Next Article in Journal
Phylogenetic Characterization of Rabies Virus Field Isolates Collected from Animals in European Russian Regions in 2009–2022
Next Article in Special Issue
Genetic Insights into Biofilm Formation by a Pathogenic Strain of Vibrio harveyi
Previous Article in Journal
Anti-Toxin Responses to Natural Enterotoxigenic Escherichia coli (ETEC) Infection in Adults and Children in Bangladesh
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of the Impact of Biofilm Formation on Recurrent Clostridioides difficile Infection

by
Daira Rubio-Mendoza
1,
Adrián Martínez-Meléndez
2,
Héctor Jesús Maldonado-Garza
1,
Carlos Córdova-Fletes
1 and
Elvira Garza-González
1,*
1
Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico
2
Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(10), 2525; https://doi.org/10.3390/microorganisms11102525
Submission received: 5 September 2023 / Revised: 25 September 2023 / Accepted: 29 September 2023 / Published: 10 October 2023
(This article belongs to the Special Issue Advances in Microbial Biofilm Formation)

Abstract

:
Clostridioides difficile infection (CDI) may recur in approximately 10–30% of patients, and the risk of recurrence increases with each successive recurrence, reaching up to 65%. C. difficile can form biofilm with approximately 20% of the bacterial genome expressed differently between biofilm and planktonic cells. Biofilm plays several roles that may favor recurrence; for example, it may act as a reservoir of spores, protect the vegetative cells from the activity of antibiotics, and favor the formation of persistent cells. Moreover, the expression of several virulence genes, including TcdA and TcdB toxins, has been associated with recurrence. Several systems and structures associated with adhesion and biofilm formation have been studied in C. difficile, including cell-wall proteins, quorum sensing (including LuxS and Agr), Cyclic di-GMP, type IV pili, and flagella. Most antibiotics recommended for the treatment of CDI do not have activity on spores and do not eliminate biofilm. Therapeutic failure in R-CDI has been associated with the inadequate concentration of drugs in the intestinal tract and the antibiotic resistance of a biofilm. This makes it challenging to eradicate C. difficile in the intestine, complicating antibacterial therapies and allowing non-eliminated spores to remain in the biofilm, increasing the risk of recurrence. In this review, we examine the role of biofilm on recurrence and the challenges of treating CDI when the bacteria form a biofilm.

1. Introduction

Clostridioides difficile is a Gram-positive sporulated bacterium that was considered to be the most common cause of healthcare-associated infective diarrhea over the last decades. However, it has been shown that reservoirs of C. difficile in the community may participate in the transmission of this infection [1].
CDI is transmitted by the consumption of spores and is associated with the disruption of the gut microbiota by the use of antimicrobials [2,3,4].
A C. difficile (CDI) infection ranges from simple colitis to pseudomembranous and fulminant colitis. The symptomatology of CDI is characterized by the presence of at least three loose or unformed stools in 24 h or less, along with a history of antibiotic exposure or evidence of megacolon or severe ileus with a positive laboratory diagnostic test result or colonoscopic or histopathological findings revealing pseudomembranous colitis [2]. The most common risk factors for CDI are age above 65, previous use of antibiotics, recent hospitalization [5,6], and enteral feeding [7] (Table 1).
Vancomycin and metronidazole are first-line CDI treatments [8]. Approximately 10–30% of patients develop recurrent CDI (R-CDI) [9], and the risk of recurrence increases with each successive recurrence, from 40% up to 65% [2]. An R-CDI infection is defined by symptoms within eight weeks after a resolved primary infection. The cause of R-CDI may be a relapse, infection by the same strain, reinfection, or infection by a different strain [2]. In patients with R-CDI, other alternatives, such as the fidaxomicin of a fecal microbiota transplant, may be used [8,10].
Some risk factors of R-CDI include immunosuppression [11]; infection with ribotype 027 5, 078, or 244 [11]; a previous history of CDI; severe CDI [4]; gastrointestinal intervention [6]; ≥15 days of acid-suppressive therapy [7]; and serum albumin levels of ˂2.5 g/dL [5,12] (Table 1). The biofilms in the gut (only by C. difficile or with other bacterial species of the microbiota) may contribute to recurrence [13]. Within the biofilm, interactions between bacteria, including C. difficile adhesion and chemotaxis, modulation of LuxS/AI-2 quorum sensing (QS) system activity, and regulation of intestinal bile acid levels [13], may have an impact on CDI [13].
Table 1. Risk factors for primary Clostridioides difficile infection (CDI) and recurrent Clostridioides difficile infection (R-CDI).
Table 1. Risk factors for primary Clostridioides difficile infection (CDI) and recurrent Clostridioides difficile infection (R-CDI).
Risk FactorOdds RatiopRef.
Primary CDI a
Age ≥ 65 years2.4 (1.6–3.5) c<0.001[6]
Recent hospitalization2.1 (1.5–3.1) b<0.001[6]
Enteral feeding2.9 (2.0–4.1)<0.001[7]
Vascular surgery2.4 (1.3–4.5)0.003[6]
Surgery in the preceding 12 weeks1.7 (1.3–2.4) c<0.001[6]
Surgery and gastrointestinal interventions 1.9 (1.2–3.0) b0.003[6]
Myocardial infarction 1.7 (1.2–2.6)0.003[6]
Gastrointestinal intervention2.9 (1.9–4.6)<0.001[6]
Congestive heart failure1.9 (1.4–2.5)<0.001[6]
Chronic kidney disease1.9 (1.4–2.6)<0.001[7]
Peripheral vascular disease1.5 (1.1–2.2)0.011[6]
Diabetes with organ damage2.1 (1.3–3.4)0.001[6]
Cerebrovascular disease2.0 (1.2–3.5)0.008[6]
Dementia4.0 (2.4–6.8)<0.001[7]
Connective tissue disease3.3 (1.6–6.7)<0.001[7]
Inflammatory bowel disease2.2 (1.2–4.1)0.006[7]
Urinary tract infection2.2 (1.3–4.1)0.004[6]
Dementia2.5 (1.3–4.8)0.003[6]
Chronic obstructive pulmonary disease2.0 (1.1–3.8)0.021[6]
Leukemia2.3 (1.2–4.1)0.004[7]
Charlson Comorbidity Index score 3 ≥ 3 vs. <31.5 (1.2–1.9)<0.001[7]
Duration of acid-suppressive therapy (≥15 days)3.8 (2.9–4.8)<0.001[7]
Chemotherapy1.8 (1.1–2.8)0.006[7]
Corticosteroids1.6 (1.2–2.1)<0.001[7]
Immunosuppressant agent use1.6 (1.22.14)<0.001[7]
Proton pump inhibitor use1.7 (1.3–2.4) c0.001[6]
At least one antibiotic (any class)1.3 (1.1–1.4) b<0.001[6]
Cephalosporins2.2 (1.3–3.8) b0.003[6]
2.1 (1.7–2.7)<0.001[7]
Third generation5.4 (3.0–9.8)<0.001[7]
Fourth generation2.0 (1.4–2.9)<0.001[7]
Glycopeptides3.2 (2.2–4.6)<0.001[6]
Fluoroquinolones1.4 (1.1–2.1)0.022[6]
1.6 (1.2–2.1)<0.001[7]
Meropenem1.7 (1.2–2.6)0.003[6]
Carbapenem4.8 (3.6–6.5)<0.001[7]
Clindamycin2.0 (1.4–2.9)<0.001[7]
Aminoglycoside2.7 (1.7–4.1)<0.001[7]
Tetracycline2.9 (1.3–6.3)0.005[7]
Linezolid2.3 (1.3–4.2)0.003[7]
Rifampicin3.4 (1.2–9.4)0.013[7]
Total duration of antibiotic therapy (≥15 days)3.7 (2.9–4.8)<0.001[7]
R-CDI a
Age ≥ 65 years1.6 (1.1–2.3)0.0012[14]
Additional non-CDI antibiotics during follow-up4.2 (2.1–8.5)0.001[14]
Proton pump inhibitors during follow-up2.14 (1.1–4.0)
1.6 (1.4–1.9)
0.019
NR
[14,15]
Nasogastric tube insertion8.7 (1.2–59.1)0.026[16]
Cardiovascular disease 3.0 (1.2–7.3)0.015[17]
Immunosuppressive comorbidities3.8 (1.3–11.2)0.012[17]
Dementia3.2 (1.2–8.4)0.014[17]
a Only risk factors were reported. Odds ratio values are included. b European multicenter, prospective, biannual, point-prevalence study of Clostridium difficile infection in hospitalized patients with diarrhea (EUCLID). c CDI cases/controls were identified from a single center in Germany (parallel study site).

2. Pathophysiology of C. difficile Infection

The CDI begins with the oral consumption of spores, which survive the acidic pH of the stomach and pass to the large intestine, where they interact with the primary and secondary bile acids that define whether the spore germinates or continues as a spore [18]. Once the spores germinate, the vegetative cells release enzymes, including collagenase, hyaluronidase, chondroitin sulfatase, enterotoxin A (TcdA), and cytotoxin (TcdB). These enzymes damage the cytoskeleton of intestinal cells [19], causing the condensation of cellular actin [20] and breaking the tight cell junctions between cells. This leads to fluid loss, local inflammation [19,21], and the destruction of the surface epithelium, causing the rounding and detachment of epithelial cells [20]. This process is associated with the infiltration of neutrophils into the submucosa and is mediated by several cytokines, including tumor necrosis factor alpha and interleukin-1β [22].
Additionally, it results in mast cell activation and the production of reactive oxygen species, and a pseudomembrane composed of neutrophils, fibrin, mucin, and cellular debris develops over the intestinal mucosa [21]. Overall, the extent of colonic damage is associated with the toxin concentration and duration of exposure before diagnosis [20]. A summary of the pathophysiology of CDI is presented in Figure 1.

