Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nat Commun ; 15(1): 1854, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38424106

ABSTRACT

The XBB.1.5 variant of SARS-CoV-2 has rapidly achieved global dominance and exhibits a high growth advantage over previous variants. Preliminary reports suggest that the success of XBB.1.5 stems from mutations within its spike glycoprotein, causing immune evasion and enhanced receptor binding. We present receptor binding studies that demonstrate retention of binding contacts with the human ACE2 receptor and a striking decrease in binding to mouse ACE2 due to the revertant R493Q mutation. Despite extensive evasion of antibody binding, we highlight a region on the XBB.1.5 spike protein receptor binding domain (RBD) that is recognized by serum antibodies from a donor with hybrid immunity, collected prior to the emergence of the XBB.1.5 variant. T cell assays reveal high frequencies of XBB.1.5 spike-specific CD4+ and CD8+ T cells amongst donors with hybrid immunity, with the CD4+ T cells skewed towards a Th1 cell phenotype and having attenuated effector cytokine secretion as compared to ancestral spike protein-specific cells. Thus, while the XBB.1.5 variant has retained efficient human receptor binding and gained antigenic alterations, it remains susceptible to recognition by T cells induced via vaccination and previous infection.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Animals , Mice , SARS-CoV-2/genetics , CD8-Positive T-Lymphocytes , Angiotensin-Converting Enzyme 2 , Spike Glycoprotein, Coronavirus/genetics , Antibodies
2.
Inflamm Bowel Dis ; 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37874904

ABSTRACT

BACKGROUND & AIMS: Clostridioides difficile is a toxin-secreting bacteria that is an urgent antimicrobial resistance threat, with approximately 25% of patients developing recurrent infections. Inflammatory bowel disease (IBD) patients are at increased risk of severe, recurrent C. difficile infection. METHODS: To investigate a role for C. difficile infection in IBD pathogenesis, we collected peripheral blood and stool from 20 each of ulcerative colitis patients, Crohn's disease patients, and healthy control subjects. We used a flow cytometric activation induced marker assay to quantify C. difficile toxin-specific CD4+ T cells and 16S ribosomal RNA sequencing to study microbiome diversity. RESULTS: We found IBD patients had significantly increased levels of C. difficile toxin B-specific CD4+ T cells, but not immunoglobulin G or immunoglobulin A, compared with healthy control subjects. Within antigen-specific CD4+ T cells, T helper type 17 cells and cells expressing the gut homing receptor integrin ß7 were reduced compared with healthy control subjects, similar to our previous study of non-IBD patients with recurrent C. difficile infection. Stool microbiome analysis revealed that gut homing, toxin-specific CD4+ T cells negatively associated with microbial diversity and, along with T helper type 17 cells, positively associated with bacteria enriched in healthy control subjects. CONCLUSIONS: These data suggest that IBD patients, potentially due to underlying intestinal dysbiosis, experience undiagnosed C. difficile infections that result in impaired toxin-specific immunity. This may contribute to the development of inflammatory T cell responses toward commensal bacteria and provide a rationale for C. difficile testing in IBD patients.


Crohn's disease and ulcerative colitis patients with no history of Clostridioides difficile infection had dysregulated T cell immunity to C. difficile toxin B. This was significantly different from healthy control subjects but similar to non­inflammatory bowel disease patients with recurrent C. difficile infection.

3.
Anaerobe ; 82: 102755, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37406762

ABSTRACT

BACKGROUND: Diagnosis of Clostridioides difficile Infection (CDI) entails compatible clinical presentation and laboratory findings. We evaluated real-time polymerase chain reaction (qPCR) cycle threshold (CT) as a predictor for disease severity and TcdB enzyme immunoassay (EIA) results. METHODS: Inpatients or emergency department patients who tested positive for tcdB gene by PCR were evaluated. Patients' stools underwent testing for GDH and TcdA/B by EIA. Medical health records were reviewed for demographic, clinical presentation, laboratory, treatment and outcome data. Severity of CDI was calculated using various severity score indexes. RESULTS: The median CT of cases was 32.05 ± 5.45. The optimal cut-off for predicting toxin EIA positivity and severe CDI based on chart review was 32.6 and 29.8, respectively, with the area under the receiver operator characteristics curve (AUC) of 0.74 and 0.60 respectively. CONCLUSION: CT value was an acceptable predictor for EIA toxin but less so for clinical severity. Our study potentially supports a diagnostic algorithm including CT value to reduce the number of EIA toxin assays performed.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Humans , Bacterial Toxins/genetics , Bacterial Toxins/analysis , Clostridioides difficile/genetics , Clostridioides/genetics , Immunoenzyme Techniques , Clostridium Infections/diagnosis , Real-Time Polymerase Chain Reaction , Feces/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/analysis
4.
Immunol Cell Biol ; 101(6): 491-503, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36825901

ABSTRACT

Activation-induced marker (AIM) assays have proven to be an accessible and rapid means of antigen-specific T-cell detection. The method typically involves short-term incubation of whole blood or peripheral blood mononuclear cells with antigens of interest, where autologous antigen-presenting cells process and present peptides in complex with major histocompatibility complex (MHC) molecules. Recognition of peptide-MHC complexes by T-cell receptors then induces upregulation of activation markers on the T cells that can be detected by flow cytometry. In this review, we highlight the most widely used activation markers for assays in the literature while identifying nuances and potential downfalls associated with the technique. We provide a summary of how AIM assays have been used in both discovery science and clinical studies, including studies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity. This review primarily focuses on AIM assays using human blood or peripheral blood mononuclear cell samples, with some considerations noted for tissue-derived T cells and nonhuman samples. AIM assays are a powerful tool that enables detailed analysis of antigen-specific T-cell frequency, phenotype and function without needing to know the precise antigenic peptides and their MHC restriction elements, enabling a wider analysis of immunity generated following infection and/or vaccination.


Subject(s)
COVID-19 , Leukocytes, Mononuclear , Humans , SARS-CoV-2 , T-Lymphocytes , Peptides , Antigens
5.
Sci Immunol ; 8(79): eadf0244, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36608148

ABSTRACT

The nature of flagellin-Toll-like receptor 5 (TLR5) interactions, depending on binding to and activation of TLR5, may hold a key to the distinct differences in gut microbiome and intestinal immune function in different populations around the world (see related Research Article by Clasen et al.).


Subject(s)
Flagellin , Toll-Like Receptor 5 , Flagellin/metabolism , Intestines
6.
J Autoimmun ; 134: 102961, 2023 01.
Article in English | MEDLINE | ID: mdl-36470208

ABSTRACT

Regulatory T cell (Treg) therapy is a promising strategy to treat inflammatory bowel disease (IBD). Data from animal models has shown that Tregs specific for intestinal antigens are more potent than polyclonal Tregs at inhibiting colitis. Flagellins, the major structural proteins of bacterial flagella, are immunogenic antigens frequently targeted in IBD subjects, leading to the hypothesis that flagellin-specific Tregs could be an effective cell therapy for IBD. We developed a novel chimeric antigen receptor (CAR) specific for flagellin derived from Escherichia coli H18 (FliC). We used this CAR to confer FliC-specificity to human Tregs and investigated their therapeutic potential. FliC-CAR Tregs were activated by recombinant FliC protein but not a control flagellin protein, demonstrating CAR specificity and functionality. In a humanized mouse model, expression of the FliC-CAR drove preferential migration to the colon and expression of the activation marker PD1. In the presence of recombinant FliC protein in vitro, FliC-CAR Tregs were significantly more suppressive than control Tregs and promoted the establishment of colon-derived epithelial cell monolayers. These results demonstrate the potential of FliC-CAR Tregs to treat IBD and more broadly show the therapeutic potential of CARs targeting microbial-derived antigens.


Subject(s)
Inflammatory Bowel Diseases , Receptors, Chimeric Antigen , Animals , Mice , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Flagellin/metabolism , Recombinant Proteins/metabolism , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/metabolism , T-Lymphocytes, Regulatory
7.
Immunity ; 55(6): 1051-1066.e4, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35649416

ABSTRACT

Microbial exposures are crucial environmental factors that impact healthspan by sculpting the immune system and microbiota. Antibody profiling via Phage ImmunoPrecipitation Sequencing (PhIP-Seq) provides a high-throughput, cost-effective approach for detecting exposure and response to microbial protein products. We designed and constructed a library of 95,601 56-amino acid peptide tiles spanning 14,430 proteins with "toxin" or "virulence factor" keyword annotations. We used PhIP-Seq to profile the antibodies of ∼1,000 individuals against this "ToxScan" library. In addition to enumerating immunodominant antibody epitopes, we studied the age-dependent stability of the ToxScan profile and used a genome-wide association study to find that the MHC-II locus modulates bacterial epitope selection. We detected previously described anti-flagellin antibody responses in a Crohn's disease cohort and identified an association between anti-flagellin antibodies and juvenile dermatomyositis. PhIP-Seq with the ToxScan library is thus an effective tool for studying the environmental determinants of health and disease at cohort scale.


Subject(s)
Bacteriophages , Peptide Library , Amino Acid Sequence , Antibodies , Antibody Formation , Bacteriophages/genetics , Genome-Wide Association Study , Humans , Immunodominant Epitopes , Prevalence , Virulence Factors/genetics
8.
Cell Rep ; 38(3): 110283, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045294

ABSTRACT

Acute damage to the intestinal epithelium can be repaired via de-differentiation of mature intestinal epithelial cells (IECs) to a stem-like state, but there is a lack of knowledge on how intestinal stem cells function after chronic injury, such as in inflammatory bowel disease (IBD). We developed a chronic-injury model in human colonoid monolayers by repeated rounds of air-liquid interface and submerged culture. We use this model to understand how chronic intestinal damage affects the ability of IECs to (1) respond to microbial stimulation, using the Toll-like receptor 5 (TLR5) agonist FliC and (2) regenerate and protect the epithelium from further damage. Repeated rounds of damage impair the ability of IECs to regrow and respond to TLR stimulation. We also identify mRNA expression and DNA methylation changes in genes associated with IBD and colon cancer. This methodology results in a human model of recurrent IEC injury like that which occurs in IBD.


Subject(s)
Cell Culture Techniques/methods , Intestinal Mucosa/physiology , Organoids/physiology , Colonic Neoplasms , DNA Methylation , Humans , Inflammatory Bowel Diseases , Regeneration/physiology , Stem Cells/physiology
10.
J Immunol Methods ; 488: 112931, 2021 01.
Article in English | MEDLINE | ID: mdl-33221458

ABSTRACT

Adoptive cell therapy with genetically modified regulatory T cells (Tregs) is under clinical investigation for the treatment of transplant rejection and various autoimmune conditions. A limitation of modelling this approach in mice is the lack of optimized protocols for expanding and transducing mouse Tregs. Here we describe a protocol for purifying, expanding and retrovirally transducing mouse Tregs with a vector encoding a chimeric antigen receptor as a model transgene. We found that isolation of Tregs from C57Bl/6J Foxp3EGFP mice solely based on eGFP expression resulted in sufficiently pure cells; co-sorting of CD25hi cells was not essential. Although expansion with rapamycin reduced Treg expansion, it promoted maximal in vitro suppressive activity. Retroviral transduction of Tregs following 2 days of stimulation with anti-CD3/CD28 beads achieved a transduction efficiency of ~40% and did not impair their suppressive capacity. When injected into a conventional T cell (Tconv)-transfer-induced colitis model, transduced Tregs inhibited colitis progression at ratios as low as 1 Treg to 100 Tconvs, and maintained Foxp3 and transgene expression throughout an 8-week period. This method facilitates the study of transduced Tregs in animal models and will enable the study of genetically engineered Treg therapy for a variety of inflammatory diseases.


Subject(s)
Cell Proliferation , Genetic Vectors , Receptors, Chimeric Antigen/metabolism , Retroviridae/genetics , T-Lymphocytes, Regulatory/metabolism , Transduction, Genetic , Adoptive Transfer , Animals , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/prevention & control , Disease Models, Animal , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genes, T-Cell Receptor beta , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunomagnetic Separation , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Retroviridae/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
12.
Front Cell Dev Biol ; 8: 583919, 2020.
Article in English | MEDLINE | ID: mdl-33282867

ABSTRACT

The intestinal epithelium is replenished every 3-4 days through an orderly process that maintains important secretory and absorptive functions while preserving a continuous mucosal barrier. Intestinal epithelial cells (IECs) derive from a stable population of intestinal stem cells (ISCs) that reside in the basal crypts. When intestinal injury reaches the crypts and damages IECs, a mechanism to replace them is needed. Recent research has highlighted the existence of distinct populations of acute and chronic damage-associated ISCs and their roles in maintaining homeostasis in several intestinal perturbation models. What remains unknown is how the damage-associated regenerative ISC population functions in the setting of chronic inflammation, as opposed to acute injury. What long-term consequences result from persistent inflammation and other cellular insults to the ISC niche? What particular "regenerative" cell types provide the most efficacious restorative properties? Which differentiated IECs maintain the ability to de-differentiate and restore the ISC niche? This review will cover the latest research on damage-associated regenerative ISCs and epigenetic factors that determine ISC fate, as well as provide opinions on future studies that need to be undertaken to understand the repercussions of the emergence of these cells, their contribution to relapses in inflammatory bowel disease, and their potential use in therapeutics for chronic intestinal diseases.

13.
Microorganisms ; 8(7)2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32635236

ABSTRACT

BACKGROUND: Mycobacterium avium subspecies paratuberculosis (MAP), a member of the mycobacteriaceae family, causes Johne's disease in ruminants, which resembles Crohn's disease (CD) in humans. MAP was proposed to be one of the causes of human CD, but the evidence remains elusive. Macrophages were reported to be the only cell where MAP proliferates in ruminants and humans and is likely the major producer of TNFα-associated inflammation. However, whether human dendritic cells (DCs), another major antigen-presenting cell (APC), have the ability to harbor MAP and disseminate infection, remains unknown. METHODS: Human monocyte-derived dendritic cells (moDCs) were infected with MAP and phagocytosis and intracellular survival were quantified by immunofluorescence (IF) and colony counts, respectively. MoDC cytokine expression was measured via ELISA and their activation state was measured via flow cytometry. RESULTS: We showed that MAP can infect and replicate in human moDCs as means to evade the immune system for successful infection, through inhibition of the phago-lysosome fusion via the secretion of protein tyrosine phosphatase PtpA. This mechanism initially led to a state of tolerance in moDCs and then subsequently caused a pro-inflammatory response as infection persisted, characterized by the upregulation of IL-6 and TNFα, and downregulation of IL-10. Moreover, we showed that moDCs have the ability to phagocytose up to 18% of MAP, when exposed at a multiplicity of infection of 1:1. CONCLUSION: Infection and subsequent proliferation of MAP within moDCs could provide a unique means for the dissemination of MAP to lymphoid tissue, while altering immune responses to facilitate the persistence of infection of host tissues in CD.

14.
Patient Prefer Adherence ; 14: 33-43, 2020.
Article in English | MEDLINE | ID: mdl-32021115

ABSTRACT

PURPOSE: Clostridioides difficile infection (CDI) is the most prevalent cause of nosocomial infectious diarrhea in Canada and is highly correlated with antibiotic use and contact with health care facilitates. The often-severe symptoms of CDI include diarrhea, dehydration, and abdominal pain. Patients often relapse following symptom resolution, resulting in increased morbidity. Previous research on the impact of CDI centered around the health-care system, clinician perspectives and economic burden, but not on patient experiences. The purpose of this study was to understand the impact of CDI on patients in Canada. METHODS: The Gastrointestinal Society conducted online surveys and gathered data from 167 qualifying participants, who were either patients or their non-treating caregivers. Quantitative parameters were analyzed by descriptive and comparative statistics and contextualized with qualitative insights derived from thematic analysis of open-ended questions. RESULTS: Our findings, which focused on clinical parameters such as prior exposure to health-care settings, antibiotic use, and patients' symptoms, mirrored findings from previous research. Interestingly, most surveyed respondents experienced delays in diagnosis and treatment; 29% waited 6-30 days and 10% over 30 days. This delayed diagnosis was further complicated by the report that 62% of respondents did not experience symptom resolution within 7 days of initiating treatment. Importantly, our results suggest a lasting impact after the resolution of CDI and we saw a reduction of self-assessed quality of life from prior to post CDI. Patients' priorities regarding their experience with CDI focused around concerns about the health-care system, particularly time to diagnosis and treatment, concerns about antibiotic usage and needs from health-care providers. CONCLUSION: This is the first Canadian report on patients' experience with CDI. Our data highlight the symptom-related impact on patients and the long-lasting effect on the quality of life including emotional impact. Reducing time to diagnosis and improving patient education are important priorities to attenuate the impact on patients.

15.
J Leukoc Biol ; 108(1): 309-321, 2020 07.
Article in English | MEDLINE | ID: mdl-32057139

ABSTRACT

Intestinal epithelial cells provide a front line of defense by establishing a barrier against food Ags, pathogens, and commensal microorganisms. This defense includes the establishment of a tolerogenic environment in the gastrointestinal (GI) tract. The intestinal epithelium replenishes itself by cell turnover every 4-5 days, and this process is facilitated by various pathways of communication between the intestinal epithelial cells (IECs), the underlying stromal cell network, and professional immune cells, which together help establish a proper intestinal stem cell (ISC) niche in the crypt. However, during a state of inflammation, such as in inflammatory bowel diseases (IBD), these communication pathways can be altered, and this can lead to the development of inflammatory IECs within the crypt that further drive inflammation. Here, we review the current literature looking at crosstalk between immune cells, stromal cells, and IECs: how does the immune system potentially alter the ISC niche, and how do IECs influence intestinal immunity? We discuss the latest research using single cell RNA sequencing and intestinal organoid cultures to help answer these questions. A better understanding of this complex crosstalk can help lead to a better understanding of intestinal biology in general, and more efficient therapeutic approaches to treat IBD.


Subject(s)
Cell Compartmentation/immunology , Epithelial Cells/immunology , Inflammatory Bowel Diseases/immunology , Intestines/pathology , Leukocytes/immunology , Mesenchymal Stem Cells/immunology , Animals , Humans
16.
Cell Mol Gastroenterol Hepatol ; 9(3): 485-506, 2020.
Article in English | MEDLINE | ID: mdl-31790809

ABSTRACT

BACKGROUND & AIMS: Bacterial flagellin is an important antigen in inflammatory bowel disease, but the role of flagellin-specific CD4+ T cells in disease pathogenesis remains unclear. Also unknown is how changes in intestinal microbiome intersect with those in microbiota-specific CD4+ T cells. We aimed to quantify and characterize flagellin-specific CD4+ T cells in Crohn's disease (CD) and ulcerative colitis (UC) patients and study their relationship with intestinal microbiome diversity. METHODS: Blood was collected from 3 cohorts that included CD patients, UC patients, and healthy controls. Flow cytometry analyzed CD4+ T cells specific for Lachnospiraceae-derived A4-Fla2 and Escherichia coli H18 FliC flagellins, or control vaccine antigens. Serum antiflagellin IgG and IgA antibodies were detected by enzyme-linked immunosorbent assay and stool samples were collected and subjected to 16S ribosomal DNA sequencing. RESULTS: Compared with healthy controls, CD and UC patients had lower frequencies of vaccine-antigen-specific CD4+ T cells and, as a proportion of vaccine-specific cells, higher frequencies of flagellin-specific CD4+ T cells. The proportion of flagellin-specific CD4+ T cells that were CXCR3negCCR4+CCR6+ Th17 cells was reduced in CD and UC patients, with increased proportions of CD39+, PD-1+, and integrin ß7+ cells. Microbiome analysis showed differentially abundant bacterial species in patient groups that correlated with immune responses to flagellin. CONCLUSIONS: Both CD and UC patients have relative increases in the proportion of circulating Fla2-specific CD4+ T cells, which may be associated with changes in the intestinal microbiome. Evidence that the phenotype of these cells strongly correlate with disease severity provides insight into the potential roles of flagellin-specific CD4+ T cells in inflammatory bowel disease.


Subject(s)
Antibodies, Bacterial/blood , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Dysbiosis/complications , Escherichia coli Proteins/immunology , Flagellin/immunology , Adaptive Immunity , Adolescent , Adult , Aged , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Clostridiales/genetics , Clostridiales/immunology , Colitis, Ulcerative/blood , Colitis, Ulcerative/microbiology , Crohn Disease/blood , Crohn Disease/microbiology , Cross-Sectional Studies , DNA, Bacterial/isolation & purification , Dysbiosis/diagnosis , Dysbiosis/immunology , Dysbiosis/microbiology , Enzyme-Linked Immunosorbent Assay , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Healthy Volunteers , Humans , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , T-Lymphocyte Subsets/immunology , Young Adult
17.
Crohns Colitis 360 ; 2(4): otaa085, 2020 Oct.
Article in English | MEDLINE | ID: mdl-36777762

ABSTRACT

Background: Inflammatory bowel disease is a life-changing disease resulting from recurrent intestinal inflammation. Current therapies (eg, steroids and biologics) are associated with mild to severe side effects, and none provide a cure. Recent research has focused on genetically engineering gut-specific anti-inflammatory T-regulatory cells (CAR-Tregs) to control intestinal inflammation, a logistically and conceptually complex approach. The purpose of our study was to understand patients' willingness to try CAR-Treg given 2 hypothetical scenarios-in a clinical trial or as a new treatment. Methods: We surveyed people living with inflammatory bowel disease about their willingness to try CAR-Treg. The online survey was developed using patient focus groups and associated literature. We recruited participants through email and social media. We used descriptive and inferential statistics to analyze closed-ended questions and inductive thematic analysis to analyze open-ended follow-up questions. Results: Survey participants indicated high willingness to try CAR-Treg therapy in both a clinical trial and as a new treatment. Willingness to try was not correlated with disease state or medication history. Women were less likely than men to indicate willingness to participate in a clinical trial. Participants' reasons for being willing to try CAR-Treg therapy included the wish to change their current treatment and the calling to participate in research. Participants that were not willing to try CAR-Treg mentioned the lack of long-term data and the success of their current therapy. Conclusions: This is the first study to our knowledge to investigate patient willingness to try CAR-Treg therapy. Our results demonstrate the promise of moving this therapy into clinical practice as most patients indicated willingness to try.

18.
J Crohns Colitis ; 14(7): 948-961, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-31796949

ABSTRACT

BACKGROUND AND AIMS: Endoplasmic reticulum [ER] stress in intestinal epithelial cells [IECs] contributes to the pathogenesis of inflammatory bowel disease [IBD]. We hypothesized that ER stress changes innate signalling in human IECs, augmenting toll-like receptor [TLR] responses and inducing pro-inflammatory changes in underlying dendritic cells [DCs]. METHODS: Caco-2 cells and primary human colon-derived enteroid monolayers were exposed to ATP [control stressor] or thapsigargin [Tg] [ER stress inducer], and were stimulated with the TLR5 agonist flagellin. Cytokine release was measured by an enzyme immunoassay. ER stress markers CHOP, GRP78 and XBP1s/u were measured via quantitative PCR and Western blot. Monocyte-derived DCs [moDCs] were cultured with the IEC supernatants and their activation state was measured. Responses from enteroids derived from IBD patients and healthy control participants were compared. RESULTS: ER stress enhanced flagellin-induced IL-8 release from Caco-2 cells and enteroids. Moreover, conditioned media activated DCs to become pro-inflammatory, with increased expression of CD80, CD86, MHCII, IL-6, IL-15 and IL-12p70 and decreased expression of CD103 and IL-10. Flagellin-induced IL-8 production correlated with DC activation, suggesting a common stress pathway. Moreover, there were distinct differences in cytokine expression and basal ER stress between IBD and healthy subject-derived enteroid monolayers, suggesting a dysregulated ER stress pathway in IBD-derived enteroids. CONCLUSIONS: Cellular stress enhances TLR5 responses in IECs, leading to increased DC activation, indicating a previously unknown mechanistic link between epithelial ER stress and immune activation in IBD. Furthermore, dysregulated ER stress may be propagated from the intestinal epithelial stem cell niche in IBD patients.


Subject(s)
Cytokines/metabolism , Dendritic Cells/physiology , Endoplasmic Reticulum Stress/physiology , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/physiopathology , Toll-Like Receptor 5/metabolism , Adenosine Triphosphate/pharmacology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Caco-2 Cells , Cell Differentiation , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Colon/pathology , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Dendritic Cells/drug effects , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Flagellin/pharmacology , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Integrin alpha Chains/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-15/metabolism , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lactones/pharmacology , Organoids/metabolism , RNA, Messenger/metabolism , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 5/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Gastroenterology ; 157(6): 1584-1598, 2019 12.
Article in English | MEDLINE | ID: mdl-31513797

ABSTRACT

BACKGROUND & AIMS: T-regulatory (Treg) cells suppress the immune response to maintain homeostasis. There are 2 main subsets of Treg cells: FOXP3 (forkhead box protein 3)-positive Treg cells, which do not produce high levels of effector cytokines, and type 1 Treg (Tr1) cells, which are FOXP3-negative and secrete interleukin (IL) 10. IL10 is an anti-inflammatory cytokine, so Tr1 cells might be used in the treatment of inflammatory bowel diseases. We aimed to develop methods to isolate and expand human Tr1 cells and define their functions. METHODS: We obtained blood and colon biopsy samples from patients with Crohn's disease or ulcerative colitis or healthy individuals (controls). CD4+ T cells were isolated from blood samples and stimulated with anti-CD3 and anti-CD28 beads, and Tr1 cells were purified by using an IL10 cytokine-capture assay and cell sorting. FOXP3-positive Treg cells were sorted as CD4+CD25highCD127low cells from unstimulated cells. Tr1 and FOXP3-positive Treg cells were expanded, and phenotypes and gene expression profiles were compared. T cells in peripheral blood mononuclear cells from healthy donors were stimulated with anti-CD3 and anti-CD28 beads, and the suppressive abilities of Tr1 and FOXP3-positive Treg cells were measured. Human colon organoid cultures were established, cultured with supernatants from Tr1 or FOXP3-positive cells, and analyzed by immunofluorescence and flow cytometry. T84 cells (human colon adenocarcinoma epithelial cells) were incubated with supernatants from Tr1 or FOXP3-positive cells, and transepithelial electrical resistance was measured to determine epithelial cell barrier function. RESULTS: Phenotypes of Tr1 cells isolated from control individuals vs patients with Crohn's disease or ulcerative colitis did not differ significantly after expansion. Tr1 cells and FOXP3-positive Treg cells suppressed proliferation of effector T cells, but only Tr1 cells suppressed secretion of IL1B and tumor necrosis factor from myeloid cells. Tr1 cells, but not FOXP3-positive Treg cells, isolated from healthy individuals and patients with Crohn's disease or ulcerative colitis secreted IL22, which promoted barrier function of human intestinal epithelial cells. Tr1 cell culture supernatants promoted differentiation of mucin-producing goblet cells in intestinal organoid cultures. CONCLUSIONS: Human Tr1 cells suppress proliferation of effector T cells (adaptive immune response) and production of IL1B and TNF by myeloid cells (inmate immune response). They also secrete IL22 to promote barrier function. They might be developed as a cell-based therapy for intestinal inflammatory disorders.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Interleukin-10/metabolism , Intestinal Mucosa/pathology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Biopsy , Cell Communication/immunology , Cell Proliferation , Cells, Cultured , Colitis, Ulcerative/blood , Colitis, Ulcerative/therapy , Colon/cytology , Colon/immunology , Colon/pathology , Crohn Disease/blood , Crohn Disease/therapy , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Healthy Volunteers , Humans , Interleukin-10/immunology , Interleukins/immunology , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Male , Middle Aged , Primary Cell Culture , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
20.
Mucosal Immunol ; 12(5): 1187-1200, 2019 09.
Article in English | MEDLINE | ID: mdl-31358861

ABSTRACT

Crohn's disease is an immune-mediated disease characterized by inflammation along the gastrointestinal tract. Fibrosis requiring surgery occurs in one-third of people with Crohn's disease but there are no treatments for intestinal fibrosis. Mice deficient in the SH2 domain-containing inositolpolyphosphate 5'-phosphatase (SHIP), a negative regulator of phosphatidylinositol 3-kinase (PI3K) develop spontaneous Crohn's disease-like intestinal inflammation and arginase I (argI)-dependent fibrosis. ArgI is up-regulated in SHIP deficiency by PI3Kp110δ activity. Thus, we hypothesized that SHIP-deficient mice develop fibrosis due to increased PI3Kp110δ activity. In SHIP-deficient mice, genetic ablation or pharmacological inhibition of PI3Kp110δ activity reduced intestinal fibrosis, including muscle thickening, accumulation of vimentin+ mesenchymal cells, and collagen deposition. PI3Kp110δ deficiency or inhibition also reduced ileal inflammation in SHIP-deficient mice suggesting that PI3Kp110δ may contribute to inflammation. Targeting PI3Kp110δ activity may be an effective strategy to reduce intestinal fibrosis, and may be particularly effective in the subset of people with Crohn's disease, who have low SHIP activity.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Inflammation/etiology , Inflammation/metabolism , Intestines/pathology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/deficiency , Animals , Arginase/genetics , Arginase/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Enzyme Activation , Fibrosis , Gene Expression , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...