Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Medical Microbiology 300 (2010) 387–395

Contents lists available at ScienceDirect

International Journal of Medical Microbiology


journal homepage: www.elsevier.de/ijmm

Mini Review

The emergence of ‘hypervirulence’ in Clostridium difficile


Stephen T. Cartman, John T. Heap, Sarah A. Kuehne, Alan Cockayne, Nigel P. Minton ∗
Centre for Biomolecular Sciences, School of Molecular Medical Sciences, Nottingham Digestive Diseases Centre NIHR Biomedical Research Unit, University of Nottingham,
University Park, Nottingham, NG7 2RD, UK

a r t i c l e i n f o a b s t r a c t

Keywords: The impact of Clostridium difficile-associated disease (CDAD) in healthcare settings throughout the devel-
Clostridium difficile oped world is considerable in terms of mortality, morbidity, and disease management. The incidence
Hypervirulence of CDAD has risen dramatically since the turn of this century, concomitant with the emergence of so-
Virulence factors
called hypervirulent strains which are thought to cause a more severe disease, higher relapse rates, and
ClosTron
increased mortality. Pre-eminent amongst hypervirulent strains are those belonging to ribotype 027,
Mutant generation
which were first reported in Canada in 2003 and shortly thereafter in the UK. Since its arrival in Europe,
it has spread rapidly and has now been reported in 16 member states and Switzerland. The physiolog-
ical factors responsible for the rapid emergence of hypervirulent C. difficile strains remain unclear. It is
known that they produce a binary toxin (CDT) in addition to toxins A and B, that they are resistant to
fluoroquinolones due to mutations in gyrA, and that they are resistant to erythromycin. Representative
strains have been suggested to produce more toxin A and B in the ‘laboratory flask’ (most likely due to a
frameshift mutation in the repressor gene tcdC), to be more prolific in terms of spore formation, and also
exhibit increased adherence to human intestinal epithelial cells due to altered surface proteins. However,
the contribution of these and other as yet unidentified factors to the rapid spread of certain C. difficile vari-
ants (e.g., ribotypes 027 and 078) remains unclear at present. The advent of ClosTron technology means
that it is now possible to construct genetically stable isogenic mutants of C. difficile and carry out reverse
genetic studies to elucidate the role of specific gene loci in causing disease. The identification of virulence
factors using this approach should help lead to the rational development of therapeutic countermeasures
against CDAD.
© 2010 Elsevier GmbH. All rights reserved.

1. Introduction bad name. Over 90% of clinical disease is caused by just 12 species,
and it is these organisms that tend to make the headlines. Thus,
All members of the genus Clostridium are obligate anaerobic bac- necrotic enteritis due to C. perfringens is causing devastation to
teria that are unified by their ability to form endospores, the most intensively reared fowl following the EU-directed withdrawal of
highly resistant life forms on earth. They are one of the largest bac- antibiotic growth promoters from feedstuffs. Post 9/11, there are
terial genera. More than 300 species have been described, although heightened public concerns over the use of C. botulinum by bioter-
currently less than 100 are recognised at the species level. The vast rorists, but the largest and most strident headlines are currently
majority of the genus is entirely benign, undertaking all manner reserved for C. difficile.
of useful biotransformations, including the production of biofuels.
Thus, concerns over global warming and escalating crude oil costs 2. Clostridium difficile-associated disease (CDAD)
dictate the reintroduction of the commercial production of bio-
fuels such as butanol by solventogenic clostridia from renewable Toxinogenic strains of C. difficile are carried by 7–11% of hospi-
biomass, e.g., C. acetobutylicum. Moreover, experimental advances talised patients (Poutanen and Simor, 2004). The reservoirs of C.
have now demonstrated that clostridia have a significant role in difficile include contaminated environments and surfaces within
the treatment of cancer, by capitalising on the unique ability of healthcare facilities and infected patients. Colonized individuals
spores to selectively germinate in tumours (e.g., C. sporogenes may become asymptomatic carriers or manifest symptoms, depen-
and C. novyi). However, despite the beneficial properties of many dent on their status. Healthy adults are generally resistant, a
clostridial species, it is the antics of a few that give the genus a consequence of the protective effects of their colonic flora. Per-
turbation of this flora, most often following exposure to broad
spectrum antibiotics (such as penicillins, cephalosporins, and clin-
∗ Corresponding author. Tel.: +44 115 846 7458; fax: +44 115 823 2120. damycin), leads to loss of colonisation resistance, enabling C. difficile
E-mail address: nigel.minton@nottingham.ac.uk (N.P. Minton). to proliferate and cause disease symptoms. Other risk factors

1438-4221/$ – see front matter © 2010 Elsevier GmbH. All rights reserved.
doi:10.1016/j.ijmm.2010.04.008
388 S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395

include various physical interventions (surgery, nasogastric tubes, cus faecium, and it lives in the same niche as C. difficile, transfer of
and enemas), antiperistaltic drugs and antacids, and exposure to vanA to C. difficile would appear to be just a matter of time. The
chemotherapy or immunosuppressive agents (Poutanen and Simor, emergence of metronidazole-resistant strains carrying vanA would
2004). The symptoms of those individuals that develop the disease have catastrophic consequences.
range from a mild, self-limiting diarrhoea to the potentially fatal Other approaches are being investigated (Critchley et al., 2009;
pseudomembranous colitis (PMC) (Mylonakis et al., 2001). Mortal- Hedge et al., 2008; Rohde et al., 2009), including the use of
ity rates can be as high as 30%. alternative antimicrobial agents, toxin-binding agents, immune-
Elderly patients are especially at risk of C. difficile-associated modifying agents, probiotics, phage-based approaches, and faecal
disease (CDAD). Although the carriage rate in neonates is higher replacement therapy. As yet, none have achieved wide acceptance.
(over 60%), they rarely develop disease, most likely a conse- As such, vaccination still remains an option. Current vaccine devel-
quence either of an inability of toxins to attach to the neonatal opment approaches have been based on a limited understanding
mucosa or through protection from maternally-acquired anti- of the detailed mechanisms of disease causation and consequently
bodies against the toxins. Neonatal carriage rates fall to levels have focussed primarily on the generation of antibody responses
equivalent to adults by the age of 3 years and upwards (Kelly et to neutralise the effects of toxins A and B which have histori-
al., 1994; Poutanen and Simor, 2004). Although generally regarded cally been considered key virulence determinants in C. difficile. A
as a healthcare-associated disease, the occurrence of community- vaccine consisting of toxoids prepared from toxins A and B has
associated disease is increasingly being reported, e.g., 9.3% of 703 already undergone Phase 1 clinical trials (Acambis, 2007). Evidence
patients with diarrhoea visiting German general practitioners over to support the potential of this approach comes from early exper-
a 5-month period were diagnosed with CDAD (Weil et al., 2007). C. imental vaccination studies using purified toxoids of toxins A and
difficile is now also recognised as an important cause of enteric dis- B or crude culture filtrates which showed that systemic anti-toxin
ease in animals (Songer and Anderson, 2005; Clooten et al., 2008). IgG responses are important in protecting hamsters from lethal C.
Until recently, it had been generally regarded that strains present difficile challenge and in preventing gut pathology (Giannasca et
in animals were distinct from those causing human infections. This al., 1999). Anti-TcdA responses have also been shown to protect
notion has now been modified somewhat by Debast et al. (2009) hamsters from C. difficile challenge (Kim et al., 1987), although a
who found that strains of PCR ribotype 078 (toxinotype V) isolated recent study with isogenic tcdA mutants showed that TcdA is not
from pigs with diarrhoea were identical to strains isolated from an essential virulence determinant (Lyras et al., 2009). This obser-
cases of human CDAD. vation supports findings that clinical isolates of C. difficile which
The impact of CDAD in healthcare settings is considerable. are unable to produce TcdA still cause disease in humans (Drudy
Patients require isolation, revised supportive therapy for under- et al., 2007). It is clear that the exact role played by TcdA in clinical
lying disease and for CDAD, specific therapy to eliminate C. difficile, disease requires further clarification. Other hamster immunisation
scrupulous hygiene in nursing, environmental decontamination, studies have shown varying levels of protection from lethality and
and (in outbreaks) ward closure. The financial impact of CDAD diarrhoea following vaccination with TcdB toxoids (Giannasca et
on the healthcare system is substantial and has recently been al., 1999; Torres et al., 1995). However, anti-toxin responses alone
estimated to cost at least $2 billion dollars in Europe (European do not appear to result in elimination of C. difficile from the gut, and
Centre for Disease Prevention and Control. ECDC Summary of CDAD vaccinated animals still remain colonised (Kim et al., 1987; Torres
infection, 2005–2007) and $3.2 billion dollars per year in the US et al., 1995) and potentially infectious. In addition, observed varia-
(O’Brien et al., 2007). As the disease is principally a disease of the tion in the sequence of tcdB among C. difficile isolates (Stabler et al.,
elderly, these costs can be expected to almost double over the next 2008) raises the possibility that a vaccine based on TcdB alone may
four decades in line with an increasing elderly population. ultimately encounter problems with the breadth of cover elicited.

3. Current therapeutic and management options 4. Increased incidence of CDAD

Given the recognised role of antibiotic therapy in precipitating Once regarded as relatively uncommon, there has been a
CDAD, mild disease may be treated by withdrawing antibiotics and steadily increasing upward trend in the incidence of CDAD. Analy-
providing additional supportive therapy (Hedge et al., 2008). The sis of infection rates in US hospitals from 1986 to 2001 found that
organism has long been recognised as multiply antibiotic resistant the incidence had significantly increased (incidence rate approxi-
(e.g., erythromycin, clindamycin, lincomycin, tetracycline, chlor- mately 5.5 per 10,000 patient-days) in hospital intensive care units
amphenicol), principally due to the presence of a large proportion (ICU) with >500 beds (Archibald et al., 2004). The upward trend was
of mobile elements in the genome carrying antibiotic resistance confirmed in 2 subsequent studies, where rates were perceived to
genes (Sebaihia et al., 2006). As a consequence, just 2 antibiotics are have doubled. Thus, McDonald et al. (2006) reported a doubling
available for treatment, metronidazole and vancomycin, which in of the rate (as any diagnosis) between 1996 and 2003 (from 31
combination with improved hygiene and patient isolation currently to 61 per 100,000 population) and Elixhauser and Jhung (2008)
remain the main therapeutic option. found 76.9 episodes of CDAD per 10,000 discharges in 2005, a rate
Whilst the latter has fewer side-effects and is more effective, more than twice of that in 1999 (37.6 per 10,000 discharges). More
metronidazole has until now been preferred due to its lower price recently, a survey of 12.5% of all US acute-care facilities indicated
and concerns over using a last resort antibiotic such as vancomycin. that there had been no decrease in rates reported by 82% of respon-
However, metronidazole is becoming increasingly ineffective and dents over the preceding 3 years; the overall C difficile prevalence
reports on the isolation of less susceptible strains are beginning rate was 13.1 per 1000 inpatients (Jarvis et al., 2009). Broadly sim-
to appear (Brazier et al., 2001, 2008; Peláez et al., 2005; Baines ilar infection rates were also noted in Canada (Gravel et al., 2009),
et al., 2008). This has led to moves towards adopting vancomycin where the overall incidence rate for adult patients admitted to the
as the front-line antibiotic. This is extremely worrying, as there hospitals surveyed was 4.6 cases per 1000 patient admissions and
are numerous examples of gene transfer between Enterococcus and 65 per 100,000 patient-days.
Clostridium, either demonstrated or inferred by isolation of con- Nowhere is the rise in CDAD rates more visible than in the
jugative transposons common to both genera (Roberts et al., 2001; UK. Accordingly, there has been a significant documented increase
Stinear et al., 2001; Agersø et al., 2006; Launay et al., 2006). As the through the 1990s and 2000s, rising from 1100 cases in 1990
major source of the vancomycin resistance gene vanA is Enterococ- peaking at 55,635 in 2006 (equivalent to 2.45 infections per 1000
S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395 389

