Next Article in Journal
Impact of Escherichia coli Outer Membrane Vesicles on Sperm Function
Next Article in Special Issue
Antibiotic Resistance Profile of RT 027/176 Versus Other Clostridioides difficile Isolates in Silesia, Southern Poland
Previous Article in Journal
From Alpha to Delta—Genetic Epidemiology of SARS-CoV-2 (hCoV-19) in Southern Poland
Previous Article in Special Issue
Risk Factors for Hospital Readmission for Clostridioides difficile Infection: A Statewide Retrospective Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota Composition Associated with Clostridioides difficile Colonization and Infection

1
Laboratory of Food Microbiology, Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
2
Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29590 Málaga, Spain
*
Author to whom correspondence should be addressed.
Pathogens 2022, 11(7), 781; https://doi.org/10.3390/pathogens11070781
Submission received: 7 June 2022 / Revised: 1 July 2022 / Accepted: 5 July 2022 / Published: 8 July 2022
(This article belongs to the Special Issue Advanced Research on Clostridium difficile)

Abstract

:
Clostridioides difficile is an anaerobic Gram-positive and spore-forming bacterium. The majority of C. difficile strains produce two toxins, A and B, associated with the development of acute diarrhea and/or colitis. In this review, two situations are distinguished: C. difficile infection (CDI) and asymptomatic colonization (AC). The main objective of this review is to explore the available data related to the link between the gut microbiota and the development of CDI. The secondary aim is to provide more information on why some people colonized with toxigenic C. difficile develop an infection while others show no signs of disease. Several factors, such as the use of antibiotics and proton pump inhibitors, hospitalization, and age, predispose individuals to C. difficile colonization and/or C. difficile infection. The gut microbiota of people with AC showed decreased abundances of Prevotella, Alistipes, Bacteroides, Bifidobacterium, Dorea, Coprococcus, and Roseburia. The gut microbiota of people suffering from CDI showed reductions in the abundances of Lachnospiraceae, Ruminococcaceae, Blautia spp., Prevotella spp., Dialister spp., Bifidobacterium spp., Roseburia spp., Anaerostipes spp., Faecalibacterium spp. and Coprococcus spp., in comparison with healthy people. Furthermore, increases in the abundances of Enterococcaceae and Enterococcus were associated with C. difficile infection.

1. Introduction

Clostridioides difficile is an anaerobic, Gram-positive, and spore-forming bacterium recognized as the leading cause of health care-associated diarrhea. In the United States of America (USA), in 2017, the Centers for Disease Control and Prevention (CDC) considered C. difficile infection (CDI) to be a major health threat (with 223,900 national cases) among hospitalized patients, eventually leading to 12,800 deaths [1]. In France, in 2016, CDI incidence in acute care was estimated to be 3.6 cases per 10,000 patient days [2]. In 2016, a total of 7711 CDI cases were reported to the ECDC (European Centre for Disease Prevention and Control) in Europe (20 EU countries), of which 74.7% were associated with health care settings [3].
In this review, two situations are distinguished in order to understand the trigger symptoms: CDI and AC of C. difficile. CDI is defined by the presence of diarrhea and at least one other following criterion: the carriage of a toxigenic strain of C. difficile, the presence of toxins in the stool and/or a colonoscopy result showing pseudomembranous colitis. AC by C. difficile can be defined as the absence of diarrhea with at least one other criterion: carrying a strain of C. difficile and/or the presence of toxins in the stool [4]. Crobach et al., defined AC as the presence of C. difficile but without symptoms of CDI.
The main risks factors are PPI use, antibiotics use, corticoid use, hospital stay and age.
PPI use is a risk factor for rCDI (recurrent CDI) [5]. Their use increased stomach pH from 1.2 to 5, raising the possibility of developing CDI or even carrying the bacterium asymptomatically compared with subjects without this treatment [6]. The increase in stomach pH to a value of 5 during digestion does not influence the resistance of the spores of C. difficile, as they are able to survive at a normal stomach pH [6,7,8]. The vegetative cells of C. difficile can survive in the gastric content only if the pH is equal to or greater than 5 [7].
The use of antibiotics is a well-known risk factor for C. difficile asymptomatic colonization or infection due to modification of the gut microbiota. Most of these cases are associated with the use of four antibiotics: clindamycin, cephalosporins, carbapenem and fluoroquinolone [9,10,11]. Other antibiotics, including macrolides, sulfonamide, trimethoprim and penicillin, are less associated with CDI [9,10,11]. Antibiotic exposure increases the possibility of C. difficile colonization by 3.7-fold [12] and of developing CDI by 3.55-fold [9]. A previous study showed that among antibiotic-associated diarrhea (AAD) cases, the incidence of CDI was 1.14–1.89% [13], and the frequency of toxigenic C. difficile carriage was 18.1–19.0% [13].
The use of corticosteroids increases the risk of C. difficile colonization in adults admitted to the hospital [14] and immunosuppressive therapy is a risk factor for complicated CDI [15]. Immunocompromised patients have a higher risk of developing rCDI during hospitalization [16].
A recent hospitalization or a recent intensive care unit stay increases the risk of developing CDI by 2.2 and 6.5, respectively [11]. Hospitalization in the previous 6 months increases the risk of colonization by 2.18 [14]. Previous studies showed that in an ambulatory group (n = 43), in patients with short hospital stays (n = 48) and in patients with long hospital stays (n = 102), the percentages of C. difficile carriage were 9.5%, 8% and 13%, respectively [17]. CDC hospitalization rates are significantly higher among those 65 years of age and older (by 4-fold) and those over 85 years of age (by 10-fold) compared to those under 65 years [18,19].
The percentage of AC evolves as a function of age. It is high in the first months of life and decreases until adult age, and then it increases with advancing age. The percentage of individuals with AC over time is shown in Figure 1 [4,20,21,22,23,24,25,26,27]. Patients aged >65 years old have a 10-fold higher risk of developing CDI than patients in the other age groups [28,29].
There are three lines of defense against pathogens: the epithelial barrier, innate immunity, and adaptive immunity. The first step is intestinal colonization by C. difficile. Once the bacterium can produce toxins, these toxins transgress the epithelial barrier through the activation of Rho glycosylation, which causes disruption of tight junctions. Secondly, pathogen-associated molecular patterns (PAMPs) are recognized by pattern recognition receptors (PRRs). This interaction induces a rapid innate immunity response. Finally, adaptive immunity provides a highly specific immune response against C. difficile [30].
The intestinal mucus is composed of two types of mucins: MUC1 (cell-surface) and MUC2 (secreted forms). Secreted MUC2 is found mostly in the feces of healthy people, while people who suffer from CDI have an imbalanced mucus composition; their stool mucus is composed mainly of MUC1, with significantly decreased MUC2 levels [31]. They also present an increase in terminal galactose residues (a known receptor for C. difficile toxin A in mice, hamster, rabbits and pigs) and N-acetyl glucosamine (GlcNAc) [31] and a decrease in N-acetyl galactosamine (GalNAc).
Essential to C. difficile spore germination is the presence of primary bile acids (PBAs). PBAs is produced by the liver, is discharged into the small intestine and helps with fatty digestion [20]. PBAs stimulates germination of C. difficile spores [32], and secondary bile acids (SBAs) inhibit germination [32].
Several studies have tried to understand the C. difficile pathogenesis in order of reduce the risks of development of the disease and find new therapeutic strategy. The animal experimentations using hamster have allowed to test the transplantation fecal efficiency, the use of non-toxigenic strain of C. difficile and use of monoclonal antibodies against toxin A and toxin B [33,34,35]. The piglet model of CDI is representative of the key characteristics of human CDI and helped to understand the virulence and new treatment [36]. Then, C. difficile studies use different methods in vitro in order to limit using animal experimentation: feces cultures [37], the continuous culture model [38,39], the triple-stage chemostat human gut model [40,41], and the Tim-2 model [42].
The main objective of this review is to explore the available data about the link between the gut microbiota and the development of CDI. The secondary aim is to provide more information on why some people colonized with toxigenic C. difficile develop CDI and others show no signs of disease.

2. Microbiota Associated with Asymptomatic Colonization and CDI

2.1. Composition of the Normal Human Gut Microbiota

The composition of the gut microbiota is influenced by diet, age, the use of antibiotics, etc. [43]. In a normal gut, the dominant phyla are Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucomicrobia. Firmicutes and Bacteroidetes represent 90% of the gut microbiota [44,45]. Gut Bacteroidetes is mainly composed of two genera, Bacteroides and Prevotella [44,45]. The main genera in the Firmicutes phylum are Lactobacillus, Bacillus, Clostridium, Enterococcus, Faecalibacterium, Roseburia, and Ruminococcus. The Actinobacteria phylum is represented mainly by the Bifidobacterium genus [44,45]. The Metahit consortium classified human fecal metagenomic samples from three continents into three groups called “enterotypes”: Bacteroides (enterotype I), Prevotella (enterotype II) and Ruminococcus (enterotype III) [45,46]. The enterotype concept is very controversial in the scientific community [46]. Gorvitovskaia et al., chose to validate the first two enterotypes (Prevotella and Bacteroides) [47], while Cheng et al., have shown that enterotypes are not constant over time and are influenced by age and diet [43].
The gut microbiota of infants is rich in Bifidobacterium spp. and the gut microbiota of elderly individuals has decreased proportions of Ruminococcaceae, Bifidobacterium, Lactobacillus and Faecalibacterium and increased proportions of Proteobacteria, Bacteroidetes and Clostridium spp. [44,48,49].

