Skip to main content
Advertisement
  • Loading metrics

Both pathogen and host dynamically adapt pH responses along the intestinal tract during enteric bacterial infection

  • Sarah E. Woodward,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • Laurel M. P. Neufeld,

    Roles Data curation, Formal analysis, Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada

  • Jorge Peña-Díaz,

    Roles Data curation, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – review & editing

    Affiliations Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • Wenny Feng,

    Roles Formal analysis, Investigation, Validation, Visualization, Writing – review & editing

    Affiliation Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • Antonio Serapio-Palacios,

    Roles Investigation, Methodology, Visualization, Writing – review & editing

    Affiliations Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • Isabel Tarrant,

    Roles Investigation, Writing – review & editing

    Affiliations Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • Wanyin Deng,

    Roles Investigation, Writing – review & editing

    Affiliation Michael Smith Laboratories, University of British Columbia, Vancouver, Canada

  • B. Brett Finlay

    Roles Funding acquisition, Supervision, Writing – review & editing

    bfinlay@interchange.ubc.ca

    Affiliations Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada

Abstract

Enteric pathogens navigate distinct regional microenvironments within the intestine that cue important adaptive behaviors. We investigated the response of Citrobacter rodentium, a model of human pathogenic Escherichia coli infection in mice, to regional gastrointestinal pH. We found that small intestinal pH (4.4–4.8) triggered virulence gene expression and altered cell morphology, supporting initial intestinal attachment, while higher pH, representative of C. rodentium’s replicative niches further along the murine intestine, supported pathogen growth. Gastric pH, a key barrier to intestinal colonization, caused significant accumulation of intra-bacterial reactive oxygen species (ROS), inhibiting growth of C. rodentium and related human pathogens. Within-host adaptation increased gastric acid survival, which may be due to a robust acid tolerance response (ATR) induced at colonic pH. However, the intestinal environment changes throughout the course of infection. We found that murine gastric pH decreases postinfection, corresponding to increased serum gastrin levels and altered host expression of acid secretion-related genes. Similar responses following Salmonella infection may indicate a protective host response to limit further pathogen ingestion. Together, we highlight interlinked bacterial and host adaptive pH responses as an important component of host–pathogen coevolution.

Introduction

Gastrointestinal pH is a key component of gut physiology, known to vary across regions of the intestine [1,2]. In humans, intestinal pH is characterized by a progressive increase from the duodenum to the distal ileum (pH 6.0 to 7.5), a drop at the cecum to pH 6.4, followed by increasing pH from the proximal to the distal colon (pH 7 to 7.5; Fig 1A) [13]. In mice, reported intestinal pH is much lower, with the intestinal contents of female BALB/c mice ranging from 2.98 in the stomach to 5.24 in the ileum (Fig 1A) [4]. In both species, maintaining intestinal pH is important to homeostasis and gut function. Disruption to intestinal ion transport is associated with spontaneous onset of colitis symptoms in mice and increased susceptibility to induced colitis [57]. pH also influences which microbial species colonize the gut, with mildly acidic pH resembling proximal colon conditions inhibiting the growth of Bacteroides spp. [810]. Therefore, maintenance of gut pH is important to preventing overgrowth of colonizing bacteria, whether commensal or pathogenic [8,9].

thumbnail
Fig 1. Gastrointestinal pH varies across gut regions.

(A) Average lumenal pH of gut regions in murine and human gastrointestinal tracts, as reported in the literature [14,4446]. (B) Lumenal pH measured across the gut of uninfected C57BL/6 mice (N = 8–12). Error represents mean +/− SD. Statistical analysis represents a one-way ANOVA with Tukey’s multiple comparisons test. Summary data displayed in Fig 1 can be found in S1 Data. Created with BioRender.com.

https://doi.org/10.1371/journal.pbio.3002761.g001

Intestinal pH is often overlooked with regard to enteric infection, despite its potential role in affecting infection dynamics, host–pathogen interactions, and possibly even therapeutic success. By definition, diarrheal disease agents interact with the gastrointestinal (GI) environment to establish infection. Pathogenic Escherichia coli, including enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC), are a diverse group of diarrheal disease agents which result in diarrhea, fever, vomiting, and even death in infected hosts [11,12]. Citrobacter rodentium, a related pathogen that shares similar virulence factors, is used to model murine EPEC/EHEC infection [1315]. C. rodentium naturally infects mice, causing transmissible murine colonic hyperplasia (a form of colitis) with diarrheal disease symptoms [16]. All 3 pathogens are transmitted through the fecal-oral route and use a type III secretion system (T3SS) for successful colonization [13,1721], translocating protein effectors directly into host cells for bacterial control over host cell processes [22].

It is clear that EPEC/EHEC/C. rodentium respond to intestinal pH during host colonization. In particular, the extreme acidic conditions of the gastric environment are a major bottleneck to both C. rodentium and EHEC colonization [2326]. Furthermore, within the small intestine C. rodentium responds to alkaline bicarbonate ions secreted by the host to neutralize acidic chyme by up-regulating genes involved in initial epithelial attachment [27,28]. Despite this work, much remains unclear about how EPEC/EHEC/C. rodentium respond to regional differences in gastrointestinal pH. Furthermore, while the GI environment is known to change as a consequence of diarrheal disease, little investigation has been done to profile intestinal pH postinfection [29].

This study investigates the behavior and virulence of C. rodentium in response to variations in host intestinal pH. We profiled pH across the intestine of one of the most commonly used laboratory mouse strains (C57BL/6) to define a range of physiologically relevant pH encountered by enteric pathogens upon initial host contact. We identified pH-specific regulation of the T3SS and epithelial attachment by C. rodentium, as well as significant changes to stress adaptation. Additionally, within-host adaptation increased C. rodentium’s acid tolerance, stimulated by colonic pH conditions. The pH of the gut also changed during established C. rodentium infection, with a significant drop in gastric pH which correlated with pathogen burden. This effect was consistent across additional models of intestinal damage, suggesting a possible host defense mechanism against further pathogen ingestion. Together, these data emphasize the interplay between pathogens and host intestinal pH in the context of enteric infection.

Materials and methods

Ethics statement

Animal experiments were performed in accordance with the guidelines of the Canadian Council on Animal Care and the University of British Columbia (UBC) Animal Care Committee. Approval for the work carried out in this study was granted by the UBC Animal Care Committee according to Animal Care Protocols A20-0187 and A17-0228.

Bacterial strains and culture conditions

Bacterial strains are listed in S1 Table. Unless otherwise noted, C. rodentium strain DBS100 was used in all assays [30]. A fresh bacterial stock was used for all assays as follows: 2 days in advance bacteria were streaked onto a fresh LB agar plate from frozen (−70°C in glycerol) and grown overnight at 37°C. A single colony was re-inoculated into LB (lysogeny broth, Sigma) broth and grown overnight at 37°C, shaking. To test the response of bacteria to gastrointestinal pH, LB broth or DMEM (Dulbecco’s Modified Eagle Medium, Hyclone) was adjusted to gastrointestinal pH by the addition of 37% HCl or 40% NaOH. Due to the high buffering capabilities of DMEM, media were pH-adjusted fresh for use within 12 h. pHs tested were: 3.5, 4.0, 4.4, 4.8, 5.7, 6.0, 6.4, 7.0, and 7.5. To test the effect of pH on human pathogenic E. coli, EHEC strain EDL933 [31] and EPEC strain E2348/69 [32] were used, as well as Salmonella enterica serovar Typhimurium strain CDC 6516–60 (ATCC 14028).

ldcC mutant construction

The sacB gene-based allelic exchange method and the suicide vector pRE112 [33] were used to generate a chromosomal in-frame deletion mutant of ldcC in C. rodentium strain DBS100. The immediate upstream and downstream flanking regions of ldcC were amplified by PCR using primers listed in S2 Table. Purified DNA fragments were ligated into pRE112 by Gibson assembly. The suicide vector was transformed into E. coli strain MFDpir [34] by electroporation, which was used as the donor strain to transfer the plasmid by conjugation into C. rodentium. After sucrose selection, colonies resistant to sucrose and sensitive to chloramphenicol were screened, and then further verified, for ldcC deletion by PCR using primers Check_ldcC_F and Check_ldcC_R.

In vivo pH measurements and mouse infections

As mentioned above, animal experiments were performed in accordance with the guidelines of the Canadian Council on Animal Care and the University of British Columbia (UBC) Animal Care Committee according to Animal Care Protocols A20-0187 and A17-0228. C57BL/6 and BALB/c mice were ordered from Jackson Laboratory (Bar Harbor, Maine, United States of America) and maintained in a specific pathogen-free facility at UBC on a 12-h light-dark cycle. Mice were allowed to acclimatize to the facility for 1 week following arrival. All pH measurements and infection models were run after the acclimatization period.

pH measurements were performed on fresh, undiluted intestinal content, using an Elite pH Spear Pocket Tester (Thermo Scientific). Measured pH represents the average of 2 to 3 readings per sample. All bacterial colonization experiments were done in 7-week-old female C57BL/6 mice fed the Picolab Rodent Diet 20 chow (Cat. no. 5053; LabDiet). Mice were monitored daily throughout the 4 to 8 day infections for weight loss and clinical symptoms.

C. rodentium infections were carried out by gavaging mice orally with 108 colony-forming units (CFUs) of C. rodentium DBS100 from overnight culture. Mice were euthanized at experimental endpoint by isoflurane anesthesia followed by carbon dioxide inhalation. Bacterial burden was determined by collecting colon samples into 1 mL of reduced phosphate-buffered saline (PBS) and homogenized in a FastPrep-24 (MP Biomedicals) at 5.5 m/s for 2 min. Sample homogenate was diluted for plating on MacConkey agar (Difco) and incubated for 18 to 20 h at 37°C before counting bacterial colonies.

Chemically induced colitis was triggered by exposure to 3% dextran sulfate sodium (DSS) in the drinking water for 4 days before being placed on regular drinking water. Mice were euthanized at 5 days posttreatment onset.

Colonization with both avirulent C. rodentium ΔescN [35] and commensal E. coli Mt1B1 [36] were carried out by oral gavage with 2.5–4 × 108 CFU from overnight culture, as determined by retrospective plating. Control WT-infected mice were inoculated at the same dosage. S. Typhimurium (ATCC 14028) infections were carried out by oral gavage with 1 × 106 CFU of S. Typhimurium from overnight culture. Fecal samples were processed for CFU enumeration as described above.

Analysis of rodent diets

To compare standard rodent diets, mice were fed either the Teklad Global 18% Protein Rodent Diet (Cat. no. 2918; Envigo) or Picolab Rodent Diet 20 (Cat. no. 5053; LabDiet), over a period of 2 weeks before measurement. Mouse chow pH was measured by dissolving pellets in 3 mL deionized water per 100 mg.

Bacterial growth at gastrointestinal pH

Bacterial growth at gastrointestinal pH was determined by inoculating bacteria at an optical density at 600 nm (OD600) of 0.005 from an overnight culture into 250 μL total volume of the media of interest (LB or DMEM base media adjusted to gastrointestinal pH). Cultures were incubated at 37°C with agitation for 20 h in a Synergy H1 plate reader (Biotek) with OD600 measurements at 10-min intervals.

Cell culture assays

Murine rectal carcinoma cell line CMT-93 (ATCC CCL-223) and human adenocarcinoma cell line Caco-2/TC7 were maintained in DMEM supplemented with 10% fetal bovine serum, 1% Glutamax, and 1% nonessential amino acids. Cells were used between passages 4 to 10. Cells were seeded in 12-well plates at 90,000 cells/well and incubated for 48 h prior to infection. Confluent CMT-93 cells were washed once with PBS before infection. The C. rodentium inoculum was prepared by subculturing 1:40 from overnight culture to mid-log phase (3.5 h, 37°C, shaking) in either LB or DMEM media adjusted to intestinal pH. Subcultures were washed and resuspended in cell culture media at multiplicity of infection (MOI) 100. Infected plates were centrifuged (1,000 rpm, 5 min) to synchronize the infection before incubation for 4 h at 37°C and 5% CO2. Infected plates were washed 5 times with PBS before detachment using 0.1% Triton X-100 in PBS for dilution and plating on neutral LB agar. Adherent bacterial CFU were quantified and data were normalized to exact inoculum dosage (determined by retrospective plating) to account for differences between pH conditions.

Evaluation of actin pedestal formation was performed as follows. Caco-2/TC7 and CMT-93 cells were cultured in a 6-channel slide (Cat. No. 80606; ibidi) to 80% confluence before infection as before with C. rodentium preconditioned to mid-log phase at pH 4.8 or pH 7.0. Following the 4-h infection, cells were washed with PBS, fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.1% of Triton X-100 for 10 min, and blocked with 3% BSA–PBS for 30 min. Cells were immunostained using a polyclonal rat anti-Tir antibody [17] overnight, followed by a goat anti-rat IgG (H+L) secondary antibody conjugated to Alexa Fluor 568 (Cat. no. A-11077; Thermo Fisher Scientific) for 45 min. F-actin was stained with Alexa Fluor 488 phalloidin (Cat. no. A12379). Nuclei and bacteria were stained with DAPI. Cells were imaged with a Leica Stellaris 5 confocal microscope using a 63× objective and analyzed using LAS-X software.

Bacterial and eukaryotic gene expression analysis

C. rodentium was subcultured for 3.5 h 1:40 in LB base media adjusted to gastrointestinal pH, as before. After subculture samples were preserved in RNAprotect Bacterial Reagent (Qiagen) and stored at −70°C until extraction. Bacterial RNA extractions were done using the RNAse-Free DNase Set (Cat. no. 79254, Qiagen) as per the manufacturer’s instructions. A QuantiTect Reverse Transcription Kit (Cat. no. 205313, Qiagen) was used for cDNA synthesis and Quantitative RT-qPCR was performed using a QuantiNova SYBR Green RT-PCR Kit (Cat. no. 208056) in combination with primers (S3 Table) for target gene mRNA. dnaQ was used as an endogenous control. Data were normalized to efficiency of primers by target gene (91.6% to 103.5% for all primers used).

Eukaryotic RNA was isolated from mouse tissue samples (stomach and colon) stored in RNAprotect Tissue Reagent (Qiagen) at −70°C before extraction using a GeneJET RNA Purification Kit (Cat. no. K0731, Thermo Scientific). cDNA synthesis and Quantitative RT-qPCR was performed as described above using various primer pairs (S3 Table). β-2-microglobulin (B2M) was used as an endogenous control.

Biofilm formation and metabolic activity

To assess pH-induced biofilm formation, C. rodentium was inoculated in LB at physiological pH to an OD600 of 0.5 in a black, clear bottom 96-well plate. The plate was incubated at 37°C, standing, for 48 h to allow for biofilm formation. After 48 h, culture media were removed and the OD600 of the resultant biofilm was measured in a Synergy H1 plate reader (Biotek), and 100 μL of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) at a concentration of 0.5 mg/mL in LB was added to the biofilms and allowed to incubate at 37°C for 10 min. MTT was removed and replaced with 100 μL of 100% DMSO for resuspension before measuring OD at 570 nm to approximate formazan production as a readout of metabolic activity.

Bacterial cell morphology

C. rodentium was cultured to mid-log phase (3.5 h, 37°C with agitation) at gastrointestinal pH before being spun down and resuspended in PBS with the addition of FM 1–43 Dye (N-(3-Triethylammoniumpropyl)-4-(4-(Dibutylamino) Styryl) Pyridinium Dibromide) (Cat. no. T3163, Invitrogen). Images were taken on a Zeiss X10 light microscope and cell area was measured using CellProfiler 4.2.1 [37,38].

Bacterial survival assays

To determine whether the deletion of cpxRA would affect C. rodentium survival at gastric pH, we cultured C. rodentium at pH 3.5 as described [23]. In brief, WT, ΔcpxRA [39], or ΔcpxRA::cpxRA C. rodentium strains were inoculated 1:20 from overnight culture into unadjusted LB (the baseline “untreated” condition) or LB adjusted to gastric pH 3.5. At 30-min intervals, subcultures were sampled and diluted in PBS for plating on neutral LB agar to determine viability by quantifying surviving CFU. Viable CFU were compared to the inoculum population to determine percent survival over time.

To test for the acid tolerance response (ATR) following preinduction at intestinal pH, C. rodentium was cultured overnight at either neutral pH 7 or colonic pH 5.7 under anaerobic conditions (90% N2, 5% CO2, 5% H2) or aerobically at 37°C without agitation. Overnight cultures were diluted 1:20 into unadjusted LB (the baseline “untreated” condition), or LB adjusted to gastric pH 3.5 and plated at 60-min intervals as previously described.

To determine whether C. rodentium shed from the mouse gut at early (day 2) and peak infection (day 7) differed in acid tolerance, mice were infected with C. rodentium as described above and fecal pellets were collected and homogenized fresh in 1 mL of PBS. Fecal homogenate was inoculated in LB pH 3.5 as described above and plated for enumeration of viable C. rodentium at 30-min intervals. A subset of fecal pellets was not homogenized to simulate natural encapsulation of C. rodentium during gastric acid exposure. Instead, fecal pellets were cut in half, with one half plated for CFU to determine the inoculating population, and the second half dropped into LB pH 3.5 as is for incubation as before. Pellets were removed from pH 3.5 after 120 min and homogenized for plating and CFU counts.

Intra-bacterial redox potential

Assays were performed using a protocol adapted from [40,41]. In brief, a roGFP2 C. rodentium reporter strain was grown overnight in LB containing 100 μg/mL of carbenicillin. Overnight cultures were diluted 1:25 into 50 mL of pre-warmed LB without antibiotics and were grown for 5 h at 37°C in a shaking incubator. Bacterial strains were washed and resuspended in saline solution (0.9% NaCl w/w). Cultures were then exposed to the different pH of interest in a black 96-well plate with a volume of 200 μL and an OD600 of 1. Intra-bacterial redox was assessed by measuring fluorescence with an excitation at 405 and 480 nm and an emission at 510 nm. Background fluorescence was controlled by measuring the fluorescence of WT C. rodentium. Normalized 405/480 nm ratios were calculated by using fully oxidized (100 mM H2O2) and reduced (10 mM DTT) control conditions.

ELISA analysis of fasting serum gastrin levels

Blood was collected from naïve and day 8-infected mice following a 12-h fast. Whole blood was allowed to clot for 2 h on ice before serum collection. Gastrin levels were analyzed using the mouse gastrin enzyme-linked immunosorbent assay (ELISA) kit (Novus Biologicals, Cat. No. NBP3-08148). Absorbance at 450 nm was measured using a Synergy H1 plate reader (Biotek).

ELISA analysis of inflammation

Serum calprotectin levels were analyzed using the mouse S100A9 ELISA kit (Hycult), as per the manufacturer’s instructions. Serum was collected from non-fasted mice by allowing whole blood to clot for 2 h on ice before spinning to collect serum. Absorbance at 450 nm was measured using a Synergy H1 plate reader (Biotek).

Cytokine measurements

To analyze intestinal inflammation of mice colonized with either E. coli Mt1B1, ΔescN C. rodentium, or WT C. rodentium as compared to naïve animals, colon tissue samples were collected at 4 days postinoculation by first removing intestinal contents. Samples were collected into 1 mL of PBS containing a protease inhibitor cocktail (Roche Diagnostics) and homogenized using a FastPrep-24 (MP Biomedicals) at 5.5 m/s for 2 min. Homogenate was then spun down at 16,000 × g for 20 min at 4°C for supernatant collection. Inflammatory cytokines were measured using a mouse inflammation cytometric bead array (CBA) kit (BD Biosciences, Cat. no. 552364) according to manufacturer’s instructions. Measurements were made on an Attune NxT flow cytometer (Thermo Fisher Scientific), normalizing cytokine concentrations to tissue weight.

Statistical analysis

Statistical analysis was performed in Graphpad Prism (www.graphpad.com) and clarified in figure legends. Unless otherwise stated, analysis was performed using a Mann–Whitney test to compare 2 groups and a one-way ANOVA with Šidák’s multiple comparisons test for more than 2 groups. Aggregate results represent the mean +/− SD and statistical significance is represented by *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

Results

Regional variability in gastrointestinal pH may be influenced by genetic background

To assess the response of C. rodentium to intestinal pH, we first defined regional pH ranges within its natural host, the mouse. We compared reported gastrointestinal pH (pH 2 to 6) to pH measured in healthy, female, 7-week-old C57BL/6 mice across gut regions (Fig 1A and 1B). We observed a similar pattern to reported murine and human values, which see pH increase at significant increments towards the ileum-large intestinal regions (Fig 1B). At the cecum, we found a nonsignificant drop in pH which is characteristic of human intestinal pH, but not previously observed in mice [1]. Across all regions, we observed variability between individuals, particularly in the stomach and ileum, demonstrating that gastrointestinal pH likely fluctuates over time and with varied consumption of food and water.

Overall however, the pH across both the small intestine (duodenum, jejunum, and ileum) and large intestine (cecum and colon) was more basic than reported murine values in BALB/c mice [4]. Mice in the aforementioned study were fed a different diet, the Teklad Global 18% Protein Rodent Diet (Teklad), with a lower protein content (18%) [42]. Dietary protein is thought to increase intestinal pH by increasing ammonia production as a by-product of bacterial metabolism [2,9,43]. We therefore placed C57BL/6 mice on either the Teklad diet or Picolab Rodent Diet 20 5053 (Picolab; 21% protein) for 2 weeks before euthanasia. We found no significant differences between diets, except in the distal colon where pH was significantly higher on the lower protein Teklad diet (S1A and S1B Fig). This indicates that the more alkaline pH observed across gut regions may instead be due to differences in mouse genetic background (BALB/c versus C57BL/6). Indeed, while the only comprehensive profiling of murine intestinal pH has been done in BALB/c mice, ileal, cecal, and colonic pH has been reported to be more alkaline in FVB/N and ICR mouse backgrounds, and more closely representative of values found in the human gut (Fig 1) [4446].

To verify whether mouse genetic background can indeed result in altered intestinal pH, we compared the intestinal pH of both BALB/c and C57BL/6 mice fed the Picolab diet. We found a significant difference in intestinal pH in the ileum and colon regions (S1C Fig). As reported previously, we did not observe a drop in intestinal pH of BALB/c mice from the ileum to cecum regions, rather pH increased along the length of the GI tract. This indicates that genetic differences between mouse strains may result in altered intestinal pH, with pH of C57BL/6 mice more closely reflecting the pattern found in humans. Altogether, these data establish important physiological variation within the naïve murine gastrointestinal tract, which could be used by enteric pathogens to signal their biogeographical location.

C. rodentium alters growth dynamics, epithelial attachment, and virulence gene expression in response to gastrointestinal pH

Having established the range of physiologically relevant pH, we next investigated whether C. rodentium responds to variations in pH by adjusting the pH of LB to mimic the ranges of pH found in both mouse and human GI tracts. Growth of C. rodentium was highest at the range of pH 6.4 to 7.5, with impaired growth at acidic pHs 4.4 and 4.8, resembling the gastric and duodenal regions (Fig 2A). As previously reported [23], no growth was observed at gastric pH of 3.5 to 4. Human enteropathogens EHEC, EPEC, and S. Typhimurium were similarly unable to grow at gastric pH 3.5 and displayed altered growth dynamics in response to pH below 5 (S2A–S2C Fig). This suggests that while acid tolerance and viability rates differ between pathogens [26,47], they are all affected by fluctuations in gastrointestinal pH.

thumbnail
Fig 2. C. rodentium alters its behavior and virulence in response to regional gastrointestinal pH.

(A) Growth in pH-adjusted LB across the range of gastrointestinal pH, measured as OD 600 nm at 10-min intervals over 20 h. (B) Attachment of C. rodentium to CMT-93 murine colonic epithelial cells after preinduction at gastrointestinal pH. Data represent biological replicates (N = 3–4; each the average of 3 technical replicates). Statistical analysis represents an ordinary one-way ANOVA test with Holm–Šidák’s multiple comparisons test. (C) Gene expression analysis by RT-qPCR of key virulence- and early infection-related genes across the range of gastrointestinal pH. Statistical analysis represents t tests with Bonferroni correction for multiple comparisons. Outlier tests were run using the ROUT method (Q = 1%), identifying no outliers. Error represents mean +/− SD. (D) Representative images of actin pedestal formation on Caco-2-/TC7 cells following infection with C. rodentium pre-induced at pH 4.8 and pH 7.0 (N = 3 biological replicates). Scale bar represents 30 μm. Arrows indicate representative Tir-rich pedestals. Statistics represents an unpaired t test. Summary data displayed in Fig 2 can be found in S1 Data. LB, lysogeny broth; OD, optical density.

https://doi.org/10.1371/journal.pbio.3002761.g002

We further investigated the relative metabolic activity of C. rodentium subjected to intestinal pH by measuring the reduction of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) to formazan by NADPH-dependent oxidoreductase enzymes in metabolically active bacteria. As expected, bacteria incubated at pH 3.5 produced significantly less formazan compared to neutral pH despite an increased biomass, suggesting decreased metabolic activity (S2D and S2E Fig). These results support both the absence of growth at pH 3.5 and the low recoverability of cells after removal from acidic pH [23]. The observed increase in biomass at pH 3.5 may further suggest the active encapsulation of community members within biofilms. Biofilm formation is an important stress-induced protective behavior of bacteria which allows for the survival of community members shielded within. Aside from direct death of bacteria under low pH, bacteria within biofilms are also known to decrease their metabolic activity under stress, which promotes survival and persistence, especially as many stressors affect the bacterial membrane (such as antibiotics) and have the greatest impacts on actively growing bacteria [48]. Taken together, extreme low pH of the gastric environment has considerable consequences to bacterial fitness.

To determine if the altered fitness dynamics observed across gastrointestinal pH also reflect differences in pathogen virulence, we assessed epithelial cell attachment, an essential characteristic of infection. This was done by “pre-inducing” C. rodentium via subculture in pH-adjusted LB before infection of CMT-93 cells, a murine rectal cell line chosen because the rectum is a known large intestinal niche of C. rodentium during host colonization. We expected attachment to increase in response to pH representing key replicative niches of C. rodentium, such as the cecum and colon. However, C. rodentium attachment was significantly increased after preinduction at pH 3.5 and 4.4, representing gastric-duodenal pH (Fig 2B). Attachment was lowest at pH 7.5 which represents the upper range of ileal pH, as well as pH found in the extra-intestinal environment. Assays were repeated using subculture in pH-adjusted DMEM media, which is known to activate expression of the T3SS. Despite high buffering of DMEM media making accuracy difficult, the same trends were noted in both growth and attachment (S2F and S2G Fig). Therefore, LB media was used in all subsequent assays.

To further explore pH-mediated virulence regulation, we characterized the gene expression profile of known virulence- and early infection-associated genes following C. rodentium subculture in pH-adjusted LB prior to RNA extraction. Despite the fact that LB media is not known to activate the T3SS, type III secretion-associated virulence genes ler, tir, eae, and effector gene nleB, were highly expressed at pH 4.8 which represents duodenal pH (a 4- to 5-fold increase from baseline). At gastric pH 3.5, we observed down-regulation of virulence genes associated with type III secretion, including both structural and effector-associated genes (Fig 2C), as well as virulence regulators ler and regA [49]. While a known fimbrial gene fimA was also down-regulated, we found an up-regulation of kfcC, a component of the C. rodentium type IV pilus, which may explain the increase in epithelial cell attachment observed at gastric pH [49].

To further investigate the extent to which the T3SS contributes to pH-mediated attachment, we evaluated the ability of a ΔescN C. rodentium strain lacking the T3SS to attach to CMT-93 cells following preinduction as before. We found that infection with the ΔescN strain did not result in decreased epithelial attachment following preinduction in LB at low pH 3.5 and 4.4 (S2H Fig), further supporting that low pH induced epithelial attachment is mediated by non-T3SS factors. To determine whether the induction of T3SS-related genes at pH 4.8 results in increased actin polymerization and pedestal formation—a hallmark of infection by attaching and effacing pathogens such as C. rodentium, EHEC, and EPEC–CMT-93 cells were infected as before with either C. rodentium pre-induced at pH 4.8 or neutral pH 7.0. As CMT-93 cells do not demonstrate robust visual actin polymerization, we have further performed these assays using the human intestinal adenocarcinoma cell line Caco-2/TC7. Following infection, cells were stained for translocated intimin receptor (Tir), a core component of the T3SS used to stain for actin-rich T3SS-induced pedestals on epithelial cells [50]. We found that average number of pedestals per Caco-2/TC7 epithelial cell was significantly higher following 4-h infection with pH 4.8-induced C. rodentium (Fig 2D). As expected, C. rodentium was less able to induce pedestal formation in CMT-93 cells. However, we still observed a trend towards increased pedestal formation following infection with bacteria pre-induced at pH 4.8 (S2I Fig). Together, these data demonstrate that C. rodentium regulates virulence gene expression in a pH-dependent manner, leading to increased attachment-related behavior upon initial entry to the intestinal environment.

Intestinal pH activates transcription of diverse bacterial stress responses

Bacterial stress responses and virulence are closely co-regulated [26,51]. Therefore, we next investigated the relationship between intestinal pH and the transcriptional expression of stress-related genes. A key strategy of food-borne pathogens under acid stress is to increase intracellular pH, which can be done via production of amino acid decarboxylases which allow for the consumption of cytoplasmic protons [52]. Interestingly, we did not find homology within the C. rodentium genome to either the transcriptional regulators GadW and GadX of the glutamate decarboxylase acid-resistance system, nor to AdiC of the arginine acid-resistance system found in some E. coli strains, including EHEC [53,54]. We did however identify 70% protein sequence homology between the E. coli CadA, a lysine decarboxylase, and C. rodentium LdcC (S3A Fig). As expected, we found that transcript levels of ldcC increased at low pH from 4.4 to 5.7 (Fig 3A), indicating a potential role for LdcC in survival of acid stress. To further investigate this, we challenged wild-type C. rodentium and a C. rodentium strain lacking ldcC to gastric pH 3.5 over a range of 0 to 120 min. We found that viability of the ΔldcC strain was significantly impaired at 120 min as compared to the wild-type strain (S3B Fig). These data indicate that C. rodentium uses amino acid decarboxylation to combat acid-induced stress despite lacking homology to 2 major systems in related E. coli. However, the effect of ldcC deletion was subtle, with no significant difference observed upon initial exposure to low pH, indicating that C. rodentium likely employs several strategies of acid adaptation.

thumbnail
Fig 3. Stress gene regulation is affected by gastrointestinal pH.

(A) Expression of acid stress-associated genes across the range of gastrointestinal pH by RT-qPCR. Data represent biological replicates (average of 3 technical replicates; N = 4). (B) Expression of general stress-associated genes across the range of gastrointestinal pH by RT-qPCR. Data represent biological replicates (average of 3 technical replicates; N = 3–4). (C) Intracellular ROS accumulation at 3 h postinoculation into LB at gastrointestinal pH, as determined using a roGFP2 reporter C. rodentium strain. Data represent biological replicates (average of 3 technical replicates; N = 3). Statistical analysis represents t tests with Bonferroni correction. Outlier tests were run using the ROUT method (Q = 1%), identifying no outliers. Summary data displayed in Fig 3 can be found in S1 Data. LB, lysogeny broth; ROS, reactive oxygen species.

https://doi.org/10.1371/journal.pbio.3002761.g003

Another strategy of food-borne pathogens in dealing with acid stress is proton efflux [52]. The F1F0-ATPase proton efflux membrane ATPase is conserved across many bacterial pathogens, including C. rodentium, in which it is encoded by the atpD gene. We observed a 7-fold increase in expression of atpD at pH 4.4 compared to neutral media (Fig 3A). Additionally, upon searching the C. rodentium genome for genes annotated as involved in acid resistance, we chose to further investigate the expression of yodD, a stress-induced peroxide/acid resistance protein, and asr, the acid resistance repetitive basic protein, both of which are shared with EHEC. Both genes were up-regulated at gastric pH 3.5, and asr in particular demonstrated a 300-fold increase in expression at pH 4.4, suggesting that both of these genes are important to Citrobacter’s acid resistance strategy within the upper GI tract.

Aside from direct mechanisms of acid resistance, we further profiled the effect of intestinal pH on nonspecific stress responses which have evolved as general mechanisms to overcome environmental stress. We investigated the expression of rpoS, a gene encoding the stationary phase sigma factor present in Gram-negative bacteria which controls general stress responses to stressors such as oxidative or osmotic stress. RpoS is important for C. rodentium survival under heat and H2O2 conditions, as well as full pathogen virulence [55,56]. Mutation of rpoS in EHEC has also been found to decrease resistance to acid stress [57]. While rpoS expression was therefore expected to increase with decreased pH, it was unaltered across the pH range tested (Fig 3B). This was surprising, given that when we exposed a roGFP2 reporter strain of C. rodentium, which can be used to measure intra-bacterial redox dynamics, to different pHs we found that gastric pH 3.5 to 4 significantly increased oxidative stress (Fig 3C) [41]. We further investigated expression of fur, encoding a ferric iron transcription regulator which may regulate transcription of genes that protect against ROS damage [52]. While we observed a trend towards increased fur expression at pH 4.4, there was no significant up- or down-regulation of the system induced by altered pH. Despite the lack of ROS-specific gene expression, we did observe evidence that bacteria at low pH respond to general cell damage. For example, we found that expression of recA which encodes a recombinase protein used to repair DNA double-strand breaks increased with increased acidity of the culture media, suggesting cell stress and DNA damage at pH 4.8 and lower (Fig 3B) [58].

pH-induced stress alters membrane regulation and cellular morphology

An important outcome of stress-induced gene regulation is the repair and maintenance of the bacterial cell membrane, which can even induce adaptive changes to cellular morphology. To visualize potential morphological changes induced by pH, C. rodentium was pre-induced at GI pH before staining with FM 1–43 Dye. We observed bacterial cell lengthening at pH 4.0 to 4.8, an indicator of cell stress, and a trend towards small size and abnormal cell shape at pH 3.5 (Fig 4A and 4B). We also observed a decrease in membrane intensity in a subpopulation of cells at pH 3.5, possibly reflecting a loss of membrane integrity [23].

thumbnail
Fig 4. Membrane regulation in response to intestinal pH.

(A) Representative cellular morphology after 3.5 h at relevant gastrointestinal pHs by light microscopy and cell membrane staining. Scale bar represents 5 μm. (B) Cell area measurements from light microscopy across gastrointestinal pH (N = 3 biological replicates, each the sum of 2 technical replicates). (C) Expression of genes by RT-qPCR associated with the Cpx stress response following culture at gastrointestinal pH. Statistical analysis represents t tests with Bonferroni correction for multiple comparisons. Outlier tests were run using the ROUT method (Q = 1%), identifying no outliers. Data represent biological replicates (average of 3 technical replicates; N = 3). (D) Survival of wild-type, ΔcpxRA, ΔcpxRA::cpxRA C. rodentium in gastric pH 3.5 over the range of average gastric emptying time. Each point represents the average of 3–5 biological replicates (3 technical replicates per biological replicate). Statistical analysis represents a mixed-effects model with Geisser–Greenhouse correction and Šidák’s multiple comparisons test. Color of stars indicates significance of Δcpx (pink) or complemented (green) strains compared to the WT strain. Summary data displayed in Fig 4 can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.g004

The Cpx envelope stress response is an important mechanism to overcome conditions which result in misfolding of envelope proteins, such as alkaline pH, and has been shown to be required for C. rodentium virulence [51,59]. We found that cpxA, part of the CpxRA two-component system which mediates the Cpx response, was most highly expressed at low pH 4.4 [51]. CpxRA is known to regulate both the expression of periplasmic proteases such as DegP, as well as T3SS-related virulence. We found that expression of degP, which is induced by stress [60], was significantly up-regulated in response to subculture at pH 3.5, 4.4, 5.7, and 6.0 compared to neutral base media (Fig 4C). degP was not up-regulated at pH 4.8 which resulted in a strong activation of T3SS-related genes, consistent with reported inverse regulation of these systems (Fig 2C), and supporting Cpx stress response regulation under low upper intestinal pH conditions.

The above data suggest that membrane protein synthesis and degradation must be tightly regulated to allow for successful pathogen adaptation to variable intestinal pH. If this is the case, interfering with low pH-induced membrane regulation will impact C. rodentium survival under acidic conditions. We therefore tested survival at gastric pH 3.5 of wild-type C. rodentium or a strain deficient in the CpxRA two-component system (ΔcpxRA), and which is unable to properly regulate its membrane stress response [51]. While there was no difference upon initial exposure to pH 3.5 (0 min) between wild-type, ΔcpxRA, and ΔcpxRA::cpxRA strains, the ΔcpxRA strain exhibited significantly reduced survival after 30 to 120 min, with up to an average of 3.4% viability in the absence of cpxRA after 120 min, which represents the upper limit of gastric emptying time, as compared to 53.5% in the complement strain (Fig 4D) [61]. The Cpx response is therefore not only necessary for C. rodentium virulence, but is also critical to gastric passage and overall host–host transmission. Overall, these data suggest that C. rodentium alters its membrane and overall shape under gastrointestinal pH conditions to adapt to its surrounding environment.

Host-adaptation increases C. rodentium acid tolerance which may support host transmission

During an established infection, C. rodentium shed from the gut at peak infection is known to display a hypervirulent state characterized by altered T3SS expression and tissue colonization dynamics, and the ability to colonize new hosts at a reduced infectious dose, allowing for more effective transmission. This hypervirulent state is quickly lost following ex vivo passage, indicating that it results from within-host adaptation rather than evolutionary change [62,63]. We hypothesized that a crucial part of this host-adapted (HA) state would include adaptation to better tolerate the gastric environment. While the conditions that trigger host-adaptation remain unknown, since host-adaptation occurs following passage through the mouse gut, we investigated whether in vitro culture at colonic pH representing the key in vivo niche of C. rodentium, ahead of gastric pH exposure, could induce an ATR and mimic the protection observed after gastrointestinal transit. Indeed, we observed a striking ATR following stationary phase growth at colonic pH 5.7 under anaerobic conditions (Fig 5A). While culture at neutral pH resulted in only 20.6% survival after 2 h at gastric pH, subculture at colonic pH 5.7 resulted in an average survival rate of 83.6%. Anaerobic conditions were chosen to mimic passage through the low oxygen large intestinal environment. We did not observe an ATR under aerobic conditions nor in response to short-term exposure to low pH (preinduction to mid-log phase) (S4A Fig).

thumbnail
Fig 5. HA C. rodentium is better able to survive acid-induced stress.

(A) C. rodentium survival at gastric pH after initial anaerobic preinduction at either colonic pH 5.7 or neutral pH. Each point represents the average of 3 biological replicates (3 technical replicates per biological replicate). Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. (B) Survival of C. rodentium in homogenized feces collected on days 2 and 7 postinfection, representing non-adapted and host-adapted states (N = 6–8). ND, not detected. Summary data displayed in Fig 5 can be found in S1 Data. HA, host-adapted; LB, lysogeny broth.

https://doi.org/10.1371/journal.pbio.3002761.g005

Having demonstrated a robust ATR under colonic pH conditions, we next sought to determine whether HA-C. rodentium conditioned within the host gut, demonstrates increased acid tolerance. We evaluated the survival of C. rodentium shed in the feces of infected mice at day 2, representing an early, non-HA state, and C. rodentium shed at day 7 postinfection, representing the HA state (Fig 5B). Though we found similar percent survival at 0 to 30 min postexposure to pH 3.5, none of the day 2-shed C. rodentium was able to survive 120 min at gastric pH. As these fecal samples were homogenized before exposure to pH 3.5 rather than naturally encapsulated in the fecal pellet, we repeated this assay on day 2 feces without homogenization. Despite an increase to 8% survival, we still observed significant population loss (S4B Fig). This indicates that while natural encapsulation, such as within contaminated food, contributes to “naïve” non-HA pathogen passage of the gastric barrier it must be combined with acid adaptation to facilitate high efficiency transmission. Altogether, these data suggest that C. rodentium adapts to the acidic conditions of the colon during colonization, possibly conferring a competitive advantage to initial gut seeding.

Changes to host cell regulation and circulating gastrin levels result in decreased gastric pH following C. rodentium infection

All the above experiments were done by subjecting C. rodentium to the range of intestinal pH found within the naïve murine gut. However, many global gut environmental changes occur during enteric infection. Therefore, we next sought to profile intestinal pH at peak C. rodentium infection to determine if infection influences environmental pH, either as a result of pathogen colonization or an active host response. To test this, we measured gastric and cecal pH of infected mice at an early (day 4) and peak (day 8) infection time point and compared values to the corresponding pH in uninfected control mice. Cecal pH did not change at days 4 or 8 postinfection (pi), despite a high pathogen burden (Fig 6A). However, we found that gastric pH significantly decreased by day 8 pi as compared to uninfected mice, correlating with colonic pathogen burden (Fig 6A and 6B). This was not due to decreased food consumption by the mice (S5A and S5B Fig) [4].

thumbnail
Fig 6. Murine gastric pH decreases at peak infection.

(A) Intestinal pH measurements in the murine stomach and cecum over time post-C. rodentium infection (inoculum dose of 108 CFU). Statistical analysis represents a two-way ANOVA with Dunnett’s multiple comparisons test (N = 8–14). (B) Spearman correlation of stomach content pH and colon colonization (CFU/gram tissue) at days 4 and 8 postinfection. Line represents linear regression +/− 95% CI. P- and rho-values represent Spearman correlation values. (C) Gene expression analysis of host genes associated with stomach parietal cell activation and acid secretion within stomach tissue from naïve and infected mice (N = 4–8). Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. (D) Fasted serum gastrin levels in naïve and day 8-infected mice. Statistical analysis represents an unpaired t test (N = 15–19). Summary data displayed in Fig 6 can be found in S1 Data. CFU, colony-forming unit.

https://doi.org/10.1371/journal.pbio.3002761.g006

To determine whether this decrease in gastric pH was the result of altered acid secretion, we analyzed host gene expression within the stomach tissue of naïve and infected mice at day 8 pi. We observed a nonsignificant increase in ATP4B which encodes the beta subunit of the gastric H+,K+-ATPase, a proton pump responsible for acid secretion in parietal cells (Fig 6C) [64]. We then specifically investigated changes across histamine-, gastrin-, and acetylcholine-mediated pathways of gastric cell activation. We observed a significant increase in HRH2 expression, encoding histamine receptor H2, a G protein-coupled receptor that stimulates gastric acid secretion in response to histamine (Fig 6C) [6466]. Additional genes associated with histamine-mediated acid secretion, such as HDC (encoding the histidine decarboxylase which converts L-histidine to histamine), and PAC1 (encoding a receptor involved in histamine release from gastric enterochromaffin-like cells) remained unaltered (Figs 6C and S5C).

Hormonal activation of gastric acid secretion is facilitated by release of gastrin, a hormone produced by gastric G cells and released into the circulation following food consumption [6466]. We observed a nonsignificant trend towards increased GAST gene expression, which directly encodes gastrin (Fig 6C). Expression of CCK2R, encoding the cholecystokinin B receptor which responds to gastrin, was highly variable in expression among infected mice (S5C Fig). Furthermore, although cholinergic agents also facilitate parietal cell activation, we did not observe changes to the expression of CHRM3, a gene encoding the M3 muscarinic acetylcholine receptor which responds to the release of cholinergic agents (S5C Fig).

We also investigated genes related to negative control of gastric acid secretion, specifically gastric levels of VIP gene expression encoding vasoactive intestinal peptide and the gene encoding its associated receptor in the stomach, VPAC2 [6466]. We found a nonsignificant trend towards decreased levels of VPAC2 (S5D Fig; p = 0.08), indicating that infection potentially leads to dysregulation of multiple systems involved in maintaining gastric pH. Collectively, these data suggest changes to parietal cell activation postinfection.

To assess whether transcript-level expression reflects systemic changes to regulation of gastric pH, we measured fasting serum gastrin levels by ELISA. High levels of circulating gastrin detected in a fasted state are indicative of hypergastrinemia, and gastrin levels are thereby used as a diagnostic marker in humans [65]. Indeed, we found that circulating gastrin levels in C. rodentium-infected mice at day 8 pi were significantly higher than in naïve mice (Fig 6D). Taken together, our data support decreased gastric pH post-C. rodentium infection, resulting from active changes to host acid regulation.

Gastric pH decreases in response to further infection and chemical models of epithelial barrier dysfunction

To further investigate decreased gastric pH as an active host defense mechanism, we next wished to determine whether similar changes could be observed in other enteric infection models. To address this, we infected mice with human pathogen Salmonella enterica serovar Typhimurium and measured gastric pH at peak infection on day 4 pi. We observed a significant decrease in gastric pH in response to S. Typhimurium (Fig 7A) which significantly correlated with pathogen burden (S6A Fig), demonstrating consistency across additional enteric infection models. To determine whether this response is infection specific, we next performed a DSS model of chemically induced colitis. Despite no difference between the pH of control and DSS drinking water (S6B Fig), we again observed a significant decrease in gastric pH following 4 days of chemical administration (Fig 7A). We therefore hypothesized that gastric pH may decrease in response to intestinal damage rather than infection specifically.

thumbnail
Fig 7. Murine gastric pH decreases in additional models of intestinal damage.

(A) Measurements of gastric pH in additional models of intestinal damage: Salmonella enterica serovar Typhimurium typhoid model of infection (Salmonella infection), and the DSS model of colitis (DSS colitis) (N = 10–15). Statistical analysis represents an ordinary one-way ANOVA with Tukey’s multiple comparisons test. (B) Spearman correlation of stomach pH and TJP1 expression within colonic tissues of naïve and infected mice (N = 20). Line represents linear regression +/− 95% CI. P- and rho-values represent Spearman correlation values. (C) Summary of strains used to test the impact of intestinal damage on gastric pH: wild-type C. rodentium DBS100, T3SS-deficient ΔescN C. rodentium, and commensal E. coli Mt1B1. (D) Measurement of gastric pH in mice which are uninfected, or colonized with either wild-type C. rodentium, ΔescN C. rodentium, or commensal isolate E. coli Mt1B1 at an inoculum dose of 2–4 × 109 CFU. Measurements were taken at euthanasia at day 4 postinoculation (N = 4–8). Statistical analysis represents an ordinary one-way ANOVA with Bonferroni’s multiple comparisons test. (E) Gene expression analysis of host genes HRH2 and GAST within stomach tissue from naïve mice and mice colonized with either wild-type C. rodentium, ΔescN C. rodentium, or commensal isolate E. coli Mt1B1 at day 4 postinoculation (N = 4–8). Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. (F) Colonic TJP1 expression in naïve mice and mice colonized with either wild-type C. rodentium, ΔescN C. rodentium, or commensal isolate E. coli Mt1B1 at day 4 postinoculation (N = 4–8). Statistics represent a Kruskal–Wallis test with Dunn’s multiple comparisons test. Summary data displayed in Fig 7 can be found in S1 Data. Created with BioRender.com. CFU, colony-forming unit; DSS, dextran sulfate sodium; T3SS, type III secretion system.

https://doi.org/10.1371/journal.pbio.3002761.g007

To investigate the relationship between intestinal damage and gastric pH, we measured colonic expression of tight junction protein TJP1, a marker of epithelial barrier integrity. Indeed, we observed a positive correlation between TJP1 expression and gastric pH during C. rodentium infection (rho = 0.53, p = 0.02; Figs 7B and S6C), such that lower gastric pH reflected decreased colonic barrier integrity. In contrast, serum levels of calprotectin, a general marker of systemic host inflammatory state altered by infection, did not correlate with gastric pH (rho = −0.299, p = 0.2; S6D and S6E Fig).

To further test the relationship between intestinal damage and gastric pH, we colonized mice with either wild-type C. rodentium, a type III secretion system-deficient mutant strain (ΔescN), or murine commensal E. coli isolate Mt1B1 (Fig 7C). The ΔescN and commensal Mt1B1 strains are capable of causing only a moderate to negligible level of tissue pathology and inflammation upon murine colonization, respectively [35,36,67], as confirmed by inflammatory cytokine levels in the colon (S6F Fig). Due to the inability of the ΔescN to persist within the mouse gut, we sampled mice at 4 days pi when bacterial burdens were expected to be similar across all groups. We found that gastric pH did not decrease in response to commensal E. coli Mt1B1, and that ΔescN inoculation resulted in only a mild, nonsignificant, decrease in gastric pH (Fig 7D). Furthermore, unlike in wild-type C. rodentium infection, colonization rates of ΔescN DBS100 and Mt1B1 did not correlate with gastric pH (S6G Fig), suggesting that bacterial colonization in the absence of tissue pathology does not affect gastric pH.

We further measured expression levels of HRH2, which were found to be significantly lower in response to colonization by the ΔescN strain, with no differences observed between naïve and Mt1B1-colonized mice (Fig 7E). However, we did find elevated expression of GAST in ΔescN colonized mice, likely accounting for the moderate decrease observed in gastric pH (Fig 7E). This increase in GAST expression again corresponded with decreased expression of TJP1 in the colon of mice colonized with the ΔescN strain but not E. coli Mt1B1, further supporting the relationship between colonic epithelial barrier integrity and gastric pH (Fig 7F). Together, these results indicate that decreased gastric pH may be a widespread host response to intestinal damage.

Discussion

Gastrointestinal pH varies along both the human and mouse gut, likely serving as an important indicator of intestinal geography for invading pathogens. Our study reveals that enteric pathogen C. rodentium responds to subtle pH changes representative of gut pH (even changes of <0.5), altering its growth and virulence. Exposure to pH resembling the cecal environment, C. rodentium’s optimal niche, promoted pathogen growth but did not stimulate the expected T3SS-related attachment. Instead, we found that small intestinal pH 4.4 to 5.7 increased expression of T3SS genes. Duodenal pH may signal pathogen entry to the small intestine from the acidic stomach environment, a transition that necessitates attachment to the host epithelium to prevent pathogen expulsion from the GI tract. Interestingly, gastric pH also enhanced epithelial cell attachment, this time through up-regulation of the type IV pilus Kfc (K99 fimbrial-like adhesin), an early attachment factor, rather than the T3SS (Fig 2). Collectively, our data suggest that low pH is a trigger of loose intestinal attachment upon immediate entry to the small intestine, while the higher pH of the large bowel favors pathogen expansion to support a high pathogen load [23].

Unexpectedly, we found that gastric pH was significantly decreased at peak infection (Fig 6). Up-regulation of host genes associated with acid secretion suggests that stomach acidification is an active adaptation of the host. Histamine receptor-2 (HRH2) in particular is associated with gastric ulcers such that HRH2-blockers, such as rantidine (Zantac), are used to treat conditions involving excessive stomach acid production [68,69]. We found that this response may not be pathogen specific, but instead related to intestinal damage, demonstrating the same phenotype during chemically induced colitis but not in response to colonization by commensal E. coli nor by ΔescN C. rodentium lacking key damage-inducing virulence machinery (Fig 7). The host environment has evolved many adaptative responses to clear invading bacteria such as fever, vomiting, and both specific and nonspecific immune responses [29,70]. Decreasing stomach pH may be another important host response for protection against further pathogen ingestion. Surprisingly, downstream cecal pH did not change at peak infection despite high pathogen burdens which were expected to acidify the gut content (Fig 6). Enteric pathogen Clostridium difficile has been shown to actively inhibit ion exchange to promote pathogen-favorable conditions, resulting in more alkaline stool in infected patients [71]. It may be that C. rodentium is similarly able to manipulate ion transport at key niche locations, such as in the cecum, to maintain favorable conditions for pathogen expansion.

While a decrease from an average gastric pH of 4.0 to 3.5 during peak C. rodentium infection may seem inconsequential, we observed significant differences in cellular morphology and metabolic activity between pHs. While pH 3.5 induced small cell size and decreased metabolic activity, a pH increase of only 0.5 resulted in cell elongation and metabolic recovery, and a reduction in intracellular ROS (Figs 3, 4, and S2). Therefore, even incremental host changes may present a formidable barrier to intestinal re-entry. Interestingly, “host-adapted” C. rodentium shed at peak infection demonstrated higher acid tolerance than host-naïve C. rodentium, indicating that the pathogen develops a more robust ATR during passage through the mouse gut, which may be triggered by exposure to colonic pH conditions (Fig 5). Acid adaptation may thereby contribute to the enhanced transmission of host-adapted C. rodentium as compared to host-naïve C. rodentium [62]. Taken together, it is clear that adaptive pH responses occur on both sides to maintain the balance between pathogen and host.

Given the impact of enteric infection on gastric acid secretion in mice, intestinal damage and inflammatory state may have important functional consequences to gut pH homeostasis. Few studies investigate human gastrointestinal pH following infection or other stressors. However, a known association exists between functional dyspepsia (FD) and infectious gastrointestinal disease by bacteria, viruses, and protozoa [72]. FD is a disorder of unknown etiology characterized by stomach pain and other indigestive symptoms, which occasionally overlaps with postinfectious irritable bowel syndrome [73,74]. A meta-analysis of 17 clinical studies found onset of postinfectious FD in 9.5% of adult subjects, indicating a 2.5-fold risk of FD following acute gastroenteritis [72]. Therefore, further understanding of gastrointestinal pH in response to environmental triggers or altered host health, such as infectious disease state, could be important to predicting susceptibility to secondary intestinal disease.

In summary, our study illustrates how regional GI fluctuations in pH signal changes to C. rodentium behavior and virulence, with consequences to bacterial stress and survival. We further observed infection-induced alterations to host pH which could impact the bioavailability and effectiveness of pH-dependent release drugs, or colonization of probiotic bacterial strains for the treatment of intestinal morbidities. As such, there is immense value in understanding the complex and long-lasting interactions of pathogens and their hosts.

Supporting information

S1 Fig. Effect of standard mouse chow on intestinal pH of C57BL/6 mice.

(A) Murine intestinal pH after 2 weeks on either standard Teklad 18% protein mouse chow or Picolab 21% protein mouse chow. Statistical analysis represents a mixed-effects model with Geisser–Greenhouse correction for matched values and Tukey’s multiple comparisons test (N = 6). (B) pH of Teklad 18% protein standard mouse chow compared to Picolab 21% protein standard mouse chow. Statistical analysis represents an unpaired t test (N = 3 pellets, average of 3 pH readings). (C) Murine intestinal pH of C57BL/6 and BALB/c mice after 2 weeks on Picolab 21% protein mouse chow. Statistical analysis represents a mixed-effects model with Geisser–Greenhouse correction for matched values and Tukey’s multiple comparisons test (N = 8–18). Summary data displayed in S1 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s001

(TIF)

S2 Fig. Similar patterns of growth observed in human pathogens EHEC, EPEC, and S. Typhimurium at physiological pH.

Growth of (A) enterohemorrhagic E. coli (EHEC), (B) enteropathogenic E. coli (EPEC), and (C) S. Typhimurium at physiological pH over 20 h, indicating that all 3 pathogens are unable to grow at pH 3.5. Data represent 3 biological replicates (the average of 3 technical replicates). Error represents mean +/− SD. (D) Biofilm opacity measured by optical density (OD) at 600 nm after 48 h of static growth seeded at physiological pH (N = 6, each the sum of 3 technical replicates). Statistical analysis represents a Friedman test with Dunn’s multiple comparisons test. (E) Metabolic activity within the biofilm after 48 h of static growth seeded at physiological pH, measured by the production of formazan from MTT at OD 570 nm. Error represents mean +/− SD. (F) Growth of C. rodentium at physiological pH in DMEM base media over 20 h. Data represent 3 biological replicates (the average of 3 technical replicates). Error represents mean +/− SD. (G) Attachment of C. rodentium to CMT-93 murine colonic epithelial cells after preinduction in DMEM base media adjusted to gastrointestinal pH. Data represent biological replicates (the average of 3 technical replicates). (H) Attachment of wild-type (WT) or T3SS-deficient (ΔescN) C. rodentium to colonic epithelial cells after preinduction at gastrointestinal pH. Data represent biological replicates (the average of 3 technical replicates). (I) Representative images of actin pedestal formation on CMT-93 cells following infection with C. rodentium pre-induced at pH 4.8 and pH 7.0 (N = 3 biological replicates). Scale bar represents 30 μm. Summary data displayed in S2 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s002

(TIF)

S3 Fig. Homology to the E. coli CadA lysine decarboxylase.

(A) Protein sequence homology between C. rodentium LdcC and E. coli CadA. (B) Survival of wild-type and ΔldcC C. rodentium strains in gastric pH 3.5 over the range of average gastric emptying time. Each point represents the average of 3 biological replicates (3 technical replicates per biological replicate). Statistical analysis represents a two-way ANOVA with Geisser–Greenhouse correction. Summary data displayed in S3 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s003

(TIF)

S4 Fig. C. rodentium survival at gastric pH.

(A) Survival of C. rodentium pre-induced aerobically at either neutral pH 7 or colonic pH 5.7 for 3.5 h (to mid-log phase) before exposure to gastric pH 3.5. Each point represents the average of 3 biological replicates (3 technical replicates per biological replicate). Statistics represent a mixed-effects model with Geisser–Greenhouse correction and Šidák’s multiple comparisons test. Error represents mean +/− SD. (B) Survival of C. rodentium shed in the feces on day 2 postinfection (pi) at gastric pH 3.5. Fecal pellets were not disrupted before submersion in LB pH 3.5. Statistical analysis represents a Mann–Whitney test (N = 6–7). Summary data displayed in S4 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s004

(TIF)

S5 Fig. Factors involved in gastric acid secretion postinfection.

(A) Spearman correlation of stomach pH and weight of stomach contents (N = 23). Line represents linear regression +/− 95% confidence interval. p- and rho-values represent Spearman correlation values. (B) Representative stomach images from fasted control and day 8-infected mice. Scale bar represents 1 cm. (C) Normalized fold expression of genes associated with host regulation of acid secretion within stomach tissue from naïve and infected mice (N = 4–8). Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. (D) Normalized fold expression of genes associated with host negative regulation of acid secretion within stomach tissue from naïve and infected mice (N = 7–8). Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. Summary data displayed in S5 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s005

(TIF)

S6 Fig. The relationship of stomach pH with infection, epithelial barrier integrity, and inflammation.

(A) Spearman correlation of stomach pH and fecal burden of Salmonella Typhimurium at day 4 of a murine typhoid infection model (N = 14). Line represents linear regression +/− 95% confidence interval. p- and rho-values represent Spearman correlation values. (B) pH of drinking water used in the DSS-colitis model. (C) Colonic expression of TJP1 in naïve and C. rodentium-infected mice at day 8 postinfection (N = 4–8). Statistics represent a Mann–Whitney test. (D) Serum calprotectin levels measured by ELISA in naïve and C. rodentium-infected mice at day 8 postinfection (N = 4–8). Statistics represent a Mann–Whitney test. (E) Spearman correlation of stomach pH and serum calprotectin levels of naïve and infected mice (N = 20). Line represents linear regression +/− 95% confidence interval. p- and rho-values represent Spearman correlation values. (F) Cytokine levels measured in the colons of naïve mice or mice colonized with ΔescN C. rodentium, commensal E. coli Mt1B1, or wild-type (WT) C. rodentium at day 4 postinoculation, or WT C. rodentium at day 8 postinfection, or mice treated with dextran sodium sulfate (DSS) to induce colitis (N = 4–8). (G) Spearman correlation of colonization levels by avirulent ΔescN C. rodentium and commensal isolate E. coli Mt1B1 with stomach pH (N = 10). Line represents linear regression +/− 95% confidence interval. p- and rho-values represent Spearman correlation values. (H) Cecum and colon bacterial burden at day 4 post-colonization with WT C. rodentium, ΔescN C. rodentium, or commensal E. coli Mt1B1. Statistics represent a two-way ANOVA with Šidák’s multiple comparisons test. Summary data displayed in S6 Fig can be found in S1 Data.

https://doi.org/10.1371/journal.pbio.3002761.s006

(TIF)

S1 Data. Raw data used for the generation of graphs presented in Figs 17 and S1S6.

https://doi.org/10.1371/journal.pbio.3002761.s007

(XLSX)

S1 Uncropped Images. Raw uncropped images representing the analysis shown in Figs 2, 4, S2, and S5.

https://doi.org/10.1371/journal.pbio.3002761.s008

(PDF)

S1 Table. Bacterial strains used in this study.

https://doi.org/10.1371/journal.pbio.3002761.s009

(DOCX)

S2 Table. Oligonucleotide primers and plasmids used for making deletion constructs of C. rodentium ldcC.

https://doi.org/10.1371/journal.pbio.3002761.s010

(DOCX)

S3 Table. Oligonucleotide primers used for RT-qPCR.

https://doi.org/10.1371/journal.pbio.3002761.s011

(DOCX)

Acknowledgments

The authors thank all of our colleagues in the Finlay laboratory for their support and assistance, especially L Thorson and T Bozorgmehr. Supporting images were created with BioRender.com under agreements HD270MIETZ (Fig 1A) and RE270RB9H8 (Fig 7C). Imaging was performed in the LSI Imaging Core Facility (RRID:SCR_023783) at the University of British Columbia.

References

  1. 1. Fallingborg J. Intraluminal pH of the human gastrointestinal tract. Dan Med Bull. 1999 Jun;46(3):183–96. pmid:10421978
  2. 2. Nugent SG, Kumar D, Rampton DS, Evans DF. Intestinal luminal pH in inflammatory bowel disease: possible determinants and implications for therapy with aminosalicylates and other drugs. Gut. 2001 Apr;48(4):571–7. pmid:11247905
  3. 3. Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ, Hardcastle JD. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut. 1988 Aug;29(8):1035–41. pmid:3410329
  4. 4. McConnell EL, Basit AW, Murdan S. Measurements of rat and mouse gastrointestinal pH, fluid and lymphoid tissue, and implications for in-vivo experiments. J Pharm Pharmacol. 2008 Jan;60(1):63–70. pmid:18088506
  5. 5. Laubitz D, Harrison CA, Midura-Kiela MT, Ramalingam R, Larmonier CB, Chase JH, et al. Reduced Epithelial Na+/H+ Exchange Drives Gut Microbial Dysbiosis and Promotes Inflammatory Response in T Cell-Mediated Murine Colitis. PLoS ONE. 2016 Apr 6;11(4):e0152044. pmid:27050757
  6. 6. Laubitz D, Larmonier CB, Bai A, Midura-Kiela MT, Lipko MA, Thurston RD, et al. Colonic gene expression profile in NHE3-deficient mice: evidence for spontaneous distal colitis. Am J Physiol Gastrointest Liver Physiol. 2008 Jul;295(1):G63–77. pmid:18467500
  7. 7. Engevik AC, Engevik MA. Exploring the impact of intestinal ion transport on the gut microbiota. Comput Struct Biotechnol J. 2021 Jan 1;19:134–44. pmid:33425246
  8. 8. Duncan SH, Louis P, Thomson JM, Flint HJ. The role of pH in determining the species composition of the human colonic microbiota. Environ Microbiol. 2009 Aug;11(8):2112–22. pmid:19397676
  9. 9. Walker AW, Duncan SH, McWilliam Leitch EC, Child MW, Flint HJ. pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl Environ Microbiol. 2005 Jul;71(7):3692–700. pmid:16000778
  10. 10. Ilhan ZE, Marcus AK, Kang DW, Rittmann BE, Krajmalnik-Brown R. pH-Mediated Microbial and Metabolic Interactions in Fecal Enrichment Cultures. mSphere. 2017 May 3;2(3):e00047–17. pmid:28497116
  11. 11. Croxen MA, Law RJ, Scholz R, Keeney KM, Wlodarska M, Finlay BB. Recent Advances in Understanding Enteric Pathogenic Escherichia coli. Clin Microbiol Rev. 2013 Jan 10;26(4):822–80. pmid:24092857
  12. 12. Scheiring J, Andreoli SP, Zimmerhackl LB. Treatment and outcome of Shiga-toxin-associated hemolytic uremic syndrome (HUS). Pediatr Nephrol Berl Ger. 2008;23(10):1749. pmid:18704506
  13. 13. Schauer DB, Falkow S. Attaching and effacing locus of a Citrobacter freundii biotype that causes transmissible murine colonic hyperplasia. Infect Immun. 1993 Jan 6;61(6):2486–92. pmid:8500884
  14. 14. Bhinder G, Sham HP, Chan JM, Morampudi V, Jacobson K, Vallance BA. The Citrobacter rodentium Mouse Model: Studying Pathogen and Host Contributions to Infectious Colitis. J Vis Exp JoVE. 2013 Feb 19;(72):50222. pmid:23462619
  15. 15. Crepin VF, Collins JW, Habibzay M, Frankel G. Citrobacter rodentium mouse model of bacterial infection. Nat Protoc. 2016 Oct;11(10):1851–76. pmid:27606775
  16. 16. Barthold SW, Coleman GL, Bhatt PN, Osbaldiston GW, Jonas AM. The etiology of transmissible murine colonic hyperplasia. Lab Anim Sci. 1976 Dec;26(6 Pt 1):889–94. pmid:1018473
  17. 17. Deng W, Vallance BA, Li Y, Puente JL, Finlay BB. Citrobacter rodentium translocated intimin receptor (Tir) is an essential virulence factor needed for actin condensation, intestinal colonization and colonic hyperplasia in mice. Mol Microbiol. 2003 Apr;48(1):95–115. pmid:12657048
  18. 18. Elliott SJ, Yu J, Kaper JB. The cloned locus of enterocyte effacement from enterohemorrhagic Escherichia coli O157:H7 is unable to confer the attaching and effacing phenotype upon E. coli K-12. Infect Immun. 1999 Aug;67(8):4260–3. pmid:10417201
  19. 19. Mellies JL, Elliott SJ, Sperandio V, Donnenberg MS, Kaper JB. The Per regulon of enteropathogenic Escherichia coli: identification of a regulatory cascade and a novel transcriptional activator, the locus of enterocyte effacement (LEE)-encoded regulator (Ler). Mol Microbiol. 1999 Jul;33(2):296–306. pmid:10411746
  20. 20. Jarvis KG, Girón JA, Jerse AE, McDaniel TK, Donnenberg MS, Kaper JB. Enteropathogenic Escherichia coli contains a putative type III secretion system necessary for the export of proteins involved in attaching and effacing lesion formation. Proc Natl Acad Sci U S A. 1995 Aug 15;92(17):7996–8000. pmid:7644527
  21. 21. Moon HW, Whipp SC, Argenzio RA, Levine MM, Giannella RA. Attaching and effacing activities of rabbit and human enteropathogenic Escherichia coli in pig and rabbit intestines. Infect Immun. 1983 Sep;41(3):1340–51. pmid:6350186
  22. 22. Deng W, Marshall NC, Rowland JL, McCoy JM, Worrall LJ, Santos AS, et al. Assembly, structure, function and regulation of type III secretion systems. Nat Rev Microbiol. 2017 Jun;15(6):323–37. pmid:28392566
  23. 23. Woodward SE, Vogt SL, Peña-Díaz J, Melnyk RA, Cirstea M, Serapio-Palacios A, et al. Gastric acid and escape to systemic circulation represent major bottlenecks to host infection by Citrobacter rodentium. ISME J. 2022 Sep 24;1–11. pmid:36153406
  24. 24. Jubelin G, Desvaux M, Schüller S, Etienne-Mesmin L, Muniesa M, Blanquet-Diot S. Modulation of Enterohaemorrhagic Escherichia coli Survival and Virulence in the Human Gastrointestinal Tract. Microorganisms. 2018 Nov 19;6(4). pmid:30463258
  25. 25. Kenny B, Abe A, Stein M, Finlay BB. Enteropathogenic Escherichia coli protein secretion is induced in response to conditions similar to those in the gastrointestinal tract. Infect Immun. 1997 Jul;65(7):2606–12. pmid:9199427
  26. 26. Pienaar JA, Singh A, Barnard TG. Acid-happy: Survival and recovery of enteropathogenic Escherichia coli (EPEC) in simulated gastric fluid. Microb Pathog. 2019 Jan 17;128:396–404. pmid:30660737
  27. 27. Yang J, Tauschek M, Hart E, Hartland EL, Robins-Browne RM. Virulence regulation in Citrobacter rodentium: the art of timing. J Microbial Biotechnol. 2010 May 1;3(3):259–68. pmid:21255326
  28. 28. Hart E, Yang J, Tauschek M, Kelly M, Wakefield MJ, Frankel G, et al. RegA, an AraC-Like Protein, Is a Global Transcriptional Regulator That Controls Virulence Gene Expression in Citrobacter rodentium. Infect Immun. 2008 Jan 11;76(11):5247–56. pmid:18765720
  29. 29. Woodward SE, Krekhno Z, Finlay BB. Here, there, and everywhere: How pathogenic Escherichia coli sense and respond to gastrointestinal biogeography. Cell Microbiol. 2019 Nov;21(11):e13107. pmid:31454133
  30. 30. Schauer DB, Zabel BA, Pedraza IF, O’Hara CM, Steigerwalt AG, Brenner DJ. Genetic and biochemical characterization of Citrobacter rodentium sp. nov. J Clin Microbiol. 1995 Aug;33(8):2064–8. pmid:7559949
  31. 31. Riley LW, Remis RS, Helgerson Steven D, McGee HB, Wells JG, Davis BR, et al. Hemorrhagic Colitis Associated with a Rare Escherichia coli Serotype. N Engl J Med. 1983 Mar 24;308(12):681–5. pmid:6338386
  32. 32. Levine MM, Bergquist EJ, Nalin DR, Waterman DH, Hornick RB, Young CR, et al. Escherichia coli strains that cause diarrhoea but do not produce heat-labile or heat-stable enterotoxins and are non-invasive. Lancet Lond Engl. 1978 May 27;1(8074):1119–22. pmid:77415
  33. 33. Edwards RA, Keller LH, Schifferli DM. Improved allelic exchange vectors and their use to analyze 987P fimbria gene expression. Gene. 1998 Jan 30;207(2):149–57. pmid:9511756
  34. 34. Ferrières L, Hémery G, Nham T, Guérout AM, Mazel D, Beloin C, et al. Silent mischief: bacteriophage Mu insertions contaminate products of Escherichia coli random mutagenesis performed using suicidal transposon delivery plasmids mobilized by broad-host-range RP4 conjugative machinery. J Bacteriol. 2010 Dec;192(24):6418–27. pmid:20935093
  35. 35. Deng W, Puente JL, Gruenheid S, Li Y, Vallance BA, Vázquez A, et al. Dissecting virulence: systematic and functional analyses of a pathogenicity island. Proc Natl Acad Sci U S A. 2004 Mar 9;101(10):3597–602. pmid:14988506
  36. 36. Serapio-Palacios A, Woodward SE, Vogt SL, Deng W, Creus-Cuadros A, Huus KE, et al. Type VI secretion systems of pathogenic and commensal bacteria mediate niche occupancy in the gut. Cell Rep. 2022 Apr 26;39(4):110731. pmid:35476983
  37. 37. Stirling DR, Swain-Bowden MJ, Lucas AM, Carpenter AE, Cimini BA, Goodman A. CellProfiler 4: improvements in speed, utility and usability. BMC Bioinformatics. 2021 Sep 10;22(1):433. pmid:34507520
  38. 38. Stirling DR, Carpenter AE, Cimini BA. CellProfiler Analyst 3.0: accessible data exploration and machine learning for image analysis. Bioinformatics. 2021 Nov 1;37(21):3992–4. pmid:34478488
  39. 39. Thomassin JL, Giannakopoulou N, Zhu L, Gross J, Salmon K, Leclerc JM, et al. The CpxRA Two-Component System Is Essential for Citrobacter rodentium Virulence. Infect Immun. 2015 May;83(5):1919–28. pmid:25712925
  40. 40. van der Heijden J, Finlay BB. In vitro Real-time Measurement of the Intra-bacterial Redox Potential. Bio-Protoc. 2015 Sep 5;5(17):e1579. pmid:27617271
  41. 41. van der Heijden J, Vogt SL, Reynolds LA, Peña-Díaz J, Tupin A, Aussel L, et al. Exploring the redox balance inside gram-negative bacteria with redox-sensitive GFP. Free Radic Biol Med. 2016 Feb 1;91:34–44. pmid:26627936
  42. 42. LabDiet—Irradiated Diets [Internet]. [cited 2022 Sep 16]. Available from: https://www.labdiet.com/Products/Irradiated/index.htm.
  43. 43. Cummings JH, Macfarlane GT. The control and consequences of bacterial fermentation in the human colon. J Appl Bacteriol. 1991;70(6):443–59.
  44. 44. Shimizu K, Seiki I, Goto Y, Murata T. Measurement of the Intestinal pH in Mice under Various Conditions Reveals Alkalization Induced by Antibiotics. Antibiot Basel Switz. 2021 Feb 11;10(2):180. pmid:33670214
  45. 45. Engevik MA, Aihara E, Montrose MH, Shull GE, Hassett DJ, Worrell RT. Loss of NHE3 alters gut microbiota composition and influences Bacteroides thetaiotaomicron growth. Am J Physiol Gastrointest Liver Physiol. 2013 Nov 15;305(10):G697–711. pmid:24072680
  46. 46. Flagella M, Clarke LL, Miller ML, Erway LC, Giannella RA, Andringa A, et al. Mice lacking the basolateral Na-K-2Cl cotransporter have impaired epithelial chloride secretion and are profoundly deaf. J Biol Chem. 1999 Sep 17;274(38):26946–55. pmid:10480906
  47. 47. Takumi K, de Jonge R, Havelaar A. Modelling inactivation of Escherichia coli by low pH: application to passage through the stomach of young and elderly people. J Appl Microbiol. 2000 Dec;89(6):935–43. pmid:11123466
  48. 48. Flemming HC, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S. Biofilms: an emergent form of bacterial life. Nat Rev Microbiol. 2016 Sep;14(9):563–75. pmid:27510863
  49. 49. Collins JW, Keeney KM, Crepin VF, Rathinam VAK, Fitzgerald KA, Finlay BB, et al. Citrobacter rodentium: infection, inflammation and the microbiota. Nat Rev Microbiol. 2014 Sep;12(9):612–23. pmid:25088150
  50. 50. Sinclair JF, Dean-Nystrom EA, O’Brien AD. The Established Intimin Receptor Tir and the Putative Eucaryotic Intimin Receptors Nucleolin and β1 Integrin Localize at or near the Site of Enterohemorrhagic Escherichia coli O157:H7 Adherence to Enterocytes In Vivo. Infect Immun. 2006 Feb;74(2):1255–65.
  51. 51. Vogt SL, Scholz R, Peng Y, Guest RL, Scott NE, Woodward SE, et al. Characterization of the Citrobacter rodentium Cpx regulon and its role in host infection. Mol Microbiol. 2019 Mar;111(3):700–16. pmid:30536519
  52. 52. Begley M, Hill C. Stress adaptation in foodborne pathogens. Annu Rev Food Sci Technol. 2015;6:191–210. pmid:25665171
  53. 53. Hughes DT, Terekhova DA, Liou L, Hovde CJ, Sahl JW, Patankar AV, et al. Chemical sensing in mammalian host–bacterial commensal associations. Proc Natl Acad Sci U S A. 2010 May 25;107(21):9831–6. pmid:20457895
  54. 54. Van Houdt R, Aertsen A, Moons P, Vanoirbeek K, Michiels CW. N-acyl-L-homoserine lactone signal interception by Escherichia coli. FEMS Microbiol Lett. 2006 Mar;256(1):83–9. pmid:16487323
  55. 55. Small P, Blankenhorn D, Welty D, Zinser E, Slonczewski JL. Acid and base resistance in Escherichia coli and Shigella flexneri: role of rpoS and growth pH. J Bacteriol. 1994 Mar;176(6):1729–37. pmid:8132468
  56. 56. Dong T, Coombes BK, Schellhorn HE. Role of RpoS in the virulence of Citrobacter rodentium. Infect Immun. 2009 Jan;77(1):501–7. pmid:18981255
  57. 57. Price SB, Cheng CM, Kaspar CW, Wright JC, DeGraves FJ, Penfound TA, et al. Role of rpoS in Acid Resistance and Fecal Shedding of Escherichia coli O157:H7. Appl Environ Microbiol. 2000 Feb;66(2):632–7. pmid:10653728
  58. 58. Cox MM. Regulation of bacterial RecA protein function. Crit Rev Biochem Mol Biol. 2007 Feb;42(1):41–63. pmid:17364684
  59. 59. Jones CH, Danese PN, Pinkner JS, Silhavy TJ, Hultgren SJ. The chaperone-assisted membrane release and folding pathway is sensed by two signal transduction systems. EMBO J. 1997 Nov 3;16(21):6394–406. pmid:9351822
  60. 60. Danese PN, Snyder WB, Cosma CL, Davis LJ, Silhavy TJ. The Cpx two-component signal transduction pathway of Escherichia coli regulates transcription of the gene specifying the stress-inducible periplasmic protease, DegP Genes Dev. 1995 Feb 15;9(4):387–98.
  61. 61. Schwarz R, Kaspar A, Seelig J, Künnecke B. Gastrointestinal transit times in mice and humans measured with 27Al and 19F nuclear magnetic resonance. Magn Reson Med. 2002 Aug;48(2):255–61. pmid:12210933
  62. 62. Wiles S, Dougan G, Frankel G. Emergence of a ‘hyperinfectious’ bacterial state after passage of Citrobacter rodentium through the host gastrointestinal tract. Cell Microbiol. 2005 Aug 1;7(8):1163–72. pmid:16008583
  63. 63. Wiles S, Clare S, Harker J, Huett A, Young D, Dougan G, et al. Organ specificity, colonization and clearance dynamics in vivo following oral challenges with the murine pathogen Citrobacter rodentium. Cell Microbiol. 2004 Oct 1;6(10):963–72. pmid:15339271
  64. 64. Hunt RH, Camilleri M, Crowe SE, El-Omar EM, Fox JG, Kuipers EJ, et al. The stomach in health and disease. Gut. 2015 Oct;64(10):1650–68. pmid:26342014
  65. 65. Dacha S, Razvi M, Massaad J, Cai Q, Wehbi M. Hypergastrinemia. Gastroenterol Rep. 2015 Aug;3(3):201–8. pmid:25698559
  66. 66. Phan J, Benhammou JN, Pisegna JR. Gastric Hypersecretory States: Investigation and Management. Curr Treat Options Gastroenterol. 2015 Dec;13(4):386–97. pmid:26342486
  67. 67. Lopez CA, Miller BM, Rivera-Chávez F, Velazquez EM, Byndloss MX, Chávez-Arroyo A, et al. Virulence factors enhance Citrobacter rodentium expansion through aerobic respiration. Science. 2016 16;353(6305):1249–53. pmid:27634526
  68. 68. Marquez-Gomez PL, Kruyer NS, Eisen SL, Torp LR, Howie RL, Jones EV, et al. Discovery of 8-Hydroxyquinoline as a Histamine Receptor 2 Blocker Scaffold. ACS Synth Biol. 2022 Aug 19;11(8):2820–8. pmid:35930594
  69. 69. Samuelson LC, Hinkle KL. Insights into the Regulation of Gastric Acid Secretion Through Analysis of Genetically Engineered Mice. Annu Rev Physiol. 2003;65(1):383–400. pmid:12517996
  70. 70. Fink SL, Campbell S. Chapter 3—Infection and Host Response. In: Coleman WB, Tsongalis GJ, editors. Molecular Pathology (Second Edition). Academic Press; 2018. p. 45–69. Available from: https://www.sciencedirect.com/science/article/pii/B9780128027615000031.
  71. 71. Engevik MA, Engevik KA, Yacyshyn MB, Wang J, Hassett DJ, Darien B, et al. Human Clostridium difficile infection: inhibition of NHE3 and microbiota profile. Am J Physiol Gastrointest Liver Physiol. 2015 Mar 15;308(6):G497–509. pmid:25552580
  72. 72. Futagami S, Itoh T, Sakamoto C. Systematic review with meta-analysis: post-infectious functional dyspepsia. Aliment Pharmacol Ther. 2015;41(2):177–88. pmid:25348873
  73. 73. Tuteja AK, Talley NJ, Gelman SS, Alder SC, Adler SC, Thompson C, et al. Development of functional diarrhea, constipation, irritable bowel syndrome, and dyspepsia during and after traveling outside the USA. Dig Dis Sci. 2008 Jan;53(1):271–6. pmid:17549631
  74. 74. Spiller R. Postinfectious functional dyspepsia and postinfectious irritable bowel syndrome: different symptoms but similar risk factors. Gastroenterology. 2010 May;138(5):1660–3. pmid:20346343