Vous êtes sur la page 1sur 9

REVIEW

Clostridium butyricum: from beneficial to a new emerging pathogen

N. Cassir, S. Benamar and B. La Scola


Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), UM63 CNRS 7278 IRD 198 INSERM U1095, Facultés de Médecine
et de Pharmacie, Aix-Marseille Université, Marseille, France

Abstract

Clostridium butyricum, a strictly anaerobic spore-forming bacillus, is a common human and animal gut commensal bacterium, and is also
frequently found in the environment. Whereas non-toxigenic strains are currently used as probiotics in Asia, other strains have been
implicated in pathological conditions, such as botulism in infants or necrotizing enterocolitis in preterm neonates. In terms of the latter,
within the same species, different strains have antagonist effects on the intestinal mucosa. In particular, short-chain fatty acids, which are
products of carbohydrate fermentation, have a dose-dependent paradoxical effect. Moreover, toxin genes have been identified by genome
sequencing in pathological strains. Asymptomatic carriage of these strains has also been reported. Herein, we provide an overview of the
implications of C. butyricum for human health, from the beneficial to the pathogenic. We focus on pathogenic strains associated with the
occurrence of necrotizing enterocolitis. We also discuss the need to use complementary microbiological methods, including culture, in
order to better assess gut bacterial diversity and identify new emergent enteropathogens at the strain level.
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All
rights reserved.

Keywords: Clostridium butyricum, culturomics, metagenomics, necrotizing enterocolitis, toxins


Article published online: 20 October 2015

including C. butyricum [3]. Ferraris et al. found that 44% of faecal


Corresponding author: B. La Scola, Unité de Recherche sur les
samples collected from asymptomatic preterm neonates
Maladies Infectieuses et Tropicales Emergentes, URMITE, UM63,
CNRS 7278, IRD 198, INSERM 1095, Faculté de médecine, (n = 76), whose median gestational age was 32.8 weeks, tested
Aix-Marseille Université, 27 Boulevard Jean Moulin, 13385 Marseille positive for C. butyricum [4]. Surveys have identified C. butyricum
cedex 05, France
in 10–20% of human faecal samples by microbial culture [5].
E-mail: bernard.la-scola@univ-amu.fr
Moreover, this bacterium has been widely used as a probiotic in
Asia (particularly in Japan, Korea, and China) [1]. However,
some C. butyricum strains detected in stool samples have also
Introduction been implicated in pathological conditions such as botulism in
infants and necrotizing enterocolitis (NEC) in preterm neonates
[6,7].
Clostridium butyricum is a strictly anaerobic, Gram-positive,
Several Clostridium species involved in infectious diseases are
spore-forming bacillus named for its capacity to produce high
known to be pathogenic, essentially by producing potent toxins
amounts of butyric acid. It was first isolated from pig intestines
that are responsible for life-threatening diseases in humans and
by Prazmowski in 1880 [1]. Since then, several strains of
other animals [8]. Interestingly, these species produce the
C. butyricum have been described in a variety of environments,
highest number of toxins of any type of bacteria, and the genetic
and are common human and animal gut commensal bacteria. A
characteristics of toxigenic clostridia support horizontal toxin
recent study found C. butyricum strains in 302 of 978 environ-
gene transfer [8,9]. Notably, it has been shown that toxigenic
mental samples tested (31%) [2]. The highest percentage of
and non-toxigenic Clostridium difficile strains form part of the
isolates was found in soil, vegetables, soured milk, and cheeses.
normal human gut microbiota [10]. Moreover, within the same
In humans, following birth, the neonate’s intestine is progres-
species, antagonist effects have been described. A recent pilot
sively colonized by facultative and strictly anaerobic bacteria,

Clin Microbiol Infect 2016; 22: 37–45


Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved
http://dx.doi.org/10.1016/j.cmi.2015.10.014
38 Clinical Microbiology and Infection, Volume 22 Number 1, January 2016 CMI

