Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 710
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(8): 970-979, 2019 08.
Article in English | MEDLINE | ID: mdl-31235952

ABSTRACT

Inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis are characterized by uncontrolled activation of intestinal immune cells in a genetically susceptible host. Due to the progressive and destructive nature of the inflammatory process in IBD, complications such as fibrosis, stenosis or cancer are frequently observed, which highlights the need for effective anti-inflammatory therapy. Studies have identified altered trafficking of immune cells and pathogenic immune cell circuits as crucial drivers of mucosal inflammation and tissue destruction in IBD. A defective gut barrier and microbial dysbiosis induce such accumulation and local activation of immune cells, which results in a pro-inflammatory cytokine loop that overrides anti-inflammatory signals and causes chronic intestinal inflammation. This Review discusses pathogenic cytokine responses of immune cells as well as immune cell trafficking as a rational basis for new translational therapies in IBD.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Cytokines/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Animals , Cell Movement/immunology , Colitis, Ulcerative/therapy , Crohn Disease/therapy , Dysbiosis , Humans , Inflammation/pathology , Intestinal Mucosa/cytology , Lymphocyte Activation/immunology , Mice , T-Lymphocytes/immunology
2.
Nat Immunol ; 20(4): 514, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30846879

ABSTRACT

In the version of this article initially published, a portion of the Acknowledgements section ("the Clinical Research Group CEDER of the German Research Council (DFG)") was incorrect. The correct statement is as follows: "...the Collaborative Research Center TRR241 of the German Research Council (DFG)...". The error has been corrected in the HTML and PDF version of the article.

3.
Nat Immunol ; 20(3): 288-300, 2019 03.
Article in English | MEDLINE | ID: mdl-30692620

ABSTRACT

Although tissue-resident memory T cells (TRM cells) have been shown to regulate host protection in infectious disorders, their function in inflammatory bowel disease (IBD) remains to be investigated. Here we characterized TRM cells in human IBD and in experimental models of intestinal inflammation. Pro-inflammatory TRM cells accumulated in the mucosa of patients with IBD, and the presence of CD4+CD69+CD103+ TRM cells was predictive of the development of flares. In vivo, functional impairment of TRM cells in mice with double knockout of the TRM-cell-associated transcription factors Hobit and Blimp-1 attenuated disease in several models of colitis, due to impaired cross-talk between the adaptive and innate immune system. Finally, depletion of TRM cells led to a suppression of colitis activity. Together, our data demonstrate a central role for TRM cells in the pathogenesis of chronic intestinal inflammation and suggest that these cells could be targets for future therapeutic approaches in IBD.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Immunologic Memory/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Transcription Factors/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chronic Disease , Colitis/genetics , Colitis/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Immunologic Memory/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/deficiency , Positive Regulatory Domain I-Binding Factor 1/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
4.
Immunity ; 55(11): 2059-2073.e8, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351375

ABSTRACT

T memory stem cells (TSCM) display increased self-renewal and prolonged survival capabilities, thus preventing T cell exhaustion and promoting effective anti-tumor T cell responses. TSCM cells can be expanded by Urolithin A (UA), which is produced by the commensal gut microbiome from foods rich in ellagitannins and is known to improve mitochondrial health. Oral UA administration to tumor-bearing mice conferred strong anti-tumor CD8+ T cell immunity, whereas ex vivo UA pre-treated T cells displayed improved anti-tumor function upon adoptive cell transfer. UA-induced TSCM formation depended on Pink1-mediated mitophagy triggering cytosolic release of the mitochondrial phosphatase Pgam5. Cytosolic Pgam5 dephosphorylated ß-catenin, which drove Wnt signaling and compensatory mitochondrial biogenesis. Collectively, we unravel a critical signaling pathway linking mitophagy to TSCM formation and suggest that the well-tolerated metabolic compound UA represents an attractive option to improve immune therapy.


Subject(s)
Coumarins , Mitophagy , Mice , Animals , Coumarins/pharmacology , Wnt Signaling Pathway , Stem Cells , Immunologic Memory
5.
Immunity ; 50(2): 278-280, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784573

ABSTRACT

The role of tumor necrosis factor (TNF) in the fetal intestine is poorly understood. In this issue of Immunity, Schreurs et al. (2019) identify important roles of TNF-producing T cells as both regulators of gut development and potential inducers of colitis.


Subject(s)
Immunologic Memory , Tumor Necrosis Factor-alpha , CD4-Positive T-Lymphocytes , Cytokines , Humans , Inflammation , Intestines
6.
Immunity ; 51(5): 791-793, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31747580

ABSTRACT

The cytokine TNF is thought to play a major role in the immunopathogenesis of ulcerative colitis, and anti-TNF antibodies are considered as cornerstones of clinical therapy. Two clinical trials published in The New England Journal of Medicine now challenge this paradigm and suggest new avenues for research.


Subject(s)
Colitis, Ulcerative , Cytokines , Humans , Tumor Necrosis Factor-alpha , Ustekinumab
7.
Immunity ; 51(2): 367-380.e4, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31350179

ABSTRACT

Epithelial barrier defects are implicated in the pathogenesis of inflammatory bowel disease (IBD); however, the role of microbiome dysbiosis and the cytokine networks orchestrating chronic intestinal inflammation in response to barrier impairment remain poorly understood. Here, we showed that altered Schaedler flora (ASF), a benign minimal microbiota, was sufficient to trigger colitis in a mouse model of intestinal barrier impairment. Colitis development required myeloid-cell-specific adaptor protein MyD88 signaling and was orchestrated by the cytokines IL-12, IL-23, and IFN-γ. Colon inflammation was driven by IL-12 during the early stages of the disease, but as the mice aged, the pathology shifted toward an IL-23-dependent inflammatory response driving disease chronicity. These findings reveal that IL-12 and IL-23 act in a temporally distinct, biphasic manner to induce microbiota-driven chronic intestinal inflammation. Similar mechanisms might contribute to the pathogenesis of IBD particularly in patients with underlying intestinal barrier defects.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-12/metabolism , Interleukin-23/metabolism , Intestinal Mucosa/pathology , Microbiota/immunology , Animals , Chronic Disease , Disease Models, Animal , Humans , Inflammation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-23/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Transplantation Chimera
8.
Immunity ; 51(3): 443-450.e4, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31422870

ABSTRACT

The presence of gallstones (cholelithiasis) is a highly prevalent and severe disease and one of the leading causes of hospital admissions worldwide. Due to its substantial health impact, we investigated the biological mechanisms that lead to the formation and growth of gallstones. We show that gallstone assembly essentially requires neutrophil extracellular traps (NETs). We found consistent evidence for the presence of NETs in human and murine gallstones and describe an immune-mediated process requiring activation of the innate immune system for the formation and growth of gallstones. Targeting NET formation via inhibition of peptidyl arginine deiminase type 4 or abrogation of reactive oxygen species (ROS) production, as well as damping of neutrophils by metoprolol, effectively inhibit gallstone formation in vivo. Our results show that after the physicochemical process of crystal formation, NETs foster their assembly into larger aggregates and finally gallstones. These insights provide a feasible therapeutic concept to prevent cholelithiasis in patients at risk.


Subject(s)
Extracellular Traps/immunology , Gallstones/immunology , Neutrophils/immunology , Animals , Female , Humans , Immunity, Innate/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reactive Oxygen Species/immunology
9.
Trends Immunol ; 45(8): 580-596, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060140

ABSTRACT

The guanine nucleotide exchange factor (GEF) VAV1, a previously 'undruggable' protein integral to T/B lymphocyte antigen-receptor signaling, promotes actin polymerization, immunological synapse formation, T cell activation and differentiation, and cytokine production. With the development of novel modalities for targeting proteins, we hypothesize that interventions targeting VAV1 will have therapeutic potential in T and T/B cell-mediated autoimmune and chronic inflammatory diseases. This opinion is supported by recent CRISPR-Cas9 studies showing VAV1 as a key positive regulator of T cell receptor (TCR) activation and cytokine production in primary human CD4+ and CD8+ T cells; data demonstrating that loss/suppression of VAV1 regulates autoimmunity and inflammation; and promising preclinical data from T and T/B cell-mediated disease models of arthritis and colitis showing the effectiveness of selective VAV1 targeting via protein degradation.


Subject(s)
Autoimmune Diseases , Inflammation , Proto-Oncogene Proteins c-vav , Proto-Oncogene Proteins c-vav/metabolism , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Inflammation/immunology , Chronic Disease , Molecular Targeted Therapy , Signal Transduction , Autoimmunity , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Lymphocyte Activation/immunology
10.
Nat Immunol ; 15(7): 676-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24908389

ABSTRACT

The molecular checkpoints that drive inflammatory bowel diseases are incompletely understood. Here we found more T cells expressing the transcription factor PU.1 and interleukin 9 (IL-9) in patients with ulcerative colitis. In an animal model, citrine reporter mice had more IL-9-expressing mucosal T cells in experimental oxazolone-induced colitis. IL-9 deficiency suppressed acute and chronic colitis. Mice with PU.1 deficiency in T cells were protected from colitis, whereas treatment with antibody to IL-9 suppressed colitis. Functionally, IL-9 impaired intestinal barrier function and prevented mucosal wound healing in vivo. Thus, our findings suggest that the TH9 subset of helper T cells serves an important role in driving ulcerative colitis by regulating intestinal epithelial cells and that TH9 cells represent a likely target for the treatment of chronic intestinal inflammation.


Subject(s)
Colitis/etiology , Intestinal Mucosa/immunology , Proto-Oncogene Proteins/physiology , Receptors, Interleukin-9/physiology , Signal Transduction/physiology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Helper-Inducer/immunology , Trans-Activators/physiology , Animals , Claudin-2/genetics , Colitis/immunology , Colitis, Ulcerative/immunology , Humans , Interleukin-9/immunology , Mice , Mice, Inbred BALB C , Th2 Cells/immunology , Wound Healing
11.
Gastroenterology ; 166(2): 284-297.e11, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37734420

ABSTRACT

BACKGROUND & AIMS: T cells are crucial for the antitumor response against colorectal cancer (CRC). T-cell reactivity to CRC is nevertheless limited by T-cell exhaustion. However, molecular mechanisms regulating T-cell exhaustion are only poorly understood. METHODS: We investigated the functional role of cyclin-dependent kinase 1a (Cdkn1a or p21) in cluster of differentiation (CD) 4+ T cells using murine CRC models. Furthermore, we evaluated the expression of p21 in patients with stage I to IV CRC. In vitro coculture models were used to understand the effector function of p21-deficient CD4+ T cells. RESULTS: We observed that the activation of cell cycle regulator p21 is crucial for CD4+ T-cell cytotoxic function and that p21 deficiency in type 1 helper T cells (Th1) leads to increased tumor growth in murine CRC. Similarly, low p21 expression in CD4+ T cells infiltrated into tumors of CRC patients is associated with reduced cancer-related survival. In mouse models of CRC, p21-deficient Th1 cells show signs of exhaustion, where an accumulation of effector/effector memory T cells and CD27/CD28 loss are predominant. Immune reconstitution of tumor-bearing Rag1-/- mice using ex vivo-treated p21-deficient T cells with palbociclib, an inhibitor of cyclin-dependent kinase 4/6, restored cytotoxic function and prevented exhaustion of p21-deficient CD4+ T cells as a possible concept for future immunotherapy of human disease. CONCLUSIONS: Our data reveal the importance of p21 in controlling the cell cycle and preventing exhaustion of Th1 cells. Furthermore, we unveil the therapeutic potential of cyclin-dependent kinase inhibitors such as palbociclib to reduce T-cell exhaustion for future treatment of patients with colorectal cancer.


Subject(s)
Colorectal Neoplasms , Th1 Cells , Humans , Animals , Mice , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Immunity , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinases/metabolism
12.
Gastroenterology ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38992449

ABSTRACT

BACKGROUND & AIMS: Primary sclerosing cholangitis (PSC), often associated with inflammatory bowel disease (IBD), presents a multifactorial etiology involving genetic, immunologic, and environmental factors. Gut dysbiosis and bacterial translocation have been implicated in PSC-IBD, yet the precise mechanisms underlying their pathogenesis remain elusive. Here, we describe the role of gut pathobionts in promoting liver inflammation and fibrosis due to the release of bacterial outer membrane vesicles (OMVs). METHODS: Preclinical mouse models in addition to ductal organoids were used to acquire mechanistic data. A proof-of-concept study including serum and liver biopsies of a patient cohort of PSC (n = 22), PSC-IBD (n = 45), and control individuals (n = 27) was performed to detect OMVs in the systemic circulation and liver. RESULTS: In both preclinical model systems and in patients with PSC-IBD, the translocation of OMVs to the liver correlated with enhanced bacterial sensing and accumulation of the NLRP3 inflammasome. Using ductal organoids, we were able to precisely attribute the pro-inflammatory and pro-fibrogenic properties of OMVs to signaling pathways dependent on Toll-like receptor 4 and NLRP3-gasdermin-D. The immunostimulatory potential of OMVs could be confirmed in macrophages and hepatic stellate cells. Furthermore, when we administered gut pathobiont-derived OMVs to Mdr2-/- mice, we observed a significant enhancement in liver inflammation and fibrosis. In a translational approach, we substantiated the presence of OMVs in the systemic circulation and hepatic regions of severe fibrosis using a PSC-IBD patient cohort. CONCLUSIONS: This study demonstrates the contribution of gut pathobionts in releasing OMVs that traverse the mucosal barrier and, thus, promote liver inflammation and fibrosis in PSC-IBD. OMVs might represent a critical new environmental factor that interacts with other disease factors to cause inflammation and thus define potential new targets for fibrosis therapy.

13.
Gut ; 73(2): 282-297, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37884352

ABSTRACT

OBJECTIVE: We sought to investigate the role of interleukin (IL)-20 in IBD and experimental colitis. DESIGN: Experimental colitis was induced in mice deficient in components of the IL-20 and signal transducer and activator of transcription (STAT)2 signalling pathways. In vivo imaging, high-resolution mini-endoscopy and histology were used to assess intestinal inflammation. We further used RNA-sequencing (RNA-Seq), RNAScope and Gene Ontology analysis, western blot analysis and co-immunoprecipitation, confocal microscopy and intestinal epithelial cell (IEC)-derived three-dimensional organoids to investigate the underlying molecular mechanisms. Results were validated using samples from patients with IBD and non-IBD control subjects by a combination of RNA-Seq, organoids and immunostainings. RESULTS: In IBD, IL20 levels were induced during remission and were significantly higher in antitumour necrosis factor responders versus non-responders. IL-20RA and IL-20RB were present on IECs from patients with IBD and IL-20-induced STAT3 and suppressed interferon (IFN)-STAT2 signalling in these cells. In IBD, experimental dextran sulfate sodium (DSS)-induced colitis and mucosal healing, IECs were the main producers of IL-20. Compared with wildtype controls, Il20-/-, Il20ra-/- and Il20rb-/- mice were more susceptible to experimental DSS-induced colitis. IL-20 deficiency was associated with increased IFN/STAT2 activity in mice and IFN/STAT2-induced necroptotic cell death in IEC-derived organoids could be markedly blocked by IL-20. Moreover, newly generated Stat2ΔIEC mice, lacking STAT2 in IECs, were less susceptible to experimental colitis compared with wildtype controls and the administration of IL-20 suppressed colitis activity in wildtype animals. CONCLUSION: IL-20 controls colitis and mucosal healing by interfering with the IFN/STAT2 death signalling pathway in IECs. These results indicate new directions for suppressing gut inflammation by modulating IL-20-controlled STAT2 signals.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Animals , Mice , Intestinal Mucosa/metabolism , Colitis/metabolism , Interleukins/metabolism , Inflammation/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/genetics , Dextran Sulfate/pharmacology , Mice, Inbred C57BL , STAT2 Transcription Factor/metabolism
14.
Gut ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684238

ABSTRACT

OBJECTIVE: Mutations in presenilin genes are the major cause of Alzheimer's disease. However, little is known about their expression and function in the gut. In this study, we identify the presenilins Psen1 and Psen2 as key molecules that maintain intestinal homoeostasis. DESIGN: Human inflammatory bowel disease (IBD) and control samples were analysed for Psen1 expression. Newly generated intestinal epithelium-specific Psen1-deficient, Psen2-deficient and inducible Psen1/Psen2 double-deficient mice were used to dissect the functional role of presenilins in intestinal homoeostasis. RESULTS: Psen1 expression was regulated in experimental gut inflammation and in patients with IBD. Induced deletion of Psen1 and Psen2 in mice caused rapid weight loss and spontaneous development of intestinal inflammation. Mice exhibited epithelial barrier disruption with bacterial translocation and deregulation of key pathways for nutrient uptake. Wasting disease was independent of gut inflammation and dysbiosis, as depletion of microbiota rescued Psen-deficient animals from spontaneous colitis development but not from weight loss. On a molecular level, intestinal epithelial cells lacking Psen showed impaired Notch signalling and dysregulated epithelial differentiation. CONCLUSION: Overall, our study provides evidence that Psen1 and Psen2 are important guardians of intestinal homoeostasis and future targets for barrier-promoting therapeutic strategies in IBD.

15.
Gut ; 73(4): 601-612, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38176897

ABSTRACT

OBJECTIVE: Mucosal T cells play a major role in inflammatory bowel disease (IBD). However, their immunometabolism during intestinal inflammation is poorly understood. Due to its impact on cellular metabolism and proinflammatory immune cell function, we here focus on the enzyme ATP citrate lyase (ACLY) in mucosal T cell immunometabolism and its relevance for IBD. DESIGN: ACLY expression and its immunometabolic impact on colitogenic T cell function were analysed in mucosal T cells from patients with IBD and in two experimental colitis models. RESULTS: ACLY was markedly expressed in colon tissue under steady-state conditions but was significantly downregulated in lamina propria mononuclear cells in experimental dextran sodium sulfate-induced colitis and in CD4+ and to a lesser extent in CD8+ T cells infiltrating the inflamed gut in patients with IBD. ACLY-deficient CD4+ T cells showed an impaired capacity to induce intestinal inflammation in a transfer colitis model as compared with wild-type T cells. Assessment of T cell immunometabolism revealed that ACLY deficiency dampened the production of IBD-relevant cytokines and impaired glycolytic ATP production but enriched metabolites involved in the biosynthesis of phospholipids and phosphatidylcholine. Interestingly, the short-chain fatty acid butyrate was identified as a potent suppressor of ACLY expression in T cells, while IL-36α and resolvin E1 induced ACLY levels. In a translational approach, in vivo administration of the butyrate prodrug tributyrin downregulated mucosal infiltration of ACLYhigh CD4+ T cells and ameliorated chronic colitis. CONCLUSION: ACLY controls mucosal T cell immunometabolism and experimental colitis. Therapeutic modulation of ACLY expression in T cells emerges as a novel strategy to promote the resolution of intestinal inflammation.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Intraepithelial Lymphocytes , Humans , Animals , Intraepithelial Lymphocytes/metabolism , ATP Citrate (pro-S)-Lyase/metabolism , CD8-Positive T-Lymphocytes/metabolism , Colitis/metabolism , Inflammation/metabolism , Butyrates , Intestinal Mucosa/metabolism , Dextran Sulfate , Disease Models, Animal
16.
Clin Gastroenterol Hepatol ; 22(7): 1353-1364, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38320679

ABSTRACT

Despite recent advances in the understanding of the pathogenesis of inflammatory bowel diseases (IBD) and advent of multiple targeted therapies, approximately one-third of patients are primary non-responders to initiated treatment, and half of patients lose response over time. There is currently a lack of available biomarkers that would prognosticate therapeutic effectiveness of these advanced therapies. This is partly explained by insufficient characterization of the functional roles assumed by the chosen molecular targets during disease treatment. There is a dire need for validated objective biomarkers, which could be indicators of a biological process, that can be applied in clinical practice to assist us in assigning therapies to patients with the highest probability of response. An appropriate molecular and cellular characterization that accounts for the interindividual differences in drug efficacy and potential side effects would help to guide clinicians in the management of patients with IBD and represent a major step to tailor a more personalized approach to treatment. An appropriate combination of complementing biomarkers should ideally incorporate a multimodal analysis in which genetic, microbial, transcriptional, proteomic, metabolic, and immunologic data are combined to enable a truly personalized approach. This would classify patients into disease subgroups according to molecular characteristics, which would enable us to initiate the most appropriate therapeutic substance. Emergence of single-cell technologies to map the intestinal cellular landscape and multiomic approaches have helped to further dissect the pathogenic mechanisms of mucosal inflammation, but the clinical translation of potential biomarkers remains cumbersome, and an ongoing concerted effort by the IBD community is required.


Subject(s)
Biomarkers , Inflammatory Bowel Diseases , Precision Medicine , Humans , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/drug therapy , Precision Medicine/methods
17.
Gastroenterology ; 164(2): 241-255, 2023 02.
Article in English | MEDLINE | ID: mdl-36279923

ABSTRACT

BACKGROUND & AIMS: Endoscopic and histologic remission have emerged as key therapeutic goals in the management of inflammatory bowel diseases (IBD) that are associated with favorable long-term disease outcomes. Here, we prospectively compared the predictive value of barrier healing with endoscopic and histologic remission for predicting long-term disease behavior in a large cohort of patients with IBD in clinical remission. METHODS: At baseline, patients with IBD in clinical remission underwent ileocolonoscopy with assessment of intestinal barrier function by confocal endomicroscopy. Endoscopic and histologic disease activity, as well as barrier healing, was prospectively assessed along established scores. During subsequent follow-up, patients were closely monitored for clinical disease activity and the occurrence of major adverse outcomes (MAOs): disease flares, IBD-related hospitalization or surgery, and initiation or dose escalation of systemic steroids, immunosuppressants, small molecules, or biological therapy. RESULTS: The final analysis included 181 patients, 100 with Crohn's disease [CD] and 81 with ulcerative colitis (UC). During a mean follow-up of 35 (CD) and 25 (UC) months, 73% of patients with CD and 69% of patients with UC experienced at least 1 MAO. The probability of MAO-free survival was significantly higher in patients with IBD with endoscopic remission compared with endoscopically active disease. In addition, histologic remission predicted MAO-free survival in patients with UC but not CD. Barrier healing on endomicroscopy was superior to endoscopic and histologic remission for predicting MAO-free survival in both UC and CD. CONCLUSIONS: Barrier healing is associated with decreased risk of disease progression in patients with clinically remittent IBD, with superior predictive performance compared with endoscopic and histologic remission. Analysis of barrier function might be considered as a future treatment target in clinical trials. CLINICALTRIALS: gov number, NCT05157750.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Humans , Prospective Studies , Remission Induction , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/drug therapy , Crohn Disease/drug therapy , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Severity of Illness Index
18.
Clin Gastroenterol Hepatol ; 22(5): 994-1004.e10, 2024 May.
Article in English | MEDLINE | ID: mdl-38184096

ABSTRACT

BACKGROUND & AIMS: Autoimmune pancreatitis (AIP) is an immune-mediated disease of the pancreas with distinct pathophysiology and manifestations. Our aims were to characterize type 1 AIP in a large pan-European cohort and study the effectiveness of current treatment regimens. METHODS: We retrospectively analyzed adults diagnosed since 2005 with type 1 or not-otherwise-specified AIP in 42 European university hospitals. Type 1 AIP was uniformly diagnosed using specific diagnostic criteria. Patients with type 2 AIP and those who had undergone pancreatic surgery were excluded. The primary end point was complete remission, defined as the absence of clinical symptoms and resolution of the index radiologic pancreatic abnormalities attributed to AIP. RESULTS: We included 735 individuals with AIP (69% male; median age, 57 years; 85% White). Steroid treatment was started in 634 patients, of whom 9 (1%) were lost to follow-up. The remaining 625 had a 79% (496/625) complete, 18% (111/625) partial, and 97% (607/625) cumulative remission rate, whereas 3% (18/625) did not achieve remission. No treatment was given in 95 patients, who had a 61% complete (58/95), 19% partial (18/95), and 80% cumulative (76/95) spontaneous remission rate. Higher (≥0.4 mg/kg/day) corticosteroid doses were no more effective than lower (<0.4 mg/kg/day) doses (odds ratio, 0.428; 95% confidence interval, 0.054-3.387) and neither was a starting dose duration >2 weeks (odds ratio, 0.908; 95% confidence interval, 0.818-1.009). Elevated IgG4 levels were independently associated with a decreased chance of complete remission (odds ratio, 0.639; 95% confidence interval, 0.427-0.955). Relapse occurred in 30% of patients. Relapses within 6 months of remission induction were independent of the steroid-tapering duration, induction treatment duration, and total cumulative dose. CONCLUSIONS: Patients with type 1 AIP and elevated IgG4 level may need closer monitoring. For remission induction, a starting dose of 0.4 mg/kg/day for 2 weeks followed by a short taper period seems effective. This study provides no evidence to support more aggressive regimens.


Subject(s)
Autoimmune Pancreatitis , Humans , Male , Middle Aged , Female , Retrospective Studies , Autoimmune Pancreatitis/drug therapy , Autoimmune Pancreatitis/diagnosis , Europe , Aged , Treatment Outcome , Adult , Steroids/therapeutic use , Steroids/administration & dosage , Aged, 80 and over
19.
Ultraschall Med ; 45(3): 316-322, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38171381

ABSTRACT

PURPOSE: Liver histology has prognostic relevance and is used in surveillance and therapeutic strategies. This longitudinal study was designed to evaluate the prognostic relevance of ARFI elastography in comparison to liver histology and to the FIB-4 score in a 5-year observation interval. MATERIALS AND METHODS: Based on the hospital database, patients with an elastography examination of the liver between 2010-2012, a liver biopsy, and a follow-up of 5 years were included in the study. The AUROCs of the events liver-related death, HCC, and liver decompensation/variceal bleeding were calculated for ARFI elastography, liver histology, and FIB-4 and compared using the DeLong test. RESULTS: In the final analysis 113 patients were included with 30 (26.5 %) patients having high-grade fibrosis and 19 (16.8 %) having liver cirrhosis in histology. The AUROC for liver-related death in the 5-year interval (9.7 %, n=11) was 0.80 [0.68-0.92] for ARFI elastography, 0.79 [0.66-0.92] for liver histology, and 0.66 [0.53-0.79] for FIB-4 with a p-value of 0.83 comparing ARFI to histology and a p-value of 0.02 comparing ARFI to FIB-4. The AUROC for liver decompensation/variceal bleeding (13.3 %, n=15) was 0.86 [0.76-0.94] for ARFI, which is significantly higher than the AUROC of liver histology with 0.71 [0.56-0.86] (p=0.02) and FIB-4 with 0.67 [0.54-0.80] (p=0.003). There was no significant difference for the event HCC when comparing ARFI to histology (p=0.33) or FIB-4 (p=0.14). CONCLUSION: The prognostic value of ARFI elastography seems to not be inferior to liver histology regarding liver-related survival and might even outperform histology and the FIB-4 score for predicting some liver-related complications.


Subject(s)
Carcinoma, Hepatocellular , Elasticity Imaging Techniques , Liver Cirrhosis , Liver Neoplasms , Liver , Adult , Aged , Female , Humans , Male , Middle Aged , Biopsy , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/mortality , Elasticity Imaging Techniques/methods , Esophageal and Gastric Varices/diagnostic imaging , Esophageal and Gastric Varices/pathology , Esophageal and Gastric Varices/mortality , Follow-Up Studies , Gastrointestinal Hemorrhage/diagnostic imaging , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/pathology , Liver Cirrhosis/mortality , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Liver Neoplasms/mortality , Longitudinal Studies , Prognosis
20.
Gut ; 72(11): 2164-2183, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37640443

ABSTRACT

Mucosal healing on endoscopy has emerged as a key prognostic parameter in the management of patients with IBD (Crohn's disease, ulcerative colitis/UC) and can predict sustained clinical remission and resection-free survival. The structural basis for this type of mucosal healing is a progressive resolution of intestinal inflammation with associated healing of ulcers and improved epithelial barrier function. However, in some cases with mucosal healing on endoscopy, evidence of histological activity in mucosal biopsies has been observed. Subsequently, in UC, a second, deeper type of mucosal healing, denoted histological healing, was defined which requires the absence of active inflammation in mucosal biopsies. Both levels of mucosal healing should be considered as initial events in the resolution of gut inflammation in IBD rather than as indicators of complete transmural healing. In this review, the effects of anti-inflammatory, biological or immunosuppressive agents as well as small molecules on mucosal healing in clinical studies are highlighted. In addition, we focus on the implications of mucosal healing for clinical management of patients with IBD. Moreover, emerging techniques for the analysis of mucosal healing as well as potentially deeper levels of mucosal healing such as transmural healing and functional barrier healing of the mucosa are discussed. Although none of these new levels of healing indicate a definitive cure of the diseases, they make an important contribution to the assessment of patients' prognosis. The ultimate level of healing in IBD would be a resolution of all aspects of intestinal and extraintestinal inflammation (complete healing).

SELECTION OF CITATIONS
SEARCH DETAIL