of 66 different PCR ribotypes were characterised, with ribotype 027


accounting for only 6.2% of isolates (Barbut et al., 2007).
Whilst the proportion of 027 isolates in the Barbut et al. (2007)
study is low, it is an average taken across 14 countries and may
not accurately reflect the present day situation. The speed with
which ribotype 027 can become predominant is amply demon-
strated by events in the UK. Laboratory reports amassed over the
period 1990–2003 had indicated that the predominant ribotype
was 001 (55%), with ribotype 106 a distant second (Brazier, 2005).
Over the period 2005–2007, 3 ribotypes had come to predominate
in approximately equal proportions (Brazier et al., 2007). These
were ribotypes 106 (26.2%), 027 (25.9%), and ribotype 001 (25.1%).
More recently, 677 isolates were obtained from 186 hospitals in
the 9 geographical regions of England over the period April 2007 to
Fig. 1. C. difficile incidence figures for England and Wales. Figures from The Health
Protection Agency.
the end of March 2008. Typing revealed that PCR ribotype 027 was
now the most common strain isolated from symptomatic patients,
bed-days) (Fig. 1). Not unexpectedly, there has been a concomi- accounting for over 41.3% of all isolates. Ribotype 106 was the sec-
tant increase in the number of deaths attributable to CDAD. This ond most common strain (20.2%), and Ribotype 001 had reduced to
trend has continued into 2007, where despite the decrease in only 7.8% of the total.
reported incidence (from 55,635 to 50,392), there was a 28% Typically, 027 strains possess a binary toxin gene and encode
increase in C. difficile-associated deaths in 2007 compared to 2006. a variant tcdC repressor gene (with an 18-bp deletion and a
This equates to over 5 times as many deaths (8324) in England frameshift mutation due to a single base pair deletion at position
and Wales than MRSA (1593). Similar trends have been noted 117 relative to the ATG start codon). The alterations have been
elsewhere in Europe (Borgmann et al., 2008; Suetens, 2008). For suggested to account for the observed increase in toxin produc-
example, in Spain (Asensio et al., 2008), the prevalence rates of tion when cultured in the laboratory (Warny et al., 2005). Detailed
CDAD increased from 3.9 to 12.2 cases per 10,000 hospitalised molecular analysis has shown that this variant TcdC repressor is
patients and showed a significantly increasing secular trend from indeed defective, but as a consequence of the frameshift and not the
1999 through 2007 (prevalence rate ratio per each year increment 18-bp deletion (Dupuy et al., 2008). Other data, presented at the 2nd
1.09; 95% CI 1.05–1.14). International Meeting on C. difficile (June 2007), has demonstrated
In the UK at least, various reasons have been suggested for that 027 strains possess altered surface proteins and increased
this extraordinary rise in incidence and mortality. These include adherence to human intestinal epithelial cells which may also con-
the introduction of mandatory reporting in the UK from 2004, tribute to enhanced virulence. A further contributing factor to the
the increasing age of the population and therefore the number at emergence of 027 strains may have been acquisition of resistance
risk, increased antibiotic resistance, lower standards of hygiene in to fluoroquinolones and erythromycin antibiotics due to chromoso-
healthcare facilities, overcrowding in hospitals, and the emergence mal mutations in gyrA/B and the 23s RNA genes, respectively (Pépin
of so-called hypervirulent strains. et al., 2005; Spigaglia et al., 2008; Schmidt et al., 2007). Thus, the
first 027 strain to be isolated in France during 1988 was susceptible
5. Hypervirulent strains to both antibiotics.
It should be noted that whilst ribotype 027 strains have received
In the view of many, the emergence of more highly virulent all the attention, other strains may present an equivalent threat
strains has contributed to the problem of increased CDAD inci- in terms of disease severity. In many countries, a different ribo-
dence (Kuijper et al., 2007; Pépin et al., 2004). The most prominent type can predominate, but be extremely rare elsewhere. Thus,
of these are strains which belong to ribotype 027, but are also the second most common ribotype in the UK, ribotype 106, was
characterised as toxinotype III, North American pulsed field gel entirely absent from the European study of Barbut et al. (2007). In
electrophoresis type 1 (NAP1) and restriction endonuclease anal- The Netherlands, ribotype 078 has significantly increased over the
ysis group BI. The occurrence of these strains is associated with period February 2005 to February 2008, from 3% to 13%. Patients
an excessive use of quinolone antibiotics and in North Amer- infected with ribotype 078 were younger (67.4 vs. 73.5 years;
ica has been responsible for a 5-fold increase in the historical p < 0.01) than those infected with 027 and more frequently had
average of CDAD, more severe disease (complications increased community-associated disease (17.5% vs. 6.7%; odds ratio 2.98; 95%
from 7.1% to 18.2%), higher relapse rates (from 20.8% to 47.2%), CI 2.11–8.02). The rates of severe diarrhoea and attributable mor-
increased mortality (from 4.7% to 13.8%), and greater resistance tality were essentially the same for both ribotypes. Ribotype 078
to fluoroquinolone antibiotics (Kuijper et al., 2006). Since 2005, isolates possess a number of features in common with ribotype
individual countries have developed surveillance studies to mon- 027 strains. They both harbour genes encoding toxin A and toxin
itor the spread of ribotype 027. In the United States, cases of C. B and the genes for binary toxin. They also possess a variant tcdC
difficile ribotype 027 infection have been reported from at least gene, although the deletion and frameshift are different for the 2
38 states (http://www.cdc.gov/ncidod/dhqp/id Cdiff.html). It has ribotypes (a 39-bp deletion and a single base pair deletion at posi-
been reported in 16 European countries (Kuijper et al., 2008). Ribo- tion 184 in the case of ribotype 078). Significantly, the ribotype
type 027 isolates have been responsible for outbreaks in Belgium, 078 strains isolated from humans have recently been shown to be
Germany, Finland, France, Ireland, Luxembourg, The Netherlands, genetically related to isolates from pigs (Goorhuis et al., 2008).
Switzerland, and the United Kingdom (England, Wales, Northern
Ireland, and Scotland). They have also been detected in Austria,
Denmark, Sweden, Norway, Hungary, Poland, and Spain. Three 6. Virulence factors of C. difficile
countries experienced imported patients with CDAD due to ribo-
type 027 who acquired the infection abroad (Kuijper et al., 2007). The emergence of dominant strains that cause more severe dis-
During May and June of 2005, a prospective CDAD study was con- ease is suggestive of one or more changes in bacterial encoded
ducted in 38 hospitals from 14 different European countries. A total determinants that contribute to the disease process. Despite
390 S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395

its medical importance, the pathogenesis of C. difficile infection of strain 630 and the various partial sequences available
remains poorly understood. Attention has largely focussed on the (http://www.sanger.ac.uk). However, definitive proof of their
2 large toxins, A and B. Initially, toxin A was widely acknowledged involvement in disease has yet to be obtained. It is only through
as the principle factor responsible for disease. However, the sub- mutation that the function of any particular gene can be definitively
sequent recognition that a significant number of strains do not ascribed. However, despite the best efforts of numerous laborato-
produce toxin A, yet remain highly pathogenic suggested that toxin ries around the globe, in the preceding decades there have been
A could have little if any role to play in disease. This interpretation relatively few directed mutants generated in the genus Clostrid-
has been supported by the study of Lyras et al. (2009) which demon- ium. Those that have been generated have almost exclusively been
strated that inactivation of the tcdA gene through the creation of an generated using classical recombination-based methods by select-
unstable, single cross-over mutant, had little effect on the virulence ing for the integration of either replication defective or deficient
of C. difficile. However, it should be noted that a proportionately plasmid on the basis of acquisition of an antibiotic resistance gene
stable mutant created in our laboratory using the ClosTron yield (Heap et al., 2008). The larger proportion of these mutants has been
contradictory results. Thus, a tcdB mutant producing only TcdA based on the mutagenic integration of the entire plasmid by a single
proved lethal in the hamster model (Sarah Kuehne, Steve Cart- cross-over event and has therefore been unstable. In C. difficile, the
man, John Heap, Michelle Kelly, Alan Cockayne, and Nigel P. Minton, handful of mutants generated to date have all been made through
unpublished results). integration of either a deficient (Liyanage et al., 2001) or defec-
Certain C. difficile strains, and in particular strains of ribotype tive (O’Connor et al., 2006; Dineen et al., 2007; Lyras et al., 2009)
027, also produce an actin-specific ADP-ribosyltransferase CDT, a plasmid. They are, therefore, unstable.
binary toxin encoded by cdtA-cdtB. Whilst not absolutely required
for virulence, CDT may enhance virulence. Other factors undoubt-
edly contribute to virulence, particularly the initial colonisation 8. Group II introns
process. Some putative virulence factors that could play a role in
adherence and intestinal colonization have already been identi- An alternative approach to recombination is to utilise group
fied. These include the S-layer proteins (Cerquetti et al., 2000), the II introns. These interrupt protein coding or RNA genes and are
cell wall protein Cwp66 (Waligora et al., 2001), GroEL (Hennequin widespread in both eukaryotic organelle and prokaryotic genomes.
et al., 2001), Fbp68 fibronectin-binding protein (Hennequin et al., Group II introns rely on the activity of an element-encoded
2003), and FliC-FliD, components of flagella (Tasteyre et al., 2001). multifunctional intron-encoded protein (IEP) to bring about the
In addition to mediating the attachment of bacteria to host tis- self-catalytic splicing of the intron from RNA. One of the best stud-
sues, adhesins may be biological effectors in vivo that influence ied elements is the ‘Ll.LtrB intron’ found in the ltrB gene (and
the outcome of the host–pathogen interactions. Thus, C. diffi- its cognate IEP, LtrA) of selected Lactococcus lactis plasmids. The
cile S-layer proteins may contribute to the pathogenicity of the details of the process by which this intron mediates its splicing
microorganism by perturbing the fine balance of inflammatory and and subsequent insertion (retro-homing) into intron-free copies
regulatory cytokines in human monocytes and monocyte-derived of ltrB have been thoroughly characterised. By determining those
dendritic cells (Ausiello et al., 2006). Cwp84, a cysteine protease, factors important in ltrB target specificity, the Lambowitz labora-
may contribute to the degradation of host tissue integrity, to the tory have been able to devise procedures whereby defined changes
dissemination of the infection and to Slp maturation (Janoir et al., to the intron sequence may be employed to target Ll.LtrB-derived
2007). introns to almost any gene of interest (Mohr et al., 2000; Perutka
Surface antigens frequently elicit a host immune response dur- et al., 2004). In the continued presence of LtrA, dependent on the
ing colonisation and infection. Indeed, levels of antibodies directed orientation of insertion, the intron is capable of secondary retro-
against the flagellar proteins and Cwp84 were significantly higher transposition events. To prevent this, in the system devised, ltrA
in a control group than in a patient group with CDAD (Péchiné et al., was moved from within Ll.LtrB to a distal plasmid-borne location.
2005a). Similarly, the S-layer proteins have shown to be immun- It follows that the selection of cells in which the plasmid-carrying
odominant in the host. This suggests that these proteins are able LtrA is lost, prevents any subsequent mobility of the intron element.
to induce an immune response that could play a role in the host The insertions generated are therefore stable. The retargeted intron
defence mechanisms. elements created have been termed ‘Targetrons’.
Blocking of the initial colonisation process, which involves adhe- Basic Targetron technology was first used to generate a plc
sion to the colonic mucosa, has been considered as an alternative mutant of C. perfringens (Chen et al., 2005) using a simple phe-
target for vaccination approaches. Analysis of serum antibody notypic plate assay to identify the desired mutant. It is, however,
responses in control and C. difficile-infected groups identified fla- disadvantaged by an inability to positively select cells in which the
gellar antigens FliC and FliD and the surface protease Cwp84 as intron element has inserted into the desired location. As integra-
potential vaccine candidates (Péchiné et al., 2005a,b). Subsequent tion frequencies can vary widely between target sites, if a simple
immunisation studies showed that recombinant FliD in combi- phenotypic screen is unavailable, then screening for the desired
nation with a crude flagella extract or Cwp84 and a cell surface mutant can be prohibitively laborious. To overcome this deficiency,
extract all reduced colonisation in the human flora-associated the Lambowitz laboratory constructed a modified Ll.LtrB intron in
mouse model though colonisation was not totally eliminated in which an antibiotic resistance gene was inserted that had been
any of the vaccinated animals (Péchiné et al., 2007). However, in a inactivated by insertion of a region of DNA encoding a group I
second study, immunisation with a crude surface layer extract and intron. In the ‘twintron’ element made, the constitutive elements
adjuvant failed to elicit significant protective immune responses were orientated relative to one another such that the group I intron
against lethal challenge in the hamster infection model (Ní Eidhin is lost (through self-catalytic splicing) during group II intron retro-
et al., 2008). transposition. As a consequence, the antibiotic gene in the inserted
group II intron becomes reactivated, allowing positive selection of
the desired mutant (Karberg et al., 2001; Zhong et al., 2003). This
7. Mutant generation new marker was termed a retrotransposition-activated selectable
marker (RAM). A widely used example is based on the kan gene,
Other potential virulence factors which present possible ther- and a plasmid carrying this element, pACD4K-C, is available from
apeutic targets are apparent in both the annotated genome Sigma Aldrich (http://www.sigma-genosys.com/targetron/.
S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395 391