study showed that the administration of spores of a non- with a suggested role of fermentation metabolites such as
toxigenic C. difficile strain was effective for the prevention of butyrate [21]. However, the same beneficial mechanism has
recurrent C. difficile infection caused by toxigenic strains [11]. been observed with 17 other Clostridium species belonging to
As fermentative bacteria, clostridia produce short-chain fatty clusters IV, XIVa, and XVIII [22]. This C. butyricum strain also
acids (SCFAs), primarily butyrate and acetate, through induced interleukin-10-producing macrophages in inflamed
fermentation of mainly undigested dietary carbohydrates. This mucosa to prevent acute experimental colitis in mice [19].
has the primary effect of stimulating the proliferation of Other experimental studies have shown the capacity of this
enterocytes [12]. It has also been shown that butyrate may strain to improve non-alcoholic fatty liver disease [23] and
downregulate the expression of several virulence genes [13]. prevent C. difficile infection [24], enterohaemorrhagic Escher-
Although previous reports have suggested that, besides ichia coli O157:H7 infection [25], and gastric ulcers [26]. In
C. difficile, other Clostridium species may also cause antibiotic- clinical practice, the administration of CBM588 was effective for
associated diarrhoea, anaerobic research in clinical practice is both the treatment and the prophylaxis of antibiotic-associated
frequently limited to C. difficile and its toxins [14]. Detection of diarrhoea in children [27]. It increased the level of anaerobes
clostridia in stool samples remains challenging. Although the and prevented the decrease in the level of Bifidobacterium
advent of modern molecular microbiological methods has species in subjects who received antibiotics.
expanded the degree of detection, it is also hampered by Moreover, it has been shown that CBM588 harbours a
several biases [15]. Consequently, there has been renewed in- plasmidic bacteriocin that is very similar to the previously
terest in culture methods [16,17]. In a recent comparative described butyricin 7423 [28], and is related to its probiotic
analysis of stool samples using culture and 16S rRNA gene effect [29]. The supernatant of cultured CBM588 inhibited the
sequencing data, only 15% overlap was found [16]. Comple- growth of Clostridium beijerinkii ATCC25752 and Clostridium
mentary approaches are therefore necessary to explore the gut pasteurianum, but not of C. beijerinkii ATCC6015, Clostridium
microbiota. Through the use of both techniques, we recently acetobutylicum, and C. butyricum ATCC19398. In addition, the
ascertained a specific association between NEC and C. butyricum purified recombinant bacteriocin showed bactericidal activity
[17]. These data were confirmed by the use of a specific against C. difficile and Clostridium perfringens, but had no effect on
quantitative real-time PCR (qPCR) assay to screen a larger set Gram-negative bacteria. These results suggest a narrow, strain-
of samples. Genome sequencing allowed the detection of genes specific spectrum of activity of this bacteriocin that cannot fully
encoding toxin-like proteins shared by this C. butyricum strain. explain the beneficial effects previously cited. Similarly, Gebhart
In this article, we will provide an overview of the various et al. have shown that some strains of C. difficile produce R-type
implications of C. butyricum for human health, from beneficial to bacteriocins, termed diffocins, which have specific bactericidal
pathogenic. We will focus on its role in the pathogenesis of effects against other strains of the same species [30].
NEC occurring in preterm neonates as compared with that of When orally administered, C. butyricum spores germinate and
C. difficile in pseudomembranous colitis occurring in the elderly. grow in intestinal tracts [5], and produce large amounts of
We will also discuss the need to use complementary microbi- SCFAs, such as butyrate and acetate [31]. SCFAs constitute an
ological methods, including culture, in order to better assess important energy source for intestinal cells, and have prolifer-
gut bacterial diversity and identify new emergent enter- ative effects on enterocytes [12]. Additionally, SCFAs have been
opathogens at the strain level. noted to have immune-modulatory effects on colonic inflam-
mation. They suppress inflammatory cytokine secretion in
cultured epithelial cells, facilitating tolerance of the intestinal
Probiotic
mucosa to the presence of vast quantities of living microor-
ganisms, and controlling the overgrowth of pathogens [12]. It is
The WHO has defined probiotic bacteria as ‘live microorgan- therefore hypothesized, but not proven, that the main mech-
isms which when administrated in adequate amounts confer a anism by which probiotic C. butyricum strains exert their
health benefit on the host’ [18]. Several studies have shown beneficial effect is through equilibrated SCFA production.
probiotic properties of specific C. butyricum strains, mainly
performed in Japan, Korea, and China, where those probiotic
Pathogenic
strains are commercialized [19,20]. A recent study found that
the C. butyricum strain MIYAIRI 588 (CBM588) promoted reg-
ulatory T-cell generation in the intestine through induction of Botulism
transforming growth factor-β1 from lamina propria dendritic Botulism is an acute paralytic disease caused by botulinum
cells. This pathway was mainly Toll-like receptor 2 dependent, neurotoxin (BoNT), which is most frequently secreted by
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
CMI Cassir et al. Clostridium butyricum antagonist effects 39

