Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
2.
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
3.
Immunity ; 51(5): 871-884.e6, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31628054

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) sense environmental signals that are critical for gut homeostasis and host defense. However, the metabolite-sensing G-protein-coupled receptors that regulate colonic ILC3s remain poorly understood. We found that colonic ILC3s expressed Ffar2, a microbial metabolite-sensing receptor, and that Ffar2 agonism promoted ILC3 expansion and function. Deficiency of Ffar2 in ILC3s decreased their in situ proliferation and ILC3-derived interleukin-22 (IL-22) production. This led to impaired gut epithelial function characterized by altered mucus-associated proteins and antimicrobial peptides and increased susceptibility to colonic injury and bacterial infection. Ffar2 increased IL-22+ CCR6+ ILC3s and influenced ILC3 abundance in colonic lymphoid tissues. Ffar2 agonism differentially activated AKT or ERK signaling and increased ILC3-derived IL-22 via an AKT and STAT3 axis. Our findings suggest that Ffar2 regulates colonic ILC3 proliferation and function, and they identify an ILC3-receptor signaling pathway modulating gut homeostasis and pathogen defense.


Subject(s)
Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Receptors, Cell Surface/metabolism , Animals , Biomarkers , Cytokines/metabolism , Disease Susceptibility , Gastrointestinal Microbiome/immunology , Gene Expression , Humans , Immunomodulation , Intestinal Mucosa/pathology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt , Receptors, Cell Surface/agonists , STAT3 Transcription Factor/metabolism
4.
Mol Syst Biol ; 20(4): 338-361, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38467837

ABSTRACT

Microbial biochemistry is central to the pathophysiology of inflammatory bowel diseases (IBD). Improved knowledge of microbial metabolites and their immunomodulatory roles is thus necessary for diagnosis and management. Here, we systematically analyzed the chemical, ecological, and epidemiological properties of ~82k metabolic features in 546 Integrative Human Microbiome Project (iHMP/HMP2) metabolomes, using a newly developed methodology for bioactive compound prioritization from microbial communities. This suggested >1000 metabolic features as potentially bioactive in IBD and associated ~43% of prevalent, unannotated features with at least one well-characterized metabolite, thereby providing initial information for further characterization of a significant portion of the fecal metabolome. Prioritized features included known IBD-linked chemical families such as bile acids and short-chain fatty acids, and less-explored bilirubin, polyamine, and vitamin derivatives, and other microbial products. One of these, nicotinamide riboside, reduced colitis scores in DSS-treated mice. The method, MACARRoN, is generalizable with the potential to improve microbial community characterization and provide therapeutic candidates.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Animals , Mice , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Metabolome , Bile Acids and Salts
6.
BMC Genomics ; 25(1): 409, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664626

ABSTRACT

OBJECTIVE: To evaluate the contribution of germline genetics to regulating the briskness and diversity of T cell responses in CRC, we conducted a genome-wide association study to examine the associations between germline genetic variation and quantitative measures of T cell landscapes in 2,876 colorectal tumors from participants in the Molecular Epidemiology of Colorectal Cancer Study (MECC). METHODS: Germline DNA samples were genotyped and imputed using genome-wide arrays. Tumor DNA samples were extracted from paraffin blocks, and T cell receptor clonality and abundance were quantified by immunoSEQ (Adaptive Biotechnologies, Seattle, WA). Tumor infiltrating lymphocytes per high powered field (TILs/hpf) were scored by a gastrointestinal pathologist. Regression models were used to evaluate the associations between each variant and the three T-cell features, adjusting for sex, age, genotyping platform, and global ancestry. Three independent datasets were used for replication. RESULTS: We identified a SNP (rs4918567) near RBM20 associated with clonality at a genome-wide significant threshold of 5 × 10- 8, with a consistent direction of association in both discovery and replication datasets. Expression quantitative trait (eQTL) analyses and in silico functional annotation for these loci provided insights into potential functional roles, including a statistically significant eQTL between the T allele at rs4918567 and higher expression of ADRA2A (P = 0.012) in healthy colon mucosa. CONCLUSIONS: Our study suggests that germline genetic variation is associated with the quantity and diversity of adaptive immune responses in CRC. Further studies are warranted to replicate these findings in additional samples and to investigate functional genomic mechanisms.