9. The ClosTron

As kanamycin cannot be used as a selectable marker in Clostrid-


ium spp., we elected to construct a new element incorporating a
clostridial RAM based on the ermB gene of the Enterococcus fae-
calis plasmid pAM␤1. This new element, the ClosTron (Heap et al.,
2007), enables the group II intron integration event to be positively
selected (using erythromycin or lincomycin). Therefore, it is pos-
sible to inactivate any non-essential gene, without the need for a
phenotypic screen appropriate to the gene of interest.
Insertion of the group I intron into the ermB gene to generate an
ermB-based RAM was achieved by extending the 5 end of the ermB
gene by 12 codons. In addition, the weak native promoter of ermB
was replaced with the stronger promoter of the thlA gene of C. ace-
tobutylicum ATCC 824 (Girbal et al., 2003) to increase expression
of the new ermB element. The complete ermB RAM was incorpo-
Fig. 2. The second generation ClosTron vector pMTL007C-E2 (Heap et al., 2010).
The second generation ClosTron plasmid pMTL007C-E2 uses the strong fdx pro- rated into a group II intron cassette within an E. coli/clostridial
moter from C. sporogenes to direct expression of the Gp II intron, and contains shuttle vector, giving rise to the plasmid pMTL007 (Heap et al.,
FRT sites flanking the RAM to facilitate FLP-mediated marker removal. The plas- 2007). This novel targeted intron system was termed the ‘ClosTron’
mid also features a lacZ’ stuffer sequence that is replaced during intron retargeting, to denote that it is functionally analogous to Targetron systems,
allowing clones containing successfully retargeted plasmids to be identified by
blue/white screening, restriction analysis, or PCR. Further details may be found at
but optimised for use in clostridia. Superfluous sequence has now
www.clostron.com. been removed from the original pMTL007 vector, giving rise to the
second-generation vector pMTL007C-E2 (Fig. 2). Further details on
this improved system may be found at www.clostron.com.

Fig. 3. ClosTron mutagenesis using plasmid pMTL007C-E2. (A) The plasmid is transferred into C. difficile by conjugation. (B) Expression from the C. acetobutylicum thiolase
promoter (PthlA ) does not confer erythromycin resistance, as the Grp I intron is transcribed in the wrong orientation to splice out of the ermB transcript. (C) Expression of
the Grp II intron yields a transcript which binds to the LtrA protein to form a ribonuclear protein complex (RNP). In this case, the Grp I intron is transcribed in the correct
orientation and thus splices out of the transcript. (D) The RNP locates the desired genomic target, the Grp II intron RNA is inserted, and LtrA reverse transcribes the cDNA
strand. Host enzymes then displace the RNA strand and repair the insertion site to give a permanent dsDNA insertion. The plasmid pMTL007C-E2 is subsequently lost due to
segregational instability (not shown). (E) The ermB marker can be removed from the insertion site by introducing a plasmid expressing the FLP recombinase enzyme. (F) This
catalyses recombination between the FRT sites flanking ermB, resulting in ‘flip-out’ of the ermB gene. The plasmid expressing FLP is also lost due to segregational instability
and the ClosTron can then be used again to make further mutations (Heap et al., 2010).
392 S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395

Fig. 4. PCR screening of ClosTron mutants. (A) PCR primers P1 and P2 are designed to anneal either side of the ClosTron insertion site. Primer P3 (EBS universal primer) anneals
to Grp II intron sequence. PCR screening is carried out using primer pairs P1 and P2 (B), and P2 and P3 (C). Lane M, 1 kb plus DNA ladder (Invitrogen); lane 1, H2 O-negative
control; lane 2, genomic DNA from parent C. difficile strain; lane 3, plasmid pMTL007C-E2; lane 4, erythromycin-resistant C. difficile mutant.