Clostridium botulinum [6]. Botulism can occur when BoNT- very similar to haemolysins shared by Brachyspira hyodysenteriae,
producing clostridia colonize wounds or the intestine and the aetiological agent of swine dysentery [41]. Among those
subsequently secrete the toxin, or when contaminated food is haemolysins, the pore-forming β-haemolysin has been
ingested (foodborne) [32]. In 1986, the first infant botulism case described as the main virulence factor capable of inducing
caused by a BoNT/E-producing C. butyricum strain was reported enterocyte necrotic lesions via the culture supernatant [42,43].
in Italy [6]. Since then, C. butyricum has been associated with Interestingly, the polypeptide encoded by the β-haemolysin
botulism in other countries (i.e. China, India, Japan, Ireland, and homologue gene was identified in the secretome of an NEC-
the USA) [32]. Notably, the operon encoding the BoNT/E toxin associated C. butyricum strain, confirming expression of this
harboured by the neurotoxigenic C. butyricum strains is very potential toxin [17]. However, the involvement of this toxin in
similar to that carried by group II type E toxin-producing the pathophysiology of NEC needs further confirmation.
C. botulinum strains [33]. The presence of the BoNT/E toxin Experimental studies showed the capacity of C. butyricum to
gene within either plasmids or the chromosome in different reproduce NEC-like lesions in animal models. Popoff et al.
Clostridium species is consistent with horizontal transfer events developed an experimental chicken model for C. butyricum
mediated by plasmids or phage, and recombination events caecitis with lesions mimicking neonatal NEC by using a human
mediated by mobile genetic elements such as transposons [33]. NEC strain [44]. In germ-free quails, C. butyricum was shown to
be the Clostridium species inducing the increased occurrence of
NEC NEC-like caecal wall lesions and the only Clostridium species
NEC is a devastating gastrointestinal disease causing high inducing pneumatosis intestinalis, preceded by high levels of
morbidity and mortality, affecting predominantly preterm neo- butyric acid production [31]. In this study, when quails were fed
nates during outbreaks [34]. Its clinical presentation is charac- a diet without lactose, no lesions were observed. Interestingly,
terized by abdominal distension, gastrointestinal bleeding, mucosal like preterm neonates, birds show low endogenous lactase
ulcerations and necrosis, portal venous gas, and pneumatosis activity, and their caeca have a physiological stasis favouring
intestinalis, with different degrees of severity [34]. Despite de- overgrowth with lactose-fermenting bacteria [45]. Among the
cades of research, the pathogenesis of NEC remains elusive. other Clostridium species inducing NEC-like lesions,
Although no aetiological microorganism has been definitively C. perfringens and Clostridium paraputrificum were those that
established, the most often implicated bacteria have been Clos- were also involved in human NEC cases [35,39,46]. Other
tridium species [35]. Howard et al. first described an association experimental findings showed that C. butyricum could play a
between C. butyricum and NEC in 1977 [7]. They identified primary role in the pathogenesis of NEC via the fermentation of
C. butyricum in blood and stool cultures in nine of ten preterm carbohydrate products, depending on the lactase deficiency of
neonates during an outbreak of NEC. In another study, Gorham preterm neonates [47,48]. By using a Caco-2 cell monolayer
et al. reported the presence of C. butyricum on the hands of model of the intestinal barrier, Peng et al. showed that butyrate
members of the medical and nursing staff during an NEC outbreak induced apoptosis and reduced the number of viable Caco-2
[36]. This was consistent with the effectiveness of preventive cells in a dose-dependent manner [49]. Additionally, Bousse-
measures in controlling such outbreaks [37]. Additionally, Sturm boua et al. showed that lactose fermentation was a prerequisite
et al. demonstrated a cytotoxic effect of the supernatant of a for the expression of C. butyricum enteropathogenicity.
C. butyricum strain isolated from a preterm neonate with NEC [38]. Numerous gas cysts were always observed in the caecal wall of
Recently, Smith et al. detected the presence of C. butyricum with quails mono-associated with C. butyricum [50]. Neuraminidase
high density in two surgical samples from preterm neonates with production has also been described as a major virulence factor
NEC. Both specimens were characterized by histological pneu- in C. butyricum strains causing NEC in preterm neonates [51].
matosis intestinalis [39]. In a previous study, we analysed 30 stool Azcarate-Peril et al. demonstrated, in an experimental piglet
samples from preterm neonates with and without NEC by using model of NEC, that the occurrence of disease was associated
16S rRNA pyrosequencing and culture-based methods, and 163 with significantly higher loads of C. butyricum, as ascertained by
samples by using C. butyricum qPCR [17]; C. butyricum was specif- qPCR in ileal mucosa samples, and overall bacterial dysbiosis
ically associated with NEC, and culture supernatants of [52]. These results suggest that lactose fermentation could be a
C. butyricum strains from preterm neonates with NEC showed prerequisite for the pathological changes observed in NEC
significant cytotoxic activity. patients, and stress the roles played by both the strain and the
Genome sequencing allowed the identification of various host in the expression of C. butyricum enteropathogenicity.
toxin genes present in these pathogenic C. butyricum strains Further studies are needed to confirm the toxigenic mechanism
[17,40]. We previously identified, in NEC-associated by which C. butyricum is involved in the pathogenesis of NEC.
C. butyricum strains, four genes encoding polypeptides that are Given that C. perfringens and the new species Clostridium
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
40 Clinical Microbiology and Infection, Volume 22 Number 1, January 2016 CMI

neonatale have also been associated with hospital outbreaks of studies overlooked Gram-negative populations as well as mi-
NEC [46,53], the potential involvement of other Clostridium nority species [60]. Enteropathogens that can be present with
species represents an exciting area of future research. a subdominant status (<105 bacteria/mL) may therefore be
neglected, adding to the difficulty in achieving clear, repro-
Other conditions ducible detection [15]. Third, microbial diversities measured
A new pathogenic C. butyricum strain (NOR33234) has recently in the same sample and relative taxon abundances may differ
been isolated from an elderly patient with antibiotic-associated with the DNA extraction methods, targeted 16S rRNA re-
diarrhoea; tests for C. difficile toxins gave negative results [40]. gion(s), and sequencing technology [15]. Although the V6
Among the proteins annotated from genome sequencing of this region is only 58 bp in length, it shows considerable sequence
strain, there was an enterotoxin (OA81_00270). Moreover, two variability, and appears to be the best region for distinguishing
annotated proteins had sequence similarity to phage holins in Clostridium species [61]. We have recently analysed faecal
C. botulinum. A previous report has shown that holin-like tcdE is samples from preterm neonates by using 16S rRNA (V6) gene
required for exporting enterotoxins tcdA and tcdB in C. difficile sequencing, and identified, for the first time, C. butyricum in 12
[54]. Whether the enterotoxin or holin plays a pathogenic role of 15 samples from patients with NEC [17]. The high het-
in C. butyricum infection remains to be examined. erogeneity of results obtained by previous studies character-
To the best of our knowledge, only one case has been re- izing the bacterial profile associated with NEC may, in part, be
ported of C. butyricum bacteraemia and sepsis, which occurred explained by the use of different molecular methods for bac-
in an injecting drug user [55]. terial identification, including the target 16S rRNA region
(Table 1). Finally, 16S rRNA gene sequencing results are only
semiquantitative, and cannot distinguish between living and
Complementary tools for identification
dead bacteria [62]. This is a major limitation when the clinical
relevance of the results obtained with molecular methods is
Detection of bacteria from stool samples is challenging, given considered.
that the human gut contains approximately 1014 bacteria [56],
and that, for a single person, the number of different phylotypes Culture-based analysis
has been estimated to be at least 130–200 by molecular At the strain level, clones with high virulence and conta-
methods [57]. Clostridia belong to a complex phylogenetically giousness could emerge among populations of pathogenic and
heterogeneous group, with some clusters having high genetic non-pathogenic species [63]. So far, despite all of the recent
strain diversity [58]. Depending on the microbiological method technological advances in molecular biology, pure culture is
used, the species may be incorrectly identified. For example, a the only way to characterize the physiological properties of
strain was recently isolated and identified by biochemical living bacteria and to assess their potential for virulence at the
analysis (VITEK2) as Clostridium clostridioforme, whereas 16S strain level [64]. Despite this, culture methods have rarely
analysis identified the same sample as C. butyricum with 99.0% been used in studies analysing the bacterial profile associated
nucleotide identity, within the species definition threshold [40]. with NEC (Table 2).
The following two sections discuss the available culture- In the 1970s, the application of anaerobic conditions to the
independent and culture-based methods. culture of gut microorganisms enabled the recovery of a large
variety of anaerobes, including clostridia, from clinical samples
Culture-independent analysis [65]. With the advent of metagenomics, molecular tools
Although 16S rRNA gene pyrosequencing is now widely used replaced culture methods in microbial ecological analysis; those
in microbial ecological analysis, expanding the depth of were considered to be time-consuming, to be fastidious, and to
detection within gut microbiota, it is also hampered by several lack sensibility [15]. Regarding anaerobes, Hayashi et al. stressed
biases [15]. First, automated taxonomic assignment and the the need to reintroduce culture methods [66]. They compared
short length of the sequences amplified allow only limited gut bacterial diversity assessed by 16S rRNA gene sequencing
identification from phylum to genus [59]. However, in a pre- with that assessed by anaerobic culture, and identified compa-
vious study, we showed that it was possible to optimize the rable numbers of phylotypes or species with both methods, with
assignment at species level for each separate read, by using imperfect overlap. Goodman et al. later demonstrated that the
BLAST similarity searches against the National Center for use of straightforward anaerobic culture conditions made it
Biotechnology Information database. Species level was defined possible to enlarge the repertoire of cultivable bacteria from the
with a minimum sequence identity of 98.7% as the best BLAST gut [67]. In 2012, the new approach known as ‘microbial cul-
hit [17]. Second, it has recently been shown that molecular turomics’ (large-scale culture conditions with identification of
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
CMI
TABLE 1. Gut microbiota profile associated with necrotizing enterocolitis (NEC) (case–control studies), analysed with culture-independent methods

