Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nature ; 611(7937): 787-793, 2022 11.
Article in English | MEDLINE | ID: mdl-36323781

ABSTRACT

Emerging studies indicate that cooperation between neurons and immune cells regulates antimicrobial immunity, inflammation and tissue homeostasis. For example, a neuronal rheostat provides excitatory or inhibitory signals that control the functions of tissue-resident group 2 innate lymphoid cells (ILC2s) at mucosal barrier surfaces1-4. ILC2s express NMUR1, a receptor for neuromedin U (NMU), which is a prominent cholinergic neuropeptide that promotes ILC2 responses5-7. However, many functions of ILC2s are shared with adaptive lymphocytes, including the production of type 2 cytokines8,9 and the release of tissue-protective amphiregulin (AREG)10-12. Consequently, there is controversy regarding whether innate lymphoid cells and adaptive lymphocytes perform redundant or non-redundant functions13-15. Here we generate a new genetic tool to target ILC2s for depletion or gene deletion in the presence of an intact adaptive immune system. Transgenic expression of iCre recombinase under the control of the mouse Nmur1 promoter enabled ILC2-specific deletion of AREG. This revealed that ILC2-derived AREG promotes non-redundant functions in the context of antiparasite immunity and tissue protection following intestinal damage and inflammation. Notably, NMU expression levels increased in inflamed intestinal tissues from both mice and humans, and NMU induced AREG production in mouse and human ILC2s. These results indicate that neuropeptide-mediated regulation of non-redundant functions of ILC2s is an evolutionarily conserved mechanism that integrates immunity and tissue protection.


Subject(s)
Immunity, Innate , Intestinal Mucosa , Lymphocytes , Neuropeptides , Animals , Humans , Mice , Cytokines/immunology , Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/parasitology , Inflammation/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Neuropeptides/physiology , Amphiregulin , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology
2.
Cell Mol Immunol ; 19(7): 777-790, 2022 07.
Article in English | MEDLINE | ID: mdl-35468944

ABSTRACT

The food colorant Red 40 is an environmental risk factor for colitis development in mice with increased expression of interleukin (IL)-23. This immune response is mediated by CD4+ T cells, but mechanistic insights into how these CD4+ T cells trigger and perpetuate colitis have remained elusive. Here, using single-cell transcriptomic analysis, we found that several CD4+ T-cell subsets are present in the intestines of colitic mice, including an interferon (IFN)-γ-producing subset. In vivo challenge of primed mice with Red 40 promoted rapid activation of CD4+ T cells and caused marked intestinal epithelial cell (IEC) apoptosis that was attenuated by depletion of CD4+ cells and blockade of IFN-γ. Ex vivo experiments showed that intestinal CD4+ T cells from colitic mice directly promoted apoptosis of IECs and intestinal enteroids. CD4+ T cell-mediated cytotoxicity was contact-dependent and required FasL, which promoted caspase-dependent cell death in target IECs. Genetic ablation of IFN-γ constrained IL-23- and Red 40-induced colitis development, and blockade of IFN-γ inhibited epithelial cell death in vivo. These results advance the understanding of the mechanisms regulating colitis development caused by IL-23 and food colorants and identify IFN-γ+ cytotoxic CD4+ T cells as a new potential therapeutic target for colitis.


Subject(s)
CD4-Positive T-Lymphocytes , Colitis , Food Coloring Agents , Interleukin-23 , Animals , CD4-Positive T-Lymphocytes/immunology , Colitis/chemically induced , Colitis/immunology , Food Coloring Agents/adverse effects , Interferon-gamma/metabolism , Interleukin-23/adverse effects , Mice , Mice, Inbred C57BL
3.
Mol Med Rep ; 24(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34278485