2.2. Factors Influencing the Healthy Gut Microbiota

The use of proton pump inhibitors (PPIs) influences the pH of the stomach and therefore the gut microbiota. The use of PPIs decreased gut microbial diversity and the abundances of Clostridiales and Ruminococcaceae [50,51] and increased the abundances of the Enterococcaceae and Staphylococcaceae families [50,52] and Veillonella parvula and Streptococcus mutans [53]. These modifications of the gut microbiota are strongly correlated with CDI development.
In human feces, clindamycin, cephalosporins and fluoroquinolone treatments impact the microbiota, resulting in an increase in Enterococcus abundance [54]. These three categories of antibiotics induce a reduction in the abundances of Streptococcus spp., Anaerococcus spp., Peptoniphilus spp., Porphyromonas spp. and Prevotella spp., and increase the abundance of Sphingomonas spp. [54]. Antibiotics also seem to decrease the proportions of Ruminococcaceae, Lachnospiraceae and Bifidobacterium and to increase the proportions of Lactobacilliaceae and Streptococcaceae [49]. Werkhoven and collaborators (2021) showed that the carriage of toxigenic C. difficile increased the incidence of developing CDI 10-fold after antibiotic treatment. Specifically, the use of carbapenem increased the incidence of CDI 5-fold and increased the abundance of Enterococcus 5-fold [13].
The age-modified gut microbiota. The gut microbiota of elderly individuals has decreased proportions of Ruminococcaceae, Bifidobacterium, Lactobacillus and Faecalibacterium and increased proportions of Proteobacteria, Bacteroidetes and Clostridium spp. [44,48,49]. Two interesting recent studies have addressed the gut microbiota composition in hospitalized elderly patients with CDI, showing lower microbial diversity, lower proportions of gut commensals with putative functions and a reduction in butyrate-producing bacteria in CDI samples [55,56].
The state of dysbiosis can be defined as a decrease in the obligate anaerobic bacteria and an increase in the relative abundance of facultative anaerobic bacteria, such as Enterobacteriaceae [57,58], a decrease in microbial diversity and a decrease in anti-inflammatory species such Faecalibacterium prausnitzii [59]. In Inflammatory Bowel Disease (IBD), a decrease in microbial diversity, a decrease in F. prausnitzii and an increase in Streptococcus and Escherichia/Shigella are observed [59,60].

2.3. Composition of Microbiota among Patients with AC

As previously described, AC by C. difficile can be defined as the absence of diarrhea with at least one other criterion: carrying a strain of C. difficile and/or the presence of toxins in the stool [4]. In the literature, few studies differentiate between AC by C. difficile and CDI. The composition of the microbiota of patients with AC was similar to that of the control group and included the phyla Bacteroidetes (40.95%), Firmicutes (36.23%), and Proteobacteria (15.73%) [61].
Within the phylum Bacteroidetes, decreases in two families (Bacteroidaceae and Prevotellaceae) were observed [17]. Zhang et al., showed decreases in the AC of Prevotella spp., Alistipes spp., Bacteroides spp. and an increase in the AC of Parabacteroides spp. [61].
In the phylum Firmicutes, increases in the abundances of Ruminococcaceae, Erysipelotrichaceae and Clostridiaceae and decreases in the abundances of Leuconostocacceae and Erysipelothrichaceae were observed [17]. Zhang et al., showed decreases in AC by Dorea spp., Coprococcus spp., and Roseburia spp. [61] and increases in AC by Lactobacillus spp. [61], Enterococcus spp. [51] and Oscillospira spp. [62]. Another study showed increases in AC by Blautia spp., Flavonifractor spp., and Lachnospiraceae_unclassified [63].
Within the phylum Proteobacteria, several studies showed an increase in AC by Enterococcus spp. and Klebsiella spp. [61]. In the phylum Actinobacteria, Zhang et al., showed a decrease in AC by Bifidobacterium spp. [61]. Within the phylum Verrucomicrobia, a decrease in AC by Akkermansia spp. was observed [63].
Some studies showed no differences in microbial diversity between an AC group and a healthy group (HG) who presented a negative stool test for C. difficile [63]; however, some studies showed that the microbial richness (Chao index) decreased in patients with AC compared with patients in the HG [61].

2.4. Microbiota Composition of Adults Suffering from CDI

Many studies have described the gut microbiota composition of patients with CDI. In Table 1, the modifications of gut microbiota with increases (red) and decreases (green) in the abundances of various phyla and genera when a patient is suffering from CDI are represented. These data are from the original research publications.
The prevalence of the phylum Proteobacteria is increased in adults with CDI [61,62,64,65,66]. The main family responsible for this increase is Enterobacteriaceae [62,66,67,68,69], and the main genera exhibiting increases are Klebsiella spp. [56,61,62,66,70], Escherichia/Shigella [56,61,64,66,69,70,71], Proteus spp. [56,66] and Providencia spp. [66].
The prevalence of the phylum Firmicutes is decreased in adults with CDI [61,62,65]. The main families responsible for this decrease are Lachnospiraceae and Ruminococcaceae [54,56,62,66,67,72], and the main genera are Blautia spp. [54,61,64,66,70,73,74], Roseburia spp. [54,61,64,66,74], Anaerostipes spp. [62,64,74], Faecalibacterium spp. [54,56,61,62,64,66,69,70,74] Collinsella spp. [56,64,66,69,70] and Coprococcus spp. [61,62,69,70,74]. Some of the genera that have been shown to exhibit increases are Enterococcus spp. [54,56,61,62,69,70,71], Veillonella spp. [56,61,62,74] and Lactobacillus spp. [61,70,71,74]. Metabolization of PBAs to SBAs is provided by populations from the Firmicutes phylum: Ruminococcaceae, Blautia and Lachnospiraceae [20,75]. The decreases in Lachnospiraceae and Ruminococcaceae also lead to decreases in the concentration of SCFA and the transformation of SBAs from PBAs, providing advantages to these bacteria [20,56,62,64,67]. If the abundances of these bacterial groups decrease, there is a decrease in SBAs and therefore a decrease in the inhibition of germination in the ileum. This decrease in SBAs facilitates the development of CDI. Clostridium scindens is able to restore SBAs metabolism and inhibit the germination of C. difficile [20,32,76].
Regarding the phylum Bacteroidetes, some studies showed a general decrease in abundance [61,62,67] while others showed an increase [64,68]. Depending on the studies, the abundance of the genus Bacteroides spp. has been shown to decrease [54,56,61,71] or increase [62,64,74]. Only one study showed that the abundance of Parabacteroides spp. increased [61]. Some genera, such as Prevotella spp. [54,61,62,64,70,73], Paraprevotella spp. [66], Alistipes spp. [54,56,61,69,70], and Porphyromonas spp., showed decreased abundances [54].
The abundance of the phylum Actinobacteria seems to decrease with adult CDI [61,62,64]. The main genus responsible for the decrease is Bifidobacterium spp.
Within the phylum Verrucomicrobia, some studies showed that an increase in the abundance of A. muciniphila is associated with CDI [62,68,71,77], while others reported that an increase in the abundance of Akkermansia protected against CDI [56,63,69].
Hernandez et al. (2019) classified CDI into two groups according to prognosis. Cluster A showed high abundances of Enterococcaceae and Enterococcus and decreases in the abundances of Bacteroidaceae and Lachnospiraceae. This cluster was associated with more severe diarrhea, more aggressive therapy and a poor prognosis. Cluster B had high abundances of Bacteroidaceae and Lachnospiraceae. This cluster was associated with less severe diarrhea, less aggressive therapies and a good prognosis [67].
Several recent studies have focused on comparing the gut microbiota of people with CDI with people with negative C. difficile detection and symptomatology. A significantly decrease in the bacterial population diversity (Shannon index) [54,64,65,66,70,74] and a significantly lower richness (Chao1 index) [64,66] has often been observed in patients with CDI.
People who developed a single CDI had higher levels of IgM anti-toxin A, toxin B and non-toxigenic antigens on Day 3 and significantly higher IgG anti-toxin A on Day 12 than people who developed recurrent CDI forms [30].
In Table 2, more details about these studies are provided.
Table 1. Variation in the abundance of the main phyla and genera of the gut microbiota of CDI people versus healthy people.
Table 1. Variation in the abundance of the main phyla and genera of the gut microbiota of CDI people versus healthy people.
Pathogens 11 00781 i009
Pathogens 11 00781 i010
p-values are the original value from research article, where * p < 0.05 ** p < 0.001. Bright red ( Pathogens 11 00781 i001) represents an increase of +10 to 33 %, red ( Pathogens 11 00781 i002) an increase of +34 to 66%, dark red ( Pathogens 11 00781 i003) an increase of +67 to 100%. Bright green ( Pathogens 11 00781 i004) represents a decrease of −10 to −33%, green ( Pathogens 11 00781 i005) a decrease of −34 to −66%, dark green ( Pathogens 11 00781 i006) a decrease of −67 to −100%. Striped red ( Pathogens 11 00781 i007) represents an increase by unspecified value and striped green ( Pathogens 11 00781 i008) a decrease by unspecified value.