Culture- Number of
independent Region patients Sample
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45

analysis (16S rRNA) (NEC/controls) collection Location Phylum level Genus/family level Species level Diversity Reference

454-pyrosequencing V6 15/15 Stool; NEC onset 3 NICUs, South-eastern No difference No difference Clostridium butyricum Lower Cassir, 2015 [17]
France (12 vs. 2)
V3–V5 12/26 Stool; daily until 1 NICU, Boston, USA NA Clostridium associated with NA Lower Zhou, 2015 [70]
discharge NEC in early-onset cases
and with
Gammaproteobacteria in
late-onset cases
V1–V3 21/74 Stool; 0–5 days 1 NICU, Louisiana, USA Lower prevalence and abundance Lower abundance in NA Lower McMurtry, 2015 [71]
before NEC onset in Actinobacteria and clostridia Veillonella and
Streptococcus
V3–V5 12/44 Stool; weekly until 1 NICU, London, UK No difference Clostridium associated with Clostridium OTU in 4 No Sim, 2014 [46]
NEC onset NEC NEC patients; difference
Klebsiella OTU in 7
NEC patients
V2–V4 16/10 Intestinal samples 1 NICU, Pittsburgh, USA No difference Clostridium and Bacteroides Clostridium perfringens Lower Brower-Sinning,
enrichment in 3 NEC samples; 2014 [72]
Clostridium
paraputrificum in 2
NEC samples
V3–V4 5/5 Stool; weekly until 1 NICU, Chicago, USA Proteobacteria increase; Firmicutes Enterobacteriaceae increase; NA Lower Claud, 2013 [73]
NEC onset decrease Veillonellaceae decrease
V3–V5 11/21 Stool; NEC onset 2 NICUs, Cincinatti, USA Increased Firmicutes in 4 patients Propionibacerium lack; NA Lower Morrow, 2013 [74]
with NEC increased
Enterobacteriaceae in 7
patients with NEC
V1–V3 9/9 Stool; 1 week and 3 NICUs, Florida, USA Bloom of Proteobacteria and Enterobacteriaceae NA NA Mai, 2011 [75]
72 h before NEC Firmicutes decrease in NEC cases sequences present more
onset between the 1-week and 72-h frequently
samples
V1–V2 4/6 Stool; 1 week before 1 NICU, Florida, USA No difference Increased abundance of Citrobacter-like species NA Mshvildadze,

Cassir et al.
NEC onset Enterococcus in 3 NEC samples 2010 [76]
Genome-resolved — 3/4 Stool; NEC onset 1 NICU, Pittsburgh, USA No difference No difference No difference NA Raveh-Sadka,
analysis 2015 [77]
454-pyrosequencing; V1–V3 18/35 Stool; weekly until 3 NICUs, Florida, USA Proteobacteria and Actinobacteria No difference Klebsiella granulomatis, No Torrazza, 2013 [78]
Bifidobacterium NEC onset increase; Bacteroidetes decrease Klebsiella difference
qPCR pneumoniae,
Clostridium
perfringens and

Clostridium butyricum antagonist effects


Staphylococcus
epidermidis more
frequent in NEC
samples
PCR-DGGE V3 8/17 Stool samples before 1 NICU, Newcastle, UK No difference Enterobacter, Flavobacterium, NA NA Stewart, 2012 [79]
and after NEC onset Propionibacterium and
Staphylococcus more
frequent
PSQMA- V1 24/0 Intestinal samples 1 NICU, Copenhagen, Proteobacteria (49.0%), Firmicutes Ralstonia most frequent; Clostridium butyricum NA Smith, 2011 [39]
pyrosequencing; Denmark (30.4%), Actinobacteria (17.1%) Clostridium in 4 samples and Clostridium
FISH and Bacteroidetes (3.6%) paraputrificum with
high density in 2
NEC samples with
Pneumatosis
intestinalis
PCR and T-RFLP V1–V6 10/10 <1 day after NEC 1 NICU, Chicago, USA Proteobacteria increase; Firmicutes Increased abundance of NA Lower Wang, 2009 [80]
analysis onset decrease Gammaproteobacteria
PCR-TTGE — 3/9 Stool samples at 7 1 NICU, Nantes, France NA Clostridium only in patients NA Lower De la Cochetière,
and 14 days of life with NEC 2004 [81]

DGEE, denaturing gradient gel electrophoresis; FISH, fluorescence in situ hybridization; NA, not available; NICU, neonatal intensive-care unit; OTU, operational taxonomic unit; qPCR, real-time PCR; T-RFLP, terminal restriction fragment length
polymorphism; TTGE, temporal temperature gradient electrophoresis.

