Skip to main content
  • Research article
  • Open access
  • Published:

Adenosine A2A receptor activation reduces recurrence and mortality from Clostridium difficileinfection in mice following vancomycin treatment

Abstract

Background

Activation of the A2A adenosine receptor (A2AAR) decreases production of inflammatory cytokines, prevents C. difficile toxin A-induced enteritis and, in combination with antibiotics, increases survival from sepsis in mice. We investigated whether A2AAR activation improves and A2AAR deletion worsens outcomes in a murine model of C. difficile (strain VPI10463) infection (CDI).

Methods

C57BL/6 mice were pretreated with an antibiotic cocktail prior to infection and then treated with vancomycin with or without an A2AAR agonist. A2AAR-/- and littermate wild-type (WT) mice were similarly infected, and IFNγ and TNFα were measured at peak of and recovery from infection.

Results

Infected, untreated mice rapidly lost weight, developed diarrhea, and had mortality rates of 50-60%. Infected mice treated with vancomycin had less weight loss and diarrhea during antibiotic treatment but mortality increased to near 100% after discontinuation of antibiotics. Infected mice treated with both vancomycin and an A2AAR agonist, either ATL370 or ATL1222, had minimal weight loss and better long-term survival than mice treated with vancomycin alone. A2AAR KO mice were more susceptible than WT mice to death from CDI. Increases in cecal IFNγ and blood TNFα were pronounced in the absence of A2AARs.

Conclusion

In a murine model of CDI, vancomycin treatment resulted in reduced weight loss and diarrhea during acute infection, but high recurrence and late-onset death, with overall mortality being worse than untreated infected controls. The administration of vancomycin plus an A2AAR agonist reduced inflammation and improved survival rates, suggesting a possible benefit of A2AAR agonists in the management of CDI to prevent recurrent disease.

Peer Review reports

Background

Clostridium difficile infection (CDI) is characterized by intense intestinal and systemic inflammatory reactions, especially in moderate to severe disease. Such microorganism-initiated tissue damage causes de novo production and in situ accumulation of adenosine that signals through four G protein–coupled receptors designated as A1, A2A, A2B, and A3[1]. Activation of the A2A adenosine receptor (A2AAR) produces a constellation of responses that are anti-inflammatory. Pro-inflammatory responses in bone marrow derived cells (BMDC) including platelets [2], monocytes [3], mast cells [4, 5], neutrophils [6–8] and T cells [9–11] are all inhibited by A2AAR activation.

Adenosine is a purine nucleoside that plays an important role in many biochemical processes such as energy transfer. It also acts as a secondary messenger and neurotransmitter [12]. Endogenous adenosine is produced in part by nucleotide degradation with participation of 5′ nucleotidase with or without ectonucleotidases on cell membranes, from the pool of adenosine triphosphate (ATP), adenosine diphosphate (ADP), or adenosine monophosphate (AMP) released through regulatory processes, inflammation, or cellular damage. Adenosine can further be degraded to inosine by adenosine deaminase intra- and/or extra-cellularly. Otherwise, it can be retaken up and converted back to AMP by adenosine kinase. As an endogenous purine nucleotide, adenosine modulates many physiological processes through four adenosine receptor subtypes, A1, A2A, A2B and A3.

Selective activation of the A2AAR with synthetic adenosine analogs has been demonstrated to protect many tissues, including liver, kidney, skin, heart, and spinal cord, from ischemia-reperfusion injury [13–16], to inhibit inflammatory responses in rabbit joint sepsis induced by LPS [17], and to improve mouse survival from sepsis with Escherichia coli [18, 19] or Staphylococcus aureus [18] in combination with antibiotic treatment. Previous studies have suggested that activation of A2AARs with ATL 313, or inhibition of adenosine deaminase prevents Clostridium difficile toxin A-induced enteritis by reducing the production of inflammatory cytokines in mouse or rabbit ileal loop model [20–22]. In the current study, we found that A2AAR activation during antibiotic treatment for CDI lessens disease severity, prevents relapse and increases survival of mice. Deletion of A2AARs worsens outcome of CDI by enhancing the host inflammatory response to infection. The beneficial effects of A2AR activation are probably caused by anti-inflammatory effects of A2AAR activation counteracting the pro-inflammatory effects of C. difficile toxins.

Methods

Animals

Eight-week old male C57BL/6 mice were purchased from the Jackson Laboratory (Bar Harbor, ME 04609). Food and water were provided ad libitum before and during the experiments. A2AAR -/- mice from Jiang-Fan Chen [23] of Boston University were bred to be congenic with C57BL/6 mice. A2AAR -/- mice were age- and sex-matched to wild type controls. Mouse genotyping employed a set of 3 primers (5′-GGGCTCCTCGGTGTACAT-3′, 5′-CCCACAGATCTAGCCTTA-3′, 5′-TGTCACGTCCTGCACGAC-3′) to resolve a 380-bp wild type allele versus a 500-bp knockout allele. Animals were housed in a pathogen-free isolation barrier facility with chip bedding. A previously published infection model was adapted with slight modification [24]. Briefly, all mice were started with a 3-day antibiotic cocktail pretreatment containing 4.5 mg of vancomycin, 4.2 units of colistin, 3.5 mg of gentamicin, and 21.5 mg of metronidazole per kg/day in drinking water 6 days before the infection. Clindamycin (32 mg/kg) was given intraperitoneally to each mouse the day before the infection. Mice were transferred from a pathogen-free room to a BSL-2 room within the vivarium where they were prepared for infection. Infected mice remained in the same cage and were placed in a dedicated sash in the BSL-2 room. In most experiments, 50 mg/kg/day of vancomycin was administered in drinking water starting 24 hours post infection. The vancomycin treatment was routinely terminated on day 4 post infection unless specifically stated. Treatment with an A2AAR agonist (via Alzet pump as described below) was typically started 1 day post-infection (unless specified) and lasted for either 7 or 14 days. Mice that were considered moribund were euthanized by cervical dislocation. Infection and treatment protocol was approved by the University of Virginia Animal Care and Use Committee.

Materials

The A2AAR agonists ATL370 and ATL1222 as well as ALZET mini-osmotic pumps (Model 1007D and 1002) were gifts from Dogwood Pharmaceuticals, Inc (Charlottesville, VA). The chemical structure and molecular weight of ATL370 are shown in Figure 1. The molecular weight of the ATL370 analogue, ATL1222, is 539.25 and its potency at the adenosine receptor subtypes is similar to ATL370. Injectable vancomycin hydrochloride (Hospira, Inc., Lake Forest, IL), Colistimethate (Colistin) (X-GEN Pharmaceuticals, Inc., Big Flats, NY), gentamicin sulfate (Hospira), metronidazole (Flagyl) (Baxter Healthcare Corporation, Deerfield, IL), and cleocin phosphate (clindamycin) (Pharmacia and Upjohn Company, Bridgewater, NJ) were all purchased through the University of Virginia Hospital Pharmacy. C. difficile strain VPI10463 was purchased from American Type Culture Collection (Manassas, VA). Chopped Meat Broth (CM, catalog AS-811) for Clostridium difficile growth was purchased from Anaerobe Systems (Morgan Hill, CA). Bacto Brain Heart Infusion (BHI medium) for C. difficile wash and reconstitution was purchased from BD Medical-Pharmaceutical Systems (Franklin Lakes, NJ). Bouin’s fixative was purchased from Polysciences, Inc. (Warrington, PA). QIAamp DNA Stool Mini Kit and Proteinase K were purchased from Qiagen, Inc. (Valencia, CA). iQ SYBR Green Supermix containing dNTPs, iTaq DNA polymerase, 6mM MgCl2, SYBR Green I, fluorescein, and stabilizers was obtained from Bio-Rad Laboratories (Hercules, CA). All PCR primers were ordered through Integrated DNA Technologies, Inc. (Coralville, IA).

Figure 1
figure 1

Molecular Structure and Weight of A 2A AR Agonist, ATL370.

Radioligand binding assays

The adenosine receptor binding assay methodology has been described previously [8] and was conducted by Dogwood Pharmaceuticals (Charlottesville, VA 22911). In brief, all subtypes of recombinant human and mouse ARs were stably expressed in HEK-293 cells (A2AAR, A2BAR, and A3AR), CHO-K1 cells (A1AR) or transiently expressed by baculoviral infection of Sf9 cells (A2AAR, high-affinity assay). Crude membranes were prepared from these transfected cells. An appropriate radioligand (125I-ABA for A1AR and A3AR, 125I-ZM241385 for A2AAR, 125I-ABOPX for A2B AR, or 125I-APE for A2AAR/high affinity assay) was added, then filtered through glass fiber filters, and counted in a Wallac Wizard 1470 gamma counter (Perkin Elmer, Boston MA). The non-specific binding of radiolabeled ligand was measured in the presence of the non-selective AR agonist, NECA (100 μM). G protein coupled receptors bound agonists with two affinity states (G protein coupled and G protein uncoupled). The coupled high affinity state was the more relevant assay because it reflects the active configuration of the receptor. To produce G protein coupled human A2AARs, Sf9 cells were simultaneously infected with 4 baculoviruses encoding the human A2AAR and G protein αs, β4 and γ2 subunits [25]. High affinity binding was performed using 125I-APE as described above. Competition binding curves were constructed and IC50 values calculated using a 4-parameter logistic fit (PRISM 5.0, GraphPad Software, San Diego, CA). The value of K i for competition for radioligand binding by agonist was calculated using the Cheng-Prusoff equation [26]. K i values were calculated from the mean of triplicate or quadruplicate assays and normalized to the mean value of the inter-assay standard.

Preparation of Clostridium difficileinocula

C. difficile was grown for 20 hours in CMB (37°C), transferred to a fresh tube of CMB and incubated for 5 hours to achieve log-phase growth prior to infection. Bacteria were centrifuged at 10,000 rpm for 2 minutes, washed with BHI medium three times, reconstituted with BHI medium, and quantified by spectrophotometry. An OD reading of 1.0 was calculated to be equivalent to 5×107 cfu C. difficile/ml. The final concentration of C. difficile (cfu/ml) was validated by hemocytometer reading under light microscopy. Unless the dose was specifically stated, 1×104-5 of C. difficile was gavaged to each mouse in the infected groups, and BHI medium only was given to mice in uninfected groups.

Implantation of ALZET osmotic pumps

ALZET mini-osmotic pumps (Model 1007D and 1002) were implanted in mice using a protocol approved by the University of Virginia Animal Care and Use Committee. Briefly, mice were anesthetized with a mixture of ketamine and xylazine and placed on a heating pad. A short (~5 mm) incision was made at the interscapular area after shaving and cleaning the skin with 70% ethanol and iodine. Pumps filled with A2AAR agonists (ATL370 or ATL1222 at either 1 or 10 ng/kg/min) for treated mice or an equivalent amount of the vehicle-3% dimethyl sulfoxide (DMSO) in PBS only, for untreated control mice were inserted subcutaneously. A wound clip was used to close the incision. Mice were allowed to wake up on the heating pad before returning to the BSL2 sash.

Clinical scoring system

During the entire post infection period, daily weights and clinical scores were recorded for each mouse (Table 1). The sum of all parameter scores was considered the final clinical score and ranged from 0 (normal) to 15. Mice judged to be moribund were sacrificed. Mice found dead were assigned a score of 15. Stools were collected daily and stored at -20°C until DNA extraction was performed.

Table 1 Clinical scoring system for mice infected with Clostridium difficile

Extraction of DNA from frozen mouse stools

All stool samples were weighed before DNA extraction for sample normalization. Stool DNA was extracted under the modified protocol provided in the QIAamp DNA Stool Mini Kit. Briefly, frozen stool was added to 400 μL of ASL buffer, homogenized by grinding with a wooden stick, vortexed for 15 seconds before and after heating in a water bath at 82.5°C for 5 minutes, and then centrifuged at 14,000 rpm for 2 minutes. The remaining steps followed the manufacturer’s directions. Extracted stool DNA was stored at -20°C prior to PCR testing.

Quantification of C. difficileshedding by qPCR

C. difficile DNA was analyzed from extracted stool DNAs with iQ SYBR Green Supermix in a 96-well plate performed at CFX96™ Real-Time PCR Detection System (Bio-Rad). Briefly, a PCR master mix was prepared with 23 μL aliquot containing 1 μL each of tcdB forward and reverse primers, 12.5 μL iQ SYBR Green Supermix, and 8.5 μL of H2O purified by Milli-Q Integral Water Purification System (Millipore Corporation, Billerica, MA). 2 μL of each sample was then added to each well filled with PCR master mix aliquot in 96-well. The PCR parameters were sequentially set for 3 stages: 1× cycle for 5 minutes at 94.0°C, 40 × cycle for 30 seconds each from 94.0°C and 55.0°C to72.0°C, 64 × cycle at 62.0°C for 15 seconds, and 1× cycle for hold at 25.0°C. Melt curve data collection and analysis were enabled. Copy numbers of unknown sample were extrapolated from the standard curve that was generated with the extracted DNA prepared from known C. difficile inocula. The sequence of tcdB forward primer was 5′-GGAGAGTCATCCAACTTATATG-3′; the sequence of tcdB reverse primer was 5′-CCACCAATTTCTTTTAATGCAG-3′.

Intestinal histopathology

The middle cross section of cecum and proximal cross section of colon were harvested from moribund mice or surviving mice at the end of the experiment. Tissues were fixed overnight with Bouin’s solution and stored in 70% ethanol until subsequently processed for Hematoxylin & Eosin (HE) staining at University of Virginia Research Histology Core. Slides were examined using a Leica DFC425 digital camera equipped microscope with Leica Application Suite Version 3.6.0.488 imaging software (Leica Microsystems Inc., Buffalo Grove, IL 60089). Intestinal tissues were scored from 0 to 3 (with zero as normal and 3 as the worst pathologic score) under 5 categories: overall architecture, mucosal thickness, submucosal edema, inflammation, and exudates.

ELISA for IFNγ and TNFα

One set of infection experiments with A2AAR -/- (n=8) and their littermate A2AAR +/+ (n=8) mice were set aside for blood and tissue cytokine assays at the peak of infection (day 3) and also at recovery (day 7). Blood samples were collected in heparinized tube by cardiopuncture under sedation. Plasma samples were obtained by centrifuging blood at 10,000 rpm at room temperature for 20 minutes and stored at -80°C until further analysis. Upon euthanasia, cecal samples were harvested and stored at -80°C. ELISA was performed using Thermo Scientific Pierce Mouse IFNγ and TNFα kits (Rockford, IL) with slight modification of the manufacturer’s instruction. Briefly, cecal tissue was homogenized by grinding on dry ice and suspended in diluent reagent (0.2 mg/ml). Each homogenate or plasma sample was incubated with IFNγ antibody (for 1 hour) or TNFα antibody (for 2 hours) in 1:20 final dilution (200 μl) at room temperature. After washing, the sample was incubated with 100 μl of streptavidin-horseradish peroxidase (HRP) for 30 minutes. Subsequently, 100 μl of TMB substrate was added for another 30-minute incubation in the dark. The incubation was terminated with 100 μl of stop solution. Samples were immediately measured at 450 nm and 550 nm in Gen5 1.11.5 version in BioTek Spectrophotometer (Winooski, Vermont). Standard curves were established by plotting the average absorbance obtained for each standard known concentration (pg/ml). Cytokine amount in each sample was extrapolated from the standard curves.

Statistical analysis

Statistical analyses were conducted using GraphPad Prism Version 5.02 software. When mouse was either found dead or sacrificed due to severe distress, its last body weight recording was continuously plotted against the body weights of surviving mice. Differences between groups for the entire experimental period were analyzed by 2-Way ANOVA with Bonferroni post hoc testing. Survival curves were analyzed using Log-rank (Mantel-Cox) or Log-rank test for mortality trend.

Results

A2AAR agonist reduced diarrhea and deaths in C. difficile- infected mice

To confirm the protective effect of A2AAR activation previously seen in ileal loop models [20–22], we used the A2AAR agonist ATL370 to treat wild-type mice infected with C. difficile VPI10463. The binding affinity of ATL 370 to adenosine receptors is shown in Table 2. Two dosing regimens were tested: ATL370 at 1 ng/kg/min via a 14-day (first study) and at 10 ng/kg/min via a 7-day (2nd study) Alzet pumps. Mortality rate from infected controls in the first study was 50%, with deaths occurring at days 4 and 5 (Figure 2A). All mice treated with vancomycin alone had relapsed and succumbed to infection by day 11 (deaths occurred 5 to 11 days after discontinuation of the antibiotic). In contrast, survival increased to 33% in mice treated with both vancomycin and ATL30. From day 1 post infection, infected mice progressively lost weights with a few losing up to almost 20% of their body weight at baseline (Figure 2B). Infected mice treated with vancomycin did not have weight loss until four days after termination of vancomycin treatment. Infected mice treated with both vancomycin and ATL370 at 1 ng/kg/min lost the same amount of weight as the vancomycin only-treated mice although weight loss started one day later. Infected control mice had rapid development of diarrhea and elevated clinical scores (Figure 2C), whereas treatment with vancomycin prevented signs of disease until three days after termination of treatment. Consistent with weight changes, clinical symptoms tended to be lessened in mice receiving combination treatment compared to those treated with vancomycin alone. Clostridial shedding in the stool was elevated in infected mice not given vancomycin during acute infection (data not shown). Upon discontinuation of vancomycin, clostridial shedding increased significantly and remained elevated with or without ATL370 in surviving mice indicating that the agonist does not affect fecal clostridial burden. This experiment suggested a modest benefit of the A2AAR agonist ATL370 at 1 ng/kg/min if given with vancomycin during C. difficile infection. Furthermore, this study suggested that A2AAR activation during infection is detrimental in the absence of antibiotics.

Table 2 Characterization of the A 2A AR agonist, ATL370, by radioligand binding (mean ± SD)
Figure 2
figure 2

Effect of A 2A AR Agonist ATL370 on C. difficile-Infected Mice. Mice were infected with VPI10463 at 105 inoculum and treated or not with vancomycin (50 mg/kg/day for 3 days) with or without ATL370 (1 ng/kg/min). Each group had 6 mice each except for uninfected control (n=4). For statistical analyses of weight change and clinical scores, the latest value from a dead mouse was carried through to the end of observation but not shown in the graphs. A. Survival curve. P=0.0016 by Mantel-Cox. B. Percent weight loss from day 0 (day of infection). *p<0.05 to 0.001 for uninfected (UC) vs. infected (I) controls, †p<0.01 to 0.001 for I vs I with vancomycin (Vanc), ‡p<0.01 to 0.001 for I vs. I with Vanc plus ATL370(1); by Two-way ANOVA with Bonferroni’s correction. C. Clinical scores. *p<0.05 for UC vs. I controls, †p<0.05 for I vs I with Vanc; by Two-way ANOVA with Bonferroni’s correction.

We next tested the effect of ATL370 at a higher dose but shorter duration of treatment, we used 10 ng/kg/min dosing given for 7 days (via Alzet pump). Similar to what was observed earlier, infected mice treated with vancomycin fared well for several days until discontinuation of vancomycin. Addition of ATL370 to vancomycin improved survival by 20%. Infected mice that received ATL370 alone had a 0% survival rate. Weight changes and clinical scores followed the same trend as the first study. Taken together, these data suggested that low dose ATL370 may be as beneficial as higher doses if given with vancomycin and confirmed that A2AAR agonist alone could exacerbate disease during acute infection.

Early or delayed administration of A2AAR agonist improved outcome of C. difficileinfection

To determine whether the timing of A2AAR agonist treatment would alter the response to treatment, we investigated the effect of delaying administration of ATL370 in relation to vancomycin. During the 21-day post infection period (Figure 3), we observed a 50% survival rate in the infected control group while those treated with vancomycin alone had only a 17% survival rate. Again, mortality in the vancomycin-treated mice was evident only after the antibiotic was discontinued suggesting relapse of infection. Treatment with ATL370, regardless of whether it was started at the same time as or 3 days after vancomycin was started, increased survival by 33%, indicating a benefit of A2AAR activation even at a later timepoint after infection or antibiotic treatment. Body weight change, diarrhea and clinical scores followed similar trends as shown in previous experiments.

Figure 3
figure 3

Effect of Timing of A 2A AR Agonist ATL370 Treatment on C.difficile-Infected Mice. Mice were infected with VPI10463 at 105 inoculum and treated or not with vancomycin (50 mg/kg/day for 3 days) with or without ATL370 (1 ng/kg/min started either at day 1 or day 4 after infection). A. Survival Curve, p=0.1427 by Mantel-Cox. B. Clinical Scores; **p<0.01 for I vs. I with Vanc, *p<0.05 to 0.01 for I vs. I with Vanc plus ATL370 by Two-way ANOVA.

Reduced vancomycin exposure further increased survival in the presence of A2AAR agonist in mice infected with C. difficile

Given that vancomycin administration had consistently resulted in delayed and worse mortality compared to untreated infection, we investigated whether decreasing duration of vancomycin treatment would improve survival and, therefore, enhance benefit derived from A2AAR activation. As shown in Figure 4A, short treatment durations (1-2 days) prevented recurrence of infection and significantly improved survival rate from 37.5% in longer treatment durations (3-5 days) to 87.5%. We, then, compared a short course (2-day) versus a long course (5-day) vancomycin treatment in combination with another A2AAR agonist, ATL1222. As shown in Figure 4B, infected mice treated with a 2-day treatment course had better survival rates than mice treated with a 5-day course (50% vs. 25%) of vancomycin. Furthermore, ATL1222 improved survival by 25% when given in addition to a 2-day course of vancomycin or by 50% when compared to a 5-day course of vancomycin alone. Both weights and diarrhea scores were likewise improved with the shorter course of antibiotics plus ATL1222 (Figure C&D). Together, these findings suggest that A2AAR activation enhances the benefit of a shorter course of antibiotic treatment against CDI.

Figure 4
figure 4

Effect of A 2A AR Agonist ATL1222 on Mortality of Mice Infected with C.difficile and Treated with Vancomycin of Short or Long Duration. Mice were infected with VPI10463 at 105 inoculum. Each group had 8 mice except uninfected control (n=4). A. Mice were treated or not with vancomycin (50 mg/kg/day) for 1, 2, 3 or 5 days. P=0.058 by Log-rank (Mantel-Cox) Test. B. Mice were treated with vancomycin for either 2 or 5 days with or without ATL1222 (1ng/kg/min started at day 1 post-infection). P value for Log-rank (Mantel-Cox) test at 0.29. C. Weight Change (%). D. Diarrhea score (mean). Scoring system defined in Table 1.

The absence of A2AAR worsened C. difficileinfection in mice

To confirm the role of A2AAR in CDI, A2AAR knockout (KO) and wild-type littermate mice were infected with VPI10463. As seen in Figure 5A (Exp B), only 50% of infected KO mice survived compared to 80% of infected wild-type mice. Four similar experiments were performed to compare survival rates between infected A2AAR KOs and wild-type mice (Figure 5B). The overall survival rates in infected wild-type were consistently higher than infected KO mice (75% vs. 36.67%) suggesting that the absence of the A2AAR is detrimental during infection.

Figure 5
figure 5

Survival of A 2A AR +/+ or -/- Mice Infected with C.difficile. A. Survival Curve of Exp B showing 80% of the littermate wild-type vs. 50% of knockout mice surviving infection with VPI10463. P value for Log-rank (Mantel-Cox) Test is 0.1592; P value of Logrank test for trend is 0.0666. B. Cumulative mortality from all experiments (Exp A-D). Number of mice in each experiment (number of mice surviving infection).

Regardless of their genetic background, infected animals had significantly higher total cecal histopathology score than uninfected controls (Figure 6; Exp B). Moreover, cecal tissues from infected KO mice had higher histopathology scores than those from wild-type mice. Submucosal edema, mucosal thickness and inflammation were observed more in infected than uninfected cecal tissues (Figure 6A-D). The same parameters were worse in A2AAR gene deleted mice than wild types during infection confirming A2AAR’s protective role against infection-induced epithelial injury consistent with what was previously seen in toxin-induced enteritis [22].

Figure 6
figure 6

Cecal Histopathology of A 2A AR +/+ or -/- Mice Infected with C. difficile. Cecal tissues from surviving mice were obtained on the day 14 post-infection. Histopathological scores were from 3 cecal tissues per group. *p<0.05 and **p<.001 by One-way ANOVA with Bonferroni’s correction. Representative H&E stained tissues from uninfected (A) and infected (B) A2AAR +/+ and uninfected (C) and infected (D) A2AAR -/- mice.

The absence of A2AAR activation altered the inflammatory response during CDI in mice

To determine whether enhanced inflammation contributed to the greater epithelial injury and increased mortality in A2AAR-/- mice, cecal tissues and sera were assayed for IFNγ and TNFα. IFNγ and TNFα levels were increased during infection in wild-type mice at both days 3 and 7 post-infection (Figure 7). Unexpectedly, both cytokines were significantly depressed in A2AAR-/- compared to wild-type mice at day 3 suggesting less inflammation in the absence of A2AAR at the expected peak of infection. At day 7 post-infection, IFNγ and TNFα were significantly more elevated in cecal and blood, respectively, in A2AAR-/- mice than wild-type mice indicating that onset and resolution of inflammation were both delayed in the absence of A2AARs. Furthermore, these results suggest that IFNγ may play a greater role in intestinal tissue injury while TNFα may be more involved in the systemic manifestations of infection.

Figure 7
figure 7

Intestinal Tissue and Blood IFNγ and TNFα in C. difficile infected mice. Cecal and plasma levels of IFNγ and TNFα were measured at days 3 and 7 post-infection in A2AAR -/- and A2AAR +/+ mice. Each bar from cecal levels represents 4 specimens (4 mice) per group. Each bar from blood levels represents 4 specimens per group except bars representing infected mice on day 3 (n=3/group). *P<0.05 or **p<0.01 for infected A2AAR +/+ vs. uninfected A2AAR +/+ in same day, †p<0.05, ‡p<0.01 or #p<0.001 for infected A2AAR -/- vs. infected A2AAR +/+ in same day; Two-way ANOVA with post-hoc Bonferroni’s correction.

Discussion and conclusions

Vancomycin is the drug of choice for severe CDI [27, 28]. However, its use has been associated with clinical recurrence of infection in up to 20% of cases, which has been attributed to antibiotic-induced changes of gut microbiota [29]. In the mouse model of infection, recurrence of disease and late mortality has also been observed to occur in 40-60% of mice treated with vancomycin [24, 30]. Typically, mice treated with vancomycin remain well and only develop disease several days after cessation of the antibiotic. In our study, we demonstrated that the addition of A2AAR agonists during vancomycin treatment decreased recurrence and associated late mortality in mice infected with C. difficile. We then confirmed the role of A2AARs in CDI by showing that the complete absence of A2AAR activation exacerbated disease and alteration of inflammatory cytokine expression.

Intestinal epithelial cells have a very limited capacity for de novo adenosine synthesis [31]. The villous cells can directly use absorbed dietary nucleosides but the cryptal cells depend on blood supply. Under normal adenosine homeostasis, extracellular adenosine concentrations can be lower than 1 μM [32, 33]. In response to cellular damage, adenosine concentrations are quickly elevated 100-fold higher in inflamed intestine due to ATP and adenosine secretion in inflammatory and other cell types. Control of inflammation and injury is thought to be secondary to A2AAR activation in intestinal tissue reperfusion injury [34] or experimental colitis [35]. The A2AAR has been shown to inhibit neutrophil cytotoxic activities such as expression of β2-integrins [36], adhesion to endothelium [37], production of oxygen radicals [38, 39], degranulation [40], and production of TNF-α [41]. Given intense intestinal inflammation, including neutrophilic tissue infiltration, noted in CDI in mouse [42], blockade or absence of A2AARs would, then, be expected to result in aggravation of disease. Indeed, in our study, A2AAR knockout mice had worse colitis (even in mice surviving infection) and more deaths from C. difficile infection compared to wild-type littermates suggesting that endogenous adenosine provides protection against infection through the A2AAR. A2AAR agonists added to 3 days of vancomycin treatment improved survival, clinical scores, and cecum histopathology compared to vancomycin alone when treatment was started at least one day after the animals were infected. This benefit was probably mediated by the anti-inflammatory effects of A2AAR activation counteracting the pro-inflammatory effects of C. difficile toxins.

Previous studies have shown that A2AAR activation with ATL 313 or inhibition of adenosine deaminase attenuated C. difficile toxin A-induced ileitis in mice [20, 21]. Myeloperoxidase activity, TNF-α production, cell death and histopathology were all noted to be reduced in ileal tissues treated with ATL 313. Recently, we showed that the A2AAR agonist, ATL370, decreased toxin A-induced secretion and epithelial injury in rabbit ileum and decreased KC (keratinocyte chemokine) and IL10 levels in mouse cecal tissues [22]. In the current study, we demonstrate that deletion of A2AARs in mice, resulted in delayed but augmented expression of inflammatory cytokines, specifically IFNγ and TNFα during infection suggesting that initial inflammatory response is essential in controlling the disease. Indeed, the administration of ATL 370 alone resulted in greater and quicker mortality in most of the treated C. difficile-infected mice suggesting that A2AAR activation may inhibit the natural and beneficial early immune response initiated by infection. Furthermore, antibiotic treatment is essential to control clostridial burden and development of severe disease. This is consistent with what had been shown in a mouse model of sepsis where another A2AAR agonist, ATL 146e, was shown to reduce mortality in endotoxemia from LPS but not in E. coli septicemia unless antibiotic was also given [19]. It is possible that adenosine is utilized by bacteria to enhance virulence as observed in other pathogens [43, 44]. Interestingly, although infected knockout mice had worse mortality than their infected wild-type littermates, 50% of these mice still survived in the absence of vancomycin treatment. Differences in intestinal flora (transgenic mice were bred in house) as well as other host factors may play a role in susceptibility to severe disease.

Our observation that A2AAR agonist alone (administered at the start of infection) worsened outcomes possibly due to inhibiting the natural immune response early on prompted us to investigate whether delaying A2AAR agonist treatment in addition to treating with vancomycin would improve outcomes. We showed that delaying the start of A2AAR agonist treatment post-infection was as good as starting A2AAR agonist at the same time as vancomycin administration suggesting benefit of A2AAR activation even at a later time during the course of infection and antibiotic treatment. While our study showed that antibiotic treatment is necessary to cure infection, we also demonstrated that overtreating with vancomycin may yield the worst outcomes. Reducing the antibiotic treatment to 2 days improved outcomes and adding A2AAR agonist to this regimen reduced the mortality associated with vancomycin treatment alone by 50%. These observations may have important ramifications if they translate to the clinical setting. If vancomycin (or antibiotic) treatment duration can be reduced by adding an A2AAR agonist, recovery of the gut microbiota may be facilitated and the recurrence of infection after antibiotic therapy may be improved considerably.

Although previous studies have shown that A2AAR activation confers significant protection against C. difficile toxin-induced ileitis and cecitis [20–22], protection against severe disease in the mouse model of infection seems limited. We recently reported that A2BAR inhibition or deletion, even in the absence of an anti-clostridial agent, improved outcome of CDI in mice [45]. During infection, the clostridial bacteria are localized in the lumen and mucosal surface of the intestinal tract. The A2BAR is the predominant adenosine receptor in human intestinal epithelial cells [46] and thus, may have a greater role than A2AAR in mediating local tissue inflammation in response to C. difficile infection. However, A2AAR activity may be critical in controlling inflammatory response from immune cells recruited to the intestinal tissues and/or circulating immune cells during severe disease. More studies are needed to elucidate the interactions between different adenosine receptor subtypes during enteric infection.

In conclusion, in a murine model of CDI, vancomycin treatment resulted to reduced weight loss and diarrhea during acute infection, but was associated with high recurrence and late-onset death, with overall mortality being worse than untreated infected controls. Deletion of A2AARs in mice worsened disease from CDI. The administration of an A2AAR agonist reduced the late mortality associated with vancomycin use, suggesting a possible adjunctive benefit of A2AAR agonists in the management of CDI to prevent recurrent disease and improve survival.

References

  1. Hasko G, Linden J, Cronstein B, Pacher P: Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008, 7: 759-770. 10.1038/nrd2638.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Dionisotti S, Ferrara S, Molta C, Zocchi C, Ongini E: Labeling of A(2A) adenosine receptors in human platelets by use of the new nonxanthine antagonist radioligand [H-3]SCH 58261. J Pharmacol Exp Ther. 1996, 278: 1209-1214.

    CAS  PubMed  Google Scholar 

  3. Link AA, Kino T, Worth JA, McGuire JL, Crane ML, Chrousos GP, Wilder RL, Elenkov IJ: Ligand-activation of the adenosine A2a receptors inhibits IL-12 production by human monocytes. J Immunol. 2000, 164: 436-442.

    Article  CAS  PubMed  Google Scholar 

  4. Lohse MJ, Maurer K, Gensheimer HP, Schwabe U: Dual Actions of Adenosine on Rat Peritoneal Mast-Cells. Naunyn-Schmiedebergs Arch Pharmacol. 1987, 335: 555-560.

    Article  CAS  PubMed  Google Scholar 

  5. Fenster MS, Shepherd RK, Linden J, Duling BR: Activation of adenosine A(2 alpha) receptors inhibits mast cell degranulation and mast cell-dependent vasoconstriction. Microcirculation. 2000, 7: 129-135.

    CAS  PubMed  Google Scholar 

  6. Walker BAM, Rocchini C, Boone RH, Ip S, Jacobson MA: Adenosine A(2a) receptor activation delays apoptosis in human neutrophils. J Immunol. 1997, 158: 2926-2931.

    CAS  PubMed  Google Scholar 

  7. Varani K, Gessi S, Dionisotti S, Ongini E, Borea PA: [H-3]-SCH 58261 labelling of functional A(2A) adenosine receptors in human neutrophil membranes. Br J Pharmacol. 1998, 123: 1723-1731. 10.1038/sj.bjp.0701758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sullivan GW, Linden J, Buster BL, Scheld WM: Neutrophil A2A adenosine receptor inhibits inflammation in a rat model of meningitis: synergy with the type IV phosphodiesterase inhibitor, rolipram. J Infect Dis. 1999, 180: 1550-1560. 10.1086/315084.

    Article  CAS  PubMed  Google Scholar 

  9. Lappas CM, Rieger JM, Linden J: A(2A) adenosine receptor induction inhibits IFN-gamma production in murine CD4(+) T cells. J Immunol. 2005, 174: 1073-1080.

    Article  CAS  PubMed  Google Scholar 

  10. Koshiba M, Rosin DL, Hayashi N, Linden J, Sitkovsky MV: Patterns of A(2A) extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. FASEB J. 1999, 13: A944-

    Google Scholar 

  11. Huang S, Apasov S, Koshiba M, Sitkovsky M: Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood. 1997, 90: 1600-1610.

    CAS  PubMed  Google Scholar 

  12. Fredholm BB: Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 2007, 14: 1315-1323. 10.1038/sj.cdd.4402132.

    Article  CAS  PubMed  Google Scholar 

  13. Day YJ, Li Y, Rieger JM, Ramos SI, Okusa MD, Linden J: A2A adenosine receptors on bone marrow-derived cells protect liver from ischemia-reperfusion injury. J Immunol. 2005, 174: 5040-5046.

    Article  CAS  PubMed  Google Scholar 

  14. Lappas CM, Sullivan GW, Linden J: Adenosine A2A agonists in development for the treatment of inflammation. Expert Opin Investig Drugs. 2005, 14: 797-806. 10.1517/13543784.14.7.797.

    Article  CAS  PubMed  Google Scholar 

  15. Li Y, Oskouian RJ, Day YJ, Rieger JM, Liu L, Kern JA, Linden J: Mouse spinal cord compression injury is reduced by either activation of the adenosine A2A receptor on bone marrow-derived cells or deletion of the A2A receptor on non-bone marrow-derived cells. Neuroscience. 2006, 141: 2029-2039. 10.1016/j.neuroscience.2006.05.014.

    Article  CAS  PubMed  Google Scholar 

  16. Linden J: Adenosine in tissue protection and tissue regeneration. Mol Pharmacol. 2005, 67: 1385-1387. 10.1124/mol.105.011783.

    Article  CAS  PubMed  Google Scholar 

  17. Hogan CJ, Fang GD, Scheld WM, Linden J, Diduch DR: Inhibiting the inflammatory response in joint sepsis. Arthroscopy. 2001, 17: 311-315. 10.1053/jars.2001.21492.

    Article  PubMed  Google Scholar 

  18. Moore CC, Martin EN, Lee GH, Obrig T, Linden J, Scheld WM: An A2A adenosine receptor agonist, ATL313, reduces inflammation and improves survival in murine sepsis models. BMC Infect Dis. 2008, 8: 141-10.1186/1471-2334-8-141.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sullivan GW, Fang G, Linden J, Scheld WM: A2A adenosine receptor activation improves survival in mouse models of endotoxemia and sepsis. J Infect Dis. 2004, 189: 1897-1904. 10.1086/386311.

    Article  CAS  PubMed  Google Scholar 

  20. Cavalcante IC, Castro MV, Barreto AR, Sullivan GW, Vale M, Almeida PR, Linden J, Rieger JM, Cunha FQ, Guerrant RL, Ribeiro RA, Brito GA: Effect of novel A2A adenosine receptor agonist ATL 313 on Clostridium difficile toxin A-induced murine ileal enteritis. Infect Immun. 2006, 74: 2606-2612. 10.1128/IAI.74.5.2606-2612.2006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. de Araujo Junqueira AF, Dias AA, Vale ML, Spilborghs GM, Bossa AS, Lima BB, Carvalho AF, Guerrant RL, Ribeiro RA, Brito GA: Adenosine deaminase inhibition prevents Clostridium difficile toxin A-induced enteritis in mice. Infect Immun. 2011, 79: 653-662. 10.1128/IAI.01159-10.

    Article  PubMed  Google Scholar 

  22. Warren CA, Calabrese GM, Li Y, Pawlowski SW, Figler RA, Rieger J, Ernst PB, Linden J, Guerrant RL: Effects of adenosine A(2)A receptor activation and alanyl-glutamine in Clostridium difficile toxin-induced ileitis in rabbits and cecitis in mice. BMC Infect Dis. 2012, 12: 13-10.1186/1471-2334-12-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen JF, Huang Z, Ma J, Zhu J, Moratalla R, Standaert D, Moskowitz MA, Fink JS, Schwarzschild MA: A(2A) adenosine receptor deficiency attenuates brain injury induced by transient focal ischemia in mice. J Neurosci. 1999, 19: 9192-9200.

    CAS  PubMed  Google Scholar 

  24. Chen X, Katchar K, Goldsmith JD, Nanthakumar N, Cheknis A, Gerding DN, Kelly CP: A mouse model of Clostridium difficile-associated disease. Gastroenterology. 2008, 135: 1984-1992. 10.1053/j.gastro.2008.09.002.

    Article  PubMed  Google Scholar 

  25. Murphree LJ, Marshall MA, Rieger JM, Macdonald TL, Linden J: Human A(2A) adenosine receptors: High affinity agonist binding to receptor-G protein complexes containing G beta 4. Drug Dev Res. 2002, 56: 573-

    Google Scholar 

  26. Cheng Y, Prusoff WH: Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973, 22: 3099-3108. 10.1016/0006-2952(73)90196-2.

    Article  CAS  PubMed  Google Scholar 

  27. Zar FA, Bakkanagari SR, Moorthi KM, Davis MB: A comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis. 2007, 45: 302-307. 10.1086/519265.

    Article  CAS  PubMed  Google Scholar 

  28. Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC, Pepin J, Wilcox MH: Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect Control Hosp Epidemiol. 2010, 31: 431-455. 10.1086/651706.

    Article  PubMed  Google Scholar 

  29. Tannock GW, Munro K, Taylor C, Lawley B, Young W, Byrne B, Emery J, Louie T: A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology. 2010, 156: 3354-3359. 10.1099/mic.0.042010-0.

    Article  CAS  PubMed  Google Scholar 

  30. 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. 10.1128/IAI.01336-10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ye JH, Rajendran VM: Adenosine: an immune modulator of inflammatory bowel diseases. World J Gastroenterol. 2009, 15: 4491-4498. 10.3748/wjg.15.4491.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ramakers BP, Pickkers P, Deussen A, Rongen GA, van den Broek P, van der Hoeven JG, Smits P, Riksen NP: Measurement of the endogenous adenosine concentration in humans in vivo: methodological considerations. Curr Drug Metab. 2008, 9: 679-685. 10.2174/138920008786049249.

    Article  CAS  PubMed  Google Scholar 

  33. Kimura Y, Turner JR, Braasch DA, Buddington RK: Lumenal adenosine and AMP rapidly increase glucose transport by intact small intestine. Am J Physiol Gastrointest Liver Physiol. 2005, 289: G1007-G1014. 10.1152/ajpgi.00085.2005.

    Article  CAS  PubMed  Google Scholar 

  34. Di PR, Melani A, Esposito E, Mazzon E, Paterniti I, Bramanti P, Pedata F, Cuzzocrea S: Adenosine A2A receptor-selective stimulation reduces signaling pathways involved in the development of intestine ischemia and reperfusion injury. Shock. 2010, 33: 541-551.

    Google Scholar 

  35. Antonioli L, Fornai M, Colucci R, Awwad O, Ghisu N, Tuccori M, Da SF, La MC, Natale G, Duranti E, Virdis A, Blandizzi C: The blockade of adenosine deaminase ameliorates chronic experimental colitis through the recruitment of adenosine A2A and A3 receptors. J Pharmacol Exp Ther. 2010, 335: 434-442. 10.1124/jpet.110.171223.

    Article  CAS  PubMed  Google Scholar 

  36. Thiel M, Chambers JD, Chouker A, Fischer S, Zourelidis C, Bardenheuer HJ, Arfors KE, Peter K: Effect of adenosine on the expression of beta(2) integrins and L-selectin of human polymorphonuclear leukocytes in vitro. J Leukoc Biol. 1996, 59: 671-682.

    CAS  PubMed  Google Scholar 

  37. Cronstein BN, Levin RI, Philips M, Hirschhorn R, Abramson SB, Weissmann G: Neutrophil adherence to endothelium is enhanced via adenosine A1 receptors and inhibited via adenosine A2 receptors. J Immunol. 1992, 148: 2201-2206.

    CAS  PubMed  Google Scholar 

  38. Cronstein BN, Rosenstein ED, Kramer SB, Weissmann G, Hirschhorn R: Adenosine; a physiologic modulator of superoxide anion generation by human neutrophils. Adenosine acts via an A2 receptor on human neutrophils. J Immunol. 1985, 135: 1366-1371.

    CAS  PubMed  Google Scholar 

  39. Sullivan GW, Rieger JM, Scheld WM, Macdonald TL, Linden J: Cyclic AMP-dependent inhibition of human neutrophil oxidative activity by substituted 2-propynylcyclohexyl adenosine A(2A) receptor agonists. Br J Pharmacol. 2001, 132: 1017-1026. 10.1038/sj.bjp.0703893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Richter J: Effect of adenosine analogues and cAMP-raising agents on TNF-, GM-CSF-, and chemotactic peptide-induced degranulation in single adherent neutrophils. J Leukoc Biol. 1992, 51: 270-275.

    CAS  PubMed  Google Scholar 

  41. Thiel M, Chouker A: Acting via A2 receptors, adenosine inhibits the production of tumor necrosis factor-alpha of endotoxin-stimulated human polymorphonuclear leukocytes. J Lab Clin Med. 1995, 126: 275-282.

    CAS  PubMed  Google Scholar 

  42. Pawlowski SW, Calabrese G, Kolling GL, Freire R, Alcantara Warren C, Liu B, Sartor B, Guerrant RL: Murine model of Clostridium difficile infection using gnotobiotic aged C57Bl/6 mice and a BI strain. J Infect Dis. 2010, 202: 1708-1712. 10.1086/657086. Erratum in: J Infect Dis 2011,203:1505. Platts-Mills, J [added]

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Smail EH, Cronstein BN, Meshulam T, Esposito AL, Ruggeri RW, Diamond RD: In vitro, Candida albicans releases the immune modulator adenosine and a second, high-molecular weight agent that blocks neutrophil killing. J Immunol. 1992, 148: 3588-3595.

    CAS  PubMed  Google Scholar 

  44. Thammavongsa V, Kern JW, Missiakas DM, Schneewind O: Staphylococcus aureus synthesizes adenosine to escape host immune responses. J Exp Med. 2009, 206: 2417-2427. 10.1084/jem.20090097.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Warren CA, Li Y, Calabrese GM, Freire RS, Zaja-Milatovic S, van Opstal E, Figler RA, Linden J, Guerrant RL: Contribution of Adenosine A2B Receptors in Clostridium difficile Intoxication and Infection. Infect Immun. 2012, Oct 8. [Epub ahead of print]

    Google Scholar 

  46. Strohmeier GR, Reppert SM, Lencer WI, Madara JL: The A2b adenosine receptor mediates cAMP responses to adenosine receptor agonists in human intestinal epithelia. J Biol Chem. 1995, 270: 2387-2394. 10.1074/jbc.270.5.2387.

    Article  CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Acknowledgement

We acknowledge Gina Calabrese, Edward van Opstal and Snjezana Zaja-Milatovic for their technical support and Robert Warren for reviewing the manuscript. The results of this work were partially presented at the 49th Annual Meeting of the Infectious Disease Society of America, Boston MA (Abstract #916). Oct. 19-23 2011. This study was supported by the National Institutes of Health/National Institute of Allergy and Infectious Diseases [U01 AI075526].

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cirle Alcantara Warren.

Additional information

Competing interests

YL, GK, TCB, CAW and RLG have no competing interests. RAF and JR were formerly employed by Dogwood Pharmaceuticals, Inc. JL was a consultant for Dogwood Pharmaceuticals, Inc. RWS is a consultant for Dogwood Pharmaceuticals, Inc.

Authors’ contributions

YL, RAF, TCR, GK conducted experiments. JR, RWS, JL, RLG, CAW participated in study design, critical review of data and manuscript. YL and CAW designed experiments, performed data analyses and drafted manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Li, Y., Figler, R.A., Kolling, G. et al. Adenosine A2A receptor activation reduces recurrence and mortality from Clostridium difficileinfection in mice following vancomycin treatment. BMC Infect Dis 12, 342 (2012). https://doi.org/10.1186/1471-2334-12-342

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2334-12-342

Keywords