3. Virulence Factors: Toxins and Spores

Virulence factors are structures or strategies of microbes that contribute to colonization and survival and may cause damage to the host [23]; they include secretory proteins such as toxins and spores.
Toxigenic strains of C. difficile contain a 19.6 kb chromosomal region known as the pathogenicity locus (PaLoc). This region comprises the genes encoding toxin A or TcdA (tcdA gene) and toxin B or TcdB (tcdB gene), as well as the accessory genes for TcdR, TcdE, TcdL, and TcdC proteins (tcdR, tcdE, tcdL, and tcdC genes). Toxin A and toxin B are glucosyltransferases with four domains, including an amino-terminal glucosyltransferase domain, an autoprotease, a translocation/pore-forming domain, and a C-terminal combined repetitive oligopeptide repeat domain (CROPS) [19,24,25]. Toxins A and B secreted in the colon bind to cell-surface glycans through the CROPS domain [25].
Approximately 20% of C. difficile isolates obtained from non-outbreak cases produce a third toxin called binary toxin, or C. difficile transferase (CDT) [26]. This toxin is encoded by the genes cdtA and cdtB, which are located in a 6.2 kb region known as the CDT locus (CdtLoc) [27,28]. CdtA is an ADP-ribosyl transferase that acts on actin, and CdtB forms a pore in acidified endosomes and facilitates the transfer of CdtA to the cytosol [27,28]. C. difficile is classified into 34 toxinotypes (I–XXXIV) based on changes in the PaLoc and CDT genes [29,30]. The toxinotypes represent changes in toxins A and B and, thus, the differences in functional properties and virulence [21,30,31,32].
Worldwide, the most frequent toxinotype in humans is toxinotype III (RT027- tcdA+ tcdB+ cdtA, and cdtB+), followed by toxinotype IV (RT023- tcdA+ tcdB+ cdtA, and cdtB+), V (RT078- tcdA+ tcdB+ cdtA, cdtB+), and VIII (RT017- tcdA- tcdB+ cdtA, cdtB- [30]. An increased disease severity associated with tcdA+ tcdB toxinotypes isolates has been reported, but the recurrence rate is similar to that involving tcdA+ tcdB+ toxinotypes [33,34,35].
Spore formation by C. difficile is crucial to the survival and dissemination of bacteria in the environment [36]. The various spore layers from the outside include the exosporium, coat, outer membrane, cortex, germ cell wall, inner membrane, and core [37]. The core of the spore is surrounded by an inner membrane, a peptidoglycan germ cell wall, and a large cortex layer composed of spore-specific modified crosslinked peptidoglycans that must be hydrolyzed in the spore germination and protect the bacteria against ethanol and heat. The cortex layer is surrounded by the outer membrane and the coat, which protects against enzymatic and chemical agents. Finally, there is a last layer known as the exosporium [38]. The spore of C. difficile is resistant to desiccation, numerous disinfectants, ultraviolet light, and antibiotics, allowing long-term survival against environmental insults and efficient transmission from host to host [39].

4. Biofilm

In addition to the ability to sporulate and produce toxins, other C. difficile virulence factors are associated with R-CDI, such as biofilm formation [40]. A biofilm is a community of bacteria organized and includes a single or multiple species growing attached or unattached [41] to a biotic or abiotic surface [42]. In a biofilm, cells are wrapped by an extracellular matrix composed of extracellular polymeric substances (EPSs), including polysaccharides, extracellular DNA (eDNA), proteins, glycoproteins, and glycolipids [43,44,45,46]. Bacteria may use biofilm as a protective barrier against multiple environmental stressors, such as antibiotics [40].

4.1. Biofilm Composition

C. difficile can form a multilayered biofilm in vitro with variable thicknesses depending on the duration and growth conditions [45]. Evidence regarding the chemical composition of C. difficile biofilms suggests that the proportions of the components may vary between strains and under different growth conditions. In a study that included 102 C. difficile isolates from different ribotypes grown in brain–heart infusion broth supplemented (BHIS) for 48 h, the main components reported were proteins [47] (Table 2). In contrast, when a biofilm of strain R20291 was grown in BHIS containing 0.1 M glucose for 48 h, the main component reported was eDNA [42]. In these studies, different models were used, and the results and findings cannot be compared. Several studies have shown that biofilm produced by C. difficile comprises proteins, eDNA, and polysaccharides [48,49,50].

4.2. Biofilm Formation

The biofilm formation process is often divided into three stages: initial attachment, maturation, and separation or dispersion [62]. The initial attachment stage of a biofilm, in which bacteria adhere to a surface, is mediated by different factors, including adhesins, flagella, pili, and fimbriae. This binding is initially reversible but becomes irreversible during maturation [45,62,63,64].
The maturation of biofilm is characterized by the production of an extracellular matrix [63], which forms the scaffolds of the biofilm structure and allows for interactions between the cells [45,65]. Moreover, it favors changes in the metabolism in response to the oxygen and nutrient gradient according to the location within the biofilm [65]. Stimuli such as changes in the microenvironment, antibiotic administration [45,63,64], QS, cyclic diguanosine monophosphate (cyclic-di-GMP) levels [63], and interactions with other bacterial species can favor biofilm sloughing and dispersal to colonize other sites [45,64].
An in vitro model of C. difficile biofilm on abiotic surfaces has shown that the number of spores inside the biofilm increases with time during biofilm maturation. Up to day 14 of growth, spores are considered the predominant form of C. difficile in the biofilm and are surrounded by extracellular EPSs [66,67].
The ability of C. difficile to produce biofilm in vivo was first described in 2017, using a germ-free model of CDI in an infected mouse with strains R20291 (RT 027), P30 (RT 014/020), and 630∆erm (RT 012) and the mutant 630∆erm cwp84::erm [68] (Table 2). In this study, the colonization levels by C. difficile in the jejunum and ileum were 100-fold lower than in the cecum and colon on day 7, and C. difficile bacteria were distributed heterogeneously over the intestinal tissue, without contact with epithelial cells. Bacterial cells were localized inside and outside the mucus layer, irrespective of the strains tested. Most bacterial cells of C. difficile were entrapped in 3D structures overlaying the mucus layer. For the R20291 strain, the PS-II was detected in large amounts in the 3D structure, suggesting that at least the R20291 strain is organized in the mouse model in glycan-rich biofilm architecture, which maintains bacteria outside the mucus layer [68].
C. difficile biofilms have been studied in various in vitro models, including microplates [51], black polycarbonate membranes [67], T-flasks [51,66], microfermenters [46] chemostats [40], and with different mutant strains (Table 2). According to these models, the C. difficile biofilm composition and structure depend on the incubation time [66], strain [66,69], and growth rate [42,67].
Recently, the transcriptomic profile in a biofilm model of RT001 and 027 associated with R-CDI and not associated with recurrent (NR)-CDI was analyzed to identify genes that may favor the recurrence using microarrays. In this study, CAJ70148, CAJ68100, CAJ69725, and CAJ68151 genes were differentially expressed in biofilm in strains associated with R-CDI; thus, they may support the biofilm favoring the recurrence of CDI [70].

4.3. Systems and Structures Associated with Adhesion and Biofilm Formation

Biofilm formation is a multistep and complex process, and approximately 20% of the C. difficile genome is expressed differently between biofilm and planktonic cells. Many of these genes participate in multiple pathways, and their expression changes according to the C. difficile strain, biofilm model conditions, biofilm stage, and incubation time [46]. Most genes involved in biofilm formation have been determined by directed mutation, its effect on increasing or reducing biofilm production, and the capacity to produce the disease [46].

4.3.1. Cell-Wall Proteins

C. difficile produces proteins that mediate bacterial adhesion to host cells [71], mucus layers [68], and other bacteria or surfaces and allow for attachment and colonization. Most of these proteins belong to the cell-wall protein family (Cwp), and some are involved in the formation of biofilms [48,72].
In the Cwp family, Cwp84 cleaves components of the extracellular matrix in eukaryotes, such as fibronectin, laminin, and vitronectin [48,73], and cleaves SlpA, which is necessary for achieving a paracrystalline arrangement that envelops the bacterial surface and anchors S-layer proteins [74,75]. Within the same family, Cwp66 exhibits 56% similarity to the N-acetylmuramoyl-L-alanine amidase of Bacillus subtilis [44,76].
In an in vitro model of C. difficile biofilm on a polycarbonate filter, in the first stage of biofilm formation, some planktonic cells of C. difficile undergo autolysis to produce eDNA and cellular debris [67,77]. Cwp19, a peptidoglycan hydrolase, probably triggers this process with its lytic transglycosylase activity [77] or through the differential expression of toxin-antitoxin systems, such as the MazE-MazFTA [78], CD2517.1-RCd8, CD2907.1-RCd9, and CD0956.2-RCd10 systems [53,79,80,81].

4.3.2. Quorum Sensing

QS is a complex communication system that allows bacteria to communicate with other cells from the same bacterial species or other species and is fundamental to the persistence, growth, and dispersion of bacterial cells [63]. QS in bacteria involves the production of self-secreted extracellular signaling molecules such as acyl homoserine lactones, autoinducers (AI), oligopeptides, diffusible signal factors, and autoinducing peptides (AIP) [82,83,84,85]. QS regulates cell-population density and regulates diverse bacterial processes, including C. difficile toxin production [86,87], activation of flagella [15,22,86], sporulation [26,88], and biofilm formation [49,56]. The bacterial cell detects the increase in AIs and triggers a signaling cascade that alters the gene expression [56]. The QS pathways partially identified in C. difficile are LuxS and the accessory gene regulator (Agr) system [89].

Lux S

The luxS is a 53 bp gene that encodes LuxS, an AI-2 synthase [90]. LuxS participates in the recycling pathway of methionine [91] by cleaving S-ribosylhomocysteine to form homocysteine [90] and 4,5-dihydroxy-2,3-pentanedione (DPD), an unstable furanone that spontaneously cyclizes into several different forms [90], collectively known as AI-2, a group of potent cross-species QS-signaling molecules [56,90,91]. The effect of luxS on biofilm production has been demonstrated in the luxS mutant strain R20291, which reduces biofilm production in vitro [42], with no difference in the number of cells or the production of spores detected. Additionally, it has been demonstrated that the biofilm phenotype can be restored by adding 100 nM DPD [56].
When the transcriptomic profile of mutant luxS was compared against the wild type, 21 genes were found to be differentially expressed: 2 were upregulated (2 involved in trehalose and one in glucose metabolism), and 18 were downregulated (2 prophage regions: CDR20291_1415–1464 and CDR20291_1197–1226). Transcriptomic analyses also revealed the downregulation of prophage loci in the luxS mutant biofilms compared to the wild type. The detection of phages and eDNA within biofilms suggests that DNA release by phage-mediated cell lysis contributes to C. difficile biofilm formation [56].

Agr

AIs can be synthesized through the Agr system, which encodes a transcriptional response regulator (AgrA), a protease (AgrB), a sensor histidine kinase (AgrC), and a signaling pre-peptide (AgrD). In C. difficile, three Agr systems, Agr1, Agr2, and Agr3, have been identified, with a different type of organization of the genes. It has been reported that C. difficile agr1 mutants affect the sporulation, motility, and toxin production of C. difficile strains 630 and R20291 [92].
In addition to agr1, agr2 locus is present in C. difficile strains (R20291, RT001, and RT017 [92]. C. difficile Agr2 is similar to S. aureus with genes of the AGR system (agrACDB arranged) in the inverse order of those found in the S. aureus Agr system (agrBDCA) 85 [93]. Most strains of C difficile exhibit an incomplete agr locus that contains agrDB and is called the agr1 locus. A third locus (agr3) has also been found in the C. difficile strains NAP07, NAP08, and QCD-23m63, all of which also encode agr1 [94]. The C. difficile Agr3 system consists of agrB3, agrD3, and agrC3 [95]. It has been reported that Agr3 seems to be encoded by a C. difficile bacteriophage phiCDHM1, suggesting the transmission between C. difficile strains [94]. The role of the Agr system in the biofilm formation of C. difficile is still poorly understood.
Gram-positives have a special QS system in which the receptor interacts with its cognate signaling peptide. The receptors are either Rap phosphatases or transcriptional regulators and integrate the protein family RNPP from Rap, Npr, PlcR, and PrgX. These systems control sporulation, virulence, biofilm formation, and the production of extracellular enzymes [96].

