Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Adv Ther ; 41(3): 901-914, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38286962

ABSTRACT

Dysbiosis corresponds to the disruption of a formerly stable, functionally complete microbiota. In the gut, this imbalance can lead to adverse health outcomes in both the short and long terms, with a potential increase in the lifetime risks of various noncommunicable diseases and disorders such as atopy (like asthma), inflammatory bowel disease, neurological disorders, and even behavioural and psychological disorders. Although antibiotics are highly effective in reducing morbidity and mortality in infectious diseases, antibiotic-associated diarrhoea is a common, non-negligible clinical sign of gut dysbiosis (and the only visible one). Re-establishment of a normal (functional) gut microbiota is promoted by completion of the clinically indicated course of antibiotics, the removal of any other perturbing external factors, the passage of time (i.e. recovery through the microbiota's natural resilience), appropriate nutritional support, and-in selected cases-the addition of probiotics. Systematic reviews and meta-analyses of clinical trials have confirmed the strain-specific efficacy of some probiotics (notably the yeast Saccharomyces boulardii CNCM I-745 and the bacterium Lactobacillus rhamnosus GG) in the treatment and/or prevention of antibiotic-associated diarrhoea in children and in adults. Unusually for a probiotic, S. boulardii is a eukaryote and is not therefore directly affected by antibiotics-making it suitable for administration in cases of antibiotic-associated diarrhoea. A robust body of evidence from clinical trials and meta-analyses shows that the timely administration of an adequately dosed probiotic (upon initiation of antibiotic treatment or within 48 h) can help to prevent or resolve the consequences of antibiotic-associated dysbiosis (such as diarrhoea) and promote the resilience of the gut microbiota and a return to the pre-antibiotic state. A focus on the prescription of evidence-based, adequately dosed probiotics should help to limit unjustified and potentially ineffective self-medication.


Subject(s)
Lacticaseibacillus rhamnosus , Probiotics , Saccharomyces boulardii , Adult , Child , Humans , Anti-Bacterial Agents/adverse effects , Diarrhea/chemically induced , Diarrhea/prevention & control , Dysbiosis/chemically induced , Dysbiosis/therapy , Probiotics/therapeutic use , Saccharomyces cerevisiae , Meta-Analysis as Topic , Systematic Reviews as Topic
2.
Clin Gastroenterol Hepatol ; 22(1): 22-33.e6, 2024 01.
Article in English | MEDLINE | ID: mdl-37716619

ABSTRACT

BACKGROUND & AIMS: Withdrawal of immunomodulators (IMMs) or tumor necrosis factor (TNF) antagonists in patients with inflammatory bowel diseases (IBDs) in remission on combination therapy is attractive. We evaluated the efficacy and safety of (1) IMM, or (2) TNF antagonist withdrawal in patients with IBD in sustained remission on combination therapy. METHODS: Through a systematic review till March 31, 2023, we identified randomized controlled trials (RCTs) that compared the efficacy and safety of IMM or TNF antagonist withdrawal vs continued combination therapy, in patients with IBD in sustained corticosteroid-free clinical remission for >6 months on combination therapy. Primary outcome was risk of relapse and serious adverse events at 12 months. We conducted meta-analysis to calculate relative risk (RR) and 95% confidence interval (CI) and used Grading of Recommendations Assessment, Development and Evaluation (GRADE) to appraise certainty of evidence. RESULTS: We identified 8 RCTs with 733 patients (77% with Crohn's disease, 91% on infliximab-based combination therapy). On meta-analysis of 5 RCTs, there was no difference in the risk of relapse between patients with IMM withdrawal (continued TNF antagonist monotherapy) vs continued combination therapy (16.8% vs 14.9%; RR, 1.15; 95% CI, 0.75-1.76) without heterogeneity (low certainty of evidence). TNF antagonist withdrawal (continued IMM monotherapy) was associated with 2.4-times higher risk of relapse compared with continuing combination therapy (31.5% vs 11.2%; RR, 2.35; 95% CI, 1.38-4.01), with minimal heterogeneity (low certainty of evidence). There was no difference in the risk of serious adverse events with IMM or TNF antagonist withdrawal vs continued combination therapy. CONCLUSIONS: In patients with IBD in sustained corticosteroid-free clinical remission for >6 months on combination therapy, de-escalation with TNF antagonist withdrawal, but not IMM withdrawal, was associated with an increased risk of relapse.