3. Fecal Microbiota Transplantation (FMT)

FMT is the transfer of the fecal microbiota containing bacteria from a healthy volunteer into a diseased patient [82,83]. In 1958, Dr Ben Eiseman described the “fecal enema” as a treatment for pseudomembranous enterocolitis [83]. FMT is administered in several ways: with capsules for ingestion, with an endoscope and with a nasoenteric tube [84]. The criteria for selecting a healthy donor have been described in several papers [84,85]. Briefly, the blood and the stool must be tested to ensure that no infectious diseases or pathogenic bacteria are present, and a series of criteria are checked, including recent antibiotic use, history of diarrhea, and history of immune disorders [84,85].
In the case of primary CDI, this treatment could be used before using antibiotics or in addition of antibiotics to avoid rCDI [86]. In the case of rCDI, FMT is the second line of treatment. FMT has been reported to be successful in 80–92% of patients with rCDI [84,87,88,89] and with primary CDI [89]. This treatment is safe and effective in adults [87,88], in elderly [90,91,92] and in children [93].
Regarding the composition of the gut microbiota of patients with FMT, the alpha diversity (Shannon index) seems to be lowest pretreatment among patients with CDI, and the alpha diversity is restored after FMT [87,88,90,91]. The composition of the microbiota has an impact on the recurrence of CDI and the success of FMT. After FMT, the phylum Firmicutes increased significantly in rCDI (<65 y) and the phylum Proteobacteria decreased significantly in rCDI (>65 y) [90]. Lachnospiraceae, Ruminococcaceae and Bifidobacteriaceae increased significantly [90] and Enterobacteriaceae decreased significantly after FMT in rCDI [89,90]. Blautia, Ruminococcus, Coprococcus, Bifidobacterium [90] and Faecalibacterium [89] spp. increased significantly after FMT in rCDI [89,90]. These modifications of the gut microbiota after FMT strongly suggested that these bacterial populations are associated with healthy people (See Table 1) and will favor a good prognosis. Staley et al. (2018) showed that a follow-up analysis of 16S rDNA extracted from feces can be used to predict an eventual recurrence of CDI. After FMT, high proportions of Lactobacillales, Enterobacteriaceae, Enterococcus, Klebsiella, Streptococcus and Veillonella and reductions in Roseburia, Blautia, Lachnospiraceae, Ruminococcaceae, Anaerostipes, Coprococcus, Dorea and Clostridiales [88,91] will disadvantage the patient and promote rCDI. These bacterial population are associated with the gut microbiota of CDI cases (see in Table 1).
Before using the FMT, a preventive probiotic administration before the antibiotics use is effective [94]. The use of probiotics before and at the same time as antibiotics reduces the risk of CDI by >50% in hospitalized adults [94].

4. Conclusions

Many studies have characterized the gut microbiota composition of patients with CDI, but confusion is still present in the literature between CDI and AC. Few studies have differentiated AC by C. difficile and CDI. This review explores the available data related to the link between the gut microbiota and the development of C. difficile infection. The causes of the development of CDI are clearly multifactorial. An external cause (such as a medication) can disrupt the homoeostasis of the gut microbiota. PPI and antibiotic use decrease the richness of the gut microbiota [95]. This imbalance promotes the growth of some bacteria (for example, A. muciniphila), and these bacteria can degrade the mucus layer and allow the pathogenic bacteria and toxins access to the epithelium. Moreover, the abundances of some bacteria (Lachnospiraceae, Ruminococcaceae and Blautia) decrease, and these bacteria are involved in bile metabolism and can increase the primary bile acid concentration. Higher PBAs concentrations are favorable to C. difficile germination and multiplication. Some bacteria also have a positive correlation (Enterococcus, Enterobacteriaceae) or negative correlation (Blautia, Prevotella, Roseburia, Dorea, Collinsella, Coprococcus, Ruminococcus, Ruminococcaceae, Lachnospiraceae) with C. difficile colonization and/or CDI. The gut microbiota will promote the development of the CDI. Through all the studies, the CDI has a gut microbiota footprint with the decrease and the increase in some bacteria. In this review, a lot of bacteria are singled out for giving an advantage or a disadvantage when developing CDI. Some of these bacteria have an impact on gut health. Faecalibacterium prausnitzii is considered as a species of the healthy gut microbiota. This bacterium is reduced in gut dysbiosis, in IBD, in obesity, in diabetes, etc. [96]. Lachnospiraceae is protective against CDI [20]. C. scindens, member of Lachnospiraceae have a protective effect against CDI [20,32]. Amrane et al. (2018) showed that C. scindens is present in the feces when patient developing CDI [97]. Alistipes spp. indicated a controversial pathogenicity. On the one hand, the bacteria have protective effects against liver fibrosis, cancer immunotherapy and cardiovascular disease [98]. On the other hand, this genus is associated with colorectal cancer and mental disease [98].
In this review, CDI can be associated with an increase or a decrease in A. municiphila and AC is associated with a decrease A. municiphila. The presence of this bacteria is positive against obesity, diabetes, cardiometabolic disease and low-grade inflammation [99]. It is actually used to manage obesity [100]. In human intestinal organoids, C. difficile is capable of decreasing MUC2 production, but it is not responsible for altering host mucus oligosaccharide composition [31]. Furthermore, it has been reported that C. difficile is not capable of degrading mucin glycans, although coculture with mucin-degrading Akkermansia muciniphila, Bacteroides thetaiotaomicron and Ruminococcus torques allowed the pathogen to grow in media that lacked glucose but contained purified MUC2 [101]. When mucus is degraded by bacteria, oligosaccharides (GlcNAc, GalNAc, galactose, mannose and fucose) are salted out [101], and C. difficile is capable of using these oligosaccharides [31].
The Enterobacteriaceae family is associated with the dysbiosis state [58,59]. Enterococcus spp. is a controversial bacterium [102]. It is a commensal bacterium of intestinal flora, vagina, and mouth microbiota [102]. E. faecium and E. faecalis are used as probiotics [102,103] and Enterococcus spp. is used in meat and cheese [102,104] fermentation. Recently, it was shown that E. faecalis and E. faecium are potentially pathogenic bacteria due to their ability to adapt in new environment [102,105]. Additionally, a resistance to vancomycin has emerged in this genus [102,105]. Romyasamit et al. (2020) exhibited that six E. faecalis strains have a probiotic effect and anti-C. difficile activity [106]. Klebsiella pneumonae is present in the mucus layer with C. difficile [107].
The second objective of this review was to provide more information on why some people colonized with toxigenic C. difficile develop C. difficile infection and others show no signs of disease. The answer to this question is still unknown, but some facts will improve the understanding. The response of the adaptative immune system impacts the development of the disease. Patients exhibiting AC were shown to have higher antibody levels against C. difficile than people who developed CDI. It has been reported that sixty percent of the general population has had AC or has been infected with C. difficile, as determined by the observation of detectable seric IgG and IgA antibodies to toxins A and B [108]. IgG and IgA titers against toxins A and B are significantly higher in children positive for toxigenic strains than in people carrying non-toxigenic strains [109]. IgG antibody levels against toxin A are significantly higher within 3 days of colonization in AC patients than in those who develop CDI [30,108,110]. IgG levels against toxin B and non-toxin antigens seem to be higher among individuals who develop AC [30,108,110].
Some risks factors will predispose patients to developing CDI (antibiotics use, PPI use, age, etc.). The decrease in the diversity described in elderly gut microbiota [18], the decrease in some bacterial population (Ruminococcaceae, Bifidobacterium, Faecalibacterium) and the increase in some bacterial population (Proteobacteria, Bacteroidetes and Clostridium spp.) suggest why this population have a 10-fold higher risk of developing CDI.
Some treatments involving bacteria are commonly used and effective against CDI. FMT allows the recovery of patients with recurrent CDI with an increase in the abundances of some bacteria (Blautia, Collinsella, Anaerostipes, Coprococcus, Dorea and Roseburia) and a decrease in the abundances of others (Lactobacillales, Enterobacteriaceae; Enterococcus, Klebsiella, Streptococcus and Veillonella). More research with strict inclusion criteria is needed to measure AC and CDI gut microbiota analysis. The purpose of this work was to study the impact of in vivo control measures for the gut microbiota to decrease colonization and CDI or to improve recovery.