4.3.3. Cyclic di-GMP

It has been reported that, when biofilm and planktonic cells are compared, 751 genes are differentially expressed, with 338 upregulated in biofilm. These genes are involved in metabolic pathways, including T4P production: pilA1 [97] cell-wall biosynthesis [98]; and the production of a diguanylate cyclase, which is associated with the synthesis of the second messenger cyclic di-3′,5′-guanylate (cyclic-di-GMP) [46,51,53,59,99]. Cyclic di-GMP controls several cellular functions of C. difficile, including virulence, motility, and adhesion [59,100], and it participates in the posttranscriptional regulation of biofilm formation [51]. Cyclic di-GMP also upregulates 42 genes, with 37 involved in chemotaxis and flagellar motility [53]. Additionally, it favors a sessile lifestyle by modulating the attachment of cell-wall proteins to peptidoglycans [53,59,101]. Cyclic di-GMP represses the major operon flgB through a type I cyclic-di-GMP riboswitch (Cdi1-3), which reduces motility [51,53,59,101,102].

4.3.4. Type IV pili

It has been reported that type IV pili (T4P) favor biofilm production by adhesion to abiotic or biotic surfaces, colonization [44,103], twitching motility [59], and microcolony formation [104]. Generally, regulation via cyclic-di-GMP acts as an “on switch” for T4P genes [59], which are differentially expressed during biofilm growth.
In a 7-day biofilm (C. difficile R20291 strain), it has been reported that pilA1, pilJ, and pilW are highly expressed [97], with pilA1 being the most upregulated gene compared to planktonic cells. Furthermore, the piA1 mutant formed a thinner biofilm with lower biomass [44,46].

4.3.5. Flagella

The flagellum is a rotating semi-rigid helical filament anchored within the bacterial membranes and driven by the influx of protons or Na+ ions. It allows bacteria to move within fluid environments, including through liquid films on surfaces [105]. Flagella have been found to play a role in flagellum-dependent swimming motility, providing the organism an advantage over non-flagellated strains [106]. Flagella functions as an adhesin and a type III secretion system that can regulate virulence factors and is essential for the induction of proinflammatory responses and the invasion of host cells [106,107].
An in vitro model showed reduced biofilm production in the C. difficile R20219 strain (fliC mutant) after five days of incubation on a BHIS medium containing 0.1 M glucose 42. A second in vitro biofilm model in glass coverslips after 7 days in BHI broth showed that the expression of fliC significantly decreased in biofilm compared to planktonic cells [44]. It has been proposed that flagella-mediated motility is required in the late stages of biofilm formation [69].

5. The Role of Spores and Biofilms in CDI and R-CDI

It has been proposed that the internalization of spores in intestinal cells during infection and germination under favorable conditions could favor the recurrence or persistence of the infection [108]. Bacterial biofilm formation has been associated with chronic and recurrent infections [40,109]. The protection provided by the biofilm structure prevents the entry of antibiotics and favors the formation of persister cells [110]. Moreover, the exchange of resistance genes through plasmids, eDNA, and phages may be a source of recurrence cases or therapeutic failure [40,111].
It has been reported that C. difficile biofilm can survive treatment with vancomycin (VAN), thus promoting recurrence [40]. In a previous in vitro study, a biofilm formed with normal gut microbiota was treated with clindamycin and C. difficile spores. C. difficile was incorporated into the biofilm of the normal microbiota, and greater numbers of spores, vegetative cells, and toxins were detected. Next, C. difficile was depleted using VAN, but an increase in spores, vegetative cells, and toxins was observed 100 days later, which strongly suggested recurrence [40].
Furthermore, inside the biofilm of C. difficile strains associated with recurrence, increased expressions of the QS gene (agrD1), adhesion gene (cwp84), and sporulation pathway genes (sigH, spo0A) were found [112]. In another study, mice infected with the asporogenic phenotype of C. difficile (R20291Δspo0A) treated with VAN did not present recurrent CDI. Furthermore, the R20291Δspo0A demonstrated decreased biofilm formation in vitro [113].

6. Biofilm and Antibiotic Resistance

The clinical practice guideline by the Infectious Diseases Society of America and Society for Healthcare Epidemiology of America recommended, in 2021, the use of fidaxomicin (FDX), VAN, or metronidazole (MTZ) to treat CDI episodes. In the first or second recurrence, FDX or VAN is recommended, along with a pulsed regimen plus bezlotoxumab [4]. However, no treatment is 100% effective at reducing R-CDI [114].
Therapeutic failure in R-CDI has been associated with the inadequate concentration of drugs in the intestinal tract to eliminate C. difficile [114,115], the metabolism and inactivation by the microbiota [116], the ability to generate metabolically inactive spores, and the antibiotic resistance of a biofilm [112].
The capacity of antibiotics to kill planktonic cells differs from their ability to kill cells in a biofilm, and the effects of antibiotics on biofilms depend on the type and concentration of the antibiotic used [117,118]. The presence of eDNA, the development of persister cells, and the abundance of EPSs prevent the penetration of antibiotics into biofilms, limiting the activity of antibiotics on aging cells [117,118]. Moreover, the ability of an antibiotic to penetrate a biofilm depends on the bacterial species or strain, the antimicrobial agent, and the growth conditions of the biofilm [118].
The role of biofilm formation in resistance to antibiotics has been reported. In one of these studies, the susceptibility of C. difficile (planktonic cells) to VAN and MTZ was evaluated in 123 isolates. The production of biofilms was also assessed, and 44% were strong biofilm producers [119]. Most isolates with reduced susceptibility to MTZ were strong biofilm producers (63%, 17/27), whereas 22.2% were non-biofilm producers. Furthermore, 72.7% of isolates with reduced susceptibility to VAN were strong biofilm producers, and 51% of isolates susceptible to VAN were found to be non-biofilm producers [119].
It is important to evaluate the activity of the antibiotics used to treat CDI in C. difficile biofilms to reduce the recurrence rate and clinical duration of the primary infection. Table 3 summarizes the activity of the most-used antibiotics and a clinical phase III drug called surotomycin (STM) against C. difficile biofilms [2].

6.1. Fidaxomicin

FDX inhibits the RNAP [128] and is associated with a significantly lower recurrence rate of CDI [115]. An observational cohort study in 2013–2016 that included 271 patients with CDI revealed that FDX was better than MTZ, VAN, and MTZ plus VAN for the prevention of R-CDI. In this study, even using FDX, 6.3% of patients had R-CDI, and 33.3% had a second recurrence [129].
A previous study reported that FDX distributes into C. difficile biofilms in minutes [120]. Furthermore, subminimal inhibitory concentrations (sub-MICs) of FDX (0.03x, 0.25x, and 0.50x MIC) exhibit a dose-dependent lowering effect on biofilm formation. For example, at 0.50x MIC, FDX reduced planktonic growth and biofilm formation [121]. In this study, the biofilm structures were thick, with reduced biomass at sub-MICs of 0.09x and 0.25x MIC of FDX. FDX was more effective than MTZ at reducing C. difficile spore counts within biofilms, and this may explain its effectiveness with R-CDI [120]. In another study, 0.125x MIC of FDX was added during the stationary phase of a culture of C. difficile (C. difficile UK-14 and ATCC 43255 strains), and pre-existing spores were not eliminated at 0.25x but prevented the production of spores [122]. Finally, sub-MICs of FDX have been found to induce a dose-dependent reduction in the number of viable spores, with 0.25x MIC reducing spo0A transcription in 9689 and 5325 strains [123].
The significant advantage of FDX is the reduction of the risk of R-CDI when compared to VAN [114,115,130,131], probably due to declining biofilm production, planktonic cell-killing activity, and reduction in spore count.

6.2. Vancomycin

VAN is a tricyclic glycopeptide that binds to the D-Ala-D-Ala moiety of monomers. After this binding process, the monomers cross the cell membrane, interrupting the process of cell-wall synthesis [132]. The biofilm minimum inhibitory concentrations (BMICs) of C. difficile were analyzed in 102 clinical isolates, and we detected reduced susceptibility to VAN (91.0%) and linezolid (89.21%). It was also found that the BMIC was 100 times higher than the MIC for VAN and 20 times higher for linezolid in the biofilm state than in planktonic MICs [112].
The production of biofilms and the susceptibility to VAN and MTZ in 123 C. difficile isolates have been assessed: 53.6% were biofilm producers, of which 44% were strong biofilm producers [119]. Most isolates with reduced susceptibility to MTZ were strong biofilm producers (63%, 17/27), whereas 22.2% were non-producers. Furthermore, 72.7% of isolates with reduced susceptibility to VAN were strong biofilm producers, and 51% of isolates susceptible to VAN were found to be non-producers [119]. Many studies have demonstrated that the sub-MICs of VAN do not inhibit biofilm formation or planktonic growth. However, only 0.25x MIC of VAN has been found to significantly reduce the biomass of biofilms without the expression of genes involved in biofilm production, including pilA1, cwp84, luxS, dccA, and spo0A [121].
Furthermore, the effects of DNase and proteinase K on vegetative cells and spores in the biofilm and other states have been investigated [51]. A reduced viable vegetative cell count in intact biofilms was observed with 12.5 mg/mL VAN. In contrast, disruption with DNase combined with VAN treatment reduced the vegetative cell count to 0.68% of the untreated control. Furthermore, treatment with proteinase K combined with VAN reduced the viable vegetative cell count to only 72.8% of that of the untreated biofilm. Therefore, combining DNase and VAN is more efficient than using either alone to reduce the viable vegetative cell count in a biofilm [51]. Furthermore, VAN does not affect spore viability, irrespective of biofilm disruption [51].

6.3. Metronidazole

The mechanism of action of MTZ has not yet been fully elucidated. MTZ crosses the target cell membrane via passive diffusion [133]. In this process, the nitro group of the molecule is reduced to nitro radicals by ferredoxin or flavodoxin, generating toxic metabolites. These metabolites, such as N-(2-hydroxyethyl)-oxamic acid and acetamide, can react with DNA and form adducts with guanosine [134].
MTZ increases in vitro biofilm formation at 0.25x and 0.5x MICs in the C. difficile ribotype 010 and produces a thick biofilm composed of layered aggregates [121,126].
The effect of MTZ on the sporulation of C. difficile in liquid broths has been analyzed (ribotypes 001, 012, 037, 027, 018, 027, 014, and 018). However, the results revealed that, at 0.5x MIC, the level of sporulation was not affected in most strains [124]. In a similar study, sporulation was not inhibited in NAP1/BI/RT027 and CD196 strains growing with 0.25x MIC of MTZ [122]. In contrast, 0.25x, 0.125x, and 0.0625x MICs of MNZ were found to stimulate sporulation in the epidemic 5325 strain at 48 h (~2 log increase). However, all subinhibitory concentrations of MTZ suppressed spore formation in strain 9689 [123].
A proportion of MTZ administered orally may be degraded by the microbiota, yielding sub-MIC doses. It has also been found that the germination of spores and the production of cytotoxins increase when the MIC decreases in a chemostat model with strains RT001 and 027 [116]. Similarly, cells of the toxigenic C. difficile strains BI17 (PCR ribotype 027) and J9 grown as a biofilm have exhibited tolerance to MTZ concentrations as high as 100 μg mL−1. However, antibiotic concentrations above 1 μg mL−1 inhibited the same strains in liquid cultures, suggesting a 100-fold increase in resistance to this drug in the form of a biofilm [67].

