Next Article in Journal
Dermatologic Changes in Experimental Model of Long COVID
Previous Article in Journal
Sebum Components Dampen the Efficacy of Skin Disinfectants against Cutibacterium acnes Biofilms
Previous Article in Special Issue
Update on Commonly Used Molecular Typing Methods for Clostridioides difficile
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reviewing the Clostridioides difficile Mouse Model: Insights into Infection Mechanisms

by
José L. Fachi
1,*,
Marco A. R. Vinolo
2 and
Marco Colonna
1
1
Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
2
Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083-862, SP, Brazil
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(2), 273; https://doi.org/10.3390/microorganisms12020273
Submission received: 24 December 2023 / Revised: 16 January 2024 / Accepted: 25 January 2024 / Published: 27 January 2024
(This article belongs to the Special Issue Molecular Epidemiology of Clostridium difficile)

Abstract

:
Clostridioides difficile is an anaerobic, spore-forming bacterium associated with intestinal infection, manifesting a broad spectrum of gastrointestinal symptoms, ranging from mild diarrhea to severe colitis. A primary risk factor for the development of C. difficile infection (CDI) is antibiotic exposure. Elderly and immunocompromised individuals are particularly vulnerable to CDI. A pivotal aspect for comprehending the complexities of this infection relies on the utilization of experimental models that mimic human CDI transmission, pathogenesis, and progression. These models offer invaluable insights into host–pathogen interactions and disease dynamics, and serve as essential tools for testing potential therapeutic approaches. In this review, we examine the animal model for CDI and delineate the stages of infection, with a specific focus on mice. Our objective is to offer an updated description of experimental models employed in the study of CDI, emphasizing both their strengths and limitations.

1. Introduction

Clostridioides difficile, commonly referred to as C. difficile or C. diff, is a spore-forming anaerobic bacterium of significant notoriety within the domain of infectious diseases [1]. Over recent decades, it has emerged as a prominent causative agent of healthcare-associated infections, particularly affecting individuals who have recently undergone antibiotic therapy or present a compromised immune system [2]. The infection primarily relies on the perturbation of the gut microbiota [3], creating a conducive environment for the colonization of C. difficile and the subsequent release of its toxins [4]. These toxins adversely affect host cells, undermining the integrity of the intestinal barrier and causing inflammation along with tissue damage [5,6]. The clinical manifestation of the disease in humans encompasses a spectrum of presentations, ranging from mild diarrhea to severe colitis, often leading to life-threatening complications and, in some cases, mortality [7,8].
Substantial research efforts have been dedicated to unraveling the complexities of C. difficile infection (CDI), covering aspects such as prevention, pathogenesis, and treatment approaches. An essential component of this research journey involves the establishment and utilization of animal models to replicate and analyze various aspects of diseases. For decades, hamsters and mice have been the primary animal models for studying CDI [9]. These models have been instrumental in elucidating key aspects of the disease, such as C. difficile toxins production, host immune response, and the role of microbiota in the host susceptibility to CDI. However, these models inherently exhibit limitations that should be taken into consideration for experimental applications.
In this review, we offer updated information on animal models in C. difficile infection research, with a particular focus on murine models. We aim to emphasize the valuable insights these models have brought to our comprehension of CDI and highlight their effective performance in elucidating the disease’s complexity.

2. Clostridioides difficile Infection

Bacillus difficile”, as denominated before, entered the scientific spotlight in 1935 when it was first identified by researchers Hall and O’Toole [10]. At that time, this bacterium was considered relatively innocuous, quietly inhabiting the human gut without drawing much attention. However, it was during the 1970s that “Clostridium difficile” began to reveal its pathogenic potential [11,12,13]. Researchers observed a connection between C. difficile and antibiotic-associated diarrhea, as well as pseudomembranous colitis, leading to a surge in research interest [8]. Since then, the bacterium has persisted in its evolutionary trajectory, developing resistance to various antibiotics and producing virulent strains that pose significant challenges to healthcare providers and researchers alike [14]. In 2016, the reclassification of “Clostridium difficile” to “Clostridioides difficile” marked a significant taxonomic shift, motivated by advancements in genetic research and DNA sequencing [15]. This realignment sought to more accurately reflect the bacterium’s genetic and evolutionary characteristics, setting it apart from the Clostridium genus.
C. difficile is a Gram-positive bacterium characterized by its ability to form spores [16]. These spores are the resilient, dormant form of the bacterium, exhibiting the capacity to endure adverse conditions, including high temperatures and sterilizing chemicals [17]. Their durability contributes to their persistence in the environment, making CDI prevention and control challenging. Moreover, C. difficile spores play a critical role in CDI recurrence; even after successful CDI treatment, some patients experience a recurrence due to the germination of these spores when favorable conditions arise in the colon, often as a result of disruptions to the normal gut microbiota [16,18,19]. Certainly, their germination and the onset of C. difficile infection depend not only on microbiota imbalance, but also on various host-related factors, including immunity, age, and others. In fact, individuals within healthcare settings face an increased risk for CDI, attributed to factors like the use of antibiotics, an impaired immune system, and direct exposure to an environment with a higher abundance of C. difficile spores [4]. Moreover, 2.4% to 17.5% of healthy adults can asymptomatically carry C. difficile, serving as potential sources of transmission, both in healthcare environments and the community at large [20,21,22].
The pathological consequences of CDI are primarily centered in the colon. The disease can manifest across a spectrum of severity, ranging from mild diarrhea to severe pseudomembranous colitis, and in extreme cases, leading to fulminant outcomes [23,24,25]. Specifically, the development of pseudomembranous colitis arises as a sequence of unfolding events leading to colitis: CDI often starts with the ingestion of C. difficile spores, typically encountered in the environment, which then transform into active bacteria after germination within the large intestine (cecum and colon) [16]. C. difficile produces two crucial toxins, toxin A (TcdA) and toxin B (TcdB), that specifically target epithelial cells by binding to their respective receptors [5,6]. Once internalized, these toxins cause the subsequent inactivation of Rho-family GTPases through glucosylation [5,26,27]. This event disrupts the host cell’s cytoskeleton, accelerating cell death [28,29]. Consequently, the damaged epithelial layer increases gut permeability and facilitates the translocation of luminal content into the intestinal lamina propria. Simultaneously, the accumulation of dead cells and the presence of translocated bacteria enable immune cells and fluids to infiltrate the tissue [30,31]. As a result, pseudomembranes manifest as yellowish-white patches on the inflamed colon mucosa, consisting of inflammatory cells, mucus, fibrin, and dead epithelial cells [30]. The classic symptoms of pseudomembranous colitis include severe diarrhea, abdominal pain, and fever. In the absence of treatment or in cases of heightened host susceptibility to infection, severe complications may arise, including toxic megacolon, bowel perforation, sepsis, and, in some instances, death [32].
In addition, the emergence of hypervirulent strains, such as the NAP1/B1/027 variant, has emerged as a global public health concern by being associated with more severe and recurrent cases of CDI [33,34,35,36,37]. Specifically, the PCR ribotype 027 strain, commonly denoted as RT027, distinguishes itself with heightened virulence, characterized by elevated production of TcdA and TcdB toxins [38]. Infections with the RT027 strain are known for their high recurrence rates, impacting patient outcomes and placing a significant burden on healthcare systems. Remarkably, RT027 demonstrates resistance to certain antibiotics, notably fluoroquinolones, presenting challenges in treatment and contributing to persistent infections [39,40]. Furthermore, this strain is recognized for frequently producing the binary toxin, also known as CDT (C. difficile transferase), which is associated with increased severity and mortality in patients [41,42,43]. Unlike the more well-known toxins TcdA and TcdB, CDT functions as an ADP-ribosyltransferase, comprising two subunits, CDTa and CDTb. CDTa manages enzymatic activity, while CDTb facilitates entry into host cells [42]. Research indicates that this binary toxin plays a crucial role in enhancing the virulence of C. difficile, particularly through increased activation of the inflammasome via Toll-like receptor 2 (TLR2)-mediated priming [44]. This heightened inflammasome activation is implicated in the pathogenicity of C. difficile infection. Additionally, CDT-induced TLR2 activation leads to the release of proinflammatory cytokines and increased apoptosis of eosinophils, further contributing to the overall impact of CDT on the host immune response [45]. Comprehending the role of the binary toxin is crucial for unraveling host immune responses and developing novel therapeutic strategies for CDI.
The current strategy for managing and treating patients with CDI typically involves a multifaceted approach [46]. Treatment plans are customized to address the unique clinical condition of each individual, underscoring the crucial role of early diagnosis and intervention for effectively managing the disease. The first step is discontinuing the antibiotic that may have triggered the infection, if applicable, to halt the selective advantage C. difficile gains in an altered gut microbiome. The next crucial step is the administration of specific antibiotics, such as vancomycin or fidaxomicin, to target and eliminate C. difficile bacteria [46,47,48]. Metronidazole is designated as an alternative agent and has been omitted from the treatment of nonsevere CDI since the 2021 guidelines [49]. Additionally, supportive care, including adequate hydration and electrolyte management, plays a pivotal role in treatment, particularly for patients with severe diarrhea [46]. These strategies do not contribute to the restoration of the natural microbiota’s colonization, potentially explaining their modest effectiveness in terms of treatment outcomes and increased rates of relapse episodes [3]. Approximately 15–30% of patients who respond to antimicrobial therapy may experience recurrent CDI, and the risk escalates with each subsequent episode [50]. Studies indicate that older age and female sex are associated with an increased risk of recurrence [51]. It is also noteworthy to notice that recurrent CDI is not always attributed to the same strain, with a new strain being identified in 33–56% of recurrent episodes [52,53,54]. For the treatment of recurrent CDI, Bezlotoxumab (Zinplava), a monoclonal antibody targeting the toxin TcdB, helps prevent recurrences and mitigates damage to colonic epithelial cells [55].
Remarkably, fecal microbiota transplantation (FMT) surprisingly emerges as a highly potent therapeutic approach for treating recurrent or severe CDI [56,57]. This procedure entails the infusion of fecal matter from a healthy donor into the patient’s gastrointestinal tract, with the overarching goal of reestablishing a balanced gut microbiota. The transplanted microbiome, armed with its diverse arsenal of beneficial microbes, plays a crucial role in efficiently combatting the opportunistic C. difficile [3,58]. Meta-analyses examining the efficacy of FMT for recurrent, severe, or fulminant CDI consistently indicate promising outcomes [59,60]. FMT has demonstrated high success rates in preventing recurrent CDI, with significant reductions in recurrence compared to standard antibiotic therapy. Moreover, this therapeutic approach has shown effectiveness in treating severe and fulminant cases, contributing to improved clinical outcomes and reduced mortality rates. However, it is not without its drawbacks and potential risks [61]. One of the primary concerns is the lack of long-term safety data, as the consequences of introducing a new, diverse microbial community into a patient’s gut over time remain uncertain. FMT relies on donated fecal matter, which, despite rigorous donor screening and testing, may still carry undetected pathogens or infections that can be transmitted to the recipient [62,63]. There is also a risk of unintended alterations to the recipient’s microbiome, potentially leading to unexpected health issues [64,65]. Continuing research and ongoing debates focus on the application and long-term consequences of FMT, emphasizing the need for a thorough evaluation of its risks and benefits.
Overall, effectively combating CDI requires a comprehensive understanding of its microbiological foundations, insight into the underlying pathological processes, and the ability to navigate the complex epidemiological factors that contribute to its persistence and spread. Given these considerations, animal models of C. difficile infection play a pivotal role in providing insights into the pathogenesis, treatment, and prevention of CDI.

3. Animal Models of CDI

CDI has been studied using different animal species, including hamsters, guinea pigs, rabbits, mice, and rats, as reviewed before [9]. The hamster model is the most commonly used experimental model. On hamsters, CDI can be induced after ingestion of antibiotics, and colonization occurs through experimental challenge or environmental exposure to C. difficile [66,67,68]. The disease in this model primarily affects the cecum with some involvement of the ileum, resulting in diarrhea and fatal enterocolitis when exposed to toxigenic strains [69,70]. It is important to note that this model represents well the severe and lethal forms of the disease, but does not consistently display the full spectrum of CDI symptoms seen in humans [66,67,71]. The hamster model has been employed for three decades to investigate CDI therapy and disease mechanisms. Studies involving hamster vaccination have demonstrated that C. difficile toxoids (formalin-inactivated toxins) can offer protection against fatal outcomes in animal models [72].
Mice and rats are less susceptible to CDI than hamsters [73]. Nevertheless, murine models serve as valuable tools for studying CDI, given their genetic similarity to humans and enhanced translational relevance [74]. Mouse application allows researchers to control various factors such as genetics, environment, and diet, ensuring consistent experimental conditions. The availability of transgenic mice facilitates the examination of host factors and components of immune responses throughout the progression of the disease. There are well-established protocols for inducing C. difficile infection in mice, making them a standardized and widely adopted model in the field. Typically, laboratory mice are subjected to antibiotic treatment to disturb their gut microbiota, rendering them susceptible to the germination and colonization of C. difficile—mirroring the dynamics observed in humans [75,76]. Then, the mice are orally inoculated with C. difficile spores or vegetative cells. Over time, infected mice may develop symptoms similar to those seen in humans, such as diarrhea, weight loss, and other gastrointestinal issues [54,77]. The severity of symptoms varies depending on the C. difficile strain used and the susceptibility state of the mouse [78]. Researchers can monitor various parameters, including the progression of infection, changes in gut microbiota composition, histopathological alterations in the colon, and levels of C. difficile toxins in feces [79]. The mouse model is very useful for testing potential treatments and interventions, including new antibiotics, probiotics, dietary modifications, or vaccines designed to combat C. difficile infection.
Numerous mouse models have been created for studying the mechanisms behind C. difficile pathogenesis, and each has its own strengths for investigating different aspects of the disease [75]. The selection of a particular method is guided by the research goals, the specific strain of C. difficile, and the intended disease features. One common approach is to perform a single oral gavage with C. difficile spores, as it mimics the natural route of infection from contaminated surfaces [80,81]. To study recurrent infections, some models involve multiple rounds of challenge [50,54]. Alternatively, researchers can administer purified C. difficile toxins to study the role of toxins in disease pathogenesis [82,83,84,85]. Antibiotic pretreatment disrupts the gut microbiota, making mice more susceptible to colonization, reflecting conditions seen in patients with antibiotic-associated C. difficile disease [86,87,88]. Likewise, models designed to investigate colonization resistance explore the use of probiotics or microbiota components to enhance the mechanisms of resistance [88,89,90,91].
Gnotobiotic/germ-free mice can be colonized by C. difficile and exhibit intestinal pathology, primarily in the colon, with pseudomembrane formation similar to human disease [92,93,94,95]. This model in particular enables the exploration of microbiota or isolated groups of bacteria’s role in the development of CDI. Reeves et al. unveiled a significant delay in the onset of primary CDI and relapse in germ-free mice pretreated with probiotics [96]. Notably, the microbiome of mice undergoing moderate CDI and receiving probiotic treatment revealed a striking increase in the abundance of the Lachnospiraceae family during the initial CDI phase. Intriguingly, mice that were precolonized with the Lachnospiraceae isolate demonstrated a substantial decrease in C. difficile colonization, decreased levels of intestinal cytotoxin, and exhibited milder clinical symptoms and colonic histopathology. This effect was not observed in mice solely colonized with E. coli as a control. This study suggests novel therapeutic approaches for the treatment and prevention of CDI by utilizing bacterial species that potentially inhibit C. difficile growth. Likewise, a bacterial consortium named VE303 is currently under development as a potential treatment for high-risk CDI [97,98]. This oral treatment consists of eight nonpathogenic, nontoxigenic, commensal strains of Clostridia that have been shown to effectively restore a healthy gut microbial community, mitigate inflammation, and elevate levels of protective metabolites in mice [97,98]. However, the use of gnotobiotic mouse models has a much higher cost and is less practical when compared to conventional mice. Additionally, the absence of other microorganisms in these models makes them less representative of the human situation. Nonetheless, the utilization of germ-free mice is crucial for investigating the specific role of human microbiota in the development of CDI. Humanized mouse models, engrafted with human microbiota, provide an environment closer to human C. difficile infection [99,100,101,102]. These methods offer flexibility in investigating various aspects of C. difficile infection, from pathogenesis to potential interventions.

4. Antibiotic-Induced Murine Model of CDI

Antibiotic-induced murine models of CDI are specifically designed to mimic the clinical manifestations and pathological features of human CDI within a controlled laboratory setting [80,103]. Despite some limitations, this model has greatly contributed to our understanding of CDI and continues to be an essential component of CDI research. Antibiotics are employed to initiate gut dysbiosis, a crucial step in simulating the clinical conditions that lead to C. difficile expansion [87,104,105]. Commonly used antibiotics for this purpose include vancomycin, cefoperazone, and tetracycline, typically administered orally via drinking water supplementation [73,106,107,108,109]. Likewise, clindamycin is often linked to the development of CDI in humans, attributed to its effectiveness against a wide range of bacteria. It is administered systemically as an antibiotic, usually through intraperitoneal injection in mice. In essence, antibiotic treatment eliminates competing bacteria in the gastrointestinal tract, thereby creating an environment that favors the proliferation of C. difficile [110,111].
The selection of antibiotics varies according to the specific study goals and the desired level of gut dysbiosis, with the administration of these antibiotics for a predetermined period before introducing C. difficile to initiate infection. Giel et al. conducted a study on the antibiotic-induced mouse model of CDI to investigate C. difficile spore germination [18]. Their findings revealed that spores were more likely to germinate in antibiotic-treated mice, but this germination was mitigated by cholestyramine, a treatment that chelated bile salts. This observation led the authors to suggest that cecal bacterial populations in antibiotic-treated mice had a reduced capacity to modify taurocholate, a spore germinant factor, providing further support for the role of bile salts in C. difficile spore germination and infection development in the host [18]. Similarly, a ten-day antibiotic pretreatment with broad-spectrum cefoperazone in drinking water showed an increase in mice’s susceptibility to infection with C. difficile strain VPI 10463 [105]. This particular strain of C. difficile produces elevated levels of toxins, and experimental infection with it proves lethal in hamsters and, depending on the dose, in mice [80,112].
Studies on mouse models have extensively characterized infection with C. difficile spores. Lawley et al. [113] confirmed that a low number of spores (seven) of C. difficile per square centimeter of mucosal tissue constitutes the minimum infectious dose. In another study by the same authors, Lawley et al. explored spore transmission between healthy and immunocompromised mice, shaping infection control strategies [73]. This study demonstrated that virulent C. difficile can asymptomatically colonize the intestines of healthy mice, establishing a persistent carrier state. The authors convincingly illustrated that when C. difficile was shed in significant amounts, referred to as the “super-shedder” state, it promoted efficient transmission. In contrast, the carrier state, in which C. difficile shedding was lower, did not facilitate transmission to the same extent. In healthy mice, the infection induced mild mucosal inflammation. Conversely, transmission to mice with compromised innate immune responses (Myd88−/−) resulted in severe and frequently fatal intestinal disease, underscoring the role of host factors in CDI.
In some research scenarios, a combination of antibiotics may be utilized to induce a more profound disturbance in the gut microbiota, closely mirroring the clinical situation where patients often receive multiple antibiotics before developing CDI. Chen et al. [80] pioneered an innovative mouse model wherein C57Bl/6 mice were subjected to a 3-day oral exposure to a combination of kanamycin, gentamicin, colistin, metronidazole, and vancomycin, followed by an intraperitoneal administration of clindamycin 48 h later. Subsequently, the mice were challenged with varying doses of C. difficile. This mouse model faithfully replicated human C. difficile infections, inducing characteristic symptoms such as diarrhea, weight loss, and typical histological features. The severity of the disease in this mouse model was proportional to the challenge dose used, ranging from 2 × 102 to 105 CFU. In a parallel initiative, our research team modified the infection model, originally proposed by Chen et al., to incorporate a more aggressive dose of the vegetative form of C. difficile [79]. We found that treating 8-week-old male C57BL/6 mice with a combination of kanamycin, gentamicin, colistin, metronidazole, and vancomycin for 4 days, followed by clindamycin administration 24 h later, and subsequent infection in the next day with 108 CFU of C. difficile, effectively induced weight loss, diarrhea, and intestinal inflammation.
Furthermore, researchers investigated the progression of C. difficile infection in correlation with the age of the mice [92]. The study utilized germ-free C57BL/6 mice aged 7 to 14 months, infected with the BI/NAP1 strain. Notably, older infected mice were critically ill within 48 to 72 h after infection, exhibiting noticeable cecitis and colitis upon gross and histological examination. These infected mice also displayed elevated levels of keratinocyte chemoattractant (KC, also known as Cxcl1), interleukin 1β, monocyte chemotactic protein 1 (MCP-1, also known as Ccl2), and granulocyte colony-stimulating factor (G-CSF). Conversely, they had decreased levels of interferon-γ, interleukin-12, and interleukin-10 when compared to the younger control group. This unique model not only sheds light on the role of the host response in disease development but also highlights its connection with the aging process. These findings are particularly significant due to the rising incidence and severity of C. difficile infection in older adults, particularly those aged 65 years and above, largely associated with the emergence of the BI/NAP1 strain [114,115].

5. CDI Pathogenesis and Disease Progression in Mice

After antibiotic treatment, the mouse’s gut undergoes substantial modifications, rendering it susceptible to C. difficile colonization [87,116,117]. The highly resilient C. difficile spores colonize the intestines, primarily finding their niche in the murine cecum and colon [16,118]. Within these regions, spores undergo germination, transitioning into vegetative cells that actively grow and produce the toxins responsible for clinical symptoms (Figure 1) [118]. Toxin A (TcdA) and toxin B (TcdB) are released following C. difficile germination, typically occurring on the first day postinfection [118]. These toxins have a specific affinity for the intestinal epithelial cells, disrupting the colonic epithelium and subsequently contributing to tissue damage and bacterial translocation [119,120,121,122]. In particular, gut bacterial translocation involves the process through which bacteria, mostly those that naturally inhabit the intestinal tract, cross the protective barrier of the gut and enter the bloodstream or other distant anatomical regions. Furthermore, the compromised epithelial barrier plays a pivotal role in the development of diarrhea and inflammation during the early stages (around day 2 postinfection) [123]. This event can ultimately lead to the development of sepsis, a life-threatening condition that arises in an overwhelming and systemic inflammatory response [124]. The consequent uncontrolled immune reaction, often termed a “cytokine storm”, holds the potential to inflict substantial harm systemically, characterized by a spectrum of clinical symptoms, including a high fever, cognitive impairment, organ dysfunction, and a reduction in blood pressure [125,126]. In severe cases, mice may manifest fulminant colitis, elevating the risk of mortality significantly, while sepsis can lead to a fatal outcome [124].
In addition, the intestinal inflammatory cascade after TcdA and TcdB toxins-associated damage, which includes the recruitment of immune cells, such as neutrophils, and the release of proinflammatory cytokines, such as IL-1β, IL-23, TNF-α, and IFN-γ, occurs around 2 days postinfection, a hallmark of CDI disease progression [121,122,123,127,128,129,130]. In particular, these toxins have been noted for their ability to activate the inflammasome, a multiprotein complex that detects cellular stress and danger signals, instigating a potent inflammatory response [131]. This activation leads to the cleavage of proinflammatory cytokines, including interleukin-18 (IL-18) and IL-1β, crucial in combating the infection [132]. Excessive activation, however, can be pathogenic, contributing to heightened host morbidity and mortality [44,131]. Moreover, the existence of the binary toxin, CDT, has demonstrated an augmentation in inflammasome activation via TLR2-mediated priming [45,133]. The process involves the coordinated actions of Mitogen-Activated Protein Kinases (MAPKs), which in turn activate the Nuclear Factor κB (NF-κB) [133]. MAPKs are triggered, initiating a phosphorylation cascade that leads to the degradation of IκB, enabling the release and translocation of NF-κB into the nucleus for the transcriptional regulation of inflammatory genes [134]. This collaborative interplay among the inflammasome, NF-κB, and MAPKs forms a dynamic and orchestrated response, pivotal for the host’s early defense against C. difficile infection.
In severe cases, pseudomembrane plaques are visible on the colonic mucosa around day 3 and 4 p.i., consisting of inflammatory cells, fibrin, and mucus [135]. However, it is essential to recognize that the mouse model of CDI is typically acute, encompassing a brief one-week timeframe [54,80]. After the disease reaches its peak around day 2 and 3 p.i., a resolutive phase takes hold within the intestinal lamina propria around 5 days postinfection [79]. During this phase, we observe a diminishing inflammatory response, with increased epithelial cell proliferation and mucus production, and the gradual relief of clinical symptoms, including body weight loss and diarrhea [79,136,137]. Significant changes include a decline in neutrophil infiltration in the tissue, a decrease in proinflammatory cytokine levels, and an increase in IL-10 and IL-22 production [138,139,140,141]. Moreover, there is a notable increase in the prevalence of group 3 innate lymphoid cells (ILC3) and regulatory T cells (Treg cells) within the lamina propria, with these alterations becoming particularly pronounced after 5–6 days of infection [128,142]. IL-22-productin ILC3 play a crucial role in CDI, reinforcing the gut barrier and promoting tissue repair [121,128]. Treg cells have a dual role, modulating immune responses to prevent excessive inflammation, while potentially hampering the efficient clearance of C. difficile [142,143].
In some instances, the immune response in mice is effective in clearing the infection, and they recover from CDI. However, chronic colonization can occur in some cases (around 20% of cases), with C. difficile persisting in the gut, potentially leading to recurrent episodes of infection [144,145]. Furthermore, following a recovery period and the discontinuation of antibiotics, mirroring the clinical scenario in which patients are susceptible to recurrent CDI, mice can be subjected to a rechallenge with C. difficile, whether from the same or a different strain [54]. The rechallenge initiates a recurrent phase in mice that were previously infected, enabling the evaluation of the host’s immune response, the maturation of protective immunity, and the effective assessment of various interventions for preventing relapse. Although T cells and B cells may not play a direct role in resolving the acute phase of C. difficile infection in mice [128,141], clinical data suggest that adaptive immune responses can confer protective effects. In this context, B cells are instrumental in the production of specific antibodies, including IgG and IgA, which can neutralize C. difficile toxins and limit their damage to intestinal epithelial cells [21,146,147,148]. Furthermore, CD4+ and CD8+ T cells play pivotal roles, orchestrating the immune response by aiding in antibody production, stimulating other immune cells, and directly targeting infected host cells [149,150,151]. Importantly, the development of immunological memory within the adaptive immune system ensures a more rapid and effective response upon re-exposure to C. difficile [152,153].
In the murine model, CDI manifests as clinical symptoms similar to those seen in humans. The severity of symptoms can vary depending on factors such as the C. difficile strain used, the antibiotic regimen, and the specific study conditions. In general, symptoms include severe diarrhea, often watery in consistency, which reflects the damaging effects of C. difficile toxins on intestinal epithelial cells [4]. In mice, CDI leads to significant weight loss, typically ranging from 10% to 20% of their initial body weight [80], serving as a quantifiable marker of infection severity and its overall impact on host responses. Reduced activity, lethargy, and altered grooming behavior are also common behavioral manifestations after infection [107]. Additional clinical signs, such as a hunched posture and ruffled fur, further highlight the mice’s discomfort [77]. These signs and symptoms are commonly quantified using a standardized clinical scoring system, frequently employed in research and experimentation (Table 1) [77].
Furthermore, colonic inflammation characterized by neutrophil infiltration and tissue damage, and pseudomembranous colitis featuring visible plaque on the colonic mucosa, serve as critical histopathological features [54,123]. Additional parameters, including a decrease in goblet cell frequency and mucus reduction, also serve as indicators of disease progression and severity [154,155]. The assessment of epithelial permeability, exemplified by tests involving the ingestion of sugar molecules followed by the analysis of urine or blood samples, as well as the quantification of translocated bacteria in various peripheral organs (e.g., liver, spleen, kidney, etc.), can serve as valuable indicators of compromised epithelial barrier function following infection [120]. Finally, examination of the immune response within the intestinal tissue postinfection, conducted through techniques like flow cytometry, immunofluorescence on preserved tissue sections, or the quantification of cytokines and gene expression, serves as a valuable means to monitor the progression of CDI in murine models [6].

6. Conclusions

Over time, C. difficile is increasingly developing resistance to antibiotics and generating virulent strains, posing challenges for healthcare providers and researchers alike. Current research on CDI covers a wide range of crucial areas, including the exploration of the gut microbiome’s role, investigating alternatives to antibiotics such as fecal microbiota transplantation (FMT), and finding strategies for the prevention and management of CDI. Mouse models have played a crucial role in the understanding of this field, offering insights into pathogenesis, host responses, and potential interventions. Researchers have employed various mouse models in the field, including genetically modified and germ-free mice. Specifically, the antibiotic-induced mouse model stands as a well-established tool for studying CDI, aiding in the comprehension of host–pathogen interactions. While it does have many benefits, the model may not fully replicate the complexities of human CDI, as it possesses gut microbiota that differ from that of humans. Overall, various protocols for utilizing mice in experimental CDI have been crucial for advancing our understanding of the disease and facilitating the development of approaches that may potentially be applied for managing the disease.

Author Contributions

J.L.F. wrote the manuscript. M.A.R.V. and M.C. have meticulously reviewed and suggested modifications to the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Pew Charitable Thrust, USA (#00035299), National Institutes of Health, USA (1R01DK126969-01), Fundação de Amparo à Pesquisa do Estado de São Paulo, Brazil (FAPESP, #2017/06577-9, 2018/15313-8), the National Council for Scientific and Technological Development, Brazil (CNPq), and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, Brazil (CAPES).

Data Availability Statement

This review article does not involve the generation of new data, as it relies solely on the analysis and synthesis of information from existing literature. All referenced sources are appropriately cited within the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Balsells, E.; Shi, T.; Leese, C.; Lyell, I.; Burrows, J.; Wiuff, C.; Campbell, H.; Kyaw, M.H.; Nair, H. Global Burden of Clostridium Difficile Infections: A Systematic Review and Meta-Analysis. J. Glob. Health 2019, 9, 010407. [Google Scholar] [CrossRef] [PubMed]
  2. Bartlett, J.G.; Gerding, D.N. Clinical Recognition and Diagnosis of Clostridium Difficile Infection. Clin. Infect. Dis. 2008, 46, S12–S18. [Google Scholar] [CrossRef] [PubMed]
  3. van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.W.M.; Tijssen, J.G.P.; et al. Duodenal Infusion of Donor Feces for Recurrent Clostridium Difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef] [PubMed]
  4. Czepiel, J.; Dróżdż, M.; Pituch, H.; Kuijper, E.J.; Perucki, W.; Mielimonka, A.; Goldman, S.; Wultańska, D.; Garlicki, A.; Biesiada, G. Clostridium Difficile Infection: Review. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1211–1221. [Google Scholar] [CrossRef] [PubMed]
  5. 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]
  6. Nibbering, B.; Gerding, D.N.; Kuijper, E.J.; Zwittink, R.D.; Smits, W.K. Host Immune Responses to Clostridioides Difficile: Toxins and Beyond. Front. Microbiol. 2021, 12, 804949. [Google Scholar] [CrossRef]
  7. Tedesco, F.J. Clindamycin-Associated Colitis. Ann. Intern. Med. 1974, 81, 429. [Google Scholar] [CrossRef]
  8. Bartlett, J.G.; Moon, N.; Chang, T.W.; Taylor, N.; Onderdonk, A.B. Role of Clostridium Difficile in Antibiotic-Associated Pseudomembranous Colitis. Gastroenterology 1978, 75, 778–782. [Google Scholar] [CrossRef]
  9. 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]
  10. Hall, I.C. INTESTINAL FLORA IN NEW-BORN INFANTS. Am. J. Dis. Child. 1935, 49, 390. [Google Scholar] [CrossRef]
  11. George, W.L.; Sutter, V.L.; Goldstein, E.J.; Ludwig, S.L.; Finegold, S.M. Aetiology of Antimicrobial-Agent-Associated Colitis. Lancet 1978, 1, 802–803. [Google Scholar] [CrossRef] [PubMed]
  12. Larson, H.E.; Price, A.B.; Honour, P.; Borriello, S.P. Clostridium Difficile and the Aetiology of Pseudomembranous Colitis. Lancet 1978, 1, 1063–1066. [Google Scholar] [CrossRef] [PubMed]
  13. Bartlett, J.G. Clostridium Difficile: History of Its Role as an Enteric Pathogen and the Current State of Knowledge about the Organism. Clin. Infect. Dis. 1994, 18 (Suppl. S4), S265–S272. [Google Scholar] [CrossRef] [PubMed]
  14. Markovska, R.; Dimitrov, G.; Gergova, R.; Boyanova, L. Clostridioides Difficile, a New “Superbug”. Microorganisms 2023, 11, 845. [Google Scholar] [CrossRef] [PubMed]
  15. Lawson, P.A.; Citron, D.M.; Tyrrell, K.L.; Finegold, S.M. Reclassification of Clostridium Difficile as Clostridioides Difficile (Hall and O’Toole 1935) Prévot 1938. Anaerobe 2016, 40, 95–99. [Google Scholar] [CrossRef]
  16. Shen, A. Clostridioides Difficile Spore Formation and Germination: New Insights and Opportunities for Intervention. Annu. Rev. Microbiol. 2020, 74, 545–566. [Google Scholar] [CrossRef]
  17. 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]
  18. Giel, J.L.; Sorg, J.A.; Sonenshein, A.L.; Zhu, J. Metabolism of Bile Salts in Mice Influences Spore Germination in Clostridium Difficile. PLoS ONE 2010, 5, e8740. [Google Scholar] [CrossRef]
  19. Sorg, J.A.; Sonenshein, A.L. Bile Salts and Glycine as Cogerminants for Clostridium Difficile Spores. J. Bacteriol. 2008, 190, 2505–2512. [Google Scholar] [CrossRef] [PubMed]
  20. Curry, S.R.; Muto, C.A.; Schlackman, J.L.; Pasculle, A.W.; Shutt, K.A.; Marsh, J.W.; Harrison, L.H. Use of Multilocus Variable Number of Tandem Repeats Analysis Genotyping to Determine the Role of Asymptomatic Carriers in Clostridium Difficile Transmission. Clin. Infect. Dis. 2013, 57, 1094–1102. [Google Scholar] [CrossRef] [PubMed]
  21. 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]
  22. 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]
  23. Cleary, R.K. Clostridium Difficile-Associated Diarrhea and Colitis: Clinical Manifestations, Diagnosis, and Treatment. Dis. Colon. Rectum 1998, 41, 1435–1449. [Google Scholar] [CrossRef]
  24. Kelly, C.P.; Pothoulakis, C.; LaMont, J.T. Clostridium Difficile Colitis. N. Engl. J. Med. 1994, 330, 257–262. [Google Scholar] [CrossRef] [PubMed]
  25. Mylonakis, E.; Ryan, E.T.; Calderwood, S.B. Clostridium Difficile--Associated Diarrhea: A Review. Arch. Intern. Med. 2001, 161, 525–533. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, Z.; Zhang, Y.; Huang, T.; Feng, H. Glucosyltransferase Activity of Clostridium Difficile Toxin B Is Essential for Disease Pathogenesis. Gut Microbes 2015, 6, 221–224. [Google Scholar] [CrossRef] [PubMed]
  27. Bilverstone, T.W.; Garland, M.; Cave, R.J.; Kelly, M.L.; Tholen, M.; Bouley, D.M.; Kaye, P.; Minton, N.P.; Bogyo, M.; Kuehne, S.A.; et al. The Glucosyltransferase Activity of C. Difficile Toxin B Is Required for Disease Pathogenesis. PLoS Pathog. 2020, 16, e1008852. [Google Scholar] [CrossRef] [PubMed]
  28. Chumbler, N.M.; Farrow, M.A.; Lapierre, L.A.; Franklin, J.L.; Haslam, D.B.; Goldenring, J.R.; Lacy, D.B. Clostridium Difficile Toxin B Causes Epithelial Cell Necrosis through an Autoprocessing-Independent Mechanism. PLoS Pathog. 2012, 8, e1003072. [Google Scholar] [CrossRef]
  29. Kuehne, S.A.; Cartman, S.T.; Heap, J.T.; Kelly, M.L.; Cockayne, A.; Minton, N.P. The Role of Toxin A and Toxin B in Clostridium Difficile Infection. Nature 2010, 467, 711–713. [Google Scholar] [CrossRef]
  30. Saavedra, P.H.V.; Huang, L.; Ghazavi, F.; Kourula, S.; Vanden Berghe, T.; Takahashi, N.; Vandenabeele, P.; Lamkanfi, M. Apoptosis of Intestinal Epithelial Cells Restricts Clostridium Difficile Infection in a Model of Pseudomembranous Colitis. Nat. Commun. 2018, 9, 4846. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Li, S.; Yang, Z.; Shi, L.; Yu, H.; Salerno-Goncalves, R.; Saint Fleur, A.; Feng, H. Cysteine Protease-Mediated Autocleavage of Clostridium Difficile Toxins Regulates Their Proinflammatory Activity. Cell Mol. Gastroenterol. Hepatol. 2018, 5, 611–625. [Google Scholar] [CrossRef] [PubMed]
  32. Farooq, P.D.; Urrunaga, N.H.; Tang, D.M.; von Rosenvinge, E.C. Pseudomembranous Colitis. Dis. Mon. 2015, 61, 181–206. [Google Scholar] [CrossRef] [PubMed]
  33. Lim, S.K.; Stuart, R.L.; Mackin, K.E.; Carter, G.P.; Kotsanas, D.; Francis, M.J.; Easton, M.; Dimovski, K.; Elliott, B.; Riley, T.V.; et al. Emergence of a Ribotype 244 Strain of Clostridium Difficile Associated with Severe Disease and Related to the Epidemic Ribotype 027 Strain. Clin. Infect. Dis. 2014, 58, 1723–1730. [Google Scholar] [CrossRef] [PubMed]
  34. Shin, B.-M.; Kuak, E.Y.; Yoo, S.J.; Shin, W.C.; Yoo, H.M. Emerging Toxin A-B+ Variant Strain of Clostridium Difficile Responsible for Pseudomembranous Colitis at a Tertiary Care Hospital in Korea. Diagn. Microbiol. Infect. Dis. 2008, 60, 333–337. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, J.; Kim, Y.; Pai, H. Clinical Characteristics and Treatment Outcomes of Clostridium Difficile Infections by PCR Ribotype 017 and 018 Strains. PLoS ONE 2016, 11, e0168849. [Google Scholar] [CrossRef]
  36. López-Ureña, D.; Orozco-Aguilar, J.; Chaves-Madrigal, Y.; Ramírez-Mata, A.; Villalobos-Jimenez, A.; Ost, S.; Quesada-Gómez, C.; Rodríguez, C.; Papatheodorou, P.; Chaves-Olarte, E. Toxin B Variants from Clostridium Difficile Strains VPI 10463 and NAP1/027 Share Similar Substrate Profile and Cellular Intoxication Kinetics but Use Different Host Cell Entry Factors. Toxins 2019, 11, 348. [Google Scholar] [CrossRef]
  37. Geric, B.; Carman, R.J.; Rupnik, M.; Genheimer, C.W.; Sambol, S.P.; Lyerly, D.M.; Gerding, D.N.; Johnson, S. Binary Toxin-Producing, Large Clostridial Toxin-Negative Clostridium Difficile Strains Are Enterotoxic but Do Not Cause Disease in Hamsters. J. Infect. Dis. 2006, 193, 1143–1150. [Google Scholar] [CrossRef]
  38. Warny, M.; Pepin, J.; Fang, A.; Killgore, G.; Thompson, A.; Brazier, J.; Frost, E.; McDonald, L.C. Toxin Production by an Emerging Strain of Clostridium Difficile Associated with Outbreaks of Severe Disease in North America and Europe. Lancet 2005, 366, 1079–1084. [Google Scholar] [CrossRef]
  39. Freeman, J.; Vernon, J.; Pilling, S.; Morris, K.; Nicholson, S.; Shearman, S.; Longshaw, C.; Wilcox, M.H. Pan-European Longitudinal Surveillance of Antibiotic Resistance among Prevalent Clostridium difficile Ribotypes Study Group The ClosER Study: Results from a Three-Year Pan-European Longitudinal Surveillance of Antibiotic Resistance among Prevalent Clostridium Difficile Ribotypes, 2011–2014. Clin. Microbiol. Infect. 2018, 24, 724–731. [Google Scholar] [CrossRef]
  40. Goldstein, E.J.C.; Citron, D.M.; Sears, P.; Babakhani, F.; Sambol, S.P.; Gerding, D.N. Comparative Susceptibilities to Fidaxomicin (OPT-80) of Isolates Collected at Baseline, Recurrence, and Failure from Patients in Two Phase III Trials of Fidaxomicin against Clostridium Difficile Infection. Antimicrob. Agents Chemother. 2011, 55, 5194–5199. [Google Scholar] [CrossRef]
  41. Cohen, N.A.; Miller, T.; Na’aminh, W.; Hod, K.; Adler, A.; Cohen, D.; Guzner-Gur, H.; Santo, E.; Halpern, Z.; Carmeli, Y.; et al. Clostridium Difficile Fecal Toxin Level Is Associated with Disease Severity and Prognosis. United Eur. Gastroenterol. J. 2018, 6, 773–780. [Google Scholar] [CrossRef]
  42. Gerding, D.N.; Johnson, S.; Rupnik, M.; Aktories, K. Clostridium Difficile Binary Toxin CDT: Mechanism, Epidemiology, and Potential Clinical Importance. Gut Microbes 2014, 5, 15–27. [Google Scholar] [CrossRef]
  43. Stubbs, S.; Rupnik, M.; Gibert, M.; Brazier, J.; Duerden, B.; Popoff, M. Production of Actin-Specific ADP-Ribosyltransferase (Binary Toxin) by Strains of Clostridium Difficile. FEMS Microbiol. Lett. 2000, 186, 307–312. [Google Scholar] [CrossRef]
  44. Donlan, A.; Petri, W.A. The Inflammasome and Type-2 Immunity in Clostridium Difficile Infection. Clin. Colon. Rectal Surg. 2020, 33, 67–72. [Google Scholar] [CrossRef]
  45. Cowardin, C.A.; Buonomo, E.L.; Saleh, M.M.; Wilson, M.G.; Burgess, S.L.; Kuehne, S.A.; Schwan, C.; Eichhoff, A.M.; Koch-Nolte, F.; Lyras, D.; et al. The Binary Toxin CDT Enhances Clostridium Difficile Virulence by Suppressing Protective Colonic Eosinophilia. Nat. Microbiol. 2016, 1, 16108. [Google Scholar] [CrossRef]
  46. 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: 2017 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]
  47. Stevens, V.W.; Nelson, R.E.; Schwab-Daugherty, E.M.; Khader, K.; Jones, M.M.; Brown, K.A.; Greene, T.; Croft, L.D.; Neuhauser, M.; Glassman, P.; et al. Comparative Effectiveness of Vancomycin and Metronidazole for the Prevention of Recurrence and Death in Patients with Clostridium Difficile Infection. JAMA Intern. Med. 2017, 177, 546–553. [Google Scholar] [CrossRef]
  48. 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] [PubMed]
  49. 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): 2021 Focused Update Guidelines on Management of Clostridioides Difficile Infection in Adults. Clin. Infect. Dis. 2021, 73, e1029–e1044. [Google Scholar] [CrossRef] [PubMed]
  50. Song, J.H.; Kim, Y.S. Recurrent Clostridium Difficile Infection: Risk Factors, Treatment, and Prevention. Gut Liver 2019, 13, 16–24. [Google Scholar] [CrossRef] [PubMed]
  51. Fu, Y.; Luo, Y.; Grinspan, A.M. Epidemiology of Community-Acquired and Recurrent Clostridioides Difficile Infection. Therap Adv. Gastroenterol. 2021, 14, 17562848211016248. [Google Scholar] [CrossRef]
  52. Tang-Feldman, Y.; Mayo, S.; Silva, J.; Cohen, S.H. Molecular Analysis of Clostridium Difficile Strains Isolated from 18 Cases of Recurrent Clostridium Difficile-Associated Diarrhea. J. Clin. Microbiol. 2003, 41, 3413–3414. [Google Scholar] [CrossRef] [PubMed]
  53. Wilcox, M.H.; Fawley, W.N.; Settle, C.D.; Davidson, A. Recurrence of Symptoms in Clostridium Difficile Infection--Relapse or Reinfection? J. Hosp. Infect. 1998, 38, 93–100. [Google Scholar] [CrossRef]
  54. Sun, X.; Wang, H.; Zhang, Y.; Chen, K.; Davis, B.; Feng, H. Mouse Relapse Model of Clostridium Difficile Infection. Infect. Immun. 2011, 79, 2856–2864. [Google Scholar] [CrossRef] [PubMed]
  55. Alonso, C.D.; Mahoney, M. V Bezlotoxumab for the Prevention of Clostridium Difficile Infection: A Review of Current Evidence and Safety Profile. Infect. Drug Resist. 2019, 12, 1–9. [Google Scholar] [CrossRef]
  56. Eiseman, B.; Silen, W.; Bascom, G.S.; Kauvar, A.J. Fecal Enema as an Adjunct in the Treatment of Pseudomembranous Enterocolitis. Surgery 1958, 44, 854–859. [Google Scholar]
  57. Smits, L.P.; Bouter, K.E.C.; de Vos, W.M.; Borody, T.J.; Nieuwdorp, M. Therapeutic Potential of Fecal Microbiota Transplantation. Gastroenterology 2013, 145, 946–953. [Google Scholar] [CrossRef]
  58. Gupta, S.; Allen-Vercoe, E.; Petrof, E.O. Fecal Microbiota Transplantation: In Perspective. Therap Adv. Gastroenterol. 2016, 9, 229–239. [Google Scholar] [CrossRef] [PubMed]
  59. Baunwall, S.M.D.; Lee, M.M.; Eriksen, M.K.; Mullish, B.H.; Marchesi, J.R.; Dahlerup, J.F.; Hvas, C.L. Faecal Microbiota Transplantation for Recurrent Clostridioides Difficile Infection: An Updated Systematic Review and Meta-Analysis. EClinicalMedicine 2020, 29–30, 100642. [Google Scholar] [CrossRef]
  60. Song, Y.N.; Yang, D.Y.; Veldhuyzen van Zanten, S.; Wong, K.; McArthur, E.; Song, C.Z.; Ianiro, G.; Cammarota, G.; Kelly, C.; Fischer, M.; et al. Fecal Microbiota Transplantation for Severe or Fulminant Clostridioides Difficile Infection: Systematic Review and Meta-Analysis. J. Can. Assoc. Gastroenterol. 2022, 5, e1–e11. [Google Scholar] [CrossRef]
  61. Sadowsky, M.J.; Khoruts, A. Faecal Microbiota Transplantation Is Promising but Not a Panacea. Nat. Microbiol. 2016, 1, 16015. [Google Scholar] [CrossRef]
  62. Brandt, L.J.; Aroniadis, O.C. An Overview of Fecal Microbiota Transplantation: Techniques, Indications, and Outcomes. Gastrointest. Endosc. 2013, 78, 240–249. [Google Scholar] [CrossRef] [PubMed]
  63. Kelly, B.J.; Tebas, P. Clinical Practice and Infrastructure Review of Fecal Microbiota Transplantation for Clostridium Difficile Infection. Chest 2018, 153, 266–277. [Google Scholar] [CrossRef] [PubMed]
  64. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef] [PubMed]
  65. Smith, M.I.; Yatsunenko, T.; Manary, M.J.; Trehan, I.; Mkakosya, R.; Cheng, J.; Kau, A.L.; Rich, S.S.; Concannon, P.; Mychaleckyj, J.C.; et al. Gut Microbiomes of Malawian Twin Pairs Discordant for Kwashiorkor. Science 2013, 339, 548–554. [Google Scholar] [CrossRef]
  66. Chang, T.W.; Bartlett, J.G.; Gorbach, S.L.; Onderdonk, A.B. Clindamycin-Induced Enterocolitis in Hamsters as a Model of Pseudomembranous Colitis in Patients. Infect. Immun. 1978, 20, 526–529. [Google Scholar] [CrossRef]
  67. Toshniwal, R.; Silva, J.; Fekety, R.; Kim, K.H. Studies on the Epidemiology of Colitis Due to Clostridium Difficile in Hamsters. J. Infect. Dis. 1981, 143, 51–54. [Google Scholar] [CrossRef]
  68. Toshniwal, R.; Fekety, R.; Silva, J. Etiology of Tetracycline-Associated Pseudomembranous Colitis in Hamsters. Antimicrob. Agents Chemother. 1979, 16, 167–170. [Google Scholar] [CrossRef]
  69. Price, A.B.; Larson, H.E.; Crow, J. Morphology of Experimental Antibiotic-Associated Enterocolitis in the Hamster: A Model for Human Pseudomembranous Colitis and Antibiotic-Associated Diarrhoea. Gut 1979, 20, 467–475. [Google Scholar] [CrossRef]
  70. Bartlett, J.G.; Onderdonk, A.B.; Cisneros, R.L.; Kasper, D.L. Clindamycin-Associated Colitis Due to a Toxin-Producing Species of Clostridium in Hamsters. J. Infect. Dis. 1977, 136, 701–705. [Google Scholar] [CrossRef]
  71. Rifkin, G.D.; Silva, J.; Fekety, R. Gastrointestinal and Systemic Toxicity of Fecal Extracts from Hamsters with Clindamycin-Induced Colitis. Gastroenterology 1978, 74, 52–57. [Google Scholar] [CrossRef]
  72. Torres, J.F.; Lyerly, D.M.; Hill, J.E.; Monath, T.P. Evaluation of Formalin-Inactivated Clostridium Difficile Vaccines Administered by Parenteral and Mucosal Routes of Immunization in Hamsters. Infect. Immun. 1995, 63, 4619–4627. [Google Scholar] [CrossRef] [PubMed]
  73. Lawley, T.D.; Clare, S.; Walker, A.W.; Goulding, D.; Stabler, R.A.; Croucher, N.; Mastroeni, P.; Scott, P.; Raisen, C.; Mottram, L.; et al. Antibiotic Treatment of Clostridium Difficile Carrier Mice Triggers a Supershedder State, Spore-Mediated Transmission, and Severe Disease in Immunocompromised Hosts. Infect. Immun. 2009, 77, 3661–3669. [Google Scholar] [CrossRef]
  74. Collignon, A. Methods for Working with the Mouse Model. Methods Mol. Biol. 2010, 646, 229–237. [Google Scholar] [CrossRef]
  75. Lawley, T.D.; Young, V.B. Murine Models to Study Clostridium Difficile Infection and Transmission. Anaerobe 2013, 24, 94–97. [Google Scholar] [CrossRef]
  76. Britton, R.A.; Young, V.B. Interaction between the Intestinal Microbiota and Host in Clostridium Difficile Colonization Resistance. Trends Microbiol. 2012, 20, 313–319. [Google Scholar] [CrossRef]
  77. Li, Y.; Figler, R.A.; Kolling, G.; Bracken, T.C.; Rieger, J.; Stevenson, R.W.; Linden, J.; Guerrant, R.L.; Warren, C.A. Adenosine A2A Receptor Activation Reduces Recurrence and Mortality from Clostridium Difficile Infection in Mice following Vancomycin Treatment. BMC Infect. Dis. 2012, 12, 342. [Google Scholar] [CrossRef]
  78. Castro-Córdova, P.; Díaz-Yáñez, F.; Muñoz-Miralles, J.; Gil, F.; Paredes-Sabja, D. Effect of Antibiotic to Induce Clostridioides Difficile-Susceptibility and Infectious Strain in a Mouse Model of Clostridioides Difficile Infection and Recurrence. Anaerobe 2020, 62, 102149. [Google Scholar] [CrossRef] [PubMed]
  79. Fachi, J.L.; Felipe, J.d.S.; Pral, L.P.; da Silva, B.K.; Corrêa, R.O.; de Andrade, M.C.P.; da Fonseca, D.M.; Basso, P.J.; Câmara, N.O.S.; de Sales E Souza, É.L.; et al. Butyrate Protects Mice from Clostridium Difficile-Induced Colitis through an HIF-1-Dependent Mechanism. Cell Rep. 2019, 27, 750–761. [Google Scholar] [CrossRef] [PubMed]
  80. Chen, X.; Katchar, K.; Goldsmith, J.D.; Nanthakumar, N.; Cheknis, A.; Gerding, D.N.; Kelly, C.P. A Mouse Model of Clostridium Difficile-Associated Disease. Gastroenterology 2008, 135, 1984–1992. [Google Scholar] [CrossRef]
  81. De Wolfe, T.J.; Kates, A.E.; Barko, L.; Darien, B.J.; Safdar, N. Modified Mouse Model of Clostridioides Difficile Infection as a Platform for Probiotic Efficacy Studies. Antimicrob. Agents Chemother. 2019, 63, e00111-19. [Google Scholar] [CrossRef]
  82. Lyerly, D.M.; Saum, K.E.; MacDonald, D.K.; Wilkins, T.D. Effects of Clostridium Difficile Toxins given Intragastrically to Animals. Infect. Immun. 1985, 47, 349–352. [Google Scholar] [CrossRef]
  83. Hirota, S.A.; Iablokov, V.; Tulk, S.E.; Schenck, L.P.; Becker, H.; Nguyen, J.; Al Bashir, S.; Dingle, T.C.; Laing, A.; Liu, J.; et al. Intrarectal Instillation of Clostridium Difficile Toxin A Triggers Colonic Inflammation and Tissue Damage: Development of a Novel and Efficient Mouse Model of Clostridium Difficile Toxin Exposure. Infect. Immun. 2012, 80, 4474–4484. [Google Scholar] [CrossRef]
  84. Savidge, T.C.; Pan, W.-H.; Newman, P.; O’brien, M.; Anton, P.M.; Pothoulakis, C. Clostridium Difficile Toxin B Is an Inflammatory Enterotoxin in Human Intestine. Gastroenterology 2003, 125, 413–420. [Google Scholar] [CrossRef]
  85. Lucas, F.; Elmer, G.W.; Brot-Laroche, E.; Corthier, G. Fixation of Clostridium Difficile Toxin A and Cholera Toxin to Intestinal Brush Border Membranes from Axenic and Conventional Mice. Infect. Immun. 1989, 57, 1680–1683. [Google Scholar] [CrossRef] [PubMed]
  86. Lesniak, N.A.; Schubert, A.M.; Flynn, K.J.; Leslie, J.L.; Sinani, H.; Bergin, I.L.; Young, V.B.; Schloss, P.D. The Gut Bacterial Community Potentiates Clostridioides Difficile Infection Severity. mBio 2022, 13, e0118322. [Google Scholar] [CrossRef] [PubMed]
  87. 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]
  88. Lawley, T.D.; Clare, S.; Walker, A.W.; Stares, M.D.; Connor, T.R.; Raisen, C.; Goulding, D.; Rad, R.; Schreiber, F.; Brandt, C.; et al. Targeted Restoration of the Intestinal Microbiota with a Simple, Defined Bacteriotherapy Resolves Relapsing Clostridium Difficile Disease in Mice. PLoS Pathog. 2012, 8, e1002995. [Google Scholar] [CrossRef] [PubMed]
  89. Corthier, G.; Dubos, F.; Ducluzeau, R. Prevention of Clostridium Difficile Induced Mortality in Gnotobiotic Mice by Saccharomyces Boulardii. Can. J. Microbiol. 1986, 32, 894–896. [Google Scholar] [CrossRef]
  90. Wilson, K.H.; Freter, R. Interaction of Clostridium Difficile and Escherichia Coli with Microfloras in Continuous-Flow Cultures and Gnotobiotic Mice. Infect. Immun. 1986, 54, 354–358. [Google Scholar] [CrossRef] [PubMed]
  91. Wilson, K.H.; Perini, F. Role of Competition for Nutrients in Suppression of Clostridium Difficile by the Colonic Microflora. Infect. Immun. 1988, 56, 2610–2614. [Google Scholar] [CrossRef] [PubMed]
  92. Pawlowski, S.W.; Calabrese, G.; Kolling, G.L.; Platts-Mills, J.; Freire, R.; AlcantaraWarren, C.; Liu, B.; Sartor, R.B.; Guerrant, R.L. Murine Model of Clostridium Difficile Infection with Aged Gnotobiotic C57BL/6 Mice and a BI/NAP1 Strain. J. Infect. Dis. 2010, 202, 1708–1712. [Google Scholar] [CrossRef] [PubMed]
  93. Wilson, K.H.; Sheagren, J.N.; Freter, R.; Weatherbee, L.; Lyerly, D. Gnotobiotic Models for Study of the Microbial Ecology of Clostridium Difficile and Escherichia Coli. J. Infect. Dis. 1986, 153, 547–551. [Google Scholar] [CrossRef]
  94. Onderdonk, A.B.; Cisneros, R.L.; Bartlett, J.G. Clostridium Difficile in Gnotobiotic Mice. Infect. Immun. 1980, 28, 277–282. [Google Scholar] [CrossRef] [PubMed]
  95. Torres, J.; Jennische, E.; Lange, S.; Lönnroth, I. Enterotoxins from Clostridium Difficile; Diarrhoeogenic Potency and Morphological Effects in the Rat Intestine. Gut 1990, 31, 781–785. [Google Scholar] [CrossRef]
  96. Reeves, A.E.; Koenigsknecht, M.J.; Bergin, I.L.; Young, V.B. Suppression of Clostridium Difficile in the Gastrointestinal Tracts of Germfree Mice Inoculated with a Murine Isolate from the Family Lachnospiraceae. Infect. Immun. 2012, 80, 3786–3794. [Google Scholar] [CrossRef] [PubMed]
  97. Louie, T.; Golan, Y.; Khanna, S.; Bobilev, D.; Erpelding, N.; Fratazzi, C.; Carini, M.; Menon, R.; Ruisi, M.; Norman, J.M.; et al. VE303, a Defined Bacterial Consortium, for Prevention of Recurrent Clostridioides Difficile Infection: A Randomized Clinical Trial. JAMA 2023, 329, 1356–1366. [Google Scholar] [CrossRef]
  98. Bobilev, D.; Bhattarai, S.; Menon, R.; Klein, B.; Reddy, S.; Olle, B.; Roberts, B.; Bucci, V.; Norman, J. 1953. VE303, a Rationally Designed Bacterial Consortium for Prevention of Recurrent Clostridioides Difficile (C. Difficile) Infection (RCDI), Stably Restores the Gut Microbiota After Vancomycin (Vanco)-Induced Dysbiosis in Adult Healthy Volunteers (HV). Open Forum Infect Dis 2019, 6, S60. [Google Scholar] [CrossRef]
  99. Raibaud, P.; Ducluzeau, R.; Dubos, F.; Hudault, S.; Bewa, H.; Muller, M.C. Implantation of Bacteria from the Digestive Tract of Man and Various Animals into Gnotobiotic Mice. Am. J. Clin. Nutr. 1980, 33, 2440–2447. [Google Scholar] [CrossRef]
  100. Hazenberg, M.P.; Bakker, M.; Verschoor-Burggraaf, A. Effects of the Human Intestinal Flora on Germ-Free Mice. J. Appl. Bacteriol. 1981, 50, 95–106. [Google Scholar] [CrossRef]
  101. Péchiné, S.; Janoir, C.; Boureau, H.; Gleizes, A.; Tsapis, N.; Hoys, S.; Fattal, E.; Collignon, A. Diminished Intestinal Colonization by Clostridium Difficile and Immune Response in Mice after Mucosal Immunization with Surface Proteins of Clostridium Difficile. Vaccine 2007, 25, 3946–3954. [Google Scholar] [CrossRef] [PubMed]
  102. Collins, J.; Auchtung, J.M.; Schaefer, L.; Eaton, K.A.; Britton, R.A. Humanized Microbiota Mice as a Model of Recurrent Clostridium Difficile Disease. Microbiome 2015, 3, 35. [Google Scholar] [CrossRef] [PubMed]
  103. Schubert, A.M.; Sinani, H.; Schloss, P.D. Antibiotic-Induced Alterations of the Murine Gut Microbiota and Subsequent Effects on Colonization Resistance against Clostridium Difficile. mBio 2015, 6, e00974. [Google Scholar] [CrossRef] [PubMed]
  104. Seekatz, A.M.; Safdar, N.; Khanna, S. The Role of the Gut Microbiome in Colonization Resistance and Recurrent Clostridioides Difficile Infection. Therap. Adv. Gastroenterol. 2022, 15, 17562848221134396. [Google Scholar] [CrossRef] [PubMed]
  105. Reeves, A.E.; Theriot, C.M.; Bergin, I.L.; Huffnagle, G.B.; Schloss, P.D.; Young, V.B. The Interplay between Microbiome Dynamics and Pathogen Dynamics in a Murine Model of Clostridium Difficile Infection. Gut Microbes 2011, 2, 145–158. [Google Scholar] [CrossRef] [PubMed]
  106. Buffie, C.G.; Jarchum, I.; Equinda, M.; Lipuma, L.; Gobourne, A.; Viale, A.; Ubeda, C.; Xavier, J.; Pamer, E.G. Profound Alterations of Intestinal Microbiota following a Single Dose of Clindamycin Results in Sustained Susceptibility to Clostridium Difficile-Induced Colitis. Infect. Immun. 2012, 80, 62–73. [Google Scholar] [CrossRef]
  107. Theriot, C.M.; Koumpouras, C.C.; Carlson, P.E.; Bergin, I.I.; Aronoff, D.M.; Young, V.B. Cefoperazone-Treated Mice as an Experimental Platform to Assess Differential Virulence of Clostridium Difficile Strains. Gut Microbes 2011, 2, 326–334. [Google Scholar] [CrossRef]
  108. Theriot, C.M.; Schumacher, C.A.; Bassis, C.M.; Seekatz, A.M.; Young, V.B. Effects of Tigecycline and Vancomycin Administration on Established Clostridium Difficile Infection. Antimicrob. Agents Chemother. 2015, 59, 1596–1604. [Google Scholar] [CrossRef]
  109. Kiełbasiński, K.; Peszek, W.; Grabarek, B.O.; Boroń, D.; Wierzbik-Strońska, M.; Oplawski, M. Effect of Salinomycin on Expression Pattern of Genes Associated with Apoptosis in Endometrial Cancer Cell Line. Curr. Pharm. Biotechnol. 2020, 21, 1269–1277. [Google Scholar] [CrossRef]
  110. Tomkovich, S.; Stough, J.M.A.; Bishop, L.; Schloss, P.D. The Initial Gut Microbiota and Response to Antibiotic Perturbation Influence Clostridioides Difficile Clearance in Mice. mSphere 2020, 5, e00869-20. [Google Scholar] [CrossRef]
  111. Lesniak, N.A.; Schubert, A.M.; Sinani, H.; Schloss, P.D. Clearance of Clostridioides Difficile Colonization Is Associated with Antibiotic-Specific Bacterial Changes. mSphere 2021, 6, e01238-20. [Google Scholar] [CrossRef]
  112. Kokkotou, E.; Moss, A.C.; Michos, A.; Espinoza, D.; Cloud, J.W.; Mustafa, N.; O’Brien, M.; Pothoulakis, C.; Kelly, C.P. Comparative Efficacies of Rifaximin and Vancomycin for Treatment of Clostridium Difficile-Associated Diarrhea and Prevention of Disease Recurrence in Hamsters. Antimicrob. Agents Chemother. 2008, 52, 1121–1126. [Google Scholar] [CrossRef]
  113. Lawley, T.D.; Croucher, N.J.; Yu, L.; Clare, S.; Sebaihia, M.; Goulding, D.; Pickard, D.J.; Parkhill, J.; Choudhary, J.; Dougan, G. Proteomic and Genomic Characterization of Highly Infectious Clostridium Difficile 630 Spores. J. Bacteriol. 2009, 191, 5377–5386. [Google Scholar] [CrossRef] [PubMed]
  114. Jump, R.L. Clostridium Difficile Infection in Older Adults. Aging Health 2013, 9, 403–414. [Google Scholar] [CrossRef] [PubMed]
  115. Asempa, T.E.; Nicolau, D.P. Clostridium Difficile Infection in the Elderly: An Update on Management. Clin. Interv. Aging 2017, 12, 1799–1809. [Google Scholar] [CrossRef] [PubMed]
  116. 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]
  117. Yurist-Doutsch, S.; Arrieta, M.-C.; Vogt, S.L.; Finlay, B.B. Gastrointestinal Microbiota-Mediated Control of Enteric Pathogens. Annu. Rev. Genet. 2014, 48, 361–382. [Google Scholar] [CrossRef]
  118. Koenigsknecht, M.J.; Theriot, C.M.; Bergin, I.L.; Schumacher, C.A.; Schloss, P.D.; Young, V.B. Dynamics and Establishment of Clostridium Difficile Infection in the Murine Gastrointestinal Tract. Infect. Immun. 2015, 83, 934–941. [Google Scholar] [CrossRef]
  119. Feltis, B.A.; Kim, A.S.; Kinneberg, K.M.; Lyerly, D.L.; Wilkins, T.D.; Erlandsen, S.L.; Wells, C.L. Clostridium Difficile Toxins May Augment Bacterial Penetration of Intestinal Epithelium. Arch. Surg. 1999, 134, 1235–1241. [Google Scholar] [CrossRef]
  120. Naaber, P.; Mikelsaar, R.H.; Salminen, S.; Mikelsaar, M. Bacterial Translocation, Intestinal Microflora and Morphological Changes of Intestinal Mucosa in Experimental Models of Clostridium Difficile Infection. J. Med. Microbiol. 1998, 47, 591–598. [Google Scholar] [CrossRef]
  121. Fachi, J.L.; Sécca, C.; Rodrigues, P.B.; de Mato, F.C.P.; Di Luccia, B.; Felipe, J.d.S.; Pral, L.P.; Rungue, M.; Rocha, V.D.M.; Sato, F.T.; et al. Acetate Coordinates Neutrophil and ILC3 Responses against C. Difficile through FFAR2. J. Exp. Med. 2020, 217, e20190489. [Google Scholar] [CrossRef]
  122. Hasegawa, M.; Yamazaki, T.; Kamada, N.; Tawaratsumida, K.; Kim, Y.-G.; Núñez, G.; Inohara, N. Nucleotide-Binding Oligomerization Domain 1 Mediates Recognition of Clostridium Difficile and Induces Neutrophil Recruitment and Protection against the Pathogen. J. Immunol. 2011, 186, 4872–4880. [Google Scholar] [CrossRef]
  123. Hasegawa, M.; Kamada, N.; Jiao, Y.; Liu, M.Z.; Núñez, G.; Inohara, N. Protective Role of Commensals against Clostridium Difficile Infection via an IL-1β-Mediated Positive-Feedback Loop. J. Immunol. 2012, 189, 3085–3091. [Google Scholar] [CrossRef]
  124. Steele, J.; Chen, K.; Sun, X.; Zhang, Y.; Wang, H.; Tzipori, S.; Feng, H. Systemic Dissemination of Clostridium Difficile Toxins A and B Is Associated with Severe, Fatal Disease in Animal Models. J. Infect. Dis. 2012, 205, 384–391. [Google Scholar] [CrossRef] [PubMed]
  125. Yu, H.; Chen, K.; Sun, Y.; Carter, M.; Garey, K.W.; Savidge, T.C.; Devaraj, S.; Tessier, M.E.; von Rosenvinge, E.C.; Kelly, C.P.; et al. Cytokines Are Markers of the Clostridium Difficile-Induced Inflammatory Response and Predict Disease Severity. Clin. Vaccine Immunol. 2017, 24, e00037-17. [Google Scholar] [CrossRef] [PubMed]
  126. Abid, H.; Bischof, E. An Unusual Presentation of Severe Sepsis Due to Clostridium Difficile Enteritis. Cureus 2019, 11, e4162. [Google Scholar] [CrossRef] [PubMed]
  127. 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]
  128. Abt, M.C.; Lewis, B.B.; Caballero, S.; Xiong, H.; Carter, R.A.; Sušac, B.; Ling, L.; Leiner, I.; Pamer, E.G. Innate Immune Defenses Mediated by Two ILC Subsets Are Critical for Protection against Acute Clostridium Difficile Infection. Cell Host Microbe 2015, 18, 27–37. [Google Scholar] [CrossRef] [PubMed]
  129. Jarchum, I.; Liu, M.; Shi, C.; Equinda, M.; Pamer, E.G. Critical Role for MyD88-Mediated Neutrophil Recruitment during Clostridium Difficile Colitis. Infect. Immun. 2012, 80, 2989–2996. [Google Scholar] [CrossRef] [PubMed]
  130. McDermott, A.J.; Falkowski, N.R.; McDonald, R.A.; Frank, C.R.; Pandit, C.R.; Young, V.B.; Huffnagle, G.B. Role of Interferon-γ and Inflammatory Monocytes in Driving Colonic Inflammation during Acute Clostridium Difficile Infection in Mice. Immunology 2017, 150, 468–477. [Google Scholar] [CrossRef]
  131. Cowardin, C.A.; Kuehne, S.A.; Buonomo, E.L.; Marie, C.S.; Minton, N.P.; Petri, W.A. Inflammasome Activation Contributes to Interleukin-23 Production in Response to Clostridium Difficile. mBio 2015, 6, e02386-14. [Google Scholar] [CrossRef]
  132. Ng, J.; Hirota, S.A.; Gross, O.; Li, Y.; Ulke-Lemee, A.; Potentier, M.S.; Schenck, L.P.; Vilaysane, A.; Seamone, M.E.; Feng, H.; et al. Clostridium Difficile Toxin-Induced Inflammation and Intestinal Injury Are Mediated by the Inflammasome. Gastroenterology 2010, 139, 542–552. [Google Scholar] [CrossRef]
  133. Simpson, M.; Frisbee, A.; Kumar, P.; Schwan, C.; Aktories, K.; Petri, W.A. Clostridioides Difficile Binary Toxin Is Recognized by the Toll-Like Receptor 2/6 Heterodimer to Induce a Nuclear Factor-ΚB Response. J. Infect. Dis. 2022, 225, 1296–1300. [Google Scholar] [CrossRef]
  134. Schulze-Osthoff, K.; Ferrari, D.; Riehemann, K.; Wesselborg, S. Regulation of NF-ΚB Activation by MAP Kinase Cascades. Immunobiology 1997, 198, 35–49. [Google Scholar] [CrossRef] [PubMed]
  135. Corthier, G.; Muller, M.C.; Wilkins, T.D.; Lyerly, D.; L’Haridon, R. Protection against Experimental Pseudomembranous Colitis in Gnotobiotic Mice by Use of Monoclonal Antibodies against Clostridium Difficile Toxin A. Infect. Immun. 1991, 59, 1192–1195. [Google Scholar] [CrossRef] [PubMed]
  136. Rodrigues, R.S.; Oliveira, R.A.C.; Li, Y.; Zaja-Milatovic, S.; Costa, L.B.; Braga Neto, M.B.; Kolling, G.L.; Lima, A.A.; Guerrant, R.L.; Warren, C.A. Intestinal Epithelial Restitution after TcdB Challenge and Recovery from Clostridium Difficile Infection in Mice with Alanyl-Glutamine Treatment. J. Infect. Dis. 2013, 207, 1505–1515. [Google Scholar] [CrossRef] [PubMed]
  137. Mileto, S.J.; Jardé, T.; Childress, K.O.; Jensen, J.L.; Rogers, A.P.; Kerr, G.; Hutton, M.L.; Sheedlo, M.J.; Bloch, S.C.; Shupe, J.A.; et al. Clostridioides Difficile Infection Damages Colonic Stem Cells via TcdB, Impairing Epithelial Repair and Recovery from Disease. Proc. Natl. Acad. Sci. USA 2020, 117, 8064–8073. [Google Scholar] [CrossRef] [PubMed]
  138. Nagao-Kitamoto, H.; Leslie, J.L.; Kitamoto, S.; Jin, C.; Thomsson, K.A.; Gillilland, M.G.; Kuffa, P.; Goto, Y.; Jenq, R.R.; Ishii, C.; et al. Interleukin-22-Mediated Host Glycosylation Prevents Clostridioides Difficile Infection by Modulating the Metabolic Activity of the Gut Microbiota. Nat. Med. 2020, 26, 608–617. [Google Scholar] [CrossRef] [PubMed]
  139. Cribas, E.S.; Denny, J.E.; Maslanka, J.R.; Abt, M.C. Loss of Interleukin-10 (IL-10) Signaling Promotes IL-22-Dependent Host Defenses against Acute Clostridioides Difficile Infection. Infect. Immun. 2021, 89, e00730-20. [Google Scholar] [CrossRef] [PubMed]
  140. Kim, M.N.; Koh, S.-J.; Kim, J.M.; Im, J.P.; Jung, H.C.; Kim, J.S. Clostridium Difficile Infection Aggravates Colitis in Interleukin 10-Deficient Mice. World J. Gastroenterol. 2014, 20, 17084–17091. [Google Scholar] [CrossRef]
  141. Hasegawa, M.; Yada, S.; Liu, M.Z.; Kamada, N.; Muñoz-Planillo, R.; Do, N.; Núñez, G.; Inohara, N. Interleukin-22 Regulates the Complement System to Promote Resistance against Pathobionts after Pathogen-Induced Intestinal Damage. Immunity 2014, 41, 620–632. [Google Scholar] [CrossRef]
  142. Alam, M.Z.; Maslanka, J.R.; Denny, J.E.; Bittinger, K.; Abt, M.C. IL10 Producing Regulatory T Cells Support Fecal Microbiota Transplant (FMT)-Mediated Clearance of Clostridioides Difficile Infection. J. Immunol. 2023, 210, 61.12. [Google Scholar] [CrossRef]
  143. Littmann, E.R.; Lee, J.-J.; Denny, J.E.; Alam, Z.; Maslanka, J.R.; Zarin, I.; Matsuda, R.; Carter, R.A.; Susac, B.; Saffern, M.S.; et al. Host Immunity Modulates the Efficacy of Microbiota Transplantation for Treatment of Clostridioides Difficile Infection. Nat. Commun. 2021, 12, 755. [Google Scholar] [CrossRef]
  144. Vardakas, K.Z.; Polyzos, K.A.; Patouni, K.; Rafailidis, P.I.; Samonis, G.; Falagas, M.E. Treatment Failure and Recurrence of Clostridium Difficile Infection following Treatment with Vancomycin or Metronidazole: A Systematic Review of the Evidence. Int. J. Antimicrob. Agents 2012, 40, 1–8. [Google Scholar] [CrossRef] [PubMed]
  145. Shields, K.; Araujo-Castillo, R.V.; Theethira, T.G.; Alonso, C.D.; Kelly, C.P. Recurrent Clostridium Difficile Infection: From Colonization to Cure. Anaerobe 2015, 34, 59–73. [Google Scholar] [CrossRef] [PubMed]
  146. Shah, H.B.; Smith, K.; Scott, E.J.; Larabee, J.L.; James, J.A.; Ballard, J.D.; Lang, M.L. Human C. Difficile Toxin-Specific Memory B Cell Repertoires Encode Poorly Neutralizing Antibodies. JCI Insight 2020, 5, e138137. [Google Scholar] [CrossRef] [PubMed]
  147. Johnson, S.; Gerding, D.N.; Janoff, E.N. Systemic and Mucosal Antibody Responses to Toxin A in Patients Infected with Clostridium Difficile. J. Infect. Dis. 1992, 166, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
  148. Drudy, D.; Calabi, E.; Kyne, L.; Sougioultzis, S.; Kelly, E.; Fairweather, N.; Kelly, C.P. Human Antibody Response to Surface Layer Proteins in Clostridium Difficile Infection. FEMS Immunol. Med. Microbiol. 2004, 41, 237–242. [Google Scholar] [CrossRef] [PubMed]
  149. Johnston, P.F.; Gerding, D.N.; Knight, K.L. Protection from Clostridium Difficile Infection in CD4 T Cell- and Polymeric Immunoglobulin Receptor-Deficient Mice. Infect. Immun. 2014, 82, 522–531. [Google Scholar] [CrossRef] [PubMed]
  150. Hernández Del Pino, R.E.; Barbero, A.M.; Español, L.Á.; Morro, L.S.; Pasquinelli, V. The Adaptive Immune Response to Clostridioides Difficile: A Tricky Balance between Immunoprotection and Immunopathogenesis. J. Leukoc. Biol. 2021, 109, 195–210. [Google Scholar] [CrossRef] [PubMed]
  151. Rees, W.D.; Steiner, T.S. Adaptive Immune Response to Clostridium Difficile Infection: A Perspective for Prevention and Therapy. Eur. J. Immunol. 2018, 48, 398–406. [Google Scholar] [CrossRef] [PubMed]
  152. 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]
  153. Cook, L.; Rees, W.D.; Wong, M.Q.; Peters, H.; Levings, M.K.; Steiner, T.S. Fecal Microbiota Transplantation for Recurrent Clostridioides Difficile Infection Enhances Adaptive Immunity to C Difficile Toxin B. Gastroenterology 2021, 160, 2155–2158. [Google Scholar] [CrossRef] [PubMed]
  154. 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]
  155. Frisbee, A.L.; Saleh, M.M.; Young, M.K.; Leslie, J.L.; Simpson, M.E.; Abhyankar, M.M.; Cowardin, C.A.; Ma, J.Z.; Pramoonjago, P.; Turner, S.D.; et al. IL-33 Drives Group 2 Innate Lymphoid Cell-Mediated Protection during Clostridium Difficile Infection. Nat. Commun. 2019, 10, 2712. [Google Scholar] [CrossRef]
Figure 1. Pathogenesis of C. difficile infection in the mouse model. Mice are infected after antibiotic treatment. The disease is usually acute, lasting for less than a week for the first episode. After antibiotic treatment and microbiota disruption, C. difficile spores germinate and colonize the colon, releasing toxins that damage the intestinal epithelium. This damage can potentially lead to bacterial translocation and inflammation, primarily driven by neutrophil infiltration, which peaks around day 2 postinfection. On day 5 postinfection, a resolving phase begins, characterized by reduced inflammation and increased tissue repair mechanisms, including the involvement of ILC3 and Treg cells. Around 20–25% of infected mice may experience recurrent infection relapse, offering valuable insights into the role of adaptive immunity in CDI.
Figure 1. Pathogenesis of C. difficile infection in the mouse model. Mice are infected after antibiotic treatment. The disease is usually acute, lasting for less than a week for the first episode. After antibiotic treatment and microbiota disruption, C. difficile spores germinate and colonize the colon, releasing toxins that damage the intestinal epithelium. This damage can potentially lead to bacterial translocation and inflammation, primarily driven by neutrophil infiltration, which peaks around day 2 postinfection. On day 5 postinfection, a resolving phase begins, characterized by reduced inflammation and increased tissue repair mechanisms, including the involvement of ILC3 and Treg cells. Around 20–25% of infected mice may experience recurrent infection relapse, offering valuable insights into the role of adaptive immunity in CDI.
Microorganisms 12 00273 g001
Table 1. Clinical score for evaluating the murine C. difficile infection.
Table 1. Clinical score for evaluating the murine C. difficile infection.
CategoryScore *
0123
ActivityNormalAlert/Slow movingLethargic/ShakyInactive unless prodded
PostureNormalBack slantedHunchedHunched/Nose down
PelageNormalPiloerectionRough skinVery ruffled Puff/Ungroomed
DiarrheaNormalSoft stool/Discolored (yellowish)Wet stained tail/mucous +/− bloodLiquid or absence of stool (ileus)
Eyes/NoseNormalSquinted
1/2 closed
Squinted/DischargeClosed/Discharge
* Clinical score = sum of all parameter scores. Total possible score is 15 (death).
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

Fachi, J.L.; Vinolo, M.A.R.; Colonna, M. Reviewing the Clostridioides difficile Mouse Model: Insights into Infection Mechanisms. Microorganisms 2024, 12, 273. https://doi.org/10.3390/microorganisms12020273

AMA Style

Fachi JL, Vinolo MAR, Colonna M. Reviewing the Clostridioides difficile Mouse Model: Insights into Infection Mechanisms. Microorganisms. 2024; 12(2):273. https://doi.org/10.3390/microorganisms12020273

Chicago/Turabian Style

Fachi, José L., Marco A. R. Vinolo, and Marco Colonna. 2024. "Reviewing the Clostridioides difficile Mouse Model: Insights into Infection Mechanisms" Microorganisms 12, no. 2: 273. https://doi.org/10.3390/microorganisms12020273

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