41
42 Clinical Microbiology and Infection, Volume 22 Number 1, January 2016 CMI

TABLE 2. Gut microbiota profile associated with necrotizing enterocolitis (NEC) (case–control studies), analysed with
culture-dependent methods

Methods for Anaerobic Number of


bacterial culture patients Sample Genus/family Species
identification conditions (NEC/controls) collection Location level level Reference

MALDI-TOF MS; Anaerobic chamber; 15/15 <1 day after 4 NICUs, No difference Clostridium butyricum Cassir, 2015 [17]
16S rRNA PCR anaerobic blood NEC onset South-eastern (14 vs. 2)
culture bottles; France
anaerobic agar
MALDI-TOF MS Anaerobic conditions 12/44 1 stool sample per 1 NICU, London, — Clostridium perfringens Sim, 2014 [46]
(unspecified); alcohol week until NEC UK type A (β2 toxin gene)
shock; anaerobic agar onset in 4 NEC patients
API kit Anaerobic conditions 8/17 Stool samples 1 NICU, Newcastle, — CoNS more frequent Stewart,
(unspecified); before and after UK 2012 [79]
anaerobic agar NEC onset
API kit, gas Anaerobic conditions 24/41 <1 day after NEC 1 NICU, Rotterdam, Increased No difference Westra-Meijer,
chromatography (unspecified); onset The Netherlands abundance 1983 [82]
anaerobic agar of Klebsiella

CoNS, coagulase-negative staphylococci; NICU, neonatal intensive-care unit; MALDI-TOF MS, matrix-assisted laser desorption ionization time-of-flight mass spectrometry.

FIG. 1. The ambiguous roles of Clostridium butyricum in its host. Some C. butyricum strains have been reported to be pathogenic, expressing virulence
factors (i.e. toxins such as enterotoxins or botulinum neurotoxin; enzymes such as neuraminidase; adhesion molecules; and secretion of high levels of
butyric acid). Other C. butyricum strains have been reported to be beneficial, expressing host protective factors (i.e. bacteriocins such as butyricin,
pathogen inhibition, and secretion of butyric acid).
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
CMI Cassir et al. Clostridium butyricum antagonist effects 43