Subject(s)
Crohn Disease , Inflammatory Bowel Diseases , Humans , Immunosuppressive Agents/therapeutic use , Tumor Necrosis Factor Inhibitors/adverse effects , Immunologic Factors/adverse effects , Crohn Disease/drug therapy , Recurrence , Remission Induction , Inflammatory Bowel Diseases/drug therapy
3.
Inflamm Bowel Dis ; 30(3): 402-409, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37229656

ABSTRACT

BACKGROUND: Intestinal ultrasound (IUS) is a noninvasive tool to assess bowel inflammation. There is a paucity of data on its accuracy in pediatric patients. AIM: The aim of this study is to evaluate the diagnostic performance of bowel wall thickness (BWT) measured using IUS compared with endoscopic disease activity in children suspected of having inflammatory bowel disease (IBD). METHODS: We conducted a single-center cross-sectional pilot study of pediatric patients suspected to have previously undiagnosed IBD. Endoscopic inflammation was graded using segmental scores of the Simple Endoscopic Score for Crohn's Disease (SES-CD) and the Ulcerative Colitis Endoscopic Index of Severity (UCEIS) and classified as having healthy, mild, or moderate/severe disease activity. Association between BWT and endoscopic severity was assessed using the Kruskal-Wallis test. The diagnostic performance of BWT to detect active disease at endoscopy was evaluated using the area under the receiver operating characteristic curve; sensitivity and specificity were calculated. RESULTS: In all, 174 bowel segments in 33 children were assessed by IUS and ileocolonoscopy. An elevated median BWT was associated with increased bowel segment disease severity, classified by the SES-CD (P < .001) and the UCEIS (P < .01). Using a cutoff value of 1.9 mm, we found that the BWT had an area under the receiver operating characteristic curve of 0.743 (95% CI, 0.67-0.82), a sensitivity of 64% (95% CI, 53%-73%), and a specificity of 76% (95% CI, 65%-85%) to detect inflamed bowel. CONCLUSION: Increasing BWT is associated with increasing endoscopic activity in pediatric IBD. Our study suggests that the optimal BWT cutoff value for detecting active disease may be less than that seen in adults. Additional pediatric studies are needed.


Increasing bowel wall thickness (BWT) is associated with increasing IBD endoscopic scores on colonoscopy. There is moderate to fair agreement between the prediction of IBD diagnosis and Paris classification using intestinal ultrasound (IUS). Bowel wall thickness cutoff values to detect inflamed bowel segments are likely lower for children with IBD than for adults, although further studies with wider age ranges are needed to confirm this finding.


Subject(s)
Colitis, Ulcerative , Inflammatory Bowel Diseases , Adult , Humans , Child , Cross-Sectional Studies , Pilot Projects , Colitis, Ulcerative/diagnostic imaging , Inflammation , Patient Acuity
4.
iScience ; 26(10): 107829, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37736049

ABSTRACT

Colonic epithelial repair is a key determinant of health. Repair involves changes in epithelial differentiation, an extensive proliferative response, and upregulation of regeneration-associated "fetal-like" transcripts, including Ly6a (Sca-1), that represent Yap1 and interferon targets. However, little is known about how this regenerative program terminates and how homeostasis is restored during injury and inflammation. Here we show that, after the initial entry into the regenerative state, the subsequent upregulation of tumor necrosis factor (TNF) receptor 2 (R2, TNFR2, Tnfrsf1b) clears the regenerative signaling and restores homeostatic patterns of epithelial differentiation. Targeted deletion of epithelial TNFR2 in vivo and in colonoid cultures revealed persistent expression of Ly6a, hyperproliferation, and reduced secretory differentiation. Moreover, mice lacking epithelial TNFR2 also failed to complete colon ulcer healing, suggesting that partial resolution of regenerative signaling is essential for the completion of the repair process. These results demonstrate how epithelial cells dynamically leverage a colitis-associated cytokine to choreograph repair.

5.
Dev Cell ; 58(21): 2309-2325.e7, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37652012

ABSTRACT

Chronic colonic injury and inflammation pose high risks for field cancerization, wherein injury-associated mutations promote stem cell fitness and gradual clonal expansion. However, the long-term stability of some colitis-associated mutational fields could suggest alternate origins. Here, studies of acute murine colitis reveal a punctuated mechanism of massive, neutral clonal expansion during normal wound healing. Through three-dimensional (3D) imaging, quantitative fate mapping, and single-cell transcriptomics, we show that epithelial wound repair begins with the loss of structural constraints on regeneration, forming fused labyrinthine channels containing epithelial cells reprogrammed to a non-proliferative plastic state. A small but highly proliferative set of epithelial founder progenitor cells (FPCs) subsequently emerges and undergoes extensive cell division, enabling fluid-like lineage mixing and spreading across the colonic surface. Crypt budding restores the glandular organization, imprinting the pattern of clonal expansion. The emergence and functions of FPCs within a critical window of plasticity represent regenerative targets with implications for preneoplasia.


Subject(s)
Colitis , Mice , Animals , Colitis/genetics , Epithelial Cells , Stem Cells , Wound Healing
6.
Gastroenterology ; 162(7): 1975-1989, 2022 06.
Article in English | MEDLINE | ID: mdl-35227778

ABSTRACT

BACKGROUND & AIMS: Epithelial wound healing is compromised and represents an unleveraged therapeutic target in inflammatory bowel disease (IBD). Intestinal epithelial cells exhibit plasticity that facilitates dedifferentiation and repair during the response to injury. However, it is not known whether epithelial cells of a neighboring organ can be activated to mediate re-epithelialization in acute colitis. Histological findings of a permanent squamous tissue structure in the distal colon in human IBD could suggest diverse cellular origins of repair-associated epithelium. Here, we tested whether skin-like cells from the anus mediate colonic re-epithelialization in murine colitis. METHODS: We studied dextran sulfate sodium-induced colitis and interleukin 10-deficient colitis in transgenic mice. We performed lineage tracing, 3-dimensional (3D) imaging, single-cell transcriptomics, and biophysical modeling to map squamous cell fates and to identify squamous cell types involved in colonic repair. RESULTS: In acute and chronic colitis, we found a large squamous epithelium, called squamous neo-epithelium of the colon (SNEC), near the anorectal junction. Neighboring squamous cells of the anus rapidly migrate into the ulcerated colon and establish this permanent epithelium of crypt-like morphology. These squamous cells derive from a small unique transition zone, distal to the border of colonic and anal epithelium, that resists colitic injury. The cells of this zone have a pre-loaded program of colonic differentiation and further upregulate key aspects of colonic epithelium during repair. CONCLUSION: Transitional anal cells represent unique reserve cells capable of rebuilding epithelial structures in the colon after colitis. Further study of these cells could reveal novel approaches to direct mucosal healing in inflammation and disease.


Subject(s)
Carcinoma, Squamous Cell , Colitis , Inflammatory Bowel Diseases , Anal Canal/pathology , Animals , Carcinoma, Squamous Cell/pathology , Colitis/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Re-Epithelialization
7.
Am J Physiol Gastrointest Liver Physiol ; 321(3): G308-G324, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34260310

ABSTRACT

Intestinal mucosal healing is the primary therapeutic goal of medical treatments for inflammatory bowel disease (IBD). Epithelial stem cells are key players in the healing process. Lgr5+ stem cells maintain cellular turnover during homeostasis in the colonic crypt. However, they are lost and dispensable for repair in a wide variety of injury models, including dextran sulfate sodium (DSS) colitis, radiation, helminth infection, and T-cell activation. The direct loss of Lgr5+ cells activates a plasticity response in the epithelium in which other cell types can serve as stem cells. Whether this paradigm applies to mouse models of IBD remains unknown. In contrast to previously tested models, IBD models involve an inflammatory response rooted in the loss of immunologic tolerance to intestinal luminal contents including the microbiome. Here, we show the persistence of Lgr5+ cells in oxazolone, 2,4,6-trinitrobenzene sulfonic acid (TNBS), and Il10-/-, and Il10-/- Tnfr1-/- IBD models. This contrasts with results obtained from DSS-induced injury. Through high-throughput expression profiling, we find that these colitis models were associated with distinct patterns of cytokine expression. Direct exposure of colonic epithelial organoids to DSS, oxazolone, or TNBS resulted in increased apoptosis and loss of Lgr5+ cells. Targeted ablation of Lgr5+ cells resulted in severe exacerbation of chronic, antibody-induced IL-10-deficient colitis, but had only modest effects in TNBS-induced colitis. These results show that distinct mouse models of IBD-like colitis induce different patterns of Lgr5+ stem cell retention and function.NEW & NOTEWORTHY Acute intestinal injury and epithelial repair are associated with the loss of fast-cycling Lgr5+ stem cells and plasticity in the activation of formerly quiescent cell populations. In contrast, here we show in murine inflammatory bowel disease the persistence of the Lgr5+ stem cell population and its essential role in restricting the severity of chronic colitis. This demonstrates a diversity of stem cell responses to colitis.


Subject(s)
Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Animals , Cell Proliferation/physiology , Disease Models, Animal , Epithelium/metabolism , Homeostasis/physiology , Intestinal Mucosa/metabolism , Mice , Regeneration/physiology , Stem Cells/metabolism
8.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G990-G1001, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33826403

ABSTRACT

Proinflammatory macrophages are essential drivers of colitis and express the growth factor receptor ErbB4. This study tested the role of ErbB4 and its specific ligand, NRG4, in regulating macrophage function. We show that endogenous NRG4-ErbB4 signaling limits macrophage production of proinflammatory cytokines in vitro and limits colitis severity in vivo and thus is a potential target for therapeutic intervention.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Neuregulins/metabolism , Receptor, ErbB-4/metabolism , Signal Transduction/physiology , Animals , Colitis/metabolism , Colon/metabolism , Cytokines/metabolism , Inflammation/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophage Activation/physiology , Mice , Mice, Knockout
9.
Cell Mol Gastroenterol Hepatol ; 11(5): 1327-1345, 2021.
Article in English | MEDLINE | ID: mdl-33482393

ABSTRACT

BACKGROUND & AIMS: Colonization by gut microbiota in early life confers beneficial effects on immunity throughout the host's lifespan. We sought to elucidate the mechanisms whereby neonatal supplementation with p40, a probiotic functional factor, reprograms intestinal epithelial cells for protection against adult-onset intestinal inflammation. METHODS: p40 was used to treat young adult mouse colonic (YAMC) epithelial cells with and without deletion of a methyltransferase, su(var)3-9, enhancer-of-zeste and trithorax domain-containing 1ß (Setd1ß), and mice in early life or in adulthood. Anti-transforming growth factor ß (TGFß)-neutralizing antibodies were administered to adult mice with and without colitis induced by 2,4,6-trinitrobenzenesulfonic acid or dextran sulfate sodium. We examined Setd1b and Tgfb gene expression, TGFß production, monomethylation and trimethylation of histone H3 on the lysine 4 residue (H3K4me1/3), H3K4me3 enrichment in Tgfb promoter, differentiation of regulatory T cells (Tregs), and the inflammatory status. RESULTS: p40 up-regulated expression of Setd1b in YAMC cells. Accordingly, p40 enhanced H3K4me1/3 in YAMC cells in a Setd1ß-dependent manner. p40-regulated Setd1ß mediated programming the TGFß locus into a transcriptionally permissive chromatin state and promoting TGFß production in YAMC. Furthermore, transient exposure to p40 during the neonatal period and in adulthood resulted in the immediate increase in Tgfb gene expression. However, only neonatal p40 supplementation induced the sustained H3K4me1/3 and Tgfb gene expression that persisted into adulthood. Interfering with TGFß function by neutralizing antibodies diminished the long-lasting effects of neonatal p40 supplementation on differentiation of Tregs and protection against colitis in adult mice. CONCLUSIONS: Exposure to p40 in early life enables an epigenetic imprint on TGFß, leading to long-lasting production of TGFß by intestinal epithelial cells to expand Tregs and protect the gut against inflammation.


Subject(s)
Colitis/prevention & control , Epigenesis, Genetic , Inflammation/prevention & control , Intestinal Mucosa/drug effects , Prenatal Exposure Delayed Effects/drug therapy , Probiotics/pharmacology , Transforming Growth Factor beta/genetics , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Transforming Growth Factor beta/metabolism
10.
Cell Rep ; 33(3): 108275, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086075

ABSTRACT

Neutralization of tumor necrosis factor (TNF) represents a widely used therapeutic strategy for autoimmune diseases including inflammatory bowel disease (IBD). However, the fact that many patients with IBD are non-responsive to anti-TNF therapies suggests the need for a better understanding of TNF signaling in IBD. Here, we show that co-deletion of TNF receptor 1 (TNFR1, Tnfrsf1a) in the Il10-/- spontaneous colitis model exacerbates disease, resulting in very-early-onset inflammation after weaning. The disease can be interrupted by treatment with antibiotics. The single deletion of TNFR1 induces subclinical colonic epithelial dysfunction and mucosal immune abnormalities, including accumulation of neutrophils and depletion of B cells. During the pre-disease period (before weaning), both Tnfr1-/- and Il10-/-Tnfr1-/- animals exhibit impaired expression of pro-inflammatory cytokines compared with wild-type and Il10-/- controls, respectively. Collectively, these results demonstrate the net anti-inflammatory functions of TNF/TNFR1 signaling through the regulation of colonic immune homeostasis in early life.


Subject(s)
Colitis/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Colitis/immunology , Colitis/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate/pharmacology , Epithelial Cells/metabolism , Female , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism
11.
Front Immunol ; 11: 1428, 2020.
Article in English | MEDLINE | ID: mdl-32719681

ABSTRACT

Advances in our understanding of the contribution of the gut microbiota to human health and the correlation of dysbiosis with diseases, including chronic intestinal conditions such as inflammatory bowel disease (IBD), have driven mechanistic investigations of probiotics in intestinal homeostasis and potential clinical applications. Probiotics have been shown to promote intestinal health by maintaining and restoring epithelial function, ensuring mucosal immune homeostasis, and inhibiting pathogenic bacteria. Recent findings reveal an approach for defining previously unrecognized probiotic-derived soluble factors as potential mechanisms of probiotic action. This review focuses on the impact of probiotics and probiotic-derived functional factors, including probiotic products and metabolites by probiotics, on the cellular responses and signaling pathways involved in maintaining intestinal homeostasis. Although there is limited information regarding the translation of probiotic treatment outcomes from in vitro and animal studies to clinical applications, potential approaches for increasing the clinical efficacy of probiotics for IBD, such as those based on probiotic-derived factors, are highlighted in this review. In this era of precision medicine and targeted therapies, more basic, preclinical, and clinical evidence is needed to clarify the efficacy of probiotics in maintaining intestinal health and preventing and treating disease.


Subject(s)
Gastrointestinal Microbiome , Homeostasis , Intestines , Probiotics/pharmacology , Animals , Dysbiosis , Humans
12.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G1-G10, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32421359

ABSTRACT

The development of modern methods to induce optical transparency ("clearing") in biological tissues has enabled the three-dimensional (3D) reconstruction of intact organs at cellular resolution. New capabilities in visualization of rare cellular events, long-range interactions, and irregular structures will facilitate novel studies in the alimentary tract and gastrointestinal systems. The tubular geometry of the alimentary tract facilitates large-scale cellular reconstruction of cleared tissue without specialized microscopy setups. However, with the rapid pace of development of clearing agents and current relative paucity of research groups in the gastrointestinal field using these techniques, it can be daunting to incorporate tissue clearing into experimental workflows. Here, we give some advice and describe our own experience bringing tissue clearing and whole mount reconstruction into our laboratory's investigations. We present a brief overview of the chemical concepts that underpin tissue clearing, what sorts of questions whole mount imaging can answer, how to choose a clearing agent, an example of how to clear and image alimentary tissue, and what to do after obtaining the image. This short review will encourage other gastrointestinal researchers to consider how utilizing tissue clearing and creating 3D "maps" of tissue might deepen the impact of their studies.


Subject(s)
Gastrointestinal Tract/pathology , Tissue Culture Techniques , Animals , Cellular Microenvironment/physiology , Humans , Imaging, Three-Dimensional/methods , Research
13.
Infect Immun ; 87(7)2019 07.
Article in English | MEDLINE | ID: mdl-31010817

ABSTRACT

The symbiotic relationship between the gut microbiome and the host provides a nutrient-rich environment for gut microbes and has beneficial effects on host health. Although the composition of the gut microbiome is known to be influenced by both host genetics and environmental factors, host effects on the activities and functions of the gut microbial communities remain poorly understood. Intestinal epithelial cells exert front-line responses to gut microbes and contribute to maintaining a healthy intestinal homeostasis. Here, seeking to elucidate whether intestinal epithelial cells modulate Lactobacillus rhamnosus GG (LGG) functions, we examined the production of p40, an LGG-derived secretory protein that protects intestinal epithelial cells against inflammation. We found that growth medium conditioned with colonic epithelial cell-derived components promotes p40 protein synthesis and secretion by LGG and enhances LGG-stimulated protective responses in intestinal epithelial cells. Furthermore, when LGG was cultured with the colonic luminal contents from healthy mice, p40 production was upregulated but was attenuated with luminal contents from mice with intestinal inflammation. Importantly, the colonic epithelial cell-derived components potentiated LGG-produced p40 levels in a mouse model of colitis and enhanced LGG-mediated amelioration of intestinal inflammation in this model. Notably, we found that colonic epithelial cell-secreted extracellular vesicles participate in communicating with LGG and that heat shock protein 90 (HSP90) in these vesicles might mediate the promotion of p40 production. These results reveal a previously unrecognized mechanism by which the anti-inflammatory effect of LGG is reinforced by intestinal epithelial cells and thereby maintains intestinal health.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/microbiology , Lacticaseibacillus rhamnosus/metabolism , Secretory Vesicles/microbiology , Animals , Bacterial Proteins/genetics , Epithelial Cells/metabolism , HSP90 Heat-Shock Proteins/metabolism , Intestinal Mucosa/metabolism , Lacticaseibacillus rhamnosus/genetics , Mice , Mice, Inbred C57BL , Secretory Vesicles/metabolism
14.
Cell Host Microbe ; 24(4): 472-474, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30308153

ABSTRACT

Pediatric ulcerative colitis incidence is rapidly rising, yet improved prognostic and therapeutic strategies are needed. In this issue of Cell Host & Microbe, Schirmer et al. (2018) reveal the dynamism of pediatric patient microbiomes through initial diagnosis and treatments, providing insights into microbial targets that predict therapeutic response and disease outcomes.


Subject(s)
Colitis, Ulcerative , Microbiota , Child , Humans , Prognosis
15.
Mucosal Immunol ; 11(5): 1316-1328, 2018 09.
Article in English | MEDLINE | ID: mdl-29875401

ABSTRACT

The beneficial effects of the gut microbiota on growth in early life are well known. However, knowledge about the mechanisms underlying regulating intestinal development by the microbiota is limited. p40, a Lactobacillus rhamnosus GG-derived protein, transactivates epidermal growth factor receptor (EGFR) in intestinal epithelial cells for protecting the intestinal epithelium against injury and inflammation. Here, we developed p40-containing pectin/zein hydrogels for targeted delivery of p40 to the small intestine and the colon. Treatment with p40-containing hydrogels from postnatal day 2 to 21 significantly enhanced bodyweight gain prior to weaning and functional maturation of the intestine, including intestinal epithelial cell proliferation, differentiation, and tight junction formation, and IgA production in early life in wild-type mice. These p40-induced effects were abolished in mice with specific deletion of EGFR in intestinal epithelial cells, suggesting that transactivation of EGFR in intestinal epithelial cells may mediate p40-regulated intestinal development. Furthermore, neonatal p40 treatment reduced the susceptibility to intestinal injury and colitis and promoted protective immune responses, including IgA production and differentiation of regulatory T cells, in adult mice. These findings reveal novel roles of neonatal supplementation of probiotic-derived factors in promoting EGFR-mediated maturation of intestinal functions and innate immunity, which likely promote long-term beneficial outcomes.


Subject(s)
Bacterial Proteins/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Intestinal Mucosa/drug effects , Lacticaseibacillus rhamnosus/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Epithelial Cells/immunology , Female , Hydrogels/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoglobulin A/immunology , Mice , Mice, Inbred C57BL , Probiotics/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tight Junctions/drug effects , Tight Junctions/immunology , Time , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
16.
Sci Rep ; 8(1): 9119, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29904166

ABSTRACT

Current treatments for inflammatory bowel disease (IBD) target the overactive immune response of the intestinal mucosa. However, epidermal growth factor (EGF), an activating ligand of the EGF receptor (EGFR), has been shown to induce disease remission through direct targeting of intestinal mucosal healing. Despite promising preclinical and clinical results, this EGFR-activating therapy has not progressed, in part due to the potential for carcinogenesis associated with long-term use and the increased risk of colitis-associated cancer (CAC) in IBD. Here we tested whether pharmacological modulation of EGFR altered outcomes of CAC in the murine azoxymethane/dextran sulfate sodium model. We found that administering EGF during the period of maximum colitis severity ("early"), coincident with the initiation and early promotion of tumors, improved outcomes of colitis and reduced tumor size. In contrast, daily EGF administration beginning ~2 months after tumor initiation ("late") increased tumor size. Administration of the EGFR kinase inhibitor gefitinib increased the tumor size when the drug was given early and decreased the tumor size when the drug was administered late. EGF administration not only reduced colonic cytokine and chemokine expression during injury, but also baseline chemokine expression in homeostasis. These results suggest that EGFR activation during acute bouts of colitis may reduce the long-term burden of CAC.


Subject(s)
Colitis/metabolism , Colonic Neoplasms/metabolism , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Signal Transduction/drug effects , Animals , Azoxymethane/toxicity , Colitis/chemically induced , Colitis/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , Mice , Neoplasm Proteins/agonists , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
17.
J Cell Sci ; 130(1): 90-96, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27026527

ABSTRACT

Cell shedding from the intestinal villus is a key element of tissue turnover that is essential to maintain health and homeostasis. However, the signals regulating this process are not well understood. We asked whether shedding is controlled by epidermal growth factor receptor (EGFR), an important driver of intestinal growth and differentiation. In 3D ileal enteroid culture and cell culture models (MDCK, IEC-6 and IPEC-J2 cells), extrusion events were suppressed by EGF, as determined by direct counting of released cells or rhodamine-phalloidin labeling of condensed actin rings. Blockade of the MEK-ERK pathway, but not other downstream pathways such as phosphoinositide 3-kinase (PI3K) or protein kinase C (PKC), reversed EGF inhibition of shedding. These effects were not due to a change in cell viability. Furthermore, EGF-driven MAPK signaling inhibited both caspase-independent and -dependent shedding pathways. Similar results were found in vivo, in a novel zebrafish model for intestinal epithelial shedding. Taken together, the data show that EGF suppresses cell shedding in the intestinal epithelium through a selective MAPK-dependent pathway affecting multiple extrusion mechanisms. EGFR signaling might be a therapeutic target for disorders featuring excessive cell turnover, such as inflammatory bowel diseases.


Subject(s)
Epidermal Growth Factor/pharmacology , Epithelial Cells/metabolism , Intestines/cytology , MAP Kinase Signaling System/drug effects , Animals , Caspase Inhibitors/pharmacology , Caspases/metabolism , Dogs , Epithelial Cells/drug effects , Madin Darby Canine Kidney Cells , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Zebrafish , rho GTP-Binding Proteins/metabolism
18.
J Biol Chem ; 291(39): 20462-72, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27507810

ABSTRACT

EGF receptor (EGFR) in tumor cells serves as a tumor promoter. However, information about EGFR activation in macrophages in regulating M2 polarization and tumor development is limited. This study aimed to investigate the effects of EGFR activation in macrophages on M2 polarization and development of gastrointestinal tumors. IL-4, a cytokine to elicit M2 polarization, stimulated release of an EGFR ligand, HB-EGF, and transactivation and down-regulation of EGFR in Raw 264.7 cells and peritoneal macrophages from WT mice. Knockdown of HB-EGF in macrophages inhibited EGFR transactivation by IL-4. IL-4-stimulated STAT6 activation, Arg1 and YM1 gene expression, and HB-EGF production were further enhanced by inhibition of EGFR activity in Raw 264.7 cells using an EGFR kinase inhibitor and in peritoneal macrophages from Egfr(wa5) mice with kinase inactive EGFR and by knockdown of EGFR in peritoneal macrophages from Egfr(fl/fl) LysM-Cre mice with myeloid cell-specific EGFR deletion. Chitin induced a higher level of M2 polarization in peritoneal macrophages in Egfr(fl/fl) LysM-Cre mice than that in Egfr(fl/fl) mice. Accordingly, IL-4-conditioned medium stimulated growth and epithelial-to-mesenchymal transition in gastric epithelial and colonic tumor cells, which were suppressed by that from Raw 264.7 cells with HB-EGF knockdown but promoted by that from Egfr(wa5) and Egfr(fl/fl) LysM-Cre peritoneal macrophages. Clinical assessment revealed that the number of macrophages with EGFR expression became less, indicating decreased inhibitory effects on M2 polarization, in late stage of human gastric cancers. Thus, IL-4-stimulated HB-EGF-dependent transactivation of EGFR in macrophages may mediate inhibitory feedback for M2 polarization and HB-EGF production, thereby inhibiting gastrointestinal tumor growth.


Subject(s)
ErbB Receptors/biosynthesis , Gastrointestinal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Macrophages, Peritoneal/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , ErbB Receptors/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout , RAW 264.7 Cells
19.
J Control Release ; 230: 79-87, 2016 05 28.
Article in English | MEDLINE | ID: mdl-27063422

ABSTRACT

Probiotics have shown beneficial effects on health and prevention of diseases in humans. However, a concern for application of probiotics is the loss of viability during storage and gastrointestinal transit. The aim of this study was to develop an encapsulation system to preserve viability of probiotics when they are administrated orally and apply Lactobacillus rhamnosus GG (LGG) as a probiotic model to evaluate the effectiveness of this approach using in vitro and in vivo experiments. LGG was encapsulated in hydrogel beads prepared using pectin, a food grade polysaccharide, glucose, and calcium chloride, and lyophilized by freeze-drying. Encapsulated LGG was cultured in vitro under the condition that mimicked the physiological environment of the human gastrointestinal tract. Compared to non-encapsulated LGG, encapsulation increased tolerance of LGG in the acid condition, protected LGG from protease digestion, and improved shelf time when stored at the ambient condition, in regard of survivability and production of p40, a known LGG-derived protein involved in LGG's beneficial effects on intestinal homeostasis. To evaluate the effects of encapsulation on p40 production in vivo and prevention of intestinal inflammation by LGG, mice were gavaged with LGG containing beads and treated with dextran sulphate sodium (DSS) to induce intestinal injury and colitis. Compared to non-encapsulated LGG, encapsulated LGG enhanced more p40 production in mice, and exerted higher levels of effects on prevention of DSS-induced colonic injury and colitis and suppression of pro-inflammatory cytokine production. These data indicated that the encapsulation system developed in this study preserves viability of LGG in vitro and in vivo, leading to longer shelf time and enhancing the functions of LGG in the gastrointestinal tract. Thus, this encapsulation approach may have the potential application for improving efficacy of probiotics.


Subject(s)
Glucose/chemistry , Hydrogels/administration & dosage , Lacticaseibacillus rhamnosus , Pectins/chemistry , Probiotics/administration & dosage , Administration, Oral , Animals , Bacterial Proteins/analysis , Calcium Chloride/chemistry , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colitis/prevention & control , Colon/metabolism , Colon/pathology , Colony Count, Microbial , Dextran Sulfate , Feces/chemistry , Hydrogels/chemistry , Hydrogen-Ion Concentration , Lacticaseibacillus rhamnosus/chemistry , Mice, Inbred C57BL , Microspheres , Peroxidase/metabolism , Probiotics/chemistry
20.
Gastroenterology ; 149(4): 993-1005.e2, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26072395

ABSTRACT

BACKGROUND & AIMS: Tumor necrosis factor receptor 2 (TNFR2, Tnfrsf1b) regulates multiple aspects of immune function, but little is known about its role in the immunopathogenesis of inflammatory bowel disease (IBD). We investigated whether TNFR2 restricts the activity of specific immune cell subtypes to protect against the development of colitis in mice. METHODS: Tnfr2(-/-) mice were crossed with interleukin (Il) 10(-/-) mice, which spontaneously develop colitis, to generate Il10(-/-)Tnfr2(-/-) mice. Colonic tissues were collected from Il10(-/-)Tnfr2(-/-) mice along with Il10(-/-) mice (controls) and analyzed by flow cytometry and histology. Bone marrow was transplanted into Il10(-/-) and Il10(-/-)Tnfr2(-/-) mice from Il10(-/-) or Il10(-/-)Tnfr2(-/-) donors by intravenous injection. CD8(+) T cells were neutralized in Il10(-/-)Tnfr2(-/-) mice by intraperitoneal injection of anti-CD8 or isotype control antibodies. Colitis was induced in Rag2(-/-) mice by intravenous injections of naïve CD8(+) T cells isolated from C57BL/6 or Tnfr2(-/-) mice. RESULTS: Il10(-/-)Tnfr2(-/-) mice spontaneously developed more severe colitis compared with Il10(-/-) controls, characterized by selective expansion of colonic CD8(+) T cells. Transplantation of TNFR2-deficient bone marrow resulted in significantly increased incidence and severity of colitis. Transcriptome analyses showed that the expression of genes regulated by TNFR2 were specific to CD8(+) T cells and included genes associated with risk for IBD. Depletion of CD8(+) T cells from Il10(-/-)Tnfr2(-/-) mice prevented colonic inflammation. Adoptive transfer of TNFR2-null naïve CD8(+) T cells compared with CD8(+) T cells from control mice increased the severity of colitis that developed in Rag2(-/-) mice. CONCLUSIONS: TNFR2 protects mice from colitis by inhibiting the expansion of colonic CD8(+) T cells. TNFR2 regulates expression of genes that regulate CD8(+) T cells and have been associated with susceptibility to IBD. Disruption in TNFR2 signaling might therefore be associated with pathogenesis. Strategies to increase levels or activity of TNFR2 and thereby reduce the activity of CD8(+) T cells might be developed to treat IBD patients with CD8(+) T cell dysfunction.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Colitis/prevention & control , Colon/metabolism , Immunity, Cellular , Receptors, Tumor Necrosis Factor, Type II/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/immunology , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...