ABSTRACT

Forkhead­box gene 1 (FOXG1) has been reported to serve an important role in various malignancies, but its effects on nasopharyngeal cancer (NPC) remain unknown. Thus, the present study aimed to investigate the specific regulatory relationship between FOXG1 and NPC progression. Tumor tissues and matching para­carcinoma tissues were obtained from patients with NPC. Small interfering (si)RNA­FOXG1 and pcDNA3.1­FOXG1 were transfected into SUNE­1 and C666­1 cells to knockdown and overexpress FOXG1 expression, respectively. FOXG1 expression was detected using reverse transcription­quantitative PCR and immunohistochemistry. Cell proliferation was detected using MTT and 5­ethynyl­20­deoxyuridine assays. Transwell invasion assay, wound healing assay and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. Western blotting was conducted to detect the expression levels of mitochondrial markers (succinate dehydrogenase complex flavoprotein subunit A, heat shock protein 60 and pyruvate dehydrogenase), epithelial­mesenchymal transition (EMT) related proteins (N­cadherin, Snail and E­cadherin) and apoptosis­related proteins [Bax, Bcl­2, poly(ADP­ribose) polymerase 1 (PARP), cleaved PARP, cleaved caspase­3, cleaved caspase­8, cleaved caspase­9, caspase­3, caspase­8 and caspase­9]. The mitochondrial membrane potential was detected via flow cytometry, while the ATP/ADP ratio was determined using the ADP/ATP ratio assay kit. The present results demonstrated that FOXG1 expression was upregulated in NPC tissues and cells, and was associated with distant metastasis and TNM stage. Moreover, knockdown of FOXG1 inhibited the proliferation, migration, invasion, EMT and mitochondrial function of SUNE­1 cells, as well as promoted cell apoptosis, while the opposite results were observed in C666­1 cells. In conclusion, FOXG1 enhanced proliferation, migration and invasion, induced EMT and improved mitochondrial function in NPC cells. The current findings provide an adequate theoretical basis for the treatment of NPC.


Subject(s)
Forkhead Transcription Factors/metabolism , Mitochondria/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Adult , Aged , Apoptosis , Carcinoma/genetics , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mitochondria/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nerve Tissue Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA, Small Interfering/metabolism , Up-Regulation
4.
Front Oncol ; 11: 684462, 2021.
Article in English | MEDLINE | ID: mdl-34268117

ABSTRACT

BACKGROUND: The involvement of microRNA-338-5p in modulating NPC pathogenesis is still largely unknown, and this study aimed to investigate this issue. METHODS: The expressions of cancer associated genes were determined by Real-Time qPCR and Western Blot, and cell apoptosis was determined by flow cytometer (FCM). CCK-8 assay and colony formation assay were respectively used to determine cell proliferation and colony formation abilities. Transwell assay was used to evaluate cell migration. The expression levels of Ki67 protein in mice tissues were measured by Immunohistochemistry (IHC) assay. RESULTS: The present study found that microRNA-338-5p suppressed NPC progression by degrading its downstream target, Wnt family member 2B (WNT2B). Specifically, microRNA-338-5p tended to be low-expressed in NPC tissues and cell lines, compared to the non-tumor nasopharyngeal mucosa tissues and normal nasopharyngeal cell line (NP69). Upregulation of microRNA-338-5p inhibited proliferation, mobility, and epithelial-mesenchymal transition (EMT) in NPC cells in vitro, while silencing of microRNA-338-5p had opposite effects. Consistently, microRNA-338-5p suppressed tumorigenesis of NPC cells in vivo. In addition, microRNA-338-5p targeted WNT2B for degradation and inhibition, and the inhibiting effects of microRNA-338-5p overexpression on NPC development were reversed by upregulating WNT2B. CONCLUSIONS: Taken together, we concluded that microRNA-338-5p targeted WNT2B to hinder NPC development.

5.
Nat Immunol ; 22(6): 757-768, 2021 06.
Article in English | MEDLINE | ID: mdl-34031614

ABSTRACT

Maturation of B cells within germinal centers (GCs) generates diversified B cell pools and high-affinity B cell antigen receptors (BCRs) for pathogen clearance. Increased receptor affinity is achieved by iterative cycles of T cell-dependent, affinity-based B cell positive selection and clonal expansion by mechanisms hitherto incompletely understood. Here we found that, as part of a physiologic program, GC B cells repressed expression of decay-accelerating factor (DAF/CD55) and other complement C3 convertase regulators via BCL6, but increased the expression of C5b-9 inhibitor CD59. These changes permitted C3 cleavage on GC B cell surfaces without the formation of membrane attack complex and activated C3a- and C5a-receptor signals required for positive selection. Genetic disruption of this pathway in antigen-activated B cells by conditional transgenic DAF overexpression or deletion of C3a and C5a receptors limited the activation of mechanistic target of rapamycin (mTOR) in response to BCR-CD40 signaling, causing premature GC collapse and impaired affinity maturation. These results reveal that coordinated shifts in complement regulation within the GC provide crucial signals underlying GC B cell positive selection.


Subject(s)
B-Lymphocytes/immunology , Complement Activation , Complement C3a/metabolism , Complement C5a/metabolism , Germinal Center/immunology , Animals , Animals, Genetically Modified , B-Lymphocytes/metabolism , CD55 Antigens/genetics , CD55 Antigens/metabolism , CD59 Antigens/metabolism , Cell Line, Tumor , Clonal Hematopoiesis/immunology , Germinal Center/cytology , Germinal Center/metabolism , Humans , Lymphocyte Activation , Mice , Palatine Tonsil/cytology , Palatine Tonsil/pathology , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, Complement/genetics , Receptors, Complement/metabolism , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism
6.
Cell Metab ; 33(7): 1358-1371.e5, 2021 07 06.
Article in English | MEDLINE | ID: mdl-33989521

ABSTRACT

Both genetic predisposition and environmental factors appear to play a role in inflammatory bowel disease (IBD) development. Genetic studies in humans have linked the interleukin (IL)-23 signaling pathway with IBD, but the environmental factors contributing to disease have remained elusive. Here, we show that the azo dyes Red 40 and Yellow 6, the most abundant food colorants in the world, can trigger an IBD-like colitis in mice conditionally expressing IL-23, or in two additional animal models in which IL-23 expression was augmented. Increased IL-23 expression led to generation of activated CD4+ T cells that expressed interferon-γ and transferred disease to mice exposed to Red 40. Colitis induction was dependent on the commensal microbiota promoting the azo reduction of Red 40 and generation of a metabolite, 1-amino-2-naphthol-6-sulfonate sodium salt. Together these findings suggest that specific food colorants represent novel risk factors for development of colitis in mice with increased IL-23 signaling.


Subject(s)
Bacteria/metabolism , Colitis , Food Coloring Agents/metabolism , Interleukin-23/genetics , Intestinal Mucosa/microbiology , Animals , Colitis/genetics , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Food Coloring Agents/adverse effects , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Interferon-gamma/genetics , Interleukin-23/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Symbiosis
7.
Sci Rep ; 10(1): 13925, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32811883

ABSTRACT

The identification of suspicious microseismic events is the first crucial step in microseismic data processing. Existing automatic classification methods are based on the training of a large data set, which is challenging to apply in mines without a long-term manual data processing. In this paper, we present a method to automatically classify microseismic records with limited samples in underground mines based on capsule networks (CapsNet). We divide each microseismic record into 33 frames, then extract 21 commonly used features in time and frequency from each frame. Consequently, a 21 × 33 feature matrix is utilized as the input of CapsNet. On this basis, we use different sizes of training sets to train the classification models separately. The trained model is tested using the same test set containing 3,200 microseismic records and compared to convolutional neural networks (CNN) and traditional machine learning methods. Results show that the accuracy of our proposed method is 99.2% with limited training samples. It is superior to CNN and traditional machine learning methods in terms of Accuracy, Precision, Recall, F1-Measure, and reliability.

8.
Sci Rep ; 10(1): 8259, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32427877

ABSTRACT

Psoriasis (PS) is a chronic skin inflammation. Up to 30% of the patients with PS develop psoriatic arthritis (PsA), a condition characterized by inflammatory arthritis that affects joints or entheses. Although there is mounting evidence for a critical role of interleukin-23 (IL-23) signaling in the pathogenesis of both PS and PsA, it remains unclear whether IL-23-induced skin inflammation drives joint disease. Here, we show that mice expressing increased levels of IL-23 in the skin (K23 mice) develop a PS-like disease that is characterized by acanthosis, parakeratosis, hyperkeratosis, and inflammatory infiltrates in the dermis. Skin disease preceded development of PsA, including enthesitis, dactylitis, and bone destruction. The development of enthesitis and dactylitis was not due to high circulating levels of IL-23, as transgenic animals and controls had similar levels of this cytokine in circulation. IL-22, a downstream cytokine of IL-23, was highly increased in the serum of K23 mice. Although IL-22 deficiency did not affect skin disease development, IL-22 deficiency aggravated the PsA-like disease in K23 mice. Our results demonstrate a central role for skin expressed IL-23 in the initiation of PS and on pathogenic processes leading to PsA.


Subject(s)
Arthritis, Psoriatic/genetics , Interleukin-23/genetics , Psoriasis/genetics , Skin/immunology , Animals , Arthritis, Psoriatic/immunology , Female , Humans , Interleukin-23/immunology , Interleukins/genetics , Interleukins/immunology , Male , Mice , Psoriasis/immunology , Interleukin-22
9.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32160524

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Subject(s)
Immunity, Cellular , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-33/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Tryptophan Hydroxylase/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Disease Susceptibility , Gene Expression Regulation/immunology , Immunity, Innate , Immunity, Mucosal , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-33/genetics , Larva/growth & development , Larva/immunology , Larva/pathogenicity , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/growth & development , Nippostrongylus/pathogenicity , Primary Cell Culture , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology , Strongylida Infections/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/parasitology , Tryptophan Hydroxylase/genetics
10.
Sensors (Basel) ; 20(2)2020 Jan 20.
Article in English | MEDLINE | ID: mdl-31968704

ABSTRACT

The velocity model is a key factor that affects the accuracy of microseismic event location around tunnels. In this paper, we consider the effect of the empty area on the microseismic event location and present a 3D heterogeneous velocity model for excavated tunnels. The grid-based heterogeneous velocity model can describe a 3D arbitrarily complex velocity model, where the microseismic monitoring areas are divided into many blocks. The residual between the theoretical arrival time calculated by the fast marching method (FMM) and the observed arrival time is used to identify the block with the smallest residual. Particle swarm optimization (PSO) is used to improve the location accuracy in this block. Synthetic tests show that the accuracy of the microseismic event location based on the heterogeneous velocity model was higher than that based on the single velocity model, independent of whether an arrival time error was considered. We used the heterogeneous velocity model to locate 7 blasting events and 44 microseismic events with a good waveform quality in the Qinling No. 4 tunnel of the Yinhanjiwei project from 6 June 2017 to 13 June 2017 and compared the location results of the heterogeneous-velocity model with those of the single-velocity model. The results of this case study show that the events located by the heterogeneous velocity model were concentrated around the working face, which matched the actual conditions of the project, while the events located by the single-velocity model were scattered and far from the working face.

11.
Nat Commun ; 10(1): 4517, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586069

ABSTRACT

Neonatal inflammatory diseases are associated with severe morbidity, but the inflammatory factors underlying them and their potential effector mechanisms are poorly defined. Here we show that necrotizing enterocolitis in neonate mice is accompanied by elevation of IL-23 and IL-22 and decreased production of pancreatic enzymes. These phenotypes are mirrored in neonate mice overexpressing IL-23 in CX3CR1+ myeloid cells or in keratinocytes. The mice fail to grow and die prematurely, displaying systemic inflammation, nutrient malabsorption and decreased expression of intestinal and pancreatic genes mediating digestion and absorption of carbohydrates, proteins, and lipids. Germ-free environment improves, and genetic ablation of IL-22 restores normal growth in mice overexpressing IL-23. Mechanistically, IL-22 acts directly at the level of pancreatic acinar cells to decrease expression of the pancreas associated transcription factor 1a (PTF1a). These results show that augmented production of IL-23 and IL-22 in early life has a negative impact on pancreatic enzyme secretion and food absorption.


Subject(s)
Enterocolitis, Necrotizing/immunology , Interleukin-23/metabolism , Interleukins/metabolism , Pancreas/enzymology , Transcription Factors/metabolism , Acinar Cells/enzymology , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Humans , Interleukin-23/genetics , Interleukin-23/immunology , Interleukins/genetics , Interleukins/immunology , Intestinal Absorption/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Keratinocytes , Mice , Mice, Knockout , Myeloid Cells , Pancreas/cytology , Primary Cell Culture , Interleukin-22
12.
Gastroenterology ; 157(6): 1572-1583.e8, 2019 12.
Article in English | MEDLINE | ID: mdl-31470007

ABSTRACT

BACKGROUND & AIMS: Transgenic mice (HBUS) that express the epidermal growth factor receptor (EGFR) ligand HBEGF (heparin-binding epidermal growth factor-like growth factor) and a constitutively active G protein-coupled receptor (US28) in intestinal epithelial cells develop serrated polyps in the cecum. Development of serrated polyps depends on the composition of the gut microbiota and is associated with bacterial invasion of the lamina propria, accompanied by induction of inflammation and up-regulation of interleukin 1 beta (IL1B) and matrix metalloproteinase (MMP) 3 in the cecum. We investigated the mechanisms by which these changes contribute to development of serrated polyps. METHODS: We performed studies with C57BL/6 (control) and HBUS mice. To accelerate polyp development, we increased the exposure of the bacteria to the lamina propria by injecting HBUS mice with diphtheria toxin, which binds transgenic HBEGF expressed by the epithelial cells and causes apoptosis. Mice were given injections of IL1B-neutralizing antibody and the MMP inhibitor N-isobutyl-N-(4-methoxyphenylsulfonyl)glycyl hydroxamic acid. Intestinal tissues were collected from mice and analyzed by histology, reverse-transcription polymerase chain reaction, enzyme-linked immunosorbent assay, immunofluorescence, and flow cytometry. We examined fibroblast subsets in polyps using single-cell RNA sequencing. RESULTS: Administration of diphtheria toxin to HBUS mice accelerated development of serrated polyps (95% of treated mice developed polyps before 100 days of age, compared with 53% given vehicle). IL1B stimulated subsets of platelet-derived growth factor receptor alpha+ (PDGRFA+) fibroblasts isolated from cecum, resulting in increased expression of MMP3. Neutralizing antibodies against IL1B or administration of the MMP inhibitor reduced the number of serrated polyps that formed in the HBUS mice. Single-cell RNA sequencing analysis showed subsets of fibroblasts in serrated polyps that express genes that regulate matrix fibroblasts and inflammation. CONCLUSIONS: In studies of mice, we found that barrier breakdown and expression of inflammatory factors contribute to development of serrated polyps. Subsets of cecal PDGFRA+ fibroblasts are activated by release of IL1B from myeloid cells during the early stages of serrated polyp development. MMP3 produced by PDGFRA+ fibroblasts is important for serrated polyp development. Our findings confirm the functions of previously identified serrated polyp-associated molecules and indicate roles for immune and stromal cells in serrated polyp development.


Subject(s)
Colonic Polyps/immunology , ErbB Receptors/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/pathology , Matrix Metalloproteinase 3/metabolism , Animals , Apoptosis/immunology , Cecum/cytology , Cecum/immunology , Cecum/pathology , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , Fibroblasts/immunology , Fibroblasts/metabolism , Gefitinib/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Hydroxamic Acids/pharmacology , Interleukin-1beta/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Matrix Metalloproteinase 3/immunology , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sulfonamides/pharmacology
13.
Cytokine ; 111: 146-153, 2018 11.
Article in English | MEDLINE | ID: mdl-30145369

ABSTRACT

Interleukin 33 (IL33) is a cytokine found in the extracellular space (mature IL33) or in the cell nucleus (full-length IL33). Nuclear accumulation of IL33 has been reported in intestinal epithelial cells (IEC) during intestinal inflammation and cancer, but a functional role for this nuclear form remains unclear. To study the role of nuclear IL33 in IEC, we generated transgenic mice expressing full-length IL33 in the intestinal epithelium (Vfl33 mice). Expression of full-length IL33 in the epithelium resulted in accumulation of IL33 protein in the nucleus and secretion of IL33. Over-expression of full-length IL33 by IEC did not promote gut inflammation, but induced expression of genes in the IEC and lamina propria lymphocytes (LPL) that correlated negatively with genes expressed in inflammatory bowel diseases (IBD). Because the IL33 receptor ST2 is expressed by IEC, there was the potential that both the mature and full-length forms could mediate this effect. To specifically interrogate the transcriptional role of nuclear IL33, we intercrossed the Vfl33 mice with ST2- deficient mice. ST2 deficiency completely abrogated the transcriptional effects elicited by IL33 expression, suggesting that the transcriptional effects of IL33 on IEC are mediated by its mature, not its nuclear form.


Subject(s)
Cell Nucleus/immunology , Enterocytes/immunology , Gene Expression Regulation/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-33/immunology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Enterocytes/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/genetics , Lymphocytes/immunology , Lymphocytes/pathology , Mice , Mice, Knockout
14.
Gastroenterology ; 155(4): 1177-1191.e16, 2018 10.
Article in English | MEDLINE | ID: mdl-29909020

ABSTRACT

BACKGROUND & AIMS: Several studies have shown that signaling via the interleukin 23 (IL23) receptor is required for development of colitis. We studied the roles of IL23, dietary factors, alterations to the microbiota, and T cells in the development and progression of colitis in mice. METHODS: All mice were maintained on laboratory diet 5053, unless otherwise noted. We generated mice that express IL23 in CX3CR1-positive myeloid cells (R23FR mice) upon cyclic administration of tamoxifen dissolved in diet 2019. Diets 2019 and 5053 have minor differences in the overall composition of protein, fat, fiber, minerals, and vitamins. CX3CR1CreER mice (FR mice) were used as controls. Some mice were given antibiotics, and others were raised in a germ-free environment. Intestinal tissues were collected and analyzed by histology and flow cytometry. Feces were collected and analyzed by 16S rDNA sequencing. Feces from C57/Bl6, R23FR, or FR mice were fed to FR and R23FR germ-free mice in microbiota transplant experiments. We also performed studies with R23FR/Rag-/-, R23FR/Mu-/-, and R23FR/Tcrd-/- mice. R23FR mice were given injections of antibodies against CD4 or CD8 to deplete T cells. Mesenteric lymph nodes and large intestine CD4+ cells from R23FR or FR mice in remission from colitis were transferred into Rag-/- mice. CD4+ cells were isolated from donor R23FR mice and recipient Rag-/- mice, and T-cell receptor sequences were determined. RESULTS: Expression of IL23 led to development of a relapsing-remitting colitis that was dependent on the microbiota and CD4+ T cells. The relapses were caused by switching from the conventional diet used in our facility (diet 5053) to the diet 2019 and were not dependent on tamoxifen after the first cycle. The switch in the diet modified the microbiota but did not alter levels of IL23 in intestinal tissues compared with mice that remained on the conventional diet. Mesenteric lymph nodes and large intestine CD4+ cells from R23FR mice in remission, but not from FR mice, induced colitis after transfer into Rag-/- mice, but only when these mice were placed on the diet 2019. The CD4+ T-cell receptor repertoire of Rag-/- mice with colitis (fed the 2019 diet) was less diverse than that from donor mice and Rag-/- mice without colitis (fed the 5053 diet) because of expansion of dominant T-cell clones. CONCLUSIONS: We developed mice that express IL23 in CX3CR1-positive myeloid cells (R23FR mice) and found that they are more susceptible to diet-induced colitis than mice that do not express IL23. The R23FR mice have a population of CD4+ T cells that becomes activated in response to dietary changes and alterations to the intestinal microbiota. The results indicate that alterations in the diet, intestinal microbiota, and IL23 signaling can contribute to pathogenesis of inflammatory bowel disease.


Subject(s)
Animal Feed , CD4-Positive T-Lymphocytes/metabolism , Colitis/diet therapy , Colon/metabolism , Gastrointestinal Microbiome , Interleukin-23/metabolism , Myeloid Cells/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CX3C Chemokine Receptor 1/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Disease Models, Animal , Disease Progression , Feces/microbiology , Gene-Environment Interaction , Host-Pathogen Interactions , Interleukin-23/genetics , Mice, Inbred C57BL , Mice, Transgenic , Nutritive Value , Signal Transduction , Time Factors
15.
Sci Rep ; 7(1): 5520, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28710436

ABSTRACT

Increased expression of Interleukin (IL)-33 has been detected in intestinal samples of patients with ulcerative colitis, a condition associated with increased risk for colon cancer, but its role in the development of colorectal cancer has yet to be fully examined. Here, we investigated the role of epithelial expressed IL-33 during development of intestinal tumors. IL-33 expression was detected in epithelial cells in colorectal cancer specimens and in the Apc Min/+ mice. To better understand the role of epithelial-derived IL-33 in the intestinal tumorigenesis, we generated transgenic mice expressing IL-33 in intestinal epithelial cells (V33 mice). V33 Apc Min/+ mice, resulting from the cross of V33 with Apc Min/+ mice, had increased intestinal tumor burden compared with littermate Apc Min/+ mice. Consistently, Apc Min/+ mice deficient for IL-33 receptor (ST2), had reduced polyp burden. Mechanistically, overexpression of IL-33 promoted expansion of ST2+ regulatory T cells, increased Th2 cytokine milieu, and induced alternatively activated macrophages in the gut. IL-33 promoted marked changes in the expression of antimicrobial peptides, and antibiotic treatment of V33 Apc Min/+ mice abrogated the tumor promoting-effects of IL-33 in the colon. In conclusion, elevated IL-33 signaling increases tumor development in the Apc Min/+ mice.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Cell Transformation, Neoplastic/immunology , Epithelial Cells/metabolism , Interleukin-33/metabolism , Intestinal Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cytokines/metabolism , Epithelial Cells/cytology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Intestinal Neoplasms/genetics , Macrophage Activation , Macrophages/immunology , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Up-Regulation
16.
Infect Agent Cancer ; 12: 14, 2017.
Article in English | MEDLINE | ID: mdl-28228842

ABSTRACT

BACKGROUND: Investigations on the effects of malaria infection on cancer mortality are limited except for the incidence of Burkitt's lymphoma (BL) in African children. Our previous murine lung cancer model study demonstrated that malaria infection significantly inhibited tumor growth and prolonged the life span of tumor-bearing mice. This study aims to assess the possible associations between malaria incidence and human cancer mortality. METHODS: We compiled data on worldwide malaria incidence and age-standardized mortality related to 30 types of cancer in 56 countries for the period 1955-2008, and analyzed their longitudinal correlations by a generalized additive mixed model (GAMM), adjusted for a nonlinear year effect and potential confounders such as country's income levels, life expectancies and geographical locations. RESULTS: Malaria incidence was negatively correlated with all-cause cancer mortality, yielding regression coefficients (log scale) of -0.020 (95%CI: -0.027,-0.014) for men (P < 0.001) and-0.020 (95%CI: -0.025,-0.014) for women (P < 0.001). Among the 29 individual types of cancer studied, malaria incidence was negatively correlated with colorectum and anus (men and women), colon (men and women), lung (men), stomach (men), and breast (women) cancer. CONCLUSIONS: Our analysis revealed a possible inverse association between malaria incidence and the mortalities of all-cause and some types of solid cancers, which is opposite to the known effect of malaria on the pathogenesis of Burkitt's lymphoma. Activation of the whole immune system, inhibition of tumor angiogenesis by Plasmodium infection may partially explain why endemic malaria might reduce cancer mortality at the population level.

17.
Mucosal Immunol ; 8(2): 390-402, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25160819

ABSTRACT

Interleukin-23 (IL-23) responsive group 3 innate lymphoid cells (ILC3s) have been implicated in immune homeostasis and pathogenesis in the adult, but little is known about their roles in the newborn. Here we show that IL-23 promotes conversion of embryonic intestinal Lin(-)IL-23R(+)Thy1(+) cells into IL-22-producing Thy1(+)Sca-1(hi) ILC3s in vitro. Gut-specific expression of IL-23 also activated and expanded Thy1(+)Sca-1(hi) ILC3s, which produced IL-22, IL-17, interferon gamma (IFN-γ), and granulocyte-macrophage colony-stimulating factor (GM-CSF) and were distinct from canonical CD4(+) lymphoid tissue inducer (LTi) cells. These ILC3s accumulated under the epithelium in intercellular adhesion molecule (ICAM)-1-positive cell aggregates together with neutrophils that disrupted the epithelium, leading to the formation of discrete intestinal erosions, bleeding, and neonatal death. Genetic and antibody depletion of ILC3s rescued the mice from neonatal death. Antibiotic treatment of pregnant mothers and offspring prolonged survival of IL-23 transgenic mice, suggesting a role for the commensal flora on ILC3-induced pathogenesis. Our results reveal a novel role for the IL-23-ILC3s axis in the pathogenesis of neonatal intestinal inflammation.


Subject(s)
Immunity, Innate , Interleukin-23/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Animals , Antigens, Surface/metabolism , Cytokines/metabolism , Gene Expression , Humans , Immunity, Innate/genetics , Immunophenotyping , Interleukin-23/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/microbiology , Intestines/pathology , Mice , Mice, Transgenic , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Permeability , Phenotype
18.
ACS Med Chem Lett ; 5(1): 89-93, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24900778

ABSTRACT

Given the threat of drug resistance, there is an acute need for new classes of antimalarial agents that act via a unique mechanism of action relative to currently used drugs. We have identified a set of druglike compounds within the Tres Cantos Anti-Malarial Set (TCAMS) which likely act via inhibition of a Plasmodium aspartic protease. Structure-activity relationship analysis and optimization of these aminohydantoins demonstrate that these compounds are potent nanomolar inhibitors of the Plasmodium aspartic proteases PM-II and PM-IV and likely one or more other Plasmodium aspartic proteases. Incorporation of a bulky group, such as a cyclohexyl group, on the aminohydantion N-3 position gives enhanced antimalarial potency while reducing inhibition of human aspartic proteases such as BACE. We have identified compound 8p (CWHM-117) as a promising lead for optimization as an antimalarial drug with a low molecular weight, modest lipophilicity, oral bioavailability, and in vivo antimalarial activity in mice.

19.
J Exp Med ; 211(3): 457-72, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24590763

ABSTRACT

The preferential localization of some neoplasms, such as serrated polyps (SPs), in specific areas of the intestine suggests that nongenetic factors may be important for their development. To test this hypothesis, we took advantage of transgenic mice that expressed HB-EGF throughout the intestine but developed SPs only in the cecum. Here we show that a host-specific microbiome was associated with SPs and that alterations of the microbiota induced by antibiotic treatment or by embryo transfer rederivation markedly inhibited the formation of SPs in the cecum. Mechanistically, development of SPs was associated with a local decrease in epithelial barrier function, bacterial invasion, production of antimicrobials, and increased expression of several inflammatory factors such as IL-17, Cxcl2, Tnf-α, and IL-1. Increased numbers of neutrophils were found within the SPs, and their depletion significantly reduced polyp growth. Together these results indicate that nongenetic factors contribute to the development of SPs and suggest that the development of these intestinal neoplasms in the cecum is driven by the interplay between genetic changes in the host, an inflammatory response, and a host-specific microbiota.


Subject(s)
Cecum/pathology , Epithelium/physiology , Gene Expression Regulation, Neoplastic/immunology , Intestinal Polyps/pathology , Microbiota/physiology , Models, Molecular , Animals , Anti-Bacterial Agents/pharmacology , Cytokines/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Polyps/metabolism , Mice , Mice, Transgenic , Protein Conformation , Species Specificity
20.
J Nat Prod ; 74(10): 2122-7, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21977916

ABSTRACT

Four new ß-carboline alkaloids, designated marinacarbolines A-D (1-4), two new indolactam alkaloids, 13-N-demethyl-methylpendolmycin (5) and methylpendolmycin-14-O-α-glucoside (6), and the three known compounds 1-acetyl-ß-carboline (7), methylpendolmycin (8), and pendolmycin (9) were obtained from the fermentation broth of Marinactinospora thermotolerans SCSIO 00652, a new actinomycete belonging to the family Nocardiopsaceae. Their structures were elucidated by extensive MS and 1D and 2D NMR spectroscopic data analyses. The structure of compound 1 was further confirmed by single-crystal X-ray crystallography. The new compounds 1-6 were inactive against a panel of eight tumor cell lines (IC50>50 µM) but exhibited antiplasmodial activities against Plasmodium falciparum lines 3D7 and Dd2, with IC50 values ranging from 1.92 to 36.03 µM.


Subject(s)
Actinomycetales/chemistry , Alkaloids/isolation & purification , Antimalarials/isolation & purification , Antineoplastic Agents/isolation & purification , Carbolines/isolation & purification , Alkaloids/chemistry , Alkaloids/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbolines/chemistry , Carbolines/pharmacology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Marine Biology , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Plasmodium falciparum/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...