Subject(s)
Colorectal Neoplasms , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Tumor Microenvironment , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Male , Female , Middle Aged , Quantitative Trait Loci , Aged , Lymphocytes, Tumor-Infiltrating/immunology , Germ-Line Mutation , RNA-Binding Proteins/genetics , Genotype , Germ Cells/metabolism
7.
Cell ; 134(5): 743-56, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18775308

ABSTRACT

Inflammatory bowel disease (IBD) has been attributed to aberrant mucosal immunity to the intestinal microbiota. The transcription factor XBP1, a key component of the endoplasmic reticulum (ER) stress response, is required for development and maintenance of secretory cells and linked to JNK activation. We hypothesized that a stressful environmental milieu in a rapidly proliferating tissue might instigate a proinflammatory response. We report that Xbp1 deletion in intestinal epithelial cells (IECs) results in spontaneous enteritis and increased susceptibility to induced colitis secondary to both Paneth cell dysfunction and an epithelium that is overly reactive to inducers of IBD such as bacterial products (flagellin) and TNFalpha. An association of XBP1 variants with both forms of human IBD (Crohn's disease and ulcerative colitis) was identified and replicated (rs35873774; p value 1.6 x 10(-5)) with novel, private hypomorphic variants identified as susceptibility factors. Hence, intestinal inflammation can originate solely from XBP1 abnormalities in IECs, thus linking cell-specific ER stress to the induction of organ-specific inflammation.


Subject(s)
DNA-Binding Proteins/immunology , Endoplasmic Reticulum/immunology , Inflammatory Bowel Diseases/genetics , Transcription Factors/immunology , Animals , Apoptosis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Listeria monocytogenes/immunology , Mice , Mice, Transgenic , Paneth Cells/cytology , Paneth Cells/metabolism , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
8.
BMC Cancer ; 22(1): 1281, 2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36476111

ABSTRACT

BACKGROUND: The Tn antigen (CD175) is an O-glycan expressed in various types of human adenocarcinomas, including colorectal cancer (CRC), though prior studies have relied heavily upon poorly characterized in-house generated antibodies and lectins. In this study, we explored Tn expression in CRC using ReBaGs6, a well-characterized recombinant murine antibody with high specificity for clustered Tn antigen. METHODS: Using well-defined monoclonal antibodies, expression patterns of Tn and sialylated Tn (STn) antigens were characterized by immunostaining in CRC, in matched peritumoral [transitional margin (TM)] mucosa, and in normal colonic mucosa distant from the tumor, as well as in adenomas. Vicia villosa agglutinin lectin was used to detect terminal GalNAc expression. Histo-scoring (H scoring) of staining was carried out, and pairwise comparisons of staining levels between tissue types were performed using paired samples Wilcoxon rank sum tests, with statistical significance set at 0.05. RESULTS: While minimal intracellular Tn staining was seen in normal mucosa, significantly higher expression was observed in both TM mucosa (p < 0.001) and adenocarcinoma (p < 0.001). This pattern was reflected to a lesser degree by STn expression in these tissue types. Interestingly, TM mucosa demonstrates a Tn expression level even higher than that of the adenocarcinoma itself (p = 0.019). Colorectal adenomas demonstrated greater Tn and STn expression relative to normal mucosa (p < 0.001 and p = 0.012, respectively). CONCLUSIONS: In summary, CRC is characterized by alterations in Tn/STn antigen expression in neoplastic epithelium as well as peritumoral benign mucosa. Tn/STn antigens are seldom expressed in normal mucosa. This suggests that TM mucosa, in addition to CRC itself, represents a source of glycoproteins rich in Tn that may offer future biomarker targets.