Author Contributions

Conceptualization, E.M.; methodology and validation, E.M. and G.D.; formal analysis, E.M. and G.D.; writing—original draft preparation, E.M.; writing—review and editing, E.M., G.D., B.T. and C.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention (U.S.). Antibiotic Resistance Threats in the United States, 2019; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2019.
  2. Colomb-Cotinat, M.; Assouvie, L.; Durand, J.; Daniau, C.; Leon, L.; Maugat, S.; Soing-Altrach, S.; Gateau, C.; Couturier, J.; Arnaud, I.; et al. Epidemiology of Clostridioides difficile infections, France, 2010 to 2017. Eurosurveillance 2019, 24, 1800638. [Google Scholar] [CrossRef]
  3. European Centre for Disease Prevention and Control. Healthcare-associated infections: Clostridium difficile infections. In Annual Epidemiological Report for 2016; ECDC: Stockholm, Sweden, 2018. [Google Scholar]
  4. Furuya-Kanamori, L.; Marquess, J.; Yakob, L.; Riley, T.V.; Paterson, D.L.; Foster, N.F.; Huber, C.A.; Clements, A.C.A. Asymptomatic Clostridium difficile colonization: Epidemiology and clinical implications. BMC Infect. Dis. 2015, 15, 516. [Google Scholar] [CrossRef] [PubMed]
  5. D’Silva, K.M.; Mehta, R.; Mitchell, M.; Lee, T.C.; Singhal, V.; Wilson, M.G.; McDonald, E.G. Proton pump inhibitor use and risk for recurrent Clostridioides difficile infection: A systematic review and meta-analysis. Clin. Microbiol. Infect. 2021, 27, 697–703. [Google Scholar] [CrossRef] [PubMed]
  6. Bavishi, C.; DuPont, H.L. Systematic Review: The use of proton pump inhibitors and increased susceptibility to enteric infection: Systematic review: Proton pump inhibitors and bacterial diarrhoea. Aliment. Pharmacol. Ther. 2011, 34, 1269–1281. [Google Scholar] [CrossRef] [PubMed]
  7. Jump, R.L.P.; Pultz, M.J.; Donskey, C.J. Vegetative Clostridium difficile survives in room air on moist surfaces and in gastric contents with reduced acidity: A potential mechanism to explain the association between proton pump inhibitors and C. difficile-associated diarrhea? Antimicrob. Agents Chemother. 2007, 51, 5. [Google Scholar] [CrossRef] [Green Version]
  8. Rao, A.; Jump, R.L.P.; Pultz, N.J.; Pultz, M.J.; Donskey, C.J. In vitro killing of nosocomial pathogens by acid and acidified nitrite. Antimicrob. Agents Chemother. 2006, 50, 3901–3904. [Google Scholar] [CrossRef] [Green Version]
  9. Brown, K.A.; Khanafer, N.; Daneman, N.; Fisman, D.N. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob. Agents Chemother. 2013, 57, 2326–2332. [Google Scholar] [CrossRef] [Green Version]
  10. Nasiri, M.J.; Goudarzi, M.; Hajikhani, B.; Ghazi, M.; Goudarzi, H.; Pouriran, R. Clostridioides (Clostridium) difficile infection in hospitalized patients with antibiotic-associated diarrhea: A systematic review and meta-analysis. Anaerobe 2018, 50, 32–37. [Google Scholar] [CrossRef]
  11. Webb, B.J.; Subramanian, A.; Lopansri, B.; Goodman, B.; Jones, P.B.; Ferraro, J.; Stenehjem, E.; Brown, S.M. Antibiotic exposure and risk for hospital-associated Clostridioides difficile infection. Antimicrob. Agents Chemother. 2020, 64, e02169-19. [Google Scholar] [CrossRef]
  12. Zomer, T.P.; Van Duijkeren, E.; Wielders, C.C.H.; Veenman, C.; Hengeveld, P.; Van Der Hoek, W.; De Greeff, S.C.; Smit, L.A.M.; Heederik, D.J.; Yzermans, C.J.; et al. Prevalence and risk factors for colonization of Clostridium difficile among adults living near livestock farms in the netherlands. Epidemiol. Infect. 2017, 145, 2745–2749. [Google Scholar] [CrossRef] [Green Version]
  13. The ANTICIPATE Study Group; van Werkhoven, C.H.; Ducher, A.; Berkell, M.; Mysara, M.; Lammens, C.; Torre-Cisneros, J.; Rodríguez-Baño, J.; Herghea, D.; Cornely, O.A.; et al. Incidence and predictive biomarkers of Clostridioides difficile infection in hospitalized patients receiving broad-spectrum antibiotics. Nat. Commun. 2021, 12, 2240. [Google Scholar] [CrossRef] [PubMed]
  14. Anjewierden, S.; Han, Z.; Brown, A.M.; Donskey, C.J.; Deshpande, A. Risk factors for Clostridioides difficile colonization among hospitalized adults: A meta-analysis and systematic review. Infect. Contr. Hosp. Epidemiol. 2021, 42, 565–572. [Google Scholar] [CrossRef] [PubMed]
  15. Li, Y.; Cai, H.; Sussman, D.A.; Donet, J.; Dholaria, K.; Yang, J.; Panara, A.; Croteau, R.; Barkin, J.S. Association between immunosuppressive therapy and outcome of Clostridioides difficile infection: Systematic review and meta-analysis. Dig. Dis. Sci. 2021. [Google Scholar] [CrossRef] [PubMed]
  16. Avni, T.; Babitch, T.; Ben-Zvi, H.; Hijazi, R.; Ayada, G.; Atamna, A.; Bishara, J. Clostridioides difficile infection in immunocompromised hospitalized patients is associated with a high recurrence rate. Int. J. Infect. Dis. 2020, 90, 237–242. [Google Scholar] [CrossRef] [Green Version]
  17. Rea, M.C.; O’Sullivan, O.; Shanahan, F.; O’Toole, P.W.; Stanton, C.; Ross, R.P.; Hill, C. Clostridium difficile carriage in elderly subjects and associated changes in the intestinal microbiota. J. Clin. Microbiol. 2012, 50, 867–875. [Google Scholar] [CrossRef] [Green Version]
  18. Dumic, I.; Nordin, T.; Jecmenica, M.; Stojkovic Lalosevic, M.; Milosavljevic, T.; Milovanovic, T. Gastrointestinal Tract Disorders in Older Age. Can. J. Gastroenterol. Hepatol. 2019, 2019, 6757524. [Google Scholar] [CrossRef] [Green Version]
  19. Lucado, J.; Gould, C.; Elixhauser, A. Clostridium difficile Infections (CDI) in Hospital Stays, 2009; HCUP Statistical Brief #124; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2012. Available online: http://www.hcup-us.ahrq.gov/reports/statbriefs/sb124.pdf (accessed on 23 June 2022).
  20. Crobach, M.J.T.; Vernon, J.J.; Loo, V.G.; Kong, L.Y.; Péchiné, S.; Wilcox, M.H.; Kuijper, E.J. Understanding Clostridium difficile colonization. Clin. Microbiol. Rev. 2018, 31, e00021-17. [Google Scholar] [CrossRef] [Green Version]
  21. Nagy, E. What do we know about the diagnostics, treatment and epidemiology of Clostridioides (Clostridium) difficile infection in Europe? J. Infect. Chemother. 2018, 24, 164–170. [Google Scholar] [CrossRef] [Green Version]
  22. Galdys, A.L.; Nelson, J.S.; Shutt, K.A.; Schlackman, J.L.; Pakstis, D.L.; Pasculle, A.W.; Marsh, J.W.; Harrison, L.H.; Curry, S.R. Prevalence and duration of asymptomatic Clostridium difficile carriage among healthy subjects in Pittsburgh, pennsylvania. J Clin. Microbiol. 2014, 52, 2406–2409. [Google Scholar] [CrossRef] [Green Version]
  23. Rousseau, C.; Levenez, F.; Fouqueray, C.; Doré, J.; Collignon, A.; Lepage, P. Clostridium difficile colonization in early infancy is accompanied by changes in intestinal microbiota composition. J. Clin. Microbiol. 2011, 49, 858–865. [Google Scholar] [CrossRef] [Green Version]
  24. Lees, E.A.; Miyajima, F.; Pirmohamed, M.; Carrol, E.D. The role of Clostridium difficile in the paediatric and neonatal gut—A narrative review. Eur. J. Clin. Microbiol. Infect. Dis. 2016, 35, 1047–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Miyajima, F.; Roberts, P.; Swale, A.; Price, V.; Jones, M.; Horan, M.; Beeching, N.; Brazier, J.; Parry, C.; Pendleton, N.; et al. Characterisation and carriage ratio of Clostridium difficile strains isolated from a community-dwelling elderly population in the United Kingdom. PLoS ONE 2011, 6, e22804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Jangi, S.; Lamont, J.T. Asymptomatic colonization by Clostridium difficile in infants: Implications for disease in later life. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 2–7. [Google Scholar] [CrossRef] [PubMed]
  27. Penders, J.; Thijs, C.; Vink, C.; Stelma, F.F.; Snijders, B.; Kummeling, I.; van den Brandt, P.A.; Stobberingh, E.E. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics 2006, 118, 511–521. [Google Scholar] [CrossRef] [Green Version]
  28. Leffler, D.A. Clostridium difficile infection. N. Engl. J. Med. 2015, 372, 1539–1548. [Google Scholar] [CrossRef] [Green Version]