In ‘proof-of-principle’ studies, the ClosTron was used to inser- cell during this time is illustrated in Fig. 3. Erythromycin-resistant
tionally inactivate a total of 19 clostridial genes (Heap et al., 2007), clones are screened for insertion of the ClosTron by PCR (Fig. 4).
including 5 in the C. difficile strain 630erm (Hussain et al., 2005). Typically, 25–100% of the clones are found to have inserted in the
Typically, 1000s of erythromycin-resistant colonies were obtained correct gene. Clones confirmed to be correct by PCR are screened
in each individual experiment with C. difficile, with the majority for thiamphenicol sensitivity to confirm loss of the ClosTron plas-
of the integration events taking place at the intended target site mid (and thus loss of the ltrA gene). Both plasmids, pMTL007
(Heap et al., 2007). Predictably, no evidence was obtained for mul- and pMTL007C-E2, are based on the replicon of pCB102 (Minton
tiple insertion of the element, and the mutations generated were and Morris, 1981), which is segregationally unstable in C. diffi-
extremely stable. The latter conclusion was based on the observa- cile. Therefore, they are easily lost from this host in the absence of
tion that when late exponential cultures of the 3 clostridial pyrF selection. Indeed, simple restreaking of colonies in the absence of
mutants were obtained (in C. acetobutylicum, C. sporogenes, and C. thiamphenicol is sufficient for the loss of pMTL007 and pMTL007C-
difficile) and plated out without dilution onto minimal media lack- E2 from C. difficile.
ing uracil, no growth (reverted colonies) was obtained (Heap et al., The ClosTron is an insertional mutagen. There are practical
2007). issues to be borne in mind in its use. Not all insertions may be muta-
genic. A proximal insertion site may result in a novel ORF encoding
10. Use of the ClosTron a fore-shortened protein with potential activity. Distal insertions
can result in the production of a truncated protein which retains
The practicalities of gene inactivation using the ClosTron are activity. Insertion sites should be chosen appropriately. The poten-
extremely straightforward. The group II intron is retargeted by tial for polar effects should also be considered. Depending upon
substituting a ∼350-bp fragment in plasmid pMTL007 with a the orientation of intron insertion, downstream genes or upstream
proportionately-sized fragment carrying the desired nucleotide genes (via antisense and transcriptional interference effects) may
changes. The fragment is generated in a one-step ‘Splicing by be affected. To alleviate polar effects, second-generation versions of
Overlap Extension’ (SOE)-PCR using primers which are auto- pMTL007 have been constructed (pMTL007C-E2) in which the ermB
matically designed by a computer algorithm (http://www.sigma- can be subsequently removed using FLP:FRT technology (Heap et
genosys.com/targetron/), and template DNA supplied in the Sigma al., 2010). The use of this system has the added benefit that it
Aldrich Targetron Gene Knockout System kit. The modified plas- provides the facility to make multiple mutations. Thus, following
mid is transferred from an E. coli donor into C. difficile 630erm the removal of ermB from a ClosTron-generated mutant, a second
by conjugation (Purdy et al., 2002). C. difficile transconjugants are gene can be targeted, and its inactivation selected on the basis of
selected based on thiamphenicol resistance, which is encoded by acquisition of erythromycin resistance. More significantly, we have
the catP gene of pMTL007 vectors. In order to isolate ClosTron now provided, free-of-charge, at www.clostron.com, access to an
integrant colonies, transconjugant colonies are simply transferred automated intron design bioinformatics tool, which, in combina-
from growth medium supplemented with thiamphenicol onto tion with outsourced construction of retargeted intron plasmids,
growth medium supplemented with erythromycin (but lacking thi- has significantly simplified the process. Using the resource at
amphenicol). A detailed account of events occurring in the host www.clostron.com, the application of this technology is more rapid
S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395 393

and cost-effective, and more fully exploits the potential of group II should be noted that data in our laboratory does not support this
introns. observation (Burns et al., 2010). Further analysis is clearly required.

Acknowledgements
11. Conclusions

The authors wish to acknowledge the financial support of the


The occurrence of CDAD has now risen to epidemic propor-
supported by SysMO (project number 2; http://www.sysmo.net),
tions in healthcare facilities throughout the developed world. The
the BBSRC (grants BB/D522797/1, BD/D522289/1, BB/F003390/1),
evolution of C. difficile strains which are better adapted to the mod-
the MRC (G0601176), and the EU (grants LSHE-CT-2006-037870
ern healthcare environment is almost certainly the reason for this,
and HEALTH-F3-2008-223585).
although the specific factors involved remain unclear at present.
The apparent dominance of ribotype 027 strains and the recent
emergence of ribotype 078 strains are particularly interesting. References
Traits which are common to strains of these ribotypes, includ-
ing variant tcdC genes and the presence of binary toxin, may be Acambis, 2007. Acambis Concludes C. difficile Vaccine Formulation Work
important. However, it is not clear whether they are sufficient to On Schedule and Will Commence Proof-of-concept Trial in 2008,
http://www.acambis.co.uk/default.asp?id=2033 [online], accessed March
explain the increased incidence of CDAD, and other factors may 30, 2009.
well be involved. Fluoroquinolone resistance may be important, Asensio, A., Vaque-Rafart, J., Calbo-Torrecillas, F., Gestal-Otero, J.J., López-Fernández,
although this is also common in less prevalent strains of C. diffi- F., Trilla-Garcia, A., Canton, R., EPINE Working Group, 2008. Increasing rates
in Clostridium difficile infection (CDI) among hospitalised patients, Spain
cile (Spigaglia et al., 2008). Nonetheless, the emergence of more 1999–2007. Euro Surveill. 13 (July (31)), 18943.
antibiotic-resistant profiles amongst strains of C. difficile would Agersø, Y., Pedersen, A.G., Aarestrup, F.M., 2006. Identification of Tn5397-like and
have profound implications for how the disease is treated. Of par- Tn916-like transposons and diversity of the tetracycline resistance gene tet(M)
in enterococci from humans, pigs and poultry. J. Antimicrob. Chemother. 57,
ticular concern is the prospect of lateral transfer of van genes from 832–839.
enterococcal species which may give rise to vancomycin-resistant Akerlund, T., Persson, I., Unemo, M., Noren, T., Svenungsson, B., Wullt, M., Bur-
strains of C. difficile. man, L.G., 2008. Increased sporulation rate of epidemic Clostridium difficile type
027/NAP1. J. Clin. Microbiol. 46, 1530–1533.
It is clear that genetic studies will play a vital role in identifying
Archibald, L.K., Banerjee, S.N., Jarvis, W.R., 2004. Secular trends in hospital-acquired
the unrecognized virulence factors of C. difficile and thus, mutage- Clostridium difficile disease in the United States. J. Infect. Dis. 189, 1585–1589.
nesis tools will play a vital role. Indeed, the value of reverse genetic Ausiello, C.M., Cerquetti, M., Fedele, G., Spensieri, F., Palazzo, R., Nasso, M., Frezza, S.,
Mastrantonio, P., 2006. Surface layer proteins from Clostridium difficile induce
studies has recently been demonstrated by Lyras et al. (2009) in
inflammatory and regulatory cytokines in human monocytes and dendritic cells.
elucidating the role of toxin B in C. difficile virulence. Historically, Microbes Infect. 8, 2640–2646.
genetic manipulation of C. difficile has been extremely difficult. Baines, S.D., O’Connor, R., Freeman, J., Fawley, W.N., Harmanus, C., Mastrantonio, P.,
However, the advent of ClosTron technology now provides a facile Kuijper, E.J., Wilcox, M.H., 2008. Emergence of reduced susceptibility to metron-
idazole in Clostridium difficile. J. Antimicrob. Chemother. 62, 1046–1052.
means of generating genetically stable mutants of C. difficile (Heap Barbut, F., Mastrantonio, P., Delmée, M., Brazier, J., Kuijper, E., Poxton, I., European
et al., 2007). Since publication of the ClosTron, we have generated Study Group on Clostridium difficile (ESGCD), 2007. Prospective study of Clostrid-
over 150 different mutants between ourselves and collaborators. ium difficile infections in Europe with phenotypic and genotypic characterisation
of the isolates. Clin. Microbiol. Infect. 13, 1048–1057.
Moreover, we have made numerous mutants in C. difficile strain Borgmann, S., Kist, M., Jakobiak, T., Reil, M., Scholz, E., von Eichel-Streiber, C., Gru-
R20291, the ribotype 027 isolate responsible for the outbreaks of ber, H., Brazier, J.S., Schulte, B., 2008. Increased number of Clostridium difficile
CDAD in the UK’s Stoke Mandeville hospital in 2003–2004 and infections and prevalence of Clostridium difficile PCR ribotype 001 in southern
Germany. Euro Surveill. 13, pii: 19057.
2004–2005. It is worth noting that the ClosTron procedure is Bradshaw, M., Marshall, K.M., Heap, J.T., Tepp, W.H., Minton, N.P., Johnson, E.A., 2010.
extremely rapid (taking as few as 10–14 days per mutant), highly Construction of a nontoxigenic Clostridium botulinum strain for food challenge
efficient (100s of retargeted clones obtained per experiment), and studies. Appl. Environ. Microbiol. 76, 387–393.
Brazier, J.S., Fawley, W., Freeman, J., Wilcox, M.H., 2001. Reduced susceptibility of
reproducible (100% success rate among non-essential genes), and
Clostridium difficile to metronidazole. J. Antimicrob. Chemother. 48, 741–742.
is revolutionizing functional genomic studies in clostridia based on Brazier, J.S., 2005. Antimicrobial susceptibility of polymerase chain reaction ribo-
reverse genetics (Bradshaw et al., 2010; Burns et al., 2010; Emerson types of Clostridium difficile commonly isolated from symptomatic hospital
patients in the UK. J. Hosp. Infect. 61, 11–14.
et al., 2009; Kirby et al., 2009; Twine et al., 2009; Underwood et al.,
Brazier, J.S., Patel, B., Pearson, A., 2007. Distribution of Clostridium difficile PCR ribo-
2009). In contrast, forward genetic studies have until now been type 027 in British hospitals. Euro Surveill. 12, 3182.
prohibited due to the absence of an effective random mutagen. Brazier, J.S., Raybould, R., Patel, B., Duckworth, G., Pearson, A., Charlett, A., Duerden,
The situation has now changed with the recent development of a B.I., 2008. HPA Regional Microbiology Network, 2008. Distribution and antimi-
crobial susceptibility patterns of Clostridium difficile PCR ribotypes in English
mariner-based transposon system (Cartman and Minton, 2010). Its hospitals, 2007–2008. Euro Surveill. 13 (41), pii: 19000.
utility has been exemplified in C. difficile R20291, where transposi- Burns, D.A., Heap, J.T., Minton, N.P., 2010. SleC is essential for germination of Clostrid-
tion occurred at random genomic loci, as determined by sequencing ium difficile spores in nutrient-rich medium supplemented with the bile salt
taurocholate. J. Bacteriol. 192, 657–664.
the insertion site of 60 randomly picked transposition events. The Cartman, S.T., Minton, N.P., 2010. A mariner-based transposon system for in
only requirement for insertion is the sequence ‘TA’. vivo random mutagenesis of Clostridium difficile. Appl. Environ. Microbiol. 76,
The deployment of mutational tools will facilitate studies aimed 1103–1109.
Cerquetti, M., Molinari, A., Sebastianelli, A., Diociaiuti, M., Petruzzelli, R., Capo, C.,
at providing fundamental aspects of clostridial biology. One such Mastrantonio, P., 2000. Characterization of surface layer proteins from different
area is spore formation and germination, an as yet little stud- Clostridium difficile clinical isolates. Microb. Pathog. 28, 363–372.
ied phenomenon in Clostridium. Spore longevity is as remarkable Chen, Y., McClane, B.A., Fisher, D.J., Rood, J.I., Gupta, P., 2005. Construction of an alpha
toxin gene knockout mutant of Clostridium perfringens type A by use of a mobile
as their ability to withstand inimical agents, such as extremes of
group II intron. Appl. Environ. Microbiol. 71, 7542–7547.
temperature, desiccation, radiation, and disinfectants. Spores are Clooten, J., Kruth, S., Arroyo, L., Weese, J.S., 2008. Prevalence and risk factors for
thought to play a pivotal role in the spread of CDAD. It has been Clostridium difficile colonization in dogs and cats hospitalized in an intensive
care unit. Vet. Microbiol. 129, 209–214.
suggested that epidemic strains of C. difficile appear to be more
Critchley, I.A., Green, L.S., Young, C.L., Bullard, J.M., Evans, R.J., Price, M., Jarvis, T.C.,
proficient in their ability to form spores than standard laboratory Guiles, J.W., Janjic, N., Ochsner, U.A., 2009. Spectrum of activity and mode of
strains (Akerlund et al., 2008; Fawley et al., 2007). If this is gener- action of REP3123, a new antibiotic to treat Clostridium difficile infections. J.
ally the case, infection control measures which specifically target Antimicrob. Chemother. 63, 954–963.
Debast, S.B., van Leengoed, L.A., Goorhuis, A., Harmanus, C., Kuijper, E.J., Bergwerff,
spores and their formation offer hope as a means of controlling A.A., 2009. Clostridium difficile PCR ribotype 078 toxinotype V found in diarrhoeal
the spread of CDAD throughout healthcare settings. However, it pigs identical to isolates from affected humans. Environ. Microbiol. 11, 505–511.
394 S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395

