Skip to main content
Advertisement
  • Loading metrics

A short chain fatty acid–centric view of Clostridioides difficile pathogenesis

  • Anna L. Gregory ,

    Contributed equally to this work with: Anna L. Gregory, Daniel A. Pensinger

    Affiliations Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America, Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America, Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America

  • Daniel A. Pensinger ,

    Contributed equally to this work with: Anna L. Gregory, Daniel A. Pensinger

    Affiliations Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America, Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America

  • Andrew J. Hryckowian

    hryckowian@medicine.wisc.edu

    Affiliations Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America, Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, United States of America

Abstract

Clostridioides difficile is an opportunistic diarrheal pathogen responsible for significant morbidity and mortality worldwide. A disrupted (dysbiotic) gut microbiome, commonly engendered by antibiotic treatment, is the primary risk factor for C. difficile infection, highlighting that C. difficile–microbiome interactions are critical for determining the fitness of this pathogen. Here, we review short chain fatty acids (SCFAs): a major class of metabolites present in the gut, their production by the gut microbiome, and their impacts on the biology of the host and of C. difficile. We use these observations to illustrate a conceptual model whereby C. difficile senses and responds to SCFAs as a marker of a healthy gut and tunes its virulence accordingly in order to maintain dysbiosis. Future work to learn the molecular mechanisms and genetic circuitry underlying the relationships between C. difficile and SCFAs will help to identify precision approaches, distinct from antibiotics and fecal transplant, for mitigating disease caused by C. difficile and will inform similar investigations into other gastrointestinal pathogens.

Short chain fatty acids are major metabolic end products of gut microbiome community metabolism

The gut microbiome produces a vast array of molecules, which have local and systemic effects on our biology (reviewed in [1,2]). A sum of biogeographical and metabolic factors dictates the nutrients accessible to, and metabolites produced by, this microbial community. While amino acids are less energetically favorable to catabolize than carbohydrates, many gut microbes use amino acids for energy generation via Stickland metabolism [3]. Additionally, fats, proteins, and simple carbohydrates are efficiently absorbed by the host small intestine, leaving few of these metabolites to support the growth of microbes in the large intestine [4,5]. Conversely, complex polysaccharides (e.g., dietary fiber) are not metabolized by the host: The human genome encodes 17 carbohydrate-degrading enzymes, leaving us unable to extract calories from dietary fiber. So, due to our limited repertoire of enzymes, dietary fiber transits to the distal gut and serves as the preferred food source for many members of our microbiome: Gut-resident bacteria of humans and other animals encode tens of thousands of carbohydrate-degrading enzymes capable of breaking various polysaccharide linkages [6].

Monosaccharides that result from dietary fiber degradation by the microbiome enter microbial metabolism via glycolysis or the pentose phosphate pathway and are metabolized to pyruvate. From pyruvate, molecules are further converted into propionate through succinate or lactate, and acetate and butyrate are produced through an acetyl-CoA intermediate or through succinate via the tricarboxylic acid (TCA) cycle [7]. Glutamate and lysine are additional precursors for butyrate, and multiple amino acids are potential precursors for propionate [8]. A substantial amount of cross-feeding occurs among gut microbes. First, primary consumers ferment polysaccharides to lactate, ethanol, or acetate and excrete these metabolites. Subsequently, secondary consumers take up these metabolites and ultimately convert the intermediate products into the short chain fatty acids (SCFAs) propionate and butyrate [912]. In general, members of the phylum Bacteroidetes are dominant primary fermenters, which metabolize complex carbohydrates, while the Firmicutes are major secondary consumers [13,14]. In addition, acetate can be produced independently of carbohydrate metabolism by gut microbes via acetogenesis (also known as the Wood–Ljungdahl pathway), which produces approximately one-third of the total acetate in the colon [15].

Given the prominence of carbohydrate metabolism by the distal gut microbiome, the SCFAs acetate, propionate, and butyrate are the most abundant microbiome-produced molecules in the gut [12]. Importantly, acetate, propionate, and butyrate comprise >95% of the SCFAs in the gut, while other SCFAs such as formate, valerate, and caproate compose the remaining approximately 5% [16], and the absolute and relative abundance of these SCFAs differ based on host diet and microbiome composition [17]. By extension, low concentrations of SCFAs are often used as a biomarker of a disrupted (dysbiotic) gut microbiome. However, SCFAs are not merely microbial waste products: Substantial amounts are absorbed by host epithelial cells over the length of the colon. For example, one study in humans demonstrated that acetate, propionate, and butyrate are found at concentrations of 69, 25, and 26 mM, respectively, in the human proximal colon and 50, 19, and 17 mM, respectively, in the distal colon ([18] and Fig 1). Comparisons of SCFA levels in feces across humans, nonhuman primates, mice, and rats found high levels of SCFAs across all species examined. Therefore, animal models, especially commonly used rodent models, are useful tools to help understand how SCFAs affect microbe–microbe and microbe–host interactions in the human gastrointestinal tract [19].

thumbnail
Fig 1. Graphical representation of levels of SCFAs, O2, and pH through the longitude of the distal gastrointestinal tract.

The green line represents the partial pressure of oxygen (pO2 [mm Hg], left axis) through the longitude of the mouse gastrointestinal tract. Levels of oxygen are elevated in the proximal small intestine and are present at negligible amounts in the cecum and more distal portions of the gut. Data on pO2 were adapted from [24]. The orange, red, and blue lines represent the concentration of the SCFAs acetate, propionate, and butyrate, respectively, through the longitude of the human gastrointestinal tract (mM, left axis). In general, concentrations of these SCFAs are highest in the cecum and decrease through the more distal portions of the gut. These changes in concentration are a function of absorption by the host and continued production/consumption by gut microbes. The black line represents pH of intestinal contents through the longitude of the human gastrointestinal tract (pH, right axis). Data on SCFA concentration and pH were adapted from [18]. SCFA, short chain fatty acid.

https://doi.org/10.1371/journal.ppat.1009959.g001

Short chain fatty acids have pleiotropic beneficial effects on host biology

SCFAs are rapidly absorbed from the human colon where they can be metabolized by colonocytes or are transported via portal circulation to the liver where they are metabolized or passed into systemic circulation [18,20]. These varied fates of SCFAs underscore the diverse beneficial effects they have on host biology and highlight some of the possible consequences of a microbiome deficient in SCFA production.

Butyrate serves as the primary energy source for colonocytes, providing 60% to 70% of the cell’s energy. In colonocytes, butyrate undergoes β-oxidation to acetate, which enters the TCA cycle [21]. This process requires oxygen, which colonocytes consume from the lumen of the gut, resulting in low concentrations of oxygen in colonic epithelial cells and contributing to the anaerobic environment of the distal gut (Fig 1 and [2224]). Low levels of oxygen in the distal gut facilitate the survival of anaerobes, supporting continued gut microbiome community metabolism. Insufficient levels of butyrate shift colonocytes toward anaerobic glycolysis, leading to the accumulation of oxygen in the gut lumen, the loss of epithelial hypoxia, and dysbiosis (reviewed in [25]).

Acetate, propionate, and butyrate are metabolized by host tissues beyond the gut. SCFAs produced by the microbiome enter the liver via the portal vein. In the liver, propionate is utilized as a substrate for gluconeogenesis and acetate as a substrate for de novo lipogenesis and cholesterol synthesis. Propionate and acetate that are not metabolized by the liver enter systemic circulation, where they reach concentrations of up to 1.2 μM and 42 μM, respectively, and are then metabolized by muscle and other tissues [2629]. The liver also utilizes butyrate for lipid biosynthesis and glycolipid metabolism, though the concentrations of butyrate are low in this tissue, and negligible amounts are found in systemic circulation (reviewed in [25]).

The beneficial impacts of SCFAs extend beyond their roles as metabolites. Acetate, propionate, and butyrate also strengthen the interface between the gastrointestinal lumen and epithelium. Mucus is produced by goblet cells in the gastrointestinal tract, provides a barrier between the lumen of the gut and the colonic epithelium, and is therefore an important component of the mucosal immune system. SCFAs, most notably butyrate, promote mucus production [30,31]. Additionally, SCFAs improve intestinal barrier function by promoting tight junction (TJ) formation [32] and by stabilizing hypoxia-inducible factor (HIF), which is implicated in promoting TJ biogenesis and actin cytoskeletal regulation, to enhance epithelial barrier function [3335].

Finally, SCFAs act as signaling molecules, which modulate host immune responses through histone deacetylase (HDAC) inhibition and by acting as substrates for G protein–coupled receptors (GPCRs). For example, in peripheral blood mononuclear cells, SCFAs are HDAC inhibitors that modulate immunity by suppressing TNF-α production in an NF-κB–dependent fashion [36,37]. SCFAs are ligands for GPCRs and can influence a range of phenotypes from hormone production to leukocyte development to maintain gut homeostasis [3840]. There are 3 known intestinal GPCRs that utilize SCFAs as ligands: GPR41 (FFAR3), GPR43 (FFAR2), and GPR109a. Butyrate, propionate, and acetate are agonists for GPR41 and GPR43, while only butyrate activates GPR109a [41,42]. Activation of GPR43 by butyrate is associated with proliferation of anti-inflammatory Treg cells, while SCFA inhibition of HDAC of Foxp3 is associated with naïve CD4+ T cells differentiating into pTreg cells [4345]. Together, these findings demonstrate that SCFAs, via GPCR engagement, suppress intestinal inflammation to help maintain gut homeostasis

The varied roles of SCFAs in host metabolism, as positive effectors of gut barrier integrity, and as immune regulators support our understanding that the gut microbiome is a key player in maintaining host homeostasis. These observations also indicate the potentially drastic effects that a SCFA-deficient microbiome can have on the host. By extension, the diverse impacts that SCFAs have on host pathways suggest that these metabolites may play comparably important and diverse roles in dictating the biology of gut-resident microbes.

Short chain fatty acids negatively impact C. difficile growth

Diverse metabolites in the gastrointestinal tract impact C. difficile fitness and pathogenesis. For example, bile acids [46,47], metals [48], amino acids [49,50], and sugars [5153] have a range of effects on C. difficile in vitro and in animal models of infection. SCFAs are part of the complex metabolic milieu of the gut, and a growing body of literature demonstrates that SCFAs play varied and important roles in the fitness and virulence of phylogenetically diverse bacterial pathogens, including C. difficile. First, SCFAs have a direct inhibitory effect on the growth of C. difficile: Acetate, butyrate, propionate, and valerate all reduce the growth rate of C difficile in culture [5456]. Consistent with the in vitro observations that C. difficile growth is inhibited by SCFAs, in vivo observations illustrate an inverse correlation between SCFAs and C. difficile pathogenesis [55,56]. The first in vivo connection between SCFAs and C. difficile pathogenesis was made using a hamster model of infection, where it was observed that older animals (which have elevated levels of SCFAs in their guts relative to younger animals) were less susceptible to C. difficile infection (CDI) [57]. Indeed, antibiotic exposure, which reduces levels of SCFAs in the gut, is the major risk factor for CDI in humans and is routinely leveraged in animal models to reduce colonization resistance against CDI [51,55,58,59].

Antibiotics are commonly used to treat for individuals with CDI. However, our increasing awareness of the off-target effects that antibiotics have on beneficial microbes has prompted the design and implementation of alternative strategies for CDI. For example, antibiotics such as vancomycin are used to reduce C. difficile burdens but are often ineffective in resolving recurrent CDI [60], likely because they perpetuate dysbiosis. Furthermore, evidence from murine models of CDI demonstrate that antibiotics can induce a “super-shedder” state in mice, perhaps increasing the transmissibility to new hosts [61], which may be a contributor to the prevalence of CDIs in nosocomial settings. One highly effective alternative is fecal microbiota transplant (FMT), which mitigates recurrent CDI in humans and in animal models, presumably by restoring a “healthy” microbiome [62,63]. FMTs also lead to elevated levels of SCFAs in the recipient, which correlate with reduced C. difficile burdens and CDI disease symptoms [62]. Another possible alternative to antibiotics for the treatment of CDI is high-fiber dietary intervention, which has shown efficacy in a murine model of CDI [55]. Because dietary fiber impacts the levels of SCFAs and other metabolites in the gastrointestinal tracts of humans and other animals, it represents a promising avenue for restoring healthy microbiome community function and reducing the fitness of C. difficile in the gut.

C. difficile toxins are regulated by metabolic cues, including short chain fatty acids

The pathogenic lifestyle of C. difficile relies on 2 cytotoxins, TcdA and TcdB (also referred to as “toxins” in this Review), which inactivate Rho GTPases within host colonic epithelial cells, leading the disruption of actin cytoskeletons and epithelial barrier integrity, which ultimately cause epithelial damage and inflammation [64]. Despite the strong association between SCFA levels in the gut and reduced fitness of C. difficile, previous studies demonstrated that C. difficile toxin production is elevated in response to SCFAs. Specifically, the addition of acetate, propionate, and butyrate to cultures of C. difficile stimulates toxin production [55,65]. In addition, metabolic conditions, which lead to elevated intracellular concentrations of butyrate in C. difficile, are correlated with increased toxin production. For example, intracellular butyrate accumulates and toxins are produced by C. difficile when cysteine and mixtures of other amino acids are depleted in culture, suggesting that C. difficile senses and responds to elevated SCFAs and amino acid starvation by increasing inflammation in the gut [65]. Consistent with these observations, high-fiber dietary intervention in mice (which elevates SCFA production by the microbiome) leads to a transient increase in the production of toxins by C. difficile on a per-cell basis. Importantly, however, the sustained consumption of fiber by the host leads to exclusion of C. difficile from the community and overall reduction of C. difficile and its toxins (i.e., an increase in toxin abundance in bulk stool was not observed) [55].

C. difficile toxin production is controlled by a complex regulatory network. TcdA and TcdB are both encoded in a pathogenicity locus along with several regulatory elements (TcdR and TcdC; [64]). TcdR is a member of the Group V sigma factors and is a positive regulator of toxin expression. TcdR is negatively regulated by TcdC, a dimeric membrane-bound anti-sigma factor. TcdR is responsive to a variety of environmental conditions including temperature, cell redox state, and the abundance of specific intracellular metabolites [6670].

Intracellular metabolites further impact the virulence program of C. difficile via 4 transcription factors: CodY, Rex, CcpA, and PrdR (reviewed in [71]). These transcription factors sense the abundance of key metabolic inputs like glucose (CcpA) and amino acids (CodY and PrdR) or intracellular redox balance (Rex). Importantly, when each of these regulators is activated, they inhibit tcdAB transcription [68,70,72,73]. Although Rex is the only regulator that directly senses NAD/NADH ratios [74], all of these regulators are connected to the maintenance of the NAD/NADH pool via various metabolic steps. For example, one of the major pathways that C. difficile uses to regenerate NAD is the production of butyrate (Fig 2), and both CcpA or CodY directly regulate this metabolism. Specifically, when CcpA or CodY are activated, they inhibit expression of genes involved in the final 2 steps of butyrate production from acetyl-CoA [67,73]. In contrast to butyrate, butanol is an inhibitor of toxin production [65]. CodY, CcpA, and Rex all alter transcription of AdhE, the enzyme responsible for butanol production [67,70,73]. Taken together, these observations link the butyrate/butanol metabolic branch point to the production of toxins by C. difficile—indicating a positive association between butyrate production by C. difficile and by the microbiome and C. difficile pathogenesis.

thumbnail
Fig 2. Overview of C. difficile metabolism with a focus on SCFA synthesis.

Glycolysis, Stickland fermentation, TCA cycle, Wood–Ljungdahl pathway, and SCFA synthesis are underlined and presented in a simplified version for clarity; adapted from [77]. Selected precursors, intermediates, and end products of these pathways are labeled with the major precursors and intermediates relevant for SCFA-centric metabolism in C. difficile. Pathways resulting in net generation of NADH (red), NAD (blue), or ATP (green) are indicated. SCFA, short chain fatty acid; TCA, tricarboxylic acid.

https://doi.org/10.1371/journal.ppat.1009959.g002

The conflicting observations that SCFAs impair C. difficile growth but also increase toxin production create a conundrum on how SCFAs affect C. difficile pathogenesis. Perhaps exogenous SCFAs impair metabolic functioning of C. difficile. Given that exogenous SCFAs are a major product of a healthy microbiome, C. difficile toxin production may be a response to create more inflammation and reduce the fitness of competitors. If so, what are the metabolic pathways that are impacted in C. difficile in the presence of exogenous SCFAs? Does C. difficile respond similarly to acetate, propionate, and butyrate or are the responses distinct? By extension, how are these responses linked to toxin production? Finally, why do ecological disturbances that result in large changes in SCFA concentration, such as FMT or high-fiber diets, resolve CDI instead of exacerbate disease? Would interventions that slightly elevate SCFAs have the opposite effect? In order to answer these questions, a more detailed understanding of the links between C. difficile metabolism and toxin production in the context of complex microbial ecosystems is needed. This will likely be achieved by studies using human samples, animal models of infection, and controlled in vitro systems (e.g., minibioreactor arrays [75], which can be carried out in high throughput while maintaining complex microbial communities).

C. difficile has flexible metabolic capabilities, enabling it to occupy various niches

C. difficile has flexible metabolic capabilities, enabling it to take advantage of different niches during varied dysbiotic states ([59,76]; reviewed in [77] and summarized in Fig 2). C. difficile metabolizes monosaccharides via glycolysis, where it generates pyruvate and acetyl-CoA, both important intermediates in C. difficile metabolism. Another major energy generating pathway employed by C. difficile is Stickland fermentation, which is split into oxidative and reductive branches, which share a single initial step, transamination of an amino acid into a 2-oxo-acid [77]. A variety of amino acids are used in the oxidative portion of the pathway, although leucine and isoleucine are preferred substrates. Both NADH and ATP are generated by the oxidative branch of the pathway, and the end product of the pathway is a carboxylic acid with one carbon converted to carbon dioxide [78]. The reductive branch of the pathway regenerates NAD from NADH and produces electrons to be shuttled out of the cell by the RNF complex. Only some specific amino acids (proline, glycine, phenylalanine, tyrosine, and leucine) can be used in the reductive pathway, which similarly results in carboxylic acid end products. C. difficile has an incomplete TCA cycle that lacks major avenues to produce NADH, and it likely functions to generate metabolic intermediates [79]. Another notable metabolic pathway is the Wood–Ljungdahl pathway in which CO2 is reduced to acetate, regenerating NAD in the process [80]. The variety of energy producing pathways available to C. difficile means that in a dysbiotic gut, C. difficile can heterogeneously express these different metabolic pathways, enhancing its ability to adapt based on available metabolite pools [59,81].

Like other anaerobes, SCFAs are major metabolic end products of C. difficile metabolism. Propionate is produced via succinate from the TCA cycle, and acetate is produced either from acetyl-CoA or from the Wood–Ljungdahl pathway. As discussed above, one of the major pathways that C. difficile uses to regenerate NAD is the production of butyrate. C. difficile produces butyrate from 3 different precursors: succinate, acetyl-CoA, and glutamate. Butyrate formation via acetate can be coupled to the Wood–Ljungdahl pathway, which provides a highly efficient means for NAD regeneration [80]. Whether butyrate is produced from succinate, acetyl-CoA, or glutamate, these pathways converge on crotonyl-CoA, before a further reduction to butyryl-CoA. Butyryl-CoA is either dehydrogenated to butanol, or more commonly butyrate is produced via 2 alternate reactions with the butyrate kinase reaction also producing ATP [77]. This final step may also be important for replenishing the CoA pool of the cell. Together, the SCFA synthesis pathways, and especially butyrate synthesis, comprise important energy generation and NAD recycling pathways in C. difficile (Fig 2).

There are several potential mechanisms by which SCFAs may impair C. difficile growth

C. difficile is one of many enteric pathogens whose fitness is negatively impacted by microbiome-produced SCFAs (reviewed in [14]). Various mechanisms underlying these effects have been demonstrated for other pathogens, raising many possibilities for how SCFAs impact C. difficile. For example, SCFAs alter the physical properties of cells. Specifically, in the presence of SCFAs, cytoplasmic pH and osmotic balance are disrupted in Shigella flexneri, Escherichia coli, and Salmonella Typhimurium [8285]. Excess SCFAs can also interfere with other metabolic pathways or cellular processes, as is the case for acetate-mediated disruption of methionine synthesis or by propionate-mediated inhibition of cell wall and DNA synthesis in E. coli [86,87]. The observations that different SCFAs impact varied cellular processes within E. coli are especially important, as they suggest that similarly diverse cellular processes may be impacted by SCFAs in C. difficile. Furthermore, it is possible that protein activities are directly affected by posttranslational modification (e.g., acetylation, butyrylation, propionylation) [88], which could lead to impaired cellular signaling or enzyme function. Finally, disruption of metabolic processes through product mediated inhibition could be another way in which SCFAs affect C. difficile, especially for butyrate, as its production can be essential for recycling reducing equivalents in the cell (Fig 2). Considering that C. difficile is sensitive to low pH [89] and that SCFAs are increasingly protonated as pH decreases (and can therefore freely translocate membranes), it is possible that different facets of C. difficile biology are impacted by SCFAs at different pH values or that pH may amplify the effects of SCFAs (n.b., the pKa for acetate, propionate, and butyrate are 4.76, 4.88, and 4.82, respectively, which are below the typical pH of the human distal gut). Taken together, further investigation into the impacts of SCFAs on C. difficile, the ways in which these effects impact toxin expression, and the ways in which extracellular pH impacts these responses will enable a better understanding of the varied lifestyles of C. difficile and ways to mitigate its pathogenesis.

Toward a better understanding of how C. difficile balances dysbiosis and inflammation to thrive in the gut

The combined observations that (1) SCFAs are major metabolic end products of anaerobic metabolism in the gastrointestinal tract; (2) SCFAs play important roles in maintaining gut homeostasis; and (3) SCFAs impact C. difficile growth and virulence together highlight a conceptual model to guide future work (Fig 3).

thumbnail
Fig 3. A conceptual model for how C. difficile balances inflammation and dysbiosis to thrive in the gastrointestinal tract.

Key interactions between members of the microbiome, C. difficile, and the host are included. Pointed arrows indicate positive effects, and blunt arrows indicate negative effects, with their weights signifying their relative importance in each of 3 states: (A) A healthy gastrointestinal tract. Gut microbes produce SCFAs as by-products of anaerobic metabolism. These metabolites inhibit C. difficile growth. In the context of a healthy gut, although SCFAs are also signal for C. difficile to produce its toxins, SCFA-mediated growth inhibition dominates, and C. difficile cannot establish a niche. (B) A dysbiotic gastrointestinal tract, where C. difficile thrives. After an ecological disturbance (e.g., antibiotics), C. difficile is able to proliferate within the gut due to an abundance of available nutrients. However, as the microbiome recovers from the disturbance, the concentration of SCFAs increase, which C. difficile uses as a signal to up-regulate its toxins. These toxins disrupt host epithelial cells and lead to the production of immune effectors (including ROS), which suppress the recovery of obligate anaerobes (competitors of C. difficile) in the gut. As a result, despite the presumed negative impacts of ROS on C. difficile, it maintains its inflammation-associated niche by excluding competing microbiome members. (C) A highly inflamed gastrointestinal tract. Here, it is presumed that C. difficile is unable to tolerate the negative effects of the host immune response and its growth is inhibited. We posit that the efficacy of FMT (in humans and animal models) and dietary intervention (in animal models) is due in large part to these interventions shifting the gut ecosystem from the state illustrated in panel B to the state illustrated in panel A. A better understanding of the transitions between all 3 states represented in the figure (e.g., A↔B, B↔C) will enable precision approaches for mitigating CDI in at risk human populations. CDI, C. difficile infection; FMT, fecal microbiota transplant; ROS, reactive oxygen species; SCFA, short chain fatty acid.

https://doi.org/10.1371/journal.ppat.1009959.g003

A healthy gut is a highly competitive environment that resists CDI. In addition to a lack of available nutrients for C. difficile, high levels of SCFAs likely contribute to the inability of C. difficile to colonize or to grow to sufficient densities to impact the host (Fig 3A). However, after an ecological disturbance, such as antibiotic treatment, C. difficile populations expand in the gut. Data from humans and murine models of infection demonstrate that, though C. difficile remains a minority member of the microbiome throughout infection, there is little competition for metabolites (e.g., amino acids, organic acids, and sugars) during this expansion and that C. difficile produces no detectable toxin [49,53,59,9092]. As the microbiome recovers, the concentrations of available nutrients decrease, and the concentrations of SCFAs increase. C. difficile senses and responds to the environmental change by up-regulating its toxins, which elevate inflammation [90]. We hypothesize that SCFAs are a key signal for C. difficile in this transition and that the resulting toxin-mediated inflammation helps to reestablish facets of community composition and function where C. difficile thrives (Fig 3B).

Importantly, this view of C. difficile pathogenesis is informed by recent studies focused on pathogenic Enterobacteriaceae (e.g., Salmonella Typhimurium, E. coli), where pathogen-mediated inflammation reduces the fitness of competing members of the microbiome and enables these pathogens to access privileged nutrients and electron acceptors [9395]. Like the Enterobacteriaceae, C. difficile-induced inflammation liberates nutrients from the host that C. difficile can utilize for growth. For example, in a toxin-dependent fashion, C. difficile uses host-derived collagen breakdown products (which are rich in Stickland substrates like proline, hydroxyproline, alanine, and glycine) and sorbitol (which is used as a carbon source) [52,96]. However, unlike the Enterobacteriaceae, C. difficile is an obligate anaerobe (i.e., it cannot use O2 as a terminal electron acceptor) and is only capable of growing in the presence of relatively low levels of oxygen (<3% O2), likely due to a suite of enzymes involved in detoxifying reactive oxygen species [9799]. So, we hypothesize that if inflammation (C. difficile-mediated or otherwise) elevates both molecular O2 and reactive oxygen species in the gut to high levels, C. difficile fitness would be reduced (Fig 3C).

Taken together, we hypothesize that there is an optimal level of inflammation that allows C. difficile to outcompete members of the gut microbiome, while being able to survive as an anaerobe in an inflamed environment. This optimal level may vary between hosts, depending on variables like host genetics, lifestyle choices, or microbiome composition. Interindividual variation in this optimal level may help to explain variability in humans who can experience a range of C. difficile-related colonization states from asymptomatic carriage to recurrent and fulminant colitis. By learning the specific molecular and genetic mechanisms underlying the response of C. difficile to SCFAs, as well as host- and microbiome-specific factors that influence this response, we expect to be able to develop new concepts and approaches for coping with this pathogen. For example, though dietary fiber intervention has shown promise in reducing burdens of C. difficile in a mouse model of CDI, the path toward translating these findings to humans is unclear. Are there fiber types that clear CDI in humans while others do not? Are the effective fiber types generalizable to all humans or do dietary interventions need to be personalized based on host genetics or microbiome composition? Could a mix of fiber types or synbiotics (fiber coadministered with specific microbes that consume it) increase generalizability of the intervention? Will patients, especially those who consume low-fiber “Western” diets be able to tolerate fiber treatments or will they require an acclimation period?

Alternatively, it is possible that dietary intervention and other SCFA-focused investigations may simply be a tool to toggle the metabolic landscape of the gut and the fitness of C. difficile in the laboratory. In this case, by using these powerful tools, along with systems biology approaches, bacterial genetics, and gnotobiotic animal models, a foundation will be built for uncovering specific C. difficile effectors, microbe–microbe, or microbe–host interactions that can be drugged with novel compounds, next-generation probiotics, or other targeted approaches. Regardless, continued focus on the ways in which C. difficile responds to SCFAs will yield promising conceptual and practical advances on how we understand and treat this vexing bacterial pathogen.

References

  1. 1. Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome. 2019;7(1):91. Epub 2019/06/15. pmid:31196177; PubMed Central PMCID: PMC6567490.
  2. 2. Van Treuren W, Dodd D. Microbial Contribution to the Human Metabolome: Implications for Health and Disease. Annu Rev Pathol. 2020;15:345–69. Epub 2019/10/18. pmid:31622559; PubMed Central PMCID: PMC7678725.
  3. 3. Stickland LH. Studies in the metabolism of the strict anaerobes (genus Clostridium): The chemical reactions by which Cl. sporogenes obtains its energy. Biochem J. 1934;28(5):1746–59. Epub 1934/01/01. pmid:16745572; PubMed Central PMCID: PMC1253397.
  4. 4. Morales P, Fujio S, Navarrete P, Ugalde JA, Magne F, Carrasco-Pozo C, et al. Impact of Dietary Lipids on Colonic Function and Microbiota: An Experimental Approach Involving Orlistat-Induced Fat Malabsorption in Human Volunteers. Clin Transl Gastroenterol. 2016;7(4):e161. Epub 2016/04/08. pmid:27054579; PubMed Central PMCID: PMC4855161.
  5. 5. Bos C, Juillet B, Fouillet H, Turlan L, Daré S, Luengo C, et al. Postprandial metabolic utilization of wheat protein in humans. Am J Clin Nutr. 2005;81(1):87–94. Epub 2005/01/11. pmid:15640465.
  6. 6. Sonnenburg ED, Sonnenburg JL. Starving our microbial self: the deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab. 2014;20(5):779–86. Epub 2014/08/27. pmid:25156449; PubMed Central PMCID: PMC4896489.
  7. 7. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG, Salazar N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front Microbiol. 2016;7:185. Epub 2016/03/01. pmid:26925050; PubMed Central PMCID: PMC4756104.
  8. 8. Louis P, Flint HJ. Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol. 2017;19(1):29–41. Epub 2016/12/09. pmid:27928878.
  9. 9. Bornstein BT, Barker HA. The energy metabolism of Clostridium kluyveri and the synthesis of fatty acids. J Biol Chem. 1948;172(2):659–69. Epub 1948/02/01. pmid:18901185.
  10. 10. Belenguer A, Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, et al. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol. 2006;72(5):3593–9. Epub 2006/05/05. pmid:16672507; PubMed Central PMCID: PMC1472403.
  11. 11. Morrison DJ, Mackay WG, Edwards CA, Preston T, Dodson B, Weaver LT. Butyrate production from oligofructose fermentation by the human faecal flora: what is the contribution of extracellular acetate and lactate? Br J Nutr. 2006;96(3):570–7. Epub 2006/08/24. pmid:16925864.
  12. 12. Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc. 2003;62(1):67–72. Epub 2003/05/13. pmid:12740060.
  13. 13. Vital M, Howe AC, Tiedje JM. Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data. mBio. 2014;5(2):e00889. Epub 2014/04/24. pmid:24757212; PubMed Central PMCID: PMC3994512.
  14. 14. Sun Y, O’Riordan MX. Regulation of bacterial pathogenesis by intestinal short-chain Fatty acids. Adv Appl Microbiol. 2013;85:93–118. Epub 2013/08/15. pmid:23942149; PubMed Central PMCID: PMC4029053.
  15. 15. Miller TL, Wolin MJ. Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora. Appl Environ Microbiol. 1996;62(5):1589–92. Epub 1996/05/01. pmid:8633856; PubMed Central PMCID: PMC167932.
  16. 16. Cook SI, Sellin JH. Review article: short chain fatty acids in health and disease. Aliment Pharmacol Ther. 1998;12(6):499–507. Epub 1998/07/25. pmid:9678808.
  17. 17. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. Epub 2013/12/18. pmid:24336217; PubMed Central PMCID: PMC3957428.
  18. 18. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28(10):1221–7. Epub 1987/10/01. pmid:3678950; PubMed Central PMCID: PMC1433442.
  19. 19. Nagpal R, Wang S, Solberg Woods LC, Seshie O, Chung ST, Shively CA, et al. Comparative Microbiome Signatures and Short-Chain Fatty Acids in Mouse, Rat, Non-human Primate, and Human Feces. Front Microbiol. 2018;9:2897. Epub 2018/12/18. pmid:30555441; PubMed Central PMCID: PMC6283898.
  20. 20. Bloemen JG, Venema K, van de Poll MC, Olde Damink SW, Buurman WA, Dejong CH. Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin Nutr. 2009;28(6):657–61. Epub 2009/06/16. pmid:19523724.
  21. 21. Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13(5):517–26. Epub 2011/05/03. pmid:21531334; PubMed Central PMCID: PMC3099420.
  22. 22. Litvak Y, Byndloss MX, Bäumler AJ. Colonocyte metabolism shapes the gut microbiota. Science (New York, NY). 2018;362(6418). Epub 2018/12/01. pmid:30498100; PubMed Central PMCID: PMC6296223.
  23. 23. Albenberg L, Esipova TV, Judge CP, Bittinger K, Chen J, Laughlin A, et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology. 2014;147(5):1055–63.e8. Epub 2014/07/22. pmid:25046162; PubMed Central PMCID: PMC4252572.
  24. 24. Friedman ES, Bittinger K, Esipova TV, Hou L, Chau L, Jiang J, et al. Microbes vs. chemistry in the origin of the anaerobic gut lumen. Proc Natl Acad Sci U S A. 2018;115(16):4170–5. Epub 2018/04/04. pmid:29610310; PubMed Central PMCID: PMC5910840.
  25. 25. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200. Epub 2016/03/11. pmid:26963409; PubMed Central PMCID: PMC4939913.
  26. 26. Boets E, Gomand SV, Deroover L, Preston T, Vermeulen K, De Preter V, et al. Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: a stable isotope study. J Physiol. 2017;595(2):541–55. Epub 2016/08/12. pmid:27510655; PubMed Central PMCID: PMC5233652.
  27. 27. den Besten G, Lange K, Havinga R, van Dijk TH, Gerding A, van Eunen K, et al. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am J Physiol Gastrointest Liver Physiol. 2013;305(12):G900–10. Epub 2013/10/19. pmid:24136789.
  28. 28. Knowles SE, Jarrett IG, Filsell OH, Ballard FJ. Production and utilization of acetate in mammals. Biochem J. 1974;142(2):401–11. Epub 1974/08/01. pmid:4441381; PubMed Central PMCID: PMC1168292.
  29. 29. Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, et al. Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome. 2018;6(1):55. Epub 2018/03/23. pmid:29562936; PubMed Central PMCID: PMC5863458
  30. 30. Shimotoyodome A, Meguro S, Hase T, Tokimitsu I, Sakata T. Short chain fatty acids but not lactate or succinate stimulate mucus release in the rat colon. Comp Biochem Physiol A Mol Integr Physiol. 2000;125(4):525–31. Epub 2000/06/07. pmid:10840229.
  31. 31. Hatayama H, Iwashita J, Kuwajima A, Abe T. The short chain fatty acid, butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, LS174T. Biochem Biophys Res Commun. 2007;356(3):599–603. Epub 2007/03/22. pmid:17374366.
  32. 32. Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr. 2009;139(9):1619–25. Epub 2009/07/25. pmid:19625695; PubMed Central PMCID: PMC2728689.
  33. 33. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe. 2015;17(5):662–71. Epub 2015/04/14. pmid:25865369; PubMed Central PMCID: PMC4433427.
  34. 34. Fachi JL, Felipe JS, Pral LP, da Silva BK, Correa RO, de Andrade MCP, et al. Butyrate Protects Mice from Clostridium difficile-Induced Colitis through an HIF-1-Dependent Mechanism. Cell Rep. 2019;27(3):750–61.e7. Epub 2019/04/18. pmid:30995474.
  35. 35. Dowdell AS, Cartwright IM, Goldberg MS, Kostelecky R, Ross T, Welch N, et al. The HIF target ATG9A is essential for epithelial barrier function and tight junction biogenesis. Mol Biol Cell. 2020;31(20):2249–58. Epub 2020/07/30. pmid:32726170; PubMed Central PMCID: PMC7550696.
  36. 36. Place RF, Noonan EJ, Giardina C. HDAC inhibition prevents NF-kappa B activation by suppressing proteasome activity: down-regulation of proteasome subunit expression stabilizes I kappa B alpha. Biochem Pharmacol. 2005;70(3):394–406. Epub 2005/06/14. pmid:15950952.
  37. 37. Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda Y, et al. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res. 2008;28(5):321–8. Epub 2008/12/17. pmid:19083427.
  38. 38. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, et al. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond). 2015;39(3):424–9. Epub 2014/08/12. pmid:25109781; PubMed Central PMCID: PMC4356745.
  39. 39. Nilsson NE, Kotarsky K, Owman C, Olde B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem Biophys Res Commun. 2003;303(4):1047–52. Epub 2003/04/10. pmid:12684041.
  40. 40. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology. 2013;145(2):396–406.e1–10. Epub 2013/05/15. pmid:23665276.
  41. 41. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278(28):25481–9. Epub 2003/04/25. pmid:12711604.
  42. 42. Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD, et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 2009;69(7):2826–32. Epub 2009/03/12. pmid:19276343; PubMed Central PMCID: PMC3747834.
  43. 43. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science (New York, NY). 2013;341(6145):569–73. Epub 2013/07/06. pmid:23828891; PubMed Central PMCID: PMC3807819.
  44. 44. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5. Epub 2013/11/15. pmid:24226773; PubMed Central PMCID: PMC3869884.
  45. 45. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50. Epub 2013/11/15. pmid:24226770.
  46. 46. Winston JA, Theriot CM. Impact of microbial derived secondary bile acids on colonization resistance against Clostridium difficile in the gastrointestinal tract. Anaerobe. 2016;41:44–50. Epub 2016/05/11. pmid:27163871; PubMed Central PMCID: PMC5050083.
  47. 47. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2014. Epub 2014/10/23. pmid:25337874.
  48. 48. Zackular JP, Moore JL, Jordan AT, Juttukonda LJ, Noto MJ, Nicholson MR, et al. Dietary zinc alters the microbiota and decreases resistance to Clostridium difficile infection. Nat Med. 2016;22(11):1330–4. Epub 2016/11/01. pmid:27668938; PubMed Central PMCID: PMC5101143.
  49. 49. Battaglioli EJ, Hale VL, Chen J, Jeraldo P, Ruiz-Mojica C, Schmidt BA, et al. Clostridioides difficile uses amino acids associated with gut microbial dysbiosis in a subset of patients with diarrhea. Sci Transl Med. 2018;10(464). Epub 2018/10/26. pmid:30355801; PubMed Central PMCID: PMC6537101.
  50. 50. Leslie JL, Jenior ML, Vendrov KC, Standke AK, Barron MR, O’Brien TJ, et al. Protection from Lethal Clostridioides difficile Infection via Intraspecies Competition for Cogerminant. mBio. 2021;12(2). Epub 2021/04/01. pmid:33785619; PubMed Central PMCID: PMC8092246.
  51. 51. Collins J, Robinson C, Danhof H, Knetsch CW, van Leeuwen HC, Lawley TD, et al. Dietary trehalose enhances virulence of epidemic Clostridium difficile. Nature. 2018;553(7688):291–4. Epub 2018/01/09. pmid:29310122; PubMed Central PMCID: PMC5984069.
  52. 52. Pruss KM, Sonnenburg JL. C. difficile exploits a host metabolite produced during toxin-mediated disease. Nature. 2021;593(7858):261–5. Epub 2021/04/30. pmid:33911281.
  53. 53. Ng KM, Ferreyra JA, Higginbottom SK, Lynch JB, Kashyap PC, Gopinath S, et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature. 2013;502(7469):96–9. Epub 2013/09/03. pmid:23995682; PubMed Central PMCID: PMC3825626.
  54. 54. Kondepudi KK, Ambalam P, Nilsson I, Wadstrom T, Ljungh A. Prebiotic-non-digestible oligosaccharides preference of probiotic bifidobacteria and antimicrobial activity against Clostridium difficile. Anaerobe. 2012;18(5):489–97. Epub 2012/09/04. pmid:22940065.
  55. 55. Hryckowian AJ, Van Treuren W, Smits SA, Davis NM, Gardner JO, Bouley DM, et al. Microbiota-accessible carbohydrates suppress Clostridium difficile infection in a murine model. Nat Microbiol. 2018;3(6):662–9. Epub 2018/04/25. pmid:29686297.
  56. 56. McDonald JAK, Mullish BH, Pechlivanis A, Liu Z, Brignardello J, Kao D, et al. Inhibiting Growth of Clostridioides difficile by Restoring Valerate, Produced by the Intestinal Microbiota. Gastroenterology. 2018;155(5):1495–507.e15. Epub 2018/07/22. pmid:30025704; PubMed Central PMCID: PMC6347096.
  57. 57. Rolfe RD. Role of volatile fatty acids in colonization resistance to Clostridium difficile. Infect Immun. 1984;45(1):185–91. Epub 1984/07/01. pmid:6735467; PubMed Central PMCID: PMC263298.
  58. 58. Theriot CM, Koenigsknecht MJ, Carlson PE Jr., Hatton GE, Nelson AM, Li B, et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat Commun. 2014;5:3114. Epub 2014/01/22. pmid:24445449; PubMed Central PMCID: PMC3950275.
  59. 59. Jenior ML, Leslie JL, Young VB, Schloss PD. Clostridium difficile Colonizes Alternative Nutrient Niches during Infection across Distinct Murine Gut Microbiomes. mSystems. 2017;2(4). Epub 2017/08/02. pmid:28761936; PubMed Central PMCID: PMC5527303.
  60. 60. Kelly CP, LaMont JT. Clostridium difficile—more difficult than ever. N Engl J Med. 2008;359(18):1932–40. Epub 2008/10/31. pmid:18971494.
  61. 61. Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA, Croucher N, 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(9):3661–9. Epub 2009/07/01. pmid:19564382; PubMed Central PMCID: PMC2737984.
  62. 62. Seekatz AM, Theriot CM, Rao K, Chang YM, Freeman AE, Kao JY, et al. Restoration of short chain fatty acid and bile acid metabolism following fecal microbiota transplantation in patients with recurrent Clostridium difficile infection. Anaerobe. 2018;53:64–73. Epub 2018/04/15. pmid:29654837; PubMed Central PMCID: PMC6185828.
  63. 63. van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368(5):407–15. Epub 2013/01/18. pmid:23323867.
  64. 64. Bouillaut L, Dubois T, Sonenshein AL, Dupuy B. Integration of metabolism and virulence in Clostridium difficile. Res Microbiol. 2015;166(4):375–83. Epub 2014/12/03. pmid:25445566; PubMed Central PMCID: PMC4398617.
  65. 65. Karlsson S, Lindberg A, Norin E, Burman LG, Akerlund T. Toxins, butyric acid, and other short-chain fatty acids are coordinately expressed and down-regulated by cysteine in Clostridium difficile. Infect Immun. 2000;68(10):5881–8. Epub 2000/09/19. pmid:10992498; PubMed Central PMCID: PMC101550.
  66. 66. Karlsson S, Dupuy B, Mukherjee K, Norin E, Burman LG, Akerlund T. Expression of Clostridium difficile toxins A and B and their sigma factor TcdD is controlled by temperature. Infect Immun. 2003;71(4):1784–93. Epub 2003/03/26. pmid:12654792; PubMed Central PMCID: PMC152060.
  67. 67. Dineen SS, McBride SM, Sonenshein AL. Integration of metabolism and virulence by Clostridium difficile CodY. J Bacteriol. 2010;192(20):5350–62. Epub 2010/08/17. pmid:20709897; PubMed Central PMCID: PMC2950512.
  68. 68. Bouillaut L, Self WT, Sonenshein AL. Proline-dependent regulation of Clostridium difficile Stickland metabolism. J Bacteriol. 2013;195(4):844–54. Epub 2012/12/12. pmid:23222730; PubMed Central PMCID: PMC3562115.
  69. 69. Antunes A, Camiade E, Monot M, Courtois E, Barbut F, Sernova NV, et al. Global transcriptional control by glucose and carbon regulator CcpA in Clostridium difficile. Nucleic Acids Res. 2012;40(21):10701–18. Epub 2012/09/20. pmid:22989714; PubMed Central PMCID: PMC3510511.
  70. 70. Ravcheev DA, Li X, Latif H, Zengler K, Leyn SA, Korostelev YD, et al. Transcriptional regulation of central carbon and energy metabolism in bacteria by redox-responsive repressor Rex. J Bacteriol. 2012;194(5):1145–57. Epub 2012/01/03. pmid:22210771; PubMed Central PMCID: PMC3294762.
  71. 71. Martin-Verstraete I, Peltier J, Dupuy B. The Regulatory Networks That Control Clostridium difficile Toxin Synthesis. Toxins. 2016;8(5). Epub 2016/05/18. pmid:27187475; PubMed Central PMCID: PMC4885068.
  72. 72. Dineen SS, Villapakkam AC, Nordman JT, Sonenshein AL. Repression of Clostridium difficile toxin gene expression by CodY. Mol Microbiol. 2007;66(1):206–19. Epub 2007/08/30. pmid:17725558.
  73. 73. Antunes A, Martin-Verstraete I, Dupuy B. CcpA-mediated repression of Clostridium difficile toxin gene expression. Mol Microbiol. 2011;79(4):882–99. Epub 2011/02/09. pmid:21299645.
  74. 74. Gyan S, Shiohira Y, Sato I, Takeuchi M, Sato T. Regulatory loop between redox sensing of the NADH/NAD(+) ratio by Rex (YdiH) and oxidation of NADH by NADH dehydrogenase Ndh in Bacillus subtilis. J Bacteriol. 2006;188(20):7062–71. Epub 2006/10/04. pmid:17015645; PubMed Central PMCID: PMC1636230.
  75. 75. Auchtung JM, Robinson CD, Britton RA. Cultivation of stable, reproducible microbial communities from different fecal donors using minibioreactor arrays (MBRAs). Microbiome. 2015;3:42. Epub 2015/10/01. pmid:26419531; PubMed Central PMCID: PMC4588258.
  76. 76. Riedel T, Wetzel D, Hofmann JD, Plorin S, Dannheim H, Berges M, et al. High metabolic versatility of different toxigenic and non-toxigenic Clostridioides difficile isolates. Int J Med Microbiol. 2017;307(6):311–20. Epub 2017/06/18. pmid:28619474.
  77. 77. Neumann-Schaal M, Jahn D, Schmidt-Hohagen K. Metabolism the Difficile Way: The Key to the Success of the Pathogen Clostridioides difficile. Front Microbiol. 2019;10:219. Epub 2019/03/05. pmid:30828322; PubMed Central PMCID: PMC6384274.
  78. 78. Neumann-Schaal M, Hofmann JD, Will SE, Schomburg D. Time-resolved amino acid uptake of Clostridium difficile 630Δerm and concomitant fermentation product and toxin formation. BMC Microbiol. 2015;15:281. Epub 2015/12/19. pmid:26680234; PubMed Central PMCID: PMC4683695.
  79. 79. Dannheim H, Will SE, Schomburg D, Neumann-Schaal M. Clostridioides difficile 630Δerm in silico and in vivo—quantitative growth and extensive polysaccharide secretion. FEBS Open Bio. 2017;7(4):602–15. Epub 2017/04/12. pmid:28396843; PubMed Central PMCID: PMC5377389.
  80. 80. Gencic S, Grahame DA. Diverse Energy-Conserving Pathways in Clostridium difficile: Growth in the Absence of Amino Acid Stickland Acceptors and the Role of the Wood-Ljungdahl Pathway. J Bacteriol. 2020;202(20). Epub 2020/09/25. pmid:32967909; PubMed Central PMCID: PMC7515248.
  81. 81. Fletcher JR, Erwin S, Lanzas C, Theriot CM. Shifts in the Gut Metabolome and Clostridium difficile Transcriptome throughout Colonization and Infection in a Mouse Model. mSphere. 2018;3(2). Epub 2018/03/31. pmid:29600278; PubMed Central PMCID: PMC5874438.
  82. 82. Axe DD, Bailey JE. Transport of lactate and acetate through the energized cytoplasmic membrane of Escherichia coli. Biotechnol Bioeng. 1995;47(1):8–19. Epub 1995/07/05. pmid:18623362.
  83. 83. Roe AJ, McLaggan D, Davidson I, O’Byrne C, Booth IR. Perturbation of anion balance during inhibition of growth of Escherichia coli by weak acids. J Bacteriol. 1998;180(4):767–72. Epub 1998/02/24. PubMed Central PMCID: PMC106953. pmid:9473028
  84. 84. Diez-Gonzalez F, Russell JB. The ability of Escherichia coli O157:H7 to decrease its intracellular pH and resist the toxicity of acetic acid. Microbiology (Reading). 1997;143 (Pt 4):1175–80. Epub 1997/04/01. pmid:9141680.
  85. 85. Jacobson A, Lam L, Rajendram M, Tamburini F, Honeycutt J, Pham T, et al. A Gut Commensal-Produced Metabolite Mediates Colonization Resistance to Salmonella Infection. Cell Host Microbe. 2018;24(2):296–307.e7. Epub 2018/07/31. pmid:30057174.
  86. 86. Cherrington CA, Hinton M, Chopra I. Effect of short-chain organic acids on macromolecular synthesis in Escherichia coli. J Appl Bacteriol. 1990;68(1):69–74. Epub 1990/01/01. pmid:1690200.
  87. 87. Roe AJ, O’Byrne C, McLaggan D, Booth IR. Inhibition of Escherichia coli growth by acetic acid: a problem with methionine biosynthesis and homocysteine toxicity. Microbiology (Reading). 2002;148(Pt 7):2215–22. Epub 2002/07/09. pmid:12101308.
  88. 88. Chen Y, Sprung R, Tang Y, Ball H, Sangras B, Kim SC, et al. Lysine propionylation and butyrylation are novel post-translational modifications in histones. Mol Cell Proteomics. 2007;6(5):812–9. Epub 2007/02/03. pmid:17267393; PubMed Central PMCID: PMC2911958.
  89. 89. Wetzel D, McBride SM. The Impact of pH on Clostridioides difficile Sporulation and Physiology. Appl Environ Microbiol. 2020;86(4). Epub 2019/12/08. pmid:31811041; PubMed Central PMCID: PMC6997743.
  90. 90. Koenigsknecht MJ, Theriot CM, Bergin IL, Schumacher CA, Schloss PD, Young VB. Dynamics and establishment of Clostridium difficile infection in the murine gastrointestinal tract. Infect Immun. 2015;83(3):934–41. Epub 2014/12/24. pmid:25534943; PubMed Central PMCID: PMC4333439.
  91. 91. Ferreyra JA, Wu KJ, Hryckowian AJ, Bouley DM, Weimer BC, Sonnenburg JL. Gut Microbiota-Produced Succinate Promotes C. difficile Infection after Antibiotic Treatment or Motility Disturbance. Cell Host Microbe. 2014;16(6):770–7. pmid:25498344
  92. 92. Kim J, Cho Y, Seo MR, Bae MH, Kim B, Rho M, et al. Quantitative characterization of Clostridioides difficile population in the gut microbiome of patients with C. difficile infection and their association with clinical factors. Sci Rep. 2020;10(1):17608. Epub 2020/10/21. pmid:33077744; PubMed Central PMCID: PMC7573688.
  93. 93. Rivera-Chavez F, Zhang LF, Faber F, Lopez CA, Byndloss MX, Olsan EE, et al. Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. Cell Host Microbe. 2016;19(4):443–54. Epub 2016/04/15. pmid:27078066; PubMed Central PMCID: PMC4832419.
  94. 94. Faber F, Tran L, Byndloss MX, Lopez CA, Velazquez EM, Kerrinnes T, et al. Host-mediated sugar oxidation promotes post-antibiotic pathogen expansion. Nature. 2016;534(7609):697–9. Epub 2016/06/17. pmid:27309805; PubMed Central PMCID: PMC4939260.
  95. 95. Lopez CA, Rivera-Chávez F, Byndloss MX, Bäumler AJ. The Periplasmic Nitrate Reductase NapABC Supports Luminal Growth of Salmonella enterica Serovar Typhimurium during Colitis. Infect Immun. 2015;83(9):3470–8. Epub 2015/06/24. pmid:26099579; PubMed Central PMCID: PMC4534643.
  96. 96. Fletcher JR, Pike CM, Parsons RJ, Rivera AJ, Foley MH, McLaren MR, et al. Clostridioides difficile exploits toxin-mediated inflammation to alter the host nutritional landscape and exclude competitors from the gut microbiota. Nat Commun. 2021;12(1):462. Epub 2021/01/21. pmid:33469019; PubMed Central PMCID: PMC7815924.
  97. 97. Giordano N, Hastie JL, Carlson PE. Transcriptomic profiling of Clostridium difficile grown under microaerophillic conditions. Pathog Dis. 2018;76(2). Epub 2018/02/02. pmid:29390060.
  98. 98. Giordano N, Hastie JL, Smith AD, Foss ED, Gutierrez-Munoz DF, Carlson PE Jr., Cysteine Desulfurase IscS2 Plays a Role in Oxygen Resistance in Clostridium difficile. Infect Immun. 2018;86(8). Epub 2018/06/06. pmid:29866903; PubMed Central PMCID: PMC6056869.
  99. 99. Kint N, Alves Feliciano C, Martins MC, Morvan C, Fernandes SF, Folgosa F, et al. How the Anaerobic Enteropathogen Clostridioides difficile Tolerates Low O(2) Tensions. mBio. 2020;11(5). Epub 2020/09/10. pmid:32900801; PubMed Central PMCID: PMC7482061.