colonies by the use of matrix-assisted laser desorption ionization Bacteriol. The Firmicutes. 9th ed., vol. 3. New-York: Springer; 2009. p.
739–40.
time-of-flight mass spectrometry or 16S rRNA PCR) provided
[2] Ghoddusi HB, Sherburn R. Preliminary study on the isolation of Clos-
results comparable to those obtained with molecular methods, tridium butyricum strains from natural sources in the UK and screening
in terms of diversity [16]. However, the 15% overlap found in the isolates for presence of the type E botulinal toxin gene. Int J Food
the inaugural study demonstrates that, like molecular techniques, Microbiol 2010;142:202–6.
[3] Mountzouris KC, McCartney AL, Gibson GR. Intestinal microflora of
culture is far from identifying the totality of gut bacteria, and that human infants and current trends for its nutritional modulation. Br J
complementary approaches are therefore required. Nutr 2002;87:405–20.
Cultivation of strictly anaerobic species is challenging. [4] Ferraris L, Butel M-J, Aires J. Antimicrobial susceptibility and resistance
determinants of Clostridium butyricum isolates from preterm infants. Int
Although they are thought to be the most common microor-
J Antimicrob Agents 2010;36:420–3.
ganisms in the human microbiota, many anaerobic bacteria [5] Sato R, Tanaka M. Intestinal distribution and intraluminal localization of
detected in stool samples with metagenomic methods remain orally administered Clostridium butyricum in rats. Microbiol Immunol
1997;41:665–71.
uncultured [68]. In particular, only a few studies analysing the gut
[6] Fenicia L, Anniballi F, Aureli P. Intestinal toxemia botulism in Italy,
bacterial profile associated with NEC have used strategies to 1984–2005. Eur J Clin Microbiol Infect Dis 2007;26:385–94.
optimize the growth of strict anaerobic bacteria (i.e. heat shock [7] Howard FM, Flynn DM, Bradley JM, Noone P, Szawatkowski M.
for sporulating anaerobes, direct inoculation in anaerobic culture Outbreak of necrotising enterocolitis caused by Clostridium butyricum.
Lancet 1977;2:1099–102.
bottles, selective culture media, and the use of an anaerobic [8] Popoff MR, Bouvet P. Clostridial toxins. Future Microbiol 2009;4:
chamber) (Table 2). Promising strategies, such as a reduction in 1021–64.
the sample transport time before inoculation and the use of [9] Popoff MR, Bouvet P. Genetic characteristics of toxigenic clostridia and
toxin gene evolution. Toxicon 2013;75:63–89.
antioxidant agents such as ascorbic acid and glutathione, will
[10] Rousseau C, Poilane I, De Pontual L, Maherault A-C, Le Monnier A,
dramatically improve the culture of anaerobic bacteria, including Collignon A. Clostridium difficile carriage in healthy infants in the
potential new emerging enteropathogens [69]. community: a potential reservoir for pathogenic strains. Clin Infect Dis
2012;55:1209–15.
[11] Gerding DN, Meyer T, Lee C, Cohen SH, Murthy UK, Poirier A, et al.
Administration of spores of nontoxigenic Clostridium difficile strain M3
Conclusions
for prevention of recurrent C. difficile infection: a randomized clinical
trial. JAMA 2015;313:1719–27.
[12] Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ,
C. butyricum has various implications for human health, from Brummer R-J. The role of butyrate on colonic function. Aliment
beneficial to pathogenic (Fig. 1). Whereas a non-toxigenic strain Pharmacol Ther 2008;27:104–19.
has been validated in clinical practice for its probiotic properties, [13] Gantois I, Ducatelle R, Pasmans F, Haesebrouck F, Hautefort I,
Thompson A, et al. Butyrate specifically down-regulates Salmonella
other strains have been linked with pathological conditions such pathogenicity island 1 gene expression. Appl Environ Microbiol
as botulism in infants or NEC in preterm neonates. As toxigenic 2006;72:946–9.
and non-toxigenic clostridia form part of the normal gut [14] Polage CR, Solnick JV, Cohen SH. Nosocomial diarrhea: evaluation and
treatment of causes other than Clostridium difficile. Clin Infect Dis
microbiota, it remains challenging to understand the triggers of
2012;55:982–9.
the expression of their beneficial or virulence factors. The fact [15] Lagier J-C, Million M, Hugon P, Armougom F, Raoult D. Human gut
that different strains within the same species may have antagonist microbiota: repertoire and variations. Front Cell Infect Microbiol
2012;2:136.
effects on human health reinforces the need to accurately
[16] Lagier J-C, Armougom F, Million M, Hugon P, Pagnier I, Robert C, et al.
identify gut bacterial species at the strain level. To date, bacterial Microbial culturomics: paradigm shift in the human gut microbiome
culture is the only method that fulfils this criterion, which is also study. Clin Microbiol Infect 2012;18:1185–93.
essential to discover new emerging enteropathogenic strains. [17] Cassir N, Benamar S, Bou Khalil J, Croce O, Saint-Faust M, Jacquot A,
et al. Clostridium butyricum strains and dysbiosis linked to necrotizing
enterocolitis in preterm neonates. Clin Infect Dis 2015;61:1107–15.
[18] Sanders ME. Probiotics: definition, sources, selection, and uses. Clin
Transparency declaration Infect Dis 2008;46(Suppl. 2):S58–61. discussion S144–51.
[19] Hayashi A, Sato T, Kamada N, Mikami Y, Matsuoka K, Hisamatsu T,
et al. A single strain of Clostridium butyricum induces intestinal IL-10-
The authors declare that they have no conflicts of interest. producing macrophages to suppress acute experimental colitis in
mice. Cell Host Microbe 2013;13:711–22.
[20] Kong Q, He G-Q, Jia J-L, Zhu Q-L, Ruan H. Oral administration of
References Clostridium butyricum for modulating gastrointestinal microflora in mice.
Curr Microbiol 2011;62:512–7.
[21] Kashiwagi I, Morita R, Schichita T, Komai K, Saeki K, Matsumoto M,
et al. Smad2 and Smad3 inversely regulate TGF-β autoinduction in
[1] Vos P, Garrity G, Jones D, Krieg NR, Ludwig W, Rainey FA, et al.
Clostridium butyricum-activated dendritic cells. Immunity 2015.
Genus I. Clostridium Prazmowski, 1880. Bergey’s Manual of Systematic

Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
44 Clinical Microbiology and Infection, Volume 22 Number 1, January 2016 CMI