7. Conclusions

The evidence suggests that biofilm formation in C. difficile is affected by multiple factors, including environmental and bacterial virulence factors, and that biofilm formation affects the recurrence of CDI.
Biofilm plays several roles that may favor recurrence, for example, allowing spores to remain inside the biofilm matrix and protecting the vegetative cells from the activity of antibiotics, even at concentrations higher than therapeutic doses.
Most antibiotics recommended for the treatment of CDI do not have activity on spores and do not eliminate biofilm. This makes it challenging to eradicate C. difficile in the intestine, thus complicating antibacterial therapies and allowing non-eliminated spores to remain in the biofilm, increasing the risk of recurrence.
According to one perspective, the deep analysis of molecules participating in QS may provide potential QS inhibitors that may be therapeutic alternatives.
Finally, no direct evidence links biofilm to CDI recurrence, so further studies are needed.

Author Contributions

D.R.-M. performed most of the writing and prepared tables; H.J.M.-G., C.C.-F. and A.M.-M. provided input in drafting the paper; and E.G.-G. coordinated the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lim, S.C.; Knight, D.R.; Riley, T.V. Clostridium difficile and One Health. Clin. Microbiol. Infect. 2020, 26, 857–863. [Google Scholar] [CrossRef] [PubMed]
  2. McDonald, L.C.; Gerding, D.N.; Johnson, S.; Bakken, J.S.; Carroll, K.C.; Coffin, S.E.; Dubberke, E.R.; Garey, K.W.; Gould, C.V.; Kelly, C.; et al. Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin. Infect. Dis. 2018, 66, e1–e48. [Google Scholar] [CrossRef] [PubMed]
  3. Abreu, Y.; Abreu, A.T.; Velarde-Ruiz Velasco, J.A.; Zavala-Solares, M.R.; Remes-Troche, J.M.; Carmona-Sánchez, R.I.; Aldana-Ledesma, J.M.; Camacho-Ortiz, A.; Contreras-Omaña, R.; Díaz-Seoane, R.; et al. Consensus on the prevention, diagnosis, and treatment of Clostridium difficile infection. Rev. Gastroenterol. Mex. 2019, 84, 204–219. [Google Scholar] [CrossRef]
  4. Johnson, S.; Lavergne, V.; Skinner, A.M.; Gonzales-Luna, A.J.; Garey, K.W.; Kelly, C.P.; Wilcox, M.H. Clinical Practice Guideline by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA): Focused Update Guidelines on Management of Clostridioides difficile Infection in Adults. Clin. Infect. Dis. 2021, 73, e1029–e1044. [Google Scholar] [CrossRef] [PubMed]
  5. Abou Chakra, C.N.; Pepin, J.; Sirard, S.; Valiquette, L. Risk factors for recurrence, complications and mortality in Clostridium difficile infection: A systematic review. PLoS ONE 2014, 9, e98400. [Google Scholar] [CrossRef]
  6. Davies, K.; Lawrence, J.; Berry, C.; Davis, G.; Yu, H.; Cai, B.; Gonzalez, E.; Prantner, I.; Kurcz, A.; Macovei, I.; et al. Risk Factors for Primary. Front. Public. Health 2020, 8, 293. [Google Scholar] [CrossRef]
  7. Yu, H.; Flaster, N.; Casanello, A.L.; Curcio, D. Assessing risk factors, mortality, and healthcare utilization associated with Clostridioides difficile infection in four Latin American countries. Braz. J. Infect. Dis. 2021, 25, 101040. [Google Scholar] [CrossRef]
  8. Wang, R. infection: Microbe-microbe interactions and live biotherapeutics. Front. Microbiol. 2023, 14, 1182612. [Google Scholar] [CrossRef]
  9. Cho, J.M.; Pardi, D.S.; Khanna, S. Update on Treatment of Clostridioides difficile Infection. Mayo Clin. Proc. 2020, 95, 758–769. [Google Scholar] [CrossRef]
  10. Baunwall, S.M.D.; Terveer, E.M.; Dahlerup, J.F.; Erikstrup, C.; Arkkila, P.; Vehreschild, M.J.; Ianiro, G.; Gasbarrini, A.; Sokol, H.; Kump, P.K.; et al. The use of Faecal Microbiota Transplantation (FMT) in Europe: A Europe-wide survey. Lancet Reg. Health Eur. 2021, 9, 100181. [Google Scholar] [CrossRef]
  11. Rounds, J.; Strain, J. Bezlotoxumab for Preventing Recurrent Clostridium difficile Infections. S D Med. 2017, 70, 422–423. [Google Scholar] [PubMed]
  12. Shakov, R.; Salazar, R.S.; Kagunye, S.K.; Baddoura, W.J.; DeBari, V.A. Diabetes mellitus as a risk factor for recurrence of Clostridium difficile infection in the acute care hospital setting. Am. J. Infect. Control 2011, 39, 194–198. [Google Scholar] [CrossRef] [PubMed]
  13. Zhong, S.; Yang, J.; Huang, H. The role of single and mixed biofilms in The role of single and mixed biofilms in Clostridioides difficile infection and strategies for prevention and inhibition. Crit. Rev. Microbiol. Crit. Rev. Microbiol. 2023, 9, e1003356. [Google Scholar] [CrossRef]
  14. Garey, K.W.; Sethi, S.; Yadav, Y.; DuPont, H.L. Meta-analysis to assess risk factors for recurrent Clostridium difficile infection. J. Hosp. Infect. 2008, 70, 298–304. [Google Scholar] [CrossRef]
  15. 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]
  16. Doh, Y.S.; Kim, Y.S.; Jung, H.J.; Park, Y.I.; Mo, J.W.; Sung, H.; Lee, K.J.; Seo, Y.K.; Moon, J.S.; Hong, S.W. Long-Term Clinical Outcome of Clostridium difficile Infection in Hospitalized Patients: A Single Center Study. Intest. Res. 2014, 12, 299–305. [Google Scholar] [CrossRef] [PubMed]
  17. Negrut, N.; Bungau, S.; Behl, T.; Khan, S.A.; Vesa, C.M.; Bustea, C.; Nistor-Cseppento, D.C.; Rus, M.; Pavel, F.M.; Tit, D.M. Risk Factors Associated with Recurrent. Healthcare 2020, 8, 352. [Google Scholar] [CrossRef] [PubMed]
  18. 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] [PubMed]
  19. Di Bella, S.; Ascenzi, P.; Siarakas, S.; Petrosillo, N.; di Masi, A. Clostridium difficile Toxins A and B: Insights into Pathogenic Properties and Extraintestinal Effects. Toxins 2016, 8, 134. [Google Scholar] [CrossRef] [PubMed]
  20. Riegler, M.; Sedivy, R.; Pothoulakis, C.; Hamilton, G.; Zacherl, J.; Bischof, G.; Cosentini, E.; Feil, W.; Schiessel, R.; LaMont, J.T. Clostridium difficile toxin B is more potent than toxin A in damaging human colonic epithelium in vitro. J. Clin. Invest. 1995, 95, 2004–2011. [Google Scholar] [CrossRef]
  21. Sun, X.; Savidge, T.; Feng, H. The enterotoxicity of Clostridium difficile toxins. Toxins 2010, 2, 1848–1880. [Google Scholar] [CrossRef] [PubMed]
  22. Rocha, M.F.; Maia, M.E.; Bezerra, L.R.; Lyerly, D.M.; Guerrant, R.L.; Ribeiro, R.A.; Lima, A.A. Clostridium difficile toxin A induces the release of neutrophil chemotactic factors from rat peritoneal macrophages: Role of interleukin-1beta, tumor necrosis factor alpha, and leukotrienes. Infect. Immun. 1997, 65, 2740–2746. [Google Scholar] [CrossRef] [PubMed]
  23. Hill, C. Virulence or niche factors: What’s in a name? J. Bacteriol. 2012, 194, 5725–5727. [Google Scholar] [CrossRef]
  24. Gupta, V.; Ye, G.; Olesky, M.; Lawrence, K.; Murray, J.; Yu, K. National prevalence estimates for resistant Enterobacteriaceae and Acinetobacter species in hospitalized patients in the United States. Int. J. Infect. Dis. 2019, 85, 203–211. [Google Scholar] [CrossRef] [PubMed]
  25. Kordus, S.L.; Thomas, A.K.; Lacy, D.B. Clostridioides difficile toxins: Mechanisms of action and antitoxin therapeutics. Nat. Rev. Microbiol. 2022, 20, 285–298. [Google Scholar] [CrossRef] [PubMed]
  26. Martin-Verstraete, I.; Peltier, J.; Dupuy, B. The Regulatory Networks That Control Clostridium difficile Toxin Synthesis. Toxins 2016, 8, 153. [Google Scholar] [CrossRef] [PubMed]
  27. Abt, M.C.; McKenney, P.T.; Pamer, E.G. Clostridium difficile colitis: Pathogenesis and host defence. Nat. Rev. Microbiol. 2016, 14, 609–620. [Google Scholar] [CrossRef] [PubMed]
  28. Beer, L.A.; Tatge, H.; Schneider, C.; Ruschig, M.; Hust, M.; Barton, J.; Thiemann, S.; Fühner, V.; Russo, G.; Gerhard, R. The Binary Toxin CDT of. Toxins 2018, 10, 225. [Google Scholar] [CrossRef]
  29. Rupnik, M.; Avesani, V.; Janc, M.; von Eichel-Streiber, C.; Delmée, M. A novel toxinotyping scheme and correlation of toxinotypes with serogroups of Clostridium difficile isolates. J. Clin. Microbiol. 1998, 36, 2240–2247. [Google Scholar] [CrossRef]
  30. Rupnik, M.; Janezic, S. An Update on Clostridium difficile Toxinotyping. J. Clin. Microbiol. 2016, 54, 13–18. [Google Scholar] [CrossRef]
  31. Knight, D.R.; Riley, T.V. Genomic Delineation of Zoonotic Origins of. Front. Public. Health 2019, 7, 164. [Google Scholar] [CrossRef] [PubMed]
  32. Knight, D.R.; Elliott, B.; Chang, B.J.; Perkins, T.T.; Riley, T.V. Diversity and Evolution in the Genome of Clostridium difficile. Clin. Microbiol. Rev. 2015, 28, 721–741. [Google Scholar] [CrossRef] [PubMed]
  33. Kyne, L.; Warny, M.; Qamar, A.; Kelly, C.P. Association between antibody response to toxin A and protection against recurrent Clostridium difficile diarrhoea. Lancet 2001, 357, 189–193. [Google Scholar] [CrossRef] [PubMed]
  34. Drudy, D.; Harnedy, N.; Fanning, S.; O’Mahony, R.; Kyne, L. Isolation and characterisation of toxin A-negative, toxin B-positive Clostridium difficile in Dublin, Ireland. Clin. Microbiol. Infect. 2007, 13, 298–304. [Google Scholar] [CrossRef]
  35. Drudy, D.; Fanning, S.; Kyne, L. Toxin A-negative, toxin B-positive Clostridium difficile. Int. J. Infect. Dis. 2007, 11, 5–10. [Google Scholar] [CrossRef]
  36. Lawler, A.J.; Lambert, P.A.; Worthington, T. A Revised Understanding of Clostridioides difficile Spore Germination. Trends Microbiol. 2020, 28, 744–752. [Google Scholar] [CrossRef]
  37. Setlow, P. Spore Resistance Properties. Microbiol. Spectr. 2014, 2, 201–215. [Google Scholar] [CrossRef]
  38. Paredes-Sabja, D.; Cid-Rojas, F.; Pizarro-Guajardo, M. Assembly of the exosporium layer in Clostridioides difficile spores. Curr. Opin. Microbiol. 2022, 67, 102137. [Google Scholar] [CrossRef]
  39. Lee, C.D.; Rizvi, A.; Edwards, A.N.; DiCandia, M.A.; Vargas Cuebas, G.G.; Monteiro, M.P.; McBride, S.M. Genetic mechanisms governing sporulation initiation in Clostridioides difficile. Curr. Opin. Microbiol. 2022, 66, 32–38. [Google Scholar] [CrossRef]
  40. Normington, C.; Moura, I.B.; Bryant, J.A.; Ewin, D.J.; Clark, E.V.; Kettle, M.J.; Harris, H.C.; Spittal, W.; Davis, G.; Henn, M.R.; et al. Biofilms harbour Clostridioides difficile, serving as a reservoir for recurrent infection. NPJ Biofilms Microbiomes 2021, 7, 16. [Google Scholar] [CrossRef]