Dineen, S.S., Villapakkam, A.C., Nordman, J.T., Sonenshein, A.L., 2007. Repression of Kuijper, E.J., van Dissel, J.T., Wilcox, M.H., 2007. Clostridium difficile: changing epi-
Clostridium difficile toxin gene expression by CodY. Mol. Microbiol. 66, 206–219. demiology and new treatment options. Curr. Opin. Infect. Dis. 20, 376–383.
Drudy, D., Harnedy, N., Fanning, S., Hannan, M., Kyne, L., 2007. Emergence and con- Launay, A., Ballard, S.A., Johnson, P.D., Grayson, M.L., Lambert, T., 2006. Transfer
trol of fluoroquinolone-resistant, toxin A-negative, toxin B-positive Clostridium of vancomycin resistance transposon Tn1549 from Clostridium symbiosum to
difficile. Infect. Control. Hosp. Epidemiol. 28, 932–940. Enterococcus spp. in the gut of gnotobiotic mice. Antimicrob. Agents Chemother.
Dupuy, B., Govind, R., Antunes, A., Matamouros, S., 2008. Clostridium difficile toxin 50, 1054–1062.
synthesis is negatively regulated by TcdC. J. Med. Microbiol. 57, 685–689. Liyanage, H., Kashket, S., Young, M., Kashket, E.R., 2001. Clostridium beijerinckii and
Emerson, J.E., Reynolds, C.B., Fagan, R.P., Shaw, H.A., Goulding, D., Fairweather, N.F., Clostridium difficile detoxify methylglyoxal by a novel mechanism involving glyc-
2009. A novel genetic switch controls phase variable expression of CwpV, a erol dehydrogenase. Appl. Environ. Microbiol. 67, 2004–2010.
Clostridium difficile cell wall protein. Mol. Microbiol. 74, 541–556. Lyras, D., O’Connor, J.R., Howarth, P.M., Sambol, S.P., Carter, G.P., Phumoonna, T.,
Elixhauser, A., Jhung, M., 2008. Clostridium difficile-associated disease in US hos- Poon, R., Adams, V., Vedantamm, G., Johnson, S., Gerding, D.N., Rood, J.I., 2009.
pitals, 1993–2005. In: HCUP Statistical Brief No. 50, April 2008. US Agency for Toxin B is essential for virulence of Clostridium difficile. Nature 458, 1176–1179.
Healthcare Research and Quality, Rockville, MD, Available at: http://www.hcup- McDonald, L.C., Owings, M., Jernigan, J.B., 2006. Clostridium difficile infection
us.ahrq.gov/reports/statbriefs/sb50.pdf, accessed March 23, 2009. in patients discharged from US short-stay hospitals. Emerg. Infect. Dis. 12,
European Centre for Disease Prevention and Control, 2005. ECDC Summary of CDAD 409–415.
Infection, 2005–2007. Minton, N.P., Morris, J.G., 1981. Isolation and partial characterisation of three cryp-
Fawley, W.N., Underwood, S., Freeman, J., Baines, S.D., Saxton, K., Stephenson, K., tic plasmids from strains of Clostridium butyricum. J. Gen. Microbiol. 127, 325–
Owens Jr., R.C., Wilcox, M.H., 2007. Efficacy of hospital cleaning agents and 331.
germicides against epidemic Clostridium difficile strains. Infect. Control Hosp. Mohr, G., Smith, D., Belfort, M., Lambowitz, A.M., 2000. Rules for DNA target-site
Epidemiol. 28, 920–925. recognition by a lactococcal group II intron enable retargeting of the intron to
Giannasca, P.J., Zhang, Z.X., Lei, W.D., Boden, J.A., Giel, M.A., Monath, T.P., Thomas Jr., specific DNA sequences. Genes Dev. 14, 559–573.
W.D., 1999. Serum antitoxin antibodies mediate systemic and mucosal protec- Mylonakis, E., Ryan, E.T., Calderwood, S.B., 2001. Clostridium difficile-associated diar-
tion from Clostridium difficile disease in hamsters. Infect. Immun. 67, 527–538. rhea. Arch. Intern. Med. 161, 525–533.
Girbal, L., Mortier-Barriere, I., Raynaud, F., Rouanet, C., Croux, C., Soucaille, P., Ní Eidhin, D.B., O’Brien, J.B., McCabe, M.S., Athié-Morales, V., Kelleher, D.P., 2008.
2003. Development of a sensitive gene expression reporter system and an Active immunization of hamsters against Clostridium difficile infection using
inducible promoter-repressor system for Clostridium acetobutylicum. Appl. Env- surface-layer protein. FEMS Immunol. Med. Microbiol. 52, 207–218.
iron. Microbiol. 69, 4985–4988. O’Brien, J.A., Lahue, B.J., Caro, J.J., Davidson, D.M., 2007. The emerging infectious
Goorhuis, A., Bakker, D., Corver, J., Debast, S.B., Harmanus, C., Notermans, D.W., Berg- challenge of Clostridium difficile-associated disease in Massachusetts hospi-
werff, A.A., Dekker, F.W., Kuijper, E.J., 2008. Emergence of Clostridium difficile tals: clinical and economic consequences. Infect. Control Hosp. Epidemiol. 28,
infection due to a new hypervirulent strain, polymerase chain reaction ribotype 1219–1227.
078. Clin. Infect. Dis. 47, 1162–1170. O’Connor, J.R., Lyras, D., Farrow, K.A., Adams, V., Powell, D.R., Hinds, J., Cheung, J.K.,
Gravel, D., Miller, M., Simor, A., Taylor, G., Gardam, M., McGeer, A., Hutchinson, Rood, J.I., 2006. Construction and analysis of chromosomal Clostridium difficile
J., Moore, D., Kelly, S., Boyd, D., Mulvey, M., Canadian Nosocomial Infection mutants. Mol. Microbiol. 61, 1335–1351.
Surveillance Program, 2009. Health care-associated Clostridium difficile infec- Péchiné, S., Janoir, C., Boureau, H., Gleizes, A., Tsapis, N., Hoys, S., Fattal, E., Col-
tion in adults admitted to acute care hospitals in Canada: a Canadian Nosocomial lignon, A., 2007. Diminished intestinal colonization by Clostridium difficile and
Infection Surveillance Program study. Clin. Infect. Dis. 48, 568–576. immune response in mice after mucosal immunization with surface proteins of
Heap, J.T., Pennington, O.J., Cartman, S.T., Carter, G.P., Minton, N.P., 2007. The Clostridium difficile. Vaccine 25, 3946–3954.
ClosTron: a universal gene knock-out system for the genus Clostridium. J. Micro- Péchiné, S., Gleizes, A., Janoir, C., Gorges-Kergot, R., Barc, M.C., Delmée, M., Collignon,
biol. Methods 70, 452–464. A., 2005b. Immunological properties of surface proteins of Clostridium difficile.
Heap, J.T., Cartman, S.T., Pennington, O.J., Cooksley, C.M., Scott, J.C., Blount, B., Burns, J. Med. Microbiol. 54, 193–196.
D., Minton, N.P., 2008. Development of genetic knock-out systems for clostridia. Pechine, S., Janoir, C., Collignon, A., 2005a. Variability of Clostridium difficile sur-
In: Bruggermann, H., Gottschalk, G. (Eds.), Clostridia: Molecular Biology in the face proteins and specific serum antibody response in patients with Clostridium
Post-Genomic Era. Caister Academic Press. difficile-associated disease. J. Clin. Microbiol. 43, 5018–5025.
