. E., G. P. Campbell, G. I. Perez-Perez, and M. J. Blaser.1990.Purification and characterization of urease from Helicobacter pylori.
J. Biol. Chem. 265:94649469.

[43].Dunn, B. E., N. B. Vakil, B. G. Schneider, J. B. Zitzer, T. Puetz, andS. H. Phadnis.1997. Localization of Helicobacter pylori urease and
heatshock protein homolog in human gastric biopsies. Infect. Immun. 65:11811188.

[44].Dunn, B. E., R. M. Roop, II, C. C. Sung, S. A. Sharma, G. I. Perez-Perez,and M. J. Blaser. 1992. Identification and purification of a cpn60
heat shockprotein homolog from Helicobacter pylori. Infect. Immun. 60:19461951.

[45].Eaton, K. A., D. R. Morgan, and S. Krakowka.1992. Motility as a factor inthecolonisation of gnotobiotic piglets by Helicobacter pylori. J.
Med. Microbiol. 37:123127.

[46].Eaton, K. A., S. Suerbaum, C. Josenhans, and S. Krakowka.1996. Colonizationofgnotobiotic piglets by Helicobacter pylori deficient in two
flagellingenes. Infect. Immun. 64:24452448.

[47].Forman, D., D. G. Newell, F. Fullerton, J. W. Yarnell, A. R. Stacey, N. Wald,and F. Sitas. 1991. Association between infection with
Helicobacter pyloriand risk of gastric cancer: evidence from a prospective investigation. Br.Med. J. 302:13021305.z

[48].Goodwin, C. S. and J. A. Armstrong.1990. Microbiological aspects of Helicobacter pylori [Campylobacter pylori]. Eur. J. Clin.
Microbiol.9:113.

[49].Goodwin, C. S., R. K. McCulloch, J. A. Armstrong, and S. H. Wee.1987. Unusual cellular fatty acids and distinctive ultrastructure in a new
spiralbacterium [Campylobacter pyloridis] from the human gastric mucosa.J. Med. Microbiol. 19:257267.

[50].Huang, J., P. W. OToole, P. Doig, and T. J. Trust.1995. Stimulation of interleukin -8 production in epithelial cell lines by Helicobacter
pylori .Infect. Immun.63:17321738.

[51].Husson, M. O., D. Legrand, G. Spik, and H. Leclerc.1993. Iron acquisitionby Helicobacter pylori: importance of human lactoferrin. Infect.
Immun.61:26942697.

[52].Illingworth, D. S., K. S. Walter, P. L. Griffiths, and R.Barclay.1993.Siderophore production and iron-regulated envelope proteins
ofHelicobacterpylori. Int. J. Med. Microbiol. Virol.Parasitol. Infect. Dis. 280:113119.

[53].Josenhans, C., A. Labigne, and S. Suerbaum.1995. Comparative ultrastructuraland functional studies of Helicobacter pylori and
Helicobacter mustelaeflagellin mutants: both flagellin subunits, FlaA and FlaB, are necessary forfull motility in Helicobacter species. J.
Bacteriol. 177:30103020.patients. 54.Kirkland, T., S. Viriyakosol, G. I. Perez-Perez, and M. J. Blaser.1997.Helicobacter pylori
lipopolysaccharide can activate 70Z/3 cells via CD14.Infect. Immun. 65:604608.

[55].Leunk, R. D., P. T. Johnson, B. C. David, W. G. Kraft, and D. R. Morgan.1988. Cytotoxic activity in broth-culture filtrates of
Campylobacter pylori.J. Med. Microbiol. 26:9399.

[56].Logan, R. P., M. M. Walker, J. J. Misiewicz, P. A. Gummett, Q. N. Karim,and J. H. Baron.1995. Changes in the intragastric distribution of
Helicobacterpyloriduring treatment with omeprazole. Gut 36:1216.

[57].Macchia, G., A. Massone, D. Burroni, A. Covacci, S. Censini, and R.Rappuoli.1993. The hsp60 protein of Helicobacter pylori: structure
andimmune response in patients with gastroduodenal diseases. Mol. Microbiol.9:645652.