  41. Rossi, E.; La Rosa, R.; Bartell, J.A.; Marvig, R.L.; Haagensen, J.A.J.; Sommer, L.M.; Molin, S.; Johansen, H.K. Pseudomonas aeruginosa adaptation and evolution in patients with cystic fibrosis. Nat. Rev. Microbiol. 2021, 19, 331–342. [Google Scholar] [CrossRef] [PubMed]
  42. Dapa, T.; Unnikrishnan, M. Biofilm formation by Clostridium difficile. Gut Microbes 2013, 4, 397–402. [Google Scholar] [CrossRef] [PubMed]
  43. Pantaléon, V.; Bouttier, S.; Soavelomandroso, A.P.; Janoir, C.; Candela, T. Biofilms of Clostridium species. Anaerobe 2014, 30, 193–198. [Google Scholar] [CrossRef] [PubMed]
  44. Maldarelli, G.A.; Piepenbrink, K.H.; Scott, A.J.; Freiberg, J.A.; Song, Y.; Achermann, Y.; Ernst, R.K.; Shirtliff, M.E.; Sundberg, E.J.; Donnenberg, M.S.; et al. Type IV pili promote early biofilm formation by Clostridium difficile. Pathog. Dis. 2016, 74, ftw061. [Google Scholar] [CrossRef]
  45. Beitelshees, M.; Hill, A.; Jones, C.H.; Pfeifer, B.A. Phenotypic Variation during Biofilm Formation: Implications for Anti-Biofilm Therapeutic Design. Materials 2018, 11, 1086. [Google Scholar] [CrossRef]
  46. Poquet, I.; Saujet, L.; Canette, A.; Monot, M.; Mihajlovic, J.; Ghigo, J.M.; Soutourina, O.; Briandet, R.; Martin-Verstraete, I.; Dupuy, B. Biofilm: Remodeling Metabolism and Cell Surface to Build a Sparse and Heterogeneously Aggregated Architecture. Front. Microbiol. 2018, 9, 2084. [Google Scholar] [CrossRef]
  47. Martínez-Meléndez, A.; Morfin-Otero, R.; Villarreal-Treviño, L.; Baines, S.D.; Camacho-Ortíz, A.; Garza-González, E. Analysis of biofilm production and expression of adhesion structures of circulating Clostridioides difficile strains from Mexico. Enferm Infecc. Microbiol. Clin. 2021, 40, 445–448. [Google Scholar] [CrossRef]
  48. Pantaléon, V.; Soavelomandroso, A.P.; Bouttier, S.; Briandet, R.; Roxas, B.; Chu, M.; Collignon, A.; Janoir, C.; Vedantam, G.; Candela, T. The Clostridium difficile Protease CwpModulates both Biofilm Formation and Cell-Surface Properties. PLoS ONE 2015, 10, e0124971. [Google Scholar] [CrossRef]
  49. Ðapa, T.; Leuzzi, R.; Ng, Y.K.; Baban, S.T.; Adamo, R.; Kuehne, S.A.; Scarselli, M.; Minton, N.P.; Serruto, D.; Unnikrishnan, M. Multiple factors modulate biofilm formation by the anaerobic pathogen Clostridium difficile. J. Bacteriol. 2013, 195, 545–555. [Google Scholar] [CrossRef]
  50. Frost, L.R.; Cheng, J.K.J.; Unnikrishnan, M. Clostridioides difficile biofilms: A mechanism of persistence in the gut? PLoS Pathog. 2021, 17, e1009348. [Google Scholar] [CrossRef]
  51. Dawson, L.F.; Peltier, J.; Hall, C.L.; Harrison, M.A.; Derakhshan, M.; Shaw, H.A.; Fairweather, N.F.; Wren, B.W. Extracellular DNA, cell surface proteins and c-di-GMP promote biofilm formation in Clostridioides difficile. Sci. Rep. 2021, 11, 3244. [Google Scholar] [CrossRef] [PubMed]
  52. Chu, M.; Mallozzi, M.J.; Roxas, B.P.; Bertolo, L.; Monteiro, M.A.; Agellon, A.; Viswanathan, V.K.; Vedantam, G. A Clostridium difficile Cell Wall Glycopolymer Locus Influences Bacterial Shape, Polysaccharide Production and Virulence. PLoS Pathog. 2016, 12, e1005946. [Google Scholar] [CrossRef] [PubMed]
  53. Soutourina, O.A.; Monot, M.; Boudry, P.; Saujet, L.; Pichon, C.; Sismeiro, O.; Semenova, E.; Severinov, K.; Le Bouguenec, C.; Coppée, J.Y.; et al. Genome-wide identification of regulatory RNAs in the human pathogen Clostridium difficile. PLoS Genet. 2013, 9, e1003493. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, K.Y.; Rathod, J.; Chiu, Y.C.; Chen, J.W.; Tsai, P.J.; Huang, I.H. The Transcriptional Regulator Lrp Contributes to Toxin Expression, Sporulation, and Swimming Motility in. Front. Cell Infect. Microbiol. 2019, 9, 356. [Google Scholar] [CrossRef] [PubMed]
  55. Walter, B.M.; Cartman, S.T.; Minton, N.P.; Butala, M.; Rupnik, M. The SOS Response Master Regulator LexA Is Associated with Sporulation, Motility and Biofilm Formation in Clostridium difficile. PLoS ONE 2015, 10, e0144763. [Google Scholar] [CrossRef] [PubMed]
  56. Slater, R.T.; Frost, L.R.; Jossi, S.E.; Millard, A.D.; Unnikrishnan, M. Clostridioides difficile LuxS mediates inter-bacterial interactions within biofilms. Sci. Rep. 2019, 9, 9903. [Google Scholar] [CrossRef]
  57. Jain, S.; Smyth, D.; O’Hagan, B.M.G.; Heap, J.T.; McMullan, G.; Minton, N.P.; Ternan, N.G. Inactivation of the dnaK gene in Clostridium difficile 630 Δerm yields a temperature-sensitive phenotype and increases biofilm-forming ability. Sci. Rep. 2017, 7, 17522. [Google Scholar] [CrossRef]
  58. Dubois, T.; Tremblay, Y.D.N.; Hamiot, A.; Martin-Verstraete, I.; Deschamps, J.; Monot, M.; Briandet, R.; Dupuy, B. A microbiota-generated bile salt induces biofilm formation in. NPJ Biofilms Microbiomes 2019, 5, 14. [Google Scholar] [CrossRef]
  59. Garrett, E.M.; Sekulovic, O.; Wetzel, D.; Jones, J.B.; Edwards, A.N.; Vargas-Cuebas, G.; McBride, S.M.; Tamayo, R. Phase variation of a signal transduction system controls Clostridioides difficile colony morphology, motility, and virulence. PLoS Biol. 2019, 17, e3000379. [Google Scholar] [CrossRef]
  60. Cuenot, E.; Garcia-Garcia, T.; Douche, T.; Gorgette, O.; Courtin, P.; Denis-Quanquin, S.; Hoys, S.; Tremblay, Y.D.N.; Matondo, M.; Chapot-Chartier, M.P.; et al. The Ser/Thr Kinase PrkC Participates in Cell Wall Homeostasis and Antimicrobial Resistance in Clostridium difficile. Infect. Immun. 2019, 87, e00005-19. [Google Scholar] [CrossRef]
  61. Tremblay, Y.D.N.; Durand, B.A.R.; Hamiot, A.; Martin-Verstraete, I.; Oberkampf, M.; Monot, M.; Dupuy, B. Metabolic adaption to extracellular pyruvate triggers biofilm formation in Clostridioides difficile. ISME J. 2021, 15, 3623–3635. [Google Scholar] [CrossRef]
  62. Schulze, A.; Mitterer, F.; Pombo, J.P.; Schild, S. Biofilms by bacterial human pathogens: Clinical relevance—Development, composition and regulation—Therapeutical strategies. Microb. Cell 2021, 8, 28–56. [Google Scholar] [CrossRef] [PubMed]
  63. Martínez, L.C.; Vadyvaloo, V. Mechanisms of post-transcriptional gene regulation in bacterial biofilms. Front. Cell Infect. Microbiol. 2014, 4, 38. [Google Scholar] [CrossRef] [PubMed]
  64. Rabin, N.; Zheng, Y.; Opoku-Temeng, C.; Du, Y.; Bonsu, E.; Sintim, H.O. Biofilm formation mechanisms and targets for developing antibiofilm agents. Future Med. Chem. 2015, 7, 493–512. [Google Scholar] [CrossRef] [PubMed]
  65. Stewart, P.S.; Franklin, M.J. Physiological heterogeneity in biofilms. Nat. Rev. Microbiol. 2008, 6, 199–210. [Google Scholar] [CrossRef] [PubMed]
  66. Dawson, L.F.; Valiente, E.; Faulds-Pain, A.; Donahue, E.H.; Wren, B.W. Characterisation of Clostridium difficile biofilm formation, a role for Spo0A. PLoS ONE 2012, 7, e50527. [Google Scholar] [CrossRef]
  67. Semenyuk, E.G.; Laning, M.L.; Foley, J.; Johnston, P.F.; Knight, K.L.; Gerding, D.N.; Driks, A. Spore formation and toxin production in Clostridium difficile biofilms. PLoS ONE 2014, 9, e87757. [Google Scholar] [CrossRef]
  68. Soavelomandroso, A.P.; Gaudin, F.; Hoys, S.; Nicolas, V.; Vedantam, G.; Janoir, C.; Bouttier, S. Biofilm Structures in a Mono-Associated Mouse Model of. Front. Microbiol. 2017, 8, 2086. [Google Scholar] [CrossRef]
  69. Pantaléon, V.; Monot, M.; Eckert, C.; Hoys, S.; Collignon, A.; Janoir, C.; Candela, T. Clostridium difficile forms variable biofilms on abiotic surface. Anaerobe 2018, 53, 34–37. [Google Scholar] [CrossRef]
  70. Rubio-Mendoza, D.; Córdova-Fletes, C.; Martínez-Meléndez, A.; Morfín-Otero, R.; Maldonado-Garza, H.J.; Garza-González, E. Transcriptomic analysis of biofilm formation in strains of Clostridioides difficile associated with recurrent and non-recurrent infection reveals potential candidate markers for recurrence. PLoS ONE 2023, 18, e0289593. [Google Scholar] [CrossRef]
  71. Paredes-Sabja, D.; Sarker, M.R. Adherence of Clostridium difficile spores to Caco-2 cells in culture. J. Med. Microbiol. 2012, 61, 1208–1218. [Google Scholar] [CrossRef] [PubMed]
  72. Fagan, R.P.; Janoir, C.; Collignon, A.; Mastrantonio, P.; Poxton, I.R.; Fairweather, N.F. A proposed nomenclature for cell wall proteins of Clostridium difficile. J. Med. Microbiol. 2011, 60, 1225–1228. [Google Scholar] [CrossRef] [PubMed]
  73. Janoir, C.; Péchiné, S.; Grosdidier, C.; Collignon, A. Cwp84, a surface-associated protein of Clostridium difficile, is a cysteine protease with degrading activity on extracellular matrix proteins. J. Bacteriol. 2007, 189, 7174–7180. [Google Scholar] [CrossRef] [PubMed]
  74. Calabi, E.; Ward, S.; Wren, B.; Paxton, T.; Panico, M.; Morris, H.; Dell, A.; Dougan, G.; Fairweather, N. Molecular characterization of the surface layer proteins from Clostridium difficile. Mol. Microbiol. 2001, 40, 1187–1199. [Google Scholar] [CrossRef]
  75. Kirby, J.M.; Ahern, H.; Roberts, A.K.; Kumar, V.; Freeman, Z.; Acharya, K.R.; Shone, C.C. Cwp84, a surface-associated cysteine protease, plays a role in the maturation of the surface layer of Clostridium difficile. J. Biol. Chem. 2009, 284, 34666–34673. [Google Scholar] [CrossRef]
  76. Waligora, A.J.; Hennequin, C.; Mullany, P.; Bourlioux, P.; Collignon, A.; Karjalainen, T. Characterization of a cell surface protein of Clostridium difficile with adhesive properties. Infect. Immun. 2001, 69, 2144–2153. [Google Scholar] [CrossRef]
  77. Wydau-Dematteis, S.; El Meouche, I.; Courtin, P.; Hamiot, A.; Lai-Kuen, R.; Saubaméa, B.; Fenaille, F.; Butel, M.J.; Pons, J.L.; Dupuy, B.; et al. CwpIs a Novel Lytic Transglycosylase Involved in Stationary-Phase Autolysis Resulting in Toxin Release in. mBio 2018, 9, e00648-18. [Google Scholar] [CrossRef]
  78. Rothenbacher, F.P.; Suzuki, M.; Hurley, J.M.; Montville, T.J.; Kirn, T.J.; Ouyang, M.; Woychik, N.A. Clostridium difficile MazF toxin exhibits selective, not global, mRNA cleavage. J. Bacteriol. 2012, 194, 3464–3474. [Google Scholar] [CrossRef]
  79. Maikova, A.; Peltier, J.; Boudry, P.; Hajnsdorf, E.; Kint, N.; Monot, M.; Poquet, I.; Martin-Verstraete, I.; Dupuy, B.; Soutourina, O. Discovery of new type I toxin-antitoxin systems adjacent to CRISPR arrays in Clostridium difficile. Nucleic Acids Res. 2018, 46, 4733–4751. [Google Scholar] [CrossRef]
  80. Soutourina, O. Type I Toxin-Antitoxin Systems in Clostridia. Toxins 2019, 11, 253. [Google Scholar] [CrossRef]
  81. Peltier, J.; Hamiot, A.; Garneau, J.R.; Boudry, P.; Maikova, A.; Hajnsdorf, E.; Fortier, L.C.; Dupuy, B.; Soutourina, O. Type I toxin-antitoxin systems contribute to the maintenance of mobile genetic elements in Clostridioides difficile. Commun. Biol. 2020, 3, 718. [Google Scholar] [CrossRef] [PubMed]
  82. Parsek, M.R.; Greenberg, E.P. Acyl-homoserine lactone quorum sensing in gram-negative bacteria: A signaling mechanism involved in associations with higher organisms. Proc. Natl. Acad. Sci. USA 2000, 97, 8789–8793. [Google Scholar] [CrossRef] [PubMed]
  83. McBrayer, D.N.; Cameron, C.D.; Tal-Gan, Y. Development and utilization of peptide-based quorum sensing modulators in Gram-positive bacteria. Org. Biomol. Chem. 2020, 18, 7273–7290. [Google Scholar] [CrossRef] [PubMed]
  84. Sturme, M.H.; Kleerebezem, M.; Nakayama, J.; Akkermans, A.D.; Vaugha, E.E.; de Vos, W.M. Cell to cell communication by autoinducing peptides in gram-positive bacteria. Antonie Van. Leeuwenhoek 2002, 81, 233–243. [Google Scholar] [CrossRef] [PubMed]
  85. Ahmed, U.K.B.; Ballard, J.D. Autoinducing peptide-based quorum signaling systems in Clostridioides difficile. Curr. Opin. Microbiol. 2022, 65, 81–86. [Google Scholar] [CrossRef]
  86. Martin, M.J.; Clare, S.; Goulding, D.; Faulds-Pain, A.; Barquist, L.; Browne, H.P.; Pettit, L.; Dougan, G.; Lawley, T.D.; Wren, B.W. The agr locus regulates virulence and colonization genes in Clostridium difficile 027. J. Bacteriol. 2013, 195, 3672–3681. [Google Scholar] [CrossRef]
  87. Lee, A.S.; Song, K.P. LuxS/autoinducer-2 quorum sensing molecule regulates transcriptional virulence gene expression in Clostridium difficile. Biochem. Biophys. Res. Commun. 2005, 335, 659–666. [Google Scholar] [CrossRef]
  88. Zhu, D.; Sorg, J.A.; Sun, X. Biology: Sporulation, Germination, and Corresponding Therapies for. Front. Cell Infect. Microbiol. 2018, 8, 29. [Google Scholar] [CrossRef]
  89. Gunaratnam, S.; Millette, M.; McFarland, L.V.; DuPont, H.L.; Lacroix, M. Potential role of probiotics in reducing Clostridioides difficile virulence: Interference with quorum sensing systems. Microb. Pathog. 2021, 153, 104798. [Google Scholar] [CrossRef]
  90. Schauder, S.; Shokat, K.; Surette, M.G.; Bassler, B.L. The LuxS family of bacterial autoinducers: Biosynthesis of a novel quorum-sensing signal molecule. Mol. Microbiol. 2001, 41, 463–476. [Google Scholar] [CrossRef]
  91. Carter, G.P.; Purdy, D.; Williams, P.; Minton, N.P. Quorum sensing in Clostridium difficile: Analysis of a luxS-type signalling system. J. Med. Microbiol. 2005, 54, 119–127. [Google Scholar] [CrossRef] [PubMed]
  92. Ahmed, U.K.B.; Shadid, T.M.; Larabee, J.L.; Ballard, J.D. Combined and Distinct Roles of Agr Proteins in Clostridioides difficile Sporulation, Motility, and Toxin Production. mBio 2020, 11, e03190-20. [Google Scholar] [CrossRef] [PubMed]
  93. Sidote, D.J.; Barbieri, C.M.; Wu, T.; Stock, A.M. Structure of the Staphylococcus aureus AgrA LytTR domain bound to DNA reveals a beta fold with an unusual mode of binding. Structure 2008, 16, 727–735. [Google Scholar] [CrossRef]
  94. Hargreaves, K.R.; Kropinski, A.M.; Clokie, M.R. What does the talking?: Quorum sensing signalling genes discovered in a bacteriophage genome. PLoS ONE 2014, 9, e85131. [Google Scholar] [CrossRef] [PubMed]
  95. Darkoh, C.; DuPont, H.L.; Norris, S.J.; Kaplan, H.B. Toxin synthesis by Clostridium difficile is regulated through quorum signaling. mBio 2015, 6, e02569. [Google Scholar] [CrossRef] [PubMed]
  96. Rocha-Estrada, J.; Aceves-Diez, A.E.; Guarneros, G.; de la Torre, M. The RNPP family of quorum-sensing proteins in Gram-positive bacteria. Appl. Microbiol. Biotechnol. 2010, 87, 913–923. [Google Scholar] [CrossRef]
  97. Maldarelli, G.A.; De Masi, L.; von Rosenvinge, E.C.; Carter, M.; Donnenberg, M.S. Identification, immunogenicity, and cross-reactivity of type IV pilin and pilin-like proteins from Clostridium difficile. Pathog. Dis. 2014, 71, 302–314. [Google Scholar] [CrossRef]
  98. Siegel, S.D.; Amer, B.R.; Wu, C.; Sawaya, M.R.; Gosschalk, J.E.; Clubb, R.T.; Ton-That, H. Structure and Mechanism of LcpA, a Phosphotransferase That Mediates Glycosylation of a Gram-Positive Bacterial Cell Wall-Anchored Protein. mBio 2019, 10, e01580-18. [Google Scholar] [CrossRef]
  99. McKee, R.W.; Aleksanyan, N.; Garrett, E.M.; Tamayo, R. Type IV Pili Promote Clostridium difficile Adherence and Persistence in a Mouse Model of Infection. Infect. Immun. 2018, 86, e00943-17. [Google Scholar] [CrossRef]
  100. Vuotto, C.; Donelli, G.; Buckley, A.; Chilton, C. Clostridium difficile Biofilm. Adv. Exp. Med. Biol. 2018, 1050, 97–115. [Google Scholar] [CrossRef]
  101. Bordeleau, E.; Fortier, L.C.; Malouin, F.; Burrus, V. c-di-GMP turn-over in Clostridium difficile is controlled by a plethora of diguanylate cyclases and phosphodiesterases. PLoS Genet. 2011, 7, e1002039. [Google Scholar] [CrossRef] [PubMed]
  102. Purcell, E.B.; McKee, R.W.; McBride, S.M.; Waters, C.M.; Tamayo, R. Cyclic diguanylate inversely regulates motility and aggregation in Clostridium difficile. J. Bacteriol. 2012, 194, 3307–3316. [Google Scholar] [CrossRef] [PubMed]
  103. Craig, L.; Pique, M.E.; Tainer, J.A. Type IV pilus structure and bacterial pathogenicity. Nat. Rev. Microbiol. 2004, 2, 363–378. [Google Scholar] [CrossRef] [PubMed]
  104. Kirn, T.J.; Lafferty, M.J.; Sandoe, C.M.; Taylor, R.K. Delineation of pilin domains required for bacterial association into microcolonies and intestinal colonization by Vibrio cholerae. Mol. Microbiol. 2000, 35, 896–910. [Google Scholar] [CrossRef]
  105. Diepold, A.; Armitage, J.P. Type III secretion systems: The bacterial flagellum and the injectisome. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20150020. [Google Scholar] [CrossRef]
  106. Stevenson, E.; Minton, N.P.; Kuehne, S.A. The role of flagella in Clostridium difficile pathogenicity. Trends Microbiol. 2015, 23, 275–282. [Google Scholar] [CrossRef]
  107. Duan, Q.; Zhou, M.; Zhu, L.; Zhu, G. Flagella and bacterial pathogenicity. J. Basic. Microbiol. 2013, 53, 1–8. [Google Scholar] [CrossRef]
  108. Castro-Córdova, P.; Mora-Uribe, P.; Reyes-Ramírez, R.; Cofré-Araneda, G.; Orozco-Aguilar, J.; Brito-Silva, C.; Mendoza-León, M.J.; Kuehne, S.A.; Minton, N.P.; Pizarro-Guajardo, M.; et al. Entry of spores into intestinal epithelial cells contributes to recurrence of Clostridioides difficile infection. Nat. Commun. 2021, 12, 1140. [Google Scholar] [CrossRef]
  109. Wu, H.; Moser, C.; Wang, H.Z.; Høiby, N.; Song, Z.J. Strategies for combating bacterial biofilm infections. Int. J. Oral. Sci. 2015, 7, 1–7. [Google Scholar] [CrossRef]
  110. Fisher, R.A.; Gollan, B.; Helaine, S. Persistent bacterial infections and persister cells. Nat. Rev. Microbiol. 2017, 15, 453–464. [Google Scholar] [CrossRef]
  111. Heuler, J.; Fortier, L.C.; Sun, X. Clostridioides difficile phage biology and application. FEMS Microbiol. Rev. 2021, 45, fuab012. [Google Scholar] [CrossRef] [PubMed]
  112. Tijerina-Rodríguez, L.; Villarreal-Treviño, L.; Baines, S.D.; Morfín-Otero, R.; Camacho-Ortíz, A.; Flores-Treviño, S.; Maldonado-Garza, H.; Rodríguez-Noriega, E.; Garza-González, E. High sporulation and overexpression of virulence factors in biofilms and reduced susceptibility to vancomycin and linezolid in recurrent Clostridium [Clostridioides] difficile infection isolates. PLoS ONE 2019, 14, e0220671. [Google Scholar] [CrossRef] [PubMed]
  113. Deakin, L.J.; Clare, S.; Fagan, R.P.; Dawson, L.F.; Pickard, D.J.; West, M.R.; Wren, B.W.; Fairweather, N.F.; Dougan, G.; Lawley, T.D. The Clostridium difficile spo0A gene is a persistence and transmission factor. Infect. Immun. 2012, 80, 2704–2711. [Google Scholar] [CrossRef] [PubMed]
  114. Mullane, K. Fidaxomicin in Clostridium difficile infection: Latest evidence and clinical guidance. Ther. Adv. Chronic Dis. 2014, 5, 69–84. [Google Scholar] [CrossRef]
  115. Louie, T.J.; Miller, M.A.; Mullane, K.M.; Weiss, K.; Lentnek, A.; Golan, Y.; Gorbach, S.; Sears, P.; Shue, Y.K.; OPT-80-003 Clinical Study Group. Fidaxomicin versus vancomycin for Clostridium difficile infection. N. Engl. J. Med. 2011, 364, 422–431. [Google Scholar] [CrossRef]
  116. Freeman, J.; Baines, S.D.; Saxton, K.; Wilcox, M.H. Effect of metronidazole on growth and toxin production by epidemic Clostridium difficile PCR ribotypes 001 and 027 in a human gut model. J. Antimicrob. Chemother. 2007, 60, 83–91. [Google Scholar] [CrossRef]
  117. Lewis, K. Riddle of biofilm resistance. Antimicrob. Agents Chemother. 2001, 45, 999–1007. [Google Scholar] [CrossRef]
  118. Hall, C.W.; Mah, T.F. Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol. Rev. 2017, 41, 276–301. [Google Scholar] [CrossRef]
  119. Rahmoun, L.A.; Azrad, M.; Peretz, A. Antibiotic Resistance and Biofilm Production Capacity in. Front. Cell Infect. Microbiol. 2021, 11, 683464. [Google Scholar] [CrossRef]
  120. James, G.A.; Chesnel, L.; Boegli, L.; deLancey Pulcini, E.; Fisher, S.; Stewart, P.S. Analysis of Clostridium difficile biofilms: Imaging and antimicrobial treatment. J. Antimicrob. Chemother. 2018, 73, 102–108. [Google Scholar] [CrossRef]
  121. Hamada, M.; Yamaguchi, T.; Ishii, Y.; Chono, K.; Tateda, K. Inhibitory effect of fidaxomicin on biofilm formation in Clostridioides difficile. J. Infect. Chemother. 2020, 26, 685–692. [Google Scholar] [CrossRef] [PubMed]
  122. Babakhani, F.; Bouillaut, L.; Gomez, A.; Sears, P.; Nguyen, L.; Sonenshein, A.L. Fidaxomicin inhibits spore production in Clostridium difficile. Clin. Infect. Dis. 2012, 55, S162–S169. [Google Scholar] [CrossRef] [PubMed]