[22] Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, [43] Hutto DL, Wannemuehler MJ. A comparison of the morphologic ef-
et al. Treg induction by a rationally selected mixture of Clostridia fects of Serpulina hyodysenteriae or its beta-hemolysin on the murine
strains from the human microbiota. Nature 2013;500:232–6. cecal mucosa. Vet Pathol 1999;36:412–22.
[23] Seo M, Inoue I, Tanaka M, Matsuda N, Nakano T, Awata T, et al. [44] Popoff MR, Szylit O, Ravisse P, Dabard J, Ohayon H. Experimental
Clostridium butyricum MIYAIRI 588 improves high-fat diet-induced non- cecitis in gnotoxenic chickens monoassociated with Clostridium butyr-
alcoholic fatty liver disease in rats. Dig Dis Sci 2013;58:3534–44. icum strains isolated from patients with neonatal necrotizing entero-
[24] Woo TDH, Oka K, Takahashi M, Hojo F, Osaki T, Hanawa T, et al. colitis. Infect Immun 1985;47:697–703.
Inhibition of the cytotoxic effect of Clostridium difficile in vitro by [45] Butel MJ, Roland N, Hibert A, Popot F, Favre A, Tessedre AC, et al.
Clostridium butyricum MIYAIRI 588 strain. J Med Microbiol 2011;60: Clostridial pathogenicity in experimental necrotising enterocolitis in
1617–25. gnotobiotic quails and protective role of bifidobacteria. J Med Micro-
[25] Takahashi M, Taguchi H, Yamaguchi H, Osaki T, Komatsu A, Kamiya S. biol 1998;47:391–9.
The effect of probiotic treatment with Clostridium butyricum on [46] Sim K, Shaw AG, Randell P, Cox MJ, McClure ZE, Li M-S, et al. Dys-
enterohemorrhagic Escherichia coli O157:H7 infection in mice. FEMS biosis anticipating necrotizing enterocolitis in very premature infants.
Immunol Med Microbiol 2004;41:219–26. Clin Infect Dis 2014;60:389–97.
[26] Wang F-Y, Liu J-M, Luo H-H, Liu A-H, Jiang Y. Potential protective [47] Szylit O, Butel MJ, Rimbault A. An experimental model of necrotising
effects of Clostridium butyricum on experimental gastric ulcers in mice. enterocolitis. Lancet 1997;350:33–4.
World J Gastroenterol 2015;21:8340–51. [48] Thymann T, Møller HK, Stoll B, Støy ACF, Buddington RK, Bering SB,
[27] Seki H, Shiohara M, Matsumura T, Miyagawa N, Tanaka M, et al. Carbohydrate maldigestion induces necrotizing enterocolitis in
Komiyama A, et al. Prevention of antibiotic-associated diarrhea in preterm pigs. Am J Physiol Gastrointest Liver Physiol 2009;297:
children by Clostridium butyricum MIYAIRI. Pediatr Int 2003;45:86–90. G1115–25.
[28] Clarke DJ, Morris JG. Butyricin 7423: a bacteriocin produced by [49] Peng L, He Z, Chen W, Holzman IR, Lin J. Effects of butyrate on in-
Clostridium butyricum NCIB7423. J Gen Microbiol 1976;95:67–77. testinal barrier function in a Caco-2 cell monolayer model of intestinal
[29] Nakanishi S, Tanaka M. Sequence analysis of a bacteriocinogenic barrier. Pediatr Res 2007;61:37–41.
plasmid of Clostridium butyricum and expression of the bacteriocin gene [50] Bousseboua H, Le Coz Y, Dabard J, Szylit O, Raibaud P, Popoff MR,
in Escherichia coli. Anaerobe 2010;16:253–7. et al. Experimental cecitis in gnotobiotic quails monoassociated with
[30] Gebhart D, Williams SR, Bishop-Lilly KA, Govoni GR, Willner KM, Clostridium butyricum strains isolated from patients with neonatal
Butani A, et al. Novel high-molecular-weight, R-type bacteriocins of necrotizing enterocolitis and from healthy newborns. Infect Immun
Clostridium difficile. J Bacteriol 2012;194:6240–7. 1989;57:932–6.
[31] Waligora-Dupriet AJ, Dugay A, Auzeil N, Nicolis I, Rabot S, [51] Popoff MR, Dodin A. Survey of neuraminidase production by Clos-
Huerre MR, et al. Short-chain fatty acids and polyamines in the path- tridium butyricum, Clostridium beijerinckii, and Clostridium difficile strains
ogenesis of necrotizing enterocolitis: kinetics aspects in gnotobiotic from clinical and nonclinical sources. J Clin Microbiol 1985;22:873–6.
quails. Anaerobe 2009;15:138–44. [52] Azcarate-Peril MA, Foster DM, Cadenas MB, Stone MR, Jacobi SK,
[32] Dykes JK, Luquez C, Raphael BH, McCroskey L, Maslanka SE. Labo- Stauffer SH, et al. Acute necrotizing enterocolitis of preterm piglets is
ratory investigation of the first case of C. butyricum type E botulism in characterized by dysbiosis of ileal mucosa-associated bacteria. Gut
the United States. J Clin Microbiol 2015;53:3363–5. Microbes 2011;2:234–43.
[33] Hill KK, Xie G, Foley BT, Smith TJ, Munk AC, Bruce D, et al. [53] Alfa MJ, Robson D, Davi M, Bernard K, Van Caeseele P, Harding GKM.
Recombination and insertion events involving the botulinum neuro- An outbreak of necrotizing enterocolitis associated with a novel
toxin complex genes in Clostridium botulinum types A, B, E and F and Clostridium species in a neonatal intensive care unit. Clin Infect Dis
Clostridium butyricum type E strains. BMC Biol 2009;7:66. 2002;35:S101–5.
[34] Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med 2011;364: [54] Govind R, Dupuy B. Secretion of Clostridium difficile toxins A and B
255–64. requires the holin-like protein TcdE. PLoS Pathog 2012;8:e1002727.
[35] Waligora-Dupriet A-J, Dugay A, Auzeil N, Huerre M, Butel M-J. Evi- [55] Gardner EM, Kestler M, Beieler A, Belknap RW. Clostridium butyricum
dence for clostridial implication in necrotizing enterocolitis through sepsis in an injection drug user with an indwelling central venous
bacterial fermentation in a gnotobiotic quail model. Pediatr Res catheter. J Med Microbiol 2008;57:236–9.
2005;58:629–35. [56] Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L,
[36] Gorham P, Millar M, Godwin PG. Clostridial hand-carriage and Sargent M, et al. Diversity of the human intestinal microbial flora.
neonatal necrotising enterocolitis. J Hosp Infect 1988;12:139–41. Science 2005;308:1635–8.
[37] Book LS, Overall JC, Herbst JJ, Britt MR, Epstein B, Jung AL. Clustering [57] Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al.
of necrotizing enterocolitis. Interruption by infection-control mea- A human gut microbial gene catalogue established by metagenomic
sures. N Engl J Med 1977;297:984–6. sequencing. Nature 2010;464:59–65.
[38] Sturm R, Staneck JL, Stauffer LR, Neblett 3rd WW. Neonatal necro- [58] Kalia VC, Mukherjee T, Bhushan A, Joshi J, Shankar P, Huma N.
tizing enterocolitis associated with penicillin-resistant, toxigenic Clos- Analysis of the unexplored features of rrs (16S rDNA) of the genus
tridium butyricum. Pediatrics 1980;66:928–31. Clostridium. BMC Genomics 2011;12:18.
[39] Smith B, Bodé S, Petersen BL, Jensen TK, Pipper C, [59] Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for
Kloppenborg J, et al. Community analysis of bacteria colonizing rapid assignment of rRNA sequences into the new bacterial taxonomy.
intestinal tissue of neonates with necrotizing enterocolitis. BMC Appl Environ Microbiol 2007;73:5261–7.
Microbiol 2011;11:73. [60] Hugon P, Lagier J-C, Robert C, Lepolard C, Papazian L, Musso D, et al.
[40] Kwok JSL, Ip M, Chan T-F, Lam W-Y, Tsui SKW. Draft genome Molecular studies neglect apparently gram-negative populations in the
sequence of Clostridium butyricum strain NOR 33234, isolated from an human gut microbiota. J Clin Microbiol 2013;51:3286–93.
elderly patient with diarrhea. Genome Announc 2014;2:6. [61] Chakravorty S, Helb D, Burday M, Connell N, Alland D. A detailed
[41] Hyatt DR, Joens LA. Analysis of the lytic activity of the Serpulina hyo- analysis of 16S ribosomal RNA gene segments for the diagnosis of
dysenteriae hemolysin. Infect Immun 1997;65:4877–9. pathogenic bacteria. J Microbiol Methods 2007;69:330–9.
[42] Hsu T, Hutto DL, Minion FC, Zuerner RL, Wannemuehler MJ. Cloning [62] Gao Z, Perez-Perez GI, Chen Y, Blaser MJ. Quantitation of major
of a beta-hemolysin gene of Brachyspira (Serpulina) hyodysenteriae and human cutaneous bacterial and fungal populations. J Clin Microbiol
its expression in Escherichia coli. Infect Immun 2001;69:706–11. 2010;48:3575–81.
Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45
CMI Cassir et al. Clostridium butyricum antagonist effects 45

[63] Raoult D. Emerging clones of bacterial epidemics in the genomic area. health or necrotizing enterocolitis in preterm infants. Microbiome
Clin Microbiol Infect 2014;20:371–2. 2013;1:20.
[64] Vartoukian SR, Palmer RM, Wade WG. Strategies for culture of [74] Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z, Wang B,
‘unculturable’ bacteria. FEMS Microbiol Lett 2010;309:1–7. et al. Early microbial and metabolomic signatures predict later onset of
[65] Finegold SM, Attebery HR, Sutter VL. Effect of diet on human fecal necrotizing enterocolitis in preterm infants. Microbiome 2013;1:13.
flora: comparison of Japanese and American diets. Am J Clin Nutr [75] Mai V, Young CM, Ukhanova M, Wang X, Sun Y, Casella G, et al. Fecal
1974;27:1456–69. microbiota in premature infants prior to necrotizing enterocolitis. PloS
[66] Hayashi H, Sakamoto M, Benno Y. Phylogenetic analysis of the human One 2011;6:e20647.
gut microbiota using 16S rDNA clone libraries and strictly anaerobic [76] Mshvildadze M, Neu J, Shuster J, Theriaque D, Li N, Mai V. Intestinal
culture-based methods. Microbiol Immunol 2002;46:535–48. microbial ecology in premature infants assessed with non-culture-
[67] Goodman AL, Kallstrom G, Faith JJ, Reyes A, Moore A, Dantas G, et al. based techniques. J Pediatr 2010;156:20–5.
Extensive personal human gut microbiota culture collections charac- [77] Raveh-Sadka T, Thomas BC, Singh A, Firek B, Brooks B, Castelle CJ,
terized and manipulated in gnotobiotic mice. Proc Natl Acad Sci USA et al. Gut bacteria are rarely shared by co-hospitalized premature
2011;108:6252–7. infants, regardless of necrotizing enterocolitis development. eLife
[68] Lagier J-C, Hugon P, Khelaifia S, Fournier P-E, La Scola B, Raoult D. The 2015;4.
rebirth of culture in microbiology through the example of culturomics to [78] Torrazza RM, Neu J. The altered gut microbiome and necrotizing
study human gut microbiota. Clin Microbiol Rev 2015;28:237–64. enterocolitis. Clin Perinatol 2013;40:93–108.
[69] La Scola B, Khelaifia S, Lagier J-C, Raoult D. Aerobic culture of [79] Stewart C, Marrs E, Magorrian S, Nelson A, Lanyon C, Perry J, et al.
anaerobic bacteria using antioxidants: a preliminary report. Eur J Clin The preterm gut microbiota: changes associated with necrotising
Microbiol Infect Dis 2014;33:1781–3. enterocolitis and infection. Acta Paediatr 2012;101:1121–7.
[70] Zhou Y, Shan G, Sodergren E, Weinstock G, Walker WA, [80] Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, et al. 16S
Gregory KE. Longitudinal analysis of the premature infant intestinal rRNA gene-based analysis of fecal microbiota from preterm infants
microbiome prior to necrotizing enterocolitis: a case–control study. with and without necrotizing enterocolitis. ISME J 2009;3:944–54.
PloS One 2015;10:e0118632. [81] de la Cochetiere M-F, Piloquet H, des Robert C, Darmaun D,
[71] McMurtry VE, Gupta RW, Tran L, Blanchard EE, Penn D, Taylor CM, Galmiche J-P, Roze J-C. Early intestinal bacterial colonization and
et al. Bacterial diversity and clostridia abundance decrease with necrotizing enterocolitis in premature infants: the putative role of
increasing severity of necrotizing enterocolitis. Microbiome 2015;3:11. Clostridium. Pediatr Res 2004;56:366–70.
[72] Brower-Sinning R, Zhong D, Good M, Firek B, Baker R, Sodhi CP, et al. [82] Westra-Meijer CM, Degener JE, Dzoljic-Danilovic G, Michel MF,
Mucosa-associated bacterial diversity in necrotizing enterocolitis. PloS Mettau JW. Quantitative study of the aerobic and anaerobic faecal
One 2014;9:e105046. flora in neonatal necrotising enterocolitis. Arch Dis Child 1983;58:
[73] Claud EC, Keegan KP, Brulc JM, Lu L, Bartels D, Glass E, et al. Bacterial 523–8.
community structure and functional contributions to emergence of

Clinical Microbiology and Infection © 2015 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved, CMI, 22, 37–45

Vous aimerez peut-être aussi