  29. Pepin, J. Mortality attributable to nosocomial Clostridium difficile-associated disease during an epidemic caused by a hypervirulent strain in Quebec. Can. Med. Assoc. J. 2005, 173, 1037–1042. [Google Scholar] [CrossRef] [Green Version]
  30. Péchiné, S.; Collignon, A. Immune responses induced by Clostridium difficile. Anaerobe 2016, 41, 68–78. [Google Scholar] [CrossRef]
  31. Engevik, M.A.; Yacyshyn, M.B.; Engevik, K.A.; Wang, J.; Darien, B.; Hassett, D.J.; Yacyshyn, B.R.; Worrell, R.T. Human Clostridium Difficile infection: Altered mucus production and composition. Am. J. Physiol.-Gastrointest. Liver Physiol. 2015, 308, G510–G524. [Google Scholar] [CrossRef] [Green Version]
  32. Francis, M.B.; Allen, C.A.; Shrestha, R.; Sorg, J.A. Bile acid recognition by the Clostridium difficile germinant receptor, CspC, is important for establishing infection. PLoS Pathog. 2013, 9, e1003356. [Google Scholar] [CrossRef]
  33. Wilson, K.H.; Silva, J.; Fekety, F.R. Suppression of Clostridium difficile by Normal Hamster Cecal Flora and Prevention of Antibiotic-Associated Cecitis. Infect. Immun. 1981, 34, 626–628. [Google Scholar] [CrossRef] [Green Version]
  34. Babcock, G.J.; Broering, T.J.; Hernandez, H.J.; Mandell, R.B.; Donahue, K.; Boatright, N.; Stack, A.M.; Lowy, I.; Graziano, R.; Molrine, D.; et al. Human monoclonal antibodies directed against toxins a and b prevent Clostridium difficile-induced mortality in hamsters. Infect. Immun. 2006, 74, 6339–6347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Young, V.B. Old and new models for studying host-microbe interactions in health and disease: C. difficile as an example. Am. J. Physiol.-Gastrointest. Liver Physiol. 2017, 312, G623–G627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Steele, J.; Feng, H.; Parry, N.; Tzipori, S. Piglet models of acute or chronic Clostridium difficile Illness. J. Infect. Dis. 2010, 201, 428–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Pultz, N.J.; Donskey, C.J. Effect of antibiotic treatment on growth of and toxin production by Clostridium difficile in the cecal contents of mice. Antimicrob. Agents Chemother. 2005, 49, 3529–3532. [Google Scholar] [CrossRef] [Green Version]
  38. Yamamoto-Osaki, T.; Kamiya, S.; Sawamura, S.; Kai, M.; Ozawa, A. growth inhibition of Clostridium difficile by intestinal flora of infant faeces in continuous flow culture. J. Med. Microbiol. 1994, 40, 179–187. [Google Scholar] [CrossRef]
  39. Hopkins, M.J.; Macfarlane, G.T. Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. J. Med. Microbiol. 2002, 51, 448–454. [Google Scholar] [CrossRef] [PubMed]
  40. Macfarlane, G.T.; Macfarlane, S.; Gibson, G.R. Validation of a three-stage compound continuous culture system for investigating the effect of retention time on the ecology and metabolism of bacteria in the human colon. Microb. Ecol. 1998, 35, 180–187. [Google Scholar] [CrossRef]
  41. Best, E.L.; Freeman, J.; Wilcox, M.H. Models for the study of Clostridium difficile infection. Gut Microbes 2012, 3, 145–167. [Google Scholar] [CrossRef] [Green Version]
  42. Van Nuenen, M.H.M.C.; Diederick Meyer, P.; Venema, K. The effect of various inulins and Clostridium difficile on the metabolic activity of the human colonic microbiota in vitro. Microb. Ecol. Health Dis. 2003, 15, 137–144. [Google Scholar] [CrossRef]
  43. Cheng, M.; Ning, K. Stereotypes about enterotype: The old and new ideas. Genom. Proteom. Bioinform. 2019, 17, 4–12. [Google Scholar] [CrossRef]
  44. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.; Gasbarrini, A.; Mele, M. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. MetaHIT Consortium; Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [PubMed]
  46. Costea, P.I.; Hildebrand, F.; Arumugam, M.; Bäckhed, F.; Blaser, M.J.; Bushman, F.D.; de Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Gorvitovskaia, A.; Holmes, S.P.; Huse, S.M. Interpreting Prevotella and Bacteroides as biomarkers of diet and lifestyle. Microbiome 2016, 4, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Seekatz, A.M.; Young, V.B. Clostridium difficile and the microbiota. J. Clin. Investig. 2014, 124, 4182–4189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ross, C.L.; Spinler, J.K.; Savidge, T.C. Structural and functional changes within the gut microbiota and susceptibility to Clostridium difficile infection. Anaerobe 2016, 41, 37–43. [Google Scholar] [CrossRef] [Green Version]
  50. Jackson, M.A.; Goodrich, J.K.; Maxan, M.-E.; Freedberg, D.E.; Abrams, J.A.; Poole, A.C.; Sutter, J.L.; Welter, D.; Ley, R.E.; Bell, J.T.; et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut 2016, 65, 749–756. [Google Scholar] [CrossRef] [Green Version]
  51. Imhann, F.; Bonder, M.J.; Vich Vila, A.; Fu, J.; Mujagic, Z.; Vork, L.; Tigchelaar, E.F.; Jankipersadsing, S.A.; Cenit, M.C.; Harmsen, H.J.M.; et al. Proton pump inhibitors affect the gut microbiome. Gut 2016, 65, 740–748. [Google Scholar] [CrossRef] [Green Version]
  52. Freedberg, D.E.; Toussaint, N.C.; Chen, S.P.; Ratner, A.J.; Whittier, S.; Wang, T.C.; Wang, H.H.; Abrams, J.A. Proton pump inhibitors alter specific taxa in the human gastrointestinal microbiome: A crossover trial. Gastroenterology 2015, 149, 883–885.e9. [Google Scholar] [CrossRef] [Green Version]
  53. Vich Vila, A.; Collij, V.; Sanna, S.; Sinha, T.; Imhann, F.; Bourgonje, A.R.; Mujagic, Z.; Jonkers, D.M.A.E.; Masclee, A.A.M.; Fu, J.; et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 2020, 11, 362. [Google Scholar] [CrossRef]
  54. The ANTICIPATE Study Group; Berkell, M.; Mysara, M.; Xavier, B.B.; van Werkhoven, C.H.; Monsieurs, P.; Lammens, C.; Ducher, A.; Vehreschild, M.J.G.T.; Goossens, H.; et al. Microbiota-based markers predictive of development of Clostridioides difficile infection. Nat. Commun. 2021, 12, 2241. [Google Scholar] [CrossRef]
  55. Vakili, B.; Fateh, A.; Asadzadeh Aghdaei, H.; Sotoodehnejadnematalahi, F.; Siadat, S.D. Intestinal microbiota in elderly inpatients with Clostridioides difficile infection. IDR 2020, 13, 2723–2731. [Google Scholar] [CrossRef] [PubMed]
  56. Milani, C.; Ticinesi, A.; Gerritsen, J.; Nouvenne, A.; Lugli, G.A.; Mancabelli, L.; Turroni, F.; Duranti, S.; Mangifesta, M.; Viappiani, A.; et al. Gut Microbiota composition and Clostridium difficile infection in hospitalized elderly individuals: A metagenomic study. Sci. Rep. 2016, 6, 25945. [Google Scholar] [CrossRef] [PubMed]
  57. Winter, S.E.; Lopez, C.A.; Bäumler, A.J. The dynamics of gut-associated microbial communities during inflammation. EMBO Rep. 2013, 14, 319–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lupp, C.; Robertson, M.L.; Wickham, M.E.; Sekirov, I.; Champion, O.L.; Gaynor, E.C.; Finlay, B.B. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe 2007, 2, 119–129. [Google Scholar] [CrossRef] [Green Version]
  59. Mahnic, A.; Breskvar, M.; Dzeroski, S.; Skok, P.; Pintar, S.; Rupnik, M. Distinct types of gut microbiota dysbiosis in hospitalized gastroenterological patients are disease non-related and characterized with the predominance of either Enterobacteriaceae or Enterococcus. Front. Microbiol. 2020, 11, 120. [Google Scholar] [CrossRef]
  60. Pascal, V.; Pozuelo, M.; Borruel, N.; Casellas, F.; Campos, D.; Santiago, A.; Martinez, X.; Varela, E.; Sarrabayrouse, G.; Machiels, K.; et al. A Microbial signature for crohn’s disease. Gut 2017, 66, 813–822. [Google Scholar] [CrossRef]
  61. Zhang, L.; Dong, D.; Jiang, C.; Li, Z.; Wang, X.; Peng, Y. Insight into alteration of gut microbiota in clostridium difficile infection and asymptomatic C. difficile colonization. Anaerobe 2015, 34, 1–7. [Google Scholar] [CrossRef]
  62. Han, S.-H.; Yi, J.; Kim, J.-H.; Lee, S.; Moon, H.-W. Composition of gut microbiota in patients with toxigenic Clostridioides (Clostridium) difficile: Comparison between subgroups according to clinical criteria and toxin gene load. PLoS ONE 2019, 14, e0212626. [Google Scholar] [CrossRef] [Green Version]