  123. Aldape, M.J.; Packham, A.E.; Heeney, D.D.; Rice, S.N.; Bryant, A.E.; Stevens, D.L. Fidaxomicin reduces early toxin A and B production and sporulation in Clostridium difficile in vitro. J. Med. Microbiol. 2017, 66, 1393–1399. [Google Scholar] [CrossRef]
  124. Garneau, J.R.; Valiquette, L.; Fortier, L.C. Prevention of Clostridium difficile spore formation by sub-inhibitory concentrations of tigecycline and piperacillin/tazobactam. BMC Infect. Dis. 2014, 14, 29. [Google Scholar] [CrossRef] [PubMed]
  125. Allen, C.A.; Babakhani, F.; Sears, P.; Nguyen, L.; Sorg, J.A. Both fidaxomicin and vancomycin inhibit outgrowth of Clostridium difficile spores. Antimicrob. Agents Chemother. 2013, 57, 664–667. [Google Scholar] [CrossRef]
  126. Vuotto, C.; Moura, I.; Barbanti, F.; Donelli, G.; Spigaglia, P. Subinhibitory concentrations of metronidazole increase biofilm formation in Clostridium difficile strains. Pathog. Dis. 2016, 74, ftv114. [Google Scholar] [CrossRef]
  127. Bouillaut, L.; McBride, S.; Sorg, J.A.; Schmidt, D.J.; Suarez, J.M.; Tzipori, S.; Mascio, C.; Chesnel, L.; Sonenshein, A.L. Effects of surotomycin on Clostridium difficile viability and toxin production in vitro. Antimicrob. Agents Chemother. 2015, 59, 4199–4205. [Google Scholar] [CrossRef]
  128. Artsimovitch, I.; Seddon, J.; Sears, P. Fidaxomicin is an inhibitor of the initiation of bacterial RNA synthesis. Clin. Infect. Dis. 2012, 55, S127–S131. [Google Scholar] [CrossRef]
  129. Polivkova, S.; Krutova, M.; Capek, V.; Sykorova, B.; Benes, J. Fidaxomicin versus metronidazole, vancomycin and their combination for initial episode, first recurrence and severe Clostridioides difficile infection—An observational cohort study. Int. J. Infect. Dis. 2021, 103, 226–233. [Google Scholar] [CrossRef]
  130. Louie, T.J.; Miller, M.A.; Crook, D.W.; Lentnek, A.; Bernard, L.; High, K.P.; Shue, Y.K.; Gorbach, S.L. Effect of age on treatment outcomes in Clostridium difficile infection. J. Am. Geriatr. Soc. 2013, 61, 222–230. [Google Scholar] [CrossRef]
  131. Cornely, O.A.; Crook, D.W.; Esposito, R.; Poirier, A.; Somero, M.S.; Weiss, K.; Sears, P.; Gorbach, S.; OPT-80-004 Clinical Study Group. Fidaxomicin versus vancomycin for infection with Clostridium difficile in Europe, Canada, and the USA: A double-blind, non-inferiority, randomised controlled trial. Lancet Infect. Dis. 2012, 12, 281–289. [Google Scholar] [CrossRef] [PubMed]
  132. Rubinstein, E.; Keynan, Y. Vancomycin revisited—60 years later. Front. Public. Health 2014, 2, 217. [Google Scholar] [CrossRef] [PubMed]
  133. Hernández Ceruelos, A.; Romero-Quezada, L.C.; Ruvalcaba Ledezma, J.C.; López Contreras, L. Therapeutic uses of metronidazole and its side effects: An update. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 397–401. [Google Scholar] [CrossRef] [PubMed]
  134. Dingsdag, S.A.; Hunter, N. Metronidazole: An update on metabolism, structure-cytotoxicity and resistance mechanisms. J. Antimicrob. Chemother. 2018, 73, 265–279. [Google Scholar] [CrossRef]
Figure 1. Pathophysiology of Clostridioides difficile infection. (a) The infection begins with the consumption of spores. (b) Spores in the intestine can interact with primary bile salts that prevent its germination or with secondary bile salts and germinate. (c) Antibiotics kill the planktonic cells of C. difficile. (d) Antibiotics can inhibit the formation of spores. (e) Planktonic cells secrete toxins that affect the cytoskeleton of the intestinal cell and adhere to the colon mucous layer. (f) Planktonic cells can form a biofilm-like structure, hindering the action of antibiotics on the cells embedded in the biofilm. (g) Biofilm can be dispersed to other sites and colonized. (h) Spores can be internalized in intestinal epithelial cells. (i) It has been suggested that spores can disperse to other sites to colonize. Adapted from “Structure of Mucosal Barrier” from BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 9 May 2023).
Figure 1. Pathophysiology of Clostridioides difficile infection. (a) The infection begins with the consumption of spores. (b) Spores in the intestine can interact with primary bile salts that prevent its germination or with secondary bile salts and germinate. (c) Antibiotics kill the planktonic cells of C. difficile. (d) Antibiotics can inhibit the formation of spores. (e) Planktonic cells secrete toxins that affect the cytoskeleton of the intestinal cell and adhere to the colon mucous layer. (f) Planktonic cells can form a biofilm-like structure, hindering the action of antibiotics on the cells embedded in the biofilm. (g) Biofilm can be dispersed to other sites and colonized. (h) Spores can be internalized in intestinal epithelial cells. (i) It has been suggested that spores can disperse to other sites to colonize. Adapted from “Structure of Mucosal Barrier” from BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 9 May 2023).
Microorganisms 11 02525 g001
Table 2. Effects of gene mutations in C. difficile biofilm.
Table 2. Effects of gene mutations in C. difficile biofilm.
Strain Biofilm Model Biofilm EffectRef.
R20291::fliC430s24-WPP; BHISB + 0.1 M glucose; 120 hDecreased biofilm production on day 5[42]
R20291::luxS161a24-WPP; BHISB + 0.1 M glucose; 72 hDecreased biofilm production. Unable to form even a bacterial monolayer [42]
R20291::sleC128a24-WPP; BHISB + 0.1 M glucose; 72 hForm thick biofilm-like structures[42]
R20291::spo0A178a24-WPP; BHISB + 0.1 M glucose; 24, 72, 120 hDecreased biofilm formation. Form uneven and thick biofilm-like structures. Cellular form filamentous structures[42]
R20291Δcwp8424-WPP; BHISB + 0.1 M glucose; 24 hDecreased in biofilm production[42]
R20291- cdtA and cdtB, cwlD and cwlD *24-WPP; BHISB + 0.1 M glucose; 24, 72, 120 hUnchanged[42]
R20291ΔpilA1Glass jars with 8 mm glass beads and glass coverslips; BHISB; 24 hReduced thickness and biomass. Decreased live cell count and a decreased tendency to aggregate[44]
630Δerm::CD2214, ErmR TmS24-WMP; TYt, 48 hDenser with several short-rod bacteria and smaller micro-aggregates[46]
630Δerm::pilA1, ErmR TmS, 630Δerm::CD283124-WMP; TYt, 24 hUnchanged[46]
630Δerm ΔpilA1 (pRPF185 Ptet dccA), ErmR TmR96-well plate; TYt + anhydrotetracycline; 24 hForm a dense and homogeneous, carpet-like biofilm, slightly decreased biomass [46]
630ΔermΔCD2831 (pRPF185 Ptet dccA), ErmR TmR96-well plate; TYt + anhydrotetracycline; 24 hForm a dense and homogeneous, carpet-like biofilm without change in biofilm production [46]
630ΔermΔcwp84 and R20291Δcwp8424-WPP; BHISB + 1.8% glucose; 72 hIncreased 72-fold. Denser and thicker. Protein abundance in biofilm was altered [48]
630Δerm::2831, 630Δerm::0183, 630Δerm::3392, 630Δerm::CbpA24-WMP; BHISB; 24 hDecreased biofilm production[51]
630∆PEPP-124-WMP; BHISB, 24 hUnchanged[51]
630∆PEPP-1(Ptet2831), 630∆PEPP-1(Ptet3246)24-WMP; BHISB, 24 hIncreased biofilm production [51]
R20291::spo0A24-WMP or 24-WMP with coverslips; BHISB, 72 and 144 hDecrease in depth and breadth of the biofilm. Decrease the number of spores [51]
JIR8094:: lcpB24-WPP; BHISB + 1.8% glucose; 72 hRobust biofilm[52]
JIR8094:: lcpA24-WPP; BHISB + 1.8% glucose; 72 hUnchanged[52]
630Δerm (pRPF185 Ptet dccA), TmR96 well polystyrene plate; TYt, 48 hIncreased biofilm production. Biovolume increase of 1.6-fold. Highly homogeneous and dense[46,53]
R20291::lrp, 630Δerm::lrp24-WPP; BHISB + 0.1 M glucose; 72 hUnchanged[54]
R20291::lexA238a24-WPP; BHISB + 20 μg/mL lincomycin: 24 hIncreased biofilm production [55]
20291::luxS161a24-WPP; BHISB + 0.1 M glucose; 24 y 72 hDecreased biofilm production[56]
630Δerm::dnaK723a96-well flat-bottom polystyrene plate; BHISB + 0.9% glucose; 24, 48 and 72 hIncreased biofilm production[57]
630Δerm::CD1687, JIR8094::codY, JIR8094::ccpA, 630Δerm::spo0A24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hDecreased biofilm production[58]
630Δerm::CD1688, 630Δerm::sigB, 630Δerm::sigE, 630Δerm::sigF24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hUnchanged[58]
630Δerm::cwp1924-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hFailed to form a biofilm [58]
R20291ΔcmrR24-WPP; BHISB + 1% glucose + 50 mM sodium phosphate buffer; 24 h [59]
R20291ΔcmrT24-WPP; BHISB + 1% glucose + 50 mM sodium phosphate buffer; 24 hUnchanged[59]
630ΔermΔprkC24-WMP; BHISB + 0.1 M glucose + polymyxin B (20 μg·mL−1) or DOC (0.01%); 48 hProduce 6- and 10-fold more biofilm than the WT [60]
630∆ermpilW, 630∆ermΔpilA1, 630∆ermΔT4P2 cluster, 630∆ermΔsinR, 630∆ermΔCD630_08650, 630∆ermΔcwp29, 630∆ermΔcysk, 630∆ermΔagrBD, 630∆ermΔluxS, 630∆ermΔhprk, 630∆ermΔfumAB, 630∆ermΔnanEAT, 630∆ermΔprdB, 630∆ermΔfur, 630∆ermΔrex24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hUnchanged[61]
630∆ermΔbcsA, 630∆ermΔfliC24-WPTCTP; BHISB + 100 mM glucos e+ 240 µM DOC; 24 or 48 hUnchanged[61]
630∆ermΔsigD24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 24 hUnchanged[61]
630∆ermΔT4P1 cluster, 630∆ermΔcdsB 630∆ermΔspo0A, 630∆ermΔsigH, 630∆ermΔsigL,24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hDecreased biofilm production[61]
630∆ermΔptsI24-WPTCTP; BHISB + 100 mM glucose + 240 µM DOC; 48 hInduction of biofilm abolished by DOC [61]
DOC, deoxycholate; BHISB, brain–heart infusion supplemented with 5 g/L yeast extract and cysteine 0.1% broth; TYt broth, tryptone yeast extract broth supplemented with 0.1% sodium thioglycolate; 24-WPP, 24-well polystyrene plate; 24-WMP, 24-well microtiter plate; 24-WPTCTP, 24-well polystyrene tissue culture-treated plate; *, not specified.
Table 3. Effects of antibiotics on C. difficile biofilms, spores, and planktonic cells.
Table 3. Effects of antibiotics on C. difficile biofilms, spores, and planktonic cells.
Effect on BiofilmEffect on Spores/Planktonic CellsChanges in Gene Expression
Fidaxomicin
Penetrates biofilms within 2 min [120].
0.03x–0.25x MICs exhibit a dose-dependent inhibitory effect on biofilm formation [121].