[58].Mannick, E. E., L. E. Bravo, G. Zarama, J. L. Realpe, X. J. Zhang, B. Ruiz,E. T. Fontham, R. Mera, M. J. Miller, and P. Correa. 1996.
Inducible nitricoxide synthase, nitrotyrosine, and apoptosis in Helicobacter pylori gastritis:effect of antibiotics and antioxidants. Cancer Res.
56:32383243.

[59].Marcelli, S. W., H. T. Chang, T. Chapman, P. A. Chalk, R. J. Miles, and R. K. Poole. 1996. The respiratory chain of Helicobacter pylori:
identificationof cytochromes and the effects of oxygen on cytochrome and menaquinonelevels. FEMS Microbiol.Lett.138:5964.

[60].Marshall, B. J., and J. R. Warren.1984. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet
i:13111315.

[61].Mauch, F., G. Bode, H. Ditschuneit, and P. Malfertheiner. 1993. Demonstrationof a phospholipid-rich zone in the human gastric epithelium
damagedbyHelicobacter pylori. Gastroenterology 105:16981704.

[62.McNulty, C. M. A., and D. M. Watson.1984. Spiral bacteria of the gastricantrum. Lancet i:10681069.

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 32 | Page


[63].Melchers, K., T. Weitzenegger, A. Buhmann, W. Steinhilber, G. Sachs, andK. P. Schafer.1996. Cloning and membrane topology of a P-type
ATPasefromHelicobacter pylori. J. Biol. Chem. 271:446457.

[64].Mendz, G. L., B. P. Burns, and S. L. Hazell.1995. Characterisation of glucose transport in Helicobacter
pylori.Biochim.Biophys.Acta1244:269276.

[65].Mendz, G. L., and S. L. Hazell.1991. Evidence for a pentose phosphatepathway in Helicobacter pylori. FEMS Microbiol.Lett.84:331336.

[66].Mendz, G. L., and S. L. Hazell.1996.The urea cycle of Helicobacter pylori. Microbiology 142:29592967.

[67].Mendz, G. L., S. L. Hazell, and B. P. Burns.1994. The Entner-Doudoroffpathway in Helicobacter pylori. Arch. Biochem. Biophys.312:349
356.

[68].Mendz, G. L., S. L. Hazell, and S. Srinivasan.1995. Fumaratereductase: atarget for therapeutic intervention against Helicobacter pylori.
Arch. Biochem.Biophys.321:153159.

[69].Micots, I., C. Augeron, C. L. Laboisse, F. Muzeau, and F. Megraud.1993.Mucin exocytosis: a major target for Helicobacter pylori. J. Clin.
Pathol.46:241245.

[70].Mobley, H. L., M. D. Island, and R. P. Hausinger.1995. Molecular biology of microbial ureases. Microbiol.Rev. 59:451480.

[71].Moran, A. P. 1996. The role of lipopolysaccharide in helicobacter pylori pathogenesis. Aliment.Pharmacol.Ther.10 [Suppl. 1]:3950.
[72].Nagata, K., H. Satoh, T. Iwahi, T. Shimoyama, and T. Tamura.1993.Potent inhibitory action of the gastric proton pump inhibitor
lansoprazoleagainst urease activity of Helicobacter pylori: unique action selective forH. pyloricells. Antimicrob.Agents Chemother.37:769774.

[73.Nagata, K., S. Tsukita, T. Tamura, and N. Sone.1996. A cb-type cytochrome-c oxidase terminates the respiratory chain in Helicobacter
pylori.Microbiology142:17571763.

[74.NIH Consensus Conference.1994. Helicobacter pylori in peptic ulcer disease.NIH Consensus Development Panel on Helicobacter pylori in
pepticulcer disease. JAMA 272:6569.

[75].Ottlecz, A., J. J. Romero, S. L. Hazell, D. Y. Graham, and L. M.Lichtenberger.1993. Phospholipase activity of Helicobacter pylori and its
inhibitionby bismuth salts. Dig. Dis. Sci. 38:20712080.

[76.Phadnis, S. H., M. H. Parlow, M. Levy, D. Ilver, C. M. Caulkins, J. B.Connors, and B. E. Dunn. 1996. Surface localization of Helicobacter
pyloriurease and a heat shock protein homolog requires bacterial autolysis. Infect.Immun. 64:905912.

[77].Ricci, V., C. Ciacci, R. Zarrilli, P. Sommi, M. K. Tummuru, C. Del VecchioBlanco, C. B. Bruni, T. L. Cover, M. J. Blaser, and M. Romano.
1996. EffectofHelicobacter pylori on gastric epithelial cell migration and proliferation inVitro: role of vacAand cagA. Infect. Immun. 64:2829
2833.

[78].Sharma, S. A., M. K. Tummuru, G. G. Miller, and M. J. Blaser.1995.Interleukin-8 response of gastric epithelial cell lines to Helicobacter
pyloristimulation in vitro. Infect. Immun. 63:16811687.

[79].Smith, A. W., B. Chahal, and G. L. French.1994. The human gastricpathogenHelicobacter pylori has a gene encoding an enzyme first
classified as a mucinase in Vibrio cholerae. Mol. Microbiol. 13:153160.

[80].Smoot, D. T., H. L. Mobley, G. R. Chippendale, J. F. Lewison, and J. H. Resau.1990. Helicobacter pylori urease activity is toxic to human
gastric epithelialcells. Infect. Immun. 58:19921994.

[81].Sobhani, I., A. Bado, Y. Cherifi, L. Moizo, J. P. Laigneau, D. Pospai, M.Mignon, and M. J. Lewin. 1996. Helicobacter pylori stimulates
gastric acidsecretion via platelet activating factor. J. Physiol. Pharmacol. 47:177185.

[82].Spiegelhalder, C., B. Gerstenecker, A. Kersten, E. Schiltz, and M. Kist.1993. Purification of Helicobacter pylori superoxide dismutase and
cloningand sequencing of the gene. Infect. Immun. 61:53155325.

[83].Suerbaum, S., A. Schmitz, C. Josenhans, and A. Labigne.1994. Cloning,sequencing, and mutagenesis of the H. pylori flbAgene: a homolog
of the lcrD/flbF/invAfamily of genes associated with motility and virulence. Am. J.Gastroenterol. 89:1331.

[84].Suerbaum, S., J. M. Thiberge, I. Kansau, R. L. Ferrero, and A. Labigne. 1994. Helicobacter pylori hspA-hspBheat-shock gene cluster:
nucleotidesequence, expression, putative function and immunogenicity. Mol. Microbiol. 14:959974.

[85].Sumii, M., K. Sumii, A. Tari, H. Kawaguchi, G. Yamamoto, Y. Takehara, Y.Fukino, T. Kamiyasu, M. Hamada, and T. Tsuda.1994.
Expression ofantral gastrin and somatostatin mRNA in Helicobacter pylori-infected subjects.Am. J. Gastroenterol. 89:15151519.

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 33 | Page


[86].Talley, N. J., A. R. Zinsmeister, A. Weaver, E. P. DiMagno, H. A. Carpenter,G. I. Perez-Perez, and M. J. Blaser.1991. Gastric
adenocarcinoma andHelicobacterpyloriinfection. J. Natl. Cancer Inst. 83:17341739.

[88].Alm, R.A., Ling, L.S., Moir, D.T., King, B.L., Brown, E.D.,Doig, P.C., et al. [1999] Genomic-sequence comparison oftwo unrelated isolates
of the human gastric pathogen Helicobacterpylori. Nature 397: 176180.

[89].Alm, R.A., Bina, J., Andrews, B.M., Doig, P., Hancock, R.E.,and Trust, T.J. [2000] Comparative genomics of Helicobacterpylori: analysis
of the outer membrane proteinfamilies. Infect Immun68: 41554168.

[90].Aspholm-Hurtig, M., Dailide, G., Lahmann, M., Kalia, A., Ilver,D., Roche, N., et al. [2004] Functional adaptation of BabA,theH. pylori
ABO blood group antigen binding adhesin.Science305: 519522.

[91].Aspholm, M., Olfat, F.O., Nordn, J., Sondn, B., Lundberg,C., Sjstrm, R., et al. [2006a] SabA is the H. pylorihemagglutinin and is
polymorphic in binding to sialylatedglycans. PLoSPathog2: 9891001.

[92].Aspholm, M., Kalia, A., Ruhl, S., Schedin, S., Arnqvist, A.,Lindn, S., et al. [2006b] Helicobacter pylori adhesion tocarbohydrates.Methods
Enzymol417: 293339.

[93].Backert, S., Moese, S., Selbach, M., Brinkmann, V., andMeyer, T.F. [2001] Phosphorylation of tyrosine 972 of theHelicobacter pylori CagA
protein is essential for inductionof a scattering phenotype in gastric epithelial cells. MolMicrobiol42: 631644.

[94].Backert, S., Kwok, T., Schmid, M., Selbach, M., Moese, S.,Peek, R., et al. [2005] Subproteomes of soluble andstructure-bound Helicobacter
pylori proteins analyzedby two-dimensional gel electrophoresis and massspectrometry.Proteomics5: 13311345.
[95].Bckstrm, A., Lundberg, C., Kersulyte, D., Berg, D., Born,T., and Arnqvist, A. [2004] Metastability of Helicobacterpyloribabadhesin
genes and dynamics in Lewis b antigenbinding. ProcNatlAcadSci USA 101: 1692316928.

[97].Baltrus, D.A., Amieva, M.R., Covacci, A., Lowe, T.M., Merrell,D.S., Ottemann, K.M., et al. [2009] The complete genomesequence of
Helicobacter pylori strain G27. J Bacteriol191: 447448.

[98].Beveridge, T.J. [1999] Structures of gram-negative cell wallsand their derived membrane vesicles. J Bacteriol181:47254733.

[99].Bomberger, J., Maceachran, D., Coutermarsh, B., Ye, S.,Otoole, G., and Stanton, B. [2009] Long-distance deliveryof bacterial virulence
factors by Pseudomonas aeruginosaouter membrane vesicles. PLoSPathog5: e1000382.

[100].Born, T., Falk, P., Roth, K.A., Larson, G., and Normark, S.[1993] Attachment of Helicobacter pylori to human gastricepithelium mediated
by blood group antigens. Science262: 18921895.

[101]. Olofsson et al. _ 2010 Blackwell Publishing Ltd, Molecular Microbiology, 77, 15391555.

[103].Deatherage, B.L., Lara, J.C., Bergsbaken, T., RassoulianBarrett, S.L., Lara, S., and Cookson, B.T. [2009] Biogenesisof bacterial membrane
vesicles.MolMicrobiol72:13951407.

[104].Fiocca, R., Necchi, V., Sommi, P., Ricci, V., Telford, J., Cover,T.L., and Solcia, E. [1999] Release of Helicobacter
pylorivacuolatingcytotoxin by both a specific secretion pathwayand budding of outer membrane vesicles. Uptake ofreleased toxin and vesicles by
gastric epithelium.J Pathol188: 220226.

[105].Fischer, W., Buhrdorf, R., Gerland, E., and Haas, R. [2001] .Outer membrane targeting of passenger proteins by thevacuolatingcytotoxin
autotransporter of Helicobacterpylori.InfectImmun69: 67696775.

[106].Gerhard, M., Hirmo, S., Wadstrm, T., Miller-Podraza, H.,Teneberg, S., Karlsson, K.-A., et al. [2001] Helicobacterpylori: Molecular and
Cellular Biology. In Helicobacterpylori: An Adherent Pain in the Stomach. Achtman, M, andSuerbaum, S. [eds]. Wymondham: Horizon
ScientificPress, pp. 185206.

[107.Hamilton, S., Hamilton, R.J., and Sewell, P. [1992] Extractionof Lipids and Derivative Formation. Oxford: IRL Press.Hatakeyama, M.
[2006] Helicobacter pylori CagA a bacterialintruder conspiring gastric carcinogenesis.Int J Cancer119: 12171223.

[108].Hatakeyama, M. [2008] SagA of CagA in Helicobacter pyloripathogenesis.CurrOpinMicrobiol11: 3037.Heczko, U., Smith, V.C.,
Meloche, R.M., Buchan, A.M., andFinlay, B.B. [2000] Characteristics of Helicobacter pyloriattachment to human primary antral epithelial
cells.Microbes Infect 2: 16691676.

[109].Hessey, S.J., Spencer, J., Wyatt, J.I., Sobala, G., Rathbone,B.J., Axon, A.T., and Dixon, M.F. [1990] Bacterial adhesionand disease activity
in Helicobacter associated chronicgastritis. Gut 31: 134138.

[110].Higashi, H., Tsutsumi, R., Muto, S., Sugiyama, T., Azuma, T.,Asaka, M., and Hatakeyama, M. [2002] SHP-2 tyrosine phosphatase as an
intracellular target of Helicobacterpylori CagAprotein.Science295: 683686.
IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 34 | Page



[111].Hofreuter, D., Odenbreit, S., and Haas, R. [2001] Naturaltransformation competence in Helicobacter pylori is mediatedby the basic
components of a type IV secretionsystem. MolMicrobiol41: 379391.

[112].Horstman, A.L., and Kuehn, M.J. [2000] EnterotoxigenicEscherichia coli secretes active heat-labile enterotoxin viaouter membrane
vesicles. J BiolChem275: 1248912496.

[113].Huang, K., Mukhopadhyay, R., and Wingreen, N. [2006] Acurvature-mediated mechanism for localization of lipids tobacterial
poles.PLoSComputBiol2: 13571364.

[114].Ilver, D., Arnqvist, A., Ogren, J., Frick, I.M., Kersulyte, D.,Incecik, E.T., et al. [1998] Helicobacter pylori adhesion binding
fucosylatedhisto-blood group antigens revealed byretagging. Science 279: 373377.

[115].Inamoto, Y., Ariyama, S., Hamanaka, Y., Okita, K., Kaneta, Y.,Nagate, T., et al. [1993] Lipid analysis of
Helicobacterpylori.JClinGastroenterol17: 136139.

[116].Kadurugamuwa, J.L., and Beveridge, T.J. [1998] Delivery ofthe non-membrane-permeative antibiotic gentamicin intomammalian cells by
using Shigellaflexnerimembranevesicles. Antimicrob Agents Chemother42: 14761483.

[121.Kobayashi, M., Mitoma, J., Nakamura, N., Katsuyama, T.,Nakayama, J., and Fukuda, M. [2004] Induction of peripherallymph node
addressin in human gastric mucosainfected by Helicobacter pylori.ProcNatlAcadSci USA101: 1780717812.

[122].Kouokam, J.C., Wai, S.N., Fallman, M., Dobrindt, U., Hacker,J., and Uhlin, B.E. [2006] Active cytotoxic necrotizing factor1 associated
with outer membrane vesicles from uropathogenicEscherichia coli. Infect Immun74: 20222030.
[123].Kudo, T., Nurgalieva, Z.Z., Conner, M.E., Crawford, S., Odenbreit,S., Haas, R., et al. [2004] Correlation between Helicobacterpylori OipA
protein expression and oipAgeneswitch status.J ClinMicrobiol42: 22792281.

[124].Kuehn, M., and Kesty, N. [2005] Bacterial outer membranevesicles and the host-pathogen interaction.GenesDev19:26452655.

[125].Kutter, S., Buhrdorf, R., Haas, J., Schneider-Brachert, W.,Haas, R., and Fischer, W. [2008] Protein subassemblies oftheHelicobacter pylori
Cag type IV secretion systemrevealed by localization and interaction studies.J Bacteriol190: 21612171.

[126.Kwok, T., Zabler, D., Urman, S., Rohde, M., Hartig, R.,Wessler, S., et al. [2007] Helicobacter exploits integrin fortype IV secretion and
kinase activation. Nature 449: 862866.

[127]--Lee, E., Bang, J., Park, G., Choi, D., Kang, J., Kim, H., et al.[2007] Global proteomic profiling of native outer membranevesicles derived
from Escherichia coli. Proteomics 7:31433153.

[128].Lee, E., Choi, D., Kim, K., and Gho, Y. [2008] Proteomics ingram-negative bacterial outer membrane vesicles. MassSpectrom Rev 27:
535555.

[129.Li, Z., Clarke, A.J., and Beveridge, T.J. [1996] A major autolysinof Pseudomonas aeruginosa: subcellular distribution,potential role in cell
growth and division and secretion insurface membrane vesicles. J Bacteriol178: 24792488.

[130].Lwer, M., Weydig, C., Metzler, D., Reuter, A., Starzinski-Powitz, A., Wessler, S., and Schneider, G. [2008] Predictionof extracellular
proteases of the human pathogenHelicobacter pylori reveals proteolytic activity of theHp1018/19 protein HtrA. PLoS ONE 3: e3510.

[131.Lundmark, R., and Carlsson, S.R. [2003] Sorting nexin 9H. pylori vesicles and properties for adhesion 1553 2010 Blackwell Publishing
Ltd, Molecular Microbiology, 77, 15391555

[132].participates in clathrin-mediated endocytosis through interactionswith the core components. J BiolChem278:4677246781.

[133.Mahdavi, J., Sondn, B., Hurtig, M., Olfat, F., Forsberg, L.,Roche, N., et al. [2002] Helicobacter pylori SabAadhesion in persistent infection
and chronic inflammation.Science297: 573578.

[135].Meneses, P., and Glonek, T. [1988] High resolution 31P NMRof extracted phospholipids.J Lipid Res 29: 679689.

[136.Namork, E., and Brandtzaeg, P. [2002] Fatal meningococcalsepticaemia with blebbing meningococcus. Lancet 360:1741.

[137].Nevot, M., Deroncel, V., Messner, P., Guinea, J., andMercad, E. [2006] Characterization of outer membranevesicles released by the
psychrotolerant bacteriumPseudoalteromonasantarcticaNF3. Environ Microbiol8:15231533.

[138].Nguyen, T.T., Saxena, A., and Beveridge, T.J. [2003] Effect ofsurface lipopolysaccharide on the nature of membranevesicles liberated from
the Gram-negative bacteriumPseudomonas aeruginosa. J Electron Microsc [Tokyo] 52:465469.

IJASR International Journal of Academic Scientific Research
ISSN: 2272-6446 Volume 2, Issue 1 (February-March 2014), PP 21-35

www.ijasrjournal.org 35 | Page


[139].Odenbreit, S., Till, M., and Haas, R. [1996] Optimized BlaMtransposonshuttle mutagenesis of Helicobacter pyloriallows the identification
of novel genetic loci involved inbacterial virulence. MolMicrobiol20: 361373.

[140].Odenbreit, S., Pls, J., Sedlmaier, B., Gerland, E., Fischer,W., and Haas, R. [2000] Translocation of Helicobacterpylori CagA into gastric
epithelial cells by type IV secretion.Science287: 14971500.

[141].Odenbreit, S., Faller, G., and Haas, R. [2002a] Role of thealpAB proteins and lipopolysaccharide in adhesion of Helicobacterpylori to
human gastric tissue.Int J Med Microbiol292: 247256.

[142].Odenbreit, S., Kavermann, H., Puls, J., and Haas, R. [2002b]CagA tyrosine phosphorylation and interleukin-8 inductionbyHelicobacter
pylori are independent from AlpAB, HopZand Bab group outer membrane proteins. Int J Med Microbiol292: 257266.

[143].Oh, J.D., Karam, S.M., and Gordon, J. [2005] IntracellularHelicobacter pylori in gastric epithelial progenitors. ProcNatlAcadSci USA 102:
51865191.

[144].Oh, J.D., Kling-Backhed, H., Giannakis, M., Xu, J., Fulton,R.S., Fulton, L.A., et al. [2006] The complete genomesequence of a chronic
atrophic gastritis Helicobacter pyloristrain: evolution during disease progression. ProcNatlAcadSci USA 103: 999910004.

[145].Pettit, R.K., and Judd, R.C. [1992] The interaction of naturallyelaborated blebs from serum-susceptible and serumresistantstrains of
Neisseria gonorrhoeaewith normalhuman serum. MolMicrobiol6: 729734.

[146.Petzold, K., Olofsson, A., Arnqvist, A., Grbner, G., andSchleucher, J. [2009] Semiconstant-time P,H-COSY NMR:analysis of complex
mixtures of phospholipids originatingfromHelicobacter pylori. J Am ChemSoc131: 1415014151.

[149].Sattler, M., Schleucher, J., and Griesinger, C. [1999] Heteronuclearmultidimensional NMR experiments for the structuredetermination of
proteins in solution employing pulsedfield gradients. ProgrNuclMagnResonSpectrosc34:93158.
[ 150].Schmees, C., Prinz, C., Treptau, T., Rad, R., Hengst, L.,Voland, P., et al. [2007] Inhibition of T-cell proliferation byHelicobacter pylori
gamma-glutamyltranspeptidase.Gastroenterology132: 18201833.

[151].Segal, E.D., Cha, J., Lo, J., Falkow, S., and Tompkins, L.S.[1999] Altered states: involvement of phosphorylatedCagA in the induction of
host cellular growth changes byHelicobacter pylori. ProcNatlAcadSci USA 96: 1455914564.

[152].Semino-Mora, C., Doi, S.Q., Marty, A., Simko, V., Carlstedt,I., and Dubois, A. [2003] Intracellular and interstitialexpression of
Helicobacter pylori virulence genes in gastricprecancerous intestinal metaplasia and adenocarcinoma.J Infect Dis 187: 11651177.

[153].Shevchenko, A., Wilm, M., Vorm, O., and Mann, M. [1996]Mass spectrometric sequencing of proteins silver-stainedpolyacrylamide gels.
Anal Chem68: 850858.

[154].Shimomura, H., Hayashi, S., Yokota, K., Oguma, K., andHirai, Y. [2004] Alteration in the composition of cholesterylglucosides and other
lipids in Helicobacter pylori undergoingmorphological change from spiral to coccoidform.FEMSMicrobiolLett237: 407413.

[155].Suzuki, M., Mimuro, H., Kiga, K., Fukumatsu, M., Ishijima, N.,Morikawa, H., et al. [2009] Helicobacter pylori CagAphosphorylation-
independent function in epithelial proliferationand inflammation. Cell Host Microbe 5: 2334.

[157].Tannaes, T., Grav, H.J., and Bukholm, G. [2000] Lipid profilesof Helicobacter pylori colony variants. APMIS 108: 349356.

[158.Tannaes, T., Dekker, N., Bukholm, G., Bijlsma, J.J., andAppelmelk, B.J. [2001] Phase variation in the Helicobacterpyloriphospholipase A
gene and its role in acid adaptation.InfectImmun69: 73347340.

[159].Tannaes, T., Bukholm, I.K., and Bukholm, G. [2005] Highrelative content of lysophospholipids of Helicobacter pylorimediates increased
risk for ulcer disease. FEMS ImmunolMedMicrobiol44: 1723

[162].Yamaoka, Y., and Alm, R.A. [2008] Helicobacter pylori OuterMembrane Proteins. In Helicobacter pylori: MolecularGenetics and Cellular
Biology. Yamaoka, Y. [ed.]. Norfolk,UK: Caister Academic Press, pp. 3760.

[163].Yamaoka, Y., Kwon, D.H., and Graham, D.Y. [2000] A M[r]34,000 proinflammatory outer membrane protein [oipA]
ofHelicobacterpylori.ProcNatlAcadSci USA 97: 75337538.

Vous aimerez peut-être aussi