Heap, J.T., Kuehne, S.A., Ehsaan, M., Cartman, S.T., Cooksley, C.M., Scott, J.C., Minton, Peláez, T., Alcala, L., Alonso, R., Martín-Lopez, A., Garcia-Arias, V., Marín, M., Bouza, E.,
N.P., 2010. The ClosTron: mutagenesis in Clostridium refined and streamlined. J. 2005. In vitro activity of ramoplanin against Clostridium difficile, including strains
Microbiol. Methods 80, 49–55. with reduced susceptibility to vancomycin or with resistance to metronidazole.
Hedge, D.D., Strain, J.D., Heins, J.R., Farver, D.K., 2008. New advances in the treatment Antimicrob. Agents Chemother. 49, 1157–1159.
of Clostridium difficile infection (CDI). Ther. Clin. Risk. Manage. 4, 949–964. Pépin, J., Valiquette, L., Alary, M.E., Villemure, P., Pelletier, A., Forget, K., Pépin, K.,
Hennequin, C., Porcheray, F., Waligora-Dupriet, A., Collignon, A., Barc, M., Bourlioux, Chouinard, D., 2004. Clostridium difficile-associated diarrhea in a region of Que-
P., Karjalainen, T., 2001. GroEL (Hsp60) of Clostridium difficile is involved in cell bec from 1991 to 2003: a changing pattern of disease severity. Can. Med. Assoc.
adherence. Microbiology 147, 87–96. J. 171, 466–472.
Hennequin, C., Janoir, C., Barc, M.C., Collignon, A., Karjalainen, T., 2003. Identification Pépin, J., Saheb, N., Coulombe, M.A., Alary, M.E., Corriveau, M.P., Authier, S., Leblanc,
and characterization of a fibronectin-binding protein from Clostridium difficile. M., Rivard, G., Bettez, M., Primeau, V., Nguyen, M., Jacob, C.E., Lanthier, L., 2005.
Microbiology 149, 2779–2787. Emergence of fluoroquinolones as the predominant risk factor for Clostridium
Hussain, H.A., Roberts, A.P., Mullany, P., 2005. Generation of an erythromycin- difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin.
sensitive derivative of Clostridium difficile strain 630 (630Deltaerm) and Infect. Dis. 41, 1254–1260.
demonstration that the conjugative transposon Tn916DeltaE enters the genome Perutka, J., Wang, W., Goerlitz, D., Lambowitz, A.M., 2004. Use of computer-designed
of this strain at multiple sites. J. Med. Microbiol. 54, 137–141. group II introns to disrupt Escherichia coli DExH/D-box protein and DNA helicase
Jarvis, W.R., Schlosser, J., Jarvis, A.A., Chinn, R.Y., 2009. National point prevalence of genes. J. Mol. Biol. 336, 421–439.
Clostridium difficile in US health care facility inpatients. Am. J. Infect. Control 37, Poutanen, S.M., Simor, A.E., 2004. Clostridium difficile-associated diarrhea in adults.
263–270. Can. Med. Assoc. J. 171, 51–58.
Janoir, C., Péchiné, S., Grosdidier, C., Collignon, A., 2007. Cwp84, a surface-associated Purdy, D., O’Keeffe, T.A., Elmore, M., Herbert, M., McLeod, A., Bokori-Brown, M.,
protein of Clostridium difficile, is a cysteine protease with degrading activity on Ostrowski, A., Minton, N.P., 2002. Conjugative transfer of clostridial shuttle vec-
extracellular matrix proteins. J. Bacteriol. 189, 7174–7180. tors from Escherichia coli to Clostridium difficile through circumvention of the
Karberg, M., Guo, H., Zhong, J., Coon, R., Perutka, J., Lambowitz, A.M., 2001. Group restriction barrier. Mol. Microbiol. 46, 439–452.
II introns as controllable gene targeting vectors for genetic manipulation of Roberts, A.P., Johanesen, P.A., Lyras, D., Mullany, P., Rood, J.I., 2001. Comparison
bacteria. Nat. Biotechnol. 19, 1162–1167. of Tn5397 from Clostridium difficile, Tn916 from Enterococcus faecalis and the
Kelly, C.P., Pothoulakis, C., LaMont, J.T., 1994. Clostridium difficile colitis. N. Engl. J. CW459tet(M) element from Clostridium perfringens shows that they have similar
Med. 330, 257–262. conjugation regions but different insertion and excision modules. Microbiology
Kim, P.H., Iaconis, J.P., Rolfe, R.D., 1987. Immunization of adult hamsters against 147, 1243–1251.
Clostridium difficile-associated ileocecitis and transfer of protection to infant Rohde, C.L., Bartolini, V., Jones, N., 2009. The use of probiotics in the prevention and
hamsters. Infect. Immun. 55, 2984–2992. treatment of antibiotic-associated diarrhea with special interest in Clostridium
Kirby, J.M., Ahern, H., Roberts, A.K., Kumar, V., Freeman, Z., Acharya, K.R., Shone, C.C., difficile-associated diarrhea. Nutr. Clin. Pract. 24, 33–40.
2009. Cwp84, a surface-associated, cysteine protease, plays a role in the matura- Sebaihia, M., Wren, B.W., Mullany, P., Fairweather, N.F., Minton, N., Stabler, R., Thom-
tion of the surface layer of Clostridium difficile, J. Biol. Chem. 284, 34666–34673. son, N.R., Roberts, A.P., Cerdeno-Tarraga, A.M., Wang, H., Holden, M.T., Wright,
Kuijper, E.J., Barbut, F., Brazier, J.S., Kleinkauf, N., Eckmanns, T., Lambert, M.L., Drudy, A., Churcher, C., Quail, M.A., Baker, S., Bason, N., Brooks, K., Chillingworth, T.,
D., Fitzpatrick, F., Wiuff, C., Brown, D.J., Coia, J.E., Pituch, H., Reichert, P., Even, J., Cronin, A., Davis, P., Dowd, L., Fraser, A., Feltwell, T., Hance, Z., Holroyd, S., Jagels,
Mossong, J., Widmer, A.F., Olsen, K.E., Allerberger, F., Notermans, D.W., Delmée, K., Moule, S., Mungall, K., Price, C., Rabbinowitsch, E., Sharp, S., Simmonds, M.,
M., Coignard, B., Wilcox, M., Patel, B., Frei, R., Nagy, E., Bouza, E., Marin, M., Stevens, K., Unwin, L., Whithead, S., Dupuy, B., Dougan, G., Barrell, B., Parkhill, J.,
Akerlund, T., Virolainen-Julkunen, A., Lyytikäinen, O., Kotila, S., Ingebretsen, A., 2006. The multidrug-resistant human pathogen Clostridium difficile has a highly
Smyth, B., Rooney, P., Poxton, I.R., Monnet, D.L., 2008. Update of Clostridium mobile, mosaic genome. Nat. Genet. 38, 779–786.
difficile infection due to PCR ribotype 027 in Europe. Euro Surveill. 31 (13), 31. Schmidt, C., Löffler, B., Ackermann, G., 2007. Antimicrobial phenotypes and molec-
Kuijper, E.J., Coignard, B., Tüll, P., ESCMID Study Group for Clostridium difficile; ular basis in clinical strains of Clostridium difficile. Diagn. Microbiol. Infect. Dis.
EU Member States; European Centre for Disease Prevention and Control, 2006. 59, 1–5.
Emergence of Clostridium difficile-associated disease in North America and Songer, J.G., Anderson, M.A., 2005. Clostridium difficile: an important pathogen of
Europe. Clin. Microbiol. Infect. 12 (Suppl. 6), 2–18. food animals. Anaerobe 12, 1–4.
S.T. Cartman et al. / International Journal of Medical Microbiology 300 (2010) 387–395 395

Spigaglia, P., Barbanti, F., Mastrantonio, P., Brazier, J.S., Barbut, F., Delmée, M., Kuijper, Underwood, S., Guan, S., Vijayasubhash, V., Baines, S.D., Graham, L., Lewis, R.J.,
E., Poxton, I.R., European Study Group on Clostridium difficile (ESGCD), 2008. Wilcox, M.H., Stephenson, K., 2009. Characterisation of the sporulation initia-
Fluoroquinolone resistance in Clostridium difficile isolates from a prospective tion pathway of Clostridium difficile and the role in toxin production. J. Bacteriol.
study of C. difficile infections in Europe. J. Med. Microbiol. 57, 784–789. 191, 7296–7305.
Stabler, R.A., Dawson, L.F., Phua, L.T., Wren, B.W., 2008. Comparative analysis of Waligora, A.J., Hennequin, C., Mullany, P., Bourlioux, P., Collignon, A., Karjalainen,
BI/NAP1/027 hypervirulent strains reveals novel toxin B-encoding gene (tcdB) T., 2001. Characterization of a cell surface protein of Clostridium difficile with
sequences. J. Med. Microbiol. 57, 771–775. adhesive properties. Infect. Immun. 69, 2144–2153.
Stinear, T.P., Olden, D.C., Johnson, P.D., Davies, J.K., Grayson, M.L., 2001. Enterococ- Warny, M., Pépin, J., Fang, A., Killgore, G., Thompson, A., Brazier, J., Frost, E., McDon-
cal vanB resistance locus in anaerobic bacteria in human faeces. Lancet 357, ald, L.C., 2005. Toxin production by an emerging strain of Clostridium difficile
855–856. associated with outbreaks of severe disease in North America and Europe. Lancet
Suetens, C., 2008. Clostridium difficile: summary of actions in the European 366, 1079–1084.
Union. Euro Surveill. 13 (31), pii: 18944, Available online: http://www. Weil, H.P., Fischer-Brügge, U., Harmanus, C., Mattner, F., Gastmeier, P., Kuijper, E.J.,
eurosurveillance.org/ViewArticle.aspx?ArticleId=18944. 2007. High incidence of Clostridium difficile-associated diarrhoea with a com-
Tasteyre, A., Barc, M.C., Collignon, A., Boureau, H., Karjalainen, T., 2001. Role of FliC munity onset in a hyperendemic region in Germany. In: Abstracts of the 17th
and FliD flagellar proteins of Clostridium difficile in adherence and gut coloniza- Europ. Congr. Clin. Microbiol. Infect. Dis., ICC, Munich, Germany, March 31–April
tion. Infect. Immun. 69, 7937–7940. 4, 2007.
Torres, J.F., Lyerly, D.M., Hill, J.E., Monath, T.P., 1995. Evaluation of formalin- Zhong, J., Karberg, M., Lambowitz, A.M., 2003. Targeted and random bacte-
inactivated Clostridium difficile vaccines administered by parenteral and mucosal rial gene disruption using a group II intron (targetron) vector containing
routes of immunization in hamsters. Infect. Immun. 63, 4619–4627. a retrotransposition-activated selectable marker. Nucleic Acids Res. 31,
Twine, S.M., Reid, C.W., Aubry, A., McMullin, D.R., Fulton, K.M., Austin, J., Logan, S.M., 1656–1664.
2009. Motility and flagellar glycosylation in Clostridium difficile. J. Bacteriol. 191,
7050–7062.

You might also like