0.09x and 0.25x MICs cause thickness and biomass reduction [121].
0.50x MIC decreases vegetative cell growth and biofilm formation [121].

25x MIC decreases the spore count and kills vegetative cells within mature biofilms [120].
Spores
0.25x and 0.125x MICs during the stationary phase prevent the production of spores.
2x MIC decreases the outgrowth of vegetative cells [122].

Planktonic cells
0.25x MIC reduces viability [123].
fliC expression increases, but no change occurs in the expression of pilA1, cwp84, luxS, dccA, and spo0A [121].
0.25x MIC decreases spo0A transcription [123].
No accumulation of spoIIR or spoIIID mRNA occurs [122].
In the non-biofilm state, it suppresses the expression of both tcdA and tcdB, with maximal repression at 1/4x MIC [123].
Vancomycin
0.25x MIC is associated with reduced biomass [121].

12.5 mg/mL reduces the viable vegetative cell count in intact biofilms with an enhanced effect by adding DNase [51].
It does not affect spore viability, irrespective of biofilm disruption [51].
Spores
0.5x MIC may affect the spore count [124].
0.25x and 0.125x MICs do not affect sporulation during the stationary phase [122].
0.25x and 0.125x MICs reduce spore production in 48 h cultures [123].
2.5x MIC inhibits the outgrowth of vegetative cells and does not affect spore germination [125].
Planktonic cells
It only inhibits the growth of vegetative cells [122].
0.25x MIC does not change the mARN expression of pilA1, cwp84, luxS, dccA, and spo0A [121].
In a biofilm state, 0.5x MIC increases the transcription of tcdA and tcdB toxins [124].
Metronidazole
0.25x and 0.5x MICs increase in vitro biofilm formation.
It stimulates the production of a thick biofilm composed of layered aggregates and influences extracellular matrix production a [121,126].
Spores
0.5x MIC does not affect sporulation [124].
0.25x MIC does not inhibit sporulation [122].
0.25x–0.125x and 0.0625x MICs stimulate sporulation in strain 5325 [123].
0.25x–0.125x and 0.0625x MICs suppress spore formation in strain 9689 [123].
SurotomycinPlanktonic cells
It penetrates C. difficile biofilms in less than one hour and starts accumulating.
It exhibits a disruptive activity on biofilm structure at 24 h [120].
100x, 50x, and 25x MICs kill vegetative C. difficile strain ATCC BAA-1382 within biofilms in vitro [120].
8x and 80x MICs kill vegetative exponential-phase cells.
80x MIC kills stationary-phase cells [127].
MIC, minimum inhibitory concentration; VAN, vancomycin; MTZ, metronidazole; FDX, fidaxomicin. a Contradictory evidence.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rubio-Mendoza, D.; Martínez-Meléndez, A.; Maldonado-Garza, H.J.; Córdova-Fletes, C.; Garza-González, E. Review of the Impact of Biofilm Formation on Recurrent Clostridioides difficile Infection. Microorganisms 2023, 11, 2525. https://doi.org/10.3390/microorganisms11102525

AMA Style

Rubio-Mendoza D, Martínez-Meléndez A, Maldonado-Garza HJ, Córdova-Fletes C, Garza-González E. Review of the Impact of Biofilm Formation on Recurrent Clostridioides difficile Infection. Microorganisms. 2023; 11(10):2525. https://doi.org/10.3390/microorganisms11102525

Chicago/Turabian Style

Rubio-Mendoza, Daira, Adrián Martínez-Meléndez, Héctor Jesús Maldonado-Garza, Carlos Córdova-Fletes, and Elvira Garza-González. 2023. "Review of the Impact of Biofilm Formation on Recurrent Clostridioides difficile Infection" Microorganisms 11, no. 10: 2525. https://doi.org/10.3390/microorganisms11102525

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