  63. Rodriguez, C.; Taminiau, B.; Korsak, N.; Avesani, V.; Van Broeck, J.; Brach, P.; Delmée, M.; Daube, G. Longitudinal survey of Clostridium difficile presence and gut microbiota composition in a belgian nursing home. BMC Microbiol. 2016, 16, 229. [Google Scholar] [CrossRef] [Green Version]
  64. Jeon, Y.D.; Ann, H.W.; Lee, W.J.; Kim, J.H.; Seong, H.; Kim, J.H.; Ahn, J.Y.; Jeong, S.J.; Ku, N.S.; Yeom, J.S.; et al. Characteristics of faecal microbiota in korean patients with Clostridioides difficile-associated diarrhea. Infect. Chemother. 2019, 51, 365–375. [Google Scholar] [CrossRef] [PubMed]
  65. Amrane, S.; Hocquart, M.; Afouda, P.; Kuete, E.; Pham, T.-P.-T.; Dione, N.; Ngom, I.I.; Valles, C.; Bachar, D.; Raoult, D.; et al. Metagenomic and culturomic analysis of gut microbiota dysbiosis during Clostridium difficile infection. Sci. Rep. 2019, 9, 12807. [Google Scholar] [CrossRef] [Green Version]
  66. Gu, S.; Chen, Y.; Zhang, X.; Lu, H.; Lv, T.; Shen, P.; Lv, L.; Zheng, B.; Jiang, X.; Li, L. Identification of key taxa that favor intestinal colonization of Clostridium difficile in an adult chinese population. Microbes Infect. 2016, 18, 30–38. [Google Scholar] [CrossRef]
  67. Hernandez, D.R. Gut check: In vitro diagnostics for gut microbiome analysis. Clin. Microbiol. Newsl. 2019, 41, 57–62. [Google Scholar] [CrossRef]
  68. Sangster, W.; Hegarty, J.P.; Schieffer, K.M.; Wright, J.R.; Hackman, J.; Toole, D.R.; Lamendella, R.; Stewart, D.B. Bacterial and fungal microbiota changes distinguish C. difficile infection from other forms of diarrhea: Results of a prospective inpatient study. Front. Microbiol. 2016, 7, 789. [Google Scholar] [CrossRef] [PubMed]
  69. Stewart, D.B.; Wright, J.R.; Fowler, M.; McLimans, C.J.; Tokarev, V.; Amaniera, I.; Baker, O.; Wong, H.-T.; Brabec, J.; Drucker, R.; et al. Integrated meta-omics reveals a fungus-associated bacteriome and distinct functional pathways in Clostridioides difficile infection. mSphere 2019, 4, e00454-19. [Google Scholar] [CrossRef] [Green Version]
  70. Kim, J.; Cho, Y.; Seo, M.-R.; Bae, M.H.; Kim, B.; Rho, M.; Pai, H. Quantitative characterization of Clostridioides difficile population in the gut microbiome of patients with C. difficile infection and their association with clinical factors. Sci. Rep. 2020, 10, 17608. [Google Scholar] [CrossRef]
  71. Vakili, B.; Fateh, A.; Asadzadeh Aghdaei, H.; Sotoodehnejadnematalahi, F.; Siadat, S.D. Characterization of gut microbiota in hospitalized patients with Clostridioides difficile infection. Curr. Microbiol. 2020, 77, 1673–1680. [Google Scholar] [CrossRef]
  72. Lamendella, R.; Wright, J.R.; Hackman, J.; McLimans, C.; Toole, D.R.; Bernard Rubio, W.; Drucker, R.; Wong, H.T.; Sabey, K.; Hegarty, J.P.; et al. Antibiotic treatments for Clostridium difficile infection are associated with distinct bacterial and fungal community structures. mSphere 2018, 3, e00572-17. [Google Scholar] [CrossRef] [Green Version]
  73. Rojo, D.; Gosalbes, M.J.; Ferrari, R.; Pérez-Cobas, A.E.; Hernández, E.; Oltra, R.; Buesa, J.; Latorre, A.; Barbas, C.; Ferrer, M.; et al. Clostridium difficile heterogeneously impacts intestinal community architecture but drives stable metabolome responses. ISME J. 2015, 9, 2206–2220. [Google Scholar] [CrossRef]
  74. Antharam, V.C.; Li, E.C.; Ishmael, A.; Sharma, A.; Mai, V.; Rand, K.H.; Wang, G.P. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J. Clin. Microbiol. 2013, 51, 2884–2892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Theriot, C.M.; Koenigsknecht, M.J.; Carlson, P.E.; Hatton, G.E.; Nelson, A.M.; Li, B.; Huffnagle, G.B.; Li, J.Z.; Young, V.B. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun. 2014, 5, 3114. [Google Scholar] [CrossRef] [PubMed]
  76. Buffie, C.G.; Bucci, V.; Stein, R.R.; McKenney, P.T.; Ling, L.; Gobourne, A.; No, D.; Liu, H.; Kinnebrew, M.; Viale, A.; et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 2015, 517, 205–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Araos, R.; Andreatos, N.; Ugalde, J.; Mitchell, S.; Mylonakis, E.; D’Agata, E.M.C. Fecal microbiome among nursing home residents with advanced dementia and Clostridium difficile. Dig. Dis. Sci. 2018, 63, 1525–1531. [Google Scholar] [CrossRef]
  78. Schubert, A.M.; Rogers, M.A.M.; Ring, C.; Mogle, J.; Petrosino, J.P.; Young, V.B.; Aronoff, D.M.; Schloss, P.D. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. mBio 2014, 5, e01021-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hernández, M.; de Frutos, M.; Rodríguez-Lázaro, D.; López-Urrutia, L.; Quijada, N.M.; Eiros, J.M. Fecal microbiota of toxigenic Clostridioides difficile-associated diarrhea. Front. Microbiol. 2019, 9, 3331. [Google Scholar] [CrossRef]
  80. Pakpour, S.; Bhanvadia, A.; Zhu, R.; Amarnani, A.; Gibbons, S.M.; Gurry, T.; Alm, E.J.; Martello, L.A. Identifying predictive features of Clostridium difficile infection recurrence before, during, and after primary antibiotic treatment. Microbiome 2017, 5, 148. [Google Scholar] [CrossRef] [Green Version]
  81. Khanna, S.; Montassier, E.; Schmidt, B.; Patel, R.; Knights, D.; Pardi, D.S.; Kashyap, P.C. Gut microbiome predictors of treatment response and recurrence in primary Clostridium difficile infection. Aliment. Pharmacol. Ther. 2016, 44, 715–727. [Google Scholar] [CrossRef] [Green Version]
  82. Bakker, G.J.; Nieuwdorp, M. Fecal microbiota transplantation: Therapeutic potential for a multitude of diseases beyond Clostridium difficile. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  83. Wortelboer, K.; Nieuwdorp, M.; Herrema, H. Fecal microbiota transplantation beyond Clostridioides difficile infections. EBioMedicine 2019, 44, 716–729. [Google Scholar] [CrossRef] [Green Version]
  84. Kim, K.O.; Gluck, M. Fecal microbiota transplantation: An update on clinical practice. Clin. Endosc. 2019, 52, 137–143. [Google Scholar] [CrossRef] [PubMed]
  85. Owens, C.; Broussard, E.; Surawicz, C. Fecal microbiota transplantation and donor standardization. Trends Microbiol. 2013, 21, 443–445. [Google Scholar] [CrossRef] [PubMed]
  86. Juul, F.E.; Garborg, K.; Bretthauer, M.; Skudal, H.; Øines, M.N.; Wiig, H.; Rose, Ø.; Seip, B.; Lamont, J.T.; Midtvedt, T.; et al. Fecal microbiota transplantation for primary Clostridium difficile infection. N. Engl. J. Med. 2018, 378, 2535–2536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Kelly, C.R.; Khoruts, A.; Staley, C.; Sadowsky, M.J.; Abd, M.; Alani, M.; Bakow, B.; Curran, P.; McKenney, J.; Tisch, A.; et al. Effect of fecal microbiota transplantation on recurrence in multiply recurrent Clostridium difficile infection: A randomized trial. Ann. Intern. Med. 2016, 165, 609. [Google Scholar] [CrossRef] [Green Version]
  88. Staley, C.; Kaiser, T.; Vaughn, B.P.; Graiziger, C.T.; Hamilton, M.J.; Rehman, T.U.; Song, K.; Khoruts, A.; Sadowsky, M.J. Predicting recurrence of Clostridium difficile infection following encapsulated fecal microbiota transplantation. Microbiome 2018, 6, 166. [Google Scholar] [CrossRef] [Green Version]
  89. Haifer, C.; Paramsothy, S.; Borody, T.J.; Clancy, A.; Leong, R.W.; Kaakoush, N.O. Long-term bacterial and fungal dynamics following oral lyophilized fecal microbiota transplantation in Clostridioides difficile infection. mSystems 2021, 6, e00905-20. [Google Scholar] [CrossRef]
  90. Girotra, M.; Garg, S.; Anand, R.; Song, Y.; Dutta, S.K. Fecal microbiota transplantation for recurrent Clostridium difficile infection in the elderly: Long-term outcomes and microbiota changes. Dig. Dis. Sci. 2016, 61, 3007–3015. [Google Scholar] [CrossRef]
  91. Song, Y.; Garg, S.; Girotra, M.; Maddox, C.; von Rosenvinge, E.C.; Dutta, A.; Dutta, S.; Fricke, W.F. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PLoS ONE 2013, 8, e81330. [Google Scholar] [CrossRef] [Green Version]
  92. Friedman-Korn, T.; Livovsky, D.M.; Maharshak, N.; Aviv Cohen, N.; Paz, K.; Bar-Gil Shitrit, A.; Goldin, E.; Koslowsky, B. Fecal transplantation for treatment of Clostridium difficile infection in elderly and debilitated patients. Dig. Dis. Sci. 2018, 63, 198–203. [Google Scholar] [CrossRef]
  93. Nicholson, M.R.; Mitchell, P.D.; Alexander, E.; Ballal, S.; Bartlett, M.; Becker, P.; Davidovics, Z.; Docktor, M.; Dole, M.; Felix, G.; et al. Efficacy of fecal microbiota transplantation for Clostridium difficile infection in children. Clin. Gastroenterol. Hepatol. 2020, 18, 612–619.e1. [Google Scholar] [CrossRef]
  94. Shen, N.T.; Maw, A.; Tmanova, L.L.; Pino, A.; Ancy, K.; Crawford, C.V.; Simon, M.S.; Evans, A.T. Timely use of probiotics in hospitalized adults prevents Clostridium difficile infection: A systematic review with meta-regression analysis. Gastroenterology 2017, 152, 1889–1900.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Seto, C.T.; Jeraldo, P.; Orenstein, R.; Chia, N.; DiBaise, J.K. Prolonged use of a proton pump inhibitor reduces microbial diversity: Implications for Clostridium difficile susceptibility. Microbiome 2014, 2, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Björkqvist, O.; Rangel, I.; Serrander, L.; Magnusson, C.; Halfvarson, J.; Norén, T.; Bergman-Jungeström, M. Faecalibacterium prausnitzii increases following fecal microbiota transplantation in recurrent Clostridioides difficile infection. PLoS ONE 2021, 16, e0249861. [Google Scholar] [CrossRef] [PubMed]
  97. Amrane, S.; Bachar, D.; Lagier, J.C.; Raoult, D. Clostridium scindens is present in the gut microbiota during Clostridium difficile infection: A metagenomic and culturomic analysis. J. Clin. Microbiol 2018, 56, e01663-17. [Google Scholar] [CrossRef] [Green Version]
  98. Parker, B.J.; Wearsch, P.A.; Veloo, A.C.M.; Rodriguez-Palacios, A. The genus Alistipes: Gut bacteria with emerging implications to inflammation, cancer, and mental health. Front. Immunol. 2020, 11, 906. [Google Scholar] [CrossRef] [PubMed]
  99. Cani, P.D.; de Vos, W.M. Next-generation beneficial microbes: The case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef] [PubMed]
  100. Roshanravan, N.; Bastani, S.; Tutunchi, H.; Kafil, B.; Nikpayam, O.; Mesri Alamdari, N.; Hadi, A.; Sotoudeh, S.; Ghaffari, S.; Ostadrahimi, A. A Comprehensive systematic review of the effectiveness of Akkermansia muciniphila, a member of the gut microbiome, for the management of obesity and associated metabolic disorders. Arch. Physiol. Biochem. 2021. [Google Scholar] [CrossRef]
  101. Engevik, M.A. Mucin-degrading microbes release monosaccharides that chemoattract Clostridioides difficile and facilitate colonization of the human intestinal mucus layer. ACS Infect. Dis. 2021, 7, 1126–1142. [Google Scholar] [CrossRef]
  102. Krawczyk, B.; Wityk, P.; Gałęcka, M.; Michalik, M. The Many Faces of Enterococcus Spp.—Commensal, probiotic and opportunistic pathogen. Microorganisms 2021, 9, 1900. [Google Scholar] [CrossRef]
  103. Franz, C.M.A.P.; Huch, M.; Abriouel, H.; Holzapfel, W.; Gálvez, A. Enterococci as probiotics and their implications in food safety. Int. J. Food Microbiol. 2011, 151, 125–140. [Google Scholar] [CrossRef] [Green Version]
  104. García-Díez, J.; Saraiva, C. Use of starter cultures in foods from animal origin to improve their safety. IJERPH 2021, 18, 2544. [Google Scholar] [CrossRef] [PubMed]
  105. Gaca, A.O.; Lemos, J.A. Adaptation to adversity: The intermingling of stress tolerance and pathogenesis in Enterococci. Microbiol. Mol. Biol. Rev. 2019, 83, e00008-19. [Google Scholar] [CrossRef] [PubMed]
  106. Romyasamit, C.; Thatrimontrichai, A.; Aroonkesorn, A.; Chanket, W.; Ingviya, N.; Saengsuwan, P.; Singkhamanan, K. Enterococcus faecalis isolated from infant feces inhibits toxigenic Clostridioides (Clostridium) difficile. Front. Pediatr. 2020, 8, 572633. [Google Scholar] [CrossRef] [PubMed]
  107. Ticer, T.; Engevik, M. Klebsiella pneumoniae in the colonic mucus layer influences Clostridioides difficile pathogenesis. Am. J. Pathol. 2021, 192. [Google Scholar] [CrossRef]
  108. Kelly, C.P.; Kyne, L. The host immune response to Clostridium difficile. J. Med. Microbiol. 2011, 60, 1070–1079. [Google Scholar] [CrossRef] [Green Version]
  109. Kociolek, L.K.; Espinosa, R.O.; Gerding, D.N.; Hauser, A.R.; Ozer, E.A.; Budz, M.; Balaji, A.; Chen, X.; Tanz, R.R.; Yalcinkaya, N.; et al. Natural Clostridioides difficile toxin immunization in colonized infants. Clin. Infect. Dis. 2020, 70, 2095–2102. [Google Scholar] [CrossRef]
  110. Kyne, L.; Warny, M.; Qamar, A.; Kelly, C.P. Asymptomatic carriage of Clostridium difficile and serum levels of IgG antibody against toxin A. N. Engl. J. Med. 2000, 342, 390–397. [Google Scholar] [CrossRef]
Figure 1. Box plot illustrating the mean relative proportion of C. difficile asymptomatic carriers in function of category of age. These data were found in articles that studied the prevalence of AC [4,20,21,22,23,24,25,26,27].
Figure 1. Box plot illustrating the mean relative proportion of C. difficile asymptomatic carriers in function of category of age. These data were found in articles that studied the prevalence of AC [4,20,21,22,23,24,25,26,27].
Pathogens 11 00781 g001
Table 2. Analysis of the most important taxa exhibiting increased and decreased abundances in the CDI group compared with the control group.
Table 2. Analysis of the most important taxa exhibiting increased and decreased abundances in the CDI group compared with the control group.
Analysis MethodStudyStudy GroupDiversityIncrease in CDI Group Compared with Control GroupDecrease in CDI Group Compared with Control Group
16S rRNA gene amplicon analysis a
Illumina
[54]CDI group (n = 15)
Control group (n = 669)
Mean age: 70 y
Location: Europa
The alpha diversity (Chao1 and Shannon index) was lower in the CDI group. *Enterococcus spp. *Bifidobacterium spp., Blautia spp.
Faecalibacterium spp., Bacteroides spp. and Prevotella spp.
qPCR[71]CDI group (n = 28; 79 y)
Control group (n = 56; 75 y)
Country: Iran
NRA. muciniphila *, Lactobacillus spp. * and Escherichia coli *Bacteroides spp. *, Bifidobacterium spp.* and F. prausnitzii *
16S rRNA gene amplicon analysis a
Illumina HiSeq
[70]CDI group (n = 26; 66.5 y)
Control group (n = 61)
The alpha diversity (Shannon index) was lower in the CDI group. *Enterococcus *, Lactobacillus *, Escherichia * and Klebsiella *.Bifidobacterium *, Ruminococcus *, Eubacterium *, Faecalibacterium spp.*, Prevotella *, Blautia *, Collinsella *, Dorea *, Alistipes *, Lachnospiraceae * and Coprococcus * (p < 0.05)
16S rRNA gene amplicon analysis a
MiSeq Illumina
[65]CDI group (n = 11; 70.81 ± 20.1 y)
Control group (n = 8; 18 to 45 y)
Country: France
The alpha diversity (Shannon index) was lower in the CDI group. *Proteobacteria *nsFirmicute *ns
Actinobacteria *ns
Bacteroidetes *
454 pyrosequencing analysis of bacteria e 454 GS FLX Titanium Sequencing System (Roche)[64]CDI group (n = 24, 64.8 ± 15.7 y)
Control group (n = 13, 49.2 ± 11.5 y).
Country: Korea
The richness (Chao1 index; 283.3 vs. 642.9) and the alpha diversity (Shannon index; 3.6 vs. 4.5) were lower in the CDI group. *Proteobacteria *ns
Bacteroidetes *ns
Verrucomicrobia
Bacteroides, Streptococcus spp., and Escherichia
Actinobacteria *
Blautia, Bifidobacterium, Faecalibacterium, Collinsela, Dorea, Eubacterium, Fusicatenibacter, Prevotella, Roseburia, Subdoligranum, Ruminococcus, Clostridium, Catenibacterium, Dialister and Anaerostipes
16S rRNA gene amplicon analysis
Metatranscriptomic
Illumina Hiseq4000 platform
[69]CDI group (n = 18, 65,3 ± 17 y)
Control group (n = 31, 60 ± 18 y)
Country: United States
A lower average species evenness was observed in the CDI group with Heip’s evenness (93.4 ± 23.1 in the CDI group vs. 121.8 ± 58.2 in the control group). *nsMetagenetic
Increases in the proportions of Clostridiaceae, Peptostreptoccocaceae, and Enterococcus
Metatranscriptomic
Increases in the proportions of Clostridioides difficile, E. coli, unclassified Peptostreptococcaceae, and Enterobacteriaceae
Metagenetic
Decreases in the proportions of Faecalibacterium and Collinsella
Metatranscriptomic
Decreases in the proportions of A. municiniphila, Faecalibacterium prausnitzii, Coprococcus, Alistipes shahii, Collinsella and Verrucomicrobiaceae *
16S rRNA gene amplicon analysis b
MiSeq technology Illumina
[68]CDI group (n = 13; 55.5 ± 20.5 y)
Control group (n = 13; 51.2 ± 16.6 y)
No difference was observed in richness or evenness between groups. *nsEnterobaceriaceae *
Peptostreptococcaceae *
A. muciniphila *
Bacteroidales
Clostridales
16S rRNA gene amplicon analysis c
Ion Chez System and Ion S5 L system
[62]tcdB-positive group (n = 79, 62.5 ± 19.9 y) divided into two groups: CDI group (n = 58) and colonized group (n = 21). Control group (n = 20, 62.2 ± 14.4 y).
Country: Korea
The richness (Chao1 index; 60 vs. 95) was lower in the tcdB group. *Proteobacteria *,
Enterobacteriaceae*, Porphyromonadaceae *, Enterococcaceae *
Parabacteroides*, Enterococcus *, Veillonella *, Klebsiella * and Akkermansia *
Lachnospiraceae *, Ruminococcaceae *, Prevotellaceae *
Prevotella, Phascolarctobacterium, Haemophilus, Lachnospira, Coprococcus, Dialister, Butyricimonas, Catenibacterium, Faecalibacterium, Paraprevotella, Odoribacter and Anaerostipes
Pyrosequencing e
Roche GS Junior
[63]Control group (n = 94)
CA group (n = 24)
Mean age: 78.66 y
No difference in diversity between the groups.Blautia
Flavonifractor
Lachnospiraceae_unclassified
Akkermansia
Pyrosequencing d
Illumina MiSeq
[77]CA group (n = 7)
Control group (n = 25)
Mean age: 89.3 y
Country: United States.
No difference in the alpha diversity or beta diversity
(Shannon index: 3.47 in the CA group vs. 3.12 in the control group).
Firmicutes, Actinobacteria
Akkermansia spp., Dermabacter spp., Romboutsia spp., Meiothermus spp., Peptoclostridium spp., Ruminococcaceae UGC 009 *
Bacteroidetes, Proteobacteria
Pyrosequencing a
Roche 454 GS FLX and sequencer
[61]CDI group (n = 8, 58.9 ± 22.2 y)
CA group (n = 8, 60.5 ± 20.8 y)
Control group (n = 9, 60.8 ± 16.02)
Country: China
Reductions in richness (Chao) *ns and diversity (Simpson and Shannon indexes) * in the CDI and CD+ groups.Proteobacteria
Fusobacteria
Clostridium cluster XI
Parabacteroides
Escherichia/Shigella
Klebsiella
Enterococcus
Veillonella
Lactobacillus
Bacteroidetess and Firmicutes.
Prevotella,
Bacteroides,
Faecalibacterium,
Coprococcus,
Roseburia
Pyrosequencing d[17]Control group (n = 252)
CA group (n = 22)
Age: 65 y
Country: Ireland
NRBacteroidaceae
Ruminococcaceae
Clostridiaceceae
Erysipelothrichaceae
Aerococacceae (one patient)
Flavobacteriaceae (one patient)
Enterococcaceae
Prevotellaceae
Leuconostocacceae
Spirochaetaceae
16S rRNA gene amplicon analysis b
Illumina MiSeq sequencer
[56]CDI group (n = 25, 82.9 ± 8.5)
AB group (n = 29, 84.2 ± 8.1)
Control group (n = 30, 82.3 ± 6.8).
Country: Italy
Decreased diversity index (Chao1 and Shannon index) in the CDI group compared to the AB group.Klebsiella *, Escherichia/shigella *, Sutterella, Enterococcus *, Citrobacter, Veillonella, Proteus, Morganella, Hafnia, Corynebacterium, StaphylococcusFaecalibacterium, Bifidobacterium, Akkermansia,
Bacteroides *,
Lachnospira *,
Alistipes *
16S rRNA gene amplicon analysis b
QMiSeq Illumina
[66]CDI group (n = 15; 61 y).
Diarrhea group (n = 18; 56.5 y).
Control group (n = 25, 58 y)
Country: China
Decreased alpha diversity (Shannon index) and richness (Chao1) in the CDI group and diarrhea group. *Proteobacteria *,
Enterococcaceae, Streptococcaceae, Lactobacillaceae and Peptostreptococcaceae *ns
Clostridium spp.*ns
Actimnomyces and Rothiabacterium
Escherichia/Shigella, Klebsiella, Proteus and Providencia
Firmicutes and Bacteroidetes *
Lachnospiraceae.* BlautiaLachnospiraceae_incertae_sedis, Roseburia and Dorea
Ruminococcaceae.* Faecalibacterium, Clostridium IV, Oscillospira and Ruminococcus
454-pyrosequencing f
FLX Titanium platform
[73]All samples with diarrhea
CDI–toxins group (71 ± 15 y)
CDI–without toxins group (66.3 ± 27 y)
Control group (52.0 ± 13 y)
Country: Spain
NRBacteroides
Parabacteroides
Faecalibacterium
Clostridium XIVa
Clostridium cluster XI
CDI group with and without toxins: Phascolarctobacterium, Enterococcus, Clostridium XI cluster, flavonifractor, and Erysipelotrichaceae incertea sedis *
CDI group with toxins:
Enterococcus, Clostridium XI cluster, and Erysipelotrichaceae incerteae sedis
CDI group with and without toxins:
Blautia, Holdemania, Enterobacteriaceae, and Veillonellaceae *
CDI group with toxins:
Fusobacterium, Prevotella, Vovibrio, and Dialister
454-Pyrosequencing g[78]CDI group (n = 94; 55.9 ± 18.3)
CDN (=C. difficile-negative nosocomial diarrhea) group (n = 89; 58.7 ± 14.9)
Control group (n = 155; 52.2 ± 21.5)
Reduction in the diversity (inverse Simpson index) * in the CDI and CDN groups compared with the control group.Enterococcus *
Lachnospiraceae *
Erysipelotrichaceae *
Bacteroides species *
Pyrosequencing e[74]CDI group (n = 39; 54.7 ± 20.1)
CDN (= C. difficile-negative nosocomial diarrhea) group (n = 36; 54.6 ± 20.0)
Control group (n = 40; 60.9 ± 9.1)
Reduction in the diversity (Shannon index) * in the CDI and CDN groups compared with the control group.Bacteroides
Veillonella
Enterococcus
Lactobacillus
Fusobacterium
Firmicutes
Lachnospiraceae and Ruminococcaceae
Blautia * Pseudobutyrivibrio, Roseburia, Faecalibacterium, Anaerostipes, Subdoligranulum, Ruminococcus, Streptococcus, Dorea * and Coprococcus.
16S rRNA gene amplicon analysis a
MiSeq technology Illumina
[79]CDI group (n = 57; 69.5 y): GDH+ and TcdB
No control group
Country: Spain
The richness (Chao1) was 134.32 and the
alpha-diversity (Shannon index) was 4.01.
Bacteroides (46.51%), Firmicutes (34.70%), Proteobacteria (13.49%).
Bacteroidaceae (31.01%). Enterobacteriaceae (9.82%), Lachnospiraceae (9.33%), Tannerllaceae (6.16%) and Ruminococcaceae (5.64%)
16S rRNA gene amplicon analysis d
MiSeq technology Illumina
[80]CDI group (n = 31; 64.0 y).
Three periods: pretreatment ATB, two days after treatment, seven days after treatment or discharge.
No control group
The alpha diversity (Shannon index) was lower with CDI pretreatment in the recurrent group. *Veillonella dispar * predictor of recurrence./
16S rRNA gene amplicon analysis d
Illumina MiSeq sequencer
[81]CDI group (n = 88; 52.7 y)
G1 (ATB responder)
G2 (non-ATB responder)
G3 (recurrent CDI)
G4 (non-recurrent CDI)
No control group (other studies)
Decreased alpha diversity (Chao1 index) in the CDI group than in the control group.G1
Ruminococcaceae; Rikenellaceae; Clostridiaceae; Bacteroides; Faecalibacterium; Rothia
G2
Clostridiaceae, Lachnospiraceae, Blautia, Coprococcus, Streptococcus, Bifidobacterium, Ruminococcus and Actinomyces.
G3 with recurrent CDI
Veillonella, Enterobacteriaceae, Streptococci, Parabacteroides, and Lachnospiraceae
/
a V3–V4 regions of the 16S rRNA gene. b V3 region of the 16S rRNA gene. c V2–V3 regions of the 16S rRNA gene. d V4 regions of the 16S rRNA gene. e V1–V3 regions of the 16S rRNA gene. f V1–V2 regions of the 16S rRNA gene. g V3–V5 regions of the 16S rRNA gene. * Significant. *ns, non-significant.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martinez, E.; Taminiau, B.; Rodriguez, C.; Daube, G. Gut Microbiota Composition Associated with Clostridioides difficile Colonization and Infection. Pathogens 2022, 11, 781. https://doi.org/10.3390/pathogens11070781

AMA Style

Martinez E, Taminiau B, Rodriguez C, Daube G. Gut Microbiota Composition Associated with Clostridioides difficile Colonization and Infection. Pathogens. 2022; 11(7):781. https://doi.org/10.3390/pathogens11070781

Chicago/Turabian Style

Martinez, Elisa, Bernard Taminiau, Cristina Rodriguez, and Georges Daube. 2022. "Gut Microbiota Composition Associated with Clostridioides difficile Colonization and Infection" Pathogens 11, no. 7: 781. https://doi.org/10.3390/pathogens11070781

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop