Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
Add more filters

Publication year range
1.
Cell ; 180(1): 64-78.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923400

ABSTRACT

Enteric-associated neurons (EANs) are closely associated with immune cells and continuously monitor and modulate homeostatic intestinal functions, including motility and nutrient sensing. Bidirectional interactions between neuronal and immune cells are altered during disease processes such as neurodegeneration or irritable bowel syndrome. We investigated the effects of infection-induced inflammation on intrinsic EANs (iEANs) and the role of intestinal muscularis macrophages (MMs) in this context. Using murine models of enteric infections, we observed long-term gastrointestinal symptoms, including reduced motility and loss of excitatory iEANs, which was mediated by a Nlrp6- and Casp11-dependent mechanism, depended on infection history, and could be reversed by manipulation of the microbiota. MMs responded to luminal infection by upregulating a neuroprotective program via ß2-adrenergic receptor (ß2-AR) signaling and mediated neuronal protection through an arginase 1-polyamine axis. Our results identify a mechanism of neuronal death post-infection and point to a role for tissue-resident MMs in limiting neuronal damage.


Subject(s)
Intestinal Mucosa/immunology , Macrophages/immunology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic Agents , Animals , Arginase/metabolism , Caspases, Initiator/immunology , Caspases, Initiator/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Female , Gastrointestinal Diseases , Gastrointestinal Microbiome , Infections , Inflammation/immunology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestines/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microbiota , Neurons/physiology , Receptors, Adrenergic, beta-2/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signal Transduction
2.
Cell ; 180(2): 278-295.e23, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978345

ABSTRACT

Mutations in FAMIN cause arthritis and inflammatory bowel disease in early childhood, and a common genetic variant increases the risk for Crohn's disease and leprosy. We developed an unbiased liquid chromatography-mass spectrometry screen for enzymatic activity of this orphan protein. We report that FAMIN phosphorolytically cleaves adenosine into adenine and ribose-1-phosphate. Such activity was considered absent from eukaryotic metabolism. FAMIN and its prokaryotic orthologs additionally have adenosine deaminase, purine nucleoside phosphorylase, and S-methyl-5'-thioadenosine phosphorylase activity, hence, combine activities of the namesake enzymes of central purine metabolism. FAMIN enables in macrophages a purine nucleotide cycle (PNC) between adenosine and inosine monophosphate and adenylosuccinate, which consumes aspartate and releases fumarate in a manner involving fatty acid oxidation and ATP-citrate lyase activity. This macrophage PNC synchronizes mitochondrial activity with glycolysis by balancing electron transfer to mitochondria, thereby supporting glycolytic activity and promoting oxidative phosphorylation and mitochondrial H+ and phosphate recycling.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Adenine/metabolism , Adenosine/metabolism , Adenosine Deaminase/metabolism , Chromatography, Liquid/methods , HEK293 Cells , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mass Spectrometry/methods , Multifunctional Enzymes/genetics , Phosphorylation , Proteins/genetics , Purine Nucleotides/metabolism , Purines/metabolism
3.
Immunity ; 56(5): 1115-1131.e9, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36917985

ABSTRACT

Intestinal IL-17-producing T helper (Th17) cells are dependent on adherent microbes in the gut for their development. However, how microbial adherence to intestinal epithelial cells (IECs) promotes Th17 cell differentiation remains enigmatic. Here, we found that Th17 cell-inducing gut bacteria generated an unfolded protein response (UPR) in IECs. Furthermore, subtilase cytotoxin expression or genetic removal of X-box binding protein 1 (Xbp1) in IECs caused a UPR and increased Th17 cells, even in antibiotic-treated or germ-free conditions. Mechanistically, UPR activation in IECs enhanced their production of both reactive oxygen species (ROS) and purine metabolites. Treating mice with N-acetyl-cysteine or allopurinol to reduce ROS production and xanthine, respectively, decreased Th17 cells that were associated with an elevated UPR. Th17-related genes also correlated with ER stress and the UPR in humans with inflammatory bowel disease. Overall, we identify a mechanism of intestinal Th17 cell differentiation that emerges from an IEC-associated UPR.


Subject(s)
Endoplasmic Reticulum Stress , Intestinal Mucosa , Th17 Cells , Endoplasmic Reticulum Stress/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Cell Differentiation , Humans , Animals , Mice , Mice, Transgenic , Anti-Bacterial Agents/pharmacology
5.
Immunity ; 53(6): 1258-1271.e5, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33296686

ABSTRACT

CD4+ T cells reactive against SARS-CoV-2 can be found in unexposed individuals, and these are suggested to arise in response to common cold coronavirus (CCCoV) infection. Here, we utilized SARS-CoV-2-reactive CD4+ T cell enrichment to examine the antigen avidity and clonality of these cells, as well as the relative contribution of CCCoV cross-reactivity. SARS-CoV-2-reactive CD4+ memory T cells were present in virtually all unexposed individuals examined, displaying low functional avidity and multiple, highly variable cross-reactivities that were not restricted to CCCoVs. SARS-CoV-2-reactive CD4+ T cells from COVID-19 patients lacked cross-reactivity to CCCoVs, irrespective of strong memory T cell responses against CCCoV in all donors analyzed. In severe but not mild COVID-19, SARS-CoV-2-specific T cells displayed low functional avidity and clonality, despite increased frequencies. Our findings identify low-avidity CD4+ T cell responses as a hallmark of severe COVID-19 and argue against a protective role for CCCoV-reactive T cells in SARS-CoV-2 infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , COVID-19/immunology , Receptors, Antigen, T-Cell/metabolism , Rhinovirus/immunology , SARS-CoV-2/immunology , Antigens, Viral/immunology , Cells, Cultured , Cross Reactions , Disease Progression , Environmental Exposure , Humans , Immunologic Memory , Lymphocyte Activation , Protein Binding , Severity of Illness Index , T-Cell Antigen Receptor Specificity
6.
Immunity ; 50(5): 1289-1304.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31079916

ABSTRACT

Pathogenic lymphocytes initiate the development of chronic inflammatory diseases. The cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) (encoded by Csf2) is a key communicator between pathogenic lymphocytes and tissue-invading inflammatory phagocytes. However, the molecular properties of GM-CSF-producing cells and the mode of Csf2 regulation in vivo remain unclear. To systematically study and manipulate GM-CSF+ cells and their progeny in vivo, we generated a fate-map and reporter of GM-CSF expression mouse strain (FROG). We mapped the phenotypic and functional profile of auto-aggressive T helper (Th) cells during neuroinflammation and identified the signature and pathogenic memory of a discrete encephalitogenic Th subset. These cells required interleukin-23 receptor (IL-23R) and IL-1R but not IL-6R signaling for their maintenance and pathogenicity. Specific ablation of this subset interrupted the inflammatory cascade, despite the unperturbed tissue accumulation of other Th subsets (e.g., Th1 and Th17), highlighting that GM-CSF expression not only marks pathogenic Th cells, but that this subset mediates immunopathology and tissue destruction.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Inflammation/genetics , Inflammation/pathology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR6/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
Nat Immunol ; 16(8): 880-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147688

ABSTRACT

Secretory immunoglobulin A (SIgA) shields the gut epithelium from luminal antigens and contributes to host-microbe symbiosis. However, how antibody responses are regulated to achieve sustained host-microbe interactions is unknown. We found that mice and humans exhibited longitudinal persistence of clonally related B cells in the IgA repertoire despite major changes in the microbiota during antibiotic treatment or infection. Memory B cells recirculated between inductive compartments and were clonally related to plasma cells in gut and mammary glands. Our findings suggest that continuous diversification of memory B cells constitutes a central process for establishing symbiotic host-microbe interactions and offer an explanation of how maternal antibodies are optimized throughout life to protect the newborn.


Subject(s)
Adaptation, Physiological/immunology , Antibodies/immunology , B-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Immunoglobulin A, Secretory/immunology , Microbiota/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies/genetics , Antibodies/metabolism , B-Lymphocytes/metabolism , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunologic Memory/immunology , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microbiota/genetics , Microbiota/physiology , Mutation , Plasma Cells/immunology , Plasma Cells/metabolism , RNA, Ribosomal, 16S/genetics , Symbiosis/drug effects , Symbiosis/immunology , Young Adult
8.
Immunity ; 48(4): 608-613, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669240

ABSTRACT

Chronic inflammatory diseases represent an increasing medical burden, yet neither tools to predict the individual disease course nor causal cures are at hand. We discuss opportunities for systems medicine to derive precise, individualized disease models and outline the European consortium SYSCID as part of the roadmap to clinical practice.


Subject(s)
Inflammation , Systems Analysis , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Europe , Humans , Inflammation/diagnosis , Inflammation/genetics , Inflammation/pathology , Inflammation/therapy , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Psoriasis/diagnosis , Psoriasis/genetics , Psoriasis/pathology
9.
Immunity ; 46(2): 173-175, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228276

ABSTRACT

Mechanisms that contribute to healthy aging remain obscure. In a recent issue of Nature Medicine, Furman et al. (2017) describe that dietary caffeine inhibits the NLRC4 inflammasome, which is associated with disease-free aging.


Subject(s)
Inflammasomes , Neuronal Apoptosis-Inhibitory Protein , Apoptosis Regulatory Proteins , Calcium-Binding Proteins , Humans
10.
Nature ; 587(7834): 377-386, 2020 11.
Article in English | MEDLINE | ID: mdl-32894860

ABSTRACT

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Subject(s)
Cell- and Tissue-Based Therapy , Delivery of Health Care/methods , Delivery of Health Care/trends , Medicine/methods , Medicine/trends , Pathology , Single-Cell Analysis , Artificial Intelligence , Delivery of Health Care/ethics , Delivery of Health Care/standards , Early Diagnosis , Education, Medical , Europe , Female , Health , Humans , Legislation, Medical , Male , Medicine/standards
11.
Proc Natl Acad Sci U S A ; 120(40): e2221507120, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37751555

ABSTRACT

Antibiotics, by definition, reduce bacterial growth rates in optimal culture conditions; however, the real-world environments bacteria inhabit see rapid growth punctuated by periods of low nutrient availability. How antibiotics mediate population decline during these periods is poorly understood. Bacteria cannot optimize for all environmental conditions because a growth-longevity tradeoff predicts faster growth results in faster population decline, and since bacteriostatic antibiotics slow growth, they should also mediate longevity. We quantify how antibiotics, their targets, and resistance mechanisms influence longevity using populations of Escherichia coli and, as the tradeoff predicts, populations are maintained for longer if they encounter ribosome-binding antibiotics doxycycline and erythromycin, a finding that is not observed using antibiotics with alternative cellular targets. This tradeoff also predicts resistance mechanisms that increase growth rates during antibiotic treatment could be detrimental during nutrient stresses, and indeed, we find resistance by ribosomal protection removes benefits to longevity provided by doxycycline. We therefore liken ribosomal protection to a "Trojan horse" because it provides protection from an antibiotic but, during nutrient stresses, it promotes the demise of the bacteria. Seeking mechanisms to support these observations, we show doxycycline promotes efficient metabolism and reduces the concentration of reactive oxygen species. Seeking generality, we sought another mechanism that affects longevity and we found the number of doxycycline targets, namely, the ribosomal RNA operons, mediates growth and longevity even without antibiotics. We conclude that slow growth, as observed during antibiotic treatment, can help bacteria overcome later periods of nutrient stress.


Subject(s)
Anti-Bacterial Agents , Bacteria , Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Escherichia coli , Ribosomes , Humans
12.
Immunity ; 45(4): 931-943, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27717798

ABSTRACT

The efficacy of the anti-cancer immunomodulatory agent cyclophosphamide (CTX) relies on intestinal bacteria. How and which relevant bacterial species are involved in tumor immunosurveillance, and their mechanism of action are unclear. Here, we identified two bacterial species, Enterococcus hirae and Barnesiella intestinihominis that are involved during CTX therapy. Whereas E. hirae translocated from the small intestine to secondary lymphoid organs and increased the intratumoral CD8/Treg ratio, B. intestinihominis accumulated in the colon and promoted the infiltration of IFN-γ-producing γδT cells in cancer lesions. The immune sensor, NOD2, limited CTX-induced cancer immunosurveillance and the bioactivity of these microbes. Finally, E. hirae and B. intestinihominis specific-memory Th1 cell immune responses selectively predicted longer progression-free survival in advanced lung and ovarian cancer patients treated with chemo-immunotherapy. Altogether, E. hirae and B. intestinihominis represent valuable "oncomicrobiotics" ameliorating the efficacy of the most common alkylating immunomodulatory compound.


Subject(s)
Cyclophosphamide/pharmacology , Enterococcus hirae/immunology , Immunologic Factors/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Colon/immunology , Colon/microbiology , Immunologic Memory/immunology , Immunotherapy/methods , Interferon-gamma/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Monitoring, Immunologic , Nod2 Signaling Adaptor Protein/immunology , Th1 Cells/immunology
13.
Nucleic Acids Res ; 51(14): 7143-7162, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37351572

ABSTRACT

In the late 19th century, formalin fixation with paraffin-embedding (FFPE) of tissues was developed as a fixation and conservation method and is still used to this day in routine clinical and pathological practice. The implementation of state-of-the-art nucleic acid sequencing technologies has sparked much interest for using historical FFPE samples stored in biobanks as they hold promise in extracting new information from these valuable samples. However, formalin fixation chemically modifies DNA, which potentially leads to incorrect sequences or misinterpretations in downstream processing and data analysis. Many publications have concentrated on one type of DNA damage, but few have addressed the complete spectrum of FFPE-DNA damage. Here, we review mitigation strategies in (I) pre-analytical sample quality control, (II) DNA repair treatments, (III) analytical sample preparation and (IV) bioinformatic analysis of FFPE-DNA. We then provide recommendations that are tested and illustrated with DNA from 13-year-old liver specimens, one FFPE preserved and one fresh frozen, applying target-enriched sequencing. Thus, we show how DNA damage can be compensated, even when using low quantities (50 ng) of fragmented FFPE-DNA (DNA integrity number 2.0) that cannot be amplified well (Q129 bp/Q41 bp = 5%). Finally, we provide a checklist called 'ERROR-FFPE-DNA' that summarises recommendations for the minimal information in publications required for assessing fitness-for-purpose and inter-study comparison when using FFPE samples.


Subject(s)
Sequence Analysis, DNA , DNA/genetics , DNA/analysis , Formaldehyde , Paraffin Embedding/methods , Sequence Analysis, DNA/methods , Tissue Fixation/methods
14.
Nature ; 563(7731): E25, 2018 11.
Article in English | MEDLINE | ID: mdl-30158704

ABSTRACT

In Fig. 1d of this Letter, the third group along should have been labelled 'WT' rather than 'Tlr5'. This has been corrected online.

15.
Nature ; 560(7719): 489-493, 2018 08.
Article in English | MEDLINE | ID: mdl-30089902

ABSTRACT

Alterations in enteric microbiota are associated with several highly prevalent immune-mediated and metabolic diseases1-3, and experiments involving faecal transplants have indicated that such alterations have a causal role in at least some such conditions4-6. The postnatal period is particularly critical for the development of microbiota composition, host-microbe interactions and immune homeostasis7-9. However, the underlying molecular mechanisms of this neonatal priming period have not been defined. Here we report the identification of a host-mediated regulatory circuit of bacterial colonization that acts solely during the early neonatal period but influences life-long microbiota composition. We demonstrate age-dependent expression of the flagellin receptor Toll-like receptor 5 (TLR5) in the gut epithelium of neonate mice. Using competitive colonization experiments, we demonstrate that epithelial TLR5-mediated REG3γ production is critical for the counter-selection of colonizing flagellated bacteria. Comparative microbiota transfer experiments in neonate and adult wild-type and Tlr5-deficient germ-free mice reveal that neonatal TLR5 expression strongly influences the composition of the microbiota throughout life. Thus, the beneficial microbiota in the adult host is shaped during early infancy. This might explain why environmental factors that disturb the establishment of the microbiota during early life can affect immune homeostasis and health in adulthood.


Subject(s)
Aging/immunology , Animals, Newborn/immunology , Gastrointestinal Microbiome/immunology , Toll-Like Receptor 5/immunology , Aging/genetics , Animals , Animals, Newborn/genetics , Crosses, Genetic , Environment , Female , Flagellin/immunology , Flagellin/metabolism , Gastrointestinal Microbiome/genetics , Homeostasis , Host Microbial Interactions , Housing, Animal , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Toll-Like Receptor 5/genetics
16.
EMBO J ; 38(20): e101266, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31544965

ABSTRACT

Inflammasomes are cytosolic protein complexes, which orchestrate the maturation of active IL-1ß by proteolytic cleavage via caspase-1. Although many principles of inflammasome activation have been described, mechanisms that limit inflammasome-dependent immune responses remain poorly defined. Here, we show that the thiol-specific peroxidase peroxiredoxin-4 (Prdx4) directly regulates IL-1ß generation by interfering with caspase-1 activity. We demonstrate that caspase-1 and Prdx4 form a redox-sensitive regulatory complex via caspase-1 cysteine 397 that leads to caspase-1 sequestration and inactivation. Mice lacking Prdx4 show an increased susceptibility to LPS-induced septic shock. This effect was phenocopied in mice carrying a conditional deletion of Prdx4 in the myeloid lineage (Prdx4-ΔLysMCre). Strikingly, we demonstrate that Prdx4 co-localizes with inflammasome components in extracellular vesicles (EVs) from inflammasome-activated macrophages. Purified EVs are able to transmit a robust IL-1ß-dependent inflammatory response in vitro and also in recipient mice in vivo. Loss of Prdx4 boosts the pro-inflammatory potential of EVs. These findings identify Prdx4 as a critical regulator of inflammasome activity and provide new insights into remote cell-to-cell communication function of inflammasomes via macrophage-derived EVs.


Subject(s)
Caspase 1/metabolism , Extracellular Vesicles/metabolism , Inflammasomes/immunology , Macrophages/immunology , Peroxiredoxins/physiology , Shock, Septic/prevention & control , Animals , Caspase 1/genetics , Cytokines/metabolism , Female , Inflammasomes/metabolism , Lipopolysaccharides/toxicity , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Shock, Septic/chemically induced , Shock, Septic/immunology , Shock, Septic/pathology , Signal Transduction
17.
Gastroenterology ; 162(1): 223-237.e11, 2022 01.
Article in English | MEDLINE | ID: mdl-34599932

ABSTRACT

BACKGROUND & AIMS: Throughout life, the intestinal epithelium undergoes constant self-renewal from intestinal stem cells. Together with genotoxic stressors and failing DNA repair, this self-renewal causes susceptibility toward malignant transformation. X-box binding protein 1 (XBP1) is a stress sensor involved in the unfolded protein response (UPR). We hypothesized that XBP1 acts as a signaling hub to regulate epithelial DNA damage responses. METHODS: Data from The Cancer Genome Atlas were analyzed for association of XBP1 with colorectal cancer (CRC) survival and molecular interactions between XBP1 and p53 pathway activity. The role of XBP1 in orchestrating p53-driven DNA damage response was tested in vitro in mouse models of chronic intestinal epithelial cell (IEC) DNA damage (Xbp1/H2bfl/fl, Xbp1ΔIEC, H2bΔIEC, H2b/Xbp1ΔIEC) and via orthotopic tumor organoid transplantation. Transcriptome analysis of intestinal organoids was performed to identify molecular targets of Xbp1-mediated DNA damage response. RESULTS: In The Cancer Genome Atlas data set of CRC, low XBP1 expression was significantly associated with poor overall survival and reduced p53 pathway activity. In vivo, H2b/Xbp1ΔIEC mice developed spontaneous intestinal carcinomas. Orthotopic tumor organoid transplantation revealed a metastatic potential of H2b/Xbp1ΔIEC-derived tumors. RNA sequencing of intestinal organoids (H2b/Xbp1fl/fl, H2bΔIEC, H2b/Xbp1ΔIEC, and H2b/p53ΔIEC) identified a transcriptional program downstream of p53, in which XBP1 directs DNA-damage-inducible transcript 4-like (Ddit4l) expression. DDIT4L inhibits mechanistic target of rapamycin-mediated phosphorylation of 4E-binding protein 1. Pharmacologic mechanistic target of rapamycin inhibition suppressed epithelial hyperproliferation via 4E-binding protein 1. CONCLUSIONS: Our data suggest a crucial role for XBP1 in coordinating epithelial DNA damage responses and stem cell function via a p53-DDIT4L-dependent feedback mechanism.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Cell Transformation, Neoplastic/metabolism , DNA Damage , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , X-Box Binding Protein 1/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/drug therapy , Adenoma/genetics , Adenoma/pathology , Animals , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Databases, Genetic , Endoplasmic Reticulum Stress , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , MTOR Inhibitors/pharmacology , Mice, Knockout , Signal Transduction , Sirolimus/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , X-Box Binding Protein 1/genetics
18.
Gastroenterology ; 162(6): 1690-1704, 2022 05.
Article in English | MEDLINE | ID: mdl-35031299

ABSTRACT

BACKGROUND & AIMS: Crohn's disease (CD) globally emerges with Westernization of lifestyle and nutritional habits. However, a specific dietary constituent that comprehensively evokes gut inflammation in human inflammatory bowel diseases remains elusive. We aimed to delineate how increased intake of polyunsaturated fatty acids (PUFAs) in a Western diet, known to impart risk for developing CD, affects gut inflammation and disease course. We hypothesized that the unfolded protein response and antioxidative activity of glutathione peroxidase 4 (GPX4), which are compromised in human CD epithelium, compensates for metabolic perturbation evoked by dietary PUFAs. METHODS: We phenotyped and mechanistically dissected enteritis evoked by a PUFA-enriched Western diet in 2 mouse models exhibiting endoplasmic reticulum (ER) stress consequent to intestinal epithelial cell (IEC)-specific deletion of X-box binding protein 1 (Xbp1) or Gpx4. We translated the findings to human CD epithelial organoids and correlated PUFA intake, as estimated by a dietary questionnaire or stool metabolomics, with clinical disease course in 2 independent CD cohorts. RESULTS: PUFA excess in a Western diet potently induced ER stress, driving enteritis in Xbp1-/-IEC and Gpx4+/-IEC mice. ω-3 and ω-6 PUFAs activated the epithelial endoplasmic reticulum sensor inositol-requiring enzyme 1α (IRE1α) by toll-like receptor 2 (TLR2) sensing of oxidation-specific epitopes. TLR2-controlled IRE1α activity governed PUFA-induced chemokine production and enteritis. In active human CD, ω-3 and ω-6 PUFAs instigated epithelial chemokine expression, and patients displayed a compatible inflammatory stress signature in the serum. Estimated PUFA intake correlated with clinical and biochemical disease activity in a cohort of 160 CD patients, which was similarly demonstrable in an independent metabolomic stool analysis from 199 CD patients. CONCLUSIONS: We provide evidence for the concept of PUFA-induced metabolic gut inflammation which may worsen the course of human CD. Our findings provide a basis for targeted nutritional therapy.


Subject(s)
Crohn Disease , Enteritis , Fatty Acids, Omega-3 , Animals , Crohn Disease/drug therapy , Endoribonucleases , Enteritis/chemically induced , Enteritis/drug therapy , Fatty Acids, Unsaturated , Humans , Inflammation/drug therapy , Mice , Protein Serine-Threonine Kinases , Toll-Like Receptor 2
19.
PLoS Genet ; 16(5): e1008789, 2020 05.
Article in English | MEDLINE | ID: mdl-32453733

ABSTRACT

Over-consumption of high-fat diets (HFDs) is associated with several pathologies. Although the intestine is the organ that comes into direct contact with all diet components, the impact of HFD has mostly been studied in organs that are linked to obesity and obesity related disorders. We used Drosophila as a simple model to disentangle the effects of a HFD on the intestinal structure and physiology from the plethora of other effects caused by this nutritional intervention. Here, we show that a HFD, composed of triglycerides with saturated fatty acids, triggers activation of intestinal stem cells in the Drosophila midgut. This stem cell activation was transient and dependent on the presence of an intestinal microbiota, as it was completely absent in germ free animals. Moreover, major components of the signal transduction pathway have been elucidated. Here, JNK (basket) in enterocytes was necessary to trigger synthesis of the cytokine upd3 in these cells. This ligand in turn activated the JAK/STAT pathway in intestinal stem cells. Chronic subjection to a HFD markedly altered both the microbiota composition and the bacterial load. Although HFD-induced stem cell activity was transient, long-lasting changes to the cellular composition, including a substantial increase in the number of enteroendocrine cells, were observed. Taken together, a HFD enhances stem cell activity in the Drosophila gut and this effect is completely reliant on the indigenous microbiota and also dependent on JNK signaling within intestinal enterocytes.


Subject(s)
Bacteria/classification , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/cytology , Animals , Bacteria/drug effects , Cell Proliferation/drug effects , Drosophila , Drosophila Proteins/metabolism , Intestinal Mucosa/drug effects , MAP Kinase Kinase 4/metabolism , Models, Animal , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects
20.
Gut ; 71(11): 2194-2204, 2022 11.
Article in English | MEDLINE | ID: mdl-35264446

ABSTRACT

OBJECTIVE: One of the current hypotheses to explain the proinflammatory immune response in IBD is a dysregulated T cell reaction to yet unknown intestinal antigens. As such, it may be possible to identify disease-associated T cell clonotypes by analysing the peripheral and intestinal T-cell receptor (TCR) repertoire of patients with IBD and controls. DESIGN: We performed bulk TCR repertoire profiling of both the TCR alpha and beta chains using high-throughput sequencing in peripheral blood samples of a total of 244 patients with IBD and healthy controls as well as from matched blood and intestinal tissue of 59 patients with IBD and disease controls. We further characterised specific T cell clonotypes via single-cell RNAseq. RESULTS: We identified a group of clonotypes, characterised by semi-invariant TCR alpha chains, to be significantly enriched in the blood of patients with Crohn's disease (CD) and particularly expanded in the CD8+ T cell population. Single-cell RNAseq data showed an innate-like phenotype of these cells, with a comparable gene expression to unconventional T cells such as mucosal associated invariant T and natural killer T (NKT) cells, but with distinct TCRs. CONCLUSIONS: We identified and characterised a subpopulation of unconventional Crohn-associated invariant T (CAIT) cells. Multiple evidence suggests these cells to be part of the NKT type II population. The potential implications of this population for CD or a subset thereof remain to be elucidated, and the immunophenotype and antigen reactivity of CAIT cells need further investigations in future studies.


Subject(s)
Crohn Disease , Natural Killer T-Cells , CD8-Positive T-Lymphocytes , Crohn Disease/genetics , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL