Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
bioRxiv ; 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38405862

ABSTRACT

Crohn's disease (CD) is the chronic inflammation of the ileum and colon triggered by bacteria, but insights into molecular perturbations at the bacteria-epithelium interface are limited. We report that membrane mucin MUC17 protects small intestinal enterocytes against commensal and pathogenic bacteria. In non-inflamed CD ileum, reduced MUC17 levels correlated with a compromised glycocalyx, allowing bacterial contact with enterocytes. Muc17 deletion in mice rendered the small intestine prone to atypical infection while maintaining resistance to colitis. The loss of Muc17 resulted in spontaneous deterioration of epithelial homeostasis and extra-intestinal translocation of bacteria. Finally, Muc17-deficient mice harbored specific small intestinal bacterial taxa observed in CD. Our findings highlight MUC17 as an essential line of defense in the small intestine with relevance for early epithelial defects in CD.

2.
J Crohns Colitis ; 18(2): 233-245, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-37602969

ABSTRACT

AIM: To assess contemporary outcomes in children with acute severe ulcerative colitis [ASUC] at initial presentation. METHODS: Between April 2014 and January 2019, children aged <17 years, with new onset ASUC (Paediatric Ulcerative Colitis Activity Index [PUCAI ≥65) were prospectively followed in a Canadian inception cohort study. 16S rRNA amplicon sequencing captured microbial composition of baseline faecal samples. Primary endpoint was corticosteroid-free clinical remission with intact colon at 1 year [PUCAI <10, no steroids ≥4 weeks]. RESULTS: Of 379 children with new onset UC/IBD-unclassified, 105 [28%] presented with ASUC (42% male; median [interquartile range; [IQR]) age 14 [11-16] years; extensive colitis in all). Compared with mild UC, gut microbiome of ASUC patients had lower α-diversity, decreased beneficial anaerobes, and increased aerobes; 54 [51%] children were steroid-refractory and given infliximab [87% intensified regimen]. Corticosteroid-free remission at 1 year was achieved by 62 [61%] ASUC cohort (by 34 [63%] steroid-refractory patients, all on biologics; by 28 [55%] steroid responders,13 [25%] on 5- aminosalicylic acid [5-ASA], 5 [10%] on thiopurines, 10 [20%] on biologics). By 1 year, 78 [74%] escalated to infliximab including 24 [47%] steroid-responders failed by 5-ASA and/or thiopurines. In multivariable analysis, clinical predictors for commencing infliximab included hypoalbuminaemia, greater PUCAI, higher age, and male sex. Over 18 months, repeat corticosteroid course[s] and repeat hospitalisation were less likely among steroid-refractory versus -responsive but -dependent patients (adjusted odds ratio [aOR] 0.71 [95% CI 0.57-0.89] and 0.54 [95% CI 0.45-0.66], respectively). CONCLUSION: The majority of children presenting with ASUC escalate therapy to biologics. Predictors of need for advanced therapy may guide selection of optimal maintenance therapy.


Subject(s)
Biological Products , Colitis, Ulcerative , Humans , Child , Male , Female , Infliximab/therapeutic use , Cohort Studies , Prospective Studies , RNA, Ribosomal, 16S , Canada , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Mesalamine/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Steroids/therapeutic use , Biological Products/therapeutic use , Treatment Outcome
3.
Gastroenterology ; 166(1): 44-58, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37734419

ABSTRACT

The gut microbiota plays a significant role in the pathogenesis of both forms of inflammatory bowel disease (IBD), namely, Crohn's disease (CD) and ulcerative colitis (UC). Although evidence suggests dysbiosis and loss of beneficial microbial species can exacerbate IBD, many new studies have identified microbes with pathogenic qualities, termed "pathobionts," within the intestines of patients with IBD. The concept of pathobionts initiating or driving the chronicity of IBD has largely focused on the putative aggravating role that adherent invasive Escherichia coli may play in CD. However, recent studies have identified additional bacterial and fungal pathobionts in patients with CD and UC. This review will highlight the characteristics of these pathobionts and their implications for IBD treatment. Beyond exploring the origins of pathobionts, we discuss those associated with specific clinical features and the potential mechanisms involved, such as creeping fat (Clostridium innocuum) and impaired wound healing (Debaryomyces hansenii) in patients with CD as well as the increased fecal proteolytic activity (Bacteroides vulgatus) seen as a biomarker for UC severity. Finally, we examine the potential impact of pathobionts on current IBD therapies, and several new approaches to target pathobionts currently in the early stages of development. Despite recognizing that pathobionts likely contribute to the pathogenesis of IBD, more work is needed to define their modes of action. Determining whether causal relationships exist between pathobionts and specific disease characteristics could pave the way for improved care for patients, particularly for those not responding to current IBD therapies.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/microbiology , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Intestines , Feces
4.
Infect Immun ; 91(11): e0032223, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37800916

ABSTRACT

One of the major contributors to child mortality in the world is diarrheal diseases, with an estimated 800,000 deaths per year. Many pathogens are causative agents of these illnesses, including the enteropathogenic or enterohemorrhagic forms of Escherichia coli. These bacteria are characterized by their ability to cause attaching and effacing lesions in the gut mucosa. Although much has been learned about the pathogenicity of these organisms and the immune response against them, the role of the intestinal microbiota during these infections is not well characterized. Infection of mice with E. coli requires pre-treatment with antibiotics in most mouse models, which hinders the study of the microbiota in an undisturbed environment. Using Citrobacter rodentium as a murine model for attaching and effacing bacteria, we show that C57BL/6 mice deficient in granzyme B expression are highly susceptible to severe disease caused by C. rodentium infection. Although a previous publication from our group shows that granzyme B-deficient CD4+ T cells are partially responsible for this phenotype, in this report, we present data demonstrating that the microbiota, in particular members of the order Turicibacterales, have an important role in conferring resistance. Mice deficient in Turicibacter sanguinis have increased susceptibility to severe disease. However, when these mice are co-housed with resistant mice or colonized with T. sanguinis, susceptibility to severe infection is reduced. These results clearly suggest a critical role for this commensal in the protection against enteropathogens.


Subject(s)
Enterobacteriaceae Infections , Escherichia coli , Child , Humans , Animals , Mice , Citrobacter rodentium/genetics , Granzymes , Enterobacteriaceae Infections/microbiology , Mice, Inbred C57BL , Bacteria
5.
Proc Natl Acad Sci U S A ; 120(28): e2301115120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37399418

ABSTRACT

Enteric bacterial pathogens pose significant threats to human health; however, the mechanisms by which they infect the mammalian gut in the face of daunting host defenses and an established microbiota remain poorly defined. For the attaching and effacing (A/E) bacterial family member and murine pathogen Citrobacter rodentium, its virulence strategy likely involves metabolic adaptation to the host's intestinal luminal environment, as a necessary precursor to reach and infect the mucosal surface. Suspecting this adaptation involved the intestinal mucus layer, we found that C. rodentium was able to catabolize sialic acid, a monosaccharide derived from mucins, and utilize it as its sole carbon source for growth. Moreover, C. rodentium also sensed and displayed chemotactic activity toward sialic acid. These activities were abolished when the nanT gene, encoding a sialic acid transporter, was deleted (ΔnanT). Correspondingly, the ΔnanT C. rodentium strain was significantly impaired in its ability to colonize the murine intestine. Intriguingly, sialic acid was also found to induce the secretion of two autotransporter proteins, Pic and EspC, which possess mucinolytic and host-adherent properties. As a result, sialic acid enhanced the ability of C. rodentium to degrade intestinal mucus (through Pic), as well as to adhere to intestinal epithelial cells (through EspC). We thus demonstrate that sialic acid, a monosaccharide constituent of the intestinal mucus layer, functions as an important nutrient and a key signal for an A/E bacterial pathogen to escape the colonic lumen and directly infect its host's intestinal mucosa.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Animals , Mice , Bacteria , Citrobacter , Enterobacteriaceae Infections/microbiology , Intestinal Mucosa/microbiology , Mammals , Monosaccharides , N-Acetylneuraminic Acid
6.
bioRxiv ; 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37163036

ABSTRACT

One of the major contributors to child mortality in the world is diarrheal diseases, with an estimated 800,000 deaths per year. Many pathogens are causative agents of these illnesses, including the enteropathogenic (EPEC) or enterohemorrhagic (EHEC) forms of Escherichia coli. These bacteria are characterized by their ability to cause attaching and effacing lesions in the gut mucosa. Although much has been learned about the pathogenicity of these organisms and the immune response against them, the role of the intestinal microbiota during these infections is not well characterized. Infection of mice with E. coli requires pre-treatment with antibiotics in most mouse models, which hinders the study of the microbiota in an undisturbed environment. Using Citrobacter rodentium as a murine model for attaching and effacing bacteria, we show that C57BL/6 mice deficient in granzyme B expression are highly susceptible to severe disease caused by C. rodentium infection. Although a previous publication from our group shows that granzyme B-deficient CD4+ T cells are partially responsible for this phenotype, in this report we present data demonstrating that the microbiota, in particular members of the order Turicibacterales, have an important role in conferring resistance. Mice deficient in Turicibacter sanguinis have increased susceptibility to severe disease. However, when these mice are co-housed with resistant mice, or colonized with T. sanguinis, susceptibility to severe infection is reduced. These results clearly suggest a critical role for this commensal in the protection against entero-pathogens.

7.
Paediatr Perinat Epidemiol ; 37(6): 516-526, 2023 08.
Article in English | MEDLINE | ID: mdl-36978215

ABSTRACT

BACKGROUND: Prenatal antibiotic exposure induces changes in the maternal microbiome, which could influence the development of the infant's microbiome-gut-brain axis. OBJECTIVES: We assessed whether prenatal antibiotic exposure is associated with an increased risk of autism spectrum disorder (ASD) in offspring born at term. METHODS: This population-based retrospective cohort study included everyone who delivered a live singleton-term infant in British Columbia, Canada between April 2000 and December 2014. Exposure was defined as filling antibiotic prescriptions during pregnancy. The outcome was an ASD diagnosis from the British Columbia Autism Assessment Network, with a follow-up to December 2016. To examine the association among pregnant individuals treated for the same indication, we studied a sub-cohort diagnosed with urinary tract infections. Cox proportional hazards models were used to estimate unadjusted and adjusted hazard ratios (HR). The analysis was stratified by sex, trimester, cumulative duration of exposure, class of antibiotic, and mode of delivery. We ran a conditional logistic regression of discordant sibling pairs to control for unmeasured environmental and genetic confounding. RESULTS: Of the 569,953 children included in the cohort, 8729 were diagnosed with ASD (1.5%) and 169,922 were exposed to prenatal antibiotics (29.8%). Prenatal antibiotic exposure was associated with an increased risk of ASD (HR 1.10, 95% confidence interval [CI] 1.05, 1.15), particularly for exposure during the first and second trimesters (HR 1.11, 95% CI 1.04, 1.18 and HR 1.09, 95% CI 1.03, 1.16, respectively), and exposure lasting ≥15 days (HR 1.13, 95% CI 1.04, 1.23). No sex differences were observed. The association was attenuated in the sibling analysis (adjusted odds ratio 1.04, 95% CI 0.92, 1.17). CONCLUSIONS: Prenatal antibiotic exposure was associated with a small increase in the risk of ASD in offspring. Given the possibility of residual confounding, these results should not influence clinical decisions regarding antibiotic use during pregnancy.


Subject(s)
Autism Spectrum Disorder , Child , Female , Humans , Infant , Pregnancy , Anti-Bacterial Agents/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/epidemiology , Cohort Studies , Retrospective Studies , Term Birth , Prenatal Exposure Delayed Effects
8.
Exp Mol Med ; 55(2): 401-412, 2023 02.
Article in English | MEDLINE | ID: mdl-36759578

ABSTRACT

Endoplasmic reticulum stress is closely associated with the onset and progression of inflammatory bowel disease. ERdj5 is an endoplasmic reticulum-resident protein disulfide reductase that mediates the cleavage and degradation of misfolded proteins. Although ERdj5 expression is significantly higher in the colonic tissues of patients with inflammatory bowel disease than in healthy controls, its role in inflammatory bowel disease has not yet been reported. In the current study, we used ERdj5-knockout mice to investigate the potential roles of ERdj5 in inflammatory bowel disease. ERdj5 deficiency causes severe inflammation in mouse colitis models and weakens gut barrier function by increasing NF-κB-mediated inflammation. ERdj5 may not be indispensable for goblet cell function under steady-state conditions, but its deficiency induces goblet cell apoptosis under inflammatory conditions. Treatment of ERdj5-knockout mice with the chemical chaperone ursodeoxycholic acid ameliorated severe colitis by reducing endoplasmic reticulum stress. These findings highlight the important role of ERdj5 in preserving goblet cell viability and function by resolving endoplasmic reticulum stress.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , HSP40 Heat-Shock Proteins/metabolism , Protein Folding , Goblet Cells/metabolism , Inflammation , Mice, Knockout , Endoplasmic Reticulum Stress , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Apoptosis , Molecular Chaperones/metabolism
9.
Biochim Biophys Acta Mol Basis Dis ; 1869(4): 166647, 2023 04.
Article in English | MEDLINE | ID: mdl-36746254

ABSTRACT

Inflammatory bowel disease (IBD) is an immune-mediated disease. Autotaxin (ATX) is associated with increased inflammatory molecules, however, its effect on IBD is not well understood. Autophagy plays an important role in IBD, whether ATX and autophagy act in concert in IBD remains unknown. This study is to explore the possible mechanisms of ATX affecting autophagy leading to the disruption of intestinal epithelial barrier, thereby exacerbating colitis. The expression of ATX was upregulated in UC patients and dextran sulfate sodium (DSS)-induced colitis mice. Here, we described that providing an ATX inhibitor during DSS colitis increased autophagy and ameliorated colonic inflammation. Conversely, intrarectal administration with recombinant (r)ATX increased colitis and decreased autophagy. This pro-colitic effect was attenuated in mice treated with rapamycin, resulting in increased autophagy activity and mild colitis. Moreover, the inhibitory effect of rATX on autophagy was confirmed in vitro and was reversed by the addition of rapamycin. The damaging effects of ATX on epithelial barrier function were reversed by ATX inhibitor or rapamycin treatment. In sum, our results show that ATX can inhibit autophagy through the mTOR pathway, resulting in exaggerated damage to the intestinal epithelial barrier during colitis. These findings suggest that ATX may be a key pro-colitic factor, and represent a potential therapeutic target for treating IBD in the future.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , Autophagy , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Inflammation , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Sirolimus/pharmacology
11.
Cell Mol Gastroenterol Hepatol ; 15(2): 425-438, 2023.
Article in English | MEDLINE | ID: mdl-36244647

ABSTRACT

BACKGROUND & AIMS: Increased intestinal permeability is seen in a variety of inflammatory conditions such as enteric infections and inflammatory bowel disease. Because barrier function can provide a key biomarker of disease severity, it often is assayed in animal models. A common methodology involves gavaging mice with fluorescein isothiocyanate-conjugated dextran (FITC-D), followed by cardiac puncture to assay plasma fluorescence on a spectrophotometer. Although the FITC-D method is relatively simple, its sensitivity is limited and enables only a single measurement because the test requires killing the subject. Herein, we describe a novel flow cytometry-based method of intestinal permeability measurement based on detection of orally gavaged ovalbumin (OVA) that leaks out of the gut. Our approach uses minute blood volumes collected from the tail vein, permitting repeated testing of the same subject at multiple time points. By comparing this assay against the gold standard FITC-D method, we show the expanded utility of our OVA assay in measuring intestinal permeability. METHODS: We directly compared our OVA assay against the FITC-D assay by co-administering both probes orally to the same animals and subsequently using their respective methodologies to measure intestinal permeability by detecting probe levels in the plasma. Permeability was assessed in mice genetically deficient in intestinal mucus production or glycosylation. In addition, wild-type mice undergoing dextran sodium sulfate-induced colitis or infected by the enteric bacterial pathogen Citrobacter rodentium also were tested. RESULTS: The OVA assay showed very high efficacy in all animal models of intestinal barrier dysfunction tested. Besides identifying intestinal barrier dysfunction in mice with impaired mucin glycosylation, the assay also allowed for repeated tracking of intestinal permeability within the same animal over time, providing data that cannot be easily acquired with other currently applied methods. CONCLUSIONS: The OVA assay is a highly sensitive and effective method of measuring intestinal permeability in mouse models of barrier dysfunction and experimental colitis.


Subject(s)
Colitis , Dextrans , Mice , Animals , Dextrans/adverse effects , Intestinal Mucosa , Flow Cytometry , Fluorescein-5-isothiocyanate/adverse effects , Colitis/chemically induced , Disease Models, Animal , Permeability
12.
Gut Microbes ; 14(1): 2111951, 2022.
Article in English | MEDLINE | ID: mdl-35984746

ABSTRACT

Hyperglycemia and type 2 diabetes (T2D) are caused by failure of pancreatic beta cells. The role of the gut microbiota in T2D has been studied, but causal links remain enigmatic. Obese individuals with or without T2D were included from two independent Dutch cohorts. Human data were translated in vitro and in vivo by using pancreatic islets from C57BL6/J mice and by injecting flagellin into obese mice. Flagellin is part of the bacterial locomotor appendage flagellum, present in gut bacteria including Enterobacteriaceae, which we show to be more abundant in the gut of individuals with T2D. Subsequently, flagellin induces a pro-inflammatory response in pancreatic islets mediated by the Toll-like receptor (TLR)-5 expressed on resident islet macrophages. This inflammatory response is associated with beta-cell dysfunction, characterized by reduced insulin gene expression, impaired proinsulin processing and stress-induced insulin hypersecretion in vitro and in vivo in mice. We postulate that increased systemically disseminated flagellin in T2D is a contributing factor to beta-cell failure in time and represents a novel therapeutic target.


Subject(s)
Diabetes Mellitus, Type 2 , Flagellin , Gastrointestinal Microbiome , Insulin-Secreting Cells , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Diabetes Mellitus, Type 2/microbiology , Flagellin/genetics , Flagellin/metabolism , Humans , Inflammation/metabolism , Insulin , Insulin-Secreting Cells/metabolism , Mice
13.
Pediatrics ; 150(3)2022 09 01.
Article in English | MEDLINE | ID: mdl-36017659

ABSTRACT

OBJECTIVES: Antibiotics are commonly administered during labor and delivery, and research has suggested that fetal exposure to antibiotics can increase risk for autism spectrum disorder (ASD). We assessed whether antibiotic exposure during labor and delivery increased the risk of ASD in the offspring. METHODS: This retrospective cohort study included everyone who delivered a live singleton-term infant in British Columbia, Canada, between April 1, 2000, and December 31, 2014. This cohort included 569 953 deliveries. To examine the association among pregnant individuals being treated for the same indication, we studied a subcohort of those who tested positive for group B Streptococcus. Cox proportional hazards models were used to estimate unadjusted and adjusted hazard ratios in both cohorts. A sensitivity analysis was conducted using length of first stage of labor as a proxy measure for dose to assess for a dose-response relationship. RESULTS: In this population-based study, antibiotic use during labor and delivery was not associated with an increased risk of ASD in offspring. The unadjusted and adjusted hazard ratios were 1.29 (95% confidence interval, 1.24-1.35) and 0.99 (0.94-1.04), respectively; and 1.07 (0.90-1.27) and 0.88 (0.74-1.05), respectively, in the group B Streptococcus-positive cohort. We observed no substantial difference in the association between antibiotic exposure and ASD depending on length of the first stage of labor. CONCLUSIONS: Our findings suggest that concern for ASD should not factor into the clinical decision on whether to administer antibiotics during labor and delivery. Future research is needed to examine longer durations of prenatal antibiotic exposure.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Prenatal Exposure Delayed Effects , Anti-Bacterial Agents/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/epidemiology , Autistic Disorder/complications , Cohort Studies , Female , Humans , Infant , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/epidemiology , Retrospective Studies , Risk Factors
14.
Immunity ; 55(3): 494-511.e11, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263568

ABSTRACT

Interleukin (IL)-22 is central to immune defense at barrier sites. We examined the contributions of innate lymphoid cell (ILC) and T cell-derived IL-22 during Citrobacter rodentium (C.r) infection using mice that both report Il22 expression and allow lineage-specific deletion. ILC-derived IL-22 activated STAT3 in C.r-colonized surface intestinal epithelial cells (IECs) but only temporally restrained bacterial growth. T cell-derived IL-22 induced a more robust and extensive activation of STAT3 in IECs, including IECs lining colonic crypts, and T cell-specific deficiency of IL-22 led to pathogen invasion of the crypts and increased mortality. This reflected a requirement for T cell-derived IL-22 for the expression of a host-protective transcriptomic program that included AMPs, neutrophil-recruiting chemokines, and mucin-related molecules, and it restricted IFNγ-induced proinflammatory genes. Our findings demonstrate spatiotemporal differences in the production and action of IL-22 by ILCs and T cells during infection and reveal an indispensable role for IL-22-producing T cells in the protection of the intestinal crypts.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Animals , Anti-Bacterial Agents , Immunity, Innate , Interleukins/metabolism , Intestinal Mucosa , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism , Interleukin-22
15.
Fetal Pediatr Pathol ; 41(6): 962-976, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35094661

ABSTRACT

INTRODUCTION: Histologic diagnosis of eosinophilic esophagitis (EoE) involves peak eosinophil counts (PEC) being greater than 15 per high power field. The EoE Histology Scoring System (EoEHSS) was developed to comprehensively evaluate biopsies to better predict symptom and endoscopy response; we aimed to validate the EoEHSS in our provincial registry, where EoEHSS had not been employed. METHODS: We reviewed 186 esophageal biopsies from 16 patients at diagnosis and follow-up. Statistical analyses were conducted to quantify how grade scores correlate with active EoE status and PEC counts, and each feature's ability to predict active disease. RESULTS: Nearly all EoEHSS variables were associated with active EoE and high PEC, with basal zone hyperplasia, eosinophil abscesses, and surface epithelial alteration being most predictive in identifying active EoE. CONCLUSIONS: We validated and demonstrated each EoEHSS variable's strength in tracking traditional PEC counts, resulting in its adoption as a standard reporting element for our research registry.


Subject(s)
Eosinophilic Esophagitis , Child , Humans , British Columbia , Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/pathology , Eosinophils/pathology , Hospitals, Pediatric
16.
Ann Clin Microbiol Antimicrob ; 20(1): 83, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34911528

ABSTRACT

BACKGROUND: Solid transplant patients are susceptible to Pneumocystis jirovecii pneumonia (PJP). While the vast majority of PJP cases occur within the first 6 months after transplantation, very few PJP cases are seen beyond 1 year post-transplantation (late-onset PJP). PJP and coronavirus disease 2019 (COVID-19, caused by infection with SARS-CoV-2) share quite a few common clinical manifestations and imaging findings, making the diagnosis of PJP often underappreciated during the current COVID-19 pandemic. To date, only 1 case of kidney transplantation who developed COVID-19 and late-onset PJP has been reported, but this patient also suffered from many other infections and died from respiratory failure and multiple organ dysfunction syndrome. A successful treatment of kidney patients with COVID-19 and late-onset PJP has not been reported. CASE PRESENTATION: We present a case of a 55-year-old male kidney transplant patient with COVID-19 who also developed late-onset PJP. He received a combined treatment strategy, including specific anti-pneumocystis therapy, symptomatic supportive therapy, adjusted immunosuppressive therapy, and use of antiviral drugs/antibiotics, ending with a favorable outcome. CONCLUSIONS: This case highlights the importance of prompt and differential diagnosis of PJP in kidney transplant patients with SARS-CoV-2 infection. Further studies are required to clarify if kidney transplant patients with COVID-19 could be prone to develop late-onset PJP and how these patients should be treated.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Kidney Transplantation , Pneumonia, Pneumocystis , COVID-19/complications , Humans , Kidney Transplantation/adverse effects , Male , Middle Aged , Pneumonia, Pneumocystis/diagnosis , Pneumonia, Pneumocystis/drug therapy
17.
Cell Host Microbe ; 29(12): 1744-1756.e5, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34678170

ABSTRACT

Interactions between the microbiota and mammalian host are essential for defense against infection, but the microbial-derived cues that mediate this relationship remain unclear. Here, we find that intestinal epithelial cell (IEC)-associated commensal bacteria, segmented filamentous bacteria (SFB), promote early protection against the pathogen Citrobacter rodentium, independent of CD4+ T cells. SFB induced histone modifications in IECs at sites enriched for retinoic acid receptor motifs, suggesting that SFB may enhance defense through retinoic acid (RA). Consistent with this, inhibiting RA signaling suppressed SFB-induced protection. Intestinal RA levels were elevated in SFB mice, despite the inhibition of mammalian RA production, indicating that SFB directly modulate RA. Interestingly, RA was produced by intestinal bacteria, and the loss of bacterial-intrinsic aldehyde dehydrogenase activity decreased the RA levels and increased infection. These data reveal RA as an unexpected microbiota-derived metabolite that primes innate defense and suggests that pre- and probiotic approaches to elevate RA could prevent or combat infections.


Subject(s)
Bacteria/metabolism , Intestinal Diseases/metabolism , Symbiosis , Tretinoin/metabolism , Animals , Bacillus cereus , Bifidobacterium bifidum , CD4-Positive T-Lymphocytes , Citrobacter rodentium , Epithelial Cells , Histone Code , Host Microbial Interactions , Intestinal Diseases/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota , Nitric Oxide , Signal Transduction
18.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34625492

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) control the formation of intestinal lymphoid tissues and play key roles in intestinal defense. They express neuropeptide vasoactive intestinal peptide (VIP) receptor 2 (VPAC2), through which VIP modulates their function, but whether VIP exerts other effects on ILC3 remains unclear. We show that VIP promotes ILC3 recruitment to the intestine through VPAC1 independent of the microbiota or adaptive immunity. VIP is also required for postnatal formation of lymphoid tissues as well as the maintenance of local populations of retinoic acid (RA)-producing dendritic cells, with RA up-regulating gut-homing receptor CCR9 expression by ILC3s. Correspondingly, mice deficient in VIP or VPAC1 suffer a paucity of intestinal ILC3s along with impaired production of the cytokine IL-22, rendering them highly susceptible to the enteric pathogen Citrobacter rodentium This heightened susceptibility to C. rodentium infection was ameliorated by RA supplementation, adoptive transfer of ILC3s, or by recombinant IL-22. Thus, VIP regulates the recruitment of intestinal ILC3s and formation of postnatal intestinal lymphoid tissues, offering protection against enteric pathogens.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Lymphocytes/immunology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , Interleukins/analysis , Lymphoid Tissue/cytology , Lymphoid Tissue/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR/biosynthesis , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Tretinoin/metabolism , Vasoactive Intestinal Peptide/genetics , Interleukin-22
19.
PLoS Pathog ; 17(8): e1009719, 2021 08.
Article in English | MEDLINE | ID: mdl-34352037

ABSTRACT

Reducing food intake is a common host response to infection, yet it remains unclear whether fasting is detrimental or beneficial to an infected host. Despite the gastrointestinal tract being the primary site of nutrient uptake and a common route for infection, studies have yet to examine how fasting alters the host's response to an enteric infection. To test this, mice were fasted before and during oral infection with the invasive bacterium Salmonella enterica serovar Typhimurium. Fasting dramatically interrupted infection and subsequent gastroenteritis by suppressing Salmonella's SPI-1 virulence program, preventing invasion of the gut epithelium. Virulence suppression depended on the gut microbiota, as Salmonella's invasion of the epithelium proceeded in fasting gnotobiotic mice. Despite Salmonella's restored virulence within the intestines of gnotobiotic mice, fasting downregulated pro-inflammatory signaling, greatly reducing intestinal pathology. Our study highlights how food intake controls the complex relationship between host, pathogen and gut microbiota during an enteric infection.


Subject(s)
Bacteria/growth & development , Fasting , Gastroenteritis/prevention & control , Inflammation/prevention & control , Intestines/immunology , NF-kappa B/antagonists & inhibitors , Salmonella Infections, Animal/immunology , Salmonella typhimurium/physiology , Animals , Bacteria/immunology , Bacteria/metabolism , Female , Gastroenteritis/immunology , Gastroenteritis/microbiology , Gastrointestinal Microbiome , Inflammation/immunology , Inflammation/microbiology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/complications , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology
20.
Curr Opin Microbiol ; 63: 76-82, 2021 10.
Article in English | MEDLINE | ID: mdl-34243134

ABSTRACT

Enteric bacterial infections impose a significant and global health burden on society, and their threat is increasing in concert with a rise in antibiotic resistance. There is thus a great need to quickly develop new antimicrobial treatments and interest is growing in targeting pathogen nutrition and metabolism. In this review, we highlight recent research on the metabolism of Citrobacter rodentium, a murine-specific relative of enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC). We focus on the mechanisms by which C. rodentium acquires nutrients as well as the distinct metabolic strategies that C. rodentium employs in varying spatiotemporal niches. We propose that identifying and targeting nutrients found essential for bacterial pathogenesis is an attractive anti-microbial approach in the new post-antibiotic era.


Subject(s)
Enterobacteriaceae Infections , Enterohemorrhagic Escherichia coli , Enteropathogenic Escherichia coli , Animals , Citrobacter , Citrobacter rodentium , Meals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...