Subject(s)
Adenoma , Colorectal Neoplasms , Humans , Animals , Mice , Statistics, Nonparametric
9.
Immunity ; 39(6): 1095-107, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24290911

ABSTRACT

Cancers arising in mucosal tissues account for a disproportionately large fraction of malignancies. Immunoglobulin G (IgG) and the neonatal Fc receptor for IgG (FcRn) have an important function in the mucosal immune system that we have now shown extends to the induction of CD8(+) T cell-mediated antitumor immunity. We demonstrate that FcRn within dendritic cells (DCs) was critical for homeostatic activation of mucosal CD8(+) T cells that drove protection against the development of colorectal cancers and lung metastases. FcRn-mediated tumor protection was driven by DCs activation of endogenous tumor-reactive CD8(+) T cells via the cross-presentation of IgG complexed antigens (IgG IC), as well as the induction of cytotoxicity-promoting cytokine secretion, particularly interleukin-12, both of which were independently triggered by the FcRn-IgG IC interaction in murine and human DCs. FcRn thus has a primary role within mucosal tissues in activating local immune responses that are critical for priming efficient anti-tumor immunosurveillance.


Subject(s)
Colorectal Neoplasms/immunology , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Immunity/genetics , Receptors, Fc/genetics , Receptors, Fc/metabolism , Animals , Colorectal Neoplasms/genetics , Dendritic Cells/immunology , Disease Models, Animal , Flow Cytometry , Humans , Immunity, Active , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Cell Mol Life Sci ; 78(14): 5569-5585, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34089345

ABSTRACT

The macrophage mannose receptor (CD206, MR) is an endocytic lectin receptor which plays an important role in homeostasis and innate immunity, however, the endogenous glycan and glycoprotein ligands recognized by its C-type lectin domains (CTLD) have not been well studied. Here we used the murine MR CTLD4-7 coupled to the Fc-portion of human IgG (MR-Fc) to investigate the MR glycan and glycoprotein recognition. We probed 16 different cancer and control tissues using the MR-Fc, and observed cell- and tissue-specific binding with varying intensity. All cancer tissues and several control tissues exhibited MR-Fc ligands, intracellular and/or surface-located. We further confirmed the presence of ligands on the surface of cancer cells by flow cytometry. To characterize the fine specificity of the MR for glycans, we screened a panel of glycan microarrays. Remarkably, the results indicate that the CTLD4-7 of the MR is highly selective for specific types of pauci- and oligomannose N-glycans among hundreds of glycans tested. As lung cancer tissue and the lung cancer cell line A549 showed intense MR-Fc binding, we further investigated the MR glycoprotein ligands in those cells by immunoprecipitation and glycoproteomic analysis. All enriched glycoproteins, of which 42 were identified, contained pauci- or oligomannose N-glycans, confirming the microarray results. Our study demonstrates that the MR CTLD4-7 is highly selective for pauci- and oligomannosidic N-glycans, structures that are often elevated in tumor cells, and suggest a potential role for the MR in tumor biology.


Subject(s)
Glycoproteins/metabolism , Lectins, C-Type/metabolism , Lung Neoplasms/pathology , Mannose-Binding Lectins/metabolism , Oligosaccharides/metabolism , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , A549 Cells , Glycoproteins/genetics , Glycosylation , Humans , Lectins, C-Type/genetics , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mannose Receptor , Mannose-Binding Lectins/genetics , Models, Molecular , Receptors, Cell Surface/genetics
11.
Gastroenterology ; 158(5): 1359-1372.e9, 2020 04.
Article in English | MEDLINE | ID: mdl-31917258

ABSTRACT

BACKGROUND & AIMS: Intestinal microbes and their metabolites affect the development of colorectal cancer (CRC). Short-chain fatty acids are metabolites generated by intestinal microbes from dietary fiber. We investigated the mechanisms by which free fatty acid receptor 2 (FFAR2), a receptor for short-chain fatty acids that can affect the composition of the intestinal microbiome, contributes to the pathogenesis of CRC. METHODS: We performed studies with ApcMin/+ mice, ApcMin/+Ffar2-/- mice, mice with conditional disruption of Ffar2 in dendritic cells (DCs) (Ffar2fl/flCD11c-Cre mice), ApcMin/+Ffar2fl/flCD11c-Cre mice, and Ffar2fl/fl mice (controls); some mice were given dextran sodium sulfate to induce colitis, with or without a FFAR2 agonist or an antibody against interleukin 27 (IL27). Colon and tumor tissues were analyzed by histology, quantitative polymerase chain reaction, and 16S ribosomal RNA gene sequencing; lamina propria and mesenteric lymph node tissues were analyzed by RNA sequencing and flow cytometry. Intestinal permeability was measured after gavage with fluorescently labeled dextran. We collected data on colorectal tumors from The Cancer Genome Atlas. RESULTS: ApcMin/+Ffar2-/- mice developed significantly more spontaneous colon tumors than ApcMin/+ mice and had increased gut permeability before tumor development, associated with reduced expression of E-cadherin. Colon tumors from ApcMin/+Ffar2-/- mice had a higher number of bacteria than tumors from ApcMin/+ mice, as well as higher frequencies of CD39+CD8+ T cells and exhausted or dying T cells. DCs from ApcMin/+Ffar2-/- mice had an altered state of activation, increased death, and higher production of IL27. Administration of an antibody against IL27 reduced the numbers of colon tumors in ApcMin/+ mice with colitis. Frequencies of CD39+CD8+ T cells and IL27+ DCs were increased in colon lamina propria from Ffar2fl/flCD11c-Cre mice with colitis compared with control mice or mice without colitis. ApcMin/+Ffar2fl/flCD11c-Cre mice developed even more tumors than ApcMin/+Ffar2fl/fl mice, and their tumors had even higher numbers of IL27+ DCs. ApcMin/+ mice with colitis given the FFAR2 agonist developed fewer colon tumors, with fewer IL27+ DCs, than mice not given the agonist. DCs incubated with the FFAR2 agonist no longer had gene expression patterns associated with activation or IL27 production. CONCLUSIONS: Loss of FFAR2 promotes colon tumorigenesis in mice by reducing gut barrier integrity, increasing tumor bacterial load, promoting exhaustion of CD8+ T cells, and overactivating DCs, leading to their death. Antibodies against IL27 and an FFAR2 agonist reduce tumorigenesis in mice and might be developed for the treatment of CRC.


Subject(s)
Colitis/pathology , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , Interleukins/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Colitis/chemically induced , Colitis/immunology , Colon/drug effects , Colon/microbiology , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dendritic Cells/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Disease Progression , Fatty Acids, Nonesterified/metabolism , Female , Humans , Interleukins/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Permeability , Primary Cell Culture , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
12.
Nature ; 517(7534): 386-90, 2015 01 15.
Article in English | MEDLINE | ID: mdl-25363763

ABSTRACT

T-cell immunoglobulin domain and mucin domain-3 (TIM-3, also known as HAVCR2) is an activation-induced inhibitory molecule involved in tolerance and shown to induce T-cell exhaustion in chronic viral infection and cancers. Under some conditions, TIM-3 expression has also been shown to be stimulatory. Considering that TIM-3, like cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), is being targeted for cancer immunotherapy, it is important to identify the circumstances under which TIM-3 can inhibit and activate T-cell responses. Here we show that TIM-3 is co-expressed and forms a heterodimer with carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1), another well-known molecule expressed on activated T cells and involved in T-cell inhibition. Biochemical, biophysical and X-ray crystallography studies show that the membrane-distal immunoglobulin-variable (IgV)-like amino-terminal domain of each is crucial to these interactions. The presence of CEACAM1 endows TIM-3 with inhibitory function. CEACAM1 facilitates the maturation and cell surface expression of TIM-3 by forming a heterodimeric interaction in cis through the highly related membrane-distal N-terminal domains of each molecule. CEACAM1 and TIM-3 also bind in trans through their N-terminal domains. Both cis and trans interactions between CEACAM1 and TIM-3 determine the tolerance-inducing function of TIM-3. In a mouse adoptive transfer colitis model, CEACAM1-deficient T cells are hyper-inflammatory with reduced cell surface expression of TIM-3 and regulatory cytokines, and this is restored by T-cell-specific CEACAM1 expression. During chronic viral infection and in a tumour environment, CEACAM1 and TIM-3 mark exhausted T cells. Co-blockade of CEACAM1 and TIM-3 leads to enhancement of anti-tumour immune responses with improved elimination of tumours in mouse colorectal cancer models. Thus, CEACAM1 serves as a heterophilic ligand for TIM-3 that is required for its ability to mediate T-cell inhibition, and this interaction has a crucial role in regulating autoimmunity and anti-tumour immunity.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Immune Tolerance/immunology , Membrane Proteins/metabolism , Receptors, Virus/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, CD/chemistry , Antigens, CD/immunology , Autoimmunity/immunology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cell Line , Colorectal Neoplasms/immunology , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Inflammation/immunology , Inflammation/pathology , Ligands , Male , Membrane Proteins/chemistry , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Mucous Membrane/immunology , Mucous Membrane/pathology , Protein Conformation , Protein Multimerization , Receptors, Virus/chemistry , Receptors, Virus/immunology
13.
Br J Cancer ; 122(9): 1367-1377, 2020 04.
Article in English | MEDLINE | ID: mdl-32157241

ABSTRACT

BACKGROUND: Histological lymphocytic reaction is regarded as an independent prognostic marker in colorectal cancer. Considering the lack of adequate statistical power, adjustment for selection bias and comprehensive tumour molecular data in most previous studies, we investigated the strengths of the prognostic associations of lymphocytic reaction in colorectal carcinoma by utilising an integrative database of two prospective cohort studies. METHODS: We examined Crohn's-like reaction, intratumoural periglandular reaction, peritumoural reaction and tumour-infiltrating lymphocytes in 1465 colorectal carcinoma cases. Using covariate data of 4420 colorectal cancer cases in total, inverse probability-weighted Cox proportional hazard regression model was used to control for selection bias (due to tissue availability) and potential confounders, including stage, MSI status, LINE-1 methylation, PTGS2 and CTNNB1 expression, KRAS, BRAF and PIK3CA mutations, and tumour neoantigen load. RESULTS: Higher levels of each lymphocytic reaction component were associated with better colorectal cancer-specific survival (Ptrend < 0.002). Compared with cases with negative/low intratumoural periglandular reaction, multivariable-adjusted HRs were 0.55 (95% CI, 0.42-0.71) in cases with intermediate reaction and 0.20 (95% CI, 0.12-0.35) in cases with high reaction. These relationships were consistent in strata of MSI status or neoantigen loads (Pinteraction > 0.2). CONCLUSIONS: The four lymphocytic reaction components are prognostic biomarkers in colorectal carcinoma.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Microsatellite Instability , Aged , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/pathology , Cyclooxygenase 2/genetics , Female , Humans , Long Interspersed Nucleotide Elements/genetics , Lymphocyte Count , Lymphocytes/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , beta Catenin/genetics
14.
Radiographics ; 40(5): 1219-1239, 2020.
Article in English | MEDLINE | ID: mdl-32678699

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), an epithelial neoplasm derived from the pancreatic ductal tree, is the most common histologic type of pancreatic cancer and accounts for 85%-95% of all solid pancreatic tumors. As a highly lethal malignancy, it is the seventh leading cause of cancer death worldwide and is responsible for more than 300 000 deaths per year. PDAC is highly resistant to current therapies, affording patients a 5-year overall survival rate of only 7.2%. It is characterized histologically by its highly desmoplastic stroma embedding tubular and ductlike structures. On images, it typically manifests as a poorly defined hypoenhancing mass, causing ductal obstruction and vascular involvement. Little is known about the other histologic subtypes of PDAC, mainly because of their rarity and lack of specific patterns of disease manifestation. According to the World Health Organization, these variants include adenosquamous carcinoma, colloid carcinoma, hepatoid carcinoma, medullary carcinoma, signet ring cell carcinoma, undifferentiated carcinoma with osteoclast-like giant cells, and undifferentiated carcinoma. Depending on the subtype, they can confer a better or even worse prognosis than that of conventional PDAC. Thus, awareness of the existence and differentiation of these variants on the basis of imaging and histopathologic characteristics is crucial to guide clinical decision making for optimal treatment and patient management.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnostic imaging , Magnetic Resonance Imaging , Pancreatic Neoplasms/diagnostic imaging , Tomography, X-Ray Computed , Carcinoma, Pancreatic Ductal/pathology , Contrast Media , Diagnosis, Differential , Humans , Pancreatic Neoplasms/pathology , Prognosis
15.
Proc Natl Acad Sci U S A ; 114(1): 142-147, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27980034

ABSTRACT

Hosts and their microbes have established a sophisticated communication system over many millennia. Within mammalian hosts, this dynamic cross-talk is essential for maintaining intestinal homeostasis. In a genetically susceptible host, dysbiosis of the gut microbiome and dysregulated immune responses are central to the development of inflammatory bowel disease (IBD). Previous surveys of stool from the T-bet-/-Rag2-/- IBD mouse model revealed microbial features that discriminate between health and disease states. Enterobacteriaceae expansion and increased gene abundances for benzoate degradation, two-component systems, and bacterial motility proteins pointed to the potential involvement of a catecholamine-mediated bacterial signaling axis in colitis pathogenesis. Enterobacteriaceae sense and respond to microbiota-generated signals and host-derived catecholamines through the two-component quorum-sensing Escherichia coli regulators B and C (QseBC) system. On signal detection, QseC activates a cascade to induce virulence gene expression. Although a single pathogen has not been identified as a causative agent in IBD, adherent-invasive Escherichia coli (AIEC) have been implicated. Flagellar expression is necessary for the IBD-associated AIEC strain LF82 to establish colonization. Thus, we hypothesized that qseC inactivation could reduce LF82's virulence, and found that an absence of qseC leads to down-regulated flagellar expression and motility in vitro and reduced colonization in vivo. We extend these findings on the potential of QseC-based IBD therapeutics to three preclinical IBD models, wherein we observe that QseC blockade can effectively modulate colitogenic microbiotas to reduce intestinal inflammation. Collectively, our data support a role for QseC-mediated bacterial signaling in IBD pathogenesis and indicate that QseC inhibition may be a useful microbiota-targeted approach for disease management.


Subject(s)
Colitis/pathology , Colitis/therapy , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Quorum Sensing/drug effects , Animals , Catecholamines/metabolism , Colitis/microbiology , Flagella/genetics , Flagella/metabolism , Gastrointestinal Microbiome , Gene Expression Regulation, Bacterial/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Sulfonamides/pharmacology , Virulence/genetics
16.
Nature ; 503(7475): 272-6, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24089213

ABSTRACT

The recognition of autophagy related 16-like 1 (ATG16L1) as a genetic risk factor has exposed the critical role of autophagy in Crohn's disease. Homozygosity for the highly prevalent ATG16L1 risk allele, or murine hypomorphic (HM) activity, causes Paneth cell dysfunction. As Atg16l1(HM) mice do not develop spontaneous intestinal inflammation, the mechanism(s) by which ATG16L1 contributes to disease remains obscure. Deletion of the unfolded protein response (UPR) transcription factor X-box binding protein-1 (Xbp1) in intestinal epithelial cells, the human orthologue of which harbours rare inflammatory bowel disease risk variants, results in endoplasmic reticulum (ER) stress, Paneth cell impairment and spontaneous enteritis. Unresolved ER stress is a common feature of inflammatory bowel disease epithelium, and several genetic risk factors of Crohn's disease affect Paneth cells. Here we show that impairment in either UPR (Xbp1(ΔIEC)) or autophagy function (Atg16l1(ΔIEC) or Atg7(ΔIEC)) in intestinal epithelial cells results in each other's compensatory engagement, and severe spontaneous Crohn's-disease-like transmural ileitis if both mechanisms are compromised. Xbp1(ΔIEC) mice show autophagosome formation in hypomorphic Paneth cells, which is linked to ER stress via protein kinase RNA-like endoplasmic reticulum kinase (PERK), elongation initiation factor 2α (eIF2α) and activating transcription factor 4 (ATF4). Ileitis is dependent on commensal microbiota and derives from increased intestinal epithelial cell death, inositol requiring enzyme 1α (IRE1α)-regulated NF-κB activation and tumour-necrosis factor signalling, which are synergistically increased when autophagy is deficient. ATG16L1 restrains IRE1α activity, and augmentation of autophagy in intestinal epithelial cells ameliorates ER stress-induced intestinal inflammation and eases NF-κB overactivation and intestinal epithelial cell death. ER stress, autophagy induction and spontaneous ileitis emerge from Paneth-cell-specific deletion of Xbp1. Genetically and environmentally controlled UPR function within Paneth cells may therefore set the threshold for the development of intestinal inflammation upon hypomorphic ATG16L1 function and implicate ileal Crohn's disease as a specific disorder of Paneth cells.


Subject(s)
Intestinal Diseases/physiopathology , Intestinal Mucosa/pathology , Paneth Cells/pathology , Animals , Autophagy/genetics , Autophagy-Related Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/genetics , Inflammation , Intestinal Diseases/genetics , Intestinal Mucosa/cytology , Mice , Regulatory Factor X Transcription Factors , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Unfolded Protein Response/physiology , X-Box Binding Protein 1 , eIF-2 Kinase/metabolism
17.
Proc Natl Acad Sci U S A ; 112(25): 7803-8, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056274

ABSTRACT

Beneficial microbes that target molecules and pathways, such as oxidative stress, which can negatively affect both host and microbiota, may hold promise as an inflammatory bowel disease therapy. Prior work showed that a five-strain fermented milk product (FMP) improved colitis in T-bet(-/-) Rag2(-/-) mice. By varying the number of strains used in the FMP, we found that Lactococcus lactis I-1631 was sufficient to ameliorate colitis. Using comparative genomic analyses, we identified genes unique to L. lactis I-1631 involved in oxygen respiration. Respiration of oxygen results in reactive oxygen species (ROS) generation. Also, ROS are produced at high levels during intestinal inflammation and cause tissue damage. L. lactis I-1631 possesses genes encoding enzymes that detoxify ROS, such as superoxide dismutase (SodA). Thus, we hypothesized that lactococcal SodA played a role in attenuating colitis. Inactivation of the sodA gene abolished L. lactis I-1631's beneficial effect in the T-bet(-/-) Rag2(-/-) model. Similar effects were obtained in two additional colonic inflammation models, Il10(-/-) mice and dextran sulfate sodium-treated mice. Efforts to understand how a lipophobic superoxide anion (O2 (-)) can be detoxified by cytoplasmic lactoccocal SodA led to the finding that host antimicrobial-mediated lysis is a prerequisite for SodA release and SodA's extracytoplasmic O2 (-) scavenging. L. lactis I-1631 may represent a promising vehicle to deliver antioxidant, colitis-attenuating SodA to the inflamed intestinal mucosa, and host antimicrobials may play a critical role in mediating SodA's bioaccessibility.


Subject(s)
Colitis/metabolism , Lactococcus lactis/metabolism , Muramidase/metabolism , Superoxide Dismutase/metabolism , Animals , Colitis/enzymology , Colitis/microbiology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Reactive Oxygen Species/metabolism
19.
Proc Natl Acad Sci U S A ; 111(25): E2559-66, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24927559

ABSTRACT

T-bet(-/-).Rag2(-/-) (TRUC) mice spontaneously develop microbiota-driven, TNF-mediated large bowel inflammation that resembles human ulcerative colitis. We show here that IL-23 and IL-1-dependent secretion of IL-17A by innate lymphoid cells (ILCs; defined as CD45(+)lin(-)Thy1(hi)NKp46(-)) is a second critical pathway in this model. Using an in vitro coculture system of bone marrow-derived dendritic cells (DCs) and freshly isolated FACS-purified ILCs, we demonstrate that IL-23 and IL-1 secreted by DCs in response to microbial stimulation work together to induce IL-17A production by ILCs. TNF is not required for IL-17A secretion by ILCs in vitro but synergizes with IL-17A to induce the expression of neutrophil-attracting chemokines. Upstream, activation of the IL-23/IL-17A axis is regulated by nucleotide-binding oligomerization domain containing (Nod)/receptor-interacting serine-threonine kinase 2 (Ripk2) signals in DCs. Genetic ablation of the Nod/Ripk2 signaling pathway protects TRUC mice from developing colitis without affecting the colitogenicity of the intestinal microbiota. Our data provide insight into the complex network of interactions between IL-17A-secreting ILCs and other components of the innate immune system in the development of colitis.


Subject(s)
Bone Marrow Cells/immunology , Colitis, Ulcerative/immunology , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Interleukin-17/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , T-Box Domain Proteins/immunology , Animals , Bacteria/immunology , Bone Marrow Cells/cytology , Chemokines/genetics , Chemokines/immunology , Colitis, Ulcerative/genetics , DNA-Binding Proteins/genetics , Dendritic Cells/pathology , Humans , Interleukin-17/genetics , Interleukin-23/genetics , Interleukin-23/immunology , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , T-Box Domain Proteins/genetics
20.
Proc Natl Acad Sci U S A ; 108(17): 7137-41, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21482794

ABSTRACT

Genetic modifier loci influence the phenotypic expression of many Mendelian traits; insight into disease pathogenesis gained from their identification in animal disease models may impact the treatment of human multigenic disorders. We previously described an innate immune-driven model of spontaneous ulcerative colitis in T-bet(-/-).Rag2(-/-) double-deficient mice that resembles human ulcerative colitis. On a BALB/c background, this disease is highly penetrant and results in the development of colorectal cancer. However, we observed that colitis in T-bet(-/-).Rag2(-/-) mice on a C57BL/6 background was significantly less severe. Quantitative trait locus analysis using an N2 backcross strategy revealed a single major quantitative trait locus on chromosome 3 that mapped to the Cdcs1 (cytokine deficiency-induced colitis susceptibility-1) locus previously identified in the Il10(-/-) and Gnai2(-/-) colitis models. Congenic introduction of the susceptible Cdcs1 interval from C3H/He into the C57BL/6 background restored colitis severity. Bone marrow reconstitution experiments further mapped the effect of host genetics on disease severity to the hematopoietic compartment. There were distinct differences in the expression of several Cdcs1 genes in bone marrow-derived dendritic cells from Cdcs1 congenic mice. We conclude that the Cdcs1 locus controls colitis severity in T-bet(-/-).Rag2(-/-) mice through innate immune cells.


Subject(s)
Chromosomes, Mammalian/genetics , Colitis, Ulcerative/genetics , Gene Expression Regulation/genetics , Immunity, Innate/genetics , Quantitative Trait Loci/genetics , Animals , Bone Marrow Cells/immunology , Chromosomes, Mammalian/immunology , Colitis, Ulcerative/immunology , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression Regulation/immunology , Humans , Immunity, Innate/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Quantitative Trait Loci/immunology
SELECTION OF CITATIONS
SEARCH DETAIL