Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 52(1): 151-166.e6, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31924474

ABSTRACT

In addition to helper and regulatory potential, CD4+ T cells also acquire cytotoxic activity marked by granzyme B (GzmB) expression and the ability to promote rejection of established tumors. Here, we examined the molecular and cellular mechanisms underpinning the differentiation of cytotoxic CD4+ T cells following immunotherapy. CD4+ transfer into lymphodepleted animals or regulatory T (Treg) cell depletion promoted GzmB expression by tumor-infiltrating CD4+, and this was prevented by interleukin-2 (IL-2) neutralization. Transcriptional analysis revealed a polyfunctional helper and cytotoxic phenotype characterized by the expression of the transcription factors T-bet and Blimp-1. While T-bet ablation restricted interferon-γ (IFN-γ) production, loss of Blimp-1 prevented GzmB expression in response to IL-2, suggesting two independent programs required for polyfunctionality of tumor-reactive CD4+ T cells. Our findings underscore the role of Treg cells, IL-2, and Blimp-1 in controlling the differentiation of cytotoxic CD4+ T cells and offer a pathway to enhancement of anti-tumor activity through their manipulation.


Subject(s)
Granzymes/immunology , Neoplasms/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Adoptive Transfer , Animals , Cell Line, Tumor , Humans , Interferon-gamma/immunology , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , Tumor Microenvironment/immunology
2.
Nat Immunol ; 16(2): 207-213, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25531831

ABSTRACT

Lymphocyte recruitment maintains intestinal immune homeostasis but also contributes to inflammation. The orphan chemoattractant receptor GPR15 mediates regulatory T cell homing and immunosuppression in the mouse colon. We show that GPR15 is also expressed by mouse TH17 and TH1 effector cells and is required for colitis in a model that depends on the trafficking of these cells to the colon. In humans GPR15 is expressed by effector cells, including pathogenic TH2 cells in ulcerative colitis, but is expressed poorly or not at all by colon regulatory T (Treg) cells. The TH2 transcriptional activator GATA-3 and the Treg-associated transcriptional repressor FOXP3 robustly bind human, but not mouse, GPR15 enhancer sequences, correlating with receptor expression. Our results highlight species differences in GPR15 regulation and suggest it as a potential therapeutic target for colitis.


Subject(s)
Colitis/physiopathology , Colon/physiopathology , Gene Expression Regulation , Receptors, G-Protein-Coupled/metabolism , Receptors, Lymphocyte Homing/metabolism , Receptors, Peptide/metabolism , Animals , Cells, Cultured , Colitis/immunology , Colon/immunology , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Forkhead Transcription Factors/metabolism , Gene Knockout Techniques , Humans , Mice , Protein Binding , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Species Specificity
3.
Trends Immunol ; 43(7): 564-579, 2022 07.
Article in English | MEDLINE | ID: mdl-35618586

ABSTRACT

Mammalian innate lymphoid cells (ILCs) have functional relevance under both homeostatic and disease settings, such as inflammatory bowel disease (IBD), particularly in the context of maintaining the integrity of mucosal surfaces. Early reports highlighted group 1 and 3 ILC regulatory transcription factors (TFs), T-box expressed in T cells (T-bet; Tbx21) and RAR-related orphan nuclear receptor γt (RORγt; Rorc), as key regulators of ILC biology. Since then, other canonical TFs have been shown to have a role in the development and function of ILC subsets. In this review, we focus on recent insights into the balance between mature ILC1 and ILC3 based on these TFs and how they interact with other key cell-intrinsic molecular pathways. We outline how this TF interplay might be explored to identify novel candidate therapeutic avenues for human diseases.


Subject(s)
Immunity, Innate , Inflammatory Bowel Diseases , Transcription Factors , Animals , Gene Expression Regulation , Humans , Lymphocytes/metabolism , Transcription Factors/metabolism
4.
Immunity ; 42(3): 499-511, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25769610

ABSTRACT

CD4(+) T cells differentiate into phenotypically distinct T helper cells upon antigenic stimulation. Regulation of plasticity between these CD4(+) T-cell lineages is critical for immune homeostasis and prevention of autoimmune disease. However, the factors that regulate lineage stability are largely unknown. Here we investigate a role for retinoic acid (RA) in the regulation of lineage stability using T helper 1 (Th1) cells, traditionally considered the most phenotypically stable Th subset. We found that RA, through its receptor RARα, sustains stable expression of Th1 lineage specifying genes, as well as repressing genes that instruct Th17-cell fate. RA signaling is essential for limiting Th1-cell conversion into Th17 effectors and for preventing pathogenic Th17 responses in vivo. Our study identifies RA-RARα as a key component of the regulatory network governing maintenance and plasticity of Th1-cell fate and defines an additional pathway for the development of Th17 cells.


Subject(s)
Cell Lineage/drug effects , Receptors, Retinoic Acid/genetics , T-Lymphocytes, Helper-Inducer/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Tretinoin/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation , Gene Regulatory Networks , Homeostasis/drug effects , Homeostasis/immunology , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Retinoic Acid/immunology , Retinoic Acid Receptor alpha , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Tretinoin/immunology
5.
Nucleic Acids Res ; 50(8): 4557-4573, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35438764

ABSTRACT

Lineage-determining transcription factors (LD-TFs) drive the differentiation of progenitor cells into a specific lineage. In CD4+ T cells, T-bet dictates differentiation of the TH1 lineage, whereas GATA3 drives differentiation of the alternative TH2 lineage. However, LD-TFs, including T-bet and GATA3, are frequently co-expressed but how this affects LD-TF function is not known. By expressing T-bet and GATA3 separately or together in mouse T cells, we show that T-bet sequesters GATA3 at its target sites, thereby removing GATA3 from TH2 genes. This redistribution of GATA3 is independent of GATA3 DNA binding activity and is instead mediated by the T-bet DNA binding domain, which interacts with the GATA3 DNA binding domain and changes GATA3's sequence binding preference. This mechanism allows T-bet to drive the TH1 gene expression program in the presence of GATA3. We propose that redistribution of one LD-TF by another may be a common mechanism that could explain how specific cell fate choices can be made even in the presence of other transcription factors driving alternative differentiation pathways.


Subject(s)
GATA3 Transcription Factor , T-Box Domain Proteins/metabolism , Th2 Cells , Animals , Cell Lineage , DNA/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression , Mice , T-Box Domain Proteins/genetics , Th2 Cells/cytology , Th2 Cells/metabolism
6.
Eur J Immunol ; 52(4): 566-581, 2022 04.
Article in English | MEDLINE | ID: mdl-35092032

ABSTRACT

T-bet is the lineage-specifying transcription factor for CD4+ TH 1 cells. T-bet has also been found in other CD4+ T cell subsets, including TH 17 cells and Treg, where it modulates their functional characteristics. However, we lack information on when and where T-bet is expressed during T cell differentiation and how this impacts T cell differentiation and function. To address this, we traced the ontogeny of T-bet-expressing cells using a fluorescent fate-mapping mouse line. We demonstrate that T-bet is expressed in a subset of CD4+ T cells that have naïve cell surface markers and transcriptional profile and that this novel cell population is phenotypically and functionally distinct from previously described populations of naïve and memory CD4+ T cells. Naïve-like T-bet-experienced cells are polarized to the TH 1 lineage, predisposed to produce IFN-γ upon cell activation, and resist repolarization to other lineages in vitro and in vivo. These results demonstrate that lineage-specifying factors can polarize T cells in the absence of canonical markers of T cell activation and that this has an impact on the subsequent T-helper response.


Subject(s)
T-Box Domain Proteins , Th1 Cells , Animals , Cell Differentiation , Gene Expression Regulation , Lymphocyte Activation , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Th2 Cells
7.
Eur Respir J ; 61(5)2023 05.
Article in English | MEDLINE | ID: mdl-36922030

ABSTRACT

BACKGROUND: COVID-19 is associated with a dysregulated immune response but it is unclear how immune dysfunction contributes to the chronic morbidity persisting in many COVID-19 patients during convalescence (long COVID). METHODS: We assessed phenotypical and functional changes of monocytes in COVID-19 patients during hospitalisation and up to 9 months of convalescence following COVID-19, respiratory syncytial virus or influenza A. Patients with progressive fibrosing interstitial lung disease were included as a positive control for severe, ongoing lung injury. RESULTS: Monocyte alterations in acute COVID-19 patients included aberrant expression of leukocyte migration molecules, continuing into convalescence (n=142) and corresponding with specific symptoms of long COVID. Long COVID patients with unresolved lung injury, indicated by sustained shortness of breath and abnormal chest radiology, were defined by high monocyte expression of C-X-C motif chemokine receptor 6 (CXCR6) (p<0.0001) and adhesion molecule P-selectin glycoprotein ligand 1 (p<0.01), alongside preferential migration of monocytes towards the CXCR6 ligand C-X-C motif chemokine ligand 16 (CXCL16) (p<0.05), which is abundantly expressed in the lung. Monocyte CXCR6 and lung CXCL16 were heightened in patients with progressive fibrosing interstitial lung disease (p<0.001), confirming a role for the CXCR6-CXCL16 axis in ongoing lung injury. Conversely, monocytes from long COVID patients with ongoing fatigue exhibited a sustained reduction of the prostaglandin-generating enzyme cyclooxygenase 2 (p<0.01) and CXCR2 expression (p<0.05). These monocyte changes were not present in respiratory syncytial virus or influenza A convalescence. CONCLUSIONS: Our data define unique monocyte signatures that define subgroups of long COVID patients, indicating a key role for monocyte migration in COVID-19 pathophysiology. Targeting these pathways may provide novel therapeutic opportunities in COVID-19 patients with persistent morbidity.


Subject(s)
COVID-19 , Influenza, Human , Lung Injury , Humans , Monocytes/metabolism , Chemokines, CXC/metabolism , Receptors, Virus/metabolism , Receptors, CXCR6 , Receptors, Chemokine/metabolism , Post-Acute COVID-19 Syndrome , Ligands , Convalescence , Receptors, Scavenger/metabolism , Chemokine CXCL16 , Patient Acuity
8.
J Immunol ; 206(11): 2725-2739, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34021046

ABSTRACT

Innate lymphoid cells are central to the regulation of immunity at mucosal barrier sites, with group 2 innate lymphoid cells (ILC2s) being particularly important in type 2 immunity. In this study, we demonstrate that microRNA(miR)-142 plays a critical, cell-intrinsic role in the homeostasis and function of ILC2s. Mice deficient for miR-142 expression demonstrate an ILC2 progenitor-biased development in the bone marrow, and along with peripheral ILC2s at mucosal sites, these cells display a greatly altered phenotype based on surface marker expression. ILC2 proliferative and effector functions are severely dysfunctional following Nippostrongylus brasiliensis infection, revealing a critical role for miR-142 isoforms in ILC2-mediated immune responses. Mechanistically, Socs1 and Gfi1 expression are regulated by miR-142 isoforms in ILC2s, impacting ILC2 phenotypes as well as the proliferative and effector capacity of these cells. The identification of these novel pathways opens potential new avenues to modulate ILC2-dependent immune functions.


Subject(s)
Lymphocytes/immunology , MicroRNAs/immunology , Animals , HEK293 Cells , Homeostasis , Humans , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics
9.
PLoS Med ; 19(2): e1003911, 2022 02.
Article in English | MEDLINE | ID: mdl-35192610

ABSTRACT

BACKGROUND: There is limited evidence on the use of high-sensitivity C-reactive protein (hsCRP) as a biomarker for selecting patients for advanced cardiovascular (CV) therapies in the modern era. The prognostic value of mildly elevated hsCRP beyond troponin in a large real-world cohort of unselected patients presenting with suspected acute coronary syndrome (ACS) is unknown. We evaluated whether a mildly elevated hsCRP (up to 15 mg/L) was associated with mortality risk, beyond troponin level, in patients with suspected ACS. METHODS AND FINDINGS: We conducted a retrospective cohort study based on the National Institute for Health Research Health Informatics Collaborative data of 257,948 patients with suspected ACS who had a troponin measured at 5 cardiac centres in the United Kingdom between 2010 and 2017. Patients were divided into 4 hsCRP groups (<2, 2 to 4.9, 5 to 9.9, and 10 to 15 mg/L). The main outcome measure was mortality within 3 years of index presentation. The association between hsCRP levels and all-cause mortality was assessed using multivariable Cox regression analysis adjusted for age, sex, haemoglobin, white cell count (WCC), platelet count, creatinine, and troponin. Following the exclusion criteria, there were 102,337 patients included in the analysis (hsCRP <2 mg/L (n = 38,390), 2 to 4.9 mg/L (n = 27,397), 5 to 9.9 mg/L (n = 26,957), and 10 to 15 mg/L (n = 9,593)). On multivariable Cox regression analysis, there was a positive and graded relationship between hsCRP level and mortality at baseline, which remained at 3 years (hazard ratio (HR) (95% CI) of 1.32 (1.18 to 1.48) for those with hsCRP 2.0 to 4.9 mg/L and 1.40 (1.26 to 1.57) and 2.00 (1.75 to 2.28) for those with hsCRP 5 to 9.9 mg/L and 10 to 15 mg/L, respectively. This relationship was independent of troponin in all suspected ACS patients and was further verified in those who were confirmed to have an ACS diagnosis by clinical coding. The main limitation of our study is that we did not have data on underlying cause of death; however, the exclusion of those with abnormal WCC or hsCRP levels >15 mg/L makes it unlikely that sepsis was a major contributor. CONCLUSIONS: These multicentre, real-world data from a large cohort of patients with suspected ACS suggest that mildly elevated hsCRP (up to 15 mg/L) may be a clinically meaningful prognostic marker beyond troponin and point to its potential utility in selecting patients for novel treatments targeting inflammation. TRIAL REGISTRATION: ClinicalTrials.gov - NCT03507309.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/mortality , C-Reactive Protein/metabolism , Acute Coronary Syndrome/diagnosis , Aged , Aged, 80 and over , Biomarkers/blood , Cohort Studies , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Mortality/trends , Predictive Value of Tests , Retrospective Studies , Risk Factors , United Kingdom/epidemiology
10.
Nat Mater ; 20(2): 250-259, 2021 02.
Article in English | MEDLINE | ID: mdl-32895507

ABSTRACT

Organoids can shed light on the dynamic interplay between complex tissues and rare cell types within a controlled microenvironment. Here, we develop gut organoid cocultures with type-1 innate lymphoid cells (ILC1) to dissect the impact of their accumulation in inflamed intestines. We demonstrate that murine and human ILC1 secrete transforming growth factor ß1, driving expansion of CD44v6+ epithelial crypts. ILC1 additionally express MMP9 and drive gene signatures indicative of extracellular matrix remodelling. We therefore encapsulated human epithelial-mesenchymal intestinal organoids in MMP-sensitive, synthetic hydrogels designed to form efficient networks at low polymer concentrations. Harnessing this defined system, we demonstrate that ILC1 drive matrix softening and stiffening, which we suggest occurs through balanced matrix degradation and deposition. Our platform enabled us to elucidate previously undescribed interactions between ILC1 and their microenvironment, which suggest that they may exacerbate fibrosis and tumour growth when enriched in inflamed patient tissues.


Subject(s)
Extracellular Matrix/metabolism , Intestinal Mucosa/metabolism , Lymphocytes/metabolism , Organoids/metabolism , Animals , Female , Humans , Intestinal Mucosa/cytology , Lymphocytes/cytology , Matrix Metalloproteinase 9/metabolism , Mice , Organoids/cytology , Transforming Growth Factor beta1/metabolism
11.
J Immunol ; 205(6): 1608-1619, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32817333

ABSTRACT

CD4+ T cell functional inhibition (exhaustion) is a hallmark of malaria and correlates with impaired parasite control and infection chronicity. However, the mechanisms of CD4+ T cell exhaustion are still poorly understood. In this study, we show that Ag-experienced (Ag-exp) CD4+ T cell exhaustion during Plasmodium yoelii nonlethal infection occurs alongside the reduction in mammalian target of rapamycin (mTOR) activity and restriction in CD4+ T cell glycolytic capacity. We demonstrate that the loss of glycolytic metabolism and mTOR activity within the exhausted Ag-expCD4+ T cell population during infection coincides with reduction in T-bet expression. T-bet was found to directly bind to and control the transcription of various mTOR and metabolism-related genes within effector CD4+ T cells. Consistent with this, Ag-expTh1 cells exhibited significantly higher and sustained mTOR activity than effector T-bet- (non-Th1) Ag-expT cells throughout the course of malaria. We identified mTOR to be redundant for sustaining T-bet expression in activated Th1 cells, whereas mTOR was necessary but not sufficient for maintaining IFN-γ production by Th1 cells. Immunotherapy targeting PD-1, CTLA-4, and IL-27 blocked CD4+ T cell exhaustion during malaria infection and was associated with elevated T-bet expression and a concomitant increased CD4+ T cell glycolytic metabolism. Collectively, our data suggest that mTOR activity is linked to T-bet in Ag-expCD4+ T cells but that reduction in mTOR activity may not directly underpin Ag-expTh1 cell loss and exhaustion during malaria infection. These data have implications for therapeutic reactivation of exhausted CD4+ T cells during malaria infection and other chronic conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/therapeutic use , Malaria/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Plasmodium yoelii/physiology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Animals , Cellular Senescence , Gene Expression Regulation , Glycolysis , Humans , Immune Tolerance , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-27/metabolism , Lymphocyte Activation , Malaria/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics
12.
Lancet ; 396(10251): 623-634, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32861307

ABSTRACT

BACKGROUND: Previous trials suggest lower long-term risk of mortality after invasive rather than non-invasive management of patients with non-ST elevation myocardial infarction (NSTEMI), but the trials excluded very elderly patients. We aimed to estimate the effect of invasive versus non-invasive management within 3 days of peak troponin concentration on the survival of patients aged 80 years or older with NSTEMI. METHODS: Routine clinical data for this study were obtained from five collaborating hospitals hosting NIHR Biomedical Research Centres in the UK (all tertiary centres with emergency departments). Eligible patients were 80 years old or older when they underwent troponin measurements and were diagnosed with NSTEMI between 2010 (2008 for University College Hospital) and 2017. Propensity scores (patients' estimated probability of receiving invasive management) based on pretreatment variables were derived using logistic regression; patients with high probabilities of non-invasive or invasive management were excluded. Patients who died within 3 days of peak troponin concentration without receiving invasive management were assigned to the invasive or non-invasive management groups based on their propensity scores, to mitigate immortal time bias. We estimated mortality hazard ratios comparing invasive with non-invasive management, and compared the rate of hospital admissions for heart failure. FINDINGS: Of the 1976 patients with NSTEMI, 101 died within 3 days of their peak troponin concentration and 375 were excluded because of extreme propensity scores. The remaining 1500 patients had a median age of 86 (IQR 82-89) years of whom (845 [56%] received non-invasive management. During median follow-up of 3·0 (IQR 1·2-4·8) years, 613 (41%) patients died. The adjusted cumulative 5-year mortality was 36% in the invasive management group and 55% in the non-invasive management group (adjusted hazard ratio 0·68, 95% CI 0·55-0·84). Invasive management was associated with lower incidence of hospital admissions for heart failure (adjusted rate ratio compared with non-invasive management 0·67, 95% CI 0·48-0·93). INTERPRETATION: The survival advantage of invasive compared with non-invasive management appears to extend to patients with NSTEMI who are aged 80 years or older. FUNDING: NIHR Imperial Biomedical Research Centre, as part of the NIHR Health Informatics Collaborative.


Subject(s)
Non-ST Elevated Myocardial Infarction/mortality , Non-ST Elevated Myocardial Infarction/therapy , Age Factors , Aged, 80 and over , Cohort Studies , Female , Hospitalization , Humans , Logistic Models , Male , Non-ST Elevated Myocardial Infarction/diagnosis , Propensity Score , Survival Rate , Troponin/blood , United Kingdom
13.
Immunity ; 37(4): 674-84, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23063332

ABSTRACT

Mice lacking the transcription factor T-bet in the innate immune system develop microbiota-dependent colitis. Here, we show that interleukin-17A (IL-17A)-producing IL-7Rα(+) innate lymphoid cells (ILCs) were potent promoters of disease in Tbx21(-/-)Rag2(-/-) ulcerative colitis (TRUC) mice. TNF-α produced by CD103(-)CD11b(+) dendritic cells synergized with IL-23 to drive IL-17A production by ILCs, demonstrating a previously unrecognized layer of cellular crosstalk between dendritic cells and ILCs. We have identified Helicobacter typhlonius as a key disease trigger driving excess TNF-α production and promoting colitis in TRUC mice. Crucially, T-bet also suppressed the expression of IL-7R, a key molecule involved in controlling intestinal ILC homeostasis. The importance of IL-7R signaling in TRUC disease was highlighted by the dramatic reduction in intestinal ILCs and attenuated colitis following IL-7R blockade. Taken together, these data demonstrate the mechanism by which T-bet regulates the complex interplay between mucosal dendritic cells, ILCs, and the intestinal microbiota.


Subject(s)
Colitis, Ulcerative/immunology , DNA-Binding Proteins/immunology , Immunity, Innate , Lymphocytes/immunology , Receptors, Interleukin-7/immunology , T-Box Domain Proteins/immunology , Animals , Cells, Cultured , Chronic Disease , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , DNA-Binding Proteins/deficiency , Helicobacter/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , T-Box Domain Proteins/deficiency
14.
Gut ; 69(5): 942-952, 2020 05.
Article in English | MEDLINE | ID: mdl-31980447

ABSTRACT

The prevalence of IBD is rising in the Western world. Despite an increasing repertoire of therapeutic targets, a significant proportion of patients suffer chronic morbidity. Studies in mice and humans have highlighted the critical role of regulatory T cells in immune homeostasis, with defects in number and suppressive function of regulatory T cells seen in patients with Crohn's disease. We review the function of regulatory T cells and the pathways by which they exert immune tolerance in the intestinal mucosa. We explore the principles and challenges of manufacturing a cell therapy, and discuss clinical trial evidence to date for their safety and efficacy in human disease, with particular focus on the development of a regulatory T-cell therapy for Crohn's disease.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Crohn Disease/drug therapy , Crohn Disease/immunology , Intestinal Mucosa/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Crohn Disease/diagnosis , Female , Forecasting , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Male , Mice , Treatment Outcome
15.
Gut ; 69(3): 578-590, 2020 03.
Article in English | MEDLINE | ID: mdl-31792136

ABSTRACT

OBJECTIVE: The functional role of interleukin-22 (IL22) in chronic inflammation is controversial, and mechanistic insights into how it regulates target tissue are lacking. In this study, we evaluated the functional role of IL22 in chronic colitis and probed mechanisms of IL22-mediated regulation of colonic epithelial cells. DESIGN: To investigate the functional role of IL22 in chronic colitis and how it regulates colonic epithelial cells, we employed a three-dimentional mini-gut epithelial organoid system, in vivo disease models and transcriptomic datasets in human IBD. RESULTS: As well as inducing transcriptional modules implicated in antimicrobial responses, IL22 also coordinated an endoplasmic reticulum (ER) stress response transcriptional programme in colonic epithelial cells. In the colon of patients with active colonic Crohn's disease (CD), there was enrichment of IL22-responsive transcriptional modules and ER stress response modules. Strikingly, in an IL22-dependent model of chronic colitis, targeting IL22 alleviated colonic epithelial ER stress and attenuated colitis. Pharmacological modulation of the ER stress response similarly impacted the severity of colitis. In patients with colonic CD, antibody blockade of IL12p40, which simultaneously blocks IL12 and IL23, the key upstream regulator of IL22 production, alleviated the colonic epithelial ER stress response. CONCLUSIONS: Our data challenge perceptions of IL22 as a predominantly beneficial cytokine in IBD and provide novel insights into the molecular mechanisms of IL22-mediated pathogenicity in chronic colitis. Targeting IL22-regulated pathways and alleviating colonic epithelial ER stress may represent promising therapeutic strategies in patients with colitis. TRIAL REGISTRATION NUMBER: NCT02749630.


Subject(s)
Colitis/genetics , Crohn Disease/physiopathology , Endoplasmic Reticulum Stress/genetics , Epithelial Cells/physiology , Interleukins/pharmacology , Transcription, Genetic , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Chronic Disease , Colitis/blood , Colitis/drug therapy , Colitis/pathology , Colon/pathology , Crohn Disease/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Humans , Interleukin-17/pharmacology , Interleukin-23/antagonists & inhibitors , Interleukins/blood , Interleukins/genetics , Intestinal Mucosa/pathology , Mice , Organoids , Patient Acuity , Phenylbutyrates/pharmacology , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects , Tunicamycin/pharmacology , Unfolded Protein Response , Ustekinumab/pharmacology , Ustekinumab/therapeutic use , Interleukin-22
16.
Am J Transplant ; 20(10): 2715-2727, 2020 10.
Article in English | MEDLINE | ID: mdl-32277570

ABSTRACT

Organ transplantation is often lifesaving, but the long-term deleterious effects of combinatorial immunosuppression regimens and allograft failure cause significant morbidity and mortality. Long-term graft survival in the absence of continuing immunosuppression, defined as operational tolerance, has never been described in the context of multiple major histocompatibility complex (MHC) mismatches. Here, we show that miR-142 deficiency leads to indefinite allograft survival in a fully MHC mismatched murine cardiac transplant model in the absence of exogenous immunosuppression. We demonstrate that the cause of indefinite allograft survival in the absence of miR-142 maps specifically to the T cell compartment. Of therapeutic relevance, temporal deletion of miR-142 in adult mice prior to transplantation of a fully MHC mismatched skin allograft resulted in prolonged allograft survival. Mechanistically, miR-142 directly targets Tgfbr1 for repression in regulatory T cells (TREG ). This leads to increased TREG sensitivity to transforming growth factor - beta and promotes transplant tolerance via an augmented peripheral TREG response in the absence of miR-142. These data identify manipulation of miR-142 as a promising approach for the induction of tolerance in human transplantation.


Subject(s)
Graft Rejection , MicroRNAs , Allografts , Animals , Graft Rejection/etiology , Graft Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , T-Lymphocytes, Regulatory , Transplantation Tolerance , Transplantation, Homologous
17.
Gastroenterology ; 156(6): 1775-1787, 2019 05.
Article in English | MEDLINE | ID: mdl-30710527

ABSTRACT

BACKGROUND & AIMS: Crohn's disease (CD) is characterized by an imbalance of effector and regulatory T cells in the intestinal mucosa. The efficacy of anti-adhesion therapies led us to investigate whether impaired trafficking of T-regulatory (Treg) cells contributes to the pathogenesis of CD. We also investigated whether proper function could be restored to Treg cells by ex vivo expansion in the presence of factors that activate their regulatory activities. METHODS: We measured levels of the integrin α4ß7 on Treg cells isolated from peripheral blood or lamina propria of patients with CD and healthy individuals (controls). Treg cells were expanded ex vivo and incubated with rapamycin with or without agonists of the retinoic acid receptor-α (RARA), and their gene expression profiles were analyzed. We also studied the cells in cytokine challenge, suppression, and flow chamber assays and in SCID mice with human intestinal xenografts. RESULTS: We found that Treg cells from patients with CD express lower levels of the integrin α4ß7 than Treg cells from control patients. The pathway that regulates the expression of integrin subunit α is induced by retinoic acid (RA). Treg cells from patients with CD incubated with rapamycin and an agonist of RARA (RAR568) expressed high levels of integrin α4ß7, as well as CD62L and FOXP3, compared with cells incubated with rapamycin or rapamycin and all-trans retinoic acid. These Treg cells had increased suppressive activities in assays and migrated under conditions of shear flow; they did not produce inflammatory cytokines, and RAR568 had no effect on cell stability or lineage commitment. Fluorescently labeled Treg cells incubated with RAR568 were significantly more likely to traffic to intestinal xenografts than Treg cells expanded in control medium. CONCLUSIONS: Treg cells from patients with CD express lower levels of the integrin α4ß7 than Treg cells from control patients. Incubation of patients' ex vivo expanded Treg cells with rapamycin and an RARA agonist induced expression of α4ß7 and had suppressive and migratory activities in culture and in intestinal xenografts in mice. These cells might be developed for treatment of CD. ClinicalTrials.gov, Number: NCT03185000.


Subject(s)
Crohn Disease/immunology , Integrins/metabolism , Retinoic Acid Receptor alpha/agonists , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Antineoplastic Agents/pharmacology , Case-Control Studies , Cell Culture Techniques , Cell Movement/drug effects , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Gene Expression/drug effects , Heterografts , Humans , Immunosuppressive Agents/pharmacology , Integrins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/transplantation , L-Selectin/metabolism , Lymphocyte Activation , Male , Mice , Mice, SCID , Middle Aged , Organic Chemicals/pharmacology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , Transcriptome/drug effects , Tretinoin/pharmacology
18.
PLoS Genet ; 13(2): e1006587, 2017 02.
Article in English | MEDLINE | ID: mdl-28187197

ABSTRACT

The polarization of CD4+ T cells into distinct T helper cell lineages is essential for protective immunity against infection, but aberrant T cell polarization can cause autoimmunity. The transcription factor T-bet (TBX21) specifies the Th1 lineage and represses alternative T cell fates. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) that may be causative for autoimmune diseases. The majority of these polymorphisms are located within non-coding distal regulatory elements. It is considered that these genetic variants contribute to disease by altering the binding of regulatory proteins and thus gene expression, but whether these variants alter the binding of lineage-specifying transcription factors has not been determined. Here, we show that SNPs associated with the mucosal inflammatory diseases Crohn's disease, ulcerative colitis (UC) and celiac disease, but not rheumatoid arthritis or psoriasis, are enriched at T-bet binding sites. Furthermore, we identify disease-associated variants that alter T-bet binding in vitro and in vivo. ChIP-seq for T-bet in individuals heterozygous for the celiac disease-associated SNPs rs1465321 and rs2058622 and the IBD-associated SNPs rs1551398 and rs1551399, reveals decreased binding to the minor disease-associated alleles. Furthermore, we show that rs1465321 is an expression quantitative trait locus (eQTL) for the neighboring gene IL18RAP, with decreased T-bet binding associated with decreased expression of this gene. These results suggest that genetic polymorphisms may predispose individuals to mucosal autoimmune disease through alterations in T-bet binding. Other disease-associated variants may similarly act by modulating the binding of lineage-specifying transcription factors in a tissue-selective and disease-specific manner.


Subject(s)
Celiac Disease/genetics , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , T-Box Domain Proteins/genetics , Animals , Binding Sites/genetics , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/metabolism , Cells, Cultured , Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Gene Expression , Genome-Wide Association Study/methods , Humans , Interleukin-18 Receptor beta Subunit/genetics , Interleukin-18 Receptor beta Subunit/metabolism , Mice, Knockout , Protein Binding/genetics , Regulatory Sequences, Nucleic Acid/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism
19.
Am J Transplant ; 19(8): 2262-2273, 2019 08.
Article in English | MEDLINE | ID: mdl-30920136

ABSTRACT

Genetic variation across the human leukocyte antigen loci is known to influence renal-transplant outcome. However, the impact of genetic variation beyond the human leukocyte antigen loci is less clear. We tested the association of common genetic variation and clinical characteristics, from both the donor and recipient, with posttransplant eGFR at different time-points, out to 5 years posttransplantation. We conducted GWAS meta-analyses across 10 844 donors and recipients from five European ancestry cohorts. We also analyzed the impact of polygenic risk scores (PRS), calculated using genetic variants associated with nontransplant eGFR, on posttransplant eGFR. PRS calculated using the recipient genotype alone, as well as combined donor and recipient genotypes were significantly associated with eGFR at 1-year posttransplant. Thirty-two percent of the variability in eGFR at 1-year posttransplant was explained by our model containing clinical covariates (including weights for death/graft-failure), principal components and combined donor-recipient PRS, with 0.3% contributed by the PRS. No individual genetic variant was significantly associated with eGFR posttransplant in the GWAS. This is the first study to examine PRS, composed of variants that impact kidney function in the general population, in a posttransplant context. Despite PRS being a significant predictor of eGFR posttransplant, the effect size of common genetic factors is limited compared to clinical variables.


Subject(s)
Genetic Markers , Genetic Variation , Graft Rejection/diagnosis , Kidney Transplantation/adverse effects , Kidney/physiopathology , Postoperative Complications/diagnosis , Risk Assessment/methods , Adult , Europe/epidemiology , Female , Follow-Up Studies , Genome-Wide Association Study , Glomerular Filtration Rate , Graft Rejection/epidemiology , Graft Rejection/genetics , Graft Survival , Humans , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/surgery , Kidney Function Tests , Living Donors/statistics & numerical data , Male , Middle Aged , Postoperative Complications/epidemiology , Postoperative Complications/genetics , Prognosis , Retrospective Studies , Risk Factors , Transplant Recipients/statistics & numerical data
20.
Gastroenterology ; 164(6): 1031-1032, 2023 May.
Article in English | MEDLINE | ID: mdl-31593699
SELECTION OF CITATIONS